Archives

Thyroid Nodules and Thyroid Cancer Prior to, During, and Following Pregnancy

ABSTRACT

 

Thyroid cancer is the second most common malignancy to co-occur in pregnancy. Further, the rising prevalence of treated thyroid cancer in women of child-bearing age means that survivors of thyroid cancer are frequently presenting for obstetric care, occasionally in the setting of persisting structural disease. To ensure that optimal health outcomes are achieved for mother and child, it is essential that pre-pregnancy issues are comprehensively addressed, and that management decisions during pregnancy remain both patient and child focused, best achieved through a woman-centered multidisciplinary team. As new data emerge regarding the impact of radioactive iodine on fertility, careful balancing of risk and benefits of this treatment is required. 

 

INTRODUCTION

 

Thyroid nodules are common in women of childbearing age. Thyroid nodules may be detected due to symptoms of local compression (either due to larger size, or pressure on the trachea or esophagus), but are more commonly detected incidentally on imaging performed for other reasons. As well as determining if compressive symptoms are present, all thyroid nodules must be risk-stratified for the presence of malignancy. A third factor is to determine whether the nodule is functional (i.e., autonomously producing thyroid hormone), however this cannot be reliably assessed during pregnancy as it is dependent on radionucleotide imaging which is contra-indicated during pregnancy. 

 

In general, the investigation or treatment of any new or co-existent medical conditions in pregnancy should be weighed against the separate risks and benefits to both the mother and fetus. Although most thyroid nodules will not grow during pregnancy, and therefore permit management decisions to be deferred to after birth (thus prioritizing fetal wellbeing), a small proportion of cases will require emergent management within pregnancy to prioritize maternal wellbeing (1).

 

Thyroid carcinomas generally develop from follicular epithelial cells (termed differentiated thyroid cancer, DTC) and present morphologically as papillary (PTC) or follicular (FTC) subtypes. Anaplastic thyroid cancer (ATC) is a rare, highly aggressive de-differentiated variant of DTC. Rarely, parafollicular, neuro-endocrine derived C-cells can give rise to medullary thyroid cancers (MTC). Thyroid lymphoma and metastases from other solid organ cancers are rare. In general, DTC has an greater than 98% 10 year survival in women of child-bearing age (2).

 

EPIDEMIOLOGY OF THYROID NODULES AND THYROID CANCER 

 

Thyroid Nodules

 

There is a clear female preponderance for the development of thyroid nodules that is demonstrated in studies from varied ethic groups, and in populations that are iodine-replete and iodine deficient (3-5). Thyroid nodules are also more prevalent with increasing age (6){Reiners, 2004 #1161}.  This may partly be explained by exposure to female reproductive hormones, as studies have demonstrated associations with increasing thyroid nodularity and multiparity, older age at menopause, and the presence of uterine fibroids (6-9). In a single study, use of oral contraceptive hormones was associated with reduced thyroid volume, but not a change in thyroid nodularity (10).

 

Carcinoma of the Thyroid Gland

 

The increased prevalence of thyroid nodules in females is matched by an increased prevalence of thyroid cancer amongst women.  SEER data from the United States cancer registry reports thyroid cancer incidence at 21 cases per 100,000 females, compared to 7.1 per 100,000 males (11).  When stratified by age, it is evident that this gender-based divergence is seen as early as puberty (Figure 1). The peak incidence of thyroid cancer amongst females occurs in midlife (age 35-59), and occurs earlier than the peak incidence in males (age 65-75), which corresponds with exposure to female reproductive hormones. ATC and MTC have equal incidence between genders.

Figure 1. Incidence of thyroid cancer by age- and gender- in the United States. Data source: SEER 18 (2010-2014). https://seer.cancer.gov/faststats

Although epidemiological data would suggest a strong link between exposure to female reproductive hormones and development of thyroid cancer, firm evidence linking reproductive factors to thyroid cancer risk is less clear. Some studies have shown a small (or transient) increase risk of DTC following pregnancy compared to nulliparous women (12).  Age of menarche, menopause, and menstrual cycle patterns present conflicting data (12), however in general, longer exposure to reproductive hormones appears associated with increased thyroid cancer risk (13-15). Conversely, extended periods of breastfeeding (resulting in prolonged reductions in cirulating estradiol), have been associated with with decreased incidence of thyroid cancer (14, 16, 17).

 

Incidence of Thyroid Carcinoma in Pregnancy

 

Multiple studies confirm that carcinoma of the thyroid gland is the second most frequent pregnancy-associated cancer, behind carcinoma of the breast. Registry studies suggest that thyroid cancer is present in between 14-27 per 100,000 mothers giving birth (18, 19). In most cases, this represents newly diagnosed thyroid cancer during pregnancy, which is usually organ-confined. However, a combination of increasing diagnosis of thyroid cancer amongst young women and excellent prognosis has resulted in an increasing cohort of survivors of thyroid cancer requiring obstetric care (20). This is demonstrated by data from Taiwan, showing thyroid cancer prevalence amongst women (175 cases per 100,000 women) is 9-fold higher than the incidence (18 cases per 100,000 women) (21). Occasionally, pregnancy occurs in a woman with known or suspected metastatic disease. A recent study from the USA reports that a historical diagnosis of thyroid cancer was the most common cancer present in women presenting for obstetric care (22).

 

IMPACT OF A PREGNANCY ON NUMBER AND SIZE OF THYROID NODULES

 

Impact of Pregnancy Hormones on Thyroid Follicular Epithelium

 

Pregnancy represents a stimulatory environment for thyroid follicular cells. The pregnancy hormone human chorionic gonadotrophin (HCG) is a heterodimeric glycoprotein. Although the beta subunit is unique, the alpha subunit is common to follicle stimulating hormone, luteinizing hormone, and thyroid stimulating hormone (TSH). As a result, this structural homology causes cross-stimulation of the TSH receptor by HCG, leading to physiological TSH-independent stimulation of the TSH-receptor, predominantly in the first trimester when HCG levels are highest. As well as contributing to gestational hyperthyroidism, in this way HCG mediated TSH-receptor signaling acts to stimulate growth of the thyroid follicular epithelium (23). Sustained activation of the signaling cascade mediated by the TSH-receptor has been associated with an increased risk of thyroid cancer in large observational studies. However, it is not known whether more limited periods of increased TSH-receptor signaling, such as would occur during pregnancy, materially contributes to thyroid cancer risk (24).

 

Iodine (not a pregnancy hormone) is a trace element required for normal maternal thyroid function and fetal thyroid development and function. Pregnancy increases maternal demands for iodine and a daily intake of approximately 250-300mcg is recommended (25).  Iodine excess and iodine deficiency states are both associated with an increased prevalence of thyroid nodules (3, 26).  

 

Changes in Number and Size of Thyroid Nodules During Pregnancy

 

As previously outlined, the hormonal environment of pregnancy is associated with the development of new thyroid nodules, and with potential growth of existing thyroid nodules.  Using ultrasound screening, thyroid nodules are demonstrated in 3-21% of pregnant women (26-28), although most nodules are small (<1cm) and not detectable clinically (26).  Prospective studies of pregnant women show an increase in thyroid nodule number and size during pregnancy. In a study of 221 women in China using repeated sonographic evaluation, an increase in nodule volume during pregnancy was shown in 15% of women in whom nodules were already present at baseline evaluation. New thyroid nodules were detected in 13% of the cohort. Post-partum, the number of women with thyroid nodules had increased from 15% to 24% (26). All nodules had a benign sonographic appearance. Similarly, a study of 726 pregnant women in Belgium identified a 3% incidence of thyroid nodules at baseline (determined by two-step screening with palpation followed by ultrasound). Of those with nodules, 60% showed an increase in size of at least 50%. Further, 20% (4/20) of women with regular sonographic surveillance developed new nodules during pregnancy (27).

 

PRESENTATION OF A NEW THYROID NODULE IN THE PREGNANT PATIENT       

 

A thyroid nodule usually comes to attention in pregnancy following the identification of a palpable abnormality. Screening for thyroid nodules in asymptomatic individuals without risk factors, both in the pregnant and non-pregnant population, is not recommended (29).

Thyroid nodules should be assessed using a triple assessment, including clinical assessment, sonographic risk stratification, and biopsy (in selected cases). Scintigraphy, which is part of the standard workup for functional nodules in the non-pregnant population, is contra-indicated in pregnancy due to the risk of ionizing radiation to the fetus.   

 

Important historical factors that increase the chance of a nodule being malignant include the presence of:

 

  • A familial cancer syndrome, including multiple endocrine neoplasia 2 (MEN2), familial PTC, Cowden’s syndrome, familial adenomatosis polyposis, and Carney Complex.
  • Neck irradiation in childhood, e.g., treatment for cancers of the head and neck
  • Exposure to ionizing radiation in early life (age <18 years)

 

On clinical examination, a palpable lump should be characterized. The presence of a large, very firm or rapidly growing nodule should raise concern for malignancy. Neck lymph nodes should be evaluated. Symptoms and signs of compression of local adjacent structures should be sought. 

 

Many thyroid nodules are functional, however the determination of the functional status of a thyroid nodule in pregnancy is limited. Firstly, although TSH should be checked, a low TSH may reflect gestational hyperthyroidism and should be interpreted with reference to the current gestational age. Most functional nodules progress slowly, therefore a pre-pregnancy TSH level which is at, or below, the lower limit of the reference range may provide a helpful clue. Secondly, radioactive isotopes used for thyroid scintigraphy readily cross the placenta, and the radiation exposure to the fetus does not justify the use of this modality in pregnancy.  Therefore, conclusive determination of whether a thyroid nodule is functional (and thus of very low malignant potential), or non-functional, during pregnancy is usually not possible. 

 

Serum biomarkers for thyroid cancer are not currently in routine use.  Although serum calcitonin is highly sensitive for the diagnosis of MTC (30), it is not validated for use in pregnancy, especially as calcitonin levels rise over the course of a normal pregnancy.  Further, its use in assessment of thyroid nodules in non-pregnant women is not universally established. Carcino-embryonic antigen, also a marker of MTC, can rise during pregnancy, and should be interpreted with caution (31).

 

Neck ultrasound is the definitive tool for assessment of thyroid nodules, and is safe in pregnancy. A high-frequency linear transducer is optimal to provide detailed characterization of the sonographic features, including size, echogenicity, shape, margins, the presence of calcification, and the presence of abnormal lymph nodes in the central and lateral neck.  All nodules should be risk stratified according to a validated scoring system, such as from the American Thyroid Association (32) or the American College of Radiology (33).  If fine-needle aspiration biopsy (FNAB) is required, this can be safely performed in all trimesters of pregnancy, with indications identical to that of the non-pregnant population (32). 

 

Nodules with higher risk features, such as larger size, growth in pregnancy, suspicion of extra-thyroidal extension, presence of large-volume nodal metastases, or suspicion for MTC or ATC should be considered for biopsy and surgery during pregnancy.  However, for smaller nodules without any high-risk features, consideration should be given to deferring biopsy (and any planned intervention) until the post-partum period, as several studies have confirmed that there is no survival benefit for surgery during pregnancy for low risk DTC (34).

 

IMPACT OF PREGNANCY ON A NEW DIAGNOSIS OF THYROID CANCER   

 

Impact of Pregnancy on Outcome of Thyroid Cancer

 

A diagnosis of thyroid cancer during pregnancy has the same excellent long term survival outcomes as seen in other settings. Large retrospective studies in the US population between 1962-1999 (35-37) show similar mortality data irrespective of the diagnosis setting (inside or outside pregnancy) or the timing of surgery (during pregnancy or post-partum).  Although these studies have the benefit of long follow up periods, they are inherently retrospective. Further, the ability of these studies to assess impact of pregnancy on thyroid cancer recurrence is limited, not only due to their retrospective nature, but also due to lack of availability of highly sensitive thyroglobulin assays and high resolution neck ultrasound in historic series (34).

 

In contrast, recent studies suggest that thyroid cancer diagnosed in pregnancy may have a higher risk of recurrence. A retrospective study from Italy showed higher rates of persistent or recurrent thyroid cancer in women diagnosed either during pregnancy or within 12 months after birth (60% persistent or recurrent disease), compared to women diagnosed with thyroid cancer more than 12 months after a pregnancy (4% persistent or recurrent disease) or women who were never pregnant (13% persistent or recurrent disease) (38). However, it is important to note that most of the pregnant women with thyroid cancer underwent surgery in the second trimester (73%), and it is possible that a more limited surgical approach in this setting may have confounded these results. 

 

Similarly, a second retrospective Italian study found a higher rate of persistent or recurrent disease in women with thyroid cancer diagnosed within two years of a pregnancy (11%), compared to women diagnosed more than two years after a pregnancy (1%) or those who were never pregnant (5%) (39). 

 

A pathology study from Australia found that DTC diagnosed within 12 months of pregnancy were more likely to be larger, and have nodal metastases than matched controls (40).

 

Overall, these data should reassure clinicians and patients that the impact of pregnancy on a newly diagnosed thyroid cancer is low, with excellent overall survival outcomes. Epidemiological and clinical data would suggest that the stimulatory milieu of pregnancy may contribute to a slightly higher overall risk of recurrence, which should be taken into consideration when planning follow up strategies.  

 

Timing of Thyroidectomy

 

Expert consensus affirms that thyroidectomy can safely be performed in the second trimester, but is often more appropriately deferred to the post-partum period (34). Clinical markers of aggressive pathology, such as large primary size, rapid growth, or bulky lateral neck nodal disease, would support a strategy of earlier surgery. At present, these clinical markers of aggressiveness are detected by specialized thyroid and neck ultrasound, which should occur at first assessment, and subsequently around 20 weeks (to allow for planning of thyroidectomy in the second trimester, if indicated). Suspicion of non-DTC pathology, such as MTC and ATC, should warrant strong consideration of early surgery. 

 

The optimal timing of surgery in the peri-partum period is uncertain.  Whist many women undergo safe surgery and anesthesia in the second trimester, small risks for mother and fetus remain. However, deferring surgery to the post-partum period potentially disrupts dyadic attachment between mother and child, and may interrupt breast-feeding. As evidence is lacking, patient-centered decision making, with inputs from a multi-disciplinary team, is appropriate. 

 

Small case series support a strategy of deferred surgery for low-risk lesions. For example, 19 women with PTC diagnosed around the time of conception were followed sonographically in pregnancy (41). Nearly 70% were microcarcinomas, and 3 cases had sonographic N1 disease. During pregnancy, 3 tumors had a detectable increase in maximal diameter, while 5 increased in volume. In 2 out of 3 with N1 disease, lymph nodes increased in size although no new nodal disease was detected, and the extent of surgery was not changed. Post-partum, 16 cases proceeded to surgery around 12 months following diagnosis.

 

Maternal Supportive Management During Pregnancy

 

Thyroid stimulating hormone (TSH) is a trophic factor for follicular thyroid cells and is associated with progression of thyroid cancer (23). However, there is no evidence to support the practice of pharmacological suppression of TSH to minimize growth of a primary tumor in pregnancy, and exogenous maternal hyperthyroidism is associated with fetal risk.  Maintaining maternal serum TSH within the lower half of the pregnancy-specific reference range is a reasonable therapeutic goal, and levothyroxine should be initiated, if required, to achieve this target.

 

Dietary iodine should not be restricted, as it is essential for fetal thyroid development.  Maternal physiological demands for iodine increase in pregnancy, and maternal iodine deficiency is associated with development of goiter in the mother. 

 

A recent cohort study found that pregnancies complicated by a diagnosis of thyroid cancer prior to or during pregnancy had a higher incidence of venous thromboembolism (odds radio 2.4) and blood transfusions (odds ratio 2.1), however there was no impact on neonatal outcomes (42). Similarly, the rate of post-partum hemorrhage in women with a history of thyroid cancer was higher than controls (odds ratio 1.23) in a large retrospective observational study, however no other adverse maternal, neonatal, or child outcomes (followed to 80 months post-partum) were found (43).

 

Measurement of Serum Thyroglobulin

 

During pregnancy, maternal serum thyroglobulin levels are higher than pre-pregnancy. This may be an effect of stimulation of maternal thyrocytes by estrogen and HCG. Therefore, maternal thyroglobulin levels during pregnancy must be interpreted with caution. Maternal serum thyroglobulin levels return to baseline values within 1-6 months of pregnancy (44-46).  In general, thyroglobulin status should be assessed no earlier than 6 weeks post-partum. 

 

Considerations in the Planning of Radioiodine Therapy

 

Radioactive iodine therapy following total thyroidectomy, either for remnant ablation, or as adjuvant therapy, is recommended for a subset of DTC with higher risk of recurrence (32).  The administration of radioiodine following pregnancy poses unique challenges, both medically and socially. Firstly, radiation safety precautions necessitate that close contact between the mother and her infant (as well as other young children) must be avoided for around 7 days following a radioactive iodine dose (precise recommendations are determined at the time of therapy) (47). Radioactive iodine is contra-indicated in pregnancy, and if administered, the risks to the fetus must be carefully assessed, based on administered dose and gestational age (48). Secondly, breast tissue expresses the sodium-iodide symporter, which is upregulated during lactation to concentrate iodine in breast milk (48, 49).  Consequently, to minimize exposure of breast tissue to ionizing radiation, lactation should cease a minimum of 8 weeks before radioactive iodine and should not be recommenced so as to avoid potential breast-milk associated radioactive iodine exposure. 

 

In light of this, the timing of radioactive iodine (if required) should be considered, balancing the potential risk of DTC progression without treatment, the benefits of a period of breastfeeding, and family unit dynamics. The literature is conflicting as to whether radioiodine administered early (within 3 months) or late (within 12 months), has any impact on prognosis. For example, a large retrospective database study including more than 9,000 patients diagnosed with high-risk PTC (primary tumor >4cm, N1 disease, positive surgical margins) found that timing of radioactive iodine within the first 12 months did not impact mortality (the median survival in this cohort was 75 months), after adjustment for confounders (50). In contrast, a small retrospective study of patients with lower risk DTC (235 cases classified as either ATA Low- or ATA-Intermediate- risk) found that deferring radioactive iodine longer than 3 months post-operatively was associated with higher rates of biochemical incomplete or structural incomplete responses compared to earlier radioactive iodine ablation(19% vs 4%) (51).

 

PRE-CONCEPTION CARE OF WOMEN WITH A HISTORY OF THYROID CANCER             

 

Pregnancy following diagnosis and treatment for thyroid cancer is common, and presents specialized management issues. Nonetheless, excellent obstetric outcomes are expected (52).  Pre-conception counselling is recommended for all women with a past history of thyroid cancer.

 

Checklist: Management issues prior to pregnancy in survivors of thyroid cancer.

 

  1. Assessment of thyroid cancer status:
    • Remission? Assessment of disease status: structural and biochemical
    • Potential impact of pregnancy on disease progression
  2. Impact of prior radioiodine therapy on timing of conception and future fertility
    • Ensure > 6 months between radioactive iodine and conception
  3. Thyroid hormone replacement
    • Pre-pregnancy optimization of levothyroxine replacement
    • Pre-emptive adjustment to levothyroxine dosing following conception
    • Potential for unmasking thyroid hormone insufficiency in women with sufficient pre-pregnancy thyroid hormones from a residual hemithyroid
    • Use of pregnancy supplements that may interfere with levothyroxine absorption

 

Establishment of Thyroid Cancer Status

 

To provide a framework for discussing the potential impact of thyroid cancer on pregnancy, an assessment of disease status is valuable, such as recommended by the ATA in its 2015 guidance (Table 1) (32). Evidently, counselling and management discussions will differ depending on what treatment has previously been received (total thyroidectomy vs hemithyroidectomy), the presence of any functional thyroid hormone production (if prior hemithyroidectomy only), the timing of any radioactive iodine administration, and the presence of any residual cancer. MicroPTC under active surveillance is a distinct management issue which is discussed separately.

 

Table 1. 2015 American Thyroid Association Risk Stratification for DTC

2015 ATA Response-to-Therapy classification

Description

Excellent response

No clinical, biochemical or structural evidence of persistent or recurrent thyroid cancer.

Biochemical-incomplete response

Elevated serum thyroglobulin, or rising anti-thyroglobulin antibodies, in the absence of structural disease identifiable on imaging.

Structural-incomplete response

Persistent or recurrent thyroid cancer visible on imaging, either in neck or distant metastases

Indeterminate response

Non-specific biochemical or structural findings that are not able to be classified as benign or malignant (includes stable/declining anti-thyroglobulin antibody levels without evidence of structural disease)

2015 American Thyroid Association Risk Stratification for DTC, tabulated from Haugen et al. (2016).  Refer to ATA Guideline (32) for full discussion of each class and qualifying criteria (Table 13).

 

In women with a history of MTC, the tumor markers calcitonin and CEA are sensitive to detect residual or recurrent disease, and allow for post-operative risk stratification (53).  There are no studies examining whether pregnancy impacts the prognosis of MTC. 

 

Discussing Impact of Pregnancy on Risk of Recurrence

 

There is a growing body of evidence reporting the long-term oncological outcomes in the setting of pregnancy following treatment for thyroid cancer. Key studies are reviewed below.

 

Leboeuf et al. (46) reported outcomes of 36 women between 1997 and 2006, with pregnancy a median 4 years following treatment for DTC. Three women had structural disease present prior to pregnancy, and of these, one showed growth in a cervical lymph node. A further two women developed recurrence following pregnancy that was not present on pre-operative physical examination. Of the full cohort, 22% had a sustained >20% rise in serum thyroglobulin post-partum. 

 

Rosario et al. (54) describe the outcome of 64 pregnancies, occurring a median of 2.4 years after treatment for DTC. In this cohort, no patient had evidence of structural disease either prior to or following pregnancy. Of the subset 49 women with undetectable thyroglobulin prior to pregnancy, this remained undetectable in the post-partum period. Of the 8 patients with low level thyroglobulin prior to pregnancy, no significant post-partum change was observed. 

 

Hirsch et al. (55) studied the outcome of 63 women, where pregnancy occurred a median of 5 years after treatment for PTC.  Of the subset of 6 women with known structural disease prior to conception, 80% were found to have progressed within 12 months of birth (2 with biochemical progression, 3 with structural progression). Of the subset of 5 women with detectable pre-pregnancy thyroglobulin, no significant change was observed post-partum.  Of the remaining 39 women with undetectable pre-pregnancy thyroglobulin, no progression was observed. 

 

Finally, Rakhlin et al. reported the outcome of pregnancy in 235 women following treatment for DTC (56), retrospectively grouped into ATA Response to Therapy criteria (Table 1).  In the 197 women without structural disease prior to pregnancy, no new structural disease was detected following post-partum evaluation.  However, 8% had a significant rise in thyroglobulin.

 

Overall, these data are reassuring that women with an ATA Excellent response to therapy have a very low risk of DTC progression occurring during pregnancy, and a low risk of DTC progression following pregnancy. As such, additional monitoring of thyroid cancer status during pregnancy for these women is not required (34). 

 

However, women with biochemical or structural evidence of disease may have a progression of their thyroid cancer status as a result of pregnancy.  Based on the above studies, the degree of disease progression appears minor, only affects a subset of women, and does not appear to have an impact on the outcome of the pregnancy. 

 

Reducing the Impacts of Prior Thyroid Cancer Treatment on Pregnancy.

 

LEVOTHYROXINE REPLACEMENT  

 

It is essential that all women who are planning pregnancy receive written instructions for the management of thyroid hormone replacement prior to, and immediately following conception.  Requirements for thyroid hormone rise early in gestation, in part as a result of an increase in thyroid-binding globulin. Adequate levels of thyroid hormones are required for healthy fetal development and pregnancy progression.

 

Women previously treated with hemithyroidectomy may unmask relative thyroid hormone deficiency following conception, and may require early initiation of levothyroxine therapy in the first trimester.

 

Women who have been treated with total thyroidectomy will always require an increase in thyroid hormone replacement at conception, of a magnitude between 15-40% of the total weekly dose. A common practice is to advise women to “double the dose” of levothyroxine that they take on two days of the week as soon as pregnancy is confirmed, with further adjustment based on regular thyroid function tests throughout pregnancy (34, 57). Women who adhered to this advice were more likely to have TSH at the pregnancy target than those that deferred thyroxine adjustment until the first specialist consultation (58). 

 

Importantly, pregnancy multivitamins, iron supplements, or calcium supplements may interfere with the absorption of thyroxine, and women should be specifically instructed to take such supplements at a different time of day to minimize interference (59). 

 

Women should be reassured that levothyroxine is both safe and essential for a healthy pregnancy, as inadvertent discontinuation in early pregnancy has been reported (60). In most cases, the TSH target prior to pregnancy (usually targeting the lower half of the normal range) will remain appropriate in pregnancy. Pharmacological suppression of TSH with supra-physiological doses of levothyroxine could be continued in the setting of persistent structural disease, however care should be taken to avoid overt hyperthyroidism, which increases pregnancy risk. In settings where a TSH-suppression strategy has been pursued outside of pregnancy, but in the absence of known structural disease, a careful balancing of risk and benefit should be considered, as although mild hyperthyroidism in pregnancy has not been shown to lead to maternal or fetal complications, greater degrees of hyperthyroidism are associated with adverse pregnancy outcomes (34, 61, 62).

 

IMPLICATIONS OF PREVIOUS RADIOIODINE  

 

Women should defer conception for at least 6 months after radioactive iodine administration.  This period includes the expected time for radioactive iodine to fully decay (approximately 10 weeks), thus avoiding exposing the fetus to gamma-particle emission). A recent large population-based cohort study found that pregnancy occurring within 5 months of radioactive iodine had a higher rate of congenital malformations (odds ratio 1.74, 95%CI, 1.01-2.97; P = .04), which was not seen if conception occurred after 6 months (63). Deferring pregnancy for at least 6 months has the additional benefit of permitting assessment of the response to radioactive iodine therapy, and to determine that no additional treatment with radioactive iodine would be recommended in the following 15 months (conception, pregnancy and the post-partum period) (64). Stabilization of levothyroxine replacement can also take a period of months.

 

In the 12 months following radioactive iodine, 8-16% of women experience amenorrhea, and 12-31% have menstrual irregularities (65). Several studies (including a meta-analysis) have confirmed a small but significant fall in AMH levels following radioactive iodine, and a slightly earlier age of menopause than women who did not receive radioactive iodine (49.5 vs 51 years) (65, 66). 

 

Most studies have not shown that radioactive iodine has an impact on future fertility (65, 67, 68). However, in a retrospective database study comparing survivors of thyroid cancer, women in the age 35-39 subgroup who received radioactive iodine had a lower birth rate (11 vs 16 births per 1000 woman-years) than women who did not receive radioactive iodine.  However, as the time from diagnosis of thyroid cancer to first live birth was also prolonged in this study, it is not clear whether this finding is due to physician recommendation to delay pregnancy, or the biological effects of radioactive iodine (68). In addition, a recent population case-control study found a higher rate of infertility diagnosis amongst survivors of thyroid cancer (69), however this analysis did not take into account any disease-specific factors such as type of treatment received. 

 

In women with a history of thyroid cancer requiring assisted reproductive techniques, pregnancy outcomes were not different compared to controls, although the number of retrieved oocytes was lower (70). A history of radioactive iodine treatment was not associated with differing rates of clinical pregnancy or live birth rates in this group (71).

 

A large longitudinal study followed 2,673 pregnancies and did not show an increase in maternal or fetal adverse events in women previously administered radioactive iodine (72).  A population-based cohort study of women with thyroid cancer in Korea, comparing 59,483 women who underwent thyroidectomy alone, with 51,976 women who had thyroidectomy followed by radioactive iodine found no difference in pregnancy or obstetric outcomes in the 9.7% of the cohort where pregnancy occurred (63). A further systematic review (67), and meta-analysis (73), pooling additional studies reported similar findings, providing sufficient time had elapsed following radioactive iodine administration. 

 

In men, radioactive iodine may transiently impact testicular function, with a short-term rise in FSH, and decrease in normal sperm morphology seen in prospective studies (74). It is suggested that men avoid fathering children for 4 months following radioactive iodine (75, 76). In men who desire fertility, and who are expected to require high cumulative activities of radioactive iodine, sperm banking should be considered. 

 

Surveillance and Monitoring During Pregnancy

 

Based on available data, women with no structural or biochemical evidence of thyroid cancer do not require DTC-specific monitoring during pregnancy. At present, there is no evidence to guide whether additional post-partum surveillance should be instituted beyond that woman’s current surveillance strategy, however consideration of neck ultrasound and serum thyroglobulin at least 6 months post-partum is reasonable. 

 

For women with ‘ATA Biochemical Incomplete’ or ‘ATA Indeterminate’ classification, surveillance during pregnancy could include periodic neck ultrasound, and determination of thyroglobulin and Tg-Ab levels. Clear evidence of progression of thyroid cancer could prompt an increase in the level of TSH suppression, or rarely prompt expedited delivery. Management in the context of a multidisciplinary team is advised. 

 

CONTINUED ACTIVE SURVELLANCE OF PAPILLARY THRYOID MICROCARCINOMA DURING PREGNANCY

 

Non-operative management of microPTC (<10mm in maximal dimension) is increasing, with emerging data on implications for active surveillance during pregnancy. Shindo et al report 9 women with microPTC followed during pregnancy, finding demonstrable growth in 44% (compared to microPTC growth of 11% in non-pregnant controls) (77). Ito et al reported outcomes of 50 pregnancies with microPTC, finding growth of >1mm in 8%, reduced size in 2%. The remaining 90% of cases showed no growth in pregnancy, and there were no nodal metastases detected (78). Oh et al described 13 microPTC in pregnancy, with a single lesion demonstrating growth (41). The available evidence supports the continuation of active surveillance during pregnancy, monitored with periodic neck ultrasound. However, women contemplating pregnancy who are under active surveillance should be advised that a small number of microPTC will grow during pregnancy, and this may result in anxiety for the patient and clinicians. Further studies are awaited in this population (32).      

 

Germline RET Mutations

 

Women with clinically diagnosed MEN2, or who carry a germline mutation in the REarranged during Transfection (RET) proto-oncogene, should be under the care of a specialized clinical team, and should be offered detailed pre-natal genetic counselling.  Individual RET mutations can be characterized for their risk of early-onset MTC, allowing personalized management decisions. The highest risk mutations should prompt consideration of total thyroidectomy in early childhood (53). The presence of hyperparathyroidism and pheochromocytoma should be biochemically excluded prior to pregnancy in any woman with MEN2.

 

MANAGEMENT OF KNOWN RESIDUAL STRUCTURAL DISEASE IN PREGNANCY

 

Case series of pregnancy in women with co-existent thyroid cancer metastases have been reported. The largest study retrospectively studied a cohort of 124 women from China, aged 16-35 years, with lung metastases from thyroid cancer, stratified by whether pregnancy occurred (n=35) and followed for a median 68-82 months after completing treatment with radioactive iodine (79). This study found that pregnancy after thyroid cancer had no measurable difference in 5 year or 10-year progression free survival or overall survival. 10-year overall survival in the pregnancy group was 86%, compared to 82% in the non-pregnant group. Although the groups appeared to have similar characteristics, it remains possible that women who chose pregnancy had a lower severity of disease than those who avoided pregnancy. 

 

Another study retrospectively described outcomes for 38 women at a large cancer center in the USA (56). Included in the cohort were 10 women with pulmonary metastases at the time of diagnosis (and of whom 7 had persistent structural disease prior to pregnancy). During pregnancy, 29% of women had progression of structural disease (11/38, with 5/38 increasing size of known abnormal nodes, 3/38 with newly abnormal lymph nodes, and 1/38 with progression of distant metastases). In total, 3/38 (~8%) were considered “clinically significant” by the study team (required further treatment within 12 months of birth).

 

These data are reassuring that the clinical impact of pregnancy in the setting of persistent structural disease appears low, despite in vitro studies and smaller case series confirming that pregnancy represents a potentially stimulatory setting for thyroid cancer cells. 

 

In general, TSH suppression should be maintained where benefit is felt to outweigh risk to the pregnancy. Serial neck ultrasound during pregnancy will monitor the status of neck disease, however imaging the chest is usually avoided to minimize ionizing radiation to the chest. Where progression of lung metastases is to be monitored, serial lung function testing may be informative.  

 

Currently approved small molecule tyrosine kinase inhibitors have been shown to have embryotoxicity, fetotoxicity, and teratogenicity in rats and rabbits (80, 81), and pregnancy should be avoided in women on this treatment.  A case report of a pregnancy in a women treated with vandetanib up until 6 weeks gestation described no fetal adverse outcomes (82).

 

REFERENCES

 

  1. Oduncu FS, Kimmig R, Hepp H, Emmerich B. Cancer in pregnancy: maternal-fetal conflict. Journal of cancer research and clinical oncology. 2003;129(3):133-46.
  2. Tuttle RM, Haugen B, Perrier ND. Updated American Joint Committee on Cancer/Tumor-Node-Metastasis Staging System for Differentiated and Anaplastic Thyroid Cancer (Eighth Edition): What Changed and Why? Thyroid. 2017;27(6):751-6.
  3. Song J, Zou SR, Guo CY, Zang JJ, Zhu ZN, Mi M, et al. Prevalence of Thyroid Nodules and Its Relationship with Iodine Status in Shanghai: a Population-based Study. Biomedical and environmental sciences : BES. 2016;29(6):398-407.
  4. Moifo B, Moulion Tapouh JR, Dongmo Fomekong S, Djomou F, Manka'a Wankie E. Ultrasonographic prevalence and characteristics of non-palpable thyroid incidentalomas in a hospital-based population in a sub-Saharan country. BMC medical imaging. 2017;17(1):21.
  5. Knudsen N, Laurberg P, Perrild H, Bulow I, Ovesen L, Jorgensen T. Risk factors for goiter and thyroid nodules. Thyroid. 2002;12(10):879-88.
  6. Reiners C, Wegscheider K, Schicha H, Theissen P, Vaupel R, Wrbitzky R, et al. Prevalence of thyroid disorders in the working population of Germany: ultrasonography screening in 96,278 unselected employees. Thyroid. 2004;14(11):926-32.
  7. Wang K, Yang Y, Wu Y, Chen J, Zhang D, Liu C. The association of menstrual and reproductive factors with thyroid nodules in Chinese women older than 40 years of age. Endocrine. 2015;48(2):603-14.
  8. Spinos N, Terzis G, Crysanthopoulou A, Adonakis G, Markou KB, Vervita V, et al. Increased frequency of thyroid nodules and breast fibroadenomas in women with uterine fibroids. Thyroid. 2007;17(12):1257-9.
  9. Kim MH, Park YR, Lim DJ, Yoon KH, Kang MI, Cha BY, et al. The relationship between thyroid nodules and uterine fibroids. Endocrine journal. 2010;57(7):615-21.
  10. Knudsen N, Bulow I, Laurberg P, Perrild H, Ovesen L, Jorgensen T. Low goitre prevalence among users of oral contraceptives in a population sample of 3712 women. Clin Endocrinol (Oxf). 2002;57(1):71-6.
  11. Thyroid Cancer Bethesda, Maryland: National Cancer Institute; 2016 [Available from: http://seer.cancer.gov/statfacts/html/thyro.html.
  12. Moleti M, Sturniolo G, Di Mauro M, Russo M, Vermiglio F. Female Reproductive Factors and Differentiated Thyroid Cancer. Frontiers in endocrinology. 2017;8:111.
  13. He JL, Zhang C, Hu MJ, Wu HB, Lu XL, Hao JH, et al. Reproductive and menstrual factors for papillary thyroid cancer risk: A case-control study in Chinese women. Cancer Epidemiol. 2021;73:101964.
  14. Kim H, Kim KY, Baek JH, Jung J. Are Pregnancy, Parity, Menstruation and Breastfeeding Risk Factors for Thyroid Cancer? Results from the Korea National Health and Nutrition Examination Survey, 2010 to 2015. Clin Endocrinol (Oxf). 2018.
  15. Schubart JR, Eliassen AH, Schilling A, Goldenberg D. Reproductive Factors and Risk of Thyroid Cancer in Women: An Analysis in the Nurses' Health Study II. Womens Health Issues. 2021;31(5):494-502.
  16. Yi X, Zhu J, Zhu X, Liu GJ, Wu L. Breastfeeding and thyroid cancer risk in women: A dose-response meta-analysis of epidemiological studies. Clinical nutrition (Edinburgh, Scotland). 2016;35(5):1039-46.
  17. Wang M, Gong WW, He QF, Hu RY, Yu M. Menstrual, reproductive and hormonal factors and thyroid cancer: a hospital-based case-control study in China. BMC Womens Health. 2021;21(1):13.
  18. Smith LH, Danielsen B, Allen ME, Cress R. Cancer associated with obstetric delivery: results of linkage with the California cancer registry. American Journal of Obstetrics & Gynecology. 2003;189(4):1128-35.
  19. Cottreau CM, Dashevsky I, Andrade SE, Li DK, Nekhlyudov L, Raebel MA, et al. Pregnancy-Associated Cancer: A U.S. Population-Based Study. J Womens Health (Larchmt). 2019;28(2):250-7.
  20. Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM. Trends in Thyroid Cancer Incidence and Mortality in the United States, 1974-2013. Jama. 2017;317(13):1338-48.
  21. Liu F-C, Lin H-T, Lin S-F, Kuo C-F, Chung T-T, Yu H-P. Nationwide cohort study on the epidemiology and survival outcomes of thyroid cancer. Oncotarget. 2017;8(45):78429-51.
  22. Wu PMMD, Jordan KPP, Chew-Graham CAMMD, Mohamed MOM, Barac AMDP, Lundberg GPMD, et al. In-Hospital Complications in Pregnant Women With Current or Historical Cancer Diagnoses. Mayo Clinic Proceedings. 2021;96(11):2779-92.
  23. Rowe CW, Paul J, Geyde C, Tolosa J, Bendinelli C, McGrath S, et al. Targeting the TSH receptor in thyroid cancer. Endocrine Related Cancer. 2017.
  24. Nieto HR, Boelaert K. Thyroid stimulating hormone in thyroid cancer: does it matter? Endocrine-related cancer. 2016.
  25. Delange F. Iodine requirements during pregnancy, lactation and the neonatal period and indicators of optimal iodine nutrition. Public health nutrition. 2007;10(12a):1571-80; discussion 81-3.
  26. Kung AW, Chau MT, Lao TT, Tam SC, Low LC. The effect of pregnancy on thyroid nodule formation. J Clin Endocrinol Metab. 2002;87(3):1010-4.
  27. Glinoer D, Soto MF, Bourdoux P, Lejeune B, Delange F, Lemone M, et al. Pregnancy in patients with mild thyroid abnormalities: maternal and neonatal repercussions. J Clin Endocrinol Metab. 1991;73(2):421-7.
  28. Struve CW, Haupt S, Ohlen S. Influence of frequency of previous pregnancies on the prevalence of thyroid nodules in women without clinical evidence of thyroid disease. Thyroid. 1993;3(1):7-9.
  29. US Preventative Services Task Force. Screening for thyroid cancer: US Preventive Services Task Force Recommendation Statement. Jama. 2017;317(18):1882-7.
  30. Costante G, Meringolo D, Durante C, Bianchi D, Nocera M, Tumino S, et al. Predictive value of serum calcitonin levels for preoperative diagnosis of medullary thyroid carcinoma in a cohort of 5817 consecutive patients with thyroid nodules. Journal of Clinical Endocrinology and Metabolism. 2007;92(2):450-5.
  31. Ercan S, Kaymaz O, Yucel N, Orcun A. Serum concentrations of CA 125, CA 15-3, CA 19-9 and CEA in normal pregnancy: a longitudinal study. Archives of gynecology and obstetrics. 2012;285(3):579-84.
  32. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016;26(1):1-133.
  33. Tessler FN, Middleton WD, Grant EG, Hoang JK, Berland LL, Teefey SA, et al. ACR Thyroid Imaging, Reporting and Data System (TI-RADS): White Paper of the ACR TI-RADS Committee. Journal of the American College of Radiology : JACR. 2017;14(5):587-95.
  34. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, et al. 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017;27(3):315-89.
  35. Moosa M, Mazzaferri EL. Outcome of differentiated thyroid cancer diagnosed in pregnant women. J Clin Endocrinol Metab. 1997;82(9):2862-6.
  36. Yasmeen S, Cress R, Romano PS, Xing G, Berger-Chen S, Danielsen B, et al. Thyroid cancer in pregnancy. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2005;91(1):15-20.
  37. Herzon FS, Morris DM, Segal MN, Rauch G, Parnell T. Coexistent thyroid cancer and pregnancy. Archives of otolaryngology--head & neck surgery. 1994;120(11):1191-3.
  38. Vannucchi G, Perrino M, Rossi S, Colombo C, Vicentini L, Dazzi D, et al. Clinical and molecular features of differentiated thyroid cancer diagnosed during pregnancy. Eur J Endocrinol. 2010;162(1):145-51.
  39. Messuti I, Corvisieri S, Bardesono F, Rapa I, Giorcelli J, Pellerito R, et al. Impact of pregnancy on prognosis of differentiated thyroid cancer: clinical and molecular features. Eur J Endocrinol. 2014;170(5):659-66.
  40. Lee JC, Zhao JT, Clifton-Bligh RJ, Gill AJ, Gundara JS, Ip J, et al. Papillary thyroid carcinoma in pregnancy: a variant of the disease? Ann Surg Oncol. 2012;19(13):4210-6.
  41. Oh H-S, Kim WG, Park S, Kim M, Kwon H, Jeon MJ, et al. Serial Neck Ultrasonographic Evaluation of Changes in Papillary Thyroid Carcinoma During Pregnancy. Thyroid. 2017;27(6):773-7.
  42. Spiegel E, Spence AR, Czuzoj-Shulman N, Abenhaim HA. Pregnancy outcomes after thyroid cancer. J Perinat Med. 2019;47(7):710-6.
  43. Cho GJ, Kim SY, Lee HC, Lee KM, Han SW, Oh MJ, et al. Risk of Adverse Obstetric Outcomes and the Abnormal Growth of Offspring in Women with a History of Thyroid Cancer. Thyroid. 2019;29(6):879-85.
  44. Nakamura S, Sakata S, Komaki T, Kojima N, Kamikubo K, Miyazaki S, et al. Serum thyroglobulin concentration in normal pregnancy. Endocrinologia japonica. 1984;31(6):675-9.
  45. Rasmussen NG, Hornnes PJ, Hegedüs L, Feldt-Rasmussen U. Serum thyroglobulin during the menstrual cycle, during pregnancy, and post partum. Acta Endocrinologica. 1989;121(2):168-73.
  46. Leboeuf R, Emerick LE, Martorella AJ, Tuttle RM. Impact of pregnancy on serum thyroglobulin and detection of recurrent disease shortly after delivery in thyroid cancer survivors. Thyroid. 2007;17(6):543-7.
  47. Sisson JC, Freitas J, McDougall IR, Dauer LT, Hurley JR, Brierley JD, et al. Radiation safety in the treatment of patients with thyroid diseases by radioiodine 131I : practice recommendations of the American Thyroid Association. Thyroid. 2011;21(4):335-46.
  48. Iijima S. Effects of fetal involvement of inadvertent radioactive iodine therapy for the treatment of thyroid diseases during an unsuspected pregnancy. Eur J Obstet Gynecol Reprod Biol. 2021;259:53-9.
  49. Azizi F, Smyth P. Breastfeeding and maternal and infant iodine nutrition. Clinical endocrinology. 2009;70(5):803-9.
  50. Suman P, Wang CH, Abadin SS, Block R, Raghavan V, Moo-Young TA, et al. Timing of radioactive iodine therapy does not impact overall survival in high-risk papillary thyroid carcinoma. Endocr Pract. 2016;22(7):822-31.
  51. Li H, Zhang YQ, Wang C, Zhang X, Li X, Lin YS. Delayed Initial Radioiodine Therapy Related to Incomplete Response in Low- to Intermediate-Risk Differentiated Thyroid Cancer. Clin Endocrinol (Oxf). 2018.
  52. Liu D, Wei Y, Zhao Y, Li R, Yan J, Qiao J. Obstetric outcomes in thyroid cancer survivors: A retrospective cohort study. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2021;155(1):119-24.
  53. Wells SA, Jr., Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, et al. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid. 2015;25(6):567-610.
  54. Rosario PW, Barroso AL, Purisch S. The effect of subsequent pregnancy on patients with thyroid carcinoma apparently free of the disease. Thyroid. 2007;17(11):1175-6.
  55. Hirsch D, Levy S, Tsvetov G, Weinstein R, Lifshitz A, Singer J, et al. Impact of pregnancy on outcome and prognosis of survivors of papillary thyroid cancer. Thyroid. 2010;20(10):1179-85.
  56. Rakhlin L, Fish S, Tuttle RM. Response to Therapy Status Is an Excellent Predictor of Pregnancy-Associated Structural Disease Progression in Patients Previously Treated for Differentiated Thyroid Cancer. Thyroid. 2017;27(3):396-401.
  57. Alexander EK, Marqusee E, Lawrence J, Jarolim P, Fischer GA, Larsen PR. Timing and magnitude of increases in levothyroxine requirements during pregnancy in women with hypothyroidism. The New England journal of medicine. 2004;351(3):241-9.
  58. Minakata M, Ito M, Kishi T, Hada M, Masaki Y, Nakamura T, et al. The magnitude of increased Levothyroxine dose during pregnancy in patients on thyroid-stimulating hormone (TSH) suppression treatment after total thyroidectomy for papillary carcinoma. Endocrine journal. 2021.
  59. Zamfirescu I, Carlson HE. Absorption of Levothyroxine When Coadministered with Various Calcium Formulations. Thyroid. 2011;21(5):483-6.
  60. Rowe C, Murray K, Woods A, Gupta S, Smith R, Wynne K. Metastatic Thyroid Cancer in Pregnancy: Risk and Uncertainty. Endocrinolgy Diabetes & Metabolism Case Reports. 2016.
  61. Casey BM, Dashe JS, Wells CE, McIntire DD, Leveno KJ, Cunningham FG. Subclinical hyperthyroidism and pregnancy outcomes. Obstetrics and gynecology. 2006;107(2 Pt 1):337-41.
  62. Millar LK, Wing DA, Leung AS, Koonings PP, Montoro MN, Mestman JH. Low birth weight and preeclampsia in pregnancies complicated by hyperthyroidism. Obstetrics and gynecology. 1994;84(6):946-9.
  63. Kim HO, Lee K, Lee SM, Seo GH. Association Between Pregnancy Outcomes and Radioactive Iodine Treatment After Thyroidectomy Among Women With Thyroid Cancer. JAMA Intern Med. 2020;180(1):54-61.
  64. International Atomic Energy Agency. Radiation protection of pregnant women in nuclear medicine: International Atomic Energy Agency;[Available from: https://www.iaea.org/resources/rpop/health-professionals/nuclear-medicine/pregnant-women.
  65. Piek MW, Postma EL, van Leeuwaarde R, de Boer JP, Bos AME, Lok C, et al. The Effect of Radioactive Iodine Therapy on Ovarian Function and Fertility in Female Thyroid Cancer Patients: A Systematic Review and Meta-Analysis. Thyroid. 2021;31(4):658-68.
  66. Yaish I, Azem F, Gutfeld O, Silman Z, Serebro M, Sharon O, et al. A Single Radioactive Iodine Treatment Has a Deleterious Effect on Ovarian Reserve in Women with Thyroid Cancer: Results of a Prospective Pilot Study. Thyroid. 2018;28(4):522-7.
  67. Sawka AM, Lakra DC, Lea J, Alshehri B, Tsang RW, Brierley JD, et al. A systematic review examining the effects of therapeutic radioactive iodine on ovarian function and future pregnancy in female thyroid cancer survivors. Clin Endocrinol (Oxf). 2008;69(3):479-90.
  68. Wu JX, Young S, Ro K, Li N, Leung AM, Chiu HK, et al. Reproductive outcomes and nononcologic complications after radioactive iodine ablation for well-differentiated thyroid cancer. Thyroid. 2015;25(1):133-8.
  69. Velez MP, Richardson H, Baxter NN, McClintock C, Greenblatt E, Barr R, et al. Risk of infertility in female adolescents and young adults with cancer: a population-based cohort study. Hum Reprod. 2021;36(7):1981-8.
  70. Huang N, Zeng L, Yan J, Chi H, Qiao J. Analysis of in vitro fertilization/intracytoplasmic sperm injection outcomes in infertile women with a history of thyroid cancer: a retrospective study. Reproductive Biology and Endocrinology. 2021;19(1):82.
  71. Huang N, Zeng L, Yan J, Chi H, Qiao J. Impact of thyroid cancer treatment on assisted reproductive technology outcomes in women with infertility. J Assist Reprod Genet. 2021;38(8):2121-8.
  72. Garsi JP, Schlumberger M, Rubino C, Ricard M, Labbe M, Ceccarelli C, et al. Therapeutic administration of 131I for differentiated thyroid cancer: radiation dose to ovaries and outcome of pregnancies. J Nucl Med. 2008;49(5):845-52.
  73. Zhang L, Huang Y, Zheng Y, Cai L, Wen J, Chen G. The effect of I-131 therapy on pregnancy outcomes after thyroidectomy in patients with differentiated thyroid carcinoma: a meta-analysis. Endocrine. 2021;73(2):301-7.
  74. Bourcigaux N, Rubino C, Berthaud I, Toubert ME, Donadille B, Leenhardt L, et al. Impact on testicular function of a single ablative activity of 3.7 GBq radioactive iodine for differentiated thyroid carcinoma. Hum Reprod. 2018;33(8):1408-16.
  75. Pacini F, Gasperi M, Fugazzola L, Ceccarelli C, Lippi F, Centoni R, et al. Testicular function in patients with differentiated thyroid carcinoma treated with radioiodine. J Nucl Med. 1994;35(9):1418-22.
  76. Poppe K, Bisschop P, Fugazzola L, Minziori G, Unuane D, Weghofer A. 2021 European Thyroid Association Guideline on Thyroid Disorders prior to and during Assisted Reproduction. Eur Thyroid J. 2021;9(6):281-95.
  77. Shindo H, Amino N, Ito Y, Kihara M, Kobayashi K, Miya A, et al. Papillary thyroid microcarcinoma might progress during pregnancy. Thyroid. 2014;24(5):840-4.
  78. Ito Y, Miyauchi A, Kudo T, Ota H, Yoshioka K, Oda H, et al. Effects of Pregnancy on Papillary Microcarcinomas of the Thyroid Re-Evaluated in the Entire Patient Series at Kuma Hospital. Thyroid. 2015;26(1):156-60.
  79. Xi C, Zhang Q, Song H-J, Shen C-T, Zhang G-Q, Sun J-W, et al. Pregnancy Does not Affect the Prognoses of Differentiated Thyroid Cancer Patients With Lung Metastases. The Journal of Clinical Endocrinology & Metabolism. 2021;106(8):e3185-e97.
  80. U.S Food & Drug Administration. Lenvima Prescribing Information: FDA; 2015 [Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/208692s000lbl.pdf.
  81. U.S Food & Drug Administration. Nexavar Prescribing Information: FDA; 2013 [Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021923s016lbl.pdf.
  82. Thomas N, Glod J, Derse-Anthony C, Baple EL, Obsborne N, Sturley R, et al. Pregnancy on vandetanib in metastatic medullary thyroid carcinoma associated with multiple endocrine neoplasia type 2B. Clin Endocrinol (Oxf). 2018.

Skin Manifestations of Diabetes Mellitus

ABSTRACT

 

Diabetes mellitus is a common and debilitating disease that affects a variety of organs including the skin. Between thirty and seventy percent of patients with diabetes mellitus, both type 1 and type 2, will present with a cutaneous complication of diabetes mellitus at some point during their lifetime. A variety of dermatologic manifestations have been linked with diabetes mellitus; these conditions vary in severity and can be benign, deforming, and even life-threatening. Such skin changes can offer insight into patients’ glycemic control and may be the first sign of metabolic derangement in undiagnosed patients with diabetes. Recognition and management of these conditions is important in maximizing the quality of life and in avoiding serious adverse effects in patients with diabetes mellitus.

 

INTRODUCTION

 

The changes associated with diabetes mellitus can affect multiple organ systems. Between thirty and seventy percent of patients with diabetes mellitus, both type 1 and type 2, will present with a cutaneous complication of diabetes mellitus at some point during their lifetime (1). Dermatologic manifestations of diabetes mellitus have various health implications ranging from those that are aesthetically concerning to those that may be life-threatening. Awareness of cutaneous manifestations of diabetes mellitus can provide insight into the present or prior metabolic status of patients. The recognition of such findings may aid in the diagnosis of diabetes, or may be followed as a marker of glycemic control. The text that follows describes the relationship between diabetes mellitus and the skin, more specifically: (1) skin manifestations strongly associated with diabetes, (2) non-specific dermatologic signs and symptoms associated with diabetes, (3) dermatologic diseases associated with diabetes, (4) common skin infections in diabetes, and (5) cutaneous changes associated with diabetes medications.

 

SKIN MANIFESTATIONS STRONGLY ASSOCIATED WITH DIABETES MELLITUS

 

Acanthosis Nigricans

 

EPIDEMIOLOGY

 

Acanthosis nigricans (AN) is a classic dermatologic manifestation of diabetes mellitus that affects men and women of all ages. AN is more common in type 2 diabetes mellitus (2) and is more prevalent in those with darker-skin color. AN is disproportionately represented in African Americans, Hispanics, and Native Americans (3). AN is observed in a variety of endocrinopathies associated with resistance to insulin such as acromegaly, Cushing syndrome, obesity, polycystic ovarian syndrome, and thyroid dysfunction. Unrelated to insulin resistance, AN can also be associated with malignancies such as gastric adenocarcinomas and other carcinomas (4).

 

PRESENTATION

 

AN presents chronically as multiple poorly demarcated plaques with grey to dark-brown hyperpigmentation and a thickened velvety to verrucous texture. Classically, AN has a symmetrical distribution and is located in intertriginous or flexural surfaces such as the back of the neck, axilla, elbows, palmer hands (also known as “tripe palms”), inframammary creases, umbilicus, or groin. Affected areas are asymptomatic; however, extensive involvement may cause discomfort or fetor. Microscopy shows hyperkeratosis and epidermal papillomatosis with acanthosis. The changes in skin pigmentation are primarily a consequence of hyperkeratosis, not changes in melanin. AN can present prior to the clinical diagnosis of diabetes; the presence of AN should prompt evaluation for diabetes mellitus and for other signs of insulin resistance.

 

PATHOGENESIS

 

The pathogenesis of AN is not completely understood. The predominant theory is that a hyperinsulin state activates insulin growth factor receptors (IGF), specifically IGF-1, on keratinocytes and fibroblasts, provoking cell proliferation, resulting in the aforementioned cutaneous manifestations of AN (5,6).

 

TREATMENT

 

Treatment of AN may improve current lesions and prevent future cutaneous manifestations. AN is best managed with lifestyle changes such as dietary modifications, increased physical activity, and weight reduction. In patients with diabetes, pharmacologic adjuvants, such as metformin, that improve glycemic control and reduce insulin resistance are also beneficial (7). Primary dermatologic therapies are usually ineffective especially in patients with generalized involvement. However, in those with thickened or macerated areas of skin, oral retinoids or topical keratolytics such as ammonium lactate, retinoic acid, or salicylic acid can be used to alleviate symptoms (8-10).

 

Diabetic Dermopathy

 

EPIDEMIOLOGY

 

Dermopathy (DD), also known as pigmented pretibial patches or diabetic shin spots, is the most common dermatologic manifestations of diabetes, presenting in as many as one-half of those with diabetes (11). Although disputed, some consider the presence of DD to be pathognomonic for diabetes. DD has a strong predilection for men and those older than 50 years of age (12). Although DD may antecede the onset of diabetes, it occurs more frequently as a late complication of diabetes and in those with microvascular disease. Nephropathy, neuropathy, and retinopathy are regularly present in patients with DD. An association with cardiovascular disease has also been identified, with one study showing 53% of non-insulin-dependent diabetes mellitus with DD had coexisting coronary artery disease (13).

 

PRESENTATION

 

DD initially presents with rounded, dull, red papules that progressively evolve over one-to-two weeks into well-circumscribed, atrophic, brown macules with a fine scale (figure 1). Normally after about eighteen to twenty-four months, lesions dissipate and leave behind an area of concavity and hyperpigmentation. At any time, different lesions can present at different stages of evolution. The lesions are normally distributed bilaterally and localized over bony prominences. The pretibial area is most commonly involved, although other bony prominences such as the forearms, lateral malleoli or thighs may also be involved. Aside from the aforementioned changes, patients are otherwise asymptomatic. DD is a clinical diagnosis that should not require a skin biopsy. Histologically, DD is rather nonspecific; it is characterized by lymphocytic infiltrates surrounding vasculature, engorged blood vessels in the papillary dermis, and dispersed hemosiderin deposits. Moreover, the histology varies based on the stage of the lesion. Immature lesions present with epidermal edema as opposed to epidermal atrophy which is representative of older lesions (14).

Figure 1. Diabetic Dermopathy

PATHOGENESIS

 

The origin of DD remains unclear, however, mild trauma to affected areas (15), hemosiderin and melanin deposition (16), microangiopathic changes (17), and destruction of subcutaneous nerves (18) have all been suggested.

 

TREATMENT

 

Treatment is typically avoided given the asymptomatic and self-resolving nature of DD as well as the ineffectiveness of available treatments. However, DD often occurs in the context of microvascular complications and neuropathies (12); hence, patients need to be examined and followed more rigorously for these complications. Although it is important to manage diabetes and its complications accordingly, there is no evidence that improved glycemic control alters the development of DD.

 

Diabetic Foot Syndrome

 

EPIDEMIOLOGY

 

Diabetic Foot Syndrome (DFS) encompasses the neuropathic and vasculopathic complications that develop in the feet of patients with diabetes. Although preventable, DFS is a significant cause of morbidity, mortality, hospitalization, and reduction in quality of life of patients with diabetes. The incidence and prevalence of DFS in patients with diabetes is 1% to 4% and 4% to 10%, respectively (19). Furthermore, DFS is slightly more prevalent in type 1 diabetes compared with type 2 diabetes (20). A more comprehensive review of diabetic foot syndrome can be found in The Diabetic Foot chapter of Endotext.

 

PRESENTATION

 

DFS presents initially with callosities and dry skin related to diabetic neuropathy. In later stages, chronic ulcers and a variety of other malformations of the feet develop. Between 15% and 25% of patients with diabetes will develop ulcers (21). Ulcers may be neuropathic, ischemic, or mixed. The most common type of ulcers are neuropathic ulcers, a painless ulceration resulting from peripheral neuropathy. Ulcers associated with peripheral vascular ischemia are painful but less common. Ulcers tend to occur in areas prone to trauma, classically presenting at the site of calluses or over bony prominences. It is common for ulcers to occur on the toes, forefoot, and ankles. Untreated ulcers usually heal within one year, however, fifty percent of patients with diabetes will have recurrence of the ulcer within three years (22). The skin of affected patients, especially in those with type 2 diabetes, is more prone to fungal infection and the toe webs are a common port of entry for fungi which can then infect and complicate ulcers (23). Secondary infection of ulcers is a serious complication that can result in gangrenous necrosis, osteomyelitis, and may even require lower extremity amputation. Another complication, diabetic neuro-osteoarthropathy (also known as Charcot foot), is an irreversible debilitating and deforming condition involving progressive destruction of weight-bearing bones and joints. Diabetic neuro-osteoarthropathy occurs most frequently in the feet and can result in collapse of the midfoot, referred to as “rocker-bottom foot.” Moreover, a reduction of the intrinsic muscle volume and thickening of the plantar aponeurosis can cause a muscular imbalance that produces a clawing deformation of the toes. An additional complication of diabetes and neuropathy involving the feet is erythromelalgia. Erythromelalgia presents with redness, warmth, and a burning pain involving the lower extremities, most often the feet. Symptoms may worsen in patients with erythromelalgia with exercise or heat exposure and may improve with cooling (24).

 

PATHOGENESIS

 

The pathogenesis of DFS involves a combination of inciting factors that coexist together: neuropathy (25), atherosclerosis (25), and impaired wound healing (26). In the setting of long-standing hyperglycemia, there is an increase in advanced glycosylation end products, proinflammatory factors, and oxidative stress which results in the demyelination of nerves and subsequent neuropathy (27,28). Single-cell RNA sequencing revealed that there is a unique subset of fibroblasts that overexpress factors associated with healing within the wound bed as opposed to the wound edge (28A). Additionally, wound healers demonstrate an increase in M1 macrophages as opposed to non-wound healers which have an increase in M2 macrophages. The effect on sensory and motor nerves, can blunt the perception of adverse stimuli and produce an altered gait, increasing the likelihood of developing foot ulcers and malformations. Also damage to autonomic nerve fibers causes a reduction in sweating which may leave skin in the lower extremity dehydrated and prone to fissures and secondary infection (29). In addition to neuropathy, accelerated arterial atherosclerosis can lead to peripheral ischemia and ulceration (30). It has been reported that diabetic patients with Charcot neuroarthropathy are associated with greater impairment of cutaneous microvascular reactivity when compared to non-complicated diabetic groups (30A). Finally, hyperglycemia impairs macrophage functionality as well as increases and prolongs the inflammatory response, slowing the healing of ulcers (31).

 

TREATMENT

 

Treatment should involve an interdisciplinary team-based approach with a focus on prevention and management of current ulcers. Prevention entails daily surveillance, appropriate foot hygiene, and proper footwear, walkers, or other devices to minimize and distribute pressure. An appropriate wound care program should be used to care for ongoing ulcers. Different classes of wound dressing should be considered based on the wound type. Hydrogels, hyperbaric oxygen therapy, topical growth factors, and biofabricated skin grafts are also available (19). The clinical presentation should indicate whether antibiotic therapy or wound debridement is necessary (19). In patients with chronic treatment resistant ulcers, underlying ischemia should be considered; these patients may require surgical revascularization or bypass.

 

Diabetic Thick Skin

 

Skin thickening is frequently observed in patients with diabetes. Affected areas of skin can appear thickened, waxy, or edematous. These patients are often asymptomatic but can have a reduction in sensation and pain. Although different parts of the body can be involved, the hands and feet are most frequently involved. Ultrasound evaluation of the skin can be diagnostic and exhibit thickened skin. Subclinical generalized skin thickening is the most common type of skin thickening. Diabetic thick skin may represent another manifestation of scleroderma-like skin changes or limited joint mobility, which are each described in more detail below.

 

Scleroderma-Like Skin Changes

 

EPIDEMIOLOGY

 

Scleroderma-like skin changes are a distinct and easily overlooked group of findings that are commonly observed in patients with diabetes. Ten to fifty percent of patients with diabetes present with the associated skin findings (32). Scleroderma-like skin changes occurs more commonly in those with type 1 diabetes and in those with longstanding disease (33). There is no known variation in prevalence between males and females, or between racial groups.

 

PRESENTATION

 

Scleroderma-like skin changes develop slowly and present with painless, indurated, occasionally waxy appearing, thickened skin. These changes occur symmetrically and bilaterally in acral areas. In patients with scleroderma-like skin changes the acral areas are involved, specifically the dorsum of the fingers (sclerodactyly), proximal interphalangeal, and metacarpophalangeal joints. Severe disease may extend centrally from the hands to the arms or back. A small number of patients with diabetes may develop more extensive disease, which presents earlier and with truncal involvement. The risk of developing nephropathy and retinopathy is increased in those with scleroderma-like skin changes who also have type 1 diabetes (33,34). The aforementioned symptoms are also associated with diabetic hand syndrome which may present with limited joint mobility, palmar fibromatosis (Dupuytren's contracture), and stenosing tenosynovitis (“trigger finger”) (35). The physical exam finding known as the “prayer sign” (inability to flushly press palmar surfaces on each hand together) may be present in patients with diabetic hand syndrome and scleroderma-like skin changes (36). On histology, scleroderma-like skin changes reveal thickening of the dermis, minimal-to-absent mucin, and increased interlinking of collagen. Although on physical exam scleroderma may be difficult to distinguish from these skin changes, scleroderma-like skin changes are not associated with atrophy of the dermis, Raynaud’s syndrome, pain, or telangiectasias.

 

PATHOGENESIS

 

Although not fully understood, the pathogenesis is believed to involve the strengthening of collagen as a result of reactions associated with advanced glycosylation end products or a buildup of sugar alcohols in the upper dermis (37,38).

 

TREATMENT

 

Scleroderma-like skin changes is a chronic condition that is also associated with joint and microvascular complication. Therapeutic options are extremely limited. One observational report has suggested that very tight blood sugar control may result in the narrowing of thickened skin (39). In addition, aldose reductase inhibitors, which limit increases in sugar alcohols, may be efficacious (38). In patients with restricted ranges of motions, physical therapy can help to maintain and improve joint mobility.

 

Limited Joint Mobility

 

Limited Joint Mobility (LJM), also known as diabetic cheiroarthropathy, is a relatively common complication of long-standing diabetes mellitus. The majority of patients with LJM also present with scleroderma-like skin changes (38,40). The prevalence of LJM is 4% to 26% in patients without diabetes and 8% to 58% in patients with diabetes (41). LJM presents with progressive flexed contractures and hindered joint extension, most commonly involving the metacarpophalangeal and interphalangeal joints of the hand. The earliest changes often begin in the joints of the fifth finger before then spreading to involve the other joints of the hand (38). Patients may present with an inability to flushly press the palmar surfaces of each of their hands together (“prayer sign”) (figure 2) or against the surface of a table when their forearms are perpendicular to the surface of the table (“tabletop sign”) (42). These changes occur as a result of periarticular enlargement of connective tissue. The pathogenesis likely involves hyperglycemia induced formation of advanced glycation end-products, which accumulate to promote inflammation and the formation of stiffening cross-links between collagen (43). LJM is strongly associated with microvascular and macrovascular changes and diagnosis of LJM should prompt a workup for related sequela (44). Patients with LJM may also be at increased risk for falls (45). There are no curative treatments. Symptomatic patients may benefit from non-steroidal anti-inflammatory drugs or targeted injection of corticosteroids (43). LJM is best managed with improved glycemic control (46), as well as, regular stretching to maintain and minimize further limitations in joint mobility.

 

Figure 2. Limited Joint Mobility

Scleredema Diabetocorum

 

EPIDEMIOLOGY

 

Scleredema diabeticorum is a chronic and slowly progressive sclerotic skin disorder that is often seen in the context of diabetes. Whereas 2.5% to 14% of all patients with diabetes have scleredema, over 50% of those with scleredema present with concomitant diabetes (47). Scleredema has a proclivity for those with a long history of diabetes. It remains unclear whether there is a predilection for scleredema in those with type 1 diabetes (48) compared to those with type 2 diabetes (48). Women are affected more often than men (49). Although all ages are affected, scleredema occurs more frequently in those over the age of twenty (48,49).

 

PRESENTATION

 

Scleredema presents with gradually worsening indurated and thickened skin. These skin changes occur symmetrically and diffusely. The most commonly involved areas are the upper back, shoulders, and back of the neck. The face, chest, abdomen, buttocks, and thighs may also be involved; however, the distal extremities are classically spared. The affected areas are normally asymptomatic but there can be reduced sensation. Patients with severe longstanding disease may develop a reduced range of motion, most often affecting the trunk. In extreme cases, this can lead to restrictive respiratory problems. A full thickness skin biopsy may be useful in supporting a clinical presentation. The histology of scleredema displays increased collagen and a thickened reticular dermis, with a surrounding mucinous infiltrate, without edema or sclerosis.

 

PATHOGENESIS

 

Although many theories center on abnormalities in collagen, there is no a consensus regarding the pathogenesis of scleredema. The pathogenesis of scleredema may involve an interplay between non-enzymatic glycosylation of collagen, increased fibroblast production of collagen, or decreases in collagen breakdown (50,51).

 

TREATMENT

 

Scleredema diabeticorum is normally unresolving and slowly progressive over years. Improved glycemic control may be an important means of prevention but evidence has not shown clinical improvements in those already affected by scleredema diabeticorum. A variety of therapeutic options have been proposed with variable efficacy. Some of these therapies include immunosuppressants, corticosteroids, intravenous immunoglobulin, and electron-beam therapy (52). Phototherapy with UVA1 or PUVA may be effective in those that are severely affected (52). Independent of other treatments, physical therapy is an important therapeutic modality for patients with scleredema and reduced mobility (53).

 

Necrobiosis Lipoidica

 

EPIDEMIOLOGY

 

Necrobiosis lipoidica (NL) is a rare chronic granulomatous dermatologic disease that is seen most frequently in patients with diabetes. Although nearly one in four patients presenting with NL will also have diabetes, less than 1% of patients with diabetes will develop NL (54). For unknown reasons, NL expresses a strong predilection for women compared to men (55). NL generally occurs in type 1 diabetes during the third decade of life, as opposed to type 2 diabetes in which it commonly presents in the fourth or fifth decades of life (54). The majority of cases of NL presents years after a diagnosis of diabetes mellitus; however, 14% to 24% of cases of NL may occur prior to or at the time of diagnosis (56). One study evaluating comorbidities and diabetic complications in patients with NL found high rates of smoking, hypertension, hyperlipidemia, obesity, coronary artery disease, myocardial infarction, thyroid disease, poor kidney function, and poor glucose control (56A). The highest comorbidity rates in patients with NL were patients with type 2 diabetes.

 

PRESENTATION

 

NL begins as a single or group of firm well-demarcated rounded erythematous papules (figure 3). The papules then expand and aggregate into plaques characterized by circumferential red-brown borders and a firm yellow-brown waxen atrophic center containing telangiectasias. NL occurs bilaterally and exhibits Koebnerization. Lesions are almost always found on the pretibial areas of the lower extremities. Additional involvement of the forearm, scalp, distal upper extremities, face, or abdomen may be present on occasion, and the heel of the foot or glans penis even more infrequently. If left untreated, only about 15% of lesions will resolve within twelve years. Despite the pronounced appearance of the lesions, NL is often asymptomatic. However, there may be pruritus and hypoesthesia of affected areas, and pain may be present in the context of ulceration. Ulceration occurs in about one-third of lesions, and has been associated with secondary infections and squamous cell carcinoma. The histology of NL primarily involves the dermis and is marked by palisading granulomatous inflammation, necrobiotic collagen, a mixed inflammatory infiltrate, blood vessel wall thickening, and reduced mucin.

Figure 3. Necrobiosis Lipoidica

PATHOGENESIS

 

The pathogenesis of NL is not well understood. The relationship between diabetes and NL has led some to theorize that diabetes-related microangiopathy is related to the development of NL (54). Other theories focus on irregularities in collagen, autoimmune disease, neutrophil chemotaxis, or blood vessels (57).

 

TREATMENT

 

NL is a chronic, disfiguring condition that can be debilitating for patients and difficult for clinicians to manage. Differing degrees of success have been reported with a variety of treatments; however, the majority of such reports are limited by inconsistent treatment responses in patients and a lack of large controlled studies. Corticosteroids are often used in the management of NL and may be administered topically, intralesionally, or orally. Corticosteroids can be used to manage active lesions, but is best not used in areas that are atrophic. Success has also been reported with calcineurin inhibitors (e.g., cyclosporine), anti-tumor necrosis factor inhibitors (e.g., infliximab), pentoxifylline, antimalarials (e.g., hydroxychloroquine), PUVA, granulocyte colony stimulating factor, dipyridamole, and low-dose aspirin (54). Appropriate wound care is important for ulcerated lesions; this often includes topical antibiotics, protecting areas vulnerable to injury, emollients, and compression bandaging. Surgical excision of ulcers typically has poor results. Some ulcerated lesion may improve with split-skin grafting. Although still recommended, improved control of diabetes has not been found to lead to an improvement in skin lesions. Patients with newly diagnosed NL should be screened for hypertension, hyperlipidemia, and thyroid disease (56A). 

Bullosis Diabeticorum

 

EPIDEMIOLOGY

 

Bullosis diabeticorum (BD) is an uncommon eruptive blistering condition that presents in those with diabetes mellitus. Although BD can occasionally present in early-diabetes (58), it often occurs in long-standing diabetes along with other complications such as neuropathy, nephropathy, and retinopathy. In the United States, the prevalence of BD is around 0.5% amongst patients with diabetes and is believed to be higher in those with type 1 diabetes (13). BD is significantly more common in male patients than in female patients (59). The average age of onset is between 50 and 70 years of age (59).

 

PRESENTATION

 

BD presents at sites of previously healthy-appearing skin with the abrupt onset of one or more non-erythematous, firm, sterile bullae. Shortly after forming, bullae increase in size and become more flaccid, ranging in size from about 0.5 cm to 5 cm. Bullae frequently present bilaterally involving the acral areas of the lower extremities. However, involvement of the upper extremities and even more rarely the trunk can be seen. The bullae and the adjacent areas are nontender. BD often presents acutely, classically overnight, with no history of trauma to the affected area. Generally, the bullae heal within two to six weeks, but then commonly reoccur. Histological findings are often non-specific but are useful in distinguishing BD from other bullous diseases. Histology, typically shows an intraepidermal or subepidermal blister, spongiosis, no acantholysis, minimal inflammatory infiltrate, and normal immunofluorescence.

 

PATHOGENESIS

 

There is an incomplete understanding of the underlying pathogenesis of BD and no consensus regarding a leading theory. Various mechanisms have been proposed, some of which focus on autoimmune processes, exposure to ultraviolet light, variations in blood glucose, neuropathy, or changes in microvasculature (60).

 

TREATMENT

 

BD resolve without treatment and are therefore managed by avoiding secondary infection and the corresponding sequelae (e.g., necrosis, osteomyelitis). This involves protection of the affected skin, leaving blisters intact (except for large blisters, which may be aspirated to prevent rupture), and monitoring for infection. Topical antibiotics are not necessary unless specifically indicated, such as with secondary infection or positive culture.

NONSPECIFIC DERMATOLOGIC SIGNS AND SYMPTOMS

Ichthyosiform Changes of the Shins

 

Ichthyosiform changes of the shins presents with large bilateral areas of dryness and scaling (sometimes described as “fish scale” skin) (figure 4). Although cutaneous changes may occur on the hands or feet, the anterior shin is most classically involved. These cutaneous changes are related to rapid skin aging and adhesion defects in the stratum corneum (61). The prevalence of ichthyosiform changes of the shins in those with type 1 diabetes has been reported to be between 22% to 48% (33,62). These changes present relatively early in the disease course of diabetes. There is no known difference in prevalence between males and females (33). The development of ichthyosiform changes of the shins is related to production of advanced glycosylation end products and microangiopathic changes. Treatment is limited but topical emollients or keratolytic agents may be beneficial (61).

Figure 4. Acquired ichthyosiform changes

Xerosis

 

Xerosis is one of the most common skin presentations in patients with diabetes and has been reported to be present in as many as 40% of patients with diabetes (63). Xerosis refers to skin that is abnormally dry. Affected skin may present with scaling, cracks, or a rough texture. These skin changes are most frequently located on the feet of patients with diabetes. It has been reported that diabetic patients that are obese will experience more severe hypohidrosis of the feet (63A). In patients with diabetes, xerosis occurs often in the context of microvascular complications (40). To avoid complications such as fissures and secondary infections, xerosis can be managed with emollients like ammonium lactate (64).

 

Acquired Perforating Dermatosis

 

EPIDEMIOLOGY

 

Perforating dermatoses refers to a broad group of chronic skin disorders characterized by a loss of dermal connective tissue. A subset of perforating dermatoses, known as acquired perforating dermatoses (APD), encompasses those perforating dermatoses that are associated with systemic diseases. Although APD may be seen with any systemic diseases, it is classically observed in patients with chronic renal failure or long-standing diabetes (65). APD occurs most often in adulthood in patients between the ages of 30 and 90 years of age (65,66). The prevalence of APD is unknown. It is estimated that of those diagnosed with APD about 15% also have diabetes mellitus (67). In a review, 4.5% to 10% of patients with chronic renal failure presented with concurrent APD (68,69).

 

PRESENTATION

 

APD presents as groups of hyperkeratotic umbilicated-nodules and papules with centralized keratin plugs. The lesions undergo Koebnerization and hence the extensor surfaces of the arms and more commonly the legs are often involved; eruptions also occur frequently on the trunk. However, lesions can develop anywhere on the body. Lesions are extremely pruritic and are aggravated by excoriation. Eruptions may improve after a few months but an area of hyperpigmentation typically remains. Histologically, perforating dermatoses are characterized by a lymphocytic infiltrate, an absence or degeneration of dermal connective tissue components (e.g., collagen, elastic fibers), and transepidermal extrusion of keratotic material.

 

PATHOGENESIS

 

The underlying pathogenesis is disputed and not fully understood. It has been suggested that repetitive superficial trauma from chronic scratching may induce epidermal or dermal derangements (70). The glycosylation of microvasculature or dermal components has been suggested as well. Other hypotheses have implicated additional metabolic disturbances, or the accumulation of unknown immunogenic substances that are not eliminated by dialysis (65). APD is also considered a form of prurigo nodularis (70A).

 

TREATMENT

 

APD can be challenging to treat and many of the interventions have variable efficacy. Minimizing scratching and other traumas to involved areas can allow lesions to resolve over a period of months. This is best achieved with symptomatic relief of pruritus. Individual lesions can be managed with topical agents such as keratolytics (e.g., 5% to 7% salicylic acid), retinoids (e.g., 0.01% to 0.1% tretinoin), or high-potency steroids (71). Refractory lesions may respond to intralesional steroid injections or cryotherapy (71). A common initial approach is a topical steroid in combination with emollients and an oral antihistamine. Generalized symptoms may improve with systemic therapy with oral retinoids, psoralen plus UVA light (PUVA), allopurinol (100 mg daily for 2 to 4 months), or oral antibiotics (doxycycline or clindamycin) (72). Additionally, as APD is a form of prurigo nodularis, the use of immunomodulating agents such as dupilumab may be effective in treating the condition. There is evidence of dupilumab monotherapy effectively treating certain forms of APD (72A). Nevertheless, effective management of the underlying systemic disease is fundamental to the treatment of APD. In those with diabetes, APD is unlikely to improve without improved blood sugar control. Moreover, dialysis does not reduce symptoms; however, renal transplantation can result in the improvement and resolution of cutaneous lesions.

 

Eruptive Xanthomas

 

EPIDEMIOLOGY

 

Eruptive xanthomas (EX) is a clinical presentation of hypertriglyceridemia, generally associated with serum triglycerides above 2,000 mg/dL (73). However, in patients with diabetes, lower levels of triglycerides may be associated with EX. The prevalence of EX is around one percent in type 1 diabetes and two percent in type 2 diabetes (74,75). Serum lipid abnormalities are present in about seventy-five percent of patients with diabetes (76).

 

PRESENTATION

 

EX has been reported as the first presenting sign of diabetes mellitus, granting it can present at any time in the disease course. EX presents as eruptions of clusters of glossy pink-to-yellow papules, ranging in diameter from 1 mm to 4 mm, overlying an erythematous area (figure 5). The lesions can be found on extensor surfaces of the extremities, the buttocks, and in areas susceptible in Koebnerization. EX is usually asymptomatic but may be pruritic or tender. The histology reveals a mixed inflammatory infiltrate of the dermis which includes triglyceride containing macrophages, also referred to as foam cells.

Figure 5. Eruptive Xanthomas

PATHOGENESIS

 

Lipoprotein lipase, a key enzyme in the metabolism of triglyceride rich lipoproteins, is stimulated by insulin. In an insulin deficient state, such as poorly controlled diabetes, there is decreased lipoprotein lipase activity resulting in the accumulation of chylomicrons and other triglyceride rich lipoproteins (77). Increased levels of these substances are scavenged by macrophages (78). These lipid-laden macrophages then collect in the dermis of the skin where they can lead to eruptive xanthomas.

 

TREATMENT

 

EX can resolve with improved glycemic control and a reduction in serum triglyceride levels (79). This may be achieved with fibrates or omega-3-fatty acids in addition to an appropriate insulin regimen (80). A more comprehensive review of the treatment of hypertriglyceridemia can be found in the Triglyceride Lowering Drugs section of Endotext.

 

Acrochordons

 

Acrochordons (also known as soft benign fibromas, fibroepithlial polyps, or skin tags) are benign, soft, pedunculated growths that vary in size and can occur singularly or in groups (figure 6). The neck, axilla, and periorbital area, are most frequently involved, although other intertriginous areas can also be affected. Skin tags are common in the general population, but are more prevalent in those with increased weight or age, and in women. It has been reported that as many as three out of four patients presenting with acrochordons also have diabetes mellitus (81). Patients with acanthosis nigricans may have acrochordons overlying the affected areas of skin. Although disputed, some studies have suggested that the amount of skin tags on an individual may correspond with an individual's risk of diabetes or insulin resistance (82). Excision or cryotherapy is not medically indicated but may be considered in those with symptomatic or cosmetically displeasing lesions.

Figure 6. Acrochordons

Diabetes-Associated Pruritus

 

Diabetes can be associated with pruritus, more often localized than generalized. Affected areas can include the scalp, ankles, feet, trunk, or genitalia (83,84). Pruritus is more likely in patients with diabetes who have dry skin or diabetic neuropathy. Specifically, for type 2 diabetes, risk factors for pruritus were identified to be age, duration of disease, diabetic peripheral neuropathy, diabetic retinopathy, diabetic chronic kidney disease, and fasting plasma glucose levels (84A). Involvement of the genitalia or intertriginous areas may occur in the setting of infection (e.g., candidiasis). Treatments include topical capsaicin, topical ketamine-amitriptyline-lidocaine, oral anticonvulsants (e.g., gabapentin or pregabalin), and, in the case of candida infection, antifungals.

 

Huntley’s Papules (Finger Pebbles)

 

Huntley’s papules, also known as finger pebbles, are a benign cutaneous finding affecting the hands. Patients present with clusters of non-erythematous, asymptomatic, small papules on the dorsal surface of the hand, specifically affecting the metacarpophalangeal joints and periungual areas. The clusters of small papules can develop into coalescent plaques. Other associated cutaneous findings include hypopigmentation and induration of the skin. Huntley’s papules are strongly associated with type 2 diabetes and may be an early sign of diabetic thick skin (85,86). Topical therapies are usually ineffective; however, patients suffering from excessive dryness of the skin may benefit from 12% ammonium lactate cream (87).

 

Keratosis Pilaris

 

Keratosis pilaris is a very common benign keratotic disorder. Patients with keratosis pilaris classically present with areas of keratotic perifollicular papules with surrounding erythema or hyperpigmentation (figure 7). The posterior surfaces of the upper arms are often affected but involvement of the thighs, face, and buttocks can also be seen. Compared to the general population, keratosis pilaris occurs more frequently and with more extensive involvement of the skin in those with diabetes (33,62). Keratosis pilaris can be treated with various topical therapies, including salicylic acid, moisturizers, and emollients.

Figure 7. Keratosis Pilaris

Pigmented Purpuric Dermatoses

 

Pigmented purpuric dermatoses (also known as pigmented purpura) are associated with diabetes, more often in the elderly, and frequently coexists with diabetic dermopathy (88,89). Pigmented purpura presents with non-blanching copper-colored patches involving the pretibial areas of the legs or the dorsum of the feet. The lesions are usually asymptomatic but may be pruritic. Pigmented purpuric dermatoses occur more often in late-stage diabetes in patients with nephropathy and retinopathy as a result of microangiopathic damage to capillaries and sequential erythrocyte deposition (90).

 

Palmar Erythema

 

Palmar erythema is a benign finding that presents with symmetric redness and warmth involving the palms. The erythema is asymptomatic and often most heavily affects the hypothenar and thenar eminences of the palms. The microvascular complications of diabetes are thought to be involved in the pathogenesis of palmar erythema (91). Although diabetes associated palmar erythema is distinct from physiologic mottled skin, it is similar to other types of palmar erythema such as those related to pregnancy and rheumatoid arthritis.

 

Periungual Telangiectasias

 

As many as one in every two patients with diabetes are affected by periungual telangiectasias (92). Periungual telangiectasias presents asymptomatically with erythema and telangiectasias surrounding the proximal nail folds (71). Such findings may occur in association with “ragged” cuticles and fingertip tenderness. The cutaneous findings are due to venous capillary dilatation that occurs secondary to diabetic microangiopathy. Capillary abnormalities, such as venous capillary tortuosity, may differ and can represent an early manifestation of diabetes-related microangiopathy (93).

 

Rubeosis Faciei

 

Rubeosis faciei is a benign finding present in about 7% of patients with diabetes; however, in hospitalized patients, the prevalence may exceed 50% (94). Rubeosis faciei presents with chronic erythema of the face or neck. Telangiectasias may also be visible. The flushed appearance is often more prominent in those with lighter colored skin. The flushed appearance is thought to occur secondary to small vessel dilation and microangiopathic changes. Complications of diabetes mellitus, such as retinopathy, neuropathy, and nephropathy are also associated with rubeosis faciei (90). Facial erythema may improve with better glycemic control and reduction of caffeine or alcohol intake.

 

Yellow Skin and Nails

 

It is common for patients with diabetes, particularly elderly patients with type 2 diabetes, to present with asymptomatic yellow discolorations of their skin or fingernails. These benign changes commonly involve the palms, soles, face, or the distal nail of the first toe. The accumulation of various substances (e.g., carotene, glycosylated proteins) in patients with diabetes may be responsible for the changes in complexion; however, the pathogenesis remains controversial (95).

 

Onychocryptosis

 

Onychocryptosis, or ingrown toenails, have been reported in patients with diabetes, specifically type 2 diabetes (95A). The great toes are most affected. It is hypothesized that this nail change can occur in diabetic patients because onychocryptosis is correlated with increased body mass index, trauma, weak vascular supply, nail plate dysfunction, and subungual hyperkeratosis.

 

DERMATOLOGIC DISEASES ASSOCIATED WITH DIABETES

 

Generalized Granuloma Annulare

 

EPIDEMIOLOGY

 

Although various forms of granuloma annulare exist, only generalized granuloma annulare (GGA) is thought to be associated with diabetes. It is estimated that between ten and fifteen percent of cases of GGA occur in patients with diabetes (96). Meanwhile, less than one percent of patients with diabetes present with GGA. GGA occurs around the average age of 50 years. It occurs more frequently in women than in men, and in those with type 1 diabetes (97).

 

PRESENTATION

 

GGA initially presents with groups of skin-colored or reddish, firm papules which slowly grow and centrally involute to then form hypo- or hyper-pigmented annular rings with elevated circumferential borders. The lesions can range in size from 0.5 cm to 5.0 cm. The trunk and extremities are classically involved in a bilateral distribution. GGA is normally asymptomatic but can present with pruritus. The histology shows dermal granulomatous inflammation surrounding foci of necrotic collagen and mucin. Necrobiosis lipoidica can present similarly to GGA; GGA is distinguished from necrobiosis lipoidica by its red color, the absence of an atrophic epidermis, and on histopathology: the presence of mucin and lack of plasma cells.

 

PATHOGENESIS

 

The pathogenesis of GGA is incompletely understood. It is believed to involve an unknown stimulus that leads to the activation of lymphocytes through a delayed-type hypersensitivity reaction, ultimately initiating a proinflammatory cascade and granuloma formation (98).

 

TREATMENT

 

GGA has a prolonged often unresolving disease course and multiple treatments have been suggested to better manage GGA. However, much of the information stems from small studies and case reports. Antimalarials, retinoids, corticosteroids, dapsone, cyclosporine, PUVA, and calcineurin inhibitors have been suggested as therapies (98).

 

Psoriasis

 

Psoriasis is a chronic immune-mediated inflammatory disorder that may present with a variety of symptoms, including erythematous, indurated, and scaly areas of skin. Psoriasis has been found to be associated with a variety of risk factors, such as hypertension and metabolic syndrome, that increase the likelihood of cardiovascular disease. The development of diabetes mellitus, an additional cardiovascular risk factor, has been strongly associated with psoriasis (99). In particular, younger patients and those with severe psoriasis may be more likely to develop diabetes in the future (99)

 

Lichen Planus

 

Lichen planus is a mucocutaneous inflammatory disorder characterized by firm, erythematous, polygonal, pruritic, papules. These papules classically involve the wrists or ankles, although the trunk, back, and thighs can also be affected. A number of studies have cited an association between lichen planus and abnormalities in glucose tolerance testing. Approximately one in four patients with lichen planus have diabetes mellitus (100). Although the association is contested, it has been reported that patients with diabetes may also be more likely to develop oral lichen planus (101).

 

Vitiligo

 

Vitiligo is an acquired autoimmune disorder involving melanocyte destruction. Patients with vitiligo present with scattered well-demarcated areas of depigmentation that can occur anywhere on the body, but frequently involves the acral surfaces and the face. Whereas about 1% of the general population is affected by vitiligo, vitiligo is much more prevalent in those with diabetes mellitus. Vitiligo occurs more frequently in women and is also more common in type 1 than in type 2 diabetes mellitus (96,98). Coinciding vitiligo and type 1 diabetes mellitus may be associated with endocrine autoimmune abnormalities of the gastric parietal cells, adrenal, or thyroid (102). A more comprehensive review of polyglandular autoimmune disorders can be found in the Autoimmune Polyglandular Syndromes section of Endotext.

 

Hidradenitis Supparativa

 

Hidradenitis supparativa (HS) is a chronic inflammatory condition characterized by inflamed nodules and abscesses located in intreginious areas such as the axilla or groin. These lesions are often painful and malodorous. HS is frequently complicated by sinus formation and the development of disfiguring scars. HS occurs more often in women than men and usually presents in patients beginning in their twenties (103). Compared to the general population, diabetes mellitus is three-times more common in patients with HS (104). It is recommended that patients with HS be screened for diabetes mellitus. There is no standardized approach to the treatment of HS, although some benefits have been reported with the use of antibiotics, retinoids, antiandrogens, or immunomodulators such as tumor necrosis factor (TNF) inhibitors (105).

 

Glucagonoma

 

Glucagonoma is a rare neuroendocrine tumor that most frequently affects patients in their sixth decade of life (106). Patients with glucagonoma may present with a variety of non-specific symptoms. However, necrolytic migratory erythema (NME) is classically associated with glucagonoma and presents in 70% to 83% of patients (106) (107). NME is characterized by erythematous erosive crusted or vesicular eruptions of papules or plaques with irregular borders. The lesions may become bullous or blistered, and may be painful or pruritic. The abdomen, groin, genitals, or buttocks are frequently involved, although cheilitis or glossitis may also be present. Biopsy at the edge of the lesion may demonstrate epidermal pallor, necrolytic edema, and a perivascular inflammatory infiltrate (108). Patients with glucagonoma may also present with diabetes mellitus. In patients with glucagonoma, diabetes mellitus frequently presents prior to NME (107). Approximately 20% to 40% of patients will present with diabetes mellitus before the diagnosis of glucagonoma (107,109). Of those patients diagnosed with glucagonoma but not diabetes mellitus, 76% to 94% will eventually develop diabetes mellitus (110). A more comprehensive review of glucagonoma can be found in the Glucagonoma section of Endotext.

 

Skin Infections

 

The prevalence of cutaneous infections in patients with diabetes is about one in every five patients (111). Compared with the general population, patients with diabetes mellitus are more susceptible to infections and more prone to repeated infections. A variety of factors are believed to be involved in the vulnerability to infection in patients with uncontrolled diabetes, some of these factors include angiopathy, neuropathy, hindrance of the anti-oxidant system, abnormalities in leukocyte adherence, chemotaxis, and phagocytosis, as well as, a glucose-rich environment facilitates the growth of pathogens.

 

BACTERIAL

 

Erysipelas and cellulitis are cutaneous infections that occur frequently in patients with diabetes. Erysipelas presents with pain and well-demarcated superficial erythema. Cellulitis is a deeper cutaneous infection that presents with pain and poorly-demarcated erythema. Folliculitis is common among patients with diabetes, and is characterized by inflamed, perifollicular, papules and pustules. Treatment for the aforementioned conditions depends on the severity of the infection. Uncomplicated cellulitis and erysipelas are typically treated empirically with oral antibiotics, whereas uncomplicated folliculitis may be managed with topical antibiotics. Colonization with methicillin-resistant Staphylococcus aureus (MRSA) is not uncommon among patients with diabetes (112); however, it is debated as to whether or not colonized patients are predisposed to increased complications (113) such as bullous erysipelas, carbuncles, or perifollicular abscesses. Regardless, it is important that appropriate precautions are taken in these patients and that antibiotics are selected that account for antimicrobial resistance.

 

Infection of the foot is the most common type of soft tissue infection in patients with diabetes. If not managed properly, diabetic foot infections can become severe, possibly leading to sepsis, amputation, or even death. Although less severe, the areas between the toes and the toenails are also frequently infected in patients with diabetes. Infections can stem from monomicrobial or polymicrobial etiologies. Staphylococcal infections are the most common (114), although complications with infection by Pseudomonas aeruginosa are also common (115). Pseudomonal infection of the toenail may present with a green discoloration, which may become more pronounced with the use of a Wood’s light. Treatment frequently requires coordination of care from multiple medical providers. Topical or oral antibiotics and surgical debridement may be indicated depending on the severity of the infection.

