Archives

Aggressive Pituitary Tumors and Pituitary Carcinomas

ABSTRACT  

Aggressive pituitary tumors (APT) refer to pituitary adenomas exhibiting radiological invasiveness and an unusually rapid tumor growth rate, or clinically relevant tumor growth despite optimal standard treatments, with abandonment of the previous term ‘atypical pituitary adenoma’. Pituitary carcinomas (PC) are defined by non-contiguous craniospinal or distant metastasis. Whilst PC is exceedingly rare, comprising only 0.1-0.4% of all pituitary neoplasms, APT may account for up to 15% of all pituitary neoplasms, depending on the definition used. Typically evolving from a known pituitary macroadenomas, APT/PC is most commonly diagnosed in the fifth decade of life with corticotroph and lactotroph neoplasms predominating. Diagnosis relies on MRI, hormonal studies, and histological assessment including proliferative markers and immunohistochemistry for pituitary hormones and, most recently, transcription factors. Structural and molecular mechanisms have been proposed in the pathogenesis of APT/PC, although there appears to be no contribution from known familial pituitary tumor syndrome genes such as MEN1. Treatment is multimodal, ideally delivered by an expert team with a high-volume caseload. Surgical resection may be performed with the aim of either gross total resection or tumor debulking. Radiotherapy may be administered either as fractionated external beam radiation or stereotactic radiosurgery. Standard pituitary medical therapies such as somatostatin analogues have limited efficacy in APT/PC, whereas temozolomide yields a clear survival benefit. Evidence is emerging for the use of peptide receptor radionuclide therapy, tyrosine kinase inhibitors, VEGF inhibitors, and immunotherapy. Avenues for further research in APT/PC include molecular biomarkers, nuclear imaging, establishment of an international register, and routine pituitary tumor biobanking.

INTRODUCTION AND DEFINITIONS

Pituitary adenomas (PA) are benign, typically slow-growing neoplasms originating from cells of the adenohypophysis (1). The 2018 European Society of Endocrinology (ESE) guidelines on APT/PC (2) define aggressive pituitary tumors (APT) as PAs that demonstrate radiological invasiveness and an unusually rapid tumor growth rate, or clinically relevant tumor growth despite optimal standard treatments in the form of surgery, radiotherapy, and conventional medical therapies (2). In the absence of reliable histopathological predictors of tumor behavior (2), APTs lack specific diagnostic criteria and are instead best considered a clinical composite of various pituitary neoplasms exhibiting clinically aggressive behavior.

Efforts should be made to be as objective as possible in diagnosing APTs (2). As a follow-up to the ESE guidelines, Raverot and colleagues note the lack of supporting data but draw upon their local experience to provide some guidance in the assessment of tumor growth. The authors outline that clinically relevant growth may be evidenced by an increase in maximum tumor diameter by more than 20% or tumor growth that produces new signs or symptoms or where such signs and symptoms are predicted based on tumor location. The authors define rapid tumor growth as a 20% increase in less than 6 months, or less than 12 months if only annual MRIs are available (3). In assessing tumor refractoriness to treatment, postoperative recurrences should only be considered to be APTs when surgery was performed by an expert neurosurgeon with a high-volume caseload (2). Whether resection was intentionally limited because of reduced surgical fitness may also need to be considered. Intrinsic tumor resistance to medical therapy should be distinguished from inadequate dosing or poor compliance as well as drug toxicity, which is increasingly recognized in the setting of dopamine agonist (DA) treated prolactinomas (4). Progression or recurrence after radiotherapy is more compelling than resistance to only surgical and medical therapy (5-7).

APT is distinct from the term ‘atypical pituitary adenoma’, which was defined by the earlier 2004 World Health Organization (WHO) classification of endocrine tumors as PAs with a Ki-67 labelling index >3%, an elevated mitotic index, and extensive p53 nuclear immunostaining (8). The Ki-67 labelling index is assessed by MIB-1 immunohistochemistry (IHC). As Ki-67 is a nuclear protein with suggested roles in ribosomal RNA transcription and chromosome separation (9), MIB-1 stains cells in the S (DNA synthesis) phase of the cell cycle and thereby represents the rate of cellular proliferation (1). Immunopositivity for p53 reflects nuclear accumulation of p53 (9). In other tumors, p53 immunostaining is related to TP53 mutations that prolong the half-life of p53 and result in nuclear accumulation, although TP53 mutations are rare in p53-immunopositive pituitary neoplasms (10). A threshold of >2 mitoses per 10 HPF was also included in the 2004 WHO classification as this portends a greater risk of recurrence (2). The intent of the 2004 WHO framework was to identify more aggressive tumors warranting more intensive management and follow-up. However, the term ‘atypical pituitary adenoma’ was omitted in the 2017 WHO classification of endocrine tumors as these criteria have not been clinically validated (2, 11, 12). Although some data show correlations between the 2004 WHO criteria and tumor behavior, the criteria do not consistently and independently predict tumor behavior in individual patients, with one study showing no difference in recurrence risk and disease-free survival in atypical PAs versus other PAs (13).

Although most APTs are invasive (14, 15), invasiveness alone is insufficient to define APTs (2), partly because invasion is often subjective with variability between radiological, operative, and histological assessments (11). Moreover, highly invasive prolactinomas may respond well to DA therapy rather than following an aggressive clinical course. However, invasiveness is still considered a key component of aggressiveness. This was demonstrated by Trouillas et al in their 8-year follow-up study classifying tumors into 5 tiers: grade 1a, non-invasive and non-proliferative; 1b, non-invasive but proliferative; 2a, invasive but non-proliferative; 2b, invasive and proliferative; and 3, metastatic (15). Proliferation was defined as at least two of: Ki-67 ≥3%; p53 staining with >10 strongly positive nuclei/10 HPF; or mitotic count >2/10 HPF. Invasion was based on radiological or histological findings. Trouillas et alshowed that, compared to non-invasive and non-proliferative PAs (grade 1a), the relative risk of persistent disease was 3.1 for non-invasive but proliferative PAs (grade 1b) versus 8.0 for invasive but non-proliferative PAs (grade 2a) (15). The impact of invasiveness and proliferation on tumor aggression was synergistic. Invasive and proliferative PAs (grade 2b) carried a 25-fold higher risk of persistent disease and a 12-fold higher risk of tumor progression compared to non-invasive and non-proliferative PAs (15). Progression-free survival is more influenced by the effect of invasiveness than that of proliferation (16), with a relatively greater prognostic effect from invasiveness in lactotroph and corticotroph PAs than other subtypes (15).  

At the extreme end of the spectrum, pituitary carcinoma (PC) is defined by non-contiguous craniospinal or distant metastasis. It should be noted that there are no histopathological features that distinguish carcinomas from aggressive adenomas without metastasis. PC remains a distinct category in the 2017 WHO classification (8).

EPIDEMIOLOGY 

Because of the lack of definitive diagnostic criteria, the prevalence of APT is unclear (2). A study incorporating radiological and histological assessments of aggressiveness found ‘grade 2b’ (invasive and proliferative) tumors in 15% of patients, although this was not a consecutive series with many patients excluded due to insufficient data and others selected to balance patients with and without persistent disease (15). Aggressiveness was also not invariable in grade 2b tumors (17). Tumor recurrence and persistence, which are generally representative of APT, are more frequently seen in younger rather than older adults (15, 18, 19). PAs are overall uncommon in children but tend to be more aggressive in the pediatric setting, with 26% of prolactinomas demonstrating DA resistance (20). Some (18) but not all data (16, 19) show greater risks of recurrence and progression with larger PAs. As highlighted in Table 1, APT/PC development is more likely in certain tumor subtypes, namely, silent corticotroph PA, Crooke’s cell PA, plurihormonal PIT-1 positive PA (formerly ‘silent subtype 3 PA’), sparsely-granulated somatotroph PA, and lactotroph macroadenomas in men (11, 21-25).

Table 1. PA Subtypes with Greater Tendency for APT/PC Development (11, 12)

PA subtype

Cell lineage

Transcription factor

Hormone

Cytokeratin pattern

Prevalence of APT/PC

Crooke’s cell PA

Corticotroph

T-PIT

ACTH

Ring-like (perinuclear hyaline bodies)

Recurrence in 60%, multiple recurrence in 24%, APT/PC-related mortality in 12% (23)

Silent corticotroph PA

Corticotroph

T-PIT

ACTH

Diffuse

Multiple recurrences in 57% of recurrent silent corticotroph PA vs. 3% in other non-functioning PA (P=0.001) (22)

Plurihormonal PIT-1 positive PA (previously silent subtype 3 PA)

Acidophilic

PIT-1

GH, PRL, beta-TSH +/- alpha-subunit

Nil

Postoperative residual in 65% with tumor progression in 53% of these patients (21); greater propensity for invasion and recurrence (11)

Sparsely granulated somatotroph PA

Acidophilic

PIT-1

GH +/- PRL

Dot-like (fibrous bodies)

Higher frequency of suprasellar extension/cavernous sinus invasion, larger tumors and smaller octreotide suppression test response in sparsely granulated vs. densely granulated tumors (24)

Lactotroph macroadenoma in men

Acidophilic

PIT-1

ER-alpha

PRL (+GH in acidophilic stem cell subtype)

Nil (or fibrous bodies in acidophilic stem cell subtype)

Complete DA resistance in 8% men vs. 4% women (25); 57% of DA-resistant lactotroph PAs occur in men (25)

 

PC is rare, comprising only 0.1-0.4% of pituitary neoplasms (14, 26, 27). The incidence of PC is 4/1,000,000 person-years (28). These figures may, however, be underestimated, as up to 75% of historical PC diagnoses were only made at autopsy (29). PC typically presents in the fourth to sixth decades of life, with a mean age at diagnosis of 44 years (1, 30), but rare pediatric cases have been reported (31, 32). Whereas clinically-silent, hormone-staining tumors only account for 7% of all pituitary neoplasms (26), functioning tumors that have evolved from such tumors comprise 25% of APT/PC  (33). The commonest PC subtypes are corticotroph and lactotroph neoplasms (14, 30). In a recent review of 72 published PC cases by Yoo et al, hormone IHC was positive for ACTH in 35%, PRL in 24%, GH in 14%, TSH in 6%, FSH in 7% and LH in 4%, and 15% were null cell (34). This rate of null cell PC was lower than other reports of 30% (14, 30), possibly relating to the limited availability of prolactin IHC in historical investigations (35). Compared to pure PA series, lactotroph and corticotroph-derived neoplasms are overrepresented whilst somatotroph and null cell neoplasms are underrepresented in PC (26, 33).

The overall epidemiology of APT/PC was recently outlined in an ESE survey of clinicians treating APT/PC patients, where APT was defined by the responding clinician. The survey cohort comprised 165 patients (40 PC, 125 APT), forming the largest APT/PC cohort to date. APT and PC cases were similar in age at diagnosis (43 vs. 45 yr), predominant cell subtypes (corticotroph in 45% vs. 48%, lactotroph in 20% vs. 38%), and functional status (clinically functioning in 58% vs. 63%), but initially silent and later functioning tumors were overrepresented in PC (7% vs. 20%) (33). Both APT and PC cases demonstrated a male predilection (65% vs. 63%) (33), in agreement with Yoo et al (34), but in conflict with other data showing a slight female predominance (27, 36) or no gender predilection (14, 30).

GENETICS

Little is known about germline and somatic genetic contributors to APT/PC formation. Clinically relevant germline variants in pituitary tumorigenesis genes are found in up to 20% of PA patients who are young and/or have other personal or family history of endocrine neoplasia (37). By contrast, the rate of germline mutations specifically in the APT/PC setting is yet to be determined, although germline AIP and SDHx mutations are typically associated with more aggressive tumor behavior (9, 37). PC has been reported in patients with germline mutations, including SDHB(38), MSH2 (39), and MEN1 (40, 41). However, PC appears to be no more common in patients with germline MEN1mutations than in patients with sporadic PAs (42). To the best of our knowledge, there have been no reports of PC in patients with AIP-associated familial isolated pituitary adenoma syndrome, multiple endocrine neoplasia syndrome type 4 due to CDKN1B mutations, Carney’s complex due to PRKAR1A mutations, McCune-Albright syndrome due to GNAS mosaicism, or X-linked acrogigantism due to Xq26.3 microduplications involving GPR101.

A novel somatic role for the ATRX gene in APT/PC pathogenesis was shown in a recent study of 30 APTs and 18 PCs which revealed loss of ATRX immunolabelling and somatic inactivating ATRX variants in 9/48 cases (19%) (43). ATRX loss was especially prevalent in PCs compared to APTs, and in corticotroph neoplasms compared to other cell lineages (43).

TP53 has also been raised as a somatic contributor to APT/PC pathogenesis. Uzilov et al showed correlations between aggressive corticotroph tumours and somatic TP53 variants in a sample of 27 corticotrophinomas that was enriched for tumors at risk for aggressive behavior (44). This study also demonstrated an association between invasiveness, macroadenomas, and somatic aneuploidy independent of TP53 status (44). However, a separate study of 134 pituitary neoplasms found no association between somatic chromosomal alterations and aggressiveness (45).

Somatic gain-of-function variants in the USP8 gene have been implicated in Cushing’s disease; however, the contribution of USP8 to corticotroph APT/PC is unclear. Early data showed USP8-mutated corticotrophinomas to be smaller with lower plasma ACTH levels (46), suggesting a milder phenotype. By contrast, subsequent data have shown higher postoperative urinary free cortisol levels in patients with USP8-mutated corticotrophinomas compared to wild-type corticotrophinomas, possibly serving as a harbinger for poorer long-term outcomes in these patients (47). Uzilov et al found mixed results regarding the aggressiveness of USP8-mutated tumors, with microadenomas in 4/5 USP8-mutated tumors but a trend towards a higher number of trans-sphenoidal operations in USP8-mutated versus USP8-wild-type tumors (44).

CLINICAL PRESENTATION

APT/PC nearly always evolve from pituitary macroadenomas (maximal tumor diameter ≥1cm) (14), but, conversely, many macroadenomas and even giant prolactinomas (≥4cm) respond well to standard treatments and never exhibit aggressiveness (2). Progression of a microadenoma (<1cm) to PC is exceedingly rare (48, 49). The time from primary diagnosis with a pituitary neoplasm to presentation with APT/PC is highly variable (2). In APT, aggressiveness can be apparent from diagnosis, or take months to more than a decade to develop (2, 50). The course of APTs may be punctuated by periods of radiological and hormonal quiescence (51). One study showed that APTs are more likely to occur following incomplete surgical resection at an odds ratio of 6.3 (18), but another study showed no relationship between APTs and the primary surgical outcome (16). These conflicting results partly reflect the difficulty in distinguishing residual tumor from normal tissue and postoperative changes (18). In PC, the mean latency from primary diagnosis is 5-9 years, but can range from months to 43 years (1, 6, 14, 30, 34, 36, 52-54).

 

Some symptoms, such as headache and visual field loss, overlap between PA and APT/PC, whilst cranial nerve palsies and obstructive hydrocephalus are more suspicious for APT/PC (36). Patients with Nelson’s syndrome, which is an inherently aggressive neoplasm, often present with mass effects including cranial neuropathies from the growing primary tumor as well as hyperpigmentation from proopiomelanocortin excess; distant metastasis may also occur (55). As in PA, diabetes insipidus is rare in APT/PC (56), and should raise suspicion for sella metastasis from a non-pituitary malignancy (1). Important differential diagnoses are breast and lung carcinomas, which are the commonest primary neoplasms to metastasize to the sella (52). Small cell lung cancer can produce both ectopic ACTH syndrome and sella metastasis, which may be misdiagnosed as a corticotroph PC with distant metastasis (35). PC metastases may lead to other site-specific clinical features, such as hearing loss, ataxia, motor weakness, back pain, neck masses, and liver function derangement (1, 9).

 

Yoo et al showed the site of metastases to be craniospinal in 58%, systemic in 32%, and both craniospinal and systemic in 8% of PC cases (34). This is in contrast to an earlier series of 15 PC cases reported by Pernicone et alwhere metastasis was predominantly systemic (47%), compared to craniospinal metastases in 40% and both craniospinal and distant metastases in 13% (14). Common sites of metastasis include the brain (43%), spine (38%), liver (14%) cervical lymph nodes (11%) and bone (10%) (34). Within the CNS, metastases typically involve the cortex, cerebellum and cerebellopontine angle  (56). Dural metastases may occur and can be misdiagnosed as meningiomas (36). Rare metastatic sites include the orbit, endolymphatic sac, oropharynx, heart, pancreas, kidney, skin, ovary, myometrium and pelvic lymph nodes (1, 14, 34, 36).

 

PC subtype may influence the pattern of metastasis. In lactotroph PC compared to corticotroph PC, systemic metastases are relatively more common (71% vs. 57%), and the duration of pituitary neoplasm diagnosis to PC diagnosis is shorter (4.7 vs. 9.5 years). In patients with distant metastases, the commonest site is bone in lactotroph PC and liver in corticotroph PC (14).

 

EVALUATION

 

The principles of APT/PC assessment are outlined in the 2018 ESE guidelines (2). As in PA, the evaluation of patients with suspected or known APT/PC involves radiological, biochemical and histological investigations. Patients with APT should be followed indefinitely as recurrence and progression accumulate with time. In a study of recurrent non-functioning pituitary adenoma (NFPA), the prevalence of recurrent disease rose from 4.4% at 5 years to 10% at 10 years (6). Long-term follow-up also allows monitoring of late treatment-related complications such as radiation-induced hypopituitarism and secondary tumors, and the late development of PC which may occur decades following the initial diagnosis (2). Clinicians should be especially vigilant for metastases in patients with APTs (53), noting that metastasis often occurs insidiously and can involve various craniospinal and distant sites which may be mistakenly attributed to another primary neoplasm (2).

 

Radiological Assessment

 

The primary imaging modality in all pituitary neoplasms is MRI, ideally with thin (2-3 mm) T1- and T2-weighted slices before and after gadolinium in sagittal and coronal planes (2). T2 sequences are particularly helpful in acromegaly as T2 hyperintensity compared to normal pituitary or grey matter is often seen in sparsely granulated somatotroph PAs which tend to behave aggressively. T2 hyperintensity is also directly correlated with larger somatotroph tumors and blunted octreotide suppression test responses (57). This radiological clue is particularly helpful preoperatively, when the granulation pattern is unknown (57).

 

The Knosp criteria which grades the relationship between tumor and the cavernous portions of the internal carotid artery on MRI can be used to identify overall radiological invasion (3), noting, however, that the criteria were originally developed specifically to predict the intraoperative finding of cavernous sinus invasion (58). PAs are generally considered radiologically invasive if the Knosp grade on MRI is 3 or 4 as this correlates with surgically confirmed cavernous sinus invasion in 38% and 100% cases, respectively, compared to 1% for grade 1 and 10% for grade 2 tumors (59).

 

Serial MRI should be performed every 3-12 months as guided by previous growth rates, proximity to vital structures and timing of interventions (2). Current images should be compared against baseline and penultimate scans (1, 2). Although pituitary tumors are often irregularly shaped, comparison of the longest diameter in a 1D approach correlates well with 3D estimates (60). Pituitary tumor cases that may still benefit from volumetric analysis include multiloculated or cystic adenomas, small tumor remnants or recurrences, and multifocal and bony invasive adenomas (60). Growth rates should also take into account the PA subtype. In NFPA, volume doubling time is highly variable, ranging from 1 to 27 years, but tends to be stable for a given individual with an initially exponential growth pattern followed by deceleration of growth velocity (61). Deviation from this with unusually rapid growth rates are an important marker of APT (2). Rapid corticotrophinoma growth following bilateral adrenalectomy is a specific hallmark of Nelson’s syndrome, which precedes metastasis in over two-thirds of corticotroph PC cases (1).

 

Patients with APT and either discordant biochemical and radiological findings or site-specific symptoms should be screened for metastasis (2). In the absence of a formal staging system, patients with identified metastatic disease should undergo imaging by one or more modalities to define the extent of metastasis and to evaluate the possibility of a non-pituitary primary neoplasm (56). In patients with pituitary neoplasms and CNS symptoms, neck masses or back pain, pituitary MRI may be extended to include the whole brain and/or spine (1). CT imaging may be useful if bony involvement is suspected or in patients with contraindications to MRI (2). As PC is often hypermetabolic with somatostatin receptor (SSTR) expression including SSTR1, SSTR5 and SSTR2, nuclear imaging with 18FDG-PET and/or 68Ga-DOTATATE-PET may be valuable in delineating the overall extent of disease (53, 62). DOTATATE-PET and FDG-PET may produce discordant but useful findings. For example, the presence of uptake on FDG-PET but not on DOTATATE-PET may indicate more dedifferentiated disease. Discordant avidity may be used to guide the selection of peptide receptor radionuclide therapy (PRRT) versus chemotherapy (2, 62).  

 

Biochemical Assessment

 

Pituitary hormones should be measured every 3-12 months, as guided by tumor subtype, clinical features, and treatment interventions (2). This is imperative to identify secretory tumors responsive to medical therapies and hypopituitarism requiring hormone replacement (2). Hormone levels are also an invaluable tumor marker to guide treatment response in secretory tumors. Transition to APT/PC may be heralded by conversion of a silent PA to a clinically functioning tumor, loss of response to medical therapies, new or progressive hypopituitarism, or increasing hormonal excess despite radiological stability (1, 2, 9, 35). In particular, an initial response to DA therapy followed by ‘escape’ was documented by Pernicone et al in 4/7 (57%) lactotroph PCs (14). Decreased hormone synthesis, reflecting tumor dedifferentiation, may also be a sign of tumor progression with declining serum levels of TSH and alpha-subunit reported at the time of metastasis in a thyrotroph PC (63). Another case report described a primary FSH-staining PA followed 15 years later by metastatic disease that stained negative for all pituitary hormones (64). This notion of tumor dedifferentiation may account for the increased aggressiveness of silent corticotrophinomas compared to functioning corticotrophinomas (36).

 

Histological Assessment

 

ASSESSMENT OF PROLIFERATION

 

Despite abandonment of the 2004 WHO criteria for atypical PA and the lack of a pituitary neoplasm grading system in the 2017 WHO classification, histopathology may be incorporated with clinical features to predict the trajectory of pituitary neoplasms (1, 11, 12).

 

The 2018 ESE guidelines recommend performing IHC to evaluate pituitary hormones and the Ki-67 index, at a minimum, in all pituitary neoplasms, with the addition of mitotic count and p53 IHC when Ki-67 is ≥3%; however, it is ceded that the evidence basis for this is very low (2). The ESE guidelines suggest incorporating these histological markers in management decisions, such as the intensity of follow-up regimens and the use of adjuvant radiotherapy in patients with invasive and proliferative postoperative tumor remnants (2). The dominance of Ki-67 in the ESE guidelines reiterates the finding of a Ki-67 index ≥3% being the commonest histological marker of tumor aggressiveness in the recent ESE survey, with this threshold met in 81% of APT and 85% of PC, compared to p53 positivity in 73% APT and 78% PC and mitotic count >2/10 HPF in 63% APT and 90% PC (33). Ki-67 was also the only predictive marker for tumor aggressiveness in other studies comparing various histological and clinical markers (27, 65, 66). Ki-67 thresholds of ≥3% and >10% are considered by some experts to indicate APT and PC, respectively (2). However, this is based on limited studies with variable methodologies and a lack of robust long-term data (2). Ki-67 also overlaps between indolent PA, APT and PC. Ki-67 ranges from undetectable to 80% in PC (1, 33), and a Ki-67 ≥10% did not discriminate between APT and PC in the ESE survey (33). A mitotic index set at ≥2/10 HPF predicts a greater risk of recurrence (67), but there was again significant overlap between APT and PC in the ESE survey (33). Similarly, p53 immunopositivity, generally defined as >10 strongly positive nuclei per 10 HPF (2), is overrepresented in PC compared to PA, and incremental p53 staining has been observed in the progression from PA to PC (1), but p53 IHC may be negative in PC (68). Even the combination of all three histological markers of proliferation in the ESE survey did not reach statistical significance in differentiating APT versus PC (33). The unreliability of histological markers in predicting tumor behavior probably represents a combination of true biological variability between tumors given the observed variability in clinical features, as well as hampered histological assessment due to intratumoral heterogeneity, different fixation protocols, prior treatment effects, and antibody and interobserver variability (1, 56).

 

The European Pituitary Pathology Group recently proposed a detailed approach to the histopathological reporting of pituitary tumors (69). As in the ESE guidelines, this proposal recommends Ki-67 IHC in all pituitary tumors, but it also recommends assessing mitotic count (69). In regards to defining APTs, the European Pituitary Pathology Group endorsed the use of the 5-tier classification system developed by Trouillas et al that incorporates both histological/radiological invasion and measures of proliferation (15), on the basis of subsequent validation data supporting this system (16, 70, 71).

 

IHC for O(6)-methylguanine DNA methyltransferase (MGMT) should be considered in suspected or known APT/PC as low expression is another potential marker of aggressive behavior and is predictive of temozolomide (TMZ) response; however, these associations are not invariable and the decision to use TMZ should not rest on this result alone (33).

 

ASSESSMENT OF CELL LINEAGE

 

Histopathological evaluation is important in identifying the more aggressive subtypes of pituitary neoplasms (Table 1). Hormone IHC is critical in identifying silent corticotroph PAs and plurihormonal PIT-1 positive PA, whilst cytokeratin staining is used to define the dot-like fibrous bodies of sparsely granulated somatotroph PAs as well as patterns specific to Crooke’s cell and silent corticotroph PAs (1). Other histological features of sparsely granulated somatotroph PAs include poorly cohesive cells with sheet-like formation and nuclear polymorphism with weak and focal GH staining (24).

 

Although transcription factor IHC, as recommended in the 2017 WHO classification (12), may assist identification of aggressive pituitary neoplasm subtypes, it does not directly predict aggressiveness (2). Transcription factor IHC is considered most valuable in the differentiation of hormone immunonegative tumors (9). For example, an IHC study including 119 hormone-negative PAs found that over one-quarter of hormone-negative tumours were in fact silent corticotrophinomas based on positive T-PIT staining (72). IHC for T-PIT is attractive given the greater aggressiveness of corticotrophinomas (33), but the availability of reliable T-PIT antibodies has been a concern (9). Nonetheless, the addition of transcription factor IHC is an attempt to overcome the false negative, misleadingly weak or dubious results that may be encountered with hormone IHC (11). The clinical implications of a null cell adenoma that stains negative both for pituitary hormones and pituitary transcription factors (approx. 5% of hormone-negative PAs) are currently uncertain as previous literature has rarely defined transcription factor status (72).

 

Utrastructural analysis is not additive to the contemporary pathological assessment of pituitary neoplasms by morphology and IHC (11).

 

ASSESSMENT OF PITUITARY CARCINOMA

 

Like PAs, PCs appear microscopically as well-differentiated neuroendocrine tumors. PCs may demonstrate hypercellularity, nuclear pleomorphism, necrosis, hemorrhage and invasion, with all such features overlapping with PAs (1). Neuronal metaplasia may rarely occur in PC (1).

 

It is not possible to distinguish PC from PA on histological, immunohistochemical or ultrastructural grounds (1), and there is poor correlation between the histological and clinical features of PC metastases (53). The primary aim in the histological assessment of PC is instead to confirm a pituitary origin of metastases. Biopsy of apparent PC metastases is particularly important where another primary malignancy could explain the metastases, thereby influencing prognosis and management. Tissue diagnosis may be achieved by surgical biopsy or fine needle aspiration (FNA) biopsy of accessible sites such as cervical lymph nodes, liver, lung or vertebrae (52, 53, 73). Histological diagnosis based on FNA specimens should be cautious, given its divergence from pituitary histological diagnoses which are virtually always made by craniotomy or trans-sphenoidal surgical resection (52). Key differential diagnoses based on similar cytological appearances include metastasis from renal cell carcinoma, plasmacytoma/multiple myeloma, lymphoma, medullary thyroid carcinoma, and other neuroendocrine tumors (52)(53). In PC, metastatic lesions should bear cytological resemblance to the primary pituitary tumor (52, 73), noting that proliferative markers, particularly Ki-67, are often higher in metastases (14, 36, 53).

 

Immunohistochemical stains for neuroendocrine markers such as chromogranin A and synaptophysin aid in the differentiation of PC from non-pituitary neoplasms (1). Hormone and pituitary-specific transcription factor IHC may also be helpful in suspected metastases from a pituitary neoplasm (73, 74). To ensure the appropriate use of these histological investigations, the reporting pathologist should be notified of the potential for metastasis from a pituitary neoplasm and aware of the frequent latency between PA onset and PC development. The small possibility of dual concurrent metastatic malignancies should be considered where there is variability in the clinical, radiological or histological features of neoplastic lesions (52).

 

Genetic Testing

 

As there are currently only weak associations between pituitary tumorigenesis genes and development of APT/PC, genetic testing for either germline or somatic mutations should not be performed purely on the basis of APT/PC development (2). Germline genetic testing should follow the usual indications as for non-aggressive PAs (2), including young onset and other personal or family history of related neoplasms (37).

 

PATHOGENESIS

 

As APTs represent a composite of different tumor subtypes, the contributing pathogenic mechanisms are varied. Tumor persistence, recurrence and progression after surgery at least partly relate to greater invasiveness, lowering the chance of gross total resection (16). Cell-specific feedback sensitivity is also important. Resistance to medical therapy in somatotroph APTs may relate to reduced SSTR2 expression (75). The relative indolence of somatotroph PAs with apparent insensitivity of somatotrophs to loss of negative feedback during pegvisomant treatment contrasts sharply with the typically aggressive nature of Nelson’s syndrome following bilateral adrenalectomy with loss of endogenous cortisol feedback in corticotrophs (76). A somatic inactivating mutation in the glucocorticoid receptor gene was found in one such case of Nelson’s syndrome (77). On the other hand, Cushing’s disease requiring bilateral adrenalectomy may reflect intrinsically more aggressive corticotrophinomas that drive the clinical course of disease, rather than adrenalectomy and loss of endogenous negative feedback being the underlying driver of progression (2). DA resistance in lactotroph APTs has been associated with decreased dopamine D2 receptor (D2R) density, overall reduction in D2R mRNA production, and altered expression of D2R mRNA isoforms with lower expression of the more efficient short isoform (78). A somatic truncating DRD2 variant has been described in a lactotroph APT (79), but DRD2variants are not a consistent feature of lactotroph APTs (79, 80). DA resistance is also associated with cystic prolactinomas (81).

 

Hypothesized mechanisms of PC dissemination include: hematogenous spread through the anterior pituitary portal system into the cavernous and petrosal sinuses and finally the jugular veins; lymphatic spread via the sphenoid sinus or in the skull base and soft tissues by connections between the intracranial perineural space and lymphatic plexus; and cerebrospinal fluid seeding along the subarachnoid space of the neuroaxis (1, 14, 53, 56). However, there have been no studies comparing the sites of metastases in pituitary neoplasms with cavernous versus sphenoid sinus invasion. Increased matrix metalloproteinase-9 expression in PC and its association with vascular density in PC suggest that extracellular matrix degradation contributes to angiogenesis (82). Matrix metalloproteinase activity may also promote local tumor invasion, including entry into deep brain structures along the Virchow-Robin perivascular CNS spaces, resulting in non-contiguous cranial metastases (53).

 

Iatrogenesis has been purported in select PC cases with intimately located metastases following trans-sphenoidal surgery (14), craniotomy (83),  radiotherapy (53), and ventricular-peritoneal shunt placement (53). Hypotheses for the role of surgery in increasing PC risk include disruption of venous barriers intraoperatively and postoperative formation of friable new blood vessels (53). Radiotherapy has been postulated to increase tumor aggressiveness by inducing genetic mutations, in TP53 for example (55). However, this theory is controversial and confounded by the fact that surgery and radiotherapy are employed in most patients with APT/PC during the typically protracted progression of PA to APT and finally to PC (1, 53). Furthermore, the vast majority of operated and irradiated pituitary neoplasms never develop into PC (1), making iatrogenesis a highly unlikely cause of PC.

 

Molecular Mechanisms

 

Competing molecular models of APT/PC pathogenesis include a hyperplasia-adenoma-carcinoma sequence with accumulation of molecular alterations, versus clonal evolution of a subclone with genetic/epigenetic changes favoring cell survival, proliferation and ultimately metastasis (1, 53, 84). As most patients present with a long history of pituitary neoplasm (14, 35), de novo malignant transformation of normal adenohypophyseal cells seems unlikely. There are, however, rare reported cases of rapid progression from pituitary neoplasm diagnosis to death (49, 85). The frequent transition of PC from PA via an APT stage (1, 53) suggests that pathogenic mechanisms may be shared between PAs, APTs and PCs. Although, whilst some genes like PTTG are overexpressed in PAs compared to normal pituitary tissue and in APTs compared to other PAs (86), other genes such as the RAS gene only appear to be implicated in APT/PC (87, 88). Whilst there is some overlap between genetic changes in APT/PC and the genes underlying more common solid organ malignancies, mutations in classic oncogenes and tumor suppressor genes are relatively uncommon (36). Certain molecular events may be specific to the different elements of APT/PC pathogenesis. A transcriptomic analysis of lactotroph pituitary neoplasms found different genetic changes in purely invasive tumors (upregulation of ADAMTS6and CRMP1; downregulation of DCAMKL3) compared to tumors that were invasive and aggressive (upregulation of ADAMTS6, CRMP1, PTTG, ASK, CCNB1, AURKB and CENPE; downregulation of PITX1). Upregulation of Pttg, Aurkb, Cenpe and Crmp and absent Pitx1 expression in malignant lactotroph tumors in rats recapitulated these findings, and there is a functional basis to the involvement of these genes with ASK, PTTG, AURKB, CCNB1 and CENPE involved in the cell cycle, ADAMTS6 in the extracellular matrix, CRMP in cellular migration, and PITX1 in pituitary differentiation (89).

 

Copy number variation (CNV) at the chromosomal level is the most frequent genetic aberration in pituitary neoplasms (79, 90). CNV is particularly common in functioning neoplasms, especially prolactinomas, as well as neoplasms with high proliferative indices (90-92). The mean number of chromosomal imbalances per tumor is 1.6 in initial PAs, 3.4 in recurrent PAs and 8.3 in PC (91, 92). Aneuploidy was observed in all but one of the 15 PCs reported by Pernicone et al (14). The degree of genomic disruption is directly proportional to Ki-67 index (90). This progressive increase in CNV supports an adenoma-carcinoma sequence, as observed in other endocrine tumors such as pancreatic and adrenocortical neoplasms (92). Recurrent chromosomal aberrations in APT/PC include gains in chromosome 4q, 5, 13q and 14q and losses of chromosome 1p, 2, 8q, 10, 11, 12q, 13q and 15q (6, 91, 92). These chromosomes contain multiple genes implicated in APT/PC pathogenesis, as listed in Table 2, although the underlying evidence for the causal involvement of these genes is limited owing to the rarity of PC and variability in genomic technologies.

 

Table 2. Selected Genes Implicated in APT/PC Pathogenesis

Gene

Locus

Function

Alteration in APT/PC

Oncogenes

PTTG, pituitary tumor transforming gene

Chr 5q33.3*

Securin protein in spindle checkpoint machinery, responsible for error-free mitosis

Overexpression associated with increased risk of PA recurrence, strong correlation with Ki-67 (86)

VEGFA, vascular endothelial growth factor A (also referred to as VEGF)

Chr 6p21.1

Induces angiogenesis by promoting endothelial cell survival and proliferation

Increased VEGF staining in PC (93); PC stabilised by VEGF inhibition (bevacizumab) (94)

EGFR, epidermal growth factor receptor

Chr 7p11.2

Receptor tyrosine kinase contributing to tumor progression by increasing proliferation, decreasing apoptosis, and inducing angiogenesis and invasion

Increased EGFR expression in APT/PC (95)

HRAS, V-HA-RAS Harvey rat sarcoma viral oncogene homolog

Chr 11p15.5*

Promotes cellular proliferation and differentiation

Rare activating mutations in APT/PC (87, 88)

CCND1, cyclin D1

Chr 11q13.3*

Promotes transition at the G1-S phase cell cycle checkpoint

Germline CCND1 genotype associated with  locally invasive and malignant pituitary neoplasms (96);  increased CCND1 staining in APT vs. other PA and normal pituitary (97)

ERBB2, V-ERB-B2 avian erythroblastic leukemia viral oncogene homolog 2 (also referred to as HER2/neu)

Chr 17q12

Induces cell survival and proliferation

Increased expression in PC (49)

TOP2A, topoisomerase DNA II alpha

Chr 17q21.2

Enzyme modifying topological state of DNA, involved in DNA transcription and mitosis

Increased topoisomerase II alpha immunostaining in invasive PA, silent type 3 PA and PC; mixed results regarding correlation with Ki-67 (98)

Tumor Suppressor Genes

MSH6, MutS E. coli homolog of 6

Chr 2p16.3* 

Mismatch repair protein, removes DNA base mismatches caused by errors in DNA replication or by DNA damage

Loss of MSH6 in progression from atypical PA to PC, loss of MSH6 +/- MSH2 in TMZ-resistant atypical PA/PC (50, 99); inactivating MSH6mutations in PC (100)

MGMT, methylguanine-DNA methyltransferase

Chr 10q26.3* 

DNA repair enzyme, removes alkylating adducts in DNA

Decreased MGMT expression in APT/PC, correlates with activation of genes in DNA damage response and DNA repair pathways and genes involved in transcription (101)

CDKN1B, cyclin-dependent kinase inhibitor 1B (encoding p27Kip1)

Chr 12p13.1 

Binds cyclin/cyclin-dependent kinase complexes, regulates transition at the G1-S phase cell cycle checkpoint

Loss of normal p27 expression in PC (102)

RB1, retinoblastoma 1 gene

Chr 13q14.2*

Regulates cellular proliferation

RB1 loss of heterozygosity in highly invasive and malignant pituitary neoplasms (103)

TP53, tumor protein p53

Chr 17p13.1

Induces cellular senescence or apoptosis in response to DNA damage

Increasing cellular accumulation in APT/PC, rare inactivating mutations in APT (44, 55)

BCL2, B-cell CLL/lymphoma 2

Chr 18q21.33

Anti-apoptotic

Decreased Bcl-2 expression in PC, correlates with higher rates of apoptosis in PC vs. PA (104)

Other 

PTGS2, prostaglandin-endoperoxide synthase 2 (encoding COX2)

Chr 1q31.1 

Cyclo-oxygenase involved in angiogenesis

Increased Cox-2 expression in PC (105)

LGALS3, lectin galactoside-binding soluble 3 (encoding GAL3)

Chr 14q22.3* 

Galactose-binding lectin regulating cyclin-E-associated kinase activity

Increased Gal-3 immunopositivity in corticotroph and lactotroph PC (106)

HIF1A, hypoxia-inducible factor-1alpha

Chr 14q23.2*

Transcription factor mediating cellular responses to hypoxia

Increased HIF1A expression in PC (107)

ATRX, ATRX chromatin remodeler

Chr Xq21.1

Regulation of expression of a variety of genes, involved in telomere maintenance

Somatic inactivating ATRXvariants detected in APT and particularly PC, especially in corticotroph neoplasms (43)

Abbreviations: * chromosomal loci that are frequently gained or lost in APT/PC

 

A particular gene of interest in the pathogenesis of APT/PC is MGMT, which maps to 10q26.3. Low MGMT expression is a common feature in APT/PC (33, 101). It is also overrepresented in patients with plurihormonal PIT-1 positive PA, Crooke’s cell PA, Nelson’s syndrome and recurrent NFPA, all of which exhibit more aggressive behavior (101). Low MGMT expression is in turn associated with upregulation of genes involved in transcriptional activity, DNA damage response and DNA repair (101). Interestingly, low MGMT expression in pituitary neoplasms does not correlate with MGMT promoter hypermethylation as it does in glioblastoma, suggesting that MGMT is inactivated by alternative, currently unknown mechanisms (2, 7, 101).

 

Apart from the aforementioned limited associations, the conversion of PA to APT/PC does not appear to be explained by the key genes underlying sporadic (e.g. USP8, GNAS) and/or familial (e.g. AIP, MEN1, CDKN1B, PRKAR1A, SDHx) PAs (2). In a study of 52 patients with somatotroph PA, GNAS mutations were found in 53% of tumors but there was no difference between the more common densely granulated subtype and the more aggressive sparsely granulated subtype, and Ki-67 index, invasiveness and diameter did not differ between GNAS mutated and wild-type tumors (24). By contrast, a known activating GNAS mutation was reported to coincide with conversion of a lactotroph PA into a somatotroph APT (108). This suggests that the conversion to hormone production in APT/PC may sometimes relate to acquired genetic mutations with a true gain of secretory function. An alternative explanation is simply increased tumor bulk with increased hormonogenesis  (33).

 

A myriad of other molecular changes has been observed in APT/PC. As in other cancers, a role for telomerase in facilitating cellular immortality has been suggested with both Ki-67 and telomerase activity shown to increase with sequential resections of a lactotroph PC, whereas telomerase activity was absent in PAs (109). Increased immune tolerance may also be contributory with reduced T-cell concentration, HLA-B downregulation and upregulation of genes involved in T-lymphocyte suppression shown in plurihormonal PIT-1 positive PAs (110). The role of T-lymphocytes in pituitary immune tolerance is underscored by the high rates of hypophysitis with the use of ipilimumab in other malignancies (111), and the recent successful use of combined anti-CTLA4/PD1 therapy in a corticotroph PC (100). Changes have also been observed in microRNA, which are small non-coding RNAs that bind the 3’-untranslated regions of target mRNAs, thereby regulating post-transcriptional gene expression (112). In a study of lactotroph neoplasms, miR-183 was downregulated in APTs versus non-aggressive PAs and this was associated with increased expression of PCLAF, a gene inhibiting p53 and p21 mediated cell cycle arrest. miR-183 and PCLAF also correlated with Ki-67 and p53 expression (112). In a case of a non-functioning PC with multiple intracranial metastases, miR-20a, miR-106b and miR17-5p were upregulated in the metastases compared to the primary neoplasm, in association with decreases in the tumorigenesis genes, PTEN and TIMP2, which are downstream targets of these microRNAs (113). Another study showed upregulation of miRNA-122 and miRNA-493 in PC versus PA, with miRNA-493 shown to interact with the LGALS3 and RUNX2 genes which have been implicated in pituitary cellular proliferation (114).

 

MANAGEMENT

 

The key principle in the management of patients with APT/PC is for care to be directed by an expert multidisciplinary team. Multimodal treatment strategies are typically required. Surgery, radiotherapy, and medical therapies all have a role in the management of APT (Figure 1). Tumor location and size, the presence of single or widespread metastatic disease (in PC), prior surgery and extent of resection(s), previous radiotherapy and cumulative doses, optimization of standard medical therapies, past oncological treatments, and patient comorbidities are all important considerations in formulating management plans.

Figure 1. Treatment options in APT/PC

 

Surgical Management

 

Patients with APT frequently require repeated neurosurgical procedures. In the ESE survey cohort, patients underwent a mean of 2.7 operations while 29% had four or more pituitary operations over the course of their disease (33). Multiple studies now demonstrate improved outcomes and lower complication rates when pituitary surgery is performed by high-volume neurosurgeons (115-118). The likelihood of achieving gross total resection is consistently reduced in the presence of tumor invasion, particularly of the cavernous sinus, even in the most experienced hands (119, 120). Endoscopic endonasal surgical techniques utilizing angled endoscopes and wide exposure may facilitate safe and more extensive surgical resection compared with trans-sphenoidal microsurgical approaches (121-123). In some circumstances where tumor extends to a significant degree into suprasellar or other extrasellar regions, a transcranial approach may be favored. However, the degree of resection may be limited by the risk of morbidity, depending on tumor location.

 

Surgical resection, even as a debulking procedure, should be considered in patients with APT as it may offer significant relief of compressive symptoms, particularly when there is visual disturbance (124, 125). In patients with isolated metastatic deposits (either craniospinal or systemic disease) complete surgical excision may result in long-term disease-free progression particularly when followed by adjuvant radiotherapy (126-128). Repeat surgical resections of recurrent metastases may also prolong survival (14).

 

Radiotherapy

 

The use of radiotherapy should be considered in patients with APT as it may assist in long-term control of tumor growth (129). Radiotherapy is recommended in the setting of clinically significant tumor growth despite surgery, and in the case of functional tumors where standard medical therapy has been ineffective (2). In patients with PC, palliative radiotherapy may be delivered to sites of metastatic disease, but there is no evidence that it prolongs survival (129). Discussion about radiotherapy should take place within a multidisciplinary setting involving an expert radiation oncologist (2). The role of further debulking surgery prior to radiotherapy should be discussed. Radiotherapy applied to a smaller tumor volume is more effective, and removing tumor in close proximity to the optic apparatus may allow safer and improved radiotherapy delivery (2, 130). In previously irradiated patients, consideration must be given to the cumulative radiation dose applied to the target region. In patients with invasive tumor remnants following surgery andwhere histological markers indicate the potential for aggressive tumor behavior (high Ki-67, particularly ³ 10%; elevated mitotic count; increased p53 immunostaining), adjuvant radiotherapy should be considered (2). In the case of evident aggressive tumor behavior, combination radiotherapy and chemotherapy with TMZ may yield improved outcomes (33).

 

Fractionated external beam radiation therapy (EBRT) and stereotactic radiosurgery (SRS, delivered as a single dose or in fractions) are both highly effective in the management of PAs. In one study of NFPAs, routine use of postoperative radiotherapy was associated with a doubling of 10-year progression-free survival compared with patients who did not undergo radiotherapy (93% vs. 47% ) (131).  Success rates vary across studies because of different modalities (linear accelerators, Gamma Knife, proton beam irradiation) and variable techniques, doses and imaging protocols used between centers (132). In APT, there are limited data on the effectiveness of radiotherapy. In a series of 50 patients with persistent or recurrent adenomas despite prior radiotherapy, further focal SRS was effective in the majority of cases, although a large number of cases were treated for persistent GH excess rather than radiologically aggressive tumors (133, 134). The response to radiotherapy may only be transient in more aggressive tumors or even ineffective, particularly in cases demonstrating progression despite salvage chemotherapy (33).

 

The choice of radiotherapy technique and modality is ultimately based on safety considerations (e.g., proximity to the optic chiasm), volume of disease, and local center availability (2). Adverse effects of radiotherapy delivered to the pituitary, such as hypopituitarism or risk of secondary tumors, has rationalized the modern-day use of radiation therapy for pituitary tumors. However, considering the morbidity and excess mortality associated with APT, these adverse effects, particularly given their significant latency, should not preclude the prompt use of radiotherapy in APT.

 

Peptide Receptor Radionuclide Therapy

 

Pituitary neoplasms express somatostatin receptors and have demonstrated 68Ga-DOTATATE uptake on PET/CT, stimulating interest in the use of PRRT in the management of APT (135, 136). A variety of radionuclides have been utilized, including 111Indium-DPTA-octreotide, 177Lutetium-DOTATATE, 177Lutetium-DOTATOC and 90Yttrium-DOTATOC (2, 137). A recent review of 20 PRRT-treated APT/PC cases reported in the literature to date found limited success, with partial responses in 3/20 and stable disease in 3/20 (138). For unclear reasons, PRRT failure in APT/PC may be associated with previous use of TMZ (138), with only one reported case of a successful PRRT response following prior TMZ treatment (139).

 

Standard Medical Therapy

 

APTs typically display resistance to the standard medical therapies commonly used in the management of functional PAs, although dose escalation may be helpful. In lactotroph APTs, use of maximally tolerated DA treatment should be attempted given occasional reported responses (140, 141), with cabergoline (3.5-11mg per week) being more effective than bromocriptine or quinagolide (2). Temporary benefit from high-dose octreotide has been described in a case of thyrotroph PC (63).

 

Aggressive corticotroph tumors represent a particular challenge, and these patients often require medical therapy to reduce hypercortisolism, a common direct cause of death (129). Adrenal glucocorticoid inhibitors, such as ketoconazole or metyrapone, are frequently used in such cases. Pasireotide has been reported in 15 cases of aggressive corticotroph tumors, including nine with Nelson’s syndrome, but with only one case exhibiting a hormonal and radiological response (2, 142).

 

DA-resistant lactotroph APT/PCs are also challenging, with limited medical options apart from TMZ. Pasireotide responses have been recently described in three patients with lactotroph APT: a 41-year-old woman who experienced prolactin normalisation and tumor necrosis but without tumor shrinkage (143); a 61-year-old woman who experienced both prolactin normalisation and tumor shrinkage (144); and a 55-year-old woman with a giant silent prolactinoma who experienced tumor shrinkage (145). Pasireotide is likely most useful in lactotroph APTs with high SSTR5 expression; however, less than 15% of prolactinomas express SSTR5 (145). Tamoxifen has been unsuccessfully used in lactotroph PC (2).

 

In rare cases of somatotroph PC, use of DA treatment has been associated with GH and IGF-1 reductions and symptomatic improvement, but without tumor shrinkage (29, 146). Similarly, the use of first-generation somatostatin analogs in somatotroph APTs is largely ineffective, whereas pasireotide may improve biochemical control, although data in APT are scarce (147). Resistance to first-generation somatostatin anologs (octreotide, lanreotide) due to downregulation of SSTR2A expression has been described among AIP mutation positive individuals with somatotroph tumors, but expression of SSTR5 is often preserved and thus response to second-generation broader-spectrum somatostatin analogs, such as pasireotide, may be more effective (148).

 

Chemotherapy

 

WHEN TO INITIATE CHEMOTHERAPY

 

In patients with PC, the decision to start systemic chemotherapy is clear and associated with improved survival (2, 149, 150). In patients with isolated metastases, loco-regional therapies such as hepatic chemoembolization for low-bulk liver metastases may offer temporary tumor control (151). For APT, in cases of documented tumor growth, other treatment options may be explored first, such as further surgery or radiotherapy if appropriate, and histological parameters such as Ki-67 or tumor subtype, may play a role in decision making. However, it is increasingly recognized that apart from the presence of metastases, there is little that distinguishes APT from PC (17). Most importantly, time to death following diagnosis of pituitary tumor is similar between APT and PC (33). Prior to the recognition of TMZ efficacy in APT, chemotherapy was typically reserved as salvage therapy because of poor response rates. The mean survival rate in the pre-TMZ era for PC was 1.9 years (56). APT and PC treated with TMZ are now reported to have 5-year overall survival rates of 57.4% and 56.2%, respectively (152). In the large French cohort, median survival was 44 months in patients who responded to TMZ compared with 16 months in non-responders (153). While TMZ is still most commonly used as a last resort therapy, it has been successfully employed during or prior to radiotherapy (33, 154). In fact, the 2018 ESE guidelines suggest, in patients with rapid tumor growth where maximal doses of radiotherapy have not been reached, TMZ may be combined with radiotherapy as per the Stupp protocol used for glioblastoma (2, 155). As new therapeutic modalities emerge in the coming years, clinicians will likely employ TMZ earlier in the treatment algorithm for APT. Decisions must be made within an expert multidisciplinary team setting where risk-benefit ratios are carefully deliberated, taking into account the morbidities of repeated surgery or radiotherapy as well as the potential for rare long-term consequences of chemotherapy such as hematological malignancy (2). 

 

TEMOZOLOMIDE

 

TMZ is recommended as first-line chemotherapy for patients with APT and PC, with more than 200 cases now reported in the literature including the recent ESE survey (2, 33). The overall response rate is 37-47% across the larger cohorts, with complete responses (both biochemical and radiological) seen in approximately 5% of cases (2, 33). However, if stable disease is considered a clinically beneficial outcome, as it frequently is in oncological studies, then rates of progression-free survival are 50-87.6% in APT and PC (33, 150). Clinically functioning APT are 3.4 times more likely to respond to TMZ compared with non-functioning APT (33). APTs are just as likely as PCs to respond to TMZ, although progression may be more frequent among tumors with Ki-67 ³10% (33).

 

TMZ is a second-generation imidotetrazine alkylating agent which, when hydrolyzed, forms toxic methyl adducts with DNA bases resulting in ineffective DNA repair and ultimately cellular apoptosis (156). TMZ is given as an oral outpatient-based chemotherapy, most commonly as monotherapy. Some centers advocate use of capecitabine pre-treatment (CAPTEM) because of in vitro data in neuroendocrine tumor cell lines suggesting synergistic effects with this regimen, although evidence supporting its superiority in APT and PC is lacking (27, 157). Similarly, it has not yet been demonstrated that TMZ in combination with any other drug(s) has enhanced efficacy (2). However, where maximal doses of radiotherapy have not yet been reached in a patient, there is suggestion of improved response when TMZ is given concurrently with radiotherapy (2, 33, 158). Experimental data strongly support a radiosensitizing effect of TMZ (159, 160).

 

The TMZ doing regimen given for APT is 150-200mg/m2 for 5 consecutive days every 28 days. It is generally well-tolerated, although mild to moderate fatigue, nausea and myelosuppression are common side effects, occurring in roughly half of patients but leading to TMZ discontinuation only in a minority (2). Prophylactic anti-emetics such as ondansetron is recommended for the 5 days of treatment per cycle (161). A dose reduction or delay in treatment cycles can allow patients to continue TMZ when myelosuppression occurs. Hemorrhage into cerebral metastases has been described in a patient with PC who developed severe thrombocytopenia (162). Hepatoxicity has been reported when TMZ was used concurrently with ketoconazole therapy, and cholestatic hepatitis has also been reported in association with TMZ treatment in the wider literature (163, 164). To monitor for the risks of myelosuppression and hepatotoxicity, a complete hematological profile should be obtained on day 22 of each 28-day cycle and liver function tests should be performed at baseline, midway through the first cycle, prior to each subsequent cycle and within a month of treatment cessation (2). TMZ-induced hearing loss has been described among two pituitary cases and other rare side effects in non-pituitary literature include hypersensitivity pneumonitis, Stevens-Johnson syndrome and hematological malignancies (2). Prophylactic trimethoprim-sulfamethoxazole to protect against Pneumocystis pneumonia should be considered, particularly in the setting of active Cushing’s syndrome, high-dose glucocorticoid therapy, concurrent radiotherapy or significant lymphopenia (2, 161).

 

WHEN TO STOP TEMOZOLOMIDE

 

Response to TMZ will be evident after 3 months of therapy (165). Treatment should cease in the event of progressive disease while receiving TMZ, or if serious adverse events occur. It is recommended to continue with treatment for at least 6 months, but therapy is often extended if there is ongoing clinical benefit (2). In the ESE survey, median treatment duration was 9 months and the longest course was 36 months (33). In this patient cohort, treatment with TMZ was initiated prior to the publication of management guidelines for APT. Hence, duration of therapy was often prescribed by oncology teams at the outset and was based on experience with TMZ clinical trials in glioblastoma (155). Following cessation of TMZ treatment in APT/PC, there is frequently a period of sustained remission. Time to tumor progression is variable, and whether longer treatment courses or degree of initial response improves progression-free survival is not currently clear. The median time to progression after cessation across patients in the ESE survey cohort was 12 months (range 1-60). Two patients exhibiting the longest time to progression were PC cases with complete response to TMZ (33). In the French multicenter cohort, patients receiving more than 12 months of TMZ achieved a median relapse-free survival of 57 months compared with 18 months in those receiving less than 12 cycles (153). However, response rates were 100% in those receiving longer treatment courses versus 75% in the shorter treatment group. Nevertheless, long-term treatment has been reported to be associated with improved progression-free survival of 61% compared to 16% for short-term treatment (152). The potential benefits of long-term TMZ treatment are tempered by the cumulative toxicity to bone marrow, especially given the relatively long survival of patients with pituitary tumors compared to other neoplasms treated with TMZ (3).

 

DETERMINANTS OF RESPONSE TO TEMOZOLOMIDE

 

The most well recognized biomarker of the likely response to TMZ is MGMT expression. An endogenous DNA repair protein, MGMT is responsible for removal of the methyl group induced by TMZ therapy. In the absence of MGMT, unrepaired methylated guanine (O6-MeG) lesions incorrectly pair with thymine, triggering activation of the mismatch repair pathway (MMR). Intact MMR results in futile attempts at repair via incorrect reinsertion of thymine opposite the O6-MeG lesion. Cycles of ineffectual repair eventually result in DNA strand breaks which lead to cell cycle arrest followed by either apoptosis or cellular senescence. If MMR function is lost, then paradoxically cells can survive. However, even in the presence of intact MMR, MGMT can facilitate cell survival by direct repair of O6-MeG, targeting it for ubiquitination and degradation (Figure 2) (166).

 

Low expression of MGMT, as determined by IHC, is associated with a high response rate, at around 75%, while tumors with high MGMT expression are unlikely to respond (33, 156). Low MGMT expression also predicts longer survival in patients with TMZ-treated APT/PC (167).Evaluation of MGMT status by IHC should be performed by a neuropathologist with expertise in APT (2). Lack of standardized IHC technique, use of different expression criteria across centers, poor fixation methods, and tumor heterogeneity are among the challenges in assessment of MGMT IHC. MGMT promoter methylation analysis has not been associated with response to TMZ in pituitary neoplasms (156, 168).

 

DNA mismatch repair proteins such as MSH6, MLH1, MSH2 and PMS2 may also play a role in response to TMZ. Loss of MSH6, in the presence of low MGMT, has been described as a mechanism responsible for the development of resistance to TMZ (169). The overexpression of multidrug resistance proteins and activity of the Sonic hedgehog signalling pathway may also contribute to TMZ resistance (150).

Figure 2. Temozolomide Cytotoxicity and Mismatch Repair Pathway

 

TREATMENT OPTIONS BEYOND TMZ

 

There is a pressing need to identify alternative effective oncological therapies for patients progressing on TMZ or following an initial successful course of TMZ treatment. Given the paucity of treatment options, a second 3-cycle trial of TMZ treatment may be considered in patients who develop recurrence after a previous response to TMZ (2). However, a second treatment course has rarely been reported to be successful in such cases (2). In a recent review of nine detailed case reports of patients with APT receiving a second course of TMZ for at least 3 months, 4/9 patients had a partial response, 2/9 had stable disease and 3/9 had progressive disease (161). Patients with late relapses after the initial TMZ course and tumors with low MGMT status appeared to have a better response to TMZ retreatment (161).

 

If there is rapid tumor progression on TMZ treatment, a trial of other systemic cytotoxic therapy is recommended based on historical reports of transient regression and/or stabilization with some regimens (2). Lomustine (CCNU) and/or 5-fluorouracil (5FU) have most commonly been employed, but multiple other drugs, alone or in combination, including cyclophosphamide, doxorubicin, adriamycin, carboplatin/cisplatin, etoposide and vincristine have also been reported, with variable results (2, 33).

 

Use of targeted therapies offer some promise, but data on clinical effectiveness are lacking. In vitro data demonstrating upregulation of Raf/MEK/ERK and PI3K/Akt/mTOR pathways in pituitary tumors have thus far not translated into clinical success in APT (33, 170, 171), apart from a single case report of partial response to everolimus in an aggressive prolactinoma (172). There has been limited use of tyrosine kinase inhibitors (lapatinib, sunitib, erlotinib), with one case report of a lactotroph APT exhibiting a partial response to lapatanib (173) and a subsequent phase 2 trial showing stable disease in 3/4 lapatanib-treated lactotroph APTs and progressive disease in the remaining case (174). VEGF-targeted therapy with bevacizumab or apatinib, as monotherapy or in combination with TMZ, has produced mixed results from complete response to progressive disease (33, 94, 138, 175). 

 

Finally, there is evolving interest in the potential use of immunotherapy for the treatment of APT/PC. Combination treatment with ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) was reported by Lin et al to result in marked tumor shrinkage and hormonal response in a patient with a hypermutated corticotroph PC (100). Subsequently, Duhamel et al reported ipilimumab/nivolumab combined treatment with partial response in a patient with corticotroph carcinoma and progressive disease in a patient with an aggressive prolactinoma (176). Sol et al reported stable disease following ipilimumab/nivolumab combined treatment in a patient with corticotroph carcinoma after previous progression despite multiple lines of treatment including TMZ (177). Caccese et al described a poor response to pembrolizumab (anti-PD-1) with rapid disease progression in an aggressive corticotrophinoma (178). An open-label phase II trial of pembrolizumab in four patients with PC led to radiographic and hormonal responses in two patients, stable disease in one patient and progressive disease in the remaining patient (179). Emerging studies of the immune microenvironment of pituitary neoplasms highlight the potential for gene expression data to identify which APT/PC will respond best to anti-CTLA-4 versus anti-PD-1 therapies (180, 181).

 

PROGNOSIS

 

Morbidity and mortality are increased in APT even in the absence of progression to PC (2, 17). This is particularly true in functioning corticotroph APTs, where morbidity and mortality are further increased in relation to cortisol excess (2).

 

In the ESE survey, mortality was higher in PC (43%) compared to APT (28%) (17), but median survival from initial diagnosis of pituitary tumor was similar (11 years in APT vs. 12 years in PC) (33). The time to death from PC diagnosis ranged from 7 days to 8 years in the study by Pernicone et al, with a 66% 1-year-survival (14). The mortality rate reported by Yoo et al was 55%, with an average time to death after PC diagnosis of just 10 months (34). Amongst all endocrine carcinomas, PC demonstrates the strongest decline in survival with advancing age (28). Prognosis is especially poor in patients with corticotroph PC, systemic metastases, or progression during TMZ therapy (1, 14, 33, 182). By contrast, patients who respond to TMZ experience a clear survival benefit (153). Exceedingly long-term survival over several years has been observed in selected cases (6, 14, 27), even without TMZ (182), but predictive markers for such survival remain unknown (53).

 

FUTURE DIRECTIONS

 

Comprehensive molecular studies will hopefully identify better biomarkers for PAs that are destined to become APT/PC. The recent finding of somatic ATRX variants in almost one-fifth of APT/PC cases suggests ATRX immunohistochemistry may be a useful adverse prognostic marker pending further research in this area (43). In addition to molecular biomarkers, the growing sphere of nuclear medicine may prove useful in the assessment of PC, which currently lacks a standard method of staging. 11C-methionine, a tracer with specific avidity for neoplastic pituitary tissue, has shown superior sensitivity to 18F-FDG-PET in localising functioning PAs (183). Though yet to be studied in PC, 11C-methionine holds promise in better delineating metastatic disease. Integration of molecular, functional and clinical data may ultimately assist clinicians in better identifying tumors with the potential for more aggressive behavior. This will allow earlier and more proactive management in affected patients with the goal of improving prognosis.

 

Therapeutic questions requiring further investigation include the optimal duration of TMZ therapy and the utility of TMZ treatment earlier in the management of selected APT cases rather than reserving TMZ as a salvage therapy (3). Early use of TMZ may be especially valuable in preference to radiotherapy in rapidly growing APTs (161) and in preference to both surgery and radiotherapy in frail patients with comorbidities (184). Clinical trials on the use of immune checkpoint inhibitors in APT/PC are ongoing (185). Combination therapies with the inclusion of TMZ also warrant investigation (161). Based on pre-clinical data regarding the pathways of pituitary tumorigenesis, future therapeutic avenues may include treatments that target BRAF, fibroblast growth factors, Notch and hedgehog signaling, and PTTG (175).

 

Because of the rarity of PC and the diverse subtypes of APT, current data are plagued by small sample size driven by case reports or series, heterogeneous case mix, short follow-up and clinical rather than histological diagnoses of PC metastases, with heavy reliance on expert opinion and local practice and a dearth of randomized controlled trials. Calls by the ESE to form an international register for APT/PC should help address the multiple evidence gaps in these rare disorders (2). As APT/PC are almost invariably diagnosed retrospectively, routine pituitary tumor biobanking with methodical storage of tissue in media that circumvent formalin-induced DNA damage will be critical in studying pathogenesis. Waiting for metastasis before labelling a pituitary neoplasm as PC is particularly problematic, given the similar time-to-death from initial pituitary tumor diagnosis between patients with APT versus PC (17). Increasing use of the term ‘pituitary neuroendocrine tumor’ (PitNET) in favour of ‘pituitary adenoma’, as proposed by the International Pituitary Pathology Club and endorsed by the European Pituitary Pathology Group, is hoped to emphasize the malignant potential of a subset of these neoplasms and expand treatment intensity (9, 69, 186); however, as with all changes in nomenclature, this risks a disconnect between existing literature and contemporary clinical practice.

