Archives

Benign Breast Disease in Women

ABSTRACT

Benign breast disease in women is a very common finding and results in a diagnosis in approximately one million women annually in the United States (1). An understanding of the hormonal and growth factor control of breast development and function is key to the rational and systematic evaluation and treatment of patients. A firm understanding of benign breast disease is important since sequential steps are necessary to distinguish lesions which impart a high risk of subsequent breast cancer from those which do not. This chapter will review the physiology of breast function, provide histologic examples of common lesions, and detail practical approaches to evaluation and treatment.  For complete coverage of all related areas of Endocrinology, please see our online FREE web-book, www.endotext.org.

 

BREAST PHYSIOLOGY IN WOMEN:

Hormones and growth factors act upon stromal and epithelial cells to regulate mammary gland development, maturation and differentiation (2). Broadly summarized, estrogen mediates development of ductal tissue; progesterone facilitates ductal branching and lobulo-alveolar development; and prolactin regulates milk protein production. At puberty, estradiol and progesterone levels increase to initiate breast development. A complex tree-like structure results and comprises 5 to 10 primary milk ducts originating at the nipple, 20 to 40 segmental ducts, and 10 to 100 sub-segmental ducts ending in glandular units called terminal duct lobular units (TDLUs) (3). During the menstrual cycle the increments in estrogen and progesterone stimulate cell proliferation during the luteal phase (Figure1). Cycle dependent apoptosis balances proliferation (4). In response to enhanced proliferation, the breast can increase by as much as 15% in size during the luteal phase.

Anatomic and histologic structures of the breast undergo substantial change during the period from early adolescence to menopause (5). The normal histologic appearance represents a spectrum ranging from a predominance of ducts, lobules, and intra- and inter-lobular stroma to patterns with a predominance of fibrous change and cyst formation, a process formerly called fibrocystic disease (Figure 2). The term “fibrocystic changes” is now preferred since up to 50 to 60 percent of normal women may have this pattern histologically (6). This new term implies that women with lumpy breasts or non-discrete nodules do not have breast disease. Importantly, fibrocystic changes detected clinically incur no increased risk of breast cancer.

 

Specific changes in the breast, relating to stromal, ductal and glandular tissue occur as a function of age. During the early reproductive years, stromal hyperplasia may occur and produces juvenile breast hypertrophy (7) or rarely, the more significant problems of unilateral or bilateral macromastia (enlargement of breast tissue beyond what is considered normal) (8). Changes in glandular and ductal tissue occur uncommonly. In the middle reproductive years, glandular breast tissue continues to undergo changes in response to cyclic increments in plasma levels of estradiol and progesterone and, if substantial, is called adenosis. Ductal changes remain uncommon while stromal hyperplasia may occur resulting in areas of ill-defined fullness (“lumpy-bumpy” consistency) on physical exam or in firm areas requiring biopsy.

 

 

Figure 1. Influence of the cycle phase on breast total labeling index (TLI) of women less than 34 years of age according to whether cycles were natural or regulated by oral contraceptives (OC). (Reprinted with permission from Going JJ, Anderson TJ, Battersby et al. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles (Am. J Pathol. 130:193-204, 1988).Figure 1. Influence of the cycle phase on breast total labeling index (TLI) of women less than 34 years of age according to whether cycles were natural or regulated by oral contraceptives (OC). (Reprinted with permission from Going JJ, Anderson TJ, Battersby et al. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles (Am. J Pathol. 130:193-204, 1988).

Figure 2 . Simplified anatomy of the female breast illustrating the major structural components of the breast, the anatomic location of various lesions, and the histology of those lesions and corresponding sites of origin of potential lesions.

Figure 2 . Simplified anatomy of the female breast illustrating the major structural components of the breast, the anatomic location of various lesions, and the histology of those lesions and corresponding sites of origin of potential lesions.

In the late reproductive period, glandular tissue may become hyperplastic with sclerosing adenosis or lobular hyperplasia. The hyperplastic glandular lesions may progress to palpable or mammographically detectable abnormalities requiring biopsy. Ductal tissue also may undergo hyperplastic change with an increase in number of ductal cells but without alterations in their appearance. This change, when associated with a 2% or greater prevalence of Ki67 positive cells, is associated with an approximately two fold increase of subsequent breast cancer (9). Further proliferative changes result in lobules approaching 100µ in diameter and called hyperplastic elongated lobular units (HELUs). With progression of the HELUs, atypical ductal or lobular hyperplasia, or ductal carcinoma in situ (DCIS) may ensue. The linear pathway illustrated in Figure 3 depicts this progression but, while considered by some to be overly simplistic, provides a useful framework. This orderly progression depends upon the number of acquired genetic mutations accumulated by clonal cells in the breast. This originally was suggested by loss of heterozygosity studies. Recent specific genetic data suggests that an average of 11 “driver” mutations and 100 bystander mutations are present in established invasive breast cancer (10). Based on the number of mutations required for cancer, current opinion suggests that breast cancer is a process that takes many years to develop with the age of menarche as the earliest factor influencing this process. The progression from the earliest neoplastic changes to invasive breast cancer is considered to take a median of 16 years with a range of 1 to 30 years based on doubling time. Careful assessment of serial mammograms allows an estimation of doubling times which range from 25 to more than 700 days (Figure 4).

Figure 3 Model of the linear progresion from normal, to hyperplastic enlogated lobular units ( HELU), to atypical ductal hyperplasia (ADH), to ductal carcinoma in situ ( DCIS), to occult invasive breast cancer ( IBC), to IBC that has exceeded detection threshold and can be diagnosed by mammogram. Kinetic modeling data suggest that it takes 10 to 20 years for the progression from atypical hyperplasia to a clinically detectable tumor

Figure 3 Model of the linear progresion from normal, to hyperplastic enlogated lobular units ( HELU), to atypical ductal hyperplasia (ADH), to ductal carcinoma in situ ( DCIS), to occult invasive breast cancer ( IBC), to IBC that has exceeded detection threshold and can be diagnosed by mammogram. Kinetic modeling data suggest that it takes 10 to 20 years for the progression from atypical hyperplasia to a clinically detectable tumor

Figure 4. Cumulative frequency distribution of the doubling times of occult breast cancer obtained by serial mammograms. Figure reproduced with the permision of the author and publisher.

Figure 4. Cumulative frequency distribution of the doubling times of occult breast cancer obtained by serial mammograms. Figure reproduced with the permision of the author and publisher.

After the onset of menopause, glandular tissue undergoes involution and stroma and fatty tissue (i.e. approximately 97%) replace glandular elements (12).  The degree of involution is inversely related to the risk of subsequent breast cancer. When comparing the lowest with the highest quartile of involution, the relative risk of subsequent breast cancer is more than 2 fold increased I (OR 2.44 , 95% confidence interval (0.96-6.19) (1).  With aging, the degree of involution increases (1), which contributes substantially to the decrease in breast density. On histologic examination, lesions appearing dense on mammography contain a larger than normal proportion of stromal and glandular tissue.

SPECIFIC BENIGN BREAST LESIONS:

A wide variety of benign breast lesions have been described and the histologic appearance fully characterized. On a practical basis, these can be subdivided into those associated with no substantial increased risk of breast cancer ( i.e. < 1.49%), those with an increase of 1.5-2% and those with a >2% increase (Table 1) (13) .

Table 1. Relative risk of breast cancer imparted by specific benign breast lesions (Data from Reference 13)

No Increased risk:

The more common lesions Included in this category are fibrocystic changes, periductal fibrosis, hamartomas, lipomas, phylloides tumors, and neurofibromas. Another lesion is duct ectasia (Figure 5), characterized by distention of subareolar ducts and presence within them of yellowish-orange material. Penetration of the duct wall by this material may produce acute inflammatory changes in the surrounding tissues. Histologically, crystalline oval and round structures thought to be lipid in origin are present. Resolution may occur but residual periductal fibrosis and nodule formation often persist. This lesion is thought to be more frequent in cigarette smokers and represents a more chronic and extensive inflammatory process. A less common lesion is diabetic mastopathy which consists of localized or diffuse areas of fibrosis occurring in patients with diabetes (14).

Figure 5. Histological appearance of duct ectasia showing the characteristic crystalline formation of the intraluminal contents.

Figure 5. Histological appearance of duct ectasia showing the characteristic crystalline formation of the intraluminal contents.

Abnormalities of ductal secretion may result in discharge of clear, cloudy, blue, green or black aqueous material. Stromal hyperplasia can result in nipple retraction or in palpable lesions requiring biopsy to distinguish from breast carcinoma. Histologically, the tissue may contain fibroblasts nearly exclusively or predominantly fibroblasts with admixture of glandular epithelium. Some degree of stromal hyperplasia may occur in 50-60% of normal women, particularly those in their middle and late reproductive periods. Traumatic lesions (hematomas and fat necrosis) and inflammatory conditions (granulomas and mastitis) are also not associated with an increased breast cancers (15).

 

1.5 To 2.0 Fold Increase In Relative Risk:

 

Fibroadenomas:
During the early reproductive period, glandular components of the breast may respond to cyclic hormonal stimuli in an exaggerated fashion with the development of single fibroadenomas. These consist of lobular units which grow to larger than normal size and contain both epithelial and stromal elements. Fibroadenomas range in size from slightly larger than a normal single lobular unit to larger, more discrete palpable lesions. When exceeding 5 cm, these are called giant fibroadenomas which are seen primarily at puberty or during pregnancy. The incidence of fibroadenomas peaks at age 20-24 (Figure 6). The prevalence on physical examination in young women is 2% in young women but 15-23% when evaluated prospectively at autopsy (16). When complex and containing cysts >3mm in diameter, sclerosing adenosis, epithelial calcification or papillary changes, these lesions are associated with an increased risk of breast cancer (17) (Table 1).

Figure 6. Incidence of fibrocystic breast changes and fibroadenoma by age group. (Reprinted with permission of the publisher and authors from Goehring C, Morabia A. Epidemiology of Bengin Breast Disease, with special attention to histologic types Epidemiologic Reviews 19:310-327, 1997) (16). Note that the term “fibrocystic disease” is used in the legend but this term has now been changed to “fibrocystic changes”.

Figure 6. Incidence of fibrocystic breast changes and fibroadenoma by age group. (Reprinted with permission of the publisher and authors from Goehring C, Morabia A. Epidemiology of Bengin Breast Disease, with special attention to histologic types Epidemiologic Reviews 19:310-327, 1997) (16). Note that the term “fibrocystic disease” is used in the legend but this term has now been changed to “fibrocystic changes”.

Other lesions:

Hyperplasia without atypia, papillomas, papillomatosis, radial scar, blunt duct adenosis, and sclerosing adenosis are also associated with an increased risk.

 

Greater Than A Two-Fold Risk:

 

Atypical hyperplasia

This lesion can be subdivided into atypical ductal hyperplasia (figure 7) and atypical lobular hyperplasia but both impart a similar increase in relative risk of breast cancer of 3 to 4 fold (18) (Table 1). In the first ten years after diagnosis of AH, the breast tumors occur predominantly in the same breast but subsequently, occur with equal frequency in the contralateral as in the ipsilateral breast (19). These observations suggest the AH is both a precursor lesion for breast cancer and a marker of an underlying predisposing condition termed a “field defect” or alternatively a “mutator phenotype (15) When the amount of hyperplastic tissue increases according to specific criteria (20) the lesions are no longer called benign but are classified as ductal carcinoma in situ. Recent emphasis has been directed toward identification of molecular genetic markers which could predict which patients with AH are at increased risk of developing breast cancer. Over-expression of HER-2/neu is one such marker (21). The presence of aberrant p53, p21, interleukin 6, and TNF alpha and aneuploidly on flow cytometery have also been reported as markers of increased risk (22).

Figure 7. Photomicrograph of atypical ductal hyperplasia histology reprinted from Hartmann LC et al (23).

Figure 7. Photomicrograph of atypical ductal hyperplasia histology reprinted from Hartmann LC et al (23).

Recent data from the Mayo Clinic report the absolute rates of breast cancer during prolonged 20+ year follow-up and the important role of multifocality (23;24). As shown in Figure 8, the risk approximates 24% with one lesion, 36% with two and 47% with 3 or more lesions. The increased risk with multifocality has been confirmed in an independent cohort follow at  Vanderbilt (18;19). Although previous studies indicated that family history added to this risk (18), the updated data suggest that neither family history or other risk factors independently contribute to breast cancer risk (23). It is important to emphasize the AH appears to impart a risk of breast cancer which is lower but may approach that from BrCa1 and 2 mutations. As noted above, the presence of atypical hyperplasia imparts a risk over time of both ipsilateral and contralateral breast cancer (Figure 9).

Figure 8. Incidence of breast cancer in cohort of patients with a histologic diagnosis of atypical hyperplasia. Top panel: mean (solid line) and 95% confidence limits (dashed lines). Bottom panel: Incidence in patients with 1, 2, or 3 or more independent lesions. Reprinted with the permission of the authors and publisher from Hartmann LC et al (23).

Figure 8. Incidence of breast cancer in cohort of patients with a histologic diagnosis of atypical hyperplasia. Top panel: mean (solid line) and 95% confidence limits (dashed lines). Bottom panel: Incidence in patients with 1, 2, or 3 or more independent lesions. Reprinted with the permission of the authors and publisher from Hartmann LC et al (23).

Figure 9. Ipsilateral versus contralateral distribution of breast cancers diagnosed over time in patients initially diagnosed with atypical hyperplasia. Reproduced with the permission of the authors and publishers from Hartmann LC et al (24).

Figure 9. Ipsilateral versus contralateral distribution of breast cancers diagnosed over time in patients initially diagnosed with atypical hyperplasia. Reproduced with the permission of the authors and publishers from Hartmann LC et al (24).

Lobular Carcinoma in situ:

Even though called lobular carcinoma in situ, this lesion is not generally considered to have reached the neoplastic stage but is analogous to atypical hyperplasia. This lesion imparts approximately a 10 fold   increase in relative risk of breast cancer developing over time. The subsequent breast cancer can occur in the same or contralateral breast; accordingly, this lesion is thought to represent a filed defect or mutator phenotype which increases breast cancer risk but the lesion itself is not a precursor of breast cancer.

 

 

Mammographic Density And Breast Cancer Risk:

The percent breast density on mammography correlates with breast cancer risk as shown on Figure 10. When comparing the lowest density category with highest, the relative risk is increased by 5.3 fold. Recent studies examined the histologic composition of dense and non-dense breast tissue. When dense lesions are biopsied and compared to areas of low density, they are found to contain a higher proportion of stroma and glandular tissue and lesser amount of fat (26). Notably, dense lesions contain more of the enzyme aromatase, when quantitated by an immunologic histologic score after staining with an aromatase monoclonal antibody (27). These findings are likely associated with a higher local production of estradiol and could explain the higher incidence of breast cancer with both of these lesions. Taking these observations together, determination of breast density assessed by Bi-RADS criteria or quantitated with a computer assisted method of determining breast density, provides the most powerful means of predicting risk of breast cancer over time (Figure 10).

Figure 10. The percentage of breast tissue which is dense on a mammogram is determined by computer assisted analysis and classified as none, <10%, 10-25%, 25-50%, 50-75%, >75%.The relative risk of breast cancer increases with each step of increased breast density. Adapted from that data of Boyd et al (25).

Figure 10. The percentage of breast tissue which is dense on a mammogram is determined by computer assisted analysis and classified as none, <10%, 10-25%, 25-50%, 50-75%, >75%.The relative risk of breast cancer increases with each step of increased breast density. Adapted from that data of Boyd et al (25).

ETIOLOGY OF BENIGN BREAST DISORDERS

 

Clinical observations in women receiving estrogens and anti-estrogens suggest that hormonal events play a role in the etiology of benign breast lesions. In post-menopausal women receiving estrogens ± progestins for >8 years, the prevalence of benign breast lesions increased by 1.7 fold (95 percent CI 1.06-2.72) (28). In the Women’s Health Initiative study (WHI), the use of estrogen plus progestin was associated with a 74% increase in the risk of benign proliferative breast disease [hazard ratio, 1.74; 95% confidence interval (CI), 1.35-2.25] (29). The anti-estrogen, tamoxifen, when used for breast cancer prevention, was associated with a 28 percent (RR 0.72, 95 percent Confidence Intervals 0.65-0.79) reduction in prevalence of benign breast lesions, including adenosis, cysts, duct ectasia, and hyperplasia (30).

 

Underlying and acquired genetic changes are also associated with benign breast lesions. Loss of heterozygosity (LOH), a finding caused by deletions of small segments of DNA (31;32) is commonly found in benign breast lesions. Women frequently have multi-focal lesions, each of which exhibit loss of heterozygosity (LOH) of differing regions of DNA. Women with BRCA1/2 mutations are found to have a high frequency of multiple benign or malignant breast lesions when bilateral mastectomy specimens are meticulously examined (33). These findings support the current theory of an underlying predisposition to mutations in some patients as the cause of multiple breast lesions (34). In the past, this phenomenon was termed a “field effect” and more recently, a “mutator phenotype” (34).

 

CLINICAL MANIFESTATIONS:

Clinical presentations of benign breast disease are divided into those with pain, lumps, or breast discharge (Table 2).

Breast Pain:

Cyclic breast pain

 

Usually occurs during the late luteal phase of the menstrual cycle, in association with the premenstrual syndrome or independently (35-40), and resolves at the onset of menses (35;36;38). A recent study in 1171 healthy American women indicated that 11% experience moderate to severe cyclic breast pain and 58%, mild discomfort (39). Breast pain interfered with usual sexual activity in 48% and with physical (37%), social (12%), and school (8%) activity in others. A role for caffeine, iodine deficiency, alterations in fatty acid levels in the breast, fat intake in the diet, and psychological factors in the etiology of breast pain remain unproven. Non-cyclic breast pain is unrelated to the menstrual cycle. Detection of focal tenderness is helpful diagnostically and suggests a tender cyst, rupture through the wall of an ectatic duct, or a particularly tender area of breast nodularity. Acute enlargement of cysts and periductal mastitis may cause severe, localized pain of sudden onset.

 

Non-breast pain:

When arising from the chest wall, pain may be mistakenly attributed to the breast. Pain localized to a limited area and characterized as burning or knife like in nature suggests this possibility. Several distinct subtypes can be distinguished including localized or diffuse lateral chest wall pain, radicular pain from cervical arthritis and Tietze’s syndrome or costochondritis. The method to distinguish between breast and non-breast pain by careful physical examination is illustrated on Figure 11, panels A-D.

Figure 11. A. Focal chest wall pain—lateral. The patient is turned 90º on her side so that breast tissue is no longer under the area of palpation. The index finger elicits a focal area of pain. B. Focal chest wall pain over costochondral junctions anteriorly. C. Diffuse lateral chest wall pain. With the patient turned over 90 degrees on her side, pain is elicited over a wider area of the chest wall. D. Verification that squeezing breast tissue does not elicit pain ensures that the pain is not related to the breast but represents chest wall pain.

Figure 11. A. Focal chest wall pain—lateral. The patient is turned 90º on her side so that breast tissue is no longer under the area of palpation. The index finger elicits a focal area of pain. B. Focal chest wall pain over costochondral junctions anteriorly. C. Diffuse lateral chest wall pain. With the patient turned over 90 degrees on her side, pain is elicited over a wider area of the chest wall. D. Verification that squeezing breast tissue does not elicit pain ensures that the pain is not related to the breast but represents chest wall pain.

Breast Nodule:

Proliferation of ductal or lobular tissue causes histologic changes that are manifested by the presence of palpable lumps or nodules. Patients often present with the finding of a new breast nodule on self-exam or are found to have a lump by their health care provider. Ninety percent of these new nodules in premenopausal women are benign and usually represent fibroadenomas in the early reproductive period (16). In the middle reproductive period, focal areas of fibrosis, hyperplasia, or cyst formation are more likely. In the later reproductive period, hyperplasia, cysts, and carcinoma in situ are more common.  Some lesions present with symptoms suggesting the cause. With duct ectasia, penetration of the duct wall by lipid material may be associated with a nodule exhibiting acute redness, and causing pain, and fever; after resolution, a subareolar nodule persists.

Other specific lesions present as lumps. These include multiple papillomas, sclerosing adenosis, and radial scars. Multiple papillomas may present as breast lumps, nodules on ultrasound, or may be the cause of bloody nipple discharge and can be seen on ductography. Sclerosing adenosis is a lobular lesion with increased fibrous tissue and interspersed glandular cells. It can present as a mass or a suspicious finding on mammograms. Radial scars are a pathologic diagnosis, usually diagnosed following mammography or palpation and then biopsy. Radial scars are characterized microscopically by a fibroelastic core with radiating ducts and lobules and impart a minimally increased risk of breast cancer similar to that of proliferative changes without atypia (41).

Nipple discharge:

6.8 percent of referrals to physicians for breast concerns result from this symptom. Although particularly distressing to the patient, only 5 percent are found to have serious underlying pathology. Age is an important factor with respect to risk of malignancy (42). In one series, 3 percent of women younger than age 40, 10 percent between 40 and 60 and 32 percent >60 with nipple discharge as their only symptom were found to have a malignancy.

 

A careful history characterizes breast discharge as either spontaneous or expressible. On examination, one can detect by careful inspection whether the discharge emanates from a single or multiple ducts. Nipple discharge can be divided into physiologic and pathologic types. Characteristics of physiologic discharge include non-spontaneous, multiple duct, bilateral, and non-bloody. Pathologic discharge is characterized as spontaneous, serous or bloody, usually unilateral and usually single duct. Reassuring characteristics are that it must be expressed; is green yellow, brown or milky; that it is bilateral and involves multiple ducts. Spontaneous discharge, whether serous or bloody, requires careful evaluation. A hemoccult card or urine dipstick can be used to test for occult blood if the discharge is spontaneous, unilateral, and from one duct. Cytologic examination is not recommended. Milky discharge (galactorrhea) should be evaluated with measurement of a serum prolactin level. If the discharge is physiologic and the patient is under 35, only reassurance is necessary. Screening mammogram is recommended for patients over 35 with physiologic discharge. Pathologic discharge requires diagnostic mammogram, galactography (Figure 12) (43), and referral to a surgeon.

Figure 12. Galactogram illustrating space occupying lesion. A catheter is inserted into the duct from which the bloody discharge emerges. Contrast material is then injected through the catheter. The various branches of the duct are outlined.

Figure 12. Galactogram illustrating space occupying lesion. A catheter is inserted into the duct from which the bloody discharge emerges. Contrast material is then injected through the catheter. The various branches of the duct are outlined.

The presence of crusting, scaling, and flaking of the nipple could be a manifestation of Paget’s disease of breast and underlying cancer or of dermatologic problems. The approach is to obtain a history of other dermatologic problems, or a history of change in soap or clothing. If absent, a diagnostic mammogram should be obtained if the patient is over 35. In a large recent series of 1251 patients with nipple discharge, 433 had unilateral discharge and 194 had no breast lump in association with this symptom. Of these, the lesions found included solitary papilloma (n=49), minimal breast cancer (n=20), fibrocystic disease(n=11), papillomatosis (n=7), lobular cancer (n=5) and duct ectasia (n=2)(42) (30A). For women with bloody discharge from a single duct, galactography is warranted. Filling defects can be due to intraductal papilloma, intraductal carcinoma, papillomatosis, debris, or air bubbles.

 

 

APPROACH TO THE PRACTICAL MANAGEMENT OF BENIGN BREAST DISEASE

 

A detailed history and physical exam systematically evaluates the entire breast and chest wall and focuses on areas involving the patient’s symptoms (Table 3). Diagnostic studies may then be ordered. For lumps, “The Triple Test” is recommended which includes palpation, imaging and percutaneous biopsy (either core or fine needle aspirate <FNA>). Mammography, often in conjunction with ultrasound examination (44-47) is required for evaluation of discrete palpable lesions in women over 35 whereas ultrasound provides an optional substitute in younger women (43). Round dense lesions on mammography often represent cysts which require only ultrasonography to distinguish them from solid lesions. Complex cysts containing both fluid and solid matter require biopsy.  For solid lesions, radiographically or ultrasonically directed core biopsy provides highly discriminative information regarding the presence or absence of malignancy. Core biopsy utilizes a large cutting needle deployed with a spring loaded, automated biopsy instrument and obtains tissue suitable for histologic analysis familiar to most pathologists. FNA frequently yields sufficient cellular material to allow adequate cytologic evaluation but requires an experienced cytopathologist. However, the amount of material obtained is insufficient to render a diagnosis in 35-47% of non-palpable lesions (48) and core biopsy is recommended. The exact role of MRI in evaluating breast lesions is currently being determined (49). Galactography (ductography) is useful for detection of focal lesions in a single duct. Cytology of nipple discharge is of limited value with the sensitivity of detecting malignancy only 35 to 47 percent (50).

Diagnostic Procedures

Ideally a team including a radiologist experienced in mammography, ultrasound, MRI and core needle biopsy as well as an internist, gynecologist, or surgeon with expertise in breast diseases should be involved in the evaluation of patients with breast disorders. MRI mammography, ductography, or ultrasound may be utilized (Figures 12 and 13). Information to be obtained by a focused history and physical examination are outlined on Table 3. The method of documenting whether breast pain is chest wall related is illustrated on Figure 11 A-D. Imaging has become an integral part of the management of benign breast disorders.

 

Imaging studies:

Mammography is useful for evaluation of palpable lesions, particularly in those over 35. Digital mammography is preferred because of its ability to penetrate through dense breast tissue which is commonly found in younger women. Ultrasound is often used as initial evaluation of a palpable mass in women under age 35. If a simple cyst is present, no further evaluation is necessary (Figure 13). If not, mammography may also be necessary to fully evaluate the lump. If the mass has findings suggestive of a fibroadenoma by ultrasound and mammography, short term follow-up and re-imaging can be considered (usually performed in 6 months). Experts are divided as to the necessity to biopsy all fibroadenomas. MRI is more sensitive than digital mammography but false positives are more common (49). Routine yearly MRI is now recommended for women whose lifetime risk of breast cancer is > 20%. As an illustration of its sensitivity, 3% of the contralateral breasts in women with diagnosed breast cancer are found to have a second lesion in the opposite breast when examined by MRI (49).

Figure 13 Upper panel illustrates by ultrasound a non-dense black area representing cyst fluid. The lower panel is the corresponding area on mammogram showing a dense area. With the combination of mammogram and ultrasound, the lesion can be shown to be a cyst.

Figure 13 Upper panel illustrates by ultrasound a non-dense black area representing cyst fluid. The lower panel is the corresponding area on mammogram showing a dense area. With the combination of mammogram and ultrasound, the lesion can be shown to be a cyst.

Findings On Imaging Studies

 

Fibrocystic change typically presents on mammogram as round or oval, well defined masses that can be subsequently shown to represent cysts on ultrasound (Figure 13). Diffusely scattered dystrophic calcifications may also be found on the mammogram. Consequently, the goal of mammographic evaluation is to provide reassurance to the patient and physician that the risk of neoplasm is low. Aspiration of cysts is usually necessary only in a those cases where the mass does not fulfill all criteria for a simple cyst or if the cyst is painful. Biopsy may be necessary to confirm the benign nature of calcifications, particularly if clustered, linear or variously shaped.

For round masses or round calcifications on a first mammogram, the risk of cancer is less than 2%, and repeat mammography in 6 months is recommended. These lesions are termed “probably benign” using the lexicon terminology required by the Mammography Quality Standards Act (in the USA). If the risk is believed to be greater, core biopsy is recommended. Stereotactically directed core biopsy is ideal for evaluation of calcifications and provides highly discriminative information regarding the presence or absence of malignancy. If this technique is not available, insertion of a wire into the lesion radiographically followed by surgical excision or mere removal of a palpable lesion is warranted.

 

CLINICAL GUIDELINES FOR EVALUATION OF NODULES AND DISCHARGE

 

Careful examination distinguishes solitary, discrete, dominant, persistent masses from vague nodularity and thickening. Practice Guidelines of the Society of Surgical Oncology (51) recommend the following evaluation: In women less than age 35, all dominant discrete palpable lesions require referral to a surgeon. If vague nodularity, thickening or asymmetrical nodularity is present, the examination is repeated at midcycle after one or two menstrual cycles. If the abnormality resolves, the patient is reassured and if not, referred to a surgeon. Breast imaging may be appropriate. In women > age 35 with a dominant mass, a diagnostic mammogram (and frequently a sonogram) (44-47) is obtained and the patient referred to a surgeon. With vague nodularity or thickening, one obtains a mammogram with repeat physical exam at mid-cyle 1 to 2 months later and refers to surgeon if the abnormality persists. Post-menopausal women are referred for surgical consultation after a mammogram. For gross cysts (i.e. >4 cm), the guidelines suggest needle aspiration with repeat imaging within six months. If the aspirated fluid does not contain blood, the fluid is discarded without further histologic analysis unless the cyst contains solid components (i.e. complex cyst). If the fluid contains blood or if the cyst is complex, the, fluid is sent for cytology and consultation from a surgeon requested. With persistent refilling of the same cyst after aspiration, surgical consultation is warranted.

 

Usual practice requires “the triple test” with palpation, mammography (often in conjunction with ultrasonography) and biopsy in women over age 35 with dominant masses. When mammography is negative but a dominant mass is present, biopsy is required to rule out malignancy since lobular carcinoma may not be seen on mammograms. In those younger, mammography may be omitted if ultrasound and biopsy yield definitive information. Many experts omit biopsy in younger women with lesions characteristic of fibroadenoma on ultrasound and elect to follow carefully with serial ultrasounds at six monthly intervals for two years and yearly thereafter. Since careful studies have shown that a lesion appearing benign on mammography and ultrasound is benign >99 percent of the time, clinical judgment may allow follow-up without biopsy in experienced hands (44-47). However, other experienced surgeons disagree and believe that all fibroadenomas require diagnostic core biopsy or FNA and especially in BRCA mutation carriers in whom medullary cancer may be found. Biopsy confirmation of a fibroadenoma eliminates the need for serial ultrasounds. For those with a diagnosis of ADH (Atypical Ductal Hyperplasia) on FNA or core biopsy, excisional biopsy is then required since more complete resection often changes the diagnosis to DCIS.

 

Breast discharge is evaluated according to the algorithm illustrated below on Figure 14. Careful attention to several factors are necessary including determination whether the discharge arises from one duct or multiple ducts, is bloody, or is milky.

Figure 14. Algorithm to assess breast discharge.

Figure 14. Algorithm to assess breast discharge.

TREATMENT OF BENIGN BREAST DISEASE IN WOMEN

 

The initial step in evaluating pain is to distinguish true breast pain from chest wall pain (Figure 11). Several well designed, randomized, controlled, double blind, cross over trials have validated the efficacy of medical therapy for cyclic mastalgia. Based upon these studies, we categorize therapies as definitely effective, definitely ineffective, possibly effective, and insufficiently studied. For classification as definitely effective, two or more randomized trials are required. For the category, possibly effective, one randomized trial must be positive in some respect but others may be negative. For the category definitely ineffective, prospective trials must be uniformly negative. For the category, insufficiently studied, only one randomized trial, either negative or positive is available. For full details and references see Table 162-1 in Endocrinology, Fourth Edition, LJ DeGroot and JL Jameson, editors, WB Saunders Company, Philadelphia publishers; Chapter 162 Benign Breast Disorders, RJ Santen page 2194)

 

Danazol, bromocriptine, and tamoxifen have been proven to be effective (52-55) (Figure 15). Linoleic acid in the form of evening primrose oil has been shown effective in two randomized trials but not in the third, the largest trial. Its role in treatment therefore remains uncertain (56-58) . Vitamin E is considered definitely ineffective and iodine and vaginal progesterone possibly effective. Medroxyprogesterone acetate, caffeine avoidance, and progesterone have not been sufficiently studied. Several other therapies have not been examined in randomized controlled trials but are likely to be beneficial since they are based upon physiologic principles. For example, precise fitting of a bra to provide support for pendulous breasts has been reported to relieve pain in observational studies. GnRH agonist analogues are used to lower LH, FSH, and estradiol levels and to create a temporary post-menopausal state (59;60). Onset of menopause is known to reduce the frequency of breast pain. This therapy is reserved for patients in whom all other measures fail and the pain is considered severe. Reduction of the dosage of estrogens in post-menopausal women or addition of an androgen to estrogen replacement therapy (e.g. Estrotestâ tablets) appears to be beneficial in reducing breast pain (personal observations of author).

Figure 15. Relative efficacy of agents to treat breast pain. These data are from the Breast Clinic in Cardiff, Wales and represent observational studies and not randomized, controlled efficacy trails.

Figure 15. Relative efficacy of agents to treat breast pain. These data are from the Breast Clinic in Cardiff, Wales and represent observational studies and not randomized, controlled efficacy trails.

Relative efficacy of effective therapies:

No large randomized, controlled studies have compared the relative efficacy of danazol, bromocriptine, evening primrose oil and tamoxifen. Figure 15 rank orders them according to efficacy based upon data from individual reports from the same clinic. Minimal data are available from clinical trials which involve direct head to head comparisons. It should be noted that overall responses to danazol, bromocriptine and evening primrose oil are lower in those with non-cyclic pain than those with cyclic pain. However, not all studies have carefully excluded patients with non-breast pain and therefore conclusions regarding non-cyclic pain should be considered tentative. The approach to non-breast pain is outlined in Figure 16.

Figure 16. Algorithm to treat non-breast pain

Figure 16. Algorithm to treat non-breast pain

WOMEN AT HIGH RISK FOR BREAST CANCER

 

A major consideration for women who present with breast problems is whether they have a higher than normal risk of developing breast cancer. Certain breast lesions such as fibrocystic changes are associated with no increased risk of subsequent breast cancer (Table I). A 1.5 to 2-fold greater risk of development of breast cancer over a 20 year period of follow-up occurs with proliferative lesions including ductal hyperplasia, lobular hyperplasia without atypia, sclerosing adenosis, diffuse papillomatosis and complex fibroadenomas. A recent report also suggested that radial scars increase relative risk by 1.8, a risk similar to that found in proliferative disease without atypia. It should be noted that the Gail model for assessing breast cancer risk, which is based predominantly on reproductive factors, underestimates the long term risk of breast cancer in women with benign breast disease. On the other hand, the five year prediction is more accurate (61).

 

The presence of dense breast tissue on mammography has also been reported to be a predictor of increased incidence of breast cancer (Figure 10).Two components of this finding must be considered: one, the presence of high breast density makes it more difficult to read mammograms and masks the sensitivity of finding a breast cancer initially but identifies it later and two, there is an increased risk of breast cancer associated with increased breast density. With long tem follow-up studies, masking is not the explanation for the increased breast cancer risk (62).

 

According to classic twin studies, heritability accounts for approximately 60% of the variation in breast density (63;64). Breast cancer risk is also increased in association with high plasma estradiol and testosterone levels in postmenopausal women (62;65) and 20 kg or more weight gain (66) in the pre-menopausal years.

 

Another risk factor is use of hormone replacement therapy. Current data from the Women’s Health Initiative suggests a 26% increase in relative risk of breast cancer with conjugated equine estrogen (CEE) when combined with medroxyprogesterone acetate when used for an average of 7.1 years. (67). This risk is probably increased further in women starting this therapy shortly after the menopause (i.e. a short gap between onset of menopause and start of menopausal hormone therapy)(68). Starting this therapy a long time after experiencing menopause (long gap) is associated with a lesser relative risk (68). The use of estrogen alone in the WHI was associated with a trend toward reduction of risk of breast cancer at five years and a statistically significant reduction in those adhering to therapy (68). In a follow up report, after 5 years of use and 8 additional follow-up years, the reported decrease of 23% was statistically significant (69) (manson). Available data suggest that the effects of menopausal hormone therapy in the WHI is a class effect and not related to the specific type of estrogen or progestin. One study, however, suggests that use of crystalline progesterone as the progestogen is associated with a lesser risk than use of medroxyprogesterone acetate (70).

 

To aid in assessing breast cancer risk, a questionnaire developed by Gail, utilizes answers to 7 questions to calculate the 5 year and lifetime risk of developing breast cancer (71). This model has recognized deficiencies in that it does not consider second degree relatives with breast cancer, proliferative lesions of breast other than ADH, alcohol intake, obesity, or birth control pill and menopausal hormone therapy (MHT) use. Nonetheless, the Gail model has been prospectively validated in over 6000 women followed for an average of 4.5 years. A more recently developed model, the Tyrer–Cuzick (IBIS II) model (72) incorporates second degree relatives, obesity and use of MHT into its risk calculations and provides more robust information than the Gail model.

 

BREAST CANCER PREVENTION

 

Patients with benign breast lesions imparting an increased risk of breast cancer can be offered tamoxifen (or raloxifene) as a prevention strategy. The risk of breast cancer is determined using the Gail or Tyrer-Cuzick model and the benefits versus risks of tamoxifen evaluated. Risk factors not included in the Gail or Claus models include degree of breast density, plasma free estradiol levels, bone density, weight gain after menopause, and waist-hip ratio (25;65;66;73). Current recommendations suggest that women with a five year risk of breast cancer of over 1.67 percent and no contraindications to tamoxifen should be informed about the possibility of taking tamoxifen for five years (74-77). A recent overview has shown a 38 percent reduction of the relative risk of breast cancer with tamoxifen but benefits may be offset by increased risks of thromboembolic phenomena, endometrial cancer, and maturation of cataracts (78). The Star trial addressed whether raloxifene might be preferable to tamoxifen and (79) demonstrated relative equivalence. However, of interest was the fact that tamoxifen prevented more non-invasive breast cancers than did raloxifene (79). More intensive and frequent screening with multi-modality imaging (i.e. digital or standard mammography plus ultrasound or MRI) may be required in high risk patients. Atypical hyperplasia lesions appear to be more amenable to breast cancer prevention as reviewed from non-head-to-head studies showing prevention ranging from 62 to 75% reduction in four separate randomized studies (23) when compared to a 38% reduction in women selected to be at high risk based on reproductive factors (78).

 

REFERENCE LIST

 

(1)   Figueroa JD, Pfeiffer RM, Brinton LA, Palakal MM, Degnim AC, Radisky D et al. Standardized measures of lobular involution and subsequent breast cancer risk among women with benign breast disease: a nested case-control study. Breast Cancer Research & Treatment 2016;159(1):163-172.

(2)   Russo J. Hormonal control of breast development. In: DeGroot LJ, Jameson JL, editors. Endocrinology. fourth edition ed. Philadelphia, PA: WB Saunders; 2001. p. 2181-2188.

(3)   Osborne MP. Breast anatomy and development. In: Harris JR, Osborne CK, Morrow M, Lippman ME, editors. Diseases of the Breast. Philadelphia: Lippincott Williams and Wilkins; 2000. p. 1-13.

(4)   Going JJ, Anderson TJ, Battersby S, MacIntyre CC. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles. American Journal of Pathology 1988;130(1):193-204.

(5)   Hughes LE, Mansel RE, Webster DJ. Aberrations of normal development and involution (ANDI): a new perspective on pathogenesis and nomenclature of benign breast disorders. Lancet 1987;2:1316-1319.

(6)   Love SM, Gelman RS, Silen W. Sounding board. Fibrocystic "disease" of the breast--a nondisease? New England Journal of Medicine 1982;307(16):1010-1014.

(7)   Pearlman MD, Griffin JL. Benign breast disease. [Review] [30 refs]. Obstetrics & Gynecology 2010;116(3):747-758.

(8)   Baker SB, Burkey BA, Thornton P, LaRossa D. Juvenile gigantomastia: presentation of four cases and review of the literature. [Review] [16 refs]. Annals of Plastic Surgery 525;46(5):517-525.

(9)   Huh SJ, Oh H, Peterson MA, Almendro V, Hu R, Bowden M et al. The Proliferative Activity of Mammary Epithelial Cells in Normal Tissue Predicts Breast Cancer Risk in Premenopausal Women. Cancer Research 2016;76(7):1926-1934.

(10)   Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ et al. The genomic landscapes of human breast and colorectal cancers.[see comment]. Science 2007;318(5853):1108-1113.

(11)   Bailey SL, Sigal BM, Plevritis SK. A simulation model investigating the impact of tumor volume doubling time and mammographic tumor detectability on screening outcomes in women aged 40-49 years. Journal of the National Cancer Institute 2010;102(16):1263-1271.

(12)   Radisky DC, Visscher DW, Frank RD, Vierkant RA, Winham S, Stallings-Mann M et al. Natural history of age-related lobular involution and impact on breast cancer risk. Breast Cancer Research & Treatment 2016;155(3):423-430.

(13)   Dyrstad SW, Yan Y, Fowler AM, Colditz GA. Breast cancer risk associated with benign breast disease: systematic review and meta-analysis. [Review]. Breast Cancer Research & Treatment 2015;149(3):569-575.

(14)   Kudva YC, Reynolds C, O'Brien T, Powell C, Oberg AL, Crotty TB. "Diabetic mastopathy," or sclerosing lymphocytic lobulitis, is strongly associated with type 1 diabetes. Diabetes Care 2002;25(1):121-126.

(15)   Santen RJ, Mansel R. Benign breast disorders. [Review] [89 refs]. New England Journal of Medicine 2005;353(3):275-285.

(16)   Goehring C, Morabia A. Epidemiology of benign breast disease, with special attention to histologic types. Epidemiologic Reviews 1997;19(2):310-327.

(17)   Nassar A, Visscher DW, Degnim AC, Frank RD, Vierkant RA, Frost M et al. Complex fibroadenoma and breast cancer risk: a Mayo Clinic Benign Breast Disease Cohort Study. Breast Cancer Research & Treatment 2015;153(2):397-405.

(18)   Hartmann LC, Sellers TA, Frost MH, Lingle WL, Degnim AC, Ghosh K et al. Benign breast disease and the risk of breast cancer.[see comment]. New England Journal of Medicine 2005;353(3):229-237.

(19)   Degnim AC, Visscher DW, Berman HK, Frost MH, Sellers TA, Vierkant RA et al. Stratification of breast cancer risk in women with atypia: a Mayo cohort study.[see comment]. Journal of Clinical Oncology 2007;25(19):2671-2677.

(20)   Page DL, Dupont WD, Rogers LW, Rados MS. Atypical hyperplastic lesions of the female breast. A long-term follow-up study. Cancer 1985;55(11):2698-2708.

(21)   Stark A, Hulka BS, Joens S, Novotny D, Thor AD, Wold LE et al. HER-2/neu amplification in benign breast disease and the risk of subsequent breast cancer. Journal of Clinical Oncology 2000;18(2):267-274.

(22)   Garcia-Tunon I, Ricote M, Ruiz A, Fraile B, Paniagua R, Royuela M. Role of tumor necrosis factor-alpha and its receptors in human benign breast lesions and tumors (in situ and infiltrative). Cancer Science 2006;97(10):1044-1049.

(23)   Hartmann LC, Degnim AC, Santen RJ, Dupont WD, Ghosh K. Atypical hyperplasia of the breast--risk assessment and management options. New England Journal of Medicine 2015;372(1):78-89.

(24)   Hartmann LC, Radisky DC, Frost MH, Santen RJ, Vierkant RA, Benetti LL et al. Understanding the premalignant potential of atypical hyperplasia through its natural history: a longitudinal cohort study. Cancer Prevention Research 2014;7(2):211-217.

(25)   Boyd NF, Lockwood GA, Byng JW, Tritchler DL, Yaffe MJ. Mammographic densities and breast cancer risk. Cancer Epidemiology, Biomarkers & Prevention 1998;7(12):1133-1144.

(26)   Ghosh K, Brandt KR, Reynolds C, Scott CG, Pankratz VS, Riehle DL et al. Tissue composition of mammographically dense and non-dense breast tissue. Breast Cancer Research & Treatment 2012;131(1):267-275.

(27)   Vachon CM, Sasano H, Ghosh K, Brandt KR, Watson DA, Reynolds C et al. Aromatase immunoreactivity is increased in mammographically dense regions of the breast. Breast Cancer Research & Treatment 2011;125(1):243-252.

(28)   Rohan TE, Miller AB. Hormone replacement therapy and risk of benign proliferative epithelial disorders of the breast. European Journal of Cancer Prevention 1999;8(2):123-130.

(29)   Rohan TE, Negassa A, Chlebowski RT, Lasser NL, McTiernan A, Schenken RS et al. Estrogen plus progestin and risk of benign proliferative breast disease. Cancer Epidemiology, Biomarkers & Prevention 2008;17(9):2337-2343.

(30)   Tan-Chiu E, Wang J, Costantino JP, Paik S, Butch C, Wickerham DL et al. Effects of tamoxifen on benign breast disease in women at high risk for breast cancer. Journal of the National Cancer Institute 2003;95(4):302-307.

(31)   Allred DC, Mohsin SK. Biological features of human pre-malignant breast disease. In: Harris JR, Osborne CK, Morrow M, Lippman ME, editors. Diseases of the Breast. 2nd Edition ed. Philadelphia: Lippincott Williams and Wilkins; 2000. p. 355-366.

(32)   O'Connell P, Pekkel V, Fuqua SA, Osborne CK, Clark GM, Allred DC. Analysis of loss of heterozygosity in 399 premalignant breast lesions at 15 genetic loci. Journal of the National Cancer Institute 1998;90(9):697-703.

(33)   Hoogerbrugge N, Bult P, Widt-Levert LM, Beex LV, Kiemeney LA, Ligtenberg MJ et al. High prevalence of premalignant lesions in prophylactically removed breasts from women at hereditary risk for breast cancer. Journal of Clinical Oncology 2003;21(1):41-45.

(34)   Lundin C, Mertens F. Cytogenetics of benign breast lesions. Breast Cancer Research & Treatment 1998;51(1):1-15.

(35)   Khan SA, Apkarian AV. The characteristics of cyclical and non-cyclical mastalgia: a prospective study using a modified McGill Pain Questionnaire. Breast Cancer Research & Treatment 2002;75(2):147-157.

(36)   Kessel B. Premenstrual syndrome. Advances in diagnosis and treatment. Obstetrics & Gynecology Clinics of North America 2000;27(3):625-639.

(37)   Goodwin PJ, Neelam M, Boyd NF. Cyclical mastopathy: a critical review of therapy. British Journal of Surgery 1988;75(9):837-844.

(38)   Goodwin PJ, Miller A, Del Giudice ME, Ritchie K. Breast health and associated premenstrual symptoms in women with severe cyclic mastopathy. American Journal of Obstetrics & Gynecology 1997;176(5):998-1005.

(39)   Ader DN, Shriver CD. Cyclical mastalgia: prevalence and impact in an outpatient breast clinic sample. Journal of the American College of Surgeons 1997;185(5):466-470.

(40)   Ader DN, South-Paul J, Adera T, Deuster PA. Cyclical mastalgia: prevalence and associated health and behavioral factors. Journal of Psychosomatic Obstetrics & Gynecology 2001;22(2):71-76.

(41)   Berg JC, Visscher DW, Vierkant RA, Pankratz VS, Maloney SD, Lewis JT et al. Breast cancer risk in women with radial scars in benign breast biopsies. Breast Cancer Research & Treatment 2008;108(2):167-174.

(42)   Seltzer MH, Perloff LJ, Kelley RI, Fitts WT, Jr. The significance of age in patients with nipple discharge. Surgery, Gynecology & Obstetrics 1970;131(3):519-522.

(43)   Smith GE, Burrows P. Ultrasound diagnosis of fibroadenoma - is biopsy always necessary?[see comment]. Clinical Radiology 516;63(5):511-515.

(44)   Shetty MK, Shah YP, Sharman RS. Prospective evaluation of the value of combined mammographic and sonographic assessment in patients with palpable abnormalities of the breast. Journal of Ultrasound in Medicine 2003;22(3):263-268.

(45)   Moy L, Slanetz PJ, Moore R, Satija S, Yeh ED, McCarthy KA et al. Specificity of mammography and US in the evaluation of a palpable abnormality: retrospective review. Radiology 2002;225(1):176-181.

(46)   Soo MS, Rosen EL, Baker JA, Vo TT, Boyd BA. Negative predictive value of sonography with mammography in patients with palpable breast lesions. AJR American Journal of Roentgenology 2001;177(5):1167-1170.

(47)   Flobbe K, Bosch AM, Kessels AG, Beets GL, Nelemans PJ, von Meyenfeldt MF et al. The additional diagnostic value of ultrasonography in the diagnosis of breast cancer. Archives of Internal Medicine 2003;163(10):1194-1199.

(48)   Venta LA. Image guided biopsy of non-palpable breast lesions. Harris JR LM, Morrow M OCe, editors. Diseases of the Breast second edition. 149-164. 2000. Philadelphia, Lippincott Williams and Wilkins.

Ref Type: Generic

(49)   Smith RA. The evolving role of MRI in the detection and evaluation of breast cancer.[comment]. New England Journal of Medicine 2007;356(13):1362-1364.

(50)   Ambrogetti D, Berni D, Catarzi S, Ciatto S. The role of ductal galactography in the differential diagnosis of breast carcinoma. Radiologia Medica 1996;91(3):198-201.

(51)   Morrow M, Bland KI, Foster R. Breast cancer surgical practice guidelines. Society of Surgical Oncology practice guidelines. Oncology (Huntington) 1997;11(6):877-881.

(52)   Fentiman IS, Caleffi M, Brame K, Chaudary MA, Hayward JL. Double-blind controlled trial of tamoxifen therapy for mastalgia. Lancet 1986;1(8476):287-288.

(53)   Messinis IE, Lolis D. Treatment of premenstrual mastalgia with tamoxifen. Acta Obstetricia et Gynecologica Scandinavica 1988;67(4):307-309.

(54)   O'Brien PM, Abukhalil IE. Randomized controlled trial of the management of premenstrual syndrome and premenstrual mastalgia using luteal phase-only danazol. American Journal of Obstetrics & Gynecology 1999;180(1):1-23.

(55)   Millet AV, Dirbas FM. Clinical management of breast pain: a review. Obstetrical & Gynecological Survey 2002;57(7):451-461.

(56)   Pashby NL, Mansel RE, Hughes LE, Hanslip JI, Preece P. A clinical trial of evening primrose oil in mastalgi. British Journal of Surgery 1981;68:801.

(57)   Preece PE, Hanslip JI, Gilbert L. Evening primrose oil ( Efamol) for mastalgia. In: Horrobin DF, editor. Clinical uses of essential fatty acid. Montreal: Eden; 1982. p. 147-154.

(58)   Blommers J, de Lange-De Klerk ES, Kuik DJ, Bezemer PD, Meijer S. Evening primrose oil and fish oil for severe chronic astalgia: a randomized, double-blind, controlled trial. American Journal of Obstetrics & Gynecology 2002;187(5):1389-1394.

(59)   Hamed H, Caleffi M, Chaudary MA, Fentiman IS. LHRH analogue for treatment of recurrent and refractory mastalgia. Annals of the Royal College of Surgeons of England 1990;72(4):221-224.

(60)   Mansel RE, Goyal A, Preece P, Leinster S, Maddox PR, Gateley C et al. European randomized, multicenter study of goserelin (Zoladex) in the management of mastalgia. American Journal of Obstetrics & Gynecology 2004;191(6):1942-1949.

(61)   Tarabishy Y, Hartmann LC, Frost MH, Maloney SD, Vierkant RA, Pankratz VS. Performance of the Gail model in individual women with benign breast disease. Journal of Clinical Oncology 2011;29(15_suppl):1525.

(62)   Santen RJ, Boyd NF, Chlebowski RT, Cummings S, Cuzick J, Dowsett M et al. Critical assessment of new risk factors for breast cancer: considerations for development of an improved risk prediction model. [Review] [95 refs]. Endocrine-Related Cancer 2007;14(2):169-187.

(63)   Boyd NF, Jensen HM, Cooke G, Han HL, Lockwood GA, Miller AB. Mammographic densities and the prevalence and incidence of histological types of benign breast disease. Reference Pathologists of the Canadian National Breast Screening Study. European Journal of Cancer Prevention 2000;9(1):15-24.

(64)   Byrne C, Schairer C, Brinton LA, Wolfe J, Parekh N, Salane M et al. Effects of mammographic density and benign breast disease on breast cancer risk (United States). Cancer Causes & Control 2001;12(2):103-110.

(65)   Key T, Appleby P, Barnes I, Reeves G, Endogenous Hormones and Breast Cancer Collaborative Group. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. Journal of the National Cancer Institute 2002;94(8):606-616.

(66)   Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas 1928;38(1):103-113.

(67)   Chlebowski RT, Hendrix SL, Langer RD, Stefanick ML, Gass M, Lane D et al. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial.[see comment]. JAMA 2003;289(24):3243-3253.

(68)   Prentice RL, Manson JE, Langer RD, Anderson GL, Pettinger M, Jackson RD et al. Benefits and risks of postmenopausal hormone therapy when it is initiated soon after menopause.[see comment]. American Journal of Epidemiology 2009;170(1):12-23.

(69)   Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE, Prentice RL et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA 2013;310(13):1353-1368.

(70)   Fournier A, Mesrine S, Boutron-Ruault MC, Clavel-Chapelon F. Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks?[see comment]. Journal of Clinical Oncology 2009;27(31):5138-5143.

(71)   Gail MH, Brinton LA, Byar DP, Corle DK, Green SB, Schairer C et al. Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. Journal of the National Cancer Institute 1989;81(24):1879-1886.

(72)   Tyrer J, Duffy SW, Cuzick J. A breast cancer prediction model incorporating familial and personal risk factors.[see comment][erratum appears in Stat Med. 2005 Jan 15;24(1):156]. Statistics in Medicine 2004;23(7):1111-1130.

(73)   Kuller LH, Cauley JA, Lucas L, Cummings S, Browner WS. Sex steroid hormones, bone mineral density, and risk of breast cancer. Environmental Health Perspectives 1997;105:Suppl-9.

(74)   Chlebowski RT, Collyar DE, Somerfield MR, Pfister DG. American Society of Clinical Oncology technology assessment on breast cancer risk reduction strategies: tamoxifen and raloxifene. Journal of Clinical Oncology 1999;17(6):1939-1955.

(75)   Gail MH, Costantino JP, Bryant J, Croyle R, Freedman L, Helzlsouer K et al. Weighing the risks and benefits of tamoxifen treatment for preventing breast cancer. Journal of the National Cancer Institute 1999;91(21):1829-1846.

(76)   Levine M, Moutquin JM, Walton R, Feightner J, Canadian Task Force on Preventive Health Care and the Canadian Breast Cancer Initiative's Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. Chemoprevention of breast cancer. A joint guideline from the Canadian Task Force on Preventive Health Care and the Canadian Breast Cancer Initiative's Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. CMAJ Canadian Medical Association Journal 2001;164(12):1681-1690.

(77)   Chlebowski RT, Col N, Winer EP, Collyar DE, Cummings SR, Vogel VG, III et al. American Society of Clinical Oncology technology assessment of pharmacologic interventions for breast cancer risk reduction including tamoxifen, raloxifene, and aromatase inhibition. Journal of Clinical Oncology 2002;20(15):3328-3343.

(78)   Cuzick J, Powles T, Veronesi U, Forbes J, Edwards R, Ashley S et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet 2003;361(9354):296-300.

(79)   Vogel VG. The NSABP Study of Tamoxifen and Raloxifene (STAR) trial. [Review] [35 refs]. Expert Review of Anticancer Therapy 2009;9(1):51-60.

 

 

Medication Induced Changes in Lipid and Lipoproteins

ABSTRACT

 

Several medications and medication classes have been reported to affect the lipid profile. Risk factors include elevated lipid levels at baseline and high cardiovascular (CV) risk patients.  This should be considered when evaluating patients with elevated levels of total cholesterol (TC), low-density lipoproteins cholesterol (LDL-C), non-high-density lipoprotein cholesterol (Non-HDL-C), triglycerides (TG) and reductions in high-density lipoprotein cholesterol (HDL-C). Cardiovascular medications, antipsychotics, anticonvulsants, hormones and certain immunosuppressives are just some of the more commonly known medications to have a negative impact on lipid levels. In some cases, this is a class effect and in others it might depend on dose and specific drug. However, how this translates to atherosclerotic cardiovascular disease (ASCVD) risk remains unknown, as there is insufficient evidence on the impact of these metabolic changes on overall risk of ASCVD. While for many of these medications, there is an abundance of literature and comprehensive reviews discussing the potential harmful effects of on lipoprotein metabolism there remains much debate about the actual long-term implications, if any, of these changes. A thorough risk-benefit analysis of each treatment associated with an adverse effect on the lipid profile should be done based on individual patient factors. In general, if negative changes in the lipid profile are observed during therapy, replacement with an equivalent alternative therapy can be recommended. If no equivalent therapy is available and treatment must be initiated, then monitoring of serum lipid levels is vital. The use of existing guidelines for the management of dyslipidemia in the general population can be referred to and in extreme cases when benefits do not outweigh the risks; the use of the suspected medication should be reassessed. For complete coverage of this area and all of Endocrinology, visit www.endotext.org.

 

INTRODUCTION

 

Secondary causes of dyslipidemia are important to identify as treatment of the underlying cause may alleviate the dyslipidemia and ultimately reduce the need for drug treatment or the need for combination pharmacotherapy.1Guidelines recommend that providers should evaluate for underlying conditions that could be causing dyslipidemias before initiating treatment in patients.2One such secondary cause of abnormally altered lipid or lipoprotein levels is medications used for other indications.3Serum lipid levels can be affected both positively and negatively by certain medications. Medications can affect lipid levels either directly or indirectly through effects on weight or glucose metabolism. This should be considered when evaluating patients with elevated levels of total cholesterol (TC), low-density lipoproteins cholesterol (LDL-C), non-high-density lipoprotein cholesterol (Non-HDL-C), triglycerides (TG) and reductions in high-density lipoprotein cholesterol (HDL-C).4

 

There have also been reports of various medications causing severe drug-induced hypertriglyceridemia that leads to acute pancreatitis.5-7  While there is a paucity of data describing the exact mechanism of drug-induced pancreatitis, it is known that severe hypertriglyceridemia (TG > 1000 mg/dl) can cause acute pancreatitis.  Therefore, in the absence of other causes, medications should be evaluated in the presence of acute pancreatitis and severe hypertriglyceridemia.

 

Several medications and medication classes have been reported to affect the lipid profile (Table 1). In some cases, this is a class effect, and some instances agents belonging to the same class can have significantly different actions on lipid levels (e.g. beta blockers).8   This is a consideration to appreciate when selecting a specific agent for high-risk patients and concurrent medications known to induce lipid abnormalities should be evaluated for discontinuation or dosage reduction prior to initiating long-term lipid lowering agents. How this translates to atherosclerotic cardiovascular disease (ASCVD) risk remains unknown, as there is insufficient evidence on the impact of these metabolic changes on overall risk of ASCVD.

 

Table 1. Drugs That May Cause Dyslipidemias
  LDL Cholesterol Triglycerides HDL Cholesterol
Cardiovascular /Endocrine
Amiodarone ↑Variable
β-Blockers*** ↑10-40% ↓5-20%
Loop diuretics ↑5-10% ↑5-10%
Thiazide diuretics (high dose) ↑5-10% ↑5-15%
Sodium-glucose co-transporter 2 (SGLT2) inhibitors ↑3-8% ↔↓ ↑Variable
Steroid Hormones/Anabolic Steroids
Estrogen ↓7-20% ↑40% ↑5-20%
Select progestins ↑Variable ↓Variable ↓15-40%
Selective Estrogen Receptor Modulators ↓10-20% ↑0-30*
Danazol ↑10-40% ↓50%
Anabolic steroids ↑20% ↓20-70%
Corticosteroids ↑Variable ↑Variable
 Antiviral Therapy
Protease inhibitors ↑15-30% ↑15-200%
Direct Acting Antivirals ↑12-27% ↑14-20%
Immunosuppressants
Cyclosporine and tacrolimus ↑0-50% ↑0-70% ↑0-90%
Corticosteroids ↑Variable ↑Variable
Centrally Acting Medications
First Generation antipsychotics ↑22% ↓20%
Second Generation antipsychotics ↑20-50%
Anticonvulsants ↑Variable ↑Variable
Other Medications
Retinoids ↑15% ↑35-100% **
Growth Hormone ↑10-25% ↔↑7%
ABBREVIATIONS: LDL, low-density lipoprotein; HDL, high-density lipoprotein. *Raloxifene has not been shown to increase Triglyceride levels, while reported increases of up to 30% have been reported with use of tamoxifen**Data remains conflicting and some evidence shows a decrease, no effect, or increase***Varies based on individual drug 

 

ANTIHYPERTENSIVE DRUGS

 

There is an abundance of literature and comprehensive reviews discussing the potential harmful effects of antihypertensive drugs on lipoprotein metabolism and there remains much debate about the actual long term implications, if any, of these changes.9The diuretics and β-

adrenergic blockers have the most data to support their adverse effects on lipid levels.10-15

 

Diuretics

 

Thiazide and loop diuretics have been associated with increases in plasma cholesterol in studies of patients with hypertension. Recent recommendations from the American College of Cardiology/American Heart Association recommend thiazide diuretics as one of four specific medication classes to be considered as initial therapy for hypertension.16In view of these recommendations and widespread use of diuretics, it is important to review the adverse metabolic effects. Use of high-dose thiazide diuretics (≥50 mg/day) may negatively affect lipoprotein levels, as seen in small studies, and some investigators have suggested that as a result, diuretics could worsen coronary artery disease (CAD).12Total cholesterol levels can be increased by approximately 4% and LDL-C levels by approximately 10%.9,15,17High density lipoprotein levels are not affected, while TG concentrations can also be elevated by 5-15%.9  Low dose hydrochlorothiazide (12.5 – 25 mg/day) has been shown not to effect plasma lipids in otherwise healthy men and women.12The dose appears to be a factor in resulting cholesterol levels18; however, there are conflicting data regarding whether the effects on lipid levels is primarily caused by higher doses.12 Long term effects beyond one year remain undetermined as more recent studies showed that effects are short term and serum lipid levels return to initial levels.19Additionally, thiazide diuretics have been shown to decrease the risk of cardiovascular (CV) events despite this effect on lipid levels.20

 

Loop diuretics have similarly shown to increase LDL-C and TG with some studies showing changes of comparable magnitude and some showing effects that are less than thiazide diuretics.21,22However, the effects appear to be acute and not expected at time intervals longer than the duration of action of furosemide (6 to 8 hours). One possibility is that hormones stimulated in response to decreased intravascular volume are responsible for some changes in lipid and lipoprotein levels.22The effects of monotherapy with potassium-sparing diuretics on lipid levels is largely unknown, but the combination of a potassium-sparing diuretic and a thiazide show similar changes as monotherapy with a thiazide diuretic, suggesting no impact from potassium-sparing diuretics.

 

The mechanism of increased lipid levels caused by diuretics remains unclear. One theory is that a reduction in insulin sensitivity may cause an increase in hepatic production of cholesterol.17,23The mechanism of an increase in TGs is not well understood. It has been recently suggested that they may modulate adipocyte differentiation leading to accumulation of plasma TGs in susceptible patients with a particular genetic polymorphism in the NELL1 gene.24It is also thought that there are sex differences, as diuretics were shown to produce a greater short-term increase in TC and LDL-C in postmenopausal women than in men. Premenopausal women may have a protective effect from estrogens and have demonstrated no changes in lipid levels.15Estrogens have been theorized to increase the number of hepatic LDL binding sites and stimulate the hepatic uptake of chylomicron remnants.17

 

β-Blockers

 

The metabolic adverse effects of β-blockers depend on dose and specific drug. While β-blocking agents have negligible effect on serum TC or LDL-C, they can increase TG levels from 10 to 40% and decrease HDL-C levels by approximately 5 to 20%.9The evidence on duration of effect remains conflicting with studies citing effects to last less than 1 year19, and other studies reporting increased levels after several years of treatment.24The alterations in lipoprotein levels from β-blockers does not appear to be a class effect, and agents with intrinsic sympathomimetic activity (ISA), β1-selectivity, or vasodilatory effects (Table 2) are associated with a less pronounced effect.9Non-selective β-blockers which cause peripheral vasoconstriction through peripheral β-adrenergic receptors seem to increase insulin resistance, leading to lowering of HDL-C, and increased TG.25Whereas, agents that are cardioselective and/or have alpha-1-adrenoreceptor blocking activity do not appear to increase insulin resistance. Other potential mechanisms of β-blocker induced lipid changes are from β-blocker associated weight gain, a decreased lipid metabolism through a reduction in the muscle lipoprotein lipase enzyme, and endothelial dysfunction from peripheral vasoconstriction (Table 3).26The beneficial effects of carvedilol compared to metoprolol and atenolol on lipid parameters has been demonstrated in several small studies.13,25,27Carvedilol has selective α -1-adrenoreceptor blocking activity, causing vasodilation and a reduction in insulin resistance. It remains unknown if these beneficial metabolic effects of carvedilol are seen with other beta-blockers with vasodilating properties, including nebivolol. Conversely, selective α-blocking agents (prazosin) have a beneficial effect on lipid profile and have been shown to increase HDL-C and decrease TG.28,29

 

Table 2. Pharmacological Properties of β-Blockers  
   
  Beta Selectivity Intrinsic sympathomimetic (ISA) or α-blocking Vasodilating Properties
More pronounced effect on lipid levels
Atenolol β1 selective - -
Betaxolol β1 selective - -
Bisoprolol β1 selective - -
Metoprolol β1 selective - -
Nadolol Nonselective - Vasoconstricting
Propanolol Nonselective - Vasoconstricting
Timolol Nonselective - Vasoconstricting
Less pronounced effect on lipid levels
Acebutolol Nonselective ISA Vasoconstricting
Penbutolol Nonselective ISA Vasoconstricting
Pindolol Nonselective ISA Vasoconstricting
No effect on Lipid levels
Carvedilol Nonselective α-blocking Vasodilating
Labetolol Nonselective α-blocking Vasodilating
Nebivolol β1 Selective - Vasodilating
           

 

Thiazide diuretics and β-blockers are important agents for other cardiovascular indications. β-blockers are effective in reducing cardiovascular morbidity and mortality in congestive heart failure and CAD and diuretics are vital for symptomatic management of many CV comorbidities. While it is important to assess for increased lipid levels caused by these agents, with other compelling indications, it remains prudent to continue them while continuing to monitor serum lipid levels.

 

Table 3. Potential Mechanism of β-blocker Induced Dyslipidemia
Inhibition of insulin release
Insulin resistance
Weight gain
Inhibition of lipolysis
Reduced activity of lipoprotein lipase enzyme
Endothelial dysfunction

 

OTHER CARDIOVASCULAR MEDICATIONS

 

Amiodarone

 

Amiodarone, a potent antiarrhythmic drug, increases plasma cholesterol levels, reported in case reports.30,31Amiodarone increases LDL-C levels as a result of a decreased expression of the LDL-receptor gene.30,32,33In addition, amiodarone induced hypothyroidism can cause alterations in lipid metabolism as hypothyroidism is one of the most common causes of secondary hyperlipidemia. Amiodarone contains 39.4% iodine on weight basis which may cause hyperthyroidism or hypothyroidism.33Research demonstrates that long-term amiodarone treatment induces a dose-dependent increase in plasma cholesterol, in part due to thyroid hormone deficiency and a decrease in the number of LDL receptors.34,35

 

DIABETES MEDICATIONS

     

Sodium-Glucose Co-Transporter 2 (SGLT2) Inhibitors

 

The SGLT2 inhibitors lower blood glucose and hemoglobin A1c (HgA1c) through inhibiting SGLT2 in the proximal tubule, thereby blocking reabsorption of glucose and increasing the renal excretion of glucose.36  There are currently four SGLT2 inhibitors available and approved for the treatment of type 2 diabetes mellitus (Table 4).  In addition to their effects on glucose lowering, SGLT2 inhibitors have been shown to have positive effects on other metabolic parameters, including body weight and blood pressure. 36  Although data is mixed and the individual agents appear to affect the lipid profile to a varying degree, these agents have shown to increase LDL-C while also increasing HDL-C with variable effects (decreasing or no effect) on TG (Table 4). While a decrease in weight could explain the favorable effects seen on HDL-C and TG, it remains unclear what the mechanism behind the modest dose-related increase in LDL-C is.37  One possibility is that SGLT2 inhibitors cause a switch from carbohydrate to lipid utilization causing an increase in hepatic fatty acid levels to induce ketone production and hepatic total cholesterol levels.37  How this impacts CV health is uncertain.  To date, canagliflozin and empagliflozin have demonstrated an improvement in CV outcomes (composite of CV mortality, nonfatal myocardial infarction (MI), or nonfatal stroke) and a reduction in heart failure hospitalizations in subjects with high CV risk.38For information on the effect of other glucose lowering drugs on lipids and lipoprotein see the chapter in the Diabetes section of Endotext entitled “Role Of Glucose And Lipids In The Cardiovascular Disease Of Patients With Diabetes”.38

 

 

Table 4. Sodium-glucose co-transporter 2 (SGLT2) inhibitors and their effects on low density lipoprotein cholesterol (LDL-C)39-42

Generic Brand Dose-Related Effects on LDL-C
Canagliflozin Invokana® 4.5% - 8.0%
Dapagliflozin Farxiga® 2.9%
Empagliflozin Jardiance® 2.3% - 6.5%
Ertugliflozin Steglatro® 2.6% - 5.4%

 

STEROID HORMONES

 

Estrogens and Progestins

 

Oral estrogens have been shown to be important regulators of lipid metabolism.43,44Premenopausal women are protected from diseases such as CAD, hypertension, diabetes and dyslipidemia. This is seemingly due to estrogens having a protective effect, as unopposed estrogens beneficially affect the lipid profile. They lower TC (2-10%) and LDL-C levels (7-20%) and increase HDL-C levels (5-20%) in a dose-related manner.3,9 Studies have also shown a decrease in lipoprotein(a) levels, which is an independent risk factor for coronary heart disease (CHD), with both estrogen and progestin therapy.45However, they also increase TG levels up to 40%, and can increase the risk of pancreatitis in women with overt hypertriglyceridemia.46Ethinyl-estradiol has a more pronounced effect on lipoproteins than natural estradiol.47Oral estrogen therapy increases TG plasma levels by increasing production of very-low density lipoprotein (VLDL), reducing the concentrations of lipoprotein lipase and hepatic lipase, resulting in a reduction of TG clearance, and potentially through a change in insulin resistance.43,46Triglyceride levels vary over time in women who are taking cyclic hormone regimens and it is assumed that the increase is enhanced among those with preexisting hypertriglyceridemia. Elevations in TGs are not usually observed with transdermally administered estrogens, as the transdermal application is thought to reduce the hepatic first pass effect and reduced impact on hepatic protein synthesis.3,48,49

 

Progestins antagonize the estrogen-induced lipid changes, and can have a negative effect on TC and HDL-C.48,50,51Serum lipid levels depend on the androgenic effects of the progestin.9Progestins with more androgenic effects, such as levonorgestrel, are theorized to have larger effects on lipid levels than those with less androgenic effects.52Newer, “third generation” progestins (desogestrel, gestodene) with higher specificity have been developed to reduce this risk and have demonstrated favorable effects on LDL-C levels and HDL-C levels, but they can also increase TGs.47,53There remains insufficient evidence that third generation agents lower risk of ASCVD. Also, in recent years, the dose of ethinyl estradiol has been decreased from 50 down to just 20-30 mcg in current formulations, to also reduce adverse metabolic changes. Therefore, the effect varies with oral contraceptives based on their estrogen and progestin content and more specifically the potency of the estrogen and the adrogenicity of the progestin. Selective estrogen-receptor modulators, including raloxifene and tamoxifen, appear to have less impact on lipids, but can still elevate TG levels.48  There have been several case reports in the literature describing tamoxifen-induced hypertriglyceridemia causing acute pancreatitis.6,7,54,55

 

The American Heart Association recommends that lower estrogen-containing preparations or other forms of contraception should be considered in women who develop hypertriglyceridemia while taking therapy.48   Postmenopausal women with hypertriglyceridemia who require hormone therapy are encouraged to switch to transdermal preparations. It is not clear if transdermal application influences cardiovascular outcomes.48In addition, oral contraceptives (OC) with low doses of estrogen should be used in women with controlled dyslipidemia, as studies of low dose triphasic OC have resulted in no significant changes or only mild elevations in TC, LDL-C, and TG levels. Contraceptives can be considered for women with uncontrolled LDL-C levels or multiple CV risk factors, including using non-androgenic or anti-androgenic progestins as they have minimal influence on the lipid profile.50

 

In postmenopausal women, hormone therapy with estrogen alone or estrogen combined with a progestin is utilized for the treatment of hot flashes and other menopausal symptoms. Similar to studies evaluating OC, oral estrogen has also been shown to increase HDL-C and TG levels, and reduce LDL-C.49Combined hormone replacement therapy regimens have similar effects on TC levels and LDL-C as estrogens.9Studies have shown that unopposed estrogen has a more beneficial effect on HDL-C than estrogen and progestin in combination but both similarly lowered LDL-C and increased TG in postmenopausal women.43There is conflicting evidence if hormone replacement therapy is associated with a protective cardiovascular effect in postmenopausal women and more recent data from a systematic review demonstrated that estrogen only increased the risk of a stroke (RR 1.34; 95% CI 1.07 to 1.68) and venous thromboembolism (RR 1.32; 95% CI 1 to 1.74), and there was no significant difference compared to placebo in risk of coronary events.56Combined hormone therapy compared to placebo demonstrated a significant increase in coronary events (RR 1.89; 95% CI 1.15 to 3.1), stroke (RR 1.38; 95% CI 1.08 to 1.75), and venous thromboembolism (RR 4.28; 95% FI 2.49 to 7.34).56   Data has also shown that the effects of hormone therapy on CV outcomes are influenced by age and time since onset of menopause and that estrogen may slow down the early stages of atherosclerosis and have more favorable effects in women with more recent onset of menopause.57Overall, long term data suggests that hormone therapy may have a harmful effect on CHD risk in older women, and the results in younger women remain inconclusive. At this time, treatment for the purpose of prevention of coronary heart disease or chronic disease prevention is not recommended. For postmenopausal women, short-term therapy should be used at the lowest effective dose.58For those with hypertriglyceridemia, the use of transdermal estrogen may be a preferred alternative to oral.

 

ANABOLIC STEROIDS

 

Danazol

 

Danazol is a synthetic steroid indicated for endometriosis and fibrocystic breast disease as well as for prophylaxis in patients with hereditary angioedema (HAE).59,60A review of data for the treatment of endometriosis showed that danazol treatment can result in a rapid reduction in HDL-C by up to 50% and increase in LDL-C by 10-40%.61However, these levels return to baseline levels after stopping therapy and there is only a concern in prolonged therapy for 12 months or greater or in patients with a high risk of CV disease.60This effect is consistent in the literature.62-65The mechanism is likely from its effects on hepatic lipase, LDL receptor, and lecithin cholesterol acyl transferase activity. Data also supports an altering of lipoprotein levels in women treated for endometriosis, but there may not be an effect in the treatment of HAE.66Some possible explanations for this difference are that HAE doses are lower than doses used for the treatment of endometriosis, the duration of therapy is longer and often lifelong versus 2-6 months for endometriosis, and men are also treated for HAE. A randomized trial evaluated danazol on HDL-C in healthy volunteers and patients with HAE. Patients with CV disease or significant risk factors for CV disease were excluded in the healthy volunteer study. Short-term use in healthy subjects (n=15) demonstrated a 23% decrease in HDL-C levels; however, these were normalized by 4 weeks of treatment. There was no effect seen on LDL-C or TG. Longer-term use in patients with HAE did not appear to decrease HDL-C levels compared to matched healthy controls. This study supports the belief that the differences in study populations as well as varying duration and doses of danazol impact the effect danazol has on the lipid profile. However, other studies have shown conflicted results and also demonstrated decrease in HDL-C (as well as apolipoprotein Apo A-I; the major component of the HDL particle) and increase in LDL in long term use for the prevention of HAE.66,67However, this negative effect was not shown to translate into an increased risk of atherosclerosis.67

 

Androgens

 

Similarly, studies of bodybuilders and weight lifters using anabolic steroids have revealed reductions in HDL-C levels by 20-70% accompanied by decreases in apo A-I levels, as well as elevations in LDL-C by approximately 20%.68-71The misuse of anabolic steroids in strength athletes has previously been associated with CV events which may be in part due to the adverse lipid effects associated with their use. One small study confirmed that self-administration of anabolic steroids produced unfavorable effects on lipids and lipoproteins, including a decrease in serum concentrations of HDL-C, and Apo-A1.70Serum LDL-C levels may increase through induction of hepatic triglyceride lipase and the catabolism of VLDL. Through this process, HDL-C serum levels are also reduced. A more recent literature review described 49 reports of 1,467 athletes using anabolic-androgenic steroids corroborating the link and reports that these changes can occur within 9 weeks of self-administration and the effects seem to be reversible and normalize within 5 months after discontinuation.72The majority of the evidence is based on small, observational studies or single case reports and may reflect significant publication bias. However, use of anabolic-androgenic steroids has also been linked to elevated blood pressure, left ventricular hypertrophy, acute myocardial infarction, and sudden death and awareness of these potential adverse effects may benefit athletes and increase recognition of young otherwise healthy individuals with CV abnormalities.

 

Androgen deprivation therapy (ADT) is hormone therapy used for the treatment of prostate cancer and is associated with a variety of metabolic adverse events, including lipid alterations.73This can be done by surgical castration or medical castration with gonadotropin-releasing hormone (GnRH) agonists (also called luteinizing hormone-releasing hormone (LHRH) agonists). These agents can cause changes in lipid levels, including increases in TC, TG, and HDL-C.73Studies have shown different changes on LDL-C; with some showing an increase and others with no significant changes. Small studies have demonstrated increases in TGs of up to 25% and HDL- C increases of up to 11%. Furthermore, a longer term study over 1 year observed these changes did not persist after 6 months.74Given that ADT may increase the risk of lipid changes as well as obesity and insulin resistance, studies have also evaluated the effects of ADT on CV disease and observational studies have suggested an association between ADT and a greater risk of CV disease.75One explanation for this association is that ADT interferes with the cardioprotective property of testosterone and therefore, increases the risk for adverse events. In 2010, the FDA released a drug safety communication informing of the increased risk of diabetes and certain CV diseases (heart attack, sudden cardiac death, stroke) in men receiving GnRH agonists for the treatment of prostate cancer based on their review of several published studies.76However, this was based on mostly small observational studies and RCTs have remained conflicted on this relationship, as seen in a recent meta-analysis of randomized trials.77

 

Testosterone replacement products are approved for men who have low testosterone levels caused by various medical conditions. Nonetheless, the use of testosterone therapy is increasing, including for men who have low testosterone simply due to aging.78Recent studies have shown that the risk of MI and other CV-related events may be increased with the use of testosterone therapy and that testosterone therapy should be avoided in certain high risk patients.78,79However, most of the data remains observational and many report conflicting results. Further RCTs are needed to clarify the concern. If treatment with testosterone does increase the development of arteriosclerotic heart disease, one potential mechanism is through an adverse effect on serum lipids and apolipoprotein levels. However, studies to date have not demonstrated a significant effect on lipid or lipoprotein levels, with possibly a slight decrease in HDL-C occurring due to changes in the HDL protein composition.80-82

 

GROWTH HORMONE

 

Adults with growth hormone deficiency frequently have lipid abnormalities, decreased insulin sensitivity and an increased CV morbidity and mortality. Treatment with recombinant human growth hormone, or somatropin, for adults with growth hormone deficiency has contributed to positive lipid changes, including decreased levels of TC and LDL-C by 10-25%.83-88There appears to be no significant effect on TG levels and data is conflicted on changes in HDL-C, with most studies demonstrating an increase in serum HDL-C.84Conversely, studies have showed no effect on HDL-C, as well as a decrease by approximately 20% has been reported.89There is some data to suggest that individual response to growth hormone on lipid metabolism is partly influenced by genetic polymorphisms in genes related to lipid metabolism.90

 

A recent prospective, open-label, single-center study reports the effects of 15 years of growth hormone replacement in 156 adults with growth hormone deficiency.84After prolonged therapy, there were decreases in serum levels of TC and LDL-C; with corresponding increases in HDL-C (p<0.001 for all vs. baseline levels). There were no significant changes in serum TG levels. This long observational study demonstrates that treatment of growth hormone deficiency in adults results in sustained improvements in the serum lipid profile. This could be due to improvements in body composition or direct effects on lipid metabolism. Studies suggest that growth hormone may increase the expression of LDL receptors, improves the catabolism of LDL, increase the turnover of LDL, and increase apo B-100 turnover. Nonetheless, evidence that these improvements result in decreased mortality from ASCVD remains unknown.

 

RETINOIDS

 

Retinoids are synthetic analogues of Vitamin A effective for the treatment of psoriasis, severe acne and other related skin disorders caused by abnormal keratinization. Oral isotretinoin was first reported to cause hypertriglyceridemia; most likely due to a reduction in the clearance of VLDL particles, which interferes with lipoprotein lipase-mediated lipolysis.91,92 Retinoids have also been shown to increase plasma apo C-III concentrations by increasing the transcriptional activity of the human apo C-III gene via the retinoid X receptor (RXR), ultimately contributing to the development of hypertriglyceridemia.93,94Isotretinoin has been established as effective treatment for severe nodular acne that is unresponsive to conventional therapy, including systemic antibiotics. However, it has been reported to cause a variety of adverse events with some potential serious consequences, including case reports of pancreatitis.95Patients with significantly elevated TG levels are more likely to develop pancreatitis and therefore, patients with preexisting hypertriglyceridemia should avoid retinoid therapy or use with extreme caution until TG levels can get better controlled. In addition, a baseline lipid profile should be obtained in all patients and TG levels checked at least once after four weeks of therapy. Patients with a higher risk of developing hypertriglyceridemia should be monitored more frequently.

 

Studies with isotretinoin have demonstrated a rise in VLDL-C, TG, LDL-C, and TC with a slight decrease in HDL-C .3One study evaluated the subsequent risk in ASCVD in 104 men and women using isotretinoin for severe acne using the ratio of TC/HDL-C.84The results showed that in otherwise healthy individuals, the changes in lipid metabolism did not influence the overall risk of ASCVD significantly.

 

ANTIPSYCHOTICS

 

Antipsychotic medications can be highly effective in controlling psychiatric illnesses. However, some of these are also associated with metabolic adverse events that can increase the risk for ASCVD.96-99One such adverse event includes dyslipidemia, with increases primarily occurring in TG levels. Phenothiazines, which are first generation or ‘typical’ antipsychotics, were found to elevate serum TG and TC levels soon after their approval, with a greater effect on TG levels. Studies have shown an increase of up to 22% after a year of treatment with chlorpromazine. 3Further studies have observed similar effects with trifluronated phenothiazines and haloperidol. The main limiting adverse effects of first generation antipsychotics are extrapyramidal symptoms and other movement disorders due to their high-affinity of dopamine D2receptors. Still, the possibility that these drugs also contribute to lipoprotein abnormalities should be considered in patients with dyslipidemia or high CV risk.

 

Second generation, or ‘atypical’ antipsychotics were later developed to reduce relapse rates and adverse events. Compared to first generation antipsychotics, they have lower affinity for the D2receptors and instead act namely on serotonin and norepinephrine. They have become first line treatment due to a lower potential risk of extrapyramidal symptoms. However, metabolic side effects including an increase in serum TG levels as well as minor increased in TC, has also been demonstrated with the use of second generation antipsychotics. Some studies suggest this is a result of increased leptin levels; an adipocyte hormone that corresponds with a decrease in the synthesis of fatty acid and TG and an increase in lipid oxidation.100There are many other possible mechanisms for the drug induced hyperlipidemia and the exact mechanisms are not fully understood. Clozapine, a second-generation antipsychotic, was the first agent shown to increase serum TG levels.101A retrospective study reviewed patients on clozapine and found that men on clozapine had an average 48.13% increase in TG level and women 35.38% and there was a significant interaction between drug and gender over time (p<0.05).101

 

In addition, weight gain is a common adverse effect of using atypical antipsychotics which can also lead to an increase in both leptin and TG levels.100   The 5-HT2c receptor-blocking and/or histamine antagonism action of these medications is a possible cause of the related weight gain. One study demonstrated a significant increase in weight and serum TG and leptin levels with olanzapine and clozapine, with minimal and moderate changes in those on risperidone and quetiapine, respectively.100However, this was an extremely small study (n=56) that and it is difficult to translate these results to the general population. This is consistent with the overall evidence and it appears that clozapine and olanzapine have the greatest effect on the risk of hyperlipidemia, followed by quietapine. Risperidone, ziprasidone and aripiprazole have a relatively low risk of hyperlipidemia associated with their use.101As access to general and preventive care remains a limitation for patient populations with schizophrenia, these adverse effects can be of great concern for a population already at increased risk of CV complications. Therefore, checking baseline lipid levels and screening for the duration of therapy may be necessary in patients receiving therapy with atypical antipsychotics. If a patient develops elevated TG levels or dyslipidemia, they should be offered lipid-lowering therapy or if possible, switched to a less offending agent.

 

ANTICONVULSANTS

 

Changes in serum lipid levels have been reported with the use of various anticonvulsants with variability and inconsistency in the literature. Some observational studies have reported elevated levels of LDL-C and HDL-C while others have shown no significant effects. Triglyceride levels are not influenced by treatment with anticonvulsants. Since most anticonvsulants induce the hepatic cytochrome P450 (CYP) enzymes, it is theorized that competition between the medication and cholesterol for the enzyme occurs which results in a decreased breakdown of cholesterol to bile acids and an increase in TC.9This inconsistency has been noted in studies in both adults and pediatrics with epilepsy and it appears differences may exist based on the individual anticonvulsant used. In addition, the influence of therapy on development of atherosclerosis remains debatable.

 

Epilepsy often requires lifelong therapy and the long-term administration of some antiepileptic drugs (AEDs) is associated with metabolic side effects from dysfunction of the vessel wall. In particular, carbamazepine and phenobarbital have shown to cause alterations in the lipid profile. In children and adolescents with epilepsy, carbamazepine has demonstrated a consistent increase in TC and LDL-C levels, while some individual studies have also shown an increase in HDL-C as well as TG levels.102,103Treatment of epilepsy in children with phenobarbital has also shown increased TC, LDL-C and HDL-C, as well as lower TG levels. Valproic acid appears to have little effect, or even a slightly favorable effect, on the lipid profile.102

 

IMMUNOSUPPRESSIVES

 

Corticosteroids

 

It has been generally postulated that chronic glucocorticoid excess is a secondary cause of dyslipidemia, but the degree of lipid abnormalities in clinical conditions is extremely variable and studies have remained conflicted and inconsistent.104Observational studies of steroid treatment in patients with asthma, rheumatoid arthritis, or connective tissue disorders have shown elevations in TC, LDL-C, and serum TG levels.3,105These are all illnesses that may require long-term treatment with corticosteroids. However, a large survey demonstrated no association with an adverse lipid profile and glucocorticoid use.106One study found that pre-menopausal females who were taking corticosteroids for a mean of 3.1 years had a significant elevation in TC and decrease in HDL-C. Conversely, a study in female patients with asthma noticed a significant increase in serum TG but no changes in TC.107The potential mechanisms of the effect on the lipid profile is multifactorial. One theory for this increase in TG is due to the redistribution of body fat caused by corticosteroid treatment to the upper trunk and face with a loss of fat in the extremities resulting in cells with fewer glucocorticoid receptors in addition to the stimulation of both lipolysis and lipogenesis.104This results in a spared effect on glucose transport in cells with fewer receptors resulting in an accumulation of glucose and TG secondary to a rise in insulin levels.108Insulin resistance also plays a role in lipid abnormalities. In the liver, glucocorticoids cause hyperglycemia, increase VLDL production, enhance hepatic lipogenesis and inhibit fatty acid β-oxidation. Furthermore, they increase the synthesis and secretion of apolipoprotein AI.

