Archives

Pituitary and Adrenal Disorders of Pregnancy

ABSTRACT

 

The pituitary and adrenal glands play an integral role in the endocrine and physiological changes of normal pregnancy. These changes are associated with alterations in the normal ranges of endocrine tests and in the appearance of the glands. It is important for the medical team to be aware of these altered normal ranges in the pregnant population. Some disorders affect women more commonly during pregnancy or the puerperium, e.g., lymphocytic hypophysitis, while other pre-existing disorders such as macroprolactinomas can have worse outcomes in pregnancy. Other conditions may be incidental to pregnancy, but management strategies require modification to ensure safety of the pregnant woman or fetus. This chapter describes the normal physiological alterations in the pituitary and adrenal glands and describes the impact of disorders of these endocrine glands on pregnant women, the fetus, and children of affected women. It also describes the existing data with regard to safety of drugs used to treat pituitary and adrenal disease in pregnancy.

 

PITUITARY DISORDERS IN PREGNANCY

 

Anterior Pituitary Gland Anatomy

 

The pituitary gland enlarges in a three-dimensional fashion by approximately 136% throughout pregnancy (1). There is a progressive linear increase in pituitary height of approximately 0.12mm for each gestational week (2), and the gland is thought to reach its peak size in the first 3 days postpartum when it may reach a height of 12mm on magnetic resonance imaging (MRI) (1, 2). The superior aspect of the gland expands to adopt a dome-like contour moving closer to the optic chiasm (3). Despite this enlargement, compressive symptoms are not typically seen during pregnancy.

 

Pituitary gland enlargement is related to estrogen-stimulated hypertrophy and hyperplasia of the lactotrophs (4). While lactotroph cells make up 20% of anterior pituitary cells in the nonpregnant state, they comprise up to 60% by the third trimester of pregnancy (5). Gonadotrophs decline in number during pregnancy whilst numbers of corticotrophs and thyrotrophs remain constant (5). Somatotrophs are generally suppressed (as a consequence of negative feedback secondary to high levels of insulin like growth factor-1 which is stimulated by placental growth hormone) and may function as lactotrophs (5, 6). After the initial increase in size postpartum there is then a reduction in size back to normal by 6 months postpartum (regardless of breastfeeding status) (2).

 

An understanding of these anatomical changes is important when investigating suspected pituitary pathology during pregnancy, and also when monitoring pre-existing tumors (1). Investigation should be reserved for those patients with symptoms or signs consistent with tumor or pituitary enlargement, asymmetrical growth, deviation of the stalk, or when the height of the pituitary is larger than expected in pregnancy (7).

 

The preferred mode of pituitary imaging in pregnancy is non-contrast MRI, which is considered safe due to the absence of ionizing radiation. There is reassuring evidence for its use in all trimesters with no evidence for adverse pregnancy outcomes (8-10). As a precaution, it has been suggested that 1.5-Tesla scanners should be used rather than 3.0-Tesla scanners as the specific absorption rate quadruples when the magnetic field doubles, although the risk of this is theoretical (8, 11).

 

Prolactin

 

INTRODUCTION

 

Prolactin (PRL) is secreted by the pituitary and a number of extra pituitary sites including the hypothalamus, lymphocytes, uterus, placenta and lactating mammary gland (12, 13). Extra-pituitary secretion, however, is thought to account for only a small proportion of the overall secretion, as supported by one study which reported that hypophysectomized female rats had 10-20% lactogenic activity in their serum compared to control (14). In combination with other hormones, PRL mediates mammogenesis, lactogenesis, and galactopoiesis, and plays a role in the regulation of humoral and cellular immune responses. Placental estrogen stimulates lactotroph PRL synthesis in the first trimester (15, 16) while progesterone also stimulates prolactin secretion (17, 18). Prolactin levels progressively increase throughout gestation (approximately 10-fold) (19), and then decline postpartum in non-lactating women. Despite increased PRL levels, the normal lactotroph continues to respond to TRH and anti-dopaminergic stimulation. Postpartum, the circadian rhythm of PRL release is enhanced by the effects of suckling.

 

FERTILITY

 

Hyperprolactinemia has been reported to account for between 7% and 20% of female infertility (20). It reduces luteinizing hormone (LH) pulse amplitude and frequency through suppression of gonadotrophin-releasing hormone (GnRH) (21), and is associated with diminished positive estrogen feedback on gonadotrophin secretion at mid-cycle (22). Prolactin also has a direct effect on the ovarian granulosa cells and suppresses progesterone and estrogen secretion from the ovaries (23). It can decrease estrogen levels through a direct effect on ovarian aromatase activity and by blocking the stimulatory effects of follicle-stimulating hormone (FSH) (24, 25). At high levels PRL also inhibits progesterone production (26). As a consequence, most hyperprolactinemic women become anovulatory with resultant amenorrhea and infertility (27).

 

PRECONCEPTION MANAGEMENT AND RESTORATION OF MENSES

 

Dopamine agonists remain the treatment of choice for the majority of patients with a prolactinoma. Bromocriptine restores ovulatory menses in 70-80% of patients with 50-75% of patients experiencing an over 50% reduction in size of the pituitary tumor (28, 29). Cabergoline is more effective, restoring ovulatory menses in >90% of women and achieving >90% reduction in tumor size (28, 29). Bromocriptine, however, has a larger volume of safety data (although the data are reassuring for both), and this should be discussed during the pre-conception counselling period. In addition, bromocriptine is cheaper, and some clinicians may elect to use it as its use has not been reported to have an association with heart valve disease. However, it should be noted that there are no published reports of cardiac valve abnormalities in cabergoline-treated pregnant women or their fetuses. The disadvantages with bromocriptine include twice-daily dosing (vs twice weekly with cabergoline), although this may not be strictly necessary, a greater side effect profile, and inferior effectiveness at normalizing prolactin concentrations (30, 31). For women who cannot tolerate bromocriptine, cabergoline should be recommended as 70% of patients who have not responded to bromocriptine respond to cabergoline (32). In those who do not achieve restoration of menses, clomiphene citrate or recombinant gonadotrophin may be considered for ovulation induction (33).

 

Surgical therapy is curative in approximately 70-80% of patients with microadenomas and rarely causes hypopituitarism in expert hands. The cure rate is lower (30%) in patients with macroadenomas, and the risk of hypopituitarism and subsequent infertility is markedly increased (28).

 

All women with prolactinomas should be counselled in the pre-pregnancy period about their potential fertility and pregnancy outcomes to enable informed decision making (34).

 

EFFECTS OF DOPAMINE AGONISTS ON THE DEVELOPING FETUS

 

Bromocriptine has been shown to cross the placenta in human studies (35); cabergoline lacks human data but has been found to do so in animal studies. Current recommendations, therefore, advise that women with prolactinomas discontinue dopamine agonist therapy when they discover that they are pregnant (29). There is a subset of patients in whom this may not apply, e.g., women with macroadenomas, in particular those with an invasive tumor or where it is abutting the optic chiasm. In such cases, the management must be considered on a case-by-case basis.

 

In the majority of cases, in order to limit the exposure time to the developing fetus it is beneficial to know the timing of the normal menstrual cycle. Use of mechanical contraception may facilitate this if used for the first two to three cycles after starting treatment. As a consequence, women will know when they have missed a period, a pregnancy test can be performed in a timely manner and the dopamine agonist can be stopped in cases where a pregnancy is confirmed. This approach aims to limit the time that the fetus is exposed to bromocriptine to 3-4 weeks and cabergoline to around 5 weeks (as a consequence of its longer half-life (21).

 

Such short-term exposure to both bromocriptine and cabergoline is unavoidable but reassuringly pregnancy outcomes with respect to spontaneous abortions, terminations, ectopic pregnancies, pre-term births, multiple pregnancies, and malformations do not differ from the normal population Table 1 summarizes outcome data from 6239 pregnancies following bromocriptine treatment and 968 where the mother took cabergoline (21). There was no increased risk demonstrated by either drug but due to the greater wealth of experience with bromocriptine it is the preferred drug of choice for those wishing to become pregnant according to the European guideline (29).

 

Table 1. Pregnancy Outcomes While Taking Bromocriptine or Cabergoline Compared to the Normal Population

 

Bromocriptine

(n (%))

Cabergoline

(n (%))

Normal (%)

Pregnancies

     Spontaneous abortions

     Terminations

     Ectopic

     Hydatiform moles

6239 (100)

620 (9.9)

75 (1.2)

31 (0.5)

11 (0.2)

968 (100)

73 (7.5)

63a (6.5)

3 (0.3)

1 (0.1)

100

10-15

20

1.0-1.5

0.1-0.15

Deliveries (known duration)

     At term (>37 weeks)

     Preterm (<37 weeks)

4139 (100)

 

3620 (87.5)

519 (12.5)

705 (100)

 

634b (89.9)

71 (10.1)

100

 

87.3

12.7

Deliveries (known outcome)

     Single births

      Multiple births

5120 (100)

 

5031 (98.3)

89 (1.7)

629 (100)

 

614 (97.6)

15 (2.4)

100

 

96.8

3.2

Babies (known details)

     Normal

     With malformations

5213 (100)

5030 (98.2)

93 (1.8)

822 (100)

801 (97.4)

21 (2.4)

100

97

3.0

aEleven of these terminations were for malformations

bFive of these births were stillbirths

 

Long-term follow up studies of children conceived whilst their mothers were taking either bromocriptine or cabergoline are also reassuring, although the numbers are smaller (36-41).

 

Bromocriptine has been used throughout gestation in just over 100 women with no increase in the rate of abnormalities compared to background rates (33, 36, 42).  Of 15 reports of the use of cabergoline throughout gestation (43), 13 healthy infants were delivered at term and another was delivered at 36 weeks’ gestation. There was one intrauterine death at 34 weeks in a pregnancy complicated by severe pre-eclampsia (43). In a study of 25 pregnancies in which cabergoline was continued throughout gestation, the incidence of missed abortion, stillbirth and low birth weight was no different compared to a group of women in whom the cabergoline was not continued. There was also no difference in post-pregnancy recurrence of hyperprolactinemia or tumor remission (44).

 

EFFECT OF PREGNANCY ON PROLACTINOMA SIZE

Prolactinomas can enlarge during pregnancy as a consequence of the progressive increase in serum estrogen levels and discontinuation of dopamine agonists (21). This can lead to tumor volume enlargement with the risk of mass effect and visual field loss. In microprolactinomas, the risk of clinically significant tumor growth is less than 5%. In contrast, patients with macroprolactinomas are reported to have a 15-35% risk (33, 45). This risk can be reduced if the patient undergoes surgery or irradiation prior to the pregnancy.

 

MANAGEMENT FOLLOWING CONCEPTION

 

The risk of tumor expansion is sufficiently rare for microprolactinomas that dopamine agonists can be withdrawn on confirmation of pregnancy. For patients with a macroprolactinoma decisions about cessation of therapy should be decided on a case-by-case basis. For some women with intrasellar/ inferiorly extending macroprolactinomas there may be less concern than for those where the tumor has close proximity to the optic chiasm.  For all women with macroprolactinomas, it is appropriate to undertake a clinical assessment of the patient in each trimester with particular attention to the presence of headache or visual impairment. Where clinical findings are positive it is then pertinent to perform a MRI scan without contrast, and if evidence of tumor expansion either re-introduce a dopamine antagonist or increase the dose (33). If this fails neurosurgery (preferably during the second trimester) or delivery (if the pregnancy is sufficiently advanced, e.g., at >37 weeks’ gestation) may be appropriate (46). If clinical examination or further investigation is reassuring, assessment in each trimester can be re-commenced without changes in treatment. MRI also plays a pertinent role in distinguishing between hemorrhage into a tumor and simple tumor enlargement in those presenting with headache (21).

 

For patients with previous expansion or an invasive macroprolactinoma, options for management need to be carefully considered by an expert in the field. Options include stopping the dopamine agonist, surgical intervention, or continuing the dopamine agonist throughout pregnancy. Clinical assessment with formal visual fields can be justified every 1-3 months, and if there are positive findings an MRI scan without contrast with a view to addition of dopamine agonist, neurosurgery, or delivery where appropriate should be undertaken (Figure 1).

Figure 1. Schematic for the Management of Both Micro- and Macroprolactinomas During pregnancy.

The monitoring of prolactin levels yields no diagnostic benefit in the pregnant woman with a prolactinoma as it bears no correlation to tumor growth. In most cases, serum prolactin concentrations will rise with gestation regardless of the presence of a prolactinoma, and it is also possible for tumor size to increase without a simultaneous rise in prolactin level.

 

BREASTFEEDING

 

There are no contraindications to breastfeeding in women with prolactinomas. To date, there is no evidence to suggest a significant increase in prolactin levels or symptoms suggestive of tumor enlargement in lactating women (20). In many women dopamine agonists do not need to be reintroduced during this period. There has even been successful lactation in women with previous surgical resection of a prolactinoma whose prolactin level had not increased during pregnancy. In a study carried out by Narita et al (47), a third of such women whose prolactin levels had not risen above 30ng/mL during pregnancy were still able to lactate.

 

Growth Hormone

 

INTRODUCTION

 

Pituitary growth hormone (PGH), expressed by the somatotroph cells of the anterior pituitary, has two main isoforms: 22K-GH and 20K-GH. The 22K-GH isoform is the main circulatory isoform in normal men, nonpregnant women, and in patients with acromegaly (3). In the nonpregnant state the hypothalamus secretes growth hormone releasing hormone (GHRH) which stimulates the production of growth hormone from the pituitary in a pulsatile fashion. PGH subsequently stimulates insulin-like growth factor 1 (IGF-1) release from the liver which is required for metabolism and growth promoting effects. Somatostatin inhibits growth hormone, providing negative feedback and there is also inhibition of PGH production by IGF-1 which prevents the somatotrophs from releasing PGH and encourages the release of somatostatin from the hypothalamus. When too much PGH is secreted by a pituitary adenoma acromegaly is diagnosed. This is confirmed by an elevated age- and sex- corrected serum IGF-1 and failure of PGH to suppress to <0.4ug/L following an oral glucose tolerance test in the presence of a pituitary mass on MRI.

 

Estrogen blocks the effects of PGH on the liver, which explains why in order to achieve equivalent levels of IGF-1 women need to secrete higher levels of PGH than men (3). During normal pregnancy the rise in estrogen generates a PGH resistant state and, as such, there is an initial drop in IGF-1 levels. As the placenta grows it begins to secrete placental growth hormone (PLGH),  a single chain protein that is structurally very similar to the PGH isoform 22K-GH (6). PLGH is initially secreted at 10 weeks (48) eventually overcoming the resistance to growth hormone and resulting in an increase in IGF-1 levels. Eventually, PGH becomes suppressed as a consequence of negative feedback such that by the last week of pregnancy PLGH and IGF-1 levels peak whilst PGH is almost undetectable (49).

 

DIAGNOSIS OF ACROMEGALY DURING PREGNANCY

 

The similarities in the structure of PGH and PLGH present challenges for the diagnosis of acromegaly and monitoring of biochemical control in pregnant women with pre-existing acromegaly. Both GH peptides are composed of a single polypeptide chain with 2 disulfide bridges and 191 amino-acids. Conventional assays for GH cannot usually distinguish between the two, and therefore the confident diagnosis of acromegaly is usually reserved until after delivery. When a diagnosis during pregnancy is desired, there are a number of factors that may be considered. The pulsatile nature of PGH may help to establish a true increase in PGH as opposed to PLGH, if pulsatility can be demonstrated. Response to hypoglycemia is enhanced in PGH but decreased in PLGH, and response to arginine is enhanced in PGH and varies in PLGH (6).

 

A high serum IGF-1 concentration prior to midgestation may also be suggestive of acromegaly as levels are not expected to rise until after this stage of pregnancy. Highly specific assays may also be used to demonstrate raised PGH during the third trimester if raised above the expected 1ug/L, which is expected that that stage of normal pregnancy (3).  

 

If a new diagnosis of acromegaly is suspected, imaging of the pituitary with MRI may be warranted (50).

 

FERTILITY IN WOMEN WITH ACROMEGALY

 

It is reported that between 40-84% of acromegalic women suffer with gonadal dysfunction the causes of which are multifactorial (51-55). Mass effect of the adenoma on the gonadotrophic cells may cause gonadotrophin deficiency. In addition, compression of the stalk may reduce levels of GnRH and contribute towards an increase in PRL levels (55). PRL secretion may also be increased in cases of a mixed GH and PRL-secreting tumor. Other contributory features include the effect of GH and IGF-1 on the ovaries (inhibition of GnRH and direct ovarian inhibition) and polycystic ovarian syndrome (54, 55).

 

EFFECT OF PREGNANCY ON ACROMEGALY

 

In a recent review of 46 women with acromegaly in pregnancy, 39 received surgical treatment prior to conception, and 21 who were receiving medical treatment and drugs were continued on therapy if the risk of treatment interruption was considered to outweigh the risk of continuation (56). In this and other similar reviews (57-59), this is considered to “reflect the overall improvement in both diagnosis and treatment of patients with acromegaly in the last decades” (3). The adenomatous somatotrophic cells themselves appear to be resistant to the IGF-1 inhibitory feedback demonstrated in pregnancy, as demonstrated by continuous PGH production during pregnancy (using specific placental assays) (3, 55). Despite this, biochemical escape is often witnessed with IGF-1 levels often remaining unchanged or decreasing during pregnancy (60). This is thought to be secondary to reduced IGF-1 generation in the context of hepatic resistance to GH action in a high-estrogen environment (61). However, the degree of hepatic resistance varies from patient to patient, and this most likely explains the variability in clinical course often encountered (59, 62, 63). As might be expected, it is not uncommon for IGF-1 levels to increase rapidly post-delivery (or following termination of pregnancy), and thus vigilance must be exhibited by the clinician at this stage.

 

A multicenter study carried out by Caron et al. in 2010 retrospectively studied 59 pregnancies in 46 women with GH-secreting pituitary adenomas. In 3 out of 27 cases in whom adenoma volume was assessed by MRI 6 months post-delivery, there was an increase in size (11.1%), and two affected women had visual complications. Adenoma volume was stable in 22 women (81.5%) and decreased in two cases (7.4%) (56). A number of subsequent published reports have mirrored these figures, demonstrating that the risk of tumor expansion is small (58, 59, 64). In a retrospective analysis of 31 pregnancies in 20 patients with acromegaly, Jallad et al. observed symptomatic pituitary tumor enlargement and subsequent surgical intervention in 3 of 31 (9.6%) pregnancies. It is worth noting, however, that in these cases the patients had visual field impairment at the initiation of pregnancy (61). In a recent series of 17 pregnancies in 12 women with acromegaly no patients developed new visual field abnormalities or symptoms suggestive of tumor expansion (60). In a systematic review including 273 pregnancies in 211 women with acromegaly 9% of women experienced tumour growth (65).

 

EFFECT OF ACROMEGALY ON THE NEONATE

 

The presence of elevated PGH levels is not thought to effect the neonate as there is currently no evidence that PGH crosses the placenta or influences placental development (55). Considering the increased risk of the mother developing worsening diabetes or hypertension or gestational diabetes mellitus (GDM) or hypertension, it is important to consider the risk of macrosomia and microsomia respectively (3, 65).

 

METABOLIC AND CARDIOVASCULAR COMPLICATIONS

 

Jallad et al. described the diabetogenic effects of PLGH and PGH which include hyperinsulinemia, decreased insulin-stimulated glucose uptake and glycogen synthesis, and impairment of the ability of insulin to suppress hepatic gluconeogenesis (61).  As a consequence, acromegalic women are at higher risk of developing GDM or suffering worsening control of pre-existing diabetes mellitus (65). The risk of developing GDM in women with acromegaly appears to be in the region of 5.5%-10% (55). This figure is slightly higher than the rate of gestational diabetes in the general UK population, which is reported as 5% (66). The risk of gestational hypertension is also approximately 14% (6, 61). In a retrospective study, this was associated with poor GH/IGF-1 control prior to pregnancy (56).

 

MANAGEMENT APPROACH

 

In patients diagnosed prior to pregnancy with microadenomas or intrasellar macroadenomas, transsphenoidal surgery is the most frequent surgical approach and the majority will achieve remission. For those with residual disease repeat surgery can be considered. In cases of persistent residual disease, either a somatostatin analogue, dopamine agonist, or pegvisomant may be used. For those with parasellar disease, debulking surgery +/- introduction of medical management should be considered. It is pertinent for patients taking either a long-acting somatostatin analogue or pegvisomant to aim to stop treatment two months prior to attempting to conceive; short-acting octreotide can be used until conception (67). The advantages and disadvantages of stopping the medication should be discussed with the patient and the ultimate decision be made on a case-by-case basis. In addition, assessment of disease activity, comorbidities and fertility status should be undertaken in the pre-pregnancy period to facilitate informed decision making (34).

 

For those patients diagnosed in pregnancy, a more conservative approach is advised with introduction of medical management in those patients who require tumor or headache control. When no improvement is seen with medication, transsphenoidal surgery should be considered (Figure 2). Routine measurements of serum concentrations of GH and IGF-1 or routine MRI are not recommended during pregnancy (67).

Figure 2: Schematic for the Management of Acromegaly During Pregnancy

MEDICAL THERAPY

 

Somatostatin Analogues

 

Due to a historic lack of data, awareness that they can cross the placenta, and identification of somatostatin receptors in the placenta and fetal pituitary, somatostatin analogues are typically stopped two months prior to conception. However, due to the long-acting nature of these drugs and the need for some women to continue therapy throughout part of the entirety of gestation, data are starting to accumulate. In a recent study by Vialon et al. 67 pregnancies in 62 women treated with somatostatin analogues during pregnancy were compared with 74 pregnancies in 65 women not treated medically (68). This study included 36 pregnancies in which a somatostatin analogue was taken in the first trimester of pregnancy. Rates of malformation were not reported above the background population and no significant impact on maternal and fetal outcomes were identified. While the paucity of data continues to support withdrawal of this group of medications prior to pregnancy in the majority these data provide reassurance for women who may conceive on these drugs or may require treatment throughout pregnancy or reintroduction during pregnancy.

 

Dopamine Agonists

 

The considerations regarding the safety use of cabergoline in women with prolactinomas discussed above also apply to those with acromegaly.

 

Growth Hormone Receptor Antagonists

 

Pegvisomant, a GH receptor antagonist, has only been used in a handful of cases and as such its safety in pregnancy has not been established. Published reports include a woman who had undergone in vitro fertilization and intra-cytoplasmic sperm injection following monotherapy with pegvisomant. The drug was discontinued after conception and a healthy neonate was born at 38 weeks by elective cesarean section and remained well at 1 year (69). Another woman treated with pegvisomant throughout gestation gave birth to a healthy girl at 40 weeks by cesarean section after failure to progress following spontaneous labor. In this case there was minimal demonstration of movement of pegvisomant across the placenta, and no evidence of substantial secretion into the breast milk (70). A 2015 review of current safety data compiled in the Pfizer Global Safety Database included 27 women exposed to pegvisomant, 3 of whom continued treatment throughout pregnancy. In this report there was no evidence to suggest adverse outcomes, but it was acknowledged that numbers remain too small to provide clear evidence (71). In a more recent series of four pregnancies in three women with pegvisomant used prior to or at the time of conception no significant maternal or fetal complications were reported (72).  

 

TSH-Secreting Adenomas

 

There are only a small number of cases reported of women with TSH-secreting adenomas, four diagnosed prior to pregnancy and two after pregnancy (summarized in Table 2). All cases had positive outcomes with a variety of treatment strategies applied.

 

In 1996 Caron et al described a case in which a 31-year-old previously infertile woman was being treated with good biochemical and radiological responses with continuous subcutaneous infusion of octreotide for a TSH-secreting macroadenoma. She was found to be pregnant after four months of treatment, and the octreotide was consequently stopped. During the pregnancy TSH concentrations increased and there was symptomatic and radiological evidence of tumor expansion. Octreotide was restarted with rapid improvement in clinical signs and biochemical markers. Immunoreactive octreotide was detected in the umbilical cord but demonstrated rapidly decreasing concentrations and was undetectable at 40 days. This demonstrated evidence of maternal-fetal transfer of octreotide. Notably, despite this the usual physiological changes in neonatal thyroid parameters were not disturbed (73). In 2002 Blackhust et aldescribed the first documented twin pregnancy in a 21-year-old patient with a TSH-secreting adenoma who had been treated with propylthiouracil to control thyrotoxicosis and cabergoline for hyperprolactinemia. She subsequently underwent transsphenoidal surgery due to resistant thyrotoxicosis, followed by long-acting octreotide and postoperative radiotherapy. This patient reported that she was pregnant during the course of radiotherapy and at this stage a decision was made to continue both the octreotide, complete the course of radiotherapy (although it is not documented for how many weeks the radiotherapy was continued it is likely to have been <9 weeks), and substitute cabergoline for bromocriptine (74). In 2003 Chaiamnuay et al. described at 39-year-old woman who was treated with propylthiouracil and bromocriptine; on confirmation of pregnancy (after 5 months of treatment) the dose of propylthiouracil was reduced and the bromocriptine was continued. At 27 weeks she developed clinical and radiological signs of tumor expansion and transsphenoidal surgery was performed with subsequent normalization of TSH and prolactin (75). There are two cases described in the literature where the mother was diagnosed during the pregnancy: both declined surgery and were medically managed. Both women had uneventful pregnancies and delivered healthy babies (76, 77).

 

Perdomo et al described a 21-year-old woman who initially underwent transsphenoidal surgery and was subsequently started on octreotide when she relapsed 17 months postoperatively. The octreotide was stopped when she was found to be pregnant (78).

 

Each pregnancy was uneventful with no congenital abnormalities reported. Although the number of cases is small, previous positive outcomes give some reassurance that pregnancy should not be entirely contraindicated in women with TSH-secreting adenomas. Treatment options include medical management with bromocriptine or octreotide, conservative medical control of thyrotoxicosis using propylthiouracil or carbimazole, or surgical management using the transsphenoidal approach. Emphasis lies on the importance of individualized treatment with the support of the multidisciplinary team. As with all pituitary tumors in pregnancy, close follow-up is paramount with monitoring of visual fields for evidence of tumor expansion.

 

Table 2. Summary of Pregnancies in Women with TSH Adenomas

Tumor

Diagnosis

Prior TS

Previous medical therapy

TS in pregnancy

Medical therapy in pregnancy

Delivery

TSH-secreting macroadenoma (73)

Prior to pregnancy

No

Oct-CSI (300ug/day)

No

Oct-CSI (300ug/day) -1st month and 3rd trimester

ECS

TSH-secreting macroadenoma with hypothalamic disconnection (74)

Prior to pregnancy

Yes

PTU and CBL prior to TS

 

Oct-LAR post TS

 

Post-TS RT

No

Br throughout gestation

 

RT (<9wks)

 

Oct-LAR- throughout gestation

NA

TSH-secreting macroadenoma with hypothalamic disconnection (75)

Prior to pregnancy

No

PTU 50mg TDS and Br 2.5mg OD

Yes

Reduced dose of PTU

 

Br continued

ECS

TSH-secreting microadenoma (76)

24 weeks gestation

No

N/A

No

PTU 150mg/day in 3 divided doses from 28 wks

 

NVD

TSH-secreting macroadenoma (77)

20 weeks’ gestation

N/A

N/A

No

PTU 100mg TDS until end of 2nd trimester then carbimazole 30mg/day until delivery

NVD

TSH-secreting macroadenoma (78)

Prior to pregnancy

Yes

Octreotide

No

Octreotide stopped

NVD

Abbreviations: NA- Not available; TS- Transsphenoidal surgery; Oct-CSI- Octreotide Subcutaneous Infusion; CBL- Cabergoline; PTU- Propylthiouracil; Oct-LAR- Octreotide long-acting repeatable formulation; RT- Radiotherapy; Br- Bromocriptine; NVD- Normal vaginal delivery; ESC-Elective caesarean section; OD- once daily

 

Non-Functioning Pituitary Adenomas and Gonadotrophin-Secreting Adenomas

 

The majority of clinically non-functioning pituitary adenomas (NFPAs) in non-pregnant individuals are demonstrated to be gonadotrophin-cell adenomas when exposed to immunostaining (79). The main concern when a patient with an NFPA becomes pregnant, by virtue of the fact that there is an absence of hormone excess, is the theoretical risk of tumor enlargement causing compressive symptoms or displacement of the tumor causing compressive symptoms secondary to lactotroph hyperplasia. It is thought that in such cases patients may respond to dopamine agonist-mediated reduction of lactotroph hyperplasia. Masding et al described such as case in which a pregnant woman developed a visual defect at 18 weeks’ gestation and was demonstrated to have a NFPA which extended into the suprasellar region causing compression of the optic chiasm. This patient responded well to bromocriptine with resolution of the visual field defect (80). A UK national cohort study identified 16 cases of NFPA in pregnancy over a 3-year period, giving an incidence of 0.59 cases/100,000 maternities (45).The numbers were small, but there was no apparent increase in the rate of gestational hypertension or preterm labor in women with NFPA. However, they did have higher rates of operative delivery and induction of labor compared to women without pituitary tumors, a finding that was not reported in women with macroprolactinomas (45).

 

Gonadotrophin-secreting adenomas in pregnancy are extremely rare, and there are only three cases reported in the literature. The first reports a 29-year-old woman with a microadenoma and ovarian hyperstimulation. She was treated with bromocriptine with subsequent normalization of ovarian size and went onto conceive naturally (81). The second case also had ovarian hyperstimulation secondary to a gonadotrophin-secreting macroadenoma. This patient underwent surgical removal of the tumor with consequent normalization of FSH, LH and estradiol and natural conception (82). Both of these women delivered healthy newborns. The third presented with infertility, during work-up she was found to have an isolated elevated LH, MRI revealed an enlarged sella turcica and intrasellar mass following which she underwent transsphenoidal resection. Menses were restored within 16 days post operation and the patient reported being pregnant three months later. She was still under follow-up at the time the case report, hence, pregnancy outcomes were not reported(83).

 

Pituitary Apoplexy

 

Pituitary apoplexy is a rare clinical syndrome characterized by headache, visual disturbance, and altered mental status. It is caused by rapid expansion of the contents of the sella turcica as a consequence of hemorrhage or infarction into a pre-existing pituitary adenoma or within a physiologically enlarged gland. Pituitary apoplexy in pregnancy is extremely rare but may present as a medical emergency (due to risk of hormonal insufficiency) and may represent the first presentation of an underlying adenoma. It is therefore pertinent to consider apoplexy as a differential diagnosis for sudden onset headache and/or visual disturbance in pregnancy.

 

In a recent review of the clinical and biochemical characteristics of male and female patients presenting with pituitary apoplexy, it was found to be the first presentation of pituitary disease in 38/52 (73%) of patients. One quarter of the women (7/27) in this review experienced apoplexy in the peripartum period (84). It is thought that rapid expansion of a pituitary adenoma during pregnancy secondary to increased estradiol may be one explanation for the predisposition to apoplexy in the peripartum period (84).

 

There are approximately 40 cases of pituitary apoplexy during pregnancy described in the literature.  The most common presenting symptoms in these patients are headache (95%) and visual disturbance (63%), comparable to the literature for non-pregnant patients (84-89). Hormonal replacement is required in approximately 60% of women with apoplexy in pregnancy making it important that pituitary hormone profiles are checked (85).

 

The acute management of apoplexy includes fluid and electrolyte replacement as well as corticosteroid replacement where indicated. The treatment given to the women in the literature ranges from conservative management, the use of dopamine agonists (bromocriptine and cabergoline) as well as surgical intervention. Women have also undergone varying modes of delivery, approximately 64% by vaginal delivery and approximately 36% by caesarean section, demonstrating the importance of an individualized management plan (85, 87-89).

 

Reassuringly, in the pregnancies described in the literature to date, if apoplexy is promptly diagnosed and managed, there appears to have not been any negative consequences for the fetus.

 

Sheehan’s Syndrome

 

Sheehan’s syndrome occurs as a consequence of ischemic pituitary necrosis secondary to severe postpartum hemorrhage with hypotension and shock (90).  Possible mechanisms include vasospasm, thrombosis, and vascular compression of the hypophyseal arteries. Enlargement of the gland, disseminated intravascular coagulation, and autoimmunity have also been implicated in the pathophysiology of the condition (91). The majority of patients have an empty sella on CT or MRI (90).

 

Advances in obstetric care and the availability of rapid transfusion has resulted in a significant decline in the incidence of Sheehan’s syndrome in the Western world (91). Although rare, Sheehan’s syndrome may present as a medical emergency, and suspicion should be raised in women that are hypotensive, tachycardic, hypoglycemic, vomiting, or elicit signs of diabetes insipidus despite adequate resuscitation following post-partum hemorrhage (90, 91).

 

It is thought that the degree of ischemia and necrosis dictates the clinical course, and thus in patients with lesser degrees of ischemia the syndrome may present in a more insidious manner. Such patients may present with failure to lactate, persistent amenorrhea, light-headedness or fatigue, genital and axillary hair loss, dry skin, cold intolerance, and other symptoms of hypopituitarism. In such patients diagnosis may be delayed for over 10 years post-partum (92). In some women only partial hypopituitarism is experienced, and they may therefore go onto have further spontaneous pregnancies.

 

For those women in whom an acute form of Sheehan’s syndrome is suspected, investigations should include serum electrolytes, cortisol, prolactin, and free thyroid hormones, and possibly ACTH. Thyroxine has a half-life of seven days and so may be normal in the initial period, prolactin levels are usually low, as are ACTH and cortisol levels. Fluid replacement and stress doses of corticosteroids should then be given without delay and additional pituitary testing and subsequent therapy should be delayed until after recovery.

 

Hypopituitarism

 

The causes of hypopituitarism are vast and include neoplasms, vascular events (pituitary apoplexy, Sheehan’s syndrome, intrasellar carotid artery or subarachnoid hemorrhage), trauma, medications, infiltrative/inflammatory disease, and treatment of sellar/parasellar and hypothalamic disease such as surgery or radiotherapy. The disorder is characterized by the deficiency of one or more of the hormones secreted by the pituitary gland.

 

In those patients with gonadotrophin deficiency, fertility is often impaired making natural conception rare. Advances in fertility treatment, however, have led to increased pregnancy rates usually with the support of a reproductive endocrinologist. In such cases and with appropriate hormone replacement women can undergo uneventful pregnancies.

 

An increased demand for thyroid hormones is observed during pregnancy. This is thought to be explained by increased plasma volume and thus volume of distribution, fetal dependence on maternal thyroid hormones, increased human chorionic gonadotropin (hCG) levels (which acts as a weak TSH agonist), increased levels of total binding globulin, and increased thyroid hormone degradation. In patients recognized to have hypopituitarism, an appropriate rise in TSH may not be achieved and so it is recommended that clinicians monitor serum free T4 or total T4 levels every 4-6 weeks so that doses of thyroxine can be adjusted as required to maintain free T4 in the normal range for pregnancy.

 

n increase in glucocorticoid dose throughout gestation is not routinely recommended but rather monitoring of clinical symptoms of signs to assess for signs of glucocorticoid deficiency. Additional supplementation is, however, recommended during the active stage of labor and delivery. Current Endocrine Society guidelines suggest 50mg intravenous hydrocortisone during the second stage of labor and 100mg every 6-8 hours during caesarean section (93). The NICE guideline on the management of medical disorders in labor recommends slightly less glucocorticoid replacement in those planning a vaginal birth (50mg 6-hourly). For those having a caesarean section, this guideline suggests individualized glucocorticoid replacement strategies that will depend upon whether the woman has received hydrocortisone in labor, i.e., 50mg intravenous hydrocortisone when starting anesthesia if she has and 100mg hydrocortisone if she has not. Hydrocortisone replacement (50mg 6-hourly) is then recommended until 6 hours after birth (94). Mineralocorticoid replacement is not required in these patients.

 

Hydrocortisone is non-fluorinated glucocorticoid and therefore a good preparation to use for physiological glucocorticoid replacement in pregnancy because it is degraded by the placental enzyme 11beta-hydroxysteroid dehydrogenase-2 and thus limited quantities of the drug cross the placental barrier (95). Prednisolone is also a non-fluorinated glucocorticoid and therefore has limited passage across the placenta (96). Dexamethasone, however, is a fluorinated glucocorticoid and thus poorly metabolized by this enzyme, hence >50% will cross the placental barrier (97). Steroids with low transplacental transfer are favored to reduce the risk of neonatal adrenal suppression and neurocognitive/neurosensory disorders in childhood which have been associated with the fluorinated preparations (98).

 

Growth hormone is not currently approved for use during conception or pregnancy, and thus the current European Guidelines recommend its discontinuation during pregnancy (93). However, there is ongoing debate as to whether women with GH deficiency may benefit from such treatment. In an observational study of 201 pregnancies of patients with GH deficiency and hypopituitarism, two thirds of the women underwent fertility treatment to achieve pregnancy; 7% stopped GH replacement prior to pregnancy, 40% once the pregnancy was confirmed, and 25% at the end of the second trimester, while 28% continued the treatment throughout pregnancy. A healthy child was delivered in 80% of cases and there was no relationship between the complications and the treatment patterns (99). In a recent study reporting outcomes of 47 women with GH deficiency exposed to growth hormone between conception and delivery no new safety signals relating to GH were identified (100).

 

Lymphocytic Hypophysitis

 

Lymphocytic Hypophysitis (LH) is characterized by infiltration of the pituitary by lymphocytes and plasma cells which cause destruction of the normal parenchyma (101). The disorder is thought to have an autoimmune basis and is commonly associated with pregnancy or the post-partum period. Most patients present in the last month of pregnancy or within the first 2 weeks post-partum (101). Plausible explanations for the association between LH and pregnancy include, firstly, a change in the pattern of blood flow from predominantly systemic as opposed to portal circulation, and thus increased exposure to the immune system. Secondly, the theory that there are common autoantibodies to both the pituitary and placenta which may be implicated, although it should be noted that conflicting results and poor specificity impair the clinical usefulness of checking anti-pituitary antibodies for diagnostic purposes in the clinical setting(102, 103).

 

Patients may present with symptoms of mass effects, hypopituitarism, hyperprolactinemia, or diabetes insipidus (101). LH may be mistaken for Sheehan’s syndrome in the postpartum period, and absence of obstetric hemorrhage can be a useful distinguishing feature. A clinical diagnosis can often be made, particularly where there is a temporal relationship with pregnancy, and can be supported by typical appearances on MRI including symmetrical enlargement of the gland, a thickened but rarely displaced stalk, intact sellar floor, and pre-contrast homogeneity of the mass (101). Hypopituitarism disproportionate to the size of the lesion, preferential impairment of ACTH secretion, and the presence of anti-pituitary autoantibodies are also supporting findings (102).

 

Management is usually symptomatic including hormone replacement and focus on reducing the size of the mass. Due to the risks involved, surgical intervention is reserved for those patients with significant compressive symptoms. Glucocorticoids are favored by some to reduce inflammation, and in some cases their use has led to recovery of pituitary function (102). Other treatment options include azathioprine when pregnant and methotrexate or radiotherapy in non-pregnant individuals.

Posterior Pituitary Gland Physiology

 

The serum osmolality threshold at which arginine vasopressin (AVP) is secreted is reduced in pregnancy by approximately 5-10 mOsm/kg (dropping from 285 to 275 mOsm/kg). Consequently, pregnant women experience thirst and release AVP at lower plasma osmolarities than non-pregnant women (104, 105). These changes are thought to be related to increased levels of human chorionic gonadotrophin (105). The placenta also plays an important role in water homeostasis during pregnancy; vasopressinase, a trophoblast-derived aminopeptidase, is an enzyme produced by the placenta which inactivates endogenous vasopressin (106). Maximum concentrations of vasopressinase are reached in the third trimester, correlate with placental weight, and are higher in multi-fetal pregnancies (107). A compensatory increase in AVP synthesis and secretion is therefore observed (108).

 

The effect of AVP has been demonstrated to be mediated by one or more of a family of water channels called aquaporins (AQPs). Their discovery has facilitated our understanding of the modes of transport across the renal tubules and collecting ducts (109). Each member of the AQP family has a different sensitivity to AVP. Abnormalities in the AQP2 channel have been implicated in the pathophysiology of nephrogenic DI. An increase in the expression of AQP2 in the renal medulla of pregnant rats has recently been demonstrated (110).

 

A progressive increase in plasma oxytocin levels is observed during uncomplicated pregnancy. Levels rise dramatically during labor peaking in the second stage (111, 112).  A positive feedback loop is executed, oxytocin secretion being stimulated by uterine contraction and the oxytocin then simulating further contractions (111). Uterine sensitivity to oxytocin increases with a rise in oxytocin receptors in the myometrium. Hypophysectomy does not alter onset of labor, indicating that oxytocin provides only a facilitatory role (112). Levels increase further during suckling (113).

 

Diabetes Insipidus (DI)

 

DI is thought to complicate up to 1 in 30,000 pregnancies and presents with polyuria, polydipsia, and dehydration (114). The presentation may involve exacerbation of previously overt or subclinical cranial or nephrogenic DI (as a consequence of increased clearance of AVP by placental cystine aminopeptidase, lower osmostat for vasopressin release and elevation of vasopressinase levels) or may develop de novo in pregnancy.

 

When investigating pregnant women for diabetes insipidus (DI) one must be aware that sodium levels may be lower than expected (115). Traditional water deprivation testing (requiring 5% weight loss) should be considered on a case-by-case basis due to the risk of hypernatremia, neurological disorders, and fetal harm (116). Providing there is close medical surveillance, a water deprivation test can be performed on women with mild symptoms. It is also reasonable to use a trial of treatment with desmopressin (DDAVP) to establish whether this can correct urinary concentrating ability.

 

During pregnancy, DI tends to be broadly categorized into 3 main subtypes; central DI, nephrogenic DI and transient DI of pregnancy.

 

CENTRAL DI

 

Patients with central DI fail to release AVP from their posterior pituitary. This may occur spontaneously in pregnancy or in the postpartum period secondary to Sheehan’s syndrome, an enlarging pituitary adenoma, pituitary apoplexy, lymphocytic hypophysitis, or with the development of other conditions such as Langerhans cell histiocytosis (110, 116). The most common causes of central DI are listed in Table 3. In a series of 55 cases of central DI described by Takeda et al. there were 19 tumoral lesions; 14 cases of Sheehan’s syndrome; 10 of pituitary apoplexy; and 12 of lymphocytic hypophysitis, infundibuloneurohypophysitis and stalkitis (103).

 

The use of chlorpropamide (a sulfonylurea) for the treatment of partial central DI is not advised as it may cross the placental barrier and cause hypoglycemia in the fetus. The AVP analogue desmopressin (DDAVP) is the first-line treatment for DI in pregnancy as it is resistant to vasopressinase (116).  To date, its use has been demonstrated to be safe for both mother and fetus (104, 106, 117). For women initiated on DDAVP prior to pregnancy it can, therefore, be continued. It should be noted that some women require higher doses during pregnancy and that in such circumstance the dose should be titrated back to pre-pregnancy dose soon after delivery. DDAVP transfers minimally into breast milk and is poorly absorbed from the gastrointestinal tract, and thus should not adversely affect the infant’s water metabolism (115).

 

Table 3. Causes of Central Diabetes Insipidus

Primary

Idiopathic

-

Genetic

Wolfram syndrome (diabetes insipidus, diabetes mellitus, optic atrophy and deafness

Neurohypophyseal diabetes insipidus

Developmental Syndromes

Septic-optic dysplasia

Secondary/ Acquired

Trauma

Head injury

Post-surgery

Post radiotherapy

Vascular

Carotid aneurysm

Cavernous sinus thrombosis

Tumor

Craniopharyngioma

Germinoma

Metastases

Pituitary adenomas

Inflammatory

Sarcoidosis

Langerhans cell histiocytosis

Meningitis/ Encephalitis

Infundibuloneurohypophysitis

Guillain-Barre Syndrome

Lymphocytic hypophysitis

Infection

Tuberculosis

Fungal diseases

Post-Partum

Sheehan’s syndrome

Pituitary apoplexy

 

NEPHROGENIC DI

 

Nephrogenic DI is caused by resistance to antidiuretic hormone and water restriction is the first line treatment (118). The risks of using medical therapy must be carefully considered. Thiazide diuretics are not routinely recommended in pregnancy due to the risk of electrolyte imbalance, jaundice, and thrombocytopenia in the neonate. There is also a risk of reducing plasma volume which may pose a challenge in situations where utero-placental insufficiency arises, for example, in pre-eclampsia (110), however, they can be used if the maternal and fetal benefits are thought to outweigh the risks. Non-steroidal anti-inflammatory drugs should not be used in the third trimester of pregnancy (110).

 

TRANSIENT DI OF PREGNANCY

 

Transient DI of pregnancy (also called gestational DI) is rare, occurring in between two to four in 100,000 pregnancies (114). It most commonly develops at the end of the second or third trimester and is caused by excessive placental vasopressinase activity. It is more common in multi-fetal pregnancies because the vasopressinase activity is proportional to placental weight. It may also occur in cases of placental abruption which can result in elevated vasopressinase levels (119, 120).

 

Vasopressinase is metabolized in the liver and thus higher concentrations of the enzyme are observed in women with hepatic dysfunction. Both transient liver disease (acute fatty liver of pregnancy, hemolysis, elevated liver enzymes and low platelets (HELLP) syndrome and preeclampsia) and chronic liver diseases may result in increased circulating vasopressin. It is therefore important to check for liver dysfunction in women with a new diagnosis of DI during pregnancy (121). In a series of 50 patients described by Takeda et al of pregnancy-associated DI due to liver pathologies just over half (n=27) developed DI during pregnancy associated with pre-existing liver or coincidental diseases; 15 developed DI associated with acute fatty liver of pregnancy, and 8 with HELLP syndrome (103). DDAVP is the treatment of choice.

 

In patients with idiopathic or central DI, oxytocin levels are normal and labor may begin spontaneously (122). The oxytocinergic pathways, however, may be affected in DI which occurs secondary to trauma, infiltrative disease, or neoplasm, and this may result in poor progression of labor and uterine atony.

 

Syndrome of Inappropriate Antidiuretic Hormone (SIADH)

SIADH is rare in pregnancy. In a series of 18 cases of hyponatremia in pregnancy, seven fit the criteria for SIADH (123). SIADH has been reported in a small number of cases with pre-eclampsia but the mechanism remains unclear (124).

 

ADRENAL DISORDERS IN PREGNANCY

 

The maternal hypothalamo-pituitary-adrenal axis undergoes significant changes in pregnancy. A rise in cortisol is observed partly due to estrogen-stimulated elevation corticosteroid-binding globulin, and also because the placenta releases corticotropin-releasing hormone (CRH) during the second and third trimester which stimulates both the maternal pituitary and adrenal glands  (125, 126). A positive feedback mechanism is then initiated as maternal cortisol stimulates placental CRH synthesis leading to a further increase in cortisol levels (127). The diurnal secretion is maintained during pregnancy despite these changes. The fetus is protected from excess glucocorticoid exposure by the action of placental 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) which inactivates 80-90% of cortisol into cortisone (126).

 

The renin-angiotensin-aldosterone system (RAAS) also undergoes changes in pregnancy. Renin levels rise early in pregnancy as a consequence of extrarenal release from the ovaries and maternal decidua (128). In addition, estrogen simulates angiotensinogen synthesis in the liver resulting in increased levels of angiotensin II (129). Angiotensin-converting enzyme (ACE) levels decline in pregnancy (130). Aldosterone levels rise up to 10-fold by term (131). Despite these changes, blood pressure is often decreased for most of pregnancy, returning to baseline by delivery. This is thought to be the result of reduced responsiveness to angiotensin II in the pregnant state (130, 132). Other theories include increased progesterone and prostacyclin concentrations during pregnancy which may decrease angiotensin II sensitivity, and the monomeric state of the angiotensin receptor (AT1) in pregnancy which renders it less active (133, 134).

 

Cushing’s Syndrome During Pregnancy

 

Cushing’s syndrome (CS) during pregnancy is rare, with fewer than 200 cases reported in the literature (135). However, it is associated with high maternal and fetal morbidity and so an understanding of its management is important. Fertility is generally reduced in women with CS as a result of suppression of gonadotrophin secretion (136). This is one explanation for the fact that adrenal adenomas are more commonly found to be the cause of CS in pregnancy than in non-pregnant women (40-60% vs 10-15% of cases, respectively), although a more favored theory is that adrenal adenomas associated with Cushing’s syndrome may express the LH receptor which then responds to pregnancy-induced hCG secretion. In pregnancy Cushing’s Disease (CD) accounts for 15-40% of cases in comparison to non-pregnant patients where the figure is closer to 70% (137). Adrenal carcinomas and ectopic ACTH secretion are rare causes of CS in pregnancy, accounting for less than 10% of cases (137).

 

INVESTIGATIONS

 

Clinical diagnosis is more challenging in pregnancy because some of the signs of hypercortisolism overlap with clinical signs observed in a normal pregnancy. These include central weight gain, fatigue, emotional lability, glucose intolerance, hypertension, and edema. Useful differentiating features may include muscular weakness, purple striae, and osteoporosis, i.e., the more catabolic features of Cushing’s syndrome (136).

 

Laboratory investigations of CS are also altered in pregnancy. Serum and urinary cortisol concentrations are frequently high in normal pregnancies and the cortisol may fail to suppress during an overnight dexamethasone suppression test (138). Urinary free cortisol can only be relied upon to distinguish between CS and normal pregnancy if it is more than three times the upper range of normal, particularly in the second and third trimesters (139, 140). In contrast to a normal pregnancy, however, the circadian rhythm is lost in CS and this can be useful when confirming a diagnosis. Changes in salivary cortisol during pregnancy are less marked and night time salivary cortisol has been proposed as a potential diagnostic tool. Trimester-specific ranges have been defined with high sensitivity and specificity (141). However, this test is not available in some locations.

 

Placental secretion of ACTH and CRH may prevent the expected suppression of ACTH in women with adrenal CS, adding to the diagnostic challenge (138). High-dose dexamethasone suppression tests, CRH testing, desmopressin testing, and petrosal vein sampling have not been performed in sufficient numbers during pregnancy to be able to draw firm conclusions regarding their reliability (135). CRH has been used in a small number of cases of pregnant women with no ill effects. However, in late gestation it has the potential risk of inducing premature labor as CRH has been demonstrated to enhance the contractile response of the myometrium to oxytocin in the pregnant woman, and has been implicated as a contributory factor in the process of parturition (138, 142, 143). There are a small number of cases in whom inferior petrosal sinus corticotropin sampling has been performed with CRH stimulation (138, 143). One must balance the risk of possible thrombotic events and radiation when considering such sampling.

 

Radiological imaging may be required following initial investigations. When an adrenal cause is suspected, ultrasound may be sufficient, particularly in cases of adrenal carcinoma (144, 145). Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) appear to be equally sensitive. However, MRI is the investigation of choice due to its superior safety profile (144). Expert bodies do not advocate the use of contrast MRI as the effects on the fetus are currently unknown.  Investigation of suspected pituitary CS requires an appreciation of the normal anatomical changes of the pituitary in pregnancy (discussed at the beginning of the chapter) in order to avoid false-positive findings. Enlargement of the normal pituitary makes the assessment of microadenomas particularly difficult (145). Similar to adrenal imaging, the modality of choice is non-contrast MRI.

 

COMPLICATIONS

 

Untreated CS (in particular) may pose a risk to both mother and fetus. Maternal complications include hypertension (68%), diabetes mellitus or glucose intolerance (25%), preeclampsia (14%), osteoporosis and fractures (5%), cardiac failure (3%), psychiatric disorders (4%), wound infection (2%), and maternal death (2%) (138). Complications for the newborn include preterm delivery (43% of pregnancies), intrauterine growth restriction (21%), stillbirth (6%), spontaneous abortion or intrauterine death (5%), and hypoadrenalism (2%) (138).

 

TREATMENT

 

A review of 136 cases of CD in pregnancy demonstrated a 13% increase in the frequency of live birth rate in patients who underwent active treatment instituted prior to 20 weeks’ gestation and thus treatment is advocated during pregnancy (138). In another review the live birth rate was 15% higher and global fetal morbidity was reduced by 28% (146).

 

Surgical intervention (pituitary or adrenal) may be performed during pregnancy, ideally during the second trimester due to lower rates of maternal and fetal complications (138, 143, 147-149). Medical therapy is also an option. Metyrapone, a steroidogenesis inhibitor, is the most frequently reported option (136). It has been demonstrated to provide good control of the hypercortisolism, although there are reports of adrenal insufficiency, worsened systemic hypertension, and risk of preeclampsia due to deoxycorticosterone accumulation (138, 150-153).  Ketoconazole, another steroidogenesis also inhibitor, while effective in controlling hypercortisolism, is avoided because of its potential teratogenicity and increased abortion rate in animal studies (152, 154-157). Cyproheptadine, a nonselective 5-hydroxytryptamine, is not recommended due to lack of efficacy (138, 158). In a review of four children exposed to mitotane, an adrenal steroidogenesis blocker, during fetal life no clear teratogenic effects were observed; however, the concentrations measured were sub-therapeutic and long-term data do not exist (159). Furthermore, the number of cases was very small. In a previous report it was demonstrated to be teratogenic and thus remains contraindicated (160). Aminoglutethimide, another adrenal steroidogenesis blocker previously in use is also contraindicated as it may cause fetal masculinization (161). There are no reported cases using Pasireotide, a somatostatin analogue, during pregnancy. There are only two reported cases of cabergoline being used for the treatment of CD in pregnancy. The first in a woman who conceived one year after the therapy was initiated and was maintained on high-dose cabergoline throughout gestation undergoing an uneventful pregnancy and delivering a heathy baby (162). The other, being treated for the management of recurrent CD during pregnancy and again a healthy infant was born, on this occasion by caesarean section at term (163).

 

Adrenal Insufficiency

 

Adrenal insufficiency (AI) is relatively rare in pregnancy with an estimated incidence of 1:3000 (164). In cases of primary AI, direct destruction of the adrenal gland occurs and both deficiency in glucocorticoids and mineralocorticoids is observed. In developed countries autoimmune adrenalitis is the most common cause of primary AI which may occur in isolation or as part of an autoimmune polyglandular syndrome (APS). APS type 2 constitutes the most common form and presents as a combination of Addison’s disease, thyroid autoimmune disease, type 1 diabetes, and/or premature ovarian failure (165). This association between primary AI and the other autoimmune disease goes some way towards explaining the reduced fertility rates observed in these women (166). Fertility rates are also reduced in women with AI and no coexisting autoimmune disorders. This has been attributed to lack of libido and the reduced sense of well-being associated with androgen depletion (165). In low income countries, tuberculosis is the most common cause of primary AI (167).

 

Secondary AI occurs is most commonly caused by prolonged exogenous suppression of the hypothalamo-pituitary-adrenal axis as a result of administration of glucocorticoids to treat pre-existing conditions such as asthma. It can also be caused by decreased stimulation of ACTH as a consequence of pituitary or hypothalamic tumors and their associated treatments, resulting in atrophy of the adrenal cortex. Pregnancy-related causes of secondary AI include Sheehan’s syndrome and lymphocytic hypophysitis.  In such women decline in other pituitary hormones is frequently observed (168, 169).

 

Regardless of the cause of the AI, with appropriate hormonal substitution and, if necessary, artificial ovulation techniques, affected women are able to undergo pregnancy. Although considered high risk these women can still achieve favorable outcomes (164, 170).

 

Only a few cases of AI detected during pregnancy have been reported in the literature [40–43], and in this relatively small proportion of women with AI the diagnosis may be challenging as many of the symptoms mimic those of normal pregnancy (165). Common symptoms include weakness, light-headedness, syncope, fatigue, nausea and vomiting, hyponatremia, and increased pigmentation. One must therefore be vigilant for the possibility of a diagnosis of AI, particularly in those with excessive fatigue, weight loss, hypoglycemia, and vomiting, or in those craving salt in order to establish a timely diagnosis. Similarly, one should consider AI in those patients with persistent unexplained orthostasis or hypotension particularly following acute illness or obstetric hemorrhage. Features which may heighten clinical suspicion include hyperpigmentation on the mucous membranes, extensor surfaces, and non-exposed areas, hyponatremia, and a personal or family history of autoimmune diseases.

 

AI may present with an adrenal crisis during pregnancy. Scenarios where this may occur include hyperemesis gravidarum-associated AI in early pregnancy, or presentation secondary to infection at any stage of gestation (164, 171). Some women may present in the postpartum period as transplacental transfer of cortisol from the fetus to the mother can conceal AI until the postpartum period when it is unveiled (172).

 

COMPLICATIONS

 

Pregnancies in which AI is untreated may be complicated both for the mother and neonate. Intrauterine growth restriction, low birth weight, fetal distress, oligohydramnios, and intrauterine death have all been described. Most adverse pregnancy outcomes occur in women in whom the AI was either untreated or undiagnosed (172-176). The fetus receives glucocorticoids from the placenta and thus maternal AI tends not to interfere with fetal development (137). In addition, maternal adrenal antibodies, although capable of crossing the placental barrier, are not thought to be transferred in sufficient quantities to cause fetal/neonatal insufficiency (177).

 

INVESTIGATIONS

 

Interpretation of the investigations to confirm AI present a challenge during pregnancy. Laboratory findings may include hyponatremia, hypoglycemia, eosinophilia, and lymphocytosis. Hyperkalemia may not be present in pregnancy due to gestational effects on the RAAS system (described above).

 

Both serum cortisol and ACTH are increased during pregnancy making them unreliable markers. However, where clinical suspicion is high a low morning cortisol <5 ug/dL (181 nmol/L) in the setting of typical symptoms may be enough to confirm a diagnosis. If this finding is accompanied by a raised ACTH (>2-fold the upper limit of the reference range) a diagnosis of primary AI may be made (178). If both serum cortisol and ACTH are low the patient has secondary AI.

 

The gold standard investigation, and that advocated in the Endocrine Society guidelines, is the cosyntropin stimulation test, also called the short Synacthen test (179). Higher diagnostic cortisol cut-offs are recommended, i.e. 25 ug/dL (700 nmol/L), 29 ug/dL (800 nmol/L) and 32 ug/dL (900 nmol/L) for the first, second and third trimesters respectively (180). Some investigators have proposed the use of the 1 ug cosyntropin test as it is more physiological and may be of value in diagnosing secondary adrenal insufficiency. However, it remains controversial as to whether it improves sensitivity, and is cumbersome to use (181-183).

 

The overnight single-dose metyrapone test may be used to assess adrenal responsiveness but is not recommended in pregnancy as it may precipitate a crisis (167, 184). When used with 750mg every 4 hours for 6 doses in pregnancy 75% of normal pregnant subjects showed a diminished response whereas 25% had a normal response and thus the test did not appear to be valid in pregnancy (185). Cortisol and ACTH responses to CRH are blunted in normal pregnancy (159). As such, the CRH stimulation test is unhelpful in differentiating secondary and tertiary AI in pregnancy. The insulin tolerance test is not used in pregnancy due to the risk of the effect of hypoglycemia on the fetus.

 

Adrenal antibodies remain helpful in pregnancy and, if positive, should prompt consideration of other autoimmune endocrine deficiencies which may co-exist.

 

In cases where radiological imaging is required MRI without gadolinium is the recommended option (145).

 

MANAGEMENT

 

Despite the normal increase in plasma cortisol during pregnancy, an increase in maternal replacement doses of glucocorticoids is not routinely advised. Rather, with the primary aim of avoiding either under or over replacement it is currently recommended that patients are monitored clinically (at least once per trimester) and dose adjustments be made on an individualized basis if required (179). Glucocorticoid preparations that may be used in pregnancy include hydrocortisone, cortisone acetate, prednisolone, or prednisone (179). These glucocorticoids are safe and suppression of neonatal adrenal function is not reported when used for replacement (186). Hydrocortisone is recommended as the glucocorticoid substitution of choice due to its safety profile (167). Dexamethasone is not recommended because it is not inactivated by placental 11β-hydroxysteroid dehydrogenase type 2 and therefore may cross the placenta, this is of concern as this may result in neurocognitive and neurosensory disorders in childhood(98). Higher doses are required during periods of stress such as hyperemesis gravidarum or infection. Additional supplementation is also recommended during the active stage of labor and delivery. Current Endocrine Society guidelines suggest 50mg intravenous hydrocortisone during the second stage of labor and 100mg every 6-8 hours during caesarean section (93). The NICE guideline on management of medical disorders in labor recommends slightly less glucocorticoid replacement in those planning a vaginal birth (50mg 6-hourly). For those having a caesarean section, this guideline suggests individualized glucocorticoid replacement strategies that will depend upon whether the woman has received hydrocortisone in labor, i.e., 50mg intravenous hydrocortisone when starting anesthesia if she has and 100mg hydrocortisone if she has not. Hydrocortisone replacement (50mg 6-hourly) is then recommended until 6 hours after birth (94). The hydrocortisone dose can then be rapidly tapered down following delivery. Physiological doses are safe while breastfeeding as only very minimal quantities are transferred into the milk (187).

 

Progesterone exerts an anti-mineralocorticoid effect, and as a result mineralocorticoid requirement may increase in the third trimester. Serum sodium and potassium and evidence of orthostatic hypotension may be used to for monitoring, but plasma renin is not useful as it increases in pregnancy. If the patient develops hypertension or hypokalemia the dose of the mineralocorticoid should be reduced and if pre-eclampsia occurs it must be stopped (188).

 

Adrenal crisis may occur secondary to infection, hyperemesis gravidarum, or during the stress of labor. In such cases the priorities of treatment include fluid replacement and parenteral administration of glucocorticoid replacement (165).  Women should be advised to wear a medic-alert bracelet, or equivalent, and to carry a “steroid card”.

 

Patient and partner/family education plays a key role in the management of adrenal insufficiency both in and outside of pregnancy. Education should include the basic principles of glucocorticoid replacements, sick day rules, vigilance for symptoms suggestive of glucocorticoid deficiency, and how to self-administer intra-muscular preparations when required. If diagnosed prior to pregnancy this should be included as part of the pre-conception counselling.

 

Congenital Adrenal Hyperplasia

 

Congenital adrenal hyperplasia (CAH) encompasses a group of inherited autosomal recessive (AR) disorders that arise from defective steroidogenesis and result from a deficiency of one or more of the enzymes required for cortisol biosynthesis (189). The most common form of CAH is 21-hydroxylase (CYP21 gene) deficiency, which accounts for 90% of cases (190). CAH due to 21-hydroxylase deficiency can be classified as either classical or non-classical CAH. In classic, severe 21-hydroxylase deficiency females are exposed to excess androgens prenatally and are born with virialized external genitalia (190). Other associations include inadequate vaginal introitus, premature adrenarche, advanced somatic development, central precocious puberty, menstrual irregularity, and possibly salt wasting. Women with the non-classical form present with pubertal and post pubertal hirsutism and menstrual irregularity.

 

Fertility is reduced in these women (191). In women with 21-hydroxylase deficiency a number of rationales for this exist, including the role of progesterone in regulating GnRH, the impact of elevated androgen concentrations on the GnRH/LH pulse generator and on the ovaries (suppressing later stages of follicular development and compromising ovulation) (192). Additionally, an unfavorable anatomical structure may play a role, sometimes complicated by reconstruction surgery. Psychosexual factors may also play a role with reduced sexual desire or orientation reported in some women (192, 193). However, pregnancy is possible in women with CAH (194), and may occur spontaneously once good hormonal control has been achieved with optimized glucocorticoid and mineralocorticoid regimens (189).

 

Fetal risk depends on the carrier status of the father and thus CYP21 genotyping should be performed (195). If the father is a carrier the female fetus is at risk of virilization in cases of classic 21-hyrdoxylase deficiency in the absence of adequate adrenal androgen suppression. In this context, some advocate the use of prenatal dexamethasone which, if used to suppress ACTH and reduce androgen excess, may block the virilization of external genitalia in female fetuses. However, this is an area of ongoing debate as in order to be effective the treatment must be implemented in early pregnancy (prior to the stage of pregnancy when clinicians were previously able to establish fetal sex) thus risking unnecessary exposure in some pregnancies (196). However, the use of newer tests to establish early prenatal diagnosis is an area which will be of great value. Current options include establishing the sex of the fetus using sex determining region Y (SRY) gene testing as early as four weeks of gestation (197). Trophoblast retrieval and isolation from the cervix has also been recently proven to non-invasively and correctly identify male fetal DNA in fetuses at risk of CAH as early as five weeks gestation (189, 198). Alternative options include measurement of cell-free fetal DNA in maternal plasma as early as six weeks, or alternatively chorionic villi sampling at 14 weeks gestation (but this would be associated with a more prolonged period of exposure to glucocorticoids) (199, 200).

 

In pregnancies in which the fetus is thought to be at risk of having CAH and prenatal treatment is desired to reduce androgen excess, dexamethasone is the treatment of choice. Dexamethasone, unlike hydrocortisone, is not inactivated by the placental 11-hydroxysteroid dehydrogenase type 2 and thus may cross the placenta. The aim of this treatment is to reduce virilization, the need for reconstructive surgery, and the emotional distress associated with ambiguous genitalia. However, it does not negate the need for lifelong hormonal replacement (201). The current recommended dose of dexamethasone is 20ug/kg maternal body weight (preconception weight) per day, divided into 2-3 daily doses without exceeding 1.5mg/day (199, 202). This is ideally initiated before seven weeks’ gestation and should be maintained for the entire pregnancy if there is a female fetus (189). Lower doses may, however, be used when poorly tolerated by the mother (197, 203).  Maternal plasma or urinary estriol reflect adrenal synthesis and are monitored to assess efficiency. Therapy must be discontinued as soon as possible if a male or unaffected fetus is identified. Glucocorticoids will need to be increased to cover the stress of labor and delivery.

 

In cases where the fetus is not expected to be affected, hydrocortisone is the maternal glucocorticoid replacement of choice to avoid unnecessary transplacental passage to the fetus.

 

Primary Hyperaldosteronism During Pregnancy

 

The physiological changes observed in the renin angiotensin aldosterone system (RAAS) make the diagnosis of primary hyperaldosteronism (PA) in pregnancy difficult. Thus, despite hypertensive disorders affecting 6-8% of all pregnant women and PA being assumed to account for 10% of all hypertensive disorders, the number of cases reported in the literature is small (204). In a review of 32 pregnancies in women diagnosed with PA during pregnancy, 81% were diagnosed with hypertension during the pregnancy and 19% had been previously diagnosed with hypertension but not screened for PA (205). Of these cases, hypertension was controlled in 19% (two on diuretics), Hypertension was uncontrolled despite medical treatment in 32% of cases and 16% of cases required adrenalectomy during the pregnancy: 23% developed preeclampsia, 61% had induced labor, and the prevalence of caesarean section was 44% (205).

 

The physiological elevation of plasma aldosterone observed in healthy pregnant women is similar to the elevation observed in those with PA, and thus plasma aldosterone levels are not useful for diagnosis in pregnancy. Plasma renin, however, is usually elevated in normal pregnancy, whereas in women with PA it is often found to be suppressed during pregnancy this making an aldosterone-to-renin ratio a more useful test (204, 206, 207). Confirmatory tests such as the saline infusion test or captopril tests are not recommended during pregnancy due to the risk of volume expansion or teratogenicity respectively (204, 208).  MRI is the first line imaging modality during pregnancy. Further subtyping including adrenal vein sampling is not recommended until after the pregnancy (137, 207).

 

The optimal management for PA in pregnancy is unclear due to the condition being rare. Medical management has historically been advocated provided that hypertension and hypokalemia can be adequately managed. However, laparoscopic adrenalectomy during the second trimester has been suggested where an adrenal adenoma can be demonstrated (204, 207, 209).

 

Spironolactone, a mineralocorticoid receptor antagonist (MRA), crosses the placenta, and may have anti-androgenic effects on the male fetus, particularly in the first trimester (the most sensitive period for sex differentiation) (210). Eplerenone, a selective MRA, to date, has not been demonstrated to cause teratogenic effects and is therefore favored in pregnancy (211, 212). Amiloride has also been demonstrated to be effective and safe in a small number of cases (206, 213, 214). Women with primary aldosteronism in pregnancy have higher rates of stillbirth and preterm labor than controls with uncomplicated pregnancy (214).

 

Glucocorticosteroid-remediable aldosteronism (GRA), is a rare hereditary form of primary hyperaldosteronism that is characterized by severe hypertension, hypokalemia, volume expansion, and suppressed plasma renin activity (215). It is a monogenic form of inherited hypertension caused by a chimeric gene originating from an unequal cross-over between the 11β-hydroxylase (CYP11B1) and aldosterone synthase (CYP11 B2) gene (216). In a review of 35 pregnancies in 16 women with GRA there was no increased risk of preeclampsia observed: however, in women with chronic hypertension 39% experienced pregnancy-induced hypertension. In such women with pregnancy-induced hypertension the birth weight of the infants was lower than in those women without. The cesarean section rate was approximately double that seen in the general population (215).

 

Pheochromocytoma and Paraganglioma in Pregnancy


Pheochromocytomas and paraganglioma, collectively known as PPGL, are catecholamine-secreting neuroendocrine tumors which are rare in pregnancy (217). Early recognition of these tumors is important as they are associated with high rates of maternal and fetal complications if undiagnosed. Untreated, the estimated maternal and fetal mortality is estimated at approximately 40-50% (218) whereas when treated maternal and fetal mortality can be reduced to less than 5% and 15%, respectively (219). In a multi-center retrospective study Bancos et al. identified 232 patients with a total of 249 pregnancies in women with PPGL; 78% had a single PPGL (pheochromocytoma in 142 and paraganglioma in 41); 13% had multiple primary PPGL and 9% had metastatic PPGL(220).

 

Symptoms related to episodic catecholamine excess reflect those outside of pregnancy and include paroxysmal headaches, sweating, palpitations, dyspnea, dizziness, and most commonly paroxysmal or sustained hypertension (221). In patients presenting with hypertension, a rare diagnosis such as a PPGL may be missed, as other causes of hypertension (pregnancy-induced hypertension or preeclampsia) are much more common. It is therefore important to be able to distinguish between the different causes. Pregnancy-induced hypertension characteristically presents after 20 weeks’ gestation and pre-eclampsia typically in the last trimester (218, 222). In addition, hypertension associated with pre-eclampsia tends to be consistent throughout whereas when associated with pheochromocytoma there is a tendency towards both paroxysmal and postural changes. The proteinuria and edema associated with pre-eclampsia is not in keeping with a diagnosis of PPGL (223).

 

In some patients, symptoms may be vague and extremely periodic, in which case initial suspicion may only arise when an enlarging uterus causes compression of the neoplasm or during the stress of labor, anesthesia, or surgery. Where a diagnosis is made during such periods of stress and where the diagnosis remains unrecognized, maternal complications such as severe hypertension, hemorrhage into the neoplasm, hemodynamic collapse, myocardial infarction, cardiac arrhythmia, heart failure, or cerebral hemorrhage may occur contributing to the high mortality rate (218, 224).

 

Paraganglioma are often located at the organ of Zuckerkandl, at the bifurcation of the aorta, a region where compression may occur in the context of an enlarging/contracting uterus or during fetal movement (225). In a review of the literature maternal and fetal mortality was lower in women with paragangliomas, 3.6% and 12% respectively compared to 9.8% and 16% in women with pheochromocytomas, but rates were considerably higher than the general obstetric population (226). In the study by Bancos et al. unrecognized PPGL (OR 26.0; 95% CI 3.5-3128.0), abdominal/pelvic locations (OR 11.3; 95% CI 1.5-1441.0), and catecholamine levels of ≥10 times the upper limit of normal (OR 4.7; 95% CI 1.8-13.8) were associated with adverse outcomes. For patients in which a diagnosis was made antepartum alpha-adrenergic blockage was protective in terms of adverse outcomes (OR 3.6; 95% CI 1.1-13.2).

 

First-line investigations include measurement of plasma or 24-hour urinary fractionated metanephrines. Levels in pregnancy are comparable to outside pregnancy and have a sensitivity of 98-99% (221, 227). False positive results may occur in the context of medications such as methyldopa, labetalol, tricyclic antidepressants, ethanol, clonidine, acetaminophen (paracetamol), and phenoxybenzamines, and in other situations that may increase adrenergic activity such as surgery, myocardial infarction, ketoacidosis, obstructive sleep apnea, stroke, and severe heart disease (228). As such, it is recommended that patients stop taking any medication that might interfere with the measurements at least two weeks prior to testing (228). Catecholamine production is not observed to rise in patients with preeclampsia; however, it may rise two-four-fold in the 24-hours following a seizure in eclamptic patients (229, 230).

 

Where the clinical picture and biochemical findings are suggestive of a pheochromocytoma, it is important to establish the location of the tumor. In pregnancy, ultrasound and MRI are the preferred imaging modalities, and if not definitive a multi-detector CT or nuclear scanning may be required (145).

 

After diagnosis, genetic counselling should be considered in the follow-up period as approximately 30% of cases are found to be related to a hereditary syndrome (231). These include multiple endocrine neoplasia type 2 (MEN2), von Hippel-Lindau syndrome, neurofibromatosis, or succinate dehydrogenase subunit gene mutations (218, 219, 232-236). Malignant pheochromocytoma has only been very rarely reported in the literature during pregnancy (237-239).

 

Fetal complications can occur as a consequence of the vasoconstrictive effect of the maternal catecholamines on the uteroplacental circulation which may lead to spontaneous abortion, fetal growth restriction, preterm delivery, fetal distress, and stillbirth (214, 240). Minimal placental transfer of the catecholamines is observed, and this is likely due to high placental concentrations of catechol-O-methyltransferase and monoamine oxidase (240-242).

 

The management of patients with pheochromocytoma and paraganglioma relies on α-adrenergic receptor blockade prior to surgical removal of the tumor. In pregnancy it is important to maintain adequate uteroplacental circulation which is entirely under the influence of maternal blood pressure. Therefore, a balance must be achieved between reducing excess catecholamines and avoiding severe hypotension (231). The most commonly used α-adrenergic receptor blockers include phenoxybenzamine, doxazosin, and prazosin. Phenoxybenzamine is favored due to its long acting, stable, non-competitive blockade and has been used in a number of pregnant women with pheochromocytoma with good outcomes (243). It does, however, cross the placental barrier and neonatal hypotension and respiratory distress have been reported in babies whose mothers were treated with phenoxybenzamine; thus, careful monitoring is required with the involvement of neonatologists at the time of delivery (244). Maternal tachycardia may occur with phenoxybenzamine, and in such cases prazosin or doxazosin may have a role as they produce less tachycardia (218). Prazosin has been used in the management of hypertension in pregnancy (245) but should be used with caution as, when compared to nifedipine for control of severe hypertension in pregnancy, a greater number of intrauterine deaths occurred in the prazosin group (246). Doxazosin has also been used with good outcomes (247).

 

administered following α-blockade. Beta-blockers have been associated with fetal bradycardia and intrauterine growth restriction when used in high doses, but clearly the advantages in women with pheochromocytoma should be weighed against the relatively uncommon fetal risks (248, 249). Labetalol is a combined alpha and beta blocker but is not recommended as the α-blockade is relatively weak, and thus paroxysmal hypertension may occur. Methyldopa is also not recommended as it has been suggested that it might worsen hypertension (231). In case of a hypertensive emergency, phentolamine is advised due to its prompt onset of action. Beta-adrenoceptor blockers may be added if tachyarrhythmia occurs but must only be

 

Drugs that should be avoided in women with pheochromocytoma include corticosteroids, opioids, pethidine, metoclopramide and certain anesthetic drugs such as thiopental, ketamine, ephedrine and mivacurium, as they may induce crisis by promoting the release of catecholamines (231). 

 

Adequate α-adrenergic blockade +/- the addition of beta-blockade is the first priority in managing women with pheochromocytoma, but surgery is the definitive treatment. The optimal timing of surgery remains a topic of debate and depends on gestational age, the success of medical management, and the location of the tumor. In patients in whom the diagnosis is made within the first 24 weeks and adequate α-adrenergic blockade has been achieved, the current recommendation is that the tumor be removed in the second trimester. In patients with pheochromocytoma identified in the third trimester and/or in whom medical management is adequate it is often advised to delay surgery until following delivery (218, 250). Where possible, a laparoscopic approach in now preferred (231, 235).

 

Historically, a vaginal delivery was avoided in women with pheochromocytoma due to concerns about a catecholamine surge during labor and delivery.  However, this theoretical risk should be mitigated with adequate α-adrenergic blockade. Several vaginal deliveries with good outcomes have now been reported in the literature, with careful consideration of medical management during the labor and good analgesia with avoidance of medications which may trigger a crisis (218, 251-253). Ideally an individualized, patient-centered, and multidisciplinary approach is required to decide the best mode of delivery for the individual patient.

 

SUMMARY

 

In summary, the management of both pituitary and adrenal diseases in pregnancy relies on a good understanding of the physiological changes during gestation. Increasing evidence is becoming available regarding the drugs that are available for management of these conditions, giving more confidence to those managing affected pregnant women and providing additional information to share with patients. As with other medical problems encountered in pregnancy, it is important to provide women with evidence-based pre-conception counselling to enable informed shared decision making. Multidisciplinary team and individualized care are essential to ensure prompt diagnosis and effective management of potentially high-risk pituitary and adrenal disease in pregnancy to ensure the best outcomes for both mother and child.

 

ACKNOWLEDGEMENT

 

The authors would like to thank Professor Mark E Molitch who wrote the original version of this chapter and designed Table 1 in the current draft.

 

REFERENCES

                                                                                                                                 

  1. Gonzalez JG, Elizondo G, Saldivar D, Nanez H, Todd LE, Villarreal JZ. Pituitary gland growth during normal pregnancy: an in vivo study using magnetic resonance imaging. Am J Med. 1988;85(2):217-20.
  2. Dinç H, Esen F, Demirci A, Sari A, Resit Gümele H. Pituitary dimensions and volume measurements in pregnancy and post partum. MR assessment. Acta Radiol. 1998;39(1):64-9.
  3. Abucham J, Bronstein MD, Dias ML. MANAGEMENT OF ENDOCRINE DISEASE: Acromegaly and pregnancy: a contemporary review. Eur J Endocrinol. 2017;177(1):R1-R12.
  4. Goluboff LG, Ezrin C. Effect of pregnancy on the somatotroph and the prolactin cell of the human adenohypophysis. J Clin Endocrinol Metab. 1969;29(12):1533-8.
  5. Scheithauer BW, Sano T, Kovacs KT, Young WF, Ryan N, Randall RV. The pituitary gland in pregnancy: a clinicopathologic and immunohistochemical study of 69 cases. Mayo Clin Proc. 1990;65(4):461-74.
  6. Laway BA. Pregnancy in acromegaly. Ther Adv Endocrinol Metab. 2015;6(6):267-72.
  7. Bethesda. National Council on Radiation Protection and Measurements: Risk estimates for radiation protection. Report Number 115 ed2007.
  8. Ray JG, Vermeulen MJ, Bharatha A, Montanera WJ, Park AL. Association Between MRI Exposure During Pregnancy and Fetal and Childhood Outcomes. JAMA. 2016;316(9):952-61.
  9. Webb JA, Thomsen HS, Morcos SK, (ESUR) MoCMSCoESoUR. The use of iodinated and gadolinium contrast media during pregnancy and lactation. Eur Radiol. 2005;15(6):1234-40.
  10. Kanal E, Barkovich AJ, Bell C, Borgstede JP, Bradley WG, Froelich JW, et al. ACR guidance document for safe MR practices: 2007. AJR Am J Roentgenol. 2007;188(6):1447-74.
  11. Ciet P, Litmanovich DE. MR safety issues particular to women. Magn Reson Imaging Clin N Am. 2015;23(1):59-67.
  12. Freeman ME, Kanyicska B, Lerant A, Nagy G. Prolactin: structure, function, and regulation of secretion. Physiol Rev. 2000;80(4):1523-631.
  13. Marano RJ, Ben-Jonathan N. Minireview: Extrapituitary prolactin: an update on the distribution, regulation, and functions. Mol Endocrinol. 2014;28(5):622-33.
  14. Ben-Jonathan N, Mershon JL, Allen DL, Steinmetz RW. Extrapituitary prolactin: distribution, regulation, functions, and clinical aspects. Endocr Rev. 1996;17(6):639-69.
  15. Vician L, Shupnik MA, Gorski J. Effects of estrogen on primary ovine pituitary cell cultures: stimulation of prolactin secretion, synthesis, and preprolactin messenger ribonucleic acid activity. Endocrinology. 1979;104(3):736-43.
  16. Maurer RA. Relationship between estradiol, ergocryptine, and thyroid hormone: effects on prolactin synthesis and prolactin messenger ribonucleic acid levels. Endocrinology. 1982;110(5):1515-20.
  17. Rakoff JS, Yen SS. Progesterone induced acute release of prolactin in estrogen primed ovariectomized women. J Clin Endocrinol Metab. 1978;47(4):918-21.
  18. Maslar IA, Ansbacher R. Effects of progesterone on decidual prolactin production by organ cultures of human endometrium. Endocrinology. 1986;118(5):2102-8.
  19. Rigg LA, Lein A, Yen SS. Pattern of increase in circulating prolactin levels during human gestation. Am J Obstet Gynecol. 1977;129(4):454-6.
  20. Cocks Eschler D, Javanmard P, Cox K, Geer EB. Prolactinoma through the female life cycle. Endocrine. 2018;59(1):16-29.
  21. Molitch ME. Endocrinology in pregnancy: management of the pregnant patient with a prolactinoma. Eur J Endocrinol. 2015;172(5):R205-13.
  22. Glass MR, Shaw RW, Butt WR, Edwards RL, London DR. An abnormality of oestrogen feedback in amenorrhoea-galactorrhoea. Br Med J. 1975;3(5978):274-5.
  23. Demura R, Ono M, Demura H, Shizume K, Oouchi H. Prolactin directly inhibits basal as well as gonadotropin-stimulated secretion of progesterone and 17 beta-estradiol in the human ovary. J Clin Endocrinol Metab. 1982;54(6):1246-50.
  24. Dorrington JH, Gore-Langton RE. Antigonadal action of prolactin: further studies on the mechanism of inhibition of follicle-stimulating hormone-induced aromatase activity in rat granulosa cell cultures. Endocrinology. 1982;110(5):1701-7.
  25. Krasnow JS, Hickey GJ, Richards JS. Regulation of aromatase mRNA and estradiol biosynthesis in rat ovarian granulosa and luteal cells by prolactin. Mol Endocrinol. 1990;4(1):13-2.
  26. McNatty KP. Relationship between plasma prolactin and the endocrine microenvironment of the developing human antral follicle. Fertil Steril. 1979;32(4):433-8.
  27. Seppälä M, Ranta T, Hirvonen E. Hyperprolactinaemia and luteal insufficiency. Lancet. 1976;1(7953):229-30.
  28. Gillam MP, Molitch ME, Lombardi G, Colao A. Advances in the treatment of prolactinomas. Endocr Rev. 2006;27(5):485-534.
  29. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(2):273-88.
  30. Klibanski A. Clinical practice. Prolactinomas. N Engl J Med. 2010;362(13):1219-26.
  31. Husk, Katherine. Prolactinoma. In: Berghella V, editor. Maternal-Fetal Evidence Based Guidelines: CRC Press; 2017.
  32. Chrisoulidou A, Boudina M, Karavitaki N, Bili E, Wass J. Pituitary disorders in pregnancy. Hormones (Athens). 2015;14(1):70-80.
  33. Glezer A, Jallad RS, Machado MC, Fragoso MC, Bronstein MD. Pregnancy and pituitary adenomas. Minerva Endocrinol. 2016;41(3):341-50.
  34. Luger A, Broersen LHA, Biermasz NR, Biller BMK, Buchfelder M, Chanson P, et al. ESE Clinical Practice Guideline on functioning and nonfunctioning pituitary adenomas in pregnancy. Eur J Endocrinol. 2021;185(3):G1-G33.
  35. Bigazzi M, Ronga R, Lancranjan I, Ferraro S, Branconi F, Buzzoni P, et al. A pregnancy in an acromegalic woman during bromocriptine treatment: effects on growth hormone and prolactin in the maternal, fetal, and amniotic compartments. J Clin Endocrinol Metab. 1979;48(1):9-12.
  36. Raymond JP, Goldstein E, Konopka P, Leleu MF, Merceron RE, Loria Y. Follow-up of children born of bromocriptine-treated mothers. Horm Res. 1985;22(3):239-46.
  37. Bronstein MD. Prolactinomas and pregnancy. Pituitary. 2005;8(1):31-8.
  38. Krupp P, Monka C. Bromocriptine in pregnancy: safety aspects. Klin Wochenschr. 1987;65(17):823-7.
  39. Ono M, Miki N, Amano K, Kawamata T, Seki T, Makino R, et al. Individualized high-dose cabergoline therapy for hyperprolactinemic infertility in women with micro- and macroprolactinomas. J Clin Endocrinol Metab. 2010;95(6):2672-9.
  40. Lebbe M, Hubinont C, Bernard P, Maiter D. Outcome of 100 pregnancies initiated under treatment with cabergoline in hyperprolactinaemic women. Clin Endocrinol (Oxf). 2010;73(2):236-42.
  41. Stalldecker G, Mallea-Gil MS, Guitelman M, Alfieri A, Ballarino MC, Boero L, et al. Effects of cabergoline on pregnancy and embryo-fetal development: retrospective study on 103 pregnancies and a review of the literature. Pituitary. 2010;13(4):345-50.
  42. Canales ES, García IC, Ruíz JE, Zárate A. Bromocriptine as prophylactic therapy in prolactinoma during pregnancy. Fertil Steril. 1981;36(4):524-6.
  43. Glezer A, Bronstein MD. Prolactinomas, cabergoline, and pregnancy. Endocrine. 2014;47(1):64-9.
  44. Rastogi A, Bhadada SK, Bhansali A. Pregnancy and tumor outcomes in infertile women with macroprolactinoma on cabergoline therapy. Gynecol Endocrinol. 2017;33(4):270-3.
  45. Lambert K, Rees K, Seed PT, Dhanjal MK, Knight M, McCance DR, et al. Macroprolactinomas and Nonfunctioning Pituitary Adenomas and Pregnancy Outcomes. Obstet Gynecol. 2017;129(1):185-94.
  46. Vilar L, Abucham J, Albuquerque JL, Araujo LA, Azevedo MF, Boguszewski CL, et al. Controversial issues in the management of hyperprolactinemia and prolactinomas - An overview by the Neuroendocrinology Department of the Brazilian Society of Endocrinology and Metabolism. Arch Endocrinol Metab. 2018;62(2):236-63.
  47. Narita O, Kimura T, Suganuma N, Osawa M, Mizutani S, Masahashi T, et al. Relationship between maternal prolactin levels during pregnancy and lactation in women with pituitary adenoma. Nihon Sanka Fujinka Gakkai Zasshi. 1985;37(5):758-62.
  48. Frankenne F, Closset J, Gomez F, Scippo ML, Smal J, Hennen G. The physiology of growth hormones (GHs) in pregnant women and partial characterization of the placental GH variant. J Clin Endocrinol Metab. 1988;66(6):1171-80.
  49. Fuglsang J, Skjaerbaek C, Espelund U, Frystyk J, Fisker S, Flyvbjerg A, et al. Ghrelin and its relationship to growth hormones during normal pregnancy. Clin Endocrinol (Oxf). 2005;62(5):554-9.
  50. McIlhargey, Trina. Acromegaly in Pregnancy—An Overview of the Key Issues. In: Corenblum, Bernard, editors. US Endocrinology2012. p. 53-6.
  51. Kaltsas GA, Mukherjee JJ, Jenkins PJ, Satta MA, Islam N, Monson JP, et al. Menstrual irregularity in women with acromegaly. J Clin Endocrinol Metab. 1999;84(8):2731-5.
  52. Grynberg M, Salenave S, Young J, Chanson P. Female gonadal function before and after treatment of acromegaly. J Clin Endocrinol Metab. 2010;95(10):4518-25.
  53. Katznelson L, Kleinberg D, Vance ML, Stavrou S, Pulaski KJ, Schoenfeld DA, et al. Hypogonadism in patients with acromegaly: data from the multi-centre acromegaly registry pilot study. Clin Endocrinol (Oxf). 2001;54(2):183-8.
  54. Dogansen SC, Tanrikulu S, Yalin GY, Yarman S. Female gonadal functions and ovarian reserve in patients with acromegaly: experience from a single tertiary center. Endocrine. 2018;60(1):167-74.
  55. Assal A, Malcolm J, Lochnan H, Keely E. Preconception counselling for women with acromegaly: More questions than answers. Obstet Med. 2016;9(1):9-14.
  56. Caron P, Broussaud S, Bertherat J, Borson-Chazot F, Brue T, Cortet-Rudelli C, et al. Acromegaly and pregnancy: a retrospective multicenter study of 59 pregnancies in 46 women. J Clin Endocrinol Metab. 2010;95(10):4680-7.
  57. Cozzi R, Attanasio R, Barausse M. Pregnancy in acromegaly: a one-center experience. Eur J Endocrinol. 2006;155(2):279-84.
  58. Cheng S, Grasso L, Martinez-Orozco JA, Al-Agha R, Pivonello R, Colao A, et al. Pregnancy in acromegaly: experience from two referral centers and systematic review of the literature. Clin Endocrinol (Oxf). 2012;76(2):264-71.
  59. Dias M, Boguszewski C, Gadelha M, Kasuki L, Musolino N, Vieira JG, et al. Acromegaly and pregnancy: a prospective study. Eur J Endocrinol. 2014;170(2):301-10.
  60. Hannon AM, O'Shea T, Thompson CA, Hannon MJ, Dineen R, Khattak A, et al. Pregnancy in acromegaly is safe and is associated with improvements in IGF-1 concentrations. Eur J Endocrinol. 2019;180(4):K21-K9.
  61. Jallad RS, Shimon I, Fraenkel M, Medvedovsky V, Akirov A, Duarte FH, et al. Outcome of pregnancies in a large cohort of women with acromegaly. Clin Endocrinol (Oxf). 2018;88(6):896-907.
  62. Persechini ML, Gennero I, Grunenwald S, Vezzosi D, Bennet A, Caron P. Decreased IGF-1 concentration during the first trimester of pregnancy in women with normal somatotroph function. Pituitary. 2015;18(4):461-4.
  63. Lau SL, McGrath S, Evain-Brion D, Smith R. Clinical and biochemical improvement in acromegaly during pregnancy. J Endocrinol Invest. 2008;31(3):255-61.
  64. Dicuonzo F, Purciariello S, De Marco A, Guastamacchia E, Triggiani V. Inoperable Giant Growth Hormone-secreting Pituitary Adenoma: Radiological Aspects, Clinical Management and Pregnancy Outcome. Endocr Metab Immune Disord Drug Targets. 2018.
  65. Bandeira DB, Olivatti TOF, Bolfi F, Boguszewski CL, Dos Santos Nunes-Nogueira V. Acromegaly and pregnancy: a systematic review and meta-analysis. Pituitary. 2022;25(3):352-62.
  66. The global challenge of diabetes. Lancet. 2008;371(9626):1723.
  67. Katznelson L, Laws ER, Melmed S, Molitch ME, Murad MH, Utz A, et al. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99(11):3933-51.
  68. Vialon M, Grunenwald S, Mouly C, Vezzosi D, Bennet A, Caron P. First-generation somatostatin receptor ligands and pregnancy: lesson from women with acromegaly. Endocrine. 2020;70(2):396-403.
  69. Qureshi A, Kalu E, Ramanathan G, Bano G, Croucher C, Panahloo A. IVF/ICSI in a woman with active acromegaly: successful outcome following treatment with pegvisomant. J Assist Reprod Genet. 2006;23(11-12):439-42.
  70. Brian SR, Bidlingmaier M, Wajnrajch MP, Weinzimer SA, Inzucchi SE. Treatment of acromegaly with pegvisomant during pregnancy: maternal and fetal effects. J Clin Endocrinol Metab. 2007;92(9):3374-7.
  71. van der Lely AJ, Gomez R, Heissler JF, Åkerblad AC, Jönsson P, Camacho-Hübner C, et al. Pregnancy in acromegaly patients treated with pegvisomant. Endocrine. 2015;49(3):769-73.
  72. Guarda FJ, Gong W, Ghajar A, Guitelman M, Nachtigall LB. Preconception use of pegvisomant alone or as combination therapy for acromegaly: a case series and review of the literature. Pituitary. 2020;23(5):498-506.
  73. Caron P, Gerbeau C, Pradayrol L, Simonetta C, Bayard F. Successful pregnancy in an infertile woman with a thyrotropin-secreting macroadenoma treated with somatostatin analog (octreotide). J Clin Endocrinol Metab. 1996;81(3):1164-8.
  74. Blackhurst G, Strachan MW, Collie D, Gregor A, Statham PF, Seckl JE. The treatment of a thyrotropin-secreting pituitary macroadenoma with octreotide in twin pregnancy. Clin Endocrinol (Oxf). 2002;57(3):401-4.
  75. Chaiamnuay S, Moster M, Katz MR, Kim YN. Successful management of a pregnant woman with a TSH secreting pituitary adenoma with surgical and medical therapy. Pituitary. 2003;6(2):109-13.
  76. Bolz M, Körber S, Schober HC. [TSH secreting adenoma of pituitary gland (TSHom) -rare cause of hyperthyroidism in pregnancy]. Dtsch Med Wochenschr. 2013;138(8):362-6.
  77. Abuzaid, Hassan. Case Report: A Rare Case of Central

Hyperthyroidism During Pregnancy- Diagnostic

And Therapeutic Challenge. World Journal of Research and Review2016. p. 21-3.

  1. Perdomo CM, Árabe JA, Idoate M, Galofré JC. Management of a pregnant woman with thyrotropinoma: a case report and review of the literature. Gynecol Endocrinol. 2017;33(3):188-92.
  2. Mercado M, Melgar V, Salame L, Cuenca D. Clinically non-functioning pituitary adenomas: Pathogenic, diagnostic and therapeutic aspects. Endocrinol Diabetes Nutr. 2017;64(7):384-95.
  3. Masding MG, Lees PD, Gawne-Cain ML, Sandeman DD. Visual field compression by a non-secreting pituitary tumour during pregnancy. J R Soc Med. 2003;96(1):27-8.
  4. Murata Y, Ando H, Nagasaka T, Takahashi I, Saito K, Fukugaki H, et al. Successful pregnancy after bromocriptine therapy in an anovulatory woman complicated with ovarian hyperstimulation caused by follicle-stimulating hormone-producing plurihormonal pituitary microadenoma. J Clin Endocrinol Metab. 2003;88(5):1988-93.
  5. Sugita T, Seki K, Nagai Y, Saeki N, Yamaura A, Ohigashi S, et al. Successful pregnancy and delivery after removal of gonadotrope adenoma secreting follicle-stimulating hormone in a 29-year-old amenorrheic woman. Gynecol Obstet Invest. 2005;59(3):138-43.
  6. Zhang Y, Chen C, Lin M, Deng K, Zhu H, Ma W, et al. Successful pregnancy after operation in an infertile woman caused by luteinizing hormone-secreting pituitary adenoma: case report and literature review. BMC Endocr Disord. 2021;21(1):15.
  7. Abbara A, Clarke S, Eng PC, Milburn J, Joshi D, Comninos AN, et al. Clinical and biochemical characteristics of patients presenting with pituitary apoplexy. Endocr Connect. 2018.
  8. Grand'Maison S, Weber F, Bédard MJ, Mahone M, Godbout A. Pituitary apoplexy in pregnancy: A case series and literature review. Obstet Med. 2015;8(4):177-83.
  9. Abraham RR, Pollitzer RE, Gokden M, Goulden PA. Spontaneous pituitary apoplexy during the second trimester of pregnancy, with sensory loss. BMJ Case Rep. 2016;2016.
  10. Watson V. An unexpected headache: pituitary apoplexy in a pregnant woman on anticoagulation. BMJ Case Rep. 2015;2015.
  11. Tandon A, Alzate J, LaSala P, Fried MP. Endoscopic Endonasal Transsphenoidal Resection for Pituitary Apoplexy during the Third Trimester of Pregnancy. Surg Res Pract. 2014;2014:397131.
  12. Hayes AR, O'Sullivan AJ, Davies MA. A case of pituitary apoplexy in pregnancy. Endocrinol Diabetes Metab Case Rep. 2014;2014:140043.
  13. Keleştimur F. Sheehan's syndrome. Pituitary. 2003;6(4):181-8.
  14. Shivaprasad C. Sheehan's syndrome: Newer advances. Indian J Endocrinol Metab. 2011;15 Suppl 3:S203-7.
  15. Gei-Guardia O, Soto-Herrera E, Gei-Brealey A, Chen-Ku CH. Sheehan syndrome in Costa Rica: clinical experience with 60 cases. Endocr Pract. 2011;17(3):337-44.
  16. Fleseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, et al. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(11):3888-921.
  17. NICE. Intrapartum care for women with existing medical conditions or obstetric complications and their babies 2019 [Available from: https://www.nice.org.uk/guidance/ng121.
  18. Oliveira D, Lages A, Paiva S, Carrilho F. Treatment of Addison's disease during pregnancy. Endocrinol Diabetes Metab Case Rep. 2018;2018.
  19. van Runnard Heimel PJ, Schobben AF, Huisjes AJ, Franx A, Bruinse HW. The transplacental passage of prednisolone in pregnancies complicated by early-onset HELLP syndrome. Placenta. 2005;26(10):842-5.
  20. Khan AA, Rodriguez A, Kaakinen M, Pouta A, Hartikainen AL, Jarvelin MR. Does in utero exposure to synthetic glucocorticoids influence birthweight, head circumference and birth length? A systematic review of current evidence in humans. Paediatr Perinat Epidemiol. 2011;25(1):20-36.
  21. Magala Ssekandi A, Sserwanja Q, Olal E, Kawuki J, Bashir Adam M. Corticosteroids Use in Pregnant Women with COVID-19: Recommendations from Available Evidence. J Multidiscip Healthc. 2021;14:659-63.
  22. Vila G, Akerblad AC, Mattsson AF, Riedl M, Webb SM, Hána V, et al. Pregnancy outcomes in women with growth hormone deficiency. Fertil Steril. 2015;104(5):1210-7.e1.
  23. Biller BMK, Höybye C, Carroll P, Gordon MB, Birkegård AC, Kelepouris N, et al. Pregnancy outcomes in women receiving growth hormone replacement therapy enrolled in the NordiNet® International Outcome Study (IOS) and the American Norditropin® Studies: Web-Enabled Research (ANSWER) Program. Pituitary. 2021;24(4):611-21.
  24. Caturegli P, Newschaffer C, Olivi A, Pomper MG, Burger PC, Rose NR. Autoimmune hypophysitis. Endocr Rev. 2005;26(5):599-614.
  25. Karaca Z, Kelestimur F. The management of hypophysitis. Minerva Endocrinol. 2016;41(3):390-9.
  26. Takeda R, Demura M, Sugimura Y, Miyamori I, Konoshita T, Yamamoto H. Pregnancy-associated diabetes insipidus in Japan-a review based on quoting from the literatures reported during the period from 1982 to 2019. Endocr J. 2021;68(4):375-85.
  27. Ray JG. DDAVP use during pregnancy: an analysis of its safety for mother and child. Obstet Gynecol Surv. 1998;53(7):450-5.
  28. Lindheimer MD, Davison JM. Osmoregulation, the secretion of arginine vasopressin and its metabolism during pregnancy. Eur J Endocrinol. 1995;132(2):133-43.
  29. Barron WM, Cohen LH, Ulland LA, Lassiter WE, Fulghum EM, Emmanouel D, et al. Transient vasopressin-resistant diabetes insipidus of pregnancy. N Engl J Med. 1984;310(7):442-4.
  30. Schrier RW. Systemic arterial vasodilation, vasopressin, and vasopressinase in pregnancy. J Am Soc Nephrol. 2010;21(4):570-2.
  31. Davison JM, Sheills EA, Philips PR, Barron WM, Lindheimer MD. Metabolic clearance of vasopressin and an analogue resistant to vasopressinase in human pregnancy. Am J Physiol. 1993;264(2 Pt 2):F348-53.
  32. Schrier RW. Body water homeostasis: clinical disorders of urinary dilution and concentration. J Am Soc Nephrol. 2006;17(7):1820-32.
  33. Hague WM. Diabetes insipidus in pregnancy. Obstet Med. 2009;2(4):138-41.
  34. Russell JA, Leng G, Douglas AJ. The magnocellular oxytocin system, the fount of maternity: adaptations in pregnancy. Front Neuroendocrinol. 2003;24(1):27-61.
  35. Gibbens GL, Chard T. Observations on maternal oxytocin release during human labor and the effect of intravenous alcohol administration. Am J Obstet Gynecol. 1976;126(2):243-6.
  36. Dawood MY, Khan-Dawood FS, Wahi RS, Fuchs F. Oxytocin release and plasma anterior pituitary and gonadal hormones in women during lactation. J Clin Endocrinol Metab. 1981;52(4):678-83.
  37. Ananthakrishnan S. Diabetes insipidus in pregnancy: etiology, evaluation, and management. Endocr Pract. 2009;15(4):377-82.
  38. Aleksandrov N, Audibert F, Bedard MJ, Mahone M, Goffinet F, Kadoch IJ. Gestational diabetes insipidus: a review of an underdiagnosed condition. J Obstet Gynaecol Can. 2010;32(3):225-31.
  39. Chanson P, Salenave S. Diabetes insipidus and pregnancy. Ann Endocrinol (Paris). 2016;77(2):135-8.
  40. Källén BA, Carlsson SS, Bengtsson BK. Diabetes insipidus and use of desmopressin (Minirin) during pregnancy. Eur J Endocrinol. 1995;132(2):144-6.
  41. Seckl JR, Dunger DB. Diabetes insipidus. Current treatment recommendations. Drugs. 1992;44(2):216-24.
  42. Sainz Bueno JA, Villarejo Ortíz P, Hidalgo Amat J, Caballero Fernández V, Caballero Manzano M, Garrido Teruel R. Transient diabetes insipidus during pregnancy: a clinical case and a review of the syndrome. Eur J Obstet Gynecol Reprod Biol. 2005;118(2):251-4.
  43. Wallia A, Bizhanova A, Huang W, Goldsmith SL, Gossett DR, Kopp P. Acute diabetes insipidus mediated by vasopressinase after placental abruption. J Clin Endocrinol Metab. 2013;98(3):881-6.
  44. Marques P, Gunawardana K, Grossman A. Transient diabetes insipidus in pregnancy. Endocrinol Diabetes Metab Case Rep. 2015;2015:150078.
  45. Hawker RS, North WG, Colbert IC, Lang LP. Oxytocin blood levels in two cases of diabetes insipidus. J Obstet Gynaecol Br Commonw. 1967;74(3):430-1.
  46. Nawathe A, Govind A. Pregnancy with known syndrome of inappropriate antidiuretic hormone. J Obstet Gynaecol. 2013;33(1):9-13.
  47. Wilson HJ, Shutt LE. Syndrome of inappropriate ADH secretion in a woman with preeclampsia. Int J Obstet Anesth. 2007;16(4):360-2.
  48. Qureshi AC, Bahri A, Breen LA, Barnes SC, Powrie JK, Thomas SM, et al. The influence of the route of oestrogen administration on serum levels of cortisol-binding globulin and total cortisol. Clin Endocrinol (Oxf). 2007;66(5):632-5.
  49. Duthie L, Reynolds RM. Changes in the maternal hypothalamic-pituitary-adrenal axis in pregnancy and postpartum: influences on maternal and fetal outcomes. Neuroendocrinology. 2013;98(2):106-15.
  50. Mastorakos G, Ilias I. Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. Ann N Y Acad Sci. 2003;997:136-49.
  51. Hsueh WA, Luetscher JA, Carlson EJ, Grislis G, Fraze E, McHargue A. Changes in active and inactive renin throughout pregnancy. J Clin Endocrinol Metab. 1982;54(5):1010-6.
  52. Brown MA, Gallery ED, Ross MR, Esber RP. Sodium excretion in normal and hypertensive pregnancy: a prospective study. Am J Obstet Gynecol. 1988;159(2):297-307.
  53. Irani RA, Xia Y. Renin angiotensin signaling in normal pregnancy and preeclampsia. Semin Nephrol. 2011;31(1):47-58.
  54. Escher G. Hyperaldosteronism in pregnancy. Ther Adv Cardiovasc Dis. 2009;3(2):123-32.
  55. ABDUL-KARIM R, ASSALIN S. Pressor response to angiotonin in pregnant and nonpregnant women. Am J Obstet Gynecol. 1961;82:246-51.
  56. Gant NF, Worley RJ, Everett RB, MacDonald PC. Control of vascular responsiveness during human pregnancy. Kidney Int. 1980;18(2):253-8.
  57. AbdAlla S, Lother H, el Massiery A, Quitterer U. Increased AT(1) receptor heterodimers in preeclampsia mediate enhanced angiotensin II responsiveness. Nat Med. 2001;7(9):1003-9.
  58. Brue T, Amodru V, Castinetti F. MANAGEMENT OF ENDOCRINE DISEASE: Management of Cushing's syndrome during pregnancy: solved and unsolved questions. Eur J Endocrinol. 2018;178(6):R259-R66.
  59. Bronstein MD, Machado MC, Fragoso MC. MANAGEMENT OF ENDOCRINE DISEASE: Management of pregnant patients with Cushing's syndrome. Eur J Endocrinol. 2015;173(2):R85-91.
  60. Lindsay JR, Nieman LK. The hypothalamic-pituitary-adrenal axis in pregnancy: challenges in disease detection and treatment. Endocr Rev. 2005;26(6):775-99.
  61. Lindsay JR, Jonklaas J, Oldfield EH, Nieman LK. Cushing's syndrome during pregnancy: personal experience and review of the literature. J Clin Endocrinol Metab. 2005;90(5):3077-83.
  62. Monticone S, Auchus RJ, Rainey WE. Adrenal disorders in pregnancy. Nat Rev Endocrinol. 2012;8(11):668-78.
  63. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, et al. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2008;93(5):1526-40.
  64. Lopes LM, Francisco RP, Galletta MA, Bronstein MD. Determination of nighttime salivary cortisol during pregnancy: comparison with values in non-pregnancy and Cushing's disease. Pituitary. 2016;19(1):30-8.
  65. Quartero HW, Fry CH. Placental corticotrophin releasing factor may modulate human parturition. Placenta. 1989;10(5):439-43.
  66. Pinette MG, Pan YQ, Oppenheim D, Pinette SG, Blackstone J. Bilateral inferior petrosal sinus corticotropin sampling with corticotropin-releasing hormone stimulation in a pregnant patient with Cushing's syndrome. Am J Obstet Gynecol. 1994;171(2):563-4.
  67. Suzuki Y, Sasagawa, Suzuki H, Izumi T, Kaneko H, Nakada T. The role of ultrasonography in the detection of adrenal masses: comparison with computed tomography and magnetic resonance imaging. Int Urol Nephrol. 2001;32(3):303-6.
  68. Lowe, Sandra. Imaging of endocrine disorders in pregnancy.Oxford Handbook of Endocrinology and Diabetes. In press2019.
  69. Caimari F, Valassi E, Garbayo P, Steffensen C, Santos A, Corcoy R, et al. Cushing's syndrome and pregnancy outcomes: a systematic review of published cases. Endocrine. 2017;55(2):555-63.
  70. Abbassy M, Kshettry VR, Hamrahian AH, Johnston PC, Dobri GA, Avitsian R, et al. Surgical management of recurrent Cushing's disease in pregnancy: A case report. Surg Neurol Int. 2015;6(Suppl 25):S640-5.
  71. Casson IF, Davis JC, Jeffreys RV, Silas JH, Williams J, Belchetz PE. Successful management of Cushing's disease during pregnancy by transsphenoidal adenectomy. Clin Endocrinol (Oxf). 1987;27(4):423-8.
  72. Ross RJ, Chew SL, Perry L, Erskine K, Medbak S, Afshar F. Diagnosis and selective cure of Cushing's disease during pregnancy by transsphenoidal surgery. Eur J Endocrinol. 1995;132(6):722-6.
  73. Lim WH, Torpy DJ, Jeffries WS. The medical management of Cushing's syndrome during pregnancy. Eur J Obstet Gynecol Reprod Biol. 2013;168(1):1-6.
  74. Blanco C, Maqueda E, Rubio JA, Rodriguez A. Cushing's syndrome during pregnancy secondary to adrenal adenoma: metyrapone treatment and laparoscopic adrenalectomy. J Endocrinol Invest. 2006;29(2):164-7.
  75. Boronat M, Marrero D, López-Plasencia Y, Barber M, Schamann Y, Nóvoa FJ. Successful outcome of pregnancy in a patient with Cushing's disease under treatment with ketoconazole during the first trimester of gestation. Gynecol Endocrinol. 2011;27(9):675-7.
  76. Close CF, Mann MC, Watts JF, Taylor KG. ACTH-independent Cushing's syndrome in pregnancy with spontaneous resolution after delivery: control of the hypercortisolism with metyrapone. Clin Endocrinol (Oxf). 1993;39(3):375-9.
  77. Zieleniewski W, Michalak R. A successful case of pregnancy in a woman with ACTH-independent Cushing's syndrome treated with ketoconazole and metyrapone. Gynecol Endocrinol. 2017;33(5):349-52.
  78. Costenaro F, Rodrigues TC, de Lima PB, Ruszczyk J, Rollin G, Czepielewski MA. A successful case of Cushing's disease pregnancy treated with ketoconazole. Gynecol Endocrinol. 2015;31(3):176-8.
  79. Amado JA, Pesquera C, Gonzalez EM, Otero M, Freijanes J, Alvarez A. Successful treatment with ketoconazole of Cushing's syndrome in pregnancy. Postgrad Med J. 1990;66(773):221-3.
  80. Berwaerts J, Verhelst J, Mahler C, Abs R. Cushing's syndrome in pregnancy treated by ketoconazole: case report and review of the literature. Gynecol Endocrinol. 1999;13(3):175-82.
  81. Khir AS, How J, Bewsher PD. Successful pregnancy after cyproheptadine treatment for Cushing's disease. Eur J Obstet Gynecol Reprod Biol. 1982;13(6):343-7.
  82. Magkou D, Do Cao C, Bouvattier C, Douillard C, de Marcellus C, Cazabat L, et al. Foetal exposure to mitotane/Op'DDD: Post-natal study of four children. Clin Endocrinol (Oxf). 2018;89(6):805-12.
  83. Leiba S, Weinstein R, Shindel B, Lapidot M, Stern E, Levavi H, et al. The protracted effect of o,p'-DDD in Cushing's disease and its impact on adrenal morphogenesis of young human embryo. Ann Endocrinol (Paris). 1989;50(1):49-53.
  84. Hanson TJ, Ballonoff LB, Northcutt RC. Letter: Amino-glutethimide and pregnancy. JAMA. 1974;230(7):963-4.
  85. Woo I, Ehsanipoor RM. Cabergoline therapy for Cushing disease throughout pregnancy. Obstet Gynecol. 2013;122(2 Pt 2):485-7.
  86. Nakhleh A, Saiegh L, Reut M, Ahmad MS, Pearl IW, Shechner C. Cabergoline treatment for recurrent Cushing's disease during pregnancy. Hormones (Athens). 2016;15(3):453-8.
  87. Albert E, Dalaker K, Jorde R, Berge LN. Addison's disease and pregnancy. Acta Obstet Gynecol Scand. 1989;68(2):185-7.
  88. Anand G, Beuschlein F. MANAGEMENT OF ENDOCRINE DISEASE: Fertility, pregnancy and lactation in women with adrenal insufficiency. Eur J Endocrinol. 2018;178(2):R45-R53.
  89. Erichsen MM, Husebye ES, Michelsen TM, Dahl AA, Løvås K. Sexuality and fertility in women with Addison's disease. J Clin Endocrinol Metab. 2010;95(9):4354-60.
  90. Yuen KC, Chong LE, Koch CA. Adrenal insufficiency in pregnancy: challenging issues in diagnosis and management. Endocrine. 2013;44(2):283-92.
  91. Chandrashekar V, Zaczek D, Bartke A. The consequences of altered somatotropic system on reproduction. Biol Reprod. 2004;71(1):17-27.
  92. Milardi D, Giampietro A, Baldelli R, Pontecorvi A, De Marinis L. Fertility and hypopituitarism. J Endocrinol Invest. 2008;31(9 Suppl):71-4.
  93. Kübler K, Klingmüller D, Gembruch U, Merz WM. High-risk pregnancy management in women with hypopituitarism. J Perinatol. 2009;29(2):89-95.
  94. MacKinnon R, Eubanks A, Shay K, Belson B. Diagnosing and managing adrenal crisis in pregnancy: A case report. Case Rep Womens Health. 2021;29:e00278.
  95. Drucker D, Shumak S, Angel A. Schmidt's syndrome presenting with intrauterine growth retardation and postpartum addisonian crisis. Am J Obstet Gynecol. 1984;149(2):229-30.
  96. Ambrosi B, Barbetta L, Morricone L. Diagnosis and management of Addison's disease during pregnancy. J Endocrinol Invest. 2003;26(7):698-702.
  97. Gradden C, Lawrence D, Doyle PM, Welch CR. Uses of error: Addison's disease in pregnancy. Lancet. 2001;357(9263):1197.
  98. O'Shaughnessy RW, Hackett KJ. Maternal Addison's disease and fetal growth retardation. A case report. J Reprod Med. 1984;29(10):752-6.
  99. Seaward PG, Guidozzi F, Sonnendecker EW. Addisonian crisis in pregnancy. Case report. Br J Obstet Gynaecol. 1989;96(11):1348-50.
  100. Gamlen TR, Aynsley-Green A, Irvine WJ, McCallum CJ. Immunological studies in the neonate of a mother with Addison's disease and diabetes mellitus. Clin Exp Immunol. 1977;28(1):192-5.
  101. Langlois F, Lim DST, Fleseriu M. Update on adrenal insufficiency: diagnosis and management in pregnancy. Curr Opin Endocrinol Diabetes Obes. 2017;24(3):184-92.
  102. Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, et al. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(2):364-89.
  103. Lebbe M, Arlt W. What is the best diagnostic and therapeutic management strategy for an Addison patient during pregnancy? Clin Endocrinol (Oxf). 2013;78(4):497-502.
  104. Kazlauskaite R, Evans AT, Villabona CV, Abdu TA, Ambrosi B, Atkinson AB, et al. Corticotropin tests for hypothalamic-pituitary- adrenal insufficiency: a metaanalysis. J Clin Endocrinol Metab. 2008;93(11):4245-53.
  105. Maghnie M, Uga E, Temporini F, Di Iorgi N, Secco A, Tinelli C, et al. Evaluation of adrenal function in patients with growth hormone deficiency and hypothalamic-pituitary disorders: comparison between insulin-induced hypoglycemia, low-dose ACTH, standard ACTH and CRH stimulation tests. Eur J Endocrinol. 2005;152(5):735-41.
  106. Magnotti M, Shimshi M. Diagnosing adrenal insufficiency: which test is best--the 1-microg or the 250-microg cosyntropin stimulation test? Endocr Pract. 2008;14(2):233-8.
  107. Fiad TM, Kirby JM, Cunningham SK, McKenna TJ. The overnight single-dose metyrapone test is a simple and reliable index of the hypothalamic-pituitary-adrenal axis. Clin Endocrinol (Oxf). 1994;40(5):603-9.
  108. Beck P, Eaton CJ, Young IS, Kupperman HS. Metyrapone response in pregnancy. Am J Obstet Gynecol. 1968;100(3):327-30.
  109. Kenny FM, Preeyasombat C, Spaulding JS, Migeon CJ. Cortisol production rate. IV. Infants born of steroid-treated mothers and of diabetic mothers. Infants with trisomy syndrome and with anencephaly. Pediatrics. 1966;37(6):960-6.
  110. Sidhu RK, Hawkins DF. Prescribing in pregnancy. Corticosteroids. Clin Obstet Gynaecol. 1981;8(2):383-404.
  111. Normington EA, Davies D. Hypertension and oedema complicating pregnancy in Addison's disease. Br Med J. 1972;2(5806):148-9.
  112. Bachelot A, Grouthier V, Courtillot C, Dulon J, Touraine P. MANAGEMENT OF ENDOCRINE DISEASE: Congenital adrenal hyperplasia due to 21-hydroxylase deficiency: update on the management of adult patients and prenatal treatment. Eur J Endocrinol. 2017;176(4):R167-R81.
  113. White PC, Speiser PW. Congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Endocr Rev. 2000;21(3):245-91.
  114. Mulaikal RM, Migeon CJ, Rock JA. Fertility rates in female patients with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. N Engl J Med. 1987;316(4):178-82.
  115. Gomes LG, Bachega TASS, Mendonca BB. Classic congenital adrenal hyperplasia and its impact on reproduction. Fertil Steril. 2019;111(1):7-12.
  116. Strandqvist A, Falhammar H, Lichtenstein P, Hirschberg AL, Wedell A, Norrby C, et al. Suboptimal psychosocial outcomes in patients with congenital adrenal hyperplasia: epidemiological studies in a nonbiased national cohort in Sweden. J Clin Endocrinol Metab. 2014;99(4):1425-32.
  117. Casteràs A, De Silva P, Rumsby G, Conway GS. Reassessing fecundity in women with classical congenital adrenal hyperplasia (CAH): normal pregnancy rate but reduced fertility rate. Clin Endocrinol (Oxf). 2009;70(6):833-7.
  118. Lekarev O, New MI. Adrenal disease in pregnancy. Best Pract Res Clin Endocrinol Metab. 2011;25(6):959-73.
  119. Heland S, Hewitt JK, McGillivray G, Walker SP. Preventing female virilisation in congenital adrenal hyperplasia: The controversial role of antenatal dexamethasone. Aust N Z J Obstet Gynaecol. 2016;56(3):225-32.
  120. Tardy-Guidollet V, Menassa R, Costa JM, David M, Bouvattier-Morel C, Baumann C, et al. New management strategy of pregnancies at risk of congenital adrenal hyperplasia using fetal sex determination in maternal serum: French cohort of 258 cases (2002-2011). J Clin Endocrinol Metab. 2014;99(4):1180-8.
  121. Bolnick AD, Fritz R, Jain C, Kadam L, Bolnick JM, Kilburn BA, et al. Trophoblast Retrieval and Isolation From the Cervix for Noninvasive, First Trimester, Fetal Gender Determination in a Carrier of Congenital Adrenal Hyperplasia. Reprod Sci. 2016;23(6):717-22.
  122. New MI, Tong YK, Yuen T, Jiang P, Pina C, Chan KC, et al. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab. 2014;99(6):E1022-30.
  123. Khattab A, Yuen T, Sun L, Yau M, Barhan A, Zaidi M, et al. Noninvasive Prenatal Diagnosis of Congenital Adrenal Hyperplasia. Endocr Dev. 2016;30:37-41.
  124. Speiser PW, Azziz R, Baskin LS, Ghizzoni L, Hensle TW, Merke DP, et al. Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95(9):4133-60.
  125. Forest MG, Morel Y, David M. Prenatal treatment of congenital adrenal hyperplasia. Trends Endocrinol Metab. 1998;9(7):284-9.
  126. Coleman MA, Honour JW. Reduced maternal dexamethasone dosage for the prenatal treatment of congenital adrenal hyperplasia. BJOG. 2004;111(2):176-8.
  127. Riester A, Reincke M. Progress in primary aldosteronism: mineralocorticoid receptor antagonists and management of primary aldosteronism in pregnancy. Eur J Endocrinol. 2015;172(1):R23-30.
  128. Landau E, Amar L. Primary aldosteronism and pregnancy. Ann Endocrinol (Paris). 2016;77(2):148-60.
  129. Krysiak R, Samborek M, Stojko R. Primary aldosteronism in pregnancy. Acta Clin Belg. 2012;67(2):130-4.
  130. Kosaka K, Onoda N, Ishikawa T, Iwanaga N, Yamamasu S, Tahara H, et al. Laparoscopic adrenalectomy on a patient with primary aldosteronism during pregnancy. Endocr J. 2006;53(4):461-6.
  131. Sadeck LS, Fernandes M, Silva SM, Trindade AA, Chia CY, Ramos JL, et al. [Captopril use in pregnancy and its effects on the fetus and the newborn: case report]. Rev Hosp Clin Fac Med Sao Paulo. 1997;52(6):328-32.
  132. Morton A. Primary aldosteronism and pregnancy. Pregnancy Hypertens. 2015;5(4):259-62.
  133. Hecker A, Hasan SH, Neumann F. Disturbances in sexual differentiation of rat foetuses following spironolactone treatment. Acta Endocrinol (Copenh). 1980;95(4):540-5.
  134. Morton A, Panitz B, Bush A. Eplerenone for gitelman syndrome in pregnancy. Nephrology (Carlton). 2011;16(3):349.
  135. Cabassi A, Rocco R, Berretta R, Regolisti G, Bacchi-Modena A. Eplerenone use in primary aldosteronism during pregnancy. Hypertension. 2012;59(2):e18-9.
  136. Ghela, Anila. A challenging case of primary aldosteronism presenting in pregnancy. Endocrine Abstracts 2017.
  137. Quartermaine G, Lambert K, Rees K, Seed PT, Dhanjal MK, Knight M, et al. Hormone-secreting adrenal tumours cause severe hypertension and high rates of poor pregnancy outcome; a UK Obstetric Surveillance System study with case control comparisons. BJOG. 2018;125(6):719-27.
  138. Wyckoff JA, Seely EW, Hurwitz S, Anderson BF, Lifton RP, Dluhy RG. Glucocorticoid-remediable aldosteronism and pregnancy. Hypertension. 2000;35(2):668-72.
  139. Mulatero P, di Cella SM, Williams TA, Milan A, Mengozzi G, Chiandussi L, et al. Glucocorticoid remediable aldosteronism: low morbidity and mortality in a four-generation italian pedigree. J Clin Endocrinol Metab. 2002;87(7):3187-91.
  140. Harrington JL, Farley DR, van Heerden JA, Ramin KD. Adrenal tumors and pregnancy. World J Surg. 1999;23(2):182-6.
  141. Lenders JW. Pheochromocytoma and pregnancy: a deceptive connection. Eur J Endocrinol. 2012;166(2):143-50.
  142. Sarathi V, Lila AR, Bandgar TR, Menon PS, Shah NS. Pheochromocytoma and pregnancy: a rare but dangerous combination. Endocr Pract. 2010;16(2):300-9.
  143. Bancos I, Atkinson E, Eng C, Young WF, Neumann HPH, Group IPaPS. Maternal and fetal outcomes in phaeochromocytoma and pregnancy: a multicentre retrospective cohort study and systematic review of literature. Lancet Diabetes Endocrinol. 2021;9(1):13-21.
  144. Reisch N, Peczkowska M, Januszewicz A, Neumann HP. Pheochromocytoma: presentation, diagnosis and treatment. J Hypertens. 2006;24(12):2331-9.
  145. Santos DR, Barbisan CC, Marcellini C, dos Santos RM. Pheochromocytoma and pregnancy: A case report and review. J Bras Nefrol. 2015;37(4):496-500.
  146. Oliva R, Angelos P, Kaplan E, Bakris G. Pheochromocytoma in pregnancy: a case series and review. Hypertension. 2010;55(3):600-6.
  147. Cross DA, Meyer JS. Postoperative deaths due to unsuspected pheochromocytoma. South Med J. 1977;70(11):1320-1.
  148. Levin N, McTighe A, Abdel-Aziz MI. Extra-adrenal pheochromocytoma in pregnancy. Md State Med J. 1983;32(5):377-9.
  149. Wing LA, Conaglen JV, Meyer-Rochow GY, Elston MS. Paraganglioma in Pregnancy: A Case Series and Review of the Literature. J Clin Endocrinol Metab. 2015;100(8):3202-9.
  150. Natrajan PG, McGarrigle HH, Lawrence DM, Lachelin GC. Plasma noradrenaline and adrenaline levels in normal pregnancy and in pregnancy-induced hypertension. Br J Obstet Gynaecol. 1982;89(12):1041-5.
  151. Kudva YC, Sawka AM, Young WF. Clinical review 164: The laboratory diagnosis of adrenal pheochromocytoma: the Mayo Clinic experience. J Clin Endocrinol Metab. 2003;88(10):4533-9.
  152. Khatun S, Kanayama N, Belayet HM, Bhuiyan AB, Jahan S, Begum A, et al. Increased concentrations of plasma neuropeptide Y in patients with eclampsia and preeclampsia. Am J Obstet Gynecol. 2000;182(4):896-900.
  153. Pedersen EB, Rasmussen AB, Christensen NJ, Johannesen P, Lauritsen JG, Kristensen S, et al. Plasma noradrenaline and adrenaline in pre-eclampsia, essential hypertension in pregnancy and normotensive pregnant control subjects. Acta Endocrinol (Copenh). 1982;99(4):594-600.
  154. van der Weerd K, van Noord C, Loeve M, Knapen MFCM, Visser W, de Herder WW, et al. ENDOCRINOLOGY IN PREGNANCY: Pheochromocytoma in pregnancy: case series and review of literature. Eur J Endocrinol. 2017;177(2):R49-R58.
  155. Remón-Ruiz P, Aliaga-Verdugo A, Guerrero-Vázquez R. Pheochromocytoma in neurofibromatosis type 1 during pregnancy. Gynecol Endocrinol. 2017;33(2):93-5.
  156. Petr EJ, Else T. Pheochromocytoma and Paraganglioma in Neurofibromatosis type 1: frequent surgeries and cardiovascular crises indicate the need for screening. Clin Diabetes Endocrinol. 2018;4:15.
  157. Kaluarachchi VTS, Bulugahapitiya U, Arambewela M, Gunathilake S. Successful Management of Pheochromocytoma Detected in Pregnancy by Interval Adrenalectomy in a VHL Patient. Case Rep Endocrinol. 2018;2018:9014585.
  158. Biggar MA, Lennard TW. Systematic review of phaeochromocytoma in pregnancy. Br J Surg. 2013;100(2):182-90.
  159. Tingi E, Kyriacou A, Verghese L. Recurrence of phaeochromocytoma in pregnancy in a patient with multiple endocrine neoplasia 2A: a case report and review of literature. Gynecol Endocrinol. 2016;32(11):875-80.
  160. Ellison GT, Mansberger JA, Mansberger AR. Malignant recurrent pheochromocytoma during pregnancy: case report and review of the literature. Surgery. 1988;103(4):484-9.
  161. Liu S, Song A, Zhou X, Kong X, Li WA, Wang Y, et al. Malignant pheochromocytoma with multiple vertebral metastases causing acute incomplete paralysis during pregnancy: Literature review with one case report. Medicine (Baltimore). 2017;96(44):e8535.
  162. Remmel E, Oertli D, Holzgreve W, Hösli I, Staub JJ, Harder F. [Malignant pheochromocytoma in the 17th week of gestation]. Chirurg. 1999;70(9):1053-7.
  163. Iijima S. Impact of maternal pheochromocytoma on the fetus and neonate. Gynecol Endocrinol. 2019:1-7.
  164. Saarikoski S. Fate of noradrenaline in the human foetoplacental unit. Acta Physiol Scand Suppl. 1974;421:1-82.
  165. Dahia PL, Hayashida CY, Strunz C, Abelin N, Toledo SP. Low cord blood levels of catecholamine from a newborn of a pheochromocytoma patient. Eur J Endocrinol. 1994;130(3):217-9.
  166. Lyons CW, Colmorgen GH. Medical management of pheochromocytoma in pregnancy. Obstet Gynecol. 1988;72(3 Pt 2):450-1.
  167. Aplin SC, Yee KF, Cole MJ. Neonatal effects of long-term maternal phenoxybenzamine therapy. Anesthesiology. 2004;100(6):1608-10.
  168. Mohamed Ismail NA, Abd Rahman R, Abd Wahab N, Muhammad R, Nor Azmi K. Pheochromocytoma and pregnancy: a difficult and dangerous ordeal. Malays J Med Sci. 2012;19(1):65-8.
  169. Hall DR, Odendaal HJ, Steyn DW, Smith M. Nifedipine or prazosin as a second agent to control early severe hypertension in pregnancy: a randomised controlled trial. BJOG. 2000;107(6):759-65.
  170. Versmissen J, Koch BC, Roofthooft DW, Ten Bosch-Dijksman W, van den Meiracker AH, Hanff LM, et al. Doxazosin treatment of phaeochromocytoma during pregnancy: placental transfer and disposition in breast milk. Br J Clin Pharmacol. 2016;82(2):568-9.
  171. Tanaka K, Tanaka H, Kamiya C, Katsuragi S, Sawada M, Tsuritani M, et al. Beta-Blockers and Fetal Growth Restriction in Pregnant Women With Cardiovascular Disease. Circ J. 2016;80(10):2221-6.
  172. Boutroy MJ. Fetal and neonatal effects of the beta-adrenoceptor blocking agents. Dev Pharmacol Ther. 1987;10(3):224-31.
  173. Prete A, Paragliola RM, Salvatori R, Corsello SM. MANAGEMENT OF CATECHOLAMINE-SECRETING TUMORS IN PREGNANCY: A REVIEW. Endocr Pract. 2016;22(3):357-70.
  174. Muzannara MA, Tawfeeq N, Nasir M, Al Harbi MK, Geldhof G, Dimitriou V. Vaginal delivery in a patient with pheochromocytoma, medullary thyroid cancer, and primary hyperparathyroidism (multiple endocrine neoplasia type 2A, Sipple's syndrome). Saudi J Anaesth. 2014;8(3):437-9.
  175. Kapoor G, Salhan S, Sarda N, Sarda AK, Aggarwal D. Phaeochromocytoma in pregnancy: safe vaginal delivery, is it possible? J Indian Med Assoc. 2013;111(4):266-7.
  176. Morton A, Poad D, Harms P, Lambley J. Phaeochromocytoma in pregnancy: timing of surgery, mode of delivery and magnesium. Obstet Med. 2010;3(4):164-5.

 

Special Considerations Relevant to Pediatric Obesity

ABSTRACT

 

In most humans, body fatness is a quantitative trait reflecting the interactions of environment, genotype, and development. The metabolic predisposition to obesity and its co-morbidities in adulthood begins in the intrauterine environment, extends into early childhood, and is further impacted by puberty.  An understanding of the pathogenesis of obesity in children, and its implications for the risk of obesity in adulthood, has the potential to inform healthcare providers about early identification and use of precision medicine approaches towards both prevention and treatment. This chapter begins with a review of the epidemiology and definition of pediatric obesity followed by a discussion of risk factors for adult obesity from genetics to the prenatal environment (epigenetics) through childhood. The next section emphasizes that while some adiposity-related problems are unique to the pediatric population, multi-system co-morbidities of adult obesity are increasingly prevalent in children. The chapter concludes with a discussion of recommendations for intervention(s) and an invitation for providers to engage federal and local governments in discussions of ways to unite families, schools, and communities in the battle against the costliest nutritional problem for children in the United States.

 

INTRODUCTION

 

Obesity and its co-morbidities currently account for over $250 billion per year in health care costs (~25% of total U.S. health care budget (1)) and is projected to increase to over $900 billion by the year 2030 (2). Obesity is a complex disease reflecting interactions of an increasingly permissive environment on a background of genetic predisposition and developmental programming (3-7). Results from 2017- March 2020 National Health and Nutrition Examination Survey (NHANES, Figure 1A) indicate that an estimated 21.5% of U.S. children and adolescents aged 2-19 years have obesity (Body mass index [BMI] > 95th percentile for age and sex), an increase of 25% over the last decade, and 6.1% have severe obesity (BMI > 125% of 95th percentile for age and sex) (6,8,9). The prevalence of obesity is significantly higher in non-Hispanic Black and Hispanic children (Figure 1B). By adolescence, the prevalence increases to two-fold and nearly three-fold in these groups respectively compared to their non-Hispanic White or Asian counterparts.

Figure 1. Prevalence of obesity in youth. A. Trends in prevalence of obesity by age group in the past decade years (281). B. Obesity prevalence in youth by age, race, and Hispanic origin in United States, generated using data from National health and nutrition examination survey 2015-2018 (n=6710). Obesity was defined as BMI ≥ 95th percentile for youth 2-20 years of age using CDC 2000 growth charts and weight-for-height ≥ 97.7th percentile from birth to 2 years using WHO growth charts (282).

The prevalence of pediatric obesity and its co-morbidities, such as type 2 diabetes mellitus (T2DM), have been increasing in parallel. Pediatric obesity tracks into adulthood, especially if present in the peri-pubertal period (over 20-fold increased risk adult obesity) and if one or both parents have obesity (10). These problems disproportionately affect Black, Hispanic, and Native American communities (8,11,12). It is also worth considering that although a higher percentage of adults have obesity, the fractional magnitude of obesity prevalence among youth is growing faster. From 1971-2018 the prevalence of adults with obesity increased by about 2.8 fold (from about 15% to 42%) whereas the percentage of children with obesity increased by 3.8 fold (from about 5% to 19.7%) (8). Recent CDC reports using data from comprehensive electronic health records indicate that the monthly rate of BMI increase in children aged 2-19 years nearly doubled during the COVID pandemic period (0.100 versus 0.052 kg/m2/month; ratio = 1.93) and the prevalence of obesity increased from 19.3% in August of 2019 (pre-pandemic) to 22.4% in August of 2020. Children with higher BMI z-scores and between the ages of 6 and 11 years were most affected. (13).

 

Adults entering non-surgical weight loss treatment will typically lose weight for approximately 6-8 months followed by inexorable weight regain. Overall, only about 15% of adults with obesity are able to lose and sustain a greater than 10% weight loss, even with intensive lifestyle or pharmacological interventions. This number has not changed in over 20 years despite multiple new pharmacological and other treatment options (14-17). A key question is whether or not children are more responsive to interventions to treat, or to prevent, obesity.

 

Reviews of large lifestyle weight loss intervention studies indicate that children are more successful than adults in sustaining weight loss (usually defined by BMI z-score) provided that they remain involved in the intervention (see treatment discussions below) and that earlier intervention is more effective. Reinehr et al (18), performed retrospective quality assessments of 129 pediatric obesity programs at 6, 12, and 24 months and found that reduction of overweight was achieved by 83% of the children after 6 months, by 82% after 12 months, and by 76% after 24 months. The mean change of SDS-BMI was −0.20 ± 0.32 at 6 months, −0.19 ± 0.40 at 12 months, and −0.20 ± 0.54 at 24 months indicating an average of about 8% sustained reduction in adiposity. In adults with T2DM and overweight or obesity enrolled in the prospective LookAHEAD trial (15), the 1 year weight loss in the intensive lifestyle intervention group was 8.6± 0.1% (similar to children) but by year 2 it had fallen to 6.4±0.2% and by year 4 to 4.7±0.2% despite continued intervention.

Figure 2. Patterns of weight loss and regain in children and adults. A. On average, adults will lose weight for only about 6-9 months during lifestyle intervention to treat obesity. After this, most will then begin to regain weight (283-286). B. In contrast, children tend to lose more fatness (expressed as BMI z-score) and sustain their weight loss longer following a lifestyle intervention (61,62). This is especially true for younger children. *Based on DeJonge et al (286). †Based on Kraschnewski et al (16).

Meta-analyses of pediatric weight loss lifestyle interventions have generally noted reductions in BMI, BMI z score and weight that are influenced by both the type and duration of the intervention (19,20) and exceed results seen with lifestyle interventions in adults especially if intervention is initiated early. Figure 2 illustrates examples of patterns of weight loss followed by weight regain seen after lifestyle interventions in adults (Figure 2A) and children (Figure 2B). It is clear that most adults will lose a smaller fraction of total body fat and are less likely to sustain that loss than are children, and that younger children are more likely to reduce body fatness and keep it off than older children or adults. Similar patterns of weight loss and weight regain are seen in pharmacological interventions to treat adults with obesity.

 

These data indicate that there is a greater likelihood of successful treatment of obesity and reduced weight maintenance in children than adults but must be interpreted caution. Adult studies, such as LookAHEAD involve a continuous active intervention that may include medication while pediatric studies, such as Obeldisks (11) (Figure 2) are only single year interventions with intermittent follow-up. Prospective pediatric studies such as Obeldisks may have a much higher attrition rate (about 70%) than adult studies such as LookAHEAD (about 10%), perhaps due to less contact with participants and type of intervention, as well as participant retention resulting in a smaller and less diverse study population of pediatric completers.

 

DEFINITION AND EPIDEMIOLOGY

 

The ideal diagnostic criteria for pediatric obesity would include some assessment of adiposity-related co-morbidity, the risk of persistence of the obesity into adulthood, as well as the risk of future morbidities that would be worsened by excess weight.

 

Several basic principles are pertinent to such an assessment:

  • During the first year of life there is an increase in weight for height followed by a decline and a second increase at about 6 years of age (designated as “adiposity rebound”). Early adiposity rebound, prior to 5 years of age, is associated with a higher risk of adult obesity (21,22).
  • The risk of persistence of pediatric obesity into adulthood increases with age, independent of the length of time that the child has been obese (3,23).
  • Growth patterns are familial and may be predictive of adult adiposity. A mildly overweight adolescent with a family history of adult obesity may be at greater risk for subsequent obesity than a severely overweight youth with a negative family history (3,23).
  • The risk of adiposity-related morbidity is strongly influenced by family history, regardless of obesity in the affected family members, and varies between racial/ethnic groups (3,23,24).

 

BMI is often used as a “surrogate” for body fatness. Although it does not measure body fat, it correlates with direct measures of body fatness within a population (25,26).  In adults, obesity is frequently divided into categories– Class 1: BMI of 30-35 kg/m2; Class 2: BMI of 35-40 kg/m2; and Class 3: BMI ≥ 40 kg/m2. Class 3 is also categorized as “severe” obesity. These definitions cannot be used in children because normative values for BMI are age- and sex-dependent (27). In 2007, the AAP Expert Committee recommended that children between the ages of 2-19 years with BMI > 95th percentile are classified as “obese” and those with a BMI between the 85th and 95th percentile are classified as “overweight” (28) using the 2000 Centers for Disease Control (CDC) growth charts. These charts were constructed from data collected between 1963-1980, that included lambda-mu-sigma (LMS) parameters to calculate ten smoothed percentiles between 3rd and 97th percentile (28).  However, extreme percentiles for heavier children extrapolated using CDC LMS parameters did not match well to the empirical data for the 99th percentile obtained in the later years. Instead, a better fit to the empirical data was obtained by using 120% of the smoothed 95th percentiles (29). This modification gave rise to the extended BMI growth charts that provides a flexible approach to describe and track children with obesity. (30). The American Health Association recommended classification of BMI ≥ 120% of 95thpercentile as severe (equivalent to Class 2) obesity (31). Subsequent publications have defined overweight as BMI between 85-95th percentile, Class 1 obesity as BMI between 95th- 120% of 95th percentile, Class 2 between 120% -140% of 95th percentile and Class 3 as ≥ 140% of 95th percentile, making the classification similar to that used in adults (32-34) (Figure 3). For children less than two years of age weight/recumbent length ≥ 97.7th percentile based on the World Health Organization (WHO) charts is currently used to define obesity (24,35).

Figure 3. Normative BMI growth curves for boys and girls. Extended BMI curves for youth aged 2-20 years of age based on Gulati et al (30). Class 1 obesity defined as BMI between 95th- 120% of 95th percentile; Class 2 between 120%-140% of 9th percentile; Class 3 ≥ 140% of 95th percentiles (287).

Normative data have also been established for waist circumference during childhood (Figure 4).  Waist circumference is measured at the level of the upper border of the right superior iliac crest with horizontal alignment of the measuring tape, parallel to the floor, lying snug, but not compressing the skin. These data are most helpful in identifying children at risk for insulin resistance, type 2 diabetes, and dyslipidemia. The limitation of waist circumference is in the difficulty in properly locating anatomic landmarks such as the umbilicus and superior iliac crest, especially in individuals with severe obesity and/or a large volume of subcutaneous adipose tissue.  

 

As noted above, BMI does not directly measure body fat.  Individuals at either extreme (low or high) of percent body fat may be incorrectly labeled solely based on BMI. In such cases, if the clinician is uncertain, further evaluation may require more precise methods of assessing body fat such as bioelectrical spectroscopy (BIS), air displacement plethysmography (BOD POD), dual-energy X-ray absorptiometry (DEXA) scanning, or Quantitative Magnetic Resonance (QMR) (36,37).

 

Obesity in childhood and adolescence predisposes to obesity in adulthood. In a meta-analysis of 200,777 subjects derived from fifteen prospective studies, Simmonds et al showed that youth with obesity were five times more likely to have obesity in adulthood. Over half of the individuals with obesity in childhood will have obesity in adolescence and nearly 80% of adolescents with obesity will continue to have obesity in adulthood (38). In a separate meta-analysis of thirty-seven studies, the same group showed that high childhood BMI was associated with an increased incidence of adult diabetes (OR 1.70, 95% CI 1.30-2.22), coronary heart disease (OR 1.20, 95% CI 1.10-1.31), and a range of obesity associated cancers (39). It should be noted that while childhood obesity persists when present, not all adults with obesity or its associated co-morbidities had obesity in childhood, re-emphasizing that obesity is a result of complex interaction between familial predisposition, likely from genetics, and the environment.

Figure 4. Waist circumference (measured at the iliac crest while subjects stood and placed their hands on opposite shoulders) curves for North American Children age 5-19 years derived from NHANES III data (196) by the Canadian Pediatric Endocrine Group (https://cpeg-gcep.net/content/waist-circumference-and-waist-height-ratio-charts). Charted indices for these variables at extreme of body fatness are currently not available.

In addition to initiating therapy in childhood when it is more likely to be effective, it is also important to identify the child who is “at-risk” of becoming an obese adult. The risk of adult obesity is higher in children with a first degree relative with obesity and also increases as the child approaches puberty. Whitaker et al (23), examined health records from 854 subjects born at a health maintenance organization in Washington State between 1965 and 1971 and tracked them into early adulthood (age 21-29 years). The odds ratio for a child with obesity (defined as BMI > 85%ile for age and sex) becoming an adult with obesity rose steadily from 1.3 at age 1-2 years, to 22.3 at age 10-14 years, and 17.5 at age 15 to 17 years of age. In contrast, the effects of parental obesity on odds ratio decreased with age from 3.2 at age 1-2 years to 2.2 at age 15-17 years. More studies like this using larger populations will be informative regarding predictors of having obesity in adulthood.

 

ENERGY HOMEOSTASIS

 

The first law of thermodynamics dictates that the accumulation of stored energy (fat) must be due to caloric intake more than energy expenditure. A sustained small excess of energy intake relative to expenditure will, over time, lead to a substantial increase in body weight.  For example, a 50 kg individual who increases their daily caloric intake by 150 kcal (8 ounces of whole milk) above their usual daily energy expenditure (~1800 kcal/day) would gain approximately 8 pounds before sufficient fat-free mass (FFM) was reached to result in a new equilibrium between energy intake and expenditure (assuming approximately 30% of weight gain is FFM).  This assumes, however, that there were no metabolic adaptations to maintain body energy stores in the face of overnutrition (40,41). In fact, adults maintain a relatively constant body weight, and most children tend to grow steadily along their respective weight percentile isobars for age, with little conscious effort to regulate energy intake or expenditure, despite the potentially large effects of small imbalances in energy intake versus expenditure.

 

The high rate of recidivism to previous levels of fatness by reduced-obese children and adults (42-47), and the tendency for individuals to maintain a relatively stable body weight over long periods of time despite variations in caloric intake (48), provide empirical evidence that body weight is regulated.  It is now known that energy intake and expenditure are responsive to complex interlocking control mechanisms in which numerous afferent signals from the gastrointestinal, endocrine, central and peripheral nervous system, and adipose organs are ‘sensed’ by central nervous system tracts whose efferent systems affect energy intake and expenditure so as to maintain (or restore) weight (40,49).  Adding to the complexity of this system’s interactions, the amount of energy stored in the body as fat also exerts potent effects on growth, pubescence, fertility, autonomic nervous system activity, and thyroid function, suggesting that humoral “signals” reflecting adipose tissue mass interact directly or indirectly with many neuroendocrine systems (40,50-54). Weight loss and maintenance of a reduced body weight are accompanied by changes in autonomic nervous system function (increased parasympathetic and decreased sympathetic nervous system tone), circulating concentrations of thyroid hormones (decreased triiodothyronine and thyroxine without a compensatory increase in TSH) (55-58), and appetite (increased hunger, reduced sense of fullness) (59) that are consistent with a homeostatic resistance to altered body weight, acting, in part, through effectors that mediate energy expenditure and intake.

 

Such a neurohumoral system to protect body energy stores would convey clear evolutionary advantages.  During periods of undernutrition, the perceived reduction in energy stores would result in hyperphagia, hypometabolism, and decreased fertility (protecting females from the increased metabolic demands of pregnancy and lactation and the delivery of progeny into inhospitable environments). While carefully controlled studies of the effects of weight loss on energy expenditure in children are not yet available, the higher success rates in sustained fatness reduction in younger children versus adults discussed above suggests that these same systems appear to be more malleable in children prior to puberty (60-62).

 

MOLECULAR GENETICS OF BODY FATNESS

 

Heritability of Body Fatness

 

The storage of excess calories as fat would have been highly advantageous to our progenitors by increasing survival during periods of prolonged caloric restriction and conferring a reproductive advantage.  The opportunities for our distant forebears to consume calories to the point of becoming morbidly obese and the likelihood of their survival to an age at which such co-morbidities as T2DM, hypertension, or hyperlipidemia were both low. Thus, it is likely through natural selection that the human genome would be enriched with genes favoring the storage of calories as adipose tissue (63,64). Conversely, there would be few, if any, evolutionary pressures to discourage obesity and ‘defend’ body thinness.

 

With the possible exceptions of the rare cases of obesity due to single gene mutations (see below) or specific anatomic/endocrine lesions (see above), body fatness is a quantitative trait reflecting the interaction of development and environment with genotype. Twin and adoption studies indicate that the heritability of body fatness and of body fat distribution in adulthood is 50 to 80%, [approximately equal to the heritability of height and greater than the heritability of schizophrenia (68%) or breast cancer (45%)] (65) (66). Studies have also identified significant genetic influences (heritability greater than 30%) on resting metabolic rate, feeding behavior, food preferences, and on changes in energy expenditure that occur in response to overfeeding (67-75).  Genetic influences on resting energy expenditure (REE) are evidenced by studies demonstrating that African-American children tend to have lower REE than Caucasian-American children, even when adjusted for body composition, gender, age, and pubertal status (76).

 

The calculation of heritability in twin studies assumes that each member of a monozygotic or dizygotic pair is reared in the same environment, and that the degree to which body fatness is more similar within mono- than dizygotic twin pairs is due to the greater genetic similarity of identical vs. non-identical twins.  Studies comparing adopted children with their adoptive and their biological parents assume that each child shares little or none of the immediate environment with each biological parent, and that the degree to which body fatness is more similar between children and their biologic vs. adoptive parents is due to the 50% of their genotype that each child shares with each biological parent. Based on twin studies, the heritability of body fatness appear to increase with age (77), illustrating the complex interactions of many obesity-risk allelic variants with the environment.

 

Common Single Gene Mutations Associated with Obesity

 

The pivotal role of genetics in the control of body weight is confirmed by the existence of rare single gene variants producing extreme obesity phenotype (e.g., Prader Willi, Bardet-Biedl, Alström, and Cohen syndromes). The most common monogenic cause of obesity – variants in MC4R – does not cause syndromic features, while others cause obesity in association with other distinctive dysmorphic phenotypes (67,78 ) (Table 1).  The fact that mutations in different genes can produce obesity suggests that these genes may be part of a control system for the regulation of body weight, i.e., that feeding behavior and energy expenditure are integrated in a system with complex control mechanisms which can be disrupted at many loci. Further, the impact of a genetic change may not be a direct increase in weight – as an example – recent studies of Prader Willi Syndrome have demonstrated that the endocrine phenotype is due to a deficiency in prohormone convertase, an enzyme that has also been identified as a single gene mutation cause of obesity (79,80).

 

Table 1.  Common Single Gene Mutations Associated with Obesity (67)

Syndrome/Gene

Chromosome

Phenotype

Alström syndrome/ ALMS1

2p14-p13

(Recessive)

Childhood blindness due to retinal degeneration, nerve deafness, acanthosis nigricans, chronic nephropathy, primary hypogonadism in males only, type II diabetes mellitus, infantile obesity which may diminish in adulthood.

Bardet-Biedl syndrome (22 different genes)

16q21

15q22-q23

Retinitis pigmentosa, mental retardation, polydactyly, hypothalamic hypogonadism, rarely glucose intolerance, deafness, or renal disease

Beckwith-Wiedemann syndrome

11p15.5

(Recessive)

Hyperinsulinemia, hypoglycemia, neonatal hemihypertrophy (Beckwith-Wiedemann Syndrome), intolerance of fasting

Börjeson-Forssmann-Lehman syndrome/ PHF6

X-linked

Intellectual disability, epilepsy, microcephaly, short stature, gynecomastia, hypogonadism, obesity, tapering fingers and short toes, multiple ophthalmological problems, coarse facial features, ptosis, large and long ears, supraorbital ridge

Carpenter /RAB23and MEGF8

Unknown

(Recessive)

Mental retardation, acrocephaly, poly- or syndactyly, hypogonadism (males only)

Cohen /COH1

8q22-q23

(Recessive)

Mental retardation, microcephaly, short stature, dysmorphic facies

Leptin deficiency / LEP

7q31.3

(Recessive)

Hypometabolic rate, hyperphagia, pubertal delay, infertility, impaired glucose tolerance due to leptin deficiency.

Leptin Receptor / LEPR

1p31-p32

(Recessive)

Hypometabolic rate, hyperphagia, pubertal delay due to deranged leptin signal transduction.

Melanocortin 4 Receptor /MC4R

18q22

(Dominant)

Obesity – early onset hyperphagia, increased bone density

Neisidioblastosis

11p15.1

(Recessive or Dominant)

Hyperinsulinemia, hypoglycemia, intolerance of fasting

Prader Willi syndrome

15q11-q12

(Uniparental Maternal Disomy)

Short stature, small hands and feet, mental retardation, neonatal hypotonia, failure to thrive, cryptorchidism, almond-shaped eyes and fish-mouth

Pro-opiomelanocortin / POMC

2p23.3

(Recessive)

Red hair and hyperphagia due to low POMC production of alpha-MSH in hair follicles and the hypothalamus, respectively; adrenal insufficiency due to impaired POMC production of ACTH. 

Prohormone Convertase/ PCSK1

5q15-q21

(Recessive)

Abnormal glucose homeostasis, hypogonadotropic hypogonadism, hypocortisolism, and elevated plasma proinsulin and POMC

Pseudohypo-parathyroidism (type IA, aka Albright’s) / GNAS

20q13.2

(Dominant)

Mental retardation, short stature, short metacarpals and metatarsals, short thick neck, round facies, subcutaneous calcifications, increased frequency of other endocrinopathies (hypothyroidism, hypogonadism)

 

Genome-Wide Association Studies (GWAS) of the Obesity Phenotype

 

The single gene mutations in humans listed in Table 1 are invariably associated with distinct phenotypes and marked (if not extreme) obesity. On the other hand, polygenic obesity may be phenotypically less extreme and with a more variable and subtle phenotype without any other syndromic features. GWAS of large populations have identified over 100 genetic loci as unequivocally associated with obesity-related traits (81-83) and over 500 loci associated with obesity-susceptibility (84), these allelic variants generally have been shown to exert only a small, but cumulative, effect on BMI (85).

 

In 2007, Frayling et al  (86) reported a link between a SNP in the first intron of the FTO gene (rs9939609) and obesity in a GWAS of approximately 500,000 individuals with type 2 diabetes. Individuals homozygous for this SNP (AA) were approximately three kilograms heavier and at a 1.7-fold increased risk of obesity than those who were homozygous unaffected (TT). Since then numerous other FTO-related SNP’s have been identified that are associated with BMI (87,88). These SNP’s are especially relevant to the study of childhood obesity because of their frequency (14-18% AA; 39-50% AT; and 30-35% TT (89)) and the fact that the behavioral phenotype is evident in early childhood before obesity is manifest. Cecil et al. (90) used a three-pronged preload model to quantify energy intake in 4 to 10 year-old subjects genotyped with AA, AT, and TT alleles and found that the presence of an A allele was associated with increased energy intake and caloric density (kcal/gm) of foods chosen without any effect on energy expenditure (doubly labeled water method) or compensation index for increasing preload. Wardle et al. (89) reported that 4 to 5 year-old children who were homozygous (n=24) or heterozygous (n=66) for the FTO/FTM allele (AA or AT) and had eaten a meal to satiety, ate significantly more than control subjects (n=43, TT) when offered additional food, even when corrected for body fatness. The choice of snack was limited in this latter study and, thus, the authors were unable to comment on preference for calorically dense foods. Two separate studies of large cohorts (totaling over 36,000 individuals) reported no association of FTO genotype with increased BMI prior to the age of seven (86,91). There appears to be no effect of the A allele on energy output (88,92). Thus, behaviors that are premonitory of subsequent weight are evident and measurable in pre-obese children with allelic variants of FTO. These abnormal feeding patterns associated with increased energy intake (93) including decreased dietary restraint following a caloric preload (94,95), and ratings of hunger prior to or satiety after a meal (96) are not seen in already-overweight adults. These data emphasize the importance of studying eating behavior in subjects "at risk" for weight gain to understand the dynamics of food intake that favor the development of obesity.

 

More recently, use of polygenic risk scores (combination of the risk estimate apportioned by each common variant) using as many as 2.1 million common variants has enhanced the ability to quantify the susceptibility of obesity. Khera et al(4) used such a polygenic risk score in the 7,861 participants in the Avon Longitudinal Study of Parents and Children, a birth cohort recruited between 1991-92 and longitudinally followed to 18 years of age. The birth weight of the individuals in the top decile of the polygenic risk score was 0.06 kg (p=.02) higher compared to the bottom decile. By 8 years of age, the difference increased to 3.5 kg (p < .0001) and by 18 years, the difference in weight was 12.3 kg (p <.0001). The authors postulate that the aggregation of risk for obesity that can be conferred by having many common variants approaches the susceptibility equivalent to rare monogenic mutations in MC4R.

 

 

Epigenetics

 

The term “epigenetics” was first coined in 1942 by the British developmental biologist C.H. Waddington to refer to how gene regulation modulates development. In 1990, the molecular biologist Dr. Robin Holliday re-defined the term “epigenetics” as “the study of the mechanisms of temporal and spatial control of gene activity during the development of complex organisms.” More recently this has been understood simply as the study of changes that affect the expression or “potency” of genes without necessarily affecting the nucleotide sequences of the genes themselves (97,98).

 

Epigenetics is extremely relevant to obesity in that it has allowed examination of the effects of the intrauterine environment, primarily in the form of factors affecting DNA methylation, histone acetylation, and expression of micro RNA’s, on gene expression relevant to obesity and its co-morbidities. Increased DNA methylation decreases the transcription of relevant genes and is affected by parental obesity, maternal diet (e.g., nutrition, folic acid content and other methyl donors), gestational diabetes (see below), and maternal medications (antibiotics and antipsychotics), smoking or exposure to chemicals such as bisphenol (99,100). Histones are proteins that “package” DNA into nucleosomes and post-translational modifications in the tails of histone affect the accessibility of DNA for methylation and translation. Loss of histone demethylase leads to obesity via decreased expression of PPARα and UCP1, and de-acetylation of the GLUT4 histone tail leads to impaired glucose transport (101,102).  The human genome has been suggested to contain over 1000 micro (non-coding) RNAs (miRNAs), which may influence expression of more than 60% of mammalian genes by regulating gene expression. Each miRNA can interact with expression of multiple genes, including many involved in adipogenesis (103), that play pivotal roles in the development of obesity and its co-morbidities.

Major intrauterine environmental influences on the risk of subsequent obesity via these processes and others include maternal adiposity and gestational weight gain, under- and over- nutrition, gestational diabetes, maternal stress, and various chemicals, pharmaceuticals etc., to which the mother and fetus may be exposed during pregnancy.

 

  • Maternal weight impacts the fetus at multiple levels beyond those due to obesity risk alleles that may be inherited from either parent. This is exemplified by studies of offspring of mothers before and after bariatric surgery. The genotype of the mother is unchanged yet the fatness, blood pressure, circulating concentrations of insulin and gene expression relevant to diabetes, autoimmune disease, and vascular disease risk are all reduced in children who develop in the post-bariatric surgery intrauterine environment (104). Weight gain during pregnancy has a strong positive correlation with the incidence of large for gestational age babies and subsequent childhood obesity(105) augmented 2-5 fold in mothers with pre-partum obesity compared to those who were neither overweight nor obese prior to pregnancy.

Figure 5. U-shaped curve of odds risk for obesity at age 9-11 years based on birth weight. Curve is corrected for gestational diabets, gestional age, childs age, breast or formula feeding, highest level of parental education, sleep time, moderately vigorous physical acitivvity time (MVPA), sendentary time and healthy/unhealthy diet scores (288) weight Dashed lines identify 95% confidence intervals. Inserted text boxes indicate independent effects of small for gestational age (SGA) and large for gestational age (LGA) on various health parameters in adults.

  • Pre-Natal undernutrition (see Figure 5) reflects maternal undernutrition or compromised fuel delivery to the fetus–the latter usually due to placental dysfunction. Studies that have examined the prevalence of obesity in children conceived during periods of natural or man-made famine such as the Nazi-imposed Dutch famine of 1944-45 (the “Winter Hunger”) (106) report a small but statistically significant increase in the prevalence of obesity (defined as weight for height greater than 120% of WHO standards for 1948) in 19 year-old male military recruits whose mothers were malnourished only during the first trimester of pregnancy (2.77% prevalence if mother was in famine area vs. 1.45% if mother was outside of famine area during pregnancy) and a decrease in the prevalence of obesity among recruits whose mothers were malnourished during the child’s immediate post-natal period (0.82 % if mother was in famine area vs. 1.32% if mother was outside of famine area during pregnancy). It has been hypothesized that early intrauterine malnutrition might affect hypothalamic ("appetite center") development while the anti-obesity effects of early post-natal malnutrition might be due to suppression of adipocyte formation.

 

      Long-term tracking studies of children who are small for gestational age, possibly due to prenatal undernutrition, have reported that, even when corrected for adult adiposity, birthweight is negatively correlated with the incidence of adiposity-related morbidities, including T2DM, hypertension, stroke, and cardiovascular disease, in adulthood (107-112).  This association implies an interaction between the prenatal environment and development/function of pancreatic beta-cells and other organs such as the hypothalamus, liver, and kidneys that are involved in the regulation of adult energy homeostasis and cardiovascular function. As hypothesized by Barker (113-115), the metabolic, cardiovascular, and endocrine basis for adult adiposity-related morbidities may originate through adaptations that the fetus makes in response to undernourishment, especially when availability of calories in the environment that baby is born into is no longer limited. Therefore, the small-for-gestational-age baby should be considered to be at increased risk for adult morbidities that are exacerbated by increased adiposity (63).

 

  • Pre-Natal over nutrition (see Figure 5) is exemplified by the infant of a mother with gestational diabetes mellitus (GDM). The high ambient glucose concentrations of the prenatal environment stimulate fetal hyperinsulinemia, increased lipogenesis, and macrosomia.  Since women with gestational diabetes are often overweight or obese, it is difficult to separate the metabolic effects of gestational diabetes on subsequent adiposity of offspring of mothers with GDM from the possibility that the mother has transmitted a genetic tendency towards obesity.  Yet several studies have shown that GDM is associated with an increased risk of obesity in the offspring, independent of the degree of maternal obesity (116-119).

 

  • Maternal stress, which can be metabolic (e.g., obesity, diabetes, undernutrition, illness), psychiatric (e.g., depression, anxiety, bereavement), or pharmacological (e.g., steroids, antidepressants, antibiotics) have all been associated with increased risk of offspring obesity. These stressors affect developing neural systems regulating energy homeostasis, endocrine systems affecting risk of diabetes–including increased activity of the hypothalamic-pituitary-adrenal (HPA) axis, immune system alterations resulting in increased circulating concentrations of pro-inflammatory cytokines, decreased concentrations of adiponectin relative to fat mass, and increased risk of hypertension (120,121).

 

  • Cocaine and marijuana: Exposure of the fetus to cocaine or marijuana during pregnancy has been reported to increase the likelihood of obesity in childhood and increase risk factors for T2DM (122-124). The mechanisms have yet to be ascertained.

 

Early Feeding Practices

 

For reasons discussed above, accurate assessment of the effects of early infant feeding practices on subsequent adiposity must control for possible effects of maternal adiposity as well as socioeconomic status and other factors that may affect the ability to breastfeed (125). Meta-analyses have shown that predominantly breastfeeding for at least 3-6 months is associated with significant reductions in the prevalence of obesity of their offspring through young adulthood (126-128), even when controlled for other adiposity-risk variables.

 

In addition to any benefits of the dietary macronutrient content on subsequent adiposity, observations suggest that  the institution of a well-balanced diet in childhood may form the basis for long-term healthy dietary habits that will significantly lower adult cardiovascular disease risk even if the diet composition does not substantially affect weight (129).  Studies have also identified positive correlations between the consumption of sugar-sweetened beverages, caloric density of snacks, fast food intake, the portion size of meals, and the hours of television watched (see below) with weight gain in children (129-135).

 

Early Life BMI Trajectories

 

A prospective longitudinal study of 7,738 U.S. children starting kindergarten in 1998-1999 showed that children with overweight and obesity at 5 years of age were four times as likely as their normal-weight counterparts to have obesity at 10-year follow-up. Among children who became obese between the ages of 5 and 14 years, nearly half had been overweight and 75% had been above the 70th percentile for BMI at baseline, indicating that incident obesity for these children had occurred at younger ages (136). Many studies have examined the association of rapid weight, or weight/length or BMI trajectories in the first three years of life and noted association with overweight, obesity or severe obesity at 6 years to adolescence (137-141). One study simulated growth trajectories across life course using pooled data from nearly 42,000 children and adults representative of U.S. population in 2016 adjusting for secular trends. They estimated that a 2-year-old with obesity will continue a trajectory of rapid weight gain and has a 74.9% (95% CI 67.3 to 81.5) probability of being obese at age 35 years. These risks are higher for those with severe obesity.

 

Social Determinants of Health

 

Social determinants of health are the conditions in which people are born, grow, live, work, and age. These include the family, physical and social environment, each of which influences obesity either directly through children’s nutrition and activity or indirectly via added stress. Analysis of NHANES data has demonstrated a higher prevalence of obesity and severe obesity with greater age and lower education of the household head (142). This same study noted an association of severe obesity in youth residing in non-metropolitan statistical areas with more difficult access to large supermarkets. In a study of national sample of 3,748 children from US households receiving Supplemental Nutrition Assistance Program (SNAP) compared to those without, Gorski Findling et al noted higher odds of obesity amongst those receiving the SNAP program (OR 1.14 [95% CI 1.05-1.24]) and with access to a combination grocery/other store, compared to those with access to supermarkets with greater variety of fresh food. They also noted that in convenience stores, 26.1% of the average child’s total household food spending was on sugary beverages (SNAP 29.8% vs non-SNAP 15.5%) (143). In a study of households in New York City, Elbel et al noted that living farther than 0.025 mile (about half a city block) from the nearest fast-food restaurant was associated with lower rates of overweight and obesity, along with lower BMIz scores (144). The built and natural environments play a critical role in the access to physical activity (PA) for children. Street connectivity, defined as the directness of links and density of connection in street networks provides better access to outdoor PA such as walking, playing and cycling. Studies with perceived street connectivity by children, frequently near school, had higher odds of PA (OR 1.13, 95% CI 1.04-1.24). Similarly, higher odds of moderate to vigorous PA (OR 1.33, 95% CI 1.17-1.52) was noted with higher levels of street connectivity. No significant associations were identified with BMI or BMI z-scores (145). The same authors also conducted a metanalysis of natural environment with levels of PA in children. Ambient temperature was identified as the most consequential predictor associated with PA. An increase of 10 °F heating and cooling was associated with reduction in moderate to vigorous PA by 5 and 17 min respectively. No associations were reported with air quality (146).

 

Thus, a complex array of determinants contributes towards the risk of obesity and severe obesity in youth and will require a multipronged approach for intervention. (147-157)

 

Physical Activity, Sedentary Behavior, and Sleep

 

Behaviors related to PA and sedentary established in childhood have been shown to track well into adulthood(150,151) and are independent correlates of BMI and adiposity (158,159) (160,161). Meta-analyses of cross-sectional studies show negative  associations of PA and positive associations of sedentary behavior (SB) with adiposity in children (162-164), that are  further evident with direct objective (e.g., calorimetry) rather than subjective (self-reported) assessments (163). The implications of these findings for early intervention to treat and prevent pediatric obesity are discussed below.

 

MORBIDITIES ASSOCIATED WITH OBESITY IN CHILDREN

 

As in adulthood, obesity in childhood adversely affects every organ system (Table 2). Adiposity-related morbidities, such as hyperlipidemia, track well into adulthood (165) and pediatric obesity may be considered an independent risk factor for adult adiposity-related morbidities, even if the obesity does not persist (166). Certain morbidities, such as slipped capital femoral epiphyses, are the consequence of the biomechanical stresses associated with excess weight while others, especially cardiovascular morbidities, appear to be more closely related to central body fat distribution rather than absolute fat mass. The psychological stress of social stigmatization imposed on children with obesity may be just as damaging to some as the medical morbidities, resulting in significant body dissatisfaction, social anxiety, loneliness, and, especially in girls, somatic symptoms (167,168). These negative images of the obese are so strong that growth failure and pubertal delay have been reported in children due to self-imposed caloric restriction arising from fears of becoming obese (169).

 

Table 2. Pediatric Adiposity-Related Morbidities (165,167-173)

Cardiovascular

Hypertension,  ­ total cholesterol, ­ low density lipoproteins, ¯ high density lipoproteins, metabolic syndrome 

Respiratory

 Abnormal respiratory muscle function and central respiratory regulation, difficulty with ventilation during surgery, lower arterial oxygenation, obstructive sleep apnea, asthma, more frequent and severe upper respiratory infections

Gastrointestinal

Nonalcoholic fatty liver disease, gallstones, gastroesophageal reflux disease

Endocrine

Type 2 diabetes, precocious puberty, polycystic ovarian syndrome, Vitamin D deficiency

Orthopedic

Coxa vara, slipped capital femoral epiphyses, Blount's disease, Legg-Calve-Perthe's disease, degenerative arthritis.

Dermatologic

Intertrigo, furunculosis, acanthosis nigricans (HAIR-AN Syndrome)

Immunologic

Impaired cell-mediated immunity, polymorphonuclear leukocyte killing capacity, lymphocyte generation of migration inhibiting factor, and maturation rates of monocytes into macrophages

Psychologic

Low self-esteem, anxiety, somatization, depression, eating disorders

Lymphatic

Obesity associated lymphedema of the lower legs

Malignancy

Higher lifetime risk of obesity related cancers

 

Pediatric Obesity and Cardiovascular Risk Factors

 

Obesity, hyperlipidemia, hypertension, and other risk factors for cardiovascular disease in children track well into adulthood (23,165,170-173). In long-term follow-up studies, adolescent fatness was a powerful predictor of mortality, cardiovascular disease, colorectal cancer, gout, and arthritis, irrespective of body fatness at the time that the morbidity was diagnosed (166,172). Therefore, it is possible that the metabolic groundwork for the chronic diseases of adulthood is laid down in childhood and the overweight youth must be assessed for both current adiposity-related morbidities and their future risk.

 

Pediatric Obesity and Type 2 Diabetes Mellitus

 

The incidence of youth-onset prediabetes and T2DM is increasing parallel with the rise in obesity in the US (174,175). Between 2001 and 2017, there was a 95.3% (95% CI 77.0-115.4%) relative increase in the prevalence of T2DM in youth < 19 years of age. The greatest absolute increase were observed among non-Hispanic Black and Hispanic youth (174). In the past 2 years of COVID-19 pandemic, the burden of youth onset T2DM has increased dramatically. In a review of two U.S. medical claims databases (~500,000 individuals), persons aged < 18 years with COVID-19 infection were more likely to receive a new diagnosis of diabetes (both Type 1 and Type 2) > 30 days after infection compared to those without or those with pre-pandemic acute respiratory illness (HR = 2,66 [95% CI 1.98-3.56]) (176). The underlying causes for this increase are yet to be identified.

 

Pathologic processes associated with diabetes, including the development of insulin resistance and deterioration of beta-cell function, progress more rapidly in youth-onset T2DM than in adult-onset disease. These factors result in worse glycemic control and an increased risk of early diabetes-related complications (177-179). In the 10-year follow-up of 500 youth with new-onset T2DM enrolled in the Treatment Options for Type 2 Diabetes and Adolescents and Youth (TODAY) clinical trial, the cumulative incidence of hypertension was 67.5%, dyslipidemia 51.6%, diabetic kidney disease 54.8%, nerve disease 32.4% and retinal disease 51.0%. At least one complication occurred in 60.1% of the participants, and at least two in 28.4%. Risk factors for the development of complications included minority race or ethnic group, hyperglycemia, hypertension, and dyslipidemia (180).

 

The pathophysiology of T2DM is discussed in the Endotext Diabetes section (181). Like obesity, T2DM is a complex metabolic disorder(182). In studies of adults and children with a strong family history of T2DM, it appears that impaired pancreatic islet-cell function is the first identifiable metabolic abnormality in some subjects who subsequently develop T2DM, while in other populations, insulin resistance is the first identifiable phenotype (183,184). These data, along with the observation that subjects may be insulin-resistant but not meet clinical definition for diabetes, and that many individuals with impaired β-cell function may not go on to develop T2DM (185,186), suggest that T2DM is due to a combination of insulin-resistance and an impaired β-cell ability to respond to that state of insulin-resistance. In this sense, a state of relative insulin resistance, or the expression of an underlying tendency towards conditions associated with insulin resistance, the major causes of which in adolescence would be pubertal hormonal changes and/or obesity, may act to “unmask” a pre-diabetic state of impaired insulin secretion in some individuals. Consistent with this, available evidence suggests that the incidence of T2DM in children peaks around puberty, as do the ethnic differences in the prevalence of pediatric obesity (187,188), coincidentally with the known decline in insulin-sensitivity and increase in adiposity in the peri-pubertal period (189-191).

 

Central body fat distribution, usually defined on the basis of waist circumference or the ratio of waist-to-hip circumference, is an independent predictor of adiposity-related insulin resistance in adolescents and adults (191-193)as well as other co-morbidity risk factors (194-196). There appear to be effects of ethnicity on the relative impact of body fat distribution on insulin sensitivity. In Caucasian-American children, increasing visceral adiposity is the best correlate of increased fasting insulin levels and insulin secretion during OGTT, and of glucose disposal during hyperinsulinemic-euglycemic clamp studies (191).  In African American (but not Caucasian) pre-pubertal children, intra-abdominal adipose tissue volume was significantly correlated with fasting insulin concentrations and with insulin sensitivity as measured by area under the curve (AUC) during oral glucose tolerance testing (197-199).  Other studies of African-American prepubertal girls have found that elevated fasting insulin concentrations and reduced insulin sensitivity are significantly correlated with greater subcutaneous, but not visceral, adipose tissue volumes (200). Because of the increasing frequency of T2DM among adolescents with obesity, and the worsening of diabetes-related morbidities that may result from delayed diagnosis, the clinician should be alert to the possible of T2DM in all adolescents with generalized and central obesity, and especially those with strong family histories of early-onset (< 40 years of age, one or more parent affected) T2DM (201).

 

ENDOCRINE CHANGES ASSOCIATED WITH OBESITY IN CHILDREN

 

The most common endocrine disorders associated with obesity are secondary to excess body fat and will correct with weight loss (Table 3).

 

Table 3. Endocrine Changes Associated with Obesity in Children (202-206)

Somatotroph

¯ basal and stimulated growth hormone release, normal concentration of insulin-like growth factor-I, accelerated linear growth and bone age

Lactotroph

­ basal serum prolactin but ¯ prolactin release in response to provocative stimuli

Gonadotroph

Early entrance into puberty with normal circulating gonadotropin concentrations may be due to earlier priming of the hypothalamic-pituitary-gonadal axis by estrogens created by aromatization of androgens in adipose tissue and/or by increased circulating concentrations of leptin associated with higher adipose tissue mass.

Thyroid

Normal serum T­4 and reverse T3, normal or ­serum T3, ¯  TSH-stimulated T4 release resulting in  ­ TSH levels

Adrenal

Normal serum cortisol but ­ cortisol production and excretion, early adrenarche, ­ adrenal androgens and DHEA, normal serum catecholamines and 24-hour urinary catecholamine excretion 

Gonad

¯ circulating gonadal androgens due to ¯ sex-hormone binding globulin, dysmenorrhea, dysfunctional uterine bleeding, polycystic ovarian syndrome

 Pancreas

­  fasting plasma insulin, ­ insulin and glucagon release,  ­ resistance to insulin-mediated glucose transport

 

There are, however, several endocrine or genetic syndromes in which obesity is part of a distinct symptom complex that often includes poor statural growth (e.g., hypercortisolism, hypothyroidism) (Table 4) and/or very distinct heritable phenotypes (e.g., Prader Willi; Bardet-Biedl syndromes) (Table 1). Assessment of skeletal maturation by bone age, and physical examination for age-appropriate secondary sexual characteristics as well as syndrome-specific morphology or symptomatology (e.g., hypotension, constipation in hypothyroidism, centripetal distribution of fat in hypercortisolism) can usually rule out these syndromes as causes of obesity.

 

Table 4.  Other Diseases, Injuries, and Medications Associated with Obesity (67,206)

Disease

Structural/Biochemical Lesion

Clinical Features

Acquired hypothalamic lesions

Infectious (sarcoid, tuberculosis, arachnoiditis, encephalitis), vascular malformations, neoplasms, trauma, post-surgical

Adipocyte hypotrophy with little hyperplasia, headache and visual disturbance, hyperphagia, hypodipsia, hypersomnolence, convulsions, central hypogonadism-hypothyroidism-hypoadrenalism, diabetes insipidus, hyperprolactinemia, hyperinsulinism, type IV hyperlipidemia

Cushing’s Disease / Syndrome

Hypercortisolism   

Moon facies, central obesity, ¯ lean body mass, glucose intolerance, short stature

Hypothyroidism

Hypothalamic, pituitary, or thyroidal

Hypometabolic state (constipation, anemia, hypotension, bradycardia, cold intolerance), cretinism (if congenital)

ROHHAD or ROHHADNET syndrome*

Hypothalamic

Hyperphagia, obesity, hypoventilation, adipsic hypernatremia, thermal dysregulation, GH deficiency, hyperprolactinemia,

Medications

Tricyclic antidepressants, Glucocorticoids, Antipsychotic drugs, Antiepileptic drugs, Sulfonylureas

*ROHHAD - rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation; ROHHADNET - rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation with neural crest tumors.

 

WEIGHT STIGMATIZATION (FAT SHAMING)

 

Weight stigmatization (devaluation and denigration of a person because of obesity) includes explicit and implicit weight bias and perpetuates the view that obesity and the difficulty in weight loss are the fault of the individual’s poor diet and exercise choices (207). Weight stigmatization is so common across age, gender, race, and ethnicity that it must be considered as a co-morbidity of overweight and obesity; is prevalent in children and adolescents regardless of their socioeconomic and demographic characteristics. Between 25% and 50% of children have been bullied and/or have been discriminated against based on their weight  (167,168). Weight bias is reported among peers, families, teachers, health professionals, and multiple media outlets (207-209) and has been shown to precipitate unhealthy eating habits, psychosocial stress, and additional weight gain in children (209-211).  

 

PREVENTION AND TREATMENT OF OBESITY:  CLINICAL APPROACH TO THE PEDIATRIC PATIENT

 

Prenatal Care

 

Prevention of obesity in childhood includes early, including prenatal, identification of the child at risk for subsequent obesity and application of effective interventions to reduce that risk. Ideally, this process includes health professions involved in obstetrics, maternal-fetal medicine, and pediatrics.  

 

Pregnancy-related modifiable risk factors for maternal under- and over- nutrition, SGA, LGA, pre- and post-natal rapid weight gain as well as childhood overweight and obesity include higher maternal pregravid adiposity, excessive gestational weight gain, gestational diabetes and hypertension, and smoking during pregnancy (212). Addressing any of these risk factors is beneficial to the health of the mother as well as the fetus. The likelihood of modifying these risk factors is variable. The Institute of Medicine (USA) recommends different ranges of weight gain for women who are underweight (12.5-18.0 kg if BMI < 18.5 kg/m2), have a BMI within the normal range (11.5-16.0 kg if BMI 18.5-24.9 kg/m2), are overweight (7.0-11.5 kg if BMI 25.0-29.9 kg/m2) or are obese (5.0-9.0 kg for BMI >30 kg/m2) (213). As discussed above, there are clear offspring-health benefits of maternal bariatric surgery (104). However, it is difficult to implement non-surgical weight loss plans in preparation for pregnancy and the health benefits of lifestyle interventions both before and during pregnancy on childhood adiposity and co-morbidities smaller and less sustained than observed with bariatric surgery (212,214). Benefits of better control of gestational diabetes are more substantial and persistent (215).

 

Initial Evaluation

 

Pediatric obesity is a persistent worldwide problem, and preventing pediatric obesity and its comorbidities is of paramount importance. The authors posit that every youth with overweight, obesity, and severe obesity should have an opportunity for medical management shared with the individual, the family, and the medical home.  The 2017 Endocrine Society guidelines for pediatric obesity assessment, treatment, and prevention provide an excellent framework towards this goal (24) (Figure 6). A thorough medical and family history is crucial as in any chronic condition.

Figure 6. Algorithm for implementing the 2017 Endocrine Society guidelines on management of children and adolescents with obesity (24).

Initial, and subsequent, evaluations should include a dietary history of the child’s and family’s typical eating habits (including snacks and the frequency with which they consume sugar-added beverages and foods prepared outside of the home). A physical activity history should also be obtained, including school physical education, after-school activities, and activities of daily living (such as walking to school), family activities, and sedentary activities (such as television watching). The family history should encompass obesity, bariatric surgery, T2DM, gestational diabetes, and other comorbidities of obesity including sleep apnea and use of continuous positive airway pressure (CPAP).

 

A detailed physical examination focused on identifying possible causes of unwanted weight gain (e.g., enlarged thyroid gland, cushingoid body habitus) and weight–related co-morbidities (acanthosis nigricans, hypertension, etc. see Tables 2-4) should be performed. Laboratory studies should be guided by history and physical examination and at minimum include fasting measurements of glucose, lipids, hemoglobin A1c, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and vitamin D to screen for diabetes, dyslipidemia, fatty liver disease, and hypovitaminosis D.  Because of the increased risk of polycystic ovarian syndrome (PCOS) in adolescents with obesity, total and free testosterone, as well as sex hormone binding globulin (SHBG), should be measured in girls with signs of hyperandrogenism, oligomenorrhea, or other symptoms suggestive of this disorder (216). Routine evaluation for endocrine etiologies of pediatric obesity are not recommended by the Endocrine Society (24) except if statural growth is compromised. The authors suggest amending this to include a broader list of phenotypes that may be related to thyroid disease. These should include any possible indices of hypothyroidism in the first 3 years of life to avoid the deleterious effects of early thyroid disease on subsequent development (217). They should also include screening for symptoms of acquired hypothyroidism, especially autoimmune thyroiditis and especially in adolescent females, with later onset weight gain or autoimmune disease (218).

 

Children under 5 years of age who are extremely obese, especially if they have concurrent adiposity-related morbidities, evidence of developmental delay, or other phenotypic features associated with the rare obesity syndromes (such as Prader Willi or Bardet Biedl syndrome) discussed above (Table 1), can be referred to a physician who specializes in the treatment and genetic evaluation of pediatric obesity (219). Targeted therapies are available for some of these conditions.

 

Health care providers are often confronted by the difficulty in deciding whether or not to attempt intervention in a child who is not obese but is overweight or has a growth trajectory that may be premonitory of obesity. Assessment of the risk of progression to obesity should be based on family history of growth patterns and of adiposity-related co-morbidities, any evidence of co-morbidity in the child, and familial readiness to engage in early intervention to prevent obesity. Parents of a child who is clearly “at-risk” maybe more reluctant to begin lifestyle or other therapies since the child is not obese. If parents are amenable then the same therapies used to treat obesity can be initiated. If the parents are not amenable then the health care provider should monitor growth of the child and co-morbidity risk and keep parents involved in the discussion of progression towards obesity and/or its co-morbidities.

 

Family based interventions are most effective for management of pediatric obesity (219-221).  Efforts spent in assessing the home environment are critical to success of management. Factors such as parental and sibling adiposity, education, and the quality of the relationship between the primary caregiver and the child have all been identified as significant determinants of the likelihood of a successful pediatric weight loss intervention (222). The logical extension of these findings is that optimal therapeutic interventions must include support for the child’s family, regardless of the level of obesity of the family members (223).

 

The clinician should begin assessment of family therapeutic readiness by asking the entire family how concerned they are about the patient’s overweight, in a supportive manner designed to elicit cooperation from the family and patient.  Examples might include asking, “Do you feel that weight is a problem?” or “What do you think that you could change to help you lose weight?” rather than, “Why can’t you control what you eat?”  The discussion should emphasize the potential benefits of therapeutic intervention, including the importance of cooperation of all caregivers, the increased likelihood of diminishing adult body fatness with early adoption of consistent and long-term lifestyle intervention.

 

Treatment of the child with overweight or obesity must be individualized and the clinician should remain sensitive to issues such as ability of the parents to prepare meals for the patient, neighborhood safety or availability of adult supervision, which may impact on the availability of physical activity after school, and remain culturally sensitive in making dietary recommendations. 

 

Therapeutic Intervention

 

The approach to management of a child with overweight or obesity is in many ways more complex than the same choice in an adult because of additional concerns regarding growth if negative energy balance is excessive.  The major goal of management should be to diminish morbidity rather than to achieve a "cosmetically endorsed" body weight. While imperfect, BMI is clinically the most readily accessible parameter to assess the level of obesity. The “severity” of obesity should initially be assessed based on the BMI references provided above, presence of current morbidities such as T2DM, and risk of future adiposity-related morbidity (based on family history) (219). This increased risk of treatment-associated impairment of statural or brain growth is higher in younger children and caloric restriction to reduce weight should not be used in infants less than 2 years of age. Beginning therapy with the assumption that obesity is a choice and can be “fixed” easily by moving more and eating less is outdated and inaccurate in the current science of obesity and promotes weight stigmatization and “fat shaming”. Excessive emphasis on behavior and self-sufficiency may precipitate eating disorders, as well as other psychological disorders such as low self-esteem, anxiety, and depression – especially if long-term weight loss is unsuccessful, especially in the peri-pubertal stages (167). It is important to tailor the management for individual child and their family. Program adherence, defined as the number of contacts with the weight-management program, is a primary factor in successful weight loss for overweight children and adolescents (224). Maximizing adherence is like to include program modification over time in a given child. As more data accumulate regarding precision medicine approaches to identify genetic and other predictors of responses to different interventions, adherence and success are likely to improve. Clinicians can prescribe intensive, age-appropriate, culturally sensitive, family-centered lifestyle modifications (dietary, physical activity, behavioral) to promote a decrease in BMI (rather than weight). When weight is maintained at a constant level or weight gain is proportionally slower than height gain, BMI can reduce with increase in linear growth. In the otherwise healthy child with obesity with no evidence of co- morbidity, such modifications may be sufficient to maintain long-term health. In contrast, in a youth with severe obesity (BMI ≥ 120% of 95th percentile of BMI) or presence of co-morbidity such as T2DM or hypertension, such management can be augmented with pharmacotherapy and/or bariatric surgery, as deemed suitable (225,226).

 

DIETARY RECOMMENDATIONS

 

The Dietary Guidelines for Americans, 2020-2025 can be used as a reference for dietary counseling (227). These guidelines emphasize following a healthy dietary pattern at every life stage with a focus on meeting food group needs with nutrient (and not calorie)-dense foods and beverages and stay within the appropriate calorie limits. Hence, the entire family can be engaged in culturally appropriate dietary modifications. Studies suggest that the long-term sustenance of such intervention is most successful with a supportive family. It is also important to convey to the family on the need for sustenance of such changes for long-term favorable outcomes. Encouragement can be provided by examining growth and growth velocity curves with patients and their families to illustrate progress.  If appropriate, the significance of any evident reduction in morbidity (e.g., lowering of blood pressure or cholesterol) can be reinforced. Reasonable goals in the form of a "target" body weight at the next visit should be set at each office visit so that the patient and parents are aware of what is expected.  These goals should be modest and attainable even if patients are only moderately compliant with their diet and exercise regimens since achievement of an interval “target weight" will also encourage the patient.

 

The caloric need of a person varies maintain, gain, or lose weight are dependent upon age, sex, height, weight and level of physical activity. The Dietary Guidelines provide estimated amounts of calories needed to maintain energy balance of various age and sex groups at three different levels of physical activity from toddlers to age 2 years, as well as ages 2 and older (227). These estimates are based on the Estimated Energy Requirements (EER) equations, using average reference height and weight by age and sex. These are a useful starting point to tailor the needs to that of the patient. It is useful to get an assessment of the current caloric intake from the families. However, self-reported caloric intake is often inaccurate. For direct assessment, the child's ad libitum diet can be observed and recorded by the parents for a minimum of five consecutive days. A diet diminished to 300 to 400 kcal/day below weight-maintenance requirements as assessed by dietary history or as calculated based upon formula relating anthropometry to energy expenditure, e.g., the Harris-Benedict Equation (228) should result in weight loss of approximately one pound per week. Note that since  weight reduction per se causes decreased energy expenditure (both from decreased metabolic mass and whatever hypometabolic state is invoked by losing weight (40,49,67) and during weight loss, periodic downward adjustments of energy intake will be necessary to sustain ongoing weight reduction. The family should be instructed in long-term monitoring of caloric intake within, and outside of, the home and cautioned not to become overly critical or punitive towards the child if weight loss is slow or compliance is suboptimal.

 

The core elements of Dietary Guidelines for Americans sorted by food group are listed below:

  • Vegetables: Increased relative consumption of vegetables of all types – dark green; red and orange; beans, peas, and lentils; starchy; and other vegetables.
  • Fruits: Consumption of whole fruits rather than juices.
  • Grains: At least half of the consumed grains should be whole grains.
  • Dairy: Dairy intake should be focused on fat-free or low-fat milk, yogurt, and cheese, and/or lactose-free versions and fortified soy beverages and yoghurt as alternatives.
  • Protein: Protein intake should focus on lean meats, poultry, and eggs; seafood; beans, peas, and lentils; and nuts, seeds, and soy products.
  • Fats: Children need fats – both saturated fats and cholesterol for normal growth and brain development. On the other hand, trans fats, such as those from fried foods are unhealthy. Eggs, butter, whole dairy products and oils, including vegetable oils and those in seafood and nuts are recommended.

 

The guidelines also recommend limiting foods and beverages higher in added sugars (including those with high fructose corn syrup), saturated fat, and sodium. Less than 10% of calories per day should be derived from added sugars starting at age 2 years, and families should be advised to avoid beverages with any added sugars. In the US, 57-61% children derive > 10% of their energy from added sugars, 88% consume > 10% saturated fat and nearly 95% consume foods containing greater than the recommended sodium amount.(229-234) Simply reducing the consumption of these types of foods should in and of itself result in a net negative energy balance most likely by reducing hedonic “eating in the absence of hunger” (235,236) and other aspects of energy intake which have been found to be correlated with subsequent weight gain in children .

 

Ultra-processed (UPF) are defined as “Industrial formulations typically with 5 or more and usually many ingredients. Besides salt, sugar, oils, and fats, ingredients of UPF include food substances not commonly used in culinary preparations, such as hydrolyzed protein, modified starches, and hydrogenated or intensified oils, and additives who purpose is to imitate sensorial qualities of unprocessed or minimally processed foods and their culinary preparations or to disguise undesirable qualities of the final product, such as colorants, flavorings, non-sugar sweeteners, emulsifiers, humectants, sequestrants, bulking, de-foaming, anticaking, and glazing agents” (237,238). These “ready to eat” or “ready to heat” preparations are typically high in added sugar, trans-fat, sodium, and refined starch and low in fiber, protein, vitamins, and minerals. The consumption of UPF has increased by 20-50% per decade 2000-2015 in the USA, and to an even greater degree in low- and middle- income countries (239). Diets high in UPF is associated with adverse health outcomes including obesity, hypertension, dyslipidemia, diabetes and pre-diabetes in adults (240-243,244 ) and, more recently, in children (7); (245,246). Though the Dietary Guidelines for Americans has yet to issue recommendations regarding UPF consumption, we believe that the evidence that UPF’s promote obesity and many of its co-morbidities in children is more than sufficiently compelling to recommend avoiding them.

 

A helpful brochure to recommend healthy eating for children including Nutrition conversation starters can be obtained from https://www.dietaryguidelines.gov/professional-resources.

 

The composition of the diet should contain at least the minimal recommend amounts of protein, essential fatty acids, vitamins, and minerals. The 2017 consensus from the Endocrine Society (24) recommended the following basic principles of dietary intervention to achieve negative energy balance, which it should be noted would likely be beneficial to everyone regardless of adiposity:

 

  • Replace all sugary drinks (including juices, sodas, and whole milk) with water, noncaloric beverages, and low-fat or skim milk.
  • Create a balanced diet including vegetables, fruits, whole grains, nuts, fiber, lean meat, fish, and low-fat dairy products. Specifically encourage consumption of at least five servings of fruits and vegetables daily.
  • Reduce intake of calorie dense foods such as saturated fats, salty snacks, and high glycemic foods including candy, white bread, processed white rice, pasta, and potatoes.
  • Minimize consumption of foods outside of the home. Fast foods in particular.
  • Eat breakfast daily.

 

Based on available data it appears that dietary macronutrient composition in childhood does not significantly affect later adiposity (247) and that diets consisting of drastically altered proportions of nutrients may be dangerous and yield no better results than a limited intake of a nutritionally balanced diet (248,249). It should be noted that the results of these studies vary substantially and may be age-dependent. For example, in a retrospective study Davis et al (250)reported that synergistic effects between the duration of breastfeeding and low sugar-sweetened beverage intake in reducing the odds of obesity in toddlers who were Hispanic. In contrast, a recent study comparing the effects of the low fat versus low glycemic index diet in the treatment of obesity in a population of Hispanic American adolescents found no differences between groups based on dietary macronutrient composition (251) and a recent meta-analysis by Hall and Guo (252) found that low fat diets promoted greater fat loss than low carbohydrate diets in adults.

 

As noted above, nutritional counseling should encourage decreasing the use of calorically dense (high fat or high glycemic index) foods and adding more fruits and vegetables to the daily diet.  The substitution of  water  for non-nutritious high calorie sugar containing drinks (juices, iced teas and soda pop) may be very helpful (225), at least transiently (253){Ebbeling, 2012 #10778}.  In some cases, reductions in calorically dense foods and sugar-containing drinks through substitution and/or elimination alone can decrease calories and weight without changing the general pattern of food consumption in the family.  When families eat at restaurants and fast-food vendors, they have less control over food choices than they do at home.  Thus, reduction in the number of meals prepared outside the home may also be an effective weight-loss strategy. Parents and adult caregivers should understand the important role they play in the development of proper eating habits in their young children.  The parents’ food preferences, the quantities and variety of foods in the home, the parents’ eating behavior and physical activity patterns all determine how supportive the home environment is to the child with obesity.

 

THERAPEUTIC EXERCISE

Physical activity may promote a slightly increased muscle mass, thereby raising total metabolic rate, and the putative effects of exercise to reduce visceral adipose tissue mass independently lower the risk of hyperlipidemia and diabetes mellitus (254-256). However, the energy cost of even vigorous exercise is low when compared to the caloric content of many "fast foods" or other "snacks", and exercise should not be viewed as a "license to eat".  For example, walking at three miles per hour for one hour consumes about 200 kilocalories, about the same number of calories contained in a 1¾ ounce bag of potato chips. Use of "treats", such as ice cream, potato chips, etc., as incentives to exercise negates its impact. As with all interventions to reduce pediatric adiposity, increasing physical activity and decreasing sedentary behavior is most likely to be effective, sustained, and benefit the entire family if the entire family participates.