 

REFERENCES

 

  1. McCormack A. Pituitary carcinoma. In: Wass J, Semple R, Arlt W, editors. Oxford Textbook of Endocrinology and Diabetes. 3rd ed. Oxford: Oxford University Press. In Press.
  2. Raverot G, Burman P, McCormack A, Heaney A, Petersenn S, Popovic V, et al. European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol. 2018;178(1):G1-G24.
  3. Ilie MD, Jouanneau E, Raverot G. Aggressive Pituitary Adenomas and Carcinomas. Endocrinol Metab Clin North Am. 2020;49(3):505-15.
  4. De Sousa SMC, Baranoff J, Rushworth RL, Butler J, Sorbello J, Vorster J, et al. Impulse control disorders in dopamine agonist-treated hyperprolactinemia: prevalence and risk factors. J Clin Endocrinol Metab. 2019.
  5. Raverot G, Castinetti F, Jouanneau E, Morange I, Figarella-Branger D, Dufour H, et al. Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment. Clin Endocrinol (Oxf). 2012;76(6):769-75.
  6. Tampourlou M, Ntali G, Ahmed S, Arlt W, Ayuk J, Byrne JV, et al. Outcome of Nonfunctioning Pituitary Adenomas That Regrow After Primary Treatment: A Study From Two Large UK Centers. J Clin Endocrinol Metab. 2017;102(6):1889-97.
  7. Besser GM, Burman P, Daly AF. Predictors and rates of treatment-resistant tumor growth in acromegaly. Eur J Endocrinol. 2005;153(2):187-93.
  8. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Endocrine Organs. 3rd ed. DeLellis RA, Lloyd RV, Heitz PU, Eng C, editors. Lyon: IARC Press; 2004; 2004.
  9. Dworakowska D, Grossman AB. Aggressive and malignant pituitary tumours: state-of-the-art. Endocr Relat Cancer. 2018.
  10. Tanizaki Y, Jin L, Scheithauer BW, Kovacs K, Roncaroli F, Lloyd RV. P53 gene mutations in pituitary carcinomas. Endocr Pathol. 2007;18(4):217-22.
  11. Lopes MBS. The 2017 World Health Organization classification of tumors of the pituitary gland: a summary. Acta Neuropathol. 2017;134(4):521-35.
  12. World Health Organization Classification of Tumours of Endocrine Organs. 4th ed. Lloyd RV, Osamura RY, Klöppel G, Rosai J, editors. Lyon: IARC Press; 2017; 2017.
  13. Chiloiro S, Doglietto F, Trapasso B, Iacovazzo D, Giampietro A, Di Nardo F, et al. Typical and atypical pituitary adenomas: a single-center analysis of outcome and prognosis. Neuroendocrinology. 2015;101(2):143-50.
  14. Pernicone PJ, Scheithauer BW, Sebo TJ, Kovacs KT, Horvath E, Young WF, Jr., et al. Pituitary carcinoma: a clinicopathologic study of 15 cases. Cancer. 1997;79(4):804-12.
  15. Trouillas J, Roy P, Sturm N, Dantony E, Cortet-Rudelli C, Viennet G, et al. 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):123-35.
  16. Raverot G, Dantony E, Beauvy J, Vasiljevic A, Mikolasek S, Borson-Chazot F, et al. Risk of Recurrence in Pituitary Neuroendocrine Tumors: A Prospective Study Using a Five-Tiered Classification. J Clin Endocrinol Metab. 2017;102(9):3368-74.
  17. Trouillas J, Burman P, McCormack A, Petersenn S, Popovic V, Dekkers O, et al. Aggressive pituitary tumours and carcinomas: two sides of the same coin? Eur J Endocrinol. 2018;178(6):C7-c9.
  18. Kim JS, Lee YS, Jung MJ, Hong YK. The Predictive Value of Pathologic Features in Pituitary Adenoma and Correlation with Pituitary Adenoma Recurrence. J Pathol Transl Med. 2016;50(6):419-25.
  19. Matsuyama J. Ki-67 expression for predicting progression of postoperative residual pituitary adenomas: correlations with clinical variables. Neurol Med Chir (Tokyo). 2012;52(8):563-9.
  20. Salenave S, Ancelle D, Bahougne T, Raverot G, Kamenicky P, Bouligand J, et al. Macroprolactinomas in children and adolescents: factors associated with the response to treatment in 77 patients. J Clin Endocrinol Metab. 2015;100(3):1177-86.
  21. Mete O, Gomez-Hernandez K, Kucharczyk W, Ridout R, Zadeh G, Gentili F, et al. Silent subtype 3 pituitary adenomas are not always silent and represent poorly differentiated monomorphous plurihormonal Pit-1 lineage adenomas. Mod Pathol. 2016;29(2):131-42.
  22. Cho HY, Cho SW, Kim SW, Shin CS, Park KS, Kim SY. Silent corticotroph adenomas have unique recurrence characteristics compared with other nonfunctioning pituitary adenomas. Clin Endocrinol (Oxf). 2010;72(5):648-53.
  23. George DH, Scheithauer BW, Kovacs K, Horvath E, Young WF, Jr., Lloyd RV, et al. Crooke's cell adenoma of the pituitary: an aggressive variant of corticotroph adenoma. Am J Surg Pathol. 2003;27(10):1330-6.
  24. Larkin S, Reddy R, Karavitaki N, Cudlip S, Wass J, Ansorge O. Granulation pattern, but not GSP or GHR mutation, is associated with clinical characteristics in somatostatin-naive patients with somatotroph adenomas. Eur J Endocrinol. 2013;168(4):491-9.
  25. Delgrange E, Daems T, Verhelst J, Abs R, Maiter D. Characterization of resistance to the prolactin-lowering effects of cabergoline in macroprolactinomas: a study in 122 patients. Eur J Endocrinol. 2009;160(5):747-52.
  26. 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):203-16.
  27. Alshaikh OM, Asa SL, Mete O, Ezzat S. An Institutional Experience of Tumor Progression to Pituitary Carcinoma in a 15-Year Cohort of 1055 Consecutive Pituitary Neuroendocrine Tumors. Endocrine pathology. 2019;30(2):118-27.
  28. van der Zwan JM, Mallone S, van Dijk B, Bielska-Lasota M, Otter R, Foschi R, et al. Carcinoma of endocrine organs: results of the RARECARE project. Eur J Cancer. 2012;48(13):1923-31.
  29. Mountcastle RB, Roof BS, Mayfield RK, Mordes DB, Sagel J, Biggs PJ, et al. Pituitary adenocarcinoma in an acromegalic patient: response to bromocriptine and pituitary testing: a review of the literature on 36 cases of pituitary carcinoma. Am J Med Sci. 1989;298(2):109-18.
  30. Santos-Pinheiro F, Penas-Prado M, Kamiya-Matsuoka C, Waguespack SG, Mahajan A, Brown PD, et al. Treatment and long-term outcomes in pituitary carcinoma: a cohort study. Eur J Endocrinol. 2019;181(4):397-407.
  31. Huang AP, Yang SH, Yang CC, Kuo MF, Wu MZ, Tu YK. Malignant prolactinoma with craniospinal metastasis in a 12-year-old boy. J Neurooncol. 2008;90(1):41-6.
  32. Guzel A, Tatli M, Senturk S, Guzel E, Cayli SR, Sav A. Pituitary carcinoma presenting with multiple metastases: case report. J Child Neurol. 2008;23(12):1467-71.
  33. McCormack A, Dekkers OM, Petersenn S, Popovic V, Trouillas J, Raverot G, et al. Treatment of aggressive pituitary tumours and carcinomas: results of a European Society of Endocrinology (ESE) survey 2016. Eur J Endocrinol. 2018;178(3):265-76.
  34. Yoo F, Kuan EC, Heaney AP, Bergsneider M, Wang MB. Corticotrophic pituitary carcinoma with cervical metastases: case series and literature review. Pituitary. 2018;21(3):290-301.
  35. Scheithauer BW, Kurtkaya-Yapicier O, Kovacs KT, Young WF, Jr., Lloyd RV. Pituitary carcinoma: a clinicopathological review. Neurosurgery. 2005;56(5):1066-74.
  36. Lenders N, McCormack A. Malignant transformation in non-functioning pituitary adenomas (pituitary carcinoma). Pituitary. 2018;21(2):217-29.
  37. De Sousa SM, McCabe MJ, Wu K, Roscioli T, Gayevskiy V, Brook K, et al. Germline variants in familial pituitary tumour syndrome genes are common in young patients and families with additional endocrine tumours. Eur J Endocrinol. 2017;176(5):635-44.
  38. Tufton N, Roncaroli F, Hadjidemetriou I, Dang MN, Denes J, Guasti L, et al. Pituitary Carcinoma in a Patient with an SDHB Mutation. Endocr Pathol. 2017;28(4):320-5.
  39. Bengtsson D, Joost P, Aravidis C, Askmalm Stenmark M, Backman AS, Melin B, et al. Corticotroph Pituitary Carcinoma in a Patient With Lynch Syndrome (LS) and Pituitary Tumors in a Nationwide LS Cohort. J Clin Endocrinol Metab. 2017;102(11):3928-32.
  40. Scheithauer BW, Kovacs K, Nose V, Lombardero M, Osamura YR, Lloyd RV, et al. Multiple endocrine neoplasia type 1-associated thyrotropin-producing pituitary carcinoma: report of a probable de novo example. Hum Pathol. 2009;40(2):270-8.
  41. Gordon MV, Varma D, McLean CA, Bittar RG, Burgess JR, Topliss DJ. Metastatic prolactinoma presenting as a cervical spinal cord tumour in multiple endocrine neoplasia type one (MEN-1). Clin Endocrinol (Oxf). 2007;66(1):150-2.
  42. Elston MS, McDonald KL, Clifton-Bligh RJ, Robinson BG. Familial pituitary tumor syndromes. Nat Rev Endocrinol. 2009;5(8):453-61.
  43. Casar-Borota O, Boldt HB, Engström BE, Andersen MS, Baussart B, Bengtsson D, et al. Corticotroph Aggressive Pituitary Tumors and Carcinomas Frequently Harbor ATRX Mutations. J Clin Endocrinol Metab. 2021;106(4):1183-94.
  44. Uzilov AV, Taik P, Cheesman KC, Javanmard P, Ying K, Roehnelt A, et al. USP8 and TP53 Drivers are Associated with CNV in a Corticotroph Adenoma Cohort Enriched for Aggressive Tumors. J Clin Endocrinol Metab. 2021;106(3):826-42.
  45. Neou M, Villa C, Armignacco R, Jouinot A, Raffin-Sanson ML, Septier A, et al. Pangenomic Classification of Pituitary Neuroendocrine Tumors. Cancer cell. 2020;37(1):123-34.e5.
  46. Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F. Mutations in the deubiquitinase gene USP8 cause Cushing's disease. 2015;47(1):31-8.
  47. Perez-Rivas LG, Theodoropoulou M, Ferrau F, Nusser C, Kawaguchi K, Stratakis CA, et al. 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.
  48. Guastamacchia E, Triggiani V, Tafaro E, De Tommasi A, De Tommasi C, Luzzi S, et al. Evolution of a prolactin-secreting pituitary microadenoma into a fatal carcinoma: a case report. Minerva Endocrinol. 2007;32(3):231-6.
  49. Nose-Alberti V, Mesquita MI, Martin LC, Kayath MJ. Adrenocorticotropin-Producing Pituitary Carcinoma with Expression of c-erbB-2 and High PCNA Index: A Comparative Study with Pituitary Adenomas and Normal Pituitary Tissues. Endocr Pathol. 1998;9(1):53-62.
  50. Bengtsson D, Schroder HD, Andersen M, Maiter D, Berinder K, Feldt Rasmussen U, et al. 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):1689-98.
  51. Moisi M, Cruz AS, Benkers T, Rostad S, Broyles FB, Yuen K, et al. Treatment of Aggressive Prolactin-Secreting Pituitary Adenomas with Adjuvant Temozolomide Chemotherapy: A Review. Cureus. 2016;8(6):e658.
  52. Cartwright DM, Miller TR, Nasr AJ. Fine-needle aspiration biopsy of pituitary carcinoma with cervical lymph node metastases: a report of two cases and review of the literature. Diagn Cytopathol. 1994;11(1):68-73.
  53. Yakoushina TV, Lavi E, Hoda RS. Pituitary carcinoma diagnosed on fine needle aspiration: Report of a case and review of pathogenesis. CytoJournal. 2010;7:14.
  54. Todeschini AB, Beer-Furlan A, Montaser AS, Jamshidi AO, Ghalib L, Chavez JA, et al. Pituitary carcinomas: review of the current literature and report of atypical case. Br J Neurosurg. 2020;34(5):528-33.
  55. Pinto EM, Siqueira SA, Cukier P, Fragoso MC, Lin CJ, de Mendonca BB. Possible role of a radiation-induced p53 mutation in a Nelson's syndrome patient with a fatal outcome. Pituitary. 2011;14(4):400-4.
  56. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M, Grossman AB. Clinical review: Diagnosis and management of pituitary carcinomas. J Clin Endocrinol Metab. 2005;90(5):3089-99.
  57. Heck A, Emblem KE, Casar-Borota O, Bollerslev J, Ringstad G. Quantitative analyses of T2-weighted MRI as a potential marker for response to somatostatin analogs in newly diagnosed acromegaly. Endocrine. 2016;52(2):333-43.
  58. Knosp E, Steiner E, Kitz K, Matula C. Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery. 1993;33(4):610-7; discussion 7-8.
  59. Micko AS, Wöhrer A, Wolfsberger S, Knosp E. Invasion of the cavernous sinus space in pituitary adenomas: endoscopic verification and its correlation with an MRI-based classification. Journal of neurosurgery. 2015;122(4):803-11.
  60. Imber BS, Lin AL, Zhang Z, Keshavamurthy KN, Deipolyi AR, Beal K, et al. Comparison of Radiographic Approaches to Assess Treatment Response in Pituitary Adenomas: Is RECIST or RANO Good Enough? Journal of the Endocrine Society. 2019;3(9):1693-706.
  61. Honegger J, Zimmermann S, Psaras T, Petrick M, Mittelbronn M, Ernemann U, et al. Growth modelling of non-functioning pituitary adenomas in patients referred for surgery. Eur J Endocrinol. 2008;158(3):287-94.
  62. Garmes HM, Carvalheira JBC, Reis F, Queiroz LS, Fabbro MD, Souza VFP, et al. Pituitary carcinoma: A case report and discussion of potential value of combined use of Ga-68 DOTATATE and F-18 FDG PET/CT scan to better choose therapy. Surg Neurol Int. 2017;8:162.
  63. Mixson AJ, Friedman TC, Katz DA, Feuerstein IM, Taubenberger JK, Colandrea JM, et al. Thyrotropin-secreting pituitary carcinoma. J Clin Endocrinol Metab. 1993;76(2):529-33.
  64. Pichard C, Gerber S, Laloi M, Kujas M, Clemenceau S, Ponvert D, et al. Pituitary carcinoma: report of an exceptional case and review of the literature. J Endocrinol Invest. 2002;25(1):65-72.
  65. Ramirez C, Cheng S, Vargas G, Asa SL, Ezzat S, Gonzalez B, et al. 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):1745-51.
  66. Zaidi HA, Cote DJ, Dunn IF, Laws ER, Jr. Predictors of aggressive clinical phenotype among immunohistochemically confirmed atypical adenomas. J Clin Neurosci. 2016;34:246-51.
  67. Miermeister CP, Petersenn S, Buchfelder M, Fahlbusch R, Lüdecke DK, Hölsken A, et al. Histological criteria for atypical pituitary adenomas – data from the German pituitary adenoma registry suggests modifications. Acta Neuropathol Commun. 2015;3:50.
  68. Kumar K, Macaulay RJ, Kelly M, Pirlot T. Absent p53 immunohistochemical staining in a pituitary carcinoma. Can J Neurol Sci. 2001;28(2):174-8.
  69. Villa C, Vasiljevic A, Jaffrain-Rea ML, Ansorge O, Asioli S, Barresi V, et al. A standardised diagnostic approach to pituitary neuroendocrine tumours (PitNETs): a European Pituitary Pathology Group (EPPG) proposal. Virchows Arch. 2019;475(6):687-92.
  70. 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):237-46.
  71. Asioli S, Righi A, Iommi M, Baldovini C, Ambrosi F, Guaraldi F, et al. Validation of a clinicopathological score for the prediction of post-surgical evolution of pituitary adenoma: retrospective analysis on 566 patients from a tertiary care centre. Eur J Endocrinol. 2019;180(2):127-34.
  72. Nishioka H, Inoshita N, Mete O, Asa SL, Hayashi K, Takeshita A, et al. The Complementary Role of Transcription Factors in the Accurate Diagnosis of Clinically Nonfunctioning Pituitary Adenomas. Endocr Pathol. 2015;26(4):349-55.
  73. Wang H, Liang J, Yong WH, Sullivan P. Metastatic Pituitary Carcinoma to Cervical Lymph Node: Diagnosis by Fine Needle Aspiration and Review of the Literature. Acta Cytol. 2017;61(3):242-6.
  74. Venable ER, Kerr SE, Lopes MBS, Jones KA, Bellizzi AM, Mounajjed T, et al. Liver metastases from pituitary carcinomas mimicking visceral well-differentiated neuroendocrine tumors: a series of four cases. Diagn Pathol. 2020;15(1):81.
  75. Casar-Borota O, Heck A, Schulz S, Nesland JM, Ramm-Pettersen J, Lekva T, et al. Expression of SSTR2a, but not of SSTRs 1, 3, or 5 in somatotroph adenomas assessed by monoclonal antibodies was reduced by octreotide and correlated with the acute and long-term effects of octreotide. J Clin Endocrinol Metab. 2013;98(11):E1730-9.
  76. De Sousa SMC, Earls P, McCormack AI. Pituitary hyperplasia: case series and literature review of an under-recognised and heterogeneous condition. Endocrinol Diabetes Metab Case Rep. 2015:150017.
  77. Karl M, Von Wichert G, Kempter E, Katz DA, Reincke M, Monig H, et al. Nelson's syndrome associated with a somatic frame shift mutation in the glucocorticoid receptor gene. J Clin Endocrinol Metab. 1996;81(1):124-9.
  78. Wu ZB, Zheng WM, Su ZP, Chen Y, Wu JS, Wang CD, et al. Expression of D2RmRNA isoforms and ERmRNA isoforms in prolactinomas: correlation with the response to bromocriptine and with tumor biological behavior. J Neurooncol. 2010;99(1):25-32.
  79. De Sousa SMC, Wang PPS, Santoreneos S, Shen A, Yates CJ, Babic M, et al. The Genomic Landscape of Sporadic Prolactinomas. Endocrine pathology. 2019;30(4):318-28.
  80. Wang F, Gao H, Li C, Bai J, Lu R, Cao L, et al. Low levels of PRB3 mRNA are associated with dopamine-agonist resistance and tumor recurrence in prolactinomas. J Neurooncol. 2014;116(1):83-8.
  81. Oh MC, Aghi MK. Dopamine agonist-resistant prolactinomas. J Neurosurg. 2011;114(5):1369-79.
  82. Turner HE, Nagy Z, Esiri MM, Harris AL, Wass JA. Role of matrix metalloproteinase 9 in pituitary tumor behavior. J Clin Endocrinol Metab. 2000;85(8):2931-5.
  83. Taylor WA, Uttley D, Wilkins PR. Multiple dural metastases from a pituitary adenoma. Case report. J Neurosurg. 1994;81(4):624-6.
  84. Melmed S. Pathogenesis of pituitary tumors. Nat Rev Endocrinol. 2011;7(5):257-66.
  85. Roncaroli F, Scheithauer BW, Young WF, Horvath E, Kovacs K, Kros JM, et al. Silent corticotroph carcinoma of the adenohypophysis: a report of five cases. Am J Surg Pathol. 2003;27(4):477-86.
  86. Filippella M, Galland F, Kujas M, Young J, Faggiano A, Lombardi G, et al. Pituitary tumour transforming gene (PTTG) expression correlates with the proliferative activity and recurrence status of pituitary adenomas: a clinical and immunohistochemical study. Clin Endocrinol (Oxf). 2006;65(4):536-43.
  87. Karga HJ, Alexander JM, Hedley-Whyte ET, Klibanski A, Jameson JL. Ras mutations in human pituitary tumors. J Clin Endocrinol Metab. 1992;74(4):914-9.
  88. Pei L, Melmed S, Scheithauer B, Kovacs K, Prager D. H-ras mutations in human pituitary carcinoma metastases. J Clin Endocrinol Metab. 1994;78(4):842-6.
  89. Wierinckx A, Auger C, Devauchelle P, Reynaud A, Chevallier P, Jan M, et al. A diagnostic marker set for invasion, proliferation, and aggressiveness of prolactin pituitary tumors. Endocr Relat Cancer. 2007;14(3):887-900.
  90. Bi WL, Horowitz P, Greenwald NF, Abedalthagafi M, Agarwalla PK, Gibson WJ, et al. Landscape of Genomic Alterations in Pituitary Adenomas. Clin Cancer Res. 2017;23(7):1841-51.
  91. Rickert CH, Dockhorn-Dworniczak B, Busch G, Moskopp D, Albert FK, Rama B, et al. Increased chromosomal imbalances in recurrent pituitary adenomas. Acta Neuropathol. 2001;102(6):615-20.
  92. Rickert CH, Scheithauer BW, Paulus W. Chromosomal aberrations in pituitary carcinoma metastases. Acta Neuropathol. 2001;102(2):117-20.
  93. Lloyd RV, Scheithauer BW, Kuroki T, Vidal S, Kovacs K, Stefaneanu L. Vascular Endothelial Growth Factor (VEGF) Expression in Human Pituitary Adenomas and Carcinomas. Endocr Pathol. 1999;10(3):229-35.
  94. Ortiz LD, Syro LV, Scheithauer BW, Ersen A, Uribe H, Fadul CE, et al. Anti-VEGF therapy in pituitary carcinoma. Pituitary. 2012;15(3):445-9.
  95. Onguru O, Scheithauer BW, Kovacs K, Vidal S, Jin L, Zhang S, et al. Analysis of epidermal growth factor receptor and activated epidermal growth factor receptor expression in pituitary adenomas and carcinomas. Mod Pathol. 2004;17(7):772-80.
  96. Simpson DJ, Fryer AA, Grossman AB, Wass JA, Pfeifer M, Kros JM, et al. Cyclin D1 (CCND1) genotype is associated with tumour grade in sporadic pituitary adenomas. Carcinogenesis. 2001;22(11):1801-7.
  97. Jordan S, Lidhar K, Korbonits M, Lowe DG, Grossman AB. Cyclin D and cyclin E expression in normal and adenomatous pituitary. Eur J Endocrinol. 2000;143(1):R1-6.
  98. Vidal S, Kovacs K, Horvath E, Rotondo F, Kuroki T, Lloyd RV, et al. Topoisomerase IIalpha expression in pituitary adenomas and carcinomas: relationship to tumor behavior. Mod Pathol. 2002;15(11):1205-12.
  99. Hirohata T, Asano K, Ogawa Y, Takano S, Amano K, Isozaki O, et al. DNA mismatch repair protein (MSH6) correlated with the responses of atypical pituitary adenomas and pituitary carcinomas to temozolomide: the national cooperative study by the Japan Society for Hypothalamic and Pituitary Tumors. J Clin Endocrinol Metab. 2013;98(3):1130-6.
  100. Lin AL, Jonsson P, Tabar V, Yang TJ, Cuaron J, Beal K, et al. Marked Response of a Hypermutated ACTH-Secreting Pituitary Carcinoma to Ipilimumab and Nivolumab. J Clin Endocrinol Metab. 2018;103(10):3925-30.
  101. McCormack A, Kaplan W, Gill AJ, Little N, Cook R, Robinson B, et al. MGMT expression and pituitary tumours: relationship to tumour biology. Pituitary. 2013;16(2):208-19.
  102. Lidhar K, Korbonits M, Jordan S, Khalimova Z, Kaltsas G, Lu X, et al. Low expression of the cell cycle inhibitor p27Kip1 in normal corticotroph cells, corticotroph tumors, and malignant pituitary tumors. J Clin Endocrinol Metab. 1999;84(10):3823-30.
  103. Pei L, Melmed S, Scheithauer B, Kovacs K, Benedict WF, Prager D. Frequent loss of heterozygosity at the retinoblastoma susceptibility gene (RB) locus in aggressive pituitary tumors: evidence for a chromosome 13 tumor suppressor gene other than RB. Cancer Res. 1995;55(8):1613-6.
  104. Kulig E, Jin L, Qian X, Horvath E, Kovacs K, Stefaneanu L, et al. Apoptosis in Nontumorous and Neoplastic Human Pituitaries : Expression of the Bcl-2 Family of Proteins. Am J Pathol. 1999;154(3):767-74.
  105. Onguru O, Scheithauer BW, Kovacs K, Vidal S, Jin L, Zhang S, et al. Analysis of Cox-2 and thromboxane synthase expression in pituitary adenomas and carcinomas. Endocr Pathol. 2004;15(1):17-27.
  106. Riss D, Jin L, Qian X, Bayliss J, Scheithauer BW, Young WF, Jr., et al. Differential expression of galectin-3 in pituitary tumors. Cancer Res. 2003;63(9):2251-5.
  107. Vidal S, Horvath E, Kovacs K, Kuroki T, Lloyd RV, Scheithauer BW. Expression of hypoxia-inducible factor-1alpha (HIF-1alpha) in pituitary tumours. Histol Histopathol. 2003;18(3):679-86.
  108. Lania AG, Ferrero S, Pivonello R, Mantovani G, Peverelli E, Di Sarno A, et al. Evolution of an aggressive prolactinoma into a growth hormone secreting pituitary tumor coincident with GNAS gene mutation. J Clin Endocrinol Metab. 2010;95(1):13-7.
  109. Harada K, Arita K, Kurisu K, Tahara H. Telomerase activity and the expression of telomerase components in pituitary adenoma with malignant transformation. Surg Neurol. 2000;53(3):267-74.
  110. Richardson TE, Shen ZJ, Kanchwala M, Xing C, Filatenkov A, Shang P, et al. Aggressive Behavior in Silent Subtype III Pituitary Adenomas May Depend on Suppression of Local Immune Response: A Whole Transcriptome Analysis. J Neuropathol Exp Neurol. 2017;76(10):874-82.
  111. De Sousa SMC, Sheriff N, Tran CH, Menzies AM, Tsang VHM, Long GV, et al. Fall in thyroid stimulating hormone (TSH) may be an early marker of ipilimumab-induced hypophysitis. Pituitary. 2018;21(3):274-82.
  112. Roche M, Wierinckx A, Croze S, Rey C, Legras-Lachuer C, Morel A-P, et al. Deregulation of miR-183 and KIAA0101 in Aggressive and Malignant Pituitary Tumors. Front Med (Lausanne). 2015;2:54.
  113. Wei Z, Zhou C, Liu M, Yao Y, Sun J, Xiao J, et al. MicroRNA involvement in a metastatic non-functioning pituitary carcinoma. Pituitary. 2015;18(5):710-21.
  114. Stilling G, Sun Z, Zhang S, Jin L, Righi A, Kovacs G, et al. MicroRNA expression in ACTH-producing pituitary tumors: up-regulation of microRNA-122 and -493 in pituitary carcinomas. Endocrine. 2010;38(1):67-75.
  115. Ciric I, Ragin A, Baumgartner C, Pierce D. Complications of transsphenoidal surgery: results of a national survey, review of the literature, and personal experience. Neurosurgery. 1997;40(2):225-36; discussion 36-7.
  116. Bates PR, Carson MN, Trainer PJ, Wass JA, Group UKNARS. Wide variation in surgical outcomes for acromegaly in the UK. Clin Endocrinol (Oxf). 2008;68(1):136-42.
  117. Wang YY, Higham C, Kearney T, Davis JR, Trainer P, Gnanalingham KK. Acromegaly surgery in Manchester revisited--the impact of reducing surgeon numbers and the 2010 consensus guidelines for disease remission. Clin Endocrinol (Oxf). 2012;76(3):399-406.
  118. Barker FG, 2nd, Klibanski A, Swearingen B. Transsphenoidal surgery for pituitary tumors in the United States, 1996-2000: mortality, morbidity, and the effects of hospital and surgeon volume. J Clin Endocrinol Metab. 2003;88(10):4709-19.
  119. Park HH, Kim EH, Ku CR, Lee EJ, Kim SH. Outcomes of Aggressive Surgical Resection in Growth Hormone-Secreting Pituitary Adenomas with Cavernous Sinus Invasion. World neurosurgery. 2018;117:e280-e9.
  120. Meij BP, Lopes MB, Ellegala DB, Alden TD, Laws ER, Jr. The long-term significance of microscopic dural invasion in 354 patients with pituitary adenomas treated with transsphenoidal surgery. J Neurosurg. 2002;96(2):195-208.
  121. Komotar RJ, Starke RM, Raper DM, Anand VK, Schwartz TH. Endoscopic endonasal compared with microscopic transsphenoidal and open transcranial resection of giant pituitary adenomas. Pituitary. 2012;15(2):150-9.
  122. Juraschka K, Khan OH, Godoy BL, Monsalves E, Kilian A, Krischek B, et al. Endoscopic endonasal transsphenoidal approach to large and giant pituitary adenomas: institutional experience and predictors of extent of resection. J Neurosurg. 2014;121(1):75-83.
  123. Taghvaei M, Sadrehosseini SM, Ardakani JB, Nakhjavani M, Zeinalizadeh M. Endoscopic Endonasal Approach to the Growth Hormone-Secreting Pituitary Adenomas: Endocrinologic Outcome in 68 Patients. World neurosurgery. 2018;117:e259-e68.
  124. Shimon I, Jallad RS, Fleseriu M, Yedinak CG, Greenman Y, Bronstein MD. Giant GH-secreting pituitary adenomas: management of rare and aggressive pituitary tumors. Eur J Endocrinol. 2015;172(6):707-13.
  125. Shimon I, Sosa E, Mendoza V, Greenman Y, Tirosh A, Espinosa E, et al. Giant prolactinomas larger than 60 mm in size: a cohort of massive and aggressive prolactin-secreting pituitary adenomas. Pituitary. 2016;19(4):429-36.
  126. Park KS, Hwang JH, Hwang SK, Kim S, Park SH. Pituitary carcinoma with fourth ventricle metastasis: treatment by excision and Gamma-knife radiosurgery. Pituitary. 2014;17(6):514-8.
  127. Ayuk J, Natarajan G, Geh JI, Mitchell RD, Gittoes NJ. Pituitary carcinoma with a single metastasis causing cervical spinal cord compression. Case report. J Neurosurg Spine. 2005;2(3):349-53.
  128. Joehlin-Price AS, Hardesty DA, Arnold CA, Kirschner LS, Prevedello DM, Lehman NL. Case report: ACTH-secreting pituitary carcinoma metastatic to the liver in a patient with a history of atypical pituitary adenoma and Cushing's disease. Diagn Pathol. 2017;12(1):34.
  129. Heaney AP. Clinical review: Pituitary carcinoma: difficult diagnosis and treatment. J Clin Endocrinol Metab. 2011;96(12):3649-60.
  130. Sheehan JP, Starke RM, Mathieu D, Young B, Sneed PK, Chiang VL, et al. Gamma Knife radiosurgery for the management of nonfunctioning pituitary adenomas: a multicenter study. J Neurosurg. 2013;119(2):446-56.
  131. Gittoes NJ, Bates AS, Tse W, Bullivant B, Sheppard MC, Clayton RN, et al. Radiotherapy for non-function pituitary tumours. Clin Endocrinol (Oxf). 1998;48(3):331-7.
  132. Ding D, Starke RM, Sheehan JP. Treatment paradigms for pituitary adenomas: defining the roles of radiosurgery and radiation therapy. J Neurooncol. 2014;117(3):445-57.
  133. Edwards AA, Swords FM, Plowman PN. Focal radiation therapy for patients with persistent/recurrent pituitary adenoma, despite previous radiotherapy. Pituitary. 2009;12(1):30-4.
  134. Swords FM, Monson JP, Besser GM, Chew SL, Drake WM, Grossman AB, et al. Gamma knife radiosurgery: a safe and effective salvage treatment for pituitary tumours not controlled despite conventional radiotherapy. Eur J Endocrinol. 2009;161(6):819-28.
  135. Chinezu L, Vasiljevic A, Jouanneau E, Francois P, Borda A, Trouillas J, et al. Expression of somatostatin receptors, SSTR2A and SSTR5, in 108 endocrine pituitary tumors using immunohistochemical detection with new specific monoclonal antibodies. Hum Pathol. 2014;45(1):71-7.
  136. Xiao J, Zhu Z, Zhong D, Ma W, Wang R. Improvement in diagnosis of metastatic pituitary carcinoma by 68Ga DOTATATE PET/CT. Clin Nucl Med. 2015;40(2):e129-31.
  137. Kovacs GL, Goth M, Rotondo F, Scheithauer BW, Carlsen E, Saadia A, et al. ACTH-secreting Crooke cell carcinoma of the pituitary. Eur J Clin Invest. 2013;43(1):20-6.
  138. Ilie MD, Lasolle H, Raverot G. Emerging and Novel Treatments for Pituitary Tumors. J Clin Med. 2019;8(8):1107.
  139. Lin AL, Tabar V, Young RJ, Cohen M, Cuaron J, Yang TJ, et al. Synergism of Checkpoint Inhibitors and Peptide Receptor Radionuclide Therapy in the Treatment of Pituitary Carcinoma. Journal of the Endocrine Society. 2021;5(10):bvab133-bvab.
  140. Hurel SJ, Harris PE, McNicol AM, Foster S, Kelly WF, Baylis PH. Metastatic prolactinoma: effect of octreotide, cabergoline, carboplatin and etoposide; immunocytochemical analysis of proto-oncogene expression. J Clin Endocrinol Metab. 1997;82(9):2962-5.
  141. Gollard R, Kosty M, Cheney C, Copeland B, Bordin G. Prolactin-secreting pituitary carcinoma with implants in the cheek pouch and metastases to the ovaries. A case report and literature review. Cancer. 1995;76(10):1814-20.
  142. Daniel E, Debono M, Caunt S, Girio-Fragkoulakis C, Walters SJ, Akker SA, et al. A prospective longitudinal study of Pasireotide in Nelson's syndrome. Pituitary. 2018;21(3):247-55.
  143. Lasolle H, Vasiljevic A, Borson-Chazot F, Raverot G. Pasireotide: A potential therapeutic alternative for resistant prolactinoma. Ann Endocrinol (Paris). 2018.
  144. Coopmans EC, van Meyel SWF, Pieterman KJ, van Ipenburg JA, Hofland LJ, Donga E, et al. Excellent response to pasireotide therapy in an aggressive and dopamine-resistant prolactinoma. Eur J Endocrinol. 2019;181(2):K21-k7.
  145. Raverot G, Vasiljevic A, Jouanneau E, Lasolle H. Confirmation of a new therapeutic option for aggressive or dopamine agonist-resistant prolactin pituitary neuroendocrine tumors. Eur J Endocrinol. 2019;181(2):C1-c3.
  146. Yamashita S, Izumi M, Nagataki S. Acromegaly and pituitary carcinoma. Ann Intern Med. 1992;117(12):1057-8.
  147. Bronstein MD, Fleseriu M, Neggers S, Colao A, Sheppard M, Gu F, et al. Switching patients with acromegaly from octreotide to pasireotide improves biochemical control: crossover extension to a randomized, double-blind, Phase III study. BMC Endocr Disord. 2016;16:16.
  148. Iacovazzo D, Carlsen E, Lugli F, Chiloiro S, Piacentini S, Bianchi A, et al. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol. 2016;174(2):241-50.
  149. Kaltsas GA, Grossman AB. Malignant pituitary tumours. Pituitary. 1998;1(1):69-81.
  150. Syro LV, Rotondo F, Camargo M, Ortiz LD, Serna CA, Kovacs K. Temozolomide and Pituitary Tumors: Current Understanding, Unresolved Issues, and Future Directions. Front Endocrinol (Lausanne). 2018;9:318.
  151. Lormeau B, Miossec P, Sibony M, Valensi P, Attali JR. Adrenocorticotropin-producing pituitary carcinoma with liver metastasis. J Endocrinol Invest. 1997;20(4):230-6.
  152. Ji Y, Vogel RI, Lou E. Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports. Neurooncol Pract. 2016;3(3):188-95.
  153. Lasolle H, Cortet C, Castinetti F, Cloix L, Caron P, Delemer B, et al. Temozolomide treatment can improve overall survival in aggressive pituitary tumors and pituitary carcinomas. Eur J Endocrinol. 2017;176(6):769-77.
  154. Whitelaw B, Dworakowska D, Thomas N, Barazi S, Riordan-Eva P, King A, et al. Temozolomide in the management of dopamine agonist resistant prolactinomas. Clin Endocrinol (Oxf). 2012.
  155. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-96.
  156. 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):1133-48.
  157. Zacharia BE, Gulati AP, Bruce JN, Carminucci AS, Wardlaw SL, Siegelin M, et al. High response rates and prolonged survival in patients with corticotroph pituitary tumors and refractory Cushing disease from capecitabine and temozolomide (CAPTEM): a case series. Neurosurgery. 2014;74(4):E447-55; discussion E55.
  158. Losa M, Bogazzi F, Cannavo S, Ceccato F, Curto L, De Marinis L, et al. Temozolomide therapy in patients with aggressive pituitary adenomas or carcinomas. J Neurooncol. 2016;126(3):519-25.
  159. Kil WJ, Cerna D, Burgan WE, Beam K, Carter D, Steeg PS, et al. In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide. Clin Cancer Res. 2008;14(3):931-8.
  160. Carlson BL, Grogan PT, Mladek AC, Schroeder MA, Kitange GJ, Decker PA, et al. Radiosensitizing effects of temozolomide observed in vivo only in a subset of O6-methylguanine-DNA methyltransferase methylated glioblastoma multiforme xenografts. Int J Radiat Oncol Biol Phys. 2009;75(1):212-9.
  161. Burman P, Lamb L, McCormack A. Temozolomide therapy for aggressive pituitary tumours – current understanding and future perspectives. Reviews in Endocrine and Metabolic Disorders. 2020;21(2):263-76.
  162. Mendola M, Passeri E, Ambrosi B, Corbetta S. Multiple cerebral hemorrhagic foci from metastases during temozolomide treatment in a patient with corticotroph pituitary carcinoma. J Clin Endocrinol Metab. 2014;99(8):2623-4.
  163. Campdera M, Palacios N, Aller J, Magallon R, Martin P, Saucedo G, et al. Temozolomide for aggressive ACTH pituitary tumors: failure of a second course of treatment. Pituitary. 2016;19(2):158-66.
  164. Grieco A, Tafuri MA, Biolato M, Diletto B, Di Napoli N, Balducci N, et al. Severe cholestatic hepatitis due to temozolomide: an adverse drug effect to keep in mind. Case report and review of literature. Medicine (Baltimore). 2015;94(12):e476.
  165. Raverot G, Sturm N, de Fraipont F, Muller M, Salenave S, Caron P, et al. Temozolomide Treatment in Aggressive Pituitary Tumors and Pituitary Carcinomas: A French Multicenter Experience. J Clin Endocrinol Metab. 2010.
  166. Marchesi F, Turriziani M, Tortorelli G, Avvisati G, Torino F, De Vecchis L. Triazene compounds: mechanism of action and related DNA repair systems. Pharmacol Res. 2007;56(4):275-87.
  167. Bengtsson D, Schrøder HD, Berinder K, Maiter D, Hoybye C, Ragnarsson O, et al. Tumoral MGMT content predicts survival in patients with aggressive pituitary tumors and pituitary carcinomas given treatment with temozolomide. Endocrine. 2018;62(3):737-9.
  168. Salehi F, Scheithauer BW, Kros JM, Lau Q, Fealey M, Erickson D, et al. MGMT promoter methylation and immunoexpression in aggressive pituitary adenomas and carcinomas. J Neurooncol. 2011.
  169. Murakami M, Mizutani A, Asano S, Katakami H, Ozawa Y, Yamazaki K, et al. A mechanism of acquiring temozolomide resistance during transformation of atypical prolactinoma into prolactin-producing pituitary carcinoma: case report. Neurosurgery. 2011;68(6):E1761-7; discussion E7.
  170. Jouanneau E, Wierinckx A, Ducray F, Favrel V, Borson-Chazot F, Honnorat J, et al. New targeted therapies in pituitary carcinoma resistant to temozolomide. Pituitary. 2012;15(1):37-43.
  171. Donovan LE, Arnal AV, Wang SH, Odia Y. Widely metastatic atypical pituitary adenoma with mTOR pathway STK11(F298L) mutation treated with everolimus therapy. CNS Oncol. 2016;5(4):203-9.
  172. Zhang D, Way JS, Zhang X, Sergey M, Bergsneider M, Wang MB, et al. Effect of Everolimus in Treatment of Aggressive Prolactin-Secreting Pituitary Adenomas. J Clin Endocrinol Metab. 2019;104(6):1929-36.
  173. Cooper O, Mamelak A, Bannykh S, Carmichael J, Bonert V, Lim S, et al. Prolactinoma ErbB receptor expression and targeted therapy for aggressive tumors. Endocrine. 2014;46(2):318-27.
  174. Cooper O, Bonert VS, Rudnick J, Pressman BD, Lo J, Salvatori R, et al. EGFR/ErbB2-Targeting Lapatinib Therapy for Aggressive Prolactinomas. J Clin Endocrinol Metab. 2021;106(2):e917-e25.
  175. Lamb LS, Sim HW, McCormack AI. Exploring the Role of Novel Medical Therapies for Aggressive Pituitary Tumors: A Review of the Literature-"Are We There Yet?". Cancers (Basel). 2020;12(2).
  176. Duhamel C, Ilie MD, Salle H, Nassouri AS, Gaillard S, Deluche E, et al. Immunotherapy in Corticotroph and Lactotroph Aggressive Tumors and Carcinomas: Two Case Reports and a Review of the Literature. J Pers Med. 2020;10(3).
  177. Sol B, de Filette JMK, Awada G, Raeymaeckers S, Aspeslagh S, Andreescu CE, et al. Immune checkpoint inhibitor therapy for ACTH-secreting pituitary carcinoma: a new emerging treatment? Eur J Endocrinol. 2021;184(1):K1-k5.
  178. Caccese M, Barbot M, Ceccato F, Padovan M, Gardiman MP, Fassan M, et al. Rapid disease progression in patient with mismatch-repair deficiency pituitary ACTH-secreting adenoma treated with checkpoint inhibitor pembrolizumab. Anticancer Drugs. 2020;31(2):199-204.
  179. Majd N, Waguespack SG, Janku F, Fu S, Penas-Prado M, Xu M, et al. Efficacy of pembrolizumab in patients with pituitary carcinoma: report of four cases from a phase II study. J Immunother Cancer. 2020;8(2).
  180. Wang Z, Guo X, Gao L, Deng K, Lian W, Bao X, et al. The Immune Profile of Pituitary Adenomas and a Novel Immune Classification for Predicting Immunotherapy Responsiveness. J Clin Endocrinol Metab. 2020;105(9):e3207-23.
  181. Yeung JT, Vesely MD, Miyagishima DF. In silico analysis of the immunological landscape of pituitary adenomas. Journal of neuro-oncology. 2020;147(3):595-8.
  182. Kaltsas GA, Mukherjee JJ, Plowman PN, Monson JP, Grossman AB, Besser GM. The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumors. J Clin Endocrinol Metab. 1998;83(12):4233-8.
  183. Feng Z, He D, Mao Z, Wang Z, Zhu Y, Zhang X, et al. Utility of 11C-Methionine and 18F-FDG PET/CT in Patients With Functioning Pituitary Adenomas. Clin Nucl Med. 2016;41(3):e130-4.
  184. Whitelaw BC. How and when to use temozolomide to treat aggressive pituitary tumours. Endocrine-related cancer. 2019;26(9):R545-r52.
  185. Di Nunno V, Franceschi E, Tosoni A, Gatto L, Maggio I, Lodi R, et al. Immune-checkpoint inhibitors in pituitary malignancies. Anticancer Drugs. 2021.
  186. Asa SL, Casar-Borota O, Chanson P, Delgrange E, Earls P, Ezzat S, et al. From pituitary adenoma to pituitary neuroendocrine tumor (PitNET): an International Pituitary Pathology Club proposal. Endocr Relat Cancer. 2017;24(4):C5-c8.

 

Multiple Endocrine Neoplasia Type 2

ABSTRACT

 

Multiple Endocrine Neoplasia (MEN) type 2 A and B are rare autosomal dominant inherited cancer syndromes characterized by tumors of the C cells of the thyroid, of the adrenal medulla, and parathyroid glands. MEN2 is caused by a genetic defect in the REarranged during Transfection (RET) proto-oncogene on chromosome 10 (10q11-2), leading to a ligand-independent activation of the transmembrane RET receptor tyrosine kinase and consequently its intra-cellular pathways. Different mutations lead to different levels of activation and MEN2 is therefore characterized by a strong genotype-phenotype correlation. Nearly all patients with MEN2A have either C-cell hyperplasia (CCH) or medullary thyroid cancer (MTC), 50% have pheochromocytoma (PHEO), and 20-30% hyperparathyroidism (pHPT) but incidence of these manifestations is depending on the underlying RET mutation. Patients with MEN2B have a 100% incidence of CCH or MTC, PHEO in 30-50%, mucosal neuromas, and rarely pHPT. Endocrine tumors in MEN2 are often multifocal and bilateral. Nowadays, the diagnosis of MEN2 is made by genetic testing. After diagnosis, annual screening for associated manifestations and prophylactic thyroidectomy for preventing metastasized MTC are advised. Optimal age for preventive surgery or when to start screening for each manifestation is based upon the underlying RET mutation. In patients with MTC present at diagnosis, adequate staging is needed before surgical resection, since surgery is the only curative treatment. The most important biomarker for MTC is calcitonin. Fractionated metanephrines are used for early diagnosing PHEO and calcium and PTH for hyperparathyroidism. For PHEO, a minimal invasive surgical resection is recommended. In pHPT the surgical approach should be tailored to the amount and location of the enlarged glands visualized with imaging. Recurrent MTC requires physical examination, neck ultrasound, and measurement of serum calcitonin and carcinoembryonic antigen (CEA) levels every 6 months. For metastasized MTC, treatment can be successful with multikinase inhibitors (vandetanib, cabozantinib) and selective RET inhibitors (pralsetinib, selpercatinib). 

 

INTRODUCTION

 

Multiple Endocrine Neoplasia (MEN) type 2 is a rare autosomal dominant inherited cancer syndrome characterized by benign and malignant tumors of multiple endocrine organs. MEN2 is caused by a genetic defect in the REarranged during Transfection (RET) proto-oncogene on chromosome 10, leading to a gain-of-function in the RET tyrosine kinase receptor (1–4). As a result, cell growth, proliferation, and differentiation is promoted, leading to multiple tumor formation in all tissues were RET predominantly is expressed (C-cells of the thyroid gland, adrenal medulla, and neurons) (5,6). Major clinical manifestations in MEN2 are medullary thyroid carcinoma (MTC), pheochromocytoma (PHEO) and, in case of MEN2A, primary hyperparathyroidism (pHPT).

 

MTC is a neuroendocrine tumor arising from the calcitonin secreting parafollicular C-cells of the thyroid gland. MTC in persons with MEN2 typically presents at a younger age than sporadic MTC (sMTC) and is more often associated with C-cell hyperplasia as well as multifocality or bilaterality. There is a genotype-phenotype correlation with the age of onset of MTC being associated with the underlying RET mutation.

 

PHEOs are catecholamine-secreting tumors that arise from chromaffin cells of the adrenal medulla. The frequency of PHEO depends upon the underlying RET mutation. As with MTC, it manifests earlier in MEN2, compared to sporadic forms. PHEO usually present after MTC or concomitantly; however PHEO can be diagnosed before the (not clinically detected) MTC in 13-27% of individuals with MEN2A (7,8).

 

pHPT is suspected in patients with elevated serum calcium concentrations, in combination with a parathyroid hormone (PTH) concentration that is elevated or within the normal range but inappropriately given the patients hypercalcemia. In MEN2A, pHPT is typically mild and may range from a single parathyroid adenoma to marked parathyroid hyperplasia (9). pHPT usually presents many years after the diagnosis of MTC; the average age at onset is 38 years of age (10).

 

Nearly all patients with MEN2A have either C-cell hyperplasia (CCH) or MTC, approximately 50% have a PHEO and 20-30% have pHPT, but the incidence of these manifestations depends on the underlying RET mutation (11,12). Patients with MEN2B have a 100% incidence of CCH and MTC, frequently have PHEO (30-50%), and typically have physical characteristics including mucosal neuromas, intestinal ganglioneuromatosis (IGN), alacrima (the lack of tears), and hyperflexible joints. They rarely have pHPT. Both MEN2A and MEN2B have autosomal dominant transmission patterns and therefore children of affected individuals have a 50% chance of inheriting the genetic abnormality. However, MEN2B most frequently occurs as a de novo mutation. Offspring of MEN2B patients have not frequently been reported.

 

Early diagnosis of affected patients and families is critical to obtain the best outcomes. For MEN2A, genetic screening in individuals at risk enable early screening. For MEN2B awareness for and early recognition of presenting syndromes (IGN, alacrima, mucosal neurinomas) is of utmost importance. Management of MEN2 patients is challenging, and the decision making is often not straightforward, as impact of screening and early surgery have to be balanced with possible benefits, particularly in younger patients. Patients and families suspected of harboring or diagnosed with MEN2 should be evaluated by an experienced multidisciplinary team.

 

Classification

 

Close to 200 RET germline variants have been identified with a clear genotype-phenotype correlation (13,14). Two distinct clinical syndromes are recognized within MEN2 syndrome: MEN2A (95%) and MEN2B (5%). MEN2A is categorized in four subtypes.   

 

  1. Classic MEN2A
  2. MEN2A with cutaneous lichen amyloidosis (CLA)
  3. MEN2A with Hirschsprung’s disease (HD)
  4. Familial medullary thyroid cancer (FMTC)

 

Incidence and prevalence rates vary with the population studied. Based on an analysis of Surveillance, Epidemiology, and End Results (SEER) data, MEN2A is most common, with an incidence of about 1 patient in 2 million, compared to MEN2B with an incidence of about 1 patient in 39 million (12). A nationwide study in Denmark described higher rates with MEN2A incidence of 28 per million live births per year and a point prevalence of 24 per million (15). MEN2B had an incidence in Denmark of 2.6 per million per year and a point prevalence of 1.06 per million. The patient distribution among the subtypes varies with the series and the population studied as well. A large series from China showed that of 65 families (214 patients) with MEN2, 30 (46%) families had classical MEN2A, 5 (8%) had MEN2A with CLA, 1 (2%) MEN2A with HD, 24 (37%) had FTMC, and 5 (8%) had MEN2B (16).

 

Previously, patients with FMTC were classified as a separate entity, because affected families appeared to have MTC alone, lacking other associated endocrine or neural-tissue involvement of MEN2. With extended follow-up, some members in many of these families developed PHEO or pHPT. Data such as these suggested FMTC patients might be more appropriately classified as a variant of MEN2A (12). Furthermore, distinguishing classical MEN2A from FMTC is difficult, making the clinical relevance of this phenotype limited.

 

In some families, MEN2A was associated with CLA (17). This might be limited to families with specific mutations in the RET gene, with C634 as most frequently described (table 1). Not all family members with the same mutation develop CLA and there is a variation in clinical appearance, although the scapular region of the upper back is typically affected in MEN2A (see clinical features). CLA can precede the other MEN2A manifestations (17).

 

The same goes for the association of MEN2A and HD. This association is especially challenging, since the type of mutation leading to the different phenotypes result in different effects in the RET gene (a gain of function in the MEN2A phenotype and a loss of function in the HD phenotype). Pathogenic RET variants are mainly associated with both these conditions when located at the 620 position. Mutations that impair RET signaling cause HD owing to failure of enteric neural crest-derived cells to migrate, proliferate and/or differentiate properly within the intestine. This seemingly paradoxical occurrence has led to speculation of a ‘Janus mutation’ in RET that causes overactivation or impairment of RET activity depending on the cellular context. In animal studies an alternative explanation was suggested that the coexistence of these two seemingly opposite phenotypes can be explained by excessive RET signaling alone and without invoking a Janus mutation. In this study it was also demonstrated that the cells comprising the ganglioneuroma-like masses induced by RET activation maintain their embryonic potential to generate an enteric nervous system, suggesting that these ganglioneuromas, and possibly other MEN-associated neoplasms, may be amenable to reprogramming along normal developmental trajectories (18).

 

GENETICS

 

The RET proto-oncogene (OMIM 164761) encodes one of the receptor tyrosine kinases, cell-surface molecules that transduce signals for cell growth and differentiation (6). The RET gene was defined as an oncogene by a classical transfection assay. RET can undergo oncogenic activation in vivo and in vitro by cytogenetic rearrangement (19). The RET protein comprises an extracellular domain, a transmembrane segment that traverse the plasma membrane, and the intracellular domain that consist of the intracellular juxtamembrane segment and the tyrosine kinase domain (TKD). Activation of RET is complex and occurs throughout a binding of a ligand-coreceptor composition, leading to RET homodimerization, resulting in activation of several downstream pathways, including RAS/MAPK and PI3K/AKT pathways (20). 

 

The oncogene contains 21 exons spanning more than 60kb of genomic DNA. It is expressed in tissues of neural crest origin, and appears to have an important function in cell migration and development. RET germline mutations in MEN2 typically result in constitutive activation. Different mutations lead to different levels of activation, which may have an effect on the clinical spectrum. In 95% of patients with MEN2A syndrome, germline RET mutations cluster in cysteine C609, C611, C618, C620 (in exon 10), or C630 and C634 (in exon 11), with mutation of C634 being the most frequent. These mutations, leading to affected cysteines in the extracellular domain of the RET receptor due to replacement of other amino acids in the cysteines, cause dimerization of receptor molecules, enhanced phosphorylation and thus ligand-independent activation of intra-cellular pathways (14,21). Germline pathogenic variants in the intracellular tyrosine kinase domain of RET are less frequent and encoded in exon 13-16. MEN2B is almost exclusively associated with a mutation in RET exon 16, which causes a methionine to threonine (M918T) substitution within the activation segment of RET kinase. This substitution increases ATP-binding and auto phosphorylation activity, thereby mediating a dimerization-independent activation of RET kinase. In less than 10% of MEN2B patients, other mutations are found: the pathogenic A883F variant (exon 15) encoded in exon 15 of RET also leads to increased and independent activating of RET. Furthermore, very rare dual germline variants (E768D/L790F, V804M/Q781R, V804M/E805 K, V804M/Y806C) have been described to cause MEN2B (22,23).  

 

Since the discovery of the RET receptor tyrosine kinase in 1985 (24), somatic alterations of this protein have also been found in sporadic tumors, like non-small cell lung cancer (1-2%), papillary thyroid carcinoma (10-20%) and sMTC (21,25). Somatic RET mutations have been found in up to 60% of patients with sMTC. The prevalence is higher in patients with large tumors, and up to 85% of patients with distant metastases have somatic RET mutations (21). The most common somatic mutation is M918T, which is present in up to 40% of patients with sMTC and is associated with disease aggressiveness. Other single amino acid changes might occur at residues C611, C618, C620, C630, C634, E768, A883 and S891; small RET deletions and/or insertions have also been detected.

 

Genotype-Phenotype Correlation

 

Genotype-phenotype correlations in MEN2 are well-established and have long been used to guide clinicians in making medical management recommendations. Currently, patients are classified based on their phenotype into three groups, according to the American Thyroid Association (ATA) guideline (table 1) (12):

 

  • Highest risk: classic MEN2B - M918T carriers
  • High Risk: patients with the RET codon C634 mutations and the RET codon A883F mutation
  • Moderate risk includes patients with hereditary MTC (hMTC) and RET codon mutations other than M918T, C634, and A883F

 

These risk categories are based on the aggressiveness of MTC and used as a guidance for timing of prophylactic thyroid surgery (see therapy). Furthermore, there is evidence that certain mutations are more commonly associated with PHEO. Besides M918T and A883F, both leading to MEN2B, all C634 mutations and D631Y mutations have a high risk of PHEO (~50%). Some mutations, like RET variant R912P and E768D are not associated with PHEO. For pHPT, C634 mutations have the highest risk as well. A clear overview of all manifestation per RET variant is illustrated in table 1. Despite a higher incidence of the various manifestations in some mutations, current data remain insufficient to determine the risk of PHEO or pHPT in a given patient or family, since there is also clear, yet unexplained intrafamilial variability. This suggests a role for genetic modifiers, such as polymorphisms/haplotypes. This leads to the suggestion that multi-step carcinogenesis may be applicable to patients with MEN2, with other non-RET genetic changes or modifiers responsible for the phenotypic differences. Limited evidence demonstrated that copy number variations (CNVs) play an important role in phenotypic expression, but other possible contributing factors have been evaluated as well (13,26). Since the observed intra- and interfamilial variability is still poorly understood, a possible divergent course of disease cannot be predicted in an individual patient. Thus, all patients need to be followed for the development of these tumors. Furthermore, ongoing evaluation of new data will be needed to update the risk categories and genotype-phenotype correlations periodically.

 

Table 1. Incidence and Occurrence of MEN2 Manifestations in Relation to Different Germline Mutation and the Advised Management According to ATA Guideline 2015 (12)

ATA risk category

RET Mutation

MTC timing surgery

Incidence

of PHEO

Incidence of pHPT

Start

PHEO/pHPT

Screening

Occurrence

CLA / HD

Highest

M918T

Within first year

50%

-

At age 11 (PHEO only)

- / -

High

C634  

A883F

At 5 years or earlier if elevated calcitonin

50%

50%

20-30%

-

At age 11

 

+ / -

- / -

Moderate

All others

- C609 F/G/R/S/Y

- C611F/G/S/Y/W

- C618F/R/S

- C620F/R/S

- C630R/Y

- D6311Y

- E768D

- G553C

- K666E

- L790F

- R912P

- S891A

- V804L

- V804M

When calcitonin becomes elevated or prophylactic before apparent disease

 

10-30%

10-30%

10-30%

10-30%

10-30%

50%

-

10%

10%

10%

-

10%

10%

10%

 

10%

10%

10%

10%

10%

-

-

-

-

-

-

10%

10%

10%

At age 16

 

- / +

- / +

- / +

- / +

- / -

- / -

- / -

- / -

- / -

- / -

- / -

- / -

- / -

+ / -

Abbreviations: ATA, American thyroid association; RET, REarranged during Transfection; MTC, medullary thyroid carcinoma; PHEO; phaeochromocytoma; PHPT, primary hyperparathyroidism; CLA, cutaneous lichen amyloidosis; HD, Hirschsprung’s disease; +, yes; -, no.

 

CLINICAL FEATURES

 

MEN2A and MEN2B share the same genetic defect and common manifestations but also have specific clinical features and course of disease, making them two separate clinical entities. The occurrence of multicentric tumor formation in the thyroid and adrenal glands is shared. However, MEN2B is characterized by a specific clinical phenotype with ganglioneuromas of the lips, tongue and conjunctiva, musculoskeletal abnormalities, narrow long face, and thickened lips, among other features (see below). Clinically relevant pHPT is absent in MEN2B. In general, patients with MEN2B will develop tumors at an earlier age and these tumors will show a more aggressive behavior(26). Biochemically, the tumors that arise in patients with MEN2 are similar to those with the sporadic forms and discrimination cannot be made based on biomarkers.  

 

MEN2A 

 

Virtually all patients with MEN2A will develop CCH or MTC, but there is much inter- and intra-familial variability in the other manifestations. Approximately 50% will develop a PHEO and 20-30% will develop pHPT, but the incidence depends on the underlying mutation (table1). No clear clinical phenotype is present to recognize MEN2A patients. Most index patients present with MTC as the first manifestation (27). Some patients will develop HD which then will reveal the diagnosis of MEN2 (table 1). CLA is dermatologic disease, typically located in the interscapular region of the back and is characterized by secondary skin changes (papular, pigmented) and intense pruritus (figure 1). It becomes apparent during late adolescence or young adulthood and can be the first presenting manifestation as well. For mutations associated with CLA, see table 1.

Figure 1. Cutaneous lichen amyloidosis.

MEN2B 

 

All MEN2B patients will develop CCH or MTC, and 30-50% of them will have a PHEO (table 1). In the MEN2 literature, there is often reference to a characteristic Marfanoid physical appearance with hyperflexible joints but without the Marfanoid lens or aortic abnormalities. However, no studies have been published describing body proportions in MEN2B. A recent case series from the Netherlands described 8 MEN2B patients: all children and adults had normal body proportion(28). The typical marfanoid appearance in MEN2B is therefore questionable.

 

MEN-2B patients can have mucosal neuromas of the eyelids, lips, and tongue, and wide-spread ganglioneuromatosis of the gastrointestinal tract resulting in an abnormal gastrointestinal motility with complaints of diarrhea, constipation, colonic dilatation, or even megacolon at a young age (29).   

 

As most MEN2B patients present with de novo mutations, the diagnosis is almost always delayed, even in the presence of clinical features (29,30). Recognition of nonendocrine symptoms like ocular symptoms (tearless crying) or the highly penetrant oral manifestations is important. Recent studies re-emphasize neonatal gastro-intestinal manifestations due to ganglioneuromatosis as the most important early feature for MEN2B diagnosis while body proportions and stature were non-specific in children with MEN2B (28,31). Those diagnosed based on nonendocrine symptoms instead of symptomatic MTC or PHEO were significantly younger (mean of 5.3 vs. 17.6 years), emphasizing the importance of early recognition. 

 

Medullary Thyroid Carcinoma (MTC)

 

MTC is a tumor originating from the parafollicular cells (C-cells) of the thyroid. The production of calcitonin is a characteristic feature of this tumor. Most patients present with a solitary nodule or cervical lymphadenopathy. Compared to sMTC, hMTC occur at a younger age and has typically multifocal and a bilateral pattern. It is often localized in the middle to upper regions of the thyroid lobe (32). In sMTC, up to 60% of cases appear to harbor a (driver) RET mutation, amongst other mutations, which has therapeutic implications (21). hMTC is preceded by CCH (33). The age-related penetrance of CCH and hMTC is mutation dependent. Most MEN2A patients (85%) are asymptomatic at MTC diagnosis, while 15% have presenting complaints (34). Especially in advanced disease diarrhea can be present. Peak incidence in index patients is in the third decade of life in MEN2A.

 

In MEN2B, MTC occurs very early, over 80% have MTC in their first year of life (30), underscoring the necessity of thyroidectomy before the age of 1. Unfortunately, the substantial diagnostic delay due to the high proportions of de novo mutations, median age of thyroidectomy in MEN2B patients is 14 years (30). The outcome of MTC in MEN2B is correlated to the way the diagnosis is made. If the diagnosis is made after recognition of nonendocrine symptoms, patients are significantly younger at diagnosis and therefore have often less lymph node metastases (43% vs 100%) or distant metastases (8% vs 79%) and were more often biochemically cured after treatment (58% vs 0%) (35). If the diagnosis is made before the age of 1 year, when there is only a preliminary stage of MTC, patients can be cured.  

 

Pheochromocytoma (PHEO) 

 

PHEOs are catecholamine-secreting tumors that arise from chromaffin cells of the adrenal medulla. Prevalence of PHEO in MEN2A is 17-42% compared to 50% in MEN2B with median age at diagnosis 42 and 24 years respectively (13). Extra-adrenal PHEO is very rare in MEN2. Tumoral hypersecretion of epinephrine and norepinephrine causes episodic headache, sweating, and tachycardia with 50% of MEN2 patients being symptomatic. PHEO was the first presenting manifestation in 25% in MEN2A patients and 6% in MEN2B (8). PHEO is bilateral in most cases and rarely malignant (0-4%) (8,13). MEN2-related PHEO typically produce epinephrine or both epinephrine and norepinephrine, but not exclusively norepinephrine (36).

 

Primary Hyperparathyroidism (pHPT) 

 

pHPT is caused by a parathyroid adenoma or hyperplasia with autonomous secretion of PTH, leading to elevated calcium levels. Disease is multiglandular in the majority of patients, but can occur metachronous, with long intervals with normocalcemia, simulating single gland disease (37). The prevalence in MEN2 is RET-mutation dependent, varying from 0-35% and pHPT is rarely the first manifestation with median age at diagnosis between 35-46 years of age (23,38). Since most MEN2 patients have mild pHPT, most patients are asymptomatic at diagnosis (56-88%). Symptoms of pHPT are often nonspecific, like constipation, fatigue, depression, anorexia, nausea, and polyuria. pHPT is rarely seen in MEN-2B patients. 

 

Cutaneous Lichen Amyloidosis (CLA) 

 

CLA is an uncommon disease characterized by pruritic lichenoid papules and 

occurs rarely in MEN2A patients. In sporadic cases they occur in the pretibial bilateral regions, and in MEN2A patients the lesions typically occur in the (inter)scapular regions (fig 1) (39). Although MEN2A patients rarely develop CLA, if it occurs this will be at a mean age of 20 years and is often 11 years prior to the diagnosis of MEN2A in index patients. 

Therefore patients with an atypical location of CLA must be considered for RET testing (39). 

 

Hirschsprung’s Disease (HD) 

 

Most patients with HD will present shortly after birth and those with exon 10 RET mutation must be screened for MEN2A (12). Older MEN-2A patients with exon 10 RET mutations and colonic symptoms must be evaluated for HD.

 

DIAGNOSIS AND SCREENING

 

The diagnosis of MEN2 is established when a RET pathological variant is detected by molecular genetic testing. Genetic screening can identify gene carriers with high accuracy (98%) (40). Sanger or next generation sequencing are the recommended methods to detect RET mutations in exon 10 (codons 609, 611, 618, and 620), exon 11 (codons 630 and 634), and exons 8, 13, 14, 15, and 16 (12). For MEN2B, the M918T mutation (exon 16) and A883F mutation (exon 15) should be evaluated. If initial sequence analysis is negative, and the clinical suspicion of a genetic syndrome remains high based on family history, physical characteristics, young age at diagnosis, or pathologic findings in the thyroid such as extensive CCH or bilateral MTC, then whole gene sequencing should be considered (12). Patients who present with MTC, even with a negative family history need to be screened for RET mutations, as up to ~25% will be found to have a hereditary syndrome. At least 33% of all PHEO patients have a familial disorder, affected succinate dehydrogenase genes, Von Hippel Lindau (VHL) and MEN2 are the most frequently identified syndromes (41,42). The relative percentage of underlying germline mutation depends on the age of onset, family history, or clinical features like multifocal-, bilateral- or metastatic disease, but genetic screening need to be considered in all patients with PHEO. Those with other clinical characteristics, associated with MEN2, such as HD or CLA in the interscapular/scapular region should also be considered for testing (12). Those with physical characteristics suggestive of MEN2B such as mucosal neuromas and alacrima also need to be considered for genetic screening. Accurate evaluation of early onset severe constipation can lead to the detection of intestinal ganglioneuromatosis. These patients need to be screened as well for MEN2(31). A negative family history is not reliable in excluding patients from genetic testing, since about 40% of MEN2A gene carriers do not develop clinically apparent disease. De novo mutations are rare in MEN2A (up to 10%), in contrast to MEN2B, where de novo mutations are more frequent (45% of A883F carriers and 84% M918T carriers), emphasizing the limited value of a negative family history in MEN2B (13). First-degree relatives of patients with proven MEN2 should be offered genetic counseling. All patients of reproductive age carrying RET mutations, particularly those with mutations in codon 634 and 918, should be offered genetic counseling and be informed on the benefits and the potential risks of reproductive options, such as prenatal diagnosis and preimplantation diagnostic testing (12). Furthermore, the option of genetic testing in offspring should be discussed with future parents, where testing either directly after birth or at an older age could be discussed, based on the specific mutation in the family as well as individual preferences of the parents.

 

Patients at risk or with MTC, where genetic testing is not possible, should be under surveillance for MTC, PHEO and pHPT. There are very rare families who meet the clinical criteria for MEN2A (one or more first-degree relatives have characteristic clinical features for MEN2A), where no causative pathogenic variant in RET can be found. In these families, the periodically screening for MTC, PHEO and pHPT should be considered in the first degree relatives at risk (12).  

 

Once a mutation is identified, the carrier should be screened for related manifestations, e.g., MTC, PHEO and pHPT. Prophylactic thyroidectomy is the mainstay of the treatment with the risk classification of the mutation defining the optimal age (see section on surgical treatment for MTC). Annual biochemical screening for all three manifestations is recommended, combined with neck ultrasound and physical examination, starting at an age based on the ATA risk category (12). Patients with M918T mutation should have a thyroidectomy within the first year of life. Patients with an ATA high risk often develop MTC in the first years of life as well, so annual screening from the age of 3 with physical examination, serum calcitonin, and cervical ultrasound is recommended by the current ATA guideline (12). The phenotypes in the ATA moderate risk category varies significantly and the development of MTC is at a later age in most patients, but clinical MTC can occur before the age of 10 in this group as well. Therefore, annual screening for MTC is advised from the age of 5 (12). Family members, who have no pathological RET variant, do not need to undergo biochemical testing.

