Archives

Adrenal Insufficiency

ABSTRACT

 

Adrenal insufficiency (AI) is an uncommon clinical condition resulting from inadequate glucocorticoid secretion or action, either at the basal state or during stress. Hypothalamic-pituitary-adrenal axis disturbance or primary adrenal failure itself is responsible for this condition. AI presentation can range from asymptomatic hormonal dysfunction to adrenal crisis. Prompt diagnosis and management are crucial since AI may be fatal if unrecognized or untreated. Among the various investigations, the appropriate tests should be performed in a timely manner in order to reverse hormonal and metabolic disturbances, treat associated conditions, and prevent acute crises. In this review, we will provide background knowledge regarding pathophysiology, clinical manifestations, underlying etiologies, hormonal investigations, and related treatments in AI.

 

CLINICAL RECOGNITION

 

Adrenal insufficiency (AI) is a life-threatening condition characterized by the failure of adrenocortical function. This failure results in impaired secretion of glucocorticoids (GCs) only or of GCs and mineralocorticoids (MCs) and adrenal androgens, which are crucial for energy, salt and fluid homeostasis, and androgenic activity. The disorder may be caused by adrenocortical disease, primary adrenal insufficiency (PAI), known as Addison’s disease, which is a rare disease with reported prevalence in Europe ranging from 39 cases/million in England in 1968 (1) to 221 cases/million in Iceland in 2016 (2), the highest prevalence reported. Depending on the study, the annual incidence of AI in Europe is estimated to range between 4.4 and 6.2 new cases per million people (3,4). AI may be secondary in the setting of conditions affecting the pituitary gland and the secretion of adrenocorticotropic hormone (ACTH) (secondary AI) and/or the hypothalamus and the secretion of corticotropin-releasing hormone (CRH) and/or other ACTH secretagogues such as vasopressin (tertiary AI). Central AI is estimated to have a prevalence between 150 and 280 per million, making it more prevalent than PAI (5-7). A prevalence of AI after pituitary surgery varies, with higher rates of up to 90% after craniopharyngioma surgery, while hypopituitarism has high prevalence after cranial radiation for non-pituitary tumors, but may take several years to develop.

 

The clinical manifestations of AI depend upon the extent of loss of adrenal function, and whether MCs and androgen production are preserved, whereby the renin-angiotensin-aldosterone system (RAAS) is intact in central AI. The presentation can be acute or insidious, depending on the underlying cause of the adrenal failure. The diagnosis may be delayed until an intercurrent illness, such as serious infection, acute stress, bilateral adrenal infarction, or hemorrhage, precipitates a life-threatening adrenal crisis. The symptoms of PAI are pleiotropic and non-specific, except for salt craving (Table 1).

 

Adrenal crisis is an acute deterioration in health which is associated with hypotension, acute abdominal symptoms, and marked laboratory abnormalities, which resolve after parenteral glucocorticoid administration. In PAI, the patients may have more severe symptoms which are due to concomitant MCs deficiency. This condition is commonly presented in PAI, but less common in central AI, in which a typical example would be pituitary apoplexy. Retrospective and prospective analysis revealed a prevalence of adrenal crises 6.6-8.3 cases/100 patient-years, with mortality 0.5/100 patient-years, mainly due to gastrointestinal and other infectious diseases (8). A recent retrospective case-control analysis of the European Adrenal Insufficiency Registry (EU-AIR) reported an incidence of adrenal crisis of 6.53 cases per 100 patient-years for PAI and 3.17 cases per 100 patient-years for central AI (9).

 

A significant feature to clinically differentiate PAI from central AI is skin pigmentation, which is nearly always present in long-standing PAI. The most probable cause of the pigmentation seems to be the increased stimulation of the melanocortin-1 receptor (MC1R) by elevated levels of ACTH itself with its intrinsic α-melanocyte stimulating hormone activity. The rest of the clinical features of secondary and tertiary AI are similar to those of PAI type (Table 1). Rarely, the presentation may be more acute in patients with pituitary apoplexy. Hyponatremia and compensatory water retention may be the result of an “inappropriate” increase in vasopressin secretion. The clinical manifestations of a pituitary or hypothalamic tumor, such as symptoms and signs of deficiency of other anterior pituitary hormones, headache or visual field defects, may also be present.

 

Another condition with a dissociation in GCs and MCs secretion presenting as AI is congenital adrenal hyperplasia (CAH), with a frequency of adrenal crisis of 5.8 cases/100 patient-years (4.9 cases/100 patient-years after correction for a neonatal salt-wasting crisis) and often respiratory infections, firstly in early childhood, followed by gastrointestinal infections at older ages (10).

 

Table 1. Symptoms, Physical Findings, and Laboratory Findings Associated with Adrenal Insufficiency

SYMPTOMS AND PHYSICAL FINDINGS

Adrenal crisis: hypotension (< 110mmHg systolic) and syncope/ shock (> 90%); volume depression

Non-specific symptoms:

·       Gastrointestinal symptoms: abdominal pain, flank pain, back pain, or lower chest pain: 86%- may mimic acute abdomen

·       Fever (66%)

·       Anorexia (early feature), nausea, vomiting (47%)

·       Abdominal rigidity or rebound tenderness (22%)

·       Diarrhea, which may alternate with constipation

·       Neuropsychiatric symptoms: Confusion, lethargy, disorientation, coma (42%)

PAI>SAI/TAI

 

Psychiatric symptoms: memory impairment, depression, anxiety, psychosis,reduced consciousness, delirium

Chronic AI

General malaise, weakness, fatigue, lassitude, generalized weakness

PAI/SAI

Hypoglycemia; increased risk in children, thin women, alcohol abuse, GH deficiency

SAI>>PAI

 

Sudden severe headache, loss of vision or visual field defect

SAI (pituitary apoplexy)

Skin

·       Hyperpigmentation: sun-exposed or pressure areas, recent scars (after AI manifestation), axillae, nipples, palmar creases, mucous membranes as buccal mucosa

·       Vitiligo (as marker of autoimmune disease)

Chronic PAI

Postural hypotension due to volume depletion, or improvement in blood pressure control in previously hypertensive patients, postural dizziness

PAI>>SAI

Salt craving (22%)

PAI

Autoimmune manifestations: vitiligo, autoimmune thyroid disease, type 1 diabetes, primary ovarian failure, autoimmune gastritis

PAI

Weight loss

Chronic PAI/SAI

Decreased axillary and pubic hair, loss of libido in females (DHEA deficiency), amenorrhea in women (in 25% due to chronic illness, weight loss or associated premature ovarian failure)

PAI/SAI

Auricular calcification

 

Low grade fever

PAI

Associated endocrinopathies in the context of autoimmune polyglandular syndrome

PAI

LABORATORY FINDINGS

Electrolyte abnormalities:

·       Hyponatremia: 85-90% (PAI: MCs deficiency; CAI: dilutional effect)

·       Hyperkalemia: 60-65% due to MCs deficiency

·       Metabolic acidosis

·       Mild hypercalcemia (uncommon)

 

PAI/SAIPAI

PAI

PAI

Azotemia

PAI

Liver enzymes abnormalities: may be observed in autoimmune hepatitis

PAI

Changes in blood count:

·       Mild anemia (normocytic normochromic)

·       Eosinophilia

·       Lymphocytosis

 

TSH with normal or low normal T4 (transient with ACTH; permanent with autoimmune thyroiditis)

PAI

 Erythrocyte Sedimentation Rate

 

ACTH: adrenocorticotropic hormone; AI: adrenal insufficiency; DHEA: dehydroepiandrosterone); GCs: glucocorticoids; GH: growth hormone; MCs: mineralocorticoids; PAI: primary AI, SAI: secondary AI, T4: thyroxine; TAI: tertiary AI; TSH: thyrotropin stimulating hormone

 

PATHOPHYSIOLOGY

 

The majority of cases of PAI are the result of gradual destruction of all three layers of the adrenal cortex. Clinical manifestations of this condition appear when the loss of the adrenocortical tissue of the combined glands is greater than 90%. In the initial phase of chronic gradual destruction, adrenal reserve is decreased, and although the basal steroid secretion is normal, the secretion in response to stress is suboptimal, resulting in inadequate GCs, MCs and androgen production, leading to partial ΑΙ; this is manifested by an inadequate cortisol response during stress. Any major or even minor stressor can precipitate an acute adrenal crisis, followed by a complete AI, since with further loss of adrenocortical tissue, even basal steroid secretion is decreased, leading to the clinical manifestations of the disease. Adrenal hemorrhage or infarction may lead to adrenal crisis, a medical emergency manifesting as hypotension and acute circulatory failure crisis due to MCs deficiency when the appropriate doses of GCs are not met to cover MCs requirements. On the other hand, GCs deficiency may also contribute to hypotension by decreasing vascular responsiveness to angiotensin II, norepinephrine/noradrenaline, and other vasoconstrictive hormones, reducing the synthesis of renin substrate, and increasing the production and effects of prostacyclin and other vasodilatory hormones. Combined GCs and MCs deficiency leads to increased urinary sodium loss and hypovolemia resulting in hypotension and electrolyte imbalance including hyponatremia and hyperkalemia. ‘Inappropriate’ anti-diuretic hormone (ADH) release and action on the renal tubule due to GCs deficiency contributes to the hyponatremia, although it could be argued that this attempt at volume maintenance is far from inappropriate. Low plasma cortisol concentrations reduce GCs negative feedback, which in turn increases the production and secretion of ACTH and other POMC-peptides. These mechanisms are responsible for the well-recognized hyperpigmentation by acting on the MC1R in the skin.

 

Conversely, ACTH deficiency in central AI leads to decreased secretion of cortisol and adrenal androgens, while MCs production remains normal, as MCs are principally regulated by the RAAS. In the early stages, basal ACTH secretion is normal, while its stress-induced release is impaired. With further loss, there is atrophy of zonae fasciculata and reticularis of the adrenal cortex. Therefore, basal cortisol secretion is decreased but aldosterone secretion by the zona glomerulosa is preserved. However, hypotension in central AI may still occur due to decreased vascular tone as a result of reduced vascular responsiveness to angiotensin II and noradrenaline.

 

DIAGNOSIS and DIFFERENTIAL DIAGNOSIS

 

Table 2. Etiology of Primary Adrenal Insufficiency (11-17)

Autoimmune

·      Sporadic: not associated with other autoimmune disorders

·      APS type 1 or autoimmune polyendocrinopathy-candidiasis-ectodermal dysplasia (APECED): Addison’s disease, chronic mucocutaneous candidiasis, hypoparathyroidism, dental enamel hypoplasia, pernicious anemia, alopecia, primary gonadal failure

·      APS type 2 or Schmidt’s syndrome: Addison’s disease, autoimmune thyroid disease, primary gonadal failure, type 1 diabetes, celiac disease, pernicious anemia, myasthenia gravis, vitiligo

·      APS type 4: Addison’s disease with other autoimmune diseases excluding autoimmune thyroid disease and type 1 diabetes

Infections

·       Tuberculosis

·       Fungal infections: histoplasmosis, cryptococcosis, candidiasis, African trypanosomiasis, paracoccidioidomycosis (South America)

·       Syphilis

·       Cytomegalovirus, HIV (up to 5% patients with AIDS)

Metastases

From lung, breast, kidney, colon cancers, melanoma, lymphoma

Infiltrations

·       Sarcoidosis

·       Amyloidosis

·       Haemochromatosis

Intra-adrenal hemorrhage

·       Drugs: anticoagulant, tyrosine kinase inhibitor

·       Trauma

·       Waterhouse-Friderichsen syndrome: mostly associated with meningococcal septicemia

Infarction

Anti-phospholipid syndrome

Hematological disorders

Lymphoma

Adrenoleukodystrophy (ABCD1 and ABCD2 gene mutations): X-linked disorder of very long chain fatty acid (VLCFA) metabolism, presents in childhood, may progress to severe spinal cord problems, adrenomyeloneuropathy (AMN), and cerebral demyelination causing dementia.

Kearns-Sayre syndrome (mitochondrial DNA deletions): progressive external ophthalmoplegia, bilateral pigmentary retinopathy, cardiac conductions, CNS dysfunction, endocrine abnormalities (Addison’s disease, hypogonadism, hypothyroidism, hypoparathyroidism, diabetes mellitus)

Wolman’s disease (LIPA gene mutations): inherited disorder of lysosomal enzyme, presented with abdominal distension from accumulation of foamy lipid droplet in various organs, along with adrenal calcification and malabsorption.

Congenital adrenal hyperplasia (CAH)

Autosomal recessive disorders of enzyme deficiency in adrenal steroidogenesis pathway, which can have various manifestations depending on their subtype and severity. Hence, this disease may be diagnosed in older individuals.

·       21-Hydroxylase deficiency (CYP21A2 gene mutation): the most common type, may be presented with salt wasting form in infancy, or simple virilizing form in later life if neonatal screening is not performed

·       11β-hydroxylase deficiency (CYP11B1 gene mutation): hyperandrogenism with hypertension in older children and adults

·       3β-hydroxysteroid dehydrogenase 2 deficiency (CYP3B2 gene mutation): neonatal wasting, ambiguous genitalia in boys, hyperandrogenism in girls

·       P450 oxidoreductase deficiency (POR gene mutation): abnormal genitalia with or without skeletal malformations, and with or without maternal virilization

·       P450 side-chain cleavage deficiency (CYP11A1 mutations): may be presented with neonatal salt wasting with 46,XY under-androgenization, or later onset of PAI and ambiguous genitalia

·       Congenital lipoid adrenal hyperplasia (StAR gene mutations): may be presented with varied severity of PAI and 46,XY DSD

Congenital adrenal hypoplasia

·       X-linked form (NR0B1 mutations or deletion): variable manifestations, hypogonadotropic hypogonadism, impaired spermatogenesis, hypoaldosteronism, shock

·       Xp21 contiguous gene syndrome (deletion of NR0B1, glycerol kinase and genes for Duchenne muscular deficiency): with psychomotor retardation, hepatic iron deposition

·       SF-1 linked (NR5A1 mutations or deletions): range from isolated adrenal failure to isolated gonadal failure, XY sex reversal, 46,XX DSD, gonadoblastoma, gonadal insufficiency

·       IMAGe syndrome (CDKN1C pathogenic variant): Intrauterine growth restriction, Metaphyseal dysplasia, Adrenal hypoplasia congenita, and Genital abnormalities in males

·       MIRAGE syndrome (SAMD9 pathogenic variant): Myelodysplasia, Infection, Restriction of growth, Adrenal hypoplasia, Genital phenotypes, and Enteropathy)

·       Familial steroid-resistant nephrotic syndrome with AI (SGPL1 mutations)

Inherited unresponsiveness to ACTH syndromes

Familial glucocorticoid deficiency (FGD): autosomal recessive cause of childhood onset AI, hyperpigmentation, hypoglycemia with normal MCs activity.

·       Type 1 variant (MC2R gene mutations)

·       Type 2 variant (MRAP gene mutations)

·       Other variants (MCM4, NNT, TXNRD2, GPX1, PRDX3, partial mutation of StAR and CYP11A1)

·       Triple A or Allgrove syndrome (AAAS gene mutations): Addison’s disease, Achalasia, Alacrima, along with neurodegenerative change with or without mental retardation

Iatrogenic

Bilateral adrenalectomy

Drugs

·       Inhibition of steroidogenesis: ketoconazole, fluconazole, etomidate, aminoglutethimide, suramin

·       Acceleration of cortisol metabolism: phenytoin, phenobarbital, thyroxine, rifampicin, St John’s Wort (Hypericum perforatum)

·       Promotion of adrenolytic activity: mitotane

·       Enhancement of autoimmunity: CTLA-4 inhibitors

 

Table 3. Etiology of Central Adrenal Insufficiency (18,19)

Pituitary, parasellar and hypothalamic masses

·       Pituitary adenomas, rarely carcinomas

·       Parasellar lesions: cysts, craniopharyngioma

·       Nonadenomatous neoplasms: meningioma, chordoma, glioma, germinoma, pituicytoma

·       Metastases: breast, lung, prostate, colon, lymphoma

Infections

·       Pituitary abscess: Gram-positive cocci

·       Tuberculosis

·       Syphilis, leptospirosis

·       Fungal infections: candidiasis, aspergillosis

·       Herpes/Varicella infection, SARS-CoV-2 virus

Infiltrations

·       Hypophysitis: lymphocytic, granulomatous, xanthomatous, necrotizing, IgG4-related, immunotherapy-induced, other autoimmune-associated

·       Hemochromatosis

·       Sarcoidosis

·       Histiocytosis X

·       Wegener’s granulomatosis

Hemorrhage

·       Pituitary apoplexy

Infarction

·       Sheehan’s syndrome

Iatrogenic

·       Pituitary surgery

·       Pituitary irradiation

Drugs

·       Steroid

·       Mifepristone: impaired GCs signal transduction

·       Somatostatin analogues

·       Opiates

·       Antipsychotics and antidepressants

Trauma

·       Traumatic brain injury

Transcription factor mutations

Hereditary ACTH deficiency can manifest as an isolated pituitary defect or as a combination of pituitary hormone deficiencies.

·       HESX1: panhypopituitarism, cognitive change, septo-optic dysplasia

·       OTX2: panhypopituitarism, neonatal hypoglycemia, pituitary hypoplasia, ectopic posterior pituitary

·       LHX4: panhypopituitarism

·       PROP1: panhypopituitarism

·       SOX3: panhypopituitarism, infundibular hypoplasia, mental retardation

·       TBX19: isolated ACTH deficiency

POMC and related processing

·       POMC gene mutations: AI, severe early-onset obesity, hyperphagia, red hair, pale skin

·       PC1 mutations: AI, abnormal glucose metabolism, early-onset obesity, hypogonadotropic hypogonadism, neonatal-onset persistent malabsorptive diarrhea

Prader-Willi syndrome: hypotonia, failure to thrive, obesity, multiple endocrine abnormalities (GH deficiency, central hypothyroidism, hypogonadotropic hypogonadism, central AI)

Familial corticosteroid binding-globulin deficiency: unexplained fatigue, hypotension

Idiopathic hypopituitarism

ACTH, adrenocorticotropic hormone; AI, adrenal insufficiency; APS, autoimmune polyglandular syndrome; CAH, congenital adrenal hyperplasia; DHEA, dehydroepiandrosterone; DSD, disorders of sexual development; GCs, glucocorticoids;GH, growth hormone; HPA, hypothalamic-pituitary-adrenal; MCs, mineralocorticoids; PAI, primary AI; POMC, pro-opiomelanocortin; TSH, thyrotropin stimulating hormone.

 

When an adrenal crisis is present, there is no need for immediate investigation to confirm AI but treatment should be initiated without delay as soon as clinically suspected. Clinical suspicion could be the case of a cancer patient under treatment with immunotherapy, particularly with the anti-CTLA-4 agent ipilimumab and signs and/or symptoms suspicious for AI, where the differential diagnosis of the rare metastatic involvement of adrenal or pituitary gland has to be also considered (20,21). In any case, confirmatory testing should be deferred until the patient has been stabilized; however, a blood sample taken at this time for cortisol and ACTH levels is extremely helpful for later assessment. In cases of insidious presentation, clinical suspicion of AI should be followed by diagnostic dynamic tests to confirm the inappropriately low cortisol secretion and whether cortisol deficiency is dependent or independent of ACTH deficiency by measuring ACTH levels (Table 4). In PAI, cortisol deficiency results in decreased feedback to the HPA axis, leading to increased secretion of ACTH to stimulate the adrenal cortex. Simultaneously, MCs deficiency causes increased release of renin by the juxtaglomerular apparatus of the kidneys.

 

In a non-acute setting, the diagnosis should be suspected based on the patient’s history and physical examinationalong with low morning cortisol levels, and confirmed by an ACTH stimulation test. Basal morning cortisol levels lower than 3μg/dL suggest ACTH deficiency. Conversely, morning cortisol higher than 15μg/dL indicates sufficient ACTH reserve. However, random cortisol levels are not advised for the diagnosis of AI. Additional tests are required to establish the diagnosis of AI if the cortisol levels are in the range of these cut-offs. Based on the pathophysiology, different dynamic tests have been proposed to diagnose this condition.

 

The classic short Synacthen test (SST; 250μg of ACTH [1-24], i.m. or i.v.) is considered the standard diagnostic method to detect AI, with a sensitivity of 92% (95% confidence interval, 81–97%) for the diagnosis of AI (22). On the other hand, no statistically significant difference was found between low-dose and high-dose ACTH stimulation tests (23,24). Low levels for age and sex of dehydroepiandrosterone sulphate (DHEAS) concentration (or less frequently, dehydroepiandrosterone, DHEA) represents an additional marker to increase the level of suspicion of PAI, but it is not per se diagnostic. A CRH test may also be used, but is less common and has limited availability, while a prolonged ACTH stimulation test is rarely required other than to distinguish secondary or tertiary deficiency (Table 4).

 

The insulin-induced hypoglycemia or insulin tolerance test (ITT), also previously known as the gold standard test, is performed by injecting 0.1 or 0.15u/kg of short-acting insulin intravenously after overnight fast, followed by serial measurements of venous glucose and cortisol over 2 hours. Importantly, this procedure must only be used if there is adequate supervision and experience. However, the ITT is beneficial in assessing growth hormone (GH) response simultaneously in patients with suspected co-existing deficiency in secondary AI, in which GH may not exceed 3-5μg/dL. It must be emphasized that all normative values quoted are assay-dependent, and using immunoassays or liquid chromatography with tandem mass spectrometry (LC-MS/MS) may result in a lower cut-off than the historic value of 18μg/dL derived from polyclonal antibody assay (25). Further investigations such as imaging studies, auto-antibodies, or microbiological screening should be arranged accordingly to identify the underlying cause of AI.

 

Table 4. Diagnostic Tests Used to Diagnose and Differentiate AI

Test/ procedure

Interpretation of the result/ comments

Cortisol physiologic response

Adrenal testing

Morning serum cortisol levels at 8-9am in combination with plasma ACTH

·   8-9am cortisol levels < 3μg/dL (80nmol/L) suggest AI (26).

·   In recent onset central AI within 4-6 weeks or severe stress such as sepsis, a ‘normal’ level may still indicate AI.

·   ACTH levels > 300ng/L (66pmol/L) or > 2-fold the ULN confirms PAI.

·       Cortisol levels > 14.5μg/dL (400nmol/L) indicates normal HPA axis (27).

·       Morning cortisol levels in early postoperative pituitary surgery higher than 10µg/dL (275nmol/L) are a predictor of corticotroph reserve (28,29).

SST or cosyntropin test or ACTH test; sampling at 8-9am for cortisol and ACTH level following by 250μg Synacthen for adults, children ≥ 2y of age (15μg/ kg for infants, 125μg for children < 2y of age) ACTH i.v. or i.m.; collect samples at 0, 30 and 60min for cortisol levels.

·       Peak cortisol levels < 18μg/dL indicate AI (depending on assay) (26).

·       Indicated when morning cortisol levels 3-15μg/dL.

·       Recent-onset central AI may produce a normal response.

·       SST can be performed at any time of the day but testing for cortisol levels should be collected at least 18–24 h after the last HC dose or longer for GCs.

·       Peak cortisol levels > 18µg/dL (430-500nmol/L) at 30 or 60 minutes (depending on assay).

·       Pregnancy: higher diagnostic cortisol cut-offs of 25μg/dL (700 nmol/L), 29μg/dL (800 nmol/L), and 32μg/dL (900nmol/L) for the first, second, and third trimesters, respectively (30).

Low-dose SST; 1μg ACTH i.v.; collect samples at 0, 30min for cortisol levels (31).

·       Peak cortisol levels < 18μg/dL indicate AI (depending on assay).

·       Indicated when suspected recent-onset central AI or a shortage of Synacthen itself.

·       Insufficient response to low-dose SST should be considered for other dynamic tests.

·       Peak cortisol level > 18µg/dL (500nmol/L)

Pituitary testing

CRH stimulation test; 1µg/kg or 100µg ovine or human CRH i.v.; collect samples at -5, -1, 0, 15, 30, 60, 90, and 120min for cortisol and ACTH levels (32).

·       Central AI demonstrates low ACTH levels that do not respond to CRH.

·       PAI shows high ACTH levels that rise after administration of CRH.

·       Limited use due to wide variation of responses.

·       Peak ACTH response should be 2-4 folds above baseline at 15 or 30min.

·       Peak cortisol level > 20µg/dL between 30 and 60min or incremental cortisol > 10µg/dL above baseline (33).

Prolonged ACTH stimulation test;

(A) 8-h protocol - 1mg depot Synacthen i.m. or 250μg cosyntropin i.v. infusion over 8h; collect serum samples hourly for cortisol and additional samples at 0, 1, 7, 8h for plasma ACTH levels.

(B) 24-h protocol - cosyntropin 250μg i.v. infusion over 24h on 2 or 3 consecutive days; take blood sample for 9am cortisol and ACTH level; then blood sample for serum cortisol levels at 30min, 60min, 120min, 4h, 8h, 12h and 24h.

·       Central AI illustrates a delayed response and typically has a much greater value at 24 and 48 hours than at 4 hours.

·       PAI shows no response at either time.

·       Useful in differentiating primary from central AI when ACTH level is equivocal

·       Helpful in detecting more subtle degrees of AI than standard SST.

A: serum cortisol levels > 20μg/dL (550nmol/L) at 30min, 60min; 25μg/dL (695nmol/L) at 6-8h after initiation of infusion (34,35).

B: serum cortisol levels at 4h > 36μg/dL (1000nmol/L) with no further increase beyond this time (36).

Hypothalamic testing

ITT; insulin 0.1-0.15 U/kg i.v. is administrated after overnight to achieve symptomatic hypoglycemia and blood glucose level lower than 40mg/dL (<2.2mmol/L); collect blood samples at -15, 0, 15, 30, 45, 60, 90 and 120min for glucose, cortisol, ACTH.

Repeat insulin dose if blood glucose does not fall to level of less than 40mg/dL at 45 min

·       AI is confirmed when a fall of glucose to less than 40mg/dL with corresponding an inability to demonstrate a cortisol response higher than 18μg/dL (37). Contraindications in patients with

basal cortisol < 3μg/dL, untreated hypothyroidism, electrocardiographic evidence or history of ischemic heart disease, or seizure.

·       Useful in diagnosis of coexisting GH deficiency.

Peak cortisol levels > 18μg/dL (500nmol/L).

Overnight metyrapone test; 30mg/kg (max 3g) metyrapone is given at midnight; serum cortisol, 11-deoxycortisol, and ACTH levels are measured at 8am the following day.

·       Alternative test when ITT is contraindicated.

·       Test is valid only when cortisol levels fall to lower than 10μg/dL.

·      Peak ACTH response > 200ng/L

·      11-deoxycortisol level > 7mg/dL

·      Sum of cortisol and 11-deoxycortisol should exceed 16.5μg/dL (38).

17-OHCS, 17-hydroxycorticosteroids; ACTH, adrenocorticophic hormone; AI, adrenal insufficiency; CBG, cortisol-binding globulin; CRH, corticotrophin releasing hormone; HC, hydrocortisone; h-CRH, human CRH; GCs, glucocorticoids; GH, growth hormone; i.m., intramuscular; ITT, insulin tolerance test; i.v., intravenous; MCs, mineralocorticoids; PAI, primary AI; SST, short Synacthen test; ULN, upper limit of normal.

 

The notion of Critical Illness-Related Corticosteroid Insufficiency (CIRCI) emerged to describe impairment of the HPA axis during critical illness that is characterized by the dysregulated systemic inflammation caused by the inadequate intracellular GC-mediated anti-inflammatory activity. The Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) have updated the guideline for the diagnosis and management of CIRCI, which was originally proposed in 2008 (39). The task force makes no recommendation on whether to use delta cortisol after SST or random plasma cortisol levels of less than 10μg/dL to diagnose CIRCI. However, some cohort studies found that patients with CIRCI had poorer outcomes than patients without CIRCI when total cortisol levels are less than 10μg/dL or delta cortisol after SST < 9μg/dL. This evidence may be helpful in deciding upon replacement treatment when CIRCI is suspected (Table 5) (40,41). Other diagnostic tests, such as salivary cortisol or serum ACTH, are not recommended to diagnose this condition. Additionally, markers of inflammation and coagulation, morbidity, length of intensive care unit (ICU) stay, and mortality should be taken in consideration. Nevertheless, this whole concept has been questioned, especially since cortisol-binding globulin and albumin are invariably decreased in severe critical illness, implying that simple measurement of total cortisol is likely to be an inaccurate reflection of the true state of the HPA axis. However, the guideline cautions against using plasma free cortisol levels rather than plasma total cortisol levels to diagnose CIRCI. Most recent evidence suggests that CIRCI is not a useful diagnostic or therapeutic description, and that during the acute phase of the response to severe sepsis there is no failure of the HPA axis and no need for corticosteroid therapy, although in the recovery phase this may become important.

 

Table 5. Tests Used in Adrenal Insufficiency in Critical Illness (but see text above)

Test

Indicators for poorer outcome

Cortisol

Random levels < 10μg/dL (275nmol/L)

SST

Delta peak/basal levels < 9μg/dL (250nmol/L)

SST: short Synacthen test

 

In the context of the different diseases associated with AI, additional investigations may be necessary (Table 4). CT scanning of adrenals should be performed to identify infectious diseases such as tuberculosis, tumors, or adrenal hemorrhage.

 

Table 6. Additional Studies Used in Patients with AI

Primary AI

Specific tests for autoimmune antibodies

·       Autoantibodies against CYP21A2 for the vast majority of autoimmune PAI.

·       Other antibodies against 17-hydroxylase and side-chain-cleavage enzyme are also identified.

·       It should be note that tests for autoantibodies are not standardized.

Other autoimmune markers and hormonal assays for evidence of APS

·       Autoantibodies against IFNa and IFNw for APS-1.

·       Serum calcium and PTH for hypoparathyroidism.

·       Autoantibodies for autoimmune thyroid disease and thyroid function test.

·       Autoimmune for insulin, GAD65, ICA, and ZnT8 along with blood glucose for autoimmune diabetes.

·       Antibody against parietal cell with or without intrinsic factor antibody for pernicious anemia.

·       Antibodies to tissue transglutaminase and anti-endomysial IgA antibody for celiac disease.

·       Liver function along with ANA, SMA, and anti-LKM1 antibodies for autoimmune hepatitis in those with abnormal liver biochemical tests.

Microbial and serological tests

·       Tuberculosis (tuberculin testing, early morning urine samples cultured for Mycobacterium tuberculosis)

·       Other infective cause.

CT / MRI scan

·       Calcified adrenals can be found in infection, hemorrhage, and malignancy.

·       Large adrenals with or without calcification can be seen in metastatic deposits and early phase of infection.

·       Small atrophic glands with calcified foci are commonly illustrated in chronic stage of infection.

·       Adrenal hemorrhage shrinks and attenuates gradually on CT and has a variable appearance on MRI depending on the stage of blood products.

·       Adrenal venous thrombosis, in turn raises internal pressure, resulting in the same hemorrhagic finding.

Chest radiograph

·       Clue for pulmonary manifestation of tuberculosis or fungal infection.

CT guided adrenal biopsy

·       Useful in confirming the diagnosis of adrenal metastasis from extra-adrenal malignancy, or certain infiltrations such as histoplasmosis.

Adrenoleukodystrophy

·       Circulating levels of VLCFA.

17-OH progesterone and 24-hour urine steroid profile

·       Classic CAH.

Secondary AI

Pituitary hormonal assessment

·       Pituitary hormone producing tumor or co-existing pituitary hormone deficiency.

Pituitary MRI scans

·       Pituitary or parasellar lesion.

Biopsy of pituitary

·       Occasionally necessary such as hypophysitis.

Other Investigations

Measurement of plasma renin and aldosterone in PAI to determine MCs reserve

·       High renin levels in combination with an inappropriately normal or low aldosterone concentration suggests PAI.

·       In early phase of evolving PAI, MCs deficiency may predominate and may be the only sign.

Measurement of serum DHEAS

·       Decreased DHEAS levels in women in both PAI (direct effect on adrenal cortex) and central AI (decrease in ACTH stimulus).

AI, adrenal insufficiency; ANA, antinuclear antibodies; anti-LKM, anti-liver microsomal; APS, autoimmune polyglandular syndrome; CAH, congenital adrenal hyperplasia; CT, computerized tomography; DHEAS, dehydroepiandrosterone sulphate; MRI, magnetic resonance imaging; SMA, anti-smooth muscle antibodies; VLCFA, very long-chain fatty acids.

 

As previously stated, laboratory tests should be interpreted with caution in specific situations, such as those affecting CBG concentration. Low CBG levels are commonly found in conditions characterized by inflammation, nephrotic syndrome, liver disease, the immediate postoperative period or requiring intensive care, or rare genetic disorders, whereas estrogen, pregnancy and mitotane can raise CBG levels. Systemic estrogen-containing drug prescriptions should be discontinued at least four weeks prior to testing. However, using an estrogen patch has no effect on CBG levels. Different diagnostic criteria should be considered according to the cortisol assay. If patients are currently on GCs replacement, omitting the steroid dose before testing is advised. The evening and morning dose of HC or prednisolone should be omitted, whereas the duration of drug withdrawal in patients taking other synthetic GCs may be longer. In pregnancy, higher diagnostic cortisol cut-offs of 25μg/dL (700nmol/L), 29μg/dL (800nmol/L), and 32μg/dL (900nmol/L) should be considered for the first, second, and third trimesters, respectively (30). In pituitary diseases, testing of GCs reserve is suggested before and after initiation of GH replacement or upon documentation of an unexplained improvement in co-existing diabetes insipidus (DI).

 

THERAPY

 

Acute Adrenal Insufficiency (Adrenal Crisis)

 

Adrenal insufficiency is a potentially life-threatening medical emergency when presented as an adrenal crisis, which requires prompt treatment with HC and fluid replacement. When clinical suspicion exists, treatment should be initiated without any delay while awaiting definitive proof of diagnosis. Blood samples should be obtained for later cortisol concentration measurements, and the management approach should be similar to that of any critically ill patient (Table 7, 8).

 

Table 7. Treatment of Acute Adrenal Insufficiency (Adrenal Crisis) - Management During Resuscitation of Critically Ill Patient.

Maintain airway and breathing.

·       Correct hypovolemia and reverse electrolyte abnormalities; caution should be taken in correcting chronic hyponatremia (not more than 12mmol in 24h, preferably < 8mmol) to prevent central pontine myelinolysis.

·       Replace glucocorticoid; clinical improvement especially blood pressure should be seen within 4-6h.

·       The half-life of HC is 90min after i.v. injection, and more prolonged after i.m. administration; switch to oral HC 40mg in the morning and 20mg in the afternoon if oral intake is resumed; taper to a standard dose of 10-20mg on awakening and 5-10mg in the early afternoon if there is no other major illness.

·       Some experts recommend dexamethasone while dynamic tests are awaited, as dexamethasone does not interfere with the assay, but HC is preferred for its MCs activity.

·       Regarding the electrolyte imbalances, no need for fludrocortisone replacement in an acute crisis since the MCs activity of HC and 0.9% NaCl infusion is sufficient.

Establish i.v. access with two large bore cannulas.

Collect venous blood samples for urea and electrolytes, glucose, full blood count, bicarbonate, infection screen, and store samples (plasma cortisol and ACTH measurement). Do not wait for blood results.

Rapid infusion of 1L isotonic saline solution (0.9% NaCl) within the 1st hour, followed by continuous i.v. isotonic saline solution guided by individual patient needs; usually infuse 2-3 L of normal saline solution within 12 hours; after this, fluid management should be guided by volume status, urine output and biochemical results; 50g/L (5%) dextrose in saline solution if there is evidence of hypoglycemia.

Inject i.v. 100mg of HC immediately (50-100mg/m2 for children) and then followed by 200mg/day (50–100mg/m2/d for children divided q 6h) of HC (via continuous i.v. therapy or 6-8 hourly i.v. injection) for 24h, reduce to HC 100mg/day on the following day; i.m. administration should be used if venous access is not possible; prednisolone may be used as an alternative drug if unavailable HC; dexamethasone is the least preferred and should be given only if no other glucocorticoid is available.

Use additional supportive measures as needed

For hypoglycemia

·       Dextrose 0.5-1g/kg of dextrose or 2-4mL/kg of D25W should be infused slowly at rate of 2-3mL/min; standard initial glucose dose is 25g.

Cardiac monitoring.

MCs replacement is not required if the HC dose exceeds 50mg/day.

ACTH, adrenocorticotropic hormone; HC, hydrocortisone; i.m., intramuscular; i.v., intravenous; MCs, mineralocorticoid.

 

Table 8. Treatment of Acute Adrenal Insufficiency (Adrenal Crisis) - After Patient Stabilization

Continue i.v. 0.9%NaCl; rate may be slower and maintained for 24-48h.

Search for and treat possible infectious precipitating causes of adrenal crisis; treat any associated condition(s).

Perform SST to confirm the diagnosis.

Differential diagnosis if needed.

Taper parenteral glucocorticoids over 1-3 days, depending on precipitating illness.

After the first 24 hours, HC dose can be reduced to 50mg q 6h and switched to oral HC 40mg in the morning and 20mg in the afternoon, then tapered to a standard dose of 10mg on awakening, 5mg at lunchtime and 5-10mg in the early afternoon.

In aldosterone deficiency, begin MCs replacement with fludrocortisone (100μg by mouth daily) when saline infusion ceased to prevent sodium loss, intravascular volume depletion and hyperkaliemia.

However, MCs replacement is not required if the HC dose exceeds 50mg/day.

ACTH, adrenocorticopic hormone; HC, hydrocortisone; i.m., intramuscular; i.v., intravenous; MCs, mineralocorticoids; SST, short Synacthen test.

 

Management of Chronic or Insidious Onset of Adrenal Insufficiency

The aim of replacement treatment in AI is to mimic the normal cortisol secretion rate, which is around 5-8mg/m2/day(42,43). Previously this rate was thought to be approximately 25-30mg/day of HC, but normal cortisol production rates seem to be about 8-15mg/day. Most patients can cope with less than 30mg/day (usually 15-25mg/day in divided doses). Doses are usually given upon waking with a smaller dose at lunchtime and then one in late afternoon. Weight-adjusted dosing may be associated with a better safety profile. Despite the various types of cortisol replacement regimens, no head–to-head comparison data is available to advocate one over the other. Decisions regarding the form and dose of GCs replacement therapy are based on crude end-points such as weight, well-being, and blood pressure,as well as on local availability, cost and clinical need (Table 9). Bone mineral density may be reduced on conventional doses of 30mg/day HC, highlighting the importance of aiming for effective but safe doses. Long-duration GCs can be administered once daily but may be associated with higher risk of side effects. Patients should be monitored for clinical symptoms.

 

Table 9. Glucocorticoid Replacement Schemes

Drug profile

Commonly used doses

Immediate-release HC (Hydrocortisone)

Short acting, given in 2-3 divided doses; this biologically active GCs approximately mimics the endogenous diurnal rhythm; obese individuals may require more GCs replacement than lean individuals; higher frequency regimes and size-based dosing may be beneficial in individual cases; high doses in the evening may disturb sleep and alter metabolism.

15-25 mg or 5-8 mg/m2/day; the highest dose in the morning on

awakening, the next in the early afternoon (2h after lunch) (2-dose regime) or at lunch and afternoon but not later than 4-6h before bedtime) (3-dose

regime); usually 10mg upon awakening, 5mg at lunchtime and 5mg in the late afternoon.

Dual-release HC (combination of immediate-release HC in the

outer-layer coat and extended-release core, PlenadrenÒ) (44,45).

Given once daily in the morning; resulting in higher morning and lower evening cortisol levels with no overnight cortisol rise. This may be advantageous in patients with high risk of metabolic comorbidities and in patients with poor administrative compliance.

20-30mg; lower dose may be sufficient in patients with some remaining endogenous cortisol production; identical total daily dose may be given when switching and clinical response needs to be monitored due to lower bioavailability than HC.

Modified-release HC

(delayed and sustained release in multiple microcrystals with polymer sheathing,

ChronocortÒ) (46).

Given twice daily as a “toothbrush regime”, with 2/3 of the total daily dose before bedtime (11p.m.) and 1/3 administered in the morning (7a.m.), resulting in overnight rise with morning peak of cortisol and near physiological cortisol levels throughout the day. This is beneficial for CAH patients since it prevents the ACTH-driven excess production of adrenal androgens.

Usual dose of HC; then titrated based on symptoms and 17OHP along with androstenedione measurement. (Phase 3 study)

Cortisone acetate (47).

Short acting but longer than HC; the peak of serum cortisol level is delayed compared to HC since the oral form requires a conversion to cortisol at liver to become active; available in oral preparation only.

20-35mg in 2-3 divided doses.

Prednisolone /prednisone

Long-acting, once-daily dose is sufficient; some may need additional 2.5mg in the evening; does not mimic diurnal rhythm of endogenous cortisol; better choice in patients with poor administrative compliance or in patients with poor quality of life on HC replacement; prednisone must be processed in liver to become prednisolone which is then able to cross the cellular membrane; cross-reaction occurs in most cortisol assays.

3-5mg once daily on waking.

Dexamethasone

Inter-individual variable metabolism makes it difficult to predict the adequate dose; dose needs to be titrated if patient is on hepatic enzyme inducing medications; it is not recommended in PAI because of risk of Cushingoid side effects; concurrent fludrocortisone replacement is necessary in PAI patients if dexamethasone is undeniable.

0.25-0.75mg once daily.

MCs replacement

(Fludrocortisone) (48).

Required only in PAI; doses may need to temporary increase by 50-100% in hot weather or conditions that promote excessive sweating; available in oral preparation only; if parenteral action required, use DOCA if available.

50-200μg (median 100μg) once daily in the morning with starting dose at 50-100μg.

Androgen replacement (DHEA) (49,50).

A trial of replacement can be offered to PAI women with low energy, low mood, and low libido, despite otherwise optimized GCs and MCs replacement; some evidence of benefit; not generally available on prescription, but obtained as a ‘health food’ supplement.

25–50mg as a single oral dose in the morning; replacement should be discontinued if no clinical benefit after initial 6-month trial.

AI, adrenal insufficiency; DHEA, dehydroepiandrosterone; DOCA, deoxycorticosterone acetate; GCs, glucocorticoids; i.m., intramuscular; i.v., intravenous; HC, hydrocortisone; MCs, mineralocorticoids; PAI, primary adrenal insufficiency.

 

It should be noted that while HC and prednisolone are active GCs, cortisone acetate and prednisone require activation via hepatic 11β-hydroxysteroid dehydrogenase type 1 to become biologically active.

 

Another novel aspect on the management of the chronic AI is the development in glucocorticoid formulation. A dual-release HC preparation that can be administered once daily upon awakening, Plenadren® (developed by Duocort, Viropharma, Shire Pharmaceuticals), which was approved for treatment of AI in adults since 2011. Moreover, another delayed- and sustained-release preparation of HC, ChronocortÒ, given twice daily before bedtime and in the morning, is currently undergoing the approval process for treatment of CAH patients (Table 9). Advances in understanding of the normal cortisol circadian rhythm in the setting of endogenous clock genes, as well as its importance in controlling innate immunity, metabolism, and stress may imply that some of these GCs replacement formulas will be superior over the traditional ones in the future.

 

One important aspect of the management of chronic PAI is patient and family education. Careful instructions should be given to up-titrate the daily dose in the event of intercurrent febrile illness, accident, or severe mental stress, such as an important examination (Table 10). If the patient is vomiting and cannot take medication by mouth, parenteral HC must be given urgently. Ideally, patients should wear a ‘medical-alert’ bracelet or necklace and carry the Emergency Medical Information Card, which should provide information on the patient’s diagnosis, prescribed medications and daily doses, and the physician involved in the patient’s care. Patients should also have supplies of HC for emergencies, and should be educated about how and when to administer them by the subcutaneous route. Rectal suppositories of prednisolone 100 mg, enemas of prednisolone 20mg/100mL, or HC acetate enema 10% have also been used, but they are clearly ineffective when diarrhea is present.

 

Table 10. Glucocorticoid Replacement Schemes During Illness

 

Examples of commonly used schemes

During minor illness

 

Increase dose by 2-3 times of usual

dosage for 3 days; do not change MC dose.

HC 25-75mg/day twice daily per oral, then taper rapidly to maintenance dose as patient recovers (usually 1-2 days); HC 50mg/m2/day i.m. or double/triple HC replacement doses in children.

During minor-to-moderate surgery

HC 50-100mg/day twice daily per oral or i.v., then taper rapidly to maintenance dose as patient recovers.

During major illness

Increase dose up to 10 times of usual dosage with continuous infusion of HC, or equivalent dexamethasone dosage, then decrease dose by half per day, to usual maintenance dose. Half dose 2nd postoperative day, maintenance dose 3rd postoperative day.

HC 100mg i.v., followed by continuous i.v. infusion of 200mg HC in 24h, then taper rapidly by half per day regarding course of illness; alternative administration of HC 50mg iv. or i.m. q 6h may be considered.

Major surgery with general anesthesia

HC 100mg i.v. just before induction of anesthesia, followed by continuous i.v. infusion of 200mg HC in 24h (alternatively 50mg every 6h i.v. or i.m.); then taper rapidly by half each day and switch to oral regime depending on clinical state; children: HC 50mg/m2 i.v. followed by HC 50–100mg/m2/d divided q 6h. Weight-appropriate continuous i.v. fluids with 5% dextrose and 0.2 or 0.45% NaCl.

Uncomplicated, outpatient dental procedures under local anesthesia and most radiologic studies

No extra supplementation is required.

Severe stress or trauma

Inject prefilled dexamethasone (4-mg) syringe.

Moderately stressful procedures (barium enema, endoscopy, or arteriography)

Extra supplementation is required only before the procedure.

100mg i.v. dose of HC just before the procedure

Pregnancy

HC should be increased 5-10mg/day in the last trimester (20–40% from the 24th week onward) to mimic physiologic increase in free cortisol; fludrocortisone dose adjustment may need in response to serum sodium and potassium levels; HC is preferred over the other GCs, whereas dexamethasone is warned as a contraindication since it is not inactivated in the placenta and can transfer to the fetus.

·       Last trimester: 5-10mg/day of HC usually (20–40% from the 24th week onward)

·       Laboratory: HC stress dosing with HC 100mg i.v. bolus, followed by continuous i.v. infusion of 200mg HC in 24h, and rapidly taper to pre-pregnancy doses after delivery.

HC, hydrocortisone; i.m., intramuscularly; i.v., intravenously; GCs, glucocorticoids

 

Regarding MC replacement therapy, the dose of fludrocortisone is titrated individually based on clinical examination, mainly body weight and blood pressure, and the levels of plasma renin activity (PRA).

 

Hydrocortisone is generally preferred for use in patients with AI because it has both GCs and MCs activity. Patients receiving prednisone or prednisolone, on the other hand, may require additional fludrocortisone due to their relatively low MCs activity. Notably, dexamethasone should not be used to treat PAI patients because its lack of MCs activity (51). After resolution of an adrenal crisis, the adequacy of MC replacement should be assessed by measuring supine and erect blood pressure, electrolytes and PRA. Inadequate fludrocortisone dose may cause postural hypotension with elevated PRA, whereas excessive administration results in the opposite. Mineralocorticoid replacement therapy is frequently neglected in patients with adrenal failure. The dose may be temporarily increased in the summer along with an increase in salt intake, particularly if patients are exposed to temperatures above 30ºC (85ºF) or other conditions causing increased sweating. Newborns and children may also require higher fludrocortisone because MCs sensitivity is lower.

 

In chronic central AI, GCs replacement is similar to that in PAI; however, measurement of plasma ACTH concentrations cannot be used to titrate the optimal GCs dose. Replacement of other anterior pituitary deficits may be also necessary, while changing doses of growth hormone and thyroxine may affect the GCs requirements. More specifically, the HPA axis should be evaluated prior to and following the initiation of GH replacement therapy since GH inhibits the conversion of cortisone to cortisol. Consequently, patients receiving GCs replacement may require higher doses once GH treatment is introduced (18,52).

 

For patients with either primary or central AI, the beneficial effects of adrenal androgen replacement therapy with 25 to 50mg/day of DHEA have been reported. To date, the reported benefit is principally confined to female patients and includes improvement in sexual function and well-being, but the effects are variable.

 

In children with PAI, HC in three or four divided doses with total starting daily dose of 8mg/m2 body surface area are preferred over synthetic long-acting GCs, such as prednisolone or dexamethasone. In the case of documented aldosterone deficiency, fludrocortisone treatment with a typical dosage of 100μg/day without body surface area adjustment is suggested, while sodium chloride supplements are needed in the newborn period until the age of 12 months. Most experience has been gained from the treatment of children with CAH; however, since in this case under treatment is confirmed by hyperandrogenism higher GCs therapy is usually given (22).

 

Other therapies have been studied such as rituximab with or without depot tetracosactide in newly diagnosed autoimmune PAI patients (53). The regenerative potential of adrenocortical stem cells combined with immunomodulatory treatment to stop the autoimmune destruction, adrenal transplantation, or gene therapy in forms of monogenic PAI may be a useful option in the future.

 

For CIRCI with septic shock, it has been suggested to use i.v. HC < 400mg/day for at least 3 days at full dose (rather than high-dose and short-course regimes) when they are not responsive to fluid and moderate- to high-dose vasopressor therapy (> 0.1μg/kg/min of norepinephrine or equivalent) (39). In hospitalized adults with community-acquired pneumonia a daily dose of < 400mg i.v. HC or equivalent has been suggested. GCs are also suggested in patients suffering from meningitis, cardiac arrest (methylprednisolone given during resuscitation or HC given for post-resuscitation shock), and cardiopulmonary bypass surgery (CPB) (250mg i.v. of methylprednisolone at anesthesia induction and at onset of CPB) or dexamethasone (1mg/kg perioperatively). Corticosteroids are not suggested for patients after major trauma or suffering from influenza. Nevertheless, it should be emphasized that this is a rapidly changing situation, and the use of corticosteroids in the ICU with severely ill patients remains controversial.

 

Despite concerns that patients with AI were more vulnerable to infection during the recent COVID-19 pandemic,adequately treated and well-trained AI patients demonstrated the same incidence of COVID-19-suggestive symptoms and disease severity as the people without AI.

 

FOLLOW-UP

 

Patients with chronic AI should be closely followed-up by an endocrinologist or a healthcare provider with endocrine expertise at least annually (for infants every 3 to 4 months) to ensure the adequacy of their GCs and/or MCsreplacement dose (Table 11, 12), to reduce the risk of adrenal crisis, to provide necessary education to patients, and to confirm that they have a prompt updated emergency pack with HC to treat an emergency. Clinical symptoms including body weight, postural blood pressure and energy levels should be monitored in order to avoid signs of frank GCs excess and adjust accordingly the dose of steroids. The ‘mapping’ of the dose has been suggested to assess the compliance and also help decide when tablets should be taken in problematic cases of reduced quality of life where the detailed questioning for daily habits, working patterns, general feelings of energy, mental concentration, daytime somnolence, and energy dips. Hormonal monitoring of GCs replacement, such as serum or salivary cortisol ‘day-curves’ is not routinely recommended, but can be useful in specific situations where the clinical response cannot be reliably used to adjust treatment or when malabsorption is suspected (54,55). In addition, the use of ACTH levels to assess GCs replacement is not suggested since it leads to over-replacement.

 

Similarly, MCs replacement should be monitored based on clinical assessment, which includes postural hypotension as measured by lying and standing blood pressure and pulse, symptoms of salt craving, oedema, blood electrolyte measurements. Reported well-being, normal blood pressure without orthostatic hypotension and electrolytes within the normal range indicate adequate replacement. PRA in the upper reference range has been found to be a useful predictor for a correct MCs dose (56,57). Liquorice and grapefruit juice enhance the MC effect of HC and should be avoided. Phenytoin potentiates hepatic clearance of GCs and increase fludrocortisone metabolism; therefore, and higher doses are needed when it is co-administered.

 

It is of note that in patients who develop hypertension while receiving fludrocortisone, the dose of fludrocortisone should be reduced and HC replacement should be adjusted. If blood pressure remains uncontrolled, anti-hypertensive treatment should be initiated without fludrocortisone discontinuation. First-line anti-hypertensive drugs to be selected are the angiotensin II receptor blockers or angiotensin-converting enzyme blockers to counterbalance the vasoconstrictive effects of elevated angiotensin II, second-line a dihydropyridine calcium blocker, while diuretics should be avoided and aldosterone receptor blockers are contraindicated.

 

Since AI might mask the presence of partial DI, urine output should be monitored after starting GCs replacement, or conversely, when DI has unreasonably improved, patients have to be tested for HPA reserve. With DHEA replacement, morning serum DHEAS levels should be measured before the intake of the daily dose aiming at the mid-normal range. An annual screening for autoimmune diseases, which includes autoimmune thyroid disease, type 1 diabetes, premature ovarian failure, coeliac disease, and autoimmune gastritis should be performed in PAI patients without other obvious cause of adrenal failure.

 

Special populations, like pregnant females, should be surveyed for clinical symptoms and signs of GCs over- and under-replacement including weight gain, fatigue, postural hypotension or hypertension, hyperglycemia, with at least once per trimester. Only sodium and potassium can be reliably monitored in blood and urine for the adequacy of replacement. In contrast, PRA monitoring is not advised since plasma renin physiologically increases during this time.

In children, monitoring of GCs replacement includes clinical assessment such as growth velocity, body weight, blood pressure, and energy levels (58).

 

Although familial autoimmune PAI is less frequent, genetic counselling is suggested in particular situations due to monogenic disorders.

 

Table 11. Assessment of Glucocorticoid Replacement

Under replacement

Lethargy, tiredness, nausea, poor appetite, weight loss, hyperpigmentation.
Low serum cortisol level on cortisol day curve (useful in specific cases for HC or cortisone replacement only)

Over replacement

Cushingoid appearance, weight gain, insomnia, peripheral edema, low bone mineral density.
High 24-h UFC, high serum cortisol on hydrocortisone day curve (useful in specific cases for HC or cortisone replacement only)

 

 

Table 12. Assessment of Mineralocorticoid Replacement

Inadequate replacement

Postural hypotension, light-headedness
High PRA (plasma renin activity, should be at the upper limit of normal)

Over replacement      

Hypertension, peripheral oedema, hypernatremia, hypokalemia

 

Patient Education

 

This is very crucial in the management of AI and for the prevention of adrenal crisis. All patients and their relatives should be educated about their condition and the emergency measures they should take at home to avoid crises, particularly concerning GCs adjustments in stressful events and preventive strategies including parenteral self- or lay-administration of emergency GCs particularly in situations of intercurrent illness, fever, or any type of stress. This information should be reinforced during annual follow-up visits by clinicians and if possible, through a structured patient education program. All patients should be given a steroid emergency card and medical-alert identification to inform health personnel of the need for increased GCs doses to avert or immediately treat adrenal crisis. Every patient should be equipped with a GCs injection kit for emergency use and be educated on how to use it (Table 13).

 

Table 13: Information and Equipment for Patients with AI

Steroid Sick Day Rules

Sick day rule 1: Patients should be advised to increase the oral GCs when the patient experiences fever or illness requiring bed rest, or when requiring antibiotics for an infection. For instance, the total oral GCs dose should be doubled (> 38°C) or tripled (> 39°C) for at least 72 hours; if the patient remains unwell after 72 hours, they should contact their caring physician. There is no need to increase the MCs dose.

 

Sick day rule 2: There should always be a supply of additional oral GCs on prescription for sick days and HC emergency injection kit prescription.

 

Sick day rule 3: Every patient should carry a medical alert bracelet or leaflet with information stating their conditions, treatment, physician, emergency phone number of endocrine specialist team, and proposed GCs regimen during adrenal crisis.

 

Sick day rule 4: Parenteral injection of GCs preparation, either i.m. or i.v. should be provided in case of severe illness, trauma, persistent vomiting, before moderately stressful procedure (i.e., barium enema endoscopy, arteriography), or before surgical intervention.

 

Steroid Emergency Pack

·                Every patient should be provided with this pack to keep at home.

·                The pack contains a vial of 100mg HC or 4mg dexamethasone, syringes, needles, also oral HC or prednisolone suppositories.

·                The patient and/ or any responsible family member should be educated to administer this medication i.m. or s.c. during an emergency situation including severe accident, significant hemorrhage, fracture, unconsciousness, diarrhea and vomiting, and they should call the emergency medical personal immediately (adults, i.m. or s.c. HC 100mg; children, i.m. HC 50mg/m2; infants, 25mg; school-age children, 50mg; adolescents, 100mg).

·                The expiry date on the pack should be checked regularly and replaced with a new pack if expired.

·                The patient should be advised to take the pack when travelling.

Medical-Alert bracelet or pendant and emergency steroid card

·                Every patient should wear or carry these in which the diagnosis and daily medication should be clearly documented.

Follow up

·                A regular visit in order to reinforce education and confirm understanding should be scheduled at least annually in a patient without specific problems or recent crises. Other circumstances, such as monitoring during infancy, may necessitate more frequent visits.

GCs, glucocorticoids; HC, hydrocortisone; i.m., intramuscularly; i.v., intravenously; HC, hydrocortisone; MCs, mineralocorticoids; s.c., subcutaneously

 

PROGNOSIS

 

The mortality of patients with PAI was increased in some studies and adrenal crisis was a significant cause of death, emphasizing the importance of educating patients with AI to prevent crises. Recent large cohorts confirmed the increased mortality of patients with ΑΙ, especially PAI. Although cardiovascular disease (CVD) was the leading cause of death, infectious diseases were found to pose the greatest risk in comparison to controls. Adrenal crisis was also found to be a common contributor, particularly in those with co-existing CVD. In addition, despite an adequate replacement dose, the quality of life of PAI patients remains impaired. This appears to be related to the delay in diagnosis.

 

GUIDELINES

 

Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, Husebye ES, Merke DP, Murad MH, Stratakis CA, Torpy DJ. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016 Feb;101(2):364-89.

 

Fleseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, Samuels MH. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016 Nov;101(11):3888-3921.

 

Annane D, Pastores SM, Rochwerg B, Arlt W, Balk RA, Beishuizen A, Briegel J, Carcillo J, Christ-Crain M, Cooper MS, Marik PE, Umberto Meduri G, Olsen KM, Rodgers SC, Russell JA, Van den Berghe G. Guidelines for the Diagnosis and Management of Critical Illness-Related Corticosteroid Insufficiency (CIRCI) in Critically Ill Patients (Part I): Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) 2017. Crit Care Med. 2017 Dec;45(12):2078-2088.

 

Pastores SM, Annane D, Rochwerg B. Corticosteroid Guideline Task Force of SCCM and ESICM. Guidelines for the Diagnosis and Management of Critical Illness-Related Corticosteroid Insufficiency (CIRCI) in Critically Ill Patients (Part II): Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) 2017. Crit Care Med. 2018 Jan;46(1):146-148.

 

Speiser PW, Arlt W, Auchus RJ, Baskin LS, Conway GS, Merke DP, Meyer-Bahlburg HFL, Miller WL, Muraf 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;103(11):4043-4088.