 

Combining the 2017 Endocrine Society statement on pediatric obesity (24) with other recommendations for physical activity in children (147), the following guidelines are suggested which again could be applied to the entire family, regardless of their adiposity:

 

  • Exercise should be fun, age-appropriate, and tailored to the child’s fitness level and ability and should involve large muscle groups (e.g., quadriceps) to increase energy expenditure. Exercise frequency, duration, and intensity should increase over time.
  • Moderate-to-vigorous physical activity should, on the average, encompass 90-120 minutes of the day in preschoolers and toddlers (usually unstructured physical activity) and at least 1 hour of the day in children 6 years or older (usually structured physical activity such as after school sports).
  • Improve sleep hygiene (10-13 hours per night for preschoolers and 8-10 hours per night for adolescents) in response to numerous studies demonstrating associations of decreased sleep duration and weight gain (257-259).
  • In order to address the issue of increased sedentary behavior due to screen time, the American Academy of Pediatrics provides a downloadable Family Media Plan in English and Spanish (healthychildren.org/MediaUsePlan) (260). This plan is for all children and can be personalized for every family depending on the children’s age(s), family priorities, time of the year (e.g., academic year versus vacation), etc., and includes elements such as screen free zones, screen free times, choosing good content, using medial together and digital privacy and safety. In its 2017 recommendations specifically for children with obesity, the Endocrine Society suggested that nonacademic screen time should be reduced to 1-2 hours per day and that other sedentary behaviors, such as digital activities, should be decreased (24).

 

While no specific aspect of the sedentary lifestyle has been shown to directly cause obesity, behaviors such as television viewing, reading, working at a computer, driving a car or commuting do exert effects on health. Television viewing appears to be directly associated with the incidence of obesity, and inversely associated with the remission of obesity. The impact of television viewing on obesity seems to be due to both displacing more vigorous activities and its effect on diet. Not only is television viewing a sedentary behavior, but food has also constituted the most heavily advertised product on children’s television in the United States. In Mexican-American children, adiposity was significantly correlated with time spent watching television but not with time spent watching videos (261), suggesting that the bulk of the positive association of television watching and adiposity is due to the approximately 60% of advertising that is devoted to food (134).  Children and adolescents should be encouraged to view as little television as possible. Limitation of television, video games, and internet viewing will encourage greater participation in physical activity. Clinicians should encourage children to participate in organized or individual sports (participate, not watch from the bench) and advocate for better community- and school-based- activity programs.

 

If the patient is unable to lose weight and/or co-morbid conditions persist, consideration should be given to referral of the child to a physician specializing in the treatment of pediatric obesity. Weight-loss programs, weight-reduction camps, etc. are often not covered by medical insurance and should be considered for the child who is morbidly obese with some caution.  Enrollment in a highly supervised environment may demonstrate to an overweight child that weight loss is possible and encourage them to continue. However, rapid weight loss may precipitate cholelithiasis (262) or eating disorders.  A child may become overly pre-occupied with his/her weight and, even if a moderate degree of weight-loss is achieved, lose self-esteem. Obsession with weight on the part of the child or their family may lead to serious deterioration of intra-family relationships.

 

DIGITAL INTERVENTIONS

 

Technology based interventions provide a novel tool to add to the armamentarium for weight management in youth. Technologies can include information and communication technology, web-based interventions, mobile phone applications and smart-phone based interventions, text-messaging, and wearable technology. In a systematic review of 8 studies (n=582 youth) of technology-based interventions with or without wearable devices with a spread of intervention ranging from behavioral counseling via telehealth to text-message based reminders and family-based therapies, significant differences in BMI were reported by 5 of the 8 studies. Pooled analysis showed standardized mean difference of -0.61 (95% CI -1.10, -0.13, p <.01), albeit with significant heterogeneity. Interestingly, as is seen with in-person interventions, the effect was lower in the sub-group with parental involvement (263). Similarly, in a separate meta-analysis of 12 randomized controlled trials (3227 youth), use of wearable devices, such as pedometers or wristband activity trackers, had statistically significant reduction in BMI, BMI z-score and body fat, but not in waist circumference. The impact was higher in individuals with obesity compared to those with normal weight (for prevention of obesity) (264). Where accessible, such technologies can provide an additional tool for weight management in youth.

 

PHARMACOLOGICAL AND SURGICAL INTERVENTIONS

For youth with severe obesity or those with concomitant co-morbidity, both pharmacotherapy and surgical interventions can augment intensive lifestyle management prescribed above. Several pharmacological therapies have been approved by FDA for use in youth ≥ 12 years of age in the past 5 years and clinical trials with additional medications are ongoing at the time of this publication. Professional associations such as The Obesity Society, Pediatric Endocrine Society as well as other experts have provided guidelines for clinical considerations on the use of obesity pharmacotherapy(265-267). Figure 7 provides a mechanistic overview of pharmacotherapies

Figure 7. Mechanism of action of the available medications. Many of the currently used medications for obesity impact the centers for weight regulation in the brain including hypothalamus and the prefrontal cortex, as well as other organs. Abbreviations: NorEpi: norepinephrine; POMC: pro-opiomelanocortin; CART: cocaine- and amphetamine regulated transcript; AGRP: agouti-related polypetide; NPY: neuropeptide Y; GLP1R: glucagon like polypeptide receptor 1; LEPR: leptin receptor; GABA: gamma amino butyric acid; MC4R: melanocyte 4 receptor. Bupropion and Naltrexone are not approved for use in pediatrics for weight loss. The therapeutic preparation of leptin is called metreleptin. Figure created using biorender.com

Both the indications for pharmacotherapy and the available approved pharmacological interventions are different in children than in adults. General recommendations for use of pharmacotherapy include: a) availability of a multidisciplinary team including at least one pediatric specialist; b) severe obesity (BMI ≥ 120% of 95th percentile or BMI ≥ 35 kg/m2) or presence of a co-morbidity with BMI ≥ 95th percentile (or BMI ≥ 30 kg/m2); c) concomitant lifestyle intervention; d) continuation of medication(s) if there is ≥ 5% BMI reduction from baseline at 12 weeks on the optimal dose or arrest or slowing of weight gain; e) discontinuation if not tolerated or if dangerous side effects occur or persist despite dose adjustment (265). A list of available therapies and evidence for pediatric use are listed below with guidance on administration is provided in figure 8.

 

Figure 8. Pharmacotherapy for youth with obesity, approval status and available pediatric data.

Bariatric surgery is only approved in adolescents and, although the frequency of adolescent bariatric surgery is increasing, it still accounts for only about 1% of total U.S. bariatric surgery cases (268). Outcome studies of adolescent bariatric surgery have shown significant improvements in weight, cardiometabolic co-morbidity risk, and quality of life tempered a high incidence (57%) of hypoferritinemia and need for additional abdominal procedures (13%) (269). The American Society for Metabolic and Bariatric Surgery recommends the following selection criteria for adolescents eligible for bariatric surgery:

 

  • Body mass index ≥ 35 kg/m2 and a severe comorbidity, with significant comorbidity with short-term effects on health or BMI 40 kg/m2 or above with more minor comorbidities.
  • Physical maturity, defined as completing 95% of predicted adult stature based on bone age or reaching Tanner stage IV. This criterion is based on theoretical concerns that rapid weight loss might inhibit statural growth if an adolescent has not reached near adult height.
  • History of lifestyle efforts to lose weight through changes in diet and physical activity.
  • Ability and motivation of the patient and family to adhere to recommended treatments pre- and postoperatively, including vitamin and mineral supplementation.
  • Appropriate understanding of the risks and benefits of surgery on behalf of the adolescents
  • Supportive but not coercive family.

 

CONTRINDICATIONS TO BARIATRIC SURGERY INCLUDE:

 

  • Medically correctable cause of obesity
  • An ongoing substance abuse problem (within the preceding year).
  • A medical, psychiatric, psychosocial, or cognitive condition that prevents adherence to postoperative dietary and medication regimens or impairs decisional capacity.
  • Current or planned pregnancy within 12 to 18 months of the procedure.
  • Inability on the part of the patient or parent to comprehend the risks and benefits of the surgical procedure.

 

Both the American Society for Metabolic and Bariatric Surgery and the Endocrine Society have recommended that a multidisciplinary team consisting of a bariatric surgeon, a pediatrician specializing in obesity, a nutritionist, a mental health professional, an exercise physiologist, and a health care coordinator should be established to evaluate optimal therapy for a child who is a candidate for bariatric surgery based on the presence of co-morbidities and failure of other interventions.

 

ADDRESSING WEIGHT STIGMATIZATION

 

Health care providers have an opportunity to improve the quality of life and intervention outcomes for children with obesity by addressing weight bias (209). Recent specific recommendations include:

 

  • Avoid oversimplification: Recognize the multifactorial nature of obesity as a disease that may require long-term, or even lifelong, attention and challenge stereotypes that obesity, or the difficulty in losing weight, is a lifestyle choice rather than a biological issue.
  • Avoid weight bias: When speaking with the patient or their family focus on the chief complaint (even if it is not weight-related), feel free to discuss implicit and explicit weight bias with families, and support evidence-based care including medication or surgery.
  • Encourage a collaborative relationship: Ask if it is okay with the patient and family to discuss weight during an appointment, use person-first language (“having obesity” rather than “is obese”), acquaint them with the multifactorial complex nature of weight management, and explore alternative factors that contribute to higher BMI.

 

OTHER INTERVENTIONS

 

There are new types of intervention that are only recently being vetted in pediatric randomized clinical trials.  Prebiotics, probiotics, and other manipulations of the gut microbiome have been suggested as possible means of treating or preventing pediatric obesity with some initial promising results in relatively small studies (270-272).   There is a wide variability in the efficacy of school-based interventions but with more attention to the methodological differences between those that are more successful and those that are not, it may be possible to create a cost-effective practical means of addressing the burgeoning problem of pediatric obesity (273).

 

There are also a number of bills languishing in Washington that have been left in committee and not allowed to be aired for public debate. The Sugar-sweetened beverage excise task (SWEET) act,  the Stop Subsidizing Childhood Obesity Act, and  establishment of nutrition standards for all foods served and sold in schools have all been projected to return between 4 and 35 times the number of dollars invested in health care cost savings over the next 10 years (274). The failure of the SWEET Act, and other legislation that might affect childhood obesity rates, to get into open debate suggests that health care professionals dealing with the problem of pediatric obesity could be more vocal regardless of whether they support the legislation. Implementation of the improved school meals endorsed by the Healthy, Hunger-free Kids Act has been shown to result in significant improvement in school-meals and to be increasingly acceptable to students, with improvement in participation in school-based breakfast programs since its implementation (275,276). Any efforts to remove funding from the Healthy, Hunger-free, Kids Act (277) or the Supplemental Nutrition Assistance Program (SNAP), in particular SNAP-Ed, will potentially promote poor dietary habits and food insecurity (278-280) and should provoke a similar level of discussion by health professionals in public forums. These are important issues and commentary from those most familiar with the problem should be helpful in their evaluation.

 

REFERENCES

 

  1. Biener A, Cawley J, Meyerhofer C. The high and rising costs of obesity to the US health care system. J Gen Inter Med. 2017;32:6-8.
  2. Cawley J, Biener A, Meyerhoefer C, Ding Y, Zvenyach T, Smolarz B, Ramasamy A. Direct medical costs of obesity in the United States and the most populous states. J Manag Care Spec Pharm. 2021;27:354-366.
  3. Rosenbaum M. Epidemiology of pediatric obesity. Ped Annals. 2007;36:89-95.
  4. Khera AV, Chaffin M, Wade KH, Zahid S, Brancale J, Xia R, Distefano M, Senol-Cosar O, Haas ME, Bick A, Aragam KG, Lander ES, Smith GD, Mason-Suares H, Fornage M, Lebo M, Timpson NJ, Kaplan LM, Kathiresan S. Polygenic Prediction of Weight and Obesity Trajectories from Birth to Adulthood. Cell. 2019;177(3):587-596.e589.
  5. Perng W, Oken E, Dabelea D. Developmental overnutrition and obesity and type 2 diabetes in offspring. Diabetologia. 2019;62:1779-1788.
  6. CDC National Center for Chronic Disease Prevention and Health Promotion (NCCDPHP). Chronic diseases in America. https://wwwcdcgov/chronicdisease/resources/infographic/chronic-diseaseshtm. 2021.
  7. Wang L, Steele E, Du M, Pomeranz J, O'Connor L, Herrick K, Luo H, Zhabg X, Mozaffarian D, Zhang F. Trends in consumption of ultraprocessed foods among us youths aged 2-19 years, 1999-2018. JAMA. 2021;326:529-530.
  8. Fryar C, Carroll M, Afful J. Prevalence of overweight, obesity, and severe obesity among children and adolescents aged 2–19 years: United States, 1963–1965 through 2017–2018. 2020 https://www.cdc.gov/nchs/data/hestat/obesity-child-17-18/obesity-child.htm2020.
  9. Hu K, Staiano A. Trends in obesity prevalence among children and adolescents aged 2 to 19 years in the US from 2011 to 2020. JAMA Pediatr. 2022:JAMA Pediatr. Published online July 25, 2022. doi:2010.1001/jamapediatrics.2022.2052.
  10. Rundle AG, Factor-Litvak P, Suglia SF, Susser ES, Kezios KL, Lovasi GS, Cirillo PM, Cohn BA, Link BG. Tracking of obesity in childhood into adulthood: Effects on body mass index and fat mass index at age 50. Child Obes. 2020;16:226-233.
  11. Ogden C, Fryar C, Martin C, Freedman D, Carooll M, Gu Q, Hales C. Trends in obesity prevalence by race and hispanic origin-1999-2000 to 2017-2018. JAMA. 2020;324:1208-1210.
  12. Ogden C, Martin C, Freedman D, Hales C. Trends in obesity disparities during childhood. Pediatrics. 2022;doi: 10.1542/peds.2022-056547.
  13. Lange S, Komaniyets L, Freedman D, Draus E, Porter R, Balnck H, Goodman A. Longitudinal trends in body mass index before and during the covid-19 pandemic among persons aged 2-19 years - United States, 2018-2020. MMWR. 2021;70:1278-1283.
  14. Phelan S, Wing R. Prevalance of successful weight loss. Arch Int Med. 2005;165:2430.
  15. Wadden T, Neiberg R, Wing R, Clark J, Delahanty L, Hill J, Krakoff J, Otto A, Ryan D, Vitolins M, Look AHEAD Research Group. Four-year weight losses in the Look AHEAD study: factors associated with long-term success. Obesity. 2011;19:1987-1998.
  16. Kraschnewski J, Boan J, Esposito J, Sherwood N, Lehman E, Kephart D, Sciamanna C. Long-term weight loss maintenance in the United States. Int J Obes. 2010;34:1644-1654.
  17. Normo M, Danielsen Y, Nordmo M. The challenge of keeping it off, a descriptive systematic review of high-quality, follow-up studies of obesity treatments. Obes Rev. 2020;21:e12949.
  18. Reinehr T, Widhalm K, l'Allemand D, Wiegand S, Wabitsch M, Holl R, Obesity TA-WSGaGCN. Two-year follow-up in 21,784 overweight children and adolescents with lifestyle intervention. Obesity. 2009;17:1196-1199.
  19. Bondyra-Wisnewska B, Muszkowaska-Ryciak J, Harton A. Impact of Lifestyle Intervention Programs for Children and Adolescents with Overweight or Obesity on Body Weight and Selected Cardiometabolic Factors—A Systematic Review. Int J Environ Res Public Health. 2021;18:2061.
  20. Mead E, Brown T, Rees K, Azevedo L, Whittaker V, Jones D, Olajide J, Mainardi G, Carpeleijn E, O'Malley C, Beardsmore E, Al-Khudairy L, Baur L, Metzenfort M-I, Demaio A, Ells L. Diet, physical activity and behavioural interventions for the treatment of overweight or obese children from the age of 6 to 11 years. Cochrane Database Syst Rev. 2017;6:CD012651. doi: 012610.011002/14651858.CD14012651.
  21. Whitaker R, Pepe M, Wright J, Seidel K, Dietz W. Early adiposity rebound and the the risk of adult obesity. Pediatr. 1998;101:E5.
  22. Freedman D, Kettel Kahn L, Serdula M, Srinivasan S, Berenson G. BMI rebound, childhood height and obesity among adults: the Bogalusa Heart Study. Int J Obes. 2001;25:543-549.
  23. Whitaker R, Wright J, Pepe M, Seidel K, Dietz W. Predicting obesity in young adulthood from childhood and parental obesity. N Eng J Med. 1997;337:869-873.
  24. Styne D, Arslanian S, Connor E, Farooqi I, Murad M, Silverstein J. Pediatric obesity-assessment, treatment, and prevention: An endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2017;102:709-757.
  25. Pietrobelli A, Faith M, Allison D, Gallagher D, Chiumello G, Heymsfield S. Body mass index as a measure of adiposity among children and adolescents: a validation study. J Pediatr. 1998;132:204-210.
  26. Reilly J, Dorosty A, Emmet P, Study TA. Identification of the obese child: adequacy of the body mass index for clinical practice and epidemiology. Int J Obes. 2000;24:1623-1627.
  27. Troiano R, Flegal K. Overweight children and adolescents: description, epidemiology, and demographics. Pediatr. 1998;101(suppl):497-504.
  28. SE Barlow and the Expert Committee. Expert committee recommendations regarding the prevention, assessment, and treatment of child and adolescent overweight and obesity: Summary report. Pediatr. 2007;120 (Suppl. 4):S164-192.
  29. Flegal K, Wei R, Ogden C, Freedman D, Johnson C, Curtin L. Characterizing extreme values of body mass index-for-age by using the 2000 Centers for Disease Control and Prevention growth charts. Am J Clin Nutr.2009;90:1314-1320.
  30. Gulati A, Kaplan D, Daniels S. Clinical tracking of severely obese children: A new growth chart. Pediatrics.2012;130:1136-1140.
  31. Kelly A, Barlow S, Rao G, Inge T, Hayman L, Steinberger J, EM U, Ewing L, Daniels S, American Heart Association Atherosclerosis H, and Obesity in the Young Committee of the Council on Cardiovascular Disease in the Young, Council on Nutrition, Physical Activity and Metabolism, and Council on Clinical Cardiology,. Severe obesity in children and adolescents: identification, associated health risks, and treatment approaches: a scientific statement from the American Heart Association. Circulation. 2013;128:1689-1712.
  32. Skinner A, Ravanbakht S, Skelton J, Perrin E, Armstrong S. Prevalence of obesity and severe obesity in US children, 1999-2016. Pediatrics. 2018;141:e20173459. doi: 20173410.20171542/peds.20172017-20173459.
  33. Skinner A, Perrin E, Skelton J. Prevalence of obesity and severe obesity in US children, 1999-2014. Obesity.2016;24:1116-1123.
  34. Skinner A, Skelton J. Prevalence and trends in obesity and severe obesity among children in the United States, 1999-2012. JAMA Pediatr. 2014;168:561-566.
  35. Organization WH. Growth Charts. https://wwwcdcgov/growthcharts/data/who/GrChrt_Boys_24HdCirc-L4W_rev90910pdf. 2006.
  36. Chen L, Tint M, Fortier M, Aris I, Shek L, Tan K, Rajadurai V, GLuckman P, Chong Y, Godfrey K, Kramer M, Henry C, Yap F, Lee Y. Body composition measurement in young children using quantitative magnetic resonance: a comparison with air displacement plethysmography. Pediatr Obes. 2017;Epub Ahead of Print.
  37. Rosenbaum M, Agurs-Collins T, Bray M, Hall K, Hopkins M, Laughlin M, MacLean P, Maruvada P, Savage C, Small D, Stoekel L. The Accumulating Data to Optimally Predict Obesity Treatment (ADOPT): Recommendations from the biological domain. Obesity. 2018;26.
  38. Simmonds M, Llesellyn A, Owen C, Woolacott N. Predicting adult obesity from childhood obesity: a systematic review and meta-analysis. Obes Rev. 2016;17:95-107.
  39. Llewellyn A, Simmonds M, Owen C, Woolacott N. Childhood obesity as a predictor of morbidity in adulthood: a systematic review and meta-analysis. Obes Rev. 2016;17:56-67.
  40. Rosenbaum M, Leibel R, Hirsch J. Medical Progress: Obesity. N Engl J Med. 1997;337:396-407.
  41. Dhurandhar J, Kaiser K, Dawson J, Alcorn A, Keating K, Allison D. Predicting adult weight change in the real world: a systematic review and meta-analysis accounting for compensatory changes in energy intake or expenditure. Int J Obes. 2015;39:1181-1187.
  42. Wadden T. Treatment of obesity by moderate and severe caloric restriction. Ann Intern Med. 1993;119:688-693.
  43. Knip M, Nuutinen O. Long-term weight control in obese children: persistence of treatment outcome and metabolic changes. Int J Obes. 1992;16:279-287.
  44. McGuire W, Wing R, Hill J. The prevalence of weight loss maintenance among American adults. Int J Obes.1999;23:1314-1319.
  45. Klem M, Wing R, Lang W, McGuire M, Hill J. Does weight loss maintenance become easier over time. Obes Res. 2000;8:438-444.
  46. Klem M, Wing R, McGuire M, Seagle H, Hill J. A descriptive study of individuals successful at long term maintenance of substantial weight loss. Am J Clin Nutr. 1998;66:239-246.
  47. Wing R, Hill J. Successful weight loss maintenance. Annu Rev Nutr. 2001;21:323-341.
  48. Belanger B, Cupples L, D'Agostino R. The Framingham study: An epidemiologic study investigation of cardiovascular disease. Section 36: Measures at each examination and interexamination consistency of specified characteristics.Framingham publication No. 88-2970. 1988.
  49. Rosenbaum M, Leibel R. The physiology of body weight regulation:relevance to the etiology of obesity in children. Pediatr. 1998;101:525-538.
  50. Rosenbaum M, Hirsch J, Murphy E, Leibel R. The effects of changes in body weight on carbohydrate metabolism, catecholamine excretion, and thyroid function. Amer J Clin Nutr. 2000;71:1421-1432.
  51. Wardlaw S. Clinical review 127: Obesity as a neuroendocrine disease: lessons to be learned from proopiomelanocortin and melanocortin receptor mutations in mice and men. J Clin Endocrinol Metab.2001;86:1442-1446.
  52. Rosenbaum M, Leibel R. Leptin: a molecule integrating somatic energy stores, energy expenditure, and fertility. Trends Endorcinol Metab. 1998;9:117-123.
  53. Ahima R, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, Flier J. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382:250-252.
  54. Ahima R, Kelly J, Elmquist J, Flier J. Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinol. 1999;140:4923-4931.
  55. Rosenbaum M, Leibel R. 20 years of leptin: role of leptin in energy homeostasis in humans. J Endocrinol.2014;223:T83-96.
  56. Rosenbaum M, Leibel R. Chapter 7: Adaptive response to weight loss. In: Kushner R, Bessesen D, eds. Treatment of the obese patient. Second ed. New York, NY: Springer-Verlag; 2014.
  57. Knuth N, Johannsen D, Tamboli R, Marks-Shulman P, Huizenga R, Chen K, Abumrad N, Ravussin E, Hall K. Metabolic adaptation following massive weight loss is related to the degree of energy imbalance and changes in circulating leptin. Obesity. 2014;22:2563-2569.
  58. Sumithran P, Prendergast L, Delbridge E, Purcell K, Shulkes A, Kriketos A, Proietto J. Long-term persistance of hormonal adaptations to weight loss. N Eng J Med. 2011;365:1597-1604.
  59. Maclean P, Blundell J, Mennella J, Batterham R. Biological control of appetite: A daunting complexity. Obesity.2017;25, Suppl 1:S8-16.
  60. Kiortsis D, Duraced I, Turpin G. Effects of a low-calorie diet on resting metabolic rate and serum tri-iodothyronine levels in obese children. Eur J Pediatr. 1999;158:446-450.
  61. Magarey A, Perry R, Baur L, Steinbeck K, Sawyer M, Hills A, Wilson G, Lee A, Daniels L. A parent-led family-focused treatment program for overweight children aged 5 to 9 years: the PEACH RCT. Pediatr. 2011;127:214-222.
  62. Reinehr T, Kleber M, Lass N, Toschke A. Body mass index patterns over 5 y in obese children motivated to participate in a 1-y lifestyle intervention: age as a predictor of long-term success. Amer J Clin Nutr.2010;91:1156-1171.
  63. Stern M, Bartley M, Duggirala R, Bradshaw B. Birth weight and the metabolic syndrome: thrifty phenotype or thrifty genotype? Diab Met Res Rev. 2000;16:88-93.
  64. Garrow JS, Webster J. Are pre-obese people energy thrifty? Paper presented at: Lancet1985
  65. Stunkard A, Foch T, Hrubec Z. A twin study of human obesity. JAMA. 1986;256:51-54.
  66. Elder S, Roberts S, McCrory M, Das S, Fuss P, Pittas A, Greenberg A, Heymsfield S, Dawson-Hughes B, Bouchard Jr. T, Saltzman E, Neale M. Effect of body composition methodology on heritability estimation of body fatness. Open Nutr J. 2012;5:48-58.
  67. Leibel R, Chua S, Rosenbaum M. Chapter 157. Obesity. In: Scriver C, Beaudet A, Sly W, Valle D, eds. The metabolic and molecular bases of inherited disease. Vol III. eigth ed. New York: McGraw-Hill; 2001:3965-4028.
  68. Oppert J-M, Dussault JH, Tremblay A, Despres J-P. Thyriod hormones and thyrotropin variations during long term overfeeding in identical twins. J Clin Endocrinol Metab. 1994;79:547-553.
  69. Poehlman E, Tremblay A, fontaine E. Genotype dependency of the thermic effect of a meal and associated hormonal changes following short-term overfeeding. Metabolism. 1986;35:30-36.
  70. Poehlman E, Despres J, Marcotte M, Tremblay A, Theriault G, Bouchard C. Genotype dependency of adaptation in adipose tissue metabolism after short-term overfeeding. Am J Physiol. 1986;250:E480-E485.
  71. Poehlman ET, Tremblay A, Despres JP. Genotype controlled changes in body composition and fat morphology following overfeeding in twins. Am J Clin Nutr. 1986;43:723-731.
  72. Tremblay A, Poehlman ET, Nadeau A, Dessault J, Bouchard C. Heredity and overfeeding-induced changes in submaximal exercise VO2. J Appl Physiol. 1987;62:539-544.
  73. Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, Dussault J, Moorjani S, Pinault S, Fournier G. The response to long-term overfeeding in identical twins. N Engl J Med. 1990;322:1477-1482.
  74. Rosenbaum M, Leibel R. Pathophysiology of childhood obesity. Advances in Pediatrics. 1988;35:73-137.
  75. Lehtovirta M, Kaprio J, Forsblom C, Eriksson J, Tuomilehto J, Groop L. Insulin sensitivity and insulin secretion in monozygotic and dizygotic twins. Diabetologia. 2000;43:285-293.
  76. Sun M, Gower B, Bartolucci A, Hunter G, Figueroa-Colon R, Goran M. A longitudinal study of resting energy expenditure relative to body composition during puberty in african american and white children. Am J Clin Nutr.2001;73:308-315.
  77. Llewellyn C, Trzaskowski M, Plomin R, Wardle J. From modeling to measurement: developmental trends in genetic influence on adiposity in childhood. Obes. 2014;22:1756-1761.
  78. Leibel R, Bahary N, Friedman J. Genetic variation and nutrition in obesity. In: Simopoulos A, Childs B, eds. Genetic variation and nutrition. Basel: Karger; 1990:90-101.
  79. Burnett L, Hubner G, LeDuc C, Morabito M, Carli J, Leibel R. Loss of the imprinted, non-coding Snord116 gene cluster in the interval deleted in the Prader Willi syndrome results in murine neuronal and endocrine pancreatic developmental phenotypes. Hum Mol Genet. 2017;26:4606-4616.
  80. Burnett L, LeDuc C, Sulsona C, Paull D, Rausch R, Eddiry S, Carli J, Morabito M, Skowronski A, Hubner G, Zimmer M, Wang L, Day R, Levy B, Fennoy I, Dubern B, Clement CPK, Butler M, Rosenbaum M, Salles J, Tauber M, Driscoll D, Egli D, Leibel R. Deficiency in prohormone convertase PC1 impairs prohormone processing in Prader-Willi syndrome. J Clin Invest. 2017;127:293-305.
  81. Manco M, Callapiccola B. Geneticsof pediatric obesity. Pediatr. 2012;130:123-133.
  82. Loos R. Genetic determinants of common obesity and their value in prediction. Best Pract Res Clin Endocrinol Metab. 2012;26:211-226.
  83. Ghosh S, Bouchard C. Convergence between biological, behavioural and genetic determinants of obesity. Nat Rev Genet. 2017;18:731-748.
  84. Vimaleswaran K, Tachmazidou J, Zhao J, Hirschhorm J, Dudbridge F, Loos R. Candidate genes for obesity-susceptibility show enriched association within a large genome-wide association study for BMI. Hum Mol Gen.2012;21:4537-4542.
  85. Speliotes EK WC, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, Lango Allen H, Lindgren CM, Luan J, Mägi R, Randall JC, Vedantam S, Winkler TW, Qi L, Workalemahu T, Heid IM, Steinthorsdottir V, Stringham HM, Weedon MN, Wheeler E, Wood AR, Ferreira T, Weyant RJ, Segrè AV, Estrada K, Liang L, Nemesh J, Park JH, Gustafsson S, Kilpeläinen TO, Yang J, Bouatia-Naji N, Esko T, Feitosa MF, Kutalik Z, Mangino M, Raychaudhuri S, Scherag A, Smith AV, Welch R, Zhao JH, Aben KK, Absher DM, Amin N, Dixon AL, Fisher E, Glazer NL, Goddard ME, Heard-Costa NL, Hoesel V, Hottenga JJ, Johansson A, Johnson T, Ketkar S, Lamina C, Li S, Moffatt MF, Myers RH, Narisu N, Perry JR, Peters MJ, Preuss M, Ripatti S, Rivadeneira F, Sandholt C, Scott LJ, Timpson NJ, Tyrer JP, van Wingerden S, Watanabe RM, White CC, Wiklund F, Barlassina C, Chasman DI, Cooper MN, Jansson JO, Lawrence RW, Pellikka N, Prokopenko I, Shi J, Thiering E, Alavere H, Alibrandi MT, Almgren P, Arnold AM, Aspelund T, Atwood LD, Balkau B, Balmforth AJ, Bennett AJ, Ben-Shlomo Y, Bergman RN, Bergmann S, Biebermann H, Blakemore AI, Boes T, Bonnycastle LL, Bornstein SR, Brown MJ, Buchanan TA, Busonero F, Campbell H, Cappuccio FP, Cavalcanti-Proença C, Chen YD, Chen CM, Chines PS, Clarke R, Coin L, Connell J, Day IN, den Heijer M, Duan J, Ebrahim S, Elliott P, Elosua R, Eiriksdottir G, Erdos MR, Eriksson JG, Facheris MF, Felix SB, Fischer-Posovszky P, Folsom AR, Friedrich N, Freimer NB, Fu M, Gaget S, Gejman PV, Geus EJ, Gieger C, Gjesing AP, Goel A, Goyette P, Grallert H, Grässler J, Greenawalt DM, Groves CJ, Gudnason V, Guiducci C, Hartikainen AL, Hassanali N, Hall AS, Havulinna AS, Hayward C, Heath AC, Hengstenberg C, Hicks AA, Hinney A, Hofman A, Homuth G, Hui J, Igl W, Iribarren C, Isomaa B, Jacobs KB, Jarick I, Jewell E, John U, Jørgensen T, Jousilahti P, Jula A, Kaakinen M, Kajantie E, Kaplan LM, Kathiresan S, Kettunen J, Kinnunen L, Knowles JW, Kolcic I, König IR, Koskinen S, Kovacs P, Kuusisto J, Kraft P, Kvaløy K, Laitinen J, Lantieri O, Lanzani C, Launer LJ, Lecoeur C, Lehtimäki T, Lettre G, Liu J, Lokki ML, Lorentzon M, Luben RN, Ludwig B; MAGIC, Manunta P, Marek D, Marre M, Martin NG, McArdle WL, McCarthy A, McKnight B, Meitinger T, Melander O, Meyre D, Midthjell K, Montgomery GW, Morken MA, Morris AP, Mulic R, Ngwa JS, Nelis M, Neville MJ, Nyholt DR, O'Donnell CJ, O'Rahilly S, Ong KK, Oostra B, Paré G, Parker AN, Perola M, Pichler I, Pietiläinen KH, Platou CG, Polasek O, Pouta A, Rafelt S, Raitakari O, Rayner NW, Ridderstråle M, Rief W, Ruokonen A, Robertson NR, Rzehak P, Salomaa V, Sanders AR, Sandhu MS, Sanna S, Saramies J, Savolainen MJ, Scherag S, Schipf S, Schreiber S, Schunkert H, Silander K, Sinisalo J, Siscovick DS, Smit JH, Soranzo N, Sovio U, Stephens J, Surakka I, Swift AJ, Tammesoo ML, Tardif JC, Teder-Laving M, Teslovich TM, Thompson JR, Thomson B, Tönjes A, Tuomi T, van Meurs JB, van Ommen GJ, Vatin V, Viikari J, Visvikis-Siest S, Vitart V, Vogel CI, Voight BF, Waite LL, Wallaschofski H, Walters GB, Widen E, Wiegand S, Wild SH, Willemsen G, Witte DR, Witteman JC, Xu J, Zhang Q, Zgaga L, Ziegler A, Zitting P, Beilby JP, Farooqi IS, Hebebrand J, Huikuri HV, James AL, Kähönen M, Levinson DF, Macciardi F, Nieminen MS, Ohlsson C, Palmer LJ, Ridker PM, Stumvoll M, Beckmann JS, Boeing H, Boerwinkle E, Boomsma DI, Caulfield MJ, Chanock SJ, Collins FS, Cupples LA, Smith GD, Erdmann J, Froguel P, Grönberg H, Gyllensten U, Hall P, Hansen T, Harris TB, Hattersley AT, Hayes RB, Heinrich J, Hu FB, Hveem K, Illig T, Jarvelin MR, Kaprio J, Karpe F, Khaw KT, Kiemeney LA, Krude H, Laakso M, Lawlor DA, Metspalu A, Munroe PB, Ouwehand WH, Pedersen O, Penninx BW, Peters A, Pramstaller PP, Quertermous T, Reinehr T, Rissanen A, Rudan I, Samani NJ, Schwarz PE, Shuldiner AR, Spector TD, Tuomilehto J, Uda M, Uitterlinden A, Valle TT, Wabitsch M, Waeber G, Wareham NJ, Watkins H; Procardis Consortium, Wilson JF, Wright AF, Zillikens MC, Chatterjee N, McCarroll SA, Purcell S, Schadt EE, Visscher PM, Assimes TL, Borecki IB, Deloukas P, Fox CS, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, Mohlke KL, O'Connell JR, Peltonen L, Schlessinger D, Strachan DP, van Duijn CM, Wichmann HE, Frayling TM, Thorsteinsdottir U, Abecasis GR, Barroso I, Boehnke M, Stefansson K, North KE, McCarthy MI, Hirschhorn JN, Ingelsson E, Loos RJ. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet. 2010;42:937-948.
  86. Frayling T, Timpson N, Weedon M, Zeggini E, Greathy R, Lindgren M, Perry J, Elliott K, Lango H, Rayner N, Shields B, Harries L, Barrett J, Ellard S, Groves C, Knight B, Patch A, Ness A, Ebrahim S, Lawlor D, Ring S, Ben-Shiomo Y, Jarvelin M, Sovio U, Bennett A, Meltzer D, Ferrucci L, Loos R, Barroso I, Wareham N, Karpe F, Owen K, Cardon L, Walker M, Hitman G, Palmer C, Doney A, Morris A, Smith G, Hattersley A, McCarthy M. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316:888-894.
  87. Liu G, Zhu H, Dong Y, Pdoolsky R, Treiber F, Snieder H. Inluence of common variants in FTO and near INSIG2 and MC4R on growth curves for adiposity in African- and European-American youth. Eur J Epidemiol.2011;26:463-473.
  88. Haupt A, Thamer C, Staiger H, Tschritter O, Kirchhoff K, Machiago F, Haring H, Stefan N, Fritsche A. Variation in the FTO gene influences food intake but not energy expenditure. Exp Clin Endocrinol Diabetes.2009;117:194-197.
  89. Wardle J, Llewellyn C, Sanderson S, Plomin R. The FTO gene and measured food intake in children. Int J Obes. 2008;33:42-45.
  90. Cecil J, Tavendale R, Watt P, Hetherington M, Palmer C. An obesity-associated FTO genevariant and increased energy intake in children. N Eng J Med. 2008;359:2558-2566.
  91. Hakanen M, Raitakari O, Lehtmake T, Peltonen N, Pahkala K, Silanmaki L, Lagstrom H, Biikari H, Simell O, Tonnemaa T. FTO genotype Is associated with Body Mass Index after the age of 7 years but not with energy intake or leisure-time physical activity

J Clin Endocrinol Metab. 2009;Epub ahead of print.

  1. Speakman J, Rance K, Johnstone A. Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity. 2008;16:1961-1965.
  2. Barkeling B, Rossner S, Sjoberg A. Methodological studies on single meal food intake characteristics in normal weight and obese men and women. Int J Obes. 1995;19:284-290.
  3. Himaya A, Louis-Sylvestre J. The effect of soup on satiation. Appetite. 1998;30:199-210.
  4. Bowen J, Noakes M, Clifton P. Appetite regulatory hormone responses to various dietary proteins differ by body mass index status despite similar reductions in ad libitum energy intake. J Clin Endocrinol Metab. 2006;91:2913-2919.
  5. Kissileff H, Thornton J, Torres M, Mayer L, Kalari V, Leibel R, Rosenbaum M. Leptin reverses decline in satiation in weight-reduced obese individuals. Am J Clin Nutr. 2012;95:309-317.
  6. Holliday R. DNA methylation and epigenetic inheritance. Phils Trans R Soc Lond B Biol Sci. 1990;326:329-338.
  7. Holliday R. Epigenetics: a historical overview. Epigenetics. 2006;1:76-80.
  8. Reynolds R, Jacobsen G, Drake A. What is the evidence in humans that DNA methylation changes link events in utero and later life disease? Clin Endocrinol. 2013;78:814-822.
  9. Inadera H. Developmental origins of obesity and type 2 diabetes: molecular aspects and role of chemicals. Environ Health Prev Med. 2013;18:185-197.
  10. Kirchner H, Osler M, Krook A, Zierath J. Epigenetic flexibility in metabolic regulation: disease cause and prevention? Trends Cell Biol. 2013;23:203-209.
  11. Menzies K, Zhang H, Katsyuba E, Auwerx J. Protein acetylation in metabolism - metabolites and cofactors. Nat Rev Endocrinol. 2016;12:43-60.
  12. Zaiou M, El Amri H, Bakillah A. The clinical potential of adipogenesis and obesity-related microRNAs. Nutr Metab Cardiovasc Dis. 2017;Epub ahead of print.
  13. Guenard F, Deshaies Y, Cianflone K, Kral J, Marceau P, Vohl M. Differential methylation in glucoregulatory genes of offspring born before vs. after maternal gastrointestinal bypass surgery. Proc Nat Acad Sci USA.2013;110:11439-11444.
  14. Oken E, Kleinman K, Belfort M, Hammitt J, GIllman M. Associations of gestational weight gain with short- and longer-term maternal and child health outcomes. Am J Epidemiol. 2009;170:123-180.
  15. Ravelli G, Stein Z, Susser M. Obesity in young men after famine exposure in utero and early infancy. N Engl J Med. 1976;295:349-353.
  16. Ravelli A, Meulen Jvd, Michels R, Osmond C, Barker D, Hales C, Bleker O. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 1998;351:173-177.
  17. Moore V, Cockington R, Ryan P, Robinson J. The relationship between birthweight and blood pressure amplifies from childhood to adulthood. J Hypertens. 1999;17:883-888.
  18. Hattersley A, Tooke J. The fetal insulin hypothesis: an alternative explanation of the association oflow birthweight with diabetes and vascular disease. Lancet. 1999;353:1789-1792.
  19. Yarborough D, Barrett-Connor E, Kritz-Silverstein D, Wingard D. Birth weight, adult weight, and girth as predictors of the metabolic syndrome in postmenopausal women: the Rancho Bernardo Study. Diab Care.1998;21:1652-1658.
  20. Barker D. Maternal nutrition, fetal nutrition, and diseases later in life. Nutr. 1997;13:807-813.
  21. Godfrey K, Barker D. Fetal nutrition and adult disease. Am J Clin Nutr. 2000;71:1344S-1352s.
  22. Barker D, Clark P. Fetal undernutrition and disease in later life. Rev Reprod. 1997;2:105-112.
  23. Barker D. Maternal nutrtion, fetal nutrtion, and disease in later life. Nutr. 1997;13:807-813.
  24. Barker D. Fetal origins of cardiovascular disease. Ann Med. 1999;31(Suppl):3-6.
  25. Pettit D, Baird H, Allech K, Knowler W. Excessive obesity in offspring of Pima Indian women with diabetes during pregnancy. N Engl J Med. 1983;308:242-245.
  26. Pettitt DJ, Baird R, Aleck KA, Bennett PH, Knowler WC. Excessive obesity in offspring of Pima Indian women with diabetes during pregnancy. N Engl J Med. 1983;308:242-245.
  27. Pettitt DJ, Knowler WC, Bennett PH, Aleck KA, Baird HR. Obesity in offspring of diabetic Pima Indian women.Despite normal birth weight. Diabetes Care. 1987;10(1):76-80.
  28. Pettitt DJ, Aleck KA, Baird HR, Carraher MJ, Bennett PH, Knowler WC. Congential susceptibility to NIDDM: Role of interauterine environment. Diabetes. 1988;37:622-628.
  29. Entringer S, Buss C, Swanson J, Cooper D, Wing D, Waffarm F, Wakhwa P. Fetal programming of body composition, obesity, and metabolic function: the role of intrauterine stress and stress biology. J Nutr Metab.2012;2012:http://dx.doi.org/10.1155/2012/632548.
  30. Entringer S, Wadhwa P. Developmental programming of obesity and metabolic dysfunction: role of prenatal stress and stress biology. Nestle Nutr Inst Workshop Ser. 2013;74:107-120.
  31. LaGasse L, Gaskins R, Bada H, Shankaran S, Liu J, Lester B, Bauer C, Higgins R, Das A, Roberts M. Prenatal cocaine exposure and childhood obesity at nine years. 2011;33:188-197.
  32. Moore B, Sauder K, Shapiro A, Crume T, Kinney G, Dabelea D. Fetal exposure to cannabis and childhood metabolic outcomes: The Healthy Start Study. J Clin Endocrinol Metab. 2022;107:e2863-2869.
  33. Rosenbaum M. Passive prenatal exposure to cannabinoids promotes weight gain and dysglycemia in childhood. J Clin Endocrinol Metab. 2022;2022 Apr 18:dgac227. doi: 10.1210/clinem/dgac227. Epub ahead of print.
  34. Hediger M, Overpeck M, Kuczmarski R, Ruan W. Association between infant breastfeeding and overweight in young children. JAMA. 2001;285:2506-2507.
  35. Horta B, de Mola C, Victoria C. Long term consequences of breastfeeding on cholesterol, obesity, systolic blood pressure, and type 2 diabetes: A systematic review and meta-analysis. Acta Pediatric. 2015;105:30-37.
  36. Yan J, Liu L, Zhu Y, Huaming G, Wang P. The association between breastfeeding and childhood obesity: A meta-analysis. BMC Public Health. 2014;14:1267.
  37. Rzehak P, Oddy W, Mearin M, Grote V, Mori T, Szajewska H, Shamir R, Koletzko S, Beilin L, Hng R, Koletzko B, WP10 working group of the Early Nutriton Project. Infant feeding and growth trajectory patterns in childhood and body composition in young adulthoo. Amer J Clin Nutr. 2017;106:568-580.
  38. Osei-Assibey G, Dick S, Macdiarmid J, Semple S, Reily J, Ellaway A, Cowie H, McNeill G. The influence of the food environment on overweight and obesity in young children: a systematic review. BMJ Open 2012;2.
  39. Dietz W, Gortmaker S. Do we fatten our children at the television set ? Obesity and television viewing in children and adolescents. Pediatrics. 1985;75:807-812.
  40. Salbe A, Nicolson M, Ravussin E. Total energy expenditure and physical activity correlate with plasma leptin concentrations in five-year-old children. J Clin Invest. 1997;99:592-595.
  41. Ku L, Shapiro L, Crayford P, Huenemann R. Body composition and physical activity in 8 year old children. Am J Clin Nutr. 1981;34:2770-2775.
  42. Davies P, Gregory J, White A. Physical activity and body fatness in pre-school children. Int J Obes. 1995;19:6-10.
  43. Borzekowski D, Robinson T. The 30-second effect: an experiment revealing the impact of television commercials on food preferences of preschoolers. J Am Diet Assoc. 2001;101:42-46.
  44. Krespo C, Smit E, Troiano R, Bartlett S, Macera C, Andersen R. Television watching, energy intake, and obesity in US children: results from the third National Health and Nutrition Examination Survey, 1988-1994. Arch Pediatr Adolesc Med. 2001;155:360-365.
  45. Cunningham S, Kramer M, Narayan K. Incidence of childhood obesity in the United States. N Eng J Med.2014;370:403-411.
  46. Nanri H, Shirasawa T, Ochiai J, Nomoto S, Hoshino H, Kokaze A. Rapid weight gain during infancy and early childhood is related to higher anthropometric measurements in preadolescence. Child Care Health Dev.2017;43:435-440.
  47. Munthali R, Kagura J, Lombard Z, Norris S. Early life growth predictors of childhood adiposity trajectories and future risk for obesity: Birth to twenty cohort. Child Obes. 2017;13:384-391.
  48. Braun J, Kalkwarf H, Papandonatos G, Chen A, Lamphear B. Patterns of early life body mass index and childhood overweight and obesity status at eight years of age. BMC Pediatr. 2018;18:161.
  49. Lu Y, Pearce A, Li L. Weight gain in early years and subsequent body mass index trajectories across birth weight groups: a prospective longitudinal study. Eur J Publ Heal. 2020;30:316-322.
  50. Stock K, Nagrani R, Gande N, Bernar B, Staudt A, Willeit P, Geiger R, Knoflach M, UKiechl-Kohlendorfer. Birth weight and weight changes from infancy to early childhood as predictors of body mass index in adolescence. J Pediatr. 2020;222:120-126.
  51. Ogden C, Fryar C, Hales C, Carroll M, Aoki Y, Freedman D. Differences in obesity prevalence by demographic characteristics and urbanization level US children and adolescents, 2013-2016. JAMA. 2018;319:2410-2418.
  52. Gorski Findling M, Wolfson J, Rimm E, Bleich S. Differences in the neighborhood retail food environment and obesity among US children and adolescents by SNAP participation. Obesity. 2018;26:1063-1071.
  53. Elbel B, Tamura K, McDermott Z, Wu E, Schwartz A. Childhood Obesity and the food environment: A population-based sample of public school children in New York City. Obesity. 2020;28:65-72.
  54. Jia P, Zou Y, Wu Z, Zhang D, Wu T, Smith M, Xiao Z. Street connectivity, physical activity, and childhood obesity: A systematic review and meta-analysis. Obes Rev. 2021;22:e12943.
  55. Jia P, Das S, Rohli K, Rohli R, Ma Y, Yu C, Pan X, Zhou W. Natural environment and childhood obesity: A systematic review. Obes Rev. 2021;22:e13097.
  56. Foster C, Moore J, Singletary C, Skelton J. Physical activity and family-based obesity treatment: a review of expert recommendations on physical activity in youth. Clinical Obesity. 2018;8:68-79.
  57. Rosenbaum M, Nonas C, Weil R, Horlick M, Fennoy I, Vargas I, Kringas P, El Camino Diabetes Prevention Group. School-based intervention acutely improves insulin sensitivity and decreases inflammatory markers in early adolescence. J Clin Endocrinol Metab. 2007;92:504-508.
  58. Bruce A, Martin L, Savage C. Neural correlates of pediatric obesity. Prev Med. 2011;52:S29-35.
  59. Telama R, Yang X, Vikari J, Valimaki I, Wanne O, Raitakari O. Physical activity from childhood to adulthood: a 21-year tracking study. Am J Prev Med. 2005;28:267-273.
  60. Craigie A, Lake A, Kelly S, Adamson A, Mathers J. Tracking of obesity-related behaviours from childhood to adulthood: a systematic review. Maturitas. 2011;70:266-284.
  61. Marson E, Delevatti R, Pardo A, Netto N, Kruel L. Effects of aerobic, resistance, and combined exercise training on insulin resistance markers in overweight or obese children and adolescents: A systemic review and meta-analysis. Prev Med. 2016;93:211-218.
  62. Estaki M, Pither J, Baumeister P, Little J, Gill S, Ghost S, Ahmadi-Vand Z, Marsden K, Gibson D. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome. 2016;4:42-55.
  63. Dias K, Green D, Ingul C, Pavey T, Coombes J. Exercise and vascular function in child obesity: A meta-analysis. Pediatr. 2015;136:e648-659.
  64. Gonzalez-Ruiz K, Ramirez-Velez R, Correra-Bautista J, Peterson M, Garcia-Hermoso A. The effects of exercise on abdominal fat and liver enzymes in pediatric obesity: A systematic review and meta-analysis. Child Obes.2017;13:272-282.
  65. Nooijen C, Galanti M, Engstrom K, Moller J, Forsell Y. Effectiveness of interventions on physical activity in overweight or obese children: a systematic review and meta-analysis including studies with objectively measured outcomes. Obes Rev. 2017;18:1950213.
  66. Mei H, Xiong Y, Xie S, Guo S, Li Y, Guo B, Zhang J. The impact of long-term school-based physical activity interventions on body mass index of primary school children - a meta-analysis of randomized controlled trials. BMC Public Health. 2016;16:205.
  67. Chaput J, Lambert M, Mathieu M, Tremblay M, Loughlin J, Tremblay A. Physical activity vs. sedentary time: independent associations with adiposity in children. Pediatr Obes. 2012;7:251-258.
  68. Cappucio F, Taggart F, Kandala N-B, Currie A, Stranges S, Miller M. Meta-analysis of short sleep duration and obesity in children and adults. Sleep. 2008;31:619-626.
  69. Julian V, Haschke F, Fearnbach N, Bomahr J, Pixner T, Furthner D, Weghuber D, Thivel D. Effects of movement behaviors on overall health and appetite control: Current evidence and perspectives in children and adolescents. Curr Obes Rep. 2022;11:10-22.
  70. Jones R, Hinkley T, Okely A, Salmon J. Tracking physical activity and sedentary behavior in childhood: a systematic review. Amer J Prevent Med. 2013;44(651-58).
  71. Janssen I, LeBlanc A. Int J Behav Nutr Phys Act. 7. 2010:40.
  72. Poitras V, Gray C, Borghese M, Carson V, Chaput J, Jansses I, Katzmarzyk P, Pate R, Corber S, Kho M, Sampson M, Tremblay M. Systematic review of the relationships between objectively measured physical activity and health indicators in school-aged children and youth. Appl Physiol Nutr Metab. 2016;41:S197-239.
  73. Carson V, Tremblay M, Chaput J, CHastin S. Associations between sleep duration, sedentary time, physical activity, and health indicators among Canadian children and youth using compositional analyses. Appl Physiol Nutr Metab. 2016;4(a294-302).
  74. Webber L, Srinivasan S, Wattigneyy W, Berenson G. Tracking of serum lipids and lipoproteins from childhood to adulthood: the Bogalusa Heart Study. Am J Epidemiol. 1991;133:884-899.
  75. Must A, Jacques P, Dallai G, Bajema D, Dietz W. Long-term morbidity and mortality of overweight adolescents. N Eng J Med. 1992;327:1350-1355.
  76. Pont S, Puhl R, Cook S, Slusser W, Section on Pediatric Obesity tOS. Stigma experienced by children and adolescents with obesity. Pediatr. 2017;140:e20173034.
  77. Juvonen J, Lessard L, Shater H, Suchlit L. Emotional implicaiton of weight stigma across middle school: The role of weight-based peer discrimiatnion. J Clin Child Adolesc Psychol. 2017;46:150-158.
  78. Pugliese M, Lifshitz F, Grad G, Fort P, Marks-Katz M. Fear of obesity. N Engl J Med. 1983;309:513-518.
  79. Magnussen C, Venn A, Thomson R, Huonala J, Srinivasan S, Vilkari J, Berenson G, Dwyer T, Taitakari O. The association of pediatric low- and high-density lipoprotein cholesterol dyslipidemia classifications and change in dyslipidemia status with carotid intima-media thickness in adulthood evidence from the cardiovascular risk in Young Finns study, the Bogalusa Heart study, and the CDAH (Childhood Determinants of Adult J Am Coll Cardiol. 2009;53:860-869.
  80. Juhola J, Magnussen C, Viikari J, Kahonen M, Hutri-Kahonen N, Julia A, Lehtimaki T, Akerblom H, Pietikainen M, Laitinen T, Jokinen E, Taittonen L, Raitakari O, Juonala M. Tracking of serum lipid levels, blood pressure, and body mass index from childhood to adulthood: the Cardiovascular Risk in Young Finns Study. J Pediatr.2011;159:584-590.
  81. Freedman D, Patel D, Srinivasan S, Chen W, Tang R, Bond M, Berenson G. The contribution of childhood obesity to adult carotid intima-media thickness: the Bogalusa Heart Study. Int J Obes. 2008;32:749-756.
  82. Clarke W, Schrott H, Leaverton P, Connor W, Lauer R. Tracking of blood lipids and blood pressure in school age children. The Muscatine study. Circulation. 1978;58:626-634.
  83. Lawrence M, Divers J, Isom S, Sayadh S, Imperatore G, Pihoker C, Marcovina S, Mayer-Davis E, Hamman R, Dolan L, Dabelea D, Pettt D, Liese A, for the SEARCH for Diabetes in Youth Study Group. Trends in Prevalence of Type 1 and Type 2 Diabetes in Children and Adolescents in the US, 2001-2017. JAMA. 2021;326:717-727.
  84. Liu J, Li Y, Zhang S, Yi S, Liu J. Trends in Prediabetes Among Youths in the US From 1999 Through 2018. JAMA Pediatr. 2022;176:608-611.
  85. Barrett C, Kouama A, Alvarez P, Chow W, Lundeen E, Perrine C, Pavkov M, Rolka D, Wiltz J, Bull-Otterson L, Gray S, Boehmer T, Gundlapalli A, Seigel D, Kompaniyets L, Goodman A, Mahon B, Tauxe R, Remley K, Saydah S. Risk for newly diagnosed diabetes >30 days after SARS-COV-2 infection among persons aged <18 years — United States, March 1, 2020–June 28, 2021. MMWR. 2022;71:59-65.
  86. The Rise Consortium. Metabolic contrasts between youth and adults with impaired glucose tolerance or recently diagnosed type 2 diabetes: II. Observations using the oral glucose tolerance test. Diab Care. 2018;41:1707-1716.
  87. Consortium TR. Obesity and insulin sensitivity effects on cardiovascular risk factors: Comparisons of obese dysglycemic youth and adults. Pediatr Diab. 2019;20(849-60).
  88. The Today Study Group. Rapid rise in hypertension and nephropathy in youth with type 2 diabetes: the TODAY clinical trial. Diab Care. 2013;36:1735-1741.
  89. The Today Study Group, P Bjornstad, Drews K, Caprio S, Gubitosi-Klug R, Nathan D, Tesfaldet B, Truggestad J, White N, Zeitler P. Long-term complications in youth-onset type 2 diabetes. N Eng J Med. 2021;385:416-426.
  90. Cersosimo E, Triplitt C, Mandarino L, DeFronzo R. Pathogenesis of type 2 diabetes mellitus. In: DeGroot L, Rebar R, Singer F, Vinik A, eds. Endotext. South Dartmoutn (MA)2000.
  91. Kahn C, Vincent D, Doria A. Genetics of non-insulin-dependent (type II) diabetes mellitus. Annu Rev Med.1996;47:509-531.
  92. Natali A, Muscelli E, Mari A, Balkau B, Walker M, Tura A, Anderwald C, Ferrannini E. Insulin sensitivity and beta-cell function in the offspring of type 2 diabetes patients: impact of line of inheritance. J Clin Endocrinol Metab. 2010;95:4703-4711.
  93. Chernausek S, Arslanian S, Caprio S, Copeland K, El ghromi L, Kelsey M, Koontz M, Orsi C, Wilfley D. Relationship between parental diabetes and presentation of metabolic and glycemic function in youth with type 2 diabetes: baseline findings from the TODAY Trial. Diab Care. 2016;39:110-117.
  94. Johnston C, Ward W, Beard J, McKnight B, Porte D. Islet function and insulin sensitivity in the non-diabetic offspring of conjugal type 2 diabetic patients. Diab Med. 1990;7:119-125.
  95. Polonsky KS, Sturis J, Bell GI. Non-insulin dependent diabetes mellitus - A genetically programmed failure of the beta cell to compensate for insulin resistance. N Engl J Med. 1996;334(12):777-783.
  96. Dabelea D, Pettitt D, Jones K, Arslanian S. Type 2 diabetes mellitus in minority children and adolescents. Endocrinol Metab Clin N Amer. 1999;28(709-29).
  97. Troiano R, Flegal K, Kuczmarski R, Campbell S, Johnson C. Overweight prevalence and trends for children and adolescents. Arch Pediatr Adol Med. 1995;149:1085-1091.
  98. Caprio S, Hyman L, McCarthy S, Lange R, Bronson M, Tamborlane W. Fat distribution and cardiovascular risk factors in obese adolescent girls: importance of the intraabdominal fat depot. Am J Clin Nutr. 1996;64:12-17.
  99. Caprio L. Central adiposity and its metabolic correlates in obese adolescent girls. N Engl J Med. 1995.
  100. Caprio S, Tamborlane W. The metabolic impact of obesity in childhood. Endocrinol Metab Clin N Amer.1999;28:731-747.
  101. Frerichs R, Webber L, Voors A, Srinivasan S, Berenson G. Cardiovascular disease risk factor variables in children at two successive years - the Bogalusa heart study. J Chron Dis. 1979;32:251-262.
  102. Freedman D, Srinivasan S, Burke G, Shear C, Smoak C, Harsha D, Webber L, Berenson G. Relationship of body fat distribution to hyperinsulinemia in children and adolescents: The Bogalusa heart study. Am J Clin Nutr.1987;46:403-410.
  103. Kelishadi R, Mirmoghtadaee P, Majafi H, Keikha M. Systematic review on the association of abdominal obesity in children and adolescents with cardio-metabolic risk factors. J Res Med Sci. 2015;20:294-307.
  104. Katzmarzyk P, Shen W, Baxter-Jones A, Bell J, Butte N, Demerath E, Gilsanz V, Goran M, Hirschler V, Hu H, Maffeis C, Malina R, Muller M, Pietrobelli A, Wella J. Adiposity in children and adolescents: correlates and clinical consequences of fat stored in specific body depots. Pediatr Obes. 2012;7:e42-61.
  105. Sharma A, Metzger D, Daymont C, Hadjiyannakis S, Rodd C. LMS tables for waist-circumference and waist-height ratio Z-scores in children aged 5–19 y in NHANES III: association with cardio-metabolic risks. Pediatr Res. 2015;78:723-729.
  106. Osei K, Schuster D. Effects of race and ethnicity on insulin sensitivity, blood pressure, and heart rate in three ethnic populations: comparative studies in African-Americans, African immigrants (Ghanaians), and white Americans using ambulatory bloodpressure monitoring. Am J Hypertens. 1996;9:1157-1164.
  107. Gower B, Nagy T, Trowbridge C, Dezenberg C, Goran M. Fat distribution and insulin response in prepubertal African American and white children. Am J Clin Nutr. 1998;67:821-827.
  108. Gower B, Nagy T, Goran M. Visceral fat, insulin sensitivity, and lipids in prepubertal children. Diabetes.1999;48:1515-1521.
  109. Yanovski J, Yanovski S, Filmer K, Hubbard V, Avila N, B; BL, Reynolds J, Flood M. Differences in body composition of black and white girls. Am J Clin Nutr. 1996;64:833-839.
  110. Mitchell B, Kammerer C, Reinhart L, Stern M. NIDDM in Mexican-American families. Heterogeneity by age of onset. Diab Care. 1994;17:567-573.
  111. Daniels S. Obesity in the pediatric patient: cardiovascular complications. Prog Pediatr Cardiol. 2001;12:161-167.
  112. Sothern M, Loftin M, Blecker U, Udall J. Impact of significant weight loss on maximal oxygen uptake in obese children and adolescents. J Investig Med. 2000;48:411-416.
  113. C Boucher-Berry PS, DE Carey, SP Shelov, S Accacha IF, R Rapaport, Y Espinal, M Rosenbaum. Vitamin D, osteocalcin, and risk for adiposity as comorbidities in middle school children. J Bone Miner Res. 2012;27:283-293.
  114. Smotkin-Tangorra M, Purushothaman R, Gupta A, Nejati G, Anhalt H, Ten S. Prevalence of vitamin D insufficient in obese children and adolescents. J Pediatr Endocrinol Metab. 2007;20:817-823.
  115. Kumar S, Kelly A. Review of childhood obesity: from epidemiology, etiology, and comorbidities to clinical assessment and treatment. Mayo Clin Proc. 2017;92:251-265.
  116. Rubino F, Puhl R, Cummings D, Eckel R, Ryan D, Mechanick J, Nadglowski J, X Ramos Salas, Schauer P, Twenefour D, Apovian C, Aronne L, Batterham R, Berthoud H, Boza C, Busetto L, Dicker D, M De Groot, Eisenberg D, Flint S, Huang T, Kaplan L, Kirwan J, Korner J, Kyle T, Laferrere B, CW Le Roux, McIve L, Mingrone G, Nece P, Reid T, Rogers A, Rosenbaum M, R Seeley, AJ Torres, Dixon J. Joint international consensus statement for ending stigma of obesity. Nat Med. 2020;26:485-497.
  117. Delichatsios H, Hauser M, Burgess J, Eisenberg D. Sared medical appointments: A portal for nutrition and culinary education in primary care-a pilot feasibility project. Glob Adv Health Med. 2015;4:22-26.
  118. Roberts K, Polfuss M. Weight Stigma in children and adolescents , Recommendations for practice and policy. Nursing. 2022;52:17-24.
  119. Tomiyama A, Carr D, Granberg E, Major B, Robinson E, Sutin A, Brewis A. How and why weight stigma drives the obesity ‘epidemic’ and harms health. BMC Med. 2018;16:https://doi.org/10.1186/s12916-12018-11116-12915.
  120. Palad C, Yarlagadda S, Stanford F. Weight stigma and its impact on paediatric care. Curr Opin Endocrinol Diabetes Oges. 2019;26:19-24.
  121. Grobler L, Visser M, Seigfried N. Healthy Life Trajectories Initiative: Summary of the evidence base for pregnancy-related interventions to prevent overweight and obesity in children. Obes Rev. 2019;20:18-30.
  122. Institute of Medicine (US) and National Research Council (US) Committee to Reexamine IOM Pregnancy Weight Guidelines. Weight gain during pregnancy: Reexamining the guidelines. In: Rasmussen K, Yaktine A, eds. The National Academies Collection: Reports funded by National Institutes of Health. Washington (DC)2009.
  123. Chen Y, Ma G, Hu Y, Yang Q, Deavila J, Zhu M-J, Due M. Effects of maternal exercise during pregnancy on perinatal growth and childhood obesity outcomes: A meta-analysis and meta-regression. Sports Med.2021;51:2329-2347.
  124. Murray S, Reynolds R. Short- and long-term outcomes of gestational diabetes and its treatment on fetal development. Prenat Diagn. 2020;40:1085-1091.
  125. Legro R, Arslanian S, Ehrmann D, Hoeger K, Murad M, Pasquali R, Welt C. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab.2013;98:4565-4592.
  126. van Trotsenburg P, Stoupa A, Leger J, Rohrer T, Peters C, Fugazzola L, Cassio A, Heinrichs C, Beauloye V, Pohlenz J, Rodien P, Coutant R, Szinnai G, Murray P, Bartes B, Luton D, Salerno M, de Sanctis L, VIgone M, Krude H, Persani L, Polak M. Congenital Hypothyroidism: A 2020-2021 Consensus Guidelines Update-An ENDO-European Reference Network Initiative Endorsed by the European Society for Pediatric Endocrinology and the European Society for Endocrinology. Thyroid. 2021;31:387-419.
  127. Segni M. Disorders of the thyroid gland in infancy, childhood and adolescence. In: Feingold K, Anawalt B, BOyce A, CHrousos G, de Herder W, Dhatariya K, Dunga K, Hershman J, Hlfland J, Kaira S, Kaltsas G, Kosh C, Kopp P, Korbonits M, Kovacs C, Kuohung W, Laferrere B, Levy M, McGee E, McLachlan R, Morley J, New M, Stratakis C, Trence D, WIlson D, eds. Endotext [Internet]. South Dartmouth (MA): MDText.com; 2017.
  128. Barlow S, Dietz W. Obesity evaluation and treatment: Expert Committee recommendations. The Maternal and Child Health Bureau, Health Resources and Services Administration and the Department of Health and Human Services. Pediatr. 1998;102:E29.
  129. Epstein L. Family-based behavioural intervention for obese children. Int J Obes. 1996;20:S14-S21.
  130. Epstein L, Valoski A, Wing R, McCurley J. Ten-year follow-up of behavioral, family-based treatment of obese children. JAMA. 1990;264:2519-2523.
  131. Frohlich G, Pott W, Albavrak O, Hedebrand H, Pauli-Pott U. Conditions of long-term success in a lifestyle intervention for overweight and obese youths. Pediatr. 2011;12:8e779-785.
  132. Pott W, Albvarak O, Hedebrand J, Pauli-Pott U. Treating childhood obesity: family background variables and the child's success in a weight-control intervention. Int J Eat Disord. 2009;42:284-289.
  133. Reinher R, Brylak D, Alexy U, Dersting M, Andler W. Predictors to success in outpatient training to obese children and adolescents. Int J Obes. 2002;27:1087-1092.
  134. Epstein L, Gordy C, Raynor H, Beddome M, Kilanowski C, Paluch R. Increasing fruit and vegetable intake and decreasing fat and sugar intake in families at risk for childhood obesity. Obes Res. 2001;9:171-178.
  135. Vander Wal J, Mitchell E. Psychological complications of pediatric obesity. Pediatr Clin N Amer. 2011;58:1393-1401.
  136. U.S. Department of Agriculture and U.S. Department of Health and Human Services. Dietary guidelines for Americans. https://www.dietaryguidelines.gov/.2020.
  137. Roza AM, Shizgal HM, FRCS(C), FACS. The Harris Benedict equation reevaluated: resting energy requirements and the body cell mass. Am J Clin Nutr. 1984;40:168-182.
  138. Malik V, Hu F. Fructose and cardiometabolic health: What the evidence form sugar sweetened beverages tells us. J Am Coll Cardiol. 2015;66:1615-1624.
  139. Raissouni N, Kolesnikov A, Purushothaman R, Sinha S, Bhandari S, Bhangoo A, Malik S, Matthew R, Baillargeon J-P, Hernandez M, Rosenbaum M, Ten S, Geller D. Altered glucose disposition and insulin sensitivity in peri-pubertal first-degree relatives of women with polycystic ovary syndrome. . Int J Pedaitr Endocrinol. 2012 doi: 10.1186/1687-9856-2012-14.
  140. Malik V, Schulze M, Hu G. Intake of sugar-sweetened beverages and weight gain: a systematic review. Am J Clin Nutr. 2006;84:274-288.
  141. Wang Y, Bleich S, Gortmaker S. Increasing caloric contribution from sugar-sweetened beverages and 100% fruit juices among US children and adolescents, 1988-2004. Pediatr. 2008;121:e:1604-1614.
  142. Ford C, Slining M, Popkin B. Trends in dietary intake among US 2- to 6- year old children, 1989-2008. J Acad Nutr Diet. 2013;113:35-42.
  143. Banfield E, Liu Y, Davis J, Chang S, Frazier-Wood A. Poor adherence to US Dietary Guidelines for children and adolescents in the National Health and Nutrition Examination Survey population. J Acad Nutr Diet. 2016;115:21-27.
  144. Asta K, Miller A, Retzloff L, Rosenblum K, Kaciroti N, Lumeng J. Eating in the absence of hunger and weight gain in low-income toddlers. Pediatr. 2016;137:e20153786.
  145. Lansigan R, Edmond J, Gilbert-Diamond D. Understanding eating in the absence of hunger among young children: a systematic review of existing studies. Appetite. 2015;85:36-47.
  146. Steele E, Baraldi L, da Costa Louzada M, Moubarac J, Mozaffarian D, monteiro C. Ultra-processed foods and added sugars in the US diet: evidence from a nationally representative cross-sectional study. BMJ Open.2016;6:e009892.
  147. GIbney M. Ultra-processed foods: Definitions and policy issues. Curr Dev Nutr. 2019;3:nzy077.
  148. Stucker D, McKee M, Ebrahim S, Basu S. Manufacturing epidemics: The role of global producers in increased consumption of unhealthy commodities including processed foods, alcohol, and tobacco. PLoS Med.2012;9:e1001235.
  149. Fardet A. Minimally processed foods are more satiating and less hyperglycemic than ultra-processed foods: A preliminary study with 98 ready-to-eat foods. Food Funct. 2016;7:2338-2346.
  150. Srour B, Fezeu L, Kesse-Guyot E, Alles B, Mejean C, Andrianasolo T, Chazelas E, Deschasaux M, Hercberg S, Galan P, Monteiro C, Julia C, Touvier M. Ultra-processed food intake and risk of cardiovascular disease: prospective cohort study (NutriNet-Santé). BMJ. 2019;465:I1451.
  151. Juul F, Martinez-Steele E, Parekh N, Monteiro C, CHang V. Ultra-processed food consumption and excess weight among US adults. Br J Nutr. 2018;120:90–100.
  152. de Miranda R, Rauber F, Levy R. Impact of ultra-processed food consumption on metabolic health. Curr Opin Lipidol. 2021;32:24-37.
  153. Hall K, Ayuketah A, Bruchta R, Cai H, Cassimatis T, Chen K, Chung S, Costa E, Courville A, Darcey V, Fletcher L, Forde C, Gharib A, Guo J, Howard R, Joseph P, McGehee S, Ouwerkerk R, Raisinger K, Rozga I, Stagliano M, Walter M, Walter P, Yang S, Zhou M. Ultra-Processed Diets Cause Excess Calorie Intake and Weight Gain: An Inpatient Randomized Controlled Trial of Ad Libitum Food Intake. Cell Metab. 2019;30:67-77.
  154. Costa C, Del-ponte B, Assuncao M, Santos I. Consumption of ultra-processed foods and body fat during childhood and adolescence: a systematic review. Pub Health Nutr. 2018;21:148-159.
  155. Leffa P, Hoffman D, Rauber F, Sangalli C, Valmorbida J, Vitolo M. Longitudinal associations between ultra-processed foods and blood lipids in childhood. Br J Nutr. 2020;124:341-348.
  156. Gow M, Ho M, BUrrows T, Baur L, Hutchesson M, Cowell C, Collins C, Garnett S. Impact of dietary macronutrient distribution on BMI and cardiometabolic outcomes in overweight and obese children and adolescents: a systematic review. Nutr Rev. 2014;72:453-470.
  157. Foster G, Wyatt H, Hill J, Makris A, Rosenbaum D, Brill C, Stein R, Mohammed B, Miller B, Rader D, Zemel B, Wadden T, Tenhave T, Newcomb C, Klein S. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a randomized trial. Ann Int Med. 2010;153:147-157.
  158. Mirza N, Palmer M, Sinclair K, McCarter R, He J, Evvling C, Ludwig D, Yanovski J. Effects of a low glycemic load or a low-fat dietary intervention on body weight in obese Hispanic American children and adolescents: a randomized controlled trial. Amer J Clin Nutr. 2013;97:276-285.
  159. Davis J, Whaley S, Goran M. Effects of breastfeeding and low sugar-sweetened beverage intake on obesity prevalence in Hispanic toddlers. Amer J Clin Nutr. 2012;95:3-8.
  160. Mirza N, Palmer M, Sinclair K, McCarter R, HE J, Ebbeling C, Ludwig D, Yanofski J. Effects of a low glycermic load or low fat-dietary intervention on body weight in obese Hispanic American children and adolescents: a randomized controlled trial. Am J Clin Nutr. 2013;97:276-285.
  161. Hall K, Guo J. Obesity energetics: Body weight regualtion and effects of diet composition. Gastroenterol.2017;152:1718-1727.
  162. Ebbeling C, Feldman H, CHomitz V, Antonelli T, Fortmaker S, Osganian S, Ludwig D. A randomized trial of sugar-sweetened beverages and adolescent body weight. N Eng J Med. 2012;367:1407-1416.
  163. Albright A, Franz M, Hornsby G, A AK, Marrero D, Ullrich I, Verity L. American College of Sports Medicine position stand. Exercise and type 2 diabetes. Med Sci Sports. 2000;32:1345-1360.
  164. Epstein L, Wing R, Penner B, Kress M, Koeske R. The effect of controlled exercise on weight loss in obese children. J Pediatr. 1985;107:358-361.
  165. Kelley D, Goodpaster B. Effects of exercise on glucose homeostasis in type 2 diabetes mellitus. Med Sci Sports Exerc. 2001;33:S495-501.
  166. Sekine M, Yamagami T, Handa K, Saito T, Nanri S, Kawaminami K, Tokue N, Yoshida K, Kagamimori S. A dose-response relationship between short sleeping hours and childhood obesity: results of the Toyama Birth Cohort Study. Child Care Health Dev. 2002;28:163-170.
  167. Anderson S, Whitaker R. Household routines and obesity in US pre-school-aged children. Pediatr.2010;125:420-428.
  168. Felso R, Lohner S, Hollody K, Erhardy E, Molnar D. Relationship between sleep duration and childhood obesity: Systematic review including the potential underlying mechanisms. Nutr Metab Cardiovasc Dis. 2017;27:751-761.
  169. Council on Commnications and Media, Hill D, Ameenuddin N, Chassiokos Y, Cross C, Radesky J, Hitchinson J, Levine A, Boyd R, Mendelson T, Moreno M, Swanson W. Media use in school-aged children and adolescents. Pediatr. 2017;138: e20162592.
  170. Hernandez B, Gortmaker S, Colditz G, Peterson K, Laird N, Parra-Cabrera S. Association of obesity with physical activity, television programs and other forms of video viewing among children in Mexico city. Int J Obes.1999;23:845-854.
  171. Schweizer P, Lenz M, Kirschner H. Pathogenesis and symptomatology of cholelithiasis in childhood. A prospective study. Dig Surg. 2000;17:459-467.
  172. Kouvari M, Karipidou M, Tsiampalis T, Mamalaki E, Poulimeneas D, Bathrellou E, Panagiotakos D, Yannakoulia M. Digital health interventions for weight management in children and adolescents: Systematic review and meta-analysis. J Med Internet Res. 2022;24:e30675.
  173. Wang W, Cheng J, Song W, Shen Y. The effectiveness of wearable devices as physical activity interventions for preventing and treating obesity in children and adolescents: Systematic review and meta-analysis. JMIR MHealth UHealth. 2022;10:e32435.
  174. Srivastava G, Fox C, Kelly A, Jastreboff A, Browne A, Browne N, Pratt J, Bolling C, Michalsky M, Cook S, Lenders C, Apovian C. Clinical considerations regarding the use of obesity pharmacotherapy in adolescents with obesity. Obesity. 2019;27:190-204.
  175. Czepiel K, Perez N, Campoverde Reyes K, Sabharwal S, Stanford F. Pharmacotherapy for the treatment of overweight and obesity in children, adolescents, and young adults in a large health system in the US. Front Endocrinol. 2020;11:290.
  176. Singhal V, Sella A, Malhotra S. Pharmacotherapy for the treatment of overweight and obesity in children, adolescents, and young adults in a large health system in the US. Curr Opin Endocrinol Diabetes Obes.2020;28:55-63.
  177. Zwintscher N, Azarow K, Horton J, Newton C, Martin M. The increasing incidence of adolescent bariatric surgery. J Pediatr Surg. 2013;48:2401-2407.
  178. Inge T, Courcoulas A, Jenkins T, Mihalsky M, Helmrath M, Brandy M, Harmon C, Zeller M, Chen M, Xanthakos S, Horlick M, Buncher C, Teen-LABS Consrtium. Weight loss and health status 3 years after bariatric surgery in adolescents. N Eng J Med. 2016;374:113-123.
  179. Sanchez M, Panahi S, Tremblay A. Childhood obesity: a role for gut microbiota? Int J Environ Res Public Health. 2014;23:162-175.
  180. Hume M, Nicolucci A, Reimer R. Prebiotic supplementation improves appetite control in children with overweight and obesity: a randomized controlled trial. Am J Clin Nutr. 2017;105:790-799.
  181. Nicolucci A, Hume M, Martinez I, Mayengbam S, Walter J, Reimer R. Prebiotics reduce body and alter intestinal microbiota in children who are overweight or with obesity. Gastroenterol. 2017;153:711-722.
  182. Oosterhoff M, Joore M, Ferreira I. The effects of school-based lifestyle interventions on body mass index and blood pressure: a multivariate multilevel meta-analysis of randomized controlled trials. Obes Rev. 2016;17:1131-1153.
  183. Gortmaker S, Wang Y, Long M, Giles C, Ward Z, Barrett J, Kenney E, Sonneville K, Afzal A, Resch S, Cardock A. Three interventions that reduced chilhood obesity are projected to save more than they cost to implement. Health Aff. 2015;34:1932039.
  184. Mansfield J, Saviano D. Effect of school wellness policies and the Healthy, Hunger-Free Kids Act on food-consumption behaviors of students, 2006-2016: a systematic review. Nutr Rev. 2017;75:533-532.
  185. Vaudrin N, Lloyd K, Yedidia M, Todd M, Ohri-Vacjaspati P. Impact of the 2010 US Healthy, Hunger-Free Kids Act on school breakfast and lunch participation rates between 2008 and 2015. Am J Pub Heal. 2018;108:84-86.
  186. United States Department of Agriculture. Ag Secretary Perdue moves to make school meals great again. USDA Press Release. 2017:https://www.usda.gov/media/press-releases/2017/2005/2001/ag-secretary-perdue-moves-make-school-meals-great-again.
  187. Kenney E, Barrett J, Bleich S, Ward Z. Impact of The Healthy, Hunger-Free Kids Act on obesity trends. Health Aff. 2021;39:1122-1129.
  188. Burke M, Gleason S, Singh A, Wilkin M. Policy, systems, and environmental change strategies in the Supplemental Nutrition Assistance Program-Education (SNAP-Ed). J Nutr Educ Behav. 2022;54:320-326.
  189. Hudak K, Racine E. Do additional SNAP benefits matter for child weight?: Evidence from the 2009 benefit increase. Econ Hum Biol. 2021;41:100966.
  190. Fryar CD, Carroll MD, Afful J. Prevalence of overweight, obesity, and severe obesity among children and adolescents aged 2–19 years: United States, 1963–1965 through 2017–2018. NCHS Health E-Stats. Vol 20212020.
  191. Ogden CL, Martin CB, Freedman DS, Hales CM. Trends in Obesity Disparities During Childhood. Pediatrics.2022.
  192. Bray G, Siri-Tarino P. The role of macronutrient content in the diet for weight management. Endocinol Metab Clin North Am. 2016;45:581-604.
  193. Unick J, Beavers D, Bond D, Clark J, Jakicic J, Kitabchi A, Knowler W, Wadden T, Wagenknecht L, Wing R, Look AHEAD Research Group. The long-term effectiveness of a lifestyle intervention in severely obese individuals. Am J Med. 2013;126:236-242.
  194. Sacks F, Bray G, Carey V, Smith S, Ryan D, Anton S, McManus K, Champagne C, Bishop L, Laranjo N, Leborr M, Rood J, de Jonge L, Greenway F, Loria C, Obarzanek E, Williamson D. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. N Eng J Med. 2009;360:859-873.
  195. De Jonge L, Bray G, Smith S, Ryan D, de Souza R, Loria C, Champagne C, Ryan D, Williamson D, Sacks F. Effect of diet composition on energy expenditure during weight loss: the POUNDS LOST Study. Obes.2012;20:2384-2389.
  196. Skinner AC, Skelton JA. Prevalence and trends in obesity and severe obesity among children in the United States, 1999-2012. JAMA Pediatr. 2014;168(6):561-566.
  197. Qiao Y, Ma J, Wang Y, Li W, Katzmarzyk P, Chaput J-P, Fogelholm M, Johnson W, Kuriyan R, Kurpad A, Lambert E, Maher C, Maia J, Matsudo V, Olds T, Onywera V, Sarmiento O, Standage M, Tremblay M, Tudor-Locke C, Church T, Zhao P, Hu G, Group TIR. Birth weight and childhood obesity: a 12-country study. Int J Obes Suppl. 2015;5(S74-79).

 

Behavioral Approaches to Obesity Management

ABSTRACT

 

Obesity is extremely prevalent, affecting 42.5% of people in the United States alone. Advisory panels recommend a 5-10% reduction in initial weight for adults with obesity, or for those who are overweight, with a weight-related comorbidity. This loss can significantly reduce the risk of developing type 2 diabetes and improve other cardiovascular disease (CVD) risk factors, as seen in the Diabetes Prevention Program and Look AHEAD trials.  Greater reductions in weight produce even greater improvements in CVD risk factors. Weight loss can be achieved with a comprehensive lifestyle program that consists of dietary change, increased physical activity, and behavior therapy, provided in individual or group sessions. Behavioral treatment can be combined with diets of varying macronutrient composition as long as they induce a caloric deficit. Physical activity should be gradually increased over a period of 6 months, and although it is not effective as a stand-alone intervention for inducing a clinically meaningful mean weight loss, it is very important for facilitating weight maintenance and improving health outcomes. Principles of behavioral treatment include self-monitoring, stimulus control, and goal setting. Weight regain is common after an initial treatment period of 6-12 months, but frequent follow-up with an interventionist, which includes at least monthly counseling, can mitigate it. Treatments delivered by telephone, internet, or smartphone can be more easily disseminated to larger populations and can produce clinically meaningful mean weight losses if they include content similar to that of in-person lifestyle interventions and provide personalized feedback.

 

INTRODUCTION

 

Obesity, defined by a body mass index (BMI) ≥ 30 kg/m², is the most common nutritional disease in the United States, affecting 42.5% of adults (1) and 19% of children and adolescents (2). An additional 31% of American adults have a BMI in the overweight range of 25.0-29.9 kg/m². Obesity is associated with an increased risk of developing cardiovascular disease (3), hypertension, dyslipidemia, and type 2 diabetes mellitus (4), along with other clinical conditions including nonalcoholic fatty liver disease, gastroesophageal reflux, obstructive sleep apnea, and osteoarthritis (5-7). A weight loss of 5-10% of initial body weight improves these complications and has been recommended by expert panels sponsored by the World Health Organization (8), the National Institutes of Health (9), and several professional societies. Losses of this magnitude can be achieved with a high-intensity lifestyle intervention (also known as lifestyle modification or behavioral weight loss treatment), as described in the Guidelines for the Management of Overweight and Obesity in Adults (i.e., Obesity Guidelines) (10) developed by The American College of Cardiology, American Heart Association, and the Obesity Society.

 

Comprehensive lifestyle interventions include three key components: diet, physical activity, and behavior therapy. This chapter describes each intervention component and reviews the short-term and long-term effectiveness of this approach. Lifestyle interventions have traditionally been delivered in 30-90 minute, in-person, group or individual sessions by a trained interventionist (usually a registered dietitian, psychologist, exercise physiologist, or other health-care professional). Although this is by far the best-researched treatment modality, the past two decades have seen an exponential growth in digital and other remote treatment approaches, which are reviewed in the final section.

 

EFFICACY OF HIGH-INTENSITY, IN-PERSON LIFESTYLE INTERVENTION PROGRAMS

 

Interventions categorized as “high-intensity” by the Obesity Guidelines provide a minimum of 14 treatment sessions during the first 6 months (10). Maintenance sessions may be delivered at a reduced frequency thereafter. In trials conducted in academic medical centers, participants treated by a 1200-1500 kcal/day diet, combined with regular exercise and a comprehensive program of group or individual behavior modification, lose an average of 5-8% of initial weight in 6 months (9-11), and approximately 60-65% of patients lose ≥5% of their initial weight. The lifestyle programs provided in the Diabetes Prevention Program and the Look AHEAD study provide excellent examples of high-intensity interventions.

 

Diabetes Prevention Program

 

In the Diabetes Prevention Program (DPP), more than 3,200 participants with obesity or overweight and impaired glucose tolerance were randomly assigned to a placebo, metformin, or an intensive lifestyle intervention, with the goal of inducing a 7% weight loss in the latter group (12). Participants in the lifestyle intervention group were given 16 individual on-site counseling sessions with a registered dietitian in the first 24 weeks, followed by at least one contact every other month for the remainder of the study. They were prescribed a reduced-calorie, low-fat diet (1200-2000 kcal/day, depending on initial body weight), and 150 min/week of physical activity. After an average of 2.8 years, participants in the lifestyle intervention group lost a mean of 5.6 kg, compared to 0.1 and 2.1 kg in the placebo and metformin groups, respectively. The 5.6 kg weight loss translated to a 58% relative reduction in the risk of developing type 2 diabetes. Ten years after randomization, the lifestyle intervention group had regained most of their lost weight, but their incidence of type 2 diabetes remained 34% below that in the placebo group (13).

 

Look AHEAD (Action for Health in Diabetes) Study

 

The Look AHEAD study enrolled more than 5,100 individuals with overweight/obesity and type 2 diabetes mellitus, and participants were randomly assigned to a diabetes support and education (DSE) group or an intensive lifestyle intervention (ILI) group, with the aim of examining the long-term effects of a 7% weight loss on cardiovascular morbidity and mortality (14). Participants randomized to the DSE group received three group education sessions each year in the first 4 years, whereas participants in the ILI group received treatment similar to that in the DPP with some modification. During the first 6 months, ILI participants had 3 weekly group treatment sessions and one individual visit per month and replaced two meals per day with a liquid supplement (i.e., shake). They were instructed to consume 1200-1800 kcal/day (with calories adjusted based on initial weight). During months 7 to 12, ILI participants had two group sessions and one individual visit each month, and used meal replacements for one meal per day. For the next 3 years, participants were offered one individual on-site visit and one phone (or e-mail) contact per month.

 

After 1 year, ILI participants lost 8.6% of baseline weight, compared with 0.6% for the DSE group, and at year 4, mean weight losses were 4.7% versus 1.1%, respectively. These latter losses were maintained at 8 years, at which time patients in the ILI group lost 4.7% of initial weight, compared with 2.1% for DSE participants. The study was ended at a mean of 9.6 years of post-randomization follow-up because there were no differences in cardiovascular morbidity and mortality between groups. However, patients in ILI, compared to DSE, had significantly greater reductions in HbA1C, lost more weight, had larger improvements in cardiovascular disease risk factors (i.e., reductions in systolic and diastolic blood pressure and levels of triglycerides), and used fewer diabetes, hypertension, and lipid-lowering medications. Analyses showed that the greater the weight loss, the greater the improvements in those risk factors (Figure 1) (15).

Figure 1. Change in risk factors by weight loss categories for the Look AHEAD cohort. Data in all figures are presented as least square means and 95% CIs adjusted for clinical sites, age, sex, race/ethnicity, baseline weight, baseline measurement of the outcome variable, and treatment group assignment. Figure is reprinted with permission from reference (15).

 

Compared to DSE, additional benefits in the ILI group included greater reduction of depression symptoms and remission or reduced severity of obstructive sleep apnea. The Look AHEAD and DPP studies both demonstrate that weight loss and long-term benefits to health can be achieved through participation in a lifestyle modification program. However, a follow-up assessment of ILI and DSE participants 16 years post-randomization continued to reveal no significant differences in CVD morbidity and mortality between the two groups (16).

 

LIFESTYLE INTERVENTION COMPONENTS

 

Dietary Recommendations

 

The primary goal of the dietary prescription in a behavioral weight loss program is to induce a 500-750 kcal/day deficit (10,11).  For women, this involves consuming about 1200-1500 kcal/day, while for men the goal is about 1500-1800 kcal/day. Calorie targets also can be based on body weight, with 1200-1500 kcal/day recommended for people who weigh less than 250 lbs. at baseline and 1500-1800 kcal/day for those >250 lbs. (10,11). The ideal composition of dietary macronutrients for producing weight loss has been studied extensively, with options including low-glycemic index diets, Mediterranean-type diets, low-fat diets, and reduced-carbohydrate diets (17). A low glycemic index is based on eating a diet containing foods with a lower glycemic load, that are less likely to cause large increases in postprandial blood glucose levels (19,20). A Mediterranean diet focuses on consuming higher amounts of plant-based foods, including fruits, legumes, vegetables, monounsaturated fats such as olive oil, and fish; and reduced consumption of foods high in saturated fats, like red meat and butter (21). Low-fat diets provide 10% to 20% of calories from fat and recommend plant-based foods including whole-grains, fruits, and vegetables (22). A low carbohydrate diet approach, like an Atkins or “ketogenic” diet, is characterized by consuming as few as 20 g/day of carbohydrates, and focusing on foods that are higher in protein and fat (23).

 

The outcomes of comparative studies of these different types of diets have consistently concluded that adequate weight loss depends less on the macronutrient content of the diet and more on the caloric deficit (17).  The POUNDS LOST trial supported this conclusion in a large, 2-year study that randomized patients to one of four diets with different macronutrient compositions, varying in proportions of fat, protein, and carbohydrate content (fat/protein/carbohydrate content: 20/15/65%; 20/25/55%; 40/15/45%; and 40/25/35%, respectively) (24). The study showed no difference in the amount of weight lost among the diet groups, all of which were designed to produce an energy deficit of approximately 750 kcal per day.  Several other studies have also found that different dietary approaches produce weight losses that are comparable, provided there is a sufficient reduction in calories (25,26) (Figure 2). The use of portion-controlled diets have been shown to facilitate greater weight losses than diets of conventional foods, but this is primarily due to improved adherence to calorie goals and not to their macronutrient profile (27).

Figure 2. Change in body weight for participants in low-fat and low-carbohydrate diet groups after 24 months, based on random-effects linear model. Figure is reprinted with permission from reference (25).

 

Because it appears that caloric restriction contributes to weight loss more than the macronutrient composition of the diet, diets should be chosen based on patients’ personal preferences and by the presence of comorbid conditions. For example, Fabricatore et al. (28) demonstrated that a low-glycemic index diet produced greater improvements in HbA1cin patients with overweight and type 2 diabetes than did a traditional low-fat diet, even though the two diets produced comparable weight losses. Low-fat diets appear to be associated with greater reductions in low-density lipoprotein cholesterol (24,25,29), compared to low-carbohydrate diets. The latter diets, by contrast, are associated with greater reductions in triglycerides (26,29-33), increases in high-density lipoprotein cholesterol (25,26,29-33), and improvements in HbA1C in patients with type 2 diabetes (33). Table 1 summarizes the results of selected randomized trials that examined the effects of macronutrient composition on changes in weight and health outcomes.

 

Table 1. Weight Loss Results from Randomized Trials that Compared diets with Varying Macronutrient Compositions

Study

N

No. Lifestyle Sessions Provided

Dietary Intervention

Weight Change

 Month

 Comment/ Other Results

Dansinger et al(26)

   160 (51% F)

   58%    completed

 

 

4

   Atkins (low-carb)

   Zone (even distribution)

   Weight Watchers (points based)

   Ornish (low-fat)

-2.1 kg a

-3.2 kg a

 

-3.0 kg a

 

-3.3 kg a

 

12

     All participants had hypertension, dyslipidemia, and/or fasting hyperglycemia.

     Weight loss was associated with level of adherence.

     Each diet decreased LDL/HDL ratio.

 

N  No significant changes in blood pressure or blood glucose at 12 months in either group.

Das et al(34)*

34 (% F  unknown)

85% completed

52

    Low-glycemic load

    High-glycemic load

-7.8% a

 

-8.0% a

12

     Triglycerides, total, HDL, and LDL cholesterol decreased in both groups.

Fabricatore et al(28)

  79 (80% F)

63% completed

30

Low-glycemic load

     Low-fat

-4.5% a

 

    -6.4% a

9

All participants had type 2 diabetes.

     Larger reductions in HbA1cin the low-glycemic load group.

Foster et al(29)

63 (68% F)

59% completed

3

     Low-carbohydrate (high protein, high fat)

    Conventional (high-carbohydrate, low-fat)

-4.4% a

 

 

 

-2.5% a

12

     HDL cholesterol increased more and triglycerides decreased more in the low-carbohydrate group.

    Greater reductions in LDL and total cholesterol in the low-fat group at 3 months.

Foster et al(25)

307 (68% F)

63% completed

38

   Low-carbohydrate

   Low-fat

-6.3 kg a

 

-7.4 kg a

24

     HDL cholesterol increased more and triglycerides were lower only in the low-carbohydrate group.

     Greater decrease in LDL at 3 and 6 months in the low-fat group.

Gardner et al(30)

 

311 (100% F)

80% completed

 

 

8

    Atkins (low-carbohydrate)

    Zone (even distribution)

    LEARN (calorie-restricted)

    Ornish (low-fat)

-4.7 kg a

 

-1.6 kg b

 

-2.2 kg ab

 

-2.6 kg ab

12

 HDL cholesterol increased more in Atkins than Ornish group. Triglyceride levels decreased more in Atkins than Zone group.

     No differences in insulin or blood glucose between groups.

     Systolic blood pressure decreased more in Atkins than in all other groups.  Diastolic blood pressure decreased more in Atkins group than in Ornish group.

Sacks et al(24)

   811 (64% F)

7 9.5%   completed

 

66

    Low-fat, average protein (highest carbohydrate)

   Low-fat, high-protein

   High-fat, average-protein

    High-fat, high-protein (lowest carbohydrate)

-3.0 kg a

 

 

-3.8 kg a

 

-3.2 kg a

 

-3.4 kg a

24

     LDL cholesterol decreased more in lowest fat than in highest fat group. HDL cholesterol increased more with lowest carbohydrate than with the highest carbohydrate diet. All diets decreased triglyceride levels similarly.

     All diets, except the highest carbohydrate, decreased fasting insulin (greater decrease in the high protein vs average protein diets).

Shai et al(32)

 

   322 (14% F)

8 4.6%   completed

 

 

24

    Low-fat

    Mediterranean (moderate fat, restricted calorie with fat predominantly from olive oil and nuts)

    Low-carbohydrate

-2.9 kg a

-4.4 kg b

 

 

 

 

 

 

-4.7 kg b

 

 

24

     No significant change in LDL cholesterol in any group.

     HDL cholesterol increased in all groups, significantly more in the low-carbohydrate than low-fat group.

     Triglyceride levels decreased more in the low-carbohydrate than in the low-fat group.

     In diabetic participants, only the Mediterranean diet group had a decrease in fasting glucose. 

     Insulin decreased in all groups, for both diabetic and non-diabetic participants.

     All groups had a significant decrease in blood pressure.

     Adiponectin levels increased, and leptin levels decreased, in all groups. 

Stern et al(33)

 

132 (17% F)

66% completed

15

    Low-carbohydrate

    Conventional (low-fat)

-5.1 kg a

 

-3.1 kg a

12

     Triglyceride levels decreased more in the low-carbohydrate group than in the low-fat group.

     HDL cholesterol decreased less in the low-carbohydrate group than in the low-fat group.

     Changes in total and LDL cholesterol were not significantly different between groups.

Yancy et al(35)

120 (76% F)

66% completed

9

 

 

 

 

    Low-fat diet

    Low-carbohydrate, ketogenic diet with nutritional supplements

-6.7% a

-12.9% b

6

     All participants were hyperlipidemic.

     Triglycerides decreased more and HDL cholesterol increased more in low-carbohydrate group.

Table is reprinted with permission from reference (18).

All studies were analyzed by use of an intention-to-treat population, with the exception as indicated by an asterisk (*).

Different letters (in superscript) indicate statistically significant differences in weight loss between groups.

F indicates female; LDL, low-density lipoprotein; HDL, high-density lipoprotein; VLDL, very low-density lipoprotein; HbA1c, hemoglobin A1c; MR, meal replacements; CVD, cardiovascular disease.

*A completer’s population was examined. †Results reported as “greater,” “larger,” “increased more,” etc. represent statistically significant differences between treatment conditions.

 

Physical Activity Recommendations

 

Physical activity is an important component of a comprehensive lifestyle intervention, in which participants are typically instructed to increase their physical activity gradually to approximately 150-180 min/week over the first 6 months. This goal can be achieved by engaging in moderate physical activity (e.g., brisk walking) for 30 minutes on 5 days per week (10,11,13). Physical activity can be increased by incorporating short bouts of lifestyle activity into individuals’ daily routines, such as increasing the amount of daily walking or using the stairs when possible, or by longer bouts of structured physical activity (e.g., at the gym). Individuals should be encouraged to engage in physical activities that they enjoy rather than be prescribed a particular activity. The recommended physical activity levels for facilitating long-term weight management are higher (225-300 min/week) than those for losing weight (36).  The effects of physical activity on weight loss, the maintenance of weight loss, and CVD risk factors have been investigated extensively.

 

PHYSICAL ACTIVITY AND WEIGHT LOSS

 

Physical activity has a modest impact on weight loss when compared with the effect of caloric restriction (36). This was demonstrated in a 12-week study in which participants achieved losses of 0.3-0.6% (male vs female) of initial weight from physical activity alone, compared to 5.5-8.4% (female vs male) and 7.5-11.4% (female vs male) losses for participants who reduced their calorie intake and those who changed both diet and physical activity, respectively (37). The exercise performed in this study consisted of 30 min/day of moderate activity on 5 days per week. Similarly, Wing et al (38) reported weight losses of 2.1, 9.1, and 10.3 kg after 6 months in participants assigned to physical activity alone, diet alone, and diet plus physical activity groups, respectively, all of whom were provided behavioral intervention.

 

PHYSICAL ACTIVITY AND WEIGHT MAINTENANCE

 

Although exercise has a limited impact on weight loss during the initial phase of treatment, it plays an important role in weight loss maintenance. Several studies have shown that the more physical activity the patient engages in, the better the maintenance of lost weight (39,40). Jakicic et al (40), in a secondary analysis of a randomized controlled trial, demonstrated that women who exercised more than 200 min/week maintained greater weight losses than those who exercised 150-199 min/week or <150 min/week. Data from the National Weight Control Registry have also provided evidence that high levels of physical activity are characteristic of individuals who report long-term, sustained weight loss (41). The Registry follows patients who have lost a minimum of 13.6 kg (i.e., 30 lb.) in six months and maintained this loss for at least 1 year.  Of these successful weight loss maintainers, 91% reported that they were exercising consistently, with women expending 2,545 kcal/week and men 3,293 kcal/week (42). Based on these findings and other evidence, the current recommendation by the American College of Sports Medicine is that, for weight maintenance, individuals should exercise at a minimum level equivalent to an hour of brisk walking per day (36). 

 

PHYSICAL ACTIVVITY AND CARDIOVASCULAR HEALTH

 

Physical activity also is crucial for improving cardiovascular health for both individuals with obesity and those of average-weight. Even in the absence of significant weight loss, regular bouts of aerobic activity have been found to reduce blood pressure (43), lipids (44), and visceral fat (45), the latter of which is associated with improved glucose tolerance and insulin sensitivity in non-diabetic individuals and glycemic control in patients with type 2 diabetes (46,47). Several authors have evaluated the independent effects of cardiorespiratory fitness and adiposity on subsequent CVD mortality and have suggested that high levels of cardiorespiratory fitness significantly decrease the CVD mortality risk in individuals with overweight and obesity, regardless of adiposity. Barry et al (48) performed a meta-analysis of 10 studies and concluded that, compared to individuals who were fit and had normal weight, unfit individuals had twice the risk of all-cause mortality regardless of their BMI, whereas individuals who were fit and had obesity had similar mortality risks as normal-weight, fit individuals. Similarly, in a longitudinal study of 25,000 men, Lee et al (49) found that those who were lean but unfit had double the mortality rate of those who were fit and lean. These findings indicate that all individuals should increase their physical activity to improve their health, regardless of its impact on body weight.

 

Principles of Behavior Therapy

 

The third component of lifestyle intervention is behavior therapy, which refers to a set of principles and techniques used to help patients adopt dietary and activity recommendations. Behavioral principles were first applied to the treatment of obesity in the 1960’s and early 1970’s and, since then, have been developed into a program of behavioral and cognitive strategies (11). The core components of behavior therapy include goal setting, self-monitoring, stimulus control, and problem solving.

 

GOAL SETTING

 

In behavioral weight loss treatment, goal setting refers to setting specific targets for making changes to the patient’s calorie intake, physical activity, and eating and exercise habits (50, 51).  Goals need to be objective and easily measurable in order to facilitate patients’ assessment of their progress. Patients are encouraged to have a target range for their total daily caloric intake (or other dietary goals), a daily or weekly exercise goal in minutes, and short- and long-term weight loss goals. Other behavioral goals are introduced as treatment proceeds. Patients should set goals that facilitate their losing about 0.5-1.0 kg per week, for a total loss of 5-10 percent of initial body weight at the end of the weight loss phase (at about 6 months). These goals should be trackable and should specify when and how the goal will be accomplished. During a typical treatment session, the lifestyle interventionist reviews each patient’s progress in achieving goals from the previous session and helps the patient set new goals. If the goals from a previous session are not met, the interventionist assists individuals with identifying and reducing barriers to goal achievement or with modifying their goals accordingly. In group programs, this information is often shared with the entire group to further increase accountability and support problem-solving.

 

SELF-MONITORING  

 

Monitoring target behaviors in a systematic way is a crucial aspect of the behavioral approach to weight loss. Self-monitoring provides instant feedback about the effectiveness of behavior change efforts. It can answer the most important question about behaviors: are they getting better, staying the same, or getting worse? Daily records also function to increase patients’ awareness of target behaviors and their effect on weight change. Self-monitoring is strongly linked to success in weight loss.  Individuals who regularly monitor their weight, activity levels, and eating patterns usually achieve the largest weight losses (52,53). 

 

Patients are encouraged to record all foods and beverages consumed and their calorie content (or an alternative dietary target) to determine if they have met their dietary goals. A thorough self-monitoring report might also include the individual’s feelings that day, particularly those that were associated with excess or unplanned eating, or other individually-identified triggers for overeating. Tracking the minutes and type of physical activity or pedometer step counts can be used to monitor improvements in the patient’s activity level. Patients also should be instructed to weigh themselves regularly at home, at least once per week, and to keep a record of their weekly weights.

 

Although some patients prefer traditional paper records, the majority now track these targets using smartphone applications (apps) and other digital devices such as wearable physical activity trackers and “smart” scales that automatically record body weight. Although these digital tools increase self-monitoring consistency and are preferred by most patients (54), they have not been found to enhance weight loss when compared to traditional tracking methods (52, 55, 56). Novel tracking tools such as digital food photography and bite counting devices may further reduce the burden of active recording, but some studies have suggested that these methods are less strongly correlated with weight loss and may produce smaller mean losses than active recording methods (57).

 

In lifestyle intervention programs, patients review their self-monitoring records with an interventionist who helps them to assess their progress, set goals, and problem solve barriers to goal adherence. Individuals often underestimate calorie intake and overestimate physical activity (58), and interventionists can help patients who report meeting their calorie and activity goals but do not lose weight to identify additional sources of caloric intake. These may come from underestimates of portion sizes or hidden sources of fat and/or sugar intake. Interventionists also can help patients address barriers to effective self-monitoring, or set more flexible self-monitoring goals (e.g., record on fewer days per week), as appropriate.

 

STIMULUS CONTROL

 

The goal of stimulus control is to alter external and internal cues that influence eating and exercise behaviors (11, 50, 51). In classical conditioning, cues develop when two stimuli (e.g., objects, activities) are repeatedly experienced together, which creates an association between the two. The appearance of one stimulus can invoke the other stimulus. Food cues are cues that cause an individual to think about eating or about specific foods. These may include external cues, such as the sight or smell of food, or an activity that is frequently engaged in while eating, such as watching television. Internal food cues include both physical sensations and thoughts or emotions that the person has come to associate with eating. Similarly, activity cues include internal and external experiences that the person has come to associate either with being active (e.g., the sight of sneakers by the door) or being inactive (e.g., the couch). 

 

Patients learn to reduce negative food and activity cues -- either by avoiding problem cues or by creating new habits in response to those cues -- and to enhance cues for desired behaviors. Examples of cue reduction include avoiding places that sell or serve high-calorie foods, staying away from all-you-can-eat buffets, and keeping any high-calorie foods that are associated with overeating out of the house. The patient can instead be encouraged to buy single portions of these foods on planned occasions. For cues that cannot be avoided, the patient may be encouraged to identify an appropriate alternative behavior, such as taking a 5-minute break instead of snacking when bored at work. To increase cues for healthy eating, patients can be taught to improve the visibility and availability of healthy, low-calorie foods in their home or workplace, such as by storing these foods at eye-level.  They can also add cues that promote physical activity, such as arranging to walk at a certain time every day with a partner or leaving their gym bag in their car so that it is the first thing that they see when they leave work. By making these changes, patients can ensure that their work and home environments facilitate (rather than interfere with) weight loss.

 

COGNITIVE STRATEGIES

 

Strategies from cognitive-behavioral therapy (CBT) have been incorporated into many lifestyle interventions. CBT focuses on identifying, testing, and correcting maladaptive thoughts in order to change emotions or behavior. For example, thoughts like “I’ll never reach my weight loss goal; I might as well eat whatever I want.”) can reduce the likelihood that a patient will adhere to their dietary goals. Patients are taught to create a rational response to these negative thoughts, such as by noting that “My weight loss may be slower than I would like, but continuing to make healthy choices gives me the best chance of long-term success,” or by highlighting some of the benefits that they experience when they make healthier eating choices (50, 51).

 

Some lifestyle programs also have incorporated strategies from motivational interviewing that are designed to help patients resolve ambivalence about the costs and benefits of behavior change, identify reasons for change, and improve self-efficacy. More recently, alternative cognitive strategies derived from mindfulness and acceptance-based psychological treatments have been incorporated into weight loss interventions. These treatments promote non-judgmental, present-moment awareness and willingness to experience discomfort in order to pursue long-term goals rather than cognitive change. Thus far, programs that place a significant emphasis on any of these cognitive techniques have not consistently enhanced weight loss when compared to standard lifestyle interventions, and those shown to be superior have only increased weight loss by 1-2 kg (59). However, because fewer studies have incorporated these techniques into comprehensive, high-intensity lifestyle interventions, they remain promising targets for future research.

 

STRUCTURE OF IN-PERSON BEHAVIORAL TREATMENT: SHORT- AND LONG-TERM

 

The lifestyle interventions provided in studies like the DPP and Look AHEAD followed a structured curriculum that gradually introduced different behavior change skills. Detailed treatment descriptions can be obtained from the intervention manuals for these two studies (50, 51) or an adaptation of the DPP protocol provided by Wadden, Tsai, and Tronieri for in-person delivery in primary care settings (60). Behavioral weight loss interventions are most commonly delivered in group sessions, which have been found to be as effective as individual counseling for weight loss in several studies (61,62). It may be that any weight loss benefit of receiving personalized support with individual counseling is roughly equivalent to the benefits of a greater degree of social support, empathic understanding, and healthy competition among group members. However, group treatment is more cost effective than individual counseling.

 

Frequency and duration of contact during the weight loss period are additional predictors of weight loss success (10,61). This benefit is apparent in trials comparing high-intensity lifestyle intervention programs to programs that provided identical dietary and physical activity recommendations with a lower session frequency, as well as in systematic reviews and meta-analyses of the efficacy of lifestyle interventions. For example, in a study by Perri and colleagues (63) that compared three different visit schedules to a control condition, the group that received 8 treatment sessions in the first 6 months had a weight loss of 3.5 kg at month 24 that did not differ significantly from the 2.9 kg loss of the control group, whereas patients who received 16 sessions had a loss of 6.7 kg that was superior to both groups. Of note, the group that received 24 sessions in the first 6 month did not differ in weight loss from the 16-session group at any time, suggesting that there may not be a benefit of further increasing visit intensity (while increasing costs). In 2012, the United States Preventative Services Task Force recommended that weight loss programs include at least 12-26 intervention sessions per year for optimal weight loss (64). This recommendation was based on their systematic review, which reported weight losses of 4 to 7 kg for programs with that level of intensity compared to 1.5 to 4 kg in programs offering fewer than 12 sessions (61). These findings were consistent with the Obesity Guideline’s conclusion that programs that provided at least 14 sessions in the first 6 months produce a weight loss of 5 to 8 kg, those that provide 6-13 sessions (1-2 sessions per month) produce a 2 to 4 kg loss, and those that provide less than monthly sessions induce minimal weight loss (10).

 

For weight loss maintenance, frequent, long-term contact with an interventionist is the most successful method for preventing weight regain. Weight loss maintenance sessions are important for providing individuals with the support and motivation needed to continue with the behavior changes they have made, such as engaging in physical activity, eating a low-calorie diet, and self-monitoring. Wing et al (65) demonstrated that monthly in-person sessions were more effective in preventing weight regain over 18 months of intervention than was an education-control group or an internet-based intervention. Participants in the three groups regained an average of 2.5, 4.9, and 4.7 kg, respectively, after an initial weight loss of 19 kg.

 

Table 2. Recommended Components of a High-Intensity Comprehensive Lifestyle Intervention to Achieve and Maintain a 5-to-10% Reduction in Body Weight.*

Component

Weight Loss

Weight-loss Maintenance

Counseling

≥14 in-person counseling sessions (individual or group) with a trained interventionist during a 6-mo period; recommendations for similarly structured, comprehensive Web-based interventions, as well as evidence-based commercial programs

Monthly or more frequent in-person or telephone sessions for ≥1 yr. with a trained interventionist

Diet

Low-calorie diet (typically 1200–1500 kcal per day for women and 1500–1800 kcal per day for men), with macronutrient composition based on patient’s preferences and health status

Reduced-calorie diet, consistent with reduced body weight, with macronutrient composition based on patient’s preferences and health status

Physical activity

≥150 min per week of aerobic activity (e.g., brisk walking)

200–300 min per week of aerobic activity (e.g., brisk walking)

Behavioral therapy

Daily monitoring of food intake and physical activity, facilitated by paper diaries or smart-phone applications; weekly monitoring of weight; structured curriculum of behavioral change (e.g., DPP), including goal setting, problem solving, and stimulus control; regular feedback and support from a trained interventionist

Occasional or frequent monitoring of food intake and physical activity, as needed; weekly-to-daily monitoring of weight; curriculum of behavioral change, including problem solving, cognitive restructuring, and relapse prevention; regular feedback from a trained interventionist

*Data are from the Guidelines (2013) for the Management of Overweight and Obesity in Adults, reported by Jensen et al. (10). The guidelines concluded that a variety of dietary approaches that differ widely in macronutrient composition, including ad libitum approaches (in which a lower calorie intake is achieved by restriction or elimination of particular food groups or by the provision of prescribed foods), can lead to weight loss provided they induce an adequate energy deficit. The guidelines recommended that practitioners, in selecting a weight-loss diet, consider its potential contribution to the management of obesity-related coexisting disorders (e.g., type 2 diabetes and hypertension). The guidelines did not address the possible benefits of strength training, in addition to aerobic activity. DPP denotes Diabetes Prevention Program. Table is reprinted with permission from reference (66)

 

REMOTELY-DELIVERED LIFESTYLE MODIFICATION INTERVENTIONS

 

In-person interventions can be costly because they require adequate facilities for hosting the intervention, staff for checking in patients, and the time of trained providers to deliver the intervention. Travel time also can represent a cost and inconvenience for patients, and many individuals, particularly those in rural and economically disadvantaged urban areas, do not have adequate access to evidence-based care. Over the past two decades, a growing body of research has investigated the use of telephone, computer, and smartphone-based methods for delivering lifestyle interventions to patients. Larger numbers of individuals can be reached with these methods at a cost that is significantly less than that of in-person interventions, particularly if little to no provider input is required. The COVID-19 pandemic further highlighted the need to identify effective ways of delivering lifestyle interventions remotely, as in-person treatment programs were either suspended or quickly migrated to phone calls or videoconferencing platforms due to stay-at-home orders and social distancing policies.

 

Telehealth Delivery

 

Remote interventions delivered live by a provider via telephone or videoconferencing, often referred to as telehealth, produce weight loss outcomes that are most consistent with those of in-person interventions. This delivery method improves treatment access and reduces travel time and cost for participants, but it has minimal impact on provider time and training costs. Several large trials have compared individual or group telephone calls to in-person treatment delivery. For example, Donnelly et al (67) achieved median 26-week weight losses of 13.0% with group conference calls which did not differ from the 12.7% loss of patients who attended on-site groups (both also received a 12-week 1200-1500 kcal/day portion-controlled diet). Similarly, Appel et al (68) showed comparable weight losses at 24 months for participants who received telephone-delivered sessions compared to those that received in-person visits (4.6 kg and 5.1 kg, respectively). Telephone-based interventions also have shown to be effective for weight maintenance and appear to attenuate weight regain to a similar degree as ongoing in-person sessions (62,65,69).

 

In the past several years, videoconferencing platforms have become more widely accessible. These platforms provide the capability for remotely delivered face-to-face interactions, which allow for visual demonstrations and may enhance feelings of connection with the interventionist and/or group (70). This delivery format has yet to be compared to in-person intervention in a randomized trial; however, pilot and short-term studies report weight losses that are 3 to 8 kg larger than control or minimal intervention conditions (71-73), which suggests that videoconferencing also may produce weight losses that are similar in magnitude to those of in-person interventions.

 

Digital Delivery via the Internet or Smartphone

 

Digitally-delivered programs in which standardized intervention content is delivered via digitally-accessible articles, messages (e.g., e-mail or SMS), or pre-recorded videos further reduce costs and interventionist burden when compared to live interventionist delivery either in person or through telehealth. Some of the earliest interventions with digital session content were developed for delivery via the internet. In an early study, Tate el al. (74) demonstrated that an Internet-based behavioral approach consisting of email-based lessons, online self-monitoring of diet and physical activity, and e-mail feedback from an interventionist produced greater 6-month weight losses of 4.1 kg compared to the 1.6 kg loss achieved by participants who received an educational program (i.e., Internet resources with no specific instruction in changing eating and activity habits). As technology has evolved, digital programs have more typically been developed for mobile delivery via smartphone apps or in formats accessible via either the computer or smartphone. Intervention delivery via text message also has been evaluated, but typically produces small mean weight losses (1-2 kg) when used as a stand-alone intervention format (75).

 

Relatively few studies have directly compared the efficacy of digitally-delivered to in-person treatment. Harvey-Berino and colleagues (76) compared the same 24-session group lifestyle intervention delivered weekly: 1) in-person; 2) by internet (including online content, self-monitoring tools, and weekly chat groups); or 3) in a hybrid format (the internet program with monthly in-person meetings). Weight losses were 8.0, 5.5, and 6.0 kg, respectively, with in-person treatment superior to the other two groups. These findings, along with the results of multiple systematic reviews, suggest that the strongest digitally-delivered interventions produce short-term losses that are at least 20-35% smaller than those achieved with in-person counseling (77, 78). Such interventions are valuable given their wide reach and low cost, and the difference between the results of these digital interventions and in-person programs is likely to wane over time with regain. However, the average effect of digitally-delivered interventions is small (1-3 kg), highlighting the importance of identifying features associated with effective interventions (77, 78).

 

The provision of tailored feedback is by far the most commonly identified characteristic that differentiates effective from less effective digital interventions (77, 78). In earlier digital trials, feedback was provided directly by an interventionist. Increasingly, digital programs provide fully-automated, personalized feedback, generated from algorithms that analyze participants’ self-monitoring data. This tailored automated feedback appears to produce weight losses that are similar in magnitude to programs with interventionist-delivered feedback (e.g., 79, 80). A 2015 study by Martin and colleagues (81) evaluated a combined approach providing participants with highly personalized automated and weekly interventionist-initiated feedback (by phone, email, or app), in addition to app-based lesson materials, in an effort to maximize weight loss. Participants were given activity monitors and smart scales, and the app delivered automated graphic feedback comparing their physical activity and weight loss to expected targets (calculated based on their starting weight and calorie prescription). If participants’ weight losses fell outside of the expected range, they were prompted to select a behavioral strategy (e.g., use portion-controlled foods) to get back on track. In this 12-week pilot study, intervention participants lost 9.4 kg compared to 0.6 kg in the control group (81).Additional research is investigating the potential for just-in-time adaptive interventions (JITAIs) that use machine learning to identify individual risk factors for behavioral lapses and provide tailored feedback and intervention strategies at the times when an individual is most at risk. An initial evaluation of a JITAI intervention that was designed to promote dietary adherence by predicting dietary lapses produced a 10-week weight loss of 4.7% when combined with an app-based commercial weight loss program (82).

 

User engagement has been found to correlate with weight loss in several digital trials, making it another potential target for improving the efficacy of digital interventions. One approach for enhancing engagement is to increase the interactive quality of the digital program. Thomas, Leahey, and Wing (83) tested the efficacy of a 12-week online program that provided interactive lessons that incorporated videos, quizzes, and practical exercises. The program also provided self-monitoring tools and fully automated weekly feedback based on participants’ recorded data. At 6 months, intervention participants lost 5.4 kg, compared to 1.3 kg for control participants who received static lessons about the benefits of weight loss (without behavioral strategies). Other efforts to increase interactive engagement have incorporated lifestyle programs into social media platforms, virtual reality, or online games, and several of these interventions also have produced mean weight losses of 4-5 kg (84). A recent study by Vaz and colleagues (85) combined several of these techniques into a smartphone app that provided automated feedback on weight and physical activity recorded via smart scale and activity tracker, respectively; text- and app-initiated engagement prompts from an interventionist; social networking and sharing of food and exercise data; and peer competitions based on dietary and physical activity adherence. The app produced a mean weight loss of 7.2 kg at 6 months, which was 4.2 kg larger than a control group that received two weight management visits.  

 

Unfortunately, the efficacy of most commercially-available weight loss apps has not been systematically evaluated. A majority of these apps include only a small percentage of the behavioral strategies typically featured in intensive lifestyle programs (86), and most do not provide tailored feedback. Such programs are not likely to induce a clinically meaningful weight loss for most individuals. For example, the highly popular app, MyFitnessPal, which helps users set a calorie goal and track food intake, produced a mean loss of only 0.03 kg in 6 months in primary care patients, compared with a gain of 0.3 kg in controls (87). The frequency of logins declined sharply after the first month (to close to 0), which again underscores the problem of maintaining user engagement with digitally-delivered interventions that do not provide interactive content. Results have been more promising for online and app-based commercial programs that do provide comprehensive intervention content. Weight losses of 4-5 kg were achieved at 3-6 months in a randomized trial evaluating an online commercial program that provided nine weekly e-mail delivered video lessons, online content (e.g., recipes), self-monitoring tools, personalized summaries of self-monitoring data, and the option to chat with an interventionist online (88). Overall, these findings suggest that providers can support their patients’ weight loss by helping them to identify digital programs that offer comprehensive session content and personalized feedback.

 

CONCLUSION

 

There is clear evidence that intensive lifestyle interventions are effective in helping patients with obesity to lose 5-10% of initial body weight, a loss that is associated with improvements in CVD risk factors and other obesity-related comorbidities. Lifestyle approaches emphasize prescriptions for dietary intake, increased physical activity, and behavioral skills such as self-monitoring. Traditionally, these interventions have been delivered in-person by a trained interventionist, which limits their potential dissemination. It is also possible to achieve a clinically meaningful weight loss with digitally-delivered programs that include little to no contact from an interventionist, provided the intervention provides comprehensive session content, tailored feedback, and features that promote user engagement.

 

One of the most challenging aspects of behavioral weight control is keeping off lost weight. Several strategies can facilitate this goal, including maintaining patient-provider contact beyond the initial weight loss intervention, either in-person or remotely, and prescribing high levels of physical activity after weight is lost in the first 6 months. In addition, the more that patients practice the skills used by participants in the National Weight Control Registry, the more likely they will be to maintain their weight loss.

 

REFERENCES

 

  1. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of obesity and severe obesity among adults: United States, 2017–2018. NCHS Data Brief, no 360. Hyattsville, MD: National Center for Health Statistics. 2020.
  2. Fryar CD, Carroll MD, Afful J. Prevalence of overweight, obesity, and severe obesity among children and adolescents aged 2–19 years: United States, 1963–1965 through 2017–2018. NCHS Health E-Stats. 2020.
  3. Manson JE, Colditz GA, Stampfer MJ, Willett WC, Rosner B, Monson RR, Speizer FE, Hennekens CH. A prospective study of obesity and risk of coronary heart disease in women. The New England Journal of Medicine. 1990; 322:882-889.
  4. Colditz GA, Willett WC, Stampfer MJ, Manson JE, Hennekens CH, Arky RA, Speizer FE. Weight as a risk factor for clinical diabetes in women. American Journal of Epidemiology. 1990; 132:501-513.
  5. Carman WJ, Sowers M, Hawthorne VM, Weissfeld LA. Obesity as a risk factor for osteoarthritis of the hand and wrist: a prospective study. American Journal of Eepidemiology. 1994; 139:119-129.
  6. Must A, Spadano J, Coakley EH, Field AE, Colditz G, Dietz WH. The disease burden associated with overweight and obesity. JAMA. 1999; 282:1523-1529.
  7. Mokdad AH, Ford ES, Bowman BA, Dietz WH, Vinicor F, Bales VS, Marks JS. Prevalence of obesity, diabetes, and obesity-related health risk factors, 2001. Jama 2003; 289:76-79
  8. Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organization Technical Report Series 2000; 894:i-xii, 1-253
  9. Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults--The Evidence Report. National Institutes of Health. Obesity Research. 1998; 6 Suppl 2:51s-209s.
  10. Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA, Hu FB, Hubbard VS, Jakicic JM, Kushner RF, Loria CM, Millen BE, Nonas CA, Pi-Sunyer FX, Stevens J, Stevens VJ, Wadden TA, Wolfe BM, Yanovski SZ, Jordan HS, Kendall KA, Lux LJ, Mentor-Marcel R, Morgan LC, Trisolini MG, Wnek J, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Curtis LH, DeMets D, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Smith SC, Jr., Tomaselli GF. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014; 129:S102-138.
  11. Wadden TA, Tronieri JS, Butryn ML. Lifestyle modification approaches for the treatment of obesity in adults. American Psychologist. 2020;75(2):235.
  12. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. The New England Journal of Medicine.2002; 346:393-403.
  13. Knowler WC, Fowler SE, Hamman RF, Christophi CA, Hoffman HJ, Brenneman AT, Brown-Friday JO, Goldberg R, Venditti E, Nathan DM. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet (London, England). 2009; 374:1677-1686.
  14. Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. The New England Journal of Medicine. 2013; 369:145-154.
  15. Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni AG, Hill JO, Brancati FL, Peters A, Wagenknecht L. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care. 2011; 34:1481-1486.
  16. Look AHEAD Research Group. Effects of Intensive Lifestyle Intervention on All-Cause Mortality in Older Adults With Type 2 Diabetes and Overweight/Obesity: Results From the Look AHEAD Study. Diabetes Care. 2022;45(5):1252-9.
  17. Chao AM, Quigley KM, Wadden TA. Dietary interventions for obesity: clinical and mechanistic findings. The Journal of Clinical Investigation. 2021;131(1).
  18. Wadden TA, Webb VL, Moran CH, Bailer BA. Lifestyle modification for obesity: new developments in diet, physical activity, and behavior therapy. Circulation. 2012; 125:1157-1170.
  19. Thomas D, Elliott EJ. Low glycaemic index, or low glycaemic load, diets for diabetes mellitus. The Cochrane Database of Systematic Reviews. 2009:Cd006296.
  20. Jenkins DJ, Wolever TM, Taylor RH, Barker H, Fielden H, Baldwin JM, Bowling AC, Newman HC, Jenkins AL, Goff DV. Glycemic index of foods: A physiological basis for carbohydrate exchange. The American Journal of Clinical Nutrition. 1981; 34:362-366.
  21. Willett W, Skerrett PJ, Giovannucci EL, Callahan M. Eat, drink, and be healthy the Harvard Medical School guide to healthy eating. New York: Simon & Schuster Source.
  22. Krauss RM, Eckel RH, Howard B, Appel LJ, Daniels SR, Deckelbaum RJ, Erdman JW, Jr., Kris-Etherton P, Goldberg IJ, Kotchen TA, Lichtenstein AH, Mitch WE, Mullis R, Robinson K, Wylie-Rosett J, St Jeor S, Suttie J, Tribble DL, Bazzarre TL. AHA Dietary Guidelines: revision 2000: A statement for healthcare professionals from the Nutrition Committee of the American Heart Association. Circulation. 2000; 102:2284-2299.
  23. Atkins RC. Dr. Atkins' new diet revolution. 1st Avon pbk. ed. New York: Avon Books.
  24. Sacks FM, Bray GA, Carey VJ, Smith SR, Ryan DH, Anton SD, McManus K, Champagne CM, Bishop LM, Laranjo N, Leboff MS, Rood JC, de Jonge L, Greenway FL, Loria CM, Obarzanek E, Williamson DA. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. The New England Journal of Medicine. 2009; 360:859-873.
  25. Foster GD, Wyatt HR, Hill JO, Makris AP, Rosenbaum DL, Brill C, Stein RI, Mohammed BS, Miller B, Rader DJ, Zemel B, Wadden TA, Tenhave T, Newcomb CW, Klein S. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a randomized trial. Annals of Internal Medicine. 2010; 153:147-157.
  26. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the Atkins, Ornish, Weight Watchers, and Zone diets for weight loss and heart disease risk reduction: a randomized trial. JAMA. 2005; 293:43-53.
  27. Tsai AG, Wadden TA. The evolution of very-low-calorie diets: an update and meta-analysis. Obesity (Silver Spring, Md). 2006; 14:1283-1293.
  28. Fabricatore AN, Wadden TA, Ebbeling CB, Thomas JG, Stallings VA, Schwartz S, Ludwig DS. Targeting dietary fat or glycemic load in the treatment of obesity and type 2 diabetes: a randomized controlled trial. Diabetes Research and Clinical Practice. 2011; 92:37-45.
  29. Foster GD, Wyatt HR, Hill JO, McGuckin BG, Brill C, Mohammed BS, Szapary PO, Rader DJ, Edman JS, Klein S. A randomized trial of a low-carbohydrate diet for obesity. The New England Journal of Medicine. 2003; 348:2082-2090.
  30. Gardner CD, Kiazand A, Alhassan S, Kim S, Stafford RS, Balise RR, Kraemer HC, King AC. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA. 2007; 297:969-977.
  31. Samaha FF, Iqbal N, Seshadri P, Chicano KL, Daily DA, McGrory J, Williams T, Williams M, Gracely EJ, Stern L. A low-carbohydrate as compared with a low-fat diet in severe obesity. The New England Journal of Medicine.2003; 348:2074-2081.
  32. Shai I, Schwarzfuchs D, Henkin Y, Shahar DR, Witkow S, Greenberg I, Golan R, Fraser D, Bolotin A, Vardi H, Tangi-Rozental O, Zuk-Ramot R, Sarusi B, Brickner D, Schwartz Z, Sheiner E, Marko R, Katorza E, Thiery J, Fiedler GM, Bluher M, Stumvoll M, Stampfer MJ. Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. The New England Journal of Medicine. 2008; 359:229-241.
  33. Stern L, Iqbal N, Seshadri P, Chicano KL, Daily DA, McGrory J, Williams M, Gracely EJ, Samaha FF. The effects of low-carbohydrate versus conventional weight loss diets in severely obese adults: one-year follow-up of a randomized trial. Annals of Internal Medicine. 2004; 140:778-785.
  34. Das SK, Gilhooly CH, Golden JK, Pittas AG, Fuss PJ, Cheatham RA, Tyler S, Tsay M, McCrory MA, Lichtenstein AH, Dallal GE, Dutta C, Bhapkar MV, Delany JP, Saltzman E, Roberts SB. Long-term effects of 2 energy-restricted diets differing in glycemic load on dietary adherence, body composition, and metabolism in CALERIE: a 1-y randomized controlled trial. The American Journal of Clinical Nutrition. 2007; 85:1023-1030.
  35. Yancy WS, Jr., Olsen MK, Guyton JR, Bakst RP, Westman EC. A low-carbohydrate, ketogenic diet versus a low-fat diet to treat obesity and hyperlipidemia: a randomized, controlled trial. Annals of Internal Medicine. 2004; 140:769-777.
  36. Donnelly JE, Blair SN, Jakicic JM, Manore MM, Rankin JW, Smith BK. American College of Sports Medicine Position Stand. Appropriate physical activity intervention strategies for weight loss and prevention of weight regain for adults. Medicine and Science in Sports and Exercise. 2009; 41:459-471.
  37. Hagan RD, Upton SJ, Wong L, Whittam J. The effects of aerobic conditioning and/or caloric restriction in overweight men and women. Medicine and Science in Sports and Exercise. 1986; 18:87-94.
  38. Wing RR, Venditti E, Jakicic JM, Polley BA, Lang W. Lifestyle intervention in overweight individuals with a family history of diabetes. Diabetes Care. 1998; 21:350-359.
  39. Fogelholm M, Kukkonen-Harjula K. Does physical activity prevent weight gain--a systematic review. Obesity reviews : an official journal of the International Association for the Study of Obesity. 2000; 1:95-111.
  40. Jakicic JM, Marcus BH, Gallagher KI, Napolitano M, Lang W. Effect of exercise duration and intensity on weight loss in overweight, sedentary women: a randomized trial. JAMA. 2003; 290:1323-1330.
  41. Thomas JG, Bond DS, Phelan S, Hill JO, Wing RR. Weight-loss maintenance for 10 years in the National Weight Control Registry. American Journal of Preventive Medicine. 2014; 46:17-23.
  42. Klem ML, Wing RR, McGuire MT, Seagle HM, Hill JO. A descriptive study of individuals successful at long-term maintenance of substantial weight loss. The American Journal of Clinical Nutrition. 1997; 66:239-246.
  43. Whelton SP, Chin A, Xin X, He J. Effect of aerobic exercise on blood pressure: a meta-analysis of randomized, controlled trials. Annals of Internal Medicine. 2002; 136:493-503.
  44. Kraus WE, Houmard JA, Duscha BD, Knetzger KJ, Wharton MB, McCartney JS, Bales CW, Henes S, Samsa GP, Otvos JD, Kulkarni KR, Slentz CA. Effects of the amount and intensity of exercise on plasma lipoproteins. The New England Journal of Medicine. 2002; 347:1483-1492.
  45. Ross R, Dagnone D, Jones PJ, Smith H, Paddags A, Hudson R, Janssen I. Reduction in obesity and related comorbid conditions after diet-induced weight loss or exercise-induced weight loss in men. A randomized, controlled trial. Annals of Internal Medicine. 2000; 133:92-103.
  46. Hayes C, Kriska A. Role of physical activity in diabetes management and prevention. Journal of the American Dietetic Association. 2008; 108:S19-23.
  47. Sigal RJ, Kenny GP, Wasserman DH, Castaneda-Sceppa C, White RD. Physical activity/exercise and type 2 diabetes: a consensus statement from the American Diabetes Association. Diabetes Care. 2006; 29:1433-1438.
  48. Barry VW, Baruth M, Beets MW, Durstine JL, Liu J, Blair SN. Fitness vs. fatness on all-cause mortality: a meta-analysis. Progress in Cardiovascular Diseases. 2014; 56:382-390.
  49. Lee CD, Blair SN, Jackson AS. Cardiorespiratory fitness, body composition, and all-cause and cardiovascular disease mortality in men. The American Journal of Clinical Nutrition. 1999; 69:373-380.
  50. American Diabetes Association. The Diabetes Prevention Program. Design and methods for a clinical trial in the prevention of type 2 diabetes. Diabetes care. 1999;22(4):623-34.
  51. Look AHEAD Research Group. The Look AHEAD study: a description of the lifestyle intervention and the evidence supporting it. Obesity. 2006;14(5):737-52.
  52. Burke LE, Wang J, Sevick MA. Self-monitoring in weight loss: a systematic review of the literature. Journal of the American Dietetic Association. 2011;111(1):92-102.
  53. Patel ML, Wakayama LN, Bennett GG. Self‐monitoring via digital health in weight loss interventions: a systematic review among adults with overweight or obesity. Obesity. 2021;29(3):478-99.
  54. Hutchesson MJ, Rollo ME, Callister R, Collins CE. Self-monitoring of dietary intake by young women: online food records completed on computer or smartphone are as accurate as paper-based food records but more acceptable. Journal of the Academy of Nutrition and Dietetics. 2015;115(1):87-94.
  55. Spring B, Pellegrini CA, Pfammatter A, Duncan JM, Pictor A, McFadden HG, Siddique J, Hedeker D. Effects of an abbreviated obesity intervention supported by mobile technology: the ENGAGED randomized clinical trial. Obesity. 2017;25(7):1191-8.
  56. Jakicic JM, Davis KK, Rogers RJ, King WC, Marcus MD, Helsel D, Rickman AD, Wahed AS, Belle SH. Effect of wearable technology combined with a lifestyle intervention on long-term weight loss: the IDEA randomized clinical trial. JAMA. 2016;316(11):1161-71.
  57. Turner‐McGrievy GM, Wilcox S, Boutté A, Hutto BE, Singletary C, Muth ER, Hoover AW. The Dietary Intervention to Enhance Tracking with Mobile Devices (DIET Mobile) study: a 6‐month randomized weight loss trial. Obesity. 2017;25(8):1336-42.

58      Lichtman SW, Pisarska K, Berman ER, Pestone M, Dowling H, Offenbacher E, Weisel H, Heshka S, Matthews DE, Heymsfield SB. Discrepancy between self-reported and actual caloric intake and exercise in obese subjects. The New England Journal of Medicine. 1992; 327:1893-1898.

  1. Tronieri JS, Wadden TA. In-person behavioral approaches for weight management. G. Bray, C.Bouchard, P. Katzmarzyk, L. Redman, and P. Schauer, eds. In: Handbook of Obesity - Volume II: Clinical Applications, 5th ed. CRC Press; in press.
  2. Wadden TA, Tsai AG, Tronieri JS. A protocol to deliver intensive behavioral therapy (IBT) for obesity in primary care settings: the MODEL‐IBT program. Obesity. 2019;27(10):1562-6.
  3. Leblanc ES, O'Connor E, Whitlock EP, Patnode CD, Kapka T. Effectiveness of primary care-relevant treatments for obesity in adults: a systematic evidence review for the U.S. Preventive Services Task Force. Annals of Internal Medicine. 2011; 155:434-447.
  4. Perri MG, Shankar MN, Daniels MJ, Durning PE, Ross KM, Limacher MC, Janicke DM, Martin AD, Dhara K, Bobroff LB, Radcliff TA. Effect of telehealth extended care for maintenance of weight loss in rural US communities: a randomized clinical trial. JAMA Network Open. 2020;3(6):e206764.
  5. Perri MG, Limacher MC, von Castel‐Roberts K, Daniels MJ, Durning PE, Janicke DM, Bobroff LB, Radcliff TA, Milsom VA, Kim C, Martin AD. Comparative effectiveness of three doses of weight‐loss counseling: Two‐year findings from the rural LITE trial. Obesity. 2014;22(11):2293-300.
  6. Moyer VA. Screening for and management of obesity in adults: U.S. Preventive Services Task Force recommendation statement. Annals of Internal Medicine. 2012; 157:373-378.
  7. Wing RR, Tate DF, Gorin AA, Raynor HA, Fava JL. A self-regulation program for maintenance of weight loss. The New England Journal of Medicine. 2006; 355:1563-1571.
  8. Heymsfield SB, Wadden TA. Mechanisms, Pathophysiology, and Management of Obesity. The New England Journal of Medicine. 2017; 376:254-266.
  9. Donnelly JE, Smith BK, Dunn L, Mayo MM, Jacobsen DJ, Stewart EE, Gibson C, Sullivan DK. Comparison of a phone vs clinic approach to achieve 10% weight loss. International Journal of Obesity. 2007; 31:1270-1276.
  10. Appel LJ, Clark JM, Yeh HC, Wang NY, Coughlin JW, Daumit G, Miller ER, 3rd, Dalcin A, Jerome GJ, Geller S, Noronha G, Pozefsky T, Charleston J, Reynolds JB, Durkin N, Rubin RR, Louis TA, Brancati FL. Comparative effectiveness of weight-loss interventions in clinical practice. The New England Journal of Medicine. 2011; 365:1959-1968.
  11. Perri MG, Limacher MC, Durning PE, Janicke DM, Lutes LD, Bobroff LB, Dale MS, Daniels MJ, Radcliff TA, Martin AD. Extended-care programs for weight management in rural communities: the treatment of obesity in underserved rural settings (TOURS) randomized trial. Archives of Internal Medicine. 2008; 168:2347-2354.
  12. Cliffe M, Di Battista E, Bishop S. Can you see me? Participant experience of accessing a weight management programme via group videoconference to overcome barriers to engagement. Health Expectations. 2021 Feb;24(1):66-76.
  13. Azar KM, Aurora M, Wang EJ, Muzaffar A, Pressman A, Palaniappan LP. Virtual small groups for weight management: an innovative delivery mechanism for evidence-based lifestyle interventions among obese men. Translational Behavioral Medicine. 2015 Mar 1;5(1):37-44.
  14. Johnson KE, Alencar MK, Coakley KE, Swift DL, Cole NH, Mermier CM, Kravitz L, Amorim FT, Gibson AL. Telemedicine-based health coaching is effective for inducing weight loss and improving metabolic markers. Telemedicine and E-Health. 2019;25(2):85-92.
  15. Alencar MK, Johnson K, Mullur R, Gray V, Gutierrez E, Korosteleva O. The efficacy of a telemedicine-based weight loss program with video conference health coaching support. Journal of Telemedicine and Telecare. 2019;25(3):151-7.
  16. Tate DF, Wing RR, Winett RA. Using Internet technology to deliver a behavioral weight loss program. JAMA.2001; 285:1172-1177.
  17. Steinberg DM, Levine EL, Askew S, Foley P, Bennett GG. Daily text messaging for weight control among racial and ethnic minority women: randomized controlled pilot study. Journal of Medical Internet Research. 2013;15(11):e2844.
  18. Harvey-Berino J, West D, Krukowski R, Prewitt E, VanBiervliet A, Ashikaga T, Skelly J. Internet delivered behavioral obesity treatment. Preventive Medicine. 2010; 51:123-128.
  19. Podina IR, Fodor LA. Critical review and meta-analysis of multicomponent behavioral e-health interventions for weight loss. Health Psychology. 2018;37(6):501.
  20. Schippers M, Adam PC, Smolenski DJ, Wong HT, De Wit JB. A meta‐analysis of overall effects of weight loss interventions delivered via mobile phones and effect size differences according to delivery mode, personal contact, and intervention intensity and duration. Obesity Reviews. 2017;18(4):450-9.
  21. Steinberg DM, Tate DF, Bennett GG, Ennett S, Samuel‐Hodge C, Ward DS. The efficacy of a daily self‐weighing weight loss intervention using smart scales and e‐mail. Obesity. 2013;21(9):1789-97.
  22. Tate DF, Jackvony EH, Wing RR. A randomized trial comparing human e-mail counseling, computer-automated tailored counseling, and no counseling in an Internet weight loss program. Archives of Internal Medicine. 2006;166(15):1620-5.
  23. Martin CK, Miller AC, Thomas DM, Champagne CM, Han H, Church T. Efficacy of Smart LossSM, a smartphone‐based weight loss intervention: Results from a randomized controlled trial. Obesity. 2015;23(5):935-42.
  24. Forman EM, Goldstein SP, Crochiere RJ, Butryn ML, Juarascio AS, Zhang F, Foster GD. Randomized controlled trial of OnTrack, a just-in-time adaptive intervention designed to enhance weight loss. Translational Behavioral Medicine. 2019;9(6):989-1001.
  25. Thomas JG, Leahey TM, Wing RR. An automated internet behavioral weight-loss program by physician referral: a randomized controlled trial. Diabetes Care. 2015;38(1):9-15.
  26. Hales S, Turner-McGrievy GM, Wilcox S, Fahim A, Davis RE, Huhns M, Valafar H. Social networks for improving healthy weight loss behaviors for overweight and obese adults: a randomized clinical trial of the social pounds off digitally (Social POD) mobile app. International Journal of Medical Informatics. 2016;94:81-90.
  27. Vaz CL, Carnes N, Pousti B, Zhao H, Williams KJ. A randomized controlled trial of an innovative, user‐friendly, interactive smartphone app‐based lifestyle intervention for weight loss. Obesity Science & Practice. 2021;7(5):555-68.
  28. Pagoto S, Schneider K, Jojic M, DeBiasse M, Mann D. Evidence-based strategies in weight-loss mobile apps. American Journal of Preventive Medicine. 2013;45(5):576-82.
  29. Laing BY, Mangione CM, Tseng CH, Leng M, Vaisberg E, Mahida M, Bholat M, Glazier E, Morisky DE, Bell DS. Effectiveness of a smartphone application for weight loss compared with usual care in overweight primary care patients: a randomized, controlled trial. Annals of internal medicine 2014; 161:S5-12
  30. Thomas JG, Raynor HA, Bond DS, Luke AK, Cardoso CC, Wojtanowski AC, Vander Veur S, Tate D, Wing RR, Foster GD. Weight loss and frequency of body‐weight self‐monitoring in an online commercial weight management program with and without a cellular‐connected ‘smart’scale: a randomized pilot study. Obesity Science & Practice. 2017;3(4):365-72.

 

 

Islet Transplantation

ABSTRACT

 

Transplanting islets of Langerhans consists of implantation in the recipient’s hepatic portal system of endocrine pancreatic tissue, with a variable degree of purification. The field of islet transplantation has evolved significantly since the initial attempts by doctors Minkowski and von Mering in 1882, with remarkable acceleration over the last four decades, thanks to the incredible efforts of the research community worldwide, with continuous improvements in cell processing and transplantation techniques, patient management and development of specific immunotherapy protocols. Restoration of beta-cell function can be obtained by transplantation of allogeneic islets in both non-uremic (Islet Transplant Alone, ITA) and uremic (Simultaneous Islet and Kidney, SIK and Islet After Kidney, IAK) patients with diabetes, providing long-term sustained function and improved metabolic control even when requiring exogenous insulin (i.e., suboptimal islet mass transplanted or development of graft dysfunction). Preservation of beta-cell function is now attained in virtually all recipients of islet autografts, a therapeutic option that should be considered for individuals undergoing total pancreatectomy for non-malignant conditions and, as recently reported for selected cases with malignant conditions. In addition, islet transplantation represents an excellent platform toward the development of cellular therapies aimed at the restoration of beta-cell function using stem cells in the near future.  In this chapter, we will review the state-of-the art of clinical islet transplantation.

 

INTRODUCTION

Diabetes affects 537 million adults (20-79 years) throughout the world (2021) and this number will rise to 643 million by 2030 and 783 million by 2045 (IDF Diabetes Atlas 10th edition, https://diabetesatlas.org/). Many cases of diabetes are successfully treated with life-long multiple daily injections of exogenous insulin and monitoring of blood glucose levels. In the last decades significant improvements in insulin therapy thanks to new  preparations (i.e., ultrafast and long-lasting insulin analogues) and the adoption of intensive diabetes management (infusion pumps and continuous glucose monitoring system) have resulted in an overall improvement of patients’ glycemic control and a decreased incidence of chronic complications of diabetes (1,2). However, exogenous insulin administration cannot attain the desirable tight control in the majority of diabetics (3-5), cannot avoid the long-term complications of diabetes in all patients and the life expectancy of patients with diabetes is still shorter compared to that of the general population (6-8).  A broad international assessment of treatment outcomes in children and adults with T1D (including 324,501 people from 19 countries in Australasia, Europe and North America) showed that the proportion of patients with HbA1c <7.5% (58 mmol/mol)  varied from 15.7% to 46.4% among 44,058 people aged < 15 years, from 8.9% to 49.5% among 50,766 people aged 15-24 years and from 20.5% to 53.6% among 229 677 people aged ≥ 25 years (9). Diabetes is one of the leading causes of end-stage renal disease, blindness and amputation (10). In principle, the treatment for type 1 diabetes, type 3c diabetes and many cases of type 2 diabetes lies in the possibility of replacing destroyed or exhausted beta cell mass in order to restore two essential functions: sensing blood sugar levels and secreting appropriate amounts of insulin in the vascular bed, ideally into the portal system. Currently, the only available clinical approach of restoring beta cell mass in patients with diabetes is the allogenic/autologous transplantation of beta cells (i.e., pancreas or islet transplantation). Clinical trials performed in the last three decades have shown that restoration of beta-cell function via transplantation of isolated islet cells or vascularized pancreas allows reproducibly achievement of a more physiological release of endocrine hormones than exogenous insulin in subjects with diabetes (11). Transplanting islets of Langerhans consists of implantation in the recipient’s hepatic portal system of endocrine pancreatic tissue, with a variable degree of purification. Isolated islets are transplanted using minimally invasive techniques with lower morbidity than vascularized pancreas transplantation, which requires major surgery. The field of islet transplantation has evolved significantly since the initial attempts by doctors Minkowski andvon Mering in 1882 (12), with remarkable acceleration over the last three decades, thanks to the incredible efforts of the research community worldwide, with continuous improvements in cell processing and transplantation techniques, patient management and development of specific immunotherapy protocols. In addition, islet transplantation represents an excellent platform toward the development of cellular therapies aimed at the restoration of beta-cell function using stem cells in the near future.  In this chapter, we will review the state-of-the art of clinical islet transplantation.

 

WHEN TO CONSIDER ISLET TRANSPLANTATION?

 

Transplantation of pancreatic islet may be considered as a therapeutic option in several conditions associated with loss of beta-cell function (Table 1).  The procedure may be performed as Islet Transplant Alone (ITA) in non-uremic subjects, an option generally indicated for the treatment of iatrogenic (surgery-induced) diabetes and for non-uremic patients with Type 1 Diabetes.  Subjects with end-stage renal disease (ESRD) may be considered for Simultaneous Islet-Kidney (SIK) or, if already undergone renal transplantation, Islet After Kidney (IAK) transplantation, respectively. In special situations, transplantation of islets may be considered in combination with other organs (i.e., in the context of multi-visceral transplantation following exenteration comprising the pancreas) (13). 

 

The source of the islets for transplantation may be the patient’s own pancreas (autologous or auto-transplant) mainly when surgical removal of the gland is required due to different conditions.  After total pancreatectomy, the subject develops surgery-induced (iatrogenic) insulin-requiring diabetes.  Introduced in the early 1970’s (14), islet auto-transplantation allows achieving optimal metabolic control without the need for exogenous insulin in approximately 70% of the cases when adequate islet numbers can be recovered from the pancreas (generally >250,000 islet equivalents).  More than 500 auto-transplant in patients with near-total or total pancreatectomy have been performed to date (15). The largest series were published by the University of Minnesota (16-19), the University of Cincinnati (20,21), and Leicester (22-25). Even when an inadequate islet mass to attain insulin-independence has been recovered, stable metabolic control and excellent management can be achieved in most subjects undergoing autologous islet transplantation (18,26-31).  Islet auto-transplantation is currently reimbursed by health insurance in the United States.  In the past auto-transplant has been performed almost exclusively in patients undergoing pancreatectomy because of chronic pancreatitis, successfully preserving β-cell mass and preventing diabetes after major pancreatic resections (15,16,32,33). Additional indications for auto-transplant other than chronic pancreatitis are still controversial (34), and have been limited to the procedure performed only in small case series (35-40) of benign enucleable tumors or pancreatic trauma. Recently, broader selection criteria for auto-transplant were published (39,41), exploring the possibility of extending auto-transplant to patients with known malignancy, either having completion pancreatectomy as treatment for severe pancreatic fistulae or extensive distal pancreatectomy for neoplasms of the pancreatic neck or pancreatoduodenectomy because at high risk of pancreatic fistula (Table 1). Of note, a randomized, open-label, controlled, bicentric trial (NCT01346098) aimed to compare pancreaticoduodenectomy (PD) and Total Pancreatectomy with Islet AutoTransplantation (TP-IAT) in patients at high risk of Post-Operative Pancreatic Fistula (POPF) was recently published. The results indicate that TP-IAT can be considered a valid alternative to PD in these patients, as it reduced complication number, severity and length of hospital stay. Of note, a trend toward a reduction of mortality, even for patients with malignancy was also evident. As expected, TPIAT was associated to a higher risk of diabetes, but IAT was able to preserve, at least in part, the endogenous insulin secretion, mitigating the impact of the pancreoprivic diabetes and assuring a good metabolic control without severe hypoglycemic episodes. In the field of islet transplantation, this study definitively confirmed IAT could be indicated for pancreas diseases other than chronic pancreatitis, suggesting the possibility to extend IAT indications (Milan protocol(42)). For the first time in a randomized prospective design, it was  confirmed that IAT is feasible, safe and effective in patients with periampullary cancer,  in agreement with previous series of patients undergoing IAT after pancreatic resection for a wide spectrum of disease besides chronic pancreatitis (43) (44) (37). This approach will be tested in further studies in the next years, such as the recently started TPIAT-01 trial (NCT05116072), which hypothesize that TPIAT rather than PD may improve the access to adjuvant chemotherapy in patients with adenocarcinoma.

 

In the case of subjects who lost islet function (mainly patients with Type 1 Diabetes or, more rarely, previous total pancreatectomy) the only option currently available for transplantable islet cells is allogeneic donor pancreata. These are generally obtained through multi-organ donation after cerebral death, following conventional donor:recipient ABO blood type matching.  The use of a segment of the pancreas from living-related donors is technically feasible (45,46), but at the present time not preferred for islet transplantation due to the limited duration of graft function after transplantation of suboptimal islet numbers under standard immunosuppressive protocols, as well as the intrinsic risks for the donor (i.e., morbidity and risk to develop diabetes)(47). 

 

Table 1.  Indication for Islet Transplantation

Condition

Procedure

Type of Transplant

Diabetes Mellitus

Type 1

ITA, SIK, IAK

Allogeneic

Type 2

ITA, SIK, IAK

Allogeneic

Surgery-Induced Diabetes
(Iatrogenic)

Chronic pancreatitis

ITA

Autologous/Allogeneic

Trauma

ITA

Autologous/Allogeneic

Multi-visceral transplantation

Different combinations: Liver-Islet Transplantation, Bowel-Liver-Islet Transplantation, etc.

Allogeneic

Cystic Fibrosis

ITA

Lung-Islet Transplantation

Autologous/Allogeneic

Allogeneic

Benign enucleable tumors

ITA

Autologous

Borderline/malignant pancreatic neoplasms

ITA

Autologous/Allogeneic

 

Grade C pancreatic fistula requiring completion pancreatectomy

ITA

Autologous/Allogeneic

 

ITA- Islet Transplant Alone; SIK- Simultaneous Islet-Kidney; IAK- Islet After Kidney

 

The current main indication for an allogeneic islet transplant is Type 1 Diabetes, which is characterized by the selective destruction of islet beta cells due to an autoimmune process.  Ongoing clinical trials of allogeneic islet transplantation are recruiting subjects with unstable Type 1 Diabetes 18-65 years of age, either sex, with frequent metabolic instability requiring medical treatment (hypo-, hyper-glycemia, ketoacidosis) despite intensive insulin therapy; hypoglycemia unawareness (<54mg/dL); severe metabolic lability (mean amplitude of glycemic excursion >11,1 mmol/L or 200 mg/dl).  The inadequate efficacy of medical therapy to attain the desirable metabolic control in this specific patient population with unstable diabetes justifies the use of transplantation of pancreatic islets (either isolated cellular graft or vascularized whole pancreas) (48).  The main objective of the transplant is to correct the high susceptibility to severe hypoglycemia and glycemic imbalance that are associated with high mortality (8% in nonuremic subjects in the waiting list for 4 years to receive pancreas transplantation).  Further indications for an islet transplant are presence of progressive complications of diabetes and psychological problems with insulin therapy that may compromise adherence to the therapeutic regimen.  Islet transplant is indicated also for cases of subcutaneous insulin resistance requiring intraperitoneal or intravenous infusions, which are associated with substantial management hurdles and morbidity.

 

Table 2.  Inclusion and Exclusion Criteria for Allogeneic Islet Transplantation in T1DM*

Inclusion Criteria:

·                Mentally stable and able to comply with study procedures

·                Clinical history compatible with type 1 diabetes with onset of disease at <40 years of age, insulin dependence for at least 5 years at study entry, and a sum of age and insulin dependent diabetes duration of at least 28

·                Absent stimulated C-peptide (<0.3 ng/ml) 60 and 90 minutes post-mixed-meal tolerance test

·                Involvement of intensive diabetes management, defined as:

o        Self-monitoring of glucose values no less than a mean of three times each day averaged over each week

o        Administration of three or more insulin injections each day or insulin pump therapy

o        Under the direction of an endocrinologist, diabetologist, or diabetes specialist with at least three clinical evaluations during the past 12 months prior to study enrollment

·                At least one episode of severe hypoglycemia in the past 12 months, defined as an event with one of the following symptoms: memory loss; confusion; uncontrollable behavior; irrational behavior; unusual difficulty in awakening; suspected seizure; seizure; loss of consciousness; or visual symptoms, compatible with hypoglycemia in which the individual required assistance of another subject was unable to treat him/herself person and which was associated with either a blood glucose level <54 mg/dl or prompt recovery after oral carbohydrate, intravenous glucose, or glucagon administration in the 12 months prior to study enrollment

·                Reduced awareness of hypoglycemia

 

Exclusion Criteria:

·                Body mass index (BMI) >30 kg/m2 or weight ≤50 kg

·                Insulin requirement of >1.0 IU/kg/day or <15 U/day

·                HbA1c >10%

·                Untreated proliferative diabetic retinopathy

·                Systolic blood pressure >160 mmHg or diastolic blood pressure >100 mmHg

·                Measured glomerular filtration rate using iohexol of <80 ml/min/1.73mm2.

·                Presence or history of macroalbuminuria (>300 mg/g creatinine)

·                Presence or history of panel-reactive anti-HLA antibody levels greater than background by flow cytometry.

·                Pregnant, breastfeeding, or unwilling to use effective contraception throughout the study and 4 months after study completion

·                Presence or history of active infection, including hepatitis B, hepatitis C, HIV, or tuberculosis.

·                Negative for Epstein-Barr virus by IgG determination

·                Invasive aspergillus, histoplasmosis, or coccidioidomycosis infection in the past year

·                History of malignancy except for completely resected squamous or basal cell carcinoma of the skin

·                Known active alcohol or substance abuse

·                Baseline Hgb below the lower limits of normal, lymphopenia, neutropenia, or thrombocytopenia

·                History of Factor V deficiency

·                Any coagulopathy or medical condition requiring long-term anticoagulant therapy after transplantation or individuals with an INR greater than 1.5

·                Severe coexisting cardiac disease, characterized by any one of the following conditions:

o        Heart attack within the last 6 months

o        Evidence of ischemia on functional heart exam within the year prior to study entry

o        Left ventricular ejection fraction <30%

·                Persistent elevation of liver function tests at the time of study entry

·                Symptomatic cholecystolithiasis

·                Acute or chronic pancreatitis

·                Symptomatic peptic ulcer disease

·                Severe unremitting diarrhea, vomiting, or other gastrointestinal disorders that could interfere with the ability to absorb oral medications

·                Hyperlipidemia despite medical therapy, defined as fasting LDL cholesterol >130 mg/dl (treated or untreated) and/or fasting triglycerides >200 mg/dl

·                Currently receiving treatment for a medical condition that requires chronic use of systemic steroids except for the use of 5 mg or less of prednisone daily, or an equivalent dose of hydrocortisone, for physiological replacement only

·                Treatment with any antidiabetic medication other than insulin within the past 4 weeks

·                Use of any study medications within the past 4 weeks

·                Received a live attenuated vaccine(s) within the past 2 months

·                Any medical condition that, in the opinion of the investigator, might interfere with safe participation in the trial

o        Treatment with any immunosuppressive regimen at the time of enrollment.

o        A previous islet transplant.

·                A previous pancreas transplant, unless the graft failed within the first week due to thrombosis, followed by pancreatectomy and the transplant occurred more than 6 months prior to enrollment.

 

*Modified from the information relative to active trials from the Clinical Islet Transplant Consortium (www.citisletstudy.org/) as listed at http://clinicaltrials.gov/ct2/show/NCT00434811.

 

MULTIDISCIPLINARY TEAM

 

Islet Transplant Programs require the integration of multidisciplinary expertise.  The endocrinologist expert in diabetes diagnosis and management is essential member of the team, and can identifying subjects who may benefit of beta-cell replacement therapy, and help with the evaluation of metabolic control during all phases of the follow-up.  The psychologist is involved in the evaluation of islet transplant candidates to assess their motivation, mental fit to enroll in the trial, and ability to adhere to the therapy.  Psychometric and psychological evaluations are performed during the follow-up period after transplantation.  Transplant surgeons provide the expertise in organ procurement, with transplant procedures, overall management of patients and immunosuppression.  A dedicated Cell Transplant Center with specialized experts in pancreatic cell isolation, purification, culture, potency assessment and quality assurance warrant that islet cell products are manufactured for clinical transplantation following cGMP standards and FDA regulations.  The interventional radiologist performs the noninvasive cannulation of the portal vein and participates to the post-transplant monitoring of the liver using noninvasive imagine techniques.  The organ procurement organizations and organ distribution networks (UNOS in the U.S.) contribute to the identification and allocation of donor organs matching the recipient’s characteristics.  The ophthalmologist and nephrologist are involved to monitor and treat progressive diabetic complications (i.e., retinopathy and renal function, respectively).

 

ISLET ISOLATION AND TRANSPLANTATION

 

Islets are highly vascularized cell clusters ranging <50um to ~800um of diameter that constitute the endocrine component of the pancreas.  It has been estimated that a healthy pancreas may contain approximately 106 islets scattered throughout the gland, and accounting for only ~1% of total pancreatic tissue.  Each cluster comprises several thousands of endocrine cell subsets that are closely in touch with capillaries and with each other.  Complex cell-cell interactions between different cell subsets, innervation, incretins and metabolites (sugar and amino acids, amongst other) in the blood and interstitial space all contribute to the proper control of glucose homeostasis (49).  Preservation of the integrity of islet cell cluster is a prerequisite for their optimal function.  The procedure currently used to extract islets from human pancreas is the so called automated method for isolation of the islets of Langerhans, established in 1987 by Ricordi and colleagues (50). Before the beginning of the isolation procedure, the spleen and the duodenum are removed from the pancreas and an accurate dissection and removal of the peripancretic fat, lymph nodes and vessels is performed. Then, the pancreas is divided at the neck and two 16-20 gauge angiocatheters are inserted into the main pancreatic ducts. The organ is then perfused with cooled perfusion solution containing collagenase and serine – protease inhibitor – dissolved in buffer at a pressure of 140-180 mmHg. After 10 minutes of cold perfusion, the distended pancreas is further cut into smaller sections, and placed into the Ricordi chamber. This chamber is composed of a superior and an inferior part, separated by a filter that has pores of about 700μm. Seven to nine stainless steel balls and the fragments of the pancreas are placed into the inferior part of the chamber, which is then filled with the digestion solution and closed together with the superior part of the chamber. A peristaltic pump connected to the system is activated creating a flow of 40 ml/min. The digestion runs in a closed circuit where warm Hank’s solution is pumped in the inferior chamber and the tissue released in the solution passes in the superior chamber through the filter. The collagenase is re-circulated at a temperature not exceeding 37°C and the chamber is agitated. When most of the islets are free of the surrounding acinar tissue, and intact islets are observed, the heating circuit is bypassed. The temperature is progressively decreased to 10°C and the collagenase diluted with cold RPMI. The free islets are then collected in containers, washed several times, re-suspended in cold organ preservation solution and purified with a continuous ficoll gradient using a Cobe 2991 cell separator. At the end of the procedure samples of the islet preparation are collected and evaluated through staining with dithizone (DTZ) which marks zinc in the insulin granules, resulting in a characteristic red stain. Adding few drops of DTZ solution to a sample allows easy evaluation of the morphology and number of isolated islets through computerized digital analysis. The islet manufacturing processes must be controlled by different assays and the islet batch product validated and characterized. Then safety testing is carried out for sterility and pyrogenicity, identity (insulin content), cell number (amount of tissue, counting of islets), purity (percentage of ductal, acinar, beta, and other cells), viability (islet nucleotide content), potency (insulin secretory response) and finally stability (storage in culture). Specific features of the final islet preparation are a required for islet preparations used in islet transplantation, in particular purity (> 20% of the preparation being islets), adequate number of islets (>5,000 islet equivalent recipient body weight for the first infusion, >3,000 for further infusions) and total tissue volume (< 5 ml). The infusion of the islets can be performed a few hours after the end of the isolation process or up to 72 hours thereafter. The implantation site is usually the hepatic parenchyma through the portal system of the recipient. Recently other implantation sites have been proposed (51) in the clinical setting, like the bone marrow (52,53), the subcutaneous site(54), the gastric submucosa (55), the omentum(56,57) or striated muscle (58,59), which in the future, may prove to be valid alternative sites for islet transplantation. The adequate amount of islets obtained is calculated with respect to the body weight of the recipient and re-suspended immediately before intrahepatic transplantation in 40-60 mL of a solution suitable for injection (Ringer Lactate, 1% Human Albumin and 2000 IE of heparin). Percutaneous trans hepatic catheterization is the most common access route, as well as a mini-laparotomy and cannulation of an omental or mesenteric vein, or recanalization of the umbilical vein. Access to the portal vein is usually provided by interventional radiologists. If the portal pressure is documented to be below 20 mmHg, the islet infusion bag is connected with the portal vein catheter and infused over a period of 15 to 60 minutes. Islet infusion is halted if the portal pressure exceeds 22 mmHg. After completion of the islet infusion, the catheter is withdrawn; coils and gelatin-sponges are deployed in the puncture tract to prevent bleeding. A schematic animation of the islet isolation and transplant procedures is available online [http://www.youtube.com/watch?v=aMNKu-ZVUls].

 

THE CONSORTIUM CONCEPT

 

A major development in the field of islet transplantation is the combination of individual centers into larger groups such as the GRAGIL network in France and Switzerland, the Nordic Network for Clinical Islet Transplantation (NNCIT) in the Scandinavian countries and the Clinical Islet Transplant Consortium (CITC) internationally but concentrated in North America. The need for dedicated infrastructures and personnel specialized in islet cell processing, quality assessment and cGMP standards impose an enormous financial burden on any Clinical Islet Transplant Program.  Acquiring and maintaining the specialized expertise in islet cell processing requires a steep learning curve and continuous refinements and training that add to the costly procedure.  Recent data have shown that the experience of the clinical islet transplant team in cell processing and management of immunosuppression are critically important in determining the success of a clinical trial (60).  Based on these premises, the development of regional cell processing centers that are part of consortia that are integrated with distant transplant centers is increasingly being considered as a practical and cost-effective strategy (Figure 1).  Initial reports of successful clinical trials carried on in the context of Consortia both in Europe and North America (61-64) support the feasibility of such an approach, which may be of assistance in reducing the operational costs while enhancing the success rate of clinical trials (i.e., better utilization of donor pancreata, more reproducible success in obtaining adequate numbers of functional islets from a donor pancreata, etc.). 

Figure 1.  Islet Transplant Consortium Models.  A.  The centralized (or ‘regional’) Cell Processing Facility receives the donor pancreas from a distant Transplant Center and isolates islet cell products that are sent back for implant.  B.  The centralized Cell Processing facility receives the donor pancreas from one of the Transplant Centers and distributes the isolated islets to any of the Transplant Center in the Consortium according to the best match of the cell product for the transplant candidate on the waiting list for transplant (that is, the islet cell product is not necessary returned to the center recovering the pancreas). 

 

ISLET TRANSPLANT ACTIVITY

 

The Collaborative Islet Transplant Registry (CITR)

 

In 2001, the National Institute of Diabetes & Digestive & Kidney Diseases established the Collaborative Islet Transplant Registry (CITR) to compile data from all islet transplant programs in North America from 1999 to the present. The Juvenile Diabetes Research Foundation (JDRF) granted additional funding to include the participation of JDRF-funded European and Australian centers from 2006 through 2015. The cumulated North American, European and Australian data are pooled for analyses included in the annual report. CITR Annual Reports are publicly available as open access and can be downloaded or requested in hard copy at www.citregistry.org. From 1999 through 2020 – the cut-off for the last Eleventh Annual Report – CITR has collected data on the following groups of study subjects:

 

  1. Allogeneic islet transplantation (typically cadaveric donor), performed as either islet transplant alone (ITA) or islet-after-kidney (IAK). A small number of cases have been performed as islet simultaneous with kidney (SIK) or kidney-after-islet (KAI).
  2. Autologous islet transplantation, performed after total pancreatectomy (N=1,233) are also reported to CITR.

 

As of December 15, 2020, the CITR Registry included data on 1,399 allogeneic islet transplant recipients (1,108 islet transplant alone, ITA, and 236 islet after kidney, IAK, 49 simultaneous islet kidney, SIK, and 6 kidney after islet, KAI), who received 2,832 infusions from 3,326 donors. From 1999 through 2020, 28 National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) sponsored North American and 12 international Eurasian and Australian islet transplant centers (40 total) contributed data to the Collaborative Islet Transplant Registry (CITR).  Combining the ITA and IAK recipients, 27.4% received a single islet infusion, 48.1% received two, 20.4% received three, and 4.1% received 4-6 infusions. Of 26 North American sites performing Auto-ITx from 1999 through December 2020, 15 reported data to CITR along with 5 European and Australian islet transplant centers. These sites registered 1233 autoislet transplant recipients. Of these, 1123 recipients were in North America, 98 in Europe, and

12 in Australia. One-hundred eight-five (185) were aged less than 18, and 1,057 were 18 or older at the time of their transplant.

 

Outside The Collaborative Islet Transplant Registry (CITR)

 

Although the CITR is an extraordinary source of valuable data, a recent publication indicates that it does not capture a major part of the international islet transplant activities and outcomes (65). In fact, a global online survey was recently administered to 69 islet transplantation programs. After integration of all data obtained, 103 islet transplant centers were identified, of which 94, in 25 countries, had reported allotransplantation activity during the 2000–2020 period: 15 in Asia (16%), 39 in Europe (42%), 34 North America (36%), 3 in Oceania (3%) and 3 in South America (3%) and between January 2000 and December 2020, 4,321 islet allotransplants in 2,149 patients were reported worldwide.  Most islet transplants were performed in Europe (2,608, 59.7%), followed by North America (1,475, 33.8%), Asia (135, 3.1%), Oceania (119, 2.7%) and South America (28, 0.6%). Actually the ANZIPTR (Australia and New Zealand Islet and Pancreas Transplant Registry) and NHS-BT (UK National Health Service-Blood and Transplant) registry are publicly available registries containing a wealth of data on islet and pancreas transplantation in Australia/New Zealand and UK, respectively, including outcomes (66) (67). The European Pancreas and Islet Transplant Registry (EPITR) is a current effort from ESOT/EPITA aiming at covering these needs for Europe (https://esot.org/epita/epita-epitr/).

 

Clinical Management of Islet Transplant Recipients

 

The clinical management of islet transplant recipients requires the concerted effort of endocrinologist and transplant teams.

 

Immunosuppression

 

Preexisting and transplant-induced auto- and allo-specific cellular immune responses play a crucial role in the loss of islets and islet function infused in the liver (68-70) along with non-specific immune responses predominantly mediated by innate inflammatory processes related to mechanics and site (71-74). Islet graft rejection occurs without clinical symptoms. Neither guidelines nor formal consensus on the “best” or “standard” immunosuppressive strategy for human islet transplantation are currently available. Multiple induction and maintenance agents are administered peri- and post- every infusion in the same recipient. According to CITR data (75), a substantial shift in immunosuppression strategies has been documented during the last years.

 

Induction with interleukin-2 receptor antagonist (e.g., daclizumab) only, which comprised about 53% of all initial infusions in 1999-2002, was replaced or supplemented with regimens that included T-cell depletion with/without TNF antagonists in about 67% of the new infusions performed since 2015 (11,76-83). In 1999-2002, maintenance immunosuppression was predominantly (64%) calcineurin (CNI) +mTOR inhibitors (60). It was increasingly replaced or supplemented throughout the eras by a CNI and IMPDH-inhibitor combination (77,84-86).; in the most recent era, CNI+mTOR inhibitors were used in 15% of new infusions while CNI+IMPDH inhibitors were used in about 62%. Moreover, the use of alemtuzumab-induction therapy was recently reported and associated with encouraging longer-term function (87,88). New biologic agents with potentially lower islet cell and organ toxicity profiles are currently being evaluated in ongoing clinical trials.  Amongst these are agents that target co-stimulation pathways in immune cells and/or adhesion molecules (CTLA4-Ig, LFA-1 PD-1/PD-L1 CD40 ) (89-95) or chemokine receptors (CXCR1/2) (71,96). Finally, calcineurin inhibitor-free immunosuppressive regimen was reported (97).

 

Antibiotic and Antiviral Prophylaxis

 

Subjects receiving immunosuppression therapy are more susceptible to opportunistic infections, as well as reactivation or de novo occurrence of viral infections.  Antibiotic prophylaxis for Pneumocystis carinii consists in trimethoprim and sulfamethoxazole three times a week.  Antiviral prophylaxis is aimed are reducing the risk, or treating the occurrence, of cytomegalovirus infections (which have been recognized increasing the risk of graft loss in solid organ transplantation) and of reactivation of Epstein - Barr virus infection (which has been associated with the dreadful post-transplant lymphoproliferative disease, PTLD).  Current protocols utilize antiviral therapy with vanglancyclovir daily for the first trimester post-transplant.  Monitoring of viremia in peripheral blood samples by PCR is becoming a routine during the follow-up as it allowed for the early detection of reactivation or de novo infections that may be treatable without compromising graft outcome (98,99). 

 

Thromboembolism Prophylaxis

 

It has been recognized that isolated islets produce tissue factor and other pro-inflammatory molecules that may trigger an instant blood-mediated inflammatory reaction upon infusion into the blood stream.  This, in turn, may enhance the generation of noxious stimuli after embolization in hepatic sinusoids of the liver, significantly reducing the mass of functional islets engrafting.  An aggressive heparin treatment is generally implemented in the early period after transplant.  Heparin is added to the transplant medium used during the islet infusion, while low molecular weight heparin injections are administered in the post-transplant period. This is aimed at enhancing islet engraftment in the hepatic portal system while reducing the risk of portal thrombosis. 

 

Peri-transplant Insulin Management

 

Islet engraftment may take up a few weeks to allow for neovascularization of the clusters in the transplant microenvironment.  The monitoring of glycemic control in the immediate post-transplant period should be intense to attain tight glycemic values in order to avoid excessive workload for the newly transplanted islets as well as to prevent hypoglycemic episodes.  This is generally done by providing basal exogenous insulin that is then progressively reduced and withdrawn according to the glycemic values measured.

 

POST-TRANSPLANT CLINICAL MONITORING

 

Monitoring of cell blood counts (erythrocytes, white blood cells and differential), hemoglobin, platelets and coagulation markers is routinely performed in the post-transplant period.  These tests allow assessing the myelosuppressive effects of anti-rejection drugs.  In the case of anemia with clinical relevance, iron supplementation may be indicated, while for more severe cases erythropoietin treatment is implemented.  In the case of severe neutropenia, marrow stimulation with granulocyte-colony stimulating factor (G-CSF) is promptly implemented.

 

Renal function is monitored periodically in the follow-up of islet transplant recipients to assess the impact of restoring beta-cell function on the progression of diabetic nephropathy, and also to identify and timely correct potential nephrotoxicity of anti-rejection drugs (i.e., CNI and mTOR inhibitors).  Standard serological tests (serum creatinine, azotemia), urine tests (spot and 24-hr collections) are frequently performed during the follow-up and glomerular filtration rates (GFR) estimated using different algorithms (i.e., MDRD).  The nephroprotective effect of ACE inhibitors and of antagonists of angiotensin-receptor (ARB) in subjects with diabetes has been recognized, and their use is particularly indicated in transplant recipients treated with immunosuppressive drugs known for their negative effects on renal function (100-104).  Elevations of blood pressure from the range 130/80 mmHg are promptly treated pharmacologically.

 

Monitoring of lipid levels and prompt treatment of dyslipidemia are important in transplanted patients.  Some of the anti-rejection drugs (i.e., mTOR inhibitors) are prone to induce dyslipidemia, which in turn may have toxic effects on beta-cells or contribute to creating an unfavorable environment (i.e., steatosis) in the liver (105).  Prophylactic use of statins targeting LDL cholesterol levels <100mg/dL can be contemplated for islet transplant recipients. 

 

Liver function is monitored in the post-transplant period.  It is common to observe a transient and self-limited elevation of liver enzymes (transaminitis) because of the embolization of islets into the liver sinusoids (106,107).  This is often associated with hyper-echoic pattern of the liver parenchyma at ultrasound evaluation in the early days post-transplant.  This phenomenon resolves spontaneously without the need for medical treatment.  Ultrasound evaluation of the liver and abdominal cavity in the days post-transplant also allows identifying timely possible procedural complication of the transplant, such as portal thrombosis, peritoneal hemorrhage and alterations of echogenicity of hepatic parenchyma (108).

 

The immune monitoring after islet transplantation does not differ much from that of any other organ transplant (109).  Basal and serial evaluation of Panel Reactive Antibodies (PRA) is performed to determine possible allosensitization against Human Leukocyte Antigens (HLA) class I and II of the Histocompatibility complex of transplanted tissue.  Generally, maintenance of an adequate immunosuppressive regimen can prevent the development of alloantibodies, thereby preventing their deleterious effects on graft survival and function (i.e., chronic rejection leading to graft loss) (110-112).  Nonetheless, development of alloreactivity against donor or non-specific antigens may develop whenever reduction (i.e., during infections, toxicity and drug conversion, amongst other causes) or suspension (i.e., after complete graft loss) of immunosuppression is needed (112,113).  The autoimmune process underlying Type 1 Diabetes is associated with the appearance of antibodies against self-antigens (autoantibodies; i.e., towards GAD, IA-1 and insulin).  Serial titration of autoantibody levels during the follow-up period may enable detecting a reactivation of the autoimmune process, measured as conversion to positive values in previously negative subjects, or increase of antibody titers.  These have been associated with a lower rate of insulin independence and shorter duration of graft function after islet transplantation (60,70).  New assays for additional autoantibodies (i.e., ZnT8) and for autoreactive T cells are under evaluation to help enhancing the sensitivity of immune monitoring for early detection of recurrence of autoimmunity, which may enable implementation of timely immune interventions to rescue the transplanted cells (114-116).  

 

MONITORING ISLET GRAFT FUNCTION

 

Several metabolic parameters allow monitoring the function of transplanted islets (Table 3).  Since only subjects with Type 1 Diabetes who have undetectable stimulated c-peptide (<0.3 ng/dl) before transplant are recruited for an islet transplant, monitoring of basal and stimulated c-peptide offers an excellent biomarker of graft function, even when exogenous insulin is required.  There is no consensus on which approach is most suited to accurately assess functional islet mass.  Algorithms and indices that combine multiple parameters have been developed and proposed to help simplifying and obtaining objective functional assessment of islet transplant recipients (117-126).  The main goal is to identify early changes that indicate propensity to graft dysfunction (i.e., functional impairment during an infection, drug-induced toxicity).  Stimulation tests are performed before (at enrollment) and during the follow-up after transplant to evaluate the functional potency of the transplanted islets in response to different secretagogues (i.e., glucose, arginine, or mixed meal test).  Insulin therapy is generally implemented when random glycemic sampling demonstrates on three subsequent occasions within the same week fasting values >140 mg/dl (7.8 mmol/L) and postprandial values >180 mg/dl (10.0 mmol/L), or after recording two consecutive A1c values >6.5%.

 

Table 3.  Monitoring of Islet Graft Function

Standard

Stimulation

Indices

Glycosylated Hb (A1c)

Fasting glycemia

Postprandial glycemia

MAGE*

CGMS*

Basal C-peptide

Daily insulin requirement

 

Mixed Meal

Intravenous glucose

Intravenous arginine

Hypo score

Liability index

Βeta score

Beta 2 score

Basal C-peptide/Glucose ratio

HOMA-B*

HOMA-IR*

TEF*

*Abbreviations.  CGMS: Continuous Glucose Monitoring System. MAGE: Mean Amplitude of Glucose Excursions. HOMA-B: Homeostasis Model Assessment – functional beta cell mass.  HOMA-IR: Homeostasis Model Assessment – Insulin-Resistance. TEF: Transplant Estimated Function

 

The Igls Score

 

The lack of standardized definition of graft functional and clinical outcomes remains a source of concern in β-cell replacement influencing its recognition as a valid clinical option from the endocrinology community. In order to address this issue, the International Pancreas & Islet Transplant Association (IPITA) joined with the European Pancreas & Islet Transplant Association (EPITA) for a two-day workshop on “Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes” in January 2017 in Igls, Austria. The main objective was to develop consensus on the definition of function and failure of current and future forms of β-cell replacement therapies.  As result of the workshop, an IPITA/EPITA Statement was recently published (127,128). This Statement introduces some relevant innovations in the field including a new classification for the definition of clinically successful outcome. The functional status and clinical success of a β-cell graft should be defined separately using the same components of assessment: the HbA1c, severe hypoglycemic events, insulin requirements, and C-peptide. Concordantly, a four-tiered system was proposed to classify the functional outcomes of β-cell replacement:

 

  • optimal β-cell graft function: HbA1c ≤6.5%, the absence of any severe hypoglycemia, the absence of any requirement for exogenous insulin or other anti-diabetic drugs, and documentation of an increase over pre-transplant measurement of C-peptide
  • good β-cell graft function: defined by: HbA1c <7.0%, the absence of any severe hypoglycemia, a reduction by more than 50% from baseline in insulin requirements or the use of non-insulin anti-diabetic drugs, and documentation of an increase over pre-transplant measurement of C-peptide.
  • marginal β-cell graft function: no modification of HbA1c, the reduction of severe hypoglycemia, a reduction by less than 50% from baseline in insulin requirements, and documentation of an increase over pre-transplant measurement of C-peptide.
  • failure β-cell graft function: absence of any evidence for a clinical impact (no modification of HbA1c, incidence of severe hypoglycemia and insulin requirement) and clinically insignificant levels of C-peptide.

 

Clinically successful outcomes includes both optimal and good functional outcomes, implying that the use of exogenous insulin or other anti-diabetic drugs is not synonymous with graft loss or failure. Neither a marginal β-cell graft nor a failed β-cell graft is considered a clinically successful.  However, if documented impairment in hypoglycemia awareness, frequent occurrence or exposure to severe hypoglycemia, or marked glycemic variability/lability is convincingly improved, then it may be appropriate to consider that the β-cell graft is clinically impactful also in marginal function and the benefit of maintaining β-cell graft function may outweigh risks of maintaining immunosuppression. This implies that hypoglycemia awareness, serious hypoglycemia, and glycemic variability/lability must be evaluated at baseline for monitoring whether a marginally functioning graft is continuing to provide any clinical impact.

IPITA / EPITA Statement has the merit of having introduced a defined concept of clinical success based on easily measurable parameters over time and with a wide consensus of international experts Implementation of this new β-cell replacement outcome definition and its use in publication will  be critical to improve the performance and to reliably compare the different β-cell replacement  strategies (129).

 

In July 2019, a symposium at the 17th IPITA World Congress was held to examine the Igls criteria after 2 years in clinical practice, including validation against continuous glucose monitoring (CGM)-derived glucose targets, and to propose future refinements that would allow for comparison of outcomes with artificial pancreas system approaches. A new Igls 2.0 form composite criteria was suggested (130), in which clinical outcome based on glucose regulation is separated from β-cell graft function, with the latter considered only for further qualification of β-cell replacement modalities (Table 4-5).

 

Table 4. Proposed Igls Criteria 2.0

Treatment outcome

Glycemic control

Hypoglycemia

Treatment success

 

HbA1c, % (mmol/mol)a

CGM, % time-in-range

Severe hypoglycemia, events per y

CGM, % time < 54 mg/dl (3.0 mmol/L)

 

Optimal

≤6.5 (48)

≥80

None

0

Yes

Good

<7.0 (53)

≥70

None

<1

Yes

Marginal

≤Baseline

>Baseline

<Baselineb

<Baseline

Noc

Failure

~Baseline

~Baseline

~Baselined

~Baseline

No

Baseline, pretransplant assessment (not applicable to total pancreatectomy with islet autotransplantation patients).

Abbreviations: CGM, continuous glucose monitoring; HbA1c, glycated hemoglobin.

a Mean glucose should be used to provide an estimate of the HbA1c, termed the glucose management indicator, in the setting of disordered red blood cell life span.

b Should severe hypoglycemia occur following treatment, then continued benefit may require assessment of hypoglycemia awareness, exposure to serious hypoglycemia (<54 mg/dL [3.0 mmol/L]), and/or glycemic variability/lability with demonstration of improvement from baseline.

c Clinically, benefits of maintaining and monitoring β-cell graft function may outweigh risks of maintaining immunosuppression.

d If severe hypoglycemia was not present before β-cell replacement therapy, then a return to baseline measures of glycemic control used as the indication for treatment (6, 7) may be consistent with β-cell graft failure.

 

Table 5. Proposed Igls Criteria 2.0

β-cell graft functione

C-peptide, ng/mL (nmol/L) f

Insulin use or noninsulin antihyperglycemic therapy

Optimal

Any

None

Good

>0.5 (0.17) stimulated
≥0.2 (0.07) fasting

Any

Marginal

0.3-0.5 (0.10-0.17) stimulated
0.1-<0.2 (0.04-<0.07) fasting

Any

Failure

<0.3 (0.10) stimulated
<0.1 (0.04) fasting

Any

e Categorization of β-cell graft function must first meet treatment outcome based on measures of glucose regulation.

f May not be reliable in uremic patients and/or in those patients with evidence of C-peptide production before β-cell replacement therapy.

 

IMPACT OF ISLET TRANSPLANTATION ON METABOLIC CONTROL (TABLE 6)

 

Four successful large-scale Phase 3 clinical trials in islet transplantation have been published recently: CIT-07 (multicenter, single-arm)(131), CIT06 (pivotal trial) (132), TRIMECO (multicenter, open-label, randomized) (133) and REP0211 (multicenter, Double blind, randomized) (134).  All these studies demonstrate that human islets, when transplanted in patients with T1D with impaired awareness of hypoglycemia and severe hypoglycemic events, can safely and efficaciously maintain optimal glycemic control (135). The clinical experience confirms that the most remarkable effect of the islet transplant is the abrogation of severe hypoglycemia and the recovery of hypoglycemia awareness, which persists after development of graft dysfunction and even several months after graft failure (and loss of detectable c-peptide) (136,137). Following islet transplantation, the restoration of β cell responses to secretagogue stimulation is observed, with improved insulin secretion (‘first phase’) in response to intravenous glucose, as well as increased c-peptide secretion in response to oral glucose. Normalization of glycemic threshold triggering the release of counter-regulatory hormones can be demonstrated during hypoglycemic clamp studies, even if without reaching normalization of the magnitude of the vegetative response; furthermore, quasi-normal glucagon secretion in response to hypoglycemia can be observed (138-142). In addition to controlling hypoglycemia, insulin independence can be achieved when an adequate islet mass is transplanted (143). After islet transplantation, 5-year insulin independence may be as high as 50%. A quarter of patients may remain insulin independent, with HbA1c concentrations of less than or equal to 6·5%, for at least 10 years, with either islet transplantation alone or islet-after-kidney transplantation (144)(145). Moreover, the glucose control associated with excellent islet graft function closely matches glucose values measured in healthy adults: median glucose 103 mg/dl (95-112), glucose standard deviation around the mean value 14 (11-20), 0% time >180 mg/dl, 0% time <54 mg/dl, HbA1c 5.6 (5-5.8) (146). Additionally, a significant improvement of quality of life after islet transplant has been documented by using standardized psychometric instruments and interviews carried on by psychologists (147-155). Associated with the better glucose control and the evidence of islet function (c-peptide secretion), a positive impact on the microvascular complications of  T1D has been described while is less evident on the macrovascular complications (156). More specifically, a stabilization/slower progression of retinopathy (104,157-159) and neuropathy (158,160-162) and an improvement of micro- and macroangiopathy (79,101,102,154,157,163-168) have been described. Some studies reported also a reduction of atherothrombotic profile paralleled by reduced incidence of cardiovascular accidents, an amelioration of cardiovascular and endothelial function, improved longevity of renal transplant (165) and a higher survival rates after islet transplantation in IAK recipients (162,165,169-173).

 

By combining donor selection criteria with improved isolation techniques and adequate immunomodulation of the recipient, insulin independence after single donor islet preparation is becoming more reproducibly possible to achieve.  Islet preparations obtained from more than one donor pancreas can be transplanted at once after pooling them, or sequentially based on the metabolic needs of each subject.  Data from the Clinical Islet Transplant Registry and independent trial reports have shown that insulin independence at one year from completion of the transplant is up to 70% with virtually 100% of the subjects maintaining graft function (c-peptide) if adequately immunosuppressed (75,82).  A progressive loss of insulin independence with approximately 90% of subjects requiring reintroduction of exogenous insulin (most of them with detectable c-peptide) has been reported in recent clinical trials based on the ‘Edmonton protocol’ (induction with anti-IL2R antibody; maintenance with sirolimus and tacrolimus) and some variants of it (60,77,84,86,174).  More recent trials using more potent lymphodepletion (i.e., thymoglobulin, anti-CD3 or anti-CD52 antibodies) and/or biologics (anti-IL2R, anti-TNF, anti-LFA-1 antibody or CTLA4Ig) have shown great promise with approximately 50% of insulin independence at 5 years after islet transplantation (86,175-179), which is comparable to some of the data in whole pancreas transplantation in subjects with Type 1 Diabetes (80,83,86,91,92). In light of the results of the last decade of clinical islet transplant trials, achievement of insulin independence, although desirable, no longer should be considered the main goal of islet transplantation.  The sizable improvement of metabolic control in the absence of severe hypoglycemic events, the amelioration of diabetes complications and the achievement of sustained better quality of life, which are quite cumbersome to reproduce by the means of medical treatment, justify the risks associated with the islet transplant procedure and immunosuppression in this high-risk population of subjects with unstable diabetes.

 

Regarding auto transplantation the largest published series are from the University of Minnesota (16-19), University of Cincinnati (20,21), and Leicester (22-25,180). Overall, one-third of patients in the Minnesota series achieves insulin independence, and the majority have islet graft function, as documented by C-peptide positivity (16,22). Cincinnati, Leicester, and other centers have published similar results, with 22-40% of the patients being insulin independent after islet transplant (21,181,182). A significant association between insulin independence and the IEQ/kg transplanted (i.e., islet mass standardized by patient’s weight) was described. Bellin et al. (19) and White et al. (24) reported that insulin independence is related to the number of transplanted islet cells (>2,000 IEQ/kg and >3,000 IEQ/kg, respectively). Similarly, Sutherland et al. (183) reported that insulin independence at 1 year was observed in 63 % of the patients who received greater than 5,000 IE/kg. Moreover, pancreatectomy recipients benefit from an islet autograft ways apart from insulin independence. In fact, the major goal of IAT in these patients is a good glycemic control without brittle diabetes. Ninety percent of patients in the Minnesota series and 100% of those in the Leicester series were C-peptide positive after the procedure (16,22).  The majority of patients receiving an islet auto transplant maintained good glycemic control, with 82% of all recipients having average HbA1c levels <7.0% (16).

 

IMPACT OF ISLET TRANSPLANTATION ON DIABETES COMPLICATIONS

 

Encouraging results have been reported in recent years on the multiple beneficial effects of islet transplantation on the progression of diabetes complications [reviewed in (184)].  Although based on nonrandomized pilot studies, which should be cautiously evaluated, they provide the proof of concept of the importance of restoring beta-cell function in patients with diabetes.  In particular, improvement of micro- and macro-angiopathy (main causes of diabetic nephropathy) (79,101,102,154,157,163-168) and stabilization/reduced progression of retinopathy (104,157-159) and neuropathy (158,160-162) have been described.  Amelioration of cardiovascular and endothelial function, reduction of atherothrombotic profile paralleled by reduced incidence of cardiovascular accidents and higher survival rates were reported In IAK recipients (169-172) (162,165,169,171,173).  Furthermore, significantly improved longevity of a renal transplant was observed after islet transplantation (165).  It is likely that these benefits are the consequence of improve metabolic control conferred by the islet transplant.  In addition, it has been proposed a contribution of restored c-peptide secretion and its effects on multiple targets (185).

 

Table 6.  Benefits of Islet Transplantation

Metabolic control

-                Reduction of exogenous insulin requirements or insulin independence

-                Reduction of MAGE

-                Reduction or normalization of A1c

-                Absence of severe hypoglycemia

Quality of Life

-                Reduced fear of hypoglycemia

-                Improvement of Diabetes Quality of Life

Diabetes complications

-                Improvement of micro- and micro-angiopathy

-                Improvement of cardiovascular and endothelial function

-                Reduced incidence of acute cardiovascular events

-                Reduced nephropathy progression

-                Stabilization/slower neuropathy progression

-                Stabilization/slower retinopathy progression

 

COMMON ADVERSE EVENTS AND THEIR MANAGEMENT (TABLE 7)

The procedure of islet transplantation has proven to be very safe, especially when compared with whole pancreas transplant (177,186,187).  For allogenic islet transplantation bleeding, either intraperitoneal or liver subcapsular, is the most common procedure-related complication, occurring with an incidence as high as 13%  (188). The use of fibrin tissue sealant and embolization coils in the hepatic catheter tract seems to effectively minimize the bleeding risk (188,189). Partial portal vein thrombosis complicates fewer than 5% of islet infusion procedures (174), and complete portal venous thrombosis is rare. The use of purer islet preparations, greater expertise in portal vein catheterization and new radiological devices (catheters medicated with anticoagulation) will continue reducing the risk of portal vein thrombosis, although the risk is unlikely be completely eliminated. Other complications of islet cell transplantation include transient liver enzyme elevation (50% incidence) (106), abdominal pain (50% incidence), focal hepatic steatosis (20% incidence) (190,191), and severe hypoglycemia (< 3% incidence). Another complication related to the intrahepatic islet transplantation procedure is portal hypertension that can occur acutely during the islet infusion, especially in the case of infusions other than the first one (192). Finally, severe hypoglycemia is a risk associated with the infusion of islets. Iatrogenic hypoglycemia in the immediate post-transplant period is a rare event.  Frequent blood glucose monitoring immediately following islet transplantation is recommended to avoid severe unrecognized hypoglycemia in the early post-transplant period. The risk of transmission of CMV disease from donor to recipient has been surprisingly low in recipients of islet allografts, particularly in the most recent period with routine use of purified islet preparations (140-144). As with any allogeneic transplant, islet transplant recipients may become sensitized to islet donor histocompatibility antigens (HLA), leading to the development of panel reactive alloantibodies (PRA).Data on the development of cytotoxic antibodies against donor HLA in islet allotransplant recipients with failing grafts have been reported from several islet transplant centers (148-152). A potential consequence of high PRA levels in recipients of a failed islet transplants is that if these individuals develop diabetic nephropathy in the future, a high PRA may increase their time on a transplant list for a suitable kidney graft.

 

The need to implement anti-rejection therapy exposes transplant recipients to an increased risk of untoward side effects expected in any immunosuppressed subjects (Table 6) (107). Opportunistic infections of urinary tract, upper respiratory tract and skin are frequent, along with myelosuppressive and gastrointestinal effects of the immunosuppressive drugs.  In the majority of the cases, these effects are not severe and resolve without sequel with medical treatment. Elevation of viremic titers for cytomegalovirus (CMV) or Epstein-Barr virus (EBV) in the presence of overt clinical symptoms (i.e., de novo infection or reactivation in seropositve subjects) imposes the implementation of anti-viral therapy and reduction of immunosuppressive drug dose (98). Timely intervention may result in faster resolution of the symptoms without compromising graft survival. Direct organ toxicity of immunosuppressive drugs has been recognized. Symptoms associated with neuro- and/or nephro-toxicity are relatively frequent in subjects receiving chronic immunosuppressive agents currently in use in the clinical arena.   In these cases, modification of the anti-rejection regimen is indicated, with dose reduction or conversion to a different combination of drugs.  In the majority of cases, these changes resolve the symptoms without compromising graft survival (193,194). Nephrotoxicity from sirolimus and/or tacrolimus has been described in patients with T1D undergoing islet transplantation, particularly when kidney function is already impaired because of pre-existing diabetic nephropathy (195,196).