 

Necrotizing fasciitis is an acute life-threatening infection of the skin and the underlying tissue. Those with poorly controlled diabetes are at an increased risk for necrotizing fasciitis. Necrotizing fasciitis presents early with erythema, induration, and tenderness which may then progress within days to hemorrhagic bullous. Patients will classically present with severe pain out of proportion to their presentation on physical exam. Palpation of the affected area often illicit crepitus. Involvement can occur on any part of the body but normally occurs in a single area, most commonly affecting the lower extremities. Fournier’s gangrene refers to necrotizing fasciitis of the perineum or genitals, often involving the scrotum and spreading rapidly to adjacent tissues. The infection in patients with diabetes is most often polymicrobial. Complications of necrotizing fasciitis include thrombosis, gangrenous necrosis, sepsis, and organ failure. Necrotizing fasciitis has a mortality rate of around twenty percent (116). In addition, those patients with diabetes and necrotizing fasciitis are more likely to require amputation during their treatment (117). Treatment is emergent and includes extensive surgical debridement and broad-spectrum antibiotics.

 

Erythrasma is a chronic asymptomatic cutaneous infection, most often attributed to Corynebacterium minutissiumum. Diabetes mellitus, as well as obesity and older age are associated with erythrasma. Erythrasma presents with non-pruritic non-tender clearly demarcated red-brown finely scaled patches or plaques. These lesions are commonly located in intriginuous areas such as the axilla or groin. Given the appearance and location, erythrasma can be easily mistaken for tinea or Candidia infection; in such cases, the presence of coral-red fluorescence under a Wood’s light can confirm the diagnosis of erythrasma. Treatment options include topical erythromycin or clindamycin, Whitfield’s ointment, and sodium fusidate ointment. More generalized erythrasma may respond better to oral erythromycin.

 

Malignant otitis externa is a rare but serious infection of the external auditory canal that occurs most often in those with a suppressed immune system, diabetes mellitus, or of older age. Malignant otitis externa develops as a complication of otitis externa and is associated with infection by Pseudomonas aeruginosa. Patients with malignant otitis externa present with severe otalgia and purulent otorrhea. The infection can spread to nearby structures and cause complications such as chondritis, osteomyelitis, meningitis, or cerebritis. If untreated, malignant otitis externa has a mortality rate of about 50%; however, with aggressive treatment the mortality rate can been reduced to 10% to 20% (118). Treatment involves long-term systemic antibiotics with appropriate pseudomonal coverage, hyperbaric oxygen, and possibly surgical debridement.

 

FUNGAL

 

Candidiasis is a frequent presentation in patients with diabetes. Moreover, asymptomatic patients presenting with recurrent candidiasis should be evaluated for diabetes mellitus. Elevated salivary glucose concentrations (119) and elevated skin surface pH in the intertriginous regions of patients with diabetes (120) may promote an environment in which candida can thrive. Candida infection can involve the mucosa (e.g., thrush, vulvovaginitis), intertriginous areas (e.g., intertrigo, erosion interdigital, balanitis), or nails (e.g., paronychia). Mucosal involvement presents with pruritus, erythema, and white plaques which can be removed when scraped. Intertriginous Candida infections may be pruritic or painful and present with red macerated, fissured plaques with satellite vesciulopustules. Involvement of the nails may present with periungual inflammation or superficial white spots. Onchyomycosis may be due to dermatophytes (discussed below) or Candidal infection. Onchomycosis, characterized by subungual hyperkeratosis and oncholysis, is present in nearly one in two patients with type 2 diabetes mellitus. Candidiasis is treated with topical or oral antifungal agents. Patients also benefit from improved glycemic control and by keeping the affected areas dry.

 

Although it remains controversial, dermatophyte infections appear to be more prevalent among patients with diabetes (121-123). Various regions of the body may be affected but tinea pedis (foot) is the most common dermatophyte infection effecting patients; it presents with pruritus or pain and erythematous keratotic or bullous lesions. Relatively benign dermatophyte infections like tinea pedis can lead to serious sequela, such as secondary bacterial infection, fungemia, or sepsis, in patients with diabetes if not treated hastily. Patients with diabetic neuropathy may be especially vulnerable (124). Treatment may include topical or systemic antifungal medications depending on the severity.

 

Mucormycosis is a serious infection that is associated with type 1 diabetes mellitus, particularly common in those who develop diabetic ketoacidosis. A variety of factors including hyperglycemia and a lower pH, create an environment in which Rhizopus oryzae, a common pathogen responsible for mucormycosis, can prosper. Mucormycosis may present in different ways. Rhino-orbital-cerebral mucormycosis is the most common presentation; it develops quickly and presents with acute sinusitis, headache, facial edema, and tissue necrosis. The infection may worsen to cause extensive necrosis and thrombosis of nearby structures such as the eye. Mucormycosis should be treated urgently with surgical debridement and intravenous amphotericin B. When it is not suitable to administer amphotericin B in patients, the alternative use of new triazoles, posaconazole and isavuconazole, may be beneficial treatments (124A).

 

Lastly, abnormal toe web findings (e.g., maceration, scale, or erythema) may be an early marker of irregularities in glucose metabolism and of undiagnosed diabetes mellitus (125). Additionally, such findings may be a sign of epidermal barrier disruption, a precursor of infection (125).

 

CUTANEOUS CHANGES ASSOCIATED WITH DIABETES MEDICATIONS

 

Insulin

 

A number of localized changes are associated with the subcutaneous injection of insulin. The most common local adverse effect is lipohypertrophy, which affects less than thirty percent of patients with diabetes that use insulin (126,127). Lipohypertrophy is characterized by localized adipocyte hypertrophy and presents with soft dermal nodules at injection sites. Continued injection of insulin at sites of lipohypertrophy can result in delayed systemic insulin absorption and capricious glycemic control. With avoidance of subcutaneous insulin at affected sites, lipohypertrophy normally improves over the course of a few months.

 

Furthermore, lipoatrophy is an uncommon cutaneous finding which occurred more frequently prior to the introduction of modern purified forms of insulin. Lipoatrophy presents at insulin injection sites over a period of months with round concave areas of adipose tissue atrophy. Allergic reactions to the injection of insulin may be immediate (within one hour) or delayed (within one day) and can present with localized or systemic symptoms. These reactions may be due to a type one hypersensitivity reaction to insulin or certain additives. However, allergic reactions to subcutaneous insulin are rare, with systemic allergic reactions occurring in only 0.01% of patients (126). Other cutaneous changes at areas of injection include the development of pruritus, induration, erythema, nodular amyloidosis, or calcification.

 

Oral Medications

 

Oral hypoglycemic agents may cause a number of different cutaneous adverse effects such as erythema multiforme or urticaria. DPP-IV inhibitors, such as vildagliptin, can be associated with inflamed blistering skin lesions, including bullous pemphigoid and Stevens-Johnson syndrome, as well as, angioedema (128,129). Allergic skin and photosensitivity reactions may occur with sulfonylureas (130). The sulfonylureas, chlorpropamide and tolbutamide, are associated with the development of a maculopapular rash during the initial two months of treatment; the rash quickly improves with stoppage of the medication (131,132). In certain patients with genetic predispositions, chlorpropamide may also cause acute facial flushing following alcohol consumption (133). SGLT-2 inhibitors have been associated with an increased risk of genital fungal infections and Fournier’s gangrene (134) (for details see Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes) (135).

 

CONCLUSION

 

Diabetes mellitus is associated with a broad array of dermatologic conditions (Table 1). Many of the sources describing dermatologic changes associated with diabetes mellitus are antiquated; larger research studies utilizing modern analytic tools are needed to better understand the underlying pathophysiology and treatment efficacy. Although each condition may respond to a variety of specific treatments, many will improve with improved glycemic control. Hence, patient education and lifestyle changes are key in improving the health and quality of life of patients with diabetes mellitus.

 

Table 1. Frequent Skin Manifestations of Diabetes Mellitus

DISEASE

APPEARANCE

COMMON LOCATIONS

SYMPTOMS

TREATMENT

Acanthosis Nigricans

Multiple poorly demarcated plaques with grey to dark-brown hyperpigmentation, and a thickened velvety to verrucous texture

Back of the neck, axilla, elbows, palmer hands, inframammary creases, umbilicus, groin

Typically, asymptomatic

Improved glycemic control, oral retinoids, ammonium lactate, retinoic acid, salicylic acid

Diabetic Dermopathy

Rounded, dull, red papules that progressively evolve over one-to-two weeks into well-circumscribed, atrophic, brown macules with a fine scale; lesions present in different stages of evolution at the same time

Pretibial area, lateral meoli, thighs

Typically, asymptomatic

Self-resolving

Diabetic Foot Syndrome

Chronic ulcers, secondary infection, diabetic neuro-osteoarthropathy, clawing deformity

Feet

Typically, asymptomatic but may have abnormal gait

Interdisciplinary team-based approach involving daily surveillance, appropriate foot hygiene, proper footwear/walker, wound care, antibiotics, wound debridement, surgery

Scleroderma-like Skin Changes

Slowly developing painless, indurated, occasionally waxy appearing, thickened skin

Acral areas: dorsum of the fingers, proximal interphalangeal areas, metacarpophalangeal joints

Typically, asymptomatic but may have reduced range of motion

Improved glycemic control, aldose reductase inhibitors, physical therapy

Ichthyosiform Skin Changes

Large bilateral areas of dryness and scaling (may be described as “fish scale” skin)

Anterior shins, hands, feet

Typically, asymptomatic

Emollients, Keratolytics

Xerosis

Abnormally dry skin that may also present with scaling or fissures

Most common on the feet

Typically, asymptomatic

Emollients

Pruritus

Normal or excoriated skin

Often localized to the scalp, ankles, feet, trunk, or genitalia; however, it may be generalized

Pruritus

Topical capsaicin, topical ketamine-amitriptyline-lidocaine, oral anticonvulsants, antifungals

 

REFERENCES

 

  1. Meurer M, Stumvoll M, Szeimies R-M. Hautveränderungen bei Diabetes mellitus. Der Hautarzt 2004;55(5):428-35
  2. Stuart CA, Gilkison CR, Smith MM, Bosma AM, Keenan BS, Nagamani M. Acanthosis nigricans as a risk factor for non-insulin dependent diabetes mellitus. Clinical Pediatrics 1998;37(2):73-79
  3. Stuart CA, Driscoll MS, Lundquist KF, Gilkison CR, Shaheb S, Smith MM. Acanthosis nigricans. Journal of basic and clinical physiology and pharmacology 1998;9(2-4):407-18
  4. Yeh JS, Munn SE, Plunkett TA, Harper PG, Hopster DJ, du Vivier AW. Coexistence of acanthosis nigricans and the sign of Leser-Trelat in a patient with gastric adenocarcinoma: a case report and literature review. Journal of the American Academy of Dermatology 2000;42(2):357-62
  5. Bhagyanathan M, Dhayanithy D, Parambath VA, Bijayraj R. Acanthosis nigricans: A screening test for insulin resistance–An important risk factor for diabetes mellitus type-2. Journal of family medicine and primary care 2017;6(1):43
  6. Hermanns-Lê T, Scheen A, Piérard GE. Acanthosis nigricans associated with insulin resistance. American journal of clinical dermatology 2004;5(3):199-203
  7. Higgins SP, Freemark M, Prose NS. Acanthosis nigricans: a practical approach to evaluation and management. Dermatology online journal 2008;14(9)
  8. Blobstein SH. Topical therapy with tretinoin and ammonium lactate for acanthosis nigricans associated with obesity. Cutis 2003;71(1):33-34
  9. Ehsani A, Noormohammadpour P, Goodarzi A, et al. Comparison of long-pulsed alexandrite laser and topical tretinoin-ammonium lactate in axillary acanthosis nigricans: A case series of patients in a before-after trial. Caspian journal of internal medicine 2016;7(4):290
  10. Sinha S, Schwartz RA. Juvenile acanthosis nigricans. Journal of the American Academy of Dermatology 2007;57(3):502-08
  11. Bustan RS, Wasim D, Yderstræde KB, Bygum A. Specific skin signs as a cutaneous marker of diabetes mellitus and the prediabetic state-a systematic review. Danish medical journal 2017;64(1)
  12. Morgan AJ, Schwartz RA. Diabetic dermopathy: A subtle sign with grave implications. Journal of the American Academy of Dermatology 2008;58(3):447-51
  13. Romano G, Moretti G, Di Benedetto A, et al. Skin lesions in diabetes mellitus: prevalence and clinical correlations. Diabetes research and clinical practice 1998;39(2):101-06
  14. Huntley AC. The cutaneous manifestations of diabetes mellitus. Journal of the American Academy of Dermatology 1982;7(4):427-55
  15. Houck GM, Morgan MB. A reappraisal of the histologic findings of pigmented pretibial patches of diabetes mellitus. Journal of cutaneous pathology 2004;31(2):141-44
  16. McCASH S, Emanuel PO. Defining diabetic dermopathy. The Journal of dermatology 2011;38(10):988-92
  17. Kaňková K, Záhejský J, Márová I, et al. Polymorphisms in the RAGE gene influence susceptibility to diabetes-associated microvascular dermatoses in NIDDM. Journal of Diabetes and its Complications 2001;15(4):185-92
  18. Kiziltan M, Benbir G. Clinical and nerve conduction studies in female patients with diabetic dermopathy. Acta diabetologica 2008;45(2):97-105
  19. Amin N, Doupis J. Diabetic foot disease: from the evaluation of the “foot at risk” to the novel diabetic ulcer treatment modalities. World journal of diabetes 2016;7(7):153
  20. Sämann A, Tajiyeva O, Müller N, et al. Prevalence of the diabetic foot syndrome at the primary care level in Germany: a cross‐sectional study. Diabetic Medicine 2008;25(5):557-63
  21. Singh N, Armstrong DG, Lipsky BA. Preventing foot ulcers in patients with diabetes. Jama 2005;293(2):217-28
  22. Boulton AJ, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J. The global burden of diabetic foot disease. The Lancet 2005;366(9498):1719-24
  23. Mlinarić-Missoni E, Kalenić S, Važić-Babić V. Species distribution and frequency of isolation of yeasts and dermatophytes from toe webs of diabetic patients. Acta dermatovenerologica Croatica 2005;13(2):0-0
  24. Davis MD, O'fallon WM, Rogers III RS, Rooke TW. Natural history of erythromelalgia: presentation and outcome in 168 patients. Archives of Dermatology 2000;136(3):330-36
  25. Jeffcoate WJ, Harding KG. Diabetic foot ulcers. The lancet 2003;361(9368):1545-51
  26. Lobmann R, Schultz G, Lehnert H. Proteases and the diabetic foot syndrome: mechanisms and therapeutic implications. Diabetes care 2005;28(2):461-71
  27. Volmer-Thole M, Lobmann R. Neuropathy and diabetic foot syndrome. International journal of molecular sciences 2016;17(6):917
  28. Sandireddy R, Yerra VG, Areti A, Komirishetty P, Kumar A. Neuroinflammation and oxidative stress in diabetic neuropathy: futuristic strategies based on these targets. International journal of endocrinology 2014;2014

28A. Theocharidis G, Thomas BE, Sarkar D, Mumme HL, Pilcher WJR, Dwivedi B, Sandoval-Schaefer T, Sîrbulescu RF, Kafanas A, Mezghani I, Wang P, Lobao A, Vlachos IS, Dash B, Hsia HC, Horsley V, Bhasin SS, Veves A, Bhasin M. Single cell transcriptomic landscape of diabetic foot ulcers. Nat Commun. 2022 Jan 10;13(1):181. doi: 10.1038/s41467-021-27801-8. PMID: 35013299; PMCID: PMC8748704.

  1. Vinik AI, Erbas T. Recognizing and treating diabetic autonomic neuropathy. Cleve Clin J Med 2001;68(11):928-30, 32, 34-44
  2. Peripheral Arterial Disease in People With Diabetes. Diabetes Care 2003;26(12):3333-41 .

30A. Lanting SM, Chan TL, Casey SL, Peterson BJ, Chuter VH. Cutaneous microvascular reactivity in Charcot neuroarthropathy: a systematic review and meta-analysis. J Foot Ankle Res. 2022 Mar 1;15(1):17. doi: 10.1186/s13047-022-00522-x. PMID: 35232466; PMCID: PMC8886937.

  1. Khanna S, Biswas S, Shang Y, et al. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PloS one 2010;5(3):e9539
  2. Ahn C, Yosipovitch G, Huang W. Diabetes and the Skin. In: Callen J, Jorizzo J, Zone J, Piette W, Rosenbach M, Vleugels RA, eds. Dermatological Signs of Systemic Disease. Edinburgh: Elsevier, 2017:205-14.
  3. Yosipovitch G, Hodak E, Vardi P, et al. The prevalence of cutaneous manifestations in IDDM patients and their association with diabetes risk factors and microvascular complications. Diabetes care 1998;21(4):506-09
  4. Brik R, Berant M, Vardi P. The scleroderma‐like syndrome of insulin‐dependent diabetes mellitus. Diabetes/Metabolism Research and Reviews 1991;7(2):121-28
  5. Yosipovitch G, Yosipovitch Z, Karp M, Mukamel M. Trigger finger in young patients with insulin dependent diabetes. The Journal of rheumatology 1990;17(7):951-52
  6. Kim RP, Edelman SV, Kim DD. Musculoskeletal complications of diabetes mellitus. Clinical Diabetes 2001;19(3):132-35
  7. Haustein UF. Scleroderma‐like lesions in insulin‐dependent diabetes mellitus. Journal of the European Academy of Dermatology and Venereology 1999;13(1):50-53
  8. Yosipovitch G, Loh KC, Hock OB. Medical pearl: Scleroderma-like skin changes in patients with diabetes mellitus. Journal of the American Academy of Dermatology 2003;49(1):109-11
  9. Rho YW, Suhr KB, Lee JH, Park JK. A clinical observation of scleredema adultorum and its relationship to diabetes. The Journal of dermatology 1998;25(2):103-07
  10. Sawatkar G, Kanwar A, Dogra S, Bhadada S, Dayal D. Spectrum of cutaneous manifestations of type 1 diabetes mellitus in 500 south Asian patients. British Journal of Dermatology 2014;171(6):1402-06
  11. Gerrits EG, Landman GW, Nijenhuis-Rosien L, Bilo HJ. Limited joint mobility syndrome in diabetes mellitus: A minireview. World journal of diabetes 2015;6(9):1108
  12. Fitzgibbons PG, Weiss A-PC. Hand manifestations of diabetes mellitus. The Journal of hand surgery 2008;33(5):771-75
  13. Abate M, Schiavone C, Pelotti P, Salini V. Limited joint mobility in diabetes and ageing: recent advances in pathogenesis and therapy: SAGE Publications Sage UK: London, England, 2010.
  14. Frost D, Beischer W. Limited joint mobility in type 1 diabetic patients. Diabetes care 2001;24(1):95-99
  15. Lopez-Martin I, Benito Ortiz L, Rodriguez-Borlado B, Cano Langreo M, Garcia-Martinez FJ, Martin Rodriguez MF. (Association between limited joint mobility syndrome and risk of accidental falls in diabetic patients). Semergen 2015;41(2):70-5
  16. Lindsay JR, Kennedy L, Atkinson AB, et al. Reduced prevalence of limited joint mobility in type 1 diabetes in a UK clinic population over a 20-year period. Diabetes Care 2005;28(3):658-61
  17. Rongioletti F, Kaiser F, Cinotti E, et al. Scleredema. A multicentre study of characteristics, comorbidities, course and therapy in 44 patients. Journal of the European Academy of Dermatology and Venereology 2015;29(12):2399-404
  18. Cole GW, Headley J, Skowsky R. Scleredema Diabeticorum: A Common and Distinct Cutaneous Manifestation of Diabetes Mellitus. Diabetes Care 1983;6(2):189-92.
  19. Carrington PR, Sanusi I, Winder PR, Turk LL, Jones C, Millikan LE. Scleredema adultorum. International journal of dermatology 1984;23(8):514-22
  20. Martin C, Requena L, Manrique K, Manzarbeitia F, Rovira A. Scleredema diabeticorum in a patient with type 2 diabetes mellitus. Case reports in endocrinology 2011;2011
  21. Varga J, Gotta S, Li L, Sollberg S, Leonardo M. Scleredema adultorum: case report and demonstration of abnormal expression of extracellular matrix genes in skin fibroblasts in vivo and in vitro. British Journal of Dermatology 1995;132(6):992-99
  22. Ferreli C, Gasparini G, Parodi A, Cozzani E, Rongioletti F, Atzori L. Cutaneous Manifestations of Scleroderma and Scleroderma-Like Disorders: a Comprehensive Review. Clinical Reviews in Allergy & Immunology 2017:1-31
  23. Bray SM, Varghese S, English JC. Ultrasonic massage and physical therapy for scleredema: improving activities of daily living. Archives of dermatology 2010;146(4):453-54
  24. Reid SD, Ladizinski B, Lee K, Baibergenova A, Alavi A. Update on necrobiosis lipoidica: a review of etiology, diagnosis, and treatment options. Journal of the American Academy of Dermatology 2013;69(5):783-91
  25. O’toole E, Kennedy U, Nolan J, Young M, Rogers S, Barnes L. Necrobiosis lipoidica: only a minority of patients have diabetes mellitus. Br J Dermatol 1999;140(2):283-6
  26. Muller SA, Winkelmann R. Necrobiosis lipoidica diabeticorum: a clinical and pathological investigation of 171 cases. Archives of dermatology 1966;93(3):272-81

56A. Severson KJ, Patel MH, Brumfiel CM, Breen I, Butterfield RJ, Nelson SA, Sekulic A, Pittelkow MR, Mangold AR. Comorbidities and diabetic complications in patients with necrobiosis lipoidica. J Am Acad Dermatol. 2022 Apr;86(4):891-894. doi: 10.1016/j.jaad.2021.03.026. Epub 2021 Mar 17. PMID: 33722550.

  1. Kota SK, Jammula S, Kota SK, Meher LK, Modi KD. Necrobiosis lipoidica diabeticorum: A case-based review of literature. Indian journal of endocrinology and metabolism 2012;16(4):614
  2. Lopez PR, Leicht S, Sigmon JR, Stigall L. Bullosis diabeticorum associated with a prediabetic state. Southern medical journal 2009;102(6):643-44
  3. Larsen K, Jensen T, Karlsmark T, Holstein PE. Incidence of bullosis diabeticorum–a controversial cause of chronic foot ulceration. International wound journal 2008;5(4):591-96
  4. Lipsky BA, Baker PD, Ahroni JH. Diabetic bullae: 12 cases of a purportedly rare cutaneous disorder. International journal of dermatology 2000;39(3):196-200
  5. Patel N, Spencer LA, English JC, Zirwas MJ. Acquired ichthyosis. Journal of the American Academy of Dermatology 2006;55(4):647-56
  6. Pavlović MD, Milenković T, Dinić M, et al. The prevalence of cutaneous manifestations in young patients with type 1 diabetes. Diabetes care 2007;30(8):1964-67
  7. Goyal A, Raina S, Kaushal SS, Mahajan V, Sharma NL. Pattern of cutaneous manifestations in diabetes mellitus. Indian journal of dermatology 2010;55(1):39

63A. Iacopi E, Riitano N, Dini V, Berta R, Pieruzzi L, Janowska A, Anselmino M, Piaggesi A, Romanelli M. Using Skin Bioengineering to Highlight How Weight and Diabetes Mellitus Modify the Skin in the Lower Limbs of Super-Obese Patients. Diabetes Metab Syndr Obes. 2020 Mar 16;13:729-738. doi: 10.2147/DMSO.S229042. PMID: 32214836; PMCID: PMC7083633.

  1. Pavicic T, Korting HC. Xerosis and callus formation as a key to the diabetic foot syndrome: dermatologic view of the problem and its management. JDDG: Journal der Deutschen Dermatologischen Gesellschaft 2006;4(11):935-41
  2. Saray Y, Seçkin D, Bilezikçi B. Acquired perforating dermatosis: clinicopathological features in twenty‐two cases. Journal of the European Academy of Dermatology and Venereology 2006;20(6):679-88
  3. Karpouzis A, Giatromanolaki A, Sivridis E, Kouskoukis C. Acquired reactive perforating collagenosis: current status. The Journal of dermatology 2010;37(7):585-92
  4. Poliak SC, Lebwohl MG, Parris A, Prioleau PG. Reactive perforating collagenosis associated with diabetes mellitus. New England Journal of Medicine 1982;306(2):81-84
  5. Abdelbaqi‐Salhab M, Shalhub S, Morgan MB. A current review of the cutaneous manifestations of renal disease. Journal of cutaneous pathology 2003;30(9):527-38
  6. Robles-Mendez J, Vazquez-Martinez O, Ocampo-Candiani J. Skin manifestations of chronic kidney disease. Actas Dermo-Sifiliográficas 2015;106(8):609-22
  7. Morton C, Henderson I, Jones M, Lowe J. Acquired perforating dermatosis in a British dialysis population. British Journal of Dermatology 1996;135(5):671-77

70A. Forouzandeh M, Stratman S, Yosipovitch G. The treatment of Kyrle's disease: a systematic review. J Eur Acad Dermatol Venereol. 2020 Jul;34(7):1457-1463. doi: 10.1111/jdv.16182. Epub 2020 Feb 5. PMID: 31919924.

  1. Van Hattem S, Bootsma AH, Thio HB. Skin manifestations of diabetes. Cleve Clin J Med 2008;75(11):772-74
  2. Wagner G, Sachse MM. Acquired reactive perforating dermatosis. JDDG: Journal der Deutschen Dermatologischen Gesellschaft 2013;11(8):723-29 doi: 10.1111/ddg.12131(published Online First: Epub Date)|.

72A. Ying Y, Shuang C, Zhen-Ying Z. Dupilumab may be an alternative option in the treatment of acquired reactive perforating collagenosis combined with AD. Immun Inflamm Dis. 2022 Mar;10(3):e574. doi: 10.1002/iid3.574. Epub 2021 Dec 24. PMID: 34953055; PMCID: PMC8926492.

  1. Sugandhan S, Khandpur S, Sharma VK. Familial chylomicronemia syndrome. Pediatric dermatology 2007;24(3):323-25
  2. Mahajan S, Koranne R, Sharma S. Cutaneous manifestation of diabetes melitus. Indian Journal of Dermatology, Venereology, and Leprology 2003;69(2):105
  3. Ahmed K, Muhammad Z, Qayum I. Prevalence of cutaneous manifestations of diabetes mellitus. J Ayub Med Coll Abbottabad 2009;21(2):76-9
  4. Vergès B. Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia 2015;58(5):886-99
  5. Naik NS. Eruptive xanthomas. Dermatology online journal 2001;7(2)
  6. Zaremba J, Zaczkiewicz A, Placek W. Eruptive xanthomas. Advances in Dermatology and Allergology/Postȩpy Dermatologii i Alergologii 2013;30(6):399
  7. Parker F. Xanthomas and hyperlipidemias. Journal of the American Academy of Dermatology 1985;13(1):1-30
  8. Ales Z, Zeman M, Slaby A, Vecka M. Xanthomas: clinical and pathophysiological relations. Biomedical Papers 2014;158(2):181-88
  9. Margolis J. Letter: Skin tags in diabetes mellitus. N Engl J Med 1976;295:172-3
  10. Levy L, Zeichner JA. Dermatologic manifestation of diabetes. Journal of diabetes 2012;4(1):68-76
  11. Yamaoka H, Sasaki H, Yamasaki H, et al. Truncal pruritus of unknown origin may be a symptom of diabetic polyneuropathy. Diabetes care 2010;33(1):150-55
  12. Valdes-Rodriguez R, Mollanazar NK, González-Muro J, et al. Itch prevalence and characteristics in a Hispanic geriatric population: a comprehensive study using a standardized itch questionnaire. Acta dermato-venereologica 2015;95(4):417-21

84A. Yang QP, Chen YY, Li Z, Xu M. Major Risk Factors Analysis of Pruritus Complicated by Type 2   Diabetes Mellitus and the Effect of Comprehensive Nursing Intervention. Front Surg. 2022 Feb 15;9:842884. doi: 10.3389/fsurg.2022.842884. PMID: 35242808; PMCID: PMC8887712.

  1. Guarneri C, Guarneri F, Borgia F, Vaccaro M. Finger pebbles in a diabetic patient: Huntley's papules. International journal of dermatology 2005;44(9):755-56
  2. Tabor CA, Parlette EC. Cutaneous manifestations of diabetes: signs of poor glycemic control or new-onset disease. Postgraduate medicine 2006;119(3):38-44
  3. Murphy-Chutorian B, Han G, Cohen SR. Dermatologic manifestations of diabetes mellitus. Endocrinology and Metabolism Clinics 2013;42(4):869-98
  4. Liau M, Long V, Yang S, Tan K, Aw D. Pigmented purpuric dermatosis in Singapore: a clinic-epidemiological characterisation. Hong Kong Journal of Dermatology & Venereology 2016;24(2):65-69
  5. Lithner F. Purpura, pigmentation and yellow nails of the lower extremities in diabetics. Journal of Internal Medicine 1976;199(1‐6):203-08
  6. Demirseren DD, Emre S, Akoglu G, et al. Relationship between skin diseases and extracutaneous complications of diabetes mellitus: clinical analysis of 750 patients. American journal of clinical dermatology 2014;15(1):65-70
  7. Serrao R, Zirwas M, English JC. Palmar erythema. American journal of clinical dermatology 2007;8(6):347-56
  8. Landau J, Davis E. The small blood-vessels of the conjunctiva and nailbed in diabetes mellitus. The Lancet 1960;276(7153):731-34
  9. Oumeish OY. Skin disorders in patients with diabetes. Clinics in dermatology 2008;26(3):235-42
  10. Duff M, Demidova O, Blackburn S, Shubrook J. Cutaneous manifestations of diabetes mellitus. Clinical Diabetes 2015;33(1):40-48
  11. Bristow I. Non‐ulcerative skin pathologies of the diabetic foot. Diabetes/metabolism research and reviews 2008;24(S1)

95A. Yesudian PD, Nwabudike LC, de Berker D. Nail changes in diabetes. Clin Exp Dermatol. 2022 Jan;47(1):9-15. doi: 10.1111/ced.14859. Epub 2021 Sep 21. PMID: 34293827.

  1. Mendes AL, Miot HA, Haddad Junior V. Diabetes mellitus and the skin. Anais brasileiros de dermatologia 2017;92(1):8-20
  2. Yun JH, Lee JY, Kim MK, et al. Clinical and pathological features of generalized granuloma annulare with their correlation: a retrospective multicenter study in Korea. Annals of dermatology 2009;21(2):113-19
  3. Cyr PR. Diagnosis and management of granuloma annulare. American family physician 2006;74(10)
  4. Armstrong AW, Harskamp CT, Armstrong EJ. Psoriasis and the risk of diabetes mellitus: A systematic review and meta-analysis. JAMA Dermatology 2013;149(1):84-91
  5. Seyhan M, Özcan H, Sahin I, Bayram N, Karincaoğlu Y. High prevalence of glucose metabolism disturbance in patients with lichen planus. Diabetes research and clinical practice 2007;77(2):198-202
  6. Guggenheimer J, Moore PA, Rossie K, et al. Insulin-dependent diabetes mellitus and oral soft tissue pathologies: I. Prevalence and characteristics of non-candidal lesions. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology 2000;89(5):563-69
  7. Van den Driessche A, Eenkhoorn V, Van Gaal L, De Block C. Type 1 diabetes and autoimmune polyglandular syndrome: a clinical review. Neth J Med 2009;67(11):376-87
  8. Von der Werth J, Williams H. The natural history of hidradenitis suppurativa. Journal of the European Academy of Dermatology and Venereology 2000;14(5):389-92
  9. Bui T-L, Silva-Hirschberg C, Torres J, Armstrong AW. Hidradenitis suppurativa and diabetes mellitus: A systematic review and meta-analysis. Journal of the American Academy of Dermatology 2017
  10. Alikhan A, Lynch PJ, Eisen DB. Hidradenitis suppurativa: a comprehensive review. Journal of the American Academy of Dermatology 2009;60(4):539-61
  11. Eldor R, Glaser B, Fraenkel M, Doviner V, Salmon A, Gross DJ. Glucagonoma and the glucagonoma syndrome–cumulative experience with an elusive endocrine tumour. Clinical endocrinology 2011;74(5):593-98
  12. Wermers RA, Fatourechi V, Wynne AG, Kvols LK, Lloyd RV. The Glucagonoma Syndrome Clinical and Pathologic Features in 21 Patients. Medicine 1996;75(2):53-63
  13. Kovács RK, Korom I, Dobozy A, Farkas G, Ormos J, Kemény L. Necrolytic migratory erythema. Journal of cutaneous pathology 2006;33(3):242-45
  14. Kindmark H, Sundin A, Granberg D, et al. Endocrine pancreatic tumors with glucagon hypersecretion: a retrospective study of 23 cases during 20 years. Medical Oncology 2007;24(3):330-37
  15. Halvorson SA, Gilbert E, Hopkins RS, et al. Putting the pieces together: necrolytic migratory erythema and the glucagonoma syndrome. Journal of general internal medicine 2013;28(11):1525-29
  16. Macedo GMC, Nunes S, Barreto T. Skin disorders in diabetes mellitus: an epidemiology and physiopathology review. Diabetology & metabolic syndrome 2016;8(1):63
  17. Kutlu SS, Cevahir N, Akalin S, et al. Prevalence and risk factors for methicillin-resistant Staphylococcus aureus colonization in a diabetic outpatient population: a prospective cohort study. American journal of infection control 2012;40(4):365-68
  18. Hartemann‐Heurtier A, Robert J, Jacqueminet S, et al. Diabetic foot ulcer and multidrug‐resistant organisms: risk factors and impact. Diabetic Medicine 2004;21(7):710-15
  19. Casqueiro J, Casqueiro J, Alves C. Infections in patients with diabetes mellitus: A review of pathogenesis. Indian journal of endocrinology and metabolism 2012;16(Suppl1):S27
  20. Driscoll JA, Brody SL, Kollef MH. The epidemiology, pathogenesis and treatment of Pseudomonas aeruginosa infections. Drugs 2007;67(3):351-68
  21. Bonne SL, Kadri SS. Evaluation and management of necrotizing soft tissue infections. Infectious Disease Clinics 2017;31(3):497-511
  22. Dworkin M, Westercamp M, Park L, McIntyre A. The epidemiology of necrotizing fasciitis including factors associated with death and amputation. Epidemiology & Infection 2009;137(11):1609-14
  23. Bhandary S, Karki P, Sinha B. Malignant otitis externa: a review. Pacific health dialog 2002;9(1):64-67
  24. Sashikumar R, Kannan R. Salivary glucose levels and oral candidal carriage in type II diabetics. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology 2010;109(5):706-11
  25. Yosipovitch G, Tur E, Cohen O, Rusecki Y. Skin surface pH in intertriginous areas in NIDDM patients: possible correlation to candidal intertrigo. Diabetes Care 1993;16(4):560-63
  26. Saunte DML, Holgersen JB, Hædersdal M, et al. Prevalence of toe nail onychomycosis in diabetic patients. Acta dermato-venereologica 2006;86(5):425-28
  27. Alteras I, Saryt E. Prevalence of pathogenic fungi in the toe-webs and toe-nails of diabetic patients. Mycopathologia 1979;67(3):157-59
  28. Romano C, Massai L, Asta F, Signorini A. Prevalence of dermatophytic skin and nail infections in diabetic patients. Mycoses 2001;44(3‐4):83-86
  29. Rich P. Onychomycosis and tinea pedis in patients with diabetes. Journal of the American Academy of Dermatology 2000;43(5):S130-S34

124A. Sharma A, Goel A. Mucormycosis: risk factors, diagnosis, treatments, and challenges during COVID-19 pandemic. Folia Microbiol (Praha). 2022 Feb 26:1–25. doi: 10.1007/s12223-021-00934-5. Epub ahead of print. PMID: 35220559; PMCID: PMC8881997.

  1. Sinikumpu S-P, Auvinen J, Jokelainen J, et al. Abnormal skin in toe webs is a marker for abnormal glucose metabolism. A cross-sectional survey among 1,849 adults in Finland. Scientific reports 2017;7:9125
  2. Richardson T, Kerr D. Skin-related complications of insulin therapy. American journal of clinical dermatology 2003;4(10):661-67
  3. Lima AL, Illing T, Schliemann S, Elsner P. Cutaneous Manifestations of Diabetes Mellitus: A Review. American Journal of Clinical Dermatology 2017:1-13
  4. Attaway A, Mersfelder TL, Vaishnav S, Baker JK. Bullous pemphigoid associated with dipeptidyl peptidase IV inhibitors. A case report and review of literature. Journal of dermatological case reports 2014;8(1):24
  5. Byrd JS, Minor DS, Elsayed R, Marshall GD. DPP-4 inhibitors and angioedema: a cause for concern? Annals of Allergy, Asthma & Immunology 2011;106(5):436-38
  6. Hitselberger JF, Fosnaugh RP. Photosensitivity due to chlorpropamide. JAMA 1962;180(1):62-63
  7. Walker G, Kinsell LW. Clinical experience with chlorpropamide. California medicine 1961;94(6):344
  8. Walker G, Slater J, Westlake E, Nabarro J. Clinical experience with tolbutamide. British medical journal 1957;2(5040):323
  9. Leslie R, Pyke D. Chlorpropamide-alcohol flushing: a dominantly inherited trait associated with diabetes. Br med J 1978;2(6151):1519-21
  10. Nyirjesy P, Sobel JD, Fung A, et al. 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. Current medical research and opinion 2014;30(6):1109-19
  11. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2021 Aug 28. In: Feingold KR, Anawalt B, Boyce A, et al editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.

 

Pathophysiology and Treatment of Pancreatic Neuroendocrine Neoplasms (PNENS): New Developments

ABSTRACT  

 

Pancreatic neuroendocrine neoplasms (PNENs) are a heterogenous group of relatively rare pancreatic malignancies with a unique biology and pathophysiology. Over the last few years, there have been significant improvements in imaging and treatment strategies, which have led to advances in patient’s management and quality of life (QOL). Yet, in practice, there are still a number of unanswered questions. For example, it remains a challenge to choose the optimal treatment sequence from the plethora of options and to properly monitor PNEN patients. Therefore, in this chapter, recent advances in the pathophysiology, diagnosis, monitoring, and management of these neoplasms will be summarized and placed in a historical context.

 

INTRODUCTION  

 

Pancreatic neuroendocrine neoplasms (PNENs) are an uncommon subset of neuroendocrine neoplasms (NENs) originating from endocrine cells (1-3). PNENs represent 1-2% of all pancreatic neoplasms and according to the Surveillance, Epidemiology and End Results (SEER) program, the annual age-adjusted incidence has risen from 0.32/100,000 persons in 2004 to 0.48/100,000 persons in 2021 (2, 4-7). Improvements in and a wider availability of high-quality imaging techniques and a well-established classification system are believed to be major factors in the increasing incidence of PNENs (5, 8, 9).

 

PNENs can be divided into both functional (10-40%) and non-functional (60-90%) neoplasms (2, 6, 7, 10, 11). Functional PNENs (F-PNENs) are characterized by a specific clinical course and symptoms due to excessive hormone production (e.g., insulin, gastrin) (10-12). The most frequent, recognized F-PNENs are listed in Table 1 (1). Less common F-PNENs include somatostatinomas, ACTHomas and PNENs that cause carcinoid syndrome, acromegaly, or hypercalcemia (2). Patients with non-functional PNENs (NF-PNENs) lack symptoms related to clinical hormonal syndromes and are therefore usually diagnosed at a more advanced stage with characteristically large primary tumors (70% >5 cm) and liver metastasis in more than 60% of the cases (2, 9, 12, 13). NF-PNENs are hence frequently discovered by chance on imaging studies performed due to nonspecific abdominal pain, often caused by tumor bulk (2, 9, 12, 14). Although NF-PNENs do not secrete peptides causing clinical syndromes, they characteristically secrete a number of other peptides including chromogranin A (CgA) and pancreatic polypeptide (PP). However, elevated levels of PP or CgA are not specific for NF-PNENs as they are also observed in patients with renal failure and inflammatory conditions (2, 9, 12-14).

 

Table 1. Overview of Recognized Functional PNENs and Their Characteristics

Tumor

[syndrome]

Hormone

Clinical symptoms

Biochemical diagnosis

Insulinoma

[Whipple’s triad]

Insulin

Hypoglycemia

At hypoglycemia:

Insulin > 6 µU/L

Glucose 40 mg/dL

C-peptide 0.6 ng/mL

Proinsulin ³ 20 pmol/L

Gastrinoma

[Zollinger-Ellison]

Gastrin

Abdominal pain, Gastroesophageal reflux, Diarrhea, Duodenal ulcers

Serum fasting gastrin level ³ 10 times normal range

VIPoma

[Verner-Morrison]

Vasoactive intestinal peptide (VIP)

Severe watery diarrhea, Hypokalemia

Fasting serum VIP > 60 pmol/L

Glucagonoma

Glucagon

Rash, Glucose intolerance (diabetes), Necrolytic migratory erythema, Weight loss

Fasting glucagon > 500 pg/mL

Note: This table was assembled based on information from Gastroenterology, Metz D. and Jensen R., Gastrointestinal neuroendocrine tumors: Pancreatic Endocrine tumors, 1469-1492 © 2008 (2) and World Journal of Gastroenterology, Ma Z., Gong Y., Zhuang H. et al., Pancreatic neuroendocrine tumors: A review of serum biomarkers, staging and management, 2305-2322 © 2020 (7) and Current Opinion in Gastroenterology, Perri G., Prakash L. and Katz M., Pancreatic neuroendocrine tumors, 468-477 © 2019 (3).  

 

CLASSIFICATION AND STAGING  

 

The World Health Organization (WHO) classification from 2019 (Table 2) takes into account both differentiation status and proliferation rate of the tumor. The former is determined through a histological examination of tumor morphology in which well-differentiated neuroendocrine tumors (NETs) can be distinguished from poorly differentiated neuroendocrine carcinomas (NECs). A grade is then assigned based on the proliferation rate assessed via Ki-67 index and mitotic count. Well-differentiated NETs can be divided into low grade (G1), intermediate grade (G2), and high grade (G3) tumors that have respective Ki-67 values of <3%, 3-20%, and >20% or mitotic counts of <2, 2-20, and >20 per 2mm³ (10 high power fields (HPF)). In the poorly-differentiated NEC group (small and large cell types), only high grade G3 tumors with a Ki-67 value >20 are found. In addition, neoplasms exist that consist of neuroendocrine cells as well as non-neuroendocrine adenocarcinoma or squamous carcinoma cells (i.e., mixed non-neuroendocrine-neuroendocrine neoplasms (MiNEN)) (3, 6-8, 15, 16). Depending on tumor grade and primary site, the 5-year survival varies between 15-95% and median overall survival (OS) from approximately 12 years for patients with G1 to 10 months in patients with G3 PNENs (3, 17). PNENs most often occur sporadically, but can also occur in patients with various inherited disorders (2, 18). For example, PNENs develop in 80-100% of patients with Multiple Endocrine Neoplasia type 1 (MEN1), in 10-17% of patients with von Hippel-Lindau syndrome (VHL), and occasionally in patients with tuberous sclerosis and neurofibromatosis (3, 18). 

 

Table 2. WHO Classification (2019) of PNENs

Type

Differentiation status

Grade

Proliferation rate

Ki-67 (%)

Mitotic count (2mm²)

NEN

Well-differentiated NETs

G1

< 3

< 2

G2

3 – 20

2 – 20

G3

> 20

> 20

Poorly-differentiated NECs

Small cell (SCNECs)

Large cell (LCNECs)

G3

> 20

> 20

MiNEN

NET or NEC + ADC or SCC

G1-G3

See above

See above

Note: This table was adapted from Histopathology, Nagtegaal I., Odze R., Klimstra D. et al., The 2019 WHO classification of tumours of the digestive system, 182-188. © 2019 (16). NEC- neuroendocrine carcinomas; NET- neuroendocrine tumors; ADC- adenocarcinoma cells; SCC- squamous carcinoma cells

 

PNENs are also classified based on the tumor-node-metastasis (TNM) classification which estimates the prognosis of the tumors based on the anatomy of the tumor (3). Previously there was no generally accepted staging system, so in Europe usually the European Neuroendocrine Tumor Society (ENETS) staging system was applied, while in America the America Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) system was being used (19-21). In the 7th edition of the AJCC/UICC, the same ordering system was employed for PNENs as for pancreatic adenocarcinoma (PAAD), but due to biological differences between both tumor types, this staging system proved to have some limitations (19, 20). Consequently, in the revised 8th edition of the AJCC/UICC, the classification system of ENETS was implemented (21). Two research groups demonstrated that the system employed in this 8thedition was superior to that of the 7th edition as well as the ENETS staging system and should be considered as golden standard (20, 22).

 

INDUCTION OF PNENs

 

PNENs are also often referred to as islet cell tumors since it is presumed that they arise from the islets of Langerhans (3, 23, 24). These islets contain A-, B-, D-, D1-, and D2-cells that respectively secrete glucagon, insulin, somatostatin, pancreatic polypeptide, and vasoactive intestinal polypeptide (VIP) (25). Logically, the F-PNENs most definitely arise from these cells, but the cell of origin in NF-PNENs is still a matter of debate (26, 27). Chan et al. revealed that NF-PNENs with ATRX, DAXX, and MEN1 mutations (A-D-M mutant) had a worse clinical outcome than A-D-M wild-type (WT) tumors. In addition, they were able to demonstrate, through RNA sequencing and DNA methylation analysis, that the A-D-M mutant PNENs had high ARX and low PDX1 expression which is consistent with the expression profile found in a-cells (28). Cejas et al. found that NF-PNENs could be divided into two subgroups with epigenomes and transcriptomes very similar to those of a- and b-cells, respectively (29). These findings were confirmed by Di Domenico and colleagues who were able to demonstrate that the genome-wide DNA methylation profiles of NF-PNENs were very consistent with the methylation profiles of a- and b-cells (24). Based on these findings, it was hypothesized that NF-PNENs evolve primarily but not exclusively from the a-cell lineage and b-cell lineage (27).

Figure 1. Visualization of the pancreatic duct glandular structures (PDGs) (arrowheads) in (A) large and (B) small ducts using scanning electron microscopy (SEM). PDGs can occur as single outpouches or form a complex of sac-like dilatations as illustrated in (C). This figure has been adapted from Gastroenterology, Strobel O., Rosow D. E., Rakhlin E. Y., et al., Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia, 138 (3): 1166-77 © 2010 (30).

Others in turn suggest that PNENs develop from multipotent pancreatic progenitor (MPP) cells in the ductal and islet regions of the pancreas that would be able to generate new pancreatic islet cells (31, 32). However, it remains unclear whether these cells originate in the islets or whether they migrate from the pancreatic ducts to subsequently transform into endocrine cells (33). This hypothesis is strengthened by the fact that early endocrine progenitors in fact appear to originate from a bipotent ductal endocrine progenitor, which in turn originates from MPP cells (34). However, not a lot is known about where these MPP cells are present. One hypothesis states these could be present in the pancreatic duct glandular structures (PDGs) that can be found as specialized compartments with a gland-like outpouching look (Figure 1) in the ductal epithelium (30, 35). The actual origin and location of the MPP that can evolve into islet cells is not known to date and thus needs to be further investigated for a better understanding of the potential origin of PNENs.

 

MOLECULAR (EPI)GENETICS  

 

Genetic Syndromes

 

Although PNENs typically occur sporadically, approximately 10-20% of them develop in the context of hereditary syndromes. The syndrome most at risk for PNEN development is Multiple Endocrine Neoplasia (MEN1) (60%), an autosomal dominant disease caused by inactivating mutations in the MEN1 gene (10, 36-39). MEN1 is a tumor suppressor gene located on chromosome 11q13 that encodes for the nuclear protein menin which plays an important role in the PI3K/Akt/mTOR pathway, histone modifications, DNA repair mechanisms, and cell cycle control (10, 37, 38, 40, 41). In addition, 5 to 18% of the patients with von Hippel-Lindau (VHL) syndrome develop PNENs. These patients carry a germline mutation in the VHL gene located on the short arm of chromosome 3. The VHL protein can be found in different complexes that mediate ubiquitin-mediated degradation and stimulate angiogenesis (10, 42, 43). Other hereditary syndromes at risk include Tuberous Sclerosis (TS) and neurofibromatosis type 1, caused by mutations in TSC1, TSC2 and NF1, respectively (36-39).

 

Sporadic PNENS  

 

Through next-generation sequencing of PNENs, it became apparent that there are distinct genetic differences, strongly depending on differentiation and functionality of the tumor (36).

For example, genetic analyses of F-PNENs revealed that insulinomas are often characterized by a hotspot mutation (p. T372R) in the Yin Yang 1 (YY1) gene in 30% of the Asian and 8-33% of the Western/Caucasian population (10, 36, 44). This recurrent mutation is located in the DNA binding domain of YY1, hence strongly affecting the DNA binding capacity of this transcriptional activator/repressor (44). In NF-PNENs, on the other hand, somatic mutations were most commonly identified in MEN1 (44.1%) followed by DAXX (25%) and ATRX (17.6%) (41). Atrx interacts with DNA methyltransferases (DNMT) 3A and 3L to form the Atrx-DNMT3A-DNMT3L (ADD) complex. This interaction is crucial for maintenance of histone methylation patterns in newly replicated chromatin, hence indirectly ensures correct gene expression. Moreover, Atrx also interacts directly with Daxx. In doing so, Daxx functions as a kind of chaperone for the deposition of histone variant H3.3 at the level of CpG islands, telomeric and ribosomal repeats and the rest of the genome. Consequently, a loss of ATRX and DAXX results in changes in DNA methylation patterns throughout the genome (45). In addition, mutations in PTEN, TSC2, and PIK3CA have already been reported in respectively 7.8%, 8.8% and 1.4% of PNENs, and they all affect the PI3K/Akt/mTOR pathway (41). Later, Scarpa and colleagues identified mutations in DNA repair genes MUTYH, CHEK2, and BRCA2 as well (39).

 

Distinct genetic differences could also be observed between G3 pancreatic NETs (PNETs) and pancreatic NECs (PNECs). The latter do not carry mutations in the known genes for PNETs (MEN1, DAXX, and ATRX), but instead appear to have mutations in TP53, RB1, KRAS, and CDKN2A/p16 (10, 36, 38). Considering that these mutations tend to result in altered protein expression, IHC might facilitate in distinguishing PNETs from PNECs, which have similar Ki-67 values. Nevertheless, results should always be interpreted with caution (10, 36, 46). 

 

Besides point mutations, copy number alterations (CNAs) have also attracted attention. CNA patterns that were frequently identified included whole or partial loss of chromosomes 1, 2, 3, 6, 8, 10, 11, 15, 16, 21 and 22, while gains have been observed in chromosomes 5, 7, 12,14 and 17 (10). Moreover, PNENs appear to display very specific CNA patterns that allow to distinguish PNENs from the more common PAADs. Boons and colleagues therefore developed a classification model, based on tumor tissue, which demonstrated a sensitivity, specificity and area under the curve (AUC) of 100%, 95% and 100% in the validation cohort (47). Benign insulinomas tend to display lower rates of CNAs (36).        

 

Since genes such as MEN1, DAXX and ATRX are of importance in several epigenetic regulatory processes, it was extremely likely that also epigenetic alterations commonly occur in PNENs. In fact, both in hereditary and sporadic PNENs promotor hypermethylation is observed in tumor suppressor genes, which is associated with silencing of gene expression (10, 48). Chan and colleagues checked whether methylation profiles and expression were different in the A-D-M mutated group versus the A-D-M WT group. They observed that both groups clustered in two separate clusters and even revealed that gene expression of the A-D-M mutated group was respectively high and low in the ARX and PDX1 gene and the latter gene also displayed hypermethylation. This profile appeared to be quite similar to that of acells in the pancreas (28). These results were confirmed by Neiman et al., who observed high methylation levels in the PDX1 promotor region in a cells, while β cells tend to have low methylation in this region (49). Based on this PDX1 gene methylation, Boons and colleagues performed unsupervised hierarchical clustering and could subsequently observe two subpopulations, A and B, which respectively contained the a and β cells. Of note, the majority of the mutated PNENs was found in group A confirming the findings of Chan et al. (28, 50). These results suggest that methylation profiling of the PDX1 gene could potentially help to divide PNENs into distinct clinically relevant groups that have different prognosis and risk of relapse (50). Recently, three subgroups (T1, T2 and T3) of PNENs have been identified, based on their methylation profile. Here, the T1 group consisted of the A-D-M WT tumors, while the T2 subgroup encompassed the A-D-M mutated tumors with recurrent chromosomal losses and methylation in the gene body of the MGMT gene. The last group, T3, displayed mutations in MEN1 and recurrent loss of chromosome 11. Tumors found in the latter group tend to have a better prognosis (51).   

 

DIAGNOSIS AND MONITORING           

 

The gold standard for diagnosing PNENs remains an immunohistochemical examination of the tumor tissue, but imaging and serum markers are also extremely important in the diagnostic process. The clinical presentation often determines the sequence of examinations. For example, patients with F-PNENs will usually undergo a biochemical blood analysis first based on their hormonal symptoms, whereas NF-PNENs are often detected by chance on imaging (25, 52, 53).  

 

Immunohistochemistry

 

To correctly classify PNENs, tumor morphology and proliferation rates (Ki-67 and mitotic index) should be evaluated in tissue biopsies. These are usually obtained from surgical specimens, percutaneous core biopsies, or preoperative biopsies (52, 54, 55). The latter were mainly derived from endoscopic ultrasound (EUS) guided fine-needle aspirations (FNA) which, in recent years, have been increasingly replaced by fine-needle biopsies (FNB) as these enable histological tissue samples to be obtained, hence immunohistochemistry (IHC) to be performed (56, 57). This immunohistochemical examination is most often initiated by confirming the neuroendocrine differentiation by checking CgA and synaptophysin (SYP) expression (52, 54). Other markers such as neuron-specific enolase (NSE) and CD56 are less specific, hence less useful (58). Next, tumor morphology is assessed to determine whether the PNEN is well- or poorly-differentiated (Figure 2). In general, well-differentiated PNENs are characterized by uniform cells with a finely granular cytoplasm and round to oval nucleus which are arranged in a trabecular, glandular, or tubuloacinar pattern (54, 59). Moreover, typically all cells have a heterogeneous expression of CgA in their cytoplasm, whereas SYP stains more diffusely. Poorly-differentiated PNECs, on the other hand, consist of atypical neoplastic cells that often lack CgA and even SYP (59, 60). Ultimately, tumors are graded by proliferation rate that is influenced by two parameters, Ki-67 and mitotic count. The latter is usually reported as the number of mitoses per mm² which in practice is often complicated by a limited tissue area. The mitotically active regions are then measured again via IHC to determine the Ki-67 index (Figure 3). It is therefore logical that the Ki-67 index is usually higher than the mitotic count since it considers the entire mitotic process and not just the number of mitoses. If both values assign a different grade to the same tumor, the highest grade, associated with the worst prognosis, is assumed (17, 23, 52, 54).

Figure 2. Hematoxylin-eosin IHC staining of (A) well-differentiated PNET and (B) poorly-differentiated PNEC. This figure has been adapted from Archives of Pathology & Laboratory Medicine, Fang J. M. and Shi J., A clinicopathologic and molecular update of pancreatic neuroendocrine neoplasms with a focus on the new world health organization classification, 143 (11): 1317-1326. © 2019 (59).

Figure 3. (A) PNEN G1 with Ki-67 index of less than 3%. (B) PNEN G2 with Ki-67 index of 3% to 20%. (C) PNEN G3 with Ki-67 index higher than 20%. This figure has been adapted from Archives of Pathology & Laboratory Medicine, Fang J. M. and Shi J., A clinicopathologic and molecular update of pancreatic neuroendocrine neoplasms with a focus on the new world health organization classification, 143 (11): 1317-1326. © 2019 (59).

Imaging

 

Regardless of whether a PNEN is functional or non-functional, imaging is critical to assess the extent of the disease by localizing the primary tumor and identifying the size of metastatic disease. Localization is required preoperatively to increase the accuracy of intraoperative techniques and to reduce the need for repeated surgery. Besides, imaging is involved in patient’s management as it allows to monitor tumor growth and evaluate response to treatment (2, 25, 53, 55, 61). A multimodal approach is applied to diagnose and stage PNENs which comprises both anatomical and functional imaging modalities (2, 61-66).

 

ANATOMIC IMAGING   

 

Anatomical imaging modalities such as Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) are capable of depicting normal and diseased tissue at high spatial resolution (67). Contrast enhanced CT is the most commonly used and preferred modality as it is widely available, renders clear anatomical images of the pancreas, lymph nodes, and liver metastasis and allows to assess vascular invasion and resectability (25, 52, 62, 64, 68, 69). The more recent, multiphase multidetector CT scan exhibits even more advantages including reduced artifacts due to rapid scan time, improved arterial phase images due to accurate contrast medium tracking and improved resolution by generating thinner slices that can be studied in different anatomical planes (61, 65). In addition, the frequent hypervascular nature of PNENs results in typical high contrast uptake in the arterial phase on CT which can aid in differentiating from pancreatic adenocarcinoma. The average sensitivity of contrast-enhanced CT varies from 63% to 83% and detection rates range from 69% to 94.3% (52, 67, 70). It appears that imaging of PNENs is often influenced by their biological heterogeneity. For example, gastrinoma become more apparent on postcontrast images and large NF-PNENs often have a necrotic or cystic appearance which tend to complicate diagnosis with imaging alone. In the latter case, MRI can be useful as cystic neoplasms can be better visualized due to the higher resolution, rendering MRI complementary to CT. MRI displays a similar sensitivity to CT (79%), but has some advantages over CT as it displays a good sensitivity even without administration of contrast agents, employs non-ionizing radiation, and is hence safer for patient follow-up (52, 53, 67). Limitations on the other hand include a higher frequency of motion-related artifacts as well as a longer acquisition time compared to CT (71).

 

Still, both conventional imaging modalities depend to a large extent on the tumor size (2, 25, 61, 72, 73). More than 70% of PNENs larger than 3 cm are detected, but only 50% of PNENs smaller than 1 cm are identified. As a result, small primary PNENs, especially insulinoma and duodenal gastrinoma, are frequently missed as well as small liver metastases (2, 25, 61, 62, 72-74). For small PNENs, which cannot be detected using CT and MRI, EUS is considered the predominant imaging technique (68). Because of the high spatial resolution of this modality, it is possible to localize even very small lesions (2-3 mm) (3, 74). Additionally, it is feasible to obtain high yield tissue samples by means of an FNA/B that can be used for Ki-67 measurements. Such EUS-FNA/B have a diagnostic accuracy of 80% for pancreatic adenocarcinoma and 46% for PNENs. In patients with proven insulinoma, EUS displays a sensitivity of 94% as a first-line modality. This renders EUS extremely valuable for localizing primary insulinoma (2, 68, 74, 75). However, EUS is not generally available, can be technically challenging, and results are operator dependent. In the hands of an expert, sensitivities of 79 to 100% can be achieved (61).  

 

FUNCTIONAL IMAGING  

 

Prior to the development of the current functional imaging modalities, selective angiography and sampling for hormone gradients were employed. However, due to the highly invasive nature of these techniques, minimally invasive modalities were developed which had a great impact on patient management (2, 53, 64).

 

Although PNENs exhibit highly heterogeneous biological behavior, 80-100% of PNENs, with the exception of insulinomas (50-70%), overexpress the G-linked protein somatostatin receptors (SSTRs), mainly subtypes SSTR-2 and -5. These receptors interact with somatostatin, a peptide hormone that affects neurotransmission and cell proliferation, but also the secretion of various compounds in the digestive system (2, 52, 62-64, 67, 76). Interestingly, these SSTRs also bind synthetic, radiolabeled somatostatin analogs (SSAs) with high affinity, which constitutes the basis of the primary functional imaging tool for PNENs, namely Somatostatin Receptor Imaging (SRI). SRI will not only allow to stage PNENs, but will also predictively identify patients eligible for SSA therapy (2, 52, 53, 64, 67, 68). One of the first SSAs used to target the SSTRs was octreotide labeled with 111Indium via chelator, diethylene-triamine-pentaaceticacid (DTPA). This 111In-DTPA-octreotide emits gamma rays that are detected 24 hours after intravenous injection using Single Photon Emission CT (SPECT) or SPECT/CT, the so-called Octreoscan® (53, 62, 64, 70, 77). The Octreoscan® is often combined with CT to improve the anatomic localization making it highly sensitive (77%), detecting 50-70% of primary PNENs, but less of the insulinomas and duodenal gastrinomas (2, 62-64, 68, 78-81). Major drawbacks include the availability and price of 111In-DTPA octreotide, the staggering acquisition time as well as the intrinsic shortcomings of SPECT, such as low spatial resolution (8-12 mm) (67, 82).

 

Positron Emission Tomography (PET) could provide better spatial resolution and greater precision (71). One of the most widely employed PET radiotracers currently used to image tumors is 18Fluor-labeled deoxy-glucose (18FDG). For high-grade NETs, especially NECs, 18FDG-PET/CT is a better choice as nuclear medicine modality as SSTR expression decreases when proliferation rates increase. 18FDG-PET can even be positive in G2 and G3 NETs. To this date, no cut-off value has been determined. However, it seems like neoplasms with Ki-67 values > 15% are more likely to exhibit a positive 18FDG-PET/CT, which is also a predictor of a more aggressive course (53, 81, 83, 84). However, 18FDG-PET/CT appears to be less useful in the majority of PNENs as these often show limited glucose uptake due to a rather slow growth rate (52, 64).

 

The development of new PET/CT radiotracers has been a major breakthrough in PNEN imaging. 11Carbon-5-hydroxytryptophan-labeled or 68Gallium-labeled SSAs including DOTA-tyrosine-3-octreotide (DOTA-TOC), DOTA-octreotate (DOTA-TATE), and DOTA-1-NaI-octreotide (DOTA-NOC) showed better sensitivity and diagnostic accuracy than the conventional imaging studies (Figure 4) and the Octreoscan® (Figure 5) (2, 52, 64, 71, 85-87). A meta-analysis revealed that 68Ga-DOTA-SSA PET for the diagnosis of NETs has a pooled sensitivity and specificity of 93% and 91%, respectively (88). Admittedly, the majority of the studies involved heterogenous populations, but most included a sizable minority of 20-30% PNENs. Hence, the overall data, although far from perfect, support use of 68Ga-DOTA-SSA PET over the Octreoscan® (89). Moreover, it is highly sensitive for the detection of bony metastases and it might obviate the need for additional radiologic studies. In addition to a higher sensitivity, other advantages of 68Ga-DOTA-SSA PET include patient convenience (imaging sessions take 70-90 minutes instead of 24 hours), lower radiation exposure, utility in finding unknown primary PNENs and it can lead to changes in treatment plans in about 33-41% of the patients (67, 89-93). 68Ga-DOTA-SSA PET might also be better at quantifying SSTR expression which facilitates targeted therapy such as PRRT (89). Consequently, 68Ga-DOTA-SSA PET quickly became the imaging modality of choice (67, 68, 71). However, similar to other imaging studies, false positives may occur due to pancreatic uncinate process activity, inflammation, osteoblastic activity, and splenosis (94). No doubt other PET agents will follow since PNENs express a variety of receptors for which there are potential ligands. For example, insulinomas express SSTRs in 50% of the cases, so tracers targeting the glucagon-like peptide-1 (GLP-1) receptor might be more useful in those patients (80, 95, 96).

Figure 4. Overview of (A) 68Ga-PET/CT, multiphase (B) atrial and (C) portal vein CT scan images from patient with partially cystic PNEN. The arrow indicates a liver metastasis which is only visible on the 68Ga-PET/CT scan. This figure has been adapted from Current treatment options in oncology, Morse B., Al-Toubah T. and Montilla-Soler J., Anatomic and functional imaging of neuroendocrine tumors, 21 (9): 75 © 2020 (67).

Figure 5. Comparison of (A) planar Octreoscan®, (B) Octreoscan®/SPECT/CT fusion, (C) planar 68Ga-DOTATOC-PET and (D) 68Ga-DOTATOC-PET/CT in the same patient. Images C and D clearly display a more precise delineation of the lesions. This figure has been adapted from International journal of endocrine oncology, Maxwell J. E. and Howe J. R., Imaging in neuroendocrine tumors: and update for the clinician, 2 (2): 159-68 © 2015 (82).

All benefits taken into account, the FDA approved 68Ga-DOTATOC PET in 2016 in the US, after being available in Europe for a number of years. Furthermore, with the development of an FDA approved 68Ga generator, an on-site cyclotron is no longer required, making this technology more widely available. A multi-society workgroup has recommended that 68Ga-DOTA-SSA PET replace use of Octreoscan®, unless it is not accessible, in combination with at least one anatomic imaging technique (66, 70).

 

Assessment Through Circulating Biomarkers

 

As stated earlier, the current gold standard for diagnosing and molecularly profiling PNENs remains the analysis of surgical or biopsy tissue samples. However, these samples have a highly invasive character, rendering repetitive sampling unfeasible. Further limitations are the individual patients’ risk and procedural costs. Besides, they represent merely a snapshot of tumor heterogeneity, which strongly influences accuracy. Hence, liquid biopsies aroused strong interest since they form a cost-effective and minimally invasive way to analyze the tumor’s behavior. The most frequently used source is blood as it allows to examine the so-called tumor circulome that consists of a set of circulating components that originate from the tumor (97-101). These blood-based biomarkers play a pivotal role in diagnosing and staging PNENs, monitoring response to therapy, and detecting tumor progression. In case of F-PNENs, specific circulating biomarkers such as insulin, gastrin, and glucagon are employed in hormonal assays to correctly diagnose F-PNENs. Moreover, both F- and NF-PNENs frequently secrete non-specific markers including CgA, neuron-specific enolase (NSE), pancreastatin, etc., which can be detected in patients’ blood as well (2, 9, 13, 52, 53, 55, 102). Besides circulating proteins, PNENs also shed circulating tumor cells (CTCs), circulating tumor RNA (ctRNA) and DNA (ctDNA) which could serve as potential biomarkers (98-100, 103, 104).

 

SPECIFIC BIOMARKERS AND HORMONAL ASSAYS  

 

Depending on the type of F-PNENs (outlined in Table 1), specific biochemical tests are performed. When insulinoma is suspected, serum levels of insulin and C-peptide are measured at a confirmed hypoglycemia during prolonged period of fasting (approximately 72 hours) as patients present with increased levels (> 6 µU/L and 0.6 ng/mL, respectively) even when glucose levels are low (7, 31, 52, 53, 104). In case of gastrinomas, the serum gastrin levels will be 10 times higher than the upper limit and gastric pH will be lower or equal to 2 (3, 7, 52, 53, 105). In patients with suspected VIPoma and glucagonoma, diagnosis is confirmed by determining the fasting levels of VIP and glucagon (7, 53).  

 

Chromogranin A (CgA)

 

CgA, a glycoprotein stored in and secreted by the secretory granules of the neuroendocrine cells, plays an important role not only in immunohistochemistry, but also as a circulating marker (7, 17, 52, 53, 102). CgA is useful as a marker for both functional and non-functional PNENs, as elevated levels are noted in 50-100% of the patients with PNENs (2, 106-108), depending upon the histological subtype (104, 109, 110). For example in gastrinoma, CgA levels are consistently high due to gastrin-induced enterochromaffin-like cell hyperplasia (111), while insulinoma show significantly lower levels of circulating CgA (112). Besides, blood levels depend upon malignant nature of the tumor, tumor burden, and progression, hence small tumors may be associated with normal CgA levels (113, 114).

 

The ENETS still recommends the use of circulating serum CgA as marker during diagnosis and follow-up in NF-PNENs (7, 115). However, the actual diagnostic value of this marker is still questionable (115). Sensitivity, specificity and overall accuracy of this clinical biomarker equal 66%, 95% and 71%, respectively (7), but these values tend to vary according to the specific assays and diagnostic threshold (52). Common conditions that can falsely elevate CgA levels, thus impair specificity, include decreased renal function, treatment with proton pump inhibitors (116), and even essential hypertension (117). In addition, 30-50% of NENs do not show elevated CgA levels, limiting sensitivity (47, 115, 118). This group mostly involves small, localized, non-functional NETs where CgA levels are normal in approximately 70% of the cases (119). As a result, these patients are in a higher need for accurate biochemical markers as diagnosis is harder both clinically and by use of imaging techniques. Moreover, SSA treatment cause a decrease in CgA secretion, which is why results should always be interpret with caution (104). In terms of follow-up, prospective studies demonstrated that elevated CgA levels do not correlate with imaging and tumor progression, hence questioning the potential of CgA as follow-up biomarker (115). Compared to CgA, CgB is not impacted by for example proton pump inhibitor treatment (112, 116). However, only in 25% of the cases with elevated CgA levels, CgB was elevated as well, thus routine estimation of CgB in all patients seemed not informatic in clinical practice (120).

 

Neuron-Specific Enolase (NSE)

 

NSE is a glycolytic enzyme expressed in the neuroendocrine cells of which levels can be elevated in PNEN patients, particularly those with a poorly-differentiated tumor (58, 110). However, its clinical use is limited as a blood-based biomarkers for NETs because sensitivity and specificity are only 39-43% and 65-73% to distinguish NETs from non-NETs. Consequently, NSE is therefore inferior to CgA in clinical practice (17, 110, 121). When combined with CgA measurement, sensitivity improves and reliability of NET diagnoses increases. Moreover, elevated CgA/NSE levels appear to provide prognostic information on progression-free survival (PFS) and OS (7, 102, 122).  

 

Pancreastatin

 

Pancreastatin, a post-translational processing product of CgA, is suggested to be a useful prognostic marker of NETs as pre-treatment levels > 500 pmol/L are an independent indicator of poor prognosis. Moreover, this marker is reported to correlate with the number of liver metastases and an increase in pancreastatin levels after treatment with SSAs is associated with poorer survival (52, 123). For diagnosis of NETs, pancreastatin is less sensitive than CgA, but also less susceptible to non-specific elevation (52, 121).

 

Pancreatic Polypeptide (PP)

 

PP is a hormone predominantly produced in pancreatic polypeptide cells, located in the head of the pancreas (7, 17, 109, 124). When used alone, a sensitivity of 63% is achieved in PNENs, but when combined with CgA sensitivity increases to 94%, better than either marker alone (109). However, less than 50% of PNEN patients display elevated serum levels and increases do not correlate with tumor burden and/or aggressiveness (102, 124). Moreover, there are several clinical conditions that can induce falsely elevated levels such as physical exercise, hypoglycemia, and food intake, whereas diarrhea, laxative abuse, high age, inflammatory processes and chronic renal disease could lead to a decrease (7, 102).   

 

Other Protein-Based Markers

 

Besides the above-mentioned markers, ProGRP and Neurokinin A can be used to further improve diagnostic and especially prognostic information. ProGRP in fact stimulates cell proliferation which is why increased levels are often associated with a more aggressive tumor and therefore worse prognosis (7, 12). In addition, several markers were reported to be useful for the detection of bone metastases that can be either osteolytic or osteoblastic. Bone alkaline phosphatase (BAP) indicates osteoblast function, while urinary N-telopeptide reflects osteoclast activity or bone resorption. An increased osteoclast activity predicts a poor outcome (12, 125, 126).    

 

Circulating Tumor Cells (CTCs)

 

CTCs have been investigated in a wide range of tumor types, and have gained increased interest in PNENs due to the limitations of the current circulating markers (98-100, 104, 121, 127, 128). The recently developed platform, CellSearch®, allows to detect and isolate CTCs based on expression of the epithelial cell adhesion molecule (EpCAM) on the cell membrane. EpCAM is a transmembrane epithelial glycoprotein that is overexpressed in adenocarcinoma, but recent studies (127-129) revealed EpCAM positivity in ileal, pancreatic, unknown primary, and gastric NETs as well. However, only 21-24% of the metastatic PNEN patients had detectable CTCs in the blood stream, which could potentially be explained by a slow shedding of CTCs or loss of EpCAM expression. Presence of CTCs was associated with increasing tumor burden and grade, while CgA failed to reveal this relationship. Changes in CTC levels were associated with treatment response and OS, revealing its potential as marker during treatment follow-up (127, 129). Furthermore, presence of CTCs could distinguish between patients suffering from PNENs with and without bone metastases with an area under the curve (AUC) of 79% (130). A phase II PAZONET study, during which Pazopanib treatment was evaluated, even demonstrated that patients without baseline CTCs showed improved response and longer median PFS (131). Contrarily, the CALM-NET phase IV study reported no notable effect of the presence of CTCs at baseline on PFS in patients treated with Lanreotide (132). Lastly, CTCs provide the opportunity to detect (epi)genetic alterations in PNENs through DNA and RNA extraction, but they can also be used to determine the SSTR status via immunohistochemistry which could facilitate therapeutic management (133, 134).

 

Circulating Tumor DNA (ctDNA)

 

ctDNA is the fraction of cell-free DNA (cfDNA) that originates from the tumor and constitutes one of the most promising new markers. It provides a representation of the whole tumor and contains tumor-specific genetic and epigenetic alterations, which allow to distinguish healthy from tumoral DNA (98-100, 104, 135). However, ctDNA research regarding NENs is still in its infancy. Boons et al., published the first paper confirming the presence of ctDNA in the plasma of metastatic PNEN patients by looking for tumor-specific single nucleotide variants (SNVs) via custom digital droplet PCR (ddPCR). In patients with localized PNENs, ctDNA could not be detected (136). In the same study, they revealed a significant correlation between CNA profiles of PNEN tissue and ctDNA and demonstrated the feasibility to detect ctDNA using these profiles (136). These findings were exploited in a more recent study, where they performed a cfDNA CNA analysis in a cohort of 43 NEN patients. Using this analysis, ctDNA could be detected in 13 of the 21 PNEN patients. ctDNA positivity appeared to be significantly associated with higher WHO grade, location of the primary tumor and higher levels of CgA and NSE. Besides, a worse OS was observed in ctDNA-positive patients. In addition, they illustrated that CNA patterns in cfDNA could even assist in distinguishing PNENs from the more common PAADs. Moreover, the longitudinal tumor fraction (i.e., amount of ctDNA vs. total cfDNA) measurements were associated with PFS and could indicate tumor progression (47).  

 

MicroRNA (miRNAs)

 

miRNAs are short noncoding RNAs (< 30 nucleotides) designated to regulate many processes including cell proliferation, apoptosis, and development (134, 137), by inducing translational repression or degradation of certain mRNAs (138). In cancer, miRNA regulation is often altered as is the case in PNENs (138). Normal pancreatic islets and PNENs display a distinctly different miRNA profile as PNENs express miRNA-103 and-107, but lack miRNA-155. A set of 10 miRNAs was even able to perfectly distinguish 40 PNENs from 4 PAADs (137). miRNA-204 was overexpressed in insulinomas only, miRNA-196a had a prognostic function and overexpression of miRNA-21 was associated with higher Ki-67 rates and presence of liver metastasis (137). A more recent study, demonstrated that the combination of a set of miRNAs together with CgA measurements could improve diagnostic accuracy (139). However, data on circulating miRNA is still scarce as miRNA measurements in NETs are not properly standardized, requiring further research (140).

 

NETest

 

The NETest, a blood-based multi-analyte transcript assay, was developed in 2013 by Modlin and colleagues (141). The expression of 51 marker genes, encompassing genes associated with NENs, is examined using a quantitative PCR (qPCR) and analyzed using multivariate algorithms (142, 143). These algorithms enable the calculation of a disease-activity score, ranging from 0 to 100, with scores higher than 20 representing tumoral samples (140, 142, 143). The NETest captures accurate diagnosis and tumor biology of NETs with the most recent study demonstrating an accuracy of more than 91%. More specifically, the NETest has proven useful for diagnosing PNEN patients, as PNENs could be distinguished from other pancreatic malignancies with an accuracy of 94% (142, 143). The test also shows to be a real-time monitor of clinical status through the disease-activity score of NEN patients. Low biological activity corresponds to a score of less than 40, while intermediate and high biological activity, indicating tumor progression, have scores of 41-79 and 80-100, respectively (103, 140, 143). Stable and progressive disease could be differentiated with an accuracy of 84.5-85.6%, consistent with image-based categorizations (103). Moreover, changes in NETest disease-activity scores over time correlated with response to treatments including SSAs, PRRT, and surgery (144-147). For example, in a prospective analysis, performed by Modlin and colleagues, 35 pancreatic and small intestine NEN patients were included that all displayed elevated NETest levels prior to surgery, while only 14 of them had increased CgA levels. After tumor removal, the disease-activity scores reduced from 80 ± 5 to 29 ± 5 (p < 0.0001), whereas changes in CgA levels did not correlate with resection. Four of the 11 patients with complete tumor resection still presented increased NETest scores one month after surgery and showed positive evidence of recurrence 6 months post-surgery (144).

 

Since 2013, the NETest has proven to perform better than the single analyte tests (e.g., CgA) and these results appeared to be highly robust and reproducible (103, 140, 142). However, a large independent validation study conducted in the Netherlands has revealed that the test is more sensitive, but less specific than CgA suggesting its suitability as a marker for disease follow-up, but not as a screening tool (147). This test is not affected by food intake or specific medication, is easy to use and available which all increases clinical utility (140). The NETest possess advantages from an economic point of view too. Identifying patients with molecularly stable disease (SD) could potentially lead to fewer use of imaging modalities. Moreover, by enabling faster identification of the clinical status than with imaging, ineffective therapies can be ceased more quickly with another obvious cost-benefit effect (140, 142). Despite all advantages, NETest is currently not implemented in a clinical setting. Results of additional independent validation studies and other practical aspects such as costs and transparency will ultimately determine its integration in clinical practice.

 

MANAGEMENT OF PNENs

 

With a better understanding of the heterogeneity in PNENs, the number of treatment options has increased substantially over the years. Unfortunately, there is a lack of head-to-head comparison data. Therefore, treatment must be individualized considering the age and overall health of the patient, the specific toxicities of potential treatment(s), costs, and potential impact on quality of life (QOL). Consequently, decisions with regard to patient management must be made by an experienced, multidisciplinary team together with the primary care physician (52). Generally, the management of PNEN patients consists of a series of well-defined steps. These comprise of: 1) establishing a diagnosis, 2) determining localization and extent of tumor, 3) controlling hormone excess state in case of F-PNENs, 4) resecting tumor, if possible, 5) checking for presence of hereditary disease (MEN1), 6) treating advanced and metastatic PNENs, and 7) long-term monitoring for tumor progression (63, 148).

Figure 6. Possible treatment scheme for PNENs based on functionality and extent of the tumor. This figure has been adapted from Cancers, Akirov A., Larouche V., Alshehri S. et al., Treatment options for pancreatic neuroendocrine tumors, 11 (6) © 2019 (149).

It is crucial to consider grade/differentiation, stage/extent, and functional status of the tumor as different treatment schemes evolved based on these factors (Figure 6). For example, surgery is usually advocated for PNENs that are functional, larger than 2 cm, or intermediate-to-high grade (3, 8, 52). For patients with metastatic disease, the treatment options are extensive and encompass surgical debulking, systemic therapies including chemotherapy, or targeted therapy such as liver-directed therapy and peptide receptor radionucleotide therapy (PRRT) (149). It is not unusual for the management plan to change based on treatment response and disease progression. Failure to respond to treatment or unexpected changes in the tempo of disease due to tumor dedifferentiation and tumor heterogeneity are well-described in PNENs. Accordingly, most patients will receive multiple treatments during the course of their disease, but there is no data on the optimal treatment sequence (52, 105). The various treatment modalities are discussed below.

 

Surgical Management

 

Surgery continues to play a major role in the management of patients with PNENs as it remains the only potentially curative treatment for PNEN patients and it can alleviate clinical symptoms caused by excessive hormone production and tumor bulk (2, 3, 7, 149-151). Furthermore, several studies revealed that patients who underwent surgical resection had a reduced risk of metastases as well as showed an improved disease-free survival (DFS) (152, 153). Different approaches exist such as resection of the primary tumor and surrounding lymph node metastases through pancreaticoduodenectomy (Whipple procedure) and pancreatectomy (central or distal) as well as the more conservative methods including sparing enucleation and wait-and-see observations (7, 52).

 

Choosing the appropriate approach depends on the extent and location of the tumor, the functional status as well as the presence or absence of metastases (52, 150, 154-156). Generally, surgery is recommended in patients with localized NF-PNENs. Besides the primary tumor, peritumoral metastases should be eradicated as well since nodal metastases occur in at least 30% of NF-PNEN patients which affect tumor grade, but more importantly DFS (157, 158). Exceptions occur in patients with sporadic, low-grade (G1/G2) NF-PNENs smaller than 2 cm (3, 8, 52, 149, 150, 154-156). For those patients, optimal management is controversial as some recommend surgical interventions such as enucleation, whereas others including the ENETS advocate a wait-and-see attitude due to the indolent nature of these tumors (3, 8, 52, 148-150, 159, 160). A similar conservative approach is encouraged in MEN1 patients with NF-PNENs of 2 cm or smaller as these tend to have a low disease-specific mortality (161). Thakker and colleagues, on the other hand, suggest resection of NF-PNENs larger than 1 cm that demonstrate significant growth over 6 to 12 months (162).

Surgical excision of the tumor is also recommended for patients with F-PNENs as these display high cure rates (2, 13, 149, 163-165). The National Comprehensive Cancer Network (NCCN) guidelines describe that insulinomas and gastrinomas are preferably removed by enucleation with peritumoral node dissection if the tumor is located in the head of the pancreas. Deeper, more invasive tumors are more appropriately eradicated by pancreaticoduodenectomy. The former strategy should also be applied for small peripheral glucagonomas and VIPomas. PNENs in the distal part of the pancreas, in turn, are ideally removed through distal pancreatectomy (149). Surgery for MEN1 patients with NF-PNENs and gastrinoma remains controversial as they often present with multiple primary tumors which renders curative surgery almost impossible. Aggressive resection of all PNENs smaller than 2 cm in MEN1 patients seems contra-indicated as several studies revealed that these patients rarely develop advanced disease and have a good prognosis (2, 18, 75, 165-167).

 

The traditional surgical approach is open laparotomy as this allows thorough abdominal exploration including bimanual palpation and intraoperative ultrasound of the pancreas and liver (2, 168). However, several studies reported that certain lesions in particular those amenable to enucleation or to distal pancreatectomy may be approached with laparoscopic or robotic techniques (169). Venkat and colleagues even demonstrated that patients who underwent laparoscopic resection had less blood loss and a lower overall complication rate, and were consequently permitted to leave the hospital sooner than patients who had had open pancreatic resection (170). Gastrinomas form an exception since palpation plays an important role in the detection of these often small malignancies. Moreover, 60-90% of these patients will have lymph node metastases in addition to the primary tumor (169, 171). Adopting a purely laparoscopic approach to these tumors will depend upon improvements in haptic feedback technology. For tumors requiring a Whipple procedure both laparotomic and laparoscopic approaches are used in centers worldwide as the latter is still associated with technical difficulties. However, when performed by trained hepatobiliary or laparoscopic surgeons’ effectiveness and safety are similar and, in some cases, even superior to open surgery (168, 171).     

 

In patients with distant metastases, surgical intervention remains important, although it may no longer result in cure (52). The most common site of distant metastasis is the liver since 46-93% of NET patients develop liver metastases which can lead to liver failure, a common cause of death (52, 172-174). There are multiple options available for patients with hepatic metastases, including surgical resection which, in selected cases, appears to improve survival in uncontrolled series (157). The optimal approach depends on several factors including the extent of primary tumor and liver metastases, planned treatment as well as the age and overall health of the patient. Accordingly, the NCCN recommends complete resection (R0 resection) of primary tumor and liver metastases, if possible and otherwise consider tumor debulking (149). Aggressive resection of the primary tumor in the setting of liver metastases is associated with a survival benefit as both obstruction and further metastatic spread may be prevented. The 5-year survival rate after this surgery ranged from 65% to 73% which is significantly better than that of patients with nonresectable metastases (20%) although this difference might be at least partially explained by selection bias, where only very fit patients receive surgery (174-176). In case of the latter, numerous non-surgical options are available (see liver-directed therapy) and primary tumor, when found to be asymptomatic and stable, is not removed (52, 177). However, R0 resection can only be achieved in 10-20% of the cases as the majority of patients presents with multifocal and bilateral metastases and studies suggest that only one third of all liver metastasis are visible on imaging (52, 174, 175, 178, 179). Consequently, cytoreductive hepatic surgery is more frequently opted for, but this approach remains controversial as it is incomplete and the target population is not clearly described. It is therefore generally considered that patients with metastatic G1/G2 PNEN in which preferably less than 25% of the liver is affected are eligible for tumor debulking (2, 52). Several studies already showed that this procedure can alleviate clinical symptoms in F-PNENs, but also provide better long-term survival (2, 52, 149, 180-183). Moreover, debulking may also be associated with an improved response to concomitant therapy such as embolization (184). Radiofrequency ablation (RFA) is increasingly used in PNEN patients to address hepatic metastases and is often performed during surgery or laparoscopically (2). Patients with extensive liver involvement, who are consequently ineligible for R0 resection and tumor debulking, may be aided with a liver transplant to improve life expectancy (52, 176, 185). A non-randomized study in 88 patients who met strict criteria of transplant eligibility reported a difference in OS in the transplant (88.8%) and no transplant group (22.4%) after 10 years (185). Important concerns are the availability of the grafts as well as the lifelong immunosuppression required after transplantation. Also, the exact criteria for eligibility are very similar to those for tumor debulking which makes patient selection difficult (52).

 

Medical Therapy

 

Use of medical therapy is limited to those with locally advanced or metastatic disease. Some of the current and promising options for targeted systemic therapy are shown in Figure 7.

Figure 7. Overview of the current (blue) and promising (red) options for targeted medical therapy in (P)NETs. This figure has been retrieved from Drugs, Herrera-Martinez A. D., Hofland J., Hofland L. J., Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives 79:21–42 © 2019 (186).

Figure 8. Visualization binding affinity of each of the three FDA-approved SSAs to the different SSTR subtypes. This figure was retrieved and adapted from Drugs, Herrera-Martinez A. D., Hofland J., Hofland L. J., et al., Targeted systemic treatment of neuroendocrine tumors: current options and future perspectives, 79:21-42. © 2019 (186).

SOMATOSTATIN ANALOGS  

 

As previously described in the functional imaging section, SSTRs are often highly overexpressed in PNENs. Several SSAs including Octreotide (Sandostatin®) and Lanreotide (Somatuline®), which have affinity for different SSTR subtypes (Figure 8), were therefore marketed. These inhibit hormone secretion and thus reduce clinical symptoms in patients with F-PNENs (149, 186). Additionally, several studies revealed that SSAs are also capable to control tumor growth with a positive impact on PFS. The antiproliferative effect of SSAs in PNETs was confirmed in the Controlled Study of Lanreotide Antiproliferative Response in Neuroendocrine Tumors (CLARINET) trial. A total of 204 patients with well-differentiated, progressive NETs were included who were then randomly assigned to either Lanreotide or placebo treatment for 96 weeks. After 24 weeks, PFS rates were 65.1% and 33.0% in the Lanreotide and placebo groups respectively (Figure 9). The study also demonstrated that there was no significant difference in QOL and OS in both groups (149, 186-189).

Figure 9. PFS among patients that received Lanreotide (red) or placebo (blue). This figure was retrieved and adapted from The New England journal of medicine, Caplin M. E., Pavel M., Cwikla J. B., et al., Lanreotide in metastatic enteropancreatic neuroendocrine tumors, 371 (3): 224-33 © 2014 (189).

As an extension to the core CLARINET study, the CLARINET open-label extension (OLE) reported long-term safety and additional efficacy data. For this purpose, 88 patients with SD were selected from the core study. Forty-one patients continued their Lanreotide treatment, while 47 patients shifted from placebo to Lanreotide. Safety and tolerability were favorable during a mean treatment period of 40 months. In addition, adverse effects, that were either attributable or not to Lanreotide, were found to improve as duration of treatment, hence exposure, increased. Median PFS in patients who had already received Lanreotide in the core study was estimated at 32.8 months (Figure 10). Of the 32 placebo-treated patients who exhibited progressive disease (PD) in the core study, 17 patients persisted in PD, while the remaining 15 patients had a median time to progression (TTP) of 14 months (187, 190). Based on the findings from the CLARINET trial, the use of SSAs as first-line treatment for symptom relief and tumor control is recommended in the NCCN and ENETS guidelines for advanced, well-differentiated, unresectable PNENs, particularly those with a high burden of liver metastases (149, 188, 191).

Figure 10. PFS of patients that received Lanreotide in the core and OLE CLARINET study. The OLE data is only visualized for patients that were originally assigned to and continued the Lanreotide treatment. This figure was retrieved and adapted from Endocrine-related cancer, Caplin M. E., Pavel M., Cwikla J. B., et al., Anti-tumor effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study, 23 (3): 191-9 © 2016 (190).

MOLECULAR-TARGETES AGENTS  

 

Newly developed molecular-targeted treatments including Sunitinib and Everolimus (Figure 7) have shown to improve PFS in advanced, metastatic PNENs and represent the most common second line treatments that are currently available (52, 149). An overview of the most recent findings can be found in Table 3.    

 

The tyrosine kinase inhibitor (TKI), Sunitinib, has been approved for the treatment of patients with well-differentiated, unresectable, locally advanced or metastatic PNENs as it displays an antiangiogenic working mechanism. It actually inhibits vascular endothelial growth factor receptors (VEGFR) 1 and 3, stem cell factor (SCF) receptor as well as platelet-derived growth factor receptors (149, 186, 192). A two-cohort phase II study, examining 107 patients with advanced NETs (of which 66 PNENs), reported an overall objective response rate (ORR) of 16.7% and SD in 68% of PNEN patients. Median TTP was 7.7 months in PNENs and 10.2 months in carcinoid patients (193). A phase III multinational, randomized, double-blind, placebo-controlled trial (SUN 1111) confirmed the activity of Sunitinib in patients with advanced, well-differentiated PNENs (Figure 11A). A total of 171 patients were enrolled in this study. Median PFS was 11.4 months in the Sunitinib group compared to 5.5 months in the placebo group, with the latter group having a higher mortality rate (25% vs 10%) (194). A retrospective analysis of the previous phase III trial reported an increased PFS in both the Sunitinib and placebo group (12.6 vs. 5.8 months). Median OS after 5 years were 38.6 and 29.1 months of the Sunitinib and placebo groups, respectively. Important to note here is that 69% of the placebo-treated patients shifted to Sunitinib treatment (195). Sunitinib presented with an acceptable safety profile as the most frequent adverse effects in the sunitinib group included diarrhea, nausea, vomiting, asthenia, and fatigue which can be managed through dose interruption or modification (192, 194, 196).  

 

Everolimus (Afinitor®) is an oral, protein kinase inhibitor of the mammalian target of rapamycin (mTOR) pathway that displays proven antitumor activity in advanced PNENs, either alone or combined with Octreotide therapy. A multinational phase II study, the RADIANT 1 trial, has reported the efficacy of Everolimus alone and in combination with Octreotide in patients with metastatic PNENs that have progressed on chemotherapy (197). Monotherapy with Everolimus provided SD in 67.8% of patients and a partial response (PR) in 9.6%, while combination therapy resulted in 80% SD and 4.4% PR. Everolimus treatment also led to a decrease in CgA and NSE levels in 50.7% and 68.2% of the patients (Table 4). An early tumor marker response (i.e., > 50% decrease by 4 weeks) was associated with a significantly longer PFS (197). The RADIANT 3 trial, later on, investigated Everolimus as first line therapy in patients with advanced PNENs (Figure11B). Four hundred and ten patients with radiologic progression of disease were randomized to either Everolimus (10 mg once daily) or placebo. The median PFS was 11 months with Everolimus compared to 4.6 months with placebo representing a 65% reduction in estimated risk of progression or death. The proportion of patients alive and progression free at 18 months was 34% with Everolimus compared to 9% with placebo. Toxicities were mostly grade I or II (198). Similar PFS rates were reported regardless of whether patients were chemo-naïve or had received prior chemotherapy (199, 200). Addition of Pasireotide to Everolimus did not improve PFS compared to Everolimus alone (201).