 

REFERENCES

 

  1. Mason AS, Meade TW, Lee JA, Morris JN. Epidemiological and clinical picture of Addison's disease. Lancet 1968;2:744-747
  2. Olafsson AS, Sigurjonsdottir HA. Increasing Prevalence of Addison Disease: Results from a Nationwide Study. Endocr Pract 2016;22:30-35
  3. Erichsen MM, Lovas K, Skinningsrud B, Wolff AB, Undlien DE, Svartberg J, Fougner KJ, Berg TJ, Bollerslev J, Mella B, Carlson JA, Erlich H, Husebye ES. Clinical, immunological, and genetic features of autoimmune primary adrenal insufficiency: observations from a Norwegian registry. J Clin Endocrinol Metab 2009;94:4882-4890
  4. Chantzichristos D, Persson A, Eliasson B, Miftaraj M, Franzen S, Svensson AM, Johannsson G. Incidence, prevalence and seasonal onset variation of Addison's disease among persons with type 1 diabetes mellitus: nationwide, matched cohort studies. Eur J Endocrinol 2018;178:113-120
  5. Bates AS, Van't Hoff W, Jones PJ, Clayton RN. The effect of hypopituitarism on life expectancy. J Clin Endocrinol Metab 1996;81:1169-1172
  6. Nilsson B, Gustavasson-Kadaka E, Bengtsson BA, Jonsson B. Pituitary adenomas in Sweden between 1958 and 1991: incidence, survival, and mortality. J Clin Endocrinol Metab 2000;85:1420-1425
  7. Regal M, Paramo C, Sierra SM, Garcia-Mayor RV. Prevalence and incidence of hypopituitarism in an adult Caucasian population in northwestern Spain. Clin Endocrinol (Oxf) 2001;55:735-740
  8. Hahner S, Spinnler C, Fassnacht M, Burger-Stritt S, Lang K, Milovanovic D, Beuschlein F, Willenberg HS, Quinkler M, Allolio B. High incidence of adrenal crisis in educated patients with chronic adrenal insufficiency: a prospective study. J Clin Endocrinol Metab 2015;100:407-416
  9. Quinkler M, Murray RD, Zhang P, Marelli C, Petermann R, Isidori AM, Ekman B. Characterization of patients with adrenal insufficiency and frequent adrenal crises. Eur J Endocrinol 2021;184:761-771
  10. Reisch N, Willige M, Kohn D, Schwarz HP, Allolio B, Reincke M, Quinkler M, Hahner S, Beuschlein F. Frequency and causes of adrenal crises over lifetime in patients with 21-hydroxylase deficiency. Eur J Endocrinol 2012;167:35-42
  11. Achermann JC, Meeks JJ, Jameson JL. Phenotypic spectrum of mutations in DAX-1 and SF-1. Mol Cell Endocrinol 2001;185:17-25
  12. Aguisanda F, Thorne N, Zheng W. Targeting Wolman Disease and Cholesteryl Ester Storage Disease: Disease Pathogenesis and Therapeutic Development. Curr Chem Genom Transl Med 2017;11:1-18
  13. Fluck CE. MECHANISMS IN ENDOCRINOLOGY: Update on pathogenesis of primary adrenal insufficiency: beyond steroid enzyme deficiency and autoimmune adrenal destruction. Eur J Endocrinol 2017;177:R99-R111
  14. Prasad R, Hadjidemetriou I, Maharaj A, Meimaridou E, Buonocore F, Saleem M, Hurcombe J, Bierzynska A, Barbagelata E, Bergada I, Cassinelli H, Das U, Krone R, Hacihamdioglu B, Sari E, Yesilkaya E, Storr HL, Clemente M, Fernandez-Cancio M, Camats N, Ram N, Achermann JC, Van Veldhoven PP, Guasti L, Braslavsky D, Guran T, Metherell LA. Sphingosine-1-phosphate lyase mutations cause primary adrenal insufficiency and steroid-resistant nephrotic syndrome. J Clin Invest 2017;127:942-953
  15. Buonocore F, Achermann JC. Primary adrenal insufficiency: New genetic causes and their long-term consequences. Clin Endocrinol (Oxf) 2020;92:11-20
  16. Turk BR, Theda C, Fatemi A, Moser AB. X-linked adrenoleukodystrophy: Pathology, pathophysiology, diagnostic testing, newborn screening and therapies. Int J Dev Neurosci 2020;80:52-72
  17. Narumi S. Discovery of MIRAGE syndrome. Pediatr Int 2022;64:e15283
  18. Fleseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, Samuels MH. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2016;101:3888-3921
  19. Patti G, Guzzeti C, Di Iorgi N, Maria Allegri AE, Napoli F, Loche S, Maghnie M. Central adrenal insufficiency in children and adolescents. Best Pract Res Clin Endocrinol Metab 2018;32:425-444
  20. Angelousi A, Alexandraki K, Tsoli M, Kaltsas G, Kassi E. Hypophysitis (Including IgG4 and Immunotherapy). Neuroendocrinology 2020;110:822-835
  21. Angelousi A, Alexandraki KI, Kyriakopoulos G, Tsoli M, Thomas D, Kaltsas G, Grossman A. Neoplastic metastases to the endocrine glands. Endocr Relat Cancer 2020;27:R1-R20
  22. Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, Husebye ES, Merke DP, Murad MH, Stratakis CA, Torpy DJ. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2016;101:364-389
  23. Ambrosi B, Barbetta L, Re T, Passini E, Faglia G. The one microgram adrenocorticotropin test in the assessment of hypothalamic-pituitary-adrenal function. Eur J Endocrinol 1998;139:575-579
  24. Mayenknecht J, Diederich S, Bahr V, Plockinger U, Oelkers W. Comparison of low and high dose corticotropin stimulation tests in patients with pituitary disease. J Clin Endocrinol Metab 1998;83:1558-1562
  25. Javorsky BR, Raff H, Carroll TB, Algeciras-Schimnich A, Singh RJ, Colon-Franco JM, Findling JW. New Cutoffs for the Biochemical Diagnosis of Adrenal Insufficiency after ACTH Stimulation using Specific Cortisol Assays. J Endocr Soc 2021;5:bvab022
  26. Dorin RI, Qualls CR, Crapo LM. Diagnosis of adrenal insufficiency. Ann Intern Med 2003;139:194-204
  27. Hagg E, Asplund K, Lithner F. Value of basal plasma cortisol assays in the assessment of pituitary-adrenal insufficiency. Clin Endocrinol (Oxf) 1987;26:221-226
  28. Watts NB, Tindall GT. Rapid assessment of corticotropin reserve after pituitary surgery. JAMA 1988;259:708-711
  29. Auchus RJ, Shewbridge RK, Shepherd MD. Which patients benefit from provocative adrenal testing after transsphenoidal pituitary surgery? Clin Endocrinol (Oxf) 1997;46:21-27
  30. Lebbe M, Arlt W. What is the best diagnostic and therapeutic management strategy for an Addison patient during pregnancy? Clin Endocrinol (Oxf) 2013;78:497-502
  31. Dickstein G, Shechner C, Nicholson WE, Rosner I, Shen-Orr Z, Adawi F, Lahav M. Adrenocorticotropin stimulation test: effects of basal cortisol level, time of day, and suggested new sensitive low dose test. J Clin Endocrinol Metab 1991;72:773-778
  32. Schlaghecke R, Kornely E, Santen RT, Ridderskamp P. The effect of long-term glucocorticoid therapy on pituitary-adrenal responses to exogenous corticotropin-releasing hormone. N Engl J Med 1992;326:226-230
  33. Trainer PJ, Faria M, Newell-Price J, Browne P, Kopelman P, Coy DH, Besser GM, Grossman AB. A comparison of the effects of human and ovine corticotropin-releasing hormone on the pituitary-adrenal axis. J Clin Endocrinol Metab 1995;80:412-417
  34. Nye EJ, Grice JE, Hockings GI, Strakosch CR, Crosbie GV, Walters MM, Jackson RV. Comparison of adrenocorticotropin (ACTH) stimulation tests and insulin hypoglycemia in normal humans: low dose, standard high dose, and 8-hour ACTH-(1-24) infusion tests. J Clin Endocrinol Metab 1999;84:3648-3655
  35. Nye EJ, Grice JE, Hockings GI, Strakosch CR, Crosbie GV, Walters MM, Torpy DJ, Jackson RV. Adrenocorticotropin stimulation tests in patients with hypothalamic-pituitary disease: low dose, standard high dose and 8-h infusion tests. Clin Endocrinol (Oxf) 2001;55:625-633
  36. Stewart PM. N-PJ. The Adrenal Cortex. In: Melmed S. PK, Larsen PR., Kronenberg HM., ed. Williams Textbook of Endocrinology. 13th ed: Elsevier; 2016:489-555.
  37. Berg C, Meinel T, Lahner H, Mann K, Petersenn S. Recovery of pituitary function in the late-postoperative phase after pituitary surgery: results of dynamic testing in patients with pituitary disease by insulin tolerance test 3 and 12 months after surgery. Eur J Endocrinol 2010;162:853-859
  38. Andrioli M, Pecori Giraldi F, Cavagnini F. Isolated corticotrophin deficiency. Pituitary 2006;9:289-295
  39. Annane D, Pastores SM, Rochwerg B, Arlt W, Balk RA, Beishuizen A, Briegel J, Carcillo J, Christ-Crain M, Cooper MS, Marik PE, Umberto Meduri G, Olsen KM, Rodgers SC, Russell JA, Van den Berghe G. Guidelines for the Diagnosis and Management of Critical Illness-Related Corticosteroid Insufficiency (CIRCI) in Critically Ill Patients (Part I): Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) 2017. Crit Care Med 2017;45:2078-2088
  40. Marik PE, Zaloga GP. Adrenal insufficiency during septic shock. Crit Care Med 2003;31:141-145
  41. de Jong MF, Molenaar N, Beishuizen A, Groeneveld AB. Diminished adrenal sensitivity to endogenous and exogenous adrenocorticotropic hormone in critical illness: a prospective cohort study. Crit Care 2015;19:1
  42. Esteban NV, Loughlin T, Yergey AL, Zawadzki JK, Booth JD, Winterer JC, Loriaux DL. Daily cortisol production rate in man determined by stable isotope dilution/mass spectrometry. J Clin Endocrinol Metab 1991;72:39-45
  43. Kerrigan JR, Veldhuis JD, Leyo SA, Iranmanesh A, Rogol AD. Estimation of daily cortisol production and clearance rates in normal pubertal males by deconvolution analysis. J Clin Endocrinol Metab 1993;76:1505-1510
  44. Quinkler M, Miodini Nilsen R, Zopf K, Ventz M, Oksnes M. Modified-release hydrocortisone decreases BMI and HbA1c in patients with primary and secondary adrenal insufficiency. Eur J Endocrinol 2015;172:619-626
  45. Isidori AM, Venneri MA, Graziadio C, Simeoli C, Fiore D, Hasenmajer V, Sbardella E, Gianfrilli D, Pozza C, Pasqualetti P, Morrone S, Santoni A, Naro F, Colao A, Pivonello R, Lenzi A. Effect of once-daily, modified-release hydrocortisone versus standard glucocorticoid therapy on metabolism and innate immunity in patients with adrenal insufficiency (DREAM): a single-blind, randomised controlled trial. Lancet Diabetes Endocrinol 2018;6:173-185
  46. Whitaker M, Debono M, Huatan H, Merke D, Arlt W, Ross RJ. An oral multiparticulate, modified-release, hydrocortisone replacement therapy that provides physiological cortisol exposure. Clin Endocrinol (Oxf) 2014;80:554-561
  47. Fariss BL, Hane S, Shinsako J, Forsham PH. Comparison of absorption of cortisone acetate and hydrocortisone hemisuccinate. J Clin Endocrinol Metab 1978;47:1137-1140
  48. Williams GH, Cain JP, Dluhy RG, Underwood RH. Studies of the control of plasma aldosterone concentration in normal man. I. Response to posture, acute and chronic volume depletion, and sodium loading. J Clin Invest 1972;51:1731-1742
  49. Arlt W, Callies F, van Vlijmen JC, Koehler I, Reincke M, Bidlingmaier M, Huebler D, Oettel M, Ernst M, Schulte HM, Allolio B. Dehydroepiandrosterone replacement in women with adrenal insufficiency. N Engl J Med 1999;341:1013-1020
  50. Alkatib AA, Cosma M, Elamin MB, Erickson D, Swiglo BA, Erwin PJ, Montori VM. A systematic review and meta-analysis of randomized placebo-controlled trials of DHEA treatment effects on quality of life in women with adrenal insufficiency. J Clin Endocrinol Metab 2009;94:3676-3681
  51. Liu D, Ahmet A, Ward L, Krishnamoorthy P, Mandelcorn ED, Leigh R, Brown JP, Cohen A, Kim H. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol 2013;9:30
  52. Alexandraki KI, Grossman A. Management of Hypopituitarism. J Clin Med 2019;8
  53. Napier C, Gan EH, Mitchell AL, Gilligan LC, Rees DA, Moran C, Chatterjee K, Vaidya B, James RA, Mamoojee Y, Ashwell S, Arlt W, Pearce SHS. Residual Adrenal Function in Autoimmune Addison's Disease-Effect of Dual Therapy With Rituximab and Depot Tetracosactide. J Clin Endocrinol Metab 2020;105
  54. Howlett TA. An assessment of optimal hydrocortisone replacement therapy. Clin Endocrinol (Oxf) 1997;46:263-268
  55. Bancos I, Hahner S, Tomlinson J, Arlt W. Diagnosis and management of adrenal insufficiency. Lancet Diabetes Endocrinol 2015;3:216-226
  56. Oelkers W, L'Age M. Control of mineralocorticoid substitution in Addison's disease by plasma renin measurement. Klin Wochenschr 1976;54:607-612
  57. Flad TM, Conway JD, Cunningham SK, McKenna TJ. The role of plasma renin activity in evaluating the adequacy of mineralocorticoid replacement in primary adrenal insufficiency. Clin Endocrinol (Oxf) 1996;45:529-534
  58. 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

 

Congenital Adrenal Hyperplasia

ABSTRACT

 

Congenital Adrenal Hyperplasia (CAH) is a term used to describe a group of genetically determined disorders of defective steroidogenesis that result in variable deficiency of the end products cortisol and/or aldosterone and their deleterious, including life-threatening, effects on metabolism and electrolytes with simultaneous diversion to the accumulation of androgens and their virilizing effects. Although we discuss the various enzymatic defects that are involved, we focus on the most common enzyme deficiency, 21-hydroxylase. Depending on the residual enzymatic activity governed by the genetic mutation, 21-hydroxylase deficiency CAH is classified as either classical (salt wasting or simple virilizing) or non-classical.  In classical 21-hydroxylase deficiency CAH, there is an accumulation of 17-hydroxyprogesterone which is shunted into the intact androgen pathway and may lead to prenatally virilized external genitalia in females as early as 9 weeks of gestation.  Inadequately treated patients may develop progressive penile or clitoral enlargement, premature adrenarche, precocious puberty, rapid linear growth accompanied by premature epiphysis maturation leading to compromised final adult height and impaired fertility. Moreover, inadequately treated salt loss increases the risk for adrenal crises. In contrast, over treatment with cortisol results in “Cushingoid” effects including retarded bone development. We describe the various defects, their manifestations and goals for therapy, and emerging newer therapies for CAH to both correct the deficiency in cortisol and aldosterone secretion while suppressing overproduction of ACTH. 

 

INTRODUCTION

 

Congenital adrenal hyperplasia (CAH) refers to a group of 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 five 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 (1).

 

EPIDEMIOLOGY

 

Data from close to 6.5 million newborn screenings worldwide indicate that classical CAH occurs in about 1:13,000 to 1:15,000 live births (2). It is estimated that 75% of patients have the salt-wasting phenotype(1). Non-classical 21OHD CAH (NC21OHD) is more common. The incidence in the heterogeneous population of New York City is about 1 in 100, making NC21OHD the most frequent autosomal recessive disorder in humans. NC21OHD is particularly prevalent in certain populations, showing a high ethnic specificity. In the Ashkenazi Jewish population, 1 in 3 are carriers of the allele, and 1 in 27 are affected with the disorder (3-5).  CAH resulting from 11β-hydroxylase deficiency (11β-OHD) is the second most common cause of CAH, accounting for 5-8% of all cases (6). It occurs in about 1 of every 100,000 live births in the general population (7)  and is more common in some populations of North African origin (8). In Moroccan Jews, for example, the disease incidence was initially estimated to be 1 in 5,000 live births (9); subsequently, it was shown to occur less frequently (10), but remains more common than in other populations. The other forms of CAH are considered rare diseases and their 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. The adrenal gland architecture suggests that the adrenal acts as three separate glands: zona glomerulosa, zona fasciculate, zona reticularis (11). The hypothalamic-pituitary-adrenal feedback system is mediated through the circulating level of plasma cortisol by negative feedback of cortisol on CRF and ACTH secretion. (Figure 2) Therefore, any CAH condition that results in 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) a build-up of precursor molecules in pathways blocked by the enzyme deficiency.

Figure 1. The classical and backdoor pathways of adrenal steroidogenesis: The classical pathway is highlighted in blue and the backdoor pathway is highlighted in orange. In the classical pathway, five enzymatic steps are necessary for cortisol production. In the first step of adrenal steroidogenesis, cholesterol enters mitochondria via a carrier protein called steroidogenic acute regulatory protein (StAR). ACTH stimulates cholesterol cleavage, the first and rate limiting step of adrenal steroidogenesis. The five enzymes required for cortisol production are cholesterol side chain cleavage enzyme (SCC), 17α-hydroxylase, 3β-hydroxysteroid dehydrogenase (3βHSD2), 21-hydroxylase, and 11β-hydroxylase. The backdoor pathway is an alternative pathway producing dihydrotestosterone. The enzymes include 5α-reductase 1, aldo keto reductases, retinol dehydrogenase RoDH, 17β-hydroxysteroid dehydrogenases, 17α-hydroxylase.

Figure 2. The hypothalamic-pituitary-adrenal (HPA) axis. Corticotropin-releasing factor produced in the hypothalamus stimulates the secretion of adrenocorticotropic hormone (ACTH) from the anterior lobe of the pituitary gland. ACTH stimulates the production of cortisol and androgens in the adrenal cortex. There is negative feedback on the hypothalamus and pituitary gland by cortisol.

 

The clinical symptoms of the five different forms of CAH result from the specific hormones that are deficient and those that are produced in excess as outlined in Table 1. In the most common form 21OHD-CAH, the function of 21-hydroxylating cytochrome P450 is deficient, creating a block in the P450 cortisol production pathway. This leads to an accumulation of 17-hydroxyprogesterone (17-OHP), a precursor of the 21-hydroxylation step. Excess 17-OHP is then shunted into the intact androgen pathway, where the 17,20-lyase enzyme converts 17-OHP to Δ4-androstenedione, the major adrenal androgen. Mineralocorticoid (aldosterone) deficiency is a feature of the most severe form of the disease and hence named salt wasting CAH. The enzyme defect in the non-classical form of 21OHD CAH is mild and salt wasting does not occur. The analogy of all other enzyme deficiencies in terms of precursor retention and product deficiencies are shown in Table 1.

Table 1. Summary of the Clinical, Hormonal, and Genetic Features of Steroidogenic Defects (1)

Condition

Onset

Abnormality

Genitalia

Mineralocorticoid Effect

Typical Features

Gene

Lipoid CAH

Congenital

StAR Protein

Female, with no sexual development

Salt wasting

All steroid products low

StAR 8p11.2

Lipoid CAH

Congenital

P450scc

Female, with no sexual development

Salt wasting

All steroid products low

CYP11A 15q23-24

3β-HSD deficiency, classic

Congenital

3β-HSD

Females virilized, males under-virilized

Salt wasting

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

HSD3B2 1p13.1

3β-HSD deficiency, non-classic

Postnatal

3β-HSD

Normal genitalia with mild to moderate hyperandrogenism postnatally

None

Elevated DHEA, 17-pregnenolone, low androstenedione, testosterone

Absent or unknown

17α-OH deficiency

Congenital

P450c17

Variable sexual development

Hypokalemic low-renin hypertension

Normal or decreased androgens and estrogen, elevated DOC, corticosterone

CYP17 10q24.3

17,20-Lyase deficiency

Congenital

P450c17

Infantile female genitalia

None

Decreased androgens and estrogens

CYP17 10q24.3

Combined 17α-OH/17,20-lyase deficiency

Congenital

P450c17

Infantile female genitalia

Hypokalemic low-renin hypertension

Decreased androgens and estrogens

CYP17 10q24.3

Combined 17α-OH/17,20-lyase deficiency

Postnatal

P450c17

Infertility, Infantile female genitalia

None

Decreased follicular estradiol and increased progesterone

CYP17 10q24.3

Classic 21-OH deficiency, salt wasting

Congenital

P450c21

Females prenatally virilized, normal male genitalia, hyperpigmentation

Salt wasting

Elevated 17-OHP, DHEA, and androstenedione, elevated K, low Na, CO2

CYP21 6p21.3

Classic 21-OH deficiency, simple virilizing

Congenital

P450c21

Females prenatally virilized, normal male genitalia

None

Elevated 17-OHP, DHEA, and androstenedione, normal electrolytes

CYP21 6p21.3

Non-classic 21-OH deficiency

Postnatal

P450c21

Males and females with normal genitalia at birth, hyperandrogenism postnatally

None

Elevated 17-OHP, DHEA, and androstenedione on ACTH stimulation

CYP21 6p21.3

Classic CAH 11β-deficiency

Congenital

P450c11B1

Females virilized with atypical genitalia, males unchanged

Low-renin hypertension

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

CYP11B1 8q24.3

Non-classic CAH 11β-deficiency

Postnatal

P450c11B1

Males and females with normal genitalia at birth, hyperandrogenism postnatally

None

Elevated 11-deoxycortisol ± DOC, elevated androgens

CYP11B1 8q24.3

P450 Oxido-Reductase Deficiency

Congenital

POR

Females virilized with atypical genitalia, males under-virilized

None

Partial, combined and variable defects of P450c21, P450c17 and P450aro activity

POR

7q11.2

 

 

 

CAH, Congenital adrenal hyperplasia; DHEA, dehydroepiandrosterone; DOC, deoxycorticosterone; 3β-HSD, 3β-hydroxysteroid dehydrogenase; OH, hydroxylase; 17-OHP, 17-hydroxyprogesterone. Adapted from: Wajnrajch MP and New MI. Chapter 103: Defects of Adrenal Steroidogenesis. Endocrinology, Adult and Pediatric. 6th Edition. 2010. pp 1897-1920. Permission obtained.

 

CLINICAL FEATURES

 

External Genitalia

 

VIRILIZING FORMS OF CAH: CLASSICAL 21-HYROXYLASE DEFICIENCY AND 11β-HYDOXYLASE DEFICIENCY

 

Females with classical CAH owing to 21OHD and 11β-hydroxylase deficiency generally present at birth with virilization of their genitalia. Adrenocortical function begins at the 7th week of gestation (12); thus, a female fetus with classical CAH is exposed to adrenal androgens at the critical time of sexual differentiation (between 9 to 15 weeks gestational age). Androgens masculinize external female genitalia causing physical changes including: 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. The degree of genital virilization may range from mild clitoral enlargement alone to, in rare cases, a penile urethra (Prader V genitalia). Degrees of genital virilization are classified into five Prader stage (13) (see Figure 3).

 

Figure 3. Different degrees of virilization according to the scale developed by Prader (15). 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).

 

Internal Genitalia

 

In contrast to the virilization of the external genitalia, internal female genitalia, the uterus, fallopian tubes and ovaries, develop normally. Females with CAH do not produce anti-Müllerian hormone (AMH), which is produced by the testicular Sertoli cells. Internal female structures are Müllerian derivatives and are not androgen responsive. Therefore, the affected female born with virilized external genitalia but normal female internal genitalia has the possibility of normal fertility (1).  Surgical correction of the external genitalia (genitoplasty) may be considered in severely virilized females. This is further discussed in a later section, Treatments.

 

Postnatal Effects and Growth

 

Deficient 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 (pubarche), axillary hair, acne, and impaired fertility. Advanced somatic and epiphyseal development occurs with rapid growth during childhood. This rapid linear growth is usually accompanied by premature epiphyseal maturation and closure, resulting in a final adult height that is below that expected from parental heights (on average -1.1 to -1.5 SD below the mid-parental target height) (14). This is on average 10 cm below the mid-parental height (15). On the other hand, poor growth can occur in patients with 21OHD as a result of excess glucocorticoid treatment. Short stature occurs even in patients with good hormonal adrenal control. A study of growth hormone therapy alone or in combination with a GnRH analog and aromatase inhibitors in CAH patients with compromised height prediction showed improvement in short- and long-term growth to reduce the height deficit (15).  Aromatase inhibitor as a sole adjunct treatment reduces bone age advancement without adverse effects on bone mineral density or visceral adipose tissue (16).

 

Puberty

 

In the majority of patients treated adequately from early life, the onset of puberty in both girls and boys with classical 21OHD occurs at the expected chronological age. A careful study showed that the mean ages at onset of puberty in both males and females were somewhat younger than the general population, but did not differ significantly among the three forms of 21OHD (17).

 

In those who are inadequately treated, advanced epiphyseal development can lead to central precocious puberty. In those with advanced bone maturation at the initial presentation, such as in simple virilizing males, the exposure to elevated androgens followed by the suddenly decreased androgen levels after initiation of glucocorticoid treatment may cause an early activation of the hypothalamic-pituitary-gonadal axis. Studies suggest that excess adrenal androgens (aromatized to estrogens) inhibit the pubertal pattern of gonadotropin secretion by the hypothalamic-pituitary axis. This inhibition, via a negative feedback effect, can be reversed by glucocorticoid treatment (17, 18).

 

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 (17). In adolescents and adults, signs of hyperandrogenism may include male-pattern alopecia (temporal balding) and acne. Female patients may develop hirsutism and menstrual irregularities. Although the expected age of menarche may be delayed in females with classical CAH (18), when adequately treated many have regular menses after menarche (19, 20). Menstrual irregularity and secondary amenorrhea with or without hirsutism occur in a subset of post-menarchal females, especially those in poor hormonal control. Primary amenorrhea or delayed menarche may occur if a female with classical CAH is untreated, inadequately treated, or over treated with glucocorticoids (20). In addition, women with CAH may develop polycystic ovarian syndrome (PCOS), likely as a complication of poorly controlled CAH (21, 22).

 

Gender Role Behavior and Cognition

 

Prenatal androgen exposure in females affected with the classic forms of 21OHD CAH not only has a masculinizing effect on the development of the external genitalia, but also on childhood behavior. Both physical and behavioral masculinization were related to genotype, indicating that behavioral masculinization in childhood is a consequence of prenatal androgen exposure. Further, changes in childhood play behavior is correlated with reduced female gender satisfaction. Prenatal androgen exposure is related to a decrease in self-reported femininity in dose response manner in adulthood (23) . Affected adult females are more likely to have gender dysphoria, and experience less heterosexual interest and reduced satisfaction with the assignment to the female sex (24). In contrast to females, males affected with CAH do not show a general alteration in childhood play behavior, core gender identity and sexual orientation (24). The rates of bisexual and homosexual orientation were increased in women with all forms of 21OHD CAH. They were found to correlate with the degree of prenatal androgenization (24). Of interest, bisexual/homosexual orientation was correlated with global measures of masculinization of nonsexual behavior and predicted independently by the degree of both prenatal androgenization and masculinization of childhood behavior (25). Among 46,XX CAH patients raised as girls, reported rates of gender dysphoria varied from 6.3% to 27.2%(26).  Most expressed gender dysphoria in late adolescence and adulthood.  Cultural views may determine serious biases in reports of gender dysphoria coming from countries with disproportional societal advantages for males and/or religious restrictions.  Male gender raised patients were approximately 10% of CAH cohorts, mostly from underdeveloped countries, with a high proportion of late diagnoses (26).With regards to cognitive abilities, such as visuospatial/motor ability and handedness, the effect of prenatal androgen exposure continues to be elucidated.  A study found males and females with CAH scored higher than their siblings of the same sex in measures of visual special processing suggesting that androgens affect spatial ability (27).  The effect of CAH on intelligence is controversial.  One study showed no evidence of intellectual deficit in either females or males with CAH. Intelligence was not significantly associated with disease characteristics (28).

 

Fertility

 

Difficulty with fertility in females with CAH may arise for various reasons, including anovulation, secondary polycystic ovarian syndrome, irregular menses, non-suppressible serum progesterone levels, or an inadequate introitus. Fertility is reduced in salt-wasting 21OHD (29). In a retrospective survey of fertility rates in a large group of females with classical CAH, simple virilizers were shown to be more likely to become pregnant and carry the pregnancy to term than salt-wasters. Adequate glucocorticoid therapy is an important variable with respect to fertility outcome. The development of PCOS in CAH patients is not uncommon and may be related to both prenatal and postnatal excess androgen exposure, which can affect the hypothalamic-pituitary-gonadal axis. An inadequate vaginal introitus can affect up to a third of classical CAH adult females. Since vaginal dilation is needed to maintain good patency, vaginoplasty is delayed until sexual intercourse is regular or when the patient can assume responsibility for vaginal dilatation (30).

 

Males with CAH, particularly if poorly treated, may have reduced sperm counts and low testosterone as a result of small testes due to suppression of gonadotropins and sometimes intra-testicular adrenal rests(31, 32). All of these complications may result in diminished fertility. In male patients with classical CAH, several long-term studies indicate that those who have been adequately treated undergo normal pubertal development, have normal testicular function, and normal spermatogenesis and fertility (32, 33). However, small testes and aspermia can occur in patients as a result of inadequately controlled disease (34, 35). Testicular adrenal rest tumor can lead to end stage damage of testicular parenchyma, most probably as a result of longstanding obstruction of the seminiferous tubules (36). In contrast, some investigators have reported normal testicular maturation as well as normal spermatogenesis and fertility in patients who had never received glucocorticoid treatment (37).

 

Studies demonstrate that post pubertal males with inadequately treated CAH are at a very high risk to develop  testicular adrenal rest tumors (TARTs) In one study, almost all these patients were found to have adenomatous adrenal rests within the testicular tissue, as indicated by the presence of specific 11β-hydroxylated steroids in the blood from gonadal veins (38). These tumors have been reported to be ACTH dependent and to regress following adequate steroid therapy (39-43). These testicular adrenal rests are more frequent in males with salt-wasting CAH and are associated with an increased risk of infertility(30, 44) . Regular testicular examination and periodic testicular ultrasonography are recommended for early detection of testicular lesions. If present, dexamethasone treatment can be considered to suppress TARTs. 

 

Salt-Wasting 21-Hydroxylase Deficiency

 

When the deficiency of 21-hydroxylase is severe, adrenal aldosterone secretion is not sufficient for sodium reabsorption by the distal renal tubules, and individuals suffer from salt wasting in addition to 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. The salt wasting is presumed to result from inadequate secretion of salt-retaining steroids, primarily aldosterone. In addition, hormonal precursors of the 21-OH enzyme may act as antagonists to mineralocorticoid action in the sodium-conserving mechanism of the immature newborn renal tubule (45-47).

 

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. They may be discharged from the hospital after birth without diagnosis and experience a salt-wasting crisis at home. On the other hand, salt wasting females are born with atypical genitalia that trigger the diagnostic process and appropriate treatment. It is important to recognize that the extent of genital virilization may not differ among the two forms of classical CAH, the simple virilizing and the salt-wasting form. Thus, even a mildly virilized newborn with 21OHD should be observed carefully for signs of a potentially life-threatening crisis within the first few weeks of life. The difference between salt-wasting and simple virilizing form of 21OHD is the quantitative difference in activity of the 21-hydrozylase enzyme, which results from specific mutations. In vitro expression studies show that as little as 1% of 21-hydroxylase activity is sufficient to synthesize enough aldosterone to prevent significant salt wasting (48). It has been observed that an aldosterone biosynthetic defect apparent in infancy may ameliorate with age (49, 50).A spontaneous partial recovery of aldosterone biosynthesis in an adult patient with a homozygous deletion of the CYP21A2 gene who had documented severe salt wasting in infancy has been reported (51). Therefore, it is desirable to follow the sodium and mineralocorticoid requirements carefully by measuring plasma renin activity (PRA) in patients who have been diagnosed in the neonatal period as salt wasters.

 

Simple-Virilizing 21-Hydroxylase Deficiency

 

The salient features of classical simple virilizing 21OHD 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 usually no evidence of mineralocorticoid deficiency in this disorder.

 

Diagnosis at birth of a female with simple virilizing CAH is usually made immediately because of the apparent genital ambiguity. Since the external genitalia are not affected in newborn males, hyperpigmentation and an enlarged phallus may be the only clues suggesting increased ACTH secretion and cortisol deficiency. Diagnosis at birth in males thus rests on prenatal or newborn screening. If a female is not treated with glucocorticoid replacement therapy early post-natally, her genitalia may continue to virilize due to continued excess adrenal androgens, and pseudo precocious puberty may occur. In patients with salt-wasting 21OHD, signs of hyperandrogenism in children affected with CAH include early onset of facial, axillary, and pubic hair, adult body odor, and rapid somatic growth and bone age advancement, leading to short stature in adulthood. The same issues as discussed above related to puberty, fertility, behavior and cognition apply to patients with simple-virilizing 21OHD (1).

 

Non-Classical 21-Hydroxylase Deficiency

 

Non-classical 21OHD (NC-21OHD), previously known as late-onset 21OHD, is much more common than the classical form, with an incidence as high as 1:27 in Ashkenazi Jews (3). Individuals with the non-classical (NC) form of 21OHD 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. This form of CAH results from a mild deficiency of the 21-hydroxylase enzyme. Table 2 summarizes the main clinical characteristics of all forms of 21OHD CAH. While serum cortisol concentration is typically low in patients with the classic form of the disease, it is usually normal in patients with NC 21OHD. Similar to classical CAH, NC-CAH may cause premature development of pubic hair, advanced bone age and accelerated linear growth velocity in both males and females. Severe cystic acne has also been attributed to NC-CAH (52, 53).

 

Table 2. Clinical Features in Individuals with Classic and Non-Classic 21-Hydroxylase Deficiency in the Untreated Form

Feature

21-OH Deficiency

Classic

Non-Classic

Prenatal virilization

Females only

Absent

Postnatal virilization (hyperandrogenism)

Females and Males

Typical

Salt wasting

~75% of all individuals

Absent

 

Women may present with a variety of symptoms of androgen excess which may be highly variable and organ-specific, including hirsutism, temporal baldness, acne and infertility. Menarche in females may be normal or delayed, and secondary amenorrhea is a frequent occurrence. Further masculinization may include hirsutism, male habitus, deepening of the voice, or male-pattern alopecia (temporal recession). Polycystic ovarian syndrome may also be seen as a secondary complication in these patients. Possible reasons for the development of PCOS include reprogramming of the hypothalamic-pituitary-gonadal axis from prenatal exposure to androgens, or chronic levels of excess adrenal androgens that disrupt gonadotropin release and have direct effects on the ovary, ultimately leading to the formation of cysts. Because of the overlap of hyperandrogenic symptoms, it is important to consider NC 21OHD in a patient diagnosed with PCOS (54, 55).

 

In adult males, early balding, acne, or impaired fertility may prompt the diagnosis of NC-CAH. A highly reliable constellation of physical signs of adrenal androgen excess is the presence of pubic hair, enlarged phallus, and relatively small testes. Males may have small testes compared to the phallus, which results from suppression of the hypothalamic-pituitary-gonadal axis from adrenal androgens. They may also develop TARTs, which can cause infertility, although some untreated men have been fertile(31, 33). Signs of NC-CAH in adult males may be limited to short stature, oligo-zoospermia and impaired fertility.

 

A subset of individuals with NC-21OHD are completely asymptomatic when detected (usually as part of a family study or evaluation for infertility), but it is thought, based on longitudinal follow-up of such patients, that symptoms of hyperandrogenism may wax and wane with time. The presence of 21OHD can also be discovered during the evaluation of an incidental adrenal mass (56). One study showed that an increased incidence of adrenal incidentalomas has been found, which was reported as high as 82% in patients with 21OHD and up to 45% in subjects heterozygous for 21OHD mutations. This probably arises from hyperplastic tissue areas and does not require surgical intervention (57). Overall, however, CAH is an uncommon cause of incidentalomas, accounting for less than 1% in one series (58, 59).

 

OTHER FORMS OF CONGENITAL ADRENAL HYPERPLASIA

 

11-β Hydroxylase Deficiency

 

Virilization and low renin hypertension are the prominent clinical features of 11β hydroxylase deficiency (11β-OHD) (60). The virilizing signs and symptoms of this disorder are similar to or more severe than classical 21OHD. Despite failure of aldosterone production, overproduction of deoxycorticosterone (DOC), in vivo a less potent mineralocorticoid, causes salt retention and hypertension. Elevated blood pressure is usually not identified until later in childhood or in adolescence, although its appearance in an infant 3 months of age has been documented (61). In addition, hypertension correlates variably with biochemical values, and clinical signs of mineralocorticoid excess and the degree of virilization are not well correlated. Some severely virilized females are normotensive, whereas mildly virilized patients may experience severe hypertension leading to fatal vascular accidents (62, 63). Complications of long-standing uncontrolled hypertension, including cardiomyopathy, retinal vein occlusion and blindness have been reported in 11β-OHD patients (64, 65). Potassium depletion develops concomitantly with sodium retention, but hypokalemia is variable. Renin production is suppressed secondary to mineralocorticoid-induced sodium retention and volume expansion.

 

A mild non-classical form of 11β-OHD CAH has been reported. Unlike the common non-classical form of 21OHD, this form is very rare. Non-classical 11β-OHD has been diagnosed in normotensive children with mild virilization or precocious pubarche (6) and in adults with signs of hyper-androgen effect (66) as well as a woman with infertility (67)(69). Despite a hormonal profile consistent with 11β-OHD, mutations in the CYP11B1 gene may not always be present (66). The best biochemical marker of 11β-OHD is elevated serum 11-deoxycortisol concentration.

 

3-β Hydroxysteroid Dehydrogenase Deficiency

 

There are two forms of the 3 β -hydroxysteroid dehydrogenase enzyme (3 β -HSD): type I and type II. Type II 3 β -HSD enzyme is expressed in the adrenal cortex and gonads and is responsible for conversion of Δ5 (delta 5) to Δ4 (delta 4) steroids (1). This enzyme is essential for the formation of progesterone, which is the precursor for aldosterone, and 17-OHP, which is the precursor for cortisol in the adrenal cortex as well as for androstenedione, testosterone, and estrogen in the adrenal cortex and gonads (68, 69). Therefore, deficiency of 3ß-HSD in the classic form of 3ß-HSD deficiency CAH results in insufficient cortisol synthesis, salt-wasting in the most severe form, and virilization of external genitalia in females due to androgen effect from the peripheral conversion of circulating Δ5 precursors to active Δ4 steroids. Simultaneous type II 3ß-HSD deficiency in the gonads results in incomplete virilization of the external genitalia in males. Thus, genital ambiguity can result in both sexes (70).

 

17 α -Hydroxylase/17,20 Lyase Deficiency

 

Steroid 17 α-hydroxylase/17,20 lyase deficiency accounts for approximately 1% of all CAH cases and affects steroid synthesis in both the adrenals and gonads(71). Patients have impaired cortisol synthesis, leading to ACTH over secretion, which increases serum levels of deoxycorticosterone and especially corticosterone, resulting in low renin hypertension, hypokalemia, and metabolic alkalosis. Affected females are born with normal external genitalia, however affected males are born with under-virilized genitalia due to their deficient gonadal testosterone production. 17 α-Hydroxylase/17,20 lyase deficiency is often recognized at puberty in female patients who fail to develop secondary sex characteristics (72).

 

Congenital Lipoid Adrenal Hyperplasia

 

Congenital lipoid adrenal hyperplasia is an extremely rare and severe form of CAH which is caused by mutations in the steroidogenic acute regulatory protein (StAR). Both the adrenal glands and the gonads exhibit a severe defect in the conversion of cholesterol to pregnenolone (73, 74). More specifically, StAR mediates the acute steroidogenic response by moving cholesterol from the outer to inner mitochondrial membrane (the rate-limiting step of steroidogenesis), and when this does not occur, cholesterol and cholesterol esters accumulate (75). In the most severe form, males with congenital lipoid hyperplasia are born with female-appearing external genitalia. Females have a normal genital phenotype at birth but remain sexually infantile without treatment. Salt wasting occurs in both males and females. If not detected and treated, the severe form of lipoid CAH is usually fatal (76).  Several cases have been reported that demonstrate that lipoid CAH has a spectrum of clinical presentation, with varying degrees of genital ambiguity (including normal male genitalia in a 46, XY male) and adrenal insufficiency. Mutations in the StAR protein have been reported that retain partial protein function, leading to variable phenotype (77).

 

The cholesterol side-chain cleavage enzyme (P450scc) is a very slow enzyme located on the inner mitochondrial membrane and catalyzes the conversion of cholesterol to pregnenolone in the first and rate-limiting step in the production of corticosteroids (78).  The CYP11A1 gene encoding P450cc lies on chromosome 15q23-24. Mutations in this gene are rare with approximately 20 patients reported (77-79).

 

Cytochrome P450 OxidoReductase Deficiency

 

Cytochrome P450 oxido-reductase (POR) deficiency is another rare form of CAH that is caused by a mutation on 7q11.2(80-82) P450 oxidoreductase is an important cofactor for electron transfer from nicotinamide adenine dinucleotide phosphate (NADPH) to several enzymes of steroidogenesis including 21-hydroxylase and 17α-hydroxylase/17,20-lyase. The various constellations of partial enzymatic deficiency of 21-hydroxylase, 17α-hydroxylase/17,20-lyase, and aromatase in the developing fetus account for the broad range of genital anomalies seen in both sexes (80).  Female newborns may have severe virilization, and males may have under-virilized genitalia due to combined partial 21-hydroxylase and 17α-hydroxylase/17,20-lyase deficiencies.  Maternal virilization and low maternal estriol levels are common findings during pregnancies with affected male and female fetuses. Maternal gestational virilization is likely due to the P450 oxidoreductase effect on placental P450 aromatase (82).  Many affected patients also have Antley-Bixler syndrome (type 2) characterized by craniosynostosis, radio-humeral or radioulnar synostosis, arachnodactyly and bowing of the femur (82).  

 

GENETICS

 

In general, all forms of CAH are transmitted in an autosomal recessive mode of inheritance as a monogenic disorder. However, there have been reports of cases where none or only one mutation in the responsible gene was identified, including in cases of 21 OHD CAH (83, 84), deficiency of the cholesterol side chain cleavage enzyme (85) and POR  deficiency (80). The genes responsible for each form of CAH are shown in Table 1.

 

21-Hydroxylase Deficiency

 

The gene encoding the enzyme 21-hydroxylase, CYP21A2, is a microsomal cytochrome P450 located on the short arm of chromosome 6 (86) in the human lymphocyte antigen (HLA) complex (87). CYP21A2 and its homologue, the pseudogene CYP21P, alternate with two genes called C4B and C4A (87, 88) that encode the two isoforms of the fourth component (C4) of serum complement (89). CYP21A2 and CYP21P, which each contain 10 exons, share 98% sequence homology in exons and approximately 96% sequence homology in introns (90, 91).

More than 140 mutations have been described including point mutations, small deletions, small insertions, and complex rearrangements of the gene (92). The most common mutations appear to be the result of either of two types of meiotic recombination events between CYP21 and CYP21P: 1) misalignment and unequal crossing over, resulting in large-scale DNA deletions, and 2) apparent gene conversion events that result in the transfer to CYP21A2 of smaller-scale deleterious mutations present in the CYP21P pseudogene (1, 93).

 

Both classical and non-classical 21-hydroxylase deficiency are inherited in a recessive manner as allelic variants of the CYP21A2 gene. Classical 21-hydroxylase deficiency tends to result from the presence of two severely affected alleles and non-classical 21-hydroxylase deficiency tends to result from the presence of either two mild 21-hydroxylase deficiency alleles or one severe and one mild allele (compound heterozygote). It is important to note, however, that the 10 most common mutations observed in CYP21A2 cause variable phenotype effects and are not always concordant with genotype. One study demonstrated that the genotype-phenotype concordance was as high as 90.5% for salt-wasting CAH, 85.1% for simple-virilizing CAH, and 97.8% for non-classical CAH (94).  In a study of 1,507 subjects with CAH by New et al, a direct genotype–phenotype correlation was noted in less than 50% of the genotypes studied. However, in the salt wasting and non-classical forms of 21OHD CAH, a phenotype was strongly correlated to a genotype (95). Moreover, it was shown in 2008  that CAG repeats in the androgen receptor have a great influence on variability in virilization of external genitalia of CAH women (96).

 

Figure 4. Common mutations in CYP21A2 gene and their related phenotypes. The numbers indicated exons of the gene. From: Forest MG. Recent advances in the diagnosis and management of congenital adrenal hyperplasia due to 21-hydroxylase deficiency (97).

 

DIAGNOSIS

 

Hormonal Diagnosis

 

Potential diagnosis of CAH must be suspected in infants born with atypical 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, and an assessment of the patient’s potential for future sexual activity and fertility. Physicians are urged to recognize the physical characteristics of CAH in newborns (e.g., atypical genitalia) and to refer such cases to appropriate clinics for full endocrine evaluation. 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. Traditionally, laboratories measured urinary excretion of adrenal hormones or their urinary metabolites (e.g., 17-ketosteroids). However, collection of 24-hour urine excretion is difficult, particularly in neonates. (98) Therefore, simple and reliable immunoassays are utilized now for measuring circulating serum levels of adrenal steroids (99). Alternatively, a non-invasive random urine collection in the first days of life for steroid hormone metabolites and precursor/product ratio assessments can be measured simultaneously. It can be used independently or in conjunction with serum steroid assays to increase accuracy and confidence in making the diagnosis and distinguishing the separate enzymatic forms of the disorder (100, 101).

 

Diagnosis of the 21OHD CAH can also be confirmed biochemically by a hormonal evaluation in blood or serum. 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 21OHD. Such testing is the basis of the newborn-screening program developed to identify classically affected patients who are at risk for salt wasting crisis (102). Only 20µl blood, obtained by heel prick and blotted on microfilter paper, is used for this purpose to provide a reliable diagnostic measurement of 17-OHP. The simplicity of the test and the ease of transporting microfilter paper specimens by mail have facilitated the implementation of CAH newborn screening programs worldwide. As of 2009, all 50 states in the United States screen for CAH.  Now, more than 35 countries worldwide have also established newborn screening programs. (103) False-positive results are, however, common with premature infants (104). Appropriate references based on weight and gestational age are therefore in place in many screening programs (105). The majority of screening programs use a single screening test without retesting of questionable 17-OHP concentrations. To improve efficacy, a small number of programs perform a second screening test of the initial sample to re-evaluate borderline cases identified by the first screening. Current immunoassay methods used in newborn screening programs yield a high false positive rate. To decrease this high rate, liquid-chromatography- tandem mass spectrometry measuring different hormones (17-OHP, Δ4-androstenedione, and cortisol) has been suggested as a second-tier method of analyzing positive results (106).

 

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 5) to ascertain disease severity (107). It is important to note that 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.  Testing is typically performed between 48 to 72 hours of life in newborns suspected of classical CAH and glucocorticoid treatment is initiated while awaiting results.  In newborns who are hemodynamically unstable and adrenal crisis is suspected, stimulation testing may delay life-saving treatment.  Screening measurements of 17-OHP, cortisol, and adrenal androgen along with ACTH can be obtained before emergent glucocorticoid administration.  The corticotropin stimulation test is crucial in establishing hormonal diagnosis of non-classical form of the disease since early-morning values of 17-OHP may not be sufficiently elevated to allow accurate diagnosis.

 

Figure 5. Nomogram relating baseline to ACTH-stimulated serum concentrations of 17-hydroxyprogesterone (17-OHP). The scales are logarithmic. A regression line for all data points is shown. Data points cluster as shown into three nonoverlapping groups: classic and non-classic forms of 21-hydroxylase deficiency are readily distinguished from each other and from those that are heterozygotes and unaffected. Distinguishing unaffected from heterozygotes is difficult. (107) Adapted from: New MI, Lorenzen F, Lerner AJ, et al. 1983 Genotyping steroid 21-hydroxylase deficiency: hormonal reference data. J Clin Endocrinol Metab 57:320-6. Permission obtained.

 

Prenatal Diagnosis of 21OHD

 

A number of approaches to prenatal identification of affected fetuses have been used. In 1965, Jeffcoate et al first reported a successful prenatal diagnosis of 21OHD, based on elevated levels of 17-ketosteroids and pregnanetriol in the amniotic fluid (108). The hormonal diagnostic test for 21OHD is amniotic fluid 17-OHP. Hormonal diagnosis is rarely used and considered only when molecular diagnosis is unavailable.

 

Advances in genotyping of the CYP21A2 gene have made molecular genetic studies of extracted fetal DNA the ideal method to diagnose 21OHD CAH in the fetus. Approximately 95% to 98% of the mutations causing 21OHD have been identified through a combination of molecular genetic techniques to study large gene rearrangement and arrays of point mutations (109, 110). Of the currently available methods for prenatal diagnosis of CAH, chorionic villus sampling (CVS), rather than amniocentesis, with molecular genotyping is the preferred diagnostic method in use. CVS is performed between the 9th and 11th week of gestation, while amniocentesis is usually performed in the second trimester. The timing of prenatal diagnosis is particularly important when deciding to treat the fetus at risk for CAH with dexamethasone prenatally to prevent virilization of the genitalia (see Prenatal Treatment below). As we only wish to treat affected females until term and only 1/8 of the fetuses will be affected and 1/2 will be males, 7 out of 8 fetuses do not require treatment. Thus, amniocentesis, which is performed later in gestation, results in treatment of unaffected fetuses for a longer period of time than CVS. However, amniocentesis can be used as a reliable alternative method of prenatal diagnosis when CVS in unavailable. In such instances, the supernatant is used for hormonal measurement and the cells are cultured to obtain a genotype through DNA analysis. The supernatant hormone measurements distinguish affected status from unaffected status only in SW patients. Nonetheless, pitfalls do occur in a small percentage of the patients undergoing prenatal diagnosis utilizing genetic diagnosis, such as undetectable mutations (111), allele drop outs (112), or maternal DNA contamination. Commercially available genetic testing utilizing short range PCR to detect common mutations may miss rare de novo mutations and thus the ACTH stimulation test remains vital to the evaluation.  Determination of satellite markers may increase the accuracy of molecular genetic analysis (113).

 

Non-Invasive Prenatal Diagnosis of CAH

 

Virilization of the genitalia in a female fetus affected with CAH owing to 21OHD and 11B-OHD can be treated prenatally with dexamethasone administered to the mother (see Prenatal Treatment below). Because CAH is an autosomal recessive disorder, the risk is 1/4 of the fetus being affected with the disease and 1/8 of the fetus being a female with atypical genitalia. Therefore, 7 out of 8 pregnancies will receive unnecessary treatment until the sex and the affection status of the fetus are known. Treatment with dexamethasone must begin before the 9th week of gestation, yet chorionic villous sampling can only be done at the 9-11th week, with karyotype and DNA results available 2-3 weeks later. Non-invasive prenatal diagnosis would eliminate unnecessary treatment and invasive procedures such as CVS and amniocentesis. Dennis Lo et al. in 1997 discovered the presence of fetal DNA in the maternal circulation (113). Fetal DNA has been extracted and enriched with high accuracy and yield in fetal Rh factor identification (114), aneuploidy and monogenic disorders such as thalassemia and cystic fibrosis (115). Identification of the SRY sequence in maternal blood, performed in multiple academic centers and now available in commercial laboratories, has also achieved excellent accuracy in several studies (116, 117). In non-invasive prenatal diagnosis of CAH, by extracting fetal DNA from the maternal blood as early as 4-5 weeks gestation, the SRY sequence can be identified to determine sex (118). If the fetal genetic sex is deduced to be female (SRY sequence not identified), DNA analysis on extracted fetal DNA can be used to determine CAH affected status. Targeted massive parallel sequencing of cell-free fetal DNA in maternal plasma was used for the noninvasive prenatal diagnosis of CAH due to 21OHD(119).   In the fourteen expectant families studied, each with a previous child affected with classical CAH (proband) and parents with at least one mutant CYP21A2 gene, the fetal CAH affection status was correctly deduced using this method from maternal plasma drawn as early as 5 weeks and 6 days (119).

 

Preimplantation Diagnosis

 

Preimplantation genetic diagnosis (PGD) identifies genetic abnormalities in preimplantation embryos prior to embryo transfer, so only unaffected embryos established from IVF are transferred. The procedure has been utilized in many monogenic recessive disorders such as cystic fibrosis, hemoglobinopathies, spinal muscular atrophy and Tay Sach’s disease. PGD is being used for a growing number of genetic diseases (120). There is only one report of PGD utilized in a family whose offspring is at risk for CAH (121), however we know from experience that families are seeking PGD with greater frequency. It would be desirable to have further studies of preimplantation diagnosis in CAH families.

 

Prenatal Treatment

 

In 21OHD, prenatal treatment with dexamethasone was introduced in France in 1978 (122) and in the United States in 1986 (123). Institution of therapy before the 9th week of gestation, prior to the onset of adrenal androgen secretion, effectively suppresses excessive adrenal androgen production and prevents virilization of external female genitalia. Dexamethasone is used because it binds minimally to cortisol binding globulin (CBG) in the maternal blood, and unlike hydrocortisone, it escapes inactivation by the placental 11-dehydrogenase enzyme. Thus, dexamethasone crosses the placenta from the mother to the fetus and suppresses ACTH secretion with longer half-life compared to other synthetic steroids (123).

 

When dexamethasone administration begins as early as the 8th week of gestation, the treatment is blind to the disease status and sex of the fetus. If the fetus is later determined to be a male upon karyotype or an unaffected female upon DNA analysis, treatment is discontinued. Otherwise, treatment is continued to term. A simplified algorithm of management of potentially affected pregnancies is shown in Figure 6.  The optimal dosage is 20 µg/kg/day of dexamethasone per maternal pre-pregnancy body weight, in three divided daily doses (124). It is recommended to start the treatment as soon as pregnancy is confirmed, and no later than 9 weeks after the last menstrual period (125, 126). The mother’s blood pressure, weight, glycosuria, HbA1C, symptoms of edema, striae and other possible adverse effects of dexamethasone treatment should be carefully observed throughout pregnancy (127) . Prenatal dexamethasone treatment remains controversial and should be administered under an IRB approved research protocol(104).

 

Figure 6. Algorithm of treatment, diagnosis and decision-making for prenatal treatment of fetuses at risk for 21-hydroxylase deficiency congenital adrenal hyperplasia. Mercado AB, Wilson RC, Cheng KC, Wei JQ, New MI 1995 Extensive personal experience: Prenatal treatment and diagnosis of congenital adrenal hyperplasia owing to steroid 21-hydroxylase deficiency. J Clin Endocrinol Metab 80:2014-2020. (125) Permission obtained.

 

Outcome of Prenatal Treatment of 21OHD

 

Prenatal treatment of 21OHD has proven to be successful in significantly reducing genital virilization in affected females. Our group has performed prenatal diagnosis in over 685 pregnancies at risk for 21OHD, and 59 affected female fetuses have been treated to term (128). Treatment was highly effective in preventing genital ambiguity when the mother was compliant until term. In all the female fetuses treated to term, the degree of virilization was on average 1.69, as measured using the Prader scoring system (Figure 3). Late treatment as well as no treatment resulted in much greater virilization, and the average Prader score was 3.73 for those female fetuses not treated. 124 and unpublished data Not only does prenatal treatment effectively minimize the degree of female genital virilization in the patients, it also lessens the high-level androgen exposure of the brain during differentiation. The latter is thought to cause a higher tendency to gender ambiguity in some females with CAH (129, 130). Genital virilization in female newborns with classical 21OHD CAH has potential adverse psychosocial implications that may be alleviated by prenatal treatment (124).

 

Prenatal dexamethasone treatment has continued to be a subject of controversy (104). Some uncertainties and concerns have been expressed about the long-term safety of prenatal diagnosis and treatment (131, 132). Concerns have been raised in regards to the glucocorticoid effects on the fetal brain, which arise from studies of other conditions rather than direct studies on prenatal treatment of 21OHD CAH. These include studies whereby much higher doses of dexamethasone were given to the human subjects at the later part of pregnancy (133) or to animals (134, 135) and therefore hold little relevance to using dexamethasone prenatally in CAH. In a small-sample study of children prenatally treated with dexamethasone, Lajic and her colleagues found no effects on intelligence, handedness, memory encoding, or long-term memory, but short-term treated CAH-unaffected children had significantly poorer performance than controls on a test of verbal working memory. These patients also had lower questionnaire scores in self-perceived scholastic competence and social anxiety (136). However, parents described these children as more sociable than controls, without significant difference in psychopathology, school performance, adaptive functioning or behavioral problems (137). A larger multi-center study (US and France) indicated no adverse cognitive effects of short-term prenatal DEX exposure, including no adverse influence on verbal working memory; a small sample of dexamethasone treated girls affected with CAH showed lower scores on two of eight neuropsychological tests, however given the variability of cognitive findings in dexamethasone unexposed CAH-affected patients, this result cannot be linked to dexamethasone with certainty (138). This indicates that further studies are needed.

 

Compelling data from large cohorts of pregnancies with prenatal diagnosis and treatment of 21OHD CAH prove its efficacy and safety (126, 139). All of the mothers who received prenatal treatment (partial or full-term) stated that they would take dexamethasone again for a future pregnancy (140).  Rare adverse events have been reported in treated children, but no harmful effects have been documented that can be clearly attributed to the treatment (141). Another long-term follow-up study in Scandinavia showed that 44 children who were variably treated prenatally demonstrated normal prenatal and postnatal growth compared to matched controls. Further, there was no observed increase in fetal abnormalities or fetal death (142). Although some abnormalities in postnatal growth and behavior were observed among dexamethasone exposed offspring, none could logically be explained by the present knowledge of teratogenic effects of glucocorticoids.

 

Published studies of almost 600 pregnancies, 80 of which were prenatally treated until term and 27 who were male and received dexamethasone for a short period of time, the newborns in the dexamethasone treated group did not differ in weight, length or head circumference from untreated, unaffected siblings. No significant or enduring side effects were noted in either the mothers or the fetuses. Greater weight gain in treated versus untreated mothers did occur, as well as the presence of striae and edema. Excessive weight gain was lost after birth. No differences were found regarding gestational diabetes or hypertension (127). No cases have been reported of cleft palate, placental degeneration or fetal death, which have been observed in the rodent model of in utero exposure to high-dose glucocorticoids (143). One explanation for the safety of human versus rodent is that glucocorticoid receptor-ligand systems in human differ from that of rodents (144). A comprehensive long-term outcome study looking at 149 male and female patients 12 years of age and older, affected and unaffected with CAH, who were treated with dexamethasone partially or to term was conducted. To date, this is the largest study evaluating the long-term effects of dexamethasone. No adverse effects such as increased risk for cognitive defects, disorders of gender identity and behavior, sexual function in adulthood, hypertension, diabetes, and osteopenia were found (127).

 

Prenatal Diagnosis and Treatment of 11β-OHD CAH

 

Several approaches to prenatal identification by measuring steroid precursors in affected fetuses have been used (145-147). Advances in genotyping of the CYP11B1 gene have made molecular genetic studies of fetal DNA extracted from maternal blood, the ideal method to diagnose 11β-OHD CAH in the fetus (148, 149). The established protocol of prenatal diagnosis and treatment in 21OHD CAH can be applied to 11β-OHD CAH. Reduced virilization of affected females in prenatal diagnosis and treatment in 11β-OHD CAH have been reported (128, 148).

 

TREATMENT

 

Hormone Replacement

 

The goal of therapy in CAH is to both correct the deficiency in cortisol secretion and to suppress ACTH overproduction (104). 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 21OHD form of CAH is about 10-15 mg/m2/day divided into 2 or 3 doses per day.  Conventionally, oral hydrocortisone tablets with the smallest available dose of 5 mg have been preferred.  To administer small doses to infants and young children, 10 mg and 5 mg tablets have to be cut and crushed.  Hydrocortisone granules and tablets at smaller doses are becoming more widely available allowing for ease of administration and avoidance of excess dosing (150).

 

Dosage requirements for patients with NC-21OHD CAH are typically less. Adults may be treated with the longer-acting dexamethasone or prednisone, alone or in combination with hydrocortisone. A small dose of dexamethasone at bedtime (0.25 to 0.5 mg) is usually adequate for androgen suppression in non-classical patients. Anti-androgen treatment may be useful as adjunctive therapy in adult women who continue to have hyperandrogenic signs despite good adrenal suppression. Females with concomitant PCOS may benefit from an oral contraceptive, though this treatment would not be appropriate for patients trying to get pregnant. Treatment of adult males with NC-21OHD may not be necessary, though our group has found that it may be helpful in preventing adrenal rest tumors and preserving fertility. Optimal corticosteroid therapy is determined by adequate suppression of adrenal hormones balanced against normal physiological parameters. The goal of corticosteroid therapy is to give the lowest dose required for optimal control. 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 in newborn males). It is recommended that hormone levels are measured at a consistent time in relation to medication dosing, usually 2 hours after the morning corticosteroid. Titration of the dose should be aimed at maintaining androgen levels at age and sex-appropriate levels and 17-OHP levels of <1000 ng/dL.  Analysis of diurnal and 24-hour urine collection of 17-OHP metabolites can be utilized to further assess adrenal control. (151)  Concurrently, over-treatment should be avoided because it can lead to Cushing syndrome. Depending on the degree of stress, stress dose coverage may require doses of up to 50-100 mg/m2/day (1).

 

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, ranging from 0.05 mg to 0.2 mg daily. Infants should also be started on salt supplementation, as sodium chloride, at 1-2 g daily, divided into several feedings. Although patients with the SV and NC form of CAH can make adequate aldosterone, the aldosterone to renin ratio (ARR) has been found to be lower than normal, though not to the degree seen in the salt-wasting form (152)(155). It has not been customary to supplement conventional glucocorticoid replacement therapy with the administration of salt-retaining steroids in the SV and NC forms of CAH, though there has been some suggestion that adding fludrocortisone to patients with elevated PRA may improve hormonal control of the disease (153). The requirement for fludrocortisone appears to diminish with age, and over-suppression of the PRA should be avoided, to prevent complications from hypertension and excessive mineralocorticoid activity. Measurements of plasma renin and aldosterone are used to monitor the efficacy of mineralocorticoid therapy in all patients with the salt wasting form of the disease (1). The use of systemic cortisol injection for impending adrenal crises is discussed below.

 

In managing 11β-OHD, glucocorticoid administration provides cortisol replacement and decreases ACTH, as it does in 21OHD. This in turn removes the drive for over secretion of deoxycorticosterone (DOC) and 11-deoxycortisol and, in most cases, normalizes blood pressure. A thorough examination undertaken by endocrine challenge and suppression studies to evaluate zonal differences has shown that in 11β-OHD CAH, the zona fasciculata exhibits reduced 11β-hydroxylation and 18-hydroxylation, while both functions appear to be spared in the zona glomerulosa (154). This demonstrates that the zona glomerulosa and the zona fasciculata function as two physiologically, and likely genetically, separate glands. Glucocorticoid treatment produces natriuresis and diuresis, normalizes plasma volume and thus increases the plasma renin to levels that stimulate aldosterone production in the zona glomerulosa. In addition to normalizing blood pressure, the goal of treatment is to replace deficient steroids and in turn minimize adrenal sex hormone excess, prevent virilization, optimize growth, and protect potential fertility. Serum DOC and 11-deoxycortiol are thus the principal steroid index of the 11β-OHD. Plasma renin activity is useful as a therapeutic index as well. In poorly controlled 11β-OHD patients, DOC is elevated, whereas plasma renin is suppressed; both are normal in well-controlled patients. As in patients with 21OHD, oral hydrocortisone at a dose of 10-15 mg/m2 divided into two to three daily doses is the preferred treatment. Long-acting glucocorticoids may be used at or near the completion of linear growth. In patients who have had ongoing hypertension for some time before diagnosis is made, adding spironolactone, calcium channel blockers or amiloride may be necessary (60).

 

In patients with 3β-HSD deficiency, glucocorticoid administration also reduces the excess production of androgens. In addition, these patients have mineralocorticoid deficiency and require treatment with the salt-retaining 9α-fludrocortisone acetate. Patients with the StAR protein deficiency or SCC deficiency (lipoid form of CAH) classically have severe adrenal insufficiency with mineralocorticoid deficiency and salt wasting; they require both glucocorticoid and mineralocorticoid replacement. Patients with 17 α -hydroxylase/17,20 lyase deficiency typically have excess DOC and low-renin hypertension, and treatment with a glucocorticoid should normalize serum DOC level and lead to normalization of blood pressure. In several conditions, such as StAR protein deficiency, 3β-HSD, 17 α -hydroxylase/17,20 lyase deficiency and cytochrome P450 oxidoreductase deficiency, patients require sex steroid replacement. Sex steroids should be added at a developmentally appropriate time to allow patients to resemble their peers.