 

As for the CITR 11th Allograft Data Report Scientific Summary, the decline in eGFR (CKD-Epi) after islet transplantation is both statistically significant and clinically important. IAK had much lower pre-transplant levels than ITA, which then declined at a slower rate. Initial levels were also lower in recipients age 35 and older and declined at a slower rate compared to younger recipients. Levels were generally lower among recipients managed with CNI+IMPDH compared to other maintenance immunosuppression regimens. Compared with an age-unadjusted cohort of 1,141 participants with T1D followed by the Diabetes Control and Complications Trial and then by the Epidemiology of Diabetes Interventions and Complications (EDIC) (The DCCT/EDIC Research Group, 2011) who started with mean eGFR levels of 126 ml/min/1.73m3, CITR allograft recipients had much lower mean eGFR (91±1SE for ITA and 62±2 for IAK) at their first transplant. CITR ITA recipients exhibited a decline in eGFR of 12 ml/min/1.73m3 and IAK experienced a mean decline of 2 ml/min/1.73m3 in 5 years from last infusion, compared to a mean decline of about 9 ml/min/1.73m3 over the first 5 years in the DCCT.

 

As of 2021, by decision of the Executive Committee, only serious adverse events (SAEs) are reportable to CITR. About 11% ITA and 14% of IAK allo-islet recipients experienced a serious adverse event in the first 30 days following transplantation. There was a sharp decline in the number of patients who experienced SAEs post-2010, with 15% or more of patients experiencing SAEs in early eras compared to ~5% in 2011-2014 and 2015-2018. In the first year after islet transplantation, which includes a majority of the reinfusions that were performed, about one-fourth of participants have experienced an SAE. SAE within 1-year was slightly more common in IAK (31%) than ITA (23%) and there was a significant decline post-2010 (>25% pre vs. <15% post). Life-threatening events have occurred in 13.4% of islet-alone, in 16.5% of IAK recipients, and in 20.4% of SIK recipients. Recent eras have seen a substantial decline in the incidence of life-threatening events. The most common life-threatening events reported were abnormal granulocytes (24 events) followed by abnormal liver function (23 events) and hypoglycemia (14 events). About 75% of patients who experienced a life-threatening event recovered fully, 12% recovered with sequelae, 5% did not recover, and 9% died as a result of the event.

 

A total of 189 instances of neoplasm have been diagnosed in 101 of the 1,399 islet recipients who collectively represent a total of 7,963 person-years of observed follow-up. This equates to about 0.02 neoplasms per person-year. Of the total 189 events, 61% were deemed possibly related to immunosuppression, and 12% definitely related. Of the total events, 69% recovered, 10% did not recover, 5% recovered with sequelae, and 3% resulted in fatality. There were 41 instances in 23 patients of basal carcinoma of the skin and 86 instances in 38 patients of squamous carcinoma of the skin. There were 56 instances in 39 recipients of non-skin cancers. Eleven deaths due to cancer occurred.

 

There have been 77 or 5.5% deaths; cumulative mortality rates differed significantly by transplant type (p<0.0001) but not by era. SIK transplant recipients were disproportionately represented among fatalities comprising only 3.5% of the allo-islet recipient population, but 15.6% percent of deaths. Of the reported deaths, ten were deemed possibly related or definitely related to islet transplantation or immunosuppression. The most common causes of death were (# cases): cardiovascular (15), neoplasm (11), infection (including pneumonia) (9), hemorrhage (4), and complications of diabetes (3). Twenty-four deaths did not have a cause specified.

 

An assessment of the surgical complication of islet auto transplantation was recently reported for the entire Minnesota series (n=413) (16). Surgical complications requiring reoperation during the initial admission occurred in 15.9% of the patients. The most common reason for reoperation was bleeding, occurring in 9.5% of the procedures. Anastomotic leaks occurred in 4.2 % of the patients, biliary in 1.4% and enteric in 2.8%. Intra-abdominal infection requiring reoperation occurred in 1.9% of patients, wound infections requiring operative debridement in 2.2%. Gastrointestinal issues, such as bowel obstruction, omental infarction, bowel ischemia, delayed reconstruction because of bowel edema, tube perforation, requiring reoperation in 4.7% of the patients. Two patients (<1%) required reoperation to remove an ischemic or bleeding spleen after spleen sparing pancreatectomy (done in 30% of patients).

 

Table 7.  Most Frequent Complications in Islet Transplant Recipients

Procedure-related

-                Hemorrhage

-                Portal thrombosis

-                Transient transaminitis

Immunosuppression-related

Hematological

-                Anemia

-                Leucopenia

-                Neutropenia

Metabolic

-                Dyslipidemia

Gastrointestinal

-                Oral ulcers (Sirolimus)

-                Diarrhea (Mycophenolic acid)

-                CMV colitis

Respiratory tract

-                Upper respiratory infections

-                Interstitial pneumonitis (Sirolimus)

Neurological

-                Neurotoxicity (Tacrolimus)

Genitourinary

-                Urinary infections

-                Ovarian cysts

-                Dysmenorrhea

-                Nephropathy

-                Proteinuria

Cutaneous

-                Infections

-                Cancer

 

CURRENT CHALLENGES

 

There are many challenges that are currently limiting islet cell transplantation (Table 8) (197-199)  While significant progress has been made in the islet transplantation field, several obstacles remain precluding its widespread use. The clinical experience of islet transplantation has been developed almost exclusively using the intra-hepatic infusion through the portal vein (60). It has been suggested that the loss of as many as 50-75% of islets during engraftment is the reason why a very large number of islets are needed to achieve normoglycemia (51,72). Moreover, two additional important limitations are the currently inadequate immunosuppression for preventing islet rejection (70) and the limited oxygen supply to islet in the engraftment site (200,201). Current immunosuppressive regimens are capable of preventing islet failure for months to years, but the agents used in these treatments may increase the risk for specific malignancies and opportunistic infections. In addition, the most commonly used agents (like calcineurin inhibitors and rapamycin) are also known to impair normal islet function and/or insulin action. Furthermore, like all medications, these agents have other associated toxicities, including the harmful effect of certain widely employed immunosuppressive agents on renal function. The second very significant factor for early and late loss of islet mass is the critical lack of immediate vascularization and chronic hypo-oxygenation. Physiological supply of oxygen and nutrients in native islets is maintained by a tight capillary network, destroyed by the islet isolation procedure, restricting supply to diffusion from the portal vein and hepatic arterial capillaries until the revascularization process is completed. Oxygen tension in the liver parenchyma decreases from approximately 40 to 5 mmHg, eight-fold lower compared to the intra-pancreatic levels, leading to severe hypoxia, and β-cell death. Revascularization of the islet graft in rodent transplant requires 10-14 days and much longer in non-human primates and human recipients. Even after the revascularization of the islets is completed, the capillary’ density is significantly lower compared to the physiological intra-pancreatic situation. Finally, one of the main challenges is the cost of the procedure and some regulatory issues, as recently demonstrated by the ongoing discussion in USA (202). In fact, on April 15, the FDA's Cellular, Tissue, and Gene Therapies Advisory Committee voted in favor of approval of the biologics license application (BLA) seeking to market allogeneic islets of Langerhans for the treatment of ‘brittle” type I diabetes mellitus in adults whose symptoms are not well controlled despite intensive insulin therapy.  The FDA endorsement of islet transplantation adds to the list of national agencies in Europe, such as the Federal Office of Public Health in Switzerland, the National Health Service (NHS) in the UK, the Swedish Local Authorities and Regions, the Ministry of Health in Poland and Belgium and, more recently, the French National Authority for Health (HAS) in France that have approved islet transplantation as a reimbursed standard-of-care procedure. Unfortunately, the FDA has chosen to consider islets as a biologic that requires licensure, making the universal implementation of the procedure in the clinic very challenging.

 

Table 8. Current Challenges Faced for Islet Transplantation

Challenge

Possible impact

Potential solutions

Progressive graft dysfunction

Reintroduction of exogenous insulin;

Destabilization of metabolic control;

Supplemental islet transplant.

Incretin mimetics;

Alternative islet implantation sites;

Novel immunosuppressive protocols.

Multiple islet donors

Increased operational costs;

Shortage of deceased donor pancreata for transplantation;

Risk of allosensitization.

Improved donor selection criteria;

Optimized cell processing;

Alternative sources of transplantable tissue (i.e., stem cells-derived or xenogeneic islets;

Alternative implantation sites.

Chronic immunosuppression

Systemic toxicity;

Increased risk of opportunistic infections;

Islet cell toxicity.

Use of biologics;

Immune isolation techniques; Development of immune tolerance inducing protocols.

Allosensitization

Reduced graft survival;

Preclude/worsen outcome of subsequent transplantation (i.e., islet or renal)

Maximizing the success rate of single donor islet transplantation;

Alternative sources of transplantable tissue;

Immune isolation;

Plasmapheresis / depletion of alloantibodies;

Novel immunosuppressive protocols;

Development of immune tolerance inducing protocols.

Cumbersome graft monitoring

Mainly rely on metabolic function tests, but cannot discriminate between immunological and metabolic causes of dysfunction;

Liver needle biopsies do not provide adequate graft tissue;

MRI and PET lack the resolution to detect islets scattered throughout the liver.

Improved simple metabolic measures predictive of graft dysfunction;

Improved sensitivity of noninvasive imaging techniques (functional MRI?);

Improved immune monitoring techniques for early detection of immune events able to discriminate between rejection and autoimmunity.

 

FUTURE DEVELOPMENTS IN BETA-CELL REPLACEMENT THERAPIES

 

The field of cellular therapies for the treatment of diabetes is rapidly evolving and a new exciting era has already begun (shown in Fig. 1). Efforts are ongoing to push to a broader dimension islet transplantation (143), including: (i) implementation of a scheme for donor and recipient selection and organ allocation to increase pancreas utilization (203-205); (ii) improvement and standardization of islet isolation process and its best codification by regulatory bodies  (206-209) (iii) monitoring of transplanted islets by noninvasive imaging techniques (210,211); (iv) development of biomarkers to assess the efficacy of the immunosuppression/immunomodulation strategies (69,212-216); (v) identification of alternative transplantation sites (51); (vi) creation of  an ideal bio-artificial niche for islet survival by bioengineering approaches (217,218) and (vii) use of immune-isolation techniques, such as hydrogel polymers that shield pancreatic islet from immune cell attack (219,220). On the other hand, there is increasing new excitement for the use of unlimited alternative sources of transplantable islets, such as xenogeneic (i.e., obtained from other species such as porcine islets) [reviewed in (221)] or derived from human stem cells (222-227).  Pig islets may be available in plentiful amounts.  Importantly, the ability to obtain genetically modified pigs that lack or overexpress specific molecules may be of assistance in developing cellular products with reduced immunogenicity for transplantation into humans.  In turn, this technology may allow achieving long-term function under immunosuppressive regimens that are used for allogeneic cells or facilitating the induction of long-term acceptance of xenogeneic islet cells.  Another area reporting great progress is that of regenerative medicine using human stem cells from embryonic or adult sources. 

 

While adult stem cells, such as mesenchymal stem cells, have an immunomodulatory potential when infused at disease onset (228) (229) or as adjuvants to improve the outcomes of islet transplantation (230), the greatest enthusiasm lies in the possibility to use pluripotent stem cells to overcome the limits of islet transplantation (231,232). Human pluripotent stem cells (both embryonic stem [ES] and induced pluripotent stem [iPS] cells), are the best candidate for making β cells as they have unlimited potential for division and differentiation. Efficient protocols for the differentiation of pluripotent cells into β cells have been developed by several laboratories (233-241) and a great effort in the last year was concentrated on developing cellular products with consistent potency and safety profile for clinical application. Actually, six clinical trials using human pluripotent stem cells for the therapy of type 1 diabetes are registered in ClinicalTrial.gov: three active and recruiting (NCT03163511; NCT04678557; NCT04786262), one completed (NCT03162926), one enrolling by invitation (NCT02939118) and one active but not recruiting (NCT02239354). All these trials, except the NCT04786262, are using PEC-01 cells as a cell product. PEC-01 cells are a mixed cell population comprising pancreatic endoderm and poly-hormonal endocrine cells (233,242,243)differentiated by a pluripotent stem cell line called CyT49 (225).  These pancreatic precursor cells are fully committed to further differentiate into mature endocrine pancreatic cells after their implantation and were tested within an encapsulation device in subcutaneous space.  In December 2021, the interim results of some of these clinical trial appeared in two articles (244) . Over the follow-up period, which lasted up to 1 year, patients had 20% reduced insulin requirements, spent 13% more time in target blood glucose range, had stable average HbA1c <7.0%, had improved hypoglycemic awareness (average Clarke score decreased ∼1 point) associated with C-peptide levels that were, on average, ∼1/100th normal levels. Explanted grafts contained heterogeneous composition of pancreatic cells, including cells with mature β cell phenotype.  In both the papers: (i) induction and maintenance immunosuppression appeared to be effective in preventing allogeneic and autoimmune destruction of the graft cells, (ii) the cell product appeared to be safe and well tolerated, since no teratoma formation was observed and the great majority of mild-to-moderate adverse effects was due to surgical procedure risks and side-effects of immunosuppression. These initial data reinforce the hope that pluripotent stem cells, differentiated into pancreatic endocrine cells, may be a renewable source of β cells for patients with T1D.

 

VX-880 is a second cell product approved in 2021 as investigational cell therapy for the treatment of type 1 diabetes. VX-880 consists of fully differentiated insulin-producing pancreatic islet cells obtained from pluripotent stem cells. A Phase 1/2, single-arm, open-label clinical trial was recently approved in patients who have T1D with impaired hypoglycemic awareness and severe hypoglycemia. VX-880 is infused in the portal vein and a chronic administration of concomitant immunosuppressive therapy is be required to protect the islet cells from immune rejection. Some preliminary results have already been shared in a press release in May 2022 and suggest that β cells fully differentiated from stem cells and transplanted into the liver may engraft and start secreting insulin early after infusion. In addition to ongoing clinical experiences, others commercial or academic organizations have announced their intention to conduct clinical trials of functional stem cell derived-islets (135). At this point, the need to shield the transplanted stem-cell-derived β-cells from immune rejection becomes more and more critical. In this direction, different strategies to reduce or avoid immune rejection are under evaluation (245) including (i) generation of universally compatible pluripotent stem cells by silencing or deleting HLA or genes essential for HLA expression or function and by expressing genes encoding immunosuppressive molecules (246), (ii) development of mild immunosuppressive regimens (e.g., monoclonal antibodies targeting NK cells and/or T cell subsets) sufficient to induce tolerance, (iii) improvement in encapsulation/containment of cell product and (iv) creation of a haplobank of stem cell lines (247). 

 

A current limitation for islet transplantation is the inability to use non- or minimally-invasive predictive tests as well as biomarkers of early graft dysfunction to guide timely interventions aimed at preserving functional islet cell mass.  Metabolic tests (i.e., glycemic control, insulin requirement, HbA1c, basal and stimulated c-peptide) remain the main indicators of graft function, the alteration of which may indicate underlying distress of the graft but cannot discriminate possible causes such as metabolic overload, immunity, or drug toxicity.  In some cases, graft dysfunction may be reversible (i.e., transient metabolic overload due to an infection episode), but in many other cases at the time graft dysfunction is detected, a considerable mass of functional beta cells might already be irreversibly lost. Monitoring of transplanted islets by noninvasive imaging techniques (such as MRI, PET-CT, and US) is cumbersome, as they lack the resolution for the detection of cellular clusters the size of islets (~50-900um) that are scattered throughout the recipient’s liver [reviewed in (248)].  While encouraging preliminary studies have shown that preloading of aliquots of the islet graft with iron nanoparticles (for MRI) (249-253) or labeled glucose (for PET-CT) (254-257) can be used safely, these techniques do not allow assessing the whole mass of transplanted clusters and provide only passive and transient information on islet distribution in the transplant site.  The progress in the field of functional MRI (fMRI) and toward the development of more sensitive beta-cell specific imaging techniques may allow a more objective assessment of islet cell mass over time in a near future. Detection of biomarkers [reviewed in (109)] in blood samples to determine immune cell function (i.e., cell surface expression of specific markers by flow cytometry and cytotoxic lymphocyte gene expression profiles, amongst other)(258-260) and autoimmunity reactivation (namely, autoantibody titers) is evaluated in ongoing clinical trials to identify means of assessing the efficacy of the immunomodulation strategies, detecting rejection episodes and reactivation of autoimmunity early enough to implement timely immune interventions to prevent graft loss (69,70,261-263).  Unfortunately, some of the current tests lack adequate specificity as they may be affected also with underlying infections.  With the rapid evolution of high throughput arrays, it is likely that new and more specific molecular biomarkers of islet cell distress and immune cell function will become available in the near future. Alternative transplantation sites [reviewed in (51)] are being currently explored that may contribute enhancing islet engraftment and attain sustained graft function long-term (52).  Importantly, alternative sites may be modified using bioengineering approaches that could enable creating an ideal bio artificial endocrine pancreas [reviewed in (264)].  The use of immunoisolation techniques, such as using hydrogel polymers that shield islet cell clusters from immune cell attack, may contribute to achieve sustained function of transplanted cells without the need for life-long immunosuppression [reviewed in (265) and (264)].

 

CONCLUSIONS

In conclusion, islet transplantation as it is today cannot be the universal cure for type 1 diabetes. It represents a clinical option in few highly selected patients but it is the proof of principle that it is possible to replace efficiently β cells in patients with diabetes by a cell therapy. Restoration of physiologic metabolic control in patients with diabetes is highly desirable. Transplantation of islets of Langerhans allows the achievement of stable metabolic control in the most severe manifestations that cannot be matched with conventional medical therapies.  The steady progress of clinical islet transplantation and the promising emerging new approaches that address immunity and beta cell sources justifies cautious optimism for the potential applicable of beta-cell replacement to all cases of insulin-dependent diabetes in the near future.

 

ACKNOWLEDGMENTS

This work was partially supported by the Italian Minister of Health (Ricerca Finalizzata RF-2009-1483387, RF-2009-1469691), Ministry of EducationUniversity and Research (PRIN 2008, prot. 2008AFA7LC), Associazione Italiana per la Ricerca sul Cancro (AIRC, bando 5 x 1,000 N_12182 and Progetto IGN_ 11783), EU (HEALTH-F5-2009-241883-BetaCellTherapy) and the Juvenile Diabetes Research Foundation International.

 

The author alone is responsible for reporting and interpreting these data; the views expressed herein are those of the author and not necessarily those of the funding agencies.

 

ONLINE RESOURCES ON THE SUBJECT

 

Clinical Islet Transplant Consortium; Collaborative Islet Transplant Registry; Diabetes Research Institute Foundation; Health Resources and Services Administration; International Pancreas & Islet Transplant Association; The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK); Organ Procurement and Transplantation Network; The Cell Transplant Society; Scientific Registry of Transplant Recipients; United States Department of Health and Human Services; United Network For Organ Sharing (UNOS).

 

 

American Diabetes Association; American Society of Transplantation; American Society of Transplant Surgeons; Beta Cell Biology Consortium; European Pancreas Club; European Society for Organ Transplantation; International Pancreas Transplant Registry; International Xenotransplantation Association; Juvenile Diabetes Research Foundation.

 

REFERENCES

 

  1. Gregg EW, Sattar N, Ali MK. The changing face of diabetes complications. Lancet Diabetes Endocrinol.2016;4(6):537-547.
  2. Harding JL, Pavkov ME, Magliano DJ, Shaw JE, Gregg EW. Global trends in diabetes complications: a review of current evidence. Diabetologia. 2019;62(1):3-16.
  3. Mannucci E, Monami M, Dicembrini I, Piselli A, Porta M. Achieving HbA1c targets in clinical trials and in the real world: a systematic review and meta-analysis. J Endocrinol Invest. 2014;37(5):477-495.
  4. Edelman SV, Polonsky WH. Type 2 Diabetes in the Real World: The Elusive Nature of Glycemic Control. Diabetes Care. 2017;40(11):1425-1432.
  5. Rodbard D. State of Type 1 Diabetes Care in the United States in 2016-2018 from T1D Exchange Registry Data. Diabetes Technol Ther. 2019;21(2):62-65.
  6. Livingstone SJ, Levin D, Looker HC, Lindsay RS, Wild SH, Joss N, Leese G, Leslie P, McCrimmon RJ, Metcalfe W, McKnight JA, Morris AD, Pearson DW, Petrie JR, Philip S, Sattar NA, Traynor JP, Colhoun HM. Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008-2010. Jama. 2015;313(1):37-44.
  7. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371(21):1972-1982.
  8. Alva ML, Hoerger TJ, Zhang P, Cheng YJ. State-level diabetes-attributable mortality and years of life lost in the United States. Ann Epidemiol. 2018;28(11):790-795.
  9. McKnight JA, Wild SH, Lamb MJ, Cooper MN, Jones TW, Davis EA, Hofer S, Fritsch M, Schober E, Svensson J, Almdal T, Young R, Warner JT, Delemer B, Souchon PF, Holl RW, Karges W, Kieninger DM, Tigas S, Bargiota A, Sampanis C, Cherubini V, Gesuita R, Strele I, Pildava S, Coppell KJ, Magee G, Cooper JG, Dinneen SF, Eeg-Olofsson K, Svensson AM, Gudbjornsdottir S, Veeze H, Aanstoot HJ, Khalangot M, Tamborlane WV, Miller KM. Glycaemic control of Type 1 diabetes in clinical practice early in the 21st century: an international comparison. Diabet Med. 2015;32(8):1036-1050.
  10. Gregg EW, Li Y, Wang J, Burrows NR, Ali MK, Rolka D, Williams DE, Geiss L. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med. 2014;370(16):1514-1523.
  11. Marzorati S, Pileggi A, Ricordi C. Allogeneic islet transplantation. Expert Opin Biol Ther. 2007;7(11):1627-1645.
  12. Piemonti L PA. 25 Years of the Ricordi Automated Method for Islet Isolation. CellR4. 2013;1(1):e128.
  13. Tzakis AG, Ricordi C, Alejandro R, Zeng Y, Fung JJ, Todo S, Demetris AJ, Mintz DH, Starzl TE. Pancreatic islet transplantation after upper abdominal exenteration and liver replacement. Lancet. 1990;336(8712):402-405.
  14. Najarian JS, Sutherland DE, Matas AJ, Goetz FC. Human islet autotransplantation following pancreatectomy. Transplant Proc. 1979;11(1):336-340.
  15. Bramis K, Gordon-Weeks AN, Friend PJ, Bastin E, Burls A, Silva MA, Dennison AR. Systematic review of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Br J Surg. 2012;99(6):761-766.
  16. Sutherland DE, Radosevich DM, Bellin MD, Hering BJ, Beilman GJ, Dunn TB, Chinnakotla S, Vickers SM, Bland B, Balamurugan AN, Freeman ML, Pruett TL. Total pancreatectomy and islet autotransplantation for chronic pancreatitis. J Am Coll Surg. 2012;214(4):409-424; discussion 424-406.
  17. Bellin MD, Beilman GJ, Dunn TB, Pruett TL, Chinnakotla S, Wilhelm JJ, Ngo A, Radosevich DM, Freeman ML, Schwarzenberg SJ, Balamurugan AN, Hering BJ, Sutherland DE. Islet autotransplantation to preserve beta cell mass in selected patients with chronic pancreatitis and diabetes mellitus undergoing total pancreatectomy. Pancreas. 2013;42(2):317-321.
  18. Bellin MD, Freeman ML, Schwarzenberg SJ, Dunn TB, Beilman GJ, Vickers SM, Chinnakotla S, Balamurugan AN, Hering BJ, Radosevich DM, Moran A, Sutherland DE. Quality of life improves for pediatric patients after total pancreatectomy and islet autotransplant for chronic pancreatitis. Clin Gastroenterol Hepatol. 2011;9(9):793-799.
  19. Bellin MD, Carlson AM, Kobayashi T, Gruessner AC, Hering BJ, Moran A, Sutherland DE. Outcome after pancreatectomy and islet autotransplantation in a pediatric population. J Pediatr Gastroenterol Nutr.2008;47(1):37-44.
  20. Wilson GC, Sutton JM, Salehi M, Schmulewitz N, Smith MT, Kucera S, Choe KA, Brunner JE, Abbott DE, Sussman JJ, Ahmad SA. Surgical outcomes after total pancreatectomy and islet cell autotransplantation in pediatric patients. Surgery. 2013;154(4):777-783; discussion 783-774.
  21. Sutton JM, Schmulewitz N, Sussman JJ, Smith M, Kurland JE, Brunner JE, Salehi M, Choe KA, Ahmad SA. Total pancreatectomy and islet cell autotransplantation as a means of treating patients with genetically linked pancreatitis. Surgery. 2010;148(4):676-685; discussion 685-676.
  22. Webb MA, Illouz SC, Pollard CA, Gregory R, Mayberry JF, Tordoff SG, Bone M, Cordle CJ, Berry DP, Nicholson ML, Musto PP, Dennison AR. Islet auto transplantation following total pancreatectomy: a long-term assessment of graft function. Pancreas. 2008;37(3):282-287.
  23. Clayton HA, Davies JE, Pollard CA, White SA, Musto PP, Dennison AR. Pancreatectomy with islet autotransplantation for the treatment of severe chronic pancreatitis: the first 40 patients at the leicester general hospital. Transplantation. 2003;76(1):92-98.
  24. White SA, Davies JE, Pollard C, Swift SM, Clayton HA, Sutton CD, Weymss-Holden S, Musto PP, Berry DP, Dennison AR. Pancreas resection and islet autotransplantation for end-stage chronic pancreatitis. Ann Surg.2001;233(3):423-431.
  25. Garcea G, Weaver J, Phillips J, Pollard CA, Ilouz SC, Webb MA, Berry DP, Dennison AR. Total pancreatectomy with and without islet cell transplantation for chronic pancreatitis: a series of 85 consecutive patients. Pancreas.2009;38(1):1-7.
  26. Teuscher AU, Kendall DM, Smets YF, Leone JP, Sutherland DE, Robertson RP. Successful islet autotransplantation in humans: functional insulin secretory reserve as an estimate of surviving islet cell mass. Diabetes. 1998;47(3):324-330.
  27. Robertson RP, Lanz KJ, Sutherland DE, Kendall DM. Prevention of diabetes for up to 13 years by autoislet transplantation after pancreatectomy for chronic pancreatitis. Diabetes. 2001;50(1):47-50.
  28. Blondet JJ, Carlson AM, Kobayashi T, Jie T, Bellin M, Hering BJ, Freeman ML, Beilman GJ, Sutherland DE. The role of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Surg Clin North Am.2007;87(6):1477-1501, x.
  29. Bellin MD, Sutherland DE. Pediatric islet autotransplantation: indication, technique, and outcome. Curr Diab Rep. 2010;10(5):326-331.
  30. Bellin MD, Sutherland DE, Beilman GJ, Hong-McAtee I, Balamurugan AN, Hering BJ, Moran A. Similar islet function in islet allotransplant and autotransplant recipients, despite lower islet mass in autotransplants. Transplantation. 2011;91(3):367-372.
  31. Robertson RP. Consequences on beta-cell function and reserve after long-term pancreas transplantation. Diabetes. 2004;53(3):633-644.
  32. Chinnakotla S, Bellin MD, Schwarzenberg SJ, Radosevich DM, Cook M, Dunn TB, Beilman GJ, Freeman ML, Balamurugan AN, Wilhelm J, Bland B, Jimenez-Vega JM, Hering BJ, Vickers SM, Pruett TL, Sutherland DE. Total pancreatectomy and islet autotransplantation in children for chronic pancreatitis: indication, surgical techniques, postoperative management, and long-term outcomes. Ann Surg. 2014;260(1):56-64.
  33. Dong M, Parsaik AK, Erwin PJ, Farnell MB, Murad MH, Kudva YC. Systematic review and meta-analysis: islet autotransplantation after pancreatectomy for minimizing diabetes. Clin Endocrinol (Oxf). 2011;75(6):771-779.
  34. Dudeja V, Beilman GJ, Vickers SM. Total pancreatectomy with islet autotransplantation in patients with malignancy: are we there yet? Ann Surg. 2013;258(2):219-220.
  35. Ris F, Niclauss N, Morel P, Demuylder-Mischler S, Muller Y, Meier R, Genevay M, Bosco D, Berney T. Islet autotransplantation after extended pancreatectomy for focal benign disease of the pancreas. Transplantation.2011;91(8):895-901.
  36. Jin SM, Oh SH, Kim SK, Jung HS, Choi SH, Jang KT, Lee KT, Kim JH, Lee MS, Lee MK, Kim KW. Diabetes-free survival in patients who underwent islet autotransplantation after 50% to 60% distal partial pancreatectomy for benign pancreatic tumors. Transplantation. 2013;95(11):1396-1403.
  37. Balzano G, Nano R, Maffi P, Mercalli A, Melzi R, Aleotti F, Gavazzi F, Berra C, De Cobelli F, Venturini M, Magistretti P, Scavini M, Capretti G, Del Maschio A, Secchi A, Zerbi A, Falconi M, Piemonti L. Salvage Islet Auto Transplantation After Relaparatomy. Transplantation. 2017;101(10):2492-2500.
  38. Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, Zerbi A, De Cobelli F, Gavazzi F, Magistretti P, Scavini M, Peccatori J, Secchi A, Ciceri F, Del Maschio A, Falconi M, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy: A Broader Spectrum of Indications Beyond Chronic Pancreatitis. Am J Transplant. 2016;16(6):1812-1826.
  39. Balzano G, Maffi P, Nano R, Zerbi A, Venturini M, Melzi R, Mercalli A, Magistretti P, Scavini M, Castoldi R, Carvello M, Braga M, Del Maschio A, Secchi A, Staudacher C, Piemonti L. Extending indications for islet autotransplantation in pancreatic surgery. Ann Surg. 2013;258(2):210-218.
  40. Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, De Cobelli F, Magistretti P, Scavini M, Secchi A, Falconi M, Piemonti L. Diabetes-free survival after extended distal pancreatectomy and islet auto transplantation for benign or borderline/malignant lesions of the pancreas. Am J Transplant. 2019;19(3):920-928.
  41. Balzano G, Piemonti L. Autologous islet transplantation in patients requiring pancreatectomy for neoplasm. Curr Diab Rep. 2014;14(8):512.
  42. Balzano G, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy for Neoplasm. Current Diabetes Reports. 2014;14(8):512.
  43. Balzano G, Maffi P, Nano R, Zerbi A, Venturini M, Melzi R, Mercalli A, Magistretti P, Scavini M, Castoldi R, Carvello M, Braga M, Del Maschio A, Secchi A, Staudacher C, Piemonti L. Extending Indications for Islet Autotransplantation in Pancreatic Surgery. Annals of Surgery. 2013;258(2):210-218.
  44. Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, Zerbi A, De Cobelli F, Gavazzi F, Magistretti P, Scavini M, Peccatori J, Secchi A, Ciceri F, Del Maschio A, Falconi M, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy: A Broader Spectrum of Indications Beyond Chronic Pancreatitis. American Journal of Transplantation. 2016;16(6):1812-1826.
  45. Matsumoto S, Okitsu T, Iwanaga Y, Noguchi H, Nagata H, Yonekawa Y, Yamada Y, Nakai Y, Ueda M, Ishii A, Yabunaka E, Shapiro JA, Tanaka K. Insulin independence of unstable diabetic patient after single living donor islet transplantation. Transplant Proc. 2005;37(8):3427-3429.
  46. Matsumoto S, Tanaka K, Strong DM, Reems JA. Efficacy of human islet isolation from the tail section of the pancreas for the possibility of living donor islet transplantation. Transplantation. 2004;78(6):839-843.
  47. Matsumoto I, Shinzeki M, Asari S, Goto T, Shirakawa S, Ajiki T, Fukumoto T, Ku Y. Evaluation of glucose metabolism after distal pancreatectomy according to the donor criteria of the living donor pancreas transplantation guidelines proposed by the Japanese Pancreas and Islet Transplantation Association. Transplant Proc. 2014;46(3):958-962.
  48. Choudhary P, Rickels MR, Senior PA, Vantyghem MC, Maffi P, Kay TW, Keymeulen B, Inagaki N, Saudek F, Lehmann R, Hering BJ. Evidence-informed clinical practice recommendations for treatment of type 1 diabetes complicated by problematic hypoglycemia. Diabetes Care. 2015;38(6):1016-1029.
  49. Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Caicedo A. The unique cytoarchitecture of human pancreatic islets has implications for islet cell function. Proc Natl Acad Sci U S A. 2006;103(7):2334-2339.
  50. Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW. Automated method for isolation of human pancreatic islets. Diabetes. 1988;37(4):413-420.
  51. Cantarelli E, Piemonti L. Alternative transplantation sites for pancreatic islet grafts. Curr Diab Rep.2011;11(5):364-374.
  52. Maffi P, Balzano G, Ponzoni M, Nano R, Sordi V, Melzi R, Mercalli A, Scavini M, Esposito A, Peccatori J, Cantarelli E, Messina C, Bernardi M, Del Maschio A, Staudacher C, Doglioni C, Ciceri F, Secchi A, Piemonti L. Autologous pancreatic islet transplantation in human bone marrow. Diabetes. 2013;62(10):3523-3531.
  53. Maffi P, Nano R, Monti P, Melzi R, Sordi V, Mercalli A, Pellegrini S, Ponzoni M, Peccatori J, Messina C, Nocco A, Cardillo M, Scavini M, Magistretti P, Doglioni C, Ciceri F, Bloem SJ, Roep BO, Secchi A, Piemonti L. Islet Allotransplantation in the Bone Marrow of Patients With Type 1 Diabetes: A Pilot Randomized Trial. Transplantation. 2019;103(4):839-851.
  54. Yasunami Y, Nakafusa Y, Nitta N, Nakamura M, Goto M, Ono J, Taniguchi M. A Novel Subcutaneous Site of Islet Transplantation Superior to the Liver. Transplantation. 2018;102(6):945-952.
  55. Wszola M, Berman A, Ostaszewska A, Gorski L, Serwanska-Swietek M, Gozdowska J, Bednarska K, Krajewska M, Lipinska A, Chmura A, Kwiatkowski A. Islets Allotransplantation Into Gastric Submucosa in a Patient with Portal Hypertension: 4-year Follow-up. Transplant Proc. 2018;50(6):1910-1913.
  56. Berman DM, Molano RD, Fotino C, Ulissi U, Gimeno J, Mendez AJ, Kenyon NM, Kenyon NS, Andrews DM, Ricordi C, Pileggi A. Bioengineering the Endocrine Pancreas: Intraomental Islet Transplantation Within a Biologic Resorbable Scaffold. Diabetes. 2016;65(5):1350-1361.
  57. Baidal DA, Ricordi C, Berman DM, Alvarez A, Padilla N, Ciancio G, Linetsky E, Pileggi A, Alejandro R. Bioengineering of an Intraabdominal Endocrine Pancreas. N Engl J Med. 2017;376(19):1887-1889.
  58. Christoffersson G, Henriksnas J, Johansson L, Rolny C, Ahlstrom H, Caballero-Corbalan J, Segersvard R, Permert J, Korsgren O, Carlsson PO, Phillipson M. Clinical and experimental pancreatic islet transplantation to striated muscle: establishment of a vascular system similar to that in native islets. Diabetes. 2010;59(10):2569-2578.
  59. Bertuzzi F, Colussi G, Lauterio A, De Carlis L. Intramuscular islet allotransplantation in type 1 diabetes mellitus. Eur Rev Med Pharmacol Sci. 2018;22(6):1731-1736.
  60. Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006;355(13):1318-1330.
  61. Benhamou PY, Oberholzer J, Toso C, Kessler L, Penfornis A, Bayle F, Thivolet C, Martin X, Ris F, Badet L, Colin C, Morel P. Human islet transplantation network for the treatment of Type I diabetes: first data from the Swiss-French GRAGIL consortium (1999-2000). Groupe de Recherche Rhin Rhjne Alpes Geneve pour la transplantation d'Ilots de Langerhans. Diabetologia. 2001;44(7):859-864.
  62. Goss JA, Goodpastor SE, Brunicardi FC, Barth MH, Soltes GD, Garber AJ, Hamilton DJ, Alejandro R, Ricordi C. Development of a human pancreatic islet-transplant program through a collaborative relationship with a remote islet-isolation center. Transplantation. 2004;77(3):462-466.
  63. Goss JA, Schock AP, Brunicardi FC, Goodpastor SE, Garber AJ, Soltes G, Barth M, Froud T, Alejandro R, Ricordi C. Achievement of insulin independence in three consecutive type-1 diabetic patients via pancreatic islet transplantation using islets isolated at a remote islet isolation center. Transplantation. 2002;74(12):1761-1766.
  64. Lablanche S, Borot S, Wojtusciszyn A, Bayle F, Tetaz R, Badet L, Thivolet C, Morelon E, Frimat L, Penfornis A, Kessler L, Brault C, Colin C, Tauveron I, Bosco D, Berney T, Benhamou PY. Five-Year Metabolic, Functional, and Safety Results of Patients With Type 1 Diabetes Transplanted With Allogenic Islets Within the Swiss-French GRAGIL Network. Diabetes Care. 2015;38(9):1714-1722.
  65. Berney T, Andres A, Bellin MD, de Koning EJ, Johnson PR, Kay TW, Lundgren T, Rickels MR, Scholz H, Stock PG. A Worldwide Survey of Activities and Practices in Clinical Islet of Langerhans Transplantation. Transplant International. 2022:135.
  66. Blood N. Transplant. Organ and tissue donation and transplantation: activity report 2020/21. 2021. 2022.
  67. Webster AC, Hedley JA, Anderson PF, Hawthorne WJ, Radford T, Drogemuller C, Rogers N, Goodman D, Lee MH, Loudovaris T. Australia and New Zealand islet and pancreas transplant registry annual report 2019: islet donations, islet isolations, and islet transplants. Transplantation direct. 2020;6(7).
  68. Campbell PM, Salam A, Ryan EA, Senior P, Paty BW, Bigam D, McCready T, Halpin A, Imes S, Al Saif F, Lakey JR, Shapiro AM. Pretransplant HLA antibodies are associated with reduced graft survival after clinical islet transplantation. Am J Transplant. 2007;7(5):1242-1248.
  69. Hilbrands R, Huurman VA, Gillard P, Velthuis JH, De Waele M, Mathieu C, Kaufman L, Pipeleers-Marichal M, Ling Z, Movahedi B, Jacobs-Tulleneers-Thevissen D, Monbaliu D, Ysebaert D, Gorus FK, Roep BO, Pipeleers DG, Keymeulen B. Differences in baseline lymphocyte counts and autoreactivity are associated with differences in outcome of islet cell transplantation in type 1 diabetic patients. Diabetes. 2009;58(10):2267-2276.
  70. Piemonti L, Everly MJ, Maffi P, Scavini M, Poli F, Nano R, Cardillo M, Melzi R, Mercalli A, Sordi V, Lampasona V, Espadas de Arias A, Scalamogna M, Bosi E, Bonifacio E, Secchi A, Terasaki PI. Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes.2013;62(5):1656-1664.
  71. Citro A, Cantarelli E, Maffi P, Nano R, Melzi R, Mercalli A, Dugnani E, Sordi V, Magistretti P, Daffonchio L, Ruffini PA, Allegretti M, Secchi A, Bonifacio E, Piemonti L. CXCR1/2 inhibition enhances pancreatic islet survival after transplantation. J Clin Invest. 2012;122(10):3647-3651.
  72. Citro A, Cantarelli E, Piemonti L. Anti-inflammatory strategies to enhance islet engraftment and survival. Curr Diab Rep. 2013;13(5):733-744.
  73. Moberg L, Johansson H, Lukinius A, Berne C, Foss A, Kallen R, Ostraat O, Salmela K, Tibell A, Tufveson G, Elgue G, Nilsson Ekdahl K, Korsgren O, Nilsson B. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet. 2002;360(9350):2039-2045.
  74. Matsuoka N, Itoh T, Watarai H, Sekine-Kondo E, Nagata N, Okamoto K, Mera T, Yamamoto H, Yamada S, Maruyama I, Taniguchi M, Yasunami Y. High-mobility group box 1 is involved in the initial events of early loss of transplanted islets in mice. J Clin Invest. 2010;120(3):735-743.
  75. Barton FB, Rickels MR, Alejandro R, Hering BJ, Wease S, Naziruddin B, Oberholzer J, Odorico JS, Garfinkel MR, Levy M, Pattou F, Berney T, Secchi A, Messinger S, Senior PA, Maffi P, Posselt A, Stock PG, Kaufman DB, Luo X, Kandeel F, Cagliero E, Turgeon NA, Witkowski P, Naji A, O'Connell PJ, Greenbaum C, Kudva YC, Brayman KL, Aull MJ, Larsen C, Kay TW, Fernandez LA, Vantyghem MC, Bellin M, Shapiro AM. Improvement in outcomes of clinical islet transplantation: 1999-2010. Diabetes Care. 2012;35(7):1436-1445.
  76. Pileggi A, Ricordi C, Kenyon NS, Froud T, Baidal DA, Kahn A, Selvaggi G, Alejandro R. Twenty years of clinical islet transplantation at the Diabetes Research Institute--University of Miami. Clin Transpl. 2004:177-204.
  77. Hering BJ, Kandaswamy R, Ansite JD, Eckman PM, Nakano M, Sawada T, Matsumoto I, Ihm SH, Zhang HJ, Parkey J, Hunter DW, Sutherland DE. Single-donor, marginal-dose islet transplantation in patients with type 1 diabetes. Jama. 2005;293(7):830-835.
  78. Frank AM, Barker CF, Markmann JF. Comparison of whole organ pancreas and isolated islet transplantation for type 1 diabetes. Advances in surgery. 2005;39:137-163.
  79. Gerber PA, Pavlicek V, Demartines N, Zuellig R, Pfammatter T, Wuthrich R, Weber M, Spinas GA, Lehmann R. Simultaneous islet-kidney vs pancreas-kidney transplantation in type 1 diabetes mellitus: a 5 year single centre follow-up. Diabetologia. 2008;51(1):110-119.
  80. Froud T, Baidal DA, Faradji R, Cure P, Mineo D, Selvaggi G, Kenyon NS, Ricordi C, Alejandro R. Islet transplantation with alemtuzumab induction and calcineurin-free maintenance immunosuppression results in improved short- and long-term outcomes. Transplantation. 2008;86(12):1695-1701.
  81. Bellin MD, Kandaswamy R, Parkey J, Zhang HJ, Liu B, Ihm SH, Ansite JD, Witson J, Bansal-Pakala P, Balamurugan AN, Papas K, Sutherland DE, Moran A, Hering BJ. Prolonged insulin independence after islet allotransplants in recipients with type 1 diabetes. Am J Transplant. 2008;8(11):2463-2470.
  82. Alejandro R, Barton FB, Hering BJ, Wease S. 2008 Update from the Collaborative Islet Transplant Registry. Transplantation. 2008;86(12):1783-1788.
  83. Tan J, Yang S, Cai J, Guo J, Huang L, Wu Z, Chen J, Liao L. Simultaneous islet and kidney transplantation in seven patients with type 1 diabetes and end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction. Diabetes. 2008;57(10):2666-2671.
  84. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230-238.
  85. Froud T, Ricordi C, Baidal DA, Hafiz MM, Ponte G, Cure P, Pileggi A, Poggioli R, Ichii H, Khan A, Ferreira JV, Pugliese A, Esquenazi VV, Kenyon NS, Alejandro R. Islet transplantation in type 1 diabetes mellitus using cultured islets and steroid-free immunosuppression: Miami experience. Am J Transplant. 2005;5(8):2037-2046.
  86. Vantyghem MC, Kerr-Conte J, Arnalsteen L, Sergent G, Defrance F, Gmyr V, Declerck N, Raverdy V, Vandewalle B, Pigny P, Noel C, Pattou F. Primary graft function, metabolic control, and graft survival after islet transplantation. Diabetes Care. 2009;32(8):1473-1478.
  87. Shapiro AM. Strategies toward single-donor islets of Langerhans transplantation. Curr Opin Organ Transplant.2011;16(6):627-631.
  88. Nijhoff MF, Engelse MA, Dubbeld J, Braat AE, Ringers J, Roelen DL, van Erkel AR, Spijker HS, Bouwsma H, van der Boog PJ, de Fijter JW, Rabelink TJ, de Koning EJ. Glycemic Stability Through Islet-After-Kidney Transplantation Using an Alemtuzumab-Based Induction Regimen and Long-Term Triple-Maintenance Immunosuppression. Am J Transplant. 2015.
  89. Badell IR, Russell MC, Thompson PW, Turner AP, Weaver TA, Robertson JM, Avila JG, Cano JA, Johnson BE, Song M, Leopardi FV, Swygert S, Strobert EA, Ford ML, Kirk AD, Larsen CP. LFA-1-specific therapy prolongs allograft survival in rhesus macaques. J Clin Invest. 2010;120(12):4520-4531.
  90. Turgeon NA, Avila JG, Cano JA, Hutchinson JJ, Badell IR, Page AJ, Adams AB, Sears MH, Bowen PH, Kirk AD, Pearson TC, Larsen CP. Experience with a novel efalizumab-based immunosuppressive regimen to facilitate single donor islet cell transplantation. Am J Transplant. 2010;10(9):2082-2091.
  91. Posselt AM, Bellin MD, Tavakol M, Szot GL, Frassetto LA, Masharani U, Kerlan RK, Fong L, Vincenti FG, Hering BJ, Bluestone JA, Stock PG. Islet transplantation in type 1 diabetics using an immunosuppressive protocol based on the anti-LFA-1 antibody efalizumab. Am J Transplant. 2010;10(8):1870-1880.
  92. Posselt AM, Szot GL, Frassetto LA, Masharani U, Tavakol M, Amin R, McElroy J, Ramos MD, Kerlan RK, Fong L, Vincenti F, Bluestone JA, Stock PG. Islet transplantation in type 1 diabetic patients using calcineurin inhibitor-free immunosuppressive protocols based on T-cell adhesion or costimulation blockade. Transplantation.2010;90(12):1595-1601.
  93. Fotino C, Pileggi A. Blockade of Leukocyte Function Antigen-1 (LFA-1) in Clinical Islet Transplantation. Curr Diab Rep. 2011;11(5):337-344.
  94. Li T, Ma R, Zhu JY, Wang FS, Huang L, Leng XS. PD-1/PD-L1 costimulatory pathway-induced mouse islet transplantation immune tolerance. Transplant Proc. 2015;47(1):165-170.
  95. Watanabe M, Yamashita K, Suzuki T, Kamachi H, Kuraya D, Koshizuka Y, Ogura M, Yoshida T, Aoyagi T, Fukumori D, Shimamura T, Okimura K, Maeta K, Miura T, Sakai F, Todo S. ASKP1240, a fully human anti-CD40 monoclonal antibody, prolongs pancreatic islet allograft survival in nonhuman primates. Am J Transplant.2013;13(8):1976-1988.
  96. Citro A, Cantarelli E, Piemonti L. The CXCR1/2 Pathway: Involvement in Diabetes Pathophysiology and Potential Target for T1D Interventions. Curr Diab Rep. 2015;15(10):638.
  97. Maffi P, Berney T, Nano R, Niclauss N, Bosco D, Melzi R, Mercalli A, Magistretti P, De Cobelli F, Battaglia M, Scavini M, Demuylder-Mischler S, Secchi A, Piemonti L. Calcineurin inhibitor-free immunosuppressive regimen in type 1 diabetes patients receiving islet transplantation: single-group phase 1/2 trial. Transplantation.2014;98(12):1301-1309.
  98. Cure P, Froud T, Leitao CB, Pileggi A, Tharavanij T, Bernetti K, Baidal DA, Selvaggi G, Ricordi C, Alejandro R. Late Epstein Barr virus reactivation in islet after kidney transplantation. Transplantation. 2008;86(9):1324-1325.
  99. Hafiz MM, Poggioli R, Caulfield A, Messinger S, Geiger MC, Baidal DA, Froud T, Ferreira JV, Tzakis AG, Ricordi C, Alejandro R. Cytomegalovirus prevalence and transmission after islet allograft transplant in patients with type 1 diabetes mellitus. Am J Transplant. 2004;4(10):1697-1702.
  100. Senior PA, Paty BW, Cockfield SM, Ryan EA, Shapiro AM. Proteinuria developing after clinical islet transplantation resolves with sirolimus withdrawal and increased tacrolimus dosing. Am J Transplant.2005;5(9):2318-2323.
  101. Maffi P, Bertuzzi F, De Taddeo F, Magistretti P, Nano R, Fiorina P, Caumo A, Pozzi P, Socci C, Venturini M, del Maschio A, Secchi A. Kidney function after islet transplant alone in type 1 diabetes: impact of immunosuppressive therapy on progression of diabetic nephropathy. Diabetes Care. 2007;30(5):1150-1155.
  102. Leitao CB, Cure P, Messinger S, Pileggi A, Lenz O, Froud T, Faradji RN, Selvaggi G, Kupin W, Ricordi C, Alejandro R. Stable renal function after islet transplantation: importance of patient selection and aggressive clinical management. Transplantation. 2009;87(5):681-688.
  103. Leitao CB, Froud T, Cure P, Tharavanij T, Pileggi A, Ricordi C, Alejandro R. Nonalbumin proteinuria in islet transplant recipients. Cell Transplant. 2010;19(1):119-125.
  104. Warnock GL, Thompson DM, Meloche RM, Shapiro RJ, Ao Z, Keown P, Johnson JD, Verchere CB, Partovi N, Begg IS, Fung M, Kozak SE, Tong SO, Alghofaili KM, Harris C. A multi-year analysis of islet transplantation compared with intensive medical therapy on progression of complications in type 1 diabetes. Transplantation.2008;86(12):1762-1766.
  105. Leitao CB, Bernetti K, Tharavanij T, Cure P, Lauriola V, Berggren PO, Ricordi C, Alejandro R. Lipotoxicity and decreased islet graft survival. Diabetes Care. 2010;33(3):658-660.
  106. Barshes NR, Lee TC, Goodpastor SE, Balkrishnan R, Schock AP, Mote A, Brunicardi FC, Alejandro R, Ricordi C, Goss JA. Transaminitis after pancreatic islet transplantation. J Am Coll Surg. 2005;200(3):353-361.
  107. Hafiz MM, Faradji RN, Froud T, Pileggi A, Baidal DA, Cure P, Ponte G, Poggioli R, Cornejo A, Messinger S, Ricordi C, Alejandro R. Immunosuppression and procedure-related complications in 26 patients with type 1 diabetes mellitus receiving allogeneic islet cell transplantation. Transplantation. 2005;80(12):1718-1728.
  108. Venturini M, Maffi P, Querques G, Agostini G, Piemonti L, Sironi S, De Cobelli F, Fiorina P, Secchi A, Del Maschio A. Hepatic steatosis after islet transplantation: Can ultrasound predict the clinical outcome? A longitudinal study in 108 patients. Pharmacol Res. 2015;98:52-59.
  109. Monti P, Vignali D, Piemonti L. Monitoring Inflammation, Humoral and Cell-mediated Immunity in Pancreas and Islet Transplants. Curr Diabetes Rev. 2015;11(3):135-143.
  110. Lobo PI, Spencer C, Simmons WD, Hagspiel KD, Angle JF, Deng S, Markmann J, Naji A, Kirk SE, Pruett T, Brayman KL. Development of anti-human leukocyte antigen class 1 antibodies following allogeneic islet cell transplantation. Transplant Proc. 2005;37(8):3438-3440.
  111. Rickels MR, Kearns J, Markmann E, Palanjian M, Markmann JF, Naji A, Kamoun M. HLA sensitization in islet transplantation. Clin Transpl. 2006:413-420.
  112. Cardani R, Pileggi A, Ricordi C, Gomez C, Baidal DA, Ponte GG, Mineo D, Faradji RN, Froud T, Ciancio G, Esquenazi V, Burke GW, 3rd, Selvaggi G, Miller J, Kenyon NS, Alejandro R. Allosensitization of islet allograft recipients. Transplantation. 2007;84(11):1413-1427.
  113. Campbell PM, Senior PA, Salam A, Labranche K, Bigam DL, Kneteman NM, Imes S, Halpin A, Ryan EA, Shapiro AM. High risk of sensitization after failed islet transplantation. Am J Transplant. 2007;7(10):2311-2317.
  114. Bosi E, Bottazzo GF, Secchi A, Pozza G, Shattock M, Saunders A, Gelet A, Touraine JL, Traeger J, Dubernard JM. Islet cell autoimmunity in type I diabetic patients after HLA-mismatched pancreas transplantation. Diabetes.1989;38 Suppl 1:82-84.
  115. Burke GW, 3rd, Vendrame F, Pileggi A, Ciancio G, Reijonen H, Pugliese A. Recurrence of autoimmunity following pancreas transplantation. Curr Diab Rep. 2011;11(5):413-419.
  116. Vendrame F, Pileggi A, Laughlin E, Allende G, Martin-Pagola A, Molano RD, Diamantopoulos S, Standifer N, Geubtner K, Falk BA, Ichii H, Takahashi H, Snowhite I, Chen Z, Mendez A, Chen L, Sageshima J, Ruiz P, Ciancio G, Ricordi C, Reijonen H, Nepom GT, Burke GW, 3rd, Pugliese A. Recurrence of type 1 diabetes after simultaneous pancreas-kidney transplantation, despite immunosuppression, is associated with autoantibodies and pathogenic autoreactive CD4 T-cells. Diabetes. 2010;59(4):947-957.
  117. Ryan EA, Paty BW, Senior PA, Lakey JR, Bigam D, Shapiro AM. Beta-score: an assessment of beta-cell function after islet transplantation. Diabetes Care. 2005;28(2):343-347.
  118. Faradji RN, Monroy K, Messinger S, Pileggi A, Froud T, Baidal DA, Cure PE, Ricordi C, Luzi L, Alejandro R. Simple measures to monitor beta-cell mass and assess islet graft dysfunction. Am J Transplant. 2007;7(2):303-308.
  119. Matsumoto S, Noguchi H, Hatanaka N, Shimoda M, Kobayashi N, Jackson A, Onaca N, Naziruddin B, Levy MF. SUITO index for evaluation of efficacy of single donor islet transplantation. Cell Transplant. 2009;18(5):557-562.
  120. Takita M, Matsumoto S, Qin H, Noguchi H, Shimoda M, Chujo D, Itoh T, Sugimoto K, Onaca N, Naziruddin B, Levy MF. Secretory Unit of Islet Transplant Objects (SUITO) Index can predict severity of hypoglycemic episodes in clinical islet cell transplantation. Cell Transplant. 2011.
  121. Ryan EA, Lakey JR, Rajotte RV, Korbutt GS, Kin T, Imes S, Rabinovitch A, Elliott JF, Bigam D, Kneteman NM, Warnock GL, Larsen I, Shapiro AM. Clinical outcomes and insulin secretion after islet transplantation with the Edmonton protocol. Diabetes. 2001;50(4):710-719.
  122. Ryan EA, Lakey JR, Paty BW, Imes S, Korbutt GS, Kneteman NM, Bigam D, Rajotte RV, Shapiro AM. Successful islet transplantation: continued insulin reserve provides long-term glycemic control. Diabetes.2002;51(7):2148-2157.
  123. Caumo A, Maffi P, Nano R, Luzi L, Hilbrands R, Gillard P, Jacobs-Tulleneers-Thevissen D, Secchi A, Keymeulen B, Pipeleers D, Piemonti L. Comparative evaluation of simple indices of graft function after islet transplantation. Transplantation. 2011;92(7):815-821.
  124. Caumo A, Maffi P, Nano R, Bertuzzi F, Luzi L, Secchi A, Bonifacio E, Piemonti L. Transplant estimated function: a simple index to evaluate beta-cell secretion after islet transplantation. Diabetes Care. 2008;31(2):301-305.
  125. Forbes S, Oram RA, Smith A, Lam A, Olateju T, Imes S, Malcolm AJ, Shapiro AM, Senior PA. Validation of the BETA-2 Score: An Improved Tool to Estimate Beta Cell Function After Clinical Islet Transplantation Using a Single Fasting Blood Sample. Am J Transplant. 2016;16(9):2704-2713.
  126. Golebiewska JE, Solomina J, Thomas C, Kijek MR, Bachul PJ, Basto L, Golab K, Wang LJ, Fillman N, Tibudan M, Cieply K, Philipson L, Debska-Slizien A, Millis JM, Fung J, Witkowski P. Comparative evaluation of simple indices using a single fasting blood sample to estimate beta cell function after islet transplantation. Am J Transplant. 2018;18(4):990-997.
  127. Rickels MR, Stock PG, Koning EJPd, Piemonti L, Pratschke J, Alejandro R, Bellin MD, Berney T, Choudhary P, Johnson PR, Kandaswamy R, Kay TWH, Keymeulen B, Kudva YC, Latres E, Langer RM, Lehmann R, Ludwig B, Markmann JF, Marinac M, Odorico JS, Pattou F, Senior PA, Shaw JAM, Vantyghem M-C, White S. Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes: a Consensus Report on the Igls Criteria from the IPITA/EPITA Opinion Leaders Workshop. Transplant International. 2018.
  128. Rickels MR, Stock PG, Koning EJPd, Piemonti L, Pratschke J, Alejandro R, Bellin MD, Berney T, Choudhary P, Johnson PR, Kandaswamy R, Kay TWH, Keymeulen B, Kudva YC, Latres E, Langer RM, Lehmann R, Ludwig B, Markmann JF, Marinac M, Odorico JS, Pattou F, Senior PA, Shaw JAM, Vantyghem M-C, White S. Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes: a Consensus Report on the Igls Criteria from the IPITA/EPITA Opinion Leaders Workshop. Transplantation. 2018.
  129. Piemonti L, de Koning EJP, Berney T, Odorico JS, Markmann JF, Stock PG, Rickels MR. Defining outcomes for beta cell replacement therapy: a work in progress. Diabetologia. 2018;61(6):1273-1276.
  130. Landstra CP, Andres A, Chetboun M, Conte C, Kelly Y, Berney T, De Koning EJ, Piemonti L, Stock PG, Pattou F. Examination of the Igls Criteria for Defining Functional Outcomes of β-cell Replacement Therapy: IPITA Symposium Report. The Journal of Clinical Endocrinology & Metabolism. 2021;106(10):3049-3059.
  131. Hering BJ, Clarke WR, Bridges ND, Eggerman TL, Alejandro R, Bellin MD, Chaloner K, Czarniecki CW, Goldstein JS, Hunsicker LG, Kaufman DB, Korsgren O, Larsen CP, Luo X, Markmann JF, Naji A, Oberholzer J, Posselt AM, Rickels MR, Ricordi C, Robien MA, Senior PA, Shapiro AM, Stock PG, Turgeon NA. Phase 3 Trial of Transplantation of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia. Diabetes Care.2016;39(7):1230-1240.
  132. Markmann JF, Rickels MR, Eggerman TL, Bridges ND, Lafontant DE, Qidwai J, Foster E, Clarke WR, Kamoun M, Alejandro R, Bellin MD, Chaloner K, Czarniecki CW, Goldstein JS, Hering BJ, Hunsicker LG, Kaufman DB, Korsgren O, Larsen CP, Luo X, Naji A, Oberholzer J, Posselt AM, Ricordi C, Senior PA, Shapiro AMJ, Stock PG, Turgeon NA. Phase 3 trial of human islet-after-kidney transplantation in type 1 diabetes. Am J Transplant.2021;21(4):1477-1492.
  133. Lablanche S, Vantyghem MC, Kessler L, Wojtusciszyn A, Borot S, Thivolet C, Girerd S, Bosco D, Bosson JL, Colin C, Tetaz R, Logerot S, Kerr-Conte J, Renard E, Penfornis A, Morelon E, Buron F, Skaare K, Grguric G, Camillo-Brault C, Egelhofer H, Benomar K, Badet L, Berney T, Pattou F, Benhamou PY. Islet transplantation versus insulin therapy in patients with type 1 diabetes with severe hypoglycaemia or poorly controlled glycaemia after kidney transplantation (TRIMECO): a multicentre, randomised controlled trial. Lancet Diabetes Endocrinol.2018;6(7):527-537.
  134. Maffi P, Lundgren T, Tufveson G, Rafael E, Shaw JAM, Liew A, Saudek F, Witkowski P, Golab K, Bertuzzi F, Gustafsson B, Daffonchio L, Ruffini PA, Piemonti L. Targeting CXCR1/2 Does Not Improve Insulin Secretion After Pancreatic Islet Transplantation: A Phase 3, Double-Blind, Randomized, Placebo-Controlled Trial in Type 1 Diabetes. Diabetes Care. 2020;43(4):710-718.
  135. Piemonti L. Felix dies natalis, insulin... ceterum autem censeo "beta is better". Acta Diabetol. 2021.
  136. Johnson JA, Kotovych M, Ryan EA, Shapiro AJ. Reduced fear of hypoglycemia in successful islet transplantation. Diabetes Care. 2004;27(2):624-625.
  137. Barton FB, Rickels MR, Alejandro R, Hering BJ, Wease S, Naziruddin B, Oberholzer J, Odorico JS, Garfinkel MR, Levy M. Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care.2012;35(7):1436-1445.
  138. Paty BW, Ryan EA, Shapiro AJ, Lakey JR, Robertson RP. Intrahepatic islet transplantation in type 1 diabetic patients does not restore hypoglycemic hormonal counterregulation or symptom recognition after insulin independence. Diabetes. 2002;51(12):3428-3434.
  139. Rickels MR, Schutta MH, Markmann JF, Barker CF, Naji A, Teff KL. β-Cell function following human islet transplantation for type 1 diabetes. Diabetes. 2005;54(1):100-106.
  140. Rickels MR, Schutta MH, Mueller R, Markmann JF, Barker CF, Naji A, Teff KL. Islet cell hormonal responses to hypoglycemia after human islet transplantation for type 1 diabetes. Diabetes. 2005;54(11):3205-3211.
  141. Paty BW, Senior PA, Lakey JR, Shapiro AJ, Ryan EA. Assessment of glycemic control after islet transplantation using the continuous glucose monitor in insulin-independent versus insulin-requiring type 1 diabetes subjects. Diabetes technology & therapeutics. 2006;8(2):165-173.
  142. Rickels MR, Naji A, Teff KL. Acute insulin responses to glucose and arginine as predictors of β-cell secretory capacity in human islet transplantation. Transplantation. 2007;84(10):1357-1360.
  143. Vantyghem MC, de Koning EJP, Pattou F, Rickels MR. Advances in beta-cell replacement therapy for the treatment of type 1 diabetes. Lancet. 2019;394(10205):1274-1285.
  144. Vantyghem M-C, Chetboun M, Gmyr V, Jannin A, Espiard S, Le Mapihan K, Raverdy V, Delalleau N, Machuron F, Hubert T. Ten-year outcome of islet alone or islet after kidney transplantation in type 1 diabetes: A prospective parallel-arm cohort study. Diabetes Care. 2019;42(11):2042-2049.
  145. Marfil-Garza BA, Imes S, Verhoeff K, Hefler J, Lam A, Dajani K, Anderson B, O'Gorman D, Kin T, Bigam D. Pancreatic islet transplantation in type 1 diabetes: 20-year experience from a single-centre cohort in Canada. The lancet Diabetes & endocrinology. 2022.
  146. Vantyghem MC, Raverdy V, Balavoine AS, Defrance F, Caiazzo R, Arnalsteen L, Gmyr V, Hazzan M, Noel C, Kerr-Conte J, Pattou F. Continuous glucose monitoring after islet transplantation in type 1 diabetes: an excellent graft function (beta-score greater than 7) Is required to abrogate hyperglycemia, whereas a minimal function is necessary to suppress severe hypoglycemia (beta-score greater than 3). J Clin Endocrinol Metab.2012;97(11):E2078-2083.
  147. Johnson JA, Kotovych M, Ryan EA, Shapiro AM. Reduced fear of hypoglycemia in successful islet transplantation. Diabetes Care. 2004;27(2):624-625.
  148. Poggioli R, Faradji RN, Ponte G, Betancourt A, Messinger S, Baidal DA, Froud T, Ricordi C, Alejandro R. Quality of life after islet transplantation. Am J Transplant. 2006;6(2):371-378.
  149. Toso C, Shapiro AM, Bowker S, Dinyari P, Paty B, Ryan EA, Senior P, Johnson JA. Quality of life after islet transplant: impact of the number of islet infusions and metabolic outcome. Transplantation. 2007;84(5):664-666.
  150. Leitao CB, Tharavanij T, Cure P, Pileggi A, Baidal DA, Ricordi C, Alejandro R. Restoration of hypoglycemia awareness after islet transplantation. Diabetes Care. 2008;31(11):2113-2115.
  151. Tharavanij T, Betancourt A, Messinger S, Cure P, Leitao CB, Baidal DA, Froud T, Ricordi C, Alejandro R. Improved long-term health-related quality of life after islet transplantation. Transplantation. 2008;86(9):1161-1167.
  152. Radosevich DM, Jevne R, Bellin M, Kandaswamy R, Sutherland DE, Hering BJ. Comprehensive health assessment and five-yr follow-up of allogeneic islet transplant recipients. Clin Transplant. 2013;27(6):E715-724.
  153. Benhamou PY, Milliat-Guittard L, Wojtusciszyn A, Kessler L, Toso C, Baertschiger R, Debaty I, Badet L, Penfornis A, Thivolet C, Renard E, Bayle F, Morel P, Morelon E, Colin C, Berney T. Quality of life after islet transplantation: data from the GRAGIL 1 and 2 trials. Diabet Med. 2009;26(6):617-621.
  154. Cure P, Pileggi A, Froud T, Messinger S, Faradji RN, Baidal DA, Cardani R, Curry A, Poggioli R, Pugliese A, Betancourt A, Esquenazi V, Ciancio G, Selvaggi G, Burke GW, 3rd, Ricordi C, Alejandro R. Improved metabolic control and quality of life in seven patients with type 1 diabetes following islet after kidney transplantation. Transplantation. 2008;85(6):801-812.
  155. Barshes NR, Vanatta JM, Mote A, Lee TC, Schock AP, Balkrishnan R, Brunicardi FC, Goss JA. Health-related quality of life after pancreatic islet transplantation: a longitudinal study. Transplantation. 2005;79(12):1727-1730.
  156. Reid L, Baxter F, Forbes S. Effects of Islet Transplantation on Microvascular and Macrovascular Complications in Type 1 Diabetes. Diabet Med. 2021:e14570.
  157. Thompson DM, Meloche M, Ao Z, Paty B, Keown P, Shapiro RJ, Ho S, Worsley D, Fung M, Meneilly G, Begg I, Al Mehthel M, Kondi J, Harris C, Fensom B, Kozak SE, Tong SO, Trinh M, Warnock GL. Reduced progression of diabetic microvascular complications with islet cell transplantation compared with intensive medical therapy. Transplantation. 2011;91(3):373-378.
  158. Lee TC, Barshes NR, O'Mahony CA, Nguyen L, Brunicardi FC, Ricordi C, Alejandro R, Schock AP, Mote A, Goss JA. The effect of pancreatic islet transplantation on progression of diabetic retinopathy and neuropathy. Transplant Proc. 2005;37(5):2263-2265.
  159. Venturini M, Fiorina P, Maffi P, Losio C, Vergani A, Secchi A, Del Maschio A. Early increase of retinal arterial and venous blood flow velocities at color Doppler imaging in brittle type 1 diabetes after islet transplant alone. Transplantation. 2006;81(9):1274-1277.
  160. Del Carro U, Fiorina P, Amadio S, De Toni Franceschini L, Petrelli A, Menini S, Martinelli Boneschi F, Ferrari S, Pugliese G, Maffi P, Comi G, Secchi A. Evaluation of polyneuropathy markers in type 1 diabetic kidney transplant patients and effects of islet transplantation: neurophysiological and skin biopsy longitudinal analysis. Diabetes Care. 2007;30(12):3063-3069.
  161. Vantyghem MC, Quintin D, Caiazzo R, Leroy C, Raverdy V, Cassim F, Glowacki F, Hubert T, Gmyr V, Noel C, Kerr-Conte J, Pattou F. Improvement of electrophysiological neuropathy after islet transplantation for type 1 diabetes: a 5-year prospective study. Diabetes Care. 2014;37(6):e141-142.
  162. D'Addio F, Maffi P, Vezzulli P, Vergani A, Mello A, Bassi R, Nano R, Falautano M, Coppi E, Finzi G, D'Angelo A, Fermo I, Pellegatta F, La Rosa S, Magnani G, Piemonti L, Falini A, Folli F, Secchi A, Fiorina P. Islet transplantation stabilizes hemostatic abnormalities and cerebral metabolism in individuals with type 1 diabetes. Diabetes Care. 2014;37(1):267-276.
  163. Toso C, Baertschiger R, Morel P, Bosco D, Armanet M, Wojtusciszyn A, Badet L, Philippe J, Becker CD, Hadaya K, Majno P, Buhler L, Berney T. Sequential kidney/islet transplantation: efficacy and safety assessment of a steroid-free immunosuppression protocol. Am J Transplant. 2006;6(5 Pt 1):1049-1058.
  164. Fiorina P, Folli F, Zerbini G, Maffi P, Gremizzi C, Di Carlo V, Socci C, Bertuzzi F, Kashgarian M, Secchi A. Islet transplantation is associated with improvement of renal function among uremic patients with type I diabetes mellitus and kidney transplants. J Am Soc Nephrol. 2003;14(8):2150-2158.
  165. Fiorina P, Venturini M, Folli F, Losio C, Maffi P, Placidi C, La Rosa S, Orsenigo E, Socci C, Capella C, Del Maschio A, Secchi A. Natural history of kidney graft survival, hypertrophy, and vascular function in end-stage renal disease type 1 diabetic kidney-transplanted patients: beneficial impact of pancreas and successful islet cotransplantation. Diabetes Care. 2005;28(6):1303-1310.
  166. Senior PA, Zeman M, Paty BW, Ryan EA, Shapiro AM. Changes in renal function after clinical islet transplantation: four-year observational study. Am J Transplant. 2007;7(1):91-98.
  167. Gillard P, Rustandi M, Efendi A, Lee DH, Ling Z, Hilbrands R, Kuypers D, Mathieu C, Jacobs-Tulleneers-Thevissen D, Gorus F, Pipeleers D, Keymeulen B. Early Alteration of Kidney Function in Nonuremic Type 1 Diabetic Islet Transplant Recipients Under Tacrolimus-Mycophenolate Therapy. Transplantation. 2014.
  168. Fung MA, Warnock GL, Ao Z, Keown P, Meloche M, Shapiro RJ, Ho S, Worsley D, Meneilly GS, Al Ghofaili K, Kozak SE, Tong SO, Trinh M, Blackburn L, Kozak RM, Fensom BA, Thompson DM. The effect of medical therapy and islet cell transplantation on diabetic nephropathy: an interim report. Transplantation. 2007;84(1):17-22.
  169. Fiorina P, Folli F, Bertuzzi F, Maffi P, Finzi G, Venturini M, Socci C, Davalli A, Orsenigo E, Monti L, Falqui L, Uccella S, La Rosa S, Usellini L, Properzi G, Di Carlo V, Del Maschio A, Capella C, Secchi A. Long-term beneficial effect of islet transplantation on diabetic macro-/microangiopathy in type 1 diabetic kidney-transplanted patients. Diabetes Care. 2003;26(4):1129-1136.
  170. Fiorina P, Folli F, Maffi P, Placidi C, Venturini M, Finzi G, Bertuzzi F, Davalli A, D'Angelo A, Socci C, Gremizzi C, Orsenigo E, La Rosa S, Ponzoni M, Cardillo M, Scalamogna M, Del Maschio A, Capella C, Di Carlo V, Secchi A. Islet transplantation improves vascular diabetic complications in patients with diabetes who underwent kidney transplantation: a comparison between kidney-pancreas and kidney-alone transplantation. Transplantation.2003;75(8):1296-1301.
  171. Fiorina P, Gremizzi C, Maffi P, Caldara R, Tavano D, Monti L, Socci C, Folli F, Fazio F, Astorri E, Del Maschio A, Secchi A. Islet transplantation is associated with an improvement of cardiovascular function in type 1 diabetic kidney transplant patients. Diabetes Care. 2005;28(6):1358-1365.
  172. Del Carro U, Fiorina P, Amadio S, De Toni Franceschini L, Petrelli A, Menini S, Boneschi FM, Ferrari S, Pugliese G, Maffi P, Comi G, Secchi A. Evaluation of polyneuropathy markers in type 1 diabetic kidney transplant patients and effects of islet transplantation: neurophysiological and skin biopsy longitudinal analysis. Diabetes Care. 2007;30(12):3063-3069.
  173. Danielson KK, Hatipoglu B, Kinzer K, Kaplan B, Martellotto J, Qi M, Mele A, Benedetti E, Oberholzer J. Reduction in carotid intima-media thickness after pancreatic islet transplantation in patients with type 1 diabetes. Diabetes Care. 2013;36(2):450-456.
  174. Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, Lakey JR, Shapiro AM. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54(7):2060-2069.
  175. Vantyghem MC, Defrance F, Quintin D, Leroy C, Raverdi V, Prevost G, Caiazzo R, Kerr-Conte J, Glowacki F, Hazzan M, Noel C, Pattou F, Diamenord AS, Bresson R, Bourdelle-Hego MF, Cazaubiel M, Cordonnier M, Delefosse D, Dorey F, Fayard A, Fermon C, Fontaine P, Gillot C, Haye S, Le Guillou AC, Karrouz W, Lemaire C, Lepeut M, Leroy R, Mycinski B, Parent E, Siame C, Sterkers A, Torres F, Verier-Mine O, Verlet E, Desailloud R, Durrbach A, Godin M, Lalau JD, Lukas-Croisier C, Thervet E, Toupance O, Reznik Y, Westeel PF. Treating diabetes with islet transplantation: Lessons from the past decade in Lille. Diabetes Metab. 2014;40(2):108-119.
  176. Bellin MD, Kandaswamy R, Parkey J, Zhang HJ, Liu B, Ihm SH, Ansite JD, Witson J, Bansal-Pakala P, Balamurugan AN, Papas KK, Sutherland DE, Moran A, Hering BJ. Prolonged insulin independence after islet allotransplants in recipients with type 1 diabetes. Am J Transplant. 2008;8(11):2463-2470.
  177. Maffi P, Scavini M, Socci C, Piemonti L, Caldara R, Gremizzi C, Melzi R, Nano R, Orsenigo E, Venturini M, Staudacher C, Del Maschio A, Secchi A. Risks and benefits of transplantation in the cure of type 1 diabetes: whole pancreas versus islet transplantation. A single center study. Rev Diabet Stud. 2011;8(1):44-50.
  178. Shapiro AT, C; Imes, S; Koh, A; Kin, T; O'Gorman, D; Malcolm, A; Dinyari, P; Owen, R; Kneteman, RN; Bigam, DL; Calne, RY; Senior, PA; Roep, BO. Five-Year Results of Islet-Alone Transplantation Match Pancreas-Alone Transplantation with Alemtuzumab, Tac/MMF, with Strong Suppression of Auto and Alloreactivity. Paper presented at: 13th World Congressof the International Pancreas and Islet Transplant Association (IPITA)2011; Prague
  179. Bellin MD, Barton FB, Heitman A, Harmon JV, Kandaswamy R, Balamurugan AN, Sutherland DE, Alejandro R, Hering BJ. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant. 2012;12(6):1576-1583.
  180. Johnson PR, White SA, Robertson GS, Koppiker N, Burden AC, Dennison AR, London NJ. Pancreatic islet autotransplantation combined with total pancreatectomy for the treatment of chronic pancreatitis--the Leicester experience. J Mol Med (Berl). 1999;77(1):130-132.
  181. Ahmad SA, Lowy AM, Wray CJ, D'Alessio D, Choe KA, James LE, Gelrud A, Matthews JB, Rilo HL. Factors associated with insulin and narcotic independence after islet autotransplantation in patients with severe chronic pancreatitis. J Am Coll Surg. 2005;201(5):680-687.
  182. Morgan KA, Nishimura M, Uflacker R, Adams DB. Percutaneous transhepatic islet cell autotransplantation after pancreatectomy for chronic pancreatitis: a novel approach. HPB (Oxford). 2011;13(7):511-516.
  183. Sutherland DE, Gruessner AC, Carlson AM, Blondet JJ, Balamurugan AN, Reigstad KF, Beilman GJ, Bellin MD, Hering BJ. Islet autotransplant outcomes after total pancreatectomy: a contrast to islet allograft outcomes. Transplantation. 2008;86(12):1799-1802.
  184. Bassi R, Fiorina P. Impact of islet transplantation on diabetes complications and quality of life. Curr Diab Rep.2011;11(5):355-363.
  185. Hansen A, Johansson BL, Wahren J, von Bibra H. C-peptide exerts beneficial effects on myocardial blood flow and function in patients with type 1 diabetes. Diabetes. 2002;51(10):3077-3082.
  186. Ryan EA, Paty BW, Senior PA, Shapiro AM. Risks and side effects of islet transplantation. Curr Diab Rep.2004;4(4):304-309.
  187. Gaba RC, Garcia-Roca R, Oberholzer J. Pancreatic islet cell transplantation: an update for interventional radiologists. J Vasc Interv Radiol. 2012;23(5):583-594; quiz 594.
  188. Villiger P, Ryan EA, Owen R, O'Kelly K, Oberholzer J, Al Saif F, Kin T, Wang H, Larsen I, Blitz SL, Menon V, Senior P, Bigam DL, Paty B, Kneteman NM, Lakey JR, Shapiro AM. Prevention of bleeding after islet transplantation: lessons learned from a multivariate analysis of 132 cases at a single institution. Am J Transplant. 2005;5(12):2992-2998.
  189. Froud T, Yrizarry JM, Alejandro R, Ricordi C. Use of D-STAT to prevent bleeding following percutaneous transhepatic intraportal islet transplantation. Cell Transplant. 2004;13(1):55-59.
  190. Bhargava R, Senior PA, Ackerman TE, Ryan EA, Paty BW, Lakey JR, Shapiro AM. Prevalence of hepatic steatosis after islet transplantation and its relation to graft function. Diabetes. 2004;53(5):1311-1317.
  191. Maffi P, Angeli E, Bertuzzi F, Paties C, Socci C, Fedeli C, De Taddeo F, Nano R, Di Carlo V, Del Maschio A, Secchi A. Minimal focal steatosis of liver after islet transplantation in humans: a long-term study. Cell Transplant.2005;14(10):727-733.
  192. Casey JJ, Lakey JR, Ryan EA, Paty BW, Owen R, O'Kelly K, Nanji S, Rajotte RV, Korbutt GS, Bigam D, Kneteman NN, Shapiro AM. Portal venous pressure changes after sequential clinical islet transplantation. Transplantation. 2002;74(7):913-915.
  193. Molinari M, Al-Saif F, Ryan EA, Lakey JR, Senior PA, Paty BW, Bigam DL, Kneteman NM, Shapiro AM. Sirolimus-induced ulceration of the small bowel in islet transplant recipients: report of two cases. Am J Transplant. 2005;5(11):2799-2804.
  194. Ponte GM, Baidal DA, Romanelli P, Faradji RN, Poggioli R, Cure P, Froud T, Selvaggi G, Pileggi A, Ricordi C, Alejandro R. Resolution of severe atopic dermatitis after tacrolimus withdrawal. Cell transplantation.2007;16(1):23-30.
  195. Andres A, Toso C, Morel P, Demuylder-Mischler S, Bosco D, Baertschiger R, Pernin N, Bucher P, Majno PE, Buhler LH, Berney T. Impairment of renal function after islet transplant alone or islet-after-kidney transplantation using a sirolimus/tacrolimus-based immunosuppressive regimen. Transpl Int. 2005;18(11):1226-1230.
  196. Gala-Lopez BL, Senior PA, Koh A, Kashkoush SM, Kawahara T, Kin T, Humar A, Shapiro AM. Late cytomegalovirus transmission and impact of T-depletion in clinical islet transplantation. Am J Transplant.2011;11(12):2708-2714.
  197. Pileggi A, Cobianchi L, Inverardi L, Ricordi C. Overcoming the challenges now limiting islet transplantation: a sequential, integrated approach. Ann N Y Acad Sci. 2006;1079:383-398.
  198. Mineo D, Pileggi A, Alejandro R, Ricordi C. Point: steady progress and current challenges in clinical islet transplantation. Diabetes Care. 2009;32(8):1563-1569.
  199. Pellegrini S, Cantarelli E, Sordi V, Nano R, Piemonti L. The state of the art of islet transplantation and cell therapy in type 1 diabetes. Acta Diabetol. 2016.
  200. Lo JF, Wang Y, Li Z, Zhao Z, Hu D, Eddington DT, Oberholzer J. Quantitative and temporal control of oxygen microenvironment at the single islet level. J Vis Exp. 2013(81):e50616.
  201. Barkai U, Weir GC, Colton CK, Ludwig B, Bornstein SR, Brendel MD, Neufeld T, Bremer C, Leon A, Evron Y, Yavriyants K, Azarov D, Zimermann B, Maimon S, Shabtay N, Balyura M, Rozenshtein T, Vardi P, Bloch K, de Vos P, Rotem A. Enhanced oxygen supply improves islet viability in a new bioartificial pancreas. Cell Transplant.2013;22(8):1463-1476.
  202. Piemonti L, Andres A, Casey J, de Koning E, Engelse M, Hilbrands R, Johnson P, Keymeulen B, Kerr‐Conte J, Korsgren O. US Food and Drug Administration (FDA) Panel Endorses Islet Cell Treatment for Type 1 Diabetes: A Pyrrhic victory? Transplant International. 2021.
  203. Hudson A, Bradbury L, Johnson R, Fuggle SV, Shaw JA, Casey JJ, Friend PJ, Watson CJ. The UK Pancreas Allocation Scheme for Whole Organ and Islet Transplantation. Am J Transplant. 2015;15(9):2443-2455.
  204. Cornateanu SM, O'Neill S, Dholakia S, Counter CJ, Sherif AE, Casey JJ, Friend P, Oniscu GC. Pancreas utilization rates in the UK - an 11-year analysis. Transpl Int. 2021;34(7):1306-1318.
  205. Nordheim E, Lindahl JP, Carlsen RK, Asberg A, Birkeland KI, Horneland R, Boye B, Scholz H, Jenssen TG. Patient selection for islet or solid organ pancreas transplantation: experiences from a multidisciplinary outpatient-clinic approach. Endocr Connect. 2021;10(2):230-239.
  206. Nano R, Kerr-Conte JA, Scholz H, Engelse M, Karlsson M, Saudek F, Bosco D, Antonioli B, Bertuzzi F, Johnson PRV, Ludwing B, Ling Z, De Paep DL, Keymeulen B, Pattou F, Berney T, Korsgren O, de Koning E, Piemonti L. Heterogeneity of Human Pancreatic Islet Isolation Around Europe: Results of a Survey Study. Transplantation.2020;104(1):190-196.
  207. Perrier Q, Lavallard V, Pernin N, Wassmer CH, Cottet-Dumoulin D, Lebreton F, Bellofatto K, Andres A, Berishvili E, Bosco D, Berney T, Parnaud G. Failure mode and effect analysis in human islet isolation: from the theoretical to the practical risk. Islets. 2021;13(1-2):1-9.
  208. Witkowski P, Philipson LH, Kaufman DB, Ratner LE, Abouljoud MS, Bellin MD, Buse JB, Kandeel F, Stock PG, Mulligan DC, Markmann JF, Kozlowski T, Andreoni KA, Alejandro R, Baidal DA, Hardy MA, Wickrema A, Mirmira RG, Fung J, Becker YT, Josephson MA, Bachul PJ, Pyda JS, Charlton M, Millis JM, Gaglia JL, Stratta RJ, Fridell JA, Niederhaus SV, Forbes RC, Jayant K, Robertson RP, Odorico JS, Levy MF, Harland RC, Abrams PL, Olaitan OK, Kandaswamy R, Wellen JR, Japour AJ, Desai CS, Naziruddin B, Balamurugan AN, Barth RN, Ricordi C. The demise of islet allotransplantation in the United States: A call for an urgent regulatory update. Am J Transplant. 2021;21(4):1365-1375.
  209. Piemonti L, Andres A, Casey J, de Koning E, Engelse M, Hilbrands R, Johnson P, Keymeulen B, Kerr-Conte J, Korsgren O, Lehmann R, Lundgren T, Maffi P, Pattou F, Saudek F, Shaw J, Scholz H, White S, Berney T. US food and drug administration (FDA) panel endorses islet cell treatment for type 1 diabetes: A pyrrhic victory? Transpl Int. 2021;34(7):1182-1186.
  210. Arifin DR, Bulte JWM. In Vivo Imaging of Pancreatic Islet Grafts in Diabetes Treatment. Front Endocrinol (Lausanne). 2021;12:640117.
  211. Murakami T, Fujimoto H, Inagaki N. Non-invasive Beta-cell Imaging: Visualization, Quantification, and Beyond. Front Endocrinol (Lausanne). 2021;12:714348.
  212. Piemonti L, Everly MJ, Maffi P, Scavini M, Poli F, Nano R, Cardillo M, Melzi R, Mercalli A, Sordi V, Lampasona V, De Arias AE, Scalamogna M, Bosi E, Bonifacio E, Secchi A, Terasaki PI. Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes. 2013;62(5):1656-1664.
  213. Sabbah S, Liew A, Brooks AM, Kundu R, Reading JL, Flatt A, Counter C, Choudhary P, Forbes S, Rosenthal MJ, Rutter MK, Cairns S, Johnson P, Casey J, Peakman M, Shaw JA, Tree TIM. Autoreactive T cell profiles are altered following allogeneic islet transplantation with alemtuzumab induction and re-emerging phenotype is associated with graft function. Am J Transplant. 2021;21(3):1027-1038.
  214. Buron F, Reffet S, Badet L, Morelon E, Thaunat O. Immunological Monitoring in Beta Cell Replacement: Towards a Pathophysiology-Guided Implementation of Biomarkers. Curr Diab Rep. 2021;21(6):19.
  215. Anteby R, Lucander A, Bachul PJ, Pyda J, Grybowski D, Basto L, Generette GS, Perea L, Golab K, Wang LJ, Tibudan M, Thomas C, Fung J, Witkowski P. Evaluating the Prognostic Value of Islet Autoantibody Monitoring in Islet Transplant Recipients with Long-Standing Type 1 Diabetes Mellitus. J Clin Med. 2021;10(12).
  216. Suwandi JS, Nikolic T, Roep BO. Translating Mechanism of Regulatory Action of Tolerogenic Dendritic Cells to Monitoring Endpoints in Clinical Trials. Front Immunol. 2017;8:1598.
  217. Tremmel DM, Odorico JS. Rebuilding a better home for transplanted islets. Organogenesis. 2018;14(4):163-168.
  218. Citro A, Moser PT, Dugnani E, Rajab TK, Ren X, Evangelista-Leite D, Charest JM, Peloso A, Podesser BK, Manenti F, Pellegrini S, Piemonti L, Ott HC. Biofabrication of a vascularized islet organ for type 1 diabetes. Biomaterials. 2019;199:40-51.
  219. Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater. 2021.
  220. Basta G, Montanucci P, Calafiore R. Microencapsulation of cells and molecular therapy of type 1 diabetes mellitus: The actual state and future perspectives between promise and progress. J Diabetes Investig.2021;12(3):301-309.
  221. Marigliano M, Bertera S, Grupillo M, Trucco M, Bottino R. Pig-to-nonhuman primates pancreatic islet xenotransplantation: an overview. Curr Diab Rep. 2011;11(5):402-412.
  222. Pagliuca FW, Millman JR, Gurtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of Functional Human Pancreatic beta Cells In Vitro. Cell. 2014;159(2):428-439.
  223. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O'Dwyer S, Quiskamp N, Mojibian M, Albrecht T, Yang YH, Johnson JD, Kieffer TJ. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121-1133.
  224. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D'Amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol.2008;26(4):443-452.
  225. Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, Bang AG, Bhoumik A, Cepa I, Cesario RM, Haakmeester C, Kadoya K, Kelly JR, Kerr J, Martinson LA, McLean AB, Moorman MA, Payne JK, Richardson M, Ross KG, Sherrer ES, Song X, Wilson AZ, Brandon EP, Green CE, Kroon EJ, Kelly OG, D'Amour KA, Robins AJ. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS One. 2012;7(5):e37004.
  226. Pellegrini S, Piemonti L, Sordi V. Pluripotent stem cell replacement approaches to treat type 1 diabetes. Curr Opin Pharmacol. 2018;43:20-26.
  227. Odorico J, Markmann J, Melton D, Greenstein J, Hwa A, Nostro C, Rezania A, Oberholzer J, Pipeleers D, Yang L, Cowan C, Huangfu D, Egli D, Ben-David U, Vallier L, Grey ST, Tang Q, Roep B, Ricordi C, Naji A, Orlando G, Anderson DG, Poznansky M, Ludwig B, Tomei A, Greiner DL, Graham M, Carpenter M, Migliaccio G, D'Amour K, Hering B, Piemonti L, Berney T, Rickels M, Kay T, Adams A. Report of the Key Opinion Leaders Meeting on Stem Cell-derived Beta Cells. Transplantation. 2018;102(8):1223-1229.
  228. Carlsson P-O, Schwarcz E, Korsgren O, Le Blanc K. Preserved β-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes. 2015;64(2):587-592.
  229. Madani S, Setudeh A, Aghayan HR, Alavi-Moghadam S, Rouhifard M, Rezaei N, Rostami P, Mohsenipour R, Amirkashani D, Bandarian F. Placenta derived Mesenchymal Stem Cells transplantation in Type 1 diabetes: preliminary report of phase 1 clinical trial. Journal of Diabetes & Metabolic Disorders. 2021;20(2):1179-1189.
  230. Arzouni AA, Vargas-Seymour A, Nardi N, JF King A, Jones PM. Using mesenchymal stromal cells in islet transplantation. Stem Cells Translational Medicine. 2018;7(8):559-563.
  231. Pellegrini S, Piemonti L, Sordi V. Pluripotent stem cell replacement approaches to treat type 1 diabetes. Current Opinion in Pharmacology. 2018;43:20-26.
  232. Migliorini A, Nostro MC, Sneddon JB. Human pluripotent stem cell-derived insulin-producing cells: A regenerative medicine perspective. Cell Metabolism. 2021;33(4):721-731.
  233. D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE. Production of pancreatic hormone–expressing endocrine cells from human embryonic stem cells. Nature biotechnology. 2006;24(11):1392-1401.
  234. Zhang D, Jiang W, Liu M, Sui X, Yin X, Chen S, Shi Y, Deng H. Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell research. 2009;19(4):429-438.
  235. Tateishi K, He J, Taranova O, Liang G, D'Alessio AC, Zhang Y. Generation of insulin-secreting islet-like clusters from human skin fibroblasts. Journal of Biological Chemistry. 2008;283(46):31601-31607.
  236. Nostro MC, Sarangi F, Ogawa S, Holtzinger A, Corneo B, Li X, Micallef SJ, Park I-H, Basford C, Wheeler MB. Stage-specific signaling through TGFβ family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development. 2011;138(5):861-871.
  237. Hrvatin S, O’Donnell CW, Deng F, Millman JR, Pagliuca FW, DiIorio P, Rezania A, Gifford DK, Melton DA. Differentiated human stem cells resemble fetal, not adult, β cells. Proceedings of the National Academy of Sciences. 2014;111(8):3038-3043.
  238. Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of functional human pancreatic β cells in vitro. Cell. 2014;159(2):428-439.
  239. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O'dwyer S, Quiskamp N, Mojibian M, Albrecht T. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nature biotechnology. 2014;32(11):1121.
  240. Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, Guo T, Puri S, Haataja L, Cirulli V. Controlled induction of human pancreatic progenitors produces functional beta‐like cells in vitro. The EMBO journal. 2015;34(13):1759-1772.
  241. Sambathkumar R, Migliorini A, Nostro MC. Pluripotent stem cell-derived pancreatic progenitors and β-Like cells for Type 1 diabetes treatment. Physiology. 2018;33(6):394-402.
  242. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nature biotechnology. 2008;26(4):443-452.
  243. Kelly OG, Chan MY, Martinson LA, Kadoya K, Ostertag TM, Ross KG, Richardson M, Carpenter MK, D'amour KA, Kroon E. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nature biotechnology. 2011;29(8):750-756.
  244. Ramzy A, Thompson DM, Ward-Hartstonge KA, Ivison S, Cook L, Garcia RV, Loyal J, Kim PTW, Warnock GL, Levings MK, Kieffer TJ. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell. 2021;28(12):2047-2061.e2045.
  245. Sordi V, Pellegrini S, Piemonti L. Immunological Issues After Stem Cell-Based beta Cell Replacement. Curr Diab Rep. 2017;17(9):68.
  246. Zheng D, Wang X, Xu RH. Concise review: one stone for multiple birds: generating universally compatible human embryonic stem cells. Stem Cells. 2016;34(9):2269-2275.
  247. Barry J, Hyllner J, Stacey G, Taylor CJ, Turner M. Setting up a haplobank: issues and solutions. Current stem cell reports. 2015;1(2):110-117.
  248. Borot S, Crowe LA, Toso C, Vallee JP, Berney T. Noninvasive imaging techniques in islet transplantation. Curr Diab Rep. 2011;11(5):375-383.
  249. Wang P, Schuetz C, Vallabhajosyula P, Medarova Z, Tena A, Wei L, Yamada K, Deng S, Markmann JF, Sachs DH, Moore A. Monitoring of Allogeneic Islet Grafts in Nonhuman Primates Using MRI. Transplantation.2015;99(8):1574-1581.
  250. Malosio ML, Esposito A, Brigatti C, Palmisano A, Piemonti L, Nano R, Maffi P, De Cobelli F, Del Maschio A, Secchi A. Mr Imaging Monitoring of Iron Labeled Pancreatic Islets in a Small Series of Patients: Islets Fate in Successful, Unsuccessful and Auto-Transplantation. Cell Transplant. 2014.
  251. Esposito A, Palmisano A, Maffi P, Malosio ML, Nano R, Canu T, De Cobelli F, Piemonti L, Ironi G, Secchi A, Del Maschio A. Liver perfusion changes occurring during pancreatic islet engraftment: a dynamic contrast-enhanced magnetic resonance study. Am J Transplant. 2014;14(1):202-209.
  252. Saudek F, Jirak D, Girman P, Herynek V, Dezortova M, Kriz J, Peregrin J, Berkova Z, Zacharovova K, Hajek M. Magnetic resonance imaging of pancreatic islets transplanted into the liver in humans. Transplantation.2010;90(12):1602-1606.
  253. Toso C, Vallee JP, Morel P, Ris F, Demuylder-Mischler S, Lepetit-Coiffe M, Marangon N, Saudek F, James Shapiro AM, Bosco D, Berney T. Clinical magnetic resonance imaging of pancreatic islet grafts after iron nanoparticle labeling. Am J Transplant. 2008;8(3):701-706.
  254. Toso C, Zaidi H, Morel P, Armanet M, Andres A, Pernin N, Baertschiger R, Slosman D, Buhler LH, Bosco D, Berney T. Positron-emission tomography imaging of early events after transplantation of islets of Langerhans. Transplantation. 2005;79(3):353-355.
  255. Eriksson O, Alavi A. Imaging the islet graft by positron emission tomography. Eur J Nucl Med Mol Imaging.2012;39(3):533-542.
  256. Kriz J, Jirak D, Berkova Z, Herynek V, Lodererova A, Girman P, Habart D, Hajek M, Saudek F. Detection of pancreatic islet allograft impairment in advance of functional failure using magnetic resonance imaging. Transpl Int. 2012;25(2):250-260.
  257. Eich T, Eriksson O, Lundgren T. Visualization of early engraftment in clinical islet transplantation by positron-emission tomography. N Engl J Med. 2007;356(26):2754-2755.
  258. Han D, Leith J, Alejandro R, Bolton W, Ricordi C, Kenyon NS. Peripheral blood cytotoxic lymphocyte gene transcript levels differ in patients with long-term type 1 diabetes compared to normal controls. Cell Transplant.2005;14(6):403-409.
  259. Han D, Xu X, Baidal D, Leith J, Ricordi C, Alejandro R, Kenyon NS. Assessment of cytotoxic lymphocyte gene expression in the peripheral blood of human islet allograft recipients: elevation precedes clinical evidence of rejection. Diabetes. 2004;53(9):2281-2290.
  260. Han D, Xu X, Pastori RL, Ricordi C, Kenyon NS. Elevation of cytotoxic lymphocyte gene expression is predictive of islet allograft rejection in nonhuman primates. Diabetes. 2002;51(3):562-566.
  261. Huurman VA, Velthuis JH, Hilbrands R, Tree TI, Gillard P, van der Meer-Prins PM, Duinkerken G, Pinkse GG, Keymeulen B, Roelen DL, Claas FH, Pipeleers DG, Roep BO. Allograft-specific cytokine profiles associate with clinical outcome after islet cell transplantation. Am J Transplant. 2009;9(2):382-388.
  262. Roep BO, Stobbe I, Duinkerken G, van Rood JJ, Lernmark A, Keymeulen B, Pipeleers D, Claas FH, de Vries RR. Auto- and alloimmune reactivity to human islet allografts transplanted into type 1 diabetic patients. Diabetes. 1999;48(3):484-490.
  263. Stobbe I, Duinkerken G, van Rood JJ, Lernmark A, Keymeulen B, Pipeleers D, De Vries RR, Glass FH, Roep BO. Tolerance to kidney allograft transplanted into Type I diabetic patients persists after in vivo challenge with pancreatic islet allografts that express repeated mismatches. Diabetologia. 1999;42(11):1379-1380.
  264. Hallé JP, deVos P, Rosenberg L. The bioartificial pancreas and other biohybrid therapies. Kerala, India: Transworld Research Network.
  265. Basta G, Calafiore R. Immunoisolation of pancreatic islet grafts with no recipient's immunosuppression: actual and future perspectives. Curr Diab Rep. 2011;11(5):384-391.

 

Acromegaly

ABSTRACT

 

Acromegaly is a rare condition with an approximate incidence of 3-11 new cases per million of population per year and a prevalence of approximately 60 per million (1). There are approximately 3000 identified individuals in the UK and 15000 in the USA, although it is possible that more cases exist but do not come to clinical attention. More recent studies suggest a higher incidence of acromegaly, up to 6.9 per 100,000 according to Italian data and 7.7 patients per million per year in Iceland (2,3). The condition was named by Pierre Marie in 1886 using the Greek words akron- extremities and megas- large to describe the typical clinical appearance of the condition (4).The disease occurs as a result of excessive secretion of growth hormone. In more than 99% of cases this is due to a benign pituitary growth hormone secreting adenoma. Pituitary carcinomas are exceedingly rare. Extremely infrequently acromegaly occurs as a result of ectopic secretion of growth hormone releasing hormone (GHRH) from a peripheral neuroendocrine tumor (5,6), excessive hypothalamic GHRH secretion (7), or can result after long term exogenous GH abuse (8). Approximately 5% of cases are associated with familial syndromes, most commonly multiple endocrine neoplasia type 1 (MEN1) syndrome, but also McCune Albright syndrome, familial acromegaly, Carney syndrome, and Familial Isolated Pituitary Adenoma (FIPA). Both genders are equally affected and the diagnosis is typically made in adults aged 40-60 years of age. Younger patients often have more aggressive disease due to more rapidly growing adenomas. Acromegaly is associated with multiple systemic complications and a higher risk of mortality if untreated. Very often a multi-modal treatment approach is required to manage the condition, including surgery, radiotherapy, somatostatin analogues, GH receptor antagonist, and dopamine agonist. The management should be individualized to the patient using best practice guidelines, clinical experience, and individual patient circumstances and guided by biomarkers and clinical predictors.

 

PHYSIOLOGY: GROWTH HORMONE- STRUCTURE AND PHYSIOLOGY

 

Growth hormone is a 191 amino acid single chain protein containing two disulphide bonds. It has considerable structural homology with prolactin. Approximately 70% circulates as a 22 kD protein, 10% as a 20 kD isoform and the remainder as dimers or sulphated and glycosylated isoforms (Figure 1). Growth hormone secretion occurs in pulsatile bursts, numbering between 4 and 11 in 24 hours, especially at night, with extremely low or undetectable levels occurring in the nadir between pulses. Thus, a random single serum measurement is very limited as a means of assessing the overall level of secretion. Secretion of growth hormone is governed by both secretory and inhibitory hypothalamic factors. GHRH (growth hormone releasing hormone), ghrelin, and klotho act to stimulate release (9), whereas hypothalamic somatostatin (a 14 amino acid peptide) exerts marked inhibitory effects on GH release. Cortistatin has been found to exert dual, stimulatory and inhibitory effects on GH secretion (10). These stimulatory and inhibitory factors are subject not only to higher influences within the brain but also to peripheral signals such that the overall secretion of growth hormone can vary widely under different physiological and pathological conditions (11) . These are summarized in Table 1.

 

Table 1. Factors Affecting Growth Hormone Secretion (12)

PHYSIOLOGICAL

 

PATHOLOGICAL

Factors which increase GH secretion

Factors which reduce GH levels

Factors which increase GH secretion

Factors which reduce GH levels

Sleep

Overeating

Acute glucocorticoid excess

Chronic excess Cortisol/ glucocorticoids

Fasting

Obesity

Type 1 DM

Hyperthyroidism

Stress

Aging

Renal failure

Type 2 DM

Exercise

Increased IGF-1

Acute use of opiods

 

Hypoglycemia

 

Anorexia

 

Dopamine

 

Depression

 

Increased Amino acids i.e., high protein meal

 

Cirrhosis

 

Reduced Free Fatty acids

 

 

 

Glucagon

 

 

 

Testosterone and estradiol

 

 

 

Figure 1. The 2-D structure of human growth hormone.

Growth hormone circulates in blood bound to a specific binding protein, called GH binding-protein (GHBP). This protein comprises the extracellular portion of the growth hormone receptor (GHR), which is widely distributed and present in most tissues. Activation of the growth hormone receptor occurs when the growth hormone molecule binds two adjacent receptors resulting in dimerization of the growth hormone receptors. Dimerization of the growth hormone receptor results in its activation and binding of the intracellular Janus kinase (Jak 2) tyrosine kinase. The activated JAK2-GHR complex induces multiple signaling pathways responsible for the diverse actions of GH (13,14). These include phosphorylation of a) signal transduction and activators of transcription (STAT) proteins STAT1, STAT3 and most importantly STAT 5, b) SRC family kinases which trigger the MAP kinase pathway, c) insulin receptor substrate (IRS) proteins which activate phosphatidylinositol-3-kinase (PI3K) and Akt pathway, and d) SH2B1, a scaffold protein that upregulates GH action in the actin cytoskeleton (13). Intracellular growth hormone signaling is suppressed by several proteins, especially the suppressors of cytokine signaling (SOCS) 1-3 and protein tyrosine phosphatases SHP1, SHP2 (14,15). While there have been suggestions that GHR polymorphism could play a role in variable responses to the GHR antagonist Pegvisomant therapy, so far studies have not convincingly demonstrated this relationship (16).

Figure 2. The growth hormone molecule binding to the membrane surface growth hormone receptor. Signaling and transduction only occur when adjacent receptors bind the two specific binding sites on the growth hormone moiety to form a dimer.

One of the major proteins induced by growth hormone is insulin-like growth factor-1 (IGF-1)   Although classical endocrinology states that it is hepatic derived IGF-1 acting in an endocrine manner that is responsible for most, if not all, of the effects of growth hormone, it is becoming increasingly clear that local production of IGF-1 acting either in a paracrine (nearby cells) or autocrine (on the same cell) manner also has important biological effects, predominant of which is stimulating cell proliferation and inhibiting apoptosis (17). Elegant gene 'knock-out' experiments have demonstrated that animals with selective hepatic IGF-1 loss have a normal phenotype and growth, despite marked reduction in serum IGF-1 levels (18). Furthermore, patients with severe GH deficiency, perhaps as a result of pituitary surgery, usually have serum IGF-1 levels just below or at the lower end of the normal range. Thus, rather than being the sole effector of growth hormone, serum IGF-1 should perhaps be more accurately regarded as a marker of serum growth hormone concentrations. Circulating IGF-1 does however have important effects in regulating pulsatile growth hormone secretion with IGF-1 acting in a negative feedback fashion suppressing growth hormone release.

 

Central Regulation of GH Secretion

 

GHRH consists of 44 amino acids, the first 27 from the N-terminus being essential for physiological activity (19). GHRH containing neurons are located in the arcuate nucleus and surrounding the venteromedial nucleus which is considered the major site of GHRH activity. Somatostatin producing neurons are predominantly present in the dorsolateral arcuate nucleus and share close synaptic connections with the GHRH neurons (20). While these two hormones form the key components of local autocrine short feedback regulation of GH secretion; it is further modulated by interactions with central neuropeptides. Dopamine, serotonin, norepinephrine, neuropeptide Y regulate GH output through their interactions with GHRH and Somatostatin neurons (20). GH exerts its own local negative feedback along with local IGF-1 feedback. The Neuropeptide Y system contributes to the rise in circulating GH seen in the fasting state (21).

 

A long negative feedback also exists and growth hormone induces hepatic secretion of IGF-1, which in turn inhibits growth hormone release through various mechanisms including modulating GH gene transcription, reducing GH mRNA expression through POUF1 /CREB protein interactions and altering somatostatin release through paracrine activity (20,22,23).

 

Ghrelin and Growth Hormone Secretion

 

GH pulsatility is primarily driven by nutritional status. The potent growth hormone stimulating action of ghrelin is now well established.  Ghrelin was identified as a natural ligand responsible for regulating GH secretion in the acylated form (24). It is a 28 amino acid peptide, which is modified by Ghrelin-O-Acyl-transferase (GOAT) enzyme to mediate its GH secretory action through IP3 signaling pathway (25). Expression of ghrelin is found in many tissues including both the gastrointestinal tract and the CNS, with the strongest concentrations located in the stomach. Ghrelin mediates its orexigenic actions through the vagal nerve stimulation and eventually acting on the hypothalamic appetite center though the noradrenergic pathway (26). Gastric expression of ghrelin is reduced following feeding and increased by fasting, hypoglycemia and after leptin administration. On other hand, leptin seems to variably effect GH secretion depending on the nutritional status. The exact mechanisms of leptin driven GH changes are not well understood (27).

Figure 3. The growth hormone/ insulin-like growth factor-I axis.

Peripheral Regulation of GH Secretion

 

Systemic energy homeostasis is a potent influence on GH secretion, with interactions by glucose, free fatty acids, adipokines, leptin, ghrelin, and insulin (28-30).  Several hormone systems have regulatory effects on growth hormone secretion. Hypothyroidism is associated with low levels of both growth hormone and IGF-1, and in children leads to short stature (31). Thyroxine replacement has been shown to reverse these deficits. Further evidence from studies in rodents indicates that growth hormone gene expression is regulated by thyroid hormone acting through a thyroid hormone responsive element in the promoter region of the growth hormone gene (32,33). GH replacement has been shown to reduce the Free T4 levels and increase Free T3 levels, by the effect on type 2 iodothyronine deiodinase (34). Glucocorticoids are inhibitors of somatic growth both in humans and experimental animals and individuals with either Cushing’s syndrome or taking exogenous corticosteroids have been shown to have reduced growth hormone secretion.

 

Gonadal hormones also play a role in the neuroregulation of growth hormone secretion. In both sexes spontaneous growth hormone secretion is increased during puberty, and reduced in those with delayed puberty (35), suggesting that both estrogen and testosterone influence growth hormone secretion. The estrogen related reduction of IGF-1 seems to be responsible for relatively higher GH levels in females compared to men, with similar IGF-1 levels (36,37). Hypoglycemia is a potent inducer of growth hormone secretion, and insulin induced hypoglycemia remains the most employed provocative test of growth hormone reserve in humans. Hypoglycemia reduces hypothalamic somatostatin secretion facilitating growth hormone release. A delayed rise in GH levels is noted after acute hyperglycemia and is likely a result of GHRH rise (38) In contrast, hyperglycemia suppresses growth hormone secretion from the healthy pituitary. The availability of amino acids as in the post-prandial state stimulates growth hormone secretion whilst elevated non-esterified fatty acid levels suppress growth hormone release.

 

IGF-1: Structure and Function

 

IGF-1 is a single chain polypeptide of 70 amino acids with three intrachain disulphide bridges, coded by a gene situated on the long arm of chromosome (39). It has 48% amino acid sequence homology to pro-insulin, the A and B domains of IGF-1 have 60-70% homology but there is no homology with the C domain. IGF-1 has a specific receptor, which is structurally and functionally very similar to the insulin receptor. It consists of two extracellular α-subunits which are the hormone binding sites and two transmembrane β-subunits which are involved in initiating intracellular signaling. Post-receptor signaling mechanisms are also similar for IGF-1 and insulin receptors, both activating the tyrosine kinase and IRS-1 cascades. IGF-1 can bind to the insulin receptor but with only 1-5% affinity compared to insulin. Under normal physiological conditions it is thought that IGF-1 acts via the specific IGF-1 receptor, but in the presence of high concentrations of IGF-1 there is likely to be cross activation with the insulin receptor. IGF-1 receptors are found on most tissues with the notable exceptions of liver and adipose tissue. Hybrid IGF-1/insulin receptors have now been well documented and sequenced but their role is unclear (40).

 

The majority of circulating IGF-1 is produced by the liver with bone, adipose tissue, kidney, muscle and many other tissues producing a smaller quantity. Plasma concentrations of IGF-1 in the human are regulated by growth hormone, insulin, age, and nutritional state. Bioavailability of IGF-1 is determined by its binding proteins (see below). Growth hormone and insulin are the main regulators of hepatic IGF-1 production. The precise regulation of local IGF-1 synthesis is uncertain, but it is influenced by many other trophic hormones such as ACTH, fibroblast growth factor, and TSH (41).

 

Figure 4. The regulation of growth hormone secretion.

IGF Binding Proteins (IGFBPs)

 

Unlike insulin the majority of IGF-1 circulates in plasma bound to a variety of binding proteins which determine its bioavailability and modulate its biological action (42). The majority of IGF-1 is bound in a 150 KDa complex with IGFBP-3 and form an acid labile subunit ALS (42). This large molecule (termed the ternary complex) is unable to pass through endothelium and acts as an intravascular reservoir of inactive IGF-1. The half-life of IGF-1 in the complex with IGFBP-3 and ALS is 12-15 hours compared with 10-12 minutes for free IGF-1. The exact mechanisms by which IGF-1 is released from the ternary complex to allow access into the tissues is not known; however, IGFBP degrading protease activity has been well documented in many biological fluids and clinical states.

 

Current knowledge suggests that IGFBP-1 and IGFBP-3/ALS are the binding proteins which have the major effects on the bioavailability of IGF-1(40). IGFBP-1 is inversely related to insulin levels, has a circadian variation with the highest levels being found overnight when insulin levels are lowest, and inhibits the hypoglycemic action of IGF-1 (43). Growth hormone secretory status is the main regulator of plasma levels of ALS (42).

 

PATHOLOGY OF ACROMEGALY

 

Acromegaly is most commonly the result of pituitary adenoma and rarely due to non-pituitary neuroendocrine tumor/ neoplasia (NET/ NEN).  Pituitary tumors are commonly monohormonal or plurihormonal in nature with further distinct subtypes. These subtypes of tumors are responsible for their characteristic behavior and can sometimes guide management (44). The common subtypes are discussed below.

 

1) Monohormonal densely granulated somatotroph adenomas are the most common type of GH-secreting pituitary tumors. They have predominance of the large, dense secretory granules which contain GH and appear deeply eosinophilic on staining.  They account for 30-40% tumors and while tend to lead to higher GH levels, they are usually slow growing tumors, in older patients with mild disease, and retain a predictable response to SSA therapy. Somatic mutation in Gs-α subunit of GNAS has found to be the most common abnormality in GH secreting tumors leading to increased cAMP activity (45).

2) Sparsely granulated somatotroph adenomas form the second most common type of tumor. As the name suggests, they are lightly eosinophilic on HE staining due to the increased presence of keratin aggregates and reduced GH containing granules. While they present with lower GH levels, the tumor behavior is relatively aggressive. They tend to be larger, more invasive with higher ki67 proliferation indices and are less responsive to SSA therapy. Further studies suggest reduced expression of E cadherin and SSTR2 in sparsely granulated tumors as factors likely to be responsible for poor response to SSA (44).  

3) Mammosomatotroph adenoma from a monohormonal Pit-1 lineage cells is a common pathology found in younger individuals with acromegaly. They are densely granulated and co-stain for GH and PRL. They behave in a more benign fashion with high GH and PRL levels leading to early presentation, when the tumor is smaller at diagnosis. They respond to SSA therapy similarly to densely granulated tumors (46).

4) Mixed somatotroph and lactotroph tumors are formed of bihormonal cells with variable combination of somatrotroph and lactotrophs. In variable combinations they comprise of sparsely granulated and densely granulated cells all of which express Pit 1. They tend to be less amenable to treatment and are reported to have frequent disease recurrence (47).

5) Mature Plurihormomal Pit1-Lineage tumors frequently immunostain for TSH along with GH, PRL and are found to express GATA3 (48). Clinical presentation includes features of thyroid overactivity with thyrotoxicosis with non-suppressed TSH.

6) Acidophil stem cell adenomata are tumors comprising of immature GH and PRL secreting tumor cells of a single precursor. The histology shows chromophobic or slightly acidophilic cells, with abundant granular cytoplasm of oncocytic distribution (44). Patients present with hyperprolactinemia which is disproportionate to the size of the tumor. They are frequently invasive and less responsive to dopamine agonist therapy.

7) Poorly differentiated Pit-1 lineage tumors comprise of tumor cells with strong expression of Pit-1 and variable expression of estrogen and GATA3. They are polyglonal and spindle shaped poorly differentiated cells which stain variably for GH, PRL, TSH, and alpha subunit. They are usually macroadenomas, with invasion of surrounding structures and high risk of recurrence (46).

8) Pituitary carcinomas are very rare and form less than 1% of cases. They are difficult to distinguish clinically, and diagnosis is confirmed with evidence of distant metastasis and high ki67 index (e.g., >10%) on histology. These tumors frequently require multi-modal therapy, including chemotherapy, temozolamide and radiation (44).

9) Pituitary hyperplasia is suspected radiologically when there is a uniformly enlarged pituitary gland with no distinct focus of gadolinium enhancement. Hyperplasia is confirmed on histology when the pathology shows expanded pituitary acini containing all of the adenohypophysial cell types, but with increased numbers of somatotrophs and/or mammosomatotrophs (46). The histological diagnosis should prompt the clinician to explore for a GHRH secreting tumor elsewhere or consider investigations for specific genetic conditions associated with acromegaly (highlighted in table 4) such as MEN1, Carney Complex, and McCune Albright Syndrome (49).

 

Non-pituitary sources of disease include GHRH or GH secreting central and peripheral tumors. Hypothalamic tumors such as hamartomas, choristomas, gliomas, and gangliocytomas producing GHRH result in pituitary hyperplasia and very often the diagnosis remains elusive until patient has undergone pituitary surgery. Carcinoid tumors secreting GHRH are recognized as rare peripheral cause of acromegaly and usually are bronchial in origin (50). An ectopic location of pituitary adenomas has been reported in the tract of dorsal migration of the adenohypophysial cells. While a significant number of peripheral tissues have been found to secrete GH (51), reports of GH secreting peripheral tumors include lung, pancreatic and adrenal tumors.

 

Table 2. Pathology Associated with Acromegaly (44,52-54)

Pituitary adenoma

Densely granulated somatotroph adenoma

Sparsely granulated somatotroph adenoma

Mixed cell somatotroph and lactrotroph adenoma

Mammosomatrotroph (monohormonal Pit-1 lineage) adenoma

Acidophil stem cell adenoma

Plurihormonal adenoma

Poorly differentiated Pit-1lineage tumor

Pituitary hyperplasia

Pituitary carcinoma

 

Rare: ectopic pituitary adenomas identified in sphenoid sinus or parapharyngeal tissue

Ectopic hormone secretion

Central:

Hypothalamic tumors

Ganglioneuroma

 

Peripheral: Bronchial carcinoid, small cell lung cancer, adrenal tumor, pancreatic neuroendocrine tumor

Exogenous GH replacement or abuse

 

Pseudoacromegaly

Pachydermoperiostosis

IGF signaling pathway diseases

Severe insulin resistance

 

CLINICAL FEATURES OF ACROMEGALY

 

The clinical manifestations of acromegaly evolve gradually over a long time and as a consequence there is a lag time of about 5-10 years, from symptom onset to diagnosis (55,56). In the recent decades, there seems to be some early recognition of the condition, particular in individuals investigated for pituitary ‘incidentaloma’, for example with hypogonadism as a presenting feature (57). One third of the cases, have co-existent symptoms of hyperprolactinemia, which aids in early diagnosis (58).

 

 

Growth hormone secreting pituitary adenomas are frequently (more than 70%) large tumors (macroadenoma, ≥ 10 mm in diameter) which may present with local mass effects such as headache (often severe and out of proportion to the size of the pituitary tumor), hydrocephalus, visual field defects, ophthalmoplegia, or other cranial nerve palsies (59). As the lesion increases in size deficiencies of other anterior pituitary hormones may also occur. Microadenomas (< 10 mm in diameter) are conventionally thought to be less common, but tend to represent one third of the cases (60,61). However patients presenting with pituitary tumors, without clear features of acromegaly may have elevated IGF-1, and GH positive immunohistochemistry on the resected tumor specimen (62) and such silent growth hormone tumors seem to be more common in females with a higher risk of recurrence (63). The term micromegaly is used to describe such clinical presentations (64,65). Recognition of such presentations should prompt the endocrine specialist to consider GH secreting tumors in all presentations, but especially in the younger patient with pituitary tumor. 

 

Hypopituitarism has been found to occur in about 40% cases with variable frequency of hypogonadism, adrenal insufficiency, and secondary hypothyroidism (66,67). Hypogonadism, presenting as decreased libido, infertility or oligo/amenorrhea is a common finding at presentation; it may be due to both gonadotrophin deficiency as well as hyperprolactinemia, either from coexistent excessive secretion of prolactin or from stalk compression. Hypogonadism has been reported even in patients with microadenomas with normal prolactin, thereby suggesting an independent effect of GH hypersecretion (68). Menstrual irregularities, PCOS, subfertility and erectile dysfunction can occur as a consequence of GH excess (69,70). The occurrence of diabetes insipidus in relation to a pituitary adenoma is extremely rare and should raise the possibility of an invasive pathology (71,72).

 

Soft Tissue and Skeletal Changes

 

The most characteristic feature and one that usually precipitates the diagnosis is a change in appearance as a consequence of soft tissue and bony changes. The common changes include coarsening of the facial features, broadening of the nose, thickening of the lips, macroglossia, and prominence of the supraorbital ridges. There is enlargement of the hands resulting in their characteristic 'spade-like' appearance and soft dough-like consistency of the palms. Ring size increases; a sensitive objective assessment of disease activity and response to treatment. Similar changes occur in the feet which become wider with increase in shoe size. Elongation of the jaw results in prognathism which contributes to dental malocclusion, interdental separation, and temporomandibular joint pain (73,74).

 

Greasiness of the skin is a frequent finding with excessive sweating, one of the most sensitive signs of growth hormone excess. Skin tags are a frequent finding, likely related to epithelial cell hyperproliferation in response to IGF-1 (74). Additional dermatological manifestations include hypertrichosis, psoriasis, acanthosis nigricans, and cutis verticis gyrata, with the latter two seen more commonly in severe cases (75). Skin changes are a result of deposition of glycosaminoglycans in the subcutaneous tissue, along with increased proliferation of dermal fibroblasts as a consequence of GH and IGF-1 action (74). These changes tend to reverse after treatment, at least partially if not completely. The lean body mass is higher in individuals with acromegaly, while there is increase in adipose tissue post therapeutic intervention (76). This reversal of body composition tends to stabilize by three months of the surgery (77). In addition to the negative impact on body fat, reversal of GH excess has been found to increase intrahepatic lipid accumulation (78).

 

Generalized organomegaly is not well reported with acromegaly, in contrast some earlier assumptions of the disease process but enlargement of thyroid, prostate, salivary glands, heart, liver and spleen has been recognized (79). Macroglossia, increased thickness of laryngeal structures and vocal cord enlargement increases the risk of anesthesia and makes intubation difficult (80). Ultrasound evidence suggests the presence of increased organ stiffness and commonly reported features include renal cysts, thyroid nodules, multinodular goiter, gallbladder polyps, and polycystic ovaries (79). Mucosal edema and hypertrophy of vocal cord can result in voice changes, but true existence of voice abnormalities is debated (81,82). Patient reported questionnaire and evaluation of voice parameters seem to demonstrate presence of micro perturbations, lower amplitude and poor quality of voice in individuals with active disease (82,83).

 

The skeletal manifestations of acromegaly result from multiple factors which include direct effect of GH, IGF-1, altered calcium phosphate metabolism, hypogonadism, diabetes, and over replacement of steroids (84,85).  Acromegaly results in increased bone resorption and altered bone formation according to various cross-sectional studies, but the effect of GH, IGF-1 on bone health is complex. GH mediates its effect on bone through systemic IGF-1 with action on cortical bone, whereas bone IGF-1 seems to be responsible for cancellous bone health. There is emerging evidence of correlation of sclerostin levels with acromegaly related bone disease (86). The high prevalence of vertebral fractures (VF) in active acromegaly has been known for a long time with some studies reporting incidence as high as 60%, and dependent on the duration of disease and gender (males>females) (84,87).  In a French study, authors have suggested that the skeletal abnormalities are more likely vertebral deformities than true fractures (88). Screening for VF is recommended in all patients with active acromegaly as it has a significant impact on quality of life, morbidity, and development of cardiac and pulmonary complications (85,89). Standard DXA seems to be less reliable in predicting the risk and Volumetric DXA with quantitative assessment of the trabecular bone density seems to provide more reliable information of acromegaly related bone disease (87,90). Unfortunately it is difficult to undertake this test routinely and therefore alternative use of newer methods such as non-invasive 3D-SHAPER and TBS Trabecular bone score assessments may be considered (90). While vertebral changes are most discussed, consequences of acromegaly include development degenerative diseases in all weight bearing and non- weight bearing joints, predominantly shoulder, hip and knees (84,91). The prevalence of radiographic evidence of at least one joint involvement has been found to be as high as 99% (92).  Patients with active disease seem to be have higher prevalence of reduced cortical density at hip compared to patients with non-functioning pituitary adenomas (93). Degenerative joint changes in early acromegaly related arthropathy appear different from usual osteoarthritis, and are noted as widened joint spaces contributed by cartilage hypertrophy and marked osteophytosis, in contrast to standard OA (94). Acromegaly related arthropathy is associated with a significant impact on quality of life and tends to progress despite improvement in disease status (89). Routine use of anti-resorptive therapy is currently not well established, but considering the wider implications of the disease on bone health in active disease, it seems reasonable to consider offering bone protective therapy in patients with early evidence of bone disease (90).

 

Figure 5. The typical facial appearance of acromegaly. Evolution of the appearances over 2 decades

Sleep Disordered Breathing

 

Sleep apnea syndrome (SAS) is a well-recognized manifestation of GH excess and seems to have been variably reported with incidences of 40-80% and about 11.7-20 times higher prevalence than in general population (55,95). Evaluation of the bony changes in the facial skeleton showed significant differences in patients with acromegaly and SAS, compared to patients without SAS. But soft tissue enlargement of upper airways contribute more to the narrowing of the pharyngeal airway space than the craniofacial skeleton changes (95). CT and MRI assessment of upper airways demonstrate pharyngeal hypertrophy and upper airway stenosis correlating with the severity of obstructive sleep apnea (96). Patients with SAS have been found to have more features consistent with metabolic syndrome, such as hypertension and DM compared to patients without SAS (97). Improvement in SAS noted after intervention seems to correlate with positive changes in tongue volume and pharyngeal soft tissue (98). Unfortunately, complete reversal of SAS does not occur and persistent SAS is seen in about 40% of treated cases. It is likely due to effect of additional factors on SAS such as age, male gender, smoking, and obesity (84). A direct inhibitory effect of GH on central respiratory center can result in a non-obstructive central pattern of sleep apnea (99). This phenomenon seems to be much less prevalent than obstructive disease, while a mixed pattern has also been reported (100).

 

Muscular Changes

 

Musculoskeletal pain is typically progressively evident during the disease course, with close to 90% patients reporting pain as a dominant symptom (101). Irrespective of the severity of the acromegaly, pain contributes significantly to reduced quality of life (101). Studies demonstrate that GH excess leads to hypertrophy of the type 1 muscle fibers with variable findings for type 2 fibers (102-104). The predominant abnormality of type 1 muscle fibers strength seems to be responsible for evident muscle weakness noted in high velocity activities (105). While earlier studies consistently reported muscle weakness in acromegaly, a recent study used quantitative measures suggest patients with active acromegaly may have higher proximal muscle strength, but reduced hand grip which normalizes after treatment (103). It is postulated that the difference in findings could be related to pain interfering with true assessment of muscle function (103). Ultrasound evidence showed increased tendon thickness, enthesisitis, soft tissue enlargement but reduced muscle volume in some lower limb muscles in a cohort of thirty nine patients (106). There is emerging evidence that acromegaly has a complex effect on muscle strength and volume (107). Differential interactions of GH, Muscle Ring Factor -1 (MuRF-1), and myostatin seem to be responsible for chronic effects of GH excess on skeletal muscles (108).

 

Neurological Abnormalities

 

Carpal tunnel syndrome is present in approximately 60% of patients at diagnosis but about 80% will have electrophysiological evidence of median nerve neuropathy (109). The pathophysiology is due to swelling of the median nerve itself within the carpal tunnel rather than extrinsic compression from increased volume of the carpal tunnel contents (110). This is well evident on MRI and Ultrasound studies of the median nerve, and seems to correlate with the abnormal nerve conduction studies (110-112). Similar peripheral nerve abnormalities and abnormal nerve conduction studies have been noted in other peripheral nerves including peroneal, tibial, ulnar, sural nerves (112,113)and polyneuropathy is more common in uncontrolled disease (114). Unfortunately it seems some of the changes are not completely reversible, despite normalization of disease markers (115). Patients with active acromegaly have altered cardiac autonomic function, which contributes to the cardiovascular risk (116,117). This seems to respond to active intervention (118). There is not much evidence of central nervous involvement related to the disease (113), with some report of delayed brainstem auditory evoked potentials (119). Restless leg syndrome has been reported to be present in 20% of cases and negatively impacts the quality of life (120).

 

Cardiac Complications

 

Cardiovascular diseases continue to remain one of the most common causes of morbidity in patients with acromegaly and account for 60% of the mortality with this condition (121,122). The range of abnormalities detected at the time of diagnosis have been reported to be hypertension, cardiac hypertrophy, arrhythmias, coronary artery disease, and systolic heart failure, in the order of prevalence (123,124).  Hypertension presents with higher diastolic readings than systolic BP measurements (125). Based on ambulatory BP monitoring readings, it seems that the prevalence of hypertension is about 22% at diagnosis, much lower than earlier studies with single office measurements (126). Anti-natriuretic effect of GH is a direct consequence of GH action on the epithelial sodium channel ENaC in cortical collecting ducts of the kidney (127). This sodium retention leads to volume expansion and further compounded by GH effects on cardiac output, impaired endothelial function, increased peripheral resistance and co-existent sleep apnea are some factors that lead to development of hypertension (128).

 

Acromegaly related cardiomyopathy is a consequence of GH and IGF-1 effect on cardiac myocytes, regulation of cardiac muscle specific gene transcription, and increased fibrosis (129). It has been described to undergo three stages of disease progression (130). In the first phase biventricular concentric hypertrophy is described related to muscle hypertrophy and increased contractility and leads to a hyperkinetic syndrome. As the disease progresses, patients tend to develop diastolic dysfunction with more prominent ventricular hypertrophy. Patients report reduced exercise tolerance and it is common for disease to be diagnosed in this phase. If untreated patients may progress to develop overt diastolic and systolic dysfunction presenting as congestive heart failure in about 3-4% cases and is a poor prognostic marker (123). The presence of left ventricular hypertrophy correlates with disease duration and various studies report prevalence from 11-78% (84,131). Cardiac Magnetic Resonance (CMR) studies have demonstrated variable prevalence of left ventricular hypertrophy compared to echocardiographic studies (132,133). CMR is more reliable in identifying myocardial fibrosis and RV systolic dysfunction then echocardiography (133). Despite the presence of cardiovascular abnormalities associated with ischemic heart disease, recent studies report no increase in prevalence of Ischemic heart disease, in comparison to normal population (55,134).

 

Cardiac arrhythmias have been reported to occur in about 7-40% cases of acromegaly. A wide range of rhythm disturbances described in patients with acromegaly include paroxysmal atrial tachycardia, supraventricular tachycardia, sick sinus syndrome, ventricular ectopic, and ventricular tachycardia (130). A typical acromegaly related left ventricular rhythm disturbance, results from abnormal and dyssynchronous loss of peak contraction of corresponding cardiac segments (135). Mitral and aortic regurgitation have been commonly associated with acromegaly and seem to correlate with the duration of the disease (136). Unlike cardiomyopathy and arrhythmias which improve or even completely reverse with disease control, valvular disease is irreversible and only tends to stabilize with intervention (130).

 

Metabolic Complications

 

Growth hormone is a potent insulin antagonist and acromegaly results in abnormal glucose tolerance in many patients with frank diabetes mellitus in up to 50% cases at diagnosis. Lipid abnormalities, in particular elevation of serum triglycerides, reduced HDL levels, increased small dense LDL particles, and increased lipoprotein-a (Lp(a)) may be an accompanying feature of insulin resistance and is noted in one third of the cases (137,138). Chronic GH excess results development of insulin resistance by several mechanisms. Reduced glucose uptake occurs by increased levels of free fatty acids and reduced expression of GLUT1 and GLUT 4 receptors (139). GH also results in development of a pro inflammatory state in the adipose tissues with alterations of the genes coding visfatin and IL6 (140). The degree of insulin resistance correlates with IGF-1 levels (141) and improves with management of the disease. Development of IGF receptor resistance beyond a threshold for IGF-1 has been reported in states of chronic GH excess leading to further insulin resistance (142). Visfatin and irisin levels have been suggested to correlate with metabolic abnormalities and cardiovascular risk factors (143,144). When choosing treatment, octreotide, and lanreotide have less impact on the glycemic variations, while pasireotide can aggravate hyperglycemia. Pegvisomant has a favorable on the metabolic parameters (145).

 

Table 3. Clinical Manifestations and Complications Reported with Acromegaly

Tumor related local effects

Headache

Visual field defects

Cranial nerve abnormalities

Hydrocephalus

Temporal lobe epilepsy

Hyperprolactinemia

Hypopituitarism

Systemic effects

Skin changes

Hyperhidrosis

Oily skin

Skin tags

Hypertrichosis

Acanthosis

Cutis verticis gyrata

Cardiac

HT

Cardiomyopathy

Valvular heart disease

Arrhythmia

Heart failure

Soft tissues changes

Acral enlargement

Change in voice quality

Visceromegaly (thyroid, prostate, liver, salivary glands)

Neurological

Peripheral nerve abnormalities

Autonomic dysregulation

Lumbar canal stenosis

Narcolepsy

Restless leg syndrome

Orofacial changes

Prognathism

Frontal prominence

Dental malocclusion

TMJ pain

Gingival enlargement

Macroglossia

Pulmonary

Sleep apnea

Restrictive lung disease

Subclinical hypoxemia

 

Musculoskeletal

Vertebral deformities

Kyphosis

Arthralgia and arthritis

Myopathy

Degenerative arthropathy

Calcific discopathy

Hypermobility

Neoplastic

Colon polyps

Thyroid cancer

Breast cancer

Endocrine and Metabolic

Hypogonadism

PCOS

DM, insulin resistance

Hypertriglyceridemia

Erectile dysfunction

 

Renal

Increased GFR

Hypercalciuria

Glomerulosclerosis

 

Hematological

Increased thrombosis risk

Psychiatric

Depression

 

Ocular

Increased risk of diabetic retinopathy

Extraocular myopathy

Glaucoma

Epiphora

 

 

 

Thyroid Abnormalities

 

Patients with GH excess have been demonstrated to have a rise in TSH and T3 levels with no significant relation with the FT4 levels (146). There is a direct correlation of IGF-1, GH levels with thyroid volume. Multinodular goiter is one of the most common thyroid abnormality in patients with acromegaly with frequencies of 69.5 to 79.1% being reported by some authors (147). Patients in remission after surgery have been shown to have change in the consistency of the thyroid nodules, reduced vascularity, and volume (148). Over the course of follow up of patients with active acromegaly, thyroid nodule enlargement seemed to correlate with IGF-1 levels with increased prevalence of differentiated thyroid cancer (papillary thyroid carcinoma) in this subgroup (149,150). Despite the increasing understanding, routine screening for thyroid nodules is not yet recommended but assessment should certainly be considered in patients with a palpable nodule (151).

 

Neoplasia

 

The true risk of cancer continues to remain debated with concerns of heterogeneity in the study population, selection biases, variable screening strategies, and limitations of using standardized incidence ratios as the reporting indices (84). In the recent years, some large cohort population studies suggest higher cancer risk than general population. Acromegaly has been associated increased risk of cancers, particularly colon, kidney, and thyroid cancer in a large Italian survey (152). A similarly large Danish cohort has reported increased incidence of colon, thyroid, breast, gastric, and urinary bladder cancers (153).

 

Animal models and in vitro studies suggest anti-apoptotic and tissue proliferative role of GH and IGF-1. IGF-1 deficiency has been found to result in protection from tumor development. GH signaling pathways and autocrine GH action contribute to tumorigenesis and in colonic tissue this mechanism results in reduced action of tumor suppressor proteins (84,154). Colon cancer has been studied in detail in patients with acromegaly. Patients have a higher prevalence of adenomatous and non-adenomatous polyps than general population (155) with reported trend varying between 6-30% (156). Colonic pathology is related to disease activity with patients with elevated serum growth hormone and IGF-1 levels being particularly prone to developing colonic adenomas (157). Although the exact pathogenesis of these tumors remains uncertain it is likely to involve altered homeostasis of cell numbers within the colonic epithelial crypts; increased proliferation and decreased apoptosis within the crypts of patients with acromegaly have both been documented (158). Colorectal neoplasia in acromegaly has different characteristics compared to the general population, in that the adenomas are more likely to be located in the right side of the colon, tend to be bigger and are more often multiple as well as demonstrating increased dysplasia (159).

 

It is now generally accepted that patients with acromegaly should be regarded as a high-risk group for colorectal cancer and regular colonoscopy screening should be offered to all patients. Current evidence suggests that this should begin at the age of 40 years with the subsequent interval depending both on disease activity and the findings at the original colonoscopy screening (160). In the presence of a polyp (hyperplastic or adenoma) or elevated serum IGF-1 levels screening should be repeated after five years, whilst a normal colonoscopy screening, or serum IGF-1 level within the normal range suggests screening every 10 years may be appropriate. As approximately 30% of lesions occur at the cecum or in the ascending colon, total full-length colonoscopy is required. This should be performed by an experienced colonoscopist, as the cecum is reached in only about 70% of patients in inexperienced hands. Due to their slow bowel transit time and elongated colon, patients with acromegaly require rigorous bowel preparation, often twice that necessary for the patient without acromegaly. Failure to visualize the cecum necessities a repeat colonoscopy or failing this examination using CT virtual colonoscopy (156).

 

Lung Complications

 

Pulmonary complications are common in acromegaly. Total lung volume and residual volume are increased, along with narrowing of both large upper airways and more commonly small airways (161,162). In a large study Storrmann et al reported higher prevalence of small airway obstruction in females. They also highlighted presence of subclinical hypoxemia in patients with acromegaly. The findings did not correlate with levels of IGF-1 of duration of disease (163). Tracheal structural abnormalities have been found to be responsible for large airway disease (164).

 

Ocular and Auditory Complications

 

A wide variety of ocular complications have been reported in patients with acromegaly, apart from visual field defects. The prevalence of proliferative diabetic retinopathy has been variably reported to be higher in patients with acromegaly (165) in some studies, while others suggest it is likely that there is increased retinal vessel branching, and noted no difference in retinopathy rates (166). Extraocular muscle enlargement has been reported by few authors and rarely has resulted in presentation of diplopia (167-169). Studies have highlighted increased intraocular pressure, increased corneal thickness and increased retinal thickness in patients with acromegaly, with rare reports of epiphora (170-172). The association of acromegaly with hearing disturbances has not been well reported. There have suggestions of abnormal bony changes, changes in middle ear pressures and internal acoustic meatus contributing to variable hearing abnormalities (173,174). But findings have not been widely validated and association of acromegaly with hearing loss is not well established (175).

 

Other Systemic Complications

 

Hematological abnormalities are not common with acromegaly. Recent evidence suggests patients with active acromegaly may be at a higher thrombotic risk and this could contribute to cardiovascular risk (176,177). Higher levels of fibrinogen, factor VIII and thrombin tend to result in hypercoagulable state in active untreated disease (178). Rare case reports of polycythemia, myeloma, and Waldenstrom’s macroglobulinemia have been reported (179,180).

 

GH and IGF-1 receptors have been found in kidneys and suggest local autocrine and endocrine activity of GH at the level of nephrons (51). The effect of GH excess on kidneys has not been well described. Chronic GH exposure leads to renal hypertrophy and structural changes to include glomerulosclerosis (181) . Patients with acromegaly have been reported to have increased GFR, reduced renal excretion of sodium, potassium, hypercalciuria, hyperphosphaturia, greater prevalence of microalbuminuria and micro-nephrolithiasis, irrespective of comorbidities (182-185). 

 

Morbidity and Mortality in Acromegaly

 

It is established that uncontrolled acromegaly results in a considerable increase in morbidity with an overall mortality at least two-fold that of the general population (186). In early epidemiological reviews more than 50% of patients had died by the age of 60 years, usually as a result of diabetes, cardiovascular, respiratory or cerebrovascular disease (187). With improved treatment of both the underlying disease and these complications, patients are now surviving longer although may then be susceptible to other complications such as malignancy (188). The determinants of mortality included older age and IGF-1 levels at diagnosis, treatment modality, and malignancy (121). Prolonged diagnostic delay has been found to positively correlate with morbidity and mortality (189). Longitudinal population cohorts indicate a negative impact of female gender on the presence of comorbidities and mortality (37,190).

 

DIAGNOSIS OF ACROMEGALY

 

The diagnosis is made using a combination of clinical examination and biochemical assessment. Serum growth hormone concentrations are typically elevated, and although pulsatility may be reduced, levels may fluctuate widely in acromegaly. Due to the pulsatile nature of growth hormone secretion, a single growth hormone measurement is of little use in either monitoring or confirming the diagnosis of acromegaly. GH levels are affected by age, gender, and comorbid disease states and these factors need to be taken into account when interpreting results. These factors have been outlined in table 1. Due to variations between assays, it is recommended that a standardized and similar performing assay is used for monitoring of the disease activity. Most modern assays detect the highly prevalent GH 22kDa isoform and the most common preparation used to calibrate GH assays is the latest recombinant IRP 98/574 (197). It is useful to be aware of the interferences of the local assay with biotin and Pegvisomant. Various tests used in screening, diagnosis, and management of acromegaly are discussed below.

 

Biochemical Tests at Diagnosis

 

  1. IGF-1 levels: A single serum IGF-1 level has been advocated as being a useful first line test for the diagnosis of acromegaly as it is elevated in the majority of subjects. It is an indirect assessment of growth hormone secretion with approximately 25% of patients having a discrepancy between the mean value of a growth hormone day curve and an IGF-1 level. IGF-1 secretion is subject to several influences including liver and renal dysfunction, nutrition, diabetes mellitus, physiological factors such as age, gender, and the presence of a statistical correlation between its levels and those of growth hormone should not be used as proof that they are interchangeable (198). However, despite these limitations, from a practical point of view, an elevated serum IGF-1 measurement may be useful as confirmatory evidence, assuming that age and sex matched normal ranges are used, and for monitoring treatment (199). Standardized IGF-1 (IS 02/254) is recommended for manufacturers of IGF-1 assays (52,200).
  2. GH day curve: The assessment of growth hormone hypersecretion requires the mean value of serial samples taken throughout the day (e.g., 5 samples over a 12-hour period). The samples should be taken through an indwelling venous cannula to avoid the stress effects of repeated venipuncture. In normal subjects, the majority of values throughout the day are undetectable, but in acromegaly typically each value is measurable, often with a fixed rate of secretion (201).
  3. Oral glucose tolerance test: Failure of normal suppression of serum growth hormone following administration of oral glucose remains the ‘gold-standard’ biochemical test (202). 75 g of oral glucose is given at 9 am to the fasting patient and plasma glucose and serum growth hormone levels are measured at baseline, 30, 60, 90, 120 (and 150) minutes thereafter. In normal subjects, growth hormone levels suppress to undetectable values (typically <0.1 ng/ml) when ultrasensitive assays are used, whilst in acromegaly serum growth hormone remains detectable, and in approximately 30% of cases there is a paradoxical increase (199). In conventional practice failure to suppress serum growth hormone to a level < 0.4 ng/ml following ingestion of glucose supports the diagnosis of acromegaly. The use of this test also detects those patients with impaired glucose tolerance or diabetes mellitus other than individuals with poor diabetes control (203). False-positive results can be seen in conditions where GH levels are elevated such as stress, type 1 diabetes mellitus, cirrhosis, chronic renal failure, during adolescence, and by drug use (L -dopa, heroin, estrogen 201,204).
  4. TRH test: In cases of remaining doubt about the diagnosis of acromegaly, a TRH test can be used (200 mg of thyrotrophin releasing hormone given intravenously with serum measurement at 0, 20 and 60 minutes). In normal subjects TRH inhibits growth hormone secretion with a fall in serum concentration, whilst approximately 60% of patients with acromegaly demonstrate a paradoxical rise in growth hormone levels (205). In mild disease with relatively low GH levels, TRH stimulation has been suggested to confirm early diagnosis (206).
  5. Others: IGFBP3 levels correlate with mean 24 hour GH levels and IGF-1 levels, but due to the wide overlap with normal and diseased individuals, is a poor parameter for measuring disease activity (207). In the rare patient in whom a non-pituitary etiology is suspected, measurement of serum GHRH may be performed, typically with very elevated levels occurring in ectopic GHRH syndromes such as neuroendocrine tumors. Basal serum prolactin should also be measured as prolactin may be co-secreted with growth hormone in up to a third of patients with acromegaly, which often indicates therapeutic responsiveness to the use of dopamine agonists. In those with hyperprolactinemia the presence of macroprolactin should be excluded (208).

 

In patients with atypical clinical symptoms such as GI symptoms, hypoglycemia, renal stones, clinicians should consider exploring possibility of coexisting genetic tumors or extra pituitary source of GH abnormality, such as carcinoid tumors.

 

Biochemical Tests Used to Define Disease Activity to Guide Outcomes

 

  1. Octreotide test: Acute challenge with octreotide 100mcg sc dose can help predict response to SSA when hourly values are measured over 6 hours (209,210). Not all clinicians perform this test and many treat with a long-acting SSA independent of a challenge test (211).
  2. IGF-1 levels: IGF-1 levels can take over three months to normalize in the post-operative period and until then cannot be reliably used to guide management. In longer term the aim of treatment is to keep levels within the normal ranges (52). In patients with Pegvisomant therapy, IGF-1 is the variable that guides response to treatment (212).
  3. Random GH levels: Random levels can be used to define surgical cure and values as early as day 1 can be undertaken if no pre-operative GH suppressing therapy was used, bearing in mind the effect of post-operative stress. A serum GH <0.4 µg/L favors disease remission and a level <1 µg/L indicates good control and normalization of the mortality risk (151).
  4. OGTT: OGTT can be undertaken to evaluate post-operative outcome if random GH values >1µg/L. A nadir GH cut off of <1µg/L is associated with better long-term outcomes and would be considered as good control. Endocrine Society guidelines suggest GH nadir of less than 0.4mcg/ L could be used to define disease remission (151) It has been suggested that post-operative OGTT at 3 months is more appropriate than an early test in 3 weeks and is likely to help avoid false positives (213).
  5. Other tests: The GH day curve test is rarely required for long term monitoring. The mean GH value of <2.5µg/L correlates better with disease control but it is unreliable in patients who have undergone radiotherapy due to alterations of GH pulsatility pattern (214). Post-operative TRH test has been suggested to predict long term disease remission, but again is rarely required (211).

 

Radiological Assessment

 

Historically the diagnosis of pituitary tumors causing acromegaly was made on the basis of changes to skull bones with demonstration of enlargement of the fossa. With advances in radiology, pituitary MRI with gadolinium enhancement is considered the optimal modality and should be undertaken to determine size of the tumor, define tumor characteristics and assess threat to surrounding structures. At diagnosis, more than 70% of patients with acromegaly have a macroadenoma (≥10 mm in diameter) which often extends laterally to the cavernous sinus or superiorly to the supra-sellar region. Younger patients often present with more aggressive disease, with more invasive tumors which often extend inferiorly. On T1 weighted images the pituitary adenoma tends to be of lower signal intensity than the surrounding normal gland and enhances less briskly than the normal gland after injection with intravenous gadolinium contrast. Tumors >15mm, supra-sellar extension, and cavernous sinus extension of the tumor has been associated with lower rate of surgical cure (215). Radiologic grading of pituitary tumors using KNOSP classification is widely used in predict disease invasiveness and tumor response. Some studies report cavernous sinus invasion as the strongest factor predicting surgical outcome (216,217).

 

In the last decade there is substantial evidence to suggest that hypointensity on T2 weighted MRI is suggestive of good response to somatostatin analogue SSA therapy (218,219). Based on histological characteristics, hypointense T2 weighted tumors correlate with a densely granulated histological subtype and tend to be less aggressive in behavior (220,221). In the post-operative phase, MRI should be considered approximately 3 months after surgery to allow post-operative changes to settle. Subsequent surveillance scans should be guided by biochemical markers as they guide correlation with tumor recurrence. Unless concerned, unenhanced imaging may be preferred for patients expected to require long term surveillance to reduce exposure to gadolinium (222). There are limitations of MRI when it comes to evaluation of persistent or residual disease. In these cases, use of C11 Methionine Positron Emission technology MET-PET with co-registration of volumetric MRI has been found to provide valuable information to guide repeat surgery or targeted gamma knife surgery (223,224). In patients with previously reported empty sella or residual parasellar tissue where surgeons would struggle to consider intervention, the information provided by MET-PET has been found to be useful with positive outcomes. Other tracers that have been studied and reported to guide management include N13 ammonia PET, DOTATATE PET (225,226).

 

In rare cases of ectopic GHRH secretion, a pituitary MRI can sometimes help differentiate between a pituitary adenoma and hyperplastic pituitary tissue (53). The use of somatostatin receptor scintigraphy (particularly Gallium Dotatate) is useful to correlate the source of ectopic hormone production to an unexpected finding on a conventional body imaging.

 

Figure 6. Enlargement of pituitary fossae on lateral skull x-ray.

Figure 7. MRI demonstrating a somatotroph macroadenoma of the pituitary gland.

 

Neuro-Ophthalmological Testing

 

Neuro-ophthalmological assessment should be undertaken in all patients with macroadenomas, especially where tumor is visibly contacting the optic chiasm. At the initial consultation visual acuity should be assessed with the use of Snellen charts and fundoscopy performed to exclude optic atrophy, retinal vein engorgement, or papilledema from pressure on the visual pathways. Visual fields may be assessed by confrontation using a red pin. Patients with any clinical symptoms or evidence of optic chiasmal compression from imaging studies require formal assessment of visual fields with formal perimetry or visual evoked responses, stimulating each half field in turn.  Optical coherence tomography should be used to assess retinal nerve fiber layer as a marker of chiasmal damage.

 

Although permanent loss of vision and/or visual field defects usually result from long standing optic chiasmal compression, the shorter the time of compression the easier and more complete is the reversal of any visual field deficit. Surgical decompression may result in rapid improvement in visual fields within hours or days, although the presence of optic atrophy reduces the likelihood of this occurring. Because onset is often insidious, patients may be unaware of any alteration in their vision, although once documented its presence requires them to inform the vehicle licensing authority as driving ability may be impaired. An exception to this usual gradual deterioration is pituitary hemorrhage when visual loss may be sudden with a loss of central vision and development of bitemporal field defects and possible ophthalmoplegia often accompanied by changes in higher mental function.

 

Assessment of Pituitary Function

 

Assessment of the integrity of the other pituitary hormones needs to be performed by a combination of the appropriate basal and dynamic tests. These are mentioned in other Endotext chapters. Prior to and following pituitary surgery, both residual pituitary function and the growth hormone secretory status should be evaluated. Basal endocrine testing for early morning cortisol, thyroxine, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, testosterone/estrogen, prolactin, and serum & urinary osmolality, should be performed. Where there is doubt, a provocative test should be made of ACTH reserve.

 

Histological Assessment

 

Histology provides valuable information to determine tumor characteristics. Keratin staining is essential to distinguish between densely granulated and sparsely granulated tumors. Further use of immunohistochemical IHC staining modalities is increasing found to help understand tumor behavior. Details of various histological subtypes are highlighted in table 2. IHC staining for different transcription factors Pit-1, SF-1, or Tpit along with hormonal staining is used to understand the tissue of origin. Ki67 labelling index is widely used to determine proliferation rates, though it does not always predict tumor behavior. Evaluation of somatostatin receptor SSTR expression is used to guide response to SSA therapy. Additional immunostaining modalities used in recognition of familial tumors are menin, p27, AIP and SDH expression (227).

 

Genetic Testing

 

Over the last decade, we have seen an increasing understanding of the role of specific genes and mechanisms responsible for development of pituitary tumors. The genetic disorders associated with acromegaly are summarized in table 4.

 

The most exciting developments relate to recognition that aryl hydrocarbon receptor interacting protein (AIP) mutations can account for familial pituitary adenomas. Such cases may present in younger patients with more aggressive tumors, including somatotroph adenomas. Testing for AIP mutations (FIPA) should be considered for any patient with a family history of pituitary tumor, especially GH and/or prolactin secreting tumors (228).  This is particularly so, in those presenting at younger age (<30 years) with aggressive pituitary adenoma. There is some evidence that the response to somatostatin analogue treatment is reduced in acromegaly associated with AIP mutation (229).  Data from the German Pituitary Registry suggest that even in younger patients with acromegaly that the prevalence of AIP mutations is low (<5%) (230). X-LAG X-linked acrogigantism mutation is reported to lead to about 80% cases of pre-pubertal acromegaly (231). GH secreting adenoma is reported to occur in approximately 6% of patients with MEN1 (232).  In practice patients presenting at a young age with acromegaly, co-existent hypercalcemia, and certainly those with a family history of pituitary tumors should be considered for MEN1 and AIP gene sequencing, respectively.

 

Table 4. Genetic Disorders Associated with Acromegaly (233,234)

Genetic condition

Gene implicated

Inheritance

Clinical features

X-LAG

GPR101

X-linked dominant or sporadic

Mostly females

Age <5 years

Hyperprolactinemia at diagnosis

Can be de-novo and family history may be absent

Familial isolated pituitary adenoma

AIP

Autosomal dominant or sporadic

Younger males

Higher GH levels

Poor response to SSA therapy

MEN1

MEN1

Autosomal dominant or sporadic

Presence of primary hyperparathyroidism, pancreatic disease along with pituitary adenoma

MEN4

CDKN1B

Autosomal dominant

Presence of pituitary and

parathyroid neoplasms with pheochromocytomas, thyroid and other tumors

Carney complex

PRKAR1A

Autosomal dominant

Skin pigmentation, Atrial myxoma, GH or PRL excess, Cushing’s due to primary pigmented nodular adrenocortical disease

Somatrotroph hyperplasia or multifocal adenoma

Mc Cune Albright

GNAS

Autosomal dominant or sporadic

Average age of diagnosis 23 years

Acromegaly seen in 20-30% cases

Hyperprolactinemia present at diagnosis

Café-au-lait spots

Peripheral Precocious puberty

Somatrotroph hyperplasia

SDH mutation

SDHx

Autosomal dominant or sporadic

Phaeochromocytoma, paraganglioma and primary hyperparathyroidism present

 

Neurofibromatosis

NF1

Autosomal dominant

Few case reports of children and adolescents with optic glioma, rarely adenoma reported

Mechanisms unclear

 

Investigations for Complication Screening

 

Acromegaly is associated with a significant number of co-morbidities most of which are present at time of diagnosis. Once the diagnosis is established it is essential to evaluate for complications such as DM, hypertension, dyslipidemia, osteoporosis, sleep apnea, carpal tunnel syndrome, quality of life, and neoplasia.  A suggested screening approach is outlined in table 5.

 

Table 5. Suggested Strategy for Screening for Complications (52,145,222)

Co-morbidity to evaluate

Screening test

Frequency

Hypertension

Measurement of BP

Ambulatory BP monitoring in selected cases

6 monthly

DM

Hba1c, FBG

OGTT in selected cases

At diagnosis and 6 monthly if normal

OSA

Clinical evaluation and Epworth scale Polysomnography for confirmation

At diagnosis and annually

Cardiomyopathy

ECG

Echocardiography

At diagnosis and 3-5 yearly if normal

Dyslipidemia

Lipid profile

At diagnosis and 6 monthly

Colon polyps

Colonoscopy

At diagnosis in patients >40 years age, and 10 yearly if normal, 3-5 yearly if polyp noted and IGF-1 elevated

Thyroid nodules

Clinical evaluation

Thyroid US guided by examination

Annually

Vertebral disease

Bone morphometric study using thoracic x-ray, thoracic and lumbar spine x-ray

DEXA

 

At Diagnosis and yearly, guided by symptoms

Cerebral aneurysm

Cerebral MR angiography

Infrequently

QOL

ACROQOL

Annually

 

DIFFERENTIAL DIAGNOSIS

 

Patients with tall stature are frequently suspected to have acromegaly, but absence of soft tissue changes and normal biochemistry should prompt investigations for alternative causes such as Marfans syndrome, homocystinuria, or a familial trait. Pseudoacromegaly or acromegaly mimics are rare disorders with clinical manifestations strongly suggestive of GH excess, but without any biochemical evidence favoring the diagnosis. Pachydermoperiostosis is reported more commonly in males and has an autosomal recessive inheritance. It has been reported secondary to HPGD mutations or SLCO2A1 mutation, which lead to an increase in prostaglandin E2 (235).  

 

MANAGEMENT OF ACROMEGALY

 

Given the chronic nature and associated significant increased morbidity and mortality of acromegaly, treatment is required for almost all patients. Three modalities of treatment are available: surgery, pituitary irradiation, and medical therapy. All of these have advantages and disadvantages and more than one modality is frequently needed, sometimes all three. The decision as to whether to treat and the modality employed must be based on a number of factors, including patient age and general health, wish for fertility, severity of disease and any associated complications, and the risk/benefit ratio of the proposed treatment modality. The goals of treatment are summarized in Table 6.

 

Consensus guidelines define goals for treatment of acromegaly.  These include achieving an age-matched normal range IGF-1 and GH <0.4 mcg/L (236). 

 

Table 6. Acromegaly- Aims of Treatment

1. Removal of the pituitary tumor and resolution of mass effects

2. Relief of the symptoms and signs of acromegaly

3. Restoration of normal rates of secretion of growth hormone and IGF-1

4. Maintenance of normal anterior pituitary function

5. Prevention of recurrence

6. Assessment and treatment of chronic complications

 

Whilst the general principles of these aims are accepted by all endocrinologists, there remains considerable controversy as to the degree of growth hormone reduction that should be the target and what level should be regarded as normal. The use of sensitive growth hormone assays has demonstrated that abnormal patterns of growth hormone secretion can remain despite reduction in mean circulating concentrations to extremely low levels, and thus complete restoration to normality is often not achieved. Early epidemiological reviews, particularly those documenting the results of surgery, tended to regard a mean level of less than 5 ng/ml as being satisfactory. It has become clear in recent years that the excess mortality associated with acromegaly can be significantly reduced and indeed restored to that of the normal population by aggressive treatment and reduction of serum growth hormone concentrations to a mean level of less than 1 ng/ml and/or a serum IGF-1 within the aged-matched reference range. Thus, rather than using the word cure, it is may be more appropriate to consider an average growth hormone concentration of ≤ 1 ng/ml as representing a "safe" level, whereas current consensus is to aim for <0.4 ng/ml (236). 

 

Surgery for Acromegaly

 

Trans-sphenoidal surgery is the initial treatment of choice for most patients. Originally performed by Harvey Cushing in 1910, the lack of adequate visualization prevented its reintroduction for routine use until the mid-1970's. With modern equipment and in experienced hands, it is a safe procedure with a low complication rate and mortality of less than 0.5%. The most commonly used approach is with the patient in a semi-reclining position via a mid-line nasal route. Using a sub-labial or direct nasal approach, the mucosa is cleaved off the nasal septum providing access to the sphenoid sinus and subsequent removal of the fossa floor. A less satisfactory alternative approach is via the ethmoidal sinus. Pituitary adenomas are usually soft and easily removed with curettes although firmer and larger tumors may require piecemeal removal. Using this technique, even tumors with a significant suprasellar extension can be removed via the trans-sphenoidal route, although massive tumors may require a craniotomy. Such transcranial surgery is however associated with increased morbidity and mortality and is rarely required. More recent surgical techniques include the use of intra-operative MRI (237) and intra-operative growth hormone measurement (238). The development of endoscopic trans-sphenoidal surgery offers several advantages over the conventional technique, and is the now the method of choice. Reported advantages over the microscopic technique include superior tumor clearance, especially suprasellar extension, less surgical morbidity, fewer complications, and reduced post-operative discomfort (239), though evidence suggests endoscopic surgery and microsurgery yield similar outcomes in the most experienced hands (240).  Endonasal endoscopic surgery, with ability to resect cavernous sinus located adenoma will increasingly be standard of care in pituitary surgery.

 

A key recent development in the management of acromegaly internationally is the formalization of a team approach, with endocrinologist, neurosurgeon, specialist nurses, oncologists, radiologists, and histopathologists increasingly working as a single-team, making consensus decisions in a timely and coordinated fashion. There is general acknowledgement that functioning pituitary tumors are best managed in centers with larger volume and experience of rarer conditions (236). Certainly, this practice is increasingly the case in Europe and the USA.

 

The success rate of trans-sphenoidal surgery depends on several factors: (i) the size of the tumor, (ii) pre-operative growth hormone values and (iii) the skill and experience of the surgeon and (iv) most importantly cavernous sinus invasion. A predictive model using age, KNOSP classification, and pre-operative GH levels has been proposed to predict surgical remission and guide pre-operative medical management and long term management (241). Although different series have often used different criteria to determine success rates, in experienced hands post-operative mean growth hormone levels of less than 1 ng/ml should be achieved in 70%-90% of microadenomas and 30%-50% of macroadenomas (52). Pre-treatment of patients with somatostatin analogues before trans-sphenoidal surgery is increasingly becoming standard practice, even if early surgery is being planned, as it results in significant shrinkage (approximately 50%) of the adenoma and may improve the subsequent surgical cure rates (242).

 

Complications of trans-sphenoidal surgery include diabetes insipidus, CSF rhinorrhea, meningitis, and hypopituitarism. Diabetes Insipidus is usually transient but may be permanent in approximately 5% of cases depending on the criteria for its diagnosis. A serum osmolality of greater than 295 mosmols/l with a simultaneous urine osmolality of less than 150 mosm/l is confirmatory. It responds well to desmopressin (DDAVP, subcutaneous, oral, or intranasal).

 

Radiotherapy in Acromegaly

 

Radiotherapy in the management of pituitary disorders including acromegaly is discussed in detail in another Endotext chapter. Pituitary irradiation is usually used as an adjunct to pituitary surgery when growth hormone levels remain elevated. In elderly patients or those unfit for surgery, it may rarely be used as first-line therapy (243). There are several techniques that have been used: conventional mega-voltage external irradiation, stereotactic single high dose irradiation, interstitial implantation of yttrium 90 seeds, and whole particle proton beam therapy. Only the first two will be discussed here.   

 

Conventional mega-voltage irradiation has been in routine use for over 40 years and consequently there is a wealth of experience principally relating to it being both a safe and effective technique. A linear accelerator is used as the source; less satisfactory is a cobalt source. Irradiation is focused onto the pituitary fossa using modern CT/MRI imaging and planning, which allows for accurate dosimetry and minimal variation in the daily dosage to surrounding structures, using IMRT. This is particularly so for the optic chiasm, damage to which is avoided by the use of daily fractions of less than 200 cGy. The majority of centers advocate a total dose of 4500 cGy given in 25 fractions of 180 cGy over 5-6 weeks via a minimum of three fields (one frontal and two temporal). Numerous studies have confirmed the efficacy of such mega-voltage irradiation with a 50% fall in growth hormone values occurring in the first two years, regardless of basal levels, followed by a continuing exponential decline thereafter (244). The majority of patients therefore do eventually achieve a level of less than 2 ng/ml, although the interval to reach this depends on the baseline levels. A similar response is seen with IGF-1 with approximately 60% of patients eventually achieving a normal serum level after 10 years. Although it is recognized that pituitary irradiation is associated with several potential adverse consequences, these are rare when irradiation is delivered properly, other than an increased prevalence of hypopituitarism. At ten years after irradiation, approximately 60% of patients are hypogonadal, 50% ACTH deficient, and 40% requiring thyroxine replacement. However, the prevalence prior to irradiation, either due to the pituitary tumor itself or previous surgery should be taken into account, with baseline figures being 40% hypogonadal, 35% ACTH, and 15% TSH deficient (244). Other concerns include development of secondary tumors in up to 2% of cases, radiation induced optic neuropathy in up to 5% cases, cerebrovascular events in up to 20% of cases over 20 years duration, brain necrosis, and psychocognitive impairment (245-247).

 

Stereotactic single high dose pituitary irradiation using either the gamma knife (radiosurgery) or stereotactic multiple arc radiotherapy (SMART) has received increasing attention in recent years as an alternative to conventional irradiation. These techniques permit the delivery of a single high dose of irradiation to a previously mapped area whilst also ensuring a rapid reduction in radiation exposure to surrounding structures. Median dose delivered is 15-35Gy and invariably achieves good tumor control at 5 years follow up and about 50% biochemical remission at 5 years (247)Care needs to be taken with tumors close to the optic chiasm. Initial impressions suggest that growth hormone levels fall to normal earlier than after conventional radiotherapy, but that hypopituitarism occurs just as often (248). Side effects of secondary brain tumors and cerebral vasculopathy seem to be lower but long term studies are awaited (247). Although the stereotactic technique has clear advantages over conventional external irradiation in terms of precise mapping to a specified tumor volume, it may not encompass tumor tissue that is not visualized radiologically. This is in contrast to conventional irradiation which is usually configured to encompass the whole of the pre-operative tumor volume, and thus will treat tumor beyond the resolution of imaging techniques. It is for this reason that stereotactic irradiation should be seen as complementary to conventional irradiation.

 

Medical Therapy of Acromegaly

 

Three different types of medical therapy are currently used in the treatment of acromegaly, dopamine agonists, somatostatin analogs and growth hormone antagonists.

 

DOPAMINE AGONISTS IN THE TREATMENT OF ACROMEGALY

 

From their discovery and synthesis in 1971 until the introduction of somatostatin analogs in the mid-1980’s, dopamine agonists, such as bromocriptine, were the sole medical therapy for acromegaly. However, they are relatively ineffective and whilst approximately 80% of patients will show a reduction in growth hormone levels, only about 10-15% achieved a mean level of less than 2 ng/ml (249). Furthermore, the doses required, often 20 - 30 mg of bromocriptine per day, are much higher than those needed for prolactin-secreting pituitary adenoma. Consequently, the side effects of nausea, headache, dizziness, postural hypotension, and nasal stuffiness tend to be worse, although can be minimized by taking the drug in the middle of a main meal to slow absorption and most patients will demonstrate tachyphylaxis. Unlike in patients with prolactinomas (where an excellent treatment response is expected), there may be only a modest reduction in tumor size but this is usually insignificant. Cessation of treatment results in rebound growth hormone hypersecretion. The use of bromocriptine in acromegaly is limited. The development of the long-acting dopamine agonists such as cabergoline offered greater convenience and reduced side effects, although again high doses of up to 4 mg per day may be needed (250). A meta-analysis has demonstrated that its use can achieve normalization of IGF-1 levels in 34% of patients (251). There are no accurate predictive tests as to which patients will respond to dopamine agonists, but it has a place in the management of mixed growth hormone and prolactin secreting tumors and patients with mild IGF-1 elevation. Combination therapy with SSA is frequently used in patients with co-secreting tumors with good effect (252).

 

Fig 8. Plasma IGF-1 and GH responses to dopamine agonist suppressive therapy in patients with pure GH secreting tumors. The upper squares indicate the pretreatment levels and lower squares correspond to the concentrations obtained by progressively increasing the weekly dose of cabergoline i.e., 1.0, 1.75, and 3.5 mg, respectively. Note the log scale for GH.

 

SOMATOSTATIN ANALOG TREATMENT OF ACROMEGALY

 

The development of octreotide (Sandostatin, Novartis, Basel, Switzerland) a synthetic somatostatin analog, represented a major advance in the treatment of acromegaly. In contrast to the short half-life of native somatostatin (approximately 90-seconds), the 8 amino acid octreotide has a half-life of about two hours. Following a single 100 mcg dose, there is prolonged suppression of growth hormone which lasts for several hours, and indeed this response to a single dose can be used to predict the long-term efficacy of octreotide. It is administered by subcutaneous injection and thus a thrice-daily regimen results in stable drug concentrations and maximal effect. More than 90% of patients show a reduction in growth hormone levels, with approximately 50-60% achieving levels of less than 2 ng/ml and a normal serum IGF-1 level. The usual doses are between 100-200 mg three times daily although occasional patients may require higher doses. This biochemical improvement is matched by rapid clinical improvement. The efficiency of octreotide and other somatostatin analogs (SSAs) such as lanreotide is linked to their preferential binding of the human somatostatin receptor type 2 (SSTR2) with reduced or absent binding of SSTR1, SSTR3, SSTR4 or SSTR5. Somatostatin analogs also have additional and independent, but poorly understood, analgesic properties on the headache associated with acromegaly.

 

Since the introduction of short-acting octreotide, depot formulations of somatostatin analogs have become available. These consist of the active drug incorporated with microspheres of biodegradable polylactide and polyglycolide polymers which allow the slow release of analog after intramuscular injection. There are currently three such preparations available, octreotide LAR (Sandostatin LAR, Novartis) which is given at a variable dose of 10 mg, 20 mg or 30 mg at recommended four weekly intervals, lanreotide (Somatuline Autogel, Ipsen Biotech, Paris, France), which is given as a single dose of 60-120 mg every 28 days as a sub-cutaneous depot formulation, and the more recently licensed Pasireotide LAR.   Pasireotide has increased affinity for SSTR5, and this has led to a license for the treatment of Cushing’s disease in addition to acromegaly.  The SSA medications are also used in the treatment of neuroendocrine tumors arising outside the pituitary gland, in particular small bowel carcinoid tumors and pancreatic neuroendocrine tumors.  Sandostatin and Lanreotide Autogel are of similar efficacy in suppression of growth hormone and IGF-1 with safe growth hormone levels (<2 ng/ml) occurring in approximately 60-70% of patients (253), although a meta-analysis of patients unselected for somatostatin responsiveness indicated that normalized IGF-1 levels and safe growth hormone levels occurred in a higher proportion of LAR treated than lanreotide treated patients (254).

 

Regardless, of the comparative effects, there is variability in individual patient’s sensitivity to these analogs and more than 90% of patients who achieve adequate control with 4 weekly octreotide LAR injections will also do so with 6 weekly injections (255). Consequently, careful dose titration needs to be performed on each patient. This is particularly important given the cost of these depot formulations; in the UK, the approximate annual cost of octreotide LAR given 4-weekly is £8000 for 10 mg injections, £11000 for 20 mg and £14000 for 30 mg, whilst the cost for lanreotide Autogel 90 mg is approximately £10000 per annum.  Biosimilar agents will increasingly become available perhaps with reduced cost.

 

Pasireotide is a novel cyclohexapeptide somatostatin analogue which is selective for SSTR2, 3 and 5, but also shows increased binding to SSTR1 compared to octreotide (256). The extended receptor affinity of pasireotide has led to it being referred to as a “second generation somatostatin analogue” with the original depot formulations being termed “first generation” analogues.  More recent clinical trial data relating to Pasireotide in acromegaly indicates that this agent is modestly more potent than Sandostatin LAR in achieving control of GH and IGF-1, has long term safety, and also has a place in the management of seemingly octreotide resistant disease (257).  A Phase III study showed that in new presentations of acromegaly achievement of control of GH and IGF-1 is superior with pasreotide (over octreotide) with about 20% patients achieving complete remission at 6 months, in a group resistant to first generation SSA therapy, suggesting that pasireotide may replace the earlier SSAs in treatment strategies in the future (258). Recent data from the PAOLA extension study showed that of the patients who achieved biochemical control at some point, 65.6% cases did so after 6 months of treatment. Increasing dose from 40-60mg allowed better remission rates (additional 28%) with reasonable safety profile (257). The drug seems to be particularly useful in the management of severe headaches in patient with acromegaly.

 

Oral octreotide as an agent coupled to a transient gut absorption enhancer and has been very recently approved by the FDA. Phase 3 studies have shown that it helps achieve about 65% control of IGF-1 and GH when switched from injectable SSA and sustains the benefit in about 90% individuals for at least 13 months of follow up (259). It is prescribed in the dose of 40-80mg per day and studies have demonstrated better absorption in a fasting state (260). The most commonly reported adverse effects include headache, nausea, and arthralgia.

 

The side effects of somatostatin analogs are related to the widespread distribution of somatostatin and include effects on the gastrointestinal system, comprising colic type abdominal pain, diarrhea, flatulence, and nausea, although these tend to resolve with time. In the long-term gastritis occurs in a significant proportion of patients and perhaps most significantly gallstones form in approximately 50% of patients after two years of use. This is due to both an inhibition of gall bladder contraction and alterations in the composition of bile with cholesterol supersaturation. However, perhaps due to the gall bladder paresis, the majority of these remain asymptomatic. The effects of SSAs on glucose metabolism are multifactorial. While they improve insulin sensitivity by reducing growth hormone levels, they also exert direct inhibitory actions on insulin secretion by the pancreatic cells. The net result is normal glucose tolerance in the majority of patients. With their improved patient convenience, there have been suggestions that these depot formulations should be used as first-line treatment for acromegaly. However, their increased cost and the need for continuing treatment should be borne in mind. At present, there remains general consensus that whilst they may have a role prior to surgery to try and decrease tumor size, their major place is post-operatively as an adjunct to irradiation whilst waiting for growth hormone levels to fall. Provisional evidence suggests that treatment of acromegaly with somatostatin analogs prior to surgery improves the cardiovascular risk and respiratory status and may therefore have a place in larger and invasive tumors (261,262). Patients who remain uncontrolled despite the use of these somatostatin analogs may gain additional benefit with the addition of a dopamine agonist, but this is the exception rather than the rule. The incidence of hyperglycemia or diabetes is higher when patients are treated with pasireotide. This and the cost of the drug have thus far limited its use in the UK, though other health care economies have more readily incorporated pasireotide into the acromegaly treatment algorithm. 

 

GROWTH HORMONE ANTAGONISTS IN ACROMEGALY

 

The development of Pegvisomant, the novel growth hormone receptor antagonist, is a major advance in the treatment of acromegaly. The development of this molecule utilizes the knowledge that the growth hormone molecule contains two distinct sites which bind to two corresponding unique sites on the respective growth hormone receptor dimer. Pegvisomant is a modified recombinant growth hormone molecule which has increased affinity to the first growth hormone receptor binding site but with decreased affinity to the second binding site. Thus, receptor dimerization and subsequent signal transduction is prevented. Its conjugation with polyethylene glycol (PEG) increases its molecular size, prolongs its half-life and reduces its antigenicity. Based on long term experience, some authors propose Pegvisomant to be most effective medical therapy to date and suitable as first line intervention in selected cases (263). In a study of 152 patients treated for up to 18 months, normalization of IGF-1 occurred in 90% of patients, although doses of up to 40 mg a day were required (264). Growth hormone levels cannot be measured in routine assays as the drug itself interferes with growth hormone assays and pituitary-derived growth hormone increases modestly. Pegvisomant is currently administered as a daily subcutaneous injection of approximately 1 ml in volume. Theoretical concerns exist regarding the increase in circulating growth hormone levels due to the loss of any negative feedback effects on the tumor, but although experience is still limited there is no evidence to date of risk of pituitary tumor growth (263).

 

Pegvisomant is generally well tolerated although abnormalities of liver function occur in some patients. Its major use is for patients who are resistant to SSAs, either as a sole agent or as an additive agent. A study observed that the combination of 4-weekly octreotide LAR and weekly Pegvisomant normalized IGF-1 in more than 90% of patients with active disease who were not controlled with octreotide alone (265). Other suggestions for its use have been in patients with diabetes or impaired glucose tolerance, in whom SSAs might worsen glycemic control. Higher doses may be required in patients with severe disease, DM, and obesity. However, the change in dosing frequency and additional cost needs to be weighed against the use, if required, of simple oral hypoglycemic agents. The major drawback of Pegvisomant other than its usual requirement for daily injection, as opposed to the 4-6 weekly administration of SSAs, is its cost of approximately £3000 per per month, which can amount to £36000 per annum for patients resistant to SSAs (266).

 

A combination study demonstrated improved IGF-1 control with Pegvisomant and cabergoline, an approach which might enable a lower dose of the Pegvisomant to be used with reduced costs (267).  Emerging data suggest that Pegvisomant may be an effective long-term treatment for acromegaly (263,268).  Several European countries have registries providing regular outcome data related to Pegvisomant treatment in acromegaly.  However, cost and approval restrictions mean that Pegvisomant is not yet universally available. Combination treated with SSA and Pegvisomant is likely the most effective medical strategy. Table 7 contains a summary of reported studies assessing effectiveness and safety of this treatment approach.

 

 

 

 

PEG pegvisomant, SRL long-acting somatostatin receptor ligand, QoL Quality of life, OL open-label, LAN Lanreotide, OCT Octreotide LAR, OGTT 75 g oral glucose tolerance test, NA not available

aInclusion criteria: responders to daily PEG monotherapy (presumed previously uncontrolled on SRL therapy), or partial responders to the highest marketed doses of either PEG at 3 months or SRL at 6 months

bPost hoc analysis: eight patients whose mean IGF-1 levels were similar while on pegvisomant monotherapy and during the co-administration period were able to reduce their weekly pegvisomant dose by 50 %

cDefined as > 2 × ULN in this study

dDifferent study criteria for IGF-1 normalization: defined by either end-of–study IGF-1, or lowest IGF-1 achieved

ePrevious pituitary surgery: 1/4; Primary medical therapy: 3/4; none had radiotherapy

fPrevious pituitary surgery: 2/14; Primary medical therapy: 11/14; one patient had radiotherapy

g12/21 patients who did not achieve normal IGF-1 received PEG < 20 mg/day

h2/3 patients with elevations >10 × ULN received OCT 60 mg/28 days

iNone had radiotherapy

 

Table 8. Medical Agents for Acromegaly Including Drugs Under Trial (Most Relevant Targets in Bold). (269-272)

Agent

Route of administration

Molecule

Target

Dose

Side effects

Cabergoline

Oral

Dopamine agonist

DR2

1-4mg /day

Nausea, headache, dizziness, postural hypotension, and nasal stuffiness. Rare concerns of mood disorders and valvular fibrosis

Octreotide LAR

IM

Somatostatin analog

SSTR2 - SSTR5

10-40mg 4 weekly

Gastrointestinal side effects, GB sludge, reduced GB contractility, cholelithiasis, hypothyroidism. Variable effect on glucose. Rare sinus bradycardia, alopecia

Lanreotide ATG

Deep SC

Somatostatin analog

SSTR2 - SSTR5

60-120mg 4 weekly

Pasireotide LAR

IM

Somatostatin analog

SSTR1, SSTR2, SSTR3, SSTR5

40-60mg 4 weekly

Same as above. Also, Hyperglycemia

Pegvisomant

SC

GH receptor antagonist

GH receptor

10-40mg / day

Injection site reactions, abnormal liver enzymes, increase tumor size?

Tamoxifen

Oral

Selective estrogen receptor modulator

 

20-40mg / day

Bone marrow suppression, gynecologic malignancies, hepatotoxicity, ocular effects, thromboembolic events (271)

Octreolin®

Oral

Somatostatin analog

SSTR2 - SSTR5

40-80mg/day

Nausea, bloating, diarrhea, GB stones, dysglycemia

THERAPIES UNDER TRIAL

Glide Octreotide Acetate (GP02)

Needle-free version of regular octreotide acetate

Somatostatin analog

SSTR2 - SSTR5

Immediate release drug, details unclear

Trial data not available

IF-2984®

SC

Somatostatin analog

SSTR1, SSTR2, SSTR3, SSTR5

Immediate release drug, details unclear

Trial data not available

CAM2029

SC

Somatostatin analog

 

20mg monthly depot

Similar to Other SSA

DG3173 (Somatoprim, now called as Veldoreotide)

SC

Somatostatin analog

SSTR2, SSTR4, SSTR5

100-1800µG TDS

Injection site reactions, GI side effects

ATL1103

SC

Antisense molecule

GH receptor (mRNA)

200mg twice weekly

Injection site reaction

Q-chip Octreotide

SC

Somatostatin analog

SSTR2 - SSTR5

10-30mg weekly

Diarrhea, DM

Botulinum neurotoxin SXN101959

Engineered neurotoxin

GHRH receptor

1mg/kg

 

Intravail Octreotide ProTek ®

Oral/nasal

Somatostatin analog

SSTR2 - SSTR5

 

 

VP-003 hydrogel formulation

SC implant

Somatostatin analog

 

84mg 6 monthly

Similar to SSA

 

PERSONALISED MANAGEMENT OF THE PATIENT WITH ACROMEGALY

 

Acromegaly is a rare condition and is best managed by expert teams with a personalized approach. Patient factors, the presence of co-morbidity, quality of life, and resource availability each influence the approach to management.  The Endocrine Society guidance is commonly considered and increasingly biomarkers of disease status and activity are used to guide decision making.

 

Surgery with an intention of cure, or debulking remains the first line of management in most cases. Current evidence suggests that in cases of growth hormone secreting microadenoma, surgery alone will result in achievement of ‘safe’ growth hormone levels in approximately 70-90% of patients. As per the Knosp criteria tumors that cross the lateral tangent of the intracavernous and supracavernous internal carotid arteries are classified as grades 3A, 3B or 4 and are considered invasive (273,274). Surgical remission rate falls when an invasive macroadenoma (<50%) or a giant adenoma (<20%) is present, in contrast to non-invasive tumors (76%). Those with the highest pre-operative growth hormone concentrations, large tumors, or ones with invasion of cavernous sinus are least likely to be ‘cured’ by surgery alone (275). Older age and lower GH levels are associated with likelihood of cure (276).  There may circumstances where consideration could be given for the use of cabergoline for IGF-1 values below twice the upper limit of normal. Use of SSA could be considered pre-operatively in patients with an intention to reduce disease burden or tumor volume prior to surgery to facilitate intervention.

 

Several biomarkers have been suggested to help guide response to first generation SSA. In a pre-operative patient T2 hypointense lesion is more likely to respond to SSA. Most of the other biomarkers are guided by histology outcome such as presence of densely granulated tumor, anti-Cam5.2 staining pattern, SSTR2a expression, and Ki-67 are well-established IHC biomarkers for response to first-generation SSA therapy (277). SSTR2a expression is also as useful marker for Pasireotide response (278). Other markers suggested to predict poor SSA response include low AIP expression, low zinc finger protein ZAC1 (a zinc finger protein expression), poor response during acute octreotide test, the presence of a gsp mutation, low expression of E-cadherin, the expression of sst5 and its truncated isoform (sst5TMD4), higher expression of miR-34a, the expression of β-arrestin, and Raf kinase expression, but these are not well validated (274). While presence of AIP expression is useful to guide use of first generation SSA, it does not seem to correlate with effectiveness of use of Pasireotide (279). There are no IHC markers to guide use of Pegvisomant, but patients with lower IGF-1 respond better (280).

 

Figure 9. Algorithm for management of acromegaly: Colao 2019.

 

TREATMENT STRATEGY IN ACROMEGALY

 

Figure 9 summarizes the initial and subsequent strategies and options used at each stage of patient management.  Practical issues including medication and treatment availability, patient factors, and surgical expertise will each have an influence on treatment.  Following confirmation of the diagnosis of acromegaly surgical treatment should be considered for all patients with a confirmed somatotroph adenoma (80). Current evidence suggests that in cases of growth hormone secreting microadenoma, surgery alone will result in achievement of ‘safe’ growth hormone levels in approximately 70-90% of patients. This figure falls when a macroadenoma (<50%) or a giant adenoma (<20%) is present. Those with the highest pre-operative growth hormone concentrations are least likely to be ‘cured’ by surgery alone. In those post-operative patients with continuing growth hormone excess, further treatment is indicated, and this can be medical or radiotherapy treatment.  A second surgical procedure will result in ‘safe’ growth hormone levels in only 20% of patients. Recognizing that radiotherapy does not result in an instant lowering of growth hormone levels, medical treatment is commonly required, especially in the short-term. On average, two years following external beam irradiation growth hormone levels have decreased by approximately 50% with a further fall resulting in 75% reduction at 5 years. Newer stereotactic radiotherapy techniques, when used appropriately, may affect a more rapid reduction in growth hormone levels. However, since the tumor in such cases is usually a macroadenoma, we would only use radiosurgery as “salvage therapy” in the face of poor control of tumor secretion or regrowth following conventional radiotherapy. Available adjunctive medical options include the use of dopamine agonists, somatostatin analogs (first and second generation), and Pegvisomant. Bromocriptine will normalize growth hormone levels in only 10% of patients, although this may rise to 30% with cabergoline. Octreotide and lanreotide, particularly in their depot formulations which last 4-6 weeks, will normalize mean growth hormone levels in 70-80% of patients, and are therefore highly effective, albeit expensive. Pasireotide results in more potent GH lowering, and in many countries is becoming a key part of the treatment algorithm.  The growth hormone receptor antagonist, Pegvisomant, is now well established and may be used in patients resistant to these agents. Periodic assessment with IGF-1 measurement and growth hormone profile testing should be performed at regular intervals to facilitate titration of doses and determine response to radiotherapy. Following irradiation it is reasonable to assess growth hormone status after appropriate discontinuation of medical therapies at 6-monthly intervals for 2 years and thereafter yearly. In all patients with acromegaly efforts should be made to optimize lung and cardiac function and particular attention be made to the management of cardiovascular risk factors including smoking, dyslipidemia, and abnormalities of carbohydrate metabolism.  ‘Extra-hepatic acromegaly’ describes the concept that elevated GH concentration results in tissue specific pathological effects despite normalization of serum levels of IGF-1 (295). It has been postulated that combined use of SSA (to lower GH) and pegvisomant (to control IGF-1) may be the most appropriate strategy for patients who fall into this category.

 

Treatment of Refractory Disease

 

Acromegaly with invasive non-responsive adenoma, with either persistent GH excess or invasive adenoma is a rare and difficult management problem often needing multi-modal therapy. Table 7 summarizes data from studies reporting use of combination medical therapy in cases resistant to first line SSA therapy.  Few studies have reported using higher dose Pegvisomant or Combination of Pegvisomant and Pasireotide in these refractory cases with good effect (281,282). Use of radiotherapy has been utilized for control of the tumor volume in aggressive disease, while others have considered using the alkylating agent temozolamide or even cytotoxic therapy (222,283).

 

Management of Acromegaly in Pregnancy

 

Pregnancy in a healthy non-pregnant female is associated with gradual decline in pituitary derived GH levels, as the placental GH levels rise throughout the pregnancy. IGF-1 levels initially tend to decrease due to enhanced estrogen effect on liver, but eventually the levels rise as an action of placental GH.  These physiological changes seem to explain the relatively less aggressive or rather benign course of acromegaly in pregnancy (284).

 

Infertility is more common in active disease, and patients often patients require treatment to achieve pregnancy.  Evaluation of baseline tumor volume in a planned pregnancy is useful to safely plan monitoring during pregnancy. GH and IGF-1 levels are unreliable due to assay interference and are not routinely used to guide decision making (212). As a consequence of physiological changes, prevalence of gestational diabetes and hypertension is higher in women with acromegaly, but this seems to correlate with the pre-pregnancy control and not the degree of rise of IGF-1 (285). For patients with macroadenoma, serial visual field testing is required during pregnancy. Patients with intractable headaches, cranial nerve deficits or visual manifestations are likely to require intervention. If MRI is required it is best undertaken as unenhanced study (212). As GH does not cross the placenta, no direct effect on the disease on fetus have been reported. Studies have shown that the tumor size does not usually increase during pregnancy (286). The current recommendations suggest all medical therapy should be ceased at diagnosis of pregnancy. If pregnancy is pre-planned it is recommended that long acting SSA are discontinued about two months prior to pregnancy and patient switched to short acting octreotide injections (151,287). Safety data for the use of SSA and Pegvisomant is not substantial, but the available evidence has not shown any significant impact on maternal or fetal outcomes. There are reports of SSA use being associated with small for gestation babies, without malformations and similar concerns with use of dopamine agonists (285,288). While there are concerns for premature delivery the data for use of Pegvisomant in pregnancy is more encouraging (289).

 

NOVEL AGENTS IN THE TREATMENT OF ACROMEGALY

 

A number of novel agents are in advanced stages of development for the medical treatment of acromegaly. These include agents that continue to work by the somatostatin mechanism as well as new mechanisms of action. Developments in the understanding of the molecular pathogenesis of growth hormone excess and pituitary tumor development have led to the identification of novel targets for drug development. New treatments need to be safe and well tolerated, as well as effective and importantly cost effective.

 

An anti-sense oligonucleotide has been developed directed against the growth hormone receptor. Early clinical trial data suggests that this strategy may prove effective in reducing growth hormone signaling and IGF-1 generation in patients with acromegaly (290).   The drug was well tolerated in an early clinical trial with injection site reactions the most common adverse event reported.

 

Novel compounds with combined affinity for SSTR2, SSTR5 and the dopamine D2 receptor are also being developed and in vitro show enhanced inhibition of growth hormone release (291). The ongoing development of these chimeric analogs may increase the efficiency of currently available analogs (292).

 

Somatoprim or Veldoreotide is a novel somatostatin analogue. This agent has affinity for the SST2, 4 and 5 receptors. A phase II study to investigate the efficacy of this agent in acromegaly is underway.  STAT3 signaling is an important mechanism in the regulation of growth on dependent gene expression. GH-secreting adenomas overexpress STAT3. Recently a STAT3 inhibitor has been shown to suppress growth action. Thus, there is early evidence that this novel strategy may have a role in the treatment of acromegaly in the future (293). In addition, a new formulation of subcutaneous octreotide depot has been trialed in phase II studies, demonstrating superior efficacy to intramuscular octreotide (294).

 

In summary, continuing advances in the understanding of the mechanisms responsible for pituitary tumor development and the regulation of GH secretion, are aiding the further development of existing therapeutic agents and enabling the creation of new promising treatment for patients with acromegaly.