 

Based on the recent, above-mentioned data, the European Society for Medical Oncology (EMSO) guidelines 2020 recommend the use of molecular-targeted agents such as Sunitinib and Everolimus in advanced, progressive PNENs (G1/G2) (191). Likewise, the North American Neuroendocrine Tumor Society (NANETS) guidelines 2020 recommend both treatments for well-differentiated, metastatic PNETs (G1/G2) (202). Both guidelines state there is no support to use Sunitinib nor Everolimus in treatment of PNET G3 or PNECs (191, 202). When comparing both molecular-targeted agents, response rates appear comparable (Figure 11). Since there has been no trial comparing the two agents directly, choice of agent may be based on the potential side-effects and patient’s overall health. For example, in patients with poorly-controlled hormonal symptoms, especially hyperinsulinism, congestive heart failure, hypertension, high risk of gastrointestinal bleeding or a history of myocardial infarction or stroke, Everolimus is thought to be the preferred choice (194, 202, 203). On the other hand, in patients with poorly controlled diabetes mellitus, pulmonary disease, or high risk of infection, sunitinib would be a more appropriate choice (192, 203). Moreover, up until now several biomarkers have been identified that correlated with the patient’s outcome. An overview of the currently known biomarkers can be found in Table 4 (204).

 

 Table 3. Results from Most Important Phase II and III Studies of Sunitinib and Everolimus in PNENs

Study

Patients

Active treatment

PD at entry

ORR

PFS/TTP (months)

Safety and other comments

Sunitinib

Phase II, open label (193)

N = 107 

 

- PNETs = 66

 

 

- Carcinoid = 41

 

50 mg daily

Schedule 4/2*

No

 

 

ORR = 16.7%

SD = 68%

 

ORR = 2.4%

SD = 83%

 

 

TTP = 7.7

 

 

TTP = 10.2

G3 fatigue: 24.3%

Phase III,

RCT,

SUN 1111 (194)

 

[Retrospect]

(195)

N = 171

 

- Sunitinib = 86

 

 

 

- Placebo = 85

 

 

37.5 mg daily

CDD**

Yes

 

 

ORR = 9.3%

SD = 63%

PD = 14%

 

ORR = 0%

SD = 60%

PD = 27%

 

 

PFS = 11.4

[PFS = 12.6]

 

 

PFS = 5.5

[PFS = 5.8]

Common AEs:

30%: diarrhea, nausea, asthenia, vomiting and fatigue 

 

10-12%: G3/4 neutropenia and hypertension

Everolimus

Phase II,

open label,

RADIANT 1 (197)

N = 160

 

- Stratum1 = 155

 

 

 

- Stratum2 = 45

 

 

10 mg daily

 

 

 

10 mg daily + 30 mg LAR Octreotide

Yes

 

 

PR = 9.6%

SD = 67.8%

PD = 13.9%

 

PR = 4.4%

SD = 80%

PD = 0%

 

 

PFS = 9.7

 

 

 

PFS = 16.7

 

Specific AEs:

 

5.2%: G3/4 asthenia

 

 

8.9%: G3/4 thrombocytopenia

 

Common AEs:

30%: stomatitis, rash, diarrhea, fatigue, nausea

Phase III,

RCT,

RADIANT 3 (198)

N = 410

 

- Everolimus = 207

 

 

- Placebo = 203

10 mg daily

Yes

 

 

PR = 5%

SD = 73%

 

PR = 2%

SD = 51%

 

 

PFS = 11

 

 

PFS = 4.6

 

Common AEs:

64%: stomatitis

49%: rash

34%: diarrhea

31%: fatigue

23%: infections

Abbreviations: ORR, objective response rate; PFS, progression-free survival; TTP, time to progression; SD, stable disease; PD, progressive disease; CDD, continuous daily dosing; AE, adverse event; LAR, long-acting release; PR, partial response; RCT, randomized clinical trial

* Concomitant use of SSA in 27% of PNET patients and 54% of patients with carcinoid tumors.

** Concomitant use of SSA in 26.7% of patients.  

 

Figure 11. This figure compares the PFS in patients with advanced metastatic PNENs, (A) treated with Sunitinib in the SUN 1111 trial (194) and (B) Everolimus in the RADIANT 3 trial (198). Figure A was retrieved and adapted from The New England journal of medicine, Raymond E., Dahan L., Raoul J. L., et al., Sunitinib malate for the treatment of pancreatic neuroendocrine tumors, 364 (6): 501-13 © 2011 (194). Figure B was retrieved and adapted from The New England journal of medicine, Yao J. C., Shah M. H., Ito T., et al., Everolimus for advanced pancreatic neuroendocrine tumors, 364 (6): 514-23 © 2012 (198).

 

Table 4. Current Soluble Biomarkers and Correlations with Outcomes with Targeted Therapies in PNENs

Study

Biomarker

Results

Sunitinib

(204, 205)

 

VEGF

Increased in 53% of patients after 4 weeks of treatment

Return to baseline after 2 weeks off treatment

When Sunitinib level > 50 ng/dL higher changes observed

No significant difference between PNET and carcinoids

sVEGFR

Decrease of ³ 30% in sVEGFR-2 and -3 levels

Return to baseline after 2 weeks off treatment

Reduction in sVEGFR-3 correlated with better OR and PFS

Lower baseline sVEGFR-2 with radiological SD for > 6 months

Elevated baseline sVEGFR-2 correlated with improved OS

IL-8

Increase (>2-fold) in 43% and (>3-fold) in 23% of patients after 4 weeks of treatment

Return to baseline after 2 weeks off treatment

Increase (1.8-fold) after 4 weeks on treatment

SDF-1a

Increase (20%) after 4 weeks on treatment

Elevated baseline correlated with significantly shorter TTP, PFS and OS

Everolimus

(197, 206)

 

CgA

Increase (> 2-fold) of CgA at baseline correlated with decreased PFS and OS

Reduction of > 30% in CgA levels after 4 weeks correlated with increased PFS and OS

NSE

Elevated NSE levels at baseline correlated with decreased PFS and OS

Reduction of > 30% in NSE levels after 4 weeks correlated with improved PFS

Abbreviations: VEGF, vascular endothelial growth factor; sVEGFR, soluble VEGFR; SDF-1a, stromal cell-derived factor 1 alpha.

Note: This table was adapted from Molecular Diagnosis and Therapy, Mateo, J., Heymach, J. V. and Zurita, A. J., Biomarkers of response to Sunitinib in gastroenteropancreatic neuroendocrine tumors: current data and clinical outlook, 151-161. © 2012 (204).

 

CYTOTOXIC CHEMOTHERAPY  

 

There is currently no unanimity on which cytotoxic chemotherapy would be optimal for the treatment of PNENs. Therefore, patient selection is key so factors such as primary tumor site and stage, differentiation, and proliferation index should be considered. Conventional cytotoxic chemotherapy is often used as first-line therapy for metastatic and progressive PNETs or PNECs (149, 207). ENETS guidelines describe the following indications: progression under SSA treatment, worsening clinical symptoms, and/or Ki-67 values > 10% (208). In a neoadjuvant setting, chemotherapy can play a potential role in tumor shrinkage prior to resection (7). Two major types of chemotherapeutic agents can be distinguished namely alkylating and platinum agents (7, 149, 207). In practice these are often combined with antimetabolites and anthracyclines (209). An overview of the most commonly used combinatory therapies and when to employ them is described in more detail below.

 

Alkylating agents such as Streptozocin, Dacarbazine, and Temozolomide are key in the treatment strategy of PNEN patients since they are often employed as second line treatment after progression under SSA (207). First of all, Streptozocin, a nitrosourea alkylating agent, is taken up by cells via a glucose protein 2 (GLUT2) after which cell damage is induced. Since the compound is associated with significant renal and hematological toxicity in high doses, it is often combined with 5-fluorouracil (5-FU) or Doxorubicin with dose reduction, hence reduced toxicity as a result (2, 149, 209). A comparative, phase III study conducted in 1992 reported that the combinatory therapy of Streptozocin + Doxorubicin provided more favorable results than Streptozocin + 5-FU in patients with advanced PNENs (210). However, the results described in this study have not been confirmed in any subsequent study (163, 209). Kouvaraki and colleagues retrospectively studied 84 PNEC patients treated with Streptozocin, 5-FU and Doxorubicin. Response rate was 39% with 2-year PFS and OS of 41% and 74%, respectively (211). Dacarbazine, a second alkylating agent, serves as a less toxic alternative. A phase II study tested Dacarbazine as a monotherapy in 50 PNEN patients and reported an ORR of 34% and median OS of 19.3 months (212). When combined with 5-FU, the overall response rate and PFS in advanced NENs were 38.2% and 13.9 months, respectively (213). A third alkylating agent that is primarily used as monotherapy for treatment of glioblastoma and melanoma is Temozolomide (163, 209). When combined with other compounds including Capecitabine (214), Bevacizumab (215), Bevacizumab and Octreotide (216), Thalidomide (217) and Everolimus (200) it shows significant activity in advanced PNENs (149, 209). A 2011 retrospective study reported that the combination treatment Capecitabine + Temozolomide (CAPTEM) in 30 chemonaive NEC patients resulted in an ORR of 70% with a PFS of 18 months (214). In 2018, a prospective, randomized phase II trial investigated Temozolomide therapy versus the CAPTEM combination therapy in PNEN patients. PFS was significantly better in the latter group (14.4 vs. 22.7 months) (218). However, a more recent retrospective analysis showed that CAPTEM was not able to improve PFS. Consequently, it was suggested by the authors that CAPTEM might be more useful for tumor shrinkage rather than improving PFS (219). 

 

In poorly-differentiated G3 NECs, platinum agent regimens are often used in patients with adequate performance status. The first-line chemotherapy for NEC patients encompasses Cisplatin or Carboplatin combined with Etoposide or Irinotecan, based on the reported results (52, 220-223). Moertel and colleagues investigated the effect of Cisplatin + Etoposide in 45 patients with metastatic NENs, of which 27 were well-differentiated. The ORR was 67% in the 18 poorly-differentiated NECs with complete response (CR) in 17% of the patients, while unfortunately, only 2 patients (17%) of the well-differentiated NEN patients showed a response. Moreover, they reported a median survival of 19 months which was significantly longer than those described in literature (6-7 months). However, toxicity was a major issue (220). These results were confirmed by Mitry and colleagues in 1999 (221). Lower toxicity levels were observed when patients were treated with Carboplatin, but efficacy was similar to that of Cisplatin, rendering Carboplatin a valuable alternative (222, 224). Moreover, Oxaliplatin-based therapy appeared to have a greater activity in advanced PNETs (207).

 

The role of second-line chemotherapy for NEC patients is currently unknown, but many combinatory options have been examined (223, 225). Capecitabine + Oxaliplatin (CAPOX) and 5-FU + Oxaliplatin (FOLFOX) have been evaluated in two retrospective trials in well-differentiated NENs. ORRs of 26% and 30% were reported, respectively (226, 227). In addition, FOLFIRI and FOLFIRINOX (5-FU-based chemotherapies) have recently proven some effect in NEC patients progressing on platinum-based regimens (225, 228).

 

Radiotherapy

 

PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

 

Peptide receptor radionuclide therapy (PRRT) is a therapy whereby a radiolabeled SSA (117Lutetium or 90Yttrium) is used to treat SSTR-positive, locally advanced and/or metastatic GEP-NENs, including PNENs. Adverse effects include nausea, renal toxicity, transient bone marrow suppression and seldom myelodysplastic syndrome or acute myeloid leukemia in 1-2% of patients (7, 149, 229, 230).

 

In a study of 504 patients, treatment with the analog 177Lu-DOTATATE showed activity in GEP-NENS (230). Looking specifically at the PNEN subgroup there was a 6% CR and a 36% PR in NF-PNENs and no CR and 47% PRs in functioning PNENs (230). Striking improvements in QOL of responders was also noted (231). A more recent study of 68 patients with PNENs treated with PRRT showed PRs in 41 patients (60.3%), minor responses in 8 (11.8%), SD in 9 (13.2%) and PD in 10 (14.7%) (232). The authors concluded that the outstanding response rates and survival outcomes suggest that PRRT is highly effective in advanced G1/2 PNENs when compared to other treatment modalities. Independent predictors of survival were the tumor proliferation index, the patient’s performance status, tumor burden and baseline plasma NSE level. PRRT also provided improvements in PFS compared to Octreotide in midgut NENs (232). The NETTER-1 phase III trial confirmed the efficacy in PRRT in midgut NENs in 2017 (229, 233, 234). Therefore, the FDA approved use of 177Lu-DOTATATE based on the results obtained in the NETTER-1 trial in midgut NETs (229, 234). Thus, the number of centers where this treatment is available is expected to increase in the US, although it has been used in Europe since 1996. Joint society practice guidelines have been developed (235). There are a number of ongoing international clinical trials listed on Clinical Trials.gov. Third party payer reimbursement is an ongoing issue which hopefully will be resolved.

 

For PNENs, the effects of PRRT have only been investigated in several single-arm prospective and retrospective trials (229). These, however, identified several signals in favor of PRRT use in PNENs as disease control rates and PFS varied between 84-85% and 30-34 months, respectively (232, 236). Additionally, a meta-analysis compared the efficiency of PRRT to Everolimus in GEP-NENs that were not eligible to surgical resection. An ORR of 47% was reported in the PRRT treated subgroup versus only 12% of the Everolimus treated patients. Moreover, disease control rates (81% vs. 73%) as well as PFS (25.7 vs. 14.7 months) were also superior in the PRRT treated subgroup (237). A recent retrospective study evaluated the association of upfront PRRT vs. upfront chemotherapy or targeted therapy with PFS in enteropancreatic NET patients who progressed under SSA treatment. Patients with a Ki-67 value of £10% who received upfront PRRT, were reported to have a statically and clinically meaningful prolonged PFS (238). Based on these findings, it seems important to better define the role of PRRT in the treatment of PNENs within the future.

 

Liver-Directed Therapy

 

As mentioned earlier, the liver constitutes the most common site for distant metastases (52, 172-174). Since surgical resection and RFA are only feasible in a minority of patients, there are multiple liver-directed therapies available to treat the remaining patients. These methods include transarterial chemoembolization (TACE), transarterial embolization (TAE), or radioembolization, which will be discussed below. Given the lack of randomized data, it is difficult to determine with certainty which method is preferred. Moreover, NANETS guidelines recommend to consider systemic therapy rather than liver-directed therapy when >50-75% hepatic tumor burden is present (239).  

 

A study of chemoembolization combined with SSA treatment resulted in a relief of symptoms in 78% of the patients. Monitoring serum pancreastatin levels predicted a response to this therapy in which radiographic improvement or stability were seen in 45% of patients (77). In NEN patients that underwent TACE, plasma levels of pancreastatin above 5000 pg/ml pre-treatment were associated with increased peri-procedure mortality (240).

Radioembolization (also known as selective intrahepatic radiotherapy or SIRT) involves embolization of 90Yttrium embedded either in a resin microsphere (Sir-Sphere) or a glass microsphere (TheraSphere). Acute toxicities associated with 90Yttrium microsphere embolization appear to be lower than other embolization techniques, primarily due to the fact that the procedure does not induce ischemic hepatitis. Thus, the procedure can be performed on an outpatient basis. A rare, but potentially serious complication is radiation enteritis, which can occur if particles are accidentally infused into arteries supplying the gastrointestinal tract. Chronic radiation hepatitis is another potential toxicity. Response rates associated with radioembolization in metastatic NEN patients have been encouraging. In one retrospective multi-center study of 148 patients treated with Sir-Spheres, the objective radiographic response rate was 63% with a median survival of 70 months, with no radiation-induced liver failure (241). Another study of 42 patients treated with either Sir-Spheres or TheraSpheres reported a response rate of 51%. However, only 29 of the 42 enrolled patients were evaluable (242). Grozinsky-Glasberg and colleagues examined 57 patients which underwent either TACE, TAE or SIRT. They reported symptomatic control and a stabilization of tumor growth in 95% of the patients. Noteworthy, they observed improvements regardless of the extent of the liver metastasis (243).

 

Novel Targets for the Treatment of (P)NENs

 

While there has been a quantum leap in the ability to treat NENs successfully we have a long way to go to cure the disease. Fortunately, research into improved and novel therapeutic strategies is ongoing. So far, the results of immunotherapy as monotherapy in PNET patients remain disappointing. Examples include the inhibition of the programmed death-ligand 1 (PDL-1) and cytotoxic T-lymphocyte antigen-4 (CTLA4), by treating patients with Pembrolizumab to enhance the immune response towards tumor cells (186, 244, 245). The KEYNOTE-028 phase I study treated PDL-1 positive PNEN patients with Pembrolizumab and reported an ORR of 6.3%, but no CRs occurred. Responses were better in metastatic carcinoids (ORR: 12%) (246). These findings were confirmed by the KEYNOTE-158 phase II study, in which the ORR was 3.7% with 3 PRs in PNEN and 1 in rectal NEN patients (247). Currently, several other clinical trials that are investigating the antitumor effect of immune checkpoint inhibitors include NCT02939651 for Pembrolizumab and NCT02955069 for other PDL-1 receptor antibodies (247). Moreover, there is also much speculation that PRRT cytotoxic drugs induce genotoxicity, hence increase the neoantigen load and thereby could potentially enhance the efficacy of immunotherapy (149, 244, 245, 248). Bevacizumab, a monoclonal antibody against the VEGF, showed no real benefit in PFS in a phase III trial in which Bevacizumab + Octreotide was compared to Interferon + Octreotide (249). The BETTER phase II trial, on the other hand, demonstrated that Bevacizumab + 5-FU/Streptozocin in patients with metastatic, well-differentiated PNENs could reach a PFS of 23.7 months and they reported an OS at 24 months of 88% (250). The SANET-ep (251) and SANET-p (252) phase III studies examined the efficacy and safety of Surufatinib in extrapancreatic NENs and PNENs, respectively. Surufatinib, a small-molecule inhibitor that targets VEGFRs as well as the fibroblast growth factor (FGF) receptor 1 and macrophage colony-stimulating factor 1 (CSF1) receptor, effectively prolonged PFS in both studies and was therefore suggested a potential treatment option in both patient populations (251, 252).

 

QUALITY OF LIFE IN PATIENTS WITH PNENs

 

The measurement of health-related quality of life (HRQOL) has become essential for evaluating the impact of the disease process and the treatment on patient’s symptoms, social, emotional, physiological, and physical functioning. The European Organization for Research and Treatment of Cancer (EORTC) developed the QLQ-C30 tool for oncology patients (253) and the QLQ-GINET21 tool was specifically developed for a spectrum of NEN patients (28% PNENs) (254). The Norfolk QOLNET was specifically developed for midgut NETs (carcinoids) and may provide some additional advantages for that specific group of patients (231).

 

The most commonly used QOL tool in GEP-NENs (including PNENs) is the EORTC QLQ-C30 (255). SSAs and Sunitinib have shown to improve HRQOL in diverse groups of GEP-NEN patients (255). In the CLARINET study, QLC-C30 data were mapped to EQ-5D utilities and not surprisingly worse utility values were noted with PD compared to SD. Of note, tumor location (midgut vs. pancreas) did not affect utility (256). PNEN patients treated with everolimus showed stable HRQOL scores as opposed to worse scores in non-PNEN patients (Pavel). PRRT treatment of PNEN patients resulted in significantly improved global health status, social functioning and mitigation of physical complaints (257). Thus, data are emerging on HRQOL in PNEN patients. However, most studies are too heterogenous in terms of patient populations and treatment interventions to draw firm conclusions (258). Moving forward, it will be important for HRQOL to be measured as a key component of clinical trials.

 

EXPERT COMMENTARY  

 

After many years of frustration, our knowledge regarding the biology, pathophysiology and genetics of (P)NENs increased. This has led to marked improvements in (functional) imaging, with the development of 68Gallium-labeled SSAs, as well as targeted treatments such as the tyrosine-kinase inhibitor Sunitinib, and the mTOR inhibitor Everolimus. In addition, PRRT seems to be expanding its role in treatment from midgut to PNENs. In the future, both imaging and treatment options will continue to evolve as more specific imaging agents and therapeutic targets are being developed and evaluated in numerous studies. The relatively uncommon nature of PNENs has made designing and completing randomized studies of adequate power challenging for a single institution. Therefore, we encourage the recent trend of multi-institutional, multinational studies in more homogeneous patient populations. We also strongly agree with the recommendation of NANETS, ENETS and other groups that all of these patients should be entered into clinical trials whenever feasible. Determining study availability and patient eligibility has been greatly facilitated by Clinical trials.gov as well as institutional and organizational websites. Enrolling more patients in clinical trials by overcoming barriers to participation will be required to move patient care forward.

 

Unfortunately, to date, the optimal treatment(s) and treatment sequences have yet to be defined. The lack of treatment standardization, the plethora of treatments that most patients receive, and different treatment sequences make it extremely difficult to assess the effectiveness of a particular treatment relative to others. Moreover, head-to-head comparisons are lacking as well. Available consensus guidelines establish broad principles, but are generally not helpful in managing a specific patient. Management has become even more complex given the multiplicity of effective treatments for advanced disease, none of which has convincingly been shown to be superior to the other. Hence, an experienced multidisciplinary team is essential to guide management of these patients. Given relative parity of effectiveness, decisions regarding choice of treatment need to be based on multiple considerations, including patient’s overall health, disease burden, symptomatology, rate of progression, treatment toxicity, effect on QOL, and cost. These considerations will usually lead to one treatment being favored over another.

 

Because of the relatively indolent nature of many (or most) NENs, long-term follow-up to assess differences in treatment outcomes, is required. However, biomarkers that can predict response to a particular therapy are currently not available. We expect that in the future the so-called tumor circulome, especially ctDNA, could play an important role in this as recent studies revealed its potential to diagnose, prognosticate and monitor disease progression.

 

5 Year View

 

Knowledge of the biology and genetics will continue to accumulate, which could potentially lead to further refinements in classification, staging, and personalized treatment. Genetic profiling will become clinically useful as data will accumulate on treatment effectiveness in patient subgroups leading to more tailored therapies. Moreover, biomarkers will be developed that better predict response to a particular therapy. Results of the ongoing clinical trials on newer SSAs and targeted agents will add to the number of available treatments. The role of PRRT in the treatment of PNENs will be better defined. There will be increased knowledge as to optimal treatment sequences. Designing randomized clinical trials of adequate power will remain a challenge for many reasons including the scarcity and indolent growth of these tumors. Consensus guidelines will evolve, but patient management will continue to require an experienced multidisciplinary team.

 

REFERENCES

 

  1. Clift AK, Kidd M, Bodei L, Toumpanakis C, Baum RP, Oberg K, et al. Neuroendocrine Neoplasms of the Small Bowel and Pancreas. Neuroendocrinology. 2020;110(6):444-76.
  2. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135(5):1469-92.
  3. Perri G, Prakash LR, Katz MHG. Pancreatic neuroendocrine tumors. Current opinion in gastroenterology. 2019;35(5):468-77.
  4. Hong X, Qiao S, Li F, Wang W, Jiang R, Wu H, et al. Whole-genome sequencing reveals distinct genetic bases for insulinomas and non-functional pancreatic neuroendocrine tumours: leading to a new classification system. Gut. 2020;69(5):877-87.
  5. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA oncology. 2017;3(10):1335-42.
  6. Daskalakis K. Functioning and nonfunctioning pNENs. Current Opinion in Endocrine and Metabolic Research. 2021;18:284-90.
  7. Ma ZY, Gong YF, Zhuang HK, Zhou ZX, Huang SZ, Zou YP, et al. Pancreatic neuroendocrine tumors: A review of serum biomarkers, staging, and management. World journal of gastroenterology. 2020;26(19):2305-22.
  8. Cives M, Strosberg JR. Gastroenteropancreatic Neuroendocrine Tumors. CA: a cancer journal for clinicians. 2018;68(6):471-87.
  9. Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, et al. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology. 2016;103(2):153-71.
  10. Boons G, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Clinical applications of (epi)genetics in gastroenteropancreatic neuroendocrine neoplasms: Moving towards liquid biopsies. Reviews in endocrine & metabolic disorders. 2019;20(3):333-51.
  11. Liakakos T, Roukos DH. Everolimus and sunitinib: from mouse models to treatment of pancreatic neuroendocrine tumors. Future oncology (London, England). 2011;7(9):1025-9.
  12. Vinik AI. Advances in diagnosis and treatment of pancreatic neuroendocrine tumors. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2014;20(11):1222-30.
  13. Verbeke CS. Endocrine tumours of the pancreas. Histopathology. 2010;56(6):669-82.
  14. Plöckinger U, Wiedenmann B. Diagnosis of non-functioning neuro-endocrine gastro-enteropancreatic tumours. Neuroendocrinology. 2004;80 Suppl 1:35-8.
  15. Inzani F, Petrone G, Rindi G. The New World Health Organization Classification for Pancreatic Neuroendocrine Neoplasia. Endocrinology and metabolism clinics of North America. 2018;47(3):463-70.
  16. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76(2):182-8.
  17. Ciobanu OA, Martin S, Fica S. Perspectives on the diagnostic, predictive and prognostic markers of neuroendocrine neoplasms (Review). Experimental and therapeutic medicine. 2021;22(6):1479.
  18. Jensen RT, Berna MJ, Bingham DB, Norton JA. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer. 2008;113(7 Suppl):1807-43.
  19. Li X, Gou S, Liu Z, Ye Z, Wang C. Assessment of the American Joint Commission on Cancer 8th Edition Staging System for Patients with Pancreatic Neuroendocrine Tumors: A Surveillance, Epidemiology, and End Results analysis. Cancer medicine. 2018;7(3):626-34.
  20. You Y, Jang JY, Kim SC, Yoon YS, Park JS, Cho CK, et al. Validation of the 8th AJCC Cancer Staging System for Pancreas Neuroendocrine Tumors Using Korean Nationwide Surgery Database. Cancer research and treatment. 2019;51(4):1639-52.
  21. Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging. CA: a cancer journal for clinicians. 2017;67(2):93-9.
  22. Yang M, Zeng L, Yao WQ, Ke NW, Tan CL, Tian BL, et al. A comprehensive validation of the novel 8th edition of American Joint Committee on Cancer staging manual for the long-term survivals of patients with non-functional pancreatic neuroendocrine neoplasms. Medicine. 2020;99(46):e22291.
  23. Reid MD, Balci S, Saka B, Adsay NV. Neuroendocrine tumors of the pancreas: current concepts and controversies. Endocrine pathology. 2014;25(1):65-79.
  24. Di Domenico A, Pipinikas CP, Maire RS, Bräutigam K, Simillion C, Dettmer MS, et al. Epigenetic landscape of pancreatic neuroendocrine tumours reveals distinct cells of origin and means of tumour progression. Communications biology. 2020;3(1):740.
  25. Noone TC, Hosey J, Firat Z, Semelka RC. Imaging and localization of islet-cell tumours of the pancreas on CT and MRI. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):195-211.
  26. Taskin OC, Clarke CN, Erkan M, Tsai S, Evans DB, Adsay V. Pancreatic neuroendocrine neoplasms: current state and ongoing controversies on terminology, classification and prognostication. Journal of gastrointestinal oncology. 2020;11(3):548-58.
  27. Gao HL, Wang WQ, Yu XJ, Liu L. Molecular drivers and cells of origin in pancreatic ductal adenocarcinoma and pancreatic neuroendocrine carcinoma. Experimental hematology & oncology. 2020;9:28.
  28. Chan CS, Laddha SV, Lewis PW, Koletsky MS, Robzyk K, Da Silva E, et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nature communications. 2018;9(1):4158.
  29. Cejas P, Drier Y, Dreijerink KMA, Brosens LAA, Deshpande V, Epstein CB, et al. Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nature medicine. 2019;25(8):1260-5.
  30. Strobel O, Rosow DE, Rakhlin EY, Lauwers GY, Trainor AG, Alsina J, et al. Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia. Gastroenterology. 2010;138(3):1166-77.
  31. Anderson CW, Bennett JJ. Clinical Presentation and Diagnosis of Pancreatic Neuroendocrine Tumors. Surgical oncology clinics of North America. 2016;25(2):363-74.
  32. Mimeault M, Batra SK. Recent progress on normal and malignant pancreatic stem/progenitor cell research: therapeutic implications for the treatment of type 1 or 2 diabetes mellitus and aggressive pancreatic cancer. Gut. 2008;57(10):1456-68.
  33. Jiang FX, Morahan G. Multipotent pancreas progenitors: Inconclusive but pivotal topic. World journal of stem cells. 2015;7(11):1251-61.
  34. Zhong F, Jiang Y. Endogenous Pancreatic β Cell Regeneration: A Potential Strategy for the Recovery of β Cell Deficiency in Diabetes. Frontiers in endocrinology. 2019;10:101.
  35. Yamaguchi J, Liss AS, Sontheimer A, Mino-Kenudson M, Castillo CF, Warshaw AL, et al. Pancreatic duct glands (PDGs) are a progenitor compartment responsible for pancreatic ductal epithelial repair. Stem cell research. 2015;15(1):190-202.
  36. Tirosh A, Kebebew E. Genetic and epigenetic alterations in pancreatic neuroendocrine tumors. Journal of gastrointestinal oncology. 2020;11(3):567-77.
  37. Görtz B, Roth J, Krähenmann A, de Krijger RR, Muletta-Feurer S, Rütimann K, et al. Mutations and allelic deletions of the MEN1 gene are associated with a subset of sporadic endocrine pancreatic and neuroendocrine tumors and not restricted to foregut neoplasms. The American journal of pathology. 1999;154(2):429-36.
  38. Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocrine reviews. 2019;40(2):506-36.
  39. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65-71.
  40. Crona J, Skogseid B. GEP- NETS UPDATE: Genetics of neuroendocrine tumors. European journal of endocrinology. 2016;174(6):R275-90.
  41. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science (New York, NY). 2011;331(6021):1199-203.
  42. Gläsker S, Vergauwen E, Koch CA, Kutikov A, Vortmeyer AO. Von Hippel-Lindau Disease: Current Challenges and Future Prospects. OncoTargets and therapy. 2020;13:5669-90.
  43. Zwolak A, Świrska J, Tywanek E, Dudzińska M, Tarach JS, Matyjaszek-Matuszek B. Pancreatic neuroendocrine tumours in patients with von Hippel-Lindau disease. Endokrynologia Polska. 2020;71(3):256-9.
  44. Parekh VI, Modali SD, Welch J, Simonds WF, Weinstein LS, Kebebew E, et al. Frequency and consequence of the recurrent YY1 p.T372R mutation in sporadic insulinomas. Endocrine-related cancer. 2018;25(5):L31-l5.
  45. Pipinikas CP, Berner AM, Sposito T, Thirlwell C. The evolving (epi)genetic landscape of pancreatic neuroendocrine tumours. Endocrine-related cancer. 2019;26(9):R519-r44.
  46. Singhi AD, Klimstra DS. Well-differentiated pancreatic neuroendocrine tumours (PanNETs) and poorly differentiated pancreatic neuroendocrine carcinomas (PanNECs): concepts, issues and a practical diagnostic approach to high-grade (G3) cases. Histopathology. 2018;72(1):168-77.
  47. Boons G, Vandamme T, Mariën L, Lybaert W, Roeyen G, Rondou T, et al. Longitudinal Copy-Number Alteration Analysis in Plasma Cell-Free DNA of Neuroendocrine Neoplasms is a Novel Specific Biomarker for Diagnosis, Prognosis, and Follow-up. Clinical cancer research : an official journal of the American Association for Cancer Research. 2021.
  48. House MG, Herman JG, Guo MZ, Hooker CM, Schulick RD, Lillemoe KD, et al. Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Annals of surgery. 2003;238(3):423-31; discussion 31-2.
  49. Neiman D, Moss J, Hecht M, Magenheim J, Piyanzin S, Shapiro AMJ, et al. Islet cells share promoter hypomethylation independently of expression, but exhibit cell-type-specific methylation in enhancers. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(51):13525-30.
  50. Boons G, Vandamme T, Ibrahim J, Roeyen G, Driessen A, Peeters D, et al. PDX1 DNA Methylation Distinguishes Two Subtypes of Pancreatic Neuroendocrine Neoplasms with a Different Prognosis. Cancers. 2020;12(6).
  51. Lakis V, Lawlor RT, Newell F, Patch AM, Mafficini A, Sadanandam A, et al. DNA methylation patterns identify subgroups of pancreatic neuroendocrine tumors with clinical association. Communications biology. 2021;4(1):155.
  52. Scott AT, Howe JR. Evaluation and Management of Neuroendocrine Tumors of the Pancreas. The Surgical clinics of North America. 2019;99(4):793-814.
  53. Lee DW, Kim MK, Kim HG. Diagnosis of Pancreatic Neuroendocrine Tumors. Clinical endoscopy. 2017;50(6):537-45.
  54. Rebours V, Cordova J, Couvelard A, Fabre M, Palazzo L, Vullierme MP, et al. Can pancreatic neuroendocrine tumour biopsy accurately determine pathological characteristics? Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2015;47(11):973-7.
  55. Klöppel G, Couvelard A, Perren A, Komminoth P, McNicol AM, Nilsson O, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: towards a standardized approach to the diagnosis of gastroenteropancreatic neuroendocrine tumors and their prognostic stratification. Neuroendocrinology. 2009;90(2):162-6.
  56. Eusebi LH, Thorburn D, Toumpanakis C, Frazzoni L, Johnson G, Vessal S, et al. Endoscopic ultrasound-guided fine-needle aspiration vs fine-needle biopsy for the diagnosis of pancreatic neuroendocrine tumors. Endoscopy international open. 2019;7(11):E1393-e9.
  57. Crinò SF, Ammendola S, Meneghetti A, Bernardoni L, Conti Bellocchi MC, Gabbrielli A, et al. Comparison between EUS-guided fine-needle aspiration cytology and EUS-guided fine-needle biopsy histology for the evaluation of pancreatic neuroendocrine tumors. Pancreatology : official journal of the International Association of Pancreatology (IAP)[et al]. 2021;21(2):443-50.
  58. Garcia-Carbonero R, Sorbye H, Baudin E, Raymond E, Wiedenmann B, Niederle B, et al. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016;103(2):186-94.
  59. Fang JM, Shi J. A Clinicopathologic and Molecular Update of Pancreatic Neuroendocrine Neoplasms With a Focus on the New World Health Organization Classification. Archives of pathology & laboratory medicine. 2019;143(11):1317-26.
  60. Perren A, Couvelard A, Scoazec JY, Costa F, Borbath I, Delle Fave G, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pathology: Diagnosis and Prognostic Stratification. Neuroendocrinology. 2017;105(3):196-200.
  61. Rockall AG, Reznek RH. Imaging of neuroendocrine tumours (CT/MR/US). Best practice & research Clinical endocrinology & metabolism. 2007;21(1):43-68.
  62. Virgolini I, Traub-Weidinger T, Decristoforo C. Nuclear medicine in the detection and management of pancreatic islet-cell tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):213-27.
  63. Gibril F, Jensen RT. Diagnostic uses of radiolabelled somatostatin receptor analogues in gastroenteropancreatic endocrine tumours. Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2004;36 Suppl 1:S106-20.
  64. Sundin A, Garske U, Orlefors H. Nuclear imaging of neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):69-85.
  65. Singh A, Hines JJ, Friedman B. Multimodality Imaging of the Pancreatic Neuroendocrine Tumors. Seminars in Ultrasound, CT and MRI. 2019;40(6):469-82.
  66. Chiti G, Grazzini G, Cozzi D, Danti G, Matteuzzi B, Granata V, et al. Imaging of Pancreatic Neuroendocrine Neoplasms. International journal of environmental research and public health. 2021;18(17).
  67. Morse B, Al-Toubah T, Montilla-Soler J. Anatomic and Functional Imaging of Neuroendocrine Tumors. Current treatment options in oncology. 2020;21(9):75.
  68. Khanna L, Prasad SR, Sunnapwar A, Kondapaneni S, Dasyam A, Tammisetti VS, et al. Pancreatic Neuroendocrine Neoplasms: 2020 Update on Pathologic and Imaging Findings and Classification. Radiographics : a review publication of the Radiological Society of North America, Inc. 2020;40(5):1240-62.
  69. Rozenblum L, Mokrane FZ, Yeh R, Sinigaglia M, Besson FL, Seban RD, et al. Imaging-guided precision medicine in non-resectable gastro-entero-pancreatic neuroendocrine tumors: A step-by-step approach. European journal of radiology. 2020;122:108743.
  70. Galgano SJ, Sharbidre K, Morgan DE. Multimodality Imaging of Neuroendocrine Tumors. Radiologic clinics of North America. 2020;58(6):1147-59.
  71. Tamm EP, Bhosale P, Lee JH, Rohren EM. State-of-the-art Imaging of Pancreatic Neuroendocrine Tumors. Surgical oncology clinics of North America. 2016;25(2):375-400.
  72. Sundin A, Vullierme MP, Kaltsas G, Plöckinger U. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: radiological examinations. Neuroendocrinology. 2009;90(2):167-83.
  73. Jackson JE. Angiography and arterial stimulation venous sampling in the localization of pancreatic neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):229-39.
  74. McLean AM, Fairclough PD. Endoscopic ultrasound in the localisation of pancreatic islet cell tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):177-93.
  75. Norton JA, Jensen RT. Resolved and unresolved controversies in the surgical management of patients with Zollinger-Ellison syndrome. Annals of surgery. 2004;240(5):757-73.
  76. Krenning EP, Kwekkeboom DJ, Oei HY, de Jong RJ, Dop FJ, Reubi JC, et al. Somatostatin-receptor scintigraphy in gastroenteropancreatic tumors. An overview of European results. Annals of the New York Academy of Sciences. 1994;733:416-24.
  77. Desai H, Borges-Neto S, Wong TZ. Molecular Imaging and Therapy for Neuroendocrine Tumors. Current treatment options in oncology. 2019;20(10):78.
  78. Termanini B, Gibril F, Reynolds JC, Doppman JL, Chen CC, Stewart CA, et al. Value of somatostatin receptor scintigraphy: a prospective study in gastrinoma of its effect on clinical management. Gastroenterology. 1997;112(2):335-47.
  79. Gibril F, Reynolds JC, Chen CC, Yu F, Goebel SU, Serrano J, et al. Specificity of somatostatin receptor scintigraphy: a prospective study and effects of false-positive localizations on management in patients with gastrinomas. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 1999;40(4):539-53.
  80. Christ E, Wild D, Ederer S, Béhé M, Nicolas G, Caplin ME, et al. Glucagon-like peptide-1 receptor imaging for the localisation of insulinomas: a prospective multicentre imaging study. The lancet Diabetes & endocrinology. 2013;1(2):115-22.
  81. Panagiotidis E, Alshammari A, Michopoulou S, Skoura E, Naik K, Maragkoudakis E, et al. Comparison of the Impact of 68Ga-DOTATATE and 18F-FDG PET/CT on Clinical Management in Patients with Neuroendocrine Tumors. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2017;58(1):91-6.
  82. Maxwell JE, Howe JR. Imaging in neuroendocrine tumors: an update for the clinician. International journal of endocrine oncology. 2015;2(2):159-68.
  83. Johnbeck CB, Knigge U, Kjær A. PET tracers for somatostatin receptor imaging of neuroendocrine tumors: current status and review of the literature. Future oncology (London, England). 2014;10(14):2259-77.
  84. Kornaczewski Jackson ER, Pointon OP, Bohmer R, Burgess JR. Utility of FDG-PET Imaging for Risk Stratification of Pancreatic Neuroendocrine Tumors in MEN1. The Journal of clinical endocrinology and metabolism. 2017;102(6):1926-33.
  85. Orlefors H, Sundin A, Garske U, Juhlin C, Oberg K, Skogseid B, et al. Whole-body (11)C-5-hydroxytryptophan positron emission tomography as a universal imaging technique for neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and computed tomography. The Journal of clinical endocrinology and metabolism. 2005;90(6):3392-400.
  86. Gabriel M, Decristoforo C, Kendler D, Dobrozemsky G, Heute D, Uprimny C, et al. 68Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and CT. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2007;48(4):508-18.
  87. Eriksson B, Orlefors H, Oberg K, Sundin A, Bergström M, Långström B. Developments in PET for the detection of endocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):311-24.
  88. Geijer H, Breimer LH. Somatostatin receptor PET/CT in neuroendocrine tumours: update on systematic review and meta-analysis. European journal of nuclear medicine and molecular imaging. 2013;40(11):1770-80.
  89. Deppen SA, Liu E, Blume JD, Clanton J, Shi C, Jones-Jackson LB, et al. Safety and Efficacy of 68Ga-DOTATATE PET/CT for Diagnosis, Staging, and Treatment Management of Neuroendocrine Tumors. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(5):708-14.
  90. Sadowski SM, Neychev V, Millo C, Shih J, Nilubol N, Herscovitch P, et al. Prospective Study of 68Ga-DOTATATE Positron Emission Tomography/Computed Tomography for Detecting Gastro-Entero-Pancreatic Neuroendocrine Tumors and Unknown Primary Sites. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(6):588-96.
  91. Etchebehere EC, de Oliveira Santos A, Gumz B, Vicente A, Hoff PG, Corradi G, et al. 68Ga-DOTATATE PET/CT, 99mTc-HYNIC-octreotide SPECT/CT, and whole-body MR imaging in detection of neuroendocrine tumors: a prospective trial. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2014;55(10):1598-604.
  92. Skoura E, Michopoulou S, Mohmaduvesh M, Panagiotidis E, Al Harbi M, Toumpanakis C, et al. The Impact of 68Ga-DOTATATE PET/CT Imaging on Management of Patients with Neuroendocrine Tumors: Experience from a National Referral Center in the United Kingdom. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(1):34-40.
  93. Crown A, Rocha FG, Raghu P, Lin B, Funk G, Alseidi A, et al. Impact of initial imaging with gallium-68 dotatate PET/CT on diagnosis and management of patients with neuroendocrine tumors. Journal of surgical oncology. 2020;121(3):480-5.
  94. Hofman MS, Lau WF, Hicks RJ. Somatostatin receptor imaging with 68Ga DOTATATE PET/CT: clinical utility, normal patterns, pearls, and pitfalls in interpretation. Radiographics : a review publication of the Radiological Society of North America, Inc. 2015;35(2):500-16.
  95. Lastoria S, Marciello F, Faggiano A, Aloj L, Caracò C, Aurilio M, et al. Role of (68)Ga-DOTATATE PET/CT in patients with multiple endocrine neoplasia type 1 (MEN1). Endocrine. 2016;52(3):488-94.
  96. Luo Y, Pan Q, Yao S, Yu M, Wu W, Xue H, et al. Glucagon-Like Peptide-1 Receptor PET/CT with 68Ga-NOTA-Exendin-4 for Detecting Localized Insulinoma: A Prospective Cohort Study. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(5):715-20.
  97. Elazezy M, Joosse SA. Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management. Computational and structural biotechnology journal. 2018;16:370-8.
  98. Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nature reviews Clinical oncology. 2017;14(9):531-48.
  99. De Rubis G, Rajeev Krishnan S, Bebawy M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends in pharmacological sciences. 2019;40(3):172-86.
  100. Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nature reviews Genetics. 2019;20(2):71-88.
  101. Lissa D, Robles AI. Methylation analyses in liquid biopsy. Translational lung cancer research. 2016;5(5):492-504.
  102. Bocchini M, Nicolini F, Severi S, Bongiovanni A, Ibrahim T, Simonetti G, et al. Biomarkers for Pancreatic Neuroendocrine Neoplasms (PanNENs) Management-An Updated Review. Frontiers in oncology. 2020;10:831.
  103. Öberg K, Califano A, Strosberg JR, Ma S, Pape U, Bodei L, et al. A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Annals of oncology : official journal of the European Society for Medical Oncology. 2020;31(2):202-12.
  104. Oberg K, Modlin IM, De Herder W, Pavel M, Klimstra D, Frilling A, et al. Consensus on biomarkers for neuroendocrine tumour disease. The Lancet Oncology. 2015;16(9):e435-e46.
  105. Parbhu SK, Adler DG. Pancreatic neuroendocrine tumors: contemporary diagnosis and management. Hospital practice (1995). 2016;44(3):109-19.
  106. de Herder WW. Biochemistry of neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):33-41.
  107. Ardill JE. Circulating markers for endocrine tumours of the gastroenteropancreatic tract. Annals of clinical biochemistry. 2008;45(Pt 6):539-59.
  108. Oberg K. Biochemical diagnosis of neuroendocrine GEP tumor. The Yale journal of biology and medicine. 1997;70(5-6):501-8.
  109. Panzuto F, Severi C, Cannizzaro R, Falconi M, Angeletti S, Pasquali A, et al. Utility of combined use of plasma levels of chromogranin A and pancreatic polypeptide in the diagnosis of gastrointestinal and pancreatic endocrine tumors. Journal of endocrinological investigation. 2004;27(1):6-11.
  110. Bajetta E, Ferrari L, Martinetti A, Celio L, Procopio G, Artale S, et al. Chromogranin A, neuron specific enolase, carcinoembryonic antigen, and hydroxyindole acetic acid evaluation in patients with neuroendocrine tumors. Cancer. 1999;86(5):858-65.
  111. Stabile BE, Howard TJ, Passaro E, Jr., O'Connor DT. Source of plasma chromogranin A elevation in gastrinoma patients. Archives of surgery (Chicago, Ill : 1960). 1990;125(4):451-3.
  112. Qiao XW, Qiu L, Chen YJ, Meng CT, Sun Z, Bai CM, et al. Chromogranin A is a reliable serum diagnostic biomarker for pancreatic neuroendocrine tumors but not for insulinomas. BMC endocrine disorders. 2014;14:64.
  113. Nobels FR, Kwekkeboom DJ, Coopmans W, Schoenmakers CH, Lindemans J, De Herder WW, et al. Chromogranin A as serum marker for neuroendocrine neoplasia: comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. The Journal of clinical endocrinology and metabolism. 1997;82(8):2622-8.
  114. Bílek R, Šafařík L, Ciprová V, Vlcek P, Lisá L. Chromogranin A, a member of neuroendocrine secretory proteins as a selective marker for laboratory diagnosis of pheochromocytoma. Physiological research. 2008;57 Suppl 1:S171-s9.
  115. Marotta V, Zatelli MC, Sciammarella C, Ambrosio MR, Bondanelli M, Colao A, et al. Chromogranin A as circulating marker for diagnosis and management of neuroendocrine neoplasms: more flaws than fame. Endocrine-related cancer. 2018;25(1):R11-r29.
  116. Stridsberg M, Eriksson B, Fellström B, Kristiansson G, Tiensuu Janson E. Measurements of chromogranin B can serve as a complement to chromogranin A. Regulatory peptides. 2007;139(1-3):80-3.
  117. Takiyyuddin MA, Cervenka JH, Hsiao RJ, Barbosa JA, Parmer RJ, O'Connor DT. Chromogranin A. Storage and release in hypertension. Hypertension (Dallas, Tex : 1979). 1990;15(3):237-46.
  118. Lindholm DP, Oberg K. Biomarkers and molecular imaging in gastroenteropancreatic neuroendocrine tumors. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. 2011;43(12):832-7.
  119. Jilesen AP, Busch OR, van Gulik TM, Gouma DJ, Nieveen van Dijkum EJ. Standard pre- and postoperative determination of chromogranin a in resectable non-functioning pancreatic neuroendocrine tumors--diagnostic accuracy: NF-pNET and low tumor burden. Digestive surgery. 2014;31(6):407-14.
  120. Monaghan PJ, Lamarca A, Valle JW, Hubner RA, Mansoor W, Trainer PJ, et al. Routine measurement of plasma chromogranin B has limited clinical utility in the management of patients with neuroendocrine tumours. Clinical endocrinology. 2016;84(3):348-52.
  121. Hofland J, Zandee WT, de Herder WW. Role of biomarker tests for diagnosis of neuroendocrine tumours. Nature reviews Endocrinology. 2018;14(11):656-69.
  122. van Adrichem RC, Kamp K, Vandamme T, Peeters M, Feelders RA, de Herder WW. Serum neuron-specific enolase level is an independent predictor of overall survival in patients with gastroenteropancreatic neuroendocrine tumors. Annals of oncology : official journal of the European Society for Medical Oncology. 2016;27(4):746-7.
  123. Stronge RL, Turner GB, Johnston BT, McCance DR, McGinty A, Patterson CC, et al. A rapid rise in circulating pancreastatin in response to somatostatin analogue therapy is associated with poor survival in patients with neuroendocrine tumours. Annals of clinical biochemistry. 2008;45(Pt 6):560-6.
  124. Walter T, Chardon L, Chopin-laly X, Raverot V, Caffin AG, Chayvialle JA, et al. Is the combination of chromogranin A and pancreatic polypeptide serum determinations of interest in the diagnosis and follow-up of gastro-entero-pancreatic neuroendocrine tumours? European journal of cancer (Oxford, England : 1990). 2012;48(12):1766-73.
  125. Brown JE, Cook RJ, Major P, Lipton A, Saad F, Smith M, et al. Bone turnover markers as predictors of skeletal complications in prostate cancer, lung cancer, and other solid tumors. Journal of the National Cancer Institute. 2005;97(1):59-69.
  126. Lipton A, Costa L, Ali S, Demers L. Use of markers of bone turnover for monitoring bone metastases and the response to therapy. Seminars in oncology. 2001;28(4 Suppl 11):54-9.
  127. Khan MS, Kirkwood A, Tsigani T, Garcia-Hernandez J, Hartley JA, Caplin ME, et al. Circulating tumor cells as prognostic markers in neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31(3):365-72.
  128. Mandair D, Khan MS, Lopes A, Furtado O'Mahony L, Ensell L, Lowe H, et al. Prognostic Threshold for Circulating Tumor Cells in Patients With Pancreatic and Midgut Neuroendocrine Tumors. The Journal of clinical endocrinology and metabolism. 2021;106(3):872-82.
  129. Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV, et al. Circulating tumor cells and EpCAM expression in neuroendocrine tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17(2):337-45.
  130. Rizzo FM, Vesely C, Childs A, Marafioti T, Khan MS, Mandair D, et al. Circulating tumour cells and their association with bone metastases in patients with neuroendocrine tumours. British journal of cancer. 2019;120(3):294-300.
  131. Grande E, Capdevila J, Castellano D, Teulé A, Durán I, Fuster J, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Annals of oncology : official journal of the European Society for Medical Oncology. 2015;26(9):1987-93.
  132. Meyer T, Caplin M, Khan MS, Toumpanakis C, Shetty S, Ramage JK, et al. Circulating tumor cells and tumor biomarkers in functional midgut neuroendocrine tumors. Journal of Neuroendocrinology. 2022.
  133. Childs A, Vesely C, Ensell L, Lowe H, Luong TV, Caplin ME, et al. Expression of somatostatin receptors 2 and 5 in circulating tumour cells from patients with neuroendocrine tumours. British journal of cancer. 2016;115(12):1540-7.
  134. Rizzo FM, Meyer T. Liquid Biopsies for Neuroendocrine Tumors: Circulating Tumor Cells, DNA, and MicroRNAs. Endocrinology and metabolism clinics of North America. 2018;47(3):471-83.
  135. Mader S, Pantel K. Liquid Biopsy: Current Status and Future Perspectives. Oncology research and treatment. 2017;40(7-8):404-8.
  136. Boons G, Vandamme T, Peeters M, Beyens M, Driessen A, Janssens K, et al. Cell-Free DNA From Metastatic Pancreatic Neuroendocrine Tumor Patients Contains Tumor-Specific Mutations and Copy Number Variations. Frontiers in oncology. 2018;8:467.
  137. Malczewska A, Kidd M, Matar S, Kos-Kudla B, Modlin IM. A Comprehensive Assessment of the Role of miRNAs as Biomarkers in Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology. 2018;107(1):73-90.
  138. Li SC, Khan M, Caplin M, Meyer T, Öberg K, Giandomenico V. Somatostatin Analogs Treated Small Intestinal Neuroendocrine Tumor Patients Circulating MicroRNAs. PloS one. 2015;10(5):e0125553.
  139. Kövesdi A, Kurucz PA, Nyírő G, Darvasi O, Patócs A, Butz H. Circulating miRNA Increases the Diagnostic Accuracy of Chromogranin A in Metastatic Pancreatic Neuroendocrine Tumors. Cancers. 2020;12(9).
  140. Malczewska A, Kos-Kudła B, Kidd M, Drozdov I, Bodei L, Matar S, et al. The clinical applications of a multigene liquid biopsy (NETest) in neuroendocrine tumors. Advances in medical sciences. 2020;65(1):18-29.
  141. Modlin IM, Drozdov I, Kidd M. The identification of gut neuroendocrine tumor disease by multiple synchronous transcript analysis in blood. PloS one. 2013;8(5):e63364.
  142. Modlin IM, Kidd M, Falconi M, Filosso PL, Frilling A, Malczewska A, et al. A multigenomic liquid biopsy biomarker for neuroendocrine tumor disease outperforms CgA and has surgical and clinical utility. Annals of oncology : official journal of the European Society for Medical Oncology. 2021;32(11):1425-33.
  143. Modlin IM, Kidd M, Malczewska A, Drozdov I, Bodei L, Matar S, et al. The NETest: The Clinical Utility of Multigene Blood Analysis in the Diagnosis and Management of Neuroendocrine Tumors. Endocrinology and metabolism clinics of North America. 2018;47(3):485-504.
  144. Modlin IM, Frilling A, Salem RR, Alaimo D, Drymousis P, Wasan HS, et al. Blood measurement of neuroendocrine gene transcripts defines the effectiveness of operative resection and ablation strategies. Surgery. 2016;159(1):336-47.
  145. Ćwikła JB, Bodei L, Kolasinska-Ćwikła A, Sankowski A, Modlin IM, Kidd M. Circulating Transcript Analysis (NETest) in GEP-NETs Treated With Somatostatin Analogs Defines Therapy. The Journal of clinical endocrinology and metabolism. 2015;100(11):E1437-45.
  146. Bodei L, Kidd M, Modlin IM, Severi S, Drozdov I, Nicolini S, et al. Measurement of circulating transcripts and gene cluster analysis predicts and defines therapeutic efficacy of peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors. European journal of nuclear medicine and molecular imaging. 2016;43(5):839-51.
  147. van Treijen MJC, Korse CM, van Leeuwaarde RS, Saveur LJ, Vriens MR, Verbeek WHM, et al. Blood Transcript Profiling for the Detection of Neuroendocrine Tumors: Results of a Large Independent Validation Study. Frontiers in endocrinology. 2018;9:740.
  148. Lee L, Ito T, Jensen RT. Imaging of pancreatic neuroendocrine tumors: recent advances, current status, and controversies. Expert review of anticancer therapy. 2018;18(9):837-60.
  149. Akirov A, Larouche V, Alshehri S, Asa SL, Ezzat S. Treatment Options for Pancreatic Neuroendocrine Tumors. Cancers. 2019;11(6).
  150. Belotto M, Crouzillard B, Araujo KO, Peixoto RD. PANCREATIC NEUROENDOCRINE TUMORS: SURGICAL RESECTION. Arquivos brasileiros de cirurgia digestiva : ABCD = Brazilian archives of digestive surgery. 2019;32(1):e1428.
  151. Zerbi A, Capitanio V, Boninsegna L, Pasquali C, Rindi G, Delle Fave G, et al. Surgical treatment of pancreatic endocrine tumours in Italy: results of a prospective multicentre study of 262 cases. Langenbeck's archives of surgery. 2011;396(3):313-21.
  152. Hill JS, McPhee JT, McDade TP, Zhou Z, Sullivan ME, Whalen GF, et al. Pancreatic neuroendocrine tumors: the impact of surgical resection on survival. Cancer. 2009;115(4):741-51.
  153. Gomez-Rivera F, Stewart AE, Arnoletti JP, Vickers S, Bland KI, Heslin MJ. Surgical treatment of pancreatic endocrine neoplasms. American journal of surgery. 2007;193(4):460-5.
  154. Martin RC, Kooby DA, Weber SM, Merchant NB, Parikh AA, Cho CS, et al. Analysis of 6,747 pancreatic neuroendocrine tumors for a proposed staging system. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2011;15(1):175-83.
  155. Scarpa A, Mantovani W, Capelli P, Beghelli S, Boninsegna L, Bettini R, et al. Pancreatic endocrine tumors: improved TNM staging and histopathological grading permit a clinically efficient prognostic stratification of patients. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2010;23(6):824-33.
  156. Toste PA, Kadera BE, Tatishchev SF, Dawson DW, Clerkin BM, Muthusamy R, et al. Nonfunctional pancreatic neuroendocrine tumors <2 cm on preoperative imaging are associated with a low incidence of nodal metastasis and an excellent overall survival. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2013;17(12):2105-13.
  157. Partelli S, Gaujoux S, Boninsegna L, Cherif R, Crippa S, Couvelard A, et al. Pattern and clinical predictors of lymph node involvement in nonfunctioning pancreatic neuroendocrine tumors (NF-PanNETs). JAMA surgery. 2013;148(10):932-9.
  158. Titan AL, Norton JA, Fisher AT, Foster DS, Harris EJ, Worhunsky DJ, et al. Evaluation of Outcomes Following Surgery for Locally Advanced Pancreatic Neuroendocrine Tumors. JAMA network open. 2020;3(11):e2024318.
  159. Bettini R, Partelli S, Boninsegna L, Capelli P, Crippa S, Pederzoli P, et al. Tumor size correlates with malignancy in nonfunctioning pancreatic endocrine tumor. Surgery. 2011;150(1):75-82.
  160. Gaujoux S, Partelli S, Maire F, D'Onofrio M, Larroque B, Tamburrino D, et al. Observational study of natural history of small sporadic nonfunctioning pancreatic neuroendocrine tumors. The Journal of clinical endocrinology and metabolism. 2013;98(12):4784-9.
  161. Triponez F, Sadowski SM, Pattou F, Cardot-Bauters C, Mirallié E, Le Bras M, et al. Long-term Follow-up of MEN1 Patients Who Do Not Have Initial Surgery for Small ≤2 cm Nonfunctioning Pancreatic Neuroendocrine Tumors, an AFCE and GTE Study: Association Francophone de Chirurgie Endocrinienne & Groupe d'Etude des Tumeurs Endocrines. Annals of surgery. 2018;268(1):158-64.
  162. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, et al. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). The Journal of clinical endocrinology and metabolism. 2012;97(9):2990-3011.
  163. Krug S, Gress TM, Michl P, Rinke A. The Role of Cytotoxic Chemotherapy in Advanced Pancreatic Neuroendocrine Tumors. Digestion. 2017;96(2):67-75.
  164. Akerström G, Hellman P. Surgery on neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):87-109.
  165. Jensen RT, Norton JA. Treatment of Pancreatic Neuroendocrine Tumors in Multiple Endocrine Neoplasia Type 1: Some Clarity But Continued Controversy. Pancreas. 2017;46(5):589-94.
  166. Jensen RT, Niederle B, Mitry E, Ramage JK, Steinmuller T, Lewington V, et al. Gastrinoma (duodenal and pancreatic). Neuroendocrinology. 2006;84(3):173-82.
  167. Norton JA, Fraker DL, Alexander HR, Venzon DJ, Doppman JL, Serrano J, et al. Surgery to cure the Zollinger-Ellison syndrome. The New England journal of medicine. 1999;341(9):635-44.
  168. Al-Kurd A, Chapchay K, Grozinsky-Glasberg S, Mazeh H. Laparoscopic resection of pancreatic neuroendocrine tumors. World journal of gastroenterology. 2014;20(17):4908-16.
  169. Drymousis P, Raptis DA, Spalding D, Fernandez-Cruz L, Menon D, Breitenstein S, et al. Laparoscopic versus open pancreas resection for pancreatic neuroendocrine tumours: a systematic review and meta-analysis. HPB : the official journal of the International Hepato Pancreato Biliary Association. 2014;16(5):397-406.
  170. Venkat R, Edil BH, Schulick RD, Lidor AO, Makary MA, Wolfgang CL. Laparoscopic distal pancreatectomy is associated with significantly less overall morbidity compared to the open technique: a systematic review and meta-analysis. Annals of surgery. 2012;255(6):1048-59.
  171. Kendrick ML, van Hilst J, Boggi U, de Rooij T, Walsh RM, Zeh HJ, et al. Minimally invasive pancreatoduodenectomy. HPB : the official journal of the International Hepato Pancreato Biliary Association. 2017;19(3):215-24.
  172. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97(4):934-59.
  173. Chamberlain RS, Canes D, Brown KT, Saltz L, Jarnagin W, Fong Y, et al. Hepatic neuroendocrine metastases: does intervention alter outcomes? Journal of the American College of Surgeons. 2000;190(4):432-45.
  174. Hodul PJ, Strosberg JR, Kvols LK. Aggressive surgical resection in the management of pancreatic neuroendocrine tumors: when is it indicated? Cancer control : journal of the Moffitt Cancer Center. 2008;15(4):314-21.
  175. Zhou B, Zhan C, Ding Y, Yan S, Zheng S. Role of palliative resection of the primary pancreatic neuroendocrine tumor in patients with unresectable metastatic liver disease: a systematic review and meta-analysis. OncoTargets and therapy. 2018;11:975-82.
  176. Mazzaferro V, Pulvirenti A, Coppa J. Neuroendocrine tumors metastatic to the liver: how to select patients for liver transplantation? Journal of hepatology. 2007;47(4):460-6.
  177. Shah MH, Goldner WS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, et al. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018. Journal of the National Comprehensive Cancer Network : JNCCN. 2018;16(6):693-702.
  178. Solorzano CC, Lee JE, Pisters PW, Vauthey JN, Ayers GD, Jean ME, et al. Nonfunctioning islet cell carcinoma of the pancreas: survival results in a contemporary series of 163 patients. Surgery. 2001;130(6):1078-85.
  179. Elias D, Lefevre JH, Duvillard P, Goéré D, Dromain C, Dumont F, et al. Hepatic metastases from neuroendocrine tumors with a "thin slice" pathological examination: they are many more than you think. Annals of surgery. 2010;251(2):307-10.
  180. Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM, Que FG. Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. Journal of the American College of Surgeons. 2003;197(1):29-37.
  181. Norton JA, Sugarbaker PH, Doppman JL, Wesley RA, Maton PN, Gardner JD, et al. Aggressive resection of metastatic disease in selected patients with malignant gastrinoma. Annals of surgery. 1986;203(4):352-9.
  182. Sarmiento JM, Que FG. Hepatic surgery for metastases from neuroendocrine tumors. Surgical oncology clinics of North America. 2003;12(1):231-42.
  183. House MG, Cameron JL, Lillemoe KD, Schulick RD, Choti MA, Hansel DE, et al. Differences in survival for patients with resectable versus unresectable metastases from pancreatic islet cell cancer. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2006;10(1):138-45.
  184. Gupta S, Johnson MM, Murthy R, Ahrar K, Wallace MJ, Madoff DC, et al. Hepatic arterial embolization and chemoembolization for the treatment of patients with metastatic neuroendocrine tumors: variables affecting response rates and survival. Cancer. 2005;104(8):1590-602.
  185. Mazzaferro V, Sposito C, Coppa J, Miceli R, Bhoori S, Bongini M, et al. The Long-Term Benefit of Liver Transplantation for Hepatic Metastases From Neuroendocrine Tumors. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2016;16(10):2892-902.
  186. Herrera-Martínez AD, Hofland J, Hofland LJ, Brabander T, Eskens F, Gálvez Moreno MA, et al. Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives. Drugs. 2019;79(1):21-42.
  187. Gomes-Porras M, Cárdenas-Salas J, Álvarez-Escolá C. Somatostatin Analogs in Clinical Practice: a Review. International journal of molecular sciences. 2020;21(5).
  188. Hauser H, Gerson DS, Reidy-Lagunes D, Raj N. Systemic Therapies for Metastatic Pancreatic Neuroendocrine Tumors. Current treatment options in oncology. 2019;20(12):87.
  189. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. The New England journal of medicine. 2014;371(3):224-33.
  190. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocrine-related cancer. 2016;23(3):191-9.
  191. Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of oncology : official journal of the European Society for Medical Oncology. 2020;31(7):844-60.
  192. Vinik AI, Raymond E. Pancreatic neuroendocrine tumors: approach to treatment with focus on sunitinib. Therapeutic advances in gastroenterology. 2013;6(5):396-411.
  193. Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008;26(20):3403-10.
  194. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England journal of medicine. 2011;364(6):501-13.
  195. Faivre S, Niccoli P, Castellano D, Valle JW, Hammel P, Raoul JL, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Annals of oncology : official journal of the European Society for Medical Oncology. 2017;28(2):339-43.
  196. Vinik E, Carlton CA, Silva MP, Vinik AI. Development of the Norfolk quality of life tool for assessing patients with neuroendocrine tumors. Pancreas. 2009;38(3):e87-95.
  197. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(1):69-76.
  198. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. The New England journal of medicine. 2011;364(6):514-23.
  199. Lombard-Bohas C, Yao JC, Hobday T, Van Cutsem E, Wolin EM, Panneerselvam A, et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015;44(2):181-9.
  200. Chan JA, Blaszkowsky L, Stuart K, Zhu AX, Allen J, Wadlow R, et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer. 2013;119(17):3212-8.
  201. Kulke MH, Ruszniewski P, Van Cutsem E, Lombard-Bohas C, Valle JW, De Herder WW, et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Annals of oncology : official journal of the European Society for Medical Oncology. 2017;28(6):1309-15.
  202. Halfdanarson TR, Strosberg JR, Tang L, Bellizzi AM, Bergsland EK, O'Dorisio TM, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(7):863-81.
  203. Yao J, Phan AT. Optimising Therapeutic Options for Patients with Advanced Pancreatic Neuroendocrine Tumours. European Oncology & Haematology. 2012;8(4):217-23.
  204. Mateo J, Heymach JV, Zurita AJ. Circulating biomarkers of response to sunitinib in gastroenteropancreatic neuroendocrine tumors: current data and clinical outlook. Molecular diagnosis & therapy. 2012;16(3):151-61.
  205. Bello C, Deprimo SE, Friece C, Smeraglia J, Sherman L, Tye L, et al. Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. Journal of Clinical Oncology. 2006;24(18_suppl):4045-.
  206. Yao JC, Pavel M, Phan AT, Kulke MH, Hoosen S, St Peter J, et al. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. The Journal of clinical endocrinology and metabolism. 2011;96(12):3741-9.
  207. Palmieri LJ, Dermine S, Barré A, Dhooge M, Brezault C, Cottereau AS, et al. Medical Treatment of Advanced Pancreatic Neuroendocrine Neoplasms. Journal of clinical medicine. 2020;9(6).
  208. Pavel M, O'Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016;103(2):172-85.
  209. Okusaka T, Ueno H, Morizane C, Kondo S, Sakamoto Y, Takahashi H, et al. Cytotoxic chemotherapy for pancreatic neuroendocrine tumors. Journal of hepato-biliary-pancreatic sciences. 2015;22(8):628-33.
  210. Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. The New England journal of medicine. 1992;326(8):519-23.
  211. Kouvaraki MA, Ajani JA, Hoff P, Wolff R, Evans DB, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004;22(23):4762-71.
  212. Ramanathan RK, Cnaan A, Hahn RG, Carbone PP, Haller DG. Phase II trial of dacarbazine (DTIC) in advanced pancreatic islet cell carcinoma. Study of the Eastern Cooperative Oncology Group-E6282. Annals of oncology : official journal of the European Society for Medical Oncology. 2001;12(8):1139-43.
  213. de Mestier L, Walter T, Brixi H, Evrard C, Legoux JL, de Boissieu P, et al. Comparison of Temozolomide-Capecitabine to 5-Fluorouracile-Dacarbazine in 247 Patients with Advanced Digestive Neuroendocrine Tumors Using Propensity Score Analyses. Neuroendocrinology. 2019;108(4):343-53.
  214. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268-75.
  215. Chan JA, Stuart K, Earle CC, Clark JW, Bhargava P, Miksad R, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(24):2963-8.
  216. Koumarianou A, Antoniou S, Kanakis G, Economopoulos N, Rontogianni D, Ntavatzikos A, et al. Combination treatment with metronomic temozolomide, bevacizumab and long-acting octreotide for malignant neuroendocrine tumours. Endocrine-related cancer. 2012;19(1):L1-4.
  217. Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24(3):401-6.
  218. Kunz PL, Catalano PJ, Nimeiri H, Fisher GA, Longacre TA, Suarez CJ, et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: A trial of the ECOG-ACRIN Cancer Research Group (E2211). Journal of Clinical Oncology. 2018;36(15_suppl):4004-.
  219. de Mestier L, Walter T, Evrard C, de Boissieu P, Hentic O, Cros J, et al. Temozolomide Alone or Combined with Capecitabine for the Treatment of Advanced Pancreatic Neuroendocrine Tumor. Neuroendocrinology. 2020;110(1-2):83-91.
  220. Moertel CG, Kvols LK, O'Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991;68(2):227-32.
  221. Mitry E, Baudin E, Ducreux M, Sabourin JC, Rufié P, Aparicio T, et al. Treatment of poorly differentiated neuroendocrine tumours with etoposide and cisplatin. British journal of cancer. 1999;81(8):1351-5.
  222. Thomas KEH, Voros BA, Boudreaux JP, Thiagarajan R, Woltering EA, Ramirez RA. Current Treatment Options in Gastroenteropancreatic Neuroendocrine Carcinoma. The oncologist. 2019;24(8):1076-88.
  223. Ilett EE, Langer SW, Olsen IH, Federspiel B, Kjær A, Knigge U. Neuroendocrine Carcinomas of the Gastroenteropancreatic System: A Comprehensive Review. Diagnostics (Basel, Switzerland). 2015;5(2):119-76.
  224. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Annals of oncology : official journal of the European Society for Medical Oncology. 2013;24(1):152-60.
  225. Hentic O, Hammel P, Couvelard A, Rebours V, Zappa M, Palazzo M, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocrine-related cancer. 2012;19(6):751-7.
  226. Bajetta E, Catena L, Procopio G, De Dosso S, Bichisao E, Ferrari L, et al. Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumours? Cancer chemotherapy and pharmacology. 2007;59(5):637-42.
  227. Spada F, Antonuzzo L, Marconcini R, Radice D, Antonuzzo A, Ricci S, et al. Oxaliplatin-Based Chemotherapy in Advanced Neuroendocrine Tumors: Clinical Outcomes and Preliminary Correlation with Biological Factors. Neuroendocrinology. 2016;103(6):806-14.
  228. Zhu J, Strosberg JR, Dropkin E, Strickler JH. Treatment of High-Grade Metastatic Pancreatic Neuroendocrine Carcinoma with FOLFIRINOX. Journal of gastrointestinal cancer. 2015;46(2):166-9.
  229. Camus B, Cottereau AS, Palmieri LJ, Dermine S, Tenenbaum F, Brezault C, et al. Indications of Peptide Receptor Radionuclide Therapy (PRRT) in Gastroenteropancreatic and Pulmonary Neuroendocrine Tumors: An Updated Review. Journal of clinical medicine. 2021;10(6).
  230. Kwekkeboom DJ, Teunissen JJ, Bakker WH, Kooij PP, de Herder WW, Feelders RA, et al. Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23(12):2754-62.
  231. Vinik E, Silva MP, Vinik AI. Measuring the relationship of quality of life and health status, including tumor burden, symptoms, and biochemical measures in patients with neuroendocrine tumors. Endocrinology and metabolism clinics of North America. 2011;40(1):97-109, viii.
  232. Ezziddin S, Khalaf F, Vanezi M, Haslerud T, Mayer K, Al Zreiqat A, et al. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. European journal of nuclear medicine and molecular imaging. 2014;41(5):925-33.
  233. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. The New England journal of medicine. 2017;376(2):125-35.
  234. Food and Drug Administration. FDA approves lutetium Lu 177 dotatate for treatment of GEP-NETS. 2018.
  235. Bodei L, Mueller-Brand J, Baum RP, Pavel ME, Hörsch D, O'Dorisio MS, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. European journal of nuclear medicine and molecular imaging. 2013;40(5):800-16.
  236. Brabander T, van der Zwan WA, Teunissen JJM, Kam BLR, Feelders RA, de Herder WW, et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2017;23(16):4617-24.
  237. Satapathy S, Mittal BR. 177Lu-DOTATATE peptide receptor radionuclide therapy versus Everolimus in advanced pancreatic neuroendocrine tumors: a systematic review and meta-analysis. Nuclear medicine communications. 2019;40(12):1195-203.
  238. Pusceddu S, Prinzi N, Tafuto S, Ibrahim T, Filice A, Brizzi MP, et al. Association of Upfront Peptide Receptor Radionuclide Therapy With Progression-Free Survival Among Patients With Enteropancreatic Neuroendocrine Tumors. JAMA network open. 2022;5(2):e220290.
  239. Howe JR, Merchant NB, Conrad C, Keutgen XM, Hallet J, Drebin JA, et al. The North American Neuroendocrine Tumor Society Consensus Paper on the Surgical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(1):1-33.
  240. Bloomston M, Al-Saif O, Klemanski D, Pinzone JJ, Martin EW, Palmer B, et al. Hepatic artery chemoembolization in 122 patients with metastatic carcinoid tumor: lessons learned. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2007;11(3):264-71.
  241. Kennedy AS, Dezarn WA, McNeillie P, Coldwell D, Nutting C, Carter D, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. American journal of clinical oncology. 2008;31(3):271-9.
  242. Rhee TK, Lewandowski RJ, Liu DM, Mulcahy MF, Takahashi G, Hansen PD, et al. 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Annals of surgery. 2008;247(6):1029-35.
  243. Grozinsky-Glasberg S, Kaltsas G, Kaltsatou M, Lev-Cohain N, Klimov A, Vergadis V, et al. Hepatic intra-arterial therapies in metastatic neuroendocrine tumors: lessons from clinical practice. Endocrine. 2018;60(3):499-509.
  244. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, et al. Phase I Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients with Advanced Solid Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21(19):4286-93.
  245. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science (New York, NY). 2016;351(6280):1463-9.
  246. Mehnert JM, Bergsland E, O'Neil BH, Santoro A, Schellens JHM, Cohen RB, et al. Pembrolizumab for the treatment of programmed death-ligand 1-positive advanced carcinoid or pancreatic neuroendocrine tumors: Results from the KEYNOTE-028 study. Cancer. 2020;126(13):3021-30.
  247. Strosberg J, Mizuno N, Doi T, Grande E, Delord JP, Shapira-Frommer R, et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results From the Phase II KEYNOTE-158 Study. Clinical cancer research : an official journal of the American Association for Cancer Research. 2020;26(9):2124-30.
  248. Naing A, Papadopoulos KP, Autio KA, Ott PA, Patel MR, Wong DJ, et al. Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(29):3562-9.
  249. Yao JC, Guthrie KA, Moran C, Strosberg JR, Kulke MH, Chan JA, et al. Phase III Prospective Randomized Comparison Trial of Depot Octreotide Plus Interferon Alfa-2b Versus Depot Octreotide Plus Bevacizumab in Patients With Advanced Carcinoid Tumors: SWOG S0518. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(15):1695-703.
  250. Ducreux M, Dahan L, Smith D, O'Toole D, Lepère C, Dromain C, et al. Bevacizumab combined with 5-FU/streptozocin in patients with progressive metastatic well-differentiated pancreatic endocrine tumours (BETTER trial)--a phase II non-randomised trial. European journal of cancer (Oxford, England : 1990). 2014;50(18):3098-106.
  251. Xu J, Shen L, Zhou Z, Li J, Bai C, Chi Y, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology. 2020;21(11):1500-12.
  252. Xu J, Shen L, Bai C, Wang W, Li J, Yu X, et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology. 2020;21(11):1489-99.
  253. Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. Journal of the National Cancer Institute. 1993;85(5):365-76.
  254. Yadegarfar G, Friend L, Jones L, Plum LM, Ardill J, Taal B, et al. Validation of the EORTC QLQ-GINET21 questionnaire for assessing quality of life of patients with gastrointestinal neuroendocrine tumours. British journal of cancer. 2013;108(2):301-10.
  255. Jiménez-Fonseca P, Carmona-Bayonas A, Martín-Pérez E, Crespo G, Serrano R, Llanos M, et al. Health-related quality of life in well-differentiated metastatic gastroenteropancreatic neuroendocrine tumors. Cancer metastasis reviews. 2015;34(3):381-400.
  256. Meng Y, McCarthy G, Berthon A, Dinet J. Patient-reported health state utilities in metastatic gastroenteropancreatic neuroendocrine tumours - an analysis based on the CLARINET study. Health and quality of life outcomes. 2017;15(1):131.
  257. Marinova M, Mücke M, Mahlberg L, Essler M, Cuhls H, Radbruch L, et al. Improving quality of life in patients with pancreatic neuroendocrine tumor following peptide receptor radionuclide therapy assessed by EORTC QLQ-C30. European journal of nuclear medicine and molecular imaging. 2018;45(1):38-46.
  258. Martini C, Gamper EM, Wintner L, Nilica B, Sperner-Unterweger B, Holzner B, et al. Systematic review reveals lack of quality in reporting health-related quality of life in patients with gastroenteropancreatic neuroendocrine tumours. Health and quality of life outcomes. 2016;14(1):127.

Bacterial Infections In Diabetes

ABSTRACT

Bacteria are microscopic single-celled organisms that exist in millions inside and outside the human body. Some bacteria are harmful and can cause a multitude of diseases in human beings. Diabetes mellitus, being a global pandemic, serves as an important cause of susceptibility to bacterial infections. Uncontrolled hyperglycemia is associated with impaired innate and adaptive immune responses that predispose to bacterial infections. In addition, chronic complications of diabetes like neuropathy (sensorimotor and autonomic) and peripheral vascular disease can lead to skin ulcerations with secondary bacterial infections. Diabetes also increases the incidence of infection related mortality. The relationship of diabetes and bacterial infections can be reciprocal, with certain infections like periodontitis exacerbating insulin resistance. Abnormalities in the bacterial flora of the gastrointestinal tract can play a role in the development of diabetes. Bacteria can infect any organ in the human body, the most common sites of infection in diabetes being the urinary tract, respiratory tract, skin, and soft tissues. Certain bacterial infections are very specific for diabetes like emphysematous pyelonephritis, emphysematous cholecystitis, and malignant otitis externa. Different antibiotic regimens (empirical and culture-specific) have been recommended for different bacterial infections, depending upon the site and severity. Our chapter gives an overview of the various bacteria, important from the standpoint of diabetes. We have also discussed the epidemiology and pathogenesis of bacterial infections in diabetes. In addition, we have highlighted the spectrum of bacterial infections and their management in diabetes. Stringent glycemic control, vaccination, adequate foot care practices, source control are some of the preventive measures to avoid bacterial infections in diabetes. Adequate knowledge about the spectrum and management of bacterial infections is important to prevent morbidity and mortality in diabetes.

INTRODUCTION

Diabetes is on the rise worldwide, with a global prevalence in adults in 2019 being 9.3% of the world population. In total numbers, this reflects a population of 463 million people with diabetes worldwide in 2019, with a projection of an increase to 700 million adults by 2045. A further 1.1 million children and adolescents under the age of 20, live with type 1 diabetes (1). The association between diabetes and bacterial infections is well recognized clinically and further adds to the morbidity associated with diabetes and its complications (2).

 

Patients with diabetes have a two-fold higher risk of community-acquired bacterial infections such as pneumococcal, streptococcal, and enterobacterial infections as compared with patients without diabetes (3-5). Urinary tract infections are more frequent in patients with diabetes. Janifer et al reported a high prevalence of 42.8% in 1157 South Indian subjects with type 2 diabetes (6). In a large retrospective cohort study in England comparing 102,493 patients with diabetes mellitus vs. n = 203,518 matched control subjects, incidence rate ratios (IRR) for infection-related hospitalizations were 3.71 (95% CI, 3.27 to 4.21) in those with type 1 diabetes mellitus and 1.88 (95% CI, 1.83 to 1.92) in those with type 2 diabetes mellitus (7). Diabetes is also associated with an average twofold higher risk of infection related mortality compared with individuals without diabetes (8).

 

Increased incidence and severity of bacterial infections in diabetes has been linked to an impaired innate and adaptive immune responses within the hyperglycemic environment (9).

Apart from hyperglycemia, other chronic complications of diabetes may also predispose patients to infections. For example, neuropathy in combination with peripheral vascular disease in diabetes can lead to ulcerations in the skin and secondary infections (10).

There is a bidirectional relationship between diabetes and bacterial infections. While diabetes increases the susceptibility to bacterial infections and its complications, chronic infections such as periodontitis is associated with increased pro inflammatory cytokines which can exacerbate insulin resistance and worsen glycemic control (11). There is a recent growing evidence that abnormalities in the microbiota composition can have a major role in the development of diabetes (12).

 

Awareness regarding the complex inter relationships between diabetes and associated bacterial infections is important for prevention and prompt treatment. A wide spectrum of bacterial infections such as malignant otitis externa, emphysematous pyelonephritis, emphysematous cholecystitis tend to be more common in diabetics than in others, and other infections may be more severe in diabetics than in nondiabetics (13). Infections may also be the first manifestation of long-standing unrecognized diabetes (14). The following figure illustrates the classification of medically important bacteria (15).

Fig 1. Classification of medically important bacteria

 

EPIDEMIOLOGY OF COMMON BACTERIAL INFECTIONS IN DIABETES

Epidemiology of common bacterial infections in diabetes with associated pathogens is shown in Table 1.

Table 1. Epidemiology of Common Bacterial Infections in Diabetes with Associated Pathogens

 

Epidemiology

Pathogens

Ref

Bacterial meningitis

Relative Risk = 2.2 (95% CI, 1.9–2.6) in diabetes compared to patients without diabetes

S pneumonia

Listeria monocytogenes

16

Malignant otitis externa

Odds ratio of prior diabetes in Malignant otitis externa is 10.07 (95% CI, 8.15-12.44)

 

Pseudomonas aeruginosa

17,18

Periodontitis

Odds Ratio = 1.34 (95% CI, 1.07–1.74) for periodontitis in diabetes  compared to patients without diabetes

 

Staphylococcus species

Streptococcus species

Bacillus species

E. Coli

19,20

Community Acquired pneumonia

(CAP)

Relative risk = 1.64 (95% CI 1.55–1.73) for CAP in patients with diabetes

 

Streptococcus pneumoniae

Legionella

Haemophilus influenza

21,22

Hospital Acquired pneumonia

Incidence Rate Ratio = 1.21, (95% CI,1.03–1.42) for postoperative pneumonia in diabetes

 

Pseudomonas species

Staphylococcus aureus

 

23, 24

Infective endocarditis

Odds ratio =1.9 (95% confidence interval 1.8-2.1)

 

Streptococcus viridans

Staphylococus aureus

Enterococcus species

25,26

Emphysematous Cholecystitis (EC)

60% of patients with EC had diabetes

Clostridium perfringens

Escherichia coli

27,28

Pyogenic liver abscess

Relative Risk = 3.6 (95% CI 2.9-4.5) in diabetes

 

 

 

Klebsiella pneumoniae

29,30

Urinary tract Infections

In patients with type 1 DM, adjusted odds ratio = 1.96 (95 % CI, 1.49–2.58)

In patients  with type 2 diabetes, adjusted Odds ratio = 1.24 (95 % CI, 1.10–1.39)

 

Escherichia coli

 

Other Enterobacteriaceae such as Klebsiella spp., Proteus spp., Enterobacter spp., and Enterococci

 

31,32

Bacterial skin and mucous membrane infections

In patients with type 1 DM, adjusted odds ratio = 1.59 (95 % CI, 1.12–2.24)

In patients  with type 2 diabetes, adjusted Odds ratio = 1.33 (95 % CI, 1.15–1.54)

 

Folliculitis        Group A streptococcus

                         Staphylococcus Aureus

 

 

Furunculosis     Streptococcus pneumoniae

Cellulitis

 

 

31, 33

Osteomyelitis of foot

20% of diabetic foot infections were associated with osteomyelitis.

 

More often poly-microbial

 

Gram positive : Staphylococcus aureus, Staphylococcus epidermidis, Streptococci, Enterobacteriaceae

 

Gram Negative : Escherichia coliKlebsiella pneumonia,  ProteusPseudomonas aeruginosa

 

34,35

GLYCEMIC CONTROL AND RISK OF INFECTIONS

Poor glycemic control increases the risk of infections in diabetes. A recent study examined the association between glycemic control in 85,312 patients with diabetes mellitus aged 40–89 years and the incidence of infection (36).  Infection rates rose steadily with HbA1c, which was particularly evident among those with HbA1c >11% (36).

 INCREASED INCIDENCE OF INFECTIONS IN DIABETES: PATHOPHYSIOLOGY

Infections are an important concern in individuals with diabetes due to the immune system’s failure to fight off invading pathogens (37). Diabetes progression itself is associated with immune dysfunction; autoimmunity in T1DM and low-grade chronic inflammation in T2DM (38).

 

Numerous studies have investigated the diabetes-related mechanisms that impair the host’s defence against pathogens. These mechanisms include a complex interplay between the host’s innate immunity and adaptive immunity (39, 40, 41). As noted earlier, chronic complications of diabetes can also predispose to infections (10).

The proposed mechanisms for increased susceptibility to infections in diabetes are depicted in figure 2.

Fig 2. Complex interactions between immune dysregulation (both innate and adaptive) from glycemic status, organism specific factors and diabetic complications plays major role in development of diabetes related infections.

Innate immunity

Cellular innate immunity is affected in uncontrolled diabetes. The steps involved in pathogen elimination by polymorphonuclear (PMN) leucocytes are:

(a) PMN adhesion to vascular endothelium, initially via the cell surface adhesion molecule L-selectin and then integrins

(b) transmigration through the vessel wall down a chemotactic gradient

(c) phagocytosis and microbial killing (2).

Hyperglycemia induces an increase in intracellular calcium concentration thereby reducing adenosine triphosphate (ATP) levels, which in turn leads to reduced phagocytic ability of polymorphonuclear cells. Correction of hyperglycemia leads to a significant reduction in intracellular calcium levels, an increase in ATP content, and improved phagocytosis (42). The hyperglycemic environment also inhibits glucose-6-phosphate dehydrogenase (G6PD) with resultant increase in apoptosis of polymorphonuclear leukocytes, and reduced polymorphonuclear leukocyte transmigration through the endothelium. Superoxide production is reduced in parallel with increasing glycemic exposure and consequently results in decreased microbial killing (2). Hyperglycemia is associated with increased formation of advanced glycation end products (AGE). AGE albumin has been shown to bind to the receptor for AGE (RAGE) present on neutrophils. This binding inhibits transendothelial migration and Staphylococcus aureus induced production of reactive oxygen species (ROS), resulting in impaired bacterial killing (43). Hyperglycemia also adversely affects the humoral component of innate immunity. Deficiency of C4 complement as well as decreased complement activation has been demonstrated in diabetes. This results in decreased opsonisation and phagocytosis of microbes. (44,45). Increased duration of cytokine response, increased pro-inflammatory cytokine gene expression and impaired local cytokine production leads to a dysregulated cytokine response in uncontrolled diabetes further increasing susceptibility to severe infections (46, 47, 48). 

Adaptive Immunity

There are two broad classes of adaptive immunity responses—antibody responses and cell-mediated immune responses, which are carried out by B cells and T cells respectively. In antibody responses, B cells are activated to secrete immunoglobulins which bind to the invading microbial antigens and block their binding to receptors on host cells. Antibody binding also marks invading pathogens for destruction by the phagocytes (49). Decreased levels of circulating immunoglobulins (IgG antibodies) as well as increased non enzymatic glycation of IgG antibodies leading to quantitative and qualitative defects in the humoral responses have been demonstrated in uncontrolled diabetes (50,51).

 

In cell-mediated immune responses, the second class of adaptive immune response, T cells which are activated by certain cytokines and antigen presenting cells, react directly against a foreign antigen that is presented to them on the surface of a host cell or themselves secrete cytokines that activate macrophages to destroy the invading microbes after phagocytosis (47). Dysregulation between anti-inflammatory and proinflammatory cytokines and defects at the level of antigen presenting cells in uncontrolled diabetes leads to dysfunction of T cells (52, 53). The role of immune systems and pathogenesis of bacterial infections is depicted in figure 3.

Fig 3. Pathogenesis of bacterial infections in diabetes. Describes role of various components of innate and adaptive immunity in pathogenesis of bacterial infection in diabetes; G6PD-Glucose 6 phosphate dehydrogenase; PMN-Polymorphonuclear cells; NADPH – Nicotinamide adenine dinucleotide phosphate; ROS- Reactive oxygen species; ATP -Adenosine triphosphate; AGEs- Advanced glycation end products; RAGE-Receptor for advanced glycation end products. 

Chronic Complications of Diabetes Predisposing to Infections

Over 50% of men and women with diabetes have bladder dysfunction which may impair voiding and increase the risk for urinary tract infections (54). The presence of renal disease and urinary incontinence in women are also predisposing factors for urinary tract infections. Diabetic cystopathy secondary to autonomic nervous dysfunction in long standing diabetes is characterized by a loss of sensation of bladder distension leading to decreased frequency of voiding and increased post-void residual urine volume. The possibility that voiding disorders may contribute to UTI should be considered in all diabetic patients (55).  

 

Peripheral diabetic neuropathy contributes to motor, autonomic, and sensory components of neuropathic foot ulcers. Damage to motor neurons of the foot musculature may lead to an imbalance of flexors and extensors, anatomic deformities, and eventual skin ulcerations. Damage to autonomic nerves impairs sweat gland function in the foot leading to a decreased ability to moisturize skin, resulting in epidermal cracks and skin breakdown. Lastly, the affected sensory component results in a loss of sensation of foot and reduced awareness of minor injuries (56). With ischemia, often as a result of related peripheral arterial disease, neuropathy can result in impaired barrier defences, skin ulcers with poor healing, and an increased risk of secondary infections and gangrene (57).

 

Pulmonary autonomic neuropathy in diabetes reduces mucociliary clearance and predisposes the lung to infections. Furthermore, hyperglycemia and insulin resistance impair collective surfactant D-mediated host defences of the lung in diabetes. Loose junctions between airway epithelial cells, which increase the transepithelial glucose gradient along with an increase in the glucose concentration of the airway surface liquid due to hyperglycemia, may dampen the airway defence against infection, resulting in lung bacterial overgrowth in diabetes (58).

SPECTRUM OF BACTERIAL INFECTIONS

Head and Neck Infections

BACTERIAL MENINGITIS

The majority of bacterial meningitis cases in adults is caused by Streptococcus pneumoniae. Listeria monocytogenes meningitis is more often found in elderly patients (>60 years) and those with acquired immune-deficiencies, such as diabetes. Immunodeficiency associated with diabetes is also a predisposing factor for pneumococcal and Haemophilus influenzae meningitis. Patients with bacterial meningitis and diabetes mellitus are older, have more comorbidities, frequently present with altered mental status and have higher mortality. In patients with diabetes, empirical antibiotics should include Cefotaxime/ ceftriaxone plus amoxicillin/ampicillin/ penicillin G (16, 59, 60).

MALIGNANT OTITIS EXTERNA

 Malignant otitis externa (MOE) is an invasive, potentially life-threatening infection of the external ear and skull base. MOE affects immunocompromised individuals and its presentation in an otherwise healthy individual should prompt an investigation for diabetes mellitus or other immune-deficiencies. In most cases, the causative agent of MOE is Pseudomonas aeruginosa. Typical patients with MOE are elderly individuals who have diabetes and severe, unremitting otalgia, aural fullness, otorrhea, and conductive hearing loss. Headache, temporomandibular joint pain, and decreased oral intake secondary to trismus may also be present. Findings of pain disproportionate to the examination, otorrhea, and granulation tissue along the floor of the ear canal at the bony–cartilaginous junction are usually the first nonspecific signs and symptoms of MOE. Important principles of treatment include aggressive control of diabetes and culture directed antibiotic therapy for at least 6-8 weeks. Although surgical intervention is no longer standard of care for MOE, it does require biopsy and culture, and may require local debridement of granulation tissue and bony sequestration or drainage of associated abscess. Long-term monotherapy with oral ciprofloxacin (750 mg twice daily) has been proposed as the preferred initial antibiotic regimen. However, microbial resistance to ciprofloxacin has been described and numerous studies have proposed carbapenem or third- generation cephalosporins as the initial empirical treatment. Recurrence rates of 15% to 20% have been reported for MOE (18, 61, 62). The risk factors for malignant otitis externa and its pathogenesis in diabetes are depicted in figure 4.

