Archives

Neonatal Hyperthyroidism

INTRODUCTION

 

Neonatal hyperthyroidism in most cases is transient and results from the transplacental passage of maternal stimulating TSH receptor antibodies (TRAb) known as neonatal Graves’ disease (GD).  Permanent non autoimmune neonatal hyperthyroidism is rare and is due to activating mutations of TSH receptor or due to somatic activating mutations in the stimulatory alpha subunit of the guanine nucleotide-binding protein (GNAS gene) in McCune-Albright syndrome. Exposure to topical iodine has also been reported as a rare cause of hyperthyroidism in newborns.

 

TRANSIENT NEONATAL HYPERTHYROIDISM

 

Neonatal Graves’ disease (GD) is usually a self-limited disease, but it can be life threatening and permanently damage the brain.  Neonatal GD is caused by transplacental passage of TSH receptor antibodies (TRAb) with stimulatory activity.

 

TRAb are Immunoglobulin of G class and freely cross the placenta. Different types of TRAb can be found: TRAb that bind to the TSH receptor and stimulates the production of thyroid hormones, (TSH receptor stimulating antibodies, TSI), TRAb that bind to the TSH receptor, do not stimulate the production of thyroid hormones and can block the binding of TSH (TSH receptor blocking antibodies TBI).

 

Hyperthyroidism develops only in babies born to mothers with the most potent stimulatory activity in serum. This corresponds to 1-2% of mothers with Graves’ disease, or 1 in 50,000 newborns, an incidence that is approximately four times higher than is that for transient neonatal hypothyroidism due to maternal TSH receptor blocking antibodies. Some mothers have mixtures of stimulating and blocking antibodies in their circulation, the relative proportion of which may change over time. Not surprisingly, the clinical picture in the fetus and neonate of these mothers is more complex and depends not only on the relative proportion of each activity in the maternal circulation at any one time but on the rate of their clearance from the neonatal circulation postpartum.

 

Occasionally, neonatal hyperthyroidism may even occur in infants born to hypothyroid mothers. A prospective study showed that 40% of patients treated for Graves’ disease with radioactive iodine had TRAb detectable after 5 years (13). In these situations, the maternal thyroid has been destroyed either by prior radioablation, surgery, or by coincident destructive autoimmune processes so that potent thyroid stimulating antibodies, present in the maternal circulation, are silent in contrast to the neonate whose thyroid gland is normal. Persistence of TRAb after thyroidectomy is higher in females with Graves’ ophthalmopathy or smokers. Fetal/neonatal thyrotoxicosis can occur also in newborn from hypothyroid mothers with chronic lymphocytic thyroiditis.

 

CLINICAL MANIFESTATIONS

 

TABLE 1. Situations That Should Prompt Consideration of Neonatal Hyperthyroidism

·       Unexplained tachycardia, goiter or stare

Unexplained petechiae, hyperbilirubinemia, or hepatosplenomegaly

·       History of persistently high TSH receptor antibody titer in mother during pregnancy

·       History of persistently high requirement for antithyroid medication in mother during pregnancy

·       History of thyroid ablation for hyperthyroidism in mother

·       History of previously affected sibling

 

Maternal TSH receptor antibody-mediated hyperthyroidism may present in utero. Fetal hyperthyroidism is suspected in the presence of fetal tachycardia (pulse greater than 160/min) especially if there is evidence of failure to thrive. Obstetric complications are common. Fetal goiter (fetal neck circumference >95%) can by monitored by ultrasound using nomograms for fetal thyroid growth. Fetal goiter can cause esophageal and/or tracheal obstructions and polyhydramnios. Fetal goiter can also be due to transplacental passage of antithyroid drugs that cause hypothyroidism in the fetus.

 

TABLE 2. Clinical Manifestations in the Fetus

Unexplained tachycardia,

Failure to thrive

Intrauterine growth retardation

Goiter

Advanced bone age

Prematurity

Craniosynostosis, microcephaly

Fetal death

 

In the neonate infant typically, the onset is during the first one two weeks of life but can occur by 45 days. This is due both to the clearance of maternally administered antithyroid drug (propylthiouracil- PTU, methimazole- MMI, or carbimazole) from the infant ’s circulation and to the increased conversion of T4 to the more metabolically active T3 after birth. Rarely, as noted earlier, the onset of neonatal hyperthyroidism may be delayed until later if higher affinity blocking antibodies are also present.

In the newborn infant, characteristic signs and symptoms include tachycardia, irritability, poor weight gain, and prominent eyes.  Goiter, when present, may be related to maternal antithyroid drug treatment as well as to the neonatal Graves’ disease itself.

 

Rarely, infants with neonatal Graves’ disease present with thrombocytopenia, jaundice, hepatosplenomegaly, and hypoprothrombinemia, a picture that may be confused with congenital infections such as toxoplasmosis, rubella, or cytomegalovirus. In addition, arrhythmias and cardiac failure may develop and may cause death, particularly if treatment is delayed or

inadequate. In addition to a significant mortality rate that approximates 20% in some older series, untreated fetal and neonatal hyperthyroidism is associated with deleterious long-term consequences, including premature closure of the cranial sutures (cranial synostosis), failure to

thrive, and developmental delay. The half-life of TSH receptor antibodies is 1 to 2 weeks. The duration of neonatal hyperthyroidism, a function of antibody potency and the rate of their metabolic clearance, is usually 2 to 3 months but may be longer.

 

TABLE 3. Clinical Manifestations in the Neonate

Irritability, hyperexcitability, sleep disorders

Tachycardia, hypertension, cardiac failure

Flushing, sweating

Respiratory distress, pulmonary hypertension

Goiter, stare

Feeding difficulties, increased appetite but no/poor weight gain

Diarrhea

Unexplained petechiae, hyperbilirubinemia, jaundice, or hepatosplenomegaly

Craniosynostosis, microcephaly,

Death

 

LABORATORY EVALUATION

 

The recent guidelines for management of hyperthyroidism and the updated guidelines for the management of thyroid disease during pregnancy released from the American Thyroid Association ATA both suggest determining TRAb levels in pregnant women with Graves’ disease at 18-22 weeks instead of 20-24 weeks of gestation because a severe case of fetal Graves’ disease has occurred at 18 weeks of pregnancy.

 

Because of the importance of early diagnosis and treatment, infants at risk for neonatal hyperthyroidism should undergo both clinical and biochemical assessment as soon as possible.

All neonates born from a woman with TRAb positivity in pregnancy should undergo determination of TRAb from cord blood at delivery. If TRAb is negative, the risk to neonatal hyperthyroidism is negligible (Sensitivity is around 100%). FT3, FT4 and TSH determination from cord blood did not predict neonatal hyperthyroidism. Increases in FT4 on day 3 to 5 seems to better indicate the onset of hyperthyroidism. Situations that should prompt consideration of neonatal hyperthyroidism are listed in Table 1. A high index of suspicion is necessary in babies of women who have had thyroid ablation because in them a high titer of TSH receptor antibodies would not be evident clinically. Similarly, women with persistently elevated TSH receptor antibodies and with a high requirement for antithyroid medication are at an increased risk of having an affected child.

 

The diagnosis of hyperthyroidism is confirmed by the demonstration of an increased concentration of circulating T4 (and free T4, and T3, if possible) accompanied by a suppressed TSH level in neonatal or fetal blood. Results should be compared with normal values during gestation. Fetal ultrasonography may be helpful in detecting the presence of a fetal goiter and in monitoring fetal growth. Demonstration in the baby or mother of a high titer of TSH receptor antibodies will confirm the etiology of the hyperthyroidism and, in babies whose thyroid function testing is normal initially, indicate the degree to which the baby is at risk.

 

 In general, babies likely to become hyperthyroid have the highest TSH receptor antibody titer but levels of maternal TRAb in the serum as low as 4.4 U/L has been associated with neonatal thyrotoxicosis.  If TSH receptor antibodies are not detectable, the baby is very unlikely to become hyperthyroid. In the latter case, it can be anticipated that the baby will be euthyroid, have transient hypothalamic-pituitary suppression, or have a transiently elevated TSH, depending on the relative contribution of maternal hyperthyroidism versus the effects of maternal antithyroid medication, respectively. Close follow up of all babies with abnormal thyroid function tests or detectable TSH receptor antibodies is mandatory.

 

THERAPY

 

In the fetus, treatment is accomplished by maternal administration of antithyroid medication. Until recently PTU was the preferred drug for pregnant women in North America, but current recommendations suggest the use of MMI rather than PTU after the first trimester because of concerns about potential PTU-induced hepatotoxicity. The goals of therapy are to utilize the minimal dosage necessary to normalize the fetal heart rate and render the mother euthyroid or slightly hyperthyroid. In the neonate MMI (0.25 to 1.0 mg/kg/day) has been used initially in 3 divided doses. If the hyperthyroidism is severe, a strong iodine solution (Lugol’s solution or SSKI, 1 drop every 8 hours) is added to block the release of thyroid hormone immediately. Often the effect of MMI is not as delayed in infants as it is in older children or adults, a consequence of decreased intrathyroidal thyroid hormone storage. Therapy with both antithyroid drug and iodine is adjusted subsequently, depending on the response. Propranolol (2 mg/kg/day in 2 or 3 divided doses) is added if sympathetic overstimulation is severe, particularly in the presence of pronounced tachycardia. If cardiac failure develops, treatment with digoxin should be initiated, and propranolol should be discontinued. Rarely, prednisone (2 mg/kg/day) is added for immediate inhibition of thyroid hormone secretion. Measurement of TSH receptor antibodies in treated babies may be helpful in predicting when antithyroid medication can be safely discontinued. Lactating mothers on antithyroid medication can continue nursing as long as the dosage of PTU or MMI does not exceed 400 mg or 40 mg, respectively. The milk/serum ratio of PTU is 1/10 that of MMI, a consequence of pH differences and increased protein binding, so one might anticipate less transmission to the infant, but concerns about potential PTU toxicity need to be considered. At higher dosages of antithyroid medication, close supervision of the infant is advisable. A review about management of neonates born to mothers with Graves’ disease has been recently published.

 

PERMANENT NEONATAL HYPERTHYROIDISM

 

Rarely, neonatal hyperthyroidism is permanent and is due to a germline mutation in the TSH receptor (TSH-R) resulting in its constitutive activation. A gain of function mutation of the TSH-R should be suspected if persistent neonatal hyperthyroidism occurs in the absence of detectable TSH-R antibodies in the maternal circulation. Prematurity, low birth weight, and advanced bone age are common. Most cases result from a mutation in exon 10 which encodes the transmembrane domain and intracytoplasmic tail of the TSH-R, a member of the G protein coupled receptor superfamily. Less frequently, a mutation encoding the extracellular domain has been described. An autosomal dominant inheritance has been noted in many of these infants; other cases have been sporadic, arising from a de novo mutation.

 

Early recognition is important because the thyroid function of affected infants is frequently difficult to manage medically and, when diagnosis and therapy is delayed, irreversible sequelae, such as cranial synostosis and developmental delay may result. For this reason, early, aggressive therapy with either thyroidectomy or even radioablation has been recommended.

 

Two clinical forms were described: the first one is the “familial non-autoimmune autosomal dominant hyperthyroidism” (FNAH). High variable age of manifestation from neonatal period to 60 years, with variability also within the same family is reported. Goiter is present in children, with nodules in older age. The second one is “Persistent sporadic congenital non autoimmune hyperthyroidism” (PSNAH) includes forms with sporadic (de novo) germline mutations in the TSH-R. PSNAH is characterized by fetal-neonatal onset or within 11 months and more severe hyperthyroidism requiring early aggressive therapy.

 

ThyroidfunctioninbabieswithagainoffunctionmutationoftheTSH receptormaybe difficult tomanagemedically and,whendiagnosis andtherapyisdelayed,irreversible sequelae,suchas cranial synostosis anddevelopmental delaymayresult.Thyroidablationmayberequired.Thyroidsurgeryis thepreferredapproachif anexperienced pediatric surgeonisavailable.Thetimingatwhichthyroidectomy canbeperformedwilldependoninstitutionalpreference.  Ifthis is notfeasible,thenradioablationmaybenecessary. Guidelines about this rare condition have recently been published.

 

MCCUNE ALBRIGHT SYNDROME

 

McCune Albright is a syndrome due to somatic activating mutations in Gsαgene and can rarely present with neonatal hyperthyroidism.

 

ACKNOWLEDGEMENTS

 

This chapter is, in part, based on the previous version written by Professor Rosalind Brown.

 

GUIDELINES

 

Ross DS, Burch HB, Cooper DS, Greenlee MC, Launberg P, Maia AL, Rivkees S, Samuels M, Sosa JA, Stan MN, Walter MA. 2016 American Thyroid Association Guidelines for Diagnosis and management of hyperthyroidism and other causes of thyrotoxicosis. Thyroid. 2016; 2:1343-1421.

 

Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, Grobman WA, Launberg P, Lazarus JH, Mandel SJ, Peeters RP, Sullivan S. 2017 Guidelines of the American Thyroid Association for the diagnosis and management of thyroid disease during pregnancy and the postpartum. Thyroid: 27:315.

 

REFERENCES

 

Samuels SL, Namoc SM, Bauer AJ. Neonatal thyrotoxicosis Clin Perinatol 45:31-40 2018.

 

Barbesino G, Tomer Y. Clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab.

2013; 98:2247-2255.

 

van Dijk MM, Smits IH, Fliers E, Bisschop PH. Maternal thyrotropin receptor antibody concentration and the risk of fetal and neonatal thyrotoxicosis: a systematic review. Thyroid 2018: 28:257-

 

Rivkees S, Pediatric Graves’ disease: management in the post -propylthiouracil Era. Int J Pediatr Endocrinol. 2014 10

 

Van der Kaay D, Wasserman JD, Palmert MR. Management of neonates born to mothers with Graves’ disease. Pediatrics. 2016;137:e20151878

 

Paschke R, Niedzela M, Vaidya B, Persani L, Rapaport B, Leclere J. The management of

familial and persistent sporadic non-autoimmune hyperthyroidism caused by thyroid stimulating

hormone receptor germline mutations. Eur Thyroid J. 2012; 1:142-147.

 

Segni M. Disorders of the Thyroid Gland in Infancy, Childhood and Adolescence. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2017 Mar 18.

 

Kleinau G, Vassart G. TSH Receptor Mutations and Diseases. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2017 Jul 24.

 

 

 

Congenital Adrenal Hyperplasia: Diagnosis and Emergency Treatment

CLINICAL RECOGNITION


Congenital adrenal hyperplasia (CAH) is a group of autosomal recessive disorders that arise from defective steroidogenesis. The production of cortisol in the zona fasciculata of the adrenal cortex occurs in five major enzyme-mediated steps. CAH results from deficiency in any one of these enzymes. Impaired cortisol synthesis leads to chronic elevations of ACTH via the negative feedback system, causing overstimulation of the adrenal cortex and resulting in hyperplasia and over-secretion of the precursors to the enzymatic defect. The forms of CAH are summarized in Table 1. Impaired enzyme function at each step of adrenal cortisol biosynthesis leads to a unique combination of elevated precursors and deficient products. The most common enzyme deficiency that accounts for more than 90% of all CAH cases is 21-hydroxylase deficiency (21OHD).

 

Table 1. Types of Congenital Adrenal Hyperplasia

Condition

Onset

Abnormality

Genitalia

Mineralocorticoid Effect

Gene

Chromosomal Location

Typical Features

Lipoid CAH
Congenital
StAR Protein

Female, with no sexual development
Salt wasting

StAR 

8p11.2
All steroid products low

Lipoid CAH

Congenital

P450scc

Female, with no sexual development
Salt wasting

CYP11A

15q23-24
All steroid products low

3β-HSD deficiency Congenital
3β-HSD

Females virilized, males hypovirilized
Salt wasting

HSD3B2

1p13.1
Elevated DHEA, 17-pregnenolone, low androstenedione, testosterone, elevated K, low Na, CO2

17α-OH deficiency Congenital

P450c17

Males hypovirilized, Hyperkalemic low-renin hypertension

CYP17

CYP17

10q24.3
Decreased androgens and estrogen, elevated DOC, corticosterone

Classic 21-OH deficiency, salt wasting
Congenital

P450c21

Females prenatally virilized, males unchanged
Salt wasting occurs in ¾ of 21OHD patients

CYP21 

6p21.3
Elevated 17-OHP, DHEA, and androstenedione, elevated K, low Na, CO2, low aldosterone, high plasma renin

Classic 21-OH deficiency, simple virilizing
Congenital P450c21

Females prenatally virilized, males unchanged
No salt wasting

CYP21 

6p21.3
Elevated 17-OHP, DHEA, and androstenedione, normal electrolytes

Non-classic 21-OH deficiency

Postnatal
P450c21

All with normal genitalia at birth, hyperandrogenism postnatally
No salt wasting

CYP21 

6p21.3
Elevated 17-OHP, DHEA, and
androstenedione on ACTH stimulation

11β-OH deficiency Congenital

P450c11B1

Females virilized, males unchanged
Low-renin hypertension

CYP11B1  

8q24.3
Elevated DOC, 11-deoxycortisol (S); androgens, low K, elevated Na, CO2

P450 Oxidoreductase deficiency (POR), Congenital

P450 oxidoreductase

 

Males undervirilized, females unchanged

Variable degree of mineralocorticoid deficiency

P450 Oxidoreductase gene (POR)

7q11.2

Combined and variable enzymatic defects of P450c21, P450c17 and P450aro

Wide range of phenotypes: normal to genital ambiguity +/- skeletal abnormalities (Antley Bixler type)

 

 

Classical CAH occurs in 1:13,000 to 1:15,000 live births. It is estimated that 75% of patients have the salt-wasting (SW) phenotype and the rest have simple-virilizing (SV) phenotype. Non-classical 21-OHD CAH (NC-CAH) is more common, and is one of the most common disorders in the Ashkenazi Jewish population with 1 in 27 Jews affected.  CAH owing to 11β-hydroxylase deficiency (11β-OHD) is the second most common cause of CAH, accounting for 5-8% of all cases. The other forms of CAH are considered rare diseases and the incidence is unknown in the general population.

 

PATHOPHYSIOLOGY

Adrenal steroidogenesis occurs in three major pathways: glucocorticoids, mineralocorticoids, and sex steroids as shown in Figure 1. Glucocorticoids (particularly cortisol), androgens, and estrogens are synthesized in the zona fasciculata and reticularis; and aldosterone in the zona glomerulosa. The HPA feedback system is mediated through the circulating level of plasma cortisol by negative feedback of cortisol on CRF and ACTH secretion. Therefore, a decrease in cortisol secretion leads to increased ACTH production, which in turn stimulates (1) excessive synthesis of adrenal products in those pathways unimpaired by the enzyme deficiency and (2) an increase of precursor molecules in pathways blocked by the enzyme deficiency.

Figure 1. Pathways of Adrenal Steroidogenesis: Five enzymatic steps necessary for cortisol production are shown in numbers. 1= 20, 22 desmolase, 2= 17 hydroxylase (17-OH), 3=3ß-hydroxysteroid dehydrogenase (3ß HSD), 4=21 hydroxylase (21-OHD), 5=11ß hydroxylase (11-OH) In the first step of adrenal steroidogenesis, cholesterol enters mitochondria via a carrier protein called StAR. ACTH stimulates cholesterol cleavage, the rate limiting step of adrenal steroidogenesis.

 

The clinical symptoms of the five different forms of CAH result from the particular hormones that are deficient and those that are produced in excess as outlined in Table 1. In 21 OHD-CAH, there is an accumulation of 17-hydroxyprogesterone (17-OHP), a precursor to the 21-hydroxylation step, which is then shunted into the intact androgen pathway, where the 17,20-lyase enzyme converts the 17-OHP to D4-androstenedione, which is converted into androgens. Mineralocorticoid deficiency is a feature of SW-CAH, the most severe form of CAH. The enzyme defect in NC-CAH is only partial and salt wasting in this mild form of the disease does not occur. The analogy of all other enzyme deficiencies in terms of precursor retention and product deficiencies are shown in Table 1.

 

CLINICAL FEATURES

Genitalia

Females with Classical 21-OHD and 11β-hydroxylase deficiency CAH present at birth with virilization of their genitalia. Adrenocortical function begins around the 7th week of gestation; thus, a female fetus with classical CAH is exposed to adrenal androgens at the critical time of sexual differentiation (approximately 9 to 15 weeks gestational age). This leads to clitoral enlargement, fusion and scrotalization of the labial folds, and rostral migration of the urethral/vaginal perineal orifice, placing the phallus in the male position. Degrees of genital virilization are classified into five Prader stages (see Figure 2).

Figure 2. Different degrees of virilization according to the scale developed by Prader

Stage I: clitoromegaly without labial fusion

Stage II: clitoromegaly and posterior labial fusion

Stage III: greater degree of clitoromegaly, single perineal urogenital orifice, and almost complete labial fusion

Stage IV: increasingly phallic clitoris, urethra-like urogenital sinus at base of clitoris, and complete labial fusion

Stage V: penile clitoris, urethral meatus at tip of phallus, and scrotum-like labia (appear like males without palpable gonads)

Prader, A. Helv Paediatr Acta, 1954. 9:230-248.

 

Internal female genitalia, such as the uterus, fallopian tubes and ovaries, develop normally. Females with classical CAH maintain the internal genitalia potential for fertility.

 

Postnatal Effects, Growth and Puberty

Lack of appropriate postnatal treatment in boys and girls results in continued exposure to excessive androgens, causing progressive penile or clitoral enlargement, the development of premature pubic hair, axillary hair and acne. Advanced somatic and epiphyseal development occurs with exaggerated growth and is usually accompanied by premature epiphyseal maturation and closure, resulting in a final adult height that is typically significantly below that expected from parental heights. Excess glucocorticoid treatment can also lead to poor growth. The mean age at onset of puberty in both males and females is slightly younger than the general population. In those who are inadequately treated, central precocious puberty can occur. Following the onset of puberty, in a majority of successfully treated patients, the milestones of further development of secondary sex characteristics in general appear to be normal. In female adolescents and adults, signs of hyperandrogenism may include male-pattern alopecia (temporal balding), acne, hirsutism, menstrual irregularities, secondary PCOS and impaired fertility. Although the expected age of menarche may be delayed in females with classical CAH, when adequately treated many have regular menses after menarche.  In males, short stature and impaired fertility are observed.

 

Gender Role Behavior and Cognition

Prenatal androgen exposure in females affected with classical forms of CAH not only has a masculinizing effect on the development of the external genitalia, but also on childhood behavior. Both physical and behavioral masculinization are related to each other and to genotype, indicating that behavioral masculinization in childhood is a consequence of prenatal androgen exposure. The majority of genetic females with CAH retain the female gender identity even in the setting of prenatal androgen exposure and postnatal hyperandrogenism.

 

Fertility

Difficulty with fertility in females with CAH may be due to anovulation, secondary polycystic ovarian syndrome, irregular menses, non-suppressible serum progesterone levels, or an inadequate introitus. Fertility is reduced in SW-CAH with rare reports of pregnancy. Non-classical CAH is an important and frequently unrecognized form of infertility. Males with CAH, particularly if poorly treated, may have reduced sperm counts and low testosterone as a result of high androstenedione concentrations which suppress gonadotropins and testicular adrenal rest tumors. Testicular adrenal rest tumors (TART) are thought to arise from aberrant adrenal cells in the testes; TARTs are always benign and mostly bilateral. Microscopic examination shows that adrenal rest cells are present in the testicles of all male patients with CAH and often detected radiographically in those with longstanding poorly controlled disease. Regular testicular examination and periodic testicular ultrasonography are recommended for early detection of adrenal rest tumors of the testes. However, MRI studies have been increasingly used to diagnose TARTs.

 

Salt-Wasting 21-Hydroxylase Deficiency

When the loss of 21-hydroxylase function is severe, adrenal aldosterone secretion is not sufficient for sodium reabsorption by the distal renal tubules, and individuals suffer from salt wasting as well as cortisol deficiency and androgen excess. Infants with renal salt wasting have poor feeding, weight loss, failure to thrive, vomiting, dehydration, hypotension, hyponatremia, and hyperkalemic metabolic acidosis progressing to adrenal crisis (azotemia, vascular collapse, shock, and death). Adrenal crisis can occur as early as age one to four weeks. Affected males who are not detected in a newborn screening program are at high risk for a salt-wasting adrenal crisis because their normal male genitalia do not alert medical professionals to their condition. It is important to recognize that the extent of genital virilization may not differ among SV-CAH and SW-CAH.

 

Simple-Virilizing 21-Hydroxylase Deficiency

The salient features of classical SV-CAH are prenatal virilization and progressive postnatal masculinization with rapid somatic growth and advanced epiphyseal maturation leading to early epiphyseal closure and likely short stature. There is no evidence of mineralocorticoid deficiency in this disorder and serum electrolyte concentrations are normal. Diagnosis at birth of a female with SV-CAH is usually made immediately because of the apparent genital ambiguity. Since the external genitalia are not affected in newborn males, hyperpigmentation may be the only clue suggesting increased ACTH secretion and cortisol deficiency. Diagnosis at birth in males thus rests on prenatal or newborn screening.

 

Non-Classical 21-Hydroxylase Deficiency

Individuals with the non-classical (NC) form of 21-OHD have only mild to moderate enzyme deficiency and present postnatally, eventually developing signs of hyperandrogenism. Females with NC-CAH do not have virilized genitalia at birth. NC-CAH may present at any age after birth with a variety of hyperandrogenic symptoms. While serum cortisol concentration is typically low in patients with the classic form of the disease, it is usually normal in patients with NC 21-OHD. Similar to classical CAH, NC-CAH may cause premature development of pubic hair, acne, secondary PCOS, advanced bone age with accelerated linear growth velocity, and short stature. In adult males, early balding, acne, infertility or short stature may prompt the diagnosis of NC-CAH.

 

DIAGNOSIS

Diagnosis of CAH must be suspected in infants born with ambiguous genitalia. The physician is obliged to make the diagnosis as quickly as possible to initiate therapy. The diagnosis and rational decision of sex assignment must rely on the determination of genetic sex, the hormonal determination of the specific deficient enzyme, genotype, and an assessment of the patient's potential for future sexual activity and fertility. As indicated in Table 1, each form of CAH has its own unique hormonal profile, consisting of elevated levels of precursors and elevated or diminished levels of adrenal steroid products. Diagnosis of the 21-OHD CAH can also be confirmed biochemically by a hormonal evaluation. In a randomly timed blood sample, a very high concentration of 17-hydroxyprogesterone (17-OHP), the precursor of the defective enzyme, is diagnostic of classical 21-OHD. Such testing is the basis of the newborn-screening program developed to identify classically affected patients who are at risk for salt wasting crisis. False-positive results are, however, common with premature infants. Appropriate references based on weight and gestational age are therefore in place in many screening programs. False negative results may occur if samples are drawn late in the afternoon as adrenal hormones exhibit diurnal variation.  The gold standard for hormonal diagnosis is the corticotropin stimulation test (250 μg cosyntropin intravenously), measuring levels of 17-OHP and Δ4 androstenedione at baseline and 60 min. These values can then be plotted in the published nomogram (Figure 4) to ascertain disease severity. The corticotropin stimulation test should not be performed during the initial 24 hours of life as samples from this period are typically elevated in all infants and may yield false-positive results. Establishing a genetic diagnosis is not only important for the genotype-phenotype correlation, but also for genetic counseling for future pregnancies and for genetic counseling for the patient and his/her reproductive future.

 

For 21-OHD CAH, genetic analysis of the CYP21A2 gene may provide more clues to predict phenotypic severity. In about 50% of the causative genotypes, genotype-phenotype correlation can be found, although certain mutations can lead to variable phenotypes in different population groups especially in the simple virilizer group. Sequencing of the entire gene should be performed to detect rare mutations when genotype–phenotype non-concordance is observed in patients with CAH.

 

Newborn screening for CAH, which utilizes 17 hydroxyprogesterone levels, is a useful tool for early detection of CAH prior to the development of adrenal crisis in the affected neonate.   However, screening is associated with a high rate of false positive results as levels are affected by prematurity and birth weight. Molecular genetics, especially genotyping of the CYP21A2 gene should be considered as a second-tier screening test in the new born screening program.

 

Prenatal testing for CAH in utero has historically utilized invasive techniques like amniocentesis and chorionic villus sampling which cannot be done prior to 14 weeks of gestation. Prenatal dexamethasone treatment must begin prior to genital formation occurring at approximately 9 weeks, in order to avoid genital ambiguity in the affected female fetus. Massive parallel sequencing using hybridization probes on cell-free fetal DNA in maternal plasma indicated that the fetal CAH status was correctly deduced as early as 5 weeks 6 days of gestation. This is a noninvasive technique that accurately diagnoses CAH before the ninth week of gestation.

 

TREATMENT

Routine Treatment

br />

The goal of therapy in CAH is to both correct the deficiency in cortisol secretion and to suppress ACTH overproduction. Proper treatment with glucocorticoid reduces stimulation of the androgen pathway, thus preventing further virilization and allowing normal growth and development. The usual requirement of hydrocortisone (or its equivalent) for the treatment of classical 21-OHD form of CAH is about 10-15 mg/m2/day divided into 2 or 3 doses per day and for non-classical 21-OHD 5-8 mg/m2/day divided into 2 or 3 doses per day. Hydrocortisone is the glucocorticoid of choice in the pediatric age group. Prednisolone and dexamethasone are not used in growing children given growth suppressive effects. A small dose of dexamethasone at bedtime (0.25 to 0.5 mg) is usually adequate for androgen suppression in non-classical adult patients. Adequate biochemical control is assessed by measuring serum levels 17-OHP and androstenedione; serum testosterone can be used in females and prepubertal males (but not newborn males). We recommend that hormone levels are measured at a consistent time in relation to medication dosing, usually 1-2 hours after the morning corticosteroid. Titration of the dose should be aimed at maintaining 17-OHP concentrations below 1000 ng/dL and androstenedione concentrations below 200 ng/dl. Over-treatment should be avoided because it can lead to Cushing syndrome. Patients with salt wasting CAH have elevated plasma renin in response to the sodium-deficient state, and they require treatment with the salt-retaining 9α-fludrocortisone acetate. The average dose is 0.1 mg daily (0.05-0.2 mg daily). Infants should also be started on salt supplementation, as sodium chloride, at 1-2 g daily, divided into several feedings. Measurements of plasma renin and aldosterone are used to monitor the efficacy of mineralocorticoid therapy.   Advancement of bone age is monitored by bone age x-rays. Growth hormone therapy, in conjunction with a GnRH analogue, has been shown to be effective in improving final adult height. Patients may also experience peripheral precocious puberty, which requires treatment with gonadotropin-releasing hormone analogues. Aromatase inhibitors and growth hormone therapy should only be used in patients with a very short predicted final stature or in clinical trials. Use of aromatase inhibitors in CAH has been shown decrease bone maturation rates and some increase in adult height but the differences were not statistically significant.

 

Treatment During Illness and Emergency


Adrenal crisis can present as hypotension or shock and serum electrolyte abnormalities (hypoglycemia, hyponatremia, hyperkalemia, acidosis). During adrenal crisis, an immediate bolus of hydrocortisone 50-100 mg can be given intravenously or intramuscularly followed by hydrocortisone 100 mg/m2/day given as either continuous infusion or divided at least every 6 hours. Rehydration can be started with 20ml/kg isotonic saline with D5 as rapid bolus followed by repeat boluses or continuous infusion guided by level of dehydration. Hypoglycemia may require dextrose bolus and an initial bolus of 0.5-1 gram/kg of dextrose can be given intravenously at 2-3 ml per minute. If hyperkalemia is present, cardiac monitoring should be done to monitor for EKG changes. If changes are present, hyperkalemia should be treated using insulin with glucose infusion with or without other measures.

 

In non-life-threatening periods of illness or physiologic stress, the corticosteroid dose should be increased to 2 or 3 times the maintenance dose for the duration of that period, divided into 3 daily doses. Each family should be given injection kits of hydrocortisone, i.e. Solu-Cortef, for emergency use, and all family members should be trained in its intramuscular administration. The injectable dose of hydrocortisone in an emergency is 25 mg for infants, 50 mg for children under 40 kg, and 100 mg for children over 40 kg and for adults. In the event of a surgical procedure, 5-10 times the daily maintenance dose of hydrocortisone is needed, with 25-100 mg hydrocortisone IM/IV administered before and during a surgical procedure (as per infant, child, adult recommendations above), followed by high doses of hydrocortisone during the first 24-48 post-operative hours; the dose can then be tapered over the following days to the normal preoperative schedule. Stress doses of dexamethasone should not be given because of the delayed onset of action. It is not necessary for increased mineralocorticoid doses during these periods of stress. It is imperative for all patients who are receiving corticosteroid replacement therapy, such as patients with CAH, to wear a Medical Alert bracelet or medallion that will enable correct and appropriate therapy in case of emergencies. It is also crucial to re-educate parents at regular intervals on the life-threatening nature of this emergency.