 

Changes in lipid metabolism due to corticosteroid treatment has also been evaluated in women with systemic lupus erythematous (SLE).109Patients with SLE may be at an increased risk for atherosclerotic CAD, which could be potentiated by the changes in lipid serum levels from corticosteroid administration. When compared to women with SLE not treated with prednisone, patients on prednisone had higher TG, TC, and LDL-C levels. 109It appears that women may be more prone to these changes than men, and in many of these chronic illnesses, the use of corticosteroids is unavoidable. It is prudent to educate patients about the risks and benefits associated with long-term therapy with corticosteroids and to support life-style changes that help prevent ASCVD.

 

Cyclosporine and Tacrolimus

 

Cyclosporine and tacrolimus are immunosuppressant agents used as mainstay therapy for solid organ transplant recipients. Several metabolic abnormalities are associated with the use of both of these medications, including glucose intolerance, bone loss, and elevations in TC and LDL-C and apo B-100 levels. Effects on HDL-C levels are inconsistent, but trials have also demonstrated increases in HDL-C.9Hyperlipidemia can occur in up to 60% in post-transplant patients.110This is due to a combination of factors, including post-transplant obesity, multiple drug therapy (including steroids and other immunosuppressants) and diabetes. These drug effects are much greater with cyclosporine than tacrolimus, which has minimal effects on TC and LDL-C, and smaller effects on TG levels than cyclosporine.111A randomized prospective trial compared a tacrolimus-based regimen to a cyclosporine-based regimen in patients undergoing a cardiac transplant. After 12 months of therapy, serum TC, LDL-C, HDL-C and TG were significantly higher in the cyclosporine group compared to tacrolimus and more patients received medical treatment for elevated lipids (71% vs. 41%; p=0.01).100The impact of cyclosporine on lipid levels appears to be dose dependent and trough blood levels correlate with the elevations in TC and LDL-C, as well as reductions in HDL-C levels.112The mechanisms by which cyclosporine causes hyperlipidemia are unclear, as the effects in humans are confounded by many other factors in transplant patients.

 

Due to the complex state of transplant patients and the increased risk of ASCVD, attention should be given to these adverse events and other risk factors. Serum drug levels should be monitored during treatment as increases in drug levels are associated with negative adverse events. In addition, if appropriate, conversion from cyclosporine to tacrolimus can be considered if post-transplant hyperlipidemia occurs, and several studies have demonstrated this.113-115Although patients may benefit from therapy with a HMG-CoA reductase inhibitor, concomitant use of cyclosporine and HMG-CoA reductase inhibitors has been shown to increase the risk of myopathy and rhabdomyolysis due to a potential drug-drug interaction through inhibition of the CYP3A-mediated metabolism of simvastatin and cyclosporine inhibition of the organic anion transporter protein (OATP1B1)-mediated hepatic uptake of simvastatin.116,117As pravastatin and fluvastatin are not significantly metabolized by CYP enzymes, they may be a favorable choice in this patient population due to the decreased risk of drug-drug interactions.118However, pravastatin and fluvastatin doses should still be lowered due to cyclosporine inhibition of the OATP1B1-mediated hepatic uptake. Furthermore, fluvastatin has been shown to reduce CV events and lower LDL-C concentrations in transplant recipients receiving immunosuppressive therapy with cyclosporine.119

 

ANTIVIRAL THERAPY

 

Protease Inhibitors

 

Protease inhibitors (PIs) are potent antiretroviral drugs used in combination with other therapy as part of a antiretorivral regimen for the treatment of human immunodeficiency virus (HIV).120These PIs have substantial clinical benefits, but can also produce lipodystrophy, hyperlipidemia, and insulin resistance.121,122The hyperlipidemia is thought to be caused by increases in VLDL production and intermediate density lipoproteins (IDL) with the potential to cause endothelial dysfunction and atheroslcerosis. Enzymes imperative for the removal of triglyceride rich lipoproteins are also decreased in HIV patients. This includes lipoprotein lipase and hepatic lipase. PI associated insulin resistance and abnormal expression of the apolipoprotein C-III gene may also induce dyslipidemia.123

 

Studies evaluating PIs have shown increases in TC as well as triglycerides with little to no effects on HDL-C and inconsistent increases in LDL-C levels.124There is insufficient evidence directly linking dyslipidemia and the risk of CHD in HIV infected individuals. The main increase being in triglyceride-containing lipoproteins supports the mechanism of a release of free fatty acids and resulting increase in synthesis of VLDL causing these changes. While all PIs can change lipid levels, ritonavir appears to have the greatest effects and has also been reported to cause cases of extreme hyperlipidemia. During a randomized, 4-week double blind study, ritonavir was associated with at least a doubling of the serum triglyceride level in 24 (61%) patients compared to only 4 (19%) patients on placebo (p=0.003) and seven subjects had triglyceride levels exceeding 1000 mg/dl.125Patients with high serum triglyceride levels are at a higher risk of pancreatitis, which has been reported after protease inhibitor therapy.126Therefore, the long-term complications of elevated lipids in the setting of HIV should be taken into consideration in patients treated with PIs. Guideline supported lipid lowering therapy and efforts to modify other CV risk factors should be initiated in patients.127Guidelines from the HIV Medicine Association and Infectious Disease Society of America (IDSA)/Adult Aids Clinical Trials Group recommend pravastatin or atorvastatin as initial therapy for elevated LDL-C to avoid potential drug-drug interactions with PIs mediated through the CYP450 enzyme system.123  Gemfibrozil or fenofibrate are recommended when triglyceride concentrations are greater than 500 mg/dl.123In higher risk patients, switching to a treatment regimen without a PI is another option.

 

Direct Acting Antivirals

 

Regimens of direct acting antivirals (DAAs) have vastly changed the treatment of chronic hepatitis C (CHC) since the approval of sofosbuvir in 2014. The DAAs have shown to be more effective than previous standard of care (interferon-based treatments such as pegylated interferon and ribavirin) in producing sustained virologic response (SVR) of ≥90% and are associated with fewer side effects.128There are currently four classes of DAAs, defined by their mechanism of action and therapeutic target (NS3/4A inhibitors, protease inhibitors [PIs], NS5B inhibitors and NS5A inhibitors).128  Recommended regimens today consist of multiple antivirals that target different sites to improve efficacy and decrease resistance rates.

 

While the hepatitis C virus itself can impact lipid metabolism, treatment with a DAA regimen has been associated with short term increases in LDL-C, TC, and HDL-C. 129There do not appear to be any significant effects on TG’s.  This effect may result from a cancellation of the suppressive effect from HCV viral replication or from direct pharmacologic activity of the DAA’s themselves.130  The magnitude of effect does seem to vary based on DAA regimen with an increase in LDL-C of up to 27% reported.  More studies are needed to elucidate the exact mechanism of action and which regimens have the greatest impact.

 

Since treatment duration is defined and short term (12-24 weeks), it is unlikely that these changes will negatively impact long term CV health. However, it is important to acknowledge that many of these agents have pharmacokinetic drug-drug interactions with statins.  Clinicians may elect to temporarily hold statin therapy which may also contribute to changes in the lipid profile during the treatment period.

 

 

INTERFERONS

 

Interferons are associated with a wide range of systemic complications including neuropsychiatric changes, fatigue, and bone marrow suppression. Although metabolic side effects are less frequent, α interferon is known to inhibit lipoprotein lipase, stimulate hepatic lipogenesis and is associated with an increase in TG.131,132A cohort study of patients with chronic hepatitis C on treatment with various forms α interferon were evaluated for changes in TG and TC.132   Overall, mean serum TG levels rose 40% at 12 weeks and returned to baseline by 24 weeks after stopping therapy. However, the effect on individual patients was variable and 41 patients (27%) experienced TGs that more than doubled from baseline. There was no significant change in TC noted. The long-term complications of this have not been evaluated and no patients in the study developed acute pancreatitis. It appears that any significant clinical consequence from this rise in TG is extremely rare. There did not seem to be a difference in change in levels based on the specific form of α interferon that was used, including the long-acting PEGylated forms.

 

As the landscape for the treatment of chronic hepatitis C transitions to interferon-free regimens with DAAs, this may be irrelevant in these patients. However, this effect on TG has been seen in the treatment with interferons for other illnesses, including chronic myelogenous leukemia and other cancers.133,134It has recently been elucidated that interleukin-10 plays a key role in the linkage between inflammation and lipoprotein metabolism and that patients with cancer or other autoimmune diseases associated with elevated interleukin-10 levels can present with markedly decreased HDL-C, low LDL-C and elevated TG. 135Patients receiving interferon treatment for the treatment of cancer can be considered for anti-dyslipidemic medications to manage secondary hypertriglyceridemia.136

 

OTHER DRUGS THAT AFFECT LIPOPROTEIN LEVELS

 

Various other drugs have been reported to affect lipid and/or lipoprotein levels (Table 4). Lipid changes from these drugs are based on limited data, are reported inconsistently, and could be due to other disease related aspects. Thus, the effects on serum lipid levels cannot fully be

substantiated.

 

Table 4. Other Drugs that Affect Lipid and/or Lipoproteins
Antacids
Ascorbic Acid
Aspirin
Cimetidine/ranitidine
Cyclophosphamide
Interferons
Ketoconazole
L-asparaginase
Neomycin
Aminosalicylid acid

 

 

MANAGEMENT

 

Several medication classes or individual medications can affect the lipid profile, both positively and negatively. Risk factors include elevated lipid levels at baseline and high cardiovascular risk patients. Identifying potential medications as the cause of these changes and monitoring the lipid profile while on therapy can provide value to the care of the patient. However, the long-term implications of these drugs on ASCVD mortality and morbidity remains unknown and there is limited evidence on the overall impact of these drug-induced changes.

 

A thorough risk-benefit analysis of each treatment should be done based on individual patient factors. In general, if negative changes in the lipid profile are observed during therapy, replacement with an equivalent alternative therapy can be recommended. If no equivalent therapy is available and treatment must be initiated, then monitoring of serum lipid levels is vital. The use of existing guidelines for the management of dyslipidemia in the general population can be referred to and in extreme cases; the use of the suspected medication should be reassessed.

 

REFERENCES

 

  1. Feingold KR, Grunfeld C. Approach to the Patient with Dyslipidemia. In: De Groot LJ, Chrousos G, Dungan K, et al., eds. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2018.
  2. Vodnala D, Rubenfire M, Brook RD. Secondary causes of dyslipidemia. The American journal of cardiology. 2012;110(6):823-825.
  3. Henkin Y, Como JA, Oberman A. Secondary dyslipidemia. Inadvertent effects of drugs in clinical practice. Jama. 1992;267(7):961-968.
  4. Jacobson TA, Ito MK, Maki KC, et al. National Lipid Association recommendations for patient-centered management of dyslipidemia: part 1 - executive summary. J Clin Lipidol. 2014;8(5):473-488.
  5. Patni N, Li X, Adams-Huet B, Garg A. The prevalence and etiology of extreme hypertriglyceridemia in children: Data from a tertiary children's hospital. J Clin Lipidol. 2018;12(2):305-310.
  6. Sakhri J, Ben Salem C, Harbi H, Fathallah N, Ltaief R. Severe acute pancreatitis due to tamoxifen-induced hypertriglyceridemia with positive rechallenge. JOP : Journal of the pancreas. 2010;11(4):382-384.
  7. Wadood A, Chesner R, Mirza M, Zaman S. Tamoxifen precipitation of familial hypertriglyceridaemia: a rare cause of acute pancreatitis. BMJ case reports. 2016;2016.
  8. Tziomalos K, Athyros VG, Karagiannis A, Mikhailidis DP. Dyslipidemia induced by drugs used for the prevention and treatment of vascular diseases. The open cardiovascular medicine journal. 2011;5:85-89.
  9. Mantel-Teeuwisse AK, Kloosterman JM, Maitland-van der Zee AH, Klungel OH, Porsius AJ, de Boer A. Drug-Induced lipid changes: a review of the unintended effects of some commonly used drugs on serum lipid levels. Drug safety. 2001;24(6):443-456.
  10. Ames RP. The effects of antihypertensive drugs on serum lipids and lipoproteins. II. Non-diuretic drugs. Drugs. 1986;32(4):335-357.
  11. Krone W, Nagele H. Effects of antihypertensives on plasma lipids and lipoprotein metabolism.Am Heart J. 1988;116(6 Pt 2):1729-1734.
  12. Ott SM, LaCroix AZ, Ichikawa LE, Scholes D, Barlow WE. Effect of low-dose thiazide diuretics on plasma lipids: results from a double-blind, randomized clinical trial in older men and women. Journal of the American Geriatrics Society. 2003;51(3):340-347.
  13. Ozbilen S, Eren MA, Turan MN, Sabuncu T. The impact of carvedilol and metoprolol on serum lipid concentrations and symptoms in patients with hyperthyroidism. Endocrine research. 2012;37(3):117-123.
  14. Weidmann P, Ferrier C, Saxenhofer H, Uehlinger DE, Trost BN. Serum lipoproteins during treatment with antihypertensive drugs. Drugs.1988;35 Suppl 6:118-134.
  15. Weir MR, Moser M. Diuretics and beta-blockers: is there a risk for dyslipidemia? Am Heart J. 2000;139(1 Pt 1):174-183.
  16. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension (Dallas, Tex : 1979). 2017.
  17. Ferrari P, Rosman J, Weidmann P. Antihypertensive agents, serum lipoproteins and glucose metabolism. The American journal of cardiology. 1991;67(10):26b-35b.
  18. Kasiske BL, Ma JZ, Kalil RS, Louis TA. Effects of antihypertensive therapy on serum lipids. Annals of internal medicine. 1995;122(2):133-141.
  19. Lakshman MR, Reda DJ, Materson BJ, Cushman WC, Freis ED. Diuretics and beta-blockers do not have adverse effects at 1 year on plasma lipid and lipoprotein profiles in men with hypertension. Department of Veterans Affairs Cooperative Study Group on Antihypertensive Agents. Archives of internal medicine. 1999;159(6):551-558.
  20. van der Heijden M, Donders SH, Cleophas TJ, et al. A randomized, placebo-controlled study of loop diuretics in patients with essential hypertension: the bumetanide and furosemide on lipid profile (BUFUL) clinical study report. Journal of clinical pharmacology. 1998;38(7):630-635.
  21. Campbell N, Brant R, Stalts H, Stone J, Mahallati H. Fluctuations in blood lipid levels during furosemide therapy: a randomized, double-blind, placebo-controlled crossover study. Archives of internal medicine. 1998;158(13):1461-1463.
  22. Neutel JM. Metabolic manifestations of low-dose diuretics. The American journal of medicine. 1996;101(3a):71s-82s.
  23. Del-Aguila JL, Beitelshees AL, Cooper-Dehoff RM, et al. Genome-wide association analyses suggest NELL1 influences adverse metabolic response to HCTZ in African Americans. The pharmacogenomics journal. 2014;14(1):35-40.
  24. Howes LG, Lykos D, Rennie GC. Effects of antihypertensive drugs on coronary artery disease risk: a meta-analysis. Clinical and experimental pharmacology & physiology. 1996;23(6-7):555-558.
  25. Bell DS, Bakris GL, McGill JB. Comparison of carvedilol and metoprolol on serum lipid concentration in diabetic hypertensive patients. Diabetes, obesity & metabolism. 2009;11(3):234-238.
  26. Ripley TL, Saseen JJ. beta-blockers: a review of their pharmacological and physiological diversity in hypertension. The Annals of pharmacotherapy. 2014;48(6):723-733.
  27. Giugliano D, Acampora R, Marfella R, et al. Metabolic and cardiovascular effects of carvedilol and atenolol in non-insulin-dependent diabetes mellitus and hypertension. A randomized, controlled trial. Annals of internal medicine. 1997;126(12):955-959.
  28. Goto Y. Effects of alpha- and beta-blocker antihypertensive therapy on blood lipids: a multicenter trial. The American journal of medicine. 1984;76(2a):72-78.
  29. Reid JL, Elliott HL, Vincent J, Meredith PA. Clinical pharmacology of selective alpha blockers. Hemodynamics and effects on lipid levels. The American journal of medicine. 1987;82(1a):15-20.
  30. Al-Sarraf A, Li M, Frohlich J. Statin resistant dyslipidemia in a patient treated with amiodarone. BMJ case reports. 2011;2011.
  31. Politi A, Poggio G, Margiotta A. Can amiodarone induce hyperglycaemia and hypertriglyceridaemia? British medical journal (Clinical research ed). 1984;288(6413):285.
  32. Hudig F, Bakker O, Wiersinga WM. Amiodarone-induced hypercholesterolemia is associated with a decrease in liver LDL receptor mRNA. FEBS letters. 1994;341(1):86-90.
  33. Hudig F, Bakker O, Wiersinga WM. Amiodarone decreases gene expression of low-density lipoprotein receptor at both the mRNA and the protein level. Metabolism: clinical and experimental. 1998;47(9):1052-1057.
  34. Lakhdar AA, Farish E, Hillis WS, Dunn FG. Long-term amiodarone therapy raises serum cholesterol. European journal of clinical pharmacology. 1991;40(5):477-480.
  35. Wiersinga WM, Trip MD, van Beeren MH, Plomp TA, Oosting H. An increase in plasma cholesterol independent of thyroid function during long-term amiodarone therapy. A dose-dependent relationship. Annals of internal medicine. 1991;114(2):128-132.
  36. Yanai H, Hakoshima M, Adachi H, et al. Effects of Six Kinds of Sodium-Glucose Cotransporter 2 Inhibitors on Metabolic Parameters, and Summarized Effect and Its Correlations With Baseline Data. Journal of clinical medicine research. 2017;9(7):605-612.
  37. Cha SA, Park YM, Yun JS, et al. A comparison of effects of DPP-4 inhibitor and SGLT2 inhibitor on lipid profile in patients with type 2 diabetes. Lipids in health and disease. 2017;16(1):58.
  38. Feingold KR, Grunfeld C. Role Of Glucose And Lipids In The Cardiovascular Disease Of Patients With Diabetes. In: De Groot LJ, Chrousos G, Dungan K, et al., eds. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2018.
  39. Invokana® [package insert]. Titusville, NJ: Janssen Pharmaceuticals. 2017.
  40. Farxiga® [package insert]. Wilmington, DE. AstraZeneca Pharmaceuticals; 2017.
  41. Jardiance® [package insert]. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals; 2017.
  42. Steglatro® [package insert]. Whitehouse Station, NJ: Merck; 2017.
  43. Barton M. Cholesterol and atherosclerosis: modulation by oestrogen. Current opinion in lipidology. 2013;24(3):214-220.
  44. Bradley DD, Wingerd J, Petitti DB, Krauss RM, Ramcharan S. Serum high-density-lipoprotein cholesterol in women using oral contraceptives, estrogens and progestins. The New England journal of medicine. 1978;299(1):17-20.
  45. Shlipak MG, Simon JA, Vittinghoff E, et al. Estrogen and progestin, lipoprotein(a), and the risk of recurrent coronary heart disease events after menopause. Jama. 2000;283(14):1845-1852.
  46. Goldenberg NM, Wang P, Glueck CJ. An observational study of severe hypertriglyceridemia, hypertriglyceridemic acute pancreatitis, and failure of triglyceride-lowering therapy when estrogens are given to women with and without familial hypertriglyceridemia. Clinica chimica acta; international journal of clinical chemistry. 2003;332(1-2):11-19.
  47. Sitruk-Ware R, Nath A. Characteristics and metabolic effects of estrogen and progestins contained in oral contraceptive pills. Best practice & research Clinical endocrinology & metabolism. 2013;27(1):13-24.
  48. Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123(20):2292-2333.
  49. Ogita H, Node K, Kitakaze M. The role of estrogen and estrogen-related drugs in cardiovascular diseases. Current drug metabolism. 2003;4(6):497-504.
  50. ACOG practice bulletin. No. 73: Use of hormonal contraception in women with coexisting medical conditions. Obstetrics and gynecology. 2006;107(6):1453-1472.
  51. Sitruk-Ware R, Nath A. Metabolic effects of contraceptive steroids. Reviews in endocrine & metabolic disorders. 2011;12(2):63-75.
  52. Godsland IF, Crook D, Simpson R, et al. The effects of different formulations of oral contraceptive agents on lipid and carbohydrate metabolism. The New England journal of medicine. 1990;323(20):1375-1381.
  53. Berenson AB, Rahman M, Wilkinson G. Effect of injectable and oral contraceptives on serum lipids. Obstetrics and gynecology. 2009;114(4):786-794.
  54. Artac M, Sari R, Altunbas H, Karayalcin U. Asymptomatic acute pancreatitis due to tamoxifen-induced severe hypertriglyceridemia in a patient with diabetes mellitus and breast cancer. Journal of chemotherapy (Florence, Italy). 2002;14(3):309-311.
  55. Singh HK, Prasad MS, Kandasamy AK, Dharanipragada K. Tamoxifen-induced hypertriglyceridemia causing acute pancreatitis. Journal of pharmacology & pharmacotherapeutics. 2016;7(1):38-40.
  56. Marjoribanks J, Farquhar CM, Roberts H, Lethaby A. Cochrane corner: long-term hormone therapy for perimenopausal and postmenopausal women. Heart (British Cardiac Society). 2018;104(2):93-95.
  57. Manson JE, Chlebowski RT, Stefanick ML, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. Jama. 2013;310(13):1353-1368.
  58. Main C, Knight B, Moxham T, et al. Hormone therapy for preventing cardiovascular disease in post-menopausal women. The Cochrane database of systematic reviews. 2013(4):Cd002229.
  59. Danazol® [Prescribing Information]. New York, NY: Sanofi-Synthelabo Inc; 2003.
  60. Birjmohun RS, Kees Hovingh G, Stroes ES, et al. Effects of short-term and long-term danazol treatment on lipoproteins, coagulation, and progression of atherosclerosis: two clinical trials in healthy volunteers and patients with hereditary angioedema. Clinical therapeutics. 2008;30(12):2314-2323.
  61. Packard CJ, Shepherd J. Action of danazol on plasma lipids and lipoprotein metabolism. Acta obstetricia et gynecologica Scandinavica Supplement. 1994;159:35-40.
  62. Allen JK, Fraser IS. Cholesterol, high density lipoprotein and danazol. The Journal of clinical endocrinology and metabolism. 1981;53(1):149-152.
  63. Fahraeus L, Larsson-Cohn U, Ljungberg S, Wallentin L. Plasma lipoproteins during and after danazol treatment. Acta obstetricia et gynecologica Scandinavica Supplement. 1984;123:133-135.
  64. Fahraeus L, Larsson-Cohn U, Ljungberg S, Wallentin L. Profound alterations of the lipoprotein metabolism during danazol treatment in premenopausal women. Fertility and sterility. 1984;42(1):52-57.
  65. Telimaa S, Penttila I, Puolakka J, Ronnberg L, Kauppila A. Circulating lipid and lipoprotein concentrations during danazol and high-dose medroxyprogesterone acetate therapy of endometriosis. Fertility and sterility. 1989;52(1):31-35.
  66. Szeplaki G, Varga L, Valentin S, et al. Adverse effects of danazol prophylaxis on the lipid profiles of patients with hereditary angioedema. The Journal of allergy and clinical immunology. 2005;115(4):864-869.
  67. Szegedi R, Szeplaki G, Varga L, et al. Long-term danazol prophylaxis does not lead to increased carotid intima-media thickness in hereditary angioedema patients. Atherosclerosis. 2008;198(1):184-191.
  68. Baldo-Enzi G, Giada F, Zuliani G, et al. Lipid and apoprotein modifications in body builders during and after self-administration of anabolic steroids. Metabolism: clinical and experimental. 1990;39(2):203-208.
  69. Haffner SM, Kushwaha RS, Foster DM, Applebaum-Bowden D, Hazzard WR. Studies on the metabolic mechanism of reduced high density lipoproteins during anabolic steroid therapy. Metabolism: clinical and experimental. 1983;32(4):413-420.
  70. Hartgens F, Rietjens G, Keizer HA, Kuipers H, Wolffenbuttel BH. Effects of androgenic-anabolic steroids on apolipoproteins and lipoprotein (a). British journal of sports medicine. 2004;38(3):253-259.
  71. Webb OL, Laskarzewski PM, Glueck CJ. Severe depression of high-density lipoprotein cholesterol levels in weight lifters and body builders by self-administered exogenous testosterone and anabolic-androgenic steroids. Metabolism: clinical and experimental. 1984;33(11):971-975.
  72. Achar S, Rostamian A, Narayan SM. Cardiac and metabolic effects of anabolic-androgenic steroid abuse on lipids, blood pressure, left ventricular dimensions, and rhythm. The American journal of cardiology. 2010;106(6):893-901.
  73. Saylor PJ, Smith MR. Metabolic complications of androgen deprivation therapy for prostate cancer. The Journal of urology. 2013;189(1 Suppl):S34-42; discussion S43-34.
  74. Salvador C, Planas J, Agreda F, et al. Analysis of the lipid profile and atherogenic risk during androgen deprivation therapy in prostate cancer patients. Urologia internationalis. 2013;90(1):41-44.
  75. O'Farrell S, Garmo H, Holmberg L, Adolfsson J, Stattin P, Van Hemelrijck M. Risk and timing of cardiovascular disease after androgen-deprivation therapy in men with prostate cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(11):1243-1251.
  76. US Food and Drug Administration: FDA drug safety communication: Update to ongoing safety review of GnRH agonists and notification to manufacturers of GNRH agonists to add new safety information to labelling regarding increased risk of diabetes and certain cardiovascular diseases. http:// www.fda.gov/Drugs/DrugSafety/ucm229986.htm.
  77. Nguyen PL, Je Y, Schutz FA, et al. Association of androgen deprivation therapy with cardiovascular death in patients with prostate cancer: a meta-analysis of randomized trials. Jama. 2011;306(21):2359-2366.
  78. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC medicine. 2013;11:108.
  79. Finkle WD, Greenland S, Ridgeway GK, et al. Increased risk of non-fatal myocardial infarction following testosterone therapy prescription in men. PloS one. 2014;9(1):e85805.
  80. Rubinow KB, Vaisar T, Tang C, Matsumoto AM, Heinecke JW, Page ST. Testosterone replacement in hypogonadal men alters the HDL proteome but not HDL cholesterol efflux capacity. Journal of lipid research. 2012;53(7):1376-1383.
  81. Ruige JB, Ouwens DM, Kaufman JM. Beneficial and adverse effects of testosterone on the cardiovascular system in men. The Journal of clinical endocrinology and metabolism. 2013;98(11):4300-4310.
  82. Snyder PJ, Peachey H, Berlin JA, et al. Effect of transdermal testosterone treatment on serum lipid and apolipoprotein levels in men more than 65 years of age. The American journal of medicine. 2001;111(4):255-260.
  83. Diez JJ, Cordido F. [Benefits and risks of growth hormone in adults with growth hormone deficiency]. Medicina clinica. 2014;143(8):354-359.
  84. Elbornsson M, Gotherstrom G, Bosaeus I, Bengtsson BA, Johannsson G, Svensson J. Fifteen years of GH replacement improves body composition and cardiovascular risk factors. European journal of endocrinology. 2013;168(5):745-753.
  85. Johannsson G, Oscarsson J, Rosen T, et al. Effects of 1 year of growth hormone therapy on serum lipoprotein levels in growth hormone-deficient adults. Influence of gender and Apo(a) and ApoE phenotypes. Arteriosclerosis, thrombosis, and vascular biology. 1995;15(12):2142-2150.
  86. Rosen T, Johannsson G, Johansson JO, Bengtsson BA. Consequences of growth hormone deficiency in adults and the benefits and risks of recombinant human growth hormone treatment. A review paper. Hormone research. 1995;43(1-3):93-99.
  87. Russell-Jones DL, Watts GF, Weissberger A, et al. The effect of growth hormone replacement on serum lipids, lipoproteins, apolipoproteins and cholesterol precursors in adult growth hormone deficient patients. Clinical endocrinology. 1994;41(3):345-350.
  88. Ziegler TR. Growth hormone treatment improves serum lipids and lipoproteins in adults with growth hormone deficiency. JPEN Journal of parenteral and enteral nutrition. 1994;18(6):558-559.
  89. Leese GP, Wallymahmed M, VanHeyningen C, Tames F, Wieringa G, MacFarlane IA. HDL-cholesterol reductions associated with adult growth hormone replacement. Clinical endocrinology. 1998;49(5):673-677.
  90. Barbosa EJ, Glad CA, Nilsson AG, et al. Genotypes associated with lipid metabolism contribute to differences in serum lipid profile of GH-deficient adults before and after GH replacement therapy. European journal of endocrinology. 2012;167(3):353-362.
  91. Bershad S, Rubinstein A, Paterniti JR, et al. Changes in plasma lipids and lipoproteins during isotretinoin therapy for acne. The New England journal of medicine. 1985;313(16):981-985.
  92. Stoll D, Binnert C, Mooser V, Tappy L. Short-term administration of isotretinoin elevates plasma triglyceride concentrations without affecting insulin sensitivity in healthy humans. Metabolism: clinical and experimental. 2004;53(1):4-10.
  93. Davies PJ, Berry SA, Shipley GL, et al. Metabolic effects of rexinoids: tissue-specific regulation of lipoprotein lipase activity. Molecular pharmacology. 2001;59(2):170-176.
  94. Vu-Dac N, Gervois P, Torra IP, et al. Retinoids increase human apo C-III expression at the transcriptional level via the retinoid X receptor. Contribution to the hypertriglyceridemic action of retinoids. The Journal of clinical investigation. 1998;102(3):625-632.
  95. McLane J. Analysis of common side effects of isotretinoin. Journal of the American Academy of Dermatology. 2001;45(5):S188-194.
  96. Chaggar PS, Shaw SM, Williams SG. Effect of antipsychotic medications on glucose and lipid levels. Journal of clinical pharmacology. 2011;51(5):631-638.
  97. Koro CE, Meyer JM. Atypical antipsychotic therapy and hyperlipidemia: a review. Essential psychopharmacology. 2005;6(3):148-157.
  98. Melkersson K, Dahl ML. Adverse metabolic effects associated with atypical antipsychotics: literature review and clinical implications. Drugs. 2004;64(7):701-723.
  99. Meyer JM. Effects of atypical antipsychotics on weight and serum lipid levels. The Journal of clinical psychiatry. 2001;62 Suppl 27:27-34; discussion 40-21.
  100. Atmaca M, Kuloglu M, Tezcan E, Ustundag B. Serum leptin and triglyceride levels in patients on treatment with atypical antipsychotics. The Journal of clinical psychiatry. 2003;64(5):598-604.
  101. Gaulin BD, Markowitz JS, Caley CF, Nesbitt LA, Dufresne RL. Clozapine-associated elevation in serum triglycerides. The American journal of psychiatry. 1999;156(8):1270-1272.
  102. Eiris J, Novo-Rodriguez MI, Del Rio M, Meseguer P, Del Rio MC, Castro-Gago M. The effects on lipid and apolipoprotein serum levels of long-term carbamazepine, valproic acid and phenobarbital therapy in children with epilepsy. Epilepsy research. 2000;41(1):1-7.
  103. Jakubus T, Michalska-Jakubus M, Lukawski K, Janowska A, Czuczwar SJ. Atherosclerotic risk among children taking antiepileptic drugs. Pharmacological reports : PR. 2009;61(3):411-423.
  104. Arnaldi G, Scandali VM, Trementino L, Cardinaletti M, Appolloni G, Boscaro M. Pathophysiology of dyslipidemia in Cushing's syndrome. Neuroendocrinology. 2010;92 Suppl 1:86-90.
  105. Jefferys DB, Lessof MH, Mattock MB. Corticosteroid treatment, serum lipids and coronary artery disease. Postgraduate medical journal. 1980;56(657):491-493.
  106. Choi HK, Seeger JD. Glucocorticoid use and serum lipid levels in US adults: the Third National Health and Nutrition Examination Survey. Arthritis and rheumatism. 2005;53(4):528-535.
  107. el-Shaboury AH, Hayes TM. Hyperlipidaemia in asthmatic patients receiving long-term steroid therapy. British medical journal. 1973;2(5858):85-86.
  108. McKay LI, Cidlowski JA. Physiologic and Pharmacologic Effects of Corticosteroids. 2003.http://www.ncbi.nlm.nih.gov/books/NBK13780/. Accessed February 16, 2015.
  109. Ettinger WH, Goldberg AP, Applebaum-Bowden D, Hazzard WR. Dyslipoproteinemia in systemic lupus erythematosus. Effect of corticosteroids. The American journal of medicine. 1987;83(3):503-508.
  110. Kockx M, Kritharides L. Cyclosporin A-Induced Hyperlipidemia. In: Kostner G, ed. Lipoproteins - Role in Health and Diseases. InTech; 2012. http://www.intechopen.com/books/lipoproteins-role-in-health-and-diseases/cyclosporin-a-induced-hyperlipidemia. Accessed January 2, 2015.
  111. Taylor DO, Barr ML, Radovancevic B, et al. A randomized, multicenter comparison of tacrolimus and cyclosporine immunosuppressive regimens in cardiac transplantation: decreased hyperlipidemia and hypertension with tacrolimus. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 1999;18(4):336-345.
  112. Kuster GM, Drexel H, Bleisch JA, et al. Relation of cyclosporine blood levels to adverse effects on lipoproteins. Transplantation.1994;57(10):1479-1483.
  113. Fazal MA, Idrees MK, Akhtar SF. Dyslipidaemia among renal transplant recipients: cyclosporine versus tacrolimus. JPMA The Journal of the Pakistan Medical Association. 2014;64(5):496-499.
  114. McCune TR, Thacker LR, II, Peters TG, et al. Effects of tacrolimus on hyperlipidemia after successful renal transplantation: a Southeastern Organ Procurement Foundation multicenter clinical study. Transplantation.1998;65(1):87-92.
  115. Seymen P, Yildiz M, Turkmen MF, Titiz MI, Seymen HO. Effects of cyclosporine-tacrolimus switching in posttransplantation hyperlipidemia on high-density lipoprotein 2/3, lipoprotein a1/b, and other lipid parameters. Transplantation proceedings. 2009;41(10):4181-4183.
  116. Holdaas H, Julian D. The use of statins after solid organ transplantation. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2002;17(8):1537.
  117. Wanner C, Bartens W, Galle J. Clinical utility of antilipidemic therapies in chronic renal allograft failure. Kidney international Supplement. 1995;52:S60-62.
  118. Christians U, Jacobsen W, Floren LC. Metabolism and drug interactions of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors in transplant patients: are the statins mechanistically similar? Pharmacology & therapeutics. 1998;80(1):1-34.
  119. Holdaas H, Fellstrom B, Jardine AG, et al. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet (London, England). 2003;361(9374):2024-2031.
  120. Fernandez-Montero JV, Barreiro P, Soriano V. HIV protease inhibitors: recent clinical trials and recommendations on use. Expert opinion on pharmacotherapy. 2009;10(10):1615-1629.
  121. Carr A, Samaras K, Burton S, et al. A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS (London, England). 1998;12(7):F51-58.
  122. Spector AA. HIV protease inhibitors and hyperlipidemia: a fatty acid connection. Arteriosclerosis, thrombosis, and vascular biology. 2006;26(1):7-9.
  123. Dube MP, Stein JH, Aberg JA, et al. Guidelines for the evaluation and management of dyslipidemia in human immunodeficiency virus (HIV)-infected adults receiving antiretroviral therapy: recommendations of the HIV Medical Association of the Infectious Disease Society of America and the Adult AIDS Clinical Trials Group.Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2003;37(5):613-627.
  124. Berthold HK, Parhofer KG, Ritter MM, et al. Influence of protease inhibitor therapy on lipoprotein metabolism. Journal of internal medicine. 1999;246(6):567-575.
  125. Markowitz M, Saag M, Powderly WG, et al. A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection. The New England journal of medicine. 1995;333(23):1534-1539.
  126. Mirete G, Masia M, Gutierrez F, Mora A, Escolano C, Maestre A. Acute pancreatitis as a complication of ritonavir therapy in a patient with AIDS. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology. 1998;17(11):810-811.
  127. Penzak SR, Chuck SK. Management of protease inhibitor-associated hyperlipidemia. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2002;2(2):91-106.
  128. Jakobsen JC, Nielsen EE, Feinberg J, et al. Direct-acting antivirals for chronic hepatitis C. The Cochrane database of systematic reviews. 2017;9:Cd012143.
  129. Endo D, Satoh K, Shimada N, Hokari A, Aizawa Y. Impact of interferon-free antivirus therapy on lipid profiles in patients with chronic hepatitis C genotype 1b. World journal of gastroenterology. 2017;23(13):2355-2364.
  130. Kanda T, Moriyama M. Direct-acting antiviral agents against hepatitis C virus and lipid metabolism. World journal of gastroenterology. 2017;23(31):5645-5649.
  131. Borden EC, Rosenzweig IB, Byrne GI. Interferons: from virus inhibitor to modulator of amino acid and lipid metabolism. Journal of interferon research. 1987;7(5):591-596.
  132. Naeem M, Bacon BR, Mistry B, Britton RS, Di Bisceglie AM. Changes in serum lipoprotein profile during interferon therapy in chronic hepatitis C. The American journal of gastroenterology. 2001;96(8):2468-2472.
  133. Kurzrock R, Rohde MF, Quesada JR, et al. Recombinant gamma interferon induces hypertriglyceridemia and inhibits post-heparin lipase activity in cancer patients. The Journal of experimental medicine. 1986;164(4):1093-1101.
  134. Penarrubia MJ, Steegmann JL, Lavilla E, et al. Hypertriglyceridemia may be severe in CML patients treated with interferon-alpha. American journal of hematology. 1995;49(3):240-241.
  135. Moraitis AG, Freeman LA, Shamburek RD, et al. Elevated interleukin-10: a new cause of dyslipidemia leading to severe HDL deficiency. J Clin Lipidol. 2015;9(1):81-90.
  136. Wong SF, Jakowatz JG, Taheri R. Management of hypertriglyceridemia in patients receiving interferon for malignant melanoma. The Annals of pharmacotherapy. 2004;38(10):1655-1659.

 

Postpartum Thyroiditis

CLINICAL RECOGNITION

 

Postpartum thyroiditis is the term used for patients who develop painless thyroiditis in the postpartum period. It occurs within 6 months (typically 2 to 4 months) after delivery and runs a clinical course identical to that of painless thyroiditis occurring without relation to pregnancy. The clinical course of thyroid dysfunction is similar to subacute thyroiditis but with no anterior neck pain or tenderness of the thyroid. The prevalence of postpartum thyroiditis ranges from 3 to 8 per cent of all pregnancies. Most women with subclinical autoimmune thyroiditis and antithyroid microsomal antibodies of more than 1:5120 before pregnancy develop postpartum thyroiditis. After delivery, other forms of autoimmune thyroid dysfunction may also occur, including Graves' disease, transient hypothyroidism without preceding destructive thyrotoxicosis, and persistent hypothyroidism (Fig. 1).

Fig. 1 Various clinical courses of thyroid dysfunction after delivery. A typical form of postpartum thyroiditis is “(III) Destructive thyrotoxicosis”. RAIU, radioactive iodine uptake.

 

The typical course is characterized by three sequential phases: the thyrotoxic, the hypothyroid and recovery phase. The thyrotoxic phase occurs 1–3 months after parturition and lasts for a few months, followed by hypothyroidism at 3–6 months after delivery. Finally, normal thyroid function is usually achieved within a year. Most patients have a complete remission, but some develop persistent hypothyroidism.

 

PATHOPHYSIOLOGY

 

This disorder is believed to be an autoimmune disorder and is characterized by lymphocytic infiltration of the thyroid gland and by transient thyrotoxicosis followed by hypothyroidism or by one or the other occurring in the first year after parturition. An immune rebound mechanism has been established for the induction of postpartum thyroiditis. Thyrotoxicosis is induced by leakage of intrathyroidal hormones into the circulation caused by damage to thyroid epithelial cells from inflammation. The early phase of thyrotoxicosis in postpartum thyroiditis can be classified as destruction-induced thyrotoxicosis or simply as destructive thyrotoxicosis.

 

DIAGNOSIS AND DIFFERENTIAL

 

Diagnostic Tests Needed and Suggested

 

Serum TSH is suppressed, associated with an increase in serum FT3 and FT4 levels. The thyroid radioactive iodine uptake (RAIU) is low. When the measurement of RAIU is difficult due to nursing, the measurement of anti-TSH receptor antibody and/or thyroid blood flow by ultrasonography may be useful to differentiate between destruction-induced thyrotoxicosis and Graves' thyrotoxicosis. Positive TPOAb or TGAb indicate the autoimmune nature of the disease.

 

THERAPY

 

If symptoms or signs of thyrotoxicosis are severe, beta-blocker drugs can be administered for the duration of the thyrotoxic phase. Propylthiouracil can be used to inhibit conversion of thyroxine to triiodothyronine. Obviously, RAI treatment is excluded since RAIU is suppressed in the toxic phase. During the hypothyroid phase, L-T4 at replacement doses is recommended.

 

FOLLOW-UP

 

Patients who have developed permanent hypothyroidism should be treated with replacement doses of L-T4. All patients should be monitored for thyroid function test at their next pregnancy, delivery and postpartum.

.

REFERENCES

 

  1. Akamizu T, Amino N. Hashimoto’s Thyroiditis. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2017 Jul 17.
  2. Lazarus J, Okosieme OE. Hypothyroidism in Pregnancy. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2015 Apr 12.
  3. Roti E, Uberti E. Post-partum thyroiditis--a clinical update. Eur J Endocrinol. 2002 Mar;146(3):275-9.
  4. Azizi F, Amouzegar A. Management of hyperthyroidism during pregnancy and lactation. Eur J Endocrinol. 2011 Jun;164(6):871-6

Lipodystrophy Syndromes: Presentation and Treatment

ABSTRACT

 

Lipodystrophy syndromes are a heterogeneous group of diseases, characterized by selective absence of adipose tissue. In one sense, these diseases are lipid-partitioning disorders, where the primary defect is the loss of functional adipocytes, leading to ectopic steatosis, severe dyslipidemia and insulin resistance. These syndromes have attracted significant attention since the mid-1990s as the understanding of adipose tissue biology grew, initially spurred by the discovery of the pathways leading to adipocyte differentiation and maturation, and then by the discovery of leptin. Although lipodystrophy syndromes are known since the beginning of the 20th century, significant progress in understanding these syndromes were made in the last two decades, placing these syndromes at the forefront of the translational metabolism field.  Currently, more than 15 distinctive molecular etiologies have been attributed to cause human diseases most of which map to adipocyte differentiation or lipid droplet pathways. Seemingly acquired syndromes are recently reported to have a genetic basis, suggesting that our “pre-genome” understanding of the syndromes was inadequate and that we need to likely change our classification schemes. Regardless of the etiology, it is the selective absence of adipose tissue and the reduced ability to store long-term energy that perturbs insulin sensitivity and lipid metabolism. The treatment of these syndromes has also attracted considerable interest. The most successful example of the treatment of these syndromes came from the demonstration that leptin replacement strategy improved insulin resistance and dyslipidemia in the most severely affected forms of the disease, leading to an FDA approved therapy for generalized lipodystrophy syndromes. In the partial forms of the disease, the phenotypes are more complex and the efficacy of leptin is not as uniform. Currently, numerous trials are in progress for the development of potential new treatments for the partial forms of the disease. These rare metabolic diseases are likely to fuel novel breakthroughs in the metabolism field in the foreseeable future. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

 

INTRODUCTION

 

Lipodystrophy syndromes comprise a heterogeneous group of disorders characterized by either generalized or partial lack of adipose tissue depending on the type of lipodystrophy 1,2. Lipodystrophy classically has been classified as congenital or acquired. Patients with partial lipodystrophy may exhibit excess adipose tissue accumulation in other areas of the body. Lipodystrophy syndromes usually manifest with several metabolic abnormalities associated with severe insulin resistance that include diabetes mellitus, hypertriglyceridemia, and hepatic steatosis which can progress to steatohepatitis. Other common manifestations are acanthosis nigricans, polycystic ovarian syndrome (PCOS), and eruptive xanthomas (due to severe hypertriglyceridemia) 3,4. Metabolic derangements are mostly responsible for the serious comorbidities associated with lipodystrophy; some of which are chronic complications of poorly controlled diabetes, acute pancreatitis, hepatic cirrhosis, proteinuria and renal failure, and premature cardiovascular disease (Fig.1) 1,2. Typically, standard treatments fail to achieve good glycemic control in most patients with lipodystrophy, although episodes of diabetic ketoacidosis have been rarely reported5. The severity of the comorbidities depends on the subtype, extent of fat loss, and other clinical characteristics such as gender and age. Major causes of mortality are cardiovascular diseases 6-9, liver diseases2,10, acute pancreatitis 2, renal failure 10, and sepsis 3. Clinical characteristics of lipodystrophy are shown in Table 1. It is important to note that there are additional components of the disease that may be specific to each molecular etiology. In addition, we are beginning to recognize that patients often report reduced quality of life with increased overall pain (requiring frequent use of pain medications), sleep disturbances and sleep apnea, gastrointestinal dysmotility, mood disturbances such as depression and anxiety and psychiatric diseases 11,12.

Figure 1. Consequences of Lipodystrophy: The figure summarizes metabolic derangements and end-organ complications in patients with lipodystrophy (Left: MRI showing near total lack of adipose tissue; Right top: Liver biopsy shows hepatic steatosis in lipodystrophy (Hematoxylin and eosin staining; magnification 100X), Right bottom: CT of abdomen obtained during an episode of acute pancreatitis in lipodystrophy; Middle top: Diabetic foot ulcer in a patient with generalized lipodystrophy; Middle bottom: Renal biopsy specimens (Left: Electron microscopy image reveals lipid vacuoles which suggest ectopic lipid accumulation; Right: Light microscopy image documents chronic kidney disease in lipodystrophy (Hematoxylin and eosin staining; magnification 40X).

Table 1: Clinical Characteristics That Raise Suspicion for Lipodystrophy

Loss or absence of adipose tissue in a partial or generalized fashion
Disproportionate hyperphagia (inability to stop eating, waking up to eat, fighting for food)
Muscle hypertrophy and prominent veins (phlebomegaly)
Cushingoid appearance
Pseudo-acromegaloid appearance
Progeroid appearance
Acanthosis nigricans (associated with insulin resistance)
Proteinuria, renal dysfunction
Reproductive dysfunction (reduced fertility, hyperandrogenism, oligomenorrhea, hirsutism and/or polycystic ovaries)
Musculoskeletal abnormalities (occasionally)
Cardiomyopathy (occasionally)
Low intelligence (occasionally)

Metabolic abnormalities

·       Relatively early onset of insulin resistant diabetes which is severe in some patients with requirement for high doses of insulin, e.g., requiring ≥200 U/day, ≥2 U/kg/day, or U-500 insulin, early development of complications

·       Dyslipidemia which is characterized by elevated triglycerides and low HDL cholesterol. Hypertriglyceridemia can be very severe (≥500 mg/dL) and is unresponsive to treatment with associated history of acute pancreatitis

·       Hepatomegaly and/or elevated transaminases in the absence of a known cause of liver disease (e.g., viral hepatitis). Hepatic steatosis (e.g. radiologic evidence), Hepatomegaly, non-alcoholic steatohepatitis (NASH), cirrhosis.

 

Lipodystrophy is an exciting rare disease that helps us obtain a better understanding of the pathophysiology of metabolic abnormalities associated with insulin resistance. The main cause of insulin resistance in lipodystrophy is the fact that the excess energy cannot be stored in adipose tissue, which is secondary to either the near total lack of adipocyte expandability in patients with generalized lipodystrophy or a limited capacity to expand in partial lipodystrophy. Limited lipid storage capacity causes the failure of buffering postprandial lipids and secreting substantial adipokines, which in turn results in excessive levels of triglycerides and lipid intermediates in circulation. The body stores fat at ectopic sites such as the liver as a result of inability to store energy in the subcutaneous adipose depots (Fig.1). Levels of adipokines and hormones secreted from the adipose tissue, most characteristically leptin, are decreased in these patients 1,2,9,13,14. Leptin has a fundamental role in glucose and lipid homeostasis. Leptin is the key hormone responsible for regulating appetite 15. Low levels of leptin in lipodystrophy trigger hyperphagia, which is often extreme 16-18. Leptin protects pancreatic beta cells from lipotoxicity at least in rodent models. Leptin improves insulin sensitivity by increasing glucose uptake in peripheral tissues such as muscle via sympathetic nervous system activation. Leptin also decreases hepatic gluconeogenesis 19-21.

 

ANIMAL MODELS OF LIPODYSTROPHY

 

Several animal models of lipodystrophy have shown that adipose tissue dysfunction triggers the development of severe insulin resistance, which is associated with metabolic abnormalities and end-organ complications as mentioned above and also shown in Fig.1. An extensive and authoritative review of these studies can be found in an article by Dr. David B. Savage 22. The introduction of these animal models has allowed researchers to explore the fundamental characteristics of lipodystrophy and insulin resistance and allowed studies of the effects of different treatment approaches. Regardless of the strategy used, ablation of white adipose tissue led to the development of insulin resistance, hypertriglyceridemia, and hepatic steatosis (sometimes 6-fold elevation in total liver weight). In now classical experiments of Reitman and colleagues, fat transplantation from littermates rescued metabolic derangements in the famous A-ZIP mice 23-25. Dr. Beutler’s group recently identified kelch repeat and BTB (POZ) domain containing 2 (KBTBD2) deficiency as a cause of lipodystrophy associated with insulin resistance and diabetes and they also showed that transplantation of wild-type adipose tissue rescued diabetes and the hepatic steatosis phenotypes of Kbtbd2−/−mice 26. The infusion of leptin into aP2–SREBP-1c transgenic mice from the Brown and Goldstein laboratory resulted in dramatic benefits in glycemic parameters, insulin action, and hepatic steatosis, which could not be explained by its effect on food intake alone, providing the premise to undertake leptin replacement in human patients 27. What was also striking was that if fat from the leptin deficient obese mice was transplanted into littermates of the A-ZIP mice, the metabolic rescue was far less effective, suggesting that leptin played an important role in the regulation of metabolism in lipodystrophy in rodents 28. The replacement of deficient leptin in a small but severely affected cohort of human patients with lipodystrophy with recombinant human leptin (metreleptin) was first reported in 2002 and attracted further attention to lipodystrophy research 29. Longer-term studies subsequently confirmed the role of metreleptin therapy in lipodystrophy syndromes especially in the most severe forms 30-32.

 

DIAGNOSIS

 

The diagnosis of lipodystrophy is usually made clinically based on history, body distribution of adipose tissue, physical examination, and metabolic profile. Lipodystrophy should be suspected in any person with partial or complete lack of subcutaneous adipose tissue. However, the diagnosis of lipodystrophy is often delayed because of the rarity of these syndromes and the failure of the physicians to recognize this disease. Although patients with congenital generalized lipodystrophy lack subcutaneous adipose tissue from birth, specific diagnosis is usually made during childhood or even adulthood when they start developing metabolic abnormalities. This is at least partly because of the fact that the awareness of lipodystrophy is still low among physicians. The problem of recognition is much more common for partial lipodystrophy. The distribution of fat loss varies in different types of partial lipodystrophy. At first glance, certain types of partial lipodystrophy cannot be clearly distinguished from other common metabolic diseases (e.g. poorly controlled diabetes mellitus with truncal obesity) based on phenotype unless the physicians is suspicious for lipodystrophy and checks carefully for certain characteristic such as the appearance of the limbs which look thinner than in a normal person. Also, the onset of fat loss may be gradual and delay the diagnosis both in genetic and acquired forms 3,4. Lipodystrophy syndromes should be considered in the differential diagnosis in patients with relatively early onset insulin resistant diabetes mellitus, persistent hypertriglyceridemia, hepatic steatosis, PCOS and hepatosplenomegaly. Other diseases that should be considered in the differential diagnosis of lipodystrophy are listed in Table 2.

 

Table 2: Differential Diagnosis of Lipodystrophy Syndromes

Generalized Lipodystrophy Syndromes Uncontrolled diabetes mellitus
HIV-associated wasting
Anorexia nervosa, cachexia and starvation
Chronic infections
Adrenocortical insufficiency
Thyrotoxicosis
Diencephalic syndrome
Partial Lipodystrophy Syndromes Cushing’s syndrome
Truncal obesity
Multiple symmetric lipomatosis
Progeroid syndromes
Acromegaly/gigantism

 

A thorough physical examination is required for clinical diagnosis of lipodystrophy. Clinicians should pay specific attention to evaluating the extremities and the gluteal region for leanness and muscularity. In addition, other body parts should be examined for accumulation of excessive amounts of fat. Due to marked abdominal obesity and excessive fat accumulation in the neck, patients with familial partial lipodystrophy (FPLD) may be misdiagnosed as Cushing’s syndrome 2. In the genetic forms of lipodystrophy, parental consanguinity and the mode of inheritance should be questioned 2.

 

The absence of subcutaneous fat can be quantified by using conventional anthropometric measurements, dual energy x-ray absorptiometry (DXA) scan, whole-body magnetic resonance imaging (MRI), and computed tomography (CT) scan 4.  Anthropometry including skinfold thickness and limb circumference measurements are easy and affordable ways to estimate the fat loss and redistribution 9. For the facial fat loss, serial photography may be used to evaluate the gradual loss of facial fat. DXA, MRI, and CT scans are non-invasive modalities that may be used for quantification of fat on a tissue-specific basis, but at least in the United States, none are covered for this purpose by insurance companies 3,4,33.

 

Laboratory testing is a valuable tool for physicians to support the diagnosis. If the physical phenotype is not recognized, hyperglycemia, insulin resistance and severe hypertriglyceridemia that is non-responsive to therapy may provide important clues for the diagnosis. When fat loss is not confirmed by the physical examination or by an imaging modality, hyperglycemia and hypertriglyceridemia that are resistant or unresponsive to conventional treatment may serve as surrogate indicators to the clinician that a patient may have lipodystrophy. Lipodystrophy should be suspected in patients requiring ≥200 units/day (≥2 units/kg/day) of insulin or triglyceride levels that remain persistently elevated (≥500 mg/dL) despite fully optimized therapy and diet modifications. All patients except those with localized lipodystrophy, should be tested for blood glucose levels, glycated hemoglobin (HbA1c), serum lipids (especially triglyceride levels), and liver function tests on the initial evaluation and during subsequent encounters. In addition to these laboratory evaluations, leptin levels may be used in support of the diagnosis. However, it should be noted that low leptin levels may be observed in other conditions such as hypothalamic amenorrhea and malnutrition. Thus, low leptin level is not specific for the diagnosis lipodystrophy 4,30. Circulating adiponectin though not a clinically available test, may be helpful in differentiating patients with generalized lipodystrophy from those who have constitutional leanness, fat loss due to calorie imbalance or excessive exercise as well as poorly controlled diabetes mellitus with insulin deficiency. In all of the cases except lipodystrophy, adiponectin levels will be normal or even higher than normal whereas in lipodystrophy including familial partial lipodystrophy, serum adiponectin levels are usually low.

 

Genetic testing is available for several genes in certain clinical and research laboratories. Because additional loci for genetic lipodystrophy syndromes are presumed to be present, negative genetic tests do not rule out a genetic condition.

 

Lipodystrophy syndromes can be classified as genetic or acquired. However, they are simply classified as generalized and partial in the clinical practice most of the time (Table 3).

 

 

Table 3: Classification of Lipodystrophy Syndromes

 

Type

 

Lipodystrophy Phenotype

 

Subtype

(Genes Involved)

 

Key Clinical Features

 

 

Congenital Generalized Lipodystrophy (CGL)

 

Near total absence of the body fat starting at birth or shortly after, generalized muscularity, metabolic abnormalities

CGL1 (AGPAT2)

Autosomal recessive

Loss of metabolically active fat with sparing of mechanically functioning fat

CGL2 (BSCL2)

Autosomal recessive

Generalized absence of adipose tissue

CGL3 (CAV1)

Autosomal recessive

Short stature, vitamin D resistance, hypocalcemia, hypomagnesemia

CGL4 (PTRF)

Autosomal recessive

Myopathy, skeletal abnormalities, pyloric stenosis and gastrointestinal motility problems, cardiac arrhythmias

 

Acquired Generalized Lipodystrophy (AGL)

Near total absence of the body fat commonly develops during childhood or adolescence, metabolic abnormalities Autoimmune AGL follows an autoimmune disease; e.g. JDM
Panniculitis-associated Tender subcutaneous nodules that herald the onset of AGL
Idiopathic No history of auto-immune disease or panniculitis

 

 

 

 

 

 

 

 

Familial Partial Lipodystrophy (FPLD)

 

 

 

 

 

 

 

 

Loss of fat from the limbs, metabolic abnormalities

FPLD1, Kobberling (Unknown) Loss of subcutaneous fat from the limbs, although they usually have truncal obesity. Palpable “ledge” formation between the normal and lipodystrophic areas

FPLD2, Dunnigan (LMNA)

Autosomal dominant

Increased muscularity and loss of fat in the limbs, excess fat accumulation in the face and neck

FPLD3 (PPARG)

Autosomal dominant

Loss of subcutaneous fat from the limbs, specifically distally

FPLD4 (PLIN1)

Autosomal dominant

Loss of subcutaneous fat from the limbs, histologically; small adipocytes, macrophage infiltration and fibrosis of adipose tissue

FPLD5 (CIDEC)

Autosomal recessive

Loss of subcutaneous fat from the limbs, small, multilocular lipid droplets in adipocytes

 

FPLD6 (LIPE)

Autosomal recessive

Increased visceral fat, dyslipidemia, hepatosteatosis, insulin resistance, and diabetes, some may present with muscular dystrophy and elevated serum creatine phosphokinase
Progeria associated lipodystrophy   LMNA, ZMPSTE24, POLD1, WRN, FBN1, BANF1, KCNJ6, SPRTN Progeroid features
Other genes associated with lipodystrophy   AKT2, PCYT1A, PIK3R1, MFN2, PSMB8, ADRA2A Various presentations of lipodystrophy

 

 

 

Acquired Partial Lipodystrophy (APL)

Loss of subcutaneous fat starts from the face, neck, upper extremities, and progresses to the trunk. Lower limbs are typically spared, some patients have excess fat over the gluteal region, thighs and calves

 

Autoimmune

Coinciding autoimmune disorders; dermatomyositis/polymyositis and SLE are most commonly associated disorders

 

MPGN-associated

Low serum complement 3, glomerulonephritis, hematuria, urinary casts, proteinuria, nephritic syndrome, renal failure
Idiopathic No history of auto-immune disease or MPGN

 

It is important to note that this classical method of disease classification will likely become inadequate as more disease-causing genes and pathways are identified.

 

GENERALIZED LIPODYSTROPHY SYNDROMES

 

Generalized lipodystrophy syndromes are rare disorders that are either inherited (Berardinelli-Seip Syndrome) 31,32,34or acquired (Lawrence Syndrome) 9.

 

Congenital Generalized Lipodystrophy

 

Congenital Generalized Lipodystrophy (CGL) or Berardinelli-Seip syndrome is a rare syndrome which manifests with near total absence of adipose tissue. It is inherited in an autosomal recessive manner. Fat loss is usually recognized shortly after birth or in the first years of life, although patients may be diagnosed later during teenage years or adulthood. There have been over 300 reported cases to date 13,35,36.

 

In addition to lack of subcutaneous fat, patients may present with hepatomegaly and umbilical protuberance during infancy. Extensive acanthosis nigricans and prominent musculature may also contribute to the striking phenotype of these patients 37. Affected females may have irregular menstrual cycles, oligomenorrhea, cliteromegaly, and hirsutism. Premature menarche and pubarche are also rarely seen. Most males were reported to be fertile whereas only a few females had successful pregnancies 38. Sperm abnormalities have been reported in CGL similar to BSCL2 knock-out mice that also exhibit male sterility 39. Other clinical manifestations include advanced bone age, mild mental retardation, cardiomyopathy and cardiac rhythm disturbances 40.

 

Children with CGL usually have a voracious appetite and accelerated growth. Basal metabolic rate may be increased. Hypertriglyceridemia usually presents with high levels of chylomicrons and very low-density lipoproteins (VLDL) and reduced levels of high density lipoproteins (HDL). Severe hypertriglyceridemia usually results in recurrent acute pancreatitis. Insulin resistance commonly results in diabetes in adolescence or later. Diabetes is rarely responsive to insulin therapy. Serum leptin levels are very low 30.

The genetic defect can be determined in vast majority of patients with CGL. There are at least four molecularly distinct types of CGL. Of note, several patients with CGL have been reported who do not possess any pathogenic variant in any of the following four genes.

 

CONGENITAL GENERALIZED LIPODYSTROPHY TYPE 1 (CGL1)

 

1-acylglycerol-3-phophate O-acyltransferase 2 (AGPAT2), a key enzyme in triglyceride synthesis, is deficient in CGL1. AGPAT2gene is located on chromosome 9q34. AGPAT2 catalyzes the acylation of lysophosphaditic acid to form phosphaditic acid, a key intermediate in the biosynthesis of triglyceride and glycerophospholipids 41. Precisely how AGPAT2 deficiency causes lipodystrophy remains unsolved, but possible mechanisms include impaired lipogenesis, altered differentiation of preadipocytes to adipocytes, altering normal activation of phosphatidylinositol 3-kinase (PI3K)/Akt and PPARγpathways in the early stages of adipogenesis, and apoptosis/necrosis of adipocytes 2,42,43. Adiposity is preserved in certain body parts such as orbits, palms and soles, which constitute the mechanically adipose tissue 30,44-46(Fig.2). AGPAT2pathogenic variants along with BSCL2pathogenic variants are responsible for the majority of the CGL cases.

Figure 2: Near total absence of adipose tissue in CGL1 (2A, 2C, 2D). Magnetic resonance images document the lack of subcutaneous fat (2B). Liver biopsy reveals severe hepatic steatosis with both micro and macrovesicular steatosis (Hematoxylin and eosin staining; magnification 200X), 2E).

CONGENITAL GENERALIZED LIPODYSTROPHY TYPE 2 (CGL2)

 

CGL2 is caused by pathogenic variants in the BSCL2gene which have been mapped to chromosome 11q13. This gene encodes a 398-amino acid integral endoplasmic reticulum membrane protein calledseipin47. This protein is assumed to take part in lipid droplet formation and adipocyte differentiation 48,49. Patients with BSCL2pathogenic variants have the most severe disease and are born without any adipose tissue. Hypertriglyceridemia and hepatic steatosis can be detected in early childhood; and hepatic involvement can be more severe in CGL2 than other subtypes 50. Intellectual disability and cardiomyopathy are more common than in CGL1. CGL2 patients are also distinguished from the CGL1 patients with the loss of mechanical adipose tissue 51(Fig.3). Although the mechanism is not clear, adiponectin levels are relatively higher in patients with CGL2 despite severely suppressed leptin levels which can help in the differential diagnosis 52.

Figure 3. Near total absence of adipose tissue in a patient with CGL2 (3A, 3B). Also note that the patient shown now deceased was only 29 years old at the time the picture was taken, suggesting the possibility of accelerated aging.

CONGENITAL GENERALIZED LIPODYSTROPHY TYPE 3 (CGL3)

 

CGL3 is caused by pathogenic variants in the CAV1gene which are located on chromosome 7q31 9,14,53. This gene encodes the protein caveolin-1, which is an integral part of caveolaefound in plasma membranes. Caveolin 1 binds fatty acids on the plasma membranes and translocates them into lipid droplets. Mutated caveolin 1 disrupts lipid droplet formation and adipocyte differentiation 54. CGL3 is distinguished from other CGLs by the presence of unique features such as preserved bone marrow fat, vitamin D resistance, hypocalcemia, hypomagnesemia, and decreased bone density 40. In addition to this classical presentation, whole exome sequencing has identified de novoheterozygous null CAV1pathogenic variants in two patients of European origin with generalized fat loss, thin mottled skin, and progeroid features at birth; however, no differences in the number and morphology of caveolae have been found in dermal fibroblasts 55, which suggests that this observation needs to be confirmed in further pedigrees. Heterozygous CAV1frameshift mutations have also been reported to be associated with partial lipodystrophy (Fig.4). Several features such as congenital cataracts and cerebellar progressive ataxia were also present 56.

 

Figure 4. Partial lipodystrophy associated with heterozygous CAV1 frameshift mutations in a male (4A, 4B, 4C) and female subject (4D, 4E). Patients shown were a father and a daughter pair.

CONGENITAL GENERALIZED LIPODYSTROPHY TYPE 4 (CGL4)

 

Type 4 CGL (CGL4) is caused by pathogenic variants in the PTRFgene. The product of this gene, cavin, is a polymerase 1 and transcript release factor which regulates caveolae 1 and 3 57. CGL4 can be recognized by distinct clinical characteristics. This rare subtype of CGL is associated with myopathy, pyloric stenosis, gastrointestinal dysmotility, arrhythmias that include exercise-induced ventricular tachycardia and sudden death, and skeletal abnormalities such as atlantoaxial instability and scoliosis 58-60(Fig.5).

Figure 5. Lack of subcutaneous fat (5A), scoliosis (5A), gastrointestinal dysmotility (5B), and exercise-induced ventricular arrhythmia (5C) in CGL4.

OTHER GENES ASSOCIATED WITH GENERALIZED LIPODYSTROPHY

 

Biallellic loss-of-function pathogenic variants in phosphate cytidylyltransferase 1 alpha (PCYT1A), the rate-limiting enzyme in the Kennedy pathway of de novo phosphatidylcholine synthesis, have been reported to be associated with generalized lipodystrophy, severe hepatic steatosis and low HDL cholesterol levels 61. Although widely involved in the familial partial lipodystrophy pathogenesis, several pathogenic variants in the LMNAand PPARGgenes have been associated with generalized lipodystrophy. Recently, heterozygous LMNAp.T10I pathogenic variant was reported to be associated with generalized lipodystrophy, diabetes mellitus, acanthosis nigricans, hypertriglyceridemia, and hepatomegaly (Fig.6) 62. Biallelic pathogenic variants at PPARGhas also been reported to cause generalized lipodystrophy 63.

Figure 6. Generalized lack of subcutaneous fat (6A), eruptive xanthomata (6B), and lipemia retinalis (6C) secondary to severe hypertriglyceridemia in a patient with heterozygous LMNA p.T10I pathogenic variant.

Acquired Generalized Lipodystrophy

 

Acquired generalized lipodystrophy (AGL), also known as Lawrence Syndrome, is very rare. Generalized fat loss is not present at birth but develops later in life. It occurs over a variable time period, ranging from a few weeks to years (Fig.7) 9.

 

Figure 7. Generalized loss of subcutaneous fat in two patients with AGL (7A-D). Note the distal fat loss around the feet as opposed to patients with CGL phenotypes.

Although the pathogenesis of AGL is unknown, it is hypothesized to be linked to autoimmune destruction of adipocytes. Autoantibodies against adipocyte membranes have been reported 64-66. AGL is associated with panniculitis in approximately 25% of the patients. This type may manifest with subcutaneous inflammatory nodules (panniculitis), which heal by localized loss of fat and eventually results in complete loss of subcutaneous fat 9. Another one fourth of the AGL patients present with an autoimmune disease that include juvenile dermatomyositis (JDM), Sjogren’s syndrome, rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus 9,65. Of these, JDM particularly correlates with AGL. 8-40% of patients with JDM develop AGL (Fig.8) 66-68. In the remaining 50% of the cases, AGL is not associated with any autoimmune or inflammatory condition 9. Some patients with AGL exhibit low serum complement 4 levels and auto-immune hepatitis, sometimes together with Type 1 diabetes, which suggests the involvement of classical complement pathway in AGL pathogenesis 69.

As mentioned above, it is of note that some of the patients with AGL are recently recognized to have additional progeroid features and may harbor a specific pathogenic of LMNAgene at position 10 (p.T10I). We have reported clinical presentations of these patients recently in a case series report. One of these patients also had biopsy proven juvenile dermatomyositis suggesting that the long-recognized association between AGL and JDM may be linked through distinctive molecular mechanisms 62.

 

In patients with AGL, as a result of generalized fat loss, metabolic abnormalities associated with severe insulin resistance that include hypertriglyceridemia, diabetes mellitus, hepatic steatosis, acanthosis nigricans, menstrual irregularities and PCOS may develop soon after the recognition of fat loss. Patients have suppressed levels of leptin and adiponectin 9,30.

 

Figure 8. Generalized loss of subcutaneous fat in a patient with juvenile dermatomyositis associated AGL (8A, 8B). Note the absence of muscle tissue as well in this severely affected patient.

PARTIAL LIPODYSTROPHY

 

Fat loss affects only part of the body in partial lipodystrophy. Partial lipodystrophy is categorized into inherited (familial partial lipodystrophy, FPLD) and acquired forms (acquired partial lipodystrophy, APL). Both patients with FPLD and APL start losing fat at some point during their life. Lower limbs are most frequently affected in FPLD. There might be accumulation of adipose tissue in the face and neck. On the other hand, APL is characterized by fat loss that spreads through a cephalocaudal distribution from the face, neck, shoulders, arms, and forearms and that extends to the thoracic region and upper abdomen. There are numerous genes associated with FPLD. Despite the growing number of proven genetic markers, about half of the patients do not have a discernible single gene variation.

 

Inherited Partial Lipodystrophy Syndromes

 

Patients with these syndromes usually notice partial fat loss around puberty. Fat loss pattern is very heterogeneous in patients with FPLD. Even among patients with pathogenic variants of the same gene, fat loss patterns may vary.

 

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 1 (FPLD1)

 

The loss of adipose tissue is mainly limited to the extremities in patients with FPLD1 or Kobberling-type lipodystrophy 70. There is a normal or slightly increased fat in the face and neck. Truncal obesity is a common finding. The hallmark of this syndrome is the formation of a palpable “ledge” between the normal and lipodystrophic areas 71. It is believed that women are diagnosed more easily as they usually present with a more severe disease. Metabolic complications usually develop in early adulthood. Insulin resistant diabetes and metabolic syndrome are common and may cause premature coronary artery disease. Hypertriglyceridemia may trigger episodes of acute pancreatitis. Acanthosis nigricans is commonly seen. Leptin levels are variable and correlate with body mass index (BMI) which suggests that the levels of leptin are appropriate for the fat content in FPLD1 71. The Cambridge group recently reported that this form of lipodystrophy may have a polygenic etiology 72. There is a remarkable phenotypical heterogeneity among patients with FPLD1. In this spectrum of FPLD1, patients with significant central obesity are likely polygenic. This type of presentation is relatively more common, and it is sometimes difficult to make a distinction between FPLD1 and truncal obesity complicated with metabolic syndrome. The use of radiological methods such as DXA, CT or MRI can help in this population to further define body fat distribution in addition to physical examination and skinfold measurements. On the other hand, some FPLD patients without increased truncal fat are classified as FPLD1 by definition, if no disease-causing gene is identified to date. These latter patients will likely turn out to have a monogenic form of FPLD eventually. These two different presentations of FPLD1 are shown in Fig.9.

Figure 9. Heterogeneity in FPLD1. Patient in A to D presented with decrease in peripheral fat depots and preservation of abdominal fat. Patient in E to H has increased abdominal adiposity. The formation of a palpable “ledge” between the normal and lipodystrophic areas is shown (9C and 9E). (Images E-H used with permission by Dr. Jonathan Q. Purnell from publication Diabetes Care 2003;26(6):1819-24)

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 2 (FPLD2)

 

FPLD2 or Dunnigan Variety lipodystrophy is an autosomal dominant syndrome which is characterized by gradual onset of subcutaneous fat loss from the extremities during puberty. Affected individuals have prominent muscularity in their extremities. Excess fat accumulates in the neck causing a buffalo hump (Fig.10). This phenotype sometimes can be misdiagnosed as Cushing’s syndrome at first glance 14. Pathogenic variants in the LMNAgene, which is located on chromosome 1q21-22, cause FPLD2. The LMNAgene codes nuclear lamina proteins, lamin A and C. Pathogenic variants in the LMNAgene can be scattered across many exons of the gene and are missense mutations 73. Mutant lamins disrupt the interaction between nuclear lamina and chromatin and may result in apoptosis, which may be  followed by premature adipocyte death 74.

Figure 10. Subcutaneous adipose tissue loss from the extremities, excess fat accumulation in the face and neck, and Cushingoid appearance in FPLD2 (10A-D; Note that one of the patients (10A) previously underwent liposuction for removal of unwanted excess fat from the neck).

Females have a more recognizable phenotype and more severe metabolic complications 75. Most patients with FPLD2 develop diabetes in their twenties and thirties. Other components of insulin resistance are usually present. Patients with FPLD2 are at high risk for cardiovascular diseases that usually develop at relatively younger ages 76.

 

There is phenotypic heterogeneity among patients with FPLD2. For instance, less severe loss of fat has been reported in patients with exon 11 LMNApathogenic variants which affects only lamin A protein 77. LMNAR349W pathogenic variant (exon 6) is associated with facial fat loss which is uncommon in FPLD2 76,78,79. Exon 1 variants are associated with severe cardiac disease that require cardiac transplant at an early age and may be coupled with arrhythmias and conduction system abnormalities. Variants across exon 4 through 8 have been noted to cause muscular dystrophy related symptoms together with fat distribution abnormalities. LMNAgene pathogenic variants are also involved in the pathogenesis of progeroid disorders including Hutchinson-Gilford progeria syndrome (HGPS), mandibuloacral dysplasia, and atypical progeroid syndrome (APS).

 

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 3 (FPLD3)

 

FPLD3 is caused by pathogenic variants in the PPARGgene, a key regulator of adipocyte differentiation. Patients with FPLD3 usually show milder fat loss; and there is no accumulation of adipose tissue in the face and neck (Fig.11); however, they manifest metabolic complications at a similar rate and severity to those with FPLD2 76,80-84.

Figure 11. Moderate partial subcutaneous adipose tissue loss in a patient with FPLD3 (11A-C).

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 4 (FPLD4)

 

FPLD4 is caused by pathogenic variants in the PLIN1gene encoding perilipin 1, which is an essential lipid droplet coat protein 85. Perilipin plays a key role in coordinating access of lipases to the core triacylglycerol. It is characterized by the loss of adipose tissue which is most striking in the lower limbs and femorogluteal depot, severe insulin resistance, diabetes, hypertriglyceridemia, and hepatic steatosis 86,87.

 

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 5 (FPLD5)

 

FPLD5 is an autosomal recessive syndrome caused by pathogenic variants in the CIDECgene. It is characterized by partial lipodystrophy, acanthosis nigricans, severe insulin resistance leading to diabetes, and hepatic steatosis. The CIDECgene is located on chromosome 3 (3p25.3) and encodes the CIDEC protein, which is expressed in the lipid droplets. Pathogenic variants of the CIDECgene are postulated to result in the loss of ability of lipid droplets to store fat 88.

 

FAMILIAL PARTIAL LIPODYSTROPHY TYPE 6 (FPLD6)

 

FPLD type 6 is caused by pathogenic variants in the LIPE(lipase E, hormone sensitive type) gene which has an autosomal recessive inheritance 89. This FPLD subtype is characterized by late-onset partial fat loss from the lower extremities and also multiple symmetric lipomatosis and progressive distal symmetric myopathy at least in some cases 89,90. Hormone sensitive lipase is the predominant regulator of lipolysis from adipocytes. Pathogenic variants in the LIPEgene appear to result in impaired lipolysis which may induce lipomatosis and partial fat loss at the same time that is associated with hypertriglyceridemia, hepatic steatosis, and insulin resistant diabetes 90.

 

OTHER GENES ASSOCIATED WITH FAMILIAL PARTIAL LIPODYSTROPHY

 

FPLD has been reported to be caused by pathogenic variants in the AKT2gene 91. AKT is a serine/threonine protein kinase, which is involved in cell signaling/growth, glycogen synthesis, and insulin-stimulated glucose transport. Lipodystrophy in patients with AKT2mutations is thought to be due to defective adipocyte differentiation and post-receptor insulin signaling 92. Exome sequencing has identified a heterozygous variant in the adrenoceptor α 2A (ADRA2A) gene, which encodes the main presynaptic inhibitory feedback G protein–coupled receptor regulating norepinephrine release, in an African-American pedigree with atypical FPLD 93, which needs to be confirmed in additional pedigrees.

 

Progeroid Syndromes And Lipodystrophy

 

Mandibuloacral Dysplasia (MAD) is a rare progeroid syndrome which manifests with craniofacial, skeletal and cutaneous abnormalities and lipodystrophy (Fig.12) 94. The clinical manifestations present gradually over time, most commonly during childhood. There are two types of MAD currently recognized. Mandibuloacral dysplasia type A (MADA) is characterized by the loss of subcutaneous fat from the extremities along with normal or excessive fat in the face and the neck. Mandibuloacral dysplasia type B manifests with a more generalized loss of subcutaneous fat 94-97.

Figure 12. Hypoplasia of the mandible in a patient with Mandibuloacral Dysplasia.

MAD is caused by mutations in the LMNAgene which results in the accumulation of prelamin A protein 98. This, in return disrupts the interaction between nuclear lamina and chromatin 97-99. Compound heterozygous pathogenic variants in the zinc metalloproteinase (ZMPSTE24) gene have been reported to cause MADB associated lipodystrophy 100,101. ZMPSTE24is essential in the post-translational proteolytic cleavage of carboxy terminal residues of farnesylated prelamin A to form mature lamin A and vimentin processing 100,102,103.

 

MDP (mandibular hypoplasia, deafness and progeroid features syndrome) has been reported to be caused by pathogenic variants of thePOLD1gene that encodes catalytic subunit of DNA polymerase δ which play an essential role in the lagging-strand DNA synthesis during DNA replication 104. In addition to progressive lipodystrophy and severe insulin resistance, patients with MDP suffer from mandibular hypoplasia, sensorineural deafness, progeroid features, scleroderma and skin telangiectasia, ligament contractures, reduced mass of limb muscles, hypogonadism and undescended testes in males 104-107.  We recently observed a mother daughter pair with a different POLD1variant near the carboxyl terminal of the protein at a very highly conserved residue (Fig.13).

 

Figure 13. Partial lipodystrophy in a patient with POLD1 variant (13A-E).

Biallelic WRNnull mutations linked to partial lipodystrophy with severe insulin resistance in adult progeria Werner syndrome (Fig.14) 108. The WRNgene encodes a RecQ DNA helicase which plays a critical role in repairing damaged DNA 109.

Figure 14. Partial lipodystrophy in a patient with adult progeria also known as Werner syndrome.

Fibrillin-1 (FBN1) gene pathogenic variants are found in more than 90% of patients with Marfan syndrome 110. Pathogenic variants in the penultimate exon of FBN1have been reported to be associated with a distinct phenotype of generalized lipodystrophy that share some clinical features with neonatal progeroid syndrome (Wiedemann–Rautenstrauch syndrome), a very severe disorder with only a few patients described who could reach their late childhood 111-113. Although these patients have marfanoid/progeroid appearance, skeletal features, dilated aortic bulb, bilateral subluxation of the lens, myopia in addition to the severe generalized lipodystrophy, no significant metabolic abnormality caused by the lack of adipose tissue has been reported 111,112,114.

 

Pathogenic variants in BANF1have been reported to be associated with progeroid features, growth retardation, decreased subcutaneous fat, thin limbs, and stiff joints. This disease is also called Néstor-Guillermo progeria syndrome (NGPS) 115.

 

Heterozygous pathogenic variants in KCNJ6(GIRK2), which encodes an inwardly rectifying potassium channel, cause Keppen-Lubinsky syndrome that is characterized by severe developmental delay and intellectual disability, microcephaly, large prominent eyes, an open mouth, progeroid appearance, and generalized lipodystrophy 116.

 

Pathogenic variants of the Spartan (SPRTN) gene, which encodes a protein that is essential in the maintenance of genomic stability, have reported to be associated progeroid features, lipodystrophy and hepatocellular carcinoma 117.

 

Other Syndromes And Genes Associated With Lipodystrophy

 

Pathogenic variants in the phosphatidylinositol 3-kinase, regulatory subunit 1 (PIK3R1), which mediates insulin’s metabolic actions, have been reported in patients with SHORT syndrome (short stature, joint hyperextensibility, ocular depression, Rieger anomaly, and teething delay) that is associated with lipodystrophy in many patients 118,119. It has also been reported that patients with C-terminal PIK3R1pathogenic variants exhibit severe insulin resistance but normolipidemia and no hepatic steatosis 120.

 

Pathogenic variants in the proteasome subunit, beta-type, 8 (PSMB8) gene, which encodes a catalytic subunit of the 20S immunoproteasomes called β5i, has been linked to an autosomal-recessive autoinflammatory syndrome characterized by joint contractures, muscle atrophy, microcytic anemia, and panniculitis-induced lipodystrophy (JMP syndrome)121-123.

 

CANDLE syndrome is another rare autoinflammatory syndrome characterized by chronic atypical neutrophilic dermatitis, recurrent fever, and partial loss of adipose tissue from the upper limbs and face 124. An eponym for the syndrome was proposed as Nakajo–Nishimura syndrome 125,126.   Homozygous or compound heterozygous mutations in the gene PSMB8have been reported in patients with CANDLE syndrome 127,128.

 

Recently a pathogenic variant in the MFN2gene that encodes mitofusin 2, a membrane-bound mediator of mitochondrial membrane fusion and inter-organelle communication, have been reported to be associated with partial lipodystrophy, upper body adipose hyperplasia, and suppression of leptin expression 129(Fig.15).

Figure 15. Disease progression in a patient with a pathogenic variant in the MFN2 gene (15A-D).

Two families with AREDYLD syndrome that is characterized by acrorenal field defect, ectodermal dysplasia, generalized lipodystrophy, and multiple abnor

malities have been reported 130,131. The genetic basis of this very rare syndrome is still unknown.

 

Acquired Partial Lipodystrophy

 

Acquired partial lipodystrophy (APL) is characterized by fat loss typically starting in childhood or early adulthood. Loss of adipose tissue first manifests in the face and gradually progresses to the upper extremities, thorax and upper abdomen symmetrically. It typically proceeds in a cephalocaudal fashion but spares the lower extremities (Fig.16). There might be accumulation of fat in the lower abdomen, gluteal region, and lower extremities.

Figure 16. Typical cephalocaudal adipose tissue loss pattern in two patients with APL (16A-D). Note preservation of fat depots below the waist line.

Although the etiology of APL is still unknown, some patients may have coinciding autoimmune conditions. Systemic lupus erythematosus and dermatomyositis/polymyositis are among the most frequently associated auto-immune diseases 132. APL has been associated with abnormalities of the alternative complement pathway that may cause membranoproliferative glomerulonephritis (MPGN) 133. Subsequent chronic renal disease constitutes the major cause of morbidity in these patients. It has been suggested that C3-nephritic factor might be the cause for the lysis of adipocytes expressing factor D, although there is no solid evidence supporting this hypothesis 134.

 

Rare variants in LMNB2were previously reported in five patients with APL, but two of four variants were also present in normal controls 135. In addition, subcutaneous loss of fat from the legs and the gluteal region, presence of diabetes, type IV and V hyperlipoproteinemias were atypical presentations in these patients 135.

 

Metabolic complications are less common compared to other types of lipodystrophy syndromes 4. Not all patients develop insulin resistance, diabetes, or hypertriglyceridemia. Leptin levels vary from hypoleptinemia to normal range 30,132. However, patients may develop metabolic abnormalities such as diabetes, hypertriglyceridemia, low HDL cholesterol levels and hepatic steatosis in later stages of the disorder. In addition, several patients with APL has been reported to develop diabetes or other metabolic abnormalities at a relatively young age, which are apparently associated with insulin resistance 136. Thus, patients with APL should also be followed for metabolic abnormalities as is done for other subtypes of lipodystrophy.

 

TREATMENT

 

Currently, treatment modalities are restricted to ameliorating or preventing the comorbidities of the lipodystrophic syndromes. There is no cure for these syndromes. For the metabolic disturbances, lifestyle modification (diet and exercise as needed), metformin, and fibrates (and/or statins) are generally required. Insulin or other antidiabetics (e.g., metformin, thiazolidinediones) can also be used if needed. Metreleptin, a leptin analog, is indicated as an adjunct to diet as replacement therapy to treat the complications of leptin deficiency in patients with generalized lipodystrophy.

 

Lifestyle Modification

 

There is limited knowledge on the effectiveness of diet and exercise in the management of metabolic disturbances in patients with lipodystrophy. In general, a balanced macronutrient composition is recommended. In patients with severe hypertriglyceridemia, a balanced low- fat diet (<15% of daily caloric intake) is appropriate. To control diabetes, increased physical activity and carbohydrate restriction are advised. Dietary fiber intake and foods that are rich in omega-3 fatty acids are suggested 3.

 

Most patients with lipodystrophy are encouraged to be physically active. In patients with cardiomyopathy and cardiac arrhythmias strenuous exercise should be avoided. Patients with CGL4 should avoid exercise as they may develop exercise-induced ventricular arrhythmias 57,60. Contact sports are not advised to patients with severe hepatosplenomegaly and CGL patients presenting with lytic bone lesions.

 

Patients should abstain from drinking alcohol due to the risk of developing acute pancreatitis and non-alcoholic steatohepatitis (NASH). Patients should also be advised to avoid smoking and maintain an optimal blood pressure to decrease the risk of cardiovascular disease.