 

Medullary Thyroid Cancer

 

For MTC, biochemical testing is similar to patients with sporadic disease. Patients with clinical MTC have elevated serum calcitonin. Calcitonin is a neuropeptide derived from the parafollicular cells (C-cells) of the thyroid and calcitonin levels are directly related with C-cell (tumor) volume (43,44).

 

Patients with CCH or subclinical MTC usually do not have elevated basal serum levels of calcitonin. Physicians screening patients for CCH or subclinical MTC must be thoroughly familiar with the particular calcitonin assay being used, as normal ranges vary. Reference ranges for calcitonin differ among laboratories, and are also gender and age dependent (45,46). Calcitonin is higher in boys and in both sexes, a significant decrease in calcitonin levels is observed after the second year of life. Several studies defined age-, and gender specific calcitonin levels in a pediatric population, but since the normal range of calcitonin varies between the different assays and laboratories, basal calcitonin is of limited help in the initial management of very young children and infants (45,46). Since, there are no globally accepted calcitonin cutoff levels to predict MTC, each institution has to define its own reference ranges and clinicians should use the same laboratory and essay for serial measurements. Elevated calcitonin levels are an indication for thyroidectomy.  

 

Provocative tests like pengastrin- or calcium stimulated calcitonin tests have no added value anymore since identification of mutation carriers is replaced by genetic testing and the newest immunochemiluminometric calcitonin assays are highly sensitive and specific for monomeric calcitonin. A recent study by Niederle, et al. illustrated no added value of calcium-stimulated calcitonin compared to basal calcitonin in the diagnosis of (sporadic) MTC or to differentiate between patients with CCH and micro-MTC in those with only mildly elevated calcitonin levels (47).

 

Pheochromocytoma

 

Screening for PHEO should be performed in all MEN2 patients prior to therapeutic thyroidectomy, as well as in female MEN2 patients who are considering pregnancy, to avoid a potential hypertensive crisis. Furthermore, annual screening for PHEO should be performed in all children in ATA highest and high risk categories from age 11 onwards, and in moderate risk children by age 16 (table 1) (12). PHEO is likely when elevated plasma concentrations of free metanephrines or elevated 24-hour urinary fractionated metanephrines and normetanephrines are detected. Practitioners should be aware of the pitfalls, conditions of sampling and possible influencing factors when analyzing the (nor)metanephrines to minimize the risk of false-positive results (42). MEN2-related PHEO produce epinephrine or both epinephrine and norepinephrine, but not exclusively norepinephrine (36).

 

When there is biochemical evidence for PHEO, imaging studies need to be initiated to locate the PHEO. Computed tomography (CT) or magnetic resonance imaging (MRI) of the adrenal glands is the first choice of imaging. CT has a high sensitivity (93-100%) in detecting intraadrenal tumors > 5 mm(36).  

 

Primary Hyperparathyroidism

 

Screening for pHPT is similar to those with sporadic disease and includes measurement of (ionized) calcium or serum calcium with albumin and intact PTH. Additional evaluation and follow up of bone density and kidney function in patients with pHPT is advised. ATA guidelines recommend that annual screening start at the same time as screening for PHEO: at age 11 for those in the high risk category and at age 16 for those in the moderate risk category (table 1)(12). Once pHPT is diagnosed, imaging studies are advised to visualize enlarged parathyroid glands. Neck ultrasoundshould be part of the diagnostic strategy. Combined radiology techniques increase localization accuracy, so the combination of neck ultrasound with parathyroid scintigraphy with Technetium (Tc) 99m sestamibi, 18F-fluorocholinePET/CT, or 4-dimensional CT is recommended. The type of imaging to use must be based on knowledge of the clinician regional imaging capabilities and experience (48).

 

Given the intra- and interfamilial variability in PHEO and pHPT incidence, a static surveillance was advised by the ATA guideline. Some authors have suggested to tailor the frequency of biochemical screening for PHEO and pHPT, based on age and ATA risk category (49). However, large prospective studies are needed to validate their findings on age-related penetrance before a more tailored surveillance by age can be applied.

 

SURGICAL MANAGEMENT

 

PHEO may cause a dangerous hypertensive crisis during surgery (12,50). Therefore, PHEO should be excluded preoperatively in all patients. Very young patients who undergo prophylactic thyroidectomy may be excluded from this advice, since PHEO is not described in patients <8 years old (51). If present, the PHEO should be resected prior to surgery for either MTC or pHPT (12).

 

Adrenalectomy

 

Surgical resection is the cornerstone of treatment for PHEO (52). To prevent hypertensive crisis during surgery, caused by vasoconstriction due to catecholamine release, adult as well as pediatric patients should be prepared preoperatively during 1-2 weeks using α-adrenergic blockade (52–54). In case of tachycardia, additional treatment with β-adrenergic blockade should be started (52,54). A minimally invasive technique such as the laparoscopic or retroperitoneoscopic approach is recommended (12,52). The transabdominal and retroperitoneal approach take place in the lateral and prone position, respectively. There is increasing evidence in favor of the retroperitoneoscopic approach due to less blood loss, shorter operation time and length of stay, and less postoperative pain (55,56). Sixty-five percent of MEN2 associated PHEO are initially bilateral (54). The majority of patients with unilateral PHEO develop contralateral disease within 10 years (12,54). To prevent lifelong steroid dependency, adrenal cortical function should be preserved for as long as possible. Therefore, for patients with unilateral PHEO, unilateral resection is the treatment of choice, despite the high chance of developing a contralateral PHEO (12). Moreover, cortical function can be preserved by performing a subtotal adrenalectomy. This is appropriate for patients with bilateral, as well as unilateral PHEO (12,57). An international retrospective study showed excellent results in 563 MEN2 patients of who 114 (21%) underwent adrenal-sparing surgery. Steroid dependency was avoided in 57% of patients after adrenal-sparing surgery for bilateral PHEO. Recurrence of PHEO after adrenal-sparing surgery occurred in 3% of the 153 operated glands (57).

 

Parathyroidectomy

 

The amount of affected parathyroids in MEN2A patients varies from a single to all glands (5). Only the enlarged glands should be resected (12,48). Due to the short half-life (3 minutes) of PTH, intraoperative PTH monitoring can help determine whether parathyroidectomy has been successful. If PTH values remain elevated, it is necessary to look for other enlarged parathyroids (48). The surgical approach is tailored to the amount and location of the enlarged glands. In case of one affected parathyroid, minimally invasive adenomectomy is the treatment of choice. In case of more enlarged glands a conventional exploration is performed. If all (four) glands are enlarged, a part of one parathyroid should be left in situ on a vascular pedicle or transplanted heterotopically to preserve parathyroid function (12,48). In some cases, pHPT is diagnosed at the same time as MTC. For these patients, thyroidectomy and parathyroidectomy can be performed during one surgery.

 

Preoperative imaging and marking of the location of the parathyroid adenoma is essential to perform minimally invasive adenomectomy. The incision is made right above the parathyroid, to reduce incision length and limit dissection. Conventional neck exploration is a more extensive procedure. After incision of the skin and passage of the platysma, the linea alba colli is dissected and the strap muscles are lateralized. Hereafter, mindful of the recurrent laryngeal nerve, the parathyroids can be identified and removed beyond the thyroid (58).

 

Thyroidectomy

 

Surgery is the only curative option for patients with MTC and remains the cornerstone of MTC treatment. Since virtually all MEN2 patients develop MTC, the question is not whether MEN2 patients should undergo thyroidectomy, but at what age? To prevent recurrence, it is essential that the thyroid is entirely removed, given the multifocal and bilateral growth in inherited MTC. If possible, thyroidectomy should be performed prophylactically, before developing (clinically relevant) MTC (12). Central neck dissection is not indicated in MEN2A patients with normal calcitonin and neck ultrasound undergoing prophylactic thyroidectomy. Genetic screening in family members of RET mutation carriers have led to the early diagnosis of MEN2 in a significant proportion of patients. Early prophylactic thyroidectomy in these patients is associated with excellent results and minimal operative morbidity: biochemical cure rates approximating 100% over 7-16 years of follow up (59,60). However, timing of thyroidectomy in known mutation carriers is challenging as risk of surgery in younger infants should be balanced with the probability of curing the patient. Surgery in (young) pediatric patients should be performed in specialized centers by experienced surgeons. Current decision making is mostly based on specific RET mutation (ATA risk category), age, and calcitonin levels (12,61).

 

Patients in the highest ATA risk category (table 1) should undergo total thyroidectomy within the first year of life. Timing of surgery in this group cannot be based on calcitonin levels since calcitonin levels are naturally high in the first months after birth. Risk of complications, especially the risk of hypoparathyroidism due to the inability to identify the parathyroids, is increased in children and infants (62). Therefore, if there are no suspicious lymph nodes and the parathyroids cannot be identified, central neck dissection might not be necessary.

 

The ATA high risk category consists of patients with RET codon C634 and A883F mutation. Children in this category should undergo screening for possible MTC from the age of three. These patients should undergo total thyroidectomy before the age of five, or earlier based on their calcitonin values (12). Considering the risk of complications, surgery before the age of three is not advised in this group (63). If lymph node metastases are suspected or calcitonin levels are >40 pg/ml, central neck dissection is needed.

 

Patients in the ATA moderate risk category generally develop a less aggressive type of MTC at an older age. These patients should be screened every 6-12 months from the age of five and should undergo thyroidectomy in childhood or early adulthood primarily based on calcitonin levels. Alternatively, thyroidectomy can be timed before calcitonin is elevated as yearly screening and calcitonin elevation might impose a psychological burden. Timing of thyroidectomy should be in consultation with child’s parents and involved pediatricians and surgeons. ‘Prophylactic’ thyroidectomy is advised from the age of five in this moderate risk group (12).

 

The best calcitonin cut off point to prevent loco regional disease varies between studies. A large French multicenter study showed that no lymph node metastases were detected when calcitonin was <31 pg/mL while a Norwegian study found that all patients were cured when calcitonin was <40 pg/mL before total thyroidectomy (64,65). These studies on calcitonin levels illustrate that progression from CCH to MTC is imminent once calcitonin levels exceeds the upper limit of normal and the ‘window of opportunity’ to perform surgery without addition of extended node dissection is closing. Therefore, thyroidectomy should be performed once calcitonin levels exceed the upper limit of normal (12).   

 

For patients with de novo mutations or unknown MEN2, MTC is often already present at first presentation. In adults with normal calcitonin values, the ATA guideline advises yearly screening and surgery when calcitonin becomes elevated (12). Two studies show that calcitonin values <20 and <60 pg/ml are associated with intrathyroidal MTC, and that calcitonin can be safely used to determine timing of surgery (61,66). Unfortunately, these patients usually present with higher calcitonin values and advanced disease. Ultrasound of the neck is performed prior to therapeutic surgery as it may give important information as to the extent of macroscopic thyroid and lymph node disease, not apparent on clinical exam. Patients with clinically apparent MTC on either clinical exam or neck ultrasound should undergo screening for metastatic disease, with attention to the most common sites such as the lungs, mediastinal lymph nodes, liver, and bones. Patients with markedly elevated serum calcitonin levels (>500 pg/ml) or extensive neck disease should also be screened for distant metastatic disease.   

 

The standard treatment for MTC consists of total thyroidectomy and central neck dissection. There is no controversy regarding the need for central neck (level 6) lymph node dissection for MEN2 patients with clinically apparent MTC. Metastases to central neck lymph nodes are noted in up to 81% of patients with palpable tumors (67,68).

 

In case of cervical lymph node metastases in the lateral compartment of the neck, patients should undergo additional dissection of the lateral neck compartments (levels II–V) (12). Unfortunately, patients with cervical lymph node metastases can only be cured by total thyroidectomy and extensive lymph node dissection in 10% of cases (54,69,70). Lateral neck dissection in the absence of clinical or radiologic pathological lymph nodes (prophylactic lateral dissection) has no prognostic value (71). Patients with distant metastases cannot be treated with curative intent. In case of poor prognosis and locally advanced disease invading the surrounding structures, a less aggressive palliative resection should be considered for the purpose of local control and preservation of quality of life (12).

 

Standard procedure for thyroidectomy is the open approach. The patient’s neck is positioned in hyperextension. After Kocher incision 1 cm above jugulum, the thyroid is visualized by dissection of the platysma, opening of the linea alba colli and lateralization of the strap muscles. The thyroid is dissected and mobilized starting at the superior or inferior pole, depending on the surgeon’s preference. The thyroid vasculature should be ligated, clipped or sealed. Identification and preservation of the recurrent laryngeal nerve (located in the tracheoesophageal groove) and parathyroids (located posterior to the thyroid) is essential to prevent complications. After mobilization of one lobe, the same procedure is performed on the contralateral lobe. Thereafter, dissection of the central neck is performed (72,73).

 

RECURRENT OR METASTATIC DISEASE

 

Surveillance After Surgery

 

Patients who underwent surgery for one of the manifestations should be screened for disease recurrence. For PHEO and pHPT, screening is the same as initial screening, i.e. annual biochemical evaluation with plasma fractionated metanephrines and calcium (see diagnosis and screening), with the first evaluation 2-4 weeks after surgery, followed by imaging in case of biochemical evidence for disease (42).

 

After thyroidectomy, the risk of recurrence is based on the American Joint Committee on Cancer (AJCC) Tumor, Node, Metastases (TNM) classification, the total number of metastatic lymph nodes resected and pre- and postoperative calcitonin (12,64,65,69,74). Since calcitonin is a marker for tumor volume, pre- and post-operative calcitonin correlate with the extend of disease. A fairly standard post-operative follow up regimen to detect recurrent MTC includes physical examination, neck ultrasound, and measurement of serum calcitonin and carcinoembryonic antigen (CEA) levels every 6-12 months, depending on the initial postoperative findings (12). Post-operative calcitonin should be measured 3 months after surgery and undetectable following complete removal of thyroid tissue (75).

 

In patients with undetectable post-operative calcitonin, surveillance should be repeated after 6 months and then every 12 months if it remains undetectable. These patients are considered biochemically cured with 10-year disease specific survival rate of 100% (61).

 

In patients with detectable calcitonin <150 pg/ml, persistent disease is mostly confined to cervical lymph nodes. Calcitonin should be repeated every 6 months in these cases together with ultrasound of the neck to detect persistent disease (12,76). Fine-needle aspiration for cytology can be used to confirm recurrence or residual disease.

 

Post-operative calcitonin >150 pg/ml is an indication for evaluation of distant metastasis by imaging procedures (CT or MRI) with focus on the lungs, liver and axial skeleton. The clinical utility of PET/CT with various radiopharmaceutical tracers in MTC is limited (77). Currently, only 18F-FDOPA PET/CT have an acceptable sensitivity for the detection of distant metastatic disease in MEN2 with a patient detection rate of 66% in patients suspected of recurrent MTC (77,78).      

 

CEA is a non-specific marker which may be elevated in patients with MTC. It is not useful for early diagnosis, but has a role for monitoring disease progression and for detecting recurrence after thyroidectomy. CEA levels should be obtained concurrently with calcitonin measurements. In rare cases, serum CEA increases progressively while calcitonin remains stable or decreases. This suggests cellular dedifferentiation and a more aggressive course of disease, or secondary non-MEN2 related malignancy like colon carcinoma.

 

In case of biochemical or radiological evidence for residual disease, recurrence, or metastatic disease, the tumor growth rate can be estimated from sequential imaging studies using response evaluation criteria in solid tumors (RECIST) or by CEA and calcitonin serial measurements over time (12). Calcitonin and CEA doubling times are efficient biomarkers for assessing tumor progression. In one study, 94% of patients with doubling times within two years showed RECIST defined disease progression (79). Consequently, the doubling time of each biomarker has an important prognostic value, especially when values double within a year, survival rates are much lower compared to longer doubling times (80,81). Disease progression should be confirmed on imaging before initiating systemic treatment.  

 

Treatment of Recurrent or Metastatic Disease in MTC

 

LOCAL RECURRENCE  

 

The treatment of recurrent MTC in patients with genetic syndromes is similar to their sporadic counterparts and remains challenging. In patients with disease confined to the neck, an aggressive surgical approach may be considered since surgery is the only curative treatment in MTC. There is generally little role for external beam radiation therapy (EBRT) to the neck. The intent of EBRT is to achieve local control since there is no survival benefit demonstrated (82,83). The recent ATA guidelines suggest that EBRT may be helpful in selected circumstances where the risk of local recurrence is felt to be high (12). These circumstances are very limited since the benefits must outweigh toxicity and the potential for making subsequent neck re-exploration, if required, more difficult. Post-operative radiation therapy is reasonable in those patients in whom there is gross residual disease, particularly if there is a concern for potential airway compromise.

 

METASTATIC DISEASE

 

Patients with metastatic disease should be carefully evaluated and the course of disease must be determined to optimize and tailor treatment. The timing of treatment initiation and the choice of treatment depends on the rate of disease progression and symptoms versus the quality of life, treatment efficacy and toxicity.

 

In patients with an indolent course of disease or low tumor burden without symptoms, a watch-and-wait strategy can be followed. Imaging should be repeated every 6 months in these patients. If calcitonin doubling time is less than 6 months, imaging should be more frequent (84).   

 

In patients presenting with local symptoms or complications such as airway- or spinal cord compression, neurological symptoms, or pathological bone fractures, local treatment modalities such as surgery, glucocorticoid therapy, and/or ERBT must be used before initiating systemic therapy. Treatment of symptomatic bone or brain metastases from MTC is similar to metastases due to other histologic types. Those with painful bone metastases may have a good symptomatic response to EBRT, and bisphosphonate therapy may also be helpful (12,85).

 

In patients with symptomatic or RECIST defined progressive disease and a significant tumor burden, systemic therapy should be considered. A significant tumor burden is defined as multiple lesions >1-2 centimeters in diameter (84).      

 

The choice of first line systemic treatment is not clear, since the armamentarium of therapeutic options is still expanding, direct head-to-head comparison is missing, and availability and approval is varying between countries. There is no role for radioactive iodine (RAI). C-cells do not take up RAI and no benefit was observed in a multicenter study (86). Current options in metastatic MTC are chemotherapy, multikinase inhibitors (vandetanib, cabozantinib) and selective RET inhibitors (pralsetinib, selpercatinib).

 

Data on cytotoxic chemotherapy is heterogeneous and the study population is small in most papers. The response rate is around 20% in most studies and 5FU/dacarbazine regimen or doxorubicin, alone or in combination with cisplatin seems to be the best choice (84). Because of the poor response rates and adverse events, chemotherapy is no longer first line treatment.  

 

Two multikinase inhibitors (MKI) have been approved by both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of MTC patients: vandetanib and cabozantinib. Other multikinase inhibitors are under investigation (23). The kinases of RET and of vascular endothelial growth factor receptor-2 (VEGFR-2) are the main targets in MTC. Vandetanib and cabozantinib target both these kinases among others. Significant increases in progression free survival (PFS) were observed (vandetanib +11 months; cabozantinib +7 months), compared with placebo but toxicity was also significant (toxicity (>grade 2) for vandetanib and cabozantinib: 55% (24%) and 69% (33%)) respectively, leading to discontinuation of treatment or dose reduction in a significant proportion of study participants (87,88). Studies in hMTC showed better response rates. Vandetanib had an overall disease control rate of 73% in 30 patients with hMTC with an estimated PFS of >27 months (89). In 17 adolescents and children with MEN2B-associated MTC, 10 had partial response and 6 stable disease with vandetanib (90). PFS was prolonged with cabozantinib versus placebo (60 vs 20 weeks) in a subgroup of patients with RET mutations (91).

 

Selpercatinib and pralsetinib are selective RET kinase inhibitors with more specific RET-targeting. This have led to an improved side effect profile and higher response rates in phase I/II studies (92,93).

 

Selpercatinib (former LOXO-292) showed a 69% response rate and 82% 1-year PFS in 55 patients with progressive RET-mutant MTC who had previously received vandetanib or cabozantinib. Response rate in MKI treatment naïve patients (n=88) was 73% and 1-years PFS 92%. Hypertension (21%) and diarrhea (6%) were the most important grade 3 side effects and 2% discontinued medication because of adverse events (92). 

 

Pralsetinib (former BLU-667) is being evaluated in the ARROW trial (ongoing). Response rate in previously MKI treated patients was 60% and 71% in TKI-naïve patients. Serious treatment related adverse events were reported in 15% and 4% discontinued medication.  

 

Several other potential therapies are under investigation, including immunotherapy and peptide receptor radionuclide therapy (PRRT) (23). Despite the advances of MKI over conventional chemotherapy, the clinical utility is still limited. The absence of overall survival benefit and severe toxicity profiles leading to a reduction in quality of life are thresholds for treatment initiation. Drug resistance is another major issue in MKI treatment and still poorly understood. A selective RET kinase inhibitor seems to be a better treatment choice in patients harboring RET germline mutations, regarding the high objective response rates and better toxicity profiles in phase I/II studies. However, trials are still ongoing and large, phase 3 trials are needed before we can determine its true value in the clinical armamentarium.   

 

Treatment of Metastatic Disease in PHEO

 

Malignant PHEO is rare in MEN2 (0-4%). Surgery is the only potential curative option in malignant PHEO. In recurrent or low volume metastatic disease, local inventions such as surgical resection, ablation, or ERBT must be discussed in an experienced multidisciplinary team. For the time being, systemic treatment is similar to sporadic malignant PHEO. With ongoing advances in MKI and selective RET inhibitors, tailored management for MEN2-related PHEO could be possible in the future. Current systemic options in metastatic disease are Iodine-131 metaiodobenzylguanidine (131I-MIBG), PRRT with radiolabeled somatostatin analogues (PRRT), MKI (i.e. sunitinib), or conventional chemotherapy with cyclophosphamide, vincristine, and dacarbazine (13,94).      

 

PROGNOSIS

 

MTC is the major cause of death in MEN2. PHEO does not seem to have an association with shorter survival: median survival is 499 months in patients with PHEO vs. 444 months without (95). Prognosis of patients with sMTC and those with inherited disease is similar after adjustment of age and disease stage at diagnosis in multivariate analysis (34). Together with the presence of certain RET mutations, serum calcitonin, and number of lymph node metastases, these parameters are important factors influencing prognosis. The 10-year overall survival for patients with MTC was 64 % in population based study from Denmark (34)

 

Disease stage is one of the most important prognostic factors: 10-year disease specific survival for patients with MTC is 98%, 93%, 87%, and 53% for disease stage I-IV, respectively (34). In children with MTC, higher disease stage also portends a worse prognosis (90).

 

Age as an independent prognostic factor is preserved in some studies but not all studies (23). A poorer prognosis in older patients may be related to a more advanced tumor stage at diagnosis. The 5-year and 15-year survival rates in children with MTC is 95% and 86%, respectively. Mean survival after diagnosis in children is 28 years (96).  

 

10-year survival rate in MEN2A patients (97%) is better than in patients with MEN2B (76%) which might be influenced by an earlier onset of disease and delay in diagnosis of MTC in (de novo) MEN2B patients (97).

 

Furthermore, as mentioned above, a rapid CEA and/or calcitonin doubling time and high number of lymph node metastases at presentation are all harbingers of an aggressive disease course. 

 

To conclude, the amount of data which has accumulated over the last decade has truly been staggering, and has resulted in significant changes in MEN2A and MEN2B patient management. Further refinements in risk stratification will undoubtedly occur as additional long-term data become available on genotype-phenotype, effects of prophylactic thyroid surgery, and effects of surveillance. Molecular based therapies now offer hope to those with advanced or metastatic MTC. The increasing molecular knowledge and selective RET inhibitors will hopefully lead to new treatment strategies or therapies useful not only for metastatic MTC, but as adjuvant treatment in high-risk patients, or perhaps even in prevention. 

 

REFERENCES

 

1        Donis-keller H, Dou S, Chi D, Carlson KM, Toshima K, Lairmore TC, et al. Mutations in the RET proto-oncogene are associated with MEN 2a and FMTC. Hum Mol Genet. 1993 DOI: 10.1093/hmg/2.7.851

2        Mulligan LM, Kwok JBJ, Healey CS, Elsdon MJ, Eng C, Gardner E, et al. Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature. 1993 DOI: 10.1038/363458a0

3        Carlson KM, Dou S, Chi D, Scavarda N, Toshima K, Jackson CE, et al. Single missense mutation in the tyrosine kinase catalytic domain of the RET protooncogene is associated with multiple endocrine neoplasia type 2B. Proc Natl Acad Sci U S A. 1994 DOI: 10.1073/pnas.91.4.1579

4        Hofstra RMW, Landsvater RM, Ceccherini I, Stulp RP, Stelwagen T, Luo Y, et al. A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature. 1994 DOI: 10.1038/367375a0

5        Takaya K, Yoshimasa T, Arai H, Tamura N, Miyamoto Y, Itoh H, et al. Expression of the RET proto-oncogene in normal human tissues, pheochromocytomas, and other tumors of neural crest origin. J Mol Med. 1996 DOI: 10.1007/s001090050065

6        Arighi E, Borrello MG, Sariola H. RET tyrosine kinase signaling in development and cancer. Cytokine Growth Factor Rev. 2005 DOI: 10.1016/j.cytogfr.2005.05.010

7        Inabnet WB, Caragliano P, Pertsemlidis D. Pheochromocytoma: Inherited associations, bilaterality, and cortex preservation. Surgery. 2000 DOI: 10.1067/msy.2000.110846

8        Modigliani E, Vasen HM, Raue K, Dralle H, Frilling A, Gheri RG, et al. Pheochromocytoma in multiple endocrine neoplasia type 2: European study. Journal of Internal Medicine. 1995. DOI: 10.1111/j.1365-2796.1995.tb01211.x

9        Brandi ML, Gagel RF, Angeli A, Bilezikian JP, Beck-Peccoz P, Bordi C, et al. Consensus: Guidelines for diagnosis and therapy of MEN type 1 and type 2. Journal of Clinical Endocrinology and Metabolism. 2001. DOI: 10.1210/jcem.86.12.8070

10      Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib H, et al. Medullary thyroid cancer: management guidelines of the American Thyroid Association. Thyroid. 2009 DOI: 10.1089/thy.2008.0403

11      Raue F, Frank-Raue K, Grauer A. Multiple endocrine neoplasia type 2: Clinical features and screening. Endocrinol Metab Clin North Am. 1994 DOI: 10.1016/s0889-8529(18)30121-x

12      Wells SA, Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, et al. Revised American thyroid association guidelines for the management of medullary thyroid carcinoma. Thyroid. 2015;25(6):567–610.

13      Mathiesen JS, Effraimidis G, Rossing M, Rasmussen ÅK, Hoejberg L, Bastholt L, et al. Multiple endocrine neoplasia type 2: A reveiw. Semin Cancer Biol. 2021;(April). DOI: 10.1016/j.semcancer.2021.03.035

14      Eng C, Clayton D, Schuffenecker I, Lenoir G, Cote G, Gagel RF, et al. The relationship between specific ret proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2: International RET mutation consortium analysis. J Am Med Assoc. 1996 DOI: 10.1001/jama.276.19.1575

15      Mathiesen JS, Kroustrup JP, Vestergaard P, Stochholm K, Poulsen PL, Rasmussen ÅK, et al. Incidence and prevalence of multiple endocrine neoplasia 2a in denmark 1901–2014: A nationwide study. Clin Epidemiol. 2018;10:1479–87.

16      Qi XP, Zhao JQ, Fang XD, Lian BJ, Li F, Wang HH, et al. Spectrum of Germline RET variants identified by targeted sequencing and associated Multiple Endocrine Neoplasia type 2 susceptibility in China. BMC Cancer. 2021 DOI: 10.1186/s12885-021-08116-9

17      Qi XP, Peng JZ, Yang XW, Cao ZL, Yu XH, Fang XD, et al. The RET C611Y mutation causes MEN 2A and associated cutaneous lichen amyloidosis. Endocr Connect. 2018 DOI: 10.1530/EC-18-0220

18      Nagy N, Guyer RA, Hotta R, Zhang D, Newgreen DF, Halasy V, et al. RET overactivation leads to concurrent Hirschsprung disease and intestinal ganglioneuromas. Dev. 2020 DOI: 10.1242/dev.190900

19      Grieco M, Santoro M, Berlingieri MT, Melillo RM, Donghi R, Bongarzone I, et al. PTC is a novel rearranged form of the ret proto-oncogene and is frequently detected in vivo in human thyroid papillary carcinomas. Cell. 1990 DOI: 10.1016/0092-8674(90)90659-3

20      Goodman KM, Kjær S, Beuron F, Knowles PP, Nawrotek A, Burns EM, et al. RET recognition of GDNF-GFRα1 ligand by a composite binding site promotes membrane-proximal self-association. Cell Rep. 2014 DOI: 10.1016/j.celrep.2014.08.040

21      Romei C, Ciampi R, Elisei R. A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat Rev Endocrinol. 2016 DOI: 10.1038/nrendo.2016.11

22      Cranston AN, Carniti C, Oakhill K, Radzio-Andzelm E, Stone EA, McCallion AS, et al. RET is constitutively activated by novel tandem mutations that alter the active site resulting in multiple endocrine neoplasia type 2B. Cancer Res. 2006 DOI: 10.1158/0008-5472.CAN-06-0884

23      Mathiesen JS, Effraimidis G, Rossing M, Rasmussen ÅK, Hoejberg L, Bastholt L, et al. Multiple endocrine neoplasia type 2: A reveiw. Semin Cancer Biol. 2021 DOI: 10.1016/j.semcancer.2021.03.035

24      Takahashi M, Ritz J, Cooper GM. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell. 1985 DOI: 10.1016/0092-8674(85)90115-1

25      Takeuchi K, Soda M, Togashi Y, Suzuki R, Sakata S, Hatano S, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med. 2012 DOI: 10.1038/nm.2658

26      Bergholm U, Bergström R, Ekbom A. Long term follow-up of patients with medullary carcinoma of the thyroid. Cancer. 1997 DOI: 10.1002/(SICI)1097-0142(19970101)79:1<132::AID-CNCR19>3.0.CO;2-5

27      Iihara M, Yamashita T, Okamoto T, Kanbe M, Yamazaki K, Egawa S, et al. A nationwide clinical survey of patients with multiple endocrine neoplasia type 2 and familial medullary thyroid carcinoma in Japan. Jpn J Clin Oncol. 1997 DOI: 10.1093/jjco/27.3.128

28      van den Broek MFM, van Santen HM, Valk GD, Verrijn Stuart AA. Children with multiple endocrine neoplasia type 2B: Not tall and marfanoid, but short with normal body proportions. Clin Endocrinol (Oxf). 2021 DOI: 10.1111/cen.14536

29      Wray CJ, Rich TA, Waguespack SG, Lee JE, Perrier ND, Evans DB. Failure to recognize multiple endocrine neoplasia 2B: More common than we think? Ann Surg Oncol. 2008 DOI: 10.1245/s10434-007-9665-4

30      Castinetti F, Waguespack SG, Machens A, Uchino S, Hasse-Lazar K, Sanso G, et al. Natural history, treatment, and long-term follow up of patients with multiple endocrine neoplasia type 2B: an international, multicentre, retrospective study. Lancet Diabetes Endocrinol. 2019 DOI: 10.1016/S2213-8587(18)30336-X

31      van den Broek MFM, Rijks EBG, Nikkels PGJ, Wolters VM, van Es RJJ, van Santen HM, et al. Timely diagnosis of multiple endocrine neoplasia 2B by identification of intestinal ganglioneuromatosis: a case series. Endocrine. 2021 DOI: 10.1007/s12020-021-02607-2

32      Block MA, Jackson CE, Greenawald KA, Yott JB, Tashjian AH. Clinical Characteristics Distinguishing Hereditary From Sporadic Medullary Thyroid Carcinoma: Treatment Implications. Arch Surg. 1980 DOI: 10.1001/archsurg.1980.01380020012004

33      Wolfe HJ, Melvin KEW, Cervi-Skinner SJ, AL Saadi AA, Juliar JF, Jackson CE, et al. C-Cell Hyperplasia Preceding Medullary Thyroid Carcinoma. N Engl J Med. 1973 DOI: 10.1056/nejm197308302890901

34      Mathiesen JS, Kroustrup JP, Vestergaard P, Stochholm K, Poulsen PL, Rasmussen ÅK, et al. Survival and Long-Term Biochemical Cure in Medullary Thyroid Carcinoma in Denmark 1997-2014: A Nationwide Study. Thyroid. 2019 DOI: 10.1089/thy.2018.0564

35      Brauckhoff M, Machens A, Lorenz K, Bjøro T, Varhaug JE, Dralle H. Surgical curability of medullary thyroid cancer in multiple endocrine neoplasia 2b: A changing perspective. Ann Surg. 2014 DOI: 10.1097/SLA.0b013e3182a6f43a

36      Pacak K, Eisenhofer G, Ilias I. Diagnosis of pheochromocytoma with special emphasis on MEN2 syndrome. Hormones. 2009 DOI: 10.14310/horm.2002.1227

37      Iacobone M, Carnaille B, Palazzo FF, Vriens M. Hereditary hyperparathyroidism—a consensus report of the European Society of Endocrine Surgeons (ESES). Langenbeck’s Arch Surg. 2015 DOI: 10.1007/s00423-015-1342-7

38      Larsen LV, Mirebeau-Prunier D, Imai T, Alvarez-Escola C, Hasse-Lazar K, Censi S, et al. Primary hyperparathyroidism as first manifestation in multiple endocrine neoplasia type 2A: An international multicenter study. Endocr Connect. 2020 DOI: 10.1530/EC-20-0163

39      Scapineli JO, Ceolin L, Puñales MK, Dora JM, Maia AL. MEN 2A-related cutaneous lichen amyloidosis: report of three kindred and systematic literature review of clinical, biochemical and molecular characteristics. Fam Cancer. 2016 DOI: 10.1007/s10689-016-9892-6

40      Taïeb D, Kebebew E, Castinetti F, Chen CC, Henry JF, Pacak K. Diagnosis and preoperative imaging of multiple endocrine neoplasia type 2: Current status and future directions. Clin Endocrinol (Oxf). 2014 DOI: 10.1111/cen.12513

41      Neumann HPH, Young WF, Eng C. Pheochromocytoma and Paraganglioma. N Engl J Med. 2019 DOI: 10.1056/nejmra1806651

42      Lenders JWM, Duh QY, Eisenhofer G, Gimenez-Roqueplo AP, Grebe SKG, Murad MH, et al. Pheochromocytoma and paraganglioma: An endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014 DOI: 10.1210/jc.2014-1498

43      Costante G, Meringolo D, Durante C, Bianchi D, Nocera M, Tumino S, et al. Predictive value of serum calcitonin levels for preoperative diagnosis of medullary thyroid carcinoma in a cohort of 5817 consecutive patients with thyroid nodules. J Clin Endocrinol Metab. 2007 DOI: 10.1210/jc.2006-1590

44      Costante G, Durante C, Francis Z, Schlumberger M, Filetti S. Determination of calcitonin levels in C-cell disease: Clinical interest and potential pitfalls. Nat Clin Pract Endocrinol Metab. 2009 DOI: 10.1038/ncpendmet1023

45      Eckelt F, Vogel M, Geserick M, Kirsten T, Bae YJ, Baber R, et al. Calcitonin measurement in pediatrics: Reference ranges are gender-dependent, validation in medullary thyroid cancer and effects of thyroid diseases. Clin Chem Lab Med. 2019 DOI: 10.1515/cclm-2018-1186

46      Castagna MG, Fugazzola L, Maino F, Covelli D, Memmo S, Sestini F, et al. Reference range of serum calcitonin in pediatric population. J Clin Endocrinol Metab. 2015 DOI: 10.1210/jc.2014-4508

47      Niederle MB, Scheuba C, Riss P, Selberherr A, Koperek O, Niederle B. Early Diagnosis of Medullary Thyroid Cancer: Are Calcitonin Stimulation Tests Still Indicated in the Era of Highly Sensitive Calcitonin Immunoassays? Thyroid. 2020 DOI: 10.1089/thy.2019.0785

48      Wilhelm SM, Wang TS, Ruan DT, Lee JA, Asa SL, Duh QY, et al. The American association of endocrine surgeons guidelines for definitive management of primary hyperparathyroidism. JAMA Surg. 2016 DOI: 10.1001/jamasurg.2016.2310

49      Machens A, Lorenz K, Dralle H. Peak incidence of pheochromocytoma and primary hyperparathyroidism in multiple endocrine neoplasia 2: Need for age-adjusted biochemical screening. J Clin Endocrinol Metab. 2013 DOI: 10.1210/jc.2012-3192

50      Fassnacht M, Dekkers OM, Else T, Baudin E, Berruti A, De Krijger RR, et al. European society of endocrinology clinical practice guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Network for the study of adrenal tumors. Eur J Endocrinol. 2018 DOI: 10.1530/EJE-18-0608

51      Amodru V, Taieb D, Guerin C, Romanet P, Paladino N, Brue T, et al. MEN2-related pheochromocytoma: current state of knowledge, specific characteristics in MEN2B, and perspectives. Endocrine. 2020 DOI: 10.1007/s12020-020-02332-2

52      Shah MH, Goldner WS, Benson AB, Bergsland E, Blaszkowsky LS, Brock P, et al. Neuroendocrine and Adrenal Tumors, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw. 2021 DOI: 10.6004/jnccn.2021.0032

53      Buitenwerf E, Osinga TE, Timmers HJLM, Lenders JWM, Feelders RA, Eekhoff EMW, et al. Efficacy of α-blockers on hemodynamic control during pheochromocytoma resection: A randomized controlled trial. J Clin Endocrinol Metab. 2020 DOI: 10.1210/clinem/dgz188

54      Wells SA, Pacini F, Robinson BG, Santoro M. Multiple endocrine neoplasia type 2 and familial medullary thyroid carcinoma: An update. J Clin Endocrinol Metab. 2013 DOI: 10.1210/jc.2013-1204

55      Dickson P V., Alex GC, Grubbs EG, Ayala-Ramirez M, Jimenez C, Evans DB, et al. Posterior retroperitoneoscopic adrenalectomy is a safe and effective alternative to transabdominal laparoscopic adrenalectomy for pheochromocytoma. Surgery. 2011 DOI: 10.1016/j.surg.2011.07.004

56      Vrielink OM, Wevers KP, Kist JW, Borel Rinkes IHM, Hemmer PHJ, Vriens MR, et al. Laparoscopic anterior versus endoscopic posterior approach for adrenalectomy: a shift to a new golden standard? Langenbeck’s Arch Surg. 2017 DOI: 10.1007/s00423-016-1533-x

57      Castinetti F, Qi XP, Walz MK, Maia AL, Sansó G, Peczkowska M, et al. Outcomes of adrenal-sparing surgery or total adrenalectomy in phaeochromocytoma associated with multiple endocrine neoplasia type 2: An international retrospective population-based study. Lancet Oncol. 2014 DOI: 10.1016/S1470-2045(14)70154-8

58      Moalem J, Guerrero M, Kebebew E. Bilateral neck exploration in primary hyperparathyroidism-when is it selected and how is it performed? World J Surg. 2009 DOI: 10.1007/s00268-009-9941-5

59      Machens A, Elwerr M, Lorenz K, Weber F, Dralle H. Long-term outcome of prophylactic thyroidectomy in children carrying RET germline mutations. Br J Surg. 2018 DOI: 10.1002/bjs.10746

60      Febrero B, Rodríguez JM, Ríos A, Segura P, Pérez-Sánchez B, Torregrosa N, et al. Prophylactic thyroidectomy in multiple endocrine neoplasia 2 (MEN2) patients with the C634Y mutation: A long-term follow-up in a large single-center cohort. Eur J Surg Oncol. 2019 DOI: 10.1016/j.ejso.2018.09.002

61      Elisei R, Romei C, Renzini G, Bottici V, Cosci B, Molinaro E, et al. The timing of total thyroidectomy in RET gene mutation carriers could be personalized and safely planned on the basis of serum calcitonin: 18 Years experience at one single center. J Clin Endocrinol Metab. 2012 DOI: 10.1210/jc.2011-2046

62      Machens A, Dralle H. Advances in risk-oriented surgery for multiple endocrine neoplasia type 2. Endocr Relat Cancer. 2018 DOI: 10.1530/ERC-17-0202

63      Kluijfhout WP, Van Beek DJ, Stuart AAV, Lodewijk L, Valk GD, Zee DCV Der, et al. Postoperative complications after prophylactic thyroidectomy for very young patients with multiple endocrine neoplasia type 2: Retrospective cohort analysis. Med (United States). 2015 DOI: 10.1097/MD.0000000000001108

64      Rohmer V, Vidal-Trecan G, Bourdelot A, Niccoli P, Murat A, Wemeau JL, et al. Prognostic factors of disease-free survival after thyroidectomy in 170 young patients with a RET germline mutation: A multicenter study of the Groupe Français d’Etude des Tumeurs Endocrines. J Clin Endocrinol Metab. 2011 DOI: 10.1210/jc.2010-1234

65      Opsahl EM, Brauckhoff M, Schlichting E, Helset K, Svartberg J, Brauckhoff K, et al. A Nationwide Study of Multiple Endocrine Neoplasia Type 2A in Norway: Predictive and Prognostic Factors for the Clinical Course of Medullary Thyroid Carcinoma. Thyroid. 2016. DOI: 10.1089/thy.2015.0673

66      Machens A, Dralle H. Biomarker-based risk stratification for previously untreated medullary thyroid cancer. J Clin Endocrinol Metab. 2010 DOI: 10.1210/jc.2009-2368

67      Moley JF, DeBenedetti MK. Patterns of nodal metastases in palpable medullary thyroid carcinoma: Recommendations for extent of node dissection. Annals of Surgery. 1999. DOI: 10.1097/00000658-199906000-00016

68      Weber T, Schilling T, Frank-Raue K, Colombo-Benkmann M, Hinz U, Ziegler R, et al. Impact of modified radical neck dissection on biochemical cure in medullary thyroid carcinomas. Surgery. 2001 DOI: 10.1067/msy.2001.118380a

69      Machens A, Schneyer U, Holzhausen HJ, Dralle H. Prospects of remission in medullary thyroid carcinoma according to basal calcitonin level. J Clin Endocrinol Metab. 2005 DOI: 10.1210/jc.2004-1836

70      Machens A, Hofmann C, Hauptman S, Dralle H. Locoregional recurrence and death from medullary thyroid carcinoma in a contemporaneous series: 5-years results. Eur J Endocrinol. 2007 DOI: 10.1530/EJE-07-0095

71      Spanheimer PM, Ganly I, Chou JF, Capanu M, Nigam A, Ghossein RA, et al. Prophylactic Lateral Neck Dissection for Medullary Thyroid Carcinoma is not Associated with Improved Survival. Ann Surg Oncol. 2021 DOI: 10.1245/s10434-021-09683-8

72      Roman BR, Randolph GW, Kamani D. Conventional Thyroidectomy in the Treatment of Primary Thyroid Cancer. Endocrinol Metab Clin North Am. 2019 DOI: 10.1016/j.ecl.2018.11.003

73      Bliss RD, Gauger PG, Delbridge LW. Surgeon’s approach to the thyroid gland: Surgical anatomy and the importance of technique. World J Surg. 2000 DOI: 10.1007/s002680010173

74      MacHens A, Dralle H. Prognostic impact of N staging in 715 medullary thyroid cancer patients: Proposal for a revised staging system. Ann Surg. 2013 DOI: 10.1097/SLA.0b013e318268301d

75      Engelbach M, Görges R, Forst T, Pfützner A, Dawood R, Heerdt S, et al. Improved diagnostic methods in the follow-up of medullary thyroid carcinoma by highly specific calcitonin measurements. J Clin Endocrinol Metab. 2000 DOI: 10.1210/jc.85.5.1890

76      Pellegriti G, Leboulleux S, Baudin E, Bellon N, Scollo C, Travagli JP, et al. Long-term outcome of medullary thyroid carcinoma in patients with normal postoperative medical imaging. Br J Cancer. 2003 DOI: 10.1038/sj.bjc.6600930

77      Giovanella L, Treglia G, Iakovou I, Mihailovic J, Verburg FA, Luster M. EANM practice guideline for PET/CT imaging in medullary thyroid carcinoma. Eur J Nucl Med Mol Imaging. 2020 DOI: 10.1007/s00259-019-04458-6

78      July M, Santhanam P, Giovanella L, Treglia G. Role of positron emission tomography imaging in Multiple Endocrine Neoplasia syndromes. Clin Physiol Funct Imaging. 2018 DOI: 10.1111/cpf.12391

79      Giraudet AL, Al Ghulzan A, Aupérin A, Leboulleux S, Chehboun A, Troalen F, et al. Progression of medullary thyroid carcinoma: Assessment with calcitonin and carcinoembryonic antigen doubling times. Eur J Endocrinol. 2008 DOI: 10.1530/EJE-07-0667

80      Meijer JAA, Le Cessie S, Van Den Hout WB, Kievit J, Schoones JW, Romijn JA, et al. Calcitonin and carcinoembryonic antigen doubling times as prognostic factors in medullary thyroid carcinoma: A structured meta-analysis. Clin Endocrinol (Oxf). 2010 DOI: 10.1111/j.1365-2265.2009.03666.x

81      Barbet J, Campion L, Kraeber-Bodéré F, Chatal JF. Prognostic impact of serum calcitonin and carcinoembryonic antigen doubling-times in patients with medullary thyroid carcinoma. J Clin Endocrinol Metab. 2005 DOI: 10.1210/jc.2005-0044

82      Martinez SR, Beal SH, Chen A, Chen SL, Schneider PD. Adjuvant external beam radiation for medullary thyroid carcinoma. J Surg Oncol. 2010 DOI: 10.1002/jso.21557

83      Jin M, Megwalu UC, Noel JE. External Beam Radiotherapy for Medullary Thyroid Cancer Following Total or Near-Total Thyroidectomy. Otolaryngol - Head Neck Surg (United States). 2021 DOI: 10.1177/0194599820947696

84      Hadoux J, Schlumberger M. Chemotherapy and tyrosine-kinase inhibitors for medullary thyroid cancer. Best Pract Res Clin Endocrinol Metab. 2017 DOI: 10.1016/j.beem.2017.04.009

85      Vitale G, Fonderico F, Martignetti A, Caraglia M, Ciccarelli A, Nuzzo V, et al. Pamidronate improves the quality of life and induces clinical remission of bone metastases in patients with thyroid cancer. Br J Cancer. 2001 DOI: 10.1054/bjoc.2001.1832

86      Meijer JAA, Bakker LEH, Valk GD, De Herder WW, De Wilt JHW, Netea-Maier RT, et al. Radioactive iodine in the treatment of medullary thyroid carcinoma: A controlled multicenter study. Eur J Endocrinol. 2013 DOI: 10.1530/EJE-12-0943

87      Wells SA, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: A randomized, double-blind phase III trial. J Clin Oncol. 2012 DOI: 10.1200/JCO.2011.35.5040

88      Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, et al. Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol. 2013 DOI: 10.1200/JCO.2012.48.4659

89      Ceolin L, Da Silveira Duval MA, Benini AF, Ferreira CV, Maia AL. Medullary thyroid carcinoma beyond surgery: Advances, challenges, and perspectives. Endocr Relat Cancer. 2019 DOI: 10.1530/ERC-18-0574

90      Kraft IL, Akshintala S, Zhu Y, Lei H, Derse-Anthony C, Dombi E, et al. Outcomes of children and adolescents with advanced hereditary medullary thyroid carcinoma treated with vandetanib. Clin Cancer Res. 2018 DOI: 10.1158/1078-0432.CCR-17-2101

91      Sherman SI, Clary DO, Elisei R, Schlumberger MJ, Cohen EEW, Schöffski P, et al. Correlative analyses of RET and RAS mutations in a phase 3 trial of cabozantinib in patients with progressive, metastatic medullary thyroid cancer. Cancer. 2016 DOI: 10.1002/cncr.30252

92      Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, et al.  Efficacy of Selpercatinib in RET -Altered Thyroid Cancers . N Engl J Med. 2020 DOI: 10.1056/nejmoa2005651

93      Subbiah V, Hu MI, Wirth LJ, Schuler M, Mansfield AS, Curigliano G, et al. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 2021 DOI: 10.1016/S2213-8587(21)00120-0

94      Young WF. Metastatic pheochromocytoma: In search of a cure. Endocrinol (United States). 2020 DOI: 10.1210/endocr/bqz019

95      Thosani S, Ayala-Ramirez M, Palmer L, Hu MI, Rich T, Gagel RF, et al. The characterization of pheochromocytoma and its impact on overall survival in multiple endocrine neoplasia type 2. J Clin Endocrinol Metab. 2013 DOI: 10.1210/jc.2013-1653

96      Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric Thyroid Carcinoma: Incidence and Outcomes in 1753 Patients. J Surg Res. 2009 DOI: 10.1016/j.jss.2009.03.098

97      Modigliani E, Cohen R, Campos JM, Conte-Devolx B, Maes B, Boneu A, et al. Prognostic factors for survival and for biochemical cure in medullary thyroid carcinoma: results in 899 patients. The GETC Study Group. Groupe d’étude des tumeurs à calcitonine. Clin Endocrinol (Oxf). 1998

98      Shepet K, Alhefdhi A, Lai N, Mazeh H, Sippel R, Chen H. Hereditary medullary thyroid cancer: Age-appropriate thyroidectomy improves disease-free survival. Ann Surg Oncol. 2013 DOI: 10.1245/s10434-012-2757-9

99      Sandoval JA, Fernandez-Pineda I, Malkan AD. Risk-reduction surgery in pediatric surgical oncology: A perspective. J Pediatr Surg. 2016 DOI: 10.1016/j.jpedsurg.2016.01.004

Multiple Endocrine Neoplasia Type 1

ABSTRACT

Multiple Endocrine Neoplasia Type 1 (MEN1) is a rare autosomal dominantly inherited endocrine tumor predisposition syndrome, caused by mutations in the MEN1 gene. Cardinal manifestations are primary hyperparathyroidism (pHPT), pituitary adenomas (PA), and neuroendocrine tumors (NETs) of the pancreas (PanNET) and duodenum. Other manifestations are NETs of thymus, lung, and stomach, adrenal tumors, and an increased breast cancer risk in women. Malignant NETs are the most important cause of disease-related mortality, mainly NF-PanNETs, gastrinomas and thymus NETs. MEN1 can be diagnosed genetically and a clinical diagnosis in patients with negative comprehensive testing has been debated. Timely recognition of MEN1, referral for genetic testing and swift cascade screening is essential. MEN1-related pHPT (penetrance >95%) is a multiglandular disease and recurrence after initial operation is to be expected. Subtotal parathyroidectomy is the preferred initial operation. Prolactinomas are the most prevalent PA in MEN1, followed by non-functioning (NF) PAs. Treatment and treatment results do not differ from sporadic PAs. Life-time penetrance of duodenopancreatic NETs is >80%. NF-PanNETs are most frequent, followed by gastrinomas and insulinomas. Surgical resection is the mainstay of treatment, and is indicated in non-gastrinoma functional PanNETs and NF-PanNETs >2cm or with progression during follow-up. No consensus exists on the surgical treatment of MEN1-related gastrinoma. MEN1-related dpNETs are currently detected at earlier stages and more indolent small dpNETs are seen. The main challenge is to identify patients at risk for an aggressive disease course. Thymic NETs (2-8%) occur predominantly in males and have a poor prognosis. Bronchopulmonary NETs are more frequent than previously thought, occur in both sexes, and are usually indolent although cases with a deviant progressive course occur. Adrenal tumors are mostly indolent non-functioning adenomas, but adrenocortical carcinomas and pheochromocytomas do occur. Women with MEN1 have an increased (RR 2.8) risk of breast cancer, at a younger age than the general population. Given the complexity of the disease, it is strongly advised that patients, whenever possible, be followed and treated in centers of expertise.

 

 

INTRODUCTION

 

Multiple Endocrine Neoplasia Type 1 (MEN1) is an inherited endocrine tumor predisposition syndrome. The prevalence is estimated at 1 in 20.000 to 1 in 40.000, and is therefore considered a rare disease (1). The syndrome predisposes mutation carriers to develop several endocrine tumors (Figure 1) with a high lifetime incidence of primary hyperparathyroidism (pHPT), pituitary adenomas (PA), and neuroendocrine tumors (NETs) of the pancreas (PanNET) and duodenum (dNET) (2). These tumors are considered the cardinal manifestations of the syndrome. Besides the cardinal manifestations, patients with MEN1 are at a higher risk for developing NETs of the thymus, lung, and stomach. In addition, there is a higher risk for adrenal tumors and a higher risk for developing breast cancer in women (3, 4). Next to these endocrine manifestations, patients are at risk for developing several non-endocrine lesions of the skin and subcutaneous tumors such as lipomas.

 

Figure1. Manifestations of MEN1

After identification of the causative MEN1 mutation an intensive lifelong surveillance program follows, if possible, starting at childhood because of the high life-time risk for developing tumors (2). This intensive follow-up is aimed at early detection of tumors to enable timely interventions in order to prevent complications and metastases of tumors and thereby preventing premature death and improving the quality of the life of patients. In 2001 the first set of clinical practice guidelines was published by Brandi et al (5). Because of paucity of scientific evidence, these guidelines were mainly based on expert opinion. Although more evidence was available, the updated clinical guidelines of Thakker, et al. which were published in 2012 were also written in the absence of robust scientific evidence (2). However, publishing the clinical practice guidelines led to more structured care of patients, which facilitated studies of the natural course of the disease and the effect of follow-up and treatment strategies. Therefore, in the last two decades large and sometimes nation-wide MEN1 cohort studies were initiated which led to new insights into the course of the disease and knowledge about more optimal follow-up and treatment. However, to date there remains a paucity of prospective data from interventional trials designed specifically for MEN1 patients.

 

GENETICS

 

The MEN1 gene (OMIM 613733 (gene) and OMIM 131100 (phenotype)), identified in 1997  (6, 7), consists of 10 exons and is localized on chromosome 11q13. Exon 1, the 5’ region upstream of exon 2 and the 3’ region of exon 10 are non-coding, so there are 9 coding exons (exons 2 through 10). The MEN1 gen encodes the protein menin, a 610 amino-acid nuclear scaffold protein that regulates gene transcription by coordinating chromatin remodeling.

 

In 2008, Lemos and Thakker published an overview of the 459 different germline mutations reported in the first decade since the discovery of the gene (8). In 2016, Concolino, et al. identified an additional 208 novel germline variants, of which 76 were reported as Variant of Unknown Significance (VUS) (9). Around 40% of all identified mutations are frameshift mutations, 25% nonsense, 20% missense, 10% splice-site, and the remainder 5% are rarer mutations such as in-frame deletions/insertions and partial or whole gene deletions (10). Frameshift, nonsense, and splice-site mutations, which are the majority, are predicted to be loss of function mutations that lead to truncated forms of menin (8). The variants are scattered throughout the MEN1 gene with no evident hotspots, although some mutations are found in apparently unrelated families. This is considered to be a founder effect (11). Sequence analysis of the MEN1 gene will reveal most of the variants. Since 1-2% of the variants are (partial) deletions of the MEN1 gene (8), multiplex ligation-dependent probe amplification (MLPA) or Copy Number Variation (CNV) analysis should also be included in the diagnostic DNA testing.

 

When a sequence variant is identified in the MEN1 gene this is classified by the genetic laboratory as either benign or likely benign (which is considered a “negative” result i.e., no disease-causing variant is found, clinically similar to no sequence variant identified), as a VUS (uncertain if the variant is disease causing) or as pathogenic or likely pathogenic (which is considered a “positive” result, a disease-causing variant is found). Especially new non-truncating (mostly missense) mutations may be difficult to classify (12).

 

The approximate prevalence of MEN1 has been reported as 1 in 30,000 individuals with no apparent gender bias. MEN1 follows an autosomal dominant pattern of inheritance with >95% penetrance by age 40–50 years (11). The cardinal clinical manifestations of MEN1 are primary hyperparathyroidism (pHPT), anterior pituitary adenomas (PA), and NET of the duodenum and pancreas, the so called three P’s.

 

If a patient is diagnosed with MEN1, he or she should be advised to undergo screening to detect manifestations and remain under lifelong surveillance in a center of expertise where care is provided by multidisciplinary teams (MDTs) comprising relevant specialists with experience in the diagnosis and treatment of patients with endocrine tumors (2).

 

Single center cohorts have identified certain genotypes that are associated with a more aggressive course of the disease (especially related to duodenopancreatic NETs), such as mutation in the JUND (13) or the CHES1 (14)interacting domain or nonsense/frameshift versus missense mutations (15), but since none of these associations have been independently validated, genotype cannot be used to individualize surveillance.

 

Evaluation of 10 Dutch families suggest genetic anticipation (decreased age of disease onset or an increased disease severity in successive generations), a known phenomenon which to date cannot be explained in autosomal dominant inherited disease genes without trinucleotide repeat expansions (“growing genes”) (16). Somatic mosaicism with subsequent germline inheritance has been described (17).

 

Function of the MEN1 Gene

 

MEN1 is considered to act as a tumor suppressor gene which is demonstrated by the identification of inactivating mutations, together with loss of heterozygosity (LOH) in MEN1-related tumors. Biochemical, proteomics, genetics and genomics approaches have identified various potential roles, which converge on the regulation of gene expression. The most consistent findings show that menin connects transcription factors including JUND (OMIM 165162), NFKB (OMIM 164011), and SMAD3 (OMIM 603109) and modulates their activities. In the nucleus, menin acts as a scaffold protein to regulate gene transcription by coordinating chromatin remodeling interacting with chromatin regulatory proteins in the MLL1 /MLL2 complex. Menin is implicated in both histone deacetylase and histone methyltransferase activity (HMT), and via the latter it regulates the expression of cyclin-dependent kinase inhibitor (CDKI) and homeobox domain genes (18, 19). While the MEN1 gene functions as a tumor suppressor gene in MEN1, it has an oncogenic role in sporadic breast cancer cells (18). Some excellent reviews on the function of the MEN1 gene can be found elsewhere (11, 18, 20).

 

Potential Therapeutic Opportunities (18)

 

Loss of menin in MEN1-associated tissues, leads to the disruption of anti-proliferative gene expression programs and to the development of endocrine tumors. Restoration of the epigenetic perturbations or correction of the function of aberrantly expressed genes in the absence of menin hold promise for molecular mechanism-based means to treat or prevent MEN1-related tumors. The fate and function of a cell are determined by its gene expression signature. As menin is a transcriptional regulator, MEN1-related tumorigenesis is likely to be the result of aberrant tumor suppressive gene expression due to the loss of menin. Restoration of the expression of menin target genes in MEN1-affected tissues could therefore have therapeutic consequences. This has been shown in a preclinical study in mice, where MEN1 replacement in pituitary tumors of Men1 (+/-) mice led to a decrease in proliferation of the pituitary tumors (21).

 

DIAGNOSIS AND GENETIC TESTING

 

Diagnosis of MEN1

 

Patients with MEN1 are at risk to develop different endocrine and non-endocrine manifestations. The most important of which are (with approximate lifetime prevalence in parentheses):

  • Cardinal manifestations/Major criteria: pHPT (>95%), duodenopancreatic NETs (dpNETs) (>80%) and anterior pituitary tumors (50–65%)
  • Minor clinical criteria: adrenal adenomas (11–35%) and bronchopulmonary, thymic, and gastric NETs (20–30%) (22, 23).

 

Additionally, there might be an association with meningioma (<10%). Cutaneous manifestations such as subcutaneous lipomas (but visceral, pleural, or retroperitoneal lipomas have also been described), facial angiofibromas (22-88%) and collagenomas (0-72%) are also seen (2). Women with MEN1 have a 2-3-fold elevated risk of developing breast cancer (3, 4).

Presently, MEN1 can be diagnosed genetically by identifying the germline heterozygous (likely) pathogenic variant in the MEN1 gene through DNA analysis. According to the guidelines, a diagnosis of MEN1 can also be made on familial grounds in a patient with one of the cardinal MEN1 manifestations and a first-degree family member with MEN1 (2). Additionally, a clinical diagnosis can be made in individuals with two of the three cardinal manifestations (2). However, with modern-day sensitive DNA testing, the value of the clinical criterion in patients with negative DNA testing is under debate. There is mounting evidence that patients who have clinical MEN1, but negative DNA testing have a different clinical course from patients with positive DNA testing (24, 25). The same could also be argued for patients with a familial diagnosis with negative DNA testing for the family mutation, as these may have a sporadically occurring endocrine tumor. This subject is discussed in more detail in the paragraph on genetic heterogeneity.

 

Patients with MEN1 suffer from high morbidity and a decreased life expectancy. In the present day and age, MEN1-related malignancy is the main MEN1-related cause of death, particularly due to duodenopancreatic and thymic NETs (26, 27). A timely and accurate diagnosis of MEN1 is paramount to improve disease outcomes. This enables early identification of tumor manifestations allowing timely treatment to reduce morbidity and improve survival (28).

 

It is therefore important for clinicians to consider the diagnosis of MEN1 not only in those patients meeting clinical or familial criteria, but also in patients with a suspicious family or personal medical history, but not meeting clinical or familial diagnostic criteria. In patients presenting with an endocrine tumor within the MEN1 spectrum, taking a family history of MEN1-related tumors is very important. Additionally, a young age at presentation or multifocality of tumors within a single organ may point to a diagnosis of MEN1. The combination of a major and minor criterion or two minor criteria should also raise suspicion of MEN1. In all these cases of suspected MEN1, the patient should be referred to a clinical geneticist or genetic counselor for counseling and consideration of DNA testing. For patients presenting with sporadically occurring endocrine tumors, de Laat, et al. developed and validated a prediction rule to predict the presence of an MEN1 mutation (29). In this model, recurrent pHPT, non-recurrent pHPT, dpNETs, PA, NET of the stomach, lung and thymus, a positive family history for a NET and age, predicted the risk of having MEN1. The authors developed a nonogram for clinical practice, allowing the clinician to calculate the risk of MEN1 in patients suspected of MEN1 with sporadically occurring endocrine tumors (29).

 

DNA Testing

 

According to current practice guidelines diagnostic DNA testing for MEN1 should be offered to (2):

  • all patients fulfilling the diagnostic criteria of a clinical or familial MEN1 diagnosis
  • all patients with a pHPT under the age of 30 or multiple (synchronous) parathyroid adenomas under the age of 40 or recurrent parathyroid adenomas
  • patients with a gastrin-producing NET (irrespective the age of presentation)
  • patients with multiple PanNETs (irrespective the age of presentation)
  • patients with two different minor criteria
  • a patient with an MEN1-related tumor with a positive family history of MEN1-related tumors

 

Once MEN1 is diagnosed in the proband, genetic counseling and DNA testing should be offered to family members, preferably by means of cascade screening. Data from the DutchMEN Study Group (DMSG) emphasize the importance of timely genetic testing of family members and prompt clinical screening according to MEN1 guidelines. In a study determining lag time between MEN1 diagnosis in index cases and their non-index family members, they found a median lag time of 3.5 years (range 0-30) years, in which clinically significant manifestations occurred in the non-index family members with MEN1 (30). Genetic testing in asymptomatic family members of MEN1 patients is called pre-symptomatic or predictive genetic testing and involves testing the at-risk family members for the familial MEN1 mutation. This is single-site testing, and outcome is whether the family mutation is present or absent in this particular family member.

 

Genetic Heterogeneity

 

Some studies have reported that between 5% and 10% of patient who fulfill the clinical criteria for MEN1 will not harbor mutations in the coding region or adjacent splice sites. In some of these patients a (likely) pathogenic variant can be found in the CDKN1A (OMIM 600778 ), CDKN1B (OMIM 116899 ), CDKN2B (OMIM 600431 ) or CDKN2Cgene (OMIM 603369 ). Mutations in CDKN1B are the cause of the MEN4 syndrome, the latest of the MEN syndromes and most rare, with <50 cases reported in the literature to date (31-33). Rather than being a separate phenotype, (likely) pathogenic variants in these genes are more likely to cause a MEN1 phenotype, with pHPT, PA and gastroenteropancreatic NETs as the main features, and are best met with the same guidelines for surveillance, until more is known about the phenotype of this rare syndrome.

 

Recent data have shown that patients with a clinical diagnosis of MEN1, in whom no (likely) pathogenic variant in the MEN1 gene can be found (genotype-negative MEN1/GN-MEN1), and who do not have another known germline mutation, have a different phenotype and clinical course compared to mutation positive patients (24, 25). Genotype-negative patients develop MEN1 manifestations at higher age, rarely develop a third main MEN1 manifestation, and have a life expectancy comparable with the general population (24, 25).

 

Additionally, regarding the individual manifestations, it seems that GN-MEN1 patients have less recurrent or multigland pHPT, less multifocal PanNETs, and more somatotrophinomas and less prolactinomas compared to genotype-positive patients (25). Most patients with GN-MEN1 present with the combination of pHPT and PA, followed by pHPT/dpNET and dpNET/PA (25). The apparent differences in clinical course suggest that GN-MEN1 patients do not have true MEN1, but another MEN1-like syndrome or sporadic co-incidence of two NETs (2, 33). In these patients there is usually a negative family history for MEN1-related disease. Although not specified in the current guidelines, these patients may benefit from a separate classification with alternative surveillance recommendations based on the clinical picture, as has been suggested by Pieterman, et al. (25). Important baseline considerations for an alternative surveillance are genetic counseling, comprehensive genetic testing based on the personal and family history, and baseline screening to identify any unrecognized manifestations. In these patients, there is generally no cause for surveillance of the first-degree relatives, although these decisions should be individualized and discussed in multidisciplinary teams.

 

GN-MEN1 patients with a positive family history of clinical MEN1 or a foregut NET and those presenting with all three main MEN1-related tumors, should be followed according to MEN1 guidelines as should their relatives. In these patients, a “false-negative result” of DNA testing should also be considered. This may either be because deletion/duplication analyses are not performed, a sequence variant exists outside of the assayed region, or polymerase chain reaction primer selection led to selective amplification of wild-type DNA (25). Additionally, somatic mosaicism or alternative mechanisms of MEN1 gene silencing could lead to inactivation of normal menin (25).

 

Depending on the presenting clinical picture and the family history, other hereditary syndromes causing endocrine tumors should also be considered.

 

If pHPT is the primary phenotype, other genes associated with hereditary pHPT are for example CDC73, CASR and RET (MEN2). Germline CDC73 (formerly HRPT2) (OMIM 607393) analysis is recommended in individuals with (suspected) Hyperparathyroidism-Jaw Tumor (HPT-JT) syndrome, familial isolated pHPT, atypical or malignant parathyroid histology, and young individuals with pHPT. These criteria would increase germline CDC73 mutation detection, enabling optimal clinical management of pHPT as well as genetic counseling and surveillance for family members at risk for developing CDC73-related disorders(34).