 

GUIDELINES

 

Speiser PW, Arlt W, Auchus RJ, Baskin LS, Conway GS, Merke DP, Meyer-Bahlburg HFL, Miller WL, Murad MH, Oberfield SE, White PC. Congenital Adrenal Hyperplasia Due to Steroid 21-Hydroxylase Deficiency: An Endocrine Society Clinical Practice Guideline.

J Clin Endocrinol Metab. 2018 Nov 1;103(11):4043-4088

 

Rodriguez A, Ezquieta B, Labarta JI, Clemente M, Espino R, Rodriguez A, et al. Recommendations for the diagnosis and treatment of classic forms of 21-hydroxylase-deficient congenital adrenal hyperplasia. An Pediatr (Barc). 2017;87(2):116 e1- e10.

 

REFERENCES

 

New M, Yau M, Lekarev O, Lin-Su K, Parsa A, Pina C, Yuen T, Khattab A. Congenital Adrenal Hyperplasia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2017 Mar 15

 

El-Maouche D, Arlt W, Merke DP. Congenital adrenal hyperplasia. Lancet. 2017 Nov 11;390(10108):2194-2210

 

Fluck CE, Miller WL. P450 oxidoreductase deficiency: a new form of congenital adrenal hyperplasia. Curr Opin Pediatr. 2006;18(4):435-41.

 

Yilmaz R, Sahin D, Aghayev A, Erol OB, Poyrazoglu S, Saka N, et al. Sonography and Magnetic Resonance Imaging Characteristics of Testicular Adrenal Rest Tumors. Pol J Radiol. 2017;82:583-8.

 

New MI, Abraham M, Gonzalez B, Dumic M, Razzaghy-Azar M, Chitayat D, et al. Genotype-phenotype correlation in 1,507 families with congenital adrenal hyperplasia owing to 21-hydroxylase deficiency. Proc Natl Acad Sci U S A. 2013;110(7):2611-6.

 

Balsamo A, Baldazzi L, Menabo S, Cicognani A. Impact of molecular genetics on congenital adrenal hyperplasia management. Sex Dev. 2010;4(4-5):233-48.

 

New MI, Tong YK, Yuen T, Jiang P, Pina C, Chan KC, et al. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab. 2014;99(6):E1022-30.

 

Lin-Su K, Harbison MD, Lekarev O, Vogiatzi MG, New MI. Final adult height in children with congenital adrenal hyperplasia treated with growth hormone. J Clin Endocrinol Metab. 2011;96(6):1710-7.

 

 

 

 

Disorders of Sexual Differentiation in Newborns

INTRODUCTION      

 

In 2006, new nomenclature for conditions previously referred to as intersex was proposed in a consensus statement from the Lawson Wilkins Pediatric Endocrine Society and European Society of Pediatric Endocrinology in response to advanced identification of molecular genetic causes of sex. Disorders of sexual differentiation (DSD) are congenital conditions within which the development of chromosomal, gonadal and phenotypic sex is atypical. These disorders have a broad differential including variations in sex chromosomes, variations in genes involved in gonadal and genital development, disorders in steroidogenesis within the gonads and adrenals, maternal factors, and endocrine disruptors.   Classification of these disorders is based on sex chromosomes as such 46XX DSD, 46XY DSD, Ovotesticular DSD, and 46XX testicular DSD. 

Approximately 1-2% of live births are affected with atypical genitalia, including isolated hypospadias in males. The incidence of 46XX DSD is 1:15,000. The incidence of 46XY DSD is higher at 1:5000.

CLINICAL RECOGNITION

Given the potential association with glucocorticoid and mineralocorticoid deficiencies in CAH, the birth of a child with atypical genitalia constitutes a medical emergency requiring immediate evaluation. Further, the parents’ reaction to the birth of a child with atypical genitalia is one of shock and concern about which gender to assign, whether or not to decide for early genital surgery, and what to expect regarding the long-term outcome in terms of gender, sexual function, fertility, and general quality of life.  In order to provide appropriate counseling to the family, there is an urgency to determine the etiology.

PATHOPHYSIOLOGY

The phenotypic sex of a newborn is the result of external genital development that is under the influence of sex-determining genes as well as both endogenous and exogenous hormone exposures. 

The commitment of the bipotential primordial gonads to become testes or ovaries begins at 6 weeks and is fully achieved at 13-14 weeks.  Gonadal differentiation is controlled by a number of time and dosage-sensitive genes including the SRY gene on the Y chromosome, SOX9, and WNT4 genes.  The expression SRY and SOX9 and suppression of WNT4 expression is crucial to testicular differentiation.  The expression of WNT4 in the absence of SRY and SOX9 expression allows for ovarian differentiation. Leydig cells produce insulin like factor 3 (INSL3) which is responsible for transabdominal phase of testicular descent.

Fetal productions of androgens from the Leydig cells within the testes and from the adrenal glands begins at approximately 8-9 weeks. External genitalia develop concurrently around the 9th week of gestation under the influence of androgens, mainly dihydrotestosterone (DHT). Testosterone is the principal hormone produced by the testes and is required for the onset of virilization and promotion of Wolffian ducts.  Testosterone is converted to DHT by 5-alpha reductase.  DHT leads to the development of the prostate, scrotum and phallus. 

Anti-Mullerian Hormone (AMH) produced from Sertoli cells in the testes is required to support the development of Wolffian ducts including vas deferens, epididymis and seminal tubules in males. In females, Mullerian ductal structures including the uterus, fallopian tubes and cervix develop in the absence of AMH.

Disorders of Sexual Differentiation

46XX DSD

Patients with 46XX DSD are genotypic females with virilized characteristics. In 46XX DSD, the degree of genital virilization can be classified into five Prader stages. Stage 1, with the mildest degree of virilization, is characterized by clitoromegaly without labial fusion.  Stage 5, with the highest degree of virilization, is characterized by clitoromegaly with the urethral meatus at the tip, labial fusion, and scrotal-like appearance of the labia.  

46XX DSD can result from exogenous androgen exposure, endogenous adrenal androgen production or placental aromatase deficiency. Congenital adrenal hyperplasia (CAH) is the most common cause of 46XX DSD. The most common enzyme defects leading to CAH are 21-hydroxylase deficiency and 11-hydroxylase deficiency.  In a very rare form of CAH owing to p450 oxidoreductase deficiency, there is a mutation in the P450 oxidoreductase (POR) enzyme which causes partial deficiency of 21-hydroxylase and 17a-hydroxylase/17,20 lyase activities. Affected females can present with virilization of the external genitalia, glucocorticoid deficiency, and skeletal malformations such as craniosynostosis.

 

Maternal hyperandrogenism during gestation can cause virilization of the external genitalia in females when the placental aromatase is overwhelmed. The hyperandrogenism can be due to luteomas, androgen producing tumors and exogenous exposure. 

Maternal aromatase deficiency leads to decreased production of estrogen from androgen precursors. This leads to conversion of fetal DHEAS to androstenedione and testosterone by placental 3-beta hydroxysteroid dehydrogenase and virilization of female fetus. 

The majority of 46XX testicular DSD cases are caused by translocation between the X and Y chromosome, involving the SRY gene.

46XY DSD

Patients with 46XY DSD are genotypic males with under-virilization.  Micropenis is defined as a penile length less than 2.5 standard deviations below the mean penile length (<2.5 cm in a full-term newborn).  The severity of hypospadias is based on the distance of the urethral opening from its normal position at the tip of the phallus.  Lack of testicular palpation in the scrotum may signify cryptorchidism, vanishing testes, or gonadal dysgenesis.

46XY DSD can be caused by atypical testicular formation, low testosterone or dihydrotesterone production, or defects in the androgen receptor. In complete gonadal dysgenesis, there is no testicular development and patients present as phenotypic female with delayed puberty or amenorrhea.  Up to 20% of these cases occur due to deletion or mutation of the SRY gene.

Defects in androgen biosynthesis can lead to under-virilization in a 46XY patient. These defects can occur at various points along the production pathway of testosterone from cholesterol. Adrenal dysfunction is associated with defects in steroidogenic enzymes such as steroidogenic acute regulatory protein (StAR), p450 side chain cleavage enzyme, 3 beta HSD type 2, 17 alpha hydroxylase/17,20 lyase. Other defects of testosterone production can occur in the following enzymes: 7-dehydrocholesterol reductase causing Smith Lemli Opitz syndrome and 17 beta hydroxysteroid dehydrogenase. Affected males with 5-alpha-reductase deficiency have atypical genitalia (small phallus and perineal hypospadias).  With rises in testosterone at puberty, progressive virilization with phallic enlargement and testicular descent is seen.

Androgen insensitivity syndrome has been reported to be the main cause of 46XY DSD and is due to mutation in the androgen receptor.  In complete androgen insensitivity, the androgenic effects of testosterone and dihydrotestosterone are abolished and patients have unambiguously female appearing external genitalia. In partial androgen insensitivity, the androgenic effects are attenuated and patients can present on a spectrum of under-virilization.

Mutations in genes responsible for sex determination such as SRY, SOX9, and SF1 lead to 46XY complete gonadal dysgenesis.  Duplication of the DAX1 gene is associated with male to female sex reversal. 

Endocrine disruptors with anti-androgenic effects such as diethylstilbestrol or phthalates can also lead to atypical genitalia in males.

Table 1.  Laboratory Values to Differentiate Between Etiologies of Ambiguous Genitalia in Newborns with a 46, XY Chromosomal Complement.

Diagnosis

T

DHT

MIS

Androgen Insensitivity
Syndrome (AIS)

Normal/up

Normal/up

Normal

5α-Reductase Deficiency

Normal/up

Low

Normal

Testosterone Biosynthetic Defect orLeydig Cell Hypoplasia

Low

Low

 

Normal

Gonadal Dysgenesis

Low

Low

Low

T=testosterone, DHT=dihydrotestosterone and MIS=müllerian inhibiting substance.

OVOTESICULAR DSD

Ovotesticular DSD, one of the rarest forms of DSD, describes patients that were previously categorized as true hermaphrodites.  The gonads of patients with ovotesticular DSD contain both ovarian and testicular tissue.  Thus, the presentation of genital ambiguity can be variable. In ovotesticular DSD in which the gonads contain both ovarian and testicular tissue, the majority have an XX chromosomal constitution.  Complex mosaicism (XX/XY) are seen in approximately 10% of cases. Patients can present with a wide variety of genital ambiguity as well as a mixture of Wolffian and Mullerian structures.

DIAGNOSIS

Determination of chromosomal sex by karyotype with FISH analysis for SRY and pelvic ultrasound to evaluate for the presence of a uterus should be performed immediately. Currently, the only newborn screening test for steroid disorders is the measurement of 17-hydroxyprogesterone for 21-hydroxylase deficiency.   Further laboratory evaluation to accurately diagnose the specific underlying defect should be directed by a pediatric endocrinologist.  If CAH is suspected, measurement of adrenal hormones, ACTH stimulation testing, and molecular genetic testing can elucidate the form of CAH. Each form of CAH has its own unique hormonal profile, consisting of elevated levels of precursors and elevated or diminished levels of adrenal steroid products. HCG stimulation testing to assess testosterone and dihydrotestosterone response may be particularly helpful in 46XY DSD to assess testicular androgen production.  Molecular genetic evaluations should be guided by chromosomal and hormonal evaluations.

Chromosomal sex can be determined prenatally invasively by chorionic villus sampling and amniocentesis and noninvasively via free fetal DNA in the maternal blood. Thus, DSD may be suspected in utero if the phenotype on prenatal ultrasonogram is discordant with the chromosomal sex. 

THERAPY

When considering the gender of rearing, the prognosis for masculinization of brain and behavior, the anatomic and physiologic character of the reproductive tract with its potential for development and function in regard to both sexuality and fertility, and the social environment of the infant should be taken into account along with the genetic sex.  Both male and female gender assignment should be thoroughly considered.

Sex hormone replacement is needed to induce pubertal development.  Testosterone is used in the treatment of patients with testosterone deficiency (46XY DSD).  Different forms of testosterone (topical and intramuscular) are available and treatment will vary depending on what is best for the patient.  A short course of testosterone can be given during infancy to induce penile growth prior to surgical correction.  For 46XY DSD patients with functioning Sertoli cells, HCG can be used to stimulate testicular production.  Estrogen is used in the treatment of those reared female.  Estrogen is available as an oral tablet or transdermal patch.  Estrogen doses should be initiated at the lowest dose possible and slowly increased to a maximum of 0.625 mg/day of conjugated estrogen to allow for gradual breast development.  Progesterone supplementation with estrogen is recommended in patients with a uterus.

 

Glucocorticoids are needed to treat congenital adrenal hyperplasia. They suppress the pituitary glands oversecretion of adrenocorticotropic hormone and thus decrease the production of precursor hormones.  This also leads to a decrease in adrenal androgen production in forms of CAH associated with 46XX DSD.

The aim of surgical repair in patients with atypical genitalia reared in the female gender is generally to remove the redundant erectile tissue, preserve the sexually sensitive glans clitoris, and provide a normal vaginal orifice.  A medical indication for early surgery other than for sex assignment is recurrent urinary tract infections as a result of pooling of urine in the vagina or urogenital sinus. In the past, it was routine to recommend early corrective surgery for neonates born with ambiguous genitalia. However, in recent years, the implementation of early corrective surgery has become increasingly controversial due to lack of data on long-term functional outcome.  It is advised that all surgical decisions remain the prerogative of families in conjunction with experienced surgical consultants.

The process of assigning and accepting a gender of rearing for a child with ambiguous genitalia and of deciding the necessity of genital surgery is challenging. A team approach that combines the insights of the DSD-experienced pediatrician, endocrinologist, psychologist/psychiatrist, surgeon, and the child’s parents or guardian is essential. Although there is no consensus as to the appropriate age to disclose a condition, it is recommended to proceed gradually in line with the child’s cognitive and psychological development.

GUIDELINES

Audi L, Ahmed SF, Krone N, Cools M, McElreavey K, Holterhus PM, Greenfield A, Bashamboo A, Hiort O, Wudy SA, McGowan R, The EUCA: GENETICS IN ENDOCRINOLOGY: Approaches to molecular genetic diagnosis in the management of differences/disorders of sex development (DSD): position paper of EU COST Action BM 1303 'DSDnet'. Eur J Endocrinol 2018;179:R197-R206.

Lee PA, Houk CP, Ahmed SF, Hughes IA, International Consensus Conference on Intersex organized by the Lawson Wilkins Pediatric Endocrine S, the European Society for Paediatric E: Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics 2006;118:e488-500.

Speiser PW, Arlt W, Auchus RJ, Baskin LS, Conway GS, Merke DP, Meyer-Bahlburg HFL, Miller WL, Murad MH, Oberfield SE, White PC: Congenital Adrenal Hyperplasia Due to Steroid 21-Hydroxylase Deficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2018;103:4043-4088.

REFERENCES

Lee PA, Nordenstrom A, Houk CP, Ahmed SF, Auchus R, Baratz A, Baratz Dalke K, Liao LM, Lin-Su K, Looijenga LH, 3rd, Mazur T, Meyer-Bahlburg HF, Mouriquand P, Quigley CA, Sandberg DE, Vilain E, Witchel S, Global DSDUC: Global Disorders of Sex Development Update since 2006: Perceptions, Approach and Care. Horm Res Paediatr 2016; 85:158-180.

 

Blackless M, Charuvastra A, Derryck A, Fausto-Sterling A, Lauzanne K, Lee E: How sexually dimorphic are we? Review and synthesis. Am J Hum Biol 2000; 12:151-166.

Krishnan S W, AB: Ambiguous Genitalia in Newborns; in New MI LO, Parsa A, Yuen T, O'Malley B, Hammer G (ed) Genetic Steroid Disorders. San Diego, CA, Elsevier, 2014, pp 87-97.

New M, Yau M, Lekarev O, Lin-Su K, Parsa A, Pina C, Yuen T, Khattab A. Congenital Adrenal Hyperplasia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2017 Mar 15.

Domenice S, Arnhold IJP, Costa EMF, Mendonca BB. 46,XY Disorders of Sexual Development. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2017 May 3.

 

Fibrous Dysplasia

ABSTRACT

 

Fibrous dysplasia (FD) is an uncommon mosaic disorder falling along a broad clinical spectrum. It arises from post-zygotic mutations in GNAS, resulting in constitutive activation of the cAMP pathway-associated G-protein, Gsα, and proliferation of undifferentiated skeletal progenitor cells.  FD may occur in isolation, or in association with skin pigmentation and hyperfunctioning endocrinopathies, termed McCune-Albright syndrome (MAS). Disease may involve any part or combination of the skeleton, ranging from an isolated, asymptomatic monostotic lesion, to severe polyostotic disease resulting in fractures, deformity, functional impairment, and progressive scoliosis. FD may be diagnosed clinically in patients with polyostotic disease and/or extraskeletal features of MAS; however, biopsy is typically required to diagnose monostotic disease. Management is focused on treating endocrinopathies, preventing fractures, optimizing function, and treating pain. All patients should be evaluated and treated for extraskeletal features of MAS at the time of diagnosis. In particular control of growth hormone excess is important to prevent craniofacial FD expansion, and control of FGF23-mediated hypophosphatemia is important to prevent fracture, deformity, and bone pain. A mainstay of FD treatment is surgical, and practitioners should be aware that techniques and procedures used in other skeletal disorders, such as bone grafting and prophylactic optic nerve decompression, are frequently ineffective in FD. There are currently no medical therapies capable of altering the disease course in FD. Bisphosphonates may be effective in treating FD-related bone pain but are unlikely to impact bone quality or lesion expansion. There is a critical need to develop novel therapies capable of altering the disease activity of FD lesions.  Ongoing efforts include developing drugs to target the mutant Gsα, and devising strategies for targeting mutant skeletal progenitor cells. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

 

INTRODUCTION

 

Fibrous dysplasia of bone (FD) (OMIM#1174800) is an uncommon skeletal disorder resulting in fractures, pain, and functional impairment. Disease is mosaic and may occur in a single bone (monostotic) or multiple bones (polyostotic). FD thus falls along a broad clinical spectrum, ranging from an incidental, asymptomatic lesion to severe disabling disease. Lesions may occur in isolation or in association with extraskeletal features, most commonly café-au-lait skin macules and hyperfunctioning endocrinopathies including precocious puberty, hyperthyroidism, growth hormone excess, hypercortisolism, and fibroblast growth factor-23 (FGF23)-mediated hypophosphatemia. The combination of FD and/or any of these extraskeletal features is termed McCune-Albright syndrome (MAS).  

 

PATHOGENESIS

 

FD/MAS results from post zygotic somatic activating (gain-of-function) mutations in GNAS, which encodes the cyclic AMP pathway-associated G-protein, Gsα. GNAS mutations in FD/MAS are known to occur at one of two amino acid residues: Arg201 (>95% of reported cases) [1] or Gln227 (<5%) [2]. These mutations disrupt the intrinsic GTPase activity of Gsα, causing constitutive activation of adenylyl cyclase and inappropriate cyclic AMP signaling.  Involvement of the skin, bone, and endocrine systems is consistent with a mutational event early in embryogenesis, occurring prior to derivation of the 3 germ layers. The phenotype in individuals with FD/MAS is thus the result of the distribution of tissues containing the GNAS mutation, and the role played by Gsα signaling in those tissues. This mosaicism is reflected in the clinical features of FD/MAS, including the distinctive appearance of café-au-lait macules which follow developmental lines of Blaschko (Fig 1A), the patchy bone disease (Fig 1B) with extreme variability between individuals, and the lack of any reported cases of vertical transmission [3].  

The pathogenesis of FD results from replacement of normal bone and bone marrow by fibro-osseous lesions. Histologically lesions are comprised of skeletal progenitor cells, which proliferate in the multipotent state without the expected differentiation into osteoblast, adipocyte, and hematopoietic-supporting cell lines. Skeletal progenitor cell proliferation and abnormal bone formation result in the characteristic histologic features of FD, including marrow fibrosis, abnormally-shaped trabeculae, and abnormal skeletal matrix formation [4] (Fig 2).  Also characteristic is skeletal undermineralization with prominent osteoid, due in part to increased production of FGF23 by FD cells [5, 6].

Figure 1. Clinical and radiographic features of fibrous dysplasia/McCune-Albright syndrome. A. Fibrous dysplasia involving the lower extremities has resulted in a windswept deformity and impaired ambulation. Note the café-au-lait macules (white arrowheads) on the posterior trunk with characteristic jagged borders and location respecting the midline of the body. B. Technetium-99 bone scan shows patchy tracer uptake in affected areas in the skull and limbs (black arrows). C. Radiograph of a patient with axial fibrous dysplasia resulting in severe thoracolumbar scoliosis.

Figure 2. Histopathologic features of fibrous dysplasia. Hematoxylin-eosin stained sections in low (A) and high power (B) show irregular, discontinuous trabeculae (b) within a fibrous stroma (ft), demonstrating the typical “discontinuous pattern. Goldner’s trichrome stained sections in low (C) and high power (D) reveal osteomalacic changes including excess osteoid (asterisks) and severe undermineralization of the dysplastic bone.

 

CLINICAL FEATURES

 

The clinical features in individuals with FD are variable depending upon the location and extent of skeletal lesions. The most commonly involved areas are the proximal femurs and skull base; however, any combination of affected bones is possible. FD in the appendicular skeleton often presents with fractures, limp, and/or pain. Recurrent fractures and FD expansion may lead to deformity with pain and loss of ambulation (Fig 1A). In the craniofacial area FD may present with facial asymmetry or a painless “lump” in the skull. Progressive expansion of craniofacial lesions may rarely lead to functional deficits including vision loss, malocclusion, and obstruction of the nasal and otic canals. Rarely, cranial base deformities may lead to serious neurologic complications [7]. Axial FD commonly leads to scoliosis, which in rare cases may be progressive and potentially fatal [8-10] (Fig 1C).

 

Radiographic features of FD also vary depending upon location. In the appendicular skeleton lesions appear radiolucent with cortical thinning and a characteristic “ground glass” appearance (Fig 3A). Craniofacial FD may appear sclerotic on radiographs, while computed tomography shows expansile lesions with a homogeneous “ground glass” appearance (Fig 3B). In older individuals craniofacial FD may appear more heterogeneous with focal cystic and sclerotic areas (Fig 3C).

Figure 3. Radiographic appearance of fibrous dysplasia (FD). A. Extensive FD involvement of the bilateral femurs in an 11-year-old girl demonstrates the characteristic radiographic findings, including the homogeneous “ground glass” appearance, diffuse cortical thinning, and coxa vara (“shepherd’s crook”) deformities (white arrows). B. Computed tomography images of craniofacial FD from a 10-year-old girl demonstrate the typical homogeneous, “ground glass” appearance. White arrows indicate the optic nerves, which are encased in FD. C. Computed tomography images from the same patient at age 19 years demonstrates typical age-related changes, including the development of mixed solid and radiolucent, cystic-appearing lesions. As is typical in FD, this patient has had persistently normal vision despite encasement of the optic nerves.  

 

The natural course of FD is to progress during childhood and adolescence. Patients have grossly normal skeletal development in utero, without obvious features of bone disease at birth.  Skeletal lesions appear during the first several years of life and expand during linear growth.  The majority of FD lesions are apparent by age 10 years, typically with no new clinically significant lesions occurring after age 15 years [11]. FD lesions typically become less active in adulthood, which is mirrored histologically by age-dependent apoptosis of GNAS mutation-bearing cells in older patients [12], and biochemically by a progressive decline in bone turnover markers over time [13].

 

Malignant transformation occurs rarely in FD, consistent with the slight increased oncogenic potential associated with GNAS mutations [14]. Malignancies appear to be more common in patients exposed to ionizing radiation, including those receiving pituitary radiation for treatment of growth hormone excess [15, 16]. Clinicians should be alerted to the possibility of malignant transformation in lesions that are rapidly expanding or show signs of cortical disruption on radiographs. Another potential complication of FD is aneurysmal bone cysts, which typically present with acute onset of expansion and localized pain. These lesions may progress rapidly, and frequently require urgent surgical treatment.

 

EVALUATION AND MANAGEMENT

 

FD is typically diagnosed based on clinical and radiologic evaluation.  All patients with FD should undergo a complete skeletal evaluation at the time of diagnosis to determine the extent of the disease. This is best accomplished with technetium-99 scintigraphy or 18F-NaF PET/CT imaging to identify the areas of involvement, followed by radiographs of affected areas to better visualize lesions anatomically.

 

The differential diagnosis in patients with FD/MAS is variable depending upon the clinical presentation. Patients with prominent skin findings may be misdiagnosed with neurofibromatosis type 1 (NF-1), which also presents with café-au-lait macules and skeletal abnormalities. The location and distribution of skin lesions in NF-1 includes multiple smooth-bordered macules, which are generally distinct from the jagged bordered lesions in MAS that typically respect the midline of the body. Skeletal findings in NF-1 are less common and include tibial pseudoarthroses and kyphoscoliosis. There are multiple types of fibro-osseous lesions with similar radiographic and histologic features to FD, including ossifying fibromas, osteofibrous dysplasia, and giant cell tumors of bone [17, 18]. These lesions are generally solitary and not associated with extraskeletal features. 

 

The utility of molecular testing for diagnostic purposes should be considered on a case-by-case basis. GNAS mutation detection is highly variable depending upon the level of mosaicism and the sensitivity of the sequencing technique [1, 19].  In patients with monostotic FD and no additional features of MAS, mutation testing may be helpful in distinguishing FD from other fibro-osseous skeletal lesions. In patients with polyostotic disease and/or typical extraskeletal features of MAS, mutation testing is not required to establish the diagnosis, and is unlikely to inform clinical management.  

 

Management in FD is focused on treating endocrinopathies, preventing fractures, optimizing function, and treating pain. There are currently no medical therapies which are capable of altering the disease course. Orthopedic surgery is an important component of management however, practitioners should be aware that techniques used in other skeletal disorders may be unsuccessful in FD. In particular bone grafting, curettage, and external fixation are commonly used techniques that were previously considered standard in FD, but are now known to be frequently ineffective [20]. Diagnosis and treatment of scoliosis is extremely important, as it may be rapidly progressive and lead to fatal respiratory compromise. Surgical fusion has been shown to be effective in treating FD patients with progressive scoliosis [13].  

 

Surgical management of craniofacial FD lesions should similarly be approached cautiously, as complete resection is typically not possible, and partially resected lesions frequently regrow post-operatively [21]. Optic canal involvement is common but only rarely leads to vision loss (Fig 3). Conservative management of optic canal FD has been shown to be superior in asymptomatic patients; prophylactic decompression without objective evidence of optic neuropathy may lead to vision loss and is contraindicated [22]. All patients with craniofacial FD should undergo baseline and yearly ophthalmologic, otolaryngologic, and audiologic evaluations. Patients with FD involving the skull base should also be undergo screening imaging for cranial base deformities around skull age, and should be evaluated regularly for associated neurologic symptoms [7].

 

Evaluation and treatment for extraskeletal features of MAS is an important component of clinical management in FD. Patients should undergo a staging workup at the time of diagnosis, including complete history and physical examination, review of growth curves, biochemical testing for endocrinopathies, and ultrasonography of the thyroid and testes [23]. 

 

Hyperthyroidism may have a deleterious effect on bone density, increasing the risk of skeletal deformities in patients with FD [7, 10]. Hyperthyroidism is treated with anti-thyroidal drugs and thyroidectomy [23]. Peripheral precocious puberty results in increased linear growth and premature epiphyseal maturation, which may compromise final adult height. Symptoms are generally well-controlled with the aromatase inhibitor letrozole in girls, which may be used in combination with a testosterone blocker in boys, as well as leuprolide in children with co-existent central precocious puberty [24-26]. Growth hormone excess has been shown to increase FD expansion, particularly in the craniofacial region, leading to an increased risk of macrocephaly and vision loss [27, 28]. This risk may be partially ameliorated with early diagnosis and treatment [29]. Therapeutic options include somatostatin analogues and pegvisomant, with total hypophysectomy generally reserved for refractory cases [30].

 

FGF23-mediated hypophosphatemia may have a significant impact on disease severity in FD.  Although urinary phosphate wasting is common in FD patients, frank hypophosphatemia occurs less frequently due to a compensatory increase in cleavage of FGF23 into its inactive fragments [31]. The degree of FGF23 overproduction appears to be related to overall disease burden, with frank hypophosphatemia occurring more frequently in individuals with greater amounts of FD. The course of FGF23-mediated hypophosphatemia in any individual may therefore be variable, with low phosphate levels “unmasked” during periods of rapid growth or disease progression. Hypophosphatemia may also potentially normalize as growth and disease activity wanes. In addition to the classic rachitic findings of growth deceleration and long bone deformities, FD patients with uncontrolled hypophosphatemia are also at higher risk for fractures [32], deformities [7, 10] and bone pain [33]. Treatment is similar to other disorders of FGF23 excess, and includes oral phosphorus and calcitriol. 

 

Bone pain is a common feature of FD, and pain management is an important component of clinical care. The pathophysiology of FD-related bone pain is not well-understood [34], and there does not appear to be a clear relationship between pain and overall FD burden [33]. The prevalence and severity of pain is generally greater in adults, however pain is frequently unrecognized and untreated in children [33]. In patients who present with pain, it is important to exclude acute injury, impending fracture, or other causes that may require orthopedic intervention. Patients should also be evaluated and treated for hypophosphatemia, in which pain is frequently the presenting symptom. In the absence of structural or metabolic causes, a reasonable management strategy for bone pain is to use a step-wise approach, starting with conservative measures such as non-steroidal anti-inflammatory medications, rest, and application of heat or cold packs. In pain not responsive to conservative measures, intravenous bisphosphonates such as zoledronic acid or pamidronate may be helpful. In general, these should be administered based on symptoms rather than a set schedule, using the lowest effective dose and interval. 

 

A number of reports have investigated the efficacy of bisphosphonates, medications which increase bone density by inhibiting bone-resorbing osteoclasts, as a potential treatment for FD.  Early uncontrolled case series reported subjective improvements in pain and variable effects on the radiographic appearance of FD lesions [35-37]. A placebo-controlled trial of the oral bisphosphonate alendronate showed no effects on pain or FD lesion appearance [38]. Retrospective analyses have demonstrated that bisphosphonates do not appear to prevent progression of FD lesions in children [13], and do not appear to prevent progression of spinal deformity in patients with scoliosis [10]. Concerningly, osteonecrosis of the jaw has been reported in patients with FD, with a prevalence of 5.4% in one series [39]. The role of bisphosphonates in FD management has not been fully elucidated; however, at present there is little evidence to support any effect of bisphosphonates on FD quality or lesion expansion, and the author recommends their use be limited to treatment of bone pain.   

 

FUTURE DIRECTIONS

 

Current therapeutic options for treatment of FD are inadequate, and there is a critical need to develop medical therapies capable of altering the disease course. Ongoing efforts include developing drugs to target activity of the mutant Gsα [40], and devising strategies for targeting mutant skeletal progenitor cells [41]. Recent evidence suggests a possible role for denosumab, a monoclonal inhibitor of receptor activator of nuclear kappa-B ligand, for treatment of FD. This medication has been approved for treatment of giant cell tumors [42], and showed preliminary positive effects on bone turnover and pain in case reports of FD [43-45]. Of concern are severe, life-threatening metabolic derangements associated with denosumab treatment in one FD patient [44]. Currently there is insufficient evidence to support safety or efficacy of denosumab treatment for FD, and its use should be limited to research protocols.

 

ACKOWLEDGEMENTS

 

This research was supported by the Intramural Research Program of the NIH, NIDCR.