 

Insulin Resistance

 

In patients presenting with lipodystrophy and diabetes, both metformin and thiazolidinediones are somewhat effective to treat hyperglycemia and hyperlipidemia 137-141. Metformin is used as the first-line agent in insulin resistant diabetes. Thiazolidinediones may improve the metabolic profile in partial lipodystrophy syndromes 141. The very first thiazolidinedione to be approved in the United States troglitazone actually worked remarkably well in lowering both HbA1c and triglyceride levels in a cohort of patients with predominantly partial lipodystrophy syndromes. However, data on the currently approved thiazolidinediones are limited and contradictory 139,142,143. Thiazolidinediones should be considered in the management of diabetes in patients with partial lipodystrophy, however they should not be routinely used in generalized lipodystrophy as their efficacy has not been studied 3,141. Insulin is usually needed in very high doses and concentrated forms, such as U-500. Patients with extreme insulin resistance, however, may not respond to concentrated insulin. Administration of insulin-like growth factor-1 (IGF-1) has been shown to be effective in maintaining glycemic control and insulin resistance in short-term studies, as well as in type 2 diabetes 144-146. Many other hypoglycemic agents have been used in lipodystrophy, but their efficacy has not been studied 3.

 

Dyslipidemia

 

Statins are normally used as first-line agents to treat hypercholesterolemia but patients with FPLD have low tolerance to statins. Rosuvastatin and pravastatin have been proven to reduce total LDL cholesterol levels 147,148. Statins are used with caution to prevent side effects such as myopathy and hepatotoxicity. Along with diet, fibrates and fish oil rich in omega-3 fatty acids, should be prescribed for serum triglyceride levels >500 mg/dL and may be considered for triglycerides >200 mg/dL. Combining fibrates with statins has proved to be effective in dyslipidemia; however, there is an increased risk for muscle toxicity. Therapeutic apheresis is used in extreme hypertriglyceridemia to prevent recurrent episodes of acute pancreatitis 29.

 

Cosmetic Treatment

 

Cosmetic correction of lipoatrophy and fat excess is associated with improved quality of life in patients with lipodystrophy. Autologous adipose tissue transplantation, facial reconstruction with free flaps and silicone or other implants have been used in lipoatrophic areas. In addition, liposuction or surgical excision is used for removal of unwanted excess fat from body parts such as; the chin, buffalo hump and vulvar region.

 

Bariatric Surgery

 

Roux-en-Y Gastric Bypass Surgery (RYGB) is associated with effective weight loss and resolution of metabolic comorbidities in patients with obesity 149. RYGB was used with success in several patients with FPLD1 and with FPLD2 150-152. RYGB resulted in weight loss and significant improvements in metabolic parameters in patients with FPLD1 that allowed patients to stop using insulin 150. FPLD2 patients also benefited from RYBG. Substantial improvements in metabolic parameters and a significant weight loss were reported after the surgery 152,153.

 

Leptin

 

A large group of lipodystrophy patients present with low leptin levels. Metreleptin (r-metHuLeptin) is an analog of human leptin made through recombinant DNA technology. It has been tested in congenital and acquired forms of lipodystrophy and has been shown to ameliorate the metabolic derangements 29,34. Several studies show that leptin (0.04 to 0.08 mg/kg/day by subcutaneous injection) in patients with generalized lipodystrophy results in significant weight loss due to its effect on appetite and resting energy expenditure 34. Leptin replacement therapy is approved in Japan as a therapy indicated specifically for the treatment of diabetes and/or hypertriglyceridemia in patients with congenital or acquired lipodystrophy. In the United States, metreleptin, now called MYALEPT, has been approved by the FDA in 2014 for use in patients with congenital generalized or acquired generalized lipodystrophy for the treatment of metabolic complications of these diseases as an adjunct to diet and lifestyle modifications. There is no lower age limit for initiation of Myalept nor a specific degree of metabolic abnormality so long as the diagnosis of generalized lipodystrophy can be substantiated.  However, it is not approved for use in human immunodeficiency virus (HIV)-related lipodystrophy, or in patients with metabolic diseases such as diabetes and hypertriglyceridemia, or partial lipodystrophy. The effects of leptin treatment in patients with lipodystrophy are summarized below.

 

APPETITE

 

Metreleptin decreases hyperphagia, leading to weight loss that usually stabilize with long-term treatment 29,154-156. This effect can be noted by the patients right after the treatment with metreleptin. Functional MRI studies combined with behavioral assessments showed that metreleptin treatment is associated with long-term improvements of hedonic and homeostatic central nervous networks regulating appetite and food intake 157-159. Food related neural activity and formation of satiety feeling have been shown to be effectively restored by leptin replacement in lipodystrophy 160.

 

METABOLIC PARAMETERS

 

Metabolic changes become prominent in several weeks after metreleptin. Metreleptin therapy has been shown to improve fasting plasma glucose levels starting from the first week 151. In a subset of patients undergoing hyperinsulinemic-euglycemic clamp studies, leptin replacement therapy improved peripheral glucose disposal and decreased both hepatic glucose output and hepatic steatosis 161. Metreleptin lowered HbA1c by 2% within the first year 162. It is recommended to lower the insulin doses by 50% on initiation of metreleptin therapy to avoid hypoglycemia in well-controlled diabetic patients. Metreleptin treatment has no suppressive effect on beta cell function in patients with lipodystrophy 163. On the contrary it has been reported that metreleptin therapy improves insulin secretion in diabetic patients with lipodystrophy 164.

 

Metreleptin lowers triglycerides starting from the first week. It is effective in providing a 60% reduction in triglyceride levels within the first year 162. It should be noted that acute withdrawal of metreleptin therapy might result in acute pancreatitis episodes 165,166. Metreleptin also decreased total cholesterol and LDL-cholesterol levels but did not alter HDL cholesterol levels 165,167.

 

The beneficial effect of metreleptin on glycemic and lipid measures in generalized lipodystrophy are clear and usually dramatic. Although the response is variable in patients with partial lipodystrophy and it is not approved yet in this patient population, studies have shown that these patients can benefit from metreleptin treatment. A selected cohort of partial lipodystrophy patients with moderately to severely low leptin and significant baseline metabolic abnormalities is more likely to benefit from metreleptin therapy 12,162,168,169.

 

LIVER

 

Leptin replacement therapy improves hepatic steatosis and lowers serum transaminases within 6-12 months (Fig.17) 156,161,170. The liver volume decreases 155,170. Although the mechanism is not fully understood, leptin therapy resulted in significant increase in insulin suppression of hepatic glucose production 161. This improvement in insulin action helps reverse hepatic steatosis by decreasing triglyceride content 161. Nonalcoholic steatohepatitis (NASH) score has been reported to improve after metreleptin treatment and no progression in hepatic fibrosis has been reported 171. When treated at least for a year, the vast majority of patients showed improved liver histology, steatosis and hepatocyte ballooning, and only 33% of patients continued fulfilling the criteria for NASH after 1 year of treatment with metreleptin 172,173. A significant improvement in the non-alcoholic fatty liver disease (NAFLD) score has been reported after metreleptin treatment in pediatric patients who underwent liver biopsies 174. Metreleptin has also been reported to result in rapid clearance of fat from the liver and normalization of liver histology in an AGL patient with recurrence of NAFLD in the first few months of liver transplantation 175.

Figure 17. Liver histology shows regression of hepatic steatosis and ballooning injury after metreleptin treatment (left before metreleptin and right 4 months on metreleptin treatment, Hematoxylin and eosin staining; magnification 200X).

KIDNEYS

 

Patients with lipodystrophy may develop proteinuric kidney disease. Metreleptin decreased proteinuria in most patients 156,176. The reduction in proteinuria coincided with improvement in hyperfiltration in 11 of 15 patients treated with metreleptin. However, four patients had worsening renal function. Hence, renal functions should be closely monitored during metreleptin therapy 176.

 

REPRODUCTIVE SYSTEM

 

In females, metreleptin was found to normalize gonadotropin secretion. It led to normal progression of puberty, normalized menstrual cycles, and improved fertility 156,177-179. Leptin replacement improved low estradiol levels and corrected the attenuated luteinizing hormone (LH) response to luteinizing hormone-releasing hormone (LHRH) in young women with lipodystrophy and leptin deficiency 177. One-year treatment with metreleptin resulted a significant decrease in testosterone and sex hormone binding globulin (SHBG) levels in lipodystrophic women with PCOS 180. Several pregnancies have occurred in patients with lipodystrophy while they were on metreleptin without any evidence for teratogenicity 38,181, although it has not been approved for use in pregnancy. Leptin replacement was associated with a small increase (clinically non-significant) in serum testosterone and SHBG in males. No change was observed in serum LH response to LHRH 178. No impact of leptin therapy on bone mineral density and content and bone metabolism has been reported in both sexes 170,182,183.

 

ADVERSE EFFECTS

 

Approximately 30% of patients treated with metreleptin experienced adverse effects 165. The most common side effects were hypoglycemia, and injection site reactions e.g. erythema and urticaria. Headache, fatigue, weight loss, and abdominal pain were also seen. There may also be a need to make dose adjustments or cessation of concomitant treatments such as insulin, oral antidiabetics, and lipid lowering drugs after metreleptin therapy. In some cases, in vivo neutralizing antibodies to metreleptin have been reported 184,185and is the main reason underlying the FDA's restriction of metreleptin use. Anti-metreleptin antibodies developed in most patients with lipodystrophy; however, neutralizing activity concurrent with worsened metabolic control has been reported only in a small number of patients treated with metreleptin 181,184. In addition, in the few patients who presented with neutralizing antibody formation, occurrence of severe infections such as sepsis has been reported. Two of these patients developed multiple sepsis episodes around the time of detection of neutralizing antibody 184. T-cell lymphoma has been reported in three patients with acquired generalized lipodystrophy receiving metreleptin 186. In acquired lipodystrophy patients with autoimmunity and immunodeficiency before metreleptin therapy, T-cell lymphoma development was also described 186,187, suggesting that lymphoma development in acquired lipodystrophy is more likely to be associated with the disease itself rather than being related to metreleptin treatment.

 

In other cases with acquired generalized lipodystrophy, progression of kidney disease and liver disease have been observed while receiving metreleptin therapy 188. Since patients with AGL with distinct autoimmune conditions clearly benefit from metreleptin, treatment for their metabolic abnormalities should be considered in patients with AGL with close clinical follow up in light of the cautionary preclinical data 189,190.

 

More recently attention has been devoted to emergence of new cancers while on metreleptin therapy. Data on these parameters will be reported in short order.

 

Investigational Treatments For Lipodystrophy

 

Since in the United States partial lipodystrophy has been without medical therapy, a number of companies with interesting compounds have initiated development of their products for this indication. Current investigational treatments with registered studies in ClinicalTrials.gov are presented in Table 4. Data on these trials will be updated as results become available.

 

Table 4: Investigational Therapies For Lipodystrophy

 

Investigational agent

 

Status

 

Type of lipodystrophy

 

Primary outcome

Volanesorsen

(anti-sense oligonucleotide to apoC-III)

Active, not recruiting Familial partial lipodystrophy Change in fasting triglycerides

Obeticholic Acid

(farnesoid X receptor agonist)

Recruiting Familial partial lipodystrophy Change in liver triglycerides

Cholic Acid

(primary bile acid)

Active, not recruiting Various forms of lipodystrophy Reduction in liver triglyceride content

Setmelanotide

(melanocortin-4 receptor agonist)

Expanded access in a single patient Partial lipodystrophy associated with leptin deficiency Treatment of refractory hypertriglyceridemia leading to recurrent bouts of pancreatitis

Gemcabene

(monocalcium salt of a dialkyl ether dicarboxylic acid)

Recruiting Familial partial lipodystrophy Change in fasting triglycerides, hepatic steatosis

Baricitinib

(inhibitor of Janus kinases 1 and 2 “JAK1/2”)

Expanded access available Autoinflammatory syndromes Clinical benefit from JAK 1/2 inhibition

Evinacumab

(Anti-ANGPTL3)

Not yet recruiting Patients with severe hypertriglyceridemia Percent lowering of triglycerides

 

CONCLUSION

 

Lipodystrophy syndromes are a group of fascinating diseases that are caused by mechanisms that disrupt predominantly adipocyte differentiation or lipid droplet formation. LMNAgene defects, the most common single gene defects leading to the development of lipodystrophy syndromes, leads to lipodystrophy possibly due to inducing adipocyte apoptosis or death, but more work is needed on this front. Regardless of the mechanism and whether the diseases present with generalized or partial fat loss, common metabolic complications include severe insulin resistance, hypertriglyceridemia, and ectopic fat deposition, especially hepatic steatosis. This common theme is recapitulated in numerous animal models as well. The diseases are typically progressive and lead to multi-organ involvement and increased mortality. Molecular advances in the understanding of disease mechanisms may lead to better and specific treatments for lipodystrophy syndromes.  So far, the most exciting therapeutic development for the treatment of lipodystrophy syndromes has been the approval of leptin replacement therapy for generalized lipodystrophy in the form of metreleptin. While Metreleptin is not approved for treatment of partial lipodystrophy syndromes in the United States, there are a number of global studies ongoing for the treatment of predominant partial lipodystrophy syndromes with other agents at this time.

 

REFERENCES

 

  1. Chan JL, Oral EA. Clinical classification and treatment of congenital and acquired lipodystrophy. Endocr Pract 2010;16:310-23.
  2. Garg A. Clinical review#: Lipodystrophies: genetic and acquired body fat disorders. J Clin Endocrinol Metab 2011;96:3313-25.
  3. Brown RJ, Araujo-Vilar D, Cheung PT, et al. The Diagnosis and Management of Lipodystrophy Syndromes: A Multi-Society Practice Guideline. The Journal of clinical endocrinology and metabolism 2016;101:4500-11.
  4. Handelsman Y, Oral EA, Bloomgarden ZT, et al. The clinical approach to the detection of lipodystrophy - an AACE consensus statement. Endocr Pract 2013;19:107-16.
  5. Robbins DC, Sims EA. Recurrent ketoacidosis in acquired, total lipodystrophy (lipoatrophic diabetes). Diabetes Care 1984;7:381-5.
  6. Jackson SN, Howlett TA, McNally PG, O'Rahilly S, Trembath RC. Dunnigan-Kobberling syndrome: an autosomal dominant form of partial lipodystrophy. QJM 1997;90:27-36.
  7. Lupsa BC, Sachdev V, Lungu AO, Rosing DR, Gorden P. Cardiomyopathy in congenital and acquired generalized lipodystrophy: a clinical assessment. Medicine (Baltimore) 2010;89:245-50.
  8. Bjornstad PG, Semb BK, Trygstad O, Seip M. Echocardiographic assessment of cardiac function and morphology in patients with generalised lipodystrophy. Eur J Pediatr 1985;144:355-9.
  9. Misra A, Garg A. Clinical features and metabolic derangements in acquired generalized lipodystrophy: case reports and review of the literature. Medicine (Baltimore) 2003;82:129-46.
  10. Seip M. Generalized lipodystrophy. Ergeb Inn Med Kinderheilkd 1971;31:59-95.
  11. Ajluni N, Meral R, Neidert AH, et al. Spectrum of disease associated with partial lipodystrophy: lessons from a trial cohort. Clin Endocrinol (Oxf) 2017;86:698-707.
  12. Ajluni N, Dar M, Xu J, Neidert AH, Oral EA. Efficacy and Safety of Metreleptin in Patients with Partial Lipodystrophy: Lessons from an Expanded Access Program. J Diabetes Metab 2016;7.
  13. Garg A, Misra A. Lipodystrophies: rare disorders causing metabolic syndrome. Endocrinol Metab Clin North Am 2004;33:305-31.
  14. Garg A. Acquired and inherited lipodystrophies. N Engl J Med 2004;350:1220-34.
  15. Caron A, Lee S, Elmquist JK, Gautron L. Leptin and brain-adipose crosstalks. Nat Rev Neurosci 2018;19:153-65.
  16. Musso C, Cochran E, Moran SA, et al. Clinical course of genetic diseases of the insulin receptor (type A and Rabson-Mendenhall syndromes): a 30-year prospective. Medicine (Baltimore) 2004;83:209-22.
  17. Oral EA, Chan JL. Rationale for leptin-replacement therapy for severe lipodystrophy. Endocr Pract 2010;16:324-33.
  18. Melvin A, O'Rahilly S, Savage DB. Genetic syndromes of severe insulin resistance. Curr Opin Genet Dev 2018;50:60-7.
  19. D'Souza A M, Neumann UH, Glavas MM, Kieffer TJ. The glucoregulatory actions of leptin. Mol Metab 2017;6:1052-65.
  20. Triantafyllou GA, Paschou SA, Mantzoros CS. Leptin and Hormones: Energy Homeostasis. Endocrinol Metab Clin North Am 2016;45:633-45.
  21. Meek TH, Morton GJ. The role of leptin in diabetes: metabolic effects. Diabetologia 2016;59:928-32.
  22. Savage DB. Mouse models of inherited lipodystrophy. Dis Model Mech 2009;2:554-62.
  23. Moitra J, Mason MM, Olive M, et al. Life without white fat: a transgenic mouse. Genes Dev 1998;12:3168-81.
  24. Reitman ML, Gavrilova O. A-ZIP/F-1 mice lacking white fat: a model for understanding lipoatrophic diabetes. Int J Obes Relat Metab Disord 2000;24 Suppl 4:S11-4.
  25. Gavrilova O, Marcus-Samuels B, Graham D, et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin Invest 2000;105:271-8.
  26. Zhang Z, Turer E, Li X, et al. Insulin resistance and diabetes caused by genetic or diet-induced KBTBD2 deficiency in mice. Proc Natl Acad Sci U S A 2016;113:E6418-E26.
  27. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL. Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature 1999;401:73-6.
  28. Colombo C, Cutson JJ, Yamauchi T, et al. Transplantation of adipose tissue lacking leptin is unable to reverse the metabolic abnormalities associated with lipoatrophy. Diabetes 2002;51:2727-33.
  29. Oral EA, Simha V, Ruiz E, et al. Leptin-replacement therapy for lipodystrophy. N Engl J Med 2002;346:570-8.
  30. Haque WA, Shimomura I, Matsuzawa Y, Garg A. Serum adiponectin and leptin levels in patients with lipodystrophies. J Clin Endocrinol Metab 2002;87:2395.
  31. Seip M. Lipodystrophy and gigantism with associated endocrine manifestations. A new diencephalic syndrome? Acta Paediatr 1959;48:555-74.
  32. Berardinelli W. An undiagnosed endocrinometabolic syndrome: report of 2 cases. J Clin Endocrinol Metab 1954;14:193-204.
  33. Goodpaster BH. Measuring body fat distribution and content in humans. Curr Opin Clin Nutr Metab Care 2002;5:481-7.
  34. Tsoukas MA MC. Endocrinology Adult and Pediatric. In: Jameson JL DL, ed. 7 ed: Saunders, In Press.
  35. Agarwal AK, Simha V, Oral EA, et al. Phenotypic and genetic heterogeneity in congenital generalized lipodystrophy. The Journal of clinical endocrinology and metabolism 2003;88:4840-7.
  36. Van Maldergem L, Magre J, Khallouf TE, et al. Genotype-phenotype relationships in Berardinelli-Seip congenital lipodystrophy. J Med Genet 2002;39:722-33.
  37. Capeau J, Magre J, Caron-Debarle M, et al. Human lipodystrophies: genetic and acquired diseases of adipose tissue. Endocr Dev 2010;19:1-20.
  38. Maguire M, Lungu A, Gorden P, Cochran E, Stratton P. Pregnancy in a woman with congenital generalized lipodystrophy: leptin's vital role in reproduction. Obstet Gynecol 2012;119:452-5.
  39. Jiang M, Gao M, Wu C, et al. Lack of testicular seipin causes teratozoospermia syndrome in men. Proc Natl Acad Sci U S A 2014;111:7054-9.
  40. Patni N, Garg A. Congenital generalized lipodystrophies--new insights into metabolic dysfunction. Nat Rev Endocrinol 2015;11:522-34.
  41. Garg A, Wilson R, Barnes R, et al. A gene for congenital generalized lipodystrophy maps to human chromosome 9q34. J Clin Endocrinol Metab 1999;84:3390-4.
  42. Fernandez-Galilea M, Tapia P, Cautivo K, Morselli E, Cortes VA. AGPAT2 deficiency impairs adipogenic differentiation in primary cultured preadipocytes in a non-autophagy or apoptosis dependent mechanism. Biochem Biophys Res Commun 2015;467:39-45.
  43. Subauste AR, Das AK, Li X, et al. Alterations in lipid signaling underlie lipodystrophy secondary to AGPAT2 mutations. Diabetes 2012;61:2922-31.
  44. Simha V, Agarwal AK, Aronin PA, Iannaccone ST, Garg A. Novel subtype of congenital generalized lipodystrophy associated with muscular weakness and cervical spine instability. Am J Med Genet A 2008;146A:2318-26.
  45. Garg A, Fleckenstein JL, Peshock RM, Grundy SM. Peculiar distribution of adipose tissue in patients with congenital generalized lipodystrophy. J Clin Endocrinol Metab 1992;75:358-61.
  46. Simha V, Garg A. Phenotypic heterogeneity in body fat distribution in patients with congenital generalized lipodystrophy caused by mutations in the AGPAT2 or seipin genes. J Clin Endocrinol Metab 2003;88:5433-7.
  47. Magre J, Delepine M, Khallouf E, et al. Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q13. Nat Genet 2001;28:365-70.
  48. Cartwright BR, Goodman JM. Seipin: from human disease to molecular mechanism. J Lipid Res 2012;53:1042-55.
  49. Cartwright BR, Binns DD, Hilton CL, Han S, Gao Q, Goodman JM. Seipin performs dissectible functions in promoting lipid droplet biogenesis and regulating droplet morphology. Mol Biol Cell 2015;26:726-39.
  50. Akinci B, Onay H, Demir T, et al. Natural History of Congenital Generalized Lipodystrophy: A Nationwide Study From Turkey. J Clin Endocrinol Metab 2016;101:2759-67.
  51. Altay C, Secil M, Demir T, et al. Determining residual adipose tissue characteristics with MRI in patients with various subtypes of lipodystrophy. Diagnostic and Interventional Radiology 2017;23:428-34.
  52. Antuna-Puente B, Boutet E, Vigouroux C, et al. Higher Adiponectin Levels in Patients with Berardinelli-Seip Congenital Lipodystrophy due to Seipin as compared with 1-Acylglycerol-3-Phosphate-O-Acyltransferase-2 Deficiency. Journal of Clinical Endocrinology & Metabolism 2010;95:1463-8.
  53. Kim CA, Delepine M, Boutet E, et al. Association of a homozygous nonsense caveolin-1 mutation with Berardinelli-Seip congenital lipodystrophy. J Clin Endocrinol Metab 2008;93:1129-34.
  54. Garg A, Agarwal AK. Caveolin-1: a new locus for human lipodystrophy. J Clin Endocrinol Metab 2008;93:1183-5.
  55. Garg A, Kircher M, Del Campo M, Amato RS, Agarwal AK, University of Washington Center for Mendelian G. Whole exome sequencing identifies de novo heterozygous CAV1 mutations associated with a novel neonatal onset lipodystrophy syndrome. Am J Med Genet A 2015;167A:1796-806.
  56. Cao H, Alston L, Ruschman J, Hegele RA. Heterozygous CAV1 frameshift mutations (MIM 601047) in patients with atypical partial lipodystrophy and hypertriglyceridemia. Lipids Health Dis 2008;7:3.
  57. Hayashi YK, Matsuda C, Ogawa M, et al. Human PTRF mutations cause secondary deficiency of caveolins resulting in muscular dystrophy with generalized lipodystrophy. J Clin Invest 2009;119:2623-33.
  58. Rajab A, Straub V, McCann LJ, et al. Fatal cardiac arrhythmia and long-QT syndrome in a new form of congenital generalized lipodystrophy with muscle rippling (CGL4) due to PTRF-CAVIN mutations. PLoS Genet 2010;6:e1000874.
  59. Shastry S, Delgado MR, Dirik E, Turkmen M, Agarwal AK, Garg A. Congenital generalized lipodystrophy, type 4 (CGL4) associated with myopathy due to novel PTRF mutations. Am J Med Genet A 2010;152A:2245-53.
  60. Akinci G, Topaloglu H, Akinci B, et al. Spectrum of clinical manifestations in two young Turkish patients with congenital generalized lipodystrophy type 4. Eur J Med Genet 2016;59:320-4.
  61. Payne F, Lim K, Girousse A, et al. Mutations disrupting the Kennedy phosphatidylcholine pathway in humans with congenital lipodystrophy and fatty liver disease. Proc Natl Acad Sci U S A 2014;111:8901-6.
  62. Hussain I, Patni N, Ueda M, et al. A Novel Generalized Lipodystrophy-associated Progeroid Syndrome due to recurrent heterozygous LMNA p.T10I Mutation. J Clin Endocrinol Metab 2017.
  63. Dyment DA, Gibson WT, Huang L, Bassyouni H, Hegele RA, Innes AM. Biallelic mutations at PPARG cause a congenital, generalized lipodystrophy similar to the Berardinelli-Seip syndrome. Eur J Med Genet 2014;57:524-6.
  64. Arioglu E, Andewelt A, Diabo C, Bell M, Taylor SI, Gorden P. Clinical course of the syndrome of autoantibodies to the insulin receptor (type B insulin resistance): a 28-year perspective. Medicine (Baltimore) 2002;81:87-100.
  65. Pope E, Janson A, Khambalia A, Feldman B. Childhood acquired lipodystrophy: a retrospective study. J Am Acad Dermatol 2006;55:947-50.
  66. Bingham A, Mamyrova G, Rother KI, et al. Predictors of acquired lipodystrophy in juvenile-onset dermatomyositis and a gradient of severity. Medicine (Baltimore) 2008;87:70-86.
  67. Verma S, Singh S, Bhalla AK, Khullar M. Study of subcutaneous fat in children with juvenile dermatomyositis. Arthritis Rheum 2006;55:564-8.
  68. Billings JK, Milgraum SS, Gupta AK, Headington JT, Rasmussen JE. Lipoatrophic panniculitis: a possible autoimmune inflammatory disease of fat. Report of three cases. Arch Dermatol 1987;123:1662-6.
  69. Eren E, Ozkan TB, Cakir ED, Saglam H, Tarim O. Acquired generalized lipodystrophy associated with autoimmune hepatitis and low serum C4 level. J Clin Res Pediatr Endocrinol 2010;2:39-42.
  70. Guillin-Amarelle C, Sanchez-Iglesias S, Castro-Pais A, et al. Type 1 familial partial lipodystrophy: understanding the Kobberling syndrome. Endocrine 2016;54:411-21.
  71. Herbst KL, Tannock LR, Deeb SS, Purnell JQ, Brunzell JD, Chait A. Kobberling type of familial partial lipodystrophy: an underrecognized syndrome. Diabetes Care 2003;26:1819-24.
  72. Lotta LA, Gulati P, Day FR, et al. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat Genet 2017;49:17-26.
  73. Hegele RA, Joy TR, Al-Attar SA, Rutt BK. Thematic review series: Adipocyte Biology. Lipodystrophies: windows on adipose biology and metabolism. J Lipid Res 2007;48:1433-44.
  74. Agarwal AK, Barnes RI, Garg A. Genetic basis of congenital generalized lipodystrophy. Int J Obes Relat Metab Disord 2004;28:336-9.
  75. Garg A. Gender differences in the prevalence of metabolic complications in familial partial lipodystrophy (Dunnigan variety). J Clin Endocrinol Metab 2000;85:1776-82.
  76. Akinci B, Onay H, Demir T, et al. Clinical presentations, metabolic abnormalities and end-organ complications in patients with familial partial lipodystrophy. Metabolism 2017;72:109-19.
  77. Garg A, Vinaitheerthan M, Weatherall PT, Bowcock AM. Phenotypic heterogeneity in patients with familial partial lipodystrophy (dunnigan variety) related to the site of missense mutations in lamin a/c gene. J Clin Endocrinol Metab 2001;86:59-65.
  78. Fountas A, Giotaki Z, Dounousi E, et al. Familial partial lipodystrophy and proteinuric renal disease due to a missense c.1045C > T LMNA mutation. Endocrinol Diabetes Metab Case Rep 2017;2017.
  79. Mory PB, Crispim F, Freire MB, et al. Phenotypic diversity in patients with lipodystrophy associated with LMNA mutations. Eur J Endocrinol 2012;167:423-31.
  80. Monajemi H, Zhang L, Li G, et al. Familial partial lipodystrophy phenotype resulting from a single-base mutation in deoxyribonucleic acid-binding domain of peroxisome proliferator-activated receptor-gamma. J Clin Endocrinol Metab 2007;92:1606-12.
  81. Ludtke A, Buettner J, Wu W, et al. Peroxisome proliferator-activated receptor-gamma C190S mutation causes partial lipodystrophy. J Clin Endocrinol Metab 2007;92:2248-55.
  82. Al-Shali K, Cao H, Knoers N, Hermus AR, Tack CJ, Hegele RA. A single-base mutation in the peroxisome proliferator-activated receptor gamma4 promoter associated with altered in vitro expression and partial lipodystrophy. J Clin Endocrinol Metab 2004;89:5655-60.
  83. Agarwal AK, Garg A. A novel heterozygous mutation in peroxisome proliferator-activated receptor-gamma gene in a patient with familial partial lipodystrophy. J Clin Endocrinol Metab 2002;87:408-11.
  84. Demir T, Onay H, Savage DB, et al. Familial partial lipodystrophy linked to a novel peroxisome proliferator activator receptor -gamma (PPARG) mutation, H449L: a comparison of people with this mutation and those with classic codon 482 Lamin A/C (LMNA) mutations. Diabet Med 2016;33:1445-50.
  85. Gandotra S, Lim K, Girousse A, Saudek V, O'Rahilly S, Savage DB. Human frame shift mutations affecting the carboxyl terminus of perilipin increase lipolysis by failing to sequester the adipose triglyceride lipase (ATGL) coactivator AB-hydrolase-containing 5 (ABHD5). J Biol Chem 2011;286:34998-5006.
  86. Gandotra S, Le Dour C, Bottomley W, et al. Perilipin deficiency and autosomal dominant partial lipodystrophy. N Engl J Med 2011;364:740-8.
  87. Kozusko K, Tsang V, Bottomley W, et al. Clinical and molecular characterization of a novel PLIN1 frameshift mutation identified in patients with familial partial lipodystrophy. Diabetes 2015;64:299-310.
  88. Rubio-Cabezas O, Puri V, Murano I, et al. Partial lipodystrophy and insulin resistant diabetes in a patient with a homozygous nonsense mutation in CIDEC. EMBO Mol Med 2009;1:280-7.
  89. Farhan SM, Robinson JF, McIntyre AD, et al. A novel LIPE nonsense mutation found using exome sequencing in siblings with late-onset familial partial lipodystrophy. Can J Cardiol 2014;30:1649-54.
  90. Zolotov S, Xing C, Mahamid R, Shalata A, Sheikh-Ahmad M, Garg A. Homozygous LIPE mutation in siblings with multiple symmetric lipomatosis, partial lipodystrophy, and myopathy. Am J Med Genet A 2017;173:190-4.
  91. George S, Rochford JJ, Wolfrum C, et al. A family with severe insulin resistance and diabetes due to a mutation in AKT2. Science 2004;304:1325-8.
  92. Tan K, Kimber WA, Luan J, et al. Analysis of genetic variation in Akt2/PKB-beta in severe insulin resistance, lipodystrophy, type 2 diabetes, and related metabolic phenotypes. Diabetes 2007;56:714-9.
  93. Garg A, Sankella S, Xing C, Agarwal AK. Whole-exome sequencing identifies ADRA2A mutation in atypical familial partial lipodystrophy. JCI Insight 2016;1.
  94. Simha V, Garg A. Body fat distribution and metabolic derangements in patients with familial partial lipodystrophy associated with mandibuloacral dysplasia. J Clin Endocrinol Metab 2002;87:776-85.
  95. Garavelli L, D'Apice MR, Rivieri F, et al. Mandibuloacral dysplasia type A in childhood. Am J Med Genet A 2009;149A:2258-64.
  96. Lombardi F, Gullotta F, Columbaro M, et al. Compound heterozygosity for mutations in LMNA in a patient with a myopathic and lipodystrophic mandibuloacral dysplasia type A phenotype. J Clin Endocrinol Metab 2007;92:4467-71.
  97. Agarwal AK, Zhou XJ, Hall RK, et al. Focal segmental glomerulosclerosis in patients with mandibuloacral dysplasia owing to ZMPSTE24 deficiency. J Investig Med 2006;54:208-13.
  98. Novelli G, Muchir A, Sangiuolo F, et al. Mandibuloacral dysplasia is caused by a mutation in LMNA-encoding lamin A/C. Am J Hum Genet 2002;71:426-31.
  99. Garg A, Cogulu O, Ozkinay F, Onay H, Agarwal AK. A novel homozygous Ala529Val LMNA mutation in Turkish patients with mandibuloacral dysplasia. J Clin Endocrinol Metab 2005;90:5259-64.
  100. Agarwal AK, Fryns JP, Auchus RJ, Garg A. Zinc metalloproteinase, ZMPSTE24, is mutated in mandibuloacral dysplasia. Hum Mol Genet 2003;12:1995-2001.
  101. Miyoshi Y, Akagi M, Agarwal AK, et al. Severe mandibuloacral dysplasia caused by novel compound heterozygous ZMPSTE24 mutations in two Japanese siblings. Clin Genet 2008;73:535-44.
  102. Peinado JR, Quiros PM, Pulido MR, et al. Proteomic profiling of adipose tissue from Zmpste24-/- mice, a model of lipodystrophy and premature aging, reveals major changes in mitochondrial function and vimentin processing. Mol Cell Proteomics 2011;10:M111 008094.
  103. Akinci B, Sankella S, Gilpin C, Ozono K, Garg A, Agarwal AK. Progeroid syndrome patients with ZMPSTE24 deficiency could benefit when treated with rapamycin and dimethylsulfoxide. Cold Spring Harb Mol Case Stud 2017;3:a001339.
  104. Weedon MN, Ellard S, Prindle MJ, et al. An in-frame deletion at the polymerase active site of POLD1 causes a multisystem disorder with lipodystrophy. Nat Genet 2013;45:947-50.
  105. Shastry S, Simha V, Godbole K, et al. A novel syndrome of mandibular hypoplasia, deafness, and progeroid features associated with lipodystrophy, undescended testes, and male hypogonadism. J Clin Endocrinol Metab 2010;95:E192-7.
  106. Pelosini C, Martinelli S, Ceccarini G, et al. Identification of a novel mutation in the polymerase delta 1 (POLD1) gene in a lipodystrophic patient affected by mandibular hypoplasia, deafness, progeroid features (MDPL) syndrome. Metabolism 2014;63:1385-9.
  107. Elouej S, Beleza-Meireles A, Caswell R, et al. Exome sequencing reveals a de novo POLD1 mutation causing phenotypic variability in mandibular hypoplasia, deafness, progeroid features, and lipodystrophy syndrome (MDPL). Metabolism 2017;71:213-25.
  108. Donadille B, D'Anella P, Auclair M, et al. Partial lipodystrophy with severe insulin resistance and adult progeria Werner syndrome. Orphanet J Rare Dis 2013;8:106.
  109. Sidorova JM. Roles of the Werner syndrome RecQ helicase in DNA replication. DNA Repair (Amst) 2008;7:1776-86.
  110. Becerra-Munoz VM, Gomez-Doblas JJ, Porras-Martin C, et al. The importance of genotype-phenotype correlation in the clinical management of Marfan syndrome. Orphanet J Rare Dis 2018;13:16.
  111. Graul-Neumann LM, Kienitz T, Robinson PN, et al. Marfan syndrome with neonatal progeroid syndrome-like lipodystrophy associated with a novel frameshift mutation at the 3' terminus of the FBN1-gene. Am J Med Genet A 2010;152A:2749-55.
  112. Takenouchi T, Hida M, Sakamoto Y, et al. Severe congenital lipodystrophy and a progeroid appearance: Mutation in the penultimate exon of FBN1 causing a recognizable phenotype. Am J Med Genet A 2013;161A:3057-62.
  113. Rautenstrauch T, Snigula F, Wiedemann HR. [Neonatal progeroid syndrome (Wiedemann-Rautenstrauch). A follow-up study]. Klin Padiatr 1994;206:440-3.
  114. Davis MR, Arner E, Duffy CR, et al. Expression of FBN1 during adipogenesis: Relevance to the lipodystrophy phenotype in Marfan syndrome and related conditions. Mol Genet Metab 2016;119:174-85.
  115. Cabanillas R, Cadinanos J, Villameytide JA, et al. Nestor-Guillermo progeria syndrome: a novel premature aging condition with early onset and chronic development caused by BANF1 mutations. Am J Med Genet A 2011;155A:2617-25.
  116. Masotti A, Uva P, Davis-Keppen L, et al. Keppen-Lubinsky syndrome is caused by mutations in the inwardly rectifying K+ channel encoded by KCNJ6. Am J Hum Genet 2015;96:295-300.
  117. Lessel D, Vaz B, Halder S, et al. Mutations in SPRTN cause early onset hepatocellular carcinoma, genomic instability and progeroid features. Nat Genet 2014;46:1239-44.
  118. Chudasama KK, Winnay J, Johansson S, et al. SHORT syndrome with partial lipodystrophy due to impaired phosphatidylinositol 3 kinase signaling. Am J Hum Genet 2013;93:150-7.
  119. Thauvin-Robinet C, Auclair M, Duplomb L, et al. PIK3R1 mutations cause syndromic insulin resistance with lipoatrophy. Am J Hum Genet 2013;93:141-9.
  120. Huang-Doran I, Tomlinson P, Payne F, et al. Insulin resistance uncoupled from dyslipidemia due to C-terminal PIK3R1 mutations. JCI Insight 2016;1:e88766.
  121. Agarwal AK, Xing C, DeMartino GN, et al. PSMB8 encoding the beta5i proteasome subunit is mutated in joint contractures, muscle atrophy, microcytic anemia, and panniculitis-induced lipodystrophy syndrome. Am J Hum Genet 2010;87:866-72.
  122. Garg A, Hernandez MD, Sousa AB, et al. An autosomal recessive syndrome of joint contractures, muscular atrophy, microcytic anemia, and panniculitis-associated lipodystrophy. J Clin Endocrinol Metab 2010;95:E58-63.
  123. Kitamura A, Maekawa Y, Uehara H, et al. A mutation in the immunoproteasome subunit PSMB8 causes autoinflammation and lipodystrophy in humans. J Clin Invest 2011;121:4150-60.
  124. Torrelo A, Patel S, Colmenero I, et al. Chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome. J Am Acad Dermatol 2010;62:489-95.
  125. Kanazawa N. Nakajo-Nishimura syndrome: an autoinflammatory disorder showing pernio-like rashes and progressive partial lipodystrophy. Allergol Int 2012;61:197-206.
  126. Torrelo A. CANDLE Syndrome As a Paradigm of Proteasome-Related Autoinflammation. Front Immunol 2017;8:927.
  127. Cavalcante MP, Brunelli JB, Miranda CC, et al. CANDLE syndrome: chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature-a rare case with a novel mutation. Eur J Pediatr 2016;175:735-40.
  128. Arima K, Kinoshita A, Mishima H, et al. Proteasome assembly defect due to a proteasome subunit beta type 8 (PSMB8) mutation causes the autoinflammatory disorder, Nakajo-Nishimura syndrome. Proc Natl Acad Sci U S A 2011;108:14914-9.
  129. Rocha N, Bulger DA, Frontini A, et al. Human biallelic MFN2 mutations induce mitochondrial dysfunction, upper body adipose hyperplasia, and suppression of leptin expression. Elife 2017;6.
  130. Pinheiro M, Freire-Maia N, Chautard-Freire-Maia EA, Araujo LM, Liberman B. AREDYLD: a syndrome combining an acrorenal field defect, ectodermal dysplasia, lipoatrophic diabetes, and other manifestations. Am J Med Genet 1983;16:29-33.
  131. Breslau-Siderius EJ, Toonstra J, Baart JA, Koppeschaar HP, Maassen JA, Beemer FA. Ectodermal dysplasia, lipoatrophy, diabetes mellitus, and amastia: a second case of the AREDYLD syndrome. Am J Med Genet 1992;44:374-7.
  132. Misra A, Peethambaram A, Garg A. Clinical features and metabolic and autoimmune derangements in acquired partial lipodystrophy: report of 35 cases and review of the literature. Medicine (Baltimore) 2004;83:18-34.
  133. Savage DB, Semple RK, Clatworthy MR, et al. Complement abnormalities in acquired lipodystrophy revisited. J Clin Endocrinol Metab 2009;94:10-6.
  134. Mathieson PW, Wurzner R, Oliveria DB, Lachmann PJ, Peters DK. Complement-mediated adipocyte lysis by nephritic factor sera. J Exp Med 1993;177:1827-31.
  135. Hegele RA, Cao H, Liu DM, et al. Sequencing of the reannotated LMNB2 gene reveals novel mutations in patients with acquired partial lipodystrophy. Am J Hum Genet 2006;79:383-9.
  136. Akinci B, Koseoglu FD, Onay H, et al. Acquired partial lipodystrophy is associated with increased risk for developing metabolic abnormalities. Metabolism 2015;64:1086-95.
  137. Vantyghem MC, Vigouroux C, Magre J, et al. Late-onset lipoatrophic diabetes. Phenotypic and genotypic familial studies and effect of treatment with metformin and lispro insulin analog. Diabetes Care 1999;22:1374-6.
  138. Luedtke A, Boschmann M, Colpe C, et al. Thiazolidinedione response in familial lipodystrophy patients with LMNA mutations: a case series. Horm Metab Res 2012;44:306-11.
  139. Moreau F, Boullu-Sanchis S, Vigouroux C, et al. Efficacy of pioglitazone in familial partial lipodystrophy of the Dunnigan type: a case report. Diabetes Metab 2007;33:385-9.
  140. McLaughlin PD, Ryan J, Hodnett PA, O'Halloran D, Maher MM. Quantitative whole-body MRI in familial partial lipodystrophy type 2: changes in adipose tissue distribution coincide with biochemical improvement. AJR Am J Roentgenol 2012;199:W602-6.
  141. Arioglu E, Duncan-Morin J, Sebring N, et al. Efficacy and safety of troglitazone in the treatment of lipodystrophy syndromes. Ann Intern Med 2000;133:263-74.
  142. Sleilati GG, Leff T, Bonnett JW, Hegele RA. Efficacy and safety of pioglitazone in treatment of a patient with an atypical partial lipodystrophy syndrome. Endocr Pract 2007;13:656-61.
  143. Iwanishi M, Ebihara K, Kusakabe T, et al. Clinical characteristics and efficacy of pioglitazone in a Japanese diabetic patient with an unusual type of familial partial lipodystrophy. Metabolism 2009;58:1681-7.
  144. Kuzuya H, Matsuura N, Sakamoto M, et al. Trial of insulinlike growth factor I therapy for patients with extreme insulin resistance syndromes. Diabetes 1993;42:696-705.
  145. Moses AC, Morrow LA, O'Brien M, Moller DE, Flier JS. Insulin-like growth factor I (rhIGF-I) as a therapeutic agent for hyperinsulinemic insulin-resistant diabetes mellitus. Diabetes Res Clin Pract 1995;28 Suppl:S185-94.
  146. Satoh M, Yoshizawa A, Takesue M, Saji T, Yokoya S. Long-term effects of recombinant human insulin-like growth factor I treatment on glucose and lipid metabolism and the growth of a patient with congenital generalized lipodystrophy. Endocr J 2006;53:639-45.
  147. Johns KW, Bennett MT, Bondy GP. Are HIV positive patients resistant to statin therapy? Lipids Health Dis 2007;6:27.
  148. Macallan DC, Baldwin C, Mandalia S, et al. Treatment of altered body composition in HIV-associated lipodystrophy: comparison of rosiglitazone, pravastatin, and recombinant human growth hormone. HIV Clin Trials 2008;9:254-68.
  149. Lager CJ, Esfandiari NH, Subauste AR, et al. Roux-En-Y Gastric Bypass Vs. Sleeve Gastrectomy: Balancing the Risks of Surgery with the Benefits of Weight Loss. Obes Surg 2017;27:154-61.
  150. Melvin A, Adams C, Flanagan C, et al. Roux-en-Y Gastric Bypass Surgery in the Management of Familial Partial Lipodystrophy Type 1. J Clin Endocrinol Metab 2017;102:3616-20.
  151. Utzschneider KM, Trence DL. Effectiveness of gastric bypass surgery in a patient with familial partial lipodystrophy. Diabetes Care 2006;29:1380-2.
  152. Ciudin A, Baena-Fustegueras JA, Fort JM, Encabo G, Mesa J, Lecube A. Successful treatment for the Dunnigan-type familial partial lipodystrophy with Roux-en-Y gastric bypass. Clin Endocrinol (Oxf) 2011;75:403-4.
  153. Grundfest-Broniatowski S, Yan J, Kroh M, Kilim H, Stephenson A. Successful Treatment of an Unusual Case of FPLD2: The Role of Roux-en-Y Gastric Bypass-Case Report and Literature Review. J Gastrointest Surg 2017;21:739-43.
  154. McDuffie JR, Riggs PA, Calis KA, et al. Effects of exogenous leptin on satiety and satiation in patients with lipodystrophy and leptin insufficiency. J Clin Endocrinol Metab 2004;89:4258-63.
  155. Moran SA, Patten N, Young JR, et al. Changes in body composition in patients with severe lipodystrophy after leptin replacement therapy. Metabolism 2004;53:513-9.
  156. Ebihara K, Kusakabe T, Hirata M, et al. Efficacy and safety of leptin-replacement therapy and possible mechanisms of leptin actions in patients with generalized lipodystrophy. J Clin Endocrinol Metab 2007;92:532-41.
  157. Schlogl H, Muller K, Horstmann A, et al. Leptin Substitution in Patients With Lipodystrophy: Neural Correlates for Long-term Success in the Normalization of Eating Behavior. Diabetes 2016;65:2179-86.
  158. Schlogl H, Muller K, Horstmann A, et al. Leptin-substitution in patients with congenital lipodystrophy increases connectivity in reward-related brain structures: an fMRI study. Exp Clin Endocr Diab 2014;122.
  159. Schlogl H, Muller K, Horstmann A, et al. Leptin-substitution increases connectivity in reward-related brain areas in patients with congenital lipodystrophy. Diabetologia 2015;58:S71-S.
  160. Aotani D, Ebihara K, Sawamoto N, et al. Functional magnetic resonance imaging analysis of food-related brain activity in patients with lipodystrophy undergoing leptin replacement therapy. The Journal of clinical endocrinology and metabolism 2012;97:3663-71.
  161. Petersen KF, Oral EA, Dufour S, et al. Leptin reverses insulin resistance and hepatic steatosis in patients with severe lipodystrophy. J Clin Invest 2002;109:1345-50.
  162. Diker-Cohen T, Cochran E, Gorden P, Brown RJ. Partial and generalized lipodystrophy: comparison of baseline characteristics and response to metreleptin. J Clin Endocrinol Metab 2015;100:1802-10.
  163. Muniyappa R, Brown RJ, Mari A, et al. Effects of leptin replacement therapy on pancreatic beta-cell function in patients with lipodystrophy. Diabetes Care 2014;37:1101-7.
  164. Vatier C, Fetita S, Boudou P, et al. One-year metreleptin improves insulin secretion in patients with diabetes linked to genetic lipodystrophic syndromes. Diabetes Obes Metab 2016;18:693-7.
  165. Chan JL, Lutz K, Cochran E, et al. Clinical effects of long-term metreleptin treatment in patients with lipodystrophy. Endocr Pract 2011;17:922-32.
  166. Kamran F, Rother KI, Cochran E, Safar Zadeh E, Gorden P, Brown RJ. Consequences of stopping and restarting leptin in an adolescent with lipodystrophy. Hormone research in paediatrics 2012;78:320-5.
  167. Chong AY, Lupsa BC, Cochran EK, Gorden P. Efficacy of leptin therapy in the different forms of human lipodystrophy. Diabetologia 2010;53:27-35.
  168. Simha V, Subramanyam L, Szczepaniak L, et al. Comparison of efficacy and safety of leptin replacement therapy in moderately and severely hypoleptinemic patients with familial partial lipodystrophy of the Dunnigan variety. J Clin Endocrinol Metab 2012;97:785-92.
  169. Park JY, Javor ED, Cochran EK, DePaoli AM, Gorden P. Long-term efficacy of leptin replacement in patients with Dunnigan-type familial partial lipodystrophy. Metabolism 2007;56:508-16.
  170. Simha V, Szczepaniak LS, Wagner AJ, DePaoli AM, Garg A. Effect of leptin replacement on intrahepatic and intramyocellular lipid content in patients with generalized lipodystrophy. Diabetes Care 2003;26:30-5.
  171. Javor ED, Ghany MG, Cochran EK, et al. Leptin reverses nonalcoholic steatohepatitis in patients with severe lipodystrophy. Hepatology 2005;41:753-60.
  172. Safar Zadeh E, Lungu AO, Cochran EK, et al. The liver diseases of lipodystrophy: the long-term effect of leptin treatment. Journal of hepatology 2013;59:131-7.
  173. Machado MV, Cortez-Pinto H. Leptin in the treatment of lipodystrophy-associated nonalcoholic fatty liver disease: are we there already? Expert Rev Gastroenterol Hepatol 2013;7:513-5.
  174. Brown RJ, Meehan CA, Cochran E, et al. Effects of Metreleptin in Pediatric Patients With Lipodystrophy. J Clin Endocrinol Metab 2017;102:1511-9.
  175. Casey SP, Lokan J, Testro A, et al. Post-liver transplant leptin results in resolution of severe recurrence of lipodystrophy-associated nonalcoholic steatohepatitis. Am J Transplant 2013;13:3031-4.
  176. Javor ED, Moran SA, Young JR, et al. Proteinuric nephropathy in acquired and congenital generalized lipodystrophy: baseline characteristics and course during recombinant leptin therapy. J Clin Endocrinol Metab 2004;89:3199-207.
  177. Oral EA, Ruiz E, Andewelt A, et al. Effect of leptin replacement on pituitary hormone regulation in patients with severe lipodystrophy. J Clin Endocrinol Metab 2002;87:3110-7.
  178. Musso C, Cochran E, Javor E, Young J, Depaoli AM, Gorden P. The long-term effect of recombinant methionyl human leptin therapy on hyperandrogenism and menstrual function in female and pituitary function in male and female hypoleptinemic lipodystrophic patients. Metabolism 2005;54:255-63.
  179. Abel BS, Muniyappa R, Stratton P, Skarulis MC, Gorden P, Brown RJ. Effects of Recombinant Human Leptin (Metreleptin) on Nocturnal Luteinizing Hormone Secretion in Lipodystrophy Patients. Neuroendocrinology 2016;103:402-7.
  180. Lungu AO, Zadeh ES, Goodling A, Cochran E, Gorden P. Insulin resistance is a sufficient basis for hyperandrogenism in lipodystrophic women with polycystic ovarian syndrome. J Clin Endocrinol Metab 2012;97:563-7.
  181. Meehan CA, Cochran E, Kassai A, Brown RJ, Gorden P. Metreleptin for injection to treat the complications of leptin deficiency in patients with congenital or acquired generalized lipodystrophy. Expert Rev Clin Pharmacol 2016;9:59-68.
  182. Christensen JD, Lungu AO, Cochran E, et al. Bone mineral content in patients with congenital generalized lipodystrophy is unaffected by metreleptin replacement therapy. J Clin Endocrinol Metab 2014;99:E1493-500.
  183. Simha V, Zerwekh JE, Sakhaee K, Garg A. Effect of subcutaneous leptin replacement therapy on bone metabolism in patients with generalized lipodystrophy. J Clin Endocrinol Metab 2002;87:4942-5.
  184. Chan JL, Koda J, Heilig JS, et al. Immunogenicity associated with metreleptin treatment in patients with obesity or lipodystrophy. Clin Endocrinol (Oxf) 2016;85:137-49.
  185. Beltrand J, Lahlou N, Le Charpentier T, et al. Resistance to leptin-replacement therapy in Berardinelli-Seip congenital lipodystrophy: an immunological origin. Eur J Endocrinol 2010;162:1083-91.
  186. Brown RJ, Chan JL, Jaffe ES, et al. Lymphoma in acquired generalized lipodystrophy. Leuk Lymphoma 2016;57:45-50.
  187. Aslam A, Savage DB, Coulson IH. Acquired generalized lipodystrophy associated with peripheral T cell lymphoma with cutaneous infiltration. Int J Dermatol 2015;54:827-9.
  188. MYALEPT PRESCRIBING INFORMATION. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125390s000lbl.pdf.
  189. Lebastchi J, Ajluni N, Neidert A, Oral EA. A Report of Three Cases With Acquired Generalized Lipodystrophy With Distinct Autoimmune Conditions Treated With Metreleptin. J Clin Endocrinol Metab 2015;100:3967-70.
  190. Park JY, Chong AY, Cochran EK, et al. Type 1 diabetes associated with acquired generalized lipodystrophy and insulin resistance: the effect of long-term leptin therapy. The Journal of clinical endocrinology and metabolism 2008;93:26-31.

Florid Cushing’s Syndrome

CLINICAL RECOGNITION

 

Cushing’s syndrome (CS) results from long-standing exposure to supraphysiologic concentrations of circulating glucocorticoids. Untreated CS is associated with a high morbidity and increased mortality rates mainly because of its metabolic abnormalities and the risk of infection. When the presentation is florid, the diagnosis is usually straightforward (Tables 1 and 2). However, diagnosis might be complicated by the non-specificity of some of the clinical symptoms; notably, the signs that most reliably distinguish CS from obesity are those of protein wasting – the presence of thin skin, easy bruising, and proximal muscle weakness.

Table 1 Signs, Symptoms of Cushing’s Syndrome

Signs Symptoms
Buffalo hump, moon face; flushing/red face 
Weight gain/ central obesity
Easy bruising/ecchymosis
Proximal myopathy/wasting/muscular atrophy 
Purplish skin striae 
Skin pigmentation
Thin skin/ skin disorders 
Edema; 
Fundal abnormalities 
Hair thin, dry
Loss of weight
Poor growth/short stature; 
Facial hirsutism (±frontal balding)
Fine non-pigmented vellus hair
Acne
Virilization
Galactorrhea
Fatigue, lethargy, poor exercise ability 
Headaches/migraines 
Joint aches 
Sweating 
Visual disturbances
Thirst/polydipsia 
Loss of libido 
Poor memory
Poor concentration
Slowed responses

Table 2 Disorders Associated with Cushing’s Cyndrome

Hypertension/ CVD/ prolonged QTc dispersion/ LVH

Hyperhomocysteinemia/ increased thrombotic tendency

Diabetes

Obesity/ increased visceral fat/ hepatic steatosis

Infection

Depression/ emotional lability/ anxiety/ sleep disorders/ lethargy/ psychosis/ psychiatric disorders

Hypogonadotrophic hypogonadism

Osteoporosis/ vertebral fractures

Cognitive/memory impairment

Renal stones

Thyroid disorders

Growth retardation

Menstrual disorders/ PCO

 

PATHOPHYSIOLOGY

 

See http://www.endotext.org/neuroendo/neuroendo7/neuroendoframe7.htm

 

DIAGNOSIS and DIFFERENTIAL DIAGNOSIS

 

The most common cause of CS is the use of supraphysiological amounts of exogenous glucocorticoids. (Table 3) Typically this is of moderate severity, and not an emergency. A detailed drug history is essential for diagnosis.

Table 3 Etiology of Cushing's Syndrome

Exogenous causes
Exogenous glucocorticoid administration
Iatrogenic

Drug--drug interactions (via hepatic enzyme CYP3A4 and P-glycoprotein (PGP) export pump)

Factitious

Exogenous ACTH administration
Iatrogenic
Endogenous causes
ACTH-dependent (80-85%)
Cushing's disease (80%)

Ectopic ACTH syndrome (20%) 
Ectopic CRH syndrome (<1%)

ACTH-independent (20%)
Adrenal adenoma (60%)
Adrenal carcinoma (40%)
ACTH-independent bilateral macronodular adrenal hyperplasia (AIMAH)±secondary to abnormal hormone receptor expression/function or armadillo repeat-containing-5 (ARMC5) gene mutations (<1%)
Sporadic or associated with Carney complex primary pigmented nodular adrenal disease (PPNAD) or micronodular adrenal disease(<1%)
Bilateral nodular adrenal disease in McCune-Albright syndrome (<1%)
Constitutive activation ACTH receptor by missense mutation (<1%)

 

Endogenous CS is more common in women. Corticotrophin (ACTH)-dependent CS is caused mainly by a pituitary corticotroph adenoma (Cushing’s disease, CD) secreting ACTH, or by an extra-pituitary tumor (ectopic ACTH syndrome, EAS, (Table 4). ACTH-independent CS is caused by unilateral adrenocortical tumors or by bilateral adrenal hyperplasia or dysplasia.

 

In ACTH-dependent CS, elevated ACTH secretion results in excess adrenal gland cortisol secretion. The normal cortisol feedback mechanism of the hypothalamic-pituitary-adrenal (HPA) axis is distorted, with loss of the circadian rhythm, excess cortisol production, and loss of normal suppression to the exogenous administration of glucocorticoids. EAS can have a rapid onset with severe features, although in some patients a paraneoplastic wasting syndrome can mask the hypercortisolism. The metabolic abnormalities such as hyperglycemia and hypokalemia tend to be more florid in EAS.

 

Table 4 Tumors More Frequently Associated with Ectopic ACTH Syndrome

Tumors Percentage reported (%)
Small cell lung carcinoma 3.3-50
Bronchial carcinoids 5-40
Islet cell tumors/ pancreatic carcinoids 7.5-25
Thymic carcinoids 5-42
Pheochromocytoma 2.5-25
Medullary thyroid carcinoma 2-8
Gastrinoma 5
Tumour not identified 12-37.5

 

In ACTH-independent CS, the most common pathology is an adrenal adrenocortical adenoma (AAA) or carcinoma (AAC) (Table 3). Adrenal adenomas occur most often around 35 years of age and are more common in women with an incidence of approximately 0.6 per million per year. The incidence of ACC is approximately 0.2 per million per year. Different frequencies have been observed in childhood (Table 5).

 

Table 5 Cushing’s Syndrome Presence In Children

Age group Mean age (yr)
McCune Albright syndrome infants 1.2
Adrenal adrenocortical carcinoma young children 4.5
Ectopic ACTH syndrome (rare) older children 10.1
Primary pigmented nodular adrenal disease adolescents 13.0
Cushing’s disease adolescents 14.1

 

DIAGNOSTIC TESTS NEEDED AND SUGGESTED

 

Diagnostic assessment is usually prompted by clinical suspicion in cases of florid CS seen as medical emergencies. In the investigation of CS, the initial biochemical tests should ideally have maximal sensitivity rather than specificity in order to identify individuals with the mild forms of this rare disease; later, more specific tests are used to exclude false positives (Table 6). Hypercortisolemia must be established before any attempt at the differential diagnosis. A combination of the following tests is initially used: 24-h urinary free cortisol (UFC), ideally measured by liquid chromatography tandem-mass spectrometry to improve accuracy, low-dose dexamethasone suppression test (LDDST) or overnight dexamethasone test (ODST), and assessment of midnight serum cortisol (MSeC) or late-night salivary cortisol (LNSaC) (Tables 6 and 7). However, it should be emphasized that in cases presenting with severe disease, a massively elevated serum cortisol at any time, or a urinary cortisol more than 4x the upper limit of normal, is sufficient to confirm the diagnosis. No other tests may be required for the diagnosis.

Table 6 Tests Used For The Establishment Of Hypercortisolemia

Normal
24-hr UFC free cortisol/ creatinine measurement 3 normal collections
ODST 1mg dexamethasone midnight <50nmol/L(<1.8μg/dL) 9:00 
next morning
LDDST 0.5mg dexamethasone/6hr for 48hrs (09.00 day0; post-48hrs) <50nmol/L(<1.8μg/dL)
MSeC/ LNSaC Midnight/23:00hr Saliva: Local

range; serum: asleep<50nmol/L, awake: 207 nmol/l (7 μg/dL)

LDDST: Low-dose dexamethasone suppression test;
LNSaC: late-night salivary cortisol
MSeC: midnight serum cortisol;
ODST: overnight dexamethasone suppression test;
UFC: urinary free cortisol

 

The second step in the diagnostic cascade of CS is to establish the cause by measuring plasma ACTH. Values in the ‘grey zone’ are the most challenging since patients with both CD and adrenal pathologies might have intermediate values. However, a plasma ACTH above 20 ng/L will immediately establish ACTH-dependence, while levels below 10 ng/L will lead to the search for adrenal pathology. If ACTH is present, then the patient either has Cushing’s disease or an ectopic source. A positive ACTH and/or cortisol response to the CRH test will suggest Cushing’s disease, a poor response to either the LDDST or the high-dose test likewise, but bilateral inferior petrosal sinus sampling (BIPSS) is generally advised in all cases except when (1) there is an obvious macroadenoma on MRI of the pituitary, or (2) the patient is too ill and requires immediate medical therapy. Data on the utility of these tests are given in Table 7.

Table 7 Test Used For The Differential Diagnosis Of Hypercortisolemia

HDDST 2mg dexamethasone/6hr for 48hrs (09.00 day0; post-48hrs) cortisol suppression >50%: CD;
sensitivity:60-100%; specificity:65-100%
hCRH/oCRH test iv-bolus 1µg/kg 
or 100 µg
o-CRH: ACTH > 35%/ or cortisol > 20% specific for CD; h-CRH: ACTH > 105%/ or cortisol >14; sensitivity:94%;
BIPPS ACTH pituitary-to-periphery gradient Basal central-to-peripheral ratio > 2,
or post-CRH>3: CD; sensitivity/specificity: 94%
ACTH levels (±potassium, bicarbonate) < 1.1pmol/L (5pg/mL) ACTH-independent CS; > 3.3pmol/L (15pg/mL) ACTH-dependent pathologies; in-between further investigation
BIPPS: Bilateral inferior petrosal sinus sampling;
CRH: corticotrophin releasing hormone;
CS: Cushing’s syndrome;
h-CRH: recombinant human;
CRH HDDST: High-dose dexamethasone suppression test;
iv: intravenous; o-CRH: ovine-sequence CRH

If ACTH is very low or undetectable, then the next step is imaging of the adrenals. (Table 8). High-resolution computed tomography (CT) scanning of the adrenal glands gives the best resolution of adrenal anatomy and it is accurate for masses >1cm allowing evaluation of the contralateral gland. A mass >5 cm in diameter is considered to be malignant until proven otherwise.

Table 8 Adrenal Gland Imaging In Different Types Of Cushing’s Syndrome

Disease Adrenal gland morphology
Adrenal tumours typically unilateral mass + atrophic contralateral gland
PPNAD normal or slightly lumpy (multiple 
small nodules); not enlarged
AIMAH bilaterally huge (>5cm) with nodular pattern
ACTH-dependent forms of CS enlarged (70%)
Cushing’s disease Enlarged ± nodules; adrenal hyperplasia not always symmetrical ± adrenal autonomy
EAS virtually always homogeneously enlarged
Exogenous administrationof glucocorticoids adrenal atrophy and very 
small glands.
AIMAH: • ACTH-independent bilateral macronodular adrenal hyperplasia; EAS: ectopic ACTH syndrome; PPNAD: primary pigmented nodular adrenal disease

Go to:

THERAPY

 

The goals of treatment are the normalization of cortisol levels with a reversal of clinical symptoms. However, it is important in the short-term to manage the metabolic problems associated with florid CS. Diabetes needs to be controlled in the standard manner, often requiring insulin, while the blood pressure will also require urgent attention. Hypokalemia is a problem in almost all patients with EAS and some 10% of patients with other etiologies. Spironolactone at a dose of 50 or 100mg is usually effective, but triamterene is sometimes a good alternative. These patients also have a high pro-thrombotic tendency, and we would usually use sc heparin at prophylactic doses as opposed to severe cases where low molecular weight heparins should be used at therapeutic doses. Where the mental changes are severe and causing problems in management, haloperidol may be necessary to calm the patient, although there is also some experience with olanzapine.

 

These patients are at high risk of sepsis, often with minimal clinical signs, and any such infection must be vigorously treated. This includes bacterial, fungal and viral causes, as is seen in other immunosuppressed patients.

 

In terms of specific treatment of the hypercortisolemia (Table 9), where available metyrapone is rapid in onset and highly effective, but doses up to 1g qid may be required. Osilodrostat, displaying a similar profile but with higher potency and a better adverse effect profile, seems a promising currently experimental drug. Ketoconazole can be used additionally or in place of metyrapone, although its onset of action is slower, occurring over several days: up to 400mg tid may be used. As the dose is titrated upwards close monitoring of liver function tests is important. Levoketoconazole may have a better safety profile particularly regarding hepatotoxicity and is currently under trial. When neither drug alone or in combination is effective or tolerated, then intravenous etomidate at sub-anesthetic doses may be very useful: it acts within hours and is almost always very effective but should be administered in an intensive care unit. Finally, if all else fails, mifepristone 400-800mg daily can reduce the symptoms and signs of CS but there two caveats; serum cortisol cannot be used as marker of efficacy and the patient can become Addisonian unless care is taken, and severe hypokalemia may be induced (this is treatable with spironolactone).

 

In patients with severe infection, the serum cortisol should be lowered to a level compatible with that seen in other patients with life-threatening infection, which we take as 600-1000 nmol/L. Some would prophylactically treat for the possibility of pneumocytis carinii pnemonia.

 

In the long-term, surgical removal of the tumor of ACTH-dependent or ACTH-independent origin is the first-line therapeutic approach. Anti-glucocorticoid medical treatment is usually required before surgery to reverse the metabolic consequences and poor healing or in patients who cannot undergo surgical procedures because of co-morbidities, or who are unwilling to receive other types of treatment.

 

Table 9 Medical Treatment: Adrenal Secretion Inhibitors Or Adrenolytic Drugs

Drugs Drawbacks Blockage
Metyrapone Escape phenomenon; hypertension, hypokalemia, edema;
women: hirsutism
11ß-hydroxylase
Osilodrostat Possible escape phenomenon, weight gain, edema, hypernatremia, gastrointestinal effects, fatigue, headache, hypokalemia, 11ß-hydroxylase and aldosterone synthase
Ketoconazole Escape phenomenon; men: gynecomastia, hypogonadism; mild liver enzyme elevation; rarely: liver failure cytochromeP450 enzymes
(17,20-lyase; cholesterol
side-chain cleavage,16a-/ 17a-/ 18-/ 11ß-hydroxylase
Levoketoconazole better safety and efficacy compared to ketoconazole mainly 21-hydroxylase, 17α-hydroxylase, 11β-hydroxylase
Etomidate Sedation 11ß-hydroxylase; 17ß-hydroxylase 17,20-lyase; cholesterol side chain cleavage
Fluconazole Not well studied; possibly better tolerated  compared to ketoconazole As ketoconazole
Mitotane Slow onset of action; digestive symptoms;
neurotoxicity; hypercholesterolemia
Cholesterol side-chain cleavage 
11ß- and 18-hydroxylase 
3ß-hydroxysteroid dehydrogenase
Compounds targeting glucocorticoid function: Glucocorticoid antagonists
mifepristone No follow-up marker Competitive binding to the glucocorticoid, androgen and progestin receptors

 

In patients with CD trans-sphenoidal surgery (TSS) offers the potential to leave the remaining pituitary function intact. Initial remission rate is 60-80% but with a recurrence rate of up to 20% after prolonged follow-up. Macroadenoma remission rates are lower. Reoperation is possible. Hypocortisolemia in the immediate postoperative period needs glucocorticoid replacement treatment until HPA axis recovery. Postoperative concentration of cortisol <50nmol/L defines cure but is not predictive of no recurrence. After surgery failure, conventional fractionated external beam radiotherapy achieves control of hypercortisolemia in approximately 50–60% of patients within 3-5 years but with long-term hypopituitarism, and delayed effectiveness; this treatment seems more effective in children. Stereotactic radiosurgery has also been reported to be effective with probably the same time of onset of control, but as the beam is more focused there may be less hypopituitarism; the tumor needs to be well clear of the optic chiasm.

 

Resection of the causative tumor is the optimum treatment for EAS. If this is not feasible because of metastatic or occult disease an individualized approach has to be used.

 

In any cause of ACTH-dependent CS, bilateral adrenalectomy induces a rapid resolution of the clinical features after first-line treatment failure or when drugs are not effective or tolerated or when the rapid control of hypercortisolemia is crucial; however, patients will need lifelong treatment with glucocorticoids and mineralocorticoids besides the careful education and the meticulous evaluation of patients.

 

Adrenal gland removal laparoscopically is the treatment of choice for unilateral adrenal adenomas. Prognosis after removal of an adenoma is good, as opposed to ACC which is poor. Those latter tumors are not usually radiosensitive or chemosensitive and the most important predictor of favorable outcome in this disease is complete resection.

 

In AIMAH, cortisol secretion can be controlled in some cases by blocking the corresponding aberrantly expressed receptor (propranolol for aberrant β-adrenergic receptor expression; somatostatin analogues in gastric inhibitory peptide responsive AIMAH or leuprolide in luteinizing hormone dependent CS). However, most patients need bilateral adrenalectomy.

 

FOLLOW-UP

 

Once the CS has been adequately treated, then long-term follow-up is mandated for all patients.

 

GUIDELINES

 

Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2008 May;93(5):1526-40.

http://www.ncbi.nlm.nih.gov/pubmed/18334580

 

Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price J, Savage MO, Tabarin A; Endocrine Society. Treatment of Cushing's Syndrome: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2015 Aug;100(8):2807-31.

http://www.ncbi.nlm.nih.gov/pubmed/26222757

 

REFERENCES

Morris DG and Grossman A. Cushing’s Syndrome,. in the on-line web-book,WWW.ENDOTEXT.ORG http://www.endotext.org/neuroendo/neuroendo7

 

Guignat L, Bertherat J. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline: commentary from a European perspective. Eur J Endocrinol. 2010 Jul;163(1):9-13 http://www.ncbi.nlm.nih.gov/pubmed/20375177

 

Biller BM, Grossman AB, Stewart PM, Melmed S, Bertagna X, Bertherat J, Buchfelder M, Colao A, Hermus AR, Hofland LJ, Klibanski A, Lacroix A, Lindsay JR, Newell-Price J, Nieman LK, Petersenn S, Sonino N, Stalla GK, Swearingen B, Vance ML, Wass JA, Boscaro M. Treatment of adrenocorticotropin-dependent Cushing's syndrome: a consensus statement. J Clin Endocrinol Metab. 2008 Jul;93(7):2454-62. http://www.ncbi.nlm.nih.gov/pubmed/18413427

 

Arnaldi G, Angeli A, Atkinson AB, Bertagna X, Cavagnini F, Chrousos GP, Fava GA, Findling JW, Gaillard RC, Grossman AB, Kola B, Lacroix A, Mancini T, Mantero F, Newell-Price J, Nieman LK,,Sonino N, Vance ML, Giustina A, Boscaro M. Diagnosis and complications of Cushing's syndrome: a consensus statement. J Clin Endocrinol Metab. 2003 Dec;88(12):5593-602. http://www.ncbi.nlm.nih.gov/pubmed/14671138

 

Alexandraki KI, Grossman AB. Therapeutic Strategies for the Treatment of Severe Cushing's Syndrome. Drugs. 2016 Mar;76(4):447-58. http://www.ncbi.nlm.nih.gov/pubmed/26833215

 

Graves’ Disease: Complications

ABSTRACT

Thyroid storm is an acute and life-threatening worsening of hyperthyroidism, characterized by an exacerbation of symptoms and signs of hyperthyroidism, with high fever, dehydration, marked tachycardia or tachyarrhytmias, heart failure, hepatomegaly, respiratory distress, abdominal pain, delirium, possibly seizures. It may occur in patients submitted to thyroidectomy or radioactive iodine treatment while hyperthyroid, or as a consequence of infections in unteated hyperthyroid patients. Treatment consists of antithyroid drug treatmnt, rest, sedation, fluid and electrolyte replacement, cardio-supportive therapy, oxygen therapy, antibiotics, cooling. Mortality is about 10%.

Graves’ orbitopathy (GO) is the main extrathyroidal manifestation of Graves’ disease, found in about 25% of patients at diagnosis, often mild and self-remitting. Removal of risk factors (refrain from smoking, correction of thyroid dysfunction, oral steroid prophylaxis after radioactive iodine therapy, antioxidant therapy with seleniomethionine) are fundamental to prevent progression of mild GO to more severe forms. In moderate-to-severe and active GO, intravenous glucocorticoids are the first-line treatment, second line treatments include cyclosporine, orbital radiotherapy, rituximab (controversial). Novel biologicals, such as teprotumumab and tocilizumab are under investigation. Rehabilitative surgery (orbital decompression, squint surgery, eyelid surgery) is often required. Thyroid dermopathy (pretibial myxedema) is a rare complication of Graves’ disease, usually observed in patients who also have severe GO. Topical glucocorticoids are usually effective. Thyroid acropachy (clubbing of fingers and toes, with swelling of hands and feet) is an extremely rare conditions, for which no treatment is available.

Hypertyroidism may be complicated by severe cardiovascular manifestations, such as tachyarrhythmias (most commonly atrial fibrillation), congestive heart failure, angina, particularly in the elderly or in patients with preexisting heart abnormalities. Prompt restoration of euthyroidism is, therefore, warranted, as well as specific treatments for the heart.

 

 

THYROID STORM

Thyroid (or thyrotoxic) storm is an acute, life-threatening syndrome due to an exacerbation of thyrotoxicosis. It is now an infrequent condition, because of earlier diagnosis and treatment of thyrotoxicosis, better pre- and postoperative medical management. However, acute exacerbation of thyrotoxicosis caused by intercurrent illness, especially infections, may still occur. Thyroid storm in the past most frequently occurred after surgery, but now it is usually a complication of untreated or partially treated thyrotoxicosis, rather than a postoperative complication.

Clinical pattern

Classic features of thyroid storm are indicative of a sudden and severe exacerbation of thyrotoxicosis, with fever, marked tachycardia, tremor, nausea and vomiting, diarrhea, dehydration, restlessness, extreme agitation, delirium or coma. Fever is typical and may be higher than 105.8 F (41 C). Patients may present with a true psychosis or a marked deterioration of previously abnormal behavior. Sometimes thyroid storm takes a strikingly different form, called apathetic storm, with extreme weakness, emotional apathy, confusion, absent or low fever

Signs and symptoms of multiple organ failure may be present. Delirium is one example. Congestive heart failure may also occur, with peripheral edema, congestive hepatomegaly, and respiratory distress. Marked sinus tachycardia or tachyarrhythmias, such as atrial fibrillation, are common. Liver damage and jaundice may derive from congestive heart failure or a direct action of thyroid hormone on the liver coupled with malnutrition (Chapter 10). Fever and vomiting may produce dehydration and prerenal azotemia. Abdominal pain may be a prominent feature. The clinical picture may be masked by a secondary infection such as pneumonia, a viral infection, or infection of the upper respiratory tract. Death may be caused by cardiac arrhythmia, congestive heart failure, hyperthermia, or other unidentified factors.

Storm is typically associated with Graves' disease, but it may occur in patients with toxic nodular goiter (1, 2). At present, although still life-threatening, death from thyroid storm is rarer if it is promptly recognized and aggressively treated in an intensive care unit. In recent nationwide studies from Japan mortality rate was >10% (3, 4).

Incidence

In Nelson and Becker's series reported in 1969 (5), there were 21 cases of thyroid storm among 2,329 admissions due to thyrotoxicosis (about 1%). Other series, which included all cases with fever of 38.3 C or more in the postoperative period, reported an incidence of thyroid storm as high as 10% of patients operated on (6). Few patients are now seen with the classic pattern of thyroid storm, but patients are occasionally encountered with marked accentuation of symptoms of thyrotoxicosis in conjunction with infection. The incidence of thyroid storm currently may currently be as low as 0.2 cases/100,000 population (3).

Cause

Thyroid storm classically began a few hours after thyroidectomy performed on a patient prepared for surgery by potassium iodide alone. Many such patients were not euthyroid and would not be considered appropriately prepared for surgery by current standards. Exacerbation of thyrotoxicosis is still seen in patients sent too soon to surgery, but it is unusual in the antithyroid drug-controlled patient. Thyroid storm occasionally occurs in patients operated on for some other illness while severely thyrotoxic. Severe exacerbation of thyrotoxicosis is rarely seen following 131-I therapy for hyperthyroidism; some of these may be defined as thyroid storm (7). Thyroid storm appears most commonly following infection (1), which seems to induce an escape from control of thyrotoxicosis. Pneumonia, upper respiratory tract infection, enteric infections, or any other infection can cause this condition. The decreased incidence of thyroid storm can be largely attributed to improved diagnosis and therapy. In most cases, thyrotoxicosis is recognized early and treated by measures of predictable therapeutic value. Patients are routinely made euthyroid before thyroidectomy or 131-I therapy (8). Using thionamides preoperatively, thyroid glands have only minimal amounts of stored hormones, thus minimizing thyroid hormone release due to manipulation.

Diagnosis

Diagnosis of thyroid storm is made on clinical grounds and involves the usual diagnostic measures for thyrotoxicosis. Semi-quantitative scales and related scores evaluating the presence and severity of clinical manifestations may be of some help in confirming the diagnosis (1, 3, 9). There are no peculiar laboratory abnormalities. Free T4 and, if possible, free T3 should be measured. Serum total T3 may be not particularly elevated or even normal, due to reduced T4 to T3 conversion as observed in nonthyroidal illness (1). Electrolytes, blood urea nitrogen (BUN), blood glucose, liver function tests, and plasma cortisol should be monitored.

Therapy

Thyroid storm is an endocrine emergency that has to be treated in an intensive care unit (Table 12-1).

Table 1. Treatment of Thyroid Storm

       

Supportive Measures

       

     1. Rest

     2. Mild sedation, or anticonvulsant therapy if convulsions occur

     3. Fluid and electrolyte replacement

     4. Nutritional support and vitamins as needed

     5. Oxygen therapy

     6. Nonspecific therapy as indicated

     7. Antibiotics

     8. Cardio-supportive

     9. Cooling

 

 Specific therapy

       

     1. Propranolol (20 to 200 mg orally every 6 hours, or 1 to 3 mg intravenously every 4 to 6 hours)

     2. Antithyroid drugs (150 to 250 mg PTU or 15 to 25 mg methimazole, every 6 hours)

     3. Potassium iodide (one hour after first dose of antithyroid drugs):

     4. 100 mg KI every 12 hours

     5. Dexamethasone (2 mg every 6 hours)

 

Possibly useful therapy

     1. Ipodate (Oragrafin) or other iodinated contrast agents, if available

     2. Plasmapheresis or exchange

     3. Oral T4 and T3 binding resins

     4. Dialysis

 

 

If drugs cannot be given orally (e.g., in the unconscious patient), they can be administered by naso-gastric tube or enemas (1). In some European countries intravenous preparations have been used (10).  If the thyrotoxic patient is untreated, an antithyroid drug should be given. PTU, 150-250 mg every 6 hours should be given, if possible, rather than methimazole, since PTU also prevents peripheral conversion of T4 to T3, thus more rapidly reduces circulating T3 levels. Methimazole (15-25 mg every 6 hours) can be given orally, or if necessary, the pure compound can be made up in a 10 mg/ml solution for parenteral administration. Methimazole is also absorbed when given rectally in a suppository. An hour after a thionamide has been given, iodide should be administered. A dosage of 100 mg twice daily is more than sufficient. Unless congestive heart failure contraindicates it, propranolol or other beta-blocking agents should be given at once, orally or parenterally in large doses, depending on the patient's clinical status. Permanent correction of thyrotoxicosis by either 131-I or immediate thyroidectomy should be postponed until euthyroidism is restored. Other supporting measures should fully be exploited, including sedation, oxygen, treatment for tachycardia or congestive heart failure, rehydration, multivitamins, occasionally supportive transfusions, and cooling the patient to lower body temperature down. Antibiotics may be given on the presumption of infection while results of culture are awaited.

The adrenal gland may be limited in its ability to increase steroid production during thyrotoxicosis. If there is any suspicion of hypoadrenalism, hydrocortisone (100-200 mg/day) or its equivalent should be given. The dose can rapidly be reduced when the acute process subsides. Pharmacological doses of glucocorticoids (2 mg dexamethasone every 6 h) acutely depress serum T3 levels by reducing T4 to T3 conversion. This effect of glucocorticoids is beneficial in thyroid storm and supports their routine use in this clinical setting. Propranolol controls tachycardia, restlessness, and other symptoms.

Usually rehydration, repletion of electrolytes, treatment of concomitant disease, such as infection, and specific agents (antithyroid drugs, iodine, propranolol, and corticosteroids) produce a marked improvement within 24 hours. A variety of additional approaches have been reported, but indications for their use are not well defined. For example, oral gallbladder contrast agents such as ipodate and iopanoic acid in doses of 1-2 g, which inhibit peripheral T4 to T3 conversion, might have value. Unfortunately, these agents are no longer available. Peritoneal dialysis can remove circulating thyroid hormone, and plasmapheresis can do likewise, but at the expense of serum protein loss. Orally administered ion-exchange resin (20-30g/day as Colestipol-HCl) can trap hormone in the intestine and prevent recirculation. These treatments are rarely needed.

Antithyroid treatment should be continued until euthyroidism is achieved, when a final decision regarding antithyroid drugs, surgery, or 131-I therapy can be made.

GRAVES’ ORBITOPATHY

Graves’ orbitopathy (GO) is the main and most frequent extrathyroidal manifestation of Graves’ disease, although it may less frequently occur in patients with Hashimoto’s thyroiditis or apparently without thyroid abnormalities (so-called Euthyroid Graves’ disease) (11-15).

Epidemiology

Fig. 1: Prevalence of GO in a series of 346 patients with newly diagnosed Graves’ hyperthyroidism. Moderate-to-severe GO includes one case of sight-threatening dysthyroid optic neuropathy (DON). Derived from Tanda ML et al. (17).

Data on the incidence of GO are limited (11, 14). In a population-based setting in USA, an adjusted rate of 16 cases per 100.000 per year in women and 2.9 cases per 100.000 in men was reported (16). In a recent study of a large cohort of newly diagnosed Graves’ patients, about 75% had no ocular involvement at diagnosis, only 6% had moderate-to-severe GO, and 0.3% showed sight-threatening GO due to dysthyroid optic neuropathy (DON) (17) (Figure 1). In a Danish population, moderate-to-severe GO showed an incidence of 16.1/million per year (women: 26.7; men: 5.4) (18). Ocular involvement is in most cases bilateral, although often asymmetrical, but it may be unilateral in up to 15% of cases (12, 14). As recently reviewed by the European Group on Graves’ Orbitopathy (EUGOGO), the overall prevalence of GO in Europe is about 10/10,000 patients, but the prevalence of its variants (hypothyroid GO, GO associated with dermopathy, GO associated with acropachy, asymmetrical or unilateral GO) is much lower, and recently euthyroid GO has been listed as a rare disease in Europe (19). The onset of GO apparently has a bimodal peak in the fifth and seventh decades of life, but eye disease may occur at any age (20). It is more frequent in women, but men tend to have a more severe disease (21-23), as suggested by a decrease in the female/male ratio from 9.3 in mild GO, to 3.2 in moderately severe GO, and 1.4 in severe GO (20). There is a close temporal relationship between the onset of GO and the onset of hyperthyroidism. In approximately 85% of cases GO and hyperthyroidism occur within 18 months of each other (20), although GO may both precede (about 20% of cases) or follow (about 40% of cases) the onset of hyperthyroidism (20).

The natural history of GO is poorly understood. However, in a longitudinal cohort study, spontaneous amelioration was observed in two thirds of cases, while ocular involvement did not change with time in 20% and progressed in 14% (22). The observation that mild GO rarely progresses and often spontaneously remits was recently confirmed by a large prospective study of patients with recent onset Graves’ hyperthyroidism (17) and summarized in a review of published studies (24). It is worth noting that GO seems to be less frequent than in the past. A review of the first 100 consecutive patients seen at the same joint thyroid-eye unit in 1960 and 1990 revealed a decrease in the proportion of Graves’ patients with clinical relevant GO from 57% to 32% (23); likewise, a reduction in the proportion of severe forms of GO compared to milder forms was observed (18), likely reflecting an earlier diagnosis and treatment of both hyperthyroidism and orbitopathy. It should be noted that a multicenter study carried out by the European Group on Graves’ Orbitopathy (EUGOGO) reported that 40% of GO patients had mild disease, 33% had moderate GO, and 28% had severe eye disease (25). It should be noted that these figures were clearly influenced by the fact that EUGOGO centers are all referral centers where it is likely to see more complicated cases of GO. Accordingly, a recent single-center study confirmed that most patients newly diagnosed with Graves’ disease have mild GO (26), although whether these forms are chronic remitting or a transient disease (27) or whether GO ever disappears completely (28) is unsettled. In summary, based on recent studies and reviews of the available literature, it can be concluded that GO is a rare disease, particularly in its severe expressions (19).

An important epidemiologic feature of GO is its relation with cigarette smoking (29,30). The prevalence of smokers among Graves’ women with orbitopathy is much higher than that in Graves’ women apparently without GO or in normal controls (Figure 2) (31). Smoking is a predictor of Graves’ hyperthyroidism, with a hazard ratio of 1.93 in current smokers, 1.27 in ex-smokers, and 2.65 in heavy smokers (32). In a case-control study, the odds ratio of cigarette smoking for Graves’ hyperthyroidism without GO was 1.7, but raised to 7.7 for Graves’ disease with GO (33). Whether passive smoking may have the same impact as active smoking is unsettled; however, in a recent European survey of GO in childhood, the highest prevalence of Graves’ children with GO was found in countries where the prevalence of smokers among teenagers was also highest: since >50% of children were <10 years of age, it is likely that passive smoking rather than active smoking influenced GO occurrence (34). Mechanisms whereby smoking may affect the development and course of GO are unclear. In addition to direct irritative effects and modulation of immune reactions in the orbit (35), smoking was associated with an increase in the orbital connective tissue volume as assessed by MRI (36), and with an increased adipogenesis and hyaluronic acid production in in vitro cultured orbital fibroblasts (37). Whatever the mechanism(s) involved, cigarette smoking is strong (probably the strongest) predictor of GO occurrence in patients with Graves’ hyperthyroidism (38).

Figure 2. Prevalence of smokers among women with Graves’ disease with (GO) or without (GD) associated orbitopathy. NTG: Non-toxic goiter; C: controls. Derived from Bartalena et al (31).