Fig 4. Risk factors for Malignant Otitis Externa and its pathogenesis in diabetes

PERIODONTITIS

Periodontitis is a complex chronic inflammatory condition in which inflammation in the periodontal tissues is stimulated by the long-term presence of the subgingival biofilm (figure 5). Periodontitis is a slowly progressing disease but the tissue destruction that occurs is largely irreversible. In the early stages, the condition is typically asymptomatic, is not usually painful, and many patients are unaware until the condition has progressed enough to result in tooth mobility. Advanced periodontitis is characterized by gingival erythema and edema, gingival bleeding, gingival recession, tooth mobility, suppuration from periodontal pockets, and tooth loss. In a randomized clinical trial, intensive periodontal treatment was associated with better glycemic control (A1C 8.3% vs 7.8% in control subjects and intensive treatment group respectively). Oral and periodontal health should be promoted as integral components of diabetes management (63, 64).

Fig 5. Chronic periodontitis with gingival inflammation in a patient with poorly controlled diabetes

DEEP NECK SPACE INFECTIONS/ABSCESS

Patients with diabetes are susceptible to spreading deep neck infections with a high frequency of complications, including tracheostomy and prolonged hospital stay. Odontogenic infections and upper airway infections are the leading reported causes of deep neck infections and the most common organism isolated is Klebsiella pneumoniae.  Early open surgical drainage remains the most appropriate method of treating deep neck abscesses. The choice of empirical antimicrobial agents in diabetic patients should take into account the agents effective against Klebsiella pneumoniae (65).

Respiratory Infections

COMMUNITY ACQUIRED PNEUMONIA

Patients with diabetes are at high risk of hospitalization due to community acquired pneumonia (CAP) (figure 6). Atypical clinical features like impaired consciousness and more severe pneumonia at admission are reported in patients with diabetes. Acute onset of disease, cough, purulent sputum, and pleuritic chest pain are less frequent among patients with diabetes. S. pneumonia, Legionella, and H influenza are frequent causative organisms of pneumonia in diabetes (22). Studies have also reported increased incidence of Klebsiella and pneumococcal pneumonia (3, 66). Independent risk factors for mortality in patients with diabetes and CAP are advanced age, bacteremia, septic shock at admission, and gram-negative pneumonia (22). The American Thoracic Society guidelines recommend combination therapy with amoxicillin/ clavulanic acid/ cephalosporin and macrolide/ doxycycline or monotherapy with respiratory fluoroquinolone for initial outpatient treatment in patients with diabetes. Beta lactam + macrolide or beta-lactam + fluoroquinolone is recommended in cases of severe in-patient pneumonia. Coverage for Pseudomonas aeruginosa is recommended in case of prior respiratory isolation, recent hospitalization with parenteral antibiotics treatment, and locally validated risk factors for Pseudomonas aeruginosa (67). The American Diabetes Association recommends vaccination against pneumococcal strains with one dose of PPSV23 (pneumococcal polysaccharide vaccine) between the ages of 19–64 years and another dose after 65 years of age. The PCV13 (pneumococcal conjugate vaccine) is no longer routinely recommended for patients over 65 years of age because of the declining rates of pneumonia due to these strains. All children are recommended to receive a four-dose series of PCV13 by 15 months of age. For children with diabetes who have incomplete series by ages 2–5 years, a catch-up schedule is recommended to ensure that these children have four doses. Children with diabetes between 6–18 years of age are also advised to receive one dose of PPSV23, preferably after receipt of PCV13 (68).

Fig 6. Radiographs of lower respiratory tract infection. A- Postero-anterior view radiograph of chest showing right middle lobe and left lower lobe consolidation in a patient with diabetes. B- Postero-anterior view radiograph of chest showing right lower lobe consolidation in a patient with diabetes

Cardiovascular Infections  

INFECTIVE ENDOCARDITIS

Infective endocarditis (IE) in diabetes is associated with poorer outcomes (figures 7 and 8). Diabetes mellitus was associated with increased mortality, acute heart failure, stroke, atrioventricular block, septic shock, and cardiogenic shock. The clinical profile of native valve infective endocarditis (NVIE) patients with diabetes is reported to be different compared to those without diabetes. Patients with diabetes had higher rates of comorbidities, and IE risk factors such as older age, and hemodialysis. They were less likely to have structural heart disease (valvular heart disease and congenital heart disease) and intravenous drug abuse. Patients with diabetes had higher rates of staphylococcus species, enterococci, and gram-negative microorganisms reflecting the increased health care utilization in DM patients, exposing them to nosocomial infections (26). Ampicillin with flucloxacillin or oxacillin with gentamicin is recommended as initial empirical therapy in community acquired native valves or late prosthetic valves (≥ 12 months post-surgery) endocarditis. Vancomycin with gentamicin and rifampicin is recommended in early PVE (<12 months post-surgery) or nosocomial and non-nosocomial healthcare associated endocarditis (69).

Fig 7. Two-dimensional Echocardiography of a patient with diabetes showing aortic root abscess (red arrowhead) and vegetations attached to aorto-mitral continuity (blue arrowhead), suggestive of infective endocarditis

 

Fig 8. Two-dimensional echocardiography in a patient with diabetes, showing large vegetation (blue arrowhead) attached to the posterior mitral leaflet, suggestive of infective endocarditis

Gastrointestinal Infections  

EMPHYSEMATOUS CHOLECYSTITIS

Emphysematous cholecystitis (EC) is an uncommon but serious biliary tract infection that occurs in increased frequency with male preponderance among diabetics. The common causative organisms are Clostridium perfringens and E. coli (28). Clinical findings of EC may be indistinguishable from those of uncomplicated cholecystitis although occasional crepitus may be present in some patients. The emphysematous infection is diagnosed by radiographic demonstration of gas on plain films or by CT. The treatment of choice is rapid surgical removal of the gallbladder and broad-spectrum antimicrobial therapy. Mortality caused by this infection is substantially higher than that of uncomplicated cholecystitis, ranging 15% to 25% compared with less than 4 percent (13).

LIVER ABSCESS

Diabetes is a strong, potentially modifiable risk factor for pyogenic liver abscess (figure 9). Pyogenic liver abscess patients with diabetes are older, with isolate of Klebsiella. pneumoniae being the predominant pathogen and require an increased use of combined antibiotic therapy with carbapenems. However, these patients have fewer abdominal surgeries and fewer E. coli infections as compared to patients without diabetes. In addition, poorly controlled glycemia in pyogenic liver abscess patients is associated with high incidence of fever and abscesses in both the lobes of the liver (29, 30).

Fig 9. Contrast enhanced axial (A) and sagittal (B) CT images showing multifocal well defined hypodense lesions involving both lobes of liver suggestive of liver abscesses in a patient with diabetes 

Urinary Tract Infections

The urinary tract is the most frequent site of infection in patients with diabetes (8, 70, 71). The spectrum of urinary tract infections in these patients ranges from asymptomatic bacteriuria (ASB) to lower UTI (cystitis), pyelonephritis, and severe urosepsis. Serious complications of UTI, such as emphysematous cystitis and pyelonephritis (figure 10), renal abscesses and renal papillary necrosis, are all encountered more frequently in type 2 diabetes than in the general population (72, 73).

Figure 10. Emphysematous pyelonephritis. Non contrast CT abdomen of a 45-year-old female with emphysematous pyelonephritis showing bilateral enlarged kidney with evidence of abscess formation on either side (black arrowheads) and air pockets in left kidney

 

The most common pathogens isolated from diabetic patients with UTI are E. coli, other Enterobacteriaceae such as Klebsiella spp., Proteus spp., Enterobacter spp., and Enterococci. Patients with diabetes are more prone to have resistant pathogens as the cause of their UTI, including extended-spectrum β-lactamase-positive Enterobacteriaceae, fluoroquinolone-resistant uropathogens, carbapenem-resistant Enterobacteriaceae, and vancomycin-resistant Enterococci. (32, 74).

 

As a general rule, treatment of UTI in diabetic patients is similar to that of UTI in non-diabetic patients. Antibiotic choice should be guided by local susceptibility patterns of uropathogens. First-line treatment recommendations for various types of UTI are detailed in Table 2 (74).

 

Table 2. First Line Antibiotics for Various Types of UTI in Diabetes

Type of urinary tract infection (UTI)

Gender

Antibiotic treatment

Route

Dosage

Duration of treatment

Asymptomatic bacteriuria

Male and female

None

 

 

 

Acute cystitis

Female

Nitrofurantoin

Per oral

100 mg BD/TDS

5 days

Complicated lower UTI  (catheter associated UTI)

Male and female

Ciprofloxacin

Per oral

200-500 mg BD

7-14 days

Ofloxacin

Per oral

200 mg BD

7-14 days

Trimethoprim-Sulfamethoxazole

Per oral

960 mg BD

7-14 days

Cefuroxime

Per oral

500 mg BD

7-14 days

Uncomplicated pyelonephritis

Female

Ciprofloxacin

Intravenous

400 mg BD

7 days

Ciprofloxacin

Per oral

500 mg BD

7 days

Ofloxacin

Intravenous

400 mg BD

7 days

Gentamicin

Intravenous

5 mg/kg OD

7 days

Cefuroxime

Intravenous

750 mg TDS

7-14 days

Cefuroxime

Per oral

500 mg BD

7-14 days

Complicated pyelonephritis/urosepsis

Male and female

Ciprofloxacin

Intravenous

400 mg BD

10-14 days

Ofloxacin

Intravenous

400 mg BD

10-14 days

Gentamicin

Intravenous

5 mg/kg OD

10-14 days

Amikacin

Intravenous

15 mg/kg OD

10-14 days

Piperacillin-Tazobactum

Intravenous

4.5 g TDS

10-14 days

Ertapenem

Intravenous

1 g OD

10-14 days

OD-once daily, BD-twice daily, TDS-thrice daily

Skin and Soft Tissue Infections 

Skin and soft tissue infections (SSTI) cause a substantial morbidity in patients with diabetes (75). SSTIs commonly seen in diabetes include cellulitis, abscess, decubitus ulcer, folliculitis, impetigo, carbuncle and furuncle, and surgical site infections. SSTI-associated complications such as gangrene, osteomyelitis, bacteremia, sepsis, and SSTI-associated hospitalizations are higher in patients with diabetes compared to those without diabetes (76).

FOOT INFECTIONS IN DIABETES

Foot infections in diabetes remain the most frequent complication requiring hospitalization and the most common precipitating event leading to lower extremity amputation (figure 11) (77-79).

Fig 11.  A-Trophic changes in the bilateral feet of a patient with diabetes with clawing of toes, thickened toe nails, loss of hair and shiny skin texture. B-Infected foot ulcer with slough in the plantar aspect of heel of a patient with diabetes. C-Another infected foot ulcer involving the entire sole in a patient with diabetes, the ulcer shows presence of granulation tissue along with oozing of pus and slough

 

Outcomes in patients presenting with an infected foot ulcer are poor. In one large prospective study at the end of one year, the ulcer had healed in only 46% (and it later recurred in 10% of these), while 15% had died and 17% required a lower extremity amputation (80). There are various validated classification systems to assess the severity and prognosis of foot ulcers and infection. One such scoring system is the SINBAD system which grades area, depth, sepsis, arteriopathy, and denervation plus site as either 0 or 1 point creating an easy to use scoring system that can achieve a maximum of 6 points (81). The IWGDF (International Working Group on the Diabetic Foot) infection classification is recommended to characterize and guide infection management in diabetic foot infections. The IWGDF/IDSA (Infectious Diseases Society of America) classification consists of four grades of severity for diabetic foot infection (Table 3) (82, 83).

 

Table 3. IWGDF/IDSA Classification for Foot Infections

Clinical classification of infection, with definitions

IWGDF classification

Uninfected

No systemic or local symptoms or signs of infection

 

1 (Uninfected)

Infected

·       At least, 2 of these items are present

·       Local swelling or induration

·       Erythema >0.5 cm around the wound

·       Local tenderness or pain

·       Local increased warmth

·       Purulent discharge

And no other cause(s) of an inflammatory response of the skin (eg. trauma, gout, acute Charcot neuro-osteoarthropathy, fracture, thrombosis or venous stasis)

 

Infection with no systemic manifestation involving:

·       only the skin or subcutaneous tissue (not any deeper tissues) and

·       any erythema present does not extend >2 cm around the wound

2 (mild infection)

Infection with no systemic manifestation involving:

·       erythema extending ≥ 2 cm from the wound margin, and/or

·       tissue deeper than skin and subcutaneous tissue (e.g., tendon, muscle, joint, bone)

3 (moderate infection)

Any foot infection with associated systemic manifestations (of the systemic inflammatory response syndrome [SIRS]), as mentioned by ≥2 of the following:

·       Temperature >38 degree celsius or <36 degree Celsius

·       Heart rate >90 beats/ minute

·       Respiratory rate >20 breaths/minute or PaCO2 <4.3 kPa (32 mm Hg)

·       White blood cell count >12,000/mm3 or <4000/mm3 or >10% immature (band) forms

 

4 (Severe infection)

Infection involving bone (osteomyelitis)

Add ‘O’ after 3 or 4

 

The empirical antibiotic choice is guided by the history, clinical examination, severity of infection, likely etiological agent, and previous antimicrobial sensitivity pattern. Studies from temperate climates in North America and Europe have consistently demonstrated that the most common pathogens in diabetic foot infections are aerobic gram-positive cocci, especially Staphylococus aureus, and to a lesser extent, streptococci and coagulase-negative staphylococci. More recent studies of diabetic foot infections from patients in tropical/subtropical climates (mainly Asia and northern Africa) have shown that aerobic gram-negative bacilli are often isolated, either alone or in combination with gram-positive cocci. Empirical treatment aimed at Pseudomonas aeruginosa, which usually requires either an additional or broad-spectrum agent should be considered in tropical/subtropical climates or if Pseudomonas aeruginosa has been isolated from previous cultures of the affected patient. Obligate anaerobes can play a role in diabetic foot infections, especially in ischemic limbs and in case of abscesses. Empirical treatment of these pathogens, e.g., with an imidazole (metronidazole), or beta-lactam with beta lactamase inhibitor, should be considered for diabetic foot infection associated with ischemia or a foul-smelling discharge. THE IWGDF guidelines on empirical antibiotic therapy for diabetic foot infections are outlined in table 4 (83).

 

Table 4. Empirical Antibiotic Therapy Recommended by IWGDF Guidelines for Diabetic Foot Infections

Severity of infection

Additional factors

Usual pathogen(s)

Potential empirical regimens

Mild

No complicating features

Gram positive cocci

Semi synthetic penicillin; 1st generation cephalosporins

Beta lactam allergy or intolerance

Gram positive cocci

Clindamycin;Fluroquinolone;Trimethoprim-sulfamethoxazole;Macrolide;Doxycycline

 

Recent antibiotic exposure

Gram positive cocci + Gram negative rods

β-lactamase inhibitor-amoxicillin/clavulanate; Trimethorpim-sulfamethoxazole; Fluoroquinolone

High risk for MRSA

MRSA

Linezolid; Trimethoprim-sulfamethoxazole; doxycycline; macrolide

Moderate or severe

No complicating features

Gram positive cocci ± Gram negative rods

β-lactamase inhibitor-amoxicillin/clavulanate; second or third generation cephalosoporins

 

Recent antibiotic exposure

Gram positive cocci ± Gram negative rods

β-lactamase 2-ticarcillin/clavulanate, piperacillin/tazobactum; 3rd generation cephalosporins; group I carbapenems (depends on prior therapy)

 

Macerated ulcer or warm climate

Gram negative rods including pseudomonas

β-lactamase 2-ticarcillin/clavulanate, piperacillin/tazobactum; semi synthetic penicillins + ceftazidime; semi synthetic penicillins + ciprofloxacin; group 2 carbapenems

 

Ischemic limb/necrosis/gas forming

Gram positive cocci ± Gram negative rods ± Anaerobes

β-lactamase inhibitor or 2; group 1 or 2 carbapenems; 2nd or 3rd generation cephalosporins + clindamycin or metronidazole

 

MRSA risk factors

MRSA

Consider adding or substituting with glycopeptides; linezolid; daptomycin; fusidic acid; trimethoprim-sulfamethoxazole ± rifampicin; doxycycline

 

Risk factors for resistant gram negative rods

ESBL

(Extended spectrum beta lactamase producing bacteria)

Carbapenem; Aminoglycoside and Colistin; Fluoroquinolone

MRSA-Methicillin resistant Staph aureus ; 1st generation cephalosporins-Cefadroxil, cefazolin, cephalexin; 2nd generation cephalosporins-Cefotetan, cefoxitin, cefuroxime, cefprozil; 3rd generation cephalosporins-Cefixime, cefotaxime, cefpodoxime; β-lactamase 2-ticarcillin/clavulanate, piperacillin/tazobactum; group 1 carbapenem: ertapenem; group 2 carbapenem: imipenem, meropenem, doripenem

FOURNIER’S GANGRENE

Fournier's gangrene (FG) is a fulminant form of infective necrotising fasciitis of the perineal, genital, or perianal regions, which commonly affects men with diabetes (figure 12) (84). Diabetes mellitus is reported to be present in 20%–70% of patients with Fournier’s gangrene (85). FG shows vast heterogeneity in clinical presentation, from insidious onset and slow progression to rapid onset and fulminant course, the latter being the more common presentation. The local signs and symptoms are usually dramatic with significant pain and swelling. The patient also has pronounced systemic signs; usually out of proportion to the local extent of the disease. Crepitus of the inflamed tissues is a common feature because of the presence of gas forming organisms. As the subcutaneous inflammation worsens, necrotic patches start appearing over the overlying skin and progress to extensive necrosis (86).

 

There has been an associated increased incidence of FG with the use of SGLT2 inhibitors in diabetes. The US Food and Drug Administration (FDA) has identified 55 cases of FG in patients receiving SGLT2 inhibitors between 2013 and 2019, out of which 39 were men and 16 were women (87). Time to onset of FG after initiation of SGLT2-inhibitors varied considerably, ranging from 5 days to 49 months (87). All patients were sick and had surgical debridement. Three patients died (87).  SGLT2-inhibitors cause glycosuria that can enhance the growth of bacterial flora in the urogenital milieu. This in turn increases the risk of urogenital infections, including FG. All types of SGLT2-inhibitors have been associated with FG. The FDA has issued a warning about the risk of FG to be added to the prescribing information of all SGLT2-inhibitors and to the patient medication guide.

 

Cultures from the wounds commonly show poly microbial infections by aerobes and anaerobes, which include coliforms, klebsiella, streptococci, staphylococci, clostridia, bacteroides, and corynebacteria (88). FG has a high mortality rate of 40% (85) and warrants an aggressive multimodal approach, which includes haemodynamic stabilisation, broad spectrum antibiotics and surgical debridement (86).

Fig 12. Fournier’s gangrene. Redness, swelling of the scrotum, penis and perineal tissues with necrosis and sloughing of the overlying skin

NECROTIZING FASCIITIS

Necrotizing fasciitis (NF) has been defined as a severe soft-tissue infection that causes extensive necrosis of subcutaneous tissue and fascia, relatively sparing the muscle and skin tissue (figure 13) (89). Based on bacterial culture results, NF is classified into the following categories: type I, which consists of synergistic polymicrobial infection; type II, representing infections caused by group A Streptococcus alone or combined with Staphylococcus; and type III, which comprises infections caused by Vibrio species (90).  Diabetic NF patients are reported to be more susceptible to polymicrobial and monomicrobial Klebsiella pneumoniae infections, which should be considered when choosing empirical antibiotics for these patients (91).

Fig 13. A and B Necrotizing fasciitis; black necrotic tissue and slough seen invading the subcutaneous tissues and fascia

INFECTION MIMICS IN DIABETES

Charcot Neuroarthropathy   

Charcot neuroarthropathy is a limb-threatening, destructive process that occurs in patients with neuropathy associated with medical diseases such as diabetes mellitus. Clinicians treating diabetic patients should be aware that the early signs of acute Charcot neuroarthropathy, such as pain, warmth, edema mimic foot infection. Early detection and prompt treatment can prevent joint and bone destruction, which, if untreated, can lead to morbidity and high-level amputation. The differentiation between acute presentations of Charcot’s joint and osteomyelitis is often difficult because the two conditions have many features in common. However, the lack of systemic sepsis or fever, significant hyperglycemia and leukocytosis may direct the diagnosis towards neuropathic joint (92, 93).

INFECTIONS AS A RISK FACTOR FOR DIABETES

Infections have been documented as a predisposing factor for Type 2 Diabetes Mellitus. Recent studies have revealed H. pylori infections to be significantly higher among diabetic patients than in non-diabetic patients (94, 95). Evidence suggests that advanced periodontitis also compromises glycemic control. Furthermore, periodontal treatment has been associated with improvement in glycemic control (63, 64). Abnormalities in the microbiota composition can have a major role in the development of obesity and diabetes. A reduced microbial diversity is associated with inflammation, insulin-resistance, and adiposity.  A rise in the Firmicutes/Bacteroidetes ratio is found to be related to a low-grade inflammation and to an increased capability of harvesting energy from food. Changes in some metabolites, such as short-chain fatty acids (SCFAs), produced by gut microbiota, and decreased amounts of the Akkermansia muciniphila are associated with the presence of type 2 diabetes (12). Increased pro inflammatory cytokine response in infections leads to insulin resistance. Even pathogen products, such as lipopolysaccharide and peptidoglycans, can cause insulin resistance leading to development of diabetes (96).

CONCLUSION

Awareness regarding the spectrum and severity of infections, in diabetes, is essential for prevention and prompt treatment. Strict glycemic control, proper choice of antibiotics and source control form the cornerstones of management. Preventive measures like vaccination and foot care practises go a long way in reducing infection related morbidity and mortality in diabetes.

REFERENCES

  1. International Diabetes Federation. IDF Diabetes Atlas, 9th edn. 2017. 
  2. Peleg AY, Weerarathna T, McCarthy JS, Davis TM. Common infections in diabetes: Pathogenesis, management and relationship to glycaemic control. Diabetes Metab Res Rev. 2007; 23:3–13.
  3. Thomsen RW, Hundborg HH, Lervang HH, et al. Risk of community-acquired pneumococcal bacteremia in patients with diabetes: a population-based case-control study. Diabetes Care 2004; 27:1143–7.
  4. Thomsen RW, Riis AH, Kjeldsen S, et al. Impact of diabetes and poor glycaemic control on risk of bacteremia with haemolytic streptococci groups A, B, and G. J Infect 2011;63:8–16.
  5. Thomsen RW, Hundborg HH, Lervang HH, et al. Diabetes mellitus as a risk and prognostic factor for community-acquired bacteremia due to enterobacteremia: a 10-year population based study among adults. Clin Infect Dis 2005; 40:628–31
  6. Janifer J, Geethalakshmi S, Satyavani K, Viswanathan V. Prevalence of lower urinary tract infection in South Indian type 2 diabetic subjects. Indian J Nephrol. 2009 Jul; 19(3):107.
  7. Carey IM, Critchley JA, DeWilde S, Harris T, Hosking FJ, Cook DG. Risk of infection in type 1 and type 2 diabetes compared with the general population: a matched cohort study. Diabetes Care. 2018 Mar 1;41(3):513-21.
  8. Shah BR, Hux JE. Quantifying the risk of infectious diseases for people with diabetes. Diabetes Care. 2003 Feb 1;26(2):510-3.
  9. Frydrych LM, Fattahi F, He K, Ward PA, Delano MJ. Diabetes and sepsis: risk, recurrence, and ruination. Front Endocrinol (Lausanne).2017 Oct 30;8:271.
  10. Shaw JE, Boulton AJ. The pathogenesis of diabetic foot problems: an overview. Diabetes. 1997 Sep 1;46(Supplement 2):S58-61.
  11. Mirza BA, Syed A, Izhar F, Ali Khan A. Bidirectional relationship between diabetes and periodontal disease: review of evidence. J Pak Med Assoc. 2010 Sep 1;60(9):766-8.
  12. Pascale A, Marchesi N, Govoni S, Coppola A, Gazzaruso C. The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: new insights into old diseases. Curr Opin Pharmacol. 2019 Dec 1;49:1-5.
  13. Calvet HM, Yoshikawa TT. Infect Dis Clin North Am. 2001 Jun 1;15(2):407-21.
  14. Clive S, Cockram C, Wong BCK. Diabetes and Infections. Textbook of Diabetes. Holt RI, Cockram C, Flyvbjerg A, Goldstein BJ (editors). John Wiley & Sons; 2017 Mar 6. 799-820.
  15. Harvey RA, Champe PC, Fisher BD, Strohl WA. Microbiology. Philadelphia: Lippincott Williams & Wilkins. 2007
  16. Van Veen KE, Brouwer MC, Van Der Ende A, Van De Beek D. Bacterial meningitis in diabetes patients: a population-based prospective study. Sci Rep. 2016 Nov 15;6(1):1-7.
  17. Yang TH, Xirasagar S, Cheng YF, Wu CS, Kao YW, Shia BC, Lin HC. Malignant Otitis Externa is Associated with Diabetes: A Population-Based Case-Control Study. Ann Otol Rhinol Laryngol. 2020 Jun;129(6):585-90.
  18. Carlton DA, Perez EE, Smouha EE. Malignant external otitis: the shifting treatment paradigm. Am J Otolaryngol. 2018;39(1):41-45.
  19. Wang TT, Chen TH, Wang PE, Lai H, Lo MT, Chen PY, Chiu SY. A population- based study on the association between type 2 diabetes and periodontal disease in 12,123 middle-aged Taiwanese (KCIS no. 21). J Clin Periodontol. 2009;36(5):372–9.
  20. Ali T, Rumnaz A, Urmi UL, Nahar S, Rana M, Sultana F, Iqbal S, Rahman MM, Rahman NA, Islam S, Haque M. Type-2 Diabetes Mellitus Individuals Carry Different Periodontal Bacteria. Pesqui. Bras. Odontopediatria Clín. Integr. 2021 Apr 2;21.
  21. Brunetti VC, Ayele HT, Yu OH, Ernst P, Filion KB. Type 2 diabetes mellitus and risk of community-acquired pneumonia: a systematic review and meta-analysis of observational studies. CMAJ Open. 2021 Jan;9(1):E62.
  22. Di Yacovo S, Garcia-Vidal C, Viasus D, Adamuz J, Oriol I, Gili F, Vilarrasa N, García-Somoza MD, Dorca J, Carratala J. Clinical features, etiology, and outcomes of community-acquired pneumonia in patients with diabetes mellitus. Medicine (Baltimore). 2013 Jan;92(1):42-50.
  23. Akbar DH. Bacterial pneumonia: comparison between diabetics and non-diabetics. Acta Diabetol. 2001 Jun 1;38(2):77-82.
  24. López-de-Andrés A, Perez-Farinos N, de Miguel-Díez J, Hernández-Barrera V, Jiménez-Trujillo I, Méndez-Bailón M, de Miguel-Yanes JM, Jiménez-García R. Type 2 diabetes and postoperative pneumonia: an observational, population-based study using the Spanish hospital discharge database, 2001-2015. PLoS One. 2019 Feb 6;14(2):e0211230.
  25. Movahed MR, Hashemzadeh M, Jamal MM. Increased prevalence of infectious endocarditis in patients with type II diabetes mellitus. J Diabetes Complications. 2007 Nov 1;21(6):403-6.
  26. Abe T, Eyituoyo HO, De Allie G, Olanipekun T, Effoe VS, Olaosebikan K, Mather P. Clinical outcomes in patients with native valve infective endocarditis and diabetes mellitus. World J Cardiol. 2021 Jan 26;13(1):11.
  27. Abengowe CU, McManamon PJ. Acute emphysematous cholecystitis. Can Med Assoc J. 1974 Nov 16;111(10):1112.
  28. Safwan M, Penny SM. Emphysematous cholecystitis: a deadly twist to a common disease. J Diagn Med Sonogr. 2016 May;32(3):131-7.
  29. Thomsen RW, Jepsen P, Sørensen HT. Diabetes mellitus and pyogenic liver abscess: risk and prognosis. Clin Infect Dis. 2007 May 1;44(9):1194-201.
  30. Li W, Chen H, Wu S, Peng J. A comparison of pyogenic liver abscess in patients with or without diabetes: a retrospective study of 246 cases. BMC Gastroenterol. 2018 Dec;18(1):1-9.
  31. Muller LM, Gorter KJ, Hak E, et al. Increased risk of common infections in patients with type 1 and type 2 diabetes mellitus. Clin Infect Dis. 2005;41:281–8.
  32. Geerlings SE, Meiland R, van Lith EC, Brouwer EC, Gaastra W, Hoepelman AI. Adherence of type 1-fimbriated Escherichia coli to uroepithelial cells: more in diabetic women than in control subjects.Diabetes Care. 2002 Aug; 25(8):1405-9
  33. Atreja A, Kalra S. Infections in diabetes. J Pak Med Assoc. 2015 Sep 1;65(9):1028-30.
  34. Lavery LA, Armstrong DG, Peters EJ, Lipsky BA. Probe- to-bone test for diagnosing diabetic foot osteomyelitis: reliable or relic? Diabetes Care. 2007 Feb;30(2):270-4
  35. Giurato L, Meloni M, Izzo V, Uccioli L. Osteomyelitis in diabetic foot: a comprehensive overview. World J Diabetes. 2017 Apr 15;8(4):135.
  36. Critchley JA, Carey IM, Harris T, DeWilde S, Hosking FJ, Cook DG. Glycemic control and risk of infections among people with type 1 or type 2 diabetes in a large primary care cohort study. Diabetes Care. 2018 Oct 1;41(10):2127-35.
  37. American Diabetes Association. AD Standards of medical care in diabetes--2013.Diabetes Care. 2013;36 (Suppl. 1):S11–S66.
  38. Pearson-Stuttard J, Blundell S, Harris T, Cook DG, Critchley J. Diabetes and infection: assessing the association with glycaemic control in population-based studies. Lancet Diabetes Endocrinol. 2016;4:148–158
  39. Casqueiro J, Casqueiro J, Alves C. Infections in patients with diabetes mellitus: A review of pathogenesis. Indian J Endocrinol Metab. 2012 Mar;16(Suppl1):S27.
  40. Schuetz P, Castro P, Shapiro NI. Diabetes and sepsis: preclinical findings and clinical relevance. Diabetes Care. 2011 Mar 1;34(3):771-8.
  41. Gan YH. Host susceptibility factors to bacterial infections in type 2 diabetes. PLoS Pathog. 2013 Dec 26;9(12):e1003794.
  42. Alexiewicz JM, Kumar D, Smogorzewski M, Klin M, Massry SG. Polymorpho- nuclear leukocytes in non-insulin-dependent diabetes mellitus: abnormalities in metabolism and function. Ann Intern Med. 1995;123:919–924.
  43. Collison KS, Parhar RS, Saleh SS, Meyer BF, Kwaasi AA, et al. (2002) RAGE- mediated neutrophil dysfunction is evoked by advanced glycation end products (AGEs). J Leukoc Biol. 71: 433–444.
  44. Stoeckle M, Kaech C, Trampuz A, Zimmerli W. The role of diabetes mellitus in patients with bloodstream infections. Swiss Med Wkly. 2008;138:512-9. 

  45. Ilyas R, Wallis R, Soilleux EJ, Townsend P, Zehnder D, et al. (2010) High glucose disrupts oligosaccharide recognition function via competitive inhibition: A potential mechanism for immune dysregulation in diabetes mellitus. Immunobiology 216: 126–131.
  46. Geerlings SE, Brouwer EC, VanKessel KC, Gaastra W, Stolk RP, Hoepelman AI. Cy- tokine secretion is impaired in women with diabetes mellitus. Eur J Clin Invest 2000;30:995–1001
  47. Graves DT, Naguib G, Lu H, Leone C, Hsue H, Krall E. Inflammation is more persistent in type 1 diabetic mice. J Dent Res. 2005;84:324–328
  48. Stegenga ME, van der Crabben SN, Dessing MC, et al. Effect of acute hyper- glycaemia and/or hyperinsulinaemia on proinflammatory gene expression, cyto- kine production and neutrophil function in humans. Diabet Med. 2008;25:157–164.
  49. Alberts A, Johnson A, Lewis J, Raff M, Roberts K, Walter P. The Adaptive Immune System. Molecular Biology of the Cell. New York: Garland Science; 2002.
  50. Liberatore RR Jr, Barbosa SF, Alkimin MG, et al. Is immunity in diabetic patients influencing the susceptibility to infections? Immunoglobulins, complement and phagocytic function in children and adolescents with type 1 diabetes mellitus. Pediatr Diabetes. 2005;6:206–212
  51. Lapolla A, Tonani R, Fedele D, et al. Non- enzymatic glycation of IgG: an in vivo study. Horm Metab Res. 2002;34:260– 264
  52. Spatz M, Eibl N, Hink S, et al. Impaired primary immune response in type-1 diabetes. Functional impairment at the level of APCs and T-cells. Cell Immunol. 2003; 221:15–26
  53. Rubinstein R, Genaro AM, Motta A, Cremaschi G, Wald MR. Impaired immune responses in streptozotocin-induced type I diabetes in mice. Involvement of high glucose. Clin Exp Immunol. 2008;154: 235–246.
  54. Frimodt-moller C. Diabetic cystopathy : epidemiology and related disorders. Ann Intern Med. 1980; 92: 318-321.
  55. Fünfstück R, Nicolle LE, Hanefeld M, Naber KG. Urinary tract infection in patients with diabetes mellitus. Clin Nephrol. 2012 Jan 1;77(1):40.
  56. Aumiller WD, Dollahite HA. Pathogenesis and management of diabetic foot ulcers. JAAPA. 2015 May 1;28(5):28-34.
  57. American Diabetes Association. Peripheral arterial disease in people with diabetes. Diabetes Care. 2003 Dec 1;26(12):3333-41.
  58. Kolahian S, Leiss V, Nürnberg B. Diabetic lung disease: fact or fiction? Rev Endocr Metab Disord. 2019 Sep;20(3):303-19.
  59. van de Beek D, Cabellos C, Dzupova O, Esposito S, Klein M, Kloek AT, Leib SL, Mourvillier B, Ostergaard C, Pagliano P, Pfister HW. ESCMID guideline: diagnosis and treatment of acute bacterial meningitis. Clin Microbiol Infect. 2016 May 1;22:S37-62.
  60. Pomar V, de Benito N, Mauri A, Coll P, Gurguí M, Domingo P. Characteristics and outcome of spontaneous bacterial meningitis in patients with diabetes mellitus. BMC Infect Dis. 2020 Dec;20:1-9.
  61. Carfrae MJ, Kesser BW. Malignant otitis externa. Otolaryngol Clin North Am. 2008 Jun 1;41(3):537-49.
  62. Ciorba A, Cultrera R, Di Laora A, Grilli A, Bianchini C, Aimoni C. Malignant otitis externa in the antibiotic resistance era: key to successful treatment. B-ENT. 2018. 14:119-123
  63. Preshaw PM, Alba AL, Herrera D, Jepsen S, Konstantinidis A, Makrilakis K, Taylor R. Periodontitis and diabetes: a two-way relationship. Diabetologia. 2012 Jan;55(1):21-31.
  64. D'Aiuto F, Gkranias N, Bhowruth D, Khan T, Orlandi M, Suvan J, Masi S, Tsakos G, Hurel S, Hingorani AD, Donos N. Systemic effects of periodontitis treatment in patients with type 2 diabetes: a 12 month, single-centre, investigator-masked, randomised trial. Lancet Diabetes Endocrinol. 2018 Dec 1;6(12):954-65.
  65. Huang TT, Tseng FY, Liu TC, Hsu CJ, Chen YS. Deep neck infection in diabetic patients: comparison of clinical picture and outcomes with nondiabetic patients. Otolaryngol Head Neck Surg. 2005 Jun;132(6):943-7.
  66. Saibal MA, Rahman SH, Nishat L, Sikder NH, Begum SA, Islam MJ, Uddin KN. Community acquired pneumonia in diabetic and non-diabetic hospitalized patients: presentation, causative pathogens and outcome. Bangladesh Med Res Counc Bull. 2012;38(3):98-103.
  67. Metlay JP, Waterer GW, Long AC, Anzueto A, Brozek J, Crothers K, Cooley LA, Dean NC, Fine MJ, Flanders SA, Griffin MR. Diagnosis and treatment of adults with community-acquired pneumonia. An official clinical practice guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019 Oct 1;200(7):e45-67.
  68. American Diabetes Association. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Medical Care in Diabetes—2021. Diabetes Care. 2021 Jan;44(Supplement 1):S40-52.
  69. Habib G, Lancellotti P, Antunes MJ, Bongiorni MG, Casalta JP, Del Zotti F, Dulgheru R, El Khoury G, Erba PA, Iung B, Miro JM. 2015 ESC guidelines for the management of infective endocarditis: the task force for the management of infective endocarditis of the European Society of Cardiology (ESC) endorsed by: European Association for Cardio-Thoracic Surgery (EACTS), the European Association of Nuclear Medicine (EANM). Eur Heart J. 2015 Nov 21;36(44):3075-128.
  70. Patterson JE, Andriole VT. Bacterial urinary tract infections in diabetes. Infect Dis Clin North Am. 1997;11(3):735–750.
  71. Joshi N, Caputo GM, Weitekamp MR, Karchmer AW. Infections in patients with diabetes mellitus.N Engl J Med. 1999;341(25):1906–1912
  72. Mnif MF, Kamoun M, Kacem FH, et al. Complicated urinary tract infections associated with diabetes mellitus: pathogenesis, diagnosis and management.Indian J Endocrinol Metab. 2013;17(3):442–445.
  73. Kofteridis DP, Papadimitraki E, Mantadakis E, et al. Effect of diabetes mellitus on the clinical and microbiological features of hospitalized elderly patients with acute pyelonephritis. J Am Geriatr Soc. 2009;57(11):2125–2128.
  74. Nitzan O, Elias M, Chazan B, Saliba W. Urinary tract infections in patients with type 2 diabetes mellitus: review of prevalence, diagnosis, and management. Diabetes Metab Synd Obes. 2015;8:129.
  75. Lipsky BA, Tabak YP, Johannes RS, Vo L, Hyde L, Weigelt JA. Skin and soft tissue infections in hospitalised patients with diabetes: culture isolates and risk factors associated with mortality, length of stay and cost. Diabetologia. 2010 May;53(5):914-23.
  76. Suaya JA, Eisenberg DF, Fang C, Miller LG. Skin and soft tissue infections and associated complications among commercially insured patients aged 0–64 years with and without diabetes in the US. PLoS One. 2013 Apr 10;8(4):e60057.
  77. Ndosi M, Wright-Hughes A, Brown S, et al. Prognosis of the infected diabetic foot ulcer: a 12-month prospective observational study. Diabet Med 2018;35:78-88.
  78. Lavery LA, Armstrong DG, Murdoch DP, Peters EJ, Lipsky BA. Validation of the Infectious Diseases Society of America's diabetic foot infection classification system. Clin Infect Dis 2007;44:562-5.
  79. Tan TW, Shih CD, Concha-Moore KC, et al. Disparities in outcomes of patients admitted with diabetic foot infections. PLoS One 2019;14:e0211481.
  80. Ndosi M, Wright-Hughes A, Brown S, et al. Prognosis of the infected diabetic foot ulcer: a 12-month prospective observational study. Diabet Med 2018;35:78-88.
  81. Ince P, Abbas ZG, Lutale JK, Basit A, Ali SM, Chohan F, et al. Use of the SINBAD classification system and score in comparing outcome of foot ulcer management on three continents. Diabetes Care. 2008;31(5):964-7.
  82. Monteiro‐Soares M, Russell D, Boyko EJ, Jeffcoate W, Mills JL, Morbach S, Game F, International Working Group on the Diabetic Foot (IWGDF). Guidelines on the classification of diabetic foot ulcers (IWGDF 2019). Diabetes Metab Res Rev. 2020 Mar;36:e3273.
  83. Lipsky BA, Senneville É, Abbas ZG, Aragón‐Sánchez J, Diggle M, Embil JM, Kono S, Lavery LA, Malone M, van Asten SA, Urbančič‐Rovan V. Guidelines on the diagnosis and treatment of foot infection in persons with diabetes (IWGDF 2019 update).Diabetes Metab Res Rev. 2020 Mar;36:e3280.
  84. Smith G L, Bunker C B, Dineeen M D. Fournier's gangrene.Br J Urol 1998 Mar;81(3):347-55.
  85. Morpurgo E, Galandiuk S. Fournier's gangrene. Surg Clin North Am. 2002 Dec 1;82(6):1213-24.
  86. Thwaini A, Khan A, Malik A, Cherian J, Barua J, Shergill I, Mammen K. Fournier’s gangrene and its emergency management. Postgrad Med J. 2006 Aug 1;82(970):516-9.
  87. 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 Med. 2019 Jun 4;170(11):764-769.
  88. Yaghan RJ, Al-Jaberi TM, Bani-Hani I. Fournier's gangrene. Dis Colon Rectum. 2000 Sep 1;43(9):1300-8.
  89. Lin C, Yeh FL, Lin JT, Ma H, Hwang CH, Shen BH, et al. Necrotizing fasciitis of the head and neck: an analysis of 47 cases.Plast Reconstr Surg. 2001;107(7):1684–93.
  90. Giuliano A, Lewis F, Jr, Hadley K, Blaisdell FW. Bacteriology of necrotizing fasciitis.Am J Surg. 1977;134(1):52–7.
  91. Cheng NC, Tai HC, Chang SC, Chang CH, Lai HS. Necrotizing fasciitis in patients with diabetes mellitus: clinical characteristics and risk factors for mortality. BMC Infect Dis. 2015 Dec;15(1):1-9.
  92. Jain R, Brown M, Boynton EL. Not another Diabetic Foot Infection: A Case Report of Charcot's Joint. Can J Plastic Surg. 1996 Dec;4(4):1-6.
  93. Varma AK. Charcot neuroarthropathy of the foot and ankle: a review. J Foot Ankle Surg. 2013 Nov 1;52(6):740-9.
  94. Mabeku LB, Ngamga ML, Leundji H. Helicobacter pylori infection, a risk factor for Type 2 diabetes mellitus: a hospital-based cross-sectional study among dyspeptic patients in Douala-Cameroon. Sci Rep. 2020 Jul 22;10(1):1-1.
  95. Bener A, Ağan AF, Al-Hamaq AO, Barisik CC, Öztürk M, Ömer A. Prevalence of Helicobacter pylori infection among type 2 diabetes mellitus. Adv Biomed Res. 2020;9:27.
  96. Chakraborty S, Bhattacharyya R, Banerjee D. Infections: a possible risk factor for type 2 diabetes. Adv Clin Chem. 2017 Jan 1;80:227-51.

 

 

 

Etiologic Classification of Diabetes Mellitus

 

Table 1 lists the various disorders that can either cause or contribute to the development of diabetes and the Endotext chapters where these disorders are discussed in detail.

 

 

Table 1. Etiologic Classification Of Diabetes Mellitus

Disorders

Endotext Chapter

Type 1 Diabetes

Pathogenesis of Type 1A Diabetes

Type 2 Diabetes

Pathogenesis of Type 2 Diabetes

Gestational Diabetes

Gestational Diabetes

Genetic defects of beta-cell development and function

MODY

Diagnosis and Clinical Management of Monogenic Diabetes

Neonatal Diabetes

Diagnosis and Clinical Management of Monogenic Diabetes

Mitochondrial DNA

Atypical Forms of Diabetes

Genetic defects in insulin action

Type A insulin resistance

Atypical Forms of Diabetes

Leprechaunism

Atypical Forms of Diabetes

Rabson-Mendenhall syndrome

Atypical Forms of Diabetes

Lipoatrophic diabetes

Lipodystrophy Syndromes: Presentation and Treatment*

Diseases of the exocrine pancreas

Pancreatitis

Atypical Forms of Diabetes

Trauma/pancreatectomy

Atypical Forms of Diabetes

Neoplasia

Atypical Forms of Diabetes

Cystic fibrosis

Atypical Forms of Diabetes

Iron overload (hemochromatosis, thalassemia, etc.)

Atypical Forms of Diabetes

Fibrocalculous pancreatic diabetes

Fibrocalculous Pancreatic Diabetes**

Endocrinopathies

Acromegaly

Cushing’s syndrome

Glucagonoma

Pheochromocytoma

Hyperthyroidism

Somatostatinoma

Primary Hyperaldosteronism

Atypical Forms of Diabetes

Diabetes Mellitus After Solid Organ Transplantation

Diabetes Mellitus After Solid Organ Transplantation

Drug- or chemical-induced hyperglycemia

Vacor

Pentamidine

Nicotinic acid

Glucocorticoids

Growth Hormone

Diazoxide

Check point inhibitors

Dilantin

Interferon alpha

Immune suppressants

Others (statins, psychotropic drugs, b-Adrenergic agonists, thiazides, etc.)

Atypical Forms of Diabetes

Infections

Congenital rubella

Atypical Forms of Diabetes

HCV

Atypical Forms of Diabetes

COVID-19

Atypical Forms of Diabetes

HIV

Diabetes in People Living with HIV

Immune-mediated diabetes

Latent Autoimmune Diabetes in Adults (LADA)

Atypical Forms of Diabetes

Stiff-man syndrome

Atypical Forms of Diabetes

Anti-insulin receptor antibodies

Atypical Forms of Diabetes

Autoimmune Polyglandular Syndromes

Autoimmune Polyglandular Syndromes***

Diabetes of unknown cause

Ketosis-prone diabetes (Flatbush diabetes)

Atypical Forms of Diabetes

Other genetic syndromes sometimes associated with diabetes

Down syndrome

Klinefelter syndrome

Turner syndrome

Wolfram syndrome

Friedreich ataxia

Huntington chorea

Bardet-Biedl syndrome (Laurence-Moon-Biedl syndrome)

Myotonic dystrophy

Porphyria

Prader-Willi syndrome

Alström syndrome

Others

Atypical Forms of Diabetes

Unless indicated chapters are located in the

*Chapter in Diagnosis and Treatment of Diseases of Lipid and Lipoprotein Metabolism section

**Chapter in Tropical Medicine section

***Chapter in Disorders that Affect Multiple Organs section

 

 

 

Control of Energy Expenditure in Humans

ABSTRACT

 

Resting and meal-related energy requirements can be assessed by measuring energy expenditure with indirect calorimetry. The indicated method to assess free-living energy expenditure is the doubly labelled water technique. Variation in energy expenditure is mainly a function of body size and composition (resting energy expenditure) and of physical activity (activity energy expenditure). Thus, energy expenditure can be calculated with a prediction equation for resting energy expenditure, based on height, age, weight and sex, in combination with the measurement of the physical activity level of a subject with a doubly labelled water validated accelerometer for movement registration. Energy balance in humans is maintained by adjusting energy intake to energy expenditure. Over- and underfeeding induces changes in activity-induced and maintenance energy expenditure as a function of changes in body weight and body composition. Additionally, underfeeding causes a metabolic adaptation as reflected in a reduction of maintenance energy expenditure below predicted values and defined as adaptive thermogenesis. When intake exceeds energy requirements, the excess is primarily stored as body fat. As a substrate for energy metabolism, fat is less likely to be oxidized for fuel than carbohydrate or protein. Consumed fat is mostly stored before oxidation, especially in heavier people, increasing the likelihood of creating a positive energy balance. An activity-induced increase in energy requirement is typically followed by an increase in energy intake, whereas a reduction in physical activity does not result in an equivalent reduction of energy intake. Thus, preventing weight gain is more effectively reached by eating less than by moving more.

 

MEASURING ENERGY EXPENDITURE

 

Living can be regarded as a combustion process. The metabolism of an organism requires energy production by the combustion of fuel in the form of carbohydrate, protein, fat, or alcohol. In this process oxygen is consumed and carbon dioxide produced. Measuring energy expenditure means measuring heat production or heat loss, and this is known as direct calorimetry. The measurement of heat production by measuring oxygen consumption and/or carbon dioxide production is called indirect calorimetry.

 

Early calorimeters for the measurement of energy expenditure were direct calorimeters. At the end of the 18th century Lavoisier constructed one of the first calorimeters, measuring energy expenditure in a guinea pig. The animal was placed in a wire cage, which occupied the center of an apparatus. The surrounding space was filled with chunks of ice (Figure 1). As the ice melted from the animal's body heat, the water was collected in a container, and weighed. The ice cavity was surrounded by a space filled with snow to maintain a constant temperature. Thus, no heat could dissipate from the surroundings to the inner ice jacket. Today, heat loss is measured in a calorimeter by removing the heat with a cooling stream of air or water or measuring the heat flow through the wall. In the first case, heat conduction through the wall of the calorimeter is prevented and the flow of heat is measured by the product of temperature difference between inflow and outflow and the rate of flow of the cooling medium. In the latter case instead of preventing heat flow through the wall, the rate of this flow is measured from differences in temperature over the wall. This method is known as gradient layer calorimetry.

Figure 1. Lavoisier’s calorimeter. Heat expended by the animal melts the ice in the inner jacket. Snow in the outer jacket prevents heat exchange with the surrounding environment (From reference (1)).

In indirect calorimetry, heat production is calculated from chemical processes. Knowing, for example, that the oxidation of 1 mol glucose requires 6 mol oxygen and produces 6 mol water, 6 mol carbon dioxide and 2.8 MJ heat, the heat production can be calculated from oxygen consumption or carbon dioxide production. Heat production and the energy equivalent of oxygen and carbon dioxide varies with the nutrient oxidized (Tables 1 and 2).

 

Table 1. Gaseous Exchange and Heat Production of Metabolized Nutrients

Nutrient

Consumption oxygen (l/g)

Production carbon dioxide (l/g)

Heat (kJ/g)

Carbohydrate

0.829

0.829

17.5

Protein

0.967

0.775

18.1

Fat

2.019

1.427

39.6

 

Brouwer (2) drew up simple formulae for calculating the heat production and the quantities of carbohydrate (C), protein (P) and fat (F) oxidized from oxygen consumption, carbon dioxide production and urine-nitrogen loss. The principle of the calculation consists of three equations with the mentioned three measured variables:

 

Oxygen consumption              = 0.829 C + 0.967 P + 2.019 F

Carbon dioxide production      = 0.829 C + 0.775 P + 1.427 F

Heat production                       = 21.1 C + 18.7 P + 19.6 F

 

Usually, only urine nitrogen is measured when information on the contribution of C, P, and F to energy production is needed. Protein oxidation (g) is calculated as 6.25 x urine-nitrogen (g), and subsequently oxygen consumption and carbon dioxide production can be corrected for protein oxidation to allow calculation of carbohydrate and fat oxidation. The general formula for the calculation of energy production (E) derived from these figures is: 

 

E = 16.20 * oxygen consumption + 5.00 * carbon dioxide production - 0.95 P.

 

In this formula the contribution of protein (P) to energy production (E), the so-called protein correction, is very small. In the case of a normal protein oxidation of 10-15 per cent of the daily energy production, the protein correction for the calculation of E is about one per cent. For this reason, in the calculation of energy production, the protein correction is often neglected.

 

Metabolizable energy is available for energy production in the form of heat and for external work. At present, the state of the art for assessing total energy expenditure is with indirect calorimetry. With indirect calorimetry, the energy expenditure is calculated from gaseous exchange of oxygen and carbon dioxide. The result is the total energy expenditure of the body for heat production and work output. With direct calorimetry, only heat loss is measured. At rest, total energy expenditure is converted to heat. During physical activity, there is work output as well. The proportion of energy expenditure for external work is the work efficiency. At rest, indirect calorimetry-assessed energy expenditure matches heat loss as measured with direct calorimetry. During physical activity, heat loss is systematically lower than indirect calorimetry-assessed energy expenditure and can be up to 25% lower than total energy expenditure during endurance exercise. The difference increases with exercise intensity. For example, during cycling, indirect calorimetry assessed energy expenditure matches the sum of heat loss and power output (3) and work efficiency during cycling, the power output divided by energy expenditure, is in the range of 15 to 25%.

 

Current techniques utilizing indirect calorimetry for the measurement of energy expenditure in humans include a facemask or ventilated hood, respiration chamber (whole room calorimeter), and the doubly labelled water method. A facemask is typically used to measure energy expenditure during standardized activities on a treadmill or a cycle ergometer. A ventilated hood is used to measure resting energy expenditure and energy expenditure during nutrient processing and absorption (diet-induced energy expenditure). A respiration chamber is an airtight room that is ventilated with fresh air, with the only difference between a usually, ventilated hood system and respiration chamber being size. In a respiration chamber the subject is fully enclosed instead of enclosing the head only, allowing physical activity depending on the size of the chamber. For measurements under a hood or in a respiration chamber, air is pumped through the system and blown into a mixing chamber where a sample is taken for analysis. Measurements taken are those of the airflow and of the oxygen and carbon dioxide concentrations of the air flowing in and out. The most common device to measure the airflow is a dry gas meter comparable to that used to measure natural gas consumption at home. The oxygen and carbon dioxide concentrations are commonly measured with a paramagnetic oxygen analyzer and an infrared carbon dioxide analyzer respectively. The airflow is adjusted to keep differences in oxygen and carbon dioxide concentrations between inlet and outlet within a range of 0.5 to 1.0%. For adults, this means airflow rates around 50 l/min at rest under a hood, 50-100 l/min when sedentary in a respiration chamber, while in exercising subjects the flow has to be increased to over 100 l/min. In the latter situation, one has to choose a compromise for the flow rate when measurements are to be continued over 24 hours that include active and inactive intervals. During exercise bouts, the 1% carbon dioxide level should not be surpassed for long. During times of rest, like an overnight sleep, the level should not fall too far below the optimal measuring range of 0.5-1.0%. Changing the flow rate during an observation interval reduces the accuracy of the measurements due to the response time of the system. Though the flow rate of a hood and a chamber system is comparable, the volume of a respiration chamber is more than 20 times the volume of a ventilated hood. Consequently, the minimum length of an observation period under a hood is about 0.5 hours and in a respiration chamber in the order of 5-10 hours.

 

The doubly labelled water method is an innovative variant on indirect calorimetry based on the discovery that oxygen in the respiratory carbon dioxide is in isotopic equilibrium with the oxygen in body water. This technique involves enriching the body water with an isotope of oxygen and an isotope of hydrogen and then determining the washout kinetics of both isotopes. Doubly labelled water provides an excellent method to measure total energy expenditure in unrestrained humans in their normal surroundings over a time period of one to four weeks. After enriching the body water with labelled oxygen and hydrogen by drinking doubly labelled water, most of the oxygen isotope is lost as water, but some is also lost as carbon dioxide because CO2 in body fluids is in isotopic equilibrium with body water due to exchange in the bicarbonate pools (4). The hydrogen isotope is lost as water only. Thus, the washout for the oxygen isotope is faster than for the hydrogen isotope, and the difference represents the CO2 production. The isotopes of choice are the stable, heavy, isotopes of oxygen and hydrogen, oxygen-18 (18O) and deuterium (2H), since these avoid the need to use radioactivity and can be used safely. Both isotopes naturally occur in drinking water and thus in body water. The CO2 production, calculated from the difference in elimination between the two isotopes, is a measure of metabolism. In practice, the observation duration is set by the biological half-life of the isotopes as a function of the level of the energy expenditure. The minimum observation duration is about three days in subjects with high energy turnover like premature infants or endurance athletes. The maximum duration is 30 days or about 4 weeks in elderly (sedentary) subjects. An observation period begins with collection of a baseline sample. Then, a weighed isotope dose is administered, usually a mixture of 10% 18O and 6% 2H in water. For a 70 kg adult, between 100-150 cc water would be used. Subsequently, the isotopes equilibrate with the body water and the initial sample is collected. The equilibration time is dependent on body size and metabolic rate. For an adult the equilibration would take between 4-8 hours. During equilibration, the subject usually does not consume any food or drink. After collecting the initial sample, the subject performs routines according to the instructions of the experimenter. Body water samples (blood, saliva or urine) are collected at regular intervals until the end of the observation period. The doubly labelled water method gives precise and accurate information on carbon dioxide production. Converting carbon dioxide production to energy expenditure needs information on the energy equivalent of CO2 (Table 2), which can be calculated with additional information on the substrate mixture being oxidized. One option is the calculation of the energy equivalent from the macronutrient composition of the diet. In energy balance, substrate intake and substrate utilization are assumed to be identical.

 

Table 2. Energy Equivalents of Oxygen and Carbon Dioxide

Nutrient

Oxygen (kJ/l)

Carbon dioxide (kJ/l)

Carbohydrate

21.1

21.1

Protein

18.7

23.4

Fat

19.6

27.8

 

ENERGY EXPENDITURE AND COMPONENTS

 

Daily energy expenditure consists of four components: 1) sleeping metabolic rate, 2) the energy cost of arousal, 3) the thermic effect of food (or diet-induced energy expenditure (DEE)), and 4) the energy cost of physical activity or activity-induced energy expenditure (AEE). Usually, sleeping metabolic rate and the energy cost of arousal are combined and referred to as resting energy expenditure (REE). Overnight when one sleeps quietly, food intake and physical activity are generally low or absent and energy expenditure gradually decreases to a daily minimum before increasing upon awakening (Figure 2). Then, increases in energy expenditure during arousal are primarily the result of activity-induced energy expenditure as well as diet-induced energy expenditure. Thus, energy expenditure varies throughout a day as a function of body size and body composition (the major components determining REE), physical activity as determinant of AEE, and food intake as determinant of DEE.

Figure 2. Average energy expenditure (upper line) and physical activity (lower line) as measured over a 24-h interval in a respiration chamber. Arrows denote meal times. Data are the average of 37 subjects, 17 women and 20 men, age 20-35 y and body mass index 20-30 kg/m2 (5).

Resting energy expenditure is defined as the metabolic rate required to maintain vital physiological functions of an individual that is in rest, awake, in a fasted state, and in a thermoneutral environment. To perform an accurate measurement of REE, a subject is instructed not to exercise the day before, to fast overnight, transported to a laboratory after waking up in the morning and habituated for 15-30 min to the testing procedure under a ventilated hood, before the actual measurement of 20-30 min, at a comfortable room temperature of 22-24 0C (6).

 

Standardizing to fat-free mass as an estimate of metabolic body size is most commonly used in the literature to compare REE between individuals. However, although fat-free body mass is a strong predictor of REE, energy expenditure should not be solely divided by the absolute fat-free mass value as the relationship between energy expenditure and fat-free mass has an Y-intercept (the value for energy expenditure when fat-free mass is theoretically absent) that is not zero (Figure 3). For example, fat-free adjusted REE is significantly different between women and men (Figure 3, 0.143±0.012 and 0.128±0.080 MJ/kg for women and men, respectively, P < 0.0001). The smaller the fat-free mass, the higher the REE/ fat-free mass ratio and thus the REE per kg fat-free mass is on average higher in women than men. Instead, a more accurate approach for comparing REE data is by regression analysis that includes both fat-free mass and fat mass as covariates.

 

REE (MJ/d) = 1.39 + 0.93 fat-free mass (kg) + 0.039 fat mass (kg), r2 = 0.93.

 

Using this equation, gender no longer comes out as a significant contributor to the explained variation in the group of women and men (Figure 3).

 

Figure 3. Resting energy expenditure (REE) plotted as a function of fat-free mass for the subjects from reference 5 as described in Figure (2) (17 women: closed symbols; 20 men: open symbols) with the calculated linear regression line (REE (MJ/d) = 2.27 + 0.091 fat-free mass (kg), r2 = 0.78).

Diet-induced energy expenditure is defined as the energy-required for intestinal absorption of nutrients, the initial steps of their metabolism and the storage of the absorbed but not immediately oxidized nutrients during the post-prandial period. As such, the amount of food ingested quantified as the energy content of the food is a determinant of DEE. The most common way to express DEE is derived from the difference between energy expenditure after food consumption and REE, divided by the rate of nutrient energy administration. Theoretically, based on the amount of ATP required for the initial steps of metabolism and storage, the DEE is different for each nutrient. Reported DEE values for separate nutrients are 0 to 3% for fat, 5 to 10% for carbohydrate, and 20 to 30% for protein (7). In healthy subjects in energy balance with a mixed diet, DEE represents about 10% of the total amount of energy ingested over 24 hours.

 

A typical mean pattern of DEE throughout the day is presented in Figure 4. Data are from a study where DEE was calculated by plotting the residual of the individual relationship between energy expenditure and physical activity in time, as measured over 30-min intervals from a 24-h observation in a respiration chamber. The level of REE after waking up in the morning, and directly before the first meal, was defined as basal metabolic rate. Resting metabolic rate had still not returned to basal metabolic rate before lunch four hours after breakfast, or before dinner at five hours after lunch. Instead, basal metabolic rate was restored overnight, approximately eight hours after dinner consumption.

Figure 4. The mean pattern of resting energy expenditure throughout the day, where arrows denote meal times (adapted from reference (8)).

Activity-induced energy expenditure, the most variable component of daily energy expenditure, is derived from total energy expenditure (TEE) minus resting energy expenditure and diet-induced energy expenditure.

 

AEE = TEE – REE – DEE.

 

Total energy expenditure is measured with doubly labelled water as described above. When diet induced energy expenditure is assumed to be 10% of TEE in subjects consuming the average mixed diet and being in energy balance, AEE can be calculated as: AEE = 0.9 TEE – REE.

 

A frequently used method to quantify the physical activity level (PAL) of a subject is to express TEE as a multiple of REE:

 

PAL = TEE/REE.

 

This assumes that the variation in total energy expenditure is due to body size and physical activity. The effect of body size is corrected for by expressing TEE as a multiple of REE. Data on daily energy expenditure, as measured with doubly labelled water, permit the evaluation of limits to the physical activity level. In our site, data were compiled for more than 500 subjects, where energy expenditure was measured over an interval of two weeks with the same protocol. The sample excludes individuals aged less than 18 years, involved in interventions of restricted or forced excess energy intake, whose physical activity including athletic performance, who were pregnant or lactating, and with an acute or chronic illness. The sample includes similar numbers of women and men, with a wide range for age, height, weight, and body mass index. Despite the wide variation in subject characteristics, a narrow range of the physical activity level (between 1.1 and 2.75) amongst the subjects was found (Figure 5) with no sex differences (9).

 

The physical activity level of a subject can be classified in three categories as defined by the last Food and Agriculture Organization/World Health (FAO/WHO/UNU) expert consultation on human energy requirements (10). The physical activity for sedentary and light activity lifestyles ranges between 1.40 and 1.69, for moderately active or active lifestyles between 1.70 and 1.99, and for vigorously active lifestyles between 2.00 and 2.40. An active lifestyle improves heath parameters like insulin sensitivity (11). Higher PAL values, while difficult to maintain over a long period, generally result in weight loss.

 

An alternative for the measurement of energy expenditure with indirect calorimetry is a prediction equation for resting energy expenditure, in combination with an estimation of activity energy expenditure from measurement of body movement with an accelerometer. Typically, prediction equations for resting energy expenditure can explain 70-80% of the variation from race, height, age, weight and gender of a subject (12). Doubly labelled water studies show the best accelerometers for movement registration so far can explain 50-70% of variation in activity energy expenditure (13).

Figure 5. Frequency distribution of the value of the physical activity level (PAL) calculated as the total energy expenditure / resting energy expenditure, in a group of 556 healthy adults, women closed bars and men open bars (data from reference (9)).

DETERMINANTS OF ENERGY EXPENDITURE

 

The main determinants of energy expenditure are body size and body composition, food intake, and physical activity. Additional determinants are ambient temperature and health. As most people are able to live in a thermoneutral environment or prevent heat loss with appropriate clothing, energy expenditure is not affected by ambient temperature for longer time intervals.

Body size and body composition determine REE, the largest component of daily energy expenditure (Figure 6). Energy expenditure is generally higher in men than in women because men generally have a larger metabolic body size. They are on average heavier than women and for the same weight men have relatively more fat-free mass. For similar reasons, gaining weight implicates gaining fat mass and fat-free mass, and daily energy expenditure is generally higher in people who are overweight and have obesity compared with people who are lean matched for age, height and gender. This higher energy expenditure in people with obesity is mainly a consequence of higher resting energy expenditure than people who are lean (Figure 6).

Figure 6. The three components of energy expenditure: resting energy expenditure (closed bar), diet-induced energy expenditure (stippled bar), and activity-induced energy expenditure (open bar) as observed in subjects who are lean and who have obesity. In the lean group, women and men weighed 61 kg and 74 kg with 29% and 17% body fat, respectively. In the group with obesity, subjects were, on average, 40 kg heavier, where 70% of the additional weight was fat mass and 30% fat-free mass. The figure illustrates the higher energy expenditure (primarily in resting energy expenditure) in men than women and in those with obesity compared to those who are lean. (After reference (14)).

Food intake affects all three components of daily (total) energy expenditure: REE, DEE and AEE. The most obvious effect is on DEE, which represents about 10% of the amount of daily energy ingested. Thus, changing energy intake changes total energy expenditure accordingly. Overeating induces an additional increase for storage of excess energy, estimated at about 10 % of the energy surplus (15). When overfeeding is lower than twice the maintenance requirements, there does not seem to be an effect of this overfeeding on physical activity (16). Undereating induces a decrease in REE, DEE and AEE. Undereating induces weight loss accompanied by adaptive thermogenesis, a disproportional or greater than expected reduction of REE. The reduction in REE is sustained even while weight loss is maintained (17). Weight loss due to a negative energy balance is accompanied by a decrease in AEE as well. Here, the decrease is due to less body movement and a lower cost to move a smaller body mass. The reduction in body movement recovers to baseline values or higher when weight loss in maintained (18). A classic example of the effect of undereating on energy expenditure is the Minnesota Experiment from the 1950’s (19). Energy intake of normal-weight men was reduced for 24 weeks from 14.6 MJ/d to 6.6 MJ/d. The subjects reached a new energy balance by saving 8 MJ/d (Table 3). Of the total saving of 8 MJ/d the main part stemmed from reduced AEE, which was mainly due to moving less.

 

Table 3. Energy Saved by 24 Weeks Underfeeding in the Minnesota Experiment (19)

 

MJ/d

% of saving

Explanation

Resting energy expenditure

2.6

32

65% for a decreased bodyweight

35% for a lowered tissue metabolism

Diet-induced expenditure

0.8

10

 

Activity-induced expenditure

4.7

58

40% for a decreased bodyweight

60% for less body movement

Total

8.0

   

 

Activity induced energy expenditure is the most variable component of daily expenditure and can be increased through exercise. Variation in energy expenditure between subjects is a function of body size and physical activity, where AEE is an important contributor. Most of the variation in AEE is accounted for by genetic factors. Genes determine for a large part whether a person is prone to engage in activities and how much energy is expended for these activities (20). Exercise training can increase AEE. However, under some conditions the added exercise expenditure is compensated for by a reduction of non-training activity. Examples are non-ad libitum food intake and older age (Figure 7).

 

Figure 7. The physical activity level, total energy expenditure as a multiple of resting energy expenditure, before (open bar) and at the end of a training program (closed bar), for eight studies displayed in a sequence of age of the participants as displayed on the horizontal axis (After reference (21)).

Activity-induced energy expenditure does not increase linearly with increasing physical activity. For example, novice runners training to run a half marathon could increase the training amount without a change in AEE (22). In the selected group of sedentary subjects, the initial training-induced increase in AEE was twice as high as predicted from the training load. However, subsequent training allowed a doubling of the training load for the same AEE, probably through an improvement of exercise economy. Similarly, exercise training has been shown to decrease the energetic cost of walking in older adults (23).

 

Physical activity level reaches a maximum value of 2.0-2.4 (Figure 7). Higher values can be reached over shorter time intervals. For example, runners in a 140-day transcontinental race across the USA showed an initial increase in PAL from a pre-race value of 1.76 to 3.76 over the first five days of running (24). In the final week (week 20) of running, PAL had decreased to a mean value of 2.81. This subsequent decrease in PAL during sustained physical activity was hypothesized to have resulted from a limit in alimentary energy supply.

 

During negative energy balance, additional exercise is compensated by a reduction of non-training activity. In elderly subjects, exercise training has a similar compensatory effect on spontaneous physical activity, even under ad-libitum food conditions. Despite the absence of an effect of exercise training on total energy expenditure in elderly people, there are many beneficial effects of exercise training like aerobic capacity, endurance, flexibility, and range of motion.

 

ENERGY BALANCE

 

Adult humans maintain weight stability through a balance between energy intake and energy expenditure. When weight is stable, the energy store of the body does not fluctuate much, as evident by constancy in body weight and body composition. This weight constancy can be achieved through the balanced control of energy intake and expenditure. This balance does not, however, take place on an immediate basis. For example, on days with high energy expenditure, energy intake is usually normal or even below normal. The 'matching' increase in energy intake comes several days afterwards (25). Energy intake can change by at least a factor of three when adapting to changes in energy expenditure. Under sedentary living conditions the energy balance is maintained at about 1.5 times basal metabolic rate (BMR), while during sustained exercise levels of 4.5 times BMR are reached (26).

 

Humans are discontinuous eaters and continuous metabolizers. An animal that takes its food in meals, such as a human, periodically consumes more than their physiological needs even when in (daily) energy balance. During meal-related hyperphagia, metabolites are initially stored then mobilized during inter-meal intervals of energy deficiency. This pattern of intermittent feeding and fasting has consequences for energy expenditure (Figure 4). During and after a meal, expended energy increases to process the ingested food, while energy deficiency before a new meal is started can lead to a reduction of energy expenditure. The latter probably does not occur during short-term energy deficiency. However, people tend to be less energetic during prolonged inter-meal intervals or extended fasts.

 

Disturbances of energy balance result in energy mobilization from, or energy storage in, body reserves. Energy intake occurs via macronutrients consumed in meals in the form of carbohydrate, protein, fat and alcohol. During positive energy balance, excess energy is stored as carbohydrate in glycogen, primarily in the liver, and as fat in adipose depots. The storage capacity for carbohydrate is small, typically covering energy needs during the overnight fast that accompanies sleep. Longer-term shortages are mainly covered by mobilization of the larger energy stores in fat. On days with a positive energy balance, protein and carbohydrate intake match protein and carbohydrate oxidation and the difference between energy intake and energy expenditure shows up in a positive fat balance (27). In the early morning, at arousal, carbohydrate oxidation goes up and continues to increase at the first food intake of the day (28). After awakening, initial energy (‘fast’) requirements are met by glycogen reserves. Subsequently, carbohydrate requirement is higher at breakfast, and one eats relatively more fat at the evening dinner (29,30).

 

Energy balance does not equate to substrate balance, and when in substrate balance one does not produce energy just from the foods consumed. Fat, as a substrate for energy metabolism is at the bottom of the oxidation hierarchy that determines fuel selection and studies show a direct link between macronutrient balance for fat and energy balance. Changes in alcohol, protein, and carbohydrate intake elicit auto regulatory adjustments in oxidation whereas a change in fat intake fails to elicit such a response, or only in the long term (31).

 

One explanation for this macronutrient oxidation disparity is the routing of dietary fat. Fat metabolism can be traced with isotope-labelled fatty acids. Oxidation and adipose tissue uptake of dietary fat can be measured by adding fatty acid labelled with heavy hydrogen (2H) to meals. Upon oxidation, these deuterated fatty acids enrich the body water with deuterium, which is subsequently detectable in urine. Therefore, the urine enrichment for deuterium is a measure of dietary fat oxidation. The first label appears in the urine in about two hours and the peak concentration is reached after 12-24h (Figure 8). After 24 hours, 5-30% of the fat from a meal is oxidized and the remaining part partitioned to the reserves. The percentage of dietary fat oxidation is independent of the composition of the meal with respect to protein, carbohydrate and fat. However, there is a clear relation of dietary fat oxidation with the body fat content. The larger the fat mass, the lower the fractional oxidation of the fat consumed on the same day (32). The observed reduction in dietary fat oxidation in subjects with greater body fat may therefore play a role in expression and maintenance of human obesity. This low dietary fat oxidation makes subjects prone to weight gain.

Figure 8. Cumulative oxidation (mean ± standard deviation) of dietary fat as a percentage of intake, over time after ingestion, as calculated from tracer recovery in urine produced at two-hour intervals (From reference (32)).

REFERENCES

 

  1. Kleiber M. The fire of life; an introduction to animal energetics. New York,: Wiley.
  2. Brouwer E. On simple formulae for calculating the heat expenditure and the quantities of carbohydrate and fat oxidized in metabolism of men and animals, from gaseous exchange (Oxygen intake and carbonic acid output) and urine-N. Acta Physiol Pharmacol Neerl. 1957;6:795-802.
  3. Webb P, Saris WH, Schoffelen PF, Van Ingen Schenau GJ, Ten Hoor F. The work of walking: a calorimetric study. Med Sci Sports Exerc. 1988;20(4):331-337.
  4. Speakman JR. The history and theory of the doubly labeled water technique. Am J Clin Nutr. 1998;68(4):932S-938S.
  5. Westerterp KR, Verboeket-van de Venne WP, Bouten CV, de Graaf C, van het Hof KH, Weststrate JA. Energy expenditure and physical activity in subjects consuming full-or reduced-fat products as part of their normal diet. Br J Nutr. 1996;76(6):785-795.
  6. Adriaens MP, Schoffelen PF, Westerterp KR. Intra-individual variation of basal metabolic rate and the influence of daily habitual physical activity before testing. Br J Nutr. 2003;90(2):419-423.
  7. Tappy L. Thermic effect of food and sympathetic nervous system activity in humans. Reprod Nutr Dev. 1996;36(4):391-397.
  8. Westerterp KR. Diet induced thermogenesis. Nutr Metab (Lond). 2004;1(1):5.
  9. Speakman JR, Westerterp KR. Associations between energy demands, physical activity, and body composition in adult humans between 18 and 96 y of age. Am J Clin Nutr. 2010;92(4):826-834.
  10. Human energy requirements: report of a joint FAO/ WHO/UNU Expert Consultation. Food Nutr Bull. 2005;26(1):166.
  11. Camps SG, Verhoef SP, Westerterp KR. Physical activity and weight loss are independent predictors of improved insulin sensitivity following energy restriction. Obesity (Silver Spring). 2016;24(2):291-296.
  12. Sabounchi NS, Rahmandad H, Ammerman A. Best-fitting prediction equations for basal metabolic rate: informing obesity interventions in diverse populations. Int J Obes (Lond). 2013;37(10):1364-1370.
  13. Westerterp KR. Reliable assessment of physical activity in disease: an update on activity monitors. Curr Opin Clin Nutr Metab Care. 2014;17(5):401-406.
  14. Ekelund U, Aman J, Yngve A, Renman C, Westerterp K, Sjostrom M. Physical activity but not energy expenditure is reduced in obese adolescents: a case-control study. Am J Clin Nutr. 2002;76(5):935-941.
  15. Westerterp KR, Donkers JH, Fredrix EW, Boekhoudt P. Energy intake, physical activity and body weight: a simulation model. Br J Nutr. 1995;73(3):337-347.
  16. Westerterp KR. Physical activity, food intake, and body weight regulation: insights from doubly labeled water studies. Nutr Rev. 2010;68(3):148-154.
  17. Camps SG, Verhoef SP, Westerterp KR. Weight loss, weight maintenance, and adaptive thermogenesis. Am J Clin Nutr. 2013;97(5):990-994.
  18. Camps SG, Verhoef SP, Westerterp KR. Weight loss-induced reduction in physical activity recovers during weight maintenance. Am J Clin Nutr. 2013;98(4):917-923.
  19. Taylor HL, Keys A. Adaptation to caloric restriction. Science. 1950;112(2904):215-218.
  20. Joosen AM, Gielen M, Vlietinck R, Westerterp KR. Genetic analysis of physical activity in twins. Am J Clin Nutr. 2005;82(6):1253-1259.
  21. Westerterp KR, Plasqui G. Physical activity and human energy expenditure. Curr Opin Clin Nutr Metab Care. 2004;7(6):607-613.
  22. Westerterp KR, Meijer GA, Janssen EM, Saris WH, Ten Hoor F. Long-term effect of physical activity on energy balance and body composition. Br J Nutr. 1992;68(1):21-30.
  23. Valenti G, Bonomi AG, Westerterp KR. Multicomponent Fitness Training Improves Walking Economy in Older Adults. Med Sci Sports Exerc. 2016;48(7):1365-1370.
  24. Thurber C, Dugas LR, Ocobock C, Carlson B, Speakman JR, Pontzer H. Extreme events reveal an alimentary limit on sustained maximal human energy expenditure. Sci Adv. 2019;5(6):eaaw0341.
  25. Edholm OG, Fletcher JG, Widdowson EM, McCance RA. The energy expenditure and food intake of individual men. Br J Nutr. 1955;9(3):286-300.
  26. Westerterp KR. Limits to sustainable human metabolic rate. J Exp Biol. 2001;204(Pt 18):3183-3187.
  27. Schrauwen P, Lichtenbelt WD, Saris WH, Westerterp KR. Fat balance in obese subjects: role of glycogen stores. Am J Physiol. 1998;274(6):E1027-1033.
  28. Verboeket-van de Venne WP, Westerterp KR. Influence of the feeding frequency on nutrient utilization in man: consequences for energy metabolism. Eur J Clin Nutr. 1991;45(3):161-169.
  29. Westerterp-Plantenga MS, MJ IJ, Wijckmans-Duijsens NE. The role of macronutrient selection in determining patterns of food intake in obese and non-obese women. Eur J Clin Nutr. 1996;50(9):580-591.
  30. Almoosawi S, Prynne CJ, Hardy R, Stephen AM. Time-of-day and nutrient composition of eating occasions: prospective association with the metabolic syndrome in the 1946 British birth cohort. Int J Obes (Lond). 2013;37(5):725-731.
  31. Schrauwen P, van Marken Lichtenbelt WD, Saris WH, Westerterp KR. Changes in fat oxidation in response to a high-fat diet. Am J Clin Nutr. 1997;66(2):276-282.
  32. Westerterp KR, Smeets A, Lejeune MP, Wouters-Adriaens MP, Westerterp-Plantenga MS. Dietary fat oxidation as a function of body fat. Am J Clin Nutr. 2008;87(1):132-135.

An Overview of Glucocorticoid-Induced Osteoporosis

ABSTRACT

 

Glucocorticoid (GC)-induced osteoporosis (GCOP) is the most common cause of iatrogenic osteoporosis (OP). Fractures may occur in 30-50% of patients on chronic GC therapy. Most of the epidemiological data associating fracture risk with GC therapy are from the use of oral GCs. The process of bone remodeling is complex, regulated by an intricate network of local and systemic factors. With prolonged GC administration, cortical bone becomes increasingly affected and long bones show increased fragility. As some patients on a low GC dose show bone loss at a much higher rate than others on a higher GC dose, genetics may play a role in determining this difference. Any patient that is treated with long-term GCs should be suspected as suffering from GCOP. Laboratory evaluation for GCOP should include total blood cell count, markers of renal and liver function, serum electrophoresis, serum and 24-hr urine calcium, serum levels of 25-hydroxyvitamin D, alkaline phosphatase, thyroid-stimulating hormone and parathyroid hormone, estradiol in women and total and free testosterone in men. Changes in BMD early on during GC therapy can be detected by dual-energy X-ray absorptiometry (DXA). In patients under GC treatment fractures tend to occur at BMD values that are lower than the conventional threshold T-score of -2.5. Recently simple adjustments for the calculated fracture risk have been presented that take into account glucocorticoid dosage for the Fracture Risk Assessment tool (FRAX). Guidelines for the prevention and treatment of GCOP have been put forth from various authorities. Prevention of GCOP should start as soon as GCs are administered; bone loss is more rapid in the first months of therapy. Patients on GCs should receive supplementation with calcium and vitamin D. There are several antiresorptive agents available for the prevention and treatment of GCOP - bisphosphonates are the most widely used. Teriparatide and denosumab can also be therapies of choice for patients on GC treatment with or without GCOP.

 

INTRODUCTION

 

Glucocorticoid (GC)-induced osteoporosis (GCOP) is often the result of secondary osteoporosis (OP) (1). It is the most common cause of iatrogenic OP; adults aged 20 to 45 years are mainly affected (1-3). Important bone loss may occur with or without other manifestations and its severity is dependent on both the dose and duration of GC treatment (4). From a retrospective study conducted in the United Kingdom the prevalence of chronic use of oral GCs in the general population was shown to be 0.5%; the prevalence was higher in women over 55 years (1.7%) and as high as 2.5% in subjects older than 70 years (5, 6); more recently experts argued that approximately 2% of the population receives long-term GC treatment (7). It is of practical interest to note that only 4%-14% of patients taking oral steroids were receiving treatment for prevention of osteoporosis (mainly by rheumatologists), indicating that GCOP is often underestimated and left untreated (5, 8).

 

EPIDEMIOLOGY

 

The association between glucocorticoid (GC) excess and osteoporosis was first described nearly 80 years ago, but its importance in clinical practice has only recently been recognized (9). Although it shares some similarities with postmenopausal osteoporosis, glucocorticoid-induced osteoporosis (GCOP) has distinct characteristics, including the rapidity of bone loss early after initiation of therapy, the accompanying increase in fracture risk during this time, and the combination of suppressed bone formation and increased bone resorption during the early phase of therapy (10).

 

Although awareness of GCOP amongst health-care professionals has increased over recent years, several studies indicate that its management remains suboptimal (11, 12). Although increased rates of diagnosis and treatment have been reported, possibly as a result of national guidelines, but overall these rates remain low (12, 13).

 

There is clear epidemiological association between GC therapy and fracture risk (14-16). Oral GC therapy is prescribed in up to 2.5% of the elderly population (aged 70-79 years) for a wide range of medical disorders (17). Fractures may occur in 30-50% of patients on chronic GC therapy (18). The vertebrae and femoral neck of the hip are specifically involved (19), whereas risk at the forearm (predominantly consisting of cortical bone), is not increased, confirming that GCs affect predominantly cancellous bone (15). Vertebral fractures associated with GC therapy may be asymptomatic (20). When assessed by X-ray-based morphometric measurements of vertebral bodies, more than 1/3 of postmenopausal women on chronic (> 6 months) oral GC treatment have sustained at least one vertebral fracture (20).

 

Along with the demonstration that fractures can occur early in the course of GC therapy, fracture incidence is also related to the dose and duration of GC exposure (16).

 

Doses as low as 2.5 mg of prednisone equivalents per day can be a risk factor for fracture, but the risk is clearly greater with higher doses. Chronic use is also associated with greater fracture risk (1, 16). When daily amounts of prednisone - or its equivalent - exceed 10 mg on a continuous basis and duration of therapy is greater than 90 days, the risk of fractures at the hip and vertebral sites is increased by 7- and 17-fold respectively (16). The risk of fractures declines after discontinuation of GCs although the recovery of the lost bone is gradual and often incomplete (1, 16).

 

Most of the epidemiological data associating fracture risk with GC therapy, come from the use of oral GCs. There is less data about risk associated with inhaled GCs (21-25); from the data available it can be extrapolated that a small but persistent and clinically significant growth retardation may be expected in children receiving inhaled GCs (26). It is also important to bear in mind that the underlying disorder for which inhaled or systemic GCs is used may also be a cause of bone loss (27). The systemic release of local bone-resorbing cytokines in some of these disorders could stimulate bone loss (28, 29). In addition, there are also local factors to consider. In inflammatory bowel disease, bone loss may be due, in part, to malabsorption of vitamin D, calcium, and other nutrients (28). In chronic lung disease, hypoxia, acidosis, reduced physical activity, and smoking may all contribute to bone loss, independently of the use of inhaled GCs (14, 25, 30, 31).

 

SECONDARY CAUSES/RISK FACTORS OF BONE LOSS

 

Factors, such as advancing age, race, sex, menopausal status, family history of OP and fractures, and secondary causes of OP, such as hyperthyroidism, hyperparathyroidism, Cushing’s syndrome, hypogonadism, diabetes (particularly type 1), renal failure, inflammatory bowel disease, and rheumatoid arthritis can add to the effects of GCOP (14, 32-36). Some of the risk factors for GCOP are common to other forms of OP and can be modified; these include: low calcium and high sodium intake (37), high caffeine intake (when calcium intake is low) (38), tobacco and alcohol use, decreased physical activity, immobilization, and a number of medications (32, 39, 40). Medications/treatments that are administered concomitantly with GCs (such as methotrexate, cyclosporine, heparin, medroxyprogesterone acetate, gonadotropin releasing hormone (GnRH) analogs, levothyroxine, anticonvulsants, or radiotherapy) may add to the disease burden of GCOP.

 

The emerging use of aromatase inhibitors (41), androgen-deprivation therapy in men with prostate cancer (42), and the growing field of bariatric surgery (43) have emerged as novel and important etiologies of secondary osteoporosis.

 

Patients with classical congenital adrenal hyperplasia (CAH)  can be over-treated with GC and show loss of bone mineral density (BMD) (44). The iatrogenic suppression of adrenal androgens production in women with CAH is associated with increased risk for bone loss (45). Young adult men on GCs apparently show more rapid bone loss compared to older men or postmenopausal or premenopausal women. Of note, men are more susceptible to depression-associated bone loss, which may be in part, GC-mediated (46). Postmenopausal women receiving GCs show higher fracture risk compared to premenopausal women that is attributed to lower bone mass when starting GC therapy) (47, 48). Patients with sarcoidosis and those taking steroids to prevent rejection of grafts after heart or kidney transplant, are also more likely to experience rapid bone loss (49-51).

 

CELLULAR AND MOLECULAR MECHANISMS OF GCOP

 

The process of bone remodeling is complex, regulated by an intricate network of local and systemic factors. Although normal bone needs endogenous GCs for its development (for osteoblast differentiation in particular, via inhibition of mesenchymal stem-cell differentiation to adipocytes) (52-54), GCs, at least in mice models, exert negative effects on bone maintenance in old age (by lowering survival of osteoblasts and osteocytes and limiting angiogenesis) (52). Quiescent bone is covered by osteoblasts and osteoclasts. In response to bone-resorbing stimuli, osteoclastic migration and bone resorption are activated. Osteoclasts remove both the organic matrix and the mineral component of the bone, producing a pit. This bone remodeling cycle takes place under a canopy of osteoprogenitor cells (55). In the formation phase, osteoblasts deposit osteoid in the pit, which is then mineralized. In normal bone there is – apparently – no appreciable effect of GCs on osteoclasts (52). Quiescence is restored at completion of the cycle (56). GCs can influence bone remodeling in a number of ways and at any stage of the remodeling cycle (Figure 1). We have to note that regarding animal studies of GCOP experts point to the heterogeneity of used models and the need for their standardization (57).

 

Figure 1. Overview of the mechanisms of glucocorticoid-induced osteoporosis (GCOP). Osteoporosis results from an imbalance between osteoblast and osteoclast activity. BMP-2: bone morphogenic protein-2; Cbfa1: core binding factor a1; Bcl-2: B-cell leukemia/lymphoma-2 apoptosis regulator; Bax: BCL-2-associated X protein; IGF-I: insulin-like growth factor-I; IGFBP: IGF binding protein; IGFBP-rPs: IGFBP-related proteins; HGF: hepatocyte growth factor; RANKL: receptor activator of the nuclear factor-κB ligand; CSF-1: colony-stimulating factor-1; OPG: osteoprotegerin; PGE2: Prostaglandin E 2; PGHS-2 prostaglandin synthase-2

Bone Histomorphometry Under GCs

 

Trabecular bones and the cortical rim of vertebral bodies are more susceptible to the effects GCs compared to the cortical component of long bones (radius, humerus) (58-62). Under GC treatment, lumbar bone shows significantly greater bone loss compared to distal radius. Bone loss is also observed in the proximal femur (particularly at Ward’s triangle, an area rich in trabecular bone) (63, 64). Although bone remodeling is initially turned on with higher bone resorption, over time, resorption parameters fall and bone becomes quiescent (65, 66). Thus, with prolonged GC administration, cortical bone becomes increasingly affected and long bones show increased fragility.

 

Bone biopsies of patients on GC therapy for longer than 12 months show increased bone resorption, a decline in all aspects of bone formation, and decreased trabecular volume. Histomorphometric studies on subjects with GCOP show increased osteoclasts and bone-resorbing sites; bone loss is higher in the metaphyses compared to the diaphyses (67-69). A specific feature of GCOP is the decrease in canopy coverage of bone remodeling sites (52, 55). GCOP differs from post-menopausal OP in terms of microanatomical appearance; in GCOP the number of trabeculae and their surface area are relatively preserved, and individual plates are very thin (trabecular attenuation), although still connected, whereas in post-menopausal OP, trabecular width is relatively preserved but the lamellae are perforated by resorption, with a loss of trabecular surface and continuity (70). Such changes may lead to lower mechanical strength of bone. The particular histology of GCOP may have important implications for pharmacologic intervention: the preservation of thinned trabeculae in GCOP may provide the foundation for new bone apposition. With excess GCs, osteoclasts, over time, preferentially deepen their resorption pits than migrate to new resorption sites (52).

 

Glucocorticoid Receptors (GRs) and Bone

 

There is still no consensus on whether genomic or non-genomic actions of GCs are the major players in GCOP (71). Genomic actions result from the binding of GCs steroids to specific cytoplasmic receptors that belong to the nuclear receptor superfamily. The GC-GR complex can either activate or repress the expression of target genes. While activation requires binding of a dimerized receptor to GC-responsive elements (GREs) in the promoter region of target genes, repression is mainly mediated by interaction between receptor monomers and transcription factors (72). GC-induced osteoblast apoptosis does not require GR dimerization (52). Translation of GR mRNAs produces two GR isoforms; GRα, which is transcriptionally active and GRβ, which can heterodimerize with GRα inhibiting its transcriptional activity (73). In humans, normal osteoblasts, and specific osteoblastic cell lines show GRα expression, whereas mature osteoclasts show no GRα expression. Osteoclasts, in contrast, predominantly show GRβ expression. Osteoblasts and osteoclasts also express mineralocorticoid receptors (MRs) that bind to cortisol and form heterodimers with both GRα and GRβ (74). IL-6, in human osteoblasts, acts as an autocrine positive modulator that upregulates the number of GRs (75, 76). Cortisol, even at physiologic concentrations, modulates negatively the secretion of IL-11, a cytokine that decreases GR expression (77). Consequently, this interplay of cytokines through autocrine/paracrine loops may modulate bone sensitivity to GCs (78).

 

GCs and Osteoblast Activity

 

In response to pharmacologic doses of GCs, osteocytes trigger the protective process of autophagy; with excessive doses of GCs autophagy leads to apoptosis (79). GCs increase the apoptosis of osteoblasts and mature osteocytes via activation of caspase 3 (1, 80-83). Osteoblast/osteocyte apoptosis may involve decreased expression of the pro-survival factor BclXL and increased expression of the proapoptotic factors Bim and Bak (through induction of the leucine zipper E4bp4) (52, 84). Apoptosis is also assisted by GC-induced excess reactive oxygen species (ROS) production and inhibition of Akt, leading to suppression of the Wnt/β-catenin pathway, which is necessary for osteoblastogenesis as well as for cell survival (52, 85). Studies on the proaptototic effect of GCs on osteoblasts/osteocytes, indicate that it may be mediated by the process of endoplasmic reticulum stress (86). Furthermore, GCs reduce osteoblast proliferation and differentiation (62), possibly as a result of GC-induced repression of bone morphogenic protein-2 (BMP-2) and expression of core binding factor a1 (Cbfa1) (84). GCs also modify the expression of osteoblast specific genes, such as osteocalcin. Osteocalcin expression during the development of bone is tightly regulated by GCs, and multiple GREs have been identified on the human and rat osteocalcin promoter region (87, 88). The osteocalcin gene also contains several activator protein-1 (AP-1) sites that apparently contribute to the basal activity of the promoter. Therefore, repression of osteocalcin promoter activity by GCs may also involve interaction between GR and components of the AP-1 complex, independently of DNA binding, as it has been postulated for the collagenase promoter (89, 90).

 

The Wnt signaling pathway is important for osteoblast differentiation and function, bone development and level of peak bone mass (91). Mechanical loading results in increased bone mass in animals that carry activating mutations of Lrp5 (coding for a Wnt coreceptor)(91). Wnt signaling may be implicated in the osseous response to mechanical loading (91) and the observed inhibition of skeletal growth by GCs may be mediated by effects on Wnt signaling (92)by enhancing Dickkopf 1 (Dkk1) expression (which is a Wnt antagonist) and Sost (sclerostin, which is a disruptor of the Wnt-induced Fz-Lrp5/6 complex leading to β-catenin ubiquitination) (52, 62, 93). Interestingly, both short- and long-term GC administration decreases Dkk1 expression in humans whereas only long-term GC administration decreases Sost expression; Wnt signaling involvement in GCOP appears to be time-dependent (52). The inhibition of Wnt signaling is also involved in GC-induced adipocyte differentiation (52).