 

REFERENCES

 

  1. Lumbroso, S., F. Paris, and C. Sultan, Activating Gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome--a European Collaborative Study. J Clin Endocrinol Metab, 2004. 89(5): p. 2107-13.
  2. Idowu, B.D., et al., A sensitive mutation-specific screening technique for GNAS1 mutations in cases of fibrous dysplasia: the first report of a codon 227 mutation in bone. Histopathology, 2007. 50(6): p. 691-704.
  3. Happle, R., The McCune-Albright syndrome: a lethal gene surviving by mosaicism. Clin Genet, 1986. 29(4): p. 321-4.
  4. Riminucci, M., et al., The histopathology of fibrous dysplasia of bone in patients with activating mutations of the Gs alpha gene: site-specific patterns and recurrent histological hallmarks. J Pathol, 1999. 187(2): p. 249-58.
  5. Riminucci, M., et al., Osteoclastogenesis in fibrous dysplasia of bone: in situ and in vitro analysis of IL-6 expression. Bone, 2003. 33(3): p. 434-42.
  6. Riminucci, M., et al., FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest, 2003. 112(5): p. 683-92.
  7. Pan, K.S., et al., Chiari I Malformation and Basilar Invagination in Fibrous Dysplasia: Prevalence, Mechanisms, and Clinical Implications. J Bone Miner Res, 2018. 33(11): p. 1990-1998.
  8. Leet, A.I., et al., Fibrous dysplasia in the spine: prevalence of lesions and association with scoliosis. J Bone Joint Surg Am, 2004. 86-A(3): p. 531-7.
  9. Mancini, F., et al., Scoliosis and spine involvement in fibrous dysplasia of bone. Eur Spine J, 2009. 18(2): p. 196-202.
  10. Berglund, J.A., et al., Scoliosis in Fibrous Dysplasia/McCune-Albright Syndrome: Factors Associated with Curve Progression and Effects of Bisphosphonates. J Bone Miner Res, 2018.
  11. Hart, E.S., et al., Onset, progression, and plateau of skeletal lesions in fibrous dysplasia and the relationship to functional outcome. J Bone Miner Res, 2007. 22(9): p. 1468-74.
  12. Kuznetsov, S.A., et al., Age-dependent demise of GNAS-mutated skeletal stem cells and "normalization" of fibrous dysplasia of bone. J Bone Miner Res, 2008. 23(11): p. 1731-40.
  13. Florenzano, P., et al., Age-Related Changes and Effects of Bisphosphonates on Bone Turnover and Disease Progression in Fibrous Dysplasia of Bone. J Bone Miner Res, 2019.
  14. Ruggieri, P., et al., Malignancies in fibrous dysplasia. Cancer, 1994. 73(5): p. 1411-24.
  15. Hansen, M.R. and J.C. Moffat, Osteosarcoma of the Skull Base after Radiation Therapy in a Patient with McCune-Albright Syndrome: Case Report. Skull Base, 2003. 13(2): p. 79-83.
  16. Liu, F., et al., A case of McCune-Albright syndrome associated with pituitary GH adenoma: therapeutic process and autopsy. J Pediatr Endocrinol Metab, 2011. 24(5-6): p. 283-7.
  17. Kumar, K.A., et al., Management and Treatment Outcomes of Maxillofacial Fibro-osseous Lesions: A Retrospective Study. J Maxillofac Oral Surg, 2015. 14(3): p. 728-34.
  18. Steffner, R., Benign bone tumors. Cancer Treat Res, 2014. 162: p. 31-63.
  19. Narumi, S., et al., Quantitative and sensitive detection of GNAS mutations causing mccune-albright syndrome with next generation sequencing. PLoS One, 2013. 8(3): p. e60525.
  20. Stanton RP, I.E., Springfield D, Lindaman L, Wientroub S, Leet A., The surgical management of fibrous dysplasia of bone. Orphanet J Rare Dis, 2012. 24 (7): p. Suppl 1:S1.
  21. Boyce, A.M., et al., Surgical Management of Polyostotic Craniofacial Fibrous Dysplasia: Long-Term Outcomes and Predictors for Postoperative Regrowth. Plast Reconstr Surg, 2016. 137(6): p. 1833-9.
  22. Amit, M., et al., Surgery versus watchful waiting in patients with craniofacial fibrous dysplasia--a meta-analysis. PLoS One, 2011. 6(9): p. e25179.
  23. Boyce, A.M. and M.T. Collins, Fibrous Dysplasia/McCune-Albright Syndrome, in GeneReviews(R), R.A. Pagon, et al., Editors. 1993, University of Washington, Seattle: Seattle WA.
  24. Feuillan, P., et al., Letrozole treatment of precocious puberty in girls with the McCune-Albright syndrome: a pilot study. J Clin Endocrinol Metab, 2007. 92(6): p. 2100-6.
  25. Boyce, A.M., et al., Characterization and management of testicular pathology in McCune-Albright syndrome. J Clin Endocrinol Metab, 2012. 97(9): p. E1782-90.
  26. Estrada, A., et al., Long-term outcomes of letrozole treatment for precocious puberty in girls with McCune-Albright syndrome. Eur J Endocrinol, 2016. 175(5): p. 477-483.
  27. Roszko, K.L., M.T. Collins, and A.M. Boyce, Mosaic Effects of Growth Hormone on Fibrous Dysplasia of Bone. N Engl J Med, 2018. 379(20): p. 1964-1965.
  28. Akintoye, S.O., et al., Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. J Clin Endocrinol Metab, 2002. 87(11): p. 5104-12.
  29. Boyce, A.M., et al., Optic Neuropathy in McCune-Albright Syndrome: Effects of Early Diagnosis and Treatment of Growth Hormone Excess. J Clin Endocrinol Metab, 2012.
  30. Salenave, S., et al., Acromegaly and McCune-Albright syndrome. J Clin Endocrinol Metab, 2014: p. jc20133826.
  31. Bhattacharyya, N., et al., Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner Res, 2012. 27(5): p. 1132-41.
  32. Leet, A.I., et al., Fracture incidence in polyostotic fibrous dysplasia and the McCune-Albright syndrome. J Bone Miner Res, 2004. 19(4): p. 571-7.
  33. Kelly, M.H., B. Brillante, and M.T. Collins, Pain in fibrous dysplasia of bone: age-related changes and the anatomical distribution of skeletal lesions. Osteoporos Int, 2008. 19(1): p. 57-63.
  34. Chapurlat, R.D., et al., Pathophysiology and medical treatment of pain in fibrous dysplasia of bone. Orphanet J Rare Dis, 2012. 7 Suppl 1: p. S3.
  35. Liens, D., P.D. Delmas, and P.J. Meunier, Long-term effects of intravenous pamidronate in fibrous dysplasia of bone. Lancet, 1994. 343(8903): p. 953-4.
  36. Chapurlat, R.D., et al., Treatment of fibrous dysplasia of bone with intravenous pamidronate: long-term effectiveness and evaluation of predictors of response to treatment. Bone, 2004. 35(1): p. 235-42.
  37. Plotkin, H., et al., Effect of pamidronate treatment in children with polyostotic fibrous dysplasia of bone. J Clin Endocrinol Metab, 2003. 88(10): p. 4569-75.
  38. Boyce, A.M., et al., A randomized, double blind, placebo-controlled trial of alendronate treatment for fibrous dysplasia of bone. J Clin Endocrinol Metab, 2014: p. jc20141371.
  39. Metwally, T., et al., Fibrous Dysplasia and Medication-Related Osteonecrosis of the Jaw. J Oral Maxillofac Surg, 2016. 74(10): p. 1983-99.
  40. Bhattacharyya, N., et al., A high throughput screening assay system for the identification of small molecule inhibitors of gsp. PLoS One, 2014. 9(3): p. e90766.
  41. Piersanti, S., et al., Transfer, analysis, and reversion of the fibrous dysplasia cellular phenotype in human skeletal progenitors. J Bone Miner Res, 2010. 25(5): p. 1103-16.
  42. Singh, A.S., N.S. Chawla, and S.P. Chawla, Giant-cell tumor of bone: treatment options and role of denosumab. Biologics, 2015. 9: p. 69-74.
  43. Benhamou, J., D. Gensburger, and R. Chapurlat, Transient improvement of severe pain from fibrous dysplasia of bone with denosumab treatment. Joint Bone Spine, 2014. 81(6): p. 549-50.
  44. Boyce, A.M., et al., Denosumab treatment for fibrous dysplasia. J Bone Miner Res, 2012. 27(7): p. 1462-70.
  45. Ganda, K. and M.J. Seibel, Rapid biochemical response to denosumab in fibrous dysplasia of bone: report of two cases. Osteoporos Int, 2014. 25(2): p. 777-82.

 

Emergencies in Childhood Diabetes

INTRODUCTION

 

Diabetes Mellitus, after asthma, is the second most common chronic condition in children.  It can be diagnosed at any age and the incidence has been rising. Type 1 Diabetes (T1D) is by far the most common type of diabetes in children and its incidence has been increasing slowly but steadily. By definition T1D is an autoimmune condition caused by immune-mediated destruction of beta cells, with resultant development of pancreatic auto-antibodies (Type 1a).  However, 10-15% of patients with a clinical phenotype of T1D do not have detectable antibodies and are classified as having Type 1b.  Besides autoimmunity, beta cell loss can be caused by absence or loss of the pancreas, cystic fibrosis, and drug-induced diabetes.  In these cases, the management of diabetes is very similar to the management of autoimmune T1D.  Type 2 Diabetes (T2D) in children is characterized by progressive loss of beta cell function due to severe insulin resistance and often requires insulin therapy.  The incidence of T2D has also been increasing in children, commensurate with the rise of obesity. Therefore, the key to treating most children with diabetes is replacement of insulin and most of the emergencies in children with diabetes are associated with either inadequate or excess insulin administration.  

 

NEW ONSET DIABETIC KETOACIDOSIS (DKA)

Clinical Recognition

 

The onset of T1D can be insidious because the immune-mediated loss of b-cells is typically slow, occurring over several months to years before hyperglycemia is manifest.  The earliest signs of hyperglycemia begin with polyuria, followed shortly by polydipsia.  A typical scenario in children is that of a few weeks’ history of increasing fatigue, intermittent polyuria and polydipsia, a recent viral illness and/or travel followed by exacerbation of these symptoms.  If not recognized at this stage, hyperglycemia further impairs b-cell function and insulin secretion, which eventually leads to increasing lipolysis to provide free fatty acids as an alternative substrate for energy generation.  Oxidation of free fatty acids leads to accumulation of acetoacetic and b-hydroxybutyric acids (ketones).  When the level of ketones exceeds the child’s capacity to buffer the acidosis, the blood pH begins to decrease to below 7.3.  In addition, the osmotic diuresis caused by the hyperglycemia leads to dehydration and lactic acidosis which further contributes to the acidosis and to increasing insulin resistance, all leading to a vicious cycle and rapid worsening in the hemodynamic status and development of severe DKA.  The severity of DKA is divided into mild (pH 7.2-7.3, or bicarbonate <15 mmol/L), moderate (pH 7.1-7.2, or bicarbonate <10 mmol/L), and severe (pH <7.1, or bicarbonate <5 mmol/L).  The younger the child the faster the progression towards DKA, and the more severe the acidosis can be, with ensuing classic signs of dehydration, Kussmaul breathing, and potential obtundation and even coma.

 

Diagnosis

 

The diagnosis of new onset DKA relies on a high index of suspicion because most children presenting with new onset T1D have no family history of T1D.  In addition, infants and toddlers may not have the classical series of symptoms that usually precede presentation with DKA and may present with non-specific symptoms such as increased irritability, difficulty sleeping, or poor feeding.  Because the initial hyperglycemia can be exacerbated by a viral illness, these non-specific symptoms can be attributed to the viral illness and not to diabetes.  Therefore, a careful review of the history of the illness and the presenting symptoms is essential for making the right diagnosis.  Important clues include increased number of daily diapers or parental description of “heavy diapers especially in the morning”, the presence of diaper rash in infants or vaginal yeast infection in older girls, increased frequency of using the bathroom during school, nocturnal enuresis in a child who has already been trained, increased appetite yet continued weight loss, deterioration of performance in sports or school, and the presence of a sweet fruity smell from the child.

Once suspected, new onset T1D is easily confirmed by obtaining a basic metabolic panel which includes glucose and bicarb measurements.  A glucose level >200 mg/dl, with symptoms of hyperglycemia confirms the diagnosis of diabetes, while a lower than normal bicarb level suggests the presence of DKA, which can be confirmed by measuring urine or serum ketone levels. 

 

Treatment

 

Once diagnosed, new onset DKA in children is best managed if possible in a pediatric specialty center with expertise in treating children with DKA.  This is due to the fact that a) fluid replacement in children must be calculated with greater precision based on body weight and surface area, and be provided in a manner that minimizes the risk of rapid shifting in osmolality between intra- and extracellular spaces, which can occur more easily in children because of their relatively immature hemodynamic and cerebral autoregulatory processes.  Rapid shifting of fluids can lead to cerebral edema, the most serious complication of DKA especially in younger children presenting with new onset T1D; b) children, especially pre-pubertal children, are far more sensitive to insulin, requiring much smaller doses than what is usually needed in adults; and c) even within the pediatric population, the approach to managing DKA in infants and toddlers is quite different than in older patients. Adolescents in peak puberty present a different challenge because of their often-severe resistance to insulin.  For these and other reasons, we strongly recommend that children in DKA get transferred to a center with pediatric intensive care with the ability to provide close monitoring and management according to childhood specific treatment protocols.  If initial stabilization of the child is required before transfer, it should be done in consultation with a pediatric intensivist and/or endocrinologist if possible.

The first steps in the treatment of DKA are shown in Table 1.

 

Table 1. The First Steps in the Treatment of DKA

1) Determine current weight and height (or length) of the child, to calculate body surface area.

2) A careful physical examination looking for signs of infection, the presence of acanthosis nigricans, and assess the neurological status and degree of dehydration.

3) Initial blood sampling for laboratory measurements of electrolytes and glucose, BUN, creatinine, a blood gas, ketones, and a complete blood count.

4) Provide oxygen if needed.

5) Obtain samples for culturing if evidence of infection exists.  Antibiotics should be considered if the child is febrile.

6) Establish peripheral intravenous access.  Central line access is rarely needed

 

FLUID REPLACEMENT

 

All children presenting with DKA have some degree of dehydration, and must receive fluid replacement immediately.  When thinking of fluid replacement, a few important principles should be kept in mind:

a) Estimating total fluid deficit in a child with DKA can be difficult: High concentrations of glucose in the extracellular space pulls water out of the cells and the osmotic diuresis maintains a relatively large urine output even when severe fluid depletion has occurred. Therefore, intravascular volume and urinary volume are not good indicators of the degree of total fluid deficit.  A decreased urine excretion in the face of hyperglycemia is often a late sign of severe dehydration.  Therefore, it is a good practice to start with an assumption of 10% dehydration in patients with severe DKA and 5-7% dehydration with moderate DKA. 

b) Serum sodium is often falsely low due to dilution of the extracellular fluid (ECF) caused by high glucose concentration in this compartment.  Corrected, true sodium concentration can be calculated by adding 1.6 x current measured serum glucose – 100 to the measured serum sodium. Serum urea nitrogen and hematocrit levels are more reliable indicators of the status of the ECF and can be used for monitoring changes in the ECF after fluid replacement.
 
c) While fluid repletion is essential, caution must be taken as to the speed of fluid administration.  Rapid fluid infusion can result in rapid changes in the differential osmolality between the ECF and intracellular space, leading to a shift of water into the cells which is the main cause of cerebral edema.  Naturally, when the peripheral circulation is compromised, resuscitation should be provided as quickly as possible using normal saline (or Ringer’s lactate solution).  Once adequate circulation and tissue perfusion have been established, it is essential to slow down the rates of fluid replacement.  Additional boluses of fluids should be given over 1-2 hours whenever possible.  Subsequently, fluid administration for deficit replacement must be provided evenly over the next 48 hours, and should not include replacement of ongoing urinary losses.  At this stage, normal saline can be used for the initial 6 hours, followed by half normal saline, with added potassium.

d) Fluid administration will result in a significant decrease in glucose concentration.  Once serum glucose has decreased to 250-300 mg, a glucose-containing solution (5-12.5% dextrose) can be added to the saline solution as part of the fluid replacement.  This is aimed at slowing down the rate of decrease in serum osmolality and to provide sufficient substrate for administering insulin (see below).

 

INSULIN

A major component of the acidosis in DKA is caused by the accumulation of ketones which is the result of absent or insufficient insulin secretion or action.  While rehydration improves the hyperglycemia and acid-base status, it is insufficient to correct the pH back to normal.  Insulin administration is essential for restarting glucose metabolism and halting fatty acid oxidation and further ketone production.

Because children’s sensitivity to insulin varies markedly, starting with an intravenous insulin infusion provides the ability to titrate rapidly in response to changes in serum glucose, primarily to avoid hypoglycemia and relative rapid drops in serum glucose.  Most children can start at a rate of 0.05-0.1 unit/kg/hour.  Smaller children often require the lower end of this range; However, it is not surprising to occasionally see a toddler requiring up to 0.8 unit/kg/hour in the presence of a concomitant infection.

Insulin infusion rates can be titrated down if serum glucose concentrations are decreasing, but only if acidosis is correcting and the anion gap is closing.  If not, it is important to keep the insulin infusion rates higher and increase the glucose infusion rate to raise serum glucose above 200 mg/dL.  Failure to provide adequate insulin delays normalization of the acid base status.

 

POTASSIUM REPLACEMENT

An important consideration is potassium (K) replacement.  Most children presenting with new onset DKA have a relative depletion of intracellular potassium even if initial serum K is in the normal range.  Once hydration is provided and insulin is started, K shifts from the ECF into the intracellular space resulting in hypokalemia and potential arrythmias.  Thus, K should always be added into the fluids administered for the slow phase of rehydration.  On the contrary, it has been established that administration of bicarbonate should not be routine, and in fact may be harmful to the CNS, except in extreme cases with severe acidosis (pH <6.9) and severe hypokalemia.

 

LABORATORY MONITORING

 

Initial laboratory monitoring should include a CBC, a chemistry panel (Na, K, Cl, bicarb, BUN, Creatinine, glucose, Ca, Phosphate, Mg), and a venous blood gas.  The chemistry panel and blood gas should be done hourly initially, and can be reduced in frequency to every 2-4 hours after significant improvement in the clinical and the acid base status of the child.  After complete resolution of the acidosis, laboratory monitoring can be less frequent, and should focus on serum K and phosphate.  Assessment of serum glucose must continue at intervals of 3-4 hours and additional rapid acting insulin is provided when serum glucose values exceed 200 mg/dl.

 

Additional laboratory testing, which can be done after stabilization and resolution of the DKA, includes pancreatic auto-antibodies, thyroid function tests, serum total IgA level, and a tissue transglutaminase antibody measurement to screen for Celiac disease.  Pancreatic auto-antibodies against the following 5 antigens should be included: Islet cells, GAD 65, Zn-T8, IA-2 (or ICA 512), and insulin. TSH and Free T4 should be done only after complete resolution of the acute phase of the illness and any concomitant infection to avoid falsely abnormal values.

 

Follow-Up

After recovery from the DKA and normalization of pH and anion gap, the child can be transitioned from intravenous to subcutaneous insulin administration.  It is essential to recognize that subcutaneous insulin takes time to begin and reach peak action, which means that intravenous insulin should be continued for a few hours after the first injection of insulin.  Ideally, iv insulin should be continued until 1-2 hours after the first injection of SQ rapid acting insulin (Aspart or Lispro), or 4 hours after the first injection of long acting insulin (Glargine or Detemir).

Although it is obviously important to differentiate between children with new onset T1D and T2D because of the implications for long term therapy, the initial management of DKA in both populations should not be different.  With increasing rates of obesity in children, more youth are presenting with T2D at younger ages.  It is thought that children who develop T2D at a younger age do so because of more progressive beta cell failure than in adults.  Thus, the initial DKA at presentation with T2D is physiologically and biochemically similar to DKA in new onset T1D, with the main targets of therapy being fluid and insulin replacement.  Measuring pancreatic auto-antibodies and documentation of careful family history, will eventually aid in differentiating T1D from T2D, especially in children who are overweight or obese.  

RECURRENT DKA

When DKA occurs in a child with an established diagnosis of diabetes, it is almost always due to insulin omission (for a variety of reasons), inadequate administration of insulin, or relative insulin insufficiency due to inadequate adjustment of insulin dosing in the context of conditions that cause temporary insulin resistance, such as generalized or localized infections.

With wider use of insulin pumps, one of the most common causes of recurrent DKA, especially in young children, is unrecognized pump failure.  Patients treated with an insulin pump receive only rapid acting insulin as a continuous infusion with superimposed boluses for meals and for correcting high blood glucose, without the use of long acting insulin.  When pump failures occur, delivery of rapid acting insulin stops, and ketones can begin to accumulate within 2-4 hours.  If unrecognized and uncorrected within this time frame, mild-moderate DKA can result, with varying degrees of symptomatology, from minor nausea and stomach ache, to vomiting, headache, and acidosis.

Pump failures are often mechanical, ranging from a kinked or clogged catheter of the infusion set, to presence of air bubbles in the tubing (which can interrupt delivery of insulin for several hours, especially in small children), dislodging of the infusion set, or simply a spontaneous pump malfunction.  Other common causes include use of ineffective insulin that has been degraded or contaminated, injection of insulin (by a pump or syringes) into a severely hypertrophied area, which interferes with insulin absorption, errors in programming the pump, and running out of insulin or battery in the pump, especially at night.

Management of DKA in these cases is similar to new onset DKA, often associated with faster recovery and return to baseline once insulin administration is initiated.  It is important in such cases to work with the child and family to identify and remedy the cause of the DKA.  Whenever possible, the treating physician should communicate with the diabetes provider(s) to arrange for prompt follow up with emphasis on specific training to avoid recurrence in the future.

Another common cause of recurrent DKA in adolescents is intentional insulin omission.  Particular attention should be given to adolescents with history or evidence of eating disorders and recent weight loss, as they may be more sensitive to insulin than anticipated.  Psychological evaluations and therapy should be arranged upon discharge of these patients.

 

DKA WITHOUT HYPERGLYCEMIA IN T1D



These cases are actually more challenging than those presenting with hyperglycemia, and are more prone to mismanagement.  A typical scenario is that of a young child with T1D, who has not been eating well for at least one day due to a gastro-intestinal infection with or without vomiting and diarrhea.  In young children, a greater proportion of the daily insulin is given for carbohydrate consumption, up to 75% of total daily dose.  Therefore, a child who is not eating even for one day will miss significant amounts of insulin and begin the process of lipolysis very quickly and become ketonemic.  The lack of food consumption is associated with euglycemia. Fluid loss in euglycemic DKA is modest because of the absence of an osmotic diuresis. If fasting is prolonged with extended consumption of glycogen stores, hypoglycemia can develop, along with ketonemia and acidosis.

A careful history review with the parents will clarify these scenarios and direct the management of DKA towards earlier start of intravenous glucose infusion and insulin administration to block further production of ketones.  In these cases, even if the child is able to take some fluids orally, intestinal absorption is generally not optimal and should not be relied upon.  With adequate intravenous hydration with intravenous or SQ insulin, the DKA can resolve fairly quickly.

These scenarios are not limited to small children and can certainly occur in older children and even adolescents with poor oral intake due to a GI illness, prolonged fasting, or severe carbohydrate restriction.  The management principles are similar and rely on providing glucose which in turn allows for more insulin dosing.  The latter can be provided by intravenous infusion, SQ intermittent dosing, or even with an insulin pump if the child already has one, as long as there is adequate peripheral tissue perfusion to ensure appropriate absorption of insulin.

An emerging cause of euglycemic DKA is the use of a new class of drugs, the SGLT-2 inhibitors.  Although still rare in children, some older adolescents with T1D or T2D are prescribed SGLT-2 inhibitors, which primarily cause forced glucosuria, preventing a rise in blood glucose, even when insulin boluses for food are omitted.  Management of these cases is similar to what is described above in this section, and patients should be counseled against use of SGLT-2 inhibitors without further consultation with a pediatric endocrinology team.

One final note is in regard to patients who are using a continuous glucose monitor (CGM).  With significantly improved accuracy of CGM devices, more and more pediatric patients are using CGM, which can be used for treatment decisions.  However, in the presence of moderate to severe dehydration and impaired tissue perfusion, CGM readings may not be accurate and should not be relied upon in an emergency setting.

 

DKA IN PATIENTS WITH T2D



As stated above, generally speaking, when children develop T2D at such a young age, they are likely to have lost significant endogenous beta cell function.  Most children with T2D are either treated with metformin alone, insulin alone, or a combination.  Those who require insulin have little endogenous insulin and can develop DKA in the same way children with T1D do, and their DKA should be treated in the same way.  Children on metformin or metformin + insulin, may have some insulin secretion capacity, but do not have the ability to increase their insulin production at times of physiological stress which exacerbates their insulin resistance.  Therefore, if they do present with DKA, this is an indication of relative insulin insufficiency and they should also be treated similarly to children with T1D.  These scenarios include a special group labelled ketosis-prone T2D, who can be effectively treated with metformin alone, but have a tendency to develop DKA despite sufficient insulin production capacity at baseline conditions.

Because of their general insulin resistance, children with T2D presenting with recurrent DKA generally have concomitant hyperglycemia and rarely present with DKA and euglycemia except when treated with SGLT-2 inhibitors as described above.

A special group that is being recognized more often lately are children with monogenic diabetes, who can eventually progress towards partial insulin insufficiency and can present for the first time with either new onset or recurrent DKA.  In particular, maturity onset diabetes of the young (MODY) type 1 and 3 are the most common.  They usually have negative pancreatic auto-antibodies, their phenotype is not suggestive of insulin resistance, and there is a strong family history of non-type 1 diabetes in several generations.  Once again, when presenting in DKA their management follows the same principles.

HYPOGLYCEMIA



Hypoglycemia is the most common acute complication of diabetes in children and represents the greatest challenge in managing children with diabetes.  Surprisingly, the definition of hypoglycemia in children remains controversial and somewhat nebulous, and varies for different ages.

 

Clinical Recognition

 
While in the general population, a blood glucose (BG) level of <70 mg/dL is considered low (hypoglycemia) and results in clinical symptoms, children with diabetes spend significant time with BG levels above 200 mg/dL, thus shifting upward the brain threshold for exhibiting signs and symptoms of hypoglycemia.  Conversely, recent episodes of hypoglycemia can shift downward the threshold for exhibiting and recognizing hypoglycemia.  Recent consensus statements from the American Diabetes Association (ADA) and the International Society for Pediatric and Adolescent Diabetes (ISPAD) have defined hypoglycemia in a child with diabetes as any BG level that results in symptoms or signs of impaired cognitive function.  These hypoglycemic levels frequently change as the range of BG levels shift up or down over time, even within short periods of time.

In the context of diabetes, any level of hypoglycemia (resulting in symptoms) can be considered an emergency because of the potential for the BG level to drop further within minutes.  Therefore, any BG <70-80 mg/dL is generally considered a “hypoglycemia alert” and should be managed urgently.  This is particularly true in children with diabetes because of their higher sensitivity to both insulin and physical activity, resulting in more rapid drops in BG levels. 

The current consensus is that a true emergency is considered a BG <54 mg/dL (3 mMol/L), a level that indicates a clinically important hypoglycemia that is likely to be associated with defective counter-regulatory response and impaired cognitive function.  At these levels, a child is at a much higher risk for more severe cognitive impairment, with or without loss of consciousness or seizure, and requiring external assistance (severe hypoglycemia).

 

Pathophysiology



By far the most common cause of hypoglycemia in children with diabetes is excess insulin.  Most children are treated with intensive insulin therapy with a basal/bolus regimen, by multiple daily injections or via an insulin pump.  Boluses of rapid acting insulin are determined for each meal based on current BG value at the time before the meal and on the amount of carbohydrates (carbs) to be consumed in that meal.  This requires fairly accurate carb counting, followed by entry of the estimated carbs and the BG number into a dose calculator, which can be a simple sliding scale hard copy sheet, a smart phone app, or a smart insulin pump.  This process involves multiple steps, which naturally presents opportunities for estimation and transcription errors, leading sometimes to over-dosing of insulin.

A common scenario, especially in young children, is refusal to eat or to finish a meal after a bolus had been given, or vomiting shortly after a meal.  A history of repeated episodes of vomiting and/or repeated hypoglycemia after meals in an adolescent should alert the provider to the possibility of an eating disorder.

The second frequent cause of hypoglycemia in children is exercise.  Patients and caregivers are trained on adjusting insulin dosing to compensate for the enhanced sensitivity to insulin during and after any physical activity.  These include lowering doses of long acting insulin or rapid acting insulin, lowering a bolus amount for the meal before or after exercise, or consuming extra carbs which are not dosed for.  For patients on insulin pumps, a temporary basal rate reduction for several hours beginning before the activity is planned is often necessary.  Omission of such adjustments can lead to hypoglycemia which can occur during or up to few hours after the exercise.

 

Management

Most hypoglycemic episodes in children should be managed in a timely manner, at the patient location, such as home, school, or sports fields, to avoid extended brain deprivation of glucose and prevent acute and long-term sequelae.  Parents of children and persons with diabetes who are taking insulin or insulin secretagogues (sulfonylureas), should be trained to recognize the signs and symptoms of hypoglycemia and how to manage hypoglycemia.

Once a child exhibits signs of hypoglycemia, treatment should be initiated after measuring the BG regardless of level.  If the child is cooperative and able to take anything by mouth, simple and fast absorbing carbohydrates should be given.  Examples include glucose tablets, clear juice, soda, regular sugar, cake frosting, and a variety of candies.  In late adolescents, like in adults, 10-20 grams of carbs can be given to treat any hypoglycemia, and can be repeated 15 minutes later if needed.  However, in smaller children, it is imperative to recognize that smaller amounts of carbs should be used to prevent a rebound hyperglycemia.  As little as 2-4 grams are often sufficient in toddlers to raise the BG 30-60 mg/dL.

If a child is combative, unconscious, or seizing, oral treatment should not be attempted, as this carries the risk of aspiration or injury.  Instead, glucagon should be given intramuscularly as soon as possible.  For most children, a dose of 0.5 mg (half the amount provided in glucagon emergency kits) is effective in raising BG to >100 mg/dL, but up to 1.0 mg can be given to older adolescents.  Although rare, a dose of glucagon can be repeated within 30 minutes if the BG level does not rise above 100 mg/dL.

HYPERGLYCEMIC HYPEROSMOLAR STATES

The hyperglycemic hyperosmolar state (HHS) is very rare in children, but is worth reviewing because of the associated high mortality rate, which approximates 20%.  As the name indicates, HHS is characterized by severe hyperglycemia, hyperosmolarity, and dehydration in the absence of ketoacidosis.  Treatment of HHS is directed at replacing the usually severe fluid deficit and slowly correcting the hyperglycemia, osmolality, and electrolyte disturbances with low-dose insulin infusion similar to treating DKA.

 

Pathophysiology



HHS is typically initiated by severe hyperglycemia caused by both relative insulin deficiency and elevation of counter regulatory hormones, which cause a decrease in peripheral glucose utilization and increase glucose production, respectively.  The resulting severe hyperglycemia leads to polyuria and severe dehydration, with subsequent decrease in glomerular filtration and glucose clearance, which further exacerbates the hyperglycemia and hyperosmolality.  The absence of ketones in HHS is attributed to the relative but not absolute deficiency of insulin, and the higher insulin to glucagon ratio than that in DKA. 

As in adults, HHS occurs in children with T2D, but it can also be the first presentation of new onset T1D, caused by use of beverages with high carbohydrates, such as regular soda or juice, to alleviate the accompanying polydipsia.  Other common precipitating factors of HHS are infections, cystic fibrosis, and use of certain anti-psychotic medications.

Diagnosis



The progression of HHS is typically slower than DKA, resulting in delayed diagnosis and presentation with more severe hyperglycemia, dehydration, and altered mental status.  Diagnostic criteria for HHS include a BG >600 mg/dL (33.3 mmol/L), absence of appreciable acidosis (pH>7.30 and bicarb >15), anion gap <12, and an effective serum osmolality >320 mmol/kg.  Recent consensus guideline recommended calculating the effective serum osmolality as [2 (serum Na)] + [glucose in mmol/L].  This is based on the fact that altered mental status is usually manifest at serum Na >160 mmol/L, or calculated effective serum osmolality >320 mmol/L.

 

Treatment

The main objectives of treating HHS are very similar to treating DKA: restoration of circulatory volume and tissue perfusion; correction of hyperglycemia and electrolyte imbalance; and identification and treatment of the precipitating event(s).  Because HHS is rare in children, most published guidelines are based on experiences with adults.  Therefore, practical management of HHS in children follow the same guidelines as for DKA, including estimation of fluid deficit, careful replacement of the deficit after initial management of shock if present, keeping in mind that fluid administration results in significant decrease in BG concentration and drop in serum osmolality.  Once osmolality stops declining, insulin infusion can be started while fluid replacement continues, with the goal of maintaining BG at 250-300 mg/dL.