 

If PAs are the primary phenotype, mutations in the AIP gene (OMIM 605555) should be considered as these can cause (familial) pituitary adenomas. Predictors of a genetic cause of sporadic pituitary adenomas are young age of diagnosis, and also in AIP pathogenic variants there is an association with gigantism and macroadenomas (35).

 

PanNETs can also be seen in neurofibromatosis type 1 (NF1), Von Hippel-Lindau (VHL), and Tuberous Sclerosis Complex (TSC).

 

Pretest Counseling

 

Before DNA testing, pretest counseling is of utmost importance. The patient should be informed by genetic counseling about all aspects (medical, psychological, social, and familial implications) of the possible outcome of genetic testing. This should lead to an individual decision whether or not to opt for DNA testing. In case of DNA testing in minors the counseling should be offered to the parents, and include the minor if possible (which is obligatory over the age of 12 in the country where the authors practice (the Netherlands) to obtain informed consent for testing.

 

In case of diagnostic testing the patient must be informed about the possible outcomes of the DNA test (finding an (likely)pathogenic variant, finding a VUS, and not finding (likely) pathogenic variants) and the implications of these findings for the patient and family members.

 

In diagnostic DNA testing the MEN1 gene should be analyzed, for which Sanger sequencing can be used, or Next Generation Sequencing (NGS) techniques. To exclude deletions MLPA or CNV analysis must be performed. As mentioned earlier there is genetic heterogeneity and panel DNA diagnostics can be considered. In particular CDKN1A, CDKN1B, CDKN2B, CDKN2C, CDC73 and AIP can be added to the panel using NGS techniques, also to be completed with CNV analysis, depending on the clinical picture. In case of panel testing, the patient must be prepared for the possible findings in the different genes, differentiation of the consequences, and implications of these findings.

 

In case of testing for a familial (likely) pathogenic variant (so-called presymptomatic or predictive DNA testing) presence or absence of the familial mutation can be ascertained, but the differences in expression of the MEN1 syndrome, both within and between families must be emphasized.

 

In case of future pregnancy the possible options like invasive prenatal diagnostics and Preimplantation Genetic Diagnosis (PGD) should be discussed so the prospective parents can make an informed decision about the desired pregnancy.

 

Periodical Screening

 

The identification of an MEN1 mutation in patients and family members at risk is followed by the advice to remain under lifelong surveillance, with at least annual clinic visits including history, physical examination, biochemical screening, and radiological screening at specific intervals (2). This should preferably be carried out in centers of expertise with a dedicated multidisciplinary team well versed in management of patients with MEN1. In MEN1 there are no prophylactic treatments, so the goal of this screening & surveillance program is early detection of MEN1-related tumors to minimize morbidity by hormonal hypersecretion and to prevent malignant NETs by timely intervention. In the absence of known genotype-phenotype correlations and with a heterogeneous clinical course, even within families, the specific mutation or family history cannot solely guide the surveillance program. In the following sections screening and surveillance is discussed within each manifestation.

 

Screening at the Pediatric Age

 

The current clinical guidelines for MEN1 suggest starting clinical and biochemical screening at the age of 5 years (2), which is based on the earliest reported case of a patients with a clinical MEN1 manifestation (36). For radiologic screening in asymptomatic children, pituitary imaging is suggested from age 5 years onward (every 3 years), with abdominal imaging starting at age 10 years (every 1-3 years) and thoracic imaging starting at age 15 years (every 1-2 years) (2). However, this intensive surveillance at the pediatric age has been questioned by some groups who suggest postponing routine screening of asymptomatic patients until ages 15 or 16 years while counseling parents about typical clinical signs of MEN1 manifestations and contacting providers if they occur (37).

 

PRIMARY HYPERPARATHYROIDISM

 

Primary hyperparathyroidism (pHPT) (Figure 1) is one of the cardinal manifestations of MEN1 and has an almost complete lifetime penetrance (24, 38). It is often the first clinical manifestation of the disease and biochemical (asymptomatic) pHPT can be diagnosed several years before symptoms arise. The reported mean age of pHPT diagnosis in published MEN1 cohorts is in the fourth decade of life (39-45), with wide ranges. When interpreting these mean ages at diagnosis it is important to realize that these cohorts often span multiple decades, are made up of both index cases and family members, and contain patients who did and did not follow prospective screening programs. Recent studies reporting on MEN1 at the pediatric age show that in a screened population at least half of the pediatric patients already have primary hyperparathyroidism, although rarely seen before the age of 10 (46-49). In most cases, patients diagnosed at a pediatric age are asymptomatic and the diagnosis is made biochemically by screening (46-49). Clinical and symptomatic pHPT is usually seen in the third decade of life.

 

Primary hyperparathyroidism in MEN1 is a multiglandular disease, affecting all parathyroid glands, although often asymmetrically and asynchronously. Parathyroid tumors in adults with MEN1 usually represent mono- or oligoclonal proliferations that probably arise independently in each parathyroid gland (50). Tumorigenesis is initiated when the remaining normal allele of the MEN1 gene is lost (the second hit), and as this cumulative chance increases with age, normal parathyroid tissue is less often seen with increasing age (51). Supernumerary glands (that is, more than four parathyroid glands) are frequently seen in MEN1, with reported ranges between 12-30% (52). Parathyroid glands at ectopic locations are also not uncommon in MEN1, especially in the thymus.

 

The diagnosis of primary hyperparathyroidism can be made when there is hypercalcemia in combination with an elevated or inadequately normal parathyroid hormone (PTH). In patients with MEN1 who follow a prospective screening program from an early age, the diagnosis is often made while they are still asymptomatic. Classic objective symptoms of pHPT include polyuria and polydipsia, gastro-intestinal complaints (nausea, abdominal pain, constipation, pancreatitis), (symptomatic) urolithiasis, and decreased bone mineral density (BMD) which can lead to pathological fractures. Non-specific symptoms occurring in pHPT are fatigue, musculoskeletal complaints, neuropsychiatric symptoms such as anxiety, depression, concentration disturbance and sleep-disturbances, and a general decrease in quality of life.

 

The diagnosis of pHPT in patients with known MEN1 or from a known MEN1 kindred is straightforward. However, pHPT can also be the first clinical clue to an MEN1 diagnosis in a patient or family without prior MEN1 diagnosis or suspected history. The prevalence of pHPT in the general population can be up to 1% (53, 54) and among cohorts of patients with pHPT, depending on the characteristics, the incidence of MEN1 is 1-18% (2). Considering MEN1 in patients presenting with pHPT is extremely important, because the diagnosis alters the management and prognosis of pHPT, allows screening and surveillance for other endocrine tumors associated with MEN1, and allows for cascade screening within the family to identify MEN1 germline mutations carriers. Important clues to an MEN1 diagnosis in a patient presenting with pHPT are young age of onset, a family history of pHPT or other MEN1-related tumors, a personal history of other MEN1-related tumors, and multiglandular disease or persistent/recurrent pHPT (29). Recurrent pHPT is one of the strongest predictors for the presence of an MEN1 mutation (29). Compared to sporadic pHPT, patients with MEN1-related pHPT present at an earlier age, have an almost equal gender distribution compared to female predominance in sporadic pHPT, and present with lower levels of calcium and PTH (55, 56). Even though they have biochemically milder disease, BMD seems to be lower in patients with MEN1-related pHPT and renal involvement similar compared to patient with sporadic pHPT, which may reflect longer standing disease (55). MEN1-related pHPT is a multiglandular disease, as already stipulated, while sporadic pHPT is predominantly caused by single-gland adenomas (56, 57). This also affects recurrence rates which are much higher in MEN1-related pHPT (56, 57). The American Association of Endocrine Surgeons (AAES) guidelines advise genetic counseling for patients younger than 40 years with pHPT and multiglandular disease and to consider this for those with a family history or syndromic manifestations (57). The European guidelines slightly differ suggesting genetic testing for MEN1 in patients with pHPT before the age of 40, multiglandular disease, or persistent/recurrent pHPT (58).

 

When comparing, several studies show that patients with MEN1-related pHPT have lower BMD compared to patients with sporadic pHPT (55, 59, 60), although a Chinese study found no significant difference (61). In patients with MEN1-related pHPT, decreased BMD is frequently seen and already present at a young age (62-64). When measured the 1/3 distal radius seems most affected, so including this location in dual-energy X-ray absorptiometry (DEXA) should be considered in patients with MEN1 (62, 64). Parathyroidectomy improves BMD (59, 65), although in one small study improvement was less for patients with MEN1 compared to patients with sporadic pHPT (59). A factor contributing to the earlier and more severe bone involvement in MEN1-related pHPT may be the early-onset of the disease thereby also influencing peak bone formation. In addition, other MEN1-related diseases may also contribute to bone loss such as pituitary insufficiency caused by pituitary adenomas or their treatment, hypercortisolism (although infrequent in MEN1), and gastro-intestinal surgery (66).

 

Urolithiasis is also frequently seen and at a young age in patients MEN1-related pHPT (55, 62, 64). In addition, a recent study showed that patients with MEN1 age 20-59 had a higher prevalence of chronic kidney disease stage 3 compared to the general US population (67).

 

It is therefore important to perform Dual-energy X-ray absorptiometry (DEXA) to assess BMD as well as a renal ultrasound and 24-hour urine for calcium excretion to asses risk of urolithiasis in patients with MEN1 diagnosed with pHPT. And if initial observation is chosen, DEXA should be repeated every 2 years (68).

 

In patients with MEN1 and pHPT, the interplay with Zollinger-Ellison Syndrome (ZES; increased gastric acid section due to gastrinomas) is also relevant, as calcium can increase gastrin levels. In a study among 84 patients with MEN1-pHPT and ZES, successful parathyroidectomy resulted in biochemical cure of ZES without any resection of duodenal or pancreatic NETs in 20% of the patients (69). In a recent perspective paper Hackeng and colleagues propose a parathyroid-gut axis arguing that hypercalcemia may promote the gastrin-cell hyperplasia to neoplasia sequence through the calcium-sensing receptor (70). The reverse, a more severe form of pHPT among patients with MEN1-ZES has also been suggested, because in the aforementioned study of 84 patients with MEN1-pHTP and ZES, patients had a higher frequency of urolithiasis at presentation, higher serum PTH, and higher recurrences rates after initial subtotal parathyroidectomy compared to the literature (69).

 

Parathyroidectomy

 

The treatment of hyperparathyroidism in MEN1 is surgical. Intervention is aimed at achieving eucalcemia for as long as possible, while preventing permanent hypoparathyroidism and facilitating potential subsequent surgery.

 

The optimal timing of the initial operation is still a matter of debate, especially in (asymptomatic) children and young adults. The guidelines for the management of asymptomatic pHPT recommend surgical intervention in case of significant hypercalcemia (1 mg/dL or 0.25 mmol/L above the upper limit of normal), skeletal abnormalities (a T-score of < -2.5 at the Lumbar Spine, Total Hip, Femoral Neck or 1/3 Distal Radius or a vertebral fracture), risk of renal complications (creatinine clearance below 60 ml/min, 24-h urine calcium excretion of >400 mg/d (>10 mmol/L)), the presence of nephrolithiasis/nephrocalcinosis, or age below 50 (68). However, these guidelines are not intended for patients with MEN1 and most patients with MEN1 will meet the age-criterion regardless of other values. In patients with MEN1 surgery is indicated in case of symptoms, significant hypercalcemia, and renal or skeletal complications. In addition, concomitant gastrinoma may also provide an indication for surgical intervention of pHPT. For patients not meeting any of these criteria, there is no evidence to determine timing of surgery. Arguments have been made in favor of observation to avoid the risk of symptomatic hypoparathyroidism, multiple operations, and by allowing the disease to progress a little bit more, making the glands more easily identifiable upon intervention. However, on the other hand, data showing early bone and renal complications have made others suggest and prefer early intervention to prevent downstream disabilities (71).

 

For initial parathyroidectomy in patients with MEN1 there are theoretically four different strategies: focused parathyroidectomy (removing a single affected parathyroid gland), unilateral clearance (resection of all parathyroid tissue on one side, including unilateral cervical thymectomy), subtotal parathyroidectomy with concomitant cervical thymectomy, or total parathyroidectomy, cervical thymectomy and immediate auto-transplantation of parathyroid tissue (usually to the non-dominant forearm).

 

The initial operation recommended by most experts and guidelines is a bilateral cervical exploration, identifying all four parathyroid glands and performing a subtotal parathyroidectomy (leaving a vascularized remnant about 1.5-2 times the size of a normal gland) with concomitant cervical thymectomy (2, 57, 58, 71-73). The latter serves the dual purpose of removing any ectopic/supranumerary parathyroid glands and potentially decreases the risk of subsequent development of thymic NETs. This approach offers the best balance between persistence (persisting pHPT after operation or recurrence within 6 months after operation) and recurrence (recurrent pHPT 6 months or more after the operation preceded by a eucalcemic period) on the one hand and permanent (lasting >6 months after the operation) hypoparathyroidism on the other hand. Persistence is infrequent in subtotal (0-22%) and total parathyroidectomy (0-19%), but rates range from 0-53% in less than subtotal parathyroidectomy(58, 72). Recurrence rates are also significantly higher after less than subtotal parathyroidectomy (0-100%) compared to subtotal (0-65%) or total parathyroidectomy (0-56%) (58, 72) and occur earlier (74). Permanent hypoparathyroidism on the other hand is rarely seen after less than subtotal parathyroidectomy. When comparing subtotal with total parathyroidectomy, hypoparathyroidism is significantly more frequent after total parathyroidectomy (RR 1.61 (95%CI 1.12-2.31) (72).

 

Pre-operative imaging plays a limited role at initial parathyroidectomy in patients with MEN1, because the recommended initial operation always constitutes bilateral neck exploration( 58). In addition, data has shown that pre-operative imaging (consisting of neck ultrasound and sestamibi scan as first line and parathyroid computed tomography (CT) or magnetic resonance imaging (MRI) as second line) only identified 68% of the largest glands pre-operatively (75). Pre-operative imaging may have some use for identifying ectopic glands (7% of ectopic glands were identified by pre-operative imaging in one series) and for identifying concomitant thyroid abnormalities that need attention (71, 75). Similar, intra-operative PTH monitoring seems of little value during the initial parathyroidectomy (76).

 

Recently, several groups have advocated unilateral clearance as an initial operation, especially for young patients with MEN1 (74, 77-79). The rationale behind this approach is to provide several years of eucalcemia during acquisition of peak bone mass, while preventing hypoparathyroidism and allowing subsequent reoperations to be performed in a non-operated neck (the contra-lateral side). A prerequisite for this strategy is that pre-operative imaging concordantly shows unilateral disease. Intra-operative PTH monitoring should be used to ensure there is an adequate drop in PTH after the resection. Although persistence rates between 10-15% after unilateral clearance or single-gland excision have been reported by these groups (74, 77), others state that less than subtotal parathyroidectomy has an unacceptable failure rate (69% in one study) (80). Several remarks must be made when using retrospective studies to evaluate this strategy. The first being that intentional less than subtotal resection is a different entity from an intended subtotal or total resection in which not all glands were identified (78). Secondly, true unilateral clearance in which all parathyroid tissue on one side of the neck is removed including unilateral cervical thymectomy is a very different operative strategy from minimal invasive parathyroidectomy/single gland excision and in retrospective studies these are often lumped together under “less than subtotal” resections. Thirdly, the success of such an approach is dependent on the sensitivity of pre-operative imaging and in most retrospective studies, more sensitive imaging modalities such as 18F-fluorcholine positron emission tomography (PET)/CT have not been used. Finally, since in MEN1 inherently all parathyroids are affected, although asynchronously, such an approach may be more successful in younger patients, where there may still be normal parathyroid glands (51). Currently this approach is controversial. Therefore, prospective data are needed to determine if and when unilateral clearance can benefit patients with MEN1 at the time of their initial parathyroidectomy.

 

Currently, subtotal parathyroidectomy remains the initial procedure of choice, but total parathyroidectomy or unilateral clearance can be considered depending on individual circumstances. Single gland excision is generally not recommended.

 

After initial subtotal parathyroidectomy, the 10-year recurrence rate is approximately 50% (2). Reoperation is therefore a frequent necessity in patients with MEN1. Recurrence can be caused by parathyroid glands missed during the initial operation, parathyroid glands intentionally left in situ, growth of the remnant of a partially resected gland, supranumerary and/or ectopic glands, and hyperplasia of autotransplanted parathyroid tissue. As reoperations are more complex and have a higher risk of complications (12% not including hypoparathyroidism in one study of reoperative parathyroidectomy in MEN1 (81)), the timing of the reoperation is individualized and patients with mild biochemical recurrence are usually initially observed. When reoperation is indicated, careful examination of the operation notes and pathology reports of previous procedures, if available, is very important. In contrast to the initial surgery, pre-operative imaging is essential for surgical planning in reoperations. First-line imaging studies are neck ultrasound and Tc99m-sestamibi scan, although this may not show all enlarged glands. Second-line imaging studies are 4-dimentional CT or MRI and PET (18F-fluorocholine or 11C-methionine) (82, 83). In a small study 18F-fluorocholine PET-CT has also shown to be of added value in MEN1-related pHPT (84). If first- and second-line studies are inconclusive more invasive localization studies can be considered such as arteriography, venous sampling, and neck ultrasound with fine needle aspiration and PTH measurement (83). The exact operative strategy (bilateral or unilateral neck exploration or focused resection) is individualized based on previous operation(s) and results of preoperative imaging. If the thymus was not removed during the initial operation, its removal is recommended at reoperation (81). Intra-operative PTH monitoring is valuable for the reoperative setting in MEN1 as it can inform when the exploration can be ended (71, 81).

 

As a consequence of the extended initial operation necessary, as well as frequent reoperation, life-time risk of postoperative hypoparathyroidism is relatively high for patients with MEN1. Transient hypoparathyroidism, defined as lasting less than six months after parathyroidectomy, may be seen in more than 50% of patients and its absence after subtotal parathyroidectomy may even be associated with recurrence (85, 86). Rates of permanent hypoparathyroidism are dependent on the procedure performed and vary greatly between series. It is important to realize that, unless patients are truly aparathyroid, recovery of parathyroid function can occur after 6 months up to several years, and permanent hypoparathyroidism may therefore be more aptly termed “prolonged” hypoparathyroidism (86). To prevent hypoparathyroidism immediate autotransplantation is used when it is suspected that all parathyroid glands are resected or when there is concern about parathyroid tissue viability in situ (57). Cryopreservation with delayed autotransplantation can also be used as a rescue from permanent hypoparathyroidism, but is not available everywhere and its use has been under debate (57).

 

Non-Surgical Interventions

 

For those patients who require intervention, but who are not surgical candidates, cinacalcet, an allosteric agonist of the calcium receptor, can be used. It has been shown to reduce/normalize calcium and PTH in small studies in patients with MEN1, although it has no effect on bone and renal complications(87-89). Cinacalcet should be used with great caution in children, as a death from acute hypocalcemia has been reported in a 14-year-old (90). Another alternative may be ethanol ablation of enlarged parathyroid glands. A study from the Mayo Clinic reported results from 37 patients who had an average of 2.2 treatments and a mean duration of eucalcemia of 25 months. Complications were hypocalcemia in 8%, hoarseness in 5%, and cough in 1% (91).

 

Parathyroid Carcinoma

 

Parathyroid carcinoma is a very rare endocrine malignancy seen in <1% of all patients with pHPT (92). It is likewise very rare in patients with MEN1, with only 21 reported cases in the literature (based on a review published in 2020) (93). In three large series from The University of Texas MD Anderson Cancer Center, the Mayo Clinic, and The Peking Union Medical College Hospital the prevalence of parathyroid carcinomas was 2/242 (0.8%), 1/348 (0.3%) and 1/153 (0.7%) respectively and the prevalence of atypical parathyroid neoplasm was 1/242 (0.4%), 0, and 2/153 (1.3%) respectively (93-95).

 

Conclusion

 

In conclusion, pHPT in MEN1 has an almost complete penetrance and is responsible for most MEN1-related surgeries. It is a multiglandular disease and recurrence after initial operation is to be expected. End-organ damage (bone, renal) can occur early and in asymptomatic patients and should be systematically looked for. Recognizing MEN1 in a patient presenting with apparently sporadic pHPT has important consequences for both the patient and his/her family. Surgical decision making is complex both for initial and reoperations and patients with MEN1 should whenever possible be treated in centers of expertise by a high-volume endocrine surgeon. Treatment decisions are made by multidisciplinary teams in shared decision making with the patient taking into account not only medical information but also the patient’s individual situation, such as but not limited to, ability to adhere to follow-up and insurance issues.

PITUITARY ADENOMAS

 

In 1903, the first description of a case with MEN1 was published by Erdheim. The necropsy report of a patient with acromegaly revealed a pituitary adenoma and enlarged parathyroid glands (96). Pituitary adenomas (PAs) are one of the three cardinal features associated with MEN1 and part of the so-called ‘three Ps’ (Figure 1). PAs are in general benign lesions and do not seem to negatively affect survival in patients with MEN1(26), although cases of mortality due to PAs have been reported (27). However, they can cause significant morbidity due to mass effect on the optic chiasm or hormone secretion leading to functional symptoms or hormone deficiency.

 

As in other main manifestations of MEN1, loss of heterozygosity (LOH) at the MEN1 locus has been demonstrated in pituitary adenomas in patients with MEN1, confirming the role of MEN1 in the pathogenesis of these tumors (97-100). However, in contrast to PanNETs, the role of MEN1/menin in tumorigenesis of sporadic PAs seems to be limited. Although initially, before the identification of the MEN1 gene, 19-33% of sporadic PAs showed allelic loss on chromosome 11 (101, 102), subsequent studies investigating LOH, somatic mutations, and messenger mRNA expression found limited involvement of MEN1 in sporadic PAs (103-107).

 

As the prevalence of clinically relevant PAs is 68-98/100,000 in the general population and in general <3% of patients with a PA will have MEN1, the question is when to think of MEN1 in a patient presenting with a PA (2, 35). Obviously, MEN1 should be considered in a patient with a family history of MEN1-related tumors or presenting with other MEN1-related tumors. For patients with apparently sporadic PAs (no suspicious family history or syndromic features), a recent systematic review has shown that MEN1 mutation analysis is recommended in patients ≤ 30 years, although this was a weak recommendation based on low quality of evidence (35).

 

Characteristics of Pituitary Adenoma in MEN1

 

From the earliest descriptions of MEN1 in the 1950s PAs have been recognized as one of the main characteristics of the syndrome. However, since the original description of MEN1, the clinical picture of MEN1-related PAs has changed. In a summary of the first 85 reported cases of MEN1 (many of which were autopsy cases), Ballard found a very high prevalence of 65% of PA, with 42% being chromophobe adenomas and more than one in four being acromegaly/eosinophilic adenoma (108). With the discovery of prolactin, it was soon realized that in fact prolactinomas were the most frequently occurring PA in patients with MEN1. The discovery of the MEN1 gene in 1997 (6, 7), and more advanced genetic testing techniques such as NGS and MLPA, have allowed better identification of patients as having MEN1. This has led to the recognition that patients with a clinical diagnosis of MEN1 because they have two out of the three main MEN1-related tumors, but negative mutation analysis, have a different clinical course than mutation positive patients and arguably do not have true MEN1, but rather an MEN1-like syndrome or a co-occurrence of two sporadic tumors (24, 25). Most patients in this group have a clinical MEN1 diagnosis based on the combination PA and pHPT. As these patients may have been included in older MEN1 cohorts, before the widespread availability of genetic testing, and these patients seem to have macro-adenomas and somatotrophinomas more often, this can be one of the reasons of the changing clinical picture of MEN1-related PAs. Additionally, imaging techniques have markedly improved over the last decades and guidelines have been developed for the screening and surveillance of patients with MEN1 including regular pituitary imaging and biochemical screening using Insulin-like Growth Factor-1 (IGF-1) and prolactin (2, 5). All this has led to earlier identification of PAs in patients with MEN1 and more frequent detection of (small) non-functioning PA (NFPAs).

 

After the discovery of the MEN1 gene (1997), six cohorts of MEN1-PA have been published, the first two by the French multicenter Groupe d’étude des Tumeurs Endocrines (GTE) in 2002 (109) and 2008 (110), in 2015 the DutchMEN Study Group (DMSG) published the results from their national population-based database (111), which was followed by two single-center cohort from China (112) and the Mayo Clinic respectively (113). Recently, the GTE have published an update to their previous study, only including patients diagnosed since January 1st 2000 (114).

 

As in sporadic PAs, PAs in patients with MEN1 show a slight female predominance (52-69%) (109, 111-114). With exception of the Chinese cohort, where the mean age of diagnosis was 54 years (112), the mean/median age of diagnosis of MEN1-related PAs is in the fourth decade. Lifetime prevalence of a PA in patients with MEN1 is 49-58% (38, 111).

 

Although not as frequent as pHPT, PAs are often the first clinical manifestation of the MEN1 syndrome. In the Dutch cohort, in 29% of the patients with a PA, it was the first manifestation (111). In the most recent GTE cohort, 88/202 patients with a PA were the index case in their family and in 84% of these patients a PA was (one of) the first manifestation(s) (114).

 

Prolactinomas are the most prevalent PA in patients with MEN1 and account for 30-80% of adenomas diagnosed in patients with clinically evident disease (42, 43, 109, 111-114). Second most prevalent are non-functioning PA comprising 36-48% in the most recent cohorts (111-114). Other functioning PAs are seen in <10%, and are in decreasing order of prevalence somatotropinomas, ACTH-producing adenomas (Cushing’s disease), and TSHomas and gonadotropinomas (the latter two being equally rare) (109, 111-114). Co-secreting tumors are seen in less than 10% (109, 111-114).

 

Multifocal PAs are rare in MEN1, and are found in 1.5% in the most recent GTE cohort (114) and in 4% in the 2008 GTE cohort of surgically resected MEN1-related PAs (110). In this latter cohort the prevalence of multifocal tumors was compared to that in non-MEN1 resected PAs and was found to be significantly larger. Additionally, MEN1-related resected PAs were more often plurihormonal on immunostaining (110).

 

Signs and symptoms in MEN1-related PAs (Table 1) are not different from those observed in sporadic PAs and are caused by size effects (chiasm compression, compression of nerves in the cavernous sinus, hypopituitarism) and effects of hormonal hypersecretion in functioning tumors.

 

Table 1. Signs and Symptoms of Pituitary Adenomas in MEN1

Related to tumor size/ growth

headache, visual field defects (usually bitemporal hemianopsia), diplopia, hypopituitarism

Prolactinoma

females: amenorrhea, galactorrhea, infertility

males: hypogonadism, impotence, lack of libido, galactorrhea (rare), infertility

Somatotrophinoma

Acromegaly: local overgrowth of bone (most often mandible, skull), soft tissue growth (acral enlargement, coarse facial features), hyperhidrosis, fatigue, hyperglycemia, hypertension, sleep apnea, skin tags, hypogonadism.

Corticotrophinoma

Cushing syndrome: central obesity, hypertension, hyperglycemia, gonadal dysfunction, moon facies, plethora, osteoporosis, proximal muscle weakness, psychological disturbance, wide purple striae, easy bruising

Thyrotropinoma

heat intolerance, unintentional weight loss, anxiety, tremor, palpitations, frequent bowel movements

Gonadotropinoma

hypogonadism, ovarian hyperstimulation in women

Pediatric specific

delayed or halted pubertal development, primary amenorrhea (females), accelerated linear growth, poor growth velocity, decline in school performance

 

Presently, most non-functioning PAs in MEN1 are microadenomas detected by prospective screening. These micro-adenomas show indolent behavior during follow-up. In the Dutch series after a median follow-up of 5.3 yrs, 9.7% showed minimal tumor growth which was without clinical significance in all and none progressed to macro-adenoma (111). In the Mayo Clinic cohort, in those with asymptomatic non-functioning PA (size not specified) progression to surgery was seen only in 1.7/100yr (113). In the most recent GTE cohort, after a median follow-up of 2 years (IQR 0-4), progression in Hardy classification was only seen in 1 out of 63 patients with a non-functioning micro-adenoma (2%) (114). In the Chinese cohort, of the 19 patients with non-functioning micro-adenomas, no progression to macro-adenomas was seen during a median follow-up of 3 years (112).

 

Prolactinomas are also mostly micro-adenoma, while 30-38% are macro-adenomas (111, 113, 114). As in sporadic PAs, GH-secreting tumors are more often macro-adenomas and ACTH-secreting tumors are generally microadenomas.

 

Although the youngest patient with a clinical manifestation of MEN1 described in the literature is a 5-year-old boy with gigantism and a lactosomatotroph macro-adenoma (36), PAs are rare in patients with MEN1 below the age of 10 (37, 46-49). However, pediatric cohorts show that in children and adolescents who have clinical manifestations of MEN1 up to 1/3 have PAs (37, 47-49). As in adults, most PAs the pediatric and adolescent age are prolactinomas followed by non-functioning PAs and more rarely GH or ACTH producing tumors (37, 46-49, 115). In the two largest pediatric cohorts, PAs were symptomatic in 50% of the cases and were macro-adenomas in 33-51% (47, 48).

 

Treatment

 

The treatment of MEN1-related pituitary adenomas follows the same strategy as sporadic pituitary adenomas. Management is aimed at tumor reduction, normalization of hormone secretion, and preservation of pituitary function.

 

Dopamine agonists are the first line of treatment for patients with prolactinoma (116), in which cabergoline has proven to be most effective at restoring normal prolactin concentrations and achieving tumor shrinkage than other dopamine agonists. With regards to adverse effects, cabergoline shows fewer side effects than bromocriptine. In case of treatment resistance, or treatment intolerance, surgery or radiotherapy are considered as second-line treatment options (116).

 

In Cushing’s disease(117) and acromegaly(118) surgery is the first treatment option. In addition, non-functioning PAs with mass effect or rapid progressive adenomas will also benefit from surgery.

 

MEN1-related functional PAs were initially considered more resistant to medical treatment than those with sporadic disease (109). However, the latest reports do not confirm this (111, 114) and treatment results seem to be in line with what is reported in sporadic PAs. The latter cohorts consist of a population with meticulous surveillance and therefore PAs are detected in an early phase (111, 114). 

 

Pituitary Carcinoma

 

Pituitary carcinoma is extremely rare, and this is equally so in patients with MEN1. Although at higher risk for PA than the general population, there does not seem to be an increased risk of pituitary carcinoma. Single cases of malignant, metastatic prolactinoma (119, 120), gonadotropinoma (121), thyrotropin secreting adenoma (122), and non-functioning PA (123) have been reported.

 

Surveillance for Pituitary Adenoma

 

Current guidelines recommend examination by MRI of the pituitary gland every three years from the age of five years, and an annual blood test of IGF-I and prolactin concentrations, together with a clinical assessment (2). The young starting age – which was based on a single case-report – has been disputed, given that PAs are rarely seen before the age of 10.

 

The aim of surveillance imaging is to detect the PAs in an early phase before clinical symptoms become apparent. In general, surveillance leads to detection of smaller non-functioning PAs (111, 113, 114). However, early diagnosis by surveillance is not associated with smaller prolactinomas, but treatment is required less frequently and a longer safe observation period can be conducted (111). There are currently no specific recommendations for the follow-up of MEN1-related (micro-)adenomas under observation, on medical treatment or after surgical resection.

 

DUODENOPANCREATIC NEUROENDOCRINE TUMORS AND GASTRIC NETS

 

General

 

Duodenopancreatic neuroendocrine tumors (dpNETs) (Figure 1) are one of the cardinal features of MEN1 and highly penetrant, with a prevalence of over 80% at the age of 80 in recent cohorts (24, 38, 124). Malignant dpNETs are the most important cause of MEN1-related death (26, 125).

 

Duodenopancreatic NETs in MEN1 can secrete hormones that produce a clinical syndrome or be functionally silent (non-functioning, NF). Due to improved imaging techniques in the past decades, including endoscopic ultrasound (EUS) and somatostatin receptor (SSTR) imaging, non-functioning pancreatic NETs (NF-PanNETs) are now recognized as the most frequent type of dpNET in patients with MEN1. Of the functional dpNETs, gastrinomas are the most frequent, seen in approximately 30% of patients with dpNETS. In patients with MEN1, gastrinomas are almost exclusively of duodenal origin (126). Insulinomas (pancreatic in origin) are the second most common functional dpNET and occur in approximately 10-15% of patients with MEN1. More rare functional PanNETs such as glucagonomas, vipomas, somatostatinomas (127) or even rarer PanNETs secreting GHRH (128), calcitonin or PTH-related peptide (129), can also occur. Upon histological examination of the duodenum in patients with MEN1, small somatostatin-positive tumors can also be found (130) although they do not seem to give rise to the somatostatinoma syndrome.

 

The hallmark of duodenopancreatic involvement in MEN1 is multifocality, with the pancreas usually containing multiple NETs <5mm, called micro-adenomas, combined with one or more macroscopic PanNETs (130). These micro-adenomas already have loss of heterogeneity (LOH) of the MEN1 locus and are considered precursors to PanNETs (130). Similarly, duodenal gastrinomas in MEN1 are usually multiple and accompanied by gastrin cell hyperplasia, although LOH was demonstrated in duodenal gastrinomas, but not in gastrin cell hyperplasia (131). This multiplicity sets MEN1-related dpNETs apart from sporadic duodenal and pancreatic NETs, which are usually single tumors.

 

For patients with MEN1, the cumulative probability of having a dpNET increases with age, however the age of onset varies somewhat per tumor type. In a recent study from the Dutch MEN1 cohort, the modeled cumulative probability of having developed a NF-PanNET was 8.6% (95%CI 0.8-15.3%) at age 15, 12% (95% CI 5.9-17.0) at age 18, 16.1% (11.2-21.5) at age 21 and rising to 80% at age 70 (72.2-97.0)(15). Insulinomas can also occur at a young age and the prevalence of insulinoma among the larger (n>50) cohorts describing pediatric and adolescent MEN1 ranges from 6-25% (37, 46-48). Data from a recent multicenter cohort study show that half of the patients with MEN1-related insulinoma were diagnosed before the age of 30 (96 patients who underwent surgery for MEN1-related insulinoma from 46 centers in Europe and North-America between 1990-2016) (132). The onset of gastrinomas is usually later, with a reported mean age of onset around 30-35 years in the National Institutes of Health (NIH) MEN1-ZES cohort (133, 134) to 51 years in the Dutch MEN1 cohort (135). The occurrence of MEN1-related gastrinoma in childhood or adolescence is rare.

 

Duodenopancreatic NETs can be the first manifestation of MEN1, both in patients from known MEN1 families but also in the index case. Approximately 20-25% of all patients with gastrinoma have MEN1 (136), a rate much lower for insulinomas (approximately 5%) (2). Therefore, genetic testing for MEN1 is recommended in all patients diagnosed with gastrinoma (137). For patients presenting with a non-gastrinoma dpNET without a family history of MEN1, referral for genetic testing should be guided by the individual clinical characteristics, such as patient age, concomitant other MEN1-related tumors, multifocality of dpNETs, and family history of endocrine tumors. If a new diagnosis of MEN1 is made in a family, cascade screening and subsequent screening and lifelong surveillance of affected family members is of utmost importance, as delays may lead to preventable morbidity and mortality in non-index cases in the family (30).

 

Distant metastases occur in approximately 15-30% of MEN1-related dpNETs and are the most important prognostic factor for disease-related survival (125, 134, 138, 139). In the Dutch MEN1 cohort, 5- and 10-year overall survival rates were 95% and 86% for patients with dpNETs without liver metastases, compared to 65% and 50% for those with liver metastases (139). Non-functional pancreatic NETs and duodenal gastrinomas are the most frequent cause of distant metastases. Regional lymph node metastases are seen more often, but the exact reported prevalence highly depends on the type of cohort, primary dpNET, and the manner of diagnosis (i.e., surgical cohorts versus observational cohorts, surgery with or without systematic lymph node dissection, imaging with or without SSTR-PET imaging, etc.). In a recent publication from the Dutch MEN1 cohort, in 350 patients with MEN1-related NF-PanNETs without metastases at diagnosis, metastases (regional and/or distant) developed in 18%, while the cumulative probability of having any PanNET-related metastases at the age of 70 was 41.2% (95%CI 31.3-50.3) (15). Since patients with MEN1 often have multiple concomitant dpNETs and most patients with duodenal gastrinomas have concomitant NF-PanNETs, it may be difficult to determine the primary tumor for regional and distant metastases.

 

Unlike in MEN2, in MEN1 there is no clear genotype-phenotype correlation. Several groups however have studied the association between MEN1 germline mutation and the disease course of dpNETs in their cohorts, to see if genotype might be able to identify a subset of patients with a more aggressive clinical course. This was in part fueled by the clinical observation that in some families dpNETs seem to be more prevalent, occur at a younger age and have a higher proportion of metastatic disease.

 

Several associations have been reported: in the French GTE cohort mutations in the JUND interacting domain were associated with death (13), in the German Marburg cohort CHES1 loss of interaction was associated with aggressive pNETs and pNET-related mortality (14), in the Italian Florence cohort mutations in exon 8 were associated with higher risk of progression and mortality (140), in the MD Anderson cohort mutations in exon 2 were associated with a higher risk of distant metastases (141), and in the Dutch MEN1 cohort nonsense/frameshift mutations were associated with a higher cumulative probability of developing metastases in NF-PanNET (regional and/or distant) compared to missense mutations 53.9 (37.8-74.3%) vs 10% (2.6-82.7%)) (15). However, these associations up until now have not been independently validated, either because associations were not confirmed in other cohorts or validation was not performed.

 

In patients with MEN1, dpNETs are usually diagnosed at an early stage, especially in patients from families with MEN1 or who have had predictive genetic testing. Additionally, even in index cases, benign MEN1 manifestations may lead to the diagnosis of MEN1 and dpNETs can be diagnosed early. In the French GTE cohort and the Dutch MEN1 cohort, both spanning multiple decades, synchronous metastases were seen in 6.5 and 6.4% of patients with a dpNET respectively (125, 139). In MEN1-related dpNETs the focus of care therefore lies before the onset of metastatic disease and with a younger population than is seen in sporadic dpNETs. The goals of follow-up and treatment are to prevent metastatic disease, cure hormonal hypersecretion, and prevent complication from hormonal hypersecretion, while minimizing treatment-related complications and preserving Quality of Life. It is therefore of utmost importance that whenever possible patients with MEN1 and MEN1-related dpNETs are treated in centers of expertise with a knowledgeable and experienced multidisciplinary team.

 

Staging and Grading

 

MEN1-related dpNET are graded according to the latest WHO classification (Table 2) of digestive system tumors (2019, 5th edition) and the WHO Classification of Tumors of Endocrine Organs (2017, 4th edition) (142). Where previously dpNET grading was only covered in the Classification of Tumors of Endocrine Organs, it is now included in the classification of digestive system tumors as well (142).

.

Table 2. WHO Classification of Digestive Neuroendocrine Tumors

Classification

Ki-67 proliferation index

Mitotic rate (mitoses/2mm2)

Well-differentiated Neuroendocrine Tumors (NET)

NET, G1

<3%

<2

NET, G2

3-20%

2-20

NET, G3

>20%

>20

Poorly-differentiated Neuroendocrine Carcinomas (NEC)

NEC (G3)

Small-cell type

Large-cell type

>20%

>20

 

Pancreatic NETs are staged according to the AJCC UICC 8th edition Neuroendocrine tumors of the pancreas (Table 3a and b).

 

Table 3a. TNM Staging of Pancreatic Neuroendocrine Tumors (AJCC UICC 8thedition)

Primary Tumor (T)

For any T add (m) for multiple tumors e.g., T2(m).

TX

Tumor cannot be assessed

T1

Tumor limited to the pancreas*, <2 cm

T2

Tumor limited to the pancreas*, 2-4 cm

T3

Tumor limited to the pancreas*, >4 cm; or tumor invading the duodenum or CBD

T4

Tumor invading adjacent organs (stomach, spleen, colon, adrenal gland) or the wall of large vessels (celiac axis or the superior mesenteric artery)

Regional lymph Nodes (N)

NX

Regional lymph nodes cannot be assessed

N0

No regional lymph node involvement

N1

Regional lymph node involvement

Distant Metastases (M)

M0

No distant metastases

M1

Distant metastases

M1a

Hepatic metastases only

M1b

Extra-hepatic metastases only

M1c

Both hepatic and extra-hepatic metastases

* Limited to the pancreas means no invasion of adjacent organs or the wall of large vessels. Extension into peripancreatic adipose tissue is included in “limited to the pancreas”. CBD common bile duct

 

Table 3b Stage Grouping

Stage I

T1 N0 M0

Stage II

T2-3 N0 M0

Stage III

T4 N0 M0

Any T N1 M0

Stage IV

Any T Any N M1

 

Duodenal NETs are staged according to the AJCC UICC 8th edition Neuroendocrine Tumors of the duodenum and ampulla of Vater (Table 4a and b).

 

Table 4a. TNM Staging of Duodenal Neuroendocrine Tumors (AJCC UICC 8th edition)

Primary Tumor (T)

If the number of tumors is known use T (#), if unavailable or too numerous T(m) e.g., T2(3) or T2(m)

TX

Tumor cannot be assessed

T1

Tumor invades the mucosa or submucosa only and is ≤ 1 cm (duodenal)

Tumor ≤ 1 cm and confined within the sphincter of Oddi (ampullary)

T2

Tumor invades the muscularis propria or is >1 cm (duodenal).

Tumor invades through sphincter into duodenal submucosa or muscularis propria, or is >1 cm (ampullary).

T3

Tumor invades the pancreas or peripancreatic adipose tissue

T4

Tumor invades the visceral peritoneum (serosa) or other organs

Regional lymph Nodes (N)

NX

Regional lymph nodes cannot be assessed

N0

No regional lymph node involvement

N1

Regional lymph node involvement

Distant Metastases (M)

M0

No distant metastases

M1

Distant metastases

M1a

Hepatic metastases only

M1b

Extra-hepatic metastases only

M1c

Both hepatic and extra-hepatic metastases

 

Table 4b. Stage Grouping

Stage I

T1 N0 M0

Stage II

T2-3 N0 M0

Stage III

T4 N0 M0

Any T N1 M0

Stage IV

Any T Any N M1

 

Non-Functioning Pancreatic NETs

 

In patients with known MEN1, screening is advised for early detection of NF-PanNETs. When NF-PanNETs are diagnosed and there is no immediate indication for intervention, surveillance should be performed at regular intervals to re-evaluate indications for intervention, as well as to detect newly developing dpNETs. Current guidelines suggest to start screening for NF-PanNETs in MEN1 below the age of 10 by a combination of biochemical tests and yearly imaging (either MRI, CT or EUS) (2).

 

Since the publication of the guidelines, it has become clear that the diagnosis of NF-PanNETs in patients with MEN1 relies heavily on imaging, since tumors markers chromogranin A, pancreatic polypeptide, and glucagon have low accuracy for the diagnosis of NF-PanNETs as summarized in a recent systematic review (143). Additionally, since the publication of the guidelines SSTR-PET-CT has emerged as a high-sensitive diagnostic imaging tool for dpNETs and its role within the screening and surveillance of MEN1-related dpNETs has yet to be determined.

 

There is currently no consensus regarding the best imaging modality and interval for screening and surveillance of MEN1-related NF-PanNETs (144). In a recent systematic review on the diagnosis of NF-PanNETs in MEN1, it was concluded that for lifelong screening and surveillance CT was probably least suitable given the inferior sensitivity compared to EUS and SSTR-PET-CT in combination with the cumulative exposure to ionizing radiation, although head-to-head comparisons with MRI are not available (143). This does not mean that a CT scan cannot still be indicated and be the best imaging for specific clinical situations (for example pre-operative imaging). EUS is the most sensitive method for the diagnosis of NF-PanNETs and offers the possibility of obtaining tissue for analysis pre-operatively. However, it is also invasive, operator dependent, clinically significant PanNETs can be missed in the pancreatic tail, and for the diagnosis of NF-PanNETs in MEN1 histological confirmation is usually not necessary given the high pre-test likelihood and the typical appearance on imaging. On the other hand, tissue-based analysis prior to intervention may become more relevant in MEN1 as more novel prognostic factors are identified. MRI has the advantage of performing more homogenously throughout the pancreas and the absence of ionizing radiation although a significant proportion of NF-PanNETs >2 cm is missed. The authors of the systematic review therefore suggest alternate use of EUS and MRI (143).

 

Given a reported growth rate of 0.1-1.32 mm/year for small NF-PanNETs, if initial screening imaging is negative, the next imaging can be performed with an interval of two to three years, providing no clinical reason for earlier imaging. For prevalent NF-PanNETs without an indication for intervention, the interval for active surveillance imaging should be individualized according to growth rate. Initial repeat imaging should be done after 6-12 months to assess growth rate, but afterwards in small stable NF-PanNETs the interval can be extended to once every 1-2 years. The imaging modality can be either MRI or alternating with EUS. EUS alone should always be combined with an imaging method for metastases detection, since it does not offer complete abdominal imaging. The exact role of SSTR-PET-CT in the screening and surveillance of MEN1-related NF-PanNETs is still to be determined, however, based on currently available evidence, it is best employed when results may change management such as in prevalent NF-PanNETs >10 mm for early detection of metastases, or as comprehensive staging before planned interventions (143, 145).

 

Another dilemma is when to start radiological screening for NF-pNET in children with MEN1. As mentioned before, current guidelines advise initiating screening before the age of 10 (2). However, others have advocated postponing until the age of 16, in the absence of signs and symptoms (37). Recently, modeled data from the Dutch population-based MEN1 cohort show that the estimated age at a 1%, 2,5% and 5% risk of having developed a clinically significant NF-PanNET (≥ 20mm or documented growth of ≥1.6 mm within one year above a baseline size of ≥ 15mm) is 9.5, 13.5 and 17.8 years respectively and they conclude that there is medical indication to initiate radiological screening during the second decade of life and that starting between 13-14 years of age is justifiable (15).

 

It is important to remember that each screening and surveillance schedule should be tailored to the needs of the individual patient in his or her unique circumstances, should be based on well-informed shared decisions making between providers and patients (and parents if applicable), with multidisciplinary team input when necessary.

 

The only curative treatment for NF-PanNETs in MEN1 is surgical resection, and the goal of surgical intervention in NF-PanNETs is to prevent metastases and thereby NF-PanNET-related mortality, while preserving as much pancreatic tissue as possible and limiting treatment-related morbidity and mortality. Although theoretically, total duodenopancreatectomy would prevent metastatic disease altogether, short-term morbidity associated with this complex major surgery is high and the subsequent life-long brittle diabetes that follows rarely justifies such major intervention when balanced against the risk of distant metastases and PanNET-related death.

 

Since the risk of future metastases and disease-related death must be balanced against short- and long-term treatment-related morbidity and mortality, information regarding prognosis in MEN1-related NF-PanNETs is of vital importance to make well-informed decisions regarding timing and extent of intervention. However, presently there is a paucity of prognostic factors on which to base these decisions (146). The most important factor to date is tumor size, with the risk of (distant) metastases increasing with increased size. Recent data from retrospective cohort studies have shown that small (<2cm) NF-PanNETs generally have an indolent course, that surgical resection of small NF-PanNETs does not seem to offer benefit over active surveillance, and that the risk of metastases and disease-related death is low, albeit not zero (124, 146-150). Most small NF-PanNETs are stable during follow-up, but there is a subset with progression in size (150). Generally, size progression is also considered to be a prognostic factor. An important tissue-based prognostic factor is tumor grade, with grade 2 tumors being more often associated with metastases (146). Grade 3 NF-PanNETs or NECs are rarely seen in patients with MEN1, but are associated with a worse prognosis. More recently, advancements in molecular techniques have identified several potential prognostic biomarkers for NF-PanNETs, mostly in sporadic NF-PanNETs, but limited data in MEN1-related NF-PanNETs is also available. Mutations in alpha-thalassemia/mental retardation X-linked (ATRX) and death domain-associated protein (DAXX), which lead to the alternative lengthening of telomeres (ALT) phenotype have been found to be associated with decreased disease-free survival and higher rates of distant metastases (146, 151). Mutations in DAXX and ATRX result in loss of nuclear expressions of their proteins by immunohistochemistry (IHC) and ALT can be identified in tissue-samples by telomere-specific fluorescence in situ hybridization (FISH). Next to DAXX/ATRX and ALT, the differential expression of transcription factors aristaless-related homeobox gene (ARX) and pancreatic and duodenal homeobox 1 (PDX1) as assessed by IHC was also found to be associated with risk of metastases (152, 153). In patients with MEN1-related NF-PanNETs, one study showed that liver metastases were only seen in ARX+ or ARX-/PDX1- tumors and that ALT positivity was only seen in ARX+ or ARX-/PDX1- tumors and significantly correlated with relapse rate (152). However, since the publication of these data, a large international cohort of 1322 NETs (not including MEN1-related NETs), was evaluated by immunolabelling for ARX/PDX1, ATRX/DAXX and by telomere-specific FISH for ALT and it was found that ATRX/DAXX and ALT, but not ARX/PDX1 were independent negative prognostic factors (151).

 

A recent study by Fahrmann, et al. identified a 3-marker polyamine signature that distinguished patients with metastatic dpNETs from controls and which yield an AUC of 0.84 (95% CI: 0.62-1.00) with 66.7% sensitivity at 95% specificity for distinguishing cases form controls in an independent test set (154). These results form the basis for prospective testing of plasma polyamines as a prognostic factor for MEN1-related dpNETs.

 

Further validation of these molecular markers in MEN1, may also change the role of pre-intervention EUS-guided aspiration or biopsy.

 

So, when to intervene in MEN1-related NF-PanNETs? Presently, these decisions are mostly based on tumor size and growth, with the current guidelines suggesting considering surgical resection in NF-PanNETs >10 mm or those that show significant growth during follow-up (doubling of tumor size over a 3–6-month interval and exceeding 10 mm) (2). Given the emerging evidence that most  NF-PanNETs <20 mm are indolent in nature as described above, a more recent consensus statement states that surgical resection is indicated for  NF-PanNETs >20 mm and those that progress during surveillance (155). Additionally, the presence of suspicious lymph nodes, or a higher grade on EUS-guided aspiration may guide intervention decisions. In all cases these decisions should be made in multidisciplinary teams and in shared decision making with the patient. The extent of resection depends on multiple patient-, tumor- and MEN1-related factors and should be individualized.

 

Gastrinoma

 

Gastrinomas, NETs secreting gastrin, cause the Zollinger-Ellison Syndrome (ZES). ZES is a syndrome characterized by tumor-related hypergastrinemia leading to gastric acid hypersecretion.

 

Sign and symptoms of ZES/Gastrinoma are gastro-esophageal reflux disease (GERD), (proton-pump inhibitor (PPI) responsive) diarrhea, abdominal pain, nausea/vomiting, weight loss, and peptic ulcer disease. Complications may arise from the peptic ulcer disease including upper gastro-intestinal bleeding, strictures, and bowel perforation.

 

Before the introduction of PPIs, complications from gastric acid hypersecretion were an important cause of death in patients with MEN1 (134). With the arrival of proton pump inhibitors, gastric acid hypersecretion can be effectively treated, although higher dosages are needed than for the treatment of non-ZES hyperacidity.

 

The diagnosis of gastrinoma in MEN1 is challenging at present. The gold standard for the diagnosis is the demonstration of inappropriate fasting hypergastrinemia without the use of antisecretory drugs. The diagnosis is established if the fasting serum gastrin (FSG) is more than tenfold the upper limit of normal with a gastric pH of less than two (after ruling out retained antrum) (156, 157). When gastric pH is low and FSG is <10-fold upper limit of normal, additional testing is needed to establish the diagnosis, such as a secretin provocative test or measuring basal acid output (156, 157). The latter situation occurs in 60% of ZES, and this might even be higher in MEN1, given the early detection through prospective screening programs. Due to unreliable gastrin assays, the limited availability of secretin and therewith the loss of expertise in performing the secretin provocative test, the wide-spread use of PPIs and the risk associated with cessation of PPI for proper testing, the diagnosis of gastrinoma is challenging (157, 158). Additionally, the value of non-biochemical tests in the diagnosis of gastrinoma (such as SSTR-PET-CT and EUS/ esophagogastroduodenoscopy (EGD)-guided cytology/biopsy) might also need re-evaluation (157). Recently experts have therefore suggested possible new criteria for diagnosis of ZES in patients with fasting hypergastrinemia with and without PPI use (157).

 

Patients with MEN1 are screened for the presence of a gastrinoma by at least annually assessing clinical symptoms and fasting serum gastrin (2). If a diagnosis of gastrinoma is established or suspected, EGD should be performed to assess the presence of complications of gastric acid hypersecretion, type II gastric NETs, and possibly to identify duodenal gastrinomas. Duodenal gastrinomas in MEN1 are small, but despite their small size 70-80% are metastatic to the regional lymph nodes at the time of diagnosis (159). However, these regional lymph node metastases do not seem to have a negative impact on overall survival (159). In MEN1, attributing locoregional lymph nodes to the correct primary dpNET is important for adequate treatment planning and prognostic inferences. It is also challenging however, given that most patients with MEN1 and duodenal gastrinoma(s) also have concomitant PanNETs, and the duodenal gastrinoma(s) may not be visible on imaging due to their small size. Hackeng, et al. studied 137 microscopic and macroscopic dpNETs and 36 matched metastases (lymph node and distant) in 10 patients with MEN1 to unravel the relationship between the multiple primary dpNETs in MEN1 and the metastases (160). They found that most patients had a single NET of origin for their metastases, but multiple metastatic primaries were also seen. In addition, and very important for MEN1-related gastrinomas, in 6 patients with MEN1 and hypergastrinemia, periduodenopancreatic lymph node metastases clustered with minute duodenal gastrinomas and not with larger pancreatic NETs. So a duodenal origin for periduodenopancreatic lymph node metastases in patients with MEN1 and hypergastrinemia should always be considered (160).

 

Although its role still needs to be delineated, in MEN1-gastrinoma next to EUS/EGD, SSTR-PET-CT may be very useful for staging to visualize duodenal gastrinomas, lymph node metastases, and concomitant PanNETs.

 

Presently, as in other MEN1-related dpNETs, surgery is the only potentially curative treatment. And although MEN1-related ZES has been historically been considered a surgically incurable disease, more recent small studies have shown, that when the correct target organ is addressed, namely the duodenum and not the pancreas, biochemical cure can be achieved after partial pancreaticoduodenectomy(PD), combined with regional lymph node dissection (159). However, this must be balanced against the risk of peri-operative and long-term complications and loss of quality of life. Overall survival is generally good in duodenum-preserving operations as well, but persistence or recurrence of ZES occurs in 6-100% (159). Most often, hyperacidity can be adequately controlled with PPIs, making the main goal of surgical resection the prevention of distant metastases and disease-related death. The majority of MEN1-ZES patients with associated small PanNETs have an indolent disease course with excellent overall survival even without surgical intervention (161). Still, in retrospective studies around a quarter of the patients develop liver metastases and around 15% shows aggressive growth (138), and presently there are no good markers to predict which patients with MEN1-ZES will have a more aggressive disease course. In a study from the NIH age at ZES diagnosis (≤33), FSG levels ≥10,000 pg/mL, pancreatic tumors >3 cm, presence of liver/bone metastases and presence of gastric carcinoids were associated with aggressive tumor growth (138). In a study from the DutchMEN Study Group, overall survival rates of MEN1-gastrinoma were 83% and 65% at 5 and 10 years respectively, which was significantly worse than age- and gender-matched patients without gastrinoma. FSG ≥ 20x upper limit of normal, PanNETs≥ 2cm, synchronous liver metastases, EGD suspicious for gastric NETs, and multiple concurrent NETs were associated with decreased overall survival (135). A recent study from the French GTE, ZES was independently associated with a higher risk of distant metastases, but did not significantly seem to be associated with decreased overall survival (125).

 

So presently, if surgical intervention should be performed, when surgical intervention should be performed and how (to what extent) surgical intervention should be performed for gastrinoma in MEN1 are all controversial topics. Treatment of patients in centers of expertise with a highly dedicated multidisciplinary team and experienced surgeons is therefore very important. Treatment decisions for MEN1-ZES should be made after MDT discussion in shared decision making with the patient.

 

As stands to reason, treatment of MEN1-ZES patients with (high-dose) PPI is mandatory and patients should not cease this treatment without consultation with their provider. If specific testing without PPI is needed this needs to be performed under close supervision in centers of expertise.

 

As mentioned in the section on parathyroid tumors, the interplay between pHPT and ZES in MEN1 is important to realize as hypercalcemia can increase gastrin levels. Additionally, a recent paper on the Tasmanian MEN1 cohort showed an association between H. pylori seropositivity and hypergastrinemia and severe ZES-range hypergastrinemia. Further work is needed to fully elucidate this relationship, but testing for H. Pylori and eradication if positive might be consider in patients with MEN1-ZES (162).

 

Insulinoma

 

As already stated, MEN1-related insulinomas occur at a young age and are the most frequent functional PanNET at the pediatric age. Early recognition of signs and symptoms of insulinoma is of extreme importance in both children and adults. Signs and symptoms may be erroneously attributed to epilepsy or behavioral or neurological disorders, especially if insulinoma is the presenting manifestation of MEN1 in an index case. In children this can lead to decline in school performance and in children and adults alike episodes of hypoglycemia can lead to accidents or irrational behavior.

 

As insulinomas secrete insulin inappropriately and lead to hypoglycemia the signs and symptoms are those of hypoglycemia; both adrenergic symptoms (such as fast heartbeat, jitteriness/shakiness, sweating and pale skin) as well as neuroglycopenic symptoms (such as mental status changes and irritability). Symptoms are relieved with food (glucose) intake. They usually occur during fasting, before meals or after exercise, but can occasionally occur at other times. In patients fulfilling Whipple’s triad (symptoms and/ or signs consistent with hypoglycemia, a low plasma glucose concentration, and resolution of symptoms/signs after plasma glucose concentration is raised) diagnosis can be established by a supervised fast (163). In patients with MEN1, screening for insulinoma is advised from the age of five by careful history taking and measurement of fasting insulin and – more importantly – glucose (2). However, almost all patients with MEN1 and insulinoma are symptomatic, therefore the history is probably the most important element.

 

When the diagnosis of insulinoma is made, localization in MEN1 can be challenging, if there are multiple PanNETs. These usually are concomitant NF-PanNETs, since in surgical series multiple insulin-positive PanNETs in patients operated for insulinoma were seen in 8-40% (132, 164-166).

 

For MEN1-related insulinoma, especially if conventional imaging shows multiple PanNETs and correctly identifying the insulinoma(s) among them would change surgical strategy, 86Ga-Exendin-4 PET-CT is very promising. Although there is limited data in MEN1 patients, a recent meta-analysis showed a positive predictive value (PPV) of 94%, with a negative predictive value (NPV) of 67%; In MEN1 PPV was 95% with NPV 96%, although based on a limited number of patients (167, 168).

 

Surgical resection is the treatment of choice for MEN1-related insulinomas and is associated with a high cure rate. In a retrospective cohort study of 40 European and 6 North-American institutes 92 patients with MEN1-related insulinomas who underwent surgical resection were followed for a median of 8 years after surgery (132). Overall, after different surgical procedures, only 1 patient had persistence of hypoglycemia and six had recurrent hypoglycemia, four due to new primaries and 2 due to development of liver metastases, leading to a 10-year hypoglycemia-free survival of 91% (95% CI 80-96). For those with unifocal insulinoma based on pre- and intra-operative assessment (n=63), 1/46 (2.2%) undergoing pancreas resection had persistent disease, while among those who underwent enucleation 1/17 (6%) had recurrence of hypoglycemia based on a new primary insulinoma. For those with multifocal insulinoma (n=33), of whom 30 underwent pancreatic resection, mostly distal pancreatic resection, and three had multiple enucleations, 15% had recurrent hypoglycemia (9% based on new primaries and 6% based on liver metastases) (132).

 

Therefore, given the better outcomes of pancreatic function over the long-term and young age of the patients, if surgically feasible, enucleation seems the better option for solitary insulinomas in MEN1, provided of course that concomitant functional and non-functional tumors do not make a different strategy necessary (132).

 

Among MEN1-related dpNETs, insulinomas have the best oncological prognosis (26, 125, 169). Data from the international MEN1 Insulinoma Study Group and the DutchMEN Study Group show that for surgically resected insulinomas 10-yr liver-metastases free survival was 87% (72-91%) (169). Malignant insulinoma is rare, both in sporadic and MEN1-related insulinoma. In the two largest MEN1-insulinoma cohorts synchronous liver metastases were seen in 3.8-8.1% and metachronous liver metastases in 0-2.2% after a median follow-up of 8-9 years (132, 165).

 

Rare Functional dpNETS

 

Functional dpNETs besides gastrinomas and insulinomas, are rare in MEN1 and are seen in <1% of patients with dpNETs(2). These include PanNETs producing vaso-active intestinal peptide (VIPoma), somatostatin, glucagon and other (ectopic) hormones such as growth hormone releasing hormone (GHRH), calcitonin, or PTH-related peptide (PTHrP). A rare functional tumor is considered if there are elevated hormone levels in conjunction with a fitting clinical syndrome. Without a clinical syndrome, tumors are not considered functional but merely hypersecreting. This is relevant as for example glucagon can be elevated in patients with MEN1 and PanNETs without the patient having the glucagonoma syndrome. VIPomas lead to watery diarrhea, hypokalemia, achlorhydria and dehydration, the somatostatinoma syndrome consists of diabetes mellitus, diarrhea, steatorrhea and cholelithiasis, while glucagonomas give rise to necrolytic migratory erythema, diabetes mellitus, and weight loss. Tumors producing GHRH, calcitonin, and PTHrP lead to acromegaly, diarrhea and hypercalcemia, respectively. In these rare functional dpNETs without synchronous distant metastases surgery is generally indicated.

 

Non-Surgical Treatments of Non-Metastatic dpNETs in MEN1

 

Although for most non-metastatic functional dpNETs in MEN1 surgery is indicated, there may be a (temporary) need to control the hormonal syndrome medically. As such gastrinomas are treated with high-dose PPI, insulinomas with diazoxide or frequent feedings and in all cases somatostatin analogues might be considered if needed to control the functional syndrome.

 

Local resection of sporadic small dNETs is increasingly considered as an alternative to surgery (170), and current European Neuroendocrine Tumor Society (ENETS) guidelines recommend endoscopic management for dNETs ≤ 10mm in size, confined to the submucosal layer and without lymph node and distant metastases (171). However, since in MEN1 dNETs are usually multiple, may grow beyond the submucosa and, in case of gastrinomas, are associated with lymph nodes in up to 80% of the cases, this is not generally recommended (159).

 

Similarly, for PanNETs EUS-guided intervention using ethanol or radio-frequent ablation has been reported in around 80 and 70 cases respectively in the literature, with only a handful procedures performed in patients with MEN1 (172). Whether or not interventional EUS may play a role in treatment of MEN1-related PanNETs is therefore unclear at the present time.

 

There is much interest in chemoprevention in small NF-PanNETs using somatostatin analogues (SSA). SSA have proven anti-proliferative effect in advanced PanNETs (173, 174) and the question has been raised if SSA may be used to prevent progression and metastases of small NF-PanNETs in patients with MEN1. In mouse models of Men1PanNET lanreotide and pasireotide showed the ability to decrease tumor proliferation. In a retrospective non-controlled study of 20 patients with small NF-PanNETs who received long-acting octreotide for 12-75 months 10% had an objective tumor response, 80% stable disease, and 10% showed progression (175). In another small (n=8) prospective series patients with small NF-PanNETs were treated with SSA for up to 72 months, with stable disease in all, however again without a control group (176). In a recent observational cohort study lanreotide was compared with standard of care active surveillance in n=42 patients with pNETs <2 cm (N=23 lanreotide vs n=19 active surveillance) during a median follow-up of 6 years (177). The study showed improved RECIST-defined progression-free survival (PFS) in the lanreotide group. In both groups however, one patient developed distant (liver) metastases (177). Limitations include sample size, non-experimental and therefore non-randomized design, and non-blinded outcome evaluation. In addition, improved RECIST PFS is not yet known to predict longer overall survival for MEN1 patients with small NF-PanNETs. Ideally this is further evaluated in a randomized double-blind trial. The most important challenge in the design of such a study however, is the definition of appropriate surrogate endpoint for distant metastases and overall survival (144).

 

Metastatic dpNET in MEN1

The treatment of stage IV dpNET in patients with MEN1 is similar to that of patients with sporadic dpNETs(178). There is very limited evidence regarding MEN1-specific outcome data, and from the limited evidence available there seems to be no difference with sporadic NETs. In landmark studies leading to approval of lanreotide (173), everolimus (179), sunitinib (180) and peptide receptor radionuclide therapy (PPRT) (181), patients with MEN1 were either excluded, only single cases included or MEN1-status was not mentioned (182). With the advancing molecular understanding of MEN1-related NETs, MEN1-specific targeted therapies might be possible in the future, which in turn might benefit the almost 50% MEN1-mutated sporadic PanNETs (182).

Gastric NETs in MEN1 (Type II Gastric NETs)

 

NETs of the stomach (Figure 1), formerly called gastric carcinoids or carcinoids of the stomach, are classified into three different types(171):

  • Type I, associated with atrophic gastritis
  • Type II, associated with MEN1/ZES
  • Type III, without associated conditions

 

Gastric NETs are graded according to the latest WHO classification of digestive system tumors (2019, 5th edition) as described above for dpNETs (Table 2) (142). TNM staging is shown in Table 5a and b.

 

Table 5a. TNM Staging of Neuroendocrine Tumors of the Stomach (AJCC UICC 8th edition)

Primary Tumor (T)

For any T, add (m) for multiple tumors, for multiple tumors with different Ts, use the highest (e.g., if three tumors’ sizes 0.5, 0.5, and 1.5 cm, T stage should be T2(m).