 

GCs are required for the differentiation of mesenchymal stem cells to bone cells; they can also promote an osteoblastic phenotype (by inhibiting collagenases (MMPs) and reducing collagen type 1 breakdown) (94-96). Impaired osteoblastogenesis by excess GCs involves the reduction in expression of microRNAs (endogenous RNAs of 18-25 nucleotides each that interact with mRNA to alter protein expression) (97), such as miR-29a/miR-34a-5p and reductions in the mRNA expression of Dkk1/receptor activator of the nuclear factor-κB ligand (RANKL) (98).

           

GCs and Osteoclast Activity

 

Compared to effects of GCs on osteoblasts, the effects of GCs on osteoclasts are less known as osteoclast isolation from bone is technically difficult and bone marrow cultures, hematopoietic cell lines and cells derived from giant-cell tumors (used as model systems to study osteoclast differentiation and activity) have produced varying results. GCs stimulate bone resorption (99-101). It has been shown that GCs stimulate osteoclastogenesis through their capacity to bind to the bone surface by altering the expression of N-acetylglucosamine and N-acetylgalactosamine (85, 102). Osteocyte apoptosis, induced by GCs, reduces osteoprotegerin (OPG, the decoy RANKL ligand) (52). GCs may decrease apoptosis and prolong the lifespan of mature osteoclasts (52, 62) but cannot affect directly their bone-resorbing activity, since these cells apparently lack functional GRs (103). GCs suppress calpain 6 (Capn 6) which is enmeshed in β-integrin (a mediator of osteocyte interaction with the osseous matrix) and expression of microtubules’ acetylation/stability within the bone cells cytoskeleton (52). Higher expression of the GR gene in subjects with lower BMD may lead to higher sensitivity of their monocytes/macrophages to GCs to differentiate into osteoclasts (104). Cytokines are also implicated in these actions (see next section on regulation of local bone factors by GCs) (105).

 

GCs and Local Bone Factors (Cytokines, Growth Factors, Prostanoids and Kinases)

 

CYTOKINES

 

Interleukin-1 (IL-1) and -6 (IL-6) induce bone resorption and inhibit bone formation. GCs partially inhibit the production of IL-1 and IL-6 and inhibit the bone resorbing activity of these cytokines (GC therapy could paradoxically protect osseous tissue from IL-induced bone resorption) (106-109). Transforming growth factor beta 1 (TGF-b, which inhibits IL-1-induced bone resorption and stimulates osteoblast activity) is decreased by GCs. (110). Lower levels of TGF-bmay increase the susceptibility of bone to the resorbing effects of IL-1. IL-1 suppression also inhibits the generation of nitric oxide, which modulates osteoclast activity (111). Excess GCs inhibit the expression of IL-11 on osteoblasts (and hinder this cytokine’s effect on their differentiation) independently of GR dimerization (52). GCs interfere with the RANKL-OPG axis. RANKL (which is expressed at high levels in pre-osteoblast/stromal cells) induces osteoclast differentiation in the presence of colony-stimulating factor-1 (CSF-1) by binding to the receptor activator of the nuclear factor-κB (RANK; a member of the TNF family on the surface of octeoclasts(108). OPG is also produced by osteoblasts and is found on their surface. OPG acts as a decoy receptor of RANKL: it binds RANKL and prevents it from binding its osteoclast receptor, therefore inhibiting osteoclast differentiation. GCs enhance RANKL and CSF-1 expression (78), and lower OPG expression in human osteoblasts cells in vitro (112). Serum OPG concentrations are significantly reduced in patients undergoing systemic GC therapy (113). This decrease in OPG is more marked than the GC-induced increase in RANKL (via suppression of miR-17/20a, which targets Rankl) (52), leading to an increased RANKL/OPG ratio that may mediate GC-induced bone resorption (114).

 

GROWTH FACTORS

 

Insulin-like growth factors (IGFs) have an anabolic effect on bone cells that affect IGF-I and IGF-II receptors. IGF-I and IGF-II are weak mitogens (they increase the replication of osteoblasts), they increase type I collagen synthesis and matrix apposition rates and decrease collagenase-3 (metalloproteinase-13) expression by osteoblasts (115, 116). Synthesis of IGF-I in osteoblasts is decreased by GCs via increased expression of the CAAT/enhancer binding protein (C/EBP) β and δ (transcription factors that bind to the IGF-I promoter and halt its transcription) (117). GCs inhibit IGF-II receptor expression in osteoblasts (while they have no effect on IGF-I receptor expression)(118, 119). Since the IGF-II receptor functions as an IGF-binding protein (IGFBP) its inhibition by GCs may result in higher levels of available growth factors although it may also lead to faster degradation of IGF-II. The activity of IGF-I and -II is regulated by at least six IGFBPs that are expressed by osteoblasts (120, 121). IGFBPs in skeletal cells are considered to be local reservoirs and modulators of IGFs. GCs decrease the expression of IGFBP-3, -4, and -5 in osteoblasts (122, 123). IGFBP-5 stimulates bone cell growth (and enhances the effects of IGF-I); its reduction in the bone microenvironment may be relevant to the inhibitory actions of GCs on bone formation and the process of GCOP (124). GCs also increase the synthesis of IGFBP-related proteins (IGFBP-rPs; a family of peptides related to IGFBPs that bind IGFs and are involved in cell growth) (125). Chondrocytes are involved in fracture healing and in OP this process is delayed. Among others, GCs inhibit the activation of GH and IGF-I receptors in chondrocytes and reduce IGF-I and GH receptor expression in these cells (126).

 

Bone cells express transforming growth factor-b (TGF-b) -1, -2, and -3 genes (127). TGF-b stimulates bone collagen synthesis and matrix apposition rates, modifies bone cell replication, stimulates growth and proliferation of osteoblasts but inhibits their differentiation and the expression of osteocalcin (128, 129). TGF-b1 expression in osteoblasts is not modified by GCs. GCs, instead, induce activation of the latent form of TGF-b1 by increasing the levels of bone proteases (130, 131). Two signal-transducing TGF-b receptors are expressed in osteoblasts. GCs shift the binding of TGF-b from these receptors to betaglycan (by increasing the synthesis of this proteoglycan) and oppose the effects of TGF-b osteoblastic cell replication (130).                     

 Hepatocyte growth factor (HGF) is produced by both osteoblasts and osteoclasts. HGF is a potent stimulator of osteoblastic function and a potent suppressor of bone resorption in isolated rat osteoclasts (132). Osteoclast-produced HGF (in an autocrine fashion), may lead to changes in osteoclast shape and stimulate osteoclast migration and chemotaxis, while (in a paracrine fashion) may lead osteoblasts to enter the cell cycle, via DNA synthesis stimulation (132, 133). GCs inhibit the release of HGF in vitro, which suggests that the inhibitory effects on bone resorption of GCs may be in part mediated via regulation of osteoblast-produced HGF (134, 135).

 

Platelet-derived growth factor (PDGF) is a mitogen of bone cells (136). PDGF-A and PDGF–B are expressed in a limited fashion in osteoblasts, and neither the synthesis nor the binding of PDGF appear to be modified by GCs. Specific PDGF-A/B binding proteins are lacking, although SPARC (secreted protein acid rich in cysteine) and osteonectin (a protein abundant in bone matrix) bind and prevent the biologic actions of PDGF-B (137). Since GCs enhance osteonectin expression in osteoblastic cells they may also decrease the activity of PDGF-B in bone (138).

 

PROSTANOIDS

 

Prostaglandins (PGs) are produced by bone cells and affect both bone formation and resorption. PGs (and PGE2 in particular) stimulate bone collagen and non-collagen protein synthesis (139-141). PGs inhibit directly the activity of isolated osteoclasts and increase bone resorption in organ cultures, (probably by promoting osteoclastogenesis) (142). GC-induced inhibition of collagen synthesis in bone, down-regulation of c-fos oncogene expression and reduced osteoblast proliferation are all reversed by exogenous PGE2in vitro, suggesting an important pathogenic role for this PG in GCOP (143-147). GCs interfere with the production of PGs in bone (especially of PGE2) via the decreased expression of cyclooxygenases (the enzymes that convert arachidonic acid into PGs) (148, 149). Osteoblasts express two cyclooxygenases: constitutive prostaglandin synthase-1 (PGHS-1) and inducible prostaglandin synthase-2 (PGHS-2). Apparently, GC-inhibited PG-production in bone is mediated through a decrease in agonist-induced PGHS-2 expression.

 

KINASES

 

GCs modulate intracellular kinases (ERKs, MAPK/JNK and Pyk2) with a proapoptotic effect on the osteoblastic lineage  (150)

 

EXTRASKELETAL MECHANISMS OF GCOP

 

Effects of GCs on Calcium Absorption and Excretion

 

Although there is no consensus regarding the effect of GCs on calcium absorption, they mainly impair intestinal calcium absorption (151-158). GCs have no effect on the intestinal brush border membrane vesicles (159), but decrease synthesis of calcium binding protein and deplete mitochondrial ATP (160). Patients treated with GCs show increased renal calcium loss occasionally leading to the development of secondary hyperparathyroidism (161). In normal subjects receiving GCs the elevation of fasting urinary calcium proceeds the rise in immunoreactive parathyroid hormone (iPTH) (162). In patients on long-term GC therapy, hypercalciuria is most likely due to increased skeletal mobilization of calcium and decreased renal tubular reabsorption that occurs in spite of elevated PTH levels. The GC-induced decrease in bone formation lowers calcium uptake by newly formed bone and elevates the filtered load of calcium. High dietary sodium intake increases renal loss of calcium whereas sodium restriction and thiazide diuretics lower its renal loss (163).

 

Effects of GCs on the Excretion of Phosphorus

 

GCs, acting directly on the kidney and indirectly, via induction of secondary hyperparathyroidism, lower tubular reabsorption of phosphate, leading to phosphaturia (164, 165). Furthermore, GCs increase the amiloride-sensitive Na+/H+ exchange activity in the renal proximal tubule brush border vesicles and decrease the Na+ gradient-dependent phosphate uptake, resulting in  increased acid secretion and phosphaturia (166).

 

GC Effects on Parathyroid Hormone (PTH)

 

A direct stimulatory effect of GCs on PTH secretion may also exist (164, 167, 168). GCs induce a negative calcium balance that leads to secondary hyperparathyroidism; in patients receiving GCs iPTH is increased, that can be suppressed with exogenous calcium and vitamin D (168, 169). Chronic GC administration is accompanied by altered secretory dynamics of PTH; more particularly, it reduces its tonic secretion and increases its pulses (170). However, elevated iPTH levels can also be suppressed following calcium infusion, suggesting that its  elevation is more likely to be secondary to a negative calcium balance caused by GCs, rather than to direct stimulation of PTH secretion (171).

 

Effects of GCs on Vitamin D Metabolism

 

Low, normal, or increased circulating levels of 1,25-dihydroxyvitamin D (1,25-(OH)2D) have been reported in subjects taking GCs (171-174). These differences may originate from variations in the dietary intake and absorption of vitamin D and in exposure to sunlight. The rate of synthesis and clearance of 1,25-(OH)2D is normal in subjects receiving GCs (175). Although the administration in humans of 1,25-(OH)2D improves calcium transport, it does not normalize it (176).

 

GC Effects on Sex Hormones

 

GCs inhibit the secretion of gonadotropins and also show direct effects on the gonads and the target tissues of gonadal steroids. In rats, GCs reduce the action of follicle-stimulating hormone (FSH) on granulosa cells and inhibit the response of luteinizing hormone (LH) to gonadotropin-releasing hormone (GnRH) (177-179).In rats and primates, GCs also decrease GnRH secretion; furthermore, in rats, overexposure to GCs renders their pituitary insensitive to exogenously administered GnRH (180-182).In men and women given GCs the plasma concentrations of estradiol, estrone, dehydroepiandrosterone (DHEAS), androstenedione, and progesterone are decreased (183-185). High-dose GC therapy in women may lead to amenorrhea. Although the exact targets of GC inhibition of steroidogenesis in Leydig or granulosa-theca cells are not fully defined, recent studies have found a GC-responsive upstream promoter region of the cholesterol side-chain cleavage gene (186).  In postmenopausal women an additive effect of GC treatment with estrogen deficiency on bone loss is observed (187, 188).

 

GC Effects on Growth Hormone (GH)

 

GH is an important regulator of both bone formation and bone resorption. in vitro studies have shown that the GH-induced increase in bone formation is twofold: by direct interaction with GH receptors on osteoblasts, and through induction of an endocrine and autocrine/paracrine IGF-I effect (189). In contrast, in animals high endogenous GCs or exogenous exposure can inhibit linear growth and GH secretion in animals. In patients with GCOP a lower GH response to growth hormone–releasing hormone (GHRH) and a positive correlation between GH increment and osteocalcin are observed. This inhibitory effect of GCs on the secretion of GH may be dependent on an increase in somatostatin synthesis and secretion, which inhibits pituitary GH secretion. Arginine, which decreases hypothalamic somatostatin tone, normalizes the GH response to GHRH (190, 191). Bone sensitivity to GH may also reduce by GCs: an up-regulatory effect on GH receptor expression may be implicated (192).

 

GC Effects on Connective Tissue

 

Excess GCs hinder wound healing via suppression of DNA and protein synthesis in fibroblasts and impaired local macrophage recruitment (193, 194).

 

GC Effects on Muscle

 

Common side effects of GC excess include muscle weakness and loss of muscle mass. Alterations of muscle biopsies of GC-treated patients include selective atrophy of type IIa muscle fibers, relative increase in the number of type IIb fibers and decrease in the number of type I fibers (195-197). The main mechanisms implicated in GC-induced myopathy are increased protein catabolism, inhibition of glycogen synthesis, and interference with fatty acid β-oxidation (83). In fact, GCs stimulate ubiquitin-proteasome-dependent protein breakdown in skeletal muscle and regulate calcium-dependent proteolysis (198, 199).Moreover, levels of glycogen synthase, beta-hydroxyacyl-CoA dehydrogenase and citric acid synthase, are lower in muscle from GC-treated patients compared to muscle from disease-matched controls (200). A strong association between steroid myopathy and OP has been described (201).

 

INDIVIDUAL SUSCEPTIBILITY TO GCOP

 

Some patients on a low GC dose show bone loss at a much higher rate than others on a higher GC dose (202). Genetics may play a role in determining this difference. Little is known about the mechanisms of cellular sensitivity to GCs. Individual factors are also important in determining the risk of fractures when GCs are used. Polymorphisms in the GR gene have been linked to the varied degrees of susceptibility to GCs; these could explain the different rates of GC-associated fractures (97). Individuals that are heterozygous for a polymorphism at nucleotide 1,220 (resulting in an Asparagine-to-Serine change at codon 360), had increased BMI, increased blood pressure and lower spine BMD compared to control subjects (203, 204).

 

Another explanation for inter-individual variability among those exposed to GCs is related to differential activity of 11b-hydroxysteroid dehydrogenase (11b-HSD) (205). This enzyme system plays a critical role in the regulation of GCs activity (206). Two distinct 11β-HSD enzymes have been described; 11b-HSD1 (converting cortisone [E] to cortisol [F] and 11b-HSD2 (converting F to E) modulate GC and mineralocorticoid hormone action in target organs (205, 207, 208). 11β-HSD1 is widely expressed in GCs target tissues, including bone (206). The reductase activity does not show a large inter-individual variability, whereas the oxidase activity of 11b-HSD2 has a large inter-individual variability. Subjects with higher oxidase activity at bone level may be at greater risk of developing GCOP (209). Men with OP were shown to have increased endogenous GC availability, via apparent 11b-HSD1 activation (210). The activity of 11β-HSD1 and the potential to generate F from E in human osteoblasts is increased by pro-inflammatory cytokines (TNFa, IL-1b and IL-6) and by GCs themselves (211, 212). During inflammation pro-inflammatory cytokines may potentiate GC actions in bone through an “intracrine” mechanism (209, 213). An increase of 11β-SD1 activity occurs with aging, possibly providing an explanation for the enhanced GC effects in the skeleton of elderly subjects (214).

 

In the future, the characterization of factors accounting for the variability to GC-related bone loss among individuals may identify subjects at higher risk of developing GCOP and, possibly, customize treatment.

 

DIAGNOSIS OF GCOP

 

Medical History and Clinical Evaluation

 

Table 1 summarizes elements from medical history suggestive of GCOP and the modalities available for its diagnosis. Any patient that is treated with long-term (for over a month) GCs should be suspected as suffering from GCOP (215). The risk for GCOP is higher in postmenopausal women, transplant recipients, and patients with sarcoidosis (216-220). Bone loss depends on the dose, route, and duration of GC administration (218-220).

 

Table 1. Clues and Diagnostic Means for GCOP

Medical history

Sex and age

History of OP and/or trauma fractures

History of allergy, chronic inflammatory or autoimmune disease, hematologic, skin and renal disorders, transplantation

Calcium and alcohol intake, smoking, physical activity

Chronic use of anticonvulsants, heparin, immunosuppressants

Menstrual, menopausal or fertility status 

Clinical evaluation

Truncal obesity, edemas, striae, skin atrophy and ecchymoses

Myopathy (myalgias, weakness of the proximal muscles and pelvic girdle)

Assessment of temporal baldness, loss of body hair, gynecomastia, altered pubic hair pattern, decreased testicle and prostate size

Laboratory evaluation

Complete blood cell count, liver and renal function, serum electrophoresis

Serum calcium and phosphate, serum 25-OH-vitamin D, serum alkaline phosphatase, PTH

Osteocalcin, bone-specific alkaline phosphatase, procollagen type I extension propeptides)

Hydroxyproline, hydroxylysine glycosides, hydroxypyridinium cross-links, type I collagen telopeptides)

Thyroid hormone profile, total and free testosterone, estradiol, luteinizing hormone, prolactin, ferritin

Bone mineral density assessment

 

 

Lateral scan (vertebral bodies) and anteroposterior scans (spine, hip) with dual-energy X-ray absorptiometry (DXA) – Trabecular Bone Score (TBS) in lumbar spine (if available)

·                  Assessment of vertebral compression fractures with X-ray        

 

 

Cushingoid clinical features include truncal obesity, skin atrophy with increased fragility and ecchymoses, fluid retention, hyperglycemia, and symptoms of vertebral compression and myopathy. Muscle strength needs to be assessed by a trained physician or specialized physical therapist, with special attention to the testing of proximal muscle groups. A brief exposure to GCs may trigger myopathy that is not always dose-dependent, and is often difficult to differentiate from inflammatory myopathy. However, GC myopathy is characterized by creatinuria and normal muscle enzymes, including aspartate aminotransferase, creatine kinase, and aldolase (195, 201).

 

Men and women on chronic treatment with GCs often have symptoms of hypogonadism, such as decreased libido and sexual activity, and may show low rates of fertility or even infertility. In premenopausal women history taking should assess menstrual periods, since subtle changes, including less bleeding and shortened menstrual periods, may be indications of low estrogen levels. Menstrual irregularities are also common in women with endogenous GC excess.

 

Various respiratory, dermatologic, musculoskeletal, neurologic and gastrointestinal disorders are frequently treated with GCs. Signs and symptoms of such disorders need to be evaluated.

 

Laboratory Tests and Markers of Bone Turnover

 

Laboratory evaluation for GCOP should include total blood cell count, markers of renal and liver function, serum electrophoresis, serum and 24-hr urine calcium, serum levels of 25-hydroxyvitamin D, alkaline phosphatase, thyroid-stimulating hormone and parathyroid hormone, estradiol in women and total and free testosterone in men (218-221).

 

In patients receiving GCs a dose-dependent decrease in serum osteocalcin is found; this is a good indicator of the degree of inhibition of osteoblastic activity (222, 223). Other markers of bone formation, such as total and bone specific alkaline phosphatase and procollagen type I carboxy-propeptide are also lower in under GC therapy (162, 224). In subjects on GC therapy baseline levels of osteocalcin do not always correlate with subsequent bone loss (225-227). In some, but not all, studies of patients treated with GCs, markers of bone resorption (like urinary collagen N-telopeptides [NTX]) are elevated (165, 228-230). In view of such discrepancies, the measurement of serum markers of bone formation and resorption is considered to be of little clinical utility and it is not currently advocated for routine use (217).

 

Bone Mineral Density (BMD) Assessment

 

Changes in BMD early on during GC therapy can be detected by dual-energy X-ray absorptiometry (DXA) and quantitative computed tomography (QCT); classic X-ray studies are useful to detect vertebral compression fractures. Both QCT and DXA can measure cortical and trabecular bone density, however, the former is mostly used to evaluate trabecular bone density, whereas the latter is used to measure cortical and trabecular bone density (231, 232). DXA also helps estimate the risk for fractures, and provides an objective measurement to judge the efficacy of treatment (221, 233, 234). BMD measurement techniques that focus on the vertebral body and exclude the cortical bone of posterior processes, such as lateral DXA scanning, are more sensitive in detecting GCOP (61, 235). However, the selection of a BMD assessment method is influenced by the presence of vertebral deformities, osteophytes, or of calcifications in the aorta that may spuriously elevate spinal BMD values. If this is the case, lateral views of the vertebral bodies are considerably less precise than antero-posterior scans, and therefore less appropriate for following up changes in bone mass. When marked osteophytosis or scoliosis of the spine is seen, proximal femoral densitometry (in the femoral neck) should be chosen (63). The trabecular bone score (TBS), which is a DXA analytical tool that hones on lumber vertebral microarchitecture, may be useful in assessing GCOP (236, 237).

 

In patients under glucocorticoid treatment fractures tend to occur at BMD values that are lower than the conventional threshold T-score of -2.5 (238, 239). A T-score threshold value of – 1.5 SD is usually the cutoff for GCOP in Europe (5), whereas the American College of Rheumatology (ACR) has defined the T-score cut off to – 1.0 SD to separate “normal” from “not normal” BMD (220). Furthermore, the ACR recommends BMD baseline measurements at the lumbar spine and/or hip before starting any GC treatment longer than 6 months (220). At 6 month intervals from the baseline assessment, or at 12 month intervals, if the patient is receiving therapy to prevent bone loss, follow-up measurements should be done (240, 241). For the United States in particular, Medicare reimburses BMD evaluation for patients on chronic treatment with GC doses higher than 7.5 mg/day of prednisolone equivalent (242).

 

The Fracture Risk Assessment tool (FRAX) estimates the 10-year risk for osteoporotic fractures at the hip and other sites. FRAX is criticized since it uses hip BMD, whereas vertebral fractures may be more common than hip fractures in subjects receiving GCs (243). Recently simple adjustments for the calculated fracture risk have been presented that take into account glucocorticoid dosage (244) (Figure 2). Use of FRAX is currently advised to stratify GC-treated patients in low, moderate and high fracture risk categories (245, 246).

Figure 2. Fracture risk stratification and FRAX fracture risk corrections according to glucocorticoid usage (modified from (245); # fracture; T: T-score; postmenop: postmenopausal; corr: corrected; * x 1.15 if glucocorticoid dose > equivalent to 7.5 mg prednisone/day; **x 1.20 if glucocorticoid dose > equivalent to 7.5 mg prednisone/day; ***for > 6months; Z: Z-score; GC Rx: glucocorticoid therapy

PREVENTION AND TREATMENT OF GCOP

 

Guidelines for the prevention and treatment of GCOP have been put forth from the ACR in 2001, in 2010 (220, 247)and more recently in 2017 (245), the UK Consensus group in Management of GCOP (240) and the Belgian Bone Club (248), among others.

 

General Preventive Strategies

 

As soon as GCs are administered prevention of GCOP should start; bone loss is more rapid in the first months of therapy. The minimal effective GC dose should be used. Although alternate day therapy seems attractive it has not been proven to hasten bone loss in adults (202, 249); the persistent depression of adrenal androgen production may be the culprit (250).

 

The concept of “safe dose” for the treatment with oral GCs is controversial (66). More particularly, prednisone given at low doses (2.5-9 mg/d) may affect BMD whereas lower doses (1-4 mg/d) were reported to have very little or no skeletal effect (251, 252). Intravenous high-dose (up to 1 g) methylprednisolone administration is not onerous to bone (253) but even a single oral dose of 2.5 mg of prednisone has an almost immediate negative effect on osteocalcin secretion (254). Alternate-day GC administration may prevent growth retardation in children but not bone loss (202, 249). Thus, despite the ambiguity of the literature, an equivalent dose equal to or higher than 2.5 mg of prednisone per day for a month seems a sensible threshold to give protection against GCOP.

 

Inhaled GCs may be better than oral or systemic GCs vis-à-vis bone health, but still have their osseous tissue complications (22, 255). Newer inhaled GCs (such as budesonide), seem to have less adverse effects on the bone, as indicated by measurements in bone markers (256, 257). Dosing of the inhaled GC is important; beclomethasone dipropionate or budesonide given at low doses for more than one year did not affect spine BMD in asthmatic subjects (257). However, patients treated with high doses of inhaled budesonide or beclomethasone (1.5 mg/day, for at least 12 months) and without prior oral GC treatment for more than 1 month, had a significant decrease in BMD and bone formation markers, with no changes in bone resorption markers (258). In another study, inhaled GCs in adults with chronic lung disease were not associated with increased fracture risk (and more in detail no dose-response curve was verified) (259). Moreover, in children treated with beclomethasone for bronchial asthma, analysis after adjustment for the severity of the underlying disease did not show any association between inhaled GCs and fracture risk (260). Thus, in children, other factors, such as excess body weight, low muscle mass and limited exercise capacity may predispose to low BMD (261).

 

Another factor that should be noted is the change in lifestyle for the prevention of GCOP. Diet should be rich in calcium and protein (262). Alcohol and sodium intake should be reduced (to 1-2 units of alcohol/day (245)), smoking should be stopped and a regular exercise program should be followed (37). Subjects on GCs may benefit if they are protected from falls (217, 263).

 

An important, yet often neglected by most prescribing physicians (93), facet of GC-treatment is the need for proper patient information and acknowledgement regarding untoward effects. A signed relevant patient acknowledgement form should be included in medical charts/files to avoid malpractice litigation (243). 

 

Therapeutic Options

 

Therapy for GCOP aims to prevent and minimize bone loss, to increase BMD and, at least partially, to reverse the effects of GC excess. Some therapies should be continued for as long as GC treatment is pursued. The usual primary outcome in most reported – to date - trials of GCOP-specific treatments, is the change from baseline in vertebral BMD vis-à-vis placebo or other treatments; few trials have also assessed fracture rates (264, 265). 

 CALCIUM AND VITAMIN D SUPPLEMENTATION Patients on GCs should receive supplementation with calcium and vitamin D; this is better than no supplementation or calcium alone (262). A daily dose of 1,500 mg calcium and 800 IU vitamin D (1 μg/day of α-calcidiol or 0.5 of μg/day calcitriol) effectively oppose negative calcium balance (220). A two-year randomized clinical trial demonstrated the efficacy of combined calcium and vitamin D supplementation in preventing bone loss in patients with rheumatoid arthritis treated with low doses of GCs (266). However, these encouraging findings were not replicated in a three-year follow-up study, where the same combination did not show any benefit (267). From randomized clinical trials and meta-analyses it was shown that active metabolites of vitamin D (α-calcidiol and calcitriol) are more effective than vitamin D in maintaining bone density during medium-to-high dose GC treatment (268-271). Treatment with active forms of vitamin D entails a risk of hypercalciuria and hypercalcemia, consequently periodic assessment of serum calcium and creatinine levels at the beginning of the therapy, after 2-4 weeks, and thereafter every 2-3 months is advised (272, 273). Currently - according to the ACR (245) - optimal intake for calcium is set at 1000 mg/day and at 600-800 IU/day for vitamin D.

 

Thiazide diuretics lower urinary calcium excretion. Chronic treatment with thiazides decreased the incidence of hip fracture in elderly patients, and increased BMD in the general population (274-276). This evidence suggests that, together with sodium restriction, they may be useful in opposing calcium loss and secondary hyperparathyroidism caused by chronic GC therapy. However, there are currently no studies showing long-term effect of thiazide diuretics on BMD in patients treated with GCs.

 

ANTIRESORPTIVE THERAPY

 

There are several antiresorptive agents available for the prevention and treatment of GCOP.

 

Bisphosphonates decrease the resorptive activity of osteoclasts, increase osteoclast apoptosis and decrease osteoblast and osteocyte apoptosis (277). Their efficacy in preventing and treating GCOP has been clearly shown in large randomized controlled clinical trials (278-280). Treatment with alendronate for 18 months or two years increased total body BMD, and – according to some studies - significantly decreased risk of vertebral fractures in patients taking GC (281, 282). In a one-year study of patients on GCs having undergone cardiac transplantation subjects given alendronate had lower bone loss compared to subjects on calcitriol or no other treatment (-0.7%, -1.6% and -3.2% for the lumbar spine and -1.7%, -2.1% and -6.2% for the femoral neck BMD, respectively); vertebral fracture rates were not different in the three groups though (283).  In a meta-analysis of published randomized clinical trials of patients with GCOP who were given alendronate for 6-24 months, BMD in the lumbar spine as well as in the femoral neck increased but the fracture rate was not different compared to that of patients who were given only calcium, serving as a control group (284). Similarly, a one-year study with risedronate in patients taking prednisone (7.5 mg/day for at least 6 months) showed an increase in lumbar spine and femoral neck BMD and an impressive – though prone to bias due to limited sample size -  70% decrease in the relative risk of vertebral fractures (285). Zoledronic acid, a long-acting potent bisphosphonate given intravenously (4-10 mg once or twice a year) has excellent anti-OP results (286-291) and has been assessed in GCOP. The HORIZON study lasted for one year and tested the effectiveness of 5 mg intravenous zoledronic acid (n=416) vs. risedronate (n=417) in subjects with GCOP; the former led to greater increase in lumbar bone mineral density and greater decrease in bone turnover compared to the latter (292). The study did not show differences in fracture risk most probably because of its short duration. Pyrexia (particularly in the first three days post-infusion) and worsening of rheumatoid arthritis were noted more often in the zoledronic acid group (292).  

 

Oral bisphosphonates are a first choice for anti-resorptive therapy, followed by intravenous bisphosphonates (245), the latter are a first choice in pediatric GCOP (293). Currently, alendronate po (70 mg/week), risedronate po (35 mg/week or 75 mg on two consecutive days per month) and zolendronic acid iv (5 mg once a year) are recommended to treat men and women receiving GC treatment (247); therapy is advised for at least two years (294). Oral ibandronate (150 mg once a month) given for GCOP in men and women has positive results – particularly regarding spine BMD and vertebral fractures (295).

 

In patients with rheumatoid arthritis and connective tissue diseases who are treated with the RANKL inhibitor denosumab, lumbar spine (296-298) and femoral neck (297) BMD increase. Denosumab sc (60 mg every six months) is henceforth also proposed as treatment for GCOP (245, 299); it is considered to be superior in therapeutic effect on lumbar spine BMD, total hip/femoral neck BMD and vertebral fractures’ incidence compared to bisphosphonates (300, 301). The downside of Denosumab is that its discontinuation is followed by rapid bone loss (302); some experts consider that this makes it less attractive as a treatment for GCOP (303). Denosumab can also be a therapeutic option in patients with renal insufficiency who cannot receive bisphosphonates or teriparatide (243).

 

ANABOLIC THERAPY

 

Anabolic medications enhance bone formation, therefore antagonizing the suppressive effect of GCs on osteoblasts. However, some of the information on the use of these compounds to prevent or treat GCOP comes from small studies.

 

Recombinant PTH administration (400 IU of PTH 1-34; teriparatide) to postmenopausal women on prolonged estrogen replacement, who had developed OP after chronic GC therapy, resulted in increased lumbar spine bone mass, assessed by both DXA and QCT, which was maintained after discontinuation of teriparatide (304, 305). An 18-month long randomized double-blind trial compared teriparatide vs alendronate in subjects with GCOP; the increase in lumbar BMD was higher with teriparatide (+4.6 to +8.1% vs. +2.3 to +3.6%) than for alendronate at 18 months. Better results were noted for those taking low GC doses and fewer vertebral fractures occurred with teriparatide compared to alendronate (0.6% vs 6.1%) whereas the non-vertebral fracture rate did not differ between treatment groups (306). Analogous results were noted when the trial was extended to 3 years: lumbar spine BMD increased by +11.0% for teriparatide vs +5.3% for alendronate whereas the respective femoral neck BMD change was +6.3% vs +3.4% (307). Teriparatide can be a therapy of choice (20 microg/day sc) for patients on GC treatment and/or with GCOP, following intravenous bisphosphonates on a par with denosumab as proposed in the ACR guidelines (245, 247, 308, 309). The combination of teriparatide and bisphosphonates may not have an additive effect on bone (310); it is not advised for GCOP. Nevertheless, bisphosphonates given after stopping teriparatide therapy help maintain the bone formed by teriparatide (311).

 

Sodium fluoride, in combination with either calcium and vitamin D, or cyclic etidronate, improved lumbar spine BMD and trabecular bone volume in GC-treated patients. However, no reduction in the incidence of fractures was observed. Moreover, fluoride induced bone loss at the femoral neck (312, 313). Since most of the evidence indicates that sodium fluoride does not provide architecturally competent bone, its use is currently not recommended for GCOP (220).

 

Anabolic steroids have also been tested in GCOP. Cyclic nandrolone decanoate (50 mg i.m. every three weeks for six months) increased the forearm bone density in GC treated women, 10% of which developed virilizing side effects (314). The typical negative effects of steroids on bone are not present with nandrolone because it is metabolized to dihydrotestosterone (DHT). Similarly, cyclic medroxyprogesterone acetate (200 mg i.m. every 6 weeks for one year) augmented lumbar spine BMD in treated men (315). Currently, there is no recommendation for the use of anabolic steroids for GCOP.

 

GONADAL HORMONE THERAPY

 

Sex hormone treatment should be considered whenever a patient with GC excess develops hypogonadism (278). A retrospective study in postmenopausal women taking GCs found an increased BMD in those who were taking estrogens, compared to increasing bone loss in those who were not (316). Moreover, in a randomized controlled clinical trial of postmenopausal women taking GCs for rheumatoid arthritis, a significant increase in lumbar spine BMD was observed in those receiving hormone replacement therapy (HT) compared to those receiving placebo (317). This evidence suggests the potential benefit of HT in hypoestrogenic women treated with GCs. However, a large randomized clinical trial in postmenopausal women treated with a combination of estrogen and progestin planned to last 8.5 years was interrupted after 5 years, because the overall risks exceeded the benefits of the treatment (318). In the past the ACR recommended oral contraceptives (unless contraindicated) in premenopausal women on GCs who develop oligo-amenorrhea (220) but this option is no longer included in the more recent ACR guidelines. Similarly, adult men with GC excess who develop hypogonadism benefit from testosterone replacement. In GC-treated asthmatic men with testosterone deficiency, i.m. testosterone injections increased lumbar spine but not hip BMD (319). There are no data on the potential benefit of testosterone therapy in GC- treated eugonadal men (247). However, since most studies have shown an increase in prostate size and prostate-specific antigen levels in older men on testosterone supplementation/therapy (320-323), testosterone administration should be monitored with yearly digital examinations and prostate-specific antigen measurements.

 

OTHER THERAPIES

 

In addition to different combinations of the treatments so far discussed, selective estrogen receptor modulators (SERMs) alone or conjugated estrogens/SERMs belong to the pharmaceutic armamentarium against GCOP. SERMs, have positive effects on the bone. Tamoxifen reduces in vitro some of the deleterious effects of GC on the bone (324). Raloxifene, which is currently approved by the United States’ Food and Drug Administration (FDA) for the prevention and treatment of postmenopausal OP, might be a safer alternative to HT in the treatment of GCOP that develops in postmenopausal women (246, 325), given its favorable effects on serum lipids, together with the lack of growth stimulation on endometrial and breast tissues (326-328).

 

FUTURE THERAPEUTIC OPTIONS

 

Currently, denosumab is being evaluated for pediatric GCOP (293). Other newer agents that are tentatively evaluated for the treatment of osteoporosis either inhibit osteoclast resorption or stimulate osteoblast bone forming activity. These include antibodies against RANKL (RANKL inhibitors), recombinant osteoprotegrin, inhibitors of osteoclast enzymes, integrin antagonists and agonists to LRP5 (308).

 

At the time of writing, abaloparatide (PTHrp) and romosozumab (humanized monoclonal antibody that targets sclerostin) have been cleared by the FDA for the treatment of OP in women only (8, 329, 330). One would expect the former to be a good candidate for GCOP in analogy to teriparatide. However, this therapy is not yet approved for GCOP and to the best of our knowledge there are no relevant clinical studies to support its use in GCOP (331). Furthermore, we have to bear in mind that administration of GC > 15 mg/day may attenuate the osseous effects of teriparatide, and this has also been shown with abaloparatide in rodent GCOP models (331, 332). There is an ongoing trial of romosozumab in GCOP but at present this medication has no firm indication for GCOP (313); experimental studies in rodents were encouraging (333).

 

Other promising future therapeutic options target GC therapy per se. These include the use of disease-modifying antirheumatic drugs or tumor-necrosis factor agents, which could lead to the need for lower GC dosage for autoimmune disease. Furthermore, deflazocort (a prednisone derivative) and liposomal prednisone may be less onerous to bone (334). The search continues to find selective GR agonists (SGRMs) that possess the anti-inflammatory benefits of traditional GCs without the associated adverse effects (335). The SGRMs are selective ligands of the GR, which maintain the transrepressive properties of GCs (usually associated with their beneficial anti-inflammatory effect) while they do not have their transactivating properties (usually associated with metabolic negative effects, including perhaps those on the bone). Some of these molecules may represent an alternative to traditional GCs in the chronic treatment of inflammatory disorders (334, 336). Inhibitors to cathepsin K (which is involved in systemic bone resorption) (337) hold promise for treating GCOP (295, 338). There is interest in therapeutic inhibitors of 11b-HSD1 for patients with endogenous hypercortisolemia such as Cushing’s disease; these inhibitors – in theory – could also mitigate GCOP but no relevant research has been put forth (53). 

 

GLUCOCORTICOID DISCONTINUATION AND REVERSIBILITY OF GCOP

 

There is no consensus on the reversibility of GCOP. Bone mineral density increases after curative surgery for Cushing’s disease or interruption of exogenous GC treatment (339-341). A prospective study in patients with rheumatoid arthritis showed partial bone regain after discontinuation of low-dose GC therapy that was given for five months (67). If GCs are discontinued and treatment for GCOP is continued, a return to baseline BMD is to be expected within 9 to 15 months (303). In patients with sarcoidosis younger than 45 years, full recovery of bone mass was reported two years after cessation of therapy (342). However, it is unlikely that the large (10% or more) bone mass that is lost during high-dose GC therapy can be completely regained, with full recovery of the mechanical properties of the bone. The likelihood of bone regain may be negatively correlated with the duration of treatment as well as unknown host-related factors. Most complications of osteoporotic fractures, such as vertebral deformities and chronic back pain, are permanent. A sensible approach is to stop anti-osteoporotic treatment 6 to 12 months after discontinuation of GCs administration (303).

 

REFERENCES

 

  1. Mazziotti G, Angeli A, Bilezikian JP, Canalis E, Giustina A. Glucocorticoid-induced osteoporosis: an update. Trends Endocrinol Metab. 2006 May-Jun;17(4):144-9.
  2. Khosla S, Lufkin EG, Hodgson SF, Fitzpatrick LA, Melton LJ, 3rd. Epidemiology and clinical features of osteoporosis in young individuals. Bone. 1994 Sep-Oct;15(5):551-5.
  3. Alesci S, De Martino MU, Ilias I, Gold PW, Chrousos GP. Glucocorticoid-induced osteoporosis: from basic mechanisms to clinical aspects. Neuroimmunomodulation. 2005;12(1):1-19.
  4. Canalis E. Clinical review 83: Mechanisms of glucocorticoid action in bone: implications to glucocorticoid-induced osteoporosis. J Clin Endocrinol Metab. 1996 Oct;81(10):3441-7.
  5. Orcel P. Prise en charge de l'osteoporose cortisonique. Presse Med. 2006 Oct;35(10 Pt 2):1571-7.
  6. Walsh LJ, Wong CA, Pringle M, Tattersfield AE. Use of oral corticosteroids in the community and the prevention of secondary osteoporosis: a cross sectional study. Brit Med J. 1996 Aug 10;313(7053):344-6.
  7. Lupsa BC, Insogna KL, Micheletti RG, Caplan A. Corticosteroid use in chronic dermatologic disorders and osteoporosis. Int J Womens Dermatol. 2021 Dec;7(5Part A):545-51.
  8. Ayub N, Faraj M, Ghatan S, Reijers JAA, Napoli N, Oei L. The Treatment Gap in Osteoporosis. J Clin Med. 2021 Jul 5;10(13):3002.
  9. Compston J. Management of glucocorticoid-induced osteoporosis. Nat Rev Rheumatol. 2010;6:82-8.
  10. Compston JE. Emerging consensus on prevention and treatment of glucocorticoid-induced osteoporosis. Curr Rheumatol Rep. 2007;9:78-84.
  11. Feldstein AC, Elmer PJ, Nichols GA, Herson M. Practice patterns in patient at risk for glucocorticoid-induced osteoporosis. Osteoporos Int. 2005;16:2168-74.
  12. Curtis JR, Westfall AO, Allison JJ, Becker A, Casebeer L, Freeman A, et al. Longitudinal patterns in the prevention of osteoporosis in glucocorticoid-treated patients. Arthritis Rheum. 2005;52:2485-94.
  13. Duyvendak M, Naunton M, Atthobari J, van den Berg PB, Brouwers JR. Corticosteroid-induced osteoporosis prevention: longitudinal practice patterns in The Nederlands 2001-2005. Osteoporos Int. 2007;18:1429-33.
  14. van Staa TP. The pathogenesis, epidemiology and management of glucocorticoid-induced osteoporosis. Calcif Tissue Int. 2006 Sep;79(3):129-37.
  15. van Staa TP, Leufkens HGM, Cooper C. The epidemiology of corticosteroid-induced osteoporosis: a meta-analysis. Osteoporos Int. 2002;13:777-87.
  16. Steinbuch M, Youket TE, Cohen S. Oral glucocorticoid use is associated with an increased risk of fractures. Osteoporos Int. 2004;15:323-8.
  17. van Staa TP, Leufkens HG, Abenhaim L, Begaud B, Zhang B, Cooper C. Use of oral corticosteroids in the United Kingdom. QJM. 2000;93:105-11.
  18. Shaker JL, Lukert BP. Osteoporosis associated with excess glucocorticoids. Endocrinol Metab Clin North Am. 2005 Jun;34(2):341-56, viii-ix.
  19. Chotiyarnwong P, McCloskey EV. Pathogenesis of glucocorticoid-induced osteoporosis and options for treatment. Nat Rev Endocrinol. 2020 Aug;16(8):437-47.
  20. Angeli A, Guglielmi G, Dovio A, Capelli G, de Feo D, Giannini S. High prevalence of asymptomatic vertebral fractures in post-menopausal women receiving chronic glucocorticoid therapy: a cross-sectional outpatient study. Bone. 2006;39:253-9.
  21. Vestergaard P, Rejnmark L, Mosekilde L. Fracture risk associated with systemic and topical corticosteroids. J Intern Med. 2005;257:374-84.
  22. Van Staa TP, Leufkens HGM, Cooper C. Use of inhaled corticosteroids and risk of fractures. J Bone Miner Res. 2001;16:581-8.
  23. van Staa TP, Leufkens HGM, Cooper C. Bone loss and inhaled glucocorticoids. N Engl J Med. 2002;346:533-5.
  24. Hubbard RB, Smith CJP, Smeeth L, Harrison TW, Tattersfield AE. Inhaled corticosteroids and hip fractures: a population-based case-control study. Am J Respir Crit Care Med. 2002;166:1563-6.
  25. de Vries F, Pouwels S, Lammers JW, Leufkens HG, Bracke M, Cooper C. Use of inhaled and oral glucocorticoids, severity of inflammatory disease and risk of hip/femur fracture: a population-based case-control study. J Intern Med. 2007;261:170-7.
  26. Buehring B, Viswanathan R, Binkley N, Busse W. Glucocorticoid-induced osteoporosis: an update on effects and management. J Allergy Clin Immunol. 2013 Nov;132(5):1019-30.
  27. Mazziotti G, Giustina A, Canalis E, Bilezikian JP. Glucocorticoid-induced Osteoporosis: clinical and therapeutic aspects. Arq Bras Endocrinol Metab. 2007;51:1404-12.
  28. van Hogezand RA, Hamdy NA. Skeletal morbidity in inflammatory bowel disease. Scan J Gastroenterol. 2006;243:S59-S64.
  29. Romas E. Bone loss in inflammatory arthritis: mechanisms and therapeutic approaches with bisphosphonates. Best Pract Res Clin Rheumatol. 2005;19:1065-79.
  30. Lekamwasam S, Trivedi DP, Khaw KT. An association between respiratory function an bone mineral density in women from the general community: a cross sectional study. Osteoporos Int. 2002;13:710-5.
  31. Sin DD, Man JP, Man SF. The risk of osteoporosis in Caucasian men and women with obstructive airways disease. Am J Med. 2003;114:10-4.
  32. Dykman TR, Gluck OS, Murphy WA, Hahn TJ, Hahn BH. Evaluation of factors associated with glucocorticoid-induced osteopenia in patients with rheumatic diseases. Arthritis Rheum. 1985 Apr;28(4):361-8.
  33. Hofbauer LC, Brueck CC, Singh SK, Dobnig H. Osteoporosis in patients with diabetes mellitus. J Bone Miner Res. 2007;22:1317-28.
  34. Nicodemus KK, Folsom AR. Type 1 and type 2 diabetes and incident hip fractures in postmenopausal women. Diabetes Care. 2001;24:1192-7.
  35. Zhukouskaya VV, Shepelkevich AP, Chiodini I. Bone Health in Type 1 Diabetes: Where We Are Now and How We Should Proceed. Advances in Endocrinology. 2014;DOI: 10.1155/2014/982129.
  36. Botushanov NP, Orbetzova MM. Bone mineral density and fracture risk in patients with type 1 and type 2 diabetes mellitus. Folia Med (Plovdiv). 2009;51:12-7.
  37. Prentice A. Diet, nutrition and the prevention of osteoporosis. Public Health Nutr. 2004;7:227-43.
  38. Heaney RP. Effects of caffeine on bone and the calcium economy. Food and Chemical Toxicology. 2002 2002/9;40(9):1263-70.
  39. Heaney RP. Long-latency deficiency disease: insights from calcium and vitamin D. Am J Clin Nutr. 2003 November 1, 2003;78(5):912-9.
  40. Packard PT, Heaney RP. Medical Nutrition Therapy for Patients with Osteoporosis. J Am Diet Assoc. 1997 1997/4;97:414-7.
  41. Eastell R, Hannon RA, Cuzick J, Dowsett M, Clack G, Adams JE. Effect of an aromatase inhibitor on BMD and bone turnover markers: 2-years results of the anastrozole, tamoxifen, alone or in combination (ATAC) trial. J Bone Miner Res. 2006;21:1215-23.
  42. Ebeling PR. Osteoporosis in men. N Engl J Med. 2008;358:1474-82.
  43. Coates PS, Fernstrom JD, Fernstrom MH, Schauer PR, Greenspan SL. Gastric bypass surgery for a morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metab. 2004;89:1061-5.
  44. Bachelot A, Chakhtoura Z, Samara-Boustani D, Dulon J, Touraine P, Polak M. “Bone health should be an important concern in the care of patients affected by 21 hydroxylase deficiency. Int J Pediatr Endocrinol. 2010;2010:326275
  45. King JA, Wisniewski AB, Bankowski BJ, Carson KA, Zacur HA, Migeon CJ. Long-term corticosteroid replacement and bone mineral density in adult women with classical congenital adrenal hyperplasia. J Clin Endocrinol Metab. 2006 Mar;91(3):865-9.
  46. Cizza G, Ravn P, Chrousos GP, Gold PW. Depression: a major, unrecognized risk factor for osteoporosis? Trends Endocrinol Metab. 2001 Jul;12(5):198-203.
  47. Thompson JM, Modin GW, Arnaud CD, Lane NE. Not all postmenopausal women on chronic steroid and estrogen treatment are osteoporotic: predictors of bone mineral density. Calcif Tissue Int. 1997 Nov;61(5):377-81.
  48. Varonos S, Ansell BM, Reeve J. Vertebral collapse in juvenile chronic arthritis: its relationship with glucocorticoid therapy. Calcif Tissue Int. 1987 Aug;41(2):75-8.
  49. Rizzato G, Tosi G, Mella C, Montemurro L, Zanni D, Sisti S. Prednisone-induced bone loss in sarcoidosis: a risk especially frequent in postmenopausal women. Sarcoidosis. 1988 Sep;5(2):93-8.
  50. Rich GM, Mudge GH, Laffel GL, LeBoff MS. Cyclosporine A and prednisone-associated osteoporosis in heart transplant recipients. J Heart Lung Transplant. 1992 Sep-Oct;11(5):950-8.
  51. Shane E, Rivas MC, Silverberg SJ, Kim TS, Staron RB, Bilezikian JP. Osteoporosis after cardiac transplantation. Am J Med. 1993 Mar;94(3):257-64.
  52. Komori T. Glucocorticoid Signaling and Bone Biology. Horm Metab Res. 2016 Nov;48(11):755-63.
  53. Martin CS, Cooper MS, Hardy RS. Endogenous Glucocorticoid Metabolism in Bone: Friend or Foe. Front Endocrinol (Lausanne). 2021;12:733611.
  54. Peng CH, Lin WY, Yeh KT, Chen IH, Wu WT, Lin MD. The molecular etiology and treatment of glucocorticoid-induced osteoporosis. Tzu Chi Med J. 2021 Jul-Sep;33(3):212-23.
  55. Jensen PR, Andersen TL, Hauge EM, Bollerslev J, Delaisse JM. A joined role of canopy and reversal cells in bone remodeling--lessons from glucocorticoid-induced osteoporosis. Bone. 2015 Apr;73:16-23.
  56. Katagiri T, Takahashi N. Regulatory mechanisms of osteoblast and osteoclast differentiation. Oral Dis. 2002 May;8(3):147-59.
  57. Xavier A, Toumi H, Lespessailles E. Animal Model for Glucocorticoid Induced Osteoporosis: A Systematic Review from 2011 to 2021. Int J Mol Sci. 2021 Dec 29;23(1).
  58. Ehrlich PJ, Lanyon LE. Mechanical strain and bone cell function: a review. Osteoporos Int. 2002 Sep;13(9):688-700.
  59. Laan RF, Buijs WC, van Erning LJ, Lemmens JA, Corstens FH, Ruijs SH, et al. Differential effects of glucocorticoids on cortical appendicular and cortical vertebral bone mineral content. Calcif Tissue Int. 1993 Jan;52(1):5-9.
  60. Reid IR, Heap SW. Determinants of vertebral mineral density in patients receiving long-term glucocorticoid therapy. Arch Intern Med. 1990 Dec;150(12):2545-8.
  61. Reid IR, Evans MC, Stapleton J. Lateral spine densitometry is a more sensitive indicator of glucocorticoid-induced bone loss. J Bone Miner Res. 1992 Oct;7(10):1221-5.
  62. Yao W, Dai W, Jiang JX, Lane NE. Glucocorticoids and osteocyte autophagy. Bone. 2013 Jun;54(2):279-84.
  63. Reid IR, Evans MC, Wattie DJ, Ames R, Cundy TF. Bone mineral density of the proximal femur and lumbar spine in glucocorticoid-treated asthmatic patients. Osteoporos Int. 1992 Mar;2(2):103-5.
  64. Sambrook P, Birmingham J, Kempler S, Kelly P, Eberl S, Pocock N, et al. Corticosteroid effects on proximal femur bone loss. J Bone Miner Res. 1990 Dec;5(12):1211-6.
  65. Lane NE. An update on glucocorticoid-induced osteoporosis. Rheum Dis Clin North Am. 2001 Feb;27(1):235-53.
  66. Saag KG. Glucocorticoid-induced osteoporosis. Endocrinol Metab Clin North Am. 2003 Mar;32(1):135-57.
  67. Adinoff AD, Hollister JR. Steroid-induced fractures and bone loss in patients with asthma. N Engl J Med. 1983 Aug 4;309(5):265-8.
  68. Dempster DW. Bone histomorphometry in glucocorticoid-induced osteoporosis. J Bone Miner Res. 1989 Apr;4(2):137-41.
  69. Hahn TJ, Boisseau VC, Avioli LV. Effect of chronic corticosteroid administration on diaphyseal and metaphyseal bone mass. J Clin Endocrinol Metab. 1974 Aug;39(2):274-82.
  70. Aaron JE, Francis RM, Peacock M, Makins NB. Contrasting microanatomy of idiopathic and corticosteroid-induced osteoporosis. Clin Orthop. 1989 Jun(243):294-305.
  71. Hartmann K, Koenen M, Schauer S, Wittig-Blaich S, Ahmad M, Baschant U, et al. Molecular Actions of Glucocorticoids in Cartilage and Bone During Health, Disease, and Steroid Therapy. Physiol Rev. 2016 Apr;96(2):409-47.
  72. Bamberger CM, Schulte HM, Chrousos GP. Molecular determinants of glucocorticoid receptor function and tissue sensitivity to glucocorticoids. Endocr Rev. 1996 Jun;17(3):245-61.
  73. Vottero A, Chrousos GP. Glucocorticoid Receptor beta: View I. Trends Endocrinol Metab. 1999 Oct;10(8):333-8.
  74. Beavan S, Horner A, Bord S, Ireland D, Compston J. Colocalization of glucocorticoid and mineralocorticoid receptors in human bone. J Bone Miner Res. 2001 Aug;16(8):1496-504.
  75. Masera RG, Dovio A, Sartori ML, Racca S, Angeli A. Interleukin-6 upregulates glucocorticoid receptor numbers in human osteoblast-like cells. Z Rheumatol. 2000;59 Suppl 2:II/103-7.
  76. Dovio A, Masera RG, Sartori ML, Racca S, Angeli A. Autocrine up-regulation of glucocorticoid receptors by interleukin-6 in human osteoblast-like cells. Calcif Tissue Int. 2001 Nov;69(5):293-8.
  77. Dovio A, Sartori ML, Masera RG, Ceoloni B, Reimondo G, Prolo P, et al. Autocrine down-regulation of glucocorticoid receptors by interleukin-11 in human osteoblast-like cell lines. J Endocrinol. 2003 Apr;177(1):109-17.
  78. Angeli A, Dovio A, Sartori ML, Masera RG, Ceoloni B, Prolo P, et al. Interactions between glucocorticoids and cytokines in the bone microenvironment. Ann N Y Acad Sci. 2002 Jun;966:97-107.
  79. Piemontese M, Onal M, Xiong J, Wang Y, Almeida M, Thostenson JD, et al. Suppression of autophagy in osteocytes does not modify the adverse effects of glucocorticoids on cortical bone. Bone. 2015 Jun;75:18-26.
  80. Weinstein RS, Jilka RL, Michael Parfitt A, Manolagas SC. Inhibition of Osteoblastogenesis and Promotion of Apoptosis of Osteoblasts and Osteocytes by Glucocorticoids . Potential Mechanisms of Their Deleterious Effects on Bone. J Clin Invest. 1998;102(2):274-82.
  81. O'Brien CA, Jia D, Plotkin LI, Bellido T, Powers CC, Stewart SA, et al. Glucocorticoids act directly on osteoblasts and osteocytes to induce their apoptosis and reduce bone formation and strength. Endocrinology. 2004;145:1835-41.
  82. Toth M, Grossman A. Glucocorticoid-induced osteoporosis: lessons from Cushing's syndrome. Clin Endocrinol (Oxf). 2013 Jul;79(1):1-11.
  83. Webster JM, Fenton CG, Langen R, Hardy RS. Exploring the Interface between Inflammatory and Therapeutic Glucocorticoid Induced Bone and Muscle Loss. Int J Mol Sci. 2019 Nov 16;20(22).
  84. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000 Apr;21(2):115-37.
  85. Shi J, Wang L, Zhang H, Jie Q, Li X, Shi Q, et al. Glucocorticoids: Dose-related effects on osteoclast formation and function via reactive oxygen species and autophagy. Bone. 2015 Oct;79:222-32.
  86. Sato AY, Tu X, McAndrews KA, Plotkin LI, Bellido T. Prevention of glucocorticoid induced-apoptosis of osteoblasts and osteocytes by protecting against endoplasmic reticulum (ER) stress in vitro and in vivo in female mice. Bone. 2015 Apr;73:60-8.
  87. Stromstedt PE, Poellinger L, Gustafsson JA, Carlstedt-Duke J. The glucocorticoid receptor binds to a sequence overlapping the TATA box of the human osteocalcin promoter: a potential mechanism for negative regulation. Mol Cell Biol. 1991 Jun;11(6):3379-83.
  88. Heinrichs AA, Bortell R, Rahman S, Stein JL, Alnemri ES, Litwack G, et al. Identification of multiple glucocorticoid receptor binding sites in the rat osteocalcin gene promoter. Biochemistry. 1993 Oct 26;32(42):11436-44.
  89. Jonat C, Rahmsdorf HJ, Park KK, Cato AC, Gebel S, Ponta H, et al. Antitumor promotion and antiinflammation: down-modulation of AP-1 (Fos/Jun) activity by glucocorticoid hormone. Cell. 1990 Sep 21;62(6):1189-204.
  90. Morrison N, Eisman J. Role of the negative glucocorticoid regulatory element in glucocorticoid repression of the human osteocalcin promoter. J Bone Miner Res. 1993 Aug;8(8):969-75.
  91. Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J Clin Invest. 2005 Dec;115(12):3318-25.
  92. Ohnaka K, Tanabe M, Kawate H, Nawata H, Takayanagi R. Glucocorticoid suppresses the canonical Wnt signal in cultured human osteoblasts. Biochem Biophys Res Commun. 2005 Apr 1;329(1):177-81.
  93. Weinstein RS. Glucocorticoid-induced bone disease. In: Rosen CJ, editor. Primer on the metabolic bone diseases and disorders of mineral metabolism. Ames, Iowa: John Wiley & Sons/American Society for Bone and Mineral Research; 2013. p. 473-81.
  94. Moutsatsou P, Kassi E, Papavassiliou AG. Glucocorticoid receptor signaling in bone cells. Trends Mol Med. 2012 Jun;18(6):348-59.
  95. Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, et al. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab. 2010 Jun 9;11(6):517-31.
  96. Derfoul A, Perkins GL, Hall DJ, Tuan RS. Glucocorticoids promote chondrogenic differentiation of adult human mesenchymal stem cells by enhancing expression of cartilage extracellular matrix genes. Stem Cells. 2006 Jun;24(6):1487-95.
  97. Wood CL, Soucek O, Wong SC, Zaman F, Farquharson C, Savendahl L, et al. Animal models to explore the effects of glucocorticoids on skeletal growth and structure. J Endocrinol. 2018 Jan;236(1):R69-R91.
  98. Kang H, Chen H, Huang P, Qi J, Qian N, Deng L, et al. Glucocorticoids impair bone formation of bone marrow stromal stem cells by reciprocally regulating microRNA-34a-5p. Osteoporos Int. 2016;27:1493-505.
  99. Gronowicz G, McCarthy MB, Raisz LG. Glucocorticoids stimulate resorption in fetal rat parietal bones in vitro. J Bone Miner Res. 1990 Dec;5(12):1223-30.
  100. Lowe C, Gray DH, Reid IR. Serum blocks the osteolytic effect of cortisol in neonatal mouse calvaria. Calcif Tissue Int. 1992 Feb;50(2):189-92.
  101. Conaway HH, Grigorie D, Lerner UH. Differential effects of glucocorticoids on bone resorption in neonatal mouse calvariae stimulated by peptide and steroid-like hormones. J Endocrinol. 1997 Dec;155(3):513-21.
  102. Bar-Shavit Z, Kahn AJ, Pegg LE, Stone KR, Teitelbaum SL. Glucocorticoids modulate macrophage surface oligosaccharides and their bone binding activity. J Clin Invest. 1984 May;73(5):1277-83.
  103. Teitelbaum SL, Bar-Shavit Z, Fallon MD, Imbimbo C, Malone JD, Kahn AJ. Glucocorticoids and bone resorption. Adv Exp Med Biol. 1984;171:121-9.
  104. Liu YZ, Dvornyk V, Lu Y, Shen H, Lappe JM, Recker RR, et al. A novel pathophysiological mechanism for osteoporosis suggested by an in vivo gene expression study of circulating monocytes. J Biol Chem. 2005 Aug 12;280(32):29011-6.
  105. Takuma A, Kaneda T, Sato T, Ninomiya S, Kumegawa M, Hakeda Y. Dexamethasone enhances osteoclast formation synergistically with transforming growth factor-beta by stimulating the priming of osteoclast progenitors for differentiation into osteoclasts. J Biol Chem. 2003 Nov 7;278(45):44667-74.
  106. Heymann D, Guicheux J, Gouin F, Passuti N, Daculsi G. Cytokines, growth factors and osteoclasts. Cytokine. 1998 Mar;10(3):155-68.
  107. Rifas L. Bone and cytokines: beyond IL-1, IL-6 and TNF-alpha. Calcif Tissue Int. 1999 Jan;64(1):1-7.
  108. Horowitz MC, Xi Y, Wilson K, Kacena MA. Control of osteoclastogenesis and bone resorption by members of the TNF family of receptors and ligands. Cytokine Growth Factor Rev. 2001 Mar;12(1):9-18.
  109. Hofbauer LC, Heufelder AE. Clinical review 114: hot topic. The role of receptor activator of nuclear factor-kappaB ligand and osteoprotegerin in the pathogenesis and treatment of metabolic bone diseases. J Clin Endocrinol Metab. 2000 Jul;85(7):2355-63.
  110. Pfeilschifter J, Seyedin SM, Mundy GR. Transforming growth factor beta inhibits bone resorption in fetal rat long bone cultures. J Clin Invest. 1988 Aug;82(2):680-5.
  111. Ake Y, Saegusa Y, Matsubara T, Mizuno K. Cultured osteoblast synthesize nitric oxide in response to cytokines and lipopolysaccharide. Kobe J Med Sci. 1994 Aug;40(3-4):125-37.
  112. Vidal NO, Brandstrom H, Jonsson KB, Ohlsson C. Osteoprotegerin mRNA is expressed in primary human osteoblast-like cells: down-regulation by glucocorticoids. J Endocrinol. 1998;159:191-5.
  113. Bornefalk E, Dahlen I, Johannsson G, Ljunggren O, Ohlsson C. Serum levels of osteoprotegerin: effects of glucocorticoids and growth hormone. Bone. 1998;23:S486.
  114. Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg TC, et al. Stimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: potential paracrine mechanisms of glucocorticoid-induced osteoporosis. Endocrinology. 1999 Oct;140(10):4382-9.
  115. Hock JM, Centrella M, Canalis E. Insulin-like growth factor I has independent effects on bone matrix formation and cell replication. Endocrinology. 1988 Jan;122(1):254-60.
  116. Canalis E, Rydziel S, Delany AM, Varghese S, Jeffrey JJ. Insulin-like growth factors inhibit interstitial collagenase synthesis in bone cell cultures. Endocrinology. 1995 Apr;136(4):1348-54.
  117. Delany AM, Durant D, Canalis E. Glucocorticoid suppression of IGF I transcription in osteoblasts. Mol Endocrinol. 2001 Oct;15(10):1781-9.
  118. Bennett A, Chen T, Feldman D, Hintz RL, Rosenfeld RG. Characterization of insulin-like growth factor I receptors on cultured rat bone cells: regulation of receptor concentration by glucocorticoids. Endocrinology. 1984 Oct;115(4):1577-83.
  119. Rydziel S, Canalis E. Cortisol represses insulin-like growth factor II receptor transcription in skeletal cell cultures. Endocrinology. 1995 Oct;136(10):4254-60.
  120. Rechler MM. Insulin-like growth factor binding proteins. Vitam Horm. 1993;47:1-114.
  121. Okazaki R, Riggs BL, Conover CA. Glucocorticoid regulation of insulin-like growth factor-binding protein expression in normal human osteoblast-like cells. Endocrinology. 1994 Jan;134(1):126-32.
  122. Gabbitas B, Pash JM, Delany AM, Canalis E. Cortisol inhibits the synthesis of insulin-like growth factor-binding protein-5 in bone cell cultures by transcriptional mechanisms. J Biol Chem. 1996 Apr 12;271(15):9033-8.
  123. Andress DL, Birnbaum RS. Human osteoblast-derived insulin-like growth factor (IGF) binding protein-5 stimulates osteoblast mitogenesis and potentiates IGF action. J Biol Chem. 1992 Nov 5;267(31):22467-72.
  124. Mehls O, Himmele R, Homme M, Kiepe D, Klaus G. The interaction of glucocorticoids with the growth hormone-insulin-like growth factor axis and its effects on growth plate chondrocytes and bone cells. J Pediatr Endocrinol Metab. 2001;14:1475-82.
  125. Pereira RC, Blanquaert F, Canalis E. Cortisol enhances the expression of mac25/insulin-like growth factor-binding protein-related protein-1 in cultured osteoblasts. Endocrinology. 1999 Jan;140(1):228-32.
  126. Wong SC, Dobie R, Altowati MA, Werther GA, Farquharson C, Ahmed SF. Growth and the Growth Hormone-Insulin Like Growth Factor 1 Axis in Children With Chronic Inflammation: Current Evidence, Gaps in Knowledge, and Future Directions. Endocrine Reviews. 2016;37(1):62-110.
  127. Canalis E, Pash J, Varghese S. Skeletal growth factors. Crit Rev Eukaryot Gene Expr. 1993;3(3):155-66.
  128. Gurlek A, Kumar R. Regulation of osteoblast growth by interactions between transforming growth factor-beta and 1alpha,25-dihydroxyvitamin D3. Crit Rev Eukaryot Gene Expr. 2001;11(4):299-317.
  129. Alliston T, Choy L, Ducy P, Karsenty G, Derynck R. TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. Embo J. 2001 May 1;20(9):2254-72.
  130. Centrella M, McCarthy TL, Canalis E. Glucocorticoid regulation of transforming growth factor beta 1 activity and binding in osteoblast-enriched cultures from fetal rat bone. Mol Cell Biol. 1991 Sep;11(9):4490-6.
  131. Oursler MJ, Riggs BL, Spelsberg TC. Glucocorticoid-induced activation of latent transforming growth factor-beta by normal human osteoblast-like cells. Endocrinology. 1993 Nov;133(5):2187-96.
  132. Fuller K, Owens J, Chambers TJ. The effect of hepatocyte growth factor on the behaviour of osteoclasts. Biochem Biophys Res Commun. 1995 Jul 17;212(2):334-40.
  133. Sato T, Hakeda Y, Yamaguchi Y, Mano H, Tezuka K, Matsumoto K, et al. Hepatocyte growth factor is involved in formation of osteoclast-like cells mediated by clonal stromal cells (MC3T3-G2/PA6). J Cell Physiol. 1995 Jul;164(1):197-204.
  134. Grano M, Galimi F, Zambonin G, Colucci S, Cottone E, Zallone AZ, et al. Hepatocyte growth factor is a coupling factor for osteoclasts and osteoblasts in vitro. Proc Natl Acad Sci U S A. 1996 Jul 23;93(15):7644-8.
  135. Ferracini R, Di Renzo MF, Scotlandi K, Baldini N, Olivero M, Lollini P, et al. The Met/HGF receptor is over-expressed in human osteosarcomas and is activated by either a paracrine or an autocrine circuit. Oncogene. 1995 Feb 16;10(4):739-49.
  136. Hock JM, Canalis E. Platelet-derived growth factor enhances bone cell replication, but not differentiated function of osteoblasts. Endocrinology. 1994 Mar;134(3):1423-8.
  137. Raines EW, Lane TF, Iruela-Arispe ML, Ross R, Sage EH. The extracellular glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-AB and -BB and inhibits the binding of PDGF to its receptors. Proc Natl Acad Sci U S A. 1992 Feb 15;89(4):1281-5.
  138. Ng KW, Manji SS, Young MF, Findlay DM. Opposing influences of glucocorticoid and retinoic acid on transcriptional control in preosteoblasts. Mol Endocrinol. 1989 Dec;3(12):2079-85.
  139. Raisz LG. Prostaglandins and bone: physiology and pathophysiology. Osteoarthr Cartilage. 1999 Jul;7(4):419-21.
  140. Chyun YS, Raisz LG. Stimulation of bone formation by prostaglandin E2. Prostaglandins. 1984 Jan;27(1):97-103.
  141. Canalis E. The hormonal and local regulation of bone formation. Endocr Rev. 1983 Winter;4(1):62-77.
  142. Kenkre JS, Bassett JHD. The bone remodelling cycle. Annals of Clinical Biochemistry 2017;10.1177/0004563218759371.
  143. Raisz LG, Dietrich JW, Canalis EM. Factors influencing bone formation in organ culture. Isr J Med Sci. 1976 Feb;12(2):108-14.
  144. Raisz LG, Fall PM. Biphasic effects of prostaglandin E2 on bone formation in cultured fetal rat calvariae: interaction with cortisol. Endocrinology. 1990 Mar;126(3):1654-9.
  145. Hughes-Fulford M. Prostaglandin regulation of gene expression and growth in normal and malignant tissues. Adv Exp Med Biol. 1997;400A:269-78.
  146. Hughes-Fulford M, Appel R, Kumegawa M, Schmidt J. Effect of dexamethasone on proliferating osteoblasts: inhibition of prostaglandin E2 synthesis, DNA synthesis, and alterations in actin cytoskeleton. Exp Cell Res. 1992 Nov;203(1):150-6.
  147. Kimberg DV, Baerg RD, Gershon E, Graudusius RT. Effect of cortisone treatment on the active transport of calcium by the small intestine. J Clin Invest. 1971 Jun;50(6):1309-21.
  148. Fuller K, Chambers TJ. Effect of arachidonic acid metabolites on bone resorption by isolated rat osteoclasts. J Bone Miner Res. 1989 Apr;4(2):209-15.
  149. Klein DC, Raisz LG. Prostaglandins: stimulation of bone resorption in tissue culture. Endocrinology. 1970 Jun;86(6):1436-40.
  150. Bellido T, Hill Gallant KM. Hormonal Effects on Bone Cells. In: Burr DB, Allen MR, editors. Basic and Applied Bone Biology. London: Elsevier; 2014. p. 299-314.
  151. Wajchenberg BL, Pereira VG, Kieffer J, Ursic S. Effect of dexamethasone on calcium metabolism and 47Ca kinetics in normal subjects. Acta Endocrinol (Copenh). 1969 May;61(1):173-92.
  152. Morris HA, Need AG, O'Loughlin PD, Horowitz M, Bridges A, Nordin BE. Malabsorption of calcium in corticosteroid-induced osteoporosis. Calcif Tissue Int. 1990 May;46(5):305-8.
  153. Binder HJ. Effect of dexamethasone on electrolyte transport in the large intestine of the rat. Gastroenterology. 1978 Aug;75(2):212-7.
  154. Sjoberg HE. Retention of orally administered 47-calcium in man under normal and diseased conditions studied with a whole-body counter technique. Acta Med Scand Suppl. 1970;509:1-28.
  155. Lee DB. Unanticipated stimulatory action of glucocorticoids on epithelial calcium absorption. Effect of dexamethasone on rat distal colon. J Clin Invest. 1983 Feb;71(2):322-8.
  156. Hahn TJ, Halstead LR, Strates B, Imbimbo B, Baran DT. Comparison of subacute effects of oxazacort and prednisone on mineral metabolism in man. Calcif Tissue Int. 1980;31(2):109-15.
  157. Lekkerkerker JF, Van Woudenberg F, Doorenbos H. Influence of low dose of steroid therapy on calcium absorption. Acta Endocrinol (Copenh). 1972 Mar;69(3):488-96.
  158. Aloia JF, Semla HM, Yeh JK. Discordant effects of glucocorticoids on active and passive transport of calcium in the rat duodenum. Calcif Tissue Int. 1984 May;36(3):327-31.
  159. Shultz TD, Bollman S, Kumar R. Decreased intestinal calcium absorption in vivo and normal brush border membrane vesicle calcium uptake in cortisol-treated chickens: evidence for dissociation of calcium absorption from brush border vesicle uptake. Proc Natl Acad Sci U S A. 1982 Jun;79(11):3542-6.
  160. Kimura S, Rasmussen H. Adrenal glucocorticoids, adenine nucleotide translocation, and mitochondrial calcium accumulation. J Biol Chem. 1977 Feb 25;252(4):1217-25.
  161. Suzuki Y, Ichikawa Y, Saito E, Homma M. Importance of increased urinary calcium excretion in the development of secondary hyperparathyroidism of patients under glucocorticoid therapy. Metabolism. 1983 Feb;32(2):151-6.
  162. Nielsen HK, Thomsen K, Eriksen EF, Charles P, Storm T, Mosekilde L. The effects of high-dose glucocorticoid administration on serum bone gamma carboxyglutamic acid-containing protein, serum alkaline phosphatase and vitamin D metabolites in normal subjects. Bone Miner. 1988 Apr;4(1):105-13.
  163. Adams JS, Wahl TO, Lukert BP. Effects of hydrochlorothiazide and dietary sodium restriction on calcium metabolism in corticosteroid treated patients. Metabolism. 1981 Mar;30(3):217-21.
  164. Au WY. Cortisol stimulation of parathyroid hormone secretion by rat parathyroid glands in organ culture. Science. 1976 Sep 10;193(4257):1015-7.
  165. Cosman F, Nieves J, Herbert J, Shen V, Lindsay R. High-dose glucocorticoids in multiple sclerosis patients exert direct effects on the kidney and skeleton. J Bone Miner Res. 1994 Jul;9(7):1097-105.
  166. Freiberg JM, Kinsella J, Sacktor B. Glucocorticoids increase the Na+-H+ exchange and decrease the Na+ gradient-dependent phosphate-uptake systems in renal brush border membrane vesicles. Proc Natl Acad Sci U S A. 1982 Aug;79(16):4932-6.
  167. Heersche JN, Jez DH, Aubin J, Sodek J. Regulation of hormone responsiveness of bone in vitro by corticosteroids, PTH, PGE2, and calcitonin. In: Talmage RV, Cohn DV, Mathews JL, editors. Hormonal control of calcium metabolism. Amsterdam: Excerpta Medica; 1981. p. 157-62.
  168. Fucik RF, Kukreja SC, Hargis GK, Bowser EN, Henderson WJ, Williams GA. Effect of glucocorticoids on function of the parathyroid glands in man. J Clin Endocrinol Metab. 1975 Jan;40(1):152-5.
  169. Lukert BP, Adams JS. Calcium and phosphorus homeostasis in man. Effect of corticosteroids. Arch Intern Med. 1976 Nov;136(11):1249-53.
  170. Bonadonna S, Burattin A, Nuzzo M, Bugari G, Rosei EA, Valle D, et al. Chronic glucocorticoid treatment alters spontaneous pulsatile parathyroid hormone secretory dynamics in human subjects. Eur J Endocrinol. 2005 Feb;152(2):199-205.
  171. Lukert BP, Stanbury SW, Mawer EB. Vitamin D and intestinal transport of calcium: effects of prednisolone. Endocrinology. 1973 Sep;93(3):718-22.
  172. Hahn TJ, Halstead LR, Haddad JG, Jr. Serum 25-hydroxyvitamin D concentrations in patients receiving chronic corticosteroid therapy. J Lab Clin Med. 1977 Aug;90(2):399-404.
  173. Chesney RW, Mazess RB, Hamstra AJ, DeLuca HF, O'Reagan S. Reduction of serum-1, 25-dihydroxyvitamin-D3 in children receiving glucocorticoids. Lancet. 1978 Nov 25;2(8100):1123-5.
  174. Zerwekh JE, Emkey RD, Harris ED, Jr. Low-dose prednisone therapy in rheumatoid arthritis: effect on vitamin D metabolism. Arthritis Rheum. 1984 Sep;27(9):1050-2.
  175. Seeman E, Kumar R, Hunder GG, Scott M, Heath H, 3rd, Riggs BL. Production, degradation, and circulating levels of 1,25-dihydroxyvitamin D in health and in chronic glucocorticoid excess. J Clin Invest. 1980 Oct;66(4):664-9.
  176. Colette C, Monnier L, Pares Herbute N, Blotman F, Mirouze J. Calcium absorption in corticoid treated subjects effects of a single oral dose of calcitriol. Horm Metab Res. 1987 Jul;19(7):335-8.
  177. Luton JP, Thieblot P, Valcke JC, Mahoudeau JA, Bricaire H. Reversible gonadotropin deficiency in male Cushing's disease. J Clin Endocrinol Metab. 1977 Sep;45(3):488-95.
  178. Sakakura M, Takebe K, Nakagawa S. Inhibition of luteinizing hormone secretion induced by synthetic LRH by long-term treatment with glucocorticoids in human subjects. J Clin Endocrinol Metab. 1975 May;40(5):774-9.
  179. Hsueh AJ, Erickson GF. Glucocorticoid inhibition of FSH-induced estrogen production in cultured rat granulosa cells. Steroids. 1978 Dec;32(5):639-48.
  180. Swift AD, Crighton DB. The effects of certain steroid hormones on the activity of ovine hypothalamic luteinizing hormone-releasing hormone (LH-RH)-degrading enzymes. FEBS Lett. 1979 Apr 1;100(1):110-2.
  181. Dubey AK, Plant TM. A suppression of gonadotropin secretion by cortisol in castrated male rhesus monkeys (Macaca mulatta) mediated by the interruption of hypothalamic gonadotropin-releasing hormone release. Biol Reprod. 1985 Sep;33(2):423-31.
  182. Sapolsky RM. Stress-induced suppression of testicular function in the wild baboon: role of glucocorticoids. Endocrinology. 1985 Jun;116(6):2273-8.
  183. Crilly R, Cawood M, Marshall DH, Nordin BE. Hormonal status in normal, osteoporotic and corticosteroid-treated postmenopausal women. J R Soc Med. 1978 Oct;71(10):733-6.
  184. Montecucco C, Caporali R, Caprotti P, Caprotti M, Notario A. Sex hormones and bone metabolism in postmenopausal rheumatoid arthritis treated with two different glucocorticoids. J Rheumatol. 1992 Dec;19(12):1895-900.
  185. MacAdams MR, White RH, Chipps BE. Reduction of serum testosterone levels during chronic glucocorticoid therapy. Ann Intern Med. 1986 May;104(5):648-51.
  186. Urban RJ, Veldhuis JD. Endocrine control of steroidogenesis in granulosa cells. Oxf Rev Reprod Biol. 1992;14:225-62.
  187. Buyon JP, Dooley MA, Meyer WR, Petri M, Licciardi F. Recommendations for exogenous estrogen to prevent glucocorticoid-induced osteoporosis in premenopausal women with oligo- or amenorrhea: comment on the American College of Rheumatology recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Rheum. 1997 Aug;40(8):1548-9.
  188. Goulding A, Gold E. Effects of chronic prednisolone treatment on bone resorption and bone composition in intact and ovariectomized rats and in ovariectomized rats receiving beta-estradiol. Endocrinology. 1988 Feb;122(2):482-7.
  189. Ohlsson C, Bengtsson BA, Isaksson OG, Andreassen TT, Slootweg MC. Growth hormone and bone. Endocr Rev. 1998 Feb;19(1):55-79.
  190. Giustina A, Bussi AR, Jacobello C, Wehrenberg WB. Effects of recombinant human growth hormone (GH) on bone and intermediary metabolism in patients receiving chronic glucocorticoid treatment with suppressed endogenous GH response to GH-releasing hormone. J Clin Endocrinol Metab. 1995 Jan;80(1):122-9.
  191. Manelli F, Carpinteri R, Bossoni S, Burattin A, Bonadonna S, Agabiti Rosei E, et al. Growth hormone in glucocorticoid-induced osteoporosis. Front Horm Res. 2002;30:174-83.
  192. King AP, Tseng MJ, Logsdon CD, Billestrup N, Carter-Su C. Distinct cytoplasmic domains of the growth hormone receptor are required for glucocorticoid- and phorbol ester-induced decreases in growth hormone (GH) binding. These domains are different from that reported for GH-induced receptor internalization. J Biol Chem. 1996 Jul 26;271(30):18088-94.
  193. Pratt WB, Aronow L. The effect of glucocorticoids on protein and nucleic acid synthesis in mouse fibroblasts growing in vitro. J Biol Chem. 1966 Nov 25;241(22):5244-50.
  194. Leibovich SJ, Ross R. The role of the macrophage in wound repair. A study with hydrocortisone and antimacrophage serum. Am J Pathol. 1975 Jan;78(1):71-100.
  195. Afifi AK, Bergman RA, Harvey JC. Steroid myopathy. Clinical, histologic and cytologic observations. Johns Hopkins Med J. 1968 Oct;123(4):158-73.
  196. Pleasure DE, Walsh GO, Engel WK. Atrophy of skeletal muscle in patients with Cushing's syndrome. Arch Neurol. 1970 Feb;22(2):118-25.
  197. Rebuffe-Scrive M, Krotkiewski M, Elfverson J, Bjorntorp P. Muscle and adipose tissue morphology and metabolism in Cushing's syndrome. J Clin Endocrinol Metab. 1988 Dec;67(6):1122-8.
  198. Wang L, Luo GJ, Wang JJ, Hasselgren PO. Dexamethasone stimulates proteasome- and calcium-dependent proteolysis in cultured L6 myotubes. Shock. 1998 Oct;10(4):298-306.
  199. Auclair D, Garrel DR, Chaouki Zerouala A, Ferland LH. Activation of the ubiquitin pathway in rat skeletal muscle by catabolic doses of glucocorticoids. Am J Physiol. 1997 Mar;272(3 Pt 1):C1007-16.
  200. Danneskiold-Samsoe B, Grimby G. The influence of prednisone on the muscle morphology and muscle enzymes in patients with rheumatoid arthritis. Clin Sci (Lond). 1986 Dec;71(6):693-701.
  201. Askari A, Vignos PJ, Jr., Moskowitz RW. Steroid myopathy in connective tissue disease. Am J Med. 1976 Oct;61(4):485-92.
  202. Ruegsegger P, Medici TC, Anliker M. Corticosteroid-induced bone loss. A longitudinal study of alternate day therapy in patients with bronchial asthma using quantitative computed tomography. Eur J Clin Pharmacol. 1983;25(5):615-20.
  203. Huizenga NA, Koper JW, De Lange P, Pols HA, Stolk RP, Burger H, et al. A polymorphism in the glucocorticoid receptor gene may be associated with and increased sensitivity to glucocorticoids in vivo. J Clin Endocrinol Metab. 1998 Jan;83(1):144-51.
  204. van Rossum EF, Lamberts SW. Polymorphisms in the glucocorticoid receptor gene and their associations with metabolic parameters and body composition. Recent Prog Horm Res. 2004;59:333-57.
  205. White PC, Mune T, Agarwal AK. 11 beta-Hydroxysteroid dehydrogenase and the syndrome of apparent mineralocorticoid excess. Endocr Rev. 1997 Feb;18(1):135-56.
  206. Tomlinson JW, Walker EA, Bujalska IJ, Draper N, Lavery GG, Cooper MS. 11beta-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Endocr Rev. 2004;25:31-66.
  207. Tannin GM, Agarwal AK, Monder C, New MI, White PC. The human gene for 11 beta-hydroxysteroid dehydrogenase. Structure, tissue distribution, and chromosomal localization. J Biol Chem. 1991 Sep 5;266(25):16653-8.
  208. Stewart PM, Krozowski ZS. 11 beta-Hydroxysteroid dehydrogenase. Vitam Horm. 1999;57:249-324.
  209. Cooper MS, Walker EA, Bland R, Fraser WD, Hewison M, Stewart PM. Expression and functional consequences of 11beta-hydroxysteroid dehydrogenase activity in human bone. Bone. 2000 Sep;27(3):375-81.
  210. Arampatzis S, Pasch A, Lippuner K, Mohaupt M. Primary male osteoporosis is associated with enhanced glucocorticoid availability. Rheumatology (Oxford). 2013 Nov;52(11):1983-91.
  211. Cooper MS, Bujalska I, Rabbit E, Walker EA, Bland R, Sheppard MC. Modulation of 11beta-hydroxysteroid dehydrogenase isoenzymes by proinflammatory cytokines in osteoblasts: an autocrine switch from glucocorticoid inactivation to activation. J Bone Mineral Res. 2001;16:1037-44.
  212. Williams LJ, Lyons V, MacLeod I, Rajan V, Darlington GJ, Poli V. C/EBP regulates hepatic transcription of 11beta-hydroxysteroid dehydrogenase type 1. A novel mechanism for cross-talk between the C/EBP and glucocorticoid signaling pathways. J Biol Chem. 2000;275:30232-9.
  213. Escher G, Galli I, Vishwanath BS, Frey BM, Frey FJ. Tumor necrosis factor alpha and interleukin 1beta enhance the cortisone/cortisol shuttle. J Exp Med. 1997 Jul 21;186(2):189-98.
  214. Cooper MS, Rabbit EH, Goddard PE, Bartlett WA, Hewison M, Stewart PM. Osteoblastic 11beta-hydroxysteroid dehydrogenase type 1 activity increases with age and glucocorticoid exposure. J Bone Mineral Res. 2002;17:979-86.
  215. Mundell L, Lindemann R, Douglas J. Monitoring long-term oral corticosteroids. BMJ Open Quality. 2017;6(2).
  216. Gonnelli S, Rottoli P, Cepollaro C, Pondrelli C, Cappiello V, Vagliasindi M, et al. Prevention of corticosteroid-induced osteoporosis with alendronate in sarcoid patients. Calcif Tissue Int. 1997;61:382-5.
  217. Adler RA, Hochberg MC. Suggested Guidelines for Evaluation and Treatment of Glucocorticoid-Induced Osteoporosis for the Department of Veterans Affairs. Arch Intern Med. 2003 November 24, 2003;163(21):2619-24.
  218. Anonymous. Consensus development conference: diagnosis, prophylaxis, and treatment of osteoporosis. Am J Med. 1993 Jun;94(6):646-50.
  219. Anonymous. Osteoporosis prevention, diagnosis, and therapy. JAMA. 2001 Feb 14;285(6):785-95.
  220. Anonymous. Recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis: 2001 update. American College of Rheumatology Ad Hoc Committee on Glucocoritcoid-Induced Osteoporosis. Arthritis Rheum. 2001 Jul;44(7):1496-503.
  221. Eastell R, Reid DM, Compston J, Cooper C, Fogelman I, Francis RM, et al. A UK Consensus Group on management of glucocorticoid-induced osteoporosis: an update. J Intern Med. 1998 Oct;244(4):271-92.
  222. Lukert BP, Higgins JC, Stoskopf MM. Serum osteocalcin is increased in patients with hyperthyroidism and decreased in patients receiving glucocorticoids. J Clin Endocrinol Metab. 1986 May;62(5):1056-8.
  223. Ekenstam E, Stalenheim G, Hallgren R. The acute effect of high dose corticosteroid treatment on serum osteocalcin. Metabolism. 1988 Feb;37(2):141-4.
  224. Prummel MF, Wiersinga WM, Lips P, Sanders GT, Sauerwein HP. The course of biochemical parameters of bone turnover during treatment with corticosteroids. J Clin Endocrinol Metab. 1991 Feb;72(2):382-6.
  225. Reid IR, Chapman GE, Fraser TR, Davies AD, Surus AS, Meyer J, et al. Low serum osteocalcin levels in glucocorticoid-treated asthmatics. J Clin Endocrinol Metab. 1986 Feb;62(2):379-83.
  226. Hall GM, Spector TD, Delmas PD. Markers of bone metabolism in postmenopausal women with rheumatoid arthritis. Effects of corticosteroids and hormone replacement therapy. Arthritis Rheum. 1995 Jul;38(7):902-6.
  227. Reeve J, Loftus J, Hesp R, Ansell BM, Wright DJ, Woo PM. Biochemical prediction of changes in spinal bone mass in juvenile chronic (or rheumatoid) arthritis treated with glucocorticoids. J Rheumatol. 1993 Jul;20(7):1189-95.
  228. Ton FN, Gunawardene SC, Lee H, Neer RM. Effects of low-dose prednisone on bone metabolism. J Bone Miner Res. 2005;20:464-70.
  229. Lems WF, Gerrits MI, Jacobs JW, van Vugt RM, van Rijn HJ, Bijlsma JW. Changes in (markers of) bone metabolism during high dose corticosteroid pulse treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 1996 May;55(5):288-93.
  230. Gennari C, Imbimbo B, Montagnani M, Bernini M, Nardi P, Avioli LV. Effects of prednisone and deflazacort on mineral metabolism and parathyroid hormone activity in humans. Calcif Tissue Int. 1984 May;36(3):245-52.
  231. Baran DT, Faulkner KG, Genant HK, Miller PD, Pacifici R. Diagnosis and management of osteoporosis: guidelines for the utilization of bone densitometry. Calcif Tissue Int. 1997;61:433-40.
  232. Guglielmi G, Lang TF. Quantitative computed tomography. Semin Musculoskelet Radiol. 2002;6:219-27.
  233. Hui SL, Slemenda CW, Johnston CC, Jr. Baseline measurement of bone mass predicts fracture in white women. Ann Intern Med. 1989 Sep 1;111(5):355-61.
  234. Blake GM, Fogelman I. Applications of bone densitometry for osteoporosis. Endocrinol Metab Clin North Am. 1998 Jun;27(2):267-88.
  235. Finkelstein JS, Cleary RL, Butler JP, Antonelli R, Mitlak BH, Deraska DJ, et al. A comparison of lateral versus anterior-posterior spine dual energy x-ray absorptiometry for the diagnosis of osteopenia. J Clin Endocrinol Metab. 1994 Mar;78(3):724-30.
  236. Sandru F, Carsote M, Dumitrascu MC, Albu SE, Valea A. Glucocorticoids and Trabecular Bone Score. J Med Life. 2020 Oct-Dec;13(4):449-53.
  237. Kobza AO, Herman D, Papaioannou A, Lau AN, Adachi JD. Understanding and Managing Corticosteroid-Induced Osteoporosis. Open Access Rheumatol. 2021;13:177-90.
  238. Van Staa TP, Laan RF, Barton IP, Cohen S, Reid DM, Cooper C. Bone density threshold and other predictors of vertebral fracture in patients receiving oral glucocorticoid therapy. Arthritis Rheum. 2003;48:3224-9.
  239. Kanis JA, Johannsson H, Oden A. A meta-analysis of prior corticosteroid use and fracture risk. J Bone Miner Res. 2004;19:893-9.
  240. Kanis JA, Torgerson D, Cooper C. Comparison of the European and USA practice guidelines for Osteoporosis. Trends Endocrinol Metab. 2000 Jan-Feb;11(1):28-32.
  241. Park SY, Gong HS, Kim KM, Kim D, Kim HY, Jeon CH, et al. Korean Guideline for the Prevention and Treatment of Glucocorticoid-induced Osteoporosis. J Bone Metab. 2018 Nov;25(4):195-211.
  242. Adler GS, Shatto A. Screening for osteoporosis and colon cancer under Medicare. Health Care Financ Rev. 2002 Summer;23(4):189-200.
  243. Weinstein RS. Glucocorticoid-induced osteoporosis and osteonecrosis. Endocrinol Metab Clin North Am. 2012 Sep;41(3):595-611.
  244. Kanis JA, Johansson H, Oden A, McCloskey EV. Guidance for the adjustment of FRAX according to the dose of glucocorticoids. Osteoporosis Int. 2011;DOI 10.1007/s00198-010-1524-7.
  245. Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, et al. 2017 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Rheumatol. 2017 Aug;69(8):1521-37.
  246. Lee TH, Song YJ, Kim H, Sung YK, Cho SK. Intervention Thresholds for Treatment in Patients with Glucocorticoid-Induced Osteoporosis: Systematic Review of Guidelines. J Bone Metab. 2020 Nov;27(4):247-59.
  247. Grossman JM, Gordon R, Ranganath VK, Deal C, Caplan L, Chen W, et al. American College of Rheumatology 2010 recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Care Res (Hoboken). 2010;62:1515-26.
  248. Devogelaer JP, Goemaere S, Boonen S, Body JJ, Kaufman JM, Reginster JY, et al. Evidence-based guidelines for the prevention and treatment of glucocorticoid-induced osteoporosis: a consensus document of the Belgian Bone Club. Osteoporos Int. 2006 Jan;17(1):8-19.
  249. Gluck OS, Murphy WA, Hahn TJ, Hahn B. Bone loss in adults receiving alternate day glucocorticoid therapy. A comparison with daily therapy. Arthritis Rheum. 1981 Jul;24(7):892-8.
  250. Avgerinos PC, Cutler GB, Jr., Tsokos GC, Gold PW, Feuillan P, Gallucci WT, et al. Dissociation between cortisol and adrenal androgen secretion in patients receiving alternate day prednisone therapy. J Clin Endocrinol Metab. 1987 Jul;65(1):24-9.
  251. Martinati LC, Bertoldo F, Gasperi E, Fortunati P, Lo Cascio V, Boner AL. Longitudinal evaluation of bone mass in asthmatic children treated with inhaled beclomethasone dipropionate or cromolyn sodium. Allergy. 1998 Jul;53(7):705-8.
  252. Briot K, Roux C. Glucocorticoid-induced osteoporosis. RMD Open. 2015;1(1):e000014.
  253. Frediani B, Falsetti P, Bisogno S, Baldi F, Acciai C, Filippou G, et al. Effects of high dose methylprednisolone pulse therapy on bone mass and biochemical markers of bone metabolism in patients with active rheumatoid arthritis: a 12-month randomized prospective controlled study. J Rheumatol. 2004;31:1083-7.
  254. Nielsen HK, Charles P, Mosekilde L. The effect of single oral doses of prednisone on the circadian rhythm of serum osteocalcin in normal subjects. J Clin Endocrinol Metab. 1988;67:1025-30.
  255. Richy F, Bousquet J, Ehrlich GE, Meunier PJ, Israel E, Morii H, et al. Inhaled corticosteroids effects on bone in asthmatic and COPD patients: a quantitative systematic review. Osteoporos Int. 2003;14:179-90.
  256. Bootsma GP, Dekhuijzen PN, Festen J, Mulder PG, Swinkels LM, van Herwaarden CL. Fluticasone propionate does not influence bone metabolism in contrast to beclomethasone dipropionate. Am J Respir Crit Care Med. 1996 Mar;153(3):924-30.
  257. Luengo M, del Rio L, Pons F, Picado C. Bone mineral density in asthmatic patients treated with inhaled corticosteroids: a case-control study. Eur Respir J. 1997 Sep;10(9):2110-3.
  258. Ebeling PR, Erbas B, Hopper JL, Wark JD, Rubinfeld AR. Bone mineral density and bone turnover in asthmatics treated with long-term inhaled or oral glucocorticoids. J Bone Miner Res. 1998 Aug;13(8):1283-9.
  259. Aris R, Donohue JF, Ontjes D. Inhaled corticosteroids and fracture risk: having our cake and eating it too. Chest. 2005 Jan;127(1):5-7.
  260. van Staa TP, Bishop N, Leufkens HG, Cooper C. Are inhaled corticosteroids associated with an increased risk of fracture in children? Osteoporos Int. 2004;15:785-91.
  261. Pennisi P, Trombetti A, Rizzoli R. Glucocorticoid-induced osteoporosis and its treatment. Clin Orthop Relat Res. 2006 Feb;443:39-47.
  262. Boonen S, inventor IOF, assignee. Corticosteroid-induced osteoporosis. Geneva2011 February 1-3, 2011.
  263. Prestwood KM, Raisz LG. Prevention and treatment of osteoporosis. Clin Cornerstone. 2002;4:31-41.
  264. Adami G, Rahn EJ, Saag KG. Glucocorticoid-induced osteoporosis: from clinical trials to clinical practice. Ther Adv Musculoskelet Dis. 2019;11:1759720X19876468.
  265. Liu Z, Zhang M, Shen Z, Ke J, Zhang D, Yin F. Efficacy and safety of 18 anti-osteoporotic drugs in the treatment of patients with osteoporosis caused by glucocorticoid: A network meta-analysis of randomized controlled trials. PLoS One. 2020;15(12):e0243851.
  266. Buckley LM, Leib ES, Cartularo KS, Vacek PM, Cooper SM. Calcium and vitamin D3 supplementation prevents bone loss in the spine secondary to low-dose corticosteroids in patients with rheumatoid arthritis. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 1996 Dec 15;125(12):961-8.
  267. Adachi JD, Bensen WG, Bianchi F, Cividino A, Pillersdorf S, Sebaldt RJ, et al. Vitamin D and calcium in the prevention of corticosteroid induced osteoporosis: a 3 year followup. J Rheumatol. 1996 Jun;23(6):995-1000.
  268. Reginster JY, Kuntz D, Verdickt W, Wouters M, Guillevin L, Menkes CJ, et al. Prophylactic use of alfacalcidol in corticosteroid-induced osteoporosis. Osteoporos Int. 1999;9(1):75-81.
  269. Amin S, LaValley MP, Simms RW, Felson DT. The role of vitamin D in corticosteroid-induced osteoporosis: a meta-analytic approach. Arthritis Rheum. 1999 Aug;42(8):1740-51.
  270. Homik J, Suarez-Almazor ME, Shea B, Cranney A, Wells G, Tugwell P. Calcium and vitamin D for corticosteroid-induced osteoporosis. Cochrane Database Syst Rev. 2000(2):CD000952.
  271. Fuji N, Hamano T, Mikami S, Nagasawa Y, Isaka Y, Moriyama T, et al. Risedronate, an effective treatment for glucocorticoid-induced bone loss in CKD patients with or without concomitant active vitamin D (PRIUS-CKD). Nephrol Dial Transplant. 2007;22:1601-7.
  272. Dechant KL, Goa KL. Calcitriol. A review of its use in the treatment of postmenopausal osteoporosis and its potential in corticosteroid-induced osteoporosis. Drugs Aging. 1994;5:300-17.
  273. Adverse Drug Reactions Advisory Committee A. Calcitriol and hypercalcaemia. Aust Adv Drug React Bull. 1997;16:2.
  274. LaCroix AZ, Wienpahl J, White LR, Wallace RB, Scherr PA, George LK, et al. Thiazide diuretic agents and the incidence of hip fracture. N Engl J Med. 1990 Feb 1;322(5):286-90.
  275. Felson DT, Sloutskis D, Anderson JJ, Anthony JM, Kiel DP. Thiazide diuretics and the risk of hip fracture. Results from the Framingham Study. JAMA. 1991 Jan 16;265(3):370-3.
  276. Cauley JA, Cummings SR, Seeley DG, Black D, Browner W, Kuller LH, et al. Effects of thiazide diuretic therapy on bone mass, fractures, and falls. The Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1993 May 1;118(9):666-73.
  277. Rodan GA, Fleisch HA. Bisphosphonates: mechanisms of action. J Clin Invest. 1996 Jun 15;97(12):2692-6.
  278. Sambrook PN. How to prevent steroid induced osteoporosis. Ann Rheum Dis. 2005 Feb;64(2):176-8.
  279. Compston J, Bowring C, Cooper A, Cooper C, Davies C, Francis R, et al. Diagnosis and management of osteoporosis in postmenopausal women and older men in the UK: National Osteoporosis Guideline Group (NOGG) update 2013. Maturitas. 2013 Aug;75(4):392-6.
  280. Lekamwasam S, Adachi JD, Agnusdei D, Bilezikian J, Boonen S, Borgstrom F, et al. A framework for the development of guidelines for the management of glucocorticoid-induced osteoporosis. Osteoporos Int. 2012 Sep;23(9):2257-76.
  281. Adachi JD, Saag KG, Delmas PD, Liberman UA, Emkey RD, Seeman E, et al. Two-year effects of alendronate on bone mineral density and vertebral fracture in patients receiving glucocorticoids: a randomized, double-blind, placebo-controlled extension trial. Arthritis Rheum. 2001 Jan;44(1):202-11.
  282. de Nijs RN, Jacobs JW, Lems WF, Laan RF, Algra A, Huisman AM, et al. Alendronate or alfacalcidol in glucocorticoid-induced osteoporosis. N Engl J Med. 2006 Aug 17;355(7):675-84.
  283. Shane E, Addesso V, Namerow PB, McMahon DJ, Lo SH, Staron RB, et al. Alendronate versus calcitriol for the prevention of bone loss after cardiac transplantation. N Engl J Med. 2004;350:767-76.
  284. Wang YK, Zhang YM, Qin SQ, Wang X, Ma T, Guo JB, et al. Effects of alendronate for treatment of glucocorticoid-induced osteoporosis: A meta-analysis of randomized controlled trials. Medicine (Baltimore). 2018 Oct;97(42):e12691.
  285. Reid DM, Hughes RA, Laan RF, Sacco-Gibson NA, Wenderoth DH, Adami S, et al. Efficacy and safety of daily risedronate in the treatment of corticosteroid-induced osteoporosis in men and women: a randomized trial. European Corticosteroid-Induced Osteoporosis Treatment Study. J Bone Miner Res. 2000 Jun;15(6):1006-13.
  286. Reid IR, Brown JP, Burckhardt P, Horowitz Z, Richardson P, Trechsel U, et al. Intravenous Zoledronic Acid in Postmenopausal Women with Low Bone Mineral Density. N Engl J Med. 2002 February 28, 2002;346(9):653-61.
  287. Reid IR. Bisphosphonates: new indications and methods of administration. Curr Opin Rheumatol. 2003 Jul;15(4):458-63.
  288. Theriault RL. Zoledronic acid (Zometa) use in bone disease. Expert Rev Anticancer Ther. 2003 Apr;3(2):157-66.
  289. Biskobing DM. Novel therapies for osteoporosis. Expert Opin Investig Drugs. 2003 Apr;12(4):611-21.
  290. Biskobing DM, Novy AM, Downs R, Jr. Novel therapeutic options for osteoporosis. Curr Opin Rheumatol. 2002 Jul;14(4):447-52.
  291. Doggrell SA. Zoledronate once-yearly increases bone mineral density--implications for osteoporosis. Expert Opin Pharmacother. 2002 Jul;3(7):1007-9.
  292. Reid DM, Devogelaer JP, Saag K, Roux C, Lau CS, Reginster JY, et al. Zoledronic acid and risedronate in the prevention and treatment of glucocorticoid-induced osteoporosis HORIZON): a multicentre, double-blind, double-dummy, randomised controlled trial. Lancet. 2009;373:1253-63.
  293. Ward LM. Glucocorticoid-Induced Osteoporosis: Why Kids Are Different. Front Endocrinol (Lausanne). 2020;11:576.
  294. Teitelbaum SL, Seton MP, Saag KG. Should bisphosphonates be used for long-term treatment of glucocorticoid-induced osteoporosis? Arthritis and rheumatism. 2011;63(2):325-8.
  295. Bultink IE, Baden M, Lems WF. Glucocorticoid-induced osteoporosis: an update on current pharmacotherapy and future directions. Expert Opin Pharmacother. 2013 Feb;14(2):185-97.
  296. Dore RK, Cohen SB, Lane NE, Palmer W, Shergy W, Zhou L, et al. Effects of denosumab on bone mineral density and bone turnover in patients with rheumatoid arthritis receiving concurrent glucocorticoids or bisphosphonates. Ann Rheum Dis. 2010;69:872-5.
  297. Akashi K, Nishimura K, Kageyama G, Ichikawa S, Shirai T, Yamamoto Y, et al. FRI0553 The efficacy of 2-years denosumab treatment for glucocorticoid-induced osteoporosis (GIOP). Annals of the Rheumatic Diseases. 2017;76(Suppl 2):699.
  298. Denosumab for glucocorticoid-induced osteoporosis. Birmingham, UK: NIHR Horizon Scanning Centre, University of Birmingham2014.
  299. Amgen. FDA Accepts Supplemental Biologics License Application For Prolia® (Denosumab) In Glucocorticoid-Induced Osteoporosis.2017 [February 17, 2018]; Available from: https://www.amgen.com/media/news-releases/2017/10/fda-accepts-supplemental-biologics-license-application-for-prolia-denosumab-in-glucocorticoidinduced-osteoporosis/.
  300. Yanbeiy ZA, Hansen KE. Denosumab in the treatment of glucocorticoid-induced osteoporosis: a systematic review and meta-analysis. Drug Des Devel Ther. 2019;13:2843-52.
  301. Yamaguchi Y, Morita T, Kumanogoh A. The therapeutic efficacy of denosumab for the loss of bone mineral density in glucocorticoid-induced osteoporosis: a meta-analysis. Rheumatol Adv Pract. 2020;4(1):rkaa008.
  302. Saag KG, McDermott MT, Adachi J, Lems W, Lane NE, Geusens P, et al. The Effect of Discontinuing Denosumab in Patients With Rheumatoid Arthritis Treated With Glucocorticoids. Arthritis Rheumatol. 2021 Sep 17.
  303. Hayes KN, Baschant U, Hauser B, Burden AM, Winter EM. When to Start and Stop Bone-Protecting Medication for Preventing Glucocorticoid-Induced Osteoporosis. Front Endocrinol (Lausanne). 2021;12:782118.
  304. Lane NE, Sanchez S, Modin GW, Genant HK, Pierini E, Arnaud CD. Bone mass continues to increase at the hip after parathyroid hormone treatment is discontinued in glucocorticoid-induced osteoporosis: results of a randomized controlled clinical trial. J Bone Miner Res. 2000 May;15(5):944-51.
  305. Lane NE, Sanchez S, Modin GW, Genant HK, Pierini E, Arnaud CD. Parathyroid hormone treatment can reverse corticosteroid-induced osteoporosis. Results of a randomized controlled clinical trial. J Clin Invest. 1998 Oct 15;102(8):1627-33.
  306. Devogelaer JP, Adler RA, Recknor C, See K, Warner MR, Wong M, et al. Baseline glucocorticoid dose and bone mineral density response with teriparatide or alendronate therapy in patients with glucocorticoid-induced osteoporosis. J Rheumatol. 2010;37:141-8.
  307. Saag KG, Zanchetta JR, Devogelaer JP, Adler RA, Eastell R, See K, et al. Effects of teriparatide versus alendronate for treating glucocorticoid-induced osteoporosis: thirty-six-month results of a randomized, double-blind, controlled trial. Arthritis Rheum. 2009;60:3346-55.
  308. Summey BT, Yosipovitch G. Glucocorticoid-induced bone loss in dermatologic patients: an update. Arch Dermatol. 2006 Jan;142(1):82-90.
  309. Hodsman AB, Bauer DC, Dempster D. Parathyroid hormone and teriparatide for the treatment of osteoporosis: a review of the evidence and suggested guidelines for its use. Endocr Rev. 2005;26:688-703.
  310. Li W, Chen W, Lin Y. The Efficacy of Parathyroid Hormone Analogues in Combination With Bisphosphonates for the Treatment of Osteoporosis: A Meta-Analysis of Randomized Controlled Trials. Medicine. 2015;94(38):e1156.
  311. Rittmaster RS, Bolognese M, Ettinger MP. Enhancement of bone mass in osteoporotic women with parathyroid hormone followed by alendronate. J Clin Endocrinol Metab. 2000;85:2129-34.
  312. Lems WF, Jacobs WG, Bijlsma JW, Croone A, Haanen HC, Houben HH, et al. Effect of sodium fluoride on the prevention of corticosteroid-induced osteoporosis. Osteoporos Int. 1997;7(6):575-82.
  313. Lems WF, Jacobs JW, Bijlsma JW, van Veen GJ, Houben HH, Haanen HC, et al. Is addition of sodium fluoride to cyclical etidronate beneficial in the treatment of corticosteroid induced osteoporosis? Ann Rheum Dis. 1997 Jun;56(6):357-63.
  314. Adami S, Fossaluzza V, Rossini M, Bertoldo F, Gatti D, Zamberlan N, et al. The prevention of corticosteroid-induced osteoporosis with nandrolone decanoate. Bone Miner. 1991 Oct;15(1):73-81.
  315. Grecu EO, Weinshelbaum A, Simmons R. Effective therapy of glucocorticoid-induced osteoporosis with medroxyprogesterone acetate. Calcif Tissue Int. 1990 May;46(5):294-9.
  316. Lukert BP, Johnson BE, Robinson RG. Estrogen and progesterone replacement therapy reduces glucocorticoid-induced bone loss. J Bone Miner Res. 1992 Sep;7(9):1063-9.
  317. Hall GM, Daniels M, Doyle DV, Spector TD. Effect of hormone replacement therapy on bone mass in rheumatoid arthritis patients treated with and without steroids. Arthritis Rheum. 1994 Oct;37(10):1499-505.
  318. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002 Jul 17;288(3):321-33.
  319. Reid IR, Wattie DJ, Evans MC, Stapleton JP. Testosterone therapy in glucocorticoid-treated men. Arch Intern Med. 1996 Jun 10;156(11):1173-7.
  320. Gruenewald DA, Matsumoto AM. Testosterone Supplementation Therapy for Older Men: Potential Benefits and Risks. J Am Geriatr Soc. 2003;51:101-15.
  321. Oettel M. The endocrine pharmacology of testosterone therapy in men. Naturwissenschaften. 2004 Feb;91(2):66-76.
  322. Hafez B, Hafez ES. Andropause: endocrinology, erectile dysfunction, and prostate pathophysiology. Arch Androl. 2004 Mar-Apr;50(2):45-68.
  323. Rhoden EL, Morgentaler A. Risks of testosterone-replacement therapy and recommendations for monitoring. N Engl J Med. 2004 Jan 29;350(5):482-92.
  324. Fritz PC, Ward WE, Atkinson SA, Tenenbaum HC. Tamoxifen attenuates the effects of exogenous glucocorticoid on bone formation and growth in piglets. Endocrinology. 1998 Aug;139(8):3399-403.
  325. Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, et al. 2017 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Care Res (Hoboken). 2017 Aug;69(8):1095-110.
  326. Delmas PD, Bjarnason NH, Mitlak BH, Ravoux AC, Shah AS, Huster WJ, et al. Effects of raloxifene on bone mineral density, serum cholesterol concentrations, and uterine endometrium in postmenopausal women. N Engl J Med. 1997 Dec 4;337(23):1641-7.
  327. Ettinger B, Black DM, Mitlak BH, Knickerbocker RK, Nickelsen T, Genant HK, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA. 1999 Aug 18;282(7):637-45.
  328. Rizzoli R, Biver E. Glucocorticoid-induced osteoporosis: who to treat with what agent? Nat Rev Rheumatol 2015;11:98-109.
  329. Radius. FDA Approves Radius Health's TYMLOS™ (abaloparatide), a Bone Building Agent for the Treatment of Postmenopausal Women with Osteoporosis at High Risk for Fracture.2017 [February 17, 2018]; Available from: http://investors.radiuspharm.com/releasedetail.cfm?releaseid=1023557.
  330. Shakeri A, Adanty C. Romosozumab (sclerostin monoclonal antibody) for the treatment of osteoporosis in postmenopausal women: A review. J Popul Ther Clin Pharmacol. 2020 Jan 6;27(1):e25-e31.
  331. Brent MB. Abaloparatide: A review of preclinical and clinical studies. Eur J Pharmacol. 2021 Oct 15;909:174409.
  332. Hauser B, Alonso N, Riches PL. Review of Current Real-World Experience with Teriparatide as Treatment of Osteoporosis in Different Patient Groups. J Clin Med. 2021 Apr 1;10(7).
  333. Yao W, Dai W, Jiang L, Lay EYA, Zhong Z, Ritchie RO, et al. Sclerostin-antibody treatment of glucocorticoid-induced osteoporosis maintained bone mass and strength. Osteoporosis Int. 2016;27(1):283-94.
  334. Devogelaer JP. New perspectives in the management of glucocorticoid-induced osteoporosis. European Musculuskeletal Review 2010;5:40-3.
  335. Sundahl N, Bridelance J, Libert C, De Bosscher K, Beck IM. Selective glucocorticoid receptor modulation: New directions with non-steroidal scaffolds. Pharmacol Ther. 2015 Aug;152:28-41.
  336. Resche-Rigon M, Gronemeyer H. Therapeutic potential of selective modulators of nuclear receptor action. Curr Opin Chem Biol. 1998 Aug;2(4):501-7.
  337. Fardellone P, Séjourné A, Paccou J, Goëb V. Bone Remodelling Markers in Rheumatoid Arthritis. Mediators of Inflammation. 2014;2014:484280.
  338. Gifre L, Ruiz-Gaspà S, Monegal A, Nomdedeu B, Filella X, Guañabens N, et al. Effect of glucocorticoid treatment on Wnt signalling antagonists (sclerostin and Dkk-1) and their relationship with bone turnover. Bone. 2013;57:272-6.
  339. Leong GM, Mercado-Asis LB, Reynolds JC, Hill SC, Oldfield EH, Chrousos GP. The effect of Cushing's disease on bone mineral density, body composition, growth, and puberty: a report of an identical adolescent twin pair. J Clin Endocrinol Metab. 1996 May;81(5):1905-11.
  340. Catargi B, Tabarin A, Basse-Cathalinat B, Ducassou D, Roger P. [Development of bone mineral density after cure of Cushing's syndrome]. Ann Endocrinol (Paris). 1996;57(3):203-8.
  341. Manning PJ, Evans MC, Reid IR. Normal bone mineral density following cure of Cushing's syndrome. Clin Endocrinol (Oxf). 1992 Mar;36(3):229-34.
  342. Rizzato G, Montemurro L. Reversibility of exogenous corticosteroid-induced bone loss. Eur Respir J. 1993 Jan;6(1):116-9.