 

REFERENCES

 

  1. Bengtsson BA, Edén S, Ernest I, Odén A, Sjögren B. Epidemiology and long-term survival in acromegaly. A study of 166 cases diagnosed between 1955 and 1984. Acta Med Scand. 1988;223(4):327-335.
  2. Gatto F, Trifirò G, Lapi F, Cocchiara F, Campana C, Dell'Aquila C, Ferrajolo C, Arvigo M, Cricelli C, Giusti M, Ferone D. Epidemiology of acromegaly in Italy: analysis from a large longitudinal primary care database. Endocrine. 2018;61(3):533-541.
  3. Hoskuldsdottir GT, Fjalldal SB, Sigurjonsdottir HA. The incidence and prevalence of acromegaly, a nationwide study from 1955 through 2013. Pituitary. 2015;18(6):803-807.
  4. Marie P. Sur deuxcas d'acromegalie hypertrophie singuliere, non conge-nitale des extremites supe rieures, inferieures, et cephaligue. Rev de Med. 1886;6:297-333.
  5. Krug S, Boch M, Rexin P, Pfestroff A, Gress T, Michl P, Rinke A. Acromegaly in a patient with a pulmonary neuroendocrine tumor: case report and review of current literature. BMC Res Notes. 2016;9:326.
  6. Glikson M, Gil-Ad I, Galun E, Dresner R, Zilberman S, Halperin Y, Okon E, Laron Z, Rubinow A. Acromegaly due to ectopic growth hormone-releasing hormone secretion by a bronchial carcinoid tumour. Dynamic hormonal responses to various stimuli. Acta Endocrinol (Copenh). 1991;125(4):366-371.
  7. Asa SL, Scheithauer BW, Bilbao JM, Horvath E, Ryan N, Kovacs K, Randall RV, Laws ER, Singer W, Linfoot JA. A case for hypothalamic acromegaly: a clinicopathological study of six patients with hypothalamic gangliocytomas producing growth hormone-releasing factor. J Clin Endocrinol Metab. 1984;58(5):796-803.
  8. Nelson AE, Ho KK. Abuse of growth hormone by athletes. Nat Clin Pract Endocrinol Metab. 2007;3(3):198-199.
  9. Rubinek T, Modan-Moses D. Chapter Four - Klotho and the Growth Hormone/Insulin-Like Growth Factor 1 Axis: Novel Insights into Complex Interactions. In: Litwack G, ed. Vitamins & Hormones. Vol 101: Academic Press; 2016:85-118.
  10. Luque RM, Peinado JR, Gracia-Navarro F, Broglio F, Ghigo E, Kineman RD, Malagón MM, Castaño JP. Cortistatin mimics somatostatin by inducing a dual, dose-dependent stimulatory and inhibitory effect on growth hormone secretion in somatotropes. J Mol Endocrinol. 2006;36(3):547-556.
  11. Cuttler L. The regulation of growth hormone secretion. Endocrinol Metab Clin North Am. 1996;25(3):541-571.
  12. Müller EE, Locatelli V, Cocchi D. Neuroendocrine control of growth hormone secretion. Physiol Rev.1999;79(2):511-607.
  13. Carter-Su C, Schwartz J, Argetsinger LS. Growth hormone signaling pathways. Growth Horm IGF Res.2016;28:11-15.
  14. Dehkhoda F, Lee CMM, Medina J, Brooks AJ. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Front Endocrinol (Lausanne). 2018;9:35.
  15. Pilecka I, Patrignani C, Pescini R, Curchod M-L, Perrin D, Xue Y, Yasenchak J, Clark A, Magnone MC, Zaratin P, Valenzuela D, Rommel C, van Huijsduijnen RH. Protein-tyrosine Phosphatase H1 Controls Growth Hormone Receptor Signaling and Systemic Growth. Journal of Biological Chemistry. 2007;282(48):35405-35415.
  16. Boguszewski CL, Barbosa EJL, Svensson PA, Johannsson G, Glad CAM. MECHANISMS IN ENDOCRINOLOGY: Clinical and pharmacogenetic aspects of the growth hormone receptor polymorphism. Eur J Endocrinol. 2017;177(6):R309-R321.
  17. Le Roith D, Bondy C, Yakar S, Liu JL, Butler A. The somatomedin hypothesis: 2001. Endocr Rev.2001;22(1):53-74.
  18. Le Roith D, Scavo L, Butler A. What is the role of circulating IGF-I? Trends Endocrinol Metab. 2001;12(2):48-52.
  19. Frohman LA, Downs TR, Chomczynski P, Frohman MA. Growth hormone-releasing hormone: structure, gene expression and molecular heterogeneity. Acta Paediatr Scand Suppl. 1990;367:81-86.
  20. Steyn FJ, Tolle V, Chen C, Epelbaum J. Neuroendocrine Regulation of Growth Hormone Secretion. Compr Physiol. 2016;6(2):687-735.
  21. Huang L, Tan HY, Fogarty MJ, Andrews ZB, Veldhuis JD, Herzog H, Steyn FJ, Chen C. Actions of NPY, and its Y1 and Y2 receptors on pulsatile growth hormone secretion during the fed and fasted state. J Neurosci.2014;34(49):16309-16319.
  22. Romero CJ, Pine-Twaddell E, Sima DI, Miller RS, He L, Wondisford F, Radovick S. Insulin-like growth factor 1 mediates negative feedback to somatotroph GH expression via POU1F1/CREB binding protein interactions. Molecular and cellular biology. 2012;32(21):4258-4269.
  23. Butler AA, Le Roith D. Control of growth by the somatropic axis: growth hormone and the insulin-like growth factors have related and independent roles. Annu Rev Physiol. 2001;63:141-164.
  24. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656-660.
  25. Khatib N, Gaidhane S, Gaidhane AM, Khatib M, Simkhada P, Gode D, Zahiruddin QS. Ghrelin: ghrelin as a regulatory Peptide in growth hormone secretion. J Clin Diagn Res. 2014;8(8):MC13-17.
  26. Kojima M, Kangawa K. Ghrelin: more than endogenous growth hormone secretagogue. Annals of the New York Academy of Sciences. 2010;1200(1):140-148.
  27. Chan JL, Williams CJ, Raciti P, Blakeman J, Kelesidis T, Kelesidis I, Johnson ML, Thorner MO, Mantzoros CS. Leptin does not mediate short-term fasting-induced changes in growth hormone pulsatility but increases IGF-I in leptin deficiency states. Journal of Clinical Endocrinology and Metabolism. 2008;93:2819-2827.
  28. Steyn FJ. Nutrient Sensing Overrides Somatostatin and Growth Hormone-Releasing Hormone to Control Pulsatile Growth Hormone Release. J Neuroendocrinol. 2015;27(7):577-587.
  29. Giovanni T. How treatments with endocrine and metabolic drugs influence pituitary cell function. Endocrine Connections. 2020;9(2):R14-R27.
  30. Kato Y, Murakami Y, Sohmiya M, Nishiki M. Regulation of human growth hormone secretion and its disorders. Intern Med. 2002;41(1):7-13.
  31. Leung AM, Brent GA. The Influence of Thyroid Hormone on Growth Hormone Secretion and Action. In: Cohen LE, ed. Growth Hormone Deficiency: Physiology and Clinical Management. Cham: Springer International Publishing; 2016:29-46.
  32. Root AW, Shulman D, Root J, Diamond F. The interrelationships of thyroid and growth hormones: effect of growth hormone releasing hormone in hypo- and hyperthyroid male rats. Acta Endocrinol Suppl (Copenh).1986;279:367-375.
  33. Behan LA, Monson JP, Agha A. The interaction between growth hormone and the thyroid axis in hypopituitary patients. Clin Endocrinol (Oxf). 2011;74(3):281-288.
  34. Yamauchi I, Sakane Y, Yamashita T, Hirota K, Ueda Y, Kanai Y, Yamashita Y, Kondo E, Fujii T, Taura D, Sone M, Yasoda A, Inagaki N. Effects of growth hormone on thyroid function are mediated by type 2 iodothyronine deiodinase in humans. Endocrine. 2018;59(2):353-363.
  35. Mauras N, Blizzard RM, Link K, Johnson ML, Rogol AD, Veldhuis JD. Augmentation of growth hormone secretion during puberty: evidence for a pulse amplitude-modulated phenomenon. J Clin Endocrinol Metab.1987;64(3):596-601.
  36. Frantz AG, Rabkin MT. Effects of estrogen and sex difference on secretion of human growth hormone. Journal of Clinical Endocrinology and Metabolism. 1965;25:1470-1480.
  37. Lenders NF, McCormack AI, Ho KKY. MANAGEMENT OF ENDOCRINE DISEASE: Does gender matter in the management of acromegaly? Eur J Endocrinol. 2020;182(5):R67-R82.
  38. Hage M, Kamenický P, Chanson P. Growth Hormone Response to Oral Glucose Load: From Normal to Pathological Conditions. Neuroendocrinology. 2019;108(3):244-255.
  39. Daughaday WH, Rotwein P. Insulin-like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr Rev. 1989;10(1):68-91.
  40. Martin JL, Baxter RC. Signalling pathways of insulin-like growth factors (IGFs) and IGF binding protein-3. Growth Factors. 2011;29(6):235-244.
  41. Rotwein P. Structure, evolution, expression and regulation of insulin-like growth factors I and II. Growth Factors.1991;5(1):3-18.
  42. Baxter RC. Insulin-like growth factor binding proteins in the human circulation: a review. Horm Res. 1994;42(4-5):140-144.
  43. Ferry RJ, Jr., Katz LE, Grimberg A, Cohen P, Weinzimer SA. Cellular actions of insulin-like growth factor binding proteins. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme.1999;31(2-3):192-202.
  44. Akirov A, Asa SL, Amer L, Shimon I, Ezzat S. The Clinicopathological Spectrum of Acromegaly. J Clin Med.2019;8(11).
  45. Hayward BE, Barlier A, Korbonits M, Grossman AB, Jacquet P, Enjalbert A, Bonthron DT. Imprinting of the G(s)alpha gene GNAS1 in the pathogenesis of acromegaly. J Clin Invest. 2001;107(6):R31-36.
  46. Asa SL, Silverberg SG. Tumors of the Pituitary Gland. ARP Press.
  47. Syro LV, Rotondo F, Serna CA, Ortiz LD, Kovacs K. Pathology of GH-producing pituitary adenomas and GH cell hyperplasia of the pituitary. Pituitary. 2017;20(1):84-92.
  48. Mete O, Kefeli M, Çalışkan S, Asa SL. GATA3 immunoreactivity expands the transcription factor profile of pituitary neuroendocrine tumors. Modern Pathology. 2019;32(4):484-489.
  49. Gomez-Hernandez K, Ezzat S, Asa SL, Mete Ö. Clinical Implications of Accurate Subtyping of Pituitary Adenomas: Perspectives from the Treating Physician. Turk Patoloji Derg. 2015;31 Suppl 1:4-17.
  50. Melmed S. Extrapituitary acromegaly. Endocrinol Metab Clin North Am. 1991;20(3):507-518.
  51. Pérez-Ibave DC, Rodríguez-Sánchez IP, Garza-Rodríguez MeL, Barrera-Saldaña HA. Extrapituitary growth hormone synthesis in humans. Growth Horm IGF Res. 2014;24(2-3):47-53.
  52. Colao A, Grasso LFS, Giustina A, Melmed S, Chanson P, Pereira AM, Pivonello R. Acromegaly. Nat Rev Dis Primers. 2019;5(1):20.
  53. Kyriakakis N, Trouillas J, Dang MN, Lynch J, Belchetz P, Korbonits M, Murray RD. Diagnostic challenges and management of a patient with acromegaly due to ectopic growth hormone-releasing hormone secretion from a bronchial carcinoid tumour. Endocrinol Diabetes Metab Case Rep. 2017;2017.
  54. Weiss DE, Vogel H, Lopes MB, Chang SD, Katznelson L. Ectopic acromegaly due to a pancreatic neuroendocrine tumor producing growth hormone-releasing hormone. Endocr Pract. 2011;17(1):79-84.
  55. Dal J, Feldt-Rasmussen U, Andersen M, Kristensen L, Laurberg P, Pedersen L, Dekkers OM, Sørensen HT, Jørgensen JO. Acromegaly incidence, prevalence, complications and long-term prognosis: a nationwide cohort study. Eur J Endocrinol. 2016;175(3):181-190.
  56. Ben-Shlomo A, Melmed S. Acromegaly. Endocrinol Metab Clin North Am. 2008;37(1):101-122, viii.
  57. Ioachimescu AG, Handa T, Goswami N, Pappy AL, Veledar E, Oyesiku NM. Gender differences and temporal trends over two decades in acromegaly: a single center study in 112 patients. Endocrine. 2020;67(2):423-432.
  58. Nyquist P, Laws ER, Elliott E. Novel features of tumors that secrete both growth hormone and prolactin in acromegaly. Neurosurgery. 1994;35(2):179-183; discussion 183-174.
  59. Mestron A, Webb SM, Astorga R, Benito P, Catala M, Gaztambide S, Gomez JM, Halperin I, Lucas-Morante T, Moreno B, Obiols G, de Pablos P, Paramo C, Pico A, Torres E, Varela C, Vazquez JA, Zamora J, Albareda M, Gilabert M. Epidemiology, clinical characteristics, outcome, morbidity and mortality in acromegaly based on the Spanish Acromegaly Registry (Registro Espanol de Acromegalia, REA). Eur J Endocrinol. 2004;151(4):439-446.
  60. Cuevas-Ramos D, Carmichael JD, Cooper O, Bonert VS, Gertych A, Mamelak AN, Melmed S. A structural and functional acromegaly classification. J Clin Endocrinol Metab. 2015;100(1):122-131.
  61. Anagnostis P, Efstathiadou ZA, Polyzos SA, Adamidou F, Slavakis A, Sapranidis M, Litsas ID, Katergari S, Selalmatzidou D, Kita M. Acromegaly: presentation, morbidity and treatment outcomes at a single centre. Int J Clin Pract. 2011;65(8):896-902.
  62. Wade AN, Baccon J, Grady MS, Judy KD, O'Rourke DM, Snyder PJ. Clinically silent somatotroph adenomas are common. Eur J Endocrinol. 2011;165(1):39-44.
  63. Langlois F, Lim DST, Varlamov E, Yedinak CG, Cetas JS, McCartney S, Dogan A, Fleseriu M. Clinical profile of silent growth hormone pituitary adenomas; higher recurrence rate compared to silent gonadotroph pituitary tumors, a large single center experience. Endocrine. 2017;58(3):528-534.
  64. Butz LB, Sullivan SE, Chandler WF, Barkan AL. "Micromegaly": an update on the prevalence of acromegaly with apparently normal GH secretion in the modern era. Pituitary. 2016;19(6):547-551.
  65. Espinosa de Los Monteros AL, Sosa-Eroza E, Gonzalez B, Mendoza V, Mercado M. Prevalence, Clinical and Biochemical Spectrum, and Treatment Outcome of Acromegaly With Normal Basal GH at Diagnosis. J Clin Endocrinol Metab. 2018;103(10):3919-3924.
  66. Greenman Y, Tordjman K, Kisch E, Razon N, Ouaknine G, Stern N. Relative sparing of anterior pituitary function in patients with growth hormone-secreting macroadenomas: comparison with nonfunctioning macroadenomas. J Clin Endocrinol Metab. 1995;80(5):1577-1583.
  67. Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest. 2009;119(11):3189-3202.
  68. Katznelson L, Kleinberg D, Vance ML, Stavrou S, Pulaski KJ, Schoenfeld DA, Hayden DL, Wright ME, Woodburn CJ, Klibanski A, Stravou S. Hypogonadism in patients with acromegaly: data from the multi-centre acromegaly registry pilot study. Clin Endocrinol (Oxf). 2001;54(2):183-188.
  69. Kaltsas GA, Androulakis II, Tziveriotis K, Papadogias D, Tsikini A, Makras P, Dimitriou K, Stathopoulou A, Piaditis G. Polycystic ovaries and the polycystic ovary syndrome phenotype in women with active acromegaly. Clin Endocrinol (Oxf). 2007;67(6):917-922.
  70. Lotti F, Rochira V, Pivonello R, Santi D, Galdiero M, Maseroli E, Balestrieri A, Faustini-Fustini M, Peri A, Sforza A, Colao A, Maggi M, Corona G. Erectile Dysfunction is Common among Men with Acromegaly and is Associated with Morbidities Related to the Disease. J Sex Med. 2015;12(5):1184-1193.
  71. Castro Cabezas M, Zelissen PM, Jansen GH, Van Gils AP, Koppeschaar HP. Acromegaly: report of two patients with an unusual presentation. Neth J Med. 1999;54(4):163-166.
  72. Genka S, Soeda H, Takahashi M, Katakami H, Sanno N, Osamura Y, Fuchinoue T, Teramoto A. Acromegaly, diabetes insipidus, and visual loss caused by metastatic growth hormone-releasing hormone-producing malignant pancreatic endocrine tumor in the pituitary gland. Case report. J Neurosurg. 1995;83(4):719-723.
  73. Ezzat S, Forster MJ, Berchtold P, Redelmeier DA, Boerlin V, Harris AG. Acromegaly. Clinical and biochemical features in 500 patients. Medicine (Baltimore). 1994;73(5):233-240.
  74. Ben-Shlomo A, Melmed S. Skin manifestations in acromegaly. Clin Dermatol. 2006;24(4):256-259.
  75. Degirmentepe EN, Gungor S, Kocaturk E, Kiziltac U, Adas M, Ozekinci S, Khachemoune A. Dermatologic manifestations of acromegaly: A case in point and a focused review. Dermatol Online J. 2017;23(8).
  76. Katznelson L. Alterations in body composition in acromegaly. Pituitary. 2009;12(2):136-142.
  77. Bengtsson BA, Brummer RJ, Edén S, Bosaeus I, Lindstedt G. Body composition in acromegaly: the effect of treatment. Clin Endocrinol (Oxf). 1989;31(4):481-490.
  78. Bredella MA, Schorr M, Dichtel LE, Gerweck AV, Young BJ, Woodmansee WW, Swearingen B, Miller KK. Body Composition and Ectopic Lipid Changes With Biochemical Control of Acromegaly. J Clin Endocrinol Metab.2017;102(11):4218-4225.
  79. Parolin M, Dassie F, Vettor R, Maffei P. Acromegaly and ultrasound: how, when and why? J Endocrinol Invest.2020;43(3):279-287.
  80. Murrant NJ, Gatland DJ. Respiratory problems in acromegaly. J Laryngol Otol. 1990;104(1):52-55.
  81. Wolters TLC, Roerink SHPP, Drenthen LCA, Wagenmakers MAEM, van den Broek GB, Rutten KIM, Herruer JM, Hermus ARMM, Netea-Maier RT. Voice Characteristics in Patients with Acromegaly during Treatment. J Voice. 2020.
  82. Aydin K, Turkyilmaz D, Ozturk B, Dagdelen S, Ozgen B, Unal F, Erbas T. Voice characteristics of acromegaly. Eur Arch Otorhinolaryngol. 2013;270(4):1391-1396.
  83. Bogazzi F, Nacci A, Campomori A, La Vela R, Rossi G, Lombardi M, Fattori B, Bartalena L, Ursino F, Martino E. Analysis of voice in patients with untreated active acromegaly. J Endocrinol Invest. 2010;33(3):178-185.
  84. Gadelha MR, Kasuki L, Lim DST, Fleseriu M. Systemic Complications of Acromegaly and the Impact of the Current Treatment Landscape: An Update. Endocr Rev. 2019;40(1):268-332.
  85. Anthony JR, Ioachimescu AG. Acromegaly and bone disease. Curr Opin Endocrinol Diabetes Obes.2014;21(6):476-482.
  86. Pekkolay Z, Kılınç F, Gozel N, Önalan E, Tuzcu AK. Increased Serum Sclerostin Levels in Patients With Active Acromegaly. J Clin Endocrinol Metab. 2020;105(3).
  87. Giustina A. Acromegaly and Vertebral Fractures: Facts and Questions. Trends Endocrinol Metab.2020;31(4):274-275.
  88. Plard C, Hochman C, Hadjadj S, Goff BL, Maugars Y, Cariou B, Drui D, Guillot P. Acromegaly is associated with vertebral deformations but not vertebral fractures: results of a cross-sectional monocentric study. Joint Bone Spine. 2020.
  89. Bima C, Chiloiro S, Mormando M, Piacentini S, Bracaccia E, Giampietro A, Tartaglione L, Bianchi A, De Marinis L. Understanding the effect of acromegaly on the human skeleton. Expert Rev Endocrinol Metab.2016;11(3):263-270.
  90. Mazziotti G, Lania AGA, Canalis E. MANAGEMENT OF ENDOCRINE DISEASE: Bone disorders associated with acromegaly: mechanisms and treatment. Eur J Endocrinol. 2019;181(2):R45-R56.
  91. Barkan AL. Acromegalic arthropathy. Pituitary. 2001;4(4):263-264.
  92. Wassenaar MJ, Biermasz NR, van Duinen N, van der Klaauw AA, Pereira AM, Roelfsema F, Smit JW, Kroon HM, Kloppenburg M, Romijn JA. High prevalence of arthropathy, according to the definitions of radiological and clinical osteoarthritis, in patients with long-term cure of acromegaly: a case-control study. Eur J Endocrinol.2009;160(3):357-365.
  93. Godang K, Lekva T, Normann KR, Olarescu NC, Øystese KAB, Kolnes A, Ueland T, Bollerslev J, Heck A. Hip Structure Analyses in Acromegaly: Decrease of Cortical Bone Thickness After Treatment: A Longitudinal Cohort Study. JBMR Plus. 2019;3(12):e10240.
  94. Wassenaar MJ, Biermasz NR, Bijsterbosch J, Pereira AM, Meulenbelt I, Smit JW, Roelfsema F, Kroon HM, Romijn JA, Kloppenburg M. Arthropathy in long-term cured acromegaly is characterised by osteophytes without joint space narrowing: a comparison with generalised osteoarthritis. Ann Rheum Dis. 2011;70(2):320-325.
  95. Chevallier M, Pontier S, Sedkaoui K, Caron P, Didier A. [Characteristics of sleep apnea syndrome in a cohort of patients with acromegaly]. Rev Mal Respir. 2012;29(5):673-679.
  96. Guo X, Gao L, Zhao Y, Wang M, Jiang B, Wang Q, Wang Z, Liu X, Feng M, Wang R, Zhang Z, Xing B. Characteristics of the upper respiratory tract in patients with acromegaly and correlations with obstructive sleep apnoea/hypopnea syndrome. Sleep Med. 2018;48:27-34.
  97. Roemmler J, Gutt B, Fischer R, Vay S, Wiesmeth A, Bidlingmaier M, Schopohl J, Angstwurm M. Elevated incidence of sleep apnoea in acromegaly-correlation to disease activity. Sleep Breath. 2012;16(4):1247-1253.
  98. Herrmann BL, Wessendorf TE, Ajaj W, Kahlke S, Teschler H, Mann K. Effects of octreotide on sleep apnoea and tongue volume (magnetic resonance imaging) in patients with acromegaly. Eur J Endocrinol.2004;151(3):309-315.
  99. Grunstein RR, Ho KY, Berthon-Jones M, Stewart D, Sullivan CE. Central sleep apnea is associated with increased ventilatory response to carbon dioxide and hypersecretion of growth hormone in patients with acromegaly. Am J Respir Crit Care Med. 1994;150(2):496-502.
  100. Davì MV, Giustina A. Sleep apnea in acromegaly: a review on prevalence, pathogenetic aspects and treatment. Expert Rev Endocrinol Metab. 2012;7(1):55-62.
  101. Miller A, Doll H, David J, Wass J. Impact of musculoskeletal disease on quality of life in long-standing acromegaly. Eur J Endocrinol. 2008;158(5):587-593.
  102. Mastaglia FL. Pathological changes in skeletal muscle in acromegaly. Acta Neuropathol. 1973;24(4):273-286.
  103. Füchtbauer L, Olsson DS, Bengtsson B, Norrman LL, Sunnerhagen KS, Johannsson G. Muscle strength in patients with acromegaly at diagnosis and during long-term follow-up. Eur J Endocrinol. 2017;177(2):217-226.
  104. Nagulesparen M, Trickey R, Davies MJ, Jenkins JS. Muscle changes in acromegaly. Br Med J.1976;2(6041):914-915.
  105. Walchan EM, Guimarães FS, Soares MS, Kasuki L, Gadelha MR, Lopes AJ. Parameters of knee isokinetic dynamometry in individuals with acromegaly: association with growth hormone levels and general fatigue. Isokinetics and Exercise Science. 2016;24:331-340.
  106. Ozturk Gokce B, Gogus F, Bolayir B, Tecer D, Gokce O, Eroglu Altinova A, Balos Toruner F, Akturk M. The evaluation of the tendon and muscle changes of lower extremity in patients with acromegaly. Pituitary. 2020.
  107. Freda PU, Shen W, Reyes-Vidal CM, Geer EB, Arias-Mendoza F, Gallagher D, Heymsfield SB. Skeletal muscle mass in acromegaly assessed by magnetic resonance imaging and dual-photon x-ray absorptiometry. J Clin Endocrinol Metab. 2009;94(8):2880-2886.
  108. Consitt LA, Saneda A, Saxena G, List EO, Kopchick JJ. Mice overexpressing growth hormone exhibit increased skeletal muscle myostatin and MuRF1 with attenuation of muscle mass. Skeletal muscle. 2017;7(1):17-17.
  109. Colao A, Auriemma RS, Pivonello R, Galdiero M, Lombardi G. Medical consequences of acromegaly: what are the effects of biochemical control? Rev Endocr Metab Disord. 2008;9(1):21-31.
  110. Jenkins PJ, Sohaib SA, Akker S, Phillips RR, Spillane K, Wass JA, Monson JP, Grossman AB, Besser GM, Reznek RH. The pathology of median neuropathy in acromegaly. Ann Intern Med. 2000;133(3):197-201.
  111. Sasagawa Y, Tachibana O, Doai M, Tonami H, Iizuka H. Median nerve conduction studies and wrist magnetic resonance imaging in acromegalic patients with carpal tunnel syndrome. Pituitary. 2015;18(5):695-700.
  112. Tagliafico A, Resmini E, Nizzo R, Bianchi F, Minuto F, Ferone D, Martinoli C. Ultrasound measurement of median and ulnar nerve cross-sectional area in acromegaly. J Clin Endocrinol Metab. 2008;93(3):905-909.
  113. Ozata M, Ozkardes A, Beyhan Z, Corakci A, Gundogan MA. Central and peripheral neural responses in acromegaly. Endocr Pract. 1997;3(3):118-122.
  114. Alibas H, Gogas Yavuz D, Kahraman Koytak P, Uygur M, Tanridag T, Uluc K. Peripheral nervous system assessment in acromegaly patients under somatostatin analogue therapy. J Endocrinol Invest. 2017;40(1):33-40.
  115. Resmini E, Tagliafico A, Nizzo R, Bianchi F, Minuto F, Derchi L, Martinoli C, Ferone D. Ultrasound of peripheral nerves in acromegaly: changes at 1-year follow-up. Clin Endocrinol (Oxf). 2009;71(2):220-225.
  116. Dural M, Kabakcı G, Cınar N, Erbaş T, Canpolat U, Gürses KM, Tokgözoğlu L, Oto A, Kaya EB, Yorgun H, Sahiner L, Dağdelen S, Aytemir K. Assessment of cardiac autonomic functions by heart rate recovery, heart rate variability and QT dynamicity parameters in patients with acromegaly. Pituitary. 2014;17(2):163-170.
  117. Oz O, Taşlıpınar A, Yücel M, Akgün H, Ulaş UH, Bolu E, Kütükçü Y, Odabaşı Z. Electrophysiological assessment of the autonomic nervous system in male patients with acromegaly. Eur Neurol. 2011;66(1):1-5.
  118. Comunello A, Dassie F, Martini C, De Carlo E, Mioni R, Battocchio M, Paoletta A, Fallo F, Vettor R, Maffei P. Heart rate variability is reduced in acromegaly patients and improved by treatment with somatostatin analogues. Pituitary. 2015;18(4):525-534.
  119. Pilecki W, Bolanowski M, Janocha A, Daroszewski J, Kałuzny M, Sebzda T, Kałka D, Sobieszczańska M. Assessment of brainstem auditory evoked potentials (BAEPs) in patients with acromegaly. Neuro Endocrinol Lett. 2008;29(3):373-378.
  120. Cannavò S, Condurso R, Ragonese M, Ferraù F, Alibrandi A, Aricò I, Romanello G, Squadrito S, Trimarchi F, Silvestri R. Increased prevalence of restless legs syndrome in patients with acromegaly and effects on quality of life assessed by Acro-QoL. Pituitary. 2011;14(4):328-334.
  121. Arosio M, Reimondo G, Malchiodi E, Berchialla P, Borraccino A, De Marinis L, Pivonello R, Grottoli S, Losa M, Cannavò S, Minuto F, Montini M, Bondanelli M, De Menis E, Martini C, Angeletti G, Velardo A, Peri A, Faustini-Fustini M, Tita P, Pigliaru F, Borretta G, Scaroni C, Bazzoni N, Bianchi A, Appetecchia M, Cavagnini F, Lombardi G, Ghigo E, Beck-Peccoz P, Colao A, Terzolo M, Acromegaly ISGo. Predictors of morbidity and mortality in acromegaly: an Italian survey. Eur J Endocrinol. 2012;167(2):189-198.
  122. Vitale G, Pivonello R, Lombardi G, Colao A. Cardiovascular complications in acromegaly. Minerva Endocrinol.2004;29(3):77-88.
  123. Goldberg MD, Vadera N, Yandrapalli S, Frishman WH. Acromegalic Cardiomyopathy: An Overview of Risk Factors, Clinical Manifestations, and Therapeutic Options. Cardiol Rev. 2018;26(6):307-311.
  124. Petrossians P, Daly AF, Natchev E, Maione L, Blijdorp K, Sahnoun-Fathallah M, Auriemma R, Diallo AM, Hulting AL, Ferone D, Hana V, Filipponi S, Sievers C, Nogueira C, Fajardo-Montañana C, Carvalho D, Stalla GK, Jaffrain-Réa ML, Delemer B, Colao A, Brue T, Neggers SJCM, Zacharieva S, Chanson P, Beckers A. Acromegaly at diagnosis in 3173 patients from the Liège Acromegaly Survey (LAS) Database. Endocr Relat Cancer. 2017;24(10):505-518.
  125. Vitale G, Pivonello R, Auriemma RS, Guerra E, Milone F, Savastano S, Lombardi G, Colao A. Hypertension in acromegaly and in the normal population: prevalence and determinants. Clin Endocrinol (Oxf). 2005;63(4):470-476.
  126. Costenaro F, Martin A, Horn RF, Czepielewski MA, Rodrigues TC. Role of ambulatory blood pressure monitoring in patients with acromegaly. J Hypertens. 2016;34(7):1357-1363.
  127. Kamenicky P, Viengchareun S, Blanchard A, Meduri G, Zizzari P, Imbert-Teboul M, Doucet A, Chanson P, Lombès M. Epithelial sodium channel is a key mediator of growth hormone-induced sodium retention in acromegaly. Endocrinology. 2008;149(7):3294-3305.
  128. Puglisi S, Terzolo M. Hypertension and Acromegaly. Endocrinol Metab Clin North Am. 2019;48(4):779-793.
  129. Sharma AN, Tan M, Amsterdam EA, Singh GD. Acromegalic cardiomyopathy: Epidemiology, diagnosis, and management. Clin Cardiol. 2018;41(3):419-425.
  130. Ramos-Leví AM, Marazuela M. Cardiovascular comorbidities in acromegaly: an update on their diagnosis and management. Endocrine. 2017;55(2):346-359.
  131. Colao A, Pivonello R, Grasso LF, Auriemma RS, Galdiero M, Savastano S, Lombardi G. Determinants of cardiac disease in newly diagnosed patients with acromegaly: results of a 10 year survey study. Eur J Endocrinol. 2011;165(5):713-721.
  132. Bogazzi F, Lombardi M, Strata E, Aquaro G, Di Bello V, Cosci C, Sardella C, Talini E, Martino E. High prevalence of cardiac hypertophy without detectable signs of fibrosis in patients with untreated active acromegaly: an in vivo study using magnetic resonance imaging. Clin Endocrinol (Oxf). 2008;68(3):361-368.
  133. Guo X, Cao J, Liu P, Cao Y, Li X, Gao L, Wang Z, Fang L, Jin Z, Wang Y, Xing B. Cardiac Abnormalities in Acromegaly Patients: A Cardiac Magnetic Resonance Study. Int J Endocrinol. 2020;2020:2018464.
  134. Schöfl C, Petroff D, Tönjes A, Grussendorf M, Droste M, Stalla G, Jaursch-Hancke C, Störmann S, Schopohl J. Incidence of myocardial infarction and stroke in acromegaly patients: results from the German Acromegaly Registry. Pituitary. 2017;20(6):635-642.
  135. Kırış A, Erem C, Turan OE, Civan N, Kırış G, Nuhoğlu I, Ilter A, Ersöz HO, Kutlu M. Left ventricular synchronicity is impaired in patients with active acromegaly. Endocrine. 2013;44(1):200-206.
  136. Pereira AM, van Thiel SW, Lindner JR, Roelfsema F, van der Wall EE, Morreau H, Smit JW, Romijn JA, Bax JJ. Increased prevalence of regurgitant valvular heart disease in acromegaly. J Clin Endocrinol Metab.2004;89(1):71-75.
  137. Mercado M, Ramírez-Rentería C. Metabolic Complications of Acromegaly. Front Horm Res. 2018;49:20-28.
  138. Feingold KR, Brinton EA, Grunfeld C. The Effect of Endocrine Disorders on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA): MDText.com, Inc.

Copyright © 2000-2020, MDText.com, Inc.; 2000.

  1. Frara S, Maffezzoni F, Mazziotti G, Giustina A. Current and Emerging Aspects of Diabetes Mellitus in Acromegaly. Trends Endocrinol Metab. 2016;27(7):470-483.
  2. Olarescu NC, Bollerslev J. The Impact of Adipose Tissue on Insulin Resistance in Acromegaly. Trends Endocrinol Metab. 2016;27(4):226-237.
  3. Reid TJ, Jin Z, Shen W, Reyes-Vidal CM, Fernandez JC, Bruce JN, Kostadinov J, Post KD, Freda PU. IGF-1 levels across the spectrum of normal to elevated in acromegaly: relationship to insulin sensitivity, markers of cardiovascular risk and body composition. Pituitary. 2015;18(6):808-819.
  4. Janssen JAMJ. Mechanisms of putative IGF-I receptor resistance in active acromegaly. Growth Horm IGF Res.2020;52:101319.
  5. Ciresi A, Amato MC, Pizzolanti G, Giordano C. Serum visfatin levels in acromegaly: Correlation with disease activity and metabolic alterations. Growth Horm IGF Res. 2015;25(5):240-246.
  6. Calan M, Demirpence M. Increased circulating levels of irisin are associated with cardiovascular risk factors in subjects with acromegaly. Hormones (Athens). 2019;18(4):435-442.
  7. Giustina A, Barkan A, Beckers A, Biermasz N, Biller BMK, Boguszewski C, Bolanowski M, Bonert V, Bronstein MD, Casanueva FF, Clemmons D, Colao A, Ferone D, Fleseriu M, Frara S, Gadelha MR, Ghigo E, Gurnell M, Heaney AP, Ho K, Ioachimescu A, Katznelson L, Kelestimur F, Kopchick J, Krsek M, Lamberts S, Losa M, Luger A, Maffei P, Marazuela M, Mazziotti G, Mercado M, Mortini P, Neggers S, Pereira AM, Petersenn S, Puig-Domingo M, Salvatori R, Shimon I, Strasburger C, Tsagarakis S, van der Lely AJ, Wass J, Zatelli MC, Melmed S. A Consensus on the Diagnosis and Treatment of Acromegaly Comorbidities: An Update. J Clin Endocrinol Metab. 2020;105(4).
  8. Glynn N, Kenny H, Salim T, Halsall DJ, Smith D, Tun TK, McDermott JH, Tormey W, Thompson CJ, McAdam B, O' Gorman D, Agha A. ALTERATIONS IN THYROID HORMONE LEVELS FOLLOWING GROWTH HORMONE REPLACEMENT EXERT COMPLEX BIOLOGICAL EFFECTS. Endocr Pract. 2018;24(4):342-350.
  9. Dogan S, Atmaca A, Dagdelen S, Erbas B, Erbas T. Evaluation of thyroid diseases and differentiated thyroid cancer in acromegalic patients. Endocrine. 2014;45(1):114-121.
  10. Xu D, Wu B, Li X, Cheng Y, Chen D, Fang Y, Du Q, Chen Z, Wang X. Evaluation of the thyroid characteristics of patients with growth hormone-secreting adenomas. BMC Endocr Disord. 2019;19(1):94.
  11. Dogansen SC, Salmaslioglu A, Yalin GY, Tanrikulu S, Yarman S. Evaluation of the natural course of thyroid nodules in patients with acromegaly. Pituitary. 2019;22(1):29-36.
  12. Boguszewski CL, Ayuk J. MANAGEMENT OF ENDOCRINE DISEASE: Acromegaly and cancer: an old debate revisited. Eur J Endocrinol. 2016;175(4):R147-156.
  13. Katznelson L, Laws ER, Melmed S, Molitch ME, Murad MH, Utz A, Wass JA, Society E. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99(11):3933-3951.
  14. Terzolo M, Reimondo G, Berchialla P, Ferrante E, Malchiodi E, De Marinis L, Pivonello R, Grottoli S, Losa M, Cannavo S, Ferone D, Montini M, Bondanelli M, De Menis E, Martini C, Puxeddu E, Velardo A, Peri A, Faustini-Fustini M, Tita P, Pigliaru F, Peraga G, Borretta G, Scaroni C, Bazzoni N, Bianchi A, Berton A, Serban AL, Baldelli R, Fatti LM, Colao A, Arosio M, Acromegaly ISGo. Acromegaly is associated with increased cancer risk: a survey in Italy. Endocr Relat Cancer. 2017;24(9):495-504.
  15. Dal J, Leisner MZ, Hermansen K, Farkas DK, Bengtsen M, Kistorp C, Nielsen EH, Andersen M, Feldt-Rasmussen U, Dekkers OM, Sørensen HT, Jørgensen JOL. Cancer Incidence in Patients With Acromegaly: A Cohort Study and Meta-Analysis of the Literature. J Clin Endocrinol Metab. 2018;103(6):2182-2188.
  16. Perry JK, Wu ZS, Mertani HC, Zhu T, Lobie PE. Tumour-Derived Human Growth Hormone As a Therapeutic Target in Oncology. Trends Endocrinol Metab. 2017;28(8):587-596.
  17. Rokkas T, Pistiolas D, Sechopoulos P, Margantinis G, Koukoulis G. Risk of colorectal neoplasm in patients with acromegaly: a meta-analysis. World J Gastroenterol. 2008;14(22):3484-3489.
  18. Dworakowska D, Grossman AB. Colonic Cancer and Acromegaly. Front Endocrinol (Lausanne). 2019;10:390.
  19. Jenkins PJ, Frajese V, Jones AM, Camacho-Hubner C, Lowe DG, Fairclough PD, Chew SL, Grossman AB, Monson JP, Besser GM. Insulin-like growth factor I and the development of colorectal neoplasia in acromegaly. J Clin Endocrinol Metab. 2000;85(9):3218-3221.
  20. Jenkins PJ, Besser GM, Fairclough PD. Colorectal neoplasia in acromegaly. Gut. 1999;44(5):585-587.
  21. Jenkins PJ, Besser M. Clinical perspective: acromegaly and cancer: a problem. J Clin Endocrinol Metab.2001;86(7):2935-2941.
  22. Dworakowska D, Gueorguiev M, Kelly P, Monson JP, Besser GM, Chew SL, Akker SA, Drake WM, Fairclough PD, Grossman AB, Jenkins PJ. Repeated colonoscopic screening of patients with acromegaly: 15-year experience identifies those at risk of new colonic neoplasia and allows for effective screening guidelines. Eur J Endocrinol. 2010;163(1):21-28.
  23. Evans CC, Hipkin LJ, Murray GM. Pulmonary function in acromegaly. Thorax. 1977;32(3):322-327.
  24. Camilo GB, Carvalho AR, Machado DC, Mogami R, Melo PL, Lopes AJ. CT pulmonary densitovolumetry in patients with acromegaly: a comparison between active disease and controlled disease. Br J Radiol.2015;88(1054):20150315.
  25. Störmann S, Gutt B, Roemmler-Zehrer J, Bidlingmaier M, Huber RM, Schopohl J, Angstwurm MW. Assessment of lung function in a large cohort of patients with acromegaly. Eur J Endocrinol. 2017;177(1):15-23.
  26. Camilo GB, Guimarães FS, Mogami R, Faria AC, Melo PL, Lopes AJ. Functional changes are associated with tracheal structural abnormalities in patients with acromegaly. Arch Med Sci. 2016;12(1):78-88.
  27. Wu TE, Chen HS. Increased prevalence of proliferative retinopathy in patients with acromegaly. J Chin Med Assoc. 2018;81(3):230-235.
  28. Füchtbauer L, Olsson DS, Coopmans EC, Bengtsson B, Norrman LL, Neggers SJCM, Hellström A, Johannsson G. Increased number of retinal vessels in acromegaly. Eur J Endocrinol. 2020;182(3):293-302.
  29. Zafar A, Jordan DR. Enlarged extraocular muscles as the presenting feature of acromegaly. Ophthalmic Plast Reconstr Surg. 2004;20(4):334-336.
  30. Heireman S, Delaey C, Claerhout I, Decock CE. Restrictive extraocular myopathy: a presenting feature of acromegaly. Indian J Ophthalmol. 2011;59(6):517-519.
  31. Patrinely JR, Osborn AG, Anderson RL, Whiting AS. Computed tomographic features of nonthyroid extraocular muscle enlargement. Ophthalmology. 1989;96(7):1038-1047.
  32. Mehra M, Mohsin M, Sharma P, Dewan T, Taneja A, Kulshreshtha B. Epiphora and proptosis as a presenting complaint in acromegaly: Report of two cases with review of literature. Indian J Endocrinol Metab.2013;17(Suppl 1):S149-151.
  33. Akay F, Akmaz B, Işik MU, Güven YZ, Örük GG. Evaluation of the retinal layers and microvasculature in patients with acromegaly: a case-control OCT angiography study. Eye (Lond). 2020.
  34. Polat SB, Ugurlu N, Ersoy R, Oguz O, Duru N, Cakir B. Evaluation of central corneal and central retinal thicknesses and intraocular pressure in acromegaly patients. Pituitary. 2014;17(4):327-332.
  35. Tabur S, Korkmaz H, Baysal E, Hatipoglu E, Aytac I, Akarsu E. Auditory changes in acromegaly. J Endocrinol Invest. 2017;40(6):621-626.
  36. Aydin K, Ozturk B, Turkyilmaz MD, Dagdelen S, Ozgen B, Unal F, Erbas T. Functional and structural evaluation of hearing in acromegaly. Clin Endocrinol (Oxf). 2012;76(3):415-419.
  37. Teixeira LS, Silva IBO, Sampaio ALL, Oliveira CAP, Bahamad Júnior F. Hearing Loss in Acromegaly - A Review. Int Arch Otorhinolaryngol. 2018;22(3):313-316.
  38. Kyriakakis N, Pechlivani N, Lynch J, Oxley N, Phoenix F, Seejore K, Orme SM, Ajjan R, Murray RD. Prothrombotic fibrin network characteristics in patients with acromegaly: a novel mechanism for vascular complications. Eur J Endocrinol. 2020;182(5):511-521.
  39. Elarabi AM, Mosleh E, Alamlih LI, Albakri MM, Ibrahim WH. Massive Pulmonary Embolism as the Initial Presentation of Acromegaly: Is Acromegaly a Hypercoagulable Condition? Am J Case Rep. 2018;19:1541-1545.
  40. Campello E, Marobin M, Barbot M, Radu CM, Voltan G, Spiezia L, Gavasso S, Ceccato F, Scaroni C, Simioni P. The haemostatic system in acromegaly: a single-centre case-control study. J Endocrinol Invest. 2020.
  41. Zoppoli G, Bianchi F, Bruzzone A, Calvia A, Oneto C, Passalia C, Balleari E, Bedognetti D, Ponomareva E, Nazzari E, Castelletti L, Castellan L, Minuto F, Ghio R, Ferone D. Polycythemia as rare secondary direct manifestation of acromegaly: management and single-centre epidemiological data. Pituitary. 2012;15(2):209-214.
  42. Barbosa FR, Vieira Neto L, Lima GA, Wildemberg LE, Portugal R, Gadelha MR. Hematologic neoplasias and acromegaly. Pituitary. 2011;14(4):377-381.
  43. Kawaguchi H, Itoh K, Mori H, Hayashi Y, Makino S. Renal pathology in rats bearing tumor-secreting growth hormone. Pediatric Nephrology. 1991;5:533-538.
  44. Fujio S, Takano K, Arimura H, Habu M, Bohara M, Hirano H, Hanaya R, Nishio Y, Koriyama C, Kinoshita Y, Arita K. Treatable glomerular hyperfiltration in patients with active acromegaly. Eur J Endocrinol. 2016;175(4):325-333.
  45. Auriemma RS, Galdiero M, De Martino MC, De Leo M, Grasso LF, Vitale P, Cozzolino A, Lombardi G, Colao A, Pivonello R. The kidney in acromegaly: renal structure and function in patients with acromegaly during active disease and 1 year after disease remission. Eur J Endocrinol. 2010;162(6):1035-1042.
  46. Libório AB, Figueiredo PR, Montenegro Junior RM, Montenegro RM, Martins MR, Silva Junior GB, Porto IA, Mota JI, Daher E. Urinary calcium excretion and insulin resistance in patients with acromegaly. Int Urol Nephrol.2012;44(5):1473-1477.
  47. Sindelka G, Skrha J, Hilgertová J, Justová V. [Early diagnosis of impaired glomerular and renal tubule function in patients with acromegaly]. Cas Lek Cesk. 1996;135(20):657-659.
  48. Orme SM, McNally RJ, Cartwright RA, Belchetz PE. Mortality and cancer incidence in acromegaly: a retrospective cohort study. United Kingdom Acromegaly Study Group. J Clin Endocrinol Metab.1998;83(8):2730-2734.
  49. Alexander L, Appleton D, Hall R, Ross WM, Wilkinson R. Epidemiology of acromegaly in the Newcastle region. Clin Endocrinol (Oxf). 1980;12(1):71-79.
  50. Kasuki L, Rocha PDS, Lamback EB, Gadelha MR. Determinants of morbidities and mortality in acromegaly. Arch Endocrinol Metab. 2019;63(6):630-637.
  51. Esposito D, Ragnarsson O, Johannsson G, Olsson DS. Prolonged diagnostic delay in acromegaly is associated with increased morbidity and mortality. Eur J Endocrinol. 2020;182(6):523-531.
  52. Park KH, Lee EJ, Seo GH, Ku CR. Risk for Acromegaly-related Comorbidities by Sex in Korean Acromegaly. J Clin Endocrinol Metab. 2020;105(4).
  53. Giustina A, Bevan JS, Bronstein MD, Casanueva FF, Chanson P, Petersenn S, Thanh XM, Sert C, Houchard A, Guillemin I, Melmed S, Group SI. SAGIT®: clinician-reported outcome instrument for managing acromegaly in clinical practice--development and results from a pilot study. Pituitary. 2016;19(1):39-49.
  54. van der Lely AJ, Gomez R, Pleil A, Badia X, Brue T, Buchfelder M, Burman P, Clemmons D, Ghigo E, Jørgensen JOL, Luger A, van der Lans-Bussemaker J, Webb SM, Strasburger CJ. Development of ACRODAT. Pituitary. 2017;20(6):692-701.
  55. Paisley AN, Rowles SV, Roberts ME, Webb SM, Badia X, Prieto L, Shalet SM, Trainer PJ. Treatment of acromegaly improves quality of life, measured by AcroQol. Clin Endocrinol (Oxf). 2007;67(3):358-362.
  56. Fleseriu M, Fogelfeld L, Gordon MB, Sisco J, Crosby RD, Ludlam WH, Haviv A, Mathias SD. An evaluation of the Acromegaly Treatment Satisfaction Questionnaire (Acro-TSQ) in adult patients with acromegaly, including correlations with other patient-reported outcome measures: data from two large multicenter international studies. Pituitary. 2020.
  57. Wolters TLC, Roerink SHPP, Sterenborg RBTM, Wagenmakers MAEM, Husson O, Smit JWA, Hermus ARMM, Netea-Maier RT. The effect of treatment on quality of life in patients with acromegaly: a prospective study. Eur J Endocrinol. 2020;182(3):319-331.
  58. Webb SM, Badia X. Quality of Life in Acromegaly. Neuroendocrinology. 2016;103(1):106-111.
  59. Schilbach K, Bidlingmaier M. Laboratory investigations in the diagnosis and follow-up of GH-related disorders. Arch Endocrinol Metab. 2019;63(6):618-629.
  60. Junnila RK, Strasburger CJ, Bidlingmaier M. Pitfalls of insulin-like growth factor-i and growth hormone assays. Endocrinol Metab Clin North Am. 2015;44(1):27-34.
  61. Freda PU. Pitfalls in the biochemical assessment of acromegaly. Pituitary. 2003;6(3):135-140.
  62. Clemmons DR, on behalf of the conference p. Consensus Statement on the Standardization and Evaluation of Growth Hormone and Insulin-like Growth Factor Assays. Clinical Chemistry. 2011;57(4):555-559.
  63. Duncan E, Wass JA. Investigation protocol: acromegaly and its investigation. Clin Endocrinol (Oxf).1999;50(3):285-293.
  64. Camacho-Hübner C. Assessment of growth hormone status in acromegaly: what biochemical markers to measure and how? Growth Horm IGF Res. 2000;10 Suppl B:S125-129.
  65. Dobri G, Niwattisaiwong S, Bena JF, Gupta M, Kirwan J, Kennedy L, Hamrahian AH. Is GH nadir during OGTT a reliable test for diagnosis of acromegaly in patients with abnormal glucose metabolism? Endocrine.2019;64(1):139-146.
  66. Tzanela M. Dynamic tests and basal values for defining active acromegaly. Neuroendocrinology. 2006;83(3-4):200-204.
  67. Hulting AL, Theodorsson E, Werner S. Thyrotropin-releasing hormone increases serum levels of growth hormone-releasing hormone and growth hormone in patients with acromegaly. J Intern Med. 1992;232(3):229-235.
  68. Kageyama K, Moriyama T, Sakihara S, Takayasu S, Nigawara T, Suda T. Usefulness of the thyrotropin-releasing hormone test in pre-clinical acromegaly. Tohoku J Exp Med. 2005;206(4):291-297.
  69. de Herder WW, van der Lely AJ, Janssen JA, Uitterlinden P, Hofland LJ, Lamberts SW. IGFBP-3 is a poor parameter for assessment of clinical activity in acromegaly. Clin Endocrinol (Oxf). 1995;43(4):501-505.
  70. Freda PU. Current concepts in the biochemical assessment of the patient with acromegaly. Growth Hormone & IGF Research. 2003;13(4):171-184.
  71. Wang M, Shen M, He W, Yang Y, Liu W, Lu Y, Ma Z, Ye Z, Zhang Y, Zhao X, Lu B, Hu J, Huang Y, Shou X, Wang Y, Ye H, Li Y, Li S, Zhao Y, Zhang Z. The value of an acute octreotide suppression test in predicting short-term efficacy of somatostatin analogues in acromegaly. Endocr J. 2016;63(9):819-834.
  72. Karavitaki N, Botusan I, Radian S, Coculescu M, Turner HE, Wass JA. The value of an acute octreotide suppression test in predicting long-term responses to depot somatostatin analogues in patients with active acromegaly. Clin Endocrinol (Oxf). 2005;62(3):282-288.
  73. Cazabat L, Souberbielle JC, Chanson P. Dynamic tests for the diagnosis and assessment of treatment efficacy in acromegaly. Pituitary. 2008;11(2):129-139.
  74. Cozzi R, Ambrosio MR, Attanasio R, Bozzao A, De Marinis L, De Menis E, Guastamacchia E, Lania A, Lasio G, Logoluso F, Maffei P, Poggi M, Toscano V, Zini M, Chanson P, Katznelson L. ITALIAN ASSOCIATION OF CLINICAL ENDOCRINOLOGISTS (AME) and ITALIAN AACE CHAPTER POSITION STATEMENT FOR CLINICAL PRACTICE: ACROMEGALY - PART 1: DIAGNOSTIC AND CLINICAL ISSUES. Endocr Metab Immune Disord Drug Targets. 2020.
  75. Kristof RA, Neuloh G, Redel L, Klingmüller D, Schramm J. Reliability of the oral glucose tolerance test in the early postoperative assessment of acromegaly remission. J Neurosurg. 2002;97(6):1282-1286.
  76. Peacey SR, Toogood AA, Veldhuis JD, Thorner MO, Shalet SM. The relationship between 24-hour growth hormone secretion and insulin-like growth factor I in patients with successfully treated acromegaly: impact of surgery or radiotherapy. J Clin Endocrinol Metab. 2001;86(1):259-266.
  77. Bourdelot A, Coste J, Hazebroucq V, Gaillard S, Cazabat L, Bertagna X, Bertherat J. Clinical, hormonal and magnetic resonance imaging (MRI) predictors of transsphenoidal surgery outcome in acromegaly. Eur J Endocrinol. 2004;150(6):763-771.
  78. Antunes X, Ventura N, Camilo GB, Wildemberg LE, Guasti A, Pereira PJM, Camacho AHS, Chimelli L, Niemeyer P, Gadelha MR, Kasuki L. Predictors of surgical outcome and early criteria of remission in acromegaly. Endocrine. 2018;60(3):415-422.
  79. Nishioka H, Fukuhara N, Horiguchi K, Yamada S. Aggressive transsphenoidal resection of tumors invading the cavernous sinus in patients with acromegaly: predictive factors, strategies, and outcomes. J Neurosurg.2014;121(3):505-510.
  80. Puig-Domingo M, Resmini E, Gomez-Anson B, Nicolau J, Mora M, Palomera E, Martí C, Halperin I, Webb SM. Magnetic resonance imaging as a predictor of response to somatostatin analogs in acromegaly after surgical failure. J Clin Endocrinol Metab. 2010;95(11):4973-4978.
  81. Potorac I, Petrossians P, Daly AF, Alexopoulou O, Borot S, Sahnoun-Fathallah M, Castinetti F, Devuyst F, Jaffrain-Rea ML, Briet C, Luca F, Lapoirie M, Zoicas F, Simoneau I, Diallo AM, Muhammad A, Kelestimur F, Nazzari E, Centeno RG, Webb SM, Nunes ML, Hana V, Pascal-Vigneron V, Ilovayskaya I, Nasybullina F, Achir S, Ferone D, Neggers SJ, Delemer B, Petit JM, Schöfl C, Raverot G, Goichot B, Rodien P, Corvilain B, Brue T, Schillo F, Tshibanda L, Maiter D, Bonneville JF, Beckers A. T2-weighted MRI signal predicts hormone and tumor responses to somatostatin analogs in acromegaly. Endocr Relat Cancer. 2016;23(11):871-881.
  82. Hagiwara A, Inoue Y, Wakasa K, Haba T, Tashiro T, Miyamoto T. Comparison of growth hormone-producing and non-growth hormone-producing pituitary adenomas: imaging characteristics and pathologic correlation. Radiology. 2003;228(2):533-538.
  83. Potorac I, Petrossians P, Daly AF, Schillo F, Ben Slama C, Nagi S, Sahnoun M, Brue T, Girard N, Chanson P, Nasser G, Caron P, Bonneville F, Raverot G, Lapras V, Cotton F, Delemer B, Higel B, Boulin A, Gaillard S, Luca F, Goichot B, Dietemann JL, Beckers A, Bonneville JF. Pituitary MRI characteristics in 297 acromegaly patients based on T2-weighted sequences. Endocr Relat Cancer. 2015;22(2):169-177.
  84. Cozzi R, Ambrosio MR, Attanasio R, Bozzao A, De Marinis L, De Menis E, Guastamacchia E, Lania A, Lasio G, Logoluso F, Maffei P, Poggi M, Toscano V, Zini M, Chanson P, Katznelson L. Italian Association Of Clinical Endocrinologists (Ame) And Italian Aace Chapter Position Statement For Clinical Practice: Acromegaly - Part 2: Therapeutic Issues. Endocr Metab Immune Disord Drug Targets. 2020.
  85. Rodriguez-Barcelo S, Gutierrez-Cardo A, Dominguez-Paez M, Medina-Imbroda J, Romero-Moreno L, Arraez-Sanchez M. Clinical usefulness of coregistered 11C-methionine positron emission tomography/3-T magnetic resonance imaging at the follow-up of acromegaly. World Neurosurg. 2014;82(3-4):468-473.
  86. Feng Z, He D, Mao Z, Wang Z, Zhu Y, Zhang X, Wang H. Utility of 11C-Methionine and 18F-FDG PET/CT in Patients With Functioning Pituitary Adenomas. Clin Nucl Med. 2016;41(3):e130-134.
  87. Wang Z, Mao Z, Zhang X, He D, Wang X, Du Q, Xiao Z, Zhu D, Zhu Y, Wang H. Utility of (13)N-Ammonia PET/CT to Detect Pituitary Tissue in Patients with Pituitary Adenomas. Acad Radiol. 2019;26(9):1222-1228.
  88. Waligórska-Stachura J, Gut P, Sawicka-Gutaj N, Liebert W, Gryczyńska M, Baszko-Błaszyk D, Blanco-Gangoo AR, Ruchała M. Growth hormone-secreting macroadenoma of the pituitary gland successfully treated with the radiolabeled somatostatin analog (90)Y-DOTATATE: case report. J Neurosurg. 2016;125(2):346-349.
  89. Asa SL, Mete O. Immunohistochemical Biomarkers in Pituitary Pathology. Endocr Pathol. 2018;29(2):130-136.
  90. Chahal HS, Stals K, Unterländer M, Balding DJ, Thomas MG, Kumar AV, Besser GM, Atkinson AB, Morrison PJ, Howlett TA, Levy MJ, Orme SM, Akker SA, Abel RL, Grossman AB, Burger J, Ellard S, Korbonits M. AIP mutation in pituitary adenomas in the 18th century and today. N Engl J Med. 2011;364(1):43-50.
  91. Daly AF, Tichomirowa MA, Petrossians P, Heliövaara E, Jaffrain-Rea ML, Barlier A, Naves LA, Ebeling T, Karhu A, Raappana A, Cazabat L, De Menis E, Montañana CF, Raverot G, Weil RJ, Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E, Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo RA, Sabaté MI, Villa C, Popelier M, Salvatori R, Jennings J, Longás AF, Labarta Aizpún JI, Georgitsi M, Paschke R, Ronchi C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT, Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A, Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen LA, Beckers A. Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab. 2010;95(11):E373-383.
  92. Schöfl C, Honegger J, Droste M, Grussendorf M, Finke R, Plöckinger U, Berg C, Willenberg HS, Lammert A, Klingmüller D, Jaursch-Hancke C, Tönjes A, Schneidewind S, Flitsch J, Bullmann C, Dimopoulou C, Stalla G, Mayr B, Hoeppner W, Schopohl J. Frequency of AIP gene mutations in young patients with acromegaly: a registry-based study. J Clin Endocrinol Metab. 2014;99(12):E2789-2793.
  93. Hannah-Shmouni F, Trivellin G, Stratakis CA. Genetics of gigantism and acromegaly. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society.2016;30-31:37-41.
  94. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, Melmed S, Sakurai A, Tonelli F, Brandi ML. Clinical Practice Guidelines for Multiple Endocrine Neoplasia Type 1 (MEN1). The Journal of Clinical Endocrinology & Metabolism. 2012;97(9):2990-3011.
  95. Gadelha MR, Kasuki L, Korbonits M. The genetic background of acromegaly. Pituitary. 2017;20(1):10-21.
  96. Hannah-Shmouni F, Trivellin G, Stratakis CA. Genetics of gigantism and acromegaly. Growth Horm IGF Res.2016;30-31:37-41.
  97. Karimova MM, Halimova ZY, Urmanova YM, Korbonits M, Cranston T, Grossman AB. Pachydermoperiostosis Masquerading as Acromegaly. J Endocr Soc. 2017;1(2):109-112.
  98. Melmed S, Bronstein MD, Chanson P, Klibanski A, Casanueva FF, Wass JAH, Strasburger CJ, Luger A, Clemmons DR, Giustina A. A Consensus Statement on acromegaly therapeutic outcomes. Nat Rev Endocrinol.2018;14(9):552-561.
  99. Fahlbusch R, Keller B, Ganslandt O, Kreutzer J, Nimsky C. Transsphenoidal surgery in acromegaly investigated by intraoperative high-field magnetic resonance imaging. Eur J Endocrinol. 2005;153(2):239-248.
  100. Abe T, Lüdecke DK. Recent primary transnasal surgical outcomes associated with intraoperative growth hormone measurement in acromegaly. Clin Endocrinol (Oxf). 1999;50(1):27-35.
  101. Fathalla H, Cusimano MD, Di Ieva A, Lee J, Alsharif O, Goguen J, Zhang S, Smyth H. Endoscopic versus microscopic approach for surgical treatment of acromegaly. Neurosurg Rev. 2015;38(3):541-548; discussion 548-549.
  102. Chen CJ, Ironside N, Pomeraniec IJ, Chivukula S, Buell TJ, Ding D, Taylor DG, Dallapiazza RF, Lee CC, Bergsneider M. Microsurgical versus endoscopic transsphenoidal resection for acromegaly: a systematic review of outcomes and complications. Acta Neurochir (Wien). 2017;159(11):2193-2207.
  103. Araujo-Castro M, Pascual-Corrales E, Martínez-Vaello V, Baonza Saiz G, Quiñones de Silva J, Acitores Cancela A, García Cano AM, Rodríguez Berrocal V. Predictive model of surgical remission in acromegaly: age, presurgical GH levels and Knosp grade as the best predictors of surgical remission. J Endocrinol Invest. 2020.
  104. Jenkins PJ, Emery M, Howling SJ, Evanson J, Besser GM, Monson JP. Predicting therapeutic response and degree of pituitary tumour shrinkage during treatment of acromegaly with octreotide LAR. Horm Res.2004;62(5):227-232.
  105. Sims-Williams HP, Rajapaksa K, Sinha S, Radatz M, Walton L, Yianni J, Newell-Price J. Radiosurgery as primary management for acromegaly. Clin Endocrinol (Oxf). 2019;90(1):114-121.
  106. Jenkins PJ, Bates P, Carson MN, Stewart PM, Wass JA. Conventional pituitary irradiation is effective in lowering serum growth hormone and insulin-like growth factor-I in patients with acromegaly. J Clin Endocrinol Metab.2006;91(4):1239-1245.
  107. Cozzi R, Barausse M, Asnaghi D, Dallabonzana D, Lodrini S, Attanasio R. Failure of radiotherapy in acromegaly. Eur J Endocrinol. 2001;145(6):717-726.
  108. Minniti G, Scaringi C, Enrici RM. Radiation techniques for acromegaly. Radiat Oncol. 2011;6:167.
  109. Gheorghiu ML. Updates in outcomes of stereotactic radiation therapy in acromegaly. Pituitary. 2017;20(1):154-168.
  110. Landolt AM, Haller D, Lomax N, Scheib S, Schubiger O, Siegfried J, Wellis G. Stereotactic radiosurgery for recurrent surgically treated acromegaly: comparison with fractionated radiotherapy. J Neurosurg.1998;88(6):1002-1008.
  111. Besser GM, Wass JA, Thorner MO. Acromegaly--results of long term treatment with bromocriptine. Acta Endocrinol Suppl (Copenh). 1978;216:187-198.
  112. Abs R, Verhelst J, Maiter D, Van Acker K, Nobels F, Coolens JL, Mahler C, Beckers A. Cabergoline in the treatment of acromegaly: a study in 64 patients. J Clin Endocrinol Metab. 1998;83(2):374-378.
  113. Sandret L, Maison P, Chanson P. Place of cabergoline in acromegaly: a meta-analysis. J Clin Endocrinol Metab.2011;96(5):1327-1335.
  114. Valea A, Ghervan C, Carsote M, Morar A, Iacob I, Tomesc F, Pop DD, Georgescu C. Effects of combination therapy: somatostatin analogues and dopamine agonists on GH and IGF1 levels in acromegaly. Clujul Med.2015;88(3):310-313.
  115. Auriemma RS, Pivonello R, Galdiero M, De Martino MC, De Leo M, Vitale G, Lombardi G, Colao A. Octreotide-LAR vs lanreotide-SR as first-line therapy for acromegaly: a retrospective, comparative, head-to-head study. J Endocrinol Invest. 2008;31(11):956-965.
  116. Freda PU, Katznelson L, van der Lely AJ, Reyes CM, Zhao S, Rabinowitz D. Long-acting somatostatin analog therapy of acromegaly: a meta-analysis. J Clin Endocrinol Metab. 2005;90(8):4465-4473.
  117. Biermasz NR, van den Oever NC, Frölich M, Arias AM, Smit JW, Romijn JA, Roelfsema F. Sandostatin LAR in acromegaly: a 6-week injection interval suppresses GH secretion as effectively as a 4-week interval. Clin Endocrinol (Oxf). 2003;58(3):288-295.
  118. Bruns C, Lewis I, Briner U, Meno-Tetang G, Weckbecker G. SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol. 2002;146(5):707-716.
  119. Colao A, Bronstein MD, Brue T, De Marinis L, Fleseriu M, Guitelman M, Raverot G, Shimon I, Fleck J, Gupta P, Pedroncelli AM, Gadelha MR. Pasireotide for acromegaly: long-term outcomes from an extension to the Phase III PAOLA study. Eur J Endocrinol. 2020;182(6):583.
  120. Gadelha MR, Bronstein MD, Brue T, Coculescu M, Fleseriu M, Guitelman M, Pronin V, Raverot G, Shimon I, Lievre KK, Fleck J, Aout M, Pedroncelli AM, Colao A, Group PCS. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, phase 3 trial. Lancet Diabetes Endocrinol. 2014;2(11):875-884.
  121. Melmed S, Popovic V, Bidlingmaier M, Mercado M, van der Lely AJ, Biermasz N, Bolanowski M, Coculescu M, Schopohl J, Racz K, Glaser B, Goth M, Greenman Y, Trainer P, Mezosi E, Shimon I, Giustina A, Korbonits M, Bronstein MD, Kleinberg D, Teichman S, Gliko-Kabir I, Mamluk R, Haviv A, Strasburger C. Safety and efficacy of oral octreotide in acromegaly: results of a multicenter phase III trial. J Clin Endocrinol Metab. 2015;100(4):1699-1708.
  122. Biermasz NR. New medical therapies on the horizon: oral octreotide. Pituitary. 2017;20(1):149-153.
  123. Nunes VS, Correa JM, Puga ME, Silva EM, Boguszewski CL. Preoperative somatostatin analogues versus direct transsphenoidal surgery for newly-diagnosed acromegaly patients: a systematic review and meta-analysis using the GRADE system. Pituitary. 2015;18(4):500-508.
  124. Albarel F, Castinetti F, Morange I, Guibert N, Graillon T, Dufour H, Brue T. Pre-surgical medical treatment, a major prognostic factor for long-term remission in acromegaly. Pituitary. 2018;21(6):615-623.
  125. van der Lely AJ, Kuhn E, Muhammad A, Coopmans EC, Neggers SJ, Chanson P. Pegvisomant and not somatostatin receptor ligands (SRLs) is first-line medical therapy for acromegaly. Eur J Endocrinol.2020;182(6):D17-D29.
  126. van der Lely AJ, Hutson RK, Trainer PJ, Besser GM, Barkan AL, Katznelson L, Klibanski A, Herman-Bonert V, Melmed S, Vance ML, Freda PU, Stewart PM, Friend KE, Clemmons DR, Johannsson G, Stavrou S, Cook DM, Phillips LS, Strasburger CJ, Hackett S, Zib KA, Davis RJ, Scarlett JA, Thorner MO. Long-term treatment of acromegaly with pegvisomant, a growth hormone receptor antagonist. Lancet. 2001;358(9295):1754-1759.
  127. Feenstra J, de Herder WW, ten Have SM, van den Beld AW, Feelders RA, Janssen JA, van der Lely AJ. Combined therapy with somatostatin analogues and weekly pegvisomant in active acromegaly. Lancet.2005;365(9471):1644-1646.
  128. Leonart LP, Riveros BS, Krahn MD, Pontarolo R. Pharmacological acromegaly treatment: cost-utility and value of information analysis. Neuroendocrinology. 2020.
  129. Higham CE, Atkinson AB, Aylwin S, Bidlingmaier M, Drake WM, Lewis A, Martin NM, Moyes V, Newell-Price J, Trainer PJ. Effective combination treatment with cabergoline and low-dose pegvisomant in active acromegaly: a prospective clinical trial. J Clin Endocrinol Metab. 2012;97(4):1187-1193.
  130. Ramos-Leví AM, Bernabeu I, Álvarez-Escolá C, Aller J, Lucas T, de Miguel P, Rodríguez-Cañete L, Sampedro-Núñez MA, Halperin I, Puig-Domingo M, Marazuela M. Long-term treatment with pegvisomant for acromegaly: a 10-year experience. Clin Endocrinol (Oxf). 2016;84(4):540-550.
  131. Maffezzoni F, Frara S, Doga M, Mazziotti G, Giustina A. New medical therapies of acromegaly. Growth Horm IGF Res. 2016;30-31:58-63.
  132. Melmed S. New therapeutic agents for acromegaly. Nat Rev Endocrinol. 2016;12(2):90-98.
  133. Stone JC, Clark J, Cuneo R, Russell AW, Doi SA. Estrogen and selective estrogen receptor modulators (SERMs) for the treatment of acromegaly: a meta-analysis of published observational studies. Pituitary.2014;17(3):284-295.
  134. Gadelha MR, Chieffo C, Bai SA, Hu X, Frohman LA. A subcutaneous octreotide hydrogel implant for the treatment of acromegaly. Endocr Pract. 2012;18(6):870-881.
  135. Alexander SGM, Adelheid W, Stefan W, Engelbert K. Invasion of the cavernous sinus space in pituitary adenomas: endoscopic verification and its correlation with an MRI-based classification. Journal of Neurosurgery JNS. 2015;122(4):803-811.
  136. Kasuki L, Wildemberg LE, Gadelha MR. MANAGEMENT OF ENDOCRINE DISEASE: Personalized medicine in the treatment of acromegaly. Eur J Endocrinol. 2018;178(3):R89-R100.
  137. Briceno V, Zaidi HA, Doucette JA, Onomichi KB, Alreshidi A, Mekary RA, Smith TR. Efficacy of transsphenoidal surgery in achieving biochemical cure of growth hormone-secreting pituitary adenomas among patients with cavernous sinus invasion: a systematic review and meta-analysis. Neurological Research. 2017;39(5):387-398.
  138. Buchfelder M, Schlaffer SM. The surgical treatment of acromegaly. Pituitary. 2017;20(1):76-83.
  139. Bollerslev J, Heck A, Olarescu NC. MANAGEMENT OF ENDOCRINE DISEASE: Individualised management of acromegaly. Eur J Endocrinol. 2019;181(2):R57-R71.
  140. Muhammad A, Coopmans EC, Gatto F, Franck SE, Janssen JAMJ, van der Lely AJ, Hofland LJ, Neggers SJCM. Pasireotide Responsiveness in Acromegaly Is Mainly Driven by Somatostatin Receptor Subtype 2 Expression. J Clin Endocrinol Metab. 2019;104(3):915-924.
  141. Donato I, Eivind C, Francesca L, Sabrina C, Serena P, Antonio B, Antonella G, Marilda M, Andrew JC, Francesco D, Carmelo A, Giulio M, Libero L, Guido R, Federico R, Alfredo P, Márta K, Laura De M. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. European Journal of Endocrinology. 2016;174(2):241-250.
  142. Basavilbaso NXG, Ballarino MC, Bruera D, Bruno OD, Chervin AB, Danilowicz K, Fainstein-Day P, Fidalgo SG, Frigeri A, Glerean M, Guelman R, Isaac G, Katz DA, Knoblovits P, Librandi F, Montes ML, Mallea-Gil MS, Manavela M, Mereshian P, Moncet D, Pignatta A, Rogozinsky A, Sago LR, Servidio M, Spezzi M, Stalldecker G, Tkatch J, Vitale NM, Guitelman M. Pegvisomant in acromegaly: a multicenter real-life study in Argentina. Arch Endocrinol Metab. 2019;63(4):320-327.
  143. Ezzat S, Gaspo R, Serri O, Ur E, Chik CL. A Canadian multi-centre, open-label long-term study of Pegvisomant treatment in refractory acromegaly. Clin Invest Med. 2009;32(6):E265.
  144. Chiloiro S, Bima C, Tartaglione T, Giampietro A, Gessi M, Lauretti L, Anile C, Colosimo C, Rindi G, Pontecorvi A, De Marinis L, Bianchi A. Pasireotide and Pegvisomant Combination Treatment in Acromegaly Resistant to Second-Line Therapies: A Longitudinal Study. J Clin Endocrinol Metab. 2019;104(11):5478-5482.
  145. Dutta P, Reddy KS, Rai A, Madugundu AK, Solanki HS, Bhansali A, Radotra BD, Kumar N, Collier D, Iacovazzo D, Gupta P, Raja R, Gowda H, Pandey A, Devgun JS, Korbonits M. Surgery, Octreotide, Temozolomide, Bevacizumab, Radiotherapy, and Pegvisomant Treatment of an AIP Mutation‒Positive Child. J Clin Endocrinol Metab. 2019;104(8):3539-3544.
  146. Beckers A, Stevenaert A, Foidart J-M, Hennen G, Frankenne F. Placental and Pituitary Growth Hormone Secretion during Pregnancy in Acromegalic Women. The Journal of Clinical Endocrinology & Metabolism.1990;71(3):725-731.
  147. Caron P, Broussaud S, Bertherat J, Borson-Chazot F, Brue T, Cortet-Rudelli C, Chanson P. Acromegaly and pregnancy: a retrospective multicenter study of 59 pregnancies in 46 women. J Clin Endocrinol Metab.2010;95(10):4680-4687.
  148. Cheng S, Grasso L, Martinez-Orozco JA, Al-Agha R, Pivonello R, Colao A, Ezzat S. Pregnancy in acromegaly: experience from two referral centers and systematic review of the literature. Clin Endocrinol (Oxf).2012;76(2):264-271.
  149. Muhammad A, Neggers SJ, van der Lely AJ. Pregnancy and acromegaly. Pituitary. 2017;20(1):179-184.
  150. Abucham J, Bronstein MD, Dias ML. MANAGEMENT OF ENDOCRINE DISEASE: Acromegaly and pregnancy: a contemporary review. Eur J Endocrinol. 2017;177(1):R1-R12.
  151. van der Lely AJ, Gomez R, Heissler JF, Åkerblad AC, Jönsson P, Camacho-Hübner C, Kołtowska-Häggström M. Pregnancy in acromegaly patients treated with pegvisomant. Endocrine. 2015;49(3):769-773.
  152. Trainer PJ, Newell-Price JDC, Ayuk J, Aylwin SJB, Rees A, Drake W, Chanson P, Brue T, Webb SM, Fajardo C, Aller J, McCormack AI, Torpy DJ, Tachas G, Atley L, Ryder D, Bidlingmaier M. A randomised, open-label, parallel group phase 2 study of antisense oligonucleotide therapy in acromegaly. Eur J Endocrinol.2018;179(2):97-108.
  153. Jaquet P, Gunz G, Saveanu A, Barlier A, Dufour H, Taylor J, Dong J, Kim S, Moreau JP, Culler MD. BIM-23A760, a chimeric molecule directed towards somatostatin and dopamine receptors, vs universal somatostatin receptors ligands in GH-secreting pituitary adenomas partial responders to octreotide. J Endocrinol Invest.2005;28(11 Suppl International):21-27.
  154. Kim J, Oh JH, Harlem H, Culler MD, Ku CR, Lee EJ. Therapeutic Effect of a Novel Chimeric Molecule Targeting Both Somatostatin and Dopamine Receptors on Growth Hormone-Secreting Pituitary Adenomas. Endocrinol Metab (Seoul). 2020;35(1):177-187.
  155. Zhou C, Jiao Y, Wang R, Ren SG, Wawrowsky K, Melmed S. STAT3 upregulation in pituitary somatotroph adenomas induces growth hormone hypersecretion. J Clin Invest. 2015;125(4):1692-1702.
  156. Pavel M, Borson-Chazot F, Cailleux A, Hörsch D, Lahner H, Pivonello R, Tauchmanova L, Darstein C, Olsson H, Tiberg F, Ferone D. Octreotide SC depot in patients with acromegaly and functioning neuroendocrine tumors: a phase 2, multicenter study. Cancer Chemother Pharmacol. 2019;83(2):375-385.
  157. Neggers SJ, Kopchick JJ, Jorgensen JO, van der Lely AJ.Hypothesis : Extra-hepatic acromegaly : a new paradigm ? 

           European Journal of Endocrinology.  2011 ; 164 : 11-16.

 

Endocrine Changes In Obesity

ABSTRACT

 

Obesity can be associated with several endocrine alterations arising from changes in the hypothalamic-pituitary hormones axes. These include hypothyroidism, Cushing’s disease, hypogonadism, and growth hormone deficiency. Besides its role in energy storage, adipose tissue has many other important functions that can be mediated through hormones or substances synthesized and released by adipocytes, including leptin and adiponectin. Further, obesity is also a common feature of polycystic ovarian syndrome with hyperinsulinemia being the primary etiological factor. Here, we provide an overview of several endocrine syndromes known to result in obesity and discuss the endocrine role of adipose tissue in conjunction to its association with hypothalamic-pituitary-endocrine axes.

 

INTRODUCTION

 

This chapter will discuss the endocrine role of adipose tissue and how alterations in each of the hypothalamic-pituitary-endocrine axes can occur in association with obesity. Of particular relevance is the possible bidirectionality of the relationships between endocrine changes and obesity: whether they are secondary to obesity or, in some cases, be a contributive factor to the development and/or perpetuation of obesity.

 

The endocrine axes of the human body are dynamic systems; they frequently show changes in response to stress, disease, or other pathological states. For example, during acute and chronic illnesses, and low calorie or starvation states, levels of thyroid, gonadal, and growth hormone are altered, returning to normal as the subject recovers. These hormonal changes are, therefore, thought to be secondary to the disease state and their recovery is reflective of homeostatic responses. Often these "adaptive" changes in hormonal dynamics may not necessarily be appropriate. Likewise, therapeutic measures aimed at restoring "normal" serum level of perturbed hormones offered in hopes of hastening recovery and improve patient outcomes have generally not been shown to be beneficial.

 

The weight gain that leads to obesity is the consequence of a positive energy balance, which can result from an increased energy intake, decreased energy expenditure, or both. This misalignment may be thought of as a failure of the body's homeostatic mechanisms to match energy intake with expenditure. Different obesity phenotypes may have variable health implications. For example, abdominal obesity is considered a more hazardous condition than gluteofemoral, or gynecoid, obesity. In those with abdominal obesity, accumulation of intraperitoneal fat (omental and visceral fat) carries greater health risk than the subcutaneous compartment. Therefore, when discussing complications of and metabolic abnormalities associated with obesity, different obesity phenotypes are recognized to carry different degrees of cardiometabolic risk.

 

Our understanding of the physiology of adipose tissue has greatly advanced in the last decade and extensive research has been dedicated to the study of the interactions between the adipose tissue and other bodily systems, in particular the central nervous system. New hormones have been discovered with potentially important roles in energy balance and food intake. The roles of many of these newly discovered hormones have not been fully elucidated in humans, but the future holds promise in not only improving our knowledge of the pathophysiology of obesity but also in developing novel therapeutic approaches to complement our currently, rather limited, pharmacological arsenal.

 

ADIPOSE TISSUE AS AN ENDOCRINE ORGAN 

 

Adipose Tissue

 

Adipose tissue has many important functions other than energy storage that are mediated through hormones or substances synthesized and released by adipocytes. These substances, termed "adipocytokines," act on distant targets in an endocrine fashion or locally in paracrine and autocrine fashions. In the following paragraphs, we shall discuss a few of the important adipocytokines secreted from “white" fat. For further characterizations of other types of adipose tissue, including "brown" and “pink” fat, see the Endotext chapter (1).

 

Leptin

 

The hormone leptin (from the Greek word ''leptos'' meaning ''thin'') is a 167-amino acid peptide hormone encoded by the ob (obesity) gene and secreted by white adipocytes. Its discovery in 1994, has greatly improved our understanding of how adipose tissue "communicates" with other systems in the body, in particular with the central nervous system (CNS). Following release into the circulation, leptin crosses the blood–brain barrier and binds to presynaptic GABAergic neurons of the hypothalamus of the CNS controlling appetite and energy expenditure (2). One of leptin’s key roles is thought to be as a signal of inadequate food intake or starvation. For example, leptin levels decline during fasting, low-calorie dieting, and uncontrolled type 1 diabetes. In these situations, the reduced leptin levels stimulate hunger while decreasing energy expenditure and engendering other physiologic adaptations that restore fat stores, and in turn leptin levels, to baseline (3,4).

 

On the other hand, serum concentrations of leptin increase in proportion to increasing adiposity. As a regulatory signal in a homeostatic system, if the leptin receptor is functioning normally, then higher circulating leptin levels should result in decreased energy intake and elevated energy expenditure, but this is not the case when individuals become overweight or obese. Instead, in patients with obesity high leptin levels are associated with low circulating soluble leptin receptors (SLR) consistent with a state of leptin resistance (5). Leptin must cross the blood–brain barrier (BBB) to reach the hypothalamus and exert its anorexigenic functions. Decreased transport across the blood-brain barrier (6)and a decreased ability of leptin to activate hypothalamic signaling in diet-induced obesity (7-9) may be crucial in the pathogenesis of leptin resistance.

 

In addition, anatomical and physiological changes that obesity can cause to the hypothalamus include expression of leptin signaling inhibitors, hypothalamic inflammatory signaling and gliosis, and endoplasmic reticulum stress.Elevated leptin itself may attenuate downstream leptin action, creating a functional ceiling for leptin action (10). These changes, together with the blood-brain barrier alterations, contribute to a failure of rising leptin levels to adequately compensate for the positive energy balance and thus promote the state of unwanted weight gain and obesity. Taken together, evidence points to leptin’s primary function as a defense against decreased body weight rather than to limit increases in body weight (10)

 

Data also suggests that leptin resistance can be a pre-conditioning factor contributing to diet induced obesity. Animal studies show that rats with a pre-existing reduction in leptin sensitivity develop excessive diet-induced obesity without eating more calories or altering their leptin sensitivity (11). This postulated leptin resistance is a major target in the search for a better understanding of obesity and the development of pharmacological tools to treat this chronic disease.

 

Most people with obesity are hyperleptinemic and show little or no weight loss after leptin treatment. However, recent evidence has indicated that a subset of patients with obesity have low endogenous plasma leptin levels and robustly respond to leptin treatment (12). These findings have led to a proposed classification of obesity based in leptin secretion and action. Type 1 obesity is associated with low leptin levels and leptin replacement can be an effective treatment in these forms of diabetes and obesity. Examples of patient populations in which this is more likely to be true include children with early onset and severe obesity (congenital leptin deficiency) (13) and those with generalized non-HIV lipodystrophy in whom recombinant methionyl human leptin has been FDA approved (14,15). Type 2 obesity is associated with leptin resistance, in which case leptin replacement is not optimal and other therapeutic approaches should be pursued (12).

 

Leptin plays a significant permissive role in the physiological regulation of several neuroendocrine axes, including the hypothalamic-pituitary-gonadal, thyroid, growth hormone, and adrenal axes (16,17). Leptin regulates reproductive function by altering the sensitivity of the pituitary gland to GnRH and acting at the ovary to alter follicular and luteal steroidogenesis, proliferation, and apoptosis (17). Thus, leptin serves as a putative signal that links metabolic status with the reproductive axis.

 

Leptin receptors are also present in peripheral organs, such as the liver, skeletal muscles, pancreatic beta cells, and even adipose cells, indicating endocrine, autocrine, and paracrine roles of leptin in energy regulation. Leptin signaling in these organs is thought to mediate important metabolic effects. For example, leptin has been implicated in glucose and lipid metabolism as an insulin-sensitizer (18). It has been shown to decrease glucagon synthesis and secretion, decrease hepatic glucose production, increase insulin hepatic extraction, decrease lipogenesis in the adipose tissue, and increase lipolysis among multiple other beneficial effects on insulin and lipids metabolism (19).

 

Other identified links between leptin and biological systems include expression of leptin by placenta and in fetal tissues. In this context, leptin is thought to be important for placentation,  maternal-fetal nutrition, and stimulating hematogenesis and angiogenesis in the regulation of fetal growth and development (20). On the other hand, the pathological expansion of white adipose tissue during expression of obesity and subsequent increases in cytokines and leptin have been implicated in worsening local and systemic inflammation, sustained proliferative signaling, epithelial-to-mesenchymal transition, angiogenesis, and cellular energetics (21) in association with increased risk of endometrial, kidney, and breast cancers (21,22).

 

Adiponectin

 

Adiponectin is another important adipocytokine that influences insulin sensitivity and atherogenesis. Adiponectin mediates its effect through binding to receptors AdipoR1 and AdipoR2, leading to activation of adenosine monophosphate dependent kinase, PPAR-α, and other yet-unidentified signaling pathways (23). Lower levels of adiponectin in obesity have been associated with insulin resistance (24), dyslipidemia (25), and atherosclerosis (26) in humans. With weight loss, plasma adiponectin levels significantly increase in parallel with improvements in insulin sensitivity (27). In a study with 2258 children with overweight or obesity, independent of the degree of obesity, leptin, adiponectin, and the leptin/adiponectin (L/A) ratio were associated with insulin resistance and other cardiometabolic comorbidities (hyperglycemia and dyslipidemia), but the L/A ratio exhibited stronger associations than the respective adipokines (28).

 

Genetic analysis of single nucleotide polymorphisms (SNP) in the adiponectin locus have identified in humans a haplotype that, in presence of reduced adiponectin and obesity might alter metabolic profile posing risk towards type 2 diabetes. Presence of +10211T/G and +276G/T SNP are associated with increased fasting plasma glucose, body mass index (BMI), and hypertriglyceridemia (29). Recently, adiponectin was found to enhance exosome biogenesis and secretion, leading to a decrease in cellular ceramides, the excess of which is known to cause insulin resistance and cardiovascular disease phenotypes (30). Adiponectin has been shown to reduce the action of inflammatory cytokines such as tumor necrosis factor alpha (TNF-alpha) (31), favorably modulate natural killer cell function (32) and other immune regulatory molecules (33), and improve dyslipidemia (34) and other risk factors of cardiovascular disease (31).

 

In addition to an anti-atherogenic effect, adiponectin may also have a variety of anti-tumor effects. This is thought to be mediated, in part, through inhibition of leptin-induced tumor proliferation (35). It retards the aggressiveness of tumors and their metastatic potential. By cancer site and type, high adiponectin levels are associated with a decreased risk of breast, colorectal, and endometrial cancer (22), whereas hypoadiponectinemia has been associated with increased risk for breast, gastric, lung, and prostate cancers (36-39).

 

A recent study also linked maintenance of the balance between adiponectin and leptin levels with cellular changes in human milk that enhances the protection and decreases the indices of neonatal infection in the breastfeeding infants of women with high BMI values (40).

 

Chemerin

 

Chemerin, also known as Retinoic Acid Receptor Responder Protein 2, is a newly discovered adipokine secreted from mature adipocytes thought to play an important role in the regulation of adipogenesis as well as macrophage infiltration into adipose tissue (41,42). Overexpression of chemerin in people with obesity correlates with early vascular damage, as chemerin was demonstrated to be a better predictor of intima-media thickening than waist circumference and glycated hemoglobin. Weight loss is associated with a decrease of chemerin level and, like adiponectin, an improvement of all parameters of the metabolic syndrome (43). Also, a decrease of  chemerin is independently associated with the reduction of carotid intima-media thickening and the improvement of insulin sensitivity (44).

 

Omentin

 

Omentin is an adipokine preferentially produced by visceral adipose tissue that exerts insulin-sensitizing actions (45). Its expression is reduced in obesity, insulin resistance, and type 2 diabetes. Omentin is also positively related with adiponectin and high-density lipoprotein levels, and negatively associated with body mass index, waist circumference, insulin resistance, triglyceride, and leptin levels (46) (47). Apart from obesity, hyperandrogenism and PCOS per-se seem to have an additional role in omentin levels since omentin-1 was lower in girls with obesity, PCOS, and hyperandrogenism compared to girls with obesity but not PCOS (48).

 

Omentin has anti-inflammatory, anti-atherogenic, anti-cardiovascular disease, and anti-diabetic properties (46). Regarding its effects in the cardiovascular system, omentin causes vasodilatation of blood vessels and mitigates C-reactive protein-induced angiogenesis. The ability of omentin to reduce insulin resistance in conjunction with its anti-inflammatory and anti-atherogenic properties makes it a promising therapeutic/diagnostic target (49).

 

Omentin levels are not significantly different during pregnancy in mothers with diabetes compared to controls. However, significantly lower levels were observed in offspring of the mothers with diabetes, suggesting an increased risk for the development of insulin resistance in later life (50).

 

Retinol Binding Protein-4

 

Retinol binding protein-4 (RBP-4) belongs to the lipocalin family that transports small hydrophobic molecules and is produced primarily in the liver and mature adipocytes (51). Although the relationship between serum RBP-4 and obesity in humans has not been confirmed yet in population studies, several studies have shown positive correlations between the expression of RBP-4 and BMI and glucose concentration (52). RBP-4 levels can be reduced by weight loss, consuming a balanced diet, and exercise in association with increased insulin sensitivity (53,54).

 

Visceral Adipose Tissue-Derived Serpin: Serpin A12 (Vaspin)

 

Vaspin is a serine protease inhibitor produced by subcutaneous and visceral adipose tissue. Vaspin is also expressed in the skin, hypothalamus, pancreatic islets, and stomach. Vaspin is considered as an anti-atherogenic insulin-sensitizing factor (55).

 

Fatty Acid-Binding Proteins

 

Fatty acid binding protein A (A-FABP) is an isoform expressed in the adipose tissue and macrophages (56). It binds to hydrophobic ligands such as long chain fatty acids and facilitates their transport to specific cell compartments. Several studies have shown positive correlations between A-FABP and proinflammatory factors, such as CRP, and may also have significant importance in predicting insulin resistance (57).

 

Acylation Stimulating Protein

 

Acylation stimulating protein is synthesized and secreted by adipocytes and plays a major role in fatty acid uptake and triglyceride synthesis in these same cells, including postprandial clearance of triglycerides (58). It has been shown to induce glucose-stimulated insulin release from pancreatic beta cells, modulates cytokine synthesis by mononuclear cells, as well as inhibit cytotoxicity of natural killer cells (59).

 

Renin-Angiotensin-Aldosterone System

 

Several components of the renin-angiotensin system (renin, angiotensinogen, angiotensin-converting enzyme, and angiotensin 2 receptors) are expressed by adipose tissue (60). Recent studies have shown that adipocyte deficiency of angiotensinogen prevents obesity-induced hypertension in male mice (61). Adipocytes promote obesity-induced increases in systolic blood pressure in male high fat-fed C57BL/6 mice via angiotensin 2 dependent mechanism (62). Adipocyte angiotensinogen deficiency prevents high fat-induced elevations in plasma angiotensin 2 concentrations and therefore in systolic blood pressure (61). These results suggest that adipose tissue serves as a major source of angiotensin 2 in the development of obesity-related hypertension.

 

Other Factors Secreted by Adipose Tissue

 

Other proteins secreted by adipose tissue include plasminogen activator inhibitor-1 (PAI-1) (63) as well as complement factors adipsin, apelin, and pten, which may have roles in the pathophysiology or the progression of coronary artery disease and type 2 diabetes (64,65).

Circulating levels of Interleukin-6 (IL-6) are significantly higher in patients with overweight and obesity (66). Interleukin-6 is released by macrophages and T-cells in the adipose tissue (67) and has been implicated in regulating insulin signaling in peripheral tissues by promoting insulin-dependent hepatic glycogen synthesis and glucose uptake in adipocytes (68). Recent studies show that IL-6 deficient mice develop late-onset obesity as well as disturbed glucose metabolism (69). The mechanisms underlying the effect of IL-6 on body fat and metabolism are not completely understood. However, IL-6 may exert central effects to decrease fat mass because of increased energy expenditure. Administration of IL-6 to the CNS has, for instance, been shown to induce energy expenditure and reduce fat mass more effectively than peripheral treatment (69). It has been suggested that IL-6 potentiates the action of leptin providing a possible mechanism for its anti-obesity effect (70). In addition, IL-6 has been postulated to play an etiologic role in the increased risk of thromboembolism observed in patients with obesity (71).

 

Summary

 

Adipose tissue is an extremely active organ with multiple roles, including endocrine, paracrine, and autocrine, in human physiology and disease. How these roles are performed and their contribution to the health or risk of disease will likely be elucidated as more discoveries continue to shed light on the mechanism of the complex interaction between adipocytes and other body tissues.

 

OBESITY AND HYPOTHALAMIC-PITUITARY AXES

 

Obesity and Sex Hormones

 

Not only is obesity associated with alterations in sex hormone levels but sex hormones may conversely influence expression of different obesity phenotypes. One of the best examples of this is the relationship between obesity and androgen levels in men and women and the roles played by sex hormone-binding globulin (SHBG) and gonadotropins (72-74).

 

SEX STEROID AND SHBG

 

Most circulating testosterone and estrogen are bound to proteins, SHBG and albumin. Although a portion of the bound sex hormones may be available for use by the body target cells, only about 2% of circulating sex steroids are unbound, or free, and constitute the bioactive fraction of these hormones. Total hormone levels, therefore, reflect the bound and unbound hormone and are greatly dependent on the serum concentration of SHBG. For example, SHGB levels increase with age and bioactive testosterone levels decrease (Table 1).

 

Table 1. Common Conditions and Medications that Affect Serum Concentrations of SHBG

Increased SHBG

Decreased SHBG

Older Age

Cirrhosis

Hyperthyroidism
Estrogens

Obesity Androgens
Hypothyroidism
Glucocorticoids
Growth hormone
Insulin

 

OBESITY AND ANDROGENS IN MEN     

 

Testosterone should be measured in the morning when serum concentrations peak and we recommend repeating an abnormal measurement for confirmation. Evidence indicates that testosterone (T) deficiency in men induces adiposity and, at the same time, increased adiposity induces hypogonadism (72). An obesity-associated decline in SHBG might partially explain the observed fall in T levels (74,75). However, an increased BMI is associated with low measured, or calculated, free- and bioavailable-testosterone levels as well. In a metanalysis of sixty-eight studies including a total of 19,996 patients with obesity, prevalence of hypogonadism ranged from 22.9 to 78.8% and from 0 to 51.5% depending on whether low total testosterone or low free testosterone was used to define hypogonadism, respectively. Pooled prevalence of hypogonadism when measuring total testosterone or free testosterone was 42.8% and 32.7%, respectively (76).

 

While the specific pathogenic mechanisms linking obesity with low testosterone levels are not completely understood, both secondary (hypogonadotropic) and, to a minor degree, primary hypogonadism (testicular failure) have been described. Other potentially contributing factors include development of type 2 diabetes, hypertension, and increased adipokines (77,78). Obstructive sleep apnea predisposes to male obesity and secondary hypogonadism (MOSH) through reductions in luteinizing hormone (LH) pulse amplitude and reduced mean serum levels of LH and T in men. Obstructive sleep apnea may also disrupt the association between a rise in serum T levels and the appearance of first REM sleep (79,80).

 

At the testicular level, studies by Wagner et al have shown that obesity lowers the number of testosterone producing Leydig cells and promotes destruction of existing ones by increasing levels of proinflammatory cytokines (TNF alpha) and cells (macrophages) (81). In both the short and long term, obesity was shown to lower intra testicular levels of testosterone by way of increasing serum leptin and estradiol levels and inhibiting the expression of the gene for cytochrome p450 of the cholesterol side chain cleavage enzyme (Cyp11a1) (81).

 

Whether testosterone treatment in (MOSH) is beneficial has long been controversial. Only those with low free T levels and signs or symptoms of hypogonadism should be considered androgen deficient. Considering the limited number of rigorous testosterone therapy trials that have shown beneficial effects, the modest amplitude of these effects, and unresolved safety issues, testosterone therapy is currently not advocated in the prevention or reversal of obesity-associated metabolic disturbances (82).

 

On the other hand, true hypogonadism in men can promote increased fat mass, which in turn may worsen the hypogonadal state. Low testosterone levels lead to a reduction in muscle mass and an increase in adipose tissue within abdominal depots, especially visceral adipose tissue (VAT) that can be reversed with testosterone therapy (83,84). As adiposity increases, there is a further raise in aromatase activity that is associated with an even greater conversion of T to estradiol (often termed the 'testosterone-estradiol shunt'), which is thought to decreased GnRH secretion (85). This further decreases T levels that in turn further increases the preferential deposition of fat within abdominal depots: a 'hypogonadal-obesity cycle' (86,87). Individuals with obesity retain the capacity to reverse this gonadotrophic response with weight loss, demonstrating that MOSH is a reversible condition. This has been made evident on several studies in which weight loss normalized T levels (88,89).

 

In summary, obesity is frequently associated with low androgen levels in men and true hypogonadism can worsen adiposity and central fat deposition. The pathogenesis of obesity-related hypogonadism is complex and multifactorial, implicating obesity-related comorbidities and changes in body fat mass itself with its multiple adipokines and inflammatory mediators. Ultimately, these changes are frequently reversible with weight loss and preferred strategies to manage these conditions target lifestyle, anti-obesity medications, and weight-loss surgeries when indicated.

 

OBESITY AND SEX STEROIDS IN WOMEN

 

Increases in body weight and fat tissue are associated with abnormalities of sex steroid levels in both premenopausal and postmenopausal women. It has been shown that women with central obesity have higher circulating androgen levels, even in the absence of a clinical diagnosis of polycystic ovarian syndrome (PCOS) (90,91). These women have higher total and free testosterone levels than normal-weight woman and lower androstenedione and SHBG levels (91). Some studies examining the co-relationships between the total testosterone levels and phenotypic features of hyperandrogenism, such as hirsutism, found a strong correlation between them, regardless of the assay used for assessment (92).

 

The timing of menarche is primarily thought to be affected primarily by genetic factors (93,94), but the average age at menarche in US girls has been declining over the past 30 years (95) in conjunction with changes in nutritional status (96). A Mendelian randomization study from the United Kingdom linked a higher BMI with early menarche, suggesting a causal relationship between increasing prevalence of childhood obesity and similar trends in the prevalence of early menarche (97).

 

Studies have also shown that the earlier the onset of menarche, the higher the risk of developing obesity (98) and other comorbidities in the adult life, independently of BMI, such as: breast cancer, cardiovascular disease, cerebrovascular disease, type 2 diabetes, and adolescence at-risk behaviors (99-104). Consequently, all-cause mortality has been linked with early menarche (105). Also, there is evidence that menarche at or before 12 years of age is associated with higher androgens levels even during adulthood, suggesting that hyperandrogenemia may explain, at least in part, the higher incidence of comorbidities among these women. A recent study demonstrated that with each one-year advance in menarcheal age, the probability of having obesity decreased by 22%; interestingly, in this study women with obesity had higher androgens levels (106).

 

Menarche age also appears to affect offspring. Boys whose mothers with menarche onset ≤13 years at menarche had an adjusted relative risk of obesity 3-fold greater than sons of mothers with a later menarche onset. The increased obesity risk was not observed in daughters. However, girls who experienced menarche earlier had a less favorable anthropometric profile consisting in a reduced waist and hip circumferences and waist-to-height ratio (107). Early menarche, therefore, has emerged as a risk of later obesity and related medical problems.

 

RELATIONSHIP BETWEEN LEPTIN AND SEX HORMONES

 

Leptin participates in the regulation of hypothalamus-pituitary-gonadal (HPG) axis at multiple levels. Leptin appears to facilitate GnRH secretion indirectly by modulating several interneuron secretory neuropeptides (108,109) and directly by stimulating LH and, to a lesser extent, FSH release.

 

Leptin has a permissive role in timing puberty but is not essential nor is the only trigger for puberty onset, as has been shown in studies (110) of patients with leptin deficiency and several animal studies (111,112).

 

Kisspeptin play a central role in the modulation of GnRH pulse generator and, thus, downstream regulation of gonadotropins and testosterone secretion in men (113,114). Kisspeptins are mostly distributed in the hypothalamus, dentate gyrus and adrenal cortex. Inactivating mutations of the kisspeptin receptor have been shown to cause hypogonadotropic hypogonadism in men, while an activating mutation is associated with precocious puberty. Data from studies in animals link kisspeptin expression with hyperglycemia, inflammation, leptin and estrogen, factors known to regulate GnRH secretion. It has been hypothesized that decreased endogenous kisspeptin secretion is the common central pathway that links metabolic and endocrine factors in the pathology of T deficiency observed in MOSH and type 2 diabetes (113).

 

Serum kisspeptin levels are higher in patients with obesity and tend to decrease after weight loss intervention. (115,116). Also, data suggest a higher concentration of serum kisspeptin in women with PCOS irrespective of their BMI but further data are needed to ascertain the role of kisspeptin in PCOS (116).

 

Kiss1 neurons appear to transmit the regulatory actions of metabolic cues on pubertal maturation. Recently, it has been documented that AMPK and SIRT1 operate as major molecular effectors for the metabolic control of Kiss1 neurons and, thereby, puberty onset. Alterations of these molecular pathways may contribute to the perturbation of pubertal timing linked to conditions of metabolic stress in humans, such as undernutrition and obesity. As such, it has the potential of becoming a druggable targets for better management of pubertal disorders (117).

 

Leptin receptors are also widely expressed in the human ovaries (118) and testes (119) indicating a direct gonadal regulatory role. Studies by Ma et al. have shown that high-fat diet fed mice produce fewer oocytes compared with control mice receiving a normal diet. Leptin has been noted to act locally within the mice ovarian granulosa cells to reduce estradiol production (120). These actions are mediated via induction of the neuropeptide cocaine- and amphetamine-regulated transcript (CART) in the granulosa cells (GCs), which in turn detrimentally affects intermediate steps of estradiol synthesis including, intracellular cAMP levels, MAPK signaling, and aromatase mRNA expression (121). In humans undergoing in vitro fertilization, Ma et al. demonstrated that subjects with higher BMI had higher levels of CART mRNA and peptide in follicular fluid (121). Therefore, in women with obesity, evidence supports a role for leptin as a mediator of infertility at the level of the ovary.

 

As mentioned above, in men with obesity, intra testicular levels of testosterone are lower due to leptin and estradiol inhibition of the expression of the gene for cytochrome p450 of the cholesterol side chain cleavage enzyme (Cyp11a1) (81). Gregoraszczuk et al exposed porcine ovarian follicles obtained from prepubertal and mature animals to progressively increasing doses of super active human leptin antagonist (SHLA) and measured levels of leptin receptor (ObR), leptin, CYP11A1 and 17β-hydroxysteroid dehydrogenase (17β-HSD), progesterone (P4), and testosterone (T) in the follicles (122). These experiments showed that SHLA inhibits CYP11A and 17 beta protein expression, subsequently inhibiting leptin, ObR, and hence leptin-mediated follicular P4 and T secretion. Women with obesity and polycystic ovarian syndrome (PCOS), a condition associated with elevated androgen levels and infertility (see also below), were found to have higher levels of leptin (both bound and free form) and lower levels of s-OBR (soluble Leptin receptors) when compared to lean females with PCOS, after adjusting both groups for age, in studies by Rizk, who hypothesized that lower s-OBR may have been in response to impaired leptin function (123).

 

Leptin and its soluble receptor are thus implicated in the pathophysiology of PCOS, may act as a mediator of infertility at the level of the ovary and testes, and that leptin antagonists acting peripherally in gonadal tissues may thus be useful in modifying the physiology of reproduction.

 

OBESITY AND POLYCYSTIC OVARIAN SYNDROME

 

Polycystic ovarian syndrome is a highly prevalent condition of hyperandrogenism frequently associated with obesity. Hence, this disorder has been studied extensively in the context of interactions between sex hormones and obesity. It affects approximately 6-10% of women in reproductive age (124). About two thirds of women with PCOS are obese and 50-70% of them have insulin resistance (IR) (125).

 

Adult men have more visceral fat than premenopausal women, in which the body fat is more prominent in the periphery and subcutaneous adipose tissue. This sexual dimorphism is mainly related to the differential effects of androgens and estrogens on adipose tissue (126). Visceral adipose tissue (VAT) excess is strongly associated with metabolic disorders such as insulin resistance and dyslipidemia (127). Women with PCOS manifest what has been called "masculinization of the adipose tissue" characterized by increased VAT and even male pattern adipokine gene expression with its associated metabolic complications (128,129). Even though increased VAT plays a significant role in the development of insulin resistance in PCOS, it has been suggested that insulin resistance may represent an intrinsic characteristic of this syndrome, independent of obesity (124). Interestingly, in PCOS, despite the insulin resistance in other organs, the ovaries remain sensitive to the stimulatory effect of insulin on androgen production (130). A recent study showed that despite women with PCOS and women with the metabolic syndrome sharing many features, these are different entities, mainly due to the excess of androgens seen in PCOS, which seems the be the main culprit of its multiple co-morbidities (131) .

 

Anovulation and menstrual irregularities are major features of PCOS in part due to ovarian hyperandrogenism, hyperinsulinemia due to IR, and altered paracrine signaling within the ovary, which can disrupt follicle growth (124). Hyperinsulinemia also decreases hepatic SHBG with a subsequent increase in free androgens levels. In addition, insulin increases the androgens synthesis stimulated by LH and IGF-1.

 

An increased ratio of serum LH to FSH may be seen in about 70% of women with PCOS (132,133). The androgen excess reduces the negative feedback in the hypothalamus causing an enhanced pulsatile release of gonadotropin releasing-hormone (GnRH) which will elevate LH levels and pulse frequency (134).

 

In summary, obesity is a common feature of PCOS and hyperinsulinemia secondary to insulin resistance of the liver and muscle is believed to be the main etiological factor behind the development of PCOS. Obesity also leads to hyperestrogenism. Weight loss and/or use of insulin sensitizing agents (mainly metformin) improve insulin sensitivity, reduce insulin levels, and improve fertility in women with PCOS but not live births (135,136). Therefore, the role of metformin in improving reproductive outcomes in women with PCOS appears to be limited (137). Letrozole, an aromatase inhibitor, is headed toward replacing clomiphene, a selective estrogen receptor modulator. As the first-choice option for ovulation induction, metabolic treatments such as metformin, troglitazone, or d-chiro-inositol have failed to show promise in improving fertility outcomes. Further studies are needed of the newer agents to treat type 2 diabetes (138) .

 

A clinical trial in 120 infertile PCOS women showed that when metformin is combined to myoinositol (MI) a significant improvement in live birth rate, menstrual cycle (length and bleeding days), and HOMA index is observed compared to use of metformin alone (139). Treatment with MI has been useful also in in-vitro fertilization (IVF), as it allows a decrease in the amount of recombinant FSH administered, in the duration of the ovulation induction for follicular development (140,141) and an increase in the clinical pregnancy rate (142) [45].

 

OBESITY AND ESTROGENS

  

Estrogens play an important role in body weight, fat distribution, energy expenditure, and metabolism. In healthy premenopausal women, estrogens are mainly synthesized in the ovaries under the regulation of gonadotropins releasing hormones from the pituitary gland. They are also produced in the adipocytes via aromatization from androgenic precursors, which is especially important in men and post-menopausal women and increase in proportion to the total body adiposity (143,144).

 

Most metabolic effects of estrogens are mediated through estrogen receptor (ER) alpha, whereas most gynecologic actions are exerted through ER beta. Mice of both sexes with a targeted deletion of the ER alpha gene manifest obesity-induced insulin resistance with altered plasma adipokines and cytokines levels and increased adiposity, mainly VAT (145,146).

 

Estrogens have a positive effect in glucose homeostasis, acting as an insulin sensitizer at multiple levels, including skeletal muscle, liver. and adipocytes (147). Estrogen effects the immune system to decrease inflammation, thus favoring insulin sensitivity (148,149). Pancreatic islet-cells also have estrogens receptors, which when activated improve beta cell function and survival (150). Estrogen deficiency promotes metabolic dysfunction predisposing to obesity, metabolic syndrome, and type 2 diabetes.

 

In rodent models, estrogen has been shown to influence energy intake and energy expenditure via hypothalamic signaling. Estrogen receptor alpha is widely expressed in the ventromedial hypothalamus (VMH), area of the brain that controls food intake, energy and body weight homeostasis. In animal models, the lack of ER alpha in the VMH causes dramatic changes in energy balance leading to increased adiposity (147).

 

The gynecoid body fat distribution, characterized by increased fat depots into the subcutaneous tissue favoring gluteal/femoral areas and decreased VAT is mediated mainly by estrogens (147). Visceral fat is augmented in hypoestrogenic states, as seen in menopause. These changes in body fat composition can be prevented by estrogens replacement (151). Also, estrogen treatment of male-to-female transsexuals significantly increases fat deposition in all subcutaneous fat depots, while having little effect on the visceral fat compartment (152).

 

Obesity in both men and women is associated with elevated estrogens levels that result from aromatization of androgens in adipocytes (86). Increased adiposity is a known risk factor for the development and progression of breast cancer and this hyperestrogenic state is associated with increased risk of cancer (153), while weight loss improves prognosis of patients diagnosed with breast cancer and the reduction in estrogens levels may be, at least in part, responsible for this finding (154).

 

Obesity and Growth Hormone

 

Growth hormone (GH) is secreted by the pituitary gland. Most of GH-promoting effects are mediated by Insulin- like Growth Factor-1 (IGF-1), but GH also has effects independent of IGF-1. Serum IGF-1 concentrations represent the most accurate reflection of growth hormone biologic activity. The liver is the major, but not exclusive, source of IGF-1. About 50% of circulating growth hormone is bound to binding proteins. These include a high affinity Growth Hormone Binding Protein (GHBP), which represents the extracellular portion of the GH receptor. IGFs are mostly bound to IGF- Binding Proteins (IGFBPs) with IGF-1 is bound to IGFBP3.

 

Together GH and IGF-1 influence lipids, protein, and glucose metabolism so as to inhibit fat accumulation, promote protein accretion, and alter energy expenditure and body fat/muscle composition. Normally, GH secretion is suppressed as insulin increases in the postprandial period, which permits skeletal muscle glucose uptake promoting glycogenesis and adipogenesis (155). The opposite changes in hormonal concentrations occur during fasting to facilitate lipolysis and hepatic glucose output (156).

 

GH secretion from the anterior pituitary is modulated by the hypothalamic GH releasing hormone (GHRH) and follows a pulsatile pattern that is influenced by age, sex, sleep, feeding, physical activity and weight (157). Obesity is typically accompanied by a decrease in GH levels and increase in GHBP levels. This is the opposite picture to starvation in which GH levels are increased and GHBP levels decreased. An inverse relation exists between GH levels and BMI and percent fat mass, particularly VAT, independently of age or sex (158,159). The reduction in GH levels in obesity is multifactorial and it involves a decreased pituitary release of GH (decreased frequency of GH secretory bursts proportionate to the decree of obesity) and an accelerated GH metabolic clearance rate (160).

 

Since GH has lipolytic and anabolic properties, it has been postulated that the decline of GH seen in elderly and individuals with obesity may be partly responsible for the progression of metabolic diseases (161). GH is known to induce insulin resistance (IR). The increased IR seen during puberty and gestational diabetes is, in part, attributed to increased GH action (162). One of the clinical manifestations of acromegaly is glucose intolerance and diabetes mellitus. But interestingly, GH deficiency can also be accompanied by increased IR. A recent general population study in Danish adults revealed that both low and high-normal IGF-1 levels are related to IR (163). There are striking similarities between the metabolic syndrome and untreated adult-onset GH deficiency: increased VAT, IR, non-alcoholic fatty liver disease, dyslipidemia and the associated increased risk of premature atherosclerosis and cardiovascular disease (164,165). All these observations have led to an increasing interest in investigating the mechanisms behind the decline of GH seen in obesity since it may have important clinical and therapeutic implications. Weight loss is associated with improved stimulated GH response. However there is uncertainty on how much weight loss is required to completely normalize GH secretion (166).

 

Despite the reduced GH levels seen in obesity, IGF-1 serum levels are not significantly different between those with and without obesity. Studies have reveled mostly normal or slightly low IGF-1 serum levels in individuals with obesity (159,167,168). This suggests that lower levels of GH are accompanied by increased peripheral sensitivity to GH accounting for the relatively normal IGF-1 levels. This is supported by data from Maccario et al., who found that the administration of a low dose of rhGH had an enhanced stimulatory effect on IGF-1 secretion in subjects with obesity compared to normal weight subjects (169). In another study, the same authors showed a normal feedback inhibitory response of the somatotroph to IGF-1 (170). In addition, decreased GH levels result in up-regulation of GH receptors and increased sensitivity at the liver, as it was shown by higher IGF-1 response to a single GH bolus in subjects with obesity as compared with normal weight individuals (171).

 

PROPOSED MECHANISMS FOR LOWER GH SECRETION IN OBESITY

 

Hyperinsulinemia that accompanies obesity could be one of the stronger inhibitors of GH secretion by peripheral and central actions. Insulin produces increased peripheral sensitivity to GH, reduced IGFBP-1 levels and increased IGF-1 in spite of decreased GH secretion by the somatotroph. High free IGF-1 levels in this case exert a negative feedback mechanism on GH secretion. Central effects of insulin were shown in a study where the peak GH secretion after GHRH stimulation was inversely associated with fasting insulin in premenopausal women with obesity (172).

 

Sex steroid levels may also govern GH activity. It has been shown that testosterone activates the somatotrophic axis in men (173,174) and augments the GH-dependent stimulatory effect on IGF-I production, enhancing protein and energy metabolism (175). Estrogens, in contrast, cause GH resistance in the liver, leading to a relative reduction of IGF-I production per unit of GH secretion (176).

 

Other possible mechanisms for the altered GH response in obesity are free fatty acids (FFA) and leptin, both of which are increased in obesity. Lee et al showed that reduction in free fatty acids concentrations in subjects with obesity through use of Acipimox leads to increased GH response to GH-releasing hormone (177). In animals, leptin has an inhibitory role on GH secretion from the pituitary gland through its effects on GHRH and neuropeptide Y (NPY) at the hypothalamus level (178).

 

RECOMBINANT GROWTH HORMONE THERAPY IN PATIENTS WITH OBESITY         

 

The use of recombinant human growth hormone (rhGH) in elderly and subjects with visceral obesity results in several mild to moderate anthropometric and metabolic effects such as reduced fat mass, increased lean mass, and improved surrogate markers of cardiovascular disease (179). Recombinant growth hormone has been extensively studied as a treatment for obesity. A meta-analysis found that rhGH therapy reduces visceral adiposity and increases lean body mass as well as having beneficial changes in lipid profile in adults with obesity, but without inducing significant weight loss. In fact, the observed reductions in abdominal fat mass are modest and similar to what can be achieved by life style interventions (180). In addition, administration of rhGH was associated with increases in fasting plasma glucose and insulinemia over shorted periods of time (181). However, the dose of rhGH used in these studies was supraphysiological.

 

Investigations of rhGH in youth have reported favorable outcomes. A pilot study in young adults (18-29 years old) with obesity and non-alcoholic fatty liver disease suggested that rhGH may have benefits to reduce liver fat content (182). Also, in boys with obesity (8-18 years old) treatment with rhGH for one-year reduced body mass index standard deviation scores and insulin-like growth factor 1 levels increased. GH treatment also reduced low density lipoprotein cholesterol, total cholesterol, triglycerides, and alanine aminotransferase when compared with the baseline. (183). However, further studies of longer duration outcomes, including cardiovascular morbidity and insulin sensitivity, are warranted.

 

In conclusion, obesity is accompanied by a reduction in basal and stimulated GH secretion by the pituitary gland. The reduction in GH does not appear to translate into similar reduction in IGF-1. While some benefits of GH treatment in obesity are seen in body composition, other than in those individuals with documented GH deficiency, these are probably not enough (or greater than was is seen with lifestyle) to outweigh potential long-term side effects and the role of GH replacement in patients with obesity and normal GH axis testing, remains controversial.

 

Obesity and Adrenal Glands

 

Cortisol circulates in the bloodstream mainly bound to Cortisol-Binding Globulin (CGB or transcortin) and less to albumin. About 10% of cortisol is free or unbound and this fraction represents the bioactive portion of the hormone. CBG concentrations can be increased or decreased in several conditions and by some medications (Table 2), thus affecting total cortisol levels in these situations.

 

Table 2. Medical Conditions and Drugs that Affect Cortisol Binding Globulin (CBG) and Total Cortisol Levels

Increase CBG

Decrease CBG

Estrogens
Pregnancy
Oral contraceptives
Diabetes mellitus
Hyperthyroidism

Obesity
Cirrhosis
Testosterone
Nephrotic syndrome
Hypothyroidism

 

The dynamics of the hypothalamic-pituitary-adrenal (HPA) axis in obesity have been examined. Patients with Cushing's syndrome display several clinical features that resemble those seen in patients with the metabolic syndrome. These features include redistribution of adipose tissue from peripheral to the truncal region increasing VAT, insulin resistance, impaired glucose homeostasis, hypertension, and lipid abnormalities. These similarities led to the hypothesis that a dysregulation of the HPA axis in the form of "functional hypercortisolism" could potentially be a cause for abdominal obesity and its accompanying metabolic consequences (184).

 

The serum concentrations of cortisol are generally normal in obesity (185-188). Salivary cortisol and 24-hour urine free cortisol (UFC) excretion are usually high-normal or sometimes mildly elevated in obesity. A cross-sectional study of subjects with obesity showed a trend to increase salivary cortisol as BMI increased, but the same association was not found with UFC (189). Other studies in which UFC has been shown to be increased in obesity are due to enhanced cortisol clearance (188,190), with maintenance of normal cortisol levels and circadian appearance in those with obesity through subsequent increases in cortisol production rates (188,190,191).

 

It has been demonstrated that high-normal ACTH and cortisol levels in individuals with obesity are associated with cardiovascular risk factors, such as hypertension, insulin resistance and dyslipidemia (192,193). On the other hand, depression and/or alcoholism may slightly increase cortisol levels. These conditions have been described as pseudo-Cushing's syndrome (194). A pseudo-Cushing's state is characterized by clinical and biochemical features that resemble true Cushing's syndrome but with resolution of the signs and symptoms once the underlying primary condition is eliminated. It is thought that these primary conditions may stimulate CRH release with subsequent activation of the entire HPA axis (195,196).

 

Although serum cortisol is not increased in obesity, it is possible that the local production of cortisol in the fat tissue is increased and this, in turn, could lead to increased local action of cortisol with the subsequent metabolic consequences. Adipose tissue is involved in the metabolism of cortisol through action of the enzyme 11 Beta-hydroxysteroid dehydrogenase-1 (11HSD1), which converts cortisone (inactive corticoid) to cortisol (active corticoid) (197). Whole body 11β-HSD1 reductase activity tends to be higher in obesity (~10%) and is further increased by insulin (198). It appears that in obesity, more cortisol is derived from cortisone due to the increased activity of this hormone, which could simply be due to increased visceral fat mass. (198).

 

Some authors provide evidence that cortisol affects zinc metabolism and indicate possible repercussions on insulin signaling that might contribute to the development of resistance to the actions of insulin in obesity. Thus, alterations in the biochemical parameters of zinc observed in individuals who are obese contribute to the development of disorders in the synthesis, secretion, and action of insulin  (199).

 

Visceral adipose tissue has higher numbers of glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) than subcutaneous tissue (200,201). Glucocorticoids have higher affinity to MR than to GR. It has been shown that MR activation mediates inflammation and dysregulation of adipokines causing insulin resistance and acceleration of the development of metabolic disorder (202). Interestingly, blockade of the MR improves these outcomes (203,204). In human adipose tissue, MR mRNA levels increase in direct association with BMI and this augmentation is more significant in VAT, whereas GR mRNA levels had no apparent correlation with BMI or fat distribution (201). Even though evidence for an increased cortisol concentration within the VAT in human obesity is "possible, but unlikely" (205), it is not surprising that inhibition of 11HSD1 and MR has become a major therapeutic target in metabolic syndrome (206,207).

 

The cortisol response to a variety of stimuli such as ACTH, CRH, or meal ingestion is altered in obesity and by sex. Animal studies showed that estrogens sensitize and androgens diminish corticotropic-response to ACTH (208). In obesity these sex hormone differences are blunted. One study showed decreased ACTH potency with higher BMI in men (208) and other studies demonstrated ACTH secretion rates comparatively higher than the cortisol secretion rate in centrally obese premenopausal women; suggesting decreased responsiveness of the adrenal gland to the ACTH stimulation in these subjects (209,210). The same authors showed in a more recent publication, that premenopausal women exhibit diminished ACTH efficacy (maximal cortisol response) and sensitivity (slope of the dose-response curve) (211). This pattern is similar to what has been described in Cushing's syndrome (212). Of note, it is important to mention that older studies have revealed increased responsiveness of adrenal glands to exogenous ACTH pharmacologic stimulation (213), but this finding should not be extrapolated to the effects of endogenous ACTH stimulation.

 

A decrease in the mineralocorticoid receptor (MR) response to circulating corticosteroids was suggested by Jessop et al as an explanation for the relative insensitivity to glucocorticoid feedback in obesity (214). A more recent study showed that MR represent an important pro-adipogenic transcription factor that may mediate both aldosterone and glucocorticoid effects on adipose tissue development. Mineralocorticoid receptor thus may be of pathophysiological relevance to the development of obesity and the metabolic syndrome (215).

 

The HPA axis is also activated in response to stress along with the sympathetic nervous system, and the sympathoadrenal system. Whether stress-related obesity due to excess and/or dysfunction of cortisol activity is a distinct medical entity remains unclear and there are contradicting findings in the literature. This topic is evidently difficult to investigate due to multiple confounding variables and therefore well-defined longitudinal studies are needed (216).

 

Finally, when screening overweight and individuals with obesity for Cushing's syndrome it is imperative to follow the Endocrine Society guidelines which recommend diagnosing the disorder only if two screening tests are abnormal (196). A study of 369 overweight or subjects with obesity with at least two features of Cushing's syndrome found that 25% of these subjects had an abnormal screening test result, but none of them had two positive tests, hence none was found to have Cushing's syndrome (217).

 

In conclusion, obesity is associated with alterations in the HPA axis that may be a manifestation of a causative effect, adaptive changes to a new homeostatic state or, most likely, a combination of both. And although signs and symptoms of hypercortisolism commonly are also found in patients with central obesity, the finding of an actual case of Cushings disease is very rare in the obese population.

 

Obesity and the Thyroid

 

More than 99% of T4 and T3 circulate bound to transport proteins. Only a very small amount, less than 1%, of thyroid hormone is unbound or free and represents the biologically active fraction of the hormone. Thyroxine Binding Globulin (TBG) is the major transport protein for thyroid hormones and serum TBG concentrations are influenced by several conditions and medications, which result in altered total T4 and T3 concentrations (Table 3). Therefore, when evaluating thyroid function, we measure thyroid stimulating hormone (TSH) and free T4 (FT4). Free T3 (FT3) can also be measured in selected circumstances, such as hyperthyroidism, although it represents only a small fraction of circulation total thyroid hormone activity.

 

Table 3. Medical Conditions and Drugs that Affect Thyroxine Binding Globulin (TBG) and Total Thyroid Hormone Levels

Increase TBG

Decrease TBG

Estrogens
Pregnancy
Hypothyroidism
Acute hepatitis

Androgens
Corticosteroids
Systemic illness
Nephrotic syndrome
Hyperthyroidism
Cirrhosis

 

Thyroid dysfunction is frequently associated with changes in body weight and composition, body temperature, energy expenditure, food intake, glucose, and lipids metabolism. Hypothyroidism is linked to weight gain and decreased metabolic rate but there is also a positive association across the normal range between serum levels of TSH and BMI. Some cross-sectional population studies suggest that even a slightly elevated serum TSH might be important in determining an excess of body weight and it can be considered a risk factor for overweight and obesity (218-221). Also, individuals with obesity have an increased incidence of subclinical and overt hypothyroidism. Some studies showed a prevalence of these conditions in morbid obesity as high as almost 20% (222,223). Thyroid-stimulating hormone concentrations has also been associated with the presence of the metabolic syndrome, even when TSH is within normal levels. In a study of 2,760 euthyroid young woman, those with high-normal TSH (2.6-4.5 mIU/L) had higher prevalence of metabolic syndrome than those with low-normal TSH (0.3-2.5 mIU/L) (224) .

 

However, further investigation is needed to determine whether the relationship between TSH and BMI represents causality (mild thyroid failure leading to obesity) or just adaptive changes (physiologic or pathologic) to a new homeostatic state of increased body weight. Contradicting results from different studies illustrate this controversy. For example, a study published by Marzullo et al. supports the idea that obesity increases susceptibility for thyroid autoimmunity, since in their group of individuals with obesity they found higher rates of positive anti-thyroid peroxidase antibodies than in controls (225). This finding was not observed in other cross-sectional studies that included individuals with severe obesity (BMI > 40 kg/m2). In that study, as compared with controls subjects with severe obesity had higher levels of TSH (but with lower rates of positive thyroid antibodies than control individuals (222,223,226). Data from the NHANES III survey showed no difference in thyroid antibodies positivity among individuals with obesityand the general population (227). However, a recent metanalysis showed that even after stratification, the obese population had increased risks of overt hypothyroidism and subclinical hypothyroidism and was clearly associated with Hashimoto’s thyroiditis but not Graves' disease. In patients with Hashimoto’s thyroiditis, obesity was correlated with positive thyroid peroxidase antibody (TPOAb) levels but not with positive thyroglobulin antibody (TGAb) levels (228).

 

In a euthyroid population, when comparing metabolically healthy obese (MHO) with metabolically unhealthy obese (MUO) phenotypes, the following findings were reported: FT4 levels were negatively associated with the MUO phenotype, FT3 levels were positively associated with both the MHO and the MUO phenotypes, and TSH levels were positively associated with the metabolically unhealthy, non-obese phenotype (229).

 

Also, in population studies higher levels of T3, FT3, T4, and TSH are seen in individuals with obesity, probably the result of the reset of their central thyrostat at higher level (223).

The idea that these thyroid function tests (TFTs) changes may reflect a state of thyroid hormone resistance has also been considered. This is supported by the observation of decreased thyroid hormone receptors in circulating mononuclear cells of individuals with obesity (230) and decreased negative feedback between TSH and peripheral T3 levels.

Fat accumulation increases in parallel with TSH and FT3 levels independently of insulin sensitivity and other metabolic parameters. Also, a positive association has been described between FT3 to FT4 ratio and BMI and waist circumference (231). These findings may result from a high conversion of T4 to T3 due to increased deiodinase activity in the adipose tissue as a compensatory mechanism to increase energy expenditure (220). On the other hand, during a hypocaloric diet, serum T3 declines significantly, generating changes in the cardiovascular system like those seen in hypothyroidism, suggesting that the decline in T3 may be an adaptive response for energy preservation (232,233). This adaptive decline in T3 may be mediated, in part, by the fall in leptin levels that accompanies weight loss as it can be reversed with leptin administration (234). Subcutaneous and visceral fat showed reduced thyroid gene expression in subjects with obesity, especially TSH Receptor gene expression. These changes were reversed by major weight loss (235).

 

After weight loss from bariatric surgery, FT3 and TSH levels were significantly reduced and serum thyroperoxidase antibody (TPOAb) and thyroglobulin antibody (TgAb) levels decreased significantly from 79.3 and 177.1 IU/mL to 57.8 and 66.0 IU/mL, respectively, in participants with positive thyroid antibodies (236). Also in patients starting with a subclinical hypothyroidism state, weight loss leaded to normalization of TSH levels in most patients and none developed overt hypothyroidism (237). Furthermore, in children with obesity and overweight without circulating antithyroid antibodies, BMI reductions uniquely predict reductions in TSH, thyroid volume, and improvement in thyroid structure with an altered parenchymal pattern at thyroid ultrasound (238).

 

Body mass index is directly associated with thyroid volume and the incidence of thyroid nodules. This association appears to be in positive correlation with the degree of insulin resistance (221,239,240). Not only is the incidence of benign thyroid abnormalities increased in obesity, but a higher rate of malignancy has also been reported (241,242). Pathway analysis has identified 1,036 genes associated with thyroid cancer (TC) and 534 regulated by obesity. Five out of the 358 obesity-specific genes, FABP4, CFD, GHR, TNFRSF11B, and LTF, had significantly decreased expression in TC patients (243). Hyperinsulinemia is a common factor found in most studies linking obesity with increased thyroid cancer incidence (244,245). It is not surprising that particularly high percentage of visceral fat mass has a stronger association with thyroid cancer since VAT is highly metabolically active and associated with increased IR. Even though neck circumference as an index of upper-body adiposity, had a positive correlation with thyroid cancer tumor size and lymph node metastasis (246), other studies do not observe any association between obesity and thyroid cancer aggressive features (247,248). Whether obesity increases the risk of thyroid cancer remains controversial as several authors have concluded that obesity is associated with greater risk of thyroid cancer (249,250), while others do not (251).

 

Synthetic thyroid hormones, as well as various other thyroid hormone preparations, have been used as adjunctive measures to induce or facilitate weight loss. A systematic review reported by Kaptein el al (252) recognized 14 randomized controlled trials and prospective observational studies describing the effects of T3 and T4 therapy in comparison with placebo in euthyroid subjects with obesity during caloric deprivation. Most of these studies had a small sample size, ranging from 5 to 12 treated patients. Thyroid hormone treatment resulted in subclinical hyperthyroidism in most patients and there was no consistent effect on weight loss across the studies.

 

Since the action of thyroid hormone varies depending on the activated receptor, selective thyroid receptor agonists have been developed. In brief, thyroid hormones exert their actions through two major receptors: thyroid receptor alpha (TRA), which mainly mediates T4 effects in bone, skeletal muscle, brain and heart, and thyroid receptor beta (TRB) that regulates TRH/TSH secretion and the metabolic effects of T3 in the liver, such as lowering lipids.

Adipose tissue expresses both TRs (253). Selective TRB agonists are promising drugs for treatment of dyslipidemia and obesity without the toxic effects of thyroid hormones analogs on bones or heart in euthyroid patients. This has been tested in animal studies but there are no clinical trials in humans yet (254-256).

 

CONCLUSIONS AND CLINICAL IMPLICATIONS

 

As discussed in the previous sections, several endocrine alterations can be identified in association with obesity (Table 4). In most cases, these alterations are reversible with weight loss and, therefore, appear to be a consequence of obesity. Emphasis has been focused on the hypothalamic-pituitary hormones axes and the possibility that some "subclinical" alterations in these axes may be at the origin of increased adiposity. At this time this hypothesis needs further testing. What is true is that the interaction between the adipose tissue and the body is far more complex than once believed, and the future will certainly provide more decisive data on the precise mechanisms of these interactions and their contribution to the development and/or the maintenance of obesity.

 

Certain endocrine syndromes are known to result in obesity. From the clinical practitioner's perspective it is important to remember these syndromes and to be suspicious should a patient with obesity display one or more of the clinical features seen in these disorders. Hypothyroidism is a common clinical problem and can, of course, occur in patients with obesity and could contribute to the presence of symptoms such as fatigue and inability to concentrate. Hypothyroidism is under-diagnosed in the general population and specifically patients with obesity. Routine screening of patients who present with obesity with a sensitive TSH assay and free T4 is reasonable, although there are no specific guidelines with regards to this. Cushing's syndrome is frequently included in the differential diagnosis of obesity and patients with abdominal obesity have many features in common with patients with authentic Cushing's. However, true Cushing's disease (due to excessive endogenous corticosteroids) is rare. Nevertheless, if there is a reasonable suspicion for this condition, the patient should be screened. Attention should be focused on symptoms and signs that are more specific to Cushing's such as proximal muscle weakness, purple striae, thin and bruised skin, hypokalemia, and osteopenia.

 

Hypogonadism and growth hormone deficiencies are both associated with abdominal obesity. The former is very common and should be kept in mind in males with other symptoms or signs suggestive of androgen deficiency while the latter is usually suspected in the setting of surgery or disease of the hypothalamus-pituitary axis. The treatment of these two conditions can result directly and indirectly (by improving conditioning, muscle strength, and stamina) in weight loss, improved metabolic profile, and improved bone density but is usually reserved for those with true deficiencies, not with low-normal levels.

 

Table 4. Hormonal Changes in Obesity

Adipose tissue as an endocrine organ 

 

Type 2 obesity with leptin resistance  Leptin increases

 

Type 1 obesity with congenital leptin deficiency  Leptin decreases

 

Adiponectin decreases

 

Chemerin increases

 

Omentin decreases

 

Retinol Binding Protein increases

 

Angiotensin 2 increases

 

Plasminogen activator inhibitor-1 (PAI-1) increases

 

Interleukin-6 increases

Obesity and the pituitary axes

 

LH pulsatility decreases

 

Total testosterone decreases in men

 

Free testosterone decreases in men

 

SHBG decreases in women and men

 

Androgens increase in women

 

Free testosterone increases in women

 

Androstenedione decreases in women

 

Increase in kisspeptin levels

 

Aromatization of androgens in adipocytes leads to elevated estrogens levels

 

GH level decreases

 

GH binding protein increases

 

IGF-1 normal or slightly low

 

Cortisol normal

 

24-hour urine free cortisol (UFC) excretion high-normal

 

TSH normal or slightly increased

 

REFERENCES

 

  1. Schulz TJ, Tseng YH. Brown adipose tissue: development, metabolism and beyond. Biochem J.2013;453(2):167-178.
  2. Vong L, Ye C, Yang Z, Choi B, Chua S, Jr., Lowell BB. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron. 2011;71(1):142-154.
  3. Cooper JA, Polonsky KS, Schoeller DA. Serum leptin levels in obese males during over- and underfeeding. Obesity (Silver Spring). 2009;17(12):2149-2154.
  4. Klok MD, Jakobsdottir S, Drent ML. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev. 2007;8(1):21-34.
  5. Holm JC, Gamborg M, Ward LC, Gammeltoft S, Kaas-Ibsen K, Heitmann BL, Sorensen TI. Tracking of leptin, soluble leptin receptor, and the free leptin index during weight loss and regain in children. Obes Facts.2011;4(6):461-468.
  6. Levin BE, Dunn-Meynell AA, Banks WA. Obesity-prone rats have normal blood-brain barrier transport but defective central leptin signaling before obesity onset. Am J Physiol Regul Integr Comp Physiol.2004;286(1):R143-150.
  7. El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest. 2000;105(12):1827-1832.
  8. Rhee SD, Sung YY, Lee YS, Kim JY, Jung WH, Kim MJ, Lee MS, Lee MK, Yang SD, Cheon HG. Obesity of TallyHO/JngJ mouse is due to increased food intake with early development of leptin resistance. Exp Clin Endocrinol Diabetes. 2011;119(4):243-251.
  9. de Lartigue G, Barbier de la Serre C, Espero E, Lee J, Raybould HE. Diet-induced obesity leads to the development of leptin resistance in vagal afferent neurons. Am J Physiol Endocrinol Metab. 2011;301(1):E187-195.
  10. Pan WW, Myers MG, Jr. Leptin and the maintenance of elevated body weight. Nat Rev Neurosci.2018;19(2):95-105.
  11. de Git KCG, Peterse C, Beerens S, Luijendijk MCM, van der Plasse G, la Fleur SE, Adan RAH. Is leptin resistance the cause or the consequence of diet-induced obesity? Int J Obes (Lond). 2018;42(8):1445-1457.
  12. Friedman JM. Leptin and the endocrine control of energy balance. Nat Metab. 2019;1(8):754-764.
  13. Zhao S, Kusminski CM, Elmquist JK, Scherer PE. Leptin: Less Is More. Diabetes. 2020;69(5):823-829.
  14. Triantafyllou GA, Paschou SA, Mantzoros CS. Leptin and Hormones: Energy Homeostasis. Endocrinol Metab Clin North Am. 2016;45(3):633-645.
  15. Sinha G. Leptin therapy gains FDA approval. Nat Biotechnol. 2014;32(4):300-302.
  16. Ahima RS, Saper CB, Flier JS, Elmquist JK. Leptin regulation of neuroendocrine systems. Front Neuroendocrinol. 2000;21(3):263-307.
  17. Hausman GJ, Barb CR, Lents CA. Leptin and reproductive function. Biochimie. 2012;94(10):2075-2081.
  18. Paz-Filho G, Mastronardi C, Wong ML, Licinio J. Leptin therapy, insulin sensitivity, and glucose homeostasis. Indian J Endocrinol Metab. 2012;16(Suppl 3):S549-555.
  19. Ahima RS, Flier JS. Leptin. Annu Rev Physiol. 2000;62:413-437.
  20. Tessier DR, Ferraro ZM, Gruslin A. Role of leptin in pregnancy: consequences of maternal obesity. Placenta.2013;34(3):205-211.
  21. Andò S, Gelsomino L, Panza S, Giordano C, Bonofiglio D, Barone I, Catalano S. Obesity, Leptin and Breast Cancer: Epidemiological Evidence and Proposed Mechanisms. Cancers (Basel). 2019;11(1).
  22. Yoon YS, Kwon AR, Lee YK, Oh SW. Circulating adipokines and risk of obesity related cancers: A systematic review and meta-analysis. Obes Res Clin Pract. 2019;13(4):329-339.
  23. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest. 2006;116(7):1784-1792.
  24. Kantartzis K, Fritsche A, Tschritter O, Thamer C, Haap M, Schafer S, Stumvoll M, Haring HU, Stefan N. The association between plasma adiponectin and insulin sensitivity in humans depends on obesity. Obes Res.2005;13(10):1683-1691.
  25. Cnop M, Havel PJ, Utzschneider KM, Carr DB, Sinha MK, Boyko EJ, Retzlaff BM, Knopp RH, Brunzell JD, Kahn SE. Relationship of adiponectin to body fat distribution, insulin sensitivity and plasma lipoproteins: evidence for independent roles of age and sex. Diabetologia. 2003;46(4):459-469.
  26. Sattar N, Wannamethee G, Sarwar N, Tchernova J, Cherry L, Wallace AM, Danesh J, Whincup PH. Adiponectin and coronary heart disease: a prospective study and meta-analysis. Circulation. 2006;114(7):623-629.
  27. Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa Y, Chao CL, Chen CL, Tai TY, Chuang LM. Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab. 2001;86(8):3815-3819.
  28. Frithioff-Bøjsøe C, Lund MAV, Lausten-Thomsen U, Hedley PL, Pedersen O, Christiansen M, Baker JL, Hansen T, Holm JC. Leptin, adiponectin, and their ratio as markers of insulin resistance and cardiometabolic risk in childhood obesity. Pediatr Diabetes. 2020;21(2):194-202.
  29. Palit SP, Patel R, Jadeja SD, Rathwa N, Mahajan A, Ramachandran AV, Dhar MK, Sharma S, Begum R. A genetic analysis identifies a haplotype at adiponectin locus: Association with obesity and type 2 diabetes. Sci Rep. 2020;10(1):2904.
  30. Kita S, Maeda N, Shimomura I. Interorgan communication by exosomes, adipose tissue, and adiponectin in metabolic syndrome. J Clin Invest. 2019;129(10):4041-4049.
  31. Goldstein BJ, Scalia R. Adiponectin: A novel adipokine linking adipocytes and vascular function. J Clin Endocrinol Metab. 2004;89(6):2563-2568.
  32. Wilk S, Jenke A, Stehr J, Yang CA, Bauer S, Goldner K, Kotsch K, Volk HD, Poller W, Schultheiss HP, Skurk C, Scheibenbogen C. Adiponectin modulates NK-cell function. Eur J Immunol. 2013;43(4):1024-1033.
  33. Wilk S, Scheibenbogen C, Bauer S, Jenke A, Rother M, Guerreiro M, Kudernatsch R, Goerner N, Poller W, Elligsen-Merkel D, Utku N, Magrane J, Volk HD, Skurk C. Adiponectin is a negative regulator of antigen-activated T cells. Eur J Immunol. 2011;41(8):2323-2332.
  34. Karbowska J, Kochan Z. Role of adiponectin in the regulation of carbohydrate and lipid metabolism. J Physiol Pharmacol. 2006;57 Suppl 6:103-113.
  35. Fenton JI, Birmingham JM, Hursting SD, Hord NG. Adiponectin blocks multiple signaling cascades associated with leptin-induced cell proliferation in Apc Min/+ colon epithelial cells. Int J Cancer. 2008;122(11):2437-2445.
  36. Mantzoros C, Petridou E, Dessypris N, Chavelas C, Dalamaga M, Alexe DM, Papadiamantis Y, Markopoulos C, Spanos E, Chrousos G, Trichopoulos D. Adiponectin and breast cancer risk. J Clin Endocrinol Metab.2004;89(3):1102-1107.
  37. Ishikawa M, Kitayama J, Kazama S, Hiramatsu T, Hatano K, Nagawa H. Plasma adiponectin and gastric cancer. Clin Cancer Res. 2005;11(2 Pt 1):466-472.
  38. Abdul-Ghafar J, Oh SS, Park SM, Wairagu P, Lee SN, Jeong Y, Eom M, Yong SJ, Jung SH. Expression of adiponectin receptor 1 is indicative of favorable prognosis in non-small cell lung carcinoma. Tohoku J Exp Med.2013;229(2):153-162.
  39. Michalakis K, Williams CJ, Mitsiades N, Blakeman J, Balafouta-Tselenis S, Giannopoulos A, Mantzoros CS. Serum adiponectin concentrations and tissue expression of adiponectin receptors are reduced in patients with prostate cancer: a case control study. Cancer Epidemiol Biomarkers Prev. 2007;16(2):308-313.
  40. Morais TC, de Abreu LC, de Quental OB, Pessoa RS, Fujimori M, Daboin BEG, França EL, Honorio-França AC. Obesity as an Inflammatory Agent Can Cause Cellular Changes in Human Milk due to the Actions of the Adipokines Leptin and Adiponectin. Cells. 2019;8(6).
  41. Bozaoglu K, Curran JE, Stocker CJ, Zaibi MS, Segal D, Konstantopoulos N, Morrison S, Carless M, Dyer TD, Cole SA, Goring HH, Moses EK, Walder K, Cawthorne MA, Blangero J, Jowett JB. Chemerin, a novel adipokine in the regulation of angiogenesis. J Clin Endocrinol Metab. 2010;95(5):2476-2485.
  42. Fatima SS, Rehman R, Baig M, Khan TA. New roles of the multidimensional adipokine: chemerin. Peptides.2014;62:15-20.
  43. Niklowitz P, Rothermel J, Lass N, Barth A, Reinehr T. Link between chemerin, central obesity, and parameters of the Metabolic Syndrome: findings from a longitudinal study in obese children participating in a lifestyle intervention. Int J Obes (Lond). 2018;42(10):1743-1752.
  44. Ministrini S, Ricci MA, Nulli Migliola E, De Vuono S, D'Abbondanza M, Paganelli MT, Vaudo G, Siepi D, Lupattelli G. Chemerin predicts carotid intima-media thickening in severe obesity. Eur J Clin Invest.2020:e13256.
  45. Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC, Gong DW. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290(6):E1253-1261.
  46. Watanabe T, Watanabe-Kominato K, Takahashi Y, Kojima M, Watanabe R. Adipose Tissue-Derived Omentin-1 Function and Regulation. Compr Physiol. 2017;7(3):765-781.
  47. Rothermel J, Lass N, Barth A, Reinehr T. Link between omentin-1, obesity and insulin resistance in children: Findings from a longitudinal intervention study. Pediatr Obes. 2020;15(5):e12605.
  48. Özgen İ T, Oruçlu Ş, Selek S, Kutlu E, Guzel G, Cesur Y. Omentin-1 level in adolescents with polycystic ovarian syndrome. Pediatr Int. 2019;61(2):147-151.
  49. Zhou JY, Chan L, Zhou SW. Omentin: linking metabolic syndrome and cardiovascular disease. Curr Vasc Pharmacol. 2014;12(1):136-143.
  50. Franz M, Polterauer M, Springer S, Kuessel L, Haslinger P, Worda C, Worda K. Maternal and neonatal omentin-1 levels in gestational diabetes. Arch Gynecol Obstet. 2018;297(4):885-889.
  51. Okuno M, Caraveo VE, Goodman DS, Blaner WS. Regulation of adipocyte gene expression by retinoic acid and hormones: effects on the gene encoding cellular retinol-binding protein. J Lipid Res. 1995;36(1):137-147.
  52. Janke J, Engeli S, Boschmann M, Adams F, Bohnke J, Luft FC, Sharma AM, Jordan J. Retinol-binding protein 4 in human obesity. Diabetes. 2006;55(10):2805-2810.
  53. Haider DG, Schindler K, Prager G, Bohdjalian A, Luger A, Wolzt M, Ludvik B. Serum retinol-binding protein 4 is reduced after weight loss in morbidly obese subjects. J Clin Endocrinol Metab. 2007;92(3):1168-1171.
  54. Christou GA, Tselepis AD, Kiortsis DN. The metabolic role of retinol binding protein 4: an update. Horm Metab Res. 2012;44(1):6-14.
  55. Bluher M. Vaspin in obesity and diabetes: pathophysiological and clinical significance. Endocrine.2012;41(2):176-182.
  56. Kralisch S, Fasshauer M. Adipocyte fatty acid binding protein: a novel adipokine involved in the pathogenesis of metabolic and vascular disease? Diabetologia. 2013;56(1):10-21.
  57. Horakova D, Pastucha D, Stejskal D, Kollarova H, Azeem K, Janout V. Adipocyte fatty acid binding protein and C-reactive protein levels as indicators of insulin resistance development. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2011;155(4):355-359.
  58. Cianflone K, Xia Z, Chen LY. Critical review of acylation-stimulating protein physiology in humans and rodents. Biochimica et biophysica acta. 2003;1609(2):127-143.
  59. Munkonda MN, Lapointe M, Miegueu P, Roy C, Gauvreau D, Richard D, Cianflone K. Recombinant acylation stimulating protein administration to C3-/- mice increases insulin resistance via adipocyte inflammatory mechanisms. PloS one. 2012;7(10):e46883.
  60. Karlsson C, Lindell K, Ottosson M, Sjostrom L, Carlsson B, Carlsson LM. Human adipose tissue expresses angiotensinogen and enzymes required for its conversion to angiotensin II. J Clin Endocrinol Metab.1998;83(11):3925-3929.
  61. Gupte M, Thatcher SE, Boustany-Kari CM, Shoemaker R, Yiannikouris F, Zhang X, Karounos M, Cassis LA. Angiotensin converting enzyme 2 contributes to sex differences in the development of obesity hypertension in C57BL/6 mice. Arterioscler Thromb Vasc Biol. 2012;32(6):1392-1399.
  62. Gupte M, Boustany-Kari CM, Bharadwaj K, Police S, Thatcher S, Gong MC, English VL, Cassis LA. ACE2 is expressed in mouse adipocytes and regulated by a high-fat diet. Am J Physiol Regul Integr Comp Physiol.2008;295(3):R781-788.
  63. Shimomura I, Funahashi T, Takahashi M, Maeda K, Kotani K, Nakamura T, Yamashita S, Miura M, Fukuda Y, Takemura K, Tokunaga K, Matsuzawa Y. Enhanced expression of PAI-1 in visceral fat: possible contributor to vascular disease in obesity. Nat Med. 1996;2(7):800-803.
  64. Kalea AZ, Batlle D. Apelin and ACE2 in cardiovascular disease. Curr Opin Investig Drugs. 2010;11(3):273-282.
  65. Ortega-Molina A, Efeyan A, Lopez-Guadamillas E, Munoz-Martin M, Gomez-Lopez G, Canamero M, Mulero F, Pastor J, Martinez S, Romanos E, Mar Gonzalez-Barroso M, Rial E, Valverde AM, Bischoff JR, Serrano M. Pten positively regulates brown adipose function, energy expenditure, and longevity. Cell metabolism.2012;15(3):382-394.
  66. El-Mikkawy DME, El-Sadek MA, El-Badawy MA, Samaha D. Circulating level of interleukin-6 in relation to body mass indices and lipid profile in Egyptian adults with overweight and obesity. Egyptian Rheumatology and Rehabilitation. 2020;47(1):7.
  67. Fried SK, Bunkin DA, Greenberg AS. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6: depot difference and regulation by glucocorticoid. J Clin Endocrinol Metab. 1998;83(3):847-850.
  68. Stith RD, Luo J. Endocrine and carbohydrate responses to interleukin-6 in vivo. Circ Shock. 1994;44(4):210-215.
  69. Wallenius V, Wallenius K, Ahren B, Rudling M, Carlsten H, Dickson SL, Ohlsson C, Jansson JO. Interleukin-6-deficient mice develop mature-onset obesity. Nat Med. 2002;8(1):75-79.
  70. Sadagurski M, Norquay L, Farhang J, D'Aquino K, Copps K, White MF. Human IL6 enhances leptin action in mice. Diabetologia. 2010;53(3):525-535.
  71. Matos MF, Lourenco DM, Orikaza CM, Gouveia CP, Morelli VM. Abdominal obesity and the risk of venous thromboembolism among women: a potential role of interleukin-6. Metab Syndr Relat Disord. 2013;11(1):29-34.
  72. Pasquali R. Obesity and androgens: facts and perspectives. Fertil Steril. 2006;85(5):1319-1340.
  73. Bhasin S, Cunningham GR, Hayes FJ, Matsumoto AM, Snyder PJ, Swerdloff RS, Montori VM, Task Force ES. Testosterone therapy in men with androgen deficiency syndromes: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95(6):2536-2559.
  74. Corona G, Rastrelli G, Morelli A, Vignozzi L, Mannucci E, Maggi M. Hypogonadism and metabolic syndrome. J Endocrinol Invest. 2011;34(7):557-567.
  75. Hofstra J, Loves S, van Wageningen B, Ruinemans-Koerts J, Jansen I, de Boer H. High prevalence of hypogonadotropic hypogonadism in men referred for obesity treatment. Neth J Med. 2008;66(3):103-109.
  76. van Hulsteijn LT, Pasquali R, Casanueva F, Haluzik M, Ledoux S, Monteiro MP, Salvador J, Santini F, Toplak H, Dekkers OM. Prevalence of endocrine disorders in obese patients: systematic review and meta-analysis. Eur J Endocrinol. 2020;182(1):11-21.
  77. Saboor Aftab SA, Kumar S, Barber TM. The role of obesity and type 2 diabetes mellitus in the development of male obesity-associated secondary hypogonadism. Clin Endocrinol (Oxf). 2013;78(3):330-337.
  78. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol. 2004;25(1):4-7.
  79. Luboshitzky R, Lavie L, Shen-Orr Z, Herer P. Altered luteinizing hormone and testosterone secretion in middle-aged obese men with obstructive sleep apnea. Obes Res. 2005;13(4):780-786.
  80. Hammoud AO, Carrell DT, Gibson M, Peterson CM, Meikle AW. Updates on the relation of weight excess and reproductive function in men: sleep apnea as a new area of interest. Asian J Androl. 2012;14(1):77-81.
  81. Wagner IV, Kloting N, Atanassova N, Savchuk I, Sprote C, Kiess W, Soder O, Svechnikov K. Prepubertal onset of obesity negatively impacts on testicular steroidogenesis in rats. Mol Cell Endocrinol. 2016;437:154-162.
  82. Lapauw B, Kaufman JM. MANAGEMENT OF ENDOCRINE DISEASE: Rationale and current evidence for testosterone therapy in the management of obesity and its complications. Eur J Endocrinol. 2020;183(6):R167-r183.
  83. Isidori AM, Giannetta E, Greco EA, Gianfrilli D, Bonifacio V, Isidori A, Lenzi A, Fabbri A. Effects of testosterone on body composition, bone metabolism and serum lipid profile in middle-aged men: a meta-analysis. Clin Endocrinol (Oxf). 2005;63(3):280-293.
  84. Kapoor D, Goodwin E, Channer KS, Jones TH. Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. Eur J Endocrinol. 2006;154(6):899-906.
  85. Strain GW, Zumoff B, Miller LK, Rosner W, Levit C, Kalin M, Hershcopf RJ, Rosenfeld RS. Effect of massive weight loss on hypothalamic-pituitary-gonadal function in obese men. J Clin Endocrinol Metab. 1988;66(5):1019-1023.
  86. Belanger C, Luu-The V, Dupont P, Tchernof A. Adipose tissue intracrinology: potential importance of local androgen/estrogen metabolism in the regulation of adiposity. Horm Metab Res. 2002;34(11-12):737-745.
  87. Mammi C, Calanchini M, Antelmi A, Cinti F, Rosano GM, Lenzi A, Caprio M, Fabbri A. Androgens and adipose tissue in males: a complex and reciprocal interplay. International journal of endocrinology. 2012;2012:789653.
  88. Corona G, Rastrelli G, Monami M, Saad F, Luconi M, Lucchese M, Facchiano E, Sforza A, Forti G, Mannucci E, Maggi M. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: a systematic review and meta-analysis. Eur J Endocrinol. 2013;168(6):829-843.
  89. Hammoud A, Gibson M, Hunt SC, Adams TD, Carrell DT, Kolotkin RL, Meikle AW. Effect of Roux-en-Y gastric bypass surgery on the sex steroids and quality of life in obese men. J Clin Endocrinol Metab. 2009;94(4):1329-1332.
  90. Tchernof A, Labrie F, Belanger A, Despres JP. Obesity and metabolic complications: contribution of dehydroepiandrosterone and other steroid hormones. J Endocrinol. 1996;150 Suppl:S155-164.
  91. Segall-Gutierrez P, Du J, Niu C, Ge M, Tilley I, Mizraji K, Stanczyk FZ. Effect of subcutaneous depot-medroxyprogesterone acetate (DMPA-SC) on serum androgen markers in normal-weight, obese, and extremely obese women. Contraception. 2012;86(6):739-745.
  92. Legro RS, Schlaff WD, Diamond MP, Coutifaris C, Casson PR, Brzyski RG, Christman GM, Trussell JC, Krawetz SA, Snyder PJ, Ohl D, Carson SA, Steinkampf MP, Carr BR, McGovern PG, Cataldo NA, Gosman GG, Nestler JE, Myers ER, Santoro N, Eisenberg E, Zhang M, Zhang H, Reproductive Medicine N. Total testosterone assays in women with polycystic ovary syndrome: precision and correlation with hirsutism. J Clin Endocrinol Metab. 2010;95(12):5305-5313.
  93. Elks CE, Perry JR, Sulem P, Chasman DI, Franceschini N, He C, Lunetta KL, Visser JA, Byrne EM, Cousminer DL, Gudbjartsson DF, Esko T, Feenstra B, Hottenga JJ, Koller DL, Kutalik Z, Lin P, Mangino M, Marongiu M, McArdle PF, Smith AV, Stolk L, van Wingerden SH, Zhao JH, Albrecht E, Corre T, Ingelsson E, Hayward C, Magnusson PK, Smith EN, Ulivi S, Warrington NM, Zgaga L, Alavere H, Amin N, Aspelund T, Bandinelli S, Barroso I, Berenson GS, Bergmann S, Blackburn H, Boerwinkle E, Buring JE, Busonero F, Campbell H, Chanock SJ, Chen W, Cornelis MC, Couper D, Coviello AD, d'Adamo P, de Faire U, de Geus EJ, Deloukas P, Döring A, Smith GD, Easton DF, Eiriksdottir G, Emilsson V, Eriksson J, Ferrucci L, Folsom AR, Foroud T, Garcia M, Gasparini P, Geller F, Gieger C, Gudnason V, Hall P, Hankinson SE, Ferreli L, Heath AC, Hernandez DG, Hofman A, Hu FB, Illig T, Järvelin MR, Johnson AD, Karasik D, Khaw KT, Kiel DP, Kilpeläinen TO, Kolcic I, Kraft P, Launer LJ, Laven JS, Li S, Liu J, Levy D, Martin NG, McArdle WL, Melbye M, Mooser V, Murray JC, Murray SS, Nalls MA, Navarro P, Nelis M, Ness AR, Northstone K, Oostra BA, Peacock M, Palmer LJ, Palotie A, Paré G, Parker AN, Pedersen NL, Peltonen L, Pennell CE, Pharoah P, Polasek O, Plump AS, Pouta A, Porcu E, Rafnar T, Rice JP, Ring SM, Rivadeneira F, Rudan I, Sala C, Salomaa V, Sanna S, Schlessinger D, Schork NJ, Scuteri A, Segrè AV, Shuldiner AR, Soranzo N, Sovio U, Srinivasan SR, Strachan DP, Tammesoo ML, Tikkanen E, Toniolo D, Tsui K, Tryggvadottir L, Tyrer J, Uda M, van Dam RM, van Meurs JB, Vollenweider P, Waeber G, Wareham NJ, Waterworth DM, Weedon MN, Wichmann HE, Willemsen G, Wilson JF, Wright AF, Young L, Zhai G, Zhuang WV, Bierut LJ, Boomsma DI, Boyd HA, Crisponi L, Demerath EW, van Duijn CM, Econs MJ, Harris TB, Hunter DJ, Loos RJ, Metspalu A, Montgomery GW, Ridker PM, Spector TD, Streeten EA, Stefansson K, Thorsteinsdottir U, Uitterlinden AG, Widen E, Murabito JM, Ong KK, Murray A. Thirty new loci for age at menarche identified by a meta-analysis of genome-wide association studies. Nat Genet. 2010;42(12):1077-1085.
  94. Fernandez-Rhodes L, Demerath EW, Cousminer DL, Tao R, Dreyfus JG, Esko T, Smith AV, Gudnason V, Harris TB, Launer L, McArdle PF, Yerges-Armstrong LM, Elks CE, Strachan DP, Kutalik Z, Vollenweider P, Feenstra B, Boyd HA, Metspalu A, Mihailov E, Broer L, Zillikens MC, Oostra B, van Duijn CM, Lunetta KL, Perry JR, Murray A, Koller DL, Lai D, Corre T, Toniolo D, Albrecht E, Stockl D, Grallert H, Gieger C, Hayward C, Polasek O, Rudan I, Wilson JF, He C, Kraft P, Hu FB, Hunter DJ, Hottenga JJ, Willemsen G, Boomsma DI, Byrne EM, Martin NG, Montgomery GW, Warrington NM, Pennell CE, Stolk L, Visser JA, Hofman A, Uitterlinden AG, Rivadeneira F, Lin P, Fisher SL, Bierut LJ, Crisponi L, Porcu E, Mangino M, Zhai G, Spector TD, Buring JE, Rose LM, Ridker PM, Poole C, Hirschhorn JN, Murabito JM, Chasman DI, Widen E, North KE, Ong KK, Franceschini N. Association of adiposity genetic variants with menarche timing in 92,105 women of European descent. Am J Epidemiol. 2013;178(3):451-460.
  95. Herman-Giddens ME. Recent data on pubertal milestones in United States children: the secular trend toward earlier development. Int J Androl. 2006;29(1):241-246; discussion 286-290.
  96. Parent AS, Teilmann G, Juul A, Skakkebaek NE, Toppari J, Bourguignon JP. The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration. Endocr Rev. 2003;24(5):668-693.
  97. Mumby HS, Elks CE, Li S, Sharp SJ, Khaw KT, Luben RN, Wareham NJ, Loos RJ, Ong KK. Mendelian Randomisation Study of Childhood BMI and Early Menarche. Journal of obesity. 2011;2011:180729.
  98. Pierce MB, Leon DA. Age at menarche and adult BMI in the Aberdeen children of the 1950s cohort study. Am J Clin Nutr. 2005;82(4):733-739.
  99. He C, Zhang C, Hunter DJ, Hankinson SE, Buck Louis GM, Hediger ML, Hu FB. Age at menarche and risk of type 2 diabetes: results from 2 large prospective cohort studies. Am J Epidemiol. 2010;171(3):334-344.
  100. Stockl D, Doring A, Peters A, Thorand B, Heier M, Huth C, Stockl H, Rathmann W, Kowall B, Meisinger C. Age at menarche is associated with prediabetes and diabetes in women (aged 32-81 years) from the general population: the KORA F4 Study. Diabetologia. 2012;55(3):681-688.
  101. Elks CE, Ong KK, Scott RA, van der Schouw YT, Brand JS, Wark PA, Amiano P, Balkau B, Barricarte A, Boeing H, Fonseca-Nunes A, Franks PW, Grioni S, Halkjaer J, Kaaks R, Key TJ, Khaw KT, Mattiello A, Nilsson PM, Overvad K, Palli D, Quirós JR, Rinaldi S, Rolandsson O, Romieu I, Sacerdote C, Sánchez MJ, Spijkerman AM, Tjonneland A, Tormo MJ, Tumino R, van der AD, Forouhi NG, Sharp SJ, Langenberg C, Riboli E, Wareham NJ. Age at menarche and type 2 diabetes risk: the EPIC-InterAct study. Diabetes Care. 2013;36(11):3526-3534.
  102. Prentice P, Viner RM. Pubertal timing and adult obesity and cardiometabolic risk in women and men: a systematic review and meta-analysis. Int J Obes (Lond). 2013;37(8):1036-1043.
  103. Canoy D, Beral V, Balkwill A, Wright FL, Kroll ME, Reeves GK, Green J, Cairns BJ. Age at menarche and risks of coronary heart and other vascular diseases in a large UK cohort. Circulation. 2015;131(3):237-244.
  104. Cancer CGoHFiB. Menarche, menopause, and breast cancer risk: individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol. 2012;13(11):1141-1151.
  105. Charalampopoulos D, McLoughlin A, Elks CE, Ong KK. Age at menarche and risks of all-cause and cardiovascular death: a systematic review and meta-analysis. Am J Epidemiol. 2014;180(1):29-40.
  106. Bleil ME, Appelhans BM, Adler NE, Gregorich SE, Sternfeld B, Cedars MI. Pubertal timing, androgens, and obesity phenotypes in women at midlife. J Clin Endocrinol Metab. 2012;97(10):E1948-1952.
  107. Hong Y, Maessen SE, Dong G, Huang K, Wu W, Liang L, Wang CL, Chen X, Gibbins JD, Cutfield WS, Derraik JGB, Fu J. Associations between maternal age at menarche and anthropometric and metabolic parameters in the adolescent offspring. Clin Endocrinol (Oxf). 2019;90(5):702-710.
  108. Terasawa E. Cellular mechanism of pulsatile LHRH release. Gen Comp Endocrinol. 1998;112(3):283-295.
  109. Tartaglia LA. The leptin receptor. J Biol Chem. 1997;272(10):6093-6096.
  110. Reinehr T, Roth CL. Is there a causal relationship between obesity and puberty? Lancet Child Adolesc Health.2019;3(1):44-54.
  111. Farooqi IS. Leptin and the onset of puberty: insights from rodent and human genetics. Semin Reprod Med.2002;20(2):139-144.
  112. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351(10):987-997.
  113. George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology. 2010;91(4):302-307.
  114. Hameed S, Dhillo WS. Biology of kisspeptins. Front Horm Res. 2010;39:25-36.
  115. Kang MJ, Oh YJ, Shim YS, Baek JW, Yang S, Hwang IT. The usefulness of circulating levels of leptin, kisspeptin, and neurokinin B in obese girls with precocious puberty. Gynecol Endocrinol. 2018;34(7):627-630.
  116. Sitticharoon C, Mutirangura P, Chinachoti T, Iamaroon A, Triyasunant N, Churintaraphan M, Keadkraichaiwat I, Maikaew P, Sririwichitchai R. Associations of serum kisspeptin levels with metabolic and reproductive parameters in men. Peptides. 2021;135:170433.
  117. Vazquez MJ, Velasco I, Tena-Sempere M. Novel mechanisms for the metabolic control of puberty: implications for pubertal alterations in early-onset obesity and malnutrition. J Endocrinol. 2019;242(2):R51-r65.
  118. Spicer LJ, Francisco CC. The adipose obese gene product, leptin: evidence of a direct inhibitory role in ovarian function. Endocrinology. 1997;138(8):3374-3379.
  119. Caprio M, Isidori AM, Carta AR, Moretti C, Dufau ML, Fabbri A. Expression of functional leptin receptors in rodent Leydig cells. Endocrinology. 1999;140(11):4939-4947.
  120. Ghizzoni L, Barreca A, Mastorakos G, Furlini M, Vottero A, Ferrari B, Chrousos GP, Bernasconi S. Leptin inhibits steroid biosynthesis by human granulosa-lutein cells. Horm Metab Res. 2001;33(6):323-328.
  121. Ma X, Hayes E, Prizant H, Srivastava RK, Hammes SR, Sen A. Leptin-Induced CART (Cocaine- and Amphetamine-Regulated Transcript) Is a Novel Intraovarian Mediator of Obesity-Related Infertility in Females. Endocrinology. 2016;157(3):1248-1257.
  122. Gregoraszczuk EL, Rak A. Superactive human leptin antagonist reverses leptin-induced excessive progesterone and testosterone secretion in porcine ovarian follicles by blocking leptin receptors. J Physiol Pharmacol. 2015;66(1):39-46.
  123. Rizk NM, Sharif E. Leptin as well as Free Leptin Receptor Is Associated with Polycystic Ovary Syndrome in Young Women. International journal of endocrinology. 2015;2015:927805.
  124. Goodarzi MO, Dumesic DA, Chazenbalk G, Azziz R. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat Rev Endocrinol. 2011;7(4):219-231.
  125. DeUgarte CM, Bartolucci AA, Azziz R. Prevalence of insulin resistance in the polycystic ovary syndrome using the homeostasis model assessment. Fertil Steril. 2005;83(5):1454-1460.
  126. Wells JC. Sexual dimorphism of body composition. Best Pract Res Clin Endocrinol Metab. 2007;21(3):415-430.
  127. Goodpaster BH, Krishnaswami S, Harris TB, Katsiaras A, Kritchevsky SB, Simonsick EM, Nevitt M, Holvoet P, Newman AB. Obesity, regional body fat distribution, and the metabolic syndrome in older men and women. Arch Intern Med. 2005;165(7):777-783.
  128. Borruel S, Fernandez-Duran E, Alpanes M, Marti D, Alvarez-Blasco F, Luque-Ramirez M, Escobar-Morreale HF. Global adiposity and thickness of intraperitoneal and mesenteric adipose tissue depots are increased in women with polycystic ovary syndrome (PCOS). J Clin Endocrinol Metab. 2013;98(3):1254-1263.
  129. Martinez-Garcia MA, Montes-Nieto R, Fernandez-Duran E, Insenser M, Luque-Ramirez M, Escobar-Morreale HF. Evidence for masculinization of adipokine gene expression in visceral and subcutaneous adipose tissue of obese women with polycystic ovary syndrome (PCOS). J Clin Endocrinol Metab. 2013;98(2):E388-396.
  130. Rojas J, Chavez M, Olivar L, Rojas M, Morillo J, Mejias J, Calvo M, Bermudez V. Polycystic ovary syndrome, insulin resistance, and obesity: navigating the pathophysiologic labyrinth. Int J Reprod Med. 2014;2014:719050.
  131. Tziomalos K, Katsikis I, Papadakis E, Kandaraki EA, Macut D, Panidis D. Comparison of markers of insulin resistance and circulating androgens between women with polycystic ovary syndrome and women with metabolic syndrome. Hum Reprod. 2013;28(3):785-793.
  132. Dignam WJ, Parlow AF, Daane TA. Serum FSH and LH measurements in the evaluation of menstrual disorders. Am J Obstet Gynecol. 1969;105(5):679-695.
  133. Kopelman PG, Pilkington TR, White N, Jeffcoate SL. Abnormal sex steroid secretion and binding in massively obese women. Clin Endocrinol (Oxf). 1980;12(4):363-369.
  134. Blank SK, McCartney CR, Marshall JC. The origins and sequelae of abnormal neuroendocrine function in polycystic ovary syndrome. Hum Reprod Update. 2006;12(4):351-361.
  135. Al-Nozha O, Habib F, Mojaddidi M, El-Bab MF. Body weight reduction and metformin: Roles in polycystic ovary syndrome. Pathophysiology. 2013;20(2):131-137.
  136. Practice Committee of the American Society for Reproductive Medicine. Electronic address Aao, Practice Committee of the American Society for Reproductive M. Role of metformin for ovulation induction in infertile patients with polycystic ovary syndrome (PCOS): a guideline. Fertil Steril. 2017;108(3):426-441.
  137. Tang T, Lord JM, Norman RJ, Yasmin E, Balen AH. Insulin-sensitising drugs (metformin, rosiglitazone, pioglitazone, D-chiro-inositol) for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility. Cochrane Database Syst Rev. 2012(5):CD003053.
  138. Legro RS. Ovulation induction in polycystic ovary syndrome: Current options. Best Pract Res Clin Obstet Gynaecol. 2016;37:152-159.
  139. Agrawal A, Mahey R, Kachhawa G, Khadgawat R, Vanamail P, Kriplani A. Comparison of metformin plus myoinositol vs metformin alone in PCOS women undergoing ovulation induction cycles: randomized controlled trial. Gynecol Endocrinol. 2019;35(6):511-514.
  140. Laganà AS, Vitagliano A, Noventa M, Ambrosini G, D'Anna R. Myo-inositol supplementation reduces the amount of gonadotropins and length of ovarian stimulation in women undergoing IVF: a systematic review and meta-analysis of randomized controlled trials. Arch Gynecol Obstet. 2018;298(4):675-684.
  141. Facchinetti F, Unfer V, Dewailly D, Kamenov ZA, Diamanti-Kandarakis E, Laganà AS, Nestler JE, Soulage CO. Inositols in Polycystic Ovary Syndrome: An Overview on the Advances. Trends Endocrinol Metab.2020;31(6):435-447.
  142. Emekçi Özay Ö, Özay AC, Çağlıyan E, Okyay RE, Gülekli B. Myo-inositol administration positively effects ovulation induction and intrauterine insemination in patients with polycystic ovary syndrome: a prospective, controlled, randomized trial. Gynecol Endocrinol. 2017;33(7):524-528.
  143. Schneider G, Kirschner MA, Berkowitz R, Ertel NH. Increased estrogen production in obese men. J Clin Endocrinol Metab. 1979;48(4):633-638.
  144. Tchernof A, Despres JP, Dupont A, Belanger A, Nadeau A, Prud'homme D, Moorjani S, Lupien PJ, Labrie F. Relation of steroid hormones to glucose tolerance and plasma insulin levels in men. Importance of visceral adipose tissue. Diabetes Care. 1995;18(3):292-299.
  145. Ribas V, Nguyen MT, Henstridge DC, Nguyen AK, Beaven SW, Watt MJ, Hevener AL. Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab. 2010;298(2):E304-319.
  146. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci U S A. 2000;97(23):12729-12734.
  147. Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev. 2013;34(3):309-338.
  148. McNelis JC, Olefsky JM. Macrophages, immunity, and metabolic disease. Immunity. 2014;41(1):36-48.
  149. Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol.2011;11(11):738-749.
  150. Tiano JP, Mauvais-Jarvis F. Importance of oestrogen receptors to preserve functional beta-cell mass in diabetes. Nat Rev Endocrinol. 2012;8(6):342-351.
  151. Lundholm L, Zang H, Hirschberg AL, Gustafsson JA, Arner P, Dahlman-Wright K. Key lipogenic gene expression can be decreased by estrogen in human adipose tissue. Fertil Steril. 2008;90(1):44-48.
  152. Elbers JM, Giltay EJ, Teerlink T, Scheffer PG, Asscheman H, Seidell JC, Gooren LJ. Effects of sex steroids on components of the insulin resistance syndrome in transsexual subjects. Clin Endocrinol (Oxf). 2003;58(5):562-571.
  153. Nichols HB, Trentham-Dietz A, Egan KM, Titus-Ernstoff L, Holmes MD, Bersch AJ, Holick CN, Hampton JM, Stampfer MJ, Willett WC, Newcomb PA. Body mass index before and after breast cancer diagnosis: associations with all-cause, breast cancer, and cardiovascular disease mortality. Cancer Epidemiol Biomarkers Prev. 2009;18(5):1403-1409.
  154. Patterson RE, Cadmus LA, Emond JA, Pierce JP. Physical activity, diet, adiposity and female breast cancer prognosis: a review of the epidemiologic literature. Maturitas. 2010;66(1):5-15.
  155. Berryman DE, Glad CA, List EO, Johannsson G. The GH/IGF-1 axis in obesity: pathophysiology and therapeutic considerations. Nat Rev Endocrinol. 2013;9(6):346-356.
  156. Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev. 2009;30(2):152-177.
  157. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev. 1998;19(6):717-797.
  158. Vahl N, Jorgensen JO, Skjaerbaek C, Veldhuis JD, Orskov H, Christiansen JS. Abdominal adiposity rather than age and sex predicts mass and regularity of GH secretion in healthy adults. Am J Physiol. 1997;272(6 Pt 1):E1108-1116.
  159. Clasey JL, Weltman A, Patrie J, Weltman JY, Pezzoli S, Bouchard C, Thorner MO, Hartman ML. Abdominal visceral fat and fasting insulin are important predictors of 24-hour GH release independent of age, gender, and other physiological factors. J Clin Endocrinol Metab. 2001;86(8):3845-3852.
  160. Veldhuis JD, Iranmanesh A, Ho KK, Waters MJ, Johnson ML, Lizarralde G. Dual defects in pulsatile growth hormone secretion and clearance subserve the hyposomatotropism of obesity in man. J Clin Endocrinol Metab.1991;72(1):51-59.
  161. Vijayakumar A, Novosyadlyy R, Wu Y, Yakar S, LeRoith D. Biological effects of growth hormone on carbohydrate and lipid metabolism. Growth Horm IGF Res. 2010;20(1):1-7.
  162. Newbern D, Freemark M. Placental hormones and the control of maternal metabolism and fetal growth. Current opinion in endocrinology, diabetes, and obesity. 2011;18(6):409-416.
  163. Friedrich N, Thuesen B, Jorgensen T, Juul A, Spielhagen C, Wallaschofksi H, Linneberg A. The association between IGF-I and insulin resistance: a general population study in Danish adults. Diabetes Care.2012;35(4):768-773.
  164. Johannsson G, Bengtsson BA. Growth hormone and the metabolic syndrome. J Endocrinol Invest. 1999;22(5 Suppl):41-46.
  165. Xu L, Xu C, Yu C, Miao M, Zhang X, Zhu Z, Ding X, Li Y. Association between serum growth hormone levels and nonalcoholic fatty liver disease: a cross-sectional study. PloS one. 2012;7(8):e44136.
  166. Kelijman M, Frohman LA. Enhanced growth hormone (GH) responsiveness to GH-releasing hormone after dietary manipulation in obese and nonobese subjects. J Clin Endocrinol Metab. 1988;66(3):489-494.
  167. Gram IT, Norat T, Rinaldi S, Dossus L, Lukanova A, Tehard B, Clavel-Chapelon F, van Gils CH, van Noord PA, Peeters PH, Bueno-de-Mesquita HB, Nagel G, Linseisen J, Lahmann PH, Boeing H, Palli D, Sacerdote C, Panico S, Tumino R, Sieri S, Dorronsoro M, Quiros JR, Navarro CA, Barricarte A, Tormo MJ, Gonzalez CA, Overvad K, Paaske Johnsen S, Olsen A, Tjonneland A, Travis R, Allen N, Bingham S, Khaw KT, Stattin P, Trichopoulou A, Kalapothaki V, Psaltopoulou T, Casagrande C, Riboli E, Kaaks R. Body mass index, waist circumference and waist-hip ratio and serum levels of IGF-I and IGFBP-3 in European women. Int J Obes (Lond). 2006;30(11):1623-1631.
  168. Lukanova A, Lundin E, Zeleniuch-Jacquotte A, Muti P, Mure A, Rinaldi S, Dossus L, Micheli A, Arslan A, Lenner P, Shore RE, Krogh V, Koenig KL, Riboli E, Berrino F, Hallmans G, Stattin P, Toniolo P, Kaaks R. Body mass index, circulating levels of sex-steroid hormones, IGF-I and IGF-binding protein-3: a cross-sectional study in healthy women. Eur J Endocrinol. 2004;150(2):161-171.
  169. Maccario M, Tassone F, Gauna C, Oleandri SE, Aimaretti G, Procopio M, Grottoli S, Pflaum CD, Strasburger CJ, Ghigo E. Effects of short-term administration of low-dose rhGH on IGF-I levels in obesity and Cushing's syndrome: indirect evaluation of sensitivity to GH. Eur J Endocrinol. 2001;144(3):251-256.
  170. Maccario M, Tassone F, Gianotti L, Lanfranco F, Grottoli S, Arvat E, Muller EE, Ghigo E. Effects of recombinant human insulin-like growth factor I administration on the growth hormone (gh) response to GH-releasing hormone in obesity. J Clin Endocrinol Metab. 2001;86(1):167-171.
  171. Gleeson HK, Lissett CA, Shalet SM. Insulin-like growth factor-I response to a single bolus of growth hormone is increased in obesity. J Clin Endocrinol Metab. 2005;90(2):1061-1067.
  172. Cordido F, Garcia-Buela J, Sangiao-Alvarellos S, Martinez T, Vidal O. The decreased growth hormone response to growth hormone releasing hormone in obesity is associated to cardiometabolic risk factors. Mediators Inflamm. 2010;2010:434562.
  173. Bondanelli M, Ambrosio MR, Margutti A, Franceschetti P, Zatelli MC, degli Uberti EC. Activation of the somatotropic axis by testosterone in adult men: evidence for a role of hypothalamic growth hormone-releasing hormone. Neuroendocrinology. 2003;77(6):380-387.
  174. Veldhuis JD, Keenan DM, Mielke K, Miles JM, Bowers CY. Testosterone supplementation in healthy older men drives GH and IGF-I secretion without potentiating peptidyl secretagogue efficacy. Eur J Endocrinol.2005;153(4):577-586.
  175. Utz AL, Yamamoto A, Sluss P, Breu J, Miller KK. Androgens may mediate a relative preservation of IGF-I levels in overweight and obese women despite reduced growth hormone secretion. J Clin Endocrinol Metab.2008;93(10):4033-4040.
  176. Parkinson C, Ryder WD, Trainer PJ, Sensus Acromegaly Study G. The relationship between serum GH and serum IGF-I in acromegaly is gender-specific. J Clin Endocrinol Metab. 2001;86(11):5240-5244.
  177. Lee EJ, Kim KR, Lee HC, Cho JH, Nam MS, Nam SY, Song YD, Lim SK, Huh KB. Acipimox potentiates growth hormone response to growth hormone-releasing hormone by decreasing serum free fatty acid levels in hyperthyroidism. Metabolism. 1995;44(11):1509-1512.
  178. Dieguez C, Carro E, Seoane LM, Garcia M, Camina JP, Senaris R, Popovic V, Casanueva FF. Regulation of somatotroph cell function by the adipose tissue. Int J Obes Relat Metab Disord. 2000;24 Suppl 2:S100-103.
  179. Franco C, Brandberg J, Lonn L, Andersson B, Bengtsson BA, Johannsson G. Growth hormone treatment reduces abdominal visceral fat in postmenopausal women with abdominal obesity: a 12-month placebo-controlled trial. J Clin Endocrinol Metab. 2005;90(3):1466-1474.
  180. Mekala KC, Tritos NA. Effects of recombinant human growth hormone therapy in obesity in adults: a meta analysis. J Clin Endocrinol Metab. 2009;94(1):130-137.
  181. Rasmussen MH. Obesity, growth hormone and weight loss. Mol Cell Endocrinol. 2010;316(2):147-153.
  182. Pan CS, Weiss JJ, Fourman LT, Buckless C, Branch KL, Lee H, Torriani M, Misra M, Stanley TL. Effect of recombinant human growth hormone on liver fat content in young adults with nonalcoholic fatty liver disease. Clin Endocrinol (Oxf). 2021;94(2):183-192.
  183. Wu J, Zhao F, Zhang Y, Xue J, Kuang J, Jin Z, Zhang T, Jiang C, Wang D, Liang S. Effect of One-Year Growth Hormone Therapy on Cardiometabolic Risk Factors in Boys with Obesity. Biomed Res Int. 2020;2020:2308124.
  184. Bjorntorp P, Rosmond R. Neuroendocrine abnormalities in visceral obesity. Int J Obes Relat Metab Disord.2000;24 Suppl 2:S80-85.
  185. Aldhahi W, Mun E, Goldfine AB. Portal and peripheral cortisol levels in obese humans. Diabetologia.2004;47(5):833-836.
  186. Stimson RH, Andersson J, Andrew R, Redhead DN, Karpe F, Hayes PC, Olsson T, Walker BR. Cortisol release from adipose tissue by 11beta-hydroxysteroid dehydrogenase type 1 in humans. Diabetes. 2009;58(1):46-53.
  187. Salehi M, Ferenczi A, Zumoff B. Obesity and cortisol status. Horm Metab Res. 2005;37(4):193-197.
  188. Purnell JQ, Brandon DD, Isabelle LM, Loriaux DL, Samuels MH. Association of 24-hour cortisol production rates, cortisol-binding globulin, and plasma-free cortisol levels with body composition, leptin levels, and aging in adult men and women. J Clin Endocrinol Metab. 2004;89(1):281-287.
  189. Abraham SB, Rubino D, Sinaii N, Ramsey S, Nieman LK. Cortisol, obesity, and the metabolic syndrome: a cross-sectional study of obese subjects and review of the literature. Obesity (Silver Spring). 2013;21(1):E105-117.
  190. Strain GW, Zumoff B, Strain JJ, Levin J, Fukushima DK. Cortisol production in obesity. Metabolism.1980;29(10):980-985.
  191. Strain GW, Zumoff B, Kream J, Strain JJ, Levin J, Fukushima D. Sex difference in the influence of obesity on the 24 hr mean plasma concentration of cortisol. Metabolism. 1982;31(3):209-212.
  192. Purnell JQ, Kahn SE, Samuels MH, Brandon D, Loriaux DL, Brunzell JD. Enhanced cortisol production rates, free cortisol, and 11beta-HSD-1 expression correlate with visceral fat and insulin resistance in men: effect of weight loss. Am J Physiol Endocrinol Metab. 2009;296(2):E351-357.
  193. Prodam F, Ricotti R, Agarla V, Parlamento S, Genoni G, Balossini C, Walker GE, Aimaretti G, Bona G, Bellone S. High-end normal adrenocorticotropic hormone and cortisol levels are associated with specific cardiovascular risk factors in pediatric obesity: a cross-sectional study. BMC Med. 2013;11:44.
  194. Besemer F, Pereira AM, Smit JW. Alcohol-induced Cushing syndrome. Hypercortisolism caused by alcohol abuse. Neth J Med. 2011;69(7):318-323.
  195. Newell-Price J, Trainer P, Besser M, Grossman A. The diagnosis and differential diagnosis of Cushing's syndrome and pseudo-Cushing's states. Endocr Rev. 1998;19(5):647-672.
  196. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab.2008;93(5):1526-1540.
  197. Stewart PM. 11 beta-Hydroxysteroid dehydrogenase: implications for clinical medicine. Clin Endocrinol (Oxf).1996;44(5):493-499.
  198. Anderson AJ, Andrew R, Homer NZM, Hughes KA, Boyle LD, Nixon M, Karpe F, Stimson RH, Walker BR. Effects of Obesity And Insulin on Tissue-Specific Recycling Between Cortisol And Cortisone in Men. J Clin Endocrinol Metab. 2020.
  199. Morais JBS, Severo JS, Beserra JB, de Oiveira ARS, Cruz KJC, de Sousa Melo SR, do Nascimento GVR, de Macedo GFS, do Nascimento Marreiro D. Association Between Cortisol, Insulin Resistance and Zinc in Obesity: a Mini-Review. Biol Trace Elem Res. 2019;191(2):323-330.
  200. Bujalska IJ, Kumar S, Stewart PM. Does central obesity reflect "Cushing's disease of the omentum"? Lancet.1997;349(9060):1210-1213.
  201. Hirata A, Maeda N, Nakatsuji H, Hiuge-Shimizu A, Okada T, Funahashi T, Shimomura I. Contribution of glucocorticoid-mineralocorticoid receptor pathway on the obesity-related adipocyte dysfunction. Biochem Biophys Res Commun. 2012;419(2):182-187.
  202. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest.2004;114(12):1752-1761.
  203. Hirata A, Maeda N, Hiuge A, Hibuse T, Fujita K, Okada T, Kihara S, Funahashi T, Shimomura I. Blockade of mineralocorticoid receptor reverses adipocyte dysfunction and insulin resistance in obese mice. Cardiovasc Res. 2009;84(1):164-172.
  204. Guo C, Ricchiuti V, Lian BQ, Yao TM, Coutinho P, Romero JR, Li J, Williams GH, Adler GK. Mineralocorticoid receptor blockade reverses obesity-related changes in expression of adiponectin, peroxisome proliferator-activated receptor-gamma, and proinflammatory adipokines. Circulation. 2008;117(17):2253-2261.
  205. Alfonso B, Araki T, Zumoff B. Is there visceral adipose tissue (VAT) intracellular hypercortisolism in human obesity? Horm Metab Res. 2013;45(5):329-331.
  206. Rask E, Walker BR, Soderberg S, Livingstone DE, Eliasson M, Johnson O, Andrew R, Olsson T. Tissue-specific changes in peripheral cortisol metabolism in obese women: increased adipose 11beta-hydroxysteroid dehydrogenase type 1 activity. J Clin Endocrinol Metab. 2002;87(7):3330-3336.
  207. Walker BR, Andrew R. Tissue production of cortisol by 11beta-hydroxysteroid dehydrogenase type 1 and metabolic disease. Ann N Y Acad Sci. 2006;1083:165-184.
  208. Keenan DM, Roelfsema F, Carroll BJ, Iranmanesh A, Veldhuis JD. Sex defines the age dependence of endogenous ACTH-cortisol dose responsiveness. Am J Physiol Regul Integr Comp Physiol. 2009;297(2):R515-523.
  209. Kok P, Kok SW, Buijs MM, Westenberg JJ, Roelfsema F, Frolich M, Stokkel MP, Meinders AE, Pijl H. Enhanced circadian ACTH release in obese premenopausal women: reversal by short-term acipimox treatment. Am J Physiol Endocrinol Metab. 2004;287(5):E848-856.
  210. Roelfsema F, Kok P, Frolich M, Pereira AM, Pijl H. Disordered and increased adrenocorticotropin secretion with diminished adrenocorticotropin potency in obese in premenopausal women. J Clin Endocrinol Metab.2009;94(8):2991-2997.
  211. Roelfsema F, Pijl H, Keenan DM, Veldhuis JD. Diminished adrenal sensitivity and ACTH efficacy in obese premenopausal women. Eur J Endocrinol. 2012;167(5):633-642.
  212. van den Berg G, Frolich M, Veldhuis JD, Roelfsema F. Combined amplification of the pulsatile and basal modes of adrenocorticotropin and cortisol secretion in patients with Cushing's disease: evidence for decreased responsiveness of the adrenal glands. J Clin Endocrinol Metab. 1995;80(12):3750-3757.
  213. Pasquali R, Anconetani B, Chattat R, Biscotti M, Spinucci G, Casimirri F, Vicennati V, Carcello A, Labate AM. Hypothalamic-pituitary-adrenal axis activity and its relationship to the autonomic nervous system in women with visceral and subcutaneous obesity: effects of the corticotropin-releasing factor/arginine-vasopressin test and of stress. Metabolism. 1996;45(3):351-356.
  214. Jessop DS, Dallman MF, Fleming D, Lightman SL. Resistance to glucocorticoid feedback in obesity. J Clin Endocrinol Metab. 2001;86(9):4109-4114.
  215. Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal role of the mineralocorticoid receptor in corticosteroid-induced adipogenesis. FASEB J. 2007;21(9):2185-2194.
  216. Pasquali R. The hypothalamic-pituitary-adrenal axis and sex hormones in chronic stress and obesity: pathophysiological and clinical aspects. Ann N Y Acad Sci. 2012;1264:20-35.
  217. Baid SK, Rubino D, Sinaii N, Ramsey S, Frank A, Nieman LK. Specificity of screening tests for Cushing's syndrome in an overweight and obese population. J Clin Endocrinol Metab. 2009;94(10):3857-3864.
  218. Knudsen N, Laurberg P, Rasmussen LB, Bulow I, Perrild H, Ovesen L, Jorgensen T. Small differences in thyroid function may be important for body mass index and the occurrence of obesity in the population. J Clin Endocrinol Metab. 2005;90(7):4019-4024.
  219. Asvold BO, Bjoro T, Vatten LJ. Association of serum TSH with high body mass differs between smokers and never-smokers. J Clin Endocrinol Metab. 2009;94(12):5023-5027.
  220. Biondi B. Thyroid and obesity: an intriguing relationship. J Clin Endocrinol Metab. 2010;95(8):3614-3617.
  221. Sousa PA, Vaisman M, Carneiro JR, Guimaraes L, Freitas H, Pinheiro MF, Liechocki S, Monteiro CM, Teixeira Pde F. Prevalence of goiter and thyroid nodular disease in patients with class III obesity. Arq Bras Endocrinol Metabol. 2013;57(2):120-125.
  222. Rotondi M, Leporati P, La Manna A, Pirali B, Mondello T, Fonte R, Magri F, Chiovato L. Raised serum TSH levels in patients with morbid obesity: is it enough to diagnose subclinical hypothyroidism? Eur J Endocrinol.2009;160(3):403-408.
  223. Michalaki MA, Vagenakis AG, Leonardou AS, Argentou MN, Habeos IG, Makri MG, Psyrogiannis AI, Kalfarentzos FE, Kyriazopoulou VE. Thyroid function in humans with morbid obesity. Thyroid. 2006;16(1):73-78.
  224. Oh JY, Sung YA, Lee HJ. Elevated thyroid stimulating hormone levels are associated with metabolic syndrome in euthyroid young women. Korean J Intern Med. 2013;28(2):180-186.
  225. Marzullo P, Minocci A, Tagliaferri MA, Guzzaloni G, Di Blasio A, De Medici C, Aimaretti G, Liuzzi A. Investigations of thyroid hormones and antibodies in obesity: leptin levels are associated with thyroid autoimmunity independent of bioanthropometric, hormonal, and weight-related determinants. J Clin Endocrinol Metab. 2010;95(8):3965-3972.
  226. Rotondi M, Magri F, Chiovato L. Thyroid and obesity: not a one-way interaction. J Clin Endocrinol Metab.2011;96(2):344-346.
  227. Spencer CA, Hollowell JG, Kazarosyan M, Braverman LE. National Health and Nutrition Examination Survey III thyroid-stimulating hormone (TSH)-thyroperoxidase antibody relationships demonstrate that TSH upper reference limits may be skewed by occult thyroid dysfunction. J Clin Endocrinol Metab. 2007;92(11):4236-4240.
  228. Song RH, Wang B, Yao QM, Li Q, Jia X, Zhang JA. The Impact of Obesity on Thyroid Autoimmunity and Dysfunction: A Systematic Review and Meta-Analysis. Front Immunol. 2019;10:2349.
  229. Nie X, Ma X, Xu Y, Shen Y, Wang Y, Bao Y. Characteristics of Serum Thyroid Hormones in Different Metabolic Phenotypes of Obesity. Front Endocrinol (Lausanne). 2020;11:68.
  230. Burman KD, Latham KR, Djuh YY, Smallridge RC, Tseng YC, Lukes YG, Maunder R, Wartofsky L. Solubilized nuclear thyroid hormone receptors in circulating human mononuclear cells. J Clin Endocrinol Metab.1980;51(1):106-116.
  231. De Pergola G, Ciampolillo A, Paolotti S, Trerotoli P, Giorgino R. Free triiodothyronine and thyroid stimulating hormone are directly associated with waist circumference, independently of insulin resistance, metabolic parameters and blood pressure in overweight and obese women. Clin Endocrinol (Oxf). 2007;67(2):265-269.
  232. Osburne RC, Myers EA, Rodbard D, Burman KD, Georges LP, O'Brian JT. Adaptation to hypocaloric feeding: physiologic significance of the fall in serum T3 as measured by the pulse wave arrival time (QKd). Metabolism.1983;32(1):9-13.
  233. O'Brian JT, Bybee DE, Burman KD, Osburne RC, Ksiazek MR, Wartofsky L, Georges LP. Thyroid hormone homeostasis in states of relative caloric deprivation. Metabolism. 1980;29(8):721-727.
  234. Rosenbaum M, Goldsmith R, Bloomfield D, Magnano A, Weimer L, Heymsfield S, Gallagher D, Mayer L, Murphy E, Leibel RL. Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest. 2005;115(12):3579-3586.
  235. Nannipieri M, Cecchetti F, Anselmino M, Camastra S, Niccolini P, Lamacchia M, Rossi M, Iervasi G, Ferrannini E. Expression of thyrotropin and thyroid hormone receptors in adipose tissue of patients with morbid obesity and/or type 2 diabetes: effects of weight loss. Int J Obes (Lond). 2009;33(9):1001-1006.
  236. Xia MF, Chang XX, Zhu XP, Yan HM, Shi CY, Wu W, Zhong M, Zeng HL, Bian H, Wu HF, Gao X. Preoperative Thyroid Autoimmune Status and Changes in Thyroid Function and Body Weight After Bariatric Surgery. Obes Surg. 2019;29(9):2904-2911.
  237. Granzotto PCD, Mesa Junior CO, Strobel R, Radominski R, Graf H, de Carvalho GA. Thyroid function before and after Roux-en-Y gastric bypass: an observational study. Surg Obes Relat Dis. 2020;16(2):261-269.
  238. Licenziati MR, Valerio G, Vetrani I, De Maria G, Liotta F, Radetti G. Altered Thyroid Function and Structure in Children and Adolescents Who Are Overweight and Obese: Reversal After Weight Loss. J Clin Endocrinol Metab. 2019;104(7):2757-2765.
  239. Cakir E, Sahin M, Topaloglu O, Colak NB, Karbek B, Gungunes A, Arslan MS, Unsal IO, Tutal E, Ucan B, Delibasi T. The relationship between LH and thyroid volume in patients with PCOS. J Ovarian Res.2012;5(1):43.
  240. Yasar HY, Ertugrul O, Ertugrul B, Ertugrul D, Sahin M. Insulin resistance in nodular thyroid disease. Endocr Res.2011;36(4):167-174.
  241. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569-578.
  242. Paes JE, Hua K, Nagy R, Kloos RT, Jarjoura D, Ringel MD. The relationship between body mass index and thyroid cancer pathology features and outcomes: a clinicopathological cohort study. J Clin Endocrinol Metab.2010;95(9):4244-4250.
  243. Chen J, Cao H, Lian M, Fang J. Five genes influenced by obesity may contribute to the development of thyroid cancer through the regulation of insulin levels. PeerJ. 2020;8:e9302.
  244. Chen ST, Hsueh C, Chiou WK, Lin JD. Disease-specific mortality and secondary primary cancer in well-differentiated thyroid cancer with type 2 diabetes mellitus. PloS one. 2013;8(1):e55179.
  245. Malaguarnera R, Morcavallo A, Belfiore A. The insulin and igf-I pathway in endocrine glands carcinogenesis. J Oncol. 2012;2012:635614.
  246. Kim MR, Kim SS, Huh JE, Lee BJ, Lee JC, Jeon YK, Kim BH, Kim SJ, Wang SG, Kim YK, Kim IJ. Neck circumference correlates with tumor size and lateral lymph node metastasis in men with small papillary thyroid carcinoma. Korean J Intern Med. 2013;28(1):62-71.
  247. Grani G, Lamartina L, Montesano T, Ronga G, Maggisano V, Falcone R, Ramundo V, Giacomelli L, Durante C, Russo D, Maranghi M. Lack of association between obesity and aggressiveness of differentiated thyroid cancer. J Endocrinol Invest. 2019;42(1):85-90.
  248. Matrone A, Ceccarini G, Beghini M, Ferrari F, Gambale C, D'Aqui M, Piaggi P, Torregrossa L, Molinaro E, Basolo F, Vitti P, Santini F, Elisei R. Potential Impact of BMI on the Aggressiveness of Presentation and Clinical Outcome of Differentiated Thyroid Cancer. J Clin Endocrinol Metab. 2020;105(4).
  249. Rahman ST, Pandeya N, Neale RE, McLeod DSA, Bain CJ, Baade PD, Youl PH, Allison R, Leonard S, Jordan SJ. Obesity Is Associated with BRAF(V600E)-Mutated Thyroid Cancer. Thyroid. 2020;30(10):1518-1527.
  250. Kwon H, Chang Y, Cho A, Ahn J, Park SE, Park CY, Lee WY, Oh KW, Park SW, Shin H, Ryu S, Rhee EJ. Metabolic Obesity Phenotypes and Thyroid Cancer Risk: A Cohort Study. Thyroid. 2019;29(3):349-358.
  251. Fussey JM, Beaumont RN, Wood AR, Vaidya B, Smith J, Tyrrell J. Does Obesity Cause Thyroid Cancer? A Mendelian Randomization Study. J Clin Endocrinol Metab. 2020;105(7):e2398-2407.
  252. Kaptein EM, Beale E, Chan LS. Thyroid hormone therapy for obesity and nonthyroidal illnesses: a systematic review. J Clin Endocrinol Metab. 2009;94(10):3663-3675.
  253. Pearce EN. Thyroid hormone and obesity. Current opinion in endocrinology, diabetes, and obesity.2012;19(5):408-413.
  254. Venditti P, Chiellini G, Bari A, Di Stefano L, Zucchi R, Columbano A, Scanlan TS, Di Meo S. T3 and the thyroid hormone beta-receptor agonist GC-1 differentially affect metabolic capacity and oxidative damage in rat tissues. J Exp Biol. 2009;212(Pt 7):986-993.
  255. Baxter JD, Webb P. Thyroid hormone mimetics: potential applications in atherosclerosis, obesity and type 2 diabetes. Nat Rev Drug Discov. 2009;8(4):308-320.
  256. Ladenson PW, Kristensen JD, Ridgway EC, Olsson AG, Carlsson B, Klein I, Baxter JD, Angelin B. Use of the thyroid hormone analogue eprotirome in statin-treated dyslipidemia. N Engl J Med. 2010;362(10):906-916.