 

GUIDELINES

 

Wolfsdorf JI, Allgrove J, Craig ME, Edge J, Glaser N, Jain V, Lee WW, Mungai LN, Rosenbloom AL, Sperling MA, Hanas R; ISPAD Clinical Practice Consensus Guidelines 2014. Diabetic ketoacidosis and hyperglycemic hyperosmolar state. International Society for Pediatric and Adolescent Diabetes. Pediatr Diabetes. 2014 Sep;15 Suppl 20:154-79

 

Jones TW. On behalf of the ISPAD hypoglycemia guidelines writing group. Defining relevant hypoglycemia measures in children and adolescents with type 1 diabetes. Pediatr Diabetes. 2018; 19:354-355.

 

REFERENCES

 

Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis (DKA), And Hyperglycemic Hyperosmolar State (HHS). In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2018 May 17

 

Spanakis EK, Cryer PE, Davis SN. Hypoglycemia During Therapy of Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2018 Jun 18.

 

Umpierrez G, Korytkowski M. Diabetic emergencies - ketoacidosis, hyperglycaemic hyperosmolar state and hypoglycaemia. Nat Rev Endocrinol. 2016 Apr;12(4):222-32.

Dayna E. McGill & Lynne L. Levitsky. Management of Hypoglycemia in Children and Adolescents with Type 1 Diabetes Mellitus. Current Diabetes Reports (2016) 16(9): 88.

 

Dhatariya KK, Vellanki P. Treatment of Diabetic Ketoacidosis (DKA)/Hyperglycemic Hyperosmolar State (HHS): Novel Advances in the Management of Hyperglycemic Crises (UK Versus USA). Current Diabetes Reports. 2017 May;17(5):33.

 

Yau M, Sperling M. Treatment of Diabetes mellitus in Children and Adolescents. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2017 Sep 25

 

Bialo SR, Agrawal S, Boney CM, Quintos JB. Rare complications of pediatric diabetic ketoacidosis. World J Diabetes. 2015 Feb 15;6(1):167-74

 

Jefferies CA, Nakhla M, Derraik JG, Gunn AJ, Daneman D, Cutfield WS. Preventing Diabetic Ketoacidosis. Pediatr Clin North Am. 2015 Aug;62(4):857-71

 

 

 

Drugs that Affect Body Weight, Body Fat Distribution, and Metabolism

ABSTRACT

 

Weight gain or body fat redistribution are common side effects of many widely used drugs. Weight gain amounts varying between a few kg to an increase of 10% or more of initial body weight have been described. Often accompanying this weight gain are worsened health risks, including an increased incidence of the metabolic syndrome, type 2 diabetes, and other cardiovascular risk factors. With many drug classes, such as β-receptor antagonists, anti-psychotic drugs, corticosteroids, neurotropic drugs, and those used in the therapy of HIV, both significant weight gain and metabolic disturbances occur in susceptible patients.  In this review, we provide an overview of drugs that affect body weight, fat distribution, and metabolism. Attention is given to the possible pathogenic mechanisms underlying these effects and their metabolic consequences. Potential preventive, alternative, or therapeutic measures are suggested where applicable. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION

 

Many widely used drugs cause weight gain that—especially in susceptible individuals—may lead to patients becoming overweight or obese. Other drugs predominately influence body fat redistribution through increases in central adiposity, including visceral fat accumulation, and/or subcutaneous fat atrophy (lipodystrophy). Accompanying these changes are increases in insulin resistance, dyslipidemia, metabolic syndrome, and risk for type 2 diabetes (T2DM), non-alcoholic steatohepatitis (NASH), cardiovascular disease, cancer, and even increased mortality. These body weight and metabolic side-effects warrant close monitoring and potentially additional therapies to minimize their health impact, thereby increasing medical costs and contributing to non-compliance, which risks worsening of the underlying condition.

 

Weight gain is consistently associated with many older agents for the treatment of diabetes and with neuropsychotropic medications, including atypical antipsychotics, antidepressants, and antiepileptic drugs (1). For other drug classes, e.g. β-blocking agents, data are less consistent or well-studied. Glucocorticoids are associated with weight gain and lipodystrophy, as are retroviral agents used in the therapy of human immunodeficiency virus (HIV). Also, drugs used to manage lipid disorders, such as MTTP inhibitors and anti-sense apo-B oligonucleotides, are associated with changes in body fat distribution, especially liver lipid accumulation.  Unfortunately, the mechanisms behind these effects on body weight and fat distribution are often poorly understood, which hampers identification of high-risk patients for prevention, development of lower risk-drugs, and possible treatments (2).

 

In this chapter, drugs affecting body weight, fat distribution, and glucometabolic outcomes will be reviewed, as well as the possible mechanisms contributing to these side effects. Recent studies will be highlighted that have been undertaken to identify predictors of weight gain and metabolic complications, and where possible, options for prevention and therapy will be discussed.

 

DRUGS ASSOCIATED WITH WEIGHT GAIN     

 

Medications for Diabetes

 

Insulin, sulfonylurea (SU), and thiazolidinediones (TZD) are medications used in the management of diabetes that may cause substantial weight gain when compared to placebo (1). Metformin and Dipeptidyl Peptidase-4 (DPP-4) inhibitors are considered to be weight neutral, whereas sodium glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor analogues (GLP1RA) are associated with weight loss on average (3).

 

INSULIN AND SULFONYLUREAS

 

Insulin causes weight gain by multiple mechanisms (4). Sulfonylureas cause weight gain by increasing endogenous insulin levels. Appetite stimulation, sometimes triggered by hypoglycemia and fluctuating glycemia, is probably the most important factor in body fat increase. Defensive snacking in order to prevent hypoglycemia or compensate for it, can be observed in some patients. Poor glycemic control increases metabolic rate and consequently, improving glycemic control decreases metabolism. Improving metabolic control also reduces glycosuria and retention of otherwise lost calories. Finally, the anabolic effects of insulin can increase protein synthesis and inhibit lipolysis and proteolysis, resulting in a gain of lean body mass (3,4).

 

The weight promoting properties of insulin are dose dependent and are more pronounced in injection regiments that include rapid-acting insulin compared to basal insulin only (5). The addition of metformin to insulin therapy, reduces the effects of insulin on body weight by decreasing energy intake (6).

 

The weight gain by sulfonylurea is most pronounced in the first months of therapy and then reaches a plateau. In the UKPDS and ADOPT studies a mean weight gain of this therapeutic class is approximately 4 kg during the first year of treatment (7,8).

 

THIAZOLIDINEDIONES

 

Thiazolidinediones also cause a substantial time and dose-dependent weight gain ranging from 1.5 to 4 kg in the first year of treatment (1,9). The mechanisms by which TZD’s cause weight gain include fluid retention, promotion of lipid storage, and adipogenesis through activation of peroxisome proliferator-activator receptor gamma (PPARg) (10,11). The fat accumulation is almost uniquely subcutaneous, with stable or even decreasing amounts of visceral fat (12). Thiazolidinediones also improve hepatic steatosis and inflammation in patients with non-alcoholic steatohepatitis (NASH), although safety concerns including osteoporosis and fluid retention with pioglitazone hamper their use (11,13). Newer PPAR drugs for the treatment of NASH are currently being studied in phase 3 trials (14).

 

Antihypertensive Drugs

 

Hypertension frequently accompanies obesity and T2DM. Therefore, drugs that promote weight gain and other metabolic side effects are of obvious concern in patients with obesity and hypertension (15).

 

BETA-BLOCKERS      

 

The propensity of β-blockers to cause weight gain has been known for years (16). Their use is associated with a mean weight gain of 1.2 kg compared to controls, although among β-blockers, variable effects on weight, ranging from no significant change to an increase of 4 kg or more after one year of treatment, have been described. Most weight gain occurs in the first few months, after which no further weight gain is apparent (16).

 

Mechanisms whereby β-receptor antagonists are thought to affect body weight include reductions in total energy expenditure through lowering of basal metabolic rate and thermogenic response to meals, and by inhibition of lipolysis in response to adrenergic stimulation (17). In addition, β-receptor antagonists can promote fatigue and reductions in patient activity (18-20). Polymorphisms in human genes involved in catecholamine signal transduction affecting fat cell lipolysis might partly explain individual susceptibility to β-receptor antagonist-induced weight gain (21). β-blockers may also selectively promote the accumulation of abdominal fat, which is more sensitive to catecholamines than peripheral fat (22). This preponderance of abdominal fat accumulation may be, in part, responsible for the abnormalities related to carbohydrate and lipid metabolism associated with β-adrenergic blockade (23).

 

Several large trials have linked β-receptor antagonists to dysglycemia and new onset diabetes, even without significant weight gain (24). In particular, non-vasodilating beta-blockers (atenolol, metoprolol and propranolol) are associated with a worsening of glycemic and lipid parameters. In contrast, vasodilating beta-blockers (nebivolol, labetolol and carvedilol) have more favorable effects on glucose and lipid profiles (25). Nebivolol has been shown to induce lipolysis, reduce adipocyte lipid droplet size, and promote thermogenic and mitochondrial genes through a β3 adrenergic receptor affect (26). Therefore, selective agents with a vasodilating component such as nebivolol and carvedilol should be prioritized when β-blockers are needed in a population with high risk for metabolic side effects (15).

 

CALCIUM CHANNEL BLOCKERS

 

Calcium channel blockers are considered weight neutral and do not show adverse effects on glucose and/or lipid metabolism. However, flunarizine, a calcium channel blocker used in the prophylaxis of migraine, is associated with increased appetite and weight gain up to 4 kg. These properties have been linked to its blocking effects on both the calcium channel receptor and the dopamine receptor (27,28).

 

Psychotropic Medications

 

Obesity is two to three times more common among patients with psychiatric disorders than the general population, and individuals who are obese suffer more frequently from psychiatric illnesses than those who are normal weight. Underlying causes of this interaction between obesity and psychiatric disease likely include a clustering of adverse metabolic risk behaviors, such as unhealthy eating and insufficient physical activity, as well as substance abuse that accompany many psychiatric conditions (29). But the pathophysiological neural processes that lead to psychiatric diseases also seem to share common brain pathways with those that lead to unwanted weight gain, obesity, metabolic syndrome, and cardiovascular disease risk factors, each of which can influence the risk for the others (30). Mounting evidence points to a critical role for two major pathways: inflammatory processes including related alterations of brain functions and chronic stimulation of the hypothalamic-pituitary-adrenal (HPA) axis (30,31).

 

Psychiatric disorders are often characterized by a chronic stimulation of the HPA axis and sustained cortisol elevation, which have been linked with abdominal obesity, hepatic steatosis, insulin resistance, and cardiovascular disease (31). Chronic psychosocial stress has also been linked with inflammation and metabolic alterations, including weight gain with a predominance of visceral fat accumulation and insulin resistance (30). On the other hand, increased adiposity leads to chronic low-grade activation of inflammatory processes, which have been shown to have a potent role in the pathophysiological brain alterations associated with psychiatric disease (31). It is therefore possible that adiposity-driven inflammation contributes to the development of mood disorders and their growing prevalence worldwide.

 

Medical therapies for depression, mood disorders, and other psychiatric illnesses have been associated with sometimes very large weight gain (Table 1). Epidemiologic data show a positive correlation between weight gain and the time exposed to psychotropic medication or the number of different psychotropic drugs used (32). However, the variation in mean weight gain is large between the different drug classes and even within the same class. For most psychiatric treatments, no correlation is found with weight gain and original diagnosis or severity of the underlying psychiatric condition, treatment outcome, weight at the onset of the disease or treatment, age, or sex, which impedes prediction of those patients who will or will not have metabolic side effects (32). What has been consistently shown is that weight gain in the first month after the start of treatment is a strong predictor of long-term weight gain (33). Therefore, weight should be monitored before and shortly after starting a psychotropic drug therapy and a 5% increase above baseline weight after the first month should prompt physicians to reconsider therapeutic options or to initiate weight-controlling strategies (33,34).

 

Table 1. Overview of the Psychotropic Drugs and Their Mean Effect on Weight.  (See text for abbreviation definitions).

Drug class

 

Weight loss

Weight neutral

(< 1 kg/y)

Minor weight gain

(1-5 kg/y)

Major weight gain

(> 5 kg/y)

Antidepressants

 

Tricyclic agents

 

 

amitriptylline

nortriptylline

imipramine

desipramine

dosulepine

doxepine

clomipramine

 

SSRI

 

escitalopram

paroxetine

citalopram

fluoxetine

sertraline

 

 

SNRI

 

duloxetin

venlafaxin

 

 

MAO-inhibitors

 

moclobemide

phenelzine

 

Other

bupropion

reboxetine

trazodone

nefazodone

mirtazapine

maprotiline

mianserine

 

Antipsychotics

Typical

molindone

 

haloperidol

perphenazine

 

 

Atypical

 

aripiprazol

ziprasidone

lurasidone

paliperidone

iloperidone

asenapine

amisulpiride

quietiapine

risperidone

sertindole

 

clozapine

olanzapine

Anticonvulsants

 

topiramate

zonisamide

lamotrigine

levtiracetam

Tiagabine

oxcarbazepine

gabapentine

pregabalin

valproate

carbamazepine

Mood stabilizers

 

 

 

 

lithium

 

ANTIDEPRESSANT MEDICATIONS

 

The magnitude of weight gain during antidepressant therapy differs significantly by class.

 

Tricyclic Agents

 

The greatest potential to induce weight gain has been shown with the tricyclic agents’ amitriptyline and nortriptyline. Antidepressant–induced weight gain has been clearly established in the acute and maintenance period of depression therapy and is not related to disease severity. Medications from this drug class are also associated with weight gain when used for other indications, such as neuropathic pain or anxiety. To date, no predisposing factors to weight gain resulting from these drugs has been clearly identified (35).

 

Serotonin Agents

 

During initial treatment, several selective serotonin reuptake inhibitors (SSRI’s) (citalopram, fluoxetine, sertraline) and serotonin and norepinephrine reuptake inhibitors (SNRI’s) (venlafaxine and duloxetine) are associated with a slight weight loss. However, with chronic therapy many have shown weight gain. Paroxetine is considered to be the SSRI with the greatest long-term weight gain, possibly due to its affinity for the cholinergic receptor (35,36).

 

Bupropion

 

Bupropion, a norepinephrine and dopamine reuptake inhibitor and nicotinic antagonist, reduces appetite and food cravings (37). In combination with naltrexone, bupropion is approved as an antiobesity drug in United States of America (USA) and the European Union (EU) (38).

 

LITHIUM

 

In randomized controlled trials, the incidence of significant weight gain (more than 5% of initial body weight) has been described to be as high as 60% of the patients on lithium therapy for bipolar disorder. Risk factors for weight gain are a high baseline weight, younger age, co-administration of antidepressants, and female sex (39).

 

The exact mechanism by which lithium exerts these adverse effects on weight is still unknown. Possible mechanisms include a direct effect on hypothalamic centers controlling appetite, increased thirst and increased intake of high caloric drinks, changes in food preference, and its influence on thyroid function with increased incidence of hypothyroidism (39,40).

 

ANTIPSYCHOTICS

 

The number of individuals in the population receiving antipsychotic drugs is surprisingly high, most commonly for psychosis, although antipsychotic drugs are also widely used to treat other psychiatric conditions like bipolar disorders, attention deficit disorder, and dementia in the elderly (41,42).

 

Typical and Atypical Antipsychotics

 

Several major chemical classes of antipsychotic drugs have been developed, mainly the phenothiazines (e.g., chlorpromazine), the butyrophenones (e.g., haloperidol), and the thioxanthines (e.g., flupenthixol). All these “conventional,” or typical, neuroleptics are effective because they are dopamine D2 receptor antagonists, but they all have major neurological side effects (43). Therefore, newer drugs, the atypical antipsychotics or second-generation antipsychotics (SGAP), are increasingly replacing the conventional neuroleptics. These atypical antipsychotics are characterized by a combined activity on both the D2 and 5-HT2a receptors. Besides their antagonistic effects on these receptors, they possess diverse pharmacologic interactions with a number of neurotransmitter receptors (44).

 

Up to 80% of patients taking antipsychotic medication experience weight gain that exceeds their ideal body weight by 20% or more (45). Weight gain variability is high in between these drugs, which has been ascribed to both a high affinity for the H1-histaminic receptor as well as, to a lesser extent, the α1-adrenergic and 5-HT2c-receptors (44). The largest weight gain is consistently associated with olanzapine and clozapine. Weight gain promoting effects of the antipsychotics seem to be more pronounced in people with a normal body weight at baseline and more in women than in men. Weight gain associated with long-term treatment is time- and dose-dependent and can be predicted by weight increases in the first weeks of treatment. Drug-naïve patients gain significantly more weight than patients exposed to antipsychotics in the past and studies in pediatric patients demonstrate greater absolute weight gain in this group than in adults. Patients who have greater treatment-emergent weight gain are more likely to benefit from treatment with antipsychotics (33,46-48).

 

Although SGAP’s influence food intake by altering neurotransmitter function in the hypothalamus, thus leading to excess caloric consumption, obesity, and insulin resistance (49), weight gain is not the only concern for patients taking antipsychotic medication (Figure 2). SGAP’s can also promote lipogenesis and enhance antilipolytic effects of insulin, thereby favoring lipid accumulation and adipocyte enlargement and inducing insulin resistance (50).  Metabolic sequela includes glucose dysregulation and an increased risk for developing metabolic syndrome and type 2 diabetes (51,52). Although an increased prevalence of metabolic syndrome has been reported in drug naïve patients with diverse psychoses, there is a significant association with longer disease duration and with the intake of clozapine in particular (51,53). In one 5-year study of clozapine-treated patients, 52% experienced one or more episodes of hyperglycemia and 30% were diagnosed as having type 2 diabetes (54). Newer SGAP appear to have fewer metabolic side effects (1,2,55,56).

 

The development of diabetes in patients taking antipsychotic has also been reported in patients without significant weight changes. SGAP’s are associated with a marked increase in insulin resistance in muscle, adipose tissue, and liver (49), possibly mediated by impaired GLUT-4 and GLUT-5 glucose transporter function (57). In addition, a direct impairment of pancreatic β-cell function and decreased insulin secretion has been linked to the affinity of these drugs for the 5HT-1a and 5HT-2 serotonin receptors of the β-cells (49).

 

Manifestations of insulin resistance, impaired glucose tolerance, metabolic syndrome (including elevations in triglyceride levels and reductions in HDL cholesterol), and type 2 diabetes contribute to the higher incidence of cardiovascular disease in patients taking these drugs. People with psychosis have a 20% shorter life expectancy than the general population, mainly driven by an increase in cardiovascular disease (53,58). In view of the high cardiometabolic risk associated with antipsychotic drug use, the American Diabetes Association and American Psychiatric Association (ADA/APA) Consensus Development Conference recommends close monitoring of weight and metabolic and cardiovascular risk factors in all patients taking SGAP’s (34).

Figure 1. Schematic representation of the central and peripheral mechanisms of antipsychotic-induced weight gain and metabolic side effects as well as current and future preventive and therapeutic options. HPA axis: hypothalamic–pituitary–adrenal axis, GLP-1: glucagon like peptide-1, GLP1RA: GLP-1 receptor agonist, RMR: resting metabolic rate, IL6: interleukin-6, TNF-α: tumor necrosis factor-α, IR: insulin resistance, PKC-βi: protein kinase C-β inhibitor, CB1R: cannabinoid receptor type 1, periph CB1Ri: peripheral cannabinoid receptor type 1 inhibitor, SREBP1c: sterol regulatory element-binding proteins type 1c, AMPK: AMP-activated protein kinase (Reprinted by permission from Springer Nature Customer Service Centr GmbH: J Endocrinol Invest. 2017;40(11):1165-1174) (2).

Therapy and Prevention of Antipsychotic Weight Gain

 

Many studies have evaluated pharmacological and non-pharmacological approaches to prevent or treat weight gain that accompanies SGAP treatment. Of non-pharmacological interventions, no significant difference was found between individual and group interventions, or cognitive-behavioral versus nutritional counselling. Adherence to the weight management program appears the best prognostic factor for achieved weight loss. To promote therapeutic alliance, weight management programs should be flexible and individualized to the patient’s needs, age and stage of their disease and incorporate daily recreational-based activities. Benefits are thought to be the greatest when delivered as early as possible, before weight gain has occurred (59).

 

Switching to another antipsychotic drug with less potential for weight and cardiometabolic side effects has been endorsed by the ADA/APA consensus guidelines in those with more than 5% weight gain or worsening of their lipid or glycemia parameters, following studies that show benefits of this strategy to limit further weight gain or reduce weight and reverse components of the metabolic syndrome (34,60) (Table 2). For example, use of the SNRI reboxetine may reduce olanzapine-induced weight gain in schizophrenia patients. Weight and metabolic benefits have also been reported by switching to, or the addition of, topiramate, amantadine, fluvoxamine, and orlistat (61-63). With metformin, attenuation or reduction of weight gain and amelioration of the metabolic side effects of SGAP therapy has been demonstrated, with greater benefits the earlier metformin was started (61,64). In a rodent study, the addition of metformin and berberine prevented the loss of brown fat induced by olanzapine and was associated with favorable changes in expression of several genes controlling energy expenditure (65).

 

Table 2. Mean Weight Reducing Effects of Switching to a Less Metabolically Active SGAP or Addition of Weight Reducing Drugs (60,63).

Action

Mean weight reduction (kg)

95 % CI

Switch to:

aripiprazole or quietiapine from olanzapine

-1.94

-3.90 to 0.08

Addition of:

Metformin

-2.94

-4.89 to -0.99

Topiramate

-2.52

-4.87 to -0.16

Reboxetine

-1.90

-3.07 to -0.72

 

In patients who are obese or with diabetes, glucagon-like protein-1 receptor agonists (GLP-1 RA) demonstrate long-lasting weight loss and benefits on glucose metabolism (1,3). Growing evidence suggests that patients who are overweight and those with psychosis exhibit similar structural brain changes, cognitive deficits, and central neuropeptide alterations, suggesting an overlap between the pathophysiological pathways of these disorders (66). GLP-1 RA’s have been shown to provide neuroprotective effects in cerebral degenerative diseases such as Parkinson’s disease, Huntington’s chorea, and Alzheimer’s dementia (66). Liraglutide, a once daily injected GLP-1 RA, reverses SGAP-induced weight gain, impaired glucose tolerance, metabolic side effects and behavioral depression (67-69).

 

Mifepristone, a glucocorticoid and progestin receptor antagonist, attenuated increases in weight and reduced the metabolic changes induced by risperidone and olanzapine, suggesting mechanistic involvement of the hypothalamic-pituitary-adrenal axis in the weight and cardiometabolic side effects of antipsychotic medications (70).  The orally effective selective protein kinase C-β (PKC- β) inhibitor ruboxistaurin, which is used in treatment of diabetes-associated retinopathy and macular edema, attenuates the effects on adipose tissue differentiation by clozapine in rodents. If this is shown to be relevant for humans, it could offer a new target for the prevention of antipsychotic-induced weight gain (71).

 

Because of the associations between inflammation, adiposity and psychiatric disease, other therapeutic options being explored to improve psychiatric symptoms without adverse metabolic sequelae include COX-2 selective non- steroidal anti-inflammatory drugs, and monoclonal antibodies against anti TNF-α and Interleukin-6 (72,73).

 

Anti-Seizure Drugs

 

Many of the anti-epileptic treatments are associated with weight change. Most prominent are valproate and carbamazepine, inducing weight gain in 71% and 43% of the patients, respectively. Pregabalin and gabapentin can also induce weight gain and are of particular importance since they are used more and more in the treatment of neuropathic pain, including in patients with diabetes. Weight neutral anti-epileptic drugs include lamotrigine, levetiracetam and phenytoin. Some others are associated with weight loss, including felbamate, topiramate, and zonisamide (74).

 

Weight-inducing effects of valproate are thought to result from interactions with appetite-regulating neuropeptides and cytokines within the hypothalamus as well as effects on energy expenditure (75). Greater weight gain is associated with longer duration of treatment with valproate, although most weight gain is observed within the first year. Specific categories of patients shown to be more susceptible to weight gain with this medication include women (vs. men), post-pubertal adolescents (vs. younger children), and those who are overweight before treatment begins. On the other hand, weight gain is not related to valproate dosage or serum levels (75).

 

Peripheral actions are thought to mediate valproates adverse effects on glucose and lipid metabolism that contribute to weight-independent worsening of insulin resistance and risk for type 2 diabetes. Directs effects in adipose tissue have been shown to increase leptin resistance, decrease adiponectin levels, as well as increase free fatty acids, all resulting in insulin resistance (76). On the other hand, valproic acid is associated with β-cell dysfunction and impaired insulin secretion by increasing oxidative stress, and direct inhibition of the GLUT-1 transporter, thereby hampering insulin secretion in an insulin resistant state, promoting hyperglycemia and type 2 diabetes (76-78).

 

Besides the increased risk for hyperglycemia and type 2 diabetes, an increased risk of other features of the metabolic syndrome (e.g., dyslipidemia) as well as endothelial dysfunction has been demonstrated. It has additionally been reported that up to 60% of the patients taking valproate also develop non-alcoholic steatohepatitis, further contributing to insulin resistance, chronic inflammation, and increased risk for cardiovascular disease (79). 

 

With the other anticonvulsants, weight gain can be considerable in susceptible patients (74). However, in contrast to valproate, metabolic side effects accompanying use of carbamazepine, pregabalin and gabapentin are thought to be secondary to the induced weight gain rather than weight-independent mechanisms (80). On the other hand, topiramate and zonisamide have been shown to decrease body weight, even when studied in populations with obesity and overweight without any seizure history. Adding topiramate to another antiepileptic, antipsychotic or antidepressant drug, or changing anticonvulsant therapy for topiramate or zonisamide can help prevent or treat weight gain that accompanies psychiatric or anticonvulsant therapy (63,81). In the USA, the combination therapy topiramate/phentermine is approved as an antiobesity drug (82).

 

DRUGS ASSOCIATED WITH WEIGHT GAIN AND LIPODYSTROPHY

 

Corticosteroids

 

Although weight gain is considered a common side-effect of long-term treatment with glucocorticoids, prospective studies examining weight gain are scarce. Self-reported data in patients using chronic corticoid therapy show substantial weight gain in up to 70% of all patients (83). The weight gain associated with glucocorticoid therapy can be massive with over 10 kg increases in approximately 20% of patients in their first treatment year of treatment. The risk of weight gain with glucocorticoids is dose dependent and significantly increases with intakes above the equivalent of oral or parenteral 5 mg prednisone per day. Inhaled corticosteroids and single epidural steroid injections have no effect on body weight.

 

Glucocorticoids may induce an increase in food intake and dietary preference for high-caloric, high-fat ‘comfort foods’ through changes in the activity of AMP-activated protein kinase in the hypothalamus (84-86). Glucocorticoids decrease thermogenesis and uncoupling protein 1 (UCP-1) expression in brown adipose tissue, thereby influencing metabolic rate (87). Chronic glucocorticoid therapy or a state of chronic hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis is associated with activation of the endocannabinoid (eCB) system, which is a potent regulator of food intake and decreases energy expenditure (88). 

 

Glucocorticoids also affect body fat distribution by increasing visceral fat mass, thereby increasing insulin resistance and the risk for impaired glucose tolerance, diabetes and cardiovascular disease (89). Although the mechanisms for this are not completely understood, glucocorticoids acutely stimulate lipolysis through the activation of hormone-sensitive lipase and an increased catecholamine responsiveness. When this disproportionately affects fat stores in the extremities, it can lead to loss of these depots, or lipodystrophy, the risk of which has been reported to be higher among females and younger patients and increases with a higher baseline body mass index (90). On the other hand, during chronic administration or exposure to high endogenous plasma corticoid concentration such as in Cushing’s syndrome, they promote both adipocyte hypertrophy by increasing synthesis and storage of lipids and adipose tissue hyperplasia by increasing differentiation of preadipocytes to mature adipocytes. Visceral adipose tissue has a higher glucocorticoid receptor density as compared with other fat depots, which might favor enhanced expansion of visceral adipose tissue (89). These differential effects on visceral and subcutaneous fat may be mediated by differential regulation of key metabolic genes including lipoprotein lipase, 11-beta-hydroxysteroid-dehydrogenase-1 (11β-HSD-1) and UCP-1 (89).

 

Glucocorticoid induced overexpression of 11β-HSD-1 in adipose tissue leads to an increase of plasma triglycerides and cholesterol levels, while 11β-HSD-1 overexpression in liver promotes insulin resistance, hepatic steatosis, and increased lipid synthesis (91,92).

In the liver, glucocorticoids act through peripheral stimulation of the cannabinoid-1 receptor (CB1R), inducing hepatic lipogenesis, steatosis and dyslipidemia. By enhancing CB1R in adipose tissue, glucocorticoids induce insulin resistance (IR) and obesity. Blocking the peripheral CB1R attenuates all aspects of metabolic dysregulation by glucocorticoids, leading the path to potential therapeutic option by selective peripheral CB1R blockers (88).  These properties make specific 11β-HSD-1 inhibitors or peripheral CB1R blockers promising candidate drugs to reverse or prevent glucocorticoid-induced side effects.

 

Hypolipidemic Drugs

 

Protein convertase subtilisin kexin type 9 (PCSK9) regulates plasma low-density lipoprotein levels and low-density lipoprotein receptor expression in several tissues. Fully human monoclonal PCSK-9 inhibitors (alirocumab and evolocumab) and small interfering RNA molecules designed to target PCSK9 messenger RNA (inclisiran) have demonstrated substantial and sustained reductions in LDL-cholesterol levels, as well as significant reductions in major cardiovascular events (93).

 

Although no obvious side effects on body weight and body fat composition or on glucose tolerance were reported in these trials, patients with genetically low PCSK9 (R46L polymorphism) have a two-fold increased prevalence of hepatic steatosis and greater epicardial fat thickness (94). PCSK9 variants associated with lower LDL cholesterol have also been associated with higher fasting glucose levels, bodyweight, and waist-to-hip ratio, and an increased risk of type 2 diabetes (95). PCSK9 KO mice display higher visceral adipose tissue (but not subcutaneous adipose tissue), compared with wild type mice, suggesting that genetically determined PCSK9 deficiency might be associated with ectopic fat accumulation (94). It has been shown that besides the effects of LDL-R regulation in liver tissue, PCSK9 regulates VLDL-R, ApoE2 R and the CD36 receptor. PCSK9 deficiency results in reduced post-prandial lipemia and triglyceride rich lipoprotein production, while its overexpression promotes hepatic lipogenesis (96). Therefore, in genetic studies in animals and humans, PCSK9 is plays a pivotal role in fat metabolism: it regulates circulating cholesterol levels via hepatic LDL-R, but it also influences visceral adipogenesis via adipose VLDL-R regulation (97).

 

Therapies that reduce the rate of VLDL secretion represent an attractive alternative for reducing plasma concentrations of pro-atherogenic lipoproteins. Mipomersen is a second-generation anti-sense oligonucleotide that inhibits the translation of the apolipoprotein B by binding to the mRNA sequence of apolipoprotein B. Mipomersen reduces apolipoprotein B synthesis in the liver and the production of VLDL (and hence to IDL and LDL as well) and increases catabolism of VLDL. On the other hand, the hepatic production rate of triglycerides is unaffected (98). In clinical trials, mipomersen is associated with an approximately four times higher risk for hepatic steatosis. In contrast to NAFLD associated with common obesity, however, mipomersen-induced liver steatosis is not associated with an increase in inflammation or fibrosis (99).

 

Microsomal triglyceride transfer protein (MTP) is the key protein that delivers the lipid droplet to nascent VLDL and chylomicron particles during the assembly and secretion of lipoproteins in liver and intestine. MTP inhibition (e.g. lomitapide) decreases the secretion of chylomicrons and VLDL, thereby reducing the production of triglyceride rich lipoproteins and ultimately the production of LDL. Treatment with this drug also induces intra-hepatic fat accumulation, but in contrast to mipomersen, MTP inhibition is associated with greater (up to six-fold) increases in hepatic fat content and more severe increases in transaminase levels (100).