 

Because patients with CAH are at risk for short stature as adults, other adjunct therapies are being utilized. Two studies have demonstrated significant improvement in growth velocity, final adult height prediction (17) and final adult height (15) with the use of growth hormone in conjunction with a GnRH analogue.

 

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. Each family should be given injection kits of hydrocortisone for emergency use (25 mg for infants, 50 mg for children, and 100 mg for adults). In the event of a surgical procedure, a total of 5 to 10 times the daily maintenance dose may be required during the first 24-48 hours, which 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 (1, 104).

 

It is imperative for all patients who are receiving corticosteroid replacement therapy, such as patients with CAH, to wear a Medic-Alert bracelet, medallion, or equivalent identification that will enable correct and appropriate therapy in case of emergencies. Additionally, all responsible family members should be trained in the intramuscular administration of hydrocortisone.

 

Bone Mineral Density

 

In order to adequately suppress androgen production in patients with CAH, the usual requirement of hydrocortisone is generally higher than the endogenous secretory rate of cortisol. Chronic therapy with glucocorticoids at supraphysiologic levels can result in diminished bone accrual and lead to osteopenia and osteoporosis. Glucocorticoid induced bone loss is a well-known phenomenon and is the most prevalent form of secondary osteoporosis (155, 156).

 

Unlike other diseases treated with chronic glucocorticoid therapy, however, the effect of glucocorticoid replacement in CAH on BMD is unclear. Previous studies of patients with 21OHD have reported increased, normal, or decreased BMD (157-160). It has been postulated that the elevated androgens typically found in patients may have a protective effect on bone integrity, but the precise mechanism is unknown. The increased adrenal androgens, which are converted to estrogens, may counteract the detrimental effects of glucocorticoids on bone mass. This may explain why older CAH women, particularly those who are post-menopausal, are at higher risk for osteoporosis than younger CAH patients. It has been proposed that the inhibitory effect of corticosteroid therapy on bone formation is counteracted by estrogen’s effect on bone resorption through the RANK-L/osteoprotegerin (OPG) system (161).

 

Surgery

 

The decision for genital surgery in females with virilizing forms of CAH and males with forms of CAH associated with under virilization should be made by parents or patients themselves with the guidance of a multidisciplinary team involving pediatric endocrinology, urology, genetics, and psychology.  As part of a case-by-case approach, attention should be given to gender identity, quality of life and potential for fertility.  The aim of surgical repair in females with atypical genitalia caused by CAH, if the decision is made, is generally to preserve the sexually sensitive glans clitoris, decrease frequency of urinary tract infections and provide a vaginal orifice that functions adequately for menstruation, intromission, and delivery. 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 atypical 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. Data show that genital sensitivity is impaired in areas where feminizing genital surgery had been done, leading to difficulties in sexual function (162). Another study showed that patients with more severe mutations in the CYP21A2 gene, i.e., those with the null genotype and thus those more severely virilized, had more surgical complications that those less severely virilized and were less satisfied with their sexual life (163). Because of the scarcity of these data, the role of the parents in sex assignment becomes crucial in all aspects of the decision-making process, and should include full discussion of the controversy and all possible therapeutic options for the intersex child, particularly early versus delayed surgery. A large repository of data concerning long-term outcomes in patients affected by CAH, including psychosexual well-being, has been enhanced by the establishment of disease registries (164).

 

In a study of intersex individuals ≥ 16 years old, 66% of individuals with CAH thought that the appropriate age of genital surgery was infancy or childhood (165).  In a study of caregivers of female infants with CAH who underwent genitoplasty, 2/3 of caregivers reported no regret over their decision-making.  However, 1/3 reported some level of regret in the process (165).

 

Other Treatment Strategies and Novel Therapies

 

Glucocorticoid replacement has been an effective treatment for CAH for over 50 years and remains its primary therapy; however, the management of these patients presents a challenge because both inadequate treatment, as well as over suppression, can cause complications.  In forms of CAH with decreased cortisol production, increased ACTH production stimulates excessive synthesis of adrenal products in those pathways unimpaired by the enzyme deficiency such as androgens.  The goal of therapy in CAH is to both correct the deficiency in cortisol secretion and to suppress ACTH overproduction which drives androgen production (104).

 

Often, doses of hydrocortisone higher than physiologic replacement or dosing in reverse diurnal pattern are required to suppress androgen production driven by ACTH.   Alternatives to oral glucocorticoid administration 2-3 times a day include continuous subcutaneous pump and newer modified release formulations.  Physiologic cortisol secretion patterns can be mimicked with varied infusion rates of hydrocortisone via continuous subcutaneous pump administration.  This more intense method of administration should be considered in those with rapid cortisol metabolism or poor gastrointestinal absorption (166).  Modified release and delayed release formulations of hydrocortisone were developed recently (167).  When taken at bedtime, these formulations approximate the diurnal pattern of physiological cortisol production with an early morning peak and aim to prevent the ACTH surge at dawn that drives androgen production (168).

 

Adjunct treatments targeting hypothalamic and pituitary signaling to the adrenal glands are in development. To suppress ACTH production, corticotrophin-releasing factor (CRF) antagonists have been developed. CRF produced in the hypothalamus stimulates the release of ACTH from the pituitary gland.   CRF antagonism can reduce ACTH production and reduce ACTH driven androgen production in patients with CAH.  With the adjunct use of CRF antagonists, lower doses of hydrocortisone can be used for physiologic replacement only. Two oral agents are currently undergoing clinical trials in patients with classical CAH (169, 170).

 

To reduce androgen production, the use of abiraterone acetate indicated for prostate cancer was proposed for patients with CAH. (171)  Abiraterone acetate is a potent inhibitor of steroid enzyme 17-hydroxylase/17,20-lyase (CYP17A1) needed for androgen production and is being studied in clinical trials as adjunct therapy in adult and soon pediatric subjects. (164, 172)

 

Bilateral adrenalectomy is a radical measure that can be effective in some cases. A few patients who were extremely difficult to control with medical therapy alone showed improvement in their symptoms after bilateral adrenalectomy (164, 165). Because this approach renders the patient completely adrenal insufficient, however, it should be reserved for extreme cases and is not a good treatment option for patients who have a history of poor compliance with medication.

 

CONCLUSION

 

The pathophysiology of the various types of CAH (the most common being 21OHD) can be traced to specific, inherited defects in the genes encoding enzymes for adrenal steroidogenesis. Clinical presentation of each form is distinctive and depends largely on the underlying enzyme defect, its precursor retention and deficient end products. Treatment of CAH is targeted to replace the insufficient adrenal hormones, notably cortisol and salt retaining hormones, and to suppress the excess precursors. With proper hormone replacement therapy, normal and healthy development may be expected. Glucocorticoid and, if necessary, mineralocorticoid replacement, has been the mainstay of treatment for CAH, but new treatment strategies continue to be developed and studied to improve care. Molecular genetic techniques used postnatally and prenatally along with well described genotype phenotype correlations can help guide clinical management. 

 

REFERENCES

 

  1. New MI, L.O., Mancenido D, Parsa A, Yuen T, Congenital Adrenal Hyperplasia Owing to 21-Hydroxylase Deficiency, in Genetic Steroid Disorders, L.O. New MI, Parsa A, Yuen T, O'Malley BW, Hammer GD, Editor. 2014, Elsevier: San Diego, CA. p. 29-51.
  2. Pang, S.Y., et al., Worldwide experience in newborn screening for classical congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Pediatrics, 1988. 81(6): p. 866-74.
  3. Speiser, P.W., et al., High frequency of nonclassical steroid 21-hydroxylase deficiency. Am J Hum Genet, 1985. 37(4): p. 650-67.
  4. Sherman, S.L., et al., A segregation and linkage study of classical and nonclassical 21-hydroxylase deficiency. Am J Hum Genet, 1988. 42(6): p. 830-8.
  5. Zerah, M., et al., Prevalence of nonclassical steroid 21-hydroxylase deficiency based on a morning salivary 17-hydroxyprogesterone screening test: a small sample study. J Clin Endocrinol Metab, 1990. 70(6): p. 1662-7.
  6. Zachmann, M., D. Tassinari, and A. Prader, Clinical and biochemical variability of congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. A study of 25 patients. J Clin Endocrinol Metab, 1983. 56(2): p. 222-9.
  7. Curnow, K.M., et al., Mutations in the CYP11B1 gene causing congenital adrenal hyperplasia and hypertension cluster in exons 6, 7, and 8. Proc Natl Acad Sci U S A, 1993. 90(10): p. 4552-6.
  8. Khemiri, M., et al., (11 beta hydroxylase deficiency: a clinical study of seven cases). Tunis Med, 2006. 84(2): p. 106-13.
  9. Rosler, A., E. Leiberman, and T. Cohen, High frequency of congenital adrenal hyperplasia (classic 11 beta-hydroxylase deficiency) among Jews from Morocco. Am J Med Genet, 1992. 42(6): p. 827-34.
  10. Paperna, T., et al., Mutations in CYP11B1 and congenital adrenal hyperplasia in Moroccan Jews. J Clin Endocrinol Metab, 2005. 90(9): p. 5463-5.
  11. Xing Y, A.J., Hammer GD, Adrenal Development, in Genetic Steroid Disorders L.O. New MI, Mancenido D, Parsa A, Yuen T, Editor. 2014, Elsevier: San Diego, CA. p. 5-27.
  12. Goto, M., et al., In humans, early cortisol biosynthesis provides a mechanism to safeguard female sexual development. J Clin Invest, 2006. 116(4): p. 953-60.
  13. Prader, A. and H.P. Gurtner, (The syndrome of male pseudohermaphrodism in congenital adrenocortical hyperplasia without overproduction of androgens (adrenal male pseudohermaphrodism)). Helv Paediatr Acta, 1955. 10(4): p. 397-412.
  14. New, M.I., et al., Growth and final height in classical and nonclassical 21-hydroxylase deficiency. J Endocrinol Invest, 1989. 12(8 Suppl 3): p. 91-5.
  15. Lin-Su, K., et al., Final adult height in children with congenital adrenal hyperplasia treated with growth hormone. J Clin Endocrinol Metab, 2011. 96(6): p. 1710-7.
  16. Halper, A., et al., Use of an aromatase inhibitor in children with congenital adrenal hyperplasia: Impact of anastrozole on bone mineral density and visceral adipose tissue. Clin Endocrinol (Oxf), 2019. 91(1): p. 124-130.
  17. Trinh, L., et al., Growth and pubertal characteristics in patients with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Pediatr Endocrinol Metab, 2007. 20(8): p. 883-91.
  18. Helleday, J., et al., Subnormal androgen and elevated progesterone levels in women treated for congenital virilizing 21-hydroxylase deficiency. J Clin Endocrinol Metab, 1993. 76(4): p. 933-6.
  19. Premawardhana, L.D., et al., Longer term outcome in females with congenital adrenal hyperplasia (CAH): the Cardiff experience. Clin Endocrinol (Oxf), 1997. 46(3): p. 327-32.
  20. Richards, G.E., et al., The effect of long acting glucocorticoids on menstrual abnormalities in patients with virilizing congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1978. 47(6): p. 1208-15.
  21. Barnes, R.B., et al., Ovarian hyperandrogynism as a result of congenital adrenal virilizing disorders: evidence for perinatal masculinization of neuroendocrine function in women. J Clin Endocrinol Metab, 1994. 79(5): p. 1328-33.
  22. Abdelhamed, M.H., W.M. Al-Ghamdi, and A.E. Al-Agha, Polycystic Ovary Syndrome Among Female Adolescents With Congenital Adrenal Hyperplasia. Cureus, 2021. 13(12): p. e20698.
  23. Long, D.N., A.B. Wisniewski, and C.J. Migeon, Gender role across development in adult women with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Pediatr Endocrinol Metab, 2004. 17(10): p. 1367-73.
  24. Hines, M., C. Brook, and G.S. Conway, Androgen and psychosexual development: core gender identity, sexual orientation and recalled childhood gender role behavior in women and men with congenital adrenal hyperplasia (CAH). J Sex Res, 2004. 41(1): p. 75-81.
  25. Meyer-Bahlburg, H.F., et al., Sexual orientation in women with classical or non-classical congenital adrenal hyperplasia as a function of degree of prenatal androgen excess. Arch Sex Behav, 2008. 37(1): p. 85-99.
  26. de Jesus, L.E., E.C. Costa, and S. Dekermacher, Gender dysphoria and XX congenital adrenal hyperplasia: how frequent is it? Is male-sex rearing a good idea? J Pediatr Surg, 2019. 54(11): p. 2421-2427.
  27. Berenbaum, S.A., K.L. Bryk, and A.M. Beltz, Early androgen effects on spatial and mechanical abilities: evidence from congenital adrenal hyperplasia. Behav Neurosci, 2012. 126(1): p. 86-96.
  28. Berenbaum, S.A., K.K. Bryk, and S.C. Duck, Normal intelligence in female and male patients with congenital adrenal hyperplasia. Int J Pediatr Endocrinol, 2010. 2010: p. 853103.
  29. Reichman, D.E., et al., Fertility in patients with congenital adrenal hyperplasia. Fertil Steril, 2014. 101(2): p. 301-9.
  30. Mulaikal, R.M., C.J. Migeon, and J.A. Rock, Fertility rates in female patients with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. N Engl J Med, 1987. 316(4): p. 178-82.
  31. Engels, M., et al., Testicular Adrenal Rest Tumors: Current Insights on Prevalence, Characteristics, Origin, and Treatment. Endocr Rev, 2019. 40(4): p. 973-987.
  32. Cabrera, M.S., M.G. Vogiatzi, and M.I. New, Long term outcome in adult males with classic congenital adrenal hyperplasia. J Clin Endocrinol Metab, 2001. 86(7): p. 3070-8.
  33. Urban, M.D., P.A. Lee, and C.J. Migeon, Adult height and fertility in men with congenital virilizing adrenal hyperplasia. N Engl J Med, 1978. 299(25): p. 1392-6.
  34. Claahsen-van der Grinten, H.L., et al., Prevalence of testicular adrenal rest tumours in male children with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Eur J Endocrinol, 2007. 157(3): p. 339-44.
  35. Molitor, J.T., B.S. Chertow, and B.L. Fariss, Long-term follow-up of a patient with congenital adrenal hyperplasia and failure of testicular development. Fertil Steril, 1973. 24(4): p. 319-23.
  36. Claahsen-van der Grinten, H.L., et al., Testicular adrenal rest tumors in patients with congenital adrenal hyperplasia can cause severe testicular damage. Fertil Steril, 2008. 89(3): p. 597-601.
  37. Wilkins, L., et al., Further studies on the treatment of congenital adrenal hyperplasia with cortisone. III. The control of hypertension with cortisone, with a discussion of variations in the type of congenital adrenal hyperplasia and report of a case with probable defect of carbohydrate-regulating hormones. J Clin Endocrinol Metab, 1952. 12(8): p. 1015-30.
  38. Blumberg-Tick, J., et al., Testicular tumors in congenital adrenal hyperplasia: steroid measurements from adrenal and spermatic veins. J Clin Endocrinol Metab, 1991. 73(5): p. 1129-33.
  39. Schoen, E.J., V. Di Raimondo, and O.V. Dominguez, Bilateral testicular tumors complicating congenital adrenocortical hyperplasia. J Clin Endocrinol Metab, 1961. 21: p. 518-32.
  40. Miller, E.C., Jr. and H.L. Murray, Congenital adrenocortical hyperplasia: case previously reported as "bilateral interstitial cell tumor of the testicle". J Clin Endocrinol Metab, 1962. 22: p. 655-7.
  41. Glenn, J.F. and W.H. Boyce, Adrenogenitalism with testicular adrenal rests simulating interstitial cell tumor. J Urol, 1963. 89: p. 457-63.
  42. Srikanth, M.S., et al., Benign testicular tumors in children with congenital adrenal hyperplasia. J Pediatr Surg, 1992. 27(5): p. 639-41.
  43. Rutgers, J.L., R.H. Young, and R.E. Scully, The testicular "tumor" of the adrenogenital syndrome. A report of six cases and review of the literature on testicular masses in patients with adrenocortical disorders. Am J Surg Pathol, 1988. 12(7): p. 503-13.
  44. Sugino, Y., et al., Genotyping of congenital adrenal hyperplasia due to 21-hydroxylase deficiency presenting as male infertility: case report and literature review. J Assist Reprod Genet, 2006. 23(9-10): p. 377-80.
  45. Klein, R., Evidence for and against the existence of a salt-losing hormone. J Pediatr, 1960. 57: p. 452-60.
  46. Kowarski, A., et al., Aldosterone Secretion Rate in Congenital Adrenal Hyperplasia. A Discussion of the Theories on the Pathogenesis of the Salt-Losing Form of the Syndrome. J Clin Invest, 1965. 44: p. 1505-13.
  47. Kuhnle, U., M. Land, and S. Ulick, Evidence for the secretion of an antimineralocorticoid in congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1986. 62(5): p. 934-40.
  48. Tusie-Luna, M.T., P. Traktman, and P.C. White, Determination of functional effects of mutations in the steroid 21-hydroxylase gene (CYP21) using recombinant vaccinia virus. J Biol Chem, 1990. 265(34): p. 20916-22.
  49. Stoner, E., et al., Is salt-wasting in congenital adrenal hyperplasia due to the same gene as the fasciculata defect? Clin Endocrinol (Oxf), 1986. 24(1): p. 9-20.
  50. Luetscher, J.A., Jr., Studies of aldosterone in relation to water and electrolyte balance in man. Recent Prog Horm Res, 1956. 12: p. 175-84; discussion, 184-98.
  51. Speiser, P.W., et al., Aldosterone synthesis in salt-wasting congenital adrenal hyperplasia with complete absence of adrenal 21-hydroxylase. N Engl J Med, 1991. 324(3): p. 145-9.
  52. Rose, L.I., et al., Adrenocortical hydroxylase deficiencies in acne vulgaris. J Invest Dermatol, 1976. 66(5): p. 324-6.
  53. Lucky, A.W., et al., Adrenal androgen hyperresponsiveness to adrenocorticotropin in women with acne and/or hirsutism: adrenal enzyme defects and exaggerated adrenarche. J Clin Endocrinol Metab, 1986. 62(5): p. 840-8.
  54. Lin-Su, K., S. Nimkarn, and M.I. New, Congenital adrenal hyperplasia in adolescents: diagnosis and management. Ann N Y Acad Sci, 2008. 1135: p. 95-8.
  55. Witchel, S.F. and R. Azziz, Nonclassic congenital adrenal hyperplasia. Int J Pediatr Endocrinol, 2010. 2010: p. 625105.
  56. Mokshagundam, S. and M.I. Surks, Congenital adrenal hyperplasia diagnosed in a man during workup for bilateral adrenal masses. Arch Intern Med, 1993. 153(11): p. 1389-91.
  57. Jaresch, S., et al., Adrenal incidentaloma and patients with homozygous or heterozygous congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1992. 74(3): p. 685-9.
  58. Barzon, L., et al., Incidentally discovered adrenal tumors: endocrine and scintigraphic correlates. J Clin Endocrinol Metab, 1998. 83(1): p. 55-62.
  59. Nieman, L.K., Approach to the patient with an adrenal incidentaloma. J Clin Endocrinol Metab, 2010. 95(9): p. 4106-13.
  60. Nimkarn, S. and M.I. New, Steroid 11beta- hydroxylase deficiency congenital adrenal hyperplasia. Trends Endocrinol Metab, 2008. 19(3): p. 96-9.
  61. Khattab, A., et al., Clinical, genetic, and structural basis of congenital adrenal hyperplasia due to 11beta-hydroxylase deficiency. Proc Natl Acad Sci U S A, 2017. 114(10): p. E1933-E1940.
  62. Globerman, H., et al., An inherited defect in aldosterone biosynthesis caused by a mutation in or near the gene for steroid 11-hydroxylase. N Engl J Med, 1988. 319(18): p. 1193-7.
  63. Rosler, A., et al., Clinical variability of congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Horm Res, 1982. 16(3): p. 133-41.
  64. Hague, W.M. and J.W. Honour, Malignant hypertension in congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Clin Endocrinol (Oxf), 1983. 18(5): p. 505-10.
  65. Chabre, O., et al., Bilateral laparoscopic adrenalectomy for congenital adrenal hyperplasia with severe hypertension, resulting from two novel mutations in splice donor sites of CYP11B1. J Clin Endocrinol Metab, 2000. 85(11): p. 4060-8.
  66. Joehrer, K., et al., CYP11B1 mutations causing non-classic adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Hum Mol Genet, 1997. 6(11): p. 1829-34.
  67. Peters, C.J., et al., Cosegregation of a novel homozygous CYP11B1 mutation with the phenotype of non-classical congenital adrenal hyperplasia in a consanguineous family. Horm Res, 2007. 67(4): p. 189-93.
  68. Simard, J., et al., Molecular basis of human 3 beta-hydroxysteroid dehydrogenase deficiency. J Steroid Biochem Mol Biol, 1995. 53(1-6): p. 127-38.
  69. Labrie, F., et al., Structure and tissue-specific expression of 3 beta-hydroxysteroid dehydrogenase/5-ene-4-ene isomerase genes in human and rat classical and peripheral steroidogenic tissues. J Steroid Biochem Mol Biol, 1992. 41(3-8): p. 421-35.
  70. Pang, S., Congenital adrenal hyperplasia owing to 3 beta-hydroxysteroid dehydrogenase deficiency. Endocrinol Metab Clin North Am, 2001. 30(1): p. 81-99, vi-vii.
  71. Yanase, T., E.R. Simpson, and M.R. Waterman, 17 alpha-hydroxylase/17,20-lyase deficiency: from clinical investigation to molecular definition. Endocr Rev, 1991. 12(1): p. 91-108.
  72. Miller, W.L., R.J. Auchus, and D.H. Geller, The regulation of 17,20 lyase activity. Steroids, 1997. 62(1): p. 133-42.
  73. Bose, H.S., et al., Mutations in the steroidogenic acute regulatory protein (StAR) in six patients with congenital lipoid adrenal hyperplasia. J Clin Endocrinol Metab, 2000. 85(10): p. 3636-9.
  74. Bose, H.S., et al., The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. N Engl J Med, 1996. 335(25): p. 1870-8.
  75. Miller, W.L., Mechanism of StAR's regulation of mitochondrial cholesterol import. Mol Cell Endocrinol, 2007. 265-266: p. 46-50.
  76. Stocco, D.M. and B.J. Clark, The role of the steroidogenic acute regulatory protein in steroidogenesis. Steroids, 1997. 62(1): p. 29-36.
  77. Sahakitrungruang, T., et al., Clinical, genetic, and functional characterization of four patients carrying partial loss-of-function mutations in the steroidogenic acute regulatory protein (StAR). J Clin Endocrinol Metab, 2010. 95(7): p. 3352-9.
  78. Tee, M.K., et al., Varied clinical presentations of seven patients with mutations in CYP11A1 encoding the cholesterol side-chain cleavage enzyme, P450scc. J Clin Endocrinol Metab, 2013. 98(2): p. 713-20.
  79. Parajes, S., et al., A novel entity of clinically isolated adrenal insufficiency caused by a partially inactivating mutation of the gene encoding for P450 side chain cleavage enzyme (CYP11A1). J Clin Endocrinol Metab, 2011. 96(11): p. E1798-806.
  80. Scott, R.R. and W.L. Miller, Genetic and clinical features of p450 oxidoreductase deficiency. Horm Res, 2008. 69(5): p. 266-75.
  81. Fluck, C.E., et al., P450 oxidoreductase deficiency - a new form of congenital adrenal hyperplasia. Endocr Dev, 2008. 13: p. 67-81.
  82. Fluck, C.E., et al., Mutant P450 oxidoreductase causes disordered steroidogenesis with and without Antley-Bixler syndrome. Nat Genet, 2004. 36(3): p. 228-30.
  83. Wilson, R.C., et al., Steroid 21-hydroxylase deficiency: genotype may not predict phenotype. J Clin Endocrinol Metab, 1995. 80(8): p. 2322-9.
  84. Nimkarn, S., et al., Congenital adrenal hyperplasia (21-hydroxylase deficiency) without demonstrable genetic mutations. J Clin Endocrinol Metab, 1999. 84(1): p. 378-81.
  85. Tajima, T., et al., Heterozygous mutation in the cholesterol side chain cleavage enzyme (p450scc) gene in a patient with 46,XY sex reversal and adrenal insufficiency. J Clin Endocrinol Metab, 2001. 86(8): p. 3820-5.
  86. Nebert, D.W., et al., The P450 superfamily: update on new sequences, gene mapping, and recommended nomenclature. DNA Cell Biol, 1991. 10(1): p. 1-14.
  87. Dupont, B., et al., Close genetic linkage between HLA and congenital adrenal hyperplasia (21-hydroxylase deficiency). Lancet, 1977. 2(8052-8053): p. 1309-12.
  88. White, P.C., et al., Two genes encoding steroid 21-hydroxylase are located near the genes encoding the fourth component of complement in man. Proc Natl Acad Sci U S A, 1985. 82(4): p. 1089-93.
  89. Carroll, M.C., R.D. Campbell, and R.R. Porter, Mapping of steroid 21-hydroxylase genes adjacent to complement component C4 genes in HLA, the major histocompatibility complex in man. Proc Natl Acad Sci U S A, 1985. 82(2): p. 521-5.
  90. Belt, K.T., M.C. Carroll, and R.R. Porter, The structural basis of the multiple forms of human complement component C4. Cell, 1984. 36(4): p. 907-14.
  91. White, P.C., M.I. New, and B. Dupont, Structure of human steroid 21-hydroxylase genes. Proc Natl Acad Sci U S A, 1986. 83(14): p. 5111-5.
  92. Higashi, Y., et al., Complete nucleotide sequence of two steroid 21-hydroxylase genes tandemly arranged in human chromosome: a pseudogene and a genuine gene. Proc Natl Acad Sci U S A, 1986. 83(9): p. 2841-5.
  93. New, M.I., 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): p. 2611-6.
  94. Finkielstain, G.P., et al., Comprehensive genetic analysis of 182 unrelated families with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Clin Endocrinol Metab, 2011. 96(1): p. E161-72.
  95. Krone, N., et al., Predicting phenotype in steroid 21-hydroxylase deficiency? Comprehensive genotyping in 155 unrelated, well defined patients from southern Germany. J Clin Endocrinol Metab, 2000. 85(3): p. 1059-65.
  96. Rocha, R.O., et al., The degree of external genitalia virilization in girls with 21-hydroxylase deficiency appears to be influenced by the CAG repeats in the androgen receptor gene. Clin Endocrinol (Oxf), 2008. 68(2): p. 226-32.
  97. Forest, M.G., Recent advances in the diagnosis and management of congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Hum Reprod Update, 2004. 10(6): p. 469-85.
  98. Pang, S., et al., Serum androgen concentrations in neonates and young infants with congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Clin Endocrinol (Oxf), 1979. 11(6): p. 575-84.
  99. Korth-Schutz, S., et al., Serum androgens as a continuing index of adequacy of treatment of congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1978. 46(3): p. 452-8.
  100. Caulfield, M.P., et al., The diagnosis of congenital adrenal hyperplasia in the newborn by gas chromatography/mass spectrometry analysis of random urine specimens. J Clin Endocrinol Metab, 2002. 87(8): p. 3682-90.
  101. Shackleton, C.H., Profiling steroid hormones and urinary steroids. J Chromatogr, 1986. 379: p. 91-156.
  102. Pang, S., et al., Microfilter paper method for 17 alpha-hydroxyprogesterone radioimmunoassay: its application for rapid screening for congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1977. 45(5): p. 1003-8.
  103. Therrell, B.L., et al., Current status of newborn screening worldwide: 2015. Semin Perinatol, 2015. 39(3): p. 171-87.
  104. Speiser, P.W., et al., Congenital adrenal hyperplasia due to steroid 21-hydroxylase deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2010. 95(9): p. 4133-60.
  105. Gruneiro-Papendieck, L., et al., Neonatal screening program for congenital adrenal hyperplasia: adjustments to the recall protocol. Horm Res, 2001. 55(6): p. 271-7.
  106. Janzen, N., et al., Newborn screening for congenital adrenal hyperplasia: additional steroid profile using liquid chromatography-tandem mass spectrometry. J Clin Endocrinol Metab, 2007. 92(7): p. 2581-9.
  107. New, M.I., et al., Genotyping steroid 21-hydroxylase deficiency: hormonal reference data. J Clin Endocrinol Metab, 1983. 57(2): p. 320-6.
  108. Jeffcoate, T.N., et al., Diagnosis of the adrenogenital syndrome before birth. Lancet, 1965. 2(7412): p. 553-5.
  109. Wilson, R.C., et al., Rapid deoxyribonucleic acid analysis by allele-specific polymerase chain reaction for detection of mutations in the steroid 21-hydroxylase gene. J Clin Endocrinol Metab, 1995. 80(5): p. 1635-40.
  110. Tukel, T., et al., A novel semiquantitative polymerase chain reaction/enzyme digestion-based method for detection of large scale deletions/conversions of the CYP21 gene and mutation screening in Turkish families with 21-hydroxylase deficiency. J Clin Endocrinol Metab, 2003. 88(12): p. 5893-7.
  111. Mao, R., et al., Prenatal diagnosis of 21-hydroxylase deficiency caused by gene conversion and rearrangements: pitfalls and molecular diagnostic solutions. Prenat Diagn, 2002. 22(13): p. 1171-6.
  112. Day, D.J., et al., Identification of non-amplifying CYP21 genes when using PCR-based diagnosis of 21-hydroxylase deficiency in congenital adrenal hyperplasia (CAH) affected pedigrees. Hum Mol Genet, 1996. 5(12): p. 2039-48.
  113. Lo, Y.M., et al., Presence of fetal DNA in maternal plasma and serum. Lancet, 1997. 350(9076): p. 485-7.
  114. Bischoff, F.Z., et al., Noninvasive determination of fetal RhD status using fetal DNA in maternal serum and PCR. J Soc Gynecol Investig, 1999. 6(2): p. 64-9.
  115. Chitty, L.S. and Y.M. Lo, Noninvasive Prenatal Screening for Genetic Diseases Using Massively Parallel Sequencing of Maternal Plasma DNA. Cold Spring Harb Perspect Med, 2015. 5(9): p. a023085.
  116. Johnson, K.L., et al., Interlaboratory comparison of fetal male DNA detection from common maternal plasma samples by real-time PCR. Clin Chem, 2004. 50(3): p. 516-21.
  117. Bischoff, F.Z., D.E. Lewis, and J.L. Simpson, Cell-free fetal DNA in maternal blood: kinetics, source and structure. Hum Reprod Update, 2005. 11(1): p. 59-67.
  118. Tardy-Guidollet, V., et al., New management strategy of pregnancies at risk of congenital adrenal hyperplasia using fetal sex determination in maternal serum: French cohort of 258 cases (2002-2011). J Clin Endocrinol Metab, 2014. 99(4): p. 1180-8.
  119. New, M.I., et al., Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab, 2014. 99(6): p. E1022-30.
  120. Simpson, J.L., Preimplantation genetic diagnosis at 20 years. Prenat Diagn, 2010. 30(7): p. 682-95.
  121. Fiorentino, F., et al., Strategies and clinical outcome of 250 cycles of Preimplantation Genetic Diagnosis for single gene disorders. Hum Reprod, 2006. 21(3): p. 670-84.
  122. David, M. and M.G. Forest, Prenatal treatment of congenital adrenal hyperplasia resulting from 21-hydroxylase deficiency. J Pediatr, 1984. 105(5): p. 799-803.
  123. New, M.I., et al., Prenatal diagnosis for congenital adrenal hyperplasia in 532 pregnancies. J Clin Endocrinol Metab, 2001. 86(12): p. 5651-7.
  124. Forest, M.G., M. David, and Y. Morel, Prenatal diagnosis and treatment of 21-hydroxylase deficiency. J Steroid Biochem Mol Biol, 1993. 45(1-3): p. 75-82.
  125. Mercado, A.B., et al., Prenatal treatment and diagnosis of congenital adrenal hyperplasia owing to steroid 21-hydroxylase deficiency. J Clin Endocrinol Metab, 1995. 80(7): p. 2014-20.
  126. Carlson, A.D., et al., Congenital adrenal hyperplasia: update on prenatal diagnosis and treatment. J Steroid Biochem Mol Biol, 1999. 69(1-6): p. 19-29.
  127. New, M.I., et al., An update on prenatal diagnosis and treatment of congenital adrenal hyperplasia. Semin Reprod Med, 2012. 30(5): p. 396-9.
  128. Motaghedi, R., et al., Update on the prenatal diagnosis and treatment of congenital adrenal hyperplasia due to 11beta-hydroxylase deficiency. J Pediatr Endocrinol Metab, 2005. 18(2): p. 133-42.
  129. Saenger, P., Abnormal sex differentiation. J Pediatr, 1984. 104(1): p. 1-17.
  130. Meyer-Bahlburg, H.F., et al., Gender development in women with congenital adrenal hyperplasia as a function of disorder severity. Arch Sex Behav, 2006. 35(6): p. 667-84.
  131. Seckl, J.R. and W.L. Miller, How safe is long-term prenatal glucocorticoid treatment? JAMA, 1997. 277(13): p. 1077-9.
  132. Seckl, J.R., Prenatal glucocorticoids and long-term programming. Eur J Endocrinol, 2004. 151 Suppl 3: p. U49-62.
  133. Yeh, T.F., et al., Outcomes at school age after postnatal dexamethasone therapy for lung disease of prematurity. N Engl J Med, 2004. 350(13): p. 1304-13.
  134. Slotkin, T.A., et al., Glucocorticoid administration alters nuclear transcription factors in fetal rat brain: implications for the use of antenatal steroids. Brain Res Dev Brain Res, 1998. 111(1): p. 11-24.
  135. Uno, H., et al., Brain damage induced by prenatal exposure to dexamethasone in fetal rhesus macaques. I. Hippocampus. Brain Res Dev Brain Res, 1990. 53(2): p. 157-67.
  136. Hirvikoski, T., et al., Cognitive functions in children at risk for congenital adrenal hyperplasia treated prenatally with dexamethasone. J Clin Endocrinol Metab, 2007. 92(2): p. 542-8.
  137. Hirvikoski, T., et al., Long-term follow-up of prenatally treated children at risk for congenital adrenal hyperplasia: does dexamethasone cause behavioural problems? Eur J Endocrinol, 2008. 159(3): p. 309-16.
  138. Meyer-Bahlburg, H.F., et al., Cognitive outcome of offspring from dexamethasone-treated pregnancies at risk for congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Eur J Endocrinol, 2012. 167(1): p. 103-10.
  139. Dorr, H.G., et al., Experts' Opinion on the Prenatal Therapy of Congenital Adrenal Hyperplasia (CAH) Due to 21-Hydroxylase Deficiency - Guideline of DGKED in cooperation with DGGG (S1-Level, AWMF Registry No. 174/013, July 2015). Geburtshilfe Frauenheilkd, 2015. 75(12): p. 1232-1238.
  140. Trautman, P.D., et al., Mothers' reactions to prenatal diagnostic procedures and dexamethasone treatment of congenital adrenal hyperplasia. J Psychosom Obstet Gynaecol, 1996. 17(3): p. 175-81.
  141. Lajic, S., et al., Prenatal treatment of congenital adrenal hyperplasia. Eur J Endocrinol, 2004. 151 Suppl 3: p. U63-9.
  142. Lajic, S., et al., Long-term somatic follow-up of prenatally treated children with congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1998. 83(11): p. 3872-80.
  143. Goldman, A.S., B.H. Sharpior, and M. Katsumata, Human foetal palatal corticoid receptors and teratogens for cleft palate. Nature, 1978. 272(5652): p. 464-6.
  144. Nandi, J., et al., In vitro steroidogenesis by the adrenal glands of mice. Endocrinology, 1967. 80(4): p. 576-82.
  145. Rosler, A., et al., Prenatal diagnosis of 11beta-hydroxylase deficiency congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1979. 49(4): p. 546-51.
  146. Schumert, Z., et al., 11-deoxycortisol in amniotic fluid: prenatal diagnosis of congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Clin Endocrinol (Oxf), 1980. 12(3): p. 257-60.
  147. Sippell, W.G., et al., Concentrations of aldosterone, corticosterone, 11-deoxycorticosterone, progesterone, 17-hydroxyprogesterone, 11-deoxycortisol, cortisol, and cortisone determined simultaneously in human amniotic fluid throughout gestation. J Clin Endocrinol Metab, 1981. 52(3): p. 385-92.
  148. Cerame, B.I., et al., Prenatal diagnosis and treatment of 11beta-hydroxylase deficiency congenital adrenal hyperplasia resulting in normal female genitalia. J Clin Endocrinol Metab, 1999. 84(9): p. 3129-34.
  149. Geley, S., et al., CYP11B1 mutations causing congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. J Clin Endocrinol Metab, 1996. 81(8): p. 2896-901.
  150. Prete, A., R.J. Auchus, and R.J. Ross, Clinical advances in the pharmacotherapy of congenital adrenal hyperplasia. Eur J Endocrinol, 2021. 186(1): p. R1-R14.
  151. Jones, C.M., et al., Modified-Release and Conventional Glucocorticoids and Diurnal Androgen Excretion in Congenital Adrenal Hyperplasia. J Clin Endocrinol Metab, 2017. 102(6): p. 1797-1806.
  152. Nimkarn, S., et al., Aldosterone-to-renin ratio as a marker for disease severity in 21-hydroxylase deficiency congenital adrenal hyperplasia. J Clin Endocrinol Metab, 2007. 92(1): p. 137-42.
  153. Rosler, A., et al., The interrelationship of sodium balance, plasma renin activity and ACTH in congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1977. 45(3): p. 500-12.
  154. New, M.I. and M.P. Seaman, Secretion rates of cortisol and aldosterone precursors in various forms of congenital adrenal hyperplasia. J Clin Endocrinol Metab, 1970. 30(3): p. 361-71.
  155. Canalis, E., Clinical review 83: Mechanisms of glucocorticoid action in bone: implications to glucocorticoid-induced osteoporosis. J Clin Endocrinol Metab, 1996. 81(10): p. 3441-7.
  156. Canalis, E., et al., Perspectives on glucocorticoid-induced osteoporosis. Bone, 2004. 34(4): p. 593-8.
  157. Guo, C.Y., A.P. Weetman, and R. Eastell, Bone turnover and bone mineral density in patients with congenital adrenal hyperplasia. Clin Endocrinol (Oxf), 1996. 45(5): p. 535-41.
  158. de Almeida Freire, P.O., et al., Classical congenital adrenal hyperplasia due to 21-hydroxylase deficiency: a cross-sectional study of factors involved in bone mineral density. J Bone Miner Metab, 2003. 21(6): p. 396-401.
  159. King, J.A., et al., Long-term corticosteroid replacement and bone mineral density in adult women with classical congenital adrenal hyperplasia. J Clin Endocrinol Metab, 2006. 91(3): p. 865-9.
  160. Riehl, G., et al., Bone mineral density and fractures in congenital adrenal hyperplasia: Findings from the dsd-LIFE study. Clin Endocrinol (Oxf), 2020. 92(4): p. 284-294.
  161. Lin-Su, K. and M.I. New, Effects of adrenal steroids on the bone metabolism of children with congenital adrenal hyperplasia. Ann N Y Acad Sci, 2007. 1117: p. 345-51.
  162. Crouch, N.S., et al., Sexual function and genital sensitivity following feminizing genitoplasty for congenital adrenal hyperplasia. J Urol, 2008. 179(2): p. 634-8.
  163. Nordenstrom, A., et al., Sexual function and surgical outcome in women with congenital adrenal hyperplasia due to CYP21A2 deficiency: clinical perspective and the patients' perception. J Clin Endocrinol Metab, 2010. 95(8): p. 3633-40.
  164. Claahsen-van der Grinten, H.L., et al., Congenital Adrenal Hyperplasia-Current Insights in Pathophysiology, Diagnostics, and Management. Endocr Rev, 2022. 43(1): p. 91-159.
  165. Bennecke, E., et al., Early Genital Surgery in Disorders/Differences of Sex Development: Patients' Perspectives. Arch Sex Behav, 2021. 50(3): p. 913-923.
  166. Mallappa, A., et al., Long-term use of continuous subcutaneous hydrocortisone infusion therapy in patients with congenital adrenal hyperplasia. Clin Endocrinol (Oxf), 2018. 89(4): p. 399-407.
  167. Mallappa, A., et al., A phase 2 study of Chronocort, a modified-release formulation of hydrocortisone, in the treatment of adults with classic congenital adrenal hyperplasia. J Clin Endocrinol Metab, 2015. 100(3): p. 1137-45.
  168. Whitaker, M.J., H. Huatan, and R.J. Ross, Chronotherapy based on modified-release hydrocortisone to restore the physiological cortisol diurnal rhythm. Drug Deliv Transl Res, 2022.
  169. Sarafoglou, K., et al., Tildacerfont in Adults With Classic Congenital Adrenal Hyperplasia: Results from Two Phase 2 Studies. J Clin Endocrinol Metab, 2021. 106(11): p. e4666-e4679.
  170. Auchus, R.J., et al., Crinecerfont Lowers Elevated Hormone Markers in Adults With 21-Hydroxylase Deficiency Congenital Adrenal Hyperplasia. J Clin Endocrinol Metab, 2022. 107(3): p. 801-812.
  171. Wright, C., et al., Abiraterone acetate treatment lowers 11-oxygenated androgens. Eur J Endocrinol, 2020. 182(4): p. 413-421.
  172. Auchus, R.J., et al., Abiraterone acetate to lower androgens in women with classic 21-hydroxylase deficiency. J Clin Endocrinol Metab, 2014. 99(8): p. 2763-70.

 

Physiology of the Pineal Gland and Melatonin

ABSTRACT

 

The pineal gland was described as the “Seat of the Soul” by Renee Descartes and it is located in the center of the brain. The main function of the pineal gland is to receive information about the state of the light-dark cycle from the environment and convey this information by the production and secretion of the hormone melatonin. Changing photoperiod is indicated by the duration of melatonin secretion and is used by photoperiodic species to time their seasonal physiology. The rhythmic production of melatonin, normally secreted only during the dark period of the day, is extensively used as a marker of the phase of the internal circadian clock. Melatonin itself is used as a therapy for certain sleep disorders related to circadian rhythm abnormalities such as delayed sleep phase syndrome, non-24h sleep wake disorder and jet lag. It might have more extensive therapeutic applications in the future, since multiple physiological roles have been attributed to melatonin. It exerts physiologic immediate effects during night or darkness and when suitably administered has prospective effects during daytime when melatonin levels are undetectable. In addition to its role in regulating seasonal physiology and influencing the circadian system and sleep patterns, melatonin is involved in cell protection, neuroprotection, and the reproductive system, among other possible functions. Pineal gland function and melatonin secretion can be impaired due to accidental and developmental conditions, such as pineal tumors, craniopharyngiomas, injuries affecting the sympathetic innervation of the pineal gland, and rare congenital disorders that alter melatonin secretion. This chapter summarizes the physiology and pathophysiology of the pineal gland and melatonin.

 

PINEAL PHYSIOLOGY

 

Pineal Anatomy and Structure

 

The pineal gland in humans is a small (100-150 mg), highly vascularized, and a secretory neuroendocrine organ (1). It is located in the mid-line of the brain, outside the blood-brain barrier and attached to the roof of the third ventricle by a short stalk. In humans, the pineal gland usually shows a degree of calcification with age providing a good imaging marker (for a speculative discussion of pineal calcification see reference(2)). The principal innervation is sympathetic, arising from the superior cervical ganglia (3). Arterial vascularization of the pineal gland is supplied by both the anterior and posterior circulation, being the main artery supplying the lateral pineal artery, which originates from the posterior circulation (4). In mammals, the main cell types are pinealocytes (95%) followed by scattered glial cells (astrocytic and phagocytic subtypes) (5). Pinealocytes are responsible for the synthesis and secretion of melatonin.

 

Main Function of the Pineal Gland

 

The main function of the pineal gland is to receive and convey information about the current light-dark cycle from the environment via the production and secretion of melatonin cyclically at night (dark period) (6, 7). Although in cold-blooded vertebrates (lower-vertebrate species), the pineal gland is photosensitive, this property is lost in higher vertebrates. In higher vertebrates, light is sensed by the inner retina (retinal ganglion cells) that send neural signals to the visual areas of the brain. However, a few retinal ganglion cells contain melanopsin and have an intrinsic photoreceptor capability that sends neural signals to non-image forming areas of the brain, including the pineal gland, through complex neuronal connections. The photic information from the retina is sent to the suprachiasmatic nucleus (SCN), the major rhythm-generating system or “clock” in mammals, and from there to other hypothalamic areas. When the light signal is positive, the SCN secretes gamma-amino butyric acid, responsible for the inhibition of the neurons that synapse in the paraventricular nucleus (PVN) of the hypothalamus, consequently the signal to the pineal gland is interrupted and melatonin is not synthesized. On the contrary, when there is no light (darkness), the SCN secretes glutamate, responsible for the PVN transmission of the signal along the pathway to the pineal gland. However, it is important to note that in continuous darkness the SCN continues to generate rhythmic output without light suppression since it functions as an endogenous oscillator (master pacemaker or clock). The rhythm deviates from 24h and ‘free-runs’ in the absence of the important light time cue. Light-dark cycles serve to synchronize the rhythm to 24h.

 

The PVN nucleus communicates with higher thoracic segments of the spinal column, conveying information to the superior cervical ganglion that transmits the final signal to the pineal gland through sympathetic postsynaptic fibers by releasing norepinephrine (NE).  NE is the trigger for the pinealocytes to produce melatonin by activating the transcription of the mRNA encoding the enzyme arylalkylamine N-acetyltransferase (AA-NAT), the first molecular step for melatonin synthesis (8) (Figure 1).

 

Figure 1. Melatonin synthesis in the pineal gland

 

MELATONIN SYNTHESIS AND METABOLISM

 

Melatonin Synthesis

 

Melatonin (N-acetyl-5-methoxytryptamine) is synthesized within the pinealocytes from tryptophan, mostly occurring during the dark phase of the day, when there is a major increase in the activity of serotonin-N-acetyltransferase (arylalkylamine N-acetyltransferase, AA-NAT), responsible for the transformation of 5-hydroxytryptamine (5HT, serotonin) to N-acetylserotonin (NAS) (Figure 1). Finally, N-acetylserotonin is converted to melatonin by acetylserotonin O-methyltransferase. The rapid decline in the synthesis with light treatment at night appears to depend on proteasomal proteolysis (9). Both AA-NAT and serotonin availability play a role limiting melatonin production. AA-NAT mRNA is expressed mainly in the pineal gland, retina, and to a lesser extent in some other brain areas, pituitary, and testis. Melatonin synthesis is also described in many other sites. AA-NAT activation is triggered by the activation of β1 and α1b adrenergic receptors by NE (9). NE is the major transmitter via β-1 adrenoceptors with potentiation by α-1 stimulation. NE levels are higher at night, approximately 180 degrees out of phase with the serotonin rhythm. Both availability of NE and serotonin are stimulatory for melatonin synthesis. Pathological, surgical or traumatic sympathetic denervation of the pineal gland or administration of β-adrenergic antagonists abolishes the rhythmic synthesis of melatonin and the light-dark control of its production.

 

There is evidence that melatonin can be synthesized in other sites of the body (skin, gastrointestinal tract, retina, bone marrow, placenta and others) acting in an autocrine or paracrine manner (10). Very recently it has been demonstrated that in the mouse brain melatonin is exclusively synthesized in the mitochondrial matrix. It is released to the cytoplasm, thereby activating a mitochondrial MT1 signal-transduction pathway which inhibits stress-mediated cytochrome c release and caspase activation: these are preludes to cell death and inflammation. This is a new mechanism whereby locally synthesized melatonin may protect against neurodegeneration. It is referred to as automitocrine signaling (11). Except for the pineal gland, other structures contribute little to circulating concentrations in mammals, since after pinealectomy, melatonin levels are mostly undetectable (12). Importantly several other factors, summarized in Table 1, have been related to the secretion and production of melatonin (13, 14).

 

Table 1. Factors Influencing Human Melatonin Secretion and Production (11),(12)

Factor

Effect(s) on melatonin

Comment

Light

Suppression

>30 lux white 460-480 nm most effective

Light

Phase-shift/ Synchronization

Short wavelengths most effective

Sleep timing

Phase-shift

Partly secondary to light exposure

Posture

­ standing (night)

 

Exercise

­ phase shifts

Hard exercise

ß-adrenoceptor-A

¯ synthesis

Anti-hypertensives

5HT UI

­ fluvoxamine

Metabolic effect

NE UI

­ change in timing

Antidepressants

MAOA I

­ may change phase

Antidepressants

α-adrenoceptor-A

¯ alpha-1, ­ alpha-2

 

Benzodiazepines

Variable¯ diazepam, alprazolam

GABA mechanisms

Testosterone

¯

Treatment

OC

­

 

Estradiol

¯? Not clear

 

Menstrual cycle

Inconsistent

­ amenorrhea

Smoking

Possible changes ­¯ ?

 

Alcohol

¯

Dose dependent

Caffeine

­

Delays clearance (exogenous)

Aspirin, Ibuprofen

¯

 

Chlorpromazine

­

Metabolic effect

Benserazide

Possible phase change, Parkinson patients

Aromatic amino-acid decarboxylase-I

Abbreviations: A: antagonist, U: uptake, I: inhibitor, MAO: monoamine oxidase, OC: oral contraceptives, 5HT: 5-hydroxytryptamine.

 

Control of Melatonin Synthesis: A Darkness Hormone

 

The rhythm of melatonin production is internally generated and controlled by interacting networks of clock genes in the bilateral SCN (15). Damage to the SCN leads to a loss of the majority of circadian rhythms. The SCN rhythm is synchronized to 24 hours mainly by the light-dark cycle acting via the retina and the retinohypothalamic projection to the SCN; the longer the night the longer the duration of secretion is, and the ocular light serves to synchronize the rhythm to 24h and to suppress secretion at the end of the dark phase, as explained above. Light exposure is the most important factor related to pineal gland function and melatonin secretion. A single daily light pulse of suitable intensity and duration in otherwise constant darkness is enough to phase shift and to synchronize the melatonin rhythm to 24h (16). The amount of light required at night to suppress melatonin secretion varies across species. In humans, intensities of 2500 lux full spectrum light (domestic light is around 100 to 500 lux) or light preferably in the blue range (460 to 480 nm) are required to completely suppress melatonin at night, but lower intensities < 200 lux can suppress secretion and shift the rhythm (17-21). Previous photic history influences the response: living in dim light increases sensitivity. Furthermore, the degree of light perception between individuals is related to the incidence of circadian desynchrony; along these lines, blind people with no conscious or unconscious light perception show free-running or abnormally synchronized melatonin and other circadian rhythms (22-24). Some blind subjects retain an intact retinohypothalamic tract and therefore a normal melatonin response despite a lack of conscious light perception (25, 26). It seems clear that an intact innervated pineal gland is necessary for the response to photoperiod change (27). Melatonin functions as a paracrine signal within the retina, it enhances retinal function in low intensity light by inducing photomechanical changes and provides a closed-loop to the pineal-retina-SCN system. All together, they are the basic structures to perceive and transduce non-visual effects of light, and to generate the melatonin rhythm by a closed-loop negative feedback of genes (Clock, “Circadian locomotor output cycles kaput” and Bmal, “Brain and muscle ARNT-like” genes), positive stimulatory elements (Per, “period and Cry, “Cryptochrome” genes), and negative elements (CCG, clock-controlled genes) of clock gene expression in the SCN (Figure 2).

Figure 2. Diagrammatic representation of the control of production and the functions of melatonin, regarding seasonal and circadian timing mechanisms. Abbreviations: SCN: suprachiasmatic nucleus, PVN: paraventricular nucleus, SCG: superior cervical ganglion, NA: norepinephrine (noradrenalin), RHT: retino-hypothalamic-tract, CCG: clock-controlled genes. Based on an original diagram by Dr Elisabeth Maywood, MRC Laboratory of Molecular Biology, Neurobiology Division, Hills Road Cambridge, CB2 2QH, UK.

Melatonin Metabolism

 

Once synthesized, melatonin is released directly into the peripheral circulation (bound to albumin) and to the CSF without being stored. In humans, melatonin’s half-life in blood is around 40 minutes and it is metabolized within the liver, converted to 6-hydroxymelatonin mainly by CYP1A2 and conjugated to 6-sulfatoxymelatonin (aMT6s) for subsequent urinary excretion. The measurement of urinary aMT6s is a good marker of melatonin secretion, since it follows the same pattern with an approximate 2-hours offset (28, 29). Overall, women have slightly higher values of plasma melatonin at night than men (30). On average, the maximum levels of plasma melatonin in adults occur between 02.00 and 04.00 hours and are on average about 60 to 70 pg/mL when measured with high-specificity assays. Concentrations in saliva, like other hormones, are three times lower than in plasma. Minimum concentrations detected are below 1 pg/mL. The plasma melatonin rhythm (timing and amplitude) strongly correlates with urinary aMT6s. Although there is a large variability in amplitude of the rhythm between subjects, the normal human melatonin rhythm is reproducible from day to day intraindividually (Figure 3 and 4).

 

Figure 3. Average concentrations of melatonin in human plasma (black, N=133), saliva (blue, N=28) and 6-sulphatoxymelatonin (aMT6s) in urine (red, N=88) using radioimmunoassay measurements. Diagrammatic representation of mean normal values (healthy men and women over 18 years old) from Dr. J. Arendt. Stockgrand Ltd., University of Surrey, UK.

Figure 4. Plasma melatonin and urinary aMT6s in hourly samples to show the delay in the rhythm of urinary aMT6s compared to plasma melatonin (mean/SEM, N=14). Red lines show typical urine sample collection times over 24-48h to determine timing of the rhythm in out-patient or field studies. Redrawn from R. Naidoo, Thesis, University of Surrey, UK, 1998.

Melatonin Production During Development and Across Life

 

At birth, melatonin levels are almost undetectable, the only fetal source of melatonin being via the placental circulation. Melatonin levels in fetal umbilical circulation reflect the day-night difference as seen in the maternal circulation. Maternal melatonin sends a temporal circadian signal to the fetus (called “maternal photoperiodic adaptative programming”), preparing the CNS to properly deal with environmental day/night fluctuations after birth. A melatonin rhythm appears around 2 to 3 months of life (31), levels increasing exponentially until a lifetime peak on average in prepubertal children; melatonin concentrations in children are associated with Tanner stages of puberty (32). Thereafter, a steady decrease occurs reaching mean adult concentrations in late teens (33, 34). Values are stable until 35 to 40 years, followed by a decline in amplitude of melatonin rhythm and lower levels with ageing, associated with fragmented sleep-wake patterns (35). In people >90 years, melatonin levels are less than 20% of young adult concentrations (36). The decline in age-related melatonin production is attributed to different reasons; calcification of the pineal gland starting early in life and an impairment in the noradrenergic innervation to the gland or light detection capacity (ocular mydriases, cataracts) (2, 37). Interestingly, pinealectomy accelerates the aging process and several reports suggest that melatonin has anti-aging properties (38).

 

MELATONIN’S MECHANISMS OF ACTION

 

Melatonin Target Sites and Receptors

 

Melatonin’s target sites are both central and peripheral. Binding sites have been found in many areas of the brain, including the pars tuberalis and hypothalamus, but also in the cells of the immune system, gonads, kidney, and the cardiovascular system (39, 40). Melatonin binding sites in the brain might vary according to species. Melatonin exerts both non-receptor and receptor-mediated actions. Non-receptor-mediated actions are due to amphipathic properties (of both lipo- and hydrophilic structure) that allows melatonin to freely cross the cell and nuclear membranes. It can be easily detected in the nucleus of several cells in the brain and peripheral organs (41). Antioxidant properties are one example of non-receptor-mediated actions of melatonin. On the other hand, melatonin has receptor-mediated actions.

 

Two types of a new family of G protein coupled melatonin receptors (MT) have been cloned in mammals (42). Melatonin receptors are widely expressed, often with overlapping distributions. MT1 is primarily expressed in the pars tuberalis, the SCN together with other hypothalamic areas, pituitary, hippocampus, and adrenal glands, suggesting that circadian and reproductive effects are mediated through this receptor. MT2 is mainly expressed in the SCN, retina, pituitary and the other brain areas, and is associated with phase shifting (43). The affinity of melatonin is five-fold greater for MT1 than MT2. Melatonin administration induces (1) an acute suppression of neuronal firing in the SCN via the MT1 receptor, and (2) a phase-shifting of SCN activity through the MT2 receptor. However, agonists that exclusively act on one or another receptor have not been identified as yet, and the understanding of the role of each receptor in most of the tissues in which both receptors are present is difficult. Also, melatonin receptors are transiently expressed in neuroendocrine tissues during development, suggesting that melatonin has a role as a neuroendocrine synchronizer in developmental physiology (44).

 

Chronobiotic Effects of Melatonin

Figure 5. Phase shifting of circadian rhythms. From data in Middleton B. et al., J. Sleep Res, 2002, 11, Suppl 1, p 154, Melatonin phase shifts all measured rhythms abstr. No. 309. Rajaratnam SW, et al., J Clin Endocrinol Metab 2003;88:4303-9. Rajaratnam SW, et al., J Physiol 2004; 561:339-351.

 

Phase shifting of circadian rhythms by melatonin (Figure 5) was first described in the 1980s (45). Melatonin has chronobiotic effects and is able to synchronize and reset biological oscillations. Melatonin acts on oscillators according to a well-defined phase-response curve (PRC). PRC is characterized by phase-advance zone (early in the evening before the beginning of the nocturnal melatonin production), phase-delay zone (in the late night/early morning hours), and a non-responsive dead.  zone (when melatonin levels are high) (46). The melatonin PRC is useful for the clinical administration of melatonin as a chronobiotic agent and treatment of sleep circadian and mood  disorders (47). In addition to the circadian chronobiotic effects, melatonin importantly has chronobiotic seasonal effects and acts as a circannual synchronizer, one season determined by increasing duration of the nocturnal melatonin production (in the direction of the winter solstice) and the other season defined by the reduction of nocturnal melatonin production (in the direction of the summer solstice) (8, 48). Interestingly, most of the clock genes are expressed in the pars tuberalis with a 24h rhythmicity different from their expression in the SCN (49), and these clock genes are influenced by melatonin with numerous potential seasonal effects. However, whether melatonin modulates the activity of the SCN via regulation of clock genes is unclear. Also, a central clock, independent of the SCN, can be entrained by food availability, temperature variations, forced activity and rest, drugs (melatonin itself), and timing (50).

 

MELATONIN PHYSIOLOGY AND PATHOPHYSIOLOGY

 

As previously stated, melatonin can act through several mechanisms and at almost all levels of the organism. Therefore, it has multiple and diversified actions with immediate (endogenous melatonin, during the night) or prospective effects (exogenous melatonin, during the previous day).

 

Hypomelatoninemia is more common, and it can be due to factors that affect directly the pineal gland, innervation, melatonin synthesis as a result of congenital disease; or secondary as a consequence of environmental factors and/or medications (shift work, spinal cord cervical transection, sympathectomy, aging, neurodegenerative diseases, genetic diseases, β-blockers, calcium channel blockers, ACE inhibitors). Hypermelatoninemia is less common, and except for pharmacological effects, few conditions have been associated with high melatonin production: spontaneous hypothermia, hyperhidrosis syndrome, polycystic ovary syndrome, hypogonadotropic hypogonadism, anorexia nervosa, and Rabson-Mendenhall syndrome that induces pineal hyperplasia.

 

Melatonin During Puberty, Menstrual Cycle and Reproductive Function

 

Neuroendocrine control of sexual maturation is influenced by the pattern of melatonin secretion as a consequence of the light-dark cycle, and in some species the photoperiod via melatonin secretion determines the timing of puberty (51). In vitro studies, in cultured prepuberal rat pituitary gland, showed that melatonin inhibits gonadotrophin-releasing hormone and therefore luteinizing hormone release, providing evidence for a potential causal role of melatonin in the timing of developmental stages (52) (Figure 2). The mechanism by which melatonin inhibits GnRH secretion is not clear; recently, kisspeptin was suggested to mediate this effect. Lower melatonin levels have been associated with precocious puberty and higher levels in delayed puberty and hypothalamic amenorrhea compared to age-matched controls; however, a causal role of melatonin in pubertal development has not been described (53, 54). Data on circulating melatonin, and its variation during the menstrual cycle, are inconsistent (55). In males, high melatonin doses (100 mg daily) potentiate testosterone-induced LH suppression, and a negative correlation between nocturnal serum LH and melatonin have been reported (56, 57). Nevertheless, attempts to develop melatonin as a contraceptive pill in combination with progestin have not been successful (58). Interestingly, people living near the Arctic circle present lower conception rates during winter darkness, when melatonin levels are high, than in summer. In summary, the overall perception in human studies is that melatonin is inhibitory to human reproductive function.

 

Melatonin and Core Body Temperature

 

Melatonin plays a role in circadian thermoregulation. In particular, the melatonin peak is associated with the nadir in body temperature, together with maximum tiredness, lowest alertness and performance (59) (Figure 6). Exogenous administration of melatonin during the daytime reproduces this association, increasing fatigue and sleepiness and decreasing body temperature, especially if the subject is seated (posture dependent effect) (59). Along these lines, the rise in temperature during the ovulatory phase of the menstrual cycle is associated with a decline in the amplitude of melatonin.

 

Figure 6. Relationship of plasma melatonin to other major circadian rhythms driven by the internal clock. Abbreviations: VAS: visual analogue scale. Reproduced from Rajaratnam SMW and Arendt J. Lancet 358:999-1005, 2001 by permission.