 

 

 

Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes

ABSTRACT

 

While lifestyle changes such as dietary modification and increased physical activity can be very effective in improving glycemic control, over the long-term most individuals with Type 2 diabetes (T2DM) will require medications to achieve and maintain glycemic control. The purpose of this chapter is to provide the healthcare practitioner with an overview of the existing oral and injectable (non-insulin) pharmacological options available for the treatment of patients with T2DM. Currently, there are ten classes of orally available pharmacological agents to treat T2DM: 1) sulfonylureas, 2) meglitinides, 3) metformin (a biguanide), 4) thiazolidinediones (TZDs), 5) alpha glucosidase inhibitors, 6) dipeptidyl peptidase IV (DPP-4) inhibitors, 7) bile acid sequestrants, 8) dopamine agonists, 9) sodium-glucose transport protein 2 (SGLT2) inhibitors and 10) oral glucagon like peptide 1 (GLP-1) receptor agonists. In addition, glucagon like peptide 1 (GLP-1) receptor agonists, dual GLP-1 receptor and GIP receptor agonists, and amylin can be administered by injection. Medications from these distinct classes of pharmaceutical agents may be used as treatment by themselves (monotherapy) or in a combination of 2 or more drugs from multiple classes with different mechanisms of action. A variety of fixed combinations of 2 agents are available in the US and in many other countries. In this chapter we discuss the administration, mechanism of action, effect on glycemic control, other benefits, side effects, and the contraindications of the use of these glucose lowering drugs.

 

INTRODUCTION

 

While lifestyle changes such as dietary modification and increased physical activity can be very effective in improving glycemic control, over the long-term most individuals with Type 2 diabetes (T2DM ) will require medications to achieve and maintain glycemic control (1). The purpose of this chapter is to provide the healthcare practitioner with an overview of the existing oral and injectable (non-insulin) pharmacological options available for the treatment of patients with T2DM. The use of these drugs to treat diabetes during pregnancy, in children and adolescents, and for the prevention of diabetes are discussed in other Endotext chapters (2-4). For information on the management of T2DM and selecting amongst the available pharmacological agents see the chapter by Emily Schroeder in Endotext (5). 

 

Currently, there are ten classes of orally available pharmacological agents to treat T2DM: 1) sulfonylureas, 2) meglitinides, 3) metformin (a biguanide), 4) thiazolidinediones (TZDs), 5) alpha glucosidase inhibitors, 6) dipeptidyl peptidase IV (DPP-4) inhibitors, 7) bile acid sequestrants, 8) dopamine agonists, 9) sodium-glucose transport protein 2 (SGLT2) inhibitors and 10) oral glucagon like peptide 1 (GLP-1) receptor agonists (Table 1) (6-8). In addition, glucagon like peptide 1 (GLP-1) receptor agonists, dual GLP-1 receptor and GIP receptor agonists, and amylin can be administered by injection (Table 2) (6-8).  

 

Table 1. Currently Available (USA) Oral Hypoglycemic Drugs to Treat Type 2 Diabetes

General Class

Compound/Brand Name

Generic Available

Dose Range

Cost

1st Generation Sulfonylureas

Chlorpropamide/ Diabinese

Yes

100-750mg qd

Low

Tolazamide/ Tolinase

Yes

100mg qd to 500mg bid

Low

Tolbutamide/ Orinase

Yes

500mg qd to 1000mg tid with meals

Low

Acetohexamide/ Dymelor 

Yes

250mg qd to 750mg bid

Low

2nd Generation Sulfonylureas

Glyburide (Glibenclamide)/ Diabeta, Glynase

Yes

2.5mg qd to 10mg bid

Low

Glipizide/ Glucotrol, Glucotrol XL

Yes

2.5mg qd to 20mg bid

Low

Glimepiride/ Amaryl

Yes

0.5mg to 8mg qd

Low

Gliclazide/ Diamicron

Yes

40mg qd to 160mg bid

Low

Meglitinides

Repaglinide/ Prandin

Yes

0.5mg to 4 mg with meals. Max 16mg/day

Low

Nateglinide/ Starlix

Yes

60-120mg tid with meals

Low

Biguanide

Metformin/ Glucophage, Glucophage XR

Yes

500-2500mg qd or tid depending upon preparation

Low

Thiazolidinediones (TZDs)

Rosiglitazone/ Avandia

Yes

4-8mg qd

High

Pioglitazone/ Actos

Yes

15-45mg qd

Low

Alpha-glucosidase inhibitors

Acarbose/ Precose

Yes

25-100mg tid with meals

Low

Miglitol/ Glyset

Yes

25-100mg tid with meals

High

Voglibose/ Basen, Voglib

Yes

0.2mg tid with meals

 

Dipeptidyl peptidase-IV (DPP-4) inhibitors

Alogliptin/ Nesina

Yes

25mg qd

High

Linagliptin/ Tradjenta

No

5mg qd

High

Sitagliptin/ Januvia

No

25-100mg qd

High

Saxagliptin/ Onglyza

No

2.5-5mg qd

High

Vildagliptin/  Galvus

No

50mg qd

 

Bile Acid Sequestrant

Colesevelam/ Welchol

No

1875mg bid or 3.75-gram packet or bar qd

High

Dopamine Agonist

Bromocriptine/ Cycloset

No

0.8 - 4.8mg qAM

High

Sodium-glucose co-transporter-2 (SGLT2) inhibitors

Canagliflozin/ Invokana

No

100-300mg qd

High

Dapagliflozin/ Farxiga

No

5-10mg qd

High

Empagliflozin/ Jardiance

No

10-25mg qd

High

Ertugliflozin/ Stelgatro

No

5-15mg qd

High

Glucagon like peptide 1 (GLP-1) receptor agonists

Semaglutide/ Rybelsus

No

7-14mg qd

High

 

Table 2. Currently Available (USA) Injectable Hypoglycemic Drugs to Treat Type 2 Diabetes

General Class

Compound/Brand Name

Generic Available

Dose Range

Cost

GLP-1 Receptor Agonist

Exenatide/ Byetta

No

5-10mcg bid

High

Exenatide/ Bydureon

No

2mg once weekly

High

Liraglutide/ Victoza

No

0.6-1.8mg qd**

High

Albiglutide/ Tanzeum*

No

30-50mg once weekly

High

Dulaglutide/ Trulicity

No

0.75-4.5mg once weekly

High

Lixisenatide/ Adlyxin

No

10-20mcg qd

High

Semaglutide/ Ozempic

No

0.25-2.0mg once weekly

High

Dual GLP-1 Receptor/GIP Receptor Agonists

Tirzepatide/ Mounjaro

No

5mg-15mg once weekly

High

Amylin Mimetic

Pramlintide/ Symlin

No

15-120mcg tid with meals

High

*Withdrawn from market

 

Medications from these distinct classes of pharmaceutical agents may be used as treatment by themselves (monotherapy) or in a combination of 2 or more drugs from multiple classes with different mechanisms of action (6-8). A variety of fixed combination of 2 agents are available in the US and in many other countries (examples shown in Table 3). There are even combinations that contains 3 drugs (Qternmet XR which contains dapagliflozin, saxagliptin, and metformin and Trijardy XR which contains empagliflozin, linagliptin, and metformin). Additionally, there are combinations of GLP-1 receptor agonists and insulin (Table 3). These combination products may be useful and attractive to the patient, as they provide multiple drugs in a single tablet or injection, offering convenience and increased compliance. In the US, they also enable patients to receive two medications for a single medical insurance co-payment. Most importantly, the addition of a second drug results in an additive improvement in glycemic control. When a patient is on drug A if drug B is added to drug A, there is an improvement in glycemic control. This concept can be extended by the addition of a third drug C, and even a fourth drug D (Figure 1).

 

 

Table 3. Oral Pharmacological Fixed Combination Therapies to Treat Type 2 Diabetes

Drug 1

Drug 2

Brand Name

Generic

Glyburide

Metformin

Glucovance (discontinued by manufacturer: generic available)

Yes

Glipizide

Metformin

Metaglip (discontinued by manufacturer; generic available)

Yes

Glimepiride

Pioglitazone

Duetact

Yes

Glimepiride

Rosiglitazone

Avandaryl

Yes

Sitagliptin

Metformin

Janumet

No

Saxagliptin

Metformin

Kombiglyze XR

No

Pioglitazone

Metformin

ACTOSplus Met; ACTOSplus Met XR

Yes

Repaglinide

Metformin

PrandiMet

Yes

Rosiglitazone

Metformin

Avandamet

Yes

Linagliptin

Metformin

Jentadueto

No

Alogliptin

Metformin

Kazano

Yes

Alogliptin

Pioglitazone

Oseni

No

Canagliflozin

Metformin

Invokamet

No

Dapagliflozin

Metformin

Xigduo XR

No

Dapagliflozin

Saxagliptin

Qtern

No

Empagliflozin

Linagliptin

Glyxambi

No

Empagliflozin

Metformin

Synjardy

No

Ertugliflozin

Metformin

Segluromet

No

Ertugliflozin

Sitagliptin

Steglujan

No

Lixisenatide

Glargine Insulin

Soliqua

No

Liraglutide

Degludec Insulin

Xultophy

No

Figure 1. Efficacy When Oral Agents are Used as Add-On Therapy. When a patient is on drug A and they are changed to drug B, C, or D, often no improvement in glucose control will be seen. However, if drug B is added to drug A, there is an improvement. This concept can often be extended by the addition of a third drug (C), or even a fourth drug (D). There is decreasing benefit for each additional drug as the baseline A1c level decreases. Note that there is limited data on the use of 4 drug combinations.

OVERVIEW OF DRUGS

 

There are a number of different abnormalities that contribute to the hyperglycemia that occurs in patients with T2DM (9). Therefore, the drugs used to treat patients with T2DM can have a number of different mechanisms by which they lower glucose levels. Figure 2 shows the various sites of action of the pharmacological therapies for the treatment of T2DM.

Figure 2. Sites of Action of Pharmacological Therapies for the Treatment of Type 2 Diabetes.

A broad overview of the most commonly used drugs to treat T2DM is shown in Table 4 and the effect of drugs on blood lipid levels is shown in Table 5.

 

Table 4. Benefits and Side Effects of Commonly Used Drugs

Drugs

Ability to Lower Glucose

Risk of Hypoglycemia

Weight Change

Effect on ASCVD

Effect on CHF

Effect on Renal Disease

2ndGeneration SU

High

Yes

Increase

Neutral

Neutral

Neutral

Metformin

High

No

Neutral- modest weight loss

Potential Benefit

Neutral

Neutral

TZDs

High

No

Increase

Potential Benefit (Pioglitazone)

Increased

Neutral

DPP-4 inhibitors

Intermediate

No

Neutral

Neutral

Potential Increase (saxagliptin and alogliptin)

Neutral

SGLT2 inhibitors

Immediate

No

Decrease

Potential Benefit

Benefit

Benefit-

Reduced progression of renal failure

GLP-1 receptor agonists

High

No

Decrease

Benefit

Neutral-

Potential Benefit

Benefit-

Decreased proteinuria

 

Table 5. Effect of Glucose Lowering Drugs on Lipid Levels*

Metformin

Modestly decrease triglycerides and LDL-C

Sulfonylureas

No effect

DPP4 inhibitors

Decrease postprandial triglycerides

GLP1 analogues

Decrease fasting and postprandial triglycerides

Acarbose

Decrease postprandial triglycerides

Pioglitazone

Rosiglitazone

Decrease triglycerides and increase HDL-C. Small increase LDL-C but a decrease in small dense LDL

SGLT2 inhibitors

Small increase in LDL-C and HDL-C

Colesevelam

Decrease LDL-C. May increase triglycerides

Bromocriptine-QR

Decrease triglycerides

Insulin

No effect

*These effects are beyond benefits of glucose lowering

 

Bloomgarden et al reported results from a meta-regression analysis of 61 clinical trials evaluating the efficacy of the five major classes of oral anti-hyperglycemic agents (10). The results demonstrated that there is a strong direct correlation between baseline A1c level and the magnitude of the decrease in fasting glucose and A1c induced by these drugs (i.e., significantly greater reductions in both fasting plasma glucose and A1c were observed in groups with higher baseline A1c levels). Thus, expectations for the overall magnitude of effect from a given agent might be modest when treating patients whose baseline A1c is <7.5-8.0% while in patients with elevated A1c levels the effect of drug therapy may be more robust (figure 3). A separate meta-analysis of 59 clinical studies reached similar conclusions (11). These results indicate that comparing efficacies among different anti-diabetic medications is challenging, when the baseline HbA1c is different in the studies being compared.

 

Additionally, the population of patients studied can impact the efficacy of a particular class of drug. For example, patients with limited beta cell function will have a decreased response to sulfonylurea drugs as these agents work via stimulating insulin secretion by the beta cells while TZDs are most effective in patients with insulin resistance. Another example would be the decrease in efficacy of SGLT2 inhibitors lowering A1c levels in patients with decreased renal function. It is thus very difficult to compare the glucose lowering effects of different hypoglycemic drugs except in direct head-to-head comparison studies.

 

Figure 3. Relationship between baseline A1c level and the observed reduction in A1c with oral anti-hyperglycemic medications. Irrespective of drug class, the baseline glycemic control markedly influences the overall magnitude of efficacy. Data from Bloomgarden et al, Table 1 (10).

 

A recent model-based meta-analysis was used to compare glycemic control between a large number of drugs adjusted for important differences between studies, including duration of treatment, baseline A1c, and drug dosages (12). In this analysis 229 studies with 121,914 patients were utilized. Table 6 shows the estimated decrease in A1c levels for different drugs in patients that are drug naïve with an A1c of 8% and a weight of 90kg after 26 weeks of treatment. If one averages the effect on A1c of the highest doses for each drug in a specific drug class the reductions in A1c for each class of drug are metformin 1.09%, sulfonylureas 1.0%, TZDs 0.95%, DPP-4 inhibitors 0.66%, SGLT2 inhibitors 0.83%, and GLP-1 receptor agonists 1.24%. These data and the individual data for each drug in table 6 provides a rough estimate of the efficacy of various drugs and drug classes in lowering A1c levels. One should note that within a drug class there may be differences in the ability of different drugs to lower A1c levels. This is particularly true with the GLP-1 receptor agonist drugs. For additional information there is a website that provides updated comparisons of various agents to treat patients with T2DM (https://www.comparediabetesdrugs.com/). Figures 4, 5, and 6 show the effect of glucose lowering drugs on A1c levels, change in weight, and hypoglycemia (graphs from https://www.comparediabetesdrugs.com/ June 28, 2020).

 

Table 6. Estimated Efficacy of Hypoglycemic Drugs Available in US (13)

Drug

A1c % Decrease

Drug

A1c % Decrease

Metformin 2000mg

1.01

Dulaglutide 0.75

1.18

Metformin 2550mg

1.09

Dulaglutide 1.5mg

1.36

Glipizide 5-20mg

0.86

Exenatide 10ug BID

0.86

Glyburide 1.25-20mg

1.17

Exenatide 2mg QW

1.16

Glimepiride 1-8mg

0.97

Exenatide 2mg QWS

1.14

Pioglitazone 15mg

0.62

Liraglutide 0.6mg

0.88

Pioglitazone 30mg

0.85

Liraglutide 1.2mg

1.13

Pioglitazone 45mg

0.98

Liraglutide 1.8mg

1.25

Rosiglitazone 4mg

0.67

Lixisenatide 10ug

0.44

Rosiglitazone 8mg

0.91

Lixisenatide 20ug

0.66

Canagliflozin 100mg

0.84

Semaglutide 0.5mg

1.43

Canagliflozin 300mg

1.01

Semaglutide 1.0mg

1.77

Dapagliflozin 5mg

0.65

Alogliptin 12.5mg

0.58

Dapagliflozin 10mg

0.73

Alogliptin 25mg

0.66

Empagliflozin 10mg

0.69

Linagliptin 5mg

0.59

Empagliflozin 25mg

0.77

Saxagliptin 2.5mg

0.59

Ertugliflozin 5mg

0.73

Saxagliptin 5mg

0.67

Ertugliflozin 15mg

0.81

Sitagliptin 100mg

0.72

The decreases in A1c are modeled for drug naïve patients with an A1c of 8% and a weight of 90kg after 26 weeks of treatment.

 

Figure 4. The Effect of Hypoglycemic Drugs on A1c Levels

Figure 5. Change in Weight Induced by Hypoglycemic Drugs

Figure 6. Relative Risk of Hypoglycemia versus Placebo

The NIH is carrying out a study, Glycemia Reduction Approaches in Diabetes: A Comparative Effectiveness (GRADE) Study, that is randomizing approximately 5,000 patients on metformin therapy to sulfonylureas, DPP-4 inhibitors, GLP-1 receptor agonists, and insulin (13). The primary outcome is the time to primary failure defined as an A1c ≥ 7% over an anticipated mean observation period of 4.8 years (range 4-7 years). Preliminary results of this study were presented at the American Diabetes Association meeting (June 2021) and the results as expected demonstrated that the GLP-1 receptor agonist liraglutide was more effective than the sulfonylurea glimepiride and the DPP4 inhibitor sitagliptin in maintaining the A1c < 7% (GLP1 receptor agonist better than sulfonylurea better than DPP-4 inhibitor). It should be noted that the SGLT2 inhibitor and TZD drugs were not included in this study.

 

SULFONYLUREAS

 

Introduction

 

Sulfonylureas were developed in the 1950s and have been widely used in the treatment of patients with T2DM (14,15). First generation sulfonylureas (acetohexamide, chlorpropamide, tolazamide, and tolbutamide) possess a lower binding affinity for the ATP-sensitive potassium channel, their molecular target (vide infra), and thus require higher doses to achieve efficacy (see table 1) (14,15). These first-generation sulfonylureas are currently rarely used. Subsequently, in the 1980s 2nd generation sulfonylureas including glyburide (glibenclamide), glipizide, gliclazide, and glimepiride were developed and are now widely used (14). The 2nd generation sulfonylureas are much more potent compounds (~100-fold). Sulfonylureas can be used as monotherapy or in combination with any other class of oral diabetic medications except meglitinides because they lower glucose levels by a similar mechanism of action (14,16).

 

Key characteristics of the different sulfonylureas are shown in Table 7 (14). Of clinical importance is the duration of action, which varies with the rate of hepatic metabolism and the hypoglycemic activity of the metabolites. Drugs with a long duration of action are more likely to cause severe and prolonged hypoglycemia whereas short acting drugs need to be given multiple times per day (14). Additionally, drugs that are metabolized to active agents (for example glyburide) are also more likely to cause hypoglycemia (14). Most sulfonylureas are metabolized in the liver and are to some extent excreted by the kidney; therefore, hepatic and/or renal impairment increases the risk of hypoglycemia (14).

 

Table 7. Key Characteristics of Sulfonylureas

Drug

Duration of action

Metabolites

Excretion

Tolbutamide

6–12 h

Inactive

Kidney

Chlorpropamide

60 h

Active or unchanged

Kidney

Tolazamide

12–24 h

Inactive

Kidney

Glipizide

12–24 h

Inactive

Kidney 80%

Feces 20%

Glipizide ER

>24 h

Inactive

Kidney 80%

Feces 20%

Glyburide

16–24 h

Inactive or weakly active

Kidney 50%

Micronized glyburide

12-24 h

Inactive or weakly active

Kidney 50%

Feces 50%

Glimepiride

24 h

Inactive or weakly active

Kidney 60%

Feces 40%

 

Administration

 

Sulfonylureas should be taken 30 minutes before meals starting with a low dose with an increase in dosage until desired glycemic control has been achieved. In patients with a high risk of severe hypoglycemia a very low-dose can be the initial therapy while in patients with very high A1c levels one can initiate therapy at a higher dose.

 

The recommended starting dose of glipizide is 5 mg approximately 30 minutes before breakfast. Geriatric patients or those with liver or renal disease or other risk factors for severe hypoglycemia can be started on 2.5 mg. Patients with very high A1c levels may be started on a higher dose. Based on the glucose response the dose can be increased weekly by 2.5-5 mg. If a once-a-day dose is not satisfactory or the patient requires more than 15 mg per day one can give the drug before breakfast and dinner. The maximum daily dose is 40 mg per day.

 

The usual starting dose of extended-release glipizide is 5 mg per day with breakfast. Those patients who are at high risk of hypoglycemia may be started at a lower dose. The dose can be increased based on glucose or A1c measurements. The maximum dose is 20 mg per day.

 

The usual starting dose of glyburide is 2.5 to 5 mg daily with breakfast or the first main meal. Patients at high risk for hypoglycemia should be started on 1.25 mg per day. The dose should be increased weekly by 2.5 mg based on the glucose response. The maximum dose per day is 20 mg.

 

The usual starting dose of micronized glyburide is 1.5 to 3 mg daily with breakfast or the first main meal. Patients at high risk for hypoglycemia should be started on 0.75 mg per day. The dose should be increased weekly by 1.5 mg based on the glucose response. The maximum dose per day is 12 mg.

 

The recommended starting dose of glimepiride is 1 or 2 mg once daily. Patients at increased risk for hypoglycemia should be started on 1 mg once daily. The dose should be increased every 1-2 weeks in increments of 1 or 2 mg based upon the patient’s glycemic response. The maximum dose is 8 mg per day.

 

The recommended starting dose of gliclazide is 40 - 80mg once daily. Patients at increased risk for hypoglycemia should be started on 40 mg once daily. The dose should be increased every 1-2 weeks in increments of 40 or 80 mg based upon the patient’s glycemic response. The maximum dose is 160mg twice a day.

 

Mechanism of Action

 

Sulfonylureas are insulin secretagogues and lower blood glucose levels by directly stimulating glucose independent insulin secretion by the pancreatic beta cells (14,16). Through the concerted efforts of GLUT2 (the high Km glucose transporter), glucokinase (the enzyme that phosphorylates glucose), and glucose metabolism, pancreatic beta cells sense blood glucose levels and secrete the appropriate amount of insulin in response (17,18). Glucose metabolism leads to ATP generation and increases the intracellular ratio of ATP/ADP, which results in the closure of the ATP-sensitive potassium channel on the plasma membrane (14,17,19). Closure of this channel depolarizes the membrane and triggers the opening of voltage-sensitive calcium channels, leading to the rapid influx of calcium (14,20). Increased intracellular calcium causes an alteration in the cytoskeleton and stimulates translocation of insulin-containing secretory granules to the plasma membrane and the secretion of insulin (Figure 7) (14).

 

Figure 7. Mechanism by which glucose, sulfonylureas, and meglitinides stimulate insulin secretion by the beta cells.

The KATP channel is comprised of two subunits, both of which are required for the channel to be functional (20). One subunit contains the cytoplasmic binding sites for both sulfonylureas and ATP, and is designated as the sulfonylurea receptor type 1 (SUR1). The other subunit is the potassium channel, which acts as the pore-forming subunit (20). Either an increase in the ATP/ADP ratio or ligand binding by sulfonylureas or meglitinides to SUR1 results in the closure of the KATP channel and insulin secretion (15,20). Studies comparing sulfonylureas and non-sulfonylurea insulin secretagogues have identified several distinct binding sites on the SUR1 that cause channel closure. Some sites exhibit high affinity for sulfonylureas, while other sites exhibit high affinity for meglitinides.

 

In addition to binding to SUR1, sulfonylureas also bind to Epac2, a protein activated by cAMP (14). Sulfonylurea-stimulated insulin secretion was reduced both in vitro and in vivo in mice lacking Epac2, indicating that Epac2 also plays a role in sulfonylurea induced insulin secretion (21).

 

In addition to inducing insulin secretion sulfonylureas have other effects that could play a role in lowering blood glucose levels (14). Specifically, sulfonylureas have been shown to decrease hepatic insulin clearance, inhibit glucagon secretion from pancreatic alpha-cells (this may be secondary to increasing insulin secretion), and enhance insulin sensitivity in peripheral tissues (this may be partially due to lowering glucose levels and reducing glucotoxicity) (14). The contribution and importance of these additional effects in mediating the glucose lowering effects of sulfonylureas is uncertain.

 

Glycemic Efficacy

 

When used at maximally effective doses, results from well-controlled clinical trials have not indicated a marked superiority of one 2nd generation sulfonylurea over another in improving glycemic control (22). Similarly, 2nd generation sulfonylureas exhibit similar clinical efficacy compared to the 1st generation agents (22). Sulfonylureas do not have a linear dose-response relationship and the majority of the A1C reduction occurs at half maximum dosage. The effect of sulfonylureas as monotherapy or when added to metformin therapy on A1c levels varies but typically results in reductions in A1c of approximately 0.50-1.5% (12,15,16,23,24). If A1c levels are very high decreases in the range of 1.5-2.0% may be seen (15,16,22). Patients with a short duration of diabetes with residual beta cell function (high C-peptide levels) are likely to be most responsive to sulfonylurea therapy (22). Overtime many patients on sulfonylureas require additional therapies (secondary failure). In the ADOPT study, after 5 years 34% of the patients on glyburide monotherapy had fasting glucose levels > 180 mg/dl (i.e., secondary failure) (25). Similarly, in the United Kingdom Prospective Diabetes Study (UKPDS), only 34% of patients attained an A1c <7 % at 6 years treated with sulfonylureas (glyburide or chlorpropamide) and this number declined to 24 % at 9 years (14). This lack of durability of sulfonylurea therapy is likely to due to beta cell exhaustion. In addition, the weight gain induced by sulfonylurea therapy may also adversely affect glycemic control.

 

Other Effects

 

CARDIOVASCULAR DISEASE

 

Based on the University Group Diabetes Project (UGDP) all sulfonylureas carry a “black box” warning regarding cardiovascular disease (26,27). However, the U.K. Prospective Diabetes Study Group (UKPDS) studied a large number of newly diagnosed patients with T2DM at risk for cardiovascular disease. In this study improved glycemic control with sulfonylureas reduced cardiovascular disease by approximately 16%, which just missed being statistically significant (p=0.052) (28). In the UKPDS, A1c was reduced by approximately 0.9% and the 16% reduction in cardiovascular disease was in the range predicted based on epidemiological studies. Thus, the reduction in cardiovascular events was likely due to improvements in glycemic control and not a direct benefit of sulfonylurea treatment. In support of this conjecture is that in the UKPDS, insulin treatment resulted in a similar decrease in A1c levels and reduction in cardiovascular events (28). Additionally, a large randomized cardiovascular outcome study (Carolina Study) reported that linagliptin, a DPP-4 inhibitor, and glimepiride, a sulfonylurea, had similar effects on cardiovascular events (hazard ratio 0.98) (29). Taken together these results suggest that sulfonylureas have a neutral effect on cardiovascular disease.  

 

Side Effects

 

HYPOGLYCEMIA

 

The major side effect of sulfonylurea treatment is hypoglycemia, which is more likely to occur and is more severe with long- acting sulfonylureas (14,15). In the UKPDS severe hypoglycemia, defined by need for third-party assistance, occurred each year in 0.4–0.6/100 patients treated with a sulfonylurea while non-severe hypoglycemia was seen in 7.9/100 persons treated with a sulfonylurea (30). Other studies have found even higher rates of severe hypoglycemia with 20–40% of patients receiving sulfonylureas having hypoglycemia and severe hypoglycemia (requiring third-party assistance) occurring in 1–7% of patients (16,30). With continuous glucose monitoring 30% of well controlled patients with T2DM had episodes of hypoglycemia that were often asymptomatic and nocturnal (31). Of great concern these hypoglycemic events were associated with EKG changes, particularly QTc prolongation (31). Other studies have also observed a very high rate of hypoglycemia in patients with T2DM treated with sulfonylureas when monitored using continuous glucose monitoring (32).

 

Hypoglycemia typically occurs after periods of fasting or exercise. In light of this hypoglycemic risk, initiation of treatment with sulfonylureas should be at the lowest recommended dose and the dose slowly increased in patients with modestly elevated A1c levels. Older patients (> age 65) and patients with hepatic or renal disease are more likely to experience frequent and severe hypoglycemic reactions, particularly if the goals of therapy aim for inappropriately tight glycemic control  (14). Many clinicians avoid the use of long acting sulfonylureas (glyburide) in these high-risk patients as glyburide has a higher risk of hypoglycemia compared to other sulfonylureas (33).

 

WEIGHT GAIN

 

In the UKPDS, sulfonylurea treatment caused a net weight gain of approximately 3 kg, which occurred during the first 3-4 years of treatment and then stabilized (15,28). Other studies have similarly observed weight gain with sulfonylurea treatment (22).

 

FIRST GENERATION SIDE EFFECTS

 

Chlorpropamide can induce hyponatremia and water retention due to inappropriate secretion of antidiuretic hormone (ADH) (14). In addition, tolbutamide and chlorpropamide, in certain susceptible individuals, is associated with alcohol-induced flushing (14). Because of an increased risk of side effects 1st generation sulfonylureas are seldom used.

 

RARE SIDE EFFECTS

 

Intrahepatic cholestasis and allergic skin reactions, including photosensitivity and erythroderma may rarely occur (Package insert).

 

Contraindications and Drug Interactions

 

Sulfonylureas are best avoided in patients with a sulfa allergy who experienced prior severe allergic reactions (Package insert). Otherwise, cross-reactivity between antibacterial and nonantibacterial sulfonamide agents is rare.

 

In renal failure, the dose of the sulfonylurea agent will require adjustment based on glucose monitoring to avoid hypoglycemia (14). Because it is metabolized primarily in the liver without the formation of active metabolites, glipizide is the preferred sulfonylurea in patients with renal disease (34).

 

In the elderly long acting sulfonylureas, such as glyburide, glimepiride and chlorpropamide are not recommended (35).

 

Sulfonylureas can cause hemolytic anemia in patients with glucose 6-phosphate dehydrogenase (G6PD) deficiency and therefore should be used with caution in such patients (Package insert).

 

Certain drugs may enhance the glucose-lowering effects of sulfonylureas by inhibition of their hepatic metabolism (antifungals and monoamine oxidase inhibitors), displacing them from binding to plasma proteins (coumarins, NSAIDs, and sulfonamides), or inhibiting their excretion (probenecid) (16).

 

Summary

 

While the ability of sulfonylureas to improve glycemic control is robust, the risk of hypoglycemia and weight gain reduce the desirability of this drug class. Additionally, the shorter durability of effectiveness is also a limiting factor. In patients at high risk for the occurrence of severe hypoglycemic reactions or in obese patients, using drugs other than sulfonylureas to treat T2DM is indicated if possible. Similarly, in patients with atherosclerotic cardiovascular disease or at high risk for cardiovascular disease or renal disease other hypoglycemic drugs have advantages. Nevertheless, because sulfonylureas are generic drugs and very inexpensive, they continue to be widely used and play a role in the management of patients with T2DM.

 

Table 8. Summary of the Advantages and Disadvantages of Sulfonylureas

Advantages

Disadvantages

Inexpensive

Hypoglycemia

Rapid acting

Weight gain

Once a day administration possible

Limited durability

Long history of use

Need to titrate dose

 

MEGLINATIDES

 

Introduction

 

The meglitinides are non-sulfonylurea insulin secretagogues characterized by a very rapid onset and abbreviated duration of action (16,36). Repaglinide (Prandin), a benzoic acid derivative introduced in 1998, was the first member of the meglitinide class. Nateglinide (Starlix) is a derivative of the amino acid D-phenylalanine and was introduced to the market in 2001. Unlike sulfonylureas, repaglinide and nateglinide stimulation of insulin secretion is dependent on the presence of glucose (36,37). As glucose levels decrease, insulin secretion decreases, which reduces the risk of hypoglycemia compared with sulfonylureas.

 

Meglitinides are rapidly absorbed with maximum serum concentrations generally attained within 1 hour and then quickly metabolized by the liver cytochrome CYP3A4 and CYP2C8 pathways, producing inactive metabolites, resulting in a plasma half-life of around 1 h (16). This rapid onset and short duration of action results in the ability of this class of drugs to predominantly reduce postprandial glucose levels (36). Because of the rapid onset and short duration of action meglitinides are given 1-30 minutes prior to meals. The drug should not be administered if the patient is going to skip the meal. 

 

The pharmacokinetics of meglitinides differ with nateglinide having a faster onset and shorter duration of action than repaglinide (37). Nateglinide stimulates early insulin release faster and to a greater extent than repaglinide with insulin levels returning to baseline levels more rapidly (36,37).

 

Administration

 

The recommended starting dose of nateglinide is 120 mg three times per day before meals (1-30 minutes). In patients who are near their glycemic goal when treatment is initiated the recommended starting dose of nateglinide is 60 mg three times per day before meals. The maximum dose of nateglinide is 120 mg three times per day before meals.

 

The recommended starting dose of repaglinide for patients whose A1c is less than 8% is 0.5 mg before each meal (1-30 minutes). For patients whose A1c is 8% or greater the starting dose is 1 or 2 mg orally before each meal. The patient’s dose should be doubled up to 4mg with each meal until satisfactory glycemic control is achieved (should wait one week between increasing dose). The maximum daily dose is 16 mg per day.

 

Mechanism of Action

 

Meglitinides bind to a different site on SUR1 in β cells that is separate from the sulfonylurea binding site (Figure 7) (16,36). The effect of meglitinide binding is similar to the effect of sulfonylureas binding resulting in the closure of the KATP channel leading to cell depolarization and calcium influx resulting in insulin secretion (16,36,37). However, the relatively rapid onset and short duration of action of meglitinides suits their use as prandial glucose-lowering agents (16,36).

 

Glycemic Efficacy

 

Studies have shown that A1c reductions are similar to, or slightly less, than those observed with sulfonylurea or metformin treatment when meglitinides are used as monotherapy (16,36). In studies comparing repaglinide monotherapy with sulfonylurea or metformin therapy the decrease in A1c was similar (36,38). In contrast, a study comparing nateglinide with metformin demonstrated that metformin was more effective in lowering A1c levels (39). In a randomized trial comparing repaglinide and nateglinide in patients with T2DM previously treated with diet and exercise, repaglinide was more effective in lowering A1c levels (1.57% vs. 1.04%) (40). While postprandial glucose levels were similar repaglinide was more effective in reducing fasting glucose levels, probably due to its longer duration of action. These clinical findings can be incorporated into clinical decision making.  For example, if the main issue for the patient is postprandial hyperglycemia, and fasting glucoses are near normal, an agent, such as nateglinide, that has a limited effect on the fasting glucose would be ideal. However, if one needs reductions in both fasting and postprandial glucose levels a longer acting agent such as repaglinide is a better choice.

 

Other Effects

 

CARDIOVASCULAR DISEASE

 

The Navigator study was a double-blind, randomized clinical trial in 9,306 individuals with impaired glucose tolerance and either cardiovascular disease or cardiovascular risk factors who received nateglinide (up to 60 mg three times daily) or placebo (41). After 5 years, nateglinide administration did not alter the incidence of cardiovascular outcomes suggesting that meglitinides do not have adverse or beneficial cardiovascular effects.

 

Side Effects

 

Similar to sulfonylureas, meglitinides can cause hypoglycemia but the risk of severe hypoglycemia is less (16,36,38). The incidence of hypoglycemia is lower with nateglinide than for repaglinide and nateglinide is less likely to cause severe hypoglycemia (16). In one study, the occurrence of symptomatic hypoglycemia was 2% for nateglinide and 7% for repaglinide (37). Weight gain is also a common side effect of meglitinides (approximately 1-3 kg) with nateglinide leading to less weight gain than repaglinide (16,37).

 

Contraindications and Drug Interactions

 

Because meglitinides are metabolized by the liver these drugs should be used cautiously in patients with impaired liver function (Package insert).

 

Drugs that inhibit CYP3A4 (for example ketoconazole, itraconazole and erythromycin) or CYP2C8 (for example trimethoprim, gemfibrozil and montelukast) can result in the increased activity of meglitinides enhancing the risk of hypoglycemia and should be avoided if possible (38).

 

Summary

 

Meglitinides can be useful drugs when there is a need to specifically lower postprandial glucose levels (i.e., patients with fasting glucose in desired range but elevated post meal glucose levels). Additionally, because of their short duration of action meglitinides can be useful in patients who eat erratically as this class of drugs can be given only before meals and the duration of action will match the postprandial increase in glucose. The risk of severe hypoglycemia and weight gain is less than sulfonylureas but still must be considered in patients treated with meglitinides. The development of drugs that do not cause weight gain or severe hypoglycemia and lower postprandial glucose levels have resulted in the limited use of meglitinides.

 

Table 9. Summary of the Advantages and Disadvantages of Meglitinides

Advantages

Disadvantages

Decrease postprandial glucose

Hypoglycemia

Flexible dosing

Weight gain

Relatively inexpensive

Frequent dosing

Short action allowing for missing meals

Need to titrate dose

 

METFORMIN

 

Introduction

 

Metformin (Glucophage) is a synthetic analog of the natural product guanidine (16). Since its initial clinical use over 50 years ago, metformin has surpassed the sulfonylureas as the most widely prescribed oral agent for T2DM throughout the world because of its proven efficacy on glycemic control as monotherapy and in combination with many other available agents (16). The widespread acceptance of metformin evolved after the realization that lactic acidosis was not a major problem in individuals with normal renal function. Phenformin, a structural analog of metformin, was previously withdrawn from the market in many countries due its propensity to induce lactic acidosis (16).

 

Administration

 

The usual starting dose of metformin is 500 mg twice a day with meals. After 1-2 weeks the dose can be increased to 1500 mg per day (750 mg twice a day or 500 mg in AM and 1000 mg in PM). After another 1-2 weeks the dose can be increased to 1000 mg twice a day. The slow increase in dosage is to reduce GI side effects and the dose should not be increased if GI side effects are occurring. The maximum dose is 2550 mg per day which can be given as 850 mg three times per day with meals but most patients are treated with 1000 mg twice a day with breakfast and dinner.

 

The usual starting dose of metformin extended release is 500 mg with the evening meal (largest meal). The dose can be increased by 500 mg weekly depending upon tolerability. The maximum dose is 2000 mg with the evening meal.

 

Note the dose of metformin may need to be adjusted based on renal function (discussed below).

 

Metformin should be temporarily discontinued when patients are unable to eat or drink. Metformin is seldom used in hospitalized patients.

 

Mechanism of Action

 

Metformin decreases hepatic glucose production and improves hepatic insulin sensitivity but has only a modest impact on peripheral insulin-mediated glucose uptake (i.e., insulin resistance), which is likely due to a reduction in hyperglycemia, triglycerides, and free fatty acid levels (42,43). Hyperinsulinemia is reduced and the decrease in hepatic glucose production results in a decrease in fasting glucose levels (16). In addition, metformin also increases intestinal glucose utilization and stimulates GLP-1 secretion (42,43). Insulin secretion is not increased (16). The cellular and molecular mechanisms that account for these changes are not definitively understood.

 

LIVER

 

There are several lines of evidence indicating that the liver plays an important role in metformin’s ability to improve glycemic control (42). In humans and rodents, metformin is concentrated in the liver and blocking the uptake of metformin into the liver in mice prevents the ability of metformin to lower blood glucose levels (42,43). As noted above tracer studies in humans show that metformin lowers hepatic glucose production and increases hepatic insulin sensitivity (42). There are a number of proposed mechanisms by which metformin alters hepatic metabolism (42).

 

  • Metformin inhibits mitochondrial ATP production by inhibition of Complex I of the respiratory chain and/or inhibiting mitochondrial glycerophosphate dehydrogenase, which is required to carry reducing equivalents from the cytoplasm into the mitochondria for re-oxidation (42,43). The decrease in ATP production could decrease hepatic gluconeogenesis (43). This also leads to an increase in AMP.
  • Metformin increases hepatic AMP levels and AMP is a potent allosteric inhibitor of fructose 1,6-bisphosphatase, a key enzyme in gluconeogenesis (43). In addition, high AMP levels inhibit adenylate cyclase reducing cyclic AMP formation in response to glucagon, which also decreases glycogenolysis and gluconeogenesis (i.e., decreases glucagon activity) (43). The increase in AMP also activates AMP-activated protein kinase.
  • Metformin activates AMP-activated protein kinase, which activates catabolic pathways leading to decreased gluconeogenesis, decreased fatty acid synthesis, and increased fatty acid oxidation (42,43). The changes in fatty acid metabolism are thought to account for the improvement in hepatic insulin sensitivity and the decrease in serum triglyceride levels (42).
  • Metformin inhibits glycerol-3-phosphate dehydrogenase increasing the cytosolic redox state resulting in a decreased conversion of glycerol and lactate to glucose (44).

 

INTESTINE

 

Several lines of evidence indicate that the intestine plays an important role in explaining metformin’s ability to lower blood glucose levels. First, a decrease in hepatic glucose production can only partially account for the decrease in blood glucose (42). Second, in humans with loss-of-function variants in SLC22A1, which decrease the uptake of metformin into the liver, the ability of metformin to lower A1c levels is not impaired (42). Finally, a delayed-release metformin that is retained in the gut, with minimal systemic absorption, is as effective at lowering blood glucose as the standard metformin formulation in patients with T2DM (42,45). There are a number of proposed mechanisms for how the intestine accounts for the beneficial effects of metformin.

 

  • Metformin increases anaerobic glucose metabolism in the intestine resulting in increased intestinal glucose utilization and decreased glucose uptake into the circulation (42). This is likely due to the inhibition of mitochondrial ATP production described above. The increased utilization of glucose by anaerobic metabolism could contribute to metformin induced weight loss.
  • Metformin increases GLP-1 secretion, which could increase insulin secretion and decrease glucagon secretion (42). The increase in GLP-1 could also contribute to the weight loss or weight neutral effects of metformin.
  • Metformin alters the intestinal microbiome, which could alter glucose metabolism (42,46).

 

It is clear that there are multiple potential mechanisms by which metformin can improve glucose metabolism and further studies are required to elucidate the relative importance and contribution of these proposed mechanisms and others yet to be identified. 

 

Glycemic Efficacy

 

Metformin is often used as the initial therapy in patients with diabetes in conjunction with lifestyle changes (6,7). The typical reduction in A1c with metformin therapy is in the range of 1 to 2.0% (16,47). The decrease in A1c induced by metformin is independent of age, weight, and diabetes duration as long as some residual β-cell function remains (16). One retrospective study has reported that African-Americans have a greater decrease in A1c with metformin compared to Caucasians (48). The effect of immediate release and extended release metformin on A1c levels is similar (49). In head-to-head trials, metformin has been shown to produce equivalent reductions in A1c as sulfonylureas and thiazolidinediones but is more potent than DPP-4 inhibitors (47).

 

The durability of glycemic control with metformin is more prolonged than with sulfonylureas but shorter than with TZDs (25). After 5 years of monotherapy, 15% of individuals on rosiglitazone therapy, 21% of individuals on metformin therapy, and 34% of individuals on glyburide (glibenclamide) therapy had fasting glucose levels above the acceptable range (25). The ability to maintain an A1c <7% was 57 months with rosiglitazone, 45 months with metformin, and 33 months with glyburide (glibenclamide) (25).

 

In addition to the ability to improve glycemic control in monotherapy, metformin in combination with sulfonylureas, meglinitides, TZDs, DPP-4 inhibitors, SGLT-2 inhibitors, insulin, and GLP-1 receptor agonists lowers A1c levels and often allows for patients to achieve their A1c goals (47). As shown in Table 3 there are a large number of combination tablets that include metformin with other glucose lowering drugs.

 

Hypoglycemia does not occur with metformin monotherapy (47). Hypoglycemia may occur with metformin during concomitant use with other glucose-lowering agents such as sulfonylureas and insulin.

 

Other Effects

 

WEIGHT

 

Metformin is weight neutral or can sometimes result in a modest weight loss (up to 4 kg) (47). When used in combination with sulfonylureas or insulin it blunts the weight gain induced by these agents.

 

LIPIDS

 

Metformin decreases serum triglyceride levels and LDL-C levels without altering HDL-C (50,51). In a meta-analysis of 37 trials with 2,891 patients, metformin decreased triglycerides by 11.4mg/dl when compared with control treatment (p=0.003) (50). In an analysis of 24 trials with 1,867 patients, metformin decreased LDL-C by 8.4mg/dl compared to control treatment (p<0.001) (50). In contrast, metformin did not significantly alter HDL-C levels (50). It should be noted that in the Diabetes Prevention Program 3,234 individuals with impaired glucose metabolism were randomized to placebo, intensive lifestyle, or metformin therapy (52). In the metformin therapy group no significant changes were noted in triglyceride, LDL-C, or HDL-C levels compared to the placebo group. Thus, metformin may have small effects on lipid levels.     

 

CARDIOVASCULAR DISEASE

 

In the UKPDS, metformin, while producing a similar improvement in glycemic control as insulin or sulfonylureas, markedly reduced cardiovascular disease by approximately 40% (53). In the ten-year follow-up the patients randomized to metformin in the UKPDS continued to show a reduction in MI and all-cause mortality (54). Two other small randomized controlled trials have also demonstrated cardiovascular benefits with metformin therapy. A study by Kooy et al compared the effect of adding metformin or placebo in overweight or obese patients already on insulin therapy (55). After a mean follow-up of 4.3 years this study observed a reduction in macrovascular events (HR 0.61 CI- 0.40-0.94, p=0.02), which was partially accounted for by metformin’s beneficial effects on weight. In this study the difference in A1c between the metformin and placebo group was only 0.3%. Hong et al randomized non-obese patients with coronary artery disease to glipizide vs. metformin therapy for three years (56). A1c levels were similar, but there was a marked reduction in cardiovascular events in the metformin treated group (HR 0.54 CI 0.30- 0.90, p=0.026). These results suggest that metformin may reduce cardiovascular disease and that this effect is not due to improving glucose control. Metformin decreases weight or prevents weight gain and lowers lipid levels and these or other non-glucose effects may account for the beneficial effects on cardiovascular disease. Larger cardiovascular outcome studies are required to definitively demonstrate a beneficial effect of metformin on cardiovascular disease.

 

POLYCYSTIC OVARY SYNDROME (PCOS)

 

In patients with PCOS metformin lowers serum androgen levels, increases ovulations, and improves menstrual frequency (57). Metformin may also be associated with weight loss in some women with PCOS (57). Metformin combined with clomiphene may be the best combination in obese women with PCOS to improve fertility (57). For a detailed discussion of the treatment of PCOS see the chapter on polycystic ovary syndrome in Endotext (57). 

 

CANCER

 

Multiple epidemiological studies have demonstrated an association between metformin treatment and a reduced cancer incidence and mortality (58,59). Treatment with metformin has been associated with a decreased risk of breast, colon, liver, pancreas, prostate, endometrium and lung cancer and marked reductions in cancer-specific mortality for colon, lung and early-stage prostate cancer and improvements in survival for breast, colon, endometrial, ovarian, liver, lung, prostate and pancreatic cancer (58,59). A wide variety of different mechanisms have been proposed that could account for metformin’s anti-tumor effects providing biological plausibility (59). However, data from large randomized controlled trials have not yet definitively demonstrated whether metformin can prevent the development of cancer or is useful in the treatment of cancer (58-60). Further studies are required to elucidate the potential role of metformin in oncology.

 

Side Effects

 

GASTROINTESTINAL

 

The most common side effects of metformin are diarrhea, nausea, and/or abdominal discomfort, which can occur in up to 50% of patients (16,47). These side effects are usually mild and disappear with continued drug administration. The GI side effects are dose-related and slow titration to allow for tolerance can reduce the occurrence of these symptoms (47). Administrating metformin three times a day with meals instead of twice a day may also reduce GI side effects. A small number of patients cannot tolerate the drug, even at low doses (47). Extended-release metformin [metformin XR]) causes fewer GI symptoms and can be used in patients who do not tolerate immediate release metformin (47).

 

Studies have shown that reduced function of plasma membrane monoamine transporter or organic cation transporter 1 leads to an increase in metformin GI side effects (61,62). Use of drugs that inhibit organic cation transporter 1 activity (including tricyclic antidepressants, citalopram, proton-pump inhibitors, verapamil, diltiazem, doxazosin, spironolactone, clopidogrel, rosiglitazone, quinine, tramadol and codeine) increased intolerance to metformin (61).

 

LACTIC ACIDOSIS

 

A very rare complication of metformin therapy is lactic acidosis (47). This complication was much more common with phenformin therapy, the initial biguanide, and the risk with metformin is estimated to be 20 times less (47). The estimated incidence of metformin-associated lactic acidosis is 3–10 per 100,000 person-years (47). This is a potentially lethal complication of metformin therapy that typically occurs when renal dysfunction results in very high blood metformin levels, which inhibit mitochondrial function resulting in the overproduction of lactate (47). In addition to renal disorders other risk factors for metformin associated lactic acidosis include sepsis, cardiogenic shock, hepatic impairment, congestive heart failure, and alcoholism (47). In some circumstances the lactic acidosis observed in patients treated with metformin may not be due to metformin but rather to underlying clinical disorders such as severe sepsis.

 

VITAMIN B12 DEFICIENCY

 

Studies have demonstrated that vitamin B12 malabsorption is a side effect of metformin therapy (47). A randomized controlled trial showed that metformin 850 mg three times per day for over 4 years resulted in a 19% decrease in B12 levels compared to placebo (63). Moreover, 9.9% of patients treated with metformin developed vitamin B12 deficiency (<150 pmol/l) vs only 2.7% in the placebo group (63). The Diabetes Prevention Program Outcomes Study also demonstrated an increased risk of B12 deficiency with long term metformin use (64). It is now recommended that periodic testing of vitamin B12 levels should be considered in patients on long-term metformin therapy, particularly in the setting of anemia or neuropathy (65). 

 

OVULATION AND PREGNANCY

 

As discussed above in the polycystic ovary section, treatment of premenopausal women with PCOS with metformin may induce ovulation and thereby result in unplanned pregnancies. In premenopausal anovulatory women started on metformin one needs to discuss the need for contraception.

 

Contraindications and Drug Interactions

 

Metformin is contraindicated in patients with advanced kidney or liver disease, acute unstable congestive heart failure, conditions marked by decreased perfusion or hemodynamic instability, major alcohol abuse, or conditions characterized by acidosis (47). Metformin therapy should be suspended during serious illness or surgical procedures. Metformin is seldom used in hospitalized patients.

 

RENAL DISEASE

 

A major contraindication to the use of metformin is renal disease (47). Metformin is not metabolized and is excreted intact by the kidneys and therefore kidney function is a major determinant of blood metformin levels. eGFR should be obtained prior to initiating therapy. In patients with renal dysfunction or at risk for developing renal dysfunction eGFR should be obtained more frequently. In patients with a eGFR < 30 mL/min/1.73 m2 metformin therapy is contraindicated (47). In patients with an eGFR between 30-60mL/min/1.73 m2 metformin can be used but one should consider using lower doses (47). In patients with eGFR < 45mL/min/1.73 m2 the author typically uses ½ the maximal dose of metformin. In patients with labile renal disease, especially if frequent deteriorations in kidney function occur, metformin is best avoided.

 

IODINATED CONTRAST STUDIES

 

FDA guidelines indicate that metformin use should be withheld before iodinated contrast procedures if a) the eGFR is 30–60 mL/min/1.73 m2, b) in the setting of liver disease, alcoholism, or heart failure, or c) if intra-arterial contrast is used. The eGFR should be checked 48 hours later and metformin restarted if renal function remains stable.

 

DRUG INTERACTIONS

 

Carbonic   anhydrase   inhibitors, such as topiramate or acetazolamide, can decrease serum bicarbonate levels and induce a non-anion gap, hyperchloremic metabolic acidosis. Concomitant use of these drugs with metformin may increase the risk for lactic acidosis (Package Insert).

 

Certain drugs, such as ranolazine, vandetanib, dolutegravir, and cimetidine, may interfere with common renal tubular transport systems that are involved in the renal elimination of metformin and therefore can increase systemic exposure to metformin and may increase the risk for lactic acidosis (Package Insert).

 

Summary

 

Metformin is a commonly used as a the first drug for the treatment of diabetes because of excellent efficacy, an outstanding safety profile, low cost, and a long history of use without significant problems. 

 

Table 10. Summary of the Advantages and Disadvantages of Metformin

Advantages

Disadvantages

Inexpensive

GI side effects

No hypoglycemia

B12 deficiency

Once a day administration possible

Lactic acidosis (very rare)

Long history of use

Need to monitor renal function

No weight gain and maybe weight loss

 

May decrease cardiovascular disease

 

 

THIAZOLIDINEDIONES (TZDS)

 

Introduction

 

Troglitazone (Rezulin), pioglitazone (Actos), and rosiglitazone (Avandia) are members of the thiazolidinedione (TZD) class of insulin sensitizing compounds that activate PPAR gamma (16,66). Troglitazone was withdrawn from the US, European, and Japanese markets in 2000 due to an idiosyncratic hepatic reaction leading to hepatic failure and death in some patients (16,66). This idiosyncratic hepatic reaction has not occurred with pioglitazone or rosiglitazone (66). TZDs decrease insulin resistance and thereby enhance the biological response to endogenously produced insulin, as well as exogenous insulin (66).

 

Administration

 

Initiate pioglitazone at 15 mg or 30 mg once a day with or without food. Use 15mg in patients where there is concern of fluid retention. If there is inadequate glycemic control, the dose can be increased in 15 mg increments up to a maximum of 45 mg once daily.

 

Initiate rosiglitazone at 4 mg once a day with or without food. If there is inadequate glycemic control, the dose can be increased to a maximum of 8 mg once daily.

 

Because the maximum effect of TZDs on glycemic control may take 10-14 weeks one should wait 12 weeks before deciding whether to increase the dose of TZDs.

 

Mechanism of Action

 

The primary effect of pioglitazone and rosiglitazone is the reduction of insulin resistance resulting in an improvement of insulin sensitivity (16,66,67). Pioglitazone and rosiglitazone are selective agonists for the PPAR gamma receptor, a member of the super-family of nuclear hormone receptors that function as ligand-activated transcription factors (66,67). In the absence of ligand, PPARs bind as hetero-dimers with the 9-cis retinoic acid receptor (RXR) and a multi-component co-repressor complex to a specific response element (PPRE) within the promoter region of their target genes (66,67). Once PPAR gamma is activated by ligand, the co-repressor complex dissociates allowing the PPAR-RXR heterodimer to associate with a multi-component co-activator complex resulting in an increased rate of gene transcription (66,67). Additionally, PPAR gamma can repress target gene expression by negative feedback on other signal transduction pathways, such as the nuclear factor kB (NF-kB) signaling pathway, in a DNA binding independent manner (66). The target genes of PPAR gamma include those involved in the regulation of lipid and carbohydrate metabolism and inflammation (66,67).

 

PPAR gamma is highly expressed in adipose tissue while its expression in skeletal muscle is low (66,67). In the liver PPAR gamma expression is low but increases in obesity and thus in obese individuals it is possible that TZDs directly affect the liver (68). It is likely that the primary effects of TZDs are on adipose tissue, followed by secondary benefits on other target tissues of insulin (66). TZDs promote fatty acid uptake and storage in adipose tissue resulting in a decrease in circulating fatty acids and a decrease in fat accumulation in liver, muscle and pancreas leading to the protection of these tissues from the harmful metabolic effects of higher levels of fatty acids (16,66). This decrease in fat accumulation in liver and muscle leads to an improvement in insulin action and the decrease in the pancreas may improve insulin secretion. Additionally, PPAR gamma agonists increase the expression and circulating levels of adiponectin, an adipocyte-derived protein with insulin sensitizing activity (66). A decrease in the gene expression of other adipokines involved in induction of insulin resistance, such as TNF-alpha, resistin, etc. are likely to also contribute to the improvement in insulin resistance that occurs with TZDs (66). Finally, the activation of PPAR gamma in other tissues may contribute to the beneficial effects of TZDs.

 

Glycemic Efficacy

 

Pioglitazone and rosiglitazone decrease A1c levels to a similar degree as metformin and sulfonylurea therapy (typically a 1.0-1.5% decrease in A1c) (16,66). The decreases in fasting plasma glucose were observed as early as the second week of therapy but maximal decreases occurred after 10-14 weeks (16,69). This differs from other hypoglycemic drugs where the maximal effect occurs more rapidly. TZDs lower both fasting and postprandial glucose levels (66). TZDs are more effective in improving glycemic control in patients with marked insulin resistance (70).

 

TZDs are effective in combination with other hypoglycemic drugs including insulin (16,37,69). TZDs do not cause hypoglycemia when used as monotherapy or in combination with metformin (16,37). In combination with insulin or insulin secretagogues, TZDs can potentiate hypoglycemia. If hypoglycemia occurs one needs to adjust the dose of insulin or insulin secretagogues.

 

The durability of glycemic control with TZDs is more prolonged than with either sulfonylureas or metformin (18). After 5 years of monotherapy, 15% of individuals on rosiglitazone, 21% of individuals on metformin, and 34% of individuals on glyburide (glibenclamide) had fasting glucose levels above the acceptable range (18). The ability to maintain an A1c <7% was 57 months with rosiglitazone, 45 months with metformin, and 33 months with glyburide (glibenclamide) (18). Similar results were observed when pioglitazone therapy was compared to sulfonylurea therapy (71). After 2-years of therapy 47.8% of pioglitazone-treated patients and only 37.0% of sulfonylurea-treated patients maintained an A1c <8%. Studies have shown that TZDs improve and preserve beta cell function, which may account for their better durability (72-74).

 

Other Beneficial Effects

 

PROTEINURIA

 

A meta-analysis of 15 studies (5 with rosiglitazone and 10 with pioglitazone) involving 2,860 patients demonstrated that TZDs decreased urinary albumin excretion in patients without albuminuria, in patients with microalbuminuria, and in patients with proteinuria (75).

 

BLOOD PRESSURE

 

TZDs modestly lower BP. In a review of 37 studies TZDs lowered systolic BP by 4.70 mm Hg and diastolic BP by 3.79 mm Hg (76).  

 

LIPIDS

 

The effect of TZDs on lipids depends on which agent is used. Rosiglitazone increases serum LDL cholesterol levels, increases HDL cholesterol levels, and only decreases serum triglycerides if the baseline triglyceride levels are high [66]. In contrast, pioglitazone has less impact on LDL cholesterol levels, but increases HDL cholesterol levels, and decreases serum triglyceride levels (77). In the PROactive study, a large randomized cardiovascular outcome study, pioglitazone decreased triglyceride levels by approximately 10%, increased HDL-C levels by approximately 10%, and increased LDL-C by 1-4% (78). It should be noted that reductions in the small dense LDL subfraction and an increase in the large buoyant LDL subfraction are seen with both TZDs (77). Treatment with pioglitazone for 12 weeks resulted in a significant increase in the ability of HDL to facilitate the efflux of cholesterol from cells (79).

 

In a randomized head-to-head trial, it was shown that pioglitazone decreased serum triglyceride levels and increased serum HDL cholesterol levels to a greater degree than rosiglitazone treatment (80,81). Additionally, pioglitazone increased LDL cholesterol levels less than rosiglitazone. In contrast to the differences in lipid parameters, both rosiglitazone and pioglitazone decreased A1c and C-reactive protein to a similar extent. The mechanism by which pioglitazone induces more favorable changes in lipid levels than rosiglitazone is unclear, but differential actions of ligands for nuclear hormone receptors are well described.

 

CARDIOVASCULAR DISEASE

 

Studies with pioglitazone have suggested a beneficial effect on cardiovascular disease. The PROactive study was a randomized controlled trial that examined the effect of pioglitazone vs. placebo over a 3-year period in patients with T2DM and pre-existing macrovascular disease (82). With regard to the primary endpoint (a composite of all-cause mortality, non-fatal myocardial infarction including silent MI, stroke, acute coronary syndrome, endovascular or surgical intervention in the coronary or leg arteries, and amputation above the ankle), there was a 10% reduction in events in the pioglitazone group but this difference was not statistically significant (p=0.095). It should be noted that both leg revascularization and leg amputations are not typical primary end points in cardiovascular disease trials and these could be affected by pioglitazone induced edema. When one focuses on standard cardiovascular disease endpoints, the pioglitazone treated group did demonstrate a 16% reduction in the main secondary endpoint (composite of all-cause mortality, non-fatal myocardial infarction, and stroke) that was statistically significant (p=0.027). In the pioglitazone treated group, blood pressure, A1c, triglyceride, and HDL cholesterol levels were all improved compared to the placebo group making it very likely that the mechanism by which pioglitazone decreased vascular events was multifactorial.

 

The IRIS trial was a multicenter, double-blind trial that randomly assigned 3,876 patients with insulin resistance but without diabetes and a recent ischemic stroke or TIA to treatment with either pioglitazone or placebo (83). After 4.8 years, the primary outcome of fatal or nonfatal stroke or myocardial infarction occurred in 9.0% of the pioglitazone group and 11.8% of the placebo group (hazard ratio 0.76; P=0.007). All components of the primary outcome were reduced in the pioglitazone treated group. Additionally, in the subgroup of patients with “prediabetes” pioglitazone therapy also reduced cardiovascular events (84). Fasting glucose, fasting triglycerides, and systolic and diastolic blood pressure were lower while HDL cholesterol and LDL cholesterol levels were higher in the pioglitazone group than in the placebo group. Although this study excluded patients with diabetes the results are consistent with and support the results of a protective effect of pioglitazone observed in the PROactive study.

 

In contrast to the above results, a study compared the effect of pioglitazone vs. sulfonylurea on cardiovascular disease and did not observe a reduction in events with pioglitazone treatment (TOSCA.IT) (85). Patients with T2DM (n= 3,028), inadequately controlled with metformin monotherapy (2-3 g per day), were randomized to pioglitazone or sulfonylurea and followed for a median of 57 months. Only 11% of the participants had a previous cardiovascular event. The primary outcome was a composite of first occurrence of all-cause death, non-fatal myocardial infarction, non-fatal stroke, or urgent coronary revascularization and occurred in 6.8% of the patients treated with pioglitazone and 7.2% of the patients treated with a sulfonylurea (HR 0.96; NS). Limitations of this study are the small number of events likely due to low-risk population studied and the relatively small number of participants. Additionally, 28% of the subjects randomized to pioglitazone prematurely discontinued the medication. Thus, the results of this study should be interpreted with caution. Additionally, it should be noted that when patients in this study were analyzed based on the risk of developing cardiovascular disease those at high risk had a marked reduction in events when treated with pioglitazone compared to the sulfonylurea (86).

 

Further support for the beneficial effects of pioglitazone on atherosclerosis is provided by studies that have examined the effect of pioglitazone on carotid intima-medial thickness. Both the Chicago and Pioneer studies demonstrated favorable effects on carotid intima-medial thickness in patients treated with pioglitazone compared to patients treated with sulfonylureas (87,88). Additionally, in patients with “prediabetes” pioglitazone also slowed the progression of carotid intima-medial thickness (89). Similarly, Periscope, a study that measured atheroma volume by intravascular ultrasonography, also demonstrated less atherosclerosis in the pioglitazone treated group compared to patients treated with sulfonylureas (90).

 

There are a large number of potential mechanisms by which pioglitazone might reduce cardiovascular disease (Table 11) (74). In addition to altering risk factors pioglitazone has direct anti-atherogenic effects on the arterial wall that could reduce cardiovascular disease (74).

 

Table 11. Effect of Pioglitazone on Cardiovascular Risk Factors

Cardiovascular Risk Factor

Effect of Pioglitazone

Visceral Obesity

Decreases

Hypertension

Lowers BP

High Triglycerides

Lower TG

Low HDL cholesterol

Increases HDL cholesterol

Small dense LDL

Converts small LDL to large LDL

Endothelial dysfunction

Improves

Hyperglycemia

Lowers A1c

Inflammation

Lowers CRP

PAI-1

Lower PAI-1

Insulin resistance

Reduces

Hyperinsulinemia

Lowers insulin levels

 

While the data from a variety of different types of studies strongly suggests that pioglitazone is anti-atherogenic, the results with rosiglitazone are different. Several meta-analyses of small and short-duration rosiglitazone trials suggested that rosiglitazone was associated with an increased risk of adverse cardiovascular outcomes (91,92). However, the final results of the RECORD study, a randomized trial that was specifically designed to compare the effect of rosiglitazone vs. either metformin or sulfonylurea therapy as a second oral drug in those receiving either metformin or a sulfonylurea on cardiovascular events, have been published and did not reveal a difference in cardiovascular disease death, myocardial infarctions, or stroke (93,94). Similarly, an analysis of patients on rosiglitazone in the BARI 2D trial also did not suggest an increase or decrease in cardiovascular events in the patients treated with rosiglitazone (95).

 

Thus, while the available data indicate that pioglitazone is anti-atherogenic, the data for rosiglitazone suggests a neutral effect. Whether these differences between pioglitazone and rosiglitazone are accounted for by their differential effects on lipid levels or other factors is unknown.

 

NONALCOHOLIC FATTY LIVER DISEASE (NAFLD) AND NONALCOHOLIC STEATOHEPATITIS (NASH)

 

Studies have shown that pioglitazone has beneficial effects on NAFLD and NASH (96). In an early study 55 patients with impaired glucose tolerance or T2DM and liver biopsy-confirmed NASH were randomized to pioglitazone 45 mg/day or placebo (97). After 6 months of therapy liver enzymes improved and hepatic fat decreased, measured by magnetic resonance spectroscopy. Moreover, histologic findings improved including steatosis (P=0.003), ballooning necrosis (P=0.02), and inflammation (P=0.008). However, fibrosis was unchanged. A more recent study randomized 101 patients with prediabetes or T2DM and biopsy-proven NASH to pioglitazone 45 mg/day or placebo for 18 months (98). The primary outcome was a reduction of at least 2 points in the nonalcoholic fatty liver disease activity score in 2 histologic categories without worsening of fibrosis. Pioglitazone treatment resulted in 58% of patients achieving the primary outcome vs. only 17% of the placebo group (p<0.001) and 51% had resolution of NASH compared to 19% of the placebo group (p<0.001). Moreover, pioglitazone treatment improved the fibrosis score. 

 

A meta-analysis of 8 randomized controlled trials (5 using pioglitazone and 3 using rosiglitazone) with 516 patients with biopsy-proven NASH reported that TZD treatment was associated with improved advanced fibrosis (OR, 3.15; P = .01), fibrosis of any stage (OR, 1.66;  P = .01), and NASH resolution (OR, 3.22; P < .001) (99). Similar results were observed in patients with and without diabetes. Pioglitazone was more effective in improving NASH than rosiglitazone.

 

These studies demonstrate that pioglitazone has beneficial effects on NAFLD and NASH. Whether this will result in improved clinical outcomes will require additional studies. TZDs are not FDA approved for the treatment of NAFLD or NASH.

 

POLYCYSTIC OVARY SYNDROME

 

TZDs by improving insulin sensitivity decrease circulating androgen levels, improve ovulation rates, and improve glucose tolerance in patients with PCOS (57). Small trials have shown some benefit of TZDs for the treatment of infertility, usually in conjunction with clomiphene (57). Concerns regarding toxicity have limited the use of TZDs for the treatment of PCOS but if a patient has diabetes and TZDs are chosen for treating the diabetes one can anticipate beneficial effects on the PCOS. 

 

Side Effects

 

WEIGHT GAIN

 

TZDs lead to an increase in body weight of 2 to 3 kg for every 1 percent decrease in A1c levels (66). In some studies patients gained over 4 kg during a 26-week study (66). Weight gain to a similar degree occurred in monotherapy studies and in studies where TZDs were added to metformin, sulfonylureas, or insulin (66). However, in combination with an SGLT2 inhibitor or a GLP-1 receptor agonist the weight gain was blunted or prevented (100,101). In the ADOPT trial weight gain was greater with TZD therapy than with glyburide therapy (2.5 kg over 5 years) (25). The weight gain induced by TZDs is dose related and can be minimized by using low doses (102).

 

The TZD induced increase in body weight is due to an expansion of the subcutaneous fat depot whereas the mass of visceral fat remains unchanged or even decreases (66). While weight increases, waist circumference typically remains stable. Stimulation of PPAR gamma in subcutaneous adipocytes stimulates lipid accumulation (67). Fluid retention as discussed below may also contribute to the increase in weight.

 

FLUID RETENTION

 

Edema has been reported in 3.0 to 7.5% of patients treated with the TZDs compared with 1.0 to 2.5% in patients on placebo or treated with other oral antidiabetic therapy (103). The increase in fluid retention is dose related. The risk of developing edema is greatest when a TZD is used in combination with insulin (103). The occurrence of edema is reduced when a TZD is used in combination with an SGLT2 inhibitor (100).

 

TZD induced edema responds poorly to treatment with thiazide and loop diuretics but responds to diuretics that effect the distal tubules such as spironolactone, triamterene, and amiloride (102). Additionally, edema improves when TZD treatment is discontinued (103). The increased fluid retention can lead to an increase in plasma volume resulting in a modest decrease in hemoglobin levels (2-4%) (102).

 

The increase in fluid retention is likely due to TZDs activating PPAR gamma in the renal tubules leading to the increased expression of the epithelial Na(+) channel resulting in the increased resorption of sodium (104). TZDs have been shown to decrease urine sodium excretion and to increase plasma renin and aldosterone levels (105).

 

CONGESTIVE HEART FAILURE (CHF)

 

In a meta-analysis of seven studies with a total of 10,040 participants with 641 CHF events, pioglitazone treatment increased the risk of developing CHF by 33% (RR 1.33, 95% CI 1.14–1.54) (106). Another meta-analysis found that pioglitazone was associated with a 51% increased risk of CHF while rosiglitazone was associated with a 173% increase (107). In the RECORD trial, the rosiglitazone group had an increased rate of severe episodes of CHF resulting in hospital admission or death (OR 2.10, p = 0.001) (93). Similarly, in the PROactive trial, the pioglitazone group also had increased rates of CHF (6% vs. 4%, p = 0.007) (82). Patients treated with TZDs have a higher risk for CHF development if they have a history of cardiovascular disease (102). Interestingly, TZD-associated CHF has not been linked with increased mortality (82,108).

 

Although TZDs are associated with worsening of CHF or CHF development, they are not associated with adverse effects on cardiac function or structure (102). It is thought that the CHF is mainly due to fluid retention rather than TZDs inducing primarily cardiac dysfunction (102).

 

OSTEOPOROSIS

 

Large randomized trials have shown that TZDs increase fracture risk, particularly in women. In the ADOPT study, which compared rosiglitazone, metformin, and glyburide, there was no difference in the incidence of fractures in men (109). However, fractures in women at 5 years was increased in the group treated with rosiglitazone (rosiglitazone 15.1%, metformin 7.3%, and glyburide 7.7%) (109). The increase in fractures with rosiglitazone occurred in pre- and postmenopausal women, and were seen predominantly in the lower and upper limbs (109). In the PROactive study there was a higher rate of bone fractures in females treated with pioglitazone vs. placebo (5.1% vs 2.5%) (110). In the RECORD trial upper and distal lower limb fracture rates were increased mainly in women in the rosiglitazone treatment group (93). Hip and femur fracture were not increased with rosiglitazone treatment (93). In the IRIS trial an increased risk of fracture was seen in both males and females (men 9.4% vs 5.2%; HR, 1.83; women 14.9% vs 11.6%; HR, 1.32) (111). In a meta-analysis of 22 randomized controlled trials with 24,544 participants with 896 fracture cases there was a significantly increased incidence of fracture in women (OR=1.94; P<0.001), but not in men (OR=1.02; P=0.83) treated with TZDs (112). The risk of a fracture was similar with rosiglitazone and pioglitazone treatment and appeared to be similar for participants aged <60 years old and older than ≥60 years of age (112). Of note, in the ACCORD trial the risk of fractures in the women treated with rosiglitazone decreased after discontinuing rosiglitazone therapy (113).

 

In mice, TZDs suppress bone formation and increase bone resorption resulting in decreased bone mass (85). Additionally, TZD administration in mice results in the massive accumulation of adipocytes in the bone marrow cavity (85). In a meta-analysis of 14 trials with 1,734 participants, treatment with TZDs for 3 to 24 months decreased bone mineral density measured by DEXA at the lumbar spine (difference -1.1%; p < 0.0001), total hip (-1.0%; p < 0.0001) and forearm (-0.9%; p = 0.007) (112). In five studies TZD therapy was discontinued and after 24-52 weeks there was no increase in bone mineral density indicating no restoration of bone mineral density with cessation of TZD treatment (112). In an observation study each year of TZD use was associated with greater bone loss at the whole body (additional loss of -0.61% per year), lumbar spine (-1.23% per year), and trochanter (-0.65% per year) in women, but not men (114).The effect of TZD treatment on bone turnover markers varied considerably between individual studies (112). This reduction in bone mass induced by TZD treatment could contribute to the increase in fractures but it is possible that changes in the microarchitecture of bone also plays a role.

 

BLADDER CANCER

 

In preclinical studies pioglitazone administration increased bladder cancer in male rats but not in female rats or in mice, dogs, or monkeys (115). In the PROactive study there was a nonsignificant increase in the number of patients who developed bladder cancer (16 vs 6, p = 0.069) (82). In a number of instances, the development of bladder could not plausibly be related to treatment due to the temporal sequence of drug exposure and cancer diagnosis. After eliminating these patients there were six patients with bladder cancer in the pioglitazone group and three patients in the placebo group (82). After 10 years of follow-up, bladder cancer was reported in 0.8% of patients (n = 14) in the pioglitazone versus 1.2% (n = 21) in the placebo group (relative risk 0.65) during the follow-up period (116). In the IRIS study bladder cancer occurred in 12 patients in the pioglitazone group and in 8 in the placebo group (P=0.37) (83). Thus, in large randomized trials the data do not definitively support that pioglitazone significantly increases the risk of bladder cancer. The short duration of the randomized studies and infrequent occurrence of bladder cancer make interpretation of these studies difficult.

 

Because of the preclinical data the FDA requested that the manufacturer of pioglitazone initiate a prospective study to examine the relationship between pioglitazone and bladder cancer. This 10-year study of 193,099 persons did not find any statistically significant association between pioglitazone treatment and bladder cancer (117). Additionally, in a multinational cohort of 1.01 million patients with T2DM there was no evidence for any association between cumulative exposure to pioglitazone and bladder cancer in men or women after adjustment for age, calendar year, diabetes duration, smoking and any ever use of pioglitazone (118). Similarly, no association was observed between rosiglitazone and bladder cancer in men or women (118). In a careful review of 23 epidemiological studies Davidson concluded that there was little evidence that pioglitazone increased the risk of bladder cancer (115). The FDA still warns about the possibility of bladder cancer with pioglitazone use and recommends that pioglitazone not be used in diabetic patients with active bladder cancer or history of bladder cancer (package insert).

 

MACULA EDEMA

 

Macular edema has been reported in patients taking TZDs (119,120).  Patients may present with blurred vision or decreased visual acuity or be diagnosed on routine ophthalmologic examination. Most patients had peripheral edema at the time macular edema was diagnosed (120). Some patients had improvement in their macular edema after discontinuation of the TZD (120).

 

OVULATION AND PREGNANCY

 

As discussed above in the polycystic ovary section, treatment of premenopausal women with PCOS may induce ovulation and thereby result in unplanned pregnancies. In premenopausal anovulatory women started on a TZD one needs to discuss the need for contraception.

 

Contraindications and Drug Interactions

 

TZDs are contraindicated in patients with NYHA Class III or IV heart failure. Pioglitazone should not be used in diabetic patients with active bladder cancer or history of bladder cancer.

 

Strong CYP2C8 inhibitors (e.g., gemfibrozil) increase pioglitazone and rosiglitazone concentrations and one should limit pioglitazone dose to 15 mg daily (package insert).

 

Summary

 

TZDs are effective drugs in improving glycemic control and have significant benefits on disorders that occur commonly in patients with T2DM (cardiovascular disease, NAFLD/NASH, PCOS). Unfortunately, TZDs also have serious side effects, such as CHF, osteoporosis, and weight gain, that limit their use. Clinicians need to balance the advantages and disadvantages of TZDs for the individual patient.

 

Table 12. The Advantages and Disadvantages of Thiazolidinediones

Advantages

Disadvantages

Once a day administration

Edema

Reduces CVD (pioglitazone)

CHF

Durable Effect

Weight gain

Reduces NASH

Osteoporosis

No hypoglycemia

Bladder cancer (pioglitazone)?

Relatively inexpensive

Macula edema?

No dose adjustment for renal disease

Small increase in LDLc

Increase HDL-C and decrease triglycerides

 

 

ALPHA-GLUCOSIDASE INHIBITORS

 

Introduction

 

Acarbose (Precose, Glucobay), miglitol (Glycet) and voglibose (Basen, Voglib) are members of the α-glucosidase inhibitor class of oral anti-hyperglycemic compounds that were introduced in the 1990s (16).

 

Administration

 

The recommended starting dosage of acarbose and miglitol is 25 mg given orally three times daily at the start of each meal. The dose of acarbose and miglitol can be adjusted at 4 to 8-week intervals based on one-hour postprandial glucose or A1c levels, and on tolerance. The dosage can be increased from 25 mg tid with meals to 50 mg tid with meals. The maximum dose is 100 mg tid with meals. Note that the dose can be varied based on the amount of carbohydrate in the meal. In some patients one can initiate therapy once a day with the largest meal.

 

Mechanism of Action

 

Alpha-glucosidase inhibitors are competitive, reversible inhibitors of pancreatic α-amylase and membrane-bound intestinal α-glucosidase hydrolase enzymes (16,121). Inhibiting these enzymes prevents the metabolism of disaccharides and oligosaccharides into monosaccharides delaying carbohydrate digestion and absorption (16,121).  Carbohydrate absorption occurs more distally in the intestine reducing the postprandial increase in glucose and lowering prandial insulin levels (16,121). Acarbose and miglitol have minimal inhibitory activity against lactase and consequently will not prevent the increase in plasma glucose following the ingestion of milk or cause lactose intolerance (package insert). In addition to effecting carbohydrate absorption, alpha-glucosidase inhibitors increase postprandial GLP-1 secretion and reduce glucose-dependent insulinotropic polypeptide (GIP) secretion (16).

 

Glycemic Efficacy

 

The typical decrease in A1c levels is relatively modest with alpha-glucosidase inhibitors (0.5-1.0%) (37,121,122). The decrease in A1c is predominantly due to decreases in post meal glucose levels and alpha-glucosidase inhibitors have only modest effects on fasting glucose levels (16,121,122). Alpha-glucosidase inhibitors can be combined with other hypoglycemic drugs with additive effects and are particularly useful to lower postprandial glucose levels (37,121). Alpha-glucosidase inhibitors are most effective in patients who ingest a high carbohydrate diet and for this reason have been widely used and very effective in Asian populations (16).

 

These drugs do not cause weight gain and hypoglycemia is uncommon (16,37,122). If a patient experiences hypoglycemia while taking an alpha-glucosidase inhibitor in combination with insulin or sulfonylureas the patient should be instructed to use glucose (gel, tablets, etc.) as alpha-glucosidase inhibitors will prevent the breakdown of sucrose and delay glucose absorption resulting in a failure to quickly correct hypoglycemia. Severe hypoglycemia may require intravenous glucose or intramuscular glucagon administration.

 

Other Effects

 

CARDIOVASCULAR DISEASE

 

In the STOP-NIDDM trial 1,429 subjects with impaired glucose tolerance were randomized to placebo vs. acarbose and followed for 3.3 years (123). In the acarbose group a 49% relative risk reduction in the development of cardiovascular events (hazard ratio 0.51; P =0.03) was observed. Among cardiovascular events, the major reduction was in the risk of myocardial infarction (HR 0.09; P =0.02). In a smaller trial, 135 patients hospitalized for the acute coronary syndrome who were newly diagnosed with IGT were randomly assigned to acarbose or placebo (124). During a mean follow-up of 2.3 years the risk of recurrent major adverse cardiovascular event was decreased significantly in the acarbose group compared with that in control group (26.7% versus 46.9%, P < 0.05).

 

Despite these favorable observations a large trial failed to demonstrate a beneficial effect of acarbose in Chinese patients with impaired glucose tolerance (ACE trial) (125). In a randomized trial acarbose vs. placebo was compared in 6,522 patients with coronary heart disease and impaired glucose tolerance. The primary outcome was cardiovascular death, non-fatal myocardial infarction, non-fatal stroke, hospital admission for unstable angina, and hospital admission for heart failure and patients were followed up for a median of 5 years. The primary outcome was similar in the acarbose and placebo groups (hazard ratio 0.98; p=0.73). No significant differences were seen for death from any cause, cardiovascular death, fatal or non-fatal myocardial infarction, fatal or non-fatal stroke, hospital admission for unstable angina, hospital admission for heart failure, or impaired renal function.

 

Thus, whether acarbose favorably affects cardiovascular disease in patients at high risk for developing diabetes is uncertain. Moreover, the effect of acarbose on cardiovascular disease in patients with diabetes is unknown.

 

WEIGHT

 

Acarbose is may result in a very small decrease in weight (0.4kg) (126).

 

Side Effects

 

Gastrointestinal side effects of alpha-glucosidase inhibitors include flatulence, abdominal discomfort, and diarrhea and are very commonly encountered (16,37,122). These side effects can lead to the inability to tolerate these drugs. A high carbohydrate diet may worsen the GI adverse effects. Over time the GI symptoms tend to decrease as the intestines adapt (121). GI side effects are due to the mechanism of action of alpha-glucosidase inhibitors (121). The inhibition of carbohydrate digestion in the small intestine leads to the delivery of undigested carbohydrates to the large intestine where microorganisms metabolize them into short-chain fatty acids, methane, carbon dioxide, and hydrogen, that can cause abdominal discomfort, increased flatulence, and diarrhea (121).

 

Acarbose, particularly at doses in excess of 50 mg tid, may give rise to elevations of serum transaminases and, in rare instances, hyperbilirubinemia. It is recommended that serum transaminase levels be checked every 3 months during the first year of treatment with acarbose and periodically thereafter. If elevated transaminases are observed, a reduction in dosage or withdrawal of therapy may be indicated, particularly if the elevations persist (package insert).

 

Contraindications and Drug Interactions

 

Acarbose and miglitol are contraindicated in patients with inflammatory bowel disease, colonic ulceration, intestinal obstruction or those predisposed to intestinal obstruction, patients with chronic intestinal disease, or conditions that will be worsened by the increased gas formation in the intestine (37) (package insert). Acarbose is contraindicated in patients with cirrhosis (package insert).

 

Acarbose and miglitol should not be used in patients with a creatinine > 2 mg/dl (package insert).

 

Summary

 

Alpha-glucosidase inhibitors are excellent drugs for lowering postprandial glucose levels. Unfortunately, because of their GI side effects many patients are unable to tolerate these drugs. Additionally, the need for three times a day administration makes it difficult for patients to comply with these drugs.

 

Table 13. Advantages and Disadvantages of Alpha-Glucosidase Inhibitors

Advantages

Disadvantages

No hypoglycemia

GI side effects

Weight neutral

Frequent dosing schedule

Decreases postprandial glucose

Avoid if renal disease (creatinine> 2mg/dL

Relatively inexpensive

 

 

SODIUM-GLUCOSE TRANSPORT PROTEIN 2 (SGLT2) INHIBITORS

 

Introduction

 

There are currently four SGLT2 inhibitors available (Canagliflozin/ Invokana; Dapagliflozin/ Farxiga; Empagliflozin/Jardiance; Ertugliflozin/ Stelgatro) (127). These drugs are very similar and there are only a few differences between these agents.

 

Administration

 

The recommended starting dose of canagliflozin is 100 mg once daily, taken before the first meal of the day. In patients tolerating canagliflozin 100 mg once daily who have an eGFR of 60 mL/min/1.73 m2 or greater and require additional glycemic control, the dose can be increased to 300 mg once daily.

 

The recommended starting dose of dapagliflozin is 5 mg once daily, taken in the morning, with or without food. In patients tolerating dapagliflozin 5 mg once daily who require additional glycemic control, the dose can be increased to 10 mg once daily.

 

The recommended starting dose of empagliflozin is 10 mg once daily in the morning, taken with or without food. In patients tolerating empagliflozin, the dose may be increased to 25 mg.

 

The recommended starting dose of ertugliflozin is 5 mg once daily, taken in the morning, with or without food. In patients tolerating ertugliflozin 5 mg once daily who require additional glycemic control, the dose can be increased to 15 mg once daily.

 

Before initiating SGLT2 inhibitor therapy one should assess renal function and volume status. The dose of SGLT2 inhibitors may need to be adjusted based on renal function (see below).

 

Mechanism of Action

 

SGLT2 is a low-affinity, high-capacity glucose transporter in the proximal tubules of the kidneys, which is responsible for the reabsorption of the majority of the filtered glucose (approximately 90%) entering the tubules (16,128). SGLT1, which is predominantly expressed in the intestines is also expressed in the kidneys, is a high-affinity, low-capacity glucose transporter in the proximal tubules, which makes a minor contribution to the reabsorption of filtered glucose (approximately 10%) (16,128). SGLT 1 and 2 transporters are capable of reabsorbing virtually all the filtered glucose when blood glucose levels are less than approximately 180mg/dL. When blood glucose levels are greater than approximately 180mg/dL, glucose begins to appear in the urine (i.e., glycosuria). The higher the blood glucose level the greater the quantity of glucose in the urine. Patients with T2DM express a greater number of SGLT2 transporters in the proximal tubule than do healthy individuals and hence glucose reabsorption from the glomerular filtrate is increased in patients with diabetes and glycosuria occurs at a higher blood glucose level (typically approximately 220mg/dl (129).

 

Inhibition of SGLT2 by drugs results in glycosuria and can lead to the excretion of 60–90 grams of glucose in the urine per day (Figure 8) (16). The amount of glucose excreted in the urine can vary considerably depending on renal function and the degree of hyperglycemia (16). Decreased renal function results in a decrease in filtered glucose and less glucose in the urine while high blood glucose levels increase filtered glucose and increases the loss of glucose in the urine (16). The ability of the inhibition of SGLT2 to lower blood glucose levels is not dependent on insulin action and hence is not affected by insulin levels or insulin resistance (16). As will be discussed below many of the non-glucose lowering benefits and side effects of SGLT2 inhibitors can be explained by the increase in glucose excretion in the urine. It should be recognized that glycosuria results in an osmotic diuresis. Additionally, because the SGLT2 transporters also facilitate the reabsorption of sodium from the filtrate there is also the loss of sodium in the urine. 

 

Figure 8. Effect of SGLT2 Inhibitors on the Kidney

Glycemic Efficacy

 

A meta-analysis of 66 randomized trials found that SGLT2 inhibitors decreased A1c levels by 0.4 to 1.1% (130). In comparison to other hypoglycemic drugs, it was found that SGLT‐2 inhibitors showed a greater efficacy than DPP‐4 inhibitors and similar or slightly less efficacy compared to metformin and TZDs (12,130). Sulfonylureas appeared to be superior to SGLT‐2 inhibitors at 12 weeks, but at 24- and 52-weeks efficacy was similar or slightly lower (12,130). However, SGLT‐2 inhibitors produced a greater reduction in HbA1c than sulfonylureas at 104 weeks perhaps due to the lack of durability of sulfonylurea therapy discussed earlier (130). The A1c lowering ability of the different SGLT2 inhibitors is similar but A1c is reduced to a slightly greater extent by high-dose canagliflozin, which is probably a result of its additional action of inhibiting SGLT1 in the intestine decreasing dietary glucose absorption (127,128,130). SGLT2 inhibitors when used as an add-on therapy to metformin, insulin, thiazolidinediones, DPP-4 inhibitors, GLP-1 receptor agonists, sulfonylureas, or metformin ± DPP-4 inhibitor were similarly effective in reducing A1c levels as when used in monotherapy (16,128). The efficacy of SGLT2 inhibitors is dependent on renal function and as renal function decreases the ability of these drugs to lower A1c levels diminishes (16,128). SGLT2 inhibitors lower both fasting and postprandial glucose levels (128). In monotherapy SGLT2 inhibitors have a low risk of causing hypoglycemia but in combinations with insulin or sulfonylureas may potentiate the development of hypoglycemia (16). In patients in good glycemic control, one often decreases the insulin or sulfonylurea dose when initiating therapy with an SGLT2 inhibitor.

 

Other Effects

 

WEIGHT

 

SGLT2 inhibitors lead to weight loss (16,128). In general patient’s lose approximately 1- 3 kg on these drugs (16,127,128). SGLT2 inhibitor-induced weight loss results primarily from a decrease in fat mass, including reductions in visceral and subcutaneous adipose tissue (128). The weight loss is due to the loss of glucose in the urine, which represents the loss of calories (128,131).  The excretion of 50 grams of glucose in the urine is equivalent to the loss of 225 calories (50-grams X 4.5 calories per gram of glucose). However, the amount of glucose lost in the urine should result in a greater weight loss than is typically observed and a compensatory increase in food intake blunts the weight loss (131). There are likely to be other homeostatic mechanisms that also play a role in limiting weight loss with SGLT2 inhibitors.

 

GLUCOSE MONITORING

 

Monitoring glycemic control with 1,5-AG assay is not accurate as measurements of 1,5-AG are unreliable in patients taking SGLT2 inhibitors.

 

BLOOD PRESSURE

 

SGLT2 inhibitors decrease systolic BP by approximately 3-6 mmHg and diastolic BP by approximately 2-3 mmHg (16,128).  Patients with poorly controlled BP at baseline experience the largest reduction in BP (127). SGLT2 inhibitors lower BP by promoting an osmotic diuresis and decreasing intravascular volume (128). Weight loss may also contribute to the decrease in BP.

 

LIPID LEVELS

 

SGLT2 inhibitors cause a small increase in LDL and HDL cholesterol levels. In the EMPA-REG outcome study, described in detail below, LDL cholesterol levels were increased by 2-4 mg/dL and HDL cholesterol by 2-3 mg/dL in the group treated with empagliflozin (132). Similarly, in the CANVAS outcome study, discussed in detail below, LDL cholesterol and HDL cholesterol were also marginally increased in the canagliflozin treated group (LDL cholesterol 4-5 mg/dL and HDL cholesterol 2-3 mg/dL) (133). In a meta-analysis of 43 randomized trials with 22,528 patient’s triglyceride levels were decreased by 2 mg/dL (134). In a meta-analysis of 48 randomized controlled trials SGLT2 inhibitors significantly increased LDL-C (3.8mg/dl, p < 0.00001), HDL-C (2.3mg/dl, p < 0.00001), and decreased triglyceride levels (8.8mg/dl, p < 0.00001) (135). It is unlikely that these small changes in LDL-C, HDL-C, and triglyceride levels are of clinical significance. The mechanism for these increases in LDL and HDL cholesterol is unknown but could be due to a decrease in plasma volume. The decrease in triglycerides might be secondary to weight loss.

 

URIC ACID

 

SGLT2 inhibitors lower blood uric acid levels (136). This decrease is due to an increase in uric acid excretion by the kidneys. In an observational study 47,905 individuals receiving an SGLT2 inhibitor and 183,303 receiving a DPP4 inhibitor it was observed that the incidence of gout was 20.26 per 1000 patient-years for SGLT2 inhibitor users and 24.30 per 1000 patient-years for DPP4 inhibitor users (137).

 

CARDIOVASCULAR

 

There have been several large randomized studies of the effect of SGLT2 inhibitors on cardiovascular events published (others are in progress).

 

EMPA-REG Outcome Trial 

 

In this study, 7,020 patients with established cardiovascular disease and T2DM were randomly assigned to receive 10 mg or 25 mg of empagliflozin or placebo once daily and were followed for 3.1 years (132). In the combined empagliflozin treated groups there was a statistically significant 14% reduction in the primary outcome (death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke). As compared with placebo, empagliflozin treatment did not result in a significant difference in the occurrence of non-fatal myocardial infarction or strokes. However, empagliflozin resulted in a significantly lower risk of death from cardiovascular causes (hazard ratio, 0.62), death from any cause (hazard ratio, 0.68), and hospitalization for heart failure (hazard ratio, 0.65). The beneficial effects of empagliflozin were noted to occur very rapidly and the beneficial effects on heart failure appeared to be the dominant effect compared to effects on atherosclerotic events. Decreases in cardiovascular outcomes and mortality with empagliflozin occurred across the range of cardiovascular risk (138). Additionally, the reduction in hospitalizations for heart failure and cardiovascular death were observed both in patients with and without heart failure at baseline (139).

 

CANVAS Trial

 

The effects of placebo vs. canagliflozin 100mg or 300mg per day were determined in two combined trials involving a total of 10,142 participants with T2DM and high cardiovascular risk (approximately 70% of patients had established cardiovascular disease) (133). The primary outcome was a composite of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke and the mean follow-up was 188 weeks. The primary outcome was reduced in the canagliflozin group (hazard ratio, 0.86; P=0.02). The effect of canagliflozin on the primary outcome was similar in people with chronic kidney disease and those with preserved kidney function (140). Death from any cause (hazard ratio 0.87; 95% CI 0.74-1.01) and death from cardiovascular disease (hazard ratio 0.87; 95% CI 0.72-1.06) were reduced but were not statistically significant. Similarly, canagliflozin treatment did not result in a significant difference in non-fatal strokes or non-fatal myocardial infarctions (hazard ratio 0.90 for stroke and 0.85 for myocardial infarction). As seen with empagliflozin, hospitalization for heart failure was markedly reduced (hazard ratio 0.67; 95% CI 0.52-0.87) and this beneficial effect occurred rapidly.

 

CREDENCE Trial

 

In a second canagliflozin trial that focused on patients with kidney disease, a decrease in cardiovascular events was also observed (141). In this double-blind trial 4,401 patients with chronic kidney disease and T2DM were randomized to canagliflozin 100mg per day or placebo and followed for a median of 2.62 years. All the patients had an eGFR of 30 to <90 ml per minute per 1.73 m2 and albuminuria (ratio of albumin [mg] to creatinine [g], >300 to 5000). In this trial hospitalization for heart failure was reduced by 39%. The relative benefits of canagliflozin for cardiovascular outcomes was similar in individuals across the spectrum of eGFR levels (142)

 

DECLARE–TIMI 58 Trial

 

The effect of dapagliflozin on cardiovascular events has been reported (143). 17,160 patients with T2DM, including 10,186 without atherosclerotic cardiovascular disease, were randomized to dapagliflozin 10mg per day or placebo and followed for a median of 4.2 years. The primary outcome was a composite of major adverse cardiovascular events (MACE), defined as cardiovascular death, myocardial infarction, or ischemic stroke. The primary efficacy outcomes were MACE and a composite of cardiovascular death or hospitalization for heart failure. Dapagliflozin did not result in a lower rate of major adverse cardiovascular events (8.8% in the dapagliflozin group and 9.4% in the placebo group; hazard ratio, 0.93; P=0.17) but did result in a lower rate of cardiovascular death or hospitalization for heart failure (4.9% vs. 5.8%; hazard ratio, 0.83; P=0.005), which reflected a lower rate of hospitalization for heart failure (hazard ratio, 0.73; 95% CI, 0.61 to 0.88). Interestingly, in the patients with a history of a previous MI dapagliflozin reduced the risk of a MACE (HR 0.84; P=0.039), whereas there was no effect in patients without a previous MI (144). Dapagliflozin reduced the risk of heart failure in patients with and without a history of heart failure but the benefit was greater in patients with a history of heart failure (with heart failure HR 0.62; 95% CI, 0.45-0.86; without heart failure HR 0.88; 95% CI, 0.74-1.03) (145). Dapagliflozin also reduced the risk of heart failure in patients without a history of atherosclerotic cardiovascular disease (146). In addition, dapagliflozin reduced the risk of atrial fibrillation and atrial flutter by 19% (HR, 0.81; P=0.009) (147).

 

VERTIS CV

 

Patients with atherosclerotic cardiovascular disease and T2DM were randomized to ertugliflozin 5mg (n=2752), 15mg (2747), or placebo (n=2747) and the primary composite outcome of cardiovascular death and non-fatal MI or stroke was determined after a mean duration of follow-up of 3.5 years (148). This trial did not demonstrate a significant difference in the primary endpoint (MACE) nor any components of the primary endpoint. However, heart failure hospitalizations were significantly reduced by 30% in the patients treated with ertugliflozin (HR 0.70; CI 0.54–0.90). The benefits on heart failure were observed in both patients with a history of heart failure (decreased 37%) and patients without a history of heart failure (decreased 21%) (149).

 

DAPA HF Trial

 

In this trial 4,744 patients with New York Heart Association class II, III, or IV heart failure and an ejection fraction of 40% or less were randomized to receive either dapagliflozin 10 mg once daily or placebo for a median of 18.2 months (150). The primary outcome was a composite of worsening heart failure (hospitalization or an urgent visit resulting in intravenous therapy for heart failure) or cardiovascular death. Of note only approximately 45% of the patients had type 2 diabetes. Treatment with dapagliflozin reduced the primary outcome (HR 0.74; 95% CI, 0.65 to 0.85; P<0.001), heart failure (HR 0.70; 95% CI, 0.59 to 0.83), and death from cardiovascular disease (HR 0.82; 95% CI, 0.69 to 0.98). Symptoms of heart failure were also improved with dapagliflozin treatment. Additionally, dapagliflozin reduced the risk of any serious ventricular arrhythmia, cardiac arrest, or sudden death (151). The benefits of dapagliflozin were similar in patients with diabetes and the non-diabetic patients (152). This study demonstrates that an SGLT2 inhibitor is beneficial in patients with pre-existing heart failure and this occurs in both patients with and without diabetes.

 

EMPEROR-Reduced Trial

 

In this trial 3,730 patients with class II, III, or IV heart failure and an ejection fraction of 40% or less were randomized to empagliflozin 10 mg once daily or placebo for a median of 16 months (153). The primary outcome was a composite of cardiovascular death or hospitalization for heart failure. Approximately 50% of the patients had type 2 diabetes. Treatment with empagliflozin reduced the primary outcome (HR 0.75; 95% CI, 0.65 to 0.86; P<0.001) and hospitalization for heart failure (HR 0.69; 95% CI, 0.59 to 0.81) but did not reduce cardiovascular death (HR 0.92; 95% CI, 0.75 to 1.12). The beneficial effects were observed in patients with and without diabetes. This study is concordant with the results observed in the DAPA HF trial and demonstrates that SGLT2 inhibitors are beneficial in patients with pre-existing heart failure and this occurs in both patients with and without diabetes.

 

DAPA CKD Trial

 

This trial randomly assigned 4,304 participants with an eGFR of 25 to 75 ml/min/1.73 m2 of body-surface area and a urinary albumin-to-creatinine ratio 200 to 5000 mg/g to receive dapagliflozin 10 mg once daily or placebo for a median of 2.4 years (154). Approximately 67% of the patients had diabetes. The composite of death from cardiovascular causes or hospitalization for heart failure was decreased in the dapagliflozin group (HR 0.71 95% CI, 0.55–0.92).  

 

EMPEROR-Preserved Trial

 

This trial randomized 5,988 patients with heart failure with an ejection fraction of >40% to double-blind treatment with placebo or empagliflozin 10 mg once daily (155). Empagliflozin decreased the combined risk of cardiovascular death, hospitalization for heart failure, or an emergency or urgent heart failure visit by 23% (HR 0.77; P<0.0001). Moreover, this benefit occurred rapidly reaching statistical significance at 18 days after randomization. The benefit of empagliflozin was similar in patients with an ejection fraction of >40% to <50% and 50% to <60%, but was attenuated at higher ejection fractions. These results indicate that SGLT2 inhibitors are beneficial even in patients with a preserved ejection fraction.

 

Summary

 

Thus, nine randomized trials of SGLT2 inhibitors demonstrated a robust decrease in heart failure (table 14) with SGLT2 inhibitor therapy without a consistent strong effect on myocardial infarctions or strokes. In a meta-analysis of eight of these trials (not including Emperor Preserved) with 59,747 patients it was observed that SGLT2 inhibitors reduced the risk of all-cause mortality (HR 0.84; 95% CI [0.78-0.91]), cardiovascular mortality (HR 0.84; 95% CI [0.76-0.93]) hospitalization for heart failure (HR 0.69; 95% CI [0.64-0.74]), and myocardial infarction (HR 0.91; 95% CI [0.84-0.99]), but there was no significant effect on the risk of stroke (HR 0.98; 95% CI [0.86-1.11]) (156). The reduction in heart failure was seen in patients with and without diabetes, patients with renal disease, and patients with and without a history of heart failure. The Emperor Preserved trial demonstrated that patients with a preserved ejection fraction also benefit from treatment with a SGLT2 inhibitor. Finally, the EMPULSE trial demonstrated that starting empagliflozin during the hospitalization for heart failure was beneficial (157). For additional information see the section on drugs that inhibit both SGLT1 and 2.

 

Table 14. Summary of Effect of SGLT2 Inhibitors on Heart Failure

 

Number

Prior Heart Failure

Mean Follow-up (years)

Hazard Ratio* (95% CI)

P value

EMPA-REG

Empagliflozin

7,020

10.1%

3.1

0.65

(0.05-0.85)

0.002

CANVAS

Canagliflozin

10,142

14.4%

3.6

0.67

(0.52-0.87)

--

DECLARE-TIMI 58

Dapagliflozin

17,160

10.0%

4.2

0.73

(0.61-0.88)

0.0007

VERTIS-CV

Ertugliflozin

8,246

23.7%

3.0

0.70

(0.54-0.90)

0.006

CREDENCE

Canagliflozin

4,401

14.8%

2.6

0.61

(0.47-0.80

0.001

DAPA-HF

Dapagliflozin

4,774

100%

1.5

0.70

(0.59-0.83)

0.001

EMPEROR

Empagliflozin

3,730

100%

1.3

0.69

(0.59-0.81)

<0.001

EMPEROR Preserved

5,988

100%

2.2

0.73

(0.61 to 0.88)

<0.001

DAPA-CKD

4,304

11%

2.4

0.71**

(0.55–0.92)

<0.009

*Hospitalization for Heart Failure

** Hospitalization for Heart Failure and death from cardiovascular disease

 

The mechanisms accounting for the beneficial effects of SGLT2 inhibitors on heart failure are uncertain (158). Glycemic control was better in the SGLT2 inhibitor treated patients but it is doubtful that this modest decrease in glucose could account for the observed results (additionally benefit in non-diabetics makes a glucose effect very unlikely). SGLT2 inhibitor treatment was associated with small reductions in weight, waist circumference, uric acid level, and systolic and diastolic blood pressure, with no increase in heart rate and small increases in both LDL and HDL cholesterol. Whether these changes played a role in reducing events remains to be determined but it is unlikely that these play a major role as other treatments that effect these factors do not markedly diminish the risk of heart failure events. It is possible that hemodynamic changes secondary to the osmotic diuresis induced by SGLT2 inhibitors contributed to the beneficial effects. In an analysis of the EMPA-REG OUTCOME trial, the change in hematocrit (~3% increase), corresponding to ~7% reduction in plasma volume, accounted for approximately 50% of the benefit of the drug on cardiovascular death (159). Additionally, SGLT2 inhibitors increase free fatty acid levels and glucagon secretion, which promotes the production of ketone bodies such as beta-hydroxybutyrate that are utilized by the heart for energy production (160). It is possible that this alternative source of energy could be protective for heart function. Finally, there may be direct effects of SGLT2 inhibition on myocardial and renal metabolism (158,161,162). Further studies are required to better elucidate the mechanism of the beneficial effects of SGLT2 inhibitors on heart failure.

 

RENAL DISEASE

 

The large randomized SGLT2 inhibitor cardiovascular outcome trials described above also examined the effect of these drugs on renal disease.

 

EMPA-REG Outcome Trial

 

The effect of empagliflozin on renal outcomes was studied in 4,124 patients with T2DM who were randomized to empagliflozin (10 mg or 25 mg) or placebo (163). The prespecified outcomes were progression to macroalbuminuria, doubling of the serum creatinine level, initiation of renal-replacement therapy, or death from renal disease and incident albuminuria. Worsening nephropathy occurred in 12.7% of patients in the empagliflozin group and in 18.8% of patients in the placebo group, a relative risk reduction of 39% (P<0.001). Progression to macroalbuminuria was reduced 38%, doubling of serum creatinine by 44%, and initiation of renal replacement therapy by 55% (all statistically significant). The renal benefit was seen regardless of baseline eGFR, occurring in individuals with an eGFR as low as 30 mL/min/1.73 m2. While empagliflozin caused an initial decrease in eGFR over the long term eGFR decreased in the placebo group at a more rapid rate than the empagliflozin group. Additionally, patients treated with empagliflozin were more likely to convert from microalbuminuria to normoalbuminuria (hazard ratio [HR] 1.43; p<0·0001) or from macroalbuminuria to microalbuminuria or normoalbuminuria (HR 1.82; p<0·0001), and were less likely to experience a sustained deterioration from normoalbuminuria to microalbuminuria or macroalbuminuria (HR 0·84; p=0·0077) (164).

 

CANVAS Trial

 

Similar to the results seen with empagliflozin, canagliflozin has also been shown to decrease renal disease. 10,142 participants with T2DM and high cardiovascular risk were randomly assigned to receive canagliflozin or placebo and were followed for a mean of 188.2 weeks (133). Progression of albuminuria occurred less frequently in the canagliflozin group (hazard ratio of 0.73; 95% CI, 0.67 to 0.79). In addition, regression of albuminuria also occurred more frequently in the canagliflozin group (hazard ratio, 1.70; 95% CI, 1.51 to 1.91). Most importantly, the composite outcome of sustained 40% reduction in eGFR, the need for renal-replacement therapy, or death from renal causes occurred less frequently in the canagliflozin group (hazard ratio of 0.60; 95% CI, 0.47 to 0.77). Annual eGFR decline was slower (slope difference between groups 1.2 mL/min/1.73 m2 per year, 95% CI 1.0-1.4) and mean urinary albumin creatinine ratio was 18% lower (95% CI 16-20) in participants treated with canagliflozin than in those treated with placebo (165). The benefits of canagliflozin on renal disease occurred across a wide spectrum of eGFR ranging from 30-45 to ≥90 mL/min/1.73 m2 and in patients with moderate and severe albuminuria (140,166).

 

CREDENCE Trial

 

The CREDENCE Trial focused on patients with renal disease. In a double-blind trial 4,401 patients with T2DM and chronic kidney disease were randomized to canagliflozin or placebo and followed for a median of 2.62 years (141). All the patients had an eGFR of 30 to <90 mL/min/1.73 m2 and albuminuria (ratio of albumin [mg] to creatinine [g], >300 to 5000) and were treated with renin-angiotensin system blockade. The primary outcome was a composite of end-stage kidney disease (dialysis, transplantation, or a sustained estimated GFR of <15 mL/min/1.73 m2), a doubling of the serum creatinine level, or death from renal or cardiovascular causes. The primary outcome was 30% lower in the canagliflozin group (hazard ratio, 0.70; P = 0.00001). The relative risk of the renal-specific composite of end-stage kidney disease, a doubling of the creatinine level, or death from renal causes was 34% lower (hazard ratio, 0.66; P<0.001), and the relative risk of end-stage kidney disease was 32% lower (hazard ratio, 0.68; P = 0.002). Benefits were seen regardless of baseline eGFR.

 

DECLARE–TIMI 58 Trial

 

In this trial of 17,160 participants a secondary outcome was a renal composite outcome defined as a sustained decrease of 40% or more in eGFR to less than 60 mL/min/1.73 m2, new end-stage renal disease, or death from renal or cardiovascular causes (143). As seen in the other SGLT2 inhibitor studies there was a decrease in the development of renal disease with the incidence of the renal outcome 4.3% in the dapagliflozin group vs. 5.6% in the placebo group (hazard ratio, 0.76; 95% CI, 0.67 to 0.87). Excluding death from cardiovascular causes as part of the composite endpoint, the reduction in renal events was even more impressive (HR 0.53 p<0.0001) (167). The risk of end-stage renal disease or renal death was lower in the dapagliflozin group than in the placebo group (HR 0.41; p=0.012) (167).

 

VERTIS CV Trial

 

In VERTIS CV trial the renal composite end point of renal death, dialysis/transplant, or doubling of serum creatinine was reduced but not statistically significant in the ertugliflozin treated group (HR 0.81; CI 0.63–1.04) (148).

 

DAPA-HF Trial

 

In this trial 4,744 patients with New York Heart Association class II, III, or IV heart failure and an ejection fraction of 40% or less were randomized to receive either dapagliflozin 10 mg once daily) or placebo for a median of 18.2 months (150). The renal outcome was a composite outcome of a reduction of 50% or more in the estimated GFR sustained for at least 28 days, end-stage renal disease, or death from renal causes. End-stage renal disease was defined as an eGFR of less than 15 ml/min/1.73 m2, long-term dialysis, or kidney transplantation. There was a trend towards benefit with dapagliflozin treatment that was not statistically significant due to a small number of events (HR 0.71; 95% CI 0.44 to 1.16).

 

EMPEROR-Reduced Trial

 

In this trial 3,730 patients with class II, III, or IV heart failure and an ejection fraction of 40% or less were randomized to empagliflozin 10 mg once daily or placebo for a median of 16 months (153). The annual rate of decline in the eGFR was decreased in the empagliflozin group compared to the placebo group (-0.55 vs. -2.28 ml per minute per 1.73 m2 of body-surface area per year, P<0.001). Additionally, a composite renal outcome (chronic dialysis or renal transplantation or a profound, sustained reduction in the eGFR) was decreased in the empagliflozin group (HR 0.50; 95% CI, 0.32 to 0.77).

 

DAPA-CKD Trial

 

In this trial 4,304 individuals with an eGFR) of 25 to 75 ml/minute/m2 and a urinary albumin-to-creatinine ratio of 200 to 5000mg/g were randomized to dapagliflozin 10 mg/day or placebo for a median of 2.4 years (this study was stopped early by the data monitoring board) (154). The primary outcome was a composite of a sustained decline in the estimated GFR of at least 50%, end-stage kidney disease, or death from renal or cardiovascular causes and this was reduced by 39% in the dapagliflozin group (HR 0.61; 95%l CI 0.51 to 0.72; P<0.001; number needed to treat to prevent one primary outcome event, 19). All of the components of this primary outcome were decreased in the dapagliflozin group. A sustained decline in the estimated GFR of at least 50%, end-stage kidney disease, or death from renal causes was reduced by 44% in the dapagliflozin group (HR 0.56; P<0.001). In the subgroup of patients with Stage 4 chronic kidney disease (eGFR< 30) the benefits of dapagliflozin were similar to those described above indicating that even in patients with severe renal disease dapagliflozin is beneficial (168) Finally, the benefits of dapagliflozin were similar in participants with type 2 diabetes (36% decrease) and in those without type 2 diabetes (50% decrease). Thus, similar to the CREDENCE trial, this trial demonstrates that dapagliflozin decreases renal disease progression in patients with pre-existing renal disease. Moreover, this benefit is seen in patients with and without diabetes. Finally, benefit was observed in the dapagliflozin group regardless of the type of kidney disease (diabetic, ischemic, hypertensive, glomerulonephritis, other, or unknown) (169).

 

Summary

 

Multiple trials clearly demonstrate that SGLT2 inhibitors have beneficial effects on renal function and decrease the development and progression of renal disease (Table 15). In a meta-analysis of these 8 trials with 59,747 patients there was a robust decrease in the composite end points of renal disease (HR 0.62; 95% CI, 0.56-0.70) (156). The benefits are observed in patients with and without diabetes, with and without renal disease, and also in patients with heart failure. In a smaller meta-analysis this renal disease benefit was seen in patients with and without atherosclerosis (170). These renal benefits are independent of improvement in glycemic control and occurs in patients without diabetes (171).

 

Table 15. Summary of SGLT2 Inhibitors on Renal Disease

 

Number

Mean Follow-up (years)

Hazard Ratio* (95% CI)

EMPA-REG; Empagliflozin

7,020

3.1

0.54 (0.40-0.75

CANVAS; Canagliflozin

10,142

3.6

0.60 (0.47-0.77)

DECLARE-TIMI 58; Dapagliflozin

17,160

4.2

0.53 (0.43-0.66)

VERTIS-CV; Ertugliflozin

8,246

3.0

0.81 (0.63-1.04)

CREDENCE; Canagliflozin

4,401

2.6

0.66 (0.53-0.81)

DAPA-HF; Dapagliflozin

4,774

1.5

0.71 (0.44-1.16)

EMPEROR; Empagliflozin

3,730

1.3

0.52 (0.32-0.77)

DAPA-CKD; Dapagliflozin

4304

2.4

0.56 (0.45-0.68)

*Renal composite outcomes

 

The mechanism accounting for this effect is unknown but a leading hypothesis is that an increase of sodium chloride in the macula densa due to SGLT2 inhibition triggers a cascade that reduces GFR through constriction of the afferent glomerular arterioles (tubuloglomerular feedback) (128,171). This would reduce glomerular hydrostatic pressure and initially decrease GFR, an effect that is observed with SGLT2 treatment, but in the long run this decrease in GFR protects the kidney from damage resulting in improved kidney function long-term (128).

 

NONALCOHOLIC FATTY LIVER DISEASE (NAFLD) AND NONALCOHOLIC STEATOHEPATITIS (NASH)

 

Numerous studies have shown that treatment with SGLT-2 inhibitors decrease liver enzymes (96,172-176).  Moreover, studies have shown a decrease in liver fat and liver stiffness (96,172,173,175-177). A study of 5 patients showed an improvement in liver histology after 24 weeks of therapy with canagliflozin (178). Further studies are required to determine whether SGLT-2 inhibitors will result in clinical benefits in patients with NAFLD and NASH.

 

MORTALITY

 

A meta-analysis of 21 randomized controlled trials with 70,364 individuals reported that all-cause mortality was decreased by 14% (179). The decrease in all-cause mortality was seen with all of the SGLT2 inhibitors but was not statistically significant with ertugliflozin.

 

Side Effects

 

In a meta-analysis of 51 randomized controlled trials involving 24,371 patients it was noted that the frequency of side effects was similar with high dose and low dose SGLT-2 inhibitors (180).

 

URINARY TRACT INFECTIONS

 

In some but not all studies an increased risk of urinary tract infections was observed with SGLT2 inhibitors (16,127). In the large randomized cardiovascular outcome trials, an increase in urinary tract infections were not observed (132,133,143). In a meta-analysis of 10 large outcome trials with 71,553 participants the relative risk of urinary tract infection was minimal (RR 1.06, 95% CI 1.00-1.12) (181). In a large meta-analysis of 86 randomized trials with 50,880 patients an increase in urinary tract infections was also not observed (182). The potential increase in the occurrence and severity of urinary tract infections is due to the glycosuria as glucose is an excellent substrate for the growth of micro-organisms.

 

GENITAL MYCOTIC INFECTIONS

 

Genital mycotic infections (mainly balanitis and vulvovaginitis) are increased with SGLT2 inhibitor treatment (127). The risk of genital mycotic infections is greater in women than men. In a meta-analysis that included over 2000 patients treated with canagliflozin 100 mg or 300 mg vs. placebo, genital mycotic infections were seen in greater than 10% of women (100mg-10.4%, 300 mg-11.4%, placebo-3.2%) and around 4% of men (100 mg-4.2%, 300 mg-3.7%, placebo- 0.6%) (183). In uncircumcised men the risk of genital mycotic infections is greater than in circumcised men. Genital mycotic infections are the most common side effect seen with SGLT2 inhibitors but fortunately these infections are generally mild and relatively easy to treat (16).

 

The increase in genital mycotic infections is due to the glycosuria as glucose is an excellent substrate for the growth of Candida.

 

FOURNIER GANGRENE

 

Fournier gangrene (FG) is a necrotizing fasciitis of the perineum that is characterized by a rapidly progressive necrotizing infection of the external genitalia, perineum, and perianal region (184). Many of the patients with FG have diabetes (32-66%) (184). FG occurs most commonly in males and is a rare condition with an incidence of 3.3 in 100,000 men aged 50 to 79 years (184). In a recent case series of 59 patients over a 10-year period at a single institution, the incidence was estimated at 32 cases per 100,000 admissions (185).  Risk factors included very high A1c (mean 9.6%), obesity, immunocompromised state, and illicit drug use (185).  FG is a urologic emergency and requires treatment with broad-spectrum antibiotics and immediate surgical intervention (184).

 

A recent report described 55 FG cases in patients treated with SGLT2 inhibitors in the last 6 years since they were approved for use in the US (184). In contrast, only 19 cases of FG were reported in 35 years among patients receiving other hypoglycemic drugs. All of the SGLT2 inhibitors were associated with FG except ertugliflozin, which is likely explained by this drug only recently being approved for the treatment of diabetes. However, the authors were unable to assess the incidence of FG or whether SGLT2 inhibitors were causative. A second study compared the occurrence of FG in patients treated with SGLT2 inhibitors (15.0 per 100,000 person-years) vs DPP4 inhibitors (9.7 per 100 000 person-years) in men 65 years and older who have T2DM using large data bases (186).

 

Early recognition of FG is essential to reduce morbidity and mortality. Typical presentations include systemic symptoms, such as fatigue, fever, and malaise, and local symptoms that include tenderness, erythema, and swelling (184). Pain out of proportion to the clinical findings is highly suggestive of necrotizing fasciitis (184).

 

HYPOVOLEMIA AND HYPOTENSION

 

SGLT2 inhibitors induce an osmotic diuresis (127). This effect can result in postural dizziness, orthostatic hypotension, falls, and dehydration, particularly in elderly individuals, patients with kidney disease, patients on either diuretics or medications that interfere with the renin-angiotensin-aldosterone system (e.g., angiotensin-converting-enzyme inhibitors, angiotensin receptor blockers), and patients with low systolic blood pressure (127) (package insert). In a meta-analysis of 10 large outcome studies the risk of volume depletion was modestly increased (RR 1.14, 95% CI 1.06-1.23) (181). Volume status should be determined prior to initiating therapy with an SGLT2 inhibitor.

 

ACUTE KIDNEY INJURY

 

SGLT2 inhibitors have been reported to cause acute kidney injury (127). It is likely that volume depletion and hypotension lead to the acute kidney injury (127). In an analysis of two large health care utilization cohorts SGLT2 inhibitors were not associated with an increased risk of acute kidney injury (187). Similarly, in the cardiovascular outcome studies described earlier an increase in acute kidney injury was not observed. In fact, in a meta-analysis of 4 large studies (EMPA-REG, CANVAS, CREDENCE, and DECLARE-TIMI 58) a decrease in acute kidney injury was observed (Risk ratio 0.75; p<0.0001) (188). Similarly, a meta-analysis of 10 studies with 71,553 participants also did not observe an increase in acute kidney injury and in fact observed a decrease (RR 0.84, 95% CI 0.77-0.91) (181). Even in patients over age 75 years of age an increase in acute kidney injury was not observed with SGLT2 treatment (189).

 

Before initiating SGLT2 inhibitor therapy one should consider factors that may predispose patients to acute kidney injury including hypovolemia, chronic renal insufficiency, congestive heart failure, and concomitant medications (diuretics, ACE inhibitors, ARBs, NSAIDs). Consider temporarily discontinuing SGLT2 inhibitors in any setting of reduced oral intake (such as acute illness or fasting) or fluid losses (such as gastrointestinal illness or excessive heat exposure) (package insert).

 

DIABETIC KETOACIDOSIS

 

Diabetic ketoacidosis (DKA) has been observed in patients with T2DM treated with SGLT2 inhibitors but is a rare side effect (16,127). In some instances, the glucose levels are not very elevated despite the patient having DKA (euglycemic DKA) and this can result in a delay in diagnosing DKA (127). SGLT2 inhibitors were associated with approximately twice the risk of diabetic ketoacidosis compared to treatment with DPP-4 inhibitors (190). Additionally, in several of the large cardiovascular studies described above an increase in DKA was observed (CANVAS Trial- canagliflozin 0.6 vs. placebo 0.3 participants with an event per 1000 patient-years; CREDENCE Trial- canagliflozin 2.2 vs. placebo 0.2 per with an event per 1000 patient-years; DECLARE–TIMI 58-dapagliflozin 27 episodes vs placebo 12 episodes; VERTIS trial 0.3% 5mg ertugliflozin, 0.4% 15mg dose, and 0.1% placebo group) (133,141,143,148). In a meta-analysis of 10 studies with 71,553 participants the risk of DKA was increased (RR 2.23, 95% CI 1.36-3.63) (181).  

 

Many of the DKA events occurred in patients with T2DM treated with insulin who had reduced or stopped insulin or experienced an intercurrent illness that could precipitate DKA (16,191). In some instances, the patients were thought to have T2DM but actually had latent autoimmune diabetes of adults (LADA), a form of Type 1 diabetes (16). The hyperglycemia in DKA associated with SGLT2 inhibitors is typically mild because the SGLT2 inhibitors reduce blood glucose levels (16). SGLT2 inhibitors should be temporarily discontinued in clinical situations known to predispose to ketoacidosis (e.g., prolonged fasting due to acute illness or surgery) (package insert). DKA developing during hospitalizations has been described emphasizing the need for vigilance when continuing SGLT-2 inhibitors in patients admitted to the hospital (192). Patients should be educated regarding this potential complication and in high-risk patients (for example patients on insulin therapy with a history of poor glycemic control or DKA) one could provide the patient with methods to measure ketone levels at home to facilitate the early diagnosis of DKA.

 

A possible mechanism for the increased risk of DKA is SGLT2 inhibitors increasing plasma glucagon levels thereby increasing ketone production (127,191). In combination with the low insulin levels this could potentiate the development of DKA.

 

OSTEOPOROSIS AND FRACTURES

 

In the CANVAS cardiovascular outcome study, the rate of all fractures was higher in the canagliflozin group than in the placebo group (15.4 vs. 11.9 participants with fracture per 1000 patient-years; hazard ratio, 1.26; 95% CI, 1.04 to 1.52) (133). A similar trend was observed for low-trauma fracture events (canagliflozin 11.6 vs. placebo 9.2 participants with fracture per 1000 patient-years; hazard ratio, 1.23; 95% CI, 0.99 to 1.52) (133). The incidence of fractures in the CANVAS study was increased with canagliflozin vs. placebo across subgroups based on sex, age, duration of Type 2 diabetes, baseline eGFR, and prior fracture history (193). Notably, the increase in fractures associated with canagliflozin treatment began within weeks of drug initiation indicating that the increased risk occurs rapidly (193).

 

In contrast, both the EMPA-REG, VERTIS, and DECLARE cardiovascular outcome studies did not demonstrate an increase in fractures with empagliflozin or dapagliflozin, respectively (132,143,148). Additionally, in the CREDENCE outcome study, canagliflozin did not increase fracture risk in patients with chronic kidney disease defined as an eGFR of 30 to <90 mL/min/1.73 m2 and albuminuria (ratio of albumin [mg] to creatinine [g], >300 to 5000) (141). Similarly, in a pooled analysis of 8 randomized canagliflozin studies with 5867 participants (CANVAS trial excluded) an increase in fractures was not observed (193). Moreover, in a meta-analysis of 27 randomized controlled trials with an average duration of 64 weeks that compared the efficacy and safety of SGLT2 inhibitors to a placebo in 20,895 participants there was no increased risk of fractures with SGLT2 inhibitor treatment (RR 1.02; 95% CI 0.81- 1.28) (194). Similarly, a meta-analysis of 10 large outcome studies also did not observe an increase in fractures (RR 1.03; 95% CI 0.95- 1.12) (181).

 

Several studies have examined the effect of SGLT2 inhibitors on bone mineral density. Canagliflozin was associated with a decrease in total hip bone mineral density over 104 weeks, (placebo-subtracted changes:100mg -0.9% and 300mg -1.2%), but did not result in changes in bone mineral density in the femoral neck, lumbar spine, or distal forearm (195). In a 2-year study dapagliflozin did not significantly affect bone mineral density at the lumbar spine, femoral neck, or total hip (196). In a 26-week study ertugliflozin also had no adverse effect on bone mineral density (197).

 

Thus, the evidence that SGLT2 inhibitors increase the risk of osteoporosis and fractures, with the possible exception of canagliflozin, is not very strong. One should recognize though, that the hypovolemia and hypotension could increase the risk of falls and thereby increase the risk of fractures in susceptible individuals.

 

AMPUTATIONS

 

In the CANVAS study described above, canagliflozin was associated with an increased risk of amputations (hazard ratio, 1.97; 95% CI, 1.41 to 2.75), which were primarily at the level of the toe or metatarsal (133). Amputation risk was strongly associated with baseline history of prior amputation and risk factors for amputation (peripheral vascular disease and neuropathy). The risk of amputation was low with 6.3 of participants per 1000 patients-years in the canagliflozin group having an amputation vs. 3.4 in the placebo group. The basis for the increase in amputations is unknown.

 

However, the EMPA-REG OUTCOME trial with empagliflozin, the DECLARE-TIMI 58 trial with dapagliflozin, and the VERTIS CV trial with ertuglifozin did not report an increase in amputations in the patients treated with an SGLT2 inhibitor (132,143,148,198). Moreover, in the CREDENCE trial, canagliflozin also did not cause an increase in amputations in the patients treated with the SLGT2 inhibitor (141). In a meta-analysis of 7 large cardiovascular/renal outcome trials described above (excluding CANVAS) there was no increased risk of amputations in the SGLT2 inhibitor treated group vs. placebo group (RR 1.09; CI 95% 0.94-1.26) (199). Given that only one of eight large randomized trials has demonstrated an increased risk of amputations it is unlikely that SGLT2 inhibitors significantly increase the risk of amputations.

 

Nevertheless, before initiating SGLT2 inhibitor therapy one should consider factors in the patient history that may predispose them to the need for amputations, such as a history of prior amputation, peripheral vascular disease, severe neuropathy, and diabetic foot ulcers and weigh the risks and benefits of therapy (package insert). 

 

ACUTE ILLNESS

 

Because of the risk of hypovolemia, hypotension, and DKA the administration of SGLT2 inhibitors should be suspended during acute illness or planned surgical procedures. SGLT2 inhibitor therapy may be resumed following recovery.

 

This view needs to be modified based on the results of the DARE 19 study (200). In this study patients hospitalized with COVID-19 and with at least one cardiometabolic risk factor (i.e., hypertension, type 2 diabetes, atherosclerotic cardiovascular disease, heart failure, and chronic kidney disease) were randomized to dapagliflozin 10 mg daily or placebo for 30 days. While dapagliflozin did not result in a statistically significant risk reduction in organ dysfunction or death, or improvement in clinical recovery, the drug was well tolerated indicating that SGLT2 inhibitors can be safely given to hospitalized patients if there are strong indications for their use.

 

Contraindications and Drug Interactions

 

RENAL FUNCTION

 

The dose of SGLT2 inhibitors needs to be adjusted based on renal function. Therefore, renal function needs to be assessed prior to initiating therapy and periodically thereafter.

 

Dosage recommendations for dapagliflozin and canagliflozin are shown in tables 16 and 17.

 

 Table 16. Dose Recommendations for Dapagliflozin

eGFR > 45

To improve glycemic control, the recommended starting dose is 5 mg orally once daily. Dose can be increased to 10 mg orally once daily for additional glycemic control*. For all other indications, the recommended starting dose is 10 mg orally once daily.

eGFR 25-45

10 mg orally once daily*

eGFR < 25

Initiation is not recommended; however, patients may continue 10 mg orally once daily to reduce the risk of eGFR decline, ESKD, CV death, and heart failure.

Dialysis

Contraindicated

 

Table 17. Dose Recommendations for Canagliflozin

eGFR > 60

100 mg orally once daily, taken before the first meal of the day. Dose can be increased to 300 mg once daily for additional glycemic control.

eGFR 30-60

100 mg once daily.

eGFR < 30

Initiation is not recommended, however patients with albuminuria greater than 300 mg/day may continue 100 mg once daily to reduce the risk of ESKD, doubling of serum creatinine, CV death, and hospitalization for heart failure

Dialysis

Contraindicated

 

Empagliflozin is not recommended for glycemic control in patients with an eGFR < 30 and is contraindicated in patients on dialysis. Data are insufficient to provide a dosing recommendation in patients who have type 2 diabetes and established cardiovascular disease with an eGFR less than 30 or who have heart failure with reduced ejection fraction with an eGFR less than 20.

 

Ertugliflozin is not recommended in patients with an eGFR less than 45 and is contraindicated in patients on dialysis.

 

Summary

 

SGLT2 inhibitors are effective at lowering glucose levels and even more importantly have beneficial effects on heart failure and renal disease. They have a number of potential side effects but many are not definitively associated with SGLT2 inhibitors (fractures, urinary tract infections, amputations) or are rare (DKA, Fournier’s gangrene). The major side effect is genital mycotic infections, which usually are mild and respond to treatment. In patients with pre-existing cardiovascular disease, at high risk for cardiovascular disease particularly heart failure, or with renal disease SGLT2 inhibitors are a leading therapeutic choice.

 

Table 18. Advantages and Disadvantages of SGLT2 Inhibitors

Advantages

Disadvantages

Weight loss

Urinary Tract Infections?

No hypoglycemia

Genital Mycotic Infections

Decrease heart failure

Increased LDL (small increase)

Decreases renal dysfunction

Increased risk of DKA

Once a day administration

Postural hypotension/volume depletion

Decrease BP

Fractures/ Osteoporosis?

 

Increased risk amputations (canagliflozin)?

 

Fournier’s gangrene (rare)

 

Expensive

 

 

COMBINATION SGLT1 AND SGLT2 INHIBITORS

 

Introduction

 

Sotagliflozin (Zynquista) inhibits both SGLT1 and SGLT2 (201). Sotaglifozin’s effectiveness in inhibiting SGLT-2 is similar to that of the selective SGLT-2 inhibitors discussed above but it is > 10-fold more potent in inhibiting SGLT-1(202). Sotagliflozin was approved in Europe for the treatment of patients with type 1 diabetes but is no longer available. In the US the drug was not approved. It was used in overweight patients (BMI> 27 kg/m2) when optimal insulin on its own does not achieve adequate glycemic control (package insert- https://www.ema.europa.eu/en/documents/product-information/zynquista-epar-product-information_en.pdf).

 

Administration

 

The recommended dose of sotagliflozin was 200mg once a day before the first meal of the day. After 3 months, the dose may be increased to 400mg once a day if additional blood sugar control is needed (package insert).

 

Because of an increased risk of diabetic ketoacidosis precautions should be taken to reduce this potential side effect. It is recommended that patients obtain several baseline blood or urine ketone levels over one to two weeks prior to initiation of sotagliflozin therapy and patients should become familiar with how their behaviors and circumstances affect their ketone levels. During the initial one to two weeks of treatment with sotagliflozin, ketones should be monitored on a regular basis. Measurement of blood ketone levels is preferred over urine (package insert).

 

In order to avoid hypoglycemia with the first dose of sotagliflozin a 20% reduction in the first mealtime bolus insulin may be considered (package insert). Subsequent bolus doses should be adjusted individually based on blood glucose results. No reduction in basal insulin is recommended when initiating sotagliflozin. Subsequently, basal insulin should be adjusted based on blood glucose results. When needed, insulin dose reduction should be done cautiously to avoid ketosis and DKA (package insert).

 

Mechanism of Action

 

The mechanism by which inhibition of SGLT2 decreases glucose levels was discussed in the prior section on SGLT2 inhibitors. Inhibition of SGLT1 will have additional effects. In the kidney SGLT1 is responsible for approximately 10% of the transport of luminal glucose and thus inhibiting SGLT1 may facilitate SGLT2 induced loss of glucose in the urine (201,203). Moreover, SGLT1 is expressed in the small intestine and facilitates the absorption of dietary glucose (201,203,204). SGLT1 expression in the small intestine is increased in patients with diabetes (203,204). Inhibition of SGLT1 delays, and perhaps reduces, glucose absorption, and enhances circulating levels of GLP-1 reducing post-prandial glucose excursions (201,204-206). Finally, SGLT1 is expressed in human heart capillaries and whether this plays a role in cardiac protection remains to be determined (202).  

 

Glycemic Efficacy

 

TYPE 1 DIABETES (T1DM)

 

The inTandem1 trial was carried out in North American adults and randomized patients with T1DM to placebo (n = 268), sotagliflozin 200 mg (n = 263), or sotagliflozin 400 mg (n = 262) (207). Baseline A1c was 7.57% and the placebo-adjusted A1c reductions were 0.36% and 0.41% with sotagliflozin 200 and 400 mg, respectively, at 24 weeks and 0.25% and 0.31% at 52 weeks (all P < 0.001). At 52 weeks the difference in body weight between the placebo group and 400mg sotagliflozin group was -4.32 kg (-5.00 to -3.64). Notably hypoglycemia was not increased with sotagliflozin treatment. However, DKA occurred more frequently with sotagliflozin treatment (placebo 0.4%, sotagliflozin 200mg 3.4%, sotagliflozin 400mg 4.2%).

 

The inTandem2 trial was carried out in European adults and randomized patients with T1DM to placebo (n = 258), oral sotagliflozin 200 mg (n = 261), or 400 mg (n = 263) (208). Baseline A1c was 7.7% and the placebo-adjusted A1c reductions were 0.37% and 0.35% with sotagliflozin 200 and 400 mg, respectively, at 24 weeks and 0.21% and 0.37% at 52 weeks (all P < 0.001). At 52 weeks the difference in body weight between the placebo group and 400mg sotagliflozin group was −2.92 kg (-3.62 to −2.22). Hypoglycemia was not increased with sotagliflozin treatment. DKA occurred more frequently with sotagliflozin treatment (placebo 0%, sotagliflozin 200mg 2.3%, sotagliflozin 400mg 3.4%).

 

The inTandem3 trial was a multicenter world-wide study in patients with T1DM randomized to placebo (n=703) or sotagliflozin 400mg (n=699) for 24 weeks (209). The baseline A1c was 8.2% and sotagliflozin decreased A1 by −0.46% compared to placebo. Hypoglycemia with a blood glucose level < 55 mg/dL was significantly lower in the sotagliflozin group than in the placebo group (11.8 per person-year vs. 15.4 per person-year) but severe hypoglycemia (episode needing assistance from another person or resulting in loss of consciousness or a seizure) was similar. Notably the risk of DKA was increase with sotagliflozin treatment (sotagliflozin 3.0% and placebo 0.6%).

 

Thus, in patients with T1DM sotagliflozin causes a modest reduction in A1c and body weight but increases the risk of DKA.

 

TYPE 2 DIABETES

 

Studies of the effect of sotagliflozin on glycemic control in patients with T2DM have not been as extensive as in patients with T1DM. In a 12-week trial that compared placebo (n= 60), canagliflozin 200mg (n= 60), or sotagliflozin 400mg (n= 60) in patients with T2DM on metformin monotherapy a decrease in A1c of -0.09%, -0.50, and -0.92% occurred in patients treated with placebo, sotagliflozin 200mg, and sotagliflozin 400mg, respectively (210). As expected, there was a decrease in body weight and an increase in urinary glucose excretion with sotagliflozin treatment. Of note a study has shown that in patients with T2DM sotagliflozin treatment is effective in lowering postprandial glucose levels even in patients with an eGFR < 45 mL/min/1.73 m2 (211).

 

Other Effects

 

CARDIOVASCULAR

 

The SOLOIST-WHF Trial was a multicenter trial in which patients with T2DM who were recently hospitalized for worsening heart failure were randomly assigned to receive sotagliflozin 200 mg once daily (with a dose increase to 400 mg, depending on side effects) (n= 608), or placebo (n= 614) (212). The primary end point was the total number of deaths from cardiovascular causes and hospitalizations and urgent visits for heart failure (first and subsequent events).  Because of loss of funding from the sponsor the study was stopped early and the median duration of follow-up was 9 months. The primary end-point was reduced in the sotagliflozin group vs. placebo group (HR 0.67; 95% CI, 0.52 to 0.85; P<0.001) as was hospitalizations or urgent visits for heart failure (HR 0.64; 95% CI, 0.49 to 0.83: P <0.001). Of particular note benefit was observed in patients with reduced or preserved ejection fractions (<50% or ≥50%). This study demonstrates benefits in patients with a reduced or preserved ejection fractions and that treatment initiated during an acute heart failure episode is beneficial. DKA was uncommon in both the sotagliflozin group (0.3%) and placebo group (0.7%) but severe hypoglycemia was increased (sotagliflozin 1.5% vs placebo 0.3%).

 

The SCORED trial was a multicenter trial in which patients with T2DM and chronic kidney disease (eGFR- 25 to 60 ml/min/1.73 m2, albuminuria was not required), and risks for cardiovascular disease were randomized to sotagliflozin (200 mg once daily, with an increase to 400 mg once daily if unacceptable side effects did not occur) (n= 5292) or placebo (n= 5292) and followed for a median of 16 months (213). The primary end point was the composite of the total number of deaths from cardiovascular causes, hospitalizations for heart failure, and urgent visits for heart failure. Sotagliflozin treatment decreased the primary end point (HR 0.74; 95% CI, 0.63–0.88; P <0.001), hospitalizations or urgent visits for heart failure (HR 0.67; 95% CI, 0.55–0.82; P <0.001), and deaths from cardiovascular causes, nonfatal myocardial infarctions, and nonfatal strokes (HR 0.77; 95%CI 0.65–0.91). There was a trend towards a reduction in a renal composite endpoint consisting of a decrease of ≥50% in the eGFR from baseline for ≥30 days, long-term dialysis, renal transplantation, or sustained eGFR of <15 ml/min/1.73 m2 for ≥30 days (HR 0.71; 95% CI, 0.46–1.08). DKA while infrequent was increased in the sotagliflozin group (0.6% vs 0.3%; P=0.02).

 

RENAL

 

While outcome studies are not available in patients with T1DM it has been shown that sotagliflozin has effects on kidney function that are similar to what has been observed in patients with T2DM. Specifically, there was an acute decrease in eGFR and a decrease in albuminuria (214). These observations suggest that the beneficial effects on renal outcomes seen with SGLT2 inhibitors in patients with T2DM and non-diabetics will also occur in patients with T1DM.

 

Side Effects

 

The side effects of sotagliflozin are similar to those described previously for SGLT2 inhibitors. In addition, sotagliflozin also causes diarrhea and flatulence due to the inhibition of SGLT1 mediated glucose uptake in the small intestine.

 

Contraindications and Drug Interactions

 

Patients at high risk for DKA should not be started on sotagliflozin.

 

Summary

 

In patients with T1DM sotagliflozin modestly reduces A1c levels and body weight but increases the risk of DKA. Because of the increased risk of DKA this drug is no longer approved for use.

 

In patients with T2DM sotagliflozin use is not approved in the US or Europe. While studies have shown beneficial effects of sotagliflozin on cardiovascular disease it is not clear whether this benefit is solely due to inhibition of SGLT2 or whether inhibition of SGLT1 plays a significant role. 

 

DOPAMINE AGONIST (CYCLOSET)

 

Introduction

 

In 2009, a quick-release formulation of bromocriptine (Cycloset, bromocriptine-QR) was approved to improve glycemic control in patients with T2DM (215,216). Bromocriptine is a centrally-acting dopamine D2 receptor agonist that has been used for many years for the treatment of hyperprolactinemia and Parkinson’s disease (215,216). It can be used to improve glycemic control in patients with T2DM either as monotherapy or in combination with other hypoglycemic drugs (215,216)

 

Administration

 

Bromocriptine-QR should be initiated at one tablet (0.8 mg) within two hours after waking in the morning. The dose can be increased by one tablet per week until a maximum daily dose of 6 tablets (4.8 mg) or until the maximal tolerated number of tablets between 2 and 6 per day is reached. Taking bromocriptine-QR with food is recommended to decrease gastrointestinal side effects (215).

 

Mechanism of Action

 

Bromocriptine-QR decreases insulin resistance resulting in an increase in glucose disposal and a decrease in hepatic glucose production (215). Bromocriptine-QR does not increase insulin levels (215). Thus, the effectiveness of bromocriptine-QR will be greatest in patients that are insulin resistant and produce insulin (215). Based on animal studies it is thought that bromocriptine-QR acts on the central nervous system, particularly the hypothalamus, to increase insulin sensitivity in liver, muscle, and adipose tissue (215).

 

Glycemic Efficacy

 

In a 24 week monotherapy study the A1c level was 0.4% lower in the bromocriptine-QR group compared to placebo group (217).  Both fasting and postprandial glucose levels were decreased with bromocriptine-QR treatment (217). Bromocriptine-QR treatment was associated with a decrease in triglyceride levels (32 mg/dL) but no significant change in LDL or HDL cholesterol levels or change in body weight (217). A trial adding bromocriptine-QR to sulfonylurea therapy demonstrated a 0.55% lower A1c in the bromocriptine-QR group compared to placebo (217). As in the monotherapy study fasting glucose, postprandial glucose, and triglyceride levels were decreased with no change in LDL or HDL cholesterol levels (217). Addition of bromocriptine-QR to other hypoglycemic drugs including insulin results in an approximate decrease in A1c of 0.5 to 1.0% (215,216). Hypoglycemia is a rare side effect with use of bromocriptine-QR alone, but is increased with use of insulin secretagogue therapy or insulin (216,217).

 

Other Effects

 

BLOOD PRESSURE

 

Bromocriptine-QR modestly decreases systolic and diastolic blood pressure (216,217).

 

LIPIDS

 

Bromocriptine-QR treatment decreases triglyceride levels but has no significant effect on LDL or HDL cholesterol levels (216,217). The decrease in triglyceride levels is thought to be due to a decrease in hepatic triglyceride synthesis, likely due to a decrease in adipose tissue lipolysis resulting in decreased blood free fatty acid levels and decreased delivery of fatty acids to the liver for triglyceride synthesis (215).  

 

CARDIOVASCULAR DISEASE

 

A 52-week, randomized, double-blind, multicenter trial evaluated cardiovascular safety in 3,095 patients with T2DM treated with bromocriptine-QR or placebo (218).  The composite end point of first myocardial infarction, stroke, coronary revascularization, or hospitalization for angina or congestive heart failure occurred in 1.8% of the bromocriptine-QR treated vs. 3.2% of the placebo-treated patients resulting in a 40% decrease in cardiovascular events (HR 0.60; CI 0.37– 0.96). Clearly further studies to confirm this finding and to elucidate the mechanism of this beneficial effect are required.

 

Side Effects

 

The most common side effect of bromocriptine-QR therapy is nausea which is usually transient and improves with time (216,217). This side effect can be minimized by reducing the dose (216,217). In the pooled phase 3 trials adverse events leading to discontinuation occurred in 539 (24%) of the bromocriptine-QR treated patients and 118 (9%) of the placebo-treated patients. This between-group difference was driven mostly by gastrointestinal adverse events, particularly nausea (package insert). Similarly, in the bromocriptine-QR safety trial adverse events leading to discontinuation of drug occurred in 24% of the bromocriptine-QR treated patients and 15% of the placebo-treated patients, a difference again driven mostly by gastrointestinal adverse events, particularly nausea (package insert).

 

Hypotension resulting in syncope can occur particularly in patients on anti-hypertensive medications (package insert). Other side effects include somnolence, fatigue, vomiting, headache, and dizziness (package insert).

 

Contraindications and Drug Interactions

 

Bromocriptine-QR is metabolized by the Cyp3A4 system and therefore the drug should not be used with strong CYP3A4 inhibitors (e.g., azole antimycotics, HIV protease inhibitors) and the dose should not exceed 1.6 mg once daily during concomitant use of a moderate CYP3A4 inhibitor (e.g., erythromycin) (package insert).

 

Bromocriptine-QR is contraindicated in patients with syncopal migraine because it increases the likelihood of a hypotensive episode (package insert). The use of bromocriptine-QR in patients with severe psychotic disorders in not recommended as it may exacerbate the disorder or diminish the effectiveness of drugs used to treat the disorder (for example clozapine, olanzapine, ziprasidone) (package insert).

 

Summary

 

Bromocriptine-QR has modest effects on A1c levels by decreasing insulin resistance. In clinical trials the drug was often discontinued due to nausea. Because of the modest effects on A1c and the prominent side effects this drug is not widely used in the treatment of patients with T2DM. If further studies confirmed the decrease in cardiovascular events in patients treated with bromocriptine-QR the use of this drug would increase.

 

Table 19. Advantages and Disadvantages of Bromocriptine-QR

Advantages

Disadvantage

Decreases triglycerides

Need to titrate dose

Once a day dosing

Modest effect on A1c

Cardiovascular benefits?

Frequent discontinuation due to GI side effects

Decrease BP

Expensive

Neutral weight effect

 

Hypoglycemia uncommon

 

 

OVERVIEW OF THE INCRETIN SYSTEM

 

The incretin effect refers to a greater insulin stimulatory effect after an oral glucose load than from an intravenous glucose infusion when plasma glucose concentrations are matched (219). Thus, glucose and other nutrients delivered via the gastrointestinal tract potentiates the ability of the beta cells in the pancreas to produce insulin resulting in greater insulin secretion than with IV glucose (220). The increase in insulin levels with IV glucose is only approximately one‐third of that elicited by oral glucose. The majority of the incretin effect is due to two GI hormones, glucose-dependent insulinotropic peptide (GIP) and glucagon like peptide-1 (GLP-1) with GIP having a dominant role (Figure 9) (219). The basal plasma levels of the incretin hormones are low but after eating the levels increase reaching concentrations that augment the insulin secretory responses if glucose levels are high but are ineffective at low glucose concentrations (i.e. glucose dependent effect) (219).

 

Patients with T2DM have a significant reduction of the incretin effect but GLP-1 and GIP levels in the blood after meals are not reduced in patients with T2DM (219). Rather decreased functional beta cell mass and resistance to the effects of GLP-1 and GIP in patients with T2DM accounts for the decreased incretin effect (219). Infusion of GIP has a minimal response on insulin secretion in patients with T2DM (resistance to effect of GIP) whereas GLP-1 administration is able to stimulate insulin secretion but the response is reduced in patients with T2DM compared to normal individuals likely secondary to decreased functional beta cell mass (219). Achieving near-normoglycemia by intensified insulin regimens improved beta cell responsiveness to exogenous GIP and GLP-1, although the increase in insulin secretion was still much lower than in individuals without diabetes (219). The reduced incretin effect in patients with T2DM occurs after the diagnosis of diabetes is established, suggesting this abnormality is secondary to the diabetic state rather than the cause of diabetes (220).   

 

Glucagon Like Peptide-1 (GLP-1)

 

GLP-1 is cleaved from the pro-glucagon molecule by pro-hormone convertase enzymes in the intestine (220). GLP-1 is stored in the L-cells of the intestine, predominantly in the ileum and colon, and is released at mealtime in response to neurohormonal signals and the presence of food in the gut (219,220). GLP-1 affects postprandial glucose levels through several mechanisms, including enhancing insulin secretion by the beta cells and inhibiting postprandial glucagon secretion by the alpha cells in a glucose-dependent manner (i.e. GLP-1 does not stimulate insulin secretion or inhibit glucagon secretion unless glucose levels are elevated) (220). This glucose dependent effect accounts for why incretin-based drugs do not cause serious hypoglycemia. Activation of GLP-1 receptors on beta cells increases cAMP levels, which potentiates insulin release in the presence of elevated glucose concentrations. In addition, GLP-1 slows the rate of gastric emptying, which is often paradoxically accelerated in patients with diabetes (220). GLP-1 also acts as a postprandial satiety signal through neurohormonal networks that signal the brain to suppress appetite and food intake, which can lead to weight loss (220). Animal studies suggest that exogenous GLP-1 has the ability to increase islet size, enhance beta-cell proliferation, inhibit beta-cell apoptosis, and regulate islet growth (221). The administration of GLP-1 intravenously increases insulin secretion, reduces glucagon secretion, and decreases glucose levels during fasting and in the post-prandial state (219). GLP-1 is rapidly degraded by dipeptidyl peptidase 4 (DPP-4) into inactive peptides (half-life is minutes) (Figure 9).

 

Figure 9. Incretin Hormone Secretion and Effect on Pancreas

Glucose-Dependent Insulinotropic Peptide (GIP)

 

Within minutes after ingestion of food, GIP is secreted from the K-cells located in the proximal region of the jejunum (219,220). GIP helps maintain normal glucose homeostasis by stimulating an increase in insulin secretion by the beta cells (Figure 9). Studies have suggested that the increase in insulin with food intake (Incretin effect) is primarily mediated by GIP (219). In contrast to GLP-1, GIP does not inhibit glucagon secretion, and in fact may stimulate glucagon secretion during euglycemic states. Additionally, GIP has no effect on gastric emptying or on satiety. GIP concentrations in patients with T2DM are either normal or slightly increased following a meal indicating that the failure to secrete is not the explanation for the decreased incretin effect. Rather, beta cells in patients with T2DM are resistant to GIP. GIP is rapidly degraded by DPP-4 into inactive peptides (half-life is minutes) (Figure 9). The characteristics of GLP-1 and GIP are shown in table 20.

 

Table 20. Characteristics of GLP-1 and GIP

 

GLP-1

GIP

Post meal levels in patients with diabetes

Normal

Normal

Effect on insulin secretion

Stimulates

Stimulates

Effect on glucagon secretion

Inhibits

No effect or stimulates

Gastric emptying

Delays

No effect

Satiety

Induces

No effect

Degradation by DPP-4

Yes

Yes

 

DIPEPTIDYL PEPTIDASE-4 (DPP-4) INHIBITORS

 

Introduction

 

The currently available DPP-4 inhibitors in the US are sitagliptin (Januvia), saxagliptin (Onglyza), linagliptin (Tradjenta), and alogliptin (Nesina). Vidigliptin (Galvus) is available in Europe (222). DPP-4 inhibitors can be used as monotherapy, dual therapy, triple drug therapy, or in combination with insulin (222). These drugs are very similar and the minor differences will be discussed below.

 

Administration

 

The recommended dose of sitagliptin is 100 mg once daily with or without food. In patients with moderate renal impairment (eGFR greater than or equal to 30 mL/min/1.73 m2 but less than 45 mL/min/1.73 m2), the dose of sitagliptin is 50 mg once daily. In patients with severe renal impairment (eGFR less than 30 mL/min/1.73 m2) the dose of sitagliptin is 25 mg once daily.

 

The recommended dosage of saxagliptin is 2.5 mg or 5 mg once daily with or without food. In patients with a creatinine clearance CrCl ≤50 mL/min the dose of saxagliptin is 2.5 mg.

 

The recommended dose of linagliptin is 5 mg once daily with or without food. No dose adjustment is required for decreased renal function.

 

The recommended dose of alogliptin is 25 mg once daily with or without food. The dose of alogliptin is 12.5 mg once daily for patients with moderate renal impairment (CrCl ≥30 to <60 mL/min) and 6.25 mg with severe renal impairment (CrCl <30 mL/min).

 

Renal function should be checked prior to initiating treatment and periodically because dose adjustments are required for all DPP-4 inhibitors except linagliptin.

 

Mechanism of Action

 

DPP-4 inhibitors increase the concentration and activity of the endogenous incretins, GLP-1 and GIP, by inhibiting the proteolytic cleavage of these hormones by DPP-4, into inactive molecules (222).   As discussed above, GLP-1 is secreted by L-cells in the intestines and stimulates insulin secretion and suppresses glucagon secretion in a glucose dependent manner, inhibits gastric emptying, and has central anorexic activity that decreases food intake. GIP is secreted by the K cells in the proximal intestine and stimulates insulin secretion in a glucose dependent manner.

 

An increase in active GLP-1 and GIP potentiates glucose induced insulin secretion and an increase in GLP-1 inhibits glucagon secretion (222). Together an increase in insulin and a decrease in glucagon will result in a decrease in blood glucose levels. Of note, DPP-4 inhibition results in a 2–3-fold increase in postprandial active GLP-1 levels, which is not at a level that delays gastric emptying or increases satiety and induces weight loss. This is in contrast to GLP-1 receptor agonist administration that results in marked elevations in active GLP1 activity that is equivalent to a >10-fold increase in GLP-1, which can delay gastric emptying and increase satiety.

 

Glycemic Efficacy

 

DPP-4 inhibitors typically reduce A1c levels by 0.5-1.0% and are less effective in lowering A1c compared to metformin, TZDs, SGLT2 inhibitors, and GLP-1 receptor agonists (Table 6) (12,16,222). With regards to sulfonylureas, studies have shown a greater decrease in A1c with sulfonylureas compared to DPP-4 inhibitors in short term studies but in studies greater than one year the effect of sulfonylureas and DPP-4 inhibitors on A1c were similar (16,222). The ability of DPP-4 inhibitors to lower A1c is similar in monotherapy and when DPP-4 inhibitors are used in combination with other drugs (16,222). The decrease in A1c is similar for the different DPP-4 inhibitors (12,16). DPP-4 inhibitors are effective in lowering postprandial glucose levels. Because of their mechanism of action, DPP-4 inhibitors do not cause hypoglycemia but can potentiate the hypoglycemia induced by insulin or sulfonylureas (16,222). An adjustment in the dose of sulfonylureas or insulin may be required to reduce the risk of hypoglycemia.

 

Other Effects

 

WEIGHT

 

DPP-4 inhibitors are weight neutral (16,222).

 

BLOOD PRESSURE

 

A meta-analysis of 15 trials involving 5,636 participants found that DPP-4 inhibitors compared to placebo reduced systolic BP (mean difference, -3.04  mmHg: P < 0.00001) and diastolic BP (mean difference, -1.47 mmHg; P < 0.00001) (223).

 

LIPIDS

 

DPP-4 inhibitors decrease postprandial triglycerides by reducing circulating chylomicrons by decreasing intestinal lipoprotein production while having minimal effects on fasting lipid levels (224).

 

CARDIOVASCULAR DISEASE

 

The effect of the DPP-4 inhibitors saxagliptin, alogliptin, sitagliptin, and linagliptin on cardiovascular endpoints has been reported. In the saxagliptin study (SAVOR‐TIMI 53 trial), 16,492 patients with T2DM who had a history of cardiovascular events or who were at high risk were randomized to saxagliptin or placebo for 2.1 years (225). Saxagliptin did not increase or decrease cardiovascular death, myocardial infarction, or ischemic stroke. Interestingly more patients treated with saxagliptin were admitted to the hospital for heart failure. The risk of heart failure with saxagliptin was greatest in patients at a high overall risk of heart failure (i.e., history of heart failure, impaired renal function, or elevated baseline levels of NT-proBNP) (226). Additionally, in the patients treated with saxagliptin the increase in heart failure was an early event with a 6-month rate of 1.1% vs. 0.6% in the placebo group (HR 1.80, p=0·001) and a 12-month rate of 1·9% vs. 1·3% (1.46; p=0.002) (226). In contrast, after 12 months no difference in the rate of heart failure was observed in the saxagliptin and placebo groups indicating that the development of heart failure is an early event (226)

 

In the alogliptin trial (EXAMINE), 5,380 patients with either an acute myocardial infarction or unstable angina within the previous 15-90 days were randomized to alogliptin or placebo and followed for a median of 18 months (227). As seen in the saxagliptin study the rates of cardiovascular events (death from cardiovascular causes, non-fatal myocardial infarction, or non-fatal stroke) were similar in the alogliptin and placebo groups. The risk of hospitalization for heart failure was not statistically increased in the entire subset of patients treated with alogliptin (228). However, the hazard ratio for the subgroup of patients without heart failure at baseline was 1.76, p=0.026) (228).

 

In the sitagliptin trial (TECOS), 14,671 patients with established cardiovascular disease were randomized to sitagliptin or placebo for 3 years (229). Sitagliptin did not decrease the risk of major adverse cardiovascular events or increase hospitalization for heart failure. Finally, in the linagliptin trial (CARMELINA), 6,979 patients at high risk for cardiovascular disease were randomized to linagliptin or placebo for a median follow-up of 2.2 years (230). As in the other DPP-4 inhibitor studies, linagliptin did not have a beneficial effect on cardiovascular events. Additionally, linagliptin did not increase the risk of hospitalization for heart failure (231).

 

Thus, these results indicate that DPP-4 inhibitors do not reduce cardiovascular disease. Whether specific DPP-4 inhibitors (saxagliptin, alogliptin) increase the risk of heart failure remains to be resolved. Of note, a meta-analysis of 30 randomized controlled trials involving 29,938 patients comparing the effects of saxagliptin vs. placebo or sulfonylureas did not observe an increase in heart failure (RR 0.99, 95% CI 0.89 to 1.10; p = 0.85) (232).

 

RENAL DISEASE

 

Changes in renal function were examined in the large cardiovascular outcome trials described above. In the SAVOR-TIMI 53 trial treatment with saxagliptin decreased albuminuria but had no effect on eGFR (233). Saxagliptin reduced the development of macroalbuminuria independent of changes in A1c levels (225,233). Doubling of serum creatinine, initiation of chronic dialysis, renal transplantation, or serum creatinine >6.0 mg/dL, were similar in the saxagliptin and placebo groups (233). In the TECOS trial treatment with sitagliptin also reduced the urinary albumin to creatinine ratio with no effect on eGFR (234). In the CARMELINA trial many of the patents had pre-existing renal disease (74% of patients had prevalent diabetic kidney disease, 43% had an eGFR below 45 mL/min/1.73 m2, 15.2% had an eGFR below 30 mL/min/1.73 m2 and 80% had a urinary albumin creatinine ratio >30 mg/g) (230). Treatment with linagliptin reduced the progression of albuminuria but had no effect on death due to renal failure, ESRD, or sustained 40% or higher decrease in eGFR from baseline (230).

 

Taken together these studies indicate that DPP-4 inhibitors decrease proteinuria but do not provide data suggesting an improvement or delay in worsening of renal function.

 

Side Effects

 

DPP-4 inhibitors have been safe drugs with minimal side effects and are well tolerated by patients. Very rarely hypersensitivity reactions including urticaria, facial edema, anaphylaxis, angioedema, and exfoliative skin conditions including Stevens-Johnson syndrome have occurred (package insert). Bullous pemphigoid has also rarely been associated with DPP-4 inhibitor treatment (package insert).

 

ACUTE PANCREATITIS

 

The package insert of DPP-4 inhibitors indicates that acute pancreatitis is a complication of DPP-4 inhibitor treatment. The individual results of the SAVOR–TIMI, EXAMINE, and TECOS trials discussed above did not show an increased risk of pancreatitis or pancreatic cancer. However, two meta-analyses of these studies demonstrated an 80% increased risk of acute pancreatitis in patients using DPP-4 inhibitors compared with those receiving standard care (235,236). It should be noted that the absolute risk was small (0.13%), which would result in one to two additional cases of acute pancreatitis for every 1,000 patients treated for 2 years (236). Thus, pancreatitis appears to be a rare side effect of DPP-4 inhibitors. In patients on DPP-4 inhibitors who have GI symptoms suggestive of pancreatitis further evaluation is indicated. The diagnosis of acute pancreatitis requires the presence of two of the following three criteria: acute onset of persistent, severe, epigastric pain often radiating to the back, elevation in serum lipase or amylase to three times or greater than the upper limit of normal, and characteristic findings of acute pancreatitis on imaging (237). 

 

ARTHRALGIA

 

Severe and disabling arthralgia in patients taking DPP-4 inhibitors has been reported (238). The time to onset of symptoms following initiation of drug therapy varied from one day to years. Patients experienced relief of symptoms upon discontinuation of the medication and a subset of patients experienced a recurrence of symptoms when restarting the same drug or a different DPP-4 inhibitor. If a patient develops severe joint pain discontinue the DPP-4 inhibitor.

 

Contraindications and Drug Interactions

 

It is unknown whether patients with a history of pancreatitis or who are at increased risk for the development of pancreatitis should be started on DPP-4 inhibitors. Given the availability of other hypoglycemic drugs many clinicians avoid the use of DPP-4 inhibitors in these patients.

 

The dosage of saxagliptin is 2.5 mg once daily when co-administered with a strong cytochrome P450 3A4/5 inhibitor (e.g., ketoconazole, atazanavir, clarithromycin, indinavir, itraconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin) (package insert).

 

Summary

 

DPP-4 inhibitors, while not the most potent drugs at lowering A1c, nevertheless are very attractive to use in the treatment of patients with T2DM as they are safe drugs that do not have many side effects. They do not cause hypoglycemia, weight gain, or cardiovascular disease. Unfortunately they do not reduce the risk of cardiovascular disease or prevent loss of renal function.

 

Table 21. Advantages and Disadvantages of DPP-4 Inhibitors

Advantages

Disadvantages

No hypoglycemia

Pancreatic disease

Weight neutral

Heart failure (saxagliptin/alogliptin)?

Decreases postprandial glucose

Arthritis

Once a day

Bullous pemphigoid

Well tolerated

Relatively expensive

Decreases BP

Modest glycemic lowering

 

INJECTABLE GLUCAGON LIKE PROTEIN-1 (GLP-1) RECEPTOR AGONISTS

 

Introduction

 

There are currently six GLP-1 receptor agonists available in the US, three drugs administered daily and three drugs administered weekly (Figure 10). Albiglutide (Tanzeum) was withdrawn from the market for commercial reasons and is no longer available. GLP-1 receptor agonists can be used in combination with multiple oral anti‐diabetic drugs or in combination with insulin (239). The concentrations of GLP-1 receptor agonist activity are much higher than physiological levels of GLP-1 activity (16). The GLP-1 receptor agonists that a similar to exendin-4 (Exenatide and Lixisenatide) are eliminated by the kidneys and therefore in patients with severe renal disease these drugs are contraindicated (16). In contrast, the drugs that are analogues of GLP-1 are degraded by peptidases (16).

 

Figure 10. Structure of GLP-1 Receptor Agonists

SHORT ACTING GLP-1 RECEPTOR AGONISTS

 

Exenatide (Byetta) is a synthetic exendin-4 that is a peptide originally isolated from the saliva of the Gila monster that has a 53% homology with human GLP-1 and is resistant to degradation by DPP-4 (16,239). Lixisenatide (Adylyxin) is an exendin-4 analogue with six Lys residues added at the C terminus to confer resistance to DPP-4 (16,239).

 

LONG ACTING GLP-1 RECPTOR AGONISTS

 

Even though liraglutide (Victoza) is administered daily it is considered a long acting GLP-1 receptor agonist because its effects on fasting glucose levels are similar to weekly GLP-1 receptor agonists and its effects on gastric emptying wane as seen with weekly GLP-1 receptor agonists. Liraglutide is an analogue of GLP-1 with the addition of a 16-carbon fatty acid chain that masks the DPP-4 cleavage site preventing degradation (8,179). Once weekly exenatide (Bydureon and Bydueron BCise) is a sustained-release formulation that consists of exenatide embedded within biodegradable polymeric microspheres of poly (DL-lactic-co-glycolic acid) (16). Dulaglutide (Trulicity) has two copies of a GLP-1 analogue covalently linked to an Fc fragment of human IgG4 (16,239). Semaglutide (Ozempic) is an analogue of human GLP‐1 RA and is linked via a hydrophilic spacer and a fatty acid side chain to albumin (239).

 

Administration

 

SHORT ACTING GLP-1 RECEPTOR AGONISTS

 

Initiate exenatide at 5 ug twice daily; increase to 10 ug twice daily after 1 month based on clinical response. Inject subcutaneously within 60 minutes prior to morning and evening meals (or before the two main meals of the day).

 

The starting dose of lixisenatide is 10 ug subcutaneously once daily within one hour before the first meal of the day for 14 days and then increase the dose to the maintenance dose of 20 ug once daily.

 

LONG ACTING GLP-1 RECPTOR AGONISTS

 

Initiate liraglutide with a dose of 0.6 mg per day for one week. After one week at 0.6 mg per day, the dose should be increased to 1.2 mg. If the 1.2 mg dose does not result in acceptable glycemic control, the dose can be increased to 1.8 mg. Inject subcutaneously once-daily at any time of day, independently of meals.

 

The recommended dose of long acting exenatide is 2 mg subcutaneously once every 7 days (weekly). The dose can be administered at any time of day, with or without meals.

 

The recommended initiating dose of dulaglutide is 0.75 mg subcutaneously with or without food once weekly. The dose may be increased to 1.5 mg once weekly to achieve glycemic control. If after 4 weeks glycemic control is not achieved the dose can be increased to 3.0mg once weekly and then after another 4 weeks to 4.5mg once weekly for additional glycemic control.

 

The recommended initiating dose of semaglutide is 0.25 mg subcutaneous injection with or without food once weekly for 4 weeks. The 0.25 mg dose is intended for treatment initiation and is not effective for glycemic control. After 4 weeks on the 0.25 mg dose, increase the dosage to 0.5 mg once weekly. If additional glycemic control is needed after at least 4 weeks on the 0.5 mg dose, the dosage may be increased to 2 mg once weekly.

 

Note that exenatide and lixisenatide are contraindicated in patients with renal dysfunction (for details see Contraindications section).

 

Information on the pen delivery systems for the GLP-1 receptor agonists is shown in table 22.

 

Table 22. Characteristics of GLP-1 Receptor Agonist Pen Devices

Generic

Exenatide

Exenatide

Exenatide

Lixisenatide

Liraglutide

Dulaglutide

Semaglutide

Brand

Byetta

Bydureon

Bydureon

BCise

Lyxumia

Victoza

Trulicity

Ozempic

Single or multiple use

Multiple

Single

Single

Multiple

Multiple

Single

Multiple

Dose*

5 or 10ug

2mg

2mg

10 or 20ug

0.6, 1.2, or 1.8mg

0.75 or 1.5mg

0.25, 0.5, or 1.0mg

Preparation

None

Resuspend

Mix

None

None

None

None

*Only the liraglutide pen can deliver different doses

 

Mechanism of Action

 

GLP-1 receptor agonists potentiate glucose dependent insulin secretion increasing insulin levels and lowering glucose levels (16). In addition, GLP-1 receptor agonists potentiate the glucose dependent inhibition of glucagon secretion, which will also lower glucose levels (16). Finally, because of the supraphysiological levels of GLP-1 activity, short-acting GLP-1 receptor agonists will delay gastric emptying resulting in a decrease in postprandial glucose levels and induce satiety, which will decrease food intake (16).

 

Glycemic Efficacy

 

GLP-1 receptor agonists typically lower A1c by 1-2% (16). The efficacy of GLP-1 receptor agonists vary with semaglutide being the most potent and lixisenatide being the least potent (see table 6) (12).  Note table 6 does not include the 3.0mg and 4.5mg of dulaglutide, which lower A1c by 1.6% and 1.8% respectively (240). In general, long acting GLP-1 receptor agonists are better at lowering A1c levels compared to short acting agents (12,239). The efficacy in lowering A1c is similar in monotherapy and during combination therapy (16). The reduction in A1c is sustained over several years (164). Long acting GLP-1 receptor agonists lower fasting glucose levels more effectively than short acting drugs (239). Conversely, short acting GLP-1 receptor agonists lower postprandial glucose excursions to a greater extent than long acting agents (239). Short acting GLP-1 receptor agonists induce a substantial retardation in gastric emptying, which likely contributes significantly to the lowering of postprandial glucose excursions after meals when they are administered (239). Notably, the ability of short acting GLP-1 receptor agonists to prevent postprandial glucose excursions is greatly diminished for meals when they are not administered (239). In patients with diminished beta cell function the glycemic response to GLP-1 receptor agonist therapy is reduced (241).

 

Studies have compared adding a GLP-1 receptor agonist to basal insulin vs. adding rapid acting insulin to basal insulin (242). In a meta-analysis there were no differences in lowering A1c levels but treatment with basal insulin plus GLP-1 receptor agonist led to a significant reduction in body weight, whereas basal insulin plus rapid acting insulin treatment was associated with weight gain (difference -2.95 kg; p = 0.0001) (242). Additionally, patients treated with basal insulin plus GLP-1 receptor agonist were less likely to experience symptomatic hypoglycemia (OR: 0.52; p < 0.0001) and severe hypoglycemia (OR: 0.27; p = 0.07) than those treated with basal insulin plus rapid acting insulin. Thus, adding a GLP-1 receptor agonist to basal insulin instead of bolus insulin will result in similar improvements in glycemic control with fewer side effects.

 

Studies have also compared adding insulin therapy vs. adding a GLP-1 receptor agonist. In a meta-analysis of 19 studies GLP-1 receptor agonists reduced A1c levels slightly more than insulin therapy (difference -0.12%, P < .0001) (243). As expected, hypoglycemia was less frequent in the patients treated with the GLP-1 receptor agonists.

 

Because the effect of GLP‐1 receptor agonists on insulin and glucagon secretion are glucose dependent they have a low potential to cause hypoglycemia (16,239).  The risk of hypoglycemia increases when these GLP-1 receptor agonists are used in combination with insulin or secretagogues (239).

 

Both GLP-1 receptor agonists and SGLT-2 inhibitors have been shown to decrease cardiovascular disease (GLP-1 receptor agonists decrease primarily atherosclerotic complications while SGLT-2 inhibitors primarily decrease heart failure). Therefore, the use of these drugs in combination to prevent cardiovascular disease has been proposed. In an analysis of four randomized trials adding a GLP-1 receptor agonist to a SGLT-2 inhibitor it was reported that the addition of a GLP-1 receptor agonist resulted in a greater reduction in HbA1c (-0.74%), body weight (-1.61 kg), and systolic blood pressure (-3.32 mmHg) demonstrating the benefits of using these drugs in combination (244).  

 

Other Effects

 

WEIGHT LOSS

 

GLP-1 receptor agonists induce weight loss (16,239).  A comparison of the ability of the maximum dose of different GLP-1 receptor analogues to induce weight loss are shown in table 23. It should be recognized that the weight loss shown in Table 23 represents averages. In clinical practice some patients lose a large amount of weight with GLP-1 receptor agonists while other patients can actually gain weight. The author has personally seen patients’ loss more than 50 lbs. Higher doses of liraglutide and semaglutide are approved for the treatment of obesity, which is discussed in the Endotext chapter “Pharmacologic Treatment of Overweight and Obesity in Adults” (245) (semagulutide 2.0mg is also approved for the treatment of diabetes). Studies have compared the effect of high doses of GLP-1 receptor agonists used for weight loss and lower doses used for treating diabetes (table 24). In general, higher doses of GLP-1 receptor agonists result in a modest further lowering of A1c and a more robust decrease in body weight.

 

Table 23. Effect of GLP-Receptor Agonists on Mean Weight Loss (12)

GLP-1 Receptor Agonist

Mean Weight Loss

Dulaglutide 1.5mg weekly

1.1Kg

Exenatide 10ug bid

1.2Kg

Exenatide 2mg weekly

1.1Kg

Liraglutide 1.8mg qd

1.5Kg

Lixisenatide 20ug qd

0.7Kg

Semaglutide 1mg weekly

3.8Kg

Based on a baseline weight of 90 kg after 26 weeks of treatment

 

Table 24. Comparison of Low and High Dose GLP-1 Receptor Agonists on A1c and Body Weight

 

Change in A1c (%)

Change in Body Weight (% or kg)

SCALE Diabetes (246)

Placebo

-0.3%

-2.0%

Liraglutide 1.8mg qd

-1.1%

-4.7%

Liraglutide 3.0mg qd

-1.3%

-6.0%

STEP-2 (247)

Placebo

-0.4%

-3.4%

Semaglutide 1mg weekly

-1.5%

-7.0%

Semaglutide 2.4mg weekly

-1.6%

-9.6%

SUSTAIN FORTE (248)

Semaglutide 1mg weekly

-1.9%

-6.2%

Semaglutide 2.0mg weekly

-2.2%

-7.2%

AWARD-11 (240)

 

 

Dulaglutide 1.5mg weekly

-1.5%

-3.1kg

Dulaglutide 3.0mg weekly

-1.7%

-4.0kg

Dulaglutide 4.5mg weekly

-1.9%

-4.7kg

 

The exact mechanisms responsible for the decrease in weight are not yet fully understood but both central and peripheral mechanisms are thought to play a part in activating receptors in the central nervous system associated with weight loss (239). GLP‐1 receptor agonists are thought to reduce body weight through decreased gastrointestinal motility and the promotion of satiety via the activation of GLP‐1 receptors in various regions of the brain (239).

 

BLOOD PRESSURE

 

GLP-1 receptor agonists result in modest but significant reductions in systolic blood pressure (2-5 mmHg) (16).

 

HEART RATE

 

The effects of GLP-1 receptor agonists on heart rate differ between drugs. Short-acting GLP-1 receptor agonists result in a modest increase (1-3 beats per minute) while long-acting GLP-1 receptor agonists are associated with a more pronounced and sustained increase (3-10 beats per minute) during the day and night (249).

 

LIPIDS

 

GLP-1 receptor agonists can favorably affect the lipid profile by inducing weight loss (decreasing triglycerides and very modestly decreasing LDL-C levels) (77). In a review by Nauck and colleagues it was noted that GLP-1 receptor agonists lowered TG levels by 18 to 62mg/dl depending upon the specific GLP-1 receptor agonist while decreasing LDL-C by 3-8mg/dl and increasing HDL-C by less than 1mg/dl (224). Additionally, GLP-1 receptor agonists reduce postprandial triglycerides by reducing circulating chylomicrons by decreasing intestinal lipoprotein production (77,224).

 

CARDIOVASCULAR DISEASE

 

The effect of six GLP-1 receptor agonists on cardiovascular disease has been reported.

 

ELIXA

 

In the Elixa trial 6,068 patients with T2DM and who recently had a myocardial infarction or been hospitalized for unstable angina were randomized to placebo or lixisenatide, and followed for a median of 25 months (250). The primary end point of cardiovascular death, myocardial infarction, stroke, or hospitalization for unstable angina was similar in the placebo or lixisenatide groups.

 

LEADER Trial

 

In contrast, the LEADER trial has shown that liraglutide decreased cardiovascular events (251). In this trial 9,340 patients with T2DM at high cardiovascular risk (~ 81% with established cardiovascular disease) were randomly assigned to receive liraglutide or placebo. After a median time of 3.5 years, the primary outcome of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke occurred in significantly fewer patients in the liraglutide group (13.0%) than in the placebo group (14.9%) (hazard ratio, 0.87, P=0.01). Additionally, deaths from cardiovascular causes (hazard ratio 0.78, P=0.007) or any cause was lower in the liraglutide group than in the placebo group (hazard ratio, 0.85; P=0.02). Interestingly patients with established cardiovascular disease or decreased renal function (eGFR < 60) appeared to derive the greatest benefit of liraglutide treatment (252,253). The decrease in cardiovascular events were similar in patients with and without a history of heart failure (254). Finally, a significant reduction in amputations with liraglutide vs. placebo was observed (HR 0.65; P = 0.03]) (255).

 

SUSTAIN 6 Trial

 

In support of the beneficial effects of some GLP1 receptor agonists to reduce cardiovascular events, semaglutide has also been shown to reduce cardiovascular events (256). In this trial, 3,297 patients with T2DM with established cardiovascular disease (83%), chronic heart failure, chronic kidney disease, or age >60 with at least one cardiovascular risk factor were randomized to receive once-weekly semaglutide (0.5 mg or 1.0 mg) or placebo for 104 weeks. The primary outcome of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke occurred in 6.6% of the semaglutide group and 8.9% of the placebo group (hazard ratio, 0.74; P = 0.02).

 

EXSCEL Trial

 

The effect of once weekly exenatide vs. placebo on cardiovascular outcomes was tested in 14,752 patients with T2DM, 73% who had cardiovascular disease (257). The primary outcome was the occurrence of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke. After a median follow-up of 3.2 years (duration of drug exposure 2.4 years) the primary outcome was reduced in the exenatide treated group but this difference just missed achieving statistical significance (hazard ratio 0.91; 95% CI 0.83-1.00; p=0.06). While not statistically significant these results are consistent with the results observed with other GLP-1 receptor agonists. It should be recognized that a high percentage of patients discontinued exenatide therapy in this trial (>40%) and this could have adversely affected the ability of exenatide treatment to favorably effect cardiovascular outcomes.

 

HARMONY Outcomes Trial

 

The effect of once weekly albiglutide vs. placebo was tested in 9,463 patients with T2DM and cardiovascular disease (258). The primary outcome was first occurrence of cardiovascular death, myocardial infarction, or stroke. After a median follow-up of 1.6 years a 22% decrease in the primary endpoint was observed in the albiglutide group (hazard ratio 0.78, p<0·0001). It should be noted that albiglutide is no longer available as it was removed from the market due to commercial considerations by the manufacturer.

 

REWIND Trial

 

This was a randomized study of weekly dulaglutide (1.5 mg) or placebo in 9,901 patients with T2DM who had either a previous cardiovascular event or cardiovascular risk factors (approximately 70% of patients did not have prior cardiovascular disease) (259).  During a median follow-up of 5.4 years the primary outcome of non-fatal myocardial infarction, non-fatal stroke, or death from cardiovascular causes was decreased by 12% in the dulaglutide treated group (HR 0.88, p=0.026). The decrease in events was similar in participants with and without previous cardiovascular disease. In an analysis that focused on stroke it was noted that dulaglutide reduced ischemic stroke by 25% compared to placebo but had no effect on hemorrhagic stroke (260).

 

Summary

 

Thus, four studies have clearly demonstrated that treatment with GLP-1 receptor agonists reduces cardiovascular events, one study has provided data consistent with these results, and one study failed to demonstrate benefit. Why there are differences in results between these studies is unknown but could be due to differential effects of the GLP-1 receptor agonists, differences in the patient populations studied, or other unrecognized variables. A meta-analysis of 7 cardiovascular outcome studies using GLP-1 receptor agonists (ELIXA (lixisenatide), LEADER (liraglutide), SUSTAIN-6 (semaglutide), EXSCEL (exenatide), Harmony Outcomes (albiglutide), REWIND (dulaglutide), and PIONEER 6 (oral semaglutide) reported a 12% decrease in cardiovascular death, stroke, or myocardial infarction (p<0.0001), 12% decrease in cardiovascular deaths (p<0.003), 16% decrease in fatal or non-fatal strokes (p<0·0001), and 9% decrease in fatal or non-fatal myocardial infarctions (p=0.043) (261) (Table 25).

 

Table 25. Summary of GLP-1 Receptor Agonist Cardiovascular Outcome Trials

 

Number

Prior CVD

HbA1c

Mean Follow-up (years)

Hazard Ratio* (95% CI)

P value

ELIXA

Lixisenatide

6068

100%

7.7%

2.1

1.02

(0.89-1.17)

0.78

LEADER

Liraglutide

9340

81%

8.7%

3.8

0.87

(0.78-0.97)

0.015

SUSTAIN 6

Semaglutide

3297

83%

8.7%

2.1

0.74

(0.58-0.95)

0.016

EXSCEL

Exenatide

14,752

73%

8.0%

3.2

0.91

(0.83-1.00)

0.061

HARMONY

Albiglutide

9463

100%

8.7%

1.6

0.78

(0.68-0.90)

<0.001

REWIND

Dulaglutide

9901

31%

7.3%

5.4

0>88

(0.79-0.99)

0.026

PIONEER 6**

Semaglutide oral

3183

85%

8.2%

1.3

0.79

(0.57-1.11)

0.17

Overall (261)

 

 

 

 

0.88

(0.82-0.94)

<0.001

*CVD death, MI, Stroke. ** The Pioneer study is included in this table to provide information on all the studies examining the effect of GLP-1 receptor agonists on cardiovascular disease.

 

The mechanism accounting for this decrease in cardiovascular disease is uncertain but could be related to reductions in glycated hemoglobin, body weight, systolic blood pressure, postprandial triglyceride levels, or the direct effect of activation of GLP-1 receptors on the atherosclerotic process such as improving endothelial function (262).

 

The effect of a GLP-1 receptor agonist (efpeglenatide- not available) in patients on an SGLT-2 inhibitor was determined in the AMPLITUDE-O trial (263). The effect of efpeglenatide vs. placebo on cardiovascular and renal outcomes was similar in the absence and presence of baseline SGLT-2 inhibitors. Additional studies on the effect of the combination of GLP-1 agonists and SGLT-2 inhibitors on key outcomes are needed.  

 

HEART FAILURE

 

Two small randomized studies have specifically examined the effect of GLP-1 receptor agonists on clinical outcomes in patients with heart failure. Margulies and colleagues randomized patients recently hospitalized for heart failure with a decreased ejection fraction to liraglutide (n=154) or placebo (n = 146) (59% with T2DM) (264). Treatment with liraglutide did not lead to greater posthospitalization clinical stability or decrease the number of deaths or rehospitalizations for heart failure. Jorsal et al carried out a randomized trial of liraglutide vs. placebo in patients (n=241) with reduced left ventricular ejection fraction who were clinically stable and on optimal heart failure treatment (265). Unexpectedly, serious cardiac events were seen in 10% of patients treated with liraglutide compared with 3% of patients in the placebo group (P = 0.04).

 

Several of the large cardiovascular outcome trials have analyzed the effect of administration of GLP-1 receptor agonists in the subgroup of patients with a history of heart failure. In the EXSCEL trial patients with heart failure at baseline had no decrease in all-cause mortality

whereas mortality was reduced in the subgroup without HF (HR 0.79; CI 0.68–0.92) (266). Similarly, in the combined data from the SUSTAIN-6 and PIONEER-6, patients with prior heart failure were the only subgroup that did not have a decrease in cardiovascular events (267). In contrast, in the LEADER trial the decrease in cardiovascular events were similar in patients with and without a history of heart failure (254).

 

The large cardiovascular outcome studies have determined the effect of GLP-1 receptor agonists on heart failure events. In a meta-analysis of the seven large cardiovascular outcome trials with a combined total of 56,004 participants, hospital admission for heart failure was decreased by 9% (0.91, 0.83-0.99; p=0.028) (261) (Table 26).

 

Table 26. Effect of GLP-1 Receptor Agonists on Heart Failure

Cardiovascular Outcome Trial

Heart Failure Hospitalization Heart Failure (HR (CI))

ELIXA (lixisenatide)

0.96 (0.75–1.23)

LEADER (liraglutide)

0.87 (0.73–1.05)

SUSTAIN-6 (semaglutide)

1.11 (0.77–1.61)

EXSCEL (exenatide)

0.94 (0.78–1.13)

HARMONY (albiglutide)

0.71 (0.53–0.94)

PIONEER-6 (oral semaglutide)

0.86 (0.48–1.55)

REWIND (dulaglutide)

0.93 (0.77–1.12)

Meta-analysis (261)

0.91 (0.83–0.99)

HR= hazard ratio; CI= 95% confidence interval

 

The effect of GLP-1 receptor agonists in preventing the development of heart failure and in patients with heart failure requires further study.

 

RENAL DISEASE

 

Five of the cardiovascular outcome studies described above also examined the effect of GLP-1 receptor agonists on kidney disease.

 

ELIXA Trial

 

Lixisenatide treatment decreased urinary albumin-to-creatinine ratio in patients with pre-existing micro or macroalbuminuria (268). Additionally, lixisenatide was associated with a reduced risk of new-onset macroalbuminuria compared with placebo (268). However, no significant differences in eGFR decline or the number of patients doubling their serum creatinine levels were seen between the lixisenatide treated group vs. placebo group (268).

 

LEADER Trial

 

The renal outcome in this trial was a composite of new-onset persistent macroalbuminuria, persistent doubling of the serum creatinine level, end-stage renal disease, or death due to renal disease. The renal outcome occurred in fewer patients in the liraglutide group than in the placebo group (hazard ratio, 0.78; P=0.003) (269). This favorable outcome was driven primarily by a decrease in the development of macroalbuminuria. The renal benefits did not appear to be driven by changes in A1c, body weight, or decreases in systolic BP.

 

SUSTAIN 6 Trial

 

In this trial, new or worsening nephropathy, defined as persistent macroalbuminuria, persistent doubling of the serum creatinine, or a creatinine clearance < 45ml/min/1.73m2, occurred in 3.8% of the patients in the semaglutide group and 6.1% of the patients in the placebo group (hazard ratio, 0.64; P=0.005) (256). As seen in the LEADER trial this favorable outcome was driven primarily by a decrease in the development of macroalbuminuria.

 

EXSCEL Trial

 

Exenatide treatment resulted in a reduction in new‐onset macroalbuminuria compared with placebo (2.2% vs 2.8%, P = 0.031), with no significant changes in either microalbuminuria (7.2% vs 7.5%) or ESKD requiring renal replacement therapy (0.7% vs 0.9%) (257).

 

REWIND Trial

 

The renal outcome included the occurrence of new macroalbuminuria (UACR >33·9 mg/mmol), a sustained decline in eGFR of 30% or more from baseline, or chronic renal replacement therapy (270). During a median follow-up of 5·4 years the renal outcome developed in 17.1% of patients in the dulaglutide group and in 19.6% of patients in the placebo group (HR 0.85, p=0·0004). This beneficial effect was driven by a reduction in the development of macroalbuminuria (HR 0.77; p<0.0001)

 

Summary

 

These studies demonstrate that GLP-1 receptor agonist administration reduce albuminuria without effecting eGFR. The decrease in albuminuria without effecting eGFR is similar to what was observed in some of the DPP-4 inhibitor studies described above. The mechanism accounting for this decrease is uncertain but decreased systolic BP, weight loss, improved glycemic control, or direct effects on the kidneys could have contributed to this decrease in albuminuria.

 

While the large studies described above demonstrated that GLP-1 receptor agonists primarily decrease albuminuria the AWARD 7 trial provides data on eGFR. The Award 7 was a multicenter randomized trial of dulaglutide 0.75mg weekly (n= 190), 1.5mg weekly (n= 193), or daily insulin glargine (n= 194) in patients with T2DM and Stage 3 and 4 chronic kidney disease (271). At 52 weeks, eGFR was higher with dulaglutide 1.5 mg (34.0; p=0.005 vs insulin glargine) and dulaglutide 0.75 mg (33.8; p=0·009 vs insulin glargine) than with insulin glargine (31.3mL/min per 1·73 m2). In contrast to the cardiovascular studies described above at 52 weeks dulaglutide 1.5 mg and 0.75 mg did not affect albuminuria. Additionally, a pooled analysis of the LEADER (liraglutide) and SUSTAIN 6 trials found a preservation in eGFR with GLP-1 receptor agonists, particularly in patients with a reduced baseline eGFR (272). Further studies are in progress to better define the effect of GLP1 receptor agonists on diabetic kidney disease.

 

NONALCOHOLIC FATTY LIVER DISEASE (NAFLD) AND NONALCOHOLIC STEATOHEPATITIS (NASH)

 

Studies have suggested that GLP-1 receptor agonists have beneficial effects on NAFLD and NASH (96). A meta-analysis of liraglutide studies and a separate meta-analysis of lixisenatide studies have reported that these drugs decrease liver enzymes (273,274). A 12-week randomized trial in 60 patients with NAFLD of exenatide + basal insulin vs. rapid acting insulin + basal insulin demonstrated lower liver enzymes in the exenatide treated group (275). Moreover, the reversal rate of fatty liver was greater in the group treated with exenatide (93.3%) than the intensive insulin group (66.7%) (p < 0.01). Similarly, liraglutide and dulaglutide has also been shown to decrease intrahepatic fat (276-278).  

 

In the LEAN Trial 52 patients with NASH were randomized to liraglutide 1.8 mg daily or placebo and followed for 48 weeks (279). Resolution of NASH occurred in 39% of patients treated with liraglutide and only 9% patients in the placebo group (RR 4.3; p=0.019). Progression of fibrosis occurred in 9% of patients in the liraglutide group versus 36% patients in the placebo group (p=0.04).

 

A recent trial of semaglutide subcutaneously given daily (0.1, 0.2, and 0.4 mg) demonstrated an improvement in NASH without a beneficial effect on fibrosis (280). Whether weekly semaglutide or daily oral semaglutide would have similar effects is unknown. 

 

While these data are suggestive larger and longer studies on the effect of GLP-1 receptor agonists on NAFLD and NASH are required.

 

Side Effects

 

GASTROINTESTINAL

 

The most common adverse effects are GI and include nausea, vomiting, and diarrhea (239). These symptoms are usually transient, resolving overtime (16). The GI side effects can be reduced by slowly increasing the dose (16). GI side effects tend to be more pronounced with short acting GLP-1 receptor agonists (239). Dehydration can occur secondary to GI side effects and can result in acute kidney failure (package insert).

 

GALL BLADDER DISEASE

 

Observational studies have shown an association of treatment with GLP-1 receptor agonists and bile duct and gallbladder disease (281). Additionally, a meta-analysis of randomized trials using GLP-1 inhibitors reported an association with an increased risk of cholelithiasis (282). Higher doses and a longer duration of treatment increased the risk of gallbladder disease (283). Finally, large cardiovascular trials with liraglutide (LEADER Trial), exenatide (EXSCEL Trial), and lixisenatide (ELIXA Trial) also reported an increased risk of gall bladder or biliary tract disease (250,257,284), however the large cardiovascular trial with semaglutide (SUSTAIN 6) did not observe an increase (256). It has been hypothesized that weight loss and/or decreased gallbladder motility induced by GLP-1 receptor agonists could contribute to this increase in gall bladder disease.

 

INJECTION-SITE REACTIONS

 

Injection-site reactions (rash, erythema) are also common with GLP-1 receptor agonists (16). Subcutaneous injection-site nodules may occur with the use of weekly exenatide (package insert), an abnormality that is due to the formulation.

 

MEDULLARY THYROID CANCER

 

Thyroid C-cell hyperplasia and medullary cell carcinoma has also been raised as possible concerns based on preclinical studies in rodents, but clinical studies in humans have not shown any indication of thyroid disorders (16). A meta-analysis of the four large cardiovascular outcome studies described above did not demonstrate an increased risk of medullary thyroid cancer with GLP-1 receptor agonist treatment (285)

 

PANCREATITIS

 

Subclinical increases in pancreatic enzyme levels are commonly observed with all GLP‐1 receptor agonists and pancreatitis has been reported (239). Importantly increases in lipase and amylase were not predictive of subsequent pancreatitis (286). A meta-analysis of four large cardiovascular outcome studies described above did not demonstrate an increased risk of pancreatitis or pancreatic cancer with GLP-1 receptor agonist treatment (285,287). A meta-analysis of all seven cardiovascular outcome studies also did not demonstrate an increase in pancreatitis with GLP-1 receptor agonist treatment (288).

 

RETINOPATHY

 

In the SUSTAIN 6 trial described above the rates of retinopathy complications (vitreous hemorrhage, blindness, or conditions requiring treatment with an intravitreal agent or photocoagulation) were significantly higher in the semaglutide group compared to the placebo group (hazard ratio, 1.76; P=0.02) (256). This increased risk of retinopathy complications has been attributed to the magnitude and rapidity of A1c reduction during the first 16 weeks of treatment in patients who had pre-existing retinopathy and poor glycemic control at baseline (“early worsening”) (289). A meta-analysis of GLP-1 cardiovascular trials found an association between retinopathy and the magnitude of A1c reduction supporting the hypothesis that the increase in retinopathy in SUSTAIN 6 was due to lowering of A1c (290).

 

Of note, other trials using semaglutide did not observe an increased risk of retinopathy (289). Additionally, an increase in diabetic retinopathy was not observed in the other cardiovascular outcome trials (250,251,257,258). In a meta-analysis of 60 studies with 60,077 patients treatment with GLP-1 receptor agonists did not increase the incidence of diabetic retinopathy, macular edema, retinal detachment, or retinal hemorrhage (291). However, the incidence of vitreous hemorrhage was higher in subjects treated with GLP-1 receptor agonists compared with placebo (odds ratios 1.93; 95% CI 1.09 to 3.42). Thus, it does not appear that GLP-1 receptor agonists treatment result in an increase in diabetic eye disease. A 5 years eye safety study for semaglutide, the FOCUS trial (NCT03811561), is currently underway

 

Contraindications and Drug Interactions

 

RENAL

 

Care needs to be exercised in patients with severe renal disease as they are more susceptible to the side effects of GLP-1 receptor agonists and more likely to have serious side effects (package inserts). There is limited data in patients with end stage renal disease.