 

Acyl-CoA: cholesterol O-acyl transferase 2 (ACAT2) plays an important role in maintaining cellular cholesterol homeostasis.  When absorbed cholesterol enters the body, it is esterified by ACAT2 and directed to the liver, where it is stored within hepatocytes in lipid droplets as cholesteryl esters. Recently it was shown in high-fat fed mice that mechanisms linking accumulation of hepatic cholesteryl esters with hepatic triglyceride accumulation depend not just on de-novo triglyceride synthesis and lipogenesis secondary to hyperinsulinemia and hyperglycemia, but also on the presence of cholesteryl ester in hepatocytes limiting mobilization of triglycerides from the liver (101). Studies have shown that ACAT inhibitors are effective for the treatment of hypercholesterolemia and atherosclerosis in rodents, however data in humans have been disappointing (102). The recent data coupling lower cholesteryl esters content to higher TG mobilization, as is realized by selective ACAT2 inhibitors, needs further study not only for treating hypercholesterolemia and atherosclerosis but also for the therapy of NAFLD.

 

Antiretroviral Therapy

 

Shortly after the introduction of effective highly-active antiretroviral therapy (HAART) for the treatment of human immunodeficiency virus (HIV) disease, it became clear that patients on these medications often had disorders of fat storage and/or wasting (lipodystrophy). The term “HIV-associated lipodystrophy syndrome” was introduced to describe a typical loss of subcutaneous fat in the limbs and face (lipoatrophy) and central or truncal fat accumulation (lipohypertrophy) (Table 3). Lipohypertrophy is characterized by intra-abdominal visceral fat accumulation and localized fat accumulation in breasts, the dorsocervical region and under the skin as lipomas. While in some patients mixed patterns are observed, some patients exhibit pure lipoatrophy and others have only fat accumulation (103). The prevalence of lipodystrophy in HIV infected patients ranges from 10 to 80%. This wide range is due to differences in study population, race, age and duration of HIV infection or antiretroviral therapy, but also due to different definitions of lipodystrophy and methods of diagnosis (104).

 

 

Table 3: Classification of Antiretroviral Drugs and the Risk for Metabolic Consequences

Drug

Lipohypertrophy / weight gain

 

Lipoatrophy

Insulin resistance

Dyslipidemia

Nucleoside and nucleotide reverse transcriptase inhibitors (NRTI’s)

Abacavir (ABC)

0

0

0

+

Didanosine (ddI)

+/-

+/-

+

+

Emtricitabine (FTC)

0

0

0

0

Lamivudine (3TC)

0

0

0

+

Stavudine (d4T)

++

+++

++

++

Tenofovir (TDF)

0

0

0

0

Zidovudine (AZT or ZDV)

+

++

++

+

Non- nucleoside reverse transcriptase inhibitors (NNRTI’s)

Delavirdine(DLV)

+/-

+/-

0

+

Efavirenz (EFV)

+/-

+

+

++

Etrivirine (ETR)

+/-

0

0

0

Nevirapine (NVP)

0

0

0

++

Rilpivirine (RPV)

+

0

0

0

Protease inhibitors (PI’s)

Amprenavir (APV)

+

+

+

+

Atazanavir (ATV)

++

0

0

+

Darunavir (DRV)

+

0

+/-

+/-

Indinavir (IDV)

+

+/-

+++

+

Lopinavir (LPV)

+

+/-

+++

++

Nelfinavir (NFV)

+

+/-

+

++

Ritonavir (RTV)

+

+/-

+++

+++

Saquinavir (SQV)

+

+/-

+/-

+/-

Tipranavir (TPV)

+

+/-

+++

++

Fosamprenavir (FPV)

++

0

0

+

Fusion inhibitor

Enfuvirtide (T20)

0

0

0

0

Integrase inhibitor

Raltegravir (RAL)

+

0

0

0

Dolutegravir (DTG)

+

0

 

 

CCR5 antagonist = entry inhibitors

Maraviroc (MVC)

No or positive effects (animal data)

0

0

0

Post attachment inhibitors

Ibalizumab

No data

No data

0

0

(+ = increase; 0 = neutral: +/- = discrepant)

 

Risk factors for lipoatrophy are male gender, older age, lower weight before therapy, lower CD4 cell counts, a higher baseline viral load, and co-infection with hepatitis C. Certain mitochondrial haplotypes and nuclear genetic polymorphisms are associated with an increased risk for lipoatrophy (104,105).  In addition, there is a clear association of lipoatrophy with stavudine and zidovudine use, while switching to other retroviral drugs or using an NRTI-sparing regimen reverses lipoatrophy (106).

 

While the distribution of lipoatrophy is specific for HIV infected patients and anti-HIV therapy (Table 3), abdominal fat accumulation seems not to be associated with specific antiretroviral drugs and carries the same risk for metabolic syndrome as non-medication-associated visceral fat accumulation. Risk factors associated with fat accumulation during HAART are increasing age, female sex, weight before start of antiretroviral therapy, dietary factors, and longer duration of HIV treatment (104). In the AIDS Clinical Trial Group study (A5175), the prevalence of overweight or obesity increased from 25% to 40% after 144 weeks of HAART. In another trial a 30% increase in visceral fat mass was seen after 96 weeks of therapy (107). Whether lipohypertrophy can be directly attributed to HAART or represents the effects of treating HIV itself, is still a matter of debate. Indeed, central fat gain generally occurs at similar rates in patients randomized to different HAART regimens, is not associated with any specific antiretroviral drug or drug class, and does not reverse on switching antiretrovirals (106,108). In addition to the promotion of generalized weight gain, increases of as little as 5% of visceral adipose tissue are associated with increased metabolic risk, cardiovascular side effects, and even 5-year mortality. Stated differently, the cardiometabolic risk of weight gain in HIV patients is much higher than comparable weight gain in non-HIV infected controls (109).

 

Therefore, the European AIDS Clinical Society recommends monitoring for changes in body composition of HIV patients by using body mass index, waist circumference, waist-to-hip ratio, and to screen regularly for clinical lipodystrophy in all patients at HIV diagnosis, before starting HAART, and annually thereafter. Fat atrophy should be distinguished from general wasting associated with advanced AIDS, where besides wasting of fat mass, lean body mass is also lost. To distinguish visceral fat accumulation from simple obesity, skin fold measures can help since in HIV-induced abdominal fat accumulation, subcutaneous fat is normal or decreased, while it mostly increases in patients with simple obesity (110).

Figure 2. Major pathogenetic pathways in HIV-induced lipodystrophy and its metabolic consequences (adapted from Debarle MC et al (111))

PATHOGENETIC MECHANISMS OF HIV INDUCED LIPODYSTROPHY

 

Lipodystrophy is considered to be multifactorial, resulting from the complex interaction of host factors, HIV-related factors, and antiretroviral drug specific factors (Figure 3). While lipodystrophy is clearly linked to antiretroviral therapy, disturbances in adipose tissue gene expression are present in treatment-naïve patients with HIV, indicating that HIV-1 infection itself likely creates alterations in adipose tissue that are worsened by antiretroviral therapy (108,112).

 

When used in monotherapy, lipoatrophy is not noticed in patients using protease-inhibitors (PI). However, the co-administration of PI’s with nucleoside and nucleotide reverse transcriptase inhibitors (NRTI’s), such as stavudine and zidovudine, play an additive role in the NRTI-induced lipodystrophy (106). The older nucleoside analogues inhibit mitochondrial DNA polymerase-γ within adipocytes causing mtDNA depletion and mitochondrial dysfunction and oxidative stress in subcutaneous adipose tissue (SAT). Together with a genetic predisposition and mitochondrial dysfunction secondary to HIV itself, NRTI’s contribute in mitochondrial toxicity and increased oxidative stress, inhibiting adipogenesis and adipocyte differentiation, and promoting apoptosis, lipolysis, and dyslipidemia (105,113). Lipoatrophy is associated with inflammation, as shown by an increased macrophage number and expression of TNF-α, IL-6 and IL-8 together with increased fibrosis (114).

 

Adipose tissue serves as a reservoir for HIV virus, altering adipose tissue environment and causing adipose tissue inflammation. The HIV accessory viral protein R (Vpr) inhibits PPAR-γ, impairs the expression of genes related to adipocyte metabolism including adiponectin and activates glucocorticoid target gene expression, inducing macrophage infiltration and adipose tissue hypertrophy (108). Therefore, it appears that HIV-1 infection initiates a first wave of alterations in adipose tissue that is amplified by HAART and ultimately results in lipoatrophy.

PI’s are more closely associated with lipo-accumulation (115). They interfere with adipocyte maturation and differentiation by alterations in gene expression of several transcriptase factors (SREBP-1, PPAR-γ, C/EBPα and β genes) and genes encoding for acyl coenzyme-A synthase, lipoprotein lipase, GLUT-4, leptin and adiponectin, resulting in impaired fatty acid and glucose uptake, increased lipolysis and peripheral fat loss, increased triglyceride esterification and central fat accumulation. The imbalance in the production of adipokines (adiponectin and leptin) and infiltration of immune cells into adipose tissue exacerbate the pro-inflammatory environment (114).

 

PPAR-γ expression is also reduced by NRTI’s. The non-nucleoside reverse transcriptase inhibitor (NNRTI) efavirenz decreases expression of SREBP-1c, thus decreasing intracellular stores of triglycerides, and exerts anti-adipogenic effects in cultured adipose cells (116).

Mitochondrial DNA depletion is common to both subcutaneous and visceral or dorsocervical depots in HIV lipodystrophy and mitochondrial dysfunction in visceral adipose tissue (VAT) was found to be similar to that in SAT. In SAT, mitochondrial dysfunction is linked with lipoatrophy, whereas in VAT lipohypertrophy results. These observations indicate that different responses occurring in subcutaneous and visceral fat depots during HAART treatment are likely related to intrinsic differences in physiology between these depots (114). 

 

Increases in proinflammatory cytokines, such as TNF-α and interleukin-6 further contribute to the development of lipodystrophy and their metabolic consequences. TNF-α inhibits adipocyte differentiation, increases apoptosis and mitochondrial toxicity and activates 11β-HSD-1 resulting in increased lipid accumulation in adipocytes, lipolysis and insulin resistance. In addition, increased fat tissue fibrosis and lipo-hypertrophy, are associated with ectopic lipid accumulation in liver, muscle and heart further increasing cardiometabolic complications (111).

 

Older PI‘s (indinavir, lopinavir and ritonavir) are associated with abnormalities in glucose tolerance and their use is associated with a threefold increase in the risk of diabetes compared to other treatment options. PI‘s inhibit the uptake of glucose into cells by interfering with the GLUT-4 glucose transporter and decrease insulin secretion through effects on β-cell function. Following chronic treatment, this insulin resistance leads to an inadequate suppression of lipolysis and endogenous glucose production as well as a decreased peripheral glucose uptake.  Newer PI’s like darinavir and atazanavir and Integrase inhibitors (INSTI’s) have only limited effects on glucose metabolism. Of the nucleoside analogues, stavudine and zidovudine have been associated with the greatest increase in insulin resistance (114,117).

 

Finally, the hypothalamic-pituitary-growth hormone axis may also be involved in the metabolic changes associated with lipodystrophy. Mean growth hormone levels and growth hormone pulse amplitude are reduced in HIV-infected men with body-fat changes receiving HAART, compared with men without body-fat changes and healthy control subjects (118).

 

Abnormalities of lipid metabolism in HIV-infected patients were described before the advent of HAART and are mainly characterized by increases in triglycerides by a decreased triglyceride clearance and increased hepatic VLDL synthesis and apolipoprotein-E levels. In advanced AIDS disease, reduced HDL-cholesterol and a predominance of small, dense LDL particles have been reported (119). The chronic inflammation of the HIV infection itself and the associated increase in circulating cytokines can induce dyslipidemia. With the use of HAART, these lipid abnormalities tend to increase in severity and in their prevalence, with sometimes dramatic increases in lipid concentrations, particularly triglycerides (119,120). The prevalence and severity of lipid abnormalities varies widely depending on the type of HAART, nutritional status, and HIV disease stage. Risk factors seem to be a higher viral load, a family history of lipid abnormalities, less physical activity, increasing weight, greater BMI and greater trunk-to-limb fat ratio (104,119). Lipid changes and therapy of dyslipidemia in patients on retroviral therapy are described elsewhere in Endotext (121).

 

THERAPY OF HIV ASSOCIATED LIPODYSTROPHY

 

In patients with visceral fat accumulation, combined aerobic and strength training is generally recognized to reduce visceral fat and biomarkers for inflammation (108).  Switching from a thymidine analogue NRTI (e.g. stavudine or zidovudine) to an alternative agent is considered to be a reasonable strategy to reverse or slow progression of lipoatrophy. Switching from stavudine to other NRTIs has been shown to improve mitochondrial indices, reduce fat apoptosis, and decrease some adipose tissue markers of inflammation (114,122). Switching to newer antiretroviral drugs has no effect on VAT accumulation. A switch from NRTI and NNRTI to protease inhibitors showed no weight changes whereas a switch to newer integrase inhibitors may cause even greater weight gain (123).

 

As mentioned above, pituitary growth hormone (GH) secretion is altered in HIV patients, and about one-third of patients meet criteria for GH deficiency. Growth hormone therapy in patients with ART induced lipodystrophy, reduces visceral adiposity, but is associated with supraphysiologic levels of IGF-I and symptoms of growth hormone excess. The FDA approved tesamorelin, a recombinant human GH releasing hormone, for the treatment of excess abdominal fat in HIV-infected patients. Tesamorelin decreases VAT by 15 to 18 %, with a significant improvement of triglyceride and cholesterol levels. The effects of tesamorelin appear to be highly specific for the visceral-fat compartment, with relatively little effect on subcutaneous fat. The preferential reduction in VAT is important, given the peripheral lipoatrophy. The reduction in VAT is associated with greater baseline visceral fat mass, suggesting that larger effects might be seen among patients with more accumulation of visceral fat. Tesamorelin also improves lipid profiles, triglyceride levels and the ratio of total to HDL cholesterol. Despite this reduction in VAT, a small but statistically significant increase in HbA1c is seen in subjects receiving tesamorelin. Therefore, monitoring of IGF-1 and glycemic parameters is warranted (124).

 

In patients with glucose intolerance and central obesity, metformin treatment was associated with small reductions of VAT. However, in patients with lipoatrophy, metformin should be used with caution, since a further decrease of subcutaneous fat can be induced. Doleglutavir (an integrase inhibitor) increases metformin concentration. The total daily dose of metformin should therefore not exceed 1000 mg in patients co-administrated metformin with doleglutavir (104).

 

While the therapeutic goals and the management of diabetes in patients with HIV with or without HAART is similar to the guidelines in the general population, evidence suggests that insulin sensitizers may be preferable to insulin secretagogues. A meta-analysis of all studies with thiazolidinedione therapy (pioglitazone) showed a significantly higher limb fat mass gain in patients treated with pioglitazone. However, some studies, demonstrated a lack of effect in patients on thymidine–NRTI therapy, explained by the decreased expression of PPARγ in those patients. Any benefit appears to be small, therefore pioglitazone should be reserved to patients with severe insulin resistance and /or diabetes (125,126).

 

For patients with morbid obesity and/or major obesity-related diseases, bariatric surgery can be considered. An average of 20% reduction of initial BMI, improved body composition and metabolic status was observed in patients after bariatric surgery, similarly to obese non-HIV patients. However, ART treatment should be monitored to control HIV infection and some ART doses should be adjusted following this degree of weight loss (127).

 

Lipodystrophy associated changes in adipokine concentration could be the basis of future therapeutic options. Leptin and adiponectin decreases have been demonstrated in patients with lipodystrophy. Recombinant leptin therapy increases adiponectin levels and improve insulin sensitivity, glucose tolerance and dyslipidemia and decreases VAT without any change in SAT (128). Finally, therapy with locally injected fillers or autologous fat transplantation, are cosmetic therapies with positive results on patient wellbeing and therapy compliance (129).

 

CONCLUSION

 

Many frequently used medications can cause weight gain, preferential central (visceral) fat accumulation, ectopic fat accumulation in liver and muscle, and consequently have adverse glucolipid metabolic side effects that increase patient’s risk for type 2 diabetes and cardiovascular disease. In high-risk patients, use of alternative or less metabolically-active drugs can reverse or prevent unwanted weight gain and metabolic disturbances. In those patients who need to remain on medications that induce obesity and metabolic dysfunction, frequent monitoring and management of resulting weight gain, elevated blood pressure, dyslipidemia, and type 2 diabetes using standard therapies is warranted.

 

REFERENCES

 