Pathogenesis

Clinical manifestations of GO reflect remodeling of the orbital space related to the enhanced orbital volume, due to an increase in retroocular fibroadipose tissue and swelling of extraocular muscles (39-41). Orbital tissues, including muscles, are infiltrated by inflammatory cells, including lymphocytes, mast cells, and macrophages. Proliferation of orbital fibroblasts and adipocytes, both in the retroocular space and in the perimysial space, is also associated with an increased production of glycosaminoglycans, which are the ultimate responsible for edematous changes both in the connective tissue and the muscles, owing to their hydrophilic nature. The relative contribution of the increase in fibroadipose tissue volume and extraocular muscle swelling is not always the same, and a predominance of either component may be observed in different patients with similar clinical features (42). Furthermore, the increase in orbital fat might be a rather late phenomenon (43). Because the orbit is a rigid, bony structure anteriorly limited by the orbital septum, the increased orbital volume deriving from cell proliferation, inflammatory infiltration and edema, results into enhanced intraorbital pressure, forward displacement of the globe (proptosis or exophthalmos), extraocular muscle dysfunction causing diplopia and/or strabismus, soft tissue changes with periorbital edema, conjunctival hyperemia and chemosis. If proptosis, which can be considered a form of spontaneous decompression, is severe, subluxation of the eye may occur. Proptosis is responsible for corneal exposure which may be particularly dangerous at night for the incomplete eyelid closure (lagophthalmos), and may result into sight-threatening corneal ulceration. The enlarged muscle volume may cause optic nerve compression (dysthyroid optic neuropathy), especially if the orbital septum is tight and proptosis is minimal. Optic nerve compression is particularly evident at the orbital apex and may be responsible for sight loss. Orbital inflammation and related anatomical changes may cause venous and lymphatic congestion that contribute to periorbital edema and chemosis. With time inflammation subsides and muscle fatty degeneration and fibrosis may contribute to further extraocular muscle restriction and strabismus, which, at this stage, can only be corrected by surgery.

GO is an autoimmune inflammatory disorder related to the thyroid and triggered by the migration of autoreactive T-helper cells into the orbit, which is infiltrated by CD4+ T cells and, to a lesser extent, CD8+ T cells, B cells, fibrocytes, mast cells, and macrophages (40, 41). Orbital fibroblasts are the main target and key effector cells in the disease (44). After recognition of one or more antigens (shared with the thyroid) on fibroblast surface, facilitated by HLA class II antigen expression on antigen-presenting cells (B cells, macrophages), CD4+ T cells secrete cytokines which activate CD8+ T cells, autoantibody-synthesizing B cells (45) and stimulate orbital fibroblasts (46).  Fibroblasts proliferate, may differentiate into myofibroblasts and adipocytes, accumulate and secrete hyaluronic acid (HA), synthesize and secrete chemoattractants (interleukin-16, RANTES, CXCL10) and a number of cytokines (interleukin-1, interleukin-6, interferon-g, tumor necrosis factor-a, interleukin-8, interleukin-10, platelet-derived growth factor, transforming growth factor-b), which concur to auto-maintain the inflammatory process (47). HA is hydrophilic and thereby attracts water and causes edema of the extraocular muscles and orbital tissue (40). Orbital fibroblasts can be differentiated based on the expression or lack of expression of a cell surface glycoprotein (thymocyte antigen-1, thy-1). Thy-1+ fibroblasts are mostly represented in extraocular muscles and produce HA, thereby contributing to extraocular muscle edema and enlargement, whereas  thy-1- fibroblasts are mainly present in connective tissue and may differentiate into adipocytes, thereby contributing to fibroadipose tissue expansion (40).

The TSH receptor (TSH-R), the ultimate cause of hyperthyroidism due to Graves’ disease, is likely the shared antigen responsible for GO. TSH-R expression has been shown in the orbital tissue of GO patients, both at the mRNA and protein levels (48, 49); however, TSH-R is also expressed in several other tissues not involved in Graves’ disease and orbitopathy (50), and, although at lower levels, in normal orbital fibroadipose tissue samples and cultures (51). On the other hand, BALB/c mice injected with spleen cells primed either with a TSH-R fusion protein or with TSH-R cDNA developed thyroiditis with blocking-type TRAb, but also showed orbital pathological changes (lymphocytic and mast cell infiltration, edema, presence of glycosaminoglycans) similar to those seen in human GO (52).  This model could not be reproduced in other laboratories. More recently a novel preclinical female mouse model of GO was established (53). In this model, some of the mice immunized by human TSH A-subunit by TSH receptor plasmid in vivo electroporation showed large infiltrates surrounding the optic nerve, increased adipogenesis, orbital muscle hypertrophy, exophtahlmos, and chemosis (53, 54). The role of the TSH-R seems to be supported also by other animal models (55-57). Another putative antigen involved in the pathogenesis of GO is the IGF-1 receptor. As recently reviewed (58), increased IGF-1 receptor levels have been reported in orbital fibroblasts as well as in B and T lymphocytes from Graves’ patients, and immunoglobulins displacing IGF-1 from its binding to the IGF-1 receptor have also been found in these patients (59). Colocalization of TSH receptor and IGF-1 receptor has been shown both in orbital fibroblasts and thyrocytes, suggesting that the two receptors might form a functional complex (60). Stimulation of the TSHR by the monoclonal TSHR-stimulating antibody, M22, could be inhibited by an IGF-1R-blocking monoclonal antibody in orbital fibroblasts (61).  Involvement of the IGF-1R is not specific for Graves’ disease, since it is implicated in other autoimmune diseases, such as rheumatoid arthritis (62). Thus, although involvement of the IGF-1R in the pathogenesis of GO seems likely (63), it is tempting to speculate, for the time being, that autoimmunity to the TSH receptor be the initiating mechanism, while subsequent increased expression of the IGF-1R might be fundamental to maintain ongoing reactions in the orbit (64). Interestingly, two recent reports found that, at variance with TRAb, only a minority of patients with GO have circulating antibodies to the IGF-1R (65, 66). This may be related to the low sensitivity and specificity of tests used to detect such antibodies (62). Alternatively, it may be conceived that IGF-1 (and/or IGF-1R antibodies) locally produced in the orbit be relevant for the pathogenesis of GO (67).

Other autoantigens, including several eye muscle antigens, acetylcholine receptor, thyroperoxidase, thyroglobulin, alpha-fodrin, have been proposed as putative shared antigens, but their true role is, to say the least, uncertain (68).

The role of genetic factors in the pathogenesis of GO is not very well defined, and no striking differences have been observed between Graves’ patients with or without associated GO (69-73). An association between GO and Major Histocompatibility Complex (MHC), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or intercellular adhesion molecule 1 gene polymorphisms has been looked for, but results are not unequivocal (74-76). GO likely stems from a complex interplay between endogenous factors and exogenous (environmental) risk factors (13, 77). The latter are probably more important and include cigarette smoking, thyroid dysfunction, and, in a subset of patients, radioiodine therapy for Graves’ hyperthyroidism (13, 77). The relationship between cigarette smoking and GO has been discussed above (see paragraph on Epidemiology). Both hyperthyroidism (78, 79) and hypothyroidism (80) seem to influence negatively the course of the orbitopathy. TRAb are independent risk factors for GO and can help to predict severity and outcome of eye disease (81). Radioiodine therapy for Graves’ hyperthyroidism is associated with GO progression in about 15% of cases, although this effect may be transient (82-86). This effect is more frequently observed in patients who already have GO prior to radioiodine therapy, smoke, have high TRAb levels, or whose post-radioiodine hypothyroidism is not promptly corrected by L-thyroxine replacement therapy (13, 77). Radioiodine-associated progression of GO can be prevented by a short course of prednisone (87). Lower doses of oral prednisone (0.2 mg/Kg bw) for 6 weeks are as effective in preventing RAI-associated progression of GO as higher doses used in the past (88). Neither thyroidectomy (typically partial) nor antithyroid drugs influence the course of the orbitopathy (89-91). The above observations have important practical implications in terms of GO prevention (Table 2), because GO patients should be urged to refrain from smoking, their thyroid dysfunction (both hyper- and hypothyroidism) should be promptly corrected, and, in the case of radioiodine therapy, a short course of oral prednisone should be administered (92).

Table 2. Risk factors for the occurrence/progression of Graves’ orbitopathy and preventive   measures

 

Risk factor

 

 

Preventive measure

 

 

Cigarette smoking

 

 

Refrain from smoking

 

 

Hyperthyroidism

 

 

Restore euthyroidism by antithyroid drugs and/or obtain a permanent control by thyroid ablation (thyroidectomy, radioiodine, both)

 

 

Hypothyroidism

 

 

Restore euthyroidism by L-thyroxine replacement therapy

 

 

Radioiodine therapy for hyperthyroidism

 

 

Give oral prednisone concomitantly with radioiodine administration. Avoid leaving the patient with untreated post-radioiodine hypothyroidism

 

High TSH-receptor antibody levels

 

 

Control hyperthyroidism as soon as possible

 

Oxidative stress Give a 6-month selenium course in mild GO

 

Clinical Features

Signs & Symptoms. Clinical features of GO include soft tissue changes, exophthalmos, extraocular muscle dysfunction, corneal abnormalities, and optic nerve involvement (Figures 3-6). The NOSPECS classification (Table 3) is a useful memory aid of GO abnormalities (93). Recommendations for GO assessment in clinical practice have recently been reviewed by EUGOGO (94) and other groups (VISA classification) (95). Soft tissue changes include eyelid edema and periorbital swelling, eyelid erythema, conjunctival hyperemia and chemosis, inflammation of the caruncle or plica: their assessment and grading can be done with the aid of a color atlas (96), which can be downloaded from EUGOGO website (www.eugogo.eu). Proptosis, i.e., protrusion of the eye (exophthalmos), is usually measured by Hertel exophthalmometer; normal values are usually less than 20 mm, but vary with race, age, gender, degree of myopia, and should be established in each center or institution. Extraocular muscle dysfunction is responsible for diplopia (double vision), which can be subjectively defined as intermittent (i.e., present only when fatigued or when first waking), inconstant (i.e., present only at extremes of gaze), or constant (i.e., present also in reading positions and primary gaze); objective assessment of extraocular muscle functioning can be done by several methods, including measurement of duction  in degrees (94). Palpebral aperture may be increased due to several factors, including upper and/or lower lid retraction, and proptosis. Lid retraction and proptosis are responsible for corneal exposure, which may lead to corneal epithelium damage (punctate keratopathy), corneal ulceration and perforation. The incomplete eye closure at night (lagophthalmos) and the absence of Bell’s phenomenon (no upward eye rotation on attempted eye closure) are risk factors for corneal damage (92, 94). Intraocular pressure is often increased, particularly in upward gaze, but this abnormality rarely progresses to true glaucoma. Dysthyroid optic neuropathy, due to optic nerve compression at the orbit apex by swollen extraocular muscles, or, less frequently, to optic nerve stretching in cases of marked proptosis or eye subluxation, is a sight-threatening expression of GO. It can be diagnosed by fundoscopy showing disc swelling, reduced visual acuity, abnormal color vision test, contrast sensitivity, perimetry, visual-evoked potentials, and pupillary responses (95).

Figure 3. Female patient with moderately severe GO. Note periorbital swelling, injection of conjunctival vessels, proptosis, marked lid retraction, and proptosis.

Figure 4. Male patient with moderately severe GO. Note marked periorbital swelling, conjunctival hyperemia, esotropia (strabismus) in the left eye.

Figure 5. Male patient with moderately severe GO. Note the superior eyelid edema, mild conjunctival vessel injection, marked proptosis, and marked upper lid retraction.

Figure 6. Female patient with severe GO. Note marked periorbital swelling, palpebral hyperemia, conjunctival hyperemia, proptosis (particularly in the left eye), caruncle edema. Eye motility was markedly reduced, lagophthalmos was present, there were two corneal ulcers in the left eye, and corneal punctate staining in the right eye, reduced visual acuity in the left eye (5/10). CT scan showed enlargement of extraocular muscles (particularly medial rectus and inferior rectus) in both eyes, but no relevant compression of the optic nerve at the orbit apex.

Table 3. NOSPECS classification of eye changes of Graves’ disease

 

Class

 

 

Grade

 

 

Symptoms and Signs

 

 

0

 

 

 

 

No symptoms or signs

 

 

1

 

 

 

 

Only signs (upper lid retraction, without lid lag or proptosis)

 

 

2

 

 

 

 

Soft tissue involvement with symptoms (excess lacrimation, sandy sensation, retrobulbar discomfort, and photophobia, but not diplopia);objective signs as follows:

 

 

 

 

0

 

 

absent

 

 

 

 

a

 

 

minimal (edema of conjunctivae and lids, conjunctival injection, and fullness of lids, often with orbital fat extrusion, palpable lacrimal glands, or swollen extraocular muscles beneath lower lids)

 

 

 

 

b

 

 

Moderate (above plus chemosis, lagophthalmos lid fullness)

 

 

 

 

c

 

 

marked

 

 

3

 

 

 

 

Proptosis associated with classes 2 to 6 only (specify if inequality of 3 mm or more between eyes, or if progression of 3 mm or more under observation)

 

 

 

 

0

 

 

absent (20 mm or less)

 

 

 

 

a

 

 

minimal (21-23 mm)

 

 

 

 

b

 

 

moderate (24-27 mm)

 

 

 

 

c

 

 

marked (28 mm or more)

 

 

4

 

 

 

 

Extraocular muscle involvement (usually with diplopia)

 

 

 

 

0

 

 

absent

 

 

 

 

a

 

 

minimal (limitation of motion, evident at extremes of gaze in one or more directions)

 

 

 

 

b

 

 

moderate (evident restriction of motion without fixation of position)

 

 

 

 

c

 

 

marked (fixation of position of a globe or globes)

 

 

5

 

 

 

 

Corneal involvement (primarily due to lagophthalmos)

 

 

 

 

0

 

 

absent

 

 

 

 

a

 

 

minimal (stippling of cornea)

 

 

 

 

b

 

 

moderate (ulceration)

 

 

 

 

c

 

 

marked (clouding, necrosis, perforation)

 

 

6

 

 

 

 

Sight loss (due to optic nerve involvement)

 

 

 

 

0

 

 

absent

 

 

 

 

a

 

 

minimal (disc pallor or choking, or visual field defect, vision 20/20 to 20/60)

 

 

 

 

b

 

 

moderate (disc pallor or choking, or visual field defect, vision 20/70 to 20/200)

 

 

 

 

c

 

 

marked (blindness, i.e., failure to perceive light; vision less than 20/200)

 

 

From Werner (93).

 

 

Symptoms of GO (Table 4) include, in addition to changes in ocular appearance related to periorbital swelling and proptosis, excess lacrimation, photophobia, grittiness, pain in or behind the eyes, either spontaneous or with eye movements, diplopia of different severity with or without strabismus, blurred vision, which may clear with blinking (due to excessive lacrimation) or covering one eye (reflecting extraocular muscle impairment), or may persist (probably reflecting optic neuropathy, particularly if associated with gray areas in the field of vision). In addition to reduced visual acuity, optic nerve involvement can be heralded by decreased color perception. Diplopia may be absent if extraocular muscle involvement is symmetrical in both eyes.

Table 4. Symptoms associated with Graves’ orbitopathy

       

1. Changes in eye appearance, particularly eyelid or periorbital swelling, eye bulging

2. Excessive lacrimation, often more pronounced on waking

3. Incomplete closure of eyes at night (lagophthalmos), as reported by the partner

4. Photophobia, need to protect eyes with dark lenses

5. Increased eye “sensitivity” to irritative factors other than light (e.g., wind, smoke, pollution)

6. Ocular discomfort, described as grittiness, foreign body or sandy sensation, often defined as “allergy”

7. Ocular pain, either related or unrelated to eye movements

8. Neck ache, with abnormal head posture (torcicullum)

9. Diplopia

a. Intermittent: present only when tired or on waking

b.     Inconstant: present only at extremes of gaze

c. Constant: present also in primary and reading positions

10. Blurred vision

a. Disappearing with blinking

b.     Not disappearing with blinking

11. Reduced color perception

 

 

 

Clinical manifestations of GO have a profound negative impact on quality of life and daily activities of affected individuals (98). By the use of general health-related quality of life (HRQL) questionnaires, such as the SF-36 or its shorter forms, it was shown that GO is associated with significant changes in several functions, including physical functioning, role functioning, social functioning, mental health, general health perception, and bodily pain (99). Interestingly, these changes in HRQL parameters were similar to those found in patients with inflammatory bowel disorders, and even more marked than those observed in patients with diabetes mellitus, heart failure or emphysema (99). Since HRQL questionnaires are broad and may not address items specific for a given disease, a GO-specific quality of life (GO-QoL) questionnaire was developed and validated in clinical studies (99, 100-102). This questionnaire (dowloadable from EUGOGO website at www.eugogo.eu) is composed of 16 questions, 8 concerning the consequences of diplopia and deceased visual acuity on visual functioning, and 8 regarding the consequences of changes in physical appearance on social functioning. The Go-QoL is a useful tool for self-assessment of treatment outcomes for GO (103).

Activity & Severity. Definition of GO severity is somehow arbitrary and may reflect different views (11, 23). According to the most recent EUGOGO definition (92),  mild GO is characterized by one or more of the following features: minor lid retraction (<2 mm), mild soft tissue involvement, exophthalmos <3 mm above normal for race and gender, transient or no diplopia, and corneal exposure; the above features usually have a minor impact on daily life to justify immmuno-suppression or surgical treatment; moderate-to-severe GO have any one or more of the following: lid retraction >2 mm, moderate or severe soft tissue involvement, exophthalmos >3 mm above normal for race and gender, inconstant or constant diplopia; Patients in this category have an impact on daily life as to justify immunosuppression (if GO is active) or surgical intervention (if GO is inactive); sight-threatening GO is due to dysthyroid optic neuropathy (DON) or corneal breakdown, and warrants immediate intervention (Table 5) (92). Assessment of severity is particularly relevant to decide on whether a given patient should be treated by aggressive treatments (either medical or surgical) or simply by local or general supportive measures (see below).The other important feature of GO is its activity. Although, as stated above, GO natural history is not completely understood, it is commonly accepted that GO undergoes an initial phase of activity, characterized by progressive exacerbation of ocular manifestations until a plateau phase is reached; GO then tends to remit spontaneously, but remission is invariably partial. In the inactive phase (burnt-out GO), only residual ocular manifestations are present (e.g., proptosis, strabismus due to muscle fibrotic changes), but inflammation has subsided and it is unlikely that it may flare up. It is unknown how long this process takes to be completed, but it is widely believed that it takes between 6 months and two years. Recognition of the different phases of the disease is important, because active disease, basically characterized by the presence of inflammation, can respond to immunosuppressive treatments, which are largely ineffective when GO is burnt-out. Different indicators have been proposed to assess GO activity, including short duration of treatment (<18 months), positivity of octreoscan, decreased extraocular muscle reflectivity at orbital ultrasound, prolonged T2 relaxation time at MRI, increased urinary glycosaminoglycan levels, but they lack sufficient specificity and accuracy. A useful tool to assess GO activity is represented by the Clinical Activity Score (CAS), which can be calculated very easily and is recommended by EUGOGO in the assessment of GO in routine clinical practice, in specialist multidisciplinary clinics, and for clinical trials (92). In its original formulation (104) it included 10 items, which were subsequently reduced to 7 when revised by an ad hoc Committee of the four sister thyroid societies (105). (Table 6). If one point is given to each item when present, CAS, which basically reflect eye inflammation, may range from 0 (absent activity) to 7 (maximal activity); GO is generally defined active if CAS is >3.

Table 5. Assessment of severity of Graves’ orbitopathy

Degree of Ocular involvement Features
Mild GO

Minor lid retraction (<2 mm), mild soft tissue involvement, exophthalmos <3 mm

above normal for race and gender, transient or no diplopia, corneal exposure

responsive to lubricants

Moderate-to-severe GO

Lid retraction >2 mm, moderate-to-severe soft tissue involvement, exophthalmos >3  mm

above normal for race and gender, inconstant or constant diplopia

Sight-threatening GO Presence of dysthyroid optic neuropathy and/or corneal breakdown

Derived from Bartalena & EUGOGO (92)

 

Table 6. Clinical Activity Score (CAS).

       

1. Spontaneous retrobulbar pain

2. Pain on eye movements

3. Eyelid erythema

4. Conjunctival injection

5. Chemosis

6. Swelling of the caruncle

7. Eyelid edema or fullness

 

 

 

One point is given to each item, if present. CAS is the sum of single scores, ranging from 0 (no activity) to 7 (maximal activity). Active GO: CAS>3

 

 

From Mourits et al (104), modified from an ad hoc Committee of the four Thyroid sister Societies (105).

 

 

Diagnosis

Diagnosis of GO is usually easy on clinical grounds and by careful ophthalmological examination. Although not necessary in most Graves’ patients, CT scan or MRI of the orbit confirm diagnosis by showing enlarged extraocular muscles (without involvement of the tendon) and/or increased orbital fibroadipose tissue (106). Modest extraocular muscle enlargement and increased fibroadipose tissue volume are often found in Graves’ patients without clinical manifestations of ocular involvement. Orbital imaging is very useful to detect signs of optic nerve compression, which support the diagnosis of optic neuropathy. Imaging is required in asymmetrical or, particularly, unilateral forms of GO, to rule out that proptosis, periorbital swelling, inflammation, or diplopia be due to disorders other than GO (12, 106). The latter include primary or metastatic orbital tumors, vascular abnormalities (e.g., carotid-cavernous sinus fistula, carotid aneurysm, cavernous sinus thrombosis, subarachnoid hemorrhage, subdural hematoma), granulomatous disorders, IgG4-related ophthalmic disease (107). Occasionally, angiograms or venograms may be required for diagnosis. Octreoscan may be useful to identify patients with active GO (106), but its role in clinical practice is limited, also in view of its high cost.

Management

Management of GO is based on a multidisciplinary approach which involves endocrinologists, ophthalmologists, orbit surgeons, radiologists and radiotherapists. In a survey of GO management based on a questionnaire distributed among members of the European Thyroid Association, European Society of Ophthalmic Plastic and Reconstructive Surgery, and European Association of Nuclear Medicine, 96% of responders stated that a multidisciplinary setting for GO management is valuable, although 21% of patients were in the end not treated in a multidisciplinary setting (108). The therapeutic approach to a GO patient should be based on both severity and activity of the disease, the former being the feature to assess first.

Mild GO. Most patients have mild GO, which does not require particularly aggressive treatments and often is self-limiting (92, 110, 111). If GO activity is modest, simple local measured can be suggested to obtain symptomatic relief until GO is burnt-out (Table 7). Photophobia can be mitigated by sunglasses; grittiness due to corneal exposure can be controlled by artificial tears and topical lubricants, particularly indicated in the presence of lagophthalmos; the latter may require taping the eyelids shut at night; eyelid retraction can be controlled (with a variable degree of success) by b-blocking drops (useful for the increased intraocular pressure) or by botulinum toxin injections (112); elevation of the bed may be helpful to reduce periorbital swelling due to congestion; mild diplopia often is controlled by prisms (if they are tolerated). Reassurance is an important issue, and the patient must be informed that his/her eye disease is unlikely to progress to more severe forms, usually stabilizes, and often ameliorates spontaneously. Control of thyroid dysfunction is fundamental, because progression often is associated with hyper- or hypothyroidism (12, 92); refrain from smoking is also essential, because it is associated with a decreased chance of developing proptosis and diplopia (113), and decreases the likelihood to develop severe GO (29). Patients who do not succeed to quit smoking by themselves, should be helped by professional stop-smoking clinics, organizations, groups, where they can receive counseling, behavioral therapies, pharmacological treatments. In a subset of patients with mild GO, the impact of GO on the quality of life is so pronounced as to justify the risk of immunosuppression (or surgery) as for moderate-to-severe GO (92). A recent randomized controlled trial performed by EUGOGO in a large cohort of patients with mild GO showed that selenium supplementation for 6 months has beneficial effects on mild GO compared with placebo and can often prevent its progression to more severe forms (114). Thus, selenium, for its anti-inflammatory and immunomodulatory actions, should be considered both as a therapeutic tool for mild GO and a preventive measure (115). Whether selenium is also useful as an adjuvant therapy in patients with moderate-to-severe GO is presently unsettled.

Table 7. Management of mild Graves’ orbitopathy

 

Sign and/or symptom and/or associated problem

 

 

Therapeutic measure

 

 

Photophobia

 

 

Sunglasses

 

 

Foreign body or sandy sensation

 

 

Artificial tears and ointments

 

 

Eyelid retraction

 

 

alpha-blocking eye drops. Botulinum toxin injections

 

 

Increased intraocular pressure

 

 

alpha-blocking eye drops

 

 

Lagophthalmos

 

 

Nocturnal eye taping

 

 

Thyroid dysfunction (hyper/hypo)

 

 

Restoration of euthyroidism, as appropriate

 

 

Smoking

 

 

Refrain from smoking

 

 

Anxiety about possible further progression

 

 

Reassurance on the natural history of mild GO

Management of clinical picture and prevention of progression Selenium supplementation

 

Moderate-to-severe GO. Management of moderate-to-severe GO depends not only on severity, but also on activity of the orbitopathy (Table 8). Medical treatment is likely to be beneficial in patients with active GO, with florid signs and symptoms of inflammation, recent-onset extraocular muscle dysfunction, recent progression of the ocular abnormalities as a whole. On the contrary, in long-standing GO, with chronic proptosis and residual, stable diplopia and/or strabismus, but no no evidence of inflammation, medical treatment has little chances to produce favorable effects, and the surgical, rehabilitative approach is preferable (92). Dysthyroid optic neuropathy, the most severe expression of the orbitopathy, is a clinical, sight-threatening emergency, which requires immediate treatment. If there is no response to medical treatment (high-dose intravenous glucocorticoids), orbital decompression is warranted (92).

Table 8. Management of moderate-to-severe Graves’ orbitopathy

Treatment Validity
Glucocorticoids Established
Orbital Radiotherapy Established
Orbital Decompression Established
Rehabilitative Surgery (squint surgery, eyelid surgery) Established
Somatostatin analogs Non-validated (octreotide, lanreotide)
Intravenous immunoglobulins Non-validated
Cyclosporine Limited applications
Antioxidants Selenium (validated only for mild GO)
Rituximab Two conflicting and small randomized clinical trials
Etanercept Limited data (non-validated in randomized clinical trials)
Mycophenolate Two randomized clinical trials with positive results
Teprotumumab One randomized clinical trial with positive results
Thyroid ablation by 131-I and/or surgery Controversial

 

Glucocorticoids are the mainstay in the medical treatment of GO (12, 92, 116, 117). They have been used for decades because of their anti-inflammatory effects, but also because they exert immunosuppressive actions useful to control the course of the orbitopathy (12, 92, 116, 117). The latter include interference with the function of T and B lymphocytes, decreased recruitment of neutrophils and macrophages, down-regulation of adhesion molecules, inhibition of cytokine secretion, inhibition of glycosaminoglycan secretion. Locally (subconjunctivally or retrobulbarly) given glucocorticoids are less effective than systemically given glucocorticoids ) (11, 92), although favorable responses in terms of improvement of diplopia and reduction in extraocular muscle dysfunction have been reported with in a recent randomized clinical trial of periocular injections of triamcinolone acetate (118) . Glucocorticoids have for a long time been given mostly orally. This route of administration has several drawbacks: high doses are required every day (e.g., prednisone 60-100 mg daily as a starting dose, or equivalent doses of other steroids) (11), treatment lasts for several months (at least 5-6 months), recurrences are frequent upon drug tapering or withdrawal, side effects (particularly Cushing’s syndrome) are frequent (12, 92, 116, 117). In the last 20 years the intravenous route has become the most commonly used (101) and currently represents the first-line treatment for moderate-to-severe and active GO (92). Intravenous glucocorticoids are more effective, with a rate of favorable responses reported until few years ago of about 80-90% versus 60-65% with oral glucocorticoids (117, 119), and better tolerated than oral glucocorticoids (120). As a proof of principle, in a placebo-controlled, randomized trial, intravenous methylprednisolone (four cycles at a dose of 500 mg for 3 consecutive days at 4-week intervals) effected inflammatory changes and extraocular muscle dysfunction in 5 of 6 patients (83%) compared to only one of 11 placebo-treated patients (121) Glucocorticoids are most effective on soft tissue, inflammatory changes, recent-onset extraocular muscle dysfunction, and dysthyroid optic neuropathy, whereas proptosis and long-lasting eye muscle impairment are less responsive (11, 117). However, it should be noted that severe liver damage, heralded by a marked rise in serum concentrations of hepatic enzymes, was noted in 7 of about 800 treated patients (approximately 0.8%), three of whom died (122). The causes of this hepatotoxicity are unclear, but might include direct liver toxicity of glucocorticoids, precipitation of virus-induced hepatitis, sudden reactivation of the immune system upon drug withdrawal leading to autoimmune hepatitis. Statins are not a risk factor for liver damage associated with intravenous glucocorticoid pulse therapy for GO (123). The cumulative dose of glucocorticoids might also be important, since no cases of liver damage were reported in a recent randomized clinical trial in which lower, but equally highly effective, doses of glucocorticoids were employed (124). In this trial, the cumulative dose of methylprednisolone was 4.5 grams, subdivided in 12 weekly 2-hour infusions (500 mg for the first 6 infusions, 250 mg for the remaining 6 infusions) (124). A questionnaire-based surgery carried out among members of the European Thyroid Association showed a wide heterogeneity in the regimens of intravenous glucocorticoid therapy for moderate-to severe and active GO (125). A recent mullticenter, randomized clinical trial of a large cohort of patients with moderate-to-severe and active GO showed that a cumulative dose of about 7.5 g of methylprednisolone was associated with more favorable treatment outcomes than lower doses (about 5 g or 2.25 g), but also caused adverse events more frequently (126). Thus, the cumulative dose should somehow be tailored to the severity and activity of GO, reserving the highest dose to patients with most severe expressions of the disease.  In any case, the current recommendation is that the cumulative dose of glucocorticoids per course should not exceed 8 grams (92). Response to intravenous glucocorticoids may occur early in the course of the intravenous course, but also later; accordingly, the lack of response after the first 5-6 infusions is not an indication to stop the treatment (127). Adverse events of high-dose glucocorticoid treatment remain a relevant issue (128). Accordingly, patients should be treated in specialized centers under strict medical surveillance (92). Intravenous glucocorticoid treatment of Graves’ ophthalmopathy is not associated with secondary adrenocortical insufficiency. (129, 130),presumably because it is given for a limited period and intermittently.

Orbital radiotherapy is the other non-surgical mainstay in the management of GO (131). The rationale for its use and the indications are quite similar to those of glucocorticoids; in addition, irradiation exploits the radiosensitivity of T lymphocytes which infiltrate the orbit (131). Irradiation is currently carried out by linear accelerators, using a cumulative dose of 20 Gray fractionated in 10 daily 2-Gray doses over a 2-week period (131), although other regimens (and lower doses) might be equally effective (132). Favorable responses have been reported in about 60% of treated patients (131). Recent years have witnessed a lively debate on the true effectiveness of orbital radiotherapy (133, 134). However, the results of several randomized studies confirmed, with one exception, its efficacy (132, 135-138). In addition, orbital radiotherapy is a safe procedure devoid of relevant short-term and long-term side effects or complications (139, 140). Preexisting retinopathy associated with diabetes mellitus or hypertension represents a contraindication to its use (92). As for glucocorticoids, orbital radiotherapy is mostly effective on soft tissue inflammatory changes and recent-onset extraocular muscle dysfunction (131). Orbital radiotherapy can be used either alone or in combination with glucocorticoids. The association exploits the prompter effect of glucocorticoids and the more sustained action of irradiation; in two randomized prospective studies, combined therapy (using oral glucocorticoids) proved to be more effective than either treatment alone (141, 142). Whether the combination of intravenous glucocorticoids and orbital radiotherapy is more effective than intravenous glucocorticoids alone is presently unsettled. For this reason the recent guidelines for the management of moderate-to-severe and active suggest the combination of orbital radiotherapy and oral glucocorticoids as a second-line treatment in the case of a partial or absent response to intravenous glucocorticoids (92).

Cyclosporine, used in GO for its immunosuppressive properties, has been reported in only two randomized and controlled studies (143, 144) . Cyclosporine has a lower efficacy than glucocorticoids as a single-agent therapy, although a combination of both drugs might be more effective than either treatment alone (143, 144). Thus, the use of cyclosporine might be maintained in patients who are relatively resistant to glucocorticoids in whom persistent GO activity warrants continuing medical intervention (145). Side effects of cyclosporine are not negligible and should be carefully considered. As for orbital radiotherapy, no studies have compared the combination of cyclosporine and intravenous glucocorticoids. Therefore, the association of oral glucocorticoids and cyclosporine is considered an alternative option when the response to intravenous glucocorticoids is poor (92).

Rituximab is a CD20+ B-cell depleting monoclonal antibody originally used for B-cell non-Hodgkin lymphoma, but then utilized for autoimmune B-cell (and T-cell) driven autoimmune disorders. This drug has been used for GO in small and uncontrolled preliminary studies (reviewed in refs. 146 and 147). The rationale for using rituximab is sound, based on our understanding of pathogenesis of GO. The available data suggest that rituximab may have favorable effects on moderate-to-severe and active GO, with relatively low adverse event rate (146, 147). Two small, single-center, randomized clinical trials have recently been published. They produced conflicting results, because the first one (148) showed no difference in the effect of rituximab compared to placebo (148), whereas the second one (149) showed that rituximab was as effective as intravenous glucocorticoids in inactivating GO and was not associated with a flare-up of the disease, not infrequently seen after withdrawal of glucocorticoids. The reason for this conflicting results remains elusive and larger multicenter studies are warranted to clarify this issue (150), nevertheless rituximab may be considered as a possible second-line treatment in the case of a partial or absent response to intravenous glucocorticoids (92). Rituximab is contraindicated in patients with overt or impending dysthyroid optic neuropathy (92).

Two recent randomized clinical trials have evaluated the effect of mycophenolate, an immunosuppressant agent inhibiting both T cells and B cells, widely used for the prevention of organ transplant rejection, but also for autoimmune disorders. The first, single-center study showed that mycophenolate mofetil was more effective than intravenous glucocorticoids on moderate-to-severe and active GO, but the experimental design had several limitations (151). The second, large, multicenter study carried out by EUGOGO demonstrated that the combination of mycophenolate sodium and intravenous glucocorticoids was more effective than intravenous glucocorticoids alone (152). The safety profile at the dose used (720 mg/day for 6 months) appears to be good (153). Therefore mycophenolate sodium may represent a useful tool for the management of GO.

Teprotumumab is a monoclonal antibody inhibiting the IGF-1 receptor. Because the latter seems to be involved in the pathogenesis of GO (154), teprotumumab might play a role in the management of the disease. A recent placebo-controlled randomized clinical trial showed that teprotumumab is more effective than placebo in reducing the CAS (155). The most striking effect was marked reduction in the exophthalmos (155), a feature of GO which is very poorly modified by whatever medical treatment. This study is not devoid of limitations and further studies are warranted before it can be included among the tools for the management of active and moderate-to-severe GO (156).

Orbital decompression is a milestone in the management of GO. It is aimed at increasing the space available for the increased orbital content by removing part of the bony walls of the orbit and/or the orbital fibroadipose tissue (157). It is indicated in patients who have impending sight loss due to optic neuropathy and do not respond promptly to intravenous glucocorticoids (157). Other important indications for decompressive surgery are represented by corneal damage due to eyeball exposure in patients with marked proptosis, or by recurrent subluxation of the globe, which may stretch the optic nerve and cause sight loss (157). In recent years, thanks to the improved surgical techniques and the diminished surgical risk, the indications for orbital decompression have expanded, including also correction of residual cosmetic problems (157, 158). Several techniques of orbital decompression are available, aimed at removing part of one, two, three or four orbital walls (floor, roof, lateral wall, medial wall) as well as part of the retroorbital fibroadipose tissue. The different surgical options should be discussed with the patient, as well possible complications of the procedure, particularly the de novo occurrence or worsening of diplopia, particularly frequent after extensive removal of the orbital floor (157, 159). Removal of fibroadipose tissue can be done together with or without bone removal, but removal of fat alone is associated with a lower reduction of proptosis (159).

Rehabilitative surgery includes surgery for strabismus or eyelid retraction. Extraocular muscle surgery is aimed at correcting residual diplopia after medical and/or surgical treatment of GO. Timing of surgery is crucial, because it should not be performed when GO is active, but when it has been inactive for 6 months (160, 161). The goal of eye muscle surgery is to align the eyes, avoiding abnormal head posture and restoring single binocular vision in primary and reading positions; multiple operations may be required to achieve this goal. Eyelid surgery may rarely be an emergency procedures in patients with exposure keratitis and corneal ulcerations, but it usually is carried out to correct eyelid malposition after medical treatment or orbital decompression. Eyelid surgery usually constitutes the last step of rehabilitation (161).

Thyroid ablation. The question of whether in a patient with GO, Graves’ hyperthyroidism should be treated by non-ablative (i.e., thionamides) or ablative (i.e., radioiodine therapy, thyroidectomy, both) therapy is unanswered (162, 163). Supporters of thyroid ablation justify this approach by mentioning the pathogenic link between thyroid and orbit: removal of thyroid-orbit shared antigen(s) and autoreactive T lymphocytes might be beneficial to the eye (162); supporters of non-ablative thyroid treatment suggest that control of thyrotoxicosis by antithyroid drugs may be associated with a reduction of autoimmune phenomena which might be reflected by an amelioration of ocular conditions; furthermore, once triggered, GO might proceed independently of thyroid treatment (163). Two retrospective studies showed that total thyroid ablation (thyroidectomy followed by radioiodine therapy, as in thyroid cancer) was associated with an improvement of clinical GO (164, 165). A recent randomized, controlled clinical trial demonstrated that, as compared to total thyroidectomy alone, total thyroid ablation is followed by a better outcome of GO in patients given intravenous glucocorticoids (166). A follow-up study of this cohort recently showed that in the long run total thyroid ablation was no better than total thyroidectomy alone, although it was associated with a prompter achievement of the best possible result obtainable by medical treatment and with an earlier possibility to submit the patient to rehabilitative surgery (147). Other cohort studies emphasized the opportunity to postpone definitive treatment of hyperthyroidism until GO is cured (168, 169). Thus, the optimal thyroid treatment in patients with GO still is a dilemma (170-172).

THYROID DERMOPATHY AND ACROPACHY

Thyroid dermopathy (also called pretibial myxedema or localized myxedema) is an uncommon extrathyroidal manifestations of Graves’ disease (less frequently of chronic autoimmune thyroiditis) (13, 173). It almost always occurs in Graves’ patients who also have GO. In a review of 178 consecutive patients with thyroid dermopathy, only 4 patients had no evidence of eye disease (174). However, in a community-based epidemiologic study, only 4% of GO patients also had thyroid dermopathy, although the latter was more frequent in patients with severe GO (175). It is more common in older than in younger patients, with a large preponderance in women (176). Skin lesions are edematous and thickened plaques, typically localized in the pretibial area; however they can be less frequently found in other skin areas, such as feet, toes, upper extremities, shoulders, upper back, nose. Prevalent localization in the pretibial area is related to mechanical and dependent position. The occurrence of lesions in less common sites is often preceded (triggered?) by local trauma (177, 178). There can be three clinical types: nodular, diffuse, and elephantias-like.(179, 180) (Figures 7-9).

Fig. 7: Thyroid dermopathy. Courtesy of Dr. Vahab Fatourechi, Mayo Clinic

 

Fig. 8: thyroid dermopathy. Courtesy of Dr. Vahab Fatourechi, Mayo Clinic

 

Fig. 9: Thyroid dermopathy. Courtesy of Dr. Vahab Fatourechi, Mayo Clinic

Histopathologically, skin lesions are characterized by the accumulation of activated fibroblasts (and, to a lesser extent, mast cells), with a markedly increased production of glycosaminoglycans in the dermis and subcutaneous tissues (181). Whereas in normal skin approximately 5% of the acid mucopolysaccharides are hyaluronic acid, in pretibial myxedema this amount increases to 90%. Glycosaminoglycans are responsible for fluid retention, subsequent compression and occlusion of lymphatic vessels, and lymphedema (182). Thus, as in GO, fibroblasts seem to play a central role in the pathogenesis of localized myxedema. This notion is further supported by the finding of limited variability of T cell receptor V gene usage in pretibial myxedema, pointing to a primary immune response of antigen-specific T lymphocytes (183). Furthermore, as is the case with acropachy, lymphocytes do recognize local fibroblasts. IgG from patients with pretibial myxedema was shown to stimulate proteoglycan synthesis by human skin fibroblasts (182). As for GO, TSH-R has been implicated in the pathogenesis of localized myxedema. TSH-R is expressed in peripheral skin fibroblasts from patients with localized myxedema, both at the mRNA and protein level (13). However, TSHR is expressed also in skin from normal subjects (183, 184). Likewise, TSH-R immunoreactivity was detected in cultured fibroblasts from pretibial myxedema, although the specificity of this finding remains to be established. As mentioned above, IgG from Graves’ patients with localized myxedema was reported to stimulate glycosaminoglycan production in cultured skin fibroblasts (183), but this data is not unequivocal, because IgG from normal subjects were equally effective in other studies (185). To summarize, although pathogenic mechanisms remain to be fully elucidated, localized myxedema appear to result from autoimmune reactions leading to fibroblast proliferation and increased glycocosaminoglycan secretion.

From a clinical standpoint, localized myxedema presents as light-colored (sometimes yellowish brown) skin lesions, frequently with an orange peel texture (Figures 7-9). Skin lesions may be characterized by hyperpigmentation and hyperkeratosis. They usually represent only a cosmetic problem and are asymptomatic, but sometimes they may be associated with itching and pain, or may be functionally important, e.g., they may cause problems to wear shoes, especially the elephantiasic form of localized myxedema. Localized myxedema may remain stable, but frequently improves with time, partially or completely (13). Many cases of mild localized myxedema do not require any treatment, but in moderately severe lesions or when there is cosmetic concern, topical glucocorticoids applied with occlusive plastic dressing produce beneficial effects in a relevant proportion of patients (13). If necessary, treatment is repeated until clinical remission occurs (13). When localized mxedema is severe and extensive, steroid pulse therapy, or decongestive physiotherapy, a combination of manual lymphatic drainage, bandaging, exercise, and scrupulous skin care, may be tried (13). No substantial effect was reported by long-term octreotide treatment in three patients with localized myxedema (186). Two studies on the use of intravenous IgG in a small number of patients have reported discrepant effects (reviewed in 13). Thus, measures such as compression bandaging and topical glucocorticoids still are the most cost effective treatments for localized myxedema.

Acropachy is a very uncommon extrathyroidal expression of Graves’ disease, usually associated with severe GO (13) and localized myxedema (13), thus reflecting severity of the autoimmune process. It seems more common in women than in men (13). It is characterized by clubbing of fingers (Fig. 10) and toes, with concomitant soft-tissue swelling of hands and feet. These abnormalities are usually painless and may be asymmetric (13). As for GO, there seems to be a strong relation with cigarette smoking. X-ray of affected sites shows soft-tissue swelling and subperiosteal bone formation. There is currently no treatment that can solve the esthetic and (less frequent) functional abnormality of thyroid acropachy, which occasionally may remit spontaneously in the long-term.

Fig. 10: Thyroid acropachy. Courtesy of Dr. Vahab Fatourechi, Mayo Clinic

 

CLINICAL ABNORMALITIES OF THE HEART

The biochemical actions of thyroid hormone on the heart are described in Chapter 10.

Hyperthyroidism is usually associated with relevant cardiovascular symptoms and changes in cardiovascular hemodynamics (186, 187).Thyrotoxicosis increases the demands on the heart both by chronotropic and inotropic alterations. Cardiac output is markedly increased owing to increased stroke volume and rapid heart rate (186, 187). It is possible that the metabolic efficiency of heart muscle is decreased (186, 187). Irritability of the heart is increased. Investigation with stress echocardiography shows in hyperthyroidism impaired chronotropic, contractile, and vasodilatatory cardiovascular reserves, that are reversible upon conversion to euthyroidism (188). In a recent, large, matched case-control study, cardiovascular symptoms and signs, including palpitations, chest pain, dyspnea, cough, orthopnea, displaced apex, cardiac murmur, chest wheeze/crepitus were much more frequent in hyperthyroid patients than in controls, and some of them persisted despite effective restoration of euthyroidism by antithyroid drug treatment (182). This common finding of cardiovascular alterations in hyperthyroid patients may result from thyroid hormone excess itself, by hyperthyroidism-related worsening of preexisting cardiovascular disorders, or by the occurrence of novel cardiovascular abnormalities (186, 187). The importance of cardiovascular abnormalities is underscored by the observation that mortality of hyperthyroid patients is increased, mainly due to cardiovascular events (189, 190). Similar conclusions were reached also in a community-based study of elderly people (191), in which, however, definition of hyperthyroidism was based on the finding of low/suppressed serum TSH, which may not necessarily reflect thyroid hormone excess, but rather be the result of non-thyroidal illness syndrome.

Mitral valve prolapse was found more commonly in hyperthyroid patients (43%) than in controls (18%) (190). This increased incidence might be due to increased adrenergic tone, autoimmunity, or the augmented cardiac output associated with thyrotoxicosis. Most patients with thyrotoxicosis are adults. Many, especially those with toxic nodular goiter, are in the 50- to 70-years age group, which has a relatively high incidence of organic heart disease anyway (192). Thus, it is not surprising that cardiac abnormalities are prominent among the symptoms of thyrotoxicosis. Frequent premature beats and paroxysmal tachycardia sometimes appear in thyrotoxic patients and may be disturbing to the patient. Atrial fibrillation occurs in thyrotoxicosis with or without preexisting heart disease, but it is more frequent in older patients (193), probably reflecting an increase in the prevalence of underlying cardiac abnormalities of ischemic or different origin (194). It may be paroxysmal or persistent during the thyrotoxic period. Attempts to correct this arrhythmia to normal in patients with persistent atrial fibrillation are usually unsuccessful while they are hyperthyroid. Once euthyroidism has been restored, atrial fibrillation may revert spontaneously or may be converted pharmacologically or by electroconversion. About two-thirds of patients undergo spontaneous reversion to sinus rhythm after receiving therapy for thyrotoxicosis, usually within 4 months; later on, spontaneous conversion is unlikely (195). It is wise to always evaluate thyroid function in clinically euthyroid patients with atrial arrhythmias with or without heart disease, because in about 20% of patients TSH tests and/or FT4 point to an overactive thyroid and in 50% of these patients normal sinus rhythm resumes after treatment with antithyroid drugs (195). It is widely accepted that subclinical hyperthyroidism is associated, in individuals aged 60 years or more with a 3-to-5-fold increased risk of developing atrial fibrillation (195).

Congestive heart failure is a frequent complication in thyrotoxic patients with pre-existing organic heart disease, particularly if old (196-198). In the elderly hyperthyroid patient, cardiac symptoms may so dominate the clinical picture that diagnosis of thyrotoxicosis may be overlooked. Careful attention should be given to this possibility in all patients with congestive heart failure, especially if goiter is detected (196). Congestive heart failure may occur in patients who have no detectable preexisting organic heart disease (199). Overt hyperthyroidism may cause ventricular dilatation and persistent tachycardia, which may lead to heart failure and fatal events (200). It is often difficult to establish whether an underlying heart disease is present in a hyperthyroid patient who also has a disorder of rhythm, a cardiac murmur, or congestive heart failure, because all these conditions may be ascribed to thyrotoxicosis per se. It is frequently gratifying to observe normalization of cardiac findings once euthyroidism has been restored.

In hyperthyroidism, owing to the increased metabolic demand, angina can be worsened if pre-existing, or induced de novo (186, 201, 202). Evidence of myocardial lactate production when the heart is paced at an accelerated rate (203), and normal coronary arteries are found at angiography after episodes of angina or infarction (203), have suggested that changes in thyrotoxicosis are due to an imbalance between O2 demand and supply rather than to arterial obstruction. This possibility is corroborated by the finding that coronary artery spasm of an otherwise normal vessel may occur during thyrotoxicosis (202).

Cardiac abnormalities found in Graves' disease often are entirely reversible, except that longstanding atrial fibrillation due to hyperthyroidism is not always convertible after euthyroidism is restored. It has become evident that even in the mildest forms of thyrotoxicosis subtle cardiac abnormalities may be present. Thus, in patients with so-called "subclinical" thyrotoxicosis, i.e. suppressed TSH and normal serum free T4 and T3 concentrations, due to multinodular, autonomous goiter or TSH-suppressive T4 treatment, mean basal 24-h heart rate is increased, there is an augmented risk of atrial premature beats and atrial fibrillation, and left ventricular function and wall thickness are increased (204).There is controversy whether TSH suppressive T4 treatment leads to functional cardiac abnormalities (204, 205).

Treatment of heart failure in the presence of thyrotoxicosis does not differ from its treatment in euthyroid patients, but it may be more difficult. Rest, salt restriction, diuretic therapy, digitalization and administration of afterload-reducers, like angiotensin converting enzyme (ACE) inhibitors, betablockers, aldosterone antagonists and other specific measures, are in order (186, 196). Larger than normal doses of digoxin are required, but there is probably no change in the toxic-to-therapeutic dose ratio. Atrial fibrillation may be controlled by digoxin, propranolol, or both. Electroconversion is usually successful only after thyrotoxicosis has been resolved for a few months (206).

Hyperthyroidism should be controlled as expeditiously as possible. Congestive heart failure is a contraindication to operation. Most patients with thyrotoxicosis and clinically relevant heart disease are now treated with RAI. This treatment may be preceded by a 3-to-6-month course of antithyroid drug therapy to deplete their glands of stored thyroid hormone, a program that lessens any chance of an exacerbation of the heart disease caused by a radioiodine-induced release of thyroid hormone from the gland. Administration of 131I followed by antithyroid drugs, and potassium iodide or ipodate, that also inhibit T4 to T3 conversion, may be used in severely ill patients in whom a prompt response is needed. This method is described in Chapter 11.

Propranolol has been used successfully in the control of tachycardia, and also in patients with congestive heart failure if tachycardia appeared to be adding substantially to the problem. In these instances, possible depression of myocardial contractility by the drug was outweighed by the benefit derived from controlling the rate. In such circumstances, one must proceed with caution and often digoxin should be added.

References

  1. Klubo-Gwiezdzinska J, Wartoksky L 2012 Thyroid emergencies. Med Clin North Am 96: 385-403.
  2. Carroll R, Matfin G 2010 Endocrine and metabolic emergencies: thyroid storm. Ther Adv Endocrinol Metab 1: 139-145.
  3. Akamizu T, Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T et al. 2012 Diagnostic criteria, clinical features, and incidence of thyroid storm based on nationwide surveys. Thyroid 22: 661-679
  4. Akamizu T 2018 Thyroid storm: a Japanese perspective. Thyroid 28: 32-40
  5. Nelson NC, Becker WF 1969 Thyroid crisis: diagnosis and treatment. Ann Surg 170:263-273.
  6. Kobayashi C, Sasaki H, Kosuge K, Miyakita Y, Hayakawa M, Suzuki A, Abe E, Suzuki K, Aizawa Y.2005 Severe starvation hypoglycemia and congestive heart failure induced by thyroid crisis, with accidentally induced severe liver dysfunction and disseminated intravascular coagulation. Intern Med. 44:234-239.
  7. McDermott MT, Kidd GS, Dodson LE, Hofeldt FD 1983 Radioiodine-induced thyroid storm. Am J Med 75:353-359.
  8. Bartalena L 2013 Diagnosis and management of Graves disease: a global overview. Nat Rev Endocrinol 9: 724-734.
  9. Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, Tsuboi K, Kanamoto N, Otani H, Furukawa Y, Teramukai S, Teramukai S, Akamizu T 2016 2016 guidelines for the management of thyroid storm from the Japan Thyroid Association and Japan Endocrine Society (First edition). Endocrine J 63: 1025-1024.
  10. Thomas DJ, Hardy J, Sarwar R, Banner NR, Mumani S, Lemon K, Hillson RM 2006 Thyroid storm treated with intravenous methimazole in patients with gastrointestinal dysfunction. Br J Hosp Med. 67: 492-493.
  11. Bartalena L, Pinchera A, Marcocci C 2000 Management of Graves’ ophthalmopathy: reality and perspectives. Endocr Rev 21: 168-199.
  12. Bartalena, Tanda ML 2009 Clinical Practice: Graves’ ophthalmopathy. N Engl J Med 360: 994.1001.
  13. Bartalena L, Fatourechi V 2014 Extrathyroidal manifestations of Graves’ disease: a 2014 update. J Endocrinol Invest 37: 691-700.
  14. Smith TJ, Hegedus L 2016 Graves’ disease. N Engl J Med 375: 1552.1575.
  15. Bartalena L, Chiovato L, Vitti P 2016 Management of hyperthyroidism due to Graves’ disease: frequently asked questions and answers (if any). J Endocrinol Invest 39: 1105-1114.
  16. Bartley GB 1994 The epidemiological characteristics and clinical course of ophthalmopathy associated with autoimmune thyroid disease in Olmsted County, Minnesota. Trans Am Ophthalmol Soc 92: 477-588.
  17. Tanda ML, Piantanida E, Liparulo L, Veronesi G, Lai A, Sassi L, Pariani N, Gallo D, Azzolini C, Ferrario M, Bartalena L 2013. Prevalence and natural history of Graves’ orbitopathy in a large series of patients with newly diagnosed Graves’ hyperthyroidism seen at a single center. J Clin Endocrinol Metab 98: 1443-1449.
  18. Laurberg P, Berman DC, Bulow Pedersen I, Andersen S, Carlé A 2012 Incidence and clinical presentation of moderate to severe Graves’ orbitopathy in a Danish population before and after iodine fortification of salt. J Clin Endocrinol Metab 97: 2325-2332.
  19. Perros P, Hegedus L, Bartalena L, Marcocci C, Kahaly GJ, Baldeschi L et al. 2017 Graves orbitopathy as a are disease in Europe: a European Group on Graves’ Orbitopathy (EUGOGO) position statement. Orphanet J Rare Dis 12: 72.
  20. Marcocci C, Bartalena L, Bogazzi F, Panicucci M, Pinchera A 1989 Studies on the occurrence of ophthalmopathy in Graves’ disease. Acta Endocrinol (Copenh) 120: 473-478.
  21. Perros P, Crombie AL, Matthews JNS, Kendall-Taylor P 1993 Age and gender influence the severity of thyroid associated ophthalmopathy: a study of 101 patients attending a combined thyroid-eye clinic. Clin Endocrinol (Oxf) 38: 367-372.
  22. Perros P, Crombie AL, Kendall-Taylor P 1995 Natural history of thyroid-associated ophthalmopathy. Clin Endocrinol (Oxf) 42: 45-50.
  23. Kendall-Taylor P, Perros P 1998 Clinical presentation of thyroid associated orbitopathy. Thyroid 8: 427-428.
  24. Piantanida E, Tanda ML, Lai A, sassi L, Bartalena L 2013 Prevalence and natural history of Graves’ orbitopathy in the XXI century. J Endocrinol Invest 36: 444-449.
  25. Prummel MF, Bakker A, Wiersinga WM, Baldeschi L, Mourits MP, Kendall-Taylor P, Perros P, Neoh C, Dickinson AJ, Lazarus JH, Lane CM, Heufelder AE, Kahaly GJ, Pitz S, Orgiazzi J, Hullo A, Pinchera A, Marcocci C, Sartini MS, Rocchi R, Nardi M, Krassas GE, Halkias A 2003 Multi-center study on the characteristics and treatment strategies of patients with Graves’ orbitopathy: the first European Group on Graves’ Orbitopathy experience. Eur J Endocrinol 148: 491-495.
  26. Bartalena L, Masiello E, Magri F, Veronesi G, Bianconi F, Zerbini F et al. 2016 The phenotype of newly diagnosed Graves’ disease in Italy in recent years is milder than in the past: results of a large observational longitudinal study. J Endocrinol Invest 39: 1445-1451
  27. Anagnostis P, Boboridis K, Adamidou F, Kita M 2017 Natural course of mild Graves’ orbitopathy: is it a chronic remitting or a transient disease? J Endocrinol Invest 40: 257-261.
  28. Sabini E, Leo M, Mazzi B, Rocchi R, Latrofa F, Nardi M et al. 2017 Does Graves’ orbitopathy ever disappear? Answers to an old question. Eur Thyroid J 6: 263-270.
  29. Bartalena L, Bogazzi F, Tanda ML, Manetti L, Dell’Unto E, Martino E 1995 Cigarette smoking and the thyroid. Eur J Endocrinol 133: 507.512.
  30. Wiersinga WM 2013 Smoking and thyroid. Clin Endocrinol (Oxf) 79: 145-151.
  31. Bartalena L, Martino E, Marcocci C, Bogazzi F, Panicucci M, Velluzzi F, Loviselli A, Pinchera A 1989 More on smoking habits and Graves’ ophthalmopathy. J Endocrinol Invest 12: 733-737.
  32. Holm IA, Manson JAE, Michels KB, Alexander EK, Willett WC, Utiger RD 2005 Smoking and other lifestyle factors and the risk of Graves’ hyperthyroidism. Arch Intern Med 165: 1606-1611.
  33. Prummel MF, Wiersinga WM 1993 Smoking and risk of Graves’ disease. JAMA 269: 479-482.
  34. Krassas GE, Segni M, Wiersinga WM 2005 Childhood Graves’ ophthalmopathy: results of a European questionnaire study. Eur J Endocrinol 153: 515-521.
  35. Bartalena L, Marcocci C, Pinchera A 2002 Graves’ ophthalmopathy: a preventable disease? Eur J Endocrinol 146: 457-461.
  36. Szucs-Farkas Z, Todt J, Kollar J, Galuska L, Barman KD, Boda J, Leovey A, Varga J, Ujhelyi B, Szabo J, Berta A, Nagy EV 2005 Volume changes in intra- and extraorbital compartments in patients with Graves’ ophthalmopathy: effect of smoking. Thyroid 15: 146-151.
  37. Cawood TJ, Moriarty P, O’Farrelly C, O’Shea D 2007 Smoking and thyroid-associated ophthalmopathy: a novel explanation of the bilogical link. J Clin Endocrinol Metab 92: 59-64.
  38. Khong JJ, Finch S, De Silva C, Rylander S, Cgarig JE, Selva D, Ebeling PR 2016 Risk facrors for Graves’ orbitopathy: the Australian Thyroid-Associated Research (ATOR) study. J Clin Endocrinol Metab 101: 2711-2720.
  39. Smith TJ 2010 Pathogenesis of Graves’ orbitopathy: a 2010 update. J Endocrinol Invest 33: 414-421.
  40. Bahn RS 2015 Current insights into the pathogenesis of Graves’ ophthalmopathy. Horm Metab Res 43: 773-776.
  41. Salvi M, Berchner-Pfannschmidt U, Ludgate M 2017 Pathogenesis. In (Wiersinga WM, Kahaly GJ (Eds.) Graves’ orbitopathy – A multidisciplinary approach: questions and answers. Karger, Basel, pp. 41-60
  42. Prabhakar BS, Bahn RS, Smith TJ 2003 Current perspective on the pathogenesis of Graves’ disease and ophthalmopathy. Endocr Rev 24: 802-835.
  43. Potgieser PW, Wiersinga WM, Regensburg NI, Wiersinga WM 2015 Some studies on the natural history of Graves’ orbitopathy: increase in orbital fat is a rather late phenomenon. Eur J Endocrinol 173: 149-153.
  44. Wang Y, Smith TJ 2014 Current concepts in the molecular pathogenesis of thyroid-associated ophthalmopathy. Invest Ophthalmol Vis Sci 55: 1735-1748.
  45. Ajjan AR, Weetman AP 2004 New understanding of the role of cytokines in the pathogenesis of Graves’ ophthalmopathy. J Endocrinol Invest 27: 237-245.
  46. Feldon SE, Park DJJ, O’Loughlin CW, Nguyen VT, Landskroner-Eiger S, Chang D, Thatcher TH, Phipps RP 2005 Autologous T-lymphocytes stimulate proliferation of orbital fibroblasts derived from patients with Graves’ ophthalmopathy. Invest Ophthalmol Vis Sci 46: 3913-3921.
  47. Khong JJ, McNab AA, Ebeling PR, Craig JR, Silva D 2016 Pathogenesis of thyroid eye disease: review and update on molecular mechanisms. Br J Ophthalomol 100: 142-150.
  48. Bahn RS 2004 TSH receptor expression in orbital tissue and its role in the pathogenesis of Graves’ ophthalmopathy. J Endocrinol Invest 27: 216-220.
  49. Agretti P, De Marco G, De Servi M, Marcocci C, Vitti P, Pinchera A, Tonacchera M 2005 Evidence for protein and mRNA expression in fibroblasts from patients with thyroid-associated ophthalmopathy (TAO) after adipocytic differentiation. Eur J Endocrinol 152: 777-784.
  50. Agretti P, Chiovato L, De Marco G, Marcocci C, Mazzi B, Sellari-Franceschini S, Vitti P, Pinchera A, Tonacchera M 2002 Real-time PCR provides evidence for thyrotropin receptor mRNA expression in orbital as well as in extraorbital tissues. Eur J Endocrinol 147: 733-739.
  51. Garrity JA, Bahn RS 2006 Pathogenesis of Graves’ ophthalmopathy: implications for prediction, prevention, and treatment. Am J Ophthalmol 142: 147-153.
  52. Ludgate M, Baker G 2004 Inducing Graves’ ophthalmopathy. J Endocrinol Invest 27: 211-215.
  53. Moshkelgosha S, So P-W, Deasy N, Diaz-Cano S, Banga JP 2013 Cutting edge: retrobulbar inflammation, adipogenesis, and acute orbital congestion in a preclinical female mouse model of Graves’ orbitopathy induced by thyrotropin receptor plasmid-in vivo electroporation. Endocrinology 154: 3008-3015.
  54. Banga JP, Moshkelgosha S, Berchner-Pfannschmidt U, Eckstein A 2015 Modeling Graces’ orbitopathy in experimental Graves’ disease. Horm Metab Res 47: 797-803.
  55. Nagayama Y, Nakahara M, Abiru N 2015 Animal models of Graves’ disease and Graves’ orbitopathy. Current Opin Endocrinol Diabetes Care 22: 381-386.
  56. Tu Y, Wang Y, Ding L, Zhang Y, Wu W 2016 Development of a novel thyroid function fluctuated animal model for thyroid-associated ophthalmopathy. PLoS One 11: e0148595
  57. Ungerer M, Fassbender J, Li Z, Munch G, Holthoff H-p 2017 Review of mouse models of Graves’ disease and orbitopathy – Novel treatment by induction of tolerance. Clin Rev Allerg Immunol 52: 182-193.
  58. Smith TJ, Huetwell FGL, Hegedus L, Douglas RS 2012 Role of insulin-like growth factor-1 (IGF-1) pathway in the pathogenesis of Graves’ orbitopathy. Best Pract Res Clin Endocrinol Metab 26: 291-302.
  59. Weightman DR, Perros P, Sherif IH, Kendall-Taylor P 1993 Autoantibodies to IGF-1 binding sites in the thyroid-associated phthalmopathy. Autoimmunity 16: 251-257
  60. Tsui S, Naik V, Hoan N, Hwang CJ, Afifiyan NF, Hikim Sinha A, Gianoukakis AG, Douglas RS, Smith TJ 2008 Evidence for an association between thyroid-stimulating hormone and insulin like growth factor 1 receptors: a tale of two antigens implicated in Graves’ disease. J Immunol 181: 4397-4405.
  61. Kumar S, Iyer S, Bauer H, Coenen M, Bahn RS 2012 A stimulatory thyrotropin receptor antibody enhances hyaluronic acid synthesis in Graves’ orbital fibroblasts: inhibition by an IGF-I receptor blocking antibody. J Clin Endocrinol Metab 97: 1681-1687
  62. Pritchard J, Tsui S, Horst N, Cruikshank WW, Smith TJ 2004 Synovial fibroblasts from patients with rheumatoid arthritis, like fibroblasts from Graves’ disease, express high levels of IL-16 when treated with Igs against insulin-like growth factor-1 receptor. J Immunol 173: 3564-3569.
  63. Smith TJ 2013 Is IGF-I receptor a target for autoantibody generation in Graves’ disease? J Clin Endocrinol Metab 98: 515-518.
  64. Wiersinga WM 2011 Autoimmunity in Graves’ ophthalmopathy: the result of an unfortunate marriage between TSH receptors and IGF-1 receptors? J Clin Endocrinol Metab 96: 2386-2394.
  65. Minich WB, Dehina N, Welsink T, Schwiebert C, Morgenthaler NG, Kohrle J, Eckstein A, Schomburg L 2013 Autoantibodies to the IGF1 receptor in Graves’ orbitopathy. J Clin Endocrinol Metab 98: 752-760.
  66. Varewijck AJ, Boelen A, Lamberts SJ, Fliers E, Jofland LJ, Wiersinga WM, Janssen JAMJL 2013 Circulating IgGs may modulate IGF-I receptor stimulating activity in a subset of patients with Graves’ ophthalmopathy. J Clin Endocrinol Metab 98: 769-776.
  67. Song D, Wang R, Zhong Y, Li W, Li H, Dong F 2012 Locally produced insulin-like growth factor-1 by orbital fibroblasts as implicative pathogenic factor rather than systemically circulated IGF-1 for patients with thyroid-associated ophthalmopathy. Graefes Arch Clin Exp Ophthalmol 250: 433-440.
  68. Bartalena L 2005 Graves’ ophthalmopathy: search for shared autoantigen(s) continues. J Endocrinol Invest 28: 396-397.
  69. Vaidya B, Kendall-Taylor P, Pearce SHS 2002 The genetics of autoimmune thyroid disease. J Clin Endocrinol Metab 87: 5385-5397.
  70. Brand OJ, Gough SCL 2010 Genetics of thyroid autoimmunity and the role of the TSHR. Mol Cell Endocrinol 322: 135-143.
  71. Brix TH, Hegedus L 2012 Twin studies as a model for exploring the aetiology of autoimmune thyroid disease. Clin Endocrinol (Oxf) 76: 457-464.
  72. Tomer Y 2014 Mechanisms of autoimmune thyroid diseases: from genetics to epigenetics. Annu Rev Pathol 9: 147-156.
  73. Marinò M, Latrofa F, Menconi F, Chiovato L, Vitti P 2015 Role of genetic and non-genetic factors in the etiology of Graves’ disease. J Endocrinol Invest 38: 283-294.
  74. Tomer Y 2010 Genetic susceptibility to autoimmune thyroid disease: past, present, future. Thyroid 20: 715-725.
  75. Yin X, Latif R, Bahn RS, Davies TF 2012 Genetic profiling in Graves’ disease: further evidence for lack of a distinct genetic contribution to Graves’ orbitopathy. Thyroid 22: 730-735.
  76. Juriecka-Lubieniecka B, Ploski R, Kula D et al 2014 Association between polymorphisms in the TSHR gene and Graves’ orbitopathy 2014 PLoS One 9: e102653
  77. Bartalena L 2012 Prevention of Graves’ ophthalmopathy. Best Pract Res Clin Endocrinol Metab 26: 371-379.
  78. Prummel MF, Wiersinga WM, Mourits MP, Koornneef L, Berghout A, van der Gaag R. 1989 Amelioration of eye changes of Graves’ ophthalmopathy by achieving euthyroidism. Acta Endocrinol (Copenh) 121: 185-189.
  79. Prummel MF, Wiersinga WM, Mourits MP, Koornneef L, Berghout A, van der Gaag R. 1990 Effect of abnormal thyroid function on the severity of Graves’ ophthalmopathy. Arch Intern Med 150: 1098-1101.
  80. Karlsson AF, Westermark K, Dahlberg PA, Jansson R, Enksson P 1989 Ophthalmopathy and thyroid stimulation. Lancet 2: 691.
  81. Eckstein AK, Plicht M, Lax H, Neuhauser M, Mann K, Lederbogen S, Heckmann C, Esser J, Morgenthaler NG 2006 Thyrotropin receptor autoantibodies are independent risk factors for Graves’ ophthalmopathy and help to predict severity and outcome of the disease. J Clin Endocrinol Metab 91: 3464-3470.
  82. Tallstedt L, Lundell G, Torring O, Wallin G, Ljunggren J-G, Blomgren H, Taube A & the Thyroid Study Group. 1992 Occurrence of ophthalmopathy after treatment for Graves’ hyperthyroidism. N Engl J Med 326: 1733-1738.
  83. Bartalena L, Marcocci C, Bogazzi F, Manetti L, Tanda ML, Dell’Unto E, Bruno-Bossio G, Nardi M, Bartolomei MP, Lepri A, Rossi G, Martino E, Pinchera A 1998 Relation between therapy for hyperthyroidism and the course of Graves’ ophthalmopathy. N Engl J Med 338: 73-78.
  84. Traisk F, Tallstedt L, Abraham-Nordling M, Andersson T, Berg G, Calissendorff J, Hallengren B, Hdner P, Lantz M, Nystrom E, Ponjavic V, Taube A, Torring O, Wallin G, Asman P, Lundell G 2009 Thyroid-associated ophthalmopathy after treatment for Graves’ hyperthyroidism with antithyroid drugs or iodine-131. J Clin Endocrinol Metab 94: 3000-3007.
  85. Acharya SH, Avernell A, Philip S, Burr J, Bevan JS, Abraham P 2008 Radioiodine therapy (RAI) for Graves’ disease (GD) and the effect on the ophthalmopathy: a systematic review. Clin Endocrinol (Oxf) 69: 943-950.
  86. Li HX, Xiang N, Hu WK, Jiao XL 2016 Relation between therapy options for Graves’ disease and the course of Graves’ ophthalmopathy: a systematic review and meta-analysis. J Endocrinol Invest 38: 1225-1233.
  87. Bartalena L, Marcocci C, Bogazzi F, Panicucci M, Lepri A, Pinchera A 1989 Use of corticosteroids to prevent progression of Graves’ ophthalmopathy after radioiodine therapy for hyperthyroidism. N Engl J Med 321: 1349-1352.
  88. Lai A, Sassi L, Compri E, Marino F, Sivelli P, Piantanida E, Tanda ML, Bartalena L 2010 Lower dose prednisone prevents radioiodine-associated exacerbation of initially mild or absent Graves’ orbitopathy: a retrospective cohort study. J Clin Endocrinol Metab 95: 1333-1337.
  89. Bartalena L, Baldeschi L, Dickinson A, Eckstein A, Kendall-Taylor P, Marcocci C, Mourits M, Perros P, Boboridis K, Boschi A, Currò N, Daumerie C, Kahaly GJ, Krassas GE, Lane CM, Lazarus JH, Marinò M, Nardi M, Neoh C, Orgiazzi J, Pearce S, Pinchera A, Pitz S, Salvi M, Sivelli P, Stahl M, von Arx G, Wiersinga WM 2008 Consensus statement of the European Group on Graves’ Orbitopathy (EUGOGO) on management of GO. Eur J Endocrinol 158: 273-285.
  90. Bartalena L, Baldeschi L, Dickinson J, Eckstein A, Kendall-Taylor P, Marcocci C, Mourits M, Perros P, Boboridis K, Boschi A, Currò N, Daumerie C, Kahaly GJ, Krassas GE, Lane CM, Lazarus JH, Marinò M, Nardi M, Neoh C, Orgiazzi J, Pearce S, Pinchera A, Pitz S, Salvi M, Sivelli P, Stahl M, von Arx G, Wiersinga WM 2008 Consensus statement of the European Group on Graves’ Orbitopathy (EUGOGO) on management of Graves’ Orbitopathy. Thyroid 18: 333-346.
  91. Marcocci C, Bruno-Bossio G, Manetti L, Tanda ML, Miccoli P, Iacconi P, Bartolomei MP, Nardi M, Pinchera A, Bartalena L 1999 The course of Graves’ ophthalmopathy is not influenced by near-total thyroidectomy. Clin Endocrinol (Oxf) 51: 503-508.
  92. Bartalena L, Baldeschi L, Boboridis K et al 2016 The 2016 European Thyroid Association/European Group on Graves’ Orbitopathy guidelines for the management of Graves’ orbitopathy. Eur Thyroid J 5: 9-26.
  93. Werner SC 1977 Modification of the classification of the eye changes of Graves’ disease: recommendations of the ad hoc committee of the American Thyroid Association. J Clin Endocrinol Metab 44: 203-206.
  94. The European Group on Graves’ Orbitopathy (EUGOGO): Wiersinga WM, Perros P, Kahaly GJ, Mourits MP, Baldeschi L, Boboridis K, Boschi A, Dickinson AJ, Kendall-Taylor P, Krassas GE, Lane CM, Lazarus JH, Marcocci C, Marinò M, Nardi M, Neoh C, Orgiazzi J, Pinchera A, Pitz S, Prummel MF, Sartini MS, Stahl M, von Arx G 2006 Clinical assessment of patients with Graves’ orbitopathy: the European Group on Graves’ Orbitopathy recommendations to generalists, specialists and clinical researchers. Eur J Endocrinol 155: 387-389.
  95. Dolman PJ 2012 Evaluating Graves’ orbitopathy. Best Pract Res Clin Endocrinol Metab 26: 229-248.
  96. Dickinson AJ, Perros P 2001 Controversies in the clinical evaluation of thyroid-associated orbitopathy: use of a detailed protocl with comparative photographs for objective assessment. Clin Endocrinol (Oxf) 55: 283-303.
  97. McKeag D, Lane C, Lazarus JH, Baldeschi L, Boboridis K, Dickinson AJ. Hullo AI, Kahaly G, Krassas G, Marcocci C, Marinò M, Mourits MP, Nardi M, Neoh C, Orgiazzi J, Perros P, Pinchera A, Pitz S, Plummer MF, Sartini MS, Wiersinga WM  2007 Clinical features of of dysthyroid optic neuropathy: a European Group on Graves’ Orbitopathy (EUGOGO) survey. Br J Ophthalmol 91: 455-458.
  98. Wiersinga WM 2017 Quality of life. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 260-274.
  99. Gerding MN, Terwee CB, Dekker FW, Koornneef L, Prummel MF, Wiersinga WM 1997 Quality of life in patients with Graves’ ophthalmopathy is markedly decreased: measurement by the Medical Outcomes Study instrument. Thyroid 7: 885-889.
  100. Terwee CB, Gerding MN, Dekker FW, Prummel MF, Wiersinga WM 1998 Development of a disease-sepcific quality of life questionnaire for patients with Graves’ ophthalmopathy. Br J Ophthalmol 82: 773-779.
  101. Kahaly GJ, Petrak F, Hardt J, Pitz S, Egle UT 2005 Psychosocial morbidity of Graves’ orbitopathy. Clin Endocrinol (Oxf) 63: 395-402.
  102. Terwee CB, Dekker FW, Mourits MP, Gerding MN, Baldeschi L, Kalmann R, Prummel MF, Wiersinga WM 2001 Interpretation and validity of changes in scores in the GO-QOL after different treatments. Clin Endocrinol (Oxf) 54: 391-398.
  103. Terwee CB, Dekker FW, Mourits MP, Gerding MN, Baldeschi L, Kalmann R, Prummel MF, Wiersinga WM 2001 Interpretation and validity of changes in scores in the GO-QOL after different treatments. Clin Endocrinol (Oxf) 54: 391-398.
  104. Mourits M, Koornneef L, Wiersinga WM, Prummel MF, Berghout A, van der Gaag R 1989 Clinical criteria for the assessment of disease activity in Graves’ ophthalmopathy: a novel approach. Br J Ophthalmol 73: 639-644.
  105. Pinchera A, Wiersinga WM, Glinoer D, Kendall-Taylor P, Koornneef L, Marcocci C, Schleusener H, Romaldini J, Niepominiscze F, Solomon D, Garrity J, Gorman CA 1992 Classification of eye changes of Graves’ disease. Thyroid 2: 235-236.
  106. Pitz S, Muller-Forell W 2017 Orbital imaging. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 61-73.
  107. Bartalena L, Chiovato L 2014 Graves’-like orbitopathy: do not forget IgG4-related disease. J Endocrinol Invest 37: 1233-1235.
  108. The European Group of Graves’ Orbitopathy: Perros P, Baldeschi L, Boboridis K, Dickinson AJ, Hullo A, Kahaly GJ, Kendall-Taylor P, Krassas GE, Lane CM, Lazarus JH, Marcocci C, Marinò M, Mourits MP, Nardi M, Orgiazzi J, Pinchera A, Pitz S, Prummel MF, Wiersinga WM 2006 A questionnaire survey on the management of Graves’ orbitopathy in Europe. Eur J Endocrinol 155: 207-211.
  109. Bartalena L, Marcocci C, Pinchera A. 1997 Treating severe Graves’ ophthalmopathy. Bailliere’s Clin Endocrinol Metab 11: 521-536
  110. Tanda ML, Piantanida E, Liparulo L, Veronesi G, Lai A, Sassi L, Pariani N, Gallo D, Azzolini C, Ferrario M, Bartalena L 2013 Prevalence and natural hostory of Graves’ orbitopathy in a large series of patients with newly diagnosed Graves’ hyperthyroidism seen at a single center. J Clin Endocrinol Metab 98: 1443-1449.
  111. Bartalena L, Masiello E, Magri F et al. 2016 The phenotype of newly diagnosed Graves’ disease in Italy in recent years is milder than in the past: results of a large observational longitudinal study. J Endocrinol Invest 39: 1445-1451.
  112. Boboridis K, Anagnostis P 2017 Local treatment modalities. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 202-206.
  113. Pfeilschifter J, Ziegler R 1996 Smoking and endocrine ophthalmopathy: impact of smoking severity and current vs lifetime cigarette consumption. Clin Endocrinol (Oxf) 45: 477-481.
  114. Marcocci C, Kahaly GJ, Krassas GE, Bartalena L, Prummel M, Stahl M, Altea MA, Nardi M, Pitz S, Boboridis K, Sivelli P, von Arx G, Mourits MP, Baldeschi L, Bencivelli W, Wiersinga W and the European Group on Graves’ Orbitopathy 2011 Selenium and the course of mild Graves’ orbitopathy. N Engl J Med 364: 1920-1931.
  115. Marcocci C, Bartalena L 2013 Role of oxidative stress and selenium in Graves’ hyperthyroidism and orbitopathy. J Endocrinol Invest 36 (suppl. to no. 10): 15-20.
  116. Bartalena L, Marcocci C, Tanda ML, Piantanida E, Lai A, Marinò M, Pinchera A 2005 An update on medical management of Graves’ ophthalmopathy. J Endocrinol Invest 28: 469-478.
  117. Zang S, Ponto KA, Kahaly GJ 2011 Intravenous glucocorticoids for Graves’ orbitopathy: efficacy and morbidity. J Clin Endocrinol Metab 96: 320-332.
  118. Ebner R, Devoto MH, Weil D, Bordaberry M, Mir C, Martinez H, Bonelli L, Niepomniszcze H 2004 Treatment of thyroid associated ophthalmopathy with periocular injections of triamcinolone. Br J Ophthalmol 88: 1380-1386.
  119. Bartalena L 2005 Glucocorticoids for Graves’ ophthalmopathy: how and when. J Clin Endocrinol Metab 90: 5497-5499
  120. Marcocci C, Bartalena L, Tanda ML, Manetti L, Dell’Unto E, Rocchi R, Barbesino G, Mazzi B, Bartolomei MP, Lepri P, Cartei F, Nardi M, Pinchera A 2001 Comparison of the effectiveness and tolerability of intravenous or oral glucocorticoids associated with orbital radiotherapy in the management of severe Graves’ ophthalmopathy: results of a prospective, single-blind, randomized study. J Clin Endocrinol Metab 86: 3562-3567.
  121. van Geest RJ, Sasim IV, Koppeschaar HPF, Mourits MP 2008 Methylprednisolone pulse therapy for patients with moderately severe Graves’ orbitopathy: a prospective, randomized, placebo-controlled study. Eur J Endocrinol 158: 229-237.
  122. Marinò M, Morabito E, Brunetto MR, Bartalena L, Pinchera A, Marcocci C 2004 Acute and severe liver damage associated with intravenous glucocorticoid pulse therapy in patients with Graves’ ophthalmopathy. Thyroid 14: 403-406.
  123. Sabini E, Sisti E, Coco B, Leo M, Ionni I, Menconi F et al 2016 Statins are not a risk factor for liver damage associated with intravenous glucocorticoid pulse therapy for Graves’ orbitopathy. J Endocrinol Invest 39: 1323-1327.
  124. Kahaly GJ, Pitz S, Hommel G, Dittmar M 2005 Randomized, single blind trial of intravenous versus oral steroid monotherapy in Graves’ orbitopathy. J Clin Endocrinol Metab 90: 5234-5240.
  125. Marcocci C, Watt T, Altea MA, Rasmussen AK, Feldt-Rasmussen U, Orgiazzi J, Bartalena L and the European Group on Graves’ Orbitopathy 2012 Fatal and non-fatal adverse events of glucocorticoid therapy for Graves’ orbitopathy: a questionnaire survey among members of the European Thyroid Association. Eur J Endocrinol 166: 247-253.
  126. Bartalena L, Krassas GE, Wiersinga W, Marcocci C, Salvi M, Daumerie C, Bournaud C, Stahl M, Sassi L, Veronesi G, Azzolini C, Boboridis KG, Mourits MP, Soeters MR, Baldeschi L, Nardi M, Currò N, Boschi A, Bernard M, von Arx G and the European Group on Graves’ Orbitopathy 2012 Efficacy and safety of three different cumulative doses of intravenous methylprednisolone for moderate to severe and active Graves’ orbitopathy. J Clin Endocrinol Metab 97: 4454-4463
  127. Bartalena L, Veronesi G, Krassas GE, Wiersinga WM, Marcocci C, Marinò M et al 2017 Does early response to intravenous glucocorticoids predict the final out come in patients with moderate-to-severe and active Graves’ orbitopathy? J Endocrinol Invest 40: 547-553.
  128. Zang S, Ponto KA, Kahaly GJ 2011 Dose of intravenous steroids and therapy outcome in Graves’ orbitopathy. J Endocrinol Invest 34: 876-880.
  129. Jespersen S, Nygaard B, Ostergaard Kristensen L 2015 Methyprednisolone pulse treatment of Graves’ ophthalmopathy is not associated with secondary adrenocortical insufficiency. Eur Thyroid J 4: 222-225.
  130. Giotaki Z, Fountas A, Tsirouki T, Bargiota A, Tigas S, Tsatsoulis A 2015 Adrenal reserve following treatment of Graves’ orbitopathy with intravenous glucocorticoids. Thyroid 25: 462-463.
  131. Tanda ML, Bartalena L 2012 Efficacy and safety of orbital radiotherapy for Graves’ orbitopathy. J Clin Endocrinol Metab 97: 3857-3865.
  132. Kahaly GJ, Rosler H-P, Pitz S, Hommel G 2000 Low- versus high-dose radiotherapy for Graves’ ophthalmopathy: a randomized, single blind trial. J Clin Endocrinol Metab 85: 102-108.
  133. Bartalena L, Marcocci C, Gorman CA, Wiersinga WM, Pinchera A 2003 Orbital radiotherapy for Graves’ ophthalmopathy: useful or useless? Safe or dangerous. J Endocrinol Invest 26: 5-16.
  134. Bradley EA, Gower EW, Bradley DJ, Meyer DR, Cahill KV, Custer PL, Holck DE, Woog JJ 2008 Orbital radiation for Graves ophthalmopathy. Ophthalmology 115: 398-409.
  135. Prummel MF, Mourits MP, Blank L, Berghout A, Koornneef L, Wiersinga WM 1993 Randomised, double-blind trial of prednisone versus radiotherapy in Graves’ ophthalmopathy. Lancet 342: 949-954.
  136. Mourits MP, van Kempen-Harteveld ML, Garcia MB, Koppeschaar HPF, Tick L, Terwee CB 2000 Radiotherapy for Graves’ orbitopathy: randomised, placebo-controlled study. Lancet 355: 1505-1509.
  137. Gorman CA, Garrity JA, Fatourechi V, Bahn RS, Petersen IA, Stafford SL, Earle JD, Forbes GS, Kline RW, Bergstrahl EJ, Offord KP, Rademacher DM, Stanley NM, Bartley GB 2001 A prospective, randomized, double-blind, placebo-controlled study of orbital radiotherapy for Graves’ ophthalmopathy. Ophthalmology 108: 1523-1534.
  138. Prummel MF, Terwee CB, Gerding MN, Baldeschi L, Mourits MP, Blank L, Dekker FW, Wiersinga WM 2004 A randomized controlled trial of orbital radiotherapy versus sham irradiation in patients with mild Graves’ ophthalmopathy. J Clin Endocrinol Metab 89: 15-20.
  139. Marcocci C, Bartalena L, Rocchi R, Marinò M, Menconi F, Morabito E, Mazzi B, Mazzeo S, Sartini MS, Nardi M, Cartei F, Cionini L, Pinchbera A 2003 Long-term safety of orbital radiotherapy for Graves’ ophthalmopathy. J Clin Endocrinol Metab 88: 3561-3566.
  140. Wakelkamp IMMJ, Tan H, Saeed P, Sckingemann RO, Verbraak FD, Blank LECM, Prummel MF, Wiersinga WM 2004 Orbital irradiation for Graves’ ophthalmopathy. Is it safe? A long-term follow-up study. Ophthalmology 111: 1557-1562.
  141. Bartalena L, Marcocci C, Chiovato L, Laddaga M, Lepri G, Andreani D, Cavallacci G, Baschieri L, Pinchera A 1983 Orbital cobalt irradiation combined with systemic corticosteroids for Graves’ ophthalmopathy: comparison with systemic corticosteroids alone. J Clin Endocrinol Metab 56: 1139-1144.
  142. Marcocci C, Bartalena L, Bogazzi F, Bruno-Bossio G, Lepri A, Pinchera A 1991 Orbital radiotherapy combined with high-dose systemic glucocorticoids for Graves’ ophthalmopathy is more effective than orbital radiotherapy alone: results of a prospective study. J Endocrinol Invest 14: 853-860.
  143. Kahaly G, Schrenzemeir, Krause U, Schweikert B, Meuer S, Muller W 1986 Ciclosporin and prednisone v. Prednisone in treatment of Graves’ ophthalmopathy: a controlled, randomized and prospective study. Eur J Clin Invest 16: 415-422.
  144. Prummel MF, Mourits MP, Berghout A, Krenning EP, van der Gaag R, Koornneef L, Wiersinga AM 1989 Prednisone and cyclosporine in the treatment of severe Graves’ ophthalmopathy. N Engl J Med 321: 1353-1359.
  145. Bartalena L 2010 What to do for moderate-to-severe Graves’ orbitopathy if glucocorticoids fail? Clin Endocrinol (Oxf) 73: 149-152.
  146. Salvi M, Vannucchi G, Beck-Peccoz P 2013 Potential utility of rituximab for Graves’ orbitopathy. J Clin Endocrinol Metab 98: 4291-4299.
  147. Minakaran N, Ezra DG 2013 Rituximab for thyroid-associated ophthalmopathy. Cochrane Database Syst Rev. 2013 May 31;5:CD009226. doi: 10.1002/14651858.
  148. Stan MN, Garrity JA, Carranza Leon BG, Prabin T, Bradley EA, Bahn RS 2015 Randomized controlled trial of rituximab in patients with Graves’ orbitopathy. J Clin Endocrinol Metab 100: 432-441.
  149. Salvi M, Vannucchi G, Currò N, Campi I, Covelli D, Dazzi D et al. 2015 Efficacy of B-cell targeted therapy with rituximab in patients with active moderate to severe Graves’ orbitopathy: a randomized controlled study. J Clin Endocrinol Metab 100: 422-431.
  150. Wiersinga WM 2017 Advances in treatment of active, moderate-to-severe Graves’ ophthalmopathy. Lancet Diabetes Endocrinol 5: 134-142.
  151. Ye X, Bo X, Hu X, Cui H, Lu B, Shao J, Wang J 2017 Efficacy and safety of mycophenolate mofetil in patients with active moderate-to-severe Graves’ orbitopathy. Clin Endocrinol (Oxf) 86: 247-255.
  152. Kahaly GJ, Riedl M, Konig J, Pits S, Ponto K, Diana T et al 2018 Mycophenolate plus methylprednisolone versus methylprednisolone alone in active, moderate-to-severe Graves’ orbitopathy (MINGO): a randomized, observer-masked, multicentre trial. Lancet Diabetes Endocrinol, in press.
  153. Riedl M, Kuhn A, Kramer I, Kolbe E, Kahaly GJ 2016 Prospective, systematically recorded mycophenolate safety data in Graves’ orbitopathy. J Endocrinol Invest 39: 687-694.
  154. Smith TJ, Huetwell FGL, Hegedus L, Douglas RS 2012 Role of insulin-like growth factor-1 (IGF-1) pathway in the pathogenesis of Graves’ orbitopathy. Best Pract Res Clin Endocrinol Metab 26: 291-302.
  155. Smith TJ, Kahaly GJ, Ezra DG, Fleming JC, Dailey RA, Tang RA et al 2017 Teprotumumab for thyroid-associated ophthalmopathy. N Engl J Med 376: 1748-1761.
  156. Piantanida E, Bartalena L 2017 Teprotumumab: a new avenue for the management of moderate-to-severe and active Graves’ orbitopathy? J Endocrinol Invest 40: 885-887.
  157. Baldeschi L 2017 Orbital decompression. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 211-229.
  158. Mourits MP, Bijl H. Altea MA, Baldeschi L, Boboridis K, Currò N, Dickinson AJ, Eckstein A, Freidel M, Guastella C, Kahaly GJ, Kalmann R, Krassas GE, Lane CM, Lareida J, Marcocci C, Marinò M, Nardi M, Mohr Ch, Neoh C, Pinchera A, Orgiazzi J, Pitz S, Saeed P, Salvi M, Sellari-Franceschini S, Stahl M, von Arx G, Wiersinga WM 2009 Outcome of orbital decompression for disfiguring proptosis in patients with Graves’ orbitopathy using various surgical procedures. Br J Ophthalmol 93: 1518-1523.
  159. Garrity JA, Fatourechi V, Bartley GB, Bergstrahl EJ, DeSanto LW, Gorman CA 1994 Graves’ ophthalmopathy. Results of transantral orbital decompression in 428 patients with severe Graves’ ophthalmopathy. Am J Ophthalmol 116: 533-547.
  160. Nardi M, Eckstein A 2017 Eye muscle surgery. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 230-246.
  161. Clarke L, Eckstein A 2017 Eyelid surgery. In Wiersinga WM, Kahaly GJ (eds), Graves’ orbitopathy. A multidisciplinary approach – Questions and answers, Karger, Basel, 3rd ed., 247-259.
  162. Marcocci C, Bartalena L, Pinchera A 1998 Ablative or non-ablative therapy for Graves’ hyperthyroidism in patients with ophthalmopathy? J Endocrinol Invest 21: 468-471.
  163. Weetman AP, Harrison BJ 1998 Ablative or non-ablative therapy for Graves’ hyperthyroidism in patients with ophthalmopathy? J Endocrinol Invest 21: 472-475.
  164. DeGroot LJ, Benjasuratwong Y 1996 Evaluation of thyroid ablative therapy for ophthalmopathy of Graves’ disease. Orbit 15: 187-196.
  165. Moleti M, Mattina F, Salomone I, Violi MA, Nucera C, Baldari S, Lo Schiavo MG, Regalbuto C, Trimarchi F, Vermiglio F 2003 Effects of thyroidectomy alone or followed by radioiodine ablation of thyroid remnants on the outcome of Graves’ ophthalmopathy. Thyroid 13: 653-658.
  166. Menconi F, Marinò M, Pinchera A, Rocchi R, Mazzi B, Nardi M, Bartalena L, Marcocci C 2007 Effects of total thyroid ablation vs near-total thyroidectomy alone on mild to moderate Graves’ orbitopathy treated with intravenous glucocorticoids. J Clin Endocrinol Metab 92: 1653-1658.
  167. Leo M, Marcocci C, Pinchera A, nardi M, Megna L, Rocchi R, Latrofa F, Altea MA, Mazzi B, Sisti E, Profilo MA, Marinò M 2012 Outcome of Graves’ orbitopathy after total thyroid ablation and glucocorticoid treatment: follow-up of a randomized clinical trial. J Clin Endocrinol Metab 97: E44-E48.
  168. Elbers L, Mourits M, Wiersinga W 2011 Outcome of very long-term treatment with antithyroid drugs in Graves’ hyperthyroidism associated with Graves’ orbitopathy. Thyroid 21: 279-283.
  169. Laurberg P, Berman BC, Andersen S, Bulow Pedersen I 2011 Sustained control of Graves’ hyperthyroidism during long-term low-dose antithyroid drug therapy of patients with severe Graves’ orbitopathy. Thyroid 21: 951-956.
  170. Bartalena L 2011 The dilemma of how to manage Graves’ hyperthyroidism in patients with associated orbitopathy. J Clin Endocrinol Metab 96: 592-599.
  171. Bartalena L, Macchia PE, Marcocci C, Salvi M, Vermiglio F 2015 Effects of treatment modalities for Graves’ hyperthyroidism on Graves’ orbitopathy: a 2015 Italian Society of Endocrinology consensus statement. J Endocrinol Invest 38: 481-487.
  172. Moleti M, Sturniolo G, Vermiglio F 2016 Safety of total thyroid ablation in patients with Graves’ orbitopathy. J Endocrinol Invest 39: 1199-1201.
  173. Fatourechi V 2012 Thyroid dermopathy and acropachy. Best Pract Res Clin Endocrinol Metab 26: 553-565.
  174. Fatourechi V, Bartley GB, Eghbali-Fatourechi GZ, Powell CC, Ahmed DD, Garrity JA 2003 Graves’ dermopathy and acropachy are markers of severe Graves’ ophthalmopathy. Thyroid 13: 1141-1144.
  175. Bartley GB, Fatourechi V, Kadrmas EF, Ilstrup DM, Garrity JA, Gorman CA 1996 Clinical features of Graves’ ophthalmopathy in an incidence cohort. Am J Ophthalmol 121: 284-290.
  176. Schwartz KM, Fatourechi V, Ahmed DDF, Pond GR 2002 Dermopathy of Graves’ disease (pretibial myxedema): long-term outcome. J Clin Endocrinol Metab 87:438-446.
  177. Tong DW, Ho KK 1998 Pretibial myxedema presenting as a scar infiltrate. Australas J Dermatol 39:255-257.
  178. Misner CS, Ramsay EW, Houck He, Kauffman CL 1998 Graves’disease presenting as a localized myxedema in a thigh donor graft site. J Am Ac Dermatol 39:864-869,1998.
  179. Cohen JB, Balzer B, Wapnir I, McDougall IR 2004 Elephantiasic pretibial myxedema. Thyroid 14: 237-238.
  180. Fatourechi V 2005 Pretibial myxedema: pathophysiology and treatment options. Am J Clin Dermatol 6: 295-309.
  181. Shishiba Y, Imai Y, Odajima R, Ozawa Y, Shimizu T 1992 Immunoglobulin G of patients with circumscribed pretibial myxedema of Graves' disease stimulates proteoglycan synthesis in human skin fibroblasts in culture. Acta Endocrinol(Copenh) 127:44-51.
  182. Hanke CW, Bergfeld WF, Guirguis MN, Lewis LJ 1983 Hyaluronic acid synthesis in fibroblasts of pretibial myxedema. Cleve Clin Q 50:129-133.
  183. Heufelder AE, Dutton CM, Sarkar G, Donovan KA, Bahn RS 1993 Detection of TSH receptor RNA in cultured fibroblasts from patients with Graves’ ophthalmopathy and pretibial dermopathy. Thyroid 3: 297-300.
  184. Rapoport B, Alsabeh R, Aftergood D, McLachlan SM 2000 Elephantiasic pretibial myxedema: insight into a hypothesis regarding the pathogenesis of the extrathyroidal manifestations of Graves’ disease. Thyroid 10: 685-692.
  185. Metcalfe RA, Davies R, Weetman AP 1993 Analysis of fibroblast-stimulating activity in IgG from patients with Graves’ dermopathy. Thyroid 3: 207-212.
  186. Kahaly GJ, Dillmann WH 2005 Thyroid hormone action in the heart. Endocr Rev 26: 704-728.
  187. Danzi S, Klein I 2012 Thyroid hormone and the cardiovascular system. Med Clin N Am 96: 257-268.
  188. Kahaly GJ, Wagner S, Nieswandt J, Mohr-Kahaly S, Rayan TJ 1999 Stress echocardiography in hyperthyroidism. J Clin Endocrinol Metab 84:2308-2313.
  189. Franklyn JA, Maisonneuve P, Sheppard MC, Betteridge J, Boyle P 1998 Mortality after the treatment of hyperthyroidism with radioactive iodine. N Engl J Med 338: 712-718.
  190. Osman F, Gammage MD, Franklyn JA 2002 Hyperthyroidism and cardiovascular morbidity and mortality. Thyroid 12: 483-488.
  191. Parle JV, Maisonneuve P, Sheppard MC, Boyle P, Franklyn JA 2001 Prediction of all cause and cardiovascular mortality in elderly people from one low serum thyrotropin result : a 10-year cohort study. Lancet 358: 861-865.
  192. Kahaly GJ, Nieswandt J, Mohr-Kahaly S 1998 Cardiac risk of hyperthyroidism in the elderly. Thyroid 8:1165-1169.
  193. Agner T, Almdal T, Thorsteinsson B, Agner E 1984 A reevaluation of atrial fibrillation in thyrotoxicosis. Dan Med Bull 31: 157-159.
  194. Nakazawa HK, Sakurai K, Hamada N, Momotani N, Ito K 1982 Management of atrial fibrillation in the post-thyrotoxic state. Am J Med 72:903-906.
  195. Sawin LT, Geller A, Wolf PhA, Belanger A, Baker E 1994 Low serum thyrotropin concentrations as a risk factor for atrial fibrillation in older persons. New Engl J Med 331:1249-1253.
  196. Biondi B 2012 Heart failure and thyroid dysfunction. Eur J Endocrinol 167: 609-618.
  197. Siu CW, Yeung CY, Lau CP, Kung AW, TSE HF 2007 Incidence, clinical characteristics and outcome of congestive heart failure as the initial presentation in patients with primary hyperthyroidism. Heart 93: 483-487.
  198. Yue W-S, Chong B-H, Zhang X-H, Liao S-Y, Jim A-Y, Jim M-H, Kung AWC, Tse H-F, Siu C-W 2011 Hyperthyroidism-induced left ventricular diastolic dysfunction: implication in hyperthyroidism-related heart failure. Clin Endocrinol (Oxf) 74: 636-643.
  199. Boccalandro C, Boccalandro F, Orlander P, Wei CF 2003 Severe reversible dilated cardiomyopathy and hyperthyroidism: case report and review of the literature. Endocr Pract 9:140-6.
  200. Soh MC, Croxson M 2008 Fatala thyrotoxic cardiomyopathy in a young man. Br Med J 337: a531.
  201. Featherstone HJ, Stewart DK 1983 Angina in thyrotoxicosis: thyroid related coronary artery spasm. Arch Intern Med 143: 554-558.
  202. Moliterno D, Debold CR, Robertson RM 1992 Coronary vasospasm – relation to the hyperthyroid state. Am J Med Sci 304: 38-42
  203. Resenkov L, Falicov R 1977 Thyrotoxicosis and lactate producing angina pectoris with normal coronary arteries. Br Heart J 39:1051-1055
  204. Biondi B, Fazio S, Carella C, Amato G, Littadini A, Lupoli G, Saca L, Bellastella A, Lombardi G. 1993 Cardiac effects of long-term thyrotropin-suppressive therapy with levothyroxine. J Clin Endocrinol Metab 77:334-338.
  205. Shapiro LE, Sievart R, Ong L, Ocamo E, Chance R, Lee M, Nanna M, Ferrick K, Surks M 1997 Minimal cardiac effects in asymptomatic athyreotic patients chronically treated with thyrotropin-suppressive doses of L-thyroxine. J Clin Endocrinol Metab 82:2592-2595.
  206. Nakazawa H, Lythall DA, Noh J, Ishikawa N, Sugino K, Ito K, Hardman SM 2000 Is there a place for late cardioversion of atrial fibrillation? A long-term follow-up study of patients with post-thyrotoxic atrial fibrillation. Eur Heart J 12:327-333.