Melatonin and Energy Metabolism and Glucose Homeostasis

 

Melatonin is an important player in the regulation of energy metabolism and glucose homeostasis. It is responsible for the daily distribution of energy metabolism functions (daily phase of high insulin sensitivity, glycogen synthesis, and lipogenesis and a sleep phase associated with the usage of stored energy) (60). Interestingly, administration of melatonin in post-menopausal women induced a reduction in fat mass and increase in lean mass compared to placebo (61). Nocturnal melatonin secretion facilitates diurnal insulin sensitivity and preservation of beta cell mass and function (62) and low melatonin secretion has independently been associated to a higher risk for type 2 diabetes (T2DM) (63). In short sleepers or when there is a misalignment between waking time and melatonin secretion (melatonin secretion is not interrupted), insulin resistance and hyperglycemia can be observed. Melatonin administration can result in iatrogenic insulin resistance and hyperglycemia in the morning, depending when it is administered and on the metabolizing characteristics of the subject. Moreover, recently, some variants of the gene encoding for the MT-1B have been associated with reduced beta-cell function and increased risk for T2DM (64). Several cardiovascular effects have also been attributed to melatonin, such as, antihypertensive properties, regulation of heart rate, and vascular resistance (65, 66).

 

Melatonin, Antioxidant Properties and Cancer

 

Antioxidant and anti-aging properties have also been attributed to melatonin. Melatonin acts as a potent free radical scavenger and antioxidant in vitro, independently of the presence of the receptor (38, 67, 68) protecting lipids, protein and DNA from oxidative damage. It is more effective than glutathione in reducing oxidative stress under many circumstances, being highly concentrated in the mitochondria. Although most of these effects in humans have been observed in supraphysiological doses of melatonin, the quantity of exogenous melatonin required to generate relevant antioxidant activity is not well established. Moreover, the clinical benefits of antioxidant supplements are not clear.  Quote “research has not shown antioxidant supplements to be beneficial in preventing diseases”, https://www.nccih.nih.gov/health/antioxidants-in-depth#:~:text=Antioxidants%20are%20man%2Dmade%20or,be%20beneficial%20in%20preventing%20diseases.

 

Also, there is growing recent evidence for anti-tumoral activity of melatonin (69, 70). Melatonin seems to be useful as an oncostatic agent at the cellular level and slows the progression of cancer (most of the studies are focused on breast and prostate cancer). Interestingly, in rats when a carcinogen is given at night during the highest levels of melatonin, DNA damage is significantly lower (20%) than in rats that receive a carcinogen exposure during the day (71). Pinealectomy stimulates cancer initiation or growth in animals, and pineal calcification, that leads to a decline in melatonin production, have been associated with an increase in pediatric primary brain tumors (72).

 

An ’Umbrella review’, whereby the results of numerous meta-analyses are combined, has provided important data on which of the numerous claims for melatonin therapeutic benefit stand up to scrutiny. A simplified version is provided here (see below).

 

As mentioned previously, exposure to light during the “biological night” can suppress melatonin production, and it is also associated to a deleterious effect on health (i.e., increased risk of cancer in most epidemiology studies in night shift workers) (70, 73). Night-shift workers present a higher incidence of hormone-dependent cancer which has been related to light-induced melatonin suppression which consequently increases estrogen production. A 50% increased risk to develop breast cancer in nurses exposed to rotating shift work has been documented (74). In contrast, a reduced risk for breast cancer was observed in blind women, with potentially higher levels of melatonin throughout all day (although there is no evidence that blind people produce more melatonin than sighted people) However, the mechanisms by which melatonin exerts any of oncostatic effects remains to be established.

 

Miscellaneous

 

The circadian annual rhythm of prolactin secretion depends on the circadian melatonin signal. Melatonin regulates pars tuberalis timer cells, and coordinates prolactin-secreting cells which together function as an intrapituitary pacemaker timer system. Interestingly, several clock genes are expressed within the pituitary with a circadian rhythm independent of the SCN (75).

 

Melatonin might act on the adrenal glands as an endogenous pacemaker. Glucocorticoids levels are low after the onset of darkness and rise after the middle of the night concomitantly with a decrease in melatonin levels. This is consistent with an inhibitory effect of melatonin through MT1 on glucocorticoid production. Interestingly, many antidepressant drugs increase the availability of the precursors (tryptophan and serotonin) and the major pineal neurotransmitter NE and therefore stimulate melatonin secretion. A melatonin agonist has been developed as an antidepressant through its actions on serotonin 2C receptors (76). If there is a link between this increase in melatonin production and the efficacy of antidepressant medications this needs further evaluation.

 

Several conditions (congenital or acquired) might induce a dysregulation of the melatonin synthesis/signaling and affect rhythmic melatonin production:

 

Pathological or traumatic sympathetic denervation (i.e., injury to the spinal cord) of the pineal gland or administration of β-adrenergic antagonists abolishes the rhythmic synthesis of melatonin and the light-dark control of its production (77, 78). NE is the major transmitter via β1 adrenoceptors with potentiation by α1 stimulation. NE levels are higher at night, approximately 180 degrees out of phase with the serotonin rhythm. Both availability of NE and serotonin are stimulatory for melatonin synthesis. However, several other factors have been related to the secretion and production of melatonin (13) (Table 1). In humans, administration of atenolol suppresses melatonin and enhances the magnitude of light-induced phase shift (79). In fact, several related observations suggest that endogenous melatonin acts to counter undesirable abrupt changes in phase. Pinealectomy i.e., abolishment of the melatonin rhythm, leads to a more rapid circadian adaptation to phase shift in rats (80).

 

Craniopharyngioma patients often display low melatonin secretion as a result of SCN impairment associated with alterations in sleep pattern (81).

 

Smith-Magenis syndrome (a congenital disorder due to a haplo-insufficiency of the retinoic acid-induced 1 gene, involved in the regulation of the expression of circadian genes) patients present with an inverted rhythmic melatonin secretion and sleep difficulties that can be managed with melatonin administration in the evening and β-adrenergic blockers during the day to reduce melatonin secretion (82). Low levels of melatonin have been consistently associated with autism spectrum disorders (ASD) (83). The last enzyme in the synthesis of melatonin, acetylserotonin-O-methyltransferase, has been associated with susceptibility for ASD which could be an explanation for low melatonin levels in ASD (84). Interestingly, administration of melatonin has shown to be efficacious for insomnia in children with ASD (85).

 

MELATONIN, CLINICAL APPLICATION AND THERAPEUTIC USE

 

Literature on the clinical use of melatonin is extensive and has increased exponentially over the last decade. Melatonin effects on sleep are the most well-known; however, the finding of increased cancer risk in shift workers and that patients with neurodegenerative diseases, autism or depression present abnormal melatonin rhythms, have recently increased attention on the role of melatonin.

 

Melatonin and Sleep

 

Numerous factors and comorbidities are associated with chronic insomnia in the adult population, different from the circadian sleep-wake rhythm disorders. Some of these risk factors and comorbidities involve psychiatric conditions (i.e. depression, anxiety, posttraumatic stress disorder), medical conditions (i.e. pulmonary diseases, chronic pain, heart failure, hyperthyroidism, nocturia, gastroesophageal reflux, cancer, pregnancy, pruritus, HIV infection, obstructive sleep apnea syndrome), neurological conditions (i.e. Alzheimer’s, Parkinson’s disease, peripheral neuropathies, strokes, brain tumors, headache syndromes), and medications (i.e. central nervous system stimulants or depressants, bronchodilators, antidepressants, diuretics, glucocorticoids, caffeine, alcohol). All these conditions should be ruled out before establishing the suspicion of a circadian sleep-wake rhythm disorder. The importance of melatonin levels for human sleep was apparently demonstrated by studies on pinealectomized subjects. These patients presented a disrupted 24h circadian rhythm, and a reduction of sleep time and quality, that were reversed after the administration of melatonin (86, 87). However, another careful, prospective study using polysomnography before and following pinealectomy found no effect on sleep and similar data are reported in rats (88-90). Thus, the question is not resolved. Clearly it is not a sleep hormone since in nocturnal animals it is secreted during the active periods. Overall, exogenous melatonin has been shown consistently to reduce sleep latency, and less consistently increase total sleep time, reduce night awakenings, and ultimately improve sleep quality (91). The most obvious action is to optimize sleep timing with respect to the circadian clock: we sleep better when melatonin production (and thus the circadian clock) and sleep are correctly aligned. Diagnostic criteria of every circadian sleep-wake rhythm disorder were fully described by the American Academy of Sleep Medicine (2014) (92).

 

CIRCADIAN SLEEP WAKE-RHYTHM DISORDERS EVALUATION

 

The origin of a circadian rhythm disorder can be related either to an intrinsic abnormality in the circadian system itself (misalignment of the intrinsic circadian timing with the desired sleep schedule) or to external factors, as when individuals must be awake at times that are not synchronized with their intrinsic rhythms. Overall, sleep disorders result in clinically significant symptoms of insomnia, daytime sleepiness and impaired physical, neurocognitive (concentration, processing speed, memory), emotional and social functioning, as well as impaired functioning at work or school (92). Age and associated comorbidities can help in the differential diagnoses. Symptoms across sleeping disorders are non-specific; the key to properly identify them is the recognition of abnormal sleep-wake patterns using sleep diaries beyond the complaint of insomnia or daytime sleepiness (93). Time domain is very important for melatonin actions (immediate or prospective effects, chronobiotic, or seasonal effects); therefore, adequate melatonin measurement and time of administration are crucial. Actigraphy can supplement self-reported sleep diary information or be useful in situations of neurodevelopmental disorders where a sleep diary cannot be completed.

 

Melatonin concentrations typically increase 90 to 120 minutes prior to the habitual bedtime if bright light (>10 lux) is absent. Preferentially frequent blood sampling every 30 to 60 minutes from six hours prior to and one hour following habitual bedtime is collected to assess the individual’s dim light melatonin onset (DLMO) and circadian phase. Samples should be collected in a dark environment (red light <10 lux) without interfering with sleep. In research studies a DLMO protocol is used as a valid and reliable indicator of circadian phase position, according to the time at which melatonin levels rise, and is extremely helpful to guide the timing for melatonin therapy in each individual; however, this is rarely feasible for routine use in clinical settings (94). Alternatively, urinary excretion of 6-sulfatoxymelatonin is a good index of nocturnal melatonin production and it should be collected as sequential urine samples for 48h in order to calculate the timing of the peak or acrophase. Also, it is possible to measure melatonin in saliva, during the evening before sleep and this approach has been frequently used. But, in the case of for example very delayed melatonin rise the time of onset can be missed during sleep. Importantly, melatonin values vary individually and according to age and sex. Although, endogenous melatonin production is closely related with the onset and offset of sleep, few associations have been found between melatonin production and sleep stages (95), and sleep deprivation does not abolish the melatonin rhythm.

 

CIRCADIAN SLEEP WAKE-RHYTHM DISORDERS

 

The primary goal of treatment of circadian sleep-wake rhythm disorders is to realign the circadian timing of sleep and wake with the required sleep-wake period. Appropriately timed and dosed melatonin, melatonin receptor agonists, and light therapy seem to be useful approaches (96, 97). Following the melatonin PRC and taking the individual DLMO as the reference phase to decide the most suitable time of melatonin administration, according to the desirable effect, seem the most appropriate guide to decide when to start chronic melatonin therapy (98-100).

                                             

Delayed and advanced sleep-wake phase disorders, non-24 hour, and irregular sleep-wake rhythm disorders are considered intrinsic circadian disorders; in contrast, jet lag and shift work disorders are due to an environmentally imposed misalignment. The most potent phase shifting factor (zeitgeber) is the environmental light-dark cycle (101).

 

Light exposure during the last hours of the usual sleep period moves the circadian rhythm forward (phase advanced). On the contrary, light exposure in the evening and the first half of the usual sleep period moves the circadian rhythm back (phase delayed) (Figure 7) (96). Advanced sleep-wake phase disorder is partially due to the physiologic advance that occurs with aging together with the weakening of circadian rhythms observed with aging. It can be genetically determined in some families as well. Delayed sleep-wake phase disorder is more often seen in children with some neurodevelopmental disorders; these patients cannot sleep during the dark time and delay sleep onset until the early hours of the morning, sleeping much of the day. In these situations both bright light in the early morning and evening melatonin 5 hours before endogenous melatonin onset secretion (0.5-5 mg) has been shown to advance sleep time significantly (102), the magnitude of the advanced shift being dose-dependent. In contrast early “biological morning” administration of melatonin induces delayed shifts. The non-24-hour sleep-wake rhythm disorder is most commonly seen in blindness, since the light-dark cycle is the most powerful cue for synchronizing the hypothalamic pacemaker to the 24-hour day (103). Therefore, this disorder is characterized by a failure to maintain stable alignment to the 24-h day, and a “free-running” circadian rhythm system which usually shifts to a later and later phase position. Timed melatonin treatment has been successful in entraining free-running blind patients.

 

The irregular sleep-wake rhythm disorder is seen in patients with dementia, which a neurodegenerative process might induce a disruption of the circadian system with a consequent loss of modulatory influence on sleep and wakefulness (104).

 

Figure 7. Representation of a simplified diagram of phase shifts of the circadian system, as evidenced by changes in melatonin rhythm itself, following oral treatment with fast release melatonin at different times. Biological night” is the time of endogenous melatonin secretion and defines “circadian time” which is independent of clock time.

 

Jet lag occurs when crossing time zones, and one needs to sleep and be awake at times that are not aligned with one’s own circadian system. It is more severe when more time zones are crossed and if the direction of the travel is eastbound, as it is more difficult to advance than delay the natural circadian cycle. Melatonin accelerates the phase shift if given at the appropriate time prior to bedtime at destination, and the benefits of melatonin administration in the alleviation of jet lag seems to be greater with larger numbers of time zones (105). If melatonin is appropriately time-administered, self-rated jet lag can be reduced by 50 percent (106). The maximum advance shift obtainable with a single treatment of oral melatonin (3-5 mg) is approximately 1-1.5 h.

 

For night shift workers wakefulness is required at the time that melatonin secretion is rising, and alertness is dissipating, and the opposite for the next day when melatonin secretion decreases, and circadian rhythms promote alertness. While some studies suggested that the use of melatonin improved sleep and increase daytime alertness in night shift workers compared to placebo (107), other studies have not shown beneficial effects (108). More data are needed before recommendations about melatonin as a therapy for shift workers can be made. However anecdotal evidence suggests that it is widely used for daytime sleep.

 

Figure 8. Diagram to illustrate free-running of the sleep wake cycle and other circadian rhythms in non-24h sleep-wake disorder, frequently seen in totally blind people. Melatonin, usually in doses of 0.5 – 5mg daily can synchronize sleep and the circadian system to the 24h day with benefits for sleep, and daytime alertness.

 

Melatonin and the CNS

 

Individuals with neurodegenerative disorders (i.e., Alzheimer’s and Parkinson’s disease, Huntington’s disease, autism) had significant flattened and attenuated melatonin rhythms compared to age-matched controls (37, 109). Melatonin showed efficacy in managing the insomnia in elderly people or improving cognitive function associated with neurodegenerative disorders (110). It has reported effects on anxiety, cognitive function and memory. Knowing the wide distribution of melatonin receptors in the CNS, melatonin seems one of the promising neuroprotective agents to be tested in humans. However other studies have found deleterious effects of melatonin in elderly demented patients (111).

 

Melatonin: Therapeutic Use

 

Almost 200 randomized clinical trials on the use of melatonin and evaluation of clinical effects have been published. Moreover, several patents have been registered in relation to the therapeutic applications of melatonin and melatonin analogues (sleep disorders, neuroprotection, cancer). The American Academy of Sleep Science recommends the use of melatonin for jet lag, delayed sleep phase syndrome, and non-24h sleep wake disorder syndrome seen in blind people; however, there were few consensuses acknowledging its therapeutic benefits (112) until an ‘Umbrella’ review of meta-analyses identified consistent therapeutic targets (91). Interestingly, at least in animal models, appropriate melatonin dose administration can reverse most of the effects after a pinealectomy.

 

Table 2. Melatonin’s Reported Significant Clinical Effects

• Breast cancer, risk of death down at 1 year

• Nocturnal hypertension, systolic and diastolic reduced

• Sleep latency shortened

• Sleep duration increased

• Melatonin onset advanced

• Core body temperature decreased

• In rodents, protective effects of melatonin in ischemic stroke

Combined meta-analyses of studies meeting strict criteria (simplified from Posadzki PP et al. BMC Med (2018) 16: 18.)

 

Several melatonin receptor agonists have been synthesized, some of them have higher affinity for the receptor than endogenous melatonin. Agomelatine (activity at the serotonin-2C receptor) functions as an anti-depressant, ramelteon (selective MT1/MT2 agonist) is marketed for use in sleep onset insomnia, tasimelteon (MT1/MT2 agonist), is commercialized for the treatment of circadian rhythm disorders particularly non-24h sleep wake disorder (113, 114). Also, slow-release formulations of the natural biologic melatonin have been developed.

 

The time of melatonin administration is critical, especially in chronic treatments, and the melatonin profile shows large interindividual variation; however, the profile of an individual is highly reproducible from day to day. Also, absorption, metabolism and excretion of melatonin vary between individuals and should be considered to get the desired clinical efficacy of the therapy. Ideally, although not feasible in a daily clinical practice, DLMO should be determined for each individual, and used as a timing reference for the prescription of melatonin. Alternatively, the time each individual goes to sleep could determine the time of administration of melatonin; it is advisable to take oral melatonin around 45 minutes to 1 hour before the usual bedtime (time to reach maximal plasma concentrations following oral immediate-release formulations). Moreover, the duration of the pharmacological profile should last until the usual wake time of the patient; thus, the type of pharmaceutical formulation (slow or fast release) is also an important consideration. However, there is little evidence for greater benefit of slow-release preparations. Route of administration, as well as age, liver function and potential drug interactions (since melatonin is metabolized in the liver), may influence plasma melatonin levels and should be taken into account (115). Sensitivities and pharmacokinetics of melatonin vary between individuals, and a lower dose of 0.3-0.5 mg might be more effective than higher doses in many subjects.

 

There is no general consensus regarding dosage. A wide range of dose formulations are available, and the usage varies depending on the clinical application. The usual advice is to start with the lowest dose available. Low doses 0.1 to 0.3 mg/d that produce near physiological melatonin concentrations can be used for central clock synchronization; doses ranging from 0.6 to 5 mg/d for sleep disorders, or doses as high as 300 mg/d for neurodegenerative disorders (amyotrophic lateral sclerosis) (116, 117). There is a current tendency to recommend high pharmacological doses for ‘protective’ or antioxidant effects. What consequences these might have for the circadian aspects of melatonin function is not known. It should not be forgotten that melatonin has profound effects on the reproductive function of photoperiodic seasonal breeders and there is good evidence for residual photoperiodicity in humans. However in most clinical trials or studies using melatonin, it is well accepted that in general melatonin lacks toxic adverse events, and it is a safe drug at most of the usual tested doses from 0.5 to 5 mg/d (118). Whether dosage should be changed in chronic melatonin treatment according to the annual season requires further investigation; further studies are needed on the undesirable consequences of melatonin suppression in the long term (for example by beta blocking drugs or shiftwork).

 

To summarize, benefits of melatonin and its analogues on circadian sleep disorders are consistently reported but for more generalized “insomnia” are often of low strength. The dose must be timed and individually adjusted as optimization will vary among individuals. More studies are required to develop treatment guidelines for different conditions. The long-term consequences of taking high dose melatonin especially in pediatrics need careful evaluation. Melatonin mechanisms of action and effects need much further work, to better understand the therapeutic value of melatonin and its potential applications.

 

REFERENCES

 

  1. L V. The Pineal Organ, Handbook of Mikroscopic Anatomy1981.
  2. Tan DX, Xu B, Zhou X, Reiter RJ. Pineal Calcification, Melatonin Production, Aging, Associated Health Consequences and Rejuvenation of the Pineal Gland. Molecules. 2018;23(2).
  3. Kappers JA. Innervation of the Vertebrate Pineal Organ. In: Axelrod J, Fraschini, F., Velo, G.P. (eds) editor1983.
  4. Kahilogullari G, Ugur HC, Comert A, Brohi RA, Ozgural O, Ozdemir M, et al. Arterial vascularization of the pineal gland. Childs Nerv Syst. 2013;29(10):1835-41.
  5. Moller M, Baeres FM. The anatomy and innervation of the mammalian pineal gland. Cell Tissue Res. 2002;309(1):139-50.
  6. Goldman BD. Mammalian photoperiodic system: formal properties and neuroendocrine mechanisms of photoperiodic time measurement. J Biol Rhythms. 2001;16(4):283-301.
  7. JA. A. Melatonin and the Mammalian Pineal Gland.1995.
  8. Cipolla-Neto J, Amaral FGD. Melatonin as a Hormone: New Physiological and Clinical Insights. Endocr Rev. 2018;39(6):990-1028.
  9. Gastel JA, Roseboom PH, Rinaldi PA, Weller JL, Klein DC. Melatonin production: proteasomal proteolysis in serotonin N-acetyltransferase regulation. Science (New York, NY). 1998;279(5355):1358-60.
  10. Bubenik GA. Localization, physiological significance and possible clinical implication of gastrointestinal melatonin. Biol Signals Recept. 2001;10(6):350-66.
  11. Suofu Y, Li W, Jean-Alphonse FG, Jia J, Khattar NK, Li J, et al. Dual role of mitochondria in producing melatonin and driving GPCR signaling to block cytochrome c release. Proc Natl Acad Sci U S A. 2017;114(38):E7997-E8006.
  12. Lewy AJ, Tetsuo M, Markey SP, Goodwin FK, Kopin IJ. Pinealectomy abolishes plasma melatonin in the rat. J Clin Endocrinol Metab. 1980;50(1):204-5.
  13. Simonneaux V, Ribelayga C. Generation of the melatonin endocrine message in mammals: a review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev. 2003;55(2):325-95.
  14. Zawilska JB, Skene DJ, Arendt J. Physiology and pharmacology of melatonin in relation to biological rhythms. Pharmacol Rep. 2009;61(3):383-410.
  15. Takahashi JS, Hong HK, Ko CH, McDearmon EL. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet. 2008;9(10):764-75.
  16. Illnerova H, Vanecek J. Entrainment of the rat pineal rhythm in melatonin production by light. Reprod Nutr Dev (1980). 1988;28(2B):515-26.
  17. Laakso ML, Hatonen T, Stenberg D, Alila A, Smith S. One-hour exposure to moderate illuminance (500 lux) shifts the human melatonin rhythm. J Pineal Res. 1993;15(1):21-6.
  18. Gooley JJ, Chamberlain K, Smith KA, Khalsa SB, Rajaratnam SM, Van Reen E, et al. Exposure to room light before bedtime suppresses melatonin onset and shortens melatonin duration in humans. J Clin Endocrinol Metab. 2011;96(3):E463-72.
  19. Reiter RJ. Action spectra, dose-response relationships, and temporal aspects of light's effects on the pineal gland. Ann N Y Acad Sci. 1985;453:215-30.
  20. Bojkowski CJ, Aldhous ME, English J, Franey C, Poulton AL, Skene DJ, et al. Suppression of nocturnal plasma melatonin and 6-sulphatoxymelatonin by bright and dim light in man. Horm Metab Res. 1987;19(9):437-40.
  21. Zeitzer JM, Dijk DJ, Kronauer R, Brown E, Czeisler C. Sensitivity of the human circadian pacemaker to nocturnal light: melatonin phase resetting and suppression. J Physiol. 2000;526 Pt 3:695-702.
  22. Klerman EB, Rimmer DW, Dijk DJ, Kronauer RE, Rizzo JF, 3rd, Czeisler CA. Nonphotic entrainment of the human circadian pacemaker. Am J Physiol. 1998;274(4 Pt 2):R991-6.
  23. Lewy AJ, Newsome DA. Different types of melatonin circadian secretory rhythms in some blind subjects. J Clin Endocrinol Metab. 1983;56(6):1103-7.
  24. Lockley SW, Skene DJ, Arendt J, Tabandeh H, Bird AC, Defrance R. Relationship between melatonin rhythms and visual loss in the blind. J Clin Endocrinol Metab. 1997;82(11):3763-70.
  25. Czeisler CA, Shanahan TL, Klerman EB, Martens H, Brotman DJ, Emens JS, et al. Suppression of melatonin secretion in some blind patients by exposure to bright light. The New England journal of medicine. 1995;332(1):6-11.
  26. Hull JT, Czeisler CA, Lockley SW. Suppression of Melatonin Secretion in Totally Visually Blind People by Ocular Exposure to White Light: Clinical Characteristics. Ophthalmology. 2018;125(8):1160-71.
  27. Malpaux B, Migaud M, Tricoire H, Chemineau P. Biology of mammalian photoperiodism and the critical role of the pineal gland and melatonin. J Biol Rhythms. 2001;16(4):336-47.
  28. Bojkowski CJ, Arendt J, Shih MC, Markey SP. Melatonin secretion in humans assessed by measuring its metabolite, 6-sulfatoxymelatonin. Clin Chem. 1987;33(8):1343-8.
  29. Arendt J, Bojkowski C, Franey C, Wright J, Marks V. Immunoassay of 6-hydroxymelatonin sulfate in human plasma and urine: abolition of the urinary 24-hour rhythm with atenolol. J Clin Endocrinol Metab. 1985;60(6):1166-73.
  30. Cain SW, Dennison CF, Zeitzer JM, Guzik AM, Khalsa SB, Santhi N, et al. Sex differences in phase angle of entrainment and melatonin amplitude in humans. J Biol Rhythms. 2010;25(4):288-96.
  31. Kennaway DJ, Stamp GE, Goble FC. Development of melatonin production in infants and the impact of prematurity. J Clin Endocrinol Metab. 1992;75(2):367-9.
  32. Waldhauser F, Weiszenbacher G, Frisch H, Zeitlhuber U, Waldhauser M, Wurtman RJ. Fall in nocturnal serum melatonin during prepuberty and pubescence. Lancet. 1984;1(8373):362-5.
  33. Wetterberg L, Bergiannaki JD, Paparrigopoulos T, von Knorring L, Eberhard G, Bratlid T, et al. Normative melatonin excretion: a multinational study. Psychoneuroendocrinology. 1999;24(2):209-26.
  34. Kennaway DJ, Lushington K, Dawson D, Lack L, van den Heuvel C, Rogers N. Urinary 6-sulfatoxymelatonin excretion and aging: new results and a critical review of the literature. J Pineal Res. 1999;27(4):210-20.
  35. Bojkowski CJ, Arendt J. Factors influencing urinary 6-sulphatoxymelatonin, a major melatonin metabolite, in normal human subjects. Clin Endocrinol (Oxf). 1990;33(4):435-44.
  36. Scholtens RM, van Munster BC, van Kempen MF, de Rooij SE. Physiological melatonin levels in healthy older people: A systematic review. J Psychosom Res. 2016;86:20-7.
  37. Jengeleski CA, Powers RE, O'Connor DT, Price DL. Noradrenergic innervation of human pineal gland: abnormalities in aging and Alzheimer's disease. Brain Res. 1989;481(2):378-82.
  38. Karasek M. Melatonin, human aging, and age-related diseases. Exp Gerontol. 2004;39(11-12):1723-9.
  39. Reppert SM, Weaver DR, Godson C. Melatonin receptors step into the light: cloning and classification of subtypes. Trends Pharmacol Sci. 1996;17(3):100-2.
  40. Dubocovich ML, Markowska M. Functional MT1 and MT2 melatonin receptors in mammals. Endocrine. 2005;27(2):101-10.
  41. Costa EJ, Lopes RH, Lamy-Freund MT. Permeability of pure lipid bilayers to melatonin. J Pineal Res. 1995;19(3):123-6.
  42. Dubocovich ML, Delagrange P, Krause DN, Sugden D, Cardinali DP, Olcese J. International Union of Basic and Clinical Pharmacology. LXXV. Nomenclature, classification, and pharmacology of G protein-coupled melatonin receptors. Pharmacol Rev. 2010;62(3):343-80.
  43. Reppert SM. Melatonin receptors: molecular biology of a new family of G protein-coupled receptors. J Biol Rhythms. 1997;12(6):528-31.
  44. Johnston JD, Klosen P, Barrett P, Hazlerigg DG. Regulation of MT melatonin receptor expression in the foetal rat pituitary. J Neuroendocrinol. 2006;18(1):50-6.
  45. Arendt J, Bojkowski C, Folkard S, Franey C, Marks V, Minors D, et al. Some effects of melatonin and the control of its secretion in humans. Ciba Found Symp. 1985;117:266-83.
  46. Armstrong SM, Redman JR. Melatonin: a chronobiotic with anti-aging properties? Med Hypotheses. 1991;34(4):300-9.
  47. Lewy AJ, Ahmed S, Jackson JM, Sack RL. Melatonin shifts human circadian rhythms according to a phase-response curve. Chronobiol Int. 1992;9(5):380-92.
  48. Arendt J, Middleton B. Human seasonal and circadian studies in Antarctica (Halley, 75 degrees S). Gen Comp Endocrinol. 2018;258:250-8.
  49. Carr AJ, Johnston JD, Semikhodskii AG, Nolan T, Cagampang FR, Stirland JA, et al. Photoperiod differentially regulates circadian oscillators in central and peripheral tissues of the Syrian hamster. Curr Biol. 2003;13(17):1543-8.
  50. Fuller PM, Lu J, Saper CB. Differential rescue of light- and food-entrainable circadian rhythms. Science (New York, NY). 2008;320(5879):1074-7.
  51. Ebling FJ, Foster DL. Pineal melatonin rhythms and the timing of puberty in mammals. Experientia. 1989;45(10):946-54.
  52. Martin JE, Sattler C. Selectivity of melatonin pituitary inhibition for luteinizing hormone-releasing hormone. Neuroendocrinology. 1982;34(2):112-6.
  53. Waldhauser F, Boepple PA, Schemper M, Mansfield MJ, Crowley WF, Jr. Serum melatonin in central precocious puberty is lower than in age-matched prepubertal children. J Clin Endocrinol Metab. 1991;73(4):793-6.
  54. Berga SL, Mortola JF, Yen SS. Amplification of nocturnal melatonin secretion in women with functional hypothalamic amenorrhea. J Clin Endocrinol Metab. 1988;66(1):242-4.
  55. Parry BL, Berga SL, Mostofi N, Klauber MR, Resnick A. Plasma melatonin circadian rhythms during the menstrual cycle and after light therapy in premenstrual dysphoric disorder and normal control subjects. J Biol Rhythms. 1997;12(1):47-64.
  56. Anderson RA, Lincoln GA, Wu FC. Melatonin potentiates testosterone-induced suppression of luteinizing hormone secretion in normal men. Hum Reprod. 1993;8(11):1819-22.
  57. Luboshitzky R, Wagner O, Lavi S, Herer P, Lavie P. Abnormal melatonin secretion in hypogonadal men: the effect of testosterone treatment. Clin Endocrinol (Oxf). 1997;47(4):463-9.
  58. Voordouw BC, Euser R, Verdonk RE, Alberda BT, de Jong FH, Drogendijk AC, et al. Melatonin and melatonin-progestin combinations alter pituitary-ovarian function in women and can inhibit ovulation. J Clin Endocrinol Metab. 1992;74(1):108-17.
  59. Cagnacci A, Elliott JA, Yen SS. Melatonin: a major regulator of the circadian rhythm of core temperature in humans. J Clin Endocrinol Metab. 1992;75(2):447-52.
  60. Picinato MC, Haber EP, Carpinelli AR, Cipolla-Neto J. Daily rhythm of glucose-induced insulin secretion by isolated islets from intact and pinealectomized rat. J Pineal Res. 2002;33(3):172-7.
  61. Amstrup AK, Sikjaer T, Pedersen SB, Heickendorff L, Mosekilde L, Rejnmark L. Reduced fat mass and increased lean mass in response to 1 year of melatonin treatment in postmenopausal women: A randomized placebo-controlled trial. Clin Endocrinol (Oxf). 2016;84(3):342-7.
  62. Costes S, Boss M, Thomas AP, Matveyenko AV. Activation of Melatonin Signaling Promotes beta-Cell Survival and Function. Mol Endocrinol. 2015;29(5):682-92.
  63. McMullan CJ, Schernhammer ES, Rimm EB, Hu FB, Forman JP. Melatonin secretion and the incidence of type 2 diabetes. JAMA. 2013;309(13):1388-96.
  64. Prokopenko I, Langenberg C, Florez JC, Saxena R, Soranzo N, Thorleifsson G, et al. Variants in MTNR1B influence fasting glucose levels. Nat Genet. 2009;41(1):77-81.
  65. Scheer FA, Van Montfrans GA, van Someren EJ, Mairuhu G, Buijs RM. Daily nighttime melatonin reduces blood pressure in male patients with essential hypertension. Hypertension. 2004;43(2):192-7.
  66. Grossman E, Laudon M, Yalcin R, Zengil H, Peleg E, Sharabi Y, et al. Melatonin reduces night blood pressure in patients with nocturnal hypertension. Am J Med. 2006;119(10):898-902.
  67. Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016;61(3):253-78.
  68. Tan DX, Reiter RJ, Manchester LC, Yan MT, El-Sawi M, Sainz RM, et al. Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger. Curr Top Med Chem. 2002;2(2):181-97.
  69. Straif K, Baan R, Grosse Y, Secretan B, El Ghissassi F, Bouvard V, et al. Carcinogenicity of shift-work, painting, and fire-fighting. Lancet Oncol. 2007;8(12):1065-6.
  70. Blask DE, Dauchy RT, Sauer LA. Putting cancer to sleep at night: the neuroendocrine/circadian melatonin signal. Endocrine. 2005;27(2):179-88.
  71. Tan D, Reiter RJ, Chen LD, Poeggeler B, Manchester LC, Barlow-Walden LR. Both physiological and pharmacological levels of melatonin reduce DNA adduct formation induced by the carcinogen safrole. Carcinogenesis. 1994;15(2):215-8.
  72. Tuntapakul S, Kitkhuandee A, Kanpittaya J, Johns J, Johns NP. Pineal calcification is associated with pediatric primary brain tumor. Asia Pac J Clin Oncol. 2016;12(4):e405-e10.
  73. Blask DE, Dauchy RT, Sauer LA, Krause JA, Brainard GC. Light during darkness, melatonin suppression and cancer progression. Neuro Endocrinol Lett. 2002;23 Suppl 2:52-6.
  74. Hansen J. Light at night, shiftwork, and breast cancer risk. J Natl Cancer Inst. 2001;93(20):1513-5.
  75. Lincoln GA, Clarke IJ, Hut RA, Hazlerigg DG. Characterizing a mammalian circannual pacemaker. Science (New York, NY). 2006;314(5807):1941-4.
  76. Mocaer E, Delalleau B, Boyer PA, de Bodinat C. [Development of a new antidepressant : agomelatine]. Med Sci (Paris). 2005;21(10):888-93.
  77. Scheer FA, Zeitzer JM, Ayas NT, Brown R, Czeisler CA, Shea SA. Reduced sleep efficiency in cervical spinal cord injury; association with abolished night time melatonin secretion. Spinal Cord. 2006;44(2):78-81.
  78. Zeitzer JM, Ayas NT, Shea SA, Brown R, Czeisler CA. Absence of detectable melatonin and preservation of cortisol and thyrotropin rhythms in tetraplegia. J Clin Endocrinol Metab. 2000;85(6):2189-96.
  79. Deacon S, English J, Tate J, Arendt J. Atenolol facilitates light-induced phase shifts in humans. Neuroscience letters. 1998;242(1):53-6.
  80. Arendt J. Melatonin: Countering Chaotic Time Cues. Frontiers in endocrinology. 2019;10:391.
  81. Muller HL, Handwerker G, Wollny B, Faldum A, Sorensen N. Melatonin secretion and increased daytime sleepiness in childhood craniopharyngioma patients. J Clin Endocrinol Metab. 2002;87(8):3993-6.
  82. De Leersnyder H. Inverted rhythm of melatonin secretion in Smith-Magenis syndrome: from symptoms to treatment. Trends Endocrinol Metab. 2006;17(7):291-8.
  83. Tordjman S, Anderson GM, Pichard N, Charbuy H, Touitou Y. Nocturnal excretion of 6-sulphatoxymelatonin in children and adolescents with autistic disorder. Biological psychiatry. 2005;57(2):134-8.
  84. Melke J, Goubran Botros H, Chaste P, Betancur C, Nygren G, Anckarsater H, et al. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13(1):90-8.
  85. Maras A, Schroder CM, Malow BA, Findling RL, Breddy J, Nir T, et al. Long-Term Efficacy and Safety of Pediatric Prolonged-Release Melatonin for Insomnia in Children with Autism Spectrum Disorder. J Child Adolesc Psychopharmacol. 2018.
  86. Lehmann ED, Cockerell OC, Rudge P. Somnolence associated with melatonin deficiency after pinealectomy. Lancet. 1996;347(8997):323.
  87. Murata J, Sawamura Y, Ikeda J, Hashimoto S, Honma K. Twenty-four hour rhythm of melatonin in patients with a history of pineal and/or hypothalamo-neurohypophyseal germinoma. J Pineal Res. 1998;25(3):159-66.
  88. Slawik H, Stoffel M, Riedl L, Vesely Z, Behr M, Lehmberg J, et al. Prospective Study on Salivary Evening Melatonin and Sleep before and after Pinealectomy in Humans. J Biol Rhythms. 2016;31(1):82-93.
  89. Mendelson WB, Bergmann BM. Effects of pinealectomy on baseline sleep and response to sleep deprivation. Sleep. 2001;24(4):369-73.
  90. Fisher SP, Sugden D. Endogenous melatonin is not obligatory for the regulation of the rat sleep-wake cycle. Sleep. 2010;33(6):833-40.
  91. Posadzki PP, Bajpai R, Kyaw BM, Roberts NJ, Brzezinski A, Christopoulos GI, et al. Melatonin and health: an umbrella review of health outcomes and biological mechanisms of action. BMC Med. 2018;16(1):18.
  92. Sateia MJ. International classification of sleep disorders-third edition: highlights and modifications. Chest. 2014;146(5):1387-94.
  93. Carney CE, Buysse DJ, Ancoli-Israel S, Edinger JD, Krystal AD, Lichstein KL, et al. The consensus sleep diary: standardizing prospective sleep self-monitoring. Sleep. 2012;35(2):287-302.
  94. Lewy AJ, Sack RL. The dim light melatonin onset as a marker for circadian phase position. Chronobiol Int. 1989;6(1):93-102.
  95. Dijk DJ, Shanahan TL, Duffy JF, Ronda JM, Czeisler CA. Variation of electroencephalographic activity during non-rapid eye movement and rapid eye movement sleep with phase of circadian melatonin rhythm in humans. J Physiol. 1997;505 ( Pt 3):851-8.
  96. Auger RR, Burgess HJ, Emens JS, Deriy LV, Thomas SM, Sharkey KM. Clinical Practice Guideline for the Treatment of Intrinsic Circadian Rhythm Sleep-Wake Disorders: Advanced Sleep-Wake Phase Disorder (ASWPD), Delayed Sleep-Wake Phase Disorder (DSWPD), Non-24-Hour Sleep-Wake Rhythm Disorder (N24SWD), and Irregular Sleep-Wake Rhythm Disorder (ISWRD). An Update for 2015: An American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med. 2015;11(10):1199-236.
  97. Lockley SW, Arendt J, Skene DJ. Visual impairment and circadian rhythm disorders. Dialogues Clin Neurosci. 2007;9(3):301-14.
  98. Lewy AJ. Clinical applications of melatonin in circadian disorders. Dialogues Clin Neurosci. 2003;5(4):399-413.
  99. Smits MG, Nagtegaal EE, van der Heijden J, Coenen AM, Kerkhof GA. Melatonin for chronic sleep onset insomnia in children: a randomized placebo-controlled trial. J Child Neurol. 2001;16(2):86-92.
  100. van Geijlswijk IM, Korzilius HP, Smits MG. The use of exogenous melatonin in delayed sleep phase disorder: a meta-analysis. Sleep. 2010;33(12):1605-14.
  101. Campbell SS. Effects of timed bright-light exposure on shift-work adaptation in middle-aged subjects. Sleep. 1995;18(6):408-16.
  102. Nagtegaal JE, Kerkhof GA, Smits MG, Swart AC, Van Der Meer YG. Delayed sleep phase syndrome: A placebo-controlled cross-over study on the effects of melatonin administered five hours before the individual dim light melatonin onset. J Sleep Res. 1998;7(2):135-43.
  103. Uchiyama M, Lockley SW. Non-24-Hour Sleep-Wake Rhythm Disorder in Sighted and Blind Patients. Sleep Med Clin. 2015;10(4):495-516.
  104. Sack RL, Auckley D, Auger RR, Carskadon MA, Wright KP, Jr., Vitiello MV, et al. Circadian rhythm sleep disorders: part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. An American Academy of Sleep Medicine review. Sleep. 2007;30(11):1484-501.
  105. Arendt J. Managing jet lag: Some of the problems and possible new solutions. Sleep Med Rev. 2009;13(4):249-56.
  106. Herxheimer A, Petrie KJ. Melatonin for the prevention and treatment of jet lag. Cochrane Database Syst Rev. 2002(2):CD001520.
  107. Folkard S, Arendt J, Clark M. Can melatonin improve shift workers' tolerance of the night shift? Some preliminary findings. Chronobiol Int. 1993;10(5):315-20.
  108. Burgess HJ, Sharkey KM, Eastman CI. Bright light, dark and melatonin can promote circadian adaptation in night shift workers. Sleep Med Rev. 2002;6(5):407-20.
  109. Bordet R, Devos D, Brique S, Touitou Y, Guieu JD, Libersa C, et al. Study of circadian melatonin secretion pattern at different stages of Parkinson's disease. Clin Neuropharmacol. 2003;26(2):65-72.
  110. Xie Z, Chen F, Li WA, Geng X, Li C, Meng X, et al. A review of sleep disorders and melatonin. Neurol Res. 2017;39(6):559-65.
  111. Riemersma-van der Lek RF, Swaab DF, Twisk J, Hol EM, Hoogendijk WJ, Van Someren EJ. Effect of bright light and melatonin on cognitive and noncognitive function in elderly residents of group care facilities: a randomized controlled trial. JAMA. 2008;299(22):2642-55.
  112. Morgenthaler TI, Lee-Chiong T, Alessi C, Friedman L, Aurora RN, Boehlecke B, et al. Practice parameters for the clinical evaluation and treatment of circadian rhythm sleep disorders. An American Academy of Sleep Medicine report. Sleep. 2007;30(11):1445-59.
  113. Loiseau F, Le Bihan C, Hamon M, Thiebot MH. Antidepressant-like effects of agomelatine, melatonin and the NK1 receptor antagonist GR205171 in impulsive-related behaviour in rats. Psychopharmacology (Berl). 2005;182(1):24-32.
  114. Arendt J, Rajaratnam SM. Melatonin and its agonists: an update. Br J Psychiatry. 2008;193(4):267-9.
  115. Harpsoe NG, Andersen LP, Gogenur I, Rosenberg J. Clinical pharmacokinetics of melatonin: a systematic review. Eur J Clin Pharmacol. 2015;71(8):901-9.
  116. Weishaupt JH, Bartels C, Polking E, Dietrich J, Rohde G, Poeggeler B, et al. Reduced oxidative damage in ALS by high-dose enteral melatonin treatment. J Pineal Res. 2006;41(4):313-23.
  117. Zhdanova IV, Wurtman RJ, Morabito C, Piotrovska VR, Lynch HJ. Effects of low oral doses of melatonin, given 2-4 hours before habitual bedtime, on sleep in normal young humans. Sleep. 1996;19(5):423-31.
  118. Seabra ML, Bignotto M, Pinto LR, Jr., Tufik S. Randomized, double-blind clinical trial, controlled with placebo, of the toxicology of chronic melatonin treatment. J Pineal Res. 2000;29(4):193-200.

 

Use of the Historial Weight Trajectory to Guide an Obesity-Focused Patient Encounter

ABSTRACT

 

Obesity is a chronic, complex, and challenging disease to address by clinicians. Thus, it is crucial for practitioners to obtain a thorough weight history from patients to identify potential triggers that influence weight gain trajectories and their relationships to development of disease co-morbidity and mortality. Obtaining a weight history from a patient can be approached systematically, similar to key elements of a history of present illness, as we will discuss.  Furthermore, patient-drawn life-events graph or readily available electronic health records graphs can elucidate in visual context pertinent contributing factors to the etiology of obesity. Often, biological, social, behavioral, and psychological causes of weight gain can be elicited through the use of weight histories. In this chapter, we will also explore life-events graph more in detail as they can provide remarkable value to the overall assessment and plan of care (whether lifestyle intervention alone or in combination with pharmacological, surgical, or combination thereof) in the patient with obesity.

 

INTRODUCTION

 

Obesity adversely affects all organ systems in the human body and causes and/or exacerbates numerous medical disorders such as cardiovascular disease, diabetes, kidney disease, and some forms of cancer. Today, the average adult weight has increased (1), disproportionately skewing rightward (2) in the body mass index (BMI) distribution curve (Figure 1) with a higher percentage of the population meeting criteria for Class 1 obesity or greater (>30 kg/m2) and more disease severity (Class 2 obesity or higher;  BMI >35 kg/m2). In addition, the average waist circumference has increased across US adults since 1999 (3). Increases in abdominal girth (>35 inches for women;  >40 inches for men), commonly called central or abdominal obesity, is a surrogate for visceral adiposity, which increases risk for the metabolic syndrome, type 2 diabetes, inflammation, and cardiovascular disease (4).

Figure 1.  Changing shape of BMI distribution curve over time (2).

 

Obesity is defined as a pathologically elevated and defended body fat mass due to dysregulation of the pathways that determine energy balance. The complexity in these pathways, including biological, genetic, developmental, epigenetic, environmental, or behavioral factors, lead to substantial variability in the pathophysiological expression of both amount of unwanted weight gain experienced by an individual as well as the number and severity of co-morbid conditions (diabetes, hypertension, etc.) (5-9). 

 

In addition to the variability in phenotypic presentation of weight gain and fat distribution in individuals with obesity (10), weight loss responses to lifestyle, pharmacological, and surgical treatment are also heterogeneous. Although most patients elicit an average response to a distinct type of treatment, some patients will have an above average response to the intervention, while responses in others may be sub-optimal, or they may not respond at all. Thus, a thorough weight history in combination with a clinically applied and integrated understanding of this disease, its root causes, and etiology can help guide successful treatment.

 

THE OVERWEIGHT AND OBESITY-FOCUSED ENCOUNTER

 

History, Physical, and Laboratory Testing of the Patient who is Overweight or Has Obesity

 

The evaluation and diagnosis of a patient with obesity includes the taking of a standard medical history with special attention to identification of potentially weight-promoting medications and obesity-related complications (11), a medical examination that characterizes the amount and distribution of weight as well as possible signs of secondary causes of unwanted weight gain, and review of relevant clinical laboratory tests. In addition, the history of present illness includes generation of a chronological weight graph (ideally using the electronic health record (EHR)) that incorporates a review of life events, lifestyle patterns and preferences, and previous successful and unsuccessful interventions. 

 

The physical exam should note the distribution of weight (especially truncal and abdominal) and areas of conspicuous absence of fat characteristic of lipodystrophies (12), both of which herald increased cardiometabolic risk, as well as areas of disproportionate accumulation of excess fat, such as lipomas and lipedema (13); documentation of cardiac status looking specifically for evidence of heart failure; abdominal palpation for hepatomegaly; identification of inflammatory or degenerative joint issues that may limit activity; and skin/neurological examinations to look for evidence of hypercortisolism (wide striae, proximal muscle weakness), hypothyroidism, hirsutism/acne that might indicate polycystic ovarian syndrome, and acanthosis nigricans over extensor surfaces/neck/axilla. Laboratory evaluation at the initial visit should include a comprehensive metabolic panel, complete blood count, assessment of thyroid status, and cardiometabolic risk assessment including a lipid panel and A1c (Table 1). 

 

Table 1.  Key Elements of an Obesity-Focused Encounter

History of Present Illness (HPI)

 

Weight History and timing to life events, developmental milestones (including puberty, pregnancy, menopause), medication use, and injuries, surgeries, or illnesses.

Past Medical and Surgical History (PMH, PSH)

 

In addition to a general review, identification of obesity-associated comorbidities and procedures:  gastroesophageal reflux, hypertension, HFpEF, asthma, OSA, OA, type 2 diabetes, CAD and PVD, menstrual irregularities/infertility/PCOS, bariatric surgery.

Social History (SH)

Lifestyle, health practices, nutrition, physical activity, sleep, smoking and recreational drug use, stressors, occupation, marital status.

Family History

Parental obesity, cultural patterns, family eating patterns.

Medications

Weight-gain promoting medications.

Physical Exam

BMI, waist circumference

Distribution of body fat, lipodystrophy, lipedema, hypercortisolism, acanthosis.

Laboratory and Diagnostic Testing

Risk assessment:  comprehensive metabolic panel, complete blood count, 25-OH vitamin D, C-reactive protein, TSH, hemoglobin A1c, and lipid panel.  When indicated, screening for co-morbid conditions such as obstructive sleep apnea, non-alcoholic steatohepatitis, and PCOS.

Assessment and Plan

Based on risks, complications, comorbid conditions and barriers to care.

 

Identification of obesity-related co-morbidities during the patient encounter and lab testing may necessitate referral for further evaluation, such as non-invasive imaging or liver biopsy to establish non-alcoholic steatohepatitis, a sleep study to diagnose obstructive sleep apnea, or X-rays to assess osteoarthritis in weight-bearing joints. Patients reporting low-level chest discomfort, dyspnea on exertion, or orthopnea should be considered for referral to a cardiologist for the possibility of cardiac ischemia or heart failure with preserved ejection fraction (HFpEF), an increasingly recognized disorder of severe obesity. Findings of weight-related medical conditions during the initial visit may prompt increased motivational drive toward behavioral and lifestyle changes as well as acceptance of recommendations to use anti-obesity medications or for referral for bariatric surgery. 

 

CLINICAL IMPORTANCE OF A WEIGHT TRAJECTORY

 

During the weight-focused portion of the history of present illness, it is important to document changes in the health that led the patient to seek medical attention over time and establish a clear and chronological description of the sequential events, including weight gain or loss, leading up to the current visit (14-16) (Table 2).

 

Table 2.  Key Elements of an Obesity-Focused History of Present Illness (16)

Onset

When did your weight gain start.

What did you weigh in early, late childhood, puberty, by decade as adult?

Nadir and Maximum Weight

What were your lifetime lowest and highest weights (excluding pregnancy)?

How did you achieve your lowest weight?

Precipitating Factors

What events in your life coincided with unwanted weight gain (puberty, pregnancy, menopause, starting or stopping smoking, starting a new medication such as insulin or steroids)?

Quality of Life

What is hardest to do at your current weight?

When did you feel your best?

Weight Loss Efforts

What did you try that helped you lose weight?

What interventions were successful for you?

Setting

In what context were you successful at your previous efforts?  Why do you think those efforts worked?

Temporal Patterns

What is the nature of your weight loss and weight gain over time?

Do you ever weight cycle (yo-yo) or is it gradual or rapid over time?

 

Multiple studies have demonstrated that an early childhood upward weight trajectory can be predictive of future development of obesity, obesity-related comorbid conditions, disability, and mortality (17-21). A lifetime maximum BMI (compared to a single baseline BMI measurement) in the overweight or obesity categories, coupled with 16 or more years of weight history, is associated with an increased all-cause and cause-specific mortality including cardiovascular disease and coronary heart disease (22). 

 

As will be discussed below, temporal patterns of weight gain that raise concerns in a weight history might include: (a) an early adiposity rebound during infancy or early childhood years (23,24), (b) adolescent weight gain that correlates with progression to severe adult obesity and related medical conditions (25), and (c) excessive weight gain during pregnancy or menopause. Other temporal associations with weight gain often not appreciated by patients and providers include those that accompany smoking cessation (26), resolution of  hyperthyroidism (27), initiation of medications for depression, anxiety, and pain management (e.g., beta-blockers, amitriptyline, gabapentin, others) (11), and normal age-associated sarcopenia where  skeletal muscle mass gradually declines and visceral fat preferentially increases (28).

 

Early Growth, Childhood, and Puberty

 

The timing of excessive body weight gain during one’s life is a predictor of future disease severity. Early and rapid weight gain during youth is predictive of co-morbidities later in life and these patients often experience a more steeply inclined weight trajectory into later stages of adulthood (19). During the ages of 2-6 years, children normally have lower adiposity and are usually at their nadir weight before “rebounding” back up during normal growth (Figure 2, blue line). When adiposity rebound occurs at less than one year of age as visualized on the BMI for-age and gender appropriate growth charts, typically expressed in percentile for age and sex, syndromic, congenital, and monogenic causes of obesity should be considered. Such a very early clinical weight gain history should trigger consideration for genetic testing of genes in the canonical melanocortin pathways (LEPR, PCSK1, POMC, MC4R), which range in incidence from very rare to being implicated in up to 1-6% of early-onset severe obesity (23,29,30). Genetic screening for obesity genes can also be triggered by rapid weight gain from early infancy, development of severe obesity (> 97thBMI percentile) at early ages (especially before the age of 10), persistent food-seeking behaviors indicative of poor appetite control, parental consanguinity, and tall stature/increased growth velocity (31). A weight trajectory following an early adiposity rebound (> 1 year age, ≤ 6 years of age, Figure 2, red dotted line) is a strong risk factor for obesity in adolescence and beyond (24,32) and should be can be an indication for early family-oriented lifestyle interventions.

Figure 2. BMI Percentile Growth Chart.

 

Weight gain leading to obesity during the adolescent development period correlates highly with progression to severe adult obesity. Development of adolescent obesity can affect timing of puberty (early vs. delayed) when body composition and fat distribution are rapidly changing (33). During normal pubertal development and growth, males acquire greater fat-free and skeletal muscle mass, whereas females attain higher fat mass (34).    

Figure 3. Schematic depiction of abnormal BMI percentile growth curves for adolescents with severe obesity.  Greater than or equal to the 95th BMI percentile corresponds to cut-offs for pediatric obesity. Other important considerations that determine therapeutic criteria are if the adolescent’s trajectory falls at or above 120th% and 140th% the 95th BMI percentile for age/gender (see Table 3).

 

Most patients who experience unwanted, excess weight gain in childhood and adolescence develop obesity-related complications that, when severe, often require pharmacologic intervention and/or bariatric surgery. Early identification and treatment are recommended as for prevention (35).  Currently there are six FDA-approved anti-obesity medications available for chronic weight management (orlistat, phentermine/topiramate, liraglutide 3.0 mg, semaglutide 2.4 mg, and setmelanotide). Phentermine/topiramate and orlistat are approved for >12 years respectively, phentermine alone is approved for age >16 years, and setmelanotide can be used as early as 6 years of age  (35,36). Use of anti-obesity pharmacotherapy in adolescents with severe obesity (>95th BMI percentile plus the presence of obesity-related comorbidity or >120th of 95th BMI percentile) is now considered standard of care for pediatric obesity treatment (35).  Furthermore, vertical sleeve gastrectomy and Roux-en-Y metabolic and bariatric surgery procedures are available options for adolescents with severe obesity (>120th of 95th BMI percentile plus obesity-related comorbidity or BMI >140th of 95th BMI percentile) (37). 

 

Table 3. Available therapeutic options based on BMI percentile cut offs of weight trajectory in adolescent patients with severe obesity (38).

BMI percentile as per CDC growth chart

>95th BMI percentile

>120th of the 95thBMI percentile

>140th of the 95thBMI percentile

Intensive lifestyle intervention

Anti-obesity medication

With comorbidity

Adolescent bariatric metabolic surgery

 

With comorbidity

 

Pregnancy, Breast Feeding, and Menopausal Transition

 

Pregnancy and menopause can be a time when women’s weights and body composition may become permanently altered under the influence of dramatic shifts in sex steroid levels.  Excessive weight gain during pregnancy can result in epigenetic changes in the developing fetus leading to adult-onset chronic disease such as diabetes, cardiovascular disease, and obesity (39-45) .  Furthermore, maternal obesity and excessive gestational weight gain have been linked to maternal-fetal complications such as increased risk of C-sections, preeclampsia, shoulder dystocia, and macrosomia in the infant (46-48). Data from large population-based epidemiological studies have shown that roughly 50% to 60% of women after pregnancy will return to their pre-pregnancy weight, but the other 50% will retain extra weight, with a third of all pregnant women shifting a BMI category (normal to overweight or obesity) (49).

 

The post-partum period following delivery of the newborn infant is also a vulnerable time for weight retention. Moreover, the relationship between breastfeeding practices and postpartum weight changes is largely unclear due to the difficulties examining breastfeeding and weight management in observational research and confounding variables (50,51). Breastfeeding overall has other notable health benefits to the infant, including reducing atop and improving , cognitive development, bone health, and maternal-infant attachment (52). 

 

Weight gain during midlife is common, and about two-thirds of women ages 40 to 59 and nearly three-quarters of women older than 60 are overweight or have obesity (BMI greater than 25 kg/m2). On average, midlife women gain 1.5 pounds (0.7 kg) per year (53). Thus, it is not surprising that menopause is often depicted as a weight-gain trigger on a patient’s life-event graph, especially in older women who gain weight after a period of weight maintenance.  Towards midlife, women undergo redistribution of body composition with increase in total body fat and enhanced inclination toward central abdominal visceral adiposity (54).  Excess body weight during menopause is associated with elevated cardiovascular (55) and metabolic risk, including insulin resistance and Type 2 diabetes mellitus (56,57). Early or late-onset menopause (with final menstrual cycle age < 45 years or age > 55 years respectively) compared to age 46-55 years is associated with increased risk of Type 2 diabetes mellitus [HR 1.04, 95% CI 0.99, 1.09 and HR 1.08, 95% CI 1.01, 1.14, respectively] (58). Undergoing a hysterectomy or an oophorectomy increases diabetes risk further (RR 1.17, 95% CI 1.07-1.29) compared to peri-/post-menopausal women (59).

 

CASE LESSONS IN PATIENT-GENERATED WEIGHT GRAPHS

 

While in the clinic, having a patient generate their own drawing of weight graph accomplishes two-fold goals. First, it provides a template on which weight inflections in the patient’s life can be potentially identified with causative or contributory life events, medical conditions, and medications; and secondly, it provides a platform to guide the clinical discussion regarding appropriate goal setting and best approaches to help them achieve as close to a healthy weight range as possible. 

 

Impact of Medications

 

The patient in Figure 4 experienced steroid-induced weight gain, a very common iatrogenic cause of obesity.  Exploring reasons for why this patient was initiated on steroids and communication with other specialists regarding opportunities to switch to another non-steroid dependent medication, if available, might mitigate the weight gain and prove to be a successful weight management strategy. Similar discussions of alternative approaches may also be undertaken with other commonly prescribed medications that promote weight gain, including some birth control methods (e.g., Depo-Provera), histamines, beta-blockers, amitriptyline, gabapentin, pregabalin, sulfonylureas, thiazolidinediones, and insulin (60).

Figure 4. Life graph showing effects of repeated exposures to steroids on weight for chronic inflammatory arthritis.

 

Effects of Situational Life Changes That Impact Weight

 

In Figure 5, the patient’s weight was at its nadir during college years until graduation.  Subsequently, marriage and job change were identified as life factors timed to weight gain.  In addition, pregnancy and menopause were identified biological associations with upward weight trajectory over time. Psychological stressors appear to have further augmented weight gain over time. Resilience to major life stressors (marriage, divorce, loss of spouse, unemployment, death of a loved one, major illness or injury, moving/relocation) is, unfortunately, not as common as is thought and can precipitate psychosocial disorders such as anxiety, depression, and alcoholism  (61,62).

Figure 5.  Life graph showing effects of several situational life changes that impact weight.

 

The patient In Figure 6 had a stable weight prior to a physically traumatic incident that led to immobility and sedentary behaviors. Identification of this specific event contributing to upward weight trajectory in the patient helped tailor the treatment strategy toward physical therapy, rehabilitation, and a customized exercise prescription to mitigate the weight gain.

Figure 6.  Life graph showing effect of a traumatic incident (a motor vehicle accident in this case) on weight.

 

Identification of Response to Weight Loss Interventions

 

The patient in Figure 7 gained over 40 pounds during exposure to various antipsychotic medications for the treatment of bipolar disorder. The downward shift in the weight graph occurred after initiation of metformin 500 mg once daily (to mitigate antipsychotic medication induced weight gain (63)) and phentermine. If certain medications are critical and cannot be substituted with an alternative, weight neutral medication, as is often the case in patients requiring anti-psychotic medications, anti-obesity medications (64), or bariatric surgery can often reverse the weight gain.

Figure 7. Life graph showing the effects of antipsychotic medications on weight and effective therapeutic intervention with metformin and anti-obesity medications.

 

Obesity has a multifactorial etiology leading to wide variability in its presentation.  Understanding causation and association of weight gain promoting factors in a patient’s life can help elucidate appropriate treatment strategies. Furthermore, initial non-response to anti-obesity medication does not indicate that the medication is ineffective. Rather it may not be targeting the pathophysiological pathways involved in metabolic and body weight dysregulation in the individual patient and an alternative anti-obesity medication or combination should be trialed for synergistic or additive weight loss effects. In Figure 8, the first blue arrow shows the time of initial visit when the patient started to gain weight due to multifactorial etiology (strong family history of obesity, maladaptive stress related to work, unhealthy nutritional habits), followed by initiation of anti-obesity medicine therapy and intensive lifestyle intervention (phentermine; 2nd blue arrow). Subsequently, the patient developed side effects and phentermine monotherapy was discontinued (3rd blue arrow). Several other anti-obesity pharmacological options were trialed (4th blue arrow); however, the patient did not respond. Ultimately, the patient underwent bariatric surgery (Roux-en-Y gastric bypass (RYGB)) to achieve successful weight loss response. 