 

Exenatide should not be used in patients with severe renal impairment (creatinine clearance < 30 mL/min) or end-stage renal disease (package insert). Caution should be applied when initiating or escalating doses of exenatide from 5 mcg to 10 mcg in patients with moderate renal impairment (creatinine clearance 30 to 50 mL/min) (package insert).

 

Weekly exenatide is not recommended for use in patients with eGFR below 45 mL/min/1.73m2 or end stage renal disease (package insert).

 

Lixisenatide is not recommended in patients with end stage renal disease (eGFR <15 mL/min/1.73 m2) (package insert).

 

No dose adjustments for liraglutide, semaglutide, or dulaglutide are recommended for patients with renal impairment (package insert).

 

OTHER

 

Exenatide is not recommended in patients with gastroparesis or severe gastrointestinal disease (package insert).

 

In patients with a history of pancreatitis or at high risk for pancreatitis many clinicians avoid GLP-1 receptor agonists.

 

GLP-1 receptor agonists are contraindicated in patients with a personal or family history of Medullary Thyroid Cancer and in patients with Multiple Endocrine Neoplasia syndrome type 2 (MEN 2) (package insert).

 

Summary

 

The ability of GLP-1 receptor agonists to effectively decrease A1c levels, reduce atherosclerotic cardiovascular disease, and in some patients induce a major loss of weight make these drugs very attractive in the treatment of patients with T2DM. Additionally, once weekly administration for certain drugs in this class can improve compliance. 

 

Table 27. Advantages and Disadvantages of GLP- 1 Receptor Agonists

Advantages

Disadvantages

Weight Loss

GI side effects

No Hypoglycemia

Requires Injection

Reduce CVD (liraglutide, semaglutide, dulaglutide)

Pancreatitis?

Improve NAFLD

Thyroid cancer?

Once a week therapy possible

Gall bladder disease

Decrease albuminuria

Expensive

Decrease postprandial glucose

 

 

ORAL GLUCAGON LIKE PROTEIN-1 (GLP-1) RECEPTOR AGONISTS

 

Introduction

 

In 2019 an oral form of semaglutide became available. To facilitate absorption of semaglutide, which is a 31 amino acid peptide, the tablet contains a permeation enhancer N-(8-[2-hydroxybenzoyl]amino)caprylic acid (SNAC, Eligen® Technology, Emisphere Technologies), which is a small fatty acid derivative that accelerates the absorption of semaglutide across the gastric epithelium avoiding the activation of proteolytic enzymes and pH-induced degradation in the stomach (292). This allows for the absorption of an intact peptide. One should note that the bioavailability of oral semaglutide is very low as the dose of oral semaglutide is 7-14 mg per day vs 0.5-2.0 mg once a week with the injectable dose.

 

Administration

 

The oral form of semaglutide must be taken at least 30 minutes before the first food, beverage, or other oral medications of the day with no more than 4 ounces of plain water (package insert). Waiting less than 30 minutes, or taking with food, beverages (other than plain water), or other oral medications will adversely affect the absorption of semaglutide. Waiting more than 30 minutes to eat may increase the absorption. The starting dose is 3 mg once daily for 30 days. After 30 days on the 3 mg dose, increase the dose to 7 mg once daily. The dose may be increased to 14 mg once daily if additional glycemic control is needed after at least 30 days on the 7 mg dose (package insert). Patients treated with once weekly semaglutide 0.5 mg injections can be transitioned to oral semaglutide 7 mg or 14 mg a day. No dose adjustment is recommended for patients with renal or hepatic impairment (package insert).

 

Mechanism of Action

 

The mechanism of action is identical to injected GLP-1 receptor agonists described above.

 

Glycemic Efficacy

 

In a meta-analysis of five trials of oral semaglutide vs. placebo, treatment with oral semaglutide reduced HbA1c by 0.89% (293). In the Pioneer 1 study 703 patients were randomized (mean baseline HbA1c 8.0%) to placebo vs. various doses of oral semaglutide (294).  After 26 weeks of treatment A1c decreased by -0.6% in the 3 mg group, -0.9% in the 7 mg group, and -1.1% in the 14 mg group compared to placebo (P < 0.001 for all results). If the decrease in A1c was adjusted for premature drug discontinuation or initiation of rescue medication the estimated decreases in A1c were -0.7% in the 3 mg group, -1.2% in the 7 mg group, and -1.4% in the 14 mg group (P < 0.001 for all).

 

Studies have also examined the ability of oral semaglutide to lower A1c vs. other drugs. Compared to sitagliptin, oral semaglutide 7mg per day reduced A1c by -0.3% while 14mg per day reduced A1c by 0.5% (P < .001 for both) (295). In a similar trial with flexible dose adjustment of semaglutide, treatment with semaglutide (60% on 14mg per day) resulted in a 1.4% decrease in A1c while 100mg sitagliptin decreased A1c by 0.7% (296). In a trial comparing empagliflozin vs. oral semaglutide, treatment with semaglutide resulted in a greater decrease in A1c compared to empagliflozin (-1.3% vs. -0.9%; P < 0.0001) (297). In a comparison of liraglutide 1.8mg per day vs. oral semaglutide 14mg per day the change from baseline in A1c was -1.2% (SE 0·1) with oral semaglutide and -1.1% with subcutaneous liraglutide (298). If the decrease in A1c was adjusted for premature drug discontinuation or initiation of rescue medication then oral semaglutide treatment resulted in a slightly greater decreases in A1c than subcutaneous liraglutide (estimated treatment difference -0·2%). Finally, early in the development of oral semaglutide various doses of oral semaglutide were compared to weekly injected semaglutide (299). Compared to placebo 10mg per day of oral semaglutide reduced A1c by –1.2%, 20mg by –1.4%, while 1mg per week of injected semaglutide decreased A1c by 1.9% (not significantly different than the 20mg oral dose). Thus, oral semaglutide is more effective in lowering A1c levels than DPP-4 inhibitors or SGLT2 inhibitors and similar to liraglutide and perhaps slightly less potent than injected semaglutide.

 

Other Effects

 

WEIGHT LOSS

 

In a meta-analysis of weight loss, treatment with oral semaglutide reduced body weight by 2.99 kg compared to placebo (293). In a 26-week study comparing sitagliptin vs. oral semaglutide the 7mg dose resulted in a 1.6kg decrease and the 14mg dose a 2.5kg decrease in weight compared to sitagliptin (295). In contrast, oral semaglutide 14mg and empagliflozin 25mg resulted in a similar decrease in body weight at 26-weeks (-3.8 vs. -3.7kg) and 52-weeks (-3.8 vs. -3.6kg) (297). Finally, in a 26-week trial oral semaglutide resulted in greater weight loss (-4.4 kg than liraglutide (-3·1 kg) (298). 

 

BLOOD PRESSURE AND PULSE RATE

 

In a meta-analysis of blood pressure, treatment with oral semaglutide reduced systolic blood pressure by 3.16 mmHg and increased pulse rate by 1.90 beats per minute compared with placebo (293).

 

CARDIOVASCULAR DISEASE

 

3,183 patients with T2DM at high cardiovascular risk (age of ≥50 years with established cardiovascular or chronic kidney disease, or age of ≥60 years with cardiovascular risk factors) were randomly assigned to receive oral semaglutide or placebo (300). After a median time of 15.9 months, major adverse cardiovascular events, the primary outcome, occurred in 3.8% of the subjects treated with oral semaglutide and 4.8% of the placebo group (HR 0.79; 95% CI 0.57 to 1.11). Deaths from cardiovascular causes were 0.9% in the oral semaglutide group and 1.9% in the placebo group (HR 0.49; 95% CI, 0.27 to 0.92) while death from any cause occurred in 1.4% in the oral semaglutide group and 2.8% in the placebo group (HR 0.51; 95% CI, 0.31 to 0.84). It should be noted that the primary outcome was not statistically decreased in this study, which may be due to the relatively small number of subjects studied and the short duration of the study that together resulted in a small number of events. Additionally, more patients in the placebo group received treatment with an SGLT2 inhibitor than in the oral semaglutide group and SGLT2 inhibitors are well recognized to reduce cardiovascular disease events (see section on SGLT2 inhibitors), which could also have diminished the ability to observe a decrease in events in the oral semaglutide group. Because the glucose lowering, weight loss, and many other effects of oral semaglutide are very similar to injected semaglutide many experts consider the effects on cardiovascular and renal disease to also be similar. 

 

Side Effects

 

The most common adverse effects are GI and include nausea, vomiting, and diarrhea (292). Transient mild or moderate nausea was the most common adverse event occurring in 5-21% of subjects treated with oral semaglutide (292).

 

Severe hypoglycemia is uncommon in patients treated with oral semaglutide (292). The risk of hypoglycemia is increased when oral semaglutide is used in combination with insulin secretagogues (e.g., sulfonylureas) or insulin. Patients may require a lower dose of the secretagogue or insulin to reduce the risk of hypoglycemia when used in combination with oral semaglutide.

 

The safety profile of oral semaglutide is similar to other GLP-1 receptor agonists (see side effect section for GLP1 receptor agonists).

 

Contraindications and Drug Interactions

 

Similar to other GLP1 receptor agonists oral semaglutide is contraindicated in patients with a personal or family history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2.

 

No notable drug interactions have been described (package insert).

 

Summary

 

The delivery of a GLP1 receptor agonist via the oral route is advantageous and make oral semaglutide an attractive choice in the treatment of patients with T2DM who do want to inject medications given its ability to decrease A1c, body weight, and blood pressure with few serious side effects. It is likely that the other beneficial effects of GLP1 receptor agonists (e.g., reducing cardiovascular disease and proteinuria) will also occur with the oral formulation.

 

DUEL GLP-1 RECEPTOR AND GIP RECEPTOR AGONIST

 

Introduction

 

Tirzepatide (Mounjaro) is a 39 amino acid peptide that was engineered from the native GIP sequence and has agonist activity at both the GIP and GLP-1 receptors (301,302). A C20 fatty diacid moiety is conjugated at the position 20 lysine residue, which facilitates binding to albumin thereby resulting a half-life after administration of approximately 5 days allowing for weekly administration (301,302).

 

Administration

 

Tirzepatide is administered weekly at any time of day, with or without meals. The starting dose is 2.5mg subcutaneously and after 4 weeks the dose is increased to 5 mg (302). Depending upon the response one may increase the dosage in 2.5 mg increments every 4 weeks to a maximum dose of 15 mg per week (302). No dosage adjustment is recommended for renal or hepatic disease (package insert).

 

Mechanism of Action

 

Both GLP-1 and GIP stimulate insulin secretion in a glucose dependent fashion (303). The higher the glucose the greater the effect with no effect when glucose levels are in the normal to low range (303). As one would expect tirzepatide stimulates both first- and second-phase insulin secretion (302,304). GLP-1 inhibits glucagon secretion when glucose levels are increased while GIP will stimulate glucagon secretion, particularly when glucose levels are in the normal to low range (303). Tirzepatide reduces fasting and postprandial glucagon concentrations (304). These effects on insulin and glucagon secretion lead to decreases in glucose levels with a low risk of hypoglycemia as the increase in insulin secretion and decrease in glucagon secretion are dependent on elevated glucose levels. In addition, tirzepatide improves insulin sensitivity (304,305). While this increase in insulin sensitivity may be due to weight loss studies suggest that there may be additional factors contributing to the improved insulin sensitivity (305). GIP may have peripheral effects that could enhance insulin sensitivity.

 

Pharmacologic levels of GLP-1 slow gastric emptying and induce satiety by activating receptors in the hypothalamus thereby leading to decreased food intake and weight loss (303). GIP also appears to have central effects leading to decreased food intake in rodents but the effect in humans is not well defined (303).

 

Glycemic Efficacy

 

A number of different studies (SURPASS trials) have examined the effect of 5mg, 10mg, and 15mg of tirzepatide on glycemic control under a variety of clinical situations (Table 28). SURPASS 1 compared tirzepatide vs. placebo in patients on no medications (306), SURPASS 2 compared tirzepatide vs. semaglutide at a dose of 1 mg in patients on metformin (307), SURPASS 3 compared tirzepatide vs. degludec insulin in patients on metformin alone or in combination with an SGLT2 inhibitor (308), SURPASS 4 compared tirzepatide vs glargine insulin in patients treated with any combination of metformin, sulfonylurea, or SGLT-2 inhibitor (309), and SURPASS 5 compared tirzepatide vs. placebo in patients treated with glargine insulin with or without metformin (310). The treatment duration was 40 weeks in SURPASS 1, 2, and 5 and 52 weeks in SURPASS 3 and 4. Baseline A1c levels were between 7.9% and 8.5% in the SURPASS studies.

 

Table 28. Decrease in HbA1c with Tirzepatide Treatment 

 

SURPASS 1

SURPASS 2

SURPASS 3

SURPASS 4

SURPASS 5

 

Tirzepatide vs. Placebo

Tirzepatide vs. Semaglutide

Tirzepatide vs. Degludec

Tirzepatide vs.

Glargine

Tirzepatide vs. Placebo

Baseline A1c

7.9%

8.3%

8.2%

8.5%

8.3%

Tirzepatide 5mg

-1.8

-2.0

-1.9

-2.1

-2.1

Tirzepatide 10mg

-1.7

-2.2

-2.0

-2.3

-2.4

Tirzepatide 15mg

-1.7

-2.3

-2.1

-2.4

-2.3

Comparator

-0.1

-1.9

-1.3

-1.4

-0.9

 

It should be noted that the reduction in A1c induced by tirzepatide is quite impressive and results in an A1c level in an “intensive” control range. For example, in the SURPASS 2 trial 80% of patients had an A1c < 6.5% and 46% < 5.7% on 15mg tirzepatide. Additionally, comparison with semaglutide (SURPASS 2) demonstrated a modestly greater lowering of A1c with tirzepatide. A greater difference in the ability to decrease A1c was seen in an earlier study comparing tirzepatide vs. dulaglutide (tirzepatide 5mg- 1.6%, 10mg- 2.0%,15 mg- 2.4%; duluglutide 1.5mg- 1.1%) (311). Note the comparisons with semaglutide and dulaglutide used in these studies were not the maximal dose. Comparisons with insulin therapy (SURPASS 3 and 4) show better glycemic control with tirzepatide, which is likely due to an increased risk of hypoglycemia with insulin therapy that limits treatment. In SURPASS 3, 48% of patients on insulin therapy had a blood glucose < 70mg/dL while on tirzepatide treatment 8-14% of patients had a blood glucose < 70mg/dL. Severe hypoglycemia is not frequently observed with tirzepatide in the absence of concomitant insulin or sulfonylurea therapy. Finally, it is worth noting that the additional A1c reduction with an increased dose of tirzepatide is very modest. This is important to recognize that in patients that have side effects with higher doses of tirzepatide treatment it is not necessary to achieve maximal doses of tirzepatide to robustly improve glycemic control.

 

Other Effects

 

WEIGHT LOSS

 

Significant weight loss has been observed with tirzepatide administration. Table 29 shows the weight loss observed in the SURPASS trials. In contrast to the modest effects of increased doses of tirzepatide on A1c levels increased doses of tirzepatide have a greater effect on weight loss. At the 15mg dose over a 10% loss in weight is observed. It should be noted that in SURPASS 2 tirzepatide is compared to semaglutide 1.0mg, which is not the dose that is recommended for weight loss (the recommended dose is 2.4mg) and therefore one cannot be certain that tirzepatide is more efficacious than higher doses of semaglutide. In a comparison of tirzepatide vs. dulaglutide, tirzepatide resulted in greater weight loss (tirzepatide 5mg- 4.8kg, 10mg- 8.7kg, 15mg-11.3kg; dulaglutide 1.5mg- 2.7kg) (311).  

 

Table 29. Decrease in Weight with Tirzepatide Treatment 

 

SURPASS 1

SURPASS 2

SURPASS 3

SURPASS 4

SURPASS 5

 

Tirzepatide vs. Placebo

Tirzepatide vs. Semaglutide

Tirzepatide vs. Degludec

Tirzepatide vs.

Glargine

Tirzepatide vs. Placebo

Tirzepatide 5mg

-6.3kg/ -7.9%

-7.6kg/ -8.5%

-7.0kg/ -8.1%

-6.4kg/ -8.1%

-5.4kg/ -6.6%

Tirzepatide 10mg

-7.0kg/ -9.3%

-9.3kg/ -11.0%

-9.6kg/ -11.4%

-8.9kg/ -10.7%

-7.5kg/ -8.9%

Tirzepatide 15mg

-7.8kg/ -11.0%

-11.2kg/ -13.1%

-11.3kg/ -13.9%

-10.6kg/ -13.0%

-8.8kg/ -11.6%

Comparator

-1.0kg/ -0.9%

-5.7kg/ -6.7%

+1.9kg/ +2.7%

+1.7kg/ +2.2%

+1.6kg/ +1.7%

 

BLOOD PRESSURE AND PULSE

 

In the SURPASS studies described above tirzepatide treatment decreased systolic BP by 2.8 to 12.6 mm Hg and diastolic BP by 0.8 to 4.5 mm Hg (301). Tirzepatide treatment increased heart rate by approximately 2 to 4 beats per minute.

 

LIPIDS

 

In the SURPASS studies described above plasma triglyceride levels were consistently decreased by 13-25% (table 30). In most studies with the exception of SURPASS 5, HDL cholesterol levels increased by 3-11%. Total cholesterol and LDL cholesterol levels modestly decreased in most studies. Not unexpectedly given the decrease in triglyceride levels small LDL particles were decreased (312). The decrease in triglycerides could be related to weight loss, which is well known to affect these parameters (313). Additionally, GIP and tirzepatide increase lipoprotein lipase activity, which could increase the clearance of triglyceride rich lipoproteins (303,312). Finally, tirzepatide lowered Apo-CIII levels, which could also play a role in the decrease in triglyceride levels (312).

 

Table 30. Effect of Tirzapetide 15mg on Lipid Levels

 

SURPASS 1

SURPASS 2

SURPASS 3

SURPASS 4

SURPASS 5

 

Tirzepatide vs. Placebo

Tirzepatide vs. Semaglutide

Tirzepatide vs. Degludec

Tirzepatide vs.

Glargine

Tirzepatide vs. Placebo

Total Cholesterol

-7.6%

-1.5%

-3.0%

-5.6%

-12.6%

Triglycerides

-25.7%

-13.3%

-13.0%

-16.1%

-19.4%

LDLc

-10.8%

+1.2%

-3.8%

-9.3%

-17.3%

HDLc

+11.3%

+2.7%

+9.2%

+7.9%

-0.8%

Results are percent change in tirzepatide group minus percent change in comparator group

 

CARDIOVASCULAR DISEASE

 

A meta-analysis of seven randomized controlled trials with 4,887 participants treated with tirzepatide and 2,328 control participants found that MACE 4 (cardiovascular death, myocardial infarction, stroke, and hospitalized unstable angina) was decreased but not statistically significant (HR 0.80; 95% CI, 0.57–1.11) (314). One should note that the number of events in this meta-analysis was small because the duration of these studies was relatively short (approximately 1 year) and the population of patients included in these studies were not at high risk for cardiovascular events (only 1/3 with pre-existing cardiovascular disease). A long-term trial dedicated to determining the effect of tirzepatide on cardiovascular disease is ongoing (SURPASS-CVOT trial NCT04255433).

 

LIVER DISEASE

 

Liver fat content was decreased to a greater degree with tirzepatide treatment compared to treatment with insulin degludec (315). Additionally, tirzepatide decreased alanine aminotransferase and aspartate aminotransferase levels (316). Further studies on the effect of tirzepatide on liver disease are in progress.

 

Side Effects

 

The side effects described in the section on GLP-1 receptor agonists also are of concern with tirzepatide.

 

Patients treated with tirzepatide in combination with a sulfonylurea or insulin may have an increased risk of hypoglycemia. The risk of hypoglycemia may be decreased by a reduction in sulfonylurea or insulin dose.

 

The incidence of pancreatitis was increased in patients treated with tirzepatide compared to comparator treatment ((0.23 patients per 100 years of exposure vs. 0.11 patients per 100 years of exposure) (package insert). Additionally, acute gallbladder disease (cholelithiasis, biliary colic, and cholecystectomy) was increased with tirzepatide treatment (0.6% of tirzepatide-treated patients and 0% of placebo-treated patients) (package insert).

 

As with other GLP-1 receptor agonists nausea, diarrhea, vomiting, dyspepsia, constipation, and decreased appetite are common side effects.

 

Contraindications and Drug Interactions

 

Tirzepatide is contraindicated in patients with a personal or family history of medullary thyroid carcinoma or in patients with MEN2. Tirzepatide has not been studied in patients with a prior history of pancreatitis and it is unknown if patients with a history of pancreatitis are at higher risk for developing pancreatitis.

 

Tirzepatide delays gastric emptying and thereby has the potential to impact the absorption of concomitantly administered oral medications. The delay is largest after the first dose and diminishes over time.

 

SUMMARY

 

The major advantage of tirzepatide compared to GLP-1 receptor agonists is the greater decrease in weight and A1c levels.   

 

INSULIN-GLP-1 RECEPTOR AGONIST COMBINATIONS

 

Introduction

 

There are currently two insulin-GLP-1 receptor agonist combinations available for use; glargine insulin/lixisenatide (iGlarLixi) (Soliqua) and degludec insulin/liraglutide (iDegLira) (Xultophy). Both combine a basal insulin with a once-a-day GLP-1 receptor agonist. iGlarLixi contains 100U glargine and 33 ug lixisenatide per ml. iDegLira contains 100U degludec insulin and 3.6 mg liraglutide per ml.

 

Administration

 

In patients naive to basal insulin or to a GLP-1 receptor agonist, currently on a GLP-1 receptor agonist, or currently on less than 30 units of basal insulin daily the recommended starting dosage of iGlarLixi 100/33 is 15 units (15 units insulin glargine/5 ug lixisenatide) given subcutaneously once daily. In patients currently on 30 to 60 units of basal insulin daily, with or without a GLP-1 receptor agonist the recommended starting dosage of iGalLixi 100/33 is 30 units (30 units insulin glargine/10 ug lixisenatide) given subcutaneously once daily. After starting with the recommended dose, titrate the dosage upwards or downwards by two to four units weekly based on the patient’s glycemic control until the desired fasting plasma glucose is achieved. Administer iGlarLixi 100/33 subcutaneously once a day within an hour prior to the first meal of the day. The maximum dose of iGlarLixi 100/33 is 60 units daily (60 units insulin glargine/20 ug lixisenatide).

 

In patients naive to basal insulin or GLP-1 receptor agonist therapy the recommended starting dose of iDegLira 100/3.6 is 10 units (10 units of insulin degludec and 0.36 mg of liraglutide) given subcutaneously once-daily. In patients currently on basal insulin or a GLP-1 receptor agonist the recommended starting dose of iDegLira 100/3.6 is 16 units (16 units of insulin degludec and 0.58 mg of liraglutide) given subcutaneously once-daily. After starting the recommended starting dose, titrate the dosage upwards or downwards by two units every three to four days based on the patient’s blood glucose monitoring results and glycemic control goal until the desired fasting plasma glucose is achieved. Administer iDegLira 100/3.6 by subcutaneous injection once-daily at the same time each day with or without food. The maximum dose of iDegLira 100/3.6 is 50 units daily (50 units of insulin degludec and 1.8 mg of liraglutide).

 

Mechanism of Action

 

Basal insulin regulates fasting blood glucose levels between meals and overnight while a GLP-1 receptor agonist lowers postprandial glucose levels (317). Together this drug combination results in 24-hour glycemic control.

 

Glycemic Efficacy 

 

A number of studies have compared the ability of the combination of insulin-GLP receptor agonists to lower A1c levels compared to either insulin alone or GLP-1 receptor agonist alone (317). Table 31 shows the results of two large studies. As shown in Table 31 combination therapy was better at lowering A1c levels compared to the individual components (317). Additionally, the risk of hypoglycemia was similar with combination therapy compared to basal insulin alone. In a study of patients poorly controlled on glargine insulin adding rapid acting insulin (basal/bolus therapy) vs. switching to iDegLira was found to result in a similar reduction in A1c levels but the risk of hypoglycemia was greater with basal/bolus insulin (318). Not unexpectedly basal/bolus insulin resulted in greater weight gain (difference 3.6 kg) (318). Indirect comparisons suggest that iDegLira reduces A1C slightly more (< 0.5%) than iGlarLixi but this could be due to different study design, different patient populations, or other differences between the trials (317). A meta-analysis of 8 studies concluded that iDegLira and iGlarLixi demonstrated no significant differences in absolute HbA1c changes, fasting plasma glucose levels, or body weight changes relative to baseline (319).

 

Table 31. Effect of Combination Therapy vs Individual Components on Key Outcomes

Study

Treatment

A1c Reduction

% Subjects with Hypoglycemia

Change in Body Weight (Kg)

Rosenstock et al (320)

iGlarLixi

1.6%

26

-0.3

 

Glar

1.3%

24

+1.1

 

Lixi

0.9%

6

-2.3

Gough et al (321)

iDegLira

1.9%

32

-0.5

 

Deg

1.4%

39

+1.6

 

Lira

1.3%

7

-3.0

 

Other Effects

 

As shown in Table 31 the typical weight gain seen with insulin therapy alone is blunted with combination therapy.

 

Side Effects

 

Studies have noted that the typical GI side effects seen with GLP-1 receptor agonist therapy is blunted with combination therapy (141). The likely explanation is that the titration of the GLP-1 receptor agonist is slower with combination therapy (141).

 

Contraindications

 

The maximum daily insulin dose of 60 units for iGlarLixi and 50 units for iDegLira, may not be sufficient in patients requiring higher daily basal insulin doses (e.g., patients with severe insulin resistance). The maximum dose is determined by the GLP-1 receptor agonist dose (the max dose of iDegLira delivers 1.8 mg of liraglutide while the max dose of iGlarLixa delivers 20 ug of lixisenatide). Conversely, there may be some patients who require only a low dose of basal insulin and thus because of the fixed ratio of basal insulin to GLP-1 receptor agonist the dose of the GLP-1 receptor agonist may be too low. These examples are a limitation of fixed ratio delivery systems. In these patients one can use basal insulin and a GLP-1 receptor agonist independently. It should be noted that for the majority of patients the fixed ratio will be acceptable.

 

Summary

 

The effects of combination therapy are predictable based on studies of basal insulin and GLP-1 receptor agonists but providing them in a single injection provides convenience and makes it easier for patients to comply. Additionally, these combination drugs are titrated based on fasting glucose values and therefore frequent home blood glucose monitoring is not required, which also makes compliance easier. In patients who do not have adequate control on basal insulin alone combination therapy can be a useful therapeutic option.

 

BILE ACID SEQUESTRANTS

 

Introduction

 

Colesevelam (Welchol) is a non-absorbed, polymeric, LDL cholesterol lowering and glucose lowering agent that is a high-capacity bile acid-binding molecule (322). This drug was developed primarily to lower LDL cholesterol levels and was later noted to have favorable effects on blood glucose levels and was approved for improving glycemic control in patients with T2DM (16,322). It should be noted that other bile acid sequestrants (cholestyramine) also have favorable effects on glycemic control (323).

 

Administration

 

The recommended dose of colesevelam is 6 tablets once daily or 3 tablets twice daily with meals (tabs 625 mg). Alternatively, one can take one 3.75-gram packet once daily mixed with water, fruit juice, or diet soft drinks and taken with meals or one flavored chewable bar (80 calories per bar) with meals. For patients with difficulty swallowing tablets the use of packets or chewable bars is recommended.

 

Mechanism of Action

 

The mechanism by which bile acid sequestrants improve glucose metabolism is not well understood and the literature on this topic is often contradictory (324,325). Colesevelam does not alter hepatic or peripheral insulin sensitivity or decrease glucose GI absorption (325,326). Neither acute nor chronic treatment affect post oral glucose tolerance test blood glucose levels (326). Additionally, colesevelam treatment did not alter endogenous glucose production, gluconeogenesis, or glycogenolysis (325,326). Thus, the mechanisms accounting for the decrease in glucose effect of bile acid sequestrants remain unclear.

 

A leading hypothesis is that bile acid sequestrants improve glucose metabolism by stimulating the incretin pathway. Colesevelam increases GLP-1 and GIP concentrations suggesting that an increase in incretins contributes to the improvement in glycemic control (326-328). There are two pathways by which colesevelam increases GLP-1 secretion; (1) TGR5-mediated GLP-1 secretion in L cells and (2) intestinal proglucagon expression.

 

TGR5 is a G protein–coupled receptor expressed in many organs and tissues including the intestine (326,328). Bile acids activate TGR5 on the surface of intestinal L cells promoting GLP-1 secretion (326,328,329). Bile acid sequestrants appear to augment TGR5 activation and GLP-1 release, which occurs primarily in the distal intestine and colon (326,329,330). 

 

FXR is a nuclear hormone receptor that complexes with RXR to alter the expression of a large number of genes (328). Bile acids are a ligand for FXR and activate FXR thereby regulating gene expression (328). FXR activation in intestinal L cells decreases proglucagon expression resulting in a decrease in GLP-1 production (331). Conversely, a decrease in bile acids due to binding to colesevelam increases GLP-1 gene expression and secretion in response to glucose improving glucose metabolism (331).

 

It is likely that there are both incretin dependent and independent mechanisms that account for the improvement in glycemic control with colesevelam treatment. The exact mechanisms by which bile acid sequestrants lower A1c levels remain to be elucidated.

 

Glycemic Efficacy

 

Colesevelam has modest effects on glycemic control, lowering A1c levels by approximately 0.5% when added to metformin, sulfonylureas, pioglitazone, or insulin (16,322,332). Colesevelam does not lead to an increase in weight (322). In combination with metformin hypoglycemia is not a problem but when used in combination with insulin or sulfonylureas hypoglycemia may occur (322).

 

Other Effects

 

LIPIDS

 

Colesevelam lowers LDL cholesterol levels by 15-20% and has only a modest effect on HDL cholesterol levels (322,333). The effect of bile acid sequestrants on triglyceride levels varies (333). In patients with normal triglyceride levels, bile acid sequestrants increase triglyceride levels by a small amount. However, as baseline triglyceride levels increase, the effect of bile acid sequestrants on plasma triglyceride levels becomes greater, and can result in substantial increases in triglyceride levels (333). In patients with triglycerides > 500mg/dl the use of bile acid sequestrants is contraindicated (333).

 

CARDIOVASCULAR DISEASE

 

There have been no randomized studies that have examined the effect of bile acid sequestrants on cardiovascular end points in subjects with diabetes. In non-diabetic-subjects bile acid sequestrants have reduced cardiovascular events (334,335). Since bile acid sequestrants have a similar beneficial impact on serum lipid levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population.

 

Side Effects

 

Colesevelam does not have major systemic side effects as it is not absorbed and remains in the intestinal tract (333). However, it does cause gastrointestinal (GI) side effects (333). Constipation is a common side effect and can be severe. In addition, patients will often complain of bloating, dyspepsia, abdominal discomfort, and aggravation of hemorrhoids. Because of GI distress, a small number of patients will discontinue therapy with colesevelam. One can reduce or ameliorate these GI side effects by increasing hydration, adding fiber to the diet (psyllium), and using stool softeners.

 

Contraindications and Drug Interactions

 

Colesevelam treatment is contraindicated in patients with a history of bowel obstruction and is cautioned in those with a history of gastrointestinal motility disorders (i.e., gastroparesis) or gastrointestinal surgery (322,333). Colesevelam is contraindicated in patients with plasma triglyceride levels > 500 mg/dL or a history of hypertriglyceridemia-induced pancreatitis (package insert).

 

In the intestine bile acid sequestrants can impede the absorption of many other drugs (333). Colesevelam, which requires a much lower quantity of drug because of its high affinity and binding capacity for bile salts, has less of an effect on the absorption of other drugs than other bile acid sequestrants but can still adversely affect the absorption of certain drugs (Table 32) (333). Administration of these drugs, as well as vitamin supplements, 4 hours prior to administration of colesevelam minimizes pharmacokinetic interactions (333). This is particularly important with drugs that have a narrow toxic/therapeutic window, such as thyroid hormone, digoxin, or warfarin. It can be difficult for some patients, particularly those on multiple medications, to take colesevelam given the need to separate pill ingestion.

 

Table 32. Drugs Affected by Colesevelam

L-thyroxine

Cyclosporine

Glimepiride

Glipizide

Glyburide

Phenytoin

Olmesartan

Warfarin

Oral contraceptives

Repaglinide

Fenofibrate

Vitamin Supplements

 

Colesevelam may also decrease the absorption of fat-soluble vitamins A, D, E, and K (package insert).

 

Summary

 

Colesevelam has the advantage of lowering both A1c and LDL cholesterol levels. However, the efficacy of lowering A1c and LDL cholesterol levels is modest compared to other drugs. Additionally, in our patients with diabetes who are often on multiple medications it can be difficult to coordinate taking colesevelam with these other medications.

 

Table 33. Advantages and Disadvantages of Colesevelam

Advantages

Disadvantages

Lowers LDL cholesterol

Increases triglyceride levels particularly if already high

Minimal systemic effects

GI side effects

Once a day administration possible

Inhibits the absorption of other drugs

No hypoglycemia

Modest effect on A1c

Weight neutral

Relatively Expensive

 

PRAMLINTIDE (SYMLIN)

 

Introduction

 

Pramlintide is a soluble synthetic analog of human amylin (336). Amylin is co-sequestered and co-secreted with insulin by the pancreatic beta cells in response to nutrient stimuli (336). Amylin secretion in response to nutrients is absent in type 1 diabetes and in patients with T2DM there is impaired beta-cell secretion of amylin in response to nutrients (336). Amylin suppresses post-prandial arginine-stimulated glucagon secretion, suppresses appetite, and slows gastric emptying time through effects on the brain (336).

 

Administration

 

In patients with T2DM initiate pramlintide at 60 ug subcutaneously immediately prior to each major meal. Increase the dose from 60 to 120 ug prior to each major meal when no clinically significant nausea has occurred for at least 3 days. Note the dose used to treat patients with Type 1 diabetes differs from the dose used in patients with T2DM.

 

Mechanism of Action

 

Pramlintide attenuates post-prandial glucagon secretion, enhances satiety, and reduces food intake, which together improve glycemic control (336). These effects are mediated centrally (336)

 

Glycemic Efficacy

 

In a review of three randomized trials in patients with T2DM comparing pramlintide vs. placebo the A1c level was decreased by approximately 0.3-0.6% in the pramlintide group (337). Postprandial glucose excursions are significantly blunted with the addition of pramlintide (336). Pramlintide has only minimal effects on fasting glucose levels (337).

 

In a study comparing rapid acting insulin vs. pramlintide with meals a similar reduction in A1c was observed (338). In contrast to rapid acting insulin, patients treated with pramlintide did not gain weight (338). Additionally, the frequency of hypoglycemia was less with pramlintide compared with rapid acting insulin (338).  

 

Other Effects

 

Pramlintide treatment decreases weight (approximately 1-3 kg), which is likely due to decreased food intake (336,337). In a comparison of food intake during an ad libitum buffet meal, treatment with pramlintide resulted in an approximately 200 calorie decrease in food intake compared to placebo administration (339). Pramlintide also decreases gastric emptying (336).

 

Side Effects

 

A major side effect of pramlintide is nausea which can lead to patients discontinuing this drug (337).

 

Although pramlintide alone does not cause hypoglycemia, in combination with rapid acting meal time insulin the two drugs synergistically increase the risk of severe hypoglycemia (336). Therefore, rapid acting meal time insulin needs to be reduced upon initiation of pramlintide treatment to decrease the risk of hypoglycemia (336). Reducing rapid acting meal time insulin by 30-50% is recommended during the initial dose titration period (336).

 

Contraindications and Drug Interactions

 

Pramlintide is contraindicated in patients with hypoglycemia unawareness and confirmed gastroparesis (package insert).

 

Summary

 

Pramlintide is currently seldom used. Its modest effect on A1c levels coupled with the difficulties of administration (extra injections) and side effects has led to minimal use of this agent. Additionally, its major advantage of weight loss is now superseded by the use of GLP-1 receptor agonists.

 

Table 34. Advantages and Disadvantages of Pramlintide

Advantages

Disadvantages

Weight loss

Hypoglycemia

Decrease postprandial glucose

Frequent dosing

 

GI side effects

 

Expensive

 

Modest reduction in A1c

 

SUMMARY

 

A large number of drugs are now available for lowering glucose levels. For information on the management of T2DM and selecting amongst the available pharmacological agents see the chapter by Schroeder (5).  For information on the use of these drugs to treat diabetes during pregnancy, in children and adolescents, and for the prevention of diabetes see other Endotext chapters (2-4). 

 

REFERENCES

 

 

  1. American Diabetes Association. 5. Lifestyle Management: Standards of Medical Care in Diabetes-2019. Diabetes Care 2019; 42:S46-S60
  2. Buschur E, Stetson B, Barbour LA. Diabetes In Pregnancy. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2018.
  3. Perreault L. Prediabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  4. Yau M, Sperling M. Treatment of Diabetes mellitus in Children and Adolescents. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  5. Schroeder EB. Management of Type 2 Diabetes: Selecting Amongst Available Pharmacological Agents. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  6. Davies MJ, D'Alessio DA, Fradkin J, Kernan WN, Mathieu C, Mingrone G, Rossing P, Tsapas A, Wexler DJ, Buse JB. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care2018; 41:2669-2701
  7. American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2020. Diabetes Care 2020; 43:S98-S110
  8. Buse JB, Wexler DJ, Tsapas A, Rossing P, Mingrone G, Mathieu C, D'Alessio DA, Davies MJ. 2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2020; 43:487-493
  9. Cersosimo E, Triplitt C, Solis-Herrera C, Mandarino LJ, DeFronzo RA. Pathogenesis of Type 2 Diabetes Mellitus. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  10. Bloomgarden ZT, Dodis R, Viscoli CM, Holmboe ES, Inzucchi SE. Lower baseline glycemia reduces apparent oral agent glucose-lowering efficacy: a meta-regression analysis. Diabetes Care 2006; 29:2137-2139
  11. DeFronzo RA, Stonehouse AH, Han J, Wintle ME. Relationship of baseline HbA1c and efficacy of current glucose-lowering therapies: a meta-analysis of randomized clinical trials. Diabet Med 2010; 27:309-317
  12. Maloney A, Rosenstock J, Fonseca V. A Model-Based Meta-Analysis of 24 Antihyperglycemic Drugs for Type 2 Diabetes: Comparison of Treatment Effects at Therapeutic Doses. Clin Pharmacol Ther 2019; 105:1213-1223
  13. Nathan DM, Buse JB, Kahn SE, Krause-Steinrauf H, Larkin ME, Staten M, Wexler D, Lachin JM. Rationale and design of the glycemia reduction approaches in diabetes: a comparative effectiveness study (GRADE). Diabetes Care 2013; 36:2254-2261
  14. Thule PM, Umpierrez G. Sulfonylureas: a new look at old therapy. Curr Diab Rep 2014; 14:473
  15. Khunti K, Chatterjee S, Gerstein HC, Zoungas S, Davies MJ. Do sulphonylureas still have a place in clinical practice? Lancet Diabetes Endocrinol 2018; 6:821-832
  16. Tahrani AA, Barnett AH, Bailey CJ. Pharmacology and therapeutic implications of current drugs for type 2 diabetes mellitus. Nat Rev Endocrinol 2016; 12:566-592
  17. Prentki M, Matschinsky FM, Madiraju SR. Metabolic signaling in fuel-induced insulin secretion. Cell Metab 2013; 18:162-185
  18. Thorens B. GLUT2, glucose sensing and glucose homeostasis. Diabetologia 2015; 58:221-232
  19. MacDonald PE, Wheeler MB. Voltage-dependent K(+) channels in pancreatic beta cells: role, regulation and potential as therapeutic targets. Diabetologia 2003; 46:1046-1062
  20. Ashcroft FM, Gribble FM. ATP-sensitive K+ channels and insulin secretion: their role in health and disease. Diabetologia 1999; 42:903-919
  21. Zhang CL, Katoh M, Shibasaki T, Minami K, Sunaga Y, Takahashi H, Yokoi N, Iwasaki M, Miki T, Seino S. The cAMP sensor Epac2 is a direct target of antidiabetic sulfonylurea drugs. Science 2009; 325:607-610
  22. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med 1999; 131:281-303
  23. Sherifali D, Nerenberg K, Pullenayegum E, Cheng JE, Gerstein HC. The effect of oral antidiabetic agents on A1C levels: a systematic review and meta-analysis. Diabetes Care 2010; 33:1859-1864
  24. Phung OJ, Scholle JM, Talwar M, Coleman CI. Effect of noninsulin antidiabetic drugs added to metformin therapy on glycemic control, weight gain, and hypoglycemia in type 2 diabetes. JAMA 2010; 303:1410-1418
  25. Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, Kravitz BG, Lachin JM, O'Neill MC, Zinman B, Viberti G. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med2006; 355:2427-2443
  26. Goldner MG, Knatterud GL, Prout TE. Effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. 3. Clinical implications of UGDP results. JAMA 1971; 218:1400-1410
  27. Meinert CL, Knatterud GL, Prout TE, Klimt CR. A study of the effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. II. Mortality results. Diabetes 1970; 19:Suppl:789-830
  28. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:837-853
  29. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients With Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019;
  30. Silbert R, Salcido-Montenegro A, Rodriguez-Gutierrez R, Katabi A, McCoy RG. Hypoglycemia Among Patients with Type 2 Diabetes: Epidemiology, Risk Factors, and Prevention Strategies. Curr Diab Rep 2018; 18:53
  31. Middleton TL, Wong J, Molyneaux L, Brooks BA, Yue DK, Twigg SM, Wu T. Cardiac Effects of Sulfonylurea-Related Hypoglycemia. Diabetes Care 2017; 40:663-670
  32. Hay LC, Wilmshurst EG, Fulcher G. Unrecognized hypo- and hyperglycemia in well-controlled patients with type 2 diabetes mellitus: the results of continuous glucose monitoring. Diabetes Technol Ther 2003; 5:19-26
  33. Gangji AS, Cukierman T, Gerstein HC, Goldsmith CH, Clase CM. A systematic review and meta-analysis of hypoglycemia and cardiovascular events: a comparison of glyburide with other secretagogues and with insulin. Diabetes Care 2007; 30:389-394
  34. Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, Neumiller JJ, Patel UD, Ratner RE, Whaley-Connell AT, Molitch ME. Diabetic kidney disease: a report from an ADA Consensus Conference. Diabetes Care 2014; 37:2864-2883
  35. By the American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2019 Updated AGS Beers Criteria(R) for Potentially Inappropriate Medication Use in Older Adults. J Am Geriatr Soc2019; 67:674-694
  36. Guardado-Mendoza R, Prioletta A, Jimenez-Ceja LM, Sosale A, Folli F. The role of nateglinide and repaglinide, derivatives of meglitinide, in the treatment of type 2 diabetes mellitus. Arch Med Sci 2013; 9:936-943
  37. Tran L, Zielinski A, Roach AH, Jende JA, Householder AM, Cole EE, Atway SA, Amornyard M, Accursi ML, Shieh SW, Thompson EE. Pharmacologic treatment of type 2 diabetes: oral medications. Ann Pharmacother2015; 49:540-556
  38. Scott LJ. Repaglinide: a review of its use in type 2 diabetes mellitus. Drugs 2012; 72:249-272
  39. Horton ES, Clinkingbeard C, Gatlin M, Foley J, Mallows S, Shen S. Nateglinide alone and in combination with metformin improves glycemic control by reducing mealtime glucose levels in type 2 diabetes. Diabetes Care2000; 23:1660-1665
  40. Rosenstock J, Hassman DR, Madder RD, Brazinsky SA, Farrell J, Khutoryansky N, Hale PM. Repaglinide versus nateglinide monotherapy: a randomized, multicenter study. Diabetes Care 2004; 27:1265-1270
  41. Navigator Study Group, Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 2010; 362:1463-1476
  42. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia 2017; 60:1577-1585
  43. Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: from mechanisms of action to therapies. Cell Metab 2014; 20:953-966
  44. LaMoia TE, Shulman GI. Cellular and Molecular Mechanisms of Metformin Action. Endocr Rev 2021; 42:77-96
  45. Buse JB, DeFronzo RA, Rosenstock J, Kim T, Burns C, Skare S, Baron A, Fineman M. The Primary Glucose-Lowering Effect of Metformin Resides in the Gut, Not the Circulation: Results From Short-term Pharmacokinetic and 12-Week Dose-Ranging Studies. Diabetes Care 2016; 39:198-205
  46. Pollak M. The effects of metformin on gut microbiota and the immune system as research frontiers. Diabetologia 2017; 60:1662-1667
  47. Sanchez-Rangel E, Inzucchi SE. Metformin: clinical use in type 2 diabetes. Diabetologia 2017; 60:1586-1593
  48. Williams LK, Padhukasahasram B, Ahmedani BK, Peterson EL, Wells KE, Gonzalez Burchard E, Lanfear DE. Differing effects of metformin on glycemic control by race-ethnicity. J Clin Endocrinol Metab 2014; 99:3160-3168
  49. Fujioka K, Pans M, Joyal S. Glycemic control in patients with type 2 diabetes mellitus switched from twice-daily immediate-release metformin to a once-daily extended-release formulation. Clin Ther 2003; 25:515-529
  50. Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT. The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 2004; 256:1-14
  51. Feingold KR. Role of Glucose and Lipids in the Cardiovascular Disease of Patients with Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2020.
  52. Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, Temprosa M. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the diabetes prevention program. Diabetes Care2005; 28:888-894
  53. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:854-865
  54. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359:1577-1589
  55. Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on metabolism and microvascular and macrovascular disease in patients with type 2 diabetes mellitus. Arch Intern Med 2009; 169:616-625
  56. Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, Zhou Z, Tang W, Zhao J, Cui L, Zou D, Wang D, Li H, Liu C, Wu G, Shen J, Zhu D, Wang W, Shen W, Ning G. Effects of metformin versus glipizide on cardiovascular outcomes in patients with type 2 diabetes and coronary artery disease. Diabetes Care 2013; 36:1304-1311
  57. Legro RS. Evaluation and Treatment of Polycystic Ovary Syndrome. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2017.
  58. Heckman-Stoddard BM, DeCensi A, Sahasrabuddhe VV, Ford LG. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia 2017; 60:1639-1647
  59. Mallik R, Chowdhury TA. Metformin in cancer. Diabetes Res Clin Pract 2018; 143:409-419
  60. Goodwin PJ, Chen BE, Gelmon KA, Whelan TJ, Ennis M, Lemieux J, Ligibel JA, Hershman DL, Mayer IA, Hobday TJ, Bliss JM, Rastogi P, Rabaglio-Poretti M, Mukherjee SD, Mackey JR, Abramson VG, Oja C, Wesolowski R, Thompson AM, Rea DW, Stos PM, Shepherd LE, Stambolic V, Parulekar WR. Effect of Metformin vs Placebo on Invasive Disease-Free Survival in Patients With Breast Cancer: The MA.32 Randomized Clinical Trial. JAMA 2022; 327:1963-1973
  61. Dujic T, Zhou K, Donnelly LA, Tavendale R, Palmer CN, Pearson ER. Association of Organic Cation Transporter 1 With Intolerance to Metformin in Type 2 Diabetes: A GoDARTS Study. Diabetes 2015; 64:1786-1793
  62. Dawed AY, Zhou K, van Leeuwen N, Mahajan A, Robertson N, Koivula R, Elders PJM, Rauh SP, Jones AG, Holl RW, Stingl JC, Franks PW, McCarthy MI, t Hart LM, Pearson ER. Variation in the Plasma Membrane Monoamine Transporter (PMAT) (Encoded by SLC29A4) and Organic Cation Transporter 1 (OCT1) (Encoded by SLC22A1) and Gastrointestinal Intolerance to Metformin in Type 2 Diabetes: An IMI DIRECT Study. Diabetes Care 2019; 42:1027-1033
  63. de Jager J, Kooy A, Lehert P, Wulffele MG, van der Kolk J, Bets D, Verburg J, Donker AJ, Stehouwer CD. Long term treatment with metformin in patients with type 2 diabetes and risk of vitamin B-12 deficiency: randomised placebo controlled trial. BMJ 2010; 340:c2181
  64. Aroda VR, Edelstein SL, Goldberg RB, Knowler WC, Marcovina SM, Orchard TJ, Bray GA, Schade DS, Temprosa MG, White NH, Crandall JP. Long-term Metformin Use and Vitamin B12 Deficiency in the Diabetes Prevention Program Outcomes Study. J Clin Endocrinol Metab 2016; 101:1754-1761
  65. American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2019. Diabetes Care 2019; 42:S90-S102
  66. Yki-Jarvinen H. Thiazolidinediones. N Engl J Med 2004; 351:1106-1118
  67. Willson TM, Lambert MH, Kliewer SA. Peroxisome proliferator-activated receptor gamma and metabolic disease. Annu Rev Biochem 2001; 70:341-367
  68. Memon RA, Tecott LH, Nonogaki K, Beigneux A, Moser AH, Grunfeld C, Feingold KR. Up-regulation of peroxisome proliferator-activated receptors (PPAR-alpha) and PPAR-gamma messenger ribonucleic acid expression in the liver in murine obesity: troglitazone induces expression of PPAR-gamma-responsive adipose tissue-specific genes in the liver of obese diabetic mice. Endocrinology 2000; 141:4021-4031
  69. Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen AL, Schneider RL. Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: a 6-month randomized placebo-controlled dose-response study. The Pioglitazone 001 Study Group. Diabetes Care 2000; 23:1605-1611
  70. Dennis JM, Shields BM, Henley WE, Jones AG, Hattersley AT. Disease progression and treatment response in data-driven subgroups of type 2 diabetes compared with models based on simple clinical features: an analysis using clinical trial data. Lancet Diabetes Endocrinol 2019; 7:442-451
  71. Tan MH, Baksi A, Krahulec B, Kubalski P, Stankiewicz A, Urquhart R, Edwards G, Johns D. Comparison of pioglitazone and gliclazide in sustaining glycemic control over 2 years in patients with type 2 diabetes. Diabetes Care 2005; 28:544-550
  72. Gastaldelli A, Ferrannini E, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab 2007; 292:E871-883
  73. Xiang AH, Peters RK, Kjos SL, Marroquin A, Goico J, Ochoa C, Kawakubo M, Buchanan TA. Effect of pioglitazone on pancreatic beta-cell function and diabetes risk in Hispanic women with prior gestational diabetes. Diabetes 2006; 55:517-522
  74. DeFronzo RA, Inzucchi S, Abdul-Ghani M, Nissen SE. Pioglitazone: The forgotten, cost-effective cardioprotective drug for type 2 diabetes. Diab Vasc Dis Res 2019; 16:133-143
  75. Sarafidis PA, Stafylas PC, Georgianos PI, Saratzis AN, Lasaridis AN. Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis. Am J Kidney Dis 2010; 55:835-847
  76. Qayyum R, Adomaityte J. A meta-analysis of the effect of thiazolidinediones on blood pressure. J Clin Hypertens (Greenwich) 2006; 8:19-28
  77. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-2296
  78. Spanheimer R, Betteridge DJ, Tan MH, Ferrannini E, Charbonnel B. Long-term lipid effects of pioglitazone by baseline anti-hyperglycemia medication therapy and statin use from the PROactive experience (PROactive 14). Am J Cardiol 2009; 104:234-239
  79. Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, French BC, Phillips JA, Mucksavage ML, Wilensky RL, Mohler ER, Rothblat GH, Rader DJ. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med 2011; 364:127-135
  80. Deeg MA, Buse JB, Goldberg RB, Kendall DM, Zagar AJ, Jacober SJ, Khan MA, Perez AT, Tan MH. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2007; 30:2458-2464
  81. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, Tan MH, Khan MA, Perez AT, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2005; 28:1547-1554
  82. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366:1279-1289
  83. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr., Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O'Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med 2016; 374:1321-1331
  84. Spence JD, Viscoli CM, Inzucchi SE, Dearborn-Tomazos J, Ford GA, Gorman M, Furie KL, Lovejoy AM, Young LH, Kernan WN. Pioglitazone Therapy in Patients With Stroke and Prediabetes: A Post Hoc Analysis of the IRIS Randomized Clinical Trial. JAMA Neurol 2019; 76:526-535
  85. Vaccaro O, Masulli M, Nicolucci A, Bonora E, Del Prato S, Maggioni AP, Rivellese AA, Squatrito S, Giorda CB, Sesti G, Mocarelli P, Lucisano G, Sacco M, Signorini S, Cappellini F, Perriello G, Babini AC, Lapolla A, Gregori G, Giordano C, Corsi L, Buzzetti R, Clemente G, Di Cianni G, Iannarelli R, Cordera R, La Macchia O, Zamboni C, Scaranna C, Boemi M, Iovine C, Lauro D, Leotta S, Dall'Aglio E, Cannarsa E, Tonutti L, Pugliese G, Bossi AC, Anichini R, Dotta F, Di Benedetto A, Citro G, Antenucci D, Ricci L, Giorgino F, Santini C, Gnasso A, De Cosmo S, Zavaroni D, Vedovato M, Consoli A, Calabrese M, di Bartolo P, Fornengo P, Riccardi G. Effects on the incidence of cardiovascular events of the addition of pioglitazone versus sulfonylureas in patients with type 2 diabetes inadequately controlled with metformin (TOSCA.IT): a randomised, multicentre trial. Lancet Diabetes Endocrinol 2017; 5:887-897
  86. Vaccaro O, Lucisano G, Masulli M, Bonora E, Del Prato S, Rivellese AA, Giorda CB, Mocarelli P, Squatrito S, Maggioni AP, Riccardi G, Nicolucci A. Cardiovascular Effects of Pioglitazone or Sulfonylureas According to Pretreatment Risk: Moving Toward Personalized Care. J Clin Endocrinol Metab 2019; 104:3296-3302
  87. Langenfeld MR, Forst T, Hohberg C, Kann P, Lubben G, Konrad T, Fullert SD, Sachara C, Pfutzner A. Pioglitazone decreases carotid intima-media thickness independently of glycemic control in patients with type 2 diabetes mellitus: results from a controlled randomized study. Circulation 2005; 111:2525-2531
  88. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D'Agostino RB, Sr., Perez A, Provost JC, Haffner SM. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial. JAMA 2006; 296:2572-2581
  89. Saremi A, Schwenke DC, Buchanan TA, Hodis HN, Mack WJ, Banerji M, Bray GA, Clement SC, Henry RR, Kitabchi AE, Mudaliar S, Ratner RE, Stentz FB, Musi N, Tripathy D, DeFronzo RA, Reaven PD. Pioglitazone slows progression of atherosclerosis in prediabetes independent of changes in cardiovascular risk factors. Arterioscler Thromb Vasc Biol 2013; 33:393-399
  90. Nissen SE, Nicholls SJ, Wolski K, Nesto R, Kupfer S, Perez A, Jure H, De Larochelliere R, Staniloae CS, Mavromatis K, Saw J, Hu B, Lincoff AM, Tuzcu EM. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA2008; 299:1561-1573
  91. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007; 356:2457-2471
  92. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007; 298:1189-1195
  93. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009; 373:2125-2135
  94. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J 2013; 166:240-249 e241
  95. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation 2013; 128:785-794
  96. Kim KS, Lee BW, Kim YJ, Lee DH, Cha BS, Park CY. Nonalcoholic Fatty Liver Disease and Diabetes: Part II: Treatment. Diabetes Metab J 2019; 43:127-143
  97. Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, Balas B, Gastaldelli A, Tio F, Pulcini J, Berria R, Ma JZ, Dwivedi S, Havranek R, Fincke C, DeFronzo R, Bannayan GA, Schenker S, Cusi K. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med 2006; 355:2297-2307
  98. Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, Tio F, Hardies J, Darland C, Musi N, Webb A, Portillo-Sanchez P. Long-Term Pioglitazone Treatment for Patients With Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus: A Randomized Trial. Ann Intern Med 2016; 165:305-315
  99. Musso G, Cassader M, Paschetta E, Gambino R. Thiazolidinediones and Advanced Liver Fibrosis in Nonalcoholic Steatohepatitis: A Meta-analysis. JAMA Intern Med 2017; 177:633-640
  100. Rosenstock J, Vico M, Wei L, Salsali A, List JF. Effects of dapagliflozin, an SGLT2 inhibitor, on HbA(1c), body weight, and hypoglycemia risk in patients with type 2 diabetes inadequately controlled on pioglitazone monotherapy. Diabetes Care 2012; 35:1473-1478
  101. Abdul-Ghani MA, Puckett C, Triplitt C, Maggs D, Adams J, Cersosimo E, DeFronzo RA. Initial combination therapy with metformin, pioglitazone and exenatide is more effective than sequential add-on therapy in subjects with new-onset diabetes. Results from the Efficacy and Durability of Initial Combination Therapy for Type 2 Diabetes (EDICT): a randomized trial. Diabetes Obes Metab 2015; 17:268-275
  102. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R. Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. Diabetes Care 2004; 27:256-263
  103. Mudaliar S, Chang AR, Henry RR. Thiazolidinediones, peripheral edema, and type 2 diabetes: incidence, pathophysiology, and clinical implications. Endocr Pract 2003; 9:406-416
  104. Guan Y, Hao C, Cha DR, Rao R, Lu W, Kohan DE, Magnuson MA, Redha R, Zhang Y, Breyer MD. Thiazolidinediones expand body fluid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption. Nat Med 2005; 11:861-866
  105. Zanchi A, Chiolero A, Maillard M, Nussberger J, Brunner HR, Burnier M. Effects of the peroxisomal proliferator-activated receptor-gamma agonist pioglitazone on renal and hormonal responses to salt in healthy men. J Clin Endocrinol Metab 2004; 89:1140-1145
  106. de Jong M, van der Worp HB, van der Graaf Y, Visseren FLJ, Westerink J. Pioglitazone and the secondary prevention of cardiovascular disease. A meta-analysis of randomized-controlled trials. Cardiovasc Diabetol2017; 16:134
  107. Hernandez AV, Usmani A, Rajamanickam A, Moheet A. Thiazolidinediones and risk of heart failure in patients with or at high risk of type 2 diabetes mellitus: a meta-analysis and meta-regression analysis of placebo-controlled randomized clinical trials. Am J Cardiovasc Drugs 2011; 11:115-128
  108. Masoudi FA, Inzucchi SE, Wang Y, Havranek EP, Foody JM, Krumholz HM. Thiazolidinediones, metformin, and outcomes in older patients with diabetes and heart failure: an observational study. Circulation 2005; 111:583-590
  109. Kahn SE, Zinman B, Lachin JM, Haffner SM, Herman WH, Holman RR, Kravitz BG, Yu D, Heise MA, Aftring RP, Viberti G. Rosiglitazone-associated fractures in type 2 diabetes: an Analysis from A Diabetes Outcome Progression Trial (ADOPT). Diabetes Care 2008; 31:845-851
  110. Dormandy J, Bhattacharya M, van Troostenburg de Bruyn AR. Safety and tolerability of pioglitazone in high-risk patients with type 2 diabetes: an overview of data from PROactive. Drug Saf 2009; 32:187-202
  111. Viscoli CM, Inzucchi SE, Young LH, Insogna KL, Conwit R, Furie KL, Gorman M, Kelly MA, Lovejoy AM, Kernan WN. Pioglitazone and Risk for Bone Fracture: Safety Data From a Randomized Clinical Trial. J Clin Endocrinol Metab 2017; 102:914-922
  112. Zhu ZN, Jiang YF, Ding T. Risk of fracture with thiazolidinediones: an updated meta-analysis of randomized clinical trials. Bone 2014; 68:115-123
  113. Schwartz AV, Chen H, Ambrosius WT, Sood A, Josse RG, Bonds DE, Schnall AM, Vittinghoff E, Bauer DC, Banerji MA, Cohen RM, Hamilton BP, Isakova T, Sellmeyer DE, Simmons DL, Shibli-Rahhal A, Williamson JD, Margolis KL. Effects of TZD Use and Discontinuation on Fracture Rates in ACCORD Bone Study. J Clin Endocrinol Metab 2015; 100:4059-4066
  114. Schwartz AV, Sellmeyer DE, Vittinghoff E, Palermo L, Lecka-Czernik B, Feingold KR, Strotmeyer ES, Resnick HE, Carbone L, Beamer BA, Park SW, Lane NE, Harris TB, Cummings SR. Thiazolidinedione use and bone loss in older diabetic adults. J Clin Endocrinol Metab 2006; 91:3349-3354
  115. Davidson MB. Pioglitazone (Actos) and bladder cancer: Legal system triumphs over the evidence. J Diabetes Complications 2016; 30:981-985
  116. Erdmann E, Harding S, Lam H, Perez A. Ten-year observational follow-up of PROactive: a randomized cardiovascular outcomes trial evaluating pioglitazone in type 2 diabetes. Diabetes Obes Metab 2016; 18:266-273
  117. Lewis JD, Habel LA, Quesenberry CP, Strom BL, Peng T, Hedderson MM, Ehrlich SF, Mamtani R, Bilker W, Vaughn DJ, Nessel L, Van Den Eeden SK, Ferrara A. Pioglitazone Use and Risk of Bladder Cancer and Other Common Cancers in Persons With Diabetes. JAMA 2015; 314:265-277
  118. Levin D, Bell S, Sund R, Hartikainen SA, Tuomilehto J, Pukkala E, Keskimaki I, Badrick E, Renehan AG, Buchan IE, Bowker SL, Minhas-Sandhu JK, Zafari Z, Marra C, Johnson JA, Stricker BH, Uitterlinden AG, Hofman A, Ruiter R, de Keyser CE, MacDonald TM, Wild SH, McKeigue PM, Colhoun HM. Pioglitazone and bladder cancer risk: a multipopulation pooled, cumulative exposure analysis. Diabetologia 2015; 58:493-504
  119. Idris I, Warren G, Donnelly R. Association between thiazolidinedione treatment and risk of macular edema among patients with type 2 diabetes. Arch Intern Med 2012; 172:1005-1011
  120. Ryan EH, Jr., Han DP, Ramsay RC, Cantrill HL, Bennett SR, Dev S, Williams DF. Diabetic macular edema associated with glitazone use. Retina 2006; 26:562-570
  121. Coniff R, Krol A. Acarbose: a review of US clinical experience. Clin Ther 1997; 19:16-26; discussion 12-13
  122. Van de Laar FA, Lucassen PL, Akkermans RP, Van de Lisdonk EH, Rutten GE, Van Weel C. Alpha-glucosidase inhibitors for type 2 diabetes mellitus. Cochrane Database Syst Rev 2005:CD003639
  123. Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance: the STOP-NIDDM trial. JAMA 2003; 290:486-494
  124. Yun P, Du AM, Chen XJ, Liu JC, Xiao H. Effect of Acarbose on Long-Term Prognosis in Acute Coronary Syndromes Patients with Newly Diagnosed Impaired Glucose Tolerance. J Diabetes Res 2016; 2016:1602083
  125. Holman RR, Coleman RL, Chan JCN, Chiasson JL, Feng H, Ge J, Gerstein HC, Gray R, Huo Y, Lang Z, McMurray JJ, Ryden L, Schroder S, Sun Y, Theodorakis MJ, Tendera M, Tucker L, Tuomilehto J, Wei Y, Yang W, Wang D, Hu D, Pan C. Effects of acarbose on cardiovascular and diabetes outcomes in patients with coronary heart disease and impaired glucose tolerance (ACE): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:877-886
  126. Domecq JP, Prutsky G, Leppin A, Sonbol MB, Altayar O, Undavalli C, Wang Z, Elraiyah T, Brito JP, Mauck KF, Lababidi MH, Prokop LJ, Asi N, Wei J, Fidahussein S, Montori VM, Murad MH. Clinical review: Drugs commonly associated with weight change: a systematic review and meta-analysis. J Clin Endocrinol Metab 2015; 100:363-370
  127. Lupsa BC, Inzucchi SE. Use of SGLT2 inhibitors in type 2 diabetes: weighing the risks and benefits. Diabetologia 2018; 61:2118-2125
  128. Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, renal function and blood pressure. Diabetologia 2018; 61:2098-2107
  129. Rahmoune H, Thompson PW, Ward JM, Smith CD, Hong G, Brown J. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with non-insulin-dependent diabetes. Diabetes 2005; 54:3427-3434
  130. Monami M, Liistro F, Scatena A, Nreu B, Mannucci E. Short and medium-term efficacy of sodium glucose co-transporter-2 (SGLT-2) inhibitors: A meta-analysis of randomized clinical trials. Diabetes Obes Metab 2018; 20:1213-1222
  131. Ferrannini G, Hach T, Crowe S, Sanghvi A, Hall KD, Ferrannini E. Energy Balance After Sodium-Glucose Cotransporter 2 Inhibition. Diabetes Care 2015; 38:1730-1735
  132. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117-2128
  133. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644-657
  134. Mazidi M, Rezaie P, Gao HK, Kengne AP. Effect of Sodium-Glucose Cotransport-2 Inhibitors on Blood Pressure in People With Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis of 43 Randomized Control Trials With 22 528 Patients. J Am Heart Assoc 2017; 6
  135. Sanchez-Garcia A, Simental-Mendia M, Millan-Alanis JM, Simental-Mendia LE. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol Res 2020; 160:105068
  136. van Baar MJB, van Ruiten CC, Muskiet MHA, van Bloemendaal L, RG IJ, van Raalte DH. SGLT2 Inhibitors in Combination Therapy: From Mechanisms to Clinical Considerations in Type 2 Diabetes Management. Diabetes Care 2018; 41:1543-1556
  137. Chung MC, Hung PH, Hsiao PJ, Wu LY, Chang CH, Wu MJ, Shieh JJ, Chung CJ. Association of Sodium-Glucose Transport Protein 2 Inhibitor Use for Type 2 Diabetes and Incidence of Gout in Taiwan. JAMA Netw Open 2021; 4:e2135353
  138. Fitchett D, Inzucchi SE, Cannon CP, McGuire DK, Scirica BM, Johansen OE, Sambevski S, Kaspers S, Pfarr E, George JT, Zinman B. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019; 139:1384-1395
  139. Fitchett D, Butler J, van de Borne P, Zinman B, Lachin JM, Wanner C, Woerle HJ, Hantel S, George JT, Johansen OE, Inzucchi SE. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME(R) trial. Eur Heart J 2018; 39:363-370
  140. Neuen BL, Ohkuma T, Neal B, Matthews DR, de Zeeuw D, Mahaffey KW, Fulcher G, Desai M, Li Q, Deng H, Rosenthal N, Jardine MJ, Bakris G, Perkovic V. Cardiovascular and Renal Outcomes With Canagliflozin According to Baseline Kidney Function. Circulation 2018; 138:1537-1550
  141. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019;
  142. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, Bajaj HS, Bull S, Cannon CP, Charytan DM, de Zeeuw D, Di Tanna GL, Greene T, Heerspink HJL, Levin A, Neal B, Pollock C, Qiu R, Sun T, Wheeler DC, Zhang H, Zinman B, Rosenthal N, Perkovic V. Renal, Cardiovascular, and Safety Outcomes of Canagliflozin by Baseline Kidney Function: A Secondary Analysis of the CREDENCE Randomized Trial. J Am Soc Nephrol2020; 31:1128-1139
  143. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med2019; 380:347-357
  144. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Nicolau JC, Gause-Nilsson IAM, Fredriksson M, Langkilde AM, Sabatine MS, Wiviott SD. Dapagliflozin and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus and Previous Myocardial Infarction. Circulation 2019; 139:2516-2527
  145. Kato ET, Silverman MG, Mosenzon O, Zelniker TA, Cahn A, Furtado RHM, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Bonaca MP, Ruff CT, Desai AS, Goto S, Johansson PA, Gause-Nilsson I, Johanson P, Langkilde AM, Raz I, Sabatine MS, Wiviott SD. Effect of Dapagliflozin on Heart Failure and Mortality in Type 2 Diabetes Mellitus. Circulation 2019; 139:2528-2536
  146. Cahn A, Raz I, Leiter LA, Mosenzon O, Murphy SA, Goodrich EL, Yanuv I, Rozenberg A, Bhatt DL, McGuire DK, Wilding JPH, Gause-Nilsson IAM, Langkilde AM, Sabatine MS, Wiviott SD. Cardiovascular, Renal, and Metabolic Outcomes of Dapagliflozin Versus Placebo in a Primary Cardiovascular Prevention Cohort: Analyses From DECLARE-TIMI 58. Diabetes Care 2021; 44:1159-1167
  147. Zelniker TA, Bonaca MP, Furtado RHM, Mosenzon O, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Budaj A, Kiss RG, Padilla F, Gause-Nilsson I, Langkilde AM, Raz I, Sabatine MS, Wiviott SD. Effect of Dapagliflozin on Atrial Fibrillation in Patients With Type 2 Diabetes Mellitus: Insights From the DECLARE-TIMI 58 Trial. Circulation 2020; 141:1227-1234
  148. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med 2020; 383:1425-1435
  149. Cosentino F, Cannon CP, Cherney DZI, Masiukiewicz U, Pratley R, Dagogo Jack S, Frederich R, Charbonnel B, Mancuso J, Shih WJ, Terra SG, Cater NB, Gantz I, McGuire DK. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients with Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease: Results of the VERTIS CV Trial. Circulation 2020;
  150. McMurray JJV, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Belohlavek J, Bohm M, Chiang CE, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukat A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O'Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjostrand M, Langkilde AM. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med2019; 381:1995-2008
  151. Curtain JP, Docherty KF, Jhund PS, Petrie MC, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Bengtsson O, Langkilde AM, Sjostrand M, Solomon SD, McMurray JJV. Effect of dapagliflozin on ventricular arrhythmias, resuscitated cardiac arrest, or sudden death in DAPA-HF. Eur Heart J 2021;
  152. Petrie MC, Verma S, Docherty KF, Inzucchi SE, Anand I, Belohlavek J, Bohm M, Chiang CE, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukat A, Ge J, Howlett J, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O'Meara E, Vinh PN, Schou M, Tereshchenko S, Kober L, Kosiborod MN, Langkilde AM, Martinez FA, Ponikowski P, Sabatine MS, Sjostrand M, Solomon SD, Johanson P, Greasley PJ, Boulton D, Bengtsson O, Jhund PS, McMurray JJV. Effect of Dapagliflozin on Worsening Heart Failure and Cardiovascular Death in Patients With Heart Failure With and Without Diabetes. JAMA 2020;
  153. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Bohm M, Choi DJ, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR, Kaul S, Brunner-La Rocca HP, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med 2020; 383:1413-1424
  154. Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjostrom CD, Toto RD, Langkilde AM, Wheeler DC. Dapagliflozin in Patients with Chronic Kidney Disease. N Engl J Med 2020; 383:1436-1446
  155. Packer M, Butler J, Zannad F, Filippatos G, Ferreira JP, Pocock SJ, Carson P, Anand I, Doehner W, Haass M, Komajda M, Miller A, Pehrson S, Teerlink JR, Schnaidt S, Zeller C, Schnee JM, Anker SD. Effect of Empagliflozin on Worsening Heart Failure Events in Patients With Heart Failure and Preserved Ejection Fraction: EMPEROR-Preserved Trial. Circulation 2021; 144:1284-1294
  156. Salah HM, Al'Aref SJ, Khan MS, Al-Hawwas M, Vallurupalli S, Mehta JL, Mounsey JP, Greene SJ, McGuire DK, Lopes RD, Fudim M. Effect of sodium-glucose cotransporter 2 inhibitors on cardiovascular and kidney outcomes-Systematic review and meta-analysis of randomized placebo-controlled trials. Am Heart J 2021; 232:10-22
  157. Voors AA, Angermann CE, Teerlink JR, Collins SP, Kosiborod M, Biegus J, Ferreira JP, Nassif ME, Psotka MA, Tromp J, Borleffs CJW, Ma C, Comin-Colet J, Fu M, Janssens SP, Kiss RG, Mentz RJ, Sakata Y, Schirmer H, Schou M, Schulze PC, Spinarova L, Volterrani M, Wranicz JK, Zeymer U, Zieroth S, Brueckmann M, Blatchford JP, Salsali A, Ponikowski P. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial. Nat Med 2022; 28:568-574
  158. Lytvyn Y, Bjornstad P, Udell JA, Lovshin JA, Cherney DZI. Sodium Glucose Cotransporter-2 Inhibition in Heart Failure: Potential Mechanisms, Clinical Applications, and Summary of Clinical Trials. Circulation 2017; 136:1643-1658
  159. Inzucchi SE, Zinman B, Fitchett D, Wanner C, Ferrannini E, Schumacher M, Schmoor C, Ohneberg K, Johansen OE, George JT, Hantel S, Bluhmki E, Lachin JM. How Does Empagliflozin Reduce Cardiovascular Mortality? Insights From a Mediation Analysis of the EMPA-REG OUTCOME Trial. Diabetes Care 2018; 41:356-363
  160. Ferrannini E. Sodium-Glucose Co-transporters and Their Inhibition: Clinical Physiology. Cell Metab 2017; 26:27-38
  161. Mudaliar S, Alloju S, Henry RR. Can a Shift in Fuel Energetics Explain the Beneficial Cardiorenal Outcomes in the EMPA-REG OUTCOME Study? A Unifying Hypothesis. Diabetes Care 2016; 39:1115-1122
  162. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia 2018; 61:2108-2117
  163. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, Johansen OE, Woerle HJ, Broedl UC, Zinman B. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med 2016; 375:323-334
  164. Cherney DZI, Zinman B, Inzucchi SE, Koitka-Weber A, Mattheus M, von Eynatten M, Wanner C. Effects of empagliflozin on the urinary albumin-to-creatinine ratio in patients with type 2 diabetes and established cardiovascular disease: an exploratory analysis from the EMPA-REG OUTCOME randomised, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:610-621
  165. Perkovic V, de Zeeuw D, Mahaffey KW, Fulcher G, Erondu N, Shaw W, Barrett TD, Weidner-Wells M, Deng H, Matthews DR, Neal B. Canagliflozin and renal outcomes in type 2 diabetes: results from the CANVAS Program randomised clinical trials. Lancet Diabetes Endocrinol 2018; 6:691-704
  166. Neuen BL, Ohkuma T, Neal B, Matthews DR, de Zeeuw D, Mahaffey KW, Fulcher G, Li Q, Jardine M, Oh R, Heerspink HL, Perkovic V. Effect of Canagliflozin on Renal and Cardiovascular Outcomes across Different Levels of Albuminuria: Data from the CANVAS Program. J Am Soc Nephrol 2019; 30:2229-2242
  167. Mosenzon O, Wiviott SD, Cahn A, Rozenberg A, Yanuv I, Goodrich EL, Murphy SA, Heerspink HJL, Zelniker TA, Dwyer JP, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Kato ET, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS, Raz I. Effects of dapagliflozin on development and progression of kidney disease in patients with type 2 diabetes: an analysis from the DECLARE-TIMI 58 randomised trial. Lancet Diabetes Endocrinol 2019;
  168. Chertow G, Vart P, Jongs N, Toto R, Gorriz JL, Hou FF, McMurray J, Correa-Rotter R, Rossing P, Sjostrom CD, Stefansson B, Langkilde AM, Wheeler D, Heerspink H. Effects of Dapagliflozin in Stage 4 Chronic Kidney Disease. J Am Soc Nephrol 2021;
  169. Wheeler DC, Stefansson BV, Jongs N, Chertow GM, Greene T, Hou FF, McMurray JJV, Correa-Rotter R, Rossing P, Toto RD, Sjostrom CD, Langkilde AM, Heerspink HJL. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol 2021; 9:22-31
  170. Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Furtado RHM, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Sabatine MS. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet 2019; 393:31-39
  171. Muskiet MHA, Wheeler DC, Heerspink HJL. New pharmacological strategies for protecting kidney function in type 2 diabetes. Lancet Diabetes Endocrinol 2019; 7:397-412
  172. Shimizu M, Suzuki K, Kato K, Jojima T, Iijima T, Murohisa T, Iijima M, Takekawa H, Usui I, Hiraishi H, Aso Y. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes Obes Metab 2019; 21:285-292
  173. Eriksson JW, Lundkvist P, Jansson PA, Johansson L, Kvarnstrom M, Moris L, Miliotis T, Forsberg GB, Riserus U, Lind L, Oscarsson J. Effects of dapagliflozin and n-3 carboxylic acids on non-alcoholic fatty liver disease in people with type 2 diabetes: a double-blind randomised placebo-controlled study. Diabetologia 2018; 61:1923-1934
  174. Sattar N, Fitchett D, Hantel S, George JT, Zinman B. Empagliflozin is associated with improvements in liver enzymes potentially consistent with reductions in liver fat: results from randomised trials including the EMPA-REG OUTCOME(R) trial. Diabetologia 2018; 61:2155-2163
  175. Kuchay MS, Krishan S, Mishra SK, Farooqui KJ, Singh MK, Wasir JS, Bansal B, Kaur P, Jevalikar G, Gill HK, Choudhary NS, Mithal A. Effect of Empagliflozin on Liver Fat in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial (E-LIFT Trial). Diabetes Care 2018; 41:1801-1808
  176. Raj H, Durgia H, Palui R, Kamalanathan S, Selvarajan S, Kar SS, Sahoo J. SGLT-2 inhibitors in non-alcoholic fatty liver disease patients with type 2 diabetes mellitus: A systematic review. World J Diabetes 2019; 10:114-132
  177. Kahl S, Gancheva S, Strassburger K, Herder C, Machann J, Katsuyama H, Kabisch S, Henkel E, Kopf S, Lagerpusch M, Kantartzis K, Kupriyanova Y, Markgraf D, van Gemert T, Knebel B, Wolkersdorfer MF, Kuss O, Hwang JH, Bornstein SR, Kasperk C, Stefan N, Pfeiffer A, Birkenfeld AL, Roden M. Empagliflozin Effectively Lowers Liver Fat Content in Well-Controlled Type 2 Diabetes: A Randomized, Double-Blind, Phase 4, Placebo-Controlled Trial. Diabetes Care 2020; 43:298-305
  178. Akuta N, Watanabe C, Kawamura Y, Arase Y, Saitoh S, Fujiyama S, Sezaki H, Hosaka T, Kobayashi M, Kobayashi M, Suzuki Y, Suzuki F, Ikeda K, Kumada H. Effects of a sodium-glucose cotransporter 2 inhibitor in nonalcoholic fatty liver disease complicated by diabetes mellitus: Preliminary prospective study based on serial liver biopsies. Hepatol Commun 2017; 1:46-52
  179. Silverii GA, Monami M, Mannucci E. Sodium-glucose co-transporter-2 inhibitors and all-cause mortality: A meta-analysis of randomized controlled trials. Diabetes Obes Metab 2021; 23:1052-1056
  180. Shi FH, Li H, Yue J, Jiang YH, Gu ZC, Ma J, Lin HW. Clinical Adverse Events of High-Dose vs Low-Dose Sodium-Glucose Cotransporter 2 Inhibitors in Type 2 Diabetes: A Meta-Analysis of 51 Randomized Clinical Trials. J Clin Endocrinol Metab 2020; 105
  181. Lin DS, Lee JK, Chen WJ. Clinical Adverse Events Associated with Sodium-Glucose Cotransporter 2 Inhibitors: A Meta-Analysis Involving 10 Randomized Clinical Trials and 71 553 Individuals. J Clin Endocrinol Metab 2021; 106:2133-2145
  182. Puckrin R, Saltiel MP, Reynier P, Azoulay L, Yu OHY, Filion KB. SGLT-2 inhibitors and the risk of infections: a systematic review and meta-analysis of randomized controlled trials. Acta Diabetol 2018; 55:503-514
  183. Nyirjesy P, Sobel JD, Fung A, Mayer C, Capuano G, Ways K, Usiskin K. Genital mycotic infections with canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes mellitus: a pooled analysis of clinical studies. Curr Med Res Opin 2014; 30:1109-1119
  184. Bersoff-Matcha SJ, Chamberlain C, Cao C, Kortepeter C, Chong WH. Fournier Gangrene Associated With Sodium-Glucose Cotransporter-2 Inhibitors: A Review of Spontaneous Postmarketing Cases. Ann Intern Med2019;
  185. Chia L, Crum-Cianflone NF. Emergence of multi-drug resistant organisms (MDROs) causing Fournier's gangrene. J Infect 2018; 76:38-43
  186. Dave CV, Schneeweiss S, Patorno E. Association of Sodium-Glucose Cotransporter 2 Inhibitor Treatment With Risk of Hospitalization for Fournier Gangrene Among Men. JAMA Intern Med 2019;
  187. Nadkarni GN, Ferrandino R, Chang A, Surapaneni A, Chauhan K, Poojary P, Saha A, Ferket B, Grams ME, Coca SG. Acute Kidney Injury in Patients on SGLT2 Inhibitors: A Propensity-Matched Analysis. Diabetes Care2017; 40:1479-1485
  188. Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, Mahaffey KW, Charytan DM, Wheeler DC, Arnott C, Bompoint S, Levin A, Jardine MJ. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2019; 7:845-854
  189. Cahn A, Mosenzon O, Wiviott SD, Rozenberg A, Yanuv I, Goodrich EL, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS, Raz I. Efficacy and Safety of Dapagliflozin in the Elderly: Analysis From the DECLARE-TIMI 58 Study. Diabetes Care2020; 43:468-475
  190. Fralick M, Schneeweiss S, Patorno E. Risk of Diabetic Ketoacidosis after Initiation of an SGLT2 Inhibitor. N Engl J Med 2017; 376:2300-2302
  191. Taylor SI, Blau JE, Rother KI. SGLT2 Inhibitors May Predispose to Ketoacidosis. J Clin Endocrinol Metab 2015; 100:2849-2852
  192. Hamblin PS, Wong R, Ekinci EI, Fourlanos S, Shah S, Jones AR, Hare MJL, Calder GL, Epa DS, George EM, Giri R, Kotowicz MA, Kyi M, Lafontaine N, MacIsaac RJ, Nolan BJ, O'Neal DN, Renouf D, Varadarajan S, Wong J, Xu S, Bach LA. SGLT2 Inhibitors Increase the Risk of Diabetic Ketoacidosis Developing in the Community and During Hospital Admission. J Clin Endocrinol Metab 2019; 104:3077-3087
  193. Watts NB, Bilezikian JP, Usiskin K, Edwards R, Desai M, Law G, Meininger G. Effects of Canagliflozin on Fracture Risk in Patients With Type 2 Diabetes Mellitus. J Clin Endocrinol Metab 2016; 101:157-166
  194. Li X, Li T, Cheng Y, Lu Y, Xue M, Xu L, Liu X, Yu X, Sun B, Chen L. Effects of SGLT2 inhibitors on fractures and bone mineral density in type 2 diabetes mellitus: an updated meta-analysis. Diabetes Metab Res Rev2019:e3170
  195. Bilezikian JP, Watts NB, Usiskin K, Polidori D, Fung A, Sullivan D, Rosenthal N. Evaluation of Bone Mineral Density and Bone Biomarkers in Patients With Type 2 Diabetes Treated With Canagliflozin. J Clin Endocrinol Metab 2016; 101:44-51
  196. Bolinder J, Ljunggren O, Johansson L, Wilding J, Langkilde AM, Sjostrom CD, Sugg J, Parikh S. Dapagliflozin maintains glycaemic control while reducing weight and body fat mass over 2 years in patients with type 2 diabetes mellitus inadequately controlled on metformin. Diabetes Obes Metab 2014; 16:159-169
  197. Rosenstock J, Frias J, Pall D, Charbonnel B, Pascu R, Saur D, Darekar A, Huyck S, Shi H, Lauring B, Terra SG. Effect of ertugliflozin on glucose control, body weight, blood pressure and bone density in type 2 diabetes mellitus inadequately controlled on metformin monotherapy (VERTIS MET). Diabetes Obes Metab 2018; 20:520-529
  198. Inzucchi SE, Iliev H, Pfarr E, Zinman B. Empagliflozin and Assessment of Lower-Limb Amputations in the EMPA-REG OUTCOME Trial. Diabetes Care 2018; 41:e4-e5
  199. Patoulias D, Papadopoulos C, Doumas M. Updated meta-analysis assessing the risk of amputation with sodium-glucose co-transporter-2 inhibitors in the hallmark cardiovascular and renal outcome trials. Diabetes Obes Metab 2021; 23:1063-1065
  200. Kosiborod MN, Esterline R, Furtado RHM, Oscarsson J, Gasparyan SB, Koch GG, Martinez F, Mukhtar O, Verma S, Chopra V, Buenconsejo J, Langkilde AM, Ambery P, Tang F, Gosch K, Windsor SL, Akin EE, Soares RVP, Moia DDF, Aboudara M, Hoffmann Filho CR, Feitosa ADM, Fonseca A, Garla V, Gordon RA, Javaheri A, Jaeger CP, Leaes PE, Nassif M, Pursley M, Silveira FS, Barroso WKS, Lazcano Soto JR, Nigro Maia L, Berwanger O. Dapagliflozin in patients with cardiometabolic risk factors hospitalised with COVID-19 (DARE-19): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol 2021;
  201. Rendell MS. Efficacy and safety of sotagliflozin in treating diabetes type 1. Expert Opin Pharmacother 2018; 19:307-315
  202. Cefalo CMA, Cinti F, Moffa S, Impronta F, Sorice GP, Mezza T, Pontecorvi A, Giaccari A. Sotagliflozin, the first dual SGLT inhibitor: current outlook and perspectives. Cardiovasc Diabetol 2019; 18:20
  203. Song P, Onishi A, Koepsell H, Vallon V. Sodium glucose cotransporter SGLT1 as a therapeutic target in diabetes mellitus. Expert Opin Ther Targets 2016; 20:1109-1125
  204. Nuffer W, Williams B, Trujillo JM. A review of sotagliflozin for use in type 1 diabetes. Ther Adv Endocrinol Metab2019; 10:2042018819890527
  205. Zambrowicz B, Ogbaa I, Frazier K, Banks P, Turnage A, Freiman J, Boehm KA, Ruff D, Powell D, Sands A. Effects of LX4211, a dual sodium-dependent glucose cotransporters 1 and 2 inhibitor, on postprandial glucose, insulin, glucagon-like peptide 1, and peptide tyrosine tyrosine in a dose-timing study in healthy subjects. Clin Ther 2013; 35:1162-1173 e1168
  206. Powell DR, Zambrowicz B, Morrow L, Beysen C, Hompesch M, Turner S, Hellerstein M, Banks P, Strumph P, Lapuerta P. Sotagliflozin Decreases Postprandial Glucose and Insulin Concentrations by Delaying Intestinal Glucose Absorption. J Clin Endocrinol Metab 2020; 105
  207. Buse JB, Garg SK, Rosenstock J, Bailey TS, Banks P, Bode BW, Danne T, Kushner JA, Lane WS, Lapuerta P, McGuire DK, Peters AL, Reed J, Sawhney S, Strumph P. Sotagliflozin in Combination With Optimized Insulin Therapy in Adults With Type 1 Diabetes: The North American inTandem1 Study. Diabetes Care 2018; 41:1970-1980
  208. Danne T, Cariou B, Banks P, Brandle M, Brath H, Franek E, Kushner JA, Lapuerta P, McGuire DK, Peters AL, Sawhney S, Strumph P. HbA1c and Hypoglycemia Reductions at 24 and 52 Weeks With Sotagliflozin in Combination With Insulin in Adults With Type 1 Diabetes: The European inTandem2 Study. Diabetes Care 2018; 41:1981-1990
  209. Garg SK, Henry RR, Banks P, Buse JB, Davies MJ, Fulcher GR, Pozzilli P, Gesty-Palmer D, Lapuerta P, Simo R, Danne T, McGuire DK, Kushner JA, Peters A, Strumph P. Effects of Sotagliflozin Added to Insulin in Patients with Type 1 Diabetes. N Engl J Med 2017; 377:2337-2348
  210. Rosenstock J, Cefalu WT, Lapuerta P, Zambrowicz B, Ogbaa I, Banks P, Sands A. Greater dose-ranging effects on A1C levels than on glucosuria with LX4211, a dual inhibitor of SGLT1 and SGLT2, in patients with type 2 diabetes on metformin monotherapy. Diabetes Care 2015; 38:431-438
  211. Zambrowicz B, Lapuerta P, Strumph P, Banks P, Wilson A, Ogbaa I, Sands A, Powell D. LX4211 therapy reduces postprandial glucose levels in patients with type 2 diabetes mellitus and renal impairment despite low urinary glucose excretion. Clin Ther 2015; 37:71-82 e12
  212. Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Voors AA, Metra M, Lund LH, Komajda M, Testani JM, Wilcox CS, Ponikowski P, Lopes RD, Verma S, Lapuerta P, Pitt B. Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N Engl J Med 2021; 384:117-128
  213. Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Inzucchi SE, Kosiborod MN, Cherney DZI, Dwyer JP, Scirica BM, Bailey CJ, Diaz R, Ray KK, Udell JA, Lopes RD, Lapuerta P, Steg PG. Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N Engl J Med 2021; 384:129-139
  214. van Raalte DH, Bjornstad P, Persson F, Powell DR, de Cassia Castro R, Wang PS, Liu M, Heerspink HJL, Cherney D. The Impact of Sotagliflozin on Renal Function, Albuminuria, Blood Pressure, and Hematocrit in Adults With Type 1 Diabetes. Diabetes Care 2019; 42:1921-1929
  215. Raskin P, Cincotta AH. Bromocriptine-QR therapy for the management of type 2 diabetes mellitus: developmental basis and therapeutic profile summary. Expert Rev Endocrinol Metab 2016; 11:113-148
  216. Lamos EM, Levitt DL, Munir KM. A review of dopamine agonist therapy in type 2 diabetes and effects on cardio-metabolic parameters. Prim Care Diabetes 2016; 10:60-65
  217. Holt RI, Barnett AH, Bailey CJ. Bromocriptine: old drug, new formulation and new indication. Diabetes Obes Metab 2010; 12:1048-1057
  218. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care 2010; 33:1503-1508
  219. Nauck MA, Meier JJ. The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol 2016; 4:525-536
  220. Nauck MA, Meier JJ. Incretin hormones: Their role in health and disease. Diabetes Obes Metab 2018; 20 Suppl 1:5-21
  221. Brubaker PL, Drucker DJ. Minireview: Glucagon-like peptides regulate cell proliferation and apoptosis in the pancreas, gut, and central nervous system. Endocrinology 2004; 145:2653-2659
  222. Sesti G, Avogaro A, Belcastro S, Bonora BM, Croci M, Daniele G, Dauriz M, Dotta F, Formichi C, Frontoni S, Invitti C, Orsi E, Picconi F, Resi V, Bonora E, Purrello F. Ten years of experience with DPP-4 inhibitors for the treatment of type 2 diabetes mellitus. Acta Diabetol 2019;
  223. Zhang X, Zhao Q. Effects of dipeptidyl peptidase-4 inhibitors on blood pressure in patients with type 2 diabetes: A systematic review and meta-analysis. J Hypertens 2016; 34:167-175
  224. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation 2017; 136:849-870
  225. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire DK, Ray KK, Leiter LA, Raz I. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013; 369:1317-1326
  226. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation 2014; 130:1579-1588
  227. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Cushman WC, Zannad F. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013; 369:1327-1335
  228. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015; 385:2067-2076
  229. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S, Van de Werf F, Peterson ED, Holman RR. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2015; 373:232-242
  230. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, McGuire DK. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults With Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA 2019; 321:69-79
  231. McGuire DK, Alexander JH, Johansen OE, Perkovic V, Rosenstock J, Cooper ME, Wanner C, Kahn SE, Toto RD, Zinman B, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, Marx N, Cahn A. Linagliptin Effects on Heart Failure and Related Outcomes in Individuals With Type 2 Diabetes Mellitus at High Cardiovascular and Renal Risk in CARMELINA. Circulation 2019; 139:351-361
  232. Men P, Li XT, Tang HL, Zhai SD. Efficacy and safety of saxagliptin in patients with type 2 diabetes: A systematic review and meta-analysis. PLoS One 2018; 13:e0197321
  233. Mosenzon O, Leibowitz G, Bhatt DL, Cahn A, Hirshberg B, Wei C, Im K, Rozenberg A, Yanuv I, Stahre C, Ray KK, Iqbal N, Braunwald E, Scirica BM, Raz I. Effect of Saxagliptin on Renal Outcomes in the SAVOR-TIMI 53 Trial. Diabetes Care 2017; 40:69-76
  234. Cornel JH, Bakris GL, Stevens SR, Alvarsson M, Bax WA, Chuang LM, Engel SS, Lopes RD, McGuire DK, Riefflin A, Rodbard HW, Sinay I, Tankova T, Wainstein J, Peterson ED, Holman RR. Effect of Sitagliptin on Kidney Function and Respective Cardiovascular Outcomes in Type 2 Diabetes: Outcomes From TECOS. Diabetes Care 2016; 39:2304-2310
  235. Roshanov PS, Dennis BB. Incretin-based therapies are associated with acute pancreatitis: Meta-analysis of large randomized controlled trials. Diabetes Res Clin Pract 2015; 110:e13-17
  236. Tkac I, Raz I. Combined Analysis of Three Large Interventional Trials With Gliptins Indicates Increased Incidence of Acute Pancreatitis in Patients With Type 2 Diabetes. Diabetes Care 2017; 40:284-286
  237. Banks PA, Bollen TL, Dervenis C, Gooszen HG, Johnson CD, Sarr MG, Tsiotos GG, Vege SS. Classification of acute pancreatitis--2012: revision of the Atlanta classification and definitions by international consensus. Gut2013; 62:102-111
  238. Saito T, Ohnuma K, Suzuki H, Dang NH, Hatano R, Ninomiya H, Morimoto C. Polyarthropathy in type 2 diabetes patients treated with DPP4 inhibitors. Diabetes Res Clin Pract 2013; 102:e8-e12
  239. Gentilella R, Pechtner V, Corcos A, Consoli A. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev 2019; 35:e3070
  240. Frias JP, Bonora E, Nevarez Ruiz L, Li YG, Yu Z, Milicevic Z, Malik R, Bethel MA, Cox DA. Efficacy and Safety of Dulaglutide 3.0 mg and 4.5 mg Versus Dulaglutide 1.5 mg in Metformin-Treated Patients With Type 2 Diabetes in a Randomized Controlled Trial (AWARD-11). Diabetes Care 2021; 44:765-773
  241. Jones AG, McDonald TJ, Shields BM, Hill AV, Hyde CJ, Knight BA, Hattersley AT. Markers of beta-Cell Failure Predict Poor Glycemic Response to GLP-1 Receptor Agonist Therapy in Type 2 Diabetes. Diabetes Care 2016; 39:250-257
  242. Wysham CH, Lin J, Kuritzky L. Safety and efficacy of a glucagon-like peptide-1 receptor agonist added to basal insulin therapy versus basal insulin with or without a rapid-acting insulin in patients with type 2 diabetes: results of a meta-analysis. Postgrad Med 2017; 129:436-445
  243. Abd El Aziz MS, Kahle M, Meier JJ, Nauck MA. A meta-analysis comparing clinical effects of short- or long-acting GLP-1 receptor agonists versus insulin treatment from head-to-head studies in type 2 diabetic patients. Diabetes Obes Metab 2017; 19:216-227
  244. Castellana M, Cignarelli A, Brescia F, Perrini S, Natalicchio A, Laviola L, Giorgino F. Efficacy and safety of GLP-1 receptor agonists as add-on to SGLT2 inhibitors in type 2 diabetes mellitus: A meta-analysis. Sci Rep 2019; 9:19351
  245. Tchang BG, Kumar RB, Aronne LJ. Pharmacologic Treatment of Overweight and Obesity in Adults. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Grossman A, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  246. Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, Andreasen AH, Jensen CB, DeFronzo RA. Efficacy of Liraglutide for Weight Loss Among Patients With Type 2 Diabetes: The SCALE Diabetes Randomized Clinical Trial. JAMA 2015; 314:687-699
  247. Davies M, Faerch L, Jeppesen OK, Pakseresht A, Pedersen SD, Perreault L, Rosenstock J, Shimomura I, Viljoen A, Wadden TA, Lingvay I. Semaglutide 2.4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial. Lancet2021; 397:971-984
  248. Frias JP, Auerbach P, Bajaj HS, Fukushima Y, Lingvay I, Macura S, Sondergaard AL, Tankova TI, Tentolouris N, Buse JB. Efficacy and safety of once-weekly semaglutide 2.0 mg versus 1.0 mg in patients with type 2 diabetes (SUSTAIN FORTE): a double-blind, randomised, phase 3B trial. Lancet Diabetes Endocrinol 2021; 9:563-574
  249. Lorenz M, Lawson F, Owens D, Raccah D, Roy-Duval C, Lehmann A, Perfetti R, Blonde L. Differential effects of glucagon-like peptide-1 receptor agonists on heart rate. Cardiovasc Diabetol 2017; 16:6
  250. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med 2015; 373:2247-2257
  251. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2016; 375:311-322
  252. Mann JFE, Fonseca V, Mosenzon O, Raz I, Goldman B, Idorn T, von Scholten BJ, Poulter NR. Effects of Liraglutide Versus Placebo on Cardiovascular Events in Patients With Type 2 Diabetes Mellitus and Chronic Kidney Disease. Circulation 2018; 138:2908-2918
  253. Verma S, Poulter NR, Bhatt DL, Bain SC, Buse JB, Leiter LA, Nauck MA, Pratley RE, Zinman B, Orsted DD, Monk Fries T, Rasmussen S, Marso SP. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018; 138:2884-2894
  254. Marso SP, Baeres FMM, Bain SC, Goldman B, Husain M, Nauck MA, Poulter NR, Pratley RE, Thomsen AB, Buse JB. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Diabetes With or Without Heart Failure. J Am Coll Cardiol 2020; 75:1128-1141
  255. Dhatariya K, Bain SC, Buse JB, Simpson R, Tarnow L, Kaltoft MS, Stellfeld M, Tornoe K, Pratley RE. The Impact of Liraglutide on Diabetes-Related Foot Ulceration and Associated Complications in Patients With Type 2 Diabetes at High Risk for Cardiovascular Events: Results From the LEADER Trial. Diabetes Care 2018; 41:2229-2235
  256. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2016; 375:1834-1844
  257. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:1228-1239
  258. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018; 392:1519-1529
  259. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019;
  260. Gerstein HC, Hart R, Colhoun HM, Diaz R, Lakshmanan M, Botros FT, Probstfield J, Riddle MC, Ryden L, Atisso CM, Dyal L, Hall S, Avezum A, Basile J, Conget I, Cushman WC, Hancu N, Hanefeld M, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Munoz EGC, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol 2020; 8:106-114
  261. Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019; 7:776-785
  262. Sposito AC, Berwanger O, de Carvalho LSF, Saraiva JFK. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018; 17:157
  263. Lam CSP, Ramasundarahettige C, Branch KRH, Sattar N, Rosenstock J, Pratley R, Del Prato S, Lopes RD, Niemoeller E, Khurmi NS, Baek S, Gerstein HC. Efpeglenatide and Clinical Outcomes With and Without Concomitant Sodium-Glucose Cotransporter-2 Inhibition Use in Type 2 Diabetes: Exploratory Analysis of the AMPLITUDE-O Trial. Circulation 2022; 145:565-574
  264. Margulies KB, Hernandez AF, Redfield MM, Givertz MM, Oliveira GH, Cole R, Mann DL, Whellan DJ, Kiernan MS, Felker GM, McNulty SE, Anstrom KJ, Shah MR, Braunwald E, Cappola TP. Effects of Liraglutide on Clinical Stability Among Patients With Advanced Heart Failure and Reduced Ejection Fraction: A Randomized Clinical Trial. JAMA 2016; 316:500-508
  265. Jorsal A, Kistorp C, Holmager P, Tougaard RS, Nielsen R, Hanselmann A, Nilsson B, Moller JE, Hjort J, Rasmussen J, Boesgaard TW, Schou M, Videbaek L, Gustafsson I, Flyvbjerg A, Wiggers H, Tarnow L. Effect of liraglutide, a glucagon-like peptide-1 analogue, on left ventricular function in stable chronic heart failure patients with and without diabetes (LIVE)-a multicentre, double-blind, randomised, placebo-controlled trial. Eur J Heart Fail 2017; 19:69-77
  266. Fudim M, White J, Pagidipati NJ, Lokhnygina Y, Wainstein J, Murin J, Iqbal N, Ohman P, Lopes RD, Reicher B, Holman RR, Hernandez AF, Mentz RJ. Effect of Once-Weekly Exenatide in Patients With Type 2 Diabetes Mellitus With and Without Heart Failure and Heart Failure-Related Outcomes: Insights From the EXSCEL Trial. Circulation 2019; 140:1613-1622
  267. Husain M, Bain SC, Jeppesen OK, Lingvay I, Sorrig R, Treppendahl MB, Vilsboll T. Semaglutide (SUSTAIN and PIONEER) reduces cardiovascular events in type 2 diabetes across varying cardiovascular risk. Diabetes Obes Metab 2020; 22:442-451
  268. Muskiet MHA, Tonneijck L, Huang Y, Liu M, Saremi A, Heerspink HJL, van Raalte DH. Lixisenatide and renal outcomes in patients with type 2 diabetes and acute coronary syndrome: an exploratory analysis of the ELIXA randomised, placebo-controlled trial. Lancet Diabetes Endocrinol 2018; 6:859-869
  269. Mann JFE, Orsted DD, Brown-Frandsen K, Marso SP, Poulter NR, Rasmussen S, Tornoe K, Zinman B, Buse JB. Liraglutide and Renal Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:839-848
  270. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Botros FT, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and renal outcomes in type 2 diabetes: an exploratory analysis of the REWIND randomised, placebo-controlled trial. Lancet 2019;
  271. Tuttle KR, Lakshmanan MC, Rayner B, Busch RS, Zimmermann AG, Woodward DB, Botros FT. Dulaglutide versus insulin glargine in patients with type 2 diabetes and moderate-to-severe chronic kidney disease (AWARD-7): a multicentre, open-label, randomised trial. Lancet Diabetes Endocrinol 2018; 6:605-617
  272. Shaman AM, Bain SC, Bakris GL, Buse JB, Idorn T, Mahaffey KW, Mann JFE, Nauck MA, Rasmussen S, Rossing P, Wolthers B, Zinman B, Perkovic V. Effect of the Glucagon-Like Peptide-1 Receptor Agonists Semaglutide and Liraglutide on Kidney Outcomes in Patients With Type 2 Diabetes: Pooled Analysis of SUSTAIN 6 and LEADER. Circulation 2022; 145:575-585
  273. Gluud LL, Knop FK, Vilsboll T. Effects of lixisenatide on elevated liver transaminases: systematic review with individual patient data meta-analysis of randomised controlled trials on patients with type 2 diabetes. BMJ Open2014; 4:e005325
  274. Armstrong MJ, Houlihan DD, Rowe IA, Clausen WH, Elbrond B, Gough SC, Tomlinson JW, Newsome PN. Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: individual patient data meta-analysis of the LEAD program. Aliment Pharmacol Ther 2013; 37:234-242
  275. Shao N, Kuang HY, Hao M, Gao XY, Lin WJ, Zou W. Benefits of exenatide on obesity and non-alcoholic fatty liver disease with elevated liver enzymes in patients with type 2 diabetes. Diabetes Metab Res Rev 2014; 30:521-529
  276. Yan J, Yao B, Kuang H, Yang X, Huang Q, Hong T, Li Y, Dou J, Yang W, Qin G, Yuan H, Xiao X, Luo S, Shan Z, Deng H, Tan Y, Xu F, Xu W, Zeng L, Kang Z, Weng J. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: The Effect on Body Weight and Intrahepatic Lipid in Patients With Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology 2019; 69:2414-2426
  277. Bouchi R, Nakano Y, Fukuda T, Takeuchi T, Murakami M, Minami I, Izumiyama H, Hashimoto K, Yoshimoto T, Ogawa Y. Reduction of visceral fat by liraglutide is associated with ameliorations of hepatic steatosis, albuminuria, and micro-inflammation in type 2 diabetic patients with insulin treatment: a randomized control trial. Endocr J 2017; 64:269-281
  278. Kuchay MS, Krishan S, Mishra SK, Choudhary NS, Singh MK, Wasir JS, Kaur P, Gill HK, Bano T, Farooqui KJ, Mithal A. Effect of dulaglutide on liver fat in patients with type 2 diabetes and NAFLD: randomised controlled trial (D-LIFT trial). Diabetologia 2020; 63:2434-2445
  279. Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, Hazlehurst JM, Guo K, team Lt, Abouda G, Aldersley MA, Stocken D, Gough SC, Tomlinson JW, Brown RM, Hubscher SG, Newsome PN. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016; 387:679-690
  280. Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, Sanyal AJ, Sejling AS, Harrison SA. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med 2021; 384:1113-1124
  281. Faillie JL, Yu OH, Yin H, Hillaire-Buys D, Barkun A, Azoulay L. Association of Bile Duct and Gallbladder Diseases With the Use of Incretin-Based Drugs in Patients With Type 2 Diabetes Mellitus. JAMA Intern Med2016; 176:1474-1481
  282. Monami M, Nreu B, Scatena A, Cresci B, Andreozzi F, Sesti G, Mannucci E. Safety issues with glucagon-like peptide-1 receptor agonists (pancreatitis, pancreatic cancer and cholelithiasis): Data from randomized controlled trials. Diabetes Obes Metab 2017; 19:1233-1241
  283. He L, Wang J, Ping F, Yang N, Huang J, Li Y, Xu L, Li W, Zhang H. Association of Glucagon-Like Peptide-1 Receptor Agonist Use With Risk of Gallbladder and Biliary Diseases: A Systematic Review and Meta-analysis of Randomized Clinical Trials. JAMA Intern Med 2022; 182:513-519
  284. Nauck MA, Muus Ghorbani ML, Kreiner E, Saevereid HA, Buse JB, La Macchia O. Effects of Liraglutide Compared With Placebo on Events of Acute Gallbladder or Biliary Disease in Patients With Type 2 Diabetes at High Risk for Cardiovascular Events in the LEADER Randomized Trial. Diabetes Care 2019; 42:1912-1920
  285. Bethel MA, Patel RA, Merrill P, Lokhnygina Y, Buse JB, Mentz RJ, Pagidipati NJ, Chan JC, Gustavson SM, Iqbal N, Maggioni AP, Ohman P, Poulter NR, Ramachandran A, Zinman B, Hernandez AF, Holman RR. Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a meta-analysis. Lancet Diabetes Endocrinol 2018; 6:105-113
  286. Steinberg WM, Buse JB, Ghorbani MLM, Orsted DD, Nauck MA. Amylase, Lipase, and Acute Pancreatitis in People With Type 2 Diabetes Treated With Liraglutide: Results From the LEADER Randomized Trial. Diabetes Care 2017; 40:966-972
  287. Liu Y, Tian Q, Yang J, Wang H, Hong T. No pancreatic safety concern following glucagon-like peptide-1 receptor agonist therapies: A pooled analysis of cardiovascular outcome trials. Diabetes Metab Res Rev 2018; 34:e3061
  288. Singh AK, Gangopadhyay KK, Singh R. Risk of acute pancreatitis with incretin-based therapy: a systematic review and updated meta-analysis of cardiovascular outcomes trials. Expert Rev Clin Pharmacol 2020; 13:461-468
  289. Vilsboll T, Bain SC, Leiter LA, Lingvay I, Matthews D, Simo R, Helmark IC, Wijayasinghe N, Larsen M. Semaglutide, reduction in glycated haemoglobin and the risk of diabetic retinopathy. Diabetes Obes Metab2018; 20:889-897
  290. Bethel MA, Diaz R, Castellana N, Bhattacharya I, Gerstein HC, Lakshmanan MC. HbA1c Change and Diabetic Retinopathy During GLP-1 Receptor Agonist Cardiovascular Outcome Trials: A Meta-analysis and Meta-regression. Diabetes Care 2021; 44:290-296
  291. Avgerinos I, Karagiannis T, Malandris K, Liakos A, Mainou M, Bekiari E, Matthews DR, Tsapas A. Glucagon-like peptide-1 receptor agonists and microvascular outcomes in type 2 diabetes: A systematic review and meta-analysis. Diabetes Obes Metab 2019; 21:188-193
  292. Hedrington MS, Davis SN. Oral semaglutide for the treatment of type 2 diabetes. Expert Opin Pharmacother2019; 20:133-141
  293. Avgerinos I, Michailidis T, Liakos A, Karagiannis T, Matthews DR, Tsapas A, Bekiari E. Oral semaglutide for type 2 diabetes: A systematic review and meta-analysis. Diabetes Obes Metab 2019;
  294. Aroda VR, Rosenstock J, Terauchi Y, Altuntas Y, Lalic NM, Morales Villegas EC, Jeppesen OK, Christiansen E, Hertz CL, Haluzik M. PIONEER 1: Randomized Clinical Trial of the Efficacy and Safety of Oral Semaglutide Monotherapy in Comparison With Placebo in Patients With Type 2 Diabetes. Diabetes Care 2019; 42:1724-1732
  295. Rosenstock J, Allison D, Birkenfeld AL, Blicher TM, Deenadayalan S, Jacobsen JB, Serusclat P, Violante R, Watada H, Davies M. Effect of Additional Oral Semaglutide vs Sitagliptin on Glycated Hemoglobin in Adults With Type 2 Diabetes Uncontrolled With Metformin Alone or With Sulfonylurea: The PIONEER 3 Randomized Clinical Trial. JAMA 2019; 321:1466-1480
  296. Pieber TR, Bode B, Mertens A, Cho YM, Christiansen E, Hertz CL, Wallenstein SOR, Buse JB. Efficacy and safety of oral semaglutide with flexible dose adjustment versus sitagliptin in type 2 diabetes (PIONEER 7): a multicentre, open-label, randomised, phase 3a trial. Lancet Diabetes Endocrinol 2019; 7:528-539
  297. Rodbard HW, Rosenstock J, Canani LH, Deerochanawong C, Gumprecht J, Lindberg SO, Lingvay I, Sondergaard AL, Treppendahl MB, Montanya E. Oral Semaglutide Versus Empagliflozin in Patients With Type 2 Diabetes Uncontrolled on Metformin: The PIONEER 2 Trial. Diabetes Care 2019; 42:2272-2281
  298. Pratley R, Amod A, Hoff ST, Kadowaki T, Lingvay I, Nauck M, Pedersen KB, Saugstrup T, Meier JJ. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet 2019; 394:39-50
  299. Davies M, Pieber TR, Hartoft-Nielsen ML, Hansen OKH, Jabbour S, Rosenstock J. Effect of Oral Semaglutide Compared With Placebo and Subcutaneous Semaglutide on Glycemic Control in Patients With Type 2 Diabetes: A Randomized Clinical Trial. JAMA 2017; 318:1460-1470
  300. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2019; 381:841-851
  301. Bucheit J, Ayers J, Pamulapati L, Browning A, Sisson E. A Novel Dual Incretin Agent, Tirzepatide (LY3298176), for the Treatment of Type 2 Diabetes Mellitus and Cardiometabolic Health. J Cardiovasc Pharmacol 2022; 80:171-179
  302. Syed YY. Tirzepatide: First Approval. Drugs 2022;
  303. Nauck MA, Quast DR, Wefers J, Pfeiffer AFH. The evolving story of incretins (GIP and GLP-1) in metabolic and cardiovascular disease: A pathophysiological update. Diabetes Obes Metab 2021; 23 Suppl 3:5-29
  304. Heise T, Mari A, DeVries JH, Urva S, Li J, Pratt EJ, Coskun T, Thomas MK, Mather KJ, Haupt A, Milicevic Z. Effects of subcutaneous tirzepatide versus placebo or semaglutide on pancreatic islet function and insulin sensitivity in adults with type 2 diabetes: a multicentre, randomised, double-blind, parallel-arm, phase 1 clinical trial. Lancet Diabetes Endocrinol 2022; 10:418-429
  305. Thomas MK, Nikooienejad A, Bray R, Cui X, Wilson J, Duffin K, Milicevic Z, Haupt A, Robins DA. Dual GIP and GLP-1 Receptor Agonist Tirzepatide Improves Beta-cell Function and Insulin Sensitivity in Type 2 Diabetes. J Clin Endocrinol Metab 2021; 106:388-396
  306. Rosenstock J, Wysham C, Frias JP, Kaneko S, Lee CJ, Fernandez Lando L, Mao H, Cui X, Karanikas CA, Thieu VT. Efficacy and safety of a novel dual GIP and GLP-1 receptor agonist tirzepatide in patients with type 2 diabetes (SURPASS-1): a double-blind, randomised, phase 3 trial. Lancet 2021; 398:143-155
  307. Frias JP, Davies MJ, Rosenstock J, Perez Manghi FC, Fernandez Lando L, Bergman BK, Liu B, Cui X, Brown K, SURPASS Investigators. Tirzepatide versus Semaglutide Once Weekly in Patients with Type 2 Diabetes. N Engl J Med 2021; 385:503-515
  308. Ludvik B, Giorgino F, Jodar E, Frias JP, Fernandez Lando L, Brown K, Bray R, Rodriguez A. Once-weekly tirzepatide versus once-daily insulin degludec as add-on to metformin with or without SGLT2 inhibitors in patients with type 2 diabetes (SURPASS-3): a randomised, open-label, parallel-group, phase 3 trial. Lancet2021; 398:583-598
  309. Del Prato S, Kahn SE, Pavo I, Weerakkody GJ, Yang Z, Doupis J, Aizenberg D, Wynne AG, Riesmeyer JS, Heine RJ, Wiese RJ, SURPASS Investigators. Tirzepatide versus insulin glargine in type 2 diabetes and increased cardiovascular risk (SURPASS-4): a randomised, open-label, parallel-group, multicentre, phase 3 trial. Lancet 2021; 398:1811-1824
  310. Dahl D, Onishi Y, Norwood P, Huh R, Bray R, Patel H, Rodriguez A. Effect of Subcutaneous Tirzepatide vs Placebo Added to Titrated Insulin Glargine on Glycemic Control in Patients With Type 2 Diabetes: The SURPASS-5 Randomized Clinical Trial. JAMA 2022; 327:534-545
  311. Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C, Urva S, Gimeno RE, Milicevic Z, Robins D, Haupt A. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 2018; 392:2180-2193
  312. Wilson JM, Nikooienejad A, Robins DA, Roell WC, Riesmeyer JS, Haupt A, Duffin KL, Taskinen MR, Ruotolo G. The dual glucose-dependent insulinotropic peptide and glucagon-like peptide-1 receptor agonist, tirzepatide, improves lipoprotein biomarkers associated with insulin resistance and cardiovascular risk in patients with type 2 diabetes. Diabetes Obes Metab 2020; 22:2451-2459
  313. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2020.
  314. Sattar N, McGuire DK, Pavo I, Weerakkody GJ, Nishiyama H, Wiese RJ, Zoungas S. Tirzepatide cardiovascular event risk assessment: a pre-specified meta-analysis. Nat Med 2022; 28:591-598
  315. Gastaldelli A, Cusi K, Fernandez Lando L, Bray R, Brouwers B, Rodriguez A. Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): a substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol2022; 10:393-406
  316. Hartman ML, Sanyal AJ, Loomba R, Wilson JM, Nikooienejad A, Bray R, Karanikas CA, Duffin KL, Robins DA, Haupt A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care 2020; 43:1352-1355
  317. Perreault L, Rodbard H, Valentine V, Johnson E. Optimizing Fixed-Ratio Combination Therapy in Type 2 Diabetes. Adv Ther 2019; 36:265-277
  318. Billings LK, Doshi A, Gouet D, Oviedo A, Rodbard HW, Tentolouris N, Gron R, Halladin N, Jodar E. Efficacy and Safety of IDegLira Versus Basal-Bolus Insulin Therapy in Patients With Type 2 Diabetes Uncontrolled on Metformin and Basal Insulin: The DUAL VII Randomized Clinical Trial. Diabetes Care 2018; 41:1009-1016
  319. Cai X, Gao X, Yang W, Ji L. Comparison between insulin degludec/liraglutide treatment and insulin glargine/lixisenatide treatment in type 2 diabetes: a systematic review and meta-analysis. Expert Opin Pharmacother 2017; 18:1789-1798
  320. Rosenstock J, Aronson R, Grunberger G, Hanefeld M, Piatti P, Serusclat P, Cheng X, Zhou T, Niemoeller E, Souhami E, Davies M. Benefits of LixiLan, a Titratable Fixed-Ratio Combination of Insulin Glargine Plus Lixisenatide, Versus Insulin Glargine and Lixisenatide Monocomponents in Type 2 Diabetes Inadequately Controlled on Oral Agents: The LixiLan-O Randomized Trial. Diabetes Care 2016; 39:2026-2035
  321. Gough SC, Bode B, Woo V, Rodbard HW, Linjawi S, Poulsen P, Damgaard LH, Buse JB. Efficacy and safety of a fixed-ratio combination of insulin degludec and liraglutide (IDegLira) compared with its components given alone: results of a phase 3, open-label, randomised, 26-week, treat-to-target trial in insulin-naive patients with type 2 diabetes. Lancet Diabetes Endocrinol 2014; 2:885-893
  322. Younk LM, Davis SN. Evaluation of colesevelam hydrochloride for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol 2012; 8:515-525
  323. Garg A, Grundy SM. Cholestyramine therapy for dyslipidemia in non-insulin-dependent diabetes mellitus. A short-term, double-blind, crossover trial. Ann Intern Med 1994; 121:416-422
  324. Prawitt J, Caron S, Staels B. Glucose-lowering effects of intestinal bile acid sequestration through enhancement of splanchnic glucose utilization. Trends Endocrinol Metab 2014; 25:235-244
  325. Hansen M, Sonne DP, Knop FK. Bile acid sequestrants: glucose-lowering mechanisms and efficacy in type 2 diabetes. Curr Diab Rep 2014; 14:482
  326. Sonne DP, Hansen M, Knop FK. Bile acid sequestrants in type 2 diabetes: potential effects on GLP1 secretion. Eur J Endocrinol 2014; 171:R47-65
  327. Beysen C, Murphy EJ, Deines K, Chan M, Tsang E, Glass A, Turner SM, Protasio J, Riiff T, Hellerstein MK. Effect of bile acid sequestrants on glucose metabolism, hepatic de novo lipogenesis, and cholesterol and bile acid kinetics in type 2 diabetes: a randomised controlled study. Diabetologia 2012; 55:432-442
  328. Shapiro H, Kolodziejczyk AA, Halstuch D, Elinav E. Bile acids in glucose metabolism in health and disease. J Exp Med 2018; 215:383-396
  329. Potthoff MJ, Potts A, He T, Duarte JA, Taussig R, Mangelsdorf DJ, Kliewer SA, Burgess SC. Colesevelam suppresses hepatic glycogenolysis by TGR5-mediated induction of GLP-1 action in DIO mice. Am J Physiol Gastrointest Liver Physiol 2013; 304:G371-380
  330. Harach T, Pols TW, Nomura M, Maida A, Watanabe M, Auwerx J, Schoonjans K. TGR5 potentiates GLP-1 secretion in response to anionic exchange resins. Sci Rep 2012; 2:430
  331. Trabelsi MS, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton CA, Sebti Y, Kluza J, Briand O, Dehondt H, Vallez E, Dorchies E, Baud G, Spinelli V, Hennuyer N, Caron S, Bantubungi K, Caiazzo R, Reimann F, Marchetti P, Lefebvre P, Backhed F, Gribble FM, Schoonjans K, Pattou F, Tailleux A, Staels B, Lestavel S. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat Commun 2015; 6:7629
  332. Ooi CP, Loke SC. Colesevelam for type 2 diabetes mellitus. Cochrane Database Syst Rev 2012; 12:CD009361
  333. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  334. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  335. The Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA 1984; 251:365-374
  336. Younk LM, Mikeladze M, Davis SN. Pramlintide and the treatment of diabetes: a review of the data since its introduction. Expert Opin Pharmacother 2011; 12:1439-1451
  337. Singh-Franco D, Perez A, Harrington C. The effect of pramlintide acetate on glycemic control and weight in patients with type 2 diabetes mellitus and in obese patients without diabetes: a systematic review and meta-analysis. Diabetes Obes Metab 2011; 13:169-180
  338. Riddle M, Pencek R, Charenkavanich S, Lutz K, Wilhelm K, Porter L. Randomized comparison of pramlintide or mealtime insulin added to basal insulin treatment for patients with type 2 diabetes. Diabetes Care 2009; 32:1577-1582
  339. Chapman I, Parker B, Doran S, Feinle-Bisset C, Wishart J, Strobel S, Wang Y, Burns C, Lush C, Weyer C, Horowitz M. Effect of pramlintide on satiety and food intake in obese subjects and subjects with type 2 diabetes. Diabetologia 2005; 48:838-848

 

46,XY Differences of Sexual Development

ABSTRACT

 

The 46,XY differences of sex development (46,XY DSD) can result either from decreased synthesis of testosterone and/or DHT or from impairment of androgen action. 46,XY DSD are characterized by micropenis, atypical or female external genitalia, caused by incomplete intrauterine masculinization with or without the presence of Müllerian structures. Male gonads are identified in the majority of 46,XY DSD patients, but in some of them no gonadal tissue is found. Complete absence of virilization results in normal female external genitalia and these patients generally seek medical attention at pubertal age, due to the absence of breast development and/or primary amenorrhea. A careful clinical evaluation of the neonate is essential because most DSD patients could be recognized in this period and prompt diagnosis allows a better therapeutic approach. Family and prenatal history, complete physical examination and assessment of genital anatomy are the first steps for a correct diagnosis. The diagnostic evaluation of DSD includes hormone measurements (assessment of Leydig and Sertoli cell function), imaging (ultrasonography is always the first and often the most valuable imaging modality in DSD patients’ investigation), cytogenetic, and molecular studies. Endoscopic and laparoscopic exploitation and/or gonadal biopsy are required in very few cases. Psychological evaluation is of crucial importance to treat DSD patients. Every couple that has a child with atypical genitalia must be assessed and receive counseling by an experienced psychologist, specialized in gender identity, who must act as soon as the diagnosis is suspected, and then follow the family periodically, more frequently during the periods before and after genitoplasty. Parents must be well informed by the physician and psychologist about normal sexual development. A simple, detailed, and comprehensive explanation about what to expect regarding integration in social life, sexual activity, need of hormonal and surgical treatment and the likely possibility or not of fertility according to the sex of rearing, should also be discussed with the parents before the assignment of final social sex. Optimal care of DSD patients begins in the newborn period and sometimes in prenatal life and requires a multidisciplinary team. Most of the well-treated DSD patients present a normal quality of life in adulthood.

 

INTRODUCTION

 

Male phenotypic development is a 2-step process: 1) testis formation from the primitive gonad (sexual determination) and 2) internal and external genitalia differentiation by action of factors secreted by the fetal testis (sexual differentiation). The first step is very complex and involves interplay of several transcription factors and signaling cells (1-3). Dosage imbalances in genes involved in DSD (deletions or duplication) have also been identified as a cause of these developmental differences (Fig. 1).

Figure 1. Summary of the molecular events in sex determination indicating the genes in which molecular defects can cause gonadal disorders in animal models. Some of these disorders were confirmed in humans. Nr5a1, Wnt4 and Wt1 are expressed in the urogenital ridge whose development results in formation of the gonads, kidneys, and adrenal cortex. Several genes, Wt1, Nr5a1, M33 (CBX2 mouse homologue), Lhx9, Lim1, Gata4/Fog2, Dmrt1, Emx2 and Cited are expressed in the bipotential gonad. Nr5a1 up-regulates Cbx2 expression that is required for upregulation of the Sry gene. Nr5a1 and Wt1 up-regulate Sry expression in pre-Sertoli cells and Sry initiates male gonad development. Sry strongly up-regulates Sox9 in Sertoli cells. Sox9 up-regulates Fgf9 and Fgf9 maintains Sox9 expression, forming a positive feed-forward loop in XY gonads. The balance between Fgf9 and Rspo1/Wnt4 signals is shifted in favor of Fgf9, establishing the male pathway. If Wnt4/Rspo1 is overexpressed activating the β-catenin pathway, this system blocks Fgf9 and disrupts the feed-forward loop between Sox9 and Fgf9. Pdg2 signaling up-regulates Sox9 and Sox9 activate Ptgds. Sox9 establishes a feed-forward loop with the Pgd2. Sox9 inhibits beta-catenin-mediated Wnt signaling. Overexpression in either Dax1 (locus DSS) or Rspo1/Wnt4 antagonizes testis formation. On the other hand, Dax1 regulates the development of peritubular myoid cells and the formation of testicular cords. Dmrt1 has recently been shown to be required for the maintenance of gonadal sex and to prevent female reprogramming in postnatal testis, Cbx2 directly or indirectly repress ovarian development. Dhx37 has critical roles in early human testis determination and also in the maintenance of testicular tissue.

The second step, male sex differentiation, is a more straightforward process. Mesonephric (Wolffian) and paramesonephric (Mullerian) ducts are present in both, male and female fetuses, and originate from the anterolateral epithelium of the urogenital ridge. Anti Müllerian hormone (AMH) secreted by the testicular Sertoli cells acts on its receptor in the Müllerian ducts to cause their regression. Testosterone secreted by the testicular Leydig cells acts on the androgen receptor in the Wolffian ducts to induce the formation of epididymis, deferent ducts and seminal vesicles (Fig. 2) (4). The external genitalia of the fetus derive from mesenchyme cells from the primitive streak. Under androgen stimuli male fetal urethral folds, genital tubercle and genital swellings give rise to corpus spongiosum and primitive urethra, phallus, and scrotal swellings respectively. This process is mediated by testosterone and its further reduced dihydrotestosterone (DHT), which acts on the androgen receptor of the prostate and external genitalia leading to their masculinization (5,6) (Figs. 3 - 8).

Figure 2. Summary of the molecular events in sex differentiation indicating the genes in which molecular defects cause 46,XY DSD in humans. After testis determination, hormones produced by the male gonad induce the differentiation of internal and external genitalia acting on their specific receptor. The regulation of AMH gene requires cooperative interaction between SOX9 and NR5A1, WT1, GATA4 and HSP70 at the AMH promoter. Combined expression of DHH, MAMLD1 and NR5A1 is required for Leydig cell development. NR5A1 regulates gonadal steroidogenesis. The Leydig cells also produce INSL3, which causes the testes to descend to the scrotum.

Figure 3. The development of male internal genitalia in the human embryo. The 6-wk-end embryo is equipped with both male and female genital ducts derived from the mesonephrons.

Figure 4. The development of male internal genitalia in the human embryo. The regression of the Müllerian ducts is mediated by the action of AMH secreted by the fetal Sertoli cells.

Figure 5. The development of male internal genitalia in the human embryo. The stabilization and differentiation of the Wolffian ducts are mediated by testosterone synthesized by the fetal Leydig cells. The enzyme 5α-reductase 2 converts testosterone to dihydrotestosterone (DHT). The Wolffian ducts differentiate into epididymis, vas deferens and seminal vesicles. DHT contributes to prostate differentiation.

Figure 6. Development of male external genitalia in the human embryo. At the 8-wk-end embryo the external genitalia of both sexes are identical and have the capacity to differentiate in both directions: male or female. DHT stimulates growth of the genital tubercle and induces fusion of urethral folds and labioscrotal swellings. It also inhibits growth of the vesicovaginal septum, preventing development of the vagina.

Figure 7. Development of male external genitalia in the human embryo. At the 12-week-end embryo the male external genitalia are entirely formed.

Figure 8. Development of male internal and external genitalia in the human embryo. At the 12-week-end embryo both internal and external genitalia are completely formed.

The term differences of sex development (DSD) include- congenital conditions in which development of chromosomal, or gonadal or anatomical sex is atypical. This nomenclature has been proposed to replace terms such as intersex, pseudo-hermaphroditism and sex reversal   (6,7). These terms, previously used to describe the differences of sex development, are potentially offensive to the patients and the consensus on the management of intersex disorders recommends a new nomenclature that will be followed in this chapter (6). The proposed changes in terminology aim to integrate upcoming advances in molecular genetics in the most recent DSD classification (8).

 

The 46,XY DSDs are characterized by micro-penis, atypical or female external genitalia, caused by incomplete intrauterine masculinization with or without the presence of Müllerian structures. Male gonads are identified in the majority of 46,XY DSD patients, but in some of them no gonadal tissue is found. Complete absence of virilization results in normal female external genitalia and these patients generally seek medical attention at pubertal age, due to the absence of breast development and/or primary amenorrhea. 46,XY DSD can result either from decreased synthesis of testosterone or DHT or from impairment of androgen action (9,10). Our proposal classification of 46,XY DSD is displayed in Table 1.

 

Table 1. Classification of 46,XY DSD

46,XY DSD DUE TO ABNORMALITIES OF GONADAL DEVELOPMENT

   Gonadal agenesis

   Gonadal dysgenesis - complete and partial forms

46,XY DSD ASSOCIATED WITH CHOLESTEROL SYNTHESIS DEFECTS

   Smith-Lemli-Opitz syndrome

46,XY DSD DUE TO TESTOSTERONE PRODUCTION DEFECTS

   Impaired Leydig cell differentiation (LHCGR defects)

       Complete and partial forms

   Enzymatic defects in testosterone synthesis

       Defects in adrenal and testicular steroidogenesis

        STAR deficiency

        P450scc deficiency

        3-β-hydroxysteroid dehydrogenase II deficiency

       17α-hydroxylase and 17,20 lyase deficiency

        P450 oxidoreductase defect (electron transfer disruption)

   Defects in testicular steroidogenesis

       Isolated 17,20-lyase deficiency

       Cytochrome b5 defect (allosteric factor for P450c17 and POR interaction)

       17β-hydroxysteroid dehydrogenase III deficiency

   Alternative pathway to DHT

       3α- hydroxysteroid dehydrogenase deficiency due to AKR1C2 and AKR1C4 defects

46,XY DSD DUE TO DEFECTS IN TESTOSTERONE METABOLISM

     5α-reductase type 2 deficiency

46,XY DSD DUE TO DEFECTS IN ANDROGEN ACTION

   Androgen insensitivity syndrome

        Complete and partial forms

46,XY DSD DUE TO PERSISTENCE OF MÜLLERIAN DUCTS

   Defect in AMH synthesis

   Defect in AMH receptor

CONGENITAL NON-GENETIC 46,XY DSD

   Maternal intake of endocrine disruptors

   Associated with impaired prenatal growth

46,XY OVOTESTICULAR DSD

NON-CLASSIFIED FORMS

   Hypospadias

   46,XY gender dysphoria

 

INVESTIGATION OF DSD PATIENTS

 

Optimal care of patients with DSD requires a multidisciplinary team and begins in the newborn period. A careful clinical evaluation of the neonate is essential because most DSD patients could be recognized in this period and prompt diagnosis allows a better therapeutic approach. Family and prenatal history, complete physical examination and assessment of genital anatomy are the first steps for a correct diagnosis. The diagnostic evaluation of DSD includes hormone measurements, imaging, cytogenetic, and molecular studies (11). In very few cases, endoscopic and laparoscopic exploitation and/or gonadal biopsy are required (12).

 

The endocrinological evaluation of 46,XY DSD infants includes assessment of testicular function by basal measurements of LH, FSH, inhibin B, anti-Mullerian hormone (AMH), and steroids. AMH and inhibin B are useful markers of the presence of Sertoli cells and their assessment could help in the diagnosis of testis determination disorders. In boys with bilateral cryptorchidism serum AMH and inhibin B correlate with the presence of testicular tissue and undetectable values are highly suggestive of absence of testicular tissue (13,14).

 

In minipuberty and in postpubertal patients with testosterone synthesis defects, the diagnosis is made through basal steroid levels. Testosterone levels are low and steroids upstream from the enzymatic blockage are elevated. This pattern can be confirmed by an hCG stimulation test, which increases the accumulation of steroids before the enzymatic blockage, with a slight elevation of testosterone. In prepubertal individuals, an hCG stimulation test is essential for the diagnosis, since basal levels are not altered.

 

There are several hCG stimulation protocols and normative data must be established for each of them. We established a normal testosterone response 72 and 96 hours after the last of 4 doses of hCG, 50-100 U/kg body weight, given via intramuscular every 4 days in boys with cryptorchidism but otherwise normal external genitalia: testosterone peak levels reached 391 ± 129 ng/dL and we consider a subnormal response a value <130 ng/dL (equivalent to -2 SD) (15).

 

Imaging evaluation is indicated in the neonatal period when atypical genitalia are identified. If apparent female genitalia with clitoral hypertrophy, posterior labial fusion, foreshortened vulva with single opening or inguinal/labial mass is present, imaging studies may also be performed. A family history of DSD and later presentations as abnormal puberty or primary amenorrhea, cyclic hematuria in a male, and inguinal hernia in a female also require an imaging evaluation.

 

Ultrasonography is always the first and often the most valuable imaging modality in DSD patients’ investigation. Ultrasound shows the presence or absence of Müllerian structures at all ages and can locate the gonads and characterize their echo texture. This exam can also identify associated malformations such as kidney abnormalities (16).

 

Genitography and cystourethrography can display the type of urethra, the presence of vagina, cervix, and urogenital sinus. MRI contributes to accurate morphologic evaluation of Mullerian duct structures, the gonads, and the development of the phallus, all of which are essential for appropriate gender assignment and planning of surgical reconstruction (17).

 

CYTOGENETIC AND MOLECULAR INVESTIGATION

 

The routine use of genetic testing for reaching a diagnosis in XY DSD is increasingly playing an important role in the diagnostic process. A wide range of techniques may be used, each one having a different investigative application and genetic resolution (18,19).

 

More than 75 genes involved in gonadal development and/or sex hormone biosynthesis/action are known causes of DSDs and the molecular methodologies have contributed to identify already known as well as novel causes of DSD. These results have led to the adoption of molecular tests into clinical practice for diagnosis and genetic counseling, reducing the need of hormonal and imaging tests to reach the correct diagnosis (20). Advances in molecular biological techniques for diagnosing DSD are reviewed in recent publications (18,19).

 

Chromosomal Analysis

 

Early identification of chromosomal regions and candidate-genes involved in the DSD etiology

were established by finding microscopically visible structural changes in the karyotype, using conventional cytogenetic techniques. Many of them were achieved by positional cloning and linkage analysis, which are not widely used tools.

 

Although conventional karyotyping is still used frequently in routine clinical diagnosis, faster molecular cytogenetic techniques that do not require cell culture can be employed. Array techniques [array comparative genomic hybridization (aCGH) and single-nucleotide polymorphism (SNP) array] are all capable to identify submicroscopic genome imbalance / copy number variation (CNV), as small as 10 KB (CNVs between 10 kb and 5 Mb in size), and which may affect several genes, in patients with an apparently normal karyotype (21,22).

 

CNV affecting coding sequences or regulatory elements of critical dosage-sensitive genes are known causes of DSDs (23-26). Novel DSD candidate chromosomic regions and genes, with potential roles in sex determination and DSD, such as SUPT3H, C2ORF80, KANK1, ADCY2, VAMP7 and ZEB2, have been also identified by array studies, many of them waiting for further validation (25,27).

 

Array techniques can diagnose pathogenic CNV in almost 30% of syndromic DSD patients as a single method (27,28). Thus, a CGH or SNP-array was proposed as the first genomic test to investigate this group of DSD patients.

 

Sequencing Analysis

 

Among the genetic tests, many use a candidate-gene approach (Sanger sequencing), while targeted DSD gene panels, wider whole-exome (WES) and whole-genome (WGS) scale are high-throughput screening technologies, in which multiple short DNA target sequences are analyzed to identified the presence of allelic variants (29).

 

Sanger sequencing is often the method of choice if a specific genetic condition is highly suspected by an established clinical and hormonal diagnosis. AR and SRD5A2, in addition to almost all testosterone synthesis defects, are the most requested genes in 46,XY DSD to be sequenced using this approach (20).

 

The superiority of targeted DSD gene panel tests, that can evaluate simultaneously several and non-standard sets of genes, over single-gene testing approaches is well established, especially considering time and cost-effectiveness (26,30).

 

Whole-Exome Sequencing (WES) and Whole-Genome Sequencing (WGS) are also based on short-read sequencing. They present a clear improvement over single-gene testing in providing clinical diagnosis for DSD. The advantage of WES/WGS is the potential to identify new DSD-related genes in the research setting. On the other hand, WGS has more consistent coverage of gene sequences throughout the genome, including the non-coding regions and so it has the potential to provide a much higher diagnostic yield than WES (25,31).

 

Nevertheless, WES and WGS require significant bioinformatic resources and are expensive strategies; consequently, their application for first-line diagnostic investigation in many clinical settings are still limited (18,32).

 

The target DNA can also be read in longer fragments (several kb). The main advantage of using long-reads during the process is that repetitive elements and complex structural variants can often be resolved to a greater extent than in assemblies generated from short-read sequencing (18). Long-read sequencing offers a potential solution to genome-wide short tandem repeats analysis, which are highly variable elements, which play a pivotal role in the regulation of gene expression.

 

Studies in animal models have suggested the involvement of epigenetic regulation in the process of gonadal formation, reinforcing a probable role of epigenetic variation in the etiology of DSD (33).

 

Careful selection of the genetic test indicated for each condition remains important for a good clinical practice (Figure 9).

 

Figure 9. Algorithm for 46,XY DSD diagnosis

46,XY DSD DUE TO ABNORMALITIES IN GONADAL DEVELOPMENT

 

Uncountable allelic variants identified in several genes involved in the process of human gonadal determination have been associated with 46,XY gonadal dysgenesis. They will be described according to the period of gene expression in gonadal determination.

 

Gonadal Determination and Differentiation

 

The intermediate mesoderm is the primary embryonic tissue at gastrulation that gives rise to the urogenital ridge. This, in turns, is going to derive the primitive gonad from a condensation of the medioventral region of the urogenital ridge. The primitive gonad separates from the adrenal primordium at about 5 weeks but remains bipotential until the 6thweek after conception. Mammals sex determination is a complex process, which involves many genes acting in networks. Several genes have been involved in the development of the urogenital ridge, including Emx2, Lim1, Lhx9, Wt1, Gata-4/Fog2, Nr5a1/Sf1. Although knockout models of these genes produce abnormal gonads in mice, not all of them have been implicated in the human gonadal dysgenesis etiology.

 

To date, Emx2 null mice have absent kidneys, ureters, gonads and genital tracts and have developmental abnormalities of the brain (34). In humans, variants in EMX2 have been found in patients with schizencephaly (a rare condition in which a person is born with clefts in the brain that are filled with liquor) but no gonadal phenotypes have been described. WT1, NR5A1 and NR0B1/DAX1 are well known genes that are critical for the formation of the urogenital ridge in humans. The products of WT1 are essential for both gonadal and renal formation (35) whereas NR5A1/SF1 protein is essential for gonadal and adrenal formation (36,37). NR0B1/DAX1 is also essential for gonadal and adrenal differentiation and when mutated, results in congenital adrenal hypoplasia and hypogonadotropic hypogonadism (38).

 

After the formation of the bipotential gonad, by the 6th week after conception, in 46, XY individuals, the expression of the testis-determining gene Sry, which is transcriptionally regulated by the expression of Wt1 (39) and GATA Binding Protein 4  (Gata4), its cofactor the Friend-of-GATA (Fog2) (40) and chromobox protein homolog 2 (Cbx2) (41) trigger the gonadal masculinizing fate process. In the mammalian male embryo, the first molecular signal of sex determination is the expression of Sry within a subpopulation of somatic cells of the indifferent genital ridge (42). The transient expression of Sry drives the initial differentiation of pre-Sertoli cells that would otherwise follow a female pathway, becoming granulosa cells. Once Sry expression begins, it initiates the cascade of gene interactions and cellular events that direct the formation of a testis from the undifferentiated fetal gonad. So, pre-Sertoli cells proliferate, polarize and aggregate around the germ cells to define the testes cords. Migration of cells into the gonad from the mesonephros or the coelomic epithelium is subsequently induced by signals emanating from the pre-Sertoli cells. Peritubular myoid cells surround the testes cords and cooperate with pre-Sertoli cells to deposit the basal lamina and further define the testis cords. Signaling molecules produced by the pre-Sertoli cells promote the differentiation of somatic cells, found outside the cords, into fetal Leydig cells, thus ultimately allowing the production of testosterone. Endothelial cells are associated to form the coelomic vessel, which promotes efficient export of testosterone into plasma.

 

The gene Sox9 is up-regulated immediately after Sry expression and is involved in the initiation and maintenance of Sertoli cell differentiation during the early phases of testis differentiation. The mechanism by which NR5A1 and SRY increase endogenous SOX9 expression was clearly demonstrated in human embryonal carcinoma cell line NT2/D1 (43).

 

Extracellular signaling pathways (Fgf9 and Igf1r/Irr/Ir) play a significant role in Sox9 expression. A model has been suggested in that the fate of the bipotential gonad is controlled by mutually antagonistic signals between Fgf9 andWnt4/Rspo1. In this model Sox9 up-regulates Fgf9-Fgfr2 and Fgf9 maintains Sox9 expression, forming a positive feed-forward loop in XY gonads. The balance between Fgf9 and Wnt4/Rspo1 signals is shifted in favor of Fgf9, establishing the male pathway. In addition, Sry inhibits β-catenin-mediated Wnt signaling (44). In the absence of this feed-forward loop between Sox9 and Fgf9, Wnt4/Rspo1, the activated β-catenin pathway, blocks Fgf9 and promotes the ovarian fate (45,46). Furthermore, Sox9 directly binds to the promoter of the Ptgds gene which encodes prostaglandin D synthase that mediates the production of PGD2 (47) which, in turn, promotes nuclear translocation of Sox9, facilitating Sertoli cell differentiation (48). Antagonism between Dmrt1 and Foxl2 comprises another step for sex-determining decision. Dmrt1 has been described as essential to maintain mammalian testis determination, preventing female reprogramming in the postnatal mammalian testis (49). MAP3K1 has been described to be important to the balance between SOX9/FGF9 to WNT/beta-catenin signaling in functional studies (50,51). However, the role of MAP3K1 in human sex-determination remains unknown as the downstream effectors of MAP3K1 in the human developing testis have not been identified (52). Similarly, the precise mechanism by which DHX37 interferes with testis determination/maintenance remains to be elucidated (53). Abnormalities in the expression (underexpression or overexpression or timing of expression) of genes involved in the cascade of testis determination can cause anomalies of gonadal development and consequently, 46,XY DSD. The absence, regression, or the presence of dysgenetic testes results in abnormal development of the genital ducts and/or external genitalia in thosepatients.

 

46,XY Gonadal Agenesis

 

Total absence of gonadal tissue confirmed by laparoscopy has rarely been described in XY subjects with female external and internal genitalia indicating the absence of testicular determination (54). Mendonca et al described a pair of siblings, one XY and the other XX, born to a consanguineous marriage, with normal female external and internal genitalia associated with gonadal agenesis (55). Pathogenic allelic variants  in NR5A1 and LHX9 were later ruled out in these siblings (56). The origin of this disorder remains to be determined, but a defect in another gene essential for bipotential gonad development is the most likely cause of this disorder.

 

46,XY Gonadal Dysgenesis - Complete and Partial Forms

 

46,XY gonadal dysgenesis consists of a variety of clinical conditions, in which the development of the fetal gonad is abnormal and encompasses both a complete and a partial form. The complete form of gonadal dysgenesis was first described by Swyer et al. (57) and is characterized by female external and internal genitalia, lack of secondary sexual characteristics, normal or tall stature without somatic stigmata of Turner syndrome, eunuchoid habitus and the presence of bilateral dysgenetic gonads in XY subjects. Mild clitoromegaly is present in some cases.

 

The partial form of this syndrome is characterized by variable degrees of impaired testicular development and testicular function. These patients present a spectrum of atypical genitalia with or without Müllerian structures. Similar phenotypes can also result from a 45,X/46,XY karyotype.

Serum gonadotropin levels are elevated in both the complete and partial forms, mainly FSH levels, which predominate over LH levels. Testosterone levels are at the prepubertal range in the complete form. Meanwhile, in the partial form, it can range from prepubertal levels to normal adult male levels.

 

The clinical condition named embryonic testicular regression syndrome (ETRS) has been considered part of the clinical spectrum of partial 46,XY gonadal dysgenesis (58). In this syndrome, most of the patients present atypical genitalia or micropenis associated with complete regression of testicular tissue in one or both sides. Pathogenic/likely pathogenic variants in DHX37 were reported in patients with 46,XY GD at a frequency of 14%. Considering only the ETRS phenotype (micropenis and absence of uni- or bilateral testicular tissue), this frequency increases to 50% (53).The masculinization degrees of internal and external genitalia presented are related to the time and duration of the hormonal secretion, prior to cessation of testicular function. The dysgenetic testes showed disorganized seminiferous tubules and stroma with occasional primitive sex cords without germ cells (59). Familial cases of gonadal dysgenesis with variable degrees of genital atypia have been reported, and the nature of the underlying genetic defect is still unknown in several families, despite new genetic investigation methodologies available (58). Regarding the genetic etiology, 46,XY gonadal dysgenesis is heterogeneous and can result from defects of any gene involved in the process of gonadal formation.

 

The following review will focus on the main genes causing gonadal dysgenesis in humans, presenting as an isolated or syndromic phenotype.

 

Dysgenetic 46,XY DSD Due to Under Expression of GATA4 and FOG2/ ZFPM2 Genes

 

Gata4 (GATA-binding factor 4 gene) cooperatively interacts with several proteins to regulate the expression of genes involved in testis determination and differentiation as SRY, SOX9, NR5A1, AMH, DMRT1, STAR, CYP19A1, and others (60).

 

In humans, GATA4 variants were first described in patients with congenital heart defects without genital abnormalities (61). However, genitourinary anomalies, such as hypospadias and cryptorchidism, were described in 46,XY patients with deletion of the 8p23.1 region, in which GATA4 is located (62).

 

The p.G221R GATA4 pathogenic variant was identified in five members of a French family, three 46,XY DSD patients, two of them with cardiac anomalies, and in their apparently unaffected mothers (63).

 

The role of FOG2 in human testis development was corroborated by the identification of a balanced translocation (8;10) (q23.1;q21.1) in a patient with partial gonadal dysgenesis and congenital heart abnormalities (64). Bashamboo et al. identified missense FOG2 variants, using exome sequencing, in two patients with 46,XY gonadal dysgenesis. One patient carried the non-synonymous p.S402R heterozygous variant. The second patient carried the inherited homozygous p.M544I variant and the de novo heterozygous p.R260Q variant. The p.M544I variant by itself has little effect on the biological activity of FOG2 protein in transactivation of the gonadal promoters, but it shows reduced binding with GATA4. In the in vitro assays, a combination of both the p.R260Q and the p.M544I variants altered the biological activity of the FOG2 protein on specific downstream targets, as well as obliterated its interaction with GATA4. In the patient, the two variants together may result in an imbalance of the delicate equilibrium between antagonistic male and female pathways leading ultimately to gonadal dysgenesis (65). Although several GATA4 and FOG2/ZFPM2 variants have been identified in 46,XY DSD patients, the real role of the majority of them in the etiology of gonadal disease is still unclear. The re-study of seven GATA4 and ten FOG2/ZFPM2 variants previously identified by Eggers et al. (26) in a cohort of 46,XY DSD patients, using updated tools and testing their molecular activity in the context of gonadal signaling by in vitro assays, support that the majority of them are benign in their contribution to 46,XY DSD. Only one variant (p. W228C) located in the conserved N-terminal zinc finger of GATA4, was considered pathogenic, with functional analysis confirming differences in its ability to regulate Sox9 and AMH, and in protein interaction with ZFPM2 (66).

 

Dysgenetic 46,XY DSD Due to Under Expression of the CBX2 gene 

 

In humans, variants in both CBX2.1 and CBX2.2 isoforms were associated with 46,XY DSD (67,68).

 

The compound heterozygous CBX2.1 variants, c.C293T (p.P98L) and c.G1370C (p.R443P), inherited from the father and the mother respectively, was identified in a 46,XY patient, who was born with a completely normal female phenotype. The patient had uterus and histologically normal ovaries (67) and high serum FSH levels. Her phenotype resembles the Cbx2 knock-out XY mice phenotype (41). Cbx2 (M33) knockout mice present hypoplastic gonads in both sexes, but a small or absent ovaries are observed in the XY Cbx2 knockout, consequently to the reduced expression of Sry and Sox9 in the gonadal tissue (41). Functional studies demonstrated that these variants do not bind to, or adequately regulate the expression of target genes important for gonadal development, such as NR5A1 (67).

 

Mutated CBX2.2 isoforms were also implicated in the etiology of partial 46,XY gonadal dysgenesis in two other patients. Each patient carried a distinct variant, the p. C132R (c.394T>C) and the C154fs (c.460delT). These CBX2.2variants were shown to be related to a defective expression of EMX2 in the developing gonad (68). However, analysis of populational genetics data indicates that p.C154fs is present in general populations at high frequency, inconsistent with causing gonadal dysgenesis (69).

 

46,XY DSD Due to Under Expression of the WT1 Gene

 

The Wilms’ tumor suppressor gene (WT1) is located on 11p13, and encodes a zinc-finger transcription factor involved in the development and function of the kidneys and gonads. The WT1 contains 10 exons, of which exons 1–6 encode a proline/glutamine-rich transcriptional-regulation region and exons 7–10 encode the four zinc fingers of the DNA-binding domain. Four major species of RNA with conserved relative amounts, different binding specificities, and different subnuclear localizations are generated by two alternative splicing regions (70). Splicing at the first site results in either inclusion or exclusion of exon 5. The second alternative splicing site is in the 3’ end of exon 9 and allows the inclusion or exclusion of three amino acids lysine, threonine and serine between the third and fourth zinc fingers, resulting in either KTS-positive or negative isoforms. Isoforms that only differ by the presence or absence of the KTS amino acids have different affinities for DNA and, therefore, possibly different regulatory functions (70). A precise balance between WT1 isoforms is necessary for its normal function (71).

 

WT1 presents a complex network of interaction with several protein systems so that abnormalities in it can determine a wide phenotypic spectrum in XY and XX individuals (72). Several syndromes are associated with WT1 pathogenic variants, including: WAGR, Denys-Drash and Frasier syndrome.

 

WAGR syndrome is characterized by Wilms’ tumor, aniridia, genitourinary abnormalities and mental retardation. Genitourinary anomalies are frequently observed, such as renal agenesis or horseshoe kidney, urethral atresia, hypospadias, cryptorchidism and more rarely atypical genitalia (73). Heterozygous deletions of WT1 and othercontiguous genes are the cause of this syndrome (74). Deletions of PAX6 gene are related to the presence of aniridia in these patients. Severe obesity is present in a substantial proportion of  subjects with the WAGR syndrome, and the acronym WAGRO has been suggested for this condition (75). The phenotype of obesity and hyperphagia in WAGRO syndrome is attributable to deletions that determine haploinsufficiency of the BDNF gene (Brain-Derived Neurotrophic Factor) (76).

 

Le Caignec et al. described a 46,XY patient with an interstitial deletion of approximately 10 Mb located on 11p13, encompassing WT1 and PAX6 , who  presented a female external and internal genitalia, an unusual phenotype of WAGR syndrome (77). This report demonstrated an overlap of clinical and molecular features in WAGR, Frasier and Denys-Drash syndromes that confirms these conditions as a spectrum of disease due to WT1 alterations.

 

Denys-Drash syndrome is characterized by dysgenetic 46,XY DSD associated with early-onset renal failure (steroid-resistant nephrotic syndrome with diffuse mesangial sclerosis and progression to end-stage kidney disease) and Wilms´ tumor development in the first decade of life (78). Müllerian duct differentiation varies according to the Sertoli cells' function. The molecular defect of this syndrome is the presence of heterozygous missense allelic variants in the zinc finger encoding exons (DNA-binding domain) of WT1 gene (79). Gonadal development is impaired to variable degrees, resulting in a spectrum of 46,XY DSD (80).

 

Frasier syndrome is characterized by a female to atypical external genitalia phenotype in 46,XY patients, streak or dysgenetic gonads, which are at high risk for gonadoblastoma development, and renal failure (early steroid-resistant nephrotic syndrome with focal and segmental glomerular sclerosis). In these patients the progression to end-stage renal disease often occurs in adolescence, although the late-onset nephropathy has also been described in Frasier syndrome (81), reinforcing that patients carrying WT1 pathogenic variants should have the renal function carefully monitored (82).

 

Constitutional heterozygous variants of the WT1, almost all located at intron 9, are found in patients with Frasier syndrome, leading to a change in splicing that results in reversal of the normal KTS positive/negative ratio from 2:1 to 1:2  (78,83).  Exonic variants have been also associated with Frasier syndrome (84).

 

The report of atypical external genitalia (84) , the presence of Wilms’ tumor  (85), and the description of exonic variants in the DNA binding domain of WT1 gene (84) in patients with Frasier syndrome indicate an overlap of clinical and molecular features between  Denys Drash and Frasier syndromes.

 

46,XY DSD Due to Under Expression of the NR5A1/SF1 Gene

 

NR5A1 was originally identified as a master-regulator of steroidogenic enzymes in the early 1990s following the Keith L. Parker and Kenichirou Morohashi inspiring work  (86-88). NR5A1 has since been shown to control many aspects of adrenal and gonadal function (37,89), NR5A1, together with several signaling molecules, are also involved in adrenal stem cell maintenance, proliferation and differentiation inducing adrenal zonation, probably acting in the progenitor cells (90).

 

Homozygous 46,XY null mice (−/−) have adrenal agenesis, complete testicular dysgenesis, persistent Müllerian structures, partial hypogonadotropic hypogonadism, and other features such as late-onset obesity (91). Therefore, it was clear demonstrated that Nr5a1 is an essential factor in sexual and adrenal differentiation, and a key regulator of adrenal and gonadal steroidogenesis and also of the hypothalamic-pituitary-gonadal axis.

 

The first reported human case of NR5A1 pathogenic variant, the heterozygous p.G35E, was a 46,XY patient who presented female external genitalia and Müllerian duct derivatives, indicating the absence of male gonadal development, associated with adrenal insufficiency. This patient presented with salt-losing adrenal failure in early infancy and was thought to have a high block in steroidogenesis (e.g., in CYP11A1, STAR) affecting both adrenal and testicular functions. However, the identification of streak-like gonad and Müllerian structures was consistent with testicular dysgenesis, thereby, a disruption of a common developmental regulator such as NR5A1 was hypothesized. The patient was found to have a de novo heterozygous p.G35E change in the P-bo of NR5A1, which is important in dictating DNA binding specificity through its interaction with DNA response elements in the regulatory regions of target genes (92).

 

The second report of NR5A1 defects in humans was described by Biason-Lauber and Schoenle, in a 14-month-old 46,XX girl who had presented with primary adrenal insufficiency and seizures  (93) . She had a de novo heterozygous NR5A1 change resulting in the p.R255L variant into the proximal part of the ligand-like binding domain of the protein. The ovaries were detected by MRI scan and Inhibin A levels were normal for her age, suggesting that NR5A1 change had not disrupted ovarian function. The follow up of this girl until 16.5 years old showed a normal puberty and regular menstruation showing that phenotypic variant of NR5A1 allelic variant in a 46, XX affected person includes adrenocortical insufficiency but no ovarian dysfunction at pubertal age (94).

 

The third report of NR5A1 defects in humans was found in an infant with a similar phenotype of the first case: primary adrenal failure and 46,XY DSD. However, this child had inherited the homozygous p.R92Q alteration in a recessive manner  (95). The change lies within the A-box of NR5A1, which interferes with monomeric DNA binding stability, but in vitro functional activity was in the order of 30–40% of the wild type  (95-97). Carrier parents showed normal adrenal function suggesting that the loss of both alleles is required for the phenotype development when disrupted protein keeps this level of functional activity. In addition, another family has been reported with a homozygous missense variant (p.D293N) in the LBD of NR5A1 (98). This change also showed partial loss-of-function (50%) in gene transcription assays.

 

In 2004, we reported the fourth NR5A1 deleterious variant in humans, which brought two novel variables to the NR5A1 phenotype: it was the first frameshift variant and it was identified in a 34-year-old 46,XY DSD female with normal adrenal function (99). Another interesting aspect in this patient was the absence of gonadal tissue at laparoscopy. Since she had atypical genitalia and absence of Müllerian derivatives, we assumed that testicular tissue regressed completely late in fetal life.

 

NR5A1 changes associated with 46,XY DSD are usually frameshift, nonsense or missense changes that affect DNA-binding and gene transcription (96). Most of the point variants identified in NR5A1 are located in the DNA-binding domain of the protein. The p.L437Q variant, the first located in the ligand-binding region, was identified in a patient with a mild phenotype, a penoscrotal hypospadias. This protein retained partial function in several NR5A1-expressing cell lines and its location points to the existence of a ligand for NR5A1, considered an orphan receptor so far (97). NR5A1 is bound to sphingosine (SPH) and lyso-sphingomyelin (lysoSM) under basal conditions (100,101).

 

Progressive androgen production and virilization in adolescence has been observed in several XY patients with NR5A1 variants, in contrast to the severe under virilized external genitalia found in most patients (101,102). The almost normal testosterone levels after hCG stimulation test or at pubertal age suggest that NR5A1 action might be less implicated in pubertal steroidogenesis than during fetal life.

 

In contrast, fetal Sertoli cell function seems to be preserved in most patients with heterozygous NR5A1 variants based on the common observation of absent Müllerian derivatives and primitive seminiferous tubules in histology. The reviewed data of seventy-two 46,XY DSD patients with NR5A1 pathogenic variants reported in the literature, for whom information on the presence or absence of Müllerian derivatives was available, suggested that Müllerian derivatives are present in about 24% of the cases. However, persistently elevated FSH levels after puberty found in all patients studied suggest an impairment of Sertoli cells function in post pubertal age.

 

More than 180 different NR5A1 variants, distributed across the full length of the protein, have been described and the majority are nonsynonymous variants (103-105).

 

 Most of these variants are in the DNA binding domain and are in heterozygous state or compound heterozygous state with the p.G46A (rs1110061) variant. A clear correlation between the location of a variant, it’s in vitro functional performance and the associated phenotype is not observed. Indeed, family members bearing the same NR5A1variant may present with different phenotypes (106).

 

The contribution of other genetic modifiers has been suggested to explain phenotypic variability. Exome sequencing analyses of DSD patients have identified pathogenic variants or variants of uncertain significance in several genes involved in sexual development (42). In a 46,XY patient with atypical external genitalia, palpable inguinal gonads, absent uterus in pelvic ultrasonography and poor testosterone response to hCG stimulation, Mazen and colleagues identified, by exome sequencing, the previously described p.Arg313Cys NR5A1 variant in compound heterozygous state with a p.Gln237Arg MAP3K1 variant 27169744 (107). This NR5A1 variant was previously reported in association with mild hypospadias (108), and a possible digenic inheritance was proposed to explain the phenotypic heterogeneity (107).

 

In several cohort studies, NR5A1 changes have been reported in approximately 10–15% of the individuals with gonadal dysgenesis  (89,96). Although many of the heterozygous changes are de novo, about one-third of these changes have been shown to be inherited from the mother in a sex-limited dominant manner  (96). These women are at potential risk of primary ovarian insufficiency but while fertile they can pass NR5A1 heterozygous changes to their children. This mode of transmission can mimic X-linked inheritance (96). The features in different affected family members can be variable.

 

A different role of NR5A1 in human reproductive function was described by Bashamboo and co-workers (109). They investigated whether changes in NR5A1 could be found in a cohort of 315 men with normal external genitalia and non-obstructive male factor infertility where the underlying cause was unknown (109). Analysis of NR5A1 in this cohort identified heterozygous changes in seven individuals; all of them were located within the hinge region of the NR5A1 protein. The men who harbored NR5A1 changes had more severe forms of infertility (azoospermia, severe oligozoospermia) and in several cases low testosterone and elevated gonadotropins were found. A serial decrease in sperm count was found in one-studied men, raising the possibility that heterozygous changes in NR5A1 might be transmitted to offspring, especially if fatherhood occurs in young adulthood rather than later in life (110). As progressive gonadal dysgenesis is likely, gonadal function should be monitored in adolescence and adulthood, and early sperm cryopreservation considered in male patients, if possible. In conclusion, this study shows that changes in NR5A1 may be found in a small subset of phenotypically normal men with non- obstructive male factor infertility where the cause is currently unknown. These individuals may be at risk of low testosterone in adult life and may represent part of the adult testicular dysgenesis syndrome (110,111).

 

A novel heterozygous missense variant (p.V355M) in NR5A1 was identified in one boy with a micropenis and testicular regression syndrome (112). NR5A1 variants have also been identified in familial and sporadic forms of 46,XX primary ovarian insufficiency not associated with adrenal failure (98,113). Most of these women harbored heterozygous alterations in NR5A1 and had been identified in families with histories of 46,XY DSD and 46,XX POI. Heterozygous NR5A1 changes were also found in two girls with sporadic forms of POI (98). In one large kindred, a partial loss-of-function NR5A1 change (p.D293N) was inherited in an autosomal recessive manner. These 46,XX women with p.D293N NR5A1 variant presented with either primary or secondary amenorrhea and with a variable age of features onset. The detection of NR5A1 alterations in 46,XX ovarian failure shows that NR5A1 is also a key factor in ovarian development and function in humans. Thus, some 46,XX women with NR5A1 variants have normal ovarian function and can transmit the variant in a sex-limited dominant pattern. Therefore, the inheritance patterns associated with NR5A1 changes can be autosomal dominant, autosomal recessive or sex-limited dominant.

 

NR5A1 defects can be found in association with a wide range of human reproductive phenotypes such as 46,XY and 46,XX disorders of sex development (DSD) associated or not with primary adrenal insufficiency, male infertility, primary ovarian insufficiency and finally testicular or ovo-testicular 46,XX DSD (101) (103) (Table 4). Spleen development anomalies have been described in patients with NR5A1 variants (103).

 

Table 2. Spectrum of Phenotypes Caused by NR5A1 Defects

 

 

Karyotype

 

Phenotypes

Number of reported patients

 

Reference

46,XY 

DSD and adrenal insufficiency

2

(92,95)

DSD without adrenal insufficiency

69

(63,89,96,98,100,101,103,106) 

 

Male infertility 

10

(63)

Ovotesticular DSD and genitopatellar syndrome*

1

(114)

46,XX

Adrenal insufficiency

2

(92,93)

Female infertility, POI

14

(98,101,103) 

(Ovo) testicular DSD

without adrenal insufficiency

11

(98,115)

 

46,XY DSD Due to Under Expression of the SRY Gene

 

Most of the authors reported pathogenic allelic variants in SRY gene in less than 20% of the patients with complete 46,XY gonadal dysgenesis (116-118). In the partial form, the frequency of SRY variants is even lower than in the complete form. To date, most of the SRY variants are located in the HMG box, showing the critical role of this domain, and are predominantly de novo variants. However, some cases of fertile fathers and their XY affected children, sharing the same altered SRY sequence, have been reported (116,119). In a few of these cases, the father’s somatic mosaicism for the normal and mutant SRY gene has been proven (120) The variable penetrance of SRY variants in familial cases have been described in SRY mutant proteins with relatively well preserved in vitro activity (121).

 

Dysgenetic 46,XY DSD Associated with Campomelic Dysplasia (Under Expression of the SOX9 Gene)

 

SRY-related HMG-box gene 9 (SOX9) is a transcription factor involved in chondrogenesis and sex determination. SOX9 gene, located on human chromosome 17, is a highly conserved HMG family member and it is also implicated in the male sex-determining pathway (122,123).

 

Pathogenic allelic variants in SOX9 have been identified in heterozygous state in patients with Campomelic dysplasia (122). This syndrome is characterized by severe skeletal malformations associated with dysgenetic 46,XY DSD. These patients have variable external genitalia ranging from that of normal male with cryptorchidism to atypical or female genitalia, and the internal genitalia may include vagina, uterus, and fallopian tubes (124).

 

Intact SOX9 were also reported in patients with Campomelic dysplasia and 46,XY gonadal dysgenesis. The genomic analysis of the SOX9 locus in these patients identified a key regulatory element termed RevSex, located approximately 600 kb upstream from SOX9. RevSex is duplicated in individuals with 46,XX (ovo)testicular DSD and deleted in individuals with 46,XY GD (125,126). Moreover, structural changes involving multiple regions both upstream and downstream of the SOX9 gene have been associated with non-syndromic XY DSD (127,128). These findings indicate that variants located in the regulatory elements of SOX9 should be routinely screened in a DSD diagnostic setting (69).

 

Dysgenetic 46,XY DSD Due to Under Expression of the FGF9/FGFR2 Genes

 

The importance of Fgf9/Fgfr2 signaling pathway in mouse testis determination is well known (129,130). In the developing testis occurs a positive feedback loop among Fgf9/Fgfr2/Sox9; Fgf9 is upregulated by Sox9 and signals through Fgfr2 maintain Sox9 expression (129) and this loop represses Wnt4 (131).

 

Mice homozygous for a null variant in Fgf9 or Fgfr2 exhibit male-to-female sex reversal, with all testis-specific cellular events being disrupted, including cell proliferation, mesonephric cell migration, Sertoli cell differentiation, and testis cord formation (129,130,132). However, in human sex development the role of FGF9 and FGFR2 remains unclear.

 

In humans, the only reported pathogenic variants in FGF9 are associated with craniosynostosis or multiple synostosis phenotypes, and no FGF9 variants were identified in 46,XY GD patients (133).

 

Human FGFR2 variants have been related with some syndromes as lacrimo-auriculo-dento-digital, characterized by tear tract, ear, teeth and digit abnormalities (133) and craniosynostosis syndromes including Crouzon, Pfeiffer, Apert and Antley-Bixler syndromes (134-136). FGFR2 variants can lead to loss (LAAD syndrome) or gain (craniosynostosis syndromes) of function in these disorders  (137). No gonadal defects were described in patients with LADD or craniosynostosis syndromes.

 

A single 46,XY patient with gonadal dysgenesis and craniosynostosis was described by Bagheri-Fam et al (138). This patient had abnormalities which are identified in several craniosynostosis syndromes (short stature, brachycephaly, proptosis, down slanting palpebral fissures, low-set dorsally rotated ears, reduced extension at the elbows but absence of hand and feet anomalies). She also presented female external genitalia, primary amenorrhea and gonadal dysgenesis with dysgerminoma. DNA sequencing revealed a cysteine-to-serine substitution at position 342 in the FGFR2c isoform (p.C342S). Cys342 substitutions by Ser or other amino acids (Arg/Phe/Trp/Tyr) occur frequently in the craniosynostosis syndromes Crouzon and Pfeiffer but these patients do not present gonadal abnormalities. Variants in the 2c isoform of FGFR2 is in agreement with knockout data showing that FGFR2c is the critical isoform during sex determination in the mouse. Taken together, these data suggest that the FGFR2c c.1025G>C (p.C342S) variant might contribute to 46,XY DSD in this patient. The authors proposed that this heterozygous variant leads to gain of function in the skull, but to loss of function in the developing gonads and that she might harbor a unique set of modifier genes, which exacerbate this testicular phenotype (138).

 

The authors proposed that the p.C342S heterozygous variant in FGFR2c leads to gain of function in the skull, but loss of function in the developing gonads; and that the presence of modifier genes would exacerbate the testicular phenotype in this patient (138). However, the presence of a pathogenic variant involving other DSD genes, cannot be completely excluded in this patient.

 

Dysgenetic 46,XY DSD Due to Disruption in Hedgehog Signaling

 

DESERT HEDGEHOG (DHH) GENE

 

It is a member of the hedgehog family of signaling proteins, is located in chromosome 12-q13.1 and is one of the genes involved in the testis-determining pathway (139). Dhh seems to be necessary for Nr5a1 up-regulation in Leydig cells in mice (140). To date, six homozygous variants have been described in DHH gene in 46,XY patients conferring phenotypes ranging from partial to complete gonadal dysgenesis, associated or not with polyneuropathy. The first one, the homozygous missense variant (p.M1T) is located at the initiation codon of exon 1 and was found in a 46,XY patient with partial gonadal dysgenesis associated with polyneuropathy (141). Two other variants, one the p.L162P located at exon 2 and the other the p.L363CfsX4 located in exon 3 were identified in three patients with complete gonadal dysgenesis without polyneuropathy; two of them harbored gonadal tumors (bilateral gonadoblastoma and dysgerminoma, respectively) (142). Later, the c.1086delG variant was identified in heterozygous state in two patients with partial gonadal dysgenesis (143). In addition, two novel homozygous variants were described in two patients with complete 46,XY gonadal dysgenesis without clinically overt polyneuropathy (144). In both sisters, clinical neurological examination revealed signs of a glove and stocking like polyneuropathy. The first defect, the c.271_273delGAC resulted in deletion of one amino acid (p.D90del) and the second one, a duplication c.57_60dupAGCC resulted in a premature termination of DHH protein (144) . The p.R124Q variant was identified by exome sequencing in two sisters of a consanguineous family with 46, XY gonadal dysgenesis and testicular seminoma (145).

 

HEDGEHOG ACETYL-TRANSFERASE (HHAT) GENE

 

The HHAT protein is a member of the MBOAT family of membrane-bound acyl-transferases which catalyzes amino-terminal palmitoylation of Hh proteins. The novel variant (p.G287V) in the HHAT gene was found in a syndromic 46,XY DSD patient with complete gonadal dysgenesis and skeletal malformation by exome sequencing. This variant disrupted the ability of HHAT protein to palmitoylated Hh proteins including DHH and SHH (146) In mice, the absence of Hhat in the XY gonad did not affect testis-determination, but impaired fetal Leydig cells and testis cords development (146). The phenotype of the girl carrying the homozygous p.G287V variant is a rare combination of gonadal dysgenesis and chondrodysplasia. Moreover, a de novo dominant variant in the MBOAT domain of HHAT was reported in association with intellectual disability and apparently normal testis development (147).

 

46,XY DSD Due to Under Expression of the DMRT1 Gene

 

Raymond et al identified both DNA-binding Motif (DM) domain genes expressed in testis (DMRT1 and DMRT2) located in chromosome 9p24.3, a region associated with gonadal dysgenesis and 46,XY DSD (148-150). The human 9p monosomy syndrome is characterized by variable degrees of 46,XY DSD, from female genitalia to male external genitalia with cryptorchidism associated to agonadism, streak gonads or hypoplastic testes and internal genitalia disclosing normal Müllerian or Wolffian ducts, mental retardation and craniofacial abnormalities (151). Gonadal function varies from insufficient to near normal testicular production. It is inferred that haploinsufficiency of DMRT1and DMRT2 primarily impairs the formation of the undifferentiated gonad, leading to various degrees of testis or ovary formation defects (151).

 

Although 9p24 deletions are a relatively common cause of syndromic 46,XY gonadal dysgenesis, the pathogenic variants within DMRT1 are rarely identified (152).

 

Genomic–wide copy number variation screening revealed that DMRT1 deletions were associated with isolated 46,XY gonadal dysgenesis in addition to inactivation variants (133,148). In vitro studies to analyze the functional activity of the DMRT1 (p.R111G) variant identified by exome sequencing in a patient with 46,XY complete gonadal dysgenesis, indicated that this protein had reduced DNA affinity and altered sequence specificity. This mutant DMRT1, when mixed with the wild-type protein bound as a tetramer complex to an in vitro Sox9 DMRT1-binding site, differs from the wild-type DMRT1 that is usually bound as a trimer. This suggests that a combination of haploinsufficiency and a dominant disruption of the normal DMRT1 target binding site is the cause of the abnormal process of testis-determination seen in this patient (153).

 

Matson et al. (2011) have shown in mice that Dmrt1 and Foxl2 create another regulatory network necessary for maintenance of the testis during adulthood. Loss of Dmrt1 in mouse Sertoli cells induces the reprogramming of those into granulosa cells, due to Foxl2 upregulation. Consequently, theca cells are formed, estrogens are produced, and germ cells appear feminized (49).

 

ATR-X Syndrome (X-linked α-Thalassemia and Mental Retardation)

 

ATR-X syndrome results from variants in the gene that encodes for X-linked helicase-2, implicating ATR-X in the development of the human testis (154). Genital anomalies leading to a female sex of rearing were reported in several affected 46,XY patients with ATR-X syndrome (155).

 

ATR-X syndrome is characterized by severe mental retardation, alpha thalassemia and a range of genital abnormalities in 80% of cases (154). In addition to these definitive phenotypes, patients also present with typical facial anomalies comprising a carp-like mouth and a small triangular nose, skeletal deformities and a range of lung, kidney, and digestive problems. A variety of phenotypically overlapping conditions (Carpenter-Waziri syndrome, Holmes-Gang syndrome, Jubert-Marsidi syndrome, Smith-Fineman-Myers syndrome, Chudley-Lowry syndrome and X-linked mental retardation with spastic paraplegia without thalassemia) have also been associated with ATRX variants (154).

 

ATRX lies on the X chromosome (Xq13) and the disease has been confined to males; in female carriers of an ATRX variant, the X-inactivating pattern is skewed against the X chromosome carrying the mutant allele.

 

Urogenital abnormalities associated with variants in human ATRX range from undescended testes to testicular dysgenesis with female or atypical genitalia. Duplication of Xq12.2-Xq21.31 that encompasses ATRX along with other genes has been described in a male patient with bilateral cryptorchidism and severe mental retardation. The patient entered spontaneous puberty by the age of 12 and developed bilateral gynecomastia (156). There are two major functional domains in ATRX protein: 1- the ATRX-DNMT3-DNMT3L (ADD) domain at the N-terminus and 2- the helicase/ATPase domain at the C-terminal half of the protein, both acting as chromatin remodeling. variants in the ADD domain have been related to severe psychomotor impairment associated with urogenital abnormalities. On the other hand, variants in the C-terminus region have been related with mild psychomotor impairment without severe urogenital abnormalities (157,158).

 

Although all cases of severe genital abnormality reported in ATRX syndrome have been associated with severe mental retardation, this is not true for alpha-thalassemia. The role of ATRX in the sexual development cascade is poorly understood and it is suggested that it could be involved in the development of the Leydig cells  (159).

 

Dysgenetic 46,XY DSD Due to Under Expression of the MAP3K1 Gene

 

MAPK signaling pathway role in mammalian sex-determination is still poorly understood. In mice, it has been shown that the Map3k4 gene is essential for testicular determination, since the lack of activity of this protein leads to failure of testicular cord development and disorganization of gonadal tissue in development (160). In mice, the reduction of the Gadd45/Map3k4/p38 pathway activity is associated with a reduction in the Sry expression in the XY mice gonad at sex-determination causing sex-reversal in these animals (161). Studies with knock-in animals for the Map3k1 gene demonstrated a lower repercussion in the testicular tissue, which present a reduction in the Leydig cells number (162,163). However, in patients with 46, XY gonadal dysgenesis, different non-synonymous allelic variants were identified in the MAP3K1 gene. The first variant described was identified for mapping by linkage analysis of an autosomal sex-determining gene locus at the long arm of chromosome 5 in two families with 46,XY DSD, including patients with complete and partial gonadal dysgenesis. The splice-acceptor variant c.634-8T>A in the MAP3K1 disrupted RNA splicing and was segregated with the phenotype in the first family. Variants in the MAP3K1 were also demonstrated in the second family (p.G616R) and in two of 11 sporadic 46,XY DSD patients (p.L189P, p.L189R) studied (51,164). Subsequently, the two novel variants p.P153L and c.2180- 2A>G in the MAP3K1 were identified in non-syndromic patients with 46,XY gonadal dysgenesis. Functional studies of mutated MAP3K1 proteins identified change in phosphorylation targets in subsequent steps of the cascade of MAP3K1, p38 and ERK1/2 and enhanced the binding of the Ras homolog gene family, member A (RHOA) to the MAP3K1 complex (51). In normal male gonadal development, the binding of MAP3K1 to the RHOA protein promotes a normal phosphorylation of p38 and ERK1/2, and a blockade of the β-catenin pathway is determined by MAP3K4. In the female development, hyperphosphorylation of p38 and ERK1/2 occurs and the presence of p38 and ERK1/2 hyperphosphorylated determine the activation of the β-catenin pathway, that result in a block of the positive feedback pathway of SOX9 and the testicular development  (51) .

 

Cohorts of patients with 46,XY DSD studied by a targeted gene panel have found several new potentially deleterious variants and uncertain significance variants in the MAP3K1 (26). Although the findings strongly indicate the participation of the MAP3K1 variants in the etiology of testicular development abnormalities, a better understanding of the mechanisms of MAPK pathway in the gene regulatory networks of the human testicular determination process is still necessary (52,107).

 

46,XY DSD Due to Over Expression of the NR0B1/DAX1 Gene

 

Male patients with female or atypical external and internal genitalia due to partial duplications of Xp in the presence of an intact SRY gene have been described (28). These patients present with dysgenetic or absent gonads associated or not with mental retardation, cleft palate, and dysmorphic face. Bardoni et al. identified in these patients, a common 160-kb region of Xp2 containing NR0B1/DAX1 gene named dosage sensitive sex  locus which, when duplicated, resulted in 46,XY DSD (164).

 

The large duplications of Xp21 reported prior to array-CGH and MLPA techniques were identified by conventional karyotyping. Patients carried large genomic rearrangements involving several genes. In these patients, the presence of XY gonadal dysgenesis was part of a more complex phenotype, which also included dysmorphic features and/or mental retardation (165).

 

Interestingly, in all cases with isolated 46,XY gonadal dysgenesis, the IL1RAPL1 gene located immediately to the duplication containing NR0B1/DAX1, is not disrupted. Deletions or variants of this gene have been identified in patients with mental retardation (166). Disruption of this gene could explain the mental retardation previously described in patients with larger Xp21 duplications (167).

 

Several patients with isolated 46,XY gonadal dysgenesis due to duplications of Xp21 have been described. The first report identified a 637 kb tandem duplication on Xp21.2 that in addition to NR0B1/DAX1 includes the four MAGEBgenes in two sisters with isolated 46,XY gonadal dysgenesis and gonadoblastoma (168). The second case exhibited a duplication with approximately 800 kb in size and, in addition to NR0B1/DAX1, contains the four MAGEB, Cxorf21 and GK genes. The healthy mother was a carrier of the duplication (169).

 

Smyk et al. described a 21-years-old 46,XY patient manifesting primary amenorrhea, a small immature uterus, gonadal dysgenesis and absence of adrenal insufficiency with a submicroscopic deletion (257 kb) upstream of NR0B1/DAX1. The authors hypothesized that loss of regulatory sequences may have resulted in up-regulation of DAX1 expression, consistent with phenotypic consequences of NR0B1/DAX1 duplication (170).

 

By using array-CGH and MLPA techniques, additional NR0B1/DAX1 locus duplications have been identified in patients with isolated 46,XY gonadal dysgenesis (28,169,171).

 

Barbaro et al. identified a relatively small NR0B1/DAX1 locus duplication responsible for isolated complete 46,XY gonadal dysgenesis in a large English family (28). The duplication extends from the MAGEB genes to part of the MAP3K7IP3 gene, including NR0B1, CXorf21, and GK genes. Unfortunately, the authors were unable to set up the rearrangement mechanism and distinguish between a nonallelic homologous recombination or a nonhomologous end joining mechanism. Therefore, until now, there is not a direct proof that an isolated NR0B1/DAX1 duplication is sufficient to cause 46,XY gonadal dysgenesis in humans, suggesting that other contiguous genes located in the DSS locus, should be involved in dosage-sensitive 46,XY DSD.

 

X-inactivation patterns in fertile female carriers of each of the three small NR0B1 locus duplications were analyzed (169). They established that female carrier of macroscopic Xp21 duplications are healthy and fertile due to the preferential inactivating of the duplicated chromosome and thereby protecting them from increased NR0B1 expression (169).

 

46,XY DSD Due to the Over Expression of WNT4 Gene

 

The WNT4 (wingless-type mouse mammary tumor virus integration site member 4) gene belongs to a family that consists of structurally related genes that encode cysteine-rich secreted glycoproteins that act as extracellular signaling factors (172).

 

Overexpression of the WNT4 and RSPO1 may be a cause of 46,XY DSD. A 46,XY newborn infant, with multiple congenital anomalies including bilateral cleft lips and palate, intrauterine growth retardation, microcephaly, tetralogy of Fallot, atypical external and internal genitalia, and undescended gonads consisted of rete testes and rudimentary seminiferous tubules, who carried a duplication of 1p31-p35, including both WNT4 and RSPO1 genes, was reported (173). In vitro functional studies showed that Wnt4 up-regulates Nr0b1/Dax1 in Sertoli cells, suggesting that Nr0b1/Dax1 overexpression was the cause of 46,XY DSD in this infant (174).

 

Table 3. Phenotypic Spectrum of Defects in the Genes Involved in Human Male Sex Determination

 

Genes

Chromosome position

Molecular

defect

External

genitalia

Müllerian ducts derivatives

Testes

Associated anomalies

Associated Syndrome

ARX

Xp22

Deletion/ Inactivating variants

Atypical/ micropenis with cryptorchidism

-

Dysgenetic

Abnormal psychomotor development, epilepsy, spasticity, and intellectual disability

X-linked lissencephaly, Proud syndrome,

Ohtahara syndrome

 

ATRX

Xq13

Inactivating variants

Atypical / Male with cryptorchidism

-

Dysgenetic

Severe psycho-motor retardation, dysmorphic face, cardiac and skeletal abnormalities, thalassemia

Alpha thalassemia and mental retardation X-linked

CBX2

17q25

Inactivating variants

Female

+

Normal

Ovary

No

No

DHH

12q12

Inactivating variants

Female/Atypical

+/-

Dysgenetic / Testis

Minifascicular

neuropathy

No

DHX37

12q24.31

Inactivating variants

Male with cryptorchidism and micropenis, Atypical

+/-

Dysgenetic/

Absent

No

No

DMRT1

9p24

Deletion/Inactivating variants

Female/ Atypical/ Male with cryptorchidism

+/-

Dysgenetic/Absent/ Hypoplastic

Craniofacial Abnormalities, microcephaly, mental retardation

No

DSS locus

(DAX-1 /MAGEB)

Xp21

Gene

duplication

Female/ Atypical/ male

+/-

Dysgenetic/

Absent

Mental retardation, cleft palate, dysmorphic face

No

FGFR2

10q26

Inactivating variants

Female

ND

Dysgenetic

Short stature, craniofacial abnormalities, elbow and knee contractures

Craniosynostosis

syndrome

FOG2/ZFPM2

8q23

Balanced translocation, inactivating variants

Male

-

Probable

dysgenetic

Heart defects

No

GATA4

8p23

Inactivating variants

Atypical / male with micropenis

-

Normal/

Dysgenetic

Heart defects

No

HHAT

1q32

Inactivating variants

Female

+

Dysgenetic

Chondrodysplasia

Nivelon-Nivelon-Mabille syndrome

MAP3K1

5q11.2

Inactivating mutation

Female/Atypical

+

Dysgenetic

No

No

MYRF

11q12.2

Inactivating variants

Female/Atypical

-

ND

Congenital heart defects, urogenital anomalies, congenital diaphragmatic hernia, and pulmonary hypoplasia

Cardiac urogenital syndrome

NR5A1

9q33

Inactivating variants

 

Female/Atypical/ Male with cryptorchidism

Male with spermatogenic failure

+/-

Normal/

Dysgenetic/

Absent

Adrenal

Insufficiency

No

PPP1R12A

12q21.2- q21.31

Inactivating variants

Female/Atypical

+/-

Dysgenetic

Genitourinary and/or brain malformations

No

SOX9

17q24.3-25.1

Inactivating variants,

5’ and 3’ Rearrangements

Female/ Atypical Male

+/-

Dysgenetic

Severe skeletal defects

Campomelic

displasia

SRY

 

Yp11.3

Inactivating variants

Female/ Atypical

+

Dysgenetic

No

No

WNT4

/RSPO1 locus

1p34.3-p35

Gene duplication

Atypical

+

Dysgenetic

Cleft lips and palate, tetralogy of Fallot, intrauterine growth retardation, microcephaly

No

WT1

11p13

Inactivating variants

Female/ Atypical

+/-

Dysgenetic

Late-onset renal failure Gonadoblastoma

Frasier

Inactivating variants

Atypical

+/-

Dysgenetic

Early-onset renal failure, Wilm's tumor

Denys-Drash

Inactivating variants

Female/ Atypical / Male with cryptorchidism

-

Dysgenetic

Mental retardation, Wilm's tumor, Aniridia, renal agenesis or horseshoe kidney

WAGR

WWOX

16q23

Multi-exons deletion

Atypical

+

Dysgenetic

No

 

-

ND: data not described

 

46,XY DSD ASSOCIATED WITH CHOLESTEROL SYNTHESIS DEFECTS

 

Smith-Lemli-Opitz Syndrome (SLOS)

 

This syndrome, caused by a deficiency of 7-dehydrocholesterol reductase, is the first true metabolic syndrome leading to multiple congenital malformations (179,180).

 

This disorder is caused by variants in the sterol delta-7-reductase (DHCR7) gene, which maps to 11q12-q13. Typical facial appearance is characterized by short nose with anteverted nostrils, blepharoptosis, microcephaly, photosensitivity, mental retardation, syndactyly of toes 2 and 3, hypotonia, and atypical genitalia. Adrenal insufficiency may be present or evolve with time. Atypical external genitalia are a frequent feature of males (71%) and ranges from hypospadias to female external genitalia despite normal 46,XY karyotype and SRY sequences. Müllerian derivative ducts can also be present (181-183). The etiology of masculinization failure in SLOS remains unclear. However, the description of patients with SLOS who present with hyponatremia, hyperkalemia, and decreased aldosterone-to-renin ratio suggest that the lack of substrate to produce adrenal and testicular steroids is the cause of adrenal insufficiency and atypical genitalia (184), although, a revision of HPA axis in these patients showed normal HPA axis function (185).

 

Affected children present elevations of 7-dehydrocholesterol (7DHC) in plasma or tissues. 7DHC is best assayed using Gas Chromatography/Mass Spectroscopy (GC/MS). Considering the relative high frequency of Smith-Lemli-Opitz syndrome, approximately 1 in 20,000 to 60,000 births, we suggest that at least cholesterol levels should be routinely measured in patients with 46,XY DSD. However, although frequently low, plasma cholesterol levels can be within normal limits in affected patients.

 

DHCR7 variant analysis can confirm a diagnosis of SLOS. The human DHCR7 gene is localized on chromosome 11q13 and contains nine exons encoding a 425 amino-acid protein (64). More than 130 different variants of DHCR7have been identified and the great majority of them are located at the exons 6 to 9 (186,187). However, the genotype-phenotype correlation in SLOS is relatively poor (188).

 

Currently, most SLOS patients are treated with cholesterol supplementation that can be achieved by including high cholesterol foods and/or suspensions of pharmaceutical grade cholesterol. Data suggests that early intervention may be of benefit to SLOS patients (189). Observational studies report improved growth and muscle tone and strength, increased socialization, decreased irritability and aggression in SLOS patients treated with cholesterol supplementation. However, in a group of SLOS patients’ treatment with a high cholesterol diet did not improve developmental scores (190).

 

Treatment with simvastatin, an HMG-CoA reductase inhibitor, aiming to block the cholesterol synthesis pathway avoiding the formation of large amounts of 7DHC/8DHC, and in this manner limiting exposure to potentially toxic metabolites in SLOS patients has been proposed. Simvastatin can also cross the blood–brain barrier and may provide a means to treat the biochemical defect present in the CNS of SLOS patients (191). A major effect of statin therapy is the transcriptional upregulation of genes controlled by the transcriptional factor SREBP, including DHCR7. Thus, if any residual activity is present in the mutant DHCR7, its upregulation could increase intracellular cholesterol synthesis. Simvastatin use in SLOS patients resulted in a paradoxical increase in serum and cerebrospinal fluid cholesterol levels (191). Randomized controlled-placebo trials were performed with simvastatin in SLOS showing significant reduction in plasmatic 7DHC associated with improvement in irritability symptoms (192). Determination of residual DHCR7 enzymatic activity may be helpful in selecting SLOS patients to be considered for a beneficial response of statins (187). Recently, promising gene therapy using an adeno-associated virus vector carrying a functional copy of the DHCR7 gene was administered by intrathecal injection in a mouse model with improvement of cholesterol levels in the central nervous system (193).

 

Table 4. Phenotype of 46,XY Subjects with Smith-Lemli-Optiz Sndrome

Inheritance

Autosomal recessive

External genitalia 

Micropenis and/or hypospadias, hypoplastic or bifid scrotum; female

Müllerian duct derivatives

May be present

Wolffian duct derivatives

Absent to male

Testes

Scrotum, inguinal or intra-abdominal region

Clinical features

Facial and bone abnormalities. Heart and pulmonary defects. Renal agenesis. Mental retardation, Seizures, hypotonia, syndactyly of second and third toes.

Puberty

Apparently normal

Hormonal diagnosis

Low cholesterol, elevated 7-dehydrocholesterol. Decreased aldosterone-to-renin ratio

Gender role

Male

DHCR7 gene location

11q12-q13

Molecular defect

variants in DHCR7 gene

Treatment

Dietary cholesterol supplies accompanied by ursodeoxycholic acid, and statins

Outcome

Severe mental retardation

 

Dysgenetic 46,XY DSD Due to Under Expression of the DHX37 gene

 

46,XY gonadal dysgenesis (GD) is a heterogeneous group of disorders with a wide phenotypic spectrum, including embryonic testicular regression syndrome (ETRS) (Table 5). Screening of 87 patients with 46,XY DSD (17 familial cases from 8 unrelated families and 70 sporadic cases) using whole-exome sequencing and target gene-panel sequencing identified a new player  in the complex cascade of male gonadal differentiation and maintenance - the Asp-Glu-Ala-His-box (DHX) helicase 37 (DHX37) gene (53). The variants were especially associated with ETRS (7/14 index cases; 50%). The frequency of rare, predicted-to-be-deleterious DHX37 variants in this cohort (14%) is significantly higher than that observed in the Genome Aggregation Database (0.4%; P < 0.001). Immunohistochemistry analysis in human testis showed that DHX37 is mainly expressed in germ cells at different stages of testis maturation, in Leydig cells, and rarely in Sertoli cells. Other papers confirmed these findings, associating 46,XY gonadal dysgenesis with defects in DHX37 gene (152,175).

 

Table 5. Phenotype of 46,XY Subjects with Gonadal Dysgenesis Due to DHX37Defects

Inheritance

Autosomal dominant

External genitalia 

Micropenis, atypical genitalia or typical female

Müllerian duct derivatives

Absent uterus, Fallopian tubes may be present

Wolffian duct derivatives

Present

Testes

Abdominal region or absent

Histological analysis

Dysgenetic, no gonadal tissue

Puberty

Hypergonadotropic hypogonadism

Hormonal diagnosis

Elevated serum levels of LH and FSH; very low levels of testosterone and normal testosterone precursors levels

Gender role

Male, female, male to female

DHX37 gene location

12q24.31

Molecular defect

Heterozygous variants in DHX37 gene

Treatment

Repair of atypical genitalia; estrogen or testosterone replacement according to social sex

Outcome

Most patients keep the male social sex; some change to female social sex

 

Different modes of inheritance have been reported in familial cases of 46,XY gonadal dysgenesis, including autosomal dominant, autosomal recessive, X-linked and multifactorial inheritance (polygenic) (107,176-178). Oligogenic mode of inheritance might explain genotype/phenotype variability observed in 46,XY gonadal formation patients.  Pathogenic allelic variants in NR5A1, DHX37, MAP3K1 and SRY are the most frequent molecular causes of 46,XY gonadal dysgenesis (20).

 

46,XY DSD DUE TO TESTOSTERONE PRODUCTION DEFECTS

 

46,XY DSD Due to Impaired Leydig Cell Differentiation (Complete and Partial Forms)

 

Inactivating variants of human LHCG receptor (LHCGR) have been described in 46,XY individuals with a rare form of disorder of sex development, termed Leydig cell hypoplasia. These inactivating variants in the LHCGR prevent LH and hCG signal transduction and thus testosterone production both pre- and postnatally in genetic males (194).

 

Both hCG and LH act by stimulating a common transmembrane receptor, the LHCGR  (195) LHCGR is a member of G protein-coupled receptors, which are characterized by the canonical serpentine region, composed of seven transmembrane helices interconnected by three extracellular and three intracellular loops (196,197). The large amino-terminal extracellular domain, rich in leucine-repeats, mediates the high affinity binding of pituitary LH or placental human chorionic gonadotropin (hCG) (197).

 

LHCGR activates the Gs protein, which determines an increase in intracellular cAMP and a subsequent stimulation of steroidogenesis in gonadal cells such as testicular Leydig cells, ovarian theca cells and differentiated granulosa cells (195,198) A secondary mechanism of LHCGR stimulation is through Gq/11 protein activation and the inositol phosphate signaling pathway (197).