 

The Iodine Deficiency Disorders

ABSTRACT

This chapter provides an overview of the disorders caused by iodine deficiency. Extensively referenced, it includes data on dietary sources of iodine, goitrogens, the effects of iodine deficiency throughout the lifecycle, the pathophysiology of iodine deficiency, as well as strategies for control and monitoring of the iodine deficiency disorders, such as iodized salt and iodized oil. It emphasizes the role of iodine deficiency in the development of brain damage and neurocognitive impairment, assessment of the iodine status of a population, the potential side effects of excessive iodine intake and current worldwide epidemiological data.

INTRODUCTION

This chapter provides a global overview of the disorders caused by iodine deficiency. Special emphasis will be put on recent developments such as the role of iodine deficiency in the development of brain damage and neurocognitive impairment, assessment of the iodine status of a population, strategies for control and monitoring of the iodine deficiency disorders (IDD), as well as side effects of iodine. Up to date information on IDD can be obtained by visiting the website of the Iodine Global Network (IGN) http://www.ign.org.

ETIOLOGY

Iodine (atomic weight 126.9 g/atom) is an essential component of the hormones produced by the thyroid gland. Thyroid hormones, and therefore iodine, are essential for mammalian life. Iodine (as iodide) is widely but unevenly distributed in the earth’s environment. Most iodide is found in the oceans (≈50 μg/L), and iodide ions in seawater are oxidized to elemental iodine, which volatilizes into the atmosphere and is returned to the soil by rain, completing the cycle. However, iodine cycling in many regions is slow and incomplete, and soils and ground water become deficient in iodine. Crops grown in these soils will be low in iodine, and humans and animals consuming food grown in these soils become iodine deficient (1). In plant foods grown in deficient soils, iodine concentration may be as low as 10 μg/kg dry weight, compared to ≈1 mg/kg in plants from iodine-sufficient soils. Iodine deficient soils are most common in inland regions, mountainous areas and areas of frequent flooding, but can also occur in coastal regions (2). This arises from the distant past through glaciation, compounded by the leaching effects of snow, water and heavy rainfall, which removes iodine from the soil. The mountainous regions of Europe, the Northern Indian Subcontinent, the extensive mountain ranges of China, the Andean region in South America and the lesser ranges of Africa are all iodine deficient. Also, the Ganges Valley in India, the Irawaddy Valley in Burma, and the Songkala valley in Northern China are also areas of endemic iodine deficiency. Iodine deficiency in populations residing in these areas will persist until iodine enters the food chain through addition of iodine to foods (e.g. iodization of salt) or dietary diversification introduces foods produced in iodine-sufficient areas.

DIETARY SOURCES OF IODINE

 

The native iodine content of most foods and beverages is low, and most commonly consumed foods provide 3 to 80 µg per serving (3-7). Major dietary sources of iodine in the USA, Europe and Australia are bread, milk and to a lesser extent seafood (3,4). Based on direct food analysis, mean intake of dietary iodine is ≈140 µg/day in Switzerland and 100-180 µg/day in Libya (3,6). Boiling, baking, and canning of foods containing iodised salt cause only small losses (of foods contain(8). Iodine content in foods is also influenced by iodine-containing compounds used in irrigation, fertilizers, and livestock feed. Iodophors, used for cleaning milk cans and teats in the dairy industry, can increase the native iodine content of dairy products through contamination of iodine containing residues (9); there are few data on the bioavailability of iodine or potential health risks from these iodophors. Traditionally, iodate was used in bread making as a dough conditioner, but it is being replaced by non-iodine-containing conditioners.  Recommendations for daily iodine intake by age group are shown in Table 1.

 

 

Table 1: Recommendations for iodine intake (µg/day) by age or population group

 

Age or population groupa U.S. Institute of Medicine (ref.5) Age or population groupc World Health Organization (ref.1)
Infants 0–12 months b 110-130 Children 0-5 years 90
Children 1-8 years 90 Children 6-12 years 120
Children 9-13 years 120    
Adults n 9-13 yea 150 Adults >12 years 150
Pregnancy 220 Pregnancy 250
Lactation 290 Lactation 250

a Recommended Daily Allowance. b Adequate Intake. c Recommended Nutrient Intake.

 

IODINE DEFICIENCY IN ANIMAL MODELS

Studies in rats have been carried out using the diet consumed by the people of Jixian village in China (10-13). This village was severely iodine deficient with 11% prevalence of endemic cretinism. The diet included available main crops (maize, wheat), vegetables, and water from the area with an iodine content of 4.5 µg/kg. After the dam had received the diet for 4 months, there was obvious neonatal goiter, fetal serum T4 was 3.6 µg/L compared to controls of 10.4 µg/L and they had higher 125I uptake and reduced brain weights. The density of brain cells was increased in the cerebral hemispheres. The cerebellum showed delayed disappearance of the external granular layer with reduced incorporation of 3H leucine in comparison to the control group.

Other more detailed studies have been carried out on the number and distribution of dendritic spines along the apical shaft of the pyramidal cells of the cerebral cortex of the rat (14). These dendritic spines can be accurately measured and have been studied in relation to both iodine deficiency and hypothyroidism. Their appearance and development reflects the formation of synaptic contacts with afferents from other neurons. In normal rats there is a progressive increase in the number of spines from 10 to 80 days of age.

These studies have demonstrated a significant effect of an iodine deficient diet on the number and distribution of the spines on the pyramidal cells of the visual cortex. This effect is similar to that of thyroidectomy. More detailed studies following thyroidectomy indicated the importance of the timing of the procedure. If carried out before the 10th day of life, recovery is unlikely to occur unless there is immediate replacement with L-T4. At 40 or 70 days, replacement can restore a normal distribution of spines even if there is a 30 day delay in its initiation. These differences confirm the need for early treatment of congenital hypothyroidism and prevention of iodine deficiency in the newborn infant in order to prevent brain damage and mental retardation.

Severe iodine deficiency has been produced in the marmoset (Callithrix Jacchus Jacchus) with a mixed diet of maize (60%), peas (15%), torula yeast (10%) and dried iodine deficient mutton (10%). The newborn iodine-deficient marmosets showed some sparsity of hair growth (15). The thyroid gland was enlarged with gross reduction in plasma T4 in both mothers and newborns, and was greater in the second pregnancy than in the first, suggesting a greater severity of iodine deficiency. There was a significant reduction in brain weight in the newborns from the second pregnancy, but not from the first. The findings were more striking in the cerebellum with reduction in weight and cell number evident and histological changes indicating impaired cell acquisition. These findings demonstrate the significant effects of iodine deficiency on the primate brain.

Severe iodine deficiency has been produced in sheep (16) with a low-iodine diet of crushed maize and pelleted pea pollard (8-15 ug iodine/kg) which provided 5-8 ug iodine per day for sheep weighing 40-50 kg. The iodine deficient fetuses at 140 days were grossly different in physical appearance in comparison to the control fetuses. There was reduced weight, absence of wool growth, goiter, varying degrees of subluxation of the foot joints, and deformation of the skull. (Fig. 2) There was also delayed bone maturation as indicated by delayed appearance of epiphyses in the limbs (17). Goiter was evident from 70 days in the iodine-deficient fetuses and thyroid histology revealed hyperplasia from 56 days gestation, associated with a reduction in fetal thyroid iodine content and reduced plasma T4 values. There was a lowered brain weight and DNA content as early as 70 days, indicating a reduction in cell number probably due to delayed neuroblast multiplication which normally occurs from 40-80 days in the sheep. Findings in the cerebellum were similar to those already described in marmoset (16).

A single intramuscular injection of iodized oil (1 ml = 480 mg iodine) given to the iodine deficient mother at 100 days gestation was followed by partial restoration of the lambs’ brain weight and body weight with restoration of maternal and fetal plasma T4 values to normal (16). Studies of the mechanisms involved revealed significant effects of fetal thyroidectomy in late gestation and a significant effect of maternal thyroidectomy on brain development mid gestation. The combination of maternal thyroidectomy (carried out 6 weeks before pregnancy) and fetal thyroidectomy produced more severe effects than that of iodine deficiency, and was associated with greater reduction in both maternal and fetal thyroid hormone levels (17). These findings in animal models confirm the importance of both maternal and fetal thyroid hormones in fetal brain development.

IODINE DEFICIENCY IN THE HUMAN LIFE CYCLE

The term IDD refers to all the ill-effects of iodine deficiency in a population that can be prevented by ensuring that the population has an adequate intake of iodine (1). These effects are listed in Table 2. Brain damage and irreversible mental retardation are the most important disorders induced by iodine deficiency: in 1990 it was estimated that among the 1572 million people in the world exposed to iodine deficiency (28.9 % of the then world population), 11.2 million were affected by overt cretinism, the most extreme form of mental retardation due to iodine deficiency and that another 43 million people were affected by some degree of itellectual impairment (18). Thus, iodine deficiency was a leading global cause of preventable mental impairment.

 

Table 2. The spectrum of iodine deficiency disorders, IDD (ref.1).

Fetus

Miscarriage

Stillbirths

Congenital anomalies

Increased perinatal morbidity and mortality

Endemic cretinism

Neonate

Neonatal goiter

Neonatal hypothyroidism

Endemic neurocognitive impairment

Increased susceptibility of the thyroid gland to nuclear radiation

Child and adolescent

Goiter

(Subclinical) hypothyroidism

Impaired mental function

Retarded physical development

Increased susceptibility of the thyroid gland to nuclear radiation

Adult

Goiter with its complications

Hypothyroidism

Impaired mental function

Spontaneous hyperthyroidism in the elderly

Iodine-induced hyperthyroidism

Increased susceptibility of the thyroid gland to nuclear radiation

Iodine Deficiency In Pregnancy

Iodine deficiency in the fetus is the result of iodine deficiency in the mother. The consequence of iodine deficiency during pregnancy is impaired synthesis of thyroid hormones by the mother and the fetus. An insufficient supply of thyroid hormones to the developing brain may result in neurocognitive impairment (19-25). The physiologic role of thyroid hormones is to ensure that normal growth and development occurs through specific effects on the rate of cell differentiation and gene expression. Thyroid hormone action is exerted through the binding of T3 to nuclear receptors which regulate the expression of specific genes in different brain regions during fetal and early postnatal life. The T3 which is bound to the nuclear receptors is primarily dependent on its local intracellular production from T4 via type II deiodinase and not from circulating T3.

Brain Development In Humans

Figure 1 shows the time course of the development of the brain and of thyroid function in the human fetus and neonate. Brain growth is characterized by two periods of maximal growth velocity (26). The first one occurs during the first and second trimesters between the third and the fifth months of gestation. This phase corresponds to neuronal multiplication, migration and organization. The second phase takes place from the third trimester onwards up to the second and third years postnatally. It corresponds to glial cell multiplication, migration and myelinization. The first phase occurs before fetal thyroid has reached its functional capacity. During this phase, the supply of thyroid hormones to the growing fetus is almost exclusively of maternal origin while during the second phase, the supply of thyroid hormones to the fetus is essentially, but not solely, of fetal origin (27).

 

Ontogenesis of thyroid functions and regulation in humans.

Figure 1. Ontogenesis of thyroid function and regulation in humans during fetal and early postnatal life in relation to the velocity of brain growth. From Delange and Fisher (ref.28).

 

In humans, T4 can be found in the first trimester coelomic fluid from 6 weeks of gestational age, long before the onset of secretion of T4 by the fetal thyroid, which occurs at the 24th week of gestation (29). Nuclear T3 receptors and the amount of T3 bound to these receptors increases six to tenfold between 10 and 16 weeks (30). The T4 and T3 found in early human fetuses up to mid gestation are likely to be entirely or mostly of maternal origin. As a consequence, infants born to women with hypothyroxinemia at 12 weeks gestation (fT4 concentrations; <10th percentile) had lower developmental scores (31). In addition, substantial amounts of T4 are transferred from mother to fetus during late gestation (32). The deiodinases are involved in the action of thyroid hormones in the brain, especially deiodinase D3 that is found in the uterine implantation site and in the placenta, producing rT3 from T4 and 3’,5’-T2 from T3 and thus having a protective effect to avoid an excess of thyroid hormone reaching the fetus.

Severe Iodine Deficiency in Pregnancy: Cretinism and Increased Fetal and Perinatal Mortality

 

The most serious adverse effect of iodine deficiency is damage to the fetus. Iodine treatment of pregnant women in areas of severe deficiency reduces fetal and perinatal mortality and improves motor and cognitive performance of the offspring. Severe iodine deficiency in utero causes a condition characterized by gross mental retardation along with varying degrees of short stature, deaf mutism, and spasticity that is termed cretinism. These disorders are described in detail below.

 

Mild-To-Moderate Deficiency in Pregnancy

 

The potential adverse effects of mild-to-moderate iodine deficiency during pregnancy in humans are unclear (33). Observational studies have shown associations between both mild maternal iodine deficiency and mild maternal thyroid hypofunction and decreased child cognition (33). In Europe, several randomized controlled trials of iodine supplementation in mild-to-moderately iodine deficient pregnant women have been done, and iodine reduced maternal and newborn thyroid size, and, in some, decreased maternal TSH; however, none of the trials showed an effect on maternal and newborn total or free thyroid hormone concentrations (33). No data are yet available from randomized placebo-controlled trials in regions of mild to moderate iodine insufficiency on the relation between maternal iodine supplementation and neurobehavioral development in the offspring (33).

Iodine Deficiency in the Neonate

An increased perinatal mortality due to iodine deficiency has been shown in Zaire from the results of a controlled trial of iodized oil injections alternating with a control injection given in the latter half of pregnancy (34). There was a substantial fall in infant mortality with improved birth weight following the iodized oil injection. Low birth weight of any cause is generally associated with a higher rate of congenital anomalies and higher risk through childhood. This has been demonstrated in the longer term follow up of the controlled trial in Papua New Guinea in children up to the age of 12 years (35) and in Indonesia (36).

A reduction of infant mortality has also been reported from China following iodine supplementation of irrigation water in areas of severe iodine deficiency. Iodine replacement has probably been an important factor in the national decrease in infant mortality in China (37).

Apart from mortality, the importance of thyroid function in the neonate relates to the fact that the brain of the human infant at birth has only reached about one third of its full size and continues to grow rapidly until the end of the second year (38). Thyroid hormone, dependent on an adequate supply of iodine, is essential for normal brain development as has been confirmed by the animal studies already cited.

Studies on iodine nutrition and neonatal thyroid function in Europe in the early 1980s confirmed the continuing presence of iodine deficiency affecting neonatal thyroid function and hence a threat to early brain development (39). A series of 1076 urine samples were collected from 16 centers from 10 different countries in Europe along with an additional series from Toronto, Canada and analyzed for their iodine content. The results of these determinations are shown in Table 3. The distribution was skewed so that arithmetic means were not used, but the results were expressed in percentiles. Some very high values were seen which could be attributed to the use of iodinated contrast media for radiological investigation of the mother during pregnancy. There was a marked difference in the results from the various cities. The high levels in Rotterdam, Helsinki and Stockholm differed from the low levels in Gottingen, Heidelberg, Freiburg and Jena by a factor of more than 10. Intermediate levels were seen in Catania, Zurich and Lille.

Table 3. Frequency distributions of urinary iodine concentrations in healthy term infants in 14 cities in Europe and in Toronto, Canada

  Urinary Iodine Concentration (ug/L)
City Number of infants 10th Percentile 50th Percentile 90th Percentile

Frequency (%) of values

Below 50μg/L

Toronto 81 4.3 14.8 37.5 11.9
Rotterdam 64 4.5 16.2 33.2 15.3
Helsinki 39 4.8 11.2 31.8 12.8
Stockholm 52 5.1 11.0 25.3 5.9
Catania 14 2.2 7.1 11.0 38.4
Zurich 62 2.6 6.2 12.9 34.4
Lille 82 2.0 5.8 15.2 37.2
Brussels 196 1.7 4.8 16.7 53.2
Rome 114 1.5 4.7 13.8 53.5
Toulouse 37 1.2 2.9 9.4 69.4
Berlin 87 1.3 2.8 13.6 69.7
Gottingen 81 0.9 1.5 4.7 91.3
Heidelberg 39 1.1 1.3 4.0 89.8
Freiburg 41 1.1 1.1 2.3 100.0
Jena 54 0.4 0.8 2.2 100.0

Data on neonatal thyroid function was analyzed for four cities where enough newborns (30,000 - 102,000) had been tested. The incidence of permanent congenital hypothyroidism was very similar in the four cities but the rate of transient hypothyroidism was much greater in Freiburg, associated with the lowest level of urine iodine excretion, than in Stockholm, with intermediate findings from Rome and Brussels.

In developing countries with more severe iodine deficiency, observations have now been made using blood taken from the umbilical vein just after birth. Neonatal chemical hypothyroidism was defined by serum levels of T4 lesser than 3 ug/dl and TSH greater than 100 mIU/L). In the most severely iodine deficient environments in Northern India, where more than 50% of the population has urinary iodine levels below 25 ug per gram creatinine, the incidence of neonatal hypothyroidism was 75 to 115 per thousand births (40). By contrast in Delhi, where only mild iodine deficiency is present with low prevalence of goiter and no cretinism, the incidence drops to 6 per thousand. In control areas without goiter the level was only one per thousand.

There is similar evidence from neonatal observations in neonates in Zaire in Africa where a rate of 10% of biochemical hypothyroidism has been found (41). This hypothyroidism persists into infancy and childhood if the deficiency is not corrected, and results in retardation of physical and mental development (42). These observations indicate a much greater risk of mental impairment in severely iodine deficient populations than is indicated by the presence of cretinism.

Another important aspect of iodine deficiency in the neonate and child is an increased susceptibility of the thyroid gland to radioactive fall-out. Thyroidal uptake of radioiodine reached its maximum value in the earliest years of life and then declined progressively into adult life (43). The apparent thyroidal iodine turnover rate was much higher in young infants than in adults and decreased progressively with age. In order to provide the normal rate of T4 secretion, Delange (43) estimated the turnover rate for intrathyroidal iodine must be 25-30 times higher in young infants than in adolescents and adults. In iodine deficiency a further increase in turnover rate is required to maintain normal thyroid hormone levels. This is the reason for the greatly increased susceptibility of the neonate and fetus to iodine deficiency. Iodine deficiency also causes an increased uptake of the radioiodide resulting from nuclear radiation. Protection against this increased uptake can be provided by correction of iodine deficiency.

Iodine Deficiency in the Child

There is cross-sectional evidence that impairment of thyroid function evidenced in mothers and neonates in conditions of mild-to-moderate iodine deficiency affects the intellectual development of their offspring. Aghini-Lombardi et al. (44) reported that in children aged 6-10 years in an area in Tuscany who had mild iodine deficiency (64 μg iodine/day), the reaction time was delayed compared with matched controls from an iodine sufficient area (142 μg iodine/day). The cognitive abilities of the children were not affected. Similarly, it was reported that in an area of Southern Spain with mild iodine deficiency (median urinary iodine of 90 μg/L), the intelligence quotient (IQ) was significantly higher in children with urinary iodine levels above 100 μg/L (45).

A recent randomized controlled study in Albania in a moderately iodine deficient area showed that information processing, fine motor skill and visual problem solving significantly improved in school-children after iodine repletion of the population (46). In a randomized, placebo-controlled, trial in mildly-deficient New Zealand children aged 10-13 y, children were randomly assigned to receive a daily tablet containing either 150 μg iodine or placebo for 28 wk (47). At the end of the trial, the overall cognitive score of the iodine-supplemented group was 0.19 SDs higher than that of the placebo group (P<0.02). These controlled trials suggest that mild to moderate iodine deficiency could prevent children from attaining their full intellectual potential (46,47). As these anomalies were reversible, they probably result from lately acquired and reversible subclinical hypothyroidism, rather than from fetal and/or neonatal hypothyroidism.

In severe iodine deficiency, the frequency distribution of IQ in normal appearing children is shifted towards low values as compared to children who were not exposed to in utero iodine deficiency because of correction of the deficiency in the mothers before or during early gestation (48-49). In a meta-analysis of 19 studies on neuromotor and cognitive functions in conditions of moderate to severe iodine deficiency, Bleichrodt and Born (50) concluded that iodine deficiency resulted in a loss of 13.5 IQ points at the level of the global population.A more recent metanalysis conducted on studies in China produced a very similar result (51). Several of these studies are summarized in Table 4.

Table 4. Neurointellectual Deficits in Infants and Schoolchildren in Conditions of Mild to Moderate Iodine Deficiency

REGIONS TESTS FINDINGS AUTHORS
Spain

Locally adpated

BAYLEY

McCARTHY

CATTELL

Lower psychomotor and mental development than controls Bleichrodt et al. 1989 (52)
Italy
Sicily BENDER- GESTALT

Low preceptual integrative motor ability

Neuromuscular and neurosensorial

abnormalities

Vermiglio et al. 1990 (53)
Tuscany WECHSLER RAVEN Low verbal IQ, perception, motor and attentive functions Fenzi et al. 1990 (54)
Tuscany

WISC

Reaction time

Lower velocity of motor response to visual stimuli

Vitti et al. 1992 (55)

Aghini-Lombardi et al. 1995 (44)

India

Verbal, pictorial learning tests

Tests of motivation

Lower capacities

learning

Tiwari et al. 1996 (56)
Iran Bender-Gestalt Raven Retardation in psychomotor development Azizi et al. 1993 (57)
Malawi Psychometric tests including verbal fluency Loss of 10 IQ points as compared to iodine-supplemented controls Shrestha 1994 (58)
Benin Battery of 8 non verbal tests exploring fluid intelligence and 2 psychomotor tests Loss of 5 IQ points as compared to controls supplemented with iodine for one year van den Briel et al. 2000 (59)

 

Data from cross-sectional studies on iodine intake and child growth are mixed, with most studies finding modest positive correlations (60). In five Asian countries, household access to iodized salt was correlated with increased weight-for-age and mid-upper-arm circumference in infancy (61). However, controlled intervention studies of iodized oil alone and iodine given with other micronutrients have generally not found effects on child growth (60). In iodine-deficient children, impaired thyroid function and goiter are inversely correlated with IGF-1 and IGFBP-3 concentrations (62). Recent controlled trials found iodine repletion increased insulin-like growth factor (IGF)-1 and insulin-like growth factor binding protein (IGFBP)-3 and improved somatic growth in children (60).

Iodine deficiency in the adult

Iodine status is a key determinant of thyroid disorders in adults (63). Severe iodine deficiency causes goitre and hypothyroidism because, despite an increase in thyroid activity to maximise iodine uptake and recycling in this setting, iodine concentrations are still too low to enable sufficient production of thyroid hormone. In mild-to-moderate iodine deficiency, increased thyroid activity can compensate for low iodine intake and maintain euthyroidism in most individuals, but at a price: chronic thyroid stimulation results in an increase in the prevalence of toxic nodular goitre and hyperthyroidism in populations. Thus, a consequence of longstanding iodine deficiency in the adult (64-67) and child (68) is the development of hyperthyroidism, especially in multinodular goiters with autonomous nodules. The pathogenesis of this syndrome is discussed later in this chapter, in the section on side effects of iodine supplementation.

This high prevalence of nodular autonomy usually results in a further increase in the prevalence of hyperthyroidism if iodine intake is subsequently increased by salt iodisation. However, this increase is transient because iodine sufficiency normalises thyroid activity which, in the long term, reduces nodular autonomy. Increased iodine intake in an iodine-deficient population is associated with a small increase in the prevalence of subclinical hypothyroidism and thyroid autoimmunity; whether these increases are also transient is unclear. Thus, optimisation of population iodine intake is an important component of preventive health care to reduce the prevalence of thyroid disorders (63).

SPECIFIC IODINE DEFICIENCY DISORDERS

Endemic goiter

Epidemiology

Endemic goiter is characterized by enlargement of the thyroid gland in a significantly large fraction of a population group, and is generally considered to be due to insufficient iodine in the daily diet. Endemic goiter exists in a population when >5% of 6-12 year-old children have enlarged thyroid glands

 

 shows a young girl with a soft diffuse goitre

Figure 2 shows a young girl with a soft diffuse goitre and an elderly woman with a huge, longstanding multinodular goiter, both resulting from iodine deficiency.

 

an elderly woman with a huge, longstanding multinodular goiter

Most mountainous districts in the world have been or still are endemic goiter regions. The disease may be seen throughout the Andes, in the whole sweep of the Himalayas, in the European Alps where iodide prophylaxis has not yet reached the entire population, in Greece and the Middle Eastern countries, in many foci in the People's Republic of China, and in the highlands of New Guinea. There are or were also important endemias in non-mountainous regions, as for example, the belt extending from the Cameroon grasslands across northern Zaire and the Central African Republic to the borders of Uganda and Rwanda, as well as in Holland, Central Europe and the interior of Brazil. An endemic existed in the Great Lakes region in North America until it was corrected by iodized salt in the early 1900s.

Goiter maps of various countries have been repeatedly drawn, requiring modification as successful prophylactic measures have been introduced. Although goiter was an important problem in many regions of the United States in the past (69), more recent US surveys have shown it in no more than 4-11% of schoolchildren, with evidence of continued adequate iodine nutrition in the country since 1988 (70,71). This finding is a testimony to the effectiveness of iodine prophylaxis in preventing endemic goiter. The world or regional distribution of goiter was exhaustively reviewed by Kelly and Snedden in 1960 (72) and, most recently, in 2005 (73).

These surveys reveal striking differences in the rate of goiter in different endemic regions and even in adjacent districts. The geographic unevenness of an endemia undoubtedly has much to do with the habits of the population and their ability and/or desire to import iodine containing foods. In attempting to account for the variability in the expression of endemic goiter from one locality to the next, the availability of iodine should be investigated before searching for some other subtle dietary or genetic factors. The key to the problem almost always lies in the availability of iodine. One must also consider the possibility that an observed goiter rate may not reflect current conditions, but rather may be a legacy of pre-existing iodine deficiency that has not yet been entirely resolved by an improvement in the supply of iodine. The assessment of goiter in a population, and its limitations, are discussed in the section on assessment of the IDD status of the population.

Etiology

Iodine Deficiency

The arguments supporting iodine deficiency as the cause of endemic goiter are four: (1) the close association between a low iodine content in food and water and the appearance of the disease in the population; (2) the sharp reduction in incidence when iodine is added to the diet; (3) the demonstration that the metabolism of iodine by patients with endemic goiter fits the pattern that would be expected from iodine deficiency and is reversed by iodine repletion; and 4) iodine deficiency causes changes in the thyroid glands of animals that are similar to those seen in humans (74,75). Almost invariably, careful assessment of the iodine intake of a goitrous population reveals levels considerably below normal.

Goitrogenic factors

Although the relation of iodine deficiency to endemic goiter is well established, other factors may be involved. A whole variety of naturally occurring agents have been identified that might be goitrogenic in man (76,77). It should be recognized that goitrogens are usually active only if iodine supply is limited and/or goitrogen intake is of long duration. Many of these have only been tested in animals and/or have been shown to possess antithyroid effects in vitro. These compounds belong to the following chemical groups:

  • Sulfurated organics (like thiocyanate, isothiocyanate, goitrin and disulphides)
  • Flavonoids (polyphenols)
  • Polyhydroxyphenols and phenol derivatives
  • Pyridines, phthalate esters and metabolites,
  • Polychlorinated (PCB) and polybrominated (PBB) biphenyls
  • Organochlorines (like DDT)
  • Polycyclic aromatic hydrocarbons (PAH)
  • Inorganic iodine (in excess)
  •  

Gaitan (76) divides goitrogens into agents acting directly on the thyroid gland and those causing goiter by indirect action. The former group is subdivided into those inhibiting transport of iodide into the thyroid (like thiocyanate and isothiocyanate), those acting on the intrathyroidal oxidation and organic binding process of iodide and/or the coupling reaction (like phenolic compounds) some phthalate derivatives (disulfides and goitrin) and those interfering with proteolysis, dehalogenation and hormone release (like iodide and lithium).

Indirect goitrogens increase the rate of thyroid hormone metabolism (like 2,4-dinitrophenol, PCB's and PBB's). Soybean, an important protein source in many third world countries, interrupts the enterohepatic cycle of thyroid hormone (78) and may cause goiter when iodine intake is limited.

Some of these goitrogens are synthetic and are used medicinally. Others occur in certain widely used food plants (79). The initial recognition of dietary goitrogenesis is attributed to Chesney et al. (80) who in 1928 found that rabbits fed largely on cabbage developed goiters. In 1936, Barker (81) found that thiocyanate used in large doses to treat hypertension resulted in goiter. In 1936, Hercus and Purves (82) reported their studies on the production of goiter in rats by feeding the seeds of several species of Brassica (rape, choumoellier, turnip, etc.). Both Mackenzie and MacKenzie (83) and Astwood (84) found in the 1940’s that certain drugs such as thiourea and related compounds caused hyperplasia of the thyroid when administered to rats. Their investigations quickly led to the introduction of the thionamide series of antithyroid drugs, now so familiar in clinical therapeutics.

Thiocyanate and precursors of thiocyanate, such as the cyanogenic glycosides, form another group of widely distributed natural antithyroid substances. They have been found particularly in the widely used tuber cassava (manioc) (85). Cassava causes goiter when fed to rats (86). Certain sulfur-containing onion volatiles are also goitrogenic (87). All of these substances interfere with the accumulation of thyroidal iodide, an effect that usually can be overcome by an increasing iodine intake.

Delange et al. (88) observed a striking difference in incidence of goiter in two regions of an isolated island in the Kivu Lake in Eastern Dem. Rep. of Congo, although the iodine intake of both groups was approximately the same. There was a major difference in the use of cassava. Cassava has been implicated as a contributing factor in endemic goiter in Zaire (89,90). In a study of several communities in the Ubangi region of Zaire, a relationship between goiter, thiocyanate and iodide excretion was described. The thiocyanate was derived from intestinal breakdown of the cyanogenic glycoside, linamarin, from cassava and its conversion to thiocyanate by the liver. The results indicated a reciprocal relationship between iodide and thiocyanate in that increasing amounts of iodide protected increasingly against the thiocyanate derived from the cassava (89). Thiocyanate may cross the human placenta (89, 91) and affect the thyroid of the fetus.

Excessive intake of iodine may cause goiter. A localized endemia has been reported on the coast of Hokkaido in Northern Japan (92). In this district the diet contained a huge amount of seaweed, and excretion of 127I in the urine exceeded 20 mg/day. The uptake of RAI by the thyroid was low, and some of it could be discharged by administration of thiocyanate, indicating impairment of organification. Similar findings have been reported from coastal (93) and continental (94) China.

Firm evidence for goitrogenic action in humans has only been shown for a few compounds: thiocyanate, goitrin, resorcinol, dinitrophenol, PBB's and its oxides, excess iodine and high doses of lithium (77). A definite role in endemic goiter has only been proved for thiocyanate and sulfurated organics, although substantial and circumstantial evidence favors the view that natural goitrogens, acting in concert with iodine deficiency, may determine the pattern and severity of the condition. An example is the possible role of the consumption of pearl millet in the etiology of endemic goiter in Sudan (95).

Selenium deficiency may have profound effects on thyroid hormone metabolism and possibly also on the thyroid gland itself (96-98). In this situation the function of type I deiodinase (a selenoprotein) is impaired. Type I deiodinase plays a major role in T4 deiodination in peripheral tissues. It has been shown that when, in an area of combined iodine and selenium deficiency, only selenium is supplemented, serum T4 decreases (99). This effect is explained by restoration of type I deiodinase activity leading to normalization of T4 deiodination while T4 synthesis remains impaired because of continued iodine deficiency.

Selenium deficiency also leads to a reduction of the selenium containing enzyme glutathione peroxidase. Glutathione peroxidase detoxifies H2O2 which is abundantly present in the thyroid gland as a substrate for the thyroperoxidase that catalyzes iodide oxidation and binding to thyroglobulin, and the oxidative coupling of iodotyrosines into iodothyronines. Reduced detoxification of H2O2 may lead to thyroid cell death (96,100). Elevated H2O2 levels in thyrocytes may be more toxic under situations of increased TSH stimulation such as is present in areas with severe iodine deficiency. Extensive epidemiological data collected in China indicated that all selenium-deficient areas were IDD-endemic areas. However, the reverse is not true: IDD can be very severe in many selenium-rich areas (101).

Deficiencies of iron (102,103) and vitamin A (104) may also have a goitrogenic effect in areas of iodine deficiency (Table 5).

 

Table 5. Dietary Goitrogens

 

Goitrogen Mechanism
Foods  
Cassava, lima beans, linseed, sorghum, sweet potato Contain cyanogenic glucosides; they are metabolized to thiocyanates that compete with iodine for thyroidal uptake
Cruciferous vegetables: cabbage, kale, cauliflower, broccoli, turnips, rapeseed Contains glucosinolates; metabolites compete with iodine for thyroidal uptake
Soy, millet Flavonoids impair thyroid peroxidase activity
Nutrients  
Selenium deficiency Accumulated peroxides may damage the thyroid, and deiodinase deficiency impairs thyroid hormone synthesis
Iron deficiency Reduces heme-dependent thyroperoxidase activity in the thyroid and may blunt the efficacy of iodine prophylaxis
Vitamin A deficiency Increases TSH stimulation and goiter through decreased vitamin A-mediated suppression of the pituitary TSHβ gene

Pathology

There are no gross or microscopic features that distinguish the thyroid of endemic goiter from changes that may appear in simple and sporadic goiter. The changes evolve through stages. In the very young, or in older patients who have lived under constant iodide deprivation, the finding is extreme hyperplasia. In some instances only a cellular organ is found, with little or no colloid. (Figure 3) The evolution of pathologic findings in humans have been detailed and well illustrated by Correa (105) and Studer and Ramelli (106) and follow the pattern of events first described by Marine (107) and known as the Marine cycle. In this formulation, repeated episodes of hyperplasia induced by iodine deficiency are followed by involution and atrophy, the result being a gland containing a mixed bag of nodules, zones of hyperplasia, and involuting, degenerative, and repair elements.

showing intense hyperplasia with no colloid

Figure 3. Histological section of large goiter removed because of pressure symptoms in Papua New Guinea, showing intense hyperplasia with no colloid. From Buttfield and Hetzel (140).

 

Diagnosis

A diagnosis of endemic goiter implies that the cause is known, or at least strongly suspected. Usually water and food are found to have very low iodine content. The thyroid glands are often diffusely enlarged in childhood, but are almost always nodular in adults. The typical laboratory findings are elevated radioiodine thyroidal uptake (RAIU), normal or low T4 and FT4 levels, normal or elevated T3 levels, normal or elevated TSH levels, and diminished urinary 127I excretion. RAIU is typically suppressible when thyroid hormone is given, but not always. Scanning with radioiodine or TcO4- shows a mottled distribution of the isotope. Antithyroglobulin or thyroperoxidase antibodies are usually absent. In an area of endemic goiter, the diagnosis can be presumed if the goiter is a community problem, but one must always be wary of missing individual patients with thyroiditis, thyrotoxicosis or thyroid carcinoma.

Pathophysiology

When iodine intake is abnormally low, adequate secretion of thyroid hormones may still be achieved by marked modifications of thyroid activity. These adaptive processes include stimulation of the trapping mechanism of iodide by the thyroid as well as stimulation of the subsequent steps of the intrathyroidal metabolism of iodine leading to preferential synthesis and secretion of T3. They are triggered and maintained by increased secretion of TSH. The morphological consequence of prolonged thyrotropic stimulation is thyroid hyperplasia (108).

The first functional consequence of iodine deficiency is an increase in the uptake of iodide by the thyroid mediated via a transmembrane protein, the sodium iodide symporter (NIS) (109). There is a clear inverse relation between iodine supply and thyroidal uptake of radioiodide. The increased uptake may be accompanied by and may result from an increase in the serum levels of TSH. However, elevated TSH in endemic goiter is usually systematically found only in conditions of extreme iodine deficiency. In conditions of mild iodine deficiency, elevated TSH is typically found in only a small fraction of subjects, usually the youngest (110). It is possible that it is the sensitivity of the thyroid to TSH rather than the TSH level itself that mainly varies with iodide supply. However, whatever the relative roles of TSH levels and sensitivity to TSH, the thyroid is stimulated as demonstrated by increased secretion and elevated serum levels of thyroglobulin.

For any adequate adjustment of iodine supply to the thyroid, iodide trapping must fulfill two conditions. First, it must reduce the amount of iodide excreted in the urine to a level corresponding to the level of iodine intake in order to preserve the preexisting iodine stores. Second, it must ensure the accumulation in the thyroid of definite amounts of iodide per day, estimated at least 100 μg/day in adolescents and adults. The increase in the iodide clearance by the thyroid despite the decrease in the serum concentration of iodide maintains a normal absolute uptake of iodide by the thyroid and an organic iodine content of the thyroid which remains within the limits of normal (i.e., 10-20 mg) as long as the iodine intake remains above a threshold of about 50 μg/day. Below this critical level of iodine intake, despite a further increase of thyroid iodide clearance, the absolute uptake of iodide diminishes and the iodine content of the thyroid decreases with functional consequences resulting in the development of a goiter (111,112).

Thyroid hyperplasia induced by iodine deficiency is associated with an altered pattern of thyroid hormonogenesis: the abnormal configuration of the poorly iodinated thyroglobulin in the thyroid colloid is accompanied by an increase in poorly iodinated compounds, monoiodotyrosine (MIT) and T3, and a decrease in diiodotyrosine (DIT) and T4. The increase of the MIT/DIT and T3/T4 ratios is closely related to the degree of iodine depletion of the gland (113).

The T3/T4 ratio in the serum may be elevated in conditions of iodine deficiency because: 1) thyroidal secretion of T4 and T3 is in the proportion in which they exist within the gland; and/or 2) preferential secretion of T3 or increased peripheral conversion of T4 to T3. The shift to increased T3 secretion plays an important role in the adaptation to iodine deficiency because T3 possesses about 4 times the metabolic potency of T4 but requires only 75 % as much iodine for synthesis.

However, efficient adaptation to iodine deficiency is possible in the absence of goiter as demonstrated in nongoitrous patients in endemic goiter areas such as New Guinea (114) and the Congo (115). Moreover, adequate adaptation to iodine deficiency has been demonstrated in areas of severe iodine deficiency in the absence of endemic goiter (116). This clearly indicates that goiter is not required for achieving efficient adaptation to iodine deficiency. Rather, in these conditions, efficient adaptation to iodine deficiency is possible thanks to a high iodide trapping capacity but with only a slight enlargement of the thyroid. At this stage, the characteristic hyperplastic picture includes abundant parenchyma, high follicular epithelium and rare colloid.

On the contrary, in large goiters, the major part of the gland is occupied by extremely distended vesicles filled with colloid with a flattened epithelium. The mechanism responsible for the development of colloid goiter is not fully understood (117), but it does not appear to be TSH hyperstimulation. It must be the consequence of an imbalance between thyroglobulin synthesis and hydrolysis, i.e. secretion. In these conditions, iodide is diluted while thyroglobulin is in excess, resulting in a lesser degree of iodization of thyroglobulin and, consequently, in a decrease in iodothyronine synthesis and secretion (118). Hydrolysis of large amounts of poorly iodinated thyroglobulin will result in an important leak of iodide by the thyroid and enhanced urinary loss of iodide, further aggravating the state of iodine deficiency (119). Therefore, large colloid goiters in endemic iodine deficiency represent maladaptation instead of adaptation to iodine deficiency because they may produce a vicious cycle of iodine loss and defective thyroid hormones synthesis.

Endemic cretinism

Epidemiology

When McCarrison described cretinism in north-western India during the first decade of this century (120), he delineated a neurologic form, with predominantly neuromotor defects, including strabismus, deaf-mutism, spastic diplegia, and other disorders of gait and coordination. The patients usually had a goiter. The other form, which he called the myxedematous form, showed evidence of severe hypothyroidism, short stature, and markedly delayed bone and sexual maturation. The patients usually had a thyroid normal in size and position, and were seldom deaf.

Neurological Cretinism

The three characteristic features of neurological endemic cretinism in its fully developed form are extremely severe mental deficiency together with squint, deaf mutism and motor spasticity with disorders of the arms and legs of a characteristic nature. (Figure 4). As would be expected with a deficiency disease, there is a wide range in the severity of the clinical features in the population affected (120-122).

Male from Ecuador about 40 years old, deaf-mute, unable to stand or walk. Use of the hands was strikingly spared, despite proximal upper-extremity spasticity.

Figure 4 (a). Male from Ecuador about 40 years old, deaf-mute, unable to stand or walk. Use of the hands was strikingly spared, despite proximal upper-extremity spasticity.

Male from South Eastern China with typical facies of neurological cretinism, who is also deaf -mute and suffering from a less severe proximal muscle weakness in lower limbs.

Figure 4 (b). Male from South western China with the typical facies of neurological cretinism, who is also deaf-mute and suffering from less severe proximal muscle weakness in lower limbs.

Mental deficiency is characterized by a marked impairment of the capacity for abstract thought but vision is unaffected. Autonomic, vegetative, personal, social functions and memory appear to be relatively well preserved except in the most severe cases.

Deafness is the striking feature. This may be complete in as many as 50% cretins. It has been confirmed by auditory brain stem evoked potential studies which showed no cochlear or brain stem responses even at the highest sound frequencies. These findings suggest a cochlear lesion. In subjects with reduced hearing a high tone defect is apparent. Deafness is sometimes absent in subjects with other signs of cretinism. Nearly all totally deaf cretins are mute and many with some hearing have no intelligible speech.

The motor disorder shows a characteristic proximal rigidity of both lower and upper extremities and the trunk. There is a corresponding proximal spasticity with markedly exaggerated deep tendon reflexes at the knees, adductors and biceps. Spastic involvement of the feet and hands is unusual or, if present, is much milder than that of the proximal limbs. Function of the hands and feet is characteristically preserved so that most cretins can walk. This observation is very useful in differentiating cretinism from other forms of cerebral palsy commonly encountered in endemic areas, such as cerebral palsy from birth injury or meningitis.

In addition to frank cretinism, a larger proportion of the population suffers from some degree of intellectual impairment and coordination defect. Comparative population based neuropsychological assessments of children in areas of iodine deficiency compared with areas with adequate iodine intake confirm a shift of the intelligence curve to the left in the iodine deficient areas. Careful examination of affected individuals in such areas reveals a pattern of neurological involvement similar to that seen in frank cretins, although of milder degree. In assessing these less severe defects, nonverbal tests are most helpful and school progress is a good indicator. After the age of 3 years drawings are very useful, indicating a defect in visual motor integration. Finally, elevated hearing thresholds have been reported in children with no other signs of endemic cretinism in conditions of mild iodine deficiency (123).

DeLong (124) suggests that the neuropathological basis of the clinical picture includes underdevelopment of the cochlea for deafness; maldevelopment of the cerebral neocortex for mental retardation; and maldevelopment of the corpus striatum (especially putamen and globus pallidus) for the motor disorder. The cerebellum, hypothalamus, visual system, and hippocampus are relatively spared.

Pathophysiology of neurological cretinism

Developmental neuropathology and available epidemiologic data suggest that the period from about 12-14 weeks until 20-30 weeks of gestation may be the critical period during which damage occurs (19). Cortical and striatal neuron proliferation, migration, and early formation of neuropil occur between 12 and 18 weeks. Cochlear development occurs at the same time. These data correlate well with the data from the Papua New Guinea trial which indicated that iodine repletion must occur by three months of pregnancy to prevent cretinism (35).

Studies already cited above on the effect of iodine deficiency on brain cell development in the newborn rat, sheep and marmoset suggest that iodine deficiency has an early effect on neuroblast multiplication. Brain weight is reduced and there are a reduced number of cells, a greater density of cells in the cerebral cortex and reduced cell acquisition in the cerebellum. Because maternal thyroxine crosses the placenta, it is now envisaged that neurological cretinism is predominantly caused by maternal hypothyroidism due to iodine deficiency (125). It has been suggested that an autosomal recessive predisposition, besides maternal iodine deficiency, may play an etiological role in neurological cretinism (126).

Myxedematous Cretinism

The typical myxedematous cretin (Fig 5) has a less severe degree of mental retardation than the neurological cretin, but has all the features of extremely severe hypothyroidism present since early life, as in untreated sporadic congenital hypothyroidism (127-129): severe growth retardation, incomplete maturation of the facial features including the naso-orbital configuration, atrophy of the mandibles, puffy features, myxedematous, thickened and dry skin, dry and decreased hair, eyelashes and eyebrows and much delayed sexual maturation.

 In the first panel are four inhabitants from the Democratic Republic of Congo, aged 15-20 years being a normal male and three females with severe longstanding hypothyroidism, atrophic thyroids, shortness of stature and retarded sexual maturation.

Figure 5. Myxedematous endemic cretinism in the Democratic Republic of Congo. Four inhabitants aged 15-20 years : a normal male and three females with severe longstanding hypothyroidism with dwarfism, retarded sexual development, puffy features, dry skin and hair and severe mental retardation.

Contrasting with the general population and with neurological cretinism, goiter is usually absent and the thyroid is often not palpable, suggesting thyroid atrophy. This diagnosis is confirmed by thyroid scans that show a thyroid in the normal location but of small volume with a very heterogeneous and patchy distribution of the tracer (114). Thyroidal uptake of radioiodine is much lower than in the general population. The serum levels of T4 and T3 are extremely low, often undetectable, and TSH is dramatically high. Markedly enlarged sella turcicas have been demonstrated, suggesting pituitary adenomas (130).

Myxedematous cretinism used to be particularly common in Zaire. Early reports indicated limited neurological abnormalities in the cretins in this country, but one has to be cautious in interpreting these reports as comprehensive neurological examinations had not been performed (128). The movements are torpid and the reflex relaxation is usually much prolonged. However, hyperreflexia and Babinski signs were occasionally reported while knock knees and flat feet were obvious in the photographs of these patients in the literature. Subsequent expert neurological examination of some of these patients by De Long (120) suggested some of them had the neurological signs reported in the neurological type of cretinism, but they were partly obscured by the status of severe hypothyroidism. This is an important finding as it indicates in utero damage from hypothyroxinemia from maternal iodine deficiency does occur in myxedematous cretinism and is followed by severe, irreversible hypothyroidism in infancy and childhood.

Etiology and Pathophysiology of myxedematous cretinism

Three additional factors, acting alone or in combination, have been proposed for explaining the particularity of thyroid atrophy characteristic of the myxedematous type of cretinism (131):

1) Thiocyanate overload resulting from the chronic consumption of poorly detoxified cassava (88). Its role has been suggested in Zaire from the observation that populations in areas with severe but uniform iodine deficiency exhibit cretinism only when a critical threshold in the dietary supply of SCN is reached. SCN crosses the placenta and inhibits the trapping of iodide by the placenta and fetal thyroid (41, 90). This explanation is not necessarily relevant to other areas such as western China where myxedematous cretinism has been described.

2) Selenium deficiency. Severe selenium deficiency has been reported in Zaire in populations where myxedematous cretinism is endemic (95,98). Selenium is present in glutathione peroxidase (Gpx) that detoxifies H2O2 produced in excess in thyroid cells hyperstimulated by TSH because of iodine deficiency. Accumulation of H2O2 within the thyroid cells could induce thyroid cell destruction and thyroid fibrosis resulting in myxedematous cretinism. It has been proposed that the combination of deficiencies in iodine and selenium and SCN overload are required for the occurrence of severe thyroid failure during the perinatal period, and subsequent development of myxedematous cretinism (95).

3) Immunological mechanisms. Some authors (132,133) but not others (134) suggested immunological factors cause destruction of the thyroid, both in endemic and sporadic congenital hypothyroidism. The role of autoimmunity in the etiology endemic cretinism remains controversial.

ASSESSMENT OF IODINE STATUS IN POPULATIONS

 

Four methods are generally recommended for assessment of iodine nutrition in populations: urinary iodine concentration (UI), the goiter rate, serum thyroid stimulating hormone (TSH), and serum thyroglobulin (Tg) (see overview in Table 6). These indicators are complementary, in that UI is a sensitive indicator of recent iodine intake (days) and Tg shows an intermediate response (weeks to months), whereas changes in the goiter rate reflect long-term iodine nutrition (months to years).

 

Thyroid size

 

Two methods are available for measuring goiter: neck inspection and palpation, and thyroid ultrasonography. By palpation, a thyroid is considered goitrous when each lateral lobe has a volume greater than the terminal phalanx of the thumbs of the subject being examined. In the classification system of WHO (1), grade 0 is defined as a thyroid that is not palpable or visible, grade 1 is a goiter that is palpable but not visible when the neck is in the normal position (i.e., the thyroid is not visibly enlarged), and grade 2 goiter is a thyroid that is clearly visible when the neck is in a normal position (see Figure 6). Goiter surveys are usually done in school age children.

 

However, palpation of goiter in areas of mild iodine deficiency has poor sensitivity and specificity; in such areas, measurement of thyroid volume (Tvol) by ultrasound is preferable (135). Thyroid ultrasound is non-invasive, quickly done (2-3 mins per subject) and feasible even in remote areas using portable equipment. However, interpretation of Tvol data requires valid references from iodine-sufficient children. In a recent multicenter study, Tvol was measured in 6-12 y-old children (n=3529) living in areas of long-term iodine sufficiency on five continents. Age- and body surface area- specific 97th percentiles for Tvol were calculated for boys and girls (136). Goiter can be classified according to these international reference criteria, but they are only applicable if Tvol is determined by a standard method (1).

 

In areas of endemic goiter, although thyroid size predictably decreases in response to increases in iodine intake, thyroid size may not return to normal for months or years after correction of iodine deficiency (137,138). During this transition period, the goiter rate is difficult to interpret, because it reflects both a population’s history of iodine nutrition and its present status. Aghini-Lombardi et al. (138) suggested that enlarged thyroids in children who were iodine deficient during the first years of life may not regress completely after introduction of salt iodization. If true, this suggests that to achieve a goiter rate <5% in children may require that they grow up under conditions of iodine sufficiency. A sustained salt iodization program will decrease the goiter rate by ultrasound to <5% in school-age children and this indicates disappearance of iodine deficiency as a significant public health problem (1). WHO recommends the total goiter rate be used to define severity of iodine deficiency in populations using the following criteria: <5%, iodine sufficiency; 5.0%–19.9%, mild deficiency; 20.0%–29.9%, moderate deficiency; and >30%, severe deficiency (1).

 

Urinary iodine concentration (UIC)

 

Because >90% of ingested iodine is excreted in the urine, UI is an excellent indicator of recent iodine intake. UI can be expressed as a concentration (µg/L), in relationship to creatinine excretion (µg iodine/g creatinine), or as 24-hour excretion (µg/day), which is termed urinary iodine excretion (UIE). For populations, because it is impractical to collect 24-hour samples in field studies, UI can be measured in spot urine specimens from a representative sample of the target group, and expressed as the median, in µg/L (1). Variations in hydration among individuals generally even out in a large number of samples, so that the median UI in spot samples correlates well with that from 24-hour samples. For national, school-based surveys of iodine nutrition, the median UI from a representative sample of spot urine collections from ≈1200 children (30 sampling clusters x 40 children per cluster) can be used to classify a population’s iodine status (1) (Table 7).

 

However, the median UI is often misinterpreted. Individual iodine intakes, and, therefore, spot UI concentrations are highly variable from day-to-day (139), and a common mistake is to assume that all subjects with a spot UI <100 µg/L are iodine deficient. To estimate iodine intakes in individuals, 24-hour collections are preferable, but difficult to obtain. An alternative is to use the age-and sex adjusted iodine:creatinine ratio in adults, but this also has limitations (140). Creatinine may be unreliable for estimating daily iodine excretion from spot samples, especially in malnourished subjects where creatinine concentration is low. Daily iodine intake for population estimates can be extrapolated from UI, using estimates of mean 24-hour urine volume and assuming an average iodine bioavailability of 92% using the formula: Urinary iodine (µg/L) x 0.0235 x body weight (kg) = daily iodine intake (5). Using this formula, a median UI of 100 μg/L corresponds roughly to an average daily intake of 150 μg.

 

Thyroid stimulating hormone

 

Because serum thyroid stimulating hormone (TSH) concentration is determined mainly by the level of circulating thyroid hormone, which in turn reflects iodine intake, TSH can be used as an indicator of iodine nutrition. However, in older children and adults, although serum TSH may be slightly increased by iodine deficiency, values often remain within the normal range. TSH is therefore a relatively insensitive indicator of iodine nutrition in adults (1). In contrast, TSH is a sensitive indicator of iodine status in the newborn period (141, 142). Compared to the adult, the newborn thyroid contains less iodine but has higher rates of iodine turnover. Particularly when iodine supply is low, maintaining high iodine turnover requires increased TSH stimulation. Serum TSH concentrations are therefore increased in iodine deficient infants for the first few weeks of life, a condition termed transient newborn hypothyroidism or hyperthyrotropinemia. In areas of iodine deficiency, an increase in transient newborn hypothyroidism, indicated by >3 % of newborn TSH values above the threshold of 5 mIU/L in whole blood collected 3 to 4 days after birth, suggests but is not diagnostic of iodine deficiency in the population (141, 142). TSH is used in many countries for routine newborn screening to detect congenital hypothyroidism. If already in place, such screening offers a sensitive indicator of population iodine nutrition providing the timing of heel-stick blood collection is standardised and the TSH assay is modified to report TSH values in the range from normal to the cut-off level for diagnosis of congenital hypothyroidism (141). Newborn TSH is an important measure because it reflects iodine status during a period when the developing brain is particularly sensitive to iodine deficiency.

 

Thyroglobulin

 

Thyroglobulin (Tg) is synthesized only in the thyroid, and is the most abundant intrathyroidal protein. In iodine sufficiency, small amounts of Tg are secreted into the circulation, and serum Tg is normally <10 µg/L (143). In areas of endemic goiter, serum Tg increases due to greater thyroid cell mass and TSH stimulation. Serum Tg is well correlated with the severity of iodine deficiency as measured by UI (144). Intervention studies examining the potential of Tg as an indicator of response to iodized oil and potassium iodide have shown that Tg falls rapidly with iodine repletion, and that Tg is a more sensitive indicator of iodine repletion than TSH or T4 (145,146). However, commercially-available assays measure serum Tg, which requires venipuncture, centrifugation and frozen sample transport, which may be difficult in remote areas.

 

A new assay for Tg has been developed for dried blood spots taken by a finger prick (147,148), simplifying collection and transport. In prospective studies, dried blood spot Tg has been shown to be a sensitive measure of iodine status and reflects improved thyroid function within several months after iodine repletion (147,148). However, several questions need to be resolved before Tg can be widely adopted as an indicator of iodine status. One question is the need for concurrent measurement of anti-Tg antibodies to avoid potential underestimation of Tg; it is unclear how prevalent anti-Tg antibodies are in iodine deficiency, or whether they are precipitated by iodine prophylaxis (149). Another limitation is large interassay variability and poor reproducibility, even with the use of standardization (143). This has made it difficult to establish normal ranges and/or cutoffs to distinguish severity of iodine deficiency. However, recently international reference ranges for DBS Tg in iodine-sufficient school children (4-40 μg/L) has been made available (147) and a similar reference range proposed for DBS Tg in iodine sufficient pregnant women (150).

 

Thyroid hormone concentrations

 

In contrast, thyroid hormone concentrations are poor indicators of iodine status. In iodine-deficient populations, serum T3 increases or remains unchanged, and serum T4 usually decreases. However, these changes are often within the normal range, and the overlap with iodine-sufficient populations is large enough to make thyroid hormone levels an insensitive measure of iodine nutrition (1).

 

 

Table 6. Indicators of iodine status at population level

 

Indicator

(units)

Age Group Advantages Disadvantages Application

Median urinary iodine concentration

(μg/L)

School-age children, adults and pregnant women

§  Spot urine samples are easy to obtain

§  Relatively low cost

§  External quality control program in place

 

§ Not useful for individual assessment

§ Assesses iodine intake only over the past few days

§ Meticulous laboratory practice needed to avoid contamination

§ Sufficiently large number of samples needed to allow for varying degrees of subject hydration

See Table 7
Goiter rate by palpation (%) School-age children

§  Simple and rapid screening test:

§  Requires no specialized equipment

§ Specificity and sensitivity are low due to a high inter-observer variation

§ Responds only slowly to changes in iodine intake

Degree of IDD by goiter rate :

§  0-4.9% - None

§  5-19.9% - Mild

§  20-29.9% -  Moderate

§  ≥30% - Severe

Goiter rate by ultrasound (%) School-age children

§  More precise than palpation

§  Reference values established as a function of age, sex, and body surface area

§ Requires expensive equipment and electricity

§ Operator needs special training

§ Responds only slowly to changes in iodine intake

Thyroid stimulating hormone

(mIU/L)

Newborns

§  Measures thyroid function at a particularly vulnerable age

§  Minimal costs if a congenital hypothyroidism screening program is already in place

§  Collection by heel stick and storage on filter paper is simple

§ Not useful if iodine antiseptics used during delivery

§ Requires a standardized, sensitive assay

§ Should be taken by heel-prick at least 48 hours after birth to avoid physiological newborn surge

A <3% frequency of TSH values >5 mIU/L indicates iodine sufficiency in a population

Serum or whole blood thyroglobulin

(μg/L)

School-age children and adults

§  Collection by finger stick and storage on filter paper is simple

§  International reference range available

§  Measures improving thyroid function within several months after iodine repletion

§ Expensive immunoassay

§ Standard reference material is available, but needs validation

Reference interval in iodine-sufficient children is 4-40 μg/L

 

 

 

Table 7. Epidemiological criteria for assessing iodine nutrition in a population based on median and/or range of urinary iodine concentrations (from ref.1).

 

Median urinary iodine (μg/L) Iodine intake Iodine nutrition
School-aged children
<20 Insufficient Severe iodine deficiency
20-49 Insufficient Moderate iodine deficiency
50-99 Insufficient Mild iodine deficiency
100-199 Adequate Optimal
200-299 More than adequate Risk of iodine-induced hyperthyroidism in susceptible groups
>300 Excessive Risk of adverse health consequences (iodine-induced hyperthyroidism, autoimmune thyroid disease and hypothyroidism)
Pregnant women  
< 150 Insufficient  
150 – 249 Adequate  
250 – 499 More than adequate  
≥ 500 Excessivea  
Lactating womenb  
< 100 Insufficient  
≥ 100 Adequate  
Children less than 2 years old  
< 100 Insufficient  
≥ 100 Adequate  

The term “excessive” means in excess of the amount required to prevent and control iodine deficiency.

b  In lactating women, the figures for median urinary iodine are lower than the iodine requirements because of the iodine excreted in breast milk.

 Three women from the Himalayas with large grade 2 multinodular goiters.

Figure 6. Three women of the Himalayas with stage II goiters.

IODINE FORTIFICATION AND SUPPLEMENTATION

Iodized salt

Iodized salt is considered the most appropriate measure for iodine fortification (1). The advantage of iodized salt is that it is used by nearly all sections of a community, irrespective of social and economic status. It is consumed as a condiment at roughly the same level throughout the year. Its production is often confined to a few centers so that fortification can occur on a large scale and with better controlled conditions. There are two forms of iodine which can be used to iodize salt: iodide and iodate, usually as the potassium salt. Iodate is less soluble and more stable than iodide and is therefore preferred for tropical moist conditions. When used, both are generally referred to as "iodized" salt.

The daily requirement of iodine is 150 µg per person for adults (1). The level of iodization of salt has to be sufficient to cover this requirement, considering potential losses from the point of production to the point of consumption, including the expected shelf life. It also should take into account the per capita salt consumption in an area. Although salt consumption in the range 10-15 g per day is common in developed countries, this is regarded as excessive because of a potential increased risk of hypertension. Therefore, intakes in the range of 3-6 g per day, or even less, are being recommended. This potential reduction in salt intakes should be taken into account when setting iodine levels in fortified salt. Iodized salt can also be used as a feed supplement for cattle and other livestock in iodine deficient areas. Allowing for these factors, the level of iodine as iodate currently recommended to provide 150 ug of iodine per day is in the range of 20-40 mg per kg salt (1).

The packaging of the iodized salt is very important. Jute bags have been used extensively but in humid conditions, the salt absorbs moisture. The iodate dissolves and will drip out of the bag if it is porous, and much of the iodine will be lost; up to 75% over a period of nine months. To avoid this, waterproofing is required, achieved by a polythene lining inside the jute bag or else a plastic bag. The additional cost of a plastic bag may be justified by reduced iodine losses and the potential resale value of the bags (151,152).

The use of iodized salt in the prevention of IDD has been reviewed (151). The control of the iodine concentration in salt at production level should be performed by using titration methods to provide quantitative data or, in the case of imported salt, by using reliable test kits to provide qualitative data at the point of entry. Consignments with suspect iodine levels should be rechecked by titration. National monitoring programs should: 1) periodically check salt iodine levels in retail shops and households using reliable test kits; 2) conduct occasionally goiter prevalence surveys; and 3) regularly measure urinary iodine. In order to determine the proportion of households using adequately iodized salt in a large geographic area, it is recommended to use cluster surveys at the provincial or national levels. It is also recommended to identify high risk communities where there are inadequate proportions of households using adequately iodized salt.

Salt iodization remains the most cost-effective way of delivering iodine and of improving cognition in iodine-deficient populations (153). Worldwide, the annual costs of salt iodization are estimated at 0.02-0.05 US$ per child covered, and the costs per child death averted are US$ 1000 and per DALY gained are US$34-36 (153). Looked at in another way, prior to widespread salt iodization, the annual potential losses attributable to iodine deficiency in the developing world have been estimated to be US$35.7 billion as compared with an estimated US$0.5 billion annual cost for salt iodization, i.e., a 70:1 benefit:cost ratio (154).

 

Iodine supplements

 

In areas of iodine deficiency where iodized salt is not available, iodine supplements are recommended by expert groups for women of reproductive age, pregnant women and lactating women (1,155). All pregnant women should consume approximately 250 µg iodine daily (1,155). To achieve a total of 250 µg iodine ingestion daily, strategies may need to be varied based on country of origin; in some countries, iodized oil supplements (see below) given once a year may be appropriate, in others, potassium iodide supplements may be given daily (1,155). In most regions, including the United States, women who are planning pregnancy or currently pregnant, should supplement their diet with a daily oral supplement that contains 150 µg of iodine in the form of potassium iodide (155). This is optimally started at least 3 months in advance of planned pregnancy. There is no need to initiate iodine supplementation in pregnant women who are being treated for hyperthyroidism or who are taking LT4 (155).

Iodized oil

Iodized oil ("Lipiodol®") was first used for the correction of iodine deficiency in Papua New Guinea (73). Buttfield and Hetzel (156) demonstrated the effectiveness of a single iodized oil injection (4 ml) in correcting iodine deficiency for a period of up to 4 1/2 years. Another trial in the Western Highlands of New Guinea demonstrated prevention of endemic cretinism and a reduction in fetal and neonatal deaths in the iodine treated group, if the iodized oil injection was given before pregnancy (126). Goiter in the treated population often resolved one to three months after the injection. However, the administration of iodized oil to adults with multinodular goiters in several other countries has not seen resolution or even diminution in size of their goiters.

Extensive additional studies on the use of iodized oil in the correction and prevention of IDD have been conducted in Latin America, Africa, Asia and Eastern Europe (157). The physiology and pharmacology of iodized oil in goiter prophylaxis has been extensively reviewed (158). Experience has confirmed the convenience of 200-400 mg the oral administration of iodized oil at yearly intervals through the primary health care system at the village level. In general, the effect of oral administration lasts half the time of the same dose given by injection (159-161). In regions of moderate-to-severe iodine deficiency without effective salt iodisation, lactating women who receive one dose of 400 mg iodine as oral iodised oil soon after delivery can provide adequate iodine to their infants through breastmilk for at least 6 months, enabling the infants to achieve euthyroidism (162). Recommendations from WHO-UNICEF-ICCIDD for oral iodized oil supplementation of women and children are shown in Table 8.

.

Table 8. Recommendations for iodine supplementation in pregnancy and infancy in areas where <90% of households are using iodized salt and the median UI is <100 µg/L in schoolchildren (from ref.1)

 

Women of child bearing age

A single annual oral dose of 400 mg of iodine as iodized oil

OR

A daily oral dose of iodine as potassium iodide should be given so that the total iodine intake meets the RNI of 150 µg/d of iodine.

Women who are pregnant or lactating

A single annual oral dose of 400 mg of iodine as iodized oil

OR

A daily oral dose of iodine as potassium iodide should be given so that the total iodine intake meets the new RNI of 250 µg/d iodine.

  • Iodine supplements should not be given to a woman who has already been given iodized oil during her current pregnancy or up to 3 months before her current pregnancy started
Children aged 0-6 months

A single oral dose of 100 mg of iodine as iodized oil

OR

A daily oral dose of iodine as potassium iodide should be given so that the total iodine intake meets the of 90 µg/d of iodine

  • Should be given iodine supplements only if the mother was not supplemented during pregnancy or if the child is not being breast-fed.
Children aged 7-24 months old

A single annual oral dose of 200 mg of iodine as iodized oil as soon as possible after reaching 7 months of age

OR

A daily oral dose of iodine as potassium iodide should be given so that the total iodine intake meets the RNI of 90 µg/d of iodine

 

An iodized oil supplementation program is necessary when other methods have been found ineffective or are inapplicable. Iodized oil can be regarded as an emergency measure for the control of severe IDD until an effective iodized salt program can be introduced. The spectacular and rapid effects of iodized oil in reducing goiter can be important in demonstrating the benefits of iodization, which can lead to community demand for iodized salt. In general, iodized oil administration should be avoided over the age of 45 because of the possibility of precipitating hyperthyroidism in subjects with longstanding goiter (see further in section VI 3).

The possibility of linking up an iodized oil program with other preventative programs, such as the Child Immunization Program, has been discussed (163). Great progress has been made with child immunization programs in Africa and Asia. To this series of measures, oral iodized oil administration could readily be added to cover young children over the first 2-5 years of life. Women of reproductive age would require separate coverage through the primary health care system, especially the family planning health care system or in antenatal services at the same time as with tetanus toxoid.

Iodized bread

Iodized bread has been used effectively in the State of Tasmania in Australia (164). Successful use of iodized bread was also reported in Russia (165). Since 2009 by law in Australia and New Zealand, all salt used in baking of bread and similar edible products must be iodized salt. Early results from urinary iodine monitoring in Australia since implementation of this mandatory practice has seen correction of mild iodine deficiency in the population as a whole (6).

Iodized water

Water has some of the advantages of salt as a vehicle for iodine fortification. Both are daily necessities and thus their iodization will reach the most vulnerable groups – the poor and the isolated. Water fortified at a regular rate with iodine provides the thyroid with a steady daily ration, which is physiologically desirable (166).

Systems for iodization of drinking water can be classified as follows:

  1. Silicone elastomers releasing iodine. A commercial version of this approach is the “Rhodifuse” system of Rhône-Poulenc-Rorer-Doman (now Adventis). Silicone matrices containing 30 % sodium iodide are placed in polyethylene baskets. When the baskets are placed in wells, sodium iodide is released into the water according to the porosity and the surface/volume ratio. Initially successfully used in Mali (167), the system was then used in Burkina Fasso and the Central African Republic. Limitations to the system were climatic conditions, the cost of the diffuser and its maintenance. A similar device was manufactured locally in Malaysia (168).
  2. Iodide added to running water in pipes. A commercial example is the Hydroline system used in Sicily (169). The apparatus consisted of a canister filled with coarse crystals of iodine through which water was diverted from the line by a pressure differential. The system was highly efficient with a cost estimated at $ 0.04/person/day. A similar system used in the Sarawak region of Malaysia resulted in the reduction of goiter rate from 61% to 30% within 9 months together with improved thyroid function (170).
  3. Iodide added to run-off water. This technique was used by Cao at all (171) in Southern Xinjiang, China. It resulted in an increase of iodine intake by plants, possible benefits on rice production, increased iodine uptake by the thyroids of sheep and chickens, increased iodine in eggs and increased survival of newborn lambs.
  4. Manual addition of iodine to standing water. In this approach, iodine is added directly to vessels containing drinking water. It has been used most notably in northern Thailand, particularly to improve iodine nutrition in schoolchildren.

A review of water iodization programs (166) concluded that when properly monitored, the procedure is efficient in controlling iodine deficiency in smaller communities. But it is generally more expensive than iodized salt in large-scale national programs and that it is unlikely to be self-sustaining in poor rural countries and thus requires permanent external funding.

CURRENT GLOBAL STATUS OF IDD CONTROL PROGRAMS

Until 1990, only a few countries Switzerland, some of the Scandinavian countries, Australia, the U.S. and Canada were completely iodine sufficient. Since then, globally, the number of households using iodized salt has risen from <20% to >70%, dramatically reducing iodine deficiency (172). This effort has been achieved by a coalition of international organizations, including ICCIDD (now IGN), WHO, MI and UNICEF, working closely with national IDD control committees and the salt industry; this informal partnership was established after the World Summit for Children in 1990.

 

The two most commonly used approaches to assessing iodine nutrition on the population level are estimation of the household penetration of adequately iodized salt (HHIS) and measurement of urinary iodine concentrations (UICs) (173). UIC surveys are usually done in school aged children (SAC), because they are a convenient population, easy to reach through school based surveys and usually representative of the general population (173). Therefore, WHO use UICs from 6-12 y-old children in nationally-representative surveys, expressed as the median in µg/L, to classify a population’s iodine status (Table 1). More countries are beginning to carry out studies in high-risk population groups, i.e. women of reproductive age, pregnant women and younger children, however data is limited and the majority of countries still conduct routine iodine monitoring in SAC (174).

 

In 2017, representative UIC surveys are available for 139 countries. There are no up-to-date UIC data available for 55 countries. Available UIC data now cover >98% of the world’s population of SAC (174).

 

 

Figure 7: Shows countries classified by iodine nutrition in 2017 according to degree of public health importance based on the median UIC. Iodine intake is inadequate in 19 countries, adequate in 110 and excessive in 10. There are no up-to-date UIC data available for 55 countries. Reference (174).

Overall, approximately 75% of households worldwide have access to iodized salt. Those with the greatest access are living in the WHO regions of the Western Pacific and the Americas, and those with the least access are residing in the Eastern Mediterranean region (175,176).

 

The International Child Development Steering Group identified iodine deficiency as one of four key global risk factors for impaired child development where the need for intervention is urgent (177). But controlling IDD in the remaining 1/3rd of the global population at risk will not be easy. Although the key contributors to successful national programs have been identified (1), reaching economically disadvantaged groups living in remote areas and convincing small scale salt producers to iodize their salt are major challenges. An important strategy will be to strengthen national coalitions that include government partners, national and international agencies, the health-care sector and salt producers. In the countries that have begun iodized salt programs, sustainability will become a major focus. These programs are fragile and require a long-term commitment from governments. In several countries where iodine deficiency had been eliminated, salt iodization programs fell apart, and iodine deficiency recurred (178). Children in iodine deficient areas are vulnerable to even short-term lapses in iodized salt programs (179). To this end, countries should monitor the state of their iodine nutrition every three years and report to the World Health Assembly on their progress (180).

 

Advocacy should focus on damage to reproduction and cognitive development. Governments need to understand the serious impact of iodine deficiency; many still equate iodine deficiency with goiter, a mostly cosmetic problem and thus a low priority. IDD is one of the most important causes of preventable neurocognitive impairment worldwide, and elimination of IDD can contribute to at least five of the Millennium Development Goals (181): 1) Eradicate extreme poverty and hunger; 2) Achieve universal primary education; 3) Reduce child mortality; 4) Improve maternal health; and 5) Develop a global partnership for development. The World Bank (182) strongly recommends that governments invest in micronutrient programs, including salt iodization, to promote development, concluding: “Probably no other technology offers as large an opportunity to improve lives at such low cost and in such a short time.”

MONITORING THE IMPACT OF PROGRAMS OF SALT IODIZATION

The social process for successful implementation of a national IDD control program includes the following components (1):

  • situation assessment
  • communication of results to health professionals, political authorities and the public
  • development of an action plan
  • implementation of the plan
  • evaluation of its impact at population level

The last phase, monitoring, is often neglected not only because it is the last phase in the process, but because it may be overshadowed by other components of the program such as implementation. In addition, many countries affected by IDD are low-income countries without the financial or technical resources to support a laboratory needed to properly monitor salt quality and iodine status.

The most cost-effective way to achieve the virtual elimination of IDD is through universal salt iodization, USI. The indicators used in monitoring and evaluating IDD control programs include both indicators to monitor and evaluate the salt iodization process, as well as indicators to monitor the impact of salt iodization on the target populations (these have been discussed previously).

Table 10 summarizes the criteria for monitoring progress towards sustainable elimination of IDD as a public health problem (1). It is considered that iodine deficiency has been eliminated from a country when:

  • access to iodized salt at household level is at least 90%
  • the median urinary iodine concentration is at least 100 μg/L and with less than 20 % of the samples below 50 μg/L
  • when at least 8 of the 10 program indicators listed in Table 10 are implemented

Table 10. Summary of criteria for monitoring progress towards sustainable elimination of IDD as a public health problem (ref.1)

Indicators Goals

Salt Iodization

Proportion of households using

adequately iodized salt

> 90 %

Urinary iodine

Proportion below 100 μg/L

Proportion below 50 μg/L

< 50 %

< 20 %

Programmatic indicators

  • An effective, functional national body (council or committee) responsible to the government for the national program for the elimination of IDD (this body should be multidisciplinary, involving the relevant fields of nutrition, medicine, education, the salt industry, the media, and consumers, with a chairman appointed by the Minister of Health)
  • Evidence of political commitment to universal salt iodization and the elimination program
  • Appointment of a responsible executive officer for the IDD elimination program
  • Legislation or regulations on universal salt iodization (while ideally regulations should cover both human and agricultural salt, if the latter is not covered this does not necessarily preclude a country from being certified as IDD-free)
  • Commitment to assessment and reassessment of progress in the elimination of IDD, with access to laboratories able to provide accurate data on salt and urinary iodine
  • A program of public education and social mobilization on the importance of IDD and the consumption of iodized salt
  • Regular data on salt iodine at the factory, retail and household levels
  • Regular laboratory data on urinary iodine in school-aged children, with appropriate sampling for higher risk areas
  • Cooperation from the salt industry in maintenance of quality control
  • A database for recording of results or regular monitoring procedures, particularly for salt iodine, urinary iodine and, if available, neonatal TSH, with mandatory public reporting.