Figure 8. The life events graph depicts the response to treatment and strategies for further intervention.

 

Identification of Weight Regain after Successful Weight Loss:  Importance of Prompt Intervention and Identification of Lifetime Maximum Weight

Figure 9.  Life graph showing effect of weight loss program intervention over time with weight regain.

 

The life-events graph of a 55-year-old patient with a history of hypertension, obstructive sleep apnea, and severe obesity (BMI 41.8 kg/m2) in Figure 9 shows a 95 pounds over a 1.5-year period through self-monitoring using an electronic smart phone tracking application, 1800 kcal/day intake, and an increase in physical activity as tolerated. However, after 1.5 years of successful weight loss, despite continued intensive lifestyle changes, the patient experienced weight regain over the next 3 years. Weight regain after a period of caloric restriction is physiologic as long-term persistence of metabolic adaptation occurs and hunger and satiety signals resist weight loss (65). It important to initiate anti-obesity pharmacotherapy at this critical stage to help sustain the weight loss and prevent weight regain. In this patient, starting an anti-obesity medication after 1.5 years when weight gain had started to occur would be recommended. It is also important to identify the patient’s lifetime max as the starting point when gauging effectiveness of anti-obesity medications in this scenario. Simply using the pre-medication weight might lead to inappropriate stopping of this treatment if weight stabilization is achieved, since the absence of subsequent weight loss might be misinterpreted as medication “failure.” By determining the lifetime maximum weight, however, weight stabilization is a very successful ~20% to 30% total weight loss (50 to 100 lbs. weight loss compared to the lifetime maximum weight of 293 lbs., depending on when it was started).

Figure 10.  Life graph showing a patient status post gastric-bypass surgery with weight regain following situational life transitions (marriage, birth of children).

Figure 11.  Life graph showing weight loss in a patient after bariatric surgery followed by weight regain after high job-related stress and retirement. 

Figures 10 and 11 depict life-event graphs of two patients who underwent metabolic and bariatric surgery. Following successful weight loss, they both experienced weight regain.  Weight regain in the postoperative bariatric patient is often difficult to treat and is usually of multifactorial etiology (as examples, recurrence of obesity-related comorbidities, non-compliance and adherence to treatment recommendations and routine bariatric care, physiologic return of hunger signals, introduction of life stressors). Early recognition, evaluation and medical management, provided anatomical causes have been ruled out, with multidisciplinary team support are crucial to removing obstacles to weight maintenance and patient’s continued commitment to lifestyle improvements. Further, it presents an opportunity to intervene early and initiate anti-obesity pharmacotherapy, ideally at the expected weight loss nadir, usually 1-3 years after surgery, to help optimize weight loss response and prevent weight regain (66).    

Communicating with Patients About the Disease of Obesity

To effectively communicate obesity treatment recommendations to patients, medical professionals first need to translate obesity pathophysiology in language understood by patients, without bias or stigma. General questions, concerns, or sentiments often echoed by patients during their weight journey include:

 

  1. “Doc, are you calling me ‘fat?’”
  2. “I have tried every diet in the past. I lose weight only to regain it back.”
  3. “I know it’s my fault I am this weight.”
  4. “I have always been 300 pounds my entire life, no matter what.”
  5. “I don’t think I need surgery. I’ll just have to diet and exercise anyways.”
  6. “I don’t need medications to lose weight.”

 

Clinically, there are three important concepts to convey to patients about the disease state: 

 

  1. Obesity is a disease, like hypertension or diabetes.
  2. Because it is a disease, we can treat it as such, either starting with lifestyle and then moving on to medical and surgical treatments for obesity, or initiating lifestyle plus these treatments in patients with severe obesity.
  3. It is not the patient’s fault that they developed obesity.There are strong biological and genetic forces at play that determine body mass in addition to medical, behavioral, lifestyle, psychosocial, and environmental influences.

 

To clearly communicate with patients about the disease of obesity, a thorough understanding of energy metabolism is often necessary. Energy metabolism is complexly regulated in the arcuate nucleus of the hypothalamus to set a defended body weight (10,36,67). This body weight “set point” is oftentimes biologically and genetically determined with influences from the modern macro- and micro-environments. Chronic exposure to an obesogenic environment can increase body fat mass set point in susceptible individuals causing the body to defend a higher weight regardless of well-intended volitional efforts to lose weight. Thus, current treatment strategies include coupling intensive lifestyle therapy with anti-obesity medications and/or bariatric surgery, to allow significant and sustained reductions in the fat mass set point. Discontinuation of treatments that successfully lower the body mass set point will result in regain of body weight to the original, pathologically higher body mass set point. Therefore, patients need to understand that treatments for obesity are long-term, as is true for any medication prescribed for a chronic medical condition. Regain of weight after discontinuation of therapy is not evidence of failed treatment or lack of willpower; rather it is physiologically expected. 

 

Medical professionals can incorporate this scientific understanding in a manner that evades bias and stigma and transforms the patient’s queries into a constructive dialogue. In response to the questions or concerns posed by the patients above, medical professionals can reply in the following manner: 

 

  1. “Doc, are you calling me ‘fat’?” Response: “We only use the word ‘fat’ to describe mice and rodents, not humans. We are people. There are various parameters we check to make sure your body is healthy, not only from the outside, but also from the inside, similar to us checking your blood pressure and cholesterol. Your weight indicates that your body-mass-index is elevated. It means that it places you at high risk for, or already contributing to, other weight-related medical conditions. A high BMI is a result of a disease process inside your body, like diabetes. You have obesity, or adiposity-based chronic disease. The good news is that you have treatment options beyond just saying ‘eat less and exercise more’.” 
  2. “I have tried every diet in the past. I lose weight only to regain it back.”  Response: “Contrary to popular belief, we don’t consciously control our weight. It is regulated by a fat mass set point in our brains.  If your fat mass set point is abnormally high (again, not your fault), your body will continue to defend that weight. This abnormal physiology is why weight regain is common. There are treatments that more effectively lower that fat mass set point, so you do not have to struggle. That way, when you are making healthy food choices and are active, your body starts to respond. If you’d like, we can discuss those treatment options and how they may benefit you.” 
  3. “I know it’s my fault I am this weight.” Response:First of all, I want you to understand that it’s not your fault you developed obesity. You did not choose to ‘have’ obesity. If you were here today to discuss your diabetes diagnosis, it would be a completely different conversation, correct? Just like you did not have a choice in selecting your parents, you did not have a choice in the development of your obesity.  Again, contrary to popular belief, majority of our weight is not under our control.  The good news is that we can treat it.” 
  4. “I have always been 300 pounds my entire life, no matter what.” Response: “Have you wondered why that has been the reason?  We now know that there is a weight set point in our brain that determines what our brains think of as “normal.”. It is biologically and genetically determined with influences from the outside environment, medications, and other factors. In patients with obesity, this weight set point is abnormally high—your brain thinks that weight is now “normal.”  Thus, when you try to lose weight, you regain it back to this set point and, sometime, more. If improving your lifestyle isn’t working, we have treatments to lower this fat mass set point when it is high.  Anti-obesity medications and bariatric surgery are great tools to allow your brain to be happy with a lower weight set point so that you do not have to fight against your body.”
  5. “I don’t think I need surgery. I’ll just have to diet and exercise anyways.”  Response: “Let’s explore this further.  Can you help me understand more your reasons as to why you think you do not need surgery? What are your concerns and fears? What is your understanding about bariatric surgery? Let’s see if we can address some of those today.”
  6. “I don’t need medications to lose weight.” Response: “Can you help me understand your reasons for not considering medications? What is your understanding of obesity medications and how they work? If we try to manage your weight without medications, I would like to reassess your progress in 12 weeks.  If you are still struggling despite all your best efforts, please know that it is not your fault, and that your body is telling you that it needs something more intensive to treat the weight gain. You do have options still!”

 

CONCLUSIONS

 

An obesity-focused medical history is important for the care of the patient who is overweight or has obesity. In addition to identifying obesity-related complications by means of a thorough history, physical, and appropriate laboratory work up, using electronic health record weight graphs and identifying significant medical and life-events that influence body weight provide context and relevance for weight trajectories in the management of a patient with obesity.  For example, these events can help guide the clinical discussion in regard to potentially modifiable influences on a patient’s weight, including identifying causative medical conditions or culprit medications that promote weight gain, associating weight gain with known life-transitions that increase a patient’s risk for becoming overweight or obese (e.g., puberty, pregnancy, menopause), help patients to re-commit to improved lifestyle choices, assess an individual’s responsiveness to recommended therapies, and help with timing for when initiating anti-obesity, therapies such as weight loss medications, weight loss surgery, or both, when indicated.       

 

REFERENCES

 

  1. Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA. 2012;307(5):491-497.
  2. Penman AD, Johnson WD. The changing shape of the body mass index distribution curve in the population: implications for public health policy to reduce the prevalence of adult obesity. Prev Chronic Dis. 2006;3(3):A74.
  3. Fryar CD, Kruszon-Moran D, Gu Q, Ogden CL. Mean Body Weight, Height, Waist Circumference, and Body Mass Index Among Adults: United States, 1999-2000 Through 2015-2016. Natl Health Stat Report. 2018(122):1-16.
  4. Ritchie SA, Connell JM. The link between abdominal obesity, metabolic syndrome and cardiovascular disease. Nutr Metab Cardiovasc Dis. 2007;17(4):319-326.
  5. Bessler M, Daud A, DiGiorgi MF, Schrope BA, Inabnet WB, Davis DG. Frequency distribution of weight loss percentage after gastric bypass and adjustable gastric banding. Surg Obes Relat Dis. 2008;4(4):486-491.
  6. Gardner CD. Tailoring dietary approaches for weight loss. Int J Obes Suppl. 2012;2(Suppl 1):S11-S15.
  7. Smith SR, Prosser WA, Donahue DJ, Morgan ME, Anderson CM, Shanahan WR, Group APDS. Lorcaserin (APD356), a selective 5-HT(2C) agonist, reduces body weight in obese men and women. Obesity (Silver Spring). 2009;17(3):494-503.
  8. Hatoum IJ, Greenawalt DM, Cotsapas C, Reitman ML, Daly MJ, Kaplan LM. Heritability of the weight loss response to gastric bypass surgery. J Clin Endocrinol Metab. 2011;96(10):E1630-1633.
  9. Buford TW, Roberts MD, Church TS. Toward exercise as personalized medicine. Sports Med. 2013;43(3):157-165.
  10. Srivastava G, Apovian CM. Current pharmacotherapy for obesity. Nat Rev Endocrinol. 2018;14(1):12-24.
  11. Verhaegen AA, Van Gaal LF. Drugs That Affect Body Weight, Body Fat Distribution, and Metabolism. In: Feingold KR, Anawalt, B., Boyce, A., Chrousos, G., Dungan, K., Grossman, A., Hershman, J. M., Kaltsas, G., Koch, C., Kopp, P., Korbonits, M., McLachlan, R., Morley, J. E., New, M., Perreault, L., Purnell, J., Rebar, R., Singer, F., Trence, D. L., Vinik, A., Wilson, D. P., ed. Endotext [Internet]. South Dartmouth (MA) MDText.com, Inc.; 2000-; 2019 Feb 11.
  12. Akinci B, Sahinoz M, Oral E. Lipodystrophy Syndromes: Presentation and Treatment. In: Feingold KR, Anawalt, B., Boyce ,A., Chrousos, G., Dungan, K., Grossman, A., Hershman, J. M., Kaltsas, G., Koch, C., Kopp, P., Korbonits, M., McLachlan, R., Morley, J. E., New, M., Perreault, L., Purnell, J., Rebar, R., Singer, F., Trence, D. L., Vinik, A., Wilson, D., ed. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-. 2018 Apr 24.
  13. Herbst KL. Subcutaneous Adipose Tissue Diseases: Dercum Disease, Lipedema, Familial Multiple Lipomatosis, and Madelung Disease. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  14. Adler HM. The history of the present illness as treatment: who's listening, and why does it matter? J Am Board Fam Pract. 1997;10(1):28-35.
  15. Redelmeier DA, Schull MJ, Hux JE, Tu JV, Ferris LE. Problems for clinical judgement: 1. Eliciting an insightful history of present illness. CMAJ. 2001;164(5):647-651.
  16. Kushner RF, Batsis JA, Butsch WS, Davis N, Golden A, Halperin F, Kidambi S, Machineni S, Novick M, Port A, Rubino DM, Saunders KH, Shapiro Manning L, Soleymani T, Kahan S. Weight History in Clinical Practice: The State of the Science and Future Directions. Obesity (Silver Spring). 2020;28(1):9-17.
  17. Malhotra R, Ostbye T, Riley CM, Finkelstein EA. Young adult weight trajectories through midlife by body mass category. Obesity (Silver Spring). 2013;21(9):1923-1934.
  18. Stokes A, Ni Y, Preston SH. Prevalence and Trends in Lifetime Obesity in the U.S., 1988-2014. Am J Prev Med.2017;53(5):567-575.
  19. Zheng Y, Manson JE, Yuan C, Liang MH, Grodstein F, Stampfer MJ, Willett WC, Hu FB. Associations of Weight Gain From Early to Middle Adulthood With Major Health Outcomes Later in Life. JAMA. 2017;318(3):255-269.
  20. Zheng Y, Song M, Manson JE, Giovannucci EL, Hu FB. Group-Based Trajectory of Body Shape From Ages 5 to 55 Years and Cardiometabolic Disease Risk in 2 US Cohorts. Am J Epidemiol. 2017;186(11):1246-1255.
  21. Petrick JL, Kelly SP, Liao LM, Freedman ND, Graubard BI, Cook MB. Body weight trajectories and risk of oesophageal and gastric cardia adenocarcinomas: a pooled analysis of NIH-AARP and PLCO Studies. Br J Cancer. 2017;116(7):951-959.
  22. Yu E, Ley SH, Manson JE, Willett W, Satija A, Hu FB, Stokes A. Weight History and All-Cause and Cause-Specific Mortality in Three Prospective Cohort Studies. Ann Intern Med. 2017;166(9):613-620.
  23. Farooqi IS, O'Rahilly S. The Genetics of Obesity in Humans. In: Feingold KR, Anawalt, B., Boyce A3, Chrousos, G., Dungan, K., Grossman, A., Hershman, J. M., Kaltsas, G., Koch, C., Kopp, P., Korbonits, M., McLachlan, R., Morley, J. E., New, M., Perreault, L., Purnell, J., Rebar, R., Singer, F., Trence, D. L., Vinik, A., Wilson, D. P., ed. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-; 2017 Dec 23.
  24. Geserick M, Vogel M, Gausche R, Lipek T, Spielau U, Keller E, Pfaffle R, Kiess W, Korner A. Acceleration of BMI in Early Childhood and Risk of Sustained Obesity. N Engl J Med. 2018;379(14):1303-1312.
  25. Twig G, Yaniv G, Levine H, Leiba A, Goldberger N, Derazne E, Ben-Ami Shor D, Tzur D, Afek A, Shamiss A, Haklai Z, Kark JD. Body-Mass Index in 2.3 Million Adolescents and Cardiovascular Death in Adulthood. N Engl J Med. 2016;374(25):2430-2440.
  26. Bush T, Lovejoy JC, Deprey M, Carpenter KM. The effect of tobacco cessation on weight gain, obesity, and diabetes risk. Obesity (Silver Spring). 2016;24(9):1834-1841.
  27. Schneider DF, Nookala R, Jaraczewski TJ, Chen H, Solorzano CC, Sippel RS. Thyroidectomy as primary treatment optimizes body mass index in patients with hyperthyroidism. Ann Surg Oncol. 2014;21(7):2303-2309.
  28. Choi KM. Sarcopenia and sarcopenic obesity. Korean J Intern Med. 2016;31(6):1054-1060.
  29. Oswal A, Yeo GS. The leptin melanocortin pathway and the control of body weight: lessons from human and murine genetics. Obes Rev. 2007;8(4):293-306.
  30. Doulla M, McIntyre AD, Hegele RA, Gallego PH. A novel MC4R mutation associated with childhood-onset obesity: A case report. Paediatr Child Health. 2014;19(10):515-518.
  31. Malhotra S, Sivasubramanian R, Srivastava G. Evaluation and Mangement of Early Onset Genetic Obesity in Childhood. Journal of Pediatric Genetics. 2021.
  32. Rolland-Cachera MF, Peneau S. Growth trajectories associated with adult obesity. World Rev Nutr Diet.2013;106:127-134.
  33. Lee JM, Wasserman R, Kaciroti N, Gebremariam A, Steffes J, Dowshen S, Harris D, Serwint J, Abney D, Smitherman L, Reiter E, Herman-Giddens ME. Timing of Puberty in Overweight Versus Obese Boys. Pediatrics.2016;137(2):e20150164.
  34. Loomba-Albrecht LA, Styne DM. Effect of puberty on body composition. Curr Opin Endocrinol Diabetes Obes.2009;16(1):10-15.
  35. Srivastava G, Fox CK, Kelly AS, Jastreboff AM, Browne AF, Browne NT, Pratt JSA, Bolling C, Michalsky MP, Cook S, Lenders CM, Apovian CM. Clinical Considerations Regarding the Use of Obesity Pharmacotherapy in Adolescents with Obesity. Obesity (Silver Spring). 2019;27(2):190-204.
  36. Kumar RB, Srivastava G, Reid TJ, Aronne LJ. Understanding the pathophysiologic pathways that underlie obesity and options for treatment. Expert Rev Endocrinol Metab. 2021;16(6):321-338.
  37. Pratt JSA, Browne A, Browne NT, Bruzoni M, Cohen M, Desai A, Inge T, Linden BC, Mattar SG, Michalsky M, Podkameni D, Reichard KW, Stanford FC, Zeller MH, Zitsman J. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg Obes Relat Dis. 2018;14(7):882-901.
  38. Cardel MI, Jastreboff AM, Kelly AS. Treatment of Adolescent Obesity in 2020. JAMA. 2019;322(17):1707-1708.
  39. Stephenson J, Heslehurst N, Hall J, Schoenaker D, Hutchinson J, Cade JE, Poston L, Barrett G, Crozier SR, Barker M, Kumaran K, Yajnik CS, Baird J, Mishra GD. Before the beginning: nutrition and lifestyle in the preconception period and its importance for future health. Lancet. 2018;391(10132):1830-1841.
  40. Fleming TP, Watkins AJ, Velazquez MA, Mathers JC, Prentice AM, Stephenson J, Barker M, Saffery R, Yajnik CS, Eckert JJ, Hanson MA, Forrester T, Gluckman PD, Godfrey KM. Origins of lifetime health around the time of conception: causes and consequences. Lancet. 2018;391(10132):1842-1852.
  41. Fernandez-Twinn DS, Hjort L, Novakovic B, Ozanne SE, Saffery R. Intrauterine programming of obesity and type 2 diabetes. Diabetologia. 2019;62(10):1789-1801.
  42. Jansen MAC, Dalmeijer GW, Saldi SR, Grobbee DE, Baharuddin M, Uiterwaal CS, Idris NS. Pre-pregnancy parental BMI and offspring blood pressure in infancy. European journal of preventive cardiology.2019:2047487319858157.
  43. Heslehurst N, Vieira R, Akhter Z, Bailey H, Slack E, Ngongalah L, Pemu A, Rankin J. The association between maternal body mass index and child obesity: A systematic review and meta-analysis. PLoS medicine.2019;16(6):e1002817.
  44. Razaz N, Villamor E, Muraca GM, Bonamy AE, Cnattingius S. Maternal obesity and risk of cardiovascular diseases in offspring: a population-based cohort and sibling-controlled study. Lancet Diabetes Endocrinol.2020;8(7):572-581.
  45. Perak AM, Lancki N, Kuang A, Labarthe DR, Allen NB, Shah SH, Lowe LP, Grobman WA, Lawrence JM, Lloyd-Jones DM, Lowe WL, Jr., Scholtens DM, Group HF-USCR. Associations of Maternal Cardiovascular Health in Pregnancy With Offspring Cardiovascular Health in Early Adolescence. JAMA. 2021;325(7):658-668.
  46. Mission JF, Marshall NE, Caughey AB. Pregnancy risks associated with obesity. Obstet Gynecol Clin North Am.2015;42(2):335-353.
  47. Marshall NE, Guild C, Cheng YW, Caughey AB, Halloran DR. Maternal superobesity and perinatal outcomes. American journal of obstetrics and gynecology. 2012;206(5):417 e411-416.
  48. Getahun D, Ananth CV, Peltier MR, Salihu HM, Scorza WE. Changes in prepregnancy body mass index between the first and second pregnancies and risk of large-for-gestational-age birth. American journal of obstetrics and gynecology. 2007;196(6):530 e531-538.
  49. Wallace JM, Bhattacharya S, Horgan GW. Weight change across the start of three consecutive pregnancies and the risk of maternal morbidity and SGA birth at the second and third pregnancy. PloS one.2017;12(6):e0179589.
  50. Neville CE, McKinley MC, Holmes VA, Spence D, Woodside JV. The effectiveness of weight management interventions in breastfeeding women--a systematic review and critical evaluation. Birth. 2014;41(3):223-236.
  51. Neville CE, McKinley MC, Holmes VA, Spence D, Woodside JV. The relationship between breastfeeding and postpartum weight change--a systematic review and critical evaluation. Int J Obes (Lond). 2014;38(4):577-590.
  52. Lucas A. Scientific Evidence for Breastfeeding. Nestle Nutr Inst Workshop Ser. 2019;90:1-12.
  53. Kapoor E, Collazo-Clavell ML, Faubion SS. Weight Gain in Women at Midlife: A Concise Review of the Pathophysiology and Strategies for Management. Mayo Clin Proc. 2017;92(10):1552-1558.
  54. Kozakowski J, Gietka-Czernel M, Leszczynska D, Majos A. Obesity in menopause - our negligence or an unfortunate inevitability? Prz Menopauzalny. 2017;16(2):61-65.
  55. Ramezani Tehrani F, Behboudi-Gandevani S, Ghanbarian A, Azizi F. Effect of menopause on cardiovascular disease and its risk factors: a 9-year follow-up study. Climacteric. 2014;17(2):164-172.
  56. Davis SR, Castelo-Branco C, Chedraui P, Lumsden MA, Nappi RE, Shah D, Villaseca P, Writing Group of the International Menopause Society for World Menopause D. Understanding weight gain at menopause. Climacteric. 2012;15(5):419-429.
  57. Senechal M, Arguin H, Bouchard DR, Carpentier AC, Ardilouze JL, Dionne IJ, Brochu M. Weight gain since menopause and its associations with weight loss maintenance in obese postmenopausal women. Clin Interv Aging. 2011;6:221-225.
  58. LeBlanc ES, Kapphahn K, Hedlin H, Desai M, Parikh NI, Liu S, Parker DR, Anderson M, Aroda V, Sullivan S, Woods NF, Waring ME, Lewis CE, Stefanick M. Reproductive history and risk of type 2 diabetes mellitus in postmenopausal women: findings from the Women's Health Initiative. Menopause. 2017;24(1):64-72.
  59. Pandeya N, Huxley RR, Chung HF, Dobson AJ, Kuh D, Hardy R, Cade JE, Greenwood DC, Giles GG, Bruinsma F, Demakakos P, Simonsen MK, Adami HO, Weiderpass E, Mishra GD. Female reproductive history and risk of type 2 diabetes: A prospective analysis of 126 721 women. Diabetes Obes Metab. 2018;20(9):2103-2112.
  60. Verhaegen AA, Van Gaal LF. Drugs That Affect Body Weight, Body Fat Distribution, and Metabolism. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  61. Infurna FJ, Luthar SS. Resilience to Major Life Stressors Is Not as Common as Thought. Perspect Psychol Sci.2016;11(2):175-194.
  62. Just-Ostergaard E, Mortensen EL, Flensborg-Madsen T. Major life events and risk of alcohol use disorders: a prospective cohort study. Addiction. 2018;113(1):25-33.
  63. Hendrick V, Dasher R, Gitlin M, Parsi M. Minimizing weight gain for patients taking antipsychotic medications: The potential role for early use of metformin. Ann Clin Psychiatry. 2017;29(2):120-124.
  64. Apovian CM, Aronne LJ, Bessesen DH, McDonnell ME, Murad MH, Pagotto U, Ryan DH, Still CD, Endocrine S. Pharmacological management of obesity: an endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(2):342-362.
  65. Sumithran P, Prendergast LA, Delbridge E, Purcell K, Shulkes A, Kriketos A, Proietto J. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597-1604.
  66. Srivastava G, Buffington C. A Specialized Medical Management Program to Address Post-operative Weight Regain in Bariatric Patients. Obes Surg. 2018.
  67. Garvey WT. Is Obesity or Adiposity-Based Chronic Disease Curable: The Set Point Theory, the Environment, and Second-Generation Medications. Endocr Pract. 2022;28(2):214-222.

 

Thyrotropin-Secreting Pituitary Adenomas

ABSTRACT

 

Thyrotropin-secreting pituitary tumors (TSH-omas) are a rare cause of hyperthyroidism and account for less than 1% of all pituitary adenomas. It is, however, noteworthy that the number of reported cases increased over the last few years because of the routine use of ultrasensitive immunometric assays for measuring TSH levels. Contrary to previous RIAs, ultrasensitive TSH assays allow a clear distinction between patients with suppressed and those with non-suppressed circulating TSH concentrations, i.e., between patients with primary hyperthyroidism (Graves’ disease or toxic nodular goiter) and those with central hyperthyroidism (TSH-oma or pituitary resistance to thyroid hormone action, PRTH). Failure to recognize the presence of a TSH-oma may result in dramatic consequences, such as improper thyroid ablation that may cause the pituitary tumor volume to further expand. The presence of neurological signs and symptoms (visual defects, headache) or clinical features of concomitant hypersecretion of other pituitary hormones (acromegaly, galactorrhea/amenorrhea) strongly supports the diagnosis of TSH-oma. Nevertheless, the differential diagnosis between TSH-oma and PRTH may be difficult when the pituitary adenoma is very small, or in the case of confusing lesions, such as an empty sella or pituitary incidentalomas. First-line treatment of TSH-omas is pituitary adenomectomy followed by irradiation in the case of surgical failure. However, medical treatment with long-acting somatostatin analogues, such as octreotide and lanreotide, are effective in reducing TSH secretion in more than 90% of cases with consequent normalization of FT4 and FT3 levels and restoration of the euthyroid state.

 

INTRODUCTION

 

Thyrotropin (TSH)-secreting pituitary adenomas (TSH-omas) are a rare cause of hyperthyroidism. In this situation, TSH secretion is autonomous and refractory to the negative feedback of thyroid hormones (inappropriate TSH secretion) and TSH itself is responsible for the hyperstimulation of the thyroid gland and the consequent hypersecretion of T4 and T3 (1, 2). Therefore, this entity can be appropriately classified as a form of "central hyperthyroidism". The first case of TSH-oma was described in 1960 by measuring serum TSH levels with a bioassay (3). In 1970, Hamilton et al. (4) reported the first case of TSH-oma proved by measuring TSH by RIA.

 

Classically, TSH-omas were diagnosed at the stage of invasive macroadenoma and were considered difficult to cure. However, the advent of ultrasensitive immunometric assays, routinely performed as first line test of thyroid function, has greatly improved the diagnostic workup of hyperthyroid patients, allowing the recognition of the cases with unsuppressed TSH secretion. Therefore, TSH-omas are now more often diagnosed at an earlier stage, before they become a macroadenoma, and an increased number of patients with normal or elevated TSH levels in the presence of high free thyroid hormone concentrations have been recognized. Signs and symptoms of hyperthyroidism along with values of thyroid function tests similar to those found in TSH-oma may be recorded also among patients affected with resistance to thyroid hormones (5-7). This form of resistance to thyroid hormones is called pituitary resistance to thyroid hormones (PRTH), as the resistance to thyroid hormone action appears more severe at the pituitary than at the peripheral tissue level. The clinical importance of these rare entities is based on the diagnostic and therapeutic challenges they present.

 

Failure to recognize these different diseases may result in dramatic consequences, such as improper thyroid ablation in patients with central hyperthyroidism or unnecessary pituitary surgery in those with PRTH. Conversely, early diagnosis and the correct treatment of TSH-omas may prevent the occurrence of neurological and endocrinological complications, such as visual defects by compression of the optic chiasm, headache, and hypopituitarism, and should improve the rate of cure.

 

EPIDEMIOLOGY

 

TSH-producing adenoma is a rare disorder, accounting for about 0.5% to 2% of all pituitary adenomas, the prevalence in the general population being 1 to 2 cases per million (1, 2, 8-69). However, this figure is probably underestimated as confirmed by data obtained from The Swedish Pituitary Registry (70), demonstrating an increased incidence of TSH-omas over time (0.05 per 1 million per year in 1990-1994 to 0.26 per 1 million per year in 2005-2009), the national prevalence in 2010 being 2.8 per 1 million inhabitants. The increased number of reported cases principally results from the introduction of ultrasensitive TSH immunometric assays and from improved general practitioner and endocrinologist awareness. Based on the finding of measurable serum TSH levels in the presence of elevated FT4 and FT3 concentrations, many patients, previously thought to be affected with primary hyperthyroidism (Graves' disease on multinodular goiter), can nowadays be correctly diagnosed as patients with TSH-oma or, alternatively, with PRTH (1, 2, 5-7).

 

The presence of a TSH-oma has been reported at ages ranging from 8 to 84 years (2, 58, 58a,59), the mean age at diagnosis being 46 ± 6 years (59a). TSH-omas occur with equal frequency in men and women, in contrast with the female predominance seen in other thyroid disorders, a recent structured review of 535 adult cases of adult cases confirmed a female to male ratio of 1.07 (59a) Familial cases of TSH-oma have been reported as part of multiple endocrine neoplasia type 1 (MEN1) syndrome (10) and in familial isolated pituitary adenoma (FIPA) families with an AIP mutation (52).

 

PATHOLOGICAL ASPECTS

 

Immunostaining studies showed the presence of TSH beta subunit, either free or combined, in all adenomatous cells from every type of TSH-oma, with only few exceptions (1, 2, 8, 9, 71, 71a). Most TSH-secreting adenomas (72%) are secreting TSH alone, often accompanied by unbalanced hypersecretion of alpha-subunit of glycoprotein hormones (alpha-GSU) (Table 1). Interestingly, the existence of TSH-omas composed of two different cell types, one secreting alpha-GSU alone and another co-secreting alpha-GSU and the entire TSH molecules (mixed TSH/alpha-GSU adenomas), was documented by using double gold particle immunostaining (72). The presence of a mixed TSH/alpha-GSU adenoma is suggested by the finding of an extremely high alpha-GSU/TSH molar ratio and/or by the observation of dissociated TSH and alpha-GSU responses to TRH (1, 72). Classic mixed adenomas characterized by concomitant hypersecretion of other anterior pituitary hormones are found in about 30% of patients.

 

Table 1. Recorded Cases of TSH-Secreting Adenomas of Different Type

 

Number 

% of total

Total TSH-secreting adenomas (TSH-omas) 

598

---

Pure TSH-omas 

450

75.2

TSH-omas with associated hypersecretion of other pituitary hormones (mixed TSH-omas) 

148

24.8

Mixed TSH/GH-omas 

90

15.1

Mixed TSH/PRL-omas 

50 

  8.4

Mixed TSH/FSH/LH-omas 

  1.3

(Updated end May 2022 and personal unpublished observations)

 

Hypersecretion of GH and/or PRL, resulting in acromegaly and/or an amenorrhea/galactorrhea syndrome, are the most frequent associations (59a). This may be due to the fact that somatotroph and lactotroph cells share with thyrotropes common transcription factors, such as Prop-1 and Pit-1 (73). Rare is the occurrence of mixed TSH/gonadotropin adenoma, while no association with ACTH hypersecretion has been documented to date, probably due to the distant origin of corticotrope and thyrotrope lineages. Nonetheless, positive immunohistochemistry for one or more pituitary hormone does not necessarily correlate with its or their hypersecretion in vivo (9, 71, 74). Accordingly, clinically and biochemically silent thyrotropinomas have been reported (9, 75, 76). Moreover, true TSH-secreting tumors associated with Hashimoto’s thyroiditis and hypothyroidism have been documented (2, 39, 77, 78). Finally, an isolated pituitary gangliocytoma producing TSH and TRH has been recently reported (78a).

 

Microadenomas, with a diameter <1 cm, were recorded in less than 15% of the cases before 1996 (22), but their prevalence among all the TSH-omas is progressively increasing due to improved testing of thyroid function and awareness among endocrinologists and general practitioners. Consistently, in the series recently published by our Institution (79), up to 30% of TSH-omas were microadenomas. In this view, a recent structured review of 533 cases of adult TSH-omas confirmed that 23.1% of them were microadenomas the remaining 76.9% being macroadenoma with a mean diameter of 21.5 ± 7.9 mm (59a). Most TSH-omas had been diagnosed at the stage of macroadenomas and showed localized or diffuse invasiveness into the surrounding structures, especially into the dura mater (1, 8, 15, 22, 71). Extrasellar extension in the supra- and/or parasellar direction were present in the majority of cases. The occurrence of invasive macroadenomas is particularly high among patients with previous thyroid ablation by surgery or radioiodine (Figure 1) (2). This finding emphasizes the deleterious effects of incorrect diagnosis and treatment of these adenomas, and the relevant action on tumor growth exerted by the reduction of circulating thyroid hormone levels through an altered feedback mechanism. Such an aggressive transformation of the tumor resembles that occurring in Nelson's syndrome after adrenalectomy for Cushing's disease. Finally, some data suggest that somatic mutations of the thyroid hormone receptor beta may be responsible for the defect in negative regulation of TSH secretion in some TSH-omas (56, 80-82). In addition, alteration in iodothyronine deiodinase enzyme expression and function may contribute to the resistance of tumor cells to the feedback mechanism of elevated thyroid hormone levels (83). However, these data were not confirmed by another study on this topic (84).

Figure 1. Clinical manifestations in patients with TSH-secreting adenomas. Patients have been divided into two categories according to previous thyroid surgery. The presence of goiter is the rule, even in patients with partial thyroidectomy. Hyperthyroid features may be overshadowed by those of associated hypersecretion/deficiency of other pituitary hormones. Invasive tumors are seen in about half of the patients with previous thyroidectomy and in 1/4 of untreated patients (P<0.01 by Fisher's exact test). Intrasellar tumors show an opposite distribution pattern.

The consistency of TSH-omas is usually very fibrous and sometimes so hard that they deserve the name of "pituitary stone" (85). Increased basic fibroblast growth factor (bFGF) levels were found in blood from two patients with invasive mixed PRL/TSH-secreting adenomas characterized by marked fibrosis (86). The tumoral origin of bFGF was confirmed by the finding of specific transcript in the tissues removed at surgery, suggesting a possible autocrine role for this growth factor in tumor development.

 

By light microscopy and appropriate staining, adenoma cells usually have a chromophobe appearance. Cells are often organized in cords, and they frequently appear polymorphous and characterized by large nuclei and prominent nucleoli. Ultrastructurally, the well differentiated adenomatous thyrotropes resemble the normal ones, while the poorly differentiated adenomas are composed of elongated angular cells with irregular nuclei, poorly developed RER, long cytoplasmic processes and sparse small secretory granules (50-200 nm) usually lining up along the cell membrane (9, 71). Generally, no exocytosis is detectable. Cells with abnormal morphology or mitoses are occasionally found which may be mistaken for a pituitary malignancy or metastases from distant carcinomas (87). Nevertheless, the transformation of a TSH-oma into a pituitary carcinoma with multiple metastases has seldom been reported (35, 59a, 60, 88). Future malignant behavior might be predicted by the finding of a concomitant, spontaneous and marked decrease of both TSH and alpha-GSU serum concentrations that might indicate that the tumor is becoming less differentiated. Finally, in a mouse model of TSH-oma, the activation of phosphatidyl-inositol 3-kinase promoted aberrant pituitary growth that may induce transformation of the adenoma into a carcinoma (89).

 

MOLECULAR AND IN VITRO SECRETION STUDIES

 

The molecular mechanisms leading to the formation of TSH-omas are presently unknown, as is true for the large majority of pituitary adenomas. X-chromosomal inactivation analysis demonstrated that most pituitary adenomas, including the small number of TSH-omas investigated, derive from the clonal expansion of a single initially transformed cell (43). Accordingly, the general principles of tumorigenesis, which assume the presence of a transforming event providing gain of proliferative function followed by secondary mutations or alterations favoring tumor progression, presumably also apply to TSH-omas.

 

A large number of candidate genes, including common proto-oncogenes and tumor suppressor genes as well as pituitary specific genes, have been screened for mutations able to confer growth advantage to thyrotrope cells. In analogy with the other pituitary adenomas, no mutations in oncogenes commonly activated in human cancer, particularly RAS, have been reported in TSH-omas. In contrast, with GH-secreting adenomas in which the oncogene gsp (GNAS mutation) is frequently present, none of the TSH-oma screened has been shown to express activating mutations of genes encoding for G protein subunits, such as alpha s, alpha q, alpha 11 or alpha i2 (90). Similarly, no mutations in the TRH receptor or dopamine D2 receptor genes (90-91) have been reported in 9 and 3 TSH-omas, respectively, while these tumors were not screened for alterations in protein kinase C, previously identified in some invasive tumors. In consideration of the crucial role that the transcription factor Pit-1 exerts on cell differentiation and PRL, GH and TSH gene expression, the Pit-1 gene has been screened for mutations in 14 TSH-omas and found to be wild-type (2). By contrast, as occurs in GH-omas, Pit-1 was demonstrated to also be overexpressed in TSH-omas, although the proliferative potential of these findings remains to be elucidated (2, 71, 73).

 

In addition to activating mutations or overexpression of protooncogenes, tumors may originate from the loss of genes with antiproliferative action. As far as the loss of tumor suppressor genes is concerned, no loss of p53 was found in one TSH-oma studied, while the loss of retinoblastoma gene (Rb), which is unaltered in other pituitary adenomas, was not investigated in TSH-omas. Another candidate gene is MEN1 coding for menin. In fact, 3-30% of sporadic pituitary adenomas show loss of heterozygosity (LOH) on 11q13, where MEN1 is located, and LOH on this chromosome seems to be associated with the transition from the non-invasive to the invasive phenotype. A screening study carried out on 13 TSH-omas using polymorphic markers on 11q13 showed LOH in 3, but none of them showed a MEN1mutation after sequence analysis (92). Interestingly, hyperthyroidism due to TSH-omas has been reported in five cases within a familial setting of multiple endocrine neoplasia type 1 syndrome (1, 2, 10). In addition, LOH and in particular polymorphisms at the somatostatin receptor type 5 gene locus, seem to be associated with an aggressive phenotype and resistance to somatostatin analogue treatment (93). Moreover, germline mutations in the aryl hydrocarbon receptor interacting protein (AIP) are known to be involved in sporadic pituitary tumorigenesis, but mutations were found in two patients with TSH-omas (52, 94). Finally, a recent study based on whole-exome sequencing identified several candidate somatic mutations and change in copy numbers in 12 sporadic TSH-omas, but with low number per tumor and without recurrence of mutations (95). A recent publication by Villa and others analyzing a series of secreting and non-secreting pituitary adenomas (including 6 TSH-omas) demonstrated a higher frequency of chromosomal alterations in TSH-omas, these alterations not being related to aggressiveness (95a). The same authors also demonstrated that POU1F1/PIT1-lineage tumors (including TSH-omas) were characterized by a global hypomethylation possibly inducing chromosomal alteration through the activation of transposable elements. Finally, transcriptosome analysis demonstrated that thyrotrope tumors cluster with sparsely granulated somatotroph adenomas and plurihormonal PIT1-positive adenomas, a group characterized by a higher high interferon-α and -γ gene expression (95a).

 

The extreme refractoriness of neoplastic thyrotropes to the inhibitory action of thyroid hormones indicates mutant forms of thyroid hormone receptors (TR) as other potential candidate oncogenes. Absence of TR alpha1, TR alpha2 and TR beta1 expression was reported in two TSH-omas (82, 96), but aberrant alternative splicing of thyroid hormone receptor beta2 (THRB) mRNA encoding TR beta variant lacking T3 binding activity and other THRB mutations were shown as a mechanism for impaired T3-dependent negative regulation of both TSH and alpha-GSU in tumoral tissue (80, 81). Moreover, an aberrant expression of a novel thyroid hormone receptor β isoform (TRβ4) may partly contribute to the inappropriate secretion of TSH in TSH-omas (56). Several patients with THRB mutation and an PRTH phenotype have been described to bear pituitary lesions at imaging of the sella region, raising diagnostic and therapeutic dilemmas (30, 97-99). The results of dynamic testing of TSH secretion were consistent with PRTH, rather than TSH-omas, indicating that these lesions are likely to be pituitary incidentalomas, whose prevalence in not selected autopsy series reaches 20%.

 

Pharmacological manipulations in short-term cultures of TSH-omas indicate that these tumors express a large number of functioning receptors. Although in vivo TSH response to TRH is usually absent, several in vitro studies showed either the presence or the absence of TSH response, indicating that the majority of tumors possess TRH receptors (2). Similarly, somatostatin binding experiments indicate that almost all TSH-omas express a variable number of somatostatin receptors, the highest somatostatin-binding site densities being found in mixed GH/TSH adenomas (57, 100, 101). Since somatostatin analogues are highly effective in reducing TSH secretion by neoplastic thyrotropes (12, 13, 102, 103), the inhibitory pathway mediated by somatostatin receptors appears to be largely intact in such adenomas. Consistently, there is a good correlation between somatostatin binding capacity and maximal biological response, as quantified by inhibition of TSH secretion and in vivo restoration of euthyroid state (57, 102-104). The presence of dopamine receptors in TSH-omas was the rationale for therapeutic trials with dopaminergic agonists, such as bromocriptine (57, 105, 106). Several studies have shown a large heterogeneity of TSH responses to dopaminergic agents, either in primary cultures or in vivo (1, 41, 107, 108). The effects of these two inhibitory agents should be nowadays re-evaluated in light of the demonstration of the possible heterodimerization of somatostatin receptor subtype 5 (sst5) and dopamine D2 receptor (109).

 

CLINICAL FINDINGS

 

Patients with TSH-omas present with signs and symptoms of hyperthyroidism that are frequently associated with those related to the pressure effects of the pituitary adenomas, causing loss of vision, visual field defects, headache, and/or loss of anterior pituitary function (Figure 1) (22, 49, 63, 110). TSH-omas may occur at any age and, in contrast with the common thyroid disorders, there is no preferential incidence in females (2, 8, 22, 59, 59a). Due to the long history of thyroid dysfunction, many patients had been mistakenly diagnosed as having primary hyperthyroidism (Graves' disease or multinodular goiter), and about one third had inappropriate thyroid ablation by thyroidectomy and/or radioiodine. True coexistence of Graves’ disease and TSH-oma has been reported in 14 cases (54, 111, 111a, 111b). The majority of these cases were females (aged between 25 and 53 years old) and the dual diagnosis was confirmed within 3 years from the original diagnosis in all cases. When Graves’ disease is diagnosed initially, it has been speculated that antithyroid medications may promote the growth of a TSH-oma via the positive feedback system (111c).

 

Clinical features of hyperthyroidism are present in up to 75% of patients (59a), sometimes milder than expected given the level of thyroid hormones, probably due to their longstanding duration (112). Interestingly, only two cases of TSH-oma complicated with a thyroid storm peri- or post-operatively have been published (112a, 112b). Consistently, several untreated patients with TSH-oma were described as clinically euthyroid (59, 113). Moreover, hyperthyroid features can be overshadowed by those of acromegaly in patients with mixed TSH/GH adenomas (59a, 79, 114-118), thus emphasizing the importance of the systematic measurement of TSH and FT4 in patients with pituitary tumors. Acromegaly itself can often be associated with multinodular goiter, presenting a further possible differential diagnostic scenario.

 

Cardiotoxicosis with atrial fibrillation, cardiac failure, massive pleural and pericardial effusions have been reported in sporadic cases (119-123). Thyrotoxic heart failure and atrial fibrillation were found to be present in 11.1% of cases (59a). Typical episodes of periodic paralysis have also been described in two patients (20, 124). A high prevalence of radiological vertebral fracture has been recently documented in a series of patients with TSH-omas (125) thus confirming the deleterious effects of thyrotoxicosis on bone health.

 

The presence of a goiter is the rule, even in the patients with previous partial thyroidectomy, since thyroid residue may regrow as a consequence of TSH hyperstimulation. The occurrence of uni- or multinodular goiter is frequent (about 72% of reported cases), but progression towards functional autonomy seems to be rare (126, 127). The monitoring of the thyroid nodule(s) and the performance of fine needle aspiration biopsy are indicated in TSH-omas since differentiated thyroid carcinomas were documented in several patients (1, 11, 55, 59a, 128-131). A recent publication evaluating sixty-two patients who underwent surgery for TSH-oma demonstrated an estimated incidence of thyroid carcinoma of 4.8%, thus suggesting a possible role of TSH hypersecretion in the development of thyroid tumors (130). The prevalence of circulating antithyroid autoantibodies (anti-thyroglobulin: Tg-Ab, and anti-thyroid peroxidase: TPO-Ab) is similar to that found in the general population, but some patients developed Graves' disease after pituitary surgery and a few others presented bilateral exophthalmos due to autoimmune thyroiditis (2, 55, 132), while unilateral exophthalmos due to orbital invasion by the pituitary tumor has also been reported (3, 133).

 

Dysfunction of the gonadal axis is not rare, with menstrual disorders present in one third of the reported cases, mainly in the mixed TSH/PRL adenomas. In this respect, a recent report described a case of a 37-year-old woman who had experienced galactorrhea and menstrual disorder and undergone infertility treatment in 1 year before TSH-oma was identified (133a). Central hypogonadism, delayed puberty, and decreased libido were also found in a number of males with TSH-omas and/or mixed TSH/FSH adenomas (1, 107, 134, 135).

 

Because of suprasellar extension or invasiveness, signs and symptoms of an expanding tumor mass are predominant in many patients. Partial or total hypopituitarism was seen in about 25% of cases, headache reported in 20-25% of patients, and visual field defects are present in about 50% of patients (Figure 1).

 

BIOCHEMICAL FINDINGS

 

High concentrations of circulating free thyroid hormones in the presence of detectable TSH levels characterize the hyperthyroidism secondary to TSH-secreting pituitary adenomas. In a review of 533 cases of TSH-omas it has been shown that the median TSH at diagnosis was 6.75 (4.02–11.90) mU/L in the case series and 5.16 (3.20–7.43) mU/L in the case reports whereas FT4 averaged 35.7 ± 8.5 and 41.5 ± 15.3 pmol/L, respectively (59a). Interestingly, normal levels of total T4 were recorded in several patients with TSH-omas despite the presence of clinical signs and symptoms of hyperthyroidism. This observation indicates that the measurement of circulating free thyroid hormones (FT4 and FT3) is mandatory. In fact, these measurements show the highest sensitivity for the correct diagnosis of central hyperthyroidism and prevent misclassification in the case of excess of circulating levels of thyroxine-binding globulin (26). Many different physiological or clinical conditions, such as pregnancy or PRTH, may present with hyperthyroxinemia and detectable serum TSH levels, and should be distinguished from TSH-omas. Most of these conditions may be recognized on the basis of either a patient's clinical history or by measuring the concentrations of FT4 and FT3 with direct "two-step" methods, i.e., the methods able to avoid possible interference due to the contact between serum factors and tracer at the time of the assay (e.g., equilibrium dialysis+RIA, adsorption chromatography+RIA, and back-titration) (136, 137). In fact, some factors may interfere with the measurement of either thyroid hormones or TSH. The presence of anti-iodothyronine autoantibodies (anti-T4 and/or anti-T3) or abnormal albumin/transthyretin forms, such as those circulating in familial dysalbuminemic hyperthyroxinemia, may cause FT4 and/or FT3 to be overestimated, particularly when "one-step" analog methods are employed (137, 138). The more common factors interfering in TSH measurement and giving spuriously high levels of TSH are the circulating heterophilic antibodies, i.e., antibodies directed against mouse gamma-globulins (36, 139).

 

About 30% of TSH-oma patients with an intact thyroid gland showed TSH levels within the normal range (2). In this respect, it is worth noting that TSH with “reflex FT4 strategy” (i.e., measurement of FT4 test only in the presence of an abnormal TSH result) fails to recognize both central hypo and hyperthyroidism thus leading to TSH deficiency or TSH-oma misdiagnosis (140, 141). The diurnal rhythm is preserved in TSH-omas and TSH secretion shares many characteristics (cross-approximate entropy, cross-correlation and cosinor regression) of other pituitary hormone-secreting adenomas (142). Interestingly, a case of TSH-oma with cyclic fluctuations in serum TSH levels has recently been reported (143).

 

Furthermore, despite the TSH-dependent origin of hyperthyroidism, there is no direct correlation between free thyroid hormone and immunoreactive TSH levels (Figure 2). An increased biological activity of secreted TSH molecules likely accounts for the finding of normal TSH in the presence of high levels of FT4 and FT3 (114). TSH molecules secreted by pituitary tumors are heterogeneous and may have either a normal, reduced, or increased ratio between their biological and immunological activities, probably due to modification of glycosylation processes secondary to alterations of the post-translational processing of the hormone within the tumor cell (144, 145). Interestingly, TSH levels in patients previously treated with thyroid ablation were 6-fold higher than in untreated patients, though free thyroid hormone levels were still in the hyperthyroid range (115). Moreover, tumoral thyrotropes may undergo more active cellular proliferation in response to even small reductions in circulating thyroid hormone levels, as documented by the higher number of invasive macroadenomas found in previously treated patients (Figure 1), beyond the increase of TSH secretion.

Figure 2. Absent correlation between immunoreactive concentrations of circulating TSH and FT3 in 14 patients with TSH-secreting adenomas. Dotted lines indicate the upper limits of normal ranges for both parameters.

Independently of previous thyroid ablation, circulating free alpha-GSU levels and alpha-GSU/TSH molar ratio were clearly elevated in the majority of patients with TSH-oma, either due to unbalanced secretion of the subunit or to the presence of a mixed TSH/alpha-GSU adenoma (72). The calculation of the alpha-GSU/TSH molar ratio increases the diagnostic sensitivity of hormone measurement, and an alpha-GSU/TSH molar ratio above 1.0 was associated with the presence of a TSH-secreting pituitary adenoma (2, 8). However, data from our group show that the individual values must be compared with those of control groups matched for TSH and gonadotropin levels before drawing any diagnostic conclusions. Controls with normal levels of TSH and gonadotropins may have alpha-GSU/TSH molar ratios as high as 5.7, and values as high as 29.1 can be found in euthyroid postmenopausal women (146). Indeed, hypersecretion of alpha-GSU is not unique to TSH-omas, being present in the majority of true gonadotropinomas, in a subset of non-functioning pituitary adenomas, and in a number of GH- or PRL-secreting tumors. Moreover, high alpha-GSU levels may be observed in conditions other than pituitary adenomas, such as in patients with inflammatory bowel disease (e.g., ulcerative colitis, Crohn disease) or with other neuroendocrine tumors (e.g., carcinoids) (146).

 

PARAMETERS OF THYROID HORMONE ACTION

 

Patients with central hyperthyroidism may present with mild signs and symptoms of thyroid hormone overproduction. Therefore, the measurements of several parameters of peripheral thyroid hormone action have been proposed to quantify the degree of tissue hyperthyroidism (2, 5-8, 98). Some of them are measured in vivo (basal metabolic rate, cardiac systolic time intervals, "Achilles" reflex time) and others in vitro (sex hormone-binding globulin: SHBG, cholesterol, angiotensin converting enzyme, soluble interleukin-2 receptor, osteocalcin, carboxyterminal cross-linked telopeptide of type I collagen (ICTP), etc.). Liver (SHBG) and bone parameters (ICTP) have been successfully used to differentiate hyperthyroid patients with TSH-omas from those with pituitary RTH (Figure 3). In fact, as seen in the common forms of hyperthyroidism, patients with TSH-omas have high SHBG and ICTP levels, while they are in the normal range in patients with hyperthyroidism due to PRTH (147, 148).

 

Figure 3. Values of sex hormone-binding globulin (SHBG) and carboxyterminal cross-linked telopeptide of type 1 collagen (ICTP) in patients with PRTH or TSH-omas. Shaded areas represent the normal ranges either in premenopausal women or in postmenopausal women and men. The combined measurement of parameters from different tissues may be useful for the differential diagnosis and by-pass possible interference by different factors (age, liver or bone diseases, combined alteration of pituitary functions, treatments, etc.). Tx TSH-omas, treated TSH-omas.

DYNAMIC TESTING

 

Although both stimulatory and inhibitory tests had been proposed for the diagnosis of TSH-omas, none of them is of clear-cut diagnostic value. Classically, T3 suppression test has been used to assess the presence of a TSH-oma. A complete inhibition of TSH secretion after T3 suppression test (80-100 mcg/day for 8-10 days) has never been recorded in patients with TSH-oma. T3 suppression test can be combined with TRH testing, where normal subjects and TSH-oma patients do not show elevation of TSH in response to TRH, while PRTH patients show a brisk elevation (1). In patients with previous thyroid ablation, T3 suppression seems to be the most sensitive and specific test in assessing the presence of a TSH-oma (2, 8, 98). However, this test is strictly contraindicated in elderly patients or in those with coronary heart disease.

 

TRH testing has been widely used to investigate the presence of a TSH-oma. In the vast majority of patients, TSH and alpha-GSU levels do not increase after TRH injection. In patients with hyperthyroidism, discrepancies between TSH and alpha-GSU responses to TRH (i.e., higher alpha-GSU than TSH response) are pathognomonic of TSH-omas co-secreting other pituitary hormones. Such a discrepancy is also found in an opposite clinical condition, i.e., the congenital hypothyroidism due to a TSH beta gene mutation (149).

 

The majority of TSH-omas maintain sensitivity to native somatostatin and its analogues. Indeed, administration of native neuropeptide or its analogues (octreotide or lanreotide) induces a reduction of TSH levels in the majority of cases, and these tests may be predictive of the efficacy of long-term treatment (30, 89, 102, 104). Recently, it has been confirmed that a positive somatostatin test result is suggestive for a TSH-oma even before positive findings become apparent on pituitary imaging (104a).

 

As suggested by ETA 2013 guidelines we recommend the use of both T3 suppression and TRH tests whenever possible, because the combination of their results increases the specificity and sensitivity of the diagnostic work-up (158).

 

IMAGING STUDIES AND LOCALIZATION OF THE TUMOR

 

As for other tumors of the region of the sella turcica, nuclear magnetic resonance imaging (MRI) is nowadays the preferred tool for the visualization of a TSH-oma. High-resolution computed tomography (CT) is the alternative investigation in the case of contraindications, such as patients with pacemakers. Most TSH-omas have been diagnosed in the past at the stage of macroadenomas, and various degrees of suprasellar extension or sphenoidal sinus invasion are seen in two thirds of cases.

 

Microadenomas are now reported with increasing frequency, accounting for about 20-30% of all recorded cases in both clinical and surgical series. Pituitary scintigraphy with radiolabeled octreotide (Octreoscan) has been shown to successfully localize TSH-omas that express somatostatin receptors (8, 150, 151). However, the specificity of the Octreoscan is low, since positive scans can be seen in the case of a pituitary mass of different types, either secreting or non-secreting.

 

Finally, ectopic localization of a TSH-oma has been reported by different groups who found a nasopharyngeal mass in few patients with clinical and biochemical features of central hyperthyroidism (21, 32, 152-157a). Histological and immunohistochemical studies of both specimens collected during the operation showed unequivocally that the tumor was a TSH-oma, and the resection of the mass restored TSH and alpha-GSU levels to normal. Interestingly, in these cases either Octreoscan or Gallium 68 DOTATATE Positron Emission Tomography/Computed Tomography might be helpful in identifying an ectopic lesion (157a).

 

DIFFERENTIAL DIAGNOSIS

 

If FT4 and FT3 concentrations are elevated in the presence of measurable TSH levels, it is important to exclude methodological interference due to the presence of circulating autoantibodies (e.g., against T3 and T4) or heterophilic antibodies (e.g., for TSH). A recent publication by Campi and others indicated assay interference as the main source of error due to the widespread use of high-throughput platforms based on one-step assays that increased the frequency of assay artifact, due to interference from biotin, circulating heterophilic antibodies, or abnormal binding proteins (157b). In this respect, in an asymptomatic patient with elevated FT4/FT3 and detectable TSH levels, the so called Familial dysalbuminemic hyperthyroxinemia should be taken into account. FSH is a familial autosomal dominant condition caused by an abnormal albumin molecule with an increased affinity for serum thyroxine thus leading to a false increase of FT4 and FT3 while TSH levels are normal (138, 157c). In a patient with signs and symptoms of hyperthyroidism, the confirmed presence of elevated FT4/FT3 and detectable TSH levels rules out Graves' disease or other forms of primary hyperthyroidism. In patients on levothyroxine replacement therapy, the finding of measurable TSH in the presence of high FT4/FT3 levels may be due to poor compliance or to an incorrect high L-T4 dosage, probably administered before blood sampling.

 

When the existence of central hyperthyroidism is confirmed, several diagnostic steps have to be carried out to differentiate a TSH-oma from PRTH (Table 2) (1, 5-8, 30, 97, 98, 158). The presence of neurological signs and symptoms (visual defects, headache) of an expanding intra-cranial mass or clinical feature of concomitant hypersecretion of other pituitary hormones (acromegaly, galactorrhea/amenorrhea) points to the presence of a TSH-oma. The presence of alterations of the pituitary content on MRI or CT scanning strongly supports the diagnosis of a TSH-oma. Nevertheless, the differential diagnosis may be difficult when the pituitary adenoma is very small, or in the case of confusing lesions, such as an empty sella. Moreover, the possibility of pituitary incidentalomas should always be considered, due to their high prevalence. In our series, about 20% of PRTH patients have a pituitary lesion on MRI.

 

No significant differences in age, sex, previous thyroid ablation, TSH levels or free thyroid hormone concentrations were seen between patients with TSH-oma and those with PRTH. However, in contrast to PRTH patients, familial cases of TSH-oma have never been documented. Serum TSH levels within the normal range are more frequently found in PRTH, while elevated alpha-GSU concentrations and/or a high alpha-GSU/TSH molar ratio are typically present in patients with TSH-omas. Moreover, TSH unresponsiveness to TRH stimulation and/or to T3 suppression tests favors the presence of a TSH-oma. We have shown that chronic administration of long-acting somatostatin analogues in patients with central hyperthyroidism caused a marked decrease of FT3 and FT4 levels in all patients but one with TSH-oma, while patients with PRTH did not respond at all (30). Thus, administration of long-acting somatostatin analogues for at least 2 months can be useful in the differential diagnosis in problematic cases of central hyperthyroidism (30, 157b) (Table 2).

 

Indexes of thyroid hormone action at the tissue level (such as SHBG or ICTP levels) are in the hyperthyroid range in most patients with TSH-oma, while they are generally normal/low in PRTH (Figure 3). Exceptions are the findings of normal SHBG levels in patients with mixed GH/TSH adenoma, due to the inhibitory action of GH on SHBG synthesis and secretion, and of high SHBG in PRTH patients treated with estrogens or showing profound hypogonadism. Genetic analysis of the TR beta gene may be useful in the differential diagnosis, as TR beta mutations in leukocyte DNA have been found only in patients with PRTH (Table 2).

 

Table 2. Differential Diagnosis Between TSH-Secreting Adenomas (TSH-omas) and Resistance to Thyroid Hormones (RTH).

Parameter

TSH-omas

RTH

P

Female/Male ratio 

1.3 

1.4 

NS

Familial cases 

0 % 

85 % 

<0.0001

TSH mU/L 

3.0 ±0.4 

2.3 ±0.3

NS

FT4 pmol/L 

38.8 ±3.9 

29.9 ±2.3 

NS

FT3 pmol/L 

14.0 ±1.2 

11.3 ±0.8 

NS

Lesions at CT or MRI 

99 % 

23 % 

<0.0001

Germline THRB mutation

0%

84%

<0.0001

High biological activity of circulating serum TSH

38%

90%

NS

High alpha-GSU levels 

69 % 

3 % 

<0.0001

High alpha-GSU/TSH molar ratio 

81 % 

2 % 

<0.0001

Elevated SHBG and/or ICTP

90%

8% 

<0.0001

Blunted TSH response to TRH test (cutoff <3 mU/L in males and <5 mU/L in females)

87 % 

2 % 

<0.0001

Abnormal TSH response to T3 suppressiona

100 % 

100 %

NS

FT4/FT3 reduction/normalization during long-acting somatostatin analogc

92%

0%

<0.0001

a Werner's test (80-100 µg T3 for 8-10 days). Quantitatively normal responses to T3, i.e., complete inhibition of both basal and TRH-stimulated TSH levels, have never been recorded in either group of patients.

b Although abnormal in quantitative terms, TSH response to T3 suppression test was qualitatively normal in 45/47 PRTH patients.

c Two or more injections of somatostatin analogues (e.g., Octreotide-LAR 20-30 mg every month or Lanreotide Autogel 120 mg every 6-8 weeks)

Only patients with intact thyroid were taken into account. Data are obtained from patients followed at our department and are expressed as mean ± SE.