TX

Tumor cannot be assessed

T0

No evidence of primary tumor

T1

Invades lamina propria or submucosa and ≤ 1 cm

T2

Invades muscularis propria or >1cm

T3

Invades through the muscularis propria into subserosal tissue without penetration of overlying serosa

T4

Invades visceral peritoneum (serosal) or other organs or adjacent structures

Regional lymph Nodes (N)

NX

Regional lymph nodes cannot be assessed

N0

No regional lymph node involvement

N1

Regional lymph node involvement

Distant Metastases (M)

M0

No distant metastases

M1

Distant metastases

M1a

Hepatic metastases only

M1b

Extra-hepatic metastases only

M1c

Both hepatic and extra-hepatic metastases

 

Table 5b. Stage Grouping

Stage I

T1 N0 M0

Stage II

T2-3 N0 M0

Stage III

T4 N0 M0

Any T N1 M0

Stage IV

Any T Any N M1

 

Gastric NETs are tumors of the gastric entero-chromaffin like (ECL) cells, which develop in MEN1 due to the trophic effect of gastrin on ECL-cells combined with the predisposing germline MEN1 mutation. Both components seem to be necessary for the development of gastric NETs in patients with MEN1. Gastric NETs are rarely seen in sporadic ZES patients and loss of heterogeneity was demonstrated in 75% of MEN1-related gastric NETs (183, 184), while in patients with MEN1 gastric NETs occur almost exclusively in patients with gastrinoma and regression of gastric NETs has been reported after normalization of hypergastrinemia (185, 186). ECL-cell hyperplasia is considered a precursor lesion for gastric NETs (187).

 

In the NIH-ZES cohort, 57 patients were extensively studied for gastric ECL-cell changes. All of the patients were found to have proliferative ECL-cell changes, with advanced changes in 53% and gastric NETs in 23% (188). More recently, data on ECL-cell changes in patients with MEN1 was reported from the Marburg MEN1 database (185). They reported on 38 MEN1 patients who underwent regular screening including EGD, regardless of gastrinoma status. Sixteen of these patients had a gastrinoma diagnosis, 13 of whom had biochemical ZES at the time of first EGD. They found that ECL-changes and gastric NETs were exclusively seen in patients with MEN1-gastrinoma albeit in a lower percentage than in the NIH. They found ECL hyperplasia in 62.5% of patients with a gastrinoma diagnosis, versus 0% in those without gastrinoma. No advanced ECL-cell changes were seen and gastric NETs were found in 12.5% of patients with a gastrinoma diagnosis (185). These differences might also reflect practice changes with earlier gastrinoma diagnosis due to screening and surveillance and more frequent surgical treatment of gastrinoma in the Marburg cohort compared to the NIH cohort.

 

In the NIH cohort, higher levels of FSG as well as longer duration of ZES were associated with a higher risk of advanced ECL-cells changes and gastric NETs (188). Higher levels of FSG as risk factor could not be confirmed in the Marburg cohort, however numbers of MEN1-ZES were small (185). As mentioned above, in the section on gastrinoma, the presence of gastric NETs in patients with MEN1-ZES was associated with a more aggressive disease course (138) and decreased overall survival (135).

 

Therefore, in the current MEN1 Clinical Practice Guidelines, EGD with biopsy, is recommend every 3 years in patients with MEN1 and hypergastrinemia. Although treatment of MEN1-related gastric NETs is not well established (2, 171), the guideline suggest that lesions <10 mm may remain under endoscopic surveillance, while larger tumors require endoscopic resection or local resection, which is analogue to the treatment of type I gastric NETs (2, 171).

 

The prognosis of MEN1-related gastric NETs is generally good, with metastases (regional and distant) reported in 10-30% and disease-related death <10% (171). Nevertheless, aggressive symptomatic and metastatic cases leading to mortality have been reported (189, 190).

 

Conclusion

 

In conclusion, dpNETs are highly prevalent in patients with MEN1 reaching a more than 80% penetrance at the age of 80.  NF-PanNETs are most frequently seen, followed by gastrinomas and insulinomas. Most MEN1-related dpNETs are diagnosed at an early stage and NF-PanNETs <2 cm generally have an indolent course. However, distant metastatic dpNETs (mostly NF-PanNETs and gastrinoma) are the most important cause of MEN1-related mortality. Treatment goals for MEN1-related dpNETs are therefore to prevent metastatic disease, cure hormonal hypersecretion, and prevent complication from hormonal hypersecretion, while minimizing treatment-related complications and preserving Quality of Life. Surgical resection is the mainstay for treatment, and is indicated in non-gastrinoma functional PanNETs and NF-PanNETs >2cm or with progression during follow-up. No consensus exists on the surgical treatment of MEN1-related gastrinoma. With increasing awareness of MEN1, increasingly refined and defined screening and surveillance programs, and increasing sensitive imaging modalities, MEN1-related dpNETs are detected at earlier stage and more indolent small dpNETs are seen. The main challenge at this point is therefore identifying those patients who are at risk for a more aggressive disease course and distant metastases to be able to offer those patients close follow-up schedules and earlier and more aggressive treatment, while limiting treatment-related morbidity in patients with low risk. Novel prognostic indicators are therefore needed, ideally blood-based, so minimal invasive assessment is possible. Other future directions are the investigation of chemoprevention in small NF-PanNETs.

 

Gastric NETs in MEN1 are almost exclusively seen in patients with gastrinoma and usually have an indolent course. Screening with EGD should be performed in all patients with MEN1-ZES.

 

THORACIC NEUROENDOCRINE TUMORS

 

General

 

Thoracic NETs occurring as part of the MEN1 syndrome are thymic (thNET) and bronchopulmonary NETs (bpNET) (Figure 1), although recently it was suggested that thymomas may also be part of the MEN1-related tumor spectrum (191). These tumors are not considered main disease-defining manifestations. As in other MEN1-related tumors, loss of heterogeneity (LOH) at the MEN1 locus was demonstrated in bpNETs in patients with MEN1 (97). This in contrast to thymic NETs, where until recently no LOH at the MEN1 locus was found. However, in a recent publication by the NIH, LOH was seen at the MEN1 locus in both MEN1-related thymic NETs (8 out of 12) and two (out of two) thymomas in patients with MEN1 (191). MEN1 is also the most frequently mutated gene in sporadic well-differentiated bpNETs, this is not described in thymus NET (192). However, approximately 25% of patients with thNET have germline mutations in MEN1, therefore it is very important to consider the diagnosis of MEN1 in patients presenting with a sporadic thNET (193). Thymic and bronchopulmonary NETs in MEN1 generally develop in adults. In pediatric and adolescent series (age up to 21, 31 in one series), there is only one reported case of thNET (diagnosed at age 16) and two cases of bpNET (diagnosed at age 15 and 20 respectively) (37, 46-48).

 

Staging and Grading

 

bpNET and thNET are classified according to the WHO classification (Table 6). TNM staging for thymicNETs is shown in Table 7a and b. TNM staging of bronchopulmonary NETs follows the same classification as bronchogenic lung carcinomas (Table 8a and b).

 

Table 6. WHO Classification of Bronchopulmonary and Thymic Neuroendocrine Tumors

Classification

Mitotic Rate and Necrosis

Well-differentiated

Typical Carcinoid, NET G1

Mitotic rate <2 and absence of necrosis

Atypical Carcinoid, NET G2

Mitotic rate 2-10 and/or presence of necrosis

Poorly differentiated

Neuro-endocrine carcinomas

Small-cell type

Large-cell type

Mitotic rate >10

 

Table 7a. Staging of Thymic Neuroendocrine Tumors (AJCC UICC 8th edition)

Primary Tumor (T)

TX

Tumor cannot be assessed

T0

No evidence of primary tumor

T1

 

T1a

T1b

Tumor encapsulated or extending into the mediastinal fat; may involve the mediastinal pleura

Tumor with no mediastinal pleura involvement

Tumor with direct invasion of mediastinal pleura

T2

Tumor with direct invasion of the pericardium (either partial or full thickness)

T3

Tumor with direct invasion into any of the following: Lung, brachiocephalic vein, superior vena cava, phrenic nerve, chest wall, or extrapericardial pulmonary artery or veins

T4

Tumor with invasion into any of the following: Aorta (ascending, arch, or descending), arch vessels, intrapericardial pulmonary artery, myocardium, trachea, esophagus

Regional lymph Nodes (N)

NX

Regional lymph nodes cannot be assessed

N0

No regional lymph node involvement

N1

Metastasis in anterior (perithymic) lymph nodes

N2

Metastasis in deep intrathoracic or cervical lymph nodes

Distant Metastases (M)

M0

No pleural, pericardial, or distant metastasis

M1

Pleural, pericardial, or distant metastasis

M1a

Separate pleural or pericardial nodule(s)

M1b

Pulmonary intraparenchymal nodule or distant organ metastasis

 

Table 7b. Stage Grouping

Stage I

T1 N0 M0

Stage II

T2 N0 M0

Stage IIIa

T3 N0 M0

Stage IIIb

T4 N0 M0

Stage IVa

Any T N1 M0

Any T N0-1 M1a

Stage IVb

Any T N2 M0-M1a

 

Any T Any N M1b

 

Table 8a. TNM Staging of Bronchopulmonary Neuroendocrine Tumors (AJCC UICC 8th edition)

Primary Tumor (T)

If the number of tumors is known used T (#), if unavailable or too numerous T(m) (e.g., T2a(2) or T2a(m)).

TX

Primary tumor cannot be assessed or tumor proven by presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy

T0

No evidence of primary tumor

Tis

Tumor in situ

T1

 

 

T1a(mi)

T1a

T1b

T1c

Tumor ≤3 cm in greatest dimension surrounded by lung or visceral pleura without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus)*

Minimally invasive adenocarcinoma

Tumor ≤1 cm in greatest dimension

Tumor >1 cm but ≤2 cm in greatest dimension

Tumor >2 cm but ≤3 cm in greatest dimension

T2

 

 

 

 

 

T2a

T2b

Tumor >3 cm but ≤5 cm or tumor with any of the following features:

Involves main bronchus regardless of distance from the carina but without involvement of the carina

Invades visceral pleura

Associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung

Tumor >3 cm but ≤4 cm in greatest dimension

Tumor >4 cm but ≤5 cm in greatest dimension

T3

Tumor >5 cm but ≤7 cm in greatest dimension or associated with separate tumor nodule(s) in the same lobe as the primary tumor or directly invades any of the following structures: chest wall (including the parietal pleura and superior sulcus tumors), phrenic nerve, parietal pericardium

T4

Tumor >7 cm in greatest dimension or associated with separate tumor nodule(s) in a different ipsilateral lobe than that of the primary tumor or invades any of the following structures: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, and carina

Regional lymph Nodes (N)

NX

Regional lymph nodes cannot be assessed

N0

No regional lymph node involvement

N1

Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension

N2

Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s)

N3

Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s)

Distant Metastases (M)

M0

No distant metastasis

M1

Distant metastases

M1a

Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodule(s) or malignant pleural or pericardial effusion

M1b

Single extrathoracic metastasis

M1c

Multiple extrathoracic metastases in one or more organs

 

Table 8b. Stage Grouping

Stage 0

Tis N0 M0

Stage IA

T1 N0 M0

Stage IB

T2a N0 M0

Stage IIA

T2b N0 M0

Stage IIB

T1a-c N1 M0

T2 N1 M0

T3 N0 M0

Stage IIIA

T1a-c N2 M0

T2 N2 M0

T3 N1 M0

T4 N0-1 M0

Stage IIIB

T1a-c N3 M0

T2 N3 M0

T3 N2 M0

T4 N2 M0

Stage IIIC

T3-4 N3 M0

Stage IVA

Any T Any N M1a-b

Stage IVb

Any T Any N M1c

 

Thymic NET

 

ThNETs develop in 2.0 - 8.2% of MEN1 patients, with a median age at diagnosis of 43 years (range 16–72 years) (194-201). There is a strong male predominance (male to female ratio 4:1) in MEN1-related thNET, which is more pronounced in American and European cohorts compared to Asian series (194). Although one of the earliest studies on MEN1-related thNET suggested a higher prevalence of truncating MEN1 mutations in patients with thNET (202), no clear genotype-phenotype relationship has been described in later cohorts (196, 197, 199, 200). Furthermore, familial clustering of thNET within MEN1 families has been reported in a number of studies (193, 197, 199) but others could not find comparable results (196, 200). The suggested link between smoking and the occurrence of thNET in MEN1 remains controversial as well, as the portion of (heavy) smokers varied significantly among studies (193-196, 199, 201).

 

With the exception of a small subset of ACTH-producing tumors, most thNETs are functionally silent. As a result, the majority of patients only experiences symptoms when the tumor has reached an advanced stage, underlining the importance of periodic thoracic imaging for a timely detection.

 

MEN1-related thNET are characterized by their aggressive nature, illustrated by their frequent presentation with metastatic disease (53.5% of patients), usually located in lymph nodes, bones and lungs (194). Despite the low prevalence of thNETs among patients with MEN1, they are responsible for 19% of MEN1-related deaths (134). The poor prognosis of MEN1-related thNET has also been illustrated in a meta-analysis of 99 MEN-1 thNETs: median survival was 8.4 years, and the 10-year survival rate was 33%. An older age at diagnosis, a tumor diameter >5 cm and the presence of metastasis were associated with worse outcome (194).

 

Total (thoracic) thymectomy, including excision of the tumor, the entire thymus and perithymic fat, is the recommended treatment of choice (2, 203). Additional radiotherapy and chemotherapy may be used in patients with unresectable or metastatic disease. Data from the earlier mentioned meta-analysis suggested that adjunctive therapy after surgery tended to result in a better survival compared to surgery alone (after adjusting for gender, age at diagnosis, tumor size and smoking), but this effect did not reach statistical significance (HR 0.557, 95%CI: 0.110–2.817) (194). Prophylactic cervical thymectomy, generally performed during parathyroid surgery for primary hyperparathyroidism, may decrease the chance of the occurrence of thNET. However, several cases of thNET have been reported in patients after this procedure, indicating that surveillance imaging is still required in these patients (197, 202).

 

Bronchopulmonary NET

 

Histopathologically proven bpNETs occur in 4.7-6.6% of MEN1 patients, but a much higher proportion of MEN1 patients may be diagnosed with lesions radiologically suspect of bpNET (22.9%, 26.0% and 29.3% in the Dutch, Tasman and German cohort respectively) (195, 204-207). BpNETs are diagnosed at a median age of ± 45 years and the reported age at bpNET diagnosis ranges between 20 and 69 years. Although the earliest report suggested a female predominance among MEN1 patients with bpNET (205), later studies could not find a relationship between the occurrence of bpNET and sex (200, 204, 206, 207). Likewise, genotype (the type of mutation) or smoking status does not seem to influence the development of bpNET in MEN1 patients (204, 206, 207).

 

Only a minority of patients experience symptoms (dyspnea, cough, hemoptysis), which explains the high rate of bpNET (77–100%) diagnosed through periodic thoracic imaging surveillance (195, 206, 207). Growth analysis of lung lesions highly suspect of bpNET have demonstrated their overall indolent course, illustrated by a tumor doubling time of ±12 years at long-term follow-up in a Dutch national cohort study (204). However, a very small number of lesions showed sudden aggressive tumor growth. Unfortunately, no prognostic factors for tumor growth have been identified to date.

 

The vast majority of MEN1-related bpNETs are well-differentiated NETs (typical and atypical carcinoids); only five cases of poorly differentiated neuroendocrine carcinomas have been identified in MEN1 patients until now, all in the French Groupe d’étude des Tumeurs Endocrines (GTE) cohort (206). Considering the large cohort size (n=1023 MEN1 patients), long-term follow-up, high frequency of smokers and lack of molecular analyses confirming a causal relationship with the MEN1 syndrome, a sporadic coincidental occurrence of neuroendocrine carcinomas in MEN1 patients might also be a possible explanation for the manifestation of these carcinomas in this particular study. The overall benign histopathological characteristics of MEN1-related bpNET may explain their usually good prognosis: large cohort studies have shown that bpNETs do not significantly affect survival in MEN1 patients (204, 206), although a few (eight) aggressive cases with fatal outcome have been described (206, 207). A recent comparison between patients with MEN1-related and sporadic bpNET with comparable histopathological features showed a significantly higher disease-specific mortality in sporadic bpNET, however this has not yet been confirmed in other cohorts (208).

 

Data from the largest cohort of histologically proven bpNETs in MEN1 patients (n=51) suggested that patients with distant metastasis at diagnosis and non-operated patients had a significantly worse survival (206). Additionally, females, patients with a typical carcinoid (compared to atypical carcinoid) and those without lymph node involvement tended to have a better survival (p=0.07, p=0.08 and p=0.08, respectively (206). However, the most recent Dutch cohort study could not find any prognostic factors (204).

 

Surgical resection is considered the first treatment of choice, and should be done as lung-sparing as possible, including considering endobronchial resection if feasible (2, 203). Given their usually indolent course, watch-and-wait policy may be considered in patients with small, non-central, slow-growing lesions (203). Parallel to treatment regimens in sporadic bpNET, additional radiotherapy and/or chemotherapy could be used in case of persistent or metastasized disease, although data on the effect of these regimens in aggressive MEN1-related bpNET is very limited.

 

Surveillance

 

Current clinical guidelines recommend thoracic imaging (CT or MRI) every 1-2 years for detection of thymic and bronchopulmonary NETs (2). The optimal imaging modality still remains to be elucidated, although CT scans are used in the far majority of cases. A direct comparison between MRI and CT scans among MEN1 patients is lacking, and the role of nuclear imaging in screening programs for thoracic NETs has to be determined yet (209-212). Furthermore, the frequency of periodic surveillance is subject of debate; on one hand, the overall indolent course of bpNET and rareness of thNET might argue for less frequent screening – thereby diminishing radiation exposure, physical and psychological distress for patients, and health care costs –, but the lack of predictors for (sudden) aggressive tumor growth in bpNET and the aggressive nature of thNET plead for frequent thoracic imaging in order to enable timely intervention if necessary. Therefore, treating physicians should inform their patients about the benefits and disadvantages of a strict surveillance program, in order to come to a personalized surveillance strategy based on shared decision-making. 

 

ADRENAL TUMORS

 

Adrenal involvement (Figure 1) is frequently seen in patients with MEN1 and considered to be part of the syndrome though not one of the cardinal manifestations. Mice with heterozygous inactivation of the Men1 gene develop adrenocortical lesions to a greater proportion than Men1 wild-type controls (213, 214) and the adrenal tumors show loss of heterogeneity (LOH) and loss of menin staining. In humans with MEN1, LOH is rarely seen in benign adrenocortical tumors (215-217). It has been hypothesized that the development of adrenal tumors in MEN1 might be related to PanNETs and hyperinsulinemia, because in some cohorts an association was seen between the occurrence of PanNETs, hyperinsulinemia, and adrenal lesions. However, in the largest series to date, no difference was found in the prevalence of main MEN1 manifestations between those with and without adrenal lesions (217).

 

In retrospective cohorts on adrenal involvement in MEN1 the reported prevalence greatly differs from 20-73% (216-225). Prevalence in part differs by the way adrenal lesions are defined (i.e. also including hyperplasia) and the manner of diagnosis, with prevalence being the highest (73%) in an EUS study (n=49) including all adrenal lesions from ‘plump’ adrenals to adenomas (221). In the series (n=27) with the second-highest prevalence (63%) all CT scans were re-read with the purpose of classifying adrenal lesions and every adrenocortical lesion >5 mm was considered a nodule (225). The largest series to date from the French GTE (n=715) has the lowest prevalence of 20.4% (217). Adrenal lesions are rarely the reason for an MEN1 diagnosis or the first manifestation of the disease, and are most frequently diagnosed asymptomatically by screening/surveillance imaging during follow-up or at the time of initial comprehensive imaging after the diagnosis of MEN1 is made (217, 223). Mean age of diagnosis is usually in the fifth decade, but ranges vary widely (217, 223, 224).

 

The French GTE series compared MEN1-related adrenal lesions to a cohort of sporadic incidentalomas (n=144) and found that adrenal lesions in patients with MEN1 were diagnosed at a younger age and were similar in size and in prevalence of bilateral lesions (217).

 

Benign Adrenocortical Tumors

 

Most MEN1-related adrenal lesions are benign adrenocortical lesions and include hyperplasia, (macro)nodular hyperplasia, and adenomas. Bilateral lesions are frequently seen, but again prevalence reported varies widely from 12.5% to >50% in different series( 216-219, 221-224). Most adrenocortical lesions in MEN1 are non-functioning and generally stable over the course of follow-up (216-224). In a minority of the cases ACTH-independent hypercortisolism, autonomous cortisol secretion, or primary hyperaldosteronism are seen (216-219, 221-224). Interestingly, in the French series, when comparing MEN1-related adrenal lesions with adrenal incidentalomas, functional tumors were more common (15% vs 6.9%), especially primary hyperaldosteronism and ACTH-independent hypercortisolism (217). Pheochromocytomas on the other hand were more common among sporadic incidentalomas.

 

Adrenocortical Carcinoma

 

Adrenocortical carcinoma (ACC) is a rare occurrence among patients with MEN1, with a 2019 review of literature identifying 19 published cases (226). In the Swedish cohort, one patient had an ACC and in the tumor LOH at the MEN1 locus was seen (216, 222). Three papers reporting from the same German institutions reported two, four and one case of ACC respectively, but some of these might represent the same patients (220, 224, 227). In the large French series, eight patients with 10 ACCs were reported, which was 5% of patients with an adrenal lesion, but 13.8% of those with an adrenal tumor (>10mm) (217). ACC prevalence was also significantly higher than in the sporadic adrenal incidentaloma cohort (217). There are also several case reports describing patients with MEN1-related ACC (226, 228-233). It is important to mention that there are several cases reported were the ACC developed from an initially observed relatively small adrenocortical tumor, although when reported these lesions did not have Houndsfield Units (HU) ≤ 10. In the French cohort one patient had two nodules (8 and 13 mm, 38 HU) in one adrenal, which grew to 29 and 30 mm after 5 years, which turned out to be ACCs. Another patient had a 25 mm calcified lesion (40 HU) which grew to 40 mm in 4 years(217). One case report describes a female patient with a 2 cm left adrenal tumor which grew to 3 cm in 7 years (advised to undergo surgery but refused) and then to 4 cm in 4 years, after which the tumor was removed and turned out to be an ACC (233). In other cases more rapidly progressing tumors are described (220, 231). When ACCs are functioning, they are mostly cortisol producing or sex-steroid producing. In sporadic ACC,MEN1 is considered one of the driver genes (234).

 

Pheochromocytoma

 

Pheochromocytoma is one of the hallmark conditions of Multiple Endocrine Neoplasia type 2 (MEN2), caused by germline mutations in the RET oncogene. In MEN1, the occurrence of pheochromocytoma is rare, with a 2020 case report and review of literature describing 20 published cases (235). The authors identified LOH at the MEN1 locus in the resected pheochromocytoma of the patient they report (235), and in another published series, two resected pheochromocytomas from patients with MEN1 were examined and LOH at the MEN1 locus was found in both, with one having absent menin staining and one weak menin staining (236).

 

Screening, Treatment and Follow-up

 

In patients with MEN1, minimal recommended screening for adrenal lesions as per the current guidelines, is abdominal imaging with CT or MRI every 3 years for those without adrenal lesions (2). Since abdominal imaging is also performed to screen for and/or surveille pancreatic lesions this can often be combined. However, care should be taken that the adrenals are properly imaged in the right phase to not only judge their size but also, should a lesion be present, to judge its characteristics. The preponderance to develop adrenal lesions should be mentioned in the clinical information to the radiologist and the images should be read by a radiologist experienced in adrenal imaging.

 

If an adrenal lesion is identified hormonal screening is recommended if patients are symptomatic or lesions are >1 cm (2). The current guidelines recommend that screening be focused on hyperaldosteronism and hypercortisolism (2), but given that pheochromocytomas do occur in MEN1 as described above and the consequence of missing the diagnosis can be serious, it is prudent to screen for the presence of a pheochromocytoma by either plasma free (nor)metanephrines or urinary fractionated (nor)metanephrines.

 

Indications for surgical resection parallel those of adrenal incidentalomas, being clinically significant hormone excess and/or concerns about malignancy either due to atypical characteristics on imaging, size (>4 cm), or significant growth over a 6-month period, which in the adrenal incidentaloma guidelines is suggested as increase in 20% (in addition to at least 5mm increase in actual size) (2, 237). Given the reports of ACCs in MEN1 arising from initially small lesions, some authors recommend to use a size cut-off of 3 cm in MEN1 (220).

 

If there is no indication for surgery, surveillance imaging is indicated, initially after 6 months. In the absence of a surgical indication at this point, frequency of further imaging follow-up should be determined individually and discussed by the multidisciplinary team. Unlike in sporadic adrenal incidentalomas, surveillance cannot be ended, given that multiple adrenal lesions can arise. There may be indications during follow-up to repeat initially negative hormonal screening, such as the development of symptoms or a new adrenal lesion.

 

CUTANEOUS LESIONS

 

Facial angiofibromas and collagenomas are the main skin lesions in MEN1 (Figure 1) (238, 239). Frequencies of 64% for angiofibromas and 62% for collagenomas have been described. Multiple angiofibromas and collagenomas are present in 77–81% of the MEN1 patients (238). Primarily angiofibromas are seen in patients with MEN1 (238, 240). An odds ratio of 6.6 (95% CI, 1.09–40.43) for cutaneous lesions in MEN1 in 29 patients with MEN1 in comparison with their non- affected family members is described (240).

 

These findings are further supported by the allelic loss of the MEN1 gene in six angiofibromas, three collagenomas, and one lipoma, suggesting that loss of function of the wild-type MEN1 gene product plays a role in the development of these skin lesions in patients with MEN1(241). Melanomas and other skin lesions are also described in the MEN1 population, but not with significant prevalences.

 

Lipomas (Figure 1) are reported in 17-34% of patients with MEN1 (238-240). Loss of heterozygosity of the MEN1 gene is described in MEN1-related lipomas (97, 241, 242) and may also play a role in sporadic lipomas (242). Menin seems to be an important factor for adipogenesis and contributes to lipoma development (243, 244).

 

A case of a novel MEN1 gene mutation with a recurrent sarcoma addresses the need for cautiousness of (atypical) skin lesions in patients with MEN1 (245).

 

BREAST CANCER AND MEN1

 

A higher incidence of breast cancer (Figure 1) was found in four independent MEN1 cohorts in the Netherlands, France, Tasmania and the United States. In the Dutch cohort a relative risk of 2.83 was found, which was significantly higher than in the general Dutch population (3). The median age for breast cancer was 45 years, which is approximately 15 years younger than the general Dutch population. The increased risk for breast cancer for MEN1 carriers was not associated with other breast cancer risk factors or a familial breast cancer risk. Considering the younger age of breast cancer occurrence and an earlier age of breast cancer, surveillance should be considered. Breast cancer surveillance from the age of 40 is initiated in the Dutch MEN1 cohort (4). After the latter publication, several cases of early breast cancer in MEN1 patients were reported (246-249).

 

These epidemiological findings are supported by basic research. Loss-of-function Men1 mouse models have shown an increased incidence of both in situ and invasive mammary cancer (250). Menin, the tumor suppressor protein encoded by MEN1, is co-localized with the estrogen receptor (ER) alfa in breast cancer cells. In this manner, menin functions as a direct activator of Erα (251). In sporadic ER-positive breast cancer, menin seems to have a proliferative role, which is in contrast with breast cancer in MEN1 carriers, in whom LOH of the MEN1 gene could be found (3, 252). Recent studies showed that reduced menin staining is associated with ER-negative breast cancer and in ER-positive breast cancer with larger tumors, higher grade tumor, and luminal subtypes tumors. Providing further evidence that there is an important role of menin in ERα regulation and the breast cancer formation (253).

 

PSYCHOSOCIAL ASPECTS

 

Recently there has been more interest in the psychosocial wellbeing of patients with MEN1 (254-259).The first study was published in 2003, which showed that psychosocial outcomes such as anxiety, depression, intrusion, and avoidance are not altered by the hospital or home setting. A higher burden of disease led to more depression. Compared to the population-based norm values, patients with MEN1 scored lower for General Health and Social Functioning according to the SF-36 (260).

 

Postoperatively, quality of life (QOL) scores did not differ after pancreaticoduodenal surgery in MEN1 patients in comparison with the general population. Financial difficulties caused by the treatment were significantly worse in MEN1 patients (261). Financial burden seems to be associated with having MEN1. The degree of financial burden has a linear relationship with worse health-related QOL. Patients were three-times more likely to be unemployed in comparison with the US population (256).

 

The largest QOL-related study showed that employment status was the most consistent predictor for QOL. This is in line with the former studies. The health-related QOL according to the SF-36 was significantly lower for patients with MEN1 on all subscales except for the physical functioning scale. Patients who are aware of their PA and PanNET have worse QOL scores in comparison with patients who are not aware of having these tumors(259). The degree of fear of disease recurrence is high in patients with MEN1. This fear is negatively associated with health-related QOL and is higher in patients who consider themselves at high risk for developing a MEN1-related tumor. More MEN1-related manifestations lead to more fear of disease occurrence (258). In comparison with other chronic diseases MEN1 scores worse regarding anxiety, depression and fatigue (257).

 

QOL did not overly differ from the general population in the Italian cohort (255) and patients were more optimistic than in the Swedish cohort (260). This could be due to cultural differences, population selection, and awareness of the disease and its implications.

The high response rate in the MEN1 population illustrates the motivation of patients to participate in research and care about their wellbeing (259, 260).

 

CONCLUSION

 

In conclusion, in the past decades there have been great advances in the understanding of the natural course of MEN1-related tumors, which has had direct consequences on clinical care. In the coming decade one of the main research objectives will be the identification of individual predictors of disease course which can guide personalized treatment and surveillance. Increasing international collaborations will enable prospective studies. Given the complexity of the disease, it is strongly advised that patients, whenever possible, be followed and treated in centers of expertise. If this is not feasible, consultation with a center of expertise should be considered.

 

REFERENCES

 

  1. Lloyd, R.V., et al., WHO Classification of Tumours of Endocrine Organs. 4th ed. 2017, Lyon: IARC.
  2. Thakker, R.V., et al., Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). The Journal of clinical endocrinology & metabolism, 2012. 97(9): p. 2990-3011.
  3. Dreijerink, K.M.a., et al., Breast-Cancer Predisposition in Multiple Endocrine Neoplasia Type 1. New England Journal of Medicine, 2014. 371(6): p. 583-584.
  4. van Leeuwaarde, R.S., et al., MEN1-Dependent Breast Cancer: Indication for Early Screening? Results From the Dutch MEN1 Study Group. J Clin Endocrinol Metab, 2017. 102(6): p. 2083-2090.
  5. Brandi, M.L., et al., Guidelines for diagnosis and therapy of MEN type 1 and type 2. The Journal of clinical endocrinology & metabolism, 2001. 86(12): p. 5658-71.
  6. Chandrasekharappa, S.C., et al., Positional Cloning of the Gene for Multiple Endocrine Neoplasia-Type 1 Science, 1997. 276: p. 404-407.
  7. Lemmens, I., et al., Identification of the multiple endocrine neoplasia type 1 (MEN1) gene. Human molecular genetics, 1997. 6: p. 1177-83.
  8. Lemos, M.C. and R.V. Thakker, Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Human mutation, 2008. 29(1): p. 22-32.
  9. Concolino, P., A. Costella, and E. Capoluongo, Multiple endocrine neoplasia type 1 (MEN1): An update of 208 new germline variants reported in the last nine years. Cancer Genet, 2016. 209(1-2): p. 36-41.
  10. Kooblall, K.G., et al., Multiple Endocrine Neoplasia Type 1 (MEN1) 5'UTR Deletion, in MEN1 Family, Decreases Menin Expression. J Bone Miner Res, 2021. 36(1): p. 100-109.
  11. Agarwal, S.K., The future: genetics advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer, 2017. 24(10): p. T119-t134.
  12. Romanet, P., et al., Proposition of adjustments to the ACMG-AMP framework for the interpretation of MEN1 missense variants. Hum Mutat, 2019. 40(6): p. 661-674.
  13. Thevenon, J., et al., Higher risk of death among MEN1 patients with mutations in the JunD interacting domain: a Groupe d'etude des Tumeurs Endocrines (GTE) cohort study. Human molecular genetics, 2013. 22: p. 1940-1948.
  14. Bartsch, D.K., et al., Higher risk of aggressive pancreatic neuroendocrine tumors in MEN1 patients with MEN1 mutations affecting the CHES1 interacting MENIN domain. J Clin Endocrinol Metab, 2014. 99(11): p. E2387-91.
  15. Klein Haneveld, M.J., et al., Initiating pancreatic neuroendocrine tumour (pNET) screening in young MEN1 patients: results from the DutchMEN Study Group. J Clin Endocrinol Metab, 2021.
  16. van den Broek, M.F.M., et al., Clues For Genetic Anticipation In Multiple Endocrine Neoplasia Type 1. J Clin Endocrinol Metab, 2020. 105(7).
  17. Beijers, H., et al., Germline and somatic mosaicism in a family with multiple endocrine neoplasia type 1 (MEN1) syndrome. Eur J Endocrinol, 2019. 180(2): p. K15-k19.
  18. Dreijerink, K.M.A., H.T.M. Timmers, and M. Brown, Twenty years of menin: emerging opportunities for restoration of transcriptional regulation in MEN1. Endocr Relat Cancer, 2017. 24(10): p. T135-t145.
  19. Canaff, L., et al., Impaired transforming growth factor-β (TGF-β) transcriptional activity and cell proliferation control of a menin in-frame deletion mutant associated with multiple endocrine neoplasia type 1 (MEN1). The Journal of biological chemistry, 2012. 287: p. 8584-97.
  20. Iyer, S. and S.K. Agarwal, Epigenetic regulation in the tumorigenesis of MEN1-associated endocrine cell types. J Mol Endocrinol, 2018. 61(1): p. R13-r24.
  21. Walls, G.V., et al., MEN1 gene replacement therapy reduces proliferation rates in a mouse model of pituitary adenomas. Cancer research, 2012. 72: p. 5060-8.
  22. Pieterman, C.R., et al., Care for patients with multiple endocrine neoplasia type 1: the current evidence base. Fam Cancer, 2011. 10(1): p. 157-71.
  23. van Leeuwaarde, R.S., et al., The future: medical advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer, 2017. 24(10): p. T179-t193.
  24. de Laat, J.M., et al., MEN1 redefined, a clinical comparison of mutation-positive and mutation-negative patients. BMC Med, 2016. 14(1): p. 182.
  25. Pieterman, C.R.C., et al., Understanding the clinical course of genotype-negative MEN1 patients can inform management strategies. Surgery, 2020.
  26. Goudet, P., et al., Risk factors and causes of death in MEN1 disease. A GTE (Groupe d'Etude des Tumeurs Endocrines) cohort study among 758 patients. World journal of surgery, 2010. 34(2): p. 249-55.
  27. Geerdink, E.A., R.B. Van der Luijt, and C.J. Lips, Do patients with multiple endocrine neoplasia syndrome type 1 benefit from periodical screening? European journal of endocrinology, 2003. 149(6): p. 577-82.
  28. de Laat, J.M., R.S. van Leeuwaarde, and G.D. Valk, The Importance of an Early and Accurate MEN1 Diagnosis. Front Endocrinol (Lausanne), 2018. 9: p. 533.
  29. de Laat, J.M., et al., Predicting the risk of multiple endocrine neoplasia type 1 for patients with commonly occurring endocrine tumors. European journal of endocrinology, 2012. 167(2): p. 181-7.
  30. van Leeuwaarde, R.S., et al., Impact of delay in diagnosis in outcomes in MEN1: results from the Dutch MEN1 study group. J Clin Endocrinol Metab, 2016: p. jc20153766.
  31. Alrezk, R., F. Hannah-Shmouni, and C.A. Stratakis, MEN4 and CDKN1B mutations: the latest of the MEN syndromes. Endocr Relat Cancer, 2017. 24(10): p. T195-t208.
  32. Frederiksen, A., et al., Clinical Features of Multiple Endocrine Neoplasia Type 4: Novel Pathogenic Variant and Review of Published Cases. J Clin Endocrinol Metab, 2019. 104(9): p. 3637-3646.
  33. Georgitsi, M., et al., Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. The Journal of clinical endocrinology and metabolism, 2007. 92: p. 3321-3325.
  34. van der Tuin, K., et al., CDC73-Related Disorders: Clinical Manifestations and Case Detection in Primary Hyperparathyroidism. J Clin Endocrinol Metab, 2017. 102(12): p. 4534-4540.
  35. van den Broek, M.F.M., et al., Clinical Relevance of Genetic Analysis in Patients With Pituitary Adenomas: A Systematic Review. Front Endocrinol (Lausanne), 2019. 10: p. 837.
  36. Stratakis, C.A., et al., Pituitary macroadenoma in a 5-year-old: an early expression of multiple endocrine neoplasia type 1. J Clin Endocrinol Metab, 2000. 85(12): p. 4776-80.
  37. Manoharan, J., et al., Is Routine Screening of Young Asymptomatic MEN1 Patients Necessary? World J Surg, 2017. 41(8): p. 2026-2032.
  38. Romanet, P., et al., UMD-MEN1 Database: An Overview of the 370 MEN1 Variants Present in 1676 Patients From the French Population. J Clin Endocrinol Metab, 2019. 104(3): p. 753-764.
  39. Goudet, P., et al., Hyperparathyroidism in multiple endocrine neoplasia type I: surgical trends and results of a 256-patient series from Groupe D'etude des Néoplasies Endocriniennes Multiples Study Group. World J Surg, 2001. 25(7): p. 886-90.
  40. Pieterman, C.R., et al., Multiple endocrine neoplasia type 1 (MEN1): its manifestations and effect of genetic screening on clinical outcome. Clinical endocrinology, 2009. 70(4): p. 575-81.
  41. Marini, F., F. Giusti, and M.L. Brandi, Multiple endocrine neoplasia type 1: extensive analysis of a large database of Florentine patients. Orphanet J Rare Dis, 2018. 13(1): p. 205.
  42. Giusti, F., et al., Multiple endocrine neoplasia syndrome type 1: institution, management, and data analysis of a nationwide multicenter patient database. Endocrine, 2017. 58(2): p. 349-359.
  43. Sakurai, A., et al., Multiple endocrine neoplasia type 1 in Japan: establishment and analysis of a multicentre database. Clinical endocrinology, 2012. 76(4): p. 533-9.
  44. Soczomski, P., et al., Multiple endocrine neoplasia type 1 in Poland: a two-centre experience. Endokrynol Pol, 2019. 70(5): p. 385-391.
  45. Waldmann, J., et al., Screening of patients with multiple endocrine neoplasia type 1 (MEN-1): a critical analysis of its value. World journal of surgery, 2009. 33(6): p. 1208-18.
  46. Herath, M., et al., Paediatric and young adult manifestations and outcomes of multiple endocrine neoplasia type 1. Clin Endocrinol (Oxf), 2019. 91(5): p. 633-638.
  47. Goudet, P., et al., MEN1 disease occurring before 21 years old: a 160-patient cohort study from the Groupe d'etude des Tumeurs Endocrines. J Clin Endocrinol Metab, 2015. 100(4): p. 1568-77.
  48. Shariq, O.A., et al., Multiple endocrine neoplasia type 1 in children and adolescents: Clinical features and treatment outcomes. Surgery, 2021.
  49. Vannucci, L., et al., MEN1 in children and adolescents: Data from patients of a regional referral center for hereditary endocrine tumors. Endocrine, 2018. 59(2): p. 438-448.
  50. Marx, S.J. and D.M. Lourenço, Jr., Questions and Controversies About Parathyroid Pathophysiology in Children With Multiple Endocrine Neoplasia Type 1. Front Endocrinol (Lausanne), 2018. 9: p. 359.
  51. Doherty, G.M., T.C. Lairmore, and M.K. DeBenedetti, Multiple endocrine neoplasia type 1 parathyroid adenoma development over time. World J Surg, 2004. 28(11): p. 1139-42.
  52. d'Alessandro, A.F., et al., Supernumerary parathyroid glands in hyperparathyroidism associated with multiple endocrine neoplasia type 1. Rev Assoc Med Bras (1992), 2012. 58(3): p. 323-7.
  53. Yeh, M.W., et al., Incidence and prevalence of primary hyperparathyroidism in a racially mixed population. J Clin Endocrinol Metab, 2013. 98(3): p. 1122-9.
  54. Press, D.M., et al., The prevalence of undiagnosed and unrecognized primary hyperparathyroidism: a population-based analysis from the electronic medical record. Surgery, 2013. 154(6): p. 1232-7; discussion 1237-8.
  55. Eller-Vainicher, C., et al., Sporadic and MEN1-related primary hyperparathyroidism: differences in clinical expression and severity. J Bone Miner Res, 2009. 24(8): p. 1404-10.
  56. Twigt, B.A., et al., Differences between sporadic and MEN related primary hyperparathyroidism; clinical expression, preoperative workup, operative strategy and follow-up. Orphanet J Rare Dis, 2013. 8: p. 50.
  57. Wilhelm, S.M., et al., The American Association of Endocrine Surgeons Guidelines for Definitive Management of Primary Hyperparathyroidism. JAMA Surg, 2016. 151(10): p. 959-968.
  58. Iacobone, M., et al., Hereditary hyperparathyroidism--a consensus report of the European Society of Endocrine Surgeons (ESES). Langenbecks Arch Surg, 2015. 400(8): p. 867-86.
  59. Silva, A.M., et al., Operative intervention for primary hyperparathyroidism offers greater bone recovery in patients with sporadic disease than in those with multiple endocrine neoplasia type 1-related hyperparathyroidism. Surgery, 2017. 161(1): p. 107-115.
  60. Wang, W., et al., Impaired geometry, volumetric density, and microstructure of cortical and trabecular bone assessed by HR-pQCT in both sporadic and MEN1-related primary hyperparathyroidism. Osteoporos Int, 2020. 31(1): p. 165-173.
  61. Kong, J., et al., Clinical and Genetic Analysis of Multiple Endocrine Neoplasia Type 1-Related Primary Hyperparathyroidism in Chinese. PLoS One, 2016. 11(11): p. e0166634.
  62. Lourenço, D.M., Jr., et al., Early-onset, progressive, frequent, extensive, and severe bone mineral and renal complications in multiple endocrine neoplasia type 1-associated primary hyperparathyroidism. J Bone Miner Res, 2010. 25(11): p. 2382-91.
  63. Burgess, J.R., et al., Osteoporosis in multiple endocrine neoplasia type 1: severity, clinical significance, relationship to primary hyperparathyroidism, and response to parathyroidectomy. Arch Surg, 1999. 134(10): p. 1119-23.
  64. Lourenco, D.M., Jr., et al., Multiple endocrine neoplasia type 1 in Brazil: MEN1 founding mutation, clinical features, and bone mineral density profile. European journal of endocrinology, 2008. 159(3): p. 259-74.
  65. Coutinho, F.L., et al., Bone mineral density analysis in patients with primary hyperparathyroidism associated with multiple endocrine neoplasia type 1 after total parathyroidectomy. Clin Endocrinol (Oxf), 2010. 72(4): p. 462-8.
  66. Kann, P.H., et al., Peripheral bone mineral density in correlation to disease-related predisposing conditions in patients with multiple endocrine neoplasia type 1. J Endocrinol Invest, 2012. 35(6): p. 573-9.
  67. Green, P., et al., High prevalence of chronic kidney disease in patients with multiple endocrine neoplasia type 1 and improved kidney function after parathyroidectomy. Surgery, 2019. 165(1): p. 124-128.
  68. Bilezikian, J.P., et al., Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the Fourth International Workshop. J Clin Endocrinol Metab, 2014. 99(10): p. 3561-9.
  69. Norton, J.A., et al., Prospective study of surgery for primary hyperparathyroidism (HPT) in multiple endocrine neoplasia-type 1 and Zollinger-Ellison syndrome: long-term outcome of a more virulent form of HPT. Ann Surg, 2008. 247(3): p. 501-10.
  70. Hackeng, W.M., et al., A Parathyroid-Gut Axis: Hypercalcemia and the Pathogenesis of Gastrinoma in Multiple Endocrine Neoplasia 1. Mol Cancer Res, 2021. 19(6): p. 946-949.
  71. Nobecourt, P.F., et al., Intraoperative Decision-Making and Technical Aspects of Parathyroidectomy in Young Patients With MEN1 Related Hyperparathyroidism. Front Endocrinol (Lausanne), 2018. 9: p. 618.
  72. Nastos, C., et al., Optimal extent of initial parathyroid resection in patients with multiple endocrine neoplasia syndrome type 1: A meta-analysis. Surgery, 2021. 169(2): p. 302-310.
  73. Schreinemakers, J.M., et al., The optimal surgical treatment for primary hyperparathyroidism in MEN1 patients: a systematic review. World J Surg, 2011. 35(9): p. 1993-2005.
  74. Manoharan, J., et al., Single gland excision for MEN1-associated primary hyperparathyroidism. Clin Endocrinol (Oxf), 2020. 92(1): p. 63-70.
  75. Nilubol, N., et al., Preoperative localizing studies for initial parathyroidectomy in MEN1 syndrome: is there any benefit? World J Surg, 2012. 36(6): p. 1368-74.
  76. Nilubol, N., et al., Utility of intraoperative parathyroid hormone monitoring in patients with multiple endocrine neoplasia type 1-associated primary hyperparathyroidism undergoing initial parathyroidectomy. World J Surg, 2013. 37(8): p. 1966-72.
  77. Kluijfhout, W.P., et al., Unilateral Clearance for Primary Hyperparathyroidism in Selected Patients with Multiple Endocrine Neoplasia Type 1. World J Surg, 2016. 40(12): p. 2964-2969.
  78. Montenegro, F.L.M., et al., Could the Less-Than Subtotal Parathyroidectomy Be an Option for Treating Young Patients With Multiple Endocrine Neoplasia Type 1-Related Hyperparathyroidism? Front Endocrinol (Lausanne), 2019. 10: p. 123.
  79. Choi, H.R., et al., Benefit of diverse surgical approach on short-term outcomes of MEN1-related hyperparathyroidism. Sci Rep, 2020. 10(1): p. 10634.
  80. Nilubol, N., et al., Limited Parathyroidectomy in Multiple Endocrine Neoplasia Type 1-Associated Primary Hyperparathyroidism: A Setup for Failure. Ann Surg Oncol, 2016. 23(2): p. 416-23.
  81. Keutgen, X.M., et al., Reoperative Surgery in Patients with Multiple Endocrine Neoplasia Type 1 Associated Primary Hyperparathyroidism. Ann Surg Oncol, 2016. 23(Suppl 5): p. 701-707.
  82. Bioletto, F., et al., Comparison of the diagnostic accuracy of 18F-Fluorocholine PET and 11C-Methionine PET for parathyroid localization in primary hyperparathyroidism: a systematic review and meta-analysis. Eur J Endocrinol, 2021. 185(1): p. 109-120.
  83. Hindié, E., et al., Primary Hyperparathyroidism: Defining the Appropriate Preoperative Imaging Algorithm. J Nucl Med, 2021. 62(Suppl 2): p. 3s-12s.
  84. Gauthé, M., et al., (18)F-fluorocholine PET/CT in MEN1 Patients with Primary Hyperparathyroidism. World J Surg, 2020. 44(11): p. 3761-3769.
  85. Fyrsten, E., et al., Long-Term Surveillance of Treated Hyperparathyroidism for Multiple Endocrine Neoplasia Type 1: Recurrence or Hypoparathyroidism? World J Surg, 2016. 40(3): p. 615-21.
  86. Pieterman, C.R., et al., Primary hyperparathyroidism in MEN1 patients: a cohort study with longterm follow-up on preferred surgical procedure and the relation with genotype. Ann Surg, 2012. 255(6): p. 1171-8.
  87. Filopanti, M., et al., MEN1-related hyperparathyroidism: response to cinacalcet and its relationship with the calcium-sensing receptor gene variant Arg990Gly. Eur J Endocrinol, 2012. 167(2): p. 157-64.
  88. Giusti, F., et al., Cinacalcet therapy in patients affected by primary hyperparathyroidism associated to Multiple Endocrine Neoplasia Syndrome type 1 (MEN1). Endocrine, 2016. 52(3): p. 495-506.
  89. Moyes, V.J., et al., Clinical Use of Cinacalcet in MEN1 Hyperparathyroidism. Int J Endocrinol, 2010. 2010: p. 906163.
  90. Geslot, A., et al., New therapies for patients with multiple endocrine neoplasia type 1. Ann Endocrinol (Paris), 2021. 82(2): p. 112-120.
  91. Singh Ospina, N., et al., Safety and efficacy of percutaneous parathyroid ethanol ablation in patients with recurrent primary hyperparathyroidism and multiple endocrine neoplasia type 1. J Clin Endocrinol Metab, 2015. 100(1): p. E87-90.
  92. Williams, M.D., et al., Pathology data set for reporting parathyroid carcinoma and atypical parathyroid neoplasm: recommendations from the International Collaboration on Cancer Reporting. Hum Pathol, 2021. 110: p. 73-82.
  93. Song, A., et al., Prevalence of Parathyroid Carcinoma and Atypical Parathyroid Neoplasms in 153 Patients With Multiple Endocrine Neoplasia Type 1: Case Series and Literature Review. Front Endocrinol (Lausanne), 2020. 11: p. 557050.
  94. Christakis, I., et al., Parathyroid carcinoma and atypical parathyroid neoplasms in MEN1 patients; A clinico-pathologic challenge. The MD Anderson case series and review of the literature. Int J Surg, 2016. 31: p. 10-6.
  95. Singh Ospina, N., et al., Prevalence of parathyroid carcinoma in 348 patients with multiple endocrine neoplasia type 1 - case report and review of the literature. Clin Endocrinol (Oxf), 2016. 84(2): p. 244-249.
  96. Erdheim, J., Zur normalen und pathologischen Histologie der Glandula thyreoidea, parathyreoidea, und Hypophysis. Beitr Pathol Anat, 1903. 33: p. 158-236.
  97. Dong, Q., et al., Loss of heterozygosity at 11q13: analysis of pituitary tumors, lung carcinoids, lipomas, and other uncommon tumors in subjects with familial multiple endocrine neoplasia type 1. The Journal of clinical endocrinology & metabolism, 1997. 82(5): p. 1416-20.
  98. Thakker, R.V., The molecular genetics of the multiple endocrine neoplasia syndromes. Clinical Endocrinology, 1993. 38(1): p. 1-14.
  99. Weil, R.J., et al., 11Q13 Allelic Loss in Pituitary Tumors in Patients With Multiple Endocrine Neoplasia Syndrome Type 1. Clinical Cancer Research, 1998. 4(7): p. 1673-1678.
  100. Yoshimoto, K., et al., Allele Loss on Chromosome 11 in a Pituitary Tumor from a Patient with Multiple Endocrine Neoplasia Type 1. 1991. p. 886-889.
  101. Thakker, R.V., et al., Association of somatotrophinomas with loss of alleles on chromosome 11 and with gsp mutations. J Clin Invest, 1993. 91(6): p. 2815-21.
  102. Boggild, M.D., et al., Molecular genetic studies of sporadic pituitary tumors. J Clin Endocrinol Metab, 1994. 78(2): p. 387-92.
  103. Asa, S.L., K. Somers, and S. Ezzat, The MEN-1 gene is rarely down-regulated in pituitary adenomas. J Clin Endocrinol Metab, 1998. 83(9): p. 3210-2.
  104. Farrell, W.E., et al., Sequence analysis and transcript expression of the MEN1 gene in sporadic pituitary tumours. Br J Cancer, 1999. 80(1-2): p. 44-50.
  105. Prezant, T.R., J. Levine, and S. Melmed, Molecular characterization of the men1 tumor suppressor gene in sporadic pituitary tumors. J Clin Endocrinol Metab, 1998. 83(4): p. 1388-91.
  106. Tanaka, C., et al., Analysis of loss of heterozygosity on chromosome 11 and infrequent inactivation of the MEN1 gene in sporadic pituitary adenomas. J Clin Endocrinol Metab, 1998. 83(8): p. 2631-4.
  107. Zhuang, Z., et al., Mutations of the MEN1 tumor suppressor gene in pituitary tumors. Cancer Res, 1997. 57(24): p. 5446-51.
  108. Ballard, H.S., B. Fame, and R.J. Hartsock, Familial multiple endocrine adenoma-peptic ulcer complex. Medicine (Baltimore), 1964. 43(0025-7974 (Print)): p. 481-516.
  109. Verges, B., et al., Pituitary disease in MEN type 1 (MEN1): Data from the France-Belgium MEN1 multicenter study. Journal of Clinical Endocrinology & Metabolism, 2002. 87(2): p. 457-465.
  110. Trouillas, J., et al., Pituitary tumors and hyperplasia in multiple endocrine neoplasia type 1 syndrome (MEN1): a case-control study in a series of 77 patients versus 2509 non-MEN1 patients. Am J Surg Pathol, 2008. 32(4): p. 534-43.
  111. de Laat, J.M., et al., Long-term natural course of pituitary tumors in patients with MEN1: results from the Dutch MEN1 study group (DMSG). J Clin.Endocrinol.Metab, 2015(1945-7197 (Electronic)): p. JC20152015-JC20152015.
  112. Wu, Y., et al., Pituitary adenomas in patients with multiple endocrine neoplasia type 1: a single-center experience in China. Pituitary, 2019. 22(2): p. 113-123.
  113. Cohen-Cohen, S., et al., Pituitary adenomas in the setting of multiple endocrine neoplasia type 1: A single-institution experience. Journal of Neurosurgery, 2021. 134(4): p. 1132-1138.
  114. Bras, M.L., et al., Pituitary adenoma in patients with multiple endocrine neoplasia type 1 – a cohort study. 2021: p. 1-34.
  115. Makri, A., et al., Children with MEN1 gene mutations may present first (and at a young age) with Cushing disease. Clin Endocrinol (Oxf), 2018. 89(4): p. 437-443.
  116. Melmed, S., et al., Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2011. 96(2): p. 273-88.
  117. Nieman, L.K., et al., The diagnosis of Cushing's syndrome: An endocrine society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism, 2008. 93(5): p. 1526-1540.
  118. Katznelson, L., et al., Acromegaly: An endocrine society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism, 2014. 99(11): p. 3933-3951.
  119. Incandela, F., et al., Malignancy course of pituitary adenoma in MEN1 syndrome: Clinical-Neuroradiological signs. Eur J Radiol Open, 2020. 7: p. 100242.
  120. Philippon, M., et al., Long-term control of a MEN1 prolactin secreting pituitary carcinoma after temozolomide treatment. Ann Endocrinol (Paris), 2012. 73(3): p. 225-9.
  121. Benito, M., et al., Gonadotroph tumor associated with multiple endocrine neoplasia type 1. J Clin Endocrinol Metab, 2005. 90(1): p. 570-4.
  122. Scheithauer, B.W., et al., Multiple endocrine neoplasia type 1-associated thyrotropin-producing pituitary carcinoma: report of a probable de novo example. Hum Pathol, 2009. 40(2): p. 270-8.
  123. Morokuma, H., et al., A case of nonfunctioning pituitary carcinoma that responded to temozolomide treatment. Case Rep Endocrinol, 2012. 2012: p. 645914.
  124. Triponez, F., et al., Epidemiology data on 108 MEN 1 patients from the GTE with isolated nonfunctioning tumors of the pancreas. Annals of surgery, 2006. 243(2): p. 265-72.
  125. Vinault, S., et al., Metastatic Potential and Survival of Duodenal and Pancreatic Tumors in Multiple Endocrine Neoplasia Type 1: A GTE and AFCE Cohort Study (Groupe d'etude des Tumeurs Endocrines and Association Francophone de Chirurgie Endocrinienne). Ann Surg, 2018.
  126. Pipeleers-Marichal, M., et al., Gastrinomas in the duodenums of patients with multiple endocrine neoplasia type 1 and the Zollinger-Ellison syndrome. The New England journal of medicine, 1990. 322(11): p. 723-7.
  127. Levy-Bohbot, N., et al., Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterologie clinique et biologique, 2004. 28(11): p. 1075-81.
  128. Borson-Chazot, F., et al., Acromegaly induced by ectopic secretion of GHRH: a review 30 years after GHRH discovery. Ann Endocrinol (Paris), 2012. 73(6): p. 497-502.
  129. Milanesi, A., et al., Concurrent primary hyperparathyroidism and humoral hypercalcemia of malignancy in a patient with multiple endocrine neoplasia type 1. Pancreas, 2011. 40(4): p. 634-7.
  130. Klöppel, G., et al., Hyperplasia to neoplasia sequence of duodenal and pancreatic neuroendocrine diseases and pseudohyperplasia of the PP-cells in the pancreas. Endocr Pathol, 2014. 25(2): p. 181-5.
  131. Anlauf, M., et al., Allelic deletion of the MEN1 gene in duodenal gastrin and somatostatin cell neoplasms and their precursor lesions. Gut, 2007. 56(5): p. 637-44.
  132. van Beek, D.J., et al., Surgery for multiple endocrine neoplasia type 1-related insulinoma: long-term outcomes in a large international cohort. Br J Surg, 2020. 107(11): p. 1489-1499.
  133. Gibril, F., et al., Multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome: a prospective study of 107 cases and comparison with 1009 cases from the literature. Medicine, 2004. 83(1): p. 43-83.
  134. Ito, T., et al., Causes of death and prognostic factors in multiple endocrine neoplasia type 1: a prospective study: comparison of 106 MEN1/Zollinger-Ellison syndrome patients with 1613 literature MEN1 patients with or without pancreatic endocrine tumors. Medicine, 2013. 92(3): p. 135-81.
  135. van Beek, D.J., et al., Prognostic factors and survival in MEN1 patients with gastrinomas: Results from the DutchMEN study group (DMSG). J Surg Oncol, 2019. 120(6): p. 966-975.
  136. Jensen, R.T. and T. Ito, Gastrinoma, in Endotext, K.R. Feingold, et al., Editors. 2000, MDText.com, Inc. Copyright © 2000-2021, MDText.com, Inc.: South Dartmouth (MA).
  137. Hampel, H., et al., A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med, 2015. 17(1): p. 70-87.
  138. Gibril, F., et al., Prospective study of the natural history of gastrinoma in patients with MEN1: definition of an aggressive and a nonaggressive form. The Journal of clinical endocrinology & metabolism, 2001. 86(11): p. 5282-93.
  139. Conemans, E.B., et al., Prognostic factors for survival of MEN1 patients with duodenopancreatic tumors metastatic to the liver: results from the DMSG. Endocr Pract, 2017. 23(6): p. 641-648.
  140. Giudici, F., et al., Natural History of MEN1 GEP-NET: Single-Center Experience After a Long Follow-Up. World J Surg, 2017. 41(9): p. 2312-2323.
  141. Christakis, I., et al., Genotype-phenotype pancreatic neuroendocrine tumor relationship in multiple endocrine neoplasia type 1 patients: A 23-year experience at a single institution. Surgery, 2018. 163(1): p. 212-217.
  142. Nagtegaal, I.D., et al., The 2019 WHO classification of tumours of the digestive system. Histopathology, 2020. 76(2): p. 182-188.
  143. van Treijen, M.J.C., et al., Diagnosing Nonfunctional Pancreatic NETs in MEN1: The Evidence Base. J Endocr Soc, 2018. 2(9): p. 1067-1088.
  144. Pieterman, C.R.C., et al., HEREDITARY ENDOCRINE TUMOURS: CURRENT STATE-OF-THE-ART AND RESEARCH OPPORTUNITIES: MEN1-related pancreatic NETs: identification of unmet clinical needs and future directives. Endocr Relat Cancer, 2020. 27(8): p. T9-t25.
  145. Cuthbertson, D.J., et al., The Impact of (68)Gallium DOTA PET/CT in Managing Patients With Sporadic and Familial Pancreatic Neuroendocrine Tumours. Front Endocrinol (Lausanne), 2021. 12: p. 654975.
  146. Sadowski, S.M., et al., Prognostic factors for the outcome of nonfunctioning pancreatic neuroendocrine tumors in MEN1: a systematic review of literature. Endocr Relat Cancer, 2020. 27(6): p. R145-r161.
  147. Partelli, S., et al., Active Surveillance versus Surgery of Nonfunctioning Pancreatic Neuroendocrine Neoplasms </=2 cm in MEN1 Patients. Neuroendocrinology, 2016. 103(6): p. 779-786.
  148. Nell, S., et al., Management of MEN1 Related Nonfunctioning Pancreatic NETs: A Shifting Paradigm: Results From the DutchMEN1 Study Group. Ann Surg, 2018. 267(6): p. 1155-1160.
  149. Triponez, F., et al., Is surgery beneficial for MEN1 patients with small (< or = 2 cm), nonfunctioning pancreaticoduodenal endocrine tumor? An analysis of 65 patients from the GTE. World journal of surgery, 2006. 30(5): p. 654-62; discussion 663-4.
  150. Pieterman, C.R.C., et al., Long-Term Natural Course of Small Nonfunctional Pancreatic Neuroendocrine Tumors in MEN1-Results From the Dutch MEN1 Study Group. J Clin Endocrinol Metab, 2017. 102(10): p. 3795-3805.
  151. Hackeng, W.M., et al., Non-functional pancreatic neuroendocrine tumours: ATRX/DAXX and alternative lengthening of telomeres (ALT) are prognostically independent from ARX/PDX1 expression and tumour size. Gut, 2021.
  152. Cejas, P., et al., Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nat Med, 2019. 25(8): p. 1260-1265.
  153. Chan, C.S., et al., ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat Commun, 2018. 9(1): p. 4158.
  154. Fahrmann, J.F., et al., A Blood-Based Polyamine Signature Associated With Disease Progression in Patients With Multiple Endocrine Neoplasia Type 1-Related Duodenopancreatic Neuroendocrine Tumors. Journal of the Endocrine Society, 2021. 5(Supplement_1): p. A1009-A1009.
  155. Niederle, B., et al., Multiple Endocrine Neoplasia Type 1 (MEN1) and the Pancreas - Diagnosis and Treatment of Functioning and Non-Functioning Pancreatic and Duodenal Neuroendocrine Neoplasia within the MEN1 Syndrome - An International Consensus Statement. Neuroendocrinology, 2020.
  156. Falconi, M., et al., ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology, 2016. 103(2): p. 153-71.
  157. Metz, D.C., et al., Diagnosis of Zollinger-Ellison syndrome in the era of PPIs, faulty gastrin assays, sensitive imaging and limited access to acid secretory testing. Int J Endocr Oncol, 2017. 4(4): p. 167-185.
  158. Singh Ospina, N., et al., Assessing for Multiple Endocrine Neoplasia Type 1 in Patients Evaluated for Zollinger-Ellison Syndrome-Clues to a Safer Diagnostic Process. Am J Med, 2017. 130(5): p. 603-605.
  159. Albers, M.B., J. Manoharan, and D.K. Bartsch, Contemporary surgical management of the Zollinger-Ellison syndrome in multiple endocrine neoplasia type 1. Best Pract Res Clin Endocrinol Metab, 2019. 33(5): p. 101318.
  160. Hackeng, W.M., et al., Metastatic Patterns of Duodenopancreatic Neuroendocrine Tumors in Patients With Multiple Endocrine Neoplasia Type 1. Am J Surg Pathol, 2021.
  161. Norton, J.A., et al., Comparison of surgical results in patients with advanced and limited disease with multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome. Annals of surgery, 2001. 234(4): p. 495-505; discussion 505-6.
  162. Endall, R., et al., The Relationship of Gastrinoma in MEN 1 to Helicobacter pylori infection. J Clin Endocrinol Metab, 2020. 105(3).
  163. Cryer, P.E., et al., Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab, 2009. 94(3): p. 709-28.
  164. Bartsch, D.K., et al., Enucleation and limited pancreatic resection provide long-term cure for insulinoma in multiple endocrine neoplasia type 1. Neuroendocrinology, 2013. 98(4): p. 290-8.
  165. Vezzosi, D., et al., Long-term results of the surgical management of insulinoma patients with MEN1: a Groupe d'étude des Tumeurs Endocrines (GTE) retrospective study. Eur J Endocrinol, 2015. 172(3): p. 309-19.
  166. Tonelli, F., et al., Operation for insulinomas in multiple endocrine neoplasia type 1: When pancreatoduodenectomy is appropriate. Surgery, 2017. 161(3): p. 727-734.
  167. Antwi, K., et al., 68Ga-Exendin-4 PET/CT Detects Insulinomas in Patients With Endogenous Hyperinsulinemic Hypoglycemia in MEN-1. J Clin Endocrinol Metab, 2019. 104(12): p. 5843-5852.
  168. Shah, R., et al., Exendin-4-based imaging in insulinoma localization: Systematic review and meta-analysis. Clin Endocrinol (Oxf), 2021. 95(2): p. 354-364.
  169. van Beek, D.J., et al., Prognosis after surgery for multiple endocrine neoplasia type 1-related pancreatic neuroendocrine tumors: Functionality matters. Surgery, 2021. 169(4): p. 963-973.
  170. Dasari, B.V.M., et al., Outcomes of Surgical and Endoscopic Resection of Duodenal Neuroendocrine Tumours (NETs): a Systematic Review of the Literature. J Gastrointest Surg, 2018. 22(9): p. 1652-1658.
  171. Delle Fave, G., et al., ENETS Consensus Guidelines Update for Gastroduodenal Neuroendocrine Neoplasms. Neuroendocrinology, 2016. 103(2): p. 119-24.
  172. Rimbaş, M., et al., Interventional endoscopic ultrasound for pancreatic neuroendocrine neoplasms. Dig Endosc, 2020. 32(7): p. 1031-1041.
  173. Caplin, M.E., et al., Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med, 2014. 371(3): p. 224-33.
  174. Caplin, M.E., et al., Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer, 2016. 23(3): p. 191-9.
  175. Ramundo, V., et al., Impact of long-acting octreotide in patients with early-stage MEN1-related duodeno-pancreatic neuroendocrine tumours. Clinical endocrinology, 2014. 80(6): p. 850-5.
  176. Cioppi, F., et al., The LARO-MEN1 study: a longitudinal clinical experience with octreotide Long-Acting Release in patients with Multiple Endocrine Neoplasia type 1 Syndrome. Clin Cases Miner Bone Metab, 2017. 14(2): p. 123-130.
  177. Faggiano, A., et al., Lanreotide Therapy vs Active Surveillance in MEN1-Related Pancreatic Neuroendocrine Tumors < 2 Centimeters. J Clin Endocrinol Metab, 2020. 105(1).
  178. Pavel, M., et al., ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology, 2016. 103(2): p. 172-85.
  179. Yao, J.C., et al., Everolimus for advanced pancreatic neuroendocrine tumors. The New England journal of medicine, 2011. 364(6): p. 514-23.
  180. Raymond, E., et al., Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England journal of medicine, 2011. 364(6): p. 501-13.
  181. Strosberg, J., et al., Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med, 2017. 376(2): p. 125-135.
  182. Frost, M., K.E. Lines, and R.V. Thakker, Current and emerging therapies for PNETs in patients with or without MEN1. Nat Rev Endocrinol, 2018. 14(4): p. 216-227.
  183. Debelenko, L.V., et al., The multiple endocrine neoplasia type I gene locus is involved in the pathogenesis of type II gastric carcinoids. Gastroenterology, 1997. 113(3): p. 773-81.
  184. Lee, L., et al., Insights into Effects/Risks of Chronic Hypergastrinemia and Lifelong PPI Treatment in Man Based on Studies of Patients with Zollinger-Ellison Syndrome. Int J Mol Sci, 2019. 20(20).
  185. Manoharan, J., et al., Gastric enterochromaffin-like cell changes in multiple endocrine neoplasia type 1. Clin Endocrinol (Oxf), 2021. 95(3): p. 439-446.
  186. Richards, M.L., et al., Regression of type II gastric carcinoids in multiple endocrine neoplasia type 1 patients with Zollinger-Ellison syndrome after surgical excision of all gastrinomas. World J Surg, 2004. 28(7): p. 652-8.
  187. Kloppel, G., M. Anlauf, and A. Perren, Endocrine precursor lesions of gastroenteropancreatic neuroendocrine tumors. Endocrine pathology, 2007. 18: p. 150-155.
  188. Berna, M.J., et al., A prospective study of gastric carcinoids and enterochromaffin-like cell changes in multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome: identification of risk factors. J Clin Endocrinol Metab, 2008. 93(5): p. 1582-91.
  189. Bordi, C., et al., Aggressive forms of gastric neuroendocrine tumors in multiple endocrine neoplasia type I. Am J Surg Pathol, 1997. 21(9): p. 1075-82.
  190. Norton, J.A., et al., Gastric carcinoid tumors in multiple endocrine neoplasia-1 patients with Zollinger-Ellison syndrome can be symptomatic, demonstrate aggressive growth, and require surgical treatment. Surgery, 2004. 136(6): p. 1267-74.
  191. Mandl, A., et al., Two distinct classes of thymic tumors in patients with MEN1 show LOH at the MEN1 locus. Endocr Relat Cancer, 2021. 28(11): p. L15-l19.
  192. Volante, M., et al., Molecular Pathology of Well-Differentiated Pulmonary and Thymic Neuroendocrine Tumors: What Do Pathologists Need to Know? Endocr Pathol, 2021. 32(1): p. 154-168.
  193. Teh, B.T., et al., Clinicopathologic Studies of Thymic Carcinoids in Multiple Endocrine Neoplasia Type 1. Medicine (Baltimore), 1997. 76(1): p. 21-29.
  194. Ye, L., et al., Clinical features and prognosis of thymic neuroendocrine tumours associated with multiple endocrine neoplasia type 1: A single-centre study, systematic review and meta-analysis. Clinical Endocrinology, 2017. 87(6): p. 706-716.
  195. Singh Ospina, N., et al., Thymic and Bronchial Carcinoid Tumors in Multiple Endocrine Neoplasia Type 1: The Mayo Clinic Experience from 1977 to 2013. Hormones and Cancer, 2015. 6(5-6): p. 247-253.
  196. Sakurai, A., et al., Thymic neuroendocrine tumour in multiple endocrine neoplasia type 1: Female patients are not rare exceptions. Clinical Endocrinology, 2013. 78(2): p. 248-254.
  197. Goudet, P., et al., Thymic neuroendocrine tumors in multiple endocrine neoplasia type 1: A comparative study on 21 cases among a series of 761 MEN1 from the GTE (Groupe des Tumeurs Endocrines). World Journal of Surgery, 2009. 33(6): p. 1197-1207.
  198. Gibril, F., et al., Prospective study of thymic carcinoids in patients with multiple endocrine neoplasia type 1. Journal of Clinical Endocrinology and Metabolism, 2003. 88(3): p. 1066-1081.
  199. Ferolla, P., et al., Thymic neuroendocrine carcinoma (carcinoid) in multiple endocrine neoplasia type 1 syndrome: The Italian series. Journal of Clinical Endocrinology and Metabolism, 2005. 90(5): p. 2603-2609.
  200. de Laat, J.M., et al., Natural course and survival of neuroendocrine tumors of thymus and lung in MEN1 patients. Journal of Clinical Endocrinology and Metabolism, 2014. 99(9): p. 3325-3333.
  201. Christakis, I., et al., Clinical Features, Treatments, and Outcomes of Patients with Thymic Carcinoids and Multiple Endocrine Neoplasia Type 1 Syndrome at MD Anderson Cancer Center. Hormones and Cancer, 2016. 7(4): p. 279-287.
  202. Lim, L.C., et al., Thymic carcinoid in multiple endocrine neoplasia 1: Genotype-phenotype correlation and prevention. Journal of Internal Medicine, 2006. 259(4): p. 428-432.
  203. Sadowski, S.M., et al., The future: surgical advances in MEN1 therapeutic approaches and management strategies. Endocrine-related cancer, 2017. 24(10): p. T243-T260.
  204. van den Broek, M.F.M., et al., The Management of Neuroendocrine Tumors of the Lung in MEN1: Results From the Dutch MEN1 Study Group. The Journal of clinical endocrinology and metabolism, 2021. 106(2): p. e1014-e1027.
  205. Sachithanandan, N., R.A. Harle, and J.R. Burgess, Bronchopulmonary carcinoid in multiple endocrine neoplasia type 1. Cancer, 2005. 103(3): p. 509-515.
  206. Lecomte, P., et al., Histologically Proven Bronchial Neuroendocrine Tumors in MEN1: A GTE 51-Case Cohort Study. World Journal of Surgery, 2018. 42(1): p. 143-152.
  207. Bartsch, D.K., et al., Bronchopulmonary Neuroendocrine Neoplasms and Their Precursor Lesions in Multiple Endocrine Neoplasia Type 1. Neuroendocrinology, 2016. 103(3-4): p. 240-247.
  208. van den Broek, M.F.M., et al., Well-Differentiated Bronchopulmonary Neuroendocrine Tumors: More Than One Entity. J Thorac Oncol, 2021. 16(11): p. 1810-1820.
  209. Sadowski, S.M., et al., Results of 68Gallium-DOTATATE PET/CT Scanning in Patients with Multiple Endocrine Neoplasia Type 1. Journal of the American College of Surgeons, 2015. 221(2): p. 509-517.
  210. Lastoria, S., et al., Role of 68Ga-DOTATATE PET/CT in patients with multiple endocrine neoplasia type 1 (MEN1). Endocrine, 2016. 52(3): p. 488-494.
  211. Froeling, V., et al., Impact of Ga-68 DOTATOC PET/CT on the diagnosis and treatment of patients with multiple endocrine neoplasia. Annals of Nuclear Medicine, 2012. 26(9): p. 738-743.
  212. Albers, M.B., et al., Limited Value of Ga-68-DOTATOC-PET-CT in Routine Screening of Patients with Multiple Endocrine Neoplasia Type 1. World journal of surgery, 2017. 41(6): p. 1521-1527.
  213. Harding, B., et al., Multiple endocrine neoplasia type 1 knockout mice develop parathyroid, pancreatic, pituitary and adrenal tumours with hypercalcaemia, hypophosphataemia and hypercorticosteronaemia. Endocr Relat Cancer, 2009. 16(4): p. 1313-27.
  214. Loffler, K.a., et al., Broad tumor spectrum in a mouse model of multiple endocrine neoplasia type 1. International journal of cancer. Journal international du cancer, 2007. 120: p. 259-67.
  215. Ludwig, L., et al., Loss of wild-type MEN1 gene expression in multiple endocrine neoplasia type 1-associated parathyroid adenoma. Endocr J, 1999. 46(4): p. 539-44.
  216. Skogseid, B., et al., Clinical and genetic features of adrenocortical lesions in multiple endocrine neoplasia type 1. J Clin Endocrinol Metab, 1992. 75(1): p. 76-81.
  217. Gatta-Cherifi, B., et al., Adrenal involvement in MEN1. Analysis of 715 cases from the Groupe d'etude des Tumeurs Endocrines database. Eur J Endocrinol, 2012. 166(2): p. 269-79.
  218. Barzon, L., et al., Multiple endocrine neoplasia type 1 and adrenal lesions. J Urol, 2001. 166(1): p. 24-7.
  219. Burgess, J.R., et al., Adrenal lesions in a large kindred with multiple endocrine neoplasia type 1. Arch Surg, 1996. 131(7): p. 699-702.
  220. Langer, P., et al., Adrenal involvement in multiple endocrine neoplasia type 1. World J Surg, 2002. 26(8): p. 891-6.
  221. Schaefer, S., et al., Natural course of small adrenal lesions in multiple endocrine neoplasia type 1: an endoscopic ultrasound imaging study. Eur J Endocrinol, 2008. 158(5): p. 699-704.
  222. Skogseid, B., et al., Adrenal lesion in multiple endocrine neoplasia type 1. Surgery, 1995. 118(6): p. 1077-82.
  223. Ventura, M., M. Melo, and F. Carrilho, Outcome and long-term follow-up of adrenal lesions in multiple endocrine neoplasia type 1. Arch Endocrinol Metab, 2019. 63(5): p. 516-523.
  224. Waldmann, J., et al., Adrenal involvement in multiple endocrine neoplasia type 1: results of 7 years prospective screening. Langenbecks Arch Surg, 2007. 392(4): p. 437-43.
  225. Whitley, S.A., et al., The appearance of the adrenal glands on computed tomography in multiple endocrine neoplasia type 1. Eur J Endocrinol, 2008. 159(6): p. 819-24.
  226. Wang, W., et al., Adrenocortical carcinoma in patients with MEN1: a kindred report and review of the literature. Endocr Connect, 2019. 8(3): p. 230-238.
  227. Dotzenrath, C., et al., Malignant endocrine tumors in patients with MEN 1 disease. Surgery, 2001. 129(1): p. 91-5.
  228. Febrero, B., A. Ríos, and J.M. Rodríguez, Aggressive adrenal carcinoma in a young patient with Multiple Endocrine Neoplasia 1 syndrome. Med Clin (Barc), 2017. 149(10): p. 463.
  229. Griniatsos, J.E., et al., Bilateral adrenocortical carcinoma in a patient with multiple endocrine neoplasia type 1 (MEN1) and a novel mutation in the MEN1 gene. World J Surg Oncol, 2011. 9: p. 6.
  230. Haase, M., et al., A new mutation in the menin gene causes the multiple endocrine neoplasia type 1 syndrome with adrenocortical carcinoma. Endocrine, 2011. 39(2): p. 153-9.
  231. Harada, K., et al., A novel case of myxoid variant of adrenocortical carcinoma in a patient with multiple endocrine neoplasia type 1. Endocr J, 2019. 66(8): p. 739-744.
  232. Kharb, S., et al., Hidden diagnosis of multiple endocrine neoplasia-1 unraveled during workup of virilization caused by adrenocortical carcinoma. Indian J Endocrinol Metab, 2013. 17(3): p. 514-8.
  233. Ohara, N., et al., Lung adenocarcinoma and adrenocortical carcinoma in a patient with multiple endocrine neoplasia type 1. Respir Med Case Rep, 2017. 20: p. 77-81.
  234. Assié, G., et al., Integrated genomic characterization of adrenocortical carcinoma. Nat Genet, 2014. 46(6): p. 607-12.
  235. Tepede, A.A., et al., 18F-FDOPA PET/CT accurately identifies MEN1-associated pheochromocytoma. Endocrinol Diabetes Metab Case Rep, 2020. 2020.
  236. Dénes, J., et al., Heterogeneous genetic background of the association of pheochromocytoma/paraganglioma and pituitary adenoma: results from a large patient cohort. J Clin Endocrinol Metab, 2015. 100(3): p. E531-41.
  237. Fassnacht, M., et al., Management of adrenal incidentalomas: European Society of Endocrinology Clinical Practice Guideline in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol, 2016. 175(2): p. G1-g34.
  238. Asgharian, B., et al., Cutaneous tumors in patients with multiple endocrine neoplasm type 1 (MEN1) and gastrinomas: Prospective study of frequency and development of criteria with high sensitivity and specificity for MEN1. Journal of Clinical Endocrinology and Metabolism, 2004. 89(11): p. 5328-5336.
  239. Darling, T.N., et al., Multiple Facial Angiofibromas and Collagenomas in Patients With Multiple Endocrine Neoplasia Type 1. Arch Dermatol., 1997. 7(133): p. 853-857.
  240. Vidal, A., et al., Cutaneous lesions associated to multiple endocrine neoplasia syndrome type 1. Journal of the European Academy of Dermatology and Venereology, 2008. 22(7): p. 835-838.
  241. Pack, S., et al., Cutaneous tumors in patients with multiple endocrine neoplasia type 1 show allelic deletion of the MEN1 gene. Journal of Investigative Dermatology, 1998. 110(4): p. 438-440.
  242. Vortmeyer, A.O., et al., Multiple endocrine neoplasia 1 gene alterations in MEN1-associated and sporadic lipomas. Journal of the National Cancer Institute, 1998. 90(5): p. 398-399.
  243. Dreijerink, K.M.A., et al., The Multiple Endocrine Neoplasia Type 1 (MEN1) Tumor Suppressor Regulates Peroxisome Proliferator-Activated Receptor γ-Dependent Adipocyte Differentiation. Molecular and Cellular Biology, 2009. 29(18): p. 5060-5069.
  244. Parekh, V.I., et al., Consequence of Menin Deficiency in Mouse Adipocytes Derived by in Vitro Differentiation. International Journal of Endocrinology, 2015. 2015.
  245. Radman, M. and T. Milicevic, A novel mutation of the MEN1 gene in a patient with multiple endocrine neoplasia type 1 and recurrent fibromyxoid sarcoma- A case report. BMC Medical Genetics, 2020. 21(1): p. 1-4.
  246. Cheah, S.K., et al., Breast cancer in multiple endocrine neoplasia type 1 (MEN1). Endocrinology, Diabetes & Metabolism Case Reports, 2021. 2021(May): p. 1-7.
  247. Jeong, Y.J., H.K. Oh, and J.G. Bong, Multiple endocrine neoplasia type 1 associated with breast cancer: A case report and review of the literature. Oncology letters, 2014. 8(1): p. 230-234.
  248. Kim, S.H. and J.H. Park, Adrenal incidentaloma, breast cancer and unrecognized multiple endocrine neoplasia type 1. Acta Endocrinologica, 2019. 15(4): p. 513-517.
  249. Maria, B.C., et al., Incidental finding of MEN-1 syndrome during staging and follow-up of breast carcinoma. BMJ Case Reports, 2020. 13(12): p. 1-6.
  250. Seigne, C., et al., High incidence of mammary intraepithelial neoplasia development in Men1-disrupted murine mammary glands. The Journal of pathology, 2013. 229(4): p. 546-58.
  251. Imachi, H., et al., Menin, a product of the MENI gene, binds to estrogen receptor to enhance its activity in breast cancer cells: possibility of a novel predictive factor for tamoxifen resistance. Breast cancer research and treatment, 2010. 122(2): p. 395-407.
  252. Dreijerink, K.M., et al., Article Enhancer-Mediated Oncogenic Function of the Menin Tumor Suppressor in Breast Cancer. Cell Reports, 2017. In Press(1).
  253. Teinturier, R., et al., Reduced menin expression leads to decreased ERα expression and is correlated with the occurrence of human luminal B-like and ER-negative breast cancer subtypes. Breast Cancer Research and Treatment, 2021(0123456789).
  254. Goswami, S., et al., Disease and treatment factors associated with lower quality of life scores in adults with multiple endocrine neoplasia type I. Surgery, 2017: p. 1-8.
  255. Marini, F., et al., Management impact: effects on quality of life and prognosis in MEN1. Endocrine-Related Cancer, 2017. 24(10): p. T227-T242.
  256. Peipert, B.J., et al., Financial burden is associated with worse health-related quality of life in adults with multiple endocrine neoplasia type 1. Surgery, 2017: p. 1-8.
  257. Peipert, B.J., et al., Health-related quality of life in MEN1 patients compared with other chronic conditions and the united states general population. Surgery (United States), 2017. 163: p. 205-211.
  258. van Leeuwaarde, R.S., et al., High fear of disease occurrence is associated with low quality of life in patients with Multiple Endocrine Neoplasia type 1 (MEN1): Results from the Dutch MEN1 Study Group. The Journal of Clinical Endocrinology & Metabolism, 2018. 103(June): p. 2354-2361.
  259. van Leeuwaarde, R.S., et al., Health-related quality of life in patients with Multiple Endocrine Neoplasia type 1. Neuroendocrinology, 2020: p. 1-8.
  260. Berglund, G., et al., Quality of life in patients with multiple endocrine neoplasia type 1 ( MEN1 ). Familial Cancer, 2003. 1(2): p. 27-33.
  261. You, Y.N., et al., Pancreatoduodenal surgery in patients with multiple endocrine neoplasia type 1: Operative outcomes, long-term function, and quality of life. Surgery, 2007. 142(6): p. 829-836.