Currently, there is much less information available on the impact of salt iodization programs than on the implementation of programs. The monitoring data of all countries affected by IDD are summarized country by country in an up-to-date database held by WHO (173).

Also, surprisingly, few longitudinal or case control studies have addressed the influence of USI on disorders induced by iodine deficiency, such as impairment of thyroid function, low birth weight, perinatal mortality and morbidity and the prevention of mental retardation. The oft-quoted statement that correction of iodine deficiency protects 50-100 million neonates from brain damage and mental retardation annually is politically attractive, but scientifically questionable. It results simply from a multiplication of the birth rate of the affected countries by the percentage of access to iodized salt at household level. Both figures lack precision. Moreover, this calculation implies that 100 % of neonates born in iodine deficient areas before the implementation of programs of iodine supplementation suffered intellectual impairment, which is a gross overestimation.

THE RISKS OF EXCESS IODINE INTAKE

As discussed so far in this chapter, iodine deficiency impairs thyroid function. Similarly, iodine excess, including overcorrection of a previous state of iodine deficiency, can also impair thyroid function. The effect of iodine on the thyroid gland is complex with a “U shaped” relation between iodine intake and risk of thyroid diseases. Both low and high iodine intake are associated with an increased risk of thyroid disorders. Healthy adults with normal thyroid glands can tolerate up to 600-1100 μg iodine/day without any side effects (5,183) (Table 11). However, this upper limit is much lower in a population which has been exposed to iodine deficiency for a prolonged period in the past. The optimal level of iodine intake to prevent any thyroid disease may be a relatively narrow range around the recommended daily intake of 150 μg (184).

Table 11. Tolerable upper intake level for iodine (µg/day)

 

Age group EC/SCF, 2002 IOM, 2001
1-3 years 200 200
4-6 years 250 300
7-10 years 300 300
11-14 years 450 300
15-17 years 500 900
Adult 600 1100
Pregnant women >19 years 600 1100

 

Iodide goiter and iodine-induced hypothyroidism

When iodine intake is chronically high, as in coastal areas of Japan (91) due to daily intake of seaweeds rich in iodine or, in Eastern China, because of the high iodine content of the drinking water from shallow wells (92), the prevalence of thyroid enlargement and goiter is high, as compared to populations with normal iodine intakes. Also, the prevalence of subclinical hypothyroidism is elevated. The mechanisms behind this impairment of thyroid function are probably both iodine enhancement of thyroid autoimmunity and reversible inhibition of thyroid function by excess iodine (the Wolff-Chaikoff effect) in susceptible subjects (185). However, this type of thyroid failure has not been observed in neonates after the administration of huge doses of iodized oil to their mothers during pregnancy (162). Increased thyroid volume in children due to iodine excess has been observed when the median urinary iodine is >500 μg/L (186).

Iodine-induced hyperthyroidism, IIH

Iodine-induced hyperthyroidism (IIH) is the main complication of iodine prophylaxis. It has been reported in most iodine supplementation programs (187). But it is rare following a well executed program of iodine supplementation, for example as in Iran (188). The outbreak of IIH most extensively investigated occurred in Tasmania in the late 1960’s. This followed iodine supplementation simultaneously by tablets of iodide, iodized bread and the use of iodophors by the milk industry (189). The incidence of hyperthyroidism increased from 24/100000 in 1963 to 125/100000 in 1967. The disease occurred most frequently in individuals over 40 years of age with multinodular goiter and preexisting heart diseases (189). The most severe manifestations were cardiovascular and were occasionally fatal. The epidemic lasted about 10 to 12 years, but it was followed by an incidence of hyperthyroidism somewhat less than that existing prior to the epidemic.

The introduction of iodized salt in Zimbabwe resulted in a sharp increase in the incidence of IIH from 3/100000 to 7/100000 over 18 months (190). A high risk of IIH was also reported from Eastern Congo following the introduction of iodized salt (191). A multicenter study conducted in seven African countries, including Zimbabwe and Congo (192) showed that the occurrence of IIH in the last two countries was due to the sudden introduction of poorly monitored and excessively iodized salt in populations which had been severely iodine deficient for very long periods in the past. The conclusion of the study was that the risk of IIH was related to a rapid increment of iodine intake resulting in a state of acute iodine overload.

IIH following iodine fortification of salt cannot be entirely avoided even when fortification provides only physiological amounts of iodine. In a well-controlled longitudinal study in Switzerland, the incidence of hyperthyroidism transiently increased by 27% during one year after the iodine supply was increased from 90 μg/day to 150 μg/day (193).

The reason for the development of IIH after iodine fortification and/or supplementation is thought to be that iodine deficiency increases thyrocyte proliferation and mutation rates which, in turn, trigger the development of multifocal autonomous growth with scattered cell clones harboring activation mutations of the TSH receptors (194). Measurement of total intrathyroidal iodine by means of X-ray fluorescence scanning shows that only some nodules keep their capacity to store iodine, become autonomous and cause hyperthyroidism (195). It should be noted that there is considerable anecdotal evidence that increased iodine intake in patients with Graves’ disease may exacerbate hyperthyroidism in susceptible patients.

It thus appears that IIH can be considered one of the iodine deficiency disorders, and it may be largely unavoidable in the early phase of iodine repletion in iodine deficient populations, particularly in those with moderate to severe iodine deficiency. Its incidence reverts to normal or even below normal after one to ten years of iodine supplementation (196).

Iodine-induced thyroiditis

Another potential complication of excessive iodine intake is the aggravation or the induction of autoimmune thyroiditis by iodine supplementation. In experimental conditions, excessive iodine intake can precipitate spontaneous thyroiditis in genetically predisposed strains of beagles, rats or chickens (196). Potential mechanisms involved in iodine-induced thyroiditis in animal models include: 1) triggering of thyroid autoimmune reactivity by increasing the immunogenicity of thyroglobulin; and/or 2) damage to the thyroid and cell injury by free radicals.

Attention was drawn to iodine-induced thyroiditis in humans when studies conducted in the United States following the implementation of salt iodization showed an increased frequency of Hashimoto’s thyroiditis in goiters removed by surgery (197). Studies following the introduction of iodized oil in Greece pointed out the possible development of thyroid autoantibodies (198). Kahaly et al. (199) reported the development of thyroid autoantibodies and lymphocytic infiltration in 6 out of 31 patients with endemic goiter treated during 6 months with a dose of 500 μg potassium iodide (KI) per day. Acute massive iodine overload (daily consumption of at least 50 mg iodine daily) in healthy adults resulted in a sharp increase in thyroid peroxidase antibody titers together with elevated prevalence of goiter and serum TSH values. The prevalence of all abnormalities decreased after removal of iodine excess (202).

Finally, cross sectional studies of populations with different iodine intakes in Italy (63), Great Britain (64) and more recently in Denmark and Iceland (65) showed that the frequency of thyroid autoantibodies and hypothyroidism is higher in iodine replete populations than in iodine deficient populations. It is also recognized that the frequency of thyroid antibodies (200) and of autoimmune thyroiditis (201) is higher in the United States than in Europe, while the iodine intake is lower in Europe.

Hypothyroidism

To investigate the effects of iodine intake on thyroid disorders in China, a prospective 5-year survey was done in three rural communities with mildly deficient, more than adequate (previously mildly deficient iodine intake), and excessive iodine intake (median UIs of 88, 214 and 634 µg/L, respectively) (203,204). High iodine intakes did not increase rates of overt hypothyroidism or hyperthyroidism, but did increase cumulative incidence of subclinical hypothyroidism (0.2 percent, 2.6 percent, and 2.9 percent, respectively) and autoimmune thyroiditis (0.2 percent, 1.0 percent, and 1.3 percent, respectively). In most people, these disorders were not sustained.

 

Denmark has documented the pattern of thyroid disease after careful introduction of iodized salt. Pederson et al (205,206) prospectively identified new cases of overt hypothyroidism and hyperthyroidism in Denmark before and for the first 6-7 yr after introduction of iodized salt. There was a moderate increase in the incidence rate of overt hypothyroidism (RR = 1.35; 95% CI = 1.11-1.66) that occurred primarily in young and middle-aged subjects with previously moderate iodine deficiency. The overall incidence rate of hyperthyroidism also increased, from 102.8 to 138.7/100,000/year. But in contrast to IIH, many of the new cases were observed in younger adults (20-39 y), and were presumably of autoimmune origin.

Thyroid cancer (TC)

In animals, chronic overstimulation of the thyroid by TSH can produce thyroid neoplasms (207). However, the relationship between thyroid cancer and endemic goiter has long been debated without agreement on a possible causal relationship (208-212). The available evidence suggests iodine deficiency is a risk factor for thyroid cancer, particularly for follicular TC and possibly, for anaplastic TC (213). A recent review (213) concluded that: a) there are consistent data showing an increase in thyroid cancer (mainly follicular) in iodine deficient animals; b) there is a plausible mechanism (chronic TSH stimulation induced by iodine deficiency); c) there is consistent data from before and after studies of iodine prophylaxis showing a decrease in follicular thyroid cancer and anaplastic thyroid cancer; d) there is an indirect association between changes in iodine intake and thyroid cancer mortality in the decade from 2000 to 2010; e) autopsy studies of occult thyroid cancer show higher microcarcinoma rates with lower iodine intakes; and f) case control studies suggest a lower risk of TC with higher total iodine intakes.

It appears the prognosis of thyroid cancer is significantly improved following iodine supplementation due to a shift towards differentiated forms of thyroid cancer that are diagnosed at earlier stages. Overall, it appears that correction of iodine deficiency decreases the risk of, and the morbidity from, thyroid cancer (2013).

Thus, the benefits of correcting iodine deficiency far outweigh its risks (63, 214, 215). Iodine-induced hyperthyroidism and other adverse effects can be almost entirely avoided by adequate and sustained quality assurance and monitoring of iodine supplementation which should also confirm adequate iodine intake.

In summary, enormous progress has been made globally over the past two decades in understanding and eliminating iodine deficiency as the major cause of preventable brain damage in the fetus, newborn and infant and as a cause of thyroid disorders in adults. Three quarters of the world’s population now has access to iodine on a daily basis through edible iodized salt, but sustainability remains a challenge in many countries. Many recent excellent clinical research studies have revealed that IDD are not confined to remote, mountainous areas in developing countries as we once thought, but are a global public health problem that affects most countries, including developed countries and island nations (216). The recognition of the universality of iodine deficiency highlights the need for more research into the pathogenesis and consequences of mild to moderate iodine deficiency and the development of new strategies to establish and maintain sustainable IDD elimination.

REFERENCES

1. World Health Organization/International Council for the Control of the Iodine Deficiency Disorders/United Nations Childrens Fund (WHO/ICCIDD/UNICEF). Assessment of the iodine deficiency disorders and monitoring their elimination. Geneva: World Health Organization, 2007.
2. Assey VD, Greiner T, Mzee RK, et al. Iodine deficiency persists in the Zanzibar Islands of Tanzania. Food Nutr Bull 2006; 27: 292–99.
3. Pearce EN, Pino S, He X, Bazrafshan HR, Lee SL, Braverman LE. Sources of dietary iodine: bread, cows’ milk, and infant formula in the Boston area. J Clin Endocrinol Metab 2004; 89: 3421–24.
4. Haldimann M, Alt A, Blanc A, Blondeau K. Iodine content of food groups. J Food Comp Anal 2005; 18: 461–71.
5. Institute of Medicine of the National Academies Dietary reference intakes for vitamin A, vitamin K, arsenic, boron, chromium, copper, iodine, iron, manganese, molybdenum, nickel, silicon, vanadium and zinc. Washington, DC: National Academy Press, 2001.
6. El-Ghawi UM, Al-Sadeq AA. Determination of iodine in Libyan food samples using epithermal instrumental neutron activation analysis. Biol Trace Elem Res 2006; 111: 31–40.
7. Charlton, K., Probst, Y., Kiene, G. 2016. Dietary iodine intake of the Australian population after introduction of a mandatory iodine fortification programme. Nutrients. Nov 4 ; 8(11).
8. Chavasit V, Malaivongse P, Judprasong K. Study on stability of iodine in iodated salt by use of diff erent cooking model conditions. J Food Comp Anal 2002; 15: 265–76.
9. Phillips DI. Iodine, milk, and the elimination of endemic goitre in Britain: the story of an accidental public health triumph. J Epidemiol Community Health 1997; 51: 391–93.
10. Li, J.Q., Wang, X., Yan, Y., Wang, K., Qin, D., Xin, Z., and Wei, J. 1985. The effects of severely iodine deficient diet derived from an endemic area on fetal brain development in the rat. Observations in the first generation. Neuropathol. and Appl. Neurobiol. 12:261-270.
11. Zhong, F.G., Cao, X.M., and Liu, J.L. 1983. Experimental study on influence of iodine deficiency on fetal brain in rats. Chinese J. Pathol. 12:205-216.
12. Hetzel, B.S., and Hay, I.D. 1979. Thyroid function, iodine nutrition and fetal brain development. Clin. Endocrinol. 11:445-460.
13. McMichael, A.J., Potter, J.D., and Hetzel, B.S. 1980. Iodine deficiency, thyroid function and reproductive failure. In Endemic goiter and endemic cretinism. Iodine nutrition in health and disease. J.B. Stanbury, and B.S. Hetzel, editors. New York: John Wiley publ. 445-460.
14. Ruiz-Marcos, A., Sanchez-Toscano, F., Rey, F.E.d., and Escobar, G.M.d. 1979. Severe hypothyroidism and the maturation of the rat cerebral cortex. Brain Res. 162:315-329.
15. Mano, M.T., Potter, B.J., Belling, G.B., Chevadej, J., and Hetzel, B.S. 1987. Fetal brain in response to iodine deficiency in a primate model Callithrix Jacchus Jacchus. J. Neurolog. Sciences 79 :287-300.
16. Potter, B.J., McIntosh, G.H., and Hetzel, B.S. 1981. The effect of iodine deficiency on fetal brain development in the sheep. In Fetal brain disorders. Recent approaches to the problem of mental deficiency. B.S. Hetzel, and R.M. Smith, editors. Amsterdam: Elsevier/North Holland Biomedical Press publ. 119-148.
17. Hetzel, B.S., Chevadej, J., and Potter, B.J. 1988. The brain in iodine deficiency. Neuropathology and Applied Neurobiology 14 : 93-104.
18. WHO, UNICEF, and ICCIDD. 1994. Indicators for assessing Iodine Deficiency Disorders and their control through salt iodization. Geneva: WHO publ. WHO/NUT/94.6. 1-55 pp.
19. Morreale de Escobar,G., Obregon, M.J. and Escobar del Rey, F. Role of thyroid hormone during early brain development. Eur. J. Endocrinol. 151 : U25-U 37, 2004.
20. Auso,E., Lavado-Autric, R., Cuevas, E., Escbar del Rey, F., Morreale de Escobar, G. and Berbel, P. A moderate and transient deficiency of maternal thyroid function at the beginning of fetal neocoticogenesis alters neuronal migration. Endocrinology. 145 : 4037-4047, 2004.
21. Koibuchi, N., and Chin, W.W. 2000. Thyroid hormone action and brain development. Trends Endocrinol. Metab. 4:123-128.
22. Chan, S., and Kilby, M.D. 2000. Thyroid hormone and central nervous system development. J. Endocrinol. 165:1-8.
23. Delange, F. 2000. Endemic cretinism. In The thyroid. A fundamental and clinical text. L.E. Braverman, and R.D. Utiger, editors. Philadelphia: Lippincott publ. 743-754.
24. Glinoer, D., and Delange, F. 2000. The potential repercussions of maternal, fetal and neonatal hypothyroxinemia on the progeny. Thyroid 10:871-887.
25. Delange, F. 2001. Iodine deficiency as a cause of brain damage. Postgrad. Med. J. 77:217-220.
26. Dobbing, J., and Sands, J. 1973. Quantitative growth and development of human brain. Arch. Dis. Child. 48:757-767.
27. Morreale de Escobar, G., Obregon, M.J., and Escobar del Rey, F. 2000. Is neuropsychological development related to maternal hypothyroidism or to maternal hypothyroxinemia ? J. Clin. Endocrinol. Metab. 85:3975-3987.
28. Delange, F., and Fisher, D.A. 1995. The thyroid gland. In Clinical Paediatric Endocrinology. 3rd Ed. C. Brook, editor. Oxford: Blackwell publ. 397-433.
29. Contempré, B., Jauniaux, E., Calvo, R., Jurkovic, D., Campbell, S., and Escobar, G.M.d. 1993. Detection of thyroid hormones in human embryonic cavities during the first trimester of gestation. J. Clin. Endocrinol. Metab. 77:1719-1722.
30. Bernal, J., and Pekonen, F. 1984. Ontogenesis of the nuclear 3,5,3'-triiodothyronine receptor in the human fetal brain. Endocrinology 114:677-679.
31. Kooistra, L., Crawford, S., van Baar, A.L., Brouwers, E.P., and Pop, V.J. 2006. Neonatal effects of maternal hypothyroxinemia during early 32. pregnancy. Pediatrics 117 :161-167.
Vulsma, T., Gons, M.H., and De Vijlder, J.J. 1989. Maternal-fetal transfer of thyroxine in congenital hypothyroidism due to a total organification defect or thyroid agenesis. N. Engl. J. Med. 321:13-16.
33. Pearce, E.N., Lazarus, J.H., Moreno-Reyes, R., Zimmermann, M.B. 2016. Consequences of iodine deficiency and excess in pregnant women: an overview of current knowns and unknowns. Am. J. Clin. Nutr. 104 : 918S-23S.
34. Thilly, C.H., Lagasse, R., Roger, G., Bourdoux, P., and Ermans, A.M. 1980. Impaired fetal and postnatal development and high perinatal death-rate in a severe iodine deficient area. In Thyroid Research VIII. J.R. Stockigt, S. Nagataki, E. Meldrum, J.W. Barlow, and P.E. Harding, editors. Canberra: Australian Academy of Science publ. 20-23.
35. Pharoah, P.O.D., and Connolly, K.J. 1987. A controlled trial of iodinated oil for the prevention of endemic cretinism : a long-term follow-up. Intern. J. Epidemiol. 16:68-73.
36. Cobra, C., Muhilal, Rusmil, K., Rustama, D., Djatnika, Suwardi, S.S., Permaesih, D., Muherdiyantinigsih, Martuti, S., and Semba, R.D. 1997. Infant survival is improved by oral iodine supplementation. J. Nutr. 127:574-578.
37. Delong, G.R., Leslie, P.W., Wang, S.-H., Jiang, X.-M., Zhang, M.-L., Rakeman, M.A., Jiang, J.-Y., Ma, T., and Cao, X.-Y. 1997. Effect on infant mortality of iodination of irrigation water in a severely iodine-deficient area of China. Lancet 350:771-773.
38. Dobbing, J. 1974. The later growth of the brain and its vulnerability. 39. Pediatrics 53:2-6.
Delange, F., Heidemann, P., Bourdoux, P., Larsson, A., Vigneri, R., Klett, M., Beckers, C., and Stubbe, P. 1986. Regional variations of iodine nutrition and thyroid function during the neonatal period in Europe. Biol. Neonate 49:322-330.
40. Kochupillai, N., and Pandav, C.S. 1987. Neonatal chemical hypothyroidism in iodine-deficient environments. In The prevention and control of Iodine Deficiency Disorders. B.S. Hetzel, J.T. Dunn, and J.B. Stanbury, editors. Amsterdam: Elsevier publ. 85-93.
41. Delange, F., Thilly, C., Bourdoux, P., Hennart, P., Courtois, P., and Ermans, A.M. 1982. Influence of dietary goitrogens during pregnancy in humans on thyroid function of the newborn. In Nutritional factors involved in the goitrogenic action of cassava. F. Delange, F.B. Iteke, and A.M. Ermans, editors. Ottawa: International Development Research Centre publ. 40-50.
42. Vanderpas, J., Bourdoux, P., Lagasse, R., Rivera, M., Dramaix, M., Lody, D., Nelson, G., Delange, F., Ermans, A.M., and Thilly, C.H. 1984. Endemic infantile hypothyroidism in a severe endemic goitre area of Central Africa. Clin. Endocrinol. 20:327-340.
43. Delange, F. 1990. Iodine nutrition and risk of thyroid irradiation from nuclear accidents. In Iodine prophylaxis following nuclear accidents. E. Rubery, and E. Smales, editors: Pergamon Press publ. 45-53.
44. Aghini-Lombardi, F., Pinchera, A., Antonangeli, L., Rago, T., Chiovato, L., Bargagna, S., Bertucelli, B., Ferretti, G., Sbrana, B., Marcheschi, M., et al. 1995. Mild iodine deficiency during fetal/ neonatal life and neuropsychological impairment in Tuscany. J. Endocrinol. Invest. 18:57-62.
45. Santiago-Fernandez, P., Torres-Barahona, R., Muela-Martinez, J.A., Rojo-Martinez, G., Garcia-Fuentes,E, Garriga, M.J., Leon, A. G. and Soriguer, F. Intelligence quotient and iodine intake : a cross-sectional study in children. J. Clin. Endocrinol. Metab. 89 : 3851-3857, 2004.
46. Zimmermann, M.B., Connolly, K., Bozo, M., Bridson, J., Rohner, F., and Grimci, L. 2006. Iodine supplementation improves cognition in iodine-deficient schoolchildren in Albania : a randomized, controlled, double-blind study. Am. J. Clin. Nutr. 83 :108-114.
47. Gordon RC, Rose MC, Skeaff SA, Gray AR, Morgan KM, Ruffman T. 2009. Iodine supplementation improves cognition in mildly iodine-deficient children. Am. J. Clin. Nutr. 90(5) : 1264-71.
48. Fierro-Benitez, R., Ramirez, I., Estrella, E., and Stanbury, J.B. 1974. The role of iodine deficiency in intellectual development in an area of endemic goiter. In Endemic goiter and cretinism : continuing threats to world health. J. Dunn, and G.A. Medeiros-Neto, editors. Washington: Pan American Health Organization (PAHO) Scientific publ. 135-142.
49. Kochupillai, N., Pandav, C.S., Godbole, M.M., Metha, M., and Ahuja, M.M.S. 1986. Iodine deficiency and neonatal hypothyroidism. Bull. WHO 64:547-551.
50. Bleichrodt, N., and Born, M.P. 1994. A metaanalysis of research on iodine and its relationship to cognitive development. In The damaged brain of iodine deficiency. J.B. Stanbury, editor. New York: Cognizant Communication publ. 195-200.
51. Qian M, Wang D,Watkins WE, Gebski V, Yan YQ, Li M, Chen ZP 2005. The effects of iodine on intelleigence in children: a meta-analysis of studies conducted in China. Asia pacific J Clin Nutr 14:32-42.
52. Bleichrodt, N., Rey, F.E.D., Escobar, G.M.d., Garcia, I., and Rubio, C. 1989. Iodine deficiency. Implications for mental and psychomotor development in children. In Iodine and the brain. G.R. DeLong, J. Robbins, and P.G. Condliffe, editors. New York: Plenum Press publ. 269-287.
53. Vermiglio, F., Sidoti, M., Finocchiaro, M.D., Battiato, S., Presti, V.P.L., Benvenga, S., and Trimarchi, F. 1990. Defective neuromotor and cognitive ability in iodine-deficient schoolchildren of an endemic goiter region in Sicily. J. Clin. Endocrinol. Metab. 70:379-384.
54. Fenzi, G.F., Giusti, L.F., Aghini-Lombardi, F., Bartalena, L., Marcocci, C., Santini, F., Bargagna, S., Brizzolara, D., Ferretti, G., Falciglia, G., et al. 1990. Neuropsychological assessment in schoolchildren from an area of moderate iodine deficiency. J. Endocrinol. Invest. 13:427-431.
55. Vitti, P., Aghini-Lombardi, F., Antonangeli, L., Rago, T., Chiovato, L., Pinchera, A., Marcheschi, M., Bargagna, S., Bertuccelli, B., Ferretti, G., et al. 1992. Mild iodine deficiency in fetal/neonatal life and neuropsychological performances. Acta Medica Austriaca 19:57-59.
56. Tiwari, B.D., Godbole, M.M., Chattopadhyay, N., Mandal, A., and Mithal, A. 1996. Learning disabilities and poor motivation to achieve due to prolonged iodine deficiency. Am. J. Clin. Nutr. 63:782-786.
57. Azizi, F., Sarshar, A., Nafarabadi, M., Ghazi, A., Kimiagar, M., Noohi, S., Rahbar, N., Bahrami, A., and Kalantari, S. 1993. Impairment of neuromotor and cognitive development in iodine-deficient schoolchildren with normal physical growth. Acta Endocrinol. 129:501-504.
58. Shresta, R.M. 1994. Effect of iodine and iron supplementation on physical, psychomotor and mental development in primary schoolchildren in Malawi.: University of Wageningen. 1-105.
59. van den Briel, T., West, C.E., Bleichrodt, N., Vijver, F.J.v.d., Ategbo, E.A., and Hautvast, J.G.A.J. 2000. Improved iodine status is associated with improved mental performance of schoolchildren in Benin. Am. J. Clin. Nutr. 72:1179-1185.
60. Zimmermann MB, Jooste PL, Mabapa NS, et al. Treatment of iodine defi ciency in school-age children increases insulin-like growth factor (IGF)-I and IGF binding protein-3 concentrations and improves somatic growth. J Clin Endocrinol Metab 2007; 92: 437–42.
61. Mason JB, Deitchler M, Gilman A, et al. Iodine fortification is related to increased weight-for-age and birthweight in children in Asia. Food Nutr Bull 2002; 23: 292–308.
62. Wan Nazaimoon WM, Osman A, Wu LL, Khalid BA. Eff ects of iodine deficiency on insulin-like growth factor-I, insulin-like growth factor-binding protein-3 levels and height attainment in malnourished children. Clin Endocrinol 1996; 45: 79–83.
63. Zimmermann, M.B., Boelaert, K. 2015. Iodine deficiency and thyroid disorders. Lancet Diabetes Endocrinol. 3(4) : 286-95.
64. Aghini-Lombardi, F., Antonangeli, L., Martino, E., Vitti, P., Maccherini, D., Leoli, F., Rago, T., Grasso, L., Valeriano, R., Balestrieri, A., et al. 1999. The spectrum of thyroid disorders in an iodine-deficient community : the Pescoporgano survey. J. Clin. Endocrinol. Metab. 84:561-566.
65. Vanderpump, M.P.J., Tunbridge, W.M.G., French, J.M., Appeton, D., Bates, D., Clark, F., Evants, J.G., Hasan, D.M., Rodgers, H., Tunbridge, F., et al. 1995. The incidence of thyroid disorders in the community : a twenty-year follow-up of the Wickham Survey. Clin. Endocrinol. 43:55-68.
66. Laurberg, P., Nohr, S.B., Pedersen, K.M., Hreidarsson, A.B., Andersen, S., Pedersen, I.B., Knudsen, N., Perrild, H., Jorgensen, T., and Ovesen, L. 2000. Thyroid disorders in mild iodine deficiency. Thyroid 10:951-963.
67. Szabolcs, I., Podoba, J., Feldkamp, J., Dohan, O., Farkas, I., Sajgo, M., Takats, K.I., Goth, M., Kovacs, L., Kressinszky, K., et al. 1997. Comparative screening for thyroid disorders in old age in areas of iodine deficiency, long-term iodine prophylaxis and abundant iodine intake. Clin. Endocrinol. 47:87-92.
68. Garcia-Mayor, R.V., Rios, M., Fluiters, E., Mendes, L.F.P., Garcia-Mayor, E.G., and Andrade, A. 1999. Effect of iodine supplementation on a pediatric population with mild iodine deficiency. Thyroid 9:1089-1093.
69. Clesen, R. 1929. Distribution of endemic goiter in the United States as shown by thyroid surveys. Public Hlth. Rep. US 44:1463.
70. Hollowell, J.G., Staehling, N.W., Hannon, W.H., Flanders, D.W., Gunter, E.W., Maberly, G.F., Braverman, L.E., Pino, S., Miller, D.T., and Garbe, P.L. 1998. Iodine nutrition in the United States. Trends and public health implications : iodine excretion data from National Health and Nutrition Examination Surveys I and III (1971-1974 and 1988-1994). J. Clin. Endocrinol. Metab. 83:3401.
71. Caldwell, K.L., Jones, R. and Hollowell, J. G. 2005. Urinary iodine concentration : United States National Health and Nutrition Examination Survey 2001-2002. Thyroid 15 :692-699.
72. Kelly, F.C., and Snedden, W.W. 1960. Prevalence and geographical distribution of endemic goitre. In Endemic goitre. Geneva: World Health Organization publ. 27-234.
73. Andersson, M., Takkouche, B., Egli, I., Allen, H.E. and de Benoist, B. 2005. Current global iodine status and progress over the last decade towards the elimination of iodine deficiency. Bulletin of the WHO 83:518-525.
74. Hetzel, B.S., and Pandav, C.S. 1996. S.O.S. for a billion. The conquest of Iodine Deficiency Disorders. 2nd Ed. Dehli: Oxford University Press publ. 1-466 pp.
75. Delange, F. 2000. Iodine deficiency. In The thyroid. A fundamental and clinical text. L.E. Braverman, and R.D. Utiger, editors. Philadelphia: Lippincott publ. 295-316.
76. Gaitan, E. 1980. Goitrogens in the etiology of endemic goiter. In Endemic goiter and endemic cretinism. Iodine nutrition in health and disease. J.B. Stanbury, and B.S. Hetzel, editors. New York: John Wiley publ. 219-236.
77. Gaitan, E. 1989. Environmental goitrogenesis. Boca Raton: CRC Press publ. 1-250 pp.
78. Van Wyk, J.J., Arnold, M.B., Wynn, J., and Pepper, F. 1959. The effect of a soybean producton on thyroid function in humans. Pediatrics 24:752.
79. Van Etten, C.H. 1969. Goitrogens. In Toxic constituents of plant foodstuffs. I.E. Liener, editor. New York: Academic Press publ. 103-142.
80. Chesney, A.M., Clawson, T.A., and Webster, B. 1928. Endemic goitre in rabbits. I. Incidence and characteristics. Bull Johns Hopkins Hosp 43:261-277.
81. Barker, M.H. 1936. The blood cyanate in the treatment of hypertension. JAMA 106:762-767.
82. Hercus, C.E., and Purves, H.D. 1936. Studies on endemic and experimental goitre. J. Hyg. (Camb) 36:182.
83. MacKenzie, C.G., and MacKenzie, J.B. 1943. Effect of sulfonamides and thioureas on the thyroid gland and basal metabolism. Endocrinology 32:185-209.
84. Astwood, E.B., Greer, M.A., and Ettlinger, M.G. 1949. L-5-Vinyl-2-Thiooxazolidone, and thyroid compound from yellow turnip and from brassica seeds. J. Biol. Chem. 181:121.
85. Ermans, A.M., Mbulamoko, N.M., Delange, F., and Ahluwalia, R. 1980. Role of cassava in the etiology of endemic goitre and cretinism. Ottawa: International Development Research Centre publ. 1-182 pp.
86. Ekpechi, O.L. 1967. Pathogenesis of endemic goitre in Eastern Nigeria. Br. J. Nutr. 21:537-545.
87. Cowan, J.W., Saghir, A.R., and Salji, J.P. 1967. Antithyroid activity of onion volatiles. Aust. J. Biol. Sci. 20:683.
88. Delange, F., Thilly, C., and Ermans, A.M. 1968. Iodine deficiency, a permissive condition in the development of endemic goiter. J. Clin. Endocrinol. Metab. 28:114-116.
89. Delange, F., Iteke, F.B., and Ermans, A.M. 1982. Nutritional factors involved in the goitrogenic action of cassava. Ottawa: International Development Research Centre publ. 1-100 pp.
90. Delange, F., and Ahluwalia, R. 1983. Cassava toxicity and thyroid : research and public health issues. Ottawa: International Development Research Centre publ. 1-148 pp.
91. Roti, E., Gnudi, A., Braverman, L.E., Robuschi, G., Emanuele, R., Bandini, P., Benassi, L., Pagliani, A., and Emerson, C.H. 1981. Human cord blood concentrations of thyrotropin, thyroglobulin, and iodithyronines after maternal administration of thyrotropin-releasing hormone. J. Clin. Endocrinol. Metab. 53:813-817.
92. Suzuki, H., Higuchi, T., Sawa, K., Ohtaki, S., and Horiuchi, Y. 1965. "Endemic coast goitre" in Hokkaido, Japan. Acta Endocr (Kbh) 50:161-176.
93. Li, M., Liu, D.R., Qu, C.Y., Zhang,Y., Qian, Q.D., Jia, Q.Z., Wang, H.X., Eastman, C.J., Boyages, S.C. 1987  Endemic goitre in central China caused by excessive iodine intake. Lancet 2(8553):257-259.
94. Zhao, J., Wang, P., Shang, L., Sullivan, K.M., Haar, F.v.d., and Maberly, G. 2000. Endemic goiter associated with high iodine intake. Amer. J. Publ. Hlth 90:1633-1635.
95. Elnour, A., Hambraeus, L., Eltom, M., Dramaix, M. and Bourdoux, P. 2000. Endemic goiter with iodine sufficiency: a possible role of the consumption of pearl millet in the etiology of endemic goiter. Am. J. Clin. Nutr. 71 :59-66.
96. Contempré, B., Morrealle de Escobar, G., Denef, J.F., Dumont J.E., and Many, M.C. 2004. Thiocyanate induces cell necrosis and fibrosis in selenium – and iodine – deficient rat thyroids : a potential experimental model for myxedematous endemic cretinism in Central Africa. Endocrinology 145 :994-1002.
97. Vanderpas, J.B., Contempré, B., Duale, N.L., Goossens, W., Bebe, N., Thorpe, R., Ntambue, K., Dumont, J., Thilly, C.H., and Diplock, A.T. 1990. Iodine and selenium deficiency associated with cretinism in Northern Zaire. Am. J. Clin. Nutr. 52:1087-1093.
98. Thomson, C.D., McLachlan, S.K., Grant, A.M., Paterson, E., and Lillico, A.J. 2005. The effect of selenium on thyroid status in a population with marginal selenium and iodine status. Brit. J. Nutr. 94 :962-968.
99. Contempré, B., Dumont, J.E., Bebe, N., Thilly, C.H., Diplock, A.T., and Vanderpas, J. 1991. Effect of selenium supplementation in hypothyroid subjects of an iodine and selenium deficient area : the possible danger of indiscriminate supplementation of iodine deficient subjects with selenium. J. Clin. Endocrinol. Metab. 73:213-215.
100. Contempré, B., Denef, J.F., Dumont, J.E., and Many, M.C. 1993. Selenium deficiency aggravates the necrotizing effects of a high iodide dose in iodine deficient rats. Endocrinology 132:1866-1868.
101. Ma, T., Guo, J., and Wang, F. 1993. The epidemiology of iodine-deficiency diseases in China. Am. J. Clin. Nutr. Suppl. 57:2645-2665.
102. Zimmermann MB.The infl uence of iron status on iodine utilization and thyroid function. Annu Rev Nutr 2006; 26: 367–89.
103. Zimmermann MB, Bürgi H, Hurrell RF. Iron deficiency predicts poor maternal thyroid status during pregnancy. J Clin Endocrinol Metab 2007; 92: 3436–40.
104. Zimmermann MB, Jooste PL, Mabapa NS, et al. Vitamin A supplementation in iodine-deficient African children decreases thyrotropin stimulation of the thyroid and reduces the goiter rate. Am J Clin Nutr 2007; 86: 1040–44.
105. Correa, P. 1980. Pathology of endemic goiter. In Endemic goiter and endemic cretinism. J.B. Stanbury, and B.S. Hetzel, editors. New York: John Wiley and Sons publ. 303-332.
106. Studer, H., and Gebel, F. 1986. Sporadic goiter. In The Thyroid. S.H. Ingbar, and L.E. Braverman, editors. Philadelphia: J.P. Lippincott publ. 1311-1315.
107. Marine, D. 1935. The pathogenesis and prevention of simple or endemic goiter. JAMA 104:2334.
108. Stanbury, J.B., Brownell, G.L., Riggs, D.S., Perinetti, H., Itoiz, J., and Castillo, E.B.D.1954.Endemic goiter. The adaptation of man to iodine deficiency. Cambridge: Harvard University Press publ. 1-209 pp.
109. Spitzweg, C. and Morris, J.C. 2002. The sodium iodide symporter : its pathophysiological and therapeutic implications. Clin. Endocrinol. 57 :559-574.
110. Wayne, E.J., Koutras, D.A., and Alexander, W.D. 1964. Clinical aspects of iodine metabolism. Oxford: Blackwell publ. 1-303 pp.
111. Ermans, A.M. 1969. Intrathyroid iodine metabolism in goiter. In Endemic goiter. J.B. Stanbury, editor. Washington: Pan American Health Organization Scientific publ. 1-13.
112. Ermans, A.M., Dumont, J.E., and P.A.Bastenie. 1963. Thyroid function in a goiter endemic. I. Impairment of hormone synthesis and secretion in the goitrous gland. J. Clin. Endocrinol. Metab. 23:539-549.
113. Delange, F. 1994. The disorders induced by iodine deficiency. Thyroid. 4:107-128.
114. Choufoer, J.C., Van Rhijn, M., Kassenaar, A.A.H., and Querido, A. 1963. Endemic goiter in Western New Guinea : Iodine metabolism in goitrous and non-goitrous subjects. J. Clin. Endocrinol. Metab. 23:1203-1217.
115. Delange, F. 1974. Endemic goitre and thyroid function in Central Africa. Monographs in Pediatrics. Basel: S. Karger publ. 1-171 pp.
116. Roche, M., Perinetti, H., and Barbeito, A. 1961. Urinary excretion of stable iodine in a small group of isolated Venezuelan Indians. J. Clin. Endocrinol. Metab. 21:1009-1012.
117. Studer, H., and Greer, M.A. 1968. The regulation of thyroid function in iodine deficiency. Bern: Hans Huber publ. 1-118 pp.
118. Dumont, J.E., Ermans, A.M., Maenhaut, G., Coppée, F., and Stanbury, J.B. 1995. Large goiter as a maladaptation to iodine deficiency. Clin. Endocrinol. 43:1-10.
119. Ermans, A.M., Dumont, J.E., and Bastenie, P.A. 1963. Thyroid function in a goitrous endemic. II. Nonhormonal iodine escape from the goitrous gland. J. Clin. Endocrinol. Metab. 23:550-560.
120. McCarrisson, R. 1908. Observations on endemic cretinism in the Chitral and Gilgit valleys. Lancet ii:1275-1280.
121. DeLong, G.R., Stanbury, J.B., and Fierro-Benitez, R. 1985. Neurological signs in congenital Iodine Deficiency Disorder (Endemic cretinism). Dev. Med. Child. Neurol. 27:317-324.
122. Boyages, S.C., Halpern, J.P., Maberly, G.F., Eastman, C.J., Morris, J., J.Collins, Jupp, J.J., Chen-En, J., Zheng-Hua, W., and Chuan-Yi, Y. 1988. A comparative study of neurological and myxedematous endemic cretinism in Western China. J. Clin. Endocrinol. Metab. 67:1262-1271.
123. van Den Briel, T., West, C.E., Hautvast, J.G.A.J. and Ategbo, E.A. 2001. Mild iodine deficiency is associated with elevated hearing thresholds in children in Benin. Eur. J. Clin. Nutr. 55 :763-768.
124. DeLong, R. 1987. Neurological involvement in Iodine Deficiency Disorders. In The prevention and control of Iodine Deficiency Disorders. B.S. Hetzel, J.T. Dunn, and J.B. Stanbury, editors. Amsterdam : Elsevier publ. 49-63.
125. Cao, X.Y., Jiang, X.M., Dou, Z.H., Rakeman, M.A., Zhang, M.L., O’Donnell, K., Ma, T., Amette, K., Delong, N., and Delong, G.R. 1994. Timing of vulnerability of the brain to iodine deficiency in endemic cretinism. N. Engl. J. Med 331 :1739-1744.
126. Pharoah, P.O.D., Buttfield, I.H., and Hetzel, B.S. 1971. Neurological damage to the fetus resulting from severe iodine deficiency during pregnancy. Lancet i :308-310.
127. Dumont, J.E., Ermans, A.M., and Bastenie, P.A. 1963. Thyroidal function in a goiter endemic. IV. Hypothyroidism and endemic cretinism. J. Clin. Endocrinol. Metab. 23 :325-335.
128. Delange, F., Ermans, A.M., Vis, H.L., and Stanbury, J.B. 1972. Endemic cretinism in Idjwi Island (Kivu Lake, Republic of the Congo). J. Clin. Endocrinol. Metab. 34 :1059-1066.
129. Delange, F. 1992. Rôle de l'iode dans la nutrition infantile. Les troubles dûs à la déficience iodée et leur prévention. Doc. Scient. Guigoz 133:1-14.
130. Melot, G.J., Jeanmart-Michez, L., Dumont, J., Ermans, A.M., and Bastenie, P. 1962. Les aspects radiologiques du crétinisme endémique. J. Belg. Radiol. 45 :385-403.
131. Thilly, C.H., Vanderpas, J., Bourdoux, P., Lagasse, R., Lody, D., Smitz, J., and Ermans, A.M. 1983. Prevention of myxedematous cretinism with iodized oil during pregnancy. In Current problems in thyroid research. N. Ui, K. Torizuka, S. Nagataki, and K. Miyai, editors. Amsterdam : Excerpta Medica publ. 386-389.
132. Boyages, S.C., Maberly, G.F., Chen, J., Gaag, R.D.V.d., Halpern, J.P., Eastman, C.J., and Drexhage, H. 1989. Endemic cretinism : possible role for thyroid autoimmunity. Lancet ii :529-532.
133. Medeiros-Neto, G., Tsuboi, K., and Lima, N. 1989. Thyroid autoimmunity and endemic cretinism. Lancet ii :111.
134. Chiovato, L., Vitti, P., Bendinelli, G., Santini, F., Fiore, E., Tonacchera, M., Mammoli, C., Capaccioli, A., Ventury, S., Pretell, E., et al. 1995. Humoral thyroid autoimmunity is not involved in the pathogenesis of myxedematous endemic cretinism. J. Clin. Endocrinol. Metab. 80 :1509-1514.
135. Zimmermann M, Saad A, Hess S, Torresani T, Chaouki N. Thyroid ultrasound compared with World Health Organization 1960 and 1994 palpation criteria for determination of goiter prevalence in regions of mild and severe iodine defi ciency. Eur J Endocrinol 2000; 143: 727–31.
136. Zimmermann MB, Hess SY, Molinari L, et al. New reference values for thyroid volume by ultrasound in iodine-sufficient schoolchildren: a WHO/NHD Iodine Deficiency Study Group Report. Am J Clin Nutr 2004; 79: 231–37.
137. Zimmermann MB, Hess SY, Adou P, Toresanni T, Wegmüller R, Hurrell RF. Thyroid size and goiter prevalence after introduction of iodized salt: a 5-yr prospective study in schoolchildren in Côte d’Ivoire. Am J Clin Nutr 2003; 77: 663–67.
138. Aghini-Lombardi F, Antonangeli L, Pinchera A, Leoli F, Rago T, Bartolomei AM & Vitti P (1997) Effect of iodized salt on thyroid volume of children living in an area previously characterized by moderate iodine deficiency. J Clin Endocrinol Metab 82, 1136–1139.
139. Andersen S, Karmisholt J, Pedersen KM & Laurberg P (2008) Reliability of studies of iodine intake and recommendations for number of samples in groups and in individuals. Br J Nutr 99, 813–818.
140. Knudsen N, Christiansen E, Brandt-Christensen M, Nygaard B & Perrild H (2000) Age- and sex-adjusted iodine/creatinine ratio. A new standard in epidemiological surveys? Evaluation of three different estimates of iodine excretion based on casual urine samples and comparison to 24 h values. Eur J Clin Nutr 54, 361–363.
141. Li. M., & Eastman, C.J. (2010) .Neonatal TSH secretion: is it a sensitive and reliable tool for monitoring iodine status in the population? Best Practice & Research Clin Endocrinol & Metab 24,63-75.
142. Zimmermann MB, Aeberli I, Torresani T & Burgi H (2005) Increasing the iodine concentration in the Swiss iodized salt program markedly improved iodine status in pregnant women and children: a 5-y prospective national study. Am J Clin Nutr 82, 388–392.
143. Spencer CA & Wang CC (1995) Thyroglobulin measurement. Techniques, clinical benefits, and pitfalls. Endocrinol Metab Clin North Am 24, 841–863.
144. Knudsen N, Bulow I, Jorgensen T, Perrild H, Ovesen L & Laurberg P (2001) Serum Tg – a sensitive marker of thyroid abnormalities and iodine deficiency in epidemiological studies J Clin Endocrinol Metab 86, 3599–3603
145. Benmiloud M, Chaouki ML, Gutekunst R, Teichert HM, Wood WG & Dunn JT (1994) Oral iodized oil for correcting iodine deficiency: optimal dosing and outcome indicator selection. J Clin Endocrinol Metab 79, 20–24.
146. Missler U, Gutekunst R & Wood WG (1994) Thyroglobulin is a more sensitive indicator of iodine deficiency than thyrotropin: development and evaluation of dry blood spot assays for thyrotropin and thyroglobulin in iodine-deficient geographical areas. Eur J Clin Chem Clin Biochem 32, 137–143.
147. Zimmermann MB, de Benoist B, Corigliano S, et al. (2006) Assessment of iodine status using dried blood spot thyroglobulin: development of reference material and establishment of an international reference range in iodine-sufficient children. J Clin Endocrinol Metab 91, 4881–4887.
148. Zimmermann MB, Moretti D, Chaouki N & Torresani T (2003) Development of a dried whole-blood spot thyroglobulin assay and its evaluation as an indicator of thyroid status in goitrous children receiving iodized salt. Am J Clin Nutr 77, 1453–1458.
149. Zimmermann MB, Moretti D, Chaouki N & Torresani T (2003) Introduction of iodized salt to severely iodine-deficient children does not provoke thyroid autoimmunity: a one-year prospective trial in northern Morocco. Thyroid 13, 199–203.
150. Stinca S, Andersson M, Weibel S, Herter-Aeberli I, Fingerhut R, Gowachirapant S, Hess SY, Jaiswal N, Jukic T, Kusic Z, Mabapa NS, Nepal AK, San Luis TO, Zhen JQ, Zimmermann MB (2017) Dried Blood Spot Thyroglobulin as a Biomarker of Iodine Status in Pregnant Women. J Clin Endocrinol Metab 102(1):23-32.
151. Mannar, V.M.G., and Dunn, J.T. 1995. Salt iodization for the elimination of iodine deficiency. MI, ICCIDD, UNICEF, WHO publ:1-126.
152. Sullivan, K.M., Houston, R., Gorstein, J. and Cervinkas, J. 1995. Monitoring universal salt iodization programmes. Atlanta, USA. PAMM-MI-ICCIDD publ.:1-101.
153. Caulfield LE, Richard SA, Rivera JA, Musgrove P, Black RE. Stunting, wasting, and micronutrient deficiency disorders 2006. In: Dean T, Jamison DT, Breman JG, et al. Disease control priorities in developing countries, 2nd edn. New York: Oxford University Press, 2006: 551–68.
154. Horton S. The economics of food fortification. J Nutr 2006; 136: 1068–71.
155. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, Grobman WA, Laurberg P, Lazarus JH, Mandel SJ, Peeters RP, Sullivan S, 2017. 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid 27(3):315-389.
156. Buttfield, I.H., and Hetzel, B.S. 1967. Endemic goitre in Eastern New Guinea. With special reference to the use of iodized oil in prophylaxis and treatment. Bull WHO 36:243.
157. Dunn, J.T. 1996. The use of iodized oil and other alternatives for the elimination of Iodine Deficiency Disorders. In S.O.S. for a billion. The conquest of Iodine Deficiency Disorders. B.S. Hetzel, and C.S. Pandav, editors. New Dehli: Oxford University Press publ. 119-128.
158. Wolff, J. 2001. Physiology and pharmacology of iodized oil in goiter prophylaxis. Medicine 80:20-36.
159. Leverge, R., Bergmann, J.F., Simoneau, G., Tillet, Y., and Bonnemain, B. 2003. Bioavailability of oral vs intramuscular iodiated oil (Lipiodol ® UF) in healthy subjects. J Endocrirnolol Invest 26 (Suppl. to n° 2): 20-26.
160. Tonglet, R., Bourdoux, P., Minga, T., and Ermans, A.M. 1992. Efficacy of low oral doses of iodized oil in the control of iodine deficiency in Zaire. New Engl. J. Med. 326:236-241.
161. Delange, F. 1996. Administration of iodized oil during pregnancy : a summary of the published evidence. Bull. WHO 74:101-108.
162. Bouhouch, R.R., Bouhouch, S., Cherkaoui, M., Aboussad, A., Stinca, S., Haldimann, M., Andersson, M., Zimmermann, M.B. 2014. Direct iodine supplementation of infants versus supplementation of their breastfeeding mothers: a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2(3) : 197-209.
163. The control of Vitamin A deficiency and Iodine Deficiency Disorders. EPI report to EPI Global Advisory Group Meeting. Washington 9-13 Nov.
164. Clements, W., Gibson, H.B., and Coy, J.F. 1970. Goiter prophylaxis by addition of potassium iodate to bread. Lancet i:489-501.
165. Gerasimov, G., Shishkina, A., Mayorova, N., Sviridenko, N., Nzarou, A., Alexandrova, G., Kotova, G., Butrova, S., Arbuzova, M., Mishchenko, B., et al. 1997. Bread iodization for iodine deficient regions of Russia and other Newly Independent States. IDD Newsletter 13:12-13.
166. Anonymous. 1997. Iodized water to eliminate iodine deficiency. IDD Newsletter 13:33-39.
167. Fisch, A., Pichard, E., Prazuck, T., Sebbag, R., Torres, G., Gernez, G., and Gentilini, M.1993. A new approach to combatting iodine deficiency in developing countries : the controlled release of iodine in water by a silicone elastomer. Am J Publ Health 83 : 540-545.
168. Foo, L.C., Zainab, T., Goh, S.Y., Letchuman, G.R., Nafikudin, M., Doraisingam, P., and Khalid, B.K. 1996. Iodization of village water supply in the control of endemic iodine deficiency in rural Sarawak, Malaysia. Biomed Environ Sci 9 : 236-241.
169. Squatrito, S., Vigneri, R., Runello, F., Ermans, A.M., Polley, R.D., and Ingbar, S.H. 1986.Prevention and treatment of endemic iodine deficiency goiter by iodization of a municipal water supply. J Clin Endocr Metab 63 : 368-375.
170. Maberly, G.F., Eastman, C.J., and Corcoran, J.M. 1981. Effect of iodination of a village water-supply on goitre size and thyroid function. Lancet ii:1270-1272.
171. Cao, X.Y., Jiang, X.M., Kareem, A., Dou, Z.H., Rakeman, M.A., Zhang, M.L., Ma, T., O’Donnel, K., Delong, N., and Delong, G.R. 1994. Iodination of irrigation water as a method of supplying iodine to a severely iodine-deficient population in Xinjiang, China. Lancet 344 : 107-110.
172. Pearce, E.N., Andersson, M., Zimmermann, M.B. 2013. Global iodine nutrition: Where do we stand in 2013? Thyroid. 23(5) : 523-8.
173. Zimmermann, M.B., Andersson, M. 2012. Assessment of iodine nutrition in populations: past, present, and future. Nutr Rev. 70(10) : 553-70.
174. Iodine Global Network Global Scorecard of Iodine Status. http://www.ign.org/cm_data/Scorecard_2016_SAC_PW.pdf
175. UNICEF. The State of the World's Children 2015, UNICEF, NY. https://www.unicef.org/publications/files/SOWC_2015_Summary_and_Tables.pdf
176. UNICEF. The State of the World's Children 2016, UNICEF, NY. https://www.unicef.org/publications/files/UNICEF_SOWC_2016.pdf
177. Walker SP, Wachs TD, Gardner JM, et al; International Child Development Steering Group. Child development: risk factors for adverse outcomes in developing countries. Lancet 2007; 369: 145–57.
178. Dunn JT. Complacency: the most dangerous enemy in the war against iodine defi ciency. Thyroid 2000; 10: 681–83.
179. Zimmermann MB, Wegmüller R, Zeder C, Torresani T, Chaouki N. Rapid relapse of thyroid dysfunction and goiter in school-age children after discontinuation of salt iodization. Am J Clin Nutr 2004; 79: 642–45.
180. WHO. The 58th World Health Assembly WHA58.24: Sustaining the elimination of iodine deficiency disorders. http://www.who.int/gb/ebwha/pdf_fi les/WHA58/WHA58_24-en.pdf (accessed Nov 18, 2007).
181. The United Nations. The Millennium Development Goals (MDGs). http://www.un.org/millenniumgoals/ (accessed Nov 18, 2007).
182. McGuire J, Galloway R. Enriching lives. Overcoming vitamin and mineral malnutrition in developing countries. Washington, DC: World Bank, 1994.
183. WHO. 1994. Iodine and health. Eliminating iodine deficiency disorders safely through salt iodization. Geneva: WHO publ. 1-7 pp.
184. Knudsen, N., Bülow, I., Jorgensen, T., Laurberg, P., Ovesen, L., and Perrild, H. 2000. Comparative study of thyroid function and types of thyroid dysfunction in two areas in Denmark with slightly different iodine status. Eur. J. Endocrinol. 143:485-491.
185. Roti, E., and Vagenakis, A.G. 2000. Effect of excess iodide : clinical aspects. In The thyroid. A clinical and fundamental text. L.E. Braverman, and R.D. Utiger, editors. Philadelphia: J.B. Lippincott, Williams and Wilkins publ. 316-329.
186. Zimmermann, M.B., Ito, Y., Hess, S.Y., Fujieda, K., and Molinari, L. 2005. High thyroid volume in children with excess dietary iodine intakes. Am. J. Clin. Nutr. 81:840-844.
187. Stanbury, J.B., Ermans, A.E., Bourdoux, P., Todd, C., Oken, E., Tonglet, R., Vidor, G., Braverman, L.E., and Medeiros-Neto, G. 1998. Iodine-induced hyperthyroidism : occurrence and epidemiology. Thyroid 8:83-100.
188. Azizi, F., Hedayati, M., Rahmani, M., Sheikholeslam, R ;, Allahverdian, S.and Salarkia, N. Reappraisal of the risk of iodine-induced hyperthyroidism : an epidemiological population survey. J. Endocrinol. Invest. 28 : 23-29, 2005.
189. Connolly, R.J., Vidor, G.I., and Stewart, J.C. 1970. Increase in thyrotoxicosis in endemic goitre area after iodation of bread. Lancet i:500-502.
190. Todd, C.H., Allain, T., Gomo, Z.A.R., Hasler, J.A., Ndiweni, M., and Oken, E. 1995. Increase in thyrotoxicosis associated with iodine supplements in Zimbabwe. Lancet 346:1563-1564.
191. Bourdoux, P., Ermans, A.M., Mukalay, A.M.W., Filleti, S., and Vigneri, R. 1996. Iodine induced thyrotoxicosis in Kivu, Zaire. Lancet 347:552-553.
192. Delange, F., de Benoist, B., Alnwick, D., and al., a. 1999. Risks of Iodine-Induced Hyperthyroidism following correction of iodine deficiency by iodized salt. Thyroid 9:545-556.
193. Baltisberger, B.L., Minder, C.E., and Burgi, H. 1995. Decrease of incidence of toxic nodular goiter in a region of Switzerland after full correction of mild iodine deficiency. Eur. J. Endocrinol. 132:546-549.
194. Dremier, S., Coppée, F., Delange, F., Vassart, G., Dumont, J.E., and Sande, J.V. 1996. Thyroid autonomy : mechanism and clinical effects. J. Clin. Endocrinol. Metab. 81:4187-4193.
195. Jonckheer, M.H., Velkeniers, B., and Van Haelst, L. 1992. Further characterization of iodine-induced hyperthyroidism based on the direct measurement of intrathyroidal iodine stores. Nucl. Med. Commun. 13:114-118.
196. Delange, F., and Lecomte, P. 2000. Iodine supplementation : benefits outweigh risks. Drug Safety 22:89-95.
197. McConahey, W.M., Keating, F.R., Beahr, O.H., and Woolner, L.B. 1962. On the increasing occurrence of Hashimoto's thyroiditis. J. Clin. Endocrinol. Metab. 22:542-544.
198. Boukis, M.A., Koutras, D.A., Souvatzoglou, A., Evangelopoulou, E., Vrontakis, M., and Moulopoulos, S.D. 1983. Thyroid hormone and immunological studies in endemic goiter. J Clin Endocr Metab 57:859-862.
199. Kahaly, G.J., Dienes, H.P., Beyer, J., and Hommel, G. 1998. Iodide induces thyroid autoimmunity in patients with endemic goiter : a randomised, double-blind, placebo-controlled trial. Eur. J. Endocrinol. 139:290-297.
200. Hollowell, J.G., Staehling, N.W., Flanders, W.D., Hannon, W.H., Gunter, E.W., Spencer, C.A. and Braverman, L.E. 2002. Serum TSH, T4 and thyroid antibodies in the United States population (1988 to 1994) : national health and nutrition examination survey (NHANES III). J. Clin. Endocrinol. Metab. 87:489-499.
201. Braverman, L.E. 1994. Iodine and the thyroid : 33 years of study. Thyroid 4:351-356.
202. Pearce, E.N., Gerber, A.R., Gootnick, D.B., Khan, L.K., Li, R., Pino, S. and Braverman, L.E. 2002. Effects of chronic iodine excess in a cohort of long-term American workers in West Africa. J. Clin. Endocrinol. Metab. 87:5499-5502.
203. Yang F, Shan Z, Teng X, et al. Chronic iodine excess does not increase the incidence of hyperthyroidism: a prospective community-based epidemiological survey in China. Eur J Endocrinol 2007; 156: 403–08.
204. Teng W, Shan Z, Teng X, et al. Effect of iodine intake on thyroid diseases in China. N Engl J Med 2006; 354:2783–93.
205. Pedersen IB, Laurberg P, Knudsen N, et al. An increased incidence of overt hypothyroidism after iodine fortification of salt in Denmark: a prospective population study. J Clin Endocrinol Metab 2007; 92:3122–27.
206. Pedersen IB, Laurberg P, Knudsen N, et al. Increase in incidence of hyperthyroidism predominantly occurs in young people after iodine fortification of salt in Denmark. J Clin Endocrinol Metab 2006; 91: 3830–34.
207. Money, W.L., and Rawson, R.W. 1950. The experimental production of thyroid tumors in the rat exposed to prolonged treatment with thiouracil. Cancer 3:321-.
208. Riccabona, G. 1980. Treatment of the individual patient with endemic goiter. In Endemic goiter and endemic cretinism. J.B. Stanbury, and B.S. Hetzel, editors. New York: J. Wiley publ. 351-394.
209. Pendergrast, W.J., Milmore, B.K., and Marcus, S.C. 1961. Thyroid cancer and thyrotoxicosis in the United States : their relation to endemic goiter. J. Chronic Dis. 13:22-38.
210. Harach, H.R., Escalante, D.A., Onativia, A., Outes, J.L., Day, E.S., and Williams, E.D. 1985. Thyroid carcinoma and thyroiditis in an endemic goitre region before and after iodine prophylaxis. Acta Endocrinol. (Kbh) 108:55-60.
211. Vigneri, R., Pezzino, V., Squatrito, S., Salamone, S., Giuffrida, D., Rosa, G.L.L., Regalbuto, C., and Belfiore, A. 1998. Iodine deficiency and thyroid cancer. In Elimination of Iodine Deficiency Disorders (IDD) in Central and Eastern Europe, the Commonwealth of Independent States, and the Baltic States. F. Delange, A. Robertson, E. McLoughney, and G. Gerasimov, editors. Geneva: WHO publ. 67-72.
212. Williams, E.D. 1985. Dietary iodide and thyroid cancer. In Thyroid disorders associated with iodine deficiency and excess. R. Hall, and J. Köbberling, editors: Raven Press publ. 201-207.
213. Zimmermann MB, Galetti V. 2015. Iodine intake as a risk factor for thyroid cancer: a comprehensive review of animal and human studies. Thyroid Res. 18 : 8.
214. Braverman, L.E. 1998. Adequate iodine intake-the good far outweighs the bad. Eur. J. Endocrinol. 139:14-15.
215. Delange, F. 1998. Risks and benefits of iodine supplementation. Lancet 351:923-924.
216. Li,M., Eastman,C.J. 2012. The changing epidemiology of iodine deficiency.Nat Rev Endocrinol. 8:434-440

Molecular links between Obesity and Diabetes: “Diabesity”

ABSTRACT

 

Severe obesity represents a major risk factor for the development of type 2 diabetes mellitus (T2DM). Due to the strong association of obesity and diabetes, the term “diabesity” was coined, suggesting a causal pathophysiological link between both phenomena. The majority of individuals with T2DM are obese, highlighting the pivotal role of increased adiposity as a risk factor for diabetes. However, only a relatively small fraction of obese individuals will develop T2DM. On a population level, the link between obesity and its secondary complications is well described. However, the molecular mechanisms underlying these complications are still poorly understood. Three main hypotheses have been developed in recent years to bridge the gap between epidemiology and pathobiochemistry: (1) The “inflammation hypothesis” asserts that obesity represents a state of chronic inflammation where inflammatory molecules produced by infiltrating macrophages in adipose tissue exert pathological changes in insulin-sensitive tissues and β-cells. (2) The “lipid overflow hypothesis” predicts that obesity may result in increased ectopic lipid stores due to the limited capacity of adipose tissue to properly store fat in obese subjects. Potentially harmful lipid components and metabolites may exert cytotoxic effects on peripheral cells. (3) The “adipokine hypothesis” refers to the principal feature of white adipose cells to function as an endocrine organ, and to secrete a variety of hormones with auto- and paracrine function. Expanding fat stores can cause dysfunctional secretion of such endocrine factors, thereby resulting in metabolic impairment of insulin target tissues and eventually failure of insulin producing β-cells. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

 

INTRODUCTION

 

Severe obesity represents a major risk factor for the development of type 2 diabetes mellitus (T2DM), a disease characterized by insulin resistance, insulin hyposecretion and hyperglycaemia.1-3  According to statistics, already 415 million people worldwide were affected by diabetes in 2015 (estimated) and this number is expected to rise to 642 million by 2040.4 Due to the strong association of obesity and diabetes, the term “diabesity” was coined, suggesting a causal pathophysiological link between both phenomena.5,6

 

SIZE MATTERS (NOT ONLY)!

 

The majority (~80%) of individuals with T2DM are obese, highlighting the pivotal role of increased adiposity as a risk factor for diabetes. However, only a relatively small fraction of obese individuals develops T2DM.7 In fact, most obese, insulin-resistant individuals do not develop hyperglycemia, indicating that their pancreatic β-cells still produce and secrete sufficient amounts of insulin in order to compensate for the reduced efficiency of insulin action in the periphery.1,8,9 Thus, in addition to an increased adipose mass, additional factors are likely to determine the risk for β-cell dysfunction and the susceptibility for β-cell destruction and diabetes. Nevertheless, despite recent advances in the understanding of body weight regulation and insulin action, the risk factors that determine which obese, non-diabetic individuals will eventually develop diabetes still remain unknown.

 

ROLE OF FAT DISTRIBUTION

 

Obesity results from a period of a positive energy balance during which adipocytes store excess triglycerides, resulting in cell hypertrophy and hyperplasia. However, fat depots do not expand uniformly as they accumulate lipids, and the adverse effects of excess fat storage have been frequently attributed to intra-abdominal (i.e. visceral) fat tissue.  Using a variety of measures (oral glucose tolerance test, intravenous glucose tolerance test, euglycemic hyperinsulinemic clamps), selective excess of visceral adipose tissue (visceral adiposity) has been linked to insulin resistance in humans.10-19 Interestingly, no relationship between visceral fat and impaired glucose metabolism has been observed in studies with non-obese individuals.20,21 On the other hand, other studies found that abdominal subcutaneous fat correlates with insulin sensitivity as well as visceral fat in euglycemic clamps, thus challenging a unique role for the visceral fat depot in modulating insulin sensitivity.22,23 However, in another study, Klein and coworkers reported that large-volume abdominal liposuction of subcutaneous fat did not improve insulin sensitivity of liver, skeletal muscle, and adipose tissue (as assessed by euglycemic-hyperinsulinemic clamps), at least not within 12 weeks post surgery.24 In accordance to these results, removal of visceral fat has been found to improve insulin sensitivity in humans,25,26 supportive of a causal role of intra-abdominal fat for the insulin resistance in obese individuals. However, the relationship between the amount of visceral fat and insulin sensitivity has been controversially discussed throughout recent years and a number of clinical studies show that surgical removal of this fat depot (e.g. via omentectomy) did not lead to improved whole-body glycemia or even BMI of the patients.27-30 Interestingly, a number of adipose-tissue related sub-phenotypes have been identified, one of these the so-called “TOFI” (“thin on the outside, fat on the inside”) subjects. These patients present a normal BMI (< 25 kg/m2) but increased abdominal obesity and therefore exhibit an increased risk to develop insulin resistance and T2DM.31,32 In contrast to TOFI, the so-called “fat-fit” subjects show no gross impairments of glucose metabolism despite their elevated body adiposity (BMI ≥ 30 kg/m2).32,33

 

Visceral fat is defined as adipose tissue located inside the peritoneal cavity—within the parietal peritoneum and transversalis fascia, excluding the spine and paraspinal muscles. As such, appropriate techniques for precise measurements of visceral fat, however, have been controversial. In humans, the amount of abdominal visceral fat is determined by a number of different techniques, including anthropomorphic measurements (waist-hip-ratio, waist circumference, abdominal sagittal diameter), computed tomography (CT), magnetic resonance imaging (MRI), and ultrasound. Comparative measurements using CT and MRI have revealed a fairly high correlation between both methods.34-36 In the recent years, Dual-energy X-ray absorptiometry (DXA) has emerged as reliable technique to measure body composition with high-precision, low X-ray exposure, and short-scanning time.37,38 In studies that included these imaging techniques and common clinical measurements, the abdominal sagittal diameter was found to be the most specific predictor of visceral adipose volume, but measurements of waist circumference and sagittal diameter are also highly correlated. Even though waist circumference varies considerably between sexes and among different ethnic groups, it has been proposed as a crude, but efficient, anthropomorphic readout for abdominal adiposity.39,40 And lastly, the amount of visceral fat is correlated to total body fat even though considerable variation in individual fat distribution has been reported,41-44 Thus, despite the lack of a ”gold standard” for the quantitative assessment of regional fat distribution in humans, most of the evidence suggests a specific association between visceral fat with an increased risk for insulin resistance and diabetes.

 

TO BE (FAT) OR NOT TO BE

 

Interestingly, deficiency of fat tissue (lipodystrophy) predisposes to similar metabolic complications as an excess of fat in obesity, such as insulin resistance, T2DM, and hepatic steatosis (reviewed in Refs 45 and 4645,46). Moreover, fat transplantation into lipodystrophic mice ameliorated the diabetic phenotype of the animals either partially or completely, implicating that the failure to properly store lipids in depots is causal for lipodystrophic diabetes.47 Also in normal lean animals, fat transplantation has been shown to result in beneficial metabolic effects.48,49 Thus, adipose tissue may have both beneficial and adverse effects on whole-body metabolism, depending on where it accumulates.

 

OBESITY-INDUCED DIABETES: FACTORS AND MECHANISMS

 

On a population level, the link between obesity and its secondary complications is well described. However, the molecular mechanisms underlying these complications are still poorly understood.50 Even though the evidence indicates a detrimental role of visceral fat in terms of insulin sensitivity, relatively little is known about distinct physiological and biochemical properties of fat tissue derived from different anatomic locations. On the one hand, transplantation experiments with fat tissue from different adipose depots in mice have not conclusively revealed intrinsic differences between subcutaneous and visceral fat.47,49 On the other hand, factors that have been attributed to confer differential metabolic effects of subcutaneous versus visceral fat include increased portal release of FFA and glycerol from omental/mesenteric fat directly to the liver, and also differences in endocrine and metabolic functions of fat depots.

 

Three main hypotheses have been developed in recent years to bridge the gap between epidemiology and pathobiochemistry:

 

(1) The “inflammation hypothesis” asserts that obesity represents a state of chronic inflammation where inflammatory molecules produced by infiltrating macrophages in adipose tissue exert pathological changes in insulin-sensitive tissues and β-cells.

 

(2) The “lipid overflow hypothesis”, also known as “Adipose Tissue Expandability Hypothesis” predicts that obesity may result in increased ‘ectopic’ lipid stores (lipid that accumulates outside the normal depots, such as in the organ tissue of the liver, muscle, and pancreas) due to the limited capacity of adipose tissue to properly store fat in obese subjects. Potentially harmful lipid components and metabolites may exert cytotoxic effects on peripheral cells, including liver and β-cells, thereby impairing function, survival, and regeneration.

 

(3) The “adipokine hypothesis” refers to the principal feature of white adipose cells to function as an endocrine organ, and to secrete a variety of hormones with auto- and paracrine function. It has been proposed that expanding fat stores in obesity cause dysfunctional secretion of such endocrine factors, thereby resulting in metabolic impairment of insulin target tissues and eventually failure of insulin producing β-cells.

 

In the following, these three hypotheses are briefly discussed.

 

The “Inflammation Hypothesis”

 

Inflammatory processes are thought to play a key role in the development of obesity-related insulin resistance and type 2-diabetes. In adiposity there are fundamental changes in adipose tissue secretory functions51. An excess of adipose tissue produces a number of pro-inflammatory cytokines leading to a state of chronic subclinical inflammation associated with both insulin resistance and type-2 diabetes.52 The term “metaflammation” describes this low-grade, chronic inflammation orchestrated by metabolic cells in response to excess nutrients and energy.53

 

How does a spill-over of these inflammatory products into circulation finally lead to insulin resistance? Weisberg et al54 described that macrophages accumulate in adipose tissue of obese subjects and suggested that these macrophages are derived from the circulation. Further studies indicated that adipose tissue macrophages (ATMs) that accumulate during diet-induced obesity (DIO) are not only an important source of adipose tissue inflammation but also mediate insulin resistance in adipocytes.55 The amount of macrophages in adipose tissue correlates positively with two indices of adiposity: Body mass index (BMI) and adipocyte size. The exact mechanisms underlying ATM recruitment and activation are still not fully understood. One factor potentially responsible for obesity-induced inflammation by increasing ATM recruitment is Macrophage Migration Inhibitory Factor (MIF), a chemokine-like inflammatory regulator directly associated with the degree of peripheral insulin resistance.56 Adipose tissue macrophages are considered to be a major reservoir of pro-inflammatory molecules in adipose tissue54. These cytokines exert various functions in the pathogenesis of the disease progression (Figure 1). Some of the most important inflammatory factors are described below.

Figure 1. The “inflammation hypothesis.” Pathophysiology of obesity-induced chronic inflammation and peripheral insulin resistance.

DAG, diacylglycerol; IL-1, interleukine-1; MCP-1, monocyte chemotactic protein-1; TNF, tumor necrosis factor alpha; Toll-like receptor 4, TLR-4; Details in the text.

 

Tumor Necrosis Factor Alpha:

Tumor necrosis factor alpha (TNF-α) is a pluripotent cytokine primarily produced from macrophages.57 Its expression was shown to be elevated in different mouse and rat models of obesity and diabetes.58 In vitro, TNF-α suppresses the expression of most adipose-specific genes in murine adipocytes, including the enzymes involved in lipogenesis.59 It was also shown that TNF-α induces insulin resistance, in part through its ability to inhibit intracellular signaling from the insulin receptor.60 Moreover, addition of TNF-α to cells in vivo increased the intracellular concentration of ceramides.61 Ceramides can directly induce DNA fragmentation and apoptosis. In skeletal muscle, diacylglycerols and ceramides operate as lipotoxic mediators engaging serine kinases that disrupt the insulin signaling cascade and diminish insulin sensitivity.62 Further, it was discovered that ceramides are able to induce lipoapoptosis in β-cells.63 In addition, TNF-α was shown to induce the formation of reactive oxygen species (ROS).64 Production of ROS increased selectively in adipose tissue of obese mice, causing dysregulated production of adipocytokines (fat-derived hormones), including adiponectin, plasminogen activator inhibitor-1, interleukin-6 (IL-6), and monocyte chemotactic protein-1.65 However, clinical studies with Etanercept, a neutralizing protein for circulating TNF-α failed to demonstrate an improvement of insulin sensitivity in humans,66,67 indicating that acute reduction of systemic TNF-α may not be sufficient to induce metabolic benefits in the periphery.

 

TNF-Like Weak Inducer of Apoptosis:

TNF-like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and was shown to have pro-inflammatory action in adipocytes mediated by the nuclear factor-κB (NFκB) and ERK but not JNK signaling pathways.68 The cytokine promotes the secretion of MCP-1 and RANTES and up-regulates CCl21 and CCL19 expression. Whereas expression levels of membrane-bound TWEAK (mTWEAK) and its receptor Fn14 are increased during obesity, the amount of soluble TWEAK (sTWEAK) was decreased, thereby enhancing the pro-inflammatory activity elicited by TNF-α.69,70

 

Monocyte Chemotactic Protein-1:

The pro-inflammatory chemokine monocyte chemotactic protein-1 (MCP-1) attracts leukocytes to inflamed sites and is regulated by NFκB. 71 Monocyte chemotactic protein-1 represents the first discovered and most extensively studied human CC chemokine and is also known as CCL2 (Chemokine (C-C motif) ligand 2). CC chemokines are characterized by the conserved position of four cysteine residues responsible for protein stabilization.56 Insulin was found to induce expression and secretion of MCP-1 substantially both in vitro in insulin-resistant adipocytes and in vivo in insulin-resistant obese mice (ob/ob). It was suggested that elevated MCP-1 levels may induce adipocyte dedifferentiation and contribute to pathologic states associated with hyperinsulinemia and obesity, including type 2 diabetes.72 Expression and plasma concentration of MCP-1, however, were shown to be increased both in genetically obese diabetic (db/db) mice and in wildtype mice with high-fat diet-induced obesity, leading to the assumption that  increased MCP-1 expression contributes to the macrophage infiltration of adipose tissue and, finally, to the development of insulin resistance.73 Monocyte chemotactic protein-1 has been developed into one of the most important targets for a variety of therapeutic approaches to improve diabetic vascular conditions over the years.74

 

Interleukin-6:

The role of the cytokine interleukin-6 (IL-6) in the regulation of lipid metabolism is controversial.75 If produced in large amounts by adipose tissue, IL-6 causes insulin resistance in adipocytes and skeletal muscle.76 Contrary to the expectations, IL-6-deficient mice develop obesity. However, excess body weight was only reported in very mature animals.77 Interestingly, chronic exposure of IL-6 produces insulin resistance in skeletal muscle, whereas short-term exposure as consequence of exercise has beneficial effects on insulin sensitivity.78 Thus, despite the evidence of IL-6 as a major player in the regulation of metabolism, the role of this cytokine in the pathogenesis of insulin resistance and diabetes remains incompletely understood.

 

Interleukin-1:

Interleukin-1 (IL-1) is a cytokine that is also secreted by stimulated macrophages and has many actions that overlap those of TNF-α. For instance, IL-1 increases hepatic triglyceride secretion and serum triglyceride levels.79 Common polymorphisms of the IL-1 gene that influence IL-1 activity are also associated with fat mass in humans.80 Pro-inflammatory pathways in adipose tissue have been shown to be directly activated by free-fatty acids (FFA). In turn, the inflammatory status of macrophages is linked to body fat content. In lean mice, macrophages in WAT are in their active M2 state and produce immunosuppressive factors. However, in obese mice, macrophages are in a pro-inflammatory M1 state (F4/80+, CD11b+, CD11c+), highly responsive to the pro-inflammatory effect of FFA that bind the Toll-Like Receptors (TLRs).81 Increased cytokine release via TLRs as a consequence of FFA binding was proposed as potential pathomechanism causing insulin resistance.82 Interestingly, in a clinical study, blockade of IL-1 receptor with Anakinra, a recombinant IL-1 receptor antagonist, improved HbA1c levels and proinsulin-to-insulin ratio but had no effect on systemic insulin sensitivity.83

 

Toll-Like Receptor-4:

Toll-like receptors are membrane-spanning, non-catalytic receptors that respond to different microbial antigens, therefore representing an important factor of the innate immunity.84 Toll-like Receptor-4 (TLR-4) is thought to be another important factor in fatty acid-induced insulin resistance. Scherer and coworkers were the first ones that found it expressed on 3T3-L1 adipocytes and activated by lipopolysaccharides (LPS).85 Characterization of TLR-4 as the main endogenous sensor for LPS in adipocytes supports the relevance of fat tissue in immune processes.86 Additionally, TLR-4 was recently shown to be directly activated by dietary saturated fatty acids, thereby promoting inflammatory aspects of the metabolic syndrome and atherosclerosis.87 In addition, stimulation of TLR-4 with activation of the Erk pathway was shown to upregulate IL-6 as well as MCP-1 release in adipose tissue. Therefore, it can be suggested that activation of TLR-4 in adipocytes induces inflammation and, as a consequence, promotes the progression towards diabetes. This mechanism provides new evidence for a coupling of visceral adipose dysfunction with the development insulin resistance and T2DM.88

 

Summary:

Inflammation is thought to be a major factor in the development of insulin resistance and diabetes. Increased secretion of adipocyte-derived inflammatory cytokines and fatty acids are directly linked to impaired insulin sensitivity in obesity.89 However, inflammatory processes do not account exclusively for the development of insulin resistance since there are studies showing subjects with T2D but without any alterations in inflammatory markers.90 Inflammation alone can therefore not explain how obesity affects insulin sensitivity and certainly not why only a small fraction of obese individuals develop T2DM.

 

The “Lipid Overflow Hypothesis”

 

Healthy adipose tissue is characterized by the ability to expand passively to accommodate periods of nutrient excess. In contrast, adipose tissue in polygenic mouse models of obesity-induced diabetes, as well as in obese humans, may fail to fully accommodate excessive nutrient loads.91-93 If the adipose tissue expansion limit is reached, lipids can no longer be stored appropriately in adipose tissue and consequently “overflow” to other peripheral tissues such as skeletal muscle, liver, and pancreas.94,95 Subcutaneous adipose tissue (SAT) represents the largest adipose tissue depot and, in addition, is considered the least metabolically harmful site for lipid storage. The SAT can expand either by increasing the size of the cells (hypertrophic obesity) and/or by recruiting new cells (hyperplastic obesity). In contrast to the hyperplastic response, which seems to be protective against SAT dysfunction, hypertrophic obesity is associated with increased T2D risk.96,97 The storage of this ectopic fat in non-SAT tissues is directly linked to the progression of insulin resistance and type-2-diabetes.98,99 Thus, fat accumulates in tissues that are not adequate for lipid storage, and as a consequence, lipid metabolites might accumulate within those tissues that inhibit insulin signal transduction (Figure 2).

Figure 2: The “lipid overflow hypothesis.” Pathophysiology of obesity-induced ectopic lipid stores that cause peripheral insulin resistance and impaired β-cell function.

 

 

CPT-1, carnitine palmitoyltransferase 1; DAG, diacylglycerol; GLUT2, facilitated glucose transporter, member 2 (SLC2A2); HAD, β-hydroxyacyl dehydrogenase; IRS1, insulin receptor substrate 1; MafA, pancreatic beta-cell-specific transcriptional activator MafA; PKC, Protein kinase C; Details in the text.

 

This hypothesis is supported by several rodent models of lipodystrophy. These animals are extremely lean but often suffer from marked insulin resistance, diabetes, hypertriglyceridemia, hepatosteatosis, and low HDL (high-density lipoprotein)-cholesterol levels – a metabolic profile similar to that observed in obesity-related metabolic syndrome.100,101 Moreover, recent studies demonstrate a lipodystrophy-like phenotype also in the general human population since subjects who are of normal weight but metabolically unhealthy (∼20% of the normal weight adult population) have a greater than 3-fold higher risk of all-cause mortality and/or cardiovascular events.46 Leptin replacement in patients with generalized lipodystrophy can serve as efficient therapy to improve insulin sensitivity by reducing ectopic fat accumulation, especially in the liver.102-104

 

Thus, an increased fatty acid flux from normal fat depots towards non-adipose tissues (NAT), e.g. skeletal muscle, heart, liver, and pancreatic β-cells appears to be a critical factor in mediating lipotoxicity. Among the substances known to impair insulin signaling, the most prominent examples include diacylglycerols (DAGs) and ceramides, which have both been shown to impair insulin action in a number of peripheral tissues.105-107

 

Lipotoxicity- Skeletal Muscle and Adipocytes:

Skeletal muscle insulin resistance is associated with high levels of stored lipids in skeletal muscle cells.108 A high lipid accumulation and/or lower triglyceride turnover can induce lipotoxicity within the skeletal muscle cell.109 Lipid infusion can induce peripheral and hepatic insulin resistance in rats and humans.110,111 There are multiple regulatory sites controlling the complex process of fatty acid (FA) metabolism in skeletal muscle. Long-chain FA (LCFA) oxidation involves lipolysis and LCFA release from the adipose tissue, delivery of FFA to the skeletal muscle, transport across the plasma membrane, lipolysis of intramuscular triacylglycerol (IMTG), activation with addition of a coenzyme A thioester (LCFA-CoA), transport across the mitochondrial membranes and ultimately oxidation.112 Obese individuals display a disturbed lipid oxidation in skeletal muscle. This leads to accumulation of fatty acids and therefore to enhanced levels of triglycerides, fatty acyl CoA, diacylglycerols, and ceramides.113-115 Accumulation of these metabolites may be able to impair insulin signaling through different mechanisms, such as increased serine phosphorylation of the insulin receptor and insulin receptor substrate 1 by Protein kinase C (PKC) β and reduced serine phosphorylation of AKT.116,117 Besides disturbances in the insulin signaling cascade, several other factors could be involved in the direction of LCFA or LCA-CoA towards esterification rather than oxidation in obesity and type-2 diabetes. It has long been debated whether reduced mitochondrial function is the cause of, or secondary to, insulin resistance and T2D. Numerous studies, however, have shown that the activity of the key enzymes of fatty acid oxidation, citrate synthase (CS) and β-hydroxyacyl dehydrogenase (HAD) are significantly reduced in skeletal muscle in obesity and type 2 diabetes.118-121 Additionally, it has also been shown that the activity of carnitine palmitoyltransferase 1 (CPT1) in muscle was also reduced in association with obesity,122 and that mitochondrial oxidative capacity is low in insulin-resistant subjects.123 Carnitine palmitoyltransferase 1 converts acyl-CoA molecules to their acyl carnitine derivatives prior transport of the mitochondrial inner membrane.124

 

Plasma non-esterified free fatty acids (NEFAs) are suggested to contribute to the development of insulin resistance, since they have been shown to activate the inflammatory nuclear factor kappa-B (NFκB) pathway in human muscle biopsies.125,126 In humans, it was demonstrated that free fatty acids induce insulin resistance by inhibition of glucose transport.127 In addition to the negative impact on insulin sensitivity, there is very recent evidence that lipid droplet (LD) formation is also impaired by an overflow of lipids. It has been shown that LD formation requires some of the same components of the machinery involved in regulated fusion of vesicles including the two soluble N-ethylmaleimide-sensitive-factor attachment protein receptor (SNARE) proteins SNAP23 and syntaxin-5. SNAP23 has been shown to be an essential factor for trafficking of GLUT4-containing vesicles to the plasma membrane, and a more recent study found that SNAP23 is also involved in LD formation in adipocytes.128 Interestingly, the study reported that excessive LD formation inhibited GLUT4 translocation by competing for SNAP23 and that overexpression of Snap23 in these cells restored insulin sensitivity. Thus, SNAP23 might constitute a link between glucose and lipid metabolism, respectively.

 

Lipotoxicity- Pancreatic Beta Cell:

The development of type 2 diabetes is caused by a combination of insulin resistance and impaired pancreatic β-cell secretion.129 With progression from euglycemia to type 2 diabetes, β-cells progressively fail to compensate for the increase insulin demand in peripheral tissues. The pathogenesis is thereby characterized by different stages, leading from compensatory insulin resistance to decompensated hyperglycemia.130 In manifest type 2 diabetes, β-cells are exposed to both high doses of glucose (glucotoxicity) and lipids (lipotoxicity), respectively.131 Lipotoxicity, manifests as incorporation of large amounts of triglycerides in pancreatic islets, leading to β-cell death.132

 

While rodents are often preferred models to study disease progression, polygenic mouse models more closely resemble human physiology and preferred over the monogenic models such as in defective leptin signaling (db, ob).133,134 New Zealand Obese (NZO) mice develop a polygenic disease pattern of obesity, insulin resistance, and type 2 diabetes.134,135 The onset of hyperglycemia is characterized by an elevated proliferation rate and hypertrophy of the β-cells136 leading to β-cell failure in most of the male animals.137 The disease progression is characterized by a gradual loss of glucose transporter 2138 and the transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene family, protein A (avian) (MafA).139 Interestingly, NZO mice fed a carbohydrate-free high fat diet become obese and insulin resistant but are protected from β-cell failure.139-141 In contrast mice fed a diet rich in both carbohydrates and fat rapidly develop diabetes, indicating that the additive toxicity of an overflow of carbohydrates and lipids is important for the progression of β-cell failure.139,140,142,143

 

Summary:

Inability to store fat (lipodystrophy) or overflow of excess lipids from normal fat depots contributes to “ectopic” deposition of lipids and their metabolites in organs important for glucose metabolism, including muscle, liver, and the pancreas. Numerous studies have demonstrated involvement of these lipid metabolites in the development of insulin resistance and diabetes.  Moreover, recent evidence indicates that hyperglycemia is a critical factor contributing to lipid-induced beta cell failure and diabetes. Thus, many leading scientists consider type 2 diabetes, a disorder with manifestations of abnormal glucose metabolism, to be at its most fundamental molecular level a disorder of lipid metabolism.