 

TREATMENT AND OUTCOME

 

As stated in 2013 guideline by European Thyroid Association (158), surgical resection is the recommended therapy for TSH-secreting pituitary tumors, with the aim of removing neoplastic tissue and restoring normal pituitary/thyroid function. However, radical removal of large tumors, that still represent the majority of TSH-omas, is particularly difficult because of the marked fibrosis of these tumors and the local invasion involving the cavernous sinus, internal carotid artery, or optic chiasm. Considering this high invasiveness, surgical removal or debulking of the tumor by transsphenoidal or subfrontal adenomectomy, depending on the tumor volume and its suprasellar extension, should be undertaken as soon as possible (158a). According to the review of 535 cases of TSH-omas by De Herdt and others, surgical resection of the adenoma was performed in 87.7% of patients of which 33.5% had residual pituitary adenoma (59a). Particular attention has to be paid to presurgical preparation of the patient, particularly in the preanesthetic period (159): antithyroid drugs or octreotide along with propranolol should be used, aiming at restoration of euthyroidism. In patients with very severe hyperthyroidism, the administration of iopanoic acid may be successfully employed (25). Though the preoperative treatment with somatostatin analogues may be useful to reduce hyperthyroid signs and symptoms in a significant number of patients, as well as the adenoma size (160-161a), no correlations between FT4/FT3 normalization and a higher rate of remission has been demonstrated (68). However, a recent multicenter, single-arm, phase 3 study in Japan confirmed in a series of TSH-omas that preoperative lanreotide autogel treatment was effective in normalizing thyroid function in 10/13 patients and to induce a -23.8% median percent change in pituitary tumor size from baseline at final assessment (161b). Finally, a single report on the efficacy of pasireotide in the pre-operative treatment of a TSH-omas has been so far published (161c).

 

It is worth noting that somatostatin analogues may lead to a condition of TSH deficiency. In this respect, in the series published by Illouz and others TSH deficiency appeared in 15% of 46 treated TSH-omas after a median time of 4 weeks, the TSH deficiency occurring after one to three injections of long-acting somatostatin analogues (161d). These data suggest that thyrotropic function should be reassessed after the first three injections of somatostatin analogues in order to diagnose TSH deficiency and to reduce the frequency of injections when control of thyrotoxicosis is the aim of the treatment.

 

After surgery, partial or complete hypopituitarism may result (162, 163). However, a case of thyroid storm after pituitary surgery was documented (164). Evaluation of pituitary function, particularly ACTH secretion, should be carefully undertaken soon after surgery and hormone replacement therapy initiated, if needed. In case of failure of pituitary surgery and in the presence of life-threatening hyperthyroidism, total thyroidectomy or thyroid ablation with radioiodine is indicated (165).

 

According to the largest published series, pituitary surgery is effective in restoring euthyroidism in 75% to 83% of patients with TSH-omas (61, 79, 165a) and a recent metanalysis by Cossu and others showed that the pooled rate of postoperative biochemical remission was 69.7% and a gross total resection was observed in 54% of patients. As expected, the extent of resection was significantly increased in microadenomas and cavernous sinus invasion was predictive of lower gross total resection (68).

 

If pituitary surgery is contraindicated or declined, as well as in the case of surgical failure, pituitary radiotherapy and/or medical treatment with somatostatin analogues (octreotide or lanretotide) are valid alternatives (158). In the case of radiotherapy, the recommended dose is no less than 45 Gy fractionated at 2 Gy per day or 10-25 Gy in a single dose if a stereotactic gamma knife is available (158, 166). Radiotherapy and radiosurgery are effective in normalizing thyroid function in 37% of patients within 2 years (79). The successful experience of an invasive TSH-oma associated with an unruptured aneurysm treated by two-stage operation and gamma knife has been reported (167). Although earlier diagnosis has improved the surgical cure rate of TSH-omas, several patients have required medical therapy in order to control the hyperthyroidism. Dopamine agonists, and particularly cabergoline, have been employed in some TSH-omas with variable results, positive effects being mainly observed in some patients with mixed PRL/TSH adenoma (121, 168, 169). Today, the medical treatment of TSH-omas relies on long-acting somatostatin analogues, such as octreotide or lanreotide (12, 13, 85, 158, 170-172). Indeed, many papers suggest the use of somatostatin analogues as first-line therapy for patients with TSH-omas, particularly for invasive macroadenomas (173-176). Treatment with these analogues lead to a reduction of TSH and alpha-GSU secretion in almost all cases, with restoration of the euthyroid state in the majority of them and it is safe even during pregnancy (18, 24, 177, 178). In some cases, inhibition of tumoral TSH secretion may be so profound that hypothyroidism may even be seen. During somatostatin analogues therapy tumor shrinkage occurs in about 50% of patients and vision improvement is seen in 75% (61, 79, 179). Very rapid shrinkage of the tumor has been described (34). Resistance to octreotide treatment has been documented in a few cases. Patients on somatostatin analogues have to be carefully monitored, as untoward side effects, such as cholelithiasis and carbohydrate intolerance, may become manifest. The dose administered should be tailored for each patient, depending on therapeutic response. Tolerance is usually very good, as gastrointestinal side effects are transient with long-acting analogues (12, 13, 57, 179, 180). As a whole, post-operative treatment with a somatostatin analogue induces a biochemical remission in 76% of patients (68) and led to a stable disease in 81.3% of the cases with residual tumor (59a).

 

CRITERIA OF CURE AND FOLLOW-UP

 

Due to the rarity of the disease and the great heterogeneity of the methods used, the criteria of cure of patients operated or irradiated for TSH-omas has not been clearly established. Previous thyroid ablation makes some of these criteria inapplicable (Table 3).

 

Table 3. Criteria for the Evaluation of the Outcome of Treatment

Criteria 

Comments

Remission from hyperthyroid

 manifestations (clinical and biochemical) 

Clinical improvement may be transient

No predictive value

Disappearance of neurological

 manifestations (adenoma imaging,

visual field defects, headache) 

May be transient

Poor predictive value

Normalization of free thyroid hormone levels 

Biochemical remission may be transient

Poor predictive value

Normalization of circulating TSH levels

Not applicable to patients with normal TSH

Poor predictive value

Undetectable TSH one week after

neurosurgery

Applicable to hyperthyroid patients that stopped treatments at least 10 days before surgery

Good prognostic value

Normalization of alpha-GSU levels and

alpha-GSU/TSH molar ratio 

Not applicable to patients with normal values before neurosurgery

Lack of sensitivity

Positive T3-suppression test with

 undetectable TSH and no response to TRH (or central hypothyroidism) 

Not applicable to elderly patients or in

 those with cardiac diseases

Optimal sensitivity/specificity and predictive

 value

 

In untreated hyperthyroid patients, it is reasonable to assume that cured patients have clinical and biochemical reversal of thyroid hyperfunction. However, the findings of normal free thyroid hormone concentrations or indices of peripheral thyroid hormone action (SHBG, ICTP, etc.) are not synonymous with complete removal or destruction of tumoral cells, since transient clinical remission accompanied by normalization of thyroid function is possible (62, 63, 67, 98, 115). Disappearance of neurological signs and symptoms is a good prognostic event, but lacks both sensitivity and specificity, as even an incomplete debulking of the tumor may cause visual field defects and headache to vanish. The resolution of specific neuroradiological abnormalities is confusing, since the pituitary imaging performed soon after surgery is often difficult to interpret. The criteria of normalization of circulating TSH are not applicable to previously thyroidectomized patients and to the 26% of patients with normal basal values of TSH. In our experience, undetectable TSH levels one week after surgery are likely to indicate complete adenomectomy, provided that the patient was hyperthyroid and presurgical treatments were stopped before surgery (115). A recent publication from a Korean group analyzing the outcome of adenomectomy in a series of 31 TSH-omas found that immediate postoperative TSH level at 12 hours after surgery was the strongest predictor of cure, with a 0.62 μIU/mL cutoff (165a). Normalization of alpha-GSU and/or the alpha-GSU/TSH molar ratio is in general a good index for the evaluation of therapy efficacy (8, 115). However, both parameters are characterized by less-than-optimal sensitivity, as they are normal in about 25% of patients with TSH-oma. The most sensitive and specific test to document the complete removal of the adenoma remains, in the absence of contraindication, the T3 suppression test (115). In fact, only patients in whom T3 administration completely inhibits basal and TRH-stimulated TSH secretion, appear to be truly cured.

 

Few data on the recurrence rates of TSH-oma in patients judged cured after surgery or radiotherapy have been reported. However, the recurrence of the adenoma does not appear to be frequent, at least in the first years after successful surgery (62, 115). In general, the patient should be evaluated clinically and biochemically 2 or 3 times the first year postoperatively, and then every year. Pituitary imaging should be performed every two or three years but should be promptly done whenever an increase in TSH and thyroid hormone levels, or clinical symptoms occur. In the case of a persistent macroadenoma, close visual field follow-up is required, as visual function could be threatened. Emergency surgical decompression is not always able to reverse even a recent visual deficit.

 

REFERENCES

 

  1. Beck-Peccoz P, Brucker-Davis F, Persani L, Smallridge RC, Weintraub BD. Thyrotropin-secreting pituitary tumors. Endocr Rev 1996; 17:610-638.
  2. Beck-Peccoz P, Lania A, Persani L. TSH-producing adenomas: in Endocrinology, Adult and Pediatric (7th Edition, vol. I), edited by Jameson LJ and DeGroot LJ. Sauderns Elsevier, Philadelphia, PA, 2015 pp.266-274.
  3. Jailer JW, Holub DA. Remission of Graves' disease following radiotherapy of a pituitary neoplasm. Am J Med 1960; 28:497-500.
  4. Hamilton C, Adams LC, Maloof F Hyperthyroidism due to thyrotropin-producing pituitary chromophobe adenoma. N Engl J Med 1970; 283:1077-1080.
  5. Refetoff S., Dumitrescu AM. Impaired sensitivity to thyroid hormone: defects of transport, metabolism and action, 2010 https://www.thyroidmanager.org/chapter/thyroid-hormone-resistance-syndromes/
  6. Gurnell M, Visser TJ, Beck-Peccoz P, Chatterjee VKK. Resistance to Thyroid Hormone: in Endocrinology, Adult and Pediatric (7th Edition, vol. II), edited by Jameson LJ and DeGroot LJ. Sauderns Elsevier, Philadelphia, PA, 2015 pp. 1648-1665.
  7. Hannoush ZC, Weiss RE. Defects of Thyroid Hormone Synthesis and Action. Endocrinol Metab Clin North Am. 2017;46:375-388.
  8. Brucker-Davis F, Oldfield EH, Skarulis MC, Doppman JL, Weintraub BD. Thyrotropin-secreting pituitary tumors: diagnostic criteria, thyroid hormone sensitivity, and treatment outcome in 25 patients followed at the National Institutes of Health. J Clin Endocrinol Metab 1999; 84: 476-486.
  9. Bertholon-Grégoire M, Trouillas J, Guigard MP, Loras B, Tourniaire J. Mono- and plurihormonal thyrotropic pituitary adenomas: pathological, hormonal and clinical studies in 12 patients. Eur J Endocrinol 1999; 140: 519-527.
  10. Taylor TJ, Donlon SS, Bale AE, Smallridge RC, Francis TB, Christensen RS, Burma KD. Treatment of a thyrotropinoma with octreotide-LAR in a patient with multiple endocrine neoplasia-1. Thyroid 2000; 10: 1001-1007.
  11. Kishida M, Otsuka F, Kataoka H, Yokota K, Oishi T, Yamauchi T, Doihara H, Tamiya T, Mimura Y, Ogura T, Makino H. Hyperthyroidism in a patient with TSH-producing pituitary adenoma coexisting with thyroid papillary adenocarcinoma. Endocr J 2000; 47:731-738.
  12. Kuhn JM, Arlot S, Lefebvre H; Caron P, Cortet-Rudelli C, Archambaud F, Chanson P, Tabarin A, Goth MI, Blumberg J, Catus F, Ispas S, Beck-Peccoz P. Evaluation of the treatment of thyrotropin-secreting pituitary adenomas with a slow release formulation of the somatostatin analog lanreotide. J Clin Endocrinol Metab 2000; 85:1487-1491.
  13. Caron P, Arlot S, Bauters C, Chanson P, Kuhn JM, Pugeat M, Marechaud R, Teutsch C, Vidal E, Sassano P. Efficacy of the long-acting octreotide formulation (octreotide-LAR) in patients with thyrotropin-secreting pituitary adenomas. J Clin Endocrinol Metab 2001; 86:2849-2853.
  14. Sunthornthepvarakul T, Prachasilchai P, Komindr A, Ngowngarmaratana S, Dansirikul R. A case of thyrotropin-secreting pituitary macroadenoma. J Med Assoc Thai 2001; 84: 897-906.
  15. Kon YC, Loh KC, Tambyah JA, Lim LH, Marshall JC. Thyrotropin (TSH)-secreting pituitary macroadenoma with cavernous sinus invasion. Singapore Med J 2001; 42: 433-439.
  16. Garcia Lafuente N, Berna Gascon MT, Pujalte Lopez E, Falco Jover G, Dominguez Escribano JR. Response of thyrotropinoma to somatostatin analogues: report of a case. Ann Med Interna 2001; 18: 663.
  17. Tella OI jr, Herculano MA, Delcello R, Prandini MN. TSH pituitary adenoma: case report. Arq Neuropsiquiatr 2002; 60:129-132.
  18. Blackhurst G, Strachan MW, Collie D, Gregor A, Statham PF, Seckl JE. The treatment of a thyrotropin-secreting pituitary macroadenoma with octreotide in twin pregnancy. Clin Endocrinol (Oxf). 2002;57:401-404.
  19. Kuo CS, Ho DM, Yang AH, Lin HD. Thyrotropin-secreting pituitary adenoma with growth hormone hypersecretion. Zhonghua Yi Xue Za Zhi (Taipei). 2002;65:489-493.
  20. Hsu FS, Tsai WS, Chau T, Chen HH, Chen YC, Lin SH. Thyrotropin-secreting pituitary adenoma presenting as hypokalemic periodic paralysis. Am J Med Sci. 2003;325:48-50.
  21. Pasquini E, Faustini-Fustini M, Sciarretta V, Saggese D, Roncaroli F, Serra D, Frank G. Ectopic TSH-secreting pituitary adenoma of the vomerosphenoidal junction. Eur J Endocrinol. 2003; 148: 253-257.
  22. Socin HV, Chanson P, Delemer B, Tabarin A, Rohmer V, Mockel J, Stevenaert A, Beckers A. The changing spectrum of TSH-secreting pituitary adenomas: diagnosis and management in 43 patients. Eur J Endocrinol. 2003; 148: 433-442.
  23. Wu YY, Chang HY, Lin JD, Chen KW, Huang YY, Jung SM. Clinical characteristics of patients with thyrotropin-secreting pituitary adenoma. J Formos Med Assoc. 2003; 102: 164-171.
  24. Chaiamnuay S, Moster M, Katz MR, Kim YN. Successful management of a pregnant woman with a TSH secreting pituitary adenoma with surgical and medical therapy. Pituitary. 2003;6:109-113.
  25. Dhillon KS, Cohan P, Kelly DF, Darwin CH, Iyer KV, Chopra IJ. Treatment of hyperthyroidism associated with thyrotropin-secreting pituitary adenomas with iopanoic acid. J Clin Endocrinol Metab. 2004; 89 :708-711.
  26. Soubhi Nizam M, Kennedy L. Co-occurrence of thyroid binding globulin excess and a pituitary macroadenoma containing thyroid stimulating hormone-producing cells. Postgrad Med J. 2004;80:114-115.
  27. Hubalewska-Hola A, Fross K, Kostecka-Matyja M, Sowa-Staszczak A, Szybinski Z, Huszno B, Ptak M. Macro- and microadenoma of thyrotropin secreting pituitary tumors--two clinical cases. Przegl Lek. 2003; 60: 768-171.
  28. Teramoto A, Sanno N, Tahara S, Osamura YR. Pathological study of thyrotropin-secreting pituitary adenoma: plurihormonality and medical treatment. Acta Neuropathol (Berl). 2004; 108: 147-153.
  29. Yang D, Tao W, Liu TH, Wei SZ. Thyrotropin-secreting pituitary adenoma:a clinicopathological analysis of seven cases. Zhonghua Bing Li Xue Za Zhi. 2004; 33: 208-211.
  30. Mannavola D, Persani L, Vannucchi G, Zanardelli M, Fugazzola L, Verga U, Facchetti M, Beck-Peccoz P. Different responses to chronic somatostatin analogues in patients with central hyperthyroidism. Clin Endocrinol (Oxf). 2005; 62: 176-181.
  31. Del Monte P, Bernasconi D, Ruelle A, Marugo A, Marugo M, Torre R. Effect of long-term treatment with octreotide-lar in a TSH-secreting pituitary macroadenoma and secondary hyperthyroidism. Minerva Endocrinol. 2005; 30: 95-99.
  32. Collie RB, Collie MJ. Extracranial thyroid-stimulating hormone-secreting ectopic pituitary adenoma of the nasopharynx. Otolaryngol Head Neck Surg. 2005; 133: 453-454.
  33. Usui T, Izawa S, Sano T, Tagami T, Nagata D, Shimatsu A, Takahashi JA, Naruse M. Clinical and molecular features of a TSH-secreting pituitary microadenoma. Pituitary. 2005; 8: 127-134.
  34. Atkinson JL, Abboud CF, Lane JI. Dramatic volume reduction of a large GH/TSH secreting pituitary tumor with short term Octreotide therapy. Pituitary. 2005; 8: 89-91.
  35. Brown RL, Muzzafar T, Wollman R, Weiss RE. A pituitary carcinoma secreting TSH and prolactin: a non-secreting adenoma gone awry. Eur J Endocrinol. 2006 May;154(5):639-43.
  36. Tan MJ, Tan F, Hawkins R, Cheah WK, Mukherjee JJ. A hyperthyroid patient with measurable thyroid-stimulating hormone concentration - a trap for the unwary. Ann Acad Med Singapore. 2006; 35: 500-503.
  37. Meas T, Sobngwi E, Vexiau P, Boudou P. An unusual somatotropin and thyreotropin secreting pituitary adenoma efficiently controlled by Octreotide and Pegvisomant. Ann Endocrinol (Paris). 2006; 67: 249-52.
  38. Gołkowski F, Buziak-Bereza M, Stefańska A, Trofimiuk M, Pantofliński J, Huszno B, Czepko R, Adamek D. A case of GH and TSH secreting pituitary macroadenoma. Przegl Lek. 2006; 63: 106-108.
  39. Losa M, Mortini P, Minelli R, Giovanelli M. Coexistence of TSH-secreting pituitary adenoma and autoimmune hypothyroidism. J Endocrinol Invest. 2006; 29 : 555-559.
  40. Kienitz T, Quinkler M, Strasburger CJ, Ventz M. Long-term management in five cases of TSH-secreting pituitary adenomas: a single center study and review of the literature. Eur J Endocrinol. 2007; 157: 39-46.
  41. Foppiani L, Del Monte P, Ruelle A, Bandelloni R, Quilici P, Bernasconi D. TSH-secreting adenomas: rare pituitary tumors with multifaceted clinical and biological features. J Endocrinol Invest. 2007; 30: 603-609.
  42. Ness-Abramof R, Ishay A, Harel G, Sylvetzky N, Baron E, Greenman Y, Shimon I. TSH-secreting pituitary adenomas: follow-up of 11 cases and review of the literature. Pituitary. 2007;10: 307-310.
  43. Chien WM, Garrison K, Caufield E, Orthel J, Dill J, Fero ML. Differential gene expression of p27Kip1 and Rb knockout pituitary tumors associated with altered growth and angiogenesis. Cell Cycle. 2007; 6: 750-757.
  44. Losa M, Fortunato M, Molteni L, Peretti E, Mortini P. Thyrotropin-secreting pituitary adenomas: biological and molecular features, diagnosis and therapy. Minerva Endocrinol. 2008; 33: 329-340.
  45. Clarke MJ, Erickson D, Castro MR, Atkinson JL. Thyroid-stimulating hormone pituitary adenomas. J Neurosurg. 2008; 109: 17-22.
  46. Caron P. Thyrotropin-secreting pituitary adenomas. Presse Med. 2009; 38: 107-111.
  47. Macchia E, Gasperi M, Lombardi M, Morselli L, Pinchera A, Acerbi G, Rossi G, Martino E. Clinical aspects and therapeutic outcome in thyrotropin-secreting pituitary adenomas: a single center experience. J Endocrinol Invest. 2009; 32: 773-779.
  48. Duarte FH, Jallad RS, Salgado LR, Bronstein MD. TSH-secreting pituitary tumors: two case reports and literature review. Arq Bras Endocrinol Metabol. 2009; 53: 1157-1166.
  49. Elston MS, Conaglen JV. Clinical and biochemical characteristics of patients with TSH-secreting pituitary adenomas from one New Zealand centre. Intern Med J. 2009; 40: 214-9.
  50. Marucci G, Faustini-Fustini M, Righi A, Pasquini E, Frank G, Agati R, Foschini MP. Thyrotropin-secreting pituitary tumors: significance of "atypical adenomas" in a series of 10 patients and association with Hashimoto thyroiditis as a cause of delay in diagnosis. J Clin Pathol. 2009; 62: 455-459.
  51. Latrech H, Rousseau A, Le Marois E, Billaud L, Bertagna X, Azzoug S, Raffin-Sanson ML. Manifestations and prognosis of thyrotropin-secreting pituitary adenomas: a case series of three patients. Rev Med Interne. 2010; 31: 858-862.
  52. Daly AF, Tichomirowa MA, Petrossians P, Heliövaara E, Jaffrain-Rea ML, Barlier A, Naves LA, Ebeling T, Karhu A, Raappana A, Cazabat L, De Menis E, Montañana CF, Raverot G, Weil RJ, Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E, Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo RA, Sabaté MI, Villa C, Popelier M, Salvatori R, Jennings J, Longás AF, Labarta Aizpún JI, Georgitsi M, Paschke R, Ronchi C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT, Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A, Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen LA, Beckers A.Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab. 2010; 95: E373-E383.
  53. Raappana A, Koivukangas J, Ebeling T, Pirilä T. Incidence of pituitary adenomas in Northern Finland in 1992-2007. J Clin Endocrinol Metab. 2010; 95: 4268-4275.
  54. Lee MT, Wang CY. Concomitant Graves hyperthyroidism with thyrotrophin-secreting pituitary adenoma. South Med J. 2010; 103: 347-349.
  55. Nguyen HD, Galitz MS, Mai VQ, Clyde PW, Glister BC, Shakir MK. Management of coexisting thyrotropin/growth-hormone-secreting pituitary adenoma and papillary thyroid carcinoma: a therapeutic challenge. Thyroid. 2010; 20: 99-103.
  56. Tagami T, Usui T, Shimatsu A, Beniko M, Yamamoto H, Moriyama K, Naruse M. Aberrant expression of thyroid hormone receptor beta isoform may cause inappropriate secretion of TSH in a TSH-secreting pituitary adenoma. J Clin Endocrinol Metab. 2011; 96: E948-E952.
  57. Gatto F, Barbieri F, Gatti M, Wurth R, Schulz S, Ravetti JL, Zona G, Culler MD, Saveanu A, Giusti M, Minuto F, Hofland LJ, Ferone D, Florio T. Balance between somatostatin and D2 receptor expression drives TSH-secreting adenoma response to somatostatin analogues and dopastatins. Clin Endocrinol (Oxf). 2012; 76: 407-414.
  58. Nakayama Y, Jinguji S, Kumakura S, Nagasaki K, Natsumeda M, Yoneoka Y, Saito T, Fujii Y. Thyroid-stimulating hormone (thyrotropin)-secretion pituitary adenoma in an 8-year-old boy: case report. Pituitary. 2012; 15: 110-115.

58a.     Yang Y, Liu J, Deng K, Lu L, Zhu H, Lian X, Bao X, Duan L, Yao Y. Clinical and Therapeutic Characteristics of Pituitary TSH-Secreting Adenoma in Adolescent-Onset Patients: Six Case Studies and Literature Review. Front Endocrinol (Lausanne). 2021; 12:771673.

  1. Rabbiosi S, Peroni E, Tronconi GM, Chiumello G, Losa M, Weber G. Asymptomatic thyrotropin-secreting pituitary macroadenoma in a 13-year-old girl: successful first-line treatment with somatostatin analogues. Thyroid. 2012; 22: 1076-1079.

59a.     De Herdt C, Philipse E, De Block C. ENDOCRINE TUMORS: Thyrotropin-secreting pituitary adenoma: a structured review of 535 adult cases. Eur J Endocrinol. 2021; 185(2):R65-R74.

  1. Lee W, Cheung AS, Freilich R. TSH-secreting pituitary carcinoma with intrathecal drop metastases. Clin Endocrinol (Oxf). 2012; 76: 604-606.
  2. Yamada S, Fukuhara N, Horiguchi K, Yamaguchi-Okada M, Nishioka H, Takeshita A, Takeuchi Y, Ito J, Inoshita N. Clinicopathological characteristics and therapeutic outcomes in thyrotropin-secreting pituitary adenomas: a single-center study of 90 cases. J Neurosurg. 2014; 121: 1462-1473.
  3. Kirkman MA, Jaunmuktane Z, Brandner S, Khan AA, Powell M, Baldeweg SE. Active and silent thyroid-stimulating hormone-expressing pituitary adenomas: presenting symptoms, treatment, outcomes, and recurrence. World Neurosurg. 2014; 82: 1224-1231.
  4. van Varsseveld NC, Bisschop PH, Biermasz NR, Pereira AM, Fliers E, Drent ML. A long-term follow-up study of eighteen patients with thyrotrophin-secreting pituitary adenomas. Clin Endocrinol (Oxf). 2014; 80: 395-402.
  5. Azzalin A, Appin CL, Schniederjan MJ, Constantin T, Ritchie JC, Veledar E, Oyesiku NM, Ioachimescu AG. Comprehensive evaluation of thyrotropinomas: single-center 20-year experience. Pituitary. 2016; 19: 183-193.
  6. Astaf'eva LI, Kadashev BA, Shishkina LV, Kalinin PL, Fomichev DV, Kutin MA, Aref'eva IA, Dzeranova LK, Sidneva YG, Klochkova IS, Rotin DL. Clinical and morphological characteristics, diagnostic criteria, and outcomes of surgical treatment of TSH-secreting pituitary adenomas. Zh Vopr Neirokhir Im N N Burdenko. 2016; 80: 24-35.
  7. Tjörnstrand A, Nyström HF. DIAGNOSIS OF ENDOCRINE DISEASE: Diagnostic approach to TSH-producing pituitary adenoma. Eur J Endocrinol. 2017; 177: R183-R197.
  8. Nazato DM, Abucham J. Diagnosis and treatment of TSH-secreting adenomas: review of a longtime experience in a reference center. J Endocrinol Invest. 2018; 41: 447-454.
  9. Cossu G, Daniel RT, Pierzchala K, Berhouma M, Pitteloud N, Lamine F, Colao A, Messerer M. Thyrotropin-secreting pituitary adenomas: a systematic review and meta-analysis of postoperative outcomes and management. Pituitary. 2019 Feb;22(1):79-88.
  10. Asioli S, Righi A, Iommi M, Baldovini C, Ambrosi F, Guaraldi F, Zoli M, Mazzatenta D, Faustini-Fustini M, Rucci P, Giannini C, Foschini MP. Validation of a clinico-pathological score for the prediction of post-surgical evolution of pituitary adenoma: retrospective analysis on 566 patients from a tertiary care centre. Eur J Endocrinol. Eur J Endocrinol. 2019 Feb 1;180(2):127-134
  11. Önnestam L, Berinder K, Burman P, Dahlqvist P, Engström BE, Wahlberg J, Filipsson Nyström H. National incidence and prevalence of TSH-secreting pituitary adenomas in Sweden. J Clin Endocrinol Metab. 2013; 98: 626-635.
  12. Sanno N, Teramoto A, Osamura RY. Thyrotropin-secreting pituitary adenomas. Clinical and biological heterogeneity and current treatment. J Neurooncol 2001; 54: 179-186.

71a.     Căpraru OM, Gaillard C, Vasiljevic A, Lasolle H, Borson-Chazot F, Raverot V, Jouanneau E, Trouillas J, Raverot G. Diagnosis, pathology, and management of TSH-secreting pituitary tumors. A single-center retrospective study of 20 patients from 1981 to 2014. 2019 Ann Endocrinol (Paris). 80(4):216-224.

  1. Terzolo M, Orlandi F, Bassetti M, Medri G, Paccotti P, Cortelazzi D, Angeli A, Beck-Peccoz P. Hyperthyroidism due to a pituitary adenoma composed of two different cell types, one secreting alpha-subunit alone and another cosecreting alpha-subunit and thyrotropin. J Clin Endocrinol Metab 1991; 72:415-421.
  2. Pereira BD, Raimundo L, Mete O, Oliveira A, Portugal J, Asa SL. Monomorphous plurihormonal pituitary adenoma of Pit-1 lineage in a giant adolescent with central hyperthyroidism. Endocr Pathol. 2016; 27: 25-33.
  3. Berker D, Isik S, Aydin Y, Tutuncu Y, Akdemir G, Ozcan HN, Guler S. Thyrotropin secreting pituitary adenoma accompanying a silent somatotropinoma. Turk Neurosurg. 2011; 21: 403-407.
  4. Banerjee AK, Sharma BS, Kak VK. Clinically and biochemically silent thyrotrope adenoma with oncocytic change. Neurol India 2000; 48: 374-377.
  5. Pawlikowski M, Pisarek H, Jaranowska M, Radek M, Winczyk K, Kunert-Radek J. "Silent" thyrotropin (TSH) expression in acromegaly and clinicallynon-functioning pituitary adenomas. Endokrynol Pol. 2016; 67: 515-518.
  6. Iskandar SB, Supit E, Jordan RM, Peiris AN. Thyrotropin-secreting pituitary tumor and Hashimoto's disease: a novel association. South Med J. 2003; 96: 933-936.
  7. Ma W, Ikeda H, Watabe N, Kanno M, Yoshimoto T. A plurihormonal TSH-producing pituitary tumor of monoclonal origin in a patient with hypothyroidism. Horm Res. 2003; 59: 257-261. 78

78a.     Sakata K, Fujimori K, Komaki S, Furuta T, Sugita Y, Ashida K, Nomura M, Morioka M.
Pituitary Gangliocytoma Producing TSH and TRH:  A Review of “Gangliocytomas of the Sellar Region”J Clin Endocrinol Metab. 2020 105(10):3109-3121.

  1. Malchiodi E, Profka E, Ferrante E, Sala E, Verrua E, Campi I, Lania AG, Arosio M, Locatelli M, Mortini P, Losa M, Beck-Peccoz P, Spada A, Mantovani G. Thyrotropin-secreting pituitary adenomas: outcome of pituitary surgery and irradiation. J Clin Endocrinol Metab. 2014;99: 2069-2076.
  2. Ando S, Sarlis NJ, Krishnan J, Feng X, Refetoff S, Zhang MQ, Oldfield EH, Yen PM. Aberrant alternative splicing of thyroid hormone receptor in a TSH-secreting pituitary tumor is a mechanism for hormone resistance. Mol Endocrinol. 2001; 15: 1529-1538.
  3. Ando S, Sarlis NJ, Oldfield EH, Yen PM. Somatic mutation of TRbeta can cause a defect in negative regulation of TSH in a TSH-secreting pituitary tumor. J Clin Endocrinol Metab. 2001; 86: 5572-5576.
  4. Teng X, Jin T, Brent GA, Wu A, Teng W, Shan Z. A patient with a thyrotropin-secreting microadenoma and resistance to thyroid hormone (P453T). J Clin Endocrinol Metab. 2015; 100: 2511-2514.
  5. Tannahill LA, Visser TJ, McCabe CJ, Kachilele S, Boelaert K, Sheppard MC, Franklyn JA, Gittoes NJ. Dysregulation of iodothyronine deiodinase enzyme expression and function in human pituitary tumors. Clin Endocrinol (Oxf). 2002; 56: 735-743.
  6. Baur A, Buchfelder M, Kohrle J. Expression of 5'-deiodinase enzymes in normal pituitaries and in various human pituitary adenomas. Eur J Endocrinol. 2002; 147: 263-268.
  7. Webster J, Peters JR, John R, Smith J, Chan V, Hall R, Scanlon MF. Pituitary stone: two cases of densely calcified thyrotropin-secreting pituitary adenomas. Clin Endocrinol (Oxf) 1994; 40:137-143.
  8. Ezzat S, Horvath E, Kovacs K, Smyth HS, Singer W, Asa SL. Basic fibroblast growth factor expression by two prolactin and thyrotropin-producing pituitary adenomas. Endocr Pathol 1995; 6:125-134.
  9. Trouillas J, Girod C, Loras B, Claustrat B, Sassolas G, Perrin G, Buonaguidi R. The TSH secretion in the human pituitary adenomas. Pathol Res Pract 1988; 183: 596-600.
  10. Mixson AJ, Friedman TC, David AK, Feuerstein IM, Taubenberger JK, Colandrea JM, Doppman JL, Oldfield EH, Weintraub BD. Thyrotropin-secreting pituitary carcinoma. J Clin Endocrinol Metab 1993; 76: 529-533.
  11. Lu C, Willingham MC, Furuya F, Cheng SY. Activation of phosphatidylinositol 3-kinase signaling promotes aberrant pituitary growth in a mouse model of thyroid-stimulating hormone-secreting pituitary tumors. Endocrinology. 2008; 149: 3339-3345.
  12. Dong Q, Brucker-Davis F, Weintraub BD, Smallridge RC, Carr FE, Battey J, Spiegel AM, Shenker A. Screening of candidate oncogenes in human thyrotrope tumors: absence of activating mutations of the Gq, G11, Gs, or thyrotropin-releasing hormone receptor genes. J Clin Endocrinol Metab. 1996; 81: 1134-1140.
  13. Friedman E, Adams EF, Höög A, Gejman PV, Carson E, Larsson C, De Marco L, Werner S, Fahlbusch R, Nordenskjöld M. Normal structural dopamine type 2 receptor gene in prolactin-secreting and other pituitary tumors. J Clin Endocrinol Metab. 1994; 78: 568-574.
  14. Asteria C, Anagni M, Persani L, Beck-Peccoz P. Loss of heterozigosity of the MEN1 gene in a large series of TSH-secreting pituitary adenomas. J Endocrinol Invest. 2001; 24: 796-801.
  15. Filopanti M, Ballare E, Lania AG et al. Loss of heterozygosity at the SS receptor type 5 locus in human GH- and TSH-secreting pituitary adenomas. J Endocrinol Invest. 2004; 27: 937-942.
  16. Barlier A, Vanbellinghen JF, Daly AF, Silvy M, Jaffrain-Rea ML, Trouillas J, Tamagno G, Cazabat L, Bours V, Brue T, Enjalbert A, Beckers A. Mutations in the aryl hydrocarbon receptor interacting protein gene are not highly prevalent among subjects with sporadic pituitary adenomas. J Clin Endocrinol Metab. 2007; 92: 1952-1955.
  17. Sapkota S, Horiguchi K, Tosaka M, Yamada S, Yamada M. Whole-exome sequencing study of thyrotropin-secreting pituitary adenomas. J Clin Endocrinol Metab. 2017; 102: 566-75.

95a.     Neou M, Villa C, Armignacco R, Jouinot A, Raffin-Sanson ML, Septier A, Letourneur F, Diry S, Diedisheim M, Izac B, Gaspar C, Perlemoine K, Verjus V, Bernier M, Boulin A, Emile JF, Bertagna X, Jaffrezic F, Laloe D, Baussart B, Bertherat J, Gaillard S, Assié G. Pangenomic Classification of Pituitary Neuroendocrine Tumors. Cancer Cell. 2020;37(1):123-134.

  1. Watanabe K, Kameya T, Yamauchi A, Yamamoto N, Kuwayama A, Takei I, Maruyama H, Saruta T. Thyrotropin-producing adenoma associated with pituitary resistance to thyroid hormone. J Clin Endocrinol Metab 1993; 76:1025-1030.
  2. Safer JD, Colan SD, Fraser LM, Wondisford FE. A pituitary tumor in a patient with thyroid hormone resistance: a diagnostic dilemma. Thyroid 2001; 11: 281-291.
  3. Beck-Peccoz P, Persani L. TSH-induced hyperthyroidism caused by a pituitary tumor. Nat Clin Pract Endocrinol Metab. 2006; 2: 524-528.
  4. Sriphrapradang C, Srichomkwun P, Refetoff S, Mamanasiri S. A Novel thyroid hormone receptor beta gene mutation (G251V) in a Thai patient with resistance to thyroid hormone coexisting with pituitary incidentaloma. Thyroid. 2016; 26: 1804-806.
  5. Gancel A, Vuillermet P, Legrand A, Catus F, Thomas F, Kuhn J M. Effets of a slow-release formulation of the new somatostatin analogue lanreotide in TSH-secreting pituitary adenomas. Clin Endocrinol (Oxf). 1994; 40: 421-428.
  6. Bertherat J, Brue T, Enjalbert A, Gunz G, Rasolonjanahary R, Warnet A, Jaquet P, Epelbaum J. Somatostatin receptors on thyrotropin-secreting pituitary adenomas: comparison with the inhibitory effects of octreotide upon in vivo and in vitro hormonal secretions. J Clin Endocrinol Metab. 1992; 75: 540-546.
  7. Yoshihara A, Isozaki O, Hizuka N, Nozoe Y, Harada C, Ono M, Kawamata T, Kubo O, Hori T, Takano K. Expression of type 5 somatostatin receptor in TSH-secreting pituitary adenomas: a possible marker for predicting long-term response to octreotide therapy. Endocr J. 2007; 54: 133-138.
  8. Gatto F, Barbieri F, Castelletti L, Arvigo M, Pattarozzi A, Annunziata F, Saveanu A, Minuto F, Castellan L, Zona G, Florio T, Ferone D. In vivo and in vitro response to octreotide LAR in a TSH-secreting adenoma: characterization of somatostatin receptor expression and role of subtype 5. Pituitary. 2011; 14: 141-147.
  9. Horiguchi K, Yamada M, Umezawa R, Satoh T, Hashimoto K, Tosaka M, Yamada S, Mori M. Somatostatin receptor subtypes mRNA in TSH-secreting pituitary adenomas: a case showing a dramatic reduction in tumor size during short octreotide treatment. Endocr J. 2007; 54: 371-378.

104a    Han R, Shen L, Zhang J, Xie J, Fang W, Sun Q, Bian L, Zhou Y, Wang S, Ning G, Wang W, Ye L. Diagnosing Thyrotropin-Secreting Pituitary Adenomas by Short-Term Somatostatin Analogue Test. Thyroid. 2020; 30(9):1236-1244.

  1. Wood DF, Johnston JM, Johnston DG. Dopamine, the dopamine D2 receptor and pituitary tumors. Clin Endocrinol (Oxf). 1991; 35: 455-466.
  2. Verhoeff NPLG, Bemelman FJ, Wiersinga WM, et al: Imaging of dopamine D2 and somatostatin receptors in vivo using single-photon emission tomography in a patient with a TSH/PRL-producing pituitary macroadenoma, Eur J Nucl Med. 1993; 20: 555-560.
  3. Gesundheit N, Petrick P, Nissim M, Dahlberg PA, Doppman L, Emerson CH, Braverman LE, Oldfield EH, Weintraub BD. Thyrotropin-secreting pituitary adenomas: clinical and biochemical heterogeneity. Ann Intern Med 1989; 111: 827-835.
  4. Spada A, Bassetti M, Martino E, Giannattasio G, Beck-Peccoz P, Sartorio P, Vallar L, Baschieri L, Pinchera A, Faglia G. In vitro studies on TSH secretion and adenylate cyclase activity in a human TSH-secreting pituitary adenoma. Effects of somatostatin and dopamine. J Endocrinol Invest 1985; 8:193-198.
  5. Rocheville M, Lange DC, Kumar U, Patel SC, Patel RC, Patel YC. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science 2000; 288:154-157.
  6. Olt S, Şirik M. TSH secreting adenoma: a rare cause of severe headache. Pan Afr Med J. 2016; 23: 2.
  7. Kamoun M, d'Herbomez M, Lemaire C, Fayard A, Desailloud R, Huglo D, Wemeau JL. Coexistence of thyroid-stimulating hormone-secreting pituitary adenoma and Graves' hyperthyroidism. Eur Thyroid J. 2014; 3: 60-4.

111a.   Fu J, Wu A, Wang X, Guan H. Concurrent Graves' Disease and TSH Secreting Pituitary Adenoma Presenting Suppressed Thyrotropin Levels: A Case Report and Review of the Literature. Front Endocrinol (Lausanne). 2020 Aug 6;11:523.

111b.   Quinn M, Bashari W, Smith D, Gurnell M, Agha A. A remarkable case of thyrotoxicosis initially caused by graves' disease followed by a probable TSHoma - a case report. BMC  Endocr Disord. 2020 Aug 27;20(1):133

111c.   Arai N, Inaba M, Ichijyo T, Kagami H, Mine Y. Thyrotropin-producing pituitary adenoma simultaneously existing with Graves' disease: a case report. J Med Case Rep. 2017;11(1):9.

  1. Daya SK, Paulus AO, Braxton EE Jr, Vroman PJ, Mathis DA, Lin R, True MW. Delayed diagnosis of TSH-Secreting adenoma attributed to worsening post-traumatic stress disorder symptoms in a military veteran because of provider anchoring bias. Mil Med. 2017; 182: e1849-e1853.

112a.   Page KA, Roehmholdt BF, Jablonski M & Mayerson AB. Development of thyroid storm after surgical resection of a thyrotropin-secreting pituitary adenoma. Endocrine Practice. 2008; 14: 732-737.

112b.   Fujio S, Ashari, Habu M, Yamahata H, Moinuddin FM, Bohara M, Arimura H, Nishijima Y & Arita K. Thyroid storm induced by TSHsecreting pituitary adenoma: a case report. Endocrine Journal 2014; 61: 1131-1136.

  1. Lim EM, Bhagat CI, Walsh J. Asymptomatic thyrotropin-secreting pituitary microadenoma. Intern Med J. 2001; 31: 428-429.
  2. Beck-Peccoz P, Piscitelli G, Amr S, Ballabio M, Bassetti M, Giannattasio G, Spada A, Nissim M, Weintraub BD, Faglia G. Endocrine, biochemical, and morphological studies of a pituitary adenoma secreting growth hormone, thyrotropin (TSH), and alpha-subunit: evidence for secretion of TSH with increased bioactivity. J Clin Endocrinol Metab. 1986; 62: 704-711.
  3. Losa M, Giovanelli M, Persani L, Mortini P, Faglia G, Beck-Peccoz P. Criteria of cure and follow-up of central hyperthyroidism due to thyrotropin-secreting pituitary adenomas. J Clin Endocrinol Metab. 1996; 81: 3086-3090.
  4. Maisnam I, Dutta D, Jain R, Ghosh S, Mukhopadhyay S, Chowdhury S. Plurihormone secreting pituitary macroadenoma masquerading as thyrotoxicosis: Clinical presentation and diagnostic challenges. Indian J Endocrinol Metab. 2012;16 (Suppl 2): S315-S317.
  5. Glynn N, Agha A. Unexpected clinical course during treatment of a TSH-secreting pituitary adenoma. Endocr Pract. 2013; 19: e88-e91.
  6. Johnston PC, Hamrahian AH, Prayson RA, Kennedy L, Weil RJ. Thyrotoxicosis with absence of clinical features of acromegaly in a TSH- and GH-secreting,invasive pituitary macroadenoma. Endocrinol Diabetes Metab Case Rep. 2015; 2015: 140070.
  7. George JT, Thow JC, Matthews B, Pye MP, Jayagopal V. Atrial fibrillation associated with a thyroid stimulating hormone-secreting adenoma of the pituitary gland leading to a presentation of acute cardiac decompensation: a case report. J Med Case Reports. 2008; 2: 67.
  8. Fujio S, Ashari, Habu M, Yamahata H, Moinuddin FM, Bohara M, Arimura H, Nishijima Y, Arita K. Thyroid storm induced by TSH-secreting pituitary adenoma: a case report. Endocr J. 2014; 61: 1131-1136.
  9. Kao YH, Chang TJ, Huang TS. Thyrotropin-secreting pituitary tumor presenting with congestive heart failure and good response to dopaminergic agonist cabergoline. J Formos Med Assoc. 2013; 112: 721-724.
  10. Mousiolis AC, Rapti E, Grammatiki M, Yavropoulou M, Efstathiou M, Foroglou N, Daniilidis M, Kotsa K. Somatostatin analogue treatment of a TSH-secreting adenoma presenting with accelerated bone metabolism and a pericardial effusion: a case report. Medicine (Baltimore). 2016; 95: e2358.
  11. Lee JH, Park M, Park MJ, Jo YS. Massive pleural and pericardial effusion dueto hypothyroidism in a patient with a surgically treated thyroid-stimulating hormone-producing pituitary adenoma. Acta Clin Belg. 2018; 73: 398-401.
  12. Pappa T, Papanastasiou L, Markou A, Androulakis I, Kontogeorgos G, Seretis A, Piaditis G. Thyrotoxic periodic paralysis as the first manifestation of a thyrotropin-secreting pituitary adenoma. Hormones (Athens). 2010; 9: 82-86.
  13. Frara S, Losa M, Doga M, Formenti AM, Mortini P, Mazziotti G, Giustina A. High prevalence of radiological vertebral fractures in patients with TSH-secreting pituitary adenoma. J Endocr Soc. 2018; 2: 1089-1099.
  14. Abs R, Stevenaert A, Beckers A. Autonomously functioning thyroid nodules in a patient with a thyrotropin-secreting pituitary adenoma: possible cause-effect relationship. Eur J Endocrinol. 1994; 131: 355-358.
  15. Aksoy DY, Gedik A, Cinar N, Soylemezoglu F, Berker M, Gurlek OA. Thyrotropinoma and multinodular goiter: A diagnostic challenge for hyperthyroidism. J Res Med Sci. 2013; 18: 1008-1010.
  16. Gasparoni P, Rubello D, Persani L, Beck-Peccoz P. Unusual association between a thyrotropin-secreting pituitary adenoma and a papillary thyroid carcinoma. Thyroid 1998; 8: 181-183.
  17. Poggi M, Monti S, Pascucci C, Toscano V. A rare case of follicular thyroid carcinoma in a patient with thyrotropin-secreting pituitary adenoma. Am J Med Sci. 2009; 337: 462-465.
  18. Perticone F, Pigliaru F, Mariotti S, Deiana L, Furlani L, Mortini P, Losa M. Is the incidence of differentiated thyroid cancer increased in patients with thyrotropin-secreting adenomas? Report of three cases from a large consecutive series. Thyroid. 2015; 25: 417-424.
  19. Ünlütürk U, Sriphrapradang C, Erdoğan MF, Emral R, Güldiken S, Refetoff S, Güllü S. Management of differentiated thyroid cancer in the presence ofresistance to thyroid hormone and TSH-secreting adenomas: a report of four cases and review of the literature. J Clin Endocrinol Metab. 2013; 98: 2210-2217.
  20. Azukizawa M, Morimoto S, Miyai K, Miki T, Yabu Y, Amino N, Kuma K, Kumahara Y. TSH-producing pituitary adenoma associated with Graves' disease. In: Stockigt JR, Nagataki S (eds) Thyroid Research, vol.VIII, Australian Academy of Sciences, Canberra, 1980: 645-648.
  21. Yovos JG, Falko JM, O'Dorisio TM, Malarkey WN, Cataland S, Capen CC. Thyrotoxicosis and a thyrotropin-secreting pituitary tumor causing unilateral exophthalmos. J Clin Endocrinol Metab 1981; 53: 338-343.

133a.   Kaneto H, Kamei S, Tatsumi F, Shimoda M, Kimura T, Obata A, Anno T, Nakanishi S, Kaku K, Mune T. Syndrome of inappropriate secretion of TSH in a subject with galactorrhea and menstrual disorder and undergoing infertility treatment: Case report. Medicine (Baltimore). 2021;100(52): e28414.

  1. Sy ARG, Bernstein R, Chynn KI, Kourides IA. Reduction in size of a thyrotropin- and gonadotropin-secreting pituitary adenoma treated with octreotide acetate (somatostatin analogue). J Clin Endocrinol Metab 1992; 74: 690-694.
  2. Li J, Li J, Jiang S, Yu R, Yu Y. Case report of a pituitary thyrotropin-secreting macroadenoma with Hashimoto thyroiditis and infertility. Medicine (Baltimore). 2018; 97: e9546.
  3. Beck-Peccoz P, Piscitelli G, Cattaneo MG, Faglia G, White EL, Barlow JW, Stockigt JR. Evaluation of free thyroxine methods in the presence of iodothyronine binding autoantibodies. J Clin Endocrinol Metab. 1984; 58: 736-739.
  4. Koulouri O, Moran C, Halsall D, Chatterjee K, Gurnell M. Pitfalls in the measurement and interpretation of thyroid function tests. Best Pract Res Clin Endocrinol Metab. 2013; 27: 745-762.
  5. Schoenmakers N, Moran C, Campi I, Agostini M, Bacon O, Rajanayagam O, Schwabe J, Bradbury S, Barrett T, Geoghegan F, Druce M, Beck-Peccoz P, O'Toole A, Clark P, Bignell M, Lyons G, Halsall D, Gurnell M, Chatterjee K. A novel albumin gene mutation (R221I) in familial dysalbuminemic hyperthyroxinemia. J Clin Endocrinol Metab. 2014; 99: E1381-E1386.
  6. Zweig MH, Csako G, Spero M. Escape from blockade of interfering heterophile antibodies in a two-site immunoradiometric assay for thyrotropin. Clin Chem. 1988; 34: 2589-2591.
  7. Srivastava, R, Bartlett WA, Kennedy IM, Hiney A, Fletcher C, Murphy MJ. Reflex and reflective testing: efficiency and effectiveness of adding on laboratory tests. Ann. Clin. Biochem. 2010; 47: 223-227.
  8. Wardle CA, Fraser WD, Squire CR. Pitfalls in the use of thyrotropin concentration as a first-line thyroid-function test. Lancet 2001; 357: 1013-1014.
  9. Roelfsema F, Kok S, Kok P, Pereira AM, Biermasz NR, Smit JW, Frolich M, Keenan DM, Veldhuis JD, Romijn JA. Pituitary-hormone secretion by thyrotropinomas. Pituitary. 2009; 12: 200-210.
  10. Okuma H, Hashimoto K, Ohashi T, Mihara M, Minami I, Izumiyama H, Sasaki S, Inoshita N, Nishioka H, Yamada S, Yoshimoto T. A case of TSH-secreting pituitary adenoma with cyclic fluctuations in serum TSH levels. Endocr J. 2018; 65: 737-746.
  11. Beck-Peccoz P, Persani L. Variable biological activity of thyroid-stimulating hormone. Eur J Endocrinol 1994; 131: 331-340.
  12. Magner J, Klibanski A, Fein H, Smallridge R, Blackard W, Young W Jr, Ferriss JB, Murphy D, Kane J, Rubin D. Ricin and lentil lectin affinity chromatography reveals oligosaccharide heterogeneity of thyrotropin secreted by 12 human pituitary tumors. Metabolism 1992; 41: 1009-1015.
  13. Beck-Peccoz P, Persani L, Faglia G. Glycoprotein hormone -subunit in pituitary adenomas. Trends Endocrinol Metab 1992; 3: 41-45.
  14. Beck-Peccoz P, Roncoroni R, Mariotti S, Medri G, Marcocci C, Brabant G, Forloni F, Pinchera A, Faglia G. Sex hormone-binding globulin measurement in patients with inappropriate secretion of thyrotropin (IST): evidence against selective pituitary thyroid hormone resistance in nonneoplastic IST. J Clin Endocrinol Metab 1990; 71: 19-25.
  15. Persani L, Preziati D, Matthews CH, Sartorio A, Chatterjee VK, Beck-Peccoz P. Serum levels of carboxyterminal cross-linked telopeptide of type I collagen (ICTP) in the differential diagnosis of the syndromes of inappropriate secretion of TSH. Clin Endocrinol (Oxf.) 1997; 47: 207-214.
  16. Bonomi M, Proverbio MC, Weber G, Chiumello G, Beck-Peccoz P, Persani L. Hyperplastic pituitary gland, high serum glycoprotein hormone alpha-subunit, and variable circulating thyrotropin (TSH) levels as hallmark of central hypothyroidism due to mutations of the TSH beta gene. J Clin Endocrinol Metab. 2001; 86: 1600-1604.
  17. Losa M, Magnani P, Mortini P, Persani L, Acerno S, Giugni E, Fazio F, Beck-Peccoz P, Giovanelli M. Indium-111 pentetreotide single-photon emission tomography in patients with TSH-secreting pituitary adenomas: correlation with the effect of a single administration of octreotide on serum TSH levels. Eur J Nucl Med. 1997; 24: 728-731.
  18. Brandstätter W, Hatzl M, Weis S, Javor A, Gabriel M. Successful resection of TSH-secreting pituitary adenoma demonstrated by serial 99mTc-scintigraphy. Nuklearmedizin. 2015; 54: N23-N24.
  19. Cooper DS, Wenig BM. Hyperthyroidism caused by an ectopic TSH-secreting pituitary tumor. Thyroid. 1996; 6: 337-343.
  20. Song M, Wang H, Song L, Tian H, Ge Q, Li J, Zhu Y, Li J, Zhao R, Ji HL. Ectopic TSH-secreting pituitary tumor: a case report and review of prior cases. BMC Cancer. 2014; 14: 544.
  21. Tong A, Xia W, Qi F, Jin Z, Yang D, Zhang Z, Li F, Xing X, Lian X. Hyperthyroidism caused by an ectopic thyrotropin-secreting tumor of the nasopharynx: a case report and review of the literature. Thyroid. 2013; 23: 1172-1177.
  22. Wang Q, Lu XJ, Sun J, Wang J, Huang CY, Wu ZF. Ectopic suprasellar thyrotropin-secreting pituitary adenoma: case report and literature review. World Neurosurg. 2016; 95: 617.e13-617.e18.
  23. Yang J, Liu S, Yang Z, Shi YB. Ectopic thyrotropin secreting pituitary adenoma concomitant with papillary thyroid carcinoma: Case report. Medicine (Baltimore). 2017; 96: e8912.
  24. Hanaoka Y, Ogiwara T, Kakizawa Y, Nagm A, Seguchi T, Aoyama T, Koyama JI, Sato A, Hongo K. Calcified ectopic TSH-secreting pituitary adenoma mimicking craniopharyngioma: a rare case report and literature review. Acta Neurochir (Wien). 2018; 160: 2001-2005.

157a.   Kim S, Dillon WP, Hope TA, El-Sayed IH, van Zante A, Wu K, Masharani U. Ectopic Thyroid-Stimulating Hormone-Secreting Pituitary Adenoma of the Nasopharynx Diagnosed by Gallium 68 DOTATATE Positron Emission Tomography/Computed Tomography. World Neurosurg. 2019; 125: 400-404.

157b.   Campi I, Covelli D, Moran C, Fugazzola L, Cacciatore C, Orlandi F, Gallone G, Chatterjee K, Beck-Peccoz P, Persani L. The Differential Diagnosis of Discrepant Thyroid Function Tests: Insistent Pitfalls and Updated Flow-Chart Based on a Long-Standing Experience. Front Endocrinol (Lausanne). 2020 Jul 7;11:432

157c.   Mimoto MS, Refetoff S. Clinical recognition and evaluation of patients with inherited serum thyroid hormone-binding protein mutations. J Endocrinol Invest. 2020 Jan;43(1):31-41

  1. Beck-Peccoz P, Lania A, Beckers A, Chatterjee K, Wemeau JL. European thyroid association guidelines for the diagnosis and treatment of thyrotropin-secreting pituitary tumors. Eur Thyroid J. 2013; 2: 76-82.

158a.   Varlamov EV, McCartney S, Fleseriu M. 2019 Functioning pituitary adenomas – Current treatment options and emerging medical therapies. Eur Endocrinol. 201; 15(1):30-40.

  1. Dyer MW, Gnagey A, Jones BT, Pula RD, Lanier WL, Atkinson JLD, Pasternak JJ. Perianesthetic management of patients with thyroid-stimulating hormone-secreting pituitary adenomas. J Neurosurg Anesthesiol. 2017; 29: 341-346.
  2. Wallace IR, Healy E, Cooke RS, Ellis PK, Harper R, Hunter SJ. TSH-secreting pituitary adenoma: benefits of pre-operative octreotide. Endocrinol Diabetes Metab Case Rep. 2015; 2015: 150007.
  3. Fukuhara N, Horiguchi K, Nishioka H, Suzuki H, Takeshita A, Inoshita N, Yamada S. Short-term preoperative octreotide treatment for TSH-secreting pituitary adenoma. Endocr J. 2015; 62: 21-27.

161a.  Tokutsu A, Okada Y, Torimoto K, Otsuka T, Narisawa M, Kurozumi A, Tanaka Y. Response of thyrotropin-secreting pituitary tumors to preoperative lanreotide therapy. Report of two cases. Neuro Endocrinol Lett. 2020; 41(1):10-16.

161b.   Shimatsu A, Nakamura A, Takahashi Y, Fujio S, Satoh F, Tahara S, Nishioka H, Takano K, Yamashita M, Arima H, Tominaga A, Tateishi S, Matsushita Y. Preoperative and long-term efficacy and safety of lanreotide autogel in patients with thyrotropin-secreting pituitary adenoma: a multicenter, single-arm, phase 3 study in Japan. Endocr J. 2021; 68(7):791-805.

161c.   van Eersel MEA, Meeuwisse-Pasterkamp SH, Muller Kobold AC, Meiners LC, den Dunnen WF, Hofland LJ, van den Berg G. Treatment of a thyrotropin-secreting pituitary adenoma (TSH-oma) with pasireotide LAR. Clin Endocrinol (Oxf). 2017; 87(6):877-87.

161d.   Illouz F, Chanson P, Sonnet E, Brue T, Ferriere A, Raffin Sanson ML, Vantyghem MC, Raverot G, Munier M, Rodien P, Briet C. Somatostatin receptor ligands induce TSH deficiency in thyrotropin-secreting pituitary adenoma. Eur J Endocrinol. 2021; 184(1):1-8.

  1. McCutcheon IE, Weintraub BD, Oldfield EH. Surgical treatment of thyrotropin-secreting pituitary adenomas, J Neurosurg 1990; 73: 674-683.
  2. Losa M, Mortini P, Franzin A. Surgical management of thyrotropin-secreting pituitary adenomas, Pituitary. 1999; 2: 127-131.
  3. Page KA, Roehmholdt BF, Jablonski M, Mayerson AB. Development of thyroid storm after surgical resection of a thyrotropin-secreting pituitary adenoma. Endocr Pract. 2008; 14: 732-737.
  4. Daousi C, Foy PM, MacFarlane IA. Ablative thyroid treatment for thyrotoxicosis due to thyrotropin-producing pituitary tumors. J Neurol Neurosurg Psychiatry. 2007; 78: 93-95.

165a.   Kim SH, Ku CR, Na M, Yoo J, Kim W, Jung IH, Kim KW, Moon JH, Kim D, Lee EJ, Kim SH, Kim EH. Immediate postoperative measurement of thyroid-stimulating hormone as an early predictor of remission in thyroid-stimulating hormone-secreting pituitary adenomas. J Neurosurg. 2020; 134(3):794-800.

  1. Mouslech Z, Somali M, Sakali AK, Savopoulos C, Mastorakos G, Hatzitolios AI. TSH-secreting pituitary adenomas treated by gamma knife radiosurgery: our case experience and a review of the literature. Hormones (Athens). 2016; 15: 122-128.
  2. Ohki M, Sato K, Tuchiya D, Sato S, Saito S, Kinjo T, Kayama T, Yokura H, Yoshimoto T. A case of TSH-secreting pituitary adenoma associated with an unruptured aneurysm: successful treatment by two-stage operation and gamma knife. No To Shinkei 1999; 51: 895-899.
  3. Mulinda JR, Hasinski S, Rose LI. Successful therapy for a mixed thyrotropin-and prolactin-secreting pituitary macroadenoma with cabergoline. Endocr Pract. 1999; 5: 76-79.
  4. Mouton F, Faivre-Defrance F, Cortet-Rudelli C, Assaker R, Soto-Ares G, Defoort-Dhellemmes S, Blond S, Wemeau JL, Vantyghem MC. TSH-secreting adenoma improved with cabergoline. Ann Endocrinol (Paris). 2008; 69: 244-248.
  5. Prieto-Tenreiro A, Díaz-Guardiola P. Long term treatment of a thyrotropin-secreting microadenoma with somatostatin analogues. Arq Bras Endocrinol Metabol. 2010; 54: 502-506.
  6. Gruszka A, Zielinski GM, Kunert-Radek J. Preoperative long-acting octreotide treatment for invasive thyrotropin-secreting pituitary macroadenoma after previous radioiodine thyroid ablation. J Clin Neurosci. 2014; 21: 340-342.
  7. Zhang CF, Liang D, Zhong LY. Efficacy of the long-acting octreotide formulation in patients with thyroid-stimulating hormone-secreting pituitary adenomas after incomplete surgery and octreotide treatment failure. Chin Med J (Engl). 2012; 125: 2758-2763.
  8. Fliers E, van Furth WR, Bisschop PH. Cure of a thyrotrophin (TSH)-secreting pituitary adenoma by medical therapy. Clin Endocrinol (Oxf). 2012 ; 77: 788-790.
  9. Neggers SJ, van der Lely AJ. Medical approach to pituitary tumors. Handb Clin Neurol. 2014; 124: 303-316.
  10. Gatto F, Grasso LF, Nazzari E, Cuny T, Anania P, Di Somma C, Colao A, Zona G, Weryha G, Pivonello R, Ferone D. Clinical outcome and evidence of high ratepost-surgical anterior hypopituitarism in a cohort of TSH-secreting adenoma patients: Might somatostatin analogues have a role as first-line therapy? Pituitary. 2015; 18: 583-591.
  11. Rimareix F, Grunenwald S, Vezzosi D, Rivière LD, Bennet A, Caron P. Primary medical treatment of thyrotropin-secreting pituitary adenomas by first-generation somatostatin analogues: a case study of seven patients. Thyroid. 2015; 25: 877-882.
  12. Araujo PB, Vieira Neto L, Gadelha MR. Pituitary tumor management in pregnancy. Endocrinol Metab Clin North Am. 2015; 44: 181-197.
  13. Bolz M, Körber S, Schober HC. TSH secreting adenoma of pituitary gland (TSHom) - rare cause of hyperthyroidism in pregnancy. Dtsch Med Wochenschr. 2013; 138: 362-366.
  14. Beck-Peccoz P, Persani L. Medical management of thyrotropin-secreting pituitary adenomas. Pituitary. 2002; 5: 83-88.
  15. Chanson P, Weintraub BD, Harris AG. Octreotide therapy for thyroid stimulating-secreting pituitary adenomas. A follow-up of 52 patients. Ann Intern Med. 1993; 119: 236-240.