 

 

Lipid and Lipoprotein Levels in Patients with Covid-19 Infections

ABSTRACT

Numerous studies have observed a decrease in total cholesterol, LDL-C, HDL-C, and apolipoprotein B and A-I levels in patients with COVID-19 infections, similar to what is observed with other infections. In most studies the decrease in LDL-C and/or HDL-C was more profound the greater the severity of the illness. LDL-C and HDL-C levels were inversely correlated with C-reactive protein (CRP) levels i.e., the lower the LDL-C or HDL-C level the higher the CRP levels. Patients with low HDL-C and/or LDL-C levels at admission to the hospital were at an increased risk of developing severe disease compared to patients with high levels. With recovery from COVID-19 infections the serum lipid levels return towards levels present prior to infection. In patients that failed to survive, total cholesterol, LDL-C, and HDL-C levels were lower at admission to the hospital and continued to decline during the hospitalization. In patients with COVID-19 infections the serum triglyceride levels were variable. Lipoprotein (a) levels increase during COVID-19 infections. Several studies using the UK Biobank and other databases have shown that low HDL-C and apolipoprotein A-I levels measured many years prior to COVID-19 infections were associated with an increased risk of COVID-19 infections and death from infection while LDL-C, apolipoprotein B, lipoprotein (a), and triglyceride levels were not consistently found to be significantly associated with an increased risk. A 10 mg/dl increase in HDL-C or apolipoprotein A1 levels was associated with ∼10% reduced risk of COVID-19 infection. It should be noted that these observations are subject to the caveats of confounding variables and reverse causation effecting the results. Several studies have found that homozygosity for apolipoprotein E4/4 is associated with a 2-3- fold increased risk of COVID-19 infections and this increase was not due to dementia or Alzheimer's disease. During the COVID-19 pandemic, diet, exercise, and lipid lowering therapy should be continued. For those who become symptomatic, lipid lowering therapy, if feasible, should also be continued throughout the duration of the illness. Individuals who are naïve to treatment but for whom lipid lowering therapy is indicated should be started on treatment. Whether lipid lowering drugs have beneficial effects when given prior to or during COVID-19 infections is uncertain but randomized controlled studies are in progress. In patients with severe symptoms of COVID-19 who are too ill to take oral medications, lipid lowering medications may be temporarily suspended. Medications should be re-started when the patient has recovered and able to take oral medications. One needs to be aware that certain drugs that are used to treat COVID-19 infections may interact with lipid lowering drugs. Remdesivir and Paxlovid (nirmatrelvir and ritonavir) are metabolized by the Cyp3A4 pathway and statins that are also metabolized by this pathway should be avoided (atorvastatin, simvastatin, and lovastatin). Because drug therapy for patients with COVID-19 infections is rapidly evolving one needs to be alert for potential drug interactions.  

 

INTRODUCTION 

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has resulted in a world-wide pandemic. The infection is spread through large respiratory droplets and fine respiratory aerosols. The majority of COVID-19 infections are either asymptomatic or result in only mild disease but in a substantial proportion of patients the infection leads to a respiratory illness requiring hospital care and respiratory support, which can have a fatal outcome. Older age, male gender, obesity, diabetes, cardiovascular disease, and hypertension are some of the pre-existing factors that increase the risk of severe infection and death. As of March 15, 2022, there have been over 6 million deaths worldwide according to the John Hopkins Corona Virus Resource Center.

LIPID ABNORMALITIES IN PATIENTS WITH COVID-19 INFECTIONS

Background

 

Patients with a variety of different infections (gram positive bacterial, gram negative bacterial, viral, tuberculosis, parasites) have similar alterations in plasma lipid levels. Specifically, total cholesterol, LDL-C, and HDL-C levels are decreased while plasma triglyceride levels may be elevated or inappropriately normal for the poor nutritional status (1-12). Apolipoprotein A-I, A-II, and B levels are also reduced (1,7,8). HIV, Epstein-Barr virus, and Dengue fever are viral infections that demonstrate these lipid alterations (13-15). The alterations in lipids correlate with the severity of the underlying infection i.e., the more severe the infection the more severe the alterations in lipid and lipoprotein levels (16-18). During recovery from the infection plasma lipid and lipoprotein abnormalities return towards levels present prior to infection. Of note studies have demonstrated that the degree of reduction in total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I are predictive of mortality in patients with severe sepsis (19-26).

Studies in Patients with COVID-19 

Numerous studies have reported a decrease in total cholesterol, LDL-C, HDL-C, apolipoprotein A-I, and apolipoprotein B levels and variable changes in triglycerides in patients with COVID 19 infections (27-43). An NMR analysis in patients with severe COVID-19 infections revealed a decrease in HDL particles particularly low numbers of small HDL particles and a predominance of small LDL particles compared to larger LDL particles (44). In addition to a decrease in HDL levels changes in HDL protein concentrations occur with decreased apolipoprotein A-I, apolipoprotein A-II, pulmonary surfactant-associated protein B, and paraoxonase and increased serum amyloid A and alpha-1 antitrypsin (34,45). With recovery from the acute COVID-19 infection lipid levels return towards levels present prior to infection (27-29,46,47). LDL-C and HDL-C levels were inversely correlated with C-reactive protein (CRP) levels i.e., the lower the LDL-C or HDL-C level the higher the CRP levels (27,28,31,48,49). The lower the HDL-C and LDL-C levels the greater the severity of the COVID-19 infection (28,30-33,36-38,41,47,48,50). Low LDL-C and/or HDL-C levels at admission to the hospital predicted an increased risk of developing severe disease and mortality and in these very ill patients, lipid levels declined during the hospitalization (27,37,38,40,46,48,50,51). In a meta-analysis of 19 studies and a meta-analysis of 22 studies decreased levels of total cholesterol, HDL-C, and LDL-C was associated with severity and mortality in COVID-19 patients (52,53).

 

In patients with COVID-19 infections serum triglyceride levels were variable. This is likely due to the decreased food intake that commonly occurs in ill patients resulting in a decrease in triglyceride levels. Additionally, the timing of when blood samples were obtained, the use of medications that may affect triglyceride levels (for example glucocorticoids or propofol), or the development of disorders that effect triglyceride levels (for example poorly controlled diabetes) could have confounded the triglyceride results. Severe hypertriglyceridemia (triglycerides > 500mg/dL) occurred in 33.3% of patients with COVID-19 associated acute respiratory distress syndrome treated with propofol compared to only 4.3% of patients with non-COVID-19 acute respiratory distress treated with propofol (54). Of note it has been reported that serum triglyceride levels were elevated in patients with mild or severe infections but not in patients with critical illness (respiratory or multiple organ failure and septic shock) (31). In contrast, a study reported that triglyceride levels were higher in patients that died from COVID-19 compared to patients that were critically ill or non-critically ill (50). In another study a severe outcome was associated with lower HDL-C levels and higher triglyceride levels (55). However, a meta-analysis did not find that triglyceride levels were associated with disease severity in patients with COVID-19 (53). NMR analysis in patients with severe COVID-19 infections revealed an increase in triglyceride rich lipoprotein particles primarily due to an increase in the small and very small subfractions (44). Finally, a patient with a mild COVID-19 infection has been reported to develop marked hypertriglyceridemia due to transient inhibition of lipoprotein lipase activity presumably due to the development of autoantibodies against lipoprotein lipase similar to what has been reported in patients with autoimmune disorders such as systemic lupus erythematosus (56).

 

Lipoprotein (a) levels increase during COVID-19 and appear to be associated with an increased risk of venous thromboembolism (57). It had been hypothesized that an increase in Lp(a) could contribute to some of the clinical abnormalities, such as thrombosis, seen during severe COVID-19 infections and these results support that hypothesis (58).  

 

The potential mechanisms by which infections and inflammation alter lipid and lipoprotein levels and the consequences of these alterations are discussed in the Endotext chapter entitled “The Effect of Inflammation and Infection on Lipids and Lipoproteins” (59).

 

Table 1. Effect of COVID-19 Infection on Lipid and Lipoprotein Levels

Triglycerides- Variable but tend to be increased

Total cholesterol- Decreased

HDL-C- Decreased

LDL-C- Decreased

Small dense LDL- Increased

Lp(a)- Increased

Apolipoprotein A-I- Decreased

Apolipoprotein B- Decreased

 

DO PRE-INFECTION LIPID LEVELS PREDISPOSE TO SEVERE COVID-19 INFECTION?

Background

Numerous observational studies have suggested that low LDL-C and/or HDL-C levels increase the risk of developing infections and sepsis (60-72). Of course, it must be recognized that confounding variables could account for this association. For example, unrecognized disease (for example pulmonary or gastrointestinal disorders) could result in decreased HDL-C and LDL-C levels and independently also increase the risk of infections and sepsis.

 

Studies employing a genetic approach to epidemiology, which reduces the risk of confounding variables and reverse causation, have been used to investigate the relationship of lipid levels with the risk of infections and sepsis. In a study by Madsen and colleagues using two common variants in the genes encoding hepatic lipase and cholesteryl ester transfer protein that regulate HDL-C levels found in 97,166 individuals from the Copenhagen General Population Study that low HDL-C levels increased the risk of infection supporting the observational studies that low HDL-C levels increase the risk of infection (66). In studies by Walley and colleagues HMGCoA reductase and PCSK9 genetic variants that decrease LDL-C levels genetically were not associated with an increased mortality from sepsis suggesting that the observational studies linking low LDL-C with sepsis may have been due to confounding variables (70). In support of this contention a study demonstrated that low LDL-C levels were significantly associated with increased risk of sepsis and admission to intensive care unit, however, this association was found to be due to comorbidities (73). Finally, Trinder and colleagues using the UK Biobank data base (407,558 individuals) demonstrated that elevated levels of HDL-C and LDL-C were associated with a reduced risk of infectious disease related hospitalizations similar to prior observational studies while elevated levels of triglycerides were associated with increased risk of infectious disease related hospitalizations (74). However, this study also employed a genetic approach and found that for genetically determined lipid levels, only increased HDL-C levels were significantly associated with a reduced risk of hospitalizations for infectious disease and mortality from sepsis suggesting that HDL could be causally related to infections (74). Taken together these studies demonstrate that low LDL-C levels that are associated with an increased risk of infections are not likely to be a causal association while the low HDL-C levels that are associated with an increased risk of infection appears to be causal.

 

This protective effect of HDL could be due to HDL particles binding lipopolysaccharide and lipoteichoic acid, compounds that mediate the excessive immune activation in sepsis or to the immunomodulatory, antithrombotic, and antioxidant properties of HDL (6,75). Additionally, HDL may have direct effects on viruses that decrease their infectivity by direct viral inactivation, interference with viral entry into the cell, or inhibition of virus-induced cell fusion (76-79). Finally, HDL has an antiviral effect against SARS-CoV-2 (COVID-19) (80). 

COVID-19 Infections

Several studies using the UK Biobank and other databases have shown that elevated HDL-C and apolipoprotein AI levels measured many years prior to COVID-19 infections were associated with a reduced risk of COVID-19 infections while LDL-C, Apo B, lipoprotein (a) and triglyceride levels were not consistently found to be significantly associated with an increased risk (81-89). Hilser and colleagues found that a 10 mg/dl increase in HDL-C or apolipoprotein A1 levels were associated with ∼10% reduced risk of COVID-19 infection (82). In addition, an increased risk of death from COVID-19 infections was also inversely related to HDL-C and apolipoprotein A1 levels (82). Thus, there is consistent evidence that HDL-C and apolipoprotein A1 levels measured many years prior to COVID-19 play a role in determining the risk of developing COVID-19 infections. It should be noted that these were not genetic based analysis so these observations, as discussed above, are subject to the caveats of confounding variables and reverse causation effecting the results.

 

Aung et al reported that genetically higher exposure to LDL-C was related to increased risk of COVID-19 (84) and Zhang and colleagues reported that genetically determined higher total cholesterol and apolipoprotein B levels might increase susceptibility for COVID-19 (90). However, other studies found no evidence supporting an association of genetically induced increases in LDL-C and apolipoprotein B levels with an increased risk for severe COVID-19 infections (82,91-93). Hilser et al was also unable to demonstrate a link between genetically determined HDL-C and triglyceride levels and COVID-19 infection risk (82). Others have also not been able to demonstrate a genetic link of HDL-C, or triglyceride levels with COVID-19 infections (93). However, a Mendelian randomization study found a causal effect of higher serum triglyceride levels on a greater risk of COVID-19 severity (92). Lp(a) genetic risk scores were similar in COVID-19 infected patient and controls (89). Given the variability of results additional studies are required to determine whether LDL-C, apolipoprotein B, apolipoprotein A-I, HDL-C, or triglyceride levels have a causal role in determining the risk or severity of COVID-19 infections.

 

Several studies have found that homozygosity for apolipoprotein E4/4 is associated with a 2-3- fold increased risk of COVID-19 infections and this increase was not due to dementia or Alzheimer's disease (82,94,95). Interestingly, in patients with HIV, apolipoprotein E4/4 is associated with an accelerated disease progression and death compared with apolipoprotein E3/3 (96). Additionally, individuals who are apolipoprotein E3/4 have an increased inflammatory response to toll receptor ligands compared with patients who are apolipoprotein E3/3 (97). The mechanisms by which apolipoprotein E4/4 increases the risk of COVID 19 infections remains to be elucidated.

LIPID LOWERING DRUGS and COVID-19 INFECTIONS  

Detailed information on cholesterol and triglyceride lowering medications is provided in the Endotext chapters entitled “Cholesterol Lowering Drugs” and Triglyceride Lowering Drugs” (98,99). Only information that is of unique importance with regards to lipid lowering drugs and COVID-19 infections will be discussed in this chapter. For a detailed review of lipid lowering drug therapy in COVID-19 patients see “Managing hyperlipidaemia in patients with COVID-19 and during its pandemic: An expert panel position statement from HEART UK” (100).

Statins

Statins have pleiotropic effects, including decreasing inflammation and oxidative stress, improving endothelial function and immune response, and inhibiting the activation of coagulation cascade, all of which could be beneficial in patients infected with SARS-CoV-2 (101,102). In contrast to these potentially beneficial effects, statins upregulate the ACE2 receptor, the receptor that the SARS-CoV-2 virus uses to enter cells, which could potentially increase the severity of the infection (101,102).

 

Because of the possibility that statins could have beneficial effects on COVID-19 infections there have been a large number of observational studies comparing the severity of disease and/or mortality in patients taking statins vs. patients not taking stains. Most meta-analyses have found that statins reduce severity of disease and/or mortality (103-108). It should be appreciated that these observation studies have potential flaws and cannot definitively prove that statins are beneficial in COVID-19 infections. In a single randomized trial statin therapy did not reduce disease severity or mortality compared to placebo (109). It is worth noting that a meta-analysis of 7 randomized trials with 1720 patients examining the effect of statins in sepsis (not COVID-19 infections) did not demonstrate any benefit compared to placebo (110). However, the absence of harm from statin therapy in the majority of the COVID-19 observational studies and in the single randomized trial makes it reasonable to continue statin therapy in COVID-19 infected patients for their well-recognized benefits on cardiovascular disease.

 

One needs to be aware of potential drug interactions with statins and some of the drugs used to treat COVID-19 infections (see table 3) (100). Remdesivir is metabolized by the Cyp3A4 pathway and statins that are also metabolized by this pathway should be avoided (atorvastatin, simvastatin, and lovastatin) (100). With the antiretroviral drug, nirmatrelvir and ritonavir (Paxlovid), it is recommended to avoid statins metabolized by the Cyp3A4 pathway (atorvastatin, simvastatin, and lovastatin) and use low dose rosuvastatin therapy (100). Tocilizumab by inhibiting IL-6 can increase CYP3A4 activity thereby reducing the LDL-C lowering effect of atorvastatin, simvastatin, and lovastatin.Additionally, certain drugs (for example nirmatrelvir and ritonavir) that treat COVID-19 are only used for a short period of time and temporarily stopping statin therapy may be a reasonable approach.

Ezetimibe

A single study reported that patients taking ezetimibe had significantly reduced odds for SARS-CoV-2 hospitalization (OR=0.513, 95% CI 0.375-0.688) (111). The mechanism for this effect is not clear and additional studies are required.

PCSK9 Inhibitors, Evinacumab, and Bempedoic Acid

There is no information with regards to COVID-19 Infections and these cholesterol lowering drugs.

Bile Acid Sequestrants

There is no information with regards to COVID-19 Infections. Because bile acid sequestrants can bind drugs in the GI tract and decrease their absorption, care must be taken when using other oral medications in patients taking bile acid sequestrants.

Fibrates

Fibrates have anti-inflammatory properties (112). In a cohort study fenofibrate did not reduce the severity of COVID-19 infections (113). In patients treated with tocilizumab the use of fibrates should be suspended (100).

Omega-3-Fatty Acids

Omega-3-fatty acids have anti-inflammatory properties (114). In a randomized trial 2 grams per day of Docosahexaenoic acid (DHA) + Eicosapentaenoic acid (EPA) for 2 weeks improved the clinical symptoms of COVID-19 infection and reduced markers of inflammation (C-reactive protein and erythrocyte sedimentation rate) (115). In another randomized trial the administration of 400mg EPA and 200mg DHA per day decreased severity and improved survival in critically ill patients with COVID-19 infection (116). Additional studies are needed to confirm these intriguing results.  

Niacin

There is no information with regards to COVID-19 Infections.

Lomitapide

Lomitapide is metabolized in the liver through CYP3A4 and lomitapide is also an inhibitor of CYP3A4 (100). Therefore, one needs to be concerned about potential drug interactions.  

Volanesorsen

The major side effect of volanesorsen is thrombocytopenia. Studies have suggested that low platelet levels are associated with an increased risk of severe disease and mortality in patients with COVID-19 infections (100). Therefore, it is recommended that volanesorsen therapy be discontinued in patients infected with COVID-19 until the infection resolves.

Future Studies

There are a large number of on-going randomized trials of the effect of lipid lowering drugs in COVID-19 infections (table 2) (117). For details on these trials see reference (117).

 

Table 2. On-Going Randomized Trials of Lipid Lowering Drugs

 

Number of RCTs

Total Number of Patients

Statins

17

18,215

Fibrates

3

1,050

Niacin

5

1,200

Omega-3 fatty acids

14

21,898

RCTs- randomized controlled trials

 

Interaction Between Drugs to Treat COVID-19 and Lipid Lowering Drugs

The effect of various drugs that are used to treat COVID-19 infections and lipid lowering drugs are shown in table 3. Because drug therapy for patients with COVID-19 infections is rapidly evolving one needs to be alert for the use of new drugs with potential drug interactions.

Table 3. Interactions Between Drugs to Treat Covid-19 and Lipid Lowering Drugs

Covid-19 Drugs

Drug Interactions

Nirmatrelvir and Ritonavir (Paxlovid)

Contraindicated with drugs that are highly dependent on CYP3A for clearance and thereby increases levels of lovastatin, simvastatin, and atorvastatin. Also increases levels of rosuvastatin by a different mechanism but can use low dose.

Monoclonal antibodies against spike protein

No drug interactions

Remdesivir (Veklury)

Metabolized by the Cyp3A4 pathway and therefore should avoid lovastatin, simvastatin, and atorvastatin.

Molnupiravir (Movfor)

No drug interactions

Baricitinib (Olumiant)

No drug interactions

Tocilizumab (Actemra)

Deceasing IL-6 can upregulate CYP3A and reduce the activity of lovastatin, simvastatin, and atorvastatin.

Glucocorticoids

No drug interactions

MANAGEMENT OF HYPERLIPIDEMIA DURING THE COVID-19 PANDEMIC

During the COVID-19 pandemic diet and exercise should be continued and there is no reason to stop lipid lowering therapy. Patients on lipid lowering therapy should continue to take their medications and patients who have indications for starting lipid lowering therapy should be started on therapy (100). In patients who are asymptomatic or have only mild symptoms of COVID-19 they should also continue their lipid lowering medications (100). This is particular important as studies have shown an association with influenza and other respiratory infections and myocardial infarctions (118-120). In patients with severe symptoms of COVID-19 who are too ill to take oral medications, lipid lowering medications may be temporarily suspended (100). Medications should be re-started when the patient has recovered and is able to take oral medications.

 

Liver function test abnormalities are frequently observed in patients with severe COVID-19 infections. If the alanine transaminase (ALT) or aspartate transaminase (AST) is greater than 3 times the upper limit of normal lipid lowering therapy should be stopped (100). Creatine kinase measurements should be considered when clinically indicated and in patients who are critically ill. It is recommended that statin therapy be stopped if creatine kinase rises 10-fold (generally to levels above 2000 IU/L) in asymptomatic patients or at a lower level of 5-fold upper limit of normal in symptomatic patients (100).

REFERENCES

  1. Alvarez C, Ramos A. Lipids, lipoproteins, and apoproteins in serum during infection. Clin Chem 1986; 32:142-145
  2. Cappi SB, Noritomi DT, Velasco IT, Curi R, Loureiro TC, Soriano FG. Dyslipidemia: a prospective controlled randomized trial of intensive glycemic control in sepsis. Intensive Care Med 2012; 38:634-641
  3. Gallin JI, Kaye D, O'Leary WM. Serum lipids in infection. N Engl J Med 1969; 281:1081-1086
  4. Gordon BR, Parker TS, Levine DM, Saal SD, Wang JC, Sloan BJ, Barie PS, Rubin AL. Low lipid concentrations in critical illness: implications for preventing and treating endotoxemia. Crit Care Med 1996; 24:584-589
  5. Kerttula Y, Weber TH. Serum lipids in viral and bacterial meningitis. Scand J Infect Dis 1986; 18:211-215
  6. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res 2004; 45:1169-1196
  7. Sammalkorpi K, Valtonen V, Kerttula Y, Nikkila E, Taskinen MR. Changes in serum lipoprotein pattern induced by acute infections. Metabolism 1988; 37:859-865
  8. van Leeuwen HJ, Heezius EC, Dallinga GM, van Strijp JA, Verhoef J, van Kessel KP. Lipoprotein metabolism in patients with severe sepsis. Crit Care Med 2003; 31:1359-1366
  9. Visser BJ, Wieten RW, Nagel IM, Grobusch MP. Serum lipids and lipoproteins in malaria--a systematic review and meta-analysis. Malar J 2013; 12:442
  10. Sahin F, Yildiz P. Distinctive biochemical changes in pulmonary tuberculosis and pneumonia. Arch Med Sci 2013; 9:656-661
  11. Apostolou F, Gazi IF, Kostoula A, Tellis CC, Tselepis AD, Elisaf M, Liberopoulos EN. Persistence of an atherogenic lipid profile after treatment of acute infection with Brucella. J Lipid Res 2009; 50:2532-2539
  12. Gazi IF, Apostolou FA, Liberopoulos EN, Filippatos TD, Tellis CC, Elisaf MS, Tselepis AD. Leptospirosis is associated with markedly increased triglycerides and small dense low-density lipoprotein and decreased high-density lipoprotein. Lipids 2011; 46:953-960
  13. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab 1992; 74:1045-1052
  14. Marin-Palma D, Sirois CM, Urcuqui-Inchima S, Hernandez JC. Inflammatory status and severity of disease in dengue patients are associated with lipoprotein alterations. PLoS One 2019; 14:e0214245
  15. Apostolou F, Gazi IF, Lagos K, Tellis CC, Tselepis AD, Liberopoulos EN, Elisaf M. Acute infection with Epstein-Barr virus is associated with atherogenic lipid changes. Atherosclerosis 2010; 212:607-613
  16. Deniz O, Gumus S, Yaman H, Ciftci F, Ors F, Cakir E, Tozkoparan E, Bilgic H, Ekiz K. Serum total cholesterol, HDL-C and LDL-C concentrations significantly correlate with the radiological extent of disease and the degree of smear positivity in patients with pulmonary tuberculosis. Clin Biochem 2007; 40:162-166
  17. Deniz O, Tozkoparan E, Yaman H, Cakir E, Gumus S, Ozcan O, Bozlar U, Bilgi C, Bilgic H, Ekiz K. Serum HDL-C levels, log (TG/HDL-C) values and serum total cholesterol/HDL-C ratios significantly correlate with radiological extent of disease in patients with community-acquired pneumonia. Clin Biochem 2006; 39:287-292
  18. El-Sadr WM, Mullin CM, Carr A, Gibert C, Rappoport C, Visnegarwala F, Grunfeld C, Raghavan SS. Effects of HIV disease on lipid, glucose and insulin levels: results from a large antiretroviral-naive cohort. HIV Med 2005; 6:114-121
  19. Barlage S, Gnewuch C, Liebisch G, Wolf Z, Audebert FX, Gluck T, Frohlich D, Kramer BK, Rothe G, Schmitz G. Changes in HDL-associated apolipoproteins relate to mortality in human sepsis and correlate to monocyte and platelet activation. Intensive Care Med 2009; 35:1877-1885
  20. Chien JY, Jerng JS, Yu CJ, Yang PC. Low serum level of high-density lipoprotein cholesterol is a poor prognostic factor for severe sepsis. Crit Care Med 2005; 33:1688-1693
  21. Gruber M, Christ-Crain M, Stolz D, Keller U, Muller C, Bingisser R, Tamm M, Mueller B, Schuetz P. Prognostic impact of plasma lipids in patients with lower respiratory tract infections - an observational study. Swiss Med Wkly 2009; 139:166-172
  22. Lekkou A, Mouzaki A, Siagris D, Ravani I, Gogos CA. Serum lipid profile, cytokine production, and clinical outcome in patients with severe sepsis. J Crit Care 2014; 29:723-727
  23. Chien YF, Chen CY, Hsu CL, Chen KY, Yu CJ. Decreased serum level of lipoprotein cholesterol is a poor prognostic factor for patients with severe community-acquired pneumonia that required intensive care unit admission. J Crit Care 2015; 30:506-510
  24. Cirstea M, Walley KR, Russell JA, Brunham LR, Genga KR, Boyd JH. Decreased high-density lipoprotein cholesterol level is an early prognostic marker for organ dysfunction and death in patients with suspected sepsis. J Crit Care 2017; 38:289-294
  25. Trinder M, Genga KR, Kong HJ, Blauw LL, Lo C, Li X, Cirstea M, Wang Y, Rensen PCN, Russell JA, Walley KR, Boyd JH, Brunham LR. Cholesteryl Ester Transfer Protein Influences High-Density Lipoprotein Levels and Survival in Sepsis. Am J Respir Crit Care Med 2019; 199:854-862
  26. Guirgis FW, Black LP, Henson M, Labilloy G, Smotherman C, Hopson C, Tfirn I, DeVos EL, Leeuwenburgh C, Moldawer L, Datta S, Brusko TM, Hester A, Bertrand A, Grijalva V, Arango-Esterhay A, Moore FA, Reddy ST. A hypolipoprotein sepsis phenotype indicates reduced lipoprotein antioxidant capacity, increased endothelial dysfunction and organ failure, and worse clinical outcomes. Crit Care 2021; 25:341
  27. Fan J, Wang H, Ye G, Cao X, Xu X, Tan W, Zhang Y. Letter to the Editor: Low-density lipoprotein is a potential predictor of poor prognosis in patients with coronavirus disease 2019. Metabolism 2020; 107:154243
  28. Hu X, Chen D, Wu L, He G, Ye W. Declined serum high density lipoprotein cholesterol is associated with the severity of COVID-19 infection. Clin Chim Acta 2020; 510:105-110
  29. Tanaka S, De Tymowski C, Assadi M, Zappella N, Jean-Baptiste S, Robert T, Peoc'h K, Lortat-Jacob B, Fontaine L, Bouzid D, Tran-Dinh A, Tashk P, Meilhac O, Montravers P. Lipoprotein concentrations over time in the intensive care unit COVID-19 patients: Results from the ApoCOVID study. PLoS One 2020; 15:e0239573
  30. Wang G, Zhang Q, Zhao X, Dong H, Wu C, Wu F, Yu B, Lv J, Zhang S, Wu G, Wu S, Wang X, Wu Y, Zhong Y. Low high-density lipoprotein level is correlated with the severity of COVID-19 patients: an observational study. Lipids Health Dis 2020; 19:204
  31. Wei X, Zeng W, Su J, Wan H, Yu X, Cao X, Tan W, Wang H. Hypolipidemia is associated with the severity of COVID-19. J Clin Lipidol 2020; 14:297-304
  32. Wang D, Li R, Wang J, Jiang Q, Gao C, Yang J, Ge L, Hu Q. Correlation analysis between disease severity and clinical and biochemical characteristics of 143 cases of COVID-19 in Wuhan, China: a descriptive study. BMC Infect Dis 2020; 20:519
  33. Zhang Q, Wei Y, Chen M, Wan Q, Chen X. Clinical analysis of risk factors for severe COVID-19 patients with type 2 diabetes. J Diabetes Complications 2020; 34:107666
  34. Begue F, Tanaka S, Mouktadi Z, Rondeau P, Veeren B, Diotel N, Tran-Dinh A, Robert T, Velia E, Mavingui P, Lagrange-Xelot M, Montravers P, Couret D, Meilhac O. Altered high-density lipoprotein composition and functions during severe COVID-19. Sci Rep 2021; 11:2291
  35. Lin L, Zhong C, Rao S, Lin H, Huang R, Chen F. Clinical characteristics of 78 cases of patients infected with coronavirus disease 2019 in Wuhan, China. Exp Ther Med 2021; 21:7
  36. Lv Z, Wang W, Qiao B, Cui X, Feng Y, Chen L, Ma Q, Liu X. The prognostic value of general laboratory testing in patients with COVID-19. J Clin Lab Anal 2020:e23668
  37. Turgay Yildirim O, Kaya S. The atherogenic index of plasma as a predictor of mortality in patients with COVID-19. Heart Lung 2021; 50:329-333
  38. Zhang B, Dong C, Li S, Song X, Wei W, Liu L. Triglyceride to High-Density Lipoprotein Cholesterol Ratio is an Important Determinant of Cardiovascular Risk and Poor Prognosis in Coronavirus Disease-19: A Retrospective Case Series Study. Diabetes Metab Syndr Obes 2020; 13:3925-3936
  39. Kimhofer T, Lodge S, Whiley L, Gray N, Loo RL, Lawler NG, Nitschke P, Bong SH, Morrison DL, Begum S, Richards T, Yeap BB, Smith C, Smith KGC, Holmes E, Nicholson JK. Integrative Modeling of Quantitative Plasma Lipoprotein, Metabolic, and Amino Acid Data Reveals a Multiorgan Pathological Signature of SARS-CoV-2 Infection. J Proteome Res 2020; 19:4442-4454
  40. Ressaire Q, Dudoignon E, Moreno N, Coutrot M, Depret F. Low total cholesterol blood level is correlated with pulmonary severity in COVID-19 critical ill patients. Anaesth Crit Care Pain Med 2020; 39:733-735
  41. Sampedro-Nunez M, Aguirre-Moreno N, Garcia-Fraile Fraile L, Jimenez-Blanco S, Knott-Torcal C, Sanz-Martin P, Fernandez-Jimenez G, Marazuela M. Finding answers in lipid profile in COVID-19 patients. Endocrine 2021; 74:443-454
  42. Papotti B, Macchi C, Favero C, Iodice S, Adorni MP, Zimetti F, Corsini A, Aliberti S, Blasi F, Carugo S, Bollati V, Vicenzi M, Ruscica M. HDL in COVID-19 Patients: Evidence from an Italian Cross-Sectional Study. J Clin Med 2021; 10
  43. El Nekidy WS, Shatnawei A, Abdelsalam MM, Hassan M, Dajani RZ, Salem N, St John TJL, Rahman N, Hamed F, Mallat J. Hypertriglyceridemia in Critically Ill Patients With SARS-CoV-2 Infection. Ann Pharmacother 2021:10600280211038302
  44. Ballout RA, Kong H, Sampson M, Otvos JD, Cox AL, Agbor-Enoh S, Remaley AT. The NIH Lipo-COVID Study: A Pilot NMR Investigation of Lipoprotein Subfractions and Other Metabolites in Patients with Severe COVID-19. Biomedicines 2021; 9
  45. Souza Junior DR, Silva ARM, Rosa-Fernandes L, Reis LR, Alexandria G, Bhosale SD, Ghilardi FR, Dalcoquio TF, Bertolin AJ, Nicolau JC, Marinho CRF, Wrenger C, Larsen MR, Siciliano RF, Di Mascio P, Palmisano G, Ronsein GE. HDL proteome remodeling associates with COVID-19 severity. J Clin Lipidol 2021; 15:796-804
  46. Ouyang SM, Zhu HQ, Xie YN, Zou ZS, Zuo HM, Rao YW, Liu XY, Zhong B, Chen X. Temporal changes in laboratory markers of survivors and non-survivors of adult inpatients with COVID-19. BMC Infect Dis 2020; 20:952
  47. Qin C, Minghan H, Ziwen Z, Yukun L. Alteration of lipid profile and value of lipids in the prediction of the length of hospital stay in COVID-19 pneumonia patients. Food Sci Nutr 2020; 8:6144-6152
  48. Sun JT, Chen Z, Nie P, Ge H, Shen L, Yang F, Qu XL, Ying XY, Zhou Y, Wang W, Zhang M, Pu J. Lipid Profile Features and Their Associations With Disease Severity and Mortality in Patients With COVID-19. Front Cardiovasc Med 2020; 7:584987
  49. D'Ardes D, Rossi I, Bucciarelli B, Allegra M, Bianco F, Sinjari B, Marchioni M, Di Nicola M, Santilli F, Guagnano MT, Cipollone F, Bucci M. Metabolic Changes in SARS-CoV-2 Infection: Clinical Data and Molecular Hypothesis to Explain Alterations of Lipid Profile and Thyroid Function Observed in COVID-19 Patients. Life (Basel) 2021; 11
  50. Huang W, Li C, Wang Z, Wang H, Zhou N, Jiang J, Ni L, Zhang XA, Wang DW. Decreased serum albumin level indicates poor prognosis of COVID-19 patients: hepatic injury analysis from 2,623 hospitalized cases. Sci China Life Sci 2020; 63:1678-1687
  51. Aparisi A, Iglesias-Echeverria C, Ybarra-Falcon C, Cusacovich I, Uribarri A, Garcia-Gomez M, Ladron R, Fuertes R, Candela J, Tobar J, Hinojosa W, Duenas C, Gonzalez R, Nogales L, Calvo D, Carrasco-Moraleja M, San Roman JA, Amat-Santos IJ, Andaluz-Ojeda D. Low-density lipoprotein cholesterol levels are associated with poor clinical outcomes in COVID-19. Nutr Metab Cardiovasc Dis 2021; 31:2619-2627
  52. Mahat RK, Rathore V, Singh N, Singh N, Singh SK, Shah RK, Garg C. Lipid profile as an indicator of COVID-19 severity: A systematic review and meta-analysis. Clin Nutr ESPEN 2021; 45:91-101
  53. Zinellu A, Paliogiannis P, Fois AG, Solidoro P, Carru C, Mangoni AA. Cholesterol and Triglyceride Concentrations, COVID-19 Severity, and Mortality: A Systematic Review and Meta-Analysis With Meta-Regression. Front Public Health 2021; 9:705916
  54. Kenes MT, McSparron JI, Marshall VD, Renius K, Hyzy RC. Propofol-Associated Hypertriglyceridemia in Coronavirus Disease 2019 Versus Noncoronavirus Disease 2019 Acute Respiratory Distress Syndrome. Crit Care Explor 2020; 2:e0303
  55. Masana L, Correig E, Ibarretxe D, Anoro E, Arroyo JA, Jerico C, Guerrero C, Miret M, Naf S, Pardo A, Perea V, Perez-Bernalte R, Plana N, Ramirez-Montesinos R, Royuela M, Soler C, Urquizu-Padilla M, Zamora A, Pedro-Botet J, group S-Xr. Low HDL and high triglycerides predict COVID-19 severity. Sci Rep 2021; 11:7217
  56. Fijen LM, Grefhorst A, Levels JHM, Cohn DM. Severe acquired hypertriglyceridemia following COVID-19. BMJ Case Rep 2021; 14
  57. Nurmohamed NS, Collard D, Reeskamp LF, Kaiser Y, Kroon J, Tromp TR, Amsterdam UMCC-B, van den Born BH, Coppens M, Vlaar APJ, Beudel M, van de Beek D, van Es N, Moriarty PM, Tsimikas S, Stroes ESG. Lipoprotein(a), venous thromboembolism and COVID-19: A pilot study. Atherosclerosis 2022; 341:43-49
  58. Moriarty PM, Gorby LK, Stroes ES, Kastelein JP, Davidson M, Tsimikas S. Lipoprotein(a) and Its Potential Association with Thrombosis and Inflammation in COVID-19: a Testable Hypothesis. Curr Atheroscler Rep 2020; 22:48
  59. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland HJ, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  60. Grion CM, Cardoso LT, Perazolo TF, Garcia AS, Barbosa DS, Morimoto HK, Matsuo T, Carrilho AJ. Lipoproteins and CETP levels as risk factors for severe sepsis in hospitalized patients. Eur J Clin Invest 2010; 40:330-338
  61. Claxton AJ, Jacobs DR, Jr., Iribarren C, Welles SL, Sidney S, Feingold KR. Association between serum total cholesterol and HIV infection in a high-risk cohort of young men. J Acquir Immune Defic Syndr Hum Retrovirol 1998; 17:51-57
  62. Iribarren C, Jacobs DR, Jr., Sidney S, Claxton AJ, Feingold KR. Cohort study of serum total cholesterol and in-hospital incidence of infectious diseases. Epidemiol Infect 1998; 121:335-347
  63. Guirgis FW, Donnelly JP, Dodani S, Howard G, Safford MM, Levitan EB, Wang HE. Cholesterol levels and long-term rates of community-acquired sepsis. Crit Care 2016; 20:408
  64. Kaysen GA, Ye X, Raimann JG, Wang Y, Topping A, Usvyat LA, Stuard S, Canaud B, van der Sande FM, Kooman JP, Kotanko P, Monitoring Dialysis Outcomes I. Lipid levels are inversely associated with infectious and all-cause mortality: international MONDO study results. J Lipid Res 2018; 59:1519-1528
  65. Kaysen GA, Grimes B, Dalrymple LS, Chertow GM, Ishida JH, Delgado C, Segal M, Chiang J, Dwyer T, Johansen KL. Associations of lipoproteins with cardiovascular and infection-related outcomes in patients receiving hemodialysis. J Clin Lipidol 2018; 12:481-487 e414
  66. Madsen CM, Varbo A, Tybjaerg-Hansen A, Frikke-Schmidt R, Nordestgaard BG. U-shaped relationship of HDL and risk of infectious disease: two prospective population-based cohort studies. Eur Heart J 2018; 39:1181-1190
  67. Canturk NZ, Canturk Z, Okay E, Yirmibesoglu O, Eraldemir B. Risk of nosocomial infections and effects of total cholesterol, HDL cholesterol in surgical patients. Clin Nutr 2002; 21:431-436
  68. Shor R, Wainstein J, Oz D, Boaz M, Matas Z, Fux A, Halabe A. Low serum LDL cholesterol levels and the risk of fever, sepsis, and malignancy. Ann Clin Lab Sci 2007; 37:343-348
  69. Shor R, Wainstein J, Oz D, Boaz M, Matas Z, Fux A, Halabe A. Low HDL levels and the risk of death, sepsis and malignancy. Clin Res Cardiol 2008; 97:227-233
  70. Walley KR, Boyd JH, Kong HJ, Russell JA. Low Low-Density Lipoprotein Levels Are Associated With, But Do Not Causally Contribute to, Increased Mortality in Sepsis. Crit Care Med 2019; 47:463-466
  71. Delgado-Rodriguez M, Medina-Cuadros M, Martinez-Gallego G, Sillero-Arenas M. Total cholesterol, HDL-cholesterol, and risk of nosocomial infection: a prospective study in surgical patients. Infect Control Hosp Epidemiol 1997; 18:9-18
  72. Rodriguez-Sanz A, Fuentes B, Martinez-Sanchez P, Prefasi D, Martinez-Martinez M, Correas E, Diez-Tejedor E. High-density lipoprotein: a novel marker for risk of in-hospital infection in acute ischemic stroke patients? Cerebrovasc Dis 2013; 35:291-297
  73. Feng Q, Wei WQ, Chaugai S, Leon BGC, Mosley JD, Leon DAC, Jiang L, Ihegword A, Shaffer CM, Linton MF, Chung CP, Stein CM. Association Between Low-Density Lipoprotein Cholesterol Levels and Risk for Sepsis Among Patients Admitted to the Hospital With Infection. JAMA Netw Open 2019; 2:e187223
  74. Trinder M, Walley KR, Boyd JH, Brunham LR. Causal Inference for Genetically Determined Levels of High-Density Lipoprotein Cholesterol and Risk of Infectious Disease. Arterioscler Thromb Vasc Biol 2020; 40:267-278
  75. Catapano AL, Pirillo A, Bonacina F, Norata GD. HDL in innate and adaptive immunity. Cardiovasc Res 2014; 103:372-383
  76. Gordon SM, Hofmann S, Askew DS, Davidson WS. High density lipoprotein: it's not just about lipid transport anymore. Trends Endocrinol Metab 2011; 22:9-15
  77. Kane JP, Hardman DA, Dimpfl JC, Levy JA. Apolipoprotein is responsible for neutralization of xenotropic type C virus by mouse serum. Proc Natl Acad Sci U S A 1979; 76:5957-5961
  78. Srinivas RV, Birkedal B, Owens RJ, Anantharamaiah GM, Segrest JP, Compans RW. Antiviral effects of apolipoprotein A-I and its synthetic amphipathic peptide analogs. Virology 1990; 176:48-57
  79. Singh IP, Chopra AK, Coppenhaver DH, Ananatharamaiah GM, Baron S. Lipoproteins account for part of the broad non-specific antiviral activity of human serum. Antiviral Res 1999; 42:211-218
  80. Cho KH, Kim JR, Lee IC, Kwon HJ. Native High-Density Lipoproteins (HDL) with Higher Paraoxonase Exerts a Potent Antiviral Effect against SARS-CoV-2 (COVID-19), While Glycated HDL Lost the Antiviral Activity. Antioxidants (Basel) 2021; 10
  81. Scalsky RJ, Desai K, Chen YJ, O'Connell JR, Perry JA, Hong CC. Baseline Cardiometabolic Profiles and SARS-CoV-2 Risk in the UK Biobank. medRxiv 2020;
  82. Hilser JR, Han Y, Biswas S, Gukasyan J, Cai Z, Zhu R, Tang WHW, Deb A, Lusis AJ, Hartiala JA, Allayee H. Association of serum HDL-cholesterol and apolipoprotein A1 levels with risk of severe SARS-CoV-2 infection. J Lipid Res 2021; 62:100061
  83. Ho FK, Celis-Morales CA, Gray SR, Katikireddi SV, Niedzwiedz CL, Hastie C, Ferguson LD, Berry C, Mackay DF, Gill JM, Pell JP, Sattar N, Welsh P. Modifiable and non-modifiable risk factors for COVID-19, and comparison to risk factors for influenza and pneumonia: results from a UK Biobank prospective cohort study. BMJ Open 2020; 10:e040402
  84. Aung N, Khanji MY, Munroe PB, Petersen SE. Causal Inference for Genetic Obesity, Cardiometabolic Profile and COVID-19 Susceptibility: A Mendelian Randomization Study. Front Genet 2020; 11:586308
  85. Zhang Y, Yang H, Li S, Li WD, Wang J, Wang Y. Association analysis framework of genetic and exposure risks for COVID-19 in middle-aged and elderly adults. Mech Ageing Dev 2021; 194:111433
  86. Mostaza JM, Salinero-Fort MA, Cardenas-Valladolid J, Rodriguez-Artalejo F, Diaz-Almiron M, Vich-Perez P, San Andres-Rebollo FJ, Vicente I, Lahoz C. Pre-infection HDL-cholesterol levels and mortality among elderly patients infected with SARS-CoV-2. Atherosclerosis 2022; 341:13-19
  87. Lassale C, Hamer M, Hernaez A, Gale CR, Batty GD. Association of pre-pandemic high-density lipoprotein cholesterol with risk of COVID-19 hospitalisation and death: The UK Biobank cohort study. Prev Med Rep 2021; 23:101461
  88. Lahoz C, Salinero-Fort MA, Cardenas J, Rodriguez-Artalejo F, Diaz-Almiron M, Vich-Perez P, San Andres-Rebollo FJ, Vicente I, Mostaza JM. HDL-cholesterol concentration and risk of SARS-CoV-2 infection in people over 75 years of age: a cohort with half a million participants from the Community of Madrid. Clin Investig Arterioscler 2021;
  89. Di Maio S, Lamina C, Coassin S, Forer L, Wurzner R, Schonherr S, Kronenberg F. Lipoprotein(a) and SARS-CoV-2 infections: Susceptibility to infections, ischemic heart disease and thromboembolic events. J Intern Med 2022; 291:101-107
  90. Zhang K, Dong SS, Guo Y, Tang SH, Wu H, Yao S, Wang PF, Zhang K, Xue HZ, Huang W, Ding J, Yang TL. Causal Associations Between Blood Lipids and COVID-19 Risk: A Two-Sample Mendelian Randomization Study. Arterioscler Thromb Vasc Biol 2021; 41:2802-2810
  91. Ponsford MJ, Gkatzionis A, Walker VM, Grant AJ, Wootton RE, Moore LSP, Fatumo S, Mason AM, Zuber V, Willer C, Rasheed H, Brumpton B, Hveem K, Kristian Damas J, Davies N, Olav Asvold B, Solligard E, Jones S, Burgess S, Rogne T, Gill D. Cardiometabolic Traits, Sepsis, and Severe COVID-19: A Mendelian Randomization Investigation. Circulation 2020; 142:1791-1793
  92. Yoshikawa M, Asaba K, Nakayama T. Estimating causal effects of atherogenic lipid-related traits on COVID-19 susceptibility and severity using a two-sample Mendelian randomization approach. BMC Med Genomics 2021; 14:269
  93. Leong A, Cole JB, Brenner LN, Meigs JB, Florez JC, Mercader JM. Cardiometabolic risk factors for COVID-19 susceptibility and severity: A Mendelian randomization analysis. PLoS Med 2021; 18:e1003553
  94. Kuo CL, Pilling LC, Atkins JL, Masoli JAH, Delgado J, Kuchel GA, Melzer D. ApoE e4e4 Genotype and Mortality With COVID-19 in UK Biobank. J Gerontol A Biol Sci Med Sci 2020; 75:1801-1803
  95. Kuo CL, Pilling LC, Atkins JL, Masoli JAH, Delgado J, Kuchel GA, Melzer D. APOE e4 Genotype Predicts Severe COVID-19 in the UK Biobank Community Cohort. J Gerontol A Biol Sci Med Sci 2020; 75:2231-2232
  96. Burt TD, Agan BK, Marconi VC, He W, Kulkarni H, Mold JE, Cavrois M, Huang Y, Mahley RW, Dolan MJ, McCune JM, Ahuja SK. Apolipoprotein (apo) E4 enhances HIV-1 cell entry in vitro, and the APOE epsilon4/epsilon4 genotype accelerates HIV disease progression. Proc Natl Acad Sci U S A 2008; 105:8718-8723
  97. Gale SC, Gao L, Mikacenic C, Coyle SM, Rafaels N, Murray Dudenkov T, Madenspacher JH, Draper DW, Ge W, Aloor JJ, Azzam KM, Lai L, Blackshear PJ, Calvano SE, Barnes KC, Lowry SF, Corbett S, Wurfel MM, Fessler MB. APOepsilon4 is associated with enhanced in vivo innate immune responses in human subjects. J Allergy Clin Immunol 2014; 134:127-134
  98. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland HJ, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  99. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland HJ, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  100. Iqbal Z, Ho JH, Adam S, France M, Syed A, Neely D, Rees A, Khatib R, Cegla J, Byrne C, Qureshi N, Capps N, Ferns G, Payne J, Schofield J, Nicholson K, Datta D, Pottle A, Halcox J, Krentz A, Durrington P, Soran H, Heart UsMS, Research C. Managing hyperlipidaemia in patients with COVID-19 and during its pandemic: An expert panel position statement from HEART UK. Atherosclerosis 2020; 313:126-136
  101. Minz MM, Bansal M, Kasliwal RR. Statins and SARS-CoV-2 disease: Current concepts and possible benefits. Diabetes Metab Syndr 2020; 14:2063-2067
  102. Ganjali S, Bianconi V, Penson PE, Pirro M, Banach M, Watts GF, Sahebkar A. Commentary: Statins, COVID-19, and coronary artery disease: killing two birds with one stone. Metabolism 2020; 113:154375
  103. Vahedian-Azimi A, Mohammadi SM, Banach M, Beni FH, Guest PC, Al-Rasadi K, Jamialahmadi T, Sahebkar A. Improved COVID-19 Outcomes following Statin Therapy: An Updated Systematic Review and Meta-analysis. Biomed Res Int 2021; 2021:1901772
  104. Diaz-Arocutipa C, Melgar-Talavera B, Alvarado-Yarasca A, Saravia-Bartra MM, Cazorla P, Belzusarri I, Hernandez AV. Statins reduce mortality in patients with COVID-19: an updated meta-analysis of 147 824 patients. Int J Infect Dis 2021; 110:374-381
  105. Kollias A, Kyriakoulis KG, Kyriakoulis IG, Nitsotolis T, Poulakou G, Stergiou GS, Syrigos K. Statin use and mortality in COVID-19 patients: Updated systematic review and meta-analysis. Atherosclerosis 2021; 330:114-121
  106. Chow R, Im J, Chiu N, Chiu L, Aggarwal R, Lee J, Choi YG, Prsic EH, Shin HJ. The protective association between statins use and adverse outcomes among COVID-19 patients: A systematic review and meta-analysis. PLoS One 2021; 16:e0253576
  107. Wu KS, Lin PC, Chen YS, Pan TC, Tang PL. The use of statins was associated with reduced COVID-19 mortality: a systematic review and meta-analysis. Ann Med 2021; 53:874-884
  108. Kow CS, Hasan SS. Meta-analysis of Effect of Statins in Patients with COVID-19. Am J Cardiol 2020; 134:153-155
  109. Inspiration S Investigators. Atorvastatin versus placebo in patients with covid-19 in intensive care: randomized controlled trial. BMJ 2022; 376:e068407
  110. Deshpande A, Pasupuleti V, Rothberg MB. Statin therapy and mortality from sepsis: a meta-analysis of randomized trials. Am J Med 2015; 128:410-417 e411
  111. Israel A, Schaffer AA, Cicurel A, Cheng K, Sinha S, Schiff E, Feldhamer I, Tal A, Lavie G, Ruppin E. Identification of drugs associated with reduced severity of COVID-19 - a case-control study in a large population. Elife 2021; 10
  112. Delerive P, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors in inflammation control. J Endocrinol 2001; 169:453-459
  113. Feher M, Joy M, Munro N, Hinton W, Williams J, de Lusignan S. Fenofibrate as a COVID-19 modifying drug: Laboratory success versus real-world reality. Atherosclerosis 2021; 339:55-56
  114. Li K, Huang T, Zheng J, Wu K, Li D. Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor alpha: a meta-analysis. PLoS One 2014; 9:e88103
  115. Sedighiyan M, Abdollahi H, Karimi E, Badeli M, Erfanian R, Raeesi S, Hashemi R, Vahabi Z, Asanjarani B, Mansouri F, Abdolahi M. Omega-3 polyunsaturated fatty acids supplementation improve clinical symptoms in patients with Covid-19: A randomised clinical trial. Int J Clin Pract 2021; 75:e14854
  116. Doaei S, Gholami S, Rastgoo S, Gholamalizadeh M, Bourbour F, Bagheri SE, Samipoor F, Akbari ME, Shadnoush M, Ghorat F, Mosavi Jarrahi SA, Ashouri Mirsadeghi N, Hajipour A, Joola P, Moslem A, Goodarzi MO. The effect of omega-3 fatty acid supplementation on clinical and biochemical parameters of critically ill patients with COVID-19: a randomized clinical trial. J Transl Med 2021; 19:128
  117. Talasaz AH, Sadeghipour P, Aghakouchakzadeh M, Dreyfus I, Kakavand H, Ariannejad H, Gupta A, Madhavan MV, Van Tassell BW, Jimenez D, Monreal M, Vaduganathan M, Fanikos J, Dixon DL, Piazza G, Parikh SA, Bhatt DL, Lip GYH, Stone GW, Krumholz HM, Libby P, Goldhaber SZ, Bikdeli B. Investigating Lipid-Modulating Agents for Prevention or Treatment of COVID-19: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:1635-1654
  118. Barnes M, Heywood AE, Mahimbo A, Rahman B, Newall AT, Macintyre CR. Acute myocardial infarction and influenza: a meta-analysis of case-control studies. Heart 2015; 101:1738-1747
  119. Muscente F, De Caterina R. Causal relationship between influenza infection and risk of acute myocardial infarction: pathophysiological hypothesis and clinical implications. Eur Heart J Suppl 2020; 22:E68-E72
  120. Ruane L, Buckley T, Hoo SYS, Hansen PS, McCormack C, Shaw E, Fethney J, Tofler GH. Triggering of acute myocardial infarction by respiratory infection. Intern Med J 2017; 47:522-529

 

Diabetic Neuropathies

ABSTRACT

 

Diabetic neuropathy (DN) is the most common form of neuropathy in developed countries and may affect about half of all patients with diabetes (DM), contributing to substantial morbidity and mortality and resulting in a huge economic burden. DN encompasses multiple different disorders involving proximal, distal, somatic, and autonomic nerves. It may be acute and self-limiting or a chronic, indolent condition.  DN may progress insidiously or present with clinical symptoms and signs that may mimic those seen in many other diseases.  The proper diagnosis therefore requires a thorough history, clinical and neurological examinations, and exclusion of secondary causes. Distal peripheral neuropathy (DPN) is the most common manifestation and is characteristically symmetric, glove and stocking distribution and a length-dependent sensorimotor polyneuropathy. It develops on a background of long-standing chronic hyperglycemia superimposed upon cardiovascular risk factors. Diagnosis is mainly based on a combination of symptoms and signs and occasionally neurophysiological tests are required. Apart from optimizing glycemic control and cardiovascular risk factor management, there is no approved treatment for the prevention or reversal of DPN. Even tight glycemic control at best limits the progression of DPN in patients with type 1 DM, but not to the same extent in type 2 DM. It has been estimated that between 3 and 25% of persons with DM might experience neuropathic pain. Painful DPN can be difficult to treat, and is associated with reduced quality of life, poor sleep, depression, and anxiety. Pharmacotherapy is the mainstay symptomatic treatment for painful DPN. The reported prevalence of diabetic autonomic neuropathy (DAN) varies widely (7.7 to 90%) depending on the cohort studied and the methods used for diagnosis, and can affect any organ system. Cardiovascular autonomic neuropathy (CAN) is significantly associated with overall mortality and with morbidity, including silent myocardial ischemia, coronary artery disease, stroke, DN progression, and perioperative complications. Cardiovascular reflex tests are the criterion standard in clinical autonomic testing.

 

INTRODUCTION

 

Diabetic neuropathy (DN) is the most common and troublesome complication of diabetes mellitus, leading to the greatest morbidity and mortality resulting in a huge economic burden for diabetes care (1,2). It is the most common form of neuropathy in the developed world, accounting for more hospitalizations than all the other diabetes related complications combined. It is the primary risk factor for complications such as foot ulceration, which is responsible for 50-75% of non-traumatic amputations (3). In the United Kingdom, the cost of managing diabetic foot disease is greater than the combined cost of three of the four most common cancers – breast, lung and prostate (4,5). DN is a set of clinical syndromes that affect distinct regions of the nervous system, singly or combined.  It may be silent and go undetected while exercising its ravages; or it may present with clinical symptoms and signs that, although nonspecific and insidious with slow progression also mimics those seen in many other diseases.

 

SCOPE OF THE PROBLEM

 

Diabetic neuropathy results in a variety of syndromes and can be subdivided into focal/multifocal neuropathies, including diabetic amyotrophy, and symmetric polyneuropathies, including sensorimotor polyneuropathy (DPN). The latter is the most common type. The Toronto Diabetic Neuropathy Expert Group defined DPN as a symmetrical, length-dependent sensorimotor polyneuropathy attributable to metabolic and microvascular alterations as a result of chronic hyperglycemia exposure (diabetes) and cardiovascular risk covariates (6).  Its onset is generally insidious, and without treatment the course is chronic and progressive. The loss of small fiber-mediated sensation results in the loss of thermal and pain perception, whereas large fiber impairment results in loss of touch and vibration perception. Sensory fiber involvement may also result in “positive” symptoms, such as paresthesias and pain, although up to 50% of neuropathic patients are asymptomatic. DPN can be associated with the involvement of the autonomic nervous system, i.e., diabetic autonomic neuropathy (7,8) and in its cardiovascular form is associated with at least a three-fold increased risk for mortality (9,10). Cardiac autonomic dysfunction in patients with diabetes is strongly associated with major cardiovascular events and mortality (11).

 

Painful DPN which occurs in up to 34% of patients with diabetes is defined as ‘pain as a direct consequence of abnormalities in the peripheral somatosensory system in people with diabetes’ (12). Persistent neuropathic pain interferes significantly with quality of life (QOL), impairing sleep and recreation; it also significantly impacts emotional well-being, and is associated with – if not the cause of – depression, anxiety, loss of sleep, and noncompliance with treatment (13).  Painful DPN can pose a significant clinical management challenge and if poorly managed can lead to mood and sleep disturbances. Hence, recognition of psychosocial problems that co-exist with neuropathic pain is critical to the management of painful DPN. For many patients, optimal management of chronic pain may require a multidisciplinary team approach with appropriate behavioral therapy, as well as input from a broad range of healthcare professionals (14). 

 

CLASSIFICATION OF DIABETIC NEUROPATHIES

 

Figure 1 and Table 1 describe the classification first proposed by PK Thomas (15) and modified in a recent Position Statement by the American Diabetes Association (16).

Figure 1. Classification of diabetic neuropathy

 

Table 1.  Classification of Diabetic Neuropathies

A. Diffuse neuropathy

  Distal Symmetrical Peripheral Neuropathy

   • Primarily small-fiber neuropathy

   • Primarily large-fiber neuropathy

   • Mixed small- and large-fiber neuropathy (most common)

  Autonomic

   Cardiovascular

    • Reduced Heart Rate Variability

    • Resting tachycardia

    • Orthostatic hypotension

    • Sudden death (malignant arrhythmia)

   Gastrointestinal

    • Diabetic gastroparesis (gastropathy)

    • Diabetic enteropathy (diarrhea)

    • Colonic hypomotility (constipation)

   Urogenital

    • Diabetic cystopathy (neurogenic bladder)

    • Erectile dysfunction

    • Female sexual dysfunction

   Sudomotor dysfunction

    • Distal hypohydrosis/anhidrosis,

    • Gustatory sweating

   Hypoglycemia unawareness

   Abnormal pupillary function

B. Mononeuropathy (mononeuritis multiplex) (atypical forms)

            Isolated cranial or peripheral nerve (e.g., Cranial Nerve III, ulnar, median, femoral, peroneal)

      Mononeuritis multiplex (if confluent may resemble polyneuropathy)

C. Radiculopathy or polyradiculopathy (atypical forms)

            Radiculoplexus neuropathy (a.k.a. lumbosacral polyradiculopathy, proximal motor amyotrophy)

      Thoracic radiculopathy

D. Nondiabetic neuropathies common in diabetes

          Pressure palsies

          Chronic inflammatory demyelinating polyneuropathy

          Radiculoplexus neuropathy

          Acute painful small-fiber neuropathies (treatment-induced)

 

NATURAL HISTORY OF DIABETIC NEUROPATHIES (DN)

 

The natural history of DPN remains poorly understood, as there are few prospective studies that have examined this. The main reason for this is the lack of standardized methodologies for the diagnosis of DPN. Unlike diabetic retinopathy and nephropathy, the lack of simple, accurate and readily reproducible methods of measuring neuropathy is a major challenge. Furthermore, the methods currently used are not only subjective and reliant on the examiner’s interpretation but tend to diagnose DPN when it’s already well established. Nevertheless, it appears that the most rapid deterioration of nerve function occurs soon after the onset of type 1 diabetes; then within 2-3 years there is a slowing of the progress with a shallower slope to the curve of dysfunction (17).  In contrast, in type 2 diabetes, slowing of nerve conduction velocities (NCVs) may be one of the earliest neuropathic abnormalities and often is present even at diagnosis.  In fact, there is accumulating evidence that indicates that the risk of DPN is increased even in patients with prediabetes. In a large population study conducted in Augsburg, Southern Germany, the prevalence of DPN was 28% in subjects with known diabetes, 13% in impaired glucose tolerance (IGT), 11% among those with impaired fasting glucose and 7% in those with normal glucose tolerance (18). After diagnosis, slowing of NCV generally progresses at a steady rate of approximately 1 m/sec/year, and the level of impairment is positively correlated with duration of diabetes. Moreover, nerve conduction velocities remained stable with intensive therapy but decreased significantly with conventional therapy (19,20). In a long term follow up study of type 2 diabetes patients (9), electrophysiologic abnormalities in the lower limb increased from 8% at baseline to 42% after 10 years; in particular, a decrease in sensory and motor amplitudes (indicating axonal destruction) was more pronounced than the slowing of the NCVs. However, there now appears to be a decline in this rate of evolution. It appears that host factors pertaining to general health, management of risk factors and nerve nutrition are changing/improving. This is particularly important when doing studies on the treatment of DPN, which have always relied on differences between drug treatment and placebo, and have apparently been successful because of the decline in function occurring in placebo-treated patients (21).  Recent studies have pointed out the changing natural history of DPN with the advent of therapeutic lifestyle change and the use of statins and ACE inhibitors, which have slowed the progression of DPN and drastically changed the requirements for placebo-controlled studies (22,23).  It is also important to recognize that DPN is a disorder wherein the prevailing abnormality is loss of axons that electrophysiologically translates to a reduction in amplitudes and not conduction velocities; therefore, changes in NCV may not be an appropriate means of monitoring progress or deterioration of nerve function.  Moreover, small, unmyelinated nerve fibers are affected early in DM and are not assessed in NCV studies. Other methods such as quantitative sensory testing, autonomic function testing, skin biopsy with quantification of intraepidermal nerve fibers (IENF), or corneal confocal microscopy are necessary to identify these patients. These techniques will be discussed in greater depth later in this chapter.

 

Although, the true prevalence is unknown and reports vary, it is estimated to be 30% with a range between 6-54% of patients with diabetes (24). It largely depends on the criteria and sensitivity of the diagnostic tests used to define neuropathy, the population (e.g., hospital/community or urban/rural), or the country surveyed and even the etiology of diabetes (24,25). Eleven to 13% of patients reported DN using a questionnaire based survey (26,27); 42-54% were found to have neuropathy when more sensitive measures such as nerve conduction studies were used (28,29). Neurologic complications occur equally in type 1 and type 2 diabetes mellitus and additionally in various forms of acquired diabetes (30).

 

The major morbidity associated with somatic neuropathy is foot ulceration, the precursor of gangrene and limb loss. Neuropathy increases the risk of amputation 1.7 fold; 12 fold if there is deformity (itself a consequence of neuropathy), and 36 fold if there is a history of previous ulceration (31). For more than a decade now, it has been recognized that a limb is lost to diabetes every 30 seconds worldwide (32). According to the International Diabetes Federation (IDF), lower-limb amputations are ten times more common in people with diabetes than in people without diabetes (32, 33). Each week in England there is about 169 amputations in people with diabetes and almost all of these individuals have DN (34). Amputation is not only devastating in its impact on the individual and their family, but also leads to loss of independence and livelihood. In low-income countries, the financial costs can be equivalent to 5.7 years of annual income, potentially resulting in financial ruin for individuals and their families (35). DN also places a substantial financial burden on health-care systems and society in general.

 

MODIFIABLE RISK FACTORS FOR DPN INCIDENCE AND PROGRESSION

 

In both type 1 and 2 diabetes, chronic hyperglycemia has a key role in the pathogenesis of DPN (36). The benefit of glucose lowering is, however, more pronounced in type 1 diabetes (78% relative risk reduction) (37) compared to type 2 (5-9% relative risk reduction) (38). In fact, the benefit of intensive glucose lowering is greatest in younger patients at early stages of the disease. This treatment effects becomes weaker once nerve damage is established. The relationship between glycemic control and DPN in type 2 diabetes is less clear cut. Even when trials have shown that tighter glucose control might have a modest beneficial effect in preventing progression of DPN in type 2 diabetes, such as the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study (39), confusion has arisen when it was reported that a self-reported history of DPN at baseline was associated with an increased risk of mortality with intensive glycemic treatment (40). This highlights the differences between the pathogenesis of DPN in type 1 and 2 diabetes and emphasizes the point that many people with type 2 diabetes develop DPN despite adequate glucose control. The presence of other risk factors, weight gain and multiple comorbidities may have significant roles to play. Although hyperglycemia and duration of diabetes play an important role in DPN, other risk factors have been identified. The EURODIAB Prospective Complications study in type 1 diabetes demonstrated that the incidence of DPN is associated with other potentially modifiable cardiovascular risk factors, including hypertriglyceridemia, hypertension, obesity and smoking (41). More recently, data from the ADDITION study also implicated similar cardiovascular risk factors in the pathogenesis of DPN in type 2 diabetes (26).

 

PATHOGENESIS OF DIABETIC NEUROPATHIES

 

Despite considerable research, the pathogenesis of diabetic neuropathy remains undetermined (42).  This is one reason why, despite several clinical trials, there has been relatively little progress in the development of disease-modifying treatments (43). Historically, a number of causative factors have been identified including persistent hyperglycemia, microvascular insufficiency, oxidative and nitrosative stress, defective neurotrophism, and autoimmune-mediated nerve destruction.  Figure 2 summarizes our current view of the pathogenesis of DPN (44). Detailed discussion of the different theories is beyond the scope of this Chapter and there are several excellent recent reviews (45).

Figure 2. Pathogenesis of diabetic neuropathies. Ab, antibody; AGE, advance glycation end products; C’, complement; DAG, diacylglycerol; ET, endothelin; EDHF, endothelium-derived hyperpolarizing factor; GF, growth factor; IGF; insulin-like growth factor; NFkB, nuclear factor kB; NGF, nerve growth factor; NO, nitric oxide; NT3, neurotropin 3; PKC, protein kinase C; PGI2, prostaglandin I2; ROS, reactive oxygen species; TRK, tyrosine kinase.

CLINICAL PRESENTATION

 

The spectrum of clinical neuropathic syndromes described in patients with diabetes mellitus includes dysfunction of almost every segment of the somatic peripheral and autonomic nervous system (16). Each syndrome can be distinguished by its pathophysiologic, therapeutic, and prognostic features.

 

Focal and Multifocal Neuropathies

 

Focal neuropathies comprise focal limb neuropathies and cranial neuropathies.

Focal limb neuropathies are usually due to entrapment, and mononeuropathies must be distinguished from these entrapment syndromes (Table 2) (46). Mononeuropathies often occur in the older population; they have an acute onset, are associated with pain, and have a self-limiting course resolving in 6–8 weeks. Mononeuropathies can involve the median (5.8% of all diabetic neuropathies), ulnar (2.1%), radial (0.6%), and common peroneal nerves (47). Cranial neuropathies in patients with diabetes are extremely rare (0.05%) and occur in older individuals with a long duration of diabetes (48). The commonest cranial neuropathy is the third nerve palsy and patients present with acute onset unilateral pain in the orbit or sometimes with a frontal headache. There is typically ptosis and ophthalmoplegia, although the pupillary response to light is usually spared. Recovery occurs usually over three months (48). The clinical onset and time-scale for recovery, and the focal nature of the lesions on the third cranial nerve, on post-mortem studies suggested an ischemic etiology.  It is important to exclude any other cause of third cranial nerve palsy (aneurysm or tumor) by CT or MR scanning, where the diagnosis is in doubt. Fourth, sixth and seventh cranial nerve palsies have also been described in patients with diabetes, but the association with diabetes is not as strong as that with third cranial nerve palsy.

 

Table 2. Distinguishing Characteristics of Mononeuropathies, Entrapment Syndromes and Distal Symmetrical Polyneuropathy

Feature

Mononeuropathy

Entrapment syndrome

Neuropathy

Onset

Sudden

Gradual

Gradual

Pattern

Single nerve but may be multiple

Single nerve exposed to trauma

Distal symmetrical poly neuropathy

Nerves involved

CN III, VI, VII, ulnar, median, peroneal

Median, ulnar, peroneal, medial and lateral plantar

Mixed, Motor, Sensory, Autonomic

Natural history

Resolves spontaneously

Progressive

Progressive

Treatment

Symptomatic

Rest, splints, local steroids, diuretics, surgery

Tight Glycemic control, Pregabalin, Duloxetine, Antioxidants, “Nutrinerve”, Research Drugs.

Distribution of Sensory loss

Area supplied by the nerve

Area supplied beyond the site of entrapment

Distal and symmetrical. “Glove and Stocking” distribution.

CN, cranial nerves; NSAIDs, non-steroidal anti-inflammatory drugs

 

Entrapment Syndromes

 

These start slowly and will progress and persist without intervention. A number of nerves including the median, ulnar, radial, lateral femoral cutaneous, fibular, and plantar nerves are vulnerable to pressure damage in diabetes. The etiology is multifactorial involving metabolic and ischemic factors, impaired reinnervation, and even obesity. Carpal tunnel syndrome occurs three times as frequently in people with diabetes compared with healthy populations (49) and is found in up to one third of patients with diabetes.  Its increased prevalence in diabetes may be related to repeated undetected trauma, metabolic changes, or accumulation of fluid or edema within the confined space of the carpal tunnel. The diagnosis is confirmed by electrophysiological studies. Treatment consists of rest, aided by placement of a wrist splint in a neutral position to avoid repetitive trauma.  Anti-inflammatory medications and steroid injections are sometimes useful. Surgery should be considered if weakness appears and medical treatment fails (50).  It consists of sectioning the volar carpal ligament or unentrapping the nerves in the ulnar canal or the peroneal nerve at the head of the fibula and release of the medial plantar nerve in the tarsal tunnel amongst others. A more detailed review of other peripheral nerves vulnerable to entrapment in anatomically constraint channels are discussed elsewhere (51).

 

Proximal Motor Neuropathy (Diabetic Amyotrophy) and Chronic Demyelinating Neuropathies

 

For many years proximal neuropathy has been considered a component of DN.  Its pathogenesis was ill understood (52), and its treatment was neglected with the anticipation that the patient would eventually recover, albeit over a period of some 1-2 years and after suffering considerable pain, weakness and disability. The condition has a number of synonyms including diabetic amyotrophy and femoral neuropathy.  It can be clinically identified based on the occurrence of these common features: 1) primarily affects those aged 50 to 60 years old with type 2 diabetes; 2) onset can be gradual or abrupt; 3) presents with severe pain in the thighs, hips and buttocks, followed by significant weakness of the proximal muscles of the lower limbs with inability to rise from the sitting position (positive Gower's maneuver); 4) can start unilaterally and then spread bilaterally; 5) often coexists with distal symmetric polyneuropathy; and 6) is characterized by muscle fasciculation, either spontaneous or provoked by percussion. Pathogenesis is not yet clearly understood although immune-mediated epineural microvasculitis has been demonstrated in some cases. Despite limited evidence of efficacy some immunosuppressive therapy is recommended using high dose steroids or intravenous immunoglobulin (53). Close monitoring and appropriate management of blood glucose is advised if high dose steorids are used (54). The condition can occur secondary to a variety of causes unrelated to diabetes, but which have a greater frequency in patients with diabetes than the general population.  Hence, it is important to exclude other causes such as chronic inflammatory demyelinating polyneuropathy (CIDP), monoclonal gammopathy, circulating GM1 antibodies, and inflammatory vasculitis (55,56). In the classic form of diabetic amyotrophy, axonal loss is the predominant process (57). Electrophysiologic evaluation reveals lumbosacral plexopathy (58). In contrast, if demyelination predominates and the motor deficit affects proximal and distal muscle groups, the diagnoses of CIDP, monoclonal gammopathy of unknown significance, and vasculitis should be considered (59,60).  The diagnosis of these demyelinating conditions is often overlooked although recognition is very important because unlike DN, they are sometimes treatable. Furthermore, they occur 11 times more frequently in patients with diabetes than nondiabetic patients (61,62).  Biopsy of the obturator nerve have revealed deposition of immunoglobulin, demyelination and inflammatory cell infiltrate of the vasa nervorum (63). Cerebrospinal fluid (CSF) protein content is high and lymphocyte count increased.  Treatment options include: intravenous immunoglobulin for CIDP (64), plasma exchange for MGUS, steroids and azathioprine for vasculitis, and withdrawal of drugs or other agents that may have caused vasculitis. It is important to divide proximal syndromes into these two subcategories, because the CIDP variant responds dramatically to intervention (65), whereas amyotrophy runs its own course over months to years. Until more evidence is available, they should be considered separate syndromes.