 

The “Adipokine Hypothesis”

 

Adipose tissue is not only a storage compartment for triglycerides but also a major endocrine and secretory organ, which releases a wide range of factors (adipokines) that signal through paracrine and hormonal mechanisms.144 Some of these secreted molecules are involved in inflammatory processes, such as TNF-α, IL-1β, IL-6 and MCP-1 as described above. The expanding volume of adipose tissue during obesity raises circulating levels of these inflammatory markers and is therefore thought to contribute to insulin resistance145 and the development of T2DM (Figure 3).

Figure 3: The “adipokine hypothesis.” Pathophysiology of obesity-induced dysfunction of adipokines in adipose cells contributing to peripheral insulin resistance.

 

 

AdipoR2, adiponectin receptor 2; AMPK, AMP-activated protein kinase; PEPCK, phosphoenolpyruvate carboxykinase; RBP4, retinol binding protein 4; Details in the text.

 

More than 100 different factors secreted by adipocytes have been identified over the past years, and it seems likely that this number will increase further due to the progress in analytical chemistry.146 Some of the prominent members of hormones produced by the adipose tissue are described below.

 

Leptin:

Leptin was the first adipokine discovered to influence body fat mass. It is predominantly secreted from white adipose tissue and exerts its main function by repressing food intake and promoting energy expenditure through sites of action in the central nervous system.147 The leptin receptor is expressed in the arcuate, ventromedial, dorsomedial, and lateral hypothalamic nuclei, which are known to regulate food intake.148 Mutation of both the leptin gene (ob) as well as the leptin receptor gene (db) leads to severe obesity, hyperphagia and insulin resistance in mice.149 The ob mutation was first hypothesized in 1950, when animal caretakers of the Jackson Laboratory observed the spontaneous occurrence of an obese phenotype in a mouse.150 but was not described as a non-sense mutation in the leptin gene until more than 40 years later.151

 

Expression and secretion of leptin is correlated with the amount of body fat and adipocyte size.152 Humans with mutations in both alleles of either leptin or the leptin receptor are obese, but these homozygous mutations are extremely rare.153 To the contrary, the vast majority of obese individuals display high plasma leptin levels in proportion to their increased body fat. Consequently, attempts to treat obesity by leptin administration have been mostly unsuccessful due to an apparent leptin resistance of these patients.154 Nevertheless, leptin improves insulin sensitivity by several mechanisms. In the liver and in skeletal muscle, leptin enhances glucose homeostasis by decreasing intracellular lipid accumulation155 and, in skeletal muscle, by direct activation of AMP-activated protein kinase (AMPK)156. In addition, leptin is able to inhibit insulin secretion by both, a direct effect on pancreatic β-cells, and an indirect mechanism via activation of the SNS (sympathetic nervous system) by the CNS (central nervous system).157-160

 

Adiponectin:

Adiponectin represents another important adipokine that has to be considered in the pathogenesis of insulin resistance and type 2 diabetes. Up-regulation of this collagen-like plasma protein secreted by adipocytes or its receptor is known to improve insulin sensitivity and endothelial function.52,161,162 Adiponectin has been closely linked to diseases such as obesity, the metabolic syndrome, type 2 diabetes mellitus, dyslipidemia and essential hypertension through its anti-inflammatory effects.163,164 In obesity and diabetes, adiponectin biosynthesis is impaired, and in vitro studies demonstrate suppression of adiponectin expression by various inflammatory and oxidative stress factors.165,166

 

Adiponectin regulates glucose and lipid metabolism by targeting the liver and skeletal muscle through two transmembrane receptors (AdipoR1 and AdipoR2). While AdipoR1 is most abundant in skeletal muscle, AdipoR2 is predominantly expressed in the liver.167 Improvement of insulin sensitivity is reached through activation of AMPK as well as increased expression of PPARα target genes.168 Adiponectin also has a key role in differentiation of subcutaneous preadipocytes and in the central regulation of energy homeostasis.161,169

 

Resistin:

Resistin expression and secretion differs between humans and rodents. In rodents, resistin is predominantly secreted from mature adipocytes with some weak expression in pancreatic islets and hypothalamus. In contrast, humans express resistin primarily in macrophages where it is thought to be involved in the recruitment of other immune cells, and in the secretion of pro-inflammatory factors.170 Because of these interspecies differences, it may have a less important role in humans during the pathogenesis of insulin resistance and diabetes. However, insulin-resistant mice display increased resistin levels and treatment with a thiazolidinedione, which activates adipocyte PPAR receptors and improves insulin sensitivity, lowers plasma resistin levels.52,171 In addition, some studies describe a role for resistin in the regulation of hepatic glucose production.172

 

Opposed to adiponectin, resistin decreases AMPK phosphorylation in liver, which leads to suppression of fatty acid oxidation and stimulation of glucose production.173 In vitro data from cultured adipocytes demonstrated a decreased insulin-stimulated glucose transport and disturbed adipocyte differentiation after resistin treatment.174,175 In humans, resistin is thought to impair insulin signaling by upregulating expression of the lipid phosphatase PTEN.170

 

Retinol Binding Protein 4:

Retinol binding protein 4 (RBP4) is predominantly expressed in adipose tissue and the liver and was first linked to the pathogenesis of insulin resistance when Abel and coworkers described that RBP4 was highly expressed in adipocytes of insulin resistant GLUT4-knockout mice.176 In addition, injection or overexpression of RBP4 in mice led to impaired insulin sensitivity. On the molecular level, RBP4 was shown to induce hepatic expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) and to inhibit insulin signaling in skeletal muscle.177 Thus, at least in rodents, increased serum RBP4 leads to impaired glucose uptake in skeletal muscle with concomitant increase of hepatic glucose production.178

 

In humans, RBP4 influence on glucose homeostasis is less clear. Retinol binding protein 4 levels are elevated in plasma from obese and diabetic subjects.179 However, in larger groups a definitive correlation between RBP4 and measures of insulin sensitivity could not be demonstrated.180

 

Visfatin:

Visfatin, also known as nicotinamide phosphoribosyltransferase (NAMPT) and pre-B-cell colony enhancing factor 1 (PBEF1), is predominantly expressed in visceral adipose tissue, from which the name visfatin was derived. As an adipokine, the protein had been also found in the bloodstream where it has been shown to exert insulin-like functions. In mice, administration of visfatin was shown to lower blood glucose levels, whereas mice with a mutation in visfatin had increased levels of circulating glucose.181 However, subsequent studies have produced conflicting results regarding the association between visceral fat mass and plasma visfatin in humans.182,183 and the initial study was, in part, retracted.184 Despite these inconsistencies, a positive correlation between visfatin gene expression in visceral adipose tissue and BMI was seen in some human studies, as well as a negative correlation between BMI and visfatin gene expression in subcutaneous fat.182,185 In summary, the provided evidence of a direct link between visfatin action and human type 2 diabetes mellitus is still weak and its role in obesity and insulin resistance remains to be elucidated.52

 

Vaspin:

Visceral adipose tissue-derived serpin or serpinA12 (Vaspin) was originally identified as an adipokine predominantly secreted from visceral adipose tissue. In humans, obesity and T2DM are associated with elevated vaspin serum concentrations and expression levels in adipose tissue, suggesting a compensatory role in response to diminished insulin signaling in obesity. In obese mice, Vaspin administration improves glucose tolerance, insulin sensitivity, and reduces food intake.186,187 The exact cellular mechanisms of Vaspin action have yet to be elucidated, but a recent study demonstrated that Vaspin inhibited TNF-α- and IL-1-mediated activation of NF-κB and its downstream signaling molecules in a concentration-dependent manner and thereby protected endothelial cells from inflammation caused by pro-inflammatory cytokines.188 Moreover, a single-nucleotide polymorphism (rs2236242) was described to be positively associated with type 2 diabetes in 2759 participants in the KORA F3 study bearing an increased risk of diabetes independent of obesity, suggesting a link between vaspin and glucose metabolism.189

 

Omentin-1:

Omentin was described as a novel adipokine which is mainly produced by visceral adipose tissue and exhibits insulin-sensitizing action. Circulating levels of omentin are reduced in the obese state and in patients with T2DM. The beneficial effects of omentin are thought to be caused by a vasodilatation of blood vessels and attenuation of C-reactive protein-induced angiogenesis, potentially via the nuclear factor B signaling pathway, a potent pro-inflammatory signaling pathway.190 In addition, omentin has been shown to block TNFα-induced JNK and NF-κB activation.191 As with adiponectin, circulating levels of omentin are lower in obesity and also inversely correlated with measures of insulin resistance (HOMA-IR) and lower serum omentin concentrations were found in individuals with impaired glucose tolerance and type 2 diabetes compared to healthy individuals.192 However, it has to be elucidated whether the association of circulating omentin levels with the risk of T2DM is independent of BMI or can entirely be explained by obesity. In addition, further studies are needed to distinguish entirely between adiponectin and omentin action and whether omentin also shows a counter-regulatory increase in pro-inflammatory conditions.193

 

Apelin:

The peptide apelin is expressed among several tissues and secreted by adipocytes. Apelin gene expression levels are increased in adipose tissue from mouse models of obesity and hyperinsulinemia. Moreover, obese and hyperinsulinemic patients demonstrate elevated plasma levels of apelin. However, apelin plasma levels depend on several factors, including blood glucose levels and plasma triglyceride concentration.194 Currently, apelin is being considered as a biomarker and drug target, but its role in the development of both obesity and type 2 diabetes needs to be clarified with convincing clinical studies.195,196

 

Cardiotrophin-1:

Cardiotrophin-1 (Ctf1) is expressed in different tissues and secreted as an adipokine. Targeted disruption of cardiotrophin-1 in mice leads to obesity and insulin resistance.197 However, studies in humans have yielded contradictory results regarding cardiotrophin-1 levels and its association with obesity.198 The role of cardiothrophin-1 in the regulation of metabolic circadian rhythms is the focus of current research.199

 

WNT1-Inducible Signaling Pathway Protein-1:

Wnt1-inducible signaling pathway protein-1 (Wisp1) was recently described as a new adipokine. Its expression and secretion are increased in the course of differentiation of human adipocytes. Changes in body weight regulate both expression of Wisp-1 in adipose tissue and plasma levels of secreted Wisp-1.200 Interestingly, Wisp-1 serum levels are elevated in obese patients affected with polycystic ovary syndrome (PCOS) and in patients with gestational diabetes mellitus.201,202

 

Micro RNA (miRNA)- Containing Exosomes:

Micro RNA’s are small, noncoding sequences of RNAs that control a multiplicity of gene expression processes in diverse organs. In adipose tissue, miRNA’s are important regulators of cellular metabolism such as cell differentiation and lipid storage. Expression of miRNAs is altered in patients with obesity and type 2 diabetes.203,204 Interestingly, the action of miRNAs is not restricted to the cells of their original expression. Adipocyte-specific targeted disruption of the miRNA-processing enzyme Dicer in mice decreased the number of circulating exosomal miRNAs. Dicer (-/-) mice manifest glucose intolerance and insulin resistance, presumably mediated via increased fibroblast growth factor 21 (FGF21) plasma levels. There is evidence for a direct effect of circulating miRNAs derived from adipose tissue on FGF21 translation in the liver.205 In addition, a recent study showed that adipose tissue macrophages secrete miRNA-containing exosomes. Transfer of exosomes from obese to lean mice led to increased glucose intolerance and insulin resistance, and vice versa. The authors identified miRNA-155 as a potential target acting via the peroxisome proliferator-activated receptor gamma.206

 

Fatty Acid Esters Of Hydroxy Fatty Acids:

Recently, a novel family of lipids, the so-called fatty acid esters of hydroxy fatty acids (FAHFAs), has been identified. These branched fatty acid esters can be found in a variety of tissues with highest amounts in adipose tissues.  A specific group of FAHFAs, the PAHSAs (Palmitic acid esters of hydroxy-stearic acids), have been shown to have beneficial metabolic effects. Circulating PAHSA levels are reduced in insulin resistant people, and serum levels correlate highly with insulin sensitivity. Moreover, treatment of obese mice with PAHSAs leads to improved glucose tolerance and increased insulin secretion. In adipocytes, PAHSAs signal through the omega-3 fatty acid receptor GPR120 to enhance insulin-stimulated glucose uptake.207 The production of FAHFAs in adipose tissue is tightly linked to the abundance of the insulin-responsive glucose transporter 4 (GLUT4) and the ability of adipocytes to transport glucose into the cell. Increased glucose uptake activates the nuclear transcription factor carbohydrate response element binding protein (CREBP), thereby enhancing lipogenesis and the synthesis of FAHFA’s.208 In addition, PAHSAs have been demonstrated to exert anti-inflammatory effects by repressing macrophage-induced tissue inflammation.209

 

Summary:

Adipocyte-derived factors such as adipokines and cytokines may provide direct links between obesity and the onset and progression of type 2 diabetes. Recent advancements in analytical technologies, in particular mass spectroscopy methods, may lead to further future discoveries of novel adipokines and cytokines that play roles in regulating intra-organ cross talk and metabolism.

 

GENETIC SUSCEPTIBILITY FOR OBESITY AND INSULIN RESISTANCE

 

Genetic Factors

 

Genetics clearly plays an important role in conferring the risk for the development of metabolic diseases. Variant genes determine the individual susceptibility towards known risk factors and may explain why only a fraction of obese individuals develop T2DM whereas the majority of diabetics are obese. In recent genome-wide association studies (GWAS’s), numerous variant genes were identified that predispose to diabetes or obesity.210,211 However, due to the relatively small contribution of the individual single nuclear polymorphisms (SNPs) to the overall disease risk, the predictive value of the gene variants is relatively small, and the pathophysiological relevance of many of these SNPs remains to be clarified. When combined, the genes identified so-far by GWAS explain only 15-20% of the heritable variance of metabolic diseases.212,213 An example of contradictory results of GWAS’s versus functional in vivo data represents the fat mass and obesity associated (FTO) gene. In different GWAS’s, SNPs located in the first intron of the FTO gene were associated with an altered body mass index,214,215 whereas Fto knockout mice develop postnatal growth retardation and exhibit a reduced body length.216 In contrast, no association of FTO was detected for height in humans.214,217 Although new in vivo data exist that reflect the human pathophysiology more precisely, the discrepancies of the GWAS’s and functional approaches remain apparent.218

 

Thus, even though many studies have confirmed FTO and TCF7L2 as two major genes implicated in obesity and diabetes in humans, respectively, GWAS’s have provided only limited mechanistic insights into the pathophysiology of these diseases.214,219-221 Novel approaches combining classical familial linkage analysis methods with whole-genome sequencing (WGS) are currently emerging as an important and powerful analysis method, especially since rare variants, which are not well interrogated by GWAS’s, could be responsible for a substantial proportion of complex human diseases.222,223

 

Perspectives: Positional Cloning to Identify Novel Genes In (and Out) of the Adipocyte

 

Polygenic mouse models have proven to be important tools to investigate molecular mechanisms that link obesity and T2DM. Despite novel advancements in the sequencing technology, the most successful strategy to identify and characterize new risk alleles is represented by a positional cloning approach.224,225 This approach capitalizes on a combination of breeding of multiple recombinant congenic mouse lines and of expression profiling of critical genomic regions that confer the phenotype. Using this approach, nine gene variants were identified as candidates for type 2 diabetes and/ or obesity during the last years. Sorcs1 encodes for a protein of largely unknown function that binds to a transcription factor responsible for islet vascularization.226  Lisch-like factor was described to be responsible for reducing β-cell mass and β-cell replication rates.227 Zfp69, a zinc-finger transcription factor, was described as causal gene for the diabetogenic Nidd1 quantitative trait locus (QTL) derived from the lean SJL (Swiss Jim Lambert) mouse strain and responsible for the distribution of lipids between different organs. Recently, it was shown that Zfp69 modulates hepatic insulin sensitivity in mice.93,228,229 Ifi202b, a member of the Ifi200 family of interferon inducible transcriptional modulators modulates fat accumulation through expression of adipogenic genes such as 11β-HSD1.230,231 Syntaxin-binding protein 5-like (Stxbp5l) or tomosyn-2 was identified in an F2 intercross from the BTBR T (+) tf (BTBR) Lep(ob/ob) and C57BL/6 (B6) Lep(ob/ob) mouse strains as a key negative regulator of insulin secretion.232 The same crossbreeding approach yielded Tsc2 as a gene underlying a QTL for nonalcoholic fatty liver disease (NAFLD) on chromosome 17. It was demonstrated that Tsc2(+/-) mice exhibited an increase in lipogenic gene expression levels in the liver in an insulin-dependent manner.233 The gene encoding the bile acid transporter Slco1a6 has been presented as a candidate gene for altered transport of taurocholic acid (TCA), resulting in broad gene regulation in pancreatic islets.234 In an NZO-based crossbreeding approach, one of the components of the KATP channel in pancreatic β-cells, Abcc8, was identified as causative factor in early-phase glucose-mediated insulin secretion.235 Lastly, Tbc1d1, a Rab-GAP protein that is presumably involved in GLUT4 vesicle sorting in skeletal muscle was identified as causal variant for the Nob1 obesity QTL derived from a crossbreeding of lean SJL with obese NZO (New Zealand obese) mice.91,236,237 Interestingly, both QTL, Nidd1 and Nob1 exhibit strong epistatic interaction as well as interaction with dietary fat in an outcross model of NZO and lean SJL mice.91,93,238 and both Zfp69 and Tbc1d1 genes are directly responsible for fat storage and fatty acid oxidation, respectively. This underscores the importance of altered lipid partitioning as a common denominator in the pathogenesis of obesity-driven diabetes.

 

All nine positionally cloned genes were located within consensus QTL regions, i.e. loci that have been linked to diabetes-related traits in multiple crossbreeding experiments. In fact, our meta-analyses of 77 published genome-wide linkage scans with hundreds of QTL strongly indicated the presence of consensus regions for metabolic traits in the mouse genome, and these hotspots could provide guidance for identifying novel gene variants involved in the development of the disease.239,240 Nevertheless, generation and refinement of novel polygenic mouse models is important since complex genetics seems to contribute significantly to the pathogenesis of the human disease 241 Moreover, diverse genetic tools such as the generation of Chromosome Substitution Strains (CSSs) and combination of classical breeding approaches with high-throughput genotyping, sequencing and genetic engineering technologies, and information repositories highlight the power of the mouse for genetic, functional, and systems studies of complex traits and disease models.242

 

summary

 

Although immune system, ectopic fat, and macro/micronutrients all contribute in part to the susceptibility for diabetes in the obese state, most of the underlying molecular mechanisms are still poorly understood. The identification of susceptibility genes mediating the progression of type 2 diabetes is crucial to prevent the massive epidemics of the disease. Future research will be focused not only on gene-gene interactions but also on the interplay of genetic and environmental risk factors.

 

References

 

  1. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. Aug 2000;106(4):473-481.
  2. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. Dec 14 2006;444(7121):840-846.
  3. Mokdad AH, Ford ES, Bowman BA, et al. Prevalence of obesity, diabetes, and obesity-related health risk factors, 2001. Jama. Jan 1 2003;289(1):76-79.
  4. Ogurtsova K, da Rocha Fernandes JD, Huang Y, et al. IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes research and clinical practice. Jun 2017;128:40-50.
  5. Farag YM, Gaballa MR. Diabesity: an overview of a rising epidemic. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. Jan 2011;26(1):28-35.
  6. Kalra S. Diabesity. JPMA. The Journal of the Pakistan Medical Association. Apr 2013;63(4):532-534.
  7. WHO. http://www.who.int/mediacentre/factsheets/fs311/en/.
  8. Abbasi F, Brown BW, Jr., Lamendola C, McLaughlin T, Reaven GM. Relationship between obesity, insulin resistance, and coronary heart disease risk. J Am Coll Cardiol. Sep 4 2002;40(5):937-943.
  9. Kahn SE. Clinical review 135: The importance of beta-cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab. Sep 2001;86(9):4047-4058.
  10. Fujioka S, Matsuzawa Y, Tokunaga K, Tarui S. Contribution of intra-abdominal fat accumulation to the impairment of glucose and lipid metabolism in human obesity. Metabolism. Jan 1987;36(1):54-59.
  11. Despres JP, Nadeau A, Tremblay A, et al. Role of deep abdominal fat in the association between regional adipose tissue distribution and glucose tolerance in obese women. Diabetes. Mar 1989;38(3):304-309.
  12. Pouliot MC, Despres JP, Nadeau A, et al. Visceral obesity in men. Associations with glucose tolerance, plasma insulin, and lipoprotein levels. Diabetes. Jul 1992;41(7):826-834.
  13. Park KS, Rhee BD, Lee KU, et al. Intra-abdominal fat is associated with decreased insulin sensitivity in healthy young men. Metabolism. 1991;40(6):600-603.
  14. Marin P, Andersson B, Ottosson M, et al. The morphology and metabolism of intraabdominal adipose tissue in men. Metabolism. Nov 1992;41(11):1242-1248.
  15. Despres JP, Moorjani S, Lupien PJ, Tremblay A, Nadeau A, Bouchard C. Regional distribution of body fat, plasma lipoproteins, and cardiovascular disease. Arteriosclerosis. Jul-Aug 1990;10(4):497-511.
  16. Carey DG, Nguyen TV, Campbell LV, Chisholm DJ, Kelly P. Genetic influences on central abdominal fat: a twin study. Int J Obes Relat Metab Disord. Aug 1996;20(8):722-726.
  17. Lillioja S, Young AA, Culter CL, et al. Skeletal muscle capillary density and fiber type are possible determinants of in vivo insulin resistance in man. J Clin Invest. Aug 1987;80(2):415-424.
  18. Solini A, Bonora E, Bonadonna R, Castellino P, DeFronzo RA. Protein metabolism in human obesity: relationship with glucose and lipid metabolism and with visceral adipose tissue. J Clin Endocrinol Metab. Aug 1997;82(8):2552-2558.
  19. Weiss R, Dufour S, Taksali SE, et al. Prediabetes in obese youth: a syndrome of impaired glucose tolerance, severe insulin resistance, and altered myocellular and abdominal fat partitioning. Lancet. Sep 20 2003;362(9388):951-957.
  20. Bonora E, Del Prato S, Bonadonna RC, et al. Total body fat content and fat topography are associated differently with in vivo glucose metabolism in nonobese and obese nondiabetic women. Diabetes. Sep 1992;41(9):1151-1159.
  21. Landin K, Lonnroth P, Krotkiewski M, Holm G, Smith U. Increased insulin resistance and fat cell lipolysis in obese but not lean women with a high waist/hip ratio. Eur J Clin Invest. Oct 1990;20(5):530-535.
  22. Abate N, Garg A, Peshock RM, Stray-Gundersen J, Grundy SM. Relationships of generalized and regional adiposity to insulin sensitivity in men. J Clin Invest. Jul 1995;96(1):88-98.
  23. Goodpaster BH, Thaete FL, Simoneau JA, Kelley DE. Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes. Oct 1997;46(10):1579-1585.
  24. Klein S, Fontana L, Young VL, et al. Absence of an effect of liposuction on insulin action and risk factors for coronary heart disease. N Engl J Med. Jun 17 2004;350(25):2549-2557.
  25. Barzilai N, She L, Liu BQ, et al. Surgical removal of visceral fat reverses hepatic insulin resistance. Diabetes. Jan 1999;48(1):94-98.
  26. Thorne A, Lonnqvist F, Apelman J, Hellers G, Arner P. A pilot study of long-term effects of a novel obesity treatment: omentectomy in connection with adjustable gastric banding. Int J Obes Relat Metab Disord. Feb 2002;26(2):193-199.
  27. Csendes A, Maluenda F, Burgos AM. A prospective randomized study comparing patients with morbid obesity submitted to laparotomic gastric bypass with or without omentectomy. Obesity surgery. Apr 2009;19(4):490-494.
  28. Herrera MF, Pantoja JP, Velazquez-Fernandez D, et al. Potential additional effect of omentectomy on metabolic syndrome, acute-phase reactants, and inflammatory mediators in grade III obese patients undergoing laparoscopic Roux-en-Y gastric bypass: a randomized trial. Diabetes care. Jul 2010;33(7):1413-1418.
  29. Lima MM, Pareja JC, Alegre SM, et al. Acute effect of roux-en-y gastric bypass on whole-body insulin sensitivity: a study with the euglycemic-hyperinsulinemic clamp. J Clin Endocrinol Metab. Aug 2010;95(8):3871-3875.
  30. Fabbrini E, Tamboli RA, Magkos F, et al. Surgical removal of omental fat does not improve insulin sensitivity and cardiovascular risk factors in obese adults. Gastroenterology. Aug 2010;139(2):448-455.
  31. Thomas EL, Frost G, Taylor-Robinson SD, Bell JD. Excess body fat in obese and normal-weight subjects. Nutrition research reviews. Jun 2012;25(1):150-161.
  32. Thomas EL, Parkinson JR, Frost GS, et al. The missing risk: MRI and MRS phenotyping of abdominal adiposity and ectopic fat. Obesity (Silver Spring). Jan 2012;20(1):76-87.
  33. O'Donovan G, Thomas EL, McCarthy JP, et al. Fat distribution in men of different waist girth, fitness level and exercise habit. Int J Obes (Lond). Dec 2009;33(12):1356-1362.
  34. van der Kooy K, Seidell JC. Techniques for the measurement of visceral fat: a practical guide. Int J Obes Relat Metab Disord. Apr 1993;17(4):187-196.
  35. Abate N, Burns D, Peshock RM, Garg A, Grundy SM. Estimation of adipose tissue mass by magnetic resonance imaging: validation against dissection in human cadavers. J Lipid Res. Aug 1994;35(8):1490-1496.
  36. Thomas EL, Fitzpatrick JA, Malik SJ, Taylor-Robinson SD, Bell JD. Whole body fat: content and distribution. Progress in nuclear magnetic resonance spectroscopy. Aug 2013;73:56-80.
  37. Xia Y, Ergun DL, Wacker WK, Wang X, Davis CE, Kaul S. Relationship between dual-energy X-ray absorptiometry volumetric assessment and X-ray computed tomography-derived single-slice measurement of visceral fat. Journal of clinical densitometry : the official journal of the International Society for Clinical Densitometry. Jan-Mar 2014;17(1):78-83.
  38. Kaul S, Rothney MP, Peters DM, et al. Dual-energy X-ray absorptiometry for quantification of visceral fat. Obesity (Silver Spring). Jun 2012;20(6):1313-1318.
  39. Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. Jul 1998;15(7):539-553.
  40. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome--a new worldwide definition. Lancet. Sep 24-30 2005;366(9491):1059-1062.
  41. Seidell JC, Oosterlee A, Deurenberg P, Hautvast JG, Ruijs JH. Abdominal fat depots measured with computed tomography: effects of degree of obesity, sex, and age. Eur J Clin Nutr. Sep 1988;42(9):805-815.
  42. Ferland M, Despres JP, Tremblay A, et al. Assessment of adipose tissue distribution by computed axial tomography in obese women: association with body density and anthropometric measurements. Br J Nutr. Mar 1989;61(2):139-148.
  43. Despres JP, Prud'homme D, Pouliot MC, Tremblay A, Bouchard C. Estimation of deep abdominal adipose-tissue accumulation from simple anthropometric measurements in men. Am J Clin Nutr. Sep 1991;54(3):471-477.
  44. Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocr Rev. Feb 1993;14(1):72-93.
  45. Hegele RA, Joy TR, Al-Attar SA, Rutt BK. Thematic review series: Adipocyte Biology. Lipodystrophies: windows on adipose biology and metabolism. J Lipid Res. Jul 2007;48(7):1433-1444.
  46. Stefan N, Schick F, Haring HU. Causes, Characteristics, and Consequences of Metabolically Unhealthy Normal Weight in Humans. Cell Metab. Aug 01 2017;26(2):292-300.
  47. Gavrilova O, Marcus-Samuels B, Graham D, et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin Invest. Feb 2000;105(3):271-278.
  48. Konrad D, Rudich A, Schoenle EJ. Improved glucose tolerance in mice receiving intraperitoneal transplantation of normal fat tissue. Diabetologia. Apr 2007;50(4):833-839.
  49. Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. May 2008;7(5):410-420.
  50. Virtue S, Vidal-Puig A. It's not how fat you are, it's what you do with it that counts. PLoS Biol. Sep 23 2008;6(9):e237.
  51. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. Dec 2003;112(12):1821-1830.
  52. Rasouli N, Kern PA. Adipocytokines and the metabolic complications of obesity. J Clin Endocrinol Metab. Nov 2008;93(11 Suppl 1):S64-73.
  53. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annual review of immunology. 2011;29:415-445.
  54. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW, Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. Dec 2003;112(12):1796-1808.
  55. Hotamisligil GS. Inflammation and metabolic disorders. Nature. Dec 14 2006;444(7121):860-867.
  56. Van Coillie E, Van Damme J, Opdenakker G. The MCP/eotaxin subfamily of CC chemokines. Cytokine & growth factor reviews. Mar 1999;10(1):61-86.
  57. Old LJ. Tumor necrosis factor (TNF). Science. Nov 8 1985;230(4726):630-632.
  58. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. Jan 1 1993;259(5091):87-91.
  59. Torti FM, Dieckmann B, Beutler B, Cerami A, Ringold GM. A macrophage factor inhibits adipocyte gene expression: an in vitro model of cachexia. Science. Aug 30 1985;229(4716):867-869.
  60. Feinstein R, Kanety H, Papa MZ, Lunenfeld B, Karasik A. Tumor necrosis factor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J Biol Chem. Dec 15 1993;268(35):26055-26058.
  61. Obeid LM, Linardic CM, Karolak LA, Hannun YA. Programmed cell death induced by ceramide. Science. Mar 19 1993;259(5102):1769-1771.
  62. Muoio DM. Intramuscular triacylglycerol and insulin resistance: Guilty as charged or wrongly accused? Biochim Biophys Acta. Dec 1 2009.
  63. Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases. Biochim Biophys Acta. Dec 30 2002;1585(2-3):202-212.
  64. Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. Mar 11 2005;120(5):649-661.
  65. Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. Dec 2004;114(12):1752-1761.
  66. Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. Nov-Dec 2005;42(6):517-525.
  67. Martinez-Abundis E, Reynoso-von Drateln C, Hernandez-Salazar E, Gonzalez-Ortiz M. Effect of etanercept on insulin secretion and insulin sensitivity in a randomized trial with psoriatic patients at risk for developing type 2 diabetes mellitus. Arch Dermatol Res. Nov 2007;299(9):461-465.
  68. Vendrell J, Maymo-Masip E, Tinahones F, et al. Tumor necrosis-like weak inducer of apoptosis as a proinflammatory cytokine in human adipocyte cells: up-regulation in severe obesity is mediated by inflammation but not hypoxia. J Clin Endocrinol Metab. Jun 2010;95(6):2983-2992.
  69. Chacon MR, Richart C, Gomez JM, et al. Expression of TWEAK and its receptor Fn14 in human subcutaneous adipose tissue. Relationship with other inflammatory cytokines in obesity. Cytokine. Feb 7 2006;33(3):129-137.
  70. Maymo-Masip E, Fernandez-Veledo S, Garcia Espana A, et al. The rise of soluble TWEAK levels in severely obese subjects after bariatric surgery may affect adipocyte-cytokine production induced by TNFalpha. J Clin Endocrinol Metab. Aug 2013;98(8):E1323-1333.
  71. Aljada A, Ghanim H, Saadeh R, Dandona P. Insulin inhibits NFkappaB and MCP-1 expression in human aortic endothelial cells. J Clin Endocrinol Metab. Jan 2001;86(1):450-453.
  72. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci U S A. Jun 10 2003;100(12):7265-7270.
  73. Kanda H, Tateya S, Tamori Y, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. Jun 2006;116(6):1494-1505.
  74. Panee J. Monocyte Chemoattractant Protein 1 (MCP-1) in obesity and diabetes. Cytokine. Oct 2012;60(1):1-12.
  75. Krook A. IL-6 and metabolism-new evidence and new questions. Diabetologia. Jul 2008;51(7):1097-1099.
  76. Rotter V, Nagaev I, Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem. Nov 14 2003;278(46):45777-45784.
  77. Wallenius V, Wallenius K, Ahren B, et al. Interleukin-6-deficient mice develop mature-onset obesity. Nat Med. Jan 2002;8(1):75-79.
  78. Nieto-Vazquez I, Fernandez-Veledo S, de Alvaro C, Lorenzo M. Dual role of interleukin-6 in regulating insulin sensitivity in murine skeletal muscle. Diabetes. Dec 2008;57(12):3211-3221.
  79. Feingold KR, Soued M, Adi S, et al. Effect of interleukin-1 on lipid metabolism in the rat. Similarities to and differences from tumor necrosis factor. Arterioscler Thromb. May-Jun 1991;11(3):495-500.
  80. Strandberg L, Lorentzon M, Hellqvist A, et al. Interleukin-1 system gene polymorphisms are associated with fat mass in young men. J Clin Endocrinol Metab. Jul 2006;91(7):2749-2754.
  81. Nguyen MT, Favelyukis S, Nguyen AK, et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem. Nov 30 2007;282(48):35279-35292.
  82. Suganami T, Nishida J, Ogawa Y. A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha. Arterioscler Thromb Vasc Biol. Oct 2005;25(10):2062-2068.
  83. Larsen CM, Faulenbach M, Vaag A, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. Apr 12 2007;356(15):1517-1526.
  84. Netea MG, van der Graaf C, Van der Meer JW, Kullberg BJ. Toll-like receptors and the host defense against microbial pathogens: bringing specificity to the innate-immune system. J Leukoc Biol. May 2004;75(5):749-755.
  85. Lin Y, Lee H, Berg AH, Lisanti MP, Shapiro L, Scherer PE. The lipopolysaccharide-activated toll-like receptor (TLR)-4 induces synthesis of the closely related receptor TLR-2 in adipocytes. J Biol Chem. Aug 11 2000;275(32):24255-24263.
  86. Beutler B. Inferences, questions and possibilities in Toll-like receptor signalling. Nature. Jul 8 2004;430(6996):257-263.
  87. Fessler MB, Rudel LL, Brown JM. Toll-like receptor signaling links dietary fatty acids to the metabolic syndrome. Curr Opin Lipidol. Oct 2009;20(5):379-385.
  88. Kopp A, Buechler C, Bala M, Neumeier M, Scholmerich J, Schaffler A. Toll-like receptor ligands cause proinflammatory and prodiabetic activation of adipocytes via phosphorylation of extracellular signal-regulated kinase and c-Jun N-terminal kinase but not interferon regulatory factor-3. Endocrinology. Mar 2010;151(3):1097-1108.
  89. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. Jul 2006;116(7):1793-1801.
  90. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med. Feb 12 2004;350(7):664-671.
  91. Chadt A, Leicht K, Deshmukh A, et al. Tbc1d1 mutation in lean mouse strain confers leanness and protects from diet-induced obesity. Nat Genet. Nov 2008;40(11):1354-1359.
  92. Sethi JK, Vidal-Puig AJ. Thematic review series: adipocyte biology. Adipose tissue function and plasticity orchestrate nutritional adaptation. J Lipid Res. Jun 2007;48(6):1253-1262.
  93. Scherneck S, Nestler M, Vogel H, et al. Positional cloning of zinc finger domain transcription factor Zfp69, a candidate gene for obesity-associated diabetes contributed by mouse locus Nidd/SJL. PLoS Genet. Jul 2009;5(7):e1000541.
  94. Tan CY, Vidal-Puig A. Adipose tissue expandability: the metabolic problems of obesity may arise from the inability to become more obese. Biochem Soc Trans. Oct 2008;36(Pt 5):935-940.
  95. Smith U. Abdominal obesity: a marker of ectopic fat accumulation. J Clin Invest. May 2015;125(5):1790-1792.
  96. Krotkiewski M, Bjorntorp P, Sjostrom L, Smith U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J Clin Invest. Sep 1983;72(3):1150-1162.
  97. Lonn M, Mehlig K, Bengtsson C, Lissner L. Adipocyte size predicts incidence of type 2 diabetes in women. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. Jan 2010;24(1):326-331.
  98. Perry RJ, Camporez JP, Kursawe R, et al. Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes. Cell. Feb 12 2015;160(4):745-758.
  99. Smith U, Kahn BB. Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids. Journal of internal medicine. Nov 2016;280(5):465-475.
  100. Barak Y, Nelson MC, Ong ES, et al. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol Cell. Oct 1999;4(4):585-595.
  101. Moitra J, Mason MM, Olive M, et al. Life without white fat: a transgenic mouse. Genes Dev. Oct 15 1998;12(20):3168-3181.
  102. Simha V, Szczepaniak LS, Wagner AJ, DePaoli AM, Garg A. Effect of leptin replacement on intrahepatic and intramyocellular lipid content in patients with generalized lipodystrophy. Diabetes care. Jan 2003;26(1):30-35.
  103. Petersen KF, Oral EA, Dufour S, et al. Leptin reverses insulin resistance and hepatic steatosis in patients with severe lipodystrophy. J Clin Invest. May 2002;109(10):1345-1350.
  104. Oral EA, Simha V, Ruiz E, et al. Leptin-replacement therapy for lipodystrophy. N Engl J Med. Feb 21 2002;346(8):570-578.
  105. Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res. Jan 2006;45(1):42-72.
  106. van Herpen NA, Schrauwen-Hinderling VB. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol Behav. May 23 2008;94(2):231-241.
  107. Petersen MC, Shulman GI. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends in pharmacological sciences. Jul 2017;38(7):649-665.
  108. Turcotte LP, Fisher JS. Skeletal muscle insulin resistance: roles of fatty acid metabolism and exercise. Phys Ther. Nov 2008;88(11):1279-1296.
  109. Moro C, Bajpeyi S, Smith SR. Determinants of intramyocellular triglyceride turnover: implications for insulin sensitivity. Am J Physiol Endocrinol Metab. Feb 2008;294(2):E203-213.
  110. Boden G, Lebed B, Schatz M, Homko C, Lemieux S. Effects of acute changes of plasma free fatty acids on intramyocellular fat content and insulin resistance in healthy subjects. Diabetes. Jul 2001;50(7):1612-1617.
  111. Li L, Yang G, Li Q, Tang Y, Li K. High-fat- and lipid-induced insulin resistance in rats: the comparison of glucose metabolism, plasma resistin and adiponectin levels. Ann Nutr Metab. 2006;50(6):499-505.
  112. Holloway GP, Bonen A, Spriet LL. Regulation of skeletal muscle mitochondrial fatty acid metabolism in lean and obese individuals. Am J Clin Nutr. Jan 2009;89(1):455S-462S.
  113. Adams JM, 2nd, Pratipanawatr T, Berria R, et al. Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes. Jan 2004;53(1):25-31.
  114. Goodpaster BH, Kelley DE. Skeletal muscle triglyceride: marker or mediator of obesity-induced insulin resistance in type 2 diabetes mellitus? Curr Diab Rep. Jun 2002;2(3):216-222.
  115. Krssak M, Falk Petersen K, Dresner A, et al. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia. Jan 1999;42(1):113-116.
  116. Morino K, Petersen KF, Shulman GI. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes. Dec 2006;55 Suppl 2:S9-S15.
  117. Cooney GJ, Thompson AL, Furler SM, Ye J, Kraegen EW. Muscle long-chain acyl CoA esters and insulin resistance. Ann N Y Acad Sci. Jun 2002;967:196-207.
  118. Blaak EE, Wagenmakers AJ, Glatz JF, et al. Plasma FFA utilization and fatty acid-binding protein content are diminished in type 2 diabetic muscle. Am J Physiol Endocrinol Metab. Jul 2000;279(1):E146-154.
  119. Ortenblad N, Mogensen M, Petersen I, et al. Reduced insulin-mediated citrate synthase activity in cultured skeletal muscle cells from patients with type 2 diabetes: evidence for an intrinsic oxidative enzyme defect. Biochim Biophys Acta. Jun 30 2005;1741(1-2):206-214.
  120. Simoneau JA, Kelley DE. Altered glycolytic and oxidative capacities of skeletal muscle contribute to insulin resistance in NIDDM. J Appl Physiol. Jul 1997;83(1):166-171.
  121. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. Oct 2002;51(10):2944-2950.
  122. Hulver MW, Berggren JR, Cortright RN, et al. Skeletal muscle lipid metabolism with obesity. Am J Physiol Endocrinol Metab. Apr 2003;284(4):E741-747.
  123. Goodpaster BH, He J, Watkins S, Kelley DE. Skeletal muscle lipid content and insulin resistance: evidence for a paradox in endurance-trained athletes. J Clin Endocrinol Metab. Dec 2001;86(12):5755-5761.
  124. Kiens B. Skeletal muscle lipid metabolism in exercise and insulin resistance. Physiol Rev. Jan 2006;86(1):205-243.
  125. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes. Jul 2002;51(7):2005-2011.
  126. Boden G. Free fatty acids-the link between obesity and insulin resistance. Endocr Pract. Jan-Feb 2001;7(1):44-51.
  127. Roden M, Price TB, Perseghin G, et al. Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest. Jun 15 1996;97(12):2859-2865.
  128. Bostrom P, Andersson L, Vind B, et al. The SNARE protein SNAP23 and the SNARE-interacting protein Munc18c in human skeletal muscle are implicated in insulin resistance/type 2 diabetes. Diabetes. May 11 2010.
  129. Kahn BB. Type 2 diabetes: when insulin secretion fails to compensate for insulin resistance. Cell. Mar 6 1998;92(5):593-596.
  130. Weir GC, Bonner-Weir S. Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes. Dec 2004;53 Suppl 3:S16-21.
  131. Joost HG. Pathogenesis, Risk Assessment and Prevention of Type 2 Diabetes mellitus. Obesity Facts. JUN 2008;1(3):128-137.
  132. Unger RH. Minireview: weapons of lean body mass destruction: the role of ectopic lipids in the metabolic syndrome. Endocrinology. Dec 2003;144(12):5159-5165.
  133. Mauvais-Jarvis F, Kahn CR. Understanding the pathogenesis and treatment of insulin resistance and type 2 diabetes mellitus: what can we learn from transgenic and knockout mice? Diabetes Metab. Dec 2000;26(6):433-448.
  134. Leiter EH, Reifsnyder PC. Differential levels of diabetogenic stress in two new mouse models of obesity and type 2 diabetes. Diabetes. Feb 2004;53 Suppl 1:S4-11.
  135. Ortlepp JR, Kluge R, Giesen K, et al. A metabolic syndrome of hypertension, hyperinsulinaemia and hypercholesterolaemia in the New Zealand obese mouse. Eur J Clin Invest. Mar 2000;30(3):195-202.
  136. Lange C, Jeruschke K, Herberg L, Leiter EH, Junger E. The diabetes-prone NZO/Hl strain. Proliferation capacity of beta cells in hyperinsulinemia and hyperglycemia. Arch Physiol Biochem. Feb 2006;112(1):49-58.
  137. Junger E, Herberg L, Jeruschke K, Leiter EH. The diabetes-prone NZO/Hl strain. II. Pancreatic immunopathology. Lab Invest. Jul 2002;82(7):843-853.
  138. Chankiewitz E, Peschke D, Herberg L, et al. Did the gradual loss of GLUT2 cause a shift to diabetic disorders in the New Zealand obese mouse (NZO/Hl)? Exp Clin Endocrinol Diabetes. May 2006;114(5):262-269.
  139. Jurgens HS, Neschen S, Ortmann S, et al. Development of diabetes in obese, insulin-resistant mice: essential role of dietary carbohydrate in beta cell destruction. Diabetologia. Jul 2007;50(7):1481-1489.
  140. Dreja T, Jovanovic Z, Rasche A, et al. Diet-induced gene expression of isolated pancreatic islets from a polygenic mouse model of the metabolic syndrome. Diabetologia. Feb 2010;53(2):309-320.
  141. Kluth O, Mirhashemi F, Scherneck S, et al. Dissociation of lipotoxicity and glucotoxicity in a mouse model of obesity associated diabetes: role of forkhead box O1 (FOXO1) in glucose-induced beta cell failure. Diabetologia. Nov 24.
  142. Leiter EH, Coleman DL, Ingram DK, Reynolds MA. Influence of dietary carbohydrate on the induction of diabetes in C57BL/KsJ-db/db diabetes mice. J Nutr. Jan 1983;113(1):184-195.
  143. Mirhashemi F, Kluth O, Scherneck S, et al. High-Fat, Carbohydrate-Free Diet Markedly Aggravates Obesity but Prevents beta-Cell Loss and Diabetes in the Obese, Diabetes-Susceptible db/db Strain. Obesity Facts. DEC 2008;1(6):292-297.
  144. Ahima RS, Osei SY. Adipokines in obesity. Front Horm Res. 2008;36:182-197.
  145. Trayhurn P, Wood IS. Signalling role of adipose tissue: adipokines and inflammation in obesity. Biochem Soc Trans. Nov 2005;33(Pt 5):1078-1081.
  146. Gnacinska M, Malgorzewicz S, Stojek M, Lysiak-Szydlowska W, Sworczak K. Role of adipokines in complications related to obesity: a review. Adv Med Sci. 2009;54(2):150-157.
  147. Houseknecht KL, Baile CA, Matteri RL, Spurlock ME. The biology of leptin: a review. J Anim Sci. May 1998;76(5):1405-1420.
  148. Fei H, Okano HJ, Li C, et al. Anatomic localization of alternatively spliced leptin receptors (Ob-R) in mouse brain and other tissues. Proc Natl Acad Sci U S A. Jun 24 1997;94(13):7001-7005.
  149. Hamann A, Matthaei S. Regulation of energy balance by leptin. Exp Clin Endocrinol Diabetes. 1996;104(4):293-300.
  150. Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house mouse. J Hered. Dec 1950;41(12):317-318.
  151. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. Dec 1 1994;372(6505):425-432.
  152. Houseknecht KL, Flier SN, Frevert EU, Frederich RC, Flier JS, Kahn BB. Leptin secretion correlates with adipocyte size in genetic and dietary obesity. Diabetes. MAY 1996;45:144-144.
  153. Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature. Dec 14 2006;444(7121):847-853.
  154. Heymsfield SB, Greenberg AS, Fujioka K, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. Jama. Oct 27 1999;282(16):1568-1575.
  155. Kamohara S, Burcelin R, Halaas JL, Friedman JM, Charron MJ. Acute stimulation of glucose metabolism in mice by leptin treatment. Nature. Sep 25 1997;389(6649):374-377.
  156. Minokoshi Y, Kim YB, Peroni OD, et al. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature. Jan 17 2002;415(6869):339-343.
  157. Kulkarni RN, Wang ZL, Wang RM, et al. Leptin rapidly suppresses insulin release from insulinoma cells, rat and human islets and, in vivo, in mice. J Clin Invest. Dec 1 1997;100(11):2729-2736.
  158. Fan W, Dinulescu DM, Butler AA, Zhou J, Marks DL, Cone RD. The central melanocortin system can directly regulate serum insulin levels. Endocrinology. Sep 2000;141(9):3072-3079.
  159. Meek TH, Morton GJ. Leptin, diabetes, and the brain. Indian journal of endocrinology and metabolism. Dec 2012;16(Suppl 3):S534-542.
  160. Park S, Ahn IS, Kim DS. Central infusion of leptin improves insulin resistance and suppresses beta-cell function, but not beta-cell mass, primarily through the sympathetic nervous system in a type 2 diabetic rat model. Life sciences. Jun 5 2010;86(23-24):854-862.
  161. Kim JY, van de Wall E, Laplante M, et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest. Sep 2007;117(9):2621-2637.
  162. Kawano J, Arora R. The role of adiponectin in obesity, diabetes, and cardiovascular disease. J Cardiometab Syndr. Winter 2009;4(1):44-49.
  163. Kazumi T, Kawaguchi A, Hirano T, Yoshino G. Serum adiponectin is associated with high-density lipoprotein cholesterol, triglycerides, and low-density lipoprotein particle size in young healthy men. Metabolism. May 2004;53(5):589-593.
  164. Weyer C, Funahashi T, Tanaka S, et al. Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab. May 2001;86(5):1930-1935.
  165. Kamigaki M, Sakaue S, Tsujino I, et al. Oxidative stress provokes atherogenic changes in adipokine gene expression in 3T3-L1 adipocytes. Biochem Biophys Res Commun. Jan 13 2006;339(2):624-632.
  166. Kim KY, Kim JK, Jeon JH, Yoon SR, Choi I, Yang Y. c-Jun N-terminal kinase is involved in the suppression of adiponectin expression by TNF-alpha in 3T3-L1 adipocytes. Biochem Biophys Res Commun. Feb 11 2005;327(2):460-467.
  167. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest. Jul 2006;116(7):1784-1792.
  168. Kadowaki T, Yamauchi T. Adiponectin and adiponectin receptors. Endocr Rev. May 2005;26(3):439-451.
  169. Kusminski CM, McTernan PG, Schraw T, et al. Adiponectin complexes in human cerebrospinal fluid: distinct complex distribution from serum. Diabetologia. Mar 2007;50(3):634-642.
  170. Barnes KM, Miner JL. Role of resistin in insulin sensitivity in rodents and humans. Curr Protein Pept Sci. Feb 2009;10(1):96-107.
  171. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. Jan 2005;1(1):15-25.
  172. Rajala MW, Obici S, Scherer PE, Rossetti L. Adipose-derived resistin and gut-derived resistin-like molecule-beta selectively impair insulin action on glucose production. J Clin Invest. Jan 2003;111(2):225-230.
  173. Hardie DG. AMPK: a key regulator of energy balance in the single cell and the whole organism. Int J Obes (Lond). Sep 2008;32 Suppl 4:S7-12.
  174. Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-specific secretory factor inhibits adipocyte differentiation. J Biol Chem. Apr 6 2001;276(14):11252-11256.
  175. Antuna-Puente B, Feve B, Fellahi S, Bastard JP. Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab. Feb 2008;34(1):2-11.
  176. Abel ED, Peroni O, Kim JK, et al. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature. Feb 8 2001;409(6821):729-733.
  177. Yang Q, Graham TE, Mody N, et al. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature. Jul 21 2005;436(7049):356-362.
  178. Wolf G. Serum retinol-binding protein: a link between obesity, insulin resistance, and type 2 diabetes. Nutr Rev. May 2007;65(5):251-256.
  179. Graham TE, Yang Q, Bluher M, et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N Engl J Med. Jun 15 2006;354(24):2552-2563.
  180. von Eynatten M, Humpert PM. Retinol-binding protein-4 in experimental and clinical metabolic disease. Expert Rev Mol Diagn. May 2008;8(3):289-299.
  181. Fukuhara A, Matsuda M, Nishizawa M, et al. Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science. Jan 21 2005;307(5708):426-430.
  182. Berndt J, Kloting N, Kralisch S, et al. Plasma visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes. Oct 2005;54(10):2911-2916.
  183. Arner P. Visfatin--a true or false trail to type 2 diabetes mellitus. J Clin Endocrinol Metab. Jan 2006;91(1):28-30.
  184. Fukuhara A, Matsuda M, Nishizawa M, et al. Retraction. Science. Oct 26 2007;318(5850):565.
  185. Varma V, Yao-Borengasser A, Rasouli N, et al. Human visfatin expression: relationship to insulin sensitivity, intramyocellular lipids, and inflammation. J Clin Endocrinol Metab. Feb 2007;92(2):666-672.
  186. Bluher M. Vaspin in obesity and diabetes: pathophysiological and clinical significance. Endocrine. Apr 2012;41(2):176-182.
  187. Heiker JT. Vaspin (serpinA12) in obesity, insulin resistance, and inflammation. Journal of peptide science : an official publication of the European Peptide Society. Mar 5 2014.
  188. Liu S, Dong Y, Wang T, et al. Vaspin inhibited proinflammatory cytokine induced activation of nuclear factor-kappa B and its downstream molecules in human endothelial EA.hy926 cells. Diabetes research and clinical practice. Dec 25 2013.
  189. Kempf K, Rose B, Illig T, et al. Vaspin (SERPINA12) genotypes and risk of type 2 diabetes: Results from the MONICA/KORA studies. Exp Clin Endocrinol Diabetes. Mar 2010;118(3):184-189.
  190. Tan BK, Adya R, Randeva HS. Omentin: a novel link between inflammation, diabesity, and cardiovascular disease. Trends in cardiovascular medicine. Jul 2010;20(5):143-148.
  191. Yamawaki H, Kuramoto J, Kameshima S, Usui T, Okada M, Hara Y. Omentin, a novel adipocytokine inhibits TNF-induced vascular inflammation in human endothelial cells. Biochem Biophys Res Commun. May 6 2011;408(2):339-343.
  192. Yan P, Liu D, Long M, Ren Y, Pang J, Li R. Changes of serum omentin levels and relationship between omentin and adiponectin concentrations in type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes. Apr 2011;119(4):257-263.
  193. Herder C, Carstensen M, Ouwens DM. Anti-inflammatory cytokines and risk of type 2 diabetes. Diabetes, obesity & metabolism. Sep 2013;15 Suppl 3:39-50.
  194. Castan-Laurell I, Dray C, Attane C, Duparc T, Knauf C, Valet P. Apelin, diabetes, and obesity. Endocrine. Aug 2011;40(1):1-9.
  195. Bertrand C, Valet P, Castan-Laurell I. Apelin and energy metabolism. Frontiers in physiology. 2015;6:115.
  196. Alipour FG, Ashoori MR, Pilehvar-Soltanahmadi Y, Zarghami N. An overview on biological functions and emerging therapeutic roles of apelin in diabetes mellitus. Diabetes & metabolic syndrome. Dec 2017;11 Suppl 2:S919-S923.
  197. Moreno-Aliaga MJ, Perez-Echarri N, Marcos-Gomez B, et al. Cardiotrophin-1 is a key regulator of glucose and lipid metabolism. Cell Metab. Aug 03 2011;14(2):242-253.
  198. Escote X, Gomez-Zorita S, Lopez-Yoldi M, et al. Role of Omentin, Vaspin, Cardiotrophin-1, TWEAK and NOV/CCN3 in Obesity and Diabetes Development. International journal of molecular sciences. Aug 15 2017;18(8).
  199. Lopez-Yoldi M, Stanhope KL, Garaulet M, et al. Role of cardiotrophin-1 in the regulation of metabolic circadian rhythms and adipose core clock genes in mice and characterization of 24-h circulating CT-1 profiles in normal-weight and overweight/obese subjects. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. Apr 2017;31(4):1639-1649.
  200. Murahovschi V, Pivovarova O, Ilkavets I, et al. WISP1 is a novel adipokine linked to inflammation in obesity. Diabetes. Mar 2015;64(3):856-866.
  201. Sahin Ersoy G, Altun Ensari T, Vatansever D, Emirdar V, Cevik O. Novel adipokines WISP1 and betatrophin in PCOS: relationship to AMH levels, atherogenic and metabolic profile. Gynecological endocrinology : the official journal of the International Society of Gynecological Endocrinology. Feb 2017;33(2):119-123.
  202. Sahin Ersoy G, Altun Ensari T, Subas S, Giray B, Simsek EE, Cevik O. WISP1 is a novel adipokine linked to metabolic parameters in gestational diabetes mellitus. The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstet. Apr 2017;30(8):942-946.
  203. Arner P, Kulyte A. MicroRNA regulatory networks in human adipose tissue and obesity. Nature reviews. Endocrinology. May 2015;11(5):276-288.
  204. Hashimoto N, Tanaka T. Role of miRNAs in the pathogenesis and susceptibility of diabetes mellitus. Journal of human genetics. Feb 2017;62(2):141-150.
  205. Thomou T, Mori MA, Dreyfuss JM, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. Feb 23 2017;542(7642):450-455.
  206. Ying W, Riopel M, Bandyopadhyay G, et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell. Oct 05 2017;171(2):372-384 e312.
  207. Moraes-Vieira PM, Saghatelian A, Kahn BB. GLUT4 Expression in Adipocytes Regulates De Novo Lipogenesis and Levels of a Novel Class of Lipids With Antidiabetic and Anti-inflammatory Effects. Diabetes. Jul 2016;65(7):1808-1815.
  208. Yore MM, Syed I, Moraes-Vieira PM, et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell. Oct 09 2014;159(2):318-332.
  209. Oh DY, Talukdar S, Bae EJ, et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. Sep 03 2010;142(5):687-698.
  210. McCarthy MI, Zeggini E. Genome-wide association studies in type 2 diabetes. Curr Diab Rep. Apr 2009;9(2):164-171.
  211. Stancakova A, Laakso M. Genetics of metabolic syndrome. Reviews in endocrine & metabolic disorders. Dec 2014;15(4):243-252.
  212. Morris AP, Voight BF, Teslovich TM, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet. Sep 2012;44(9):981-990.
  213. So HC, Gui AH, Cherny SS, Sham PC. Evaluating the heritability explained by known susceptibility variants: a survey of ten complex diseases. Genetic epidemiology. Jul 2011;35(5):310-317.
  214. Frayling TM, Timpson NJ, Weedon MN, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. May 11 2007;316(5826):889-894.
  215. Dina C, Meyre D, Gallina S, et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet. Jun 2007;39(6):724-726.
  216. Fischer J, Koch L, Emmerling C, et al. Inactivation of the Fto gene protects from obesity. Nature. Apr 16 2009;458(7240):894-898.
  217. Hakanen M, Raitakari OT, Lehtimaki T, et al. FTO genotype is associated with body mass index after the age of seven years but not with energy intake or leisure-time physical activity. J Clin Endocrinol Metab. Apr 2009;94(4):1281-1287.
  218. Church C, Lee S, Bagg EA, et al. A mouse model for the metabolic effects of the human fat mass and obesity associated FTO gene. PLoS Genet. Aug 2009;5(8):e1000599.
  219. Steinthorsdottir V, Thorleifsson G, Reynisdottir I, et al. A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet. Apr 26 2007.
  220. Sladek R, Rocheleau G, Rung J, et al. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature. Feb 22 2007;445(7130):881-885.
  221. Scott LJ, Mohlke KL, Bonnycastle LL, et al. A Genome-Wide Association Study of Type 2 Diabetes in Finns Detects Multiple Susceptibility Variants. Science. Apr 26 2007.
  222. McClellan J, King MC. Genetic heterogeneity in human disease. Cell. Apr 16 2010;141(2):210-217.
  223. Ott J, Wang J, Leal SM. Genetic linkage analysis in the age of whole-genome sequencing. Nature reviews. Genetics. May 2015;16(5):275-284.
  224. Schwenk RW, Vogel H, Schurmann A. Genetic and epigenetic control of metabolic health. Molecular metabolism. Sep 25 2013;2(4):337-347.
  225. Joost HG, Schurmann A. The genetic basis of obesity-associated type 2 diabetes (diabesity) in polygenic mouse models. Mammalian genome : official journal of the International Mammalian Genome Society. Oct 2014;25(9-10):401-412.
  226. Clee SM, Yandell BS, Schueler KM, et al. Positional cloning of Sorcs1, a type 2 diabetes quantitative trait locus. Nat Genet. Jun 2006;38(6):688-693.
  227. Dokmanovic-Chouinard M, Chung WK, Chevre JC, et al. Positional cloning of "Lisch-Like", a candidate modifier of susceptibility to type 2 diabetes in mice. PLoS Genet. 2008;4(7):e1000137.
  228. Chung B, Stadion M, Schulz N, et al. The diabetes gene Zfp69 modulates hepatic insulin sensitivity in mice. Diabetologia. Oct 2015;58(10):2403-2413.
  229. Scherneck S, Vogel H, Nestler M, Kluge R, Schurmann A, Joost HG. Role of zinc finger transcription factor zfp69 in body fat storage and diabetes susceptibility of mice. Results and problems in cell differentiation. 2010;52:57-68.
  230. Vogel H, Scherneck S, Kanzleiter T, et al. Loss of function of Ifi202b by a microdeletion on chromosome 1 of C57BL/6J mice suppresses 11beta-hydroxysteroid dehydrogenase type 1 expression and development of obesity. Human molecular genetics. Sep 1 2012;21(17):3845-3857.
  231. Vogel H, Jahnert M, Stadion M, Matzke D, Scherneck S, Schurmann A. A vast genomic deletion in the C56BL/6 genome affects different genes within the Ifi200 cluster on chromosome 1 and mediates obesity and insulin resistance. BMC genomics. Feb 15 2017;18(1):172.
  232. Bhatnagar S, Oler AT, Rabaglia ME, et al. Positional cloning of a type 2 diabetes quantitative trait locus; tomosyn-2, a negative regulator of insulin secretion. PLoS Genet. Oct 2011;7(10):e1002323.
  233. Wang CY, Stapleton DS, Schueler KL, et al. Tsc2, a positional candidate gene underlying a quantitative trait locus for hepatic steatosis. J Lipid Res. Aug 2012;53(8):1493-1501.
  234. Tian J, Keller MP, Oler AT, et al. Identification of the Bile Acid Transporter Slco1a6 as a Candidate Gene That Broadly Affects Gene Expression in Mouse Pancreatic Islets. Genetics. Nov 2015;201(3):1253-1262.
  235. Andrikopoulos S, Fam BC, Holdsworth A, et al. Identification of ABCC8 as a contributory gene to impaired early-phase insulin secretion in NZO mice. The Journal of endocrinology. Jan 2016;228(1):61-73.
  236. Dokas J, Chadt A, Joost HG, Al-Hasani H. Tbc1d1 deletion suppresses obesity in leptin-deficient mice. Int J Obes (Lond). Aug 2016;40(8):1242-1249.
  237. Chadt A, Immisch A, de Wendt C, et al. "Deletion of both Rab-GTPase-activating proteins TBC1D1 and TBC1D4 in mice eliminates insulin- and AICAR-stimulated glucose transport [corrected]. Diabetes. Mar 2015;64(3):746-759.
  238. Plum L, Giesen K, Kluge R, et al. Characterisation of the mouse diabetes susceptibilty locus Nidd/SJL: islet cell destruction, interaction with the obesity QTL Nob1, and effect of dietary fat. Diabetologia. Jun 2002;45(6):823-830.
  239. Wuschke S, Dahm S, Schmidt C, Joost HG, Al-Hasani H. A meta-analysis of quantitative trait loci associated with body weight and adiposity in mice. Int J Obes (Lond). May 2007;31(5):829-841.
  240. Schmidt C, Gonzaludo NP, Strunk S, et al. A meta-analysis of QTL for diabetes-related traits in rodents. Physiol Genomics. Jun 12 2008;34(1):42-53.
  241. Leiter EH. Selecting the "right" mouse model for metabolic syndrome and type 2 diabetes research. Methods Mol Biol. 2009;560:1-17.
  242. Buchner DA, Nadeau JH. Contrasting genetic architectures in different mouse reference populations used for studying complex traits. Genome research. Jun 2015;25(6):775-791.

 

 

Contraception

ABSTRACT

Contraception is important for the prevention of unintended pregnancies worldwide.  Many contraceptives also have other medical benefits, such as decreasing endometrial and ovarian cancer risk or regulating the pain and bleeding of menstruation.  It is essential for all physicians to have a basic knowledge of contraceptives, given that 99% of women in the United States will use contraception at some point in their reproductive lives.  This chapter provides an overview of the various hormonal and non-hormonal methods.   There is an emphasis placed on efficacy, pharmacology and the mechanism of action for each category of contraceptives.  Progestin-only methods include the implant, intrauterine devices (IUDs), injectables and pills. Combined estrogen-progestin methods include pills, patch and vaginal ring.  Non-hormonal methods include the copper IUD and barrier methods such as condoms or diaphragms.  Non-contraceptive benefits and contraindications for each particular method are also described.  Finally, some future developments are discussed. For complete coverage of this and all related areas of Endocrinology, please visit our FREE on-line web-textbook, www.endotext.org.

INTRODUCTION

The global population is at an all-time high at over 7 billion people. A large percentage of this population growth is in less developed countries where access to reproductive health care is limited. However, even in industrialized countries, unintended pregnancies do occur.  The rate of unintended pregnancies in the United States is higher than much of the industrialized world and approaches 50%.[i]  About 12% of these unintended pregnancies in the U.S. occur in teenagers and 75% occur in women who are poor or low income.[ii] While 2% of U.S. women have an induced abortion every year,[iii] about half of the unplanned pregnancies are carried to term.[iv] The rates of unwanted pregnancy in the United States are at least four times higher than in some countries in Europe and Japan.[v] These differences do not appear to be explained solely by exposure to the risk of pregnancy because other countries such as the Netherlands and Sweden have teenagers who engage in sexual activity earlier than most teenagers in the United States.[vi] The cost of contraception to the consumer and lack of insurance coverage for contraception had been identified as contributors to the high unintended pregnancy rate in the U.S. prior to passage of the Affordable Care Act (ACA).3  The ACA contraceptive mandate then required most insurers to cover contraception without cost-sharing beginning in August 2011.[vii]  However, not all women have insurance and the politics of contraceptive coverage are ever shifting.  Even without cost sharing, women continue to choose less effective methods and/or use methods incorrectly and inconsistently.  While the varied reasons for inconsistent contraceptive use have yet to be fully elucidated, the rate of unintended pregnancies is declining as uptake of contraception, and long-acting reversible contraception (LARC), in particular, increases.1

Safe and reliable family planning directly improves public health. New methods are an area of ongoing research, such as the development of microbicides, which will fulfill the unmet dual need of contraception and protection against sexually transmitted infections/human immunodeficiency virus in women. Women provided with effective contraception are protected against events that may threaten their personal and professional independence. Pregnancy and childbirth pose substantial health risks that should be actively avoided unless pregnancy is desired. The chance of death due to pregnancy and childbirth varies geographically but is always higher than that associated with currently available methods of contraception.[viii]

There is an ever increasing variety of contraceptives available to couples. Each method has its own distinct advantages and disadvantages. The ideal contraceptive would be effective, reversible, easy to use, not coitally dependent, safe, free of side effects, and inexpensive. Because one objectively perfect method does not yet exist, the choice of a family planning method should be individualized to each couple and may change during a woman's reproductive life. Currently, methods may be non-hormonal, progesterone-containing, or estrogen- and progesterone-containing. There have been, and there will continue to be, great strides to modify and optimize these methods. This chapter will serve as a review of contraceptive options with focus on the efficacy, mechanism of action, and non-contraceptive effects.

 

EFFICACY AND EFFECTIVENESS

There are inherent risks in our methodology for defining the efficacy of a contraceptive method. If 100 women used a contraceptive method for a year and five became pregnant, we cannot say that the method was 95% effective. We do not know who would have become pregnant without using family planning. Thus, we rely on a failure rate to describe the effectiveness of a method. However, all failure rates are not calculated equally and have different implications.

Most estimates of a contraceptive's efficacy refer to the first year of its use. Overall, the longer a woman uses a contraceptive method, the less likely it is to fail. Thus, the failure rate in the second year is lower than the first and the failure rate in the fifth year is lower than in that of the second. There are two commonly used failure rates to compare contraceptive methods: typical use and perfect use. 'Perfect use' is a measure of efficacy if the method is used perfectly, consistently, and according to specific guidelines. It represents the efficacy of the method in the laboratory setting (method failure rate). 'Typical use' estimates the probability of pregnancy during the first year of using the method in “the real world” setting. This measure of effectiveness takes into account occasional non-use of the method, incorrect use of the method, as well as pure failure of the method. Generally speaking, methods that are coitally dependent, such as condoms and diaphragms, have a larger disparity between typical use and perfect use. The failure rate for perfect use for oral contraceptive pills is approximately 0.3%, but the typical failure rate is about 9%.[ix] Given that it is impossible to predict if women will use a method perfectly, it is appropriate to quote typical-use failure rates in clinical counseling (Table 1). Methods that are long acting and require one visit to a clinician such as an IUD or an implant have very little disparity between perfect use and typical use, and thus are the most effective methods of contraception.

 

Method Typical Use Perfect Use
Implant 0.05 0.05
Male sterilization 0.15 0.1
LNG IUD (52mg) 0.2 0.2
LNG IUD (19.5mg) 0.2 0.2
LNG IUD (13.5mg) 0.4 0.4
Female sterilization 0.5 0.5
Copper IUD 0.8 0.6
Depo Provera 6 0.2
Pill 9 0.3
Patch 9 0.3
Ring 9 0.3
Diaphragm 12 6
Male condom 18 2
Female condom 21 5
Withdrawal 22 4
Fertility awareness 24 0.4-5
No method 85 85

Table 1:  Typical use and perfect use one-year failure rates for contraceptive methods9,[x],[xi]

 

PHARMACOLOGY

Progestins

The major portion of the contraceptive effect in systemic hormonal methods is due to the progestin compound. Progestins confer most of the contraceptive benefit by suppressing LH and ovulation. Progestins in oral contraceptives are derived from 19 nor-testosterone and include norethindrone, norethindrone acetate, ethynodiol diacetate, norgestrel, levonorgestrel, norethynodrel, desogestrel, norgestimate, and gestodene.

Progestins should be categorized according to their active structure and their parent compound. There are three molecularly distinct types of progestins: estranes, gonanes, and pregnanes. Estranes include norethindrone, norethindrone acetate, ethynodiol diacetate, and lynestrenol. Gonanes include desogestrel, norgestimate, and gestodene. Gonanes and estranes differ in their half-life and with respect to their estrogenic and anti-estrogenic effects. Pregnanes are used in injectable methods. Drospirenone is a spironolactone analog with anti-mineralocorticoid and anti-androgenic activity.[xii]

Progestin-only methods have many documented mechanisms of action, including inhibition of ovulation, thickened and decreased cervical mucus, suppression of mid-cycle peaks of LH and FSH, inhibition of progesterone receptor synthesis, reduction in the number and size of endometrial glands, reduction in ciliary activity within the fallopian tube, and premature luteolysis (decreased functioning of the corpus luteum).[xiii],[xiv],[xv],[xvi] Progestins at high concentrations likely suppress the initiation of folliculogenesis at the level of the hypothalamus. At slightly lower concentrations folliculogenesis can be initiated but the progestin prevents the LH surge at the level of the pituitary and therefore prevents ovulation. At even lower concentrations, progestins alter cervical mucus, tubal motility and/or the endometrium.[xvii]

 

Estrogens

Estrogens primarily serve to regulate bleeding, but also inhibit FSH and prevent formation of the dominant follicle.[xviii] In contrast to the long list of progestin formulations, only a few estrogenic compounds are used in hormonal contraceptives: ethinyl estradiol (EE), mestranol, and estradiol valerate. Other estrogens, particularly estetrol, are being tested clinically in contraceptive preparations. EE is pharmacologically active whereas mestranol must be converted into EE before it becomes active. Most contraceptives currently on the market contain 35 micrograms of estrogen or less. Ethinyl estradiol is absorbed rapidly and undergoes extensive hepatic first pass metabolism. Its plasma half-life has been reported to be in the range of 10-27 hours. Its half-life in tissue, such as endometrium, appears to be longer.

MECHANISM OF ACTION

While there is a great increase in the number of hormonal contraceptive options, most of these methods are a "variation on a theme.” The mechanism of action of hormonal contraception is primarily through the suppression of ovulation, but progestational effects include:[xix]

  • Inhibition of ovulation by suppressing luteinizing hormone (LH);
  • Thickening of cervical mucus, thus hampering the transport of sperm;
  • Possible inhibition of sperm capacitation;
  • Hampered implantation by the production of decidualized endometrium with exhausted and atrophic glands.

Estrogenic effects include:[xx]

  • Partial inhibition of ovulation in part by the suppression of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), depending on dose;
  • Alteration of secretions and cellular structures of the endometrium within the uterus.

 

PROGESTIN ONLY HORMONAL CONTRACEPTIVES

Current progestin-only methods include an etonogestrel (ENG) subdermal implant, intrauterine devices, depot medroxyprogesterone acetate, and progestin-only pills.

Advantages: Progestin-only methods do not have an estrogen component, thereby decreasing the complications attributable to estrogen (such as, cardiovascular disease, venous thromboembolism, and thrombophlebitis). Specific non-contraceptive benefits of these methods include scanty or no menses, decreased menstrual cramps and pain, suppression of pain associated with ovulation, decrease in endometrial cancer, ovarian cancer, and pelvic inflammatory disease, and potential improvement of the pain associated with endometriosis. All progestin-only contraceptive methods are reversible. Non-oral administration of progestin provides long term, effective contraception, decreases the risk of ectopic pregnancy, and is not coitally dependent. Specific indications for these methods may include women who are breastfeeding, women who are at greater risk for thromboembolic events, and all women who cannot take estrogen.

Disadvantages: These methods do not protect against sexually transmitted disease and HIV, and can alter the menstrual cycle (including breakthrough bleeding with an increased number of days of light bleeding, and potential amenorrhea).

The contraindications to progestin-only contraceptives are listed in Table 2. The only absolute contraindications are pregnancy, unexplained abnormal vaginal bleeding suspicious for a serious underlying condition, and breast cancer. Active liver disease has been deleted from the list of contraindications to DMPA. In general, one should exercise care when using this method in women with acute, severe liver disease or liver tumors.

Table 2. Contraindications to Progestin-only Hormonal Contraceptives

Progestin-only contraception should not be used for women with the following conditions:

• Known or suspected pregnancy

• Unexplained vaginal bleeding

• Breast cancer

 

Progestin-only IUDs should not be initiated in the following conditions:

• Suspected uterine infection

• Cervical cancer

 

Caution should be used when prescribing progestin only pills in the following conditions:

• Concurrent medication that cause progestins to be metabolized more rapidly:

• anti-seizure medications: phenytoin, carbamazepine, primidone, phenylbutazone, felbamate, ozcarbazine and lamotrigine (lamotrigine levels decreased by progestins)

• antibiotics: rifampin/rifampicin

• ritonavir- boosted protease inhibitors

• Women who have undergone bariatric surgery may have difficulty with absorption.

 

 

*There is no evidence that progestin-only methods increase the risk of cardiovascular events in a fashion similar to the combined oral contraceptive pill. Even though there is no evidence that progestin-only contraceptives cause cardiovascular events, the product labeling might still list active cardiovascular disease (or history of disease) as a contraindication.

 

PROGESTIN-ONLY IMPLANTS

 

There is one progestin-only implant available in the United States.  It is a single rod that contains 68mg of etonogestrel (ENG).  It is placed subdermally, usually in the upper extremity, in the office with local anesthesia.  It is FDA approved for up to 3 years. Small studies indicate continued efficacy for 4 or even 5 years, but insufficient numbers of obese BMI women have been included to make definitive recommendations in that cohort.[xxi],[xxii]  Larger studies are needed to routinely recommend extended use.

 

Efficacy:  The typical failure rate of the ENG implant is reported to be about 1 in 1000.  However, there are compiled data that demonstrate a Pearl Index of 0.00 (1716 women with 4103 women years of implant use and no pregnancies.[xxiii],[xxiv] There does appear to be a decrease in efficacy when combined with drugs that increase hepatic metabolism (Table 2).. Because of these possible interactions, the WHO classifies the implant in women taking these medications as category 2 (benefits likely outweigh the risks).

 

Side effects:  The most common side effect and most reported reason for removal of the implant is an irregular or unpredictable bleeding pattern24,[xxv]. Approximately ¼ of women reported prolonged or frequent bleeding. Fortunately, most women state that menstrual flow overall decreases with each successive year of implant use and the most common patterns were infrequent bleeding and amenorrhea.[xxvi]  There appears to be a slightly increased incidence of clinically insignificant ovarian cysts in users of the implant.  Unlike the concerns with depot medroxyprogesterone and bone mineral density (BMD), limited data suggest that the ENG implant does not have any effect on BMD.[xxvii]  Weight gain, however, has not been well studied.  Some trials report an increase of BMI by less than 1%, and perhaps as many as 12% of women will report an increase in weight.  When looking at reasons why women have the implant removed, only 3-7% state it was because of weight gain.  Therefore, it appears that weight gain is clinically insignificant25,[xxviii],[xxix].

 

Risks: The risks and contraindications are the same as those for other progestin-only methods.

 

Non contraceptive benefits: The ENG implant likely improves dysmenorrhea.  To date there is no randomized control trial comparing the implant to other acceptable treatments of dysmenorrhea. However, An observational study26 of 315 women showed that of the 187 women who reported dysmenorrhea at baseline, 151 (81%) had improvement of their dysmenorrhea and 26 (14%) reported no change.

INTRAUTERINE DEVICES

Although intrauterine devices (IUDs) are the most widely used form of reversible contraception worldwide, they are an underutilized contraceptive method in the United States.[xxx] IUDs and implants are the most effective methods of reversible contraception (20 times as effective as the hormonal methods of the pill, patch and ring), and have high user acceptability, with few medical contraindications.[xxxi]

Mechanism of action: There are five IUDs currently available in the U.S.: a copper IUD (Paragard®) and four progestin-impregnated intrauterine systems (IUS): Liletta®, Mirena®, Kyleena®, and Skyla®. Both medicated and non-medicated intrauterine devices (IUDs) have multiple mechanisms of action that provide for contraceptive protection. Both medicated and non-medicated IUDs can alter the uterine lining so that it becomes unfavorable for implantation. Release of copper ions also alters fluid in the uterine cavity in a manner that impairs the viability of sperm, thereby inhibiting fertilization. This mechanism may be responsible for the high efficacy of copper IUDs as emergency contraception.

IUDs can also alter both sperm motility and integrity.30,[xxxii],[xxxiii],[xxxiv] Medicated, or hormonal IUDs, can interfere with sperm motility by thickening cervical mucus. Sperm head-tail disruption has been reported in the presence of a copper IUD.30 IUDs, whether hormonal or non-hormonal, do not provide protection against sexually transmitted diseases. However, it is important to recognize that IUDs are not associated with PID,[xxxv],[xxxvi] and that the historical associations that both physicians and the lay public maintain between IUDs and PID/tubal infertility are false.

Timing of IUD insertion: The IUD can be inserted at any point in the cycle as long as the provider is reasonably certain the patient is not pregnant. An IUD can be inserted immediately postpartum, post abortion, or as an 'interval' insertion. Interval Insertion: An 'interval' insertion is defined as insertion in women who are neither postpartum nor post abortion, or an insertion in women 6 weeks after delivery.[xxxvii] Traditionally, physicians were taught that the best time for IUD insertion was either during menses or immediately after menstruation. Limiting insertion to these time points, however, created a barrier to their use. Data now suggest that IUD insertion between cycle days 12 to 17 results in greater IUD continuation rates. The Centers for Disease Control and Prevention reviewed data from more than 9,000 copper T-200 IUD insertions and found that IUDs placed after cycle day 11 resulted in fewer IUD removals during the first 2 months of IUD use. Insertions after day 17 resulted in more frequent IUD removal due to pain, bleeding, or accidental pregnancy.[xxxviii]

Progestin-only IUD:  The 52mg LNG IUS (Mirena, Liletta) initially releases 20 mcg of levonorgestrel per day from a polymer cylinder mounted on a T-shaped frame (32mm x 32mm) containing 52 mg levonorgestrel; it is covered by a release rate-controlling membrane. Mirena is FDA approved for 5 years of use; Liletta is FDA approved for 4 years of use. However, large studies support continued efficacy for 7 years of use with either brand.[xxxix] The failure rate is low: 0.16 per 100 woman years of use. Its mechanisms of action include production of an atrophic and inactive endometrium, disturbed ovulation, and thickening of the cervical mucus. Ovulation may be inhibited in about 20% of women, but this is not the main mechanism of action. The mean number of bleeding and spotting days is initially increased but both the volume of menstrual flow and the number of days of bleeding are reduced over time.[xl] During the first year of use, about 16% of women will be become amenorrheic and average menstrual bleeding days decrease to 2 per month by 12 months of use.40 A recent meta-analysis of randomized controlled trials reveal that LNG-20 IUS users were significantly more likely than all other IUD users to discontinue its use because of hormonal side effects and amenorrhea,[xli] so appropriate counseling is an important component of success. Initial irregular spotting or bleeding, and hormonal side effects like acne and ovarian cysts may occur in some users. In part because there is no user-dependence, the LNG-20 IUS offers much improved effectiveness over other hormonal methods.41,31

The 19.5mg LNG IUS (Kyleena) initially releases 17.5mcg of levonorgestrel per day during the first year from a 19.5mg reservoir mounted on a smaller T-shaped frame (28 x 30mm). Consequently, it has a more narrow insertion tube.  88% women in the first year and 100% of women in the third year of use will continue to ovulate.[xlii]  Although decreased over time, average number of bleeding/spotting days is higher than with the 52mg IUS.11  Kyleena is FDA approved for 5 years and there are no studies of extended use.

The 13.5mg LNG IUS (Skyla) has the same sized T frame and inserter as the 19.5mg IUS, but contains 13.5 mg of levonorgestrel, and initially releases 14mcg levonorgestrel per day, declining to 5mcg/day after 3 years. Less than 4% of women will have ovulation inhibition, and 6% of women have amenorrhea after 1 year. It is FDA approved for 3 years and there are no studies of extended use.[xliii]

Specific populations:  One population of interest is nulliparous women.  The failure rate of IUDs is low and similar for both parous and nulliparous women.  Acceptability, using the surrogate marker of continuation rate, shows a 90% retention rate in nulliparous women at one year.  As with efficacy, expulsion rates between parous and nulliparous appear to be the same, likely between 1-5% for all comers.  The risk of infection does not seem to vary between the parous and nulliparous women.[xliv] The 19.5mg and 13.5mg IUSs are marketed toward nulliparous women, but it is unknown as to whether its structural differences confer any specific benefits over the 52mg IUS in this population.

 

Non-contraceptive uses of 52mg LNG IUS: Multiple descriptive studies and clinical trials have been performed on the non-contraceptive benefits:[xlv],[xlvi],[xlvii],[xlviii],[xlix],[l],[li]

  • Treatment of menorrhagia in women with uterine fibroids and adenomyosis
  • Treatment of pain in women with endometriosis
  • Alternative to hysterectomy for women with menorrhagia
  • Prevention of endometrial hyperplasia in menopausal women using estrogen therapy
  • Prevention of endometrial proliferation and polyps in breast cancer survivors taking tamoxifen

One such study evaluated the efficacy and performance of the LNG-IUS in 44 women with menorrhagia after medical therapy had failed.51 At 12 months, 79.5% of participants continued use of the LNG-IUS. After LNG-IUS insertion, the most common bleeding pattern at 3 months was spotting followed predominantly by amenorrhea or oligomenorrhea at 6, 9, and 12 months. Hemoglobin levels significantly increased from 10.2 g/L before insertion to 12.8 g/L at 12 months (p<0.01).

The LNG-IUS is an effective treatment for menorrhagia. It is becoming increasingly evident that the LNG-IUS is also effective in the treatment of menorrhagia due to fibroids.  All studies in a systematic review showed an increase in hemoglobin.  Early observational data shows that the LNG-IUS does not appear to decrease the size of the fibroids.  Expulsion rates also varied in these studies from 0-20%, which probably reflects the great variety of cavity size and shape among women with fibroids.[lii],[liii]

Another prospective study observed that the 20mcg LNG-IUS provided more effective treatment for endometrial hyperplasia than oral progestin.[liv]

 

PROGESTIN-ONLY INJECTABLE

Depot medroxyprogesterone acetate (DMPA; Depo-Provera®) is delivered by a deep intramuscular injection of 150 mg of medroxyprogesterone acetate (MPA) every 12 weeks.[lv] Pharmacologically active levels (>0.5mg/ml) of MPA are achieved within 24 hours of injection. Serum levels remain >1.0 ng/ml for approximately three months after administration. By the fifth month, levels drop to 0.2mg/ml.[lvi] There is also a subcutaneous preparation of Depo-Provera, which administers 104 mg of MPA.  DMPA's main mechanism of action is inhibition of ovulation.

Non-contraceptive benefits of DMPA[lvii]

Decreased risk of:

  • Endometrial cancer
  • Iron deficiency anemia
  • Pelvic Inflammatory diseases
  • Ectopic pregnancy
  • Uterine Leiomyomata

Improvements of the following conditions:

  • Menorrhagia/Dysmenorrhea
  • Premenstrual syndrome symptoms
  • Pain in women with endometriosis
  • Seizures refractory to conventional anti-convulsants
  • Hemoglobinopathy
  • Endometrial hyperplasia
  • Vasomotor symptoms in menopausal women
  • Pelvic pain/dyspareunia in ovarian origin post hysterectomy
  • Metastatic breast cancer
  • Metastatic endometrial cancer

Efficacy: With ideal use, the failure rate is 0.3 per 100 woman-years which is similar to rates found with surgical sterilization.[lviii] Variations in body weight and concurrent medications have not been shown to alter efficacy.[lix] The typical-use failure rate is 6/100 women-years.