Non-Functioning Pituitary Adenomas

ABSTRACT

 

Pituitary adenomas comprise approximately 10-20% of intracranial tumors. Non-functioning pituitary adenomas (NFPAs) are benign adenohypophyseal tumors not associated with clinical evidence of hormonal hypersecretion. NFPAs comprise different histological subtypes, classified according to their immunostaining to different adenohypophyseal hormones and transcription factors. The silent gonadotroph adenoma is the most common subtype, followed by corticotroph, PIT1 (POU1F1) gene lineage, and null cell tumors. Patients with NFPAs usually come to medical attention as a result of “mass effects” symptoms such as headaches, visual disorders, and/or cranial nerve dysfunction caused by lesions large enough to damage surrounding structures. Hypopituitarism, caused by the compression of the normal anterior pituitary, and hyperprolactinemia due to pituitary stalk deviation can also be present. Some cases may be diagnosed incidentally through imaging studies performed for other purposes. Patients with NFPAs should undergo hormonal, clinical, and laboratory evaluation to rule out hyper and hypopituitarism. Assessment of prolactin and IGF-1 levels have been recommended in all patients whereas screening for cortisol excess is suggested in the presence of clinical symptoms. Impairment of pituitary function should be assessed by baseline hormonal measurements and/or stimulatory tests, if needed. Patients in whom the tumor abuts the optic chiasm should be submitted to visual field perimetry. Surgical resection is the primary treatment for symptomatic patients with NFPAs, i.e., those with neuro-ophthalmologic complaints and/or tumors affecting the optic pathway. Visual deficits and, less commonly, hormone deficiencies may improve following surgical treatment although new hormone deficiencies may also occasionally develop after a surgical approach. For patients with residual NFPAs following transsphenoidal surgery, a therapeutic attempt using cabergoline can be made according to clinical judgment in individual cases. Radiotherapy in the postoperative period is not consensual and is generally reserved for cases of tumors not completely resected by surgery, those cases that present progressive tumor growth during follow-up, or for patients who, at diagnosis, already have tumors with aggressive features. Highly aggressive tumors need special care during follow-up, including temozolomide with or without radiotherapy complementation or new potential emerging treatments. For patients with asymptomatic NFPAs a “watch and wait” option is reasonable. Follow up is individualized and should consider tumor size, prior treatments, and clinical symptoms.

 

INTRODUCTION

 

Pituitary adenomas are common, predominantly indolent neoplasms comprising approximately 10-20% of intracranial tumors (1,2). Non-functioning pituitary adenomas (NFPAs) are benign neoplasms that originate from the adenohypophyseal cells and are not associated with clinical evidence of hormonal hypersecretion (3). They comprise a large and heterogeneous group, representing a sizeable proportion (22% to 54% in different series) of all pituitary adenomas (4-7). Malignant transformation in a non-functioning pituitary adenoma (pituitary carcinoma) is extremely rare and is characterized by craniospinal or systemic dissemination (8).

 

NFPAs can be further classified according to their pituitary hormone and transcription factor profile, as defined by the 2017 World Health Organization (WHO) classification for endocrine tumors (9). Tumors that express one or more anterior pituitary hormones or their transcription factors with immunohistochemistry (IHC) but do not secrete hormones at a clinically relevant level can be referred to as silent pituitary adenomas (SPAs) (10,11). As a consequence, the definition of “null cell adenoma” is now limited to an exceptionally rare primary adenohypophyseal tumor that shows immunonegativity for all adenohypophyseal hormones as well as for cell-type specific transcription factors (12) - Figure 1.

Figure 1 - Classification of non-functioning pituitary adenomas rely upon clinical features and histopathological data. Reprinted with permission from Drummond et al., 2019

The term “totally silent” has been proposed to be used when a patient with an NFPA presents basal and stimulated serum concentrations of the correspondent hormones within the normal range and there are no clinical signs or symptoms that can be attributed to hormone excess (13). The term “clinically silent” may be used when NFPAs secrete hormonal products that cause an elevation of the serum concentration but do not result in clinical signs or symptoms of hormonal hypersecretion (13). Some cases are referred to as “whispering” adenomas with borderline, mild, often overlooked, clinical symptoms and signs (14,15).

 

EPIDEMIOLOGY

 

The prevalence of NFPAs is variable and is often based upon autopsy or magnetic resonance imaging (MRI) series. Data from Europe, North and South America have estimated that the prevalence of clinically relevant NFPAs is 7–41.3 cases per 100,000 of population (16). This is likely an underestimate of the true prevalence, as many NFPAs go undiagnosed until they are large enough to cause mass effect or are accidentally discovered. Data are discordant about gender predominance and the peak occurrence is from the fourth to the eighth decade. A recent population-based study from South Korea showed an annual incidence of 3.5 cases/ 100,000 population for NFPAs, which is noticeably higher than the annual incidences previously reported in other countries, such as Sweden, Finland, and Argentina (17). These differences may be related to genetic and environmental factors in Asian populations or inter-study heterogeneity and may also reflect the good local access to diagnostic evaluations using magnetic resonance imaging and computed tomography.

 

CLINICAL PRESENTATION

 

The absence of clinical manifestations of hormonal hypersecretion usually results in significant diagnostic delay and therefore NFPAs may not be diagnosed until they cause mass effects to surrounding structures (3), causing symptoms such as headaches, visual disorders, and/or cranial nerve dysfunction. Other manifestations are hormone deficiencies or hyperprolactinemia due to pituitary stalk deviation and, less frequently, pituitary apoplexy (18,19). Additionally, some cases may be diagnosed incidentally through imaging studies performed for other purposes, the so-called pituitary incidentaloma.

 

Neurologic Manifestations

 

VISUAL IMPAIRMENT 

 

Impaired vision, caused by suprasellar extension of the adenoma that compresses the optic chiasm, is the most common neuro-ophthalmological symptom (19). Different types of visual defects depend on the degree and site of optic nerve compression. Both eyes are usually affected, although a significant proportion of patients may have unilateral or altitudinal problems in 33 and 16% of the cases, respectively (20). Diplopia, induced by oculomotor nerve compression resulting from parasellar expansion of the adenoma may occur, and the fourth, fifth and sixth cranial nerves may also be occasionally involved when there is parasellar expansion (16). Nevertheless, the typical visual field defect associated with pituitary tumors is bitemporal hemianopia, reported in approximately 40% of the patients.

 

HEADACHE

 

Headaches, the second most common neurologic symptom, occur in 19-75% of patients with pituitary tumors, regardless of size (21). In a retrospective case series of incidentally-discovered NFPAs, headache was present in approximately 20% of the cases (22). In a recent prospective series, 138 out of 269 patients with NFPAs complained of tumor-related symptoms and, among them, 23% presented with headaches (23). Although it is not always clear whether the presenting headache is related to the tumor, proposed mechanisms for headache include increased intrasellar pressure, stretching of dural membrane pain receptors, and activation of trigeminal pain pathways (16,24). Cerebrospinal fluid (CSF) rhinorrhea, associated or not with headache, can occur in cases where the tumor causes erosion of the sellar floor and extends inferiorly to the sphenoid sinus.

 

PITUITARY APOPLEXY

 

Pituitary apoplexy (sudden hemorrhage into a pituitary adenoma) is rare. According to a retrospective case series of 485 Mexican patients with NFPAs, pituitary apoplexy was the initial presentation in 8% of the cases (25). It causes acute onset of a severe headache associated with visual disturbances and can occur in all types of pituitary tumors, although some series suggest pituitary apoplexy might be more common in NFPAs than other adenomas subtypes (26). It has been suggested that the combination of the high metabolism of pituitary adenomas combined with their special blood supply would make them more prone to vascular events (27). Pituitary apoplexy may occur without an identified risk factor, but it has also been reported as being related to pregnancy, use of anticoagulants, surgical procedures, as well as in association with dynamic tests, such as thyrotropin-releasing hormone (TRH), gonadotropin-releasing hormone (GnRH), and insulin-tolerance stimulation tests (28).

 

The actual long-term risk of apoplexy in NFPAs has not been clearly defined (29). In a Japanese prospective cohort study of 42 asymptomatic patients with NFPAs (mean initial tumor size of 18.3 mm) followed-up for approximately four years, pituitary apoplexy was reported in 9.5% of the cases (30). A systematic review and meta-analysis evaluating the outcomes of patients with NFPAs and pituitary incidentalomas who were treated conservatively and followed-up for mean period of 3.9 years showed that the development of pituitary apoplexy was rare (0.2/100 patients-year), although a trend for a greater incidence of pituitary apoplexy was seen in macroadenomas compared with microadenomas, with a reported incidence of apoplexy of 1.1% per year (31). These findings are in line with two recent retrospective studies including a Korean series of 197 patients with NFPAs without mass effect symptoms or pituitary apoplexy at baseline who were followed-up for a median of 37 months (32) and an Italian series of 296 patients with incidentally discovered NFPAs (macroadenomas in 49% of the cases) who we followed-up for approximately three years (33). Five out of 169 patients with macroadenomas developed pituitary apoplexy with an overall incidence of 0.83 per 100 patients-years in the Korean series while no cases of pituitary apoplexy were reported in the Italian series.

 

Endocrine Manifestations

 

HORMONAL DEFICIENCIES

 

Most patients with nonfunctioning pituitary macroadenomas present with deficiency of at least one pituitary hormone resulting from the compression of the normal anterior pituitary and/or pituitary stalk, preventing the stimulation of pituitary cells by hypothalamic factors. Hypogonadism can result from either a direct compressive effect on gonadotropic cells or by stalk compression-induced hyperprolactinemia that inhibits the pulsatile secretion of gonadotropin releasing hormone via interfering with hypothalamic kisspeptin-secreting cells (34). This “disconnection hyperprolactinemia” usually <2000mIU/L (95 ng/mL) (35) is characterized by compression of the pituitary stalk, which prevents the arrival of dopamine to the anterior pituitary, the main inhibitor of prolactin (stalk effect). GH and gonadotroph axes are most commonly affected, followed by adrenal insufficiency and central hypothyroidism (16,23,32).

 

HORMONAL EXCESS

 

Gonadotroph adenomas are usually considered to be "nonfunctioning" although they can secrete intact gonadotropins, as they do not generally result in a clinical syndrome. Occasionally, gonadotroph adenomas secrete primarily FSH but also LH in quantities high enough to raise serum gonadotropin levels, which in turn, may lead to the development of some specific symptoms, such as ovarian hyperstimulation in young women (36-38) or, more rarely, precocious puberty or testicular enlargement in men. In addition, low serum LH:FSH ratios (usually < 1.0) have been described in clinically-secreting gonadotroph adenomas (39). Measurement of α-subunit may also contribute to a pre-operative diagnosis in clinically silent but biochemically-secreting NFPAs, as it may be the sole biochemical marker of the gonadotroph subtype in a number of cases. In addition, circulating FSH, LH and α-subunit levels can help the post-operative surveillance of these patients (39).

 

HISTOPATHOLOGICAL CLASSIFICATION

 

The 2017 WHO classification for endocrine tumors (9) defines pituitary adenomas, including NFPAs, according to their pituitary hormone and transcription factor profile. It is noteworthy that in this classification system, IHC forms the basis of the new categorization, in which an adenohypophyseal cell lineage designation of the pituitary adenoma has replaced the concept of a “hormone-producing pituitary adenoma” (40) -Table 1.

 

Table 1 – Classification of Silent Pituitary Adenomas According to Adenohypophyseal Hormones and Transcription Factors.

Cell Lineage

Hormone Staining

Transcription Factors and Other Co-Factors

 

Somatotroph adenoma

     Sparsely granulated

     Densely granulated

GH, α-subunit

Weak and patchy Diffuse and strong

PIT-1

 

Lactotroph adenomas

     Sparsely granulated

     Densely granulated

     Acidophil stem-cell adenoma

PRL

Perinuclear

Diffuse PRL

Focal and variable PRL, GH

PIT-1, ERα

 

Thyrotroph adenomas

TSHβ, α-subunit

PIT-1, GATA2

 

Corticotroph adenomas

    Densely granulated (type I)

    Sparsely granulated (type II)

    Crooke-cell

ACTH

Diffuse and strong ACTH

Weak and patchy ACTH

Periphery (ring-like)

TPIT

 

Gonadotroph adenomas

FSHβ, LHβ, α-subunit

SF1, GATA2, ERα

 

Null cell adenomas

None

None

 

Plurihormonal

     PIT-1-positive adenomas

     Adenomas with unusual IHC combinations

 

 

GH, PRL, TSHβ±, α-subunit

Various combinations

 

PIT-1

 

Adapted from Mete et al, 2017.

 

Indeed, the prevalence of the different histological subtypes of NFPAs is dependent on the extent of the IHC profile. According to a large retrospective case series (11), the gonadotroph adenoma is the most common subtype, followed by corticotroph, PIT1 (GH/ Prolactin/TSH) lineage and null cell - Figure 2.

 

Figure 2 - Prevalence of silent pituitary adenoma subtypes according to immunochemistry for anterior pituitary hormones and transcription factors. Adapted from Nishioka et al., 2015

Pituitary hormones gene expression analysis using real-time quantitative PCR (RT-qPCR) has been shown to complement the IHC classification of pituitary tumors, according to a large observational, cross-sectional study evaluating 268 patients (41). Addition of RT-qPCR analysis reduced the proportion of null-cell subtype in this series from 36% to 19%. Lower specific adenohypophyseal hormone gene expression was observed in some SPA variants when compared with their secreting counterparts, including somatotroph, lactotroph, mixed somatotroph-lactotroph and plurihormonal adenomas, which could contribute to the absence of endocrine manifestations.

 

The European Pituitary Pathology Group has recently proposed a standardized report for the diagnosis of pituitary adenomas which includes a multi-step approach, comprising the summary of clinical and neuroimaging features, IHC for hormones and transcription factors, assessment of proliferation and, when indicated, the use of markers predictive of treatment response (42). This proposition highlights the role of the pituitary pathologist as part of a multidisciplinary pituitary team helping the clinician define the most appropriate post-operative strategy for each patient, including appropriate follow-up and early recognition and treatment of potentially aggressive tumors (43).

 

Subtypes of NFPAs

 

NULL CELL ADENOMAS AND SILENT GONADOTROPH ADENOMAS

 

Null cell adenomas and silent gonadotroph adenomas (SGAs) were previously misunderstood to be the same type of tumor. The addition of immunostaining for steroidogenic factor 1 (SF1) as a tool in the diagnosis of pituitary lesions has shown that many LH/FSH immunonegative adenomas are in fact SGAs (44), which comprise almost 80% of resected NFPAs. The distinction between SGAs and null cell adenomas is of clinical relevance because true null cell adenomas are rare and likely to be more invasive and aggressive than SGAs (45). In addition, these rare cases of ‘null cell adenomas’ may require a broad panel of immunohistochemistry in order to exclude a sellar metastasis of a neuroendocrine tumor (NET) from another organ. Several markers, including TTF1, serotonin, ATRX, DAXX and CDX2 may be useful in the differential diagnosis between an NFPA and a metastatic NET in the sellar regionData from a retrospective case series of 516 patients with NFPAs have shown that from the 23% of the tumors initially classified as null cell adenomas by using only classical pituitary hormone IHC, only 5% of them remained as true null cell type. Indeed, immunostaining using lineage-specific markers, namely, PIT-1 (coded by the POU class 1 homeobox 1 (POU1F1) gene), SF1, TPIT (coded by the T-Box transcription factor 19 (TBX19) gene) and estrogen receptor-α (ERα) provided tumor reclassification in 95% of cases (11).

 

SILENT CORTICOTROPH ADENOMAS

 

Silent corticotroph adenomas (SCAs) are characterized by the absence of clinical features of Cushing syndrome, along with normal circadian cortisol secretion (totally silent) or elevated ACTH (clinically silent) (5,47-49). They currently account for approximately 15% of NFPAs, an underestimated proportion, since IHC for the transcription factor TPIT, a marker of corticotroph differentiation which regulates the proopiomelanocortin (POMC) lineage giving origin to the corticotrophs, is still not widely available (50). For instance, in a recent retrospective series, inclusion of IHC for TPIT allowed identification of eight SCAs out of 18 (44%) pituitary tumors previously classified as null cell adenomas (51).

 

SCAs often present themselves as macroadenomas associated with mass-related symptoms. In comparison with SGAs, SCAs show female preponderance, are more frequently giant adenomas, and are more often associated with marked cavernous sinus invasion (49). Importantly, the presence of multiple microcysts in T2-weighted pituitary MRI sequences in a NFPA has a high specificity (> 90%) for the corticotroph subtype. The first study showed that multiple microcysts were present in 76% (13/17) of SCAs as opposed to 5% (3/60) of SGAs (52) while a more recent study showed comparable results, as multiple microcysts were present in 58% (7/12) SCAs but in only 9.5% of SGAs (53).Histologically, SCAs can be further divided into type 1 (densely granulated), type 2 (sparsely granulated) and Crooke-cell adenoma. Type 1 SCAs show strong ACTH immunoreactivity whereas type 2 SCAs resemble the rare chromophobe corticotroph adenoma and show weak and focal ACTH immunoreactivity. Type 2 SCAs seem to be more common and are likely to display higher expression of migration and proliferation factors compared with type 1 SCAs (5,11,40). Clinically silent Crooke-cell adenoma is a rare yet highly aggressive subtype as it carries significant risk of morbidity (54). Since SCAs are nonfunctional, an important point to note is the absence of Crooke’s hyalinization in the normal surrounding pituitary gland, as there is no exposure to high circulating glucocorticoid levels to promote hyaline deposits of cytokeratin filaments in the cytoplasm of normal corticotrophs (49).

 

The transformation of a silent corticotroph tumor into Cushing disease has been described, although the mechanism involved in this phenomenon is not yet well understood (55,56). It has been proposed that the clinical manifestations of Cushing disease are dependent on the processing of the pro-hormone POMC in corticotrophs. The pro-hormone convertase 1/3 (PC1/3) is involved in the post-translational processing of POMC into mature and biologically active ACTH. SCAs show a decrease in PC1/3 expression associated with a down-regulation of PC1/3 genes compared with corticotroph adenomas associated with Cushing disease (47). An attractive and more likely hypothesis involves epigenetic mechanisms: DNA hypermethylation of regulatory regions could lead to reduced expression of POMCtranscripts impairing the production of secreted ACTH. Differences in methylation status were observed when comparing ACTH-secreting pituitary tumors and SCAs: the second promoter was highly methylated in SCAs, partially demethylated in normal pituitary tissue and highly demethylated in pituitary and ectopic ACTH-secreting tumors (57).

 

SILENT SOMATOTROPH ADENOMAS

 

Silent somatotroph adenomas are PIT1 and GH-immunoreactive tumors without clinical and biological signs of acromegaly. They represent approximately 2-4% of all pituitary adenomas in surgical case series (58). Patients with silent somatotroph adenomas usually present with normal pre-operative GH and IGF-1 levels but there have been few reports of "clinically silent" cases, with non-suppressible serum GH and elevated IGF-1 levels (59).

 

Similar to secreting somatotroph adenomas, silent somatotroph adenomas are classified into densely granulated and sparsely granulated types, based on the presence and pattern of the low molecular weight cytokeratin staining. More than 50% of silent somatotroph adenoma cases constitute mixed GH–PRL adenomas, a proportion twofold higher than somatotroph adenomas causing acromegaly (60). Unlike clinically functioning somatotroph tumors, the silent ones are more frequently sparsely granulated with more aggressive behavior and a lower response to somatostatin analog therapy (61). Furthermore, silent somatotroph adenomas are more frequent in females, present at a younger age, are larger, more invasive, and recur earlier and more frequently than their secreting counterparts (62).

 

SILENT THYROTROPH ADENOMAS

 

Silent thyrotroph adenomas usually present TSHβ, α-subunit, and PIT-1 expression on IHC in a variable manner (40). They are more frequent when compared with their functioning counterparts and seem to behave similarly regarding treatment outcomes and recurrence rates (63). In a recent retrospective series of 20 patients with silent thyrotroph adenomas who underwent transsphenoidal surgery, 95% were macroadenomas and 85% showed extrasellar growth. Gross total tumor resection was achieved in 45% while major tumor progression or recurrence occurred in 10% of the patients over a median follow-up period of 18.5 months (64). These results show that, although silent thyrotroph adenomas tend to be large and invasive tumors, good overall outcomes with low complication rates can be achieved.

 

SILENT LACTOTROPH ADENOMAS

 

Clinically silent lactotroph adenomas are rare. The positive prolactin staining by IHC with no clinical signs of hyperprolactinemia is usually encountered concomitantly with

GH positive staining (silent mixed somatotroph-lactotroph adenoma) (61). They can also express ERα on immunostaining (9).

 

PLURIHORMONAL ADENOMAS

 

According to the WHO 2017 classification, plurihormonal adenomas can be classified on the basis of their transcription factor expression into two groups: 1) “PIT1-positive adenomas” (previously known as silent subtype 3 pituitary adenoma); and 2) plurihormonal with more than one transcription factor, termed “plurihormonal adenomas with unusual immunohistochemical combinations” (PAWUC) (9,40). In a recent large retrospective series comprising 665 patients who underwent endoscopic endonasal transsphenoidal surgery, plurihormonal adenomas were identified in 4% of the cases (18 patients had PAWUC and 9 patients were diagnosed with PIT-1 positive adenomas); 24 (89%) were macroadenomas (including 6 giant adenomas) and cavernous sinus invasion was found in 12 patients (44%) (65).

 

PIT1-positive plurihormonal adenomas are a distinct entity, with reportedly aggressive behavior. A single-center retrospective case series reported a prevalence of 0.9% for ‘silent adenomas type 3’ among resected pituitary tumors over a period of 13 years. All tumors were macroadenomas, aggressive, invasive, with a high rate of persistent/recurrent disease (66). Interestingly, these tumors may present clinical symptoms of hormone excess, such as acromegaly, hyperthyroidism or marked hyperprolactinemia (67) and the diagnosis of a PIT-1 positive plurihormonal adenoma should be kept in mind in case of large and invasive NFPA tumors in young patients, particularly those showing TSH and often minor reactivities for PRL and GH as well as cytologic atypia in conjunction with an elevated Ki-67 labelling index (66).

 

PAWUC are also characterized by aggressive behavior and higher rates of cavernous sinus invasion. A recent single-center retrospective series comparing clinical characteristics, outcome parameters and rate of invasiveness of 22 PAWUC to 51 SGAs showed that patients with PAWUC were younger, were more likely to present with intraoperatively signs of tumor invasion and less likely to achieve gross-total resection than patients with SGAs, suggesting a more aggressive behavior of NFPAs if additional transcription factors other than SF1, GATA2/3 and ER α are expressed within the tumor cells (68).

 

EVALUATION

 

According to current practice guidelines on incidentally discovered sellar masses, all patients, including those without symptoms, should undergo hormonal, clinical, and laboratory evaluation for hyper and hypopituitarism (69). The extent of the evaluation is still debatable and has commonly relied on clinical experience - assessment of prolactin and IGF-1 levels have been recommended, whereas screening for cortisol excess (i.e., overnight 1mg dexamethasone and/or late-evening salivary cortisol and/or urinary-free cortisol) is suggested in the presence of clinical symptoms, while measurement of ACTH levels is not routinely recommended. (69).

 

Patients with macroadenoma, especially those >3 cm and with normal or slightly elevated prolactin, must have their serum prolactin diluted in order to exclude the "hook effect" (70,71). This effect occurs when excessively high levels of this hormone interfere with the formation of the complex antibody-antigen-antibody sandwich, and a prolactinoma might be mistaken for a NFPA (72).

 

It is also suggested to check for pituitary hormone deficiencies in patients with non-functioning tumors, irrespective of symptoms. Macroadenomas can cause impairment of pituitary function by the involvement of the normal gland or pituitary stalk compression, and the risk of hypopituitarism is directly related to tumor volume. Microadenomas eventually lead to pituitary dysfunction, particularly if larger than 5mm (32,69). A large retrospective cohort of 218 patients with NFPAs found at least one pituitary deficiency at diagnosis in 33.3% of microadenomas (73) although other studies have failed to identify hormonal deficiencies in microadenomas. An interesting and cost-effective approach for microincidentalomas could be no hormonal evaluation for cystic and solid lesions smaller than 5 mm. For solid microadenomas (between 6-9 mm), it is suggested a simple investigation with measurements of PRL and IGF1 (74).

 

Concerning macroadenomas, current guidelines suggest that serum cortisol levels at 8-9 AM should be done as the first-line test for diagnosing central adrenal insufficiency (75). Despite well recognized limitations of most commercial cortisol immunoassays (76), baseline cortisol levels <3 µg/dL (83 nmol/L) are suggestive of central adrenal insufficiency, while baseline cortisol levels >15 µg/dL (414 nmol/L) likely exclude it, and levels between 3 and 15 µg/dL require a corticotropin/insulin stimulation test. Central hypothyroidism is assessed by measuring serum TSH and free T4 (fT4), and fT4 level below the laboratory reference range in conjunction with a low, normal, or mildly elevated TSH in the setting of pituitary disease usually confirms the diagnosis (75). Diagnosing GH deficiency (GHD) is relatively straightforward in patients with IGF1 concentration lower than the gender- and age-specific lower limit of normal and structural pituitary disease. Low IGF1 in patients with non-functioning tumors who have at least three pituitary hormone deficiencies is also suggestive of GHD (29). Nevertheless, some adults with suspected GHD may have normal IGF1, in such cases GH stimulation testing may be useful. Central hypogonadism in males, manifests with low serum testosterone, low or inappropriately normal FSH/LH and features of testosterone deficiency. For females, pre-menopausal women with oligomenorrhea or amenorrhea should be screened with serum E2, FSH, and LH. In postmenopausal women, if the patient is not in hormonal therapy, the absence of high serum FSH and LH is sufficient for a diagnosis of central hypogonadism (75).

 

Sellar MRI with gadolinium is the best imaging study for suspected adenomas because it provides images of high resolution of the mass as well as its relationship with surrounding structures. Pituitary adenomas usually appear hypo or isointense compared to normal pituitary tissue in T1 images on MRI. In addition, while the normal pituitary tissue enhances earlier with contrast, pituitary adenomas commonly present with delayed contrast uptake (77). Patients whose tumor abuts the optic chiasm should have visual field perimetry, preferably by the Goldmann method, to assess for visual deficits (69).

 

Functional (FDG- and/or SSTR- positron emission tomography (PET) imaging studies should only be considered in aggressive pituitary adenomas/carcinomas in the setting of site-specific symptoms (neck/back pain or neurological complaints), and/or where laboratory measures are discordant with known visible extent of disease (78). Recently, 37 patients diagnosed with NFPAs enrolled in a clinical trial underwent 68Ga-DOTATATE PET/computed tomography (CT) of the head. 68Ga-DOTATATE uptake was positive in 34/37 patients (92%), demonstrating in vivo SSTR expression in NFPAs and ultimately showing superior sensitivity of PET imaging when compared with 111In-DTPA-octreotide scintigraphy (79).

 

The pre-operative differential diagnosis of NFPAs includes several additional primary non-hormonal-secreting lesions (80). This distinction is challenging in many cases because the clinical presentations and radiological aspects may be similar between adenomas and other pituitary lesions. The presence of diabetes insipidus (DI) is common in tumors of non-pituitary origin and indicates that the sellar mass is most likely not a pituitary adenoma (81). An increase in α subunit of the glycoprotein hormones (elevated in 30% of NFPA patients) can help in the differential diagnosis although normal values do not rule them out (82).

 

TREATMENT

                                   

In spite of current knowledge of definite NFPAs pathologic subtypes, initial treatment strategies are similar regardless of the subset. A small number of studies have reported treatment outcome results taking into consideration adenohypophyseal hormone immuno-profile (82-84) and, for the future, precise pathologic characterization utilizing adenohypophyseal hormones and transcription factors may potentially aid in predicting the response to specific adjunctive therapies.

 

Surgery

 

Surgical resection is the primary treatment for symptomatic patients with NFPAs (85), i.e., those with neuro-ophthalmologic complaints and/or tumors affecting the optic pathway. Surgery is also urgently indicated for patients with apoplexy who develop neuro-ophthalmologic complaints. In some experts’ opinion, tumors larger than 2cm should also be considered for surgery due to their propensity for growth (86). Treatment for hypopituitarism, primarily central adrenal insufficiency, and central hypothyroidism, should be commenced prior to surgical resection. Mortality rate is low (<1%) and reported surgical complication rates are acceptable. Postoperative complications such as CSF leakage, fistula, meningitis, vascular injury, persistent diabetes insipidus (DI), or new visual field defect occurred in ≤ 5% of patients, as reported by a systematic review and metanalysis (87). Accomplishment of total or near-total resection can be challenging and varies in different series, ranging from 20% to 80% (88,89). According to a recent large retrospective series evaluating 254 patients who underwent endoscopic endonasal surgery for a macroadenoma (including 72 patients with NFPAs), cavernous sinus invasion as assessed by the modified Knosp classification (90)effectively predicted surgical outcomes. Gross total resection was negatively correlated with tumor grade and success rates of gross-total resection were between 30% and 56% for tumors extending beyond the internal carotid artery and into the cavernous sinus compartments (grades 3A and 3B) (91).

 

A new “shape grading system” (Figure 3) has been recently proposed for predicting outcomes in patients with NFPAs operated by transsphenoidal surgery. In a retrospective single center study, 191 NFPAs were assessed according to different radiological growth patterns: spherical (Shape I), oval (Shape II), dumbbell (Shape III), mushroom (Shape IV), and polylobulated (Shape V) (92). Gross total resection was achieved in 53% of the patients, with decreasing likelihood of accomplishment in higher shape grades - shape I (82%), shape II (74%), shape III (24%), shape IV (17%) and shape 5 (0%). Furthermore, shape grades predicted resection rate better than Knosp grades. During a mean follow-up of 59 months, tumor recurrence or regrowth was observed in 6% and 12% of the patients, respectively. Higher shape grades also correlated with higher risk of tumor recurrence/regrowth as well as the need for reoperation and/or radiotherapy. Of note, SCAs more often grew as shape IV or V, while silent somatotroph adenomas were more often detected in the shape V group. Thus, the shape grading system seems to be a complimentary tool to better predict NFPA surgical outcomes, however, these findings need validation in other cohorts.

 

Figure 3 - The Shape grading system. Abbreviations: OC, optic chiasm; PS, pituitary stalk. From Berkaman et al Acta Neuropathologica 2021 (92)

Visual field deficits and, less commonly, hormone deficiencies may improve following surgical treatment although new hormone deficiencies may also occasionally develop after a surgical approach (87,93). The incidence of DI after endoscopic transsphenoidal surgery to remove NFPAs was recently investigated in 168 patients. Seventy-seven (45.8%) patients experienced postoperative DI and 10 (6.0%) patients suffered from permanent DI. A large cephalocaudal tumor diameter (cut off value-of 2.7 cm) was predictive of postoperative DI in such patients (94). Moreover, younger age and the absence or intrasellar location of the bright spot (as opposed to suprasellar location) on preoperative T1-weighted MRI were further factors predicting postoperative DI in a Japanese series of 333 patients with NFPAs undergoing transsphenoidal surgery (95).

 

Pituitary function should be reassessed one to three months after surgery and treatment of hypopituitarism introduced according to hormone deficiencies. Whether or not GH deficiency should be replaced requires a thoughtful and individualized evaluation of risks and benefits (96,97). Current data supports the absence of any stimulation of remnant or induction of recurrence by growth hormone replacement in patients with NFPAs solely treated by surgical removal (98). In a retrospective series, tumor regrowth occurred in 38/107 (36%) of non-GH treated subjects and in 8/23 (35%) of GH-treated subjects, followed up for a mean period of 6.8 years. The Cox regression analysis showed that after adjusting for sex and age at tumor diagnosis, cavernous sinus invasion at diagnosis, and type of tumor removal, GH treatment was not a significant independent predictor of recurrence.

 

A sellar MRI should be obtained three to six months after surgery to assess the extent of tumor resection. Also, as a significant number of patients with NFPAs may develop tumor re-growth, long-term imaging surveillance is recommended. In a retrospective analysis of 155 patients with NFPAs treated solely by surgery and followed-up for a mean period of six years (twenty-nine were followed up for more than 10 years), re-growth was reported in 34.8% of the cases, with 20% of relapse/re-growth occurring after 10 years (88). Likewise, in patients with NFPAs who present with classical apoplexy, tumor re-growth rate is not negligible. In a retrospective series of thirty-two patients with NFPAs who underwent surgery for pituitary apoplexy, tumor re-growth was reported in 11% of the cases at just over five years (99). Therefore, it is advisable that patients with NFPAs, particularly those with post-operative tumor remnants, be closely monitored following surgery, and follow-up surveillance needs to certainly be continued for more than 10 years.

 

Radiation Therapy

 

Radiation therapy (RT) has been shown to be effective as an adjunct to surgical resection in cases of post-operative residual tumor or recurrence. However, it carries a major long-term risk of hypopituitarism (85,100,101). Stereotactic techniques such as stereotactic radiosurgery or fractionated stereotactic radiotherapy have been developed with the purpose of delivering more localized irradiation and reducing long-term side-effects. Both techniques provide excellent tumor control in patients with NFPAs, ranging from 85% to 95% at five to ten years (102). However, at the present time, there is no consensus concerning the systematic use of RT in the postoperative period for patients with incompletely resected NFPAs (29) and whether an earlier approach would be preferable over conservative management. In general, RT is reserved for cases with large tumor remnants and for those cases that present progressive tumor growth during follow-up (85). Adjuvant RT may also be considered for patients who, at diagnosis, already present aggressive tumors, such as those invading parasellar structures or with extensive positive immunostaining for Ki-67, a proliferative index significantly associated with recurrence in NFPAs (9,103). Furthermore, NFPA subtype may be a relevant factor in the expected response to radiotherapy. A retrospective multicenter study demonstrated that overall tumor control rate after radiosurgery was lower in SCAs compared to other NFPA subtypes (104) suggesting that, in SCAs, an elevated margin dose may be considered in order to achieve a better chance of tumor control. In line with these findings, a recent and large retrospective surgical series showed a significantly lower progression-free survival in patients with SCAs compared to other NFPAs (24.5 vs 51.1 months) (105). Among the SCA cohort, progression was noted despite the use of adjuvant radiosurgery in one third of the patients. Notwithstanding, a recent systematic review and meta-analysis showed no evidence supporting higher recurrence rate after primary treatment of SCAs compared to other NFPAs (106), however the evaluated study samples included only a small number of patients who had been offered adjuvant radiotherapy.

Primary treatment of NFPAs with medical therapy is not currently recommended (15,85). Small series have demonstrated significant tumor volume stabilization under medical treatment in non-operated patients with NFPAs due to contraindications or refusal (110), whereas other studies have revealed tumor volume progression in the long term (111). The main medical agents that have been evaluated in NFPAs are dopamine agonists (DA) and somatostatin analogues (SAs), mainly in patients with residual tumor after transsphenoidal surgery. Figure 4 shows a suggested algorithm for the management of patients with NFPAs.

 

Figure 4 - Suggested algorithm for the management of patients with NFPAs. CBA: cabergoline; NFPA: non-functioning pituitary adenoma, MRI: magnetic resonance imaging; PRRT: peptide receptor radionuclide therapy RT: radiotherapy, *potential option, less evidenced-based approach; SSTR: somatostatin receptor

DOPAMINE AGONISTS

 

Dopamine receptor type 2 (D2R) expression has been demonstrated in patients with NFPAs. In a small series of 18 patients with hormone-negative NFPAs, two thirds of them (12/18) expressed D2R by real-time polymerase chain reaction. Patients who presented residual tumor (9/18) were treated with cabergoline up to 3mg per week. After 12 months of treatment, tumor shrinkage was observed in 56% of the patients and tumor reduction was significantly greater in those expressing D2R (112). A historical cohort analysis on the adjunctive role of DA in adult patients with GH and ACTH negative NFPAs showed favorable results (83). The treatment group consisted of patients who were either initiated on DA upon post-surgical residual tumor detection or when tumor growth was subsequently detected on follow-up, while the control group received no medication after surgical treatment. Tumor control was significantly superior in patients who were treated upon detection of post-operative residual tumor, compared to those who were treated after presenting tumor progression or the control group, 87% vs. 58% vs. 47%, respectively. The requirement for additional treatment (surgery and/or radiotherapy) during follow-up was significantly decreased from 47% to 16% with adjunctive DA therapy. In this series, there were no correlations between clinical response to DA and D2R expression, the isoform type or their expression levels.

 

A randomized open-label clinical trial was recently conducted aiming to compare cabergoline with non-intervention in 116 Brazilian patients with residual NFPA after transsphenoidal surgery followed-up for over two years. NFPAs were classified solely based on IHC for anterior pituitary hormones and included 59% hormone-negative adenomas and 34% silent gonadotroph adenomas. Silent corticotroph adenomas were excluded from this study. By the end of the study, residual tumor shrinkage was significantly more frequently observed in the medical-therapy group compared to patients in the control group (28.8% vs. 10.5%). Tumor response was not associated with D2R expression (113). Thus, although cabergoline has not been a consensual treatment for patients with NFPAs, a therapeutic attempt can be made according to clinical judgment in individual cases; particularly in patients with large extrasellar remnants after surgery and a high probability of tumor progression (114). On the other hand, most studies published so far included only patients with hormone-negative or silent gonadotroph adenomas, such that this proposition should not be extended to silent corticotroph adenomas or the other rarer silent tumors. It is also important to bear in mind the natural history of untreated NFPAs, which may show a spontaneous decrease in tumor volume in up to 30% of them(115).

 

Cabergoline doses are variable, but usually started at 0.5 mg weekly, increasing 0.5 mg each week until a maximum dose of 3.0 mg/week is reached. For these patients, tumor shrinkage is not the major target although it can occur in 38% of them; otherwise, prevention of tumor growth is the main treatment goal. Tumor progression can be assessed by a sellar MRI every six months for the first two years, and annually thereafter. Once stability is achieved without significant side effects, the drug can be maintained indefinitely, taking into account individual cost-benefits (82,116).

 

SOMATOSTATIN RECEPTOR LIGANDS 

 

The finding of somatostatin receptors (SSTRs) expression by NFPAs has raised the possibility that the use of somatostatin receptor ligands (SRLs) could be an effective treatment strategy (117,118). The SRL octreotide which binds with high affinity to SSTR2 was not effective in controlling tumor size or improving visual field in a small group of patients with NFPAs (119). A case-control study evaluated the results of long-acting octreotide in patients harboring post-surgical NFPA residues and it demonstrated tumor remnant stabilization in 81% (21/26) of patients in the treated group compared to 47% (6/13) of patients in the control group after a mean follow-up of 37 months (120). However, neither visual field nor pituitary function changed in any of the groups. This cohort of 39 NFPAs consisted of a heterogeneous group as IHC revealed positivity for pituitary hormones in 28 cases (20 of those showing positivity for FSH and/or LH) whereas the remaining 11 cases were negative for adenohypophyseal hormones. SSTR5 was the predominant SSTR expressed (84%), followed by SSTR3 (61%), while SSTR2 was expressed in 46% of the cases.

 

The expression of SSTRs and zinc finger protein 1 (ZAC1), a protein regulating apoptosis and cell cycle arrest, were assessed in a group of NFPAs (SGAs and hormone-negative adenomas), active somatotroph adenomas, and normal pituitary. SSTR2 and ZAC1 expression was reduced whereas SSTR3 expression was increased in SGAs compared to active somatotroph adenomas and normal pituitary (121). Likewise, other studies have suggested that SSTR3 is the predominant SSTR expressed in most NFPAs, both by IHC studies (118,122) and mRNA levels (11,122). However, a few other studies have demonstrated higher SSTR2 expression than SSTR3 or SSTR5 expression in SGAs and hormone-negative adenomas (103,123). Regarding corticotroph pituitary tumors, SSTR5 expression was significantly less frequently found in SCAs as compared to their secreting counterparts (2/23 vs. 24/39) (124). Furthermore, amongst the 23 SCAs, somatic ubiquitin-specific protease 8 (USP8) mutation was detected in only two tumors, in line with previous reports showing a significantly lower prevalence of USP8 mutations in SCAs compared to functioning corticotroph tumors (125,126).

 

There is an ongoing multicenter, randomized, double-blind, placebo-controlled trial (GALANT study) evaluating the effectiveness of first generation SRLs in patients with NFPAs and suprasellar extension, either surgery-naive or with a postoperative remnant. Forty-four patients with positive results on 68Ga-DOTATATE PET are being randomized to treatment with either the SRL lanreotide or placebo (127).

 

Pasireotide is a universal SRL with action on SSTR1, SSTR2, SSTR3 and STR5 subtypes and, therefore, seems more attractive than first-generation SRLs as an alternative medical treatment for NFPAs. A head-to-head comparison of octreotide and pasireotide in MENX-affected rat harboring a mutation I the gene encoding p27, an in vivo model of NFPAs, was recently performed. Pasireotide showed superior anti-tumor effect vs. octreotide, especially in females, which also showed higher SSTR3 expression (128). There are two phase 2 clinical trials evaluating the safety and efficacy of pasireotide for the treatment of NFPAs. Both trials have been recently completed but the results are not yet available. Passion 1 (NCT01283542) is an open-label single arm study evaluating tumor volume response to pasireotide in naive patients with NFPAs >1cm. Another phase 2 clinical trial (NCT01620138) is currently comparing the response of patients with NFPAs and surgical remnants to cabergoline vs. pasireotide. Notwithstanding, until further data are available, the use of SSRLs is not currently recommended for the treatment of patients with NFPAs.

 

Treatment of NFPAs with dopastatin, i.e., chimeric compound with dopamine and somatostatin agonist activity, has also been under investigation. BIM-23A760, a compound with potent agonist activity both at D2R and SSTR2 effectively inhibited cell proliferation in primary cultures of NFPAs (129). Interestingly, TBR-760 (formerly BIM-23A760)was recently tested in a mouse model of aggressive NFPA and resulted in nearly complete inhibition of tumor growth (130). A one year, randomized, double-blind, placebo-controlled phase 2 study of TBR-760 in adult patients with residual NFPAs > 1cm after transsphenoidal surgery (NCT04335357) is being conducted and is expected to be completed by 2023.

 

TEMOZOLOMIDE

 

Temozolomide (TMZ) was the first alkylating chemotherapeutic drug to show significant response rates in aggressive pituitary tumors (131). Notwithstanding, TMZ seems to be less effective in NFPAs compared to their secreting counterparts as 45% of 110 clinically functioning pituitary tumors showed regression on first-line TMZ while only 17% of 47 NFPAs did (132).

 

Responsiveness to TMZ is probably dependent on the immuno-expression of O (6)-methylguanine DNA methyltransferase (MGMT), a DNA repair protein that acts by removing the alkyl group and therefore is associated with TMZ resistance (133). Low immuno-expression of MGMT by pituitary tumors has been associated with higher response rates to TMZ (131). MGMT immuno-expression was assessed in a group of 45 NFPAs and the degree of expression was correlated with tumor aggressiveness (133). Low MGMT expression was observed in 50% of the aggressive NFPAs compared to 24% of the non-aggressive NFPAs. These findings suggest that aggressive NFPAs with low MGMT expression could be potential candidates for treatment with TMZ.

 

Regarding dosing regimens and indications, current guidelines suggest using TMZ (150-200 mg/m2daily for 5/28 days) as an alternative treatment for patients with aggressive NFPAs presenting tumor progression despite radiotherapy and other therapeutic measures, and for exceptional cases of pituitary carcinomas (29,132). Furthermore, in the case of rapid tumor growth in patients who have not previously reached maximal doses of radiotherapy, the use of the STUPP protocol (six weeks of concomitant fractionated radiotherapy and TMZ 75 mg/m2 daily, followed by TMZ alone 150-200 mg/m2 daily for 5/28 days) has been suggested (132). Studies on long-term administration of TMZ are scarce but clinical observations indicate that TMZ should be continued for as long as it is effective and well tolerated. Some authors suggest continuing TMZ at standard dosage for two years and later reducing to half-dose (134). Patients receiving TMZ chemotherapy are at risk for hematologic toxicity, occurring in approximately 15-20% of the cases. The most common non-hematologic side effects of TMZ are nausea, anorexia, and fatigue (135).

 

PEPTIDE RECEPTOR RADIONUCLIDE THERAPY

 

In vivo SSTR expression by NFPAs has been previously demonstrated both by positive uptake on somatostatin receptor scintigraphy (120) and on 68Gallium DOTATATE PET/ CT (79) providing a rationale for the administration of PRRT in these patients. So far, PRRT has been studied in few patients with aggressive pituitary tumors (clinically functioning and NFPAs) with different response patterns (136,137). Among the six patients with nonfunctioning pituitary adenoma or carcinoma, three patients (not previously treated with TMZ) showed stable disease (138-140), two patients had progressive disease (138,141) and one patient died in the following months while information on tumor volume was lacking (142).

 

QUALITY OF LIFE AND LONG-TERM MORBIDITY AND MORTALITY

 

A substantial number of patients with NFPAs suffer from morbidities related to the tumor itself, as well as to the treatments offered. Standardized mortality ratios in these patients seem to be higher than that of the general population with deaths associated mainly with circulatory, respiratory, and infectious causes (143). Until now, there was no consensus on predictive factors of mortality but those most consistently described are older age at diagnosis (144,145) and high doses of glucocorticoid replacement therapy (146). A retrospective series of 546 patients operated on for a macro NFPA between 1963 and 2011 and followed up for a median period of 8 years reported a standardized mortality ratio of 3.6 (95% CI, 2.9–4.5) (144). After adjustment for factors proven to be significant in univariate analysis - radiotherapy, tumor regrowth, and untreated growth hormone deficiency - age at diagnosis was an independent predictor of mortality, with shorter survival observed in older patients.

 

Following NFPA treatment, patient-reported health-related quality of life (HR-QoL) substantially improves.evertheless, there are conflicting findings about HR-QoL normalization, which may also be related to the lack of a disease-specific HR-QoL questionnaire for NFPAs (147). Some studies have described a persistent decreased HR-QoL compared to healthy controls and reference data (148,149), while others have not (150). In the latest study, HR-QoL was evaluated in 193 consecutive patients with NFPAs followed up in a tertiary endocrine referral center (150). The overall health-related quality of life and perception of subjective health in patients with NFPAs was not compromised although specific groups, such as females, patients with tumor recurrences, and with visual defects, were shown to be affected in various dimensions. Altered sleep-wake rhythmicity has also been described in a cohort of 69 patients with NFPA in long term remission after transsphenoidal surgery on stable replacement treatment for hypopituitarism (151). NFPA patients reported severely impaired QoL, sleep quality, and increased daytime sleepiness. Preoperative visual field defects were associated with sleep-wake rhythm fragmentation and vasopressin deficiency was associated with decreased sleep efficiency, independent of age, hypopituitarism, or radiotherapy.

 

More recently, the use of the Wilson–Cleary model, a conceptual biopsychosocial model of HR-QoL, suggested that elements at each stage of this model could be contributing to the impairment in HR-QoL observed in patients with a NFPA (147). The authors concluded that currently available biomedical treatments, i.e., surgery, radiotherapy, and hormone replacement therapy, are clearly not sufficient for achievement of good HR-QoL in patients with a NFPA, and further improvement should be supported by a pituitary specific care trajectory, targeting not only biological and physiological variables, but also psychosocial care.

 

PROGNOSTIC FACTORS AND FUTURE PERSPECTIVES

 

There has been a search to identify reliable factors related to aggressiveness and the risk of recurrence in NFPAs. A single-center retrospective study which evaluated 108 surgically-resected NFPAs followed for up to 15 years (152)showed that 22% of the patients required further treatment, either second surgery or radiotherapy. Factors determining recurrence were the presence of residual tumor, tumor growth rate (>80 mm3/year), and suprasellar extension (152). According to another retrospective case series evaluating patients with NFPAs who presented tumor regrowth after primary treatment, the NFPA subtype, categorized by anterior pituitary hormone immunostaining, was not a predictive factor for the requirement of secondary treatment or tumor regrowth. Significant risk factors were female gender and treatment approach (monitoring vs. interventions); secondary progression was significantly higher in those patients who were followed conservatively (63%) as compared to those who received surgery (36%), radiotherapy (13%), and surgery/adjuvant radiotherapy (13%) (84). In patients with SGAs, ERα seems to be a prognostic factor for re-intervention (reoperation or radiation) in males - the combination of the absence of ERα expression and young age served as good predictive markers of aggressiveness (122).

 

The role of cellular markers, associated with cell proliferation and apoptosis, in predicting the recurrence of NFPAs has also been investigated (153). Proliferative indexes such as a high Ki-67 index, assessed by IHC, were significantly associated with a tumor size greater than 3 cm, as well as with tumor recurrence (103). Evaluation of tumor proliferation by using Ki-67 IHC is widely available and is recommended as part of the assessment of NFPAs (40). According to a large retrospective series evaluating 601 patients with surgically resected pituitary tumors, including approximately 30% NFPAs, the optimal cut-off points of the Ki-67 proliferation index that predicted recurrence was 2.5% with 84.6% sensitivity and 47.4% specificity (154). Moreover, a recent retrospective analysis of 120 patients operated on for an NFPA showed that invasive and proliferative tumors, i.e., grade 2b tumors. according to the clinicopathological classification of Trouillas et al. (155) showed an overall likelihood of recurrence that was approximately 9 times greater than those of grade 1a, i.e., non-invasive and non-proliferative tumors (156). Further risk factors associated with an increased risk of recurrence in this cohort were a younger age and the presence of residual tumor.

 

Minichromosome maintenance 7 (MCM7), a cell-cycle regulator protein, has been recently proposed as a marker of tumor progression in NFPAs. In a cohort study of 97 surgically treated NFPAs, the probability for reintervention within 6 years for patients harboring residual tumors with high MCM7 expression was 93% (157). Ki-67 expression >3%, age ≤55 years and mitotic index≥1, but not tumor subtype, were also associated with reintervention. Attempts to develop clinical nomograms to predict post-operative recurrence of NFPAs have recently been demonstrated based on age, tumor size, cavernous sinus invasion, sphenoid sinus invasion, and surgery extension. The nomogram model proposed by Lyu et al. (158) showed an area under the ROC curve of 0.953 and correlation analyses indicated that sphenoid sinus invasion, cavernous sinus invasion and tumor size could promote the recurrence of NFPA while advanced age and gross total resection could effectively inhibit it.

 

Genetic and epigenetic mechanisms underlying the development and aggressiveness of NFPAs have not been fully elucidated (159). The mutational landscape of a cohort of pituitary tumors including 37 NFPAs was recently published (160). Aside from demonstrating gonadotroph signatures in seven out of eight SCAs, retrieving the question whether a subset of SCAs arise from a specific pituitary lineage distinct from other corticotroph (161), this study showed that most NFPAs displayed no functional somatic variant and no chromosome alterations. Further characterization of these tumors with techniques such as whole genome sequencing coupled to chromatin structure analysis may disclose mutations in non-coding regions affecting chromatin opening and/or the binding of specific transcription factors. allowing for the development of novel therapeutic strategies.

 

An adverse pituitary adenoma phenotype is defined not only by the intrinsic activity of tumor cells but also by the infiltrated immune cells in the tumor microenvironment (162). The study of the immune profile of pituitary tumors for predicting immunotherapy responsiveness is an evolving field. Wang et al (163) have recently proposed classification of these tumors on three clusters based on tumor-infiltrating immune cells and the expression of immune checkpoint molecules This study cohort included 57 “unspecified“ NFPAs since they were not pathologically classified with pituitary transcription factors. The majority of them exhibited a “hot” immune microenvironment and were predicted to exhibit higher immunotherapy responsiveness.

 

CONCLUSION

 

NFPAs are frequent in endocrine practice. Clinically they range from being completely asymptomatic (incidental findings on head MRI or computed tomography scans) to causing significant hypothalamic/pituitary dysfunction and visual symptoms due to mass effect., For microadenomas and asymptomatic/relatively small macroadenomas (1–2 cm), a “watch and wait” option is reasonable. If tumor growth, development of visual field defects, or progressive pituitary dysfunction is detected during follow up, then surgery is indicated. It is now recommended that NFPAs are classified according to their pituitary hormone and transcription factor profiles along with proliferation markers such as Ki-67. This refined stratification may potentially aid in predicting disease course and in selecting adjunctive therapies. Radiotherapy is an effective treatment, although usually reserved for those patients with aggressive tumors and significant tumor remnant after pituitary surgery as considerable side effects may occur. Medical treatment with dopamine agonists stands as an alternative in selected cases, despite the lack of placebo-controlled trials. Highly aggressive tumors need special care during follow-up, including treatment with TMZ with or without RT complementation or new potential emerging treatments. Close collaboration of a multidisciplinary pituitary team is crucial to better serve this challenging group of patients.

 

REFERENCES

 

  1. Daly AF, Rixhon M, Adam C, Dempegioti A, Tichomirowa MA, Beckers A. High prevalence of pituitary adenomas: a cross-sectional study in the province of Liege, Belgium. J Clin Endocrinol Metab. 2006;91(12):4769-4775.
  2. Ho K, Fleseriu M, Kaiser U, Salvatori R, Brue T, Lopes MB, Kunz P, Molitch M, Camper SA, Gadelha M, Syro LV, Laws E, Reincke M, Nishioka H, Grossman A, Barkan A, Casanueva F, Wass J, Mamelak A, Katznelson L, van der Lely AJ, Radovick S, Bidlingmaier M, Boguszewski M, Bollerslev J, Hoffman AR, Oyesiku N, Raverot G, Ben-Shlomo A, Fowkes R, Shimon I, Fukuoka H, Pereira AM, Greenman Y, Heaney AP, Gurnell M, Johannsson G, Osamura RY, Buchfelder M, Zatelli MC, Korbonits M, Chanson P, Biermasz N, Clemmons DR, Karavitaki N, Bronstein MD, Trainer P, Melmed S. Pituitary Neoplasm Nomenclature Workshop: Does Adenoma Stand the Test of Time? J Endocr Soc. 2021;5(3):bvaa205.
  3. Fleseriu M, Karavitaki N. Non-functioning pituitary adenomas, not all the same and certainly not boring! Pituitary. 2018;21(2):109-110.
  4. Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf). 2010;72(3):377-382.
  5. Horvath E, Kovacs K, Killinger DW, Smyth HS, Platts ME, Singer W. Silent corticotropic adenomas of the human pituitary gland: a histologic, immunocytologic, and ultrastructural study. Am J Pathol. 1980;98(3):617-638.
  6. Aflorei ED, Korbonits M. Epidemiology and etiopathogenesis of pituitary adenomas. J Neurooncol. 2014;117(3):379-394.
  7. Tjornstrand A, Gunnarsson K, Evert M, Holmberg E, Ragnarsson O, Rosen T, Filipsson Nystrom H. The incidence rate of pituitary adenomas in western Sweden for the period 2001-2011. Eur J Endocrinol. 2014;171(4):519-526.
  8. Lenders N, McCormack A. Malignant transformation in non-functioning pituitary adenomas (pituitary carcinoma). Pituitary. 2018;21(2):217-229.
  9. Lloyd R, Osamura, R., Klöppel, G. & Rosai, J (eds). World Health Organization Classification of Tumours of Endocrine Organs. Vol 10. 4th ed: IARC Publication; 2017.
  10. Saeger W, Ludecke DK, Buchfelder M, Fahlbusch R, Quabbe HJ, Petersenn S. Pathohistological classification of pituitary tumors: 10 years of experience with the German Pituitary Tumor Registry. Eur J Endocrinol. 2007;156(2):203-216.
  11. Nishioka H, Inoshita N, Mete O, Asa SL, Hayashi K, Takeshita A, Fukuhara N, Yamaguchi-Okada M, Takeuchi Y, Yamada S. The Complementary Role of Transcription Factors in the Accurate Diagnosis of Clinically Nonfunctioning Pituitary Adenomas. Endocr Pathol. 2015;26(4):349-355.
  12. Lopes MBS. Classification, Pathobiology, Molecular Markers, and Intraoperative Pathology. In: Laws JE, Cohen-Gadol A., Schwartz T., Sheehan J., ed. Transsphenoidal Surgery: Springer, Cham; 2017:113-143.
  13. Mayson SE, Snyder PJ. Silent (clinically nonfunctioning) pituitary adenomas. J Neurooncol. 2014;117(3):429-436.
  14. Grossman AB. The 2004 World Health Organization classification of pituitary tumors: is it clinically helpful? Acta Neuropathol. 2006;111(1):76-77.
  15. Drummond J, Roncaroli F, Grossman AB, Korbonits M. Clinical and Pathological Aspects of Silent Pituitary Adenomas. J Clin Endocrinol Metab. 2019;104(7):2473-2489.
  16. Ntali G, Wass JA. Epidemiology, clinical presentation and diagnosis of non-functioning pituitary adenomas. Pituitary. 2018;21(2):111-118.
  17. Oh JS, Kim HJ, Hann HJ, Kang TU, Kim DS, Kang MJ, Lee JY, Shim JJ, Lee MR, Ahn HS. Incidence, mortality, and cardiovascular diseases in pituitary adenoma in Korea: a nationwide population-based study. Pituitary. 2021;24(1):38-47.
  18. Chen L, White WL, Spetzler RF, Xu B. A prospective study of nonfunctioning pituitary adenomas: presentation, management, and clinical outcome. J Neurooncol. 2011;102(1):129-138.
  19. Ferrante E, Ferraroni M, Castrignano T, Menicatti L, Anagni M, Reimondo G, Del Monte P, Bernasconi D, Loli P, Faustini-Fustini M, Borretta G, Terzolo M, Losa M, Morabito A, Spada A, Beck-Peccoz P, Lania AG. Non-functioning pituitary adenoma database: a useful resource to improve the clinical management of pituitary tumors. Eur J Endocrinol. 2006;155(6):823-829.
  20. Ogra S, Nichols AD, Stylli S, Kaye AH, Savino PJ, Danesh-Meyer HV. Visual acuity and pattern of visual field loss at presentation in pituitary adenoma. J Clin Neurosci. 2014;21(5):735-740.
  21. Rizzoli P, Iuliano S, Weizenbaum E, Laws E. Headache in Patients With Pituitary Lesions: A Longitudinal Cohort Study. Neurosurgery. 2016;78(3):316-323.
  22. Losa M, Donofrio CA, Barzaghi R, Mortini P. Presentation and surgical results of incidentally discovered nonfunctioning pituitary adenomas: evidence for a better outcome independently of other patients' characteristics. Eur J Endocrinol. 2013;169(6):735-742.
  23. Freda PU, Bruce JN, Khandji AG, Jin Z, Hickman RA, Frey E, Reyes-Vidal C, Otten M, Wardlaw SL, Post KD. Presenting Features in 269 Patients With Clinically Nonfunctioning Pituitary Adenomas Enrolled in a Prospective Study. J Endocr Soc. 2020;4(4):bvaa021.
  24. Greenman Y, Melmed S. Diagnosis and management of nonfunctioning pituitary tumors. Annu Rev Med. 1996;47:95-106.
  25. Vargas G, Gonzalez B, Ramirez C, Ferreira A, Espinosa E, Mendoza V, Guinto G, Lopez-Felix B, Zepeda E, Mercado M. Clinical characteristics and treatment outcome of 485 patients with nonfunctioning pituitary macroadenomas. Int J Endocrinol. 2015;2015:756069.
  26. Briet C, Salenave S, Chanson P. Pituitary apoplexy. Endocrinol Metab Clin North Am. 2015;44(1):199-209.
  27. Oldfield EH, Merrill MJ. Apoplexy of pituitary adenomas: the perfect storm. J Neurosurg. 2015;122(6):1444-1449.
  28. Wildemberg LE, Glezer A, Bronstein MD, Gadelha MR. Apoplexy in nonfunctioning pituitary adenomas. Pituitary. 2018;21(2):138-144.
  29. Vieira LN, Boguszewski CL, Araujo LA, Bronstein MD, Miranda PA, Musolino NR, Naves LA, Vilar L, Ribeiro-Oliveira AJ, Gadelha MR. A review on the diagnosis and treatment of patients with clinically nonfunctioning pituitary adenoma by the Neuroendocrinology Department of the Brazilian Society of Endocrinology and Metabolism. Archives of endocrinology and metabolism. 2016;60(4):374-390.
  30. Arita K, Tominaga A, Sugiyama K, Eguchi K, Iida K, Sumida M, Migita K, Kurisu K. Natural course of incidentally found nonfunctioning pituitary adenoma, with special reference to pituitary apoplexy during follow-up examination. J Neurosurg. 2006;104(6):884-891.
  31. Fernandez-Balsells MM, Murad MH, Barwise A, Gallegos-Orozco JF, Paul A, Lane MA, Lampropulos JF, Natividad I, Perestelo-Perez L, Ponce de Leon-Lovaton PG, Erwin PJ, Carey J, Montori VM. Natural history of nonfunctioning pituitary adenomas and incidentalomas: a systematic review and metaanalysis. J Clin Endocrinol Metab. 2011;96(4):905-912.
  32. Kim JH, Dho YS, Kim YH, Lee JH, Lee JH, Hong AR, Shin CS. Developing an optimal follow-up strategy based on the natural history of nonfunctioning pituitary adenomas. J Neurosurg. 2018;131(2):500-506.
  33. Tresoldi AS, Carosi G, Betella N, Del Sindaco G, Indirli R, Ferrante E, Sala E, Giavoli C, Morenghi E, Locatelli M, Milani D, Mazziotti G, Spada A, Arosio M, Mantovani G, Lania AGA. Clinically Nonfunctioning Pituitary Incidentalomas: Characteristics and Natural History. Neuroendocrinology. 2020;110(7-8):595-603.
  34. Sonigo C, Bouilly J, Carre N, Tolle V, Caraty A, Tello J, Simony-Conesa FJ, Millar R, Young J, Binart N. Hyperprolactinemia-induced ovarian acyclicity is reversed by kisspeptin administration. J Clin Invest. 2012;122(10):3791-3795.
  35. Karavitaki N, Thanabalasingham G, Shore HC, Trifanescu R, Ansorge O, Meston N, Turner HE, Wass JA. Do the limits of serum prolactin in disconnection hyperprolactinaemia need re-definition? A study of 226 patients with histologically verified non-functioning pituitary macroadenoma. Clin Endocrinol (Oxf). 2006;65(4):524-529.
  36. Gryngarten MG, Braslavsky D, Ballerini MG, Ledesma J, Ropelato MG, Escobar ME. Spontaneous ovarian hyperstimulation syndrome caused by a follicle-stimulating hormone-secreting pituitary macroadenoma in an early pubertal girl. Horm Res Paediatr. 2010;73(4):293-298.
  37. Graillon T, Castinetti F, Chabert-Orsini V, Morange I, Cuny T, Albarel F, Brue T, Dufour H. Functioning gonadotroph adenoma with severe ovarian hyperstimulation syndrome: A new emergency in pituitary adenoma surgery? Surgical considerations and literature review. Ann Endocrinol (Paris). 2019;80(2):122-127.
  38. Broughton C, Mears J, Williams A, Lonnen K. A clinically functioning gonadotroph adenoma presenting with abdominal pain, ovarian hyperstimulation and fibromatosis. Endocrinol Diabetes Metab Case Rep. 2018;2018.
  39. Takeda M, Otsuka F, Suzuki J, Kishida M, Ogura T, Tamiya T, Makino H. Involvement of activin/BMP system in development of human pituitary gonadotropinomas and nonfunctioning adenomas. Biochem Biophys Res Commun. 2003;306(4):812-818.
  40. Mete O, Lopes MB. Overview of the 2017 WHO Classification of Pituitary Tumors. Endocr Pathol. 2017;28(3):228-243.
  41. Torregrosa-Quesada ME, García-Martínez A, Sánchez-Barbie A, Silva-Ortega S, Cámara R, Fajardo C, Lamas C, Aranda I, Pico A. The silent variants of pituitary tumors: demographic, radiological and molecular characteristics. J Endocrinol Invest. 2021; 44(8):1637-1648.
  42. Villa C, Vasiljevic A, Jaffrain-Rea ML, Ansorge O, Asioli S, Barresi V, Chinezu L, Gardiman MP, Lania A, Lapshina AM, Poliani L, Reiniger L, Righi A, Saeger W, Soukup J, Theodoropoulou M, Uccella S, Trouillas J, Roncaroli F. A standardised diagnostic approach to pituitary neuroendocrine tumours (PitNETs): a European Pituitary Pathology Group (EPPG) proposal. Virchows Arch. 2019;475(6):687-692.
  43. Trouillas J, Jaffrain-Rea ML, Vasiljevic A, Raverot G, Roncaroli F, Villa C. How to Classify the Pituitary Neuroendocrine Tumors (PitNET)s in 2020. Cancers (Basel). 2020;12(2).