  1. Domecq JP, Prutsky G, Leppin A, Sonbol MB, Altayar O, Undavalli C, Wang Z, Elraiyah T, Brito JP, Mauck KF, Lababidi MH, Prokop LJ, Asi N, Wei J, Fidahussein S, Montori VM, Murad MH. Clinical review: Drugs commonly associated with weight change: a systematic review and meta-analysis. J Clin Endocrinol Metab. 2015;100(2):363-370.
  2. Verhaegen AA, Van Gaal LF. Drug-induced obesity and its metabolic consequences: a review with a focus on mechanisms and possible therapeutic options. J Endocrinol Invest. 2017;40(11):1165-1174.
  3. Van Gaal L, Scheen A. Weight management in type 2 diabetes: current and emerging approaches to treatment. Diabetes Care. 2015;38(6):1161-1172.
  4. Russell-Jones D, Khan R. Insulin-associated weight gain in diabetes--causes, effects and coping strategies. Diabetes, obesity & metabolism. 2007;9(6):799-812.
  5. Pontiroli AE, Miele L, Morabito A. Increase of body weight during the first year of intensive insulin treatment in type 2 diabetes: systematic review and meta-analysis. Diabetes, obesity & metabolism. 2011;13(11):1008-1019.
  6. Makimattila S, Nikkila K, Yki-Jarvinen H. Causes of weight gain during insulin therapy with and without metformin in patients with Type II diabetes mellitus. Diabetologia. 1999;42(4):406-412.
  7. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352(9131):854-865.
  8. Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, Kravitz BG, Lachin JM, O'Neill MC, Zinman B, Viberti G, Group AS. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med. 2006;355(23):2427-2443.
  9. Medici V, McClave SA, Miller KR. Common Medications Which Lead to Unintended Alterations in Weight Gain or Organ Lipotoxicity. Current gastroenterology reports. 2016;18(1):2.
  10. Cignarelli A, Giorgino F, Vettor R. Pharmacologic agents for type 2 diabetes therapy and regulation of adipogenesis. Archives of physiology and biochemistry. 2013;119(4):139-150.
  11. Aleman-Gonzalez-Duhart D, Tamay-Cach F, Alvarez-Almazan S, Mendieta-Wejebe JE. Current Advances in the Biochemical and Physiological Aspects of the Treatment of Type 2 Diabetes Mellitus with Thiazolidinediones. PPAR research. 2016;2016:7614270.
  12. Wilding J. Thiazolidinediones, insulin resistance and obesity: Finding a balance. Int J Clin Pract. 2006;60(10):1272-1280.
  13. Mills EP, Brown KPD, Smith JD, Vang PW, Trotta K. Treating nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a review of efficacy and safety. Therapeutic advances in endocrinology and metabolism. 2018;9(1):15-28.
  14. Banini BA, Sanyal AJ. Current and future pharmacologic treatment of nonalcoholic steatohepatitis. Curr Opin Gastroenterol. 2017;33(3):134-141.
  15. Landsberg L, Aronne LJ, Beilin LJ, Burke V, Igel LI, Lloyd-Jones D, Sowers J. Obesity-related hypertension: pathogenesis, cardiovascular risk, and treatment--a position paper of the The Obesity Society and The American Society of Hypertension. Obesity (Silver Spring, Md). 2013;21(1):8-24.
  16. Sharma AM, Pischon T, Hardt S, Kunz I, Luft FC. Hypothesis: Beta-adrenergic receptor blockers and weight gain: A systematic analysis. Hypertension (Dallas, Tex : 1979). 2001;37(2):250-254.
  17. Newsom SA, Richards JC, Johnson TK, Kuzma JN, Lonac MC, Paxton RJ, Rynn GM, Voyles WF, Bell C. Short-term sympathoadrenal inhibition augments the thermogenic response to beta-adrenergic receptor stimulation. The Journal of endocrinology. 2010;206(3):307-315.
  18. Welle S, Schwartz RG, Statt M. Reduced metabolic rate during beta-adrenergic blockade in humans. Metabolism. 1991;40(6):619-622.
  19. Astrup A, Simonsen L, Bulow J, Madsen J, Christensen NJ. Epinephrine mediates facultative carbohydrate-induced thermogenesis in human skeletal muscle. Am J Physiol. 1989;257(3 Pt 1):E340-345.
  20. Koch G, Franz IW, Lohmann FW. Effects of short-term and long-term treatment with cardio-selective and non-selective beta-receptor blockade on carbohydrate and lipid metabolism and on plasma catecholamines at rest and during exercise. Clinical science (London, England : 1979). 1981;61 Suppl 7:433s-435s.
  21. Arner P. Genetic variance and lipolysis regulation: implications for obesity. Annals of medicine. 2001;33(8):542-546.
  22. Hoffstedt J, Arner P, Hellers G, Lonnqvist F. Variation in adrenergic regulation of lipolysis between omental and subcutaneous adipocytes from obese and non-obese men. Journal of lipid research. 1997;38(4):795-804.
  23. Nicklas BJ, Rogus EM, Colman EG, Goldberg AP. Visceral adiposity, increased adipocyte lipolysis, and metabolic dysfunction in obese postmenopausal women. The American journal of physiology. 1996;270(1 Pt 1):E72-78.
  24. Elliott WJ, Meyer PM. Incident diabetes in clinical trials of antihypertensive drugs: a network meta-analysis. Lancet. 2007;369(9557):201-207.
  25. Fonseca VA. Effects of beta-blockers on glucose and lipid metabolism. Curr Med Res Opin. 2010;26(3):615-629.
  26. Bordicchia M, Pocognoli A, D'Anzeo M, Siquini W, Minardi D, Muzzonigro G, Dessi-Fulgheri P, Sarzani R. Nebivolol induces, via beta3 adrenergic receptor, lipolysis, uncoupling protein 1, and reduction of lipid droplet size in human adipocytes. Journal of hypertension. 2014;32(2):389-396.
  27. Vecsei L, Majlath Z, Szok D, Csati A, Tajti J. Drug safety and tolerability in prophylactic migraine treatment. Expert opinion on drug safety. 2015;14(5):667-681.
  28. Manconi FM ZA, Piccardi MP, et al. . Behavioural and biochemical effects of flunarizine on the dopaminergic system in rodents. . Cephalalgia. 1987;7:420-421.
  29. Compton MT, Daumit GL, Druss BG. Cigarette smoking and overweight/obesity among individuals with serious mental illnesses: a preventive perspective. Harvard review of psychiatry. 2006;14(4):212-222.
  30. Milaneschi Y, Simmons WK, van Rossum EFC, Penninx BW. Depression and obesity: evidence of shared biological mechanisms. Mol Psychiatry. 2018.
  31. Menard C, Pfau ML, Hodes GE, Russo SJ. Immune and Neuroendocrine Mechanisms of Stress Vulnerability and Resilience. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2017;42(1):62-80.
  32. Zimmermann U, Kraus T, Himmerich H, Schuld A, Pollmacher T. Epidemiology, implications and mechanisms underlying drug-induced weight gain in psychiatric patients. Journal of psychiatric research. 2003;37(3):193-220.
  33. Vandenberghe F, Gholam-Rezaee M, Saigi-Morgui N, Delacretaz A, Choong E, Solida-Tozzi A, Kolly S, Thonney J, Gallo SF, Hedjal A, Ambresin AE, von Gunten A, Conus P, Eap CB. Importance of early weight changes to predict long-term weight gain during psychotropic drug treatment. J Clin Psychiatry. 2015;76(11):e1417-1423.
  34. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.
  35. Serretti A, Mandelli L. Antidepressants and body weight: a comprehensive review and meta-analysis. The Journal of clinical psychiatry. 2010;71(10):1259-1272.
  36. Masand PS, Gupta S. Long-term side effects of newer-generation antidepressants: SSRIS, venlafaxine, nefazodone, bupropion, and mirtazapine. Annals of clinical psychiatry : official journal of the American Academy of Clinical Psychiatrists. 2002;14(3):175-182.
  37. Gadde KM, Xiong GL. Bupropion for weight reduction. Expert review of neurotherapeutics. 2007;7(1):17-24.
  38. Billes SK, Sinnayah P, Cowley MA. Naltrexone/bupropion for obesity: an investigational combination pharmacotherapy for weight loss. Pharmacol Res. 2014;84:1-11.
  39. Livingstone C, Rampes H. Lithium: a review of its metabolic adverse effects. J Psychopharmacol. 2006;20(3):347-355.
  40. Chen Y, Silverstone T. Lithium and weight gain. Int Clin Psychopharmacol. 1990;5(3):217-225.
  41. Sohn M, Moga DC, Blumenschein K, Talbert J. National trends in off-label use of atypical antipsychotics in children and adolescents in the United States. Medicine. 2016;95(23):e3784.
  42. Verdoux H, Tournier M, Begaud B. Antipsychotic prescribing trends: a review of pharmaco-epidemiological studies. Acta Psychiatr Scand. 2010;121(1):4-10.
  43. Seeman P, Lee T, Chau-Wong M, Wong K. Antipsychotic drug doses and neuroleptic/dopamine receptors. Nature. 1976;261(5562):717-719.
  44. Allison DB, Casey DE. Antipsychotic-induced weight gain: a review of the literature. J Clin Psychiatry. 2001;62 Suppl 7:22-31.
  45. Allison DB, Mentore JL, Heo M, Chandler LP, Cappelleri JC, Infante MC, Weiden PJ. Antipsychotic-induced weight gain: a comprehensive research synthesis. Am J Psychiatry. 1999;156(11):1686-1696.
  46. Correll CU, Manu P, Olshanskiy V, Napolitano B, Kane JM, Malhotra AK. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. Jama. 2009;302(16):1765-1773.
  47. Hoffmann VP, Case M, Stauffer VL, Jacobson JG, Conley RR. Predictive value of early changes in triglycerides and weight for longer-term changes in metabolic measures during olanzapine, ziprasidone or aripiprazole treatment for schizophrenia and schizoaffective disorder post hoc analyses of 3 randomized, controlled clinical trials. Journal of clinical psychopharmacology. 2010;30(6):656-660.
  48. Raben AT, Marshe VS, Chintoh A, Gorbovskaya I, Muller DJ, Hahn MK. The Complex Relationship between Antipsychotic-Induced Weight Gain and Therapeutic Benefits: A Systematic Review and Implications for Treatment. Frontiers in neuroscience. 2017;11:741.
  49. Ballon JS, Pajvani U, Freyberg Z, Leibel RL, Lieberman JA. Molecular pathophysiology of metabolic effects of antipsychotic medications. Trends Endocrinol Metab. 2014;25(11):593-600.
  50. Vestri HS, Maianu L, Moellering DR, Garvey WT. Atypical antipsychotic drugs directly impair insulin action in adipocytes: effects on glucose transport, lipogenesis, and antilipolysis. Neuropsychopharmacology. 2007;32(4):765-772.
  51. Mitchell AJ, Vancampfort D, Sweers K, van Winkel R, Yu W, De Hert M. Prevalence of metabolic syndrome and metabolic abnormalities in schizophrenia and related disorders--a systematic review and meta-analysis. Schizophr Bull. 2013;39(2):306-318.
  52. Holt RI, Peveler RC. Association between antipsychotic drugs and diabetes. Diabetes, obesity & metabolism. 2006;8(2):125-135.
  53. De Hert M, Detraux J, van Winkel R, Yu W, Correll CU. Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nat Rev Endocrinol. 2011;8(2):114-126.
  54. Henderson DC, Cagliero E, Gray C, Nasrallah RA, Hayden DL, Schoenfeld DA, Goff DC. Clozapine, diabetes mellitus, weight gain, and lipid abnormalities: A five-year naturalistic study. Am J Psychiatry. 2000;157(6):975-981.
  55. Balf G, Stewart TD, Whitehead R, Baker RA. Metabolic adverse events in patients with mental illness treated with antipsychotics: a primary care perspective. Prim Care Companion J Clin Psychiatry. 2008;10(1):15-24.
  56. Orsolini L, Tomasetti C, Valchera A, Vecchiotti R, Matarazzo I, Vellante F, Iasevoli F, Buonaguro EF, Fornaro M, Fiengo AL, Martinotti G, Mazza M, Perna G, Carano A, De Bartolomeis A, Di Giannantonio M, De Berardis D. An update of safety of clinically used atypical antipsychotics. Expert opinion on drug safety. 2016;15(10):1329-1347.
  57. Dwyer DS, Donohoe D. Induction of hyperglycemia in mice with atypical antipsychotic drugs that inhibit glucose uptake. Pharmacology, biochemistry, and behavior. 2003;75(2):255-260.
  58. Daumit GL, Goff DC, Meyer JM, Davis VG, Nasrallah HA, McEvoy JP, Rosenheck R, Davis SM, Hsiao JK, Stroup TS, Lieberman JA. Antipsychotic effects on estimated 10-year coronary heart disease risk in the CATIE schizophrenia study. Schizophr Res. 2008;105(1-3):175-187.
  59. Alvarez-Jimenez M, Hetrick SE, Gonzalez-Blanch C, Gleeson JF, McGorry PD. Non-pharmacological management of antipsychotic-induced weight gain: systematic review and meta-analysis of randomised controlled trials. The British journal of psychiatry : the journal of mental science. 2008;193(2):101-107.
  60. Mukundan A, Faulkner G, Cohn T, Remington G. Antipsychotic switching for people with schizophrenia who have neuroleptic-induced weight or metabolic problems. The Cochrane database of systematic reviews. 2010(12):Cd006629.
  61. Gierisch JM, Nieuwsma JA, Bradford DW, Wilder CM, Mann-Wrobel MC, McBroom AJ, Wing L, Musty MD, Chobot MM, Hasselblad V, Williams JW, Jr. AHRQ Comparative Effectiveness Reviews. Interventions To Improve Cardiovascular Risk Factors in People With Serious Mental Illness. Rockville (MD): Agency for Healthcare Research and Quality (US); 2013.
  62. Mizuno Y, Suzuki T, Nakagawa A, Yoshida K, Mimura M, Fleischhacker WW, Uchida H. Pharmacological strategies to counteract antipsychotic-induced weight gain and metabolic adverse effects in schizophrenia: a systematic review and meta-analysis. Schizophr Bull. 2014;40(6):1385-1403.
  63. Maayan L, Vakhrusheva J, Correll CU. Effectiveness of medications used to attenuate antipsychotic-related weight gain and metabolic abnormalities: a systematic review and meta-analysis. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2010;35(7):1520-1530.
  64. Zheng W, Li XB, Tang YL, Xiang YQ, Wang CY, de Leon J. Metformin for Weight Gain and Metabolic Abnormalities Associated With Antipsychotic Treatment: Meta-Analysis of Randomized Placebo-Controlled Trials. Journal of clinical psychopharmacology. 2015;35(5):499-509.
  65. Hu Y, Young AJ, Ehli EA, Nowotny D, Davies PS, Droke EA, Soundy TJ, Davies GE. Metformin and berberine prevent olanzapine-induced weight gain in rats. PloS one. 2014;9(3):e93310.
  66. Ebdrup BH, Knop FK, Ishoy PL, Rostrup E, Fagerlund B, Lublin H, Glenthoj B. Glucagon-like peptide-1 analogs against antipsychotic-induced weight gain: potential physiological benefits. BMC Med. 2012;10:92.
  67. Lykkegaard K, Larsen PJ, Vrang N, Bock C, Bock T, Knudsen LB. The once-daily human GLP-1 analog, liraglutide, reduces olanzapine-induced weight gain and glucose intolerance. Schizophr Res. 2008;103(1-3):94-103.
  68. Sharma AN, Ligade SS, Sharma JN, Shukla P, Elased KM, Lucot JB. GLP-1 receptor agonist liraglutide reverses long-term atypical antipsychotic treatment associated behavioral depression and metabolic abnormalities in rats. Metab Brain Dis. 2015;30(2):519-527.
  69. Larsen JR, Vedtofte L, Jakobsen MSL, Jespersen HR, Jakobsen MI, Svensson CK, Koyuncu K, Schjerning O, Oturai PS, Kjaer A, Nielsen J, Holst JJ, Ekstrom CT, Correll CU, Vilsboll T, Fink-Jensen A. Effect of Liraglutide Treatment on Prediabetes and Overweight or Obesity in Clozapine- or Olanzapine-Treated Patients With Schizophrenia Spectrum Disorder: A Randomized Clinical Trial. JAMA psychiatry. 2017;74(7):719-728.
  70. Gross C, Blasey CM, Roe RL, Belanoff JK. Mifepristone reduces weight gain and improves metabolic abnormalities associated with risperidone treatment in normal men. Obesity (Silver Spring). 2010;18(12):2295-2300.
  71. Rimessi A, Pavan C, Ioannidi E, Nigro F, Morganti C, Brugnoli A, Longo F, Gardin C, Ferroni L, Morari M, Vindigni V, Zavan B, Pinton P. Protein Kinase C beta: a New Target Therapy to Prevent the Long-Term Atypical Antipsychotic-Induced Weight Gain. Neuropsychopharmacology. 2017;42(7):1491-1501.
  72. Capuron L, Lasselin J, Castanon N. Role of Adiposity-Driven Inflammation in Depressive Morbidity. Neuropsychopharmacology. 2017;42(1):115-128.
  73. Muller N. COX-2 inhibitors as antidepressants and antipsychotics: clinical evidence. Current opinion in investigational drugs (London, England : 2000). 2010;11(1):31-42.
  74. Ben-Menachem E. Weight issues for people with epilepsy--a review. Epilepsia. 2007;48 Suppl 9:42-45.
  75. Verrotti A, D'Egidio C, Mohn A, Coppola G, Chiarelli F. Weight gain following treatment with valproic acid: pathogenetic mechanisms and clinical implications. Obes Rev. 2011;12(5):e32-43.
  76. Belcastro V, D'Egidio C, Striano P, Verrotti A. Metabolic and endocrine effects of valproic acid chronic treatment. Epilepsy Res. 2013;107(1-2):1-8.
  77. Aycicek A, Iscan A. The effects of carbamazepine, valproic acid and phenobarbital on the oxidative and antioxidative balance in epileptic children. Eur Neurol. 2007;57(2):65-69.
  78. Wong HY, Chu TS, Lai JC, Fung KP, Fok TF, Fujii T, Ho YY. Sodium valproate inhibits glucose transport and exacerbates Glut1-deficiency in vitro. J Cell Biochem. 2005;96(4):775-785.
  79. Farinelli E, Giampaoli D, Cenciarini A, Cercado E, Verrotti A. Valproic acid and nonalcoholic fatty liver disease: A possible association? World J Hepatol. 2015;7(9):1251-1257.
  80. Swann AC. Major system toxicities and side effects of anticonvulsants. J Clin Psychiatry. 2001;62 Suppl 14:16-21.
  81. Antel J, Hebebrand J. Weight-reducing side effects of the antiepileptic agents topiramate and zonisamide. Handb Exp Pharmacol. 2012(209):433-466.
  82. Smith SM, Meyer M, Trinkley KE. Phentermine/topiramate for the treatment of obesity. The Annals of pharmacotherapy. 2013;47(3):340-349.
  83. Curtis JR, Westfall AO, Allison J, Bijlsma JW, Freeman A, George V, Kovac SH, Spettell CM, Saag KG. Population-based assessment of adverse events associated with long-term glucocorticoid use. Arthritis and rheumatism. 2006;55(3):420-426.
  84. Christ-Crain M, Kola B, Lolli F, Fekete C, Seboek D, Wittmann G, Feltrin D, Igreja SC, Ajodha S, Harvey-White J, Kunos G, Muller B, Pralong F, Aubert G, Arnaldi G, Giacchetti G, Boscaro M, Grossman AB, Korbonits M. AMP-activated protein kinase mediates glucocorticoid-induced metabolic changes: a novel mechanism in Cushing's syndrome. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2008;22(6):1672-1683.
  85. Larsen PJ, Jessop DS, Chowdrey HS, Lightman SL, Mikkelsen JD. Chronic administration of glucocorticoids directly upregulates prepro-neuropeptide Y and Y1-receptor mRNA levels in the arcuate nucleus of the rat. J Neuroendocrinol. 1994;6(2):153-159.
  86. Harfstrand A, Cintra A, Fuxe K, Aronsson M, Wikstrom AC, Okret S, Gustafsson JA, Agnati LF. Regional differences in glucocorticoid receptor immunoreactivity among neuropeptide Y immunoreactive neurons of the rat brain. Acta Physiol Scand. 1989;135(1):3-9.
  87. Soumano K, Desbiens S, Rabelo R, Bakopanos E, Camirand A, Silva JE. Glucocorticoids inhibit the transcriptional response of the uncoupling protein-1 gene to adrenergic stimulation in a brown adipose cell line. Molecular and cellular endocrinology. 2000;165(1-2):7-15.
  88. Bowles NP, Karatsoreos IN, Li X, Vemuri VK, Wood JA, Li Z, Tamashiro KL, Schwartz GJ, Makriyannis AM, Kunos G, Hillard CJ, McEwen BS, Hill MN. A peripheral endocannabinoid mechanism contributes to glucocorticoid-mediated metabolic syndrome. Proc Natl Acad Sci U S A. 2015;112(1):285-290.
  89. Fardet L, Feve B. Systemic glucocorticoid therapy: a review of its metabolic and cardiovascular adverse events. Drugs. 2014;74(15):1731-1745.
  90. Fardet L, Cabane J, Lebbe C, Morel P, Flahault A. Incidence and risk factors for corticosteroid-induced lipodystrophy: a prospective study. J Am Acad Dermatol. 2007;57(4):604-609.
  91. Dube S, Slama MQ, Basu A, Rizza RA, Basu R. Glucocorticoid Excess Increases Hepatic 11beta-HSD-1 Activity in Humans: Implications in Steroid-Induced Diabetes. J Clin Endocrinol Metab. 2015;100(11):4155-4162.
  92. Wolf G. Glucocorticoids in adipocytes stimulate visceral obesity. Nutr Rev. 2002;60(5 Pt 1):148-151.
  93. Karatasakis A, Danek BA, Karacsonyi J, Rangan BV, Roesle MK, Knickelbine T, Miedema MD, Khalili H, Ahmad Z, Abdullah S, Banerjee S, Brilakis ES. Effect of PCSK9 Inhibitors on Clinical Outcomes in Patients With Hypercholesterolemia: A Meta-Analysis of 35 Randomized Controlled Trials. J Am Heart Assoc. 2017;6(12).
  94. Baragetti A, Balzarotti G, Grigore L, Pellegatta F, Guerrini U, Pisano G, Fracanzani AL, Fargion S, Norata GD, Catapano AL. PCSK9 deficiency results in increased ectopic fat accumulation in experimental models and in humans. Eur J Prev Cardiol. 2017;24(17):1870-1877.
  95. Schmidt AF, Swerdlow DI, Holmes MV, Patel RS, Fairhurst-Hunter Z, Lyall DM, Hartwig FP, Horta BL, Hypponen E, Power C, Moldovan M, van Iperen E, Hovingh GK, Demuth I, Norman K, Steinhagen-Thiessen E, Demuth J, Bertram L, Liu T, Coassin S, Willeit J, Kiechl S, Willeit K, Mason D, Wright J, Morris R, Wanamethee G, Whincup P, Ben-Shlomo Y, McLachlan S, Price JF, Kivimaki M, Welch C, Sanchez-Galvez A, Marques-Vidal P, Nicolaides A, Panayiotou AG, Onland-Moret NC, van der Schouw YT, Matullo G, Fiorito G, Guarrera S, Sacerdote C, Wareham NJ, Langenberg C, Scott R, Luan J, Bobak M, Malyutina S, Pajak A, Kubinova R, Tamosiunas A, Pikhart H, Husemoen LL, Grarup N, Pedersen O, Hansen T, Linneberg A, Simonsen KS, Cooper J, Humphries SE, Brilliant M, Kitchner T, Hakonarson H, Carrell DS, McCarty CA, Kirchner HL, Larson EB, Crosslin DR, de Andrade M, Roden DM, Denny JC, Carty C, Hancock S, Attia J, Holliday E, O'Donnell M, Yusuf S, Chong M, Pare G, van der Harst P, Said MA, Eppinga RN, Verweij N, Snieder H, Christen T, Mook-Kanamori DO, Gustafsson S, Lind L, Ingelsson E, Pazoki R, Franco O, Hofman A, Uitterlinden A, Dehghan A, Teumer A, Baumeister S, Dorr M, Lerch MM, Volker U, Volzke H, Ward J, Pell JP, Smith DJ, Meade T, Maitland-van der Zee AH, Baranova EV, Young R, Ford I, Campbell A, Padmanabhan S, Bots ML, Grobbee DE, Froguel P, Thuillier D, Balkau B, Bonnefond A, Cariou B, Smart M, Bao Y, Kumari M, Mahajan A, Ridker PM, Chasman DI, Reiner AP, Lange LA, Ritchie MD, Asselbergs FW, Casas JP, Keating BJ, Preiss D, Hingorani AD, Sattar N. PCSK9 genetic variants and risk of type 2 diabetes: a mendelian randomisation study. The lancet Diabetes & endocrinology. 2017;5(2):97-105.
  96. Norata GD, Tavori H, Pirillo A, Fazio S, Catapano AL. Biology of proprotein convertase subtilisin kexin 9: beyond low-density lipoprotein cholesterol lowering. Cardiovasc Res. 2016;112(1):429-442.
  97. Roubtsova A, Munkonda MN, Awan Z, Marcinkiewicz J, Chamberland A, Lazure C, Cianflone K, Seidah NG, Prat A. Circulating proprotein convertase subtilisin/kexin 9 (PCSK9) regulates VLDLR protein and triglyceride accumulation in visceral adipose tissue. Arterioscler Thromb Vasc Biol. 2011;31(4):785-791.
  98. Wierzbicki AS, Viljoen A. Anti-sense oligonucleotide therapies for the treatment of hyperlipidaemia. Expert Opin Biol Ther. 2016;16(9):1125-1134.
  99. Hashemi N, Odze RD, McGowan MP, Santos RD, Stroes ES, Cohen DE. Liver histology during Mipomersen therapy for severe hypercholesterolemia. J Clin Lipidol. 2014;8(6):606-611.
  100. Berberich AJ, Hegele RA. Lomitapide for the treatment of hypercholesterolemia. Expert Opin Pharmacother. 2017;18(12):1261-1268.
  101. Alger HM, Brown JM, Sawyer JK, Kelley KL, Shah R, Wilson MD, Willingham MC, Rudel LL. Inhibition of acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2) prevents dietary cholesterol-associated steatosis by enhancing hepatic triglyceride mobilization. J Biol Chem. 2010;285(19):14267-14274.
  102. Meuwese MC, Franssen R, Stroes ES, Kastelein JJ. And then there were acyl coenzyme A:cholesterol acyl transferase inhibitors. Current opinion in lipidology. 2006;17(4):426-430.
  103. Chen D, Misra A, Garg A. Clinical review 153: Lipodystrophy in human immunodeficiency virus-infected patients. J Clin Endocrinol Metab. 2002;87(11):4845-4856.
  104. Mirza FS, Luthra P, Chirch L. Endocrinological aspects of HIV infection. J Endocrinol Invest. 2018;41(8):881-899.
  105. Hulgan T, Haubrich R, Riddler SA, Tebas P, Ritchie MD, McComsey GA, Haas DW, Canter JA. European mitochondrial DNA haplogroups and metabolic changes during antiretroviral therapy in AIDS Clinical Trials Group Study A5142. AIDS. 2011;25(1):37-47.
  106. de Waal R, Cohen K, Maartens G. Systematic review of antiretroviral-associated lipodystrophy: lipoatrophy, but not central fat gain, is an antiretroviral adverse drug reaction. PloS one. 2013;8(5):e63623.
  107. McComsey GA, Moser C, Currier J, Ribaudo HJ, Paczuski P, Dube MP, Kelesidis T, Rothenberg J, Stein JH, Brown TT. Body Composition Changes After Initiation of Raltegravir or Protease Inhibitors: ACTG A5260s. Clin Infect Dis. 2016;62(7):853-862.
  108. Erlandson KM, Lake JE. Fat Matters: Understanding the Role of Adipose Tissue in Health in HIV Infection. Curr HIV/AIDS Rep. 2016;13(1):20-30.
  109. Scherzer R, Heymsfield SB, Lee D, Powderly WG, Tien PC, Bacchetti P, Shlipak MG, Grunfeld C, Study of Fat R, Metabolic Change in HIVI. Decreased limb muscle and increased central adiposity are associated with 5-year all-cause mortality in HIV infection. AIDS. 2011;25(11):1405-1414.
  110. Lundgren JD, Battegay M, Behrens G, De Wit S, Guaraldi G, Katlama C, Martinez E, Nair D, Powderly WG, Reiss P, Sutinen J, Vigano A, Committee EE. European AIDS Clinical Society (EACS) guidelines on the prevention and management of metabolic diseases in HIV. HIV Med. 2008;9(2):72-81.
  111. Caron-Debarle M, Lagathu C, Boccara F, Vigouroux C, Capeau J. HIV-associated lipodystrophy: from fat injury to premature aging. Trends in molecular medicine. 2010;16(5):218-229.
  112. Giralt M, Domingo P, Guallar JP, Rodriguez de la Concepcion ML, Alegre M, Domingo JC, Villarroya F. HIV-1 infection alters gene expression in adipose tissue, which contributes to HIV- 1/HAART-associated lipodystrophy. Antiviral therapy. 2006;11(6):729-740.
  113. Koczor CA, Lewis W. Nucleoside reverse transcriptase inhibitor toxicity and mitochondrial DNA. Expert Opin Drug Metab Toxicol. 2010;6(12):1493-1504.
  114. Giralt M, Domingo P, Villarroya F. Adipose tissue biology and HIV-infection. Best Pract Res Clin Endocrinol Metab. 2011;25(3):487-499.
  115. Flint OP, Noor MA, Hruz PW, Hylemon PB, Yarasheski K, Kotler DP, Parker RA, Bellamine A. The role of protease inhibitors in the pathogenesis of HIV-associated lipodystrophy: cellular mechanisms and clinical implications. Toxicol Pathol. 2009;37(1):65-77.
  116. Diaz-Delfin J, del Mar Gutierrez M, Gallego-Escuredo JM, Domingo JC, Gracia Mateo M, Villarroya F, Domingo P, Giralt M. Effects of nevirapine and efavirenz on human adipocyte differentiation, gene expression, and release of adipokines and cytokines. Antiviral research. 2011;91(2):112-119.
  117. Hruz PW. Molecular mechanisms for insulin resistance in treated HIV-infection. Best Pract Res Clin Endocrinol Metab. 2011;25(3):459-468.
  118. Rietschel P, Hadigan C, Corcoran C, Stanley T, Neubauer G, Gertner J, Grinspoon S. Assessment of growth hormone dynamics in human immunodeficiency virus-related lipodystrophy. J Clin Endocrinol Metab. 2001;86(2):504-510.
  119. Dube MP, Stein JH, Aberg JA, Fichtenbaum CJ, Gerber JG, Tashima KT, Henry WK, Currier JS, Sprecher D, Glesby MJ. 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.
  120. Schmitz M, Michl GM, Walli R, Bogner J, Bedynek A, Seidel D, Goebel FD, Demant T. Alterations of apolipoprotein B metabolism in HIV-infected patients with antiretroviral combination therapy. J Acquir Immune Defic Syndr. 2001;26(3):225-235.
  121. Herink M, Ito MK. Medication Induced Changes in Lipid and Lipoproteins. 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, eds. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  122. Martin A, Smith DE, Carr A, Ringland C, Amin J, Emery S, Hoy J, Workman C, Doong N, Freund J, Cooper DA, Mitochondrial Toxicity Study G. Reversibility of lipoatrophy in HIV-infected patients 2 years after switching from a thymidine analogue to abacavir: the MITOX Extension Study. AIDS. 2004;18(7):1029-1036.
  123. Norwood J, Turner M, Bofill C, Rebeiro P, Shepherd B, Bebawy S, Hulgan T, Raffanti S, Haas DW, Sterling TR, Koethe JR. Brief Report: Weight Gain in Persons With HIV Switched From Efavirenz-Based to Integrase Strand Transfer Inhibitor-Based Regimens. J Acquir Immune Defic Syndr. 2017;76(5):527-531.
  124. Rochira V, Guaraldi G. Growth hormone deficiency and human immunodeficiency virus. Best Pract Res Clin Endocrinol Metab. 2017;31(1):91-111.
  125. Edgeworth A, Treacy MP, Hurst TP. Thiazolidinediones in the treatment of HIV/HAART-associated lipodystrophy syndrome. AIDS Rev. 2013;15(3):171-180.
  126. Raboud JM, Diong C, Carr A, Grinspoon S, Mulligan K, Sutinen J, Rozenbaum W, Cavalcanti RB, Wand H, Costagliola D, Walmsley S, Glitazone, Lipoatrophy Meta-Analysis Working G. A meta-analysis of six placebo-controlled trials of thiazolidinedione therapy for HIV lipoatrophy. HIV Clin Trials. 2010;11(1):39-50.
  127. Amouyal C, Buyse M, Lucas-Martini L, Hirt D, Genser L, Torcivia A, Bouillot JL, Oppert JM, Aron-Wisnewsky J. Sleeve Gastrectomy in Morbidly Obese HIV Patients: Focus on Anti-retroviral Treatment Absorption After Surgery. Obesity surgery. 2018;28(9):2886-2893.
  128. Tsoukas MA, Farr OM, Mantzoros CS. Leptin in congenital and HIV-associated lipodystrophy. Metabolism. 2015;64(1):47-59.
  129. Jagdeo J, Ho D, Lo A, Carruthers A. A systematic review of filler agents for aesthetic treatment of HIV facial lipoatrophy (FLA). J Am Acad Dermatol. 2015;73(6):1040-1054 e1014.

Metabolic Syndrome

ABSTRACT

 

Significant interest exists in understanding the shared metabolic dysregulation leading to obesity, diabetes, and cardiovascular disease (CVD).  Hence came the concept of the “metabolic syndrome” (MetS).  Reaven first described MetS in his 1988 Banting lecture as “Syndrome X”.  Reaven suggested that the syndrome hinged on the existence of insulin resistance and resulted in glucose intolerance, hypertension and dyslipidemia. The World Health Organization (WHO) produced the first formalized definition of the MetS in 1998.  Since then multiple definitions of the syndrome have been proposed, the most recent being the Harmonized Definition where 3 of the 5 risk factors are present: enlarged waist circumference with population-specific and country-specific criteria; triglycerides ≥ 150 mg/dL, HDL-C < 40 mg/dL in men and < 50 mg/dL in women, systolic blood pressure ≥ 130 mm Hg or diastolic blood pressure ≥ 85 mm Hg and fasting glucose > 100 mg/dL, with the inclusion of patients taking medication to manage hypertriglyceridemia, low HDL-C, hypertension, and hyperglycemia. The National Health and Nutrition Examination Survey (NHANES) estimated the overall prevalence of MetS in adults (aged ≥ 20 years) in the United States as 33% from 2003 to 2012.  The high prevalence is particularly alarming given that MetS also predisposes to a number of serious conditions beyond diabetes and CVD including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), polycystic ovarian syndrome (PCOS), obstructive sleep apnea (OSA), cancer, and many other serious disease states.  Hence, early identification and intervention are warranted. Lifestyle modification is the foundational intervention in treatment of MetS.  Specifically, a healthy low-calorie, low fat diet and moderate physical activity of at least 150 minutes/week, resulting in a weight reduction of 7%.  Obesity, hypertension and dyslipidemia may also be treated pharmacologically to meet individualized patient goals. Beyond the clinic imperative around MetS are its pathophysiologic underpinnings.  This review will focus on the investigative work into the proximal origins of the MetS.  Defects in insulin signaling occur in a shared environment of pro-inflammation, untoward adipokines coming from dysregulated fatty acid metabolism, as well as novel pathways involving the gut microbiota.  Collectively, MetS continues to exist as a fertile area of research yielding significant insights into early events leading to the most prevalent cause of human morbidity and mortality. For in depth review of all related aspects of endocrinology, visit www.endotext.org.

 

HISTORY AND DEFINITIONS

 

The metabolic syndrome (MetS) is a compilation of risk factors that predispose individuals to the development of type 2 diabetes (T2DM) and cardiovascular disease (CVD).  Reaven (1) first described MetS in his 1988 Banting lecture as “Syndrome X. ” Reaven suggested that insulin resistance clustered together with glucose intolerance, dyslipidemia, and hypertension to increase the risk of CVD.  The initial definition of metabolic syndrome included impaired glucose tolerance (IGT), hyperinsulinemia, elevated triglycerides (TG), and reduced high-density lipoprotein cholesterol (HDL-C). Hyperuricemia, microvascular angina, and elevated plasminogen activator inhibitor 1 (PAI-1) were later proposed as possible additional components of the same syndrome (1,2). Obesity was not included as part of Syndrome X as Reaven believed that insulin resistance, not obesity, was the common denominator. Reaven noted that all of the elements of Syndrome X could occur in non-obese individuals, and while he acknowledged that obesity could lead to a decrease in insulin mediated glucose uptake, he stressed that obesity was only one of the environmental factors that affect insulin sensitivity (3,4).

 

The World Health Organization (WHO) produced the first formalized definition of the MetS in 1998. The working definition included impaired glucose tolerance (IGT), impaired fasting glucose (IFG) or diabetes mellitus and/or insulin resistance (as measured using a hyperinsulinemic euglycemic clamp study) together with two or more additional components. Additional components included hypertension (defined as a blood pressure ≥160/90 mm Hg), raised plasma triglycerides (≥150 mg/dl) and/or low HDL-C (<35 mg/dl for men and <39 mg/dl for women), central obesity (defined either as body mass index (BMI) > 30 kg/m2 or waist to hip ratio>0.90 for males and >0.85 for females) and microalbuminuria (5). Critics questioned the practicality of this definition given the need for a hyperinsulinemic clamp study. Others argued that measuring waist circumference was superior in terms of convenience to the waist to hip ratio with similar correlations to obesity. Additionally, there was a question about the value of including microalbuminuria in the definition as there was insufficient evidence of a connection with insulin resistance (5).

 

These critiques led to the first revision of the definition of the syndrome in 1999 by the European Group for the Study of Insulin Resistance (EGIR). They renamed the syndrome the “insulin resistance syndrome” (IRS) as it included non-metabolic features. They excluded patients with diabetes because of the difficulty of measuring insulin resistance in these individuals. The need for hyperinsulinemic clamp studies was obviated by defining insulin resistance as a fasting insulin level above the 75th percentile for the population. Additional criteria (elements associated with increased risk of coronary artery disease by the Second Joint Task Force of European and other Societies on Coronary Prevention) were also included, namely obesity (defined as waist circumference ≥ 94 cm (37 inches) for men and ≥ 80 cm (32 inches) for women), hypertension (now defined as a blood pressure ≥140/90 mm Hg) and dyslipidemia (with triglycerides ≥ 180 mg/dl or HDL-C ≤ 39). Additionally, microalbuminuria was omitted from the definition (6).

 

The National Cholesterol Education Program (NCEP) Adult Treatment Panel III (ATP III) recognized that these multiple metabolic elements were cardiovascular risk factors and renamed the constellation of these metabolic risk factors as “The Metabolic Syndrome” (7). The criteria included any three of the following: obesity (defined as waist circumference ≥ 102 cm (40 inches) in males and ≥ 88 cm (35 inches) in females (based on the 1998 National Institutes of Health (NIH) obesity clinical guidelines; pediatric definitions use standardized Z-scores rather than waist circumference (8)), hypertension (defined as blood pressure ≥ 130/85 mm Hg based on the Joint National Committee guidelines), fasting glucose > 110 mg/dL, triglycerides ≥ 150 mg/dL and HDL-C < 40 mg/dL.  Additionally, in this report MetS was recognized as a secondary target of risk reduction therapy after the primary target of LDL cholesterol (7).

 

In 2003, the American Association of Clinical Endocrinologists (AACE) modified the ATP III criteria and restored the condition to the name “Insulin Resistance Syndrome,” again highlighting the central role of insulin resistance in the pathogenesis of the syndrome (9). This definition did not rely on strict diagnostic criteria. The components of the syndrome included some degree of glucose intolerance (but not overt diabetes), abnormal uric acid metabolism, dyslipidemia, hemodynamic changes (including hypertension), prothrombotic factors, markers of inflammation, and endothelial dysfunction. The AACE position statement also identified factors that increased the likelihood of developing the insulin resistance syndrome, including a diagnosis of CVD, hypertension, polycystic ovarian syndrome (PCOS), non-alcoholic fatty liver disease (NAFLD) or acanthosis nigricans, a family history of T2DM, hypertension or CVD, a personal history of gestational diabetes (GDM) or glucose intolerance, non-Caucasian ethnicity,  a sedentary lifestyle,  overweight/obesity (defined as BMI > 25 kg/m2 or waist circumference > 40 inches in men and > 35 inches in women) and age > 40 years (9).

 

The International Diabetes Federation (IDF) aimed to create a straightforward, clinically useful definition to identify individuals in any country worldwide at high risk of CVD and diabetes and to allow for comparative epidemiologic studies.  This resulted in the IDF consensus definition of MetS in 2005 (10). Central obesity, as defined as BMI> 30 kg/m2 or if ≤ 30 kg/m2 by ethnic specific waist circumference measurements) was a requisite for the syndrome. Additionally, the definition required the presence of two of the following four elements: triglycerides ≥ 150 mg/dL, HDL-C < 40 mg/dL  in men or < 50 mg/dL in women, systolic blood pressure ≥ 130 mmHg or diastolic blood pressure ≥ 85 mmHg, fasting glucose > 100 mg/dL ( based on the 2003 ADA definition of IFG) (11) including diabetes and those with a prior diagnosis of or treatment of any of these conditions (10).

 

In 2005, the American Heart Association (AHA)/ National Heart, Lung and Blood Institute (NHLBI) also suggested criteria for diagnosis of the metabolic syndrome. Their revised definition of the metabolic syndrome was based on the ATP III criteria and required three of any of the five following criteria: elevated waist circumference ( ≥ 102 cm (40 inches) in males and ≥ 88 cm (35 inches) in females) , triglycerides ≥ 150 mg/dL and HDL-C < 40 mg/dL in men and < 50 mg/dL in women, elevated blood pressure ≥ 130/85 mm Hg and elevated fasting glucose > 100 mg/dL (12). As suggested by the IDF, ethnic-specific waist circumferences were taken into account when using this definition. Additionally, impaired fasting glucose was defined as >100 mg/dl, which was also consistent with the IDF guidelines.

 

Finally, in an effort to provide more consistency in both clinical care and research of patients with MetS, the IDF, NHBLI, AHA, World Heart Federation, and the International Association for the Study of Obesity published a joint statement in 2009 that provided a “harmonized” definition of MetS (13). According to this joint statement, a diagnosis of the MetS is made when any 3 of the 5 following risk factors are present (Table 1): enlarged waist circumference with population-specific and country-specific criteria; elevated triglycerides, defined as ≥ 150 mg/dL, decreased HDL-C, defined as < 40 mg/dL in men and < 50 mg/dL in women, elevated blood pressure, defined as systolic blood pressure ≥ 130 mm Hg or diastolic blood pressure ≥ 85 mm Hg and elevated fasting glucose, defined as blood glucose  > 100 mg/dL, with the inclusion of patients taking medication to manage hypertriglyceridemia, low HDL-C, hypertension. and hyperglycemia. This definition is frequently referred to as the current Harmonization definition.

 

Table 1. Criteria for Diagnosis of the Metabolic Syndrome

Measure

Categorical Cut-Points

        Waist circumference

Population and country specific     definitions

        Triglycerides *

          ≥ 150 mg/dL

        High Density Lipoprotein Cholesterol (HDL-C)*

Men < 40 mg/dL       Women < 50 mg/dL

        Blood Pressure*

          ≥ 130/ ≥85

        Fasting Glucose*

          ≥ 100 mg/dL

*Drug treatment for elevated triglycerides, low HDL-C, elevated blood pressure or elevated glucose are alternate indicators

 

It is important to note that in the current Harmonization definition, obesity is diagnosed using waist circumference and not BMI as waist circumference has been shown to better correlate with visceral adiposity and insulin resistance as well as the development of T2DM and CVD than does BMI (10,14,15). Subsequent to the establishment of the harmonized definition, waist to height ratio has been demonstrated to be superior to waist circumference and BMI as a screening tool for cardiometabolic risk factors (diabetes, hypertension, cardiovascular disease, and all outcomes) as well as predicting whole-body fat percentage and visceral adipose tissue mass (16,17).  It is not clear if the definition of MetS will be revised over time to reflect these new findings. Additionally, in the current Harmonization definition, ethnic-specific waist circumference cut-off values are used, as it has been shown that certain ethnic groups, especially South Asian populations, have higher degrees of visceral adiposity for given waist circumference measurements compared to Europeans (10,13,18).

 

PREVALENCE

 

The prevalence of MetS vary greatly depending on criteria used to define MetS, the age, gender, ethnicity and environment of the population being studied and obesity prevalence of the background population studied (25). Regardless of which criteria are used, however, the prevalence of MetS is high and is on the rise in many Western societies(26).

 

The National Health and Nutrition Examination Survey (NHANES) reported the overall prevalence of MetS in adults (aged ≥ 20 years) in the United States from 2003 to 2012 was 33% with the prevalence increasing with age, a finding that has been seen in other studies (24,25,27,28).  The NHANES report indicates the highest prevalence amongst Hispanics followed by non-Hispanic whites and blacks. Other studies have shown that American Indian, Hawaiian, Polynesian, and Filipino populations develop MetS more than individuals of European descent (27,29-33). Urban populations have higher rates of MetS than rural populations (34,35).  Similar to trends in Western societies, recent studies demonstrate rising rates of MetS in many developing countries (36,37). The development of these countries, bringing along a higher calorie diet and decreased physical activity, is thought to be largely responsible for the increased rate of MetS that is being observed (26,38,39). In summary, MetS affects a significant number of individuals worldwide.

 

CLINICAL UTILITY

 

The clinical utility of a diagnosis of MetS – vs. the individual components - has been studied extensively. Most recently, Pajunen and colleages compared the predictive ability of various definitions of MetS, namely the WHO, ATP III, IDF and new Harmonization definitions, found that all these definitions of MetS were significant predictors for incident CVD and T2DM. Additionally, the new Harmonization definition was found to be a better predictor of CVD endpoint than the sum of its components, as well as when compared to the Framingham risk score, but this was not the case for the prediction of T2DM (19).  Importantly, extensive, frequently conflicting literature exists examining the ability of the various definitions of MetS to predict outcomes.  Further, skeptics argue that making the diagnosis of MetS does not change the clinical management of these patients, as treatment of patients with MetS starts with diet and exercise and most physicians would offer the same recommendations to a patient with any of the individual elements of MetS (20,21).

 

In an attempt to settle some of the controversy, a WHO Expert Consultation was undertaken in November 2008. The panel concluded that MetS has limited practical utility as a diagnostic or management tool. They determined that MetS should not be applied as a clinical diagnosis, but rather should be considered a pre-morbid condition and that people with established diabetes or known cardiovascular disease should be excluded (22). They also stated that further attempts to redefine it are inappropriate in light of current knowledge and understanding (23). Despite the conclusions of the panel, the diagnosis of MetS is still commonly encountered in clinical practice as well as in the research arena and arguably applies to roughly one-third of the US adult population (24). It also predisposes to a number of serious conditions beyond diabetes and CVD including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), polycystic ovarian syndrome (PCOS), obstructive sleep apnea (OSA), cancer, and many others.

 

PATHOGENESIS

 

There are many different factors that contribute to the development of MetS. However, as initially proposed by Reaven, insulin resistance is thought to play a central role in connecting the different components of MetS and adding to the syndrome's development (1,40). Elevated free fatty acids (FFA) and abnormal adipokine profiles can both cause and result in insulin resistance and can manifest as MetS (41). In this section, we will discuss how these factors contribute to the development of the metabolic abnormalities that characterize insulin resistance and MetS.

 

Insulin Action and Signaling

 

Through its complex signaling cascades, insulin regulates glucose and fat metabolism. Pancreatic β-cells release insulin in response to increased circulating glucose levels and subsequently decreases plasma glucose concentrations by coordinately suppressing hepatic glucose production from amino acids and other intermediates of metabolism (gluconeogenesis) and glycogen (glycogenolysis), and enhancing glucose uptake into the muscle and adipose tissue by mobilization of the insulin-responsive glucose transporter 4 (GLUT4) (Fig. 1) (42). Through actions on hormone sensitive lipase, nuclear receptor PPARγ, and fatty acid synthase, insulin inhibits lipolysis, promotes adipogenesis and adipose tissue differentiation, and under conditions of chronic hyperinsulinemia paradoxically increases fatty acid synthase(41,43).