Side Effects: The most commonly cited side effects of DMPA are changes in menstrual patterns, weight, and mood. After 3 months of use, almost one-half of DMPA users report amenorrhea with the majority of the remaining women complaining of irregular bleeding.[lx] By the end of one year, nearly 75% of users will experience amenorrhea.[lxi] Short courses of high-dose estrogen do little to reduce bleeding among DMPA users.[lxii] In addition, early re-administration of DMPA (at 8-10 weeks instead of 12 weeks) does not reduce bleeding.[lxiii] Although many users of DMPA report weight gain, recent controlled studies show that its long-term use does not cause a significant increase in body weight.[lxiv] Product labeling for DMPA includes depression as a possible side effect. Two well-designed studies addressed the issue of depression and DMPA use. Patients followed up to one year after DMPA initiation showed no worsening of depressive symptoms compared to those who did not use Depo-Provera.[lxv],[lxvi] In clinical trial settings, DMPA does not cause statistically significant weight gain.[lxvii],[lxviii] However, African American women, Navajo Native Americans, and women with high baseline body mass index may be predisposed to weight gain with the use of this method.[lxix] Studies have shown that neither DMPA nor progestin-only injectable is associated with increased risk of breast cancer.[lxx]

Risks: A recent review showed that levels of triglyceride and total cholesterol do not change with DMPA use.69 Seven of the ten studies that measured serum lipid levels revealed a decrease in high density lipoprotein (HDL) while three out of five studies showed an increase in mean low density lipoprotein (LDL) levels. The clinical significance of these findings remains to be determined. In contrast to combination OCs, Depo-Provera is not associated with increases in coagulation-related factors or in blood pressure.[lxxi] Recent studies provide reassuring evidence that decreases in bone mineral density in current and recent Depo-Provera users are reversible and appear to be similar to changes seen in lactation.[lxxii]

Bone loss: On November 17, 2004, the FDA placed a black box in DMPA package labeling regarding the long-term use of DMPA and bone loss. Anecdotally, this warning had an immediate impact on decreased prescription of DMPA.[lxxiii] Depot medroxyprogesterone acetate use has not been linked to menopausal osteoporosis or fractures. A cross-sectional study from the WHO demonstrated that even after four years of use, BMD in former adult DMPA users is similar to that of never users.72 The 2002 cohort study of BMD in adult women performed by Scholes et al.[lxxiv] demonstrated that at 3 years of follow-up, BMD of former DMPA users was comparable to that of never users of injectable contraception. While the FDA warning also specifically heightened concerns about teens and bone loss in the setting of DMPA, a 2005 report from Scholes et al.[lxxv] showed that use of DMPA in this population does not put patients at risk for osteoporosis later in life. This cohort study followed 170 adolescents (including 80 who used DMPA at baseline) and found that recovery of BMD was complete within 12 months following DMPA discontinuation. BMD was ultimately observed to be higher in the former than in the never users of DMPA. Duration of DMPA use was not observed to impact speed of BMD recovery following DMPA discontinuation.  A Cochrane review indicated that there may be a small increased risk of fracture in women with increased duration of use.[lxxvi]

PROGESTIN-ONLY ORAL CONTRACEPTIVES

Progestin-only pills, also called the 'minipill' have a dose of progestin that is very close to the threshold of contraceptive efficacy; therefore, these pills must be taken continuously at the same time each day and without a pill-free interval. Less than one percent of oral contraceptive prescriptions in the United States are for the progestin-only oral contraceptive.[lxxvii] This form of contraception is traditionally most often used in women who are breastfeeding or in women who have contraindications to estrogen.  However, most women are candidates for this method. Only one progestin-only formulation is available in the United States with 0.35mg of norethindrone per pill. It is important to note that ovulation is not always inhibited with the use of progestin-only pills. Approximately half of cycles have suppressed ovulation and thus contraceptive efficacy is dependent on the other progestin related mechanisms listed previously.77

Efficacy: The typical failure rate of progestin-only pills is similar to that with combined oral contraceptives,[lxxviii] despite the fact that efficacy is only for 27 hours and requires consistent administration. Serum levels of progestin peak 2 hours after administration and return to near baseline levels within 24 hours.77 Variation of only a few hours in administration can be the difference in the progestin-only pill providing its contraceptive protection. Women should be prepared to use a back-up method if they are three hours late in taking the pill, if one pill is missed or if there is a delay in its administration.  Furthermore there are data to suggest that the efficacy of progestin-only pills in the setting of rifampin, certain anticonvulsants (excluding lamotrigine) and ritonavir-boosted protease inhibitors is decreased.  The WHO subsequently categorizes progestin-only pills in these settings as risks may outweigh benefits.

Side Effects: The main side effect associated with progestin-only pills is menstrual cycle irregularity. Spotting or breakthrough bleeding, amenorrhea, and shortened length of menstrual cycles are the most common irregularities experienced. A randomized, double-blind study by the WHO showed that an average of 53% of users had frequent bleeding, 22% had prolonged bleeding, 13% had irregular bleeding, and 6% had amenorrhea within 3 months of initiation.[lxxix] Menstrual irregularity is a common reason for method discontinuation. Other less common side effects include nausea, dizziness, headache, and breast tenderness.

Risks: In general, any contraceptive method protects against ectopic pregnancy. However, if progestin-only pill users get pregnant, on average 6-10% of the pregnancies will be ectopic, higher than the rate seen in women not using any method of contraception (2%).77 The overall risk, however, remains lower than the general population because so few women actually become pregnant (7%) while using this method of contraception.

A recent WHO case-control study of cardiovascular disease and progestin-only pill use found no significant increase in the risk of acute myocardial infarction (RR=1.0, 95%CI, 0.2-6.0), stroke (RR=1.1, 95%CI, 0.6-1.9), or venous thromboembolism (RR=1.8, 95%CI, 0.8-4.2) (34).  Thus far, progestin-only pills appear to have little or no effect on lipid metabolism, carbohydrate metabolism, hypertension, and coagulation factors.77

COMBINED ESTROGEN AND PROGESTIN HORMONAL CONTRACEPTION

 

The bioavailability of progesterone is poor, so progestins derived from androgens are used instead.  There are many active progestins in addition to norethindrone.  They are classified into generations as shown in Table 3.18,19

 

Generation Progestins Notes
First norethynodrel, norethindrone, norethindrone acetate, and ethynodiol diacetate Low potency, well tolerated, more breakthrough bleeding with low doses of estrogen
Second Levonorgestrel, norgestrel, norgestimate Higher potency, less breakthrough bleeding, more androgenic side effects
Third desogestrel, norgestimate, gestodene Decreased androgenic side effects
Fourth drospirinone Anti-androgenic and anti-mineralocorticoid effects

 

EXTENDED AND CONTINUOUS REGIMENS:[lxxx]

While manipulation of the timing of the pill-free-interval has been practiced by gynecologists for decades, there are now marketed ways to decrease the number of withdrawal bleeds experienced on the standard pill pack, which contains 21 active pills and 7 placebo pills (“21/7”). There are “24/4” regimens with only 4 days per month of placebo pills. Pills such as 'Seasonale' are packaged such that women take the pill for 84 consecutive days, and then stop for a week before inducing a withdrawal bleed. This method allows women to bleed only once every three months. This regimen is especially useful for women who suffer from endometriosis, heavy periods or severe PMS or menstrual cramps. Clinical trials have highlighted breakthrough bleed as an adverse effect of these extended formulations. However, many women favor the decreased number of bleeding days experienced with these newer regimens. Furthermore, there is evidence of greater ovarian suppression with potential for lower failure rates with these regimens.

 

Non-contraceptive Benefits of Combined Oral Contraceptives

Today there is a range of non-oral, non-daily hormonal contraceptive available, and the face of contraceptive management has changed. However, oral contraceptives have been widely used for decades, and they represent the most extensively studied drug on the market. Research regarding oral contraceptives focused on possible health risks. Many of the concerns of these health risks have been allayed. There is a large body of evidence demonstrating non-contraceptive health benefits of oral contraception. Table 3 is a list of potential non-contraceptive benefits. Specific clinical situations are described below.

Table 3. Advantages of oral contraceptives20

 

·       Reduction of ovarian and endometrial cancer risk

·       Decreased benign breast disease

·       Reversibility and quick return to fertility

·       Favorable bone mineral density profile

·       Reduced risk of benign ovarian tumors and ovarian cysts

·       Reduced risk of colorectal cancer

·       Reduced dysfunctional uterine bleeding

·       Decrease in menstrual flow and menorrhagia

·       Decrease in primary dysmenorrhea

·       Decreased risk of iron deficiency anemia

·       Improvement in hirsutism and acne

·       Decreased perimenopausal vasomotor symptoms

·       Decreased risk of premenstrual syndrome (PMS)/premenstrual dysphoric disorder (PMDD)

 

Reduction in ovarian cancer risk: Reduction in ovarian cancer risk increases with greater oral contraceptive use although protection is provided after as little as 3 to 6 months.[lxxxi] Compared to non-users, women who have used oral contraceptives for four years or fewer have a 30% decreased risk of ovarian cancer. If they used combined OC's for 5-11 years, they have a 50% reduction of risk; if they used combined OC's for more than 12 years, there is an 80% reduction in risk. This protective effect persists for at least 15 years after OC discontinuation.[lxxxii],[lxxxiii] These data hold true for women at genetically increased risk of breast cancer.  The Society of Gynecologic Oncology (SGO) recommends OC use for women with BRCA1 and BRCA2 in the absence of contraindications. Given the devastating effects of this disease and our lack of success with screening and early diagnosis, the chemo-prophylactic effects of oral contraceptives should be emphasized.

Prevention of Endometrial Cancer: There is a 50% reduction in endometrial cancer risk in OC users compared with never users.[lxxxiv] Reduced risk depends on duration of OC use. The risk is reduced by 20% with 1 year of use, 40% with 2 years of use, and 60% with 4 or more years of use.[lxxxv] The actual duration of protection after discontinuation is unknown but is estimated to be at least 15 years.[lxxxvi]

Benign Breast Disease: OC use significantly reduces fibrocystic breast change and fibroadenoma development.[lxxxvii],[lxxxviii],[lxxxix] The Oxford Family Planning Association Study found a decreased risk of benign breast disease with increasing duration of use; current users, however, were at lowest risk.[xc] Fibrocystic change is significantly decreased after 1 to 2 years of use,[xci] and lasts up to 1 year after OC discontinuation.90

Bone Mineral Density: Studies of both premenopausal and postmenopausal women seem to favor bone-sparing effects of OC's. A past history of OC use provided protection against low bone mineral density in a cross-sectional, retrospective study (OR=0.4, 95%CI, 0.2-0.5).[xcii] The same study observed increasing protection with increasing duration of use. Few studies have shown a protective effect against fracture risk-a case-control study showed that use of any OC in women after the age of 40 years provided significant protection against hip fractures during their menopause (OR=0.7;95%CI, 0.5-0.9).[xciii] Many studies, however, have not found a favorable association between OC's and bone mass.[xciv],[xcv] No study thus far has found a detrimental effect of OC's on bone mineral density.[xcvi]

Functional Ovarian Cysts: In general, studies of current monophasic or triphasic OC formulations demonstrate that OCs do not have a significant effect on the development of functional ovarian cysts.[xcvii],[xcviii]

Colorectal Cancer: A meta-analysis of pooled relative risks of colorectal cancer for ever-use of OCs from case-control studies was 0.81 (95%CI, 0.69-0.94) and from four cohort studies was 0.84 (95%CI, 0.72-0.97).[xcix] Women using high-dose OCs (with 50mcg estrogen) for greater than 96 months had a relative risk of 0.6 (95%CI, 0.4-0.9).[c] It is unclear if the results from this study apply to women using lower-dose oral contraceptives.

Relief from menstrual disorders: A randomized clinical trial of patients with dysfunctional uterine bleeding showed an 81-87% improvement in bleeding within 3 months compared to a 36-45% improvement seen in placebo treated patients.[ci] The likelihood of iron-deficiency anemia appears to be decreased in both current and past combination OC users. Anecdotal reports of treating primary dysmenorrhea with OCs document their effectiveness.[cii]

Reduction of acne: Two randomized, placebo-controlled trials showed that nearly 50% of women treated with a triphasic OCs containing norgestimate had an improvement in acne compared to 30% of women on placebo.[ciii],[civ] Ortho Tri Cyclen and Estrostep are approved by the FDA for the treatment of acne. Another pill that contains ethinyl estradiol and drospirenone is also effective in treating this condition, and may lead to overall improvement in facial acne.[cv] Other OCs too are being evaluated for similar use.[cvi],[cvii] All combination OCs likely reduce acne via an increase in sex hormone binding protein and a subsequent decrease in serum testosterone.

Reduced risk of adverse cardiovascular outcomes: In 2004, a study on the WHI database revealed that use of OCs is associated with better cardiovascular outcomes, including any cardiovascular disease, hypercholesterolemia, angina, myocardial infarction, transient ischemic attack, peripheral vascular disease, and need for cardiac catheterization. The data showed that increasing age, elevated body mass index and smoking greatly increased the risks, even in OC users.[cviii],[cix]

Risks

Many prescribing patterns of oral contraceptive and other hormonal contraceptive methods are based on perception rather than evidence-based medicine. Evidence-based medicine relies on the integration of clinical expertise with the best evidence from systematic review of research. As clinicians, we can use this methodology to refute misconceptions about oral contraceptives and promote many non-contraceptive benefits.[cx] However, there are some important contraindications to the use of estrogen-containing hormonal contraceptives (Table 4).

 

Table 4. Contraindications for the use of combined oral contraception

 

Estrogen-containing contraception should not be used for women with the following conditions:[cxi]

 

• Known presence or history of deep venous thrombosis or pulmonary embolism;

• History of cerebral vascular accident, coronary artery or ischemic heart disease;

• Diabetes with microvascular complications (neuropathy, retinopathy), duration greater than 20 years or older than 35 years;

• Personal history of estrogen-dependent cancer including current or history of breast cancer;

• Current pregnancy;

• Migraines with aura, focal neurological symptoms, vascular risk factors, vascular disease, or age greater than 35 years;

• Smoker, age greater than 35 years;

• Hypertension

• Liver disease (benign hepatic adenoma, liver cancer, active viral hepatitis, or severe cirrhosis);

• Major surgery with prolonged immobilization or any surgery of the legs;

 

 

SPECIAL CONSIDERATIONS

Oral Contraceptive Use and The Risk of Thromboembolism

The link between estrogen use and venous thromboembolism was identified more than 20 years ago.[cxii] Since then, there has been extensive literature that describes and attempts to elucidate this risk. A summary of these data show relative risks of venous thromboembolism ranging from 2.1 - 4.4.[cxiii] It has been a demonstrated that risk increases as estrogen dose increases. Despite these risks, it is still safer for a woman to use oral contraceptives than to become pregnant. The attributable risk, or number of new cases of venous thromboembolism attributable to estrogen, is on the order of about 6 per 100,000 women years.113 This is in contrast to the risk of venous thromboembolism in pregnancy - it is estimated that there are approximately 20 cases per 100,000 pregnant women years. Table 5 provides a summary of relative risk for VTE in different populations of women.

Table 5. Relative Risk of Venous Thromboembolism (VTE)

 

Population

 

 

Relative Risk (New cases per 100,000 women/year)

 

 

General Population

 

 

1 (4-5)

 

 

Pregnant Women

 

 

20 (48-60)

 

 

High-dose (>50μg EE) OCs

 

 

6-10 (24-50)

 

 

Low-dose (<50μg EE) OCs

 

 

3-4 (12-20)

 

 

Factor V Leiden carrier

 

 

6-8 (24-40)

 

 

Factor V Leiden Homozygote

 

 

80 (320-400)

 

 

Factor V Leiden carrier + OCs

 

 

30 (120-150)

 

 

Prothrombin G20210A carrier

 

 

3-4 (12-20)

 

 

Prothrombin G20210A mutation + OCs

 

 

7 (28-35)

 

 

Protein C or S deficiency

 

 

6-8 (24-40)

 

 

Protein C or S deficiency + OCs

 

 

6-8 (24-40)

 

 

Oral Contraceptive Use and Risk of Breast Cancer:

The relationship between OC use and breast cancer remains controversial. Two studies provide evidence that OC use is not associated with an increase in breast cancer incidence. The first study was conducted by the Collaborative Group on Hormonal Factors in Breast Disease. This group reanalyzed approximately 90% of the epidemiologic data available worldwide concerning oral contraceptive use and breast cancer risk.[cxiv],[cxv] The findings included a slight increase in the relative risk of localized breast cancer associated with current oral contraceptive use (relative risk 1.24, 95% CI 1.15-1.33) or oral contraceptive use within 1-4 years (relative risk 1.16, 95% CI 1.08-1.23) compared to never use. The study also demonstrated that breast cancers diagnosed in OC users were significantly less advanced than those in never users (relative risk 0.88 for spread of disease beyond the breast). They also noted there was no change in the effect of OC use associated with breast cancer by family history. Importantly, they demonstrated no overall effect of OC use that was associated with breast cancer by duration of use, dose, formulation, age at use, or age at breast cancer diagnosis. Oral contraceptive users and non-users older than 50 years have the same cumulative risk of diagnosis of breast cancer. Oral contraceptive use may accelerate the diagnosis of breast cancer but does not affect the overall risk.115

The second study involved over 8000 women, half of which had the diagnosis of breast cancer.[cxvi] Overall, 77% of cases and 79% of controls had ever used OCs. Ever users and current users of OCs were found not to have an increased risk of breast cancer compared to women who had never used OCs (OR 0.9, 95%CI 0.8-1.0 and 1.0, 95% CI, 0.8-1.0, respectively). The relative risk did not increase with increasing duration of OC use or higher estrogen doses. In addition, family history of breast cancer did not significantly impact risk.

 

In a third study,[cxvii] the authors showed an increased annual risk of breast cancer diagnosis of one per 7,690 women per year using hormonal contraception. Important limitations exist in this recent paper. The research was conducted via multiple Danish registries, which are methods of data collection, but not a study design.  Thus, these studies do not fall directly into standard epidemiologic classifications, and are subject to the biases of retrospective cohort studies. More specifically, the method of detection of the study outcome (breast cancer diagnosis) was not specified, nor was the stage of the diagnosis, nor whether or not the mortality due to breast cancer differed among women who had and had not used hormonal contraception. The clinical significance and health outcomes of higher breast cancer detection rates in the hormonal contraceptive users could not be determined. Finally and most importantly, the reference group, women who chose never to use hormonal contraception, may also make other behavioral, health and lifestyle decisions, including having additional clinical examinations, that affect their risk of cancer. This small absolute risk is unlikely to have clinical significance for women prioritizing pregnancy prevention, especially considering that previously published research[cxviii] shows that users of oral contraceptives are protected from other cancers including colorectal, endometrial, and ovarian cancer.

 

Oral Contraceptive Use and Liver Cancer

Non-case control studies of reproductive age women in western developed nations have reported an association between oral contraceptive and liver cancer. Recent population-based data, however, do not suggest any association between liver cancer and OC use among women in five developed nations. In addition, reassuring data from two studies in developing countries, including a large WHO study, do not support an increased risk of liver cancer with oral contraceptive use.[cxix]

Oral Contraceptive Use and Gallbladder Disease

Studies suggested in the 1970's that oral contraceptives were associated with an increased risk of gall bladder disease. Since then, numerous case-control and cohort studies have described an increased risk of benign gallbladder disease in oral contraceptive users. A meta-analysis of these studies published in 1990, however, found that few of these studies could stand up to internal validity measures. The relationship between benign gallbladder disease and oral contraceptives yields a relative risk of 1.1 with a 95% CI 1.1 - 1.2.81

 

Second Versus Third Generation Oral Contraceptives and Deep Vein Thrombosis.

There is no strong biological evidence that specific progestins have differential effects on VTE risk. While clotting factors may be altered differentially by specific progestins such data are not clinically relevant because we do not have a proven surrogate marker for VTE risk.[cxx] In the mid 1990's pharmaco-epidemiological studies reported that women using "third generation" oral contraceptives had a higher risk of venous thromboembolism (VTE) compared to women using "second generation" OCs.[cxxi],[cxxii],[cxxiii] Studies performed after the initial observation demonstrate a weak association between oral contraceptive use and VTE (strength of association ranges from 0.7 to 2.3). Paradoxically, larger doses of estrogen are associated with lower risks for VTE in these studies. This finding has questioned the biological plausibility of the hypothesis of associating 'new progestins’ to an increased risk of deep venous thrombosis (DVT).

It was suggested that the original studies included newer users of oral contraceptives that may have been innately at higher risk for DVT (new-user bias), thus biasing the results.8,[cxxiv] After reanalysis of the data, the FDA issued a statement stating that the risk of DVT with the 'new progestins "is not great enough to justify switching to other products".

The association of second versus third generation progestin and the risk of VTE was further analyzed in 2 separate meta-analyses. Twelve observational studies were included in one meta-analysis of the relative risk of VTE for OCs containing either desogestrel and gestodene or levonorgestrel.[cxxv] The relative risk of VTE in users of OCs with desogestrel and gestodene v levonorgestrel was 1.7 (95% CI, 1.3-2.1), an increase of approximately 11 more cases of VTE per 100,000 women per year. When accounting for duration of use and new use (less than 1 year), this increased risk persisted. However, differences in BMI and other comorbidities that may act as confounders were not accounted for in these studies.

The results in another meta-analysis of seven cohort and case-control studies similarly show the biases in these studies. The overall adjusted odds ratio for third versus second generation oral contraceptives was 1.7 (95% CI, 1.4-2.0).[cxxvi] Among first-time users (<1 year of use), the odds ratio for third versus second generation preparations was 3.1 (95% CI, 2.0-4.6), which decreased to 2.0 (95% CI, 1.4-2.7) in longer term users (1 year of use). In this paper, the new-user effect is clearly demonstrated.

Lidegaard et al retrospectively assessed the influence of OCs on the risk of VTE in women aged 15-44 years.[cxxvii] After adjusting for age, BMI, length of OC use, and family history of coagulopathies, the odds of VTE among current second generation OC users compared to non-users was 2.9 (95% CI, 2.2-3.8) while the odds of current third generation OC users compared to nonusers was 4.0 (95% CI, 3.2-4.9). After correcting for duration of use and differences in estrogen dose, the third/second generation risk ratio was 1.3 (95% CI, 1.0-1.8; p<0.05).

If there is any increased risk of VTE with third generation OCPs, this is likely to be marginal, with small absolute risks. The majority of the risk is conferred by the estrogen, which should be factored into contraceptive decision-making. Despite the heated debate and the revealed flaws with such study designs, a similar argument is now in the literature with regard to drospirenone-containing OCPs. Several studies have investigated the risk of VTE associated with OCPs containing drosperinone vs other progestins. The EURAS study, initiated in 2001 and including more than 120,000 COC users in Europe, found comparable risk of DVT among the 3 categories of progestins.[cxxviii] This study was not only large, but also of prospective study design and the primary objective was to assess safety across COC user groups. Two other studies have shown an increased risk of VTE in drosperinone users.[cxxix],[cxxx] These studies are limited by flawed methodologies in a similar fashion to the studies that caused controversies around the 3rd generation progestins. In one study[cxxxi] only 1.2% of COC users used drosperinone-containing OCPs, which results in unstable estimates.  In the other retrospective cohort study the relative risk for drosperinone-using women was elevated, but these one-year estimates are unreliable because of left censoring (women had varying risk-levels at entry to the study). The estimates of risk after the first year are in line with the EURAS study.

 

COC concerns for women with other risk factors:

Factor V Leiden mutation may independently increase the risk of DVT. The Factor V mutation occurs in 3-5% of Caucasians and is responsible for the majority of cases of venous thrombosis in which a mechanism is identifiable. A recent study suggested that the combination of third generation oral contraceptives and the Factor V Leiden mutation may increase the risk of DVT 30-50-fold.130 This study has been criticized for its lack of validation and methodology.

Other inherited thrombophilias, such as the prothrombin G20210A defect, and Protein S or Protein C deficiency, have been associated with an increased risk of VTE. Multiple studies have shown that this risk increases in the setting of OC use.

A recent retrospective cohort study of patients with a documented VTE showed that compared to non-users, OC use increases the risk of thrombosis in carriers of antithrombin, protein C and protein S defects six fold.131 Interestingly, risk of VTE in carriers of Factor V Leiden was not significantly increased.

Another investigator retrospectively analyzed OC exposure and incidence of VTE in thirteen female patients with the prothrombin G20210A defect (12 of which were heterozygote for the defect).[cxxxii] All thirteen women took OCs for an average of 10 years without any thrombotic complication. Interestingly, the homozygote took OCs with a 'third generation' progestin for 6 years without a thrombotic event. Another investigator noted that of those patients who develop VTE soon after initiating OCs (<6 months), most are thrombophilic.[cxxxiii] Among women with protein C or protein S deficiency, antithrombin deficiency, Factor V Leiden or prothrombin 20210A mutations, the risk of developing a DVT during the first year of OC use was increased 11-fold (95% CI, 2.1-57.3).[cxxxiv]

A pooled analysis of 8 case-control studies revealed that the odds ratio for VTE associated with OC use was 10.25 (95% CI, 5.59-18.45) in factor V Leiden carriers and 7.14 (95% CI, 3.39-15.04) in carriers of the prothrombin G20210A mutation.127 The crude odds ratios for VTE (not specifically analyzing the effect of OCs) were 4.9 (95%CI, 4.1-5.9) for factor V Leiden and 3.8 (95%CI, 3.0-4.9) for the prothrombin 20210A mutation.

Therefore, all healthy women who are diagnosed with a DVT while using oral contraceptives should be tested for possible Factor V Leiden mutation or Protein S or Protein C deficiency, although screening beforehand is not warranted.

Oral contraceptive use and risk of myocardial infarction: Myocardial infarction is a very rare event in non-smoking women of reproductive age. For women younger than 35 who do not smoke, the incidence of myocardial infarction is less than 1.7 per 100,000 woman years.113 This rate is notably higher between the ages of 40 and 45 years and is about 21 per 100,000 woman years. Review of the evidence shows that the association between current combined oral contraceptive use (containing 35 micrograms of ethinyl estradiol or less) and myocardial infarction is weak, with a relative risk ranging from 0.9 to 2.5. There is no evidence to support an increased or decreased risk of myocardial infarction due to past oral contraceptive use compared to no use.[cxxxv] Smoking has been identified as an independent risk factor for myocardial infarction; the combination of smoking and oral contraceptive use can be synergistic for increasing the risk of a MI.

The World Health Organization (WHO) has the following recommendations:135,[cxxxvi]

  1. Combined oral contraceptives can be use safely by women of any age who are non-smoking, normotensive, and non-diabetic.
  2. For women who smoke, and are 35 years or younger, oral contraceptives containing 35 micrograms or less are recommended.
  3. For women who smoke and are 35 years or older, oral contraceptive use is contraindicated.

Oral contraceptive use and risk of stroke: The link between high dose oral contraceptive pills and ischemic stroke has historically been determined with a strength of association ranging from 2.9 to 5.3.113 However, as dose of estrogen decreased, the odds ratio and relative risks in further studies all decreased as well. A summary of the data by the WHO123,124 concludes that there is no significant increased risk of ischemic stroke in women younger than 45 years old who use oral contraceptives. Overall, the strength of association between the use of lower dose oral contraceptives and stroke is weak, with odds ratios ranging from 1.1 to 1.8, with most 95% confidence intervals including 1.0.113 There is no consistent strong evidence linking oral contraceptive use to hemorrhagic stroke. Women of any age who have migraines with aura should not take combination oral contraceptive pills.[cxxxvii] Since smoking, hypertension, and migraine headaches all are independent risk factors for stroke, it must be concluded that women with other independent risk factors may have a slightly increased risk of stroke while taking the oral contraceptive pill. There is ample evidence, however, to suggest that there is no significant increase in ischemic or hemorrhagic stroke in OC-using women with no other risk factors. Because the baseline risk of stroke is rare in reproductive aged women, the attributable risk of oral contraceptives is quite small. In summary, evidence shows that current low dose oral contraceptives are safe with regard to vascular disease for a great majority of healthy non-smoking women who seek an effective contraceptive method.

NEW DEVELOPMENTS IN COMBINED ORAL CONTRACEPTIVE PILLS

The number of combined oral contraceptive pill preparations on the market has increased in recent years. There are new formulations of pills with a dosage of estrogen as low as 20 micrograms. Some physicians use the 'Quick Start' approach to pill initiation: this allows for immediate ingestion of the first dose of the pill in the office after a negative pregnancy test regardless of menstrual cycle day. A clinical trial has shown that is approach is safe, and results in higher ultimate rate of pill use than the conventional approach that initiates pill use after the onset of menses.[cxxxviii] In general, the efficacy, side effects and cycle control of these new preparations are similar to those with 35 micrograms of estrogen. The "lower" dose pills offer the theoretical advantage of less estrogen and therefore fewer estrogen side effects and medical complications. It remains to be determined if the new "lower" dose pills confer the same non-contraceptive benefits of the "higher" dose pills. A recent Cochrane review found that there is no difference in contraceptive effectiveness for lower dose pills. Compared to the higher-estrogen pills, several COCs containing 20 μg EE resulted in higher rates of early clinical trial discontinuation (overall and due to adverse events such as irregular bleeding) as well as increased risk of bleeding disturbances (both amenorrhea or infrequent bleeding and irregular, prolonged, frequent bleeding, or breakthrough bleeding or spotting). So, while COCs containing 20 μg EE may be theoretically safer, this has not been proven and low-dose estrogen COCs have in higher rates of bleeding pattern disruptions.[cxxxix]

An oral contraceptive containing 30mcg ethinyl estradiol and 3mg drospirenone works in a manner similar to other oral contraceptives, effectively inhibiting ovulation and producing cervical mucus that is hostile to sperm motility. Unlike other progestins used in oral contraceptives, drospirenone is an analogue to spironolactone and has biochemical and pharmacologic profiles similar to endogenous progesterone.[cxl] Drospirenone has both anti-mineralocorticoid and anti-androgenic activity. Its anti-androgenic activity may leads to suppression of undesired symptoms such as acne and hirsutism. Its anti-mineralocorticoid activity balances the aldosterone-stimulating effects of estrogen, thereby potentially reducing water-retention and weight gain.

Another preparation offers a hormone-free, placebo length of only 2 days. Twenty-one days of 20mcg ethinyl estradiol and 150mcg of desogestrel is followed by 2 days of placebo and 5 days of 10mcg ethinyl estradiol. Within 18 months of use, absence of withdrawal bleeding and intermenstrual bleeding have been reported to occur in 5.5% and 12% of total cycles, respectively.[cxli] Nearly three-quarters of participants in a large, open-label study reported one or more side effects, including headache, breast pain, dysmenorrhea, and menstrual irregularities.

 

Psychological/behavioral effects of hormonal contraception: Data from randomized controlled trials fail to support the assertion that combined oral contraceptives cause adverse psychological symptoms. One randomized double-blind study of 462 women looked at the percentage of traditionally “hormone-related” side effects in a 6-month comparison of COC versus placebo pill users. Symptoms of emotional lability and physical symptoms of headache, nausea, breast pain, abdominal bloating, back pain, weight gain, and decreased libido were studied, and there were no differences in the incidence of these symptoms between the COC and placebo groups.[cxlii] In a 2002 review of prospective, controlled studies of the effect of COCs on mood, four studies found no significant group differences in negative affect across the entire menstrual cycle, one study found that COC users reported less negative affect across the cycle, one study found higher negative affect throughout the cycle of a monophasic but not a triphasic COC, and two studies found that COC users experienced higher positive affect.[cxliii] Common to all the studies of the psychological effects of COCs was a beneficial outcome: there was less variability in negative affect, and less negative affect during menstruation in patients taking COCs.

Many studies have focused on the role of the progestin as a contributor to dysphoric mood. Two studies have shown worsening of mood with a higher progestin to estrogen ratio.[cxliv],[cxlv] Thus, switching to a formulation with a lower progestin to estrogen ratio may improve mood in women who have negative mood symptoms with COCs. However, evidence regarding mood disturbances with COCs is sparse.

 

ALTERNATIVE METHODS

Effective, safe contraception is achieved with combination estrogen and progestin delivery via a contraceptive skin patch or a vaginal ring. The general mechanism of action of these methods is similar to that of combined oral contraceptives. These methods offer the advantage of non-daily administration, relative ease of administration, potential greater compliance and thus potential greater efficacy.

Transdermal Patch

The once-weekly contraceptive patch delivers 150mcg of norelgestromin, the active metabolite of norgestimate, and 20mcg of ethinyl estradiol daily to the systemic circulation.[cxlvi] Ten percent of American women have used or currently used the patch.[cxlvii] Typical use includes placement of the patch on the same day of each week for 3 consecutive weeks followed by a patch-free week. Serum levels of the estrogen and progestin components are maintained for 2 days beyond the recommended 7 days of wear. Therefore, patients do not have to change the patch at the exact same time each week.

The patch is composed of 3 layers: an outer protective layer of polyester, a medicated, adhesive middle layer, and a clear, polyester liner that is removed before patch application. Patients can maintain normal activity, including bathing, swimming and heavy exercise while using the patch. It is recommended that wearers avoid use of oils, creams or cosmetics that may interfere with adhesion of the patch.

Compared to OCs with 250mcg norgestimate and 35mcg ethinyl estradiol, the patch suppresses ovulation to a similar degree[cxlviii]. It is as effective as oral levonorgestrel/ethinyl estradiol in altering cervical mucus and in providing cycle control. The overall and method-failure probabilities of the transdermal patch (through 13 cycles) are 0.7% and 0.4%, respectively.146 Perfect compliance (21 days of consecutive dosing followed by 7 days of no medication) was achieved in 90% of patients in the above study. However, efficacy trials of the patch have shown that participants less than 20 years age were less likely to use the patch correctly as compared with the pill.[cxlix] The noncontraceptive effects of transdermal administration have not yet been studied, but are expected to be similar to the combined oral contraceptive pill. A recent clinical trial did not demonstrate the same improved continuation rates with the "quick start" method in patch users as was seen in oral contraceptive users.[cl]

Vaginal Ring

For numerous decades, the vagina has been identified as a potential organ for drug absorption.[cli],[clii] The anatomy of the vagina allows for the easy placement of a ring to achieve this purpose.

A combination estrogen/progestin vaginal ring was approved for use in 2001 and was used by 6% of US women of reproductive age by 2006-2010.147 It is a flexible transparent circular tube, 54 mm (2 inches) in diameter and 4 mm (1/4 inch) thick. The ring is made of ethylene vinyl acetate polymer and contains a hormone reservoir that releases 0.120 μg of etonogestrel and 0.015 μg of ethinyl estradiol each day over a three-week period.[cliii] Hormone content in the ring is sufficient to provide a "grace period" of at least 14 additional days,[cliv] so woman can leave it in place for a full month and then immediately replace it to avoid menstruation, if they so desire. Etonogestrel, also called 3-ketodesogestrel, is also a synthethormone. It is the active metabolite of desogestrel, the progestin component of commonly used oral contraceptives.

The mechanism of action of a vaginal ring is similar to other hormonal contraceptives. Initiating ring use during the first five days of a normal cycle ensures that ovulation in that cycle is suppressed. Similarly, allowing no more than seven ring-free days each month, and making sure that the ring is in place continuously, with no more than three hours out in one day are also important for efficacy.153 Overall pregnancy rates are reported to be 0.65 per 100 woman-years (all first-year users).103 This level of effectiveness is similar to that found for women using combined oral contraceptives. Adherence to rules for ring use was very high, with consistent and correct use reported in 90.8% of all cycles. Women using the ring also reported good cycle control, with expected withdrawal bleeding in 98% of cycles, and bleeding at other times in only 6.4% of cycles.155

The ring can be placed in any position in the vagina that is comfortable. A total of 8% of women note the sensation of the ring in the vagina. If the ring is removed for intercourse it can be cleaned with water and must be replaced within three hours. Risks and adverse reactions possible with use of combined hormonal oral contraceptives also are likely to apply to the vaginal contraceptive ring. The ring does not prevent against sexually transmitted disease. Some women using the ring experienced side effects related to the device itself including vaginal discomfort or problems during intercourse, vaginal discharge, or vaginitis. These device-related problems were reported by 2-5% of women.[clv] Overall acceptability and tolerability of the ring were very high in the clinical trials performed to date.

 

EMERGENCY CONTRACEPTION

Emergency contraception prevents pregnancy after unprotected sexual intercourse. Emergency contraception (EC) does not protect against sexually transmitted infections. The emergency contraceptive formulations available in the United States include the CuT380A IUD,[clvi],[clvii],[clviii] ulipristal acetate (UPA) selective progesterone receptor modulator, and 150 mcg of levonorgestrel (Plan B and Plan B One-Step). Combined oral contraceptive tablets can also be used by following the “Yuzpe regimen,” which can be found on the internet for various pill formulations.

The Copper “T” IUD can be inserted up to 7 days after unprotected intercourse and is ideal for women who desire long-term contraception going forward.[clix]  It is 99% effective for emergency contraception.

The current treatment schedule for emergency contraceptive pills (ECPs) is one dose within 120 hours of unprotected intercourse.158 Ulipristal acetate is more effective than levonorgestrel for emergency contraception, especially in obese women.  Estimates of effectiveness range from 62-85%.[clx] Effectiveness is sustained throughout the 120 hours after unprotected intercourse. Combined and progestin-only ECPs reduce the risk of pregnancy by about 75-88%.[clxi] Effectiveness declines with increasing delay between unprotected intercourse and initiation of treatment.[clxii]

Mechanism of action: Oral emergency contraception likely inhibits or delays ovulation.[clxiii] Some investigators have shown histological alterations in the endometrium suggesting impairment of endometrial receptivity to implantation[clxiv] while others have found no such effects.[clxv],[clxvi] Other possible mechanisms include interference with corpus luteum function, thickening of cervical mucus, and alterations in tubal transport of sperm, egg, or embryo.[clxvii] Emergency contraceptives do not interrupt an already established pregnancy.  The Copper “T” IUD does not have an established mechanism of action as an emergency contraceptive but likely creates a hostile environment for sperm as well as for the egg and possibly an embryo.

Progestin-only emergency contraceptives are more effective and associated with significantly less nausea and vomiting than combined emergency contraceptive pills.[clxviii] The only absolute contraindication to the use of emergency contraceptive pills is a confirmed pregnancy. The absence of contraindications is likely due to the very short duration of exposure and low total hormone content. There are no conclusive studies of women who were already pregnant when they took emergency contraceptive pills or of women pregnant after failed emergency contraception. However, there is no epidemiologic evidence that progestins are teratogenic and observational studies provide reassurance regarding birth defects.[clxix]

The FDA has granted approval for the over the counter use of progestin-only EC for prevention of pregnancy. There are no longer any restrictions on age or gender for purchasing progestin-only EC.  Evidence has been reported that making ECPs widely available does not increase risk-taking behavior or increase the incidence of unintended pregnancy.[clxx] Additionally, it has been demonstrated that women most likely to seek emergency contraception are those already concerned about or using contraception.[clxxi],[clxxii]

Copper IUDs can be inserted up to five to seven days after ovulation to prevent pregnancy. Insertion of a Copper IUD is significantly more effective than the use of hormonal emergency contraception. The use of a Copper IUD can reduce the risk of unintended pregnancy by more than 99%.[clxxiii]  Furthermore, 86% of parous and 80% of nulliparous women retain the IUD after insertion for emergency contraception.162

 

BARRIER METHODS

Multiple forms of barrier methods are currently used: male and female condoms, the diaphragm, the contraceptive sponge, and cervical cap. Vaginal barriers are easy to use and are non-invasive. They can be used with little advance planning. Consistent and correct uses are absolutely essential for barrier effectiveness; most failures occur due to improper or inconsistent use. The 'typical use' pregnancy rate for these methods can be as high as 20-30%. The male condom has a 'typical use' failure rate of 14%. Recent studies have shown that teens were most likely to use condoms for birth control and 66% used a condom when they became sexually active.173

Mechanism of action: Both the male and female condoms provide a physical barrier that prevents sperm and egg interaction. They are intended for one time use only. Condoms also provide some protection against HIV and STI.

Diaphragms and cervical caps use two different mechanisms, a physical barrier as well as a spermicidal chemical. They are available by prescription only, and must be sized by a health professional for a proper fit. They are always used with spermicidal agents. Diaphragm provides protection for 6 hours and cervical cap for 48 hours after insertion.

The contraceptive sponge is a disc shaped polyurethane device and contains a 1,000 mg of nonoxynol-9. It does not require prescription, and provides protection for up to 24 hours after insertion. The typical use pregnancy rate for this method is 10-40%.

Vaginal barriers have many advantages: protection against sexually transmitted infections, immediate protection without much prior planning, easy access, and no systemic side effects. Disadvantages include: discomfort with placement and use, possible latex allergy (for condoms and the orthoflex diaphragm), increased incidence of urinary tract infections and bacterial vaginosis; and associations have been reported with toxic shock syndrome. In addition, a health care provider may be required to do the initial fitting for diaphragms, necessitating an extra visit to the physician's office. A comparison of the ability of contraceptive methods to reduce sexually transmitted disease is found in Table 6.

Table 6. Contraceptive Methods and STD Protection

 

Contraceptive Method

 

 

Effect on Reproductive Tract

 

 

Effect on Bacterial STDs

 

 

Effect on Viral STDs

 

 

Diaphragm,

Cervical cap,

Sponge

 

 

Reduces risk of PID; associated with vaginal and urinary infections

 

 

Some protection against cervicitis; increases organisms associated with bacterial vaginosis, candidiasis and urinary tract infections

 

 

No protection against vaginal infection or external genitalia transmission; prevention of HPV controversial. No protection against HIV

 

 

Female condom

 

 

Occasional local irritation

 

 

In vivo protection against recurrent trichomonal infections suggests possible protection against other STDs

 

 

In vitro impermeability to HIV, cytomegalovirus

 

 

IUD

 

 

Foreign body reaction within the uterus;

 

 

Copper IUD: No protection

 

LNG-IUS: associated with decreased upper-genital tract infection

 

 

No protection

 

 

Latex male condom

 

 

Occasional latex allergy

 

 

Protection against most pathogens in genital fluids

 

 

Less protection against organisms transmitted from external genitalia (HSV and HPV)

 

 

Combination oral contraceptive

 

 

Increased cervical ectopy; decreased risk of symptomatic PID requiring hospitalization

 

 

No protection against bacterial STDs; possible increase in cervical chlamydia

 

 

Data on HIV transmission risks conflicting; role regarding risk of HPV infection and cervical dysplasia unclear

 

 

DMPA/ Implants

 

 

Atrophic endometrium; thickening of cervical mucus

 

 

Assume no protection

 

 

May promote HIV transmission

 

 

Spermicide with nonoxynol-9

 

 

Risk of chemical irritation of vaginal epithelium/alteration of the vaginal flora with high doses

 

 

Equivocal

 

 

Data suggests increased HIV transmission risk that is dose and frequency dependent

 

 

Tubal ligation

 

 

Changes associated with surgery

 

 

No protection

 

 

No protection

 

 

Contraceptive Vaginal rings

 

 

Increased Vaginal discharge in some users

 

 

No protection

 

 

No protection

 

Barriers

Spermicides

Spermicides can be purchased without a physician's prescription in supermarkets and pharmacies. They can be used alone but are often used in conjunction with a vaginal barrier method (diaphragm, sponge, or cap). Nonoxynol-9 (N-9), the most commonly used spermicide, is an agent that destroys the sperm cell membrane, thereby immobilizing sperm. But, recent studies have shown that N-9 does not protect against STIs and HIV.[clxxiv] Spermicidal formulations include gels, creams, suppositories, film and male condoms. Pregnancy rates among typical users range from 5% to 30% in the first year of use.19 Methodology in determining these rates has not been consistent leading to skepticism of much of the data. Like the barrier methods, the effectiveness of spermicides is dependent on their consistent and correct use. Its advantages are similar to barrier methods of contraception.

Microbicides

Efforts to combine effective contraception and protection against HIV and STI transmission are of the utmost priority because the incidence of transmission of HIV and STIs is greatest in women of reproductive age.[clxxv]

Any substance that can reduce the transmission of HIV and STI when applied to the vagina is considered a microbicide.  A randomized double-blind, placebo controlled trial studied tenofovir vaginal gel in 889 women in South Africa.  The antiretroviral tenofovir gel is applied to the vagina sometime within 12 hours prior to sex and then again within 12 hours after sex.  This study showed that it cut HIV transmission by approximately 39%.  Even more encouraging were those women who were “high adherers” meaning they reported and demonstrated using the gel with >80% of each act of vaginal intercourse.  This subcategory of women showed a 54% decrease in HIV infection compared to placebo. In summary HIV incidence in the tenofovir arm was 5.6 per 100 women years vs. 9.1 per 100 women-years in the placebo arm.[clxxvi]  While this is promising, real world adherence is low, thus promoting the search for alternative delivery mechanisms, such as vaginal rings.[clxxvii]  As many as 60 potential compounds are currently under development. These formulations will probably be used as an adjunct to condoms, but may be used as primary protection for those who are unable or unwilling to use condoms consistently. These microbicides will work by either killing or immobilizing pathogens possibly by forming a barrier between pathogen and vaginal tissues, preventing the infection from entering target cells, preventing a pathogen from replicating once it has entered cells, by boosting the vagina's or rectum's own defense system or by acting like invisible condoms. The most desirable qualities of a new formula microbicide would be that it is applicable hours before sexual intercourse, it is not messy or "leaky," and spreads rapidly and evenly over the vagina and cervix.148

Some compounds may increase the host natural defenses against certain sexually transmitted pathogens by maintaining the normal acidic pH of the vagina in the presence of semen. They contain lactobacillus which naturally resides in the human vagina and produces hydrogen peroxide to kill HIV and STDs. Examples are: Lactobacillus suppositories, Buffer gel, and Acid gel (ACIDFORM).

Invisible Condoms: Thermoreversible Gel - Prevents infection by forming a protective barrier after being inserted into the vagina or rectum. It is a liquid at room temperature and quickly turns into an impermeable gel inside the rectal/vaginal canal.

NATURAL FAMILY PLANNING METHODS:

Abstinence: Couples who avoid sexual intercourse are practicing abstinence. Effectiveness for preventing pregnancy is 100%, but HIV and STIs may spread through the oral or rectal mucus membrane.

Coitus Interruptus: Also known as the withdrawal method, coitus interruptus entails withdrawal of the penis from the vagina (and external genitalia) immediately prior to ejaculation. Effectiveness depends largely on the man's ability to withdraw prior to ejaculation. Its actual efficacy is difficult to measure but the probability of pregnancy among perfect users is estimated to be approximately 4% in the initial year of use.[clxxviii]

Fertility Awareness: Symptothermic method - Natural family planning methods use the signs, symptoms, and timing of a normal menstrual cycle to avoid intercourse during fertile intervals. These methods are effective because of periodic abstinence during the fertile period of a woman's menstrual cycle. The fertile period of a woman's menstrual cycle can be determined by using cycle beads, a calendar, measuring basal body temperature and monitoring cervical secretions.178

Calendar Method: Estimating the fertile period during each menstrual cycle is based on 3 assumptions: (1) ovulation occurs on day 14 (±2 days) before the onset of the next menstrual flow, (2) the ovum survives for approximately 24 hours, and (3) sperm remain viable up to 5 days. Past cycle lengths give an estimate of fertile days within a given cycle. Avoidance of pregnancy is achieved by abstinence beginning about 5 days before and ending nearly 5 days after ovulation.

Basal Body Temperature: Most ovulatory cycles demonstrate a biphasic temperature pattern with lower temperatures in the first half of the cycle and higher temperatures beginning at the time of ovulation and continuing for the remainder of the cycle. Because this method does not adequately predict ovulation in advance, couples are instructed to abstain from intercourse or use a barrier method of contraception for the first half of the menstrual cycle until at least 2 days after a rise in temperature signifying ovulation.

Cervical Secretions: Changes in the character of cervical mucus can signify the fertile period of a woman's menstrual cycle. Cervical mucus that is abundant, clear or white, stretchy, and slippery represents the fertile period. Ovulation most likely occurs within 1 day of the appearance of cervical mucus that is abundant, stretchy and slippery. After ovulation, cervical secretions appear thick, cloudy and sticky. Couples are counseled to avoid intercourse when cervical secretions are first noted until 4 days after the peak of clear and slippery cervical mucus.

Couples can also use a combination of natural family planning methods (i.e., basal body temperature measurements and cervical secretion monitoring) to further avoid an undesired pregnancy. In addition, recent advances in home hormonal detection kits allow for detection of ovulation and better timing of abstinence for avoidance of pregnancy.

Lactation Amenorrhea Method (LAM) - Breastfeeding: Breastfeeding provides more than 98% protection from pregnancy in the first 6 months after birth.[clxxix] This method of contraception requires complete or nearly complete infant dependence on breast milk so that frequent suckling (at least 6-10 times per day) prevents ovulation. High frequency of feeds, long duration of each feed, night feeds, and short intervals between breastfeeds delay the return of ovulation.[clxxx],[clxxxi] Infant suckling disrupts the pulsatile release of gonadotropin releasing hormone (GnRH) by the hypothalamus resulting in abnormal pulsatility of LH and subsequent anovulation.[clxxxii] Ovulation however can occur even in the absence of menstruation. The probability of ovulation occurring before menstruation increases with time after delivery; the probability that women will ovulate before the resumption of menses increases from 33-45% during the first 3 months after delivery to 87-100% more than 12 months from delivery.[clxxxiii] To maintain effective contraception, another method should be used as soon as menstruation resumes, the frequency or duration of breastfeeding is reduced, food (or bottle) supplements are introduced, or the duration since delivery is 6 months. This method provides no protection against sexually transmitted diseases, but may help reduce post partum bleeding.

FUTURE DEVELOPMENTS

The near future will bring improved modifications of current methods including barrier methods, hormonal methods and intrauterine devices. Combined oral contraceptive pills using estetrol for the estrogen component are being developed in Europe.  Phase II studies using a combination of estetrol and drospirinone in a 24/4 pill show a favorable bleeding profile and have high user satisfaction.[clxxxiv],[clxxxv]  The next generation of contraceptives will likely be focused on new mechanisms of action. Targets include all aspects of the female and male reproductive system from gamete development to gamete delivery, fertilization and implantation. The goal of these new methods will be the interruption of key components of the system allowing safe, effective contraception with a minimum of side effects. The challenge facing the development of these new methods is allowing modification or interruption of the reproductive system with a minimum of effect on other body systems. Specifically, an optimal method would provide reversible interruption of reproductive capacity of the male and/or female without affecting the endocrine system. This is particularly important in the male as secondary sexually characteristics, and even behavior, are closely linked to any change in the hormonal milieu. Other challenges include the development of simple and cost effective methods. The next breakthrough in contraceptives will be the identification of a key molecular aspect of reproduction. If a specific receptor can be identified, an antagonist can be developed. If a critical enzyme is identified, an inhibitor can be synthesized.

Areas of interest will be in disruption of ovulation with anti-progestins or blocking the action of matrix metaloproteinases needed for the physical breakdown of the follicle. Folliculogenesis may be blocked by specific FSH antagonists, or interruption of folliculogenesis at the local level by growth differentiation factor-9. Other possible targets include blocking resumption of meiosis in the oocyte, or blocking egg activation (which is necessary before an egg is capable of fertilization and to prevent polyspermic fertilization). Finally the specific steps necessary for implantation may be identified and selectively disrupted – and research on molecules involved in implantation and angiogenesis is currently being done in mouse models.

Male hormonal contraception: male hormonal contraception may come to light in the distant future. In particular, the identification of genes in various aspects of testicular steroidogenesis, spermatogenesis and sperm maturation may provide novel targets for fertility regulation. Progestins act on the hypothalamic pituitary axis and suppress spermatogenesis and production of the hormone testosterone. It has been hypothesized that progestin implants with long acting add-back testosterone injections, oral progestagens with testosterone injections, or oral desogestrol with testosterone implants may produce azoospermia without affecting the other physiological and biological effects of testosterone.[clxxxvi] Future work will determine if these targets can be useful in developing novel strategies for male contraception.

  1. Finer LB, Zolna MR. Declines in unintended pregnancy in the United States, 2008–2011. New England Journal of Medicine. 2016 Mar 3;374(9):843-52.
  2. Jerman J, Jones RK, Onda T. Characteristics of US abortion patients in 2014 and changes since 2008.
  3. Jones RK, Darroch JE, Henshaw SK. Contraceptive use among US women having abortions in 2000-2001. Perspect Sex Reprod Health 2002; 34:294-303.
  4. Finer, LK, and Henshaw SK: Unintended Pregnancy in the United States. Perspectives on Sexual and Reproductive Health, 2006, vol. 38, p 90-95.
  5. Alan Guttmacher Institute: Sharing Responsibility: Women, Society, and Abortion Worldwide. New York, NY, Alan Guttmacher Institute, 1999.
  6. Alan Guttmacher Institute: Sex and America's Teenagers. New York, NY, Alan Guttmacher Institute, 1994: p46.
  7. https://obamacarefacts.com/obamacare-birth-control/
  8. Schwingl PJ, Ory HW, Visness CW: Estimates of the Risk of Cardiovascular Death Attributable to Low Dose Oral Contraceptives in the United States. Am J Obstet Gynecol, 1999 (180) 241-249.
  9. Trussell J. Contraceptive failure in the United States.   2011 Jan 21; 83(5): 397-404.
  10. http://hcp.skyla-us.com/why-skyla/unintended-pregnancy.php
  11. Gemzell-Danielsson K, Schellschmidt I, Apter D. A randomized, phase II study describing the efficacy, bleeding profile, and safety of two low-dose levonorgestrel-releasing intrauterine contraceptive systems and Mirena. Fertility and sterility. 2012 Mar 31;97(3):616-22.
  12. Schindler AE, Campagnoli C, Druckmann R, Huber J, Pasqualini JR, Schweppe KW, Thijssen JH. Classification and pharmacology of progestins. Maturitas. 2003 Dec 10;46:7-16.
  13. Landgren BM, Diczfalusy E. Hormonal effects of the 300mcg norethisterone (NET. minipill, 1. Daily steroid levels in 43 subjects during a pretreatment cycle and during the second month of NET administration. Contraception 1980;21:87-113.
  14. Oberti C, Dabancens A, Garcia-Huidobro M, et al. Low dosage oral progestogens to control fertility. II. Morphologic modifications in the gonad and oviduct. Obstet Gynecol 1974;43:285-94.
  15. Kim-Bjorklund T, Lundgren BM, Johannisson E. Morphometric studies of the endometrium, the fallopian tube and the corpus luteum during contraception with the 300mcg norethisterone (NET) minipill. Contraception 1991;43:459-74.
  16. Martinez-Manautou J, Giner-Velasquez J, Cortes-Gallegos V, et al. Daily progestogen for contraception: a clinical study. Br Med J 1967;2:730-2.
  17. Barnhart KT, Devoto L, Pommer R, Sir-Pettermann T, Robinovic J, Segal S. Neuroendocrine Mechanism of Anovulation in Users of Contraceptive Subdermal Implant of Nomgestrol Acetate (Uniplant). Fertil Steril 1997;67:250-5.
  18. Speroff L, Fritz MA, editors. Clinical gynecologic endocrinology and infertility. lippincott Williams & wilkins; 2005.
  19. Contraceptive Technology. Robert A. Hatcher (ed). 20th Revised Edition, 2011. Bridging the Gap Communications.
  20. Kaunitz AM. Noncontraceptive health benefits of oral contraceptives. Rev Endocrin Metab Disorders. 2002;3:277-283.
  21. Ali M, Akin A, Bahamondes L, Brache V, Habib N, Landoulsi S, Hubacher D, WHO study group on subdermal contraceptive implants for women†. Extended use up to 5 years of the etonogestrel-releasing subdermal contraceptive implant: comparison to levonorgestrel-releasing subdermal implant. Human Reproduction. 2016 Oct 21;31(11):2491-8.
  22. McNicholas C, Swor E, Wan L, Peipert JF. Prolonged use of the etonogestrel implant and levonorgestrel intrauterine device: 2 years beyond Food and Drug Administration–approved duration. American Journal of Obstetrics and Gynecology. 2017 Jan 29.
  23. Power J, French R, Cowan F. Subdermal implantable contraception versus other forms of reversible contraceptives or other implants as effective methods for preventing pregnancy. Cochrane Database Syst Rev 2007;(3):CD001326
  24. McDonald-Mosley R, Burke A. Contraceptive Implants. Sem Repro Med. 2010; 28 (2): 110-117.
  25. Croxatto HB, Urbancsek J, Massai R, Coelingh Bennink H, van Beek A; Implanon Study Group. A multicentre efficacy and safety study of the single contraceptive implant Implanon. Hum Reprod 1999;14(4):976–981
  26. Funk S, Miller M, Mishell D, Archer D, Poindexter A, Schmidt J, Zapmpaglione E. Safety and efficacy of Implanon, a single-rod implantable contraceptive containing etonogestrel.  Contraception 2005 (71): 319-326.
  27. Beerthhuizen R, van BeekA, Massai R. Bone mineral density during long-term use of the progestogen ceontraceptive implant Implanon compared to a non-hormonal method of contraception. Hum Reprod. 2006; (15): 118-122.
  28. Hohmann H, Creinin M. The Contraceptive Implant. Clin Ob and Gyn. 2007; 50 (4): 907-917.
  29. dos Santos PD, Madden T, Omvig K, Peipert JF. Changes in body composition in women using long-acting reversible contraception. Contraception. 2017 Apr 30;95(4):382-9.
  30. Croxatto HB, Ortiz ME, Valdez E. IUD mechanism of action. In: Bardin CW, Mishell DR Jr, eds. Proceedings from the Fourth International Conference on IUDs. London:Butterworth-Heinemann, 1994:44-62.
  31. Winner, B., Peipert, J.F., Zhao, Q., et al. Effectiveness of long-acting reversible contraception. N Engl J Med 2012; 366; 1998-2007.
  32. WHO Scientific Group. Mechanism of action, safety and efficacy of intrauterine devices. Technical Report Series, No. 753. Geneva: World Health Organization, 1987.
  33. Alvarez F, Brache V, Fernandez E, et al. New insights on the mode of action of intrauterine contraceptive devices in women. Fertil Steril 1988;49:768-73.
  34. Ortiz ME, Croxatto HB. The mode of action of IUDs. Contraception 1987;36:37-53.
  35. Hubacher D, Lara Ricalde R, Taylor DJ, Guerra Infante F, Guzman Rodriguez R. Use of intrauterine device and the risk of infertility among nulligravid women. N Engl J Med 2001;345:561-567.
  36. Shelton JD. Risk of clinical pelvic inflammatory disease attributable to an intrauterine device. Lancet 2001;257:443.

 

  1. Chi I-C. Postpartum IUD insertion: timing, route, lactation, and uterine perforation. In: Bardin CW, Mishell DR Jr, eds. Proceedings from the Fourth International Conference on IUDs. London:Butterworth-Heinemann, 1994:219-27.
  2. White MK, Ory HW, Rooks JB, et al. Intrauterine device termination rates and the menstrual cycle day of insertion. Obstet Gynecol 1980;55:220-4.
  3. Rowe P, Farley T, Peregoudov A, Piaggio G, Boccard S, Landoulsi S, Meirik O. Safety and efficacy in parous women of a 52-mg levonorgestrel-medicated intrauterine device: a 7-year randomized comparative study with the TCu380A. Contraception. 2016 Jun 30;93(6):498-506.
  4. Lahteenmaki P, Rauramo I, Backman T. The levonorgestrel intrauterine system in contraception. Steroids 2000;65:693-7.
  5. Andersson K, Odlind V, Rybo G. Levornorgestrel-releasing and copper-releasing (Nova T) IUDs during 5 years of use: a randomized comparative trial. Contraception 1994;49:56-72.
  6. McKeage K, Lyseng-Williamson KA. Kyleena™(levonorgestrel-releasing intrauterine system) in contraception: a profile of its use. Drugs & Therapy Perspectives. 2017 May 1;33(5):202-7.
  7. http://hcp.skyla-us.com/about-skyla/#t-body
  8. Lyus, R., Lohr, P., Prager, S., Board of the Society of Family Planning. Use of the Mirena LNG-IUS and Paragard CuT380A intrauterine devices in nulliparous women Contraception 2010; 81: 367-371.
  9. Hubacher D, Grimes DA. Non contraceptive health benefits of intrauterine devices: A systematic review. Obstet Gynecol Surv 2002;57:120-8
  10. Mok M, Tarasova M, Mikhailov A. Use of a levonorgestrel releasing intrauterine system to treat bleeding related to uterine leiomyomas. Fert Steril 2003;79:1194-8
  11. Fedele L, Bianchi S, Raffaelli R, Portuese A, Dorta M. Treatment of adenomyosis associated menorrhagia with a levonorgestrel releasing intrauterine device. Fert Steril 1997;68:426-9
  12. Vercellini P, Frontino G, Giorgi OD, Aimi G, Zaina B, Crosignani PG. Comparison of levonorgestrel releasing intrauterine device versus expectant management after conservative surgery for management of endometriosis: a pilot study. Fert Steril 2002;80:305-9
  13. Hurskainen R, Teperi J, Rissanen P, Aalto AM, Grenman S, Kivela A et al. Clinical outcomes and costs with levonorgestrel releasing intrauterine system or hysterectomy for treatment of menorrhagia: Randomized trial 5 year follow up. JAMA 2004: 291:1456-1463.
  14. Monteiro I, Bahamondes L, Diaz J, Perrotti M, Petta C. Therapeutic use of levonorgestrel-releasing intrauterine system in women with menorrhagia: a pilot study (1). Contraception 2002;65(5):325-8.
  15. Raudaskoski T, Tapanainen J, Tomas E, Luotola H, Pekonen F, Ronni-Sivula H, et al. Intrauterine 10 microgram and 20 microgram levonorgestrel systems in postmenopausal women receiving oral oestrogen replacement therapy: clinical, endometrial and metabolic response. BJOG 2002;109(2):136-44.
  16. Zapata L, Whiteman M, Tepper N, Jamieson D, Marchbanks P, Curtis K. Intrauterine device use among women with uterine fibroids: a systematic review.  Contraception 2010; (82): 41-55.
  17. Naki M, Tekcan C, Ozcan N, Cebi M, Levonorgestrel-releasing intrauterine device insertion ameliorates leimyoma-dependent menorrhagia among women of reproductive age without a significant regression in the uterine and leiomyoma volumes. Fert Ster 2010; 94 (1): 371-374.
  18. Orbo, A., Arnes, M. Hancke, C. et al. Treatment results of endometrial hyperplasia after prospective D-score classification: a follow-up study comparing effect of LNG-IUD and oral progestins versus observation only. Gynecol Oncol 2008; 111; 68-73.

 

  1. World Health Organization Collaborative Study of Cardiovascular Disease and Steroid Contraception. Cardiovascular disease and use of oral and injectable progestogen-only contraceptives: results of an international, multicenter, case-control study. Contraception 1998;57:315-24.
  2. Mishell DR Jr. Pharmacokinetics of depot medroxyprogesterone acetate contraception. J Reprod Med. 1996;41(suppl 5):381-90.
  3. Cullins VE. Noncontraceptive benefits and therapeutic uses of depot-medroxyprogesterone acetate. J Reprod Med. 1996;41(suppl 5):428-33.
  4. Trussell J, Kost K. Contraceptive failure in the United States: A critical review of the literature. Stud Fam Plann. 1987;18:237-83.
  5. Kaunitz AM. Long-acting contraceptive options.Int J Fert. 1996;41:69-76.
  6. Sangi-Haghpeykar H, Poindexter AN III, Bateman L, et al. Experiences of injectable contraceptive users in an urban setting. Obstet Gynecol. 1996;88:227-33.
  7. Mainwaring R, Hales HA, Stevenson K, et al. Metabolic parameter, bleeding, and weight changes in U.S. women using progestin only contraceptives. Contraception. 1995;51:149-53.
  8. World Health Organization Task Force on Long-Acting Agents for Fertility Regulation. Clinical evaluation of the therapeutic effectiveness of ethinyl oestradiol and oestrone sulphate on prolonged bleeding in women using depot medroxyprogesterone acetate for contraception. Hum Reprod. 1996;11(suppl 2):1-13.
  9. Harel Z, Biro FM, Kollar LM. Depo-Provera in adolescents: effects of early second injection or prior oral contraception. J Adolesc Health. 1995;16:379-84.
  10. Kaunitz AM. Long-acting hormonal contraception: assessing impact on bone mineral density, weight, and mood. Int J Fert. 1999;44:110-7.
  11. Westhoff C, Truman C, Kalmuss D, et al. Depressive symptoms and Depo-Provera. Contraception. 1998;57:237-40.
  12. Westhoff C, Wieland D, Tiezzi L. Depression in users of medroxy-progesterone acetate. Contraception. 1995;51:351-4.
  13. Westhoff C. Depot Medroxyprogesterone acetate injection (Depo-Provera); a highly effective contraceptive option with proven long term safety. Contraception 2003;68:75-87.
  14. Kaunitz A. Current options for injectable contraception in the United States. Seminar in Reproductive Medicine 2001;19:331-336.
  15. Westhoff C. Depot medroxyprogesterone acetate contraception. Metabolic parameters and mood changes. J Reprod Med. 1996;41(suppl 5):401-6.
  16. Strom BL, Berlin JA, Weber AL, Norman SA, Bernstein L, Burkman RT et al. Absence of an effect of injectable and implantable progestin only contraceptive on subsequent risk of breast cancer. Contraception 2004;69:437-446.
  17. Kaunitz AM. Injectable contraception: New and existing options. Obstet Gynecol Clin North Am.2000;27:741-80.
  18. Petitti DB, Piaggio G, Mehta S, et al. Steroid hormone contraception and bone mineral density: a cross-sectional study in an international population: the WHO Study of Hormonal Contraception and Bone Health. Obstet Gynecol. 2000;95:736-44.
  19. Food and Drug Administration. Full Prescribing Information: Depo Provera Contraceptive Injection. Rev 10/2010. Accessed 2/7/2015. Available at:http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020246s036lbl.pdf.

 

  1. Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM. Injectable hormone contraception and bone density: results from a prospective study. Epidemiology. 2002;13:581-587.
  2. Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM. Change in bone mineral density among adolescent women using and discontinuing depot medroxyprogesterone acetate contraception. Arch Pediatr Adolesc Med. 2005;159:139-144.
  3. Lopez LM, Chen M, Mullins Long S, Curtis KM, Helmerhorst FM. Steroidal contraceptives and bone fractures in women: evidence from observational studies. The Cochrane Library. 2015 Jan 1.
  4. McCann MF, Potter LS. Progestin-only oral contraception: a comprehensive review. Contraception 1994;50(Suppl 1):S9-S195.
  5. Fu H, Darroch JE, Haas T, et al. Contraceptive failure rates: new estimates from the 1995 National Survey of Family Growth. Fam Plann Perspect 1999;31:58-63.
  6. WHO Task Force on Oral Contraceptives. A randomized, double-blind study of two combined and two progestogen-only oral contraceptives. Contraception 1982;25:243-52.
  7. Anderson FD, Hait H. A multicenter, randomized study of an extended cycle oral contraceptive. Contraception 2003;68:89-96.
  8. Schlesselman JJ. Net effect of oral contraceptive use on the risk of cancer in women in the United States. Obstet Gynecol 1995;85:793-801.
  9. The Cancer and Steroid Hormone Study of the Centers for Disease Control and the National Institute of Child Health and Human Development. The reduction in risk of ovarian cancer associated with oral contraceptive use. N Engl J Med 1987;316:650-5.
  10. Ness RB, Grisso JA, Cottreau C, et al. Factors related to inflammation of the ovarian epithelium and risk of ovarian cancer. Epidemiology 2000;11:111-7.
  11. Prentice RL, Thomas DB. On the epidemiology of oral contraceptives and disease. Adv Cancer Res 1987;49:285-401.
  12. Schlesselman JJ. Oral contraceptives and neoplasia of the uterine corpus. Contraception 1991;43:557-79.
  13. The Cancer and Steroid Hormone Study of the Centers for Disease Control and the National Institute of Child Health and Human Development. Combination oral contraceptive use and the risk of endometrial cancer. JAMA 1987;257:796-800.
  14. Mishell DR Jr. Noncontraceptive benefits of oral contraceptives. J Reprod Med 1993;38:1021-9.
  15. Peterson HB, Lee NC. The health effects of oral contraceptives: misperceptions, controversies, and continuing good news. Clin Obstet Gynecol 1989;32:339-55.
  16. Ory HW. The noncontraceptive health benefits from oral contraceptive use. Fam Plan Perspect 1982;14:182-4.
  17. Brinton LA, Vessey MP, Flavel R, et al. Risk factors for benign breast disease. Am J Epidemiol 1981;113:203-14
  18. Mishell DR Jr. Noncontraceptive benefits of oral contraceptives. Am J Obstet Gynecol 1982;142:809-16
  19. Kleerekoper M, Brienza RS, Schultz LR, et al. Oral contraceptive use may protect against low bone mass. Arch Intern Med 1991;151:1971-6.
  20. Michaelsson K, Baron JA, Farahmand BY, et al. Oral contraceptive use and risk of hip fracture: a case-control study. Lancet 1999;353:1481-4.
  21. Hall ML, Heavens J, Cullum ID, Ell PJ. The range of bone density in normal British women. Br J Radiol 1990;63:266-9.
  22. Mazess RB, Barden HS. Bone density in premenopausal women: effects of age, dietary intake, physical activity, smoking and birth control pills. Am J Clin Nutr 1991;53:132-42.
  23. DeCherney A. Bone-sparing properties of oral contraceptives. Am J Obstet Gynecol 1996;174:15-20.
  24. Lanes SF, Birmann B, Walker AM, Singer S. Oral contraceptive type and functional ovarian cysts. Am J Obstet Gynecol 1992;166:956-61.
  25. Holt VL, Daling JR, McKnight B, et al. Functional ovarian cysts in relation to the use of monophasic and triphasic oral contraceptives. Obstet Gynecol 1992;79:529-33.
  26. Fernandez E, La Vecchia C, Balducci A, et al. Oral contraceptives and colorectal cancer risk: a meta-analysis. Br J Cacner 2001;84:722-7.
  27. Martinez ME, Grodstein F, Giovannucci E, et al. A prospective study of reproductive factors, oral contraceptive use, and risk of colorectal cancer. Cancer Epidemiol Biomark Prev 1997;6:1-5.
  28. Davis A, Lippman J, Godwin A, et al. Triphasic norgestimate/ethinyl estradiol oral contraceptive for the treatment of dysfunctional bleeding. Obstet Gynecol 2000;95:S84.
  29. van Hooff MH, Hirasing RA, Kaptein MB, et al. The use of oral contraceptives by adolescents for contraception, menstrual problems, or acne. Acta Obstet Gynecol Scand 1998;77:898-904.
  30. Redmond GP, Olson WH, Lippman JS, et al. Norgestimate and ethinyl estradiol in the treatment of acne vulgaris: a randomized, placebo-controlled trial. Obstet Gynecol 1997;89:615-22.
  31. Lucky AW, Henderson TA, Olsen WH, et al. Effectiveness of norgestimate and ethinyl estradiol in treating moderate acne vulgaris. J Am Acad Dermatol m1997;37:746-54.
  32. Thorneycroft H, Gollnick H, Schellschmidt I. Superiority of a combined contraceptive containing drospirenone to a triphasic preparation containing norgestimate in acen treatment. Cutis 2004; 74: 123-30.
  33. Haider A, Shaw JC. Treatment of Acne Vulgaris. JAMA 2004; 292:726-735
  34. Thiboutot D, Archer DF, Lemay A, et al. A randomized controlled trial of a low dose contraceptive containing 20microg of ethinyl estradiol and 100 microg of levonorgestrel for acne treatment. Fertil Steril 2001; 76:461-468
  35. Victory R, D'Souza C, Diamond MP, McNeeley SG, Vista Dect D, Hendrix, S. Adverse cardiovascular disease outcomes are reduced in women with history of oral contraceptive use: results from the Women's Health Initiative Database. Fert Steril 2004;82(2):52-53
  36. Victory R, D'Souza C, Diamond MP, McNeeley SG, Vista Dect D, Hendrix, S. Reduced cancer risks in oral contraceptive users: Results from the Women's Health Initiative. Fert Steril 2004; 28(2): 104-5
  37. Centers for Disease Control and Prevention. U.S. Medical Eligibility for Contraceptive Use. MMWR 2010; 59 (No. RR-4): 1-86.
  38. ACOG practice bulletin. The use of hormonal contraception in women with co-existing medical conditions. Number 18, 2000, In J Gynaecol Obstet 2001;75:93-106.
  39. Inman WHM, Vessey MP, Westerholm B, et al: Thromboembolic Disease and the Steroidal Component of Oral Contraceptions - A Report of the Committee on Safety of Drugs. Brit Med J 2:203-209, 1970.
  40. Michel D: Oral Contraceptives and Cardiovascular Events: Summary and Application of Data. Inter J Fertil, 45(suppl 2):121-133,2000.
  41. Speroff L. Oral Contraceptive and Breast Cancer Risk: Summary and Application of Data. Inter J Fertil, 45(suppl 2):113-120, 2000.
  42. Collaborative Group on Hormonal Factors in Breast Cancer: Breast Cancer and Hormonal Contraceptives: Collaborative Reanalysis of Individual Data on 53,297 Women with and 100,239 Women without Breast Cancer from 54 Epidemiologic Studies. Lancet 347:1713-1727, 1996.
  43. Marchbanks PA, McDonald JA, Wilson HG, Folger SG, Mandel MG, Daling JR, et al. Oral contraceptives and the risk of breast cancer. N Engl J Med 2002;346:2025-32.
  44. Mørch LS, Skovlund CW, Hannaford PC, Iversen L, Fielding S, Lidegaard Ø. Contemporary Hormonal Contraception and the Risk of Breast Cancer. New England Journal of Medicine. 2017 Dec 7;377(23):2228-39.
  45. Iversen L, Sivasubramaniam S, Lee AJ, Fielding S, Hannaford PC. Lifetime cancer risk and combined oral contraceptives: the Royal College of General Practitioners’ Oral Contraception Study. American Journal of Obstetrics and Gynecology. 2017 Jun 30;216(6):580-e1.
  46. Oral Contraceptive and Liver Cancer. The Contraceptive Report Nov. 1997, vol. 8, #5.
  47. Sitruk-Ware R, Nath A. The use of newer progestins for contraception. Contraception. 2010 (82): 410-417.
  48. Jick H, Jick SS, Gurewich V, Myers MW, Dasilakis C: Risk of Idiopathic Cardiovascular Disease and of Non-fatal Venous Thromboembolism in Women Using Oral Contraceptives with Differing Progestagen Components. Lancet 346:1589-1593, 1995.
  49. Spitzer WO, Lewis MA, Heinemann LA, Thorogood M, McCrae KT: Third Generation Oral Contraceptives and Risk of Venous Thromboembolic Disorders: An International Case Study. Brit Med J 312:83-88, 1996.
  50. Collaborative Study of Cardiovascular Diseases and Steroid Hormone Contraception. Venous Thromboembolic Disease and Combined Oral Contraceptives: Results of International Multicentre Case-Controlled Study. Lancet 346:1575-1582, 1995.
  51. Farmer RDT, Lawrenson RA, Thompson CR, Kennedy JG, Hambleton IR: Population-based Study of Risk of Venous Thromboembolism Associated with Various Oral Contraceptives. Lancet 349:83-88, 1997.
  52. Hennessey S, Berlin JA, Kinman JL, Margolis DJ, Marcus SM, Strom BL. Risk of venous thromboembolism from oral contraceptives containing gestodene and desogestrel versus levonorgestrel: a meta-analysis and formal sensitivity analysis. Contraception 2001;62(2):125-33.
  53. Kemmeren JM, Algra A, Grobbee DE. Third generation oral contraceptives and risk of venous thrombosis: meta-analysis. BMJ 2001;323:1-9.
  54. Lidegaard O, Edstrom B, Kreiner S. Oral contraceptives and venous thromboembolism: a five-year national case-control study. Contraception 2002;65(3):187-96.
  55. Dinger JC, Heinemann LA, Kuhl-Habich D. The safety of a drospirenone-containing oral contraceptive: final results from the European Active Surveillance Study on oral contraceptives based on 142,475 women-years of observation. Contraception 2007;75:344-54.
  56. Lidegaard O, Løkkegaard E, Svendsen AL, Agger C. Hormonal contraception and risk of venous thromboembolism: national follow-up study. BMJ 2009;339:b2890.
  57. Vandenbrouke JP, Koster T, Breit E, Reitsma PH, Bertina RM, Rosendaal FR: Increased Risk of Venous Thrombosis in Oral Contraceptive Users Who are Carriers of Factor V Leiden Mutation. Lancet 344:1453-1457, 1994.
  58. A van Hylckama Vlieg, F M Helmerhorst, J P Vandenbroucke, C J M Doggen, F R Rosendaal. The venous thrombotic risk of oral contraceptives, effects of oestrogen dose and progestogen type: results of the MEGA case-control study. BMJ 2009;339:b2921.
  59. Girolami A, Tormene D, Simioni P, Zanon E. Long-tern use of oral contraceptive therapy in women with prothrombin 20210G-A polymorphism without thrombotic complications: a study of 13 women (12 heterozygotes and 1 homozygote). Thrombosis Res 2001;102:205-10.
  60. Bloemenkamp KW, Rosendaal FR, Helmerhorst FM, Vandenbroucke JP. Higher risk of venous thrombosis during early use of oral contraceptives in women with inherited clotting defects. Arch Intern Med 2000;160(1):49-52.
  61. Emmerich J, Rosendaal FR, Cataneo M, Margaglione M, De Stefano V, Cumming T, et al. Combined effect of factor V Leiden and prothrombin 20210A on the risk of venous thromboembolism-pooled analysis of 8 case-control studies including 2310 cases and 3204 controls. Study Group for Pooled-Analysis in Venous Thromboembolism. Thromb Haemost 2001;86(3):809-16.
  62. WHO Scientific Group on Cardiovascular Disease and Steroid Hormone Contraception: Cardiovascular Disease and Steroid Hormone Contraception: Report of the WHO Scientific Group. WHO Technical Report 877. Geneva, Switzerland, 1998.
  63. WHO Collaborative Study on Cardiovascular Disease and Steroid Hormone: Ischemic Stroke and Combined Oral Contraceptive: Results of an International Multicenter Case-Controlled Study. Lancet 348:498-509, 1996.
  64. Curtis KM, Mohllajee AP, Peterson HB. Use of combined oral contraceptives among women with migraine and nonmigrainous headaches: a systematic review. Contraception 2006;73:189–194.
  65. Westoff C, Kerns J, Morroni C, Cushman LF, Tiezzi L, Murphy PA. Quick Start: A novel oral contraceptive initiation method. Contraception 2002; 66:141-145.
  66. Gallo MF, Nanda K, Grimes DA, Lopez LM, Schulz KF. 20 µg versus >20 µg estrogen combined oral contraceptives for contraception. Cochrane Database of Systematic Reviews 2008, Issue 4. Art. No.: CD003989. DOI: 10.1002/14651858.CD003989.pub3
  67. Krattenmacher, R. Drospirenone: pharmacology and pharmacokinetics of a unique progestogen. Contraception 2000;62:29-38.
  68. An open-label, multi-center, noncomparative safety and efficacy study of Mircette, a low-dose estrogen-progestin oral contraceptive. The Mircette Study Group. Am J Obstet Gynecol 1998;179(1);S2-8.
  69. Redmond, G. et al. Use of placebo controls in an oral contraceptive trial: methodological issues and adverse event incidence. Contraception  60 (1999) 81–85.
  70. Oinonen, KA, Mazmanian D. To what extent do oral contraceptives influence mood and affect? J Affective Disorders 70 (2002) 229-240.
  71. Akerlund, M., Rode, A., Westergaard, J. Comparative profiles of reliability, cycle control and side effects of two oral contraceptive formulations containing 150 micrograms desogestrel and either 30 micrograms or 20 micrograms ethinyl oestradiol. Br J Obstet Gynaecol;l 1993;100 :832-838.
  72. Graham CA, Ramos R, Bancroft J, Maglaya C, Farley TM. The effects of steroidal contraceptives on the well-being and sexuality of women: a double-blind, placebo-controlled, two-centre study of combined and progestogen-only methods. Contraception 1995; 52:363-369.
  73. Creasy GW, Abrams LS, Fisher AC. Transdermal Contraception. Semin Reprod Med 2001;19(4):373-80.
  74. https://www.guttmacher.org/fact-sheet/contraceptive-use-united-states
  75. Smallwood GH, Meador ML, Lenihan JP, et al. Efficacy and safety of a transdermal contraceptive system. Obstet Gynecol 2001;98:799-805.
  76. Archer DF, Bigrigg, Smallwood GH. Assessment of compliance with a contraceptive patch (Ortho Evra TM/Evra TM) among North American women. Fertil Steril 2002;77:S27-S31
  77. Murthy AS, Creinin MD, Harwood BJ, Schreiber CA. Same day initiation of the transdermal hormonal delivery system (contraceptive patch) versus traditional initiation methods. Contraception 2005;72:333-6
  78. Macht DI.The absorption of drugs and poisons through the vagina. J Pharmacol 10:509.
  79. Barnhart K, Shalaby W: The Vagina: Physiologic Characteristics Important to Formulators of Microbicides. In: Rencher (ed): Vaginal Microbicide Formulations Workshop. Lippincott-Raven Publishers, 1998.
  80. Timmer CJ, Mulders TMT. Pharmacokinetics of etonogestrel and ethinylestradiol released from a combined contraceptive vaginal ring. Clin Pharmacokinet 2000; 39:233-242.
  81. Mulders TMT, Dieben, TOM. Use of the novel combined contraceptive vaginal ring NuvaRing for ovulation inhibition. Fertil Steril 2001; 75:865-870.
  82. Roumen FJME, Apter D, Mulders TMT, et al. Efficacy, tolerability and acceptability of a novel contraceptive vaginal ring releasing etonogestrel and ethiny oestradiol. Human Reprod 2001; 16:469-475.
  83. Van Look PFA, Stewart F: Emergency Contraception. In Hatcher RA, Trussell J, Stewart F, Katz W, Stewart GK, Guest F, Kowal D: Contraceptive Technology 17th Revised Edition, New York, NY, Ardent Media, 1998.
  84. Glasier A: Emergency Postcoital Contraception. NEJM 1997: 337; 1058-1064.
  85. Hatcher RA, Trussell J, Stewart F, Holelles S, Russell CR, Kowal D: Emergency Contraception: The Nations Best Kept Secret. Decatur GA: Bridging the Gap Communications, 1995.
  86. Zhou L, Xiao B. Emergency contraception with Multiload Cu-375 SL IUD: a multicenter clinical trial. Contraception 2001;64:107–12.
  87. Glasier, A., Cameron, S.T., Fine, P.M., et al. Ulipristal acetate versus levonorgestrel for emergency contraception: a randomized non-inferiority trial and meta-analysis. Lancet 2010; 375: 555-562.
  88. Trussell J, Rodriguez G, Ellertson C: New Estimates of the Effectiveness of the Yupze Regime of Emergency Contraception. Contraception 1998: 57; 363-369.
  89. Piggio G, von Hertzen H, Grimes DA, Van Look PFA: Timing of Emergency Contraception with Levonorgestrel or the Yuspe Regime. Lancet 1999: 353; 721.
  90. Swahn ML, Westlund P, Johansson E, Bygdmen M: Effect of Postcoital Contraceptive Methods on the Endometrium and the Menstrual Cycle. Acta Obstet Gynecol Scand 1996: 75; 738-744.
  91. Kubb AA, White JO, Guillebud J, Elder MG: The Biochemistry of Human Endometrium after Two Regimes of Postcoital Contraception: A DL-norgestrel/ethinyl estradiol combination or danazol? Fertil Steril 1986: 45; 512-516.
  92. Taskin O, Brown RW, Young DC, Poindexter AN, Wiehle RD: High Doses of Oral Contraceptives do not Alter Endometrial a1 and aVβ3 Integrins in the Late Implantation Window. Fertil Steril 1994: 61; 850-855.
  93. Raymond EG, Lovely LP, Chen-Mok M, Seppala M, Kurman RJ, Lessey BA: Effects of Yupze Regime of Emergency Contraception on Markers of Endometrial Receptivity. Human Reprod 2000: 15; 2351-2355.
  94. Ling WY, Wrixon W, Acorn T, Wilson E, Collins J: Mode of Action of DL-norgestrel and ethinyl estradiol combination in Postcoital Contraception. III Effect of Preovulatory Administration following the Luteinizing Hormone Surge of Ovarian Steroidogenesis. Fertil Steril 1983: 40; 631-636.

 

  1. Raymond EG. Creinin MD. Barnhart KT. Lovvorn AE. Rountree RW. Trussell J. Meclizine for prevention of nausea associated with use of emergency contraceptive pills: a randomized trial. Obstet Gynecol 2000, 95(2):271-7.
  2. Raman-Wilms L, Tseng AL, Wighardt S, Einarson TR, Koren G: Fetal Genital Effects of First Trimester Sex Hormone Exposure: A Meta-analysis. Obstet Gynecol 1995: 85; 141-149.
  3. Grimes DA. Emergency contraception: Politics trumps science at the US Food and Drug Administration. Obstet Gynecol 2004;104:220-221.
  4. Glasier A, Baird D: The Effects of Self-administering contraception. NEJM 1998: 339; 1-4.
  5. Kosunen E, Sihvo S, Hemminki E: Knowledge and Use of Hormonal Emergency Contraception in Finland. J Contracept 1997: 55; 153-157.
  6. Trussell J, Ellertoson C: Efficacy of Emergency Contraception. Fert Control Rev 1995: 4; 8-11.
  7. Ramjee G. Morar NS. Alary M. Mukenge-Tshibaka L. Vuylsteke B. Ettiegne-Traore V. Chandeying V. Karim SA. Van Damme L. COL 1492 study group. Challenges in the conduct of vaginal microbicide effectiveness trials in the developing world. AIDS. 2000;14(16):2553-7.
  8. Elias CJ, Leonard A., Family Planning and Sexually Transmitted Diseases: The need to enhance contraceptive choice. Curr Issues Public Health 1995; 1:191-99.
  9. Karim X, Karim S, Frohlich, Grobler A, Baxter C, Mansoor L, Kharsany A, Sibeko S, Mlisana K, Omar Z, Gengiah T, Maarschalk, Arulappan N, Mlotshwa, Morris L, Taylor D. Effectiveness and Safety of Tenofovir Gel, an Antiretroviral Microbicide, for the Prevention of HIV Infection in Women. Science.  2010 (329) 1168-1174.
  10. Notario-Pérez F, Ruiz-Caro R, Veiga-Ochoa MD. Historical development of vaginal microbicides to prevent sexual transmission of Hiv in women: from past failures to future hopes. Drug design, development and therapy. 2017;11:1767.
  11. Kowal D. Coitus Interruptus (Withdrawal). In: Hatcher RA, Trussell J (eds). Contraceptive Technology (17th Revised Edition). New York: Ardent Media, Inc., 1998:303-7.
  12. Labbok M, Cooney K, Coly S. Guidelines: breastfeeding, family planning, and the Lactational Amenorrhea Method - LAM. Washington, D.C.:Institute for Reproductive Health, Georgetown University, 1994.
  13. Campbell OMR, Gray RH. Characteristics and determinants of postpartum ovarian function in women in the United States. Curr Opin Obstet Gynecol 1993;169:55-60.
  14. Gray RH, Campbell OM, Apelo R, Eslami SS, Zacur H, Ramos RM, Gehret JC, Labbok MH. Risk of ovulation during lactation. Lancet 1990;335:25-9.
  15. McNeilly AS. Suckling and the control of gonadotropin secretion. In: Knobil E, Neill JD (eds). The physiology of reproduction (second edition). New York: Raven Press, 1994:1179-212.
  16. Lewis PR, Brown JB, Renfree MB, Short RV. The resumption of ovulation and menstruation in a well-nourished population of women breastfeeding for an extended period of time. Fertil Steril 1991;55:529-36.
  17. Apter D, Zimmerman Y, Beekman L, Mawet M, Maillard C, Foidart JM, Bennink HJ. Bleeding pattern and cycle control with estetrol-containing combined oral contraceptives: results from a phase II, randomised, dose-finding study (FIESTA). Contraception. 2016 Oct 31;94(4):366-73.
  18. Apter D, Zimmerman Y, Beekman L, Mawet M, Maillard C, Foidart JM, Coelingh Bennink HJ. Estetrol combined with drospirenone: an oral contraceptive with high acceptability, user satisfaction, well-being and favourable body weight control. The European Journal of Contraception & Reproductive Health Care. 2017 Jul 4;22(4):260-7.
  19. Brunk D. Many men would consider using hormonal methods of contraception: survey highlights experimental methods in gynecology. ObGyn News 2004.