44      Gomez-Hernandez K, Ezzat S, Asa SL, Mete O. Clinical Implications of Accurate Subtyping of Pituitary Adenomas: Perspectives from the Treating Physician. Turk Patoloji Derg. 2015;31 Suppl 1:4-17.

  1. Balogun JA, Monsalves E, Juraschka K, Parvez K, Kucharczyk W, Mete O, Gentili F, Zadeh G. Null cell adenomas of the pituitary gland: an institutional review of their clinical imaging and behavioral characteristics. Endocr Pathol. 2015;26(1):63-70.
  2. Manojlovic-Gacic E, Bollerslev J, Casar-Borota O. Invited Review: Pathology of pituitary neuroendocrine tumours: present status, modern diagnostic approach, controversies and future perspectives from a neuropathological and clinical standpoint. Neuropathol Appl Neurobiol. 2020;46(2):89-110.
  3. Raverot G, Wierinckx A, Jouanneau E, Auger C, Borson-Chazot F, Lachuer J, Pugeat M, Trouillas J. Clinical, hormonal and molecular characterization of pituitary ACTH adenomas without (silent corticotroph adenomas) and with Cushing's disease. Eur J Endocrinol. 2010;163(1):35-43.
  4. Righi A, Faustini-Fustini M, Morandi L, Monti V, Asioli S, Mazzatenta D, Bacci A, Foschini MP. The changing faces of corticotroph cell adenomas: the role of prohormone convertase 1/3. Endocrine. 2017;56(2):286-297.
  5. Ben-Shlomo A, Cooper O. Silent corticotroph adenomas. Pituitary. 2018;21(2):183-193.
  6. Sjostedt E, Bollerslev J, Mulder J, Lindskog C, Ponten F, Casar-Borota O. A specific antibody to detect transcription factor T-Pit: a reliable marker of corticotroph cell differentiation and a tool to improve the classification of pituitary neuroendocrine tumours. Acta Neuropathol. 2017;134(4):675-677.
  7. McDonald WC, McDonald KN, Helmer JA, Ho B, Wang A, Banerji N. The Role of T-box Transcription Factor in a Pituitary Adenoma Diagnostic Algorithm. Arch Pathol Lab Med. 2021;145(5):592-598.
  8. Cazabat L, Dupuy M, Boulin A, Bernier M, Baussart B, Foubert L, Raffin-Sanson ML, Caron P, Bertherat J, Gaillard S. Silent, but not unseen: multimicrocystic aspect on T2-weighted MRI in silent corticotroph adenomas. Clin Endocrinol (Oxf). 2014;81(4):566-572.
  9. Kasuki L, Antunes X, Coelho MCA, Lamback EB, Galvao S, Silva Camacho AH, Chimelli L, Ventura N, Gadelha MR. Accuracy of microcystic aspect on T2-weighted MRI for the diagnosis of silent corticotroph adenomas. Clin Endocrinol (Oxf). 2020;92(2):145-149.
  10. Di Ieva A, Davidson JM, Syro LV, Rotondo F, Montoya JF, Horvath E, Cusimano MD, Kovacs K. Crooke's cell tumors of the pituitary. Neurosurgery. 2015;76(5):616-622.
  11. Tateno T, Izumiyama H, Doi M, Yoshimoto T, Shichiri M, Inoshita N, Oyama K, Yamada S, Hirata Y. Differential gene expression in ACTH -secreting and non-functioning pituitary tumors. Eur J Endocrinol. 2007;157(6):717-724.
  12. Zheng G, Lu L, Zhu H, You H, Feng M, Liu X, Dai C, Yao Y, Wang R, Zhang H, Sun X, Lu Z. Clinical, Laboratory, and Treatment Profiles of Silent Corticotroph Adenomas That Have Transformed to the Functional Type: A Case Series With a Literature Review. Front Endocrinol (Lausanne). 2020;11:558593.
  13. Araki T, Tone Y, Yamamoto M, Kameda H, Ben-Shlomo A, Yamada S, Takeshita A, Yamamoto M, Kawakami Y, Tone M, Melmed S. Two Distinctive POMC Promoters Modify Gene Expression in Cushing Disease. J Clin Endocrinol Metab. 2021;106(9):e3346-e3363.
  14. Chinezu L, Vasiljevic A, Trouillas J, Lapoirie M, Jouanneau E, Raverot G. Silent somatotroph tumour revisited from a study of 80 patients with and without acromegaly and a review of the literature. Eur J Endocrinol. 2017;176(2):195-201.
  15. Sidhaye A, Burger P, Rigamonti D, Salvatori R. Giant somatotrophinoma without acromegalic features: more "quiet" than "silent": case report. Neurosurgery. 2005;56(5):E1154; discussion E1154.
  16. Langlois F, Woltjer R, Cetas JS, Fleseriu M. Silent somatotroph pituitary adenomas: an update. Pituitary. 2018;21(2):194-202.
  17. Langlois F, Lim DST, Yedinak CG, Cetas I, McCartney S, Cetas J, Dogan A, Fleseriu M. Predictors of silent corticotroph adenoma recurrence; a large retrospective single center study and systematic literature review. Pituitary. 2018;21(1):32-40.
  18. Naritaka H, Kameya T, Sato Y, Furuhata S, Otani M, Kawase T. Morphological characterization and subtyping of silent somatotroph adenomas. Pituitary. 1999;1(3-4):233-241.
  19. Kirkman MA, Jaunmuktane Z, Brandner S, Khan AA, Powell M, Baldeweg SE. Active and silent thyroid-stimulating hormone-expressing pituitary adenomas: presenting symptoms, treatment, outcomes, and recurrence. World Neurosurg. 2014;82(6):1224-1231.

64      Cyprich J, Donoho DA, Brunswick A, Hurth K, Carmichael JD, Weiss MH, Zada G. Surgical management of clinically silent thyrotropin pituitary adenomas: A single center series of 20 patients. J Clin Neurosci. 2020;71:70-75.

  1. Aydin S, Comunoglu N, Ahmedov ML, Korkmaz OP, Oz B, Kadioglu P, Gazioglu N, Tanriover N. Clinicopathologic Characteristics and Surgical Treatment of Plurihormonal Pituitary Adenomas. World Neurosurg. 2019;130:e765-e774.
  2. Erickson D, Scheithauer B, Atkinson J, Horvath E, Kovacs K, Lloyd RV, Young WF, Jr. Silent subtype 3 pituitary adenoma: a clinicopathologic analysis of the Mayo Clinic experience. Clin Endocrinol (Oxf). 2009;71(1):92-99.
  3. Mete O, Gomez-Hernandez K, Kucharczyk W, Ridout R, Zadeh G, Gentili F, Ezzat S, Asa SL. Silent subtype 3 pituitary adenomas are not always silent and represent poorly differentiated monomorphous plurihormonal Pit-1 lineage adenomas. Mod Pathol. 2016;29(2):131-142.
  4. Micko A, Rotzer T, Hoftberger R, Vila G, Oberndorfer J, Frischer JM, Knosp E, Wolfsberger S. Expression of additional transcription factors is of prognostic value for aggressive behavior of pituitary adenomas. J Neurosurg. 2020;134(3):1139-1146.
  5. Freda PU, Beckers AM, Katznelson L, Molitch ME, Montori VM, Post KD, Vance ML, Endocrine S. Pituitary incidentaloma: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(4):894-904.
  6. do Carmo Dias Gontijo M, de Souza Vasconcellos L, Ribeiro-Oliveira A, Jr. Hook effect and linear range in prolactin assays: distinct confounding entities. Pituitary. 2016;19(4):458-459.
  7. Vilar L, Abucham J, Albuquerque JL, Araujo LA, Azevedo MF, Boguszewski CL, Casulari LA, Cunha Neto MBC, Czepielewski MA, Duarte FHG, Faria MDS, Gadelha MR, Garmes HM, Glezer A, Gurgel MH, Jallad RS, Martins M, Miranda PAC, Montenegro RM, Musolino NRC, Naves LA, Ribeiro-Oliveira Junior A, Silva CMS, Viecceli C, Bronstein MD. Controversial issues in the management of hyperprolactinemia and prolactinomas - An overview by the Neuroendocrinology Department of the Brazilian Society of Endocrinology and Metabolism. Archives of endocrinology and metabolism. 2018;62(2):236-263.
  8. Glezer A, Bronstein MD. Hyperprolactinemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  9. Carosi G, Malchiodi E, Ferrante E, Sala E, Verrua E, Profka E, Giavoli C, Filopanti M, Beck-Peccoz P, Spada A, Mantovani G. Hypothalamic-Pituitary Axis in Non-Functioning Pituitary Adenomas: Focus on the Prevalence of Isolated Central Hypoadrenalism. Neuroendocrinology. 2015;102(4):267-273.
  10. Boguszewski CL, de Castro Musolino NR, Kasuki L. Management of pituitary incidentaloma. Best Pract Res Clin Endocrinol Metab. 2019;33(2):101268.
  11. Fleseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, Samuels MH. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(11):3888-3921.
  12. El-Farhan N, Pickett A, Ducroq D, Bailey C, Mitchem K, Morgan N, Armston A, Jones L, Evans C, Rees DA. Method-specific serum cortisol responses to the adrenocorticotrophin test: comparison of gas chromatography-mass spectrometry and five automated immunoassays. Clin Endocrinol (Oxf). 2013;78(5):673-680.
  13. Bonneville F. Silent Corticotroph Adenoma. In: Bonneville J, F B, Cattin F, S N, eds. MRI of the pituitary glan: Springer; 2016.
  14. Raverot G, Burman P, McCormack A, Heaney A, Petersenn S, Popovic V, Trouillas J, Dekkers OM, European Society of E. European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol. 2018;178(1):G1-G24.
  15. Boertien TM, Booij J, Majoie C, Drent ML, Pereira AM, Biermasz NR, Simsek S, Veldman RG, Stokkel MPM, Bisschop PH, Fliers E. (68)Ga-DOTATATE PET imaging in clinically non-functioning pituitary macroadenomas. Eur J Hybrid Imaging. 2020;4(1):4.
  16. Kaltsas GA, Evanson J, Chrisoulidou A, Grossman AB. The diagnosis and management of parasellar tumours of the pituitary. Endocr Relat Cancer. 2008;15(4):885-903.
  17. Samejima N, Yamada S, Takada K, Sano T, Ozawa Y, Shimizu T, Usui M, Shishiba Y. Serum alpha-subunit levels in patients with pituitary adenomas. Clin Endocrinol (Oxf). 2001;54(4):479-484.
  18. Vieira Neto L, Wildemberg LE, Moraes AB, Colli LM, Kasuki L, Marques NV, Gasparetto EL, de Castro M, Takiya CM, Gadelha MR. Dopamine receptor subtype 2 expression profile in nonfunctioning pituitary adenomas and in vivo response to cabergoline therapy. Clin Endocrinol (Oxf). 2015;82(5):739-746.
  19. Greenman Y, Cooper O, Yaish I, Robenshtok E, Sagiv N, Jonas-Kimchi T, Yuan X, Gertych A, Shimon I, Ram Z, Melmed S, Stern N. Treatment of clinically nonfunctioning pituitary adenomas with dopamine agonists. Eur J Endocrinol. 2016;175(1):63-72.
  20. Tampourlou M, Ntali G, Ahmed S, Arlt W, Ayuk J, Byrne JV, Chavda S, Cudlip S, Gittoes N, Grossman A, Mitchell R, O'Reilly MW, Paluzzi A, Toogood A, Wass JAH, Karavitaki N. Outcome of Nonfunctioning Pituitary Adenomas That Regrow After Primary Treatment: A Study From Two Large UK Centers. J Clin Endocrinol Metab. 2017;102(6):1889-1897.
  21. Lucas JW, Bodach ME, Tumialan LM, Oyesiku NM, Patil CG, Litvack Z, Aghi MK, Zada G. Congress of Neurological Surgeons Systematic Review and Evidence-Based Guideline on Primary Management of Patients With Nonfunctioning Pituitary Adenomas. Neurosurgery. 2016;79(4):E533-535.
  22. Huang W, Molitch ME. Management of nonfunctioning pituitary adenomas (NFAs): observation. Pituitary. 2018;21(2):162-167.
  23. Murad MH, Fernandez-Balsells MM, Barwise A, Gallegos-Orozco JF, Paul A, Lane MA, Lampropulos JF, Natividad I, Perestelo-Perez L, Ponce de Leon-Lovaton PG, Albuquerque FN, Carey J, Erwin PJ, Montori VM. Outcomes of surgical treatment for nonfunctioning pituitary adenomas: a systematic review and meta-analysis. Clin Endocrinol (Oxf). 2010;73(6):777-791.
  24. Reddy R, Cudlip S, Byrne JV, Karavitaki N, Wass JA. Can we ever stop imaging in surgically treated and radiotherapy-naive patients with non-functioning pituitary adenoma? Eur J Endocrinol. 2011;165(5):739-744.
  25. Oystese KA, Evang JA, Bollerslev J. Non-functioning pituitary adenomas: growth and aggressiveness. Endocrine. 2016;53(1):28-34.
  26. Micko AS, Wohrer A, Wolfsberger S, Knosp E. Invasion of the cavernous sinus space in pituitary adenomas: endoscopic verification and its correlation with an MRI-based classification. J Neurosurg. 2015;122(4):803-811.
  27. Buchy M, Lapras V, Rabilloud M, Vasiljevic A, Borson-Chazot F, Jouanneau E, Raverot G. Predicting early post-operative remission in pituitary adenomas: evaluation of the modified knosp classification. Pituitary. 2019;22(5):467-475.
  28. Berkmann S, Lattmann J, Schuetz P, Diepers M, Remonda L, Fandino J, Buchfelder M, Mueller B. The Shape grading system: a classification for growth patterns of pituitary adenomas. Acta Neurochir (Wien). 2021;163(11):3181-3189.
  29. Losa M, Mortini P, Barzaghi R, Ribotto P, Terreni MR, Marzoli SB, Pieralli S, Giovanelli M. Early results of surgery in patients with nonfunctioning pituitary adenoma and analysis of the risk of tumor recurrence. J Neurosurg. 2008;108(3):525-532.
  30. Oh H, Cheun H, Kim YJ, Yoon HK, Kang H, Lee HC, Park HP, Kim JH, Kim YH. Cephalocaudal tumor diameter is a predictor of diabetes insipidus after endoscopic transsphenoidal surgery for non-functioning pituitary adenoma. Pituitary. 2021;24(3):303-311.
  31. Kinoshita Y, Taguchi A, Tominaga A, Sakoguchi T, Arita K, Yamasaki F. Predictive factors of postoperative diabetes insipidus in 333 patients undergoing transsphenoidal surgery for non-functioning pituitary adenoma. Pituitary. 2021.
  32. Gasco V, Caputo M, Lanfranco F, Ghigo E, Grottoli S. Management of GH treatment in adult GH deficiency. Best Pract Res Clin Endocrinol Metab. 2017;31(1):13-24.
  33. Chanson P, Raverot G, Castinetti F, Cortet-Rudelli C, Galland F, Salenave S, French Endocrinology Society non-functioning pituitary adenoma w-g. Management of clinically non-functioning pituitary adenoma. Ann Endocrinol (Paris). 2015;76(3):239-247.
  34. Arnold JR, Arnold DF, Marland A, Karavitaki N, Wass JA. GH replacement in patients with non-functioning pituitary adenoma (NFA) treated solely by surgery is not associated with increased risk of tumour recurrence. Clin Endocrinol (Oxf). 2009;70(3):435-438.
  35. Pal A, Capatina C, Tenreiro AP, Guardiola PD, Byrne JV, Cudlip S, Karavitaki N, Wass JA. Pituitary apoplexy in non-functioning pituitary adenomas: long term follow up is important because of significant numbers of tumour recurrences. Clin Endocrinol (Oxf). 2011;75(4):501-504.

100    Colao A, Cerbone G, Cappabianca P, Ferone D, Alfieri A, Di Salle F, Faggiano A, Merola B, de Divitiis E, Lombardi G. Effect of surgery and radiotherapy on visual and endocrine function in nonfunctioning pituitary adenomas. J Endocrinol Invest. 1998;21(5):284-290.

  1. Brochier S, Galland F, Kujas M, Parker F, Gaillard S, Raftopoulos C, Young J, Alexopoulou O, Maiter D, Chanson P. Factors predicting relapse of nonfunctioning pituitary macroadenomas after neurosurgery: a study of 142 patients. Eur J Endocrinol. 2010;163(2):193-200.
  2. Minniti G, Clarke E, Scaringi C, Enrici RM. Stereotactic radiotherapy and radiosurgery for non-functioning and secreting pituitary adenomas. Reports of practical oncology and radiotherapy : journal of Greatpoland Cancer Center in Poznan and Polish Society of Radiation Oncology. 2016;21(4):370-378.
  3. Ramirez C, Cheng S, Vargas G, Asa SL, Ezzat S, Gonzalez B, Cabrera L, Guinto G, Mercado M. Expression of Ki-67, PTTG1, FGFR4, and SSTR 2, 3, and 5 in nonfunctioning pituitary adenomas: a high throughput TMA, immunohistochemical study. J Clin Endocrinol Metab. 2012;97(5):1745-1751.
  4. Cohen-Inbar O, Xu Z, Lee CC, Wu CC, Chytka T, Silva D, Sharma M, Radwan H, Grills IS, Nguyen B, Siddiqui Z, Mathieu D, Iorio-Morin C, Wolf A, Cifarelli CP, Cifarelli DT, Lunsford LD, Kondziolka D, Sheehan JP. Prognostic significance of corticotroph staining in radiosurgery for non-functioning pituitary adenomas: a multicenter study. J Neurooncol. 2017;135(1):67-74.
  5. Strickland BA, Shahrestani S, Briggs RG, Jackanich A, Tavakol S, Hurth K, Shiroishi MS, Liu CJ, Carmichael JD, Weiss M, Zada G. Silent corticotroph pituitary adenomas: clinical characteristics, long-term outcomes, and management of disease recurrence. J Neurosurg. 2021:1-8.
  6. Fountas A, Lavrentaki A, Subramanian A, Toulis KA, Nirantharakumar K, Karavitaki N. Recurrence in silent corticotroph adenomas after primary treatment: A systematic review and meta-analysis. J Clin Endocrinol Metab. 2018.
  7. Dekkers OM, Hammer S, de Keizer RJ, Roelfsema F, Schutte PJ, Smit JW, Romijn JA, Pereira AM. The natural course of non-functioning pituitary macroadenomas. Eur J Endocrinol. 2007;156(2):217-224.
  8. Galland F, Vantyghem MC, Cazabat L, Boulin A, Cotton F, Bonneville JF, Jouanneau E, Vidal-Trecan G, Chanson P. Management of nonfunctioning pituitary incidentaloma. Ann Endocrinol (Paris). 2015;76(3):191-200.
  9. Guo BJ, Yang ZL, Zhang LJ. Gadolinium Deposition in Brain: Current Scientific Evidence and Future Perspectives. Front Mol Neurosci. 2018;11:335.
  10. Garcia EC, Naves LA, Silva AO, de Castro LF, Casulari LA, Azevedo MF. Short-term treatment with cabergoline can lead to tumor shrinkage in patients with nonfunctioning pituitary adenomas. Pituitary. 2013;16(2):189-194.
  11. Nobels FR, de Herder WW, van den Brink WM, Kwekkeboom DJ, Hofland LJ, Zuyderwijk J, de Jong FH, Lamberts SW. Long-term treatment with the dopamine agonist quinagolide of patients with clinically non-functioning pituitary adenoma. Eur J Endocrinol. 2000;143(5):615-621.
  12. Pivonello R, Matrone C, Filippella M, Cavallo LM, Di Somma C, Cappabianca P, Colao A, Annunziato L, Lombardi G. Dopamine receptor expression and function in clinically nonfunctioning pituitary tumors: comparison with the effectiveness of cabergoline treatment. J Clin Endocrinol Metab. 2004;89(4):1674-1683.
  13. Batista RL, Musolino NRC, Cescato VAS, da Silva GO, Medeiros RSS, Herkenhoff CGB, Trarbach EB, Cunha-Neto MB. Cabergoline in the Management of Residual Nonfunctioning Pituitary Adenoma: A Single-Center, Open-Label, 2-Year Randomized Clinical Trial. Am J Clin Oncol. 2019;42(2):221-227.
  14. Greenman Y, Bronstein MD. Cabergoline should be attempted in progressing non-functioning pituitary macroadenoma. Eur J Endocrinol. 2021;185(4):D11-D20.
  15. Yavropoulou MP, Tsoli M, Barkas K, Kaltsas G, Grossman A. The natural history and treatment of non-functioning pituitary adenomas (non-functioning PitNETs). Endocr Relat Cancer. 2020;27(10):R375-R390.
  16. Even-Zohar N, Greenman Y. Management of NFAs: medical treatment. Pituitary. 2018;21(2):168-175.
  17. Taboada GF, Luque RM, Bastos W, Guimaraes RF, Marcondes JB, Chimelli LM, Fontes R, Mata PJ, Filho PN, Carvalho DP, Kineman RD, Gadelha MR. Quantitative analysis of somatostatin receptor subtype (SSTR1-5) gene expression levels in somatotropinomas and non-functioning pituitary adenomas. Eur J Endocrinol. 2007;156(1):65-74.
  18. Lee M, Lupp A, Mendoza N, Martin N, Beschorner R, Honegger J, Schlegel J, Shively T, Pulz E, Schulz S, Roncaroli F, Pellegata NS. SSTR3 is a putative target for the medical treatment of gonadotroph adenomas of the pituitary. Endocr Relat Cancer. 2015;22(1):111-119.
  19. Gasperi M, Petrini L, Pilosu R, Nardi M, Marcello A, Mastio F, Bartalena L, Martino E. Octreotide treatment does not affect the size of most non-functioning pituitary adenomas. J Endocrinol Invest. 1993;16(7):541-543.
  20. Fusco A, Giampietro A, Bianchi A, Cimino V, Lugli F, Piacentini S, Lorusso M, Tofani A, Perotti G, Lauriola L, Anile C, Maira G, Pontecorvi A, De Marinis L. Treatment with octreotide LAR in clinically non-functioning pituitary adenoma: results from a case-control study. Pituitary. 2012;15(4):571-578.
  21. Vieria Neto L, Wildemberg LE, Colli LM, Kasuki L, Marques NV, Moraes AB, Gasparetto EL, Takiya CM, Castro M, Gadelha MR. ZAC1 and SSTR2 are downregulated in non-functioning pituitary adenomas but not in somatotropinomas. PLoS One. 2013;8(10):e77406.
  22. Oystese KA, Casar-Borota O, Normann KR, Zucknick M, Berg JP, Bollerslev J. Estrogen Receptor alpha, a Sex-Dependent Predictor of Aggressiveness in Nonfunctioning Pituitary Adenomas: SSTR and Sex Hormone Receptor Distribution in NFPA. J Clin Endocrinol Metab. 2017;102(9):3581-3590.
  23. Zatelli MC, Piccin D, Vignali C, Tagliati F, Ambrosio MR, Bondanelli M, Cimino V, Bianchi A, Schmid HA, Scanarini M, Pontecorvi A, De Marinis L, Maira G, degli Uberti EC. Pasireotide, a multiple somatostatin receptor subtypes ligand, reduces cell viability in non-functioning pituitary adenomas by inhibiting vascular endothelial growth factor secretion. Endocr Relat Cancer. 2007;14(1):91-102.
  24. Castellnou S, Vasiljevic A, Lapras V, Raverot V, Alix E, Borson-Chazot F, Jouanneau E, Raverot G, Lasolle H. SST5 expression and USP8 mutation in functioning and silent corticotroph pituitary tumors. Endocrine connections. 2020.
  25. Bujko M, Kober P, Boresowicz J, Rusetska N, Paziewska A, Dabrowska M, Piascik A, Pekul M, Zielinski G, Kunicki J, Bonicki W, Ostrowski J, Siedlecki JA, Maksymowicz M. USP8 mutations in corticotroph adenomas determine a distinct gene expression profile irrespective of functional tumour status. Eur J Endocrinol. 2019;181(6):615-627.
  26. Perez-Rivas LG, Theodoropoulou M, Ferrau F, Nusser C, Kawaguchi K, Stratakis CA, Faucz FR, Wildemberg LE, Assie G, Beschorner R, Dimopoulou C, Buchfelder M, Popovic V, Berr CM, Toth M, Ardisasmita AI, Honegger J, Bertherat J, Gadelha MR, Beuschlein F, Stalla G, Komada M, Korbonits M, Reincke M. The Gene of the Ubiquitin-Specific Protease 8 Is Frequently Mutated in Adenomas Causing Cushing's Disease. J Clin Endocrinol Metab. 2015;100(7):E997-1004.
  27. Boertien TM, Drent ML, Booij J, Majoie C, Stokkel MPM, Hoogmoed J, Pereira A, Biermasz NR, Simsek S, Groote Veldman R, Tanck MWT, Fliers E, Bisschop PH. The GALANT trial: study protocol of a randomised placebo-controlled trial in patients with a (68) Ga -DOTATATE PET-positive, clinically non-functioning pituitary macroadenoma on the effect of lanreotide on tumour size. BMJ Open. 2020;10(8):e038250.
  28. Gulde S, Wiedemann T, Schillmaier M, Valenca I, Lupp A, Steiger K, Yen HY, Bauerle S, Notni J, Luque R, Schmid H, Schulz S, Ankerst DP, Schilling F, Pellegata NS. Gender-Specific Efficacy Revealed by Head-to-Head Comparison of Pasireotide and Octreotide in a Representative In Vivo Model of Nonfunctioning Pituitary Tumors. Cancers (Basel). 2021;13(12).
  29. Florio T, Barbieri F, Spaziante R, Zona G, Hofland LJ, van Koetsveld PM, Feelders RA, Stalla GK, Theodoropoulou M, Culler MD, Dong J, Taylor JE, Moreau JP, Saveanu A, Gunz G, Dufour H, Jaquet P. Efficacy of a dopamine-somatostatin chimeric molecule, BIM-23A760, in the control of cell growth from primary cultures of human non-functioning pituitary adenomas: a multi-center study. Endocr Relat Cancer. 2008;15(2):583-596.
  30. Halem HA, Hochgeschwender U, Rih JK, Nelson R, Johnson GA, Thiagalingam A, Culler MD. TBR-760, a Dopamine-Somatostatin Compound, Arrests Growth of Aggressive Nonfunctioning Pituitary Adenomas in Mice. Endocrinology. 2020;161(8).
  31. McCormack AI, Wass JA, Grossman AB. Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status. Eur J Clin Invest. 2011;41(10):1133-1148.
  32. McCormack A, Dekkers OM, Petersenn S, Popovic V, Trouillas J, Raverot G, Burman P, collaborators ESEs. Treatment of aggressive pituitary tumours and carcinomas: results of a European Society of Endocrinology (ESE) survey 2016. Eur J Endocrinol. 2018;178(3):265-276.
  33. Widhalm G, Wolfsberger S, Preusser M, Woehrer A, Kotter MR, Czech T, Marosi C, Knosp E. O(6)-methylguanine DNA methyltransferase immunoexpression in nonfunctioning pituitary adenomas: are progressive tumors potential candidates for temozolomide treatment? Cancer. 2009;115(5):1070-1080.
  34. Ilie MD, Raverot G. Treatment Options for Gonadotroph Tumors: Current State and Perspectives. J Clin Endocrinol Metab. 2020;105(10).
  35. Dixit S, Baker L, Walmsley V, Hingorani M. Temozolomide-related idiosyncratic and other uncommon toxicities: a systematic review. Anticancer Drugs. 2012;23(10):1099-1106.
  36. Ilie MD, Lasolle H, Raverot G. Emerging and Novel Treatments for Pituitary Tumors. J Clin Med. 2019;8(8).
  37. Nakano-Tateno T, Lau KJ, Wang J, McMahon C, Kawakami Y, Tateno T, Araki T. Multimodal Non-Surgical Treatments of Aggressive Pituitary Tumors. Front Endocrinol (Lausanne). 2021;12:624686.
  38. Maclean J, Aldridge M, Bomanji J, Short S, Fersht N. Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: variable clinical response in preliminary evaluation. Pituitary. 2014;17(6):530-538.
  39. Komor J, Reubi JC, Christ ER. Peptide receptor radionuclide therapy in a patient with disabling non-functioning pituitary adenoma. Pituitary. 2014;17(3):227-231.
  40. Novruzov F, Aliyev JA, Jaunmuktane Z, Bomanji JB, Kayani I. The use of (68)Ga DOTATATE PET/CT for diagnostic assessment and monitoring of (177)Lu DOTATATE therapy in pituitary carcinoma. Clin Nucl Med. 2015;40(1):47-49.
  41. Giuffrida G, Ferrau F, Laudicella R, Cotta OR, Messina E, Granata F, Angileri FF, Vento A, Alibrandi A, Baldari S, Cannavo S. Peptide receptor radionuclide therapy for aggressive pituitary tumors: a monocentric experience. Endocrine connections. 2019;8(5):528-535.
  42. Bengtsson D, Schroder HD, Andersen M, Maiter D, Berinder K, Feldt Rasmussen U, Rasmussen AK, Johannsson G, Hoybye C, van der Lely AJ, Petersson M, Ragnarsson O, Burman P. Long-term outcome and MGMT as a predictive marker in 24 patients with atypical pituitary adenomas and pituitary carcinomas given treatment with temozolomide. J Clin Endocrinol Metab. 2015;100(4):1689-1698.
  43. Tampourlou M, Fountas A, Ntali G, Karavitaki N. Mortality in patients with non-functioning pituitary adenoma. Pituitary. 2018;21(2):203-207.
  44. Ntali G, Capatina C, Fazal-Sanderson V, Byrne JV, Cudlip S, Grossman AB, Wass JA, Karavitaki N. Mortality in patients with non-functioning pituitary adenoma is increased: systematic analysis of 546 cases with long follow-up. Eur J Endocrinol. 2016;174(2):137-145.

145    Chang EF, Zada G, Kim S, Lamborn KR, Quinones-Hinojosa A, Tyrrell JB, Wilson CB, Kunwar S. Long-term recurrence and mortality after surgery and adjuvant radiotherapy for nonfunctional pituitary adenomas. J Neurosurg. 2008;108(4):736-745.

  1. Hammarstrand C, Ragnarsson O, Hallen T, Andersson E, Skoglund T, Nilsson AG, Johannsson G, Olsson DS. Higher glucocorticoid replacement doses are associated with increased mortality in patients with pituitary adenoma. Eur J Endocrinol. 2017;177(3):251-256.
  2. Andela CD, Scharloo M, Pereira AM, Kaptein AA, Biermasz NR. Quality of life (QoL) impairments in patients with a pituitary adenoma: a systematic review of QoL studies. Pituitary. 2015;18(5):752-776.
  3. Dekkers OM, van der Klaauw AA, Pereira AM, Biermasz NR, Honkoop PJ, Roelfsema F, Smit JW, Romijn JA. Quality of life is decreased after treatment for nonfunctioning pituitary macroadenoma. J Clin Endocrinol Metab. 2006;91(9):3364-3369.
  4. Biermasz NR, Joustra SD, Donga E, Pereira AM, van Duinen N, van Dijk M, van der Klaauw AA, Corssmit EP, Lammers GJ, van Kralingen KW, van Dijk JG, Romijn JA. Patients previously treated for nonfunctioning pituitary macroadenomas have disturbed sleep characteristics, circadian movement rhythm, and subjective sleep quality. J Clin Endocrinol Metab. 2011;96(5):1524-1532.
  5. Capatina C, Christodoulides C, Fernandez A, Cudlip S, Grossman AB, Wass JA, Karavitaki N. Current treatment protocols can offer a normal or near-normal quality of life in the majority of patients with non-functioning pituitary adenomas. Clin Endocrinol (Oxf). 2013;78(1):86-93.
  6. Joustra SD, Kruijssen E, Verstegen MJ, Pereira AM, Biermasz NR. Determinants of altered sleep-wake rhythmicity in patients treated for nonfunctioning pituitary macroadenomas. J Clin Endocrinol Metab. 2014;99(12):4497-4505.
  7. Ratnasingam J, Lenders N, Ong B, Boros S, Russell AW, Inder WJ, Ho KKY. Predictors for secondary therapy after surgical resection of nonfunctioning pituitary adenomas. Clin Endocrinol (Oxf). 2017;87(6):717-724.
  8. Noh TW, Jeong HJ, Lee MK, Kim TS, Kim SH, Lee EJ. Predicting recurrence of nonfunctioning pituitary adenomas. J Clin Endocrinol Metab. 2009;94(11):4406-4413.
  9. Hasanov R, Aydogan BI, Kiremitci S, Erden E, Gullu S. The Prognostic Roles of the Ki-67 Proliferation Index, P53 Expression, Mitotic Index, and Radiological Tumor Invasion in Pituitary Adenomas. Endocr Pathol. 2019;30(1):49-55.
  10. Trouillas J, Roy P, Sturm N, Dantony E, Cortet-Rudelli C, Viennet G, Bonneville JF, Assaker R, Auger C, Brue T, Cornelius A, Dufour H, Jouanneau E, Francois P, Galland F, Mougel F, Chapuis F, Villeneuve L, Maurage CA, Figarella-Branger D, Raverot G, members of H, Barlier A, Bernier M, Bonnet F, Borson-Chazot F, Brassier G, Caulet-Maugendre S, Chabre O, Chanson P, Cottier JF, Delemer B, Delgrange E, Di Tommaso L, Eimer S, Gaillard S, Jan M, Girard JJ, Lapras V, Loiseau H, Passagia JG, Patey M, Penfornis A, Poirier JY, Perrin G, Tabarin A. A new prognostic clinicopathological classification of pituitary adenomas: a multicentric case-control study of 410 patients with 8 years post-operative follow-up. Acta Neuropathol. 2013;126(1):123-135.
  11. Lelotte J, Mourin A, Fomekong E, Michotte A, Raftopoulos C, Maiter D. Both invasiveness and proliferation criteria predict recurrence of non-functioning pituitary macroadenomas after surgery: a retrospective analysis of a monocentric cohort of 120 patients. Eur J Endocrinol. 2018;178(3):237-246.
  12. Hallen T, Olsson DS, Hammarstrand C, Orndal C, Engvall A, Ragnarsson O, Skoglund T, Johannsson G. MCM7 as a marker of postsurgical progression in non-functioning pituitary adenomas. Eur J Endocrinol. 2021;184(4):521-531.
  13. Lyu W, Fei X, Chen C, Tang Y. Nomogram predictive model of post-operative recurrence in non-functioning pituitary adenoma. Gland Surg. 2021;10(2):807-815.
  14. Srirangam Nadhamuni V, Korbonits M. Novel Insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev. 2020;41(6).
  15. Neou M, Villa C, Armignacco R, Jouinot A, Raffin-Sanson ML, Septier A, Letourneur F, Diry S, Diedisheim M, Izac B, Gaspar C, Perlemoine K, Verjus V, Bernier M, Boulin A, Emile JF, Bertagna X, Jaffrezic F, Laloe D, Baussart B, Bertherat J, Gaillard S, Assie G. Pangenomic Classification of Pituitary Neuroendocrine Tumors. Cancer Cell. 2020;37(1):123-134 e125.
  16. Cooper O, Ben-Shlomo A, Bonert V, Bannykh S, Mirocha J, Melmed S. Silent corticogonadotroph adenomas: clinical and cellular characteristics and long-term outcomes. Horm Cancer. 2010;1(2):80-92.
  17. Lu JQ, Adam B, Jack AS, Lam A, Broad RW, Chik CL. Immune Cell Infiltrates in Pituitary Adenomas: More Macrophages in Larger Adenomas and More T Cells in Growth Hormone Adenomas. Endocr Pathol. 2015;26(3):263-272.
  18. Wang Z, Guo X, Gao L, Deng K, Lian W, Bao X, Feng M, Duan L, Zhu H, Xing B. The Immune Profile of Pituitary Adenomas and a Novel Immune Classification for Predicting Immunotherapy Responsiveness. J Clin Endocrinol Metab. 2020;105(9).

Pituitary and Adrenal Disorders of Pregnancy

ABSTRACT

 

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

 

PITUITARY DISORDERS IN PREGNANCY

 

Anterior Pituitary Gland Anatomy

 

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

 

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

 

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

 

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

 

Prolactin

 

INTRODUCTION

 

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

 

FERTILITY

 

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

 

PRECONCEPTION MANAGEMENT AND RESTORATION OF MENSES

 

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

 

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

 

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

 

EFFECTS OF DOPAMINE AGONISTS ON THE DEVELOPING FETUS

 

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

 

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

 

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

 

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

 

Bromocriptine

(n (%))

Cabergoline

(n (%))

Normal (%)

Pregnancies

     Spontaneous abortions

     Terminations

     Ectopic

     Hydatiform moles

6239 (100)

620 (9.9)

75 (1.2)

31 (0.5)

11 (0.2)

968 (100)

73 (7.5)

63a (6.5)

3 (0.3)

1 (0.1)

100

10-15

20

1.0-1.5

0.1-0.15

Deliveries (known duration)

     At term (>37 weeks)

     Preterm (<37 weeks)

4139 (100)

 

3620 (87.5)

519 (12.5)

705 (100)

 

634b (89.9)

71 (10.1)

100

 

87.3

12.7

Deliveries (known outcome)

     Single births

      Multiple births

5120 (100)

 

5031 (98.3)

89 (1.7)

629 (100)

 

614 (97.6)

15 (2.4)

100

 

96.8

3.2

Babies (known details)

     Normal

     With malformations

5213 (100)

5030 (98.2)

93 (1.8)

822 (100)

801 (97.4)

21 (2.4)

100

97

3.0

aEleven of these terminations were for malformations

bFive of these births were stillbirths

 

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

 

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

 

EFFECT OF PREGNANCY ON PROLACTINOMA SIZE

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

 

MANAGEMENT FOLLOWING CONCEPTION

 

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

 

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

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

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

 

BREASTFEEDING

 

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

 

Growth Hormone

 

INTRODUCTION

 

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

 

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

 

DIAGNOSIS OF ACROMEGALY DURING PREGNANCY

 

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

 

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

 

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

 

FERTILITY IN WOMEN WITH ACROMEGALY

 

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

 

EFFECT OF PREGNANCY ON ACROMEGALY

 

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

 

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

 

EFFECT OF ACROMEGALY ON THE NEONATE

 

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

 

METABOLIC AND CARDIOVASCULAR COMPLICATIONS

 

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

 

MANAGEMENT APPROACH

 

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

 

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

Figure 2: Schematic for the Management of Acromegaly During Pregnancy

MEDICAL THERAPY

 

Somatostatin Analogues

 

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

 

Dopamine Agonists

 

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

 

Growth Hormone Receptor Antagonists

 

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

 

TSH-Secreting Adenomas

 

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

 

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

 

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

 

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

 

Table 2. Summary of Pregnancies in Women with TSH Adenomas

Tumor

Diagnosis

Prior TS

Previous medical therapy

TS in pregnancy

Medical therapy in pregnancy

Delivery

TSH-secreting macroadenoma (73)

Prior to pregnancy

No

Oct-CSI (300ug/day)

No

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

ECS

TSH-secreting macroadenoma with hypothalamic disconnection (74)

Prior to pregnancy

Yes

PTU and CBL prior to TS

 

Oct-LAR post TS

 

Post-TS RT

No

Br throughout gestation

 

RT (<9wks)

 

Oct-LAR- throughout gestation

NA

TSH-secreting macroadenoma with hypothalamic disconnection (75)

Prior to pregnancy

No

PTU 50mg TDS and Br 2.5mg OD

Yes

Reduced dose of PTU

 

Br continued

ECS

TSH-secreting microadenoma (76)

24 weeks gestation

No

N/A

No

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

 

NVD

TSH-secreting macroadenoma (77)

20 weeks’ gestation

N/A

N/A

No

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

NVD

TSH-secreting macroadenoma (78)

Prior to pregnancy

Yes

Octreotide

No

Octreotide stopped

NVD

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

 

Non-Functioning Pituitary Adenomas and Gonadotrophin-Secreting Adenomas

 

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

 

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

 

Pituitary Apoplexy

 

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

 

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

 

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

 

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

 

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

 

Sheehan’s Syndrome

 

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

 

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

 

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

 

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

 

Hypopituitarism

 

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

 

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

 

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

 

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

 

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

 

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

 

Lymphocytic Hypophysitis

 

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

 

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

 

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

Posterior Pituitary Gland Physiology

 

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

 

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

 

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

 

Diabetes Insipidus (DI)

 

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

 

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

 

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

 

CENTRAL DI

 

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

 

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

 

Table 3. Causes of Central Diabetes Insipidus

Primary

Idiopathic

-

Genetic

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

Neurohypophyseal diabetes insipidus

Developmental Syndromes

Septic-optic dysplasia

Secondary/ Acquired

Trauma

Head injury

Post-surgery

Post radiotherapy

Vascular

Carotid aneurysm

Cavernous sinus thrombosis

Tumor

Craniopharyngioma

Germinoma

Metastases

Pituitary adenomas

Inflammatory

Sarcoidosis

Langerhans cell histiocytosis

Meningitis/ Encephalitis

Infundibuloneurohypophysitis

Guillain-Barre Syndrome

Lymphocytic hypophysitis

Infection

Tuberculosis

Fungal diseases

Post-Partum

Sheehan’s syndrome

Pituitary apoplexy

 

NEPHROGENIC DI

 

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

 

TRANSIENT DI OF PREGNANCY

 

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

 

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

 

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

 

Syndrome of Inappropriate Antidiuretic Hormone (SIADH)

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

 

ADRENAL DISORDERS IN PREGNANCY

 

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

 

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

 

Cushing’s Syndrome During Pregnancy

 

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

 

INVESTIGATIONS

 

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

 

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

 

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

 

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

 

COMPLICATIONS

 

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

 

TREATMENT

 

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

 

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

 

Adrenal Insufficiency

 

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

 

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

 

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

 

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

 

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

 

COMPLICATIONS

 

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

 

INVESTIGATIONS

 

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

 

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

 

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

 

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

 

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

 

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

 

MANAGEMENT

 

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

 

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

 

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

 

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

 

Congenital Adrenal Hyperplasia

 

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

 

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

 

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

 

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

 

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

 

Primary Hyperaldosteronism During Pregnancy

 

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

 

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

 

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

 

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

 

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

 

Pheochromocytoma and Paraganglioma in Pregnancy


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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

SUMMARY

 

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

 

ACKNOWLEDGEMENT

 

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

 

REFERENCES

                                                                                                                                 

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

Hyperthyroidism During Pregnancy- Diagnostic

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

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

 

Special Considerations Relevant to Pediatric Obesity

ABSTRACT

 

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

 

INTRODUCTION

 

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

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

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

 

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

 

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

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

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

 

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

 

DEFINITION AND EPIDEMIOLOGY

 

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

 

Several basic principles are pertinent to such an assessment:

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

 

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

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

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

 

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

 

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

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

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

 

ENERGY HOMEOSTASIS

 

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

 

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

 

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

 

MOLECULAR GENETICS OF BODY FATNESS

 

Heritability of Body Fatness

 

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

 

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

 

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

 

Common Single Gene Mutations Associated with Obesity

 

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

 

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

Syndrome/Gene

Chromosome

Phenotype

Alström syndrome/ ALMS1

2p14-p13

(Recessive)

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

Bardet-Biedl syndrome (22 different genes)

16q21

15q22-q23

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

Beckwith-Wiedemann syndrome

11p15.5

(Recessive)

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

Börjeson-Forssmann-Lehman syndrome/ PHF6

X-linked

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

Carpenter /RAB23and MEGF8

Unknown

(Recessive)

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

Cohen /COH1

8q22-q23

(Recessive)

Mental retardation, microcephaly, short stature, dysmorphic facies

Leptin deficiency / LEP

7q31.3

(Recessive)

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

Leptin Receptor / LEPR

1p31-p32

(Recessive)

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

Melanocortin 4 Receptor /MC4R

18q22

(Dominant)

Obesity – early onset hyperphagia, increased bone density

Neisidioblastosis

11p15.1

(Recessive or Dominant)

Hyperinsulinemia, hypoglycemia, intolerance of fasting

Prader Willi syndrome

15q11-q12

(Uniparental Maternal Disomy)

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

Pro-opiomelanocortin / POMC

2p23.3

(Recessive)

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

Prohormone Convertase/ PCSK1

5q15-q21

(Recessive)

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

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

20q13.2

(Dominant)

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

 

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

 

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

 

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

 

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

 

 

Epigenetics

 

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

 

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

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

 

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

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

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

 

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

 

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

 

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

 

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

 

Early Feeding Practices

 

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

 

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

 

Early Life BMI Trajectories

 

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

 

Social Determinants of Health

 

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

 

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

 

Physical Activity, Sedentary Behavior, and Sleep

 

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

 

MORBIDITIES ASSOCIATED WITH OBESITY IN CHILDREN

 

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

 

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

Cardiovascular

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

Respiratory

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

Gastrointestinal

Nonalcoholic fatty liver disease, gallstones, gastroesophageal reflux disease

Endocrine

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

Orthopedic

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

Dermatologic

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

Immunologic

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

Psychologic

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

Lymphatic

Obesity associated lymphedema of the lower legs

Malignancy

Higher lifetime risk of obesity related cancers

 

Pediatric Obesity and Cardiovascular Risk Factors

 

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

 

Pediatric Obesity and Type 2 Diabetes Mellitus

 

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

 

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

 

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

 

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

 

ENDOCRINE CHANGES ASSOCIATED WITH OBESITY IN CHILDREN

 

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

 

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

Somatotroph

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

Lactotroph

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

Gonadotroph

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

Thyroid

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

Adrenal

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

Gonad

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

 Pancreas

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

 

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

 

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

Disease

Structural/Biochemical Lesion

Clinical Features

Acquired hypothalamic lesions

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

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

Cushing’s Disease / Syndrome

Hypercortisolism   

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

Hypothyroidism

Hypothalamic, pituitary, or thyroidal

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

ROHHAD or ROHHADNET syndrome*

Hypothalamic

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

Medications

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

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

 

WEIGHT STIGMATIZATION (FAT SHAMING)

 

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

 

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

 

Prenatal Care

 

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

 

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

 

Initial Evaluation

 

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

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

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

 

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

 

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

 

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

 

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

 

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

 

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

 

Therapeutic Intervention

 

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

 

DIETARY RECOMMENDATIONS

 

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

 

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

 

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

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

 

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

 

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

 

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

 

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

 

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

 

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

 

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

 

THERAPEUTIC EXERCISE

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

 

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

 

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

 

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

 

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

 

DIGITAL INTERVENTIONS

 

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

 

PHARMACOLOGICAL AND SURGICAL INTERVENTIONS

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

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

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

 

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

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

 

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

 

CONTRINDICATIONS TO BARIATRIC SURGERY INCLUDE:

 

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

 

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

 

ADDRESSING WEIGHT STIGMATIZATION

 

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

 

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

 

OTHER INTERVENTIONS

 

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

 

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

 

REFERENCES

 

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

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

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

 

Behavioral Approaches to Obesity Management

ABSTRACT

 

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

 

INTRODUCTION

 

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

 

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

 

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

 

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

 

Diabetes Prevention Program

 

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

 

Look AHEAD (Action for Health in Diabetes) Study

 

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

 

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

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

 

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

 

LIFESTYLE INTERVENTION COMPONENTS

 

Dietary Recommendations

 

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

 

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

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

 

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

 

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

Study

N

No. Lifestyle Sessions Provided

Dietary Intervention

Weight Change

 Month

 Comment/ Other Results

Dansinger et al(26)

   160 (51% F)

   58%    completed

 

 

4

   Atkins (low-carb)

   Zone (even distribution)

   Weight Watchers (points based)

   Ornish (low-fat)

-2.1 kg a

-3.2 kg a

 

-3.0 kg a

 

-3.3 kg a

 

12

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

     Weight loss was associated with level of adherence.

     Each diet decreased LDL/HDL ratio.

 

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

Das et al(34)*

34 (% F  unknown)

85% completed

52

    Low-glycemic load

    High-glycemic load

-7.8% a

 

-8.0% a

12

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

Fabricatore et al(28)

  79 (80% F)

63% completed

30

Low-glycemic load

     Low-fat

-4.5% a

 

    -6.4% a

9

All participants had type 2 diabetes.

     Larger reductions in HbA1cin the low-glycemic load group.

Foster et al(29)

63 (68% F)

59% completed

3

     Low-carbohydrate (high protein, high fat)

    Conventional (high-carbohydrate, low-fat)

-4.4% a

 

 

 

-2.5% a

12

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

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

Foster et al(25)

307 (68% F)

63% completed

38

   Low-carbohydrate

   Low-fat

-6.3 kg a

 

-7.4 kg a

24

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

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

Gardner et al(30)

 

311 (100% F)

80% completed

 

 

8

    Atkins (low-carbohydrate)

    Zone (even distribution)

    LEARN (calorie-restricted)

    Ornish (low-fat)

-4.7 kg a

 

-1.6 kg b

 

-2.2 kg ab

 

-2.6 kg ab

12

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

     No differences in insulin or blood glucose between groups.

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

Sacks et al(24)

   811 (64% F)

7 9.5%   completed

 

66

    Low-fat, average protein (highest carbohydrate)

   Low-fat, high-protein

   High-fat, average-protein

    High-fat, high-protein (lowest carbohydrate)

-3.0 kg a

 

 

-3.8 kg a

 

-3.2 kg a

 

-3.4 kg a

24

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

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

Shai et al(32)

 

   322 (14% F)

8 4.6%   completed

 

 

24

    Low-fat

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

    Low-carbohydrate

-2.9 kg a

-4.4 kg b

 

 

 

 

 

 

-4.7 kg b

 

 

24

     No significant change in LDL cholesterol in any group.

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

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

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

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

     All groups had a significant decrease in blood pressure.

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

Stern et al(33)

 

132 (17% F)

66% completed

15

    Low-carbohydrate

    Conventional (low-fat)

-5.1 kg a

 

-3.1 kg a

12

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

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

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

Yancy et al(35)

120 (76% F)

66% completed

9

 

 

 

 

    Low-fat diet

    Low-carbohydrate, ketogenic diet with nutritional supplements

-6.7% a

-12.9% b

6

     All participants were hyperlipidemic.

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

Table is reprinted with permission from reference (18).

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

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

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

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

 

Physical Activity Recommendations

 

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

 

PHYSICAL ACTIVITY AND WEIGHT LOSS

 

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

 

PHYSICAL ACTIVITY AND WEIGHT MAINTENANCE

 

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

 

PHYSICAL ACTIVVITY AND CARDIOVASCULAR HEALTH

 

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

 

Principles of Behavior Therapy

 

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

 

GOAL SETTING

 

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

 

SELF-MONITORING  

 

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

 

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

 

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

 

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

 

STIMULUS CONTROL

 

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

 

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

 

COGNITIVE STRATEGIES

 

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

 

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

 

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

 

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

 

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

 

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

 

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

Component

Weight Loss

Weight-loss Maintenance

Counseling

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

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

Diet

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

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

Physical activity

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

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

Behavioral therapy

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

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

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

 

REMOTELY-DELIVERED LIFESTYLE MODIFICATION INTERVENTIONS

 

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

 

Telehealth Delivery

 

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

 

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

 

Digital Delivery via the Internet or Smartphone

 

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

 

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

 

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

 

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

 

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

 

CONCLUSION

 

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

 

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

 

REFERENCES

 

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

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

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

 

 

Islet Transplantation

ABSTRACT

 

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

 

INTRODUCTION

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

 

WHEN TO CONSIDER ISLET TRANSPLANTATION?

 

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

 

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

 

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

 

Table 1.  Indication for Islet Transplantation

Condition

Procedure

Type of Transplant

Diabetes Mellitus

Type 1

ITA, SIK, IAK

Allogeneic

Type 2

ITA, SIK, IAK

Allogeneic

Surgery-Induced Diabetes
(Iatrogenic)

Chronic pancreatitis

ITA

Autologous/Allogeneic

Trauma

ITA

Autologous/Allogeneic

Multi-visceral transplantation

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

Allogeneic

Cystic Fibrosis

ITA

Lung-Islet Transplantation

Autologous/Allogeneic

Allogeneic

Benign enucleable tumors

ITA

Autologous

Borderline/malignant pancreatic neoplasms

ITA

Autologous/Allogeneic

 

Grade C pancreatic fistula requiring completion pancreatectomy

ITA

Autologous/Allogeneic

 

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

 

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

 

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

Inclusion Criteria:

·                Mentally stable and able to comply with study procedures

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

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

·                Involvement of intensive diabetes management, defined as:

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

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

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

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

·                Reduced awareness of hypoglycemia

 

Exclusion Criteria:

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

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

·                HbA1c >10%

·                Untreated proliferative diabetic retinopathy

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

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

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

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

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

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

·                Negative for Epstein-Barr virus by IgG determination

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

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

·                Known active alcohol or substance abuse

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

·                History of Factor V deficiency

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

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

o        Heart attack within the last 6 months

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

o        Left ventricular ejection fraction <30%

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

·                Symptomatic cholecystolithiasis

·                Acute or chronic pancreatitis

·                Symptomatic peptic ulcer disease

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

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

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

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

·                Use of any study medications within the past 4 weeks

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

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

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

o        A previous islet transplant.

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

 

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

 

MULTIDISCIPLINARY TEAM

 

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

 

ISLET ISOLATION AND TRANSPLANTATION

 

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

 

THE CONSORTIUM CONCEPT

 

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

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

 

ISLET TRANSPLANT ACTIVITY

 

The Collaborative Islet Transplant Registry (CITR)

 

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

 

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

 

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

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

 

Outside The Collaborative Islet Transplant Registry (CITR)

 

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

 

Clinical Management of Islet Transplant Recipients

 

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

 

Immunosuppression

 

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

 

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

 

Antibiotic and Antiviral Prophylaxis

 

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

 

Thromboembolism Prophylaxis

 

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

 

Peri-transplant Insulin Management

 

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

 

POST-TRANSPLANT CLINICAL MONITORING

 

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

 

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

 

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

 

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

 

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

 

MONITORING ISLET GRAFT FUNCTION

 

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

 

Table 3.  Monitoring of Islet Graft Function

Standard

Stimulation

Indices

Glycosylated Hb (A1c)

Fasting glycemia

Postprandial glycemia

MAGE*

CGMS*

Basal C-peptide

Daily insulin requirement

 

Mixed Meal

Intravenous glucose

Intravenous arginine

Hypo score

Liability index

Βeta score

Beta 2 score

Basal C-peptide/Glucose ratio

HOMA-B*

HOMA-IR*

TEF*

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

 

The Igls Score

 

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

 

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

 

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

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

 

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

 

Table 4. Proposed Igls Criteria 2.0

Treatment outcome

Glycemic control

Hypoglycemia

Treatment success

 

HbA1c, % (mmol/mol)a

CGM, % time-in-range

Severe hypoglycemia, events per y

CGM, % time < 54 mg/dl (3.0 mmol/L)

 

Optimal

≤6.5 (48)

≥80

None

0

Yes

Good

<7.0 (53)

≥70

None

<1

Yes

Marginal

≤Baseline

>Baseline

<Baselineb

<Baseline

Noc

Failure

~Baseline

~Baseline

~Baselined

~Baseline

No

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

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

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

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

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

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

 

Table 5. Proposed Igls Criteria 2.0

β-cell graft functione

C-peptide, ng/mL (nmol/L) f

Insulin use or noninsulin antihyperglycemic therapy

Optimal

Any

None

Good

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

Any

Marginal

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

Any

Failure

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

Any

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

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

 

IMPACT OF ISLET TRANSPLANTATION ON METABOLIC CONTROL (TABLE 6)

 

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

 

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

 

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

 

IMPACT OF ISLET TRANSPLANTATION ON DIABETES COMPLICATIONS

 

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

 

Table 6.  Benefits of Islet Transplantation

Metabolic control

-                Reduction of exogenous insulin requirements or insulin independence

-                Reduction of MAGE

-                Reduction or normalization of A1c

-                Absence of severe hypoglycemia

Quality of Life

-                Reduced fear of hypoglycemia

-                Improvement of Diabetes Quality of Life

Diabetes complications

-                Improvement of micro- and micro-angiopathy

-                Improvement of cardiovascular and endothelial function

-                Reduced incidence of acute cardiovascular events

-                Reduced nephropathy progression

-                Stabilization/slower neuropathy progression

-                Stabilization/slower retinopathy progression

 

COMMON ADVERSE EVENTS AND THEIR MANAGEMENT (TABLE 7)

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

 

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

 

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

 

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

 

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

 

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

 

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

 

Table 7.  Most Frequent Complications in Islet Transplant Recipients

Procedure-related

-                Hemorrhage

-                Portal thrombosis

-                Transient transaminitis

Immunosuppression-related

Hematological

-                Anemia

-                Leucopenia

-                Neutropenia

Metabolic

-                Dyslipidemia

Gastrointestinal

-                Oral ulcers (Sirolimus)

-                Diarrhea (Mycophenolic acid)

-                CMV colitis

Respiratory tract

-                Upper respiratory infections

-                Interstitial pneumonitis (Sirolimus)

Neurological

-                Neurotoxicity (Tacrolimus)

Genitourinary

-                Urinary infections

-                Ovarian cysts

-                Dysmenorrhea

-                Nephropathy

-                Proteinuria

Cutaneous

-                Infections

-                Cancer

 

CURRENT CHALLENGES

 

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

 

Table 8. Current Challenges Faced for Islet Transplantation

Challenge

Possible impact

Potential solutions

Progressive graft dysfunction

Reintroduction of exogenous insulin;

Destabilization of metabolic control;

Supplemental islet transplant.

Incretin mimetics;

Alternative islet implantation sites;

Novel immunosuppressive protocols.

Multiple islet donors

Increased operational costs;

Shortage of deceased donor pancreata for transplantation;

Risk of allosensitization.

Improved donor selection criteria;

Optimized cell processing;

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

Alternative implantation sites.

Chronic immunosuppression

Systemic toxicity;

Increased risk of opportunistic infections;

Islet cell toxicity.

Use of biologics;

Immune isolation techniques; Development of immune tolerance inducing protocols.

Allosensitization

Reduced graft survival;

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

Maximizing the success rate of single donor islet transplantation;

Alternative sources of transplantable tissue;

Immune isolation;

Plasmapheresis / depletion of alloantibodies;

Novel immunosuppressive protocols;

Development of immune tolerance inducing protocols.

Cumbersome graft monitoring

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

Liver needle biopsies do not provide adequate graft tissue;

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

Improved simple metabolic measures predictive of graft dysfunction;

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

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

 

FUTURE DEVELOPMENTS IN BETA-CELL REPLACEMENT THERAPIES

 

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

 

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

 

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

 

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

 

CONCLUSIONS

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

 

ACKNOWLEDGMENTS

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

 

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

 

ONLINE RESOURCES ON THE SUBJECT

 

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

 

 

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

 

REFERENCES

 

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