Snakebite Envenomation and Endocrine Dysfunction

ABSTRACT

 

Snakebite envenoming (SBE) is a life-threatening medical emergency encountered in tropical parts of Asia, Africa, and Latin America. Toxins in the venom cause local damage and multi-organ dysfunction, predominantly affecting neurological, hematological, and vascular systems. Endocrine anomalies are less frequently reported and often masked by more severe disorders. Anterior pituitary insufficiency is the most common endocrine manifestation and mainly observed after Russell’s viper (Daboia russelii and D. siamensis) bite. SBE-induced hypopituitarism can manifest early or have a delayed presentation. Primary adrenal insufficiency, hyponatremia, hypokalemia, hyperkalemia, and hyperglycemia are also described. These complications are uncommon and under-reported, as SBE occurs in remote areas and medical facilities for endocrine assessment might not be available. Timely identification and management of these problems are critical for optimum medical outcome.

 

INTRODUCTION

 

Snakebite envenoming (SBE) occurs predominantly in rural parts of Asia, Africa, and Latin America (1,2). The World Health Organization (WHO) included SBE in the priority list of neglected tropical diseases in 2018. According to the WHO, 4.5–5.4 million people get bitten by snakes annually, but many cases are not reported as people living in remote areas with limited healthcare access are affected. Clinical illness from SBE develops in 1.8–2.7 million, and the annual mortality is around 81,000 to 138,000 (3).

 

Toxins in snake venom can cause local tissue destruction, neurological damage, hemorrhagic tendency, renal failure, and cardiovascular compromise. Endocrine dysfunctions are uncommon but can have ominous consequences if not recognized. Anterior pituitary insufficiency (API) after Russell’s viper (RV) envenomation (RVE) is the most common endocrine manifestation of SBE. Electrolyte disturbances and hyperglycemia are the other complications described (4). Timely recognition and appropriate management of endocrine derangements like hypocortisolism and electrolyte imbalances can save lives.

 

SPECIES OF SNAKES AND SNAKE VENOM

 

The medically relevant poisonous snakes usually belong to the Elapidae and Viperidae families. Rare cases of envenoming from Atractaspididae and Colubridae families are also described. The common Elapidae snakes include cobras, mambas, kraits, coral snakes, death adders, and sea snakes. The Viperidae snakes of significance are vipers, including RV, adders, asps, and pit vipers. The general notion that Elapidae envenomation results in neuroparalytic manifestation and Viperidae bite induce local reaction and vasculotoxicity does not always hold.

 

Snake venoms contain mixtures of polypeptides, amines, carbohydrates, lipids, phospholipids, nucleosides, and minerals. The principal constituents are proteins belonging to the four families: phospholipase A2, metalloprotease, serine protease, and three-finger peptides. Additional secondary protein families include cysteine-rich secretory proteins, l-amino acid oxidases, kunitz peptides, C-type lectins/snaclecs, disintegrins, and natriuretic peptides (5).

 

Snake venom toxicity can be classified into three main categories: vasculotoxic, neurotoxic, and cytotoxic. Various proteins with enzymatic properties such as phospholipase A2, hyaluronidases, peptidases, and metalloprotease can cause local tissue destruction. Phospholipase A2, metalloproteases, and other protein components can cause neurotoxicity, damage the coagulation cascade, induce muscle necrosis, and sometimes exert cardiotoxic and nephrotoxic effects (6). Cardiac compromise and acute kidney injury (AKI) can also emerge as secondary complications. Endocrine disorders after SBE are uncommon and pathophysiologic mechanisms are incompletely understood.

 

ANTERIOR PITUITARY INSUFFICIENCY

 

Anterior pituitary insufficiency (API) is the most well-recognized endocrine manifestation of SBE. Most cases are from Sri Lanka, India, and Myanmar and occur after RVE (Daboia russelii and D. siamensis).

 

Etiology

 

Wolff first narrated SBE-induced hypopituitarism in 1958 after bite from Bothrops jararacussu (7). The first description of API following RVE in the Indian subcontinent came from Eapen et al. (8). Although RV is found in many south Asian countries, most accounts of RVE-induced API are almost exclusively from Myanmar, India, and Sri Lanka (9–16). It could be related to the geographic variation in venom composition among the same snake species. The incidence of API in a study from northern India was 14.6% (6/41) among patients admitted with vasculotoxic snakebites (presumed RVE) (12).

 

Pathophysiology

 

The pituitary gland is a highly vascular structure enclosed in a bony cavity called the sella tursica. The low-pressure hypothalamic-pituitary portal system originating from the superior hypophyseal artery provides blood supply to the anterior pituitary. The hypophyseal portal system is susceptible to compressive effects from an enlarged or engorged gland and renders the anterior pituitary vulnerable to vascular insults after stimulation from any cause.

 

Sheehan’s syndrome and RVE-induced hypopituitarism share similar pathophysiology (16,17). In Sheehan’s syndrome, hemorrhagic infarction of the pregnancy-induced hyperplastic gland occurs during severe postpartum bleeding-related hypovolemic shock (18). Predisposition to vascular damage in RVE could result from gland engorgement due to a generalized increase in capillary permeability as in capillary leak syndrome (19–21). Additionally, the toxins in RV venom can stimulate pituitary cells as suggested by in-vitro studies, further increasing the susceptibility to damage (22).

 

The vascular supply to an engorged and stimulated gland might be compromised due to microthrombi deposition or hemorrhage from disseminated intravascular coagulation (DIC) (16,23), circulatory or hypovolemic shock (14), thrombotic occlusion of major vessels including cerebral venous thrombosis (24,25), and increased intracranial pressure (14). Autoimmune damage has been postulated to contribute to delayed pituitary injury in Sheehan’s syndrome (26,27). The role of similar immune-mediated damage in the development of delayed hypopituitarism after RVE has not been studied.

 

Clinical Features

 

ACUTE HYPOPITUITARISM

 

Acute onset API has been observed after RVE in several series and can present as early as the first day (11,12,15). In one series of nine patients, API occurred after a median interval of nine days (range 2-14 days) (15). The usual manifestations of RVE include local reaction, coagulopathy, neuromuscular paralysis, and AKI (28,29). Circulatory shock, another feature of RVE, is multifactorial in etiology (14). In the acute phase, adrenal insufficiency (AI) dominates the clinical presentation of API. The symptoms of AI often get masked by other systemic effects of RVE. Clinical clues could be refractory hypotension and the presence of hypoglycemia or hyponatremia.

 

Central hypothyroidism may coexist with secondary AI and is diagnosed if serum thyroid-stimulating hormone (TSH) is low or normal along with decreased serum thyroxine levels. TSH can also get suppressed due to sick euthyroid syndrome and glucocorticoid administration. The diagnosis of central hypothyroidism is difficult to discern in the acute phase, and a follow-up test after recovery is necessary for confirmation. Reassessment of hypothalamic-pituitary-adrenal (HPA) and additional evaluation of the gonadotrophic and growth hormone (GH) secretion should be performed after 4-6 weeks. Acute hypopituitarism is sometimes transient, but the typical outcome is a permanent disease (12,13).

 

DELAYED HYPOPITUITARISM

 

API is often diagnosed years after RVE (30–33). The symptoms depend on the hormone axis involved and the extent of hormone deficiency (34). Delayed hypopituitarism often presents as secondary amenorrhea and infertility in females. In males, hypogonadotropic hypogonadism usually manifests as loss of libido and erectile dysfunction. Loss of secondary sexual characteristics can be present in both genders.

 

Standard features of secondary hypothyroidism include cold intolerance, weight gain, constipation, dry skin, and hoarseness. Secondary AI presents as fatigue, loss of appetite, and orthostatic dizziness (34). Involvement in early childhood can cause stunted growth and delayed or absent puberty (31). In a case series of delayed API, secondary hypothyroidism and hypogonadotropic hypogonadism were present in all the cases (8/8); and GH and secondary AI were present in 75% (6/8) (30). Table 1 depicts the recent case series describing API.

 

Predictors of Hypopituitarism

 

The presence of acute kidney injury (AKI) most consistently correlates with the development of API after RVE (13,30). Bhat et al. found that in patients (n=51) with vasculotoxic snakebite-associated AKI, the risk factors for API were younger age, the number of hemodialysis sessions, and 20-min whole blood clotting time (13). There was a history of AKI in 75% of cases of delayed hypopituitarism in another series (30). In a study describing nine patients with acute API, the predictors were multi-organ dysfunction, lower platelet counts, and more bleeding with a requirement for transfusions (15). However, coagulopathy, AKI, hemodialysis, and clinical severity scores failed to show any association with hypopituitarism in a prospective study (12).

 

Table 1. Case Series of Hypopituitarism After Snakebite Envenoming

Author, year

Region

Snake species

No. of patients

Onset,  Time

Clinical features/ hormone axes involved/ comments

Tun Pe, 1987(10)

Myanmar

Not defined

Snakebite – 220

Acute API – 3

PH (on autopsy) – 4

 

Delayed  API – 11

Acute: 21 hr - 9 d

 

Delayed: 2 wk - 24 yr

Acute - C, GH, PRL

 

Delayed

Symptomatic - 7 Asymptomatic – 4

Proby, 1989 (35)

Myanmar

RV

Acute API (probable) – 20

 

Delayed API – 11/12

Acute, NA

 

Delayed – 8 - 226 wk

C - 10/15

T - 19/20

G -12/17

Golay, 2014 (11)

West Bengal, India

Vasculotoxic snakebite

API - 9/96 cases of snakebite associated AKI

Acute and delayed, 2 wk - 10 yr

C - 6/9

G, GH, T - 9/9

1 empty sella, rest normal

Rajagopala, 2014 (15)

Puducherry, India

Vasculotoxic snakebite

9/989 cases

Acute, 2-14 d

Hypoglycemia (100%), hypotension (67%)

C - 9/9

Partial empty sella in 6/9

Naik, 2018 (12)

India

Vasculotoxic snakebite

9/41 cases

Acute (10%), Mean - 32 hr

Primary AI - 2/6,

C, GH, G - 6/6

PRL - 2/6

 

White, 2019 (36)

Myanmar

RV (85.4%), Rest - cobra, krait, green pit viper, others

20/948 cases

Acute (2%)

Coagulopathy - 68.9%,

 AKI - 72.2%

Gopalkrishnan, 2018 (14)

India

RV, saw-scaled viper

SB - 248

AI – 12

API - 4

Acute

C -19/48

Autopsy - 52.

PH or ischemic necrosis - 46%

Bilateral adrenal hemorrhage - 26%, Adrenal ischemic necrosis – 6%

Shivaprasad, 2018 (30)

Karnataka, India

RV

Delayed API - 8

Delayed, 5-11 yr

C, GH - 6/8

T, G - 8/8

Bhat, 2019 (13)

West Bengal, India

Vasculotoxic snakebite

API - 11/51 at 7 d and 13/33 at 3 mn after snakebite associated AKI

Acute and delayed,

7 d – 3 mn

C – 12/13

PRL – 9/13

G – 9/13

GH – 5/13

T – 4/13

RV - Russell’s viper, C - cortisol, GH - growth hormone, G - gonadotropin, PRL - prolactin, T - thyroid, API – anterior pituitary insufficiency, AI - adrenal insufficiency, PH – pituitary hemorrhage, AKI – acute kidney injury, SB- snake bite

 

Diagnosis

 

ACUTE HYPOPITUITARISM

 

It is challenging to diagnose API during the acute phase. The indicators associated with API are summarized in table 2. The assessment of the HPA axis is required to decide the necessity for glucocorticoid replacement. Hypocortisolism in the acute phase is diagnosed from random cortisol or with the cosyntropin stimulation test. In remote areas, if a delay is anticipated in obtaining the cortisol report, hydrocortisone replacement should be started in suspected cases. The different criteria that have been used to diagnose hypocortisolism in the acute phase are (a) fasting serum cortisol < 3 μg/dL (83 nmol/L) (13), (b) random serum cortisol < 5 μg/dL (138 nmol/L) in suspected pituitary apoplexy (37), (c) random serum cortisol < 10 μg/dL (275 nmol/L) in a critically ill patient (14), (d) post-cosyntropin peak cortisol < 18 μg/dl (500 nmol/L), and (e) post-cosyntropin delta cortisol < 9 μg/dL (250 nmol/L) (38). Note in very ill patients serum cortisol levels can be artifactually low secondary to a decrease in cortisol binding protein.

 

The interpretation of the thyroid function test can be problematic in the acute phase. Low luteinizing hormone (LH), follicle-stimulating hormone (FSH), and prolactin might be indicative but not diagnostic of API. The gonadal axis and sex hormone secretion (testosterone in males and estradiol in females) is usually suppressed during any severe illness. The pituitary function should be reassessed after 4-6 weeks of recovery. The MRI findings reveal a normal gland on imaging in acute cases (11,12,15). Pituitary hemorrhage has been demonstrated in autopsy findings but is seldom observed in imaging studies (15).

 

Table 2. Features Suggestive Of or Associated with Acute Hypopituitarism after Russell’s Viper Bite

Clinical features

Laboratory results

Imaging

Persistent or unexplained hypotension

Hypoglycemia

Pituitary hemorrhage or infarct on MRI

Acute kidney injury

Hyponatremia

Capillary leak syndrome

 

 

Disseminated intravascular coagulation

 

 

 

DELAYED HYPOPITUITARISM

 

Acute hypopituitarism may progress to chronic disease or manifest insidiously years later (11,12,30). It may be prudent to perform periodic surveillance to rule out the development of API following RVE. Hypogonadism has been described in 100% of cases, central hypothyroidism in 96.4%, secondary AI in 82%, and GH deficiency in 77% (30). Central diabetes insipidus (CDI) is very rare and discussed next. The clinical presentation depends on the age, the hormone axes involved, and the mode of onset. The tests recommended for the diagnosis of pituitary hormone deficiency are outlined in table 3. However, facilities for the dynamic tests may not be available in remote areas where SBE is prevalent (34). MRI usually reveals a normal pituitary during the initial year, but partial or complete empty sella may be found later on (4). 

 

Table 3. Investigations for Diagnosis of Chronic Anterior Pituitary Insufficiency

Hormone

Tests

Interpretation/comments

ACTH

Cortisol, ACTH between 8- 9 AM

Serum cortisol values < 3 μg/dL (83 nmol/L) at 8–9 AM on 2 occasions strongly suggest AI in an appropriate clinical setting. Intermediate levels (3-18 μg/dl; 83 – 497 nmol/L) require cosyntropin stimulation test. Concomitant normal or low ACTH levels indicate secondary AI.

 

Cosyntropin (Synacthen) stimulation test

Cosyntropin injection 250 μg (i.v. or i.m.) followed by serum cortisol at 30 min and 60 min. Peak cortisol < 18 μ/dl (500 nmol/L) is suggestive of AI.

 

Insulin tolerance test

Serum cortisol < 20 μg/dL (550 nmol/L) at the time of insulin induced hypoglycemia < 40 mg/dL (2.2 mmol/L). Extreme caution required, not practiced in many centers.

 

 

 

TSH

T4 (total or free), TSH

Low T4 with low or normal TSH suggests the diagnosis of central hypothyroidism

 

 

 

GH

IGF-1

Low IGF-1 suggestive but not diagnostic.

 

GH stimulation test

Adults: Insulin tolerance test, arginine, GHRH stimulation test, Macimorelin stimulation test, glucagon stimulation test.

Children: Clonidine stimulation test in addition to tests used for adults.

 

 

 

Gonadotrophin (Males)

LH, FSH, testosterone (total), SHBG

Low total testosterone (<300 ng/dl (10.41 nmol/L)) between 8–9 AM, preferably on 2 occasions along with low or normal LH, FSH is suggestive of gonadotrophin deficiency. SHBG and free or bioavailable testosterone measurement should be considered in borderline cases.

Gonadotrophin (Females)

LH, FSH, estradiol

Low estradiol in the setting of low or normal LH and FSH in the appropriate clinical setting (amenorrhea/ oligomeorrhea) suggests gonadotrophin deficiency.

 

 

 

Prolactin

Prolactin

Low levels found in hypopituitarism

 ACTH – adrenocorticotrophic hormone, AI – adrenal insufficiency, TSH – thyroid-stimulating hormone, T4 – thyroxine, GH - growth hormone, IGF-1 – insulin-like growth factor 1, LH – luteinizing hormone, FSH – follicle-stimulating hormone, SHBG – sex-hormone binding globulin

 

Management

 

Acute hypopituitarism typically occurs in critically ill patients with severe envenomation from RV. Intravenous hydrocortisone is required if AI is suspected or diagnosed. Thyroxine supplementation for hypothyroidism should be started only after correcting AI. There is a risk of precipitating adrenal crisis because of the accelerated metabolic clearance of cortisol, if thyroxine is administered before treatment of AI (39). Monitoring of electrolytes, and slow correction of hyponatremia when present, in order to prevent central pontine myelinolysis, are important adjuncts.

 

If oral intake is proper and the patient is hemodynamically stable, intravenous hydrocortisone can be substituted by oral glucocorticoids. Hormonal evaluation of the entire pituitary axes should be performed after 4-6 weeks of recovery. Replacement of deficient hormones as per standard practice should be instituted if API persists (34).

 

Post-mortem Findings

 

Pituitary hemorrhage and ischemic necrosis have been described in autopsy studies of 43% (36/84) cases of RVE in Myanmar (40). Areas of ischemic necrosis with hemorrhage at the center were observed in studies from India (15). Deposition of fibrin microthrombi in the pituitary and other organs, including the kidney, suggests a possible role of DIC in the pathogenesis of API (23). 

 

DIABETES INSIPIDUS

 

Involvement of the posterior pituitary gland is exceedingly rare in SBE. There are only a few case reports of central diabetes insipidus (CDI) after RVE (13,31,41,42). The posterior pituitary receives direct arterial supply from the inferior hypophyseal artery and is resistant to vascular damage. On the other hand, the anterior gland is susceptible to vascular compromise as it is supplied by the low-pressure hypophyseal-portal system (43). Moreover, CDI occurs when more than 80% of arginine vasopressin (AVP)-producing hypothalamic magnocellular neurons are lost. The posterior pituitary acts as a storage and secretory organ, and persistent CDI ensues only in the presence of significant damage to the hypothalamus (44). Polyuria, a cardinal feature of CDI, may be obscured due to concomitant hypocortisolism and manifest only after glucocorticoid replacement (45). CDI should be treated with nasal or oral desmopressin.

 

ADRENAL DISORDERS

 

Etiopathogenesis

 

Secondary AI from hypopituitarism is the classically described adrenal disorder resulting from SBE. Primary AI is exceptionally uncommon, though adrenal hemorrhage (AH) has been described in imaging studies and autopsy findings. There are cases of AH occurring after RVE and saw scale viper (Echis carinatus) bite (40,46,47).

 

The pathophysiology of AH is related to DIC and has been postulated to resemble Waterhouse–Friderichsen syndrome (48). The adrenal gland is a highly vascular structure that derives its arterial supply from three arteries but is drained by only one adrenal vein and has a dense internal network of capillaries (49). The causal factors behind predisposition to AH after RVE are summarized in table 4 (14,50,51).

 

Table 4. Factors Predisposing to Hemorrhage after Russell’s Viper Envenomation

Intrinsic predisposition of adrenal vascular structure due to arterial supply by 3 vessels but drainage by one vein

Rich subcapsular plexus with limited drainage by venules forming a “dam”

Disseminated intravascular coagulation and hemorrhagic toxins in the venom increase bleeding tendency

Formation of microthrombi in venules impair venous drainage and cause pooling of blood

Pooling of blood due to capillary leak syndrome

Stress-induced trophic effect of adrenocorticotrophic hormone induces adrenal cortical hyperplasia and increase vascularity

Stress-induced catecholamine secretion causes adrenal venous constriction resulting in pooling of blood in the adrenal gland

 

Diagnosis And Treatment

 

AH has been described in 36% of cases in an autopsy series, though primary AI is rare (40). Refractory hypotension, hypoglycemia, hyponatremia and hyperkalemia should raise suspicion of primary AI. The presence of associated secondary AI can confound the diagnosis. Bilateral AH with transient AI has been described following RVE. The hemorrhage and adrenal function resolved in weeks (47). Cases depicting chronic AI have been published after vasculotoxic SBE (12). Diagnosis and treatment are similar to secondary AI; the primary differentiating point is elevated plasma ACTH. Mineralocorticoid supplementation may be additionally required in primary AI.

 

HYPERGLYCEMIA

 

Etiopathogenesis

 

Hyperglycemia, an infrequent endocrine complication after SBE, has been described following both elapid (Bungarus multicinctus multicinctus) and viper envenomation (Vipera ammodytes ammodytes, European viper spp) (52–54). In rat models, common krait (Bungarus caeruleus) venom produces hyperglycemia (55). Intraperitoneal injection of saw-scaled viper (Echis carinatus) venom in rats suppressed plasma insulin and depleted liver glycogen stores (56). The hyperglycemic effect of Egyptian cobra (Naja haje) was also associated with concomitant depletion of liver and kidney glycogen stores. The mechanism of hyperglycemia is presumed to be triggered by a massive surge of catecholamines, a phenomenon observed after scorpion envenomation and in pheochromocytoma (4,57). Scorpion toxins stimulate sodium and inhibit potassium channels leading to intense and persistent excitation of the autonomic nervous system and release of neurotransmitters from the adrenal medulla, activating parasympathetic (early hours) and sympathetic nerve endings (4–48 hours). Catecholamine-mediated activation of the alpha receptors inhibits insulin secretion and contributes to hyperglycemia (54,58).

 

Clinical Features and Management

 

In a series of 83 children, viper envenomation resulted in hyperglycemia starting 4 hours after the bite, was moderate in severity, and usually transient. Moreover, hyperglycemia at presentation was a marker of high-grade envenomation (54). Severe hyperglycemia up to 480 mg/dl (26.7 mmol/L) occurred in a 45-day baby after two hours of bite from a nose-horned viper (V. a. ammodytes). (53). A retrospective study from Taiwan found hyperglycemia in 15% (7/44) patients of Bungarus multicinctus envenomation. Only one of them had persistent diabetes after recovery (52). Acute pancreatitis can result from the bite of the adder (Vipera berus), but associated hyperglycemia was not observed (59,60). Insulin and other antihyperglycemic drugs should be administered for management of hyperglycemia as and when necessary.

 

ELECTROLYTE DISTURBANCES

 

Hyponatremia

 

ETIOPATHOGENESIS

 

Envenomation by Malayan krait (Bungarus candidus), banded krait (Bungarus fasciatus)), and vipers can result in hyponatremia (61–68). Hyponatremia sometimes occur secondary to anterior pituitary insufficiency (API) after vasculotoxic envenoming but it has also been reported in the absence of API (4). Initial descriptions suggested that the syndrome of inappropriate antidiuretic hormone secretion (SIADH) could be responsible for hyponatremia (64). However, subsequent accounts revealed that urinary salt loss from natriuretic peptides in venom, rather than SIADH, is the pathogenic mechanism. The urinary salt loss is secondary to venom-derived natriuretic peptides, similar to endogenous natriuretic peptides, and acts on the renal tubules to decrease sodium and water reabsorption (69). Cerebral salt wasting has also been postulated to cause hyponatremia (61). An unusually high prevalence of hyponatremia (89%) was observed in a series of 14 patients with berg adder (Bitis atropos) bite in South Africa (67). Many-banded krait (Bungarus multicinctus) envenomation caused hyponatremia in 42% of cases (63).

 

Natriuretic peptides are found in the venom of Elapidae species such as Bungarus candidus,  Bungarus multicinctus, Dendroaspi sangusticeps, Oxyuranus microlepidotus, Pseudonaja textillis, and Pseudechis australis and a few Viperidae species e.g. Hypnale hypnale,  Psudocerastus persicus, and Macrovipera lebetina (66).

 

CLINICAL FEATURES

 

The presentation of hyponatremia depends on the severity and acuteness of onset. The clinical profile ranges from asymptomatic hyponatremia to varying alteration in sensorium to frank coma (61,62). Seizures occur in severe cases (62). Usually there are associated systemic features but isolated hyponatremia, hypovolemia, urinary salt loss, and generalized tonic-clonic seizures, following hump-nosed pit viper bite (Hypnale hypnale) has been described (66). In a case series of 42 patients admitted in Vietnam, 31 people (73.8%) had hyponatremia, the lowest values occurring an average of two days after the bite. Approximately 42–50% of patients who did not receive antivenom developed significant hyponatremia (< 130 mmol/L) 2–3 days post-bite. (70).  Another series of 78 cases of krait bite from Thailand reported hyponatremia in 17.6%, with severe hyponatremia (< 120 mmol/L) developing in four pediatric patients, two of whom developed seizures (71).

 

MANAGEMENT

 

Hyponatremia resulting from natriuretic peptides should be corrected by intravenous saline administration. SIADH is not the cause of hyponatremia, and fluid restriction is not recommended. If chronic hyponatremia is suspected, the correction rate should not exceed 10-12 meq/L in any 24 hours to avoid osmotic demyelination (72). If primary or secondary AI is the cause of hyponatremia, glucocorticoid supplementation is necessary.

 

Hypokalemia

 

ETIOPATHOGENESIS

 

Hypokalemia results from both elapid (73,74) and viper envenomation (75–77). Patients with hypokalemia after RV, common krait (Bungarus caeruleus), and Balkan adder (Vipera berus) bite demonstrated low trans-tubular potassium gradient (TTKG) ruling out renal potassium loss. The intracellular redistribution of potassium has been suggested as the likely pathophysiological mechanism, as gastrointestinal loss was also unlikely. Beta-adrenergic stimulation from toxin-mediated autonomic dysfunction leads to the intracellular shift of potassium and is the likely cause of hypokalemia (74,75). Concomitant hypomagnesemia and high urinary magnesium excretion were also observed in patients with hypokalemia, following Viperidae bite, presumably resulting from the direct toxic action of venom on the renal tubules (77). A high incidence (71%) of hypokalemia (<3.5 mmol/l) was found in a series of 210 patients from Sri Lanka during the first 48 hours. It was accompanied by metabolic acidosis but not respiratory alkalosis (78).

 

CLINICAL FEATURES AND MANAGEMENT

 

Hypokalemia manifests as muscular cramps or weakness, constipation, abdominal bloating, polyuria, and sometimes cause life-threatening complications like arrhythmias, rhabdomyolysis, hypokalemic paralysis, diaphragmatic palsy, and respiratory failure (75). The treatment strategy is similar to that of hypokalemic periodic paralysis. Rebound hyperkalemia is a potential complication during recovery. Potassium should be replaced orally or intravenously, along with appropriate monitoring. Magnesium deficit should be corrected if present (79).

 

Hyperkalemia

 

ETIOPATHOGENESIS

 

Hyperkalemia can complicate envenomation by nose-horned viper (Vipera ammodytes ammodytes), European viper (Vipera berus), and hump-nosed viper (Hypnale hypnale) (53,80–83). Severe envenomation from these snakes causes hyperkalemia secondary to rhabdomyolysis and AKI, and can be fatal (53,80). Hyperkalemia was present in 7% of cases of SBE in 258 patients from Thailand (77).

 

Type 4 renal tubular acidosis (T4RTA) is another possible cause of hyperkalemia. It was described during the recovery phase of bite by hump-nosed viper (81,82). Renal biopsy from these patients showed tubular atrophy and focal segmental glomerulosclerosis pattern (81). The underlying cause could be thrombotic microangiopathy caused by toxins in venom leading to patchy cortical necrosis with delayed or partial recovery of renal functions (83).

 

CLINICAL FEATURES AND MANAGEMENT

 

Hyperkalemia associated with rhabdomyolysis and AKI can cause life-threatening arrhythmias. The presence of hyperkalemia along with hyperchloremic metabolic acidosis and low trans-tubular potassium gradient (TTKG) is suggestive of T4RTA and has been described in victims of hump-nosed viper bites during recovery from AKI. Fludrocortisone has been used successfully in such cases. T4RTA, in most cases, was transient (82). Hyperkalemia associated with rhabdomyolysis and AKI will require potassium lowering therapy and, in severe cases, dialysis.

Table 5. Summary of Snakebite Envenoming Induced Endocrine Dysfunctions

Endocrine manifestation

Pathophysiology

Onset of symptoms

Clinical features

Management

Acute hypopituitarism

Hemorrhagic infarction of the anterior pituitary (pathogenesis similar to Sheehan’s syndrome)

Hours to days

Hypotension not responding to standard therapy, hypoglycemia, hyponatremia

Glucocorticoid +/- thyroxine replacement

Delayed hypopituitarism

Sequalae of vascular insult to pituitary during acute phase

Months to years

Amenorrhea, hypogonadism, hypothyroidism, secondary adrenal insufficiency, growth hormone deficiency

Replacement of deficient hormones

Diabetes Insipidus

Very rare, possible vascular insult during acute phase

Immediate or delayed

Polyuria, polydipsia

Desmopressin

Adrenal insufficiency

Hemorrhage with or without infarction in the adrenals secondary to coagulopathy

Hours to days

Hypotension and circulatory collapse

Glucocorticoid +/- mineralocorticoid replacement

Hyperglycemia

Massive catecholamine surge

First 4-6 hours

Children more than adults

Standard treatment of hyperglycemia

Hyponatremia

Venom derived natriuretic peptides – renal salt wasting

First 2-3 days

Asymptomatic to varying alteration in sensorium to coma, seizures

Intravenous saline

 

 

Pituitary or adrenal insufficiency

Glucocorticoid replacement

Hypokalemia

Intracellular redistribution of potassium secondary to autonomic dysfunction.

Within first 24 hours

Muscle cramps, constipation, abdominal bloating, paralysis, respiratory failure, arrhythmia

Replacement of potassium with precaution to avoid rebound hyperkalemia

Venom-mediated renal tubular damage

Hyperkalemia

Venom mediated thrombotic microangiopathy in the kidneys leading to type 4 renal tubular acidosis

Weeks

 

 

 

 

Arrhythmia

 

 

Fludrocortisone

 

 

 

 

Rhabdomyolysis or kidney injury related

Days

Supportive, dialysis in severe cases

 

CONCLUSION

 

Endocrine dysfunctions associated with SBE are rare. However, missing the diagnosis can have life-threatening consequences. Acute or delayed anterior pituitary insufficiency is the most common manifestation. Establishing the diagnosis of hypocortisolism and timely glucocorticoid initiation in acute hypopituitarism are critical. Reports of adrenal dysfunction are scarce, though adrenal hemorrhage following RVE has been described more often in autopsy series. Electrolyte abnormalities should be anticipated and managed appropriately. Awareness and appropriate treatment of endocrine dysfunctions in resource-limited settings are necessary for optimal outcome.

 

REFERENCES

 

  1. Feasey N, Wansbrough-Jones M, Mabey DCW, Solomon AW. Neglected tropical diseases. Br Med Bull. 2010 Mar 1;93(1):179–200.
  2. Williams DJ, Faiz MA, Abela-Ridder B, Ainsworth S, Bulfone TC, Nickerson AD, et al. Strategy for a globally coordinated response to a priority neglected tropical disease: Snakebite envenoming. PLoS Negl Trop Dis. 2019 Feb 21;13(2):e0007059.
  3. Gutiérrez JM, Calvete JJ, Habib AG, Harrison RA, Williams DJ, Warrell DA. Snakebite envenoming. Nat Rev Dis Primer. 2017 Sep 14;3:17063.
  4. Bhattacharya S, Krishnamurthy A, Gopalakrishnan M, Kalra S, Kantroo V, Aggarwal S, et al. Endocrine and Metabolic Manifestations of Snakebite Envenoming. Am J Trop Med Hyg. 2020 Oct;103(4):1388–96.
  5. Tasoulis T, Isbister GK. A Review and Database of Snake Venom Proteomes. Toxins. 2017 18;9(9).
  6. Ferraz CR, Arrahman A, Xie C, Casewell NR, Lewis RJ, Kool J, et al. Multifunctional Toxins in Snake Venoms and Therapeutic Implications: From Pain to Hemorrhage and Necrosis. Front Ecol Evol. 2019 Jun 19;7:218.
  7. Wolff H. Insuficiência hipofisária anterior por picada de ofídio. Arq Bras Endocrinol Metab. 1958;7:25–47.
  8. Animal, Plant, and Microbial Toxins - Volume 1—Biochemistry | Akira Ohsaka | Springer [Internet]. [cited 2021 May 2]. Available from: https://www.springer.com/gp/book/9781468408881
  9. Myint-Lwin, Warrell DA, Phillips RE, Tin-Nu-Swe, Tun-Pe, Maung-Maung-Lay. Bites by Russell’s viper (Vipera russelli siamensis) in Burma: haemostatic, vascular, and renal disturbances and response to treatment. Lancet Lond Engl. 1985 Dec 7;2(8467):1259–64.
  10. Tun-Pe, Phillips RE, Warrell DA, Moore RA, Tin-Nu-Swe, Myint-Lwin, et al. Acute and chronic pituitary failure resembling Sheehan’s syndrome following bites by Russell’s viper in Burma. Lancet Lond Engl. 1987 Oct 3;2(8562):763–7.
  11. Golay V, Roychowdhary A, Dasgupta S, Pandey R. Hypopituitarism in patients with vasculotoxic snake bite envenomation related acute kidney injury: a prospective study on the prevalence and outcomes of this complication. Pituitary. 2014 Apr;17(2):125–31.
  12. Naik BN, Bhalla A, Sharma N, Mokta J, Singh S, Gupta P, et al. Pituitary dysfunction in survivors of Russell’s viper snake bite envenomation: A prospective study. Neurol India. 2018 Oct;66(5):1351–8.
  13. Bhat S, Mukhopadhyay P, Raychaudhury A, Chowdhury S, Ghosh S. Predictors of hypopituitarism due to vasculotoxic snake bite with acute kidney injury. Pituitary. 2019 Dec;22(6):594–600.
  14. Gopalakrishnan M, Vinod KV, Dutta TK, Shaha KK, Sridhar MG, Saurabh S. Exploring circulatory shock and mortality in viper envenomation: a prospective observational study from India. QJM Mon J Assoc Physicians. 2018 Nov 1;111(11):799–806.
  15. Rajagopala S, Thabah MM, Ariga KK, Gopalakrishnan M. Acute hypopituitarism complicating Russell’s viper envenomation: case series and systematic review. QJM Mon J Assoc Physicians. 2015 Sep;108(9):719–28.
  16. Antonypillai CN, Wass J a. H, Warrell DA, Rajaratnam HN. Hypopituitarism following envenoming by Russell’s vipers (Daboia siamensis and D. russelii) resembling Sheehan’s syndrome: first case report from Sri Lanka, a review of the literature and recommendations for endocrine management. QJM Mon J Assoc Physicians. 2011 Feb;104(2):97–108.
  17. Burke CW. The anterior pituitary, snakebite and Sheehan’s syndrome. Q J Med. 1990 Apr;75(276):331–3.
  18. Diri H, Karaca Z, Tanriverdi F, Unluhizarci K, Kelestimur F. Sheehan’s syndrome: new insights into an old disease. Endocrine. 2016 Jan;51(1):22–31.
  19. Kendre PP, Jose MP, Varghese AM, Menon JC, Joseph JK. Capillary leak syndrome in Daboia russelii bite-a complication associated with poor outcome. Trans R Soc Trop Med Hyg. 2018 Feb 1;112(2):88–93.
  20. Lingam TMC, Tan KY, Tan CH. Capillary leak syndrome induced by the venoms of Russell’s Vipers (Daboia russelii and Daboia siamensis) from eight locales and neutralization of the differential toxicity by three snake antivenoms. Comp Biochem Physiol Toxicol Pharmacol CBP. 2021 Sep 9;250:109186.
  21. Rucavado A, Escalante T, Camacho E, Gutiérrez JM, Fox JW. Systemic vascular leakage induced in mice by Russell’s viper venom from Pakistan. Sci Rep. 2018 Oct 31;8(1):16088.
  22. Hart GR, Proby C, Dedhia G, Yeo TH, Joplin GF, Burrin JM. Burmese Russell’s viper venom causes hormone release from rat pituitary cells in vitro. J Endocrinol. 1989 Aug;122(2):489–94.
  23. Than-Thannull, Francis N, Tin-Nu-Swe  null, Myint-Lwin  null, Tun-Pe  null, Soe-Soe  null, et al. Contribution of focal haemorrhage and microvascular fibrin deposition to fatal envenoming by Russell’s viper (Vipera russelli siamensis) in Burma. Acta Trop. 1989 Jan;46(1):23–38.
  24. Hung D-Z, Wu M-L, Deng J-F, Yang D-Y, Lin-Shiau S-Y. Multiple thrombotic occlusions of vessels after Russell’s viper envenoming. Pharmacol Toxicol. 2002 Sep;91(3):106–10.
  25. Das SK, Khaskil S, Mukhopadhyay S, Chakrabarti S. A patient of Russell’s viper envenomation presenting with cortical venous thrombosis: an extremely uncommon presentation. J Postgrad Med. 2013 Sep;59(3):235–6.
  26. Goswami R, Kochupillai N, Crock PA, Jaleel A, Gupta N. Pituitary autoimmunity in patients with Sheehan’s syndrome. J Clin Endocrinol Metab. 2002 Sep;87(9):4137–41.
  27. De Bellis A, Kelestimur F, Sinisi AA, Ruocco G, Tirelli G, Battaglia M, et al. Anti-hypothalamus and anti-pituitary antibodies may contribute to perpetuate the hypopituitarism in patients with Sheehan’s syndrome. Eur J Endocrinol. 2008 Feb;158(2):147–52.
  28. Kularatne S a. M. Epidemiology and clinical picture of the Russell’s viper (Daboia russelii russelii) bite in Anuradhapura, Sri Lanka: a prospective study of 336 patients. Southeast Asian J Trop Med Public Health. 2003 Dec;34(4):855–62.
  29. Hung D-Z, Wu M-L, Deng J-F, Lin-Shiau S-Y. Russell’s viper snakebite in Taiwan: differences from other Asian countries. Toxicon Off J Int Soc Toxinology. 2002 Sep;40(9):1291–8.
  30. Shivaprasad C, Aiswarya Y, Sridevi A, Anupam B, Amit G, Rakesh B, et al. Delayed hypopituitarism following Russell’s viper envenomation: a case series and literature review. Pituitary. 2019 Feb;22(1):4–12.
  31. Golay V, Roychowdhary A, Pandey R, Pasari A, Praveen M, Arora P, et al. Growth retardation due to panhypopituitarism and central diabetes insipidus following Russell’s viper bite. Southeast Asian J Trop Med Public Health. 2013 Jul 4;44(4):697–702.
  32. Ratnakaran B, Punnoose VP, Das S, Kartha A. Psychosis in Secondary Empty Sella Syndrome following a Russell’s Viper Bite. Indian J Psychol Med. 2016;38(3):254–6.
  33. Yerawar C, Punde D, Pandit A, Deokar P. Russell’s viper bite and the empty sella syndrome. QJM Mon J Assoc Physicians. 2021 Jul 28;114(4):255–7.
  34. Chung T-T, Koch CA, Monson JP. Hypopituitarism. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Oct 17]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK278989/
  35. Proby C, Tha-Aung, Thet-Win, Hla-Mon, Burrin JM, Joplin GF. Immediate and long-term effects on hormone levels following bites by the Burmese Russell’s viper. Q J Med. 1990 Apr;75(276):399–411.
  36. White J, Alfred S, Bates D, Mahmood MA, Warrell D, Cumming R, et al. Twelve month prospective study of snakebite in a major teaching hospital in Mandalay, Myanmar; Myanmar Snakebite Project (MSP). Toxicon X. 2019 Jan;1:100002.
  37. Hannoush ZC, Weiss RE. Pituitary Apoplexy. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Oct 27]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279125/
  38. Annane D, Pastores SM, Rochwerg B, Arlt W, Balk RA, Beishuizen A, et al. 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):2078–88.
  39. Higham CE, Johannsson G, Shalet SM. Hypopituitarism. The Lancet. 2016 Nov 12;388(10058):2403–15.
  40. Thein CM, Byard RW. Characteristics and relative numbers of lethal snake bite cases in medicolegal practice in central Myanmar - A five year study. J Forensic Leg Med. 2019 Apr;63:52–5.
  41. Krishnan MN, Kumar S, Ramamoorthy KP. Severe panhypopituitarism and central diabetes insipidus following snake bite: unusual presentation as torsades de pointes. J Assoc Physicians India. 2001 Sep;49:923–4.
  42. Gupta UC, Garg OP, Kataria ML. Cranial diabetes insipidus due to viper bite. J Assoc Physicians India. 1992 Oct;40(10):686–7.
  43. Anderson JR, Antoun N, Burnet N, Chatterjee K, Edwards O, Pickard JD, et al. Neurology of the pituitary gland. J Neurol Neurosurg Psychiatry. 1999 Jun 1;66(6):703–21.
  44. Christ-Crain M, Bichet DG, Fenske WK, Goldman MB, Rittig S, Verbalis JG, et al. Diabetes insipidus. Nat Rev Dis Primer. 2019 Aug 8;5(1):1–20.
  45. Garrahy A, Moran C, Thompson CJ. Diagnosis and management of central diabetes insipidus in adults. Clin Endocrinol (Oxf). 2019 Jan;90(1):23–30.
  46. Lakhotia M, Pahadiya HR, Singh J, Gandhi R, Bhansali S. Adrenal hematoma and right hemothorax after echis carinatus bite: an unusual manifestation. Toxicol Int. 2014 Dec;21(3):325–7.
  47. Senthilkumaran S, Menezes RG, Hussain SA, Luis SA, Thirumalaikolundusubramanian P. Russell’s Viper Envenomation-Associated Addisonian Crisis. Wilderness Environ Med. 2018 Dec;29(4):504–7.
  48. Fox B. Disseminated intravascular coagulation and the Waterhouse-Friderichsen syndrome. Arch Dis Child. 1971 Oct;46(249):680–5.
  49. Gagnon R. The venous drainage of the human adrenal gland. Rev Can Biol. 1956 Feb;14(4):350–9.
  50. Tan GXV, Sutherland T. Adrenal congestion preceding adrenal hemorrhage on CT imaging: a case series. Abdom Radiol N Y. 2016 Feb;41(2):303–10.
  51. Senthilkumaran S, Menezes RG, Hussain SA, Luis SA, Thirumalaikolundusubramanian P. Russell’s Viper Envenomation-Associated Addisonian Crisis. Wilderness Environ Med. 2018 Dec;29(4):504–7.
  52. Mao Y-C, Liu P-Y, Chiang L-C, Liao S-C, Su H-Y, Hsieh S-Y, et al. Bungarus multicinctus multicinctus Snakebite in Taiwan. Am J Trop Med Hyg. 2017 Jun 7;96(6):1497–504.
  53. Lukšić B, Culić V, Stričević L, Brizić I, Poljak NK, Tadić Z. Infant death after nose-horned viper (Vipera ammodytes ammodytes) bite in Croatia: A case report. Toxicon Off J Int Soc Toxinology. 2010 Dec;56(8):1506–9.
  54. Claudet I, Grouteau E, Cordier L, Franchitto N, Bréhin C. Hyperglycemia is a risk factor for high-grade envenomations after European viper bites (Vipera spp.) in children. Clin Toxicol Phila Pa. 2016;54(1):34–9.
  55. Kiran KM, More SS, Gadag JR. Biochemical and clinicopathological changes induced by Bungarus coeruleus venom in a rat model. J Basic Clin Physiol Pharmacol. 2004;15(3–4):277–87.
  56. Al-Saleh SSM. The effect of Echis carinatus crude venom and purified protein fractions on carbohydrate metabolism in rats. Cell Biochem Funct. 2002 Mar;20(1):1–10.
  57. Bouaziz M, Bahloul M, Kallel H, Samet M, Ksibi H, Dammak H, et al. Epidemiological, clinical characteristics and outcome of severe scorpion envenomation in South Tunisia: multivariate analysis of 951 cases. Toxicon Off J Int Soc Toxinology. 2008 Dec 15;52(8):918–26.
  58. Bahloul M, Turki O, Chaari A, Bouaziz M. Incidence, mechanisms and impact outcome of hyperglycaemia in severe scorpion-envenomed patients. Ther Adv Endocrinol Metab. 2018 Jul;9(7):199–208.
  59. Pande R, Khan H. Acute pancreatitis following adder bite in the UK: a case report. Ann R Coll Surg Engl. 2010 Sep;92(6):e25–6.
  60. Kjellström BT. Acute pancreatitis after snake bite. Case report. Acta Chir Scand. 1989 May;155(4–5):291–2.
  61. Kumar Keyal N, Shrestha R, Thapa S, Adhikari P. Krait Snake Bite Presenting as a Cerebral Salt Wasting. Indian J Crit Care Med Peer-Rev Off Publ Indian Soc Crit Care Med. 2019 Jul;23(7):347–8.
  62. Höjer J, Tran Hung H, Warrell D. Life-threatening hyponatremia after krait bite envenoming – A new syndrome. Clin Toxicol. 2010 Nov 1;48(9):956–7.
  63. Hung HT, Höjer J, Du NT. Clinical features of 60 consecutive ICU-treated patients envenomed by Bungarus multicinctus. Southeast Asian J Trop Med Public Health. 2009 May;40(3):518–24.
  64. Trinh KX, Khac QL, Trinh LX, Warrell DA. Hyponatraemia, rhabdomyolysis, alterations in blood pressure and persistent mydriasis in patients envenomed by Malayan kraits (Bungarus candidus) in southern Viet Nam. Toxicon Off J Int Soc Toxinology. 2010 Nov;56(6):1070–5.
  65. Tongpoo A, Sriapha C, Pradoo A, Udomsubpayakul U, Srisuma S, Wananukul W, et al. Krait envenomation in Thailand. Ther Clin Risk Manag. 2018;14:1711–7.
  66. de Silva U, Sarathchandra C, Senanayake H, Pilapitiya S, Siribaddana S, Silva A. Hyponatraemia and seizures in Merrem’s hump-nosed pit viper (Hypnale hypnale) envenoming: a case report. J Med Case Reports. 2018 Aug 2;12(1):213.
  67. van der Walt AJ, Muller GJ. Berg adder (Bitis atropos) envenoming: an analysis of 14 cases. Clin Toxicol Phila Pa. 2019 Feb;57(2):131–6.
  68. Wium CA, Marks CJ, Du Plessis CE, Müller GJ. Berg adder (Bitis atropos): An unusual case of acute poisoning. South Afr Med J Suid-Afr Tydskr Vir Geneeskd. 2017 Nov 27;107(12):1075–7.
  69. Vink S, Jin AH, Poth KJ, Head GA, Alewood PF. Natriuretic peptide drug leads from snake venom. Toxicon Off J Int Soc Toxinology. 2012 Mar 15;59(4):434–45.
  70. Trinh KX, Khac QL, Trinh LX, Warrell DA. Hyponatraemia, rhabdomyolysis, alterations in blood pressure and persistent mydriasis in patients envenomed by Malayan kraits (Bungarus candidus) in southern Viet Nam. Toxicon Off J Int Soc Toxinology. 2010 Nov;56(6):1070–5.
  71. Tongpoo A, Sriapha C, Pradoo A, Udomsubpayakul U, Srisuma S, Wananukul W, et al. Krait envenomation in Thailand. Ther Clin Risk Manag. 2018;14:1711–7.
  72. Spasovski G, Vanholder R, Allolio B, Annane D, Ball S, Bichet D, et al. Clinical practice guideline on diagnosis and treatment of hyponatraemia. Eur J Endocrinol. 2014 Mar;170(3):G1-47.
  73. Gawarammana IB, Mudiyanselage Kularatne SA, Kularatne K, Waduge R, Weerasinghe VS, Bowatta S, et al. Deep coma and hypokalaemia of unknown aetiology following Bungarus caeruleus bites: Exploration of pathophysiological mechanisms with two case studies. J Venom Res. 2010 Dec 14;1:71–5.
  74. Kularatne S a. M. Common krait (Bungarus caeruleus) bite in Anuradhapura, Sri Lanka: a prospective clinical study, 1996-98. Postgrad Med J. 2002 May;78(919):276–80.
  75. Jeevagan V, Katulanda P, Gnanathasan CA, Warrell DA. Acute pituitary insufficiency and hypokalaemia following envenoming by Russell’s viper (Daboia russelii) in Sri Lanka: Exploring the pathophysiological mechanisms. Toxicon Off J Int Soc Toxinology. 2013 Mar 1;63:78–82.
  76. Malina T, Krecsák L, Jelić D, Maretić T, Tóth T, Siško M, et al. First clinical experiences about the neurotoxic envenomings inflicted by lowland populations of the Balkan adder, Vipera berus bosniensis. Neurotoxicology. 2011 Jan;32(1):68–74.
  77. Aye K-P, Thanachartwet V, Soe C, Desakorn V, Thwin K-T, Chamnanchanunt S, et al. Clinical and laboratory parameters associated with acute kidney injury in patients with snakebite envenomation: a prospective observational study from Myanmar. BMC Nephrol. 2017 Mar 16;18(1):92.
  78. Sellahewa K. Lessons from four studies on the management of snake bite in Sri Lanka. Ceylon Med J. 1997 Mar;42(1):8–15.
  79. Kardalas E, Paschou SA, Anagnostis P, Muscogiuri G, Siasos G, Vryonidou A. Hypokalemia: a clinical update. Endocr Connect. 2018 Apr;7(4):R135–46.
  80. Denis D, Lamireau T, Llanas B, Bedry R, Fayon M. Rhabdomyolysis in European viper bite. Acta Paediatr Oslo Nor 1992. 1998 Sep;87(9):1013–5.
  81. Weerakkody RM, Lokuliyana PN, Lanerolle RD. Transient distal renal tubular acidosis following hump nosed viper bite: Two cases from Sri Lanka. Saudi J Kidney Dis Transplant. 2016 Sep 1;27(5):1018.
  82. Karunarathne S, Udayakumara Y, Govindapala D, Fernando H. Type IV renal tubular acidosis following resolution of acute kidney injury and disseminated intravascular coagulation due to hump-nosed viper bite. Indian J Nephrol. 2013;23(4):294–6.
  83. Herath N, Wazil A, Kularatne S, Ratnatunga N, Weerakoon K, Badurdeen S, et al. Thrombotic microangiopathy and acute kidney injury in hump-nosed viper (Hypnale species) envenoming: a descriptive study in Sri Lanka. Toxicon Off J Int Soc Toxinology. 2012 Jul;60(1):61–5.