Figure 1: Normal Insulin Action: In individuals with normal insulin sensitivity, the pancreatic β-cells release insulin in response to increased circulating glucose levels (as seen in the postprandial state). Insulin then decreases the plasma glucose concentration by suppressing hepatic glucose output and enhancing glucose uptake into adipose tissue and by skeletal muscle.

 

Insulin resistance is most simply defined by its end organ effects; a decreased ability of insulin to suppress lipolysis and hepatic glucose production, as well as facilitate glucose uptake from peripheral tissues. There are numerous factors thought to mediate insulin resistance and its adverse effects in MetS. Despite its widespread appreciation in metabolic disease, insulin resistance is still not fully understood and remains an area of intense scientific investigation. In the following section, we will review the ways in which known factors affect insulin resistance in MetS.

 

It has been thoroughly documented that FFAs mediate many undesirable metabolic effects, especially insulin resistance (44). FFAs are thought to be increased in obesity secondary to increased fat mass. Additionally, under conditions of insulin resistance, insulin’s inhibitory effects on lipolysis are reduced, leading to a further increase in FFAs. Increased FFAs are not only a result of insulin resistance, but a cause as well, thus creating a vicious cycle. FFAs can lead to insulin resistance via a variety of mechanisms that include but are not limited to the Randle cycle, the accumulation of intracellular lipid derivatives such as diacylglycerol and ceramides, inflammatory signaling, oxidative stress and mitochondrial dysfunction.

 

Randle et al. first demonstrated that an elevation in FFA in the diaphragm and heart was associated with an increase in fatty acid oxidation and impaired glucose utilization (45). Via the Randle cycle effect, increased FFAs and fatty acid oxidation lead to increased intracellular glucose content and decreased glucose uptake (46). Studies in rodents and humans have demonstrated that conditions of increased FFA either via lipid infusions or secondary to T2DM lead to impaired glucose uptake and utilization in insulin sensitive tissues (47). This occurs secondary to the inhibition of the insulin signaling pathway.

 

As FFA levels increase, the capacity of the adipose tissue to take up and store FFAs can be exceeded. When this occurs, FFAs accumulate in tissues with limited ability for lipid storage, such as the liver and skeletal muscle. This phenomenon is known as ectopic fat deposition and is strongly associated with insulin resistance (48). Fatty acids accumulate in myocytes as fatty acid derivatives. Of these fatty acid derivatives, diacylglycerol (DAG), triacylglycerol, and ceramides directly correlate with insulin resistance. DAG interferes with normal insulin signaling by its interaction with a group of novel kinases, members of the protein kinase C family, that serine phosphorylate IRS, thereby impairing tyrosine phosphorylation and activation by insulin (41,48,49) Ceramide activates the enzyme protein phosphatase 2A, leading to dephosphorylation of AKT, thwarting insulin signaling and GLUT4 translocation to the cell membrane. This impairs insulin-mediated glucose uptake into the skeletal muscle (50).

 

FFAs increase inflammatory signaling pathways through direct interaction with members of the Toll-like receptor (TLR) family and indirectly through the secretion of cytokines, namely tumor necrosis factor-α (TNF-α), and interleukins (IL), IL-1β and IL-6 (49). TLR are the pathogen recognition receptors of the innate immune system that function to facilitate the detection of microbes and transmit inflammatory signaling (51,52). In vitro, FFA can signal through TLR-2 and TLR-4 on macrophages, thereby inducing pro-inflammatory gene expression (52,53). Studies in mice with a loss of function mutation of the TLR-4 receptor are protected from diet-induced obesity and saturated fatty acid-induced insulin resistance (54). Similarly, animal studies in which TLR-2 is either absent or inhibited, demonstrate a resolution of high fat diet induced insulin resistance (55,56). A recent study in humans corroborates the importance of TLR-2 and TLR-4 in the development of FFA induced insulin resistance. Jialal and colleagues studied individuals with and without MetS (according to the NCEP ATP III definition) and found that those with MetS had increased expression and activity of TLR-2 and TLR-4 (51).  TLR-4 activity leads to activation of c-Jun N- terminal kinase (JNK) and Iκβ kinase (IKK), which results in degradation of the inhibitor κβ (Iκβα) and activation of Nuclear Factor- κβ (NF- κβ).  Through JNK and IKK activation, FFA lead to Ser phosphorylation of IRS-1 and impaired insulin signaling (57,58). Ding and colleagues assessed 1628 Chinese adults and reported that levels of IL-6 and C-reactive protein were significantly associated with MetS (using the Harmonized definition) which also increased concurrent to the increased number of MetS components, further supporting that MetS is a pro-inflammatory state (59).

 

In obesity, adipose tissue infiltration by macrophages is increased. This leads to a pro-inflammatory state as macrophages produce TNF-α, IL-6 and IL-1β (60,61). Along with FFA signaling through TLR, these macrophage-derived inflammatory cytokines activate JNK and IKK to further interfere with insulin signaling and action (61). Additionally suppressor of cytokine signaling (SOCS) proteins are induced downstream of these inflammatory cytokines which terminate insulin signaling by promoting the ubiquitination and proteasomal degradation of IRS (62).

 

Reactive oxygen species (ROS) production increases with fat accumulation. FFAs activate ROS production by adipose tissue by stimulating NADPH oxidase and decreasing the expression of anti-oxidative enzymes (63). When adipose tissues is exposed to oxidative stress, there is a decrease in the anti-inflammatory adipokine, adiponectin (to be discussed in greater detail below) (64). In MetS, there is increased ROS production as a result of elevated levels of inflammatory cytokines and decreased levels of adiponectin (65). Increased levels of ROS lead to hindered insulin signaling by inducing IRS phosphorylation and impairing GLUT4 translocation and gene transcription (66).

 

It has been shown that there is a connection between mitochondrial dysfunction and insulin resistance in skeletal muscle that precedes the development of obesity and hyperglycemia. Animal studies demonstrate that mitochondrial number and function are intact, if not increased, under conditions of insulin resistance (67,68). On the other hand, studies in obese, insulin-resistant individuals as well as those with T2DM have skeletal muscle mitochondria that are fewer in size as well as number. It has also been shown that these individuals exhibit down-regulation of the genes involved in mitochondrial oxidative phosphorylation, the process by which mitochondria produce energy in the form of ATP (69-72). Studies demonstrate that PPARγ coactivator-1α (PGC-1α), a transcriptional activator involved in mitochondrial biosynthesis, has diminished expression in patients with T2DM, obesity, or a family history of T2DM (73,74). Increased FFA uptake and their incomplete oxidation have also been implicated in mediating mitochondrial dysfunction in the skeletal muscle under insulin resistant conditions (75). Furthermore, mitochondrial dysfunction leads to increased oxidative stress and the formation of ROS, which further diminishes mitochondrial mass and function.

 

As discussed above, increased FFAs in obesity and MetS are thought to lead to insulin resistance via several different mechanisms. These different mechanisms are not exclusive of one another and interact in such a way as to create a vicious cycle of insulin resistance.

 

Adipokines   

 

Adipose tissue is an active endocrine organ that releases adipokines, bioactive mediators that affect metabolism (76). It has been demonstrated that individuals with MetS have an abnormal adipokine profile that affects insulin sensitivity (77).

 

Adiponectin differs from other adipokines in that its level is inversely correlated with body adiposity and insulin resistance (78). The administration of recombinant adiponectin ameliorates insulin resistance in obese mice (78). Adiponectin transgenic mice demonstrate improvements in insulin sensitivity (79). Adiponectin increases insulin secretion in vivo and in vitro (80). In addition to its ability to improve insulin sensitivity in peripheral tissues, adiponectin has been shown to have effects on the central nervous system that affect food intake and energy expenditure (81). In humans, low levels of adiponectin have been strongly associated with insulin resistance, increased body adiposity, T2DM, and MetS (76). Genetic hypoadiponectinemia caused by a missense mutation leads to an increased propensity toward MetS (82). Longitudinal studies demonstrate that in individuals at high risk for developing T2DM, those with higher levels of adiponectin were less likely to develop T2DM than those with lower levels of adiponectin (83) . Adiponectin levels have even been proposed to be used as a cut-off for managing the risk of developing MetS in a study of male Japanese workers.  In a 3-year prospective cohort study, the risk of developing MetS, calculated by the accelerated failure-time model, demonstrated that the mean time to develop MetS declined with decreasing total adiponectin levels.

 

Adiponectin modulates glucose metabolism through its interaction with its receptors, the adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2 (AdipoR2). Binding of adiponectin to AdipoR1 and AdipoR2 results in the activation of signaling pathways affecting glucose and fatty acid metabolism. As a result of adiponectin signaling, AMP-activated protein kinase (AMPK) is phosphorylated, leading to increased glucose uptake in the muscle and reduced gluconeogenesis (84). Adiponectin also has anti-inflammatory actions, suppressing TNF-α and IL-6 expression and anti-atherogenic effects, decreasing levels of pro-atherogenic small, dense low-density lipoprotein (LDL) and TG levels (76,85).

 

In patients with insulin resistance, there is reduced responsiveness of the skeletal muscle, liver and adipose tissue to insulin. Insulin levels rise in an attempt to maintain euglycemia, and the result is hyperinsulinemia. Hyperinsulinemia has been shown to down-regulate the bioactive high-molecular weight form of adiponectin (86). Thus, the hyperinsulinemia in insulin resistance may decrease adiponectin further contributing to insulin resistance (77). Aside from the direct effects of insulin, changes that characterize the metabolic milieu of insulin resistance such as inflammation, oxidative stress and mitochondrial dysfunction have been shown to suppress adiponectin (77). This relationship is observed clinically in the same study by Ding and colleagues, showing a strong inverse association between adiponectin and HOMA-IR and an inverse trend between adiponectin and an increased number of MetS components (59). Hence, the association between insulin resistance and adiponectin appears to be complex and bidirectional. Further studies are necessary to better define this complicated relationship.

 

Leptin, another important adipokine produced by adipocytes, exerts effects on appetite and energy expenditure. When leptin binds to its receptor, signaling pathways such as the Janus Kinase-Signal Transducers and Activation of Transcription (JAK/STAT) and IRS/PI3K are activated. The result is similar to what is observed when insulin binds the IR, in that anorexigenic pathways (involving POMC) are favored over orexigenic pathways (involving neuropeptides NPY and AgRP) (87). Studies suggest that leptin affects glucose metabolism independently of its effects on food intake. Studies in rodents suggest that leptin stimulated JAK/STAT signaling is important in food intake and energy expenditure while leptin mediated PI3K signaling plays a role in regulating glucose metabolism (88-90).

 

Leptin also stimulates FFA oxidation in the liver, pancreas and skeletal muscle. Leptin opposes the action of insulin by decreasing insulin’s lipogenic effect on the adipocyte and depleting the triglyceride content of adipose tissue without increasing circulating FFA (91-93). Separate from its effects on lipid and glucose metabolism, leptin affects the immune system, by enhancing the production of inflammatory cytokines and by stimulating T–cell proliferation (94).

 

While the absence of leptin leads to extreme obesity and insulin resistance, most obese individuals are not leptin deficient. Rather, they have increased levels of leptin but are immune to its appetite suppressant effects. This observation has given rise to the concept of leptin resistance in obesity (95). Similarly, elevated leptin levels have been observed in different populations with metabolic syndrome -(96-98). Decreased sensitivity to leptin leads to increased triglyceride accumulation in adipose tissue, muscle, liver and pancreas, resulting in insulin resistance (76). An alternative perspective is the concept of hypothalamic leptin insufficiency, which states that in conditions of hyperleptinemia, the blood brain barrier prevents entry of leptin into the brain resulting in insufficiencies of leptin at important sites in the CNS (99). Regardless of whether the decreased responsiveness to leptin observed in obesity is due to leptin resistance or hypothalamic leptin insufficiency, the ability of leptin to activate hypothalamic signaling is decreased in obesity and insulin resistance (99).

 

The role of resistin in MetS is not entirely understood. Resistin is an adipokine that has been seen to be increased in rodent models of obesity, leading to impaired insulin action and β-cell dysfunction (100). Resistin is highly associated with insulin resistance and T2DM in animal models (101). Resistin activates SOCS-3, which inhibits IR phosphorylation and downstream signaling proteins, leading to impaired insulin signaling (102). It also inhibits glucose uptake by skeletal muscle and the liver and enhances hepatic gluconeogenesis (101,103).  In humans, the relationship of resistin, MetS and its components are not as clear, however associations between the components of MetS have driven an interest in further understanding its potential role.  Resistin expression in humans differs from rodents in its low expression in white adipose tissue and regulation of concentration by peripheral blood mononuclear cells, macrophages and bone marrow cells (104). Its role in the inflammatory pathway has been well described, associated with upregulation of inflammatory cytokines and to induce monocyte-endothelial cell adhesions [127]. However, the role of resistin in insulin resistance has been controversial. (76). Increased resistin levels have been demonstrated in several studies with individuals with MetS but correlations have been more consistent in women than in men (105-107). Hence, more studies are necessary to better determine the role of resistin in MetS.

 

Retinol Binding Protein-4 (RBP-4) is the vitamin A (retinol) transporter and is secreted from both adipose tissue and liver.  RBP-4 has been shown to be increased in the adipose tissue of mice with an adipose-specific knockout of GLUT4 (108). RBP-4 levels are also elevated in humans with obesity, T2DM, impaired glucose intolerance and those with a strong family history of T2DM (109,110). The suggested mechanisms by which RBP4 can mediate insulin resistance include increased gluconeogenesis and impaired insulin action in the liver and muscle (108). However, there are other studies that do not support the relationship of RBP-4 with altered glucose metabolism (111,112). As with all other adipokines, further exploration is necessary to better define the role of RBP-4 in insulin resistance and MetS.

 

Apelin, omentin and visfatin are other adipokines have been implicated in the pathogenesis of insulin resistance and MetS. However further study is necessary to better define the part they play in this process. Individuals with insulin resistance and MetS exhibit atypical adipokine profiles that not only result from insulin resistance but further contribute to its development and pathogenesis.

 

Though there are many different factors that contribute to the development of MetS. Insulin resistance, via augmented FFA levels and irregular adipokine patterns, is largely responsible for the pathogenesis of the syndrome.

 

Gut Microbiota

 

There has been considerable interest in the gut microbiota and its relationship with inflammation and metabolism.  With limited ability to digest polysaccharides, the gut microbiota in mammals represents an important system significant influence on energy harvest and efficiency(113-115).

 

In fact, mice raised in a germ-free environment, compared to conventionally raised mice, had lower body fat content and, following colonization with intestinal flora, there was an increase in body fat and hepatic triglyceride synthesis as well as the development of insulin resistance, independent of food intake and energy expenditure (113). Beyond alterations in energy harvest, the gut microbiota composition can also drive low level inflammation which has also been found to be a contributor to obesity and the metabolic syndrome (116,117). Interventional experiments with Roux en Y gastric bypass versus sham surgeries with subsequent microbiota transplant have further underscored the relationship of the microbiota with obesity (118). Observational human studies have noted differences in the microbial diversity in lean and obese subjects as well as in those with differences in microbial diversity based upon diet composition (119-121). Similar differences in microbiota composition have been seen in those with and without type 2 diabetes mellitus (122). Furthermore, infusion of microbiota via gastrointestinal probe have demonstrated alterations in insulin sensitivity (123).

 

The metabolic syndrome is a product of the complex intertwining of inflammation and insulin resistance; with its relationship to both of these, the gut microbiota has been demonstrated to have a strong influence on metabolic diseases. From observational to experimental data, the microbiota not only offers important insight into pathophysiology but also has the potential as a therapeutic target.

 

TREATMENT

 

Lifestyle modification is the foundational intervention in treatment of MetS. The Diabetes Prevention Program demonstrated that lifestyle intervention reduced the incidence of MetS by 41% compared with placebo. The intensive lifestyle intervention involved a healthy low-calorie, low fat diet and moderate physical activity of at least 150 minutes/week, resulting in a weight reduction of 7% (124). The recommended diet should include < 200 mg/day of cholesterol, < 7% saturated fat, with total fat comprising 25-35% of calories, low simple sugars and increased fruits, vegetables and whole grains (12). Smoking cessation should be instituted in all patients with MetS. Additionally, low dose aspirin is recommended in cases of moderate to high cardiovascular risk where no contraindication to aspirin therapy exists (12). For those patients in whom lifestyle intervention is not sufficient to treat their MetS, pharmacotherapy for the treatment of many of the components of MetS is available.

 

Historically, many of the medications aimed to treat obesity have failed to gain approval or have been removed from the market by the FDA due to side effects and marginal success in weight reduction (125). However, in the last decade, an increasing number of pharmacotherapies have become FDA approved.  Currently available FDA-approved pharmacotherapy for obesity includes phentermine, orlistat, phentermine/topiramate, locaserin, buproprion/naltrexone and liraglutide 3.0 mg.  Further, individuals with morbid obesity (BMI> 40 kg/m2 or >35 kg/m2 with comorbidities) may be candidates for bariatric surgery (126). Bariatric surgery has been demonstrated to be an effective treatment of obesity with improvements in weight, T2DM, hypertension, hyperlipidemia, and sleep apnea.  Resolution rates of each component reported in the literature are variable, the type of surgery highly influential on the resolution of comorbidities (127). Some studies demonstrate superiority of surgical to nonsurgical treatment in weight loss and MetS (128).     

 

There are no pharmacologic agents specifically approved for prediabetes or the prevention of T2DM. In the Diabetes Prevention Program, metformin was shown to lead to weight loss and a 31% decrease in the incidence of T2DM compared to patients receiving placebo (124). It has been suggested that GLP-1 receptor agonists, agents now commonly used in the treatment of established T2DM, may have a role in prevention of T2DM, but more studies are needed. The American Diabetes Association recommends lifestyle modification over medication for the prevention of diabetes. However, they state that metformin therapy may be considered for the prevention of T2DM in individuals with IGT, IFG or HgA1c 5.7-6.4%, especially for those individuals with BMI> 35 kg/m2, those aged < 60 years, and those with a prior diagnosis of GDM (129,130).

 

Elevated blood pressure is first approached with lifestyle modification. If this fails to bring the blood pressure to goal range <140/90 or <130/80 in patients with diabetes or CKD, medication should be added. First line medications include thiazide diuretics in uncomplicated individuals, angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) in those with diabetes, congestive heart failure or CKD, or beta blockers in individuals with angina (131).

 

Drug therapy for dyslipidemia is generally approached with the use of HMG Co-A reductase inhibitors (statins). The primary objective in CVD risk reduction is to lower LDL-C values and the drug of choice for this purposes is statins, which have been shown not only to lower LDL-C, but also to modestly raise HDL-C and lower triglycerides (132). The second targets in lipid improvement to reduce CVD risk are HDL-C and triglycerides. Niacin is effective at raising HDL-C as well as lowering triglycerides and LDL-C. Fibrates are effective at lowering triglycerides but do not have the beneficial effects on HDL-C and LDL-C. Omega-3 polyunsaturated fatty acids (n-3 PUFA) in fish oil can also be used to lower triglycerides with recent data from the REDUCE-IT trial demonstrating a reduced risk of ischemic events in patients with elevated triglycerides despite statin therapy receiving icosapent ethyl.  

 

CONCLUSION

 

The metabolic syndrome is a collection of related risk factors that predispose an individual to the development of T2DM and CVD. It affects a large number of people worldwide and its prevalence is increasing. The diagnostic criteria for MetS have been harmonized for the purpose of providing more consistency in clinical care and research of patients with MetS. Insulin resistance remains at the core of the syndrome, as it did when it was first introduced by Reaven in 1988, and appears to contribute to the development of MetS, via elevated FFA levels and abnormal adipokine profiles. Insulin resistance has both metabolic and mitogenic effects and can result in the development of hyperglycemia and T2DM, hypertension, dyslipidemia, NAFLD, PCOS, OSA, sexual dysfunction, and cancer. In patients with MetS, lifestyle modification is imperative in decreasing the risk of CVD and treating many of the associated conditions. Treatment of the individual conditions is often also required.

 

REFERENCES

 