 

The LHCGR gene is located on the short arm of chromosome 2 (2p21). It spans nearly 80 kb and has been thought to be composed of 11 exons and 10 introns. Exon 11 of the LHCGR gene encodes the entire serpentine domain as well as the carboxy-terminal portion of the hinge region (NCBI GeneID 3973; http://www.ncbi.nlm.nih.gov). The amino-terminal portion of the hinge region is encoded by exon 10 and the signal peptide and remaining portion of the extracellular domain are encoded by exons 1-9 (194,196). A novel primate-specific exon (termed exon 6A) was identified within intron 6 of the LHCGR gene. This exon is not used by the wild-type full-length receptor. It displays composite characteristics of an internal/terminal exon and possesses stop codons triggering nonsense-mediated mRNA decay in LHCGR. When exon 6A is utilized, it results in a truncated LHCGR protein (199).

 

In 1976, Berthezene et al. (200) described the first patient with Leydig cell hypoplasia and subsequently several cases have been reported (201-203). The clinical features are heterogeneous and result from a failure of intrauterine and pubertal virilization. A review of the literature allowed  delineation of the characteristics of 46,XY DSD due to the complete form of Leydig cell hypoplasia as: 1) female external genitalia leading to female sex assignment 2) no development of sexual characteristics at puberty, 3) undescended testes slightly smaller than normal with relatively preserved seminiferous tubules and absence of mature Leydig cells, 4) presence of rudimentary epididymis and vas deferens and absence of uterus and fallopian tubes, 5) low testosterone levels despite elevated gonadotropin levels, with elevated LH levels predominant over FSH levels, 6) testicular unresponsiveness to hCG stimulation, and 7) no abnormal step up in testosterone biosynthesis precursors (194,204) (table 6).

 

Several different variants in the LHCGR gene were reported in patients with Leydig cell hypoplasia in both sexes (194,205).

 

Table 6.  Phenotype of 46,XY Subjects with the Complete Form of Leydig Cell Hypoplasia

Inheritance

Autosomal recessive

External genitalia

Female, occasionally mild clitoromegaly or labial fusion

Müllerian derivatives

Absent

Wolffian ducts derivatives

Absent or vestigial

Testes

Inguinal or intra-abdominal, slightly subnormal size

Puberty

Absence of spontaneous virilization or feminization

Hormonal diagnosis

Elevated serum LH, normal or slightly elevated FSH and very low testosterone levels with normal levels of testosterone precursors

Gender role

Female

LHCGR gene location

2p21

Molecular defect

Pathogenic variants in LHCGR gene (complete inactivation) and in the internal exon 6A LHCGR(increase of nonfunctional isoform); defects in LHCGRwere not identified in several families

Treatment

Estrogen replacement at pubertal age, bilateral orchiectomy and vaginal dilation

Outcome

Female gender role and behavior, infertility

 

In contrast to the homogenous phenotype of the complete form of Leydig cell hypoplasia, the partial form features a broad spectrum, ranging from incomplete male sexual differentiation characterized by micropenis and/or hypospadias to hypergonadotropic hypogonadism without ambiguity of the male external genitalia (194,195,206,207). Testes are cryptorchidic or in the scrotum and during puberty, partial virilization occurs and testicular size is normal or only slightly reduced, while penile growth is significantly impaired. Spontaneous gynecomastia does not occur. Before puberty, the testosterone response to the hCG test is subnormal without accumulation of testosterone precursors. After puberty, LH levels are elevated as a result of insufficient negative feedback of gonadal steroid hormones on the anterior pituitary and testosterone levels are intermediate between those of children and normal males.

 

Several mutations in the LHCGR gene have also been identified in patients with the partial form of Leydig cell hypoplasia. Latronico et al. reported the first homozygous mutation in the LHCGR (p.Ser616Tyr) in a boy with micropenis (207). Subsequently, other milder mutations were identified in further patients with the partial form of Leydig cell hypoplasia (194,195,207). In vitro studies showed that cells transfected with LHCGR gene containing these mutations had an impaired hCG-stimulated cAMP production (195,207).

 

Leydig cell hypoplasia was found to be a genetic heterogenous disorder since Zenteno et al. (197) ruled out, by segregation analysis of a known polymorphism in exon 11 of the LHCG receptor gene, molecular defects in the LHCG receptor as being responsible for Leydig cell hypoplasia in three siblings with 46,XY DSD. Most inactivating mutations of the LHCGR are missense mutations that result in a single amino acid substitution in the LHCGR. In addition, mutations causing amino acid deletions, amino acid insertions, splice acceptor mutation or premature truncations of the receptor have also been reported (208). LHCGR mutations are usually located in the coding sequence, resulting in impairment of either LH/CG binding or signal transduction.

 

Although it is well known that hCG and LH act by stimulating a common receptor, a differential action of them in the LHCGR has been suggested. The identification of a deletion of exon 10 of the LHCGR in a patient with normal male genitalia at birth, but no pubertal development indicated that the mutant LHCGR was responsive to fetal hCG, but resistant to pituitary LH. The binding affinity of hCG for LHCGR was normal in vitro analysis, suggesting that exon 10 is necessary for LH, but not for hCG action (199).

 

The identification and characterization of a novel, primate-specific bona fide exon (exon 6A) within the LHCGR determined a new regulatory element within the genomic organization of this receptor and a new potential mechanism of this disorder. Kossack et al analyzing the exon 6A in 16 patients with 46,XY DSD due to Leydig cells hypoplasia without molecular diagnosis, detected mutations (p.A557C or p.G558C) in three patients. Functional studies revealed a dramatic increase in expression of the mutated internal exon 6A transcripts, resulting in the generation of predominantly nonfunctional isoforms of the LHCGR, thereby preventing its proper expression and functioning (209).

 

A new compound heterozygous mutation of the LHCGR, constituted by a previously described missense mutation (p.Cys13Arg) and a large deletion of the paternal chromosome 2 was identified by array-Comparative Genomic Hybridization (array-CGH) in a 46,XY infant with sexual ambiguity and low hCG-stimulated testosterone levels associated with high LH and FSH levels (200).

 

In addition, causative mutations in LHCGR were absent in around 50% of the patients strongly suspected to have Leydig cell hypoplasia. These findings supported the idea that other genes must be implicated in the molecular basis of this disorder. 

 

We observed that 46,XX sisters of the patients with 46,XY DSD due to Leydig cell hypoplasia, carrying the same homozygous mutation in the LHCGR, have primary or secondary amenorrhea, spontaneous breast development, infertility, normal or enlarged cystic ovaries with elevated LH and LH/FSH ratio, normal estradiol and progesterone levels for early to mid-follicular phase, but not for luteal phase levels, confirming lack of ovulation (198,207,210). Our findings were subsequently confirmed by other authors who studied 46,XX sisters of 46,XY DSD patients with Leydig cell hypoplasia (201,202,211).

 

Subsequently, a novel homozygous missense mutation, p.N400S, has been identified by whole genome sequencing in two sisters with empty follicle syndrome (204).

 

Table 7. Phenotype of 46,XY Subjects with Partial Leydig Cells Hypoplasia

Inheritance 

Autosomal recessive

External genitalia 

Atypical to male

Müllerian derivatives 

Absent

Wolffian ducts derivatives 

Rudimentary to male

Testes

Scrotum, labial folds, or inguinal regions, normal or only slightly subnormal size

Puberty

Partial virilization without gynecomastia, discrepancy between reduced penis size and normal testicular growth

Hormonal diagnosis

Elevated serum LH levels, normal or slightly elevated FSH and low T levels with normal levels of T precursors in relation to T

Gender role

Male

LHCGR gene location

2p21

Molecular defect

Variants which confer partial inactivation of LHCGR

Treatment

Repair of the hypospadias, testosterone replacement at pubertal age

Outcome

Male gender role and behavior, possible fertility under treatment

 

46,XY DSD Due to Enzymatic Defects in Testosterone Synthesis  

 

Six enzymatic defects that alter the normal synthesis of testosterone have been described to date (Figure 10). Three of them are associated with defects in cortisol synthesis leading to congenital adrenal hyperplasia. All of them present an autosomal recessive mode of inheritance and genetic counseling is mandatory since the chance of recurring synthesis defects among siblings is 25%.

Figure 10. Standard steroidogenesis and alternative pathway to DHT synthesis.

DEFECTS IN ADRENAL AND TESTICULAR STEROIDOGENESIS  

 

Adrenal hyperplasia syndromes are examples of hypoadrenocorticism or mixed hypo- and hyper cortico-adrenal steroid secretion. Synthesis of cortisol or both cortisol and aldosterone are impaired. When cortisol production is impaired, there is a compensatory increase in ACTH secretion. If mineralocorticoid production is impeded, there is a compensatory increase in renin-angiotensin production. These compensatory mechanisms may return cortisol or aldosterone production to normal or near normal levels, but at the expense of excessive production of precursors that can cause undesirable hormonal effects.

 

Lipoid Congenital Adrenal Hyperplasia due to Deficiency of the Steroidogenic Acute Regulatory Protein (StAR)

 

StAR is a mitochondrial phosphoprotein which facilitates the influx of cholesterol from the outer to the inner mitochondrial membrane for the subsequent action of the P450scc enzyme  (212).

StAR is encoded by the STAR gene and its deficiency leads to congenital lipoid adrenal hyperplasia (CLAH),  the most severe form of congenital adrenal hyperplasia (213) . Lipoid adrenal hyperplasia is rare in Europe and America, but it is thought to be the second most common form of adrenal hyperplasia in Japan where 1 in 300 individuals carries the p.Q258X variant (214).

 

Affected subjects are phenotypic females irrespective of gonadal sex or sometimes have slightly virilized external genitalia with or without cryptorchidism, underdeveloped internal male organs and an enlarged adrenal cortex, engorged with cholesterol and cholesterol esters (215). Adrenal steroidogenesis deficiency leads to salt wasting, hyponatremia, hyperkalemia, hypovolemia, acidosis, and death in infancy, although patients can survive to adulthood with appropriate mineralocorticoid- and glucocorticoid-replacement therapy (215).

 

Hormonal diagnosis is based on high ACTH and renin levels and the presence of low levels of all glucocorticoids, mineralocorticoids, and androgens.

 

The disease was firstly attributed to P450scc deficiency, but most of the cases studied through molecular analysis showed an intact P45011A gene and its RNA (216). Since StAR is also required for the conversion of cholesterol to pregnenolone, molecular studies were performed in StAR gene and variants were found in most of the affected patients (217) Congenital lipoid adrenal hyperplasia (LCAH) in most Palestinian cases is caused by a founder c.201_202delCT variant causing premature termination of the StAR protein (217) Histopathological findings of excised XY gonads included accumulation of fat in Leydig cells since 1 yr. of age, positive placental alkaline phosphatase and octamer binding transcription factor (OCT4) staining indicating a neoplastic potential (217).

 

A two-hit model has been proposed by Bose et al. (216) as the pathophysiological explanation for LCAH. In response to a stimulus (e.g., ACTH), the normal steroidogenic cell recruits cholesterol from endogenous synthesis, stored lipid droplets or low-density lipoprotein-receptor mediated endocytosis.

 

Subsequently StAR promotes the cholesterol transport from the outer to the inner mitochondrial membrane in which cholesterol is further processed to pregnenolone. In cells with mutant StAR (first hit), there is no rapid steroid synthesis, but still some StAR-independent cholesterol flows into the mitochondria, resulting in a low level of steroidogenesis. Due to increased steroidogenic stimuli in response to inadequately low steroid levels, additional cholesterol accumulates. Massive cholesterol storage and resulting biochemical reactions eventually destroy all steroidogenic capacity (second hit) (217). This two-hit model has been confirmed by clinical studies (218) as well as StAR knockout mice research (219).

 

The human STAR gene is localized on chromosome 8p11.2 and consists of seven exons (220). It is translated as a 285-amino acid protein including a mitochondrial target sequence (N terminal 62 amino acids), which guides StAR to the outer mitochondrial membrane and a cholesterol binding site, which is located at the C-terminal region. In vitro studies revealed that StAR protein lacking the N terminal targeting sequence (N-62 StAR) can still stimulate steroidogenesis in transfected COS-1 cells, whereas variants in the C-terminal region led to severely diminished or absent function (221-223). Most of the STAR gene variants associated with LCAH are located in the C-terminal coding region between exons 5 and 7 StAR related lipid transfer (START) domain (224). Mild phenotype of lipoid CAH is a recognized disorder caused by StAR variants that retain partial activity (225). Affected males can present with adrenal insufficiency resembling autoimmune Addison disease with micropenis or normal development with hypergonadotropic hypogonadism (224,225). More than 40 StAR variants causing classic lipoid CAH have been described  (217,226,227), but very few partial loss-of-function variants have been reported (224-226). Therefore, there is a broad clinical spectrum of StAR variants, however, the StAR activities in vitro correlate well with clinical phenotypes (228).

 

Three 46,XY patients with the homozygous p.R188C STAR variant causing primary adrenocortical insufficiency without atypical genitalia were reported (229). Patients with nonclassical lipoid CAH may present with male genitalia and preserved testicular function (230).

 

Table 8. Phenotype of 46,XY Subjects with StAR Deficiency

Inheritance

Autosomal recessive

External genitalia

Female

Micropenis (mild form)

Müllerian duct derivatives

Absent

Wolffian duct derivatives

Absent -> hypoplastic

Testes

Small size

Clinical Features

Early adrenal insufficiency; no pubertal development; hypergonadotropic hypogonadism

Hormonal diagnosis

Elevated ACTH and renin levels; low levels of all glucocorticoids, mineralocorticoids, and androgens

Gender role

Female

Male (mild form)

STAR gene location

8p11.2

Molecular defect

Inactivating variants in STAR

Treatment

Early gluco- and mineralocorticoid replacement; estrogen replacement at pubertal age

Outcome

Infertile, female or male gender role and behavior

 

Deficiency of P450 Side Chain Cleavage Enzyme (P450scc) Due to Variants in CYP11A1

 

The first step in the conversion of cholesterol to hormonal steroids is hydroxylation at carbon 20, with subsequent cleavage of the 20-22 side chain to form pregnenolone. In steroidogenic tissues, such as adrenal cortex, testis, ovary, and placenta, this is the initial and rate-limiting step in steroidogenesis. This reaction, known as cholesterol side-chain cleavage, is catalyzed by a specific cytochrome P450 called P450scc or CYP11A1 encoded by the CYP11A1 gene (231).

 

A number of patients with CYP11A1 variants have now been described (232-235), including late-onset non-classical forms secondary to variants that retain partial enzyme activity (236,237). Clinically, these patients are indistinguishable from those with lipoid CAH, but none of them present enlarged adrenals that characterize lipoid CAH.

 

Analyzing infants with adrenal failure and disorder of sexual differentiation compound heterozygous variants in CYP11A1 have been identified, recognizing that this disorder may be more frequent than originally thought. The phenotypic spectrum of P450scc deficiency ranges from severe loss-of-function variants associated with prematurity, complete under androgenization, and severe early-onset adrenal failure, to partial deficiencies found in children born at term with mild masculinization and later-onset adrenal failure (236,237).

 

3β-Hydroxysteroid Dehydrogenase type II Deficiency

 

3β-hydroxysteroid dehydrogenase 2 (3βHSD2) deficiency is a rare form of congenital adrenal hyperplasia (CAH), with fewer than 200 cases reported in the world literature.

 

3β-HSD converts 3β-hydroxy 5 steroids to 3-keto 4 steroids and is essential for the biosynthesis of mineralocorticoids, glucocorticoids and sex steroids Two forms of the enzyme have been described in man: the type I enzyme which is expressed in placenta and peripheral tissues such as the liver and skin, and type II that is the major form expressed in the adrenals and gonads (238). The two forms are very closely related in structure and substrate specificity, though the type I enzyme has higher substrate affinities and a 5-fold greater enzymatic activity than type II (238).

 

Male patients with 3β-HSD type II deficiency present with atypical external genitalia, characterized by microphallus, proximal hypospadias, bifid scrotum and a blind vaginal pouch associated or not with salt loss (239,240). Precocious pubarche and gynecomastia at pubertal stage is a common phenotype in 3β-HSD type II deficiency (241).

 

Serum levels of Δ-5 steroids such as pregnenolone, 17OHpregnenolone (17OHPreg), DHEA, DHEAS are elevated and basal levels of 17OHPreg and 17OHPreg/17OHP ratio are the best markers of this deficiency in both prepubertal and postpubertal stage. Δ-4 steroids are slightly increased due to the peripheral action of 3β-HSD type I enzyme but the ratio of Δ-5/Δ-4 steroids is elevated. Cortisol secretion is reduced but the response to exogenous ACTH stimulation varies from decreased (more severe deficiency) to normal. At adult age, affected males can reach normal or almost normal levels of testosterone due to the peripheral conversion of elevated Δ-5 steroids by 3β-HSD type I enzyme and also due to testicular stimulation by the high LH levels (242).

 

The human genome encodes two functional 3βHSD genes on chromosome 1p13.1. The HSD3B2 gene is expressed in adrenal and gonads and consists of four exons coding for a 372 amino acid protein (243). To date, around 40 variants in the HSD3B2 gene have been described. Most of them are base substitutions, and they are located especially at the N-terminal region of the protein. The amino acids A10, A82, P222 and T259 could be considered a hotspot since different variants were reported in these HSD3B2 positions.

 

Variants abolishing 3β-HSD type II activity lead to congenital adrenal hyperplasia (CAH) with severe salt-loss (244). Variants that reduce, but do not abolish type II activity ( > 5% of wild type 3βHSD2 activity in vitro) lead to CAH with mild or no salt-loss, which in males is associated with 46,XY DSD due to the reduction in androgen synthesis (241,242,245,246). Male subjects with 46,XY DSD due 3β-HSD type II deficiency without salt loss showed clinical features in common with the deficiencies of 17β-HSD3 and 5α-reductase 2.

 

Most of the patients were raised as males and kept the male social sex at puberty. In one Brazilian family, two cousins with 46,XY DSD due to 3β-HSD type II deficiency were reared as females; one of them was underwent orchiectomy in childhood and kept the female social sex; the other did not undergo orchiectomy at childhood and changed to male social sex at puberty (246).

 

There is little data on the outcomes of 3β-HSD type II deficiency. A mixed longitudinal and cross-sectional study from a single Algerian center reported 14 affected subjects (8 females) with pathogenic variants in HSD3B2 gene (247). Premature pubarche was observed in four patients (3F:1M). Six patients (5F:1M) entered puberty spontaneously, aged 11 (5-13) years in 5 girls and 11.5 years in one boy. Testicular adrenal rest tumors were found in three boys. Four girls reached menarche at 14.3 (11-14.5) years, with three developing adrenal masses and polycystic ovary syndrome (PCOS), with radiological evidence of ovarian adrenal rest tumor in one. The median IQ was 90 (43-105), >100 in only two patients and <70 in three of them (247).

 

Table 9. Phenotype of 46,XY Subjects with 3β-HSD Type II Deficiency

Inheritance

Autosomal recessive

External genitalia

Atypical (proximal hypospadias, bifid scrotum, urogenital sinus), precocious pubarche

Müllerian derivatives

Absent

Wolffian duct derivatives

Normal

Testes

Well developed; generally topic

Clinical features

Adrenal insufficiency or not in infancy; virilization at puberty with or without gynecomastia

Hormonal diagnosis

Elevated basal and ACTH-stimulated 17OHPreg and 17OHPreg/17OHP ratio

Gender role

Male; female to male

HSD3B2 gene location

1p13.1

Molecular defect

Inactivating variants in HSD3B2

Treatment

Glucocorticoid replacement along with mineralocorticoids in salt-losing form; at puberty variable necessity for testosterone replacement

Outcome

Variable spermatogenesis; fertility possible by in vitrofertilization

 

Combined 17-Hydroxylase and C-17-20 lyase deficiency

 

CYP17 is a steroidogenic enzyme that has dual functions: hydroxylation and lyase. It is located in the fasciculata and reticularis zone of the adrenal cortex and gonadal tissues. The first activity results in hydroxylation of pregnenolone and progesterone at the C(17) position to generate 17α-hydroxypregnenolone and 17α-hydroxyprogesterone, while the second enzyme activity cleaves the C(17)-C(20) bond of 17α-hydroxypregnenolone and 17α-hydroxyprogesterone to form dehydroepiandrosterone and androstenedione, respectively. The modulation of these two activities occurs through cytochrome b5, necessary for lyase activity (248).

 

Deficiency of adrenal 17-hydroxylation activity was first demonstrated by Biglieri et al. (249). The phenotype of 17-hydroxylase deficiency in most of the male patients described is a female-like or slightly virilized external genitalia with blind vaginal pouch, cryptorchidism and high blood pressure, usually associated with hypokalemia. New in 1970, reported the first affected patient with atypical genitalia which was assigned to the male sex (250). The 17-hydroxylase deficiency is the second most common cause of CAH in Brazil (251).

 

At puberty, patients usually present sparse axillary and pubic hair. Male internal genitalia are hypoplastic and gynecomastia can appear at puberty. Most of the male patients were reared as females and sought treatment due to primary amenorrhea or lack of breast development. Genetic female patients may also be affected and present normal development of internal and external genitalia at birth and hypergonadotropic hypogonadism and amenorrhea at post pubertal age; enlarged ovaries at adult age and infarction from twisting can occur (252,253). These patients do not present signs of glucocorticoid insufficiency, due to the elevated levels of corticosterone, which has a glucocorticoid effect. The phenotype is similar to 46,XX or 46,XY complete gonadal dysgenesis and the presence of systemic hypertension and absence of pubic hair in post pubertal patients suggests the diagnosis of 17-hydroxylase deficiency (254).

 

Serum levels of progesterone, corticosterone, and 18-OH-corticosterone are elevated, while aldosterone, 17-OH-progesterone, cortisol, androgens and estrogens are decreased. Martin et al, performed a clinical, hormonal, and molecular study of 11 patients from 6 Brazilian families with the combined 17-alpha-hydroxylase/17,20-lyase deficiency phenotype (255). All patients had elevated basal serum levels of progesterone and suppressed plasma renin activity. The authors concluded that basal progesterone measurement is a useful marker of P450c17 deficiency and suggest that its use should reduce the misdiagnosis of this deficiency in patients presenting with male DSD, primary or secondary amenorrhea, and mineralocorticoid excess syndrome.

 

Excessive production of deoxycorticosterone and corticosterone results in systemic hypertension, suppression of renin levels and inhibition of aldosterone synthesis. The CYP17A1 gene, which encodes the enzymes 17-hydroxylase and 17-20 lyase, is a member of a gene family within the P450 supergene family and is mapped at 10q24.3 (254) (256). Several variants in the CYP17A1 gene have been identified in patients with both 17-hydroxylase and 17,20 lyase deficiencies (252,253,257). Four homozygous variants, p.A302P, p.K327del, p.E331del and p.R416H, were identified by direct sequencing of the CYP17A1 gene. Both P450c17 activities were abolished in all the mutant proteins but the mutant proteins were normally expressed, suggesting that the loss of enzymatic activity is not due to defects of synthesis, stability, or localization of P450c17 proteins (257).

 

Glucocorticoid replacement for hypertension management, gonadectomy and estrogen replacement at puberty for patients reared in the female social sex are indicated. In male patients, androgen replacement is usually necessary since they present very low levels of testosterone. These patients are very sensitive to glucocorticoids and low doses of dexamethasone (0.125-0.5 mg at night) are sufficient to control blood pressure. In some patients, however, estrogens might aggravate hypertension. The control of blood pressure can be initially achieved by salt restriction although mineralocorticoid antagonists might be necessary (257).

 

Table 10. Phenotype of 46,XY Subjects with 17a-Hydroxylase and 17,20-Lyase Deficiency

Inheritance

Autosomal recessive

External genitalia

Female like --> atypical

Müllerian duct derivatives

Absent

Wolffian duct derivatives

Hypoplastic --> normal

Testes

Intra-abdominal or inguinal

Clinical features

Low renin hypertension; absent or slight virilization at puberty; gynecomastia

Hormonal diagnosis

Elevated progesterone, DOC, corticosterone; low plasma renin activity low cortisol not stimulated by ACTH

Gender role

Female in most patients

CYP17 gene location

10q24.3

Molecular defect

Variants in CYP17A1 gene

Treatment

Repair of sexual ambiguity; glucocorticoid and estrogen or testosterone replacement according to social sex

Outcome

Female behavior, infertility

 

Cytochrome P450 Reductase (POR) Deficiency (Electron Transfer Disruption)

 

The apparent combined P450C17 and P450C21 deficiency is a rare variant of congenital adrenal hyperplasia, first reported by Peterson et al in 1985 (258). Affected girls and boys are born with atypical genitalia, indicating intrauterine androgen excess in females and androgen deficiency in males. Boys and girls can also present with skeletal malformations, which in some cases resemble a pattern seen in patients with Antley-Bixler syndrome. Findings of biochemical investigations of urinary steroid excretion in affected patients have shown accumulation of steroid metabolites, indicating impaired C17 and C21 hydroxylation, suggesting concurrent partial deficiencies of the 2 steroidogenic enzymes, P450C17 and P450C21. However, sequencing of the genes encoding these enzymes showed no variants, suggesting a defect in a cofactor that interacts with both enzymes. POR is a flavoprotein that donates electrons to all microsomal P450 enzymes, including the steroidogenic enzymes P450c17, P450c21 and P450aro (259). Shephard et al. (1989) isolated and sequenced cDNA clones that encode the rat and human NADPH-dependent cytochrome P-450 reductase and located the human gene at 7q11.2 (260).

 

The underlying molecular basis of congenital adrenal hyperplasia with apparent combined P450C17 and P450C21 deficiency was defined in 3 patients, who were compound heterozygotes for variants in POR (259,260). Antley-Bixler syndrome is characterized by craniosynostosis, severe midface hypoplasia, proptosis, choanal atresia/stenosis, frontal bossing, dysplastic ears, depressed nasal bridge, radio-humeral synostosis, long bone fractures, femoral bowing, phalangeal malformation (arachno-/campto-/clinodactyly, brachy-tele-phalanges, rocker bottom feet) and urogenital abnormalities (259). The occurrence of genital abnormalities in patients with Antley-Bixler syndrome, especially females was reported in 2000 (261). In a recent large survey of patients with Antley-Bixler syndrome, it was demonstrated that individuals with an Antley-Bixler-like phenotype and normal steroidogenesis have FGFR2 variants, whereas those with atypical genitalia and altered steroidogenesis have POR deficiency (262). The skeletal malformations observed in many, but not all patients with POR deficiency, are thought to be due to disruption of enzymes involved in sterol synthesis, 14α-lanosterol demethylase (CYP51A1) and squalene epoxidase, and disruption of retinoic acid metabolism catalyzed by CYP26 isoenzymes that depend on electron transfer from POR (263).

 

Pubertal presentations in females with congenital POR deficiency were described. Incomplete pubertal development and large ovarian cysts prone to spontaneous rupture were the predominant findings in females (264).The ovarian cysts may be driven not only by high gonadotropins but possibly also by impaired CYP51A1-mediated production of meiosis-activating sterols due to mutant POR. In the two boys evaluated, pubertal development was more mildly affected, with some spontaneous progression. These findings may suggest that testicular steroidogenesis may be less dependent on POR than adrenal and ovarian steroidogenesis (265).

 

Table 11. Phenotype of 46,XY Patients with POR Deficiency

Inheritance

Autosomal recessive

External genitalia

Atypical

Müllerian duct derivatives

Normally developed

Wolffian duct derivatives

Normally developed

Testes

Well developed, frequent cryptorchidism

Hormonal diagnosis

Low T and cortisol and elevated basal ACTH, Prog and 17OHP

POR gene location

7q11.2

Molecular defect

Inactivating variants of POR gene

Puberty

Spontaneous pubertal development in males

Gender role

Male

Treatment

Repair of sexual ambiguity; glucocorticoid replacement and estrogen or testosterone replacement according to social sex

Outcome

Puberty development, fertility?

 

DEFECTS IN TESTICULAR STEROIDOGENESIS   

 

Three defects in testosterone synthesis that are not associated with adrenal insufficiency have been described: CYP17A1 deficiency, cytochrome B5 deficiency and 17-β-HSD3 deficiency

 

CYP17A1 Deficiency

 

Human male sexual differentiation requires production of fetal testicular testosterone, whose biosynthesis requires steroid 17,20-lyase activity. The existence of true isolated 17,20-lyase deficiency has been questioned because 17-α-hydroxylase and 17,20-lyase activities are catalyzed by a single enzyme and because combined deficiencies of both activities were found in functional studies of the variant found in a patient thought to have had isolated 17,20-lyase deficiency (266,267). Later, clear molecular evidence of the existence of isolated 17,20 desmolase deficiency was demonstrated (268).

 

The patients present atypical genitalia with micropenis, proximal hypospadias and cryptorchidism. Gynecomastia Tanner stage V can occur at puberty (268).

 

Elevated serum levels of 17-OHP and 17-OHPreg, with low levels of androstenedione, dehydroepiandrosterone and testosterone, are described. The hCG stimulation test results in a slight stimulation in androstenedione and testosterone secretion with an accumulation of 17-OHP and 17-OHPreg.

 

The CYP17A1 gene of two Brazilian 46,XY DSD patients with clinical and hormonal findings indicative of isolated 17,20-lyase deficiency, since they produce cortisol normally, were studied. Both were homozygous for missense variants in CYP17A1 (268). When expressed in COS-1 cells, the mutants retained 17α-hydroxylase activity and had minimal 17,20-lyase activity. Both variants alter the electrostatic charge distribution in the redox-partner binding site, so that the electron transfer for the 17,20-lyase reaction is selectively lost (268).

 

Table 12. Phenotype of 46,XY Subjects with 17,20 Lyase Deficiency

Inheritance

Autosomal recessive

External genitalia

Atypical (proximal hypospadias, bifid scrotum, urogenital sinus)

Müllerian derivatives

Absent

Wolffian ducts derivatives

Hypoplastic --> normal

Testes

In the inguinal region, small size

Clinical features

Gynecomastia variable; poor virilization at puberty

Hormonal diagnosis

Elevated 17OHP and 17OHP/A ratio after hCG stimulation and decreased A and T levels;

Gender role

Male or female

CYP17 gene location

10q24.3

Molecular defect

Variants in the redox partner binding site of CYP17A1 enzyme

Treatment

Repair of hypospadias and gynecomastia; testosterone replacement at pubertal age

Outcome

Male or female behavior

 

Cytochrome B5 deficiency (Allosteric Factor for P450c17 and POR Interaction)

 

In 1986, Hegesh et al described a 46,XY DSD patient with type IV hereditary methemoglobinemia (269). The patient had a 16-bp deletion in the cytochrome b5 mRNA leading to a new in-frame termination codon and a truncated protein. The etiology of 46,XY DSD in this patient was attributed to the cytochrome b5 defect since cytochrome b5 acts as an allosteric factor, promoting the interaction of. P450c17 and POR favoring 17,20 lyase reactions (270).

 

Two homozygous variants in CYB5 in 46,XY DSD patients with elevated methemoglobin levels but without clinical phenotype of methemoglobinemia were reported (269).

 

46,XY DSD due to 17β-HSD 3 Deficiency

 

This disorder consists of a defect in the last phase of steroidogenesis when androstenedione is converted to testosterone and estrone to estradiol. This disorder was described by Saez and his colleagues (271) and is the most common disorder of androgen synthesis, reported from several parts of the world (272,273).

 

There are 5 steroid 17β-HSD enzymes that catalyze this reaction (274) and 46,XY DSD results from variants in the gene encoding the 17β-HSD3 isoenzyme (275). Patients present female-like or atypical genitalia at birth, with the presence of a blind vaginal pouch, intra-abdominal or inguinal testes and epididymis, vasa deferentia, seminal vesicles and ejaculatory ducts. Most affected males are raised as females (276,277), but some have less severe defects in virilization and are raised as males (274). Virilization in subjects with 17β-HSD3 deficiency occurs at the time of expected puberty. This late virilization is usually a consequence of the presence of testosterone in the circulation because of the conversion of androstenedione to testosterone by some other 17β-HSD isoenzyme (presumably 17β-HSD 5) in extra-gonadal tissue and, occasionally, of the secretion of testosterone by the testes when levels of LH are elevated in subjects with some residual 17β-HSD3 function (274,277). However, the discrepancy between the failure of intrauterine masculinization and the virilization that occurs at the time of expected puberty is poorly understood. A limited capacity to convert androstenedione into testosterone in the fetal extragonadal tissues may explain the impairment of virilization of the external genitalia in the newborn. Bilateral orchiectomy resulted in a clear reduction of androstenedione levels indicating that the main origin of this androgen is the testis (278).  46,XY DSD phenotype is sufficiently variable in 17β-HSD3 deficiency to cause problems in accurate diagnosis, particularly in distinguishing it from partial androgen insensitivity syndrome  (276,279).

 

Laboratory diagnosis is based on elevated serum levels of androstenedione and estrone and low levels of testosterone and estradiol resulting in elevated androstenedione/testosterone and estrone/ estradiol ratios or low (or low testosterone/androstenedione and estradiol/estrone ratios) indicating impairment in the conversion of 17-keto into 17-hydroxysteroids. Testosterone/Androstenedione ratio of 0.4±0.2 was found in prepubertal patients with 17β-HSD3 deficiency after hCG stimulation. Based on these data, a T/A ratio below <0.8 is suggestive of 17β-HSD3 deficiency (272). At the time of expected puberty, serum LH and testosterone levels rise in all affected males and testosterone levels may reach the normal adult male range (277,278).

 

Pitfalls in the hormonal diagnosis of 17β-HSD3 deficiency had been reported in the literature. Two of the fourteen cases of 17β-HSD3 deficiency reported from the UK database had a T/A ratio > 0.8 (276). Both patients were from a consanguineous pedigree, with two affected sisters (both assigned in the female gender) and one nephew. The former patient had atypical genitalia with proximal hypospadias and was assigned as male. The hCG test was performed at 2 years and 2 months of age, respectively, resulting in a T/A ratio of 3.4 and 1.5. Two other patients with atypical genitalia, who were also assigned in the female social sex, were evaluated at 5 months and 9.2 year of age, respectively (280). After the hCG stimulation test, there was a clear elevation of serum testosterone (measured by HPLC tandem mass spectrometry) with a small increase of the androstenedione levels resulting in a high T/A ratio (2.47 and 2.27 respectively). Sequencing of the HSD17B3 gene identified deleterious molecular defects in both alleles in both patients. The possible explanation for the normal T/A ratio in these 4 children is the individual and temporal variability in the HSD17B isoenzymes activity (280).

 

The disorder is due to homozygous or compound heterozygous variants in the HSD17B3 gene which encodes the 17β-HSD3 isoenzyme. Up to now, almost 40 variants in the HSD17B3 gene have been reported. These include missense, nonsense, exonic deletion, duplication and intronic splice site variants (274,277). Although allelic variants have been described throughout HSD17B3, a variant cluster region was identified in the exon 9. The 17β-HSD3 activity was completely eliminated in the majority of the HSD17B3 variants (276). Outside exon 9, the most frequent site of variant in HSD17B3 gene is the R80 in exon 3, which primarily disrupts the binding of the NADPH cofactor to the protein. The p.R80Q variant has been found in Palestinian, Brazilian, and Turkish families (281).

 

Most patients are raised as girls during childhood. Change to male gender role behavior at puberty has been frequently described in individuals with this disorder who were reared as females (282-285), including members of a large consanguineous family in the Gaza strip (286). In a review of all adult patients with 46,XY DSD due to 17β-HSD3 deficiency reared as female and not castrated during childhood reported until now, we found that 30 of them (61%) kept the female gender and 19 of them (39%) changed to male gender (277).

 

After a histological analysis of testicular tissue stained with hematoxylin-eosin from 40 reported cases of 46,XY patients with 17β-HSD3 deficiency, the prevalence of germ cell tumor was 5%, which is lower than the estimated GCT risk for some 46,XY DSD etiologies (287-289). However, the maintenance of the testes in male patients is safe if the testes can be positioned into the scrotum (277,290).

 

Table 13. Phenotype of 46,XY Patients with 17β-HSD 3 Deficiency

Inheritance

Autosomal recessive

External genitalia

Atypical, frequently female-like at birth

Müllerian duct derivatives

Absent

Wolffian duct derivatives

Normally developed

Testes

Well developed, frequent cryptorchidism

Hormonal diagnosis

Low T and elevated basal and hCG-stimulated A and A/T ratio

HSD17B3 gene location

9q22

Molecular defect

Inactivating variants of HSD17B3

Puberty

Virilization at puberty; variable gynecomastia

Gender role

Most patients keep the female social sex; some change to male social sex

Treatment

Repair of sexual ambiguity; estrogen or testosterone replacement according to social sex

Outcome 

Male or female gender identity; in males’ fertility possible by in vitro fertilization

 

ALTERNATIVE PATHWAY TO DHT SYNTHESIS

 

46,XY DSD Due to 3α-Hydroxysteroid Dehydrogenase Deficiency (AKR1C2 and AKR1C4 Defects)

 

Back in 1972, the molecular analysis of  46,XY DSD due to isolated 17,20-lyase deficiency patients failed to find variants in the CYP17A1 (248). However, the hormonal data was inconsistent with other adrenal enzymatic deficiencies. Therefore, the alternative or backdoor pathway was considered to explain the etiology of the DSD in these patients. The backdoor pathway was firstly described in marsupials and is remarkable for having both reductive and oxidative 3α-HSD steps: the reductive reaction converts 17-OH-dihydroprogesterone (17OH-DHP) into 17OH-allopregnanolone (17OH-Allo), and the oxidative reaction converts androstanediol into DHT (291,292) (Figure 6). Therefore, synthesis of dihydrotestosterone (DHT) occurs without the intermediacy of DHEA, androstenedione or testosterone (291). All the human genes participating in the backdoor pathway have not been identified, however it has been thought that the reductive 3α-HSD activity can be catalyzed by an aldo-keto reductase called AKR1C2 (293), as well as by other enzyme, such as the oxidative 3α-HSD activity by 17β-HSD6, also called as RoDH (294)and possibly by AKR1C4 (295).

 

The first reported cases with isolated 17,20 lyase deficiency from 1972 (266) were found to carry variants in two aldo-keto reductases, AKR1C2 and AKR1C4 which catalyze 3α-hydroxysteroid dehydrogenase activity. The two affected 46,XY females were compound heterozygotes for AKR1C2 variants, the p.I79V/H90Q and p.I79V/N300T. However, the mutant AKR1C2 enzymes retained 22-82% of wild-type activity in vitro analysis suggesting that another gene might be involved (248). Analysis of AKR1C cDNA found that AKR1C4 was spliced incorrectly and gene sequencing displayed an intronic variant 106 bases upstream from exon 2 that caused this exon skipping. So, in this family, a digenetic inheritance was found to impair testicular synthesis of DHT during prenatal life (296).

 

AKR1C2 is abundantly expressed in the fetal testis, but minimally expressed in the adult testis; on the other hand, the AKR1C4 was found in fetal and adult testes at lower levels (293). Therefore, it appears that both AKR1C2 and AKR1C4 participate in the backdoor pathway to DHT in the fetal testis, and that molecular defects in these genes appear to cause incomplete male genital development (297). However, the relative roles of these two AKR1C enzymes remain unclear and testosterone levels at adult age are not available in these patients (298).

All findings described above, which substantially advanced our understanding of the underlying mechanisms of male sexual differentiation, illustrate the importance of detailed studies of rare 17,20 lyase deficiency patients.

 

46,XY DSD DUE TO DEFECTS IN TESTOSTERONE METABOLISM

 

5α-Reductase Type 2 Deficiency

 

A condition named pseudo-vaginal perineo-scrotal hypospadias in 46,XY individuals was reported in 1961, in which the phenotype included female-like external genitalia, bilateral testes, and male urogenital tracts with a blind-ending vagina (299). Thereafter, experimental studies showed that the male external genitalia virilization depended on the conversion of testosterone into dihydrotestosterone (DHT), an enzymatic reaction catalyzed by the 5α-reductase enzyme. Further, that enzymatic deficiency was biochemically and clinically reported in 24 individuals from the Dominican Republic and two siblings from North America (300,301). Typically, affected individuals are born with female-like external genitalia but develop clinical and psychological virilization at puberty with no gynecomastia (300). Both studies characterized this syndrome as a genetic condition with an autosomal recessive pattern of inheritance, resulting from the inability to convert testosterone into DHT. Later, two different genes encoding two 5α-reductase isoenzymes were isolated by cloning technology: the 5α-reductase type 1 and 2 (SRD5A1 and SRD5A2) (302). Allelic variants in the SRD5A2 gene were found in two individuals from Papua New Guinea with clinical features of 5α-reductase type 2 deficiency, whereas controls did not have variants in this gene, suggesting that variants in the SRD5A2 were the molecular basis of this condition (303). Further, the SRD5A2 gene was mapped at chromosome 2 (2p23), containing 5 exons and 4 introns, and encoding a 254 amino-acids protein (304).

 

Since then, several SRD5A2 allelic variants have been reported across the whole gene in individuals presenting this particular 46,XY DSD (305). We recently reviewed all 5α-reductase type 2 deficiency cases reported in the literature. We identified 451 cases of 5α-reductase type 2 deficiency from several countries, harboring 121 different SRD5A2 allelic variants (306). These variants have been reported in all exons of this gene, but mainly are located at exons 1 (33%) and at exon 4 (25%). Among the 254 amino acids that make up the SR5A2 protein, we found allelic variants in the SRD5A2 gene in 76 of them (306).

 

Regarding the SRD5A2 allelic variants, most are missense variants, but small deletions, variants at splicing sites, stop codons, small indels (n = 20) and large deletions have also been described. We also identified homozygosity in 70% of the SRD5A2 allelic variants causing 5α-reductase type 2 deficiency (306).

 

Neonatal diagnosis was carried out in 29.7%, whereas the remaining had the 5α-reductase type 2 deficiency diagnosis later in life. Most cases were assigned as female (69.4%), and an association between higher scores of external genitalia virilization (less virilization) and female sex assignment was identified. However, when we divided the cases into those who were diagnosed after and before 1999, the percentage of male sex assignment rose from 26.8% to 42.8%, suggesting a temporal trend pointing toward an increased likelihood of 5α-reductase type 2 deficiency patients being raised as boys (305).

 

Intriguingly, 5α-reductase type 2 deficiency is a condition with no genotype-phenotype correlation (307-309). This observation is based on several 5α-reductase type 2 deficiency families carrying the same genotype but presenting a broad range of external genitalia virilization. However, some SRD5A2 variants are consistent in the way they affect phenotype. It is the case of the p.Arg246Gln variant, which is associated with more external genitalia virilization (304,310-312), and also the case of both p.Gly183Ser and p.Gln126Arg variants, that are consistently reported with more severe external genitalia under-virilization (304,313-315). 

 

The diagnosis is usually made at birth, infancy or at puberty. In the newborn, the features of 46,XY DSD due to 5a-reductase type 2 deficiency overlap with other forms of male DSD such as androgen insensitivity syndrome (partial form) and testosterone synthesis defects (304,316).

 

At puberty or in young adult men, the basal hormonal evaluation demonstrates normal male serum testosterone levels, low or low/normal dihydrotestosterone levels, and elevated or normal serum testosterone to dihydrotestosterone ratio (307). For appropriated use of this ratio, the testosterone levels should be in the post puberal range. Likewise, in prepubertal children, a hCG stimulation to increase serum testosterone levels  is necessary (313). The biggest challenge is the diagnosis in newborns. This difficult largely arises from the fact that even when serum testosterone has undergone a neonatal surge, the ratio of serum testosterone to dihydrotestosterone may be normal, because expression of the 5a-reductase type 1 enzyme can occasionally be higher than expected (317,318).

 

Measurement of dihydrotestosterone is difficult because this steroid is present in very low concentrations and has a high rate of cross-reactions (319). To obtain an accurate dihydrotestosterone measurement, a precise assay must be utilized since serum testosterone levels are higher than dihydrotestosterone levels (about 10-fold). Consequently, the separation of testosterone from dihydrotestosterone is necessary to provide and accurate dihydrotestosterone measurement. Using such methodologies, the testosterone/dihydrotestosterone ratio for 5a-reductase type 2 deficiency hormonal diagnosis is generally over 18 in most cases (320,321). However, the testosterone/dihydrotestosterone ratio for 5a-reductase type 2 deficiency hormonal diagnosis has been debatable.

 

Another approach to 5a-reductase type 2 deficiency diagnosis is the measurement of urinary steroids by gas chromatography– mass spectrometry (GC–MS) to determine the ratio of 5a- to 5ß- reduced steroids in urine. This evaluation is very helpful for the diagnosis in subjects at prepubertal age and in orchiectomized adults. In one review, extremely low ratios of 5a- to 5ß-reduced steroid metabolites in urine were pathognomonic for 5a-reductase type 2 deficiency (322). Based on the challenges on the hormonal diagnosis, genetic analysis of the SRD5A2 gene is recommended to confirm the diagnosis [39,40].

 

The management in subjects with female social sex includes a careful psychological evaluation of gender identity (323). Subsequent management is similar to that in women with other forms of 46,XY DSD (324). Treatment must simulate a normal puberty pattern and low to normal estrogen doses, considering the height, and it should be administered at the age of expected puberty (10 – 12 years old). After complete breast development, adult estrogen doses are maintained continuously. Progesterone replacement is not necessary because these patients do not have a uterus (11). For women with this condition, feminizing genitoplasty is often necessary to provide an adequate vaginal opening, a functional vaginal introitus, full separation between urethral and vaginal orifice and phallic erectile tissue removal (11). Vaginal dilatation to promote vaginal length with acrylic molds is recommended when the patients decide to initiate sexual activity (325). Laparoscopic orchiectomy is recommended for all female patients to avoid virilization and gonadal malignancies. Usually, testosterone replacement is not often necessary for male patients since most retain testes and present adequate testicular function towards puberty (307). However, since the degree of virilization is usually unsatisfactory in male patients, a limited use of intramuscular testosterone or transdermal dihydrotestosterone may be helpful to improve virilization (307,319). Maximum penile length is obtained after 6 months of high dose testosterone therapy (e.g., 500 mg of testosterone cypionate per week) (319). The therapeutic penile response does not result in normal penile length in all individuals, even when initiated during childhood, and the final penile length is still below -2 SD in most patients (326). Surgical treatment consists of orthophalloplasty, scrotoplasty, resection of the vaginal pouch and proximal and distal urethroplasty. Correction of hypospadias is indicated in early childhood (up to two years old) (326).

 

Gender change (from female to male) is common among 5α-reductase type 2 deficiency individuals (327). It occurs in over 50% among those assigned as girls in some series (328). It may change since there is growing evidence suggesting male sex assignment for 5α-reductase type 2 deficiency newborns to avoid gender incongruence and gender dysphoria (329-331). 

 

Regarding long-term follow up in the males from the Sao Paulo cohort, most of these subjects were satisfied with the appearance of the external genitalia and sexual life, although a small penile length made sexual intercourse difficult for some of them (326). Most of the adult males patients get married, and those reared as male report a more satisfactory quality of life than the female patients (332). Among female individuals, most describe a satisfactory sexual life, but none are married or have adopted children (333).

 

Table 14. Phenotype of 46,XY Subjects with 5α-Reductase 2 Deficiency

Inheritance 

Autosomal recessive

External genitalia 

Atypical, small phallus, proximal hypospadias, bifid scrotum, blind vaginal pouch

Müllerian duct derivatives 

Absent

Wolffian duct derivatives 

Normal

Testes

Normal size at inguinal, intra-abdominal region or topic

Puberty

Virilization at puberty, absence of gynecomastia

Hormonal diagnosis 

Increased T/DHT ratio in basal and hCG-stimulation conditions in postpubertal patients and after hCG-stimulation in pre-pubertal subjects. Elevated 5β/5α C21 and C19 steroids in urine in all ages

SRD5A2 gene location

2 p23

Molecular defect 

Inactivating variants in 5RD5A2

Gender role 

Female → male in 60% of the cases

Treatment 

High doses of T and/or DHT for 6 months to increase penis size

Outcome 

Maximum penis size in males after treatment = 9 cm; fertility is possible by in vitro fertilization

 

46,XY DSD DUE TO DEFECTS IN ANDROGEN ACTION

 

Androgen Insensitivity Syndrome

 

Androgen insensitivity syndrome (AIS) is the most frequent etiology of 46,XY DSD individuals (334). The underlying molecular basis of AIS is variants in the androgen receptor gene (AR), which is located on the long arm of the X chromosome (Xq11-12) (335). AIS is an X-linked inherited condition, and up to 30% of AIS cases present de novovariants (334,336). Due to disruptive variants in the AR gene, affected individuals present a broad spectrum of under-virilization, which will depend upon the residual activity of the mutant AR. There are three phenotypes of AIS: complete (typically female external genitalia; CAIS), partial (a wide spectrum of external genitalia under virilization; PAIS) or mild (typically male external genitalia with further gynecomastia and/or infertility; MAIS) (337,338).

 

The AR contains eight exons and encodes a 920 amino-acids protein (10). The AR is composed of three major functional domains: the N-terminal transactivation domain (NTD), a central DNA-binding domain (DBD), a C-terminal ligand-binding domain (LBD), and a hinge region connecting the DBD and LBD (339,340). The main difference between the AR and other steroid receptors is the presence of a longer NTD (341). Exon 1 encodes for the NTD, while exons 2 and 3 encode for the DBD and exons 4-8 encode for the LDB (342). In the presence of androgens, the AR recruits multiple epigenetic coregulators. These co-regulators can be either co-activators or co-repressors and actupon AR influencing DNA binding, nuclear translocation, chromatin remodeling, AR stability and bridging AR with transcriptional machinery (343,344). AR coding region has two polymorphic trinucleotide repeat regions, located at exon 1, the CAG and GGC repeats (345). The length of these repeats can cause human diseases. In general, longer CAG repeats may lead to AR transactivation impairment whereas shorter CAG repeats may enhance the ARtransactivation (346). A high number of CAG repeats (>38) is the molecular cause of Spinal and Bulbar Muscular Atrophy (Kennedy’s disease) (347). This condition is characterized by severe muscular atrophy and a mild AISphenotype, including gynecomastia. On the other hand, shorter CAG repeats are related with increased risk for prostate cancer (348).

 

There are more than 800 variants in the AR gene reported in AIS patients (www.androgendb.mcgill.ca/; HGMD). Most of them are missense variants leading to amino acid substitutions (349). However, small indels, variants at splicing sites, premature stop codons and large deletions were also reported, most of them related to CAIS (350). Despite a well-characterized monogenic condition, AR variants are identified in 90-95% of CAIS, but only in 28-50% of PAIS (334,350). Therefore, the molecular diagnosis of PAIS individuals remains challenging. Advances in molecular biology have been helpful to clarify unusual molecular mechanisms or deep DNA alterations related to AIS. Alterations immediately upstream of the AR were identified in AIS patients without variants in the coding region of the AR, either by promoting aberrant AR transcripts, or disrupting AR expression by the insertion of a large portion of a long-interspersed element retrotransposon, which were proven to cause AIS (351,352). Rare synonymous variants within the encoding region of the AR gene were proven to play a role in AIS by disrupting splicing (353). The sequencing of intronic regions of the AR was able to identify a deep intronic variant leading to pseudo-exon activation in AIS (354). Additionally, studies involving AR variant-negative individuals with AIS revealed the deficiency of the androgen-responsive apolipoprotein D, indicating functional AIS, and epigenetic repression of the AR transcription was reported in a group of AIS variant-negative individuals, a condition defined as AIS type II (355). However, a specific role of certain coregulators in the pathophysiology of AIS is not established yet and the contribution of AR-associated coregulators in AIS remains poorly understood.

 

COMPLETE ANDROGEN INSENSITIVITY SYNDROME

 

Prenatal diagnosis of CAIS is possible and can be suspected based on the discordance between 46,XY karyotype on prenatal fetal sex determination and the identification of a female genitalia at prenatal ultrasound (356). At birth, CAIS individuals present typically female external genitalia. In childhood, the identification of an inguinal hernia in a girl may be a clinical indication of CAIS, since inguinal hernias in girls are rare (357).

 

At puberty, CAIS patients present with complete breast development and primary amenorrhea (358). Pubic hair and axillar hair are sparse in most of them, and Mullerian ducts are often absent in CAIS patients (337).

 

The endocrine evaluation after puberty shows normal or elevated serum testosterone levels and slightly elevated LH levels, whereas FSH levels can be slightly elevated, with normal presence of testosterone precursors (334).

 

Patients with CAIS are assigned and raised as girls and usually present a female gender identity (328,331). Estrogen replacement is recommended to induce puberty if bilateral gonadectomy has been performed before puberty. There is gonadal tumor risk in CAIS patients, but this risk is very low before puberty (359). Therefore, gonadectomy can be postponed because after puberty is complete in CAIS patients (360,361) . An increasing number of adult women with CAIS prefer to decline or delay gonadectomy for several reasons, such as fear of surgery, to avoid estrogen replacement, and expectations for future fertility (362).

 

Table 15. Phenotype of 46,XY Subjects with Complete Androgen Insensitivity Syndrome

Inheritance 

X-linked recessive

External genitalia 

Female

Müllerian duct derivatives 

Absent

Wolffian duct derivatives 

Absent or vestigial

Testes

Inguinal or intra-abdominal, slightly subnormal size

Puberty 

Complete breast development

Hormonal diagnosis

High or normal serum LH and T levels, normal or slightly elevated FSH levels

Gender role 

Female

AR gene location

Xq11-12

Molecular defect 

Pathogenic allelic variants in AR gene

Treatment

Psychological support

Estrogen replacement after gonadectomy. Vaginal dilation for sexual intercourse

Outcome

Female identity, infertility

 

PARTIAL ANDROGEN INSENSITIVITY SYNDROME

 

Patients with PAIS have a broad spectrum of virilization impairment (337). The external genitalia ranged from predominantly female with clitoromegaly and labial fusion to predominantly male with micropenis and hypospadias. Testes are in the inguinal canal or labioscrotal folds or, less frequently, intraabdominal. At puberty, under-virilization and gynecomastia are observed (334). The final height of PAIS individuals is intermediate between the average height for control males and females. In addition, PAIS individuals presented decreased bone mineral density in the lumbar spine compared to controls (363). In male PAIS, gynecomastia is common at puberty which is helpful in the differential diagnosis from other 46,XY DSD etiologies (364).

 

In the endocrine analysis, serum LH levels are in the normal upper range or slightly elevated, and testosterone levels are normal or slightly elevated (334). A definitive diagnosis of PAIS is established by identifying a variant in the ARgene, but AR variants are found only in about 40% of PAIS (350).

 

The sex of rearing is female in half of the cases, and gender change is uncommon in PAIS patients either raised as female or male (328).

 

Estrogen replacement is necessary for female patients to induce adequate puberty since most female PAIS patients undergo gonadectomy in childhood (334,365). For male patients, androgen replacement, either to induce puberty or to enhance virilization post-puberty, is commonly required (11). High doses of intramuscular testosterone preparations or topical DHT can be tried for six months to improve virilization, but it is unnecessary after that (11).

 

If the testes are at the scrotum, gonadectomy is unnecessary in male PAIS individuals. However, bilateral gonadectomy is still recommended for female PAIS due to avoid partial virilization and due to the gonadal malignancy risk (288,337).

 

Table 16. Phenotype of 46,XY Subjects with Partial Androgen Insensitivity Syndrome

Inheritance 

X-linked recessive

External genitalia

Broad spectrum from female with mild clitoromegaly to male with micropenis and/or hypospadias

Müllerian duct derivatives 

Absent

Wolffian duct derivatives 

Broad spectrum from absent or male

Testes

Eutopic, inguinal or intra-abdominal, normal or slightly subnormal size

Puberty 

Gynecomastia

Hormonal diagnosis

High or normal serum LH and T levels, normal or slightly elevated FSH levels

Gender role 

Female or male

AR gene location

Xq11-12

Molecular defect 

Pathogenic variants in AR gene

Treatment

Females: surgical feminization, gonadectomy, replacement with estrogens at the time of puberty, vaginal dilation (if necessary)

Males: hypospadias repair, bifid scrotum; high doses of T or DHT to increase penis size

Outcome 

Infertility, female or male gender role

 

46,XY DSD DUE TO PERSISTENT MÜLLERIAN DUCT

 

Defect in AMH Synthesis or AMH Receptor

 

The development of female internal genitalia in a male individual is due to the incapacity of Sertoli cells to synthesize or secrete anti-Mullerian hormone (AMH) or to alterations in the hormone receptor. Persistent Müllerian duct syndrome (PMDS) phenotype can be produced by a variant in the gene encoding anti-Müllerian hormone or by avariant in the AMH receptor. These two forms result in the same phenotype and are referred to as type I and type II, respectively (366).

 

AMH is a 145,000 MW glycoprotein homodimer produced by Sertoli cells not only during the period when it is responsible for regression of the Müllerian ducts but also in late pregnancy, after birth, and even, albeit at a reduced rate, in adulthood (13,367,368).

 

AMH is a small gene containing 5 exons, located in chromosome19p.13.3 (367) and its protein product acts through its specific receptor type 2 (AMHR2) a serine/threonine kinase, member of the family of type II receptors for TGF-β-related proteins (369).

 

Affected patients present a male phenotype, usually along with bilateral cryptorchidism and inguinal hernia (368). Leydig cell function is preserved, but azoospermia is common due to the malformation of ductus deferens or agenesis of epididymis. When the hernia is surgically corrected, the presence of a uterus, fallopian tubes and the superior part of the vagina can be verified.

 

PMDS is a heterogeneous disorder that is inherited in a sex-limited autosomal recessive manner. variants in AMHgene or AMH receptor 2 gene in similar proportions are the cause of approximately 85% of the cases of PMDS (370,371). In the remaining cases the cause of the persistent Mullerian duct syndrome is unknown (368).

 

Normally, AMH levels are measurable during childhood and decrease at puberty. Patients with AMH gene defects have low AMH levels since birth whereas patients with variants in AMH receptor gene have elevated AMH levels (372).

 

Treatment is directed toward an attempt to assure fertility in males. Early orchiopexy, proximal salpingectomy (preserving the epididymis), and a complete hysterectomy with dissection of the vas deferens from the lateral walls of the uterus are indicated (368,373).

 

CONGENITAL NON-GENETIC 46,XY DSD

 

Maternal Intake of Endocrine Disruptors

 

The use of synthetic progesterone or its analogs during the gestational period has been implicated in the etiology of 46,XY DSD (374). Some hypotheses have been proposed to explain the effect of progesterone in the development of male external genitalia, such as reduction of testosterone synthesis by the fetal testes or a decrease in the conversion of testosterone to DHT due to competition with progesterone (also a substrate for 5α-reductase 2 action). The effect of estrogen use during gestation in the etiology of 46,XY DSD has not been confirmed to date (375). Recently, a study in Japanese subjects supports the hypothesis that homozygosis for the specific estrogen receptor alpha 'AGATA' haplotype may increase the susceptibility to the development of male genital abnormalities in response to estrogenic effects of environmental endocrine disruptors (376).

 

Environmental chemicals that display anti-androgenic activity via multiple mechanisms of action have been identified. They are pesticides, fungicides, insecticides, plasticizers and herbicides. They can work as androgen receptor antagonists like pesticides, or they can decrease mRNA expression of key steroidogenic enzymes and also the peptide hormone insl3 from the fetal Leydig cells, like plasticizers and fungicides (377).

 

Daily exposure to residues of a fungicide (vinclozolin), either alone or in association with a phytoestrogen genistein (present in soy products), induce hypospadias in 41% of mice, supporting the idea that exposure to environmental endocrine disruptors during gestation could contribute to the development of hypospadias (378).

 

Supporting the idea that exposure to a mixture of chemicals can produce greater incidences of genital malformations, Rider et al examined the effects of exposure to a mixture of two chemicals that act as androgen receptor antagonists. They observed that the exposure to vinclozolin (fungicide) alone resulted in a 10% incidence of hypospadias and no vaginal pouch development in male rats, whereas procymidone, another fungicide exposure, failed to generate malformations. However, the combined exposure resulted in a 96% incidence of hypospadias and 54% incidence of vaginal pouch in treated animals. Similar results were observed in phthalate (plasticizer) mixture studies (377).

 

Given that severe alterations of sexual differentiation can be produced in animal laboratory studies, the question arises of what would be expected in exposed humans given that humans are exposed to mixtures of compounds in their environment.

 

Congenital Non-Genetic 46,XY DSD Associated With Impaired Prenatal Growth

 

Despite the multiple genetic causes of 46,XY DSD, around 30-40% of cases remain without diagnosis. Currently, there is a frequent, non-genetic variant of 46,XY DSD characterized by reduced prenatal growth and lack of evidence for any associated malformation or endocrinopathy (379,380). Using the model of monozygotic twins, hypospadias has now been linked to low birth weight (379). We have identified a pair of 46,XY monozygotic twins (identical for 13 informative DNA loci) born at term after an uneventful pregnancy sustained by one placenta who were discordant for genital development (perineal hypospadias versus normal male genitalia) and postnatal growth (low birth weight versus normal birth weight). No evidence for uniparental disomy was found (381). The most plausible cause of incomplete male differentiation associated with early-onset growth failure is a post-zygotic, micro-environmental factor since different DNA methylation patterns associated with silencing of genes important for sex differentiation has been shown (382).

 

Additionally, three cohorts of undetermined 46,XY DSD report around 30% of cases as associated with low birth weight, indicating that adverse events in early pregnancy are frequent causes of congenital non-genetic 46,XY DSD (383-385).

 

A genetic defect that clarifies the etiology of hypospadias was not found in 41 non-syndromic SGA children, supporting the hypothesis that multifactorial causes, new genes, and/or unidentified epigenetic defects may have an influence in this condition (385).

  

46,XY OVOTESTICULAR DSD

 

There are rare descriptions of 46,XY DSD patients with well characterized ovarian tissue with primordial follicles and testicular tissue, a condition that is histologically characterized 46,XY ovo-testicular DSD (386). The differential diagnosis of 46,XY ovo-testicular DSD with partial 46,XY gonadal dysgenesis should be performed considering that in the latter condition there are descriptions of dysgenetic testes with disorganized seminiferous tubules and ovarian stroma with occasional primordial follicles in the first years of life (46). To our knowledge there are no descriptions of an adult patient with 46,XY ovo-testicular DSD with functioning ovarian tissue, as occurs in all 46,XX ovo-testicular DSD. Therefore, the diagnosis of 46,XY ovo-testicular DSD is debatable.

 

NON-CLASSIFIED FORMS

 

Hypospadias

 

Hypospadias is one of the most frequent genital malformations in the male newborn and 40% of the cases are associated with other defects of the urogenital system (387). Hypospadias results from an abnormal penile and urethral development that appears to be a consequence of various mechanisms including genetic and environmental factors. It is usually a sporadic phenomenon, but familial cases can be observed, with several affected members (388,389).

 

The presence of hypospadias indicates an intra uterus interference in the correct genetic program of the complex tissue interactions and hormonal action through enzymatic activities or transduction signals. MAMLD1 (mastermind-like domain containing 1) has been reported to be a causative gene for hypospadias (390). It appears to play a supportive role in testosterone production around the critical period for sex development. To date, microdeletions involving MAMLD1 and nonsense and frameshift variants in the gene have been found in 46, XY DSD patients, suggesting that MAMLD1 variants cause 46,XY DSD primarily because of compromised fetal testosterone production, however, its role in the molecular network involved in fetal testosterone production is not known so far (391).

 

The activating transcription factor 3 (ATF3) expression was identified in the developing male urethra. Apparently ATF3 variants may influence the risk of hypospadias (392).

 

By definition, hypospadias is a form of 46,XY DSD and although most of the patients maintain fertility and masculinization at puberty, their testicular function should be assessed to rule out causes such as defects in testosterone synthesis and action, which require hormonal treatment and genetic counseling in addition to surgical treatment.

 

GONADAL TUMOR DEVELOPMENT IN 46,XY DSD PATIENTS

 

Any disturbance in the gonadal development, including testicular descent, increases the risk of developing gonadal malignancies (288). For inherited disturbances in gonadal development or endocrine alterations, patients with 46,XY DSD are at increased risk of developing type II germ cell tumors (GCT) (289). In testicular tissues, GCTs comprise both premalignant conditions, such as germ cell neoplasia in situ (GCNIS) and malignant invasive germ cell tumors, including seminomas and non-seminomas (393).

 

The term GCNIS was introduced in the 2016 WHO classification of urological tumors to define precursor lesions of invasive GCTs, since GCNIS has the potential to develop into several types of GCTs (394,395). GCNIS cells are fetal gonocytes present in the seminiferous tubules arrested during gonadal development that failed to mature into spermatogonia (396). GCNIS are often detected in testicular tissues from 46,XY DSD subjects (397). It is estimated that 50% of GCNIS progress to an invasive GCT in five years (396,398,399).

 

A high risk of GCT is found when sex determination is disrupted at an early stage of Sertoli cell differentiation (due to abnormalities in SRY, WT1, SOX9) (289,397,400). For that reason, specific etiologies of 46,XY DSD (401) have a significant risk factor for GCT development (393). Early Sertoli cell development is also disturbed in patients with 45X/46,XY mosaicism (402). The presence of the well-defined Y chromosome region, known as the gonadoblastoma Y locus (GBY), is a prerequisite for malignant transformation. Among the genes located in the GBY region the testis-specific protein Y (TSPY) seems to be the most significant candidate gene for the tumor-promoting process (288,403). The presence of undifferentiated gonadal tissue containing germ cells that abundantly express TSPY has also been identified as a gonadal differentiation pattern bearing a high risk for GCT development (404). Prolonged expression of OCT3/4 (POU5F1) and the stem cell factor KITL after one year of age are also estimated to play a role in GCNIS/GCT development. Other factors implicated in that risk include MAP3K1 variants in 46,XY patients with gonadal dysgenesis due to MAPK signaling pathway upregulation and loss of androgen receptor function in patients with androgen insensitivity syndrome (289,405). Additionally, gonads at the abdominal region are at higher risk of GCNIS/GCT development than those appropriately positioned (393,406).

 

Unfortunately,  GCNIS/GCT screening is challenging due to a lack of a predictive factor or a biomarker with adequate sensitivity and specificity (407). As far as imaging is concerned, ultrasound (US) is more sensitive than MRI at identifying dysgenetic gonads, but MRI showed better sensitivity and specificity than US at localizing non-palpable gonads (408). However, both imaging techniques are poor at identifying GCNIS/GCT, since MRI failed to identify GCNIS in patients with CAIS and the US only identified one out of ten malignant lesions in 46,XY DSD people (409). There are serum markers that are associated with GCT in non-DSD people, such as alpha-fetoprotein, beta-hCG, and lactate dehydrogenase, but there is poor evidence about how useful they are for GCT screening in 46,XY DSD individuals (410). An interesting perspective for GCT screening are microRNAs (miRNA), since some miRNA clusters are expressed in the presence of GCT (411). For non-DSD people, microRNAs are more sensitive than serum markers and imaging to detect GCT. Noteworthy, GCNIS also expresses some embryonic-type miRNAs (miR-371-3, miR-302, and miR-367) that are also expressed by GCTs (410,412). Therefore, they have the potential to serve as a biomarker even for GCNIS(407).

 

Overall, neoplastic transformation of germ cells in dysgenetic gonads (gonadoblastoma and/or an invasive germ cell tumor) occurs in 20-30% of 46,XY DSD individuals, but the risk varies among 46,XY DSD etiologies (413). Individuals with Denys-Drash syndrome (40%), Frasier syndrome (60%), and gonadal dysgenesis (15 - 35%) have the highest risk of GCNIS/GCT among 46,XY DSD etiologies (413). On the other hand, individuals with CAIS (at prepubertal age) and ovotestis DSD have lower risk of GCNIS/GCT (414). The age matters in the estimation of GCNIS/GCT risk. For example, it is as low as 1.3% in CAIS individuals before puberty, but it can reach 33% thereafter (287,415).

 

For 46,XY DSD subjects, gonadectomy is classically recommended to avoid GCNIS/GCT development, preventing additional therapies and related risks (290). Despite a very effective strategy to avoid GCNIS/GCT, gonadectomy leads to hypogonadism and infertility.

 

Regarding the time for gonadectomy, bilateral gonadectomy should be performed in early childhood in 46,XY DSD patients with gonadal dysgenesis, females with Y chromosome material, and patients with androgen biosynthesis defect, unless the gonad is functional and easily accessible to palpation and imaging studies, which should be performed yearly (11,289). Although data are limited, in the androgen insensitivity syndrome the risk seems to be markedly lower in the complete form before puberty than in the other 46,XY DSD (416). Therefore, gonadectomy can be postponed until puberty is complete in CAIS individuals (417).  Unfortunately, the GCNIS/GCT risk for other causes of 46,XY DSD patients, such as Leydig Cell Hypoplasia and 5 alpha reductase type 2 deficiency has not been estimated yet. 

 

Rarely, gonadal tumors can produce sexual steroids (418). In those cases that are able to produce estrogens, spontaneous breast development may be a clinical sign that suggests the presence of an estrogen-secreting gonadal tumor, and bilateral gonadectomy is indicated even at early childhood, regardless of the 46,XY DSD etiology.

 

Overall, 46,XY DSD patients are at increased risk for gonadal malignancy which seems to be related to 46,XY DSD etiology. While it is clear that prepubertal CAIS patients are at low risk for GCT and 46,XY DSD individuals harboring WT1 variants present a high risk for GCT development, the real GCT risk for other 46,XY DSD etiologies is not that clear (413). In the absence of a reliable predictive factor or biomarker of GCNIS/GCT as well as appropriate recommendations for GCT screening, bilateral gonadectomy will still be recommended for most 46,XY DSD etiologies.

 

FERTILITY IN PATIENTS WITH 46,XY DSD

 

Most 46,XY DSD individuals face infertility due to abnormal gonadal development, endocrine disturbances, anatomical issues, or prophylactic gonadectomy for malignancy risk (419). However, there has been growing evidence showing that fertility is relevant for several 46, XY DSD people, in addition to the possibility of delaying gonadectomy in some 46,XY DSD etiologies (420). In parallel, fertility preservation technologies have been improved in recent years along with a better social perception of non-traditional family structures (421,422).

 

In 46,XY DSD, the fertility potential varies depending on the underlying etiology as well as the severity of the condition (421). In this sense, all options for fertility should be discussed considering the 46,XY DSD etiology or the gonadal structure and internal genitalia in those in whom the 46,XY etiology is unknown (420).

 

For example, individuals with complete gonadal dysgenesis possess uterus, despite lacking gametes (423). Therefore, pregnancy by oocyte donation is an alternative for these patients. On the other hand, male individuals with partial and mild androgen insensitivity often present oligospermia, but biological fertility is possible (334,338).

 

Overall, there is limited literature about fertility potential among 46,XY DSD people. Successful biological fertility was obtained in a man with PAIS after prolonged high-dose testosterone therapy followed by intracytoplasmic sperm injection (424). The possibility of fertility seems to be more frequent among MAIS since there are six cases of successful fertility (338). There are few reported cases of successful pregnancies and live births in men with 5RD2 deficiency, both spontaneous and with assisted reproductive technology (319,425-427). Biological fertility has also been documented in individuals with nonclassical congenital lipoid adrenal hyperplasia, 3b-HSD2 deficiency, and LHCG receptor defect (420). Conversely, there are no reported cases of biological fertility in individuals with classic congenital lipoid adrenal hyperplasia, cytochrome p450 oxidoreductase deficiency, complete CYP17A1 deficiency, 17b-HSD3 deficiency, and CAIS (419,428).

 

To estimate fertility potential, a pilot study evaluated the presence of germ cells and the germ cell density in individuals with several 46,XY DSD etiologies (429). In six patients with CAIS, all presented Sertoli Cell nodules and hyperplasia, but germ cells were detected in areas between nodules. All six patients with mixed gonadal dysgenesis and two with ovo-testicular DSD presented germ cells, and ten out of twelve 46,XY DSD patients with unknown etiology presented germ cells in their gonads. On the other hand, germ cells were not found in any of the patients with either complete or partial gonadal dysgenesis. However, the number of germ cells was inversely correlated with age, suggesting that the gonadectomy delay may decrease fertility potential. It needs to be confirmed by more extensive studies, but it indicates that 46,XY DSD fertility potential may be greater than previously thought.

 

As far as desire for fertility is concerned, a large follow-up study included 1,040 DSD individuals to investigate their fertility preferences (430). The authors reported that 55% of patients expressed a desire to have had fertility treatments in the past or have it in the future, and 40% mentioned that they would like to try new fertility treatment techniques. Additionally, CAIS women reported the possibility of future fertility as one of the reasons to keep their gonads (362).

 

Indeed, fertility preservation has been primarily assessed in oncology to preserve patients' fertility under gonadotoxic treatments (431). In this sense, cryopreservation of postpubertal testicular tissue is helpful to keep fertility potential in patients having gonadectomy or those before gonadotoxic treatment. As an alternative, cryopreservation of immature testicular tissue containing spermatogonial cells or spermatogonial stem cells can be offered to prepubertal patients (432). These techniques could also be considered for 46,XY DSD patients. 

 

In summary, addressing fertility is essential in 46,XY DSD management. The fertility potential must be discussed considering the 46,XY DSD etiology and the patient’s desire. As assisted fertility and preservation techniques improve, these advancements should be offered and accessible to all 46,XY individuals.  

 

46,XY GENDER IDENTITY DISORDERS

 

Transgender Women are characterized by the wish to live as members of the female sex with conviction and consistently and progressively efforts to achieve such state. 46,XY gender identity disorders are more frequent among the male sex, although it also occurs in the female sex. Its first manifestations usually start during childhood. If it has a biological basis is still unknown, but some hormonal alterations during intrauterine life and familial factors before and after birth cannot be ruled out (433).

 

The term used to name men and women who live a relevant incongruence between their gender identity and their inborn physical phenotype has changed over time. The term “trans-sexualism” was coined by Hirschfeld in 1923 and was adopted by the International Classification of Diseases – version 10 (ICD-10). The American Psychiatric Association, in its 4th edition, adopted “gender identity disorder” to define persons who are not satisfied with their biological gender (Association, American Psychiatric. "Diagnostic and statistical manual of mental disorders (2000).

 

Finally, the current classification system of the American Psychiatric Association (DSM-5) replaced the term “gender identity disorder” with “gender dysphoria” and the upcoming version of International Classification of Diseases – version 11 (ICD-11) has proposed the term “gender incongruence” (434).

 

In this chapter we will use the current DSM-5 term, “gender dysphoria”. To refer to male to female gender-dysphoric persons we will use the term transgender woman (American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition - DSM-5; 2013). Therefore, the term “transgender woman” refers to all 46, XY individuals with typical male phenotype who wish to live and be accepted as a female.

 

Higher prevalence of addictions and suicidal thoughts or suicide attempt than those observed in the general population, revealed the need for early care of these patients by health professionals. Among transgender women, total mortality was 51% higher than in the general population, mainly from increased mortality rates due to suicide, acquired immunodeficiency syndrome, cardiovascular disease, drug abuse, and unknown cause (435). Based on these data, supervised gender-affirming treatment for gender dysphoric persons is crucial because they are at increased risk of committing suicide and self-harm (436).

 

Management of Adult Transgender Women

 

As proposed by the Harry Benjamin International Gender Dysphoria Association, now known as World Professional Association for Transgender Health (WPATH), the process of gender-affirming treatment should be given by a multi and interdisciplinary team, in which the endocrinologist has a key role (437).

 

The interdisciplinary team should consist of a psychologist, a psychiatrist, an endocrinologist, and a surgeon, at least (438). It would be ideal that they all participate in an integrated and consistent way across all the steps of the treatment (439).

 

The mental health professionals (psychologist and psychiatrist) make a distinction between gender dysphoria and conditions with similar features (body dysmorphic disorder and body identity integrity disorder), decide whether the individuals fulfill ICD-11 and DSM-5 criteria, recommend the appropriate treatment and follow-up before, during and after gender-affirming treatment. The endocrinologist will inform about the possibilities and limitations of all sorts of treatment, initiate and monitor the cross-sex hormonal treatment and participate in the indication of gender-affirming surgery. At the final step, the surgeon performs surgical procedures of the treatment (439).

 

DIAGNOSTIC ASSESSMENT AND MENTAL HEALTH CARE

 

Psychological evaluation of persons with gender dysphoria should consider the evolution of the individual as whole, using psychological assessment instruments, such as: freely structured interviews and patterned psychological assessment instruments. For the structured interview, we use a specific questionnaire developed by our mental health professionals that covers childhood, adolescence, and adulthood aspects.

 

During the psychotherapeutic follow up, besides offering an ideal condition for elaborating conflicts and issues regarding gender identity, other variables should be considered, such as individual general state of mental health, ability and manner of conflict resolution, quality of interpersonal relationships, ability to deal with frustrations and limitations, particularly regarding to surgery’s esthetic and functional results idealization. It is recommended that the relatives and/or spouses are invited for interviews to clear them up upon the offered treatment.

 

HORMONAL THERAPY FOR ADULT TRANSGENDER WOMEN

 

Endocrinologists have the responsibility to confirm that persons fulfill criteria for hormonal treatment and clarify the consequences, risks, and benefits of treatment.  

 

Hormone therapy must follow well-defined criteria. The person with gender dysphoria has to: 1) demonstrate knowledge and understanding of the expected and side effects of cross-sex hormone use; 2) complete a real life experience in the gender identity for at least three months, or psychotherapy for a period determined by the mental health professional to consolidate gender identity; and 3) be likely to take hormones appropriately (439).

 

There are two major goals of hormonal therapy: 1) to replace endogenous sex hormone levels and, thus, induce the appearance of sexual characteristics compatible with female gender identity; 2) to reduce endogenous sex hormone levels and, thereby, the secondary male sexual characteristics and 3) to establish the ideal hormones dosage which allows physiological hormone serum levels compatible with female gender identity by adopting the principles of hormone replacement treatment of hypogonadal patients (439,440).

 

Hormone therapy provides a strong relief from the suffering caused by the incongruence of the phenotype with the gender identity.

 

In our clinical practice, we observe that most transgender women consume very high doses of female sex hormones, guided by their wish to obtain fast breast development, and reduce facial hair. However, high doses of hormones are not necessary to achieve the desired effects and are frequently associated with undesirable side effects.

 

The chosen hormone to induce female secondary sexual characteristics are the estrogens. Several pharmaceutical estrogen preparations, including oral, injectable, transdermal, and intravaginal forms associated or not with progesterone are available. Due to the higher cost of the transdermal preparations, the oral route is the most widely used. Nevertheless, the transdermal route is recommended for transgender women over 40 years of age due to the lower association of transdermal 17β-estradiol replacement with thromboembolic events (441).

 

Anti-androgens are used as adjuvants to estrogen, especially in the reduction of male sexual characteristics and the suppression of testosterone to levels compatible with the female sex. Cyproterone acetate blocks testosterone binding to its receptor, and in a dose of 50-100 mg/day associated with estrogen can maintain testosterone in female levels in transgender women (442).

 

At the time, most of the patients in our clinic used conjugated equine estrogens at a dose of 0.625-1.25 mg/day associated with 50 mg/day of cyproterone acetate for an average period of 11 years. At clinical examination we observed satisfactory breast development, decrease of spontaneous erections, thinning of facial and body hair (especially after association with cyproterone acetate), body fat redistribution, enlargement of the areola and nipple and reduction of testicular volume (440).

 

Testosterone levels remained at pre- or intra-pubertal female range (< 14-99 ng/dL) in all patients; LH levels were pre-pubertal (<0.6-0.7 U/L) in 72% of the cases, and the FSH levels were suppressed (<1.0 U/L) in 40% of cases. Therefore, daily use of oral conjugated estrogens at low doses in association with cyproterone acetate is effective in suppressing the hypothalamic-pituitary-testicular axis in transgender women (440).

 

Venous thromboembolism may be a serious complication related to estrogen therapy, particularly during the first year of treatment, when the incidence of this event is 2-6% falling to 0.4% in the second year, significantly higher when compared to the overall young population (0.005 to 0.01%/year). This high incidence of thromboembolic events in transgender women seems to be more associated with ethinyl estradiol than natural oral or transdermal estrogens (441). All patients on estrogen therapy have a mild prolactin level increase. However, a small percentage of these subjects have galactorrhea. In our cohort, two patients had macroprolactinoma, which totally regressed with dopamine agonist treatment. Both had previously used high doses of estrogen (443). Endocrinologists should monitor weight, blood pressure, breast enlargement, body hair involution, body fat redistribution and testicular size every six months. The laboratory evaluation should include measurement of LH, FSH, testosterone, estradiol, prolactin, liver enzymes, complete blood count, coagulation factors, and lipid profile. Bone densitometry and breast ultrasound should be performed yearly.

 

After surgery in patients over 50 years old, the measurement of PSA should be conducted yearly (440).

 

The current key issues include avoiding supraphysiological doses of estrogen and the use of ethinyl estradiol. The preference should be given to conjugated estrogens or transdermal natural estrogen, especially in patients over 40 years of age (444). Hormone therapy provides a strong relief from the suffering caused by gender dysphoria. (440).

 

MANAGEMENT OF PATIENTS WITH 46,XY DSD

 

It is important to stress that the treatment of 46,XY DSD patients requires an appropriately trained multi-disciplinary team. Early diagnosis is important for better outcomes and should start with a careful examination of the newborn’s genitalia at birth (445-447).

 

Psychological Evaluation

 

It is of crucial importance to treat DSD patients (448). Every couple that has a child with atypical genitalia must be assessed and receive counseling by an experienced psychologist, specialized in gender identity, who must be act as soon as the diagnosis is suspected, and then follow the family periodically, more frequently during the periods before and after genitoplasty (449,450).

 

Parents must be well informed by the physician and psychologist about sexual development (451). A simple, detailed, and comprehensive explanation about what to expect regarding integration in social life, sexual activity, need of hormonal replacement and surgical treatment and fertility issues should also be discussed with the parents, before sex assignment (11).

 

The sex assignment must consider the etiological diagnosis, external genitalia, cultural and social aspects, sexual identity and the acceptance of the assigned gender by the parents (452). In case parents and health care providers disagree over the sex of rearing, the parents’ choice must be respected. The affected child and his/her family must be followed throughout life to ascertain the patient’s adjustment to his/her social sex.

 

Hormonal Therapy

 

Sex steroid replacement is an important component of management for some types of 46,XY DSD (453,454). The goals of replacement include induction and maintenance of secondary sex characteristics as well as other aspects of pubertal development including growth.  Bone mineral optimization and promotion of uterine development may also be helped by treatment with sex steroids for some patients. Hormone replacement can also impact psychosocial and psychosexual development, as well as general wellbeing, in positive ways for some people (455,456). Induction and maintenance of pubertal development is necessary in most patients affected by 46,XY DSD regardless of male or female rearing; however, specific indications depend on the underlying etiology of the condition.

 

FEMALE SOCIAL SEX  

 

The purpose of the hormonal therapy is the development of female sexual characteristics and menses in the patients with uterus. There are several options available for estrogen replacement as well as different combinations and doses of progestins (457) however, 17β-estradiol (oral or transdermal) is preferred. Estrogen therapy should be initiated at a low dose (1/6 to 1/4 of the adult dose) to avoid excessive bone maturation in short children and increase gradually at intervals of 6 months. Doses can then be adjusted to the response (Tanner stage, bone age), with the aim of completing feminization gradually over a period of 2–3 years. In 46,XY females with tall stature, adult estrogen dosage is recommended to avoid high final stature.  Transdermal delivery avoids hepatic first-pass metabolism resulting in less thrombogenicity and more neutral effects on lipids (458,459). It is also easier to administer small doses of estrogen by cutting up a patch or by using a metered-dose gel dispenser. An initial recommended dose of oral 17-βestradiol is 5 μg/kg daily, titrated every 6–12 months to an additional 5 μg/kg daily until an adult dose of 1–2 mg daily is achieved (459) .

 

In case of transdermal replacement, the initial recommended dose for the 17-β estradiol patch is 3.1–6.2 mg/24h overnight (1/8–1/4 of 25 mg/24h patch). Transdermal doses can increase 3.1–6.2 mg/24h every 6 months until an adult dose of 50-100 mg/24 h twice a week is achieved (460) Once breast development is complete, an adult dose can be maintained continuously (11). For patients who do not have a uterus, estrogen alone is indicated (458,461). Progesterone is needed to induce endometrial cycling and menses in patients with a uterus. For the latter group, medroxyprogesterone acetate (5 to 10 mg/day) or micronized progesterone (200 mg/day from the 1st to the 12th day of each month) are appropriate to maintain uterine health.

 

Some females with CAIS report decreased psychological wellbeing and sexual dissatisfaction following bilateral gonadectomy and subsequent estrogen replacement (462,463).

 

Testosterone treatment has been proposed as an alternative to estrogen for hormone replacement in these women and such treatment improves sexual desire (464). However, long-term follow-up studies on the impact of T replacement on additional psychological measures, as well as on bone metabolism and cardiovascular outcomes, are needed (465).

 

The dilation of the blind vaginal pouch with acrylic molds (325) or exceptionally surgical neovagina promote development of a vagina adequate for sexual intercourse after 6-10 months of treatment when patients desire to initiate sexual activity (466).

 

MALE SOCIAL SEX   

 

For those raised male, T replacement should strive to mimic masculine pubertal induction between 10 and 12 years of age, provided the child’s predicted height and growth are normal and he indicates a desire and readiness for puberty (5). Intramuscular, short-acting injections of T esters are the most suitable formulation to induce male puberty, although other options include oral T undecanoate and transdermal preparations (467,468).. The initial dose of short-acting T esters is 25–50 mg/month intramuscularly, with further increments of 50- 100 mg every 6–12 months, thereafter. After reaching a replacement dose of 100–150 mg/month, the delivery interval can decrease to every two weeks. 

 

An adult dose of 200-250 mg every two weeks (short-acting T esters), 1000 mg every 10-14 weeks (long-acting T esters), or 50-100 mg for T gel or other transdermal preparations applied topically are effective to maintain male secondary sex characteristics (12,468). Monitoring of T levels should be performed on the day preceding the next hormone administration, and serum levels should fall just above the lower limit of the normal range for eugonadal men.

 

In male patients with androgen insensitivity, higher doses of testosterone esters (250-500 mg twice a week) are used to increase penile length and male secondary characteristics. Maximum penis enlargement is obtained after 6 months of high doses and after that, the normal dosage is re-instituted (272,313). The use of topical DHT gel is also useful to increase penile length with the advantage of not causing gynecomastia.

 

Glucocorticoid Replacement

 

It is necessary for 46,XY DSD patients with classical forms of congenital lipoid adrenal hyperplasia, POR, 3β-HSD type II deficiency to receive glucocorticoid replacement for adrenal insufficiency and in 17α-hydroxylase/17,20-lyase deficiency for hypertension management (469) (267).

 

Mineralocorticoid replacement is also required for 46,XY DSD salt-losing patients (470).

 

Surgical Treatment

 

Surgical approach for 46,XY DSD patients includes: gonadal management, removal of internal structures that disagree with the social sex and reconstruction of the atypical external genitalia. Genital reconstruction involves the feminization or masculinization of external genitalia; these procedures are being widely discussed and controversy continues over the ideal age for genital surgery (471,472). There is a lack of data concerning this issue: a survey with 459 individuals (≥ 16 years) with a DSD diagnosis concerning patients desire about timing of genital surgery was published (473). A total of 66% of individuals with CAH and 60% of those with 46,XY DSD thought that infancy or childhood were the appropriate age for genital surgery. This report concluded that case-by-case decision-making is the best approach (473). In our experience, patients submitted to surgery in adulthood, preferred surgery in infancy and none of the patients operated during childhood regretted the surgery at that age (474).

 

Laparoscopy is the ideal method of surgical treatment of the internal genital organs in patients with 46,XY DSD (475). In these patients, the indications for laparoscopy are the removal of gonads and ductal structures that are contrary to the assigned gender and the removal of dysgenetic gonads, which are nonfunctional and present potential for malignancy. In addition to being a minimally invasive surgery, one of the main advantages of this method is the lack of scars.

 

Feminizing genitoplasty includes the reduction of enlarged clitoral size, opening the urogenital sinus to separate the urethra from the vaginal introitus, and constructing labioscrotal folds. Feminizing techniques have evolved over time to achieve better cosmetic outcomes (476,477). Many techniques have been proposed to separate the urethra from the vaginal introitus and bring both to the surface of the perineum. Fortunoff and Latimer in 1964 described the most commonly used technique until the present day, using an inverted U-shaped perineal skin flap to enlarge the vaginal introitus allowing adequate menstrual flow and future sexual activity (478). Failure to interposing an adequate flap will result in persistent urogenital sinus or vaginal introitus stenosis, requiring later revision (479). Vaginal dilation with acrylic molds in patients with short vagina or introitus stenosis showed to be a good treatment choice when these patients wished to start sexual intercourse, resulting in better outcomes (325). To reduce clitoral enlargement a number of techniques were proposed during the years   (480). Kogan described preserving the neuro-vascular bundle attached to the dorsal portion of the tunica albuginea to protect the nerves and blood supply (481) and this is the technique of choice. The redundant clitoral skin obtained during clitoroplasty is used to create the labia minora; this skin is divided longitudinally and then sutured along either side of the vagina. When necessary, the reduction of labioscrotal folds is performed to create the labia majora, often using a Y-V plasty technique (482).

 

The most common surgical complications, in feminizing genitoplasty, includes: clitoral ischemia or necrosis that can rarely occur in patients with high grade of virilization; introitus stenosis or vaginal stenosis particularly when the confluence of the vagina and urethra is far from the perineum surface and urinary infections mostly observed in patients with persistence of urogenital sinus (471,479,480).

 

In order to minimize surgical complications and dissatisfaction in adulthood, only skilled surgeons with specific training should perform these procedures in the DSD patients (8). In our experience, the single-stage feminizing genitoplasty consisting of clitoroplasty with the preservation of dorsal nerves and vessels and ventral mucosa, vulvoplasty and Y-V perineal flap, followed by vaginal dilation with acrylic molds, allowed good cosmetic and functional results (483).

 

For the males, masculinizing procedures aims to allow the patient to have micturition standing up without effort with a straight and wide stream and to have a satisfactory sexual life with straight erections. The genital surgery consists in correction of hypospadias and scrotal abnormalities, relocation of the testes to the scrotum or removal when dysgenetic, and resection of Mullerian remnants (326,484). Correction of hypospadias includes correction of phallic curvature (orthophalloplasty) and construction of a urethra to the tip of the glans (urethroplasty). Preoperative administration of testosterone is indicated for patients with a small penis (485). Usually, multistage procedures are preferred for male genital reconstruction in DSD patients, due to the severe under virilization represented by proximal hypospadias with severe curvature. The first stage repair consists in ortho-phaloplasty and scrotoplasty (486) (487). The second stage is performed 6-9 months later and consists in urethroplasty). The two-stage approach typically results in better cosmetic outcomes and fewer postoperative complications for patients with severe hypospadias and significant chordee (326,487-489). The most frequent complication in correction of hypospadias is urethral fistula (23%) followed by urethral strictures (9%) and diverticula (4%)  (490). This frequency is highly variable in the literature (490). Fistula can be observed just after surgery or months later, but urethral stenosis in some cases can occur several years after surgery. Reoperations are necessary to correct fistula, diverticula and particularly to treat severe urethral strictures. The buccal mucosa graft is commonly used to enlarge the urethra in these cases (490). For patients with undescended testes, simultaneous orchidopexy may be performed. The surgical treatment of gonads of 46, XY DSD patients aims to preserve testicular function (production of testosterone and sperm) and prevent malignancy (288,360,491). Finally, gonadectomy is recommended for patients at risk for neoplastic transformation of germ cells (gonadoblastomas and/or an invasive germ cell tumor) in dysgenetic gonads (287).

 

When gonadectomy is recommended, patients may then choose to have a testicular prosthesis placed in the scrotum (492).

 

Müllerian structures are rudimentary in some patients and present as a cystic prostatic utricle. These utricles may be left in situ when asymptomatic, but in cases of recurrent urinary tract infection, stones, or significant post-void urethral dribbling due to urinary pooling, they can be removed either laparoscopically or through a sagittal posterior incision of the perineum (475). With either approach, great care must be taken to prevent injury to the vas deferens, seminal vesicles and pelvic nerves so as to avoid subsequent infertility, erectile dysfunction and urinary incontinence (488,493). Late evaluation of 46,XY DSD patients operated in childhood due to proximal hypospadias reveals that many felt that their genitals had an unusual appearance or presented some degree of urinary or sexual dysfunction (494). Objectively, most DSD patients have a penile length below the -2.0 SD (5.2 ± 2.0 cm) (326). Dysfunctional voiding and lower urinary tract symptoms are also more frequent in these patients than in controls (495). However, between 55.6 and 91% of these patients after genitoplasty were satisfied with their overall sexual function after genitoplasty, when considering sexual contacts, libido, erections, orgasm, as well as size of the penis and volume of ejaculation (326),(494),(496),(497),(498). The long-term outcomes were evaluated for a long time concerning functional and cosmetic results that could be analyzed by objective criteria. The subjective long-term evaluation analyzing psychological and sexual implications in quality of life were often neglected in the past, but is being currently explored (332) (499) (500). Jones et al reported that 81% were satisfied with their genital appearance and that 90% were satisfied with their overall body image (500). Most of our patients were satisfied with their genital appearance and present satisfactory sexual performance as long as they present a penis size of at least 6 cm (326).

 

ACKNOWLEDGMENT

 

The authors would like to thank the postgraduate students Nathalia Lisboa Gomes and Jose Antonio D Faria Junior for their help in the update of this chapter.

 

REFERENCES

 

  1. Donahoe PK, Schnitzer JJ. Evaluation of the infant who has ambiguous genitalia, and principles of operative management. Semin Pediatr Surg. 1996;5(1):30-40.
  2. Gomes NL, Chetty T, Jorgensen A, Mitchell RT. Disorders of Sex Development-Novel Regulators, Impacts on Fertility, and Options for Fertility Preservation. Int J Mol Sci. 2020;21(7).
  3. Sreenivasan R, Gonen N, Sinclair A. SOX Genes and Their Role in Disorders of Sex Development. Sex Dev. 2022:1-12.
  4. Pask A. The Reproductive System. Adv Exp Med Biol. 2016;886:1-12.
  5. Tanaka SS, Nishinakamura R. Regulation of male sex determination: genital ridge formation and Sry activation in mice. Cell Mol Life Sci. 2014;71(24):4781-4802.
  6. Lucas-Herald AK, Bashamboo A. Gonadal development. Endocr Dev. 2014;27:1-16.
  7. Hughes IA, Houk C, Ahmed SF, Lee PA, Group LC, Group EC. Consensus statement on management of intersex disorders. Arch Dis Child. 2006;91(7):554-563.
  8. Lee PA, Houk CP, Ahmed SF, Hughes IA, Endocrinology ICCoIobtLWPESatESfP. Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics. 2006;118(2):e488-500.
  9. Russell DW, Wilson JD. Chapter 4A - Steroid 5α-Reductase 2 Deficiency. In: Hammer MINLPTYWOMD, ed. Genetic Steroid Disorders. San Diego: Academic Press; 2014:199-214.
  10. Bennett NC, Gardiner RA, Hooper JD, Johnson DW, Gobe GC. Molecular cell biology of androgen receptor signalling. Int J Biochem Cell Biol. 2010;42(6):813-827.
  11. Wisniewski AB, Batista RL, Costa EMF, Finlayson C, Sircili MHP, Dénes FT, Domenice S, Mendonca BB. Management of 46,XY Differences/Disorders of Sex Development (DSD) Throughout Life. Endocr Rev. 2019.
  12. Mendonca BB, Domenice S, Arnhold IJ, Costa EM. 46,XY disorders of sex development (DSD). Clin Endocrinol (Oxf). 2009;70(2):173-187.
  13. Josso N, Picard JY, Rey R, di Clemente N. Testicular anti-Mullerian hormone: history, genetics, regulation and clinical applications. Pediatr Endocrinol Rev. 2006;3(4):347-358.
  14. Lahlou N, Roger M. Inhibin B in pubertal development and pubertal disorders. Semin Reprod Med.2004;22(3):165-175.
  15. Arnhold IJ, Mendonça BB, Diaz JA, Nogueira C, Batista MC, Madureira G, Oliveira D, Nicolau W, Bloise W. Prepubertal male pseudohermaphroditism due to 17-ketosteroid reductase deficiency: diagnostic value of a hCG test and lack of HLA association. J Endocrinol Invest. 1988;11(4):319-322.
  16. Guerra-Junior G, Andrade KC, Barcelos IHK, Maciel-Guerra AT. Imaging Techniques in the Diagnostic Journey of Disorders of Sex Development. Sex Dev. 2018;12(1-3):95-99.
  17. Garel L. Abnormal sex differentiation: who, how and when to image. Pediatr Radiol. 2008;38 Suppl 3:S508-511.
  18. Parivesh A, Barseghyan H, Délot E, Vilain E. Translating genomics to the clinical diagnosis of disorders/differences of sex development. Curr Top Dev Biol. 2019;134:317-375.
  19. Ahmed SF, Alimusina M, Batista RL, Domenice S, Lisboa Gomes N, McGowan R, Patjamontri S, Mendonca BB. The Use of Genetics for Reaching a Diagnosis in XY DSD. Sex Dev. 2022:1-18.
  20. Gomes NL, Batista RL, Nishi MY, Lerario AM, Silva TE, de Moraes Narcizo A, Benedetti AFF, de Assis Funari MF, Faria Junior JA, Moraes DR, Quintao LML, Montenegro LR, Ferrari MTM, Jorge AA, Arnhold IJP, Costa EMF, Domenice S, Mendonca BB. Contribution of Clinical and Genetic Approaches for Diagnosing 209 Index Cases With 46,XY Differences of Sex Development. J Clin Endocrinol Metab.2022;107(5):e1797-e1806.
  21. Igarashi M, Dung VC, Suzuki E, Ida S, Nakacho M, Nakabayashi K, Mizuno K, Hayashi Y, Kohri K, Kojima Y, Ogata T, Fukami M. Cryptic genomic rearrangements in three patients with 46,XY disorders of sex development. PLoS One. 2013;8(7):e68194.
  22. Kon M, Fukami M. Submicroscopic copy-number variations associated with 46,XY disorders of sex development. Mol Cell Pediatr. 2015;2(1):7.
  23. Ahmed SF, Hughes IA. The genetics of male undermasculinization. Clin Endocrinol (Oxf). 2002;56(1):1-18.
  24. Buonocore F, Achermann JC. Human sex development: targeted technologies to improve diagnosis. Genome Biol. 2016;17(1):257.
  25. Croft B, Ohnesorg T, Sinclair AH. The Role of Copy Number Variants in Disorders of Sex Development. Sex Dev. 2018;12(1-3):19-29.
  26. Eggers S, Sadedin S, van den Bergen JA, Robevska G, Ohnesorg T, Hewitt J, Lambeth L, Bouty A, Knarston IM, Tan TY, Cameron F, Werther G, Hutson J, O'Connell M, Grover SR, Heloury Y, Zacharin M, Bergman P, Kimber C, Brown J, Webb N, Hunter MF, Srinivasan S, Titmuss A, Verge CF, Mowat D, Smith G, Smith J, Ewans L, Shalhoub C, Crock P, Cowell C, Leong GM, Ono M, Lafferty AR, Huynh T, Visser U, Choong CS, McKenzie F, Pachter N, Thompson EM, Couper J, Baxendale A, Gecz J, Wheeler BJ, Jefferies C, MacKenzie K, Hofman P, Carter P, King RI, Krausz C, van Ravenswaaij-Arts CM, Looijenga L, Drop S, Riedl S, Cools M, Dawson A, Juniarto AZ, Khadilkar V, Khadilkar A, Bhatia V, Dũng VC, Atta I, Raza J, Thi Diem Chi N, Hao TK, Harley V, Koopman P, Warne G, Faradz S, Oshlack A, Ayers KL, Sinclair AH. Disorders of sex development: insights from targeted gene sequencing of a large international patient cohort. Genome Biol. 2016;17(1):243.
  27. Norling A, Linden Hirschberg A, Iwarsson E, Persson B, Wedell A, Barbaro M. Novel candidate genes for 46,XY gonadal dysgenesis identified by a customized 1 M array-CGH platform. Eur J Med Genet.2013;56(12):661-668.
  28. Ledig S, Hiort O, Scherer G, Hoffmann M, Wolff G, Morlot S, Kuechler A, Wieacker P. Array-CGH analysis in patients with syndromic and non-syndromic XY gonadal dysgenesis: evaluation of array CGH as diagnostic tool and search for new candidate loci. Hum Reprod. 2010;25(10):2637-2646.
  29. Parivesh A, Barseghyan H, Delot E, Vilain E. Translating genomics to the clinical diagnosis of disorders/differences of sex development. Curr Top Dev Biol. 2019;134:317-375.
  30. Dong Y, Yi Y, Yao H, Yang Z, Hu H, Liu J, Gao C, Zhang M, Zhou L, Asan, Yi X, Liang Z. Targeted next-generation sequencing identification of mutations in patients with disorders of sex development. BMC Med Genet. 2016;17:23.
  31. Bocher O, Genin E. Rare variant association testing in the non-coding genome. Hum Genet.2020;139(11):1345-1362.
  32. Ahmad-Nejad P, Ashavaid T, Vacaflores Salinas A, Huggett J, Harris K, Linder MW, Baluchova K, Steimer W, Payne DA, Diagnostics ICfM. Current and future challenges in quality assurance in molecular diagnostics. Clin Chim Acta. 2021;519:239-246.
  33. Jeong YH, Lu H, Park CH, Li M, Luo H, Kim JJ, Liu S, Ko KH, Huang S, Hwang IS, Kang MN, Gong D, Park KB, Choi EJ, Park JH, Jeong YW, Moon C, Hyun SH, Kim NH, Jeung EB, Yang H, Hwang WS, Gao F. Stochastic anomaly of methylome but persistent SRY hypermethylation in disorder of sex development in canine somatic cell nuclear transfer. Sci Rep. 2016;6:31088.
  34. Miyamoto N, Yoshida M, Kuratani S, Matsuo I, Aizawa S. Defects of urogenital development in mice lacking Emx2. Development. 1997;124(9):1653-1664.
  35. Wilhelm D, Englert C. The Wilms tumor suppressor WT1 regulates early gonad development by activation of Sf1. Genes Dev. 2002;16(14):1839-1851.
  36. Parker KL, Schedl A, Schimmer BP. Gene interactions in gonadal development. Annu Rev Physiol.1999;61:417-433.
  37. Parker KL, Schimmer BP, Schedl A. Genes essential for early events in gonadal development. Cell Mol Life Sci. 1999;55(6-7):831-838.
  38. Ito M, Yu R, Jameson JL. DAX-1 inhibits SF-1-mediated transactivation via a carboxy-terminal domain that is deleted in adrenal hypoplasia congenita. Mol Cell Biol. 1997;17(3):1476-1483.
  39. Hossain A, Saunders GF. The human sex-determining gene SRY is a direct target of WT1. J Biol Chem.2001;276(20):16817-16823.
  40. Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Eicher EM, Orkin SH. Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development. 2002;129(19):4627-4634.
  41. Katoh-Fukui Y, Miyabayashi K, Komatsu T, Owaki A, Baba T, Shima Y, Kidokoro T, Kanai Y, Schedl A, Wilhelm D, Koopman P, Okuno Y, Morohashi K. Cbx2, a polycomb group gene, is required for Sry gene expression in mice. Endocrinology. 2012;153(2):913-924.
  42. Bashamboo A, Eozenou C, Rojo S, McElreavey K. Anomalies in human sex determination provide unique insights into the complex genetic interactions of early gonad development. Clin Genet.2017;91(2):143-156.
  43. Knower KC, Sim H, McClive PJ, Bowles J, Koopman P, Sinclair AH, Harley VR. Characterisation of urogenital ridge gene expression in the human embryonal carcinoma cell line NT2/D1. Sex Dev.2007;1(2):114-126.
  44. Bernard P, Sim H, Knower K, Vilain E, Harley V. Human SRY inhibits beta-catenin-mediated transcription. Int J Biochem Cell Biol. 2008;40(12):2889-2900.
  45. Kim Y, Kobayashi A, Sekido R, DiNapoli L, Brennan J, Chaboissier MC, Poulat F, Behringer RR, Lovell-Badge R, Capel B. Fgf9 and Wnt4 act as antagonistic signals to regulate mammalian sex determination. PLoS Biol. 2006;4(6):e187.
  46. Maatouk DM, DiNapoli L, Alvers A, Parker KL, Taketo MM, Capel B. Stabilization of beta-catenin in XY gonads causes male-to-female sex-reversal. Hum Mol Genet. 2008;17(19):2949-2955.
  47. Wilhelm D, Hiramatsu R, Mizusaki H, Widjaja L, Combes AN, Kanai Y, Koopman P. SOX9 regulates prostaglandin D synthase gene transcription in vivo to ensure testis development. J Biol Chem.2007;282(14):10553-10560.
  48. Moniot B, Declosmenil F, Barrionuevo F, Scherer G, Aritake K, Malki S, Marzi L, Cohen-Solal A, Georg I, Klattig J, Englert C, Kim Y, Capel B, Eguchi N, Urade Y, Boizet-Bonhoure B, Poulat F. The PGD2 pathway, independently of FGF9, amplifies SOX9 activity in Sertoli cells during male sexual differentiation. Development. 2009;136(11):1813-1821.
  49. Matson CK, Murphy MW, Sarver AL, Griswold MD, Bardwell VJ, Zarkower D. DMRT1 prevents female reprogramming in the postnatal mammalian testis. Nature. 2011;476(7358):101-104.
  50. Pearlman A, Loke J, Le Caignec C, White S, Chin L, Friedman A, Warr N, Willan J, Brauer D, Farmer C, Brooks E, Oddoux C, Riley B, Shajahan S, Camerino G, Homfray T, Crosby AH, Couper J, David A, Greenfield A, Sinclair A, Ostrer H. Mutations in MAP3K1 cause 46,XY disorders of sex development and implicate a common signal transduction pathway in human testis determination. Am J Hum Genet.2010;87(6):898-904.
  51. Loke J, Pearlman A, Radi O, Zuffardi O, Giussani U, Pallotta R, Camerino G, Ostrer H. Mutations in MAP3K1 tilt the balance from SOX9/FGF9 to WNT/beta-catenin signaling. Hum Mol Genet.2014;23(4):1073-1083.
  52. Bashamboo A, McElreavey K. Human sex-determination and disorders of sex-development (DSD). Semin Cell Dev Biol. 2015;45:77-83.
  53. Evilen da Silva T, Gomes NL, Lerário AM, Keegan CE, Nishi MY, Carvalho FM, Vilain E, Barseghyanm H, Martinez-Aguayo A, Forclaz MV, Papazian R, Pedroso de Paula LC, Costa EC, Carvalho LR, Jorge AA, Elias F, Mitchell R, Frade Costa EM, Mendonca BB, Domenice S. Genetic evidence of the association of DEAH-box helicase 37 defects with 46,XY gonadal dysgenesis spectrum. J Clin Endocrinol Metab. 2019.
  54. De Marchi M, Campagnoli C, Ghiringhello B, Ponzio G, Carbonara A. Gonadal agenesis in a phenotypically normal female with positive H-Y antigen. Hum Genet. 1981;56(3):417-419.
  55. Mendonca BB, Barbosa AS, Arnhold IJ, McElreavey K, Fellous M, Moreira-Filho CA. Gonadal agenesis in XX and XY sisters: evidence for the involvement of an autosomal gene. Am J Med Genet.1994;52(1):39-43.
  56. Ottolenghi C, Moreira-Filho C, Mendonca BB, Barbieri M, Fellous M, Berkovitz GD, McElreavey K. Absence of mutations involving the LIM homeobox domain gene LHX9 in 46,XY gonadal agenesis and dysgenesis. J Clin Endocrinol Metab. 2001;86(6):2465-2469.
  57. Swyer GI. Male pseudohermaphroditism: a hitherto undescribed form. Br Med J. 1955;2(4941):709-712.
  58. Josso N, Briard ML. Embryonic testicular regression syndrome: variable phenotypic expression in siblings. J Pediatr. 1980;97(2):200-204.
  59. Berkovitz GD, Fechner PY, Zacur HW, Rock JA, Snyder HM, 3rd, Migeon CJ, Perlman EJ. Clinical and pathologic spectrum of 46,XY gonadal dysgenesis: its relevance to the understanding of sex differentiation. Medicine (Baltimore). 1991;70(6):375-383.
  60. Viger RS, Guittot SM, Anttonen M, Wilson DB, Heikinheimo M. Role of the GATA family of transcription factors in endocrine development, function, and disease. Mol Endocrinol. 2008;22(4):781-798.
  61. Garg V, Kathiriya IS, Barnes R, Schluterman MK, King IN, Butler CA, Rothrock CR, Eapen RS, Hirayama-Yamada K, Joo K, Matsuoka R, Cohen JC, Srivastava D. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature. 2003;424(6947):443-447.
  62. Wat MJ, Shchelochkov OA, Holder AM, Breman AM, Dagli A, Bacino C, Scaglia F, Zori RT, Cheung SW, Scott DA, Kang SH. Chromosome 8p23.1 deletions as a cause of complex congenital heart defects and diaphragmatic hernia. Am J Med Genet A. 2009;149A(8):1661-1677.
  63. Lourenco D, Brauner R, Rybczynska M, Nihoul-Fekete C, McElreavey K, Bashamboo A. Loss-of-function mutation in GATA4 causes anomalies of human testicular development. Proc Natl Acad Sci U S A. 2011;108(4):1597-1602.
  64. Finelli P, Pincelli AI, Russo S, Bonati MT, Recalcati MP, Masciadri M, Giardino D, Cavagnini F, Larizza L. Disruption of friend of GATA 2 gene (FOG-2) by a de novo t(8;10) chromosomal translocation is associated with heart defects and gonadal dysgenesis. Clin Genet. 2007;71(3):195-204.
  65. Bashamboo A, Brauner R, Bignon-Topalovic J, Lortat-Jacob S, Karageorgou V, Lourenco D, Guffanti A, McElreavey K. Mutations in the FOG2/ZFPM2 gene are associated with anomalies of human testis determination. Hum Mol Genet. 2014;23(14):3657-3665.
  66. van den Bergen JA, Robevska G, Eggers S, Riedl S, Grover SR, Bergman PB, Kimber C, Jiwane A, Khan S, Krausz C, Raza J, Atta I, Davis SR, Ono M, Harley V, Faradz SMH, Sinclair AH, Ayers KL. Analysis of variants in GATA4 and FOG2/ZFPM2 demonstrates benign contribution to 46,XY disorders of sex development. Mol Genet Genomic Med. 2020;8(3):e1095.
  67. Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C, Schoenle EJ. Ovaries and female phenotype in a girl with 46,XY karyotype and mutations in the CBX2 gene. Am J Hum Genet. 2009;84(5):658-663.
  68. Sproll P, Eid W, Gomes CR, Mendonca BB, Gomes NL, Costa EM, Biason-Lauber A. Assembling the jigsaw puzzle: CBX2 isoform 2 and its targets in disorders/differences of sex development. Mol Genet Genomic Med. 2018;6(5):785-795.
  69. Elzaiat M, McElreavey K, Bashamboo A. Genetics of 46,XY gonadal dysgenesis. Best Pract Res Clin Endocrinol Metab. 2022;36(1):101633.
  70. Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM, Housman DE. Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc Natl Acad Sci U S A. 1991;88(21):9618-9622.
  71. Melo KF, Martin RM, Costa EM, Carvalho FM, Jorge AA, Arnhold IJ, Mendonca BB. An unusual phenotype of Frasier syndrome due to IVS9 +4C>T mutation in the WT1 gene: predominantly male ambiguous genitalia and absence of gonadal dysgenesis. J Clin Endocrinol Metab. 2002;87(6):2500-2505.
  72. Ferrari MTM, Watanabe A, da Silva TE, Gomes NL, Batista RL, Nishi MY, de Paula LCP, Costa EC, Costa EMF, Cukier P, Onuchic LF, Mendonca BB, Domenice S. WT1 Pathogenic Variants are Associated with a Broad Spectrum of Differences in Sex Development Phenotypes and Heterogeneous Progression of Renal Disease. Sex Dev. 2022;16(1):46-54.
  73. Riccardi VM, Sujansky E, Smith AC, Francke U. Chromosomal imbalance in the Aniridia-Wilms' tumor association: 11p interstitial deletion. Pediatrics. 1978;61(4):604-610.
  74. van Heyningen V, Bickmore WA, Seawright A, Fletcher JM, Maule J, Fekete G, Gessler M, Bruns GA, Huerre-Jeanpierre C, Junien C, et al. Role for the Wilms tumor gene in genital development? Proc Natl Acad Sci U S A. 1990;87(14):5383-5386.
  75. Tiberio G, Digilio MC, Giannotti A. Obesity and WAGR syndrome. Clin Dysmorphol. 2000;9(1):63-64.
  76. Han JC, Liu QR, Jones M, Levinn RL, Menzie CM, Jefferson-George KS, Adler-Wailes DC, Sanford EL, Lacbawan FL, Uhl GR, Rennert OM, Yanovski JA. Brain-derived neurotrophic factor and obesity in the WAGR syndrome. N Engl J Med. 2008;359(9):918-927.
  77. Le Caignec C, Delnatte C, Vermeesch JR, Boceno M, Joubert M, Lavenant F, David A, Rival JM. Complete sex reversal in a WAGR syndrome patient. Am J Med Genet A. 2007;143A(22):2692-2695.
  78. Mueller RF. The Denys-Drash syndrome. J Med Genet. 1994;31(6):471-477.
  79. Baird PN, Santos A, Groves N, Jadresic L, Cowell JK. Constitutional mutations in the WT1 gene in patients with Denys-Drash syndrome. Hum Mol Genet. 1992;1(5):301-305.
  80. Pelletier J, Bruening W, Kashtan CE, Mauer SM, Manivel JC, Striegel JE, Houghton DC, Junien C, Habib R, Fouser L, et al. Germline mutations in the Wilms' tumor suppressor gene are associated with abnormal urogenital development in Denys-Drash syndrome. Cell. 1991;67(2):437-447.
  81. da Silva TE, Nishi MY, Costa EM, Martin RM, Carvalho FM, Mendonca BB, Domenice S. A novel WT1 heterozygous nonsense mutation (p.K248X) causing a mild and slightly progressive nephropathy in a 46,XY patient with Denys-Drash syndrome. Pediatr Nephrol. 2011;26(8):1311-1315.
  82. Gwin K, Cajaiba MM, Caminoa-Lizarralde A, Picazo ML, Nistal M, Reyes-Mugica M. Expanding the clinical spectrum of Frasier syndrome. Pediatr Dev Pathol. 2008;11(2):122-127.
  83. Drash A, Sherman F, Hartmann WH, Blizzard RM. A syndrome of pseudohermaphroditism, Wilms' tumor, hypertension, and degenerative renal disease. J Pediatr. 1970;76(4):585-593.
  84. Kohsaka T, Tagawa M, Takekoshi Y, Yanagisawa H, Tadokoro K, Yamada M. Exon 9 mutations in the WT1 gene, without influencing KTS splice isoforms, are also responsible for Frasier syndrome. Hum Mutat. 1999;14(6):466-470.
  85. Barbosa AS, Hadjiathanasiou CG, Theodoridis C, Papathanasiou A, Tar A, Merksz M, Gyorvari B, Sultan C, Dumas R, Jaubert F, Niaudet P, Moreira-Filho CA, Cotinot C, Fellous M. The same mutation affecting the splicing of WT1 gene is present on Frasier syndrome patients with or without Wilms' tumor. Hum Mutat. 1999;13(2):146-153.
  86. Morohashi K, Honda S, Inomata Y, Handa H, Omura T. A common trans-acting factor, Ad4-binding protein, to the promoters of steroidogenic P-450s. J Biol Chem. 1992;267(25):17913-17919.
  87. Lala DS, Rice DA, Parker KL. Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I. Mol Endocrinol. 1992;6(8):1249-1258.
  88. Rice DA, Mouw AR, Bogerd AM, Parker KL. A shared promoter element regulates the expression of three steroidogenic enzymes. Mol Endocrinol. 1991;5(10):1552-1561.
  89. Lin L, Achermann JC. Steroidogenic factor-1 (SF-1, Ad4BP, NR5A1) and disorders of testis development. Sex Dev. 2008;2(4-5):200-209.
  90. Schimmer BP, White PC. Minireview: steroidogenic factor 1: its roles in differentiation, development, and disease. Mol Endocrinol. 2010;24(7):1322-1337.
  91. Majdic G, Young M, Gomez-Sanchez E, Anderson P, Szczepaniak LS, Dobbins RL, McGarry JD, Parker KL. Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology. 2002;143(2):607-614.
  92. Achermann JC, Ito M, Hindmarsh PC, Jameson JL. A mutation in the gene encoding steroidogenic factor-1 causes XY sex reversal and adrenal failure in humans. Nat Genet. 1999;22(2):125-126.
  93. Biason-Lauber A, Schoenle EJ. Apparently normal ovarian differentiation in a prepubertal girl with transcriptionally inactive steroidogenic factor 1 (NR5A1/SF-1) and adrenocortical insufficiency. Am J Hum Genet. 2000;67(6):1563-1568.
  94. Gerster K, Biason-Lauber A, Schoenle EJ. Clinical follow-up of the first SF-1 insufficient female patient. Ann Endocrinol (Paris). 2017;78(3):156-161.
  95. Achermann JC, Ozisik G, Ito M, Orun UA, Harmanci K, Gurakan B, Jameson JL. Gonadal determination and adrenal development are regulated by the orphan nuclear receptor steroidogenic factor-1, in a dose-dependent manner. J Clin Endocrinol Metab. 2002;87(4):1829-1833.
  96. Ferraz-de-Souza B, Lin L, Achermann JC. Steroidogenic factor-1 (SF-1, NR5A1) and human disease.Mol Cell Endocrinol. 2011;336(1-2):198-205.
  97. Lin L, Philibert P, Ferraz-de-Souza B, Kelberman D, Homfray T, Albanese A, Molini V, Sebire NJ, Einaudi S, Conway GS, Hughes IA, Jameson JL, Sultan C, Dattani MT, Achermann JC. Heterozygous missense mutations in steroidogenic factor 1 (SF1/Ad4BP, NR5A1) are associated with 46,XY disorders of sex development with normal adrenal function. J Clin Endocrinol Metab. 2007;92(3):991-999.
  98. Lourenco D, Brauner R, Lin L, De Perdigo A, Weryha G, Muresan M, Boudjenah R, Guerra-Junior G, Maciel-Guerra AT, Achermann JC, McElreavey K, Bashamboo A. Mutations in NR5A1 associated with ovarian insufficiency. N Engl J Med. 2009;360(12):1200-1210.
  99. Correa RV, Domenice S, Bingham NC, Billerbeck AE, Rainey WE, Parker KL, Mendonca BB. A microdeletion in the ligand binding domain of human steroidogenic factor 1 causes XY sex reversal without adrenal insufficiency. J Clin Endocrinol Metab. 2004;89(4):1767-1772.
  100. Urs AN, Dammer E, Sewer MB. Sphingosine regulates the transcription of CYP17 by binding to steroidogenic factor-1. Endocrinology. 2006;147(11):5249-5258.
  101. Domenice S, Zamboni Machado A, Moraes Ferreira F, Ferraz-de-Souza B, Marcondes Lerario A, Lin L, Yumie Nishi M, Lisboa Gomes N, Evelin da Silva T, Barbosa Silva R, Vieira Correa R, Ribeiro Montenegro L, Narciso A, Maria Frade Costa E, C Achermann J, Bilharinho Mendonca B. Wide spectrum of NR5A1-related phenotypes in 46,XY and 46,XX individuals. Birth Defects Res C Embryo Today. 2016;108(4):309-320.
  102. Fabbri HC, Ribeiro de Andrade JG, Maciel-Guerra AT, Guerra-Júnior G, de Mello MP. NR5A1 Loss-of-Function Mutations Lead to 46,XY Partial Gonadal Dysgenesis Phenotype: Report of Three Novel Mutations. Sex Dev. 2016;10(4):191-199.
  103. Fabbri-Scallet H, de Sousa LM, Maciel-Guerra AT, Guerra-Júnior G, de Mello MP. Mutation update for the NR5A1 gene involved in DSD and infertility. Hum Mutat. 2020;41(1):58-68.
  104. Pedace L, Laino L, Preziosi N, Valentini MS, Scommegna S, Rapone AM, Guarino N, Boscherini B, De Bernardo C, Marrocco G, Majore S, Grammatico P. Longitudinal hormonal evaluation in a patient with disorder of sexual development, 46,XY karyotype and one NR5A1 mutation. Am J Med Genet A.2014;164A(11):2938-2946.
  105. Tantawy S, Mazen I, Soliman H, Anwar G, Atef A, El-Gammal M, El-Kotoury A, Mekkawy M, Torky A, Rudolf A, Schrumpf P, Grüters A, Krude H, Dumargne MC, Astudillo R, Bashamboo A, Biebermann H, Köhler B. Analysis of the gene coding for steroidogenic factor 1 (SF1, NR5A1) in a cohort of 50 Egyptian patients with 46,XY disorders of sex development. Eur J Endocrinol. 2014;170(5):759-767.
  106. Warman DM, Costanzo M, Marino R, Berensztein E, Galeano J, Ramirez PC, Saraco N, Baquedano MS, Ciaccio M, Guercio G, Chaler E, Maceiras M, Lazzatti JM, Bailez M, Rivarola MA, Belgorosky A. Three new SF-1 (NR5A1) gene mutations in two unrelated families with multiple affected members: within-family variability in 46,XY subjects and low ovarian reserve in fertile 46,XX subjects. Horm Res Paediatr. 2011;75(1):70-77.
  107. Mazen I, Abdel-Hamid M, Mekkawy M, Bignon-Topalovic J, Boudjenah R, El Gammal M, Essawi M, Bashamboo A, McElreavey K. Identification of NR5A1 Mutations and Possible Digenic Inheritance in 46,XY Gonadal Dysgenesis. Sex Dev. 2016;10(3):147-151.
  108. Allali S, Muller JB, Brauner R, Lourenco D, Boudjenah R, Karageorgou V, Trivin C, Lottmann H, Lortat-Jacob S, Nihoul-Fekete C, De Dreuzy O, McElreavey K, Bashamboo A. Mutation analysis of NR5A1 encoding steroidogenic factor 1 in 77 patients with 46, XY disorders of sex development (DSD) including hypospadias. PLoS One. 2011;6(10):e24117.
  109. Bashamboo A, Ferraz-de-Souza B, Lourenço D, Lin L, Sebire NJ, Montjean D, Bignon-Topalovic J, Mandelbaum J, Siffroi JP, Christin-Maitre S, Radhakrishna U, Rouba H, Ravel C, Seeler J, Achermann JC, McElreavey K. Human male infertility associated with mutations in NR5A1 encoding steroidogenic factor 1. Am J Hum Genet. 2010;87(4):505-512.
  110. El-Khairi R, Achermann JC. Steroidogenic factor-1 and human disease. Semin Reprod Med.2012;30(5):374-381.
  111. Tantawy S, Lin L, Akkurt I, Borck G, Klingmuller D, Hauffa BP, Krude H, Biebermann H, Achermann JC, Kohler B. Testosterone production during puberty in two 46,XY patients with disorders of sex development and novel NR5A1 (SF-1) mutations. Eur J Endocrinol. 2012;167(1):125-130.
  112. Philibert P, Zenaty D, Lin L, Soskin S, Audran F, Léger J, Achermann JC, Sultan C. Mutational analysis of steroidogenic factor 1 (NR5a1) in 24 boys with bilateral anorchia: a French collaborative study. Hum Reprod. 2007;22(12):3255-3261.
  113. Camats N, Pandey AV, Fernandez-Cancio M, Andaluz P, Janner M, Toran N, Moreno F, Bereket A, Akcay T, Garcia-Garcia E, Munoz MT, Gracia R, Nistal M, Castano L, Mullis PE, Carrascosa A, Audi L, Fluck CE. Ten novel mutations in the NR5A1 gene cause disordered sex development in 46,XY and ovarian insufficiency in 46,XX individuals. J Clin Endocrinol Metab. 2012;97(7):E1294-1306.
  114. Smith AM RN, Robin NH. NR5A1 Pathogenic Variant Identified in Non-Syndromic 46, XY Ovotesticular Disorder of Sexual Development. Archives of Pediatrics. 2019;4(162. Knarston IM, Robevska G, van den Bergen JA, Eggers S, Croft B, Yates J, Hersmus R, Looijenga LHJ, Cameron FJ, Monhike K, Ayers KL, Sinclair AH. NR5A1 gene variants repress the ovarian-specific WNT signaling pathway in 46,XX disorders of sex development patients. Hum Mutat. 2019;40(2):207-216.
  115. Vilain E, Elreavey KM, Richaud F, Fellous M. [Sex genetics]. Presse Med. 1992;21(18):852-856.
  116. Hawkins JR. Mutational analysis of SRY in XY females. Hum Mutat. 1993;2(5):347-350.
  117. McElreavey K, Vilain E, Barbaux S, Fuqua JS, Fechner PY, Souleyreau N, Doco-Fenzy M, Gabriel R, Quereux C, Fellous M, Berkovitz GD. Loss of sequences 3' to the testis-determining gene, SRY, including the Y pseudoautosomal boundary associated with partial testicular determination. Proc Natl Acad Sci U S A. 1996;93(16):8590-8594.
  118. Harley VR, Jackson DI, Hextall PJ, Hawkins JR, Berkovitz GD, Sockanathan S, Lovell-Badge R, Goodfellow PN. DNA binding activity of recombinant SRY from normal males and XY females. Science.1992;255(5043):453-456.
  119. Schmitt-Ney M, Thiele H, Kaltwasser P, Bardoni B, Cisternino M, Scherer G. Two novel SRY missense mutations reducing DNA binding identified in XY females and their mosaic fathers. Am J Hum Genet.1995;56(4):862-869.
  120. Assumpcao JG, Benedetti CE, Maciel-Guerra AT, Guerra G, Jr., Baptista MT, Scolfaro MR, de Mello MP. Novel mutations affecting SRY DNA-binding activity: the HMG box N65H associated with 46,XY pure gonadal dysgenesis and the familial non-HMG box R30I associated with variable phenotypes. J Mol Med (Berl). 2002;80(12):782-790.
  121. Foster JW, Dominguez-Steglich MA, Guioli S, Kwok C, Weller PA, Stevanovic M, Weissenbach J, Mansour S, Young ID, Goodfellow PN, et al. Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 1994;372(6506):525-530.
  122. Wagner T, Wirth J, Meyer J, Zabel B, Held M, Zimmer J, Pasantes J, Bricarelli FD, Keutel J, Hustert E, Wolf U, Tommerup N, Schempp W, Scherer G. Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell. 1994;79(6):1111-1120.
  123. Kwok C, Weller PA, Guioli S, Foster JW, Mansour S, Zuffardi O, Punnett HH, Dominguez-Steglich MA, Brook JD, Young ID, et al. Mutations in SOX9, the gene responsible for Campomelic dysplasia and autosomal sex reversal. Am J Hum Genet. 1995;57(5):1028-1036.
  124. Croft B, Ohnesorg T, Hewitt J, Bowles J, Quinn A, Tan J, Corbin V, Pelosi E, van den Bergen J, Sreenivasan R, Knarston I, Robevska G, Vu DC, Hutson J, Harley V, Ayers K, Koopman P, Sinclair A. Human sex reversal is caused by duplication or deletion of core enhancers upstream of SOX9. Nat Commun. 2018;9(1):5319.
  125. Migale R, Neumann M, Lovell-Badge R. Long-Range Regulation of Key Sex Determination Genes. Sex Dev. 2021;15(5-6):360-380.
  126. Pop R, Conz C, Lindenberg KS, Blesson S, Schmalenberger B, Briault S, Pfeifer D, Scherer G. Screening of the 1 Mb SOX9 5' control region by array CGH identifies a large deletion in a case of campomelic dysplasia with XY sex reversal. J Med Genet. 2004;41(4):e47.
  127. Velagaleti GV, Bien-Willner GA, Northup JK, Lockhart LH, Hawkins JC, Jalal SM, Withers M, Lupski JR, Stankiewicz P. Position effects due to chromosome breakpoints that map approximately 900 Kb upstream and approximately 1.3 Mb downstream of SOX9 in two patients with campomelic dysplasia. Am J Hum Genet. 2005;76(4):652-662.
  128. Bagheri-Fam S, Sim H, Bernard P, Jayakody I, Taketo MM, Scherer G, Harley VR. Loss of Fgfr2 leads to partial XY sex reversal. Dev Biol. 2008;314(1):71-83.
  129. Colvin JS, Green RP, Schmahl J, Capel B, Ornitz DM. Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell. 2001;104(6):875-889.
  130. Jameson SA, Lin YT, Capel B. Testis development requires the repression of Wnt4 by Fgf signaling. Dev Biol. 2012;370(1):24-32.
  131. Kim Y, Bingham N, Sekido R, Parker KL, Lovell-Badge R, Capel B. Fibroblast growth factor receptor 2 regulates proliferation and Sertoli differentiation during male sex determination. Proc Natl Acad Sci U S A. 2007;104(42):16558-16563.
  132. Machado AZ, da Silva TE, Frade Costa EM, Dos Santos MG, Nishi MY, Brito VN, Mendonca BB, Domenice S. Absence of inactivating mutations and deletions in the DMRT1 and FGF9 genes in a large cohort of 46,XY patients with gonadal dysgenesis. Eur J Med Genet. 2012;55(12):690-694.
  133. Reardon W, Winter RM, Rutland P, Pulleyn LJ, Jones BM, Malcolm S. Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nat Genet. 1994;8(1):98-103.
  134. Kan SH, Elanko N, Johnson D, Cornejo-Roldan L, Cook J, Reich EW, Tomkins S, Verloes A, Twigg SR, Rannan-Eliya S, McDonald-McGinn DM, Zackai EH, Wall SA, Muenke M, Wilkie AO. Genomic screening of fibroblast growth-factor receptor 2 reveals a wide spectrum of mutations in patients with syndromic craniosynostosis. Am J Hum Genet. 2002;70(2):472-486.
  135. Roscioli T, Elakis G, Cox TC, Moon DJ, Venselaar H, Turner AM, Le T, Hackett E, Haan E, Colley A, Mowat D, Worgan L, Kirk EP, Sachdev R, Thompson E, Gabbett M, McGaughran J, Gibson K, Gattas M, Freckmann ML, Dixon J, Hoefsloot L, Field M, Hackett A, Kamien B, Edwards M, Ades LC, Collins FA, Wilson MJ, Savarirayan R, Tan TY, Amor DJ, McGillivray G, White SM, Glass IA, David DJ, Anderson PJ, Gianoutsos M, Buckley MF. Genotype and clinical care correlations in craniosynostosis: findings from a cohort of 630 Australian and New Zealand patients. Am J Med Genet C Semin Med Genet. 2013;163C(4):259-270.
  136. Shams I, Rohmann E, Eswarakumar VP, Lew ED, Yuzawa S, Wollnik B, Schlessinger J, Lax I. Lacrimo-auriculo-dento-digital syndrome is caused by reduced activity of the fibroblast growth factor 10 (FGF10)-FGF receptor 2 signaling pathway. Mol Cell Biol. 2007;27(19):6903-6912.
  137. Bagheri-Fam S, Ono M, Li L, Zhao L, Ryan J, Lai R, Katsura Y, Rossello FJ, Koopman P, Scherer G, Bartsch O, Eswarakumar JV, Harley VR. FGFR2 mutation in 46,XY sex reversal with craniosynostosis. Hum Mol Genet. 2015;24(23):6699-6710.
  138. Tate G, Satoh H, Endo Y, Mitsuya T. Assignment of desert hedgehog (DHH) to human chromosome bands 12q12-->q13.1 by in situ hybridization. Cytogenet Cell Genet. 2000;88(1-2):93-94.
  139. Yao HH, Whoriskey W, Capel B. Desert Hedgehog/Patched 1 signaling specifies fetal Leydig cell fate in testis organogenesis. Genes Dev. 2002;16(11):1433-1440.
  140. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, Mitsuya T, Osame M. A novel mutation of desert hedgehog in a patient with 46,XY partial gonadal dysgenesis accompanied by minifascicular neuropathy. Am J Hum Genet. 2000;67(5):1302-1305.
  141. Canto P, Soderlund D, Reyes E, Mendez JP. Mutations in the desert hedgehog (DHH) gene in patients with 46,XY complete pure gonadal dysgenesis. J Clin Endocrinol Metab. 2004;89(9):4480-4483.
  142. Canto P, Vilchis F, Soderlund D, Reyes E, Mendez JP. A heterozygous mutation in the desert hedgehog gene in patients with mixed gonadal dysgenesis. Mol Hum Reprod. 2005;11(11):833-836.
  143. Das DK, Sanghavi D, Gawde H, Idicula-Thomas S, Vasudevan L. Novel homozygous mutations in Desert hedgehog gene in patients with 46,XY complete gonadal dysgenesis and prediction of its structural and functional implications by computational methods. Eur J Med Genet. 2011;54(6):e529-534.
  144. Werner R, Merz H, Birnbaum W, Marshall L, Schroder T, Reiz B, Kavran JM, Baumer T, Capetian P, Hiort O. 46,XY Gonadal Dysgenesis due to a Homozygous Mutation in Desert Hedgehog (DHH) Identified by Exome Sequencing. J Clin Endocrinol Metab. 2015;100(7):E1022-1029.
  145. Callier P, Calvel P, Matevossian A, Makrythanasis P, Bernard P, Kurosaka H, Vannier A, Thauvin-Robinet C, Borel C, Mazaud-Guittot S, Rolland A, Desdoits-Lethimonier C, Guipponi M, Zimmermann C, Stévant I, Kuhne F, Conne B, Santoni F, Lambert S, Huet F, Mugneret F, Jaruzelska J, Faivre L, Wilhelm D, Jégou B, Trainor PA, Resh MD, Antonarakis SE, Nef S. Loss of function mutation in the palmitoyl-transferase HHAT leads to syndromic 46,XY disorder of sex development by impeding Hedgehog protein palmitoylation and signaling. PLoS Genet. 2014;10(5):e1004340.
  146. Agha Z, Iqbal Z, Azam M, Ayub H, Vissers LE, Gilissen C, Ali SH, Riaz M, Veltman JA, Pfundt R, van Bokhoven H, Qamar R. Exome sequencing identifies three novel candidate genes implicated in intellectual disability. PLoS One. 2014;9(11):e112687.
  147. Raymond CS, Parker ED, Kettlewell JR, Brown LG, Page DC, Kusz K, Jaruzelska J, Reinberg Y, Flejter WL, Bardwell VJ, Hirsch B, Zarkower D. A region of human chromosome 9p required for testis development contains two genes related to known sexual regulators. Hum Mol Genet. 1999;8(6):989-996.
  148. Raymond CS, Murphy MW, O'Sullivan MG, Bardwell VJ, Zarkower D. Dmrt1, a gene related to worm and fly sexual regulators, is required for mammalian testis differentiation. Genes Dev.2000;14(20):2587-2595.
  149. Raymond CS, Shamu CE, Shen MM, Seifert KJ, Hirsch B, Hodgkin J, Zarkower D. Evidence for evolutionary conservation of sex-determining genes. Nature. 1998;391(6668):691-695.
  150. Muroya K, Okuyama T, Goishi K, Ogiso Y, Fukuda S, Kameyama J, Sato H, Suzuki Y, Terasaki H, Gomyo H, Wakui K, Fukushima Y, Ogata T. Sex-determining gene(s) on distal 9p: clinical and molecular studies in six cases. J Clin Endocrinol Metab. 2000;85(9):3094-3100.
  151. Buonocore F, Clifford-Mobley O, King TFJ, Striglioni N, Man E, Suntharalingham JP, Del Valle I, Lin L, Lagos CF, Rumsby G, Conway GS, Achermann JC. Next-Generation Sequencing Reveals Novel Genetic Variants (SRY, DMRT1, NR5A1, DHH, DHX37) in Adults With 46,XY DSD. J Endocr Soc.2019;3(12):2341-2360.
  152. Zarkower D, Murphy MW. DMRT1: An Ancient Sexual Regulator Required for Human Gonadogenesis. Sex Dev. 2021:1-14.
  153. Gibbons RJ, Higgs DR. Molecular-clinical spectrum of the ATR-X syndrome. Am J Med Genet.2000;97(3):204-212.
  154. Wilkie AO, Gibbons RJ, Higgs DR, Pembrey ME. X linked alpha thalassaemia/mental retardation: spectrum of clinical features in three related males. J Med Genet. 1991;28(11):738-741.
  155. Linhares ND, Valadares ER, da Costa SS, Arantes RR, de Oliveira LR, Rosenberg C, Vianna-Morgante AM, Svartman M. Inherited Xq13.2-q21.31 duplication in a boy with recurrent seizures and pubertal gynecomastia: Clinical, chromosomal and aCGH characterization. Meta Gene. 2016;9:185-190.
  156. Badens C, Lacoste C, Philip N, Martini N, Courrier S, Giuliano F, Verloes A, Munnich A, Leheup B, Burglen L, Odent S, Van Esch H, Levy N. Mutations in PHD-like domain of the ATRX gene correlate with severe psychomotor impairment and severe urogenital abnormalities in patients with ATRX syndrome. Clin Genet. 2006;70(1):57-62.
  157. Gibbons RJ, Wada T, Fisher CA, Malik N, Mitson MJ, Steensma DP, Fryer A, Goudie DR, Krantz ID, Traeger-Synodinos J. Mutations in the chromatin-associated protein ATRX. Hum Mutat. 2008;29(6):796-802.
  158. Tang P, Park DJ, Marshall Graves JA, Harley VR. ATRX and sex differentiation. Trends Endocrinol Metab. 2004;15(7):339-344.
  159. Bogani D, Siggers P, Brixey R, Warr N, Beddow S, Edwards J, Williams D, Wilhelm D, Koopman P, Flavell RA, Chi H, Ostrer H, Wells S, Cheeseman M, Greenfield A. Loss of mitogen-activated protein kinase kinase kinase 4 (MAP3K4) reveals a requirement for MAPK signalling in mouse sex determination. PLoS Biol. 2009;7(9):e1000196.
  160. Gierl MS, Gruhn WH, von Seggern A, Maltry N, Niehrs C. GADD45G functions in male sex determination by promoting p38 signaling and Sry expression. Dev Cell. 2012;23(5):1032-1042.
  161. Warr N, Bogani D, Siggers P, Brixey R, Tateossian H, Dopplapudi A, Wells S, Cheeseman M, Xia Y, Ostrer H, Greenfield A. Minor abnormalities of testis development in mice lacking the gene encoding the MAPK signalling component, MAP3K1. PLoS One. 2011;6(5):e19572.
  162. Charlaftis N, Suddason T, Wu X, Anwar S, Karin M, Gallagher E. The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines. EMBO J. 2014;33(21):2581-2596.
  163. Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC, Tonini G, Ferrante E, Chiumello G, McCabe ER, Fraccaro M, et al. A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet. 1994;7(4):497-501.
  164. Sanlaville D, Vialard F, Thepot F, Vue-Droy L, Ardalan A, Nizard P, Corre A, Devauchelle B, Martin-Denavit T, Nouchy M, Malan V, Taillemite JL, Portnoi MF. Functional disomy of Xp including duplication of DAX1 gene with sex reversal due to t(X;Y)(p21.2;p11.3). Am J Med Genet A. 2004;128A(3):325-330.
  165. Moyses-Oliveira M, Guilherme RS, Meloni VA, Di Battista A, de Mello CB, Bragagnolo S, Moretti-Ferreira D, Kosyakova N, Liehr T, Carvalheira GM, Melaragno MI. X-linked intellectual disability related genes disrupted by balanced X-autosome translocations. Am J Med Genet B Neuropsychiatr Genet.2015;168(8):669-677.
  166. Carrie A, Jun L, Bienvenu T, Vinet MC, McDonell N, Couvert P, Zemni R, Cardona A, Van Buggenhout G, Frints S, Hamel B, Moraine C, Ropers HH, Strom T, Howell GR, Whittaker A, Ross MT, Kahn A, Fryns JP, Beldjord C, Marynen P, Chelly J. A new member of the IL-1 receptor family highly expressed in hippocampus and involved in X-linked mental retardation. Nat Genet. 1999;23(1):25-31.
  167. Barbaro M, Oscarson M, Schoumans J, Staaf J, Ivarsson SA, Wedell A. Isolated 46,XY gonadal dysgenesis in two sisters caused by a Xp21.2 interstitial duplication containing the DAX1 gene. J Clin Endocrinol Metab. 2007;92(8):3305-3313.
  168. Barbaro M, Cicognani A, Balsamo A, Lofgren A, Baldazzi L, Wedell A, Oscarson M. Gene dosage imbalances in patients with 46,XY gonadal DSD detected by an in-house-designed synthetic probe set for multiplex ligation-dependent probe amplification analysis. Clin Genet. 2008;73(5):453-464.
  169. Smyk M, Berg JS, Pursley A, Curtis FK, Fernandez BA, Bien-Willner GA, Lupski JR, Cheung SW, Stankiewicz P. Male-to-female sex reversal associated with an approximately 250 kb deletion upstream of NR0B1 (DAX1). Hum Genet. 2007;122(1):63-70.
  170. White S, Ohnesorg T, Notini A, Roeszler K, Hewitt J, Daggag H, Smith C, Turbitt E, Gustin S, van den Bergen J, Miles D, Western P, Arboleda V, Schumacher V, Gordon L, Bell K, Bengtsson H, Speed T, Hutson J, Warne G, Harley V, Koopman P, Vilain E, Sinclair A. Copy number variation in patients with disorders of sex development due to 46,XY gonadal dysgenesis. PLoS One. 2011;6(3):e17793.
  171. Stark K, Vainio S, Vassileva G, McMahon AP. Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature. 1994;372(6507):679-683.
  172. Elejalde BR, Opitz JM, de Elejalde MM, Gilbert EF, Abellera M, Meisner L, Lebel RR, Hartigan JM. Tandem dup (1p) within the short arm of chromosome 1 in a child with ambiguous genitalia and multiple congenital anomalies. Am J Med Genet. 1984;17(4):723-730.
  173. Jordan BK, Mohammed M, Ching ST, Delot E, Chen XN, Dewing P, Swain A, Rao PN, Elejalde BR, Vilain E. Up-regulation of WNT-4 signaling and dosage-sensitive sex reversal in humans. Am J Hum Genet. 2001;68(5):1102-1109.
  174. McElreavey K, Jorgensen A, Eozenou C, Merel T, Bignon-Topalovic J, Tan DS, Houzelstein D, Buonocore F, Warr N, Kay RGG, Peycelon M, Siffroi JP, Mazen I, Achermann JC, Shcherbak Y, Leger J, Sallai A, Carel JC, Martinerie L, Le Ru R, Conway GS, Mignot B, Van Maldergem L, Bertalan R, Globa E, Brauner R, Jauch R, Nef S, Greenfield A, Bashamboo A. Pathogenic variants in the DEAH-box RNA helicase DHX37 are a frequent cause of 46,XY gonadal dysgenesis and 46,XY testicular regression syndrome. Genet Med. 2020;22(1):150-159.
  175. Le Caignec C, Baron S, McElreavey K, Joubert M, Rival JM, Mechinaud F, David A. 46,XY gonadal dysgenesis: evidence for autosomal dominant transmission in a large kindred. Am J Med Genet A.2003;116A(1):37-43.
  176. Fechner PY, Marcantonio SM, Ogata T, Rosales TO, Smith KD, Goodfellow PN, Migeon CJ, Berkovitz GD. Report of a kindred with X-linked (or autosomal dominant sex-limited) 46,XY partial gonadal dysgenesis. J Clin Endocrinol Metab. 1993;76(5):1248-1253.
  177. Ostrer H. Pathogenic Variants in MAP3K1 Cause 46,XY Gonadal Dysgenesis: A Review. Sex Dev.2022:1-6.
  178. Opitz JM. RSH/SLO ("Smith-Lemli-Opitz") syndrome: historical, genetic, and developmental considerations. Am J Med Genet. 1994;50(4):344-346.
  179. Tint GS, Irons M, Elias ER, Batta AK, Frieden R, Chen TS, Salen G. Defective cholesterol biosynthesis associated with the Smith-Lemli-Opitz syndrome. N Engl J Med. 1994;330(2):107-113.
  180. Fukazawa R, Nakahori Y, Kogo T, Kawakami T, Akamatsu H, Tanae A, Hibi I, Nagafuchi S, Nakagome Y, Hirayama T. Normal Y sequences in Smith-Lemli-Opitz syndrome with total failure of masculinization. Acta Paediatr. 1992;81(6-7):570-572.
  181. Joseph DB, Uehling DT, Gilbert E, Laxova R. Genitourinary abnormalities associated with the Smith-Lemli-Opitz syndrome. J Urol. 1987;137(4):719-721.
  182. Bianconi SE, Cross JL, Wassif CA, Porter FD. Pathogenesis, Epidemiology, Diagnosis and Clinical Aspects of Smith-Lemli-Opitz Syndrome. Expert Opin Orphan Drugs. 2015;3(3):267-280.
  183. Andersson HC, Frentz J, Martinez JE, Tuck-Muller CM, Bellizaire J. Adrenal insufficiency in Smith-Lemli-Opitz syndrome. Am J Med Genet. 1999;82(5):382-384.
  184. Bianconi SE, Conley SK, Keil MF, Sinaii N, Rother KI, Porter FD, Stratakis CA. Adrenal function in Smith-Lemli-Opitz syndrome. Am J Med Genet A. 2011;155A(11):2732-2738.
  185. Correa-Cerro LS, Porter FD. 3beta-hydroxysterol Delta7-reductase and the Smith-Lemli-Opitz syndrome. Mol Genet Metab. 2005;84(2):112-126.
  186. Porter FD. Smith-Lemli-Opitz syndrome: pathogenesis, diagnosis and management. Eur J Hum Genet.2008;16(5):535-541.
  187. Correa-Cerro LS, Wassif CA, Waye JS, Krakowiak PA, Cozma D, Dobson NR, Levin SW, Anadiotis G, Steiner RD, Krajewska-Walasek M, Nowaczyk MJ, Porter FD. DHCR7 nonsense mutations and characterisation of mRNA nonsense mediated decay in Smith-Lemli-Opitz syndrome. J Med Genet.2005;42(4):350-357.
  188. Tierney E, Nwokoro NA, Porter FD, Freund LS, Ghuman JK, Kelley RI. Behavior phenotype in the RSH/Smith-Lemli-Opitz syndrome. Am J Med Genet. 2001;98(2):191-200.
  189. Sikora DM, Ruggiero M, Petit-Kekel K, Merkens LS, Connor WE, Steiner RD. Cholesterol supplementation does not improve developmental progress in Smith-Lemli-Opitz syndrome. J Pediatr.2004;144(6):783-791.
  190. Jira PE, Wevers RA, de Jong J, Rubio-Gozalbo E, Janssen-Zijlstra FS, van Heyst AF, Sengers RC, Smeitink JA. Simvastatin. A new therapeutic approach for Smith-Lemli-Opitz syndrome. J Lipid Res.2000;41(8):1339-1346.
  191. Wassif CA, Kratz L, Sparks SE, Wheeler C, Bianconi S, Gropman A, Calis KA, Kelley RI, Tierney E, Porter FD. A placebo-controlled trial of simvastatin therapy in Smith-Lemli-Opitz syndrome. Genet Med.2017;19(3):297-305.
  192. Pasta S, Akhile O, Tabron D, Ting F, Shackleton C, Watson G. Delivery of the 7-dehydrocholesterol reductase gene to the central nervous system using adeno-associated virus vector in a mouse model of Smith-Lemli-Opitz Syndrome. Mol Genet Metab Rep. 2015;4:92-98.
  193. Misrahi M, Meduri G, Pissard S, Bouvattier C, Beau I, Loosfelt H, Jolivet A, Rappaport R, Milgrom E, Bougneres P. Comparison of immunocytochemical and molecular features with the phenotype in a case of incomplete male pseudohermaphroditism associated with a mutation of the luteinizing hormone receptor. J Clin Endocrinol Metab. 1997;82(7):2159-2165.
  194. Martens JW, Verhoef-Post M, Abelin N, Ezabella M, Toledo SP, Brunner HG, Themmen AP. A homozygous mutation in the luteinizing hormone receptor causes partial Leydig cell hypoplasia: correlation between receptor activity and phenotype. Mol Endocrinol. 1998;12(6):775-784.
  195. Toledo SP, Arnhold IJ, Luthold W, Russo EM, Saldanha PH. Leydig cell hypoplasia determining familial hypergonadotropic hypogonadism. Prog Clin Biol Res. 1985;200:311-314.
  196. Zenteno JC, Canto P, Kofman-Alfaro S, Mendez JP. Evidence for genetic heterogeneity in male pseudohermaphroditism due to Leydig cell hypoplasia. J Clin Endocrinol Metab. 1999;84(10):3803-3806.
  197. Arnhold IJ, de Mendonca BB, Toledo SP, Madureira G, Nicolau W, Bisi H, Bloise W. Leydig cell hypoplasia causing male pseudohermaphroditism: case report and review of the literature. Rev Hosp Clin Fac Med Sao Paulo. 1987;42(5):227-232.
  198. Gromoll J, Eiholzer U, Nieschlag E, Simoni M. Male hypogonadism caused by homozygous deletion of exon 10 of the luteinizing hormone (LH) receptor: differential action of human chorionic gonadotropin and LH. J Clin Endocrinol Metab. 2000;85(6):2281-2286.
  199. Richard N, Leprince C, Gruchy N, Pigny P, Andrieux J, Mittre H, Manouvrier S, Lahlou N, Weill J, Kottler ML. Identification by array-Comparative Genomic Hybridization (array-CGH) of a large deletion of luteinizing hormone receptor gene combined with a missense mutation in a patient diagnosed with a 46,XY disorder of sex development and application to prenatal diagnosis. Endocr J. 2011;58(9):769-776.
  200. Stavrou SS, Zhu YS, Cai LQ, Katz MD, Herrera C, Defillo-Ricart M, Imperato-McGinley J. A novel mutation of the human luteinizing hormone receptor in 46XY and 46XX sisters. J Clin Endocrinol Metab.1998;83(6):2091-2098.
  201. Qiao J, Han B, Liu BL, Chen X, Ru Y, Cheng KX, Chen FG, Zhao SX, Liang J, Lu YL, Tang JF, Wu YX, Wu WL, Chen JL, Chen MD, Song HD. A splice site mutation combined with a novel missense mutation of LHCGR cause male pseudohermaphroditism. Hum Mutat. 2009;30(9):E855-865.
  202. Zhou HX. Effect of mixed macromolecular crowding agents on protein folding. Proteins.2008;72(4):1109-1113.
  203. Yariz KO, Walsh T, Uzak A, Spiliopoulos M, Duman D, Onalan G, King MC, Tekin M. Inherited mutation of the luteinizing hormone/choriogonadotropin receptor (LHCGR) in empty follicle syndrome. Fertil Steril. 2011;96(2):e125-130.
  204. Aktar Karakaya A, Çayır A, Unal E, Beştaş A, Ece Solmaz A, Kenan Haspolat Y. A rare cause of primary amenorrhea: LHCGR gene mutations. Eur J Obstet Gynecol Reprod Biol. 2022;272:193-197.
  205. Latronico AC, Arnhold IJ. Inactivating mutations of the human luteinizing hormone receptor in both sexes. Semin Reprod Med. 2012;30(5):382-386.
  206. Latronico AC, Anasti J, Arnhold IJ, Rapaport R, Mendonca BB, Bloise W, Castro M, Tsigos C, Chrousos GP. Brief report: testicular and ovarian resistance to luteinizing hormone caused by inactivating mutations of the luteinizing hormone-receptor gene. N Engl J Med. 1996;334(8):507-512.
  207. Segaloff DL. Diseases associated with mutations of the human lutropin receptor. Prog Mol Biol Transl Sci. 2009;89:97-114.
  208. Kossack N, Simoni M, Richter-Unruh A, Themmen AP, Gromoll J. Mutations in a novel, cryptic exon of the luteinizing hormone/chorionic gonadotropin receptor gene cause male pseudohermaphroditism. PLoS Med. 2008;5(4):e88.
  209. Arnhold IJ, Latronico AC, Batista MC, Izzo CR, Mendonca BB. Clinical features of women with resistance to luteinizing hormone. Clin Endocrinol (Oxf). 1999;51(6):701-707.
  210. Bruysters M, Christin-Maitre S, Verhoef-Post M, Sultan C, Auger J, Faugeron I, Larue L, Lumbroso S, Themmen AP, Bouchard P. A new LH receptor splice mutation responsible for male hypogonadism with subnormal sperm production in the propositus, and infertility with regular cycles in an affected sister. Hum Reprod. 2008;23(8):1917-1923.
  211. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol. 2018;179(3):R125-R141.
  212. Prader A, Gurtner HP. [The syndrome of male pseudohermaphrodism in congenital adrenocortical hyperplasia without overproduction of androgens (adrenal male pseudohermaphrodism)]. Helv Paediatr Acta. 1955;10(4):397-412.
  213. Miller WL. Molecular biology of steroid hormone synthesis. Endocr Rev. 1988;9(3):295-318.
  214. Hauffa BP, Miller WL, Grumbach MM, Conte FA, Kaplan SL. Congenital adrenal hyperplasia due to deficient cholesterol side-chain cleavage activity (20, 22-desmolase) in a patient treated for 18 years. Clin Endocrinol (Oxf). 1985;23(5):481-493.
  215. Bose HS, Sugawara T, Strauss JF, 3rd, Miller WL, International Congenital Lipoid Adrenal Hyperplasia C. The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. N Engl J Med.1996;335(25):1870-1878.
  216. Abdulhadi-Atwan M, Jean A, Chung WK, Meir K, Ben Neriah Z, Stratigopoulos G, Oberfield SE, Fennoy I, Hirsch HJ, Bhangoo A, Ten S, Lerer I, Zangen DH. Role of a founder c.201_202delCT mutation and new phenotypic features of congenital lipoid adrenal hyperplasia in Palestinians. J Clin Endocrinol Metab. 2007;92(10):4000-4008.
  217. Fujieda K, Tajima T, Nakae J, Sageshima S, Tachibana K, Suwa S, Sugawara T, Strauss JF, 3rd. Spontaneous puberty in 46,XX subjects with congenital lipoid adrenal hyperplasia. Ovarian steroidogenesis is spared to some extent despite inactivating mutations in the steroidogenic acute regulatory protein (StAR) gene. J Clin Invest. 1997;99(6):1265-1271.
  218. Hasegawa T, Zhao L, Caron KM, Majdic G, Suzuki T, Shizawa S, Sasano H, Parker KL. Developmental roles of the steroidogenic acute regulatory protein (StAR) as revealed by StAR knockout mice. Mol Endocrinol. 2000;14(9):1462-1471.
  219. Sugawara T, Lin D, Holt JA, Martin KO, Javitt NB, Miller WL, Strauss JF, 3rd. Structure of the human steroidogenic acute regulatory protein (StAR) gene: StAR stimulates mitochondrial cholesterol 27-hydroxylase activity. Biochemistry. 1995;34(39):12506-12512.
  220. Miller WL. Congenital lipoid adrenal hyperplasia: the human gene knockout for the steroidogenic acute regulatory protein. J Mol Endocrinol. 1997;19(3):227-240.
  221. Baker BY, Yaworsky DC, Miller WL. A pH-dependent molten globule transition is required for activity of the steroidogenic acute regulatory protein, StAR. J Biol Chem. 2005;280(50):41753-41760.
  222. Baker BY, Epand RF, Epand RM, Miller WL. Cholesterol binding does not predict activity of the steroidogenic acute regulatory protein, StAR. J Biol Chem. 2007;282(14):10223-10232.
  223. Lin D, Sugawara T, Strauss JF, 3rd, Clark BJ, Stocco DM, Saenger P, Rogol A, Miller WL. Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science.1995;267(5205):1828-1831.
  224. Baker BY, Lin L, Kim CJ, Raza J, Smith CP, Miller WL, Achermann JC. Nonclassic congenital lipoid adrenal hyperplasia: a new disorder of the steroidogenic acute regulatory protein with very late presentation and normal male genitalia. J Clin Endocrinol Metab. 2006;91(12):4781-4785.
  225. Nakae J, Tajima T, Sugawara T, Arakane F, Hanaki K, Hotsubo T, Igarashi N, Igarashi Y, Ishii T, Koda N, Kondo T, Kohno H, Nakagawa Y, Tachibana K, Takeshima Y, Tsubouchi K, Strauss JF, 3rd, Fujieda K. Analysis of the steroidogenic acute regulatory protein (StAR) gene in Japanese patients with congenital lipoid adrenal hyperplasia. Hum Mol Genet. 1997;6(4):571-576.
  226. Fluck CE, Pandey AV, Dick B, Camats N, Fernandez-Cancio M, Clemente M, Gussinye M, Carrascosa A, Mullis PE, Audi L. Characterization of novel StAR (steroidogenic acute regulatory protein) mutations causing non-classic lipoid adrenal hyperplasia. PLoS One. 2011;6(5):e20178.
  227. Sahakitrungruang T, Soccio RE, Lang-Muritano M, Walker JM, Achermann JC, Miller WL. Clinical, genetic, and functional characterization of four patients carrying partial loss-of-function mutations in the steroidogenic acute regulatory protein (StAR). J Clin Endocrinol Metab. 2010;95(7):3352-3359.
  228. Metherell LA, Naville D, Halaby G, Begeot M, Huebner A, Nurnberg G, Nurnberg P, Green J, Tomlinson JW, Krone NP, Lin L, Racine M, Berney DM, Achermann JC, Arlt W, Clark AJ. Nonclassic lipoid congenital adrenal hyperplasia masquerading as familial glucocorticoid deficiency. J Clin Endocrinol Metab. 2009;94(10):3865-3871.
  229. Ishii T, Hori N, Amano N, Aya M, Shibata H, Katsumata N, Hasegawa T. Pubertal and Adult Testicular Functions in Nonclassic Lipoid Congenital Adrenal Hyperplasia: A Case Series and Review. J Endocr Soc. 2019;3(7):1367-1374.
  230. Morohashi K, Fujii-Kuriyama Y, Okada Y, Sogawa K, Hirose T, Inayama S, Omura T. Molecular cloning and nucleotide sequence of cDNA for mRNA of mitochondrial cytochrome P-450(SCC) of bovine adrenal cortex. Proc Natl Acad Sci U S A. 1984;81(15):4647-4651.
  231. Hiort O, Holterhus PM, Werner R, Marschke C, Hoppe U, Partsch CJ, Riepe FG, Achermann JC, Struve D. Homozygous disruption of P450 side-chain cleavage (CYP11A1) is associated with prematurity, complete 46,XY sex reversal, and severe adrenal failure. J Clin Endocrinol Metab. 2005;90(1):538-541.
  232. Tajima T, Fujieda K, Kouda N, Nakae J, Miller WL. Heterozygous mutation in the cholesterol side chain cleavage enzyme (p450scc) gene in a patient with 46,XY sex reversal and adrenal insufficiency. J Clin Endocrinol Metab. 2001;86(8):3820-3825.
  233. Katsumata N, Ohtake M, Hojo T, Ogawa E, Hara T, Sato N, Tanaka T. Compound heterozygous mutations in the cholesterol side-chain cleavage enzyme gene (CYP11A) cause congenital adrenal insufficiency in humans. J Clin Endocrinol Metab. 2002;87(8):3808-3813.
  234. Kim CJ, Lin L, Huang N, Quigley CA, AvRuskin TW, Achermann JC, Miller WL. Severe combined adrenal and gonadal deficiency caused by novel mutations in the cholesterol side chain cleavage enzyme, P450scc. J Clin Endocrinol Metab. 2008;93(3):696-702.
  235. Rubtsov P, Karmanov M, Sverdlova P, Spirin P, Tiulpakov A. A novel homozygous mutation in CYP11A1 gene is associated with late-onset adrenal insufficiency and hypospadias in a 46,XY patient. J Clin Endocrinol Metab. 2009;94(3):936-939.
  236. Sahakitrungruang T, Tee MK, Blackett PR, Miller WL. Partial defect in the cholesterol side-chain cleavage enzyme P450scc (CYP11A1) resembling nonclassic congenital lipoid adrenal hyperplasia. J Clin Endocrinol Metab. 2011;96(3):792-798.
  237. Mason JI, Ushijima K, Doody KM, Nagai K, Naville D, Head JR, Milewich L, Rainey WE, Ralph MM. Regulation of expression of the 3 beta-hydroxysteroid dehydrogenases of human placenta and fetal adrenal. J Steroid Biochem Mol Biol. 1993;47(1-6):151-159.
  238. Bongiovanni AM. The adrenogenital syndrome with deficiency of 3 beta-hydroxysteroid dehydrogenase. J Clin Invest. 1962;41:2086-2092.
  239. Kalfa N, Gaspari L, Ollivier M, Philibert P, Bergougnoux A, Paris F, Sultan C. Molecular genetics of hypospadias and cryptorchidism recent developments. Clin Genet. 2019;95(1):122-131.
  240. Guran T, Kara C, Yildiz M, Bitkin EC, Haklar G, Lin JC, Keskin M, Barnard L, Anik A, Catli G, Guven A, Kirel B, Tutunculer F, Onal H, Turan S, Akcay T, Atay Z, Yilmaz GC, Mamadova J, Akbarzade A, Sirikci O, Storbeck KH, Baris T, Chung BC, Bereket A. Revisiting Classical 3β-hydroxysteroid Dehydrogenase 2 Deficiency: Lessons from 31 Pediatric Cases. J Clin Endocrinol Metab. 2020;105(3).
  241. Mendonça BB, Russell AJ, Vasconcelos-Leite M, Arnhold IJ, Bloise W, Wajchenberg BL, Nicolau W, Sutcliffe RG, Wallace AM. Mutation in 3 beta-hydroxysteroid dehydrogenase type II associated with pseudohermaphroditism in males and premature pubarche or cryptic expression in females. J Mol Endocrinol. 1994;12(1):119-122.
  242. Sutcliffe RG, Russell AJ, Edwards CR, Wallace AM. Human 3 beta-hydroxysteroid dehydrogenase: genes and phenotypes. J Mol Endocrinol. 1996;17(1):1-5.
  243. Simard J, Ricketts ML, Gingras S, Soucy P, Feltus FA, Melner MH. Molecular biology of the 3beta-hydroxysteroid dehydrogenase/delta5-delta4 isomerase gene family. Endocr Rev. 2005;26(4):525-582.
  244. Russell AJ, Wallace AM, Forest MG, Donaldson MD, Edwards CR, Sutcliffe RG. Mutation in the human gene for 3 beta-hydroxysteroid dehydrogenase type II leading to male pseudohermaphroditism without salt loss. J Mol Endocrinol. 1994;12(2):225-237.
  245. Mendonca BB, Bloise W, Arnhold IJ, Batista MC, Toledo SP, Drummond MC, Nicolau W, Mattar E. Male pseudohermaphroditism due to nonsalt-losing 3 beta-hydroxysteroid dehydrogenase deficiency: gender role change and absence of gynecomastia at puberty. J Steroid Biochem. 1987;28(6):669-675.
  246. Ladjouze A, Donaldson M, Plotton I, Djenane N, Mohammedi K, Tardy-Guidollet V, Mallet D, Boulesnane K, Bouzerar Z, Morel Y, Roucher-Boulez F. Genotype, Mortality, Morbidity, and Outcomes of 3β-Hydroxysteroid Dehydrogenase Deficiency in Algeria. Front Endocrinol (Lausanne).2022;13:867073.
  247. Miller WL. The syndrome of 17,20 lyase deficiency. J Clin Endocrinol Metab. 2012;97(1):59-67.
  248. Biglieri EG, Herron MA, Brust N. 17-hydroxylation deficiency in man. J Clin Invest. 1966;45(12):1946-1954.
  249. New MI. Male pseudohermaphroditism due to 17 alpha-hydroxylase deficiency. J Clin Invest.1970;49(10):1930-1941.
  250. Auchus RJ. The uncommon forms of congenital adrenal hyperplasia. Curr Opin Endocrinol Diabetes Obes. 2022;29(3):263-270.
  251. Yanase T, Simpson ER, Waterman MR. 17 alpha-hydroxylase/17,20-lyase deficiency: from clinical investigation to molecular definition. Endocr Rev. 1991;12(1):91-108.
  252. Auchus RJ. The genetics, pathophysiology, and management of human deficiencies of P450c17. Endocrinol Metab Clin North Am. 2001;30(1):101-119, vii.
  253. Zachmann M. Recent aspects of steroid biosynthesis in male sex differentiation. Clinical studies. Horm Res. 1992;38(5-6):211-216.
  254. Martin RM, Lin CJ, Costa EM, de Oliveira ML, Carrilho A, Villar H, Longui CA, Mendonca BB. P450c17 deficiency in Brazilian patients: biochemical diagnosis through progesterone levels confirmed by CYP17 genotyping. J Clin Endocrinol Metab. 2003;88(12):5739-5746.
  255. Matteson KJ, Picado-Leonard J, Chung BC, Mohandas TK, Miller WL. Assignment of the gene for adrenal P450c17 (steroid 17 alpha-hydroxylase/17,20 lyase) to human chromosome 10. J Clin Endocrinol Metab. 1986;63(3):789-791.
  256. Rosa S, Duff C, Meyer M, Lang-Muritano M, Balercia G, Boscaro M, Topaloglu AK, Mioni R, Fallo F, Zuliani L, Mantero F, Schoenle EJ, Biason-Lauber A. P450c17 deficiency: clinical and molecular characterization of six patients. J Clin Endocrinol Metab. 2007;92(3):1000-1007.
  257. Peterson RE, Imperato-McGinley J, Gautier T, Shackleton C. Male pseudohermaphroditism due to multiple defects in steroid-biosynthetic microsomal mixed-function oxidases. A new variant of congenital adrenal hyperplasia. N Engl J Med. 1985;313(19):1182-1191.
  258. Arlt W, Walker EA, Draper N, Ivison HE, Ride JP, Hammer F, Chalder SM, Borucka-Mankiewicz M, Hauffa BP, Malunowicz EM, Stewart PM, Shackleton CH. Congenital adrenal hyperplasia caused by mutant P450 oxidoreductase and human androgen synthesis: analytical study. Lancet.2004;363(9427):2128-2135.
  259. Shephard EA, Phillips IR, Santisteban I, West LF, Palmer CN, Ashworth A, Povey S. Isolation of a human cytochrome P-450 reductase cDNA clone and localization of the corresponding gene to chromosome 7q11.2. Ann Hum Genet. 1989;53(4):291-301.
  260. Reardon W, Smith A, Honour JW, Hindmarsh P, Das D, Rumsby G, Nelson I, Malcolm S, Ades L, Sillence D, Kumar D, DeLozier-Blanchet C, McKee S, Kelly T, McKeehan WL, Baraitser M, Winter RM. Evidence for digenic inheritance in some cases of Antley-Bixler syndrome? J Med Genet.2000;37(1):26-32.
  261. Huang N, Pandey AV, Agrawal V, Reardon W, Lapunzina PD, Mowat D, Jabs EW, Van Vliet G, Sack J, Fluck CE, Miller WL. Diversity and function of mutations in p450 oxidoreductase in patients with Antley-Bixler syndrome and disordered steroidogenesis. Am J Hum Genet. 2005;76(5):729-749.
  262. Schmidt K, Hughes C, Chudek JA, Goodyear SR, Aspden RM, Talbot R, Gundersen TE, Blomhoff R, Henderson C, Wolf CR, Tickle C. Cholesterol metabolism: the main pathway acting downstream of cytochrome P450 oxidoreductase in skeletal development of the limb. Mol Cell Biol. 2009;29(10):2716-2729.
  263. Finkielstain GP, Vieites A, Bergadá I, Rey RA. Disorders of Sex Development of Adrenal Origin. Front Endocrinol (Lausanne). 2021;12:770782.
  264. Idkowiak J, O'Riordan S, Reisch N, Malunowicz EM, Collins F, Kerstens MN, Kohler B, Graul-Neumann LM, Szarras-Czapnik M, Dattani M, Silink M, Shackleton CH, Maiter D, Krone N, Arlt W. Pubertal presentation in seven patients with congenital adrenal hyperplasia due to P450 oxidoreductase deficiency. J Clin Endocrinol Metab. 2011;96(3):E453-462.
  265. Zachmann M, Vollmin JA, Hamilton W, Prader A. Steroid 17,20-desmolase deficiency: a new cause of male pseudohermaphroditism. Clin Endocrinol (Oxf). 1972;1(4):369-385.
  266. Auchus RJ. Steroid 17-hydroxylase and 17,20-lyase deficiencies, genetic and pharmacologic. J Steroid Biochem Mol Biol. 2017;165(Pt A):71-78.
  267. Geller DH, Auchus RJ, Miller WL. P450c17 mutations R347H and R358Q selectively disrupt 17,20-lyase activity by disrupting interactions with P450 oxidoreductase and cytochrome b5. Mol Endocrinol.1999;13(1):167-175.
  268. Hegesh E, Hegesh J, Kaftory A. Congenital methemoglobinemia with a deficiency of cytochrome b5. N Engl J Med. 1986;314(12):757-761.
  269. Idkowiak J, Randell T, Dhir V, Patel P, Shackleton CH, Taylor NF, Krone N, Arlt W. A missense mutation in the human cytochrome b5 gene causes 46,XY disorder of sex development due to true isolated 17,20 lyase deficiency. J Clin Endocrinol Metab. 2012;97(3):E465-475.
  270. Saez JM, De Peretti E, Morera AM, David M, Bertrand J. Familial male pseudohermaphroditism with gynecomastia due to a testicular 17-ketosteroid reductase defect. I. Studies in vivo. J Clin Endocrinol Metab. 1971;32(5):604-610.
  271. Boehmer AL, Brinkmann AO, Sandkuijl LA, Halley DJ, Niermeijer MF, Andersson S, de Jong FH, Kayserili H, de Vroede MA, Otten BJ, Rouwé CW, Mendonça BB, Rodrigues C, Bode HH, de Ruiter PE, Delemarre-van de Waal HA, Drop SL. 17Beta-hydroxysteroid dehydrogenase-3 deficiency: diagnosis, phenotypic variability, population genetics, and worldwide distribution of ancient and de novo mutations. J Clin Endocrinol Metab. 1999;84(12):4713-4721.
  272. George MM, New MI, Ten S, Sultan C, Bhangoo A. The clinical and molecular heterogeneity of 17betaHSD-3 enzyme deficiency. Horm Res Paediatr. 2010;74(4):229-240.
  273. Andersson S, Moghrabi N. Physiology and molecular genetics of 17 beta-hydroxysteroid dehydrogenases. Steroids. 1997;62(1):143-147.
  274. Andersson S, Geissler WM, Wu L, Davis DL, Grumbach MM, New MI, Schwarz HP, Blethen SL, Mendonca BB, Bloise W, Witchel SF, Cutler GB, Griffin JE, Wilson JD, Russel DW. Molecular genetics and pathophysiology of 17 beta-hydroxysteroid dehydrogenase 3 deficiency. J Clin Endocrinol Metab.1996;81(1):130-136.
  275. Lee YS, Kirk JM, Stanhope RG, Johnston DI, Harland S, Auchus RJ, Andersson S, Hughes IA. Phenotypic variability in 17beta-hydroxysteroid dehydrogenase-3 deficiency and diagnostic pitfalls. Clin Endocrinol (Oxf). 2007;67(1):20-28.
  276. Mendonca BB, Gomes NL, Costa EM, Inacio M, Martin RM, Nishi MY, Carvalho FM, Tibor FD, Domenice S. 46,XY disorder of sex development (DSD) due to 17β-hydroxysteroid dehydrogenase type 3 deficiency. J Steroid Biochem Mol Biol. 2017;165(Pt A):79-85.
  277. Mendonca BB, Inacio M, Arnhold IJ, Costa EM, Bloise W, Martin RM, Denes FT, Silva FA, Andersson S, Lindqvist A, Wilson JD. Male pseudohermaphroditism due to 17 beta-hydroxysteroid dehydrogenase 3 deficiency. Diagnosis, psychological evaluation, and management. Medicine (Baltimore).2000;79(5):299-309.
  278. Bertelloni S, Balsamo A, Giordani L, Fischetto R, Russo G, Delvecchio M, Gennari M, Nicoletti A, Maggio MC, Concolino D, Cavallo L, Cicognani A, Chiumello G, Hiort O, Baroncelli GI, Faienza MF. 17beta-Hydroxysteroid dehydrogenase-3 deficiency: from pregnancy to adolescence. J Endocrinol Invest. 2009;32(8):666-670.
  279. Khattab A, Yuen T, Yau M, Domenice S, Frade Costa EM, Diya K, Muhuri D, Pina CE, Nishi MY, Yang AC, de Mendonça BB, New MI. Pitfalls in hormonal diagnosis of 17-beta hydroxysteroid dehydrogenase III deficiency. J Pediatr Endocrinol Metab. 2015;28(5-6):623-628.
  280. McKeever BM, Hawkins BK, Geissler WM, Wu L, Sheridan RP, Mosley RT, Andersson S. Amino acid substitution of arginine 80 in 17beta-hydroxysteroid dehydrogenase type 3 and its effect on NADPH cofactor binding and oxidation/reduction kinetics. Biochim Biophys Acta. 2002;1601(1):29-37.
  281. Cocchetti C, Baldinotti F, Romani A, Ristori J, Mazzoli F, Vignozzi L, Maggi M, Fisher AD. A Novel Compound Heterozygous Mutation of HSD17B3 Gene Identified in a Patient With 46,XY Difference of Sexual Development. Sex Med. 2022;10(4):100522.
  282. von Spreckelsen B, Aksglaede L, Johannsen TH, Nielsen JE, Main KM, Jørgensen A, Jensen RB. Prepubertal and pubertal gonadal morphology, expression of cell lineage markers and hormonal evaluation in two 46,XY siblings with 17β-hydroxysteroid dehydrogenase 3 deficiency. J Pediatr Endocrinol Metab. 2022;35(7):953-961.
  283. Jahagirdar R, Khadilkar V, Deshpande R, Lohiya N. Clinical, Etiological and Laboratory Profile of Children with Disorders of Sexual Development (DSD)-Experience from a Tertiary Pediatric Endocrine Unit in Western India. Indian J Endocrinol Metab. 2021;25(1):48-53.
  284. Manyas H, Eroğlu Filibeli B, Ayrancı İ, Güvenç MS, Dündar BN, Çatlı G. Early and late diagnoses of 17β-Hydroxysteroid dehydrogenase type-3 deficiency in two unrelated patients. Andrologia.2021;53(6):e14017.
  285. Rösler A, Kohn G. Male pseudohermaphroditism due to 17 beta-hydroxysteroid dehydrogenase deficiency: studies on the natural history of the defect and effect of androgens on gender role. J Steroid Biochem. 1983;19(1B):663-674.
  286. Cools M, Drop SL, Wolffenbuttel KP, Oosterhuis JW, Looijenga LH. Germ cell tumors in the intersex gonad: old paths, new directions, moving frontiers. Endocr Rev. 2006;27(5):468-484.
  287. Looijenga LH, Hersmus R, Oosterhuis JW, Cools M, Drop SL, Wolffenbuttel KP. Tumor risk in disorders of sex development (DSD). Best Pract Res Clin Endocrinol Metab. 2007;21(3):480-495.
  288. Kathrins M, Kolon TF. Malignancy in disorders of sex development. Transl Androl Urol. 2016;5(5):794-798.
  289. Abacı A, Çatlı G, Berberoğlu M. Gonadal malignancy risk and prophylactic gonadectomy in disorders of sexual development. J Pediatr Endocrinol Metab. 2015;28(9-10):1019-1027.
  290. Auchus RJ. The backdoor pathway to dihydrotestosterone. Trends Endocrinol Metab. 2004;15(9):432-438.
  291. Wilson JD, Auchus RJ, Leihy MW, Guryev OL, Estabrook RW, Osborn SM, Shaw G, Renfree MB. 5alpha-androstane-3alpha,17beta-diol is formed in tammar wallaby pouch young testes by a pathway involving 5alpha-pregnane-3alpha,17alpha-diol-20-one as a key intermediate. Endocrinology.2003;144(2):575-580.
  292. Lee HG, Kim CJ. Classic and backdoor pathways of androgen biosynthesis in human sexual development. Ann Pediatr Endocrinol Metab. 2022;27(2):83-89.
  293. Biswas MG, Russell DW. Expression cloning and characterization of oxidative 17beta- and 3alpha-hydroxysteroid dehydrogenases from rat and human prostate. J Biol Chem. 1997;272(25):15959-15966.
  294. Dufort I, Soucy P, Labrie F, Luu-The V. Molecular cloning of human type 3 3 alpha-hydroxysteroid dehydrogenase that differs from 20 alpha-hydroxysteroid dehydrogenase by seven amino acids. Biochem Biophys Res Commun. 1996;228(2):474-479.
  295. Fluck CE, Meyer-Boni M, Pandey AV, Kempna P, Miller WL, Schoenle EJ, Biason-Lauber A. Why boys will be boys: two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation. Am J Hum Genet. 2011;89(2):201-218.
  296. Mares L, Vilchis F, Chavez B, Ramos L. Molecular genetic analysis of AKR1C2-4 and HSD17B6 genes in subjects 46,XY with hypospadias. J Pediatr Urol. 2020;16(5):689 e681-689 e612.
  297. Penning TM. The aldo-keto reductases (AKRs): Overview. Chem Biol Interact. 2015;234:236-246.
  298. NOWAKOWSKI H, LENZ W. Genetic aspects in male hypogonadism. Recent Prog Horm Res.1961;17:53-95.
  299. Imperato-McGinley J, Guerrero L, Gautier T, Peterson RE. Steroid 5alpha-reductase deficiency in man: an inherited form of male pseudohermaphroditism. Science. 1974;186(4170):1213-1215.
  300. Walsh PC, Madden JD, Harrod MJ, Goldstein JL, MacDonald PC, Wilson JD. Familial incomplete male pseudohermaphroditism, type 2. Decreased dihydrotestosterone formation in pseudovaginal perineoscrotal hypospadias. N Engl J Med. 1974;291(18):944-949.
  301. Andersson S, Russell DW. Structural and biochemical properties of cloned and expressed human and rat steroid 5 alpha-reductases. Proc Natl Acad Sci U S A. 1990;87(10):3640-3644.
  302. Imperato-McGinley J, Miller M, Wilson JD, Peterson RE, Shackleton C, Gajdusek DC. A cluster of male pseudohermaphrodites with 5 alpha-reductase deficiency in Papua New Guinea. Clin Endocrinol (Oxf).1991;34(4):293-298.
  303. Thigpen AE, Davis DL, Milatovich A, Mendonca BB, Imperato-McGinley J, Griffin JE, Francke U, Wilson JD, Russell DW. Molecular genetics of steroid 5 alpha-reductase 2 deficiency. J Clin Invest.1992;90(3):799-809.
  304. Batista RL, Mendonca BB. Integrative and Analytical Review of the 5-Alpha-Reductase Type 2 Deficiency Worldwide. Appl Clin Genet. 2020;13:83-96.
  305. Batista RL, Mendonca BB. The Molecular Basis of 5alpha-Reductase Type 2 Deficiency. Sex Dev.2022:1-13.
  306. Mendonca BB, Batista RL, Domenice S, Costa EM, Arnhold IJ, Russell DW, Wilson JD. Steroid 5α-reductase 2 deficiency. J Steroid Biochem Mol Biol. 2016;163:206-211.
  307. Avendaño A, Paradisi I, Cammarata-Scalisi F, Callea M. 5-α-Reductase type 2 deficiency: is there a genotype-phenotype correlation? A review. Hormones (Athens). 2018;17(2):197-204.
  308. Gui B, Song Y, Su Z, Luo FH, Chen L, Wang X, Chen R, Yang Y, Wang J, Zhao X, Fan L, Liu X, Wang Y, Chen S, Gong C. New insights into 5α-reductase type 2 deficiency based on a multi-centre study: regional distribution and genotype-phenotype profiling of. J Med Genet. 2019;56(10):685-692.
  309. Berra M, Williams EL, Muroni B, Creighton SM, Honour JW, Rumsby G, Conway GS. Recognition of 5α-reductase-2 deficiency in an adult female 46XY DSD clinic. Eur J Endocrinol. 2011;164(6):1019-1025.
  310. Shabir I, Khurana ML, Joseph AA, Eunice M, Mehta M, Ammini AC. Phenotype, genotype and gender identity in a large cohort of patients from India with 5α-reductase 2 deficiency. Andrology.2015;3(6):1132-1139.
  311. Cheng J, Lin R, Zhang W, Liu G, Sheng H, Li X, Zhou Z, Mao X, Liu L. Phenotype and molecular characteristics in 45 Chinese children with 5α-reductase type 2 deficiency from South China. Clin Endocrinol (Oxf). 2015;83(4):518-526.
  312. Mendonca BB, Inacio M, Costa EM, Arnhold IJ, Silva FA, Nicolau W, Bloise W, Russel DW, Wilson JD. Male pseudohermaphroditism due to steroid 5alpha-reductase 2 deficiency. Diagnosis, psychological evaluation, and management. Medicine (Baltimore). 1996;75(2):64-76.
  313. Bertelloni S, Baldinotti F, Russo G, Ghirri P, Dati E, Michelucci A, Moscuzza F, Meroni S, Colombo I, Sessa MR, Baroncelli GI. 5α-Reductase-2 Deficiency: Clinical Findings, Endocrine Pitfalls, and Genetic Features in a Large Italian Cohort. Sex Dev. 2016;10(1):28-36.
  314. Vilchis F, Méndez JP, Canto P, Lieberman E, Chávez B. Identification of missense mutations in the SRD5A2 gene from patients with steroid 5alpha-reductase 2 deficiency. Clin Endocrinol (Oxf).2000;52(3):383-387.
  315. Imperato-McGinley J. 5alpha-reductase-2 deficiency and complete androgen insensitivity: lessons from nature. Adv Exp Med Biol. 2002;511:121-131; discussion 131-124.
  316. Walter KN, Kienzle FB, Frankenschmidt A, Hiort O, Wudy SA, van der Werf-Grohmann N, Superti-Furga A, Schwab KO. Difficulties in diagnosis and treatment of 5alpha-reductase type 2 deficiency in a newborn with 46,XY DSD. Horm Res Paediatr. 2010;74(1):67-71.
  317. Hochberg Z, Chayen R, Reiss N, Falik Z, Makler A, Munichor M, Farkas A, Goldfarb H, Ohana N, Hiort O. Clinical, biochemical, and genetic findings in a large pedigree of male and female patients with 5 alpha-reductase 2 deficiency. J Clin Endocrinol Metab. 1996;81(8):2821-2827.
  318. Costa EM, Domenice S, Sircili MH, Inacio M, Mendonca BB. DSD due to 5α-reductase 2 deficiency - from diagnosis to long term outcome. Semin Reprod Med. 2012;30(5):427-431.
  319. Mendonca BB, Batista RL, Domenice S, Costa EM, Arnhold IJ, Russell DW, Wilson JD. Reprint of "Steroid 5α-reductase 2 deficiency". J Steroid Biochem Mol Biol. 2017;165(Pt A):95-100.
  320. Gomes NL, Batista RL, Nishi MY, Lerario AM, Silva TE, Narcizo AM, Benedetti AFF, Funari MFA, Junior JAF, Moraes DR, Quintao LML, Montenegro LR, Ferrari MTM, Jorge AA, Arnhold IJP, Costa EMF, Domenice S, Mendonca BB. Contribution of clinical and genetic approaches for diagnosing 209 index cases with 46,XY Differences of Sex Development. J Clin Endocrinol Metab. 2022.
  321. Chan AO, But BW, Lee CY, Lam YY, Ng KL, Tung JY, Kwan EY, Chan YK, Tsui TK, Lam AL, Tse WY, Cheung PT, Shek CC. Diagnosis of 5α-reductase 2 deficiency: is measurement of dihydrotestosterone essential? Clin Chem. 2013;59(5):798-806.
  322. Cohen-Kettenis PT. Psychosocial and psychosexual aspects of disorders of sex development. Best Pract Res Clin Endocrinol Metab. 2010;24(2):325-334.
  323. Ahmed SF, Achermann JC, Arlt W, Balen A, Conway G, Edwards Z, Elford S, Hughes IA, Izatt L, Krone N, Miles H, O'Toole S, Perry L, Sanders C, Simmonds M, Watt A, Willis D. Society for Endocrinology UK guidance on the initial evaluation of an infant or an adolescent with a suspected disorder of sex development (Revised 2015). Clin Endocrinol (Oxf). 2016;84(5):771-788.
  324. Costa EM, Mendonca BB, Inácio M, Arnhold IJ, Silva FA, Lodovici O. Management of ambiguous genitalia in pseudohermaphrodites: new perspectives on vaginal dilation. Fertil Steril. 1997;67(2):229-232.
  325. Sircili MH, e Silva FA, Costa EM, Brito VN, Arnhold IJ, Dénes FT, Inacio M, de Mendonca BB. Long-term surgical outcome of masculinizing genitoplasty in large cohort of patients with disorders of sex development. J Urol. 2010;184(3):1122-1127.
  326. Cohen-Kettenis PT. Gender change in 46,XY persons with 5alpha-reductase-2 deficiency and 17beta-hydroxysteroid dehydrogenase-3 deficiency. Arch Sex Behav. 2005;34(4):399-410.
  327. Loch Batista R, Inácio M, Prado Arnhold IJ, Gomes NL, Diniz Faria JA, Rodrigues de Moraes D, Frade Costa EM, Domenice S, Bilharinho Mendonça B. Psychosexual Aspects, Effects of Prenatal Androgen Exposure, and Gender Change in 46,XY Disorders of Sex Development. J Clin Endocrinol Metab.2019;104(4):1160-1170.
  328. Meyer-Bahlburg HF, Baratz Dalke K, Berenbaum SA, Cohen-Kettenis PT, Hines M, Schober JM. Gender Assignment, Reassignment and Outcome in Disorders of Sex Development: Update of the 2005 Consensus Conference. Horm Res Paediatr. 2016;85(2):112-118.
  329. Fisher AD, Ristori J, Fanni E, Castellini G, Forti G, Maggi M. Gender identity, gender assignment and reassignment in individuals with disorders of sex development: a major of dilemma. J Endocrinol Invest.2016;39(11):1207-1224.
  330. Mendonca BB. Gender assignment in patients with disorder of sex development. Curr Opin Endocrinol Diabetes Obes. 2014;21(6):511-514.
  331. Amaral RC, Inacio M, Brito VN, Bachega TA, Domenice S, Arnhold IJ, Madureira G, Gomes L, Costa EM, Mendonca BB. Quality of life of patients with 46,XX and 46,XY disorders of sex development. Clin Endocrinol (Oxf). 2015;82(2):159-164.
  332. Cassia Amaral R, Inacio M, Brito VN, Bachega TA, Oliveira AA, Domenice S, Denes FT, Sircili MH, Arnhold IJ, Madureira G, Gomes L, Costa EM, Mendonca BB. Quality of life in a large cohort of adult Brazilian patients with 46,XX and 46,XY disorders of sex development from a single tertiary centre. Clin Endocrinol (Oxf). 2015;82(2):274-279.
  333. Batista RL, Costa EMF, Rodrigues AS, Gomes NL, Faria JA, Nishi MY, Arnhold IJP, Domenice S, Mendonca BB. Androgen insensitivity syndrome: a review. Arch Endocrinol Metab. 2018;62(2):227-235.
  334. Hiort O. Clinical and molecular aspects of androgen insensitivity. Endocr Dev. 2013;24:33-40.
  335. Mongan NP, Tadokoro-Cuccaro R, Bunch T, Hughes IA. Androgen insensitivity syndrome. Best Pract Res Clin Endocrinol Metab. 2015;29(4):569-580.
  336. Hughes IA, Werner R, Bunch T, Hiort O. Androgen insensitivity syndrome. Semin Reprod Med.2012;30(5):432-442.
  337. Batista RL, Craveiro FL, Ramos RM, Mendonca BB. Mild Androgen Insensitivity Syndrome: The Current Landscape. Endocr Pract. 2022.
  338. Yu X, Yi P, Hamilton RA, Shen H, Chen M, Foulds CE, Mancini MA, Ludtke SJ, Wang Z, O'Malley BW. Structural Insights of Transcriptionally Active, Full-Length Androgen Receptor Coactivator Complexes. Mol Cell. 2020;79(5):812-823.e814.
  339. Clinckemalie L, Vanderschueren D, Boonen S, Claessens F. The hinge region in androgen receptor control. Mol Cell Endocrinol. 2012;358(1):1-8.
  340. Schlanger S, Heemers HV. Functional Studies on Steroid Receptors. Methods Mol Biol. 2018;1786:117-130.
  341. Tan MH, Li J, Xu HE, Melcher K, Yong EL. Androgen receptor: structure, role in prostate cancer and drug discovery. Acta Pharmacol Sin. 2015;36(1):3-23.
  342. Chmelar R, Buchanan G, Need EF, Tilley W, Greenberg NM. Androgen receptor coregulators and their involvement in the development and progression of prostate cancer. Int J Cancer. 2007;120(4):719-733.
  343. Heinlein CA, Chang C. Androgen receptor (AR) coregulators: an overview. Endocr Rev.2002;23(2):175-200.
  344. Tirabassi G, Cignarelli A, Perrini S, Delli Muti N, Furlani G, Gallo M, Pallotti F, Paoli D, Giorgino F, Lombardo F, Gandini L, Lenzi A, Balercia G. Influence of CAG Repeat Polymorphism on the Targets of Testosterone Action. Int J Endocrinol. 2015;2015:298107.
  345. Huang G, Shan W, Zeng L, Huang L. Androgen receptor gene CAG repeat polymorphism and risk of isolated hypospadias: results from a meta-analysis. Genet Mol Res. 2015;14(1):1580-1588.
  346. Malek EG, Salameh JS, Makki A. Kennedy's disease: an under-recognized motor neuron disorder. Acta Neurol Belg. 2020;120(6):1289-1295.
  347. Paz-Y-Miño C, Robles P, Salazar C, Leone PE, García-Cárdenas JM, Naranjo M, López-Cortés A. Positive association of the androgen receptor CAG repeat length polymorphism with the risk of prostate cancer. Mol Med Rep. 2016;14(2):1791-1798.
  348. Gottlieb B, Beitel LK, Nadarajah A, Paliouras M, Trifiro M. The androgen receptor gene mutations database: 2012 update. Hum Mutat. 2012;33(5):887-894.
  349. Hornig NC, Holterhus PM. Molecular basis of androgen insensitivity syndromes. Mol Cell Endocrinol.2021;523:111146.
  350. Batista RL, Yamaguchi K, di Santi Rodrigues A, Nishi MY, Goodier JL, Carvalho LR, Domenice S, Costa EMF, Hazazian H, Mendonca BB. Mobile DNA in Endocrinology: LINE-1 retrotransposon causing Partial Androgen Insensitivity Syndrome. J Clin Endocrinol Metab. 2019.
  351. Hornig NC, de Beaufort C, Denzer F, Cools M, Wabitsch M, Ukat M, Kulle AE, Schweikert HU, Werner R, Hiort O, Audi L, Siebert R, Ammerpohl O, Holterhus PM. A Recurrent Germline Mutation in the 5'UTR of the Androgen Receptor Causes Complete Androgen Insensitivity by Activating Aberrant uORF Translation. PLoS One. 2016;11(4):e0154158.
  352. Batista RL, di Santi Rodrigues A, Nishi MY, Gomes NLRA, Faria JAD, de Moraes DR, Carvalho LR, Frade EMC, Domenice S, de Mendonca BB. A Recurrent Synonymous Mutation in the Human Androgen Receptor Gene Causing Complete Androgen Insensitivity Syndrome. J Steroid Biochem Mol Biol. 2017.
  353. Känsäkoski J, Jääskeläinen J, Jääskeläinen T, Tommiska J, Saarinen L, Lehtonen R, Hautaniemi S, Frilander MJ, Palvimo JJ, Toppari J, Raivio T. Complete androgen insensitivity syndrome caused by a deep intronic pseudoexon-activating mutation in the androgen receptor gene. Sci Rep. 2016;6:32819.
  354. Hornig NC, Ukat M, Schweikert HU, Hiort O, Werner R, Drop SL, Cools M, Hughes IA, Audi L, Ahmed SF, Demiri J, Rodens P, Worch L, Wehner G, Kulle AE, Dunstheimer D, Müller-Roßberg E, Reinehr T, Hadidi AT, Eckstein AK, van der Horst C, Seif C, Siebert R, Ammerpohl O, Holterhus PM. Identification of an AR Mutation-Negative Class of Androgen Insensitivity by Determining Endogenous AR Activity. J Clin Endocrinol Metab. 2016;101(11):4468-4477.
  355. Riskin A, Koren I, Bader D, Grün M, Dar H, Leibovitz Z, Kugelman A, Hiort O. The approach to a neonate with a possible prenatal diagnosis of androgen insensitivity syndrome. J Pediatr Endocrinol Metab. 2006;19(12):1437-1443.
  356. Batista RL. Complete Androgen Insensitivity in Girls with Inguinal Hernias: A Serendipity Opportunity for Early Diagnosis. J Invest Surg. 2019:1-2.
  357. Oakes MB, Eyvazzadeh AD, Quint E, Smith YR. Complete androgen insensitivity syndrome--a review. J Pediatr Adolesc Gynecol. 2008;21(6):305-310.
  358. Döhnert U, Wünsch L, Hiort O. Gonadectomy in Complete Androgen Insensitivity Syndrome: Why and When? Sex Dev. 2017.
  359. Tack LJW, Maris E, Looijenga LHJ, Hannema SE, Audi L, Köhler B, Holterhus PM, Riedl S, Wisniewski A, Flück CE, Davies JH, T Apos Sjoen G, Lucas-Herald AK, Evliyaoglu O, Krone N, Iotova V, Marginean O, Balsamo A, Verkauskas G, Weintrob N, Ellaithi M, Nordenström A, Verrijn Stuart A, Kluivers KB, Wolffenbuttel KP, Ahmed SF, Cools M. Management of Gonads in Adults with Androgen Insensitivity: An International Survey. Horm Res Paediatr. 2018:1-11.
  360. Cools M, Looijenga L. Update on the Pathophysiology and Risk Factors for the Development of Malignant Testicular Germ Cell Tumors in Complete Androgen Insensitivity Syndrome. Sex Dev. 2017.
  361. Deans R, Creighton SM, Liao LM, Conway GS. Timing of gonadectomy in adult women with complete androgen insensitivity syndrome (CAIS): patient preferences and clinical evidence. Clin Endocrinol (Oxf). 2012;76(6):894-898.
  362. Danilovic DL, Correa PH, Costa EM, Melo KF, Mendonca BB, Arnhold IJ. Height and bone mineral density in androgen insensitivity syndrome with mutations in the androgen receptor gene. Osteoporos Int. 2007;18(3):369-374.
  363. Lucas-Herald A, Bertelloni S, Juul A, Bryce J, Jiang J, Rodie M, Sinnott R, Boroujerdi M, Lindhardt-Johansen M, Hiort O, Holterhus PM, Cools M, Guaragna-Filho G, Guerra-Junior G, Weintrob N, Hannema S, Drop S, Guran T, Darendeliler F, Nordenstrom A, Hughes IA, Acerini C, Tadokoro-Cuccaro R, Ahmed SF. The Long Term Outcome Of Boys With Partial Androgen Insensitivity Syndrome And A Mutation In The Androgen Receptor Gene. J Clin Endocrinol Metab. 2016:jc20161372.
  364. Gulía C, Baldassarra S, Zangari A, Briganti V, Gigli S, Gaffi M, Signore F, Vallone C, Nucciotti R, Costantini FM, Pizzuti A, Bernardo S, Porrello A, Piergentili R. Androgen insensitivity syndrome. Eur Rev Med Pharmacol Sci. 2018;22(12):3873-3887.
  365. Josso N, di Clemente N, Gouedard L. Anti-Mullerian hormone and its receptors. Mol Cell Endocrinol.2001;179(1-2):25-32.
  366. Rey R. Anti-Müllerian hormone in disorders of sex determination and differentiation. Arq Bras Endocrinol Metabol. 2005;49(1):26-36.
  367. Loeff DS, Imbeaud S, Reyes HM, Meller JL, Rosenthal IM. Surgical and genetic aspects of persistent mullerian duct syndrome. J Pediatr Surg. 1994;29(1):61-65.
  368. Josso N, di Clemente N. TGF-beta Family Members and Gonadal Development. Trends Endocrinol Metab. 1999;10(6):216-222.
  369. Imbeaud S, Carre-Eusebe D, Rey R, Belville C, Josso N, Picard JY. Molecular genetics of the persistent mullerian duct syndrome: a study of 19 families. Hum Mol Genet. 1994;3(1):125-131.
  370. Imbeaud S, Faure E, Lamarre I, Mattei MG, di Clemente N, Tizard R, Carre-Eusebe D, Belville C, Tragethon L, Tonkin C, Nelson J, McAuliffe M, Bidart JM, Lababidi A, Josso N, Cate RL, Picard JY. Insensitivity to anti-mullerian hormone due to a mutation in the human anti-mullerian hormone receptor. Nat Genet. 1995;11(4):382-388.
  371. Orós-Millán ME, Muñoz-Calvo MT, Nishi MY, Bilharinho Mendonca B, Argente J. [Persistent Müllerian duct syndrome due to a mutation in the anti-Müllerian hormone receptor gene (AMHR2)]. An Pediatr (Barc). 2016.
  372. Saleem M, Ather U, Mirza B, Iqbal S, Sheikh A, Shaukat M, Sheikh MT, Ahmad F, Rehan T. Persistent mullerian duct syndrome: A 24-year experience. J Pediatr Surg. 2016;51(10):1721-1724.
  373. Aarskog D. Maternal progestins as a possible cause of hypospadias. N Engl J Med. 1979;300(2):75-78.
  374. Driscoll SG, Taylor SH. Effects of prenatal maternal estrogen on the male urogenital system. Obstet Gynecol. 1980;56(5):537-542.
  375. Watanabe M, Yoshida R, Ueoka K, Aoki K, Sasagawa I, Hasegawa T, Sueoka K, Kamatani N, Yoshimura Y, Ogata T. Haplotype analysis of the estrogen receptor 1 gene in male genital and reproductive abnormalities. Hum Reprod. 2007;22(5):1279-1284.
  376. Rider CV, Furr J, Wilson VS, Gray LE, Jr. A mixture of seven antiandrogens induces reproductive malformations in rats. Int J Androl. 2008;31(2):249-262.
  377. Vilela ML, Willingham E, Buckley J, Liu BC, Agras K, Shiroyanagi Y, Baskin LS. Endocrine disruptors and hypospadias: role of genistein and the fungicide vinclozolin. Urology. 2007;70(3):618-621.
  378. Fredell L, Lichtenstein P, Pedersen NL, Svensson J, Nordenskjold A. Hypospadias is related to birth weight in discordant monozygotic twins. J Urol. 1998;160(6 Pt 1):2197-2199.
  379. Francois I, van Helvoirt M, de Zegher F. Male pseudohermaphroditism related to complications at conception, in early pregnancy or in prenatal growth. Horm Res. 1999;51(2):91-95.
  380. Mendonca BB, Billerbeck AE, de Zegher F. Nongenetic male pseudohermaphroditism and reduced prenatal growth. N Engl J Med. 2001;345(15):1135.
  381. Rossignol S, Netchine I, Le Bouc Y, Gicquel C. Epigenetics in Silver-Russell syndrome. Best Pract Res Clin Endocrinol Metab. 2008;22(3):403-414.
  382. Morel Y, Rey R, Teinturier C, Nicolino M, Michel-Calemard L, Mowszowicz I, Jaubert F, Fellous M, Chaussain JL, Chatelain P, David M, Nihoul-Fekete C, Forest MG, Josso N. Aetiological diagnosis of male sex ambiguity: a collaborative study. Eur J Pediatr. 2002;161(1):49-59.
  383. Main KM, Jensen RB, Asklund C, Hoi-Hansen CE, Skakkebaek NE. Low birth weight and male reproductive function. Horm Res. 2006;65 Suppl 3:116-122.
  384. Leitao Braga B, Lisboa Gomes N, Nishi MY, Freire BL, Batista RL, JA DFJ, Funari MFA, Figueredo Benedetti AF, de Moraes Narcizo A, Cavalca Cardoso L, Lerario AM, Guerra-Junior G, Frade Costa EM, Domenice S, Jorge AAL, Mendonca BB. Variants in 46,XY DSD-Related Genes in Syndromic and Non-Syndromic Small for Gestational Age Children with Hypospadias. Sex Dev. 2022;16(1):27-33.
  385. Scarpa MG, Grazia MD, Tornese G. 46,XY ovotesticular disorders of sex development: A therapeutic challenge. Pediatr Rep. 2017;9(4):7085.
  386. van der Horst HJ, de Wall LL. Hypospadias, all there is to know. Eur J Pediatr. 2017;176(4):435-441.
  387. Mole RJ, Nash S, MacKenzie DN. Hypospadias. BMJ. 2020;369:m2070.
  388. Hutson JM. Cryptorchidism and Hypospadias. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  389. Fukami M, Wada Y, Miyabayashi K, Nishino I, Hasegawa T, Nordenskjold A, Camerino G, Kretz C, Buj-Bello A, Laporte J, Yamada G, Morohashi K, Ogata T. CXorf6 is a causative gene for hypospadias. Nat Genet. 2006;38(12):1369-1371.
  390. Ogata T, Sano S, Nagata E, Kato F, Fukami M. MAMLD1 and 46,XY disorders of sex development. Semin Reprod Med. 2012;30(5):410-416.
  391. Beleza-Meireles A, Tohonen V, Soderhall C, Schwentner C, Radmayr C, Kockum I, Nordenskjold A. Activating transcription factor 3: a hormone responsive gene in the etiology of hypospadias. Eur J Endocrinol. 2008;158(5):729-739.
  392. Batool A, Karimi N, Wu XN, Chen SR, Liu YX. Testicular germ cell tumor: a comprehensive review. Cell Mol Life Sci. 2019;76(9):1713-1727.
  393. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol. 2016;70(1):93-105.
  394. Berney DM, Looijenga LH, Idrees M, Oosterhuis JW, Rajpert-De Meyts E, Ulbright TM, Skakkebaek NE. Germ cell neoplasia in situ (GCNIS): evolution of the current nomenclature for testicular pre-invasive germ cell malignancy. Histopathology. 2016;69(1):7-10.
  395. Fink C, Baal N, Wilhelm J, Sarode P, Weigel R, Schumacher V, Nettersheim D, Schorle H, Schrock C, Bergmann M, Kliesch S, Kressin M, Savai R. On the origin of germ cell neoplasia in situ: Dedifferentiation of human adult Sertoli cells in cross talk with seminoma cells in vitro. Neoplasia.2021;23(7):731-742.
  396. Morin J, Peard L, Vanadurongvan T, Walker J, Donmez MI, Saltzman AF. Oncologic outcomes of pre-malignant and invasive germ cell tumors in patients with differences in sex development - A systematic review. J Pediatr Urol. 2020;16(5):576-582.
  397. Pierconti F, Martini M, Grande G, Larocca LM, Sacco E, Pugliese D, Gulino G, Bassi PF, Milardi D, Pontecorvi A. Germ Cell Neoplasia in situ (GCNIS) in Testis-Sparing Surgery (TSS) for Small Testicular Masses (STMs). Front Endocrinol (Lausanne). 2019;10:512.
  398. von der Maase H, Rorth M, Walbom-Jorgensen S, Sorensen BL, Christophersen IS, Hald T, Jacobsen GK, Berthelsen JG, Skakkebaek NE. Carcinoma in situ of contralateral testis in patients with testicular germ cell cancer: study of 27 cases in 500 patients. Br Med J (Clin Res Ed). 1986;293(6559):1398-1401.
  399. Jørgensen A, Lindhardt Johansen M, Juul A, Skakkebaek NE, Main KM, Rajpert-De Meyts E. Pathogenesis of germ cell neoplasia in testicular dysgenesis and disorders of sex development. Semin Cell Dev Biol. 2015;45:124-137.
  400. Baxter RM, Arboleda VA, Lee H, Barseghyan H, Adam MP, Fechner PY, Bargman R, Keegan C, Travers S, Schelley S, Hudgins L, Mathew RP, Stalker HJ, Zori R, Gordon OK, Ramos-Platt L, Pawlikowska-Haddal A, Eskin A, Nelson SF, Délot E, Vilain E. Exome sequencing for the diagnosis of 46,XY disorders of sex development. J Clin Endocrinol Metab. 2015;100(2):E333-344.
  401. Cools M, Pleskacova J, Stoop H, Hoebeke P, Van Laecke E, Drop SL, Lebl J, Oosterhuis JW, Looijenga LH, Wolffenbuttel KP, Group MC. Gonadal pathology and tumor risk in relation to clinical characteristics in patients with 45,X/46,XY mosaicism. J Clin Endocrinol Metab. 2011;96(7):E1171-1180.
  402. Spoor JA, Oosterhuis JW, Hersmus R, Biermann K, Wolffenbuttel KP, Cools M, Kazmi Z, Ahmed SF, Looijenga LHJ. Histological Assessment of Gonads in DSD: Relevance for Clinical Management. Sex Dev. 2018;12(1-3):106-122.
  403. Palma I, Garibay N, Pena-Yolanda R, Contreras A, Raya A, Dominguez C, Romero M, Aristi G, Queipo G. Utility of OCT3/4, TSPY and β-catenin as biological markers for gonadoblastoma formation and malignant germ cell tumor development in dysgenetic gonads. Dis Markers. 2013;34(6):419-424.
  404. Granados A, Alaniz VI, Mohnach L, Barseghyan H, Vilain E, Ostrer H, Quint EH, Chen M, Keegan CE. MAP3K1-related gonadal dysgenesis: Six new cases and review of the literature. Am J Med Genet C Semin Med Genet. 2017;175(2):253-259.
  405. Ferguson L, Agoulnik AI. Testicular cancer and cryptorchidism. Front Endocrinol (Lausanne). 2013;4:32.
  406. Leão R, Ahmad AE, Hamilton RJ. Testicular Cancer Biomarkers: A Role for Precision Medicine in Testicular Cancer. Clin Genitourin Cancer. 2019;17(1):e176-e183.
  407. Morin J, Peard L, Saltzman AF. Gonadal malignancy in patients with differences of sex development. Transl Androl Urol. 2020;9(5):2408-2415.
  408. Khan S, Mannel L, Koopman CL, Chimpiri R, Hansen KR, Craig LB. The use of MRI in the pre-surgical evaluation of patients with androgen insensitivity syndrome. J Pediatr Adolesc Gynecol.2014;27(1):e17-20.
  409. Rajpert-De Meyts E, Nielsen JE, Skakkebaek NE, Almstrup K. Diagnostic markers for germ cell neoplasms: from placental-like alkaline phosphatase to micro-RNAs. Folia Histochem Cytobiol.2015;53(3):177-188.
  410. Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, Nagel R, Liu YP, van Duijse J, Drost J, Griekspoor A, Zlotorynski E, Yabuta N, De Vita G, Nojima H, Looijenga LH, Agami R. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell.2006;124(6):1169-1181.
  411. Leao R, Albersen M, Looijenga LHJ, Tandstad T, Kollmannsberger C, Murray MJ, Culine S, Coleman N, Belge G, Hamilton RJ, Dieckmann KP. Circulating MicroRNAs, the Next-Generation Serum Biomarkers in Testicular Germ Cell Tumours: A Systematic Review. Eur Urol. 2021;80(4):456-466.
  412. Abaci A, Catli G, Berberoglu M. Gonadal malignancy risk and prophylactic gonadectomy in disorders of sexual development. J Pediatr Endocrinol Metab. 2015;28(9-10):1019-1027.
  413. Wünsch L, Holterhus PM, Wessel L, Hiort O. Patients with disorders of sex development (DSD) at risk of gonadal tumour development: management based on laparoscopic biopsy and molecular diagnosis. BJU Int. 2012;110(11 Pt C):E958-965.
  414. Barros BA, Oliveira LR, Surur CRC, Barros-Filho AA, Maciel-Guerra AT, Guerra-Junior G. Complete androgen insensitivity syndrome and risk of gonadal malignancy: systematic review. Ann Pediatr Endocrinol Metab. 2021;26(1):19-23.
  415. Patel V, Casey RK, Gomez-Lobo V. Timing of Gonadectomy in Patients with Complete Androgen Insensitivity Syndrome-Current Recommendations and Future Directions. J Pediatr Adolesc Gynecol.2016;29(4):320-325.
  416. Weidler EM, Linnaus ME, Baratz AB, Goncalves LF, Bailey S, Hernandez SJ, Gomez-Lobo V, van Leeuwen K. A Management Protocol for Gonad Preservation in Patients with Androgen Insensitivity Syndrome. J Pediatr Adolesc Gynecol. 2019;32(6):605-611.
  417. McNeill SA, O'Donnell M, Donat R, Lessells A, Hargreave TB. Estrogen secretion from a malignant sex cord stromal tumor in a patient with complete androgen insensitivity. Am J Obstet Gynecol.1997;177(6):1541-1542.
  418. Guercio G, Rey RA. Fertility issues in the management of patients with disorders of sex development. Endocr Dev. 2014;27:87-98.
  419. Van Batavia JP, Kolon TF. Fertility in disorders of sex development: A review. J Pediatr Urol.2016;12(6):418-425.
  420. Carson SA, Kallen AN. Diagnosis and Management of Infertility: A Review. JAMA. 2021;326(1):65-76.
  421. Foli KJ, VanGraafeiland B, Snethen JA, Greenberg CS. Caring for nontraditional families: Kinship, foster, and adoptive. J Spec Pediatr Nurs. 2022;27(3):e12388.
  422. King TF, Conway GS. Swyer syndrome. Curr Opin Endocrinol Diabetes Obes. 2014;21(6):504-510.
  423. Tordjman KM, Yaron M, Berkovitz A, Botchan A, Sultan C, Lumbroso S. Fertility after high-dose testosterone and intracytoplasmic sperm injection in a patient with androgen insensitivity syndrome with a previously unreported androgen receptor mutation. Andrologia. 2014;46(6):703-706.
  424. Matsubara K, Iwamoto H, Yoshida A, Ogata T. Semen analysis and successful paternity by intracytoplasmic sperm injection in a man with steroid 5α-reductase-2 deficiency. Fertil Steril.2010;94(7):2770.e2777-2710.
  425. Nordenskjöld A, Ivarsson SA. Molecular characterization of 5 alpha-reductase type 2 deficiency and fertility in a Swedish family. J Clin Endocrinol Metab. 1998;83(9):3236-3238.
  426. Bertelloni S, Baldinotti F, Baroncelli GI, Caligo MA, Peroni D. Paternity in 5α-Reductase-2 Deficiency: Report of Two Brothers with Spontaneous or Assisted Fertility and Literature Review. Sex Dev.2019;13(2):55-59.
  427. Guercio G, Costanzo M, Grinspon RP, Rey RA. Fertility Issues in Disorders of Sex Development. Endocrinol Metab Clin North Am. 2015;44(4):867-881.
  428. Finlayson C, Fritsch MK, Johnson EK, Rosoklija I, Gosiengfiao Y, Yerkes E, Madonna MB, Woodruff TK, Cheng E. Presence of Germ Cells in Disorders of Sex Development: Implications for Fertility Potential and Preservation. J Urol. 2017;197(3 Pt 2):937-943.
  429. Słowikowska-Hilczer J, Hirschberg AL, Claahsen-van der Grinten H, Reisch N, Bouvattier C, Thyen U, Cohen Kettenis P, Roehle R, Köhler B, Nordenström A, Group d-L. Fertility outcome and information on fertility issues in individuals with different forms of disorders of sex development: findings from the dsd-LIFE study. Fertil Steril. 2017;108(5):822-831.
  430. Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, Wallace WH, Wang ET, Loren AW. Fertility Preservation in Patients With Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36(19):1994-2001.
  431. Sadri-Ardekani H, Atala A. Testicular tissue cryopreservation and spermatogonial stem cell transplantation to restore fertility: from bench to bedside. Stem Cell Res Ther. 2014;5(3):68.
  432. Michel A, Mormont C, Legros JJ. A psycho-endocrinological overview of transsexualism. Eur J Endocrinol. 2001;145(4):365-376.
  433. Drescher J, Cohen-Kettenis P, Winter S. Minding the body: situating gender identity diagnoses in the ICD-11. Int Rev Psychiatry. 2012;24(6):568-577.
  434. Asscheman H, Giltay EJ, Megens JA, de Ronde WP, van Trotsenburg MA, Gooren LJ. A long-term follow-up study of mortality in transsexuals receiving treatment with cross-sex hormones. Eur J Endocrinol. 2011;164(4):635-642.
  435. Mustanski B, Liu RT. A longitudinal study of predictors of suicide attempts among lesbian, gay, bisexual, and transgender youth. Arch Sex Behav. 2013;42(3):437-448.
  436. Wylie K, Knudson G, Khan SI, Bonierbale M, Watanyusakul S, Baral S. Serving transgender people: clinical care considerations and service delivery models in transgender health. Lancet.2016;388(10042):401-411.
  437. Hembree WC, Cohen-Kettenis PT, Gooren L, Hannema SE, Meyer WJ, Murad MH, Rosenthal SM, Safer JD, Tangpricha V, T'Sjoen GG. Endocrine Treatment of Gender-Dysphoric/Gender-Incongruent Persons: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab.2017;102(11):3869-3903.
  438. Hembree WC, Cohen-Kettenis P, Delemarre-van de Waal HA, Gooren LJ, Meyer WJ, 3rd, Spack NP, Tangpricha V, Montori VM, Endocrine S. Endocrine treatment of transsexual persons: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2009;94(9):3132-3154.
  439. Costa EM, Mendonca BB. Clinical management of transsexual subjects. Arq Bras Endocrinol Metabol.2014;58(2):188-196.
  440. Toorians AW, Thomassen MC, Zweegman S, Magdeleyns EJ, Tans G, Gooren LJ, Rosing J. Venous thrombosis and changes of hemostatic variables during cross-sex hormone treatment in transsexual people. J Clin Endocrinol Metab. 2003;88(12):5723-5729.
  441. Jequier AM, Bullimore NJ, Bishop MJ. Cyproterone acetate and a small dose of oestrogen in the pre-operative management of male transsexuals. A report of three cases. Andrologia. 1989;21(5):456-461.
  442. Cunha FS, Domenice S, Camara VL, Sircili MH, Gooren LJ, Mendonca BB, Costa EM. Diagnosis of prolactinoma in two male-to-female transsexual subjects following high-dose cross-sex hormone therapy. Andrologia. 2015;47(6):680-684.
  443. Gooren LJ. Clinical practice. Care of transsexual persons. N Engl J Med. 2011;364(13):1251-1257.
  444. Wilson JD, Rivarola MA, Mendonca BB, Warne GL, Josso N, Drop SL, Grumbach MM. Advice on the management of ambiguous genitalia to a young endocrinologist from experienced clinicians. Semin Reprod Med. 2012;30(5):339-350.
  445. Achermann JC, Domenice S, Bachega TA, Nishi MY, Mendonca BB. Disorders of sex development: effect of molecular diagnostics. Nat Rev Endocrinol. 2015;11(8):478-488.
  446. Hiort O, Birnbaum W, Marshall L, Wünsch L, Werner R, Schröder T, Döhnert U, Holterhus PM. Management of disorders of sex development. Nat Rev Endocrinol. 2014;10(9):520-529.
  447. Bennecke E, Werner-Rosen K, Thyen U, Kleinemeier E, Lux A, Jürgensen M, Grüters A, Köhler B. Subjective need for psychological support (PsySupp) in parents of children and adolescents with disorders of sex development (dsd). Eur J Pediatr. 2015;174(10):1287-1297.
  448. Markosyan R, Ahmed SF. Sex Assignment in Conditions Affecting Sex Development. J Clin Res Pediatr Endocrinol. 2017;9(Suppl 2):106-112.
  449. Sandberg DE, Callens N, Wisniewski AB. Disorders of Sex Development (DSD): Networking and Standardization Considerations. Horm Metab Res. 2015;47(5):387-393.
  450. Streuli JC, Vayena E, Cavicchia-Balmer Y, Huber J. Shaping parents: impact of contrasting professional counseling on parents' decision making for children with disorders of sex development. J Sex Med.2013;10(8):1953-1960.
  451. Ediati A, Maharani N, Utari A. Sociocultural aspects of disorders of sex development. Birth Defects Res C Embryo Today. 2016;108(4):380-383.
  452. Moshiri M, Chapman T, Fechner PY, Dubinsky TJ, Shnorhavorian M, Osman S, Bhargava P, Katz DS. Evaluation and management of disorders of sex development: multidisciplinary approach to a complex diagnosis. Radiographics. 2012;32(6):1599-1618.
  453. Massanyi EZ, Dicarlo HN, Migeon CJ, Gearhart JP. Review and management of 46,XY disorders of sex development. J Pediatr Urol. 2013;9(3):368-379.
  454. Lee PA, Houk CP, Ahmed SF, Hughes IA. Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics. 2006;118(2):e488-500.
  455. Hewitt J, Zacharin M. Hormone replacement in disorders of sex development: Current thinking. Best Pract Res Clin Endocrinol Metab. 2015;29(3):437-447.
  456. Birnbaum W, Bertelloni S. Sex hormone replacement in disorders of sex development. Endocr Dev.2014;27:149-159.
  457. Crandall CJ, Hovey KM, Andrews C, Cauley JA, Stefanick M, Shufelt C, Prentice RL, Kaunitz AM, Eaton C, Wactawski-Wende J, Manson JE. Comparison of clinical outcomes among users of oral and transdermal estrogen therapy in the Women's Health Initiative Observational Study. Menopause.2017;24(10):1145-1153.
  458. Adami S, Rossini M, Zamberlan N, Bertoldo F, Dorizzi R, Lo Cascio V. Long-term effects of transdermal and oral estrogens on serum lipids and lipoproteins in postmenopausal women. Maturitas.1993;17(3):191-196.
  459. Ankarberg-Lindgren C, Kriström B, Norjavaara E. Physiological estrogen replacement therapy for puberty induction in girls: a clinical observational study. Horm Res Paediatr. 2014;81(4):239-244.
  460. Cools M, Nordenström A, Robeva R, Hall J, Westerveld P, Flück C, Köhler B, Berra M, Springer A, Schweizer K, Pasterski V, 1 CABwg. Caring for individuals with a difference of sex development (DSD): a Consensus Statement. Nat Rev Endocrinol. 2018;14(7):415-429.
  461. Schonbucher V, Schweizer K, Richter-Appelt H. Sexual quality of life of individuals with disorders of sex development and a 46,XY karyotype: a review of international research. J Sex Marital Ther.2010;36(3):193-215.
  462. Minto CL, Liao KL, Conway GS, Creighton SM. Sexual function in women with complete androgen insensitivity syndrome. Fertil Steril. 2003;80(1):157-164.
  463. Birnbaum W, Marshall L, Werner R, Kulle A, Holterhus PM, Rall K, Köhler B, Richter-Unruh A, Hartmann MF, Wudy SA, Auer MK, Lux A, Kropf S, Hiort O. Oestrogen versus androgen in hormone-replacement therapy for complete androgen insensitivity syndrome: a multicentre, randomised, double-dummy, double-blind crossover trial. Lancet Diabetes Endocrinol. 2018;6(10):771-780.
  464. Batista RL, Mendonca BB. Testosterone replacement in androgen insensitivity: is there an advantage? Ann Transl Med. 2018;6(Suppl 1):S85.
  465. Khen-Dunlop N, Lortat-Jacob S, Thibaud E, Clement-Ziza M, Lyonnet S, Nihoul-Fekete C. Rokitansky syndrome: clinical experience and results of sigmoid vaginoplasty in 23 young girls. J Urol.2007;177(3):1107-1111.
  466. Werner R, Grötsch H, Hiort O. 46,XY disorders of sex development--the undermasculinised male with disorders of androgen action. Best Pract Res Clin Endocrinol Metab. 2010;24(2):263-277.
  467. McGriff NJ, Csako G, Kabbani M, Diep L, Chrousos GP, Pucino F. Treatment options for a patient experiencing pruritic rash associated with transdermal testosterone: a review of the literature. Pharmacotherapy. 2001;21(11):1425-1435.
  468. El-Maouche D, Arlt W, Merke DP. Congenital adrenal hyperplasia. Lancet. 2017;390(10108):2194-2210.
  469. Fleming L, Van Riper M, Knafl K. Management of Childhood Congenital Adrenal Hyperplasia-An Integrative Review of the Literature. J Pediatr Health Care. 2017;31(5):560-577.
  470. Mouriquand PD, Gorduza DB, Gay CL, Meyer-Bahlburg HF, Baker L, Baskin LS, Bouvattier C, Braga LH, Caldamone AC, Duranteau L, El Ghoneimi A, Hensle TW, Hoebeke P, Kaefer M, Kalfa N, Kolon TF, Manzoni G, Mure PY, Nordenskjöld A, Pippi Salle JL, Poppas DP, Ransley PG, Rink RC, Rodrigo R, Sann L, Schober J, Sibai H, Wisniewski A, Wolffenbuttel KP, Lee P. Surgery in disorders of sex development (DSD) with a gender issue: If (why), when, and how? J Pediatr Urol. 2016;12(3):139-149.
  471. Creighton S, Chernausek SD, Romao R, Ransley P, Salle JP. Timing and nature of reconstructive surgery for disorders of sex development - introduction. J Pediatr Urol. 2012;8(6):602-610.
  472. Bennecke E, Bernstein S, Lee P, van de Grift TC, Nordenskjöld A, Rapp M, Simmonds M, Streuli JC, Thyen U, Wiesemann C, Group d-L. Early Genital Surgery in Disorders/Differences of Sex Development: Patients' Perspectives. Arch Sex Behav. 2021;50(3):913-923.
  473. Sircili MH, de Mendonca BB, Denes FT, Madureira G, Bachega TA, e Silva FA. Anatomical and functional outcomes of feminizing genitoplasty for ambiguous genitalia in patients with virilizing congenital adrenal hyperplasia. Clinics. 2006;61(3):209-214.
  474. Dénes FT, Cocuzza MA, Schneider-Monteiro ED, Silva FA, Costa EM, Mendonca BB, Arap S. The laparoscopic management of intersex patients: the preferred approach. BJU Int. 2005;95(6):863-867.
  475. Bernabé KJ, Nokoff NJ, Galan D, Felsen D, Aston CE, Austin P, Baskin L, Chan YM, Cheng EY, Diamond DA, Ellens R, Fried A, Greenfield S, Kolon T, Kropp B, Lakshmanan Y, Meyer S, Meyer T, Delozier AM, Mullins LL, Palmer B, Paradis A, Reddy P, Reyes KJS, Schulte M, Swartz JM, Yerkes E, Wolfe-Christensen C, Wisniewski AB, Poppas DP. Preliminary report: Surgical outcomes following genitoplasty in children with moderate to severe genital atypia. J Pediatr Urol. 2018;14(2):157.e151-157.e158.
  476. Jesus LE. Feminizing genitoplasties: Where are we now? J Pediatr Urol. 2018;14(5):407-415.
  477. FORTUNOFF S, LATTIMER JK, EDSON M. VAGINOPLASTY TECHNIQUE FOR FEMALE PSEUDOHERMAPHRODITES. Surg Gynecol Obstet. 1964;118:545-548.
  478. Sircili MH, Bachega TS, Madureira G, Gomes L, Mendonca BB, Dénes FT. Surgical Treatment after Failed Primary Correction of Urogenital Sinus in Female Patients with Virilizing Congenital Adrenal Hyperplasia: Are Good Results Possible? Front Pediatr. 2016;4:118.
  479. Baskin LS, Erol A, Li YW, Liu WH, Kurzrock E, Cunha GR. Anatomical studies of the human clitoris. J Urol. 1999;162(3 Pt 2):1015-1020.
  480. Kogan SJ, Smey P, Levitt SB. Subtunical total reduction clitoroplasty: a safe modification of existing techniques. J Urol. 1983;130(4):746-748.
  481. Rink RC, Cain MP. Urogenital mobilization for urogenital sinus repair. BJU Int. 2008;102(9):1182-1197.
  482. Sircili MH, de Mendonca BB, Denes FT, Madureira G, Bachega TA, e Silva FA. Anatomical and functional outcomes of feminizing genitoplasty for ambiguous genitalia in patients with virilizing congenital adrenal hyperplasia. Clinics (Sao Paulo). 2006;61(3):209-214.
  483. Kolon TF, Herndon CD, Baker LA, Baskin LS, Baxter CG, Cheng EY, Diaz M, Lee PA, Seashore CJ, Tasian GE, Barthold JS, Assocation AU. Evaluation and treatment of cryptorchidism: AUA guideline. J Urol. 2014;192(2):337-345.
  484. Sircili MH, Denes FT, Costa EM, Machado MG, Inacio M, Silva RB, Srougi M, Mendonca BB, Domenice S. Long-term followup of a large cohort of patients with ovotesticular disorder of sex development. J Urol. 2014;191(5 Suppl):1532-1536.
  485. Pippi Salle JL, Sayed S, Salle A, Bagli D, Farhat W, Koyle M, Lorenzo AJ. Proximal hypospadias: A persistent challenge. Single institution outcome analysis of three surgical techniques over a 10-year period. J Pediatr Urol. 2016;12(1):28.e21-27.
  486. Snodgrass WT, Granberg C, Bush NC. Urethral strictures following urethral plate and proximal urethral elevation during proximal TIP hypospadias repair. J Pediatr Urol. 2013;9(6 Pt B):990-994.
  487. Romao RLP, Pippi Salle JL. Update on the surgical approach for reconstruction of the male genitalia. Semin Perinatol. 2017;41(4):218-226.
  488. Steven L, Cherian A, Yankovic F, Mathur A, Kulkarni M, Cuckow P. Current practice in paediatric hypospadias surgery; a specialist survey. J Pediatr Urol. 2013;9(6 Pt B):1126-1130.
  489. Hafez AT, Helmy T. Tubularized incised plate repair for penoscrotal hypospadias: role of surgeon's experience. Urology. 2012;79(2):425-427.
  490. Cools M, Looijenga LH, Wolffenbuttel KP, T'Sjoen G. Managing the risk of germ cell tumourigenesis in disorders of sex development patients. Endocr Dev. 2014;27:185-196.
  491. Lee P, Schober J, Nordenström A, Hoebeke P, Houk C, Looijenga L, Manzoni G, Reiner W, Woodhouse C. Review of recent outcome data of disorders of sex development (DSD): emphasis on surgical and sexual outcomes. J Pediatr Urol. 2012;8(6):611-615.
  492. Ark JT, Moses KA. Operative considerations for late-presenting persistent Müllerian duct syndrome. Urol Ann. 2016;8(3):363-365.
  493. Andersson M, Sjöström S, Wängqvist M, Örtqvist L, Nordenskjöld A, Holmdahl G. Psychosocial and Sexual Outcomes in Adolescents following Surgery for Proximal Hypospadias in Childhood. J Urol.2018;200(6):1362-1370.
  494. Rynja SP, de Jong TP, Bosch JL, de Kort LM. Functional, cosmetic and psychosexual results in adult men who underwent hypospadias correction in childhood. J Pediatr Urol. 2011;7(5):504-515.
  495. van der Zwan YG, Callens N, van Kuppenveld J, Kwak K, Drop SL, Kortmann B, Dessens AB, Wolffenbuttel KP, DSD DSGo. Long-term outcomes in males with disorders of sex development. J Urol.2013;190(3):1038-1042.
  496. Köhler B, Kleinemeier E, Lux A, Hiort O, Grüters A, Thyen U, Group DNW. Satisfaction with genital surgery and sexual life of adults with XY disorders of sex development: results from the German clinical evaluation study. J Clin Endocrinol Metab. 2012;97(2):577-588.
  497. Meyer-Bahlburg HF, Migeon CJ, Berkovitz GD, Gearhart JP, Dolezal C, Wisniewski AB. Attitudes of adult 46, XY intersex persons to clinical management policies. J Urol. 2004;171(4):1615-1619; discussion 1619.
  498. Mureau MA, Slijper FM, van der Meulen JC, Verhulst FC, Slob AK. Psychosexual adjustment of men who underwent hypospadias repair: a norm-related study. J Urol. 1995;154(4):1351-1355.
  499. Bubanj TB, Perovic SV, Milicevic RM, Jovcic SB, Marjanovic ZO, Djordjevic MM. Sexual behavior and sexual function of adults after hypospadias surgery: a comparative study. J Urol. 2004;171(5):1876-1879.