 

Vitamin D: Production, Metabolism, and Mechanisms of Action

ABSTRACT

Vitamin D production in the skin under the influence of sunlight (UVB) is maximized at levels of sunlight exposure that do not burn the skin. Further metabolism of vitamin D to its major circulating form (25(OH)D) and hormonal form (1,25(OH)2D) takes place in the liver and kidney, respectively, but also in other tissues where the 1,25(OH)2D produced serves a paracrine/autocrine function: examples include the skin, cells of the immune system, parathyroid gland, intestinal epithelium, prostate, and breast. Parathyroid hormone, FGF23, calcium and phosphate are the major regulators of the renal 1-hydroxylase (CYP27B1, the enzyme producing 1,25(OH)2D); regulation of the extra renal 1-hydroxylase differs from that in the kidney and involves cytokines. The major enzyme that catabolizes 25(OH)D and 1,25(OH)2D is the 24-hydroxylase; like the 1-hydroxylase it is tightly controlled in the kidney in a manner opposite to that of the 1-hydroxylase, but like the 1-hydroxylase it is widespread in other tissues where its regulation is different from that of the kidney. Vitamin D and its metabolites are carried in the blood bound to vitamin D binding protein (DBP) and albumin--for most tissues it is the free (i.e., unbound) metabolite that enters the cell; however, DBP bound metabolites can enter some cells such as the kidney and parathyroid gland through a megalin/cubilin mechanism. Most but not all actions of 1,25(OH)2D are mediated by the vitamin D receptor (VDR). VDR is a transcription factor that partners with other transcription factors such as retinoid X receptor that when bound to 1,25(OH)2D regulates gene transcription either positively or negatively depending on other cofactors to which it binds or interacts.  The VDR is found in most cells, not just those involved with bone and mineral homeostasis (i.e., bone, gut, kidney) resulting in wide spread actions of 1,25(OH)2D on most physiologic and pathologic processes. Animal studies indicate that vitamin D has beneficial effects on various cancers, blood pressure, heart disease, immunologic disorders, but these non-skeletal effects have been difficult to prove in humans in randomized controlled trials. Analogs of 1,25(OH)2D are being developed to achieve specificity for non-skeletal target tissues such as the parathyroid gland and cancers to avoid the hypercalcemia resulting from 1,25(OH)2D itself. The level of vitamin D intake and achieved serum levels of 25(OH)D that are optimal and safe for skeletal health and the non-skeletal actions remain controversial, but are likely between an intake of 800-2000IU vitamin D in the diet and 20-50ng/ml 25(OH)D in the blood.

OVERVIEW

Rickets became a public health problem with the movement of the population from the farms to the cities during the Industrial Revolution. Various foods such as cod liver oil and irradiation of other foods including plants were found to prevent or cure this disease, leading eventually to the discovery of the active principle—vitamin D. Vitamin D comes in two forms (D2 and D3) which differ chemically in their side chains. These structural differences alter their binding to the carrier protein vitamin D binding protein (DBP) and their metabolism, but in general the biologic activity of their active metabolites is comparable. Vitamin D3 is produced in the skin from 7-dehydrocholesterol by UV irradiation, which breaks the B ring to form pre-D3. Pre-D3 isomerizes to D3 but with continued UV irradiation to tachysterol and lumisterol. D3 is preferentially removed from the skin, bound to DBP. The liver and other tissues metabolize vitamin D, whether from the skin or oral ingestion, to 25OHD, the principal circulating form of vitamin D. Several enzymes have 25-hydroxylase activity, but CYP2R1 is the most important. 25OHD is then further metabolized to 1,25(OH)2D principally in the kidney, by the enzyme CYP27B1, although other tissues including various epithelial cells, cells of the immune system, and the parathyroid gland contain this enzyme. 1,25(OH)2D is the principal hormonal form of vitamin D, responsible for most of its biologic actions. The production of 1,25(OH)2D in the kidney is tightly controlled, being stimulated by parathyroid hormone (PTH), and inhibited by calcium, phosphate and FGF23. Extrarenal production of 1,25(OH)2D as in keratinocytes and macrophages is under different control, being stimulated primarily by cytokines such as tumor necrosis factor alfa (TNFα) and interferon gamma (IFNg). 1,25(OH)2D reduces 1,25(OH)2D levels in cells primarily by stimulating its catabolism through the induction of CYP24A1, the 24-hydroxylase.  25OHD and 1,25(OH)2D are hydroxylated in the 24 position by this enzyme to form 24,25(OH)2D and 1,24,25(OH)3D, respectively.  This 24-hydroxylation is generally the first step in the catabolism of these active metabolites to the final end product of calcitroic acid, although 24,25(OH)2D and 1,24,25(OH)3D have their own biologic activities. CYP24A1 also has 23-hydroxylase activity that leads to a different end product. Different species differ in their ratio of 23-hydroxylase/24-hydroxyase activity in their CYP24A1 enzyme, but in humans the 24-hydroxyase activity predominates. Like CYP27B1, CYP24A1 is widely expressed. CYP24A1 is induced by 1,25(OH)2D in most tissues, which serves as an important feedback mechanism to avoid vitamin D toxicity. In the kidney, PTH inhibits CYP24A1, whereas FGF23, calcium and phosphate stimulates it, just the opposite of the actions of these hormones and minerals on CYP27B1. However, such regulation is not seen in other tissues.  In macrophages, CYP24A1 is either missing or defective, so in situations such as granulomatous diseases like sarcoidosis in which macrophage production of 1,25(OH)2D is increased, hypercalcemia and hypercalciuria due to elevated 1,25(OH)2D can occur without the counter regulation by CYP24A1.

The vitamin D metabolites are transported in blood bound to DBP and albumin. Very little circulates as the free form. The liver produces DBP and albumin, production that is decreased in liver disease, and these proteins may be lost in protein losing enteropathies or the nephrotic syndrome. Thus, individuals with liver, intestinal or renal diseases which result in low levels of these transport proteins may have low total levels of the vitamin D metabolites without necessarily being vitamin D deficient as their free concentrations may be normal.

The receptor for 1,25(OH)2D (VDR) is a transcription factor regulating the expression of genes which mediate its biologic activity. VDR is a member of a rather large family of nuclear hormone receptors which includes the receptors for glucocorticoids, mineralocorticoids, sex hormones, thyroid hormone, and vitamin A metabolites or retinoids. The VDR is widely distributed, and is not restricted to those tissues considered the classic target tissues of vitamin D. The VDR upon binding to 1,25(OH)2D heterodimerizes with other nuclear hormone receptors, in particular the family of retinoid X receptors. This complex then binds to special DNA sequences called vitamin D response elements (VDRE) generally within the genes it regulates, although these VDREs can be thousands of base pairs from the transcription start site. There are thousands of the VDREs in hundreds of genes, and the profile of active VDREs (and regulated genes) varies from cell to cell.  A variety of additional proteins called coregulators complex with the VDR to activate (coactivators) or inhibit (corepressors) VDR transcriptional activity. Coactivator factors involved in VDR mediated transcription include factors with histone acetylase activity, including steroid receptor coactivator (SRC) 1, SRC 2 and SRC 3, and CREB-binding protein p300, in addition to the SWI–SNF ATP dependent chromatin remodeling complex, methyltransferases and the Mediator complex (aka DRIP), which functions to recruit RNA polymerases. VDR binding sites are associated with sites for other transcription factors such as p63, C–EBPα, C–EBPβ, Runx2 and PU.1, which can cooperate with VDR and VDR coregulators to influence 1,25(OH)2D responses in target cells. Among other functions these coregulators reconfigure the chromatin structure to bring the VDR/VDRE to the transcription start site, explaining how such distant VDR/VDREs can regulate gene transcription.  In addition to coactivators there are a number of corepressors. One such corepressor of VDR action in the skin is called hairless, in that its loss or mutation, like that of the VDR, leads to altered hair follicle cycling resulting in baldness. Corepressors typically work by recruiting histone deacetylases (HDAC) or methyl transferases (MT) to the gene which reverses the actions of HAT, leading to a reduction in access to the gene by the transcription machinery. These coregulators can be specific for different genes, and different cells differentially express these coregulators, providing some specificity for the actions of 1,25(OH)2D and VDR.

In addition to regulating gene expression, 1,25(OH)2D has a number of non-genomic actions including the ability to stimulate calcium transport across the plasma membrane. The mechanisms mediating these non-genomic actions and their physiologic significance remain unclear. Similarly, it is not clear that all actions of the VDR require the ligand 1,25(OH)2D. The best example of this is the hair loss in animals and subjects with VDR mutations but not in animals and subjects with mutations in CYP27B1, the enzyme producing 1,25(OH)2D. As mentioned, the VDR is widely distributed, and the actions of 1,25(OH)2D are quite varied. The classic target tissues—bone, gut, and kidney—are involved with calcium homeostasis. The mechanisms by which 1,25(OH)2D regulates transcellular calcium transport are best understood in the intestine. Here 1,25(OH)2D stimulates calcium entry across the brush border membrane into the cell, transport of calcium through the cell, and removal of calcium from the cell at the basolateral membrane. Calcium entry at the brush border membrane occurs down a steep electrochemical gradient. It is controlled in large measure by a specific calcium channel called TRPV6 and in humans also by a homologous calcium channel TRPV5. Transport of calcium through the cell is regulated by a class of calcium binding proteins called calbindins. Much of the transport occurs within vesicles that form in the terminal web. Removal of calcium from the cell at the basolateral membrane requires energy and is mediated by the ATP requiring calcium pump or CaATPase (PMCA1b) as well as the sodium/calcium exchange protein (NCX1). 1,25(OH)2D induces TRPV6 and TRPV5, the calbindins, and the CaATPase, but not all aspects of transcellular calcium transport are a function of new protein synthesis. Animals null for calbindin 9k (the major calbindin in mammalian intestine) have little impairment of intestinal calcium transport. Animals null for TRPV6, on the other hand, have a reduction in intestinal calcium transport, but the deficit is not profound. Thus, it is likely that compensatory mechanisms for intestinal calcium transport exist that have yet to be discovered.  Similar mechanisms mediate 1,25(OH)2D regulated calcium reabsorption in the distal tubule of the kidney. The proteins involved are homologous but not identical (TRPV5 and Calbindin 28k, for example). The situation in bone, however, is less clear. VDR are found in osteoblasts, the bone forming cells. 1,25(OH)2D promotes the differentiation of osteoblasts and regulates the production of proteins such as collagen, alkaline phosphatase, and osteocalcin thought to be important in bone formation. 1,25(OH)2D also induces RANKL, a membrane bound protein in osteoblasts that enables osteoblasts to stimulate the formation and activity of osteoclasts. Thus 1,25(OH)2D regulates both bone formation and bone resorption. Some evidence suggests that the major effect of 1,25(OH)2D on bone is to provide adequate levels of calcium and phosphate from the intestine. The rickets of patients with a mutated VDR or of mice in which the VDR has been deleted can be prevented/corrected by normalizing serum calcium and phosphate levels by dietary means. On the other hand, normal bone formation is not restored, and with time the VDR null mice become osteoporotic despite the high calcium/phosphate diet. Moreover, the VDR in osteoblasts/osteocytes appears to control bone resorption especially when dietary calcium is limited. Whether subjects with VDR mutations also develop osteoporosis prematurely or fail to maintain serum calcium in times of calcium deficiency has not been reported.

The non-classic actions of 1,25(OH)2D include regulation of cellular proliferation and differentiation, regulation of hormone secretion, and regulation of immune function. The ability of 1,25(OH)2D to inhibit proliferation and stimulate differentiation has led to the development of a number of analogs in the hopes of treating hyperproliferative disorders such as psoriasis and cancer without raising serum calcium. Psoriasis is now successfully treated with several vitamin D analogs. Observational studies are promising with respect to adequate vitamin D nutrition and cancer prevention. However, supplementation with vitamin D of subjects with adequate vitamin D levels to start with has not been shown to decrease cancer incidence but may be beneficial for cancer mortality. 1,25(OH)2D inhibits parathyroid hormone secretion and stimulates insulin secretion. A number of analogs and 1,25(OH)2D itself are currently available for use in the treatment of secondary hyperparathyroidism accompanying renal failure. Epidemiologic evidence indicates that vitamin D deficiency is associated with increased risk of both type 1 and type 2 diabetes mellitus, but prospective clinical trials to demonstrate a role for vitamin D supplementation in preventing the conversion of prediabetes to diabetes has not shown benefit in vitamin D replete individuals. However, there may be benefit in vitamin D deficient patients.  The ability of 1,25(OH)2D to regulate immune function is likely part of its efficacy in the treatment of psoriasis. A number of other autoimmune diseases have been found in animal studies to respond favorably to vitamin D and 1,25(OH)2D or its analogs, and epidemiologic evidence linking vitamin D deficiency to increased incidence of these diseases has been reported. Similarly, epidemiologic evidence linking vitamin D deficiency to a number of respiratory illnesses is substantial, including increased risk of COVID-19 infections.

DISCOVERY

The first clear description of rickets was by Whistler (1) in 1645. However, it was not until the Industrial Revolution with the mass movement of the population from the farms to the smoke- filled cities that rickets became a public health problem, most notably in England where sunlight intensity was already marginal for much of the year. Mellanby (2) in Great Britain and McCollum (3) in the United States developed animal models for rickets and showed that rickets could be cured with cod liver oil. McCollum heated the cod liver oil to destroy its vitamin A content and found that it still had antirachitic properties; he named the antirachitic factor vitamin D. Steenbock and Black (4) then demonstrated that UV irradiation of food, in particular non saponifiable lipids, could treat rickets. Meanwhile, clinical investigations revealed that rickets could be prevented or cured in children with sunlight or artificial UV exposure (5,6) suggesting that what subsequently became known as vitamin D could be produced by irradiation of precursors in vivo. Ultimately, Askew et al. (7) isolated and determined the structure of vitamin D2 (ergocalciferol) from irradiated plant sterols (ergosterol), and Windaus et al. (8) determined the structures and pathway by which 7-dehydrocholesterol (7-DHC) in the skin is converted to vitamin D3 (cholecalciferol). The name vitamin D1 refers to what proved to be an error of an earlier identification, and is not used. The structures and pathways of production of vitamin D3 are shown in figure 1. The structures of vitamins D2 and D3 differ in the side chain where D2 contains a double bond (C22-23) and an additional methyl group attached to C24. In this chapter the designation of D will refer to both D3 and D2.

Figure 1. The production of vitamin D3 from 7-dehydrocholesterol in the epidermis. Sunlight (the ultraviolet B component) breaks the B ring of the cholesterol structure to form pre- D3. Pre-D3 then undergoes a thermal induced rearrangement to form D3. Continued irradiation of pre- D3 leads to the reversible formation of lumisterol3 and tachysterol3 which can revert back to pre-D3 in the dark.

Figure 2. The metabolism of vitamin D. The liver converts vitamin D to 25OHD. The kidney converts 25OHD to 1,25(OH)2D and 24,25(OH)2D. Other tissues contain these enzymes, but the liver is the main source for 25-hydroxylation, and the kidney is the main source for 1α-hydroxylation. Control of metabolism of vitamin D to its active metabolite, 1,25(OH)2D, is exerted primarily at the renal level where calcium, phosphorus, parathyroid hormone, FGF23, and 1,25(OH)2D regulate the levels of 1,25(OH)2D produced.

 METABOLISM

Vitamin D3 produced in the epidermis must be further metabolized to be active. The first step, 25-hydroxylation, takes place primarily in the liver, although other tissues have this enzymatic activity as well. As will be discussed below, there are several 25-hydroxylases. 25OHD is the major circulating form of vitamin D. However, in order for vitamin D metabolites to achieve maximum biologic activity they must be further hydroxylated in the 1α position by the enzyme CYP27B1; 1,25(OH)2D is the most potent metabolite of vitamin D and accounts for most of its biologic actions. The 1α hydroxylation occurs primarily in the kidney, although as for the 25-hydroxylase, other tissues have this enzyme. Vitamin D and its metabolites, 25OHD and 1,25(OH)2D, can also be hydroxylated in the 24 position. This may serve to activate the metabolite or analog as 1,25(OH)2D and 1,24(OH)2D have similar biologic potency, and 1,24,25(OH)3D has activity approximately 1/10 that of 1,25(OH)2D. However, 24-hydroxylation of metabolites with an existing 25OH group leads to further catabolism. The details of these reactions are described below.

Cutaneous Production of Vitamin D3

The precursor of vitamin D, 7-dehydrocholesterol (7-DHC) is on the Kandutsch-Russell cholesterol pathway. The final enzymatic reaction mediated by 7-dehyrocholesterol reductase converting 7-DHC to cholesterol is regulated by a number of factors including vitamin D and cholesterol which enhance its degradation thus enabling increased levels of 7-DHC for conversion to vitamin D (9). Although irradiation of 7-DHC was known to produce pre-D3 (which subsequently undergoes a temperature rearrangement of the triene structure to form D3), lumisterol, and tachysterol (figure 1), the physiologic regulation of this pathway was not well understood until the studies of Holick and his colleagues (10-12). They demonstrated that the formation of pre-D3 under the influence of solar or UV irradiation (maximal effective wavelength between 290-310) is relatively rapid and reaches a maximum within hours. UV irradiation further converts pre-D3 to lumisterol and tachysterol. Both the degree of epidermal pigmentation and the intensity of exposure correlate with the time required to achieve this maximal concentration of pre-D3, but do not alter the maximal level achieved. Although pre-D3 levels reach a maximum level, the biologically inactive lumisterol continues to accumulate with continued UV exposure. Tachysterol is also formed, but like pre-D3, does not accumulate with extended UV exposure. The formation of lumisterol is reversible and can be converted back to pre-D3 as pre-D3 levels fall. At 0oC, no D3 is formed; however, at 37oC pre-D3 is slowly converted to D3. Thus, short exposure to sunlight would be expected to lead to a prolonged production of D3 in the exposed skin because of the slow thermal conversion of pre-D3 to D3 and the conversion of lumisterol to pre-D3. Prolonged exposure to sunlight would not produce toxic amounts of D3 because of the photoconversion of pre-D3 to lumisterol and tachysterol as well as the photoconversion of D3 itself to suprasterols I and II and 5,6 transvitamin D3 (13).

Melanin in the epidermis, by absorbing UV irradiation, can reduce the effectiveness of sunlight in producing D3 in the skin. This may be one important reason for the lower 25OHD levels (a well-documented surrogate measure for vitamin D levels in the body) in Blacks and Hispanics living in temperate latitudes (14). Sunlight exposure increases melanin production, and so provides another mechanism by which excess D3 production can be prevented. The intensity of UV irradiation is also important for effective D3 production. The seasonal variation of 25OHD levels can be quite pronounced with higher levels during the summer months and lower levels during the winter. The extent of this seasonal variation depends on the latitude, and thus the intensity of the sunlight striking the exposed skin. In Edmonton, Canada (52oN) very little D3 is produced in exposed skin from mid-October to mid-April; Boston (42oN) has a somewhat longer period for effective D3 production; whereas in Los Angeles (34oN) and San Juan (18oN) the skin is able to produce D3 all year long (15). These findings apply to sea level. At higher elevations there is less atmospheric absorption of UVB, so that skiers can make vitamin D even in winter on sunny days. Peak D3 production occurs around noon, with a larger portion of the day being capable of producing D3 in the skin during the summer than other times of the year. Clothing (16) and sunscreens (17) effectively prevent D3 production in the covered areas. This is one likely explanation for the observation that the Bedouins in the Middle East, who totally cover their bodies with clothing, are more prone to develop rickets and osteomalacia than the Israeli Jews with comparable sunlight exposure.

Hepatic Production of 25OHD

The next step in the bioactivation of D2 and D3, hydroxylation to 25OHD, takes place primarily in the liver although a number of other tissues express this enzymatic activity. 25OHD is the major circulating form of vitamin D and provides a clinically useful marker for vitamin D status. DeLuca and colleagues were the first to identify 25OHD and demonstrate its production in the liver over 30 years ago, but ambiguity remains as to the actual enzyme(s) responsible for this activity. 25-hydroxylase activity has been found in both the liver mitochondria and endoplasmic reticulum, and the enzymatic activities appear to differ indicating different proteins. At this point most attention has been paid to the mitochondrial CYP27A1 and the microsomal CYP2R1. However, in mouse knockout studies and in humans with mutations in these enzymes, only CYP2R1 loss is associated with decreased 25OHD levels (18,19). However, deletion or mutation of CYP2R1 does not totally eliminate 25OHD production These are mixed function oxidases, but differ in apparent Kms and substrate specificities. 

The mitochondrial 25-hydroxylase is now well accepted as CYP27A1, an enzyme first identified as catalyzing a critical step in the bile acid synthesis pathway. This is a high capacity, low affinity enzyme consistent with the observation that 25-hydroxylation is not generally rate limiting in vitamin D metabolism. Although initial studies suggested that the vitamin D3-25-hydroxylase and cholestane triol 27-hydroxyase activities in liver mitochondria were due to distinct enzymes with differential regulation, the cloning of CYP27A1 and the demonstration that it contained both activities has put this issue to rest (20-22). CYP27A1 is widely distributed throughout different tissues with highest levels in liver and muscle, but also in kidney, intestine, lung, skin, and bone (20-23).  Mutations in CYP27A1 lead to cerebrotendinous xanthomatosis (24,25), and are associated with abnormal vitamin D and/or calcium metabolism in some but not all of these patients (25-27).  However, mice in which CYP27A1 is deleted actually have elevated 25OHD levels along with the disruption in bile acid synthesis (28). CYP27A1 can hydroxylate vitamin D and related compounds at the 24, 25, and 27 positions. However, D2 appears to be preferentially 24-hydroxylated, whereas D3 is preferentially 25-hydroxylated (29). The 1αOH derivatives of D are more rapidly hydroxylated than the parent compounds (30). These differences between D2 and D3 and their 1αOH derivatives may explain the differences in biologic activity between D2 and D3 or between 1αOHD2 and 1αOHD3.

The major microsomal 25-hydroxylase is CYP2R1, although other enzymes have been shown in in vitro studies to have 25-hydroxylase activity. This enzyme like that of CYP27A1 is widely distributed, although it is most abundantly expressed in liver, skin and testes (30). Unlike CYP27A1, CYP2R1 25-hydroxylates D2 and D3 equally (30). Several Nigerian families have been shown to have CYP2R1 mutations in family members with rickets (19,31).  These subjects respond to D therapy but suboptimally (19,31). Mice lacking CYP2R1 have reduced 25OHD levels, unlike mice lacking CYP27A1, but even the combined deletion of CYP2R1 and CYP27A1 does not reduce these levels more than about 70% (18). Thus, neither CYP27A1 nor CYP2R1 by themselves account for all 25-hydroxyase activity in the body, suggesting a role of other yet to be described 25-hydroxylases.

Studies of the regulation of 25-hydroxylation have not been completely consistent, most likely because of the initial failure to appreciate that at least two enzymatic activities were involved and because of species differences. In general, 25-hydroxylation in the liver is little affected by vitamin D status. However, CYP27A1 expression in the intestine (32) and kidney (33) is reduced by 1,25(OH)2D. Not surprisingly bile acids decrease CYP27A1 expression (34) as does insulin (35) through an unknown mechanism. Dexamethasone, on the other hand, increases CYP27A1 expression (36). CYP2R1 appears to be mediated by aspects of metabolism. Roizen et al. (37) found that the serum concentration of 25OHD, but not vitamin D, was decreased in mice fed a high fat diet to induce obesity compared with normal weight mice. Moreover, mRNA and protein levels of CYP2R1 were decreased in these obese mice.  The expression of other 25-hydroxylases (CYP27A1, CYP3A) or the catabolizing enzyme CYP24A1 was not altered. Aatsinki et al (38) examined the effect of high fat diet induced obesity, fasting, and type 2 diabetes as well as streptozotocin induced (type 1) diabetes on 25OHD levels in mice.  All these metabolic manipulations decreased the hepatic mRNA and protein concentration of CYP2R1. These authors then demonstrated that the decrease in CYP2R1 was mediated by PPARγ-coactivator-1α (PGC1α), a key metabolic regulator increased by fasting or diabetes. They then showed that the control of CYP2R1 gene expression by PGC1α involved another transcriptional regulator, estrogen-related receptor α (ERRα), which also binds to other nuclear receptors such as VDR and the glucocorticoid receptor (GR). Consistent with this is that dexamethasone, a ligand for GR, decreased hepatic CYP2R1 mRNA and protein concentrations by a mechanism mediated by increased PGC1α.

Renal Production of 1,25(OH)2D

1,25(OH)2D is the most potent metabolite of vitamin D, and mediates most of its hormonal actions. 1,25(OH)2D is produced from 25OHD by the enzyme 25OHD-1α hydroxylase (CYP27B1). The cloning of CYP27B1 by four independent groups (40-43) ended a long effort to determine the structure of this critical enzyme in vitamin D metabolism. Mutations in this gene are responsible for the rare autosomal disease of pseudovitamin D deficiency rickets (40,42,44,45). An animal model in which the gene is knocked out by homologous recombination reproduces the clinical features of this disease including retarded growth, rickets, hypocalcemia, hyperparathyroidism, and undetectable 1,25(OH)2D (46). Unlike Vdr null mice and VDR mutations in humans, alopecia is not part of this phenotype.

CYP27B1 is a mitochondrial mixed function oxidase with significant homology to other mitochondrial steroid hydroxylases including CYP27A1 (39%), CYP24A1 (30%), CYP11A1 (32%), and CYP11β (33%) (40). However, within the heme-binding domain the homology is much greater with 73% and 65% sequence identity with CYP27A1 and CYP24A1 (40). These mitochondrial P450 enzymes are located in the inner membrane of the mitochondrion, and serve as the terminal acceptor for electrons transferred from NADPH through ferrodoxin reductase and ferrodoxin. Expression of CYP27B1 is highest in epidermal keratinocytes (40), cells that previously had been shown to contain high levels of this enzymatic activity (47). However, the kidney also expresses this enzyme in the renal tubules as do the brain, placenta, testes, intestine, lung, breast, macrophages, lymphocytes, parathyroid gland, osteoblasts and chondrocytes (40,48-51). That said, the kidney is generally considered the major source of circulating levels of 1,25(OH)2D, with the extrarenal CYP27B1 activities providing for local needs under normal circumstances. However, extrarenal sources can lead to increased 1,25(OH)2D and calcium levels in some pathologic conditions to be discussed subsequently.

The principal regulators of CYP27B1 activity in the kidney are parathyroid hormone (PTH), FGF23, calcium, phosphate, and 1,25(OH)2D. Extrarenal production tends to be stimulated by cytokines such as IFN-gamma and TNF-α more effectively than PTH (52) and may be less inhibited by calcium, phosphate, and 1,25(OH)2D depending on the tissue. Administration of PTH in vivo (53) or in vitro (54,55) stimulates renal production of 1,25(OH)2D. This action of PTH can be mimicked by cAMP (53,55) and forskolin (56,57) indicating that at least part of the effect of PTH is mediated via its activation of adenylate cyclase. However, PTH activation of protein kinase C (PKC) also appears to be involved in that concentrations of PTH sufficient to stimulate PKC activation and 1,25(OH)2D production are below that required to increase cAMP levels (58). Furthermore, synthetic fragments of PTH lacking the ability to activate adenylate cyclase but which stimulate PKC activity were found to increase 1,25(OH)2D production (59). Direct activation of PKC with phorbol esters results in increased 1,25(OH)2D production. Although the promoter of CYP27B1 contains several AP-1 (PKC activated) and cAMP response elements, it is not yet clear how PTH regulates CYP27B1 gene expression (60). However, several mechanisms have been proposed. In one study the nuclear receptor 4A2 acting through a C/EBP consensus element appears to be involved (61). Another mechanism involves VDIR that is proposed to bind to a negative VDRE in the CYP27B1 promoter. When PKA is activated by PTH VDIR is phosphorylated and recruits the p300 complex with HAT activity, inducing gene transcription (62). Calcium modulates the ability of PTH to increase 1,25(OH)2D production. Calcium by itself can decrease CYP27B1activity (63,64) and block the stimulation by PTH (65). Given in vivo, calcium can exert its effect in part by reducing PTH secretion, but this does not explain its direct actions in vitro or its effects in parathyroidectomized or PTH infused animals. Phosphate deprivation can stimulate CYP27B1 activity in vivo (66,67) and in vitro (68). The in vivo actions of phosphate deprivation can be blocked by hypophysectomy (69,70) and partially restored by growth hormone (GH) (70,71) and insulin-like growth factor (IGF-I) (72). However, like PTH, the exact mechanism by which GH and/or IGF-I mediates the effects of phosphate on CYP27B1 expression remains unclear. More recently FGF23 has been shown to inhibit CYP27B1 activity in vivo and in vitro (73). FGF23 has been implicated as at least one of the factors responsible for impaired phosphate reabsorption and 1,25(OH)2D production in conditions such as X-linked and autosomal dominant hypophosphatemic rickets and oncogenic osteomalacia (74,75). FGF23 acts through FGF receptors 1 and 3 in conjunction with the coreceptor Klotho, but the mechanism by which FGF23 regulates CYP27B1 remains obscure. High phosphate stimulates FGF23 production from bone, and this is likely the major mechanism by which phosphate leads to decreased CYP27B1 activity (76).  1,25(OH)2D administration leads to reduction in CYP27B1 activity. In the kidney Meyer et al. (77) identified a region in the Cyp27b1 gene that when deleted blocked 1,25(OH)2D production. However, in other tissues no vitamin D response element has been identified in the promoter of the 1α-hydroxylase gene (60). In keratinocytes, 1,25(OH)2D has little or no effect on CYP27B1 mRNA and protein levels when given in vitro. When 24-hydroxylase activity is blocked, 1,25(OH)2D administration fails to reduce the levels of 1,25(OH)2D produced (78,79). Thus, the apparent feedback regulation of CYP27B1 activity by 1,25(OH)2D in most tissues, with the possible exception of the kidney, appears to be due to its stimulation of CYP24A1 and subsequent catabolism, not to a direct effect on CYP27B1 expression or activity. Moreover, 1,25(OH)2D stimulates FGF23 production and inhibits PTH production. Both actions will decrease, indirectly, the ability of 1,25(OH)2D to inhibit its own production (76).  Thus, renal and extrarenal regulation of CYP27B1 by 1,25(OH)2D may differ.

Renal Production of 24,25(OH)2D

The kidney is also the major producer of a second important metabolite of 25OHD, namely 24,25(OH)2D, and the enzyme responsible is 25OHD-24 hydroxylase (CYP24A1) [75]. CYP24A1 and CYP27B1 are homologous enzymes that coexist in the mitochondria of tissues where both are found, such as the kidney tubule. However, there genes are located on different chromosomes (chromosome 20q13 and chromosome 12q14 for CYP24A1 and CYP27B1, respectively, in humans). They share the same ferrodoxin and ferrodoxin reductase components. While CYP27B1 activates the parent molecule, 25OHD, CYP24A1 initiates a series of catabolic steps that lead to its inactivation. However, in some tissues 24,25(OH)2D has been shown to have biologic effects different from 1,25(OH)2D as will be described subsequently. CYP24A1 24-hydroxylates both 25OHD and 1,25(OH)2D. The 24-hydroxylation is then followed by oxidation of 24OH to a 24-keto group, 23-hydroxylation, cleavage between C23-24, and the eventual production of calcitroic acid, a metabolite with no biologic activity. CYP24A1 also has 23-hydroxylase activity, initiating steps that lead to 23/26 lactone formation. Different species have CYP24A1s that differ in their preference for the 24-hydroxylation vs 23-hydroxylation pathway. The human enzyme follows the 24-hydroxylation pathway. Analogs with differences in their side chain are also likely to differ in the pathway utilized. CYP24A1 catalyzes all the steps in this catabolic pathway (81) (82). Although CYP24A1 is highly expressed in the kidney tubule, its tissue distribution is quite broad. In general, CYP24A1 can be found wherever the VDR is found. The affinity for 1,25(OH)2D is higher than that for 25OHD, making this enzyme an efficient means for eliminating 1,25(OH)2D. Thus, CYP24A1 is likely to play the important role of protecting the body against excess 1,25(OH)2D. Indeed, inactivating mutations in CYP24A1 have been found to underlie the disease idiopathic infantile hypercalcemia (83), manifesting as the name suggests with elevated serum calcium and 1,25(OH)2D levels. These individuals may present for the first time as adults, often in the context of increased 1,25(OH)2D production as in pregnancy (84).  An animal model in which CYP24A1 has been knocked out likewise showed very high levels of 1,25(OH)2D when treated with vitamin D and impaired mineralization of intramembranous bone (85). The skeletal abnormalities could be corrected by crossing this mouse to one lacking the VDR suggesting that excess 1,25(OH)2D (which acts through the VDR) rather than deficient 24,25(OH)2D (which does not) is to blame (85).

The regulation of CYP24A1 in the kidney is almost the mirror image of that of CYP27B1. PTH and 1,25(OH)2D are the dominant regulators, but calcium, phosphate, insulin, FGF23, IGF-I, GH, and sex steroids may also play a role. 1,25(OH)2D induces CYP24A1. The promoter of CYP24A1 has two vitamin D response elements (VDREs) critical for this induction (86-88). Protein kinase C activation as by phorbol esters enhances this induction by 1,25(OH)2D (89). An AP-1 site is found adjacent to the proximal VDRE, but mutation of this site does not appear to block phorbol ester enhancement of CYP24A1 induction by 1,25(OH)2D (90). PTH, on the other hand, inhibits the expression of CYP24A1 in the kidney (91). This action can be reproduced with cAMP (92) and forskolin (56) indicating the role of PTH activated adenylate cyclase (93). PTH has no effect on intestinal CYP24A1, most likely because the intestine does not have PTH receptors. Surprisingly, however, PTH is synergistic with 1,25(OH)2D in stimulating CYP24A1 expression and activity in bone cells which do have PTH receptors, again through a cAMP mediated mechanism (94). This synergism is further potentiated by the addition of insulin (95) (96). FGF23 also induces CYP24A1 expression (97). Surprisingly this requires the VDR (97), since FGF23 also inhibits 1,25(OH)2D production and so would be expected to reduce CYP24A1 via a 1,25(OH)2D/VDR mechanism. Restriction in dietary phosphate reduces CYP24A1 expression consistent with a decrease in FGF23, but also in a manner blocked by hypophysectomy (98). GH and IGF-I can reduce CYP24A1 expression in hypophysectomized animals, suggesting that the phosphate effect on CYP24A1 like its opposing effect on CYP27B1, is mediated by GH and IGF-I (98) as well as FGF23. The region(s) of the CYP24A1 promoter mediating these actions of PTH and FGF23 as well as 1,25(OH)2D have recently been mapped (96). Similar to that for CYP27B1 this regulation differs in different cell types. Thus, although different regulators tend to have opposite effects on CYP24A1 and CYP27B1 expression the molecular mechanisms by which the regulation occurs also differ for each enzyme.

TRANSPORT IN BLOOD

The vitamin D metabolites are transported in blood bound primarily to vitamin D binding protein (DBP) (85-88%) and albumin (12-15%) (99-101). DBP concentrations are normally 4-8µM, well above the concentrations of the vitamin D metabolites, such that DBP is only about 2% saturated. DBP has high affinity for the vitamin D metabolites (Ka=5x108M-1 for 25OHD and 24,25(OH)2D, 4x107M-1 for 1,25(OH)2D and vitamin D), such that under normal circumstances only approximately 0.03% 25OHD and 24,25(OH)2D and 0.4% 1,25(OH)2D are free (100-102). Conditions such as liver disease and nephrotic syndrome resulting in reduced DBP and albumin levels will lead to a reduction in total 25OHD and 1,25(OH)2D levels without necessarily affecting the free concentrations (103) (figure 3). Similarly, DBP levels are reduced during acute illness, potentially obscuring the interpretation of total 25OHD levels (104). Earlier studies with a monoclonal antibody to measure DBP levels suggested a decreased level in African Americans consistent with their lower total 25OHD levels, but these results were not confirmed using polyvalent antibody-based assays (105). Vitamin D intoxication can increase the degree of saturation sufficiently to increase the free concentrations of 1,25(OH)2D and so cause hypercalcemia without necessarily raising the total concentrations (106).

The vitamin D metabolites bound to DBP are in general not available to most cells. Thus, the free or unbound concentration is that which is critical for cellular uptake as postulated by the free hormone hypothesis. Support for the concept that the role of DBP is to provide a reservoir for the vitamin D metabolites but that it is the free concentration that enters cells and exerts biologic function comes from studies in mice in which DBP has been deleted and in humans in which the gene is mutated. In DBP knockout mice the vitamin D metabolites are presumably all free and/or bioavailable. These mice do not show evidence of vitamin D deficiency unless placed on a vitamin D deficient diet despite having very low levels of serum 25OHD and 1,25(OH)2D (107). Tissue levels of 1,25(OH)2D were found to be normal in the DBP knockout mice as were markers of vitamin D action such as expression of intestinal TRPV6, calbindin 9k, PMCA1b, and renal TRPV5 (108). Recently a family in which a large deletion of the coding portion of the DBP gene (and adjacent NPFFR2 gene) has been reported (109). The proband had normal calcium, phosphate and PTH levels with vitamin D supplementation despite very low levels of 25OHD, 24,25(OH)2D, and 1,25(OH)2D that were not responsive to massive doses of vitamin D (oral or parenteral). The free 25OHD was nearly normal. The carrier sibling had vitamin D metabolite levels between those of the proband and the normal sibling. Thus, both the studies in DBP null mice and humans support the free hormone hypothesis while also supporting the role of DBP as a circulating reservoir for the vitamin D metabolites. Therefore, there is currently a debate as to whether the free concentration of 25OHD, for example, is a better indicator of vitamin D nutritional status than total 25OHD, given that DBP levels, and hence total 25OHD levels, can be influenced by liver disease, nephrotic syndrome, pregnancy, and inflammatory states (110,111). However, certain tissues such as the kidney, placenta, and parathyroid gland express the megalin/cubilin complex which is able to transport vitamin D metabolites bound to DBP into the cell. This is critical for preventing renal losses of the vitamin metabolites (112) and may be important for vitamin D metabolite transport into the fetus and regulation of PTH secretion. Indeed, mice lacking the megalin/cubilin complex have poor survival with evidence of osteomalacia indicating its role in vitamin D transport into critical cells involved with vitamin D signaling

Figure 3. Correlation of total 25OHD (A) and 1,25(OH)2D (C) levels to DBP; lack of correlation of free 25OHD (B) and 1,25(OH)2D (D) levels to DBP. Data from normal subjects (open triangles), subjects with liver disease (closed triangles, open circles), subjects on oral contraceptives (open triangles*), and pregnant women (open squares) are included. These data demonstrate the dependence of total 25OHD and 1,25(OH)2D concentrations on DBP levels which are reduced by liver disease. However, the free concentrations of 25OHD and 1,25(OH)2D are normal in most patients with liver disease. Reprinted with permission from the American Society for Clinical Investigation.

DBP was originally known as group specific component (Gc-globulin) before its properties as a vitamin D transport protein became known. It has three common polymorphisms which are useful in population genetics. These alleles have somewhat different affinities for the vitamin D metabolites (113), but which do not appear to alter its function. DBP is a 58kDa protein with 458 amino acids that is homologous to albumin and α-fetoprotein (αFP) (40% homology at the nucleotide level, 23% at the amino acid level) (114). These three genes cluster on chromosome 4q11-13 (115). DBP, like albumin and αFP, is made primarily but not exclusively in the liver-other sites include the kidney, testes, and fat.  DBP like other steroid hormone binding proteins is increased by oral (not transdermal) estrogens and pregnancy (100). In vitro, glucocorticoids and cytokines such as EGF, IL-6 and TGF-β have been shown to increase (glucocorticoids, EGF, IL-6) or decrease (TGF-β) DBP production (116).

Although transport of the vitamin D metabolites may be the major function for DBP, it has other properties. DBP has high affinity for actin, and may serve as a scavenger for actin released into the blood during cell death (117). DBP has also been shown to activate macrophages (118) and osteoclasts (119). However, in a mouse rendered deficient in DBP by homologous recombination (knock out) no obvious abnormality was observed except for increased turnover in vitamin D and increased susceptibility to osteomalacia on a vitamin D deficient diet (120). Evidence for osteopetrosis (indicating failure of osteoclast function) was not found.

MECHANISM OF ACTION

The hormonal form of vitamin D, 1,25(OH)2D, is the ligand for a transcription factor, the vitamin D receptor (VDR). Most if not all effects of 1,25(OH)2D are mediated by VDR acting primarily by regulating the expression of genes whose promoters contain specific DNA sequences known as vitamin D response elements (VDREs). There are thousands of VDREs throughout the gene, often thousands of base pairs away from the coding portion of the gene regulated.  However, some actions of 1,25(OH)2D are more immediate, and may be mediated by a membrane bound vitamin D receptor that has been less well characterized than the nuclear VDR or by the VDR acting outside of the nucleus. On the other hand, some actions of VDR do not require its ligand 1,25(OH)2D. Our understanding of the mechanism by which VDR regulates gene expression has increased enormously over the past few years.

VDR and Transcriptional Regulation

 The VDR was discovered in 1969 (121) (although only as a binding protein for an as yet unknown vitamin D metabolite subsequently identified as 1,25(OH)2D), and was eventually cloned and sequenced in 1987 (122,123). Inactivating mutations in the VDR result in hereditary vitamin D resistant rickets (HVDRR) (124). Animal models in which the VDR has been knocked out (125) (126) have the full phenotype of severe vitamin D deficiency indicating that the VDR is the major mediator of vitamin D action. The one major difference is the alopecia seen in HVDRR and VDR knockout animals, a feature not associated with vitamin D deficiency, suggesting that the VDR may have functions independent of 1,25(OH)2D at least in hair follicle cycling. The VDR is a member of a large family of proteins (over 150 members) that includes the receptors for the steroid hormones, thyroid hormone, vitamin A family of metabolites (retinoids), and a variety of cholesterol metabolites, bile acids, isoprenoids, fatty acids and eicosanoids. A large number of family members have no known ligands, and are called orphan receptors. VDR is widely, although not universally, distributed throughout the different tissues of the body (127). Many of these tissues were not originally considered target tissues for 1,25(OH)2D. The discovery of VDR in these tissues along with the demonstration that 1,25(OH)2D altered function of these tissues has markedly increased our appreciation of the protean effects of 1,25(OH)2D.

The VDR is a molecule of approximately 50-60kDa depending on species. The basic structure is shown in figure 4. The VDR is unusual in that it has a very short N-terminal domain before the DNA binding domain when compared to other nuclear hormone receptors. The human VDR has two potential start sites. A common polymorphism (Fok 1) alters the first ATG start site to ACG. Individuals with this polymorphism begin translation three codons downstream such that in these individuals the VDR is three amino acids shorter (424 aas vs 427 aas). This polymorphism has been correlated with reduced bone density suggesting it is of functional importance (128). The most conserved domain in VDR from different species and among the nuclear hormone receptors in general is the DNA binding domain. This domain is comprised of two zinc fingers. The name derives from the cysteines within this stretch of amino acids that form tetrahedral complexes with zinc in a manner which creates a loop or finger of amino acids with the zinc complex at its base. The proximal (N-terminal) zinc finger confers specificity for DNA binding to the VDREs while the second zinc finger and the region following provide at least one of the sites for heterodimerization of the VDR to the retinoid X receptor (RXR). The second half of the molecule is the ligand binding domain, the region responsible for binding 1,25(OH)2D, but which also contains regions necessary for heterodimerization to RXR. At the C-terminal end is the major activation domain, AF-2, which is critical for the binding to coactivators such as those in the steroid receptor coactivator (SRC) and vitamin D receptor interacting protein (DRIP) or Mediator families (129). In mutation studies of the homologous thyroid receptor, corepressors were found to bind in overlapping regions with coactivators in helices 3 and 5, a region blocked by helix 12 (the terminal portion of the AF2 domain) in the presence of ligand (130). Deletion of helix 12 promoted corepressor binding while preventing that of coactivators (130).

Figure 4. Model of the vitamin D receptor (VDR). The N terminal region is short relative to other steroid hormone receptors. This region is followed by two zinc fingers which constitute the principal DNA binding domain. Nuclear localization signals (NLS) are found within and just C-terminal to the DNA binding domain. The ligand binding domain makes up the bulk of the C-terminal half of the molecule, with the AF2 domain comprising the most C-terminal region. The AF2 domain is largely responsible for binding to co-activators such as the SRC family and DRIP (Mediator) in the presence of ligand. Regions on the second zinc finger and within the ligand binding domain facilitate heterodimerization with RXR. Corepressor binding is less well characterized but appears to overlap that of coactivators in helices 3 and 5, a region blocked by helix 12 in the presence of ligand. 

The ligand binding domain (LBD) for VDR has been crystallized and its structure solved (131). More recently the structure of the VDR/RXR heterodimer has been analyzed by high resolution cryoelectron microscopy (132).  These studies show that the VDR has a high degree of structural homology to other nuclear hormone receptors. It is comprised of 12 helices joined primarily by beta sheets. The 1,25(OH)2D is buried deep in the ligand binding pocket and covered by helix 12 (the terminal portion of the AF-2 domain). Assuming analogy with the unliganded LBD of RXRα and the ligand bound LBD of RARγ (133), the binding of 1,25(OH)2D to the VDR triggers a substantial movement of helix 12 from an open position to a closed position, covering the ligand binding pocket and putting helix 12 in position with critical residues from helices 3, 4, and 5 to bind coactivators. Coactivator complexes bridge the gap from the VDRE to the transcription machinery at the transcription start site (figure 5) (134).

Figure 5. 1,25(OH)2D-initiated gene transcription. 1,25(OH)2D enters the target cell and binds to its receptor, VDR. The VDR then heterodimerizes with the retinoid X receptor (RXR). This increases the affinity of the VDR/RXR complex for the vitamin D response element (VDRE), a specific sequence of nucleotides in the promoter region of the vitamin D responsive gene. Binding of the VDR/RXR complex to the VDRE attracts a complex of proteins termed coactivators to the VDR/RXR complex. The DRIP (Mediator) coactivator complex spans the gap between the VDRE and RNA polymerase II and other proteins in the initiation complex centered at or around the TATA box (or other transcription regulatory elements). SRC coactivators recruit histone acetyl transferases (HAT) to the gene promoting the opening up of its structure to enable the transcription machinery to work. Transcription of the gene is initiated to produce the corresponding mRNA, which leaves the nucleus to be translated to the corresponding protein.

Nuclear hormone receptors including the VDR are further regulated by protein complexes that can be activators or repressors (135). The role of corepressors in VDR function has been demonstrated (136) but is less well studied than the role of coactivators. One such corepressor, hairless, is found in the skin and may regulate 1,25(OH)2D mediated epidermal proliferation and differentiation as well as ligand independent VDR regulation of hair follicle cycling (137-139). The coactivators, which are essential for VDR function, form two distinct complexes, the interaction of which remains unclear (129). The SRC family has three members, SRC 1-3, all of which can bind to the VDR in the presence of ligand (1,25(OH)2D) (140). These coactivators recruit additional coactivators such as CBP/p300 and p/CAF that have histone acetyl transferase activity (HAT), an enzyme that by acetylation of lysines within specific histones appears to help unravel the chromatin allowing the transcriptional machinery to do its job. The domain in these molecules critical for binding to the VDR and other nuclear hormone receptors is called the NR box, and has as its central motif LxxLL where L stands for leucine and x for any amino acid. Each SRC family member contains three well conserved NR boxes in the region critical for nuclear hormone receptor binding. The DRIP (Mediator) complex is comprised of 15 or so proteins several of which contain LxxLL motifs (141). However, DRIP205 (Mediator 1) is the protein critical for binding the complex to VDR. It contains 2 NR boxes. Different NR boxes in these coactivators show specificity for different nuclear hormone receptors (142). Unlike the SRC complex, the DRIP complex does not have HAT activity (129). Rather the DRIP complex spans the gene from the VDRE to the transcription start site linking directly with RNA polymerase II and its associated transcription factors.  DRIP and SRC appear to compete for binding to the VDR. In keratinocytes DRIP binds preferentially to the VDR in undifferentiated cells, whereas SRC 2 and 3 bind in the more differentiated cells in which DRIP levels have declined (143). Thus in these cells DRIP appears to regulate the early stages of 1,25(OH)2D induced differentiation, whereas SRC may be more important in the later stages, although overlap in gene specificity is also observed (144,145) (146). These coregulators are not specific for VDR, but interact with a large number of other transcription factors. The DRIP (Mediator) complex can mark regions in the genome containing large numbers of sites for transcription factors including VDREs. These sites are known as super enhancers often regulating genes involved with cell fate determination (147).  Recently, SMAD 3, a transcription factor in the TGF-β pathway, has been found to complex with the SRC family members and the VDR, enhancing the coactivation process (148). Phosphorylation of the VDR may also control VDR function (149). Furthermore, VDR has been shown to suppress β-catenin transcriptional activity (150), whereas β-catenin enhances that of VDR (151).  Thus, control of VDR activity may involve crosstalk between signaling pathways originating in receptors at the plasma membrane as well as within the nucleus.

VDR acts in concert with other nuclear hormone receptors, in particular RXR (152). Unlike VDR, there are three forms of RXR--α, β, γ--and all three are capable of binding to VDR with no obvious differences in terms of functional effect. RXR and VDR form heterodimers that optimize their affinity for the vitamin D response elements (VDREs) in the genes being regulated. RXR appears to be responsible for keeping VDR in the nucleus in the absence of ligand (153). VDR may also partner with other receptors including the thyroid receptor (TR) and the retinoic acid receptor (RAR) (154,155), but these are the exceptions, whereas RXR is the rule. The VDR/RXR heterodimers bind to VDREs, which typically are comprised of two half sites each with six nucleotides separated by three nucleotides of nonspecific type; this type of VDRE is known as a DR3 (direct repeats with three nucleotide spacing). RXR binds to the upstream half site, while VDR binds to the downstream site (156). However, a wide range of VDRE configurations have been found at nearly any location within a gene (5’, 3’, introns) (157). Moreover, different tissues differ as to which VDREs actively bind VDR (158). 1,25(OH)2D is required for high affinity binding and activation, but the RXR ligand, 9-cis retinoic acid, may either inhibit (159) or activate (160) 1,25(OH)2D stimulation of gene transcription. A DR6 has been identified in the phospholipase C-γ1 gene that recognizes VDR/RAR heterodimers (154), and a DR4 has been found in the mouse calbindin 28k gene (161). Inverted palinodromes with 7 to 12 bases between half sites have also been found (151).  Furthermore, the half sites of the various known VDREs show remarkable degeneracy (table 1). The G in the second position of each site appears to be the only nearly invariant nucleotide. 1,25(OH)2D can also inhibit gene transcription through its VDR. This may occur by direct binding of the VDR to negative VDREs that in the PTH and PTHrP genes are remarkably similar in sequence to positive VDREs of other genes (162,163). However, inhibition may also be indirect. For example, 1,25(OH)2D inhibits IL-2 production by blocking the NFATp/AP-1 complex of transcription factors from activating this gene (164) through a mechanism not yet clear. Similarly, 1,25(OH)2D inhibits CYP27B1 in at least one renal cell line by an indirect mechanism involving VDR binding to VDIR (62,80). Thus, a variety of factors including the flanking sequences of the genes around the VDREs and tissue specific factors play a large role in dictating the ability of 1,25(OH)2D to regulate gene expression.

Non-Genomic Actions

A variety of hormones that serve as ligands for nuclear hormone receptors also exert biologic effects that do not appear to require gene regulation and may work through membrane receptors rather their cognate nuclear hormone receptors. Examples include estrogen (165), progesterone (166), testosterone (167), corticosteroids (168), and thyroid hormone (169). 1,25(OH)2D has also been shown to have rapid effects on selected cells that are not likely to involve gene regulation and that appear to be mediated by a different, probably membrane receptor. A model for such effects is shown in figure 6. Similar to other steroid hormones, 1,25(OH)2D has been shown to regulate calcium and chloride channel activity, protein kinase C activation and distribution, and phospholipase C activity in a number of cells including osteoblasts (170), liver (171), muscle (172), and intestine (173,174). These rapid effects of 1,25(OH)2D have been most extensively studied in the intestine. Norman's laboratory coined the term transcaltachia to describe the rapid onset of calcium flux across the intestine of a vitamin D replete chick perfused with 1,25(OH)2D (175). This increased flux could not be blocked with actinomycin D pretreatment (176), but was blocked by voltage gated L type channel inhibitors (177) and protein kinase C inhibitors (178). These animals had to be vitamin D replete and contain the VDR, indicating that the basic machinery for calcium transport was intact. On the other hand L type channel activators such as BAY K-8644 (179) and protein kinase C activators such as phorbol esters (177) could activate transcaltachia similar to 1,25(OH)2D.

Figure 6. Model for the non-genomic actions of 1,25(OH)2D. 1,25(OH)2D binds to a putative membrane receptor. This leads to activation of a G protein (GTP displacement of GDP and dissociation of the β and γ subunits from the now active α subunit). Gα -GTP activates phospholipase C (PLC) (β or γ) to hydrolyze phosphatidyl inositol bis phosphate (PIP2) to inositol tris phosphate (IP3) and diacyl glycerol (DG). IP3 releases calcium from intracellular stores via the IP3 receptor in the endoplasmic reticulum; DG activates protein kinase C (PKC). Both calcium and PKC may regulate the influx of calcium across the plasma membrane through various calcium channels including L-type calcium channels.  

A putative membrane receptor for 1,25(OH)2D (1,25(OH)2D membrane associated rapid response steroid binding protein (1,25D-MARRSBP) also known as ERp57) has been purified from the intestine (180) and subsequently cloned and sequenced (181). Its size is approximately 66kDa. Antibodies have been made against this putative receptor (182). These antibodies block the ability of 1,25(OH)2D to stimulate calcium uptake by isolated chick intestinal cells (183) and to stimulate protein kinase C activity in resting zone chondrocytes while inhibiting proliferation of both resting zone and proliferating zone chondrocytes (182). Analog studies also support the existence of a separate membrane receptor for 1,25(OH)2D. Because of the breaking of the B ring during vitamin D3 production from 7-dehydrocholesterol, the A ring can assume a conformation similar to the parent cholesterol molecule (6-s-cis) (shown as previtamin D3 in figure 1) or the more commonly depicted 6-s-trans form in which the A ring rotates away from the rest of the molecule (shown as vitamin D3 in figure 1). Analogs of 1,25(OH)2D can be produced which favor the 6-s-cis conformation or the 6-s-trans conformation. 1,25(OH)2-d5-previtamin D3 is one such analog locked into the 6-s-cis conformation. This analog has only weak activity with respect to VDR binding or transcriptional activation but is fully effective in terms of stimulating transcaltachia and calcium uptake by osteosarcoma cells when compared to 1,25(OH)2D (184). 6-s-trans analogs are not effective. However, some of these rapid actions of 1,25(OH)2D are not found in cells from VDR null mice suggesting that the VDR may be required for the expression and/or function of the membrane receptor or be the membrane receptor. In other cells both 1,25D-MARRSBP and VDR appear to be required for these rapid effects of 1,25(OH)2D (185,186).

The model (figure 6) emerging from these studies is that 1,25(OH)2D interacts with a membrane receptor to activate phospholipase C possibly through a G protein coupled process. Phospholipase C then hydrolyzes phosphatidyl inositol bis phosphate (PIP2) in the membrane releasing inositol tris phosphate (IP3) and diacyl glycerol (DG). These second messengers may then activate both the intracellular release of calcium from intracellular stores via the IP3 receptor and protein kinase C, either one or both of which could stimulate calcium channel activity leading to a further rise in intracellular calcium levels. In the intestine and kidney, the increased flux of calcium across the brush border membrane is then transported out of the cell at the basolateral membrane, completing transcellular transport. In other cells the increased calcium would need to be removed by other mechanisms after the signal conveyed by the rise in calcium is no longer required. Much work remains to prove this model including the physiologic requirement for a unique membrane receptor.

TARGET TISSUE RESPONSES: CALCIUM REGULATING ORGANS 

Intestine

Intestinal calcium absorption, in particular the active component of transcellular calcium absorption, is one of the oldest and best known actions of vitamin D having been first described in vitro by Schachter and Rosen (187) in 1959 and in vivo by Wasserman et al. (188) in 1961. Absorption of calcium from the luminal contents of the intestine involves both transcellular and paracellular pathways. The transcellular pathway dominates in the duodenum and cecum, and this is the pathway primarily regulated by 1,25 dihydroxyvitamin D (1,25(OH)2D) (189), although elements of the paracellular pathway such as the claudins 2 and 12 are likewise regulated by 1,25(OH)2D (reviews in (190,191). Figure 7 shows a model of our current understanding of how this process is regulated by 1,25(OH)2D. Calcium entry across the brush border membrane (BBM) occurs down a steep electrical-chemical gradient and requires no input of energy. Removal of calcium at the basolateral membrane must work against this gradient, and energy is required. This is achieved by the CaATPase (PMCA1b), an enzyme induced by 1,25(OH)2D in the intestine. Calcium movement through the cell occurs with minimal elevation of the intracellular free calcium concentration (192) by packaging the calcium in calbindin containing vesicles (193-195) that form in the terminal web following 1,25(OH)2D administration.

Figure 7. Model of intestinal calcium transport. Calcium enters the microvillus of the intestinal epithelial cell through TRPV6 (previously known as CaT1) calcium channel. Within the microvillus calcium is bound to calmodulin (CaM) which is itself bound to brush border myosin I (BBMI). BBMI may facilitate the movement of the calcium/CaM complex into the terminal web where the calcium is picked up by calbindin (CaBP) and transported through the cytoplasm in endocytic vesicles. At the basolateral membrane the calcium is pumped out of the cell by the Ca-ATPase (PMCA1b). 1,25(OH)2D enhances intestinal calcium transport by inducing TRPV6, CaBP, and PMCAb as well as increasing the amount of CaM bound to BBMI in the brush border.  

1,25(OH)2D regulates transcellular calcium transport using a combination of genomic and nongenomic actions. The first step, calcium entry across the BBM, is accompanied by changes in the lipid composition of the membrane including an increase in linoleic and arachidonic acid (196,197) and an increase in the phosphatidylcholine:phosphatidylethanolamine ratio (198). These changes are associated with increased membrane fluidity (197), which we have shown results in increased calcium flux (199). The changes in lipid composition occur within hours after 1,25(OH)2D administration and are not blocked by pretreatment with cycloheximide (198). In addition, an epithelial specific calcium channel, TRPV6, is expressed in the intestinal epithelium (200). This channel has a high degree of homology to TRPV5, a channel originally identified in the kidney (201,202). The tissue distributions of these channels are overlapping and can be found in other tissues, but TRPV6 appears to be the main form in the intestine (203,204). TRPV6 mRNA levels in the intestine of vitamin D deficient mice are markedly increased by 1,25(OH)2D, although similar changes are not found in the kidney (205).  Mice null for TRPV6 have decreased intestinal calcium transport (206).

Calcium entering the brush border must then be moved into and through the cytoplasm without disrupting the function of the cell. Electron microscopic observations indicate that in the vitamin D deficient animal, calcium accumulates along the inner surface of the plasma membrane of the microvilli (207,208). Following vitamin D or 1,25(OH)2D administration calcium leaves the microvilli and subsequently can be found in mitochondria and vesicles within the terminal web (193,194,207,208). The vesicles appear to shuttle the calcium to the lateral membrane where it is pumped out of the cell by the basolateral CaATPase, PMCA1b. These morphologic observations have been confirmed by direct measurements of calcium using x-ray microanalysis that demonstrate equivalent amounts of calcium within the microvilli of D deficient and 1,25(OH)2D treated animals but much higher amounts of calcium in the mitochondria and vesicles of the 1,25(OH)2D treated animals (194,209). Such data suggest that 1,25(OH)2D controls calcium entry into the cell primarily by regulating its removal from the microvillus and accumulation by subcellular organelles in the terminal web, although flux through calcium channels in the membrane such as TRPV6 also plays a major role.

The ability of 1,25(OH)2D to stimulate calcium entry into and transport from the microvillus does not require new protein synthesis (193,198,210). Cycloheximide does not block the ability of 1,25(OH)2D to increase the capacity of brush border membrane vesicles (BBMV) to accumulate calcium, although it does block the increase in alkaline phosphatase in the same BBMV [193]. Likewise, cycloheximide does not block the increase in mitochondrial calcium following 1,25(OH)2D administration, although it blocks the rise in calbindin and prevents the normal vesicular transport of calcium through the cytosol (193,211). Thus, nongenomic actions underlie at least some of these first steps in 1,25(OH)2D stimulated intestinal calcium transport within the microvillus, although the changes take hours, not minutes, to observe. The exact role for these nongenomic effects on calcium influx relative to the role of TRPV6 remains to be elucidated.

Calmodulin is the major calcium binding protein in the microvillus (212). Its concentration in the microvillus is increased by 1,25(OH)2D; no new calmodulin synthesis is required or observed after 1,25(OH)2D administration (213). Calmodulin is likely to play a major role in calcium transport within the microvillus, and inhibitors of calmodulin block 1,25(OH)2D stimulated calcium uptake by BBMV (214). Within the microvillus calmodulin is bound to a 110kD protein, myosin 1A (myo1A)) (previously referred to as brush border myosin 1). 1,25(OH)2D increases the binding of calmodulin to myo1A in brush border membrane preparations (213), although binding of calmodulin to the myo1A attached to the actin core following detergent extraction of the membrane appears to be reduced (215). The calmodulin/myo1A complex appears late in the development of the brush border, and is found in highest concentration in the same cells of the villus which have the highest capacity for calcium transport (216). Myo1A is located primarily in the microvillus of the mature intestinal epithelial cell, although small amounts have been detected associated with vesicles in the terminal web (217). Thus, the calmodulin/myo1A complex may be responsible for moving calcium out of the microvillus. Its exact role in calcium transport is unclear in that mice null for myo1A do not show reduced intestinal calcium transport(218)).  Calbindin is the dominant calcium binding protein in the cytoplasm (212,219), where it appears to play the major role in calcium transport from the terminal web to the basolateral membrane (190). The increase in calbindin levels in the cytosol following 1,25(OH)2D administration is blocked by protein synthesis inhibitors (210). Indeed, calbindin was the first protein discovered to be induced by vitamin D (219). Glenney and Glenney (212) observed that calbindin has a higher affinity for calcium than does calmodulin. The differential distribution of calmodulin and calbindin between microvillus and cytosol combined with the differences in affinity for calcium led Glenney and Glenney (212) to propose that in the course of calcium transport calcium flowed from calmodulin in the microvillus to calbindin in the cytosol with minimal change in the free calcium concentration in either location. However, the role of calbindin in intestinal calcium transport does not appear to be critical in that mice null for calbindin9k grow normally, have normal intestinal calcium transport, and their serum calcium levels and bone mineral content are equivalent to wildtype mice regardless of the calcium content of the diet (220). The CaATPase (PMCA1b) at the basolateral membrane and the sodium/calcium exchanger (NCX1) are responsible for removing calcium from the cell against the same steep electrochemical gradient as favored calcium entry at the brush border membrane (221). Related proteins are found in the renal distal tubule. As its name implies, the extrusion of calcium from the cell by the calcium pump requires ATP. This pump is a member of the PMCA family, and in the intestine the isoform PMCA1b is the major isoform found. This pump is induced by 1,25(OH)2D (222). Calmodulin activates the pump, but calbindin may do likewise (223). Deletion of Pmca1b reduces calcium absorption and blocks 1,25(OH)2D stimulation of such resulting in reduction in growth and bone mineralization (224)., Moreover, the deletion of protein 4.1R, which regulates PMCA1b expression in the intestine, results in decreased intestinal calcium transport (225). The role of NCX is not considered to be as important as PMCA1b for intestinal calcium transport (226).

The paracellular pathway has received less study, but accounts for the bulk of intestinal calcium transport in that the ileum accounts for around 80% of total calcium absorption essentially all by the paracellular pathway. Paracellular calcium absorption depends to a considerable extent on the gradient between the luminal calcium concentrations and the interstitial calcium concentrations. Thus, it is faster in the duodenum and upper jejunum than the ileum, but because the transit time in the ileum is so much longer than that of the upper GI tract, the ileum is where most of the calcium absorption takes place. Solvent drag plays a large part in moving calcium across the tight junctions between the epithelial cells (227) . Solvent flow follows the osmotic gradient which is maintained distal to the tight junction by the Na/K ATPase and sodium glucose cotransporter of the basolateral membrane which may be stimulated by 1,25(OH)2D (226,227). The tight junction itself provides both charge and size selectivity. The actomyosin ring around the tight junction contributes to the size selectivity (228). The claudins and occludins contribute to charge selectivity. Claudin 2, 12, 15 are negatively charged proteins enabling cations such as sodium and calcium to pass (229,230). 1,25(OH)2D stimulates the expression of claudins 2 and 12 (231). Prolactin stimulates claudin 15 expression, thought to contribute to the increased calcium absorption during pregnancy (232).

Although less studied, intestinal phosphate transport is also under the control of vitamin D. This was first demonstrated by Harrison and Harrison (233) in 1961. Active phosphate transport is greatest in the jejunum, in contrast to active calcium transport that is greatest in the duodenum. Cycloheximide blocks 1,25(OH)2D stimulated phosphate transport (234), indicating that protein synthesis is involved. Phosphate transport at both the brush border and basolateral membranes requires sodium. A sodium-phosphate transporter in the small intestine (NaPi-IIb), homologous to the type IIa sodium phosphate transporter in kidney, has been cloned and sequenced (235). Expression of NaPi-IIb is increased by 1,25(OH)2D (236). Transport of phosphate through the cytosol from one membrane to the other is poorly understood. However, cytochalasin B, a disrupter of microfilaments, has been shown to disrupt this process (237) suggesting that as for calcium, intracellular phosphate transport occurs in vesicles.