 

Diabetic Truncal Radiculoneuropathy

 

Diabetic truncal radiculoneuropathy affects middle-aged to elderly patients and has a predilection for male sex (16).  Acute onset of pain is the most important symptom and it occurs in a girdle-like distribution over the lower thoracic or abdominal wall. It can be uni- or bilaterally distributed. Motor weakness is rare but there may be local bulging of the muscle. Patchy sensory loss may be present and other causes of nerve root compression should be excluded. Resolution generally occurs within 4-6 months (16).

 

Rapidly Reversible Hyperglycemic Neuropathy

 

Reversible abnormalities of nerve function may occur in patients with recently diagnosed or poorly controlled diabetes. These are unlikely to be caused by structural abnormalities, as recovery soon follows restoration of euglycemia.  Rapidly reversible hyperglycemic neuropathy usually presents with distal sensory symptoms, and whether these abnormalities result in an increased risk of developing chronic neuropathies in the future remains unknow (8).

 

Generalized Symmetric Polyneuropathy

 

ACUTE SENSORY NEUROPATHY

 

Acute sensory (painful) neuropathy is considered by some authors a distinctive variant of distal symmetrical polyneuropathy (66). The syndrome is characterized by severe pain, cachexia, weight loss, depression and sexual dysfunction. It occurs predominantly in male patients and may appear at any time in the course of both type 1 and type 2 diabetes.  It is self-limiting and invariably responds to simple symptomatic treatment (67). Conditions such as Fabry's disease, amyloidosis, HIV infection, heavy metal poisoning (such as arsenic), and excess alcohol consumption should be excluded. Autonomic nervous system involvement can also occur and can be very disabling.

 

Patients report unremitting burning, deep pain and hyperesthesia especially in the feet. Other symptoms include sharp, stabbing, lancinating pain; “electric shock” like sensations in the lower limbs that appear more frequently during the night; paresthesia; tingling; coldness, and numbness. Signs are usually absent with a relatively normal clinical examination, except for allodynia (exaggerated response to non-noxious stimuli) during sensory testing and, occasionally, absent or reduced ankle reflexes. There are no motor signs and little or no abnormality on nerve conduction studies.

 

Acute sensory neuropathy is usually associated with poor glycemic control but may also appear after sudden improvement of glycemia. Most commonly associated with the onset of insulin therapy, being termed "insulin neuritis",it can also occur with oral hypoglycemic treatment. Patients present with severe neuropathic pain and/or autonomic symptoms with or without an acute worsening of retinopathy.  Although the pathologic basis has not been determined, one hypothesis suggests that changes in blood glucose flux produce alterations in epineural blood flow, leading to ischemia; proinflammatory cytokines from activation of microglia have also been implicated (68). Hence, rapid glycemic changes in patients with uncontrolled diabetes increases the risk of this complication and should be avoided. A 2-3% (10-42mmol/mol) decrease in HbA1c over 3 months is associated with a 20% absolute risk of developing this complication. The risk exceeds 80% with a decreased in HbA1c of >4% (20mmol/mol) (69).  A study using in vivo epineural vessel photography and fluorescein angiography demonstrated abnormalities of epineural vessels including arteriovenous shunting and proliferating new vessels in patients with acute sensory neuropathy (68). Other authors relate this syndrome to diabetic lumbosacral radiculoplexus neuropathy (DLRPN) and propose an immune mediated mechanism (70).

 

The key in the management of this syndrome is achieving and maintaining blood glucose stability (71).  Most patients also require medication for neuropathic pain. The natural history of this disease is resolution of symptoms within one year.

 

CHRONIC SENSORIMOTOR NEUROPATHY OR DISTAL SYMMETRIC POLYNEUROPATY (DPN)

 

The most common form of neuropathy in diabetes is a distal symmetrical polyneuropathy.  It occurs in both type 1 and type 2 DM with similar frequency and may already be present at the time of diagnosis of type 2 DM (18). Sensory symptoms include numbness (‘dead feeling’), paraesthesia, and neuropathic pain (hyperalgesia, allodynia, deep aching, burning and sharp stabbing sensations). Patients do occasionally present paradoxically with a painful/painless leg i.e. painful neuropathic symptoms in the presence of severe sensory loss (72). Symptoms begin in the toes before progressing in a stocking and then a glove distribution as the disease progresses. Unsteadiness or sensory ataxia leading to increased falls risk occurs in advanced neuropathy loss of proprioception, foot deformity, and abnormal muscle sensory function (73). In the absence of painful symptoms, the onset of DPN is insidious whereby patients remain completely asymptomatic and signs discovered by a detailed neurological examination. Unfortunately, DPN is often already established or well advanced when identified by bedside clinical examination.

 

It is critically important to annually (at least) examine the feet of patients with diabetes as loss of protective sensation is the strongest risk factor for diabetic foot ulceration. On physical examination, a symmetrical stocking like distribution of sensory abnormalities in both lower limbs is usually seen. In more severe cases, hands may be involved. All sensory modalities can be affected, particularly vibration, touch and position perceptions (large Aα/β fiber damage); and pain, with abnormal heat and cold temperature perception (small thinly myelinated Aδ and unmyelinated C fiber damage, see Figure 3, 4 and 5; Table 3). Deep tendon reflexes may be absent or reduced, especially in the lower extremities, although this may occur with advancing age in the absence of neuropathy. When DPN is established, small muscle wasting of the foot and extensor halluces longus may be seen but severe weakness is rare and should raise the possibility of a non-diabetic etiology of the neuropathy. High arching of the foot, clawing of the toes with prominent metatarsal heads also become apparent – increasing the risk ulceration (74). A thorough assessment of patient’s footwear is essential. A poor fit, abnormal wear from internal pressure areas and foreign objects found in footwear are common causes of trauma leading to foot ulceration (75).

Figure 3. Clinical presentation of small and large fiber neuropathies. Aα fibers are large myelinated fibers, in charge of motor functions and muscle control. Aα/β fibers are large myelinated fibers too, with sensory functions such as perception to touch, vibration, and position. Aδ fibers are small myelinated fibers, in charge of pain stimuli and cold perception. C fibers can be myelinated or unmyelinated and have both sensory (warm perception and pain) and autonomic functions (blood pressure and heart rate regulation, sweating, etc.)

Figure 4. Clinical manifestations of small fiber neuropathies

Figure 5. Nerve fibers of the skin and their functions

 

Table 3. Subtypes of Neuropathies

Clinical Manifestations of Small Fiber Neuropathies:

•           Small thinly myelinated Aδ and unmyelinated C fibers are affected.

•           Prominent symptoms with burning, superficial, or lancinating pain often accompanied by hyperalgesia, dysesthesia, and allodynia.

•           Progression to numbness and hypoalgesia (Disappearance of pain may not necessarily reflect nerve recovery but rather nerve death, and progression of neuropathy must be excluded by careful examination).

•           Abnormal cold and warm thermal sensation.

•           Abnormal autonomic function with decreased sweating, dry skin, impaired vasomotion and skin blood flow with cold feet.

•           Intact motor strength and deep tendon reflexes.

•           Negative nerve conduction velocity findings.

•           Loss of cutaneous nerve fibers on skin biopsies.

•           Can be diagnosed clinically by reduced sensitivity to 1.0 g Semmes Weinstein monofilament and prickling pain perception using the Waardenberg wheel or similar instrument.

•           Patients at risk of foot ulceration and subsequent gangrene and amputations.

Clinical Manifestations of Large Fiber Neuropathies

•           Large myelinated, rapidly conducting Aα/β fibers are affected and may involve sensory and/or motor nerves.

•           Prominent signs with sensory ataxia (waddling like a duck), wasting of small intrinsic muscles of feet and hands with hammertoe deformities and weakness of hands and feet.

•           Abnormal deep tendon reflexes.

•           Impaired vibration perception (often the first objective evidence), light touch, and joint position perception.

•           Shortening of the Achilles tendon with pes equinus.

•           Symptoms may be minimal: sensation of walking on cotton, floors feeling "strange", inability to turn the pages of a book, or inability to discriminate among coins.  In some patients with severe distal muscle weakness, inability to stand on the toes or heels.

•           Abnormal nerve conduction velocity findings

•           Increased skin blood flow with hot feet.

•           Patients at higher risk of falls, fractures, and development of Charcot Neuroarthropathy

•           Most patients with DPN, however, have a "mixed" variety of neuropathy with both large and small nerve fiber damages.

 

DIAGNOSIS OF DIABETIC NEUROPATHIES

 

Diabetic peripheral neuropathy can be diagnosed by the bedside with careful clinical examination of the feet and legs using simple tools within a few minutes. The basic neurological assessment comprises the general medical and neurological history, inspection of the feet, and neurological examination of sensation using simple semi-quantitative bed-side instruments such as the 10g Semmes-Weinstein monofilament, Neuropen (76) (to assess touch/pressure), NeuroQuick (77) or Tiptherm (78) (temperature), calibrated Rydel-Seiffer tuning fork (vibration), pin-prick (pain), and tendon reflexes (knee and ankle) (Table 4).  In addition, assessment of joint position and motor power should also be assessed. The Rydel Seiffer tuning fork is a 128 Hz tuning fork which allows quantifiable assessment of vibration perception in the feet of diabetic patients. When vibrating, two triangles appear on the graduated scale of 0–8 which join together as the amplitude decreases. The normal range for the graduated tuning fork on the dorsal distal joint of the great toe is ≥5/8 scale units in persons 21-40 years old, ≥4.5/8 in those 41-60 years old, ≥4/8 in individuals 61-71 years old, and ≥3.5/8 scale units in those 72-82 years old (79). In resource, limited settings the simple Ipswich Touch Test can be performed by lightly touching the tips of the first, third and fifth toes (80). It is recommended that a simple foot examination to detect loss of protective foot sensation, pedal pulses, and foot deformity is performed from the diagnosis of type 2 diabetes, 5-years after the diagnosis of type 1 diabetes and annually thereafter (81,82,16). This is performed in order to determine the risk of foot ulceration and prompt early referral for foot protection, regular podiatry or specialist input.

 

Table 4.  Examination - Bedside Sensory Tests

Sensory Modality

Nerve Fiber

Instrument

Associated Sensory Receptors

Vibration

Ab (large)

128 Hz

Tuning fork

Ruffini corpuscle mechanoreceptors

Pain (pinprick)

C (small)

Neuro-tips

Nociceptors for pain and warmth

Pressure

Ab, Aa (large)

1 g and 10 g

Monofilament

Pacinian  corpuscle

Light touch

Ab, Aa (large)

Wisp of cotton

Meissner’s corpuscle

Cold

Ad (small)

Cold tuning fork

Cold thermoreceptors

 

A consensus definition of DPN has been proposed by the Toronto Diabetic Neuropathy Expert Group, see below (6). In a clinical context, the diagnosis of ‘possible’ or ‘probable’ DPN is normally sufficient without the need for specialist investigations. For research purposes further tests are needed for a diagnosis of ‘confirmed’ DPN’, ‘Subclinical’ DPN or small fiber neuropathy.

 

Toronto Classification of DPN (6)

 

1)         Possible DSN: The presence of symptoms or signs of DPN may include the following: symptoms–decreased sensation, positive neuropathic sensory symptoms (e.g., “asleep numbness,” prickling or stabbing, burning or aching pain) predominantly in the toes, feet, or legs; or signs–symmetric decrease of distal sensation or unequivocally decreased or absent ankle reflexes.

 

2)         Probable DPN: The presence of a combination of symptoms and signs of neuropathy including any 2 or more of the following: neuropathic symptoms, decreased distal sensation, or unequivocally decreased or absent ankle reflexes.

 

3)         Confirmed DPN: The presence of an abnormality of nerve conduction and a symptom or symptoms, or a sign or signs, of neuropathy confirm DPN.  If nerve conduction is normal, a validated measure of small fiber neuropathy (with class 1 evidence) may be used. To assess for the severity of DPN, several approaches can be recommended: for e.g., the graded approach outlined above; various continuous measures of sum scores of neurologic signs, symptoms or nerve test scores; scores of functions of activities of daily living; or scores of predetermined tasks or of disability.

 

4)         Subclinical DPN: The presence of no signs or symptoms of neuropathy are confirmed with abnormal nerve conduction or a validated measure of small fiber neuropathy (with class 1 evidence).  Definitions 1, 2, or 3 can be used for clinical practice and definitions 3 or 4 can be used for research studies.

 

5)         Small fiber neuropathy (SFN): SFN should be graded as follows: 1) possible: the presence of length-dependent symptoms and/or clinical signs of small fiber damage; 2) probable: the presence of length-dependent symptoms, clinical signs of small fiber damage, and normal sural nerve conduction; and 3) definite: the presence of length-dependent symptoms, clinical signs of small fiber damage, normal sural nerve conduction, and altered intraepidermal nerve fiber density (IENFD) at the ankle and/or abnormal thermal thresholds at the foot (Figure 4).

 

The following findings should alert the physician to consider causes for DPN other than diabetes and referral for a detailed neurological work-up: 1.) pronounced asymmetry of the neurological deficits, 2.) predominant motor deficits, mononeuropathy, or cranial nerve involvement, 3.) rapid development or progression of the neuropathic impairments, 4.) progression of the neuropathy despite optimal glycemic control, 5.) symptoms from the upper limbs, 6.) family history of non-diabetic neuropathy, and 7.) diagnosis of DPN cannot be ascertained by clinical examination.

 

Conditions Mimicking Diabetic Neuropathy

 

An atypical pattern of presentation of symptoms or signs, i.e., the presence of relevant motor deficits, an asymmetrical or proximal distribution, or rapid progression, always requires referral for electrodiagnostic testing. Furthermore, in the presence of such atypical neuropathic signs and symptoms other forms of neuropathy should be sought and excluded.  A good medical history is essential to exclude other causes of neuropathy: a history of trauma, cancer, unexplained weight loss, fever, substance abuse, or HIV infection suggests that an alternative source should be sought. Screening laboratory tests may be considered: serum B12 with its metabolites, folic acid, thyroid function, full blood count, metabolic profile, and serum free light chains (16).

 

There are a number of conditions that can be mistaken for painful DPN: intermittent claudication in which the pain is exacerbated by walking; Morton’s neuroma, in which the pain and tenderness are localized to the intertarsal space and are elicited by applying pressure with the thumb in the appropriate intertarsal space; osteoarthritis/inflammatory arthritis, in which the pain is confined to the joints, made worse with joint movement or exercise, and associated with morning stiffness that improves with ambulation; radiculopathy in which  the pain originates in the shoulder, arm, thorax, or back and radiates into the legs and feet; Charcot neuropathy in which the pain is localized to the site of the collapse of the bones of the foot, and the foot is hot rather than cold; plantar fasciitis, in which there is shooting or burning in the heel with each step and there is exquisite tenderness in the sole of the foot; and tarsal tunnel syndrome in which the pain and numbness radiate from beneath the medial malleolus to the sole and are localized to the inner side of the foot. These contrast with the pain of DPN which is bilateral, symmetrical, covering the whole foot and particularly the dorsum, and is worse at night interfering with sleep.  

 

Scored Clinical Assessment Tools for Diabetic Peripheral Neuropathy

 

Scored Clinical assessments provide standardized, quantitative, and objective measures to assess for both the severity of symptoms and the degree of neuropathic deficits. These tools which have been subjected to strict validation studies, are sufficiently reproducible but require some minimal training. The most widely used instruments include: the Michigan Neuropathy Screening Instrument Questionnaire (MNSIQ, 15-item self-administered questionnaire), Michigan Neuropathy Screening Instrument (MNSI, MNSIQ plus a structured clinical examination), Michigan Diabetic Neuropathy Score (neurological assessment coupled with nerve conduction studies) (83), Toronto Clinical Neuropathy Score (TCNS, composite score of neuropathy symptoms sensory exam and reflexes) (84), modified TCNS (composite score of neuropathy symptoms and signs) (85), Neuropathy Disability Score (neuropathy signs, including reflexes) (86), Neurological Disability Score (neurological examination of cranial nerves, and upper and lower limbs) (87), the Neuropathy Symptom Score (assessment of sensory, motor and autonomic neuropathy symptoms) (87), and the Neuropathy impairment score (NIS) for neuropathic deficits (impairments) (87). A number of instruments have also been used to assess neuropathic pain and these include: the Neuropathy Total Symptom Score-6 (NTSS-6; measures frequency and intensity of neuropathic symptoms) (88), PainDETECT (patient administered 10-item questionnaire) (89), DN4 (Doleur Neuropathique en 4 Questions; 7 sensory descriptors and 3 clinical signs) (90) and the Neuropathic Pain Symptom Inventory (NPSI; self-administered 12-item questionnaire evaluating different symptoms of neuropathic pain) (91).

 

Objective Devices for the Diagnosis of Neuropathy

 

Nerve conduction studies are the current ‘gold’ standard for the diagnosis of DN. This robust measure also predicts foot ulceration and mortality. However, they are time consuming, labor intensive, costly, and impractical in routine clinical care.

 

Skin biopsy has become a widely used tool to investigate small caliber sensory nerves including somatic unmyelinated intraepidermal nerve fibers (IENF), dermal myelinated nerve fibers, and autonomic nerve fibers in peripheral neuropathies and other conditions (92).  Different techniques for tissue processing and nerve fiber evaluation have been used.  For diagnostic purposes in peripheral neuropathies, the current recommendation is to perform a 3-mm punch skin biopsy at the distal leg and quantification of the linear density of IENF in at least three 50-µm thick sections per biopsy, fixed in 2% PLP or Zamboni's solution, by bright-field immunohistochemistry or immunofluorescence with anti-protein gene product (PGP) 9.5 antibodies (93). Quantification of IENF density appeared more sensitive than sensory nerve conduction study or sural nerve biopsy in diagnosing SFN.

 

Quantitative sensory testing (QST) enables more accurate assessment of sensory deficits - also those related to small fiber function - by applying controlled and quantified stimuli and standardized procedures. Moreover, assessment of thermal thresholds can be a helpful tool in the diagnostic pathway of small fiber polyneuropathy (16).

 

Point of Care Devices for the Diagnosis of DN

 

Significant progress has been made to develop point-of-care (POC) devices that are capable of diagnosing early, subclinical neuropathy. Papanas et al have recently comprehensively reviewed these devices (94). Therefore, we will briefly outline the following devices: the NeuroQuick 77, NeuroPAD (95), NC-Stat DPN-Check (96), Corneal Confocal Microscopy (CCM) (97,98), and Sudoscan (99,100).

 

DPN CHECK

 

The DPN-Check is a novel, user-friendly, handheld POC devices that performs a sural nerve conduction study in three minutes (Figure 6). It is an acceptable proxy to standard nerve conduction studies which are time-consuming, expensive, and often require patients to be seen in specialist’s clinics. The DPN check has been demonstrated to have excellent reliability with an inter- and intra-observer intraclass correlation coefficients of between 0.83 and 0.97 for sensory nerve action potentials respectively (101). It also has good validity with 95% sensitivity and 71% specificity when compared against reference standard nerve conduction study (101) for the diagnosis of DN.

Figure 6. DPN Check device

As detailed above, nerve conduction studies are only an assessment of large nerve fiber function. DPN, on the other hand, usually involves both small and large nerve fibers, with some evidence suggesting small nerve fiber involvement early in its natural history (102,103). Small nerve fibers constitute 80-91% of peripheral nerve fibers and control pain perception, autonomic and sudomotor function. Although intraepidermal nerve fiber density measurement from lower limb skin biopsy is considered the gold standard for the diagnosis of small fiber neuropathy (104,92) it is invasive and hence not suitable for routine screening. However, a number of POC devices have been developed to assess small fiber dysfunction. These include:

 

NEUROQUICK

 

Thinly myelinated Aδ and unmyelinated C-fibers are small caliber nerves that mediate thermal sensation and nociceptive stimuli. Quantitative sensory testing of thermal discrimination thresholds is a non-invasive test used to examine impaired small nerve fiber function. NeuroQuick is a handheld device for quantitative bedside testing of cold thermal perception threshold. It allows near patient assessment of small fiber dysfunction avoiding the use of time-consuming and expensive quantitative sensory testing equipment in a laboratory. To date, one published clinical validation study has been performed in a diabetic population which suggests it is a valid and reliable screening tool for the assessment of small fiber dysfunction (77). Use of NeuroQuick was more sensitive in detecting early DPN compared to the traditional bedside screening tests such as the tuning fork or elaborate thermal testing (77). However, it is a psychophysical test that relies on the cognition/attention of the patient. Furthermore, the coefficients of variation for repeated NeuroQuick measurements ranged between 8.5% and 20.4% (77). Further studies are required to demonstrate whether the NeuroQuick is a useful screening tool to detect small fiber dysfunction in DPN.

 

NEUROPAD    

 

This is a 10-minute test which measures sweat production on the plantar surface of the foot (Figure 7). It is based on a color change in a cobalt compound from blue to pink which produces a categorical output with modest diagnostic performance for DPN compared to electrophysiological assessments. If the patch remains completely or partially blue within 10 min, the result is considered abnormal (105).   No training is required to administer Neuropad, nor does it require responses from the patient. Therefore, this method of assessment may be more suitable for screening in community settings and those with cognitive or communication difficulties who have to respond to other methods of assessment. A number of clinical validation studies (95, 106) have been conducted which demonstrates low sensitivity for large fiber neuropathy (50-64%) but much higher sensitivity for small fiber neuropathy (80%) 107. Neuropad has also shown good reproducibility with intra- and inter-observer coefficient of variation between 4.1% and 5.1% (108).

Figure 7. NeuroPAD

 

CORNEAL CONFOCAL MICROSCOPY 

 

Corneal confocal microscopy (CCM, Figure 8) is a noninvasive technique used to detect small nerve fiber loss in the cornea which correlates with both increasing neuropathic severity and reduced IENFD in patients with diabetes (103,109). A novel technique of real-time mapping permits an area of 3.2 mm² to be mapped with a total of 64 theoretically non-overlapping single 400 µm² images (110). There have been a number of clinical validation studies including one 3.5-year prospective study in T1DM which demonstrated relatively modest to high sensitivity (82%) and specificity (69%) of CCM for the incipient DPN (98). It has good reproducibility for corneal nerve fiber length measurements with intra- and inter-observer intraclass correlation coefficients of 0.72 and 0.73 respectively. Currently, CCM is used in specialist centers, but would suit widespread application given its easy application for patient follow-up. However, large, multicenter, prospective studies are now required to confirm that corneal nerve changes unequivocally reflect the complex pathological processes in the peripheral nerve. Moreover, the establishment of a normative database and technical improvements in automated fiber measurements and wider-area image analysis may be useful to increase diagnostic performance.

Figure 8. Examples of corneal nerve fiber density in a patient with no diabetic neuropathy on the left and with established diabetic neuropathy on the right.

CONTACT HEAT EVOKED POTENTIALS  

 

Contact Heat Evoked Potentials (CHEPS) has been studied in healthy controls, newly diagnosed and established patients with diabetes, and patients with the metabolic syndrome. It does appear that CHEPS is capable of detecting small fiber neuropathy in the absence of other indices, and that CHEPS correlates with quantitative sensory perception and objective tests of small fiber structure (intraepidermal nerve fiber density) (111) and function (cooling detection threshold and cold pain) (112) .

 

SUDOSCAN

 

Sudoscan®, an instrument capable of detecting chloride ion flux in response to a very low current (Figure 9), is an objective and quantitative sudomotor function test with promising sensitivity and specificity in the investigation of DPN (113). The entire evaluation takes only 2 minutes and can be done in an ambulatory setting. A measurement of electrochemical skin conductance (ESC) for the hands and feet, that are rich in sweat glands, is generated from the derivative current associated with the applied voltage. Sensitivity and specificity of foot ESC for classifying DPN were 87.5% and 76.2%, respectively. The area under the ROC curve (AUC) was 0.85 (99).

Figure 9. SUDOSCAN test of sudomotor function being performed

SUMMARY OF POINT OF CARE DEVICES

 

In summary, the sensitivity of point of care devices seems acceptable and perhaps a combination of devices may be used in the future for detecting DPN. However, there is high heterogeneity and patient selection bias in most of the studies. Further studies are needed to evaluate the performance of point of care devices against Wilson criteria for screening of undiagnosed DPN at the population level. Prospective studies of hard endpoints (e.g., foot ulcerations and lower limb amputations) are also necessary to ensure that the benefits of screening are important for patients. The cost-effectiveness of implementing screening using these devices also needs to be carefully appraised. Point of care devices provide rapid, non-invasive tests that could be used as an objective screening test for DPN in busy diabetic clinics, ensuring adherence to current recommendation of annual assessment for all patients with diabetes that remains unfulfilled.

 

Summary of Clinical Assessment of DPN

 

Symptoms of neuropathy can vary markedly from one patient to another. For this reason, a number of symptom screening questionnaires with similar scoring systems have been developed. These questionnaires are useful for patient follow-up and to assess response to treatment. A detailed clinical examination is the key to the diagnosis of DPN.  The latest position statement of the American Diabetes Association recommends that all patients with diabetes be screened for DPN at diagnosis in type 2 DM and 5 years after diagnosis in type 1 DM. DPN screening should be repeated annually and must include sensory examination of the feet and ankle reflexes (16).  One or more of the following can be used to assess sensory function: pinprick (using the Waardenberg wheel or similar instrument), temperature, vibration perception (using 128-Hz tuning fork) or 10-g monofilament pressure perception at the distal halluces. For this last test a simple substitute is to use 25 lb strain fishing line cut into 4 cm and 8 cm lengths, which translate to 10 and 1 g monofilaments respectively (114). The most sensitive measure has been shown to be the vibration detection threshold, although sensitivity of 10-g Semmes-Weinstein monofilament to identify feet at risk varies from 86 to 100% (115,116). Combinations of more than one test have more than 87% sensitivity in detecting DPN (117). Longitudinal studies have shown that these simple tests are good predictors of foot ulcer risk (118). Numerous composite scores to evaluate clinical signs of DN, such as the Neuropathy Impairment Score (NIS) are currently available. These, in combination with symptom scores, are useful in documenting and monitoring neuropathic patients in the clinic (119). Feet should always be examined in detail to detect ulcers, calluses, and deformities, and footwear must be inspected at every visit. However, these simple bedside tests are crude and detect DN very late in its natural history. Even the benefits gained by standardising clinical assessment using scored clinical assessments such as the Michigan Neuropathy Screening Instrument (MNSI) (120), the Toronto Clinical Neuropathy Score (TCNS) (84,85) and the United Kingdom Screening Test (UKST) (86), remain subjective, heavily reliant on the examiners’ interpretations (121). Bedside tests used to aid diagnosis of neuropathy such as the 10g monofilament (122), the Ipswich Touch Test (80), and vibration perception threshold using the tuning fork (123) are not only reliant on patients’ subjective response but are mainly utilised to identify the loss of protective foot sensation and risk of ulceration (124). As such, these tests tend to diagnose DPN when it is already well-established (125). Late diagnosis hampers the benefits of early identification which includes a focus on early, intensified diabetes control, and the prevention of neuropathy-related sequelae. Conversely, the situation is different for the detection of diabetic retinopathy using digital camera-based retinal photography (126) or diabetic kidney disease using blood and urine tests. These developments led to the institution of a robust annual screening program that has led to significant reduction in blindness, such that retinopathy is no longer the commonest cause of blindness in working age adults (127) and reductions in end stage renal failure (128). Unfortunately, by the time neuropathy is detected using these crude tests, it is often very well established and consequently impossible to reverse or even to halt the inexorable neuropathic process.

 

In the clinical research settings nerve conduction studies, quantitative sensory testing, and skin biopsy is used to identify and quantify early, subclinical neuropathy. Multiple studies have proven the value of Quantitative Sensory Testing (QST) measures in the detection of subclinical neuropathy (small fiber neuropathy), the assessment of progression of neuropathy, and the prediction of risk of foot ulceration (117,129,130). These standardized measures of vibration and thermal thresholds also play an important role in multicenter clinical trials as primary efficacy endpoints. A consensus subcommittee of the American Academy of Neurology stated that QST receive a Class II rating as a diagnostic test with a type B strength of recommendation (131).

 

The use of electrophysiologic measures (nerve conduction velocity, NCV) in both clinical practice and multicenter clinical trials is recommended (6, 132). In a long term follow-up study of type 2 patients with diabetes (28) NCV abnormalities in the lower limbs increased from 8% at baseline to 42% after 10 years of disease. A slow progression of NCV abnormalities was seen in the Diabetes Control and Complication Trial (DCCT). The sural and peroneal nerve conduction velocities diminished by 2.8 and 2.7 m/s respectively, over a 5-year period (21). Furthermore, in the same study, patients who were free of neuropathy at baseline had a 40% incidence of abnormal NCV in the conventionally treated group versus 16% in the intensive therapy treated group after 5 years. However, the neurophysiologic findings vary widely depending on the population tested and the type and distribution of the neuropathy. Patients with painful, predominantly small fiber neuropathy have normal studies. There is consistent evidence that small, unmyelinated fibers are affected early in DM and these alterations are not diagnosed by routine NCV studies (45). Therefore, other methods, such as QST, autonomic testing, or skin biopsy with quantification of intraepidermal nerve fibers (IENF) are needed to detect these patients (22,133,134). Nevertheless electrophysiological studies play a key role in ruling out other causes of neuropathy and are essential for the identification of focal and multifocal neuropathies (46,8).

 

Intraepithelial Nerve Fiber Density

 

The importance of the skin biopsy as a diagnostic tool for DPN is increasingly being recognized (45, 135). This technique quantitates small epidermal nerve fibers through antibody staining of the pan-axonal marker protein gene product 9.5 (PGP 9.5). Though minimally invasive (3-mm diameter punch biopsy), it enables a direct study of small fibers, which cannot be evaluated by NCV studies. It has led to the recognition of the small nerve fiber syndrome as part of IGT and the metabolic syndrome (Figure 10). When patients present with the “burning foot or hand syndrome”, evaluation for glucose tolerance and the metabolic syndrome (including waist circumference, blood pressure, and plasma triglyceride and HDL-C levels) becomes mandatory.  Therapeutic life style changes (136) can result in nerve fiber regeneration, reversal of the neuropathy, and alleviation of symptoms (see below). 

Figure 10. Intraepidermal nerve fiber loss in small vessel neuropathy. Loss of cutaneous nerve fibers that stain positive for the neuronal antigen protein gene product 9.5 (PGP 9.5) in metabolic syndrome and diabetes.

It is widely recognized that neuropathy per se can affect the quality of life (QOL) of patients with diabetes. A number of instruments have been developed and validated to assess QOL in DPN. The NeuroQoL measures patients’ perceptions of the impact of neuropathy and foot ulcers (137). The Norfolk QOL questionnaire for DPN is a validated tool addressing specific symptoms and the impact of large, small, and autonomic nerve fiber functions (138). The tool has been used in clinical trials and is available in several validated language versions. It was tested in 262 subjects (healthy controls, controls with diabetes, and DPN patients): differences between DN patients and both diabetes and healthy controls were significant (p<0.05) for all item groupings (small fiber, large fiber, and autonomic nerve function; symptoms; and activities of daily living (ADL). Total QOL scores correlated with total neuropathy scores. The ADL, total scores, and autonomic scores were also greater in controls with diabetes compared to healthy controls (p<0.05), suggesting that diabetes per se impacts some aspects of QO (137).

 

The diagnosis of DPN is mainly a clinical one with the aid of specific diagnostic tests according to the type and severity of the neuropathy. However other non-diabetic causes of neuropathy must always be excluded, depending on the clinical findings (B12 deficiency, hypothyroidism, uremia, CIDP, etc.) (Figure 11).

Figure 11. A diagnostic algorithm for assessment of neurologic deficit and classification of neuropathic syndromes: B12, vitamin B12; BUN, blood urea nitrogen; CHEPS, Contact Heat Evoked Potentials CIDP, chronic inflammatory demyelinating polyneuropathy; EMG, electromyogram; Hx, history; MGUS, monoclonal gammopathy of unknown significance; NCV, nerve conduction studies; NIS, neurologic impairment score (sensory and motor evaluation); NSS, neurologic symptom score; QAFT, quantitative autonomic function tests; QST, quantitative sensory tests; Sudo, sudomotor function testing.

Central Nervous System Involvement

 

Hitherto considered a disease of the peripheral nervous system, there is now mounting evidence of central nervous system (CNS) involvement in DN (Figure 12). Several magnetic resonance imaging studies provide valuable insight into CNS alterations in DN. From the spinal cord (139,140) to the cerebral cortex, structural (141), biochemical (142,143), perfusion (144), and functional changes (145,146) have been described. Although the initial injury may occur in the peripheral nerves, concomitant changes within the CNS may have a crucial role in the pathogenesis and determining clinical phenotype and even treatment response in painful DN.

 

Central nervous system involvement was first recognized in the 1960’s when post-mortem autopsy studies of patients with advanced diabetes found evidence of spinal cord atrophy, demyelination, and axonal loss (147,148). These findings were largely dismissed as being secondary to poor diabetes control and infection (e.g., syphilis) rather than DN. Indeed, the pathological abnormalities in the spinal cord were reported in isolation and not examined in the context of DN related peripheral nerve changes. Subsequent studies performed in the late 70’s and 80’s utilized advances in somatosensory evoked potentials and demonstrated central (brain and spinal cord) slowing in humans with DN (149) and rodent models (150). With the advent and accessibility of demonstrated magnetic resonance imaging in the 90’s and early 00’s, investigators were able to demonstrate clear spinal cord involvement in the form of cervical cord atrophy not only in patients with established DN (140) but also in those with early subclinical DN (139). Subsequent studies have sought to apply advances multimodal magnetic resonance imaging to gain unique insights into brain involvement, particularly brain regions involved with somatosensory and nociception in DN – e.g. primary somatosensory cortex (141) and the thalamus (142). Accompanying the reduction in cervical spine volume is a reduction in primary somatosensory cortical volume in both painful and painless DN (141). Proton magnetic resonance spectroscopy studies have demonstrated evidence of thalamic neuronal dysfunction in painless but not in painful DN – indicating that preservation of thalamic neuronal function may be a prerequisite for the perception of pain in DN (142). In addition, there was also an increase in thalamic vascularity (144), altered thalamic-cortical functional connectivity (146), and a reorganization of the primary somatosensory cortex in patients with painful DN (146). Thus, the involvement of the central nervous system in DN has opened a whole new area of further research and has great potential for future patient stratification and development of new therapeutic targets.

Figure 12. Multimodal magnetic resonance imaging studies of the central nervous system in diabetic neuropathy.

Risk Factors for Diabetic Polyneuropathies

 

Diabetic neuropathy is the end results of a culmination of several etiologically linked pathophysiological processes – some not fully understood. Although hyperglycemia and duration of diabetes play an important role in DN, other risk factors have been identified. The EURODIAB Prospective Complications study demonstrated that the incidence of DN is associated with other potentially modifiable cardiovascular risk factors, including hypertriglyceridemia, hypertension, obesity and smoking (41). In the Look AHEAD study in patients with type 2 diabetes, there was a greater increase in neuropathic symptoms (but not neuropathic signs) in the control cohort (diabetes support and education program) compared to the cohort receiving intensive diet and exercise lifestyle intervention programmed focused on weight loss (151).

 

TREATMENT OF DIABETIC POLYNEUROPATHIES

 

Treatment of DN should be targeted towards a number of different aspects: firstly, treatment of specific underlying pathogenic mechanisms; secondly, treatment of symptoms and improvement in QOL; and thirdly, prevention of progression and treatment of complications of neuropathy.

 

Targeting Risk Factors

 

GLYCEMIC AND METABOLIC CONTROL

 

Several long-term prospective studies that assessed the effects of intensive diabetes therapy on the prevention and progression of chronic diabetic complications have been published. The large randomized trials such as the Diabetes Control and Complications Trial (DCCT) and the UK Prospective Diabetes Study (UKPDS) were not designed to evaluate the effects of intensive diabetes therapy on DPN, but rather to study the influence of such treatment on the development and progression of the chronic diabetic complications (152,153). Thus, only a minority of the patients enrolled in these studies had symptomatic DPN at entry. Studies in patients with type 1 diabetes show that intensive diabetes therapy retards but does not completely prevent the development of DPN.  In the DCCT/EDIC cohort, the benefits of former intensive insulin treatment persisted for 13-14 years after DCCT closeout and provided evidence of a durable effect of prior intensive treatment on DPN and cardiac autonomic neuropathy (“hyperglycemic memory”) (154,155).

 

In contrast, in patients with type 2 diabetes, who represent the vast majority of people with diabetes, the results were largely negative. The UKPDS showed a lower rate of impaired vibration perception threshold (VPT) (VPT >25 V) after 15 years for intensive therapy (IT) vs. conventional therapy (CT) (31% vs. 52%). However, the only additional time point at which VPT reached a significant difference between IT and CT was the 9-year follow-up, whereas the rates after 3, 6, and 12 years did not differ between the groups. Likewise, the rates of absent knee and ankle reflexes as well as the heart rate responses to deep breathing did not differ between the groups (153). In the ADVANCE study including 11,140 patients with type 2 diabetes randomly assigned to either standard glucose control or intensive glucose control, the relative risk reduction (95% CI) for new or worsening neuropathy for intensive vs. standard glucose control after a median of 5 years of follow-up was −4 (−10 to 2), without a significant difference between the groups (156).  Likewise, in the VADT study including 1,791 military veterans (mean age, 60.4 years) who had a suboptimal response to therapy for type 2 diabetes, after a median follow-up of 5.6 years no differences between the two groups on intensive or standard glucose control were observed for DPN or microvascular complications (157). In the ACCORD trial (39), intensive therapy aimed at HbA1c <6.0% was stopped before study end because of higher mortality in that group, and patients were transitioned to standard therapy after 3.7 years on average. At transition, loss of sensation to light touch was significantly improved on intensive vs standard diabetes therapy. At study end after 5 years, MNSI score >2 and loss of sensation to vibration and light touch were significantly improved on intensive vs standard diabetes therapy. However, because of the premature study termination and the aggressive HbA1c goal, the neuropathy outcome in the ACCORD trial is difficult to interpret.

 

In the Steno 2 Study (158), intensified multifactorial risk intervention including intensive diabetes treatment, angiotensin converting enzyme (ACE)-inhibitors, antioxidants, statins, aspirin, and smoking cessation in patients with microalbuminuria showed no effect on DPN after 7.8 (range: 6.9-8.8) years and again at 13.3 years, after the patients were subsequently followed for a mean of 5.5 years.  However, the progression of cardiac autonomic neuropathy (CAN) was reduced by 57%. Thus, there is no evidence that intensive diabetes therapy or a target-driven intensified intervention aimed at multiple risk factors favorably influences the development or progression of DPN as opposed to CAN in patients with type 2 diabetes.  However, the Steno study used only vibration detection, which measures exclusively the changes in large fiber function.

 

DYSLIPIDEMIA  

 

Observational and cross-sectional studies have demonstrated, to varying degrees, an association between lipids and DPN (159). The strongest evidence, however, is for the association of elevated levels of triglycerides and DPN (160). In a study of patients with T2DM there was a graded relationship between triglyceride levels and the risk of lower-limb amputations (160). Likewise, another study demonstrated that hypertriglyceridemia was an independent risk factor of loss of sural (myelinated) nerve fiber density and lower limb amputations (161). In addition to hypertriglyceridemia, low-level of HDL cholesterol is reported to as an independent risk factor for DPN (159). However, clinical studies investigating the effects of statins on the development of DPN are far from conclusive. This is partly because several large statin studies that included patients with diabetes did not report data on the development of microvascular disease (162,163) let alone DPN. The Freemantle Diabetes Study, an observational study with cross-sectional and longitudinal analysis, suggested that statin or fibrate therapy may protect against DPN in T2DM (164). Two subsequent, relatively small, randomized clinical studies have reported improvements in nerve conduction parameters of DPN following 6 to 12 weeks of statin treatment (165,166). The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study has since, demonstrated that fibrates are beneficial in preventing microvascular complications (retinopathy and nephropathy) and non-traumatic lower limb amputations but DPN outcomes have not been reported (167). Subsequently, a patient registry study from Denmark, found that the use of statins before diagnosis of incident diabetes was protective against the development of DPN (168). In summary, whether lipid lowering treatment reduces the risk of DPN —a possibility raised by these data—will need to be addressed in other studies preferably in randomized controlled trials.

 

HYPERTENSION

 

An association between hypertension and DPN has been demonstrated in several observational studies in both T2DM (169,170) and T1DM (171). There is some preliminary evidence from relatively small randomized control trials with improvements in DPN based on clinical and nerve conduction parameters following antihypertensive treatment with angiotensin converting enzyme (ACE) inhibitors (172) and calcium channel blockers (173). However, the significance of this relationship is uncertain as several large intervention studies targeting hypertension (26) studies failed to show a reduction in DPN despite clear benefits in renal and retinal complications (174). One possible explanation is that the methods used in these intervention studies to diagnose/quantify DPN lacked the necessary sensitivity or reliability to diagnose/quantity DPN let alone examine differences between study groups. The heterogeneity in effect size estimates for this outcome in many of these studies supports this view. Another possible explanation for this finding could be the strengthening of guidelines for diabetes care and the more widespread routine use antihypertensive treatment.

 

OBESITY  

 

Several studies have revealed an association between obesity and polyneuropathy even in the presence of normoglycemia (175,176) The prevalence of polyneuropathy, however, increases in obese patients with prediabetes and diabetes (177). Subsequent studies appear to demonstrate that adopting a healthy lifestyle incorporating a balanced diet, regular aerobic and weight-resistance physical activities may reverse the process, particularly if they are undertaken at an early stage of DPN (136,178,179). A randomized control study of a 2.5-hour, weekly supervised treadmill exercise and dietary intervention program aimed at normalizing body mass index or losing 7% baseline body weight in T2DM demonstrated significant improvement in markers (intraepithelial nerve fiber density and regenerative capacity) of DPN (180). However, once DPN is established, restoration of normal weight did not show significant improvement.

 

Targeting Underlying Pathophysiological Mechanisms

 

OXIDATIVE STRESS

 

Several studies have shown that hyperglycemia causes oxidative stress in tissues that are susceptible to complications of diabetes, including peripheral nerves. Figure 2 presents our current understanding of the mechanisms and potential therapeutic pathways for oxidative stress-induced nerve damage. Studies show that hyperglycemia induces an increased presence of markers of oxidative stress, such as superoxide and peroxynitrite ions, and that antioxidant defense moieties are reduced in patients with diabetic peripheral neuropathy (181). Therapies known to reduce oxidative stress are therefore recommended. Therapies that are under investigation include aldose reductase inhibitors (ARIs), α-lipoic acid, γ-linolenic acid, benfotiamine, and protein kinase C (PKC) inhibitors.

 

Advanced glycation end-products (AGE) are the result of non-enzymatic addition of glucose or other saccharides to proteins, lipids, and nucleotides. In diabetes, excess glucose accelerates AGE generation that leads to intra- and extracellular protein cross-linking and protein aggregation. Activation of RAGE (AGE receptors) alters intracellular signaling and gene expression, releases pro-inflammatory molecules, and results in an increased production of reactive oxygen species (ROS) that contribute to diabetic microvascular complications. Aminoguanidine, an inhibitor of AGE formation, showed good results in animal studies but trials in humans have been discontinued because of toxicity (182).  Benfotiamine is a transketolase activator that reduces tissue AGEs. Several independent pilot studies have demonstrated its effectiveness in diabetic polyneuropathy. The BEDIP 3-week study used a 200 mg daily dose, and the BENDIP 6-week study used 300 and 600 mg daily doses; both studies demonstrated subjective improvements in neuropathy scores in the groups receiving benfotiamine, with a pronounced decrease in reported pain levels (183). In a 12-week study, the use of benfotiamine plus vitamin B6/B12 significantly improved nerve conduction velocity in the peroneal nerve along with appreciable improvements in vibratory perception. An alternate combination of benfotiamine (100 mg) and pyridoxine (100 mg) has been shown to improve diabetic polyneuropathy in a small number of patients with diabetes (184,185). The use of benfotiamine in combination with other antioxidant therapies such as α-Lipoic acid (see below) are commercially available.

 

ARIs reduce the flux of glucose through the polyol pathway, inhibiting tissue accumulation of sorbitol and fructose. In a 12-month study of zenarestat a dose dependent improvement in nerve fiber density was shown (186). In a one year trial of fidarestat in Japanese patients with diabetes, improvement of symptoms was shown (187), and a 3 year study of epalrestat showed improved nerve function (NCV) as well as vibration perception (188). Epalrestat is marketed only in Japan and India. Newer ARIs are currently being explored, and some positive results have emerged (189), but it is becoming clear that these may be insufficient per se and combinations of treatments may be needed.

 

Gamma-Linolenic acid can cause significant improvement in clinical and electrophysiological tests for neuropathy (190). Alpha-Lipoic acid or thioctic acid has been used for its antioxidant properties and for its thiol-replenishing redox-modulating properties. A number of studies show its favorable influence on microcirculation and reversal of symptoms of neuropathy (191,192). A meta-analysis including 1,258 patients from four randomized clinical trials concluded that 600 mg of i.v. α-lipoic acid daily significantly reduced symptoms of neuropathy and improved neuropathic deficits (193). The SYDNEY 2 trial showed significant improvement in neuropathic symptoms and neurologic deficits in 181 diabetes patients with 3 different doses of α-lipoic acid compared to placebo over a 5-week period (194). The long-term effects of oral α-lipoic acid on electrophysiology and clinical assessments were examined during the NATHAN-1 study.  The study showed that 4 years of treatment with α-lipoic acid in mild to moderate DSP is well tolerated and improves some neuropathic deficits and symptoms, but not nerve conduction (195). Additional long-term RCTs could further strengthen the rationale for the use of these agents in clinical practice. Safety profiles of α-lipoic acid are favorable during long-term treatment. An overview on the usual dosages of α-lipoic acid and benfothiamine, most frequent adverse events and scientific evidence can be found here (193,196,197,185).

 

Protein kinase C (PKC) activation is a critical step in the pathway to diabetic microvascular complications. It is activated by both hyperglycemia and disordered fatty-acid metabolism, resulting in increased production of vasoconstrictive, angiogenic, and chemotactic cytokines including transforming growth factor β (TGF-β), vascular endothelial growth factor (VEGF), endothelin (ET-1), and intercellular adhesion molecules (ICAMs). A multinational, randomized, phase-2, double blind, placebo-controlled trial with ruboxistaurin (a PKC-β inhibitor) failed to achieve the primary endpoints although significant changes were observed in a number of domains (198). Nevertheless, in a subgroup of patients with less severe DN (sural nerve action potential greater than 0.5 μV) at baseline and clinically significant symptoms, a statistically significant improvement in symptoms and vibratory detection thresholds was observed in the ruboxistaurin-treated groups as compared with placebo (199). A smaller, single center study showed improvement in symptom scores, endothelium dependent skin blood flow measurements, and quality of life scores in the ruboxistaurin treated group (200). These studies and the NATHAN studies have pointed out the change in the natural history of DPN with the advent of therapeutic lifestyle change, statins and ACE inhibitors, which have slowed the progression of DPN and drastically altered the requirements for placebo-controlled studies. Several studies (201,202) have demonstrated that patients with type 1 diabetes who retain some β-cell activity are considerably less prone to developing microvascular complications than those who are completely C-peptide deficient, and that C-peptide may have substantial anti-oxidant, cytoprotective, anti-anabolic, and anti-inflammatory effects.  C-peptide administration for 6 months in type 1 diabetes has been shown to improve sensory nerve function (203).

 

GROWTH FACTORS  

 

There is increasing evidence that there is a deficiency of nerve growth factor (NGF) in diabetes, as well as the dependent neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) and that this contributes to the clinical perturbations in small-fiber function (204). Clinical trials with NGF have not been successful but are subject to certain caveats with regard to design; however, NGF still holds promise for sensory and autonomic neuropathies (205). The pathogenesis of DN includes loss of vasa nervorum, so it is likely that appropriate application of vascular endothelial growth factor (VEGF) would reverse the dysfunction. Introduction of VEGF gene into the muscle of DM animal models improved nerve function (206). However, VEGF gene studies with transfection of the gene into the muscle in humans failed to meet efficacy end points in painful DPN trials 207. Hepatocyte growth factor (208,209) (HGF) is another potent angiogenic cytokine under study for the treatment of painful neuropathy.  INGAP peptide comprises the core active sequence of Islet Neogenesis Associated Protein (INGAP), a pancreatic cytokine that can induce new islet formation and restore euglycemia in diabetic rodents. Maysinger et al showed significant improvement in thermal hypoalgesia in diabetic mice after a 2-week treatment with INGAP peptide (210,211).

 

IMMUNE THERAPY

 

Several different autoantibodies in human sera have been reported that can react with epitopes in neuronal cells and have been associated with DN.  Milicevic et al have reported a 12% incidence of a predominantly motor form of neuropathy in patients with diabetes associated with monosialoganglioside antibodies (anti GM1 antibodies) (63). Perhaps the clearest link between autoimmunity and neuropathy has been the demonstration of an 11-fold increased likelihood of CIDP, multiple motor polyneuropathy, vasculitis, and monoclonal gammopathies in diabetes (61). New data, however, support a predictive role of the presence of antineuronal antibodies on the later development of neuropathy, suggesting that these antibodies may not be innocent bystanders but neurotoxins (212). There may be selected cases, particularly those with autonomic neuropathy, evidence of antineuronal autoimmunity, and CIDP, that may benefit from intravenous immunoglobulin or large dose steroids (59).

 

Summary of Treatment of Diabetic Peripheral Neuropathy

 

In summary, the risk factors for DPN are well recognized and to-date only small-scale intervention studies targeting these risk factors that have used appropriate DPN biomarkers have been conducted. Nevertheless, these have provided preliminary evidence for the efficacy of multifactorial risk factor management in preventing the development and progression of DPN. Hence, early identifications of subjects with insipient/sub-clinical neuropathy using validated, yet novel non-invasive point of care devices will allow larger studies to determine if targeted intensified cardiometabolic risk factor control can prevent clinical DPN or halt disease progression. Unfortunately, despite several clinical trials, there has been relatively little progress in the development of disease modifying treatments despite some advances in the management of symptoms in painful DN, as described below.

 

PAINFUL DIABETIC PERIPHERAL NEUROPATHY

 

Pathogenesis

 

Peripheral neuropathic pain in diabetes is defined as “pain arising as a direct consequence of abnormalities in the peripheral somatosensory system” after exclusion of other causes (213). Nerve damage results in the release of inflammatory mediators which activate intracellular signal transduction pathways in the nociceptor terminal, prompting an increase in the production, transport, and membrane insertion of transducer channels and voltage-gated ion channels (214). Following nerve injury, different types of voltage-gated sodium and calcium channels are up-regulated at the site of the lesion and in the dorsal root ganglion membrane, promoting ectopic spontaneous activity along the primary afferent neuron and determining hyperexcitability associated with lowered activation threshold, hyper-reactivity to stimuli, and abnormal release of neurotransmitters such as substance P and glutamate (215, 216). As a consequence of this hyperactivity in primary afferent nociceptive neurons, important secondary changes may occur in the dorsal horn of the spinal cord and higher up in the central nervous system leading to neuron hyperexcitability. This phenomenon, called central sensitization, is a form of use-dependent synaptic plasticity, considered a major pathophysiological mechanism of neuropathic pain (217).

 

Diagnosis

 

Painful DPN is often underdiagnosed and under treated. Binns-Hall et al. trialed a ‘one-stop’ microvascular screening service, which tested a model for patients to receive combined eye, foot (DPN and painful-DPN), and renal screening (218). A new diagnosis of painful-DPN in this cohort was identified in 25% of participants using the validated screening tool for neuropathic pain, the Doleur Neuropathique en 4 Questions (DN4). Additionally, Daousi et al. found that in a community sample of 350 patients with diabetes 12.5% of patients with painful-DPN had not reported their symptoms to their treating physician (219). This study also found that 39.3% had never received treatment for their painful neuropathy. In the clinical environment, most cases of painful DPN can be diagnosed with a careful history to identify presence of typical painful neuropathic symptoms lasting > 3 months and clinical examination to demonstrate the clinical signs of DPN. In these circumstances and other causes are excluded (see above), there is no need for further investigations.

 

A number of self-administered questionnaires have been developed, validated, translated, and subjected to cross-cultural adaptation both to diagnose and distinguish neuropathic as opposed to non-neuropathic pain  (screening tools such as the Leeds Assessment of Neuropathic Symptoms and Signs (LANSS) Pain Scale (220), Douleur Neuropathique en 4 questions (DN4), Neuropathic Pain Questionnaire (NPS) (221), pain DETECT (89) and to assess pain quality and intensity such as the Short-Form McGill Pain Questionnaire (222), the Brief Pain Inventory (BPI) (223), and the Neuropathic Pain Symptom Inventory (NPSI) (224).

 

It is important to assess the intensity (severity) of neuropathic pain as it is helpful when assessing and monitoring response to therapy. The best approach is to use a simple 11-Point numerical rating scale (Likert scale) or a visual analogue scale. In clinical trials of neuropathic pain treatment a number of questionnaires are used to capture the complex, multidimensional impact of chronic pain. According to IMMPACT (Initiative on Methods, Measurement and Pain Assessment in Clinical Trials) the following assessments are performed to assess the efficacy and effectiveness of new treatments: 1. pain intensity measured on a 0 to 10 numerical rating scale (NRS); 2. physical functioning assessed by the Multidimensional Pain Inventory (MPI) and Brief Pain Inventory (BPI) Interferences scale; 3. emotional functioning, assessed by the Beck Depression Inventory (BPI) and Profile of Mood states; and 4. patient rating of overall improvement, assessed by the Patient Global Impression of Change (PGI-C) (225).

 

Quality of Life

 

Over time the persistence of extremely unpleasant painful symptoms can have a profound impact upon its sufferers’ lives. This often results in a poor quality of life (226), disruption of employment (227), and mood disturbance (13). This adds to the burden of suffering and increases the challenge of managing neuropathic effectively. This is further compounded when patients also suffer from other co-morbid conditions associated with diabetes. Painful-DPN is also an expensive condition, incurring high healthcare costs (228). Data from the US found that patients with DPN and painful-DPN have greater healthcare resource utilization and costs than those with diabetes alone (228). Patients with severe painful-DPN incurred five-fold higher annual direct medical costs (USD $30,755) than for patients with diabetes alone (USD $6632) (226).

 

Sensory Profiling  

 

For many years, sensory profiling has been the mainstay for identifying a homogenous subgroup of neuropathic pain patients in clinical pain research. The basis of this approach is that painful symptoms reflect specific pathophysiological mechanisms, which are present to varying degrees in individual patients (229,230). Detailed sensory profiling using quantitative sensory testing (QST) can be used to subgroup patients into more homogenous cohorts (pain phenotypes), which could then be targeted with treatments known to act specifically on pathophysiological pathways underlying the phenotypes (231) (Figure 13). QST refers to a battery of standardized, psychophysical tests (e.g., thermal testing, pin prick, pressure algometry, and von Frey filaments) used to assess central and peripheral nervous system sensory function (232). In DPN, QST has been used for several decades mainly for diagnosing and quantifying the extent of small and large nerve fiber impairment in individuals predominantly with painless DPN. In the context of pain somatosensory phenotyping, a standardized QST protocol was developed by the German Research Network on Neuropathic Pain (DFNS), which includes 12 sensory testing parameters (i.e., cold and warm detection thresholds, paradoxical heat sensations, thermal sensory limen procedure, cold and heat pain thresholds, mechanical detection threshold, mechanical pain threshold, mechanical pain sensitivity, dynamic mechanical allodynia, wind-up ratio, vibration detection threshold, and pressure pain threshold) (232). The positive and negative results of individual patients are obtained by comparison against a normative QST reference dataset, comprised of age- and sex-stratified healthy individuals.

Figure 13. Schematic representation of the generation of pain. (A) Normal: Central terminals of c-afferents project into the dorsal horn and make contact with secondary pain-signaling neurons. Mechanoreceptive Aβ afferents project without synaptic transmission into the dorsal columns (not shown) and also contact secondary afferent dorsal horn neurons. (B) C-fiber sensitization: Spontaneous activity in peripheral nociceptors (peripheral sensitization, black stars) induces changes in the central sensory processing, leading to spinal-cord hyperexcitability (central sensitization, gray star) that causes input from mechanoreceptive Aβ (light touch) and Aδ fibers (punctuate stimuli) to be perceived as pain (allodynia). (C) C-fiber loss: C-nociceptor degeneration and novel synaptic contacts of Aβ fibers with “free” central nociceptive neurons, causing dynamic mechanical allodynia. (D) Central disinhibition: Selective damage of cold-sensitive Aδ fibers that leads to central disinhibition, resulting in cold hyperalgesia. Sympat, sympathetic nerve

Two Distinct Pain Phenotypes – The Non-Irritable and Irritable Nociceptor

 

Application of the QST technique has shown that there are two distinct subgroups of patients who have particular patterns of sensory symptoms and signs: (a) a predominant differentiation with loss of sensory function (non-irritable nociceptor phenotype), and (b) a relatively preserved small fiber function associated with thermal/mechanical hypersensitivity (irritable nociceptor phenotype) (231). Using the DFNS protocol, the PiNS reported that the non-irritable nociceptor was the predominant phenotype in painful DPN, whilst only a minority of patients had the irritable nociceptor phenotype (6.3%) (233). Nevertheless, a small but significant proportion of patients (15%) did demonstrate signs of sensory gain with dynamic mechanical allodynia, often in combination with hyposensitivity across a range of small and large nerve fiber sensory assessments. The presence of allodynia would suggest that aberrant central processing of sensory inputs has an important role in these patients. Recent studies have demonstrated proof-of-concept for using sensory profiling to improve clinical trial efficiency by demonstrating that some treatments are more effective in patients with the irritable versus the non-irritable nociceptor phenotype (230-234). However, most of these studies examined patients with peripheral neuropathy of diverse causes.

 

Phenotype-Driven Therapeutic Experience in Painful DPN

 

Examples of studies that focused on painful DPN include an open label retrospective study using the DFNS protocol, which evaluated key phenotypic differences in sensory profiling associated with response to intravenous lidocaine in patients with severe, intractable painful DPN (235). Patients with the irritable nociceptor phenotype were more likely to respond to intravenous lidocaine, which inactivates sodium channels, compared to the non-irritable nociceptor phenotype (235). In fact, dynamic mechanical allodynia and pain summation to repetitive pinprick stimuli were the only evoked ‘gain of function’ QST parameters that informed treatment response. The presence of these sensory gain parameters suggests aberrant central processing with hyperexcitable neurons driven by abnormal sodium channel regulation, generating ectopic impulses and amplifying afferent sensory inputs. In another painful DPN study by Campbell et al. of topical clonidine, sensory profiling was performed using the capsaicin challenge test (236). The post-hoc analysis demonstrated a significant reduction in pain in the patient subgroup with increased spontaneous pain following cutaneous capsaicin administration, indicating the presence of functioning and sensitized nociceptors. Bouhassira et al. published post-hoc analysis data of treatment response based on sensory profiling using the Neuropathic Pain Symptom Inventory (NPSI) questionnaire from the Combination vs Monotherapy of pregabalin and duloxetine in Diabetic Neuropathy (COMBO-DN) study (237). This study examined the effect of high-dose duloxetine, a serotonin noradrenaline reuptake inhibitor, or pregabalin, a calcium channel blocker, as monotherapy versus combined pregabalin and duloxetine for painful DPN. The investigators showed that adding pregabalin (300 mg) to duloxetine (60 mg) improved the dimensions of ‘pressing pain’ and ‘evoked pain’ more significantly. On the other hand, increasing duloxetine from 60 mg to 120 mg daily improved the dimension ‘paresthesia/dysesthesia’ to a greater extent.

 

SENSORY PHENOTYPING TO PREDICT THERAPEUTIC RESPONSE

 

In a randomized, double-blind, placebo-controlled, and phenotype-stratified study of patients with painful DPN Demant et al. reported that oxcarbazepine was more efficacious for relief of peripheral neuropathic pain in patients with the irritable vs the nonirritable nociceptor phenotype (234).  Based on this and other recent studies, current opinion with regard to neuropathic pain clinical trials recommends a detailed sensory profiling of participants at baseline; and even if there is no significant separation of a drug with placebo, a subgroup analysis can be performed to see if the drug was efficacious in a particular subgroup. If there is a clear signal that this was the case, a further, adequately powered, phenotype stratified trial would be designed.   

 

Sensory profiling can also identify subgroups with altered endogenous pain modulation to predict treatment outcomes of drugs and other interventions that affect a given mechanism. Figure 14 describes the different nerve fibers affected and possible targeted treatments.

 

In a study of pain modulation in DPN, individuals were assessed using QST for conditioned pain modulation (CPM), a psychophysical paradigm in which central pain inhibition is measured via the phenomenon of ‘pain inhibiting pain,’ via the simultaneous administration of a conditioning painful stimulus at a distant body site. The pain in participants with abnormal CPM was more receptive to duloxetine, which is believed to increase descending inhibitory pain pathway activation, than individuals with normal pain modulation, although there was no comparison to placebo in this open-label study (238).

Figure 14. Schematic representation of the generation of pain. (A) Normal: Central terminals of c-afferents project into the dorsal horn and make contact with secondary pain-signaling neurons. Mechanoreceptive Aβ afferents project without synaptic transmission into the dorsal columns (not shown) and also contact secondary afferent dorsal horn neurons. (B) C-fiber sensitization: Spontaneous activity in peripheral nociceptors (peripheral sensitization, black stars) induces changes in the central sensory processing, leading to spinal-cord hyperexcitability (central sensitization, gray star) that causes input from mechanoreceptive Aβ (light touch) and Aδ fibers (punctuate stimuli) to be perceived as pain (allodynia). (C) C-fiber loss: C-nociceptor degeneration and novel synaptic contacts of Aβ fibers with “free” central nociceptive neurons, causing dynamic mechanical allodynia. (D) Central disinhibition: Selective damage of cold-sensitive Aδ fibers that leads to central disinhibition, resulting in cold hyperalgesia. Sympat, sympathetic nerve

Taken together, these studies support the notion that mechanism-based approaches to pain management may be feasible in painful DPN. However, in an elegant mechanistic study, Haroutounian et al examined 14 patients with neuropathic pain of mixed etiology [unilateral foot pain from nerve injury (n=7) and distal polyneuropathy (n=7)] to determine the contribution of primary afferent input in maintaining peripheral neuropathic pain (239). Each patient underwent randomized ultrasound-guided peripheral nerve block with lidocaine versus intravenous lidocaine infusion. They found that peripheral afferent input was critical for maintaining neuropathic pain, but improvement in evoked hypersensitivity was not related to improvements in spontaneous pain intensity. This suggests that further studies are needed to rationalize sensory phenotyping in order to optimize clinical trial outcomes in painful DPN. Moreover, given the rarity of the irritable-nociceptor phenotype, as determined by QST, a single assessment modality may be unlikely to help stratify patients and combining with additional modalities may be necessary (e.g., brain imaging). 

 

Brain Imaging in Painful Diabetic Peripheral Neuropathy

 

Recent advances in neuroimaging provide us with unique insights into the human central nervous system in chronic pain conditions (240). We now have a better understanding how the brain modulates nociceptive inputs to generate the pain experience, and how this is disrupted in patients with painful DPN. However, to date, brain imaging serves mainly as a research tool, with minimal direct application in clinical trials for pain or clinical practice. While mechanistic approaches that require carefully evaluating specific responses to guide therapy have significant appeal (e.g., cold, heat, von Frey etc.), in practice, these are time consuming and may be difficult to implement in busy clinical practices. Furthermore, these are psychophysical measures which rely on patient responses and may be subjective and biased. Sensory profiling methods also do not capture the complex and multidimensional pain experience, which affects emotional and cognitive processing in addition to sensory processing. For example, chronic pain patients often undergo neuropsychological changes, which include changes in emotion and motivation or changes in cognition (241). Chronic pain may also arise after the onset of depression, even in patients without a prior history of pain or depression. Collectively, these clinical insights suggest a better strategy for assessing and treating painful DPN, given it is a chronic disease of dynamic process (e.g., evolution of co-morbid phenotypes such as anxiety or depression), which is not easily reversed in most patients. It is important to determine specific targets that are relevant to pain across individuals, because modulating activation in these targets may provide evidence that a compound engages a target or attenuates nociceptive processing.

 

Structural and functional cortical plasticity is a fundamental property of the human central nervous system, which can adjust to nerve injury. However, it can have maladaptive consequences, possibly resulting in chronic pain. Studies using structural magnetic resonance (MR) neuroimaging have demonstrated a clear reduction in both spinal cord cross-sectional area (139) and primary somatosensory cortex (S1) gray matter volume in patients with DPN (141). These findings are supported by studies in other pain conditions, which have also reported dynamic structural and functional plasticity with profound effects on the brain in patients with neuropathic pain. More recently, it has been demonstrated how brain structural and functional changes are related to painful DPN clinical phenotypes (146). Patients with the painful insensate phenotype have a more pronounced reduction in S1 cortical thickness and a remapping of S1 sensory processing compared to painful DPN subjects with relatively preserved sensation (146). Furthermore, the extent to which S1 cortical structure and function is altered is related to the severity of neuropathy and the magnitude of self-reported pain. These data suggest a dynamic plasticity of the brain in DPN driven by the neuropathic process and may ultimately determine an individual’s clinical pain phenotypes.

Over the last decade, resting-state functional MR imaging (RS-fMRI) – a quick, and simple non-invasive technique – has become an increasingly appealing way to examine spontaneous brain activity in individuals, without relying on external stimulation tasks. During a typical RS-fMRI examination, the hemodynamic response to spontaneous neuronal activity (bold oxygen level dependent, BOLD) signal is acquired whilst subjects are instructed to simply rest in the MR scanner (242). The data acquired is used in brain mapping to evaluate regional interactions or functional connectivity, which occur in a resting state. Most studies use a machine learning approach to identify patterns of functional connectivity, which differentiates patients from controls. RS-fMRI experiments in painful DPN have reported greater thalamic-insula functional connectivity and decreased thalamic-somatosensory cortical functional connectivity in patients with the irritable versus non-irritable nociceptor phenotype (235). There was a significant positive correlation between thalamic-insula functional connectivity with self-reported pain scores (235). Conversely, there was a more significant reduction in thalamic-somatosensory cortical functional connectivity in those with more severe neuropathy. This demonstrates how RS-fMRI measures of functional connectivity relates to both the somatic and non-somatic assessments of painful DPN. In one study, using a machine learning approach to integrate anatomical and functional connectivity data achieved an accuracy of 92% and sensitivity of 90%, indicating good overall performance (235). Multimodal MR imaging combining structural and RS-fMRI has also been used to predict treatment response in painful DPN. Responders to intravenous lidocaine treatment have significantly greater S1 cortical volume and greater functional connectivity between the insular cortex and corticolimbic system compared to non-responders (235). The insular cortex plays a pivotal role in processing the emotion and cognitive dimensions of the chronic pain experience. The corticolimbic circuits have also long been implicated in reward, decision making, and fear learning. Hence, these findings suggest that this network may have a role in determining treatment response in painful DPN.

 

Using advanced multimodal MR neuroimaging, a number of studies have demonstrated alterations in pain processing brain regions, which relate to clinical pain phenotype, treatment response, and behavioral/psychological factors impacted by pain. Taken together, these assessments could serve as a possible Central Pain Signature for painful DPN. The challenge now is to apply this potential pain biomarker at an individual level in order to demonstrate clinical utility. To this end, applying machine learning (243) to leverage brain imaging features from a quick 6-minute RS-fMRI scan to classify individual patients into different clinical pain phenotypes is appealing. Future studies should externally validate and optimize current models in larger patient cohorts to examine if/how such models can be used as biomarkers in clinical trials of pain therapeutics. Although many of the findings described are consistent with neuroimaging studies in other chronic pain conditions, it is difficult to assess convergence of findings across a number of relatively small cohort studies employing different analytical methods to derive complex models involving a large number of distributed brain regions (244). These are important limitations that are being addressed with 1) a number of large scale multi-center studies in progress or in preparation (MAPP consortium (245) and Placebo Imaging consortium (246), and 2) several consensus statements by key stakeholders, promoting standardized approaches and reporting and transparent/sharable models. 

 

General Principals of Managing Painful DPN

 

Managing painful symptoms in DPN may constitute a considerable treatment challenge. The efficacy of a single therapeutic agent is not the rule, and most patients require combination therapy to control the pain. The present ‘trial and error’ approach is to offer the available therapies in a stepwise fashion until an effective treatment is achieved (247,248). Effective pain treatment considers a favorable balance between pain relief and side effects without implying a maximum effect. The following general considerations in the pharmacotherapy of neuropathic pain require attention (249):

 

  • The appropriate and effective drug has to be tried and identified in each patient by carefully titrating the dose based on efficacy and side effects.
  • Lack of efficacy should be judged only after 2-4 weeks of treatment using an adequate dose.
  • As the evidence from clinical trials suggests a > 50% reduction in pain for any monotherapy, combination therapy is considered a ‘robust’ response. A reduction of pain of 30-49% may be considered a ‘clinically relevant’ response.
  • Potential drug interactions have to be considered given the frequent use of polypharmacy in patients with diabetes.

 

For many patients, optimal management of chronic pain may require a multidisciplinary team approach with appropriate behavioral therapy, as well as input from a broad range of healthcare professionals. Here we highlight the common agents used to manage painful DPN and key papers to demonstrate the evidence base. The most recent guidelines for pharmacotherapy for neuropathic pain in general and specifically in painful DPN can be found elsewhere (16,250,251,252,253,254,67, 255,256).

 

ANTIDEPRESSANTS 

 

Antidepressants are commonest agents used in the treatment of chronic neuropathic pain (217). The putative mechanisms of interrupting pain transmission by these agents include inhibition of norepinephrine and/or serotonin reuptake within the endogenous descending pain-inhibitory systems in the brain and spinal cord (257). Antagonism of N-Methyl-D-Aspartate receptors that mediate hyperalgesia and allodynia has also been proposed.

 

Tricyclic Antidepressants (TCAs)

 

Imipramine, amitriptyline, and clomipramine induce a balanced reuptake inhibition of both norepinephrine and serotonin, while desipramine is a relatively selective norepinephrine inhibitor. The most frequent adverse events of tricyclic antidepressants (TCAs) are anticholinergic symptoms including tiredness and dry mouth and may exacerbate cardiovascular and gastrointestinal autonomic neuropathy. The starting dose should be 25 mg (10 mg in frail patients) taken as a single night time dose one hour before sleep. The maximum dose is usually 150 mg per day and doses >100mg should be avoided in the elderly.

 

TCAs should be used with caution in patients with orthostatic hypotension and are contraindicated in patients with unstable angina, recent (<6 months) myocardial infarction, closed-angle glaucoma, heart failure, history of ventricular arrhythmias, significant conduction system disease, and long QT syndrome. Their use is limited by relatively high rates of adverse events and several contraindications.

 

Serotonin Noradrenaline Reuptake Inhibitors (SNRI)

 

The efficacy and safety of duloxetine has been evaluated in 7 RCTs establishing it as a mainstay treatment option in painful DPN. Several systematic reviews demonstrate a moderate strength of evidence for duloxetine reduces neuropathic pain to a clinically meaningful degree in patients with painful DPN (258,259,260). Patients with higher pain intensity tend to respond better than those with lower pain levels. The most frequent side effects of duloxetine (60/120 mg/day) include nausea (16.7/27.4%), somnolence (20.2/28.3%), dizziness (9.6/23%), constipation (4.9/10.6%), dry mouth (7.1/15%), and reduced appetite (2.6/12.4%). These adverse events are usually mild to moderate and transient. To minimize them the starting dose should be 30 mg/day for 4-5 days. In contrast to TCAs and some anticonvulsants, duloxetine does not cause weight gain, but a small increase in fasting blood glucose may occur (261).

 

Venlafaxine is another SNRI that has mixed action on catecholamine uptake. Compared to duloxetine, the strength of evidence for venlafaxine is lower and it could be considered an alternative if duloxetine is not tolerated. At lower doses, venlafaxine inhibits serotonin uptake and at higher doses it inhibits norepinephrine uptake (262). The extended release version of venlafaxine was found to be superior to placebo in painful DPN in non-depressed patients at doses of 150-225 mg daily, and when added to gabapentin there was improved pain, mood, and quality of life (263).  In a 6-week trial comprised of 244 patients the analgesic response rates were 56%, 39%, and 34% in patients given 150-225 mg venlafaxine, 75 mg venlafaxine, and placebo, respectively. Because patients with depression were excluded, the effect of venlafaxine (150-225 mg) was attributed to an analgesic, rather than antidepressant, effect. The most common adverse events were tiredness and nausea (264); additionally, clinically important electrocardiogram changes were found in seven patients in the treatment arm.

 

ANTI-EPILEPTIC DRUGS

 

Calcium Channel Modulators (a2-δ ligands)

 

Gabapentin is an anticonvulsant structurally related to g-aminobutyric acid (GABA), a neurotransmitter that plays a role in pain transmission and modulation. The exact mechanisms of action of this drug in neuropathic pain are not fully elucidated. Among others, they involve an interaction with the L-amino acid transporter system and high affinity binding to the a2-δ subunit of voltage-activated calcium channels. A Cochrane review reported 4 out of 10 patients with painful DPN achieved greater than 50% pain relief with gabapentin compared to placebo (2 out of 10). Pain was reduced by a third or more for 5 in 10 with gabapentin and 4 in 10 with placebo. Over half of those treated did not benefit from worthwhile pain relief but experienced adverse event (265).

 

In contrast to gabapentin, pregabalin is a more specific a2-δ ligand with a 6-fold higher binding affinity. It also has a more rapid onset with a dose-dependent linear pharmacokinetic profiled i.e., 600mg/day being more effective that 300mg/day (266). Hence, the administration (BD vs QDS) and dose titration of pregabalin in considerably easier compared to gabapentin. A recent Cochrane review reported moderate quality evidence for the efficacy of pregabalin in painful DPN compared to placebo (267). 3 or 4 in 10 people had pain reduced by half or more with pregabalin 300 mg or 600 mg daily, and 2 or 3 in 10 with placebo. Pain was reduced by a third or more for 5 or 6 in 10 people with pregabalin 300 mg or 600 mg daily, and 4 or 5 in 10 with placebo.

 

Common side-effects associated with the use of gabapentinoids include weight gain, edema, dizziness, and somnolence. They should be used with caution in patients with congestive cardiac failure (NYHA class III or IV) and renal impairment (dose reduction required). Pooled trial analysis of adverse events showed a higher risk of side-effects with increasing pregabalin dose but not older age (268). The misuse and abuse of gabapentinoids is a growing problem in the US and in Europe necessitating monitoring for signs of misuse/abuse and caution when used in at risk populations (269). Gabapentinoids may also increase the risk of respiratory depression, a serious concern for patients taking opioids or with underlying respiratory impairment (270,271,272).

 

TOPICAL CAPSAICIN

 

C-fibers utilize the neuropeptide substance P as their neurotransmitter, and depletion of axonal substance P (through the use of capsaicin) will often lead to amelioration of the pain. Prolonged application of capsaicin, a highly selective agonist of transient receptor potential vanilloid-1 (TRPV1), depletes stores of substance P, and possibly other neurotransmitters, from sensory nerve endings. This reduces or abolishes the transmission of painful stimuli from the peripheral nerve fibers to the higher centers (273). The 8% capsaicin patch (Qutenza) (274) is authorized for the treatment of peripheral neuropathic pain. In one RCT in painful DPN, a single application of 8% capsaicin patch applied for 30min provided modest pain relief for up to 3 months (275). Specialist trained staff are required for application which can be repeated every 2-3 months. A Cochrane review of low dose (0.025% and 0.075%) topical capsaicin cream was not able to provide any recommendations due to insufficient data (276).

 

LIDOCAINE 

 

Lidocaine has unique analgesic properties. Although the exact mechanism by which intravenous lidocaine provides systemic analgesia is unknown, it is thought to have both peripheral and central mechanisms of action (277,278,279). It exhibits state-dependent binding where sodium channels that are rapidly and repeatedly activated and inactivated are more readily blocked (280). This state-dependence is thought to be very important in limiting the hyperexcitability of cells exhibiting abnormal activity. Thus, it is likely to have greater efficacy in patients with neuropathic pain (281,282) and has been used to relieve chronic pain for over 50 years (283). A Cochrane review of 30 RCT found that intravenous lidocaine (284), which is more effective than its oral analogue (mexilitine, NNT10-38) and gastrointestinal intolerance most common side effect and major factor limiting its use) (284,285) and is more effective than placebo in decreasing neuropathic pain. It was found to be generally well tolerated with little or no side effects (286). Hence, intravenous lidocaine is a recognized treatment option for patients with severe painful DPN (287), and is included in clinical guidelines (288).

 

Although 5% lidocaine patch is being used in patients with postherpetic neuralgia (289), there is insufficient evidence to recommend its use in those with painful DPN.

 

OPIOIDS

 

Tramadol and NMDA Receptor Antagonists

 

The most examined compounds in painful DPN are tramadol, oxycodone, and tapentadol. Tramadol is a centrally acting weak opioid and SNRI for use in treating moderate to severe pain.  More severe pain requires administration of strong opioids such as oxycodone (µ-opioid agonist) or tapentadol (µ-opioid agonist and SNRI).  There is limited data available on the efficacy of these agents from relatively small-scale studies. Recent Cochrane reviews graded the available evidence as mostly of low or very low quality and likely to overestimate the efficacy of tramadol and oxycodone in the treatment of painful DPN (290,291). Side effects typical of opioids were common including somnolence, headache, and nausea. There is an increased risk of serotonergic syndrome if tramadol and tapentadol are prescribed with other agents with serotonin reuptake inhibitor properties and thus best avoided. Nevertheless, there is role for these agents as 2nd or 3rd line analgesics for painful DPN in carefully selected patients unresponsive to standard treatments. Non-pharmacological and non-opioid analgesic treatments should be optimized and established and/or not tolerated/contraindicated before opioid treatment is considered (292). Regular monitoring/evaluation of efficacy is recommended particularly if treatment is longer than 3 months. Opioids are associated with less pain relief during longer trials possibly due to opioid tolerance or opioid induced hyperalgesia. Moreover, adverse outcomes such as dependence, overdose, depression, and impaired functional status were more common in patients with neuropathic pain (painful DPN 68%) receiving long-term (>90 days) vs short term (<90 days) of treatment (293). Hence, referral to specialist or centers with experience in opioid use is recommended to avoid risks.

 

PSYCHOLOGICAL SUPPORT 

 

A psychological component to pain should not be underestimated. Hence, an explanation to the patient that even severe pain may remit, particularly in poorly controlled patients with acute painful neuropathy or in those painful symptoms precipitated by intensive insulin treatment. Thus, the empathetic approach addressing the concerns and anxieties of patients with neuropathic pain is essential for their successful management (294).

 

PHYSICAL MEASURES

 

The temperature of the painful neuropathic foot may be increased due to arterio-venous shunting. Cold water immersion may reduce shunt flow and relieve pain. Allodynia may be relieved by wearing silk pajamas or the use of a bed cradle. Patients who describe painful symptoms on walking as comparable to walking on pebbles may benefit from the use of comfortable footwear (255).

 

ACUPUNCTURE

 

A 10-week uncontrolled study with a follow-up period of 18-52 weeks in patients with diabetes showed significant pain relief after up to 6 courses of traditional Chinese acupuncture without any side effects (295). A single-blind placebo-controlled randomized trial of acupuncture in 45 subjects with painful DN recently reported an improvement in the outcome measures assessing pain in the acupuncture arm relative to sham treatment (296). However, Chen and colleagues warn that design flaws and lack of robust outcome measures of pain in acupuncture trials make meaningful conclusions difficult (297).  Larger controlled studies are needed to confirm these early findings.

 

ELECTRICAL STIMULATION

 

Transcutaneous electrical nerve stimulation (TENS) influences neuronal afferent transmission and conduction velocity, increases the nociceptive flexion reflex threshold, and changes the somatosensory evoked potentials. In a 4-week study of TENS applied to the lower limbs, each for 30 minutes daily, pain relief was noted in 83% of the patients compared to 38% of a sham-treated group. In patients who only marginally responded to amitriptyline, pain reduction was significantly greater following TENS given for 12 weeks as compared with sham treatment. Thus, TENS may be used as an adjunctive modality combined with pharmacotherapy to augment pain relief  (298).

 

Frequency-modulated electromagnetic nerve stimulation (FREMS) in 2 studies, including a recent double-blind randomized placebo controlled trial with 51 weeks of follow-up, proved to be a safe treatment for symptomatic diabetic neuropathy, with immediate but transient reduction in pain and no effect on nerve conduction velocities (299,300).  Six out of eight trials analyzed in a recent review evaluating the use of electrical stimulation in painful DN found significant pain relief in patients treated with electrical stimulation compared with placebo or sham treatment (301). 

 

Electrical spinal cord stimulation (SCS) was first reported in painful DPN in 1996 (302). With electrodes implanted between T9 and T11, 8 out of 10 patients reported greater than 50% pain relief. Most of these early devices utilized low-frequency stimulation (40-60Hz) with two RCTs demonstrating moderate utility (n=36 to 60) with 6-month to 24-month follow up (303,304,305) with responder attrition within 12 months (306). Modern iterations of SCS employ high-frequency stimulation (10kHz) provides pain relief without generating paresthesia (307,308,309,310). A recent RCT examine the use of 10kHz electrical SCS in patients with refractory painful DPN compared to conventional medical management in 216 randomized patients (311). 50% reduction in pain relief was observed in 5% in the control group compared to 79% in the electrical SCS group with 6 months follow up. The main limitation of this study was the lack of blinding and potential for placebo effects as an important confounding factor. Nevertheless, this is an interesting finding which should open a new area for further research. Overall complications of electrical SCS include wound infection and lead migration requiring reinsertion. Currently, therefore, this invasive treatment option should be reserved for patients who do not respond to analgesic combination pharmacotherapy.

 

SURGICAL DECOMPRESSION

 

Surgical decompression at the site of anatomic narrowing has been promoted as an alternative treatment for patients with symptomatic DPN. A systematic review of the literature revealed only Class IV studies concerning the utility of this therapeutic approach. Given the current evidence available, this treatment alternative should be considered unproven. Prospective randomized controlled trials with standard definitions and outcome measures are necessary to determine the value of this therapeutic intervention (312,313).

 

The odds ratios for efficacy of neuropathic pain medications are given in Figure 15. In addition, Table 5 shows the dosages of the different drugs and the commonly encountered side effects.

Figure 15. Efficacy analysis of drugs used for the treatment of PDN

Guidelines for Pharmacotherapy of Painful Neuropathy

 

Figure 16 is a pharmacotherapy algorithm that we propose for the management of painful neuropathy in diabetes. This presumes that the cause of the pain has been attributed to DPN and that all causes masquerading as DPN have been excluded. The identification of neuropathic pain as being focal or diffuse dictates the initial course of action. Focal neuropathic pain is best treated with splinting, steroid injections, and surgery to release entrapment. Diffuse neuropathies are treated with medical therapy and in a majority of cases, need combination therapy.  Essential to the DPN evaluation is the identification of the patient’s comorbidities, potential adverse events, and drug interactions. When single agents fail, combinations of drugs with different mechanisms of action should be considered. Comorbidities that accompany pain include depression, anxiety, and sleep disturbances, all of which must be addressed for successful management of pain. Treatment of peripheral neuropathic pain conditions can benefit from further understanding of the impact of pain response and QOL, including activities of daily living (ADLs) and sleep. Patients often benefit from participation in pain management groups and psychological intervention to develop/gain better coping strategies and deal with harmful/disruptive pain-related behaviors. There is currently minimal evidence for the use of combination treatment for painful DPN – hence, most guidelines recommend switching to an alternative agent. There are also few head-to-head comparator trials of commonly used agent evaluating efficacy and safety between drugs. We await the outcome of the much-anticipated OPTION-DM study – head-to-head multicenter, RCT will inform clinicians of the most cost effective monotherapy (amitriptyline, pregabalin and duloxetine) followed by combination therapy for painful DPN (314).

Figure 16. Algorithm for the Management of Symptomatic Diabetic Neuropathy. Non-pharmacological, topical or physical therapies can be useful at any time. SNRIs, serotonin and norepinephrine reuptake inhibitors; TCA, tricyclic antidepressants.

AUTONOMIC NEUROPATHY

 

Introduction

 

The autonomic nervous system (ANS) supplies all organs in the body and consists of an afferent and an efferent system, with long efferents in the vagus (cholinergic) and short postganglionic unmyelinated fibers in the sympathetic system (adrenergic). A third component is the neuropeptidergic system with its neurotransmitters substance P (SP), vasoactive intestinal polypeptide (VIP), and calcitonin gene related peptide (CGRP) amongst others. Diabetic autonomic neuropathy (DAN) is a serious and common complication of diabetes but remains among the least recognized and understood. Diabetic autonomic neuropathy (DAN) can cause dysfunction of every part of the body, and has a significant negative impact on survival and quality of life (315). The organ systems that most often exhibit prominent clinical autonomic signs and symptoms in diabetes include the pupils, sweat glands, genitourinary system, gastrointestinal tract, adrenal medullary system, and the cardiovascular system (Table 6). Clinical symptoms generally do not appear until long after the onset of diabetes. However, subclinical autonomic dysfunction can occur within a year of diagnosis in type 2 diabetes patients and within two years in type 1 diabetes patients (316).

 

 

Table 6. Clinical Manifestations of Autonomic Neuropathy

Cardiovascular

Central:

Tachycardia/ Bradycardia

Systolic and diastolic dysfunction

Decreased exercise tolerance

Orthostasis,

Orthostatic tachycardia and bradycardia syndrome

Sleep apnea

Anxiety/ depression

Cardiac denervation syndrome

Paradoxic supine or nocturnal hypertension

Intraoperative and perioperative cardiovascular instability

Peripheral:

Decreased thermoregulation

Decreased sweating

Altered blood flow

Impaired vasomotion

Edema

Gastrointestinal

Esophageal dysmotility

Gastroparesis diabeticorum

Diarrhea

Constipation

Fecal incontinence

Genitourinary

Erectile dysfunction

Retrograde ejaculation

Neurogenic bladder and cystopathy

Female sexual dysfunction (e.g., loss of vaginal lubrication)

Sudomotor

Anhidrosis

Hyperhidrosis

Heat intolerance

Gustatory sweating

Dry skin

Metabolic

Hypoglycemia unawareness

Hypoglycemia unresponsiveness

Pupillary

Pupillomotor function impairment (e.g., decreased diameter of dark-adapted pupil)

Pseudo Argyll-Robertson pupil

 

 

Microvascular flow is under the control of the ANS and is regulated by both the central and peripheral components of the ANS. Defective blood flow in the small capillary circulation is found with decreased responsiveness to mental arithmetic, cold pressor, hand grip, and heating (317). The defect is associated with a reduction in the amplitude of vasomotion (318) and resembles premature aging (277). There are differences in the glabrous and hairy skin (319) and is correctable with antioxidants (320). The clinical counterpart is a dry cold skin, loss of sweating, and development of fissures and cracks that are portals of entry for organisms leading to infectious ulcers and gangrenes. Silent myocardial infarction, respiratory failure, amputations, and sudden death are hazards for diabetes patients with cardiac autonomic neuropathy (321). Therefore, it is vitally important to make this diagnosis early so that appropriate intervention can be instituted (322).

 

Disturbances in the autonomic nervous system may be functional, e.g., gastroparesis with hyperglycemia and ketoacidosis, or organic wherein nerve fibers are actually lost. This creates inordinate difficulties in diagnosing, treating, and prognosticating as well as establishing true prevalence rates. Tests of autonomic function generally stimulate entire reflex pathways. Furthermore, autonomic control for each organ system is usually divided between opposing sympathetic and parasympathetic innervations, so that heart rate acceleration, for example, may reflect either decreased parasympathetic or increased sympathetic nervous system stimulation. Since many conditions affect the autonomic nervous system and autonomic neuropathy (AN) is not unique to diabetes, the diagnosis of DAN rests with establishing the diagnosis and excluding other causes (Table 7 and 8). The best studied diagnostic methods, for which there are large databases and evidence to support their use in clinical practice, relate to the evaluation of cardiovascular reflexes (Figure 17). In addition, the evaluation of orthostasis is fairly straightforward and is readily done in clinical practice (Figure 18), as is the establishment of the cause of gastrointestinal symptoms (Figure 19) and erectile dysfunction. The combination of cardiovascular autonomic tests with sudomotor function tests may allow a more accurate diagnosis of diabetic autonomic neuropathy (323). Tables 9 and 10 below present the diagnostic tests that would be applicable to the diagnosis of cardiovascular autonomic neuropathy. These tests can be used as a surrogate for the diagnosis of AN of any system since it is generally rare to find involvement (although it does occur) of any other division of the ANS in the absence of cardiovascular autonomic dysfunction. For example, if one entertains the possibility that the patient has erectile dysfunction due to AN, then prior to embarking upon a sophisticated and expensive evaluation of erectile status, a measure of heart rate and its variability in response to deep breathing would - if normal - exclude the likelihood that the erectile dysfunction is a consequence of disease of the autonomic nervous system. The cause thereof would have to be sought elsewhere. Similarly, it is extremely unusual to find gastroparesis secondary to AN in a patient with normal cardiovascular autonomic reflexes.

 

Table 7. Differential Diagnosis of Diabetic Autonomic Neuropathy

Clinical Manifestations

Differential Diagnosis

Cardiovascular

Resting tachycardia, Exercise intolerance

Orthostatic tachycardia and bradycardia syndromes

Cardiac denervation, painless myocardial infarction

Orthostatic hypotension

Intraoperative and perioperative cardiovascular instability

Cardiovascular disorders

Idiopathic orthostatic hypotension, multiple system atrophy with Parkinsonism, orthostatic tachycardia, hyperadrenergic hypotension

Shy-Drager syndrome

Panhypopituitarism

Pheochromocytoma

Hypovolemia

Congestive heart disease

Carcinoid syndrome

Gastrointestinal

Esophageal dysfunction

Gastroparesis diabeticorum

Diarrhea

Constipation

Fecal incontinence

Gastrointestinal disorders

Obstruction

Bezoars

Secretory diarrhea (endocrine tumors)

Biliary disease

Psychogenic vomiting

Medications

Genitourinary

Erectile dysfunction

Retrograde ejaculation

Cystopathy

Neurogenic bladder

Genitourinary disorders

Genital and pelvic surgery

Atherosclerotic vascular disease

Medications

Alcohol abuse

Neurovascular

Heat intolerance

Gustatory sweating

Dry skin

Impaired skin blood flow

Other causes of neurovascular dysfunction

Chaga's disease

Amyloidosis

Arsenic

Metabolic

Hypoglycemia unawareness

Hypoglycemia unresponsiveness

Hypoglycemia associated autonomic failure

Metabolic disorders

Other cause of hypoglycemia, intensive glycemic control and drugs that mask hypoglycemia

Pupillary

Decreased diameter of dark- adapted pupil

Argyll-Robertson type pupil

Pupillary disorders

Syphilis

 

Table 8. Diagnosis and Management of Autonomic Nerve Dysfunction

Symptoms

Assessment Modalities

Management

Resting tachycardia, exercise intolerance, early fatigue and weakness with exercise

HRV, respiratory HRV, MUGA thallium scan, 123I MIBG scan

Graded supervised exercise, beta blockers, ACE-inhibitors

Postural hypotension, dizziness, lightheadedness, weakness, fatigue, syncope, tachycardia/bradycardia

HRV, blood pressure measurement lying and standing

Mechanical measures, clonidine, midodrine, octreotide, erythropoietin, pyridostigmine

Hyperhidrosis

Sympathetic/parasympathetic balance

Clonidine, amitryptylline, trihexyphenidyl, propantheline, or scopolamine ,botox, Glycopyrrolate

 

Table 9.  Diagnostic Tests of Cardiovascular Autonomic Neuropathy

TEST

METHOD/ PARAMETERS

Resting heart rate Beat-to-beat heart rate Variation*

>100 beats/min is abnormal. With the patient at rest and supine (no overnight coffee or hypoglycemic episodes), breathing 6 breaths/min, heart rate monitored by EKG or ANSCORE device, a difference in heart rate of >15 beats/min is normal and <10 beats/min is abnormal, R-R inspiration/R-R expiration >1.17. All indices of HRV are age-dependent**.