 

Pituitary and Hypothalamic Tumor Syndromes in Childhood

ABSTRACT

 

Central nervous system (CNS) tumors are the second commonest childhood malignancy, with 10% of these affecting the suprasellar and/or intrasellar regions. Survival has increased significantly over the last decade as a result of improved multimodality cancer therapies and better supportive care. Measurements of serum prolactin, α-fetoprotein, and β-hCG as well as baseline pituitary function tests are essential at diagnosis prior to commencement of any therapy. Craniopharyngiomas and low-grade gliomas account for most of these tumors, whilst other histological subtypes such as pituitary adenomas, germinomas, and hamartomas are rare. Non-neoplastic masses include pituitary hyperplasia and Rathke’s cleft cysts. Neurological syndromes and endocrine dysfunction are often present at diagnosis, and may be missed if not sought for. Post-diagnosis, endocrinopathies can evolve over decades secondary to tumor and/or treatment, necessitating long-term follow-up of such patients. Treatment of endocrine dysfunction is crucial not just to avoid the fatal consequences of untreated secondary adrenal insufficiency and/or diabetes insipidus, but also to improve quality of survival, and should be closely supervised by a pediatric endocrinologist with experience in the management of such patients. Growth hormone therapy in replacement doses in particular has not been shown to increase the risk of tumor recurrence. The “hypothalamic syndrome”, including variable hypothalamic dysfunction (e.g., sleep-wake cycle disturbances, temperature dysregulation, adipsia, and behavioral disorders) and hypothalamic obesity, is a common and as yet untreatable sequela of both tumor and treatment. The latter is caused by dysregulation of a network anorexigenic and orexigenic hormone signals which is only beginning to be elucidated.

 

INTRODUCTION

 

Central nervous system (CNS) tumors are the second commonest childhood malignancy after leukemias, accounting for 25% of cancers in children <15 years of age with an annual incidence rate of 35 cases/million/year (1–4). As with all childhood cancers, their incidence is gradually increasing worldwide (1,2,5), an effect largely attributed to improvements in diagnosis and tumor registration (6–8), and more recently campaigns such as the UK HeadSmart project aimed at increasing awareness of pediatric brain tumor symptoms (http://www.headsmart.org.uk/) (9). Concurrently, 5-year survival for CNS tumors has increased much more steeply from 57% to 65% in the last decade (~95% in low-grade gliomas) as a result of improved multimodality cancer therapies and better supportive care (10–12).

 

However, while survival is high, increasingly intensive treatment strategies aimed at improving cure in a small minority can conversely cause a higher toxicity burden in the larger majority, with a rapidly accruing cohort of survivors faced with reduced quality of life due to late and evolving multi-organ toxicities (13–15). Over 40% of these chronic morbidities (“late effects”) are severe, disabling or life-threatening (16), and more than 80% of CNS tumor survivors develop at least one endocrinopathy, most frequently growth hormone deficiency  (17). Indeed, suprasellar tumors have been found to be the commonest cause of hypothalamo-pituitary dysfunction in adult cohort studies (18,19). However, when compared with adult CNS tumors, pediatric tumors tend to be more curable, and the early presentation of some tumors (e.g., craniopharyngiomas, primitive neuroectodermal tumors (PNET)), and their association with mutations in neural development genes blur the delineation between congenital malformations and neoplasia (20–22).

 

Tumor location and histology is distinctly age-dependent: 30% of tumors under the age of 14 years are infratentorial (medulloblastomas, posterior fossa juvenile pilocytic astrocytomas, and ependymomas), whilst 26% and 16% of tumors diagnosed in young adulthood (15 to 24 years) are supratentorial or suprasellar respectively (non-pilocytic astrocytomas, other gliomas, pituitary adenomas, and germinomas) (4,23). Supra- and intrasellar tumors constitute 10% of all pediatric CNS tumors (23,24) and their close proximity to the vital hypothalamo-pituitary axis (HPA) increases the risk of important endocrine dysfunction. This may occur secondary to tumor mass effect and/or treatment, and can therefore be manifest at presentation or evolve subsequently during or after completion of oncological therapies. Dissecting the effect of tumor from treatment on endocrinopathies diagnosed after commencement of therapy is particularly complicated. We aim here to (1) outline the epidemiology, clinical features, and management of common pediatric suprasellar tumors not readily addressed in other chapters, (2) examine the common clinical neuroendocrine presenting features and (3) summarize common themes in the neuroendocrine late effects observed at follow-up of these patients.

 

THE DIFFERENTIAL DIAGNOSIS OF PEDIATRIC SUPRA- AND INTRASELLAR MASSES

 

The definitive diagnosis of pediatric suprasellar and intrasellar masses is crucial, as therapeutic strategies differ markedly depending on histological subtype. However, a tissue diagnosis may not always be possible due to their location, as even minor procedures such as biopsies can lead to life-threatening endocrinopathies such as diabetes insipidus (DI) (25). Biochemical measurements of serum prolactin (PRL), α-fetoprotein (AFP), and β-human chorionic gonadotrophin (β-hCG) to aid the diagnosis of prolactinomas and secreting germinomas respectively are therefore absolutely essential prior to commencement of any therapy.

 

Table 1. The Differential Diagnosis of Pediatric Suprasellar Tumors and Other Disorders

Neoplastic

Craniopharyngioma

Low-grade glioma (mainly pilocytic astrocytoma)

Pituitary adenoma

Germ cell tumor (mainly germinoma)

Hamartoma

Meningeal metastases

Non-neoplastic

Pituitary hyperplasia

Pituitary stalk thickening

Langerhans cell histiocytosis*

Tuberculosis

Sarcoidosis

Rathke cleft cyst

Arachnoid cyst

Epidermoid/dermoid cyst

Meningioma

*The classification of Langerhans cell histiocytosis as a non-neoplastic disease is debatable.

 

Craniopharyngiomas

 

Figure 1. T1-weighted MRI images of a craniopharyngioma demonstrating the coexistence of solid, cystic and calcified components with the tendency for multiple progressions over seven years. (a) After initial endoscopic cyst fenestration and ventriculoperitoneal shunt insertion, (b) after first transcranial debulking, (c) first cystic progression, (d) after first cyst drainage via reservoir, (e) second cystic progression, (f) after second transcranial debulking, (g) after adjuvant radiotherapy and third cystic progression, (h) after second cyst drainage via reservoir, (a) after fourth cystic & solid progression, (j) after complete resection.

 

Craniopharyngiomas are by far the commonest suprasellar tumor of childhood, accounting for up to 50-80% of masses in this region (24,26–28) and 1.5-11.6% of all pediatric CNS tumors (3,24,26,29,30).  There is a bimodal age distribution in incidence, with the peak incidence in childhood occurring between the ages of 5-14 years at 1.4 cases/million/year (29,31). They are benign tumors originating from the embryonal epithelium lining Rathke’s pouch and are almost invariably adamantinomatous in childhood, characterized by the presence of intratumoral calcifications(32). Over-activation of the Sonic hedgehog (SHH) and Wnt/β-catenin pathways, both important in both pituitary stem cell development and carcinogenesis, have been shown to be key to their formation (20,21), but they occur typically sporadically, with only one case report of familial adamantinomatous craniopharyngiomas occurring in a consanguineous pedigree reported in the English literature (33). Contrastingly, papillary craniopharyngiomas are found almost exclusively in adults and harbor the BRAF V600E mutation instead (34).

 

Symptoms related to hypothalamo-pituitary dysfunction, such as weight gain, growth failure, prolonged recovery from infections, and abnormalities of puberty are often under-recognized but in fact constitute the third commonest group of clinical findings at diagnosis, after symptoms related to raised intracranial pressure (e.g., headaches, vomiting) and visual deterioration (22,35–47). Radiologically, 65-93% of these tumors are calcified but a plain X-ray or computerized tomography (CT) scan may be required to demonstrate this. The coexistence of solid, cystic, and calcified structures on neuroimaging, as well as the characteristic cholesterol crystals seen under microscopy of the “engine fluid” aspirated surgically from cystic components are so highly suggestive of the diagnosis that histological confirmation from biopsies of solid components may be unnecessary, particularly as this may further compromise hypothalamo-pituitary function (32,48). Anatomically, 75% of craniopharyngiomas are suprasellar with an intrasellar extension, 20% are exclusively suprasellar, and 5% are exclusively intrasellar, with over 50% involving the hypothalamus and nearly one-third invading the floor of the third ventricle (26,37,44).

 

Due to their location, a significant proportion of these tumors are not completely resectable, but their relative rarity, high rates of survival, and benign histology have precluded them from pan-European randomized trials, resulting in a lack of agreement on the optimal treatment strategy. Most recently, the first evidence- and consensus-based national UK guideline for the management of craniopharyngiomas in children and young people has been published by the UK Children’s Cancer and Leukemia Group (CCLG), with endorsement from the Royal College of Pediatrics and Child Health (RCPCH) and British Society of Pediatric Endocrinology & Diabetes (BSPED) (49).Importantly, these guidelines advocate a more conservative approach to the degree of surgical resection in the presence of significant hypothalamic involvement in order to minimize further damage to the hypothalamo-pituitary axis (39,50,51), balanced against the need to relieve symptoms of raised intracranial pressure, preserve vision, and provide long-term control and reduced recurrence rates (49,52,53). The use of adjuvant radiotherapy in combination with subtotal tumor resection has been shown to achieve survival rates which are on par with complete tumor resection (5-year progression-free survival 73-100% vs 73-82%), with the potential for less neuroendocrine dysfunction (54–56). More recently, the use of proton beam therapy has increased, with equivalent survival outcomes to conventional radiotherapy, but there remains the issue of insufficient follow-up data to ascertain its long-term toxicity profile (57,58). Experience with systemic or intracystic chemotherapy, intracystic interferon, and radioisotope instillation of 32P or 90Y have been met with conflicting success and cannot therefore be currently recommended as primary treatment approaches in children (59–62). Ultimately, despite high long-term overall survival (80% at 30 years), (37) up to 98% of survivors experience dysfunction in at least one hypothalamo-pituitary axis with high rates of morbid obesity(45,63).

 

Low-grade Gliomas (LGGs)

Figure 2. T1-weighted MRI image demonstrating appearances of a large, lobulated optic pathway astrocytoma with hydrocephalus and widespread leptomeningeal dissemination affecting the brainstem, cerebellum, and spinal cord.

 

LGGs account for >40% of all CNS tumors and are thus the commonest pediatric intracranial tumor (3,8). The optic pathway, hypothalamus, and suprasellar midline are the second most frequent location for LGGs (30-50%) after the cerebellum, cerebral hemispheres, and brainstem (12,64). Even in the suprasellar region they are the second commonest pediatric tumor after craniopharyngiomas, and are similarly regarded as benign (grade I or II), the vast majority being juvenile pilocytic astrocytomas (65). The genetic tumor predisposition syndrome neurofibromatosis type 1 (NF-1) is present in 10-16% of cases, whilst 15% of asymptomatic NF-1 children will have LGGs on neuroimaging. NF-1-associated tumors more often originate from the optic nerves (70%) than from the hypothalamochiasmatic area (27-40%) and tend to a more indolent course (11,12,64,66–69). Mutations involving KIAA1549, BRAF and Ras proto-oncogenes are associated with pilocytic astrocytomas and disruptors targeted at these pathways form the basis of current clinical therapeutic trials (70–72). Similar to craniopharyngiomas, the commonest symptoms at diagnosis are related to visual changes or raised intracranial pressure, with disorders of the LH/ FSH axis being the most prevalent endocrinopathy at presentation (25,66,73–75). In infancy, hypothalamic LGGs can present with diencephalic syndrome (see below) (11,76–78), which significantly increases the risk of future neuroendocrine dysfunction (79).

 

Complete tumor resection has been shown to be a favorable risk factor for survival (12,64) but suprasellar and/or optic pathway tumors cannot be completely resected without causing major visual and neuroendocrine morbidity. Treatment trials have thus focused on medical strategies, with radiotherapy being delayed in favor of chemotherapy in young children due to concerns of cognitive dysfunction (80), subsequent primary cancers (SPCs) (81,82) and radiation-induced vasculopathies (83), despite showing superior 5-year progression-free survival rates (65% vs. 47%) (11). However, to date none of the previous international treatment trials – LGG1 (1997-2004) or LGG2 (2005-2010) – were randomized, these being purely observational studies aimed at improving visual outcomes but with little reported success (11,12,84). At the time of writing, the first randomized interventional study of chemotherapeutic strategies (LGG3) is being designed with careful long-term prospective measurements of visual and neuroendocrine outcomes. More recently, tumors harboring BRAF mutations have been the target of MAPK/ERK kinase (MEK) and BRAF inhibitors such as trametinib and dabrafenib (72,85–87), although these can still lead to various side effects including endocrinopathies(88).

 

A 30-year survival analysis has revealed the extent of long-term neuroendocrine dysfunction affecting these patients with new endocrine deficits appearing up to 15 years post-diagnosis, and 20-year endocrine event-free survival approaching 20% (25). Hypothalamic tumor location is a more important independent risk factor for long-term anterior hypothalamo-pituitary deficits than radiotherapy exposure; however only surgical intervention (regardless of extent) has been shown to be independently associated with posterior pituitary dysfunction and life-threatening salt and water imbalances (25,64). Similar to craniopharyngiomas, overall survival is high (85% at 25 years), but ~80% of survivors experience at least one endocrinopathy (25,79).

 

Pituitary Adenomas

Figure 3. T1-weighted MRI image demonstrating appearances of a giant prolactinoma. There is obscuration of normal pituitary morphology due to the tumor arising from the pituitary gland itself.

 

Pituitary adenomas are rare in childhood, accounting for just 3% of all supratentorial tumors with an estimated annual incidence of 0.1 cases/million/year in children (89). The vast majority are functioning, with prolactinomas alone accounting for 50% of adenomas and 2% of all pediatric and adolescent intracranial tumors. Therefore, the measurement of plasma prolactin (PRL) may be diagnostic and is absolutely mandatory prior to planning surgery for any pituitary mass, as medical treatment alone may be entirely curative (90,91). ACTH- and GH-secreting adenomas are the next commonest, whilst TSH-secreting, gonadotrophin-secreting, and non-functioning adenomas are vanishingly rare (91–93).

 

A child with a pituitary adenoma may be the index case for a genetic tumor predisposition syndrome (up to 22%), particularly given their rarity, and therefore careful documentation of their family history and testing for multiple endocrine neoplasia type 1 (MEN1) and aryl-hydrocarbon receptor interacting protein (AIP) gene mutations are therefore paramount in all cases (94–96). Other genetic syndromes associated with pituitary adenomas that need to be considered are multiple endocrine neoplasia type 4 (CDKN1B), Carney complex (PRKAR1A), McCune-Albright syndrome (GNAS), SDH-related pituitary adenoma syndrome (SDHB, SDHC, SDHD), and DICER1 syndrome (97).

 

Investigation and management of pituitary adenomas depends on whether they are functioning or non-functioning, and in the case of the former, which hormones are being secreted in excess. Similar to craniopharyngiomas, an evidence- and consensus-based national UK guideline is being written for the management of pituitary adenomas in children and young people as a collaborative effort between the CCLG, RCPCH and BSPED.

 

PROLACTINOMA

 

Pituitary adenomas are classified as microadenomas (<1 cm), macroadenomas (>1 cm), and giant adenomas (>4 cm). In prolactinomas plasma PRL levels generally, but not exclusively, increase with tumor size. Hyperprolactinemia may also result from stalk compression by tumor mass (interrupting hypothalamic dopaminergic inhibition of PRL secretion) and antipsychotic medication but PRL concentrations are usually <2000 mU/l and patients rarely symptomatic (98). Laboratories should always screen for artefactual hyperprolactinemia due to macroprolactin, but levels >5000 mU/l are usually diagnostic and symptomatic. Occasionally, falsely low results can be due to interference by extreme hyperprolactinemia on antibody-antigen sandwich complex formation, a phenomenon known as the hook effect. In cases of large tumors, samples should therefore be diluted 100-fold and repeated for confirmation (99). Clinical presentation varies according to the size of tumor, gender, and pubertal status, with girls usually experiencing galactorrhea, pubertal delay, or amenorrhea and boys presenting later with larger, more aggressive tumors with raised intracranial pressure (90).

 

Given the paucity of good quality outcome data in children, treatment guidelines follow those for adults (53,91), recommending dopamine agonists (DAs) as first line, ideally cabergoline due to its high efficacy and tolerability (98). Starting doses, dose escalation and duration of therapy in children remain undefined and are critical questions given the potential for more aggressive disease and cardiac valve abnormalities with long-term cumulative exposure (100). Surgery should be reserved for those cases resistant to DAs or for neurosurgical emergencies (e.g., neuro-ophthalmic deficits, pituitary apoplexy) and both trans-sphenoidal and transcranial approaches should be considered by an experienced pediatric neurosurgeon. Radiotherapy has usually been reserved for treatment failures in view of the presumed risk of post-treatment endocrine morbidity and second primary cancers. However, the former may have been overestimated in view of the high incidence of endocrinopathies already present at diagnosis (101), and therefore this treatment modality should be considered earlier and prior to other more experimental treatments such as temozolomide chemotherapy (98). As with other hypothalamo-pituitary tumors, long-term neuroendocrine and secondary cardiovascular morbidity is significant (102).

 

CORTICOTROPHINOMAS

 

The age distribution for corticotrophinomas is younger than that of prolactinomas (where the incidence increases in adolescence and young adulthood), with Cushing disease accounting for the vast proportion of Cushing syndrome in children aged >5 years, and >70% of pituitary adenomas in the prepubertal age group (103,104). These tumors are nearly always microadenomas. Common presenting features include weight gain with linear growth arrest or short stature, change in facial appearance, fatigue, striae, hirsutism, emotional lability, hypertension, acne, headaches, or psychosis (104–106). Diagnosis is achieved by firstly screening for Cushing syndrome indicated by a raised urine free cortisol (sensitivity 89%) or midnight cortisol concentration of >50 nmol/l (sensitivity 99%, specificity 20%). This is then followed by a low-dose (sensitivity 100%, specificity 80%) then high-dose (sensitivity 94%, specificity 70%) dexamethasone suppression test (104,107–111). CRH-stimulated bilateral inferior petrosal sinus sampling (BIPSS) may help successfully localize the position of the microadenoma (104,105). Transsphenoidal resection is the first-line treatment of choice, superseding bilateral adrenalectomy which carries a risk of post-operative Nelson syndrome(112). Cure rates are 45-78% with nearly 40% requiring adjuvant radiotherapy (113–115).

 

SOMATOTROPHINOMAS

 

8-15% of all pituitary adenomas in patients <20 years of age secrete GH, with a significant proportion co-secreting PRL and TSH (103,116). Genetic syndromes associated with somatotrophinomas include MEN-1 (MEN1), Carney complex (PRKAR1A), McCune-Albright syndrome (GNAS), and familial isolated pituitary adenoma (FIPA, AIP) syndrome (97). Due to the absence of complete epiphyseal fusion, in childhood and adolescence, somatotrophinomas present with pituitary gigantism rather than acromegaly. Tall stature and increased growth velocity however can still be associated with other acromegalic features such as mild obesity, macrocephaly, acral enlargement, frontal bossing, and macrognathia (93,117). Investigations reveal high random GH and IGF-1 concentrations, loss of GH pulsatility, and failure of GH suppression to an oral glucose tolerance test (87). Like corticotrophinomas, transsphendoidal resection is the treatment of choice but a significant proportion of patients require adjuvant medical therapy with somatostatin analogues (octreotide, lanreotide), dopamine agonists (cabergoline, bromocriptine), or the GH receptor antagonist pegvisomant (118). Radiotherapy has been used with limited effect (119).

 

Germ Cell Tumors

Figure 4. T1-weighted MRI image demonstrating the appearance of a contrast-enhancing suprasellar β-hCG-secreting germinoma in a patient who presented with central diabetes insipidus.

 

Germ cell tumors (GCTs) are tumors arising from primordial germ cells normally sited in the testes and ovaries and can be subclassified into germinomatous (GGCT, usually non-secreting but can occasionally produce βhCG) and non-germinomatous germ cell tumors (NGGCT). NGGCTs and can be further classified into yolk sac tumors (secreting α-fetoprotein (AFP)), choriocarcinomas (secreting βhCG), and embryonal carcinomas. In contrast to craniopharyngiomas and LGGs, intracranial GCTs account for just 3-4% of all primary pediatric and young adult CNS tumors <24 years (23,120). There is a clear peak in incidence in adolescence and young adulthood, with age-adjusted incidence rates rising from 0.9 cases/million/year in patients <10 years to 1.3-2.1 cases/million/year in patients aged 15-24 years (23,120). Boys are affected nearly three times as often as girls, and this sex distribution is magnified in adolescence (male: female ratio of >8:1) (23). GCTs are also the commonest CNS tumor in Klinefelter and Down syndromes (121). Diabetes Insipidus (DI) and gonadotrophin-independent precocious puberty (due to βhCG acting on the LH receptor) are common findings at diagnosis and present in 30-50% and 11-12% of patients respectively. Unlike NGGCTs, GGCTs can grow indolently (if at all), meaning that both clinical and radiological features can often be subtle at onset, and delays in diagnosis up to 21 years have been reported (122–124).

 

Histologically, intracranial GCTs resemble their gonadal counterparts (ovarian teratoma or testicular seminoma) and account for 34% of all such tumors (125). They have a particular predilection for the pineal gland (37-66%) and suprasellar region (23-35%), such that synchronous (bifocal) pineal and suprasellar tumors are pathognomonic. Both GGCTs and NGGCTs are extremely chemo- and radiosensitive, and their propensity to metastasize throughout the cerebrospinal fluid (26,121,126) has meant that whole neuraxial (craniospinal) irradiation has been standard therapy for decades, with overall and progression-free survival rates approaching 100% (119). Chemotherapy alone has been shown to result in inferior survival (127), and more recent attempts to reduce the irradiation field with adjuvant chemotherapy in an effort to preserve cognitive function have shown little reduction in overall survival (121,128,129). The latest SIOP CNS GCTII however aims to reduce the radiation dose and field by stratifying treatment strategies between NGGCT and GGCTs, and based on the absence or presence of metastatic disease (https://www.skion.nl/workspace/uploads/2_siop_cns_gct_ii_final_version_2_15062011_unterschrift_hoppenheit.pdf). As for other suprasellar tumors, the rate of post-treatment endocrine morbidity is significant, with 50-60% of patients having at least one endocrinopathy (122).

 

Hypothalamic Hamartomas

Figure 5. T1-weighted MRI image demonstrating the appearances of a pedunculated hypothalamic hamartoma (arrowheads) arising from the floor of the third ventricle in a patient who presented with central precocious puberty. The pituitary morphology is otherwise normal.

 

Hypothalamic hamartomas are extremely rare congenital (rather than neoplastic) malformations consisting of grey matter heterotopia in the tuber cinereum and inferior hypothalamus (24,26,130). Their true prevalence is unknown but is estimated to occur in between 1 in 50,000 – 1 million individuals (131–133). Symptom onset occurs in infancy to early childhood, with the mean age of first seizures occurring between 6 weeks – 4.5 years (133–136). The triad of epilepsy (usually gelastic (laughing) or dacrystic (crying) seizures), central precocious puberty, and developmental delay is classic with the seizure semiology eventually evolving into multiple, more severe seizure types (130). Rarely, they are associated with Pallister-Hall syndrome, an autosomal dominant disorder characterized by polydactyly and other midline defects (imperforate anus, bifid epiglottis, panhypopituitarism and dysmorphic facies) (132,137), or with SOX2 mutations (138).

 

The intrinsic epileptogenicity of these lesions (139,140), the trend towards evolving seizure semiology, the worsening of behavioral and psychiatric co-morbidities, and the general failure of anti-epileptic drug therapy has led clinicians to explore the options of surgical or stereotactic radiosurgical resection despite their deep-seated location, with variably reported success in the remission of seizure activity and behavioral disturbances, but more modest improvements in cognitive function (130,131,141–143). Li et al.'s (144) case series reported successful remission of central precocious puberty (CPP) and little, if any, late-onset endocrinopathy; but a larger cohort study by Freeman et al. (145) suggested that clinically silent endocrine dysfunction (particularly GH and TSH deficiency) is common both at diagnosis and postoperatively. Transient posterior pituitary dysfunction leading to DI and the syndrome of inappropriate antidiuretic hormone secretion (SIADH) has also been described (145,146). One adult cohort study corroborates these findings, showing that >1/3 of these patients had endocrine dysfunction and approximately 2/3 experienced excessive weight gain postoperatively (147). More recently laser induced thermal therapy (LiTT) of these lesions has shown promising results with regards to seizure control, with little late onset additional endocrinopathies (148,149).

Langerhans Cell Histiocytosis (LCH)

 

Figure 6. T1-weighted MRI image demonstrating the appearances of a contrast-enhancing suprasellar LCH lesion. There is a small anterior pituitary and absent posterior pituitary bright spot in keeping with the known panhypopituitarism (including central DI) present at diagnosis.

 

LCH (previously “histiocytosis X”) is one of the three major histiocyte disorders, and involves clonal proliferation of bone marrow-derived dendritic antigen-presenting (“Langerhans”) cells which accumulate in various organs (150). It is a rare disease with an incidence of 2.6-8.9 cases/million/year, the majority presenting in infancy (median age at diagnosis 2-3.8 years, incidence at age <1 year 9.0-15.3 cases/million/year vs. age >5 years 0.7-4.5 cases/million/year) with no sex predilection (151–154). The variability in organ involvement causes a spectrum of clinical features ranging from a single self-healing cutaneous lesion to fatal multiorgan disease, particularly if the liver, spleen, lungs, and hemopoietic system (the “risk” organs) are involved (150). Multisystem involvement is present in 27-56% of cases, of which 28-80% have “risk” organ involvement (151–153,155,156). LCH can thus be considered a primary hematological disorder which, in a proportion of cases, infiltrates the CNS, although its etiology, whether neoplastic or reactive, remain poorly understood (155). More than half of biopsied lesions contain BRAF mutations(157).

 

In the CNS, the hypothalamo-pituitary region is involved in up to 25% of cases, which almost invariably leads to DI (previously known as Hand-Schuller-Christian disease if associated with orbital and bony lesions)(151,152,154,158,159). Commonly associated radiological findings include thickening of the pituitary stalk with progression to space-occupying tumors and an absence of the posterior pituitary bright spot (159). Indeed, LCH is the commonest underlying diagnosis in patients with central DI and an intracranial mass, occurring in 70% of this cohort(160). The presence of multisystem involvement, particularly if involving “risk” organs, craniofacial bones, gastrointestinal tract, skin, or genitalia) is a particular risk factor for DI (159,161).

 

Treatment is dependent on the number of organs involved and may range from biopsy/curettage, intralesional steroids, indomethacin, and radiotherapy/UV phototherapy for single bone and cutaneous lesions to systemic chemotherapy with steroids and vinblastine for multisystem disease (155,162,163). Refractory cases have been treated with cytarabine, cladribine, clofarabine, hemopoietic stem cell transplantation, or BRAF inhibitors (164–168). Notably, no treatment protocol has been shown to reverse existing or prevent future DI or other endocrinopathies(159), though current therapeutic recommendations are aimed at preventing disease progression and limiting endocrinopathy with prolonged, low-dose systemic chemotherapy (155,169–171). Overall, 5-year survival remains relatively high at 71-95%, but 3-25% of patients experience at least one endocrinopathy (particularly GH deficiency), with no current chemotherapeutic regimens showing superior overall- or endocrine event-free survival (151,156,158,161).

 

Pituitary Stalk Thickening

Figure 7. T1-weighted MRI image illustrating the appearances of a contrast-enhancing thickened pituitary stalk lesion (arrow) and an absent posterior pituitary bright spot in a patient presenting with central DI. The differential diagnosis included germinoma and LCH. However, approximately one year after diagnosis, the pituitary stalk lesion resolved completely, although the patient has been left with GH deficiency and central DI.

 

A thickened pituitary stalk (TPS) may be discovered either as part of the evaluation of a patient presenting with central DI, visual impairment, or other endocrine dysfunction or incidentally on neuroimaging performed for other purposes. It is discussed here as it is an important differential for germ cell tumors and Langerhans cell histiocytosis (LCH), resulting frequently in diagnostic and management dilemmas, due to a number of reasons:

 

  1. There is no clear consensus as to what constitutes abnormality for children; previous adult studies have shown that the 95th centile for the transverse dimensions of the infundibulum at the optic chiasm and pituitary insertion are 4.21-4.35 mm and 2.69-2.89 mm respectively (upper limit 4.21-4.58 mm and 2.93-3.04 mm) (172,173). Raybaud and Barkovich suggest using a pediatric threshold thickness of 3.8 mm at the optic chiasm and 2.7 mm at the pituitary insertion for investigating further pathology, particularly if there are interruptions in the normal smooth tapering of the infundibulum from median eminence to pituitary insertion (174).
  2. The radiological appearances of a TPS, LCH and germinomas cannot be easily differentiated and there is substantial overlap (Table 2). The normal infundibulum lacks a blood-brain barrier and therefore always enhances with contrast, obscuring neoplastic processes. TPS is the commonest initial radiological finding in both LCH and germinomas, and concurrent absence of the posterior pituitary bright spot is inconsistent (123,175,176). Similarly, the two commonest causes of TPS in the pediatric age group are LCH and germinomas, accounting for 7-75% and 9-71% of TPS cases respectively (176–179). Other common causes of TPS in adults such as lymphocytic hypophysitis and neurosarcoidosis are rare in children (176).
  3. Biopsies of the TPS to obtain a definitive histological diagnosis can be inconclusive and lead to further substantial endocrine morbidity, including panhypopituitarism with DI, and are thus generally avoided (178).
  4. The interval from the time of initial symptoms to diagnostic MRI can be prolonged, particularly for germinomas (up to 21 years), occasionally with initially normal neuroimaging (123,124,180,181). An initially normal MRI does not therefore preclude an occult germinoma or other pathological process in the presence of idiopathic central DI, leading some authors to recommend serial 3-6 monthly scans and follow-up, although the duration of serial scanning is unclear (174). Additionally, there have been cases of occult germinomas masquerading as radiologically or even histologically diagnosed lymphocytic hypophysitis in children (182,183).

 

In an attempt to define which patients with isolated TPS are at risk of neoplasia and therefore require more intensive follow-up or biopsy, Robison et al. suggest risk factors such as the presence of DI (strongest risk factor), the coexistence of DI with anterior pituitary dysfunction or a progressive increase in infundibular size of >15% from baseline (178). Apart from size, no other particular MRI appearances have been found to be specific for pediatric-related tumor processes (184). Various proposed diagnostic pathways have been proposed for the management of TPS and idiopathic DI (178,184,185) but most recently a national consensus-based guideline has been developed in the UK by the CCLG, RCPCH and BSPED to help achieve a more consistent approach to this finding (186).

 

Miscellaneous Non-Neoplastic Hypothalamo-Pituitary Masses

 

Other hypothalamo-pituitary malformations can mimic neoplastic processes in the suprasellar region, and should therefore be considered in the differential diagnosis particularly before commencing oncological therapies:

 

  • Pituitary hyperplasia – Hypothalamic releasing hormones are trophic on the pituitary gland, hence hypersecretion of these hormones (e.g., GHRH from a pancreatic tumor in children with MEN1 syndrome) can cause anterior pituitary enlargement and mimic a true mass. The commonest physiological cause of pituitary hyperplasia is puberty, where the maximal height of the gland can be 10 mm in girls and 7 mm in boys (187,188). Pituitary hyperplasia can also occur pathologically, for instance in chronic primary hypothyroidism leading to thyrotroph hyperplasia due to a lack of negative feedback (24,187). It is also important to note that pituitary enlargement can be associated with certain congenital forms of hypopituitarism (PROP1, LHX3, SOX3 mutations (189,190).
  • Rathke’s cleft cysts (RCCs) – RCCs are congenital cystic epithelial remnants of Rathke’s pouch which fail to involute during pituitary development, hence arising in the pars intermedia but often extending superiorly (24). Although often incidental and asymptomatic (occurring in 11% of autopsy cases (191)), cystic growth can lead to visual deficits and endocrinopathies, requiring surgical marsupialization (resection exacerbates endocrine dysfunction) (192). Unlike craniopharyngiomas (the other common cystic suprasellar lesion), RCCs do not calcify.
  • Arachnoid cysts – These are congenital collections of cerebrospinal fluid (CSF) arising from the splitting and/ or duplication of the arachnoid membranes. 16% are suprasellar and these cysts can present with symptoms of raised intracranial pressure, visual deterioration, endocrinopathies, or developmental delay (193–197). Treatment is by endoscopic fenestration (196,198,199).
  • Rare entities – In contrast to adults where autoimmune lymphocytic hypophysitis is the commonest cause of isolated thickened pituitary stalk (TPS), this is exceptionally rare in children, but should be considered in the differential together with other granulomatous diseases (neurosarcoidosis, tuberculosis (24,200).

 

Table 2. The Differential Diagnosis of Pediatric Suprasellar Masses by Radiological Features

Tumor

Primary location

T1 intensity§

T2 intensity§

Contrast enhancement

Other features

Craniopharyngioma

Supra>intrasellar

Variable, heterogenous

High

Yes (cystic rims)

Cysts, heterogenous, calcification

LGG

Suprasellar, optic pathways

Low

High

Yes

Generally homogenous

Pituitary adenoma

Intrasellar (intrapituitary)

Low

Low

No

Sella turcica expansion

Germinoma*

Suprasellar, pituitary stalk

Isointense – low

Isointense – low

Yes

Loss of posterior pituitary bright spot, coexistent pineal tumor

Hamartoma

Suprasellar (tuber cinereum)

Isointense

Isointense – high

No

-

LCH*

Suprasellar, pituitary stalk

Isointense

Isointense

Yes

Loss of posterior pituitary bright spot, coexistent osseous lesions

Lymphocytic hypophysitis*

Suprasellar, pituitary stalk, intrasellar

Isointense

Isointense

Yes

Loss of posterior pituitary bright spot

Pituitary hyperplasia

Intrasellar

Isointense

Isointense

Yes

Homogenous

RCC

Intrasellar

Isointense – high

Isointense – low

No

Round & smooth walled

Granuloma (sarcoidosis, TB)

Suprasellar, pituitary stalk

Isointense – low

Low – isointense

Yes

Coexistent parenchymal and leptomeningeal lesions

Arachnoid cyst

Suprasellar

Very low (isointense with CSF)

High (isointense with CSF)

No

-

LGG, low-grade glioma; LCH, Langerhans cell histiocytosis; RCC, Rathke’s cleft cysts. §MRI signal intensity in comparison to that of gray matter. *Note that germinomas, LCH and lymphocytic hypophysitis cannot be differentiated on radiological features alone (24,26,174,201).

 

NEUROENDOCRINE DYSFUNCTION AT DIAGNOSIS OF HYPOTHALAMO-PITUITARY TUMORS

 

Neurological Syndromes

 

RAISED INTRACRANIAL PRESSURE (RICP)

 

The proximity of hypothalamo-pituitary tumors to the floor of the third ventricle and optic chiasm accounts for the high frequency of RICP and visual symptoms at presentation. RICP symptoms (headache, vomiting, and/or papilloedema) are the commonest presenting feature of any pediatric brain tumor (30-60%) (202,203), but occur with even greater frequency in suprasellar lesions such as craniopharyngiomas (78%) and LGGs (86%) (37,66). Children may therefore present to acute neurosurgical units as a neurosurgical emergency or subacutely with a chronic course that may initially be misdiagnosed as tension/ migrainous headaches or infective gastroenteritis with unrecognized concurrent visual disturbances. Current UK recommendations are to scan all children with vomiting persisting <2 weeks, and/ or headaches occurring in children <4 years, on waking or during sleep, in association with confusion and/ or disorientation, or persisting >4 weeks (9). Persistent vomiting in the absence of other features suggestive of gastroenteritis (diarrhea, pyrexia) should also prompt consideration of an intracranial lesion. It is important to note that due to the delayed fusion of cranial sutures, children <4 years of age with hydrocephalus more often (41%) present with a rapidly increasing head circumference than classical RICP symptoms (203).

 

VISUAL DETERIORATION

 

Visual field loss and/or worsening visual acuity are the second commonest presenting feature, particularly in LGGs, where up to 100% of cases may have visual impairment due to direct involvement of the optic pathway (75). Other suprasellar tumors affect visual function by mass effect on the optic chiasm, occurring in up to 50-70% of craniopharyngiomas and 15% of pituitary adenomas (38,44,102). Contrastingly, visual symptoms are rare (~5-7%) in children with other CNS tumors (203). Other common ophthalmological symptoms that warrant urgent neuroimaging include new onset nystagmus, incomitant (paralytic) squints, optic atrophy, and proptosis, particularly given the difficulties in assessing visual function in young children and the danger of passing off a squint as being “normal” in childhood without detailed examination (9,203,204). Parinaud’s syndrome, a combination of upward gaze palsy, convergence-retraction nystagmus, and pupillary dilatation with light-near dissociation is a rare particular presentation of bifocal suprasellar/pineal germinomas due to pressure of the pineal tumor on the tectal plate (124,205). Although the aim of oncological therapy in many of these low-grade tumors is to preserve vision, this has not been generally successful, most likely due to nerve fiber dropout and optic atrophy (84), or the fact that anatomical tumor characteristics correlate poorly with the degree of visual loss at diagnosis  (206).

 

SEIZURES

 

Seizures are an uncommon presenting clinical feature of pediatric hypothalamo-pituitary tumors, occurring in <10% of craniopharyngiomas, LGGs, and germinomas (35,39,124,207,208), and are more often the result of reversible metabolic causes such as hypoglycemia (from cortisol and/or GH insufficiency), hypernatremia (from DI), or hyponatremia (from SIADH). Gelastic or dacrystic (laughing or crying, from the Greek gelos and dakryon respectively) seizures are notoriously difficult to diagnose but are characteristic of hypothalamic hamartomas (80-90%) due to the intrinsic epileptogenicity of these lesions that are essentially disorders of neuronal migration (134,139,147).

 

OTHER NEUROLOGICAL AND COGNITIVE SYMPTOMS

 

Hemiparesis and ataxia are less common but significant presenting features of intracranial tumors, as are cognitive impairment, delayed development, behavioral changes, and psychiatric symptoms, all of which mandate detailed neuro-ophthalmological examination in such cases, particularly in the presence of the neurocutaneous stigmata of tumor-predisposing syndromes such as neurofibromatosis and tuberous sclerosis. 

 

Endocrine Dysfunction

 

Although neuro-ophthalmological symptoms are the commonest presenting feature of hypothalamo-pituitary lesions, they are often preceded by symptoms associated with undiagnosed endocrinopathies in as many as two-thirds of patients (209). Endocrine dysfunction may be due to hormone excess (e.g., secreting pituitary adenomas, central precocious puberty) or hormone deficiency from pituitary invasion or compression by tumor mass, disrupting the various hypothalamo-pituitary endocrine pathways. The incidence of dysfunction in each of the hypothalamo-pituitary axes is partly dependent on the lesion (Table 3) though the reasons for the specificity of these presentations are largely unknown.

 

GH deficiency (GHD) and gonadotrophin dysfunction (either central precocious puberty (CPP) or gonadotrophin deficiency (GnD, i.e., pubertal delay/arrest)) are often the initial and commonest endocrinopathies at presentation of both craniopharyngiomas (GHD – up to 100%; GnD – up to 85%, CPP – up to 3%) and LGGs (CPP – up to 56%; GHD – up to 27%; GnD – up to 12%) (37,41,42,66,210). CPP is particularly prevalent in LGGs as it can occur in the context of NF-1 even in the absence of a hypothalamo-pituitary lesion (211). It is also one of key components of the hypothalamic hamartoma clinical triad, present in up to 45% of patients at diagnosis (131,145). In both these cases it is presumed to result from premature activation of hypothalamic GnRH, unlike its occurrence in up to 35% of germinomas, where gonadotrophin-independent CPP can occur due to secretion of β-hCG which shares a common alpha subunit with LH and FSH and thus stimulates the same receptors (124,126).

 

Other anterior pituitary deficits evolve only with extensive disease, and are usually only seen at presentation with craniopharyngiomas, although more subtle deficits may have previously been under-recognized with other tumors. ACTH deficiency (secondary hypoadrenalism) is particularly important to diagnose and treat pre-operatively, and is present at diagnosis in up to 71% of craniopharyngiomas, 19% of germinomas, 10% of hamartomas and 3% of LGGs (41,124,145,212). Similarly, TSH/TRH deficiency (secondary/central hypothyroidism) is present in up to 32% of craniopharyngiomas, 19% of germinomas and 10% of LGGs and hamartomas(45,124,145,213). Mild to moderate hyperprolactinemia (<2000 mU/l) is common in all non-prolactinoma hypothalamo-pituitary lesions, needs to be distinguished from true prolactinomas (>5000 mU/l), and does not usually lead to clinically significant galactorrhea.

 

Posterior pituitary dysfunction, particularly central (“cranial”) DI, is the hallmark endocrinopathy of germinomas and Langerhans cell histiocytosis (LCH), being present in up to 90% and 40% of patients respectively at diagnosis(123,158). However, DI can also occur as a presenting clinical feature for other suprasellar lesions which may be missed if symptoms of polyuria and polydipsia are not elucidated.

 

Table 3. Common Endocrinopathies at Presentation of Various Hypothalamo-Pituitary Lesions

Tumor

Commonest endocrinopathy at presentation

Craniopharyngioma

GH deficiency, pubertal delay/arrest

Optic pathway LGG

Central precocious puberty

Pituitary adenoma

Hyperprolactinemia (prolactinomas)

Suprasellar germinoma

Central diabetes insipidus, gonadotrophin-independent central precocious puberty (hCG-secreting)

Hypothalamic hamartoma

Central precocious puberty

Langerhans cell histiocytosis

Central diabetes insipidus

GH, growth hormone; LGG, low-grade glioma; hCG, human chorionic gonadotrophin.

 

Endocrine dysfunction is under-recognized at presentation, as demonstrated by the discrepancies between spontaneous reports of growth retardation, weight loss/gain, polyuria and polydipsia compared to their true incidence based on direct enquiry or assessment (44). Longitudinal retrospective studies have shown that growth failure and weight gain can occur up to 3 years before the diagnosis of a craniopharyngioma, especially in the presence of hypothalamic infiltration (214). Since the diagnosis of GH deficiency requires dynamic endocrine testing, and idiopathic CPP can be a normal variant in young girls, a significant underlying lesion may be missed without mandatory neuroimaging, despite studies showing that 14-45% of female patients with CPP have a hypothalamo-pituitary mass (215–217). DI may remain occult in the ACTH-deficient patient, or unrecognized until the patient is water-deprived or rendered effectively adipsic by general anesthesia, coma or further hypothalamic damage sustained during surgery, with potentially fatal consequences. Lethargy, recurrent infections, somnolence, and cold intolerance may be subtle symptoms of ACTH and/or TSH deficiencies, whilst hypothalamic dysfunction (discussed below) manifesting as hyperphagia, escalating obesity, sleep-wake cycle disturbance, and temperature dysregulation may be mistaken for psychosocial dysfunction.

 

PRE-OPERATIVE ENDOCRINE ASSESSMENT AND MANAGEMENT OF HYPOTHALAMO-PITUITARY TUMORS

 

Due to their relative rarity and a general lack of data on optimum treatment strategies, all pediatric hypothalamo-pituitary tumors should be discussed in a multidisciplinary forum which comprises, at minimum, a neuro-oncologist, neuroradiologist, pediatric endocrinologist, and pituitary surgeon. Careful endocrine assessment with appropriate neuroimaging is vital before definitive therapy (Table 4). Early morning cortisol/ACTH measurements should ideally be performed before any dexamethasone is given for cerebral oedema, alongside paired urine and plasma osmolarities & electrolytes as these will influence perioperative fluid management. Plasma tumor markers (prolactin, β-hCG, α-fetoprotein) should be obtained prior to any surgical intervention regardless of radiological appearances, as both prolactinomas and germinomas can be treated medically without requiring a biopsy. In some cases, cerebrospinal fluid β-hCG and α-fetoprotein may be required to aid diagnosis. Early access to a pediatric endocrinologist enhances diagnostic decision-making and ensures appropriate peri-operative fluid management particularly in the presence of life-threatening salt/water and hypocortisolemic crises. If dexamethasone has not been commenced for peritumoral edema and where a patient’s hypothalamo-pituitary-adrenal status is unknown, parenteral hydrocortisone (2 mg/kg) should be given at anesthetic induction and 6-8 hourly thereafter for 48-72 hours (or via a continuous hydrocortisone infusion), weaning to maintenance doses over 5-10 days according to clinical status until this axis can be formally assessed with a synacthen test. Clinicians should be aware of cortisol’s permissive effects on the renal tubule for free water clearance; thus, its replacement will unmask occult DI. In this situation, precise fluid balance measurements and the judicious use of desmopressin by an experienced endocrinologist are required. GH, thyroxine and estradiol/ testosterone supplementation may also be necessary. It is important to note that thyroid hormone replacement should not be commenced until a patient is cortisol replete for at least 48 hours to avoid precipitating an adrenal crisis.

 

Table 4. Recommended Minimum Pre-Treatment Endocrine Assessment for Hypothalamo-Pituitary Tumors

Clinical assessment

Height

Weight

Sitting height

BMI

Tanner pubertal stage

Bone age

Endocrine biochemistry

IGF-1/IGF-BP3

LH, FSH, estradiol/testosterone

TSH, free T4 ± free T3

Early morning (8-9 am) cortisol & ACTH

Early morning paired urine & plasma osmolarities & electrolytes

Tumor markers

PRL

AFP

β-hCG

BMI, body mass index; IGF-1, insulin-like growth factor 1; IGF-BP3, insulin-like growth factor binding protein 3; LH, luteinizing hormone; FSH, follicle-stimulating hormone; TSH, thyroid stimulating hormone; T4, thyroxine; T3, triiodothyronine; ACTH, adrenocorticotrophic hormone; PRL, prolactin; AFP, alpha-fetoprotein; β-hCG, beta-human chorionic gonadotrophin.

 

Rare Emaciation/Failure To Thrive Syndromes

 

DIENCEPHALIC SYNDROME (DS)

 

DS is a rare syndrome of severe emaciation first described over 60 years ago typically seen in infants <2 years of age in the presence of a hypothalamic tumor (218). The original description incorporated four “major” criteria – profound emaciation (often leading to a multitude of misdirected investigations for failure to thrive), preserved (or accelerated) linear growth, hyperactivity, and euphoria – and three “minor” features: pallor without anemia, hypoglycemia, and hypotension. There is marked loss of subcutaneous fat despite increased caloric intake. Other associated features result from either tumor location (nystagmus, papilloedema, optic atrophy, vomiting, ataxia) or increased sympathetic tone (sweatiness, tremor). Classically, DS occurs in <10% of hypothalamic LGGs (11,209), but has also been described in suprasellar high grade gliomas (77,219), germinomas (220,221), teratomas (222), ependymomas (223), craniopharyngiomas (224), epidermoid cysts (225), and rarely with non-suprasellar lesions such as brainstem gliomas(226). Since Russell’s original description, however, the definition for DS has now too loosely broadened to include all cancer-related cachexia (227), with <4% of patients with DS having onset of symptoms at >2 years of age (220,228), and some publications reporting adult-onset DS where growth velocity is irrelevant (224,229). It is therefore becoming increasingly difficult to determine whether the patients described in these cases truly have DS or not. Its pathophysiology remains poorly understood, although the most consistent biochemical finding is of high random plasma GH concentrations that is neither suppressed by an oral glucose tolerance test, nor further stimulated by insulin-induced hypoglycemia, with low or normal IGF-1 concentrations, indicative of a GH-resistant state(77,230,231). Studies showing increased resting energy expenditure (232,233) support the theory of a dysregulated metabolism rather than abnormal caloric intake. At the time of writing, the next LGG trial is being designed to incorporate an international study of this rare entity, which is an independent risk factor for death, progression (11) and severe endocrine morbidity (25).

ANOREXIA AND EATING DISORDERS

 

Anorexia nervosa is an over-represented symptom in multiple published case reports of patients with hypothalamic lesions (particularly slow-growing germ cell tumors), with an average delay in diagnosis of nearly 3 years (234), though symptoms tend to resolve with appropriate therapy. Given the ventromedial and lateral hypothalamic location of the hunger and satiety centers, it is reasonable to postulate the effect of a suprasellar lesion on appetite. However, current understanding of the orexigenic and anorexigenic neuroendocrine regulators of tumor-related anorexia is still incomplete, and reports of non-suprasellar CNS tumors presenting with anorexia (227,235,236) suggest dysregulation beyond the hypothalamus, whilst the effect of inflammatory cytokines present in disseminated disease (tumor necrosis factor-α (TNF- α), interleukin-1 (IL-1), interleukin-6 (IL-6), interferon-γ (IFN- γ)), may also play a role (227). An intracranial lesion needs to be differentiated from true anorexia nervosa, which should fulfil DSM-V or ICD-10 criteria(237,238)), in all patients presenting with anorexia and weight loss. A full auxological, pubertal and endocrine biochemical assessment should be performed to exclude neuroendocrine disease, particularly in boys where the lower prevalence of anorexia nervosa requires mandatory pituitary neuroimaging. Anorexia nervosa presenting with amenorrhea may be due to a suprasellar tumor causing hypogonadotrophic hypogonadism (239), and initially normal imaging may not exclude an eventual diagnosis of a tumor, particularly for germinomas (235). Severe weight loss at diagnosis may be a predictor for future hypothalamic obesity (240).

 

NEUROENDOCRINE DYSFUNCTION AFTER DIAGNOSIS AND/OR TREATMENT

 

The Evolution Of Endocrinopathy And Its Association With Treatment

 

Whilst the initial endocrinopathies present at diagnosis are fairly typical for particular tumor subtypes, the pattern of post-treatment endocrine dysfunction in survivors of these lesions is interestingly very similar in frequency and timing. It has long been recognized that there is an evolution in the incidence of dysfunction in each of the hypothalamo-pituitary axes over time, closely mimicking that seen in congenital neurodevelopmental disorders such as septo-optic dysplasia (241). Although the various axes are differentially sensitive to irradiation, with the GH axis being the most susceptible (even at doses as low as 20 Gy), and the ACTH axis being the most robust (38,242,243), the similar evolutionary pattern of endocrine dysfunction seen in patients with a wide range of hypothalamo-pituitary lesions even in the absence of therapeutic irradiation suggests that the pattern of deficits is related most strongly to the position of the tumor (and thus recurrent disease) rather than treatment. GH deficiency is thus commonest, followed by gonadotrophin dysfunction (either central precocious puberty or hypogonadotrophic hypogonadism), ACTH and TSH deficiency, and least commonly posterior pituitary dysfunction, usually presenting as central DI (which is never seen after similar pituitary irradiation doses administered to non-suprasellar tumors) (25,37,45,145,158,244–247). Hence, lifelong endocrine follow-up of these survivors with regular clinical and biochemical assessments is vital as all patients with such tumors remain at high-risk for the development of these deficits. National guidelines on the neuroendocrine long-term follow-up of tumors such as craniopharyngiomas have been developed in the UK (49).

GH Deficiency

 

GH deficiency affects virtually all survivors of pediatric hypothalamo-pituitary lesions at some stage. If not already present at diagnosis, it is virtually guaranteed to occur after pituitary-directed therapy such as radiotherapy or surgery(45,248). Diagnosis of GH deficiency requires dynamic endocrine testing with the gold standard being the insulin tolerance test, although this is contraindicated in patients with a history of seizures. It is worth noting that the GHRH stimulation test should not be used in this context as it will not detect GH deficiency of hypothalamic origin (249). Serum IGF-1 and its binding protein IGF-BP3 are less accurate markers of GH deficiency, although they may be useful in severe growth failure in the context of a hypothalamo-pituitary tumor where GH testing is considered too hazardous (250,251). They should not be used in the context of suspected GH deficiency in the context of previous irradiation (252–254). Occasionally, GH deficiency may initially present with abnormal spontaneous secretion but normal peak responses to stimulation tests (termed “neurosecretory dysfunction”) (255), although testing for this with overnight GH profiling is not currently recommended by the GH Research Society (256).

 

Paradoxical normal growth may continue despite GH deficiency either due to precocious or accelerated puberty, or the syndrome of “growth without growth hormone”, where secondary hyperinsulinemia occurs due to the rapid weight gain observed post-treatment (257). Growth failure may also be masked by concurrent central precocious puberty. Both situations deserve prompt investigation and GH substitution which, in replacement doses, does not increase tumor recurrence (25,258–260), but promotes anabolism and lean body mass. This should therefore not be delayed beyond 12 months after definitive therapy (although this cut-off is arbitrary) (261), particularly in patients who have irreversible loss of height from spinal irradiation (e.g., for germinomas) (262).  

 

Gonadotrophin Dysfunction

 

Gonadotrophin dysfunction may manifest in three ways. Firstly, central precocious puberty (CPP) (defined as a testicular volume of ≥4 ml in a boy <9 years or breast budding in a girl <8 years) which, if not already present at diagnosis (e.g., hamartomas, LGGs, germinomas) is increased particularly by radiotherapy (243). There is no evidence that surgical resection of hypothalamic hamartomas, the commonest lesion associated with CPP, improves these symptoms, despite ameliorating the seizures (145). As mentioned above, coexistence of an early puberty with GH deficiency may mask the latter as height velocity may initially appear to be maintained or even accelerated, but not when corrected for bone age. Any child in puberty should therefore concurrently have an urgent assessment of GH secretion and consideration of replacement to restore height in combination with GnRH analogues to delay skeletal maturation if it is felt psychosocially appropriate. It is worth noting that prior CPP does not preclude later pubertal delay or arrest and may in fact be a risk factor (25). Therefore, careful monitoring is required even after the cessation of GnRH analogues.

 

Pubertal delay or arrest may either be due to hypogonadotrophic hypogonadism from tumor- or treatment-related injury to the hypothalamus (causing GnRH and/or LH/FSH deficiency) or to primary gonadal failure from systemic chemotherapy (hypergonadotrophic hypogonadism). Patients may fail to enter puberty altogether by the expected age (14 years in boys, 13 years in girls), enter puberty normally and subsequently fail to progress, or demonstrate secondary amenorrhea (girls) or sexual dysfunction (boys). In this situation concurrent GH deficiency can be corrected simultaneously or 6 months prior to commencing sex steroid replacement to initiate an appropriately-timed pubertal growth spurt. There is no advantage to adult height in delaying sex steroid replacement any further, particularly in the light of the benefits on bone mineral accretion (263).

 

Most chemotherapeutic drugs used in CNS tumor regimens (e.g., carboplatin, vincristine, etoposide) are not considered gonadotoxic, but other high-risk agents such as cyclophosphamide, temozolomide, and cisplatin are occasionally used, with their effects being modulated by age at exposure and gender (264). Since it is possible to protect future fertility in boys even as young as 12 years with some masculinization (Tanner stage 3+ and/or testicular volume of 8+ mls) by sperm cryopreservation, this should be considered before definitive therapy, even in those not receiving chemotherapy (265). By contrast, girls who have achieved regular spontaneous menses should be warned of the reduced window of reproductive capacity and a premature menopause due to a reduced ovarian follicular reserve (266). Notably, patients with hypothalamo-pituitary tumors who have received chemotherapy can potentially have concurrent hypogonadotrophic hypogonadism and primary gonadal failure, compounding the future risk of subfertility.

 

ACTH Deficiency/Central Adrenal Insufficiency

 

The hypothalamo-pituitary-adrenal (HPA) axis is fortunately relatively robust to irradiation and chemotherapeutic damage. However, in the context of a hypothalamo-pituitary tumor, the most important diagnostic challenge is to accurately determine adrenal reserve and differentiate reversible dexamethasone-induced ACTH suppression (after treatment for cerebral edema) from true, permanent ACTH deficiency. Given the lifelong implications of the latter, it is our opinion that the diagnosis should be carefully made ideally with the gold standard insulin tolerance test (ITT) and repeatedly reviewed with time. This may additionally necessitate regular plasma morning cortisol and ACTH measurements and 24-hour cortisol day curves. Although the standard synacthen test (SST) is often used to test adrenal integrity in adults, this supraphysiological stimulus does not test the entire pathway and the integrity of the hypothalamus or pituitary. There is evidence to suggest that in CNS tumor survivors the SST may be less sensitive than the ITT or low dose synacthen stimulation in detecting more subtle degrees of deficiency (267–269). In patients who have received peri-operative dexamethasone for peritumoral edema, formal testing of the HPA axis may be best left until 2-3 months after substitution with maintenance hydrocortisone as doses can be more safely omitted whilst testing is performed. Testing should be performed in a tertiary pediatric endocrinology unit used to managing patients with multiple endocrinopathies, with routine glucose rescue at 25-30 minutes and hydrocortisone at the end of low-dose (0.1 units/kg) insulin-induced hypoglycemia or glucagon stimulation. Treatment of adrenal insufficiency with glucocorticoids may unmask occult DI, and the coexistence of ACTH deficiency, DI, and adipsia due to hypothalamic damage can be fatal and should be avoided where possible.

 

TRH/TSH Deficiency/Central Hypothyroidism

 

The thyroid, like the hypothalamo-pituitary-gonadal axis, can be rendered underactive by either central TRH/TSH deficiency (inappropriately normal/low TSH for a low free T4 or T3) due to the tumor itself or surgery, or primary hypothyroidism (high TSH with a normal (compensated/subclinical) or low (frank) free T4) from spinal irradiation and/or chemotherapy, with the potential for the two states coexisting in some patients. There is little evidence for the role of irradiation in the former. In the adult cohort studied by Littley et al., no patients treated with low-dose radiotherapy alone experienced TSH deficiency (242). Similarly, Gan et al. found that the only independent risk factor for TSH deficiency in LGGs was hypothalamic involvement of the tumor (25). TRH stimulation tests may not differentiate hypothalamic (tertiary) from pituitary (secondary) damage, and serial thyroid function tests with two consecutive low free T4 concentrations in association with a low or inappropriately normal TSH concentration confirm the diagnosis without the need for further testing (270–272).

 

Primary hypothyroidism can present many years after the initial irradiation or chemotherapeutic insult. Annual thyroid function tests in at-risk children are important for early detection of subclinical hypothyroidism and institution of early treatment, particularly in light of the known effects on the developing brain. Given the known risk of radiation-associated second primary thyroid cancers, the carcinogenicity of nuclear fallouts, and the long-term cardiovascular mortality risk of subclinical hypothyroidism, few clinicians would leave a persistently raised TSH in such a patient cohort untreated (273).

 

Hyperprolactinemia

 

The importance of serum prolactin (PRL) measurements in the diagnosis of prolactinomas has already been discussed. Similarly, a rise in PRL levels can occur post-treatment in two situations. In the presence of a prolactinoma, this can indicate tumor “escape” from dopamine agonist (cabergoline/bromocriptine) control requiring further therapy. The more common situation arises where hyperprolactinemia is due to stalk compression by a progressive sellar or suprasellar tumor or hypothalamic damage. In this situation PRL concentrations are usually <2000 mU/l (274) and patients are unlikely to be symptomatic, with galactorrhea being unusual(25). Chronic severe primary hypothyroidism will also lead to hyperprolactinemia due to the stimulatory effects of a raised TRH on the lactotroph.

 

Posterior Pituitary Dysfunction (PPD)

 

Posterior pituitary dysfunction can present itself in three ways – DI, SIADH, or cerebral salt-wasting syndrome (CSW), the latter attributed to hypersecretion of cerebral atrial natriuretic (ANP) and brain natriuretic peptides (BNP) in response to plasma volume expansion by ADH. The latter two syndromes are rare outside the context of an acute cerebral insult and are usually transient, whilst DI may be a presenting feature and/or a permanent post-operative deficit. The absence of a posterior pituitary bright spot on MRI is a relatively sensitive marker of a lack of neurohypophyseal integrity (275–277). DI does not develop after cranial irradiation in the absence of a hypothalamo-pituitary tumor or surgery to the area (25,99). Whilst PPD is the least common form of endocrinopathy, the rapid shifts from hyper- to hyponatremia in the acute setting can prove life-threatening, as evidenced by a recent retrospective cohort study of optic pathway LGGs with high survival showing showed that nearly 50% of the deaths that occurred were associated with uncontrolled PPD (25). This risk is further increased by coexistent ACTH deficiency, hypothalamic adipsia, and treatment with anti-epileptic medications, which have SIADH-like effects.

 

After hypothalamo-pituitary surgery, PPD presents as a well-described triphasic response in ADH secretion: firstly, immediate but transient DI up to day 2; secondly, SIADH from day 1-14; and finally, a second phase of DI, which is usually permanent if it persists beyond 21 days, the preceding SIADH is prolonged or severe, or if extensive surgery has been performed (278,279). This triphasic response is thought to result from necrosis of hypothalamic ADH-secreting magnocellular neurons and is seen more often in children than adults (23% vs. 14% in one craniopharyngioma study) (280). The three phases may also occur independently, and cerebral salt-wasting syndrome may coexist and complicate diagnosis and management. Dramatic changes in sodium concentrations can therefore occur with the inherent risk of seizures, cerebral edema and death; such patients require high intensity care with precise fluid management supervised by an experienced pediatric endocrinologist. The measurement of plasma and urinary arginine-vasopressin (AVP) may help differentiate between these different disorders, but these assays are not widely available and careful sample processing is required prior to analysis (281). More recently, measurement of plasma copeptin, the more stable by-product of cleavage of AVP from its carrier protein neurophysin II, is becoming more widely available and has been shown to be a more easily measurable, sensitive, and specific surrogate marker of AVP secretion (282–284).

 

Detailed management of these disorders is beyond the scope of this chapter, but can be summarized in the algorithm seen in Figure 8.

Figure 8. Algorithm for the management of post-operative salt-water balance disorders (53).

 

The Hypothalamic Syndrome

 

The hypothalamic “syndrome” is loosely defined and usually refers to a constellation of features attributed to hypothalamic dysfunction. Central to it is hypothalamic obesity, a morbid, inexorably escalating obesity (BMI usually >+3 SDS) first described over a century ago (285). It occurs in up to 77% of craniopharyngiomas, 53% of optic pathway LGGs, 40% of pituitary adenomas, 40% of germinomas, and 23% of hamartomas (64,145,286–288), with some symptoms occurring at diagnosis prior to any treatment. Despite this, its pathophysiology is still poorly understood, although it is becoming increasingly evident that both hyperphagia and a dysregulation of anorexigenic and orexigenic hormones contribute (289). Young age at diagnosis, hypothalamic injury by tumor, high dose irradiation or surgery (including biopsies), and multiple endocrinopathies are all risk factors (278,289). Unlike common obesity, the weight gain is largely resistant to caloric restriction, lifestyle interventions, medical and surgical therapies (290–295). Several authors have recently trialed GLP-1 agonists in hypothalamic obesity with some success (296–298), but a randomized control trial is needed to confirm these findings in the longer-term, particularly given the newly published data demonstrating long-term success with common obesity (299). More recently, the combination of tesofensine (a monoamine reuptake inhibitor) and metoprolol has shown promising results in a phase 2 trial (300).

 

Other hypothalamic symptoms include sleep-wake cycle disturbances, adipsia, temperature dysregulation, cognitive (particularly memory loss), and behavioral (particularly autistic) disorders. Children with disturbed sleep and/or behavioral difficulties should be referred to a specialist sleep laboratory and behavioral neuropsychopharmacology unit. These disorders are even more difficult to treat than replacement of the endocrine deficits. Where endocrine deficits, particularly ACTH deficiency and DI coexist, hypothalamic adipsia is potentially fatal particularly during intercurrent illness and surgery, requiring careful day-to-day fluid management with obligate daily fluid intake and desmopressin dose adjustments. The difficulties in managing patients with panhypopituitarism with concurrent hypothalamic dysfunction should not be underestimated, therefore avoiding these complications must be an important aim of initial therapy.

 

CONCLUSIONS

 

Pediatric hypothalamo-pituitary tumors are uncommon, and may present with occult or unusual clinical features posing diagnostic dilemmas that incur treatment delays or necessitate prolonged MRI surveillance. Notwithstanding their generally high survival rates, tumor- or treatment-related neuroendocrine morbidity is very significant and not always simply reversible by hormone replacement therapy. Consequently, treatment decision-making should aim to preserve not only visual, but also hypothalamo-pituitary function. Pediatric endocrinologists and pituitary surgeons should be part of the decision-making multidisciplinary team, with radiological, visual, and biochemical assessments together aiding management planning. A detailed baseline endocrine assessment is paramount to both diagnosis and treatment and will ultimately improve long-term outcome monitoring, the clarification of tumor- and treatment-related consequences and management of lifelong morbidity. Given the potentially significant reduction in health-related quality of survival, lifelong, age-appropriate follow-up and management within a dedicated multidisciplinary neuroendocrine unit familiar with the complexity of patients’ needs is recommended. To achieve this, rehabilitation, reproductive, neuropsychological, and vocational services need developing further in parallel with appropriate transition processes to adult services if we are to better manage and improve outcomes for this high-risk group of young patients. Evidence- and consensus-based guidelines are increasingly being developed to define a standard of best practice for the management of these rare tumors.

 

REFERENCES

 

  1. Baade PD, Youlden DR, Valery PC, Hassall T, Ward L, Green AC, et al. Trends in incidence of childhood cancer in Australia, 1983-2006. Br J Cancer [Internet]. 2010/01/07. 2010;102(3):620–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20051948
  2. Childhood Cancer Research Group . The National Registry of Childhood Tumours. Oxford: Childhood Cancer Research Group; 2012.
  3. Stiller C. Childhood cancer in Britain: incidence, survival, mortality. Oxford: Oxford University Press; 2007.
  4. Department of Health ., Macmillan Cancer Support ., NHS Improvement . Living with and beyond cancer: taking action to improve outcomes. London: National Cancer Survivorship Initiative (NCSI), Department of Health; 2013.
  5. Ward EM, Thun MJ, Hannan LM, Jemal A. Interpreting cancer trends. Ann N Y Acad Sci [Internet]. 2006/11/23. 2006;1076:29–53. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17119192
  6. Adamson P, Law G, Roman E. Assessment of trends in childhood cancer incidence. Lancet [Internet]. 2005/03/01. 2005;365(9461):753. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15733714
  7. Steliarova-Foucher E, Stiller C, Kaatsch P, Berrino F, Coebergh JW. Trends in childhood cancer incidence in Europe, 1970-99. Lancet [Internet]. 2005/06/21. 2005;365(9477):2088. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15964441
  8. Hjalmars U, Kulldorff M, Wahlqvist Y, Lannering B. Increased incidence rates but no space-time clustering of childhood astrocytoma in Sweden, 1973-1992: a population-based study of pediatric brain tumors. Cancer [Internet]. 1999/05/01. 1999;85(9):2077–90. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10223251
  9. Royal College of Paediatrics & Child Health ., Samantha Dickson Brain Tumour Trust ., Children’s Brain Tumour Research Centre ., The Health Foundation . The diagnosis of brain tumours in children: an evidence-based guideline to assist healthcare professionals in the assessment of children presenting with symptoms and signs that may be due to a brain tumour. 3rd ed. Nottingham: Children’s Brain Tumour Research Centre; 2011.
  10. Gatta G, Capocaccia R, Stiller C, Kaatsch P, Berrino F, Terenziani M. Childhood cancer survival trends in Europe: a EUROCARE Working Group study. J Clin Oncol [Internet]. 2005/06/01. 2005;23(16):3742–51. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15923571
  11. Gnekow AK, Falkenstein F, von Hornstein S, Zwiener I, Berkefeld S, Bison B, et al. Long-term follow-up of the multicenter, multidisciplinary treatment study HIT-LGG-1996 for low-grade glioma in children and adolescents of the German Speaking Society of Pediatric Oncology and Hematology. Neuro Oncol [Internet]. 2012/09/04. 2012;14(10):1265–84. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22942186
  12. Stokland T, Liu JF, Ironside JW, Ellison DW, Taylor R, Robinson KJ, et al. A multivariate analysis of factors determining tumor progression in childhood low-grade glioma: a population-based cohort study (CCLG CNS9702). Neuro Oncol [Internet]. 2010/09/24. 2010;12(12):1257–68. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20861086
  13. Skinner R, Wallace WH, Levitt G. Long-term follow-up of children treated for cancer: why is it necessary, by whom, where and how? Arch Dis Child [Internet]. 2007/03/06. 2007;92(3):257–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17337686
  14. Skinner R, Wallace WHB, Levitt GA. Therapy based long-term follow-up. 2nd ed. UK Children’s Cancer Study Group (UK CCSG) Late Effects Group; 2005.
  15. Wallace WH, Thompson L, Anderson RA. Long term follow-up of survivors of childhood cancer: summary of updated SIGN guidance. BMJ [Internet]. 2013/03/29. 2013;346:f1190. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23535255
  16. Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM, Meadows AT, et al. Chronic health conditions in adult survivors of childhood cancer. N Engl J Med [Internet]. 2006/10/13. 2006;355(15):1572–82. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17035650
  17. Brignardello E, Felicetti F, Castiglione A, Chiabotto P, Corrias A, Fagioli F, et al. Endocrine health conditions in adult survivors of childhood cancer: the need for specialized adult-focused follow-up clinics. Eur J Endocrinol [Internet]. 2012/12/22. 2013;168(3):465–72. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23258270
  18. Regal M, Paramo C, Sierra SM, Garcia-Mayor R v. Prevalence and incidence of hypopituitarism in an adult Caucasian population in northwestern Spain. Clin Endocrinol (Oxf) [Internet]. 2001;55(6):735–40. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11895214
  19. Tanriverdi F, Dokmetas HS, Kebapci N, Kilicli F, Atmaca H, Yarman S, et al. Etiology of hypopituitarism in tertiary care institutions in Turkish population: analysis of 773 patients from Pituitary Study Group database. Endocrine [Internet]. 2014;47(1):198–205. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24366641
  20. Andoniadou CL, Gaston-Massuet C, Reddy R, Schneider RP, Blasco MA, le Tissier P, et al. Identification of novel pathways involved in the pathogenesis of human adamantinomatous craniopharyngioma. Acta Neuropathol [Internet]. 2012/02/22. 2012;124(2):259–71. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22349813
  21. Gaston-Massuet C, Andoniadou CL, Signore M, Jayakody SA, Charolidi N, Kyeyune R, et al. Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proc Natl Acad Sci U S A [Internet]. 2011/06/04. 2011;108(28):11482–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21636786
  22. Muller HL, Emser A, Faldum A, Bruhnken G, Etavard-Gorris N, Gebhardt U, et al. Longitudinal study on growth and body mass index before and after diagnosis of childhood craniopharyngioma. J Clin Endocrinol Metab [Internet]. 2004/07/09. 2004;89(7):3298–305. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15240606
  23. Arora RS, Alston RD, Eden TO, Estlin EJ, Moran A, Birch JM. Age-incidence patterns of primary CNS tumors in children, adolescents, and adults in England. Neuro Oncol [Internet]. 2008/11/27. 2009;11(4):403–13. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19033157
  24. Schroeder JW, Vezina LG. Pediatric sellar and suprasellar lesions. Pediatr Radiol [Internet]. 2011/01/27. 2011;41(3):287–98. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21267556
  25. Gan HW, Phipps K, Aquilina K, Gaze MN, Hayward R, Spoudeas HA. Neuroendocrine Morbidity After Pediatric Optic Gliomas: A Longitudinal Analysis of 166 Children Over 30 Years. J Clin Endocrinol Metab [Internet]. 2015;100(10):3787–99. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26218754
  26. Warmuth-Metz M, Gnekow AK, Muller H, Solymosi L. Differential diagnosis of suprasellar tumors in children. Klin Padiatr [Internet]. 2004/11/27. 2004;216(6):323–30. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15565547
  27. Kaatsch P, Rickert CH, Kuhl J, Schuz J, Michaelis J. Population-based epidemiologic data on brain tumors in German children. Cancer [Internet]. 2001;92(12):3155–64. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11753995
  28. May JA, Krieger MD, Bowen I, Geffner ME. Craniopharyngioma in childhood. Adv Pediatr [Internet]. 2006;53:183–209. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17089867
  29. Bunin GR, Surawicz TS, Witman PA, Preston-Martin S, Davis F, Bruner JM. The descriptive epidemiology of craniopharyngioma. J Neurosurg [Internet]. 1998/10/07. 1998;89(4):547–51. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9761047
  30. Stiller CA, Nectoux J. International incidence of childhood brain and spinal tumours. Int J Epidemiol [Internet]. 1994/06/01. 1994;23(3):458–64. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7960369
  31. Nielsen EH, Feldt-Rasmussen U, Poulsgaard L, Kristensen LO, Astrup J, Jorgensen JO, et al. Incidence of craniopharyngioma in Denmark (n = 189) and estimated world incidence of craniopharyngioma in children and adults. J Neurooncol [Internet]. 2011/02/22. 2011;104(3):755–63. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21336771
  32. Zhang YQ, Wang CC, Ma ZY. Pediatric craniopharyngiomas: clinicomorphological study of 189 cases. Pediatr Neurosurg [Internet]. 2002/03/15. 2002;36(2):80–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11893889
  33. Boch AL, van Effenterre R, Kujas M. Craniopharyngiomas in two consanguineous siblings: case report. Neurosurgery [Internet]. 1997;41(5):1185–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9361074
  34. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-Santagata D, Thorner AR, et al. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nat Genet. 2014;46(2):161–5.
  35. Caldarelli M, Massimi L, Tamburrini G, Cappa M, di Rocco C. Long-term results of the surgical treatment of craniopharyngioma: the experience at the Policlinico Gemelli, Catholic University, Rome. Childs Nerv Syst [Internet]. 2005/07/05. 2005;21(8–9):747–57. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15995885
  36. Hoffman HJ, de Silva M, Humphreys RP, Drake JM, Smith ML, Blaser SI. Aggressive surgical management of craniopharyngiomas in children. J Neurosurg [Internet]. 1992/01/01. 1992;76(1):47–52. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1727168
  37. Karavitaki N, Brufani C, Warner JT, Adams CB, Richards P, Ansorge O, et al. Craniopharyngiomas in children and adults: systematic analysis of 121 cases with long-term follow-up. Clin Endocrinol (Oxf) [Internet]. 2005/04/06. 2005;62(4):397–409. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15807869
  38. Merchant TE, Kiehna EN, Sanford RA, Mulhern RK, Thompson SJ, Wilson MW, et al. Craniopharyngioma: the St. Jude Children’s Research Hospital experience 1984-2001. Int J Radiat Oncol Biol Phys [Internet]. 2002/06/14. 2002;53(3):533–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12062594
  39. Puget S, Garnett M, Wray A, Grill J, Habrand JL, Bodaert N, et al. Pediatric craniopharyngiomas: classification and treatment according to the degree of hypothalamic involvement. J Neurosurg [Internet]. 2007/01/20. 2007;106(1 Suppl):3–12. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17233305
  40. van Effenterre R, Boch AL. Craniopharyngioma in adults and children: a study of 122 surgical cases. J Neurosurg [Internet]. 2002/07/24. 2002;97(1):3–11. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12134929
  41. de Vries L, Lazar L, Phillip M. Craniopharyngioma: presentation and endocrine sequelae in 36 children. J Pediatr Endocrinol Metab [Internet]. 2003/07/26. 2003;16(5):703–10. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12880119
  42. Hetelekidis S, Barnes PD, Tao ML, Fischer EG, Schneider L, Scott RM, et al. 20-year experience in childhood craniopharyngioma. Int J Radiat Oncol Biol Phys [Internet]. 1993/09/30. 1993;27(2):189–95. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8407391
  43. Lin LL, el Naqa I, Leonard JR, Park TS, Hollander AS, Michalski JM, et al. Long-term outcome in children treated for craniopharyngioma with and without radiotherapy. J Neurosurg Pediatr [Internet]. 2008/03/21. 2008;1(2):126–30. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18352781
  44. Muller HL. Childhood craniopharyngioma--current concepts in diagnosis, therapy and follow-up. Nat Rev Endocrinol [Internet]. 2010/09/30. 2010;6(11):609–18. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20877295
  45. DeVile CJ, Grant DB, Hayward RD, Stanhope R. Growth and endocrine sequelae of craniopharyngioma. Arch Dis Child [Internet]. 1996/08/01. 1996;75(2):108–14. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8869189
  46. Sorva R, Heiskanen O, Perheentupa J. Craniopharyngioma surgery in children: endocrine and visual outcome. Childs Nerv Syst [Internet]. 1988/04/01. 1988;4(2):97–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/3401877
  47. de Vries L, Weintrob N, Phillip M. Craniopharyngioma presenting as precocious puberty and accelerated growth. Clin Pediatr (Phila) [Internet]. 2003;42(2):181–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12659393
  48. Molla E, Marti-Bonmati L, Revert A, Arana E, Menor F, Dosda R, et al. Craniopharyngiomas: identification of different semiological patterns with MRI. Eur Radiol [Internet]. 2002/07/12. 2002;12(7):1829–36. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12111075
  49. Children’s Cancer & Leukaemia Group (CCLG) . Craniopharyngioma: guideline for the management of children and young people (CYP) aged <19 years. Leicester, UK: CCLG; 2021.
  50. Flitsch J, Muller HL, Burkhardt T. Surgical strategies in childhood craniopharyngioma. Front Endocrinol (Lausanne) [Internet]. 2011/01/01. 2011;2:96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22645514
  51. de Vile CJ, Grant DB, Kendall BE, Neville BG, Stanhope R, Watkins KE, et al. Management of childhood craniopharyngioma: can the morbidity of radical surgery be predicted? J Neurosurg [Internet]. 1996/07/01. 1996;85(1):73–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8683285
  52. Muller HL, Albanese A, Calaminus G, Hargrave D, Garre ML, Gebhardt U, et al. Consensus and perspectives on treatment strategies in childhood craniopharyngioma: results of a meeting of the Craniopharyngioma Study Group (SIOP), Genova, 2004. J Pediatr Endocrinol Metab [Internet]. 2006;19 Suppl 1:453–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16700324
  53. Spoudeas HA, Harrison B, Spoudeas HA, Harrison B. Paediatric Endocrine Tumours: A Multidisciplinary Consensus Statement of Best Practice from a Working Group Convened Under the Auspices of the BSPED and UKCCSG (rare tumour working groups). 1st ed. Crawley: Novo Nordisk Ltd.; 2005.
  54. Clark AJ, Cage TA, Aranda D, Parsa AT, Sun PP, Auguste KI, et al. A systematic review of the results of surgery and radiotherapy on tumor control for pediatric craniopharyngioma. Childs Nerv Syst [Internet]. 2013;29(2):231–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23089933
  55. Iannalfi A, Fragkandrea I, Brock J, Saran F. Radiotherapy in craniopharyngiomas. Clin Oncol (R Coll Radiol) [Internet]. 2013;25(11):654–67. Available from: https://www.ncbi.nlm.nih.gov/pubmed/23910225
  56. Stripp DC, Maity A, Janss AJ, Belasco JB, Tochner ZA, Goldwein JW, et al. Surgery with or without radiation therapy in the management of craniopharyngiomas in children and young adults. Int J Radiat Oncol Biol Phys [Internet]. 2004/02/18. 2004;58(3):714–20. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14967425
  57. Bishop AJ, Greenfield B, Mahajan A, Paulino AC, Okcu MF, Allen PK, et al. Proton beam therapy versus conformal photon radiation therapy for childhood craniopharyngioma: multi-institutional analysis of outcomes, cyst dynamics, and toxicity. Int J Radiat Oncol Biol Phys [Internet]. 2014;90(2):354–61. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25052561
  58. Leroy R, Benahmed N, Hulstaert F, van Damme N, de Ruysscher D. Proton Therapy in Children: A Systematic Review of Clinical Effectiveness in 15 Pediatric Cancers. Int J Radiat Oncol Biol Phys [Internet]. 2016;95(1):267–78. Available from: https://www.ncbi.nlm.nih.gov/pubmed/27084646
  59. Bremer AM, Nguyen TQ, Balsys R. Therapeutic benefits of combination chemotherapy with vincristine, BCNU, and procarbazine on recurrent cystic craniopharyngioma. A case report. J Neurooncol [Internet]. 1984/01/01. 1984;2(1):47–51. Available from: http://www.ncbi.nlm.nih.gov/pubmed/6470759
  60. Lippens RJ, Rotteveel JJ, Otten BJ, Merx H. Chemotherapy with Adriamycin (doxorubicin) and CCNU (lomustine) in four children with recurrent craniopharyngioma. Eur J Paediatr Neurol [Internet]. 2000/03/22. 1998;2(5):263–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10726829
  61. Bartels U, Laperriere N, Bouffet E, Drake J. Intracystic therapies for cystic craniopharyngioma in childhood. Front Endocrinol (Lausanne) [Internet]. 2012/06/02. 2012;3:39. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22654864
  62. Cavalheiro S, di Rocco C, Valenzuela S, Dastoli PA, Tamburrini G, Massimi L, et al. Craniopharyngiomas: intratumoral chemotherapy with interferon-alpha: a multicenter preliminary study with 60 cases. Neurosurg Focus [Internet]. 2010/04/07. 2010;28(4):E12. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20367356
  63. Crom DB, Smith D, Xiong Z, Onar A, Hudson MM, Merchant TE, et al. Health status in long-term survivors of pediatric craniopharyngiomas. J Neurosci Nurs [Internet]. 2011/01/07. 2010;42(6):323–8; quiz 329–30. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21207770
  64. Armstrong GT, Conklin HM, Huang S, Srivastava D, Sanford R, Ellison DW, et al. Survival and long-term health and cognitive outcomes after low-grade glioma. Neuro Oncol [Internet]. 2010/12/24. 2011;13(2):223–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21177781
  65. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol [Internet]. 2007/07/10. 2007;114(2):97–109. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17618441
  66. Bataini JP, Delanian S, Ponvert D. Chiasmal gliomas: results of irradiation management in 57 patients and review of literature. Int J Radiat Oncol Biol Phys [Internet]. 1991/08/01. 1991;21(3):615–23. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1907959
  67. Gnekow AK, Kortmann RD, Pietsch T, Emser A. Low grade chiasmatic-hypothalamic glioma-carboplatin and vincristin chemotherapy effectively defers radiotherapy within a comprehensive treatment strategy -- report from the multicenter treatment study for children and adolescents with a low grade glioma -- HIT-LGG 1996 -- of the Society of Pediatric Oncology and Hematology (GPOH). Klin Padiatr [Internet]. 2004/11/27. 2004;216(6):331–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15565548
  68. Janss AJ, Grundy R, Cnaan A, Savino PJ, Packer RJ, Zackai EH, et al. Optic pathway and hypothalamic/chiasmatic gliomas in children younger than age 5 years with a 6-year follow-up. Cancer [Internet]. 1995/02/15. 1995;75(4):1051–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7842408
  69. Medlock MD, Madsen JR, Barnes PD, Anthony DS, Cohen LE, Scott RM. Optic chiasm astrocytomas of childhood. 1. Long-term follow-up. Pediatr Neurosurg [Internet]. 1998/04/21. 1997;27(3):121–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9548522
  70. Dasgupta B, Li W, Perry A, Gutmann DH. Glioma formation in neurofibromatosis 1 reflects preferential activation of K-RAS in astrocytes. Cancer Res [Internet]. 2005/01/25. 2005;65(1):236–45. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15665300
  71. Lawson AR, Tatevossian RG, Phipps KP, Picker SR, Michalski A, Sheer D, et al. RAF gene fusions are specific to pilocytic astrocytoma in a broad paediatric brain tumour cohort. Acta Neuropathol [Internet]. 2010/05/11. 2010;120(2):271–3. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20454969
  72. Hargrave DR, Bouffet E, Tabori U, Broniscer A, Cohen KJ, Hansford JR, et al. Efficacy and Safety of Dabrafenib in Pediatric Patients with BRAF V600 Mutation-Positive Relapsed or Refractory Low-Grade Glioma: Results from a Phase I/IIa Study. Clin Cancer Res [Internet]. 2019;25(24):7303–11. Available from: https://www.ncbi.nlm.nih.gov/pubmed/31811016
  73. Campagna M, Opocher E, Viscardi E, Calderone M, Severino SM, Cermakova I, et al. Optic pathway glioma: long-term visual outcome in children without neurofibromatosis type-1. Pediatr Blood Cancer [Internet]. 2010/10/28. 2010;55(6):1083–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20979170
  74. Jaing TH, Lin KL, Tsay PK, Hsueh C, Hung PC, Wu CT, et al. Treatment of optic pathway hypothalamic gliomas in childhood: experience with 18 consecutive cases. J Pediatr Hematol Oncol [Internet]. 2008/04/01. 2008;30(3):222–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18376285
  75. Tao ML, Barnes PD, Billett AL, Leong T, Shrieve DC, Scott RM, et al. Childhood optic chiasm gliomas: radiographic response following radiotherapy and long-term clinical outcome. Int J Radiat Oncol Biol Phys [Internet]. 1997/10/23. 1997;39(3):579–87. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9336136
  76. Brauner R, Trivin C, Zerah M, Souberbielle JC, Doz F, Kalifa C, et al. Diencephalic syndrome due to hypothalamic tumor: a model of the relationship between weight and puberty onset. J Clin Endocrinol Metab [Internet]. 2006/04/20. 2006;91(7):2467–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16621905
  77. Fleischman A, Brue C, Poussaint TY, Kieran M, Pomeroy SL, Goumnerova L, et al. Diencephalic syndrome: a cause of failure to thrive and a model of partial growth hormone resistance. Pediatrics [Internet]. 2005/06/03. 2005;115(6):e742-8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15930202
  78. Suarez JC, Viano JC, Zunino S, Herrera EJ, Gomez J, Tramunt B, et al. Management of child optic pathway gliomas: new therapeutical option. Childs Nerv Syst [Internet]. 2006/01/04. 2006;22(7):679–84. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16389565
  79. Picariello S, Cerbone M, D’Arco F, Gan HW, O’Hare P, Aquilina K, et al. A 40-Year Cohort Study of Evolving Hypothalamic Dysfunction in Infants and Young Children (<3 years) with Optic Pathway Gliomas. Cancers (Basel). 2022 Jan 31;14(3).
  80. Mulhern RK, Merchant TE, Gajjar A, Reddick WE, Kun LE. Late neurocognitive sequelae in survivors of brain tumours in childhood. Lancet Oncol [Internet]. 2004/07/03. 2004;5(7):399–408. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15231246
  81. Friedman DL, Whitton J, Leisenring W, Mertens AC, Hammond S, Stovall M, et al. Subsequent neoplasms in 5-year survivors of childhood cancer: the Childhood Cancer Survivor Study. J Natl Cancer Inst [Internet]. 2010/07/17. 2010;102(14):1083–95. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20634481
  82. Taylor AJ, Little MP, Winter DL, Sugden E, Ellison DW, Stiller CA, et al. Population-based risks of CNS tumors in survivors of childhood cancer: the British Childhood Cancer Survivor Study. J Clin Oncol [Internet]. 2010/11/17. 2010;28(36):5287–93. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21079138
  83. Ullrich NJ, Robertson R, Kinnamon DD, Scott RM, Kieran MW, Turner CD, et al. Moyamoya following cranial irradiation for primary brain tumors in children. Neurology [Internet]. 2007/03/21. 2007;68(12):932–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17372129
  84. Dalla Via P, Opocher E, Pinello ML, Calderone M, Viscardi E, Clementi M, et al. Visual outcome of a cohort of children with neurofibromatosis type 1 and optic pathway glioma followed by a pediatric neuro-oncology program. Neuro Oncol [Internet]. 2007/08/21. 2007;9(4):430–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17704361
  85. Wen PY, Stein A, van den Bent M, de Greve J, Wick A, de Vos FYFL, et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial. Lancet Oncol. 2022;23(1):53–64.
  86. Perreault S, Larouche V, Tabori U, Hawkin C, Lippe S, Ellezam B, et al. A phase 2 study of trametinib for patients with pediatric glioma or plexiform neurofibroma with refractory tumor and activation of the MAPK/ERK pathway: TRAM-01. BMC Cancer [Internet]. 2019;19(1):1250. Available from: https://www.ncbi.nlm.nih.gov/pubmed/31881853
  87. Selt F, van Tilburg CM, Bison B, Sievers P, Harting I, Ecker J, et al. Response to trametinib treatment in progressive pediatric low-grade glioma patients. J Neurooncol. 2020 Sep;149(3):499–510.
  88. Gan HW. Management of Craniopharyngiomas in the Era of Molecular Oncological Therapies: Not a Panacea. J Endocr Soc. 2021 Jul 1;5(7):bvab094.
  89. Gillam MP, Molitch ME, Lombardi G, Colao A. Advances in the treatment of prolactinomas. Endocr Rev [Internet]. 2006/05/18. 2006;27(5):485–534. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16705142
  90. Fideleff HL, Boquete HR, Suarez MG, Azaretzky M. Prolactinoma in children and adolescents. Horm Res [Internet]. 2009/09/30. 2009;72(4):197–205. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19786791
  91. Harrington MH, Casella SJ. Pituitary tumors in childhood. Curr Opin Endocrinol Diabetes Obes [Internet]. 2011/12/14. 2012;19(1):63–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22157404
  92. Colao A, Loche S. Prolactinomas in children and adolescents. Endocr Dev [Internet]. 2009/12/04. 2010;17:146–59. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19955764
  93. Diamond Jr. FB. Pituitary adenomas in childhood: development and diagnosis. Fetal Pediatr Pathol [Internet]. 2007/08/19. 2006;25(6):339–56. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17696045
  94. Beckers A, Rostomyan L, Daly AF. Overview of genetic testing in patients with pituitary adenomas. Ann Endocrinol (Paris) [Internet]. 2012/04/17. 2012;73(2):62–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22503805
  95. Gan HW, Bulwer C, Jeelani O, Levine MA, Korbonits M, Spoudeas HA. Treatment-resistant pediatric giant prolactinoma and multiple endocrine neoplasia type 1. Int J Pediatr Endocrinol [Internet]. 2015;2015(1):15. Available from: https://www.ncbi.nlm.nih.gov/pubmed/26180530
  96. Korbonits M, Storr H, Kumar A v. Familial pituitary adenomas - who should be tested for AIP mutations? Clin Endocrinol (Oxf) [Internet]. 2012;77(3):351–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22612670
  97. Alband N, Korbonits M. Familial pituitary tumors. Handb Clin Neurol [Internet]. 2014;124:339–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25248598
  98. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab [Internet]. 2011/02/08. 2011;96(2):273–88. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21296991
  99. Moraes AB, Silva CM, Vieira Neto L, Gadelha MR. Giant prolactinomas: the therapeutic approach. Clin Endocrinol (Oxf) [Internet]. 2013/05/15. 2013;79(4):447–56. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23662975
  100. Schade R, Andersohn F, Suissa S, Haverkamp W, Garbe E. Dopamine agonists and the risk of cardiac-valve regurgitation. N Engl J Med. 2007 Jan 4;356(1):29–38.
  101. Bulwer C, Gan HW, Stern E, Powell M, Jeelani O, Korbonits M, et al. Managing rare, resistant, macro- and giant prolactinomas causing raised intracranial pressure in children: lessons learnt at a single centre. Horm Res Paediatr. 2013;80(Suppl 1):165.
  102. Steele CA, MacFarlane IA, Blair J, Cuthbertson DJ, Didi M, Mallucci C, et al. Pituitary adenomas in childhood, adolescence and young adulthood: presentation, management, endocrine and metabolic outcomes. Eur J Endocrinol [Internet]. 2010/08/06. 2010;163(4):515–22. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20685833
  103. Mindermann T, Wilson CB. Pediatric pituitary adenomas. Neurosurgery [Internet]. 1995;36(2):259–68; discussion 269. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7731505
  104. Savage MO, Storr HL, Chan LF, Grossman AB. Diagnosis and treatment of pediatric Cushing’s disease. Pituitary [Internet]. 2007;10(4):365–71. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17570065
  105. Joshi SM, Hewitt RJ, Storr HL, Rezajooi K, Ellamushi H, Grossman AB, et al. Cushing’s disease in children and adolescents: 20 years of experience in a single neurosurgical center. Neurosurgery [Internet]. 2005;57(2):281–5; discussion 281-5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16094156
  106. Guemes M, Murray PG, Brain CE, Spoudeas HA, Peters CJ, Hindmarsh PC, et al. Management of Cushing syndrome in children and adolescents: experience of a single tertiary centre. Eur J Pediatr [Internet]. 2016;175(7):967–76. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27169546
  107. Batista DL, Riar J, Keil M, Stratakis CA. Diagnostic tests for children who are referred for the investigation of Cushing syndrome. Pediatrics [Internet]. 2007;120(3):e575-86. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17698579
  108. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, et al. The diagnosis of Cushing’s syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab [Internet]. 2008;93(5):1526–40. Available from: https://www.ncbi.nlm.nih.gov/pubmed/18334580
  109. Pecori Giraldi F, Pivonello R, Ambrogio AG, de Martino MC, de Martin M, Scacchi M, et al. The dexamethasone-suppressed corticotropin-releasing hormone stimulation test and the desmopressin test to distinguish Cushing’s syndrome from pseudo-Cushing’s states. Clin Endocrinol (Oxf) [Internet]. 2007;66(2):251–7. Available from: https://www.ncbi.nlm.nih.gov/pubmed/17223996
  110. Wood PJ, Barth JH, Freedman DB, Perry L, Sheridan B. Evidence for the low dose dexamethasone suppression test to screen for Cushing’s syndrome--recommendations for a protocol for biochemistry laboratories. Ann Clin Biochem [Internet]. 1997;34 ( Pt 3):222–9. Available from: https://www.ncbi.nlm.nih.gov/pubmed/9158818
  111. Magiakou MA, Mastorakos G, Oldfield EH, Gomez MT, Doppman JL, Cutler Jr. GB, et al. Cushing’s syndrome in children and adolescents. Presentation, diagnosis, and therapy. N Engl J Med [Internet]. 1994;331(10):629–36. Available from: https://www.ncbi.nlm.nih.gov/pubmed/8052272
  112. Hopwood NJ, Kenny FM. Incidence of Nelson’s syndrome after adrenalectomy for Cushing’s disease in children: results of a nationwide survey. Am J Dis Child [Internet]. 1977;131(12):1353–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/930887
  113. Atkinson AB, Kennedy A, Wiggam MI, McCance DR, Sheridan B. Long-term remission rates after pituitary surgery for Cushing’s disease: the need for long-term surveillance. Clin Endocrinol (Oxf) [Internet]. 2005;63(5):549–59. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16268808
  114. Devoe DJ, Miller WL, Conte FA, Kaplan SL, Grumbach MM, Rosenthal SM, et al. Long-term outcome in children and adolescents after transsphenoidal surgery for Cushing’s disease. J Clin Endocrinol Metab [Internet]. 1997;82(10):3196–202. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9329338
  115. Storr HL, Afshar F, Matson M, Sabin I, Davies KM, Evanson J, et al. Factors influencing cure by transsphenoidal selective adenomectomy in paediatric Cushing’s disease. Eur J Endocrinol [Internet]. 2005;152(6):825–33. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15941921
  116. Kane LA, Leinung MC, Scheithauer BW, Bergstralh EJ, Laws Jr. ER, Groover R v, et al. Pituitary adenomas in childhood and adolescence. J Clin Endocrinol Metab [Internet]. 1994;79(4):1135–40. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7525627
  117. Eugster EA, Pescovitz OH. Gigantism. J Clin Endocrinol Metab [Internet]. 1999;84(12):4379–84. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10599691
  118. van der Lely AJ, Biller BM, Brue T, Buchfelder M, Ghigo E, Gomez R, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab [Internet]. 2012;97(5):1589–97. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22362824
  119. Eugster E. Gigantism. In: de Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, et al., editors. Endotext [Internet]. South Dartmouth (MA); 2000. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25905378
  120. Surawicz TS, McCarthy BJ, Kupelian V, Jukich PJ, Bruner JM, Davis FG. Descriptive epidemiology of primary brain and CNS tumors: results from the Central Brain Tumor Registry of the United States, 1990-1994. Neuro Oncol [Internet]. 2001/09/14. 1999;1(1):14–25. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11554386
  121. Murray MJ, Horan G, Lowis S, Nicholson JC. Highlights from the Third International Central Nervous System Germ Cell Tumour symposium: laying the foundations for future consensus. Ecancermedicalscience [Internet]. 2013/07/19. 2013;7:333. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23861728
  122. Maity A, Shu HK, Janss A, Belasco JB, Rorke L, Phillips PC, et al. Craniospinal radiation in the treatment of biopsy-proven intracranial germinomas: twenty-five years’ experience in a single center. Int J Radiat Oncol Biol Phys [Internet]. 2004/03/06. 2004;58(4):1165–70. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15001260
  123. Phi JH, Kim SK, Lee YA, Shin CH, Cheon JE, Kim IO, et al. Latency of intracranial germ cell tumors and diagnosis delay. Childs Nerv Syst [Internet]. 2013/07/03. 2013;29(10):1871–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23811803
  124. Sethi R v, Marino R, Niemierko A, Tarbell NJ, Yock TI, Macdonald SM. Delayed diagnosis in children with intracranial germ cell tumors. J Pediatr [Internet]. 2013/07/31. 2013;163(5):1448–53. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23896184
  125. Cancer Research UK . CancerStats: Childhood Cancer - Great Britain & UK. London: Cancer Research UK; 2010.
  126. Wang Y, Zou L, Gao B. Intracranial germinoma: clinical and MRI findings in 56 patients. Childs Nerv Syst [Internet]. 2010/07/29. 2010;26(12):1773–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20665036
  127. da Silva NS, Cappellano AM, Diez B, Cavalheiro S, Gardner S, Wisoff J, et al. Primary chemotherapy for intracranial germ cell tumors: results of the third international CNS germ cell tumor study. Pediatr Blood Cancer [Internet]. 2010/01/12. 2010;54(3):377–83. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20063410
  128. Calaminus G, Kortmann R, Worch J, Nicholson JC, Alapetite C, Garre ML, et al. SIOP CNS GCT 96: final report of outcome of a prospective, multinational nonrandomized trial for children and adults with intracranial germinoma, comparing craniospinal irradiation alone with chemotherapy followed by focal primary site irradiation for patients with localized disease. Neuro Oncol [Internet]. 2013/03/06. 2013;15(6):788–96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23460321
  129. O’Neil S, Ji L, Buranahirun C, Azoff J, Dhall G, Khatua S, et al. Neurocognitive outcomes in pediatric and adolescent patients with central nervous system germinoma treated with a strategy of chemotherapy followed by reduced-dose and volume irradiation. Pediatr Blood Cancer [Internet]. 2011/04/16. 2011;57(4):669–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21495164
  130. Maixner W. Hypothalamic hamartomas--clinical, neuropathological and surgical aspects. Childs Nerv Syst [Internet]. 2006/06/10. 2006;22(8):867–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16763856
  131. Brandberg G, Raininko R, Eeg-Olofsson O. Hypothalamic hamartoma with gelastic seizures in Swedish children and adolescents. Eur J Paediatr Neurol [Internet]. 2004/03/17. 2004;8(1):35–44. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15023373
  132. Ng YT, Kerrigan JF, Prenger EC, White WL, Rekate HL. Successful resection of a hypothalamic hamartoma and a Rathke cleft cyst. Case report. J Neurosurg [Internet]. 2005/10/07. 2005;102(1 Suppl):78–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16206738
  133. Weissenberger AA, Dell ML, Liow K, Theodore W, Frattali CM, Hernandez D, et al. Aggression and psychiatric comorbidity in children with hypothalamic hamartomas and their unaffected siblings. J Am Acad Child Adolesc Psychiatry [Internet]. 2001/06/08. 2001;40(6):696–703. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11392348
  134. Castano De La Mota C, Martin Del Valle F, Perez Villena A, Calleja Gero ML, Losada Del Pozo R, Ruiz-Falco Rojas ML. [Hypothalamic hamartoma in paediatric patients: clinical characteristics, outcomes and review of the literature]. Neurologia [Internet]. 2012/02/22. 2012;27(5):268–76. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22341983
  135. Papayannis CE, Consalvo D, Seifer G, Kauffman MA, Silva W, Kochen S. Clinical spectrum and difficulties in management of hypothalamic hamartoma in a developing country. Acta Neurol Scand [Internet]. 2008/05/09. 2008;118(5):313–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18462479
  136. Tassinari C, Riguzzi P, Rizzi R. Gelastic seizures. In: Tuxhom I, Holthausen H, Boenigk K, editors. Paediatric Epilepsy Syndromes and Their Surgical Management. London: John Libbey; 1997. p. 429–46.
  137. Graham Jr. JM, Saunders R, Fratkin J, Spiegel P, Harris M, Klein RZ. A cluster of Pallister-Hall syndrome cases, (congenital hypothalamic hamartoblastoma syndrome). Am J Med Genet Suppl [Internet]. 1986/01/01. 1986;2:53–63. Available from: http://www.ncbi.nlm.nih.gov/pubmed/3146300
  138. Kelberman D, Rizzoti K, Avilion A, Bitner-Glindzicz M, Cianfarani S, Collins J, et al. Mutations within Sox2/SOX2 are associated with abnormalities in the hypothalamo-pituitary-gonadal axis in mice and humans. J Clin Invest [Internet]. 2006;116(9):2442–55. Available from: https://www.ncbi.nlm.nih.gov/pubmed/16932809
  139. Wu J, Xu L, Kim DY, Rho JM, St John PA, Lue LF, et al. Electrophysiological properties of human hypothalamic hamartomas. Ann Neurol [Internet]. 2005/09/01. 2005;58(3):371–82. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16130091
  140. Munari C, Kahane P, Francione S, Hoffmann D, Tassi L, Cusmai R, et al. Role of the hypothalamic hamartoma in the genesis of gelastic fits (a video-stereo-EEG study). Electroencephalogr Clin Neurophysiol [Internet]. 1995/09/01. 1995;95(3):154–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7555906
  141. Wethe J v, Prigatano GP, Gray J, Chapple K, Rekate HL, Kerrigan JF. Cognitive functioning before and after surgical resection for hypothalamic hamartoma and epilepsy. Neurology [Internet]. 2013/08/16. 2013;81(12):1044–50. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23946307
  142. Mittal S, Mittal M, Montes JL, Farmer JP, Andermann F. Hypothalamic hamartomas. Part 2. Surgical considerations and outcome. Neurosurg Focus [Internet]. 2013/06/04. 2013;34(6):E7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23724841
  143. Kerrigan JF, Ng YT, Chung S, Rekate HL. The hypothalamic hamartoma: a model of subcortical epileptogenesis and encephalopathy. Semin Pediatr Neurol [Internet]. 2005/08/24. 2005;12(2):119–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16114178
  144. Li CD, Luo SQ, Gong J, Ma ZY, Jia G, Zhang YQ, et al. Surgical treatment of hypothalamic hamartoma causing central precocious puberty: long-term follow-up. J Neurosurg Pediatr [Internet]. 2013/06/12. 2013;12(2):151–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23746126
  145. Freeman JL, Zacharin M, Rosenfeld J v, Harvey AS. The endocrinology of hypothalamic hamartoma surgery for intractable epilepsy. Epileptic Disord [Internet]. 2004/02/21. 2003;5(4):239–47. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14975793
  146. Abla AA, Wait SD, Forbes JA, Pati S, Johnsonbaugh RE, Kerrigan JF, et al. Syndrome of alternating hypernatremia and hyponatremia after hypothalamic hamartoma surgery. Neurosurg Focus [Internet]. 2011/02/03. 2011;30(2):E6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21284452
  147. Drees C, Chapman K, Prenger E, Baxter L, Maganti R, Rekate H, et al. Seizure outcome and complications following hypothalamic hamartoma treatment in adults: endoscopic, open, and Gamma Knife procedures. J Neurosurg [Internet]. 2012/06/12. 2012;117(2):255–61. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22680243
  148. Burrows AM, Marsh WR, Worrell G, Woodrum DA, Pollock BE, Gorny KR, et al. Magnetic resonance imaging-guided laser interstitial thermal therapy for previously treated hypothalamic hamartomas. Neurosurg Focus. 2016;41(4):E8.
  149. Du VX, Gandhi S v, Rekate HL, Mehta AD. Laser interstitial thermal therapy: a first line treatment for seizures due to hypothalamic hamartoma? Epilepsia. 2017;58(Suppl 2):77–84.
  150. Henter JI, Tondini C, Pritchard J. Histiocyte disorders. Crit Rev Oncol Hematol [Internet]. 2004/05/26. 2004;50(2):157–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15157664
  151. Alston RD, Tatevossian RG, McNally RJ, Kelsey A, Birch JM, Eden TO. Incidence and survival of childhood Langerhans cell histiocytosis in Northwest England from 1954 to 1998. Pediatr Blood Cancer [Internet]. 2006/05/03. 2007;48(5):555–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16652350
  152. Guyot-Goubin A, Donadieu J, Barkaoui M, Bellec S, Thomas C, Clavel J. Descriptive epidemiology of childhood Langerhans cell histiocytosis in France, 2000-2004. Pediatr Blood Cancer [Internet]. 2008/02/09. 2008;51(1):71–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18260117
  153. Salotti JA, Nanduri V, Pearce MS, Parker L, Lynn R, Windebank KP. Incidence and clinical features of Langerhans cell histiocytosis in the UK and Ireland. Arch Dis Child [Internet]. 2008/12/09. 2009;94(5):376–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19060008
  154. Stalemark H, Laurencikas E, Karis J, Gavhed D, Fadeel B, Henter JI. Incidence of Langerhans cell histiocytosis in children: a population-based study. Pediatr Blood Cancer [Internet]. 2008/02/13. 2008;51(1):76–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18266220
  155. Abla O, Egeler RM, Weitzman S. Langerhans cell histiocytosis: Current concepts and treatments. Cancer Treat Rev [Internet]. 2010/03/02. 2010;36(4):354–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20188480
  156. Kim BE, Koh KN, Suh JK, Im HJ, Song JS, Lee JW, et al. Clinical Features and Treatment Outcomes of Langerhans Cell Histiocytosis: A Nationwide Survey From Korea Histiocytosis Working Party. J Pediatr Hematol Oncol [Internet]. 2013/11/28. 2013; Available from: http://www.ncbi.nlm.nih.gov/pubmed/24276037
  157. Badalian-Very G, Vergilio JA, Degar BA, MacConaill LE, Brandner B, Calicchio ML, et al. Recurrent BRAF mutations in Langerhans cell histiocytosis. Blood [Internet]. 2010;116(11):1919–23. Available from: https://www.ncbi.nlm.nih.gov/pubmed/20519626
  158. Donadieu J, Rolon MA, Thomas C, Brugieres L, Plantaz D, Emile JF, et al. Endocrine involvement in pediatric-onset Langerhans’ cell histiocytosis: a population-based study. J Pediatr [Internet]. 2004/03/06. 2004;144(3):344–50. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15001940
  159. Grois N, Potschger U, Prosch H, Minkov M, Arico M, Braier J, et al. Risk factors for diabetes insipidus in langerhans cell histiocytosis. Pediatr Blood Cancer [Internet]. 2005/07/28. 2006;46(2):228–33. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16047354
  160. Varan A, Atas E, Aydin B, Yalcin B, Akyuz C, Kutluk T, et al. Evaluation of patients with intracranial tumors and central diabetes insipidus. Pediatr Hematol Oncol [Internet]. 2013/08/31. 2013;30(7):668–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23988090
  161. Haupt R, Nanduri V, Calevo MG, Bernstrand C, Braier JL, Broadbent V, et al. Permanent consequences in Langerhans cell histiocytosis patients: a pilot study from the Histiocyte Society-Late Effects Study Group. Pediatr Blood Cancer [Internet]. 2004/03/30. 2004;42(5):438–44. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15049016
  162. Arico M. Langerhans cell histiocytosis in children: from the bench to bedside for an updated therapy. Br J Haematol [Internet]. 2016; Available from: http://www.ncbi.nlm.nih.gov/pubmed/26913480
  163. Braier J, Rosso D, Pollono D, Rey G, Lagomarsino E, Latella A, et al. Symptomatic bone langerhans cell histiocytosis treated at diagnosis or after reactivation with indomethacin alone. J Pediatr Hematol Oncol [Internet]. 2014;36(5):e280-4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24977402
  164. Bernard F, Thomas C, Bertrand Y, Munzer M, Landman Parker J, Ouache M, et al. Multi-centre pilot study of 2-chlorodeoxyadenosine and cytosine arabinoside combined chemotherapy in refractory Langerhans cell histiocytosis with haematological dysfunction. Eur J Cancer [Internet]. 2005;41(17):2682–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16291085
  165. Heritier S, Jehanne M, Leverger G, Emile JF, Alvarez JC, Haroche J, et al. Vemurafenib Use in an Infant for High-Risk Langerhans Cell Histiocytosis. JAMA Oncol [Internet]. 2015;1(6):836–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26180941
  166. Simko SJ, McClain KL, Allen CE. Up-front therapy for LCH: is it time to test an alternative to vinblastine/prednisone? Br J Haematol [Internet]. 2015;169(2):299–301. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25400231
  167. Simko SJ, Tran HD, Jones J, Bilgi M, Beaupin LK, Coulter D, et al. Clofarabine salvage therapy in refractory multifocal histiocytic disorders, including Langerhans cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman disease. Pediatr Blood Cancer [Internet]. 2014;61(3):479–87. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24106153
  168. Veys PA, Nanduri V, Baker KS, He W, Bandini G, Biondi A, et al. Haematopoietic stem cell transplantation for refractory Langerhans cell histiocytosis: outcome by intensity of conditioning. Br J Haematol [Internet]. 2015;169(5):711–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25817915
  169. Grois N, Fahrner B, Arceci RJ, Henter JI, McClain K, Lassmann H, et al. Central nervous system disease in Langerhans cell histiocytosis. J Pediatr [Internet]. 2010/05/04. 2010;156(6):873–81, 881 e1. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20434166
  170. Abla O, Weitzman S, Minkov M, McClain KL, Visser J, Filipovich A, et al. Diabetes insipidus in Langerhans cell histiocytosis: When is treatment indicated? Pediatr Blood Cancer [Internet]. 2009/01/15. 2009;52(5):555–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19142995
  171. Gadner H, Minkov M, Grois N, Potschger U, Thiem E, Arico M, et al. Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis. Blood [Internet]. 2013;121(25):5006–14. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23589673
  172. Satogami N, Miki Y, Koyama T, Kataoka M, Togashi K. Normal pituitary stalk: high-resolution MR imaging at 3T. AJNR Am J Neuroradiol [Internet]. 2009/10/03. 2010;31(2):355–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19797792
  173. Simmons GE, Suchnicki JE, Rak KM, Damiano TR. MR imaging of the pituitary stalk: size, shape, and enhancement pattern. AJR Am J Roentgenol [Internet]. 1992/08/01. 1992;159(2):375–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1632360
  174. Raybaud C, Barkovich AJ. Intracranial, orbital and neck masses of childhood. In: Barkovich AJ, Raybaud C, editors. Pediatric Neuroimaging. Philadelphia: Wolters Kluwer Health/ Lippincott Wiliams & Wilkins; 2012. p. 714–5.
  175. Varan A, Cila A, Akyuz C, Kale G, Kutluk T, Buyukpamukcu M. Radiological evaluation of patients with pituitary langerhans cell histiocytosis at diagnosis and at follow-up. Pediatr Hematol Oncol [Internet]. 2008/08/30. 2008;25(6):567–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18728976
  176. Hamilton BE, Salzman KL, Osborn AG. Anatomic and pathologic spectrum of pituitary infundibulum lesions. AJR Am J Roentgenol [Internet]. 2007/02/22. 2007;188(3):W223-32. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17312027
  177. Jinguji S, Nishiyama K, Yoshimura J, Yoneoka Y, Harada A, Sano M, et al. Endoscopic biopsies of lesions associated with a thickened pituitary stalk. Acta Neurochir (Wien) [Internet]. 2012/10/31. 2013;155(1):119–24; discussion 124. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23108562
  178. Robison NJ, Prabhu SP, Sun P, Chi SN, Kieran MW, Manley PE, et al. Predictors of neoplastic disease in children with isolated pituitary stalk thickening. Pediatr Blood Cancer [Internet]. 2013/05/15. 2013;60(10):1630–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23670879
  179. Beni-Adani L, Sainte-Rose C, Zerah M, Brunelle F, Constantini S, Renier D, et al. Surgical implications of the thickened pituitary stalk accompanied by central diabetes insipidus. J Neurosurg [Internet]. 2005/12/24. 2005;103(2 Suppl):142–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16370280
  180. Biller BM, Colao A, Petersenn S, Bonert VS, Boscaro M. Prolactinomas, Cushing’s disease and acromegaly: debating the role of medical therapy for secretory pituitary adenomas. BMC Endocr Disord [Internet]. 2010/05/19. 2010;10:10. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20478050
  181. Mootha SL, Barkovich AJ, Grumbach MM, Edwards MS, Gitelman SE, Kaplan SL, et al. Idiopathic hypothalamic diabetes insipidus, pituitary stalk thickening, and the occult intracranial germinoma in children and adolescents. J Clin Endocrinol Metab [Internet]. 1997/05/01. 1997;82(5):1362–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9141516
  182. Mikami-Terao Y, Akiyama M, Yanagisawa T, Takahashi-Fujigasaki J, Yokoi K, Fukuoka K, et al. Lymphocytic hypophysitis with central diabetes insipidus and subsequent hypopituitarism masking a suprasellar germinoma in a 13-year-old girl. Childs Nerv Syst [Internet]. 2006;22(10):1338–43. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16565852
  183. Nishiuchi T, Imachi H, Murao K, Fujiwara M, Sato M, Nishiuchi Y, et al. Suprasellar germinoma masquerading as lymphocytic hypophysitis associated with central diabetes insipidus, delayed sexual development, and subsequent hypopituitarism. Am J Med Sci [Internet]. 2010/01/07. 2010;339(2):195–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20051818
  184. Turcu AF, Erickson BJ, Lin E, Guadalix S, Schwartz K, Scheithauer BW, et al. Pituitary stalk lesions: the Mayo Clinic experience. J Clin Endocrinol Metab [Internet]. 2013/03/28. 2013;98(5):1812–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23533231
  185. di Iorgi N, Napoli F, Allegri AE, Olivieri I, Bertelli E, Gallizia A, et al. Diabetes insipidus--diagnosis and management. Horm Res Paediatr [Internet]. 2012/03/22. 2012;77(2):69–84. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22433947
  186. Cerbone M, Visser J, Bulwer C, Ederies A, Vallabhaneni K, Ball S, et al. Management of children and young people with idiopathic pituitary stalk thickening, central diabetes insipidus, or both: a national clinical practice consensus guideline. Lancet Child Adolesc Health. 2021;5(9):662–76.
  187. Aquilina K, Boop FA. Nonneoplastic enlargement of the pituitary gland in children. J Neurosurg Pediatr [Internet]. 2011/05/03. 2011;7(5):510–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21529191
  188. Elster AD, Chen MY, Williams 3rd DW, Key LL. Pituitary gland: MR imaging of physiologic hypertrophy in adolescence. Radiology [Internet]. 1990/03/01. 1990;174(3 Pt 1):681–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/2305049
  189. Alatzoglou KS, Kelberman D, Cowell CT, Palmer R, Arnhold IJ, Melo ME, et al. Increased transactivation associated with SOX3 polyalanine tract deletion in a patient with hypopituitarism. J Clin Endocrinol Metab [Internet]. 2011;96(4):E685-90. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21289259
  190. Gan HW, Alatzoglou KS, Dattani MT. Disorders of Hypothalamo-pituitary Axis. In: Dattani MT, Brook CGD, editors. Brook’s Clinical Pediatric Endocrinology. 7th ed. Oxford, UK: John Wiley & Sons Ltd; 2020. p. 133–98.
  191. Teramoto A, Hirakawa K, Sanno N, Osamura Y. Incidental pituitary lesions in 1,000 unselected autopsy specimens. Radiology [Internet]. 1994/10/01. 1994;193(1):161–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8090885
  192. Han SJ, Rolston JD, Jahangiri A, Aghi MK. Rathke’s cleft cysts: review of natural history and surgical outcomes. J Neurooncol [Internet]. 2013/10/23. 2013; Available from: http://www.ncbi.nlm.nih.gov/pubmed/24146189
  193. Dubuisson AS, Stevenaert A, Martin DH, Flandroy PP. Intrasellar arachnoid cysts. Neurosurgery [Internet]. 2007;61(3):505–13; discussion 513. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17881962
  194. Ali ZS, Lang SS, Bakar D, Storm PB, Stein SC. Pediatric intracranial arachnoid cysts: comparative effectiveness of surgical treatment options. Childs Nerv Syst [Internet]. 2014;30(3):461–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24162618
  195. Ozek MM, Urgun K. Neuroendoscopic management of suprasellar arachnoid cysts. World Neurosurg [Internet]. 2013;79(2 Suppl):S19 e13-8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22381821
  196. El-Ghandour NM. Endoscopic treatment of suprasellar arachnoid cysts in children. J Neurosurg Pediatr [Internet]. 2011;8(1):6–14. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21721882
  197. Adan L, Bussieres L, Dinand V, Zerah M, Pierre-Kahn A, Brauner R. Growth, puberty and hypothalamic-pituitary function in children with suprasellar arachnoid cyst. Eur J Pediatr [Internet]. 2000;159(5):348–55. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10834520
  198. McCrea HJ, George E, Settler A, Schwartz TH, Greenfield JP. Pediatric Suprasellar Tumors. J Child Neurol [Internet]. 2015; Available from: http://www.ncbi.nlm.nih.gov/pubmed/26676303
  199. Ogiwara H, Morota N, Joko M, Hirota K. Endoscopic fenestrations for suprasellar arachnoid cysts. J Neurosurg Pediatr [Internet]. 2011;8(5):484–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22044374
  200. Howlett TA, Levy MJ, Robertson IJ. How reliably can autoimmune hypophysitis be diagnosed without pituitary biopsy. Clin Endocrinol (Oxf) [Internet]. 2009/12/31. 2010;73(1):18–21. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20039888
  201. Smith JK, Matheus MG, Castillo M. Imaging manifestations of neurosarcoidosis. AJR Am J Roentgenol [Internet]. 2004/01/23. 2004;182(2):289–95. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14736648
  202. Wilne S, Collier J, Kennedy C, Jenkins A, Grout J, Mackie S, et al. Progression from first symptom to diagnosis in childhood brain tumours. Eur J Pediatr [Internet]. 2011/05/20. 2012;171(1):87–93. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21594769
  203. Wilne S, Collier J, Kennedy C, Koller K, Grundy R, Walker D. Presentation of childhood CNS tumours: a systematic review and meta-analysis. Lancet Oncol [Internet]. 2007/07/24. 2007;8(8):685–95. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17644483
  204. Royal College of Ophthalmologists . Guidelines for the management of strabismus in childhood. London: Royal College of Ophthalmologists; 2012.
  205. Hawley DP, Walker DA. A symptomatic journey to the centre of the brain. Arch Dis Child Educ Pract Ed [Internet]. 2010/03/31. 2010;95(2):59–64. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20351153
  206. Aquilina K, Daniels DJ, Spoudeas H, Phipps K, Gan HW, Boop FA. Optic pathway glioma in children: does visual deficit correlate with radiology in focal exophytic lesions? Childs Nerv Syst [Internet]. 2015;31(11):2041–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26277358
  207. Chateil JF, Soussotte C, Pedespan JM, Brun M, le Manh C, Diard F. MRI and clinical differences between optic pathway tumours in children with and without neurofibromatosis. Br J Radiol [Internet]. 2001/03/03. 2001;74(877):24–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11227773
  208. Grill J, Laithier V, Rodriguez D, Raquin MA, Pierre-Kahn A, Kalifa C. When do children with optic pathway tumours need treatment? An oncological perspective in 106 patients treated in a single centre. Eur J Pediatr [Internet]. 2000/10/03. 2000;159(9):692–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11014471
  209. Taylor M, Couto-Silva AC, Adan L, Trivin C, Sainte-Rose C, Zerah M, et al. Hypothalamic-pituitary lesions in pediatric patients: endocrine symptoms often precede neuro-ophthalmic presenting symptoms. J Pediatr [Internet]. 2012/06/26. 2012;161(5):855–63. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22727865
  210. Rodriguez LA, Edwards MS, Levin VA. Management of hypothalamic gliomas in children: an analysis of 33 cases. Neurosurgery [Internet]. 1990/02/01. 1990;26(2):242–6; discussion 246-7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/2308672
  211. Virdis R, Sigorini M, Laiolo A, Lorenzetti E, Street ME, Villani AR, et al. Neurofibromatosis type 1 and precocious puberty. J Pediatr Endocrinol Metab [Internet]. 2000/09/02. 2000;13 Suppl 1:841–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10969931
  212. Ahn Y, Cho BK, Kim SK, Chung YN, Lee CS, Kim IH, et al. Optic pathway glioma: outcome and prognostic factors in a surgical series. Childs Nerv Syst [Internet]. 2006/04/22. 2006;22(9):1136–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16628460
  213. Cappelli C, Grill J, Raquin M, Pierre-Kahn A, Lellouch-Tubiana A, Terrier-Lacombe MJ, et al. Long-term follow up of 69 patients treated for optic pathway tumours before the chemotherapy era. Arch Dis Child [Internet]. 1999/01/06. 1998;79(4):334–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9875044
  214. Muller HL, Kaatsch P, Warmuth-Metz M, Flentje M, Sorensen N. Kraniopharyngeom im Kindes-und Jugendalter: Diagnostische und therapeutische Strategien (Childhood craniopharyngioma - diagnostic and therapeutic strategies). Monatsschrift Kindheilkunde. 2003;151:1056–63.
  215. Cisternino M, Arrigo T, Pasquino AM, Tinelli C, Antoniazzi F, Beduschi L, et al. Etiology and age incidence of precocious puberty in girls: a multicentric study. J Pediatr Endocrinol Metab [Internet]. 2000/09/02. 2000;13 Suppl 1:695–701. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10969911
  216. Faizah M, Zuhanis A, Rahmah R, Raja A, Wu L, Dayang A, et al. Precocious puberty in children: A review of imaging findings. Biomed Imaging Interv J [Internet]. 2012/09/13. 2012;8(1):e6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22970062
  217. Mogensen SS, Aksglaede L, Mouritsen A, Sorensen K, Main KM, Gideon P, et al. Diagnostic work-up of 449 consecutive girls who were referred to be evaluated for precocious puberty. J Clin Endocrinol Metab [Internet]. 2011/02/25. 2011;96(5):1393–401. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21346077
  218. Russell A. A diencephalic syndrome of emaciation in infancy and childhood. Arch Dis Child. 1951;26(127):270–5.
  219. Waga S, Shimizu T, Sakakura M. Diencephalic syndrome of emaciation (Russell’s syndrome). Surg Neurol [Internet]. 1982/02/01. 1982;17(2):141–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/6803375
  220. Burr IM, Slonim AE, Danish RK, Gadoth N, Butler IJ. Diencephalic syndrome revisited. J Pediatr [Internet]. 1976/03/01. 1976;88(3):439–44. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1245953
  221. Mohan SM, Dharmalingam M, Prasanna Kumar KM, Verma RG, Balaji Pai S, Krishna KN, et al. Suprasellar germ cell tumor presenting as diencephalic syndrome and precocious puberty. J Pediatr Endocrinol Metab [Internet]. 2003/04/23. 2003;16(3):443–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12705371
  222. Chipkevitch E, Fernandes AC. Hypothalamic tumor associated with atypical forms of anorexia nervosa and diencephalic syndrome. Arq Neuropsiquiatr [Internet]. 1993/06/01. 1993;51(2):270–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8274094
  223. Addy DP, Hudson FP. Diencephalic syndrome of infantile emaciation. Analysis of literature and report of further 3 cases. Arch Dis Child [Internet]. 1972/06/01. 1972;47(253):338–43. Available from: http://www.ncbi.nlm.nih.gov/pubmed/5034666
  224. Sharma RR, Chandy MJ, Lad SD. Diencephalic syndrome of emaciation in an adult associated with a suprasellar craniopharyngioma--a case report. Br J Neurosurg [Internet]. 1990/01/01. 1990;4(1):77–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/2334532
  225. Eliash A, Roitman A, Karp M, Reichental E, Manor RS, Shalit M, et al. Diencephalic syndrome due to a suprasellar epidermoid cyst. Case report. Childs Brain [Internet]. 1983/01/01. 1983;10(6):414–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/6661939
  226. Maroon JC, Albright L. “Failure to thrive” due to pontine glioma. Arch Neurol [Internet]. 1977/05/01. 1977;34(5):295–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/67836
  227. Ramos EJ, Suzuki S, Marks D, Inui A, Asakawa A, Meguid MM. Cancer anorexia-cachexia syndrome: cytokines and neuropeptides. Curr Opin Clin Nutr Metab Care [Internet]. 2004/06/12. 2004;7(4):427–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15192446
  228. DeSousa AL, Kalsbeck JE, Mealey Jr. J, Fitzgerald J. Diencephalic syndrome and its relation to opticochiasmatic glioma: review of twelve cases. Neurosurgery [Internet]. 1979/03/01. 1979;4(3):207–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/460550
  229. Miyoshi Y, Yunoki M, Yano A, Nishimoto K. Diencephalic syndrome of emaciation in an adult associated with a third ventricle intrinsic craniopharyngioma: case report. Neurosurgery [Internet]. 2002/12/21. 2003;52(1):224–7; discussion 227. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12493122
  230. Hager A, Thorell JI. Studies on growth hormone secretion in a patient with the diencephalic syndrome of emaciation. Acta Paediatr Scand [Internet]. 1973;62(3):231–40. Available from: https://www.ncbi.nlm.nih.gov/pubmed/4703018
  231. Pimstone BL, Sobel J, Meyer E, Eale D. Secretion of growth hormone in the diencephalic syndrome of childhood. J Pediatr [Internet]. 1970/06/01. 1970;76(6):886–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/5444580
  232. Kilday JP, Bartels U, Huang A, Barron M, Shago M, Mistry M, et al. Favorable survival and metabolic outcome for children with diencephalic syndrome using a radiation-sparing approach. J Neurooncol [Internet]. 2013/11/13. 2014;116(1):195–204. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24218181
  233. Vlachopapadopoulou E, Tracey KJ, Capella M, Gilker C, Matthews DE. Increased energy expenditure in a patient with diencephalic syndrome. J Pediatr [Internet]. 1993/06/01. 1993;122(6):922–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8501572
  234. Chipkevitch E. Brain tumors and anorexia nervosa syndrome. Brain Dev [Internet]. 1994/05/01. 1994;16(3):175–9, discussion 180-2. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7943600
  235. de Vile CJ, Sufraz R, Lask BD, Stanhope R. Occult intracranial tumours masquerading as early onset anorexia nervosa. BMJ [Internet]. 1995/11/18. 1995;311(7016):1359–60. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7496292
  236. Houy E, Debono B, Dechelotte P, Thibaut F. Anorexia nervosa associated with right frontal brain lesion. Int J Eat Disord [Internet]. 2007/08/09. 2007;40(8):758–61. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17683096
  237. American Psychiatric Association . Diagnostic and Statistic Manual of Mental Disorders (DSM-5). 5th ed. Arlington, VA, USA: American Psychiatric Publishing; 2013.
  238. World Health Organisation . The ICD-10 Classification of Mental and Behavioural Disorders: Clinical descriptions and diagnostic guidelines. 10th ed. Geneva, Switzerland: World Health Organisation; 1992.
  239. Diamanti A, Ubertini GM, Basso MS, Caramadre AM, Alterio A, Panetta F, et al. Amenorrhea and weight loss: not only anorexia nervosa. Eur J Obstet Gynecol Reprod Biol [Internet]. 2011/12/27. 2012;161(1):111–2. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22197307
  240. de Vile CJ, Grant DB, Hayward RD, Kendall BE, Neville BG, Stanhope R. Obesity in childhood craniopharyngioma: relation to post-operative hypothalamic damage shown by magnetic resonance imaging. J Clin Endocrinol Metab [Internet]. 1996/07/01. 1996;81(7):2734–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8675604
  241. Webb EA, Dattani MT. Septo-optic dysplasia. Eur J Hum Genet [Internet]. 2010;18(4):393–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19623216
  242. Littley MD, Shalet SM, Beardwell CG, Robinson EL, Sutton ML. Radiation-induced hypopituitarism is dose-dependent. Clin Endocrinol (Oxf) [Internet]. 1989/09/01. 1989;31(3):363–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/2559824
  243. Adan L, Trivin C, Sainte-Rose C, Zucker JM, Hartmann O, Brauner R. GH deficiency caused by cranial irradiation during childhood: factors and markers in young adults. J Clin Endocrinol Metab [Internet]. 2001/11/10. 2001;86(11):5245–51. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11701685
  244. Talbot L, Spoudeas H. Late effects in relation to childhood cancer. In: Estlin EJ, Gilbertson RJ, Wynn RF, editors. Pediatric Hematology and Oncology: Scientific Principles & Clinical Practice. Oxford: Wiley-Blackwell; 2010. p. 367–91.
  245. Collet-Solberg PF, Sernyak H, Satin-Smith M, Katz LL, Sutton L, Molloy P, et al. Endocrine outcome in long-term survivors of low-grade hypothalamic/chiasmatic glioma. Clin Endocrinol (Oxf) [Internet]. 1997/07/01. 1997;47(1):79–85. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9302376
  246. Grabenbauer GG, Schuchardt U, Buchfelder M, Rodel CM, Gusek G, Marx M, et al. Radiation therapy of optico-hypothalamic gliomas (OHG)--radiographic response, vision and late toxicity. Radiother Oncol [Internet]. 2000/03/30. 2000;54(3):239–45. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10738082
  247. Nanduri VR, Bareille P, Pritchard J, Stanhope R. Growth and endocrine disorders in multisystem Langerhans’ cell histiocytosis. Clin Endocrinol (Oxf) [Internet]. 2000;53(4):509–15. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11012577
  248. Huguenin M, Trivin C, Zerah M, Doz F, Brugieres L, Brauner R. Adult height after cranial irradiation for optic pathway tumors: relationship with neurofibromatosis. J Pediatr [Internet]. 2003/07/03. 2003;142(6):699–703. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12838200
  249. Sklar CA, Antal Z, Chemaitilly W, Cohen LE, Follin C, Meacham LR, et al. Hypothalamic-Pituitary and Growth Disorders in Survivors of Childhood Cancer: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab [Internet]. 2018;103(8):2761–84. Available from: https://www.ncbi.nlm.nih.gov/pubmed/29982476
  250. Hindmarsh PC, Swift PG. An assessment of growth hormone provocation tests. Arch Dis Child [Internet]. 1995/04/01. 1995;72(4):362–7; discussion 367-8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7539245
  251. Shah A, Stanhope R, Matthew D. Hazards of pharmacological tests of growth hormone secretion in childhood. BMJ [Internet]. 1992/01/18. 1992;304(6820):173–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1737165
  252. Sfeir JG, Kittah NEN, Tamhane SU, Jasim S, Chemaitilly W, Cohen LE, et al. Diagnosis of GH deficiency as a late effect of radiotherapy in survivors of childhood cancers. J Clin Endocrinol Metab. 2018;103(8):2785–93.
  253. Cattoni A, Clarke E, Albanese A. The predictive value of insulin-like growth factor 1 in irradiation-dependent growth hormone deficiency in childhood cancer survivors. Horm Res Paediatr. 2018;90(5):314–25.
  254. Sklar C, Sarafoglou K, Whittam E. Efficacy of insulin-like growth factor binding protein 3 in predicting the growth hormone response to provocative testing in children treated with cranial irradiation. Acta Endocrinol (Copenh). 1993;129(6):511–5.
  255. Murray PG, Dattani MT, Clayton PE. Controversies in the diagnosis and management of growth hormone deficiency in childhood and adolescence. Arch Dis Child [Internet]. 2016;101(1):96–100. Available from: https://www.ncbi.nlm.nih.gov/pubmed/26153506
  256. Growth Hormone Research S. Consensus guidelines for the diagnosis and treatment of growth hormone (GH) deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society. J Clin Endocrinol Metab [Internet]. 2000;85(11):3990–3. Available from: https://www.ncbi.nlm.nih.gov/pubmed/11095419
  257. Phillip M, Moran O, Lazar L. Growth without growth hormone. J Pediatr Endocrinol Metab [Internet]. 2003/01/04. 2002;15 Suppl 5:1267–72. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12510977
  258. Moshang Jr. T, Rundle AC, Graves DA, Nickas J, Johanson A, Meadows A. Brain tumor recurrence in children treated with growth hormone: the National Cooperative Growth Study experience. J Pediatr [Internet]. 1996;128(5 Pt 2):S4-7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8627468
  259. Muller HL, Gebhardt U, Schroder S, Pohl F, Kortmann RD, Faldum A, et al. Analyses of treatment variables for patients with childhood craniopharyngioma--results of the multicenter prospective trial KRANIOPHARYNGEOM 2000 after three years of follow-up. Horm Res Paediatr [Internet]. 2010/03/04. 2010;73(3):175–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20197669
  260. Karavitaki N, Warner JT, Marland A, Shine B, Ryan F, Arnold J, et al. GH replacement does not increase the risk of recurrence in patients with craniopharyngioma. Clin Endocrinol (Oxf) [Internet]. 2006;64(5):556–60. Available from: https://www.ncbi.nlm.nih.gov/pubmed/16649976
  261. Grimberg A, DiVall SA, Polychronakos C, Allen DB, Cohen LE, Quintos JB, et al. Guidelines for Growth Hormone and Insulin-Like Growth Factor-I Treatment in Children and Adolescents: Growth Hormone Deficiency, Idiopathic Short Stature, and Primary Insulin-Like Growth Factor-I Deficiency. Horm Res Paediatr [Internet]. 2016;86(6):361–97. Available from: https://www.ncbi.nlm.nih.gov/pubmed/27884013
  262. Lerner SE, Huang GJ, McMahon D, Sklar CA, Oberfield SE. Growth hormone therapy in children after cranial/craniospinal radiation therapy: sexually dimorphic outcomes. J Clin Endocrinol Metab [Internet]. 2004/12/08. 2004;89(12):6100–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15579765
  263. Carel JC. Management of short stature with GnRH agonist and co-treatment with growth hormone: a controversial issue. Mol Cell Endocrinol [Internet]. 2006/06/22. 2006;254–255:226–33. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16787697
  264. Brougham MF, Wallace WH. Subfertility in children and young people treated for solid and haematological malignancies. Br J Haematol [Internet]. 2005/10/04. 2005;131(2):143–55. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16197443
  265. Gan HW, Spoudeas HA. Preserving reproductive capacity in young boys with cancer. Trends Urol Men’s Health. 2013/5/23. 2013;4(3):8–12.
  266. Wallace WH, Kelsey TW. Ovarian reserve and reproductive age may be determined from measurement of ovarian volume by transvaginal sonography. Hum Reprod [Internet]. 2004/06/19. 2004;19(7):1612–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15205396
  267. Crowley S, Hindmarsh PC, Holownia P, Honour JW, Brook CG. The use of low doses of ACTH in the investigation of adrenal function in man. J Endocrinol [Internet]. 1991/09/01. 1991;130(3):475–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1940720
  268. Patterson BC, Truxillo L, Wasilewski-Masker K, Mertens AC, Meacham LR. Adrenal function testing in pediatric cancer survivors. Pediatr Blood Cancer [Internet]. 2009/07/29. 2009;53(7):1302–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19637328
  269. Schmiegelow M, Feldt-Rasmussen U, Rasmussen AK, Lange M, Poulsen HS, Muller J. Assessment of the hypothalamo-pituitary-adrenal axis in patients treated with radiotherapy and chemotherapy for childhood brain tumor. J Clin Endocrinol Metab [Internet]. 2003/07/05. 2003;88(7):3149–54. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12843158
  270. Persani L, Brabant G, Dattani M, Bonomi M, Feldt-Rasmussen U, Fliers E, et al. 2018 European Thyroid Association (ETA) Guidelines on the Diagnosis and Management of Central Hypothyroidism. Eur Thyroid J [Internet]. 2018;7(5):225–37. Available from: https://www.ncbi.nlm.nih.gov/pubmed/30374425
  271. Mehta A, Hindmarsh PC, Stanhope RG, Brain CE, Preece MA, Dattani MT. Is the thyrotropin-releasing hormone test necessary in the diagnosis of central hypothyroidism in children. J Clin Endocrinol Metab [Internet]. 2003/12/13. 2003;88(12):5696–703. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14671155
  272. Crofton PM, Tepper LA, Kelnar CJ. An evaluation of the thyrotrophin-releasing hormone stimulation test in paediatric clinical practice. Horm Res [Internet]. 2008;69(1):53–9. Available from: https://www.ncbi.nlm.nih.gov/pubmed/18059084
  273. Rodondi N, den Elzen WP, Bauer DC, Cappola AR, Razvi S, Walsh JP, et al. Subclinical hypothyroidism and the risk of coronary heart disease and mortality. JAMA [Internet]. 2010/09/23. 2010;304(12):1365–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20858880
  274. Karavitaki N, Thanabalasingham G, Shore HC, Trifanescu R, Ansorge O, Meston N, et al. Do the limits of serum prolactin in disconnection hyperprolactinaemia need re-definition? A study of 226 patients with histologically verified non-functioning pituitary macroadenoma. Clin Endocrinol (Oxf) [Internet]. 2006/09/21. 2006;65(4):524–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16984247
  275. Edate S, Albanese A. Management of electrolyte and fluid disorders after brain surgery for pituitary/suprasellar tumours. Horm Res Paediatr [Internet]. 2015;83(5):293–301. Available from: https://www.ncbi.nlm.nih.gov/pubmed/25677941
  276. Liu SY, Tung YC, Lee CT, Liu HM, Peng SF, Wu MZ, et al. Clinical characteristics of central diabetes insipidus in Taiwanese children. J Formos Med Assoc [Internet]. 2013;112(10):616–20. Available from: https://www.ncbi.nlm.nih.gov/pubmed/23916565
  277. Maghnie M, Villa A, Arico M, Larizza D, Pezzotta S, Beluffi G, et al. Correlation between magnetic resonance imaging of posterior pituitary and neurohypophyseal function in children with diabetes insipidus. J Clin Endocrinol Metab [Internet]. 1992;74(4):795–800. Available from: https://www.ncbi.nlm.nih.gov/pubmed/1548343
  278. Ghirardello S, Hopper N, Albanese A, Maghnie M. Diabetes insipidus in craniopharyngioma: postoperative management of water and electrolyte disorders. J Pediatr Endocrinol Metab [Internet]. 2006/05/17. 2006;19 Suppl 1:413–21. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16700319
  279. Finken MJ, Zwaveling-Soonawala N, Walenkamp MJ, Vulsma T, van Trotsenburg AS, Rotteveel J. Frequent occurrence of the triphasic response (diabetes insipidus/hyponatremia/diabetes insipidus) after surgery for craniopharyngioma in childhood. Horm Res Paediatr [Internet]. 2011;76(1):22–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21701131
  280. Pratheesh R, Swallow DM, Rajaratnam S, Jacob KS, Chacko G, Joseph M, et al. Incidence, predictors and early post-operative course of diabetes insipidus in paediatric craniopharygioma: a comparison with adults. Childs Nerv Syst [Internet]. 2013/02/07. 2013;29(6):941–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23386174
  281. Shimura N. Urinary arginine vasopressin (AVP) measurement in children: water deprivation test incorporating urinary AVP. Acta Paediatr Jpn [Internet]. 1993;35(4):320–4. Available from: https://www.ncbi.nlm.nih.gov/pubmed/8379325
  282. de Fost M, Oussaada SM, Endert E, Linthorst GE, Serlie MJ, Soeters MR, et al. The water deprivation test and a potential role for the arginine vasopressin precursor copeptin to differentiate diabetes insipidus from primary polydipsia. Endocr Connect [Internet]. 2015;4(2):86–91. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25712898
  283. Fenske W, Quinkler M, Lorenz D, Zopf K, Haagen U, Papassotiriou J, et al. Copeptin in the differential diagnosis of the polydipsia-polyuria syndrome--revisiting the direct and indirect water deprivation tests. J Clin Endocrinol Metab [Internet]. 2011;96(5):1506–15. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21367924
  284. Timper K, Fenske W, Kuhn F, Frech N, Arici B, Rutishauser J, et al. Diagnostic Accuracy of Copeptin in the Differential Diagnosis of the Polyuria-polydipsia Syndrome: A Prospective Multicenter Study. J Clin Endocrinol Metab [Internet]. 2015;100(6):2268–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25768671
  285. Babinski MJ. Tumeur du corps pituitaire san acromegalie et avec arret de developpement des organs genitaux. Rev Neurol (Paris). 1900;8:531–3.
  286. Steele CA, Cuthbertson DJ, MacFarlane IA, Javadpour M, Das KS, Gilkes C, et al. Hypothalamic obesity: prevalence, associations and longitudinal trends in weight in a specialist adult neuroendocrine clinic. Eur J Endocrinol [Internet]. 2013/01/08. 2013;168(4):501–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23293322
  287. Lustig RH. Hypothalamic obesity after craniopharyngiomas: mechanisms, diagnosis and treatment. Frontiers in Endocrinology. 2011;2:60.
  288. Pratheesh R, Rajaratnam S, Prabhu K, Mani SE, Chacko G, Chacko AG. The current role of transcranial surgery in the management of pituitary adenomas. Pituitary [Internet]. 2012/10/19. 2013;16(4):419–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23076713
  289. Lustig RH, Post SR, Srivannaboon K, Rose SR, Danish RK, Burghen GA, et al. Risk factors for the development of obesity in children surviving brain tumors. J Clin Endocrinol Metab [Internet]. 2003/02/08. 2003;88(2):611–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12574189
  290. Hamilton JK, Conwell LS, Syme C, Ahmet A, Jeffery A, Daneman D. Hypothalamic Obesity following Craniopharyngioma Surgery: Results of a Pilot Trial of Combined Diazoxide and Metformin Therapy. Int J Pediatr Endocrinol [Internet]. 2011/05/24. 2011;2011:417949. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21603206
  291. Harz KJ, Muller HL, Waldeck E, Pudel V, Roth C. Obesity in patients with craniopharyngioma: assessment of food intake and movement counts indicating physical activity. J Clin Endocrinol Metab [Internet]. 2003/11/07. 2003;88(11):5227–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14602754
  292. Lustig RH, Hinds PS, Ringwald-Smith K, Christensen RK, Kaste SC, Schreiber RE, et al. Octreotide therapy of pediatric hypothalamic obesity: a double-blind, placebo-controlled trial. J Clin Endocrinol Metab [Internet]. 2003/06/06. 2003;88(6):2586–92. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12788859
  293. Mason PW, Krawiecki N, Meacham LR. The use of dextroamphetamine to treat obesity and hyperphagia in children treated for craniopharyngioma. Arch Pediatr Adolesc Med [Internet]. 2002/08/29. 2002;156(9):887–92. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12197795
  294. Muller HL, Gebhardt U, Maroske J, Hanisch E. Long-term follow-up of morbidly obese patients with childhood craniopharyngioma after laparoscopic adjustable gastric banding (LAGB). Klin Padiatr [Internet]. 2011/11/05. 2011;223(6):372–3. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22052635
  295. Rakhshani N, Jeffery AS, Schulte F, Barrera M, Atenafu EG, Hamilton JK. Evaluation of a comprehensive care clinic model for children with brain tumor and risk for hypothalamic obesity. Obesity (Silver Spring) [Internet]. 2010/01/09. 2010;18(9):1768–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20057368
  296. Zoicas F, Droste M, Mayr B, Buchfelder M, Schofl C. GLP-1 analogues as a new treatment option for hypothalamic obesity in adults: report of nine cases. Eur J Endocrinol [Internet]. 2013;168(5):699–706. Available from: https://www.ncbi.nlm.nih.gov/pubmed/23392214
  297. Ando T, Haraguchi A, Matsunaga T, Natsuda S, Yamasaki H, Usa T, et al. Liraglutide as a potentially useful agent for regulating appetite in diabetic patients with hypothalamic hyperphagia and obesity. Intern Med [Internet]. 2014;53(16):1791–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25130112
  298. Lomenick JP, Buchowski MS, Shoemaker AH. A 52-week pilot study of the effects of exenatide on body weight in patients with hypothalamic obesity. Obesity (Silver Spring) [Internet]. 2016;24(6):1222–5. Available from: https://www.ncbi.nlm.nih.gov/pubmed/27133664
  299. Wilding JPH, Batterham RL, Calanna S, Davies M, van Gaal LF, Lingvay I, et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. New England Journal of Medicine. 2021 Mar 18;384(11):989–1002.
  300. Huynh K, Klose M, Krogsgaard K, Drejer J, Byberg S, Madsbad S, et al. Randomized controlled trial of Tesomet for weight loss in hypothalamic obesity. European Journal of Endocrinology. 2022 Jun 1;186(6):687–700.

 

Diabetes Insipidus

CLINICAL RECOGNITION

 

Diabetes Insipidus (DI) is the excess production of dilute urine. Diagnosis requires a targeted history, examination and confirmation through appropriate laboratory and radiological investigations. DI presents with polyuria and polydipsia. Urine output is more than 40 ml/kg /24 hours in adults and more than 100 ml/kg/24 hours in children. DI reflects either the lack of production or action of the posterior pituitary hormone vasopressin (AVP). There are three subtypes.

 

  • Cranial or hypothalamic DI (HDI): due to relative or absolute lack of AVP.
  • Nephrogenic DI (NDI): due to partial or total resistance to the renal antidiuretic effects of AVP.
  • Dipsogenic DI (DDI, primary polydipsia): where polyuria is secondary to excessive, inappropriate fluid intake.

 

All forms of DI are rare. HDI has an estimated prevalence of 1/25,000. While presentation is more common in adults, familial forms of both HDI and NDI characteristically present in childhood.

 

PATHOPHYSIOLOGY

 

The Physiology of AVP

 

AVP is a nine-amino acid peptide made within magnocellular neurones of the paraventricular (PVN) and supraoptic (SON) nuclei of the hypothalamus that project through the hypophyseal portal tract to terminate in the posterior pituitary, where AVP is released into the circulation. Together, the PVN, SON and posterior pituitary form an anatomical and functional unit- the neurohypophysis (Figure 1).

 

 

AVP is produced from a large precursor that undergoes extensive post-translational processing (figure 2).

 

The major action of AVP is in the regulation of renal water excretion. AVP increases expression of the AVP-dependent water channel Aquaporin 2, which is expressed in the renal collecting duct, facilitating water reabsorption. This action of AVP is mediated by the type 2 AVP receptor (V2-R), expressed exclusively on the interstitial surface of target cells in the distal nephron. AVP release is regulated by osmoreceptors within the lamina terminalis. There is a linear relationship between plasma osmolality and plasma AVP concentration. Not unexpectedly, thirst perception regulated in a parallel manner (Figure 3).

 

Hypothalamic DI

 

Presentation with HDI implies loss of 80%-90% of AVP production from the posterior pituitary. This, in turn, reflects either destruction of vasopressinergic magnocellular neurons in the hypothalamus or interruption of intra-axonal transport/processing of AVP. Some 50% of children and young adults with HDI have an underlying tumor or CNS malformation (e.g., craniopharyngioma, germinoma, septo-optic dysplasia). Familial HDI comprises 5% of cases. 

 

Acute HDI can occur in up to 22% of non-selected patients presenting with traumatic brain injury (TBI), persisting in some 30% of these on long term follow up. HDI may follow trauma to the pituitary or hypothalamus. HDI following surgery to the pituitary or neurohypophysis presents within 24-48h after surgery and is often transient, resolving within 10 days. Pituitary stalk trauma (including that following surgery) may lead to a tri-phasic disturbance in water balance; an immediate polyuria due to HDI followed by a more prolonged period of antidiuresis suggestive of AVP excess. The antidiuretic phase may last several weeks and can be followed by reversion to HDI or recovery. DI presenting with a pituitary mass should raise concerns about a diagnosis other than pituitary adenoma. HDI can worsen in pregnancy due to increased degradation of AVP by placental enzyme activity

 

Table 1. Etiology of HDI

Primary

Genetic

Wolfram syndrome

Autosomal dominant

Autosomal recessive

Developmental syndromes

Septo-optic dysplasia

 

Idiopathic

 

Secondary/acquired

Trauma

Head injury,

Post-surgery

Tumor

Craniopharyngioma

Germinoma

Metastases

Pituitary macroadenoma

Inflammatory

Sarcoidosis, Histiocytosis, Meningitis, Encephalitis, Infundibuloneurohypophysitis, Guillain–Barré syndrome, Autoimmune

 

Nephrogenic DI  

 

Renal resistance to AVP may reflect a toxic renal tubulopathy secondary to metabolic (e.g., hypokalemia; hypercalcemia) or drug effects (e.g., lithium). Prolonged polyuria of any cause can result in partial NDI through disruption of the intra-renal solute gradients and reduced tubular concentrating capacity.

 

X-linked familial NDI results from loss-of-function mutations in the renal AVP receptor (Figure 4). Autosomal recessive NDI is caused by loss-of-function mutations in the AVP-dependent renal water channel aquaporin-2.

 

 

Dipsogenic DI

 

Persistent high fluid intake leads to appropriate polyuria.  If intake exceeds the limit of renal free water excretion,hyponatremia may result.  DDI can be associated with abnormalities in thirst perception.

 

  • Low threshold for thirst
  • Exaggerated thirst response to osmotic challenge
  • Inability to suppress thirst at low plasma osmolalities

 

Neuroimaging is normal in most cases. DDI is associated with affective disorders.

 

DIAGNOSIS AND DIFFERENTIAL

 

History and examination may reveal important clinical information

 

-Features of systemic disease

-Associated endocrinopathy: suggestive of additional hypothalamic or pituitary dysfunction

-Neuro-ophthalmic problems suggestive of structural disease

-Evidence of drug toxicity (e.g., lithium, phenytoin)

 

There should be a standard initial diagnostic approach.

 

-Confirmation of true polyuria, distinct from simple frequency without excess urine volume

-Exclusion of common differentials such as drug (diuretics) and metabolic causes (hyperglycemia, hypercalcemia hypokalemia)

 

If polyuria is confirmed and simple causes are excluded, the clinician should proceed to a diagnostic Water Deprivation Test (Table 2)

 

Definitive diagnosis of DI requires testing of AVP production and action in response to osmolar stress. The water deprivation test is an indirect assessment of the AVP axis, measuring renal concentrating capacity in response to dehydration. It can be followed by assessment of renal response to the synthetic AVP analogue DDAVP, to determine whether any defect identified in urine concentrating ability can be corrected with AVP-replacement.

 

Table 2. Water Deprivation Test

Step 1 - Dehydration phase

Aim

Differentiate HDI and NDI from DDI

Procedure

Restrict all fluids between 8am-4pm in a controlled environment. Take baseline and 2 hourly measurements of weight, urine volume, urine osmolality, and plasma osmolality.  Abandon test if thirst becomes unbearable or if patient loses >5% initial weight.

Analysis

 

HDI and NDI:

Urine osmolality <300mOsm/kg

Plasma osmolality >290mOsm/kg

DDI:

Urine and plasma osmolality normal

Step 2 – DDAVP (desmopressin) response phase

Aim

Differentiate HDI from NDI

Procedure

At 4pm, administer desmopressin bolus (1mcg, intramuscular). Allow fluid intake up to 2x the volume of urine output in step 1. Continue to measure urine volume, urine osmolality and plasma osmolality every hour until 8pm.

Measure plasma osmolality and plasma sodium at 9am the next morning.

Interpretation

HDI:

Urine osmolality >750 mOsm/kg

NDI:

Urine osmolality remains low

 

Further Investigations

 

Water deprivation test results may be indeterminate.  If HDI is suspected but water deprivation test data are inconclusive, a reasonable approach is a therapeutic trial of 10-20 mcg intranasal DDAVP per day with close monitoring of plasma Na+. Patients with HDI note improved symptoms without significant dilutional hyponatremia. In the future, basal or stimulated measurement of copeptin may be the most useful investigation, when generally available.

 

Confirmation of HDI should lead to further pituitary function testing and cranial MRI. MRI may reveal the absence of posterior pituitary bright spot on T1- weighted sequences (Figure 5), or a pituitary mass. In the absence of structural problem, the MRI should be repeated 12 months after presentation to exclude slow growing mass lesion. NDI requires renal tract imaging and additional renal studies.

 

 

Diabetes Insipidus Combined with Defects in Thirst (Adipsic DI)

 

While the regulation of thirst and AVP are discrete, the close neuroanatomical relationship of the structures responsible for osmoregulation of both processes means that some structural, neurovascular and neuro-developmental lesions are associated with combined defects.  Absent or reduced thirst (adipsia) in association with HDI predisposes to hypernatremic dehydration. Diagnosis follows that outlined for HDI, with parallel assessment of thirst perception.

 

TREATMENT

 

Mild forms of HDI may not require treatment. Significant polyuria and polydipsia are treated effectively with DDAVP in divide doses: nasal spray 5-100 mcg per day; tablets 100-1000 mcg/day; or parenterally 0.1-2.0 mcg/day. Hyponatremia from plasma dilution can be avoided by omitting treatment for a short period on a regular basis (e.g., one dose per week).

 

NDI may respond to removal of the causal agent (such as correction of hypokalemia or cessation of Lithium). However, drug-induced NDI may persist. Symptoms may respond partly to high-dose DDAVP (e.g., 4 mcg i.m. bid.)  Hydrochlorothiazide (25 mg/day) either alone or in combination with Ibuprofen (200 mg/day) may be of some help. Urine output should not be expected to normalize.

 

The approach to DDI is reduction in fluid intake. DDAVP treatment must be avoided because of the risk of significant hyponatremia.

 

Patients with adipsic DI require careful management. Absence of normal thirst perception and/or regulation means that they may continue to drink at low plasma osmolalities that would normally suppress fluid intake. The combination of an obligate antidiuresis produced by DDAVP treatment, together with the potential for spontaneous fluid intake in excess of that required for maintenance of plasma volume and normal plasma osmolality, means they are at risk of fluid overload and dilutional hyponatremia. The same group of patients are also at risk of dehydration and hypernatremia if total body water loss is higher than a spontaneous fluid intake that is, by definition, uncoupled from normal osmo-regulatory control. In patients with adipsic DI, managing fluid balance to maintain normal plasma sodium is therefore challenging. One approach is to combine a fixed DDAVP-dependent antidiuresis (giving urine output of some 2 L/day) with a variable daily fluid intake that aims to maintain the patient’s body weight at that which is known to be associated with normal plasma volume and normal plasma sodium (the ‘target’ weight, see below).

 

e.g.  Fluid intake for given day (L) = 2 L (i.e., urine output from fixed dose DDAVP) - (weight on given day in kg - target weight in kg)

 

FOLLOW-UP 

 

Following initiation of DDAVP, patients require review for dose titration. When stable, they can be seen annually to assess symptom control and to check plasma Na+ levels to avoid over-treatment. Adipsic DI requires meticulous follow-up in a specialist service.

 

GUIDELINES

 

Baldeweg SE, Ball S, Brooke A, Gleeson HK, Levy MJ, Prentice M, Wass J. In-patient management of Cranial Diabetes Insipidus. Endocrine Connections 2018;

 

REFERENCES

 

Ball SG. The Neurohypophysis: Endocrinology of Vasopressin and Oxytocin. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2017 Apr 22. PMID: 25905380

 

Gubbi S, Hannah-Shmouni F, Koch CA, Verbalis JG. Diagnostic Testing for Diabetes Insipidus. 2019 Feb 10. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 30779536

 

Ball S 2013. Diabetes Insipidus. Medicine 41: 519-521. 10.1016/j.mpmed.2013.06.001

http://www.sciencedirect.com/science/article/pii/S1357303913001783

 

Refardt J, Winzeler B, Christ-Crain M. Endocrinol Metab Clin North Am. 2020 Sep;49(3):517-531. doi: 10.1016/j.ecl.2020.05.012. Epub 2020 Jul 15. PMID: 32741486

 

Garrahy A, Thompson CJ. Management of central diabetes insipidus. Best Pract Res Clin Endocrinol Metab. 2020 Sep;34(5):101385. doi: 10.1016/j.beem.2020.101385. Epub 2020 Jan 31. PMID: 32169331