  1. Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 1988;37(12):1595-1607.
  2. Reaven GM. Role of insulin resistance in human disease (syndrome X): an expanded definition. Annu Rev Med. 1993;44:121-131.
  3. Bogardus C, Lillioja S, Mott DM, Hollenbeck C, Reaven G. Relationship between degree of obesity and in vivo insulin action in man. Am J Physiol. 1985;248(3 Pt 1):E286-291.
  4. Reaven GM. Banting Lecture 1988. Role of insulin resistance in human disease. 1988. Nutrition. 1997;13(1):65; discussion 64, 66.
  5. 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. 1998;15(7):539-553.
  6. Balkau B, Charles MA. Comment on the provisional report from the WHO consultation. European Group for the Study of Insulin Resistance (EGIR). Diabet Med. 1999;16(5):442-443.
  7. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA. 2001;285(19):2486-2497.
  8. Eisenmann JC. On the use of a continuous metabolic syndrome score in pediatric research. Cardiovasc Diabetol. 2008;7:17.
  9. Einhorn D, Reaven GM, Cobin RH, Ford E, Ganda OP, Handelsman Y, Hellman R, Jellinger PS, Kendall D, Krauss RM, Neufeld ND, Petak SM, Rodbard HW, Seibel JA, Smith DA, Wilson PW. American College of Endocrinology position statement on the insulin resistance syndrome. Endocr Pract. 2003;9(3):237-252.
  10. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome--a new worldwide definition. Lancet. 2005;366(9491):1059-1062.
  11. Report of the expert committee on the diagnosis and classification of diabetes mellitus. Diabetes Care. 2003;26 Suppl 1:S5-20.
  12. Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, Smith SC, Jr., Spertus JA, Costa F. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation. 2005;112(17):2735-2752.
  13. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA, Fruchart JC, James WP, Loria CM, Smith SC, Jr. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation. 2009;120(16):1640-1645.
  14. Ohlson LO, Larsson B, Svardsudd K, Welin L, Eriksson H, Wilhelmsen L, Bjorntorp P, Tibblin G. The influence of body fat distribution on the incidence of diabetes mellitus. 13.5 years of follow-up of the participants in the study of men born in 1913. Diabetes. 1985;34(10):1055-1058.
  15. Yusuf S, Hawken S, Ounpuu S, Bautista L, Franzosi MG, Commerford P, Lang CC, Rumboldt Z, Onen CL, Lisheng L, Tanomsup S, Wangai P, Jr., Razak F, Sharma AM, Anand SS. Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study. Lancet. 2005;366(9497):1640-1649.
  16. Ashwell M, Gunn P, Gibson S. Waist-to-height ratio is a better screening tool than waist circumference and BMI for adult cardiometabolic risk factors: systematic review and meta-analysis. Obes Rev. 2012;13(3):275-286.
  17. Swainson MG, Batterham AM, Tsakirides C, Rutherford ZH, Hind K. Prediction of whole-body fat percentage and visceral adipose tissue mass from five anthropometric variables. PLoS One. 2017;12(5):e0177175.
  18. Lear SA, Humphries KH, Kohli S, Birmingham CL. The use of BMI and waist circumference as surrogates of body fat differs by ethnicity. Obesity (Silver Spring). 2007;15(11):2817-2824.
  19. Pajunen P, Rissanen H, Harkanen T, Jula A, Reunanen A, Salomaa V. The metabolic syndrome as a predictor of incident diabetes and cardiovascular events in the Health 2000 Study. Diabetes Metab. 2010;36(5):395-401.
  20. Gale EA. Should we dump the metabolic syndrome? Yes. BMJ. 2008;336(7645):640.
  21. Kahn R. Metabolic syndrome--what is the clinical usefulness? Lancet. 2008;371(9628):1892-1893.
  22. Beltran-Sanchez H, Harhay MO, Harhay MM, McElligott S. Prevalence and trends of metabolic syndrome in the adult U.S. population, 1999-2010. J Am Coll Cardiol. 2013;62(8):697-703.
  23. Simmons RK, Alberti KG, Gale EA, Colagiuri S, Tuomilehto J, Qiao Q, Ramachandran A, Tajima N, Brajkovich Mirchov I, Ben-Nakhi A, Reaven G, Hama Sambo B, Mendis S, Roglic G. The metabolic syndrome: useful concept or clinical tool? Report of a WHO Expert Consultation. Diabetologia. 2010;53(4):600-605.
  24. Aguilar M, Bhuket T, Torres S, Liu B, Wong RJ. Prevalence of the metabolic syndrome in the United States, 2003-2012. JAMA. 2015;313(19):1973-1974.
  25. Cornier MA, Dabelea D, Hernandez TL, Lindstrom RC, Steig AJ, Stob NR, Van Pelt RE, Wang H, Eckel RH. The metabolic syndrome. Endocr Rev. 2008;29(7):777-822.
  26. Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Metabolic syndrome: definitions and controversies. BMC Med. 2011;9:48.
  27. Cameron AJ, Shaw JE, Zimmet PZ. The metabolic syndrome: prevalence in worldwide populations. Endocrinol Metab Clin North Am. 2004;33(2):351-375, table of contents.
  28. Ford ES, Li C, Zhao G. Prevalence and correlates of metabolic syndrome based on a harmonious definition among adults in the US. J Diabetes. 2010;2(3):180-193.
  29. Araneta MR, Wingard DL, Barrett-Connor E. Type 2 diabetes and metabolic syndrome in Filipina-American women: a high-risk nonobese population. Diabetes Care. 2002;25(3):494-499.
  30. Maggi S, Noale M, Gallina P, Bianchi D, Marzari C, Limongi F, Crepaldi G. Metabolic syndrome, diabetes, and cardiovascular disease in an elderly Caucasian cohort: the Italian Longitudinal Study on Aging. J Gerontol A Biol Sci Med Sci. 2006;61(5):505-510.
  31. Mancia G, Bombelli M, Corrao G, Facchetti R, Madotto F, Giannattasio C, Trevano FQ, Grassi G, Zanchetti A, Sega R. Metabolic syndrome in the Pressioni Arteriose Monitorate E Loro Associazioni (PAMELA) study: daily life blood pressure, cardiac damage, and prognosis. Hypertension. 2007;49(1):40-47.
  32. Simmons D, Thompson CF. Prevalence of the metabolic syndrome among adult New Zealanders of Polynesian and European descent. Diabetes Care. 2004;27(12):3002-3004.
  33. Welty TK, Lee ET, Yeh J, Cowan LD, Go O, Fabsitz RR, Le NA, Oopik AJ, Robbins DC, Howard BV. Cardiovascular disease risk factors among American Indians. The Strong Heart Study. Am J Epidemiol. 1995;142(3):269-287.
  34. Feng Y, Hong X, Li Z, Zhang W, Jin D, Liu X, Zhang Y, Hu FB, Wei LJ, Zang T, Xu X. Prevalence of metabolic syndrome and its relation to body composition in a Chinese rural population. Obesity (Silver Spring). 2006;14(11):2089-2098.
  35. Lorenzo C, Serrano-Rios M, Martinez-Larrad MT, Gonzalez-Sanchez JL, Seclen S, Villena A, Gonzalez-Villalpando C, Williams K, Haffner SM. Geographic variations of the International Diabetes Federation and the National Cholesterol Education Program-Adult Treatment Panel III definitions of the metabolic syndrome in nondiabetic subjects. Diabetes Care. 2006;29(3):685-691.
  36. Azimi-Nezhad M, Herbeth B, Siest G, Dade S, Ndiaye NC, Esmaily H, Hosseini SJ, Ghayour-Mobarhan M, Visvikis-Siest S. High Prevalence of Metabolic Syndrome in Iran in Comparison with France: What Are the Components That Explain This? Metab Syndr Relat Disord. 2012.
  37. Pandit K, Goswami S, Ghosh S, Mukhopadhyay P, Chowdhury S. Metabolic syndrome in South Asians. Indian J Endocrinol Metab. 2012;16(1):44-55.
  38. McKeigue PM, Ferrie JE, Pierpoint T, Marmot MG. Association of early-onset coronary heart disease in South Asian men with glucose intolerance and hyperinsulinemia. Circulation. 1993;87(1):152-161.
  39. Thomas GN, Ho SY, Janus ED, Lam KS, Hedley AJ, Lam TH. The US National Cholesterol Education Programme Adult Treatment Panel III (NCEP ATP III) prevalence of the metabolic syndrome in a Chinese population. Diabetes Res Clin Pract. 2005;67(3):251-257.
  40. Smith DO, LeRoith D. Insulin resistance syndrome, pre-diabetes, and the prevention of type 2 diabetes mellitus. Clin Cornerstone. 2004;6(2):7-6.
  41. Gallagher EJ, Leroith D, Karnieli E. The metabolic syndrome--from insulin resistance to obesity and diabetes. Med Clin North Am. 2011;95(5):855-873.
  42. Karnieli E, Zarnowski MJ, Hissin PJ, Simpson IA, Salans LB, Cushman SW. Insulin-stimulated translocation of glucose transport systems in the isolated rat adipose cell. Time course, reversal, insulin concentration dependency, and relationship to glucose transport activity. J Biol Chem. 1981;256(10):4772-4777.
  43. Najjar SM, Yang Y, Fernstrom MA, Lee SJ, Deangelis AM, Rjaily GA, Al-Share QY, Dai T, Miller TA, Ratnam S, Ruch RJ, Smith S, Lin SH, Beauchemin N, Oyarce AM. Insulin acutely decreases hepatic fatty acid synthase activity. Cell Metab. 2005;2(1):43-53.
  44. Karpe F, Dickmann JR, Frayn KN. Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes. 2011;60(10):2441-2449.
  45. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1(7285):785-789.
  46. Randle PJ. Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev. 1998;14(4):263-283.
  47. Boden G, Jadali F, White J, Liang Y, Mozzoli M, Chen X, Coleman E, Smith C. Effects of fat on insulin-stimulated carbohydrate metabolism in normal men. J Clin Invest. 1991;88(3):960-966.
  48. Yki-Jarvinen H. Ectopic fat accumulation: an important cause of insulin resistance in humans. J R Soc Med. 2002;95 Suppl 42:39-45.
  49. Martins AR, Nachbar RT, Gorjao R, Vinolo MA, Festuccia WT, Lambertucci RH, Cury-Boaventura MF, Silveira LR, Curi R, Hirabara SM. Mechanisms underlying skeletal muscle insulin resistance induced by fatty acids: importance of the mitochondrial function. Lipids Health Dis. 2012;11:30.
  50. Summers SA. Sphingolipids and insulin resistance: the five Ws. Curr Opin Lipidol. 2010;21(2):128-135.
  51. Jialal I, Huet BA, Kaur H, Chien A, Devaraj S. Increased toll-like receptor activity in patients with metabolic syndrome. Diabetes Care. 2012;35(4):900-904.
  52. Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA, Jenn A, Liu-Bryan R, Glass CK, Neels JG, Olefsky JM. 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. 2007;282(48):35279-35292.
  53. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116(11):3015-3025.
  54. Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, Prada PO, Hirabara SM, Schenka AA, Araujo EP, Vassallo J, Curi R, Velloso LA, Saad MJ. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes. 2007;56(8):1986-1998.
  55. Caricilli AM, Nascimento PH, Pauli JR, Tsukumo DM, Velloso LA, Carvalheira JB, Saad MJ. Inhibition of toll-like receptor 2 expression improves insulin sensitivity and signaling in muscle and white adipose tissue of mice fed a high-fat diet. J Endocrinol. 2008;199(3):399-406.
  56. Ehses JA, Meier DT, Wueest S, Rytka J, Boller S, Wielinga PY, Schraenen A, Lemaire K, Debray S, Van Lommel L, Pospisilik JA, Tschopp O, Schultze SM, Malipiero U, Esterbauer H, Ellingsgaard H, Rutti S, Schuit FC, Lutz TA, Boni-Schnetzler M, Konrad D, Donath MY. Toll-like receptor 2-deficient mice are protected from insulin resistance and beta cell dysfunction induced by a high-fat diet. Diabetologia. 2010;53(8):1795-1806.
  57. Bhargava P, Lee CH. Role and function of macrophages in the metabolic syndrome. Biochem J. 2012;442(2):253-262.
  58. Hotamisligil GS. Inflammation and endoplasmic reticulum stress in obesity and diabetes. Int J Obes (Lond). 2008;32 Suppl 7:S52-54.
  59. Ding Y, Li S, Ma RL, Guo H, Zhang J, Zhang M, Liu J, Guo S. Association of homeostasis model assessment of insulin resistance, adiponectin, and low-grade inflammation with the course of the metabolic syndrome. Clin Biochem. 2015.
  60. 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. 2003;112(12):1796-1808.
  61. Harford KA, Reynolds CM, McGillicuddy FC, Roche HM. Fats, inflammation and insulin resistance: insights to the role of macrophage and T-cell accumulation in adipose tissue. Proc Nutr Soc. 2011;70(4):408-417.
  62. Lebrun P, Van Obberghen E. SOCS proteins causing trouble in insulin action. Acta Physiol (Oxf). 2008;192(1):29-36.
  63. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114(12):1752-1761.
  64. Soares AF, Guichardant M, Cozzone D, Bernoud-Hubac N, Bouzaidi-Tiali N, Lagarde M, Geloen A. Effects of oxidative stress on adiponectin secretion and lactate production in 3T3-L1 adipocytes. Free Radic Biol Med. 2005;38(7):882-889.
  65. Otani H. Oxidative stress as pathogenesis of cardiovascular risk associated with metabolic syndrome. Antioxid Redox Signal. 2011;15(7):1911-1926.
  66. Bloch-Damti A, Bashan N. Proposed mechanisms for the induction of insulin resistance by oxidative stress. Antioxid Redox Signal. 2005;7(11-12):1553-1567.
  67. Hancock CR, Han DH, Chen M, Terada S, Yasuda T, Wright DC, Holloszy JO. High-fat diets cause insulin resistance despite an increase in muscle mitochondria. Proc Natl Acad Sci U S A. 2008;105(22):7815-7820.
  68. Holloway GP, Gurd BJ, Snook LA, Lally J, Bonen A. Compensatory increases in nuclear PGC1alpha protein are primarily associated with subsarcolemmal mitochondrial adaptations in ZDF rats. Diabetes. 2010;59(4):819-828.
  69. Hojlund K, Mogensen M, Sahlin K, Beck-Nielsen H. Mitochondrial dysfunction in type 2 diabetes and obesity. Endocrinol Metab Clin North Am. 2008;37(3):713-731, x.
  70. Kim JA, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance. Circ Res. 2008;102(4):401-414.
  71. Chomentowski P, Coen PM, Radikova Z, Goodpaster BH, Toledo FG. Skeletal muscle mitochondria in insulin resistance: differences in intermyofibrillar versus subsarcolemmal subpopulations and relationship to metabolic flexibility. J Clin Endocrinol Metab. 2011;96(2):494-503.
  72. Coletta DK, Mandarino LJ. Mitochondrial dysfunction and insulin resistance from the outside in: extracellular matrix, the cytoskeleton, and mitochondria. Am J Physiol Endocrinol Metab. 2011;301(5):E749-755.
  73. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldfine AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, Mandarino LJ. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1. Proc Natl Acad Sci U S A. 2003;100(14):8466-8471.
  74. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstrale M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34(3):267-273.
  75. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, Muoio DM. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45-56.
  76. Rabe K, Lehrke M, Parhofer KG, Broedl UC. Adipokines and insulin resistance. Mol Med. 2008;14(11-12):741-751.
  77. Deng Y, Scherer PE. Adipokines as novel biomarkers and regulators of the metabolic syndrome. Ann N Y Acad Sci. 2010;1212:E1-E19.
  78. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med. 2001;7(8):941-946.
  79. Yamauchi T, Kamon J, Waki H, Imai Y, Shimozawa N, Hioki K, Uchida S, Ito Y, Takakuwa K, Matsui J, Takata M, Eto K, Terauchi Y, Komeda K, Tsunoda M, Murakami K, Ohnishi Y, Naitoh T, Yamamura K, Ueyama Y, Froguel P, Kimura S, Nagai R, Kadowaki T. Globular adiponectin protected ob/ob mice from diabetes and ApoE-deficient mice from atherosclerosis. J Biol Chem. 2003;278(4):2461-2468.
  80. Okamoto M, Ohara-Imaizumi M, Kubota N, Hashimoto S, Eto K, Kanno T, Kubota T, Wakui M, Nagai R, Noda M, Nagamatsu S, Kadowaki T. Adiponectin induces insulin secretion in vitro and in vivo at a low glucose concentration. Diabetologia. 2008;51(5):827-835.
  81. Kubota N, Yano W, Kubota T, Yamauchi T, Itoh S, Kumagai H, Kozono H, Takamoto I, Okamoto S, Shiuchi T, Suzuki R, Satoh H, Tsuchida A, Moroi M, Sugi K, Noda T, Ebinuma H, Ueta Y, Kondo T, Araki E, Ezaki O, Nagai R, Tobe K, Terauchi Y, Ueki K, Minokoshi Y, Kadowaki T. Adiponectin stimulates AMP-activated protein kinase in the hypothalamus and increases food intake. Cell Metab. 2007;6(1):55-68.
  82. Kondo H, Shimomura I, Matsukawa Y, Kumada M, Takahashi M, Matsuda M, Ouchi N, Kihara S, Kawamoto T, Sumitsuji S, Funahashi T, Matsuzawa Y. Association of adiponectin mutation with type 2 diabetes: a candidate gene for the insulin resistance syndrome. Diabetes. 2002;51(7):2325-2328.
  83. Lindsay RS, Funahashi T, Hanson RL, Matsuzawa Y, Tanaka S, Tataranni PA, Knowler WC, Krakoff J. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet. 2002;360(9326):57-58.
  84. Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, Okada-Iwabu M, Kawamoto S, Kubota N, Kubota T, Ito Y, Kamon J, Tsuchida A, Kumagai K, Kozono H, Hada Y, Ogata H, Tokuyama K, Tsunoda M, Ide T, Murakami K, Awazawa M, Takamoto I, Froguel P, Hara K, Tobe K, Nagai R, Ueki K, Kadowaki T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med. 2007;13(3):332-339.
  85. Gallagher EJ, Leroith D, Karnieli E. Insulin resistance in obesity as the underlying cause for the metabolic syndrome. Mt Sinai J Med. 2010;77(5):511-523.
  86. Basu R, Pajvani UB, Rizza RA, Scherer PE. Selective downregulation of the high molecular weight form of adiponectin in hyperinsulinemia and in type 2 diabetes: differential regulation from nondiabetic subjects. Diabetes. 2007;56(8):2174-2177.
  87. Prodi E, Obici S. Minireview: the brain as a molecular target for diabetic therapy. Endocrinology. 2006;147(6):2664-2669.
  88. Bates SH, Kulkarni RN, Seifert M, Myers MG, Jr. Roles for leptin receptor/STAT3-dependent and -independent signals in the regulation of glucose homeostasis. Cell Metab. 2005;1(3):169-178.
  89. Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, Tang V, Liu SM, Ludwig T, Chua SC, Jr., Lowell BB, Elmquist JK. The hypothalamic arcuate nucleus: a key site for mediating leptin's effects on glucose homeostasis and locomotor activity. Cell Metab. 2005;1(1):63-72.
  90. Morton GJ, Gelling RW, Niswender KD, Morrison CD, Rhodes CJ, Schwartz MW. Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons. Cell Metab. 2005;2(6):411-420.
  91. Buettner C, Muse ED, Cheng A, Chen L, Scherer T, Pocai A, Su K, Cheng B, Li X, Harvey-White J, Schwartz GJ, Kunos G, Rossetti L. Leptin controls adipose tissue lipogenesis via central, STAT3-independent mechanisms. Nat Med. 2008;14(6):667-675.
  92. Lago F, Gomez R, Gomez-Reino JJ, Dieguez C, Gualillo O. Adipokines as novel modulators of lipid metabolism. Trends Biochem Sci. 2009;34(10):500-510.
  93. Shimabukuro M, Koyama K, Chen G, Wang MY, Trieu F, Lee Y, Newgard CB, Unger RH. Direct antidiabetic effect of leptin through triglyceride depletion of tissues. Proc Natl Acad Sci U S A. 1997;94(9):4637-4641.
  94. Rasouli N, Kern PA. Adipocytokines and the metabolic complications of obesity. J Clin Endocrinol Metab. 2008;93(11 Suppl 1):S64-73.
  95. Munzberg H, Myers MG, Jr. Molecular and anatomical determinants of central leptin resistance. Nat Neurosci. 2005;8(5):566-570.
  96. Yun JE, Kimm H, Jo J, Jee SH. Serum leptin is associated with metabolic syndrome in obese and nonobese Korean populations. Metabolism. 2010;59(3):424-429.
  97. Lee SW, Jo HH, Kim MR, You YO, Kim JH. Association between metabolic syndrome and serum leptin levels in postmenopausal women. J Obstet Gynaecol. 2012;32(1):73-77.
  98. Yoshinaga M, Sameshima K, Tanaka Y, Wada A, Hashiguchi J, Tahara H, Kono Y. Adipokines and the prediction of the accumulation of cardiovascular risk factors or the presence of metabolic syndrome in elementary school children. Circ J. 2008;72(11):1874-1878.
  99. Kalra SP. Central leptin insufficiency syndrome: an interactive etiology for obesity, metabolic and neural diseases and for designing new therapeutic interventions. Peptides. 2008;29(1):127-138.
  100. Gao CL, Zhao DY, Qiu J, Zhang CM, Ji CB, Chen XH, Liu F, Guo XR. Resistin induces rat insulinoma cell RINm5F apoptosis. Mol Biol Rep. 2009;36(7):1703-1708.
  101. Yang Y, Xiao M, Mao Y, Li H, Zhao S, Gu Y, Wang R, Yu J, Zhang X, Irwin DM, Niu G, Tan H. Resistin and insulin resistance in hepatocytes: resistin disturbs glycogen metabolism at the protein level. Biomed Pharmacother. 2009;63(5):366-374.
  102. Steppan CM, Wang J, Whiteman EL, Birnbaum MJ, Lazar MA. Activation of SOCS-3 by resistin. Mol Cell Biol. 2005;25(4):1569-1575.
  103. Banerjee RR, Rangwala SM, Shapiro JS, Rich AS, Rhoades B, Qi Y, Wang J, Rajala MW, Pocai A, Scherer PE, Steppan CM, Ahima RS, Obici S, Rossetti L, Lazar MA. Regulation of fasted blood glucose by resistin. Science. 2004;303(5661):1195-1198.
  104. Abate N, Sallam HS, Rizzo M, Nikolic D, Obradovic M, Bjelogrlic P, Isenovic ER. Resistin: an inflammatory cytokine. Role in cardiovascular diseases, diabetes and the metabolic syndrome. Curr Pharm Des. 2014;20(31):4961-4969.
  105. Malo E, Ukkola O, Jokela M, Moilanen L, Kahonen M, Nieminen MS, Salomaa V, Jula A, Kesaniemi YA. Resistin is an indicator of the metabolic syndrome according to five different definitions in the Finnish Health 2000 survey. Metab Syndr Relat Disord. 2011;9(3):203-210.
  106. Norata GD, Ongari M, Garlaschelli K, Raselli S, Grigore L, Catapano AL. Plasma resistin levels correlate with determinants of the metabolic syndrome. Eur J Endocrinol. 2007;156(2):279-284.
  107. Singh AK, Tiwari S, Gupta A, Natu SM, Mittal B, Pant AB. Association of Resistin with Metabolic Syndrome in Indian Subjects. Metab Syndr Relat Disord. 2012.
  108. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, Kotani K, Quadro L, Kahn BB. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature. 2005;436(7049):356-362.
  109. Graham TE, Yang Q, Bluher M, Hammarstedt A, Ciaraldi TP, Henry RR, Wason CJ, Oberbach A, Jansson PA, Smith U, Kahn BB. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N Engl J Med. 2006;354(24):2552-2563.
  110. Meisinger C, Ruckert IM, Rathmann W, Doring A, Thorand B, Huth C, Kowall B, Koenig W. Retinol-binding protein 4 is associated with prediabetes in adults from the general population: the Cooperative Health Research in the Region of Augsburg (KORA) F4 Study. Diabetes Care. 2011;34(7):1648-1650.
  111. Janke J, Engeli S, Boschmann M, Adams F, Bohnke J, Luft FC, Sharma AM, Jordan J. Retinol-binding protein 4 in human obesity. Diabetes. 2006;55(10):2805-2810.
  112. Promintzer M, Krebs M, Todoric J, Luger A, Bischof MG, Nowotny P, Wagner O, Esterbauer H, Anderwald C. Insulin resistance is unrelated to circulating retinol binding protein and protein C inhibitor. J Clin Endocrinol Metab. 2007;92(11):4306-4312.
  113. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101(44):15718-15723.
  114. Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance and obesity. J Physiol. 2009;587(Pt 17):4153-4158.
  115. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031.
  116. Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, Sitaraman SV, Knight R, Ley RE, Gewirtz AT. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328(5975):228-231.
  117. Chassaing B, Gewirtz AT. Gut microbiota, low-grade inflammation, and metabolic syndrome. Toxicol Pathol. 2014;42(1):49-53.
  118. Liou AP, Paziuk M, Luevano JM, Jr., Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5(178):178ra141.
  119. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107(33):14691-14696.
  120. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480-484.
  121. Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C, Hardt PD. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 2010;18(1):190-195.
  122. Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, Al-Soud WA, Sorensen SJ, Hansen LH, Jakobsen M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5(2):e9085.
  123. Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS, Bartelsman JF, Dallinga-Thie GM, Ackermans MT, Serlie MJ, Oozeer R, Derrien M, Druesne A, Van Hylckama Vlieg JE, Bloks VW, Groen AK, Heilig HG, Zoetendal EG, Stroes ES, de Vos WM, Hoekstra JB, Nieuwdorp M. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913-916 e917.
  124. Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R, Marcovina S, Fowler S. The effect of metformin and intensive lifestyle intervention on the metabolic syndrome: the Diabetes Prevention Program randomized trial. Ann Intern Med. 2005;142(8):611-619.
  125. Leroith D. Pathophysiology of the metabolic syndrome: implications for the cardiometabolic risks associated with type 2 diabetes. Am J Med Sci. 2012;343(1):13-16.
  126. Buchwald H, Avidor Y, Braunwald E, Jensen MD, Pories W, Fahrbach K, Schoelles K. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292(14):1724-1737.
  127. Shuai X, Tao K, Mori M, Kanda T. Bariatric surgery for metabolic syndrome in obesity. Metab Syndr Relat Disord. 2015;13(4):149-160.
  128. O'Brien PE, Dixon JB, Laurie C, Skinner S, Proietto J, McNeil J, Strauss B, Marks S, Schachter L, Chapman L, Anderson M. Treatment of mild to moderate obesity with laparoscopic adjustable gastric banding or an intensive medical program: a randomized trial. Ann Intern Med. 2006;144(9):625-633.
  129. Executive summary: Standards of medical care in diabetes--2012. Diabetes Care. 2012;35 Suppl 1:S4-S10.
  130. American Diabetes A. (5) Prevention or delay of type 2 diabetes. Diabetes Care. 2015;38 Suppl:S31-32.
  131. Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL, Jr., Jones DW, Materson BJ, Oparil S, Wright JT, Jr., Roccella EJ. Seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Hypertension. 2003;42(6):1206-1252.
  132. Onat A. Metabolic syndrome: nature, therapeutic solutions and options. Expert Opin Pharmacother. 2011;12(12):1887-1900.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Hypothyroidism in Pregnancy

CLINICAL RECOGNITION

The prevalence of overt and subclinical hypothyroidism in pregnancy is 0.3-0.5% and 2-3% respectively. Overt hypothyroidism in pregnancy may present classically but is often subtle and difficult to distinguish from the symptoms of normal pregnancy. A high index of suspicion is therefore required, especially in women with a predisposition to thyroid disease such as a personal or family history of thyroid disease, the presence of goiter or the co-existence of other autoimmune disorders like type 1 diabetes. Subclinical hypothyroidism (high TSH with normal FT4) accounts for the majority of cases. Isolated hypo-thyroxinemia (FT4 below the trimester specific reference range without elevation of TSH) occurs in about an equal number of cases.

PATHOPHYSIOLOGY

Although endemic iodine deficiency (areas where the ambient urinary iodine concentration is less than 50µg/liter) is the most common cause of hypothyroidism worldwide, the main cause in iodine-replete populations is chronic autoimmune thyroiditis. Other causes include post-surgical or post-radioiodine ablation. Adverse effects on mother and child range from anemia in pregnancy to miscarriage, or if pregnancy is continued, preterm birth with its consequences (table 1). Even in an iodine sufficient area maternal thyroid dysfunction (hypothyroidism, subclinical hypothyroidism or hypothyroxinemia) during pregnancy results in neuro-intellectual impairment of the child; hence maternal thyroid hormones are required through gestation for proper fetal brain development. Specific effects will depend on when maternal hormone deficiency occurs during pregnancy. Low maternal thyroid hormone concentrations in early gestation can be associated with significant decrements of IQ of young children. A significant decrement in IQ has also been reported in children born to euthyroid mothers with circulating anti TPO antibodies, but this is not an established association as yet.

 

Table 1. Adverse Outcomes of Pregnancy with Maternal Hypothyroidism

Infertility

Miscarriage

Increased fetal death rate

Anemia in pregnancy

Preeclampsia

Abruptio placenta

Postpartum hemorrhage

Preterm birth

Low birth weight

Increased neonatal respiratory distress

Impaired neurointellectual child development

 

DIAGNOSIS AND DIFFERENTIAL

 

Hypothyroidism is diagnosed on the basis of a low FT4 or TT4 and high TSH. In pregnancy there are changes in the ranges of both these hormones requiring the use of gestational trimester-specific reference ranges. Thyroid antibody testing (thyroid peroxidase antibody) confirms the autoimmune nature of hypothyroidism and may also identify antibody positive women who are at risk of postpartum thyroiditis. Subclinical hypothyroidism is diagnosed when TSH is above the reference range while the T4 level is normal. The TSH level is difficult to interpret during the 1st trimester due to the weak thyromimetic action of hCG. Isolated hypothyroxinemia occurs most frequently in the 3rd trimester. The clinical significance is not clear as it may arise due to hemodilution.

 

Table 2. Etiology and Diagnosis of Hypothyroidism During Pregnancy

Autoimmune thyroiditis:  Positive thyroid antibody test (TPOAb)

Iodine deficiency: Low urinary iodine, Goiter

Post-surgical: History of Graves’ disease, toxic nodular goiter, thyroid cancer, benign goiter

 

Table 3. Patients at Risk of Thyroid Dysfunction (American Thyroid Association Case Finding Criteria)

1.         Age >30 years

2.         History of thyroid dysfunction or positive thyroid antibodies

3.         Type 1 diabetes or other autoimmune disorders

4.         Head or neck radiation

5.         Use of drugs that affect thyroid function

6.         Administration of iodinated contrast materials

7.         Goiter or symptoms or signs of thyroid dysfunction

8.         Residents in areas of moderate to severe iodine deficiency

9.         Multiple prior pregnancies (> 2)

10.       Previous pregnancy loss, preterm delivery, or infertility

11.       Family history of thyroid disease

12.       Morbid obesity (BMI > 40 kg/m2)

 

THERAPY

 

Women with overt hypothyroidism in pregnancy should be treated with levothyroxine, but there is no consensus regarding treatment for women with subclinical hypothyroidism or isolated hypothyroxinemia. Treatment should be considered in women with subclinical hypothyroidism if they have TSH concentrations above 10 mU/L, positive thyroid antibodies, or other risk factors for thyroid disease such as goiter, personal or family history of thyroid autoimmunity, or type 1 diabetes (table 3). Women with infertility or recurrent pregnancy loss could also be treated on the basis that treatment could potentially improve live delivery rates. Treatment of isolated hypothyroxinemia is controversial and is probably not indicated in the 3rd trimester.

 

In women not receiving T4 who may have risk factors for thyroid disease (e.g. personal or family history of an autoimmune disorder, positive thyroid antibodies, Type 1 DM, prior pre-term delivery, possible iodine deficiency, or neck irradiation) thyroid function should be measured pre-conception. If the TSH is above the laboratory reference range, the test should be confirmed, and supplemental thyroxine therapy should be considered, especially if thyroid antibodies or other risk factors for thyroid disease are present (table 3). Women with thyroid autoimmunity who are euthyroid in the early stages of pregnancy are at risk of developing hypothyroidism and should be monitored for elevation of TSH above the normal range for pregnancy.

 

Women receiving T4 for hypothyroidism before pregnancy should have thyroid function checked to maintain TSH levels not higher than 2.5mIU/L in the first trimester and not higher than 3.0mIU/L in subsequent trimesters. As soon as pregnancy is confirmed T4 dose should be increased by 30-50% and TFTs checked every 4 weeks. Note that TSH level is difficult to interpret in the 1st trimester due to HCG action. Not all women require an increase in T4 dosage in pregnancy. Women who are newly diagnosed to be hypothyroid in pregnancy should receive 100µg T4 daily and the dose adjusted after 4 weeks to the optimal level. In summary, women with overt hypothyroidism or with subclinical hypothyroidism who are TPO antibody positive should be treated with oral levothyroxine.

Screening

Because of the proven adverse effects of hypothyroidism on pregnancy, and the failure of testing only women at “high risk” of hypothyroidism (defined above) to detect more than 50% of thyroid problems, a case can be made for screening all women for thyroid function in early pregnancy with administration of levothyroxine in women with subnormal thyroid function. Another recommended approach is to screen only women at “high risk”. However, the issue remains unsettled.

 

FOLLOW-UP

 

In women with previously treated Graves’ hyperthyroidism who are receiving thyroxine for post ablative hypothyroidism the Thyroid Stimulating Hormone Receptor Antibodies (TRAbs) assay may be positive even many years later. The woman should be counselled if another pregnancy is planned to guard against fetal or neonatal hyperthyroidism due to transplacental passage of maternal TRAb. Before a further pregnancy thyroid function should be checked in order to keep the TSH less than 2.5mIU/l. When first pregnant the woman should increase T4 dose by 25-50% (usually by 50 micrograms per day) and then have a further thyroid function test 4 weeks later and at least in every trimester thereafter. About 25% do not require an increase in T4 dose.

 

GUIDELINE

 

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 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017 Mar;27(3):315-389.

REFERENCES

 

Teng W, Shan Z, Patil-Sisodia K, Cooper DS. Hypothyroidism in pregnancy.

Lancet Diabetes Endocrinol. 2013;1:228-237.

 

Krassas GE, Poppe K, Glinoer D. Thyroid function and human reproductive health. Endocr Rev 2010;31:702-755

Lazarus J. Thyroid dysfunction and pregnancy. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2016 Jul 21.

 

Okosieme OE, Khan I, Taylor PN. Preconception management of thyroid dysfunction. Clin Endocrinol (Oxf). 2018 Sep;89(3):269-279.

 

 

 

 

 

Hyperthyroidism in Pregnancy

CLINICAL RECOGNITION

 

Hyperthyroidism occurs in approximately 0.2-1.0% of all pregnancies. Most cases are due to Graves’ disease. The clinical recognition of Graves’ disease may prove challenging in pregnancy since the features of normal pregnancy overlap with symptoms of hyperthyroidism. Specific features which may point to Graves’ hyperthyroidism include the presence of a diffuse goiter, ophthalmopathy, or pre-tibial myxedema (Table 1).

 

Table 1. Features of Graves’ Disease

Past history of autoimmune thyroid disease

Family history of autoimmune thyroid disease

Features of hyperthyroidism such as weight loss and heat intolerance

Diffuse goiter

Ophthalmopathy

Pre-tibial myxedema

Proximal myopathy

Nail changes: onycholysis

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

Hyperthyroidism is diagnosed on the basis of elevated trimester specific serum levels of free thyroxine (FT4) or free triiodothyronine (FT3) (or comparable measures of total thyroxine or FTI) and low thyroid stimulating hormone (TSH). In subclinical hyperthyroidism FT4 and FT3 are normal but TSH is low or suppressed to undetectable levels. Treatment is generally not required for subclinical hyperthyroidism in pregnancy. In fact, most instances of a low TSH in early pregnancy are not pathological and are due to TSH suppressive effects of β-human chorionic gonadotrophin (β-HCG). Detection of thyroid stimulating hormone receptor antibodies (TRAbs) in serum is diagnostically helpful in distinguishing Graves’ disease from other pathological and non-pathological causes of a low TSH. Most cases of hyperthyroidism in pregnancy are due to Graves’ disease. Other causes include single or multiple toxic nodules and thyroiditis (Table 2).

 

Table 2. Causes of Hyperthyroidism in Pregnancy

Graves’ disease

Gestational thyrotoxicosis

Single toxic nodule

Toxic multinodular goiter

Subacute thyroiditis

Silent thyroiditis

Thyrotoxicosis factitial

 

It is important to distinguish hyperthyroidism in pregnancy from gestational transient thyrotoxicosis (GTT) which occurs as a result of the thyroid stimulatory actions of β–HCG. GTT is more common than Graves’ disease being diagnosed in about 1-3% of all pregnancies. It may be associated with hyperemesis gravidarum, choriocarcinoma, or hydatiform mole and in rare instances may result from functional mutations which increase TSH receptor hypersensitivity to β-HCG. GTT is typically mild in presentation, self-limiting and rarely requires specific treatment with antithyroid medications (Table 3). While GTT may be difficult to distinguish from Graves’ disease, features such as goiter, ophthalmopathy, or pretibial myxedema are suggestive of Graves’ disease.

 

Table 3. Clinical Differences Between Graves’ Disease in Pregnancy and Gestational Transient Thyrotoxicosis

Graves’ Disease

Gestational Thyrotoxicosis

Past history of thyroid autoimmunity

Family history of thyroid autoimmunity

May exhibit overt hyperthyroid features

Goiter may be present

Ophthalmopathy may be present

TRAb positive

TPOAb positive

No past history of thyroid autoimmunity

No family history of thyroid autoimmunity

May present with hyperemesis, dehydration and   electrolyte imbalance

No goiter

No ophthalmopathy

TRAb negative

TPOAb negative

 

Thyroid Stimulating Hormone Receptor Antibodies (TRAbs)

 Measurement of TRAbs is useful for monitoring for the risk of fetal and neonatal hyperthyroidism (Table 4). The management of such patients can be considered in three categories.

 

Table 4. Measurement of Thyroid Stimulating Hormone Receptor Antibodies (TRAbs)

Patients with active hyperthyroidism

Patients previously treated with radioiodine or surgery

Patients with high TRAb levels require serial fetal monitoring with ultrasonography

 

PATIENTS IN REMISSION FROM HYPERTHYROIDISM

 

Patients who have successfully completed treatment for hyperthyroidism who become pregnant while in remission require close monitoring since there is a risk of relapse in pregnancy. Such patients may continue to harbor TRAbs with the risk of transplacental transfer. This risk is lowest for patients who were treated with antithyroid drugs and it is recommended that TRAbs are checked in early pregnancy in patients who were treated with surgery or radioiodine. If TRAbs are positive in early pregnancy, then fetal monitoring is indicated with repeated measurement of TRAbs at 18-22 weeks, and again at 30-34 weeks if TRAbs continue to be positive.  TRAb levels >5 IU/L or more than 3 times the upper limit of normal is an indication for close fetal or neonatal monitoring due to the high risk of fetal/neonatal hyperthyroidism from transplacental TRAb transfer.

 

PATIENTS CURRENTLY UNDERGOING TREATMENT FOR HYPERTHYROIDISM

 

Women who conceive while on antithyroid treatment should have TRAbs level checked in early pregnancy and again at 18-22 weeks, and at 30-34 weeks if TRAbs continue to be positive. Women of child bearing age with Graves’ disease should be counselled against becoming pregnant until a euthyroid state is achieved.

 

PATIENTS WHO DEVELOP HYPERTHYROIDISM DURING PREGNANCY

 

Patients who develop hyperthyroidism for the first-time during pregnancy are at particular risk of adverse fetal and maternal adverse effects and should be controlled promptly and monitored carefully. TRAB levels should also be checked in late pregnancy to assess the risk of neonatal hyperthyroidism.

 

PATHOPHYSIOLOGY

 

Uncontrolled hyperthyroidism is associated with adverse feto-maternal effects including pre-eclampsia, maternal congestive cardiac failure, miscarriages, premature birth, still-birth, and low birth weight. Furthermore, neonates of hyperthyroid mothers with Graves’ disease are at risk of developing fetal hyperthyroidism and goiter due to the transplacental transfer of TRAbs. Fetal hypothyroidism may also develop due to transplacental transfer of maternal antithyroid drugs or in rare instances from transplacental transfer of maternal blocking TRAbs.

 

THERAPY

 

The natural course of Graves’ disease in pregnancy should be borne in mind during therapy. Due to the immune tolerant state of pregnancy there is a tendency for Graves’ disease to remit towards the latter stages of gestation.

 

Anti-Thyroid Drugs

 

Anti-thyroid drugs are the treatment of choice for hyperthyroidism in pregnancy (Table 5). The lowest effective dose should be used. Treatment should be monitored with FT4 and TSH. These should be measured every 2-4 weeks initially and then 4-6 weekly once thyroid hormone levels are stabilized. FT4 levels should be maintained at or just above the upper limit of the trimester specific reference range.

 

Dose reductions or even cessation of therapy with careful monitoring may be necessary in late pregnancy. The thionamides, propylthiouracil (PTU), methimazole (MMI), and its pro-drug derivative, carbimazole are the antithyroid drugs of choice. Both propylthiouracil and methimazole exhibit similar placental transfer kinetics, have similar effects on fetal and neonatal thyroid function, and are equally safe in lactation. Methimazole has greater efficacy than propylthiouracil and is associated with better compliance since it can be administered once daily whereas propylthiouracil needs to be taken twice or thrice daily. More so a growing number of reports have highlighted the association of propylthiouracil with fatal liver failure. However, methimazole is associated, rarely, with the occurrence of aplasia cutis and methimazole embryopathy in the neonate. Although this risk is slight it is most likely with methimazole administration in early pregnancy during embryogenesis. For the above reasons it is recommended that propylthiouracil is used in the first trimester and that consideration should be given to switching to methimazole in later pregnancy.

 

Table 5. Guidelines for Anti-Thyroid Drugs (ATD) in Pregnancy

PTU is recommended in first trimester

Consider switching to MMI from second trimester

Use lowest effective dose of ATD

Consider reducing dose or stopping ATD in later pregnancy

Monitor treatment with FT4 and TSH: Initially 2-4 weekly, later 4-6 weekly.

Aim for FT4 at or just above the upper end reference range

 

Beta-blocking agents such as propranolol may be used to control severe adrenergic symptoms but should be discontinued once symptoms begin to improve, usually within 2-4 weeks. The combination of thionamides with levothyroxine i.e. block and replace therapy is not recommended in pregnancy as this may lead to fetal hypothyroidism due to disproportionately greater transplacental transfer of antithyroid drugs than levothyroxine. Radioactive iodine is absolutely contraindicated in pregnancy. Adverse effects of radioiodine on fetal thyroid function include fetal hypothyroidism and this is more likely in later pregnancy since the fetal thyroid only starts to actively concentrate iodide from about 12 weeks gestation.

Surgery

 

Thyroidectomy is an option in patients with significant problems with compliance or severe adverse reactions to antithyroid medications. Surgery is best undertaken in the second trimester of pregnancy.

FOLLOW UP

Newborn

Following delivery, the infant of hyperthyroid mothers should be monitored for thyroid dysfunction. Transient neonatal hyperthyroidism due to transplacental transfer of maternal TRAbs is seen in 1-5% of neonates of mothers with Graves’ disease. The presentation may be more obvious after the first few days of life since TRAbs are cleared from the neonatal circulation at a slower rate than maternal antithyroid drugs.

Mothers

Mothers with a past history of hyperthyroidism require regular thyroid function tests after delivery since Graves’ disease may relapse in the postpartum period. Anti-thyroid drugs are safe in lactation and if indicated should be used at the lowest effective dose, preferably administered after breast feeds in divided doses. Women with risk factors for autoimmune thyroid dysfunction may develop postpartum thyroiditis (PPT).  This occurs in 5-9% of pregnancies and is characterized by a transient hyperthyroid phase followed by a hypothyroid phase before return to euthyroidism. The hyperthyroid phase will usually not require treatment but levothyroxine may be given to symptomatic women in the hypothyroid phase. Long term follow-up is necessary due to the risk of permanent hypothyroidism.

GUIDELINES

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 Guidelines of the

American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017 Mar;27(3):315-389.

 

REFERENCES

 

Cooper DS, Laurberg P. Hyperthyroidism in pregnancy. Lancet Diabetes

Endocrinol. 2013;1:238-249.

 

Krassas GE, Poppe K, Glinoer D. Thyroid function and human reproductive health. Endocr Rev 2010;31:702-755. 

 

Lazarus J. Thyroid dysfunction and pregnancy. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2016 Jul 21.

 

Okosieme OE, Khan I, Taylor PN. Preconception management of thyroid dysfunction. Clin Endocrinol (Oxf). 2018 Sep;89(3):269-279.