Heart rate response to Standing*

During continuous EKG monitoring, the R-R interval is measured at beats 15 and 30 after standing. Normally, a tachycardia is followed by reflex bradycardia. The 30:15 ratio is normally >1.03.

Heart rate response to Valsalva maneuver*

The subject forcibly exhales into the mouthpiece of a manometer to 40 mmHg for 15 s during EKG monitoring. Healthy subjects develop tachycardia and peripheral vasoconstriction during strain and an overshoot bradycardia and rise in blood pressure with release. The ratio of longest R-R shortest R-R should be >1.2.

Spectral analysis of heart rate variation, very low frequency power (VLFP 0.003-0.04) and high frequency power (HFP 0.15-0.40 Hz)

Series of sequential R-R intervals into its various frequent components. It defines two fixed spectral regions for the low-frequency and high-frequency measure.

Systolic blood pressure response to standing 

Systolic blood pressure is measured in the supine subject. The patient stands and the systolic blood pressure is measured after 2 min. Normal response is a fall of <10 mmHg, borderline is a fall of 10-29 mmHg, and abnormal is a fall of >30 mmHg with symptoms.

Diastolic blood pressure response to isometric exercise

The subject squeezes a handgrip dynamometer to establish a maximum. Grip is then squeezed at 30% maximum for 5 min. The normal response for diastolic blood pressure is a rise of >16 mmHg in the other arm.

EKG QT/QTc intervals Spectral analysis with respiratory frequency

The QTc (corrected QT interval on EKG) should be <440 ms. VLF peak (sympathetic dysfunction) LF peak (sympathetic dysfunction) HF peak (parasympathetic dysfunction) LH/HF ratio (sympathetic imbalance)

Neurovascular flow

Using noninvasive laser Doppler measures of peripheral sympathetic responses to nociception.

* These can now be performed quickly (<15 min) in the practitioners' office, with a central reference laboratory providing quality control and normative values. LF, VLF, HF =low, very low and high frequency peaks on spectral analysis. These are now readily available in most cardiologist's practice.** Lowest normal value of E/I ratio: Age 20-24:1.17, 25-29:1.15, 30-34:1.13, 35-30:1.12, 40-44:1.10, 45-49:1.08, 50-54:1.07, 55-59:1.06, 60-64:1.04, 65-69:1.03, 70-75:1.02 .

 

Table 10. Diagnostic Assessment of Cardiovascular Autonomic Function

Parasympathetic

Sympathetic

Resting heart rate

Beat to beat variation with deep breathing (E:I ratio)

30:15 heart rate ratio with standing

Valsalva ratio

Spectral analysis of heart rate variation , high frequency power (HFP 0.15-0.40 Hz)

Spectral Analysis of HRV respiratory frequency

Resting heart rate

Spectral analysis of heart rate variation, very low frequency power (VLFP 0.003-0.04)

Orthostasis BP

Hand grip BP

Cold pressor response

Sympathetic skin galvanic response (cholinergic)

Sudorimetry (cholinergic)

Cutaneous blood flow (peptidergic)

Figure 17. This is a sample power spectrum of the HRV signal from a subject breathing at an average rate of 7.5 breaths per minute (Fundamental Respiratory Frequency, FRF = 0.125 Hz). The method using HRV alone defines two fixed spectral regions for the low-frequency (LF) and high-frequency (HF) measure (dark gray and light gray, respectively). It is clear that the high-frequency (light gray) region includes very little area under the HRV spectral curve, suggesting very little parasympathetic activity. The great majority of the HRV spectral activity is under the low-frequency (dark gray) region suggesting primarily sympathetic activity. These representations are incorrect because the slow-breathing subject should have a large parasympathetic component reflective of the vagal activity. This parasympathetic component is represented correctly by the method using both HRV and respiratory activity which defines the red and blue regions of the spectrum in the graph. The blue region defined by the FRF represents purely parasympathetic activity whereas the remainder of the lower frequency regions (red region) represents purely sympathetic activity.

Figure 18. Evaluation of postural dizziness in patients with diabetes

Figure 19. Evaluation of a patient with suspected gastroparesis

The role of over-activation of the autonomic nervous system is illustrated in Figure 20 (324).

Figure 20. Role of over-activation of autonomic nervous system

There are few data on the longitudinal trends in small fiber dysfunction. Much remains to be learned of the natural history of diabetic autonomic neuropathy. Karamitsos et al (325) reported that the progression of diabetic autonomic neuropathy is significant during the 2 years subsequent to its discovery.

 

The mortality for diabetic autonomic neuropathy has been estimated to be 44% within 2.5 years of diagnosing symptomatic autonomic neuropathy (29).  In a meta-analysis, the Mantel-Haenszel estimates for the pooled prevalence rate risk for silent myocardial ischemia was 1.96, with 95% confidence interval of 1.53 to 2.51 (p<0.001; n = 1,468 total subjects). Thus, a consistent association between CAN and the presence of silent myocardial ischemia was shown (284) (Figure 21).

Figure 21. Relative risks and 95% CIs for studies of cardiovascular neuropathy (CAN) and mortality. Pooled relative risk for 10 studies with CAN define by two or more measures: 3.45 (95% CI 2.66–4.47). Pooled relative risk for 4 studies with CAN defined by a single measure: 1.20 (1.02–1.41). REF: Maser, R. E., Mitchell, B. D., Vinik, A. I., and Freeman, R. Diabetes Care. 2003;26(6):1895-1901.

Prevention and Reversibility of Autonomic Neuropathy

 

It has now become clear that strict glycemic control (37) and a stepwise progressive management of hyperglycemia, lipids, and blood pressure as well as the use of antioxidants (326) and ACE inhibitors (327) reduce the odds ratio for autonomic neuropathy to 0.32 (328). It has also been shown that early mortality is a function of loss of beat-to-beat variability with MI. This can be reduced by 33% with acute administration of insulin (329). Kendall et al (330) reported that successful pancreas transplantation improves epinephrine response and normalizes hypoglycemia symptom recognition in patients with long standing diabetes and established autonomic neuropathy. Burger et al (331) showed that a reversible metabolic component of CAN exists in patients with early CAN.

 

Management of Autonomic Neuropathy

 

POSTURAL HYPOTENSION

 

The syndrome of postural hypotension is posture-related dizziness and syncope. Patients who have Type 2 diabetes mellitus and orthostatic hypotension are hypovolemic and have sympathoadrenal insufficiency; both factors contribute to the pathogenesis of orthostatic hypotension (332). Postural hypotension in the patient with diabetic autonomic neuropathy can present a difficult management problem. Elevating the blood pressure in the standing position must be balanced against preventing hypertension in the supine position.

 

Supportive Garments: Whenever possible, attempts should be made to increase venous return from the periphery using total body stockings. But leg compression alone is less effective, presumably reflecting the large capacity of the abdomen relative to the legs (333). Patients should be instructed to put them on while lying down and to not remove them until returning to the supine position.

 

Drug Therapy: Some patients with postural hypotension may benefit from treatment with 9-flurohydrocortisone. Unfortunately, symptoms do not improve until edema occurs, and there is a significant risk of developing congestive heart failure and hypertension. If fluorohydrocortisone does not work satisfactorily, various adrenergic agonists and antagonists may be used (Table 11). Enhancement of ganglionic transmission via the use of pyridostigmine (inhibitor of acetylcholinesterase) improved symptoms and orthostatic hypotension with only modest effects in supine BP for patients with POTS. Similarly, the use of b-adrenergic blockers may benefit the tachycardia, and anticholinergics, the orthostatic bradycardia. Pyridostigmine has also been shown to improve HRV in healthy young adults.  If the adrenergic receptor status is known, then therapy can be guided to the appropriate agent.  Metoclopramide may be helpful in patients with dopamine excess or increased sensitivity to dopaminergic stimulation. Patients with α2-adrenergic receptor excess may respond to the α2-antagonist yohimbine. Those few patients in whom ß-receptors are increased may be helped with propranolol. α2-adrenergic receptor deficiency can be treated with the α2-agonist clonidine, which in this setting may paradoxically increase blood pressure. One should start with small doses and gradually increase the dose. If the preceding measures fail, midodrine, an α1-adrenergic agonist, or dihydroergotamine in combination with caffeine may help. A particularly refractory form of postural hypotension occurs in some patients post-prandially and may respond to therapy with octreotide given subcutaneously in the mornings.

 

 

Table 11. Pharmacologic Treatment of Autonomic Neuropathy

Clinical status

Drug

Dosage

Side effects

Orthostatic hypotension

 

9α flouro hydrocortisone, mineralocorticoid

0.5-2 mg/day

Congestive heart failure, hypertension

 

Clonidine, α2 adrenergic agonist

0,1-0,5 mg, at bedtime

Orthostatic Hypotension, sedation, dry mouth, constipation, dizziness, bradycardia.

 

Octreotide, somatostatin analogue

0.1-0.5 mg/kg/day

Injection site pain, diarrhea

Orthostatic tachycardia and bradycardia syndrome

 

Clonidine, α2 adrenergic agonist

0.1-0.5 mg, at bedtime

Orthostatic Hypotension, sedation, dry mouth, constipation, dizziness, bradycardia.

 

Octreotide, somatostatin analogue

0.1-0.5 μg/kg/day

Injection site pain, diarrhea

Gastroparesis diabeticorum

 

Domperidone, D2-receptor antagonist

10-20 mg, 30-60 min before meal and bedtime

Galactorrhea

 

Erythromycin, motilin receptor agonist

250 mg, 30 minutes before meals

Abdominal cramps, nausea, diarrhea, rash

 

Levosulphide, D2-receptor antagonist

25 mg, 3 times/day

Galactorrhea

Diabetic diarrhea

 

Metronidazole, broad spectrum antibiotics

250 mg, 3 times/day, minimum 3 weeks

Anorexia, rash, GI upset, urine discoloration, dizziness, disulfiram like reaction.

 

Clonidine, α2 adrenergic agonist

0.1 mg, 2-3 times/day

Orthostatic Hypotension, sedation, dry mouth, constipation, dizziness, bradycardia.

 

Cholestyramine, bile acid sequestrant

4 g, 1-6 times/day

Constipation

 

Loperamide, opiate-receptor agonist

2 mg, four times/day

Toxic megacolon

 

Octreotide, somatostatin analogue

50 μg, 3 times/day

Aggravate nutrient malabsorption (at higher doses)

Cystopathy

 

Bethanechol, acetylcholine receptor agonist

10 mg, 4 times/day

Blurred vision, abdominal cramps, diarrhea, salivation, and hypotension.

 

Doxazosin, α1 adrenergic antagonist

1-2 mg, 2-3 times/day

Hypotension, headache, palpitation

Exercise Intolerance

 

Graded supervised exercise

20 minutes, 3 times/week

Foot injury, angina.

Hyperhidrosis

 

Clonidine, α2 adrenergic agonist

0.1-0.5 mg, at bedtime and divided doses above 0.2 mg

Orthostatic Hypotension, sedation, dry mouth, constipation, dizziness, bradycardia.

 

Amitryptiline, Norepinephrine & serotonin reuptake inhibitor

150 mg/ day

Tachycardia, palpitation

 

Propantheline, Anti-muscarinic.

15 mg/ day PO

Dry mouth, blurred vision

 

Trihexyphenidyl,

2-5 mg PO

Dry mouth, blurred vision, constipation, tachycardia, photosensitivity, arrhythmias.

 

Botox,

 

 

 

Scopolamine, anti-cholinergic

1.5 mg patch/ 3 days; 0.4 to 0.8mg PO

Dry mouth, blurred vision, constipation, drowsiness, and tachycardia.

 

Glycopyrrolate, anti-cholinergic

1-2 mg, 2-3 times daily.

Constipation, tachycardia, dry mouth.

Erectile dysfunction

 

 

 

 

Sildenafil (Viagra), GMP type-5 phosphodiesterase inhibitor

50 mg before sexual activity, only once per day

Hypotension and fatal cardiac event (with nitrate-containing drugs), headache, flushing, nasal congestion, dyspepsia, musculoskeletal pain, blurred vision

 

Tadalafil (Cialis), GMP type-5 phosphodiesterase inhibitor

10 mg PO before sexual activity only once per day.

Headache, flushing, dyspepsia, rhinitis, myalgia, back pain.

 

Verdenafil (Levitra), GMP type-5 phosphodiesterase inhibitor

10 mg PO, 60 minutes before sexual activity.

Hypotension, headache, dyspepsia, priapism.

 

 

SLEEP APNEA

 

During sleep, increased sympathetic drive is a result of repetitive episodes of hypoxia, hypercapnia, and obstructive apnea (OSA) acting through chemoreceptor reflexes. Increased sympathetic drive has been implicated in increased blood pressure variability with repetitive sympathetic activation and blood pressure surges impairing baroreflex and cardiovascular reflex functions (284). A direct relationship between the severity of OSA and the increase in blood pressure has been noted. Furthermore, the use of continuous positive airway pressure (CPAP) for the treatment of OSA has been shown to lower blood pressure and improve cardiovascular autonomic nerve fiber function for individuals with OSA. Withdrawal of CPAP for even a short period (i.e., 1 week) has been shown to result in a marked increase in sympathetic activity (284).

 

GASTROPATHY

 

Gastrointestinal motor disorders are frequent and widespread in patients with type 2 diabetes, regardless of symptoms (334) and there is a poor correlation between symptoms and objective evidence of a functional or organic defect. The first step in management of diabetic gastroparesis consists of multiple, small feedings; decreased fat intake as it tends to delay gastric emptying; maintenance of glycemic control (335,336); and a low-fiber diet to avoid bezoar formation. Metoclopramide may be used. Domperidone (337,338) has been shown to be effective in some patients, although probably no more so than metoclopramide. Erythromycin given as either a liquid or suppository also may be helpful. Erythromycin acts on the motilin receptor, "the sweeper of the gut," and shortens gastric emptying time (339). Several novel drugs, including the ghrelin (orexigenic hormone) and ghrelin receptor agonists, motilin agonist (mitemcinal), 5-HT4-receptor agonists and the muscarinic antagonist are being investigated for their prokinetic effects (340,341).  If medications fail and severe gastroparesis persists, jejunostomy placement into normally functioning bowel may be needed. Different treatment modalities for gastroparesis include dietary modifications, prokinetic and antiemetic medications, measures to control pain and address psychological issues, and endoscopic or surgical options in selected instances (342).

 

For additional information see the Endotext chapter entitled “Gastrointestinal Disorders in Diabetes”.

 

ENTEROPATHY     

 

Enteropathy involving the small bowel and colon can produce both chronic constipation and explosive diabetic diarrhea, making treatment of this complication difficult.

 

Antibiotics: Stasis of bowel contents with bacterial overgrowth may contribute to the diarrhea. Treatment with broad-spectrum antibiotics is the mainstay of therapy, including tetracycline or trimethoprim and sulfamethoxazole. Metronidazole appears to be the most effective and should be continued for at least 3 weeks.

 

Cholestyramine: Retention of bile may occur and can be highly irritating to the gut. Chelation of bile salts with cholestyramine 4g tid mixed with fluid may offer relief of symptoms.

 

Diphenoxylate plus atropine: Diphenoxylate plus atropine may help to control the diarrhea; however, toxic megacolon can occur, and extreme care should be used.

 

Diet: Patients with poor digestion may benefit from a gluten-free diet, while constipation may respond to a high-soluble-fiber diet supplemented with daily hydrophilic colloid. Beware of certain fibers in the neuropathic patient that can lead to bezoar formation because of bowel stasis in gastroparetic or constipated patients.

 

For additional information see the Endotext chapter entitled “Gastrointestinal Disorders in Diabetes”.

 

SEXUAL DYSFUNCTION

 

Erectile dysfunction (ED) occurs in 50-75% of men with diabetes, and it tends to occur at an earlier age than in the general population. The incidence of ED in men with diabetes aged 20-29 years is 9% and increases to 95% by age 70. It may be the presenting symptom of diabetes. More than 50% notice the onset of ED within 10 years of the diagnosis, but it may precede the other complications of diabetes. The etiology of ED in diabetes is multifactorial. Neuropathy, vascular disease, diabetes control, nutrition, endocrine disorders, psychogenic factors as well as drugs used in the treatment of diabetes and its complications play a role (343,344). The diagnosis of the cause of ED is made by a logical stepwise progression in all instances. An approach to therapy has been presented to which the reader is referred; Figure 22 below shows a flow chart modified from Vinik et. al., 1998 (302).

Figure 22. Evaluation of patients with diabetes with erectile dysfunction

A thorough work-up for impotence will include: medical and sexual history; physical and psychological evaluations; blood tests for diabetes and levels of testosterone, prolactin, and thyroid hormones; tests for nocturnal erections; tests to assess penile, pelvic, and spinal nerve function; and a test to assess penile blood supply and blood pressure. The flow chart provided is intended as a guide to assist in defining the problem. The healthcare provider should initiate questions that will help distinguish the various forms of organic erectile dysfunction from those that are psychogenic in origin. Physical examination must include an evaluation of the autonomic nervous system, vascular supply, and the hypothalamic-pituitary-gonadal axis.

 

Autonomic neuropathy causing ED is almost always accompanied by loss of ankle jerks and absence or reduction of vibration sense over the large toes. More direct evidence of impairment of penile autonomic function can be obtained by (1) demonstrating normal perianal sensation, (2) assessing the tone of the anal sphincter during a rectal exam, and (3) ascertaining the presence of an anal wink when the area of the skin adjacent to the anus is stroked or contraction of the anus when the glans penis is squeezed, i.e., the bulbo-cavernosus reflex. These measurements are easily and quickly done at the bedside and reflect the integrity of sacral parasympathetic divisions.

 

Vascular disease is usually manifested by buttock claudication but may be due to stenosis of the internal pudendal artery. A penile/brachial index of <0.7 indicates diminished blood supply. A venous leak manifests as unresponsiveness to vasodilators and needs to be evaluated by penile Doppler sonography.

 

In order to distinguish psychogenic from organic erectile dysfunction, nocturnal penile tumescence (NPT) measurement can be done. Normal NPT defines psychogenic ED, and a negative response to vasodilators implies vascular insufficiency. Application of NPT is not so simple. It is much like having a sphygmomanometer cuff inflate over the penis many times during the night while one is trying to have a normal night's sleep and the REM sleep associated with erections. The individual may have to take home the device and become familiar with it over several nights before one has a reliable estimate of the failure of NPT.

 

Treatment of Erectile Dysfunction

 

A number of treatment modalities are available and each treatment has positive and negative effects; therefore, patients must be made aware of both aspects before a therapeutic decision is made. Before considering any form of treatment, every effort should be made to have the patient withdraw from alcohol and eliminate smoking. If possible, drugs that are known to cause erectile dysfunction should be removed. Additionally, metabolic control should be optimized.

 

Relaxation of the corpus cavernous smooth muscle cells is caused by NO and cGMP, and the ability to have and maintain an erection depends on NO and cGMP. The peripherally acting oral phosphodiesterase type 5 (PDE5) inhibitors block the action of PDE5, and cGMP accumulates, enhancing blood flow to the corpora cavernosum with sexual stimulation. This class of agents consists of sildenafil, vardenafil, and tadalafil. They have been evaluated in patients with diabetes with similar levels of efficacy of about 70%. A 50 mg tablet of sildenafil taken orally is the usual starting dose, 60 minutes before sexual activity. Lower doses should be considered in patients with renal failure and hepatic dysfunction. The duration of the drug effect is 4 hours. Generally, patients with diabetes require the maximum dose of each agent, sildenafil 100 mg, tadalafil 20 mg, and vardenafil 20 mg. Before prescribing a PDE5 inhibitor, it is important to exclude ischemic heart disease. These are absolutely contraindicated in patients being treated with nitroglycerine or other nitrate-containing drugs. Severe hypotension and fatal cardiac events can occur (345). Side-effects include headache, flushing, dyspepsia, and muscle pain (346). Direct injection of prostacyclin into the corpus cavernosum will induce satisfactory erections in a significant number of men. Also, surgical implantation of a penile prosthesis may be appropriate. The less expensive type of prosthesis is a semirigid, permanently erect type that may be embarrassing and uncomfortable for some patients. The inflatable type is three times more expensive and subject to mechanical failure, but it avoids the embarrassment caused by other devices.

 

Female Sexual Dysfunction

 

Women with diabetes mellitus may experience decreased sexual desire and more pain on sexual intercourse, and they are at risk of decreased sexual arousal, with inadequate lubrication (347). Diagnosis of female sexual dysfunction using vaginal plethysmography to measure lubrication and vaginal flushing has not been well established.

 

For additional information on this topic see the Endotext chapter entitled “Sexual Dysfunction in Diabetes”.

 

CYSTOPATHY

 

In diabetic autonomic neuropathy, the motor function of the bladder is unimpaired, but afferent fiber damage results in diminished bladder sensation. The urinary bladder can be enlarged to more than three times its normal size. Patients are seen with bladders filled to their umbilicus, yet they feel no discomfort. Loss of bladder sensation occurs with diminished voiding frequency, and the patient is no longer able to void completely. Consequently, dribbling and overflow incontinence are common complaints. A post-void residual of greater than 150cc is diagnostic of cystopathy. Cystopathy may put the patients at risk for urinary infections.

 

Treatment of Cystopathy

 

Patients with cystopathy should be instructed to palpate their bladder and, if they are unable to initiate micturition when their bladders are full, use Crede's maneuver (massage or pressure on the lower portion of abdomen just above the pubic bone) to start the flow of urine. The principal aim of the treatment should be to improve bladder emptying and to reduce the risk of urinary tract infection. Parasympathomimetics such as bethanechol are sometimes helpful, although frequently they do not help to fully empty the bladder. Extended sphincter relaxation can be achieved with an alpha-1-blocker, such as doxazosin. Self-catheterization can be particularly useful in this setting, with the risk of infection generally being low.

 

SWEATING DYSFUNCTION

 

Hyperhidrosis of the upper body, often related to eating (gustatory sweating), and anhidrosis of the lower body, are a characteristic feature of autonomic neuropathy. Gustatory sweating accompanies the ingestion of certain foods, particularly spicy foods, and cheeses. There is a suggestion that application of glycopyrrolate (an antimuscarinic compound) might benefit diabetes patients with gustatory sweating (348). Low-dose oral glycopyrrolate in the range of 1 mg to 2 mg once daily can be tolerated without problematic adverse effects to alleviate the symptoms of diabetic gustatory sweating. Although more long-term data are needed, the use of glycopyrrolate for diabetic gustatory sweating may be a viable option (349). Symptomatic relief can be obtained by avoiding the specific inciting food. Loss of lower body sweating can cause dry, brittle skin that cracks easily, predisposing one to ulcer formation that can lead to loss of the limb. Special attention must be paid to foot care.

 

METABOLIC DYSFUNCTION

 

Hypoglycemia Unawareness

 

Blood glucose concentration is normally maintained during starvation or increased insulin action by an asymptomatic parasympathetic response with bradycardia and mild hypotension, followed by a sympathetic response with glucagon and epinephrine secretion for short-term glucose counter regulation, and growth hormone and cortisol secretion for long-term regulation. The release of catecholamine alerts the patient to take the required measures to prevent coma due to low blood glucose. The absence of warning signs of impending neuroglycopenia is known as "hypoglycemic unawareness". The failure of glucose counter regulation can be confirmed by the absence of glucagon and epinephrine responses to hypoglycemia induced by a standard, controlled dose of insulin (350).

 

In patients with type 1 diabetes mellitus, the glucagon response is impaired with diabetes duration of 1-5 years; after 14-31 years of diabetes, the glucagon response is almost undetectable. Absence of the glucagon response is not present in those with autonomic neuropathy. However, a syndrome of hypoglycemic autonomic failure occurs with intensification of diabetes control and repeated episodes of hypoglycemia. The exact mechanism is not understood, but it does represent a real barrier to physiologic glycemic control. In the absence of severe autonomic dysfunction, hypoglycemia unawareness is at least in part reversible.

 

Patients with hypoglycemia unawareness and unresponsiveness pose a significant management problem for the physician. Although autonomic neuropathy may improve with intensive therapy and normalization of blood glucose, there is a risk to the patient, who may become hypoglycemic without being aware of it and who cannot mount a counterregulatory response. It is our recommendation that if a pump is used, boluses of smaller than calculated amounts should be used and, if intensive conventional therapy is used, long-acting insulin with very small boluses should be given. In general, normal glucose and HbA1 levels should not be goals in these patients to avoid the possibility of hypoglycemia. The use of continuous glucose monitoring with hypoglycemic alarms can be very helpful in warning patients of hypoglycemia and in preventing severe hypoglycemic reactions.

 

Further complicating management of some patients with diabetes is the development of a functional autonomic insufficiency associated with intensive insulin treatment, which resembles autonomic neuropathy in all relevant aspects. In these instances, it is prudent to relax therapy, as for the patient with bona fide autonomic neuropathy. If hypoglycemia occurs in these patients at a certain glucose level, it will take a lower glucose level to trigger the same symptoms in the next 24-48 hours. Avoidance of hypoglycemia for a few days will result in recovery of the adrenergic response.

 

For additional information on this topic see the Endotext chapter entitled “Hypoglycemia During Therapy of Diabetes”.

 

DIABETIC NEUROPATHIES: PROSPECTS FOR THE FUTURE

 

Management of DN encompasses a wide variety of therapies. Treatment must be individualized in a manner that addresses the particular manifestation and underlying pathogenesis of each patient's unique clinical presentation, without subjecting the patient to untoward medication effects. An increased understanding of the pathogenesis of DN will lead to more effective approaches to diagnose and treat this condition.  Refinements and adoption of new approaches to measure quantitatively and diagnose DN early is crucial, so that appropriate therapies (risk factor modification or pathogenic) can be commenced before nerve damage is established. These tests must be validated and standardized to allow comparability between studies and a more meaningful interpretation of study results. Our ability to manage successfully the many different manifestations of DN depends ultimately on our success in uncovering the pathogenic processes underlying this disorder.

 

ACKNOWLEDGEMENTS

 

This chapter updates the original Endotext chapter on this topic written by Aaron Vinik, Carolina Casellini, and Marie-Laure Nevoret.

 

REFERENCES

 

  1. Saeedi, P., et al., Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas. Diabetes research and clinical practice, 2019. 157: p. 107843.
  2. Williams, R., et al., Global and regional estimates and projections of diabetes-related health expenditure: Results from the International Diabetes Federation Diabetes Atlas. Diabetes research and clinical practice, 2020. 162: p. 108072.
  3. Moxey, P., et al., Lower extremity amputations—a review of global variability in incidence. Diabetic Medicine, 2011. 28(10): p. 1144-1153.
  4. Paisey, R., et al., Diabetes‐related major lower limb amputation incidence is strongly related to diabetic foot service provision and improves with enhancement of services: peer review of the South‐West of England. Diabetic Medicine, 2018. 35(1): p. 53-62.
  5. Programme Budgeting. 2014[cited 2019 08/10/2019]; Available from: https://bit.ly/31WeHc5.
  6. Tesfaye, S., et al., Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes care, 2010. 33(10): p. 2285-2293.
  7. Ziegler, D., Painful diabetic neuropathy: treatment and future aspects. Diabetes/metabolism research and reviews, 2008. 24(S1): p. S52-S57.
  8. Boulton, A.J., et al., Diabetic somatic neuropathies. Diabetes care, 2004. 27(6): p. 1458-1486.
  9. Vinik, A.I. and D. Ziegler, Diabetic cardiovascular autonomic neuropathy. Circulation, 2007. 115(3): p. 387-397.
  10. Vinik, A.I., R.E. Maser, and D. Ziegler, Neuropathy: the crystal ball for cardiovascular disease? 2010, Am Diabetes Assoc. p. 1688-1690.
  11. Pop-Busui, R., et al., Effects of cardiac autonomic dysfunction on mortality risk in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Diabetes care, 2010. 33(7): p. 1578-1584.
  12. Abbott, C.A., et al., Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the UK. Diabetes care, 2011. 34(10): p. 2220-2224.
  13. Selvarajah, D., et al., The contributors of emotional distress in painful diabetic neuropathy. Diabetes and Vascular Disease Research, 2014. 11(4): p. 218-225.
  14. Giordano, J. and M.E. Schatman, A crisis in chronic pain care: an ethical analysis. Part three: toward an integrative, multi-disciplinary pain medicine built around the needs of the patient. Pain Physician, 2008. 11(6): p. 775-784.
  15. Thomas, P., Classification, differential diagnosis, and staging of diabetic peripheral neuropathy. Diabetes, 1997. 46(Supplement 2): p. S54-S57.
  16. Pop-Busui, R., et al., Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes care, 2017. 40(1): p. 136-154.
  17. Ziegler, D., et al., Somatic and autonomic nerve function during the first year after diagnosis of type 1 (insulin-dependent) diabetes. Diabetes Research (Edinburgh, Scotland), 1988. 7(3): p. 123-127.
  18. Papanas, N. and D. Ziegler, Prediabetic neuropathy: does it exist? Current diabetes reports, 2012. 12(4): p. 376-383.
  19. The effect of intensive diabetes therapy on the development and progression of neuropathy. The Diabetes Control and Complications Trial Research Group. Ann Intern Med, 1995. 122(8): p. 561-8.
  20. Ziegler, D., et al., The natural course of peripheral and autonomic neural function during the first two years after diagnosis of type 1 diabetes. Klinische Wochenschrift, 1988. 66(21): p. 1085-1092.
  21. Dyck, P., et al., The Rochester Diabetic Neuropathy Study: design, criteria for types of neuropathy, selection bias, and reproducibility of neuropathic tests. Neurology, 1991. 41(6): p. 799-799.
  22. Casellini, C.M., et al., A 6-month, randomized, double-masked, placebo-controlled study evaluating the effects of the protein kinase C-β inhibitor ruboxistaurin on skin microvascular blood flow and other measures of diabetic peripheral neuropathy. Diabetes care, 2007. 30(4): p. 896-902.
  23. Chalk, C., T.J. Benstead, and F. Moore, Aldose reductase inhibitors for the treatment of diabetic polyneuropathy. Cochrane database of systematic reviews, 2007(4).
  24. Ziegler, D., et al., Epidemiology of polyneuropathy in diabetes and prediabetes. Handbook of clinical neurology, 2014. 126: p. 3-22.
  25. Hicks, C.W. and E. Selvin, Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Current diabetes reports, 2019. 19(10): p. 1-8.
  26. Andersen, S.T., et al., Risk factors for incident diabetic polyneuropathy in a cohort with screen-detected type 2 diabetes followed for 13 years: ADDITION-Denmark. Diabetes care, 2018. 41(5): p. 1068-1075.
  27. Mizokami-Stout, K.R., et al., The contemporary prevalence of diabetic neuropathy in type 1 diabetes: findings from the T1D Exchange. Diabetes care, 2020. 43(4): p. 806-812.
  28. Partanen, J., et al., Natural history of peripheral neuropathy in patients with non-insulin-dependent diabetes mellitus. New England Journal of Medicine, 1995. 333(2): p. 89-94.
  29. Martin, C.L., et al., Neuropathy and related findings in the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes care, 2014. 37(1): p. 31-38.
  30. Dyck, P.J., et al., The prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and nephropathy in a population‐based cohort: the Rochester Diabetic Neuropathy Study. Neurology, 1993. 43(4): p. 817-817.
  31. Armstrong, D.G., L.A. Lavery, and L.B. Harkless, Validation of a diabetic wound classification system: the contribution of depth, infection, and ischemia to risk of amputation. Diabetes care, 1998. 21(5): p. 855-859.
  32. East, M. and N. Africa, IDF diabetes atlas. diabetes, 2017. 20: p. 79.
  33. Hoffstad, O., et al., Diabetes, lower-extremity amputation, and death. Diabetes Care, 2015. 38(10): p. 1852-1857.
  34. Diabetes, U., Twenty devastating amputations ever day. 2018, Retrieved on 12th March.
  35. Cavanagh, P., et al., Cost of treating diabetic foot ulcers in five different countries. Diabetes/metabolism research and reviews, 2012. 28: p. 107-111.
  36. Brownlee, M. and I.B. Hirsch, Glycemic variability: a hemoglobin A1c–independent risk factor for diabetic complications. Jama, 2006. 295(14): p. 1707-1708.
  37. DCCT Research Group, B.N.D., Bethesda, MD 20892., Effect of intensive diabetes treatment on nerve conduction in the Diabetes Control and Complications Trial. Annals of Neurology, 1995. 38(6): p. 869-880.
  38. Callaghan, B.C., et al., Enhanced glucose control for preventing and treating diabetic neuropathy. Cochrane database of systematic reviews, 2012(6).
  39. Ismail-Beigi, F., et al., Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. The Lancet, 2010. 376(9739): p. 419-430.
  40. Calles-Escandón, J., et al., Effect of intensive compared with standard glycemia treatment strategies on mortality by baseline subgroup characteristics: the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Diabetes care, 2010. 33(4): p. 721-727.
  41. Tesfaye, S., et al., Vascular risk factors and diabetic neuropathy. New England Journal of Medicine, 2005. 352(4): p. 341-350.
  42. Vincent, A.M., et al., Diabetic neuropathy: cellular mechanisms as therapeutic targets. Nature Reviews Neurology, 2011. 7(10): p. 573-583.
  43. Boulton, A.J., et al., Whither pathogenetic treatments for diabetic polyneuropathy? Diabetes/Metabolism Research and Reviews, 2013. 29(5): p. 327-333.
  44. Vinik, A., et al., Diabetic neuropathies: clinical manifestations and current treatment options. Nature clinical practice Endocrinology & metabolism, 2006. 2(5): p. 269-281.
  45. Feldman, E.L., et al., Diabetic neuropathy. Nature reviews Disease primers, 2019. 5(1): p. 1-18.
  46. Vinik, A., et al., Focal entrapment neuropathies in diabetes. Diabetes care, 2004. 27(7): p. 1783-1788.
  47. Wilbourn, A., Diabetic entrapment and compression neuropathies. Diabetic neuropathy, 1999: p. 481-508.
  48. Watanabe, K., et al., Characteristics of cranial nerve palsies in diabetic patients. Diabetes research and clinical practice, 1990. 10(1): p. 19-27.
  49. Perkins, B.A., D. Olaleye, and V. Bril, Carpal tunnel syndrome in patients with diabetic polyneuropathy. Diabetes Care, 2002. 25(3): p. 565-569.
  50. Dawson, D.M., Entrapment neuropathies of the upper extremities. New England Journal of Medicine, 1993. 329(27): p. 2013-2018.
  51. Rota, E. and N. Morelli, Entrapment neuropathies in diabetes mellitus. World journal of diabetes, 2016. 7(17): p. 342-353.
  52. Llewelyn, J., P. Thomas, and R. King, Epineurial microvasculitis in proximal diabetic neuropathy. Journal of neurology, 1998. 245(3): p. 159-165.
  53. Dyck, P.J.B. and A.J. Windebank, Diabetic and nondiabetic lumbosacral radiculoplexus neuropathies: new insights into pathophysiology and treatment. Muscle & nerve, 2002. 25(4): p. 477-491.
  54. Roberts, A., J. James, and K. Dhatariya, Management of hyperglycaemia and steroid (glucocorticoid) therapy: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabet Med, 2018. 35(8): p. 1011-1017.
  55. Vinik, A.I., G.L. Pittenger, and L. Hopkins, Autoimmune mechanisms in the pathogenesis of diabetic neuropathy. 1998.
  56. Steck, A.J. and L. Kappos, Gangliosides and autoimmune neuropathies: classification and clinical aspects of autoimmune neuropathies. Journal of neurology, neurosurgery, and psychiatry, 1994. 57(Suppl): p. 26.
  57. Said, G., et al., Nerve biopsy findings in different patterns of proximal diabetic neuropathy. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society, 1994. 35(5): p. 559-569.
  58. Sander, H.W. and S. Chokroverty. Diabetic amyotrophy: current concepts. in Seminars in neurology. 1996. © 1996 by Thieme Medical Publishers, Inc.
  59. DA Kendrel, D.C., LC Hopkins, Successful treatment of neuropathies in patients with diabetes mellitus. Arch Neurol, 1995. 52: p. 1053-1061.
  60. Britland, S.T., et al., Acute and remitting painful diabetic polyneuropathy: a comparison of peripheral nerve fibre pathology. Pain, 1992. 48(3): p. 361-370.
  61. Sharma, K.R., et al., Demyelinating neuropathy in diabetes mellitus. Archives of Neurology, 2002. 59(5): p. 758-765.
  62. Krendel, D.A., A. Zacharias, and D.S. Younger, Autoimmune diabetic neuropathy. Neurologic clinics, 1997. 15(4): p. 959-971.
  63. Milicevic, Z., et al., Anti-ganglioside GM1 antibody and distal symmetric" diabetic polyneuropathy" with dominant motor features. Diabetologia, 1997. 40(11): p. 1364-1365.
  64. Ayyar, D.R. and K.R. Sharma, Chronic inflammatory demyelinating polyradiculoneuropathy in diabetes mellitus. Current diabetes reports, 2004. 4(6): p. 409-412.
  65. BARADA, A., et al., Proximal diabetic neuropathy-response to immunotherapy. Diabetes, 1999. 48(5): p. SA148-SA148.
  66. Archer, A., et al., The natural history of acute painful neuropathy in diabetes mellitus. Journal of Neurology, Neurosurgery & Psychiatry, 1983. 46(6): p. 491-499.
  67. NICE, Neuropathic pain in adults: pharmacological management in non-specialist settings. 2013.
  68. Tesfaye, S., et al., Arterio-venous shunting and proliferating new vessels in acute painful neuropathy of rapid glycaemic control (insulin neuritis). diabetologia, 1996. 39(3): p. 329-335.
  69. Gibbons, C.H. and R. Freeman, Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain, 2015. 138(1): p. 43-52.
  70. Sinnreich, M., B.V. Taylor, and P.J.B. Dyck, Diabetic neuropathies: classification, clinical features, and pathophysiological basis. The neurologist, 2005. 11(2): p. 63-79.
  71. Oyibo, S., et al., The relationship between blood glucose excursions and painful diabetic peripheral neuropathy: a pilot study. Diabetic Medicine, 2002. 19(10): p. 870-873.
  72. Ward, J., The diabetic leg. Diabetologia, 1982. 22(3): p. 141-147.
  73. Katoulis, E.C., et al., Gait abnormalities in diabetic neuropathy. Diabetes care, 1997. 20(12): p. 1904-1907.
  74. Cavanagh, P.R., J.S. Ulbrecht, and G.M. Caputo, New developments in the biomechanics of the diabetic foot. Diabetes/Metabolism Research and Reviews, 2000. 16(S1): p. S6-S10.
  75. Reiber, G.E., et al., Causal pathways for incident lower-extremity ulcers in patients with diabetes from two settings. Diabetes care, 1999. 22(1): p. 157-162.
  76. AN, P., et al., A comparison of the Neuropen against standard quantitative sensory‐threshold measures for assessing peripheral nerve function. Diabetic Medicine, 2002. 19(5): p. 400-405.
  77. Ziegler, D., et al., Validation of a novel screening device (NeuroQuick) for quantitative assessment of small nerve fiber dysfunction as an early feature of diabetic polyneuropathy. Diabetes care, 2005. 28(5): p. 1169-1174.
  78. Viswanathan, V., et al., Early recognition of diabetic neuropathy: evaluation of a simple outpatient procedure using thermal perception. Postgraduate medical journal, 2002. 78(923): p. 541-542.
  79. Martina, I., et al., Measuring vibration threshold with a graduated tuning fork in normal aging and in patients with polyneuropathy. Journal of Neurology, Neurosurgery & Psychiatry, 1998. 65(5): p. 743-747.
  80. Rayman, G., et al., The Ipswich Touch Test: a simple and novel method to identify inpatients with diabetes at risk of foot ulceration. Diabetes care, 2011. 34(7): p. 1517-1518.
  81. Association, A.D., Professional Practice Committee: Standards of Medical Care in Diabetes—2022. Diabetes Care, 2021. 45(Supplement_1): p. S3-S3.
  82. NICE Guidelines for the Management of Diabetes. Available from: https://www.nice.org.uk/guidance/conditions-and-diseases/diabetes-and-other-endocrinal--nutritional-and-metabolic-conditions/diabetes.
  83. Feldman, E.L., et al., A practical two-step quantitative clinical and electrophysiological assessment for the diagnosis and staging of diabetic neuropathy. Diabetes care, 1994. 17(11): p. 1281-1289.
  84. Bril, V. and B.A. Perkins, Validation of the Toronto Clinical Scoring System for diabetic polyneuropathy. Diabetes care, 2002. 25(11): p. 2048-2052.
  85. Bril, V., et al., Reliability and validity of the modified Toronto Clinical Neuropathy Score in diabetic sensorimotor polyneuropathy. Diabetic Medicine, 2009. 26(3): p. 240-246.
  86. Young, M., et al., A multicentre study of the prevalence of diabetic peripheral neuropathy in the United Kingdom hospital clinic population. Diabetologia, 1993. 36(2): p. 150-154.
  87. Dyck, P.J., Detection, characterization, and staging of polyneuropathy: assessed in diabetics. Muscle & Nerve: Official Journal of the American Association of Electrodiagnostic Medicine, 1988. 11(1): p. 21-32.
  88. Bastyr III, E.J., et al., Development and validity testing of the neuropathy total symptom score-6: questionnaire for the study of sensory symptoms of diabetic peripheral neuropathy. Clinical therapeutics, 2005. 27(8): p. 1278-1294.
  89. Freynhagen, R., et al., Pain DETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Current medical research and opinion, 2006. 22(10): p. 1911-1920.
  90. Spallone, V., et al., Validation of DN4 as a screening tool for neuropathic pain in painful diabetic polyneuropathy. Diabetic Medicine, 2012. 29(5): p. 578-585.
  91. Bouhassira, D., et al., Development and validation of the neuropathic pain symptom inventory. Pain, 2004. 108(3): p. 248-257.
  92. Lauria, G., et al., European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Fe‐deration of Neurological Societies and the Peripheral Nerve Society. European journal of neurology, 2010. 17(7): p. 903-e49.
  93. England, J., et al., Practice Parameter: The Evaluation of Distal Symmetric Polyneuropathy: The Role of Autonomic Testing, Nerve Biopsy, and Skin Biopsy (An Evidence‐Based Review) Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM&R, 2009. 1(1): p. 14-22.
  94. Papanas, N. and D. Ziegler, New vistas in the diagnosis of diabetic polyneuropathy. Endocrine, 2014. 47(3): p. 690-698.
  95. Papanas, N., et al., Sensitivity and specificity of a new indicator test (Neuropad) for the diagnosis of peripheral neuropathy in type 2 diabetes patients: a comparison with clinical examination and nerve conduction study. Journal of diabetes and its complications, 2007. 21(6): p. 353-358.
  96. Vinik, A.I., et al., Diabetic nerve conduction abnormalities in the primary care setting. Diabetes technology & therapeutics, 2006. 8(6): p. 654-662,Chatzikosma, G., et al., Evaluation of sural nerve automated nerve conduction study in the diagnosis of peripheral neuropathy in patients with type 2 diabetes mellitus. Archives of medical science: AMS, 2016. 12(2): p. 390.
  97. Lovblom, L.E., et al., In vivo corneal confocal microscopy and prediction of future-incident neuropathy in type 1 diabetes: a preliminary longitudinal analysis. Canadian journal of diabetes, 2015. 39(5): p. 390-397.
  98. Alam, U., et al., Diagnostic utility of corneal confocal microscopy and intra-epidermal nerve fibre density in diabetic neuropathy. PloS one, 2017. 12(7): p. e0180175.
  99. Selvarajah, D., et al., SUDOSCAN: a simple, rapid, and objective method with potential for screening for diabetic peripheral neuropathy. PloS one, 2015. 10(10): p. e0138224.
  100. Mao, F., et al., Sudoscan is an effective screening method for asymptomatic diabetic neuropathy in Chinese type 2 diabetes mellitus patients. Journal of diabetes investigation, 2017. 8(3): p. 363-368.
  101. Lee, J.A., et al., Reliability and validity of a point-of-care sural nerve conduction device for identification of diabetic neuropathy. PloS one, 2014. 9(1): p. e86515.
  102. Umapathi, T., et al., Intraepidermal nerve fiber density as a marker of early diabetic neuropathy. Muscle & Nerve: Official Journal of the American Association of Electrodiagnostic Medicine, 2007. 35(5): p. 591-598.
  103. Quattrini, C., et al., Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes, 2007. 56(8): p. 2148-2154.
  104. Malik, R., et al., Small fibre neuropathy: role in the diagnosis of diabetic sensorimotor polyneuropathy. Diabetes/metabolism research and reviews, 2011. 27(7): p. 678-684.
  105. Papanas, N., et al., Evaluation of a new indicator test for sudomotor function (Neuropad®) in the diagnosis of peripheral neuropathy in type 2 diabetic patients. Experimental and clinical endocrinology & diabetes, 2005. 113(04): p. 195-198.
  106. Ziegler, D., et al., Neuropad: evaluation of three cut‐off points of sudomotor dysfunction for early detection of polyneuropathy in recently diagnosed diabetes. Diabetic Medicine, 2011. 28(11): p. 1412-1415,Spallone, V., et al., Neuropad as a diagnostic tool for diabetic autonomic and sensorimotor neuropathy. Diabetic medicine, 2009. 26(7): p. 686-692.
  107. Ponirakis, G., et al., The diagnostic accuracy of Neuropad® for assessing large and small fibre diabetic neuropathy. Diabetic medicine, 2014. 31(12): p. 1673-1680.
  108. Papanas, N., et al., Reproducibility of the new indicator test for sudomotor function (Neuropad®) in patients with type 2 diabetes mellitus. Experimental and clinical endocrinology & diabetes, 2005. 113(10): p. 577-581.
  109. Tavakoli, M., et al., Corneal confocal microscopy: a novel noninvasive test to diagnose and stratify the severity of human diabetic neuropathy. Diabetes care, 2010. 33(8): p. 1792-1797.
  110. Zhivov, A., et al., Real-time mapping of the subepithelial nerve plexus by in vivo confocal laser scanning microscopy. British journal of ophthalmology, 2010. 94(9): p. 1133-1135.
  111. Casanova-Molla, J., et al., On the relationship between nociceptive evoked potentials and intraepidermal nerve fiber density in painful sensory polyneuropathies. Pain, 2011. 152(2): p. 410-418.
  112. Chao, C.C., et al., Effects of aging on contact heat-evoked potentials: the physiological assessment of thermal perception. Muscle Nerve, 2007. 36(1): p. 30-8.
  113. Smith, A.G., et al., The diagnostic utility of Sudoscan for distal symmetric peripheral neuropathy. Journal of Diabetes and its Complications, 2014. 28(4): p. 511-516,Yajnik, C.S., et al., Quick and simple evaluation of sudomotor function for screening of diabetic neuropathy. International Scholarly Research Notices, 2012. 2012.
  114. Bourcier, M.E., et al., Diabetic peripheral neuropathy: how reliable is a homemade 1-g monofilament for screening? A case-control study of sensitivity, specificity, and comparison with standardized sensory modalities. Journal of family practice, 2006. 55(6): p. 505-509.
  115. Kumar, S., et al., Semmes-Weinstein monofilaments: a simple, effective and inexpensive screening device for identifying diabetic patients at risk of foot ulceration. Diabetes research and clinical practice, 1991. 13(1-2): p. 63-67.
  116. Armstrong, D.G., et al., Choosing a practical screening instrument to identify patients at risk for diabetic foot ulceration. Archives of internal medicine, 1998. 158(3): p. 289-292.
  117. Vinik, A.I., et al., Quantitative measurement of cutaneous perception in diabetic neuropathy. Muscle & Nerve: Official Journal of the American Association of Electrodiagnostic Medicine, 1995. 18(6): p. 574-584.
  118. Abbott, C., et al., The North‐West Diabetes Foot Care Study: incidence of, and risk factors for, new diabetic foot ulceration in a community‐based patient cohort. Diabetic medicine, 2002. 19(5): p. 377-384.
  119. Dyck, P.J., L.J. Melton, and P.C. O'Brien, Approaches to improve epidemiological studies of diabetic neuropathy: insights from the Rochester Diabetic Neuropathy Study. Diabetes, 1997. 46(Supplement 2): p. S5-S8.
  120. Herman, W., et al., Use of the Michigan Neuropathy Screening Instrument as a measure of distal symmetrical peripheral neuropathy in Type 1 diabetes: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications. Diabetic medicine, 2012. 29(7): p. 937-944.
  121. Dyck, P.J., et al., “Unequivocally abnormal” vs “usual” signs and symptoms for proficient diagnosis of diabetic polyneuropathy: Cl vs N Phys Trial. Archives of neurology, 2012. 69(12): p. 1609-1614.
  122. Feng, Y., F.J. Schlösser, and B.E. Sumpio, The Semmes Weinstein monofilament examination as a screening tool for diabetic peripheral neuropathy. Journal of vascular surgery, 2009. 50(3): p. 675-682. e1.
  123. Richard, J.L., et al., Screening patients at risk for diabetic foot ulceration: a comparison between measurement of vibration perception threshold and 10‐g monofilament test. International wound journal, 2014. 11(2): p. 147-151.
  124. Tan, L.S., The clinical use of the 10 g monofilament and its limitations: a review. Diabetes research and clinical practice, 2010. 90(1): p. 1-7.
  125. Weisman, A., et al., Identification and prediction of diabetic sensorimotor polyneuropathy using individual and simple combinations of nerve conduction study parameters. PLoS One, 2013. 8(3): p. e58783.
  126. The Diabetic Retinopathy Barometer Report Global Findings.[cited 2019 28th December]; Available from: https://www.idf.org/our-activities/advocacy-awareness/resources-and-tools/92:diabetic-retinopathy-barometer.html
  127. Liew, G., M. Michaelides, and C. Bunce, A comparison of the causes of blindness certifications in England and Wales in working age adults (16–64 years), 1999–2000 with 2009–2010. BMJ open, 2014. 4(2): p. e004015.
  128. Marshall, S., Diabetic nephropathy in type 1 diabetes: has the outlook improved since the 1980s? Diabetologia, 2012. 55(9): p. 2301-2306.
  129. Dyck, P.J., et al., Patterns of quantitative sensation testing of hypoesthesia and hyperalgesia are predictive of diabetic polyneuropathy: a study of three cohorts. Nerve growth factor study group. Diabetes Care, 2000. 23(4): p. 510-517.
  130. Yarnitsky, D. and E. Sprecher, Thermal testing: normative data and repeatability for various test algorithms. Journal of the neurological sciences, 1994. 125(1): p. 39-45.
  131. Shy, M., et al., Quantitative sensory testing: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology, 2003. 60(6): p. 898-904.
  132. Society, P.N., Diabetic polyneuropathy in controlled clinical trials: consensus report of the Peripheral Nerve Society. Annals of Neurology, 1995. 38(3): p. 478-482.
  133. Gibbons, C.H., Small fiber neuropathies. CONTINUUM: Lifelong Learning in Neurology, 2014. 20(5): p. 1398-1412.
  134. Pittenger, G.L., et al., Intraepidermal nerve fibers are indicators of small-fiber neuropathy in both diabetic and nondiabetic patients. Diabetes care, 2004. 27(8): p. 1974-1979.
  135. Polydefkis, M., et al., Skin biopsy as a tool to assess distal small fiber innervation in diabetic neuropathy. Diabetes technology & therapeutics, 2001. 3(1): p. 23-28.
  136. Smith, A.G., et al., Lifestyle intervention for pre-diabetic neuropathy. Diabetes care, 2006. 29(6): p. 1294-1299.
  137. Vileikyte, L., et al., The development and validation of a neuropathy-and foot ulcer-specific quality of life instrument. Diabetes care, 2003. 26(9): p. 2549-2555.
  138. Vinik, E.J., et al., The development and validation of the Norfolk QOL-DN, a new measure of patients' perception of the effects of diabetes and diabetic neuropathy. Diabetes technology & therapeutics, 2005. 7(3): p. 497-508.
  139. Selvarajah, D., et al., Early involvement of the spinal cord in diabetic peripheral neuropathy. Diabetes care, 2006. 29(12): p. 2664-2669.
  140. Eaton, S.E., et al., Spinal-cord involvement in diabetic peripheral neuropathy. The Lancet, 2001. 358(9275): p. 35-36.
  141. Selvarajah, D., et al., Magnetic resonance neuroimaging study of brain structural differences in diabetic peripheral neuropathy. Diabetes care, 2014. 37(6): p. 1681-1688.
  142. Selvarajah, D., et al., Thalamic neuronal dysfunction and chronic sensorimotor distal symmetrical polyneuropathy in patients with type 1 diabetes mellitus. Diabetologia, 2008. 51(11): p. 2088-2092.
  143. Hansen, T.M., et al., Brain spectroscopy reveals that N-acetylaspartate is associated to peripheral sensorimotor neuropathy in type 1 diabetes. Journal of Diabetes and its Complications, 2019. 33(4): p. 323-328.
  144. Selvarajah, D., et al., Microvascular perfusion abnormalities of the Thalamus in painful but not painless diabetic polyneuropathy: a clue to the pathogenesis of pain in type 1 diabetes. Diabetes care, 2011. 34(3): p. 718-720.
  145. Cauda, F., et al., Low-frequency BOLD fluctuations demonstrate altered thalamocortical connectivity in diabetic neuropathic pain. BMC neuroscience, 2009. 10(1): p. 1-14.
  146. Selvarajah, D., et al., Structural and functional abnormalities of the primary somatosensory cortex in diabetic peripheral neuropathy: a multimodal MRI study. Diabetes, 2019. 68(4): p. 796-806.
  147. Reske-Nielsen, E. and K. Lundbæk, Pathological changes in the central and peripheral nervous system of young long-term diabetics. Diabetologia, 1968. 4(1): p. 34-43.
  148. Reske-Nielsen, E., et al., Pathological changes in the central and peripheral nervous system of young long-term diabetics. The terminal neuro-muscular apparatus. Diabetologia, 1970. 6(2): p. 98-103.
  149. Ziegler, D., et al., Tibial nerve somatosensory evoked potentials at various stages of peripheral neuropathy in insulin dependent diabetic patients. Journal of Neurology, Neurosurgery & Psychiatry, 1993. 56(1): p. 58-64.
  150. Biessels, G.-J., et al., Neurophysiological changes in the central and peripheral nervous system of streptozotocin-diabetic rats: course of development and effects of insulin treatment. Brain, 1999. 122(4): p. 757-768.
  151. Group, L.A.R., Effects of a long-term lifestyle modification programme on peripheral neuropathy in overweight or obese adults with type 2 diabetes: the Look AHEAD study. Diabetologia, 2017. 60(6): p. 980.
  152. Control, D. and C.T.R. Group, The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. New England journal of medicine, 1993. 329(14): p. 977-986.
  153. Group, U.P.D.S., Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). The lancet, 1998. 352(9131): p. 837-853.
  154. Albers, J.W., et al., Effect of prior intensive insulin treatment during the Diabetes Control and Complications Trial (DCCT) on peripheral neuropathy in type 1 diabetes during the Epidemiology of Diabetes Interventions and Complications (EDIC) Study. Diabetes care, 2010. 33(5): p. 1090-1096.
  155. Pop-Busui, R., et al., Effects of prior intensive insulin therapy on cardiac autonomic nervous system function in type 1 diabetes mellitus: the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications study (DCCT/EDIC). Circulation, 2009. 119(22): p. 2886-2893.
  156. Group, A.C., Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. New England journal of medicine, 2008. 358(24): p. 2560-2572.
  157. Duckworth, W., et al., Glucose control and vascular complications in veterans with type 2 diabetes. New England journal of medicine, 2009. 360(2): p. 129-139.
  158. Gæde, P., et al., Effect of a multifactorial intervention on mortality in type 2 diabetes. New England Journal of Medicine, 2008. 358(6): p. 580-591.
  159. Vincent, A.M., et al., Hyperlipidemia: a new therapeutic target for diabetic neuropathy. Journal of the Peripheral Nervous System, 2009. 14(4): p. 257-267.
  160. Callaghan, B.C., et al., Triglycerides and amputation risk in patients with diabetes: ten-year follow-up in the DISTANCE study. Diabetes care, 2011. 34(3): p. 635-640.
  161. Wiggin, T.D., et al., Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes, 2009. 58(7): p. 1634-1640.
  162. Knopp, R.H., et al., Efficacy and safety of atorvastatin in the prevention of cardiovascular end points in subjects with type 2 diabetes: the Atorvastatin Study for Prevention of Coronary Heart Disease Endpoints in non-insulin-dependent diabetes mellitus (ASPEN). Diabetes care, 2006. 29(7): p. 1478-1485,Group, H.P.S.C., MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. The Lancet, 2003. 361(9374): p. 2005-2016.
  163. Colhoun, H.M., et al., Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. The Lancet, 2004. 364(9435): p. 685-696.
  164. Davis, T., et al., Lipid-lowering therapy and peripheral sensory neuropathy in type 2 diabetes: the Fremantle Diabetes Study. Diabetologia, 2008. 51(4): p. 562-566.
  165. Zangiabadi, N., et al., Atorvastatin treatment improves diabetic polyneuropathy electrophysiological changes in non-insulin dependent diabetic patients: a double blind, randomized clinical trial. Minerva Endocrinol, 2012. 37(2): p. 195-200.
  166. Hernández-Ojeda, J., et al., Effect of rosuvastatin on diabetic polyneuropathy: a randomized, double-blind, placebo-controlled Phase IIa study. Diabetes, metabolic syndrome and obesity: targets and therapy, 2014. 7: p. 401.
  167. Ansquer, J., et al., Fibrates and microvascular complications in diabetes-insight from the FIELD study. Current pharmaceutical design, 2009. 15(5): p. 537-552.
  168. Nielsen, S.F. and B.G. Nordestgaard, Statin use before diabetes diagnosis and risk of microvascular disease: a nationwide nested matched study. The lancet Diabetes & endocrinology, 2014. 2(11): p. 894-900.
  169. Jarmuzewska, E., A. Ghidoni, and A.A. Mangoni, Hypertension and sensorimotor peripheral neuropathy in type 2 diabetes. European neurology, 2007. 57(2): p. 91-95.
  170. Estacio, R.O., et al., Effect of blood pressure control on diabetic microvascular complications in patients with hypertension and type 2 diabetes. Diabetes care, 2000. 23: p. B54.
  171. Forrest, K.Y., et al., Hypertension as a risk factor for diabetic neuropathy: a prospective study. Diabetes, 1997. 46(4): p. 665-670.
  172. Malik, R.A., et al., Effect of angiotensin-converting-enzyme (ACE) inhibitor trandolapril on human diabetic neuropathy: randomised double-blind controlled trial. The Lancet, 1998. 352(9145): p. 1978-1981.
  173. Ruggenenti, P., et al., Effects of manidipine and delapril in hypertensive patients with type 2 diabetes mellitus: the delapril and manidipine for nephroprotection in diabetes (DEMAND) randomized clinical trial. Hypertension, 2011. 58(5): p. 776-783.
  174. Callaghan, B.C., et al., Metabolic syndrome components are associated with symptomatic polyneuropathy independent of glycemic status. Diabetes care, 2016. 39(5): p. 801-807.
  175. Costa, L., et al., Aggregation of features of the metabolic syndrome is associated with increased prevalence of chronic complications in type 2 diabetes. Diabetic Medicine, 2004. 21(3): p. 252-255.
  176. Ylitalo, K.R., M. Sowers, and S. Heeringa, Peripheral vascular disease and peripheral neuropathy in individuals with cardiometabolic clustering and obesity: National Health and Nutrition Examination Survey 2001–2004. Diabetes care, 2011. 34(7): p. 1642-1647.
  177. Callaghan, B.C., et al., Association between metabolic syndrome components and polyneuropathy in an obese population. JAMA neurology, 2016. 73(12): p. 1468-1476.
  178. Kluding, P.M., et al., The effect of exercise on neuropathic symptoms, nerve function, and cutaneous innervation in people with diabetic peripheral neuropathy. Journal of Diabetes and its Complications, 2012. 26(5): p. 424-429.
  179. Balducci, S., et al., Exercise training can modify the natural history of diabetic peripheral neuropathy. Journal of diabetes and its complications, 2006. 20(4): p. 216-223.
  180. Singleton, J.R., et al., Supervised exercise improves cutaneous reinnervation capacity in metabolic syndrome patients. Annals of neurology, 2015. 77(1): p. 146-153.
  181. Ziegler, D., C.G. Sohr, and J. Nourooz-Zadeh, Oxidative stress and antioxidant defense in relation to the severity of diabetic polyneuropathy and cardiovascular autonomic neuropathy. Diabetes care, 2004. 27(9): p. 2178-2183.
  182. Miyauchi, Y., et al., Slowing of peripheral motor nerve conduction was ameliorated by aminoguanidine in streptozocin-induced diabetic rats. European journal of endocrinology, 1996. 134(4): p. 467-473.
  183. Haupt, E., H. Ledermann, and W. Köpcke, Benfotiamine in the treatment of diabetic. International journal of clinical pharmacology and therapeutics, 2005. 43(2): p. 71-77.
  184. Stracke, H., A. Lindemann, and K. Federlin, A benfotiamine-vitamin B combination in treatment of diabetic polyneuropathy. Experimental and clinical endocrinology & diabetes, 1996. 104(04): p. 311-316.
  185. Stracke, H., et al., Benfotiamine in diabetic polyneuropathy (BENDIP): results of a randomised, double blind, placebo-controlled clinical study. Experimental and clinical endocrinology & diabetes, 2008. 116(10): p. 600-605.
  186. Greene, D.A., et al., Effect of aldose reductase inhibition on nerve conduction and morphometry in diabetic neuropathy. Neurology, 1999. 53(3): p. 580-580.
  187. Hotta, N., et al., Clinical efficacy of fidarestat, a novel aldose reductase inhibitor, for diabetic peripheral neuropathy: a 52-week multicenter placebo-controlled double-blind parallel group study. Diabetes care, 2001. 24(10): p. 1776-1782.
  188. Hotta, N., et al., Long-term effects of epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy: a 3-y multicenter comparative study, ARI-Diabetes Complications Trial (ADCT). Diabetes, 2005. 54: p. A213.
  189. Bril, V. and R.A. Buchanan, Long-term effects of ranirestat (AS-3201) on peripheral nerve function in patients with diabetic sensorimotor polyneuropathy. Diabetes care, 2006. 29(1): p. 68-72.
  190. Keen, H., et al., Treatment of diabetic neuropathy with γ-linolenic acid. Diabetes Care, 1993. 16(1): p. 8-15.
  191. Ametov, A.S., et al., The sensory symptoms of diabetic polyneuropathy are improved with α-lipoic acid. Diabetes care, 2003. 26(3): p. 770-776.
  192. Reljanovic, M., et al., Treatment of diabetic polyneuropathy with the antioxidant thioctic acid (α-lipoic acid): a two year multicenter randomized double-blind placebo-controlled trial (ALADIN II). Free radical research, 1999. 31(3): p. 171-179.
  193. Ziegler, D., et al., Treatment of symptomatic diabetic polyneuropathy with the antioxidant α‐lipoic acid: a meta‐analysis. Diabetic Medicine, 2004. 21(2): p. 114-121.
  194. Ziegler, D., et al., Oral treatment with α-lipoic acid improves symptomatic diabetic polyneuropathy: the SYDNEY 2 trial. Diabetes care, 2006. 29(11): p. 2365-2370.
  195. Ziegler, D., et al., Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes care, 2011. 34(9): p. 2054-2060.
  196. McIlduff, C.E. and S.B. Rutkove, Critical appraisal of the use of alpha lipoic acid (thioctic acid) in the treatment of symptomatic diabetic polyneuropathy. Therapeutics and clinical risk management, 2011. 7: p. 377.
  197. Mijnhout, G.S., et al., Alpha lipoic acid for symptomatic peripheral neuropathy in patients with diabetes: a meta-analysis of randomized controlled trials. International Journal of Endocrinology, 2012. 2012.
  198. Vinik, A.I., et al., Treatment of symptomatic diabetic peripheral neuropathy with the protein kinase C β-inhibitor ruboxistaurin mesylate during a 1-year, randomized, placebo-controlled, double-blind clinical trial. Clinical therapeutics, 2005. 27(8): p. 1164-1180.
  199. Vinik, A.I., et al., Sural sensory action potential identifies diabetic peripheral neuropathy responders to therapy. Muscle & nerve, 2005. 32(5): p. 619-625.
  200. Boyd, A., et al., Quality of life and objective measures of diabetic neuropathy in a prospective placebo-controlled trial of ruboxistaurin and topiramate. Journal of Diabetes Science and Technology, 2011. 5(3): p. 714-722.
  201. Forst, T., et al., Molecular effects of C-Peptide in microvascular blood flow regulation. The review of diabetic studies: RDS, 2009. 6(3): p. 159.
  202. Wahren, J., Å. Kallas, and A.A. Sima, The clinical potential of C-peptide replacement in type 1 diabetes. Diabetes, 2012. 61(4): p. 761-772.
  203. Ekberg, K., et al., C-Peptide Replacement Therapy and Sensory Nerve Function in Type 1 Diabetic Neuropathy. Diabetes Care, 2007. 30(1): p. 71-76.
  204. Pittenger, G. and A. Vinik, Nerve growth factor and diabetic neuropathy. Experimental diabesity research, 2003. 4(4): p. 271-285.
  205. VINIK, A., Treatment of diabetic polyneuropathy (DPN) with recombinant human nerve growth factor (rhNGF). Diabetes, 1999. 48(5): p. SA54-SA54.
  206. Rivard, A., et al., Rescue of diabetes-related impairment of angiogenesis by intramuscular gene therapy with adeno-VEGF. The American journal of pathology, 1999. 154(2): p. 355-363.
  207. Kessler, J.A., et al., Gene therapy for diabetic peripheral neuropathy: A randomized, placebo‐controlled phase III study of VM202, a plasmid DNA encoding human hepatocyte growth factor. Clinical and Translational Science, 2021. 14(3): p. 1176-1184.
  208. Kessler, J.A., et al., Double‐blind, placebo‐controlled study of HGF gene therapy in diabetic neuropathy. Annals of clinical and translational neurology, 2015. 2(5): p. 465-478.
  209. Ajroud-Driss, S., et al., Phase 1/2 open-label dose-escalation study of plasmid DNA expressing two isoforms of hepatocyte growth factor in patients with painful diabetic peripheral neuropathy. Molecular Therapy, 2013. 21(6): p. 1279-1286.
  210. Tam, J., L. Rosenberg, and D. Maysinger, INGAP peptide improves nerve function and enhances regeneration in streptozotocin‐induced diabetic C57BL/6 mice. The FASEB journal, 2004. 18(14): p. 1767-1769.
  211. Granberg, V., et al., Autoantibodies to autonomic nerves associated with cardiac and peripheral autonomic neuropathy. Diabetes care, 2005. 28(8): p. 1959-1964.
  212. Vinik, A.I., D. Anandacoomaraswamy, and J. Ullal, Antibodies to neuronal structures: innocent bystanders or neurotoxins? Diabetes care, 2005. 28(8): p. 2067-2072.
  213. Pain, I.A.f.t.S.o., Pain terms. Neuropathic Pain. IASP Taxonomy, 2017.
  214. Colloca, L., et al., Neuropathic pain. Nature reviews Disease primers, 2017. 3(1): p. 1-19.
  215. Bennett, D.L., et al., The role of voltage-gated sodium channels in pain signaling. Physiological reviews, 2019. 99(2): p. 1079-1151.
  216. Saegusa, H., et al., Suppression of inflammatory and neuropathic pain symptoms in mice lacking the N‐type Ca2+ channel. The EMBO journal, 2001. 20(10): p. 2349-2356.
  217. Finnerup, N.B., Nonnarcotic methods of pain management. New England Journal of Medicine, 2019. 380(25): p. 2440-2448.
  218. Binns‐Hall, O., et al., One‐stop microvascular screening service: an effective model for the early detection of diabetic peripheral neuropathy and the high‐risk foot. Diabetic Medicine, 2018. 35(7): p. 887-894.
  219. Daousi, C., et al., Chronic painful peripheral neuropathy in an urban community: a controlled comparison of people with and without diabetes. Diabetic medicine, 2004. 21(9): p. 976-982.
  220. Bennett, M.I., et al., The S-LANSS score for identifying pain of predominantly neuropathic origin: validation for use in clinical and postal research. The Journal of Pain, 2005. 6(3): p. 149-158.
  221. Krause, S.J. and M.-M. Backonja, Development of a neuropathic pain questionnaire. The Clinical journal of pain, 2003. 19(5): p. 306-314.
  222. Dworkin, R.H., et al., Development and initial validation of an expanded and revised version of the Short-form McGill Pain Questionnaire (SF-MPQ-2). Pain®, 2009. 144(1-2): p. 35-42.
  223. Daut, R.L., C.S. Cleeland, and R.C. Flanery, Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases. Pain, 1983. 17(2): p. 197-210.
  224. Freeman, R., et al., Sensory profiles of patients with neuropathic pain based on the neuropathic pain symptoms and signs. PAIN®, 2014. 155(2): p. 367-376.
  225. Dworkin, R.H., et al., Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. The journal of pain, 2008. 9(2): p. 105-121.
  226. Davies, M., et al., The prevalence, severity, and impact of painful diabetic peripheral neuropathy in type 2 diabetes. Diabetes care, 2006. 29(7): p. 1518-1522.
  227. Tölle, T., X. Xu, and A.B. Sadosky, Painful diabetic neuropathy: a cross-sectional survey of health state impairment and treatment patterns. Journal of Diabetes and its Complications, 2006. 20(1): p. 26-33.
  228. Alleman, C.J., et al., Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: a review of the literature. Diabetes research and clinical practice, 2015. 109(2): p. 215-225.
  229. Woolf, C.J., et al., Towards a mechanism-based classification of pain? 1998, LWW. p. 227-229.
  230. Edwards, R.R., et al., Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. Pain, 2016. 157(9): p. 1851.
  231. Maier, C., et al., Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. Pain, 2010. 150(3): p. 439-450.
  232. Rolke, R., et al., Quantitative sensory testing: a comprehensive protocol for clinical trials. European journal of pain, 2006. 10(1): p. 77-88.
  233. Themistocleous, A.C., et al., The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. Pain, 2016. 157(5): p. 1132.
  234. Demant, D.T., et al., The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. PAIN®, 2014. 155(11): p. 2263-2273.
  235. Wilkinson, I.D., et al., Determinants of Treatment Response in Painful Diabetic Peripheral Neuropathy: A Combined Deep Sensory Phenotyping and Multimodal Brain MRI Study. Diabetes, 2020. 69(8): p. 1804-1814.
  236. Campbell, C.M., et al., Randomized control trial of topical clonidine for treatment of painful diabetic neuropathy. PAIN®, 2012. 153(9): p. 1815-1823.
  237. Bouhassira, D., et al., Neuropathic pain phenotyping as a predictor of treatment response in painful diabetic neuropathy: data from the randomized, double-blind, COMBO-DN study. Pain®, 2014. 155(10): p. 2171-2179.
  238. Yarnitsky, D., et al., Conditioned pain modulation predicts duloxetine efficacy in painful diabetic neuropathy. Pain, 2012. 153(6): p. 1193-1198.
  239. Haroutounian, S., et al., Primary afferent input critical for maintaining spontaneous pain in peripheral neuropathy. PAIN®, 2014. 155(7): p. 1272-1279.
  240. Wager, T.D., et al., An fMRI-based neurologic signature of physical pain. New England Journal of Medicine, 2013. 368(15): p. 1388-1397.
  241. Tracey, I., “Seeing” how our drugs work brings translational added value. Anesthesiology, 2013. 119(6): p. 1247-1248.
  242. Buxton, R., Interpreting oxygenation-based neuroimaging signals: the importance and the challenge of understanding brain oxygen metabolism. Frontiers in neuroenergetics, 2010. 2: p. 8.
  243. Pereira, F., T. Mitchell, and M. Botvinick, Machine learning classifiers and fMRI: a tutorial overview. Neuroimage, 2009. 45(1): p. S199-S209.
  244. Wager, T.D., et al., Evaluating the consistency and specificity of neuroimaging data using meta-analysis. Neuroimage, 2009. 45(1): p. S210-S221.
  245. Alger, J.R., et al., Multisite, multimodal neuroimaging of chronic urological pelvic pain: Methodology of the MAPP Research Network. NeuroImage: Clinical, 2016. 12: p. 65-77.
  246. Zunhammer, M., et al., Placebo effects on the neurologic pain signature: a meta-analysis of individual participant functional magnetic resonance imaging data. JAMA neurology, 2018. 75(11): p. 1321-1330.
  247. Dworkin, R.H., et al., Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain, 2007. 132(3): p. 237-251.
  248. Finnerup, N.B., S.H. Sindrup, and T.S. Jensen, The evidence for pharmacological treatment of neuropathic pain. Pain, 2010. 150(3): p. 573-581.
  249. Ziegler, D. and V. Fonseca, From guideline to patient: a review of recent recommendations for pharmacotherapy of painful diabetic neuropathy. Journal of Diabetes and its Complications, 2015. 29(1): p. 146-156.
  250. Bril, V., et al., Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Pm&r, 2011. 3(4): p. 345-352. e21.
  251. Bril, V., et al., Neuropathy. Canadian journal of diabetes, 2018. 42: p. S217-S221.
  252. Ziegler, D., et al., Diabetic neuropathy. Experimental and Clinical Endocrinology & Diabetes, 2014. 122(07): p. 406-415.
  253. Finnerup, N.B., et al., Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. The Lancet Neurology, 2015. 14(2): p. 162-173.
  254. Moisset, X., et al., Pharmacological and non-pharmacological treatments for neuropathic pain: systematic review and French recommendations. Revue neurologique, 2020. 176(5): p. 325-352.
  255. Sumitani, M., et al., Executive summary of the clinical guidelines of pharmacotherapy for neuropathic pain: by the Japanese society of pain clinicians. Journal of anesthesia, 2018. 32(3): p. 463-478.
  256. Attal, N., et al., EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. European journal of neurology, 2010. 17(9): p. 1113-e88.
  257. Max, M.B., et al., Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. New England Journal of Medicine, 1992. 326(19): p. 1250-1256.
  258. Lunn, M.P., R.A. Hughes, and P.J. Wiffen, Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database of Systematic Reviews, 2014(1).
  259. Waldfogel, J.M., et al., Pharmacotherapy for diabetic peripheral neuropathy pain and quality of life: a systematic review. Neurology, 2017. 88(20): p. 1958-1967.
  260. Dy, S.M., et al., Preventing complications and treating symptoms of diabetic peripheral neuropathy. 2017.
  261. Hardy, T., et al., Does treatment with duloxetine for neuropathic pain impact glycemic control? Diabetes Care, 2007. 30(1): p. 21-26.
  262. Sansone, R.A. and L.A. Sansone, Pain, pain, go away: antidepressants and pain management. Psychiatry (Edgmont), 2008. 5(12): p. 16.
  263. Simpson, D.A., Gabapentin and venlafaxine for the treatment of painful diabetic neuropathy. Journal of clinical neuromuscular disease, 2001. 3(2): p. 53-62.
  264. Rowbotham, M.C., et al., Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain, 2004. 110(3): p. 697-706.
  265. Wiffen, P.J., et al., Gabapentin for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews, 2017(6).
  266. Freeman, R., E. Durso-DeCruz, and B. Emir, Efficacy, safety, and tolerability of pregabalin treatment for painful diabetic peripheral neuropathy: findings from seven randomized, controlled trials across a range of doses. Diabetes care, 2008. 31(7): p. 1448-1454.
  267. Derry, S., et al., Pregabalin for neuropathic pain in adults. Cochrane Database of Systematic Reviews, 2019(1).
  268. Semel, D., et al., Evaluation of the safety and efficacy of pregabalin in older patients with neuropathic pain: results from a pooled analysis of 11 clinical studies. BMC family practice, 2010. 11(1): p. 1-12.
  269. Evoy, K.E., et al., Abuse and misuse of pregabalin and gabapentin: a systematic review update. Drugs, 2021. 81(1): p. 125-156.
  270. Gabapentin and risk of Severe Respiratory Depression. Drug Ther Bull., 2018. 53(1): p. 3-4.
  271. Meisenberg, B., et al., Implementation of solutions to reduce opioid-induced oversedation and respiratory depression. American Journal of Health-System Pharmacy, 2017. 74(3): p. 162-169.
  272. Eipe, N. and J. Penning, Postoperative respiratory depression associated with pregabalin: A case series and a preoperative decision algorithm. Pain Research and Management, 2011. 16(5): p. 353-356.
  273. Rains, C. and H.M. Bryson, Topical capsaicin. Drugs & aging, 1995. 7(4): p. 317-328.
  274. Backonja, M., et al., NGX-4010, a high-concentration capsaicin patch, for the treatment of postherpetic neuralgia: a randomised, double-blind study. The Lancet Neurology, 2008. 7(12): p. 1106-1112.
  275. Bonezzi, C., et al., Capsaicin 8% dermal patch in clinical practice: an expert opinion. Expert opinion on pharmacotherapy, 2020. 21(11): p. 1377-1387.
  276. Derry, S. and R.A. Moore, Topical capsaicin (low concentration) for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews, 2012(9).
  277. Omana-Zapata, I., et al., QX-314 inhibits ectopic nerve activity associated with neuropathic pain. Brain research, 1997. 771(2): p. 228-237.
  278. Devor, M., P.D. Wall, and N. Catalan, Systemic lidocaine silences ectopic neuroma and DRG discharge without blocking nerve conduction. Pain, 1992. 48(2): p. 261-268.
  279. Abdi, S., D.H. Lee, and J.M. Chung, The anti-allodynic effects of amitriptyline, gabapentin, and lidocaine in a rat model of neuropathic pain. Anesthesia & Analgesia, 1998. 87(6): p. 1360-1366.
  280. Sheets, P.L., B.W. Jarecki, and T.R. Cummins, Lidocaine reduces the transition to slow inactivation in Nav1. 7 voltage‐gated sodium channels. British journal of pharmacology, 2011. 164(2b): p. 719-730.
  281. Jasmin, L., et al., The cold plate as a test of nociceptive behaviors: description and application to the study of chronic neuropathic and inflammatory pain models. Pain, 1998. 75(2-3): p. 367-382.
  282. Chaplan, S.R., et al., Prolonged alleviation of tactile allodynia by intravenous lidocaine in neuropathic rats. The Journal of the American Society of Anesthesiologists, 1995. 83(4): p. 775-785.
  283. BARTLETT, E.E. and O. HUTASERANI, Xylocaine for the relief of postoperative pain. Anesthesia & Analgesia, 1961. 40(3): p. 296-304.
  284. Challapalli, V., et al., Systemic administration of local anesthetic agents to relieve neuropathic pain. Cochrane Database of Systematic Reviews, 2005(4).
  285. McGavin, C., S. Gupta, and G. McHardy, Treatment of chronic painful diabetic neuropathy with intravenous lidocaine inusion. British Medical Journal, 1986. 173.
  286. Kastrup, J., et al., Intravenous lidocaine infusion—a new treatment of chronic painful diabetic neuropathy? Pain, 1987. 28(1): p. 69-75.
  287. Viola, V., H.H. Newnham, and R.W. Simpson, Treatment of intractable painful diabetic neuropathy with intravenous lignocaine. Journal of Diabetes and its Complications, 2006. 20(1): p. 34-39.
  288. Tesfaye, S., et al., Painful diabetic peripheral neuropathy: consensus recommendations on diagnosis, assessment and management. Diabetes/metabolism research and reviews, 2011. 27(7): p. 629-638.
  289. Finnerup, N.B., S.H. Sindrup, and T.S. Jensen, Management of painful neuropathies. Handbook of Clinical Neurology, 2013. 115: p. 279-290.
  290. Duehmke, R.M., et al., Tramadol for neuropathic pain in adults. Cochrane Database of Systematic Reviews, 2017(6).
  291. Gaskell, H., et al., Oxycodone for neuropathic pain in adults. Cochrane Database of Systematic Reviews, 2016(7).
  292. Häuser, W., et al., European* clinical practice recommendations on opioids for chronic noncancer pain–Part 1: Role of opioids in the management of chronic noncancer pain. European Journal of Pain, 2021. 25(5): p. 949-968.
  293. Hoffman, E.M., et al., Association of long-term opioid therapy with functional status, adverse outcomes, and mortality among patients with polyneuropathy. JAMA neurology, 2017. 74(7): p. 773-779.
  294. Tesfaye, S., Painful diabetic neuropathy. Aetiology and nonpharmacological treatment. Clinical management of diabetic neuropathy. Humana Press, Totowa, New jersey, 1998.
  295. Abuaisha, B., J. Costanzi, and A. Boulton, Acupuncture for the treatment of chronic painful peripheral diabetic neuropathy: a long-term study. Diabetes research and clinical practice, 1998. 39(2): p. 115-121.
  296. Garrow, A.P., et al., Role of acupuncture in the management of diabetic painful neuropathy (DPN): a pilot RCT. Acupuncture in Medicine, 2014. 32(3): p. 242-249.
  297. Chen, W., et al., Manual acupuncture for treatment of diabetic peripheral neuropathy: a systematic review of randomized controlled trials. PloS one, 2013. 8(9): p. e73764.
  298. Kumar, D., et al., Diabetic peripheral neuropathy: effectiveness of electrotherapy and amitriptyline for symptomatic relief. Diabetes Care, 1998. 21(8): p. 1322-1325.
  299. Bosi, E., et al., Effectiveness of frequency-modulated electromagnetic neural stimulation in the treatment of painful diabetic neuropathy. Diabetologia, 2005. 48(5): p. 817-823.
  300. Bosi, E., et al., Frequency-modulated electromagnetic neural stimulation (FREMS) as a treatment for symptomatic diabetic neuropathy: results from a double-blind, randomised, multicentre, long-term, placebo-controlled clinical trial. Diabetologia, 2013. 56(3): p. 467-475.
  301. Thakral, G., et al., Electrical stimulation as an adjunctive treatment of painful and sensory diabetic neuropathy. Journal of diabetes science and technology, 2013. 7(5): p. 1202-1209.
  302. Tesfaye, S., et al., Electrical spinal-cord stimulation for painful diabetic peripheral neuropathy. The Lancet, 1996. 348(9043): p. 1698-1701.
  303. Kumar, K., C. Toth, and R.K. Nath, Spinal cord stimulation for chronic pain in peripheral neuropathy. Surgical neurology, 1996. 46(4): p. 363-369.
  304. de Vos, C.C., et al., Spinal cord stimulation in patients with painful diabetic neuropathy: a multicentre randomized clinical trial. PAIN®, 2014. 155(11): p. 2426-2431.
  305. van Beek, M., et al., Sustained treatment effect of spinal cord stimulation in painful diabetic peripheral neuropathy: 24-month follow-up of a prospective two-center randomized controlled trial. Diabetes Care, 2015. 38(9): p. e132-e134.
  306. Slangen, R., et al., Spinal cord stimulation and pain relief in painful diabetic peripheral neuropathy: a prospective two-center randomized controlled trial. Diabetes Care, 2014. 37(11): p. 3016-3024.
  307. van Beek, M., et al., Severity of neuropathy is associated with long-term spinal cord stimulation outcome in painful diabetic peripheral neuropathy: five-year follow-up of a prospective two-center clinical trial. Diabetes Care, 2018. 41(1): p. 32-38.
  308. Sills, S., Treatment of painful polyneuropathies of diabetic and other origins with 10 kHz SCS: a case series. Postgraduate medicine, 2020. 132(4): p. 352-357.
  309. Galan, V., et al., 10-kHz spinal cord stimulation treatment for painful diabetic neuropathy: results from post-hoc analysis of the SENZA-PPN study. Pain Management, 2020. 10(5): p. 291-300.
  310. Bradley, K. and C. Redwood City, Paresthesia-independence: an assessment of technical factors related to 10 kHz paresthesia-free spinal cord stimulation. Pain Physician, 2017. 20: p. 331-341.
  311. Petersen, E.A., et al., Effect of high-frequency (10-kHz) spinal cord stimulation in patients with painful diabetic neuropathy: a randomized clinical trial. JAMA neurology, 2021.
  312. Chaudhry, V., et al., Practice Advisory: utility of surgical decompression for treatment of diabetic neuropathy: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology, 2006. 66(12): p. 1805-1808.
  313. Chaudhry, V., J. Russell, and A. Belzberg, Decompressive surgery of lower limbs for symmetrical diabetic peripheral neuropathy. Cochrane Database of Systematic Reviews, 2008(3).
  314. Selvarajah, D., et al., Multicentre, double-blind, crossover trial to identify the Optimal Pathway for TreatIng neurOpathic paiN in Diabetes Mellitus (OPTION-DM): study protocol for a randomised controlled trial. Trials, 2018. 19(1): p. 1-12.
  315. Vinik, A.I. and T. Erbas, Diabetic autonomic neuropathy. Handbook of clinical neurology, 2013. 117: p. 279-294.
  316. Pfeifer, M.A., et al., Autonomic neural dysfunction in recently diagnosed diabetic subjects. Diabetes care, 1984. 7(5): p. 447-453.
  317. Stansberry, K.B., et al., Impairment of peripheral blood flow responses in diabetes resembles an enhanced aging effect. Diabetes care, 1997. 20(11): p. 1711-1716.
  318. Stansberry, K.B., et al., Impaired peripheral vasomotion in diabetes. Diabetes care, 1996. 19(7): p. 715-721.
  319. Stansberry, K.B., et al., Primary nociceptive afferents mediate the blood flow dysfunction in non-glabrous (hairy) skin of type 2 diabetes: a new model for the pathogenesis of microvascular dysfunction. Diabetes Care, 1999. 22(9): p. 1549-1554.
  320. Haak, E.S., et al., The effect of α-lipoic acid on the neurovascular reflex arc in patients with diabetic neuropathy assessed by capillary microscopy. Microvascular Research, 1999. 58(1): p. 28-34.
  321. Ziegler, D., Diabetic cardiovascular autonomic neuropathy: prognosis, diagnosis and treatment. Diabetes/metabolism reviews, 1994. 10(4): p. 339-383.
  322. Valensi, P., Diabetic autonomic neuropathy: what are the risks? Diabetes & metabolism, 1998. 24: p. 66-72.
  323. Vinik, A.I., et al., Neurovascular function and sudorimetry in health and disease. Current diabetes reports, 2013. 13(4): p. 517-532.
  324. Vinik, A.I., R. Maser, and A. Nakave, Diabetic cardiovascular autonomic nerve dysfunction. US Endocr Dis, 2007. 2: p. 2-9.
  325. Karamitsos, D., et al., The natural history of recently diagnosed autonomic neuropathy over a period of 2 years. Diabetes research and clinical practice, 1998. 42(1): p. 55-63.
  326. Ziegler, D. and F.A. Gries, α-Lipoic acid in the treatment of diabetic peripheral and cardiac autonomic neuropathy. Diabetes, 1997. 46(Supplement 2): p. S62-S66.
  327. Athyros, V., et al., Long-term effect of converting enzyme inhibition on circadian sympathetic and parasympathetic modulation in patients with diabetic autonomic neuropathy. Acta cardiologica, 1998. 53(4): p. 201-209.
  328. Gæde, P., et al., Intensified multifactorial intervention in patients with type 2 diabetes mellitus and microalbuminuria: the Steno type 2 randomised study. The Lancet, 1999. 353(9153): p. 617-622.
  329. Malmberg, K., et al., Glycometabolic state at admission: important risk marker of mortality in conventionally treated patients with diabetes mellitus and acute myocardial infarction: long-term results from the Diabetes and Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) study. Circulation, 1999. 99(20): p. 2626-2632.
  330. Kendall, D.M., et al., Pancreas transplantation restores epinephrine response and symptom recognition during hypoglycemia in patients with long-standing type I diabetes and autonomic neuropathy. Diabetes, 1997. 46(2): p. 249-257.
  331. Burger, A.J., et al., Effect of glycemic control on heart rate variability in type I diabetic patients with cardiac autonomic neuropathy. The American journal of cardiology, 1999. 84(6): p. 687-691.
  332. Laederach-Hofmann, K., P. Weidmann, and P. Ferrari, Hypovolemia contributes to the pathogenesis of orthostatic hypotension in patients with diabetes mellitus. The American journal of medicine, 1999. 106(1): p. 50-58.
  333. Denq, J.-C., et al., Efficacy of compression of different capacitance beds in the amelioration of orthostatic hypotension. Clinical Autonomic Research, 1997. 7(6): p. 321-326.
  334. Annese, V., et al., Gastrointestinal motor dysfunction, symptoms, and neuropathy in noninsulin-dependent (type 2) diabetes mellitus. Journal of clinical gastroenterology, 1999. 29(2): p. 171-177.
  335. Melga, P., et al., Chronic administration of levosulpiride and glycemic control in IDDM patients with gastroparesis. Diabetes Care, 1997. 20(1): p. 55-58.
  336. Stacher, G., et al., Cisapride versus placebo for 8 weeks on glycemic control and gastric emptying in insulin-dependent diabetes: a double blind cross-over trial. The Journal of Clinical Endocrinology & Metabolism, 1999. 84(7): p. 2357-2362.
  337. Barone, J.A., Domperidone: a peripherally acting dopamine2-receptor antagonist. Annals of Pharmacotherapy, 1999. 33(4): p. 429-440.
  338. Silvers, D., et al., Domperidone in the management of symptoms of diabetic gastroparesis: efficacy, tolerability, and quality-of-life outcomes in a multicenter controlled trial. Clinical therapeutics, 1998. 20(3): p. 438-453.
  339. Erbas, T., et al., Comparison of metoclopramide and erythromycin in the treatment of diabetic gastroparesis. Diabetes Care, 1993. 16(11): p. 1511-1514.
  340. Camilleri, M. and A. Acosta, Emerging treatments in Neurogastroenterology: relamorelin: a novel gastrocolokinetic synthetic ghrelin agonist. Neurogastroenterology & Motility, 2015. 27(3): p. 324-332.
  341. Alam, U., O. Asghar, and R.A. Malik, Diabetic gastroparesis: Therapeutic options. Diabetes Therapy, 2010. 1(1): p. 32-43.
  342. Abell, T., et al., Treatment of gastroparesis: a multidisciplinary clinical review: The American Motility Society Task Force on Gastroparesis (members in alphabetical order). Neurogastroenterology & Motility, 2006. 18(4): p. 263-283.
  343. Vinik, A. and D. Richardson, Erectile dysfunction in diabetes: pills for penile failure. Clinical Diabetes, 1998. 16(3): p. 108-109.
  344. Vinik, A. and D. Richardson, Erectile dysfunction in diabetes. Clinical Diabetes, 1996. 14(5): p. 111-121.
  345. Rendell, M.S., et al., Sildenafil for treatment of erectile dysfunction in men with diabetes: a randomized controlled trial. Jama, 1999. 281(5): p. 421-426.
  346. Goldstein, I., et al., Vardenafil, a new phosphodiesterase type 5 inhibitor, in the treatment of erectile dysfunction in men with diabetes: a multicenter double-blind placebo-controlled fixed-dose study. Diabetes care, 2003. 26(3): p. 777-783.
  347. Enzlin, P., et al., Diabetes mellitus and female sexuality: a review of 25 years’ research. Diabetic Medicine, 1998. 15(10): p. 809-815.
  348. Shaw, J., et al., A randomised controlled trial of topical glycopyrrolate, the first specific treatment for diabetic gustatory sweating. Diabetologia, 1997. 40(3): p. 299-301.
  349. Edick, C.M., Oral glycopyrrolate for the treatment of diabetic gustatory sweating. Annals of Pharmacotherapy, 2005. 39(10): p. 1760-1760.
  350. Meyer, C., et al., IMPROVED GLUCOSE COUNTERREGULATION AND AUTONOMIC SYMPTOMS AFTER INTRAPORTAL ISLET TRANSPLANTS ALONE IN PATIENTS WITH LONG-STANDING TYPE I DIABETES MELLITUS1. Transplantation, 1998. 66(2): p. 233-240.