 Bone

Nutritional vitamin D deficiency, altered vitamin D responsiveness such as vitamin D receptor mutations (hereditary vitamin D resistant rickets), and deficient production of 1,25(OH)2D such as mutations in the CYP27B1 gene (pseudo vitamin D deficiency) all have rickets as their main phenotype. This would suggest that vitamin D, and in particular 1,25(OH)2D, is of critical importance to bone. Furthermore, VDR are found in bone cells (238,239), and vitamin D metabolites have been shown to regulate many processes in bone. However, the rickets resulting from vitamin D deficiency or VDR mutations (or knockouts) can be corrected by supplying adequate amounts of calcium and phosphate either by infusions or orally [214-217]. Moreover, deletion of VDR from bone cells does not result in rickets (240). This would suggest either that vitamin D metabolites do not directly impact bone, or that substantial redundancy has been built into the system.  However, arguing for a physiologically non-redundant direct action of vitamin D on bone is the development of osteoporosis and decreased bone formation in these VDR or CYP27B1 null mice not corrected by the high calcium/phosphate diet (241).  A further complicating factor in determining the role of vitamin D metabolites in bone is the multitude of effects these metabolites have on systemic calcium homeostatic mechanisms which themselves impact on bone. The lack of vitamin D results in hypocalcemia and hypophosphatemia that as implied above is sufficient to cause rickets. Moreover, part of the skeletal phenotype in vitamin D deficiency is also due to the hyperparathyroidism that develops in the vitamin D deficient state as PTH has its own actions on bone and cartilage. Furthermore, within bone the vitamin D metabolites can alter the expression and/or secretion of a large number of skeletally derived factors including insulin like growth factor-1 (IGF-I) (242), its receptor (243), and binding proteins (244,245), transforming growth factor β (TGFβ) (246), vascular endothelial growth factor (VEGF) (247), interleukin-6 (IL-6) (248), IL-4 (249), and endothelin receptors (250) all of which can exert effects on bone of their own as well as modulate the actions of the vitamin D metabolites on bone. Understanding the impact of vitamin D metabolites on bone is additionally complicated by species differences, differences in responsiveness of bone and cartilage cells according to their states of differentiation, and differences in responsiveness in terms of the vitamin D metabolite being examined. Thus, the study of vitamin D on bone has had a complex history, and uncertainty remains as to how critical the direct actions of the vitamin D metabolites on bone are for bone formation and resorption.

Bone develops intramembranously (e.g., skull) or from cartilage (endochondral bone formation, e.g., long bones with growth plates). Intramembranous bone formation occurs when osteoprogenitor cells proliferate and produce osteoid, a type I collagen rich matrix. The osteoprogenitor cells differentiate into osteoblasts which then deposit calcium phosphate crystals into the matrix to produce woven bone. This bone is remodeled into mature lamellar bone. Endochondral bone formation is initiated by the differentiation of mesenchymal stem cells into chondroblasts that produce the proteoglycan rich type II collagen matrix. These cells continue to differentiate into hypertrophic chondrocytes that shift from making type II collagen to producing type X collagen. These cells also initiate the degradation and calcification of the matrix by secreting matrix vesicles filled with degradative enzymes such as metalloproteinases and phospholipases, alkaline phosphatase (thought to be critical for the mineralization process), and calcium phosphate crystals. Vascular invasion and osteoclastic resorption are stimulated by the production of VEGF and other chemotactic factors from the degraded matrix. The hypertrophic chondrocytes also begin to produce markers of osteoblasts such as osteocalcin, osteopontin, and type I collagen resulting in the initial deposition of osteoid. Moreover, at least some of these chondrocytes further differentiate (or trans differentiate) into osteoblasts (251). Terminal differentiation of the hypertrophic chondrocytes and the subsequent calcification of the matrix are markedly impaired in vitamin D deficiency leading to the flaring of the ends of the long bones and the rachitic rosary along the costochondral junctions of the ribs, classic features of rickets. Although supply of adequate amounts of calcium and phosphate may correct most of these defects in terminal differentiation and calcification, the vitamin D metabolites, 1,25(OH)2D and 24,25(OH)2D, have been shown to exert distinct roles in the process of endochondral bone formation.

The VDR makes its first appearance in the fetal rat at day 13 of gestation in the condensing mesenchyme of the vertebral column then subsequently in osteoblasts and the proliferating and hypertrophic chondrocytes by day 17 (252). However, fetal development is quite normal in vitamin D deficient rats (253) and VDR knockout mice (126) suggesting that vitamin D and the VDR are not critical for skeletal formation. Rickets develops postnatally, becoming most manifest after weaning. The impairment of endochondral bone formation observed in vitamin D deficiency is associated with decreased alkaline phosphatase activity of the hypertrophic chondrocytes (254), alterations in the lipid composition of the matrix (255) perhaps secondary to reduced phospholipase activity (256), and altered proteoglycan degradation (257) due to changes in metalloproteinase activity (257,258). Both 1,25(OH)2D and 24,25(OH)2D appear to be required for optimal endochondral bone formation (259). However, in the CYP24A1 knockout mouse, that fails to produce any 24-hydroxylated metabolites of vitamin D, the skeletal lesion is defective mineralization of intramembranous (not endochondral) bone. Furthermore, the skeletal abnormality appears to be due to high circulating 1,25(OH)2D levels in that crossing this mouse with one lacking the VDR corrects the problem (85). Whether this reflects species differences between mice and other species (most studies demonstrating the role of 24,25(OH)2D in bone and cartilage have used rats and chicks) remains unknown. Chondrocytes from the resting zone of the growth plate of rats tend to be more responsive to 24,25(OH)2D than 1,25(OH)2D, whereas the reverse is true for chondrocytes from the growth zone with respect to stimulation of alkaline phosphatase activity (260), regulation of phospholipase A2 (stimulation by 1,25(OH)2D, inhibition by 24,25(OH)2D) (261), changes in membrane fluidity (increased by 1,25(OH)2D, decreased by 24,25(OH)2D) (262), and stimulation of protein kinase C activity (263). These actions of 1,25(OH)2D and 24,25(OH)2D do not require the VDR and are non-genomic in that they take place with isolated matrix vesicles and membrane preparations from these cells (260). As discussed earlier membrane receptors for these vitamin D metabolites have been found in chondrocytes that may mediate these non-genomic actions (264). Osteoblasts also differ in their response to 1,25(OH)2D depending on their degree of maturation (265). In the latter stages of differentiation, rat osteoblasts respond to 1,25(OH)2D with an increase in osteocalcin production (266), but do not respond to 1,25(OH)2D in the early stages. Mice, however, differ from rats in that 1,25(OH)2D inhibits osteocalcin expression (266). Similarly, the effects of 1,25(OH)2D on alkaline phosphatase (267) and type I collagen (268) are inhibitory in the early stages of osteoblast differentiation but stimulatory in the latter stages (265). Osteopontin is better stimulated by 1,25(OH)2D in the early stages than the late stages of differentiation (265,269). Osteocalcin and osteopontin in human and rat cells have well described VDREs in their promoters (270-272) (the mouse does not) (273). However, alkaline phosphatase and the COL1A1 and COL1A2 genes producing type I collagen do not have clearly defined VDREs, so it remains unclear how these genes are regulated by 1,25(OH)2D. These maturation dependent effects of 1,25(OH)2D on bone cell function may explain the surprising ability of excess 1,25(OH)2D to block mineralization leading to hyperosteoidosis (274,275) as such doses may prevent the normal maturation of osteoblasts.

In addition to its role in promoting bone formation, 1,25(OH)2D also promotes bone resorption by increasing the number and activity of osteoclasts (276). Whether mature osteoclasts contain the VDR and are regulated directly by 1,25(OH)2D remains controversial (277,278), but the VDR in osteoclast precursors is not required for osteoclastogenesis. Rather, the stimulation of osteoclastogenesis by 1,25(OH)2D is mediated by osteoblasts. Rodan and Martin (279) originally proposed the hypothesis that osteoblasts were required for osteoclastogenesis, and the mechanism has now been elucidated (280). Osteoblasts produce a membrane associated protein known as RANKL (receptor activator of nuclear factor (NF)-kB ligand) that activates RANK on osteoclasts and their hematopoietic precursors. This cell-to-cell contact in combination with m-CSF also produced by osteoblasts stimulates the differentiation of precursors to osteoclasts, and promotes their activity. 1,25(OH)2D regulates this process by inducing RANKL (281) as does PTH, PGE2, and IL-11, all of which stimulate osteoclastogenesis. 1,25(OH)2D requires the VDR in osteoblasts for this purpose, although the other hormones and cytokines do not. Osteoblasts from Vdr knockout mice fail to support 1,25(OH)2D induced osteoclastogenesis, whereas osteoclast precursors from Vdr knockout mice can be induced by 1,25(OH)2D to form osteoclasts in the presence of osteoblasts from wildtype animals (282). 

Kidney

The regulation of calcium and phosphate transport by vitamin D metabolites in the kidney has received less study than that in the intestine, but the two tissues have similar although not identical mechanisms. Eight grams of calcium are filtered by the glomerulus each day, and 98% of that is reabsorbed. Most is reabsorbed in the proximal tubule. This is a paracellular, sodium dependent process with little or no regulation by PTH and 1,25(OH)2D. Approximately 20% of calcium is reabsorbed in the thick ascending limb of the loop of Henle, 10-15% in the distal tubule, and 5% in the collecting duct (283). Regulation by vitamin D takes place in the distal tubule where calcium moves against an electrochemical gradient (presumably transcellular) in a sodium independent fashion (284). Phosphate, on the other hand, is approximately 80% reabsorbed in the proximal tubule, and this process is regulated by PTH (285). In parathyroidectomized (PTX) animals Puschett et al. (286-288)) demonstrated acute effects of 25OHD and 1,25(OH)2D on calcium and phosphate reabsorption. Subsequent studies indicated that PTH could enhance or was required for the stimulation of calcium and phosphate reabsorption by vitamin D metabolites (289,290).

The molecules critical for calcium reabsorption in the distal tubule appear to be the VDR, calbindin, TRPV5, and the BLM calcium pump (PMCA1b as in the intestine), a situation similar to the mechanism for calcium transport in the intestine. However, the calbindin in the kidney in most species is 28kDa, whereas the 9kDa form is found in the intestine in most species. The kidney has mostly TRPV5, whereas the intestine is primarily TRPV6. The calcium pump is the same isoform in both tissues (PMCA1b) although other forms of PMCA are also present. Calmodulin and a brush border myosin I like protein are also found in the kidney brush border, but their role in renal calcium transport has not been explored. VDR, calbindin, TRPV5, and PMCA1b colocalize in the distal tubule, but not all distal tubules contain this collection of proteins (201,202,291,292) suggesting that not all distal tubules are involved in calcium transport. 1,25(OH)2D upregulates the VDR (234), an action opposed by PTH (237). Calbindin is also induced by 1,25(OH)2D in the kidney(293,294). The activity of the calcium pump is increased by 1,25(OH)2D (295), but it is not clear that the protein itself is induced. The increased activity may be due to the induction of calbindin that increases its activity. The effect of 1,25(OH)2D on TRPV5 expression is stimulatory (205).

Phosphate reabsorption in the proximal tubule is mediated at the brush border by sodium dependent phosphate transporters (NaPi-2a and NaPi-2c) that rely on the baso-lateral membrane Na,K ATPase to maintain the sodium gradient that drives the transport process (296). It is not clear whether 1,25(OH)2D regulates the expression or activities of these transporters as it does in the intestine, although PTH clearly does. Like PTH, FGF23 blocks phosphate reabsorption, presumably by blocking NaPi-2a activity. Unlike PTH, FGF23 also blocks the renal production of 1,25(OH)2D, as discussed earlier.  The link between phosphate reabsorption and 1,25(OH)2D production remains unclear.

TARGET TISSUE RESPONSES: NON-CALCIUM TRANSPORTING TISSUES

In addition to the its effects on tissues directly responsible for calcium homeostasis, 1,25(OH)2D regulates the function of a wide number of other tissues. These all contain the VDR. Regulation of differentiation and proliferation is one common theme; regulation of hormone secretion is another; regulation of immune function is the third. In most cases 1,25(OH)2D acts in conjunction with calcium. Selected examples follow.

Regulation of Hormone Secretion

PARATHYROID GLAND (PTH SECRETION)

As previously mentioned, PTH stimulates the production of 1,25(OH)2D. In turn 1,25(OH)2D inhibits the production of PTH (297,298). The regulation occurs at the transcriptional level. Within the promoter of the PTH gene is a region that binds the VDR and mediates the suppression of the PTH promoter by 1,25(OH)2D (162,293,299-303). However, there is substantial controversy about whether this site is a single half site (299) or a more classic DR3 (292), whether one VDRE is involved or two (300), whether only VDR binds (299,303), whether VDR/RXR heterodimers bind (162,300), or whether VDR partners with a different protein (301). Some of the differences may reflect different species, but the nature of PTH gene suppression by 1,25(OH)2D remains incompletely understood. Calcium alters the ability of 1,25(OH)2D to regulate PTH gene expression. Calcium is a potent inhibitor of PTH production and secretion, acting through the calcium sensing receptor (CaSR) on the plasma membrane of the parathyroid cell. 1,25(OH)2D induces the CaSR in the parathyroid gland making it more sensitive to calcium (304). Animals placed on a low calcium diet have an increase in PTH and 1,25(OH)2D levels indicating that the low calcium overrides the inhibition by 1,25(OH)2D on PTH secretion (305,306). One possible explanation involves the protein calreticulin that binds to nuclear hormone receptors including VDR at KXGFFKR sequences, and inhibits their activity (307,308). Low dietary calcium has been shown to increase calreticulin levels in the parathyroid gland (309). The ability of 1,25(OH)2D to inhibit PTH production and secretion has been exploited clinically in that 1,25(OH)2D and several of its analogs are used to prevent and/or treat secondary hyperparathyroidism associated with renal failure. The parathyroid gland also expresses CYP27B1 and so can produce its own 1,25(OH)2D that may act in an autocrine or paracrine fashion to regulate PTH production (310). As noted earlier, the parathyroid gland is one of several tissues expressing the megalin/cubilin complex potentially enabling it to take up 25OHD and other D metabolites still bound to DBP.

PANCREATIC BETA CELLS (INSULIN SECRETION) 

1,25(OH)2D stimulates insulin secretion, although the mechanism is not well defined (311,312). VDR, CYP27B1 and calbindin-D28k are found in pancreatic beta cells (313-315), and  studies using calbindin-D28k null mice have suggested that calbindin-D28k, by regulating intracellular calcium, can modulate depolarization-stimulated insulin release (316).  Furthermore, calbindin-D28k, by buffering calcium, can protect against cytokine mediated destruction of beta cells (317).  A number of mostly case control and observational studies have suggested that vitamin D deficiency contributes to increased risk for type 2 diabetes mellitus (318). Moreover, several randomized clinical trials evaluating the ability of vitamin D supplementation to prevent the progression of prediabetes to diabetes indicate that vitamin D has a modest protective effect especially in vitamin D deficient subjects (319,320).

FIBROBLAST GROWTH FACTOR (FGF23)

 FGF23 is produced primarily by bone, and in particular by osteoblasts and osteocytes. 1,25(OH)2D3 stimulates this process, but the mechanism is not clear (322). Inasmuch as FGF23 inhibits 1,25(OH)2D production by the kidney, this feedback loop like that for PTH secretion maintains a balance in the levels of these important hormones. Mutations in the Phosphate regulating gene with Homologies to Endopeptidases on the X chromosome (PHEX) or FGF23 itself (which prevent its proteolysis) or conditions such as McCune-Albright disease and tumor induced osteomalacia in which FGF23 is overexpressed in the involved tissue led to hypophosphatemia and inappropriately low 1,25(OH)2D accompanied by osteomalacia. The role of PHEX, which was originally thought to cleave FGF23, in regulating FGF23 levels is not clear.  In contrast mutations in UDP-N-acetyl-α-D galactosamine:polypeptide N-acetylgalactosaminyltransferase (GALNT3), which glycosylates FGF23, or in FGF23 which blocks this glycosylation result in inhibited FGF23 secretion leading to hyperphosphatemia, increased 1,25(OH)2D, and tumoral calcinosis (323).

Regulation of Proliferation and Differentiation 

CANCER

 1,25(OH)2D has been evaluated for its potential anticancer activity in animal and cell studies for nearly 40 years (324). The list of malignant cells that express VDR is now quite extensive, and the list of those same cells that express CYP27B1 is growing. The accepted basis for the promise of 1,25(OH)2D in the prevention and treatment of malignancy includes its antiproliferative, pro-differentiating effects on most cell types. The list of mechanisms proposed for these actions is extensive, and to some extent cell specific (325). Among these mechanisms 1,25(OH)2D has been shown to stimulate the expression of cell cycle inhibitors p21 and p27 (326) and the expression of the cell adhesion molecule E-cadherin (150), while inhibiting the transcriptional activity of β-catenin (150,327,328). In keratinocytes, 1,25(OH)2D has been shown to promote the repair of DNA damage induced by ultraviolet radiation (UVR) (329) (330), reduce apoptosis while increasing survival after UVR (331), and increase p53 (332).  Epidemiologic evidence supporting the importance of adequate vitamin D nutrition (including sunlight exposure) for the prevention of a number of cancers (333-337) is extensive. Although numerous types of cancers show reduction (338), most attention has been paid to cancers of the breast, colon, and prostate. I (339) recently reviewed a number of meta-analyses of epidemiologic studies evaluating the association of vitamin D intake and/or 25OHD levels and the risk of developing these cancers.  The data supporting a reduction in risk for developing colorectal cancer and breast cancer in premenopausal females with higher vitamin D intake or higher serum 25OHD levels were considerably stronger than that for the prevention of prostate cancer. Prospective randomized controlled trial data are limited. In a prospective 4 yr. trial with 1100iu vitamin D and 1400-1500 mg calcium originally designed to look at osteoporosis the authors showed a 77% reduction in cancers after excluding the initial year of study (340), including a reduction in both breast and colon cancers. In this study, vitamin D supplementation raised the 25OHD levels from a mean of 28.8ng/ml to 38.4ng/ml with no changes in the placebo or calcium only arms of the study. However, this was a relatively small study in which cancer prevention was not the primary outcome variable. A substantially larger trial involving over 25,000 subjects treated in a two by two design with vitamin D and/or omega 3 fatty acid did not find a benefit of vitamin supplementation with respect to cancer incidence but appears to have shown a beneficial effect on mortality (341). Trials of 1,25(OH)2D and its analogs for the treatment of cancer have been disappointing. In a small study involving 7 subjects with prostate cancer treated with doses of 1,25(OH)2D up to 2.5µg for 6-15 months, 6/7 showed a decrease in the rise of prostate specific antigen (PSA), a marker of tumor progression (342), and one patient showed a decline. However, hypercalciuria was common and limiting. A preliminary report of a larger study involving 250 patients with prostate cancer using 45µg 1,25(OH)2D  weekly in combination with docetaxel demonstrated a non-significant decline in PSA, although survival was significantly improved (HR 0.67) (343). A larger follow-up study did not show increased survival (344).  The incidence of either hypercalcemia or hypercalciuria was not reported. Most likely until an analog of 1,25(OH)2D is developed which is both efficacious and truly non hypercalcemic, treatment of cancer with vitamin D metabolites will remain problematic.

SKIN 

 Epidermal keratinocytes are the only cells in the body with the entire vitamin D metabolic pathway. As described earlier, production of vitamin D3 from 7-dehydrocholesterol takes place in the epidermis. However, the epidermis also contains CYP27A1 (345), the mitochondrial enzyme that 25-hydroxylates vitamin D, and CYP27B1 (40,47), the enzyme that produces 1,25(OH)2D from 25OHD. The CYP27B1 in keratinocytes is differently regulated than CYP27B1 in renal cells. Although PTH stimulates CYP27B1 activity in the keratinocyte, the mechanism appears to be independent of cAMP (346). Cytokines such as tumor necrosis factor-α and interferon-γ stimulate CYP27B1 activity (347,348). 1,25(OH)2D does not exert a direct effect on CYP27B1 expression in keratinocytes, but regulates 1,25(OH)2D levels by inducing CYP24A1 thus initiating the catabolism of 1,25(OH)2D (79). CYP27B1 is expressed primarily in the basal cells of the epidermis (50); as the cells differentiate the mRNA and protein levels of CYP27B1and its activity decline (349).

1,25(OH)2D regulates keratinocyte differentiation in part by modulating the ability of calcium to do likewise (350). Therefore, it is important to understand the actions of calcium on this cell prior to examining the influence of 1,25(OH)2D (351-356)(357). If keratinocytes are grown at calcium concentrations below 0.07mM, they continue to proliferate but either fail or are slow to develop intercellular contacts, stratify little if at all, and fail or are slow to form cornified envelopes. Acutely increasing the extracellular calcium concentration (Cao) above 0.1mM (calcium switch) leads to the rapid redistribution of desmoplakin, cadherins, integrins, catenins, plakoglobulin, vinculin, and actinin from the cytosol to the membrane where they participate in the formation of intercellular contacts. Calcium also stimulates the redistribution to the membrane of protein kinase Cα (PKCα) (358,359) and the tyrosine-phosphorylated p62 associated protein of ras GAP (360,361) where they further the calcium signaling process. These early events are accompanied by a rearrangement of actin filaments from a perinuclear to a radial pattern which if disrupted blocks the redistribution of these proteins and blocks the differentiation process. Within hours of the calcium switch keratinocytes switch from making the basal keratins K5 and K14 and begin making keratins K1 and K10 (356) followed, subsequently, by increased levels of profilaggrin (the precursor of filaggrin, an intermediate filament associated protein), involucrin and loricrin (precursors for the cornified envelope) (362,363). Loricrin, involucrin and other proteins (364) are cross linked into the insoluble cornified envelope by the calcium sensitive, membrane bound form of transglutaminase (365,366), which like involucrin and loricrin increases within 24 hours after the calcium switch (367). Within 1-2 days of the calcium switch cornified envelope formation is apparent (355,368), paralleling transglutaminase activation (369). The induction of these proteins represents a genomic action (likely indirect) of calcium as indicated by a calcium induced increase in mRNA levels and transcription rates (356,363,369,370). The relevance of calcium induced differentiation in vitro to the in vivo situation is indicated by the steep gradient of calcium within the epidermis, with the highest levels in the uppermost (most differentiated) nucleated layers (371). Current evidence for the importance of calcium in epidermal function is that barrier disruption, which results in increased proliferation, is associated with loss of the calcium gradient, whereas increasing the calcium concentration in the epidermis with sonophoresis stimulates lamellar body secretion (372-376).

The keratinocyte senses calcium via a seven transmembrane domain, G protein coupled receptor (CaSR) (377) originally cloned from the parathyroid cell by Brown et al (378,379). Knocking out the CaSR blocks calcium induced differentiation in vitro (380,381) and in vivo (382). However, keratinocytes also produce an alternatively spliced variant of the CaSR as they differentiate (383). This variant CaSR lacks exon 5 and so would be missing residues 461-537 in the extracellular domain. A mouse model in which the full length CaSR has been knocked out continues to produce the alternatively spliced form of CaSR, but its epidermis contains lower levels of the terminal differentiation markers loricrin and profilaggrin, and keratinocytes from these mice fail to respond normally to calcium (383) consistent with the results when the full length calcium receptor was deleted in vitro (380,381). We have produced a conditional knockout of the CaSR allowing us to delete CaSR in the tissue of choice using cell specific cre recombinases that avoids the problem with the original global knockout (384). When the CaSR is deleted specifically in the keratinocyte, this mouse has a reduction in epidermal differentiation and barrier repair (382), but unlike the global knockout does not have abnormalities in overall calcium homeostasis, and rather than showing an increased calcium gradient in the epidermis has a blunted one. The conditional knockout mouse also lacks the alternatively spliced CaSR.

Inositol 1,4,5 tris phosphate (IP3) and diacylglycerol levels increase within seconds to minutes after the calcium switch implicating activation of the phospholipase C (PLC) pathway (385,386). Similar to intracellular calcium levels (Cai), the levels of inositol phosphates (IPs) remain elevated for hours after the calcium switch. The prolonged increase in IPs after the calcium switch may contribute to the plateau phase of Cai elevation and a prolonged elevation of diacylglycerol (DG) that would stimulate the protein kinase C (PKC) pathway. This prolonged increase in IPs appears to be due to calcium induction and activation of PLC (154,386,387), especially PLC-γ1.  Activation of PLC-γ1 by calcium involves a chain of events involving src kinase activation of phosphatidyl inositol 3 kinase and phosphatidyl inositol 4 phosphate 5  kinase 1α within the context of a membrane complex with E-cadherin leading to the formation of phosphatidyl inositol tris phosphate in the membrane which activates PLC-γ1 via its PH domain (388).  Phosphorylation of PLC-γ1 is not part of its activation by calcium unlike its activation by EGF (389). Knocking out Plcg1 blocks the ability of calcium to increase Cai and to induce involucrin and transglutaminase (387). Thus, like CaSR, PLC-γ1 is critical for the ability of calcium to regulate keratinocyte differentiation.

Phorbol esters, which bind to and activate PKC, are well known tumor promoters in skin However, the initial effects of phorbol esters in vitro are to promote differentiation in cells grown in low calcium (358,390,391), effects which are potentiated by calcium (383). Phorbol esters stimulate PKC, and PKC inhibitors block the ability of both calcium and phorbol esters to promote differentiation (391). Phorbol esters as well as calcium stimulate the expression of both keratin 1 and involucrin gene constructs each of which contains an AP-1 site within the calcium response element (CaRE) of the promoter for these genes (392,393). If the AP-1 site within the CaRE is mutated, neither calcium nor phorbol esters are effective (392,393). These CaREs also contain VDREs (DR3), which at least in the involucrin gene has been shown to mediate 1,25(OH)2D regulation of this gene (394). Phorbol esters do not reproduce all the actions of calcium on the keratinocyte, and vice versa, but cross talk between their signaling pathways is clearly present.

The observation that 1,25(OH)2D induces keratinocyte differentiation was first made by Hosomi et al. (395) and provided a rationale for the previous and unexpected finding of 1,25(OH)2D receptors in the epidermis (396). 1,25(OH)2D increases the mRNA and protein levels for involucrin and transglutaminase, and promotes CE formation at subnanomolar concentrations in preconfluent keratinocytes (370,397-399). Calcium affects the ability of 1,25(OH)2D to stimulate keratinocyte differentiation, and vice versa. Calcium in the absence of 1,25(OH)2D and 1,25(OH)2D at low (0.03mM) calcium raise the mRNA levels for involucrin and transglutaminase in a dose dependent fashion by stimulating gene expression. The stimulation of mRNA levels by calcium and 1,25(OH)2D is synergistic at early time points; however, longer periods of incubation lead to a paradoxical fall in the mRNA levels for these proteins. This is due to the fact that although transcription is increased by calcium and 1,25(OH)2D, stability of the mRNA is reduced in cells incubated with calcium and 1,25(OH)2D.

The transcriptional regulation by 1,25(OH)2D is both direct and indirect. Several genes contain VDREs (e.g. involucrin), but VDREs have not been found in all genes that are regulated by 1,25(OH)2D. Inhibition of PKC activity or mutation of the AP-1 site in the CaRE of the involucrin gene also blocks the ability of 1,25(OH)2D to regulate expression of involucrin (394). The ability of 1,25(OH)2D to increase intracellular calcium (Cai) (298) accounts for at least part of the ability of 1,25(OH)2D to induce differentiation. A rapid (presumably nongenomic) effect of 1,25(OH)2D on Cai has been described (400), although this response is controversial (398). Our studies indicate that the ability of 1,25(OH)2D to increase Cai requires time and gene transcription. 1,25(OH)2D increases CaSR mRNA levels and prevents their fall in cells grown in 0.03mM calcium (401). This results in an enhanced Cai response to extracellular calcium (Cao). 1,25(OH)2D also induces the family of PLCs (402). PLC-γ1 contains a VDRE in its promoter (154), which unlike the usual VDRE is a DR6 which binds VDR/RAR rather than VDR/RXR. Knocking out PLCG1 blocks 1,25(OH)2D induced differentiation (403) as well as calcium induced differentiation mentioned earlier. The other PLCs have not been studied as extensively, but are likely to show similar means of regulation by 1,25(OH)2D.

Our current working model for the mechanisms by which calcium and 1,25(OH)2D regulate keratinocyte differentiation is shown in figure 8. The keratinocyte expresses a CaSR that by coupling to and activating PLC controls the production of two important second messengers, IP3 and DG. PLC-β is likely to be activated acutely by CaSR via a G protein coupled mechanism, whereas PLC-γ1 is activated acutely by calcium stimulated non receptor tyrosine kinases and subsequently by PIP3 in the membrane. Both PLCs are induced by calcium and 1,25(OH)2D. IP3 stimulates the release of calcium from intracellular stores thus raising Cai. The initial release of calcium from these stores activates the Stim1/Orai1 channel in the membrane (404) that may stimulate proliferation of the basal keratinocytes and initiate their movement out of the basal layer. The increase in Cai and DG stimulates the activation of critical PKCs and their translocation to membrane receptors (RACK). PKC-α appears to be the most critical PKC for the subsequent events triggered by calcium in the keratinocyte, although PKCδ has also been implicated.  Activated PKC leads to the induction and activation of AP-1 transcription factors which regulate the transcription of a number of genes including keratin 1, transglutaminase, involucrin, loricrin, and profilaggrin required for the differentiation process. Activation of the CaSR also activates the RhoA kinase leading to activation of src kinases which by phosphorylating various catenins leads to the formation of the Ecadherin/catenin complex in the membrane (405). This complex recruits both PI3K and PIP5K1α required to maintain the PIP2 and PIP3 levels in the membrane (357). PIP3 activates PLC-γ, that is in turn activates the TRPC channels in the membrane to enable the prolonged increase in Cai required for differentiation (406). 1,25(OH)2D, which is produced by the keratinocyte in a highly regulated fashion, modulates calcium regulated differentiation at several steps. First, 1,25(OH)2D increases CaSR expression, thus making the cell more responsive to calcium. Secondly, 1,25(OH)2D induces all the PLCs again increasing the responsiveness of the cell to calcium. Finally, 1,25(OH)2D has a direct effect on the transcription of the genes such as involucrin. The net result is that both calcium and 1,25(OH)2D promote keratinocyte differentiation through interactive mechanisms.

Figure 8. A model of 1,25(OH)2D and calcium regulated keratinocyte differentiation. The G-protein coupled calcium receptor (CaSR) when activated by extracellular calcium activates Gα as described in the legend to figure 6. Gα stimulates PLC mediated hydrolysis of PIP2 to IP3 and DG. IP3 releases Cai from intracellular stores, and DG activates PKC. Depletion of intracellular calcium stores leads to influx of calcium across store operated calcium channels. PKC stimulation leads to activation of AP-1 transcription factors which along with calcium and 1,25(OH)2D activated transcription factors stimulate the expression of genes essential for the differentiation process. 1,25(OH)2D regulates this process by inducing CaSR and PLC as well as genes essential for cornified envelope formation such as involucrin and transglutaminase.

The VDR is also critical for hair follicle (HF) cycling. Unlike epidermal differentiation, hair follicle cycling is not dependent on 1,25(OH)2D. Alopecia is a well described characteristic of mice and humans lacking VDR (125,126,407) due to failure to regenerate the cycling lower portion of the HF after the initial developmental cycle is completed. Deletion of CYP27B1 (408) and CaSR (382) do not result in alopecia. Cianferotti et al. (409) attributed the loss of HF cycling in VDR null mice to a gradual loss of the proliferative potential in the stem cells of the HF bulge region. However, this conclusion has been challenged by Palmer et al. (410), who attributed the failure of HF cycling in the VDR null mouse in part to a failure of the progeny of these stem cells to migrate out of the bulge rather than their loss of proliferative potential suggesting a loss of activation. The role of VDR in the stem cells that regulate both HF cycling and epidermal regeneration is also important in the skin wound healing process. When the skin is wounded the progeny of stem cells from all regions of the HF and interfollicular epidermis (IFE) contribute at least initially (411,412), although the stem cells in the IFE make the most lasting contribution. Tian et al. (413) observed that topical 1,25(OH)2D enhanced wound healing, suggesting that unlike HF cycling, the wound repair required this VDR ligand. Luderer et al. (414) observed that in the global VDRKO, there was a reduction in TGFβ signaling in the dermis, and subsequently demonstrated that the VDR in macrophages but not in keratinocytes was responsible for macrophage recruitment during the inflammatory phase of cutaneous wound healing (415). Our studies have focused on the VDR in epidermal keratinocytes.  We have observed that re-epithelialization by the keratinocytes over the wound is impaired when the deletion of VDR from keratinocytes is accompanied by either a low calcium diet or a deletion of the CaSR (416). Thus like the role of calcium and CaSR in vitamin D regulated keratinocyte differentiation so a similar synergism is seen in wound healing. These results are consistent with the loss of E-cadherin/catenin complex formation in the VDRKO keratinocyte, a complex that maintains stem cells in their niches (417), regulates when stem cell division is symmetric (to maintain stem cell numbers) or asymmetric (initiating differentiation) (418), and is essential for the ability of keratinocytes to migrate as a sheet to re-epithelialize the wound (419). As noted previously calcium and the CaSR along with 1,25(OH)2D and VDR are required for E-cadherin/catenin complex formation during the differentiation process and so are involved in enabling its role in wound healing (420).

Immune System

The potential role for vitamin D and its active metabolite 1,25(OH)2D3 in modulating the immune response has long been recognized since the discovery of vitamin D receptors (VDR) in macrophages, dendritic cells (DC), and activated T and B lymphocytes, the ability of macrophages and DC as well as activated T and B cells to express CYP27B1, and the ability of 1,25(OH)2D3 to regulate the proliferation and function of these cells. While these are the key cells mediating the adaptive immune response, 1,25(OH)2D, VDR, and CYP27B1 are also expressed in a large number of epithelial cells which along with the aforementioned members of the adaptive immune response contribute to host defense by their innate immune response. The totality of the immune response involves both types of responses in complex interactions involving numerous cytokines. The regulation of these different responses and their interactions by 1,25(OH)2D3 is nuanced. In general, 1,25(OH)2D3 enhances the innate immune response primarily via its ability to stimulate cathelicidin, an antimicrobial peptide important in defense against invading organisms, whereas it inhibits the adaptive immune response primarily by inhibiting the maturation of dendritic cells (DC) important for antigen presentation, reducing T cell proliferation, and shifting the balance of T cell differentiation from the Th1 and Th17 pathways to Th2 and Treg pathways. Inflammatory autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis involve Th17 activation, a cell that expresses RANKL, and so can drive osteoclastogenesis leading to bone loss.     

ADAPTIVE IMMUNE RESPONSE

The adaptive immune response is initiated by cells specialized in antigen presentation, DC and macrophages in particular, activating the cells responsible for subsequent antigen recognition, T and B lymphocytes. These cells are capable of a wide repertoire of responses that ultimately determine the nature and duration of the immune response. Activation of T and B cells occurs after a priming period in tissues of the body, e.g., lymph nodes, distant from the site of the initial exposure to the antigenic substance, and is marked by proliferation of the activated T and B cells accompanied by post translational modifications of immunoglobulin production that enable the cellular response to adapt specifically to the antigen presented. Importantly, the type of T cell activated, CD4 or CD8, or within the helper T cell class Th1, Th2, Th17, Treg, and subtle variations of those, is dependent on the context of the antigen presented by which cell and in what environment. Systemic factors such as vitamin D influence this process. Vitamin D in general exerts an inhibitory action on the adaptive immune system. 1,25(OH)2D3 decreases the maturation of DC as marked by inhibited expression of the costimulatory molecules HLA-DR, CD40, CD80, and CD86, decreasing their ability to present antigen and so activate T cells (421). Furthermore, by suppressing IL-12 production, important for Th1 development, and IL-23 and IL-6 production important for Th17 development and function, 1,25(OH)2D3 inhibits the development of Th1 cells capable of producing IFN- and IL-2, and Th17 cells producing IL-17 (422). These actions prevent further antigen presentation to and recruitment of T lymphocytes (role of IFN-γ), and T lymphocyte proliferation (role of IL-2).  Suppression of IL-12 increases the development of Th2 cells leading to increased IL-4, IL-5, and IL-13 production, which further suppresses Th1 development shifting the balance to a Th2 cell phenotype. Treatment of DCs with 1,25(OH)2D3 can also induce CD4+/CD25+ regulatory T cells (Treg) cells (423) as shown by increased FoxP3 expression, critical for Treg development.  These cells produce IL-10, which suppresses the development of the other Th subclasses. Treg are critical for the induction of immune tolerance (424).  In addition, 1,25(OH)2D3 alters the homing of properties of T cells for example by inducing expression of CCR10, the receptor for CCL27, a keratinocyte specific cytokine, while suppressing that of CCR9, a gut homing receptor (425). The actions of 1,25(OH)2D3 on B cells have received less attention, but recent studies have demonstrated a reduction in proliferation, maturation to plasma cells and immunoglobulin production (426). 

 

1,25(OH)2D3 has both direct and indirect effects on regulation of a number of cytokines involved with the immune response (review in (427)). TNF has a VDRE in its promoter to which the VDR/RXR complex binds.  1,25(OH)2D3 both blocks the activation of NFκB via an increase in IκBα expression and impedes its binding to its response elements in the genes such as IL-8 and IL-12 that it regulates. 1,25(OH)2D3 has also been shown to bring an inhibitor complex containing histone deacetylase 3 (HDAC3) to the promoter of rel B, one of the members of the NFκB family, thus suppressing gene expression. Thus, TNF/NFkB activity is markedly impaired by 1,25(OH)2D3 at multiple levels. In VDR null fibroblasts, NFκB activity is enhanced. Furthermore, 1,25(OH)2D3 suppresses IFNγ, and a negative VDRE has been found in the IFNγ promoter. GM-CSF is regulated by VDR monomers binding to a repressive complex in the promoter of this gene, competing with nuclear factor of T cells 1(NFAT1) for binding to the promoter.

The ability of 1,25(OH)2D3 to suppress the adaptive immune system appears to be beneficial for a number of conditions in which the immune system is directed at self—i.e. autoimmunity (review in (428)). In a number of experimental models including inflammatory arthritis, psoriasis, autoimmune diabetes (e.g., NOD mice), systemic lupus erythematosis (SLE), experimental allergic encephalitis (EAE) (a model for multiple sclerosis), inflammatory bowel disease (IBD), prostatitis, and thyroiditis VDR agonist administration has prevented and/or treated the disease process. As will be discussed later, a number of these conditions are associated with bone loss either directly (e.g., inflammatory arthritis) or indirectly presumably via increased serum levels of inflammatory cytokines. These actions of 1,25(OH)2D3 were originally ascribed to inhibition of Th1 function, but Th17 cells have also been shown to play important roles in a number of these conditions including psoriasis (321),  experimental colitis (422), and rheumatoid arthritis (429), conditions that respond to 1,25(OH)2D3 and its analogs. Although few prospective, randomized, placebo-controlled trials in humans have been performed, epidemiologic and case control studies indicate that a number of these diseases in humans are favorably impacted by adequate vitamin D levels. For example, the incidence of multiple sclerosis correlates inversely with 25OHD levels and vitamin D intake, and early studies suggested benefit in the treatment of patients with rheumatoid arthritis and multiple sclerosis with VDR agonists (427,428). Similarly, IBD is associated with low vitamin D levels (430). Children who are vitamin D deficient have a higher risk of developing type 1 diabetes mellitus, and supplementation with vitamin D during early childhood reduces the risk of developing type 1 diabetes (review in (421)).  In VDR null mice myelopoiesis and the composition of lymphoid organs are normal, although a number of abnormalities in the immune response have been found.  Some of the abnormalities in macrophage function and T cell proliferation in response to anti-CD3 stimulation in these animals could be reversed by placing the animals on a high calcium diet to normalize serum calcium (431), indicating the important role of calcium in vitamin D regulated immune function as in skeletal development and maintenance, hormone regulation, and keratinocyte differentiation. Other studies have noted an increased number of mature DCs in the lymph nodes of VDR null mice, which would be expected to promote the adaptive immune response (432). Somewhat surprisingly, RANKL also increases the number and retention of DCs in lymph nodes (433) suggesting that at least this mechanism is not mediated via the RANKL/RANK system in VDR null mice, which I will discuss at length subsequently.  In contrast to these inhibitory actions of 1,25(OH)2D3, Th2 function as indicated by increased IgE stimulated histamine from mast cells is increased in VDR null mice (434). The IL-10 null mouse model of IBD shows an accelerated disease profile when bred with the VDR null mouse with increased expression of Th1 cytokines (435). Surprisingly, despite a reduction in natural killer T cells and Treg cells and a decreased number of mature DCs, VDR null mice bred with NOD mice do not show accelerated development of diabetes (436). Part of the difference in tissue response in VDR null mice may relate to differences in the ability of 1,25(OH)2D3 to alter the homing of T cells to the different tissues (425).  In allergic airway disease (asthma) Th2 cells, not Th1 cells, dominate the inflammatory response. 1,25(OH)2D3 administration to normal mice protected these mice from experimentally induced asthma in one study, blocking eosinophil infiltration, IL-4 production, and limiting histologic evidence of inflammation (437).  However, a study with VDR null mice using a comparable method of inducing asthma showed that lack of VDR also protected the mice from an inflammatory response in their lungs (438). In an extension of this study the investigators showed that wildtype (WT) splenocytes were only minimally successful at restoring experimental airway inflammation to VDR null mice, whereas splenocytes from these mice were able to transfer experimental airway inflammation to the unprimed WT host (439). Thus, the impact of vitamin D signaling on adaptive immunity depends on the specifics of the immune response being evaluated. 

Inhibition of the adaptive immune response may also have benefit in transplantation procedures (440).  In experimental allograft models of the aorta, bone, bone marrow, heart, kidney, liver, pancreatic islets, skin, and small bowel VDR agonists have shown benefit generally in combination with other immunosuppressive agents such as cyclosporine, tacrolimus, sirolimus, and glucocorticoids (440). Much of the effect could be attributed to a reduction in infiltration of Th1 cells, macrophages and DC into the grafted tissue associated with a reduction in chemokines such as CXCL10, CXCL9, CCL2, and CCL5.  CXCL10, the ligand for CXCR3, may be of particular importance for acute rejection in a number of tissues, whereas CXCL9 as well as CXCL10 (both CXCR3 ligands) may be more important for chronic rejection at least in the heart and kidney, respectively. Although there are no prospective trials of the use of VDR agonists in transplant patients, several retrospective studies in patients with renal transplants treated with 1,25(OH)2D3 have suggested benefit with respect to prolonged graft survival and reduced numbers of acute rejection episodes.

Suppression of the adaptive immune system may not be without a price. Several publications have demonstrated that for some infections including Leishmania major (441) and toxoplasmosis (442), 1,25(OH)2D3 promotes the infection (442), while the mouse null for VDR is protected (441). This may be due at least in part to loss of IFNγ stimulation of ROS and NO production required for macrophage antimicrobial activity (441). Furthermore, atopic dermatitis, a disease associated with increased Th2 activity (443), and allergic airway disease, likewise associated with increased Th2 activity, (437-439), may be aggravated by 1,25(OH)2D3 and less severe in animals null for VDR.

THE INNATE IMMUNE RESPONSE

The innate immune response involves the activation of toll-like receptors (TLRs) in polymorphonuclear cells (PMNs), monocytes and macrophages as well as in a number of epithelial cells including those of the epidermis, gingiva, intestine, vagina, bladder and lungs (review in (444)). There are 10 functional TLRs in human cells (of 11 known mammalian TLRs). TLRs are an extended family of host noncatalytic transmembrane pathogen-recognition receptors that interact with specific membrane patterns (PAMP) shed by infectious agents that trigger the innate immune response in the host. A number of these TLRs signal through adapter molecules such as myeloid differentiation factor-88 (MyD88) and the TIR-domain containing adapter inducing IFN-β (TRIF).  MyD88 signaling includes translocation of NFkB to the nucleus, leading to the production and secretion of a number of inflammatory cytokines. TRIF signaling leads to the activation of interferon regulatory factor-3 (IRF-3) and the induction of type 1 interferons such as IFNβ.  MyD88 mediates signaling from TLRs 2, 4, 5, 7 and 9, whereas TRIF mediates signaling from TLR 3 and 4. TLR1/2, TLR4, TLR5, TLR2/6 respond to bacterial ligands, whereas, TLR3, TLR7, and TLR 8 respond to viral ligands. The TLR response to fungi is less well defined. CD14 serves as a coreceptor for a number of these TLRs. Activation of TLRs leads to the induction of antimicrobial peptides (AMPs) and reactive oxygen species, which kill the organism. Among these AMPs is cathelicidin. Cathelicidin plays a number of roles in the innate immune response. The precursor protein, hCAP18, must be cleaved to its major peptide LL-37 to be active. In addition to its antimicrobial properties, LL-37 can stimulate the release of cytokines such as IL-6 and IL-10 through G protein coupled receptors, and IL-18 through ERK/P38 pathways, stimulate the EGF receptor leading to activation of STAT1 and 3, induce the chemotaxis of neutrophils, monocytes, macrophages, and T cells into the skin, and promote keratinocyte proliferation and migration (445). The expression of this antimicrobial peptide is induced by 1,25(OH)2D3 in both myeloid and epithelial cells (446,447).  In addition, 1,25(OH)2D3 induces the coreceptor CD14 in keratinocytes(448). Stimulation of TLR2 by infectious organisms like tuberculosis in macrophages (449) or stimulation of TLR2 in keratinocytes by wounding the epidermis (448) results in increased expression of CYP27B1, which in the presence of adequate substrate (25OHD) stimulates the expression of cathelicidin.  Lack of substrate (25OHD) or lack of CYP27B1 blunts the ability of these cells to respond to a challenge with respect to cathelicidin and/or CD14 production (447-449). In diseases such as atopic dermatitis, the production of cathelicidin and other antimicrobial peptides (AMPs) is reduced, predisposing these patients to microbial superinfections (450). Th2 cytokines such as IL-4 and 13 suppress the induction of AMPs(451). Since 1,25(OH)2D3 stimulates the differentiation of Th2 cells, in this disease 1,25(OH)2D3 administration may be harmful.  An important role of these AMPs besides their antimicrobial properties is to help link the innate and adaptive immune response. This interplay is well demonstrated in SARS-CoV-19 infections in which a dysfunctional and/or delayed innate immune response can lead to an unchecked adaptive immune response resulting in a massive release of proinflammatory cytokines, the “cytokine storm”, leading to destruction of the lungs and death (452). Patients with vitamin D deficiency appear to be more vulnerable to this infection (453).

Although many cells are capable of the innate immune response including bone cells, most studies have focused on the macrophage and the keratinocyte. Vitamin D regulation of the innate immune response in these two cell types is comparable, but differences exist.

Macrophages

The importance of adequate vitamin D nutrition for resistance to infection has long been appreciated but poorly understood. This has been especially true for tuberculosis. Indeed, prior to the development of specific drugs for the treatment of tuberculosis, getting out of the city into fresh air and sunlight was the treatment of choice. In a recent survey of patients with tuberculosis in London (454) 56% had undetectable 25OHD levels, and an additional 20% had detectable levels but below 9 ng/ml (22 nM).  In 1986 Rook et al. (455) demonstrated that 1,25(OH)2D3 could inhibit the growth of Mycobacterium tuberculosis.  The mechanism for this remained unclear until the publication by Liu et al. (449) of their results in macrophages. They observed that activation of the Toll-like receptor TLR2/1 by a lipoprotein extracted from M. tuberculosis reduced the viability of intracellular M. tuberculosis in human monocytes and macrophages concomitant with increased expression of the VDR and of CYP27B1 in these cells. Killing of M. tuberculosis occurred only when the serum in which the cells were cultured contained adequate levels of 25OHD, the substrate for CYP27B1. This provided clear evidence for the importance of vitamin D nutrition (as manifested by adequate serum levels of 25OHD) in preventing and treating this disease, and demonstrated the critical role for endogenous production of 1,25(OH)2D3 by the macrophage to enable its antimycobacterial capacity.  Activation of TLR2/1 or directly treating these cells with 1,25(OH)2D3 induced the antimicrobial peptide cathelicidin, which is toxic for M. tuberculosis. If induction of cathelicidin is blocked as with siRNA, the ability of 1,25(OH)2D3 to enhance the killing of M. tuberculosis is prevented (456). Furthermore, 1,25(OH)2D3 also induces the production of reactive oxygen species which if blocked likewise prevents the anti-mycobacterial activity of 1,25(OH)2D3 treated macrophages (457). The murine cathelicidin gene lacks a known VDR response element in its promoter, and so might not be expected to be induced by 1,25(OH)2D3 in mouse cells, yet 1,25(OH)2D3 stimulates antimycobacterial activity in murine macrophages. Murine macrophages, unlike human macrophages, utilize inducible nitric oxide synthase (iNOS) for their TLR and 1,25(OH)2D3 mediated killing of M. tuberculosis (457,458). Clinical trials attempting to treat tuberculosis patients with high levels of vitamin D have shown mixed results (459)(460).

Keratinocytes

Cathelicidiin and CD14 expression in epidermal keratinocytes is induced by 1,25(OH)2D3 (445,448).  In these cells butyrate, which by itself has little effect, potentiates the ability of 1,25(OH)2D3 to induce cathelicidin (461).  Keratinocytes treated with 1,25(OH)2D3 are substantially more effective in killing Staphyococcus aureus than are untreated keratinocytes. Wounding the epidermis induces the expression of TLR2 and that of its co-receptor CD14 and cathelicidin (448). This does not occur in mice lacking CYP27B1 (448). Unlike macrophages, 1,25(OH)2D3 stimulates TLR2 expression in keratinocytes as well as in the epidermis when applied topically (448) providing a feed forward loop to amplify the innate immune response. Wounding also increases the expression of CYP27B1.  This may occur as a result of increased levels of cytokines such as TNF-α and IFN-γ, both of which we have shown stimulate 1,25(OH)2D3 production, as well as by TGF-β and the TLR2 ligand Malp-2 (448). When the levels of VDR or one of its principal coactivators, SRC3, are reduced using siRNA technology, the ability of 1,25(OH)2D3 to induce cathelicidin and CD14 expression in human keratinocytes is markedly blunted (461).

Other Tissues

The VDR is widespread (127,462) (reviews). In some of these tissues the functional significance of the VDR and/or the effect of 1,25(OH)2D are unclear. Since several of the functions regulated by 1,25(OH)2D in some of these tissues may have clinical relevance, this section will focus on a select number of these tissues. 

HEART

A reduction in contractility has been observed in vitamin D deficient animals (463). This may be due to lack of vitamin D or the accompanying hypocalcemia and hypophosphatemia. However, in vitro 1,25(OH)2D stimulates calcium uptake by cardiac muscle cells (464,465). In addition, 1,25(OH)2D inhibits the expression of atrial naturetic factor, one of the few genes with a negative VDRE in its promoter (466).  Deletion of the VDR specifically in cardiac muscle leads to hypertrophy and fibrosis (467). Low circulating levels of 25OHD are associated with increased risk of myocardial infarction in men [436]. However, a large randomized clinical trial failed to show a protective effective of vitamin D supplementation to individuals with normal levels of 25OHD with respect to cardiovascular disease (341) 

SKELETAL MUSCLE 

Proximal muscle weakness is a hallmark of vitamin D deficiency, and reduced high energy substrates (ATP, creatinine phosphate) have been observed in that condition (468). Myoblasts contain VDR, although the expression of VDR in mature muscle cells is controversial. Muscle weakness may reflect the lower levels of calcium and phosphate rather than a reduction in 1,25(OH)2D. However, evidence for a direct role of 1,25(OH)2D and VDR in muscle function is increasing (469). Moreover, 1,25(OH)2D may have actions on muscle that do not require the VDR, at least the genomic functions of VDR. The Boland laboratory (470) has demonstrated acute effects of 1,25(OH)2D on calcium uptake, PLC, PLA2, PLD, PKC, and adenylate cyclase activities, all of which may alter muscle function.

PITUITARY

VDR have been found primarily in thyrotropes in vivo and in GH and prolactin secreting cell lines in vitro (471,472). 1,25(OH)2D increases TRH stimulated TSH secretion by a mechanism involving increased Cai and IP3 production (473,474), suggesting that induction of PLC by 1,25(OH)2D may be involved. 

BREAST

The breast contains VDR (475), and vitamin D plays a role in normal breast development (476). Moreover, breast cancer cells also contain VDR (477), and 1,25(OH)2D and its analogs reduce their proliferation in vivo and in vitro (478,479). This has obvious clinical implications for the treatment of breast cancer.

LIVER

Low levels of VDR have been found in the liver, particularly in stellate cells (480,481). Hepatic regeneration is impaired in vitamin D deficient animals, even when the serum calcium is normalized by a high calcium diet (482), suggesting a role for 1,25(OH)2D in hepatic cell growth and in the prevention of hepatic fibrosis (481).

LUNG 

VDR have been found in type II epithelial pneumocytes (483). 1,25(OH)2D stimulates their maturation including increased phospholipid production and surfactant release [437].These results are consistent with the abnormal alveolar development observed in pups born to vitamin D deficient mothers (484). In addition 1,25(OH)2D stimulates the innate immune response in bronchial epithelial cells and may provide protection in patients with cystic fibrosis with recurrent lung infections as well as in patient with Covid-19 infections (452,485) as discussed previously.

REFERENCES

 

  1. Whistler D. De morbo puerli anglorum, quem patrio ideiomate

indigenae vocant "the rickets". Journal of History of Medicine. 1645;5:397-415.

  1. Mellanby E. An experimental investigation on rickets. Lancet. 1919;1:407-412.
  2. McCollum EV, Simmonds N, Becker JE, Shipley PG. An experimental demonstration of the existence of a vitamin which promotes calcium deposition. Journal of Biological Chemistry. 1922;53:293-298.
  3. Steenbock H, Black A. Fat-soluble vitamins. XVII. The induction of growth-promoting and calcifying properties in a ration by exposure to ultraviolet light. Journal of Biological Chemistry. 1924;61:405-422.
  4. Huldshinsky K. Heilung von rachitis durch kunstalich hohensonne. Deut Med Wochenschr. 1919;45:712-713.
  5. Hess AF, Unger LF. Cure of infantile rickets by sunlight. Journal of The American Medical Association. 1921;77:39.
  6. Askew FA, Bourdillon RB, Bruce HM, Jenkins RGC, Webster TA. The distillation of vitamin D. Proceedings of the Royal Society. 1931;B107:76-90.
  7. Windaus A, Schenck F, von Werder F. Uber das antirachitisch wirksame bestrahlungs-produkt aus 7-dehydro-cholesterin. Hoppe-Seylers Z Physiological Chemistry. 1936;241:100-103.
  8. Prabhu AV, Luu W, Sharpe LJ, Brown AJ. Cholesterol-mediated Degradation of 7-Dehydrocholesterol Reductase Switches the Balance from Cholesterol to Vitamin D Synthesis. The Journal of biological chemistry. 2016;291(16):8363-8373.
  9. Holick MF, McLaughlin JA, Clark MB, Doppelt SH. Factors that influence the cutaneous photosynthesis of previtamin D3. Science. 1981;211:590-593.
  10. Holick MF, McLaughlin JA, Clark MB, Holick SA, J.T. PJ, Anderson RR, Blank IH, Parrish JA. Photosynthesis of previtamin D3 in human and the physiologic consequences. Science. 1980;210:203-205.
  11. Holick MF, Richtand NM, McNeill SC, Holick SA, Frommer JE, Henley JW, Potts JT, Jr. Isolation and identification of previtamin D3 from the skin of rats exposed to ultraviolet irradiation. Biochemistry. 1979;18(6):1003-1008.
  12. Webb AR, DeCosta BR, Holick MF. Sunlight regulates the cutaneous production of vitamin D3 by causing its photodegradation. Journal of Clinical Endocrinology and Metabolism. 1989;68(5):882-887.
  13. Bell NH, Greene A, Epstein S, Oexmann MJ, Shaw S, Shary J. Evidence for alteration of the vitamin D-endocrine system in blacks. Journal of Clinical Investigation. 1985;76(2):470-473.
  14. Webb AR, Kline L, Holick MF. Influence of season and latitude on the cutaneous synthesis of vitamin D3: exposure to winter sunlight in Boston and Edmonton will not promote vitamin D3 synthesis in human skin. Journal of Clinical Endocrinology and Metabolism. 1988;67(2):373-378.
  15. Matsuoka LY, Wortsman J, Dannenberg MJ, Hollis BW, Lu Z, Holick MF. Clothing prevents ultraviolet-B radiation-dependent photosynthesis of vitamin D3. The Journal of clinical endocrinology and metabolism. 1992;75(4):1099-1103.
  16. Matsuoka LY, Ide L, Wortsman J, MacLaughlin JA, Holick MF. Sunscreens suppress cutaneous vitamin D3 synthesis. Journal of Clinical Endocrinology and Metabolism. 1987;64(6):1165-1168.
  17. Zhu JG, Ochalek JT, Kaufmann M, Jones G, Deluca HF. CYP2R1 is a major, but not exclusive, contributor to 25-hydroxyvitamin D production in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(39):15650-15655.
  18. Thacher TD, Fischer PR, Singh RJ, Roizen J, Levine MA. CYP2R1 Mutations Impair Generation of 25-hydroxyvitamin D and Cause an Atypical Form of Vitamin D Deficiency. The Journal of clinical endocrinology and metabolism. 2015;100(7):E1005-1013.
  19. Andersson S, Davis DL, Dahlbäck H, Jörnvall H, Russell DW. Cloning, structure, and expression of the mitochondrial cytochrome P-450 sterol 26-hydroxylase, a bile acid biosynthetic enzyme. Journal of Biological Chemistry. 1989;264(14):8222-8229.
  20. Usui E, Noshiro M, Okuda K. Molecular cloning of cDNA for vitamin D3 25-hydroxylase from rat liver mitochondria. Febs Letters. 1990;262(1):135-138.
  21. Cali JJ, Russell DW. Characterization of human sterol 27-hydroxylase. A mitochondrial cytochrome P-450 that catalyzes multiple oxidation reaction in bile acid biosynthesis. Journal of Biological Chemistry. 1991;266(12):7774-7778.
  22. Ichikawa F, Sato K, Nanjo M, Nishii Y, Shinki T, Takahashi N, Suda T. Mouse primary osteoblasts express vitamin D3 25-hydroxylase mRNA and convert 1 alpha-hydroxyvitamin D3 into 1 alpha,25-dihydroxyvitamin D3. Bone. 1995;16(1):129-135.
  23. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. Journal of Biological Chemistry. 1991;266(12):7779-7783.
  24. Leitersdorf E, Reshef A, Meiner V, Levitzki R, Schwartz SP, Dann EJ, Berkman N, Cali JJ, Klapholz L, Berginer VM. Frameshift and splice-junction mutations in the sterol 27-hydroxylase gene cause cerebrotendinous xanthomatosis in Jews or Moroccan origin. Journal of Clinical Investigation. 1993;91(6):2488-2496.
  25. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D. Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis [see comments]. Metabolism: Clinical and Experimental. 1993;42(1):69-74.
  26. Leitersdorf E, Safadi R, Meiner V, Reshef A, Björkhem I, Friedlander Y, Morkos S, Berginer VM. Cerebrotendinous xanthomatosis in the Israeli Druze: molecular genetics and phenotypic characteristics. American Journal of Human Genetics. 1994;55(5):907-915.
  27. Guo YD, Strugnell S, Jones G. Identification of a human liver mitochondrial cytochrome P-450 cDNA corresponding to the vitamin D3-25-hydroxylase. Journal of Bone and Mineral Research. 1991;6:S120.
  28. Guo YD, Strugnell S, Back DW, Jones G. Transfected human liver cytochrome P-450 hydroxylates vitamin D analogs at different side-chain positions. Proc Natl Acad Sci U S A. 1993;90(18):8668-8672.
  29. Cheng JB, Motola DL, Mangelsdorf DJ, Russell DW. De-orphanization of cytochrome P450 2R1: a microsomal vitamin D 25-hydroxilase. The Journal of biological chemistry. 2003;278(39):38084-38093.
  30. Cheng JB, Levine MA, Bell NH, Mangelsdorf DJ, Russell DW. Genetic evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(20):7711-7715.
  31. Theodoropoulos C, Demers C, Mirshahi A, Gascon-Barre M. 1,25-Dihydroxyvitamin D(3) downregulates the rat intestinal vitamin D(3)-25-hydroxylase CYP27A. Am J Physiol Endocrinol Metab. 2001;281(2):E315-325.
  32. Axen E, Postlind H, Wikvall K. Effects on CYP27 mRNA expression in rat kidney and liver by 1 alpha, 25- dihydroxyvitamin D3, a suppressor of renal 25-hydroxyvitamin D3 1 alpha- hydroxylase activity. Biochem Biophys Res Commun. 1995;215(1):136-141.
  33. Vlahcevic ZR, Jairath SK, Heuman DM, Stravitz RT, Hylemon PB, Avadhani NG, Pandak WM. Transcriptional regulation of hepatic sterol 27-hydroxylase by bile acids. Am J Physiol. 1996;270(4 Pt 1):G646-652.
  34. Twisk J, Hoekman MF, Lehmann EM, Meijer P, Mager WH, Princen HM. Insulin suppresses bile acid synthesis in cultured rat hepatocytes by down-regulation of cholesterol 7 alpha-hydroxylase and sterol 27- hydroxylase gene transcription. Hepatology. 1995;21(2):501-510.
  35. Stravitz RT, Vlahcevic ZR, Russell TL, Heizer ML, Avadhani NG, Hylemon PB. Regulation of sterol 27-hydroxylase and an alternative pathway of bile acid biosynthesis in primary cultures of rat hepatocytes. J Steroid Biochem Mol Biol. 1996;57(5-6):337-347.
  36. Roizen JD, Long C, Casella A, O'Lear L, Caplan I, Lai M, Sasson I, Singh R, Makowski AJ, Simmons R, Levine MA. Obesity Decreases Hepatic 25-Hydroxylase Activity Causing Low Serum 25-Hydroxyvitamin D. J Bone Miner Res. 2019:e3686.
  37. Aatsinki SM, Elkhwanky MS, Kummu O, Karpale M, Buler M, Viitala P, Rinne V, Mutikainen M, Tavi P, Franko A, Wiesner RJ, Chambers KT, Finck BN, Hakkola J. Fasting-Induced Transcription Factors Repress Vitamin D Bioactivation, a Mechanism for Vitamin D Deficiency in Diabetes. Diabetes. 2019;68(5):918-931.
  38. Saarem K, Pedersen JI. Sex differences in the hydroxylation of cholecalciferol and of 5 beta-cholestane-3 alpha, 7 alpha, 12 alpha-triol in rat liver. Biochemical Journal. 1987;247(1):73-78.
  39. Fu GK, Lin D, Zhang MY, Bikle DD, Shackleton CH, Miller WL, Portale AA. Cloning of human 25-hydroxyvitamin D-1 alpha-hydroxylase and mutations causing vitamin D-dependent rickets type 1. Mol Endocrinol. 1997;11(13):1961-1970.
  40. Takeyama K, Kitanaka S, Sato T, Kobori M, Yanagisawa J, Kato S. 25-Hydroxyvitamin D3 1alpha-hydroxylase and vitamin D synthesis. Science. 1997;277(5333):1827-1830.
  41. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glorieux FH. The 25-hydroxyvitamin D 1-alpha-hydroxylase gene maps to the pseudovitamin D-deficiency rickets (PDDR) disease locus. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1997;12(10):1552-1559.
  42. Shinki T, Shimada H, Wakino S, Anazawa H, Hayashi M, Saruta T, DeLuca HF, Suda T. Cloning and expression of rat 25-hydroxyvitamin D3-1alpha-hydroxylase cDNA. Proc Natl Acad Sci U S A. 1997;94(24):12920-12925.
  43. Kitanaka S, Takeyama K, Murayama A, Sato T, Okumura K, Nogami M, Hasegawa Y, Niimi H, Yanagisawa J, Tanaka T, Kato S. Inactivating mutations in the 25-hydroxyvitamin D3 1alpha-hydroxylase gene in patients with pseudovitamin D-deficiency rickets. N Engl J Med. 1998;338(10):653-661.
  44. Wang JT, Lin CJ, Burridge SM, Fu GK, Labuda M, Portale AA, Miller WL. Genetics of vitamin D 1alpha-hydroxylase deficiency in 17 families. Am J Hum Genet. 1998;63(6):1694-1702.
  45. Dardenne O, Prud'homme J, Arabian A, Glorieux FH, St-Arnaud R. Targeted inactivation of the 25-hydroxyvitamin D(3)-1(alpha)- hydroxylase gene (CYP27B1) creates an animal model of pseudovitamin D- deficiency rickets. Endocrinology. 2001;142(7):3135-3141.
  46. Bikle DD, Nemanic MK, Whitney JO, Elias PW. Neonatal human foreskin keratinocytes produce 1,25-dihydroxyvitamin D3. Biochemistry. 1986;25(7):1545-1548.
  47. Murayama A, Takeyama K, Kitanaka S, Kodera Y, Kawaguchi Y, Hosoya T, Kato S. Positive and negative regulations of the renal 25-hydroxyvitamin D3 1alpha-hydroxylase gene by parathyroid hormone, calcitonin, and 1alpha,25(OH)2D3 in intact animals. Endocrinology. 1999;140(5):2224-2231.
  48. Panda DK, Al Kawas S, Seldin MF, Hendy GN, Goltzman D. 25-hydroxyvitamin D 1alpha-hydroxylase: structure of the mouse gene, chromosomal assignment, and developmental expression. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2001;16(1):46-56.
  49. Zehnder D, Bland R, Williams MC, McNinch RW, Howie AJ, Stewart PM, Hewison M. Extrarenal expression of 25-hydroxyvitamin d(3)-1 alpha-hydroxylase. The Journal of clinical endocrinology and metabolism. 2001;86(2):888-894.
  50. Bikle DD. Extra renal synthesis of 1,25 dihydroxyvitamin D and its Health Implications. Clin Rev in Bone and Min Metab. 2009;7:114-125.
  51. Bikle DD, Pillai S. Vitamin D, calcium, and epidermal differentiation. Endocrine Review. 1993;14:3-19.
  52. Horiuchi N, Suda T, Takahashi H, Shimazawa E, Ogata E. In vivo evidence for the intermediary role of 3',5'-cyclic AMP in parathyroid hormone-induced stimulation of 1alpha,25-dihydroxyvitamin D3 synthesis in rats. Endocrinology. 1977;101(3):969-974.
  53. Rasmussen H, Wong M, Bikle D, Goodman DB. Hormonal control of the renal conversion of 25-hydroxycholecalciferol to 1,25-dihydroxycholecalciferol. The Journal of clinical investigation. 1972;51(9):2502-2504.
  54. Rost CR, Bikle DD, Kaplan RA. In vitro stimulation of 25-hydroxycholecalciferol 1 alpha-hydroxylation by parathyroid hormone in chick kidney slices: evidence for a role for adenosine 3',5'-monophosphate. Endocrinology. 1981;108(3):1002-1006.
  55. Henry HL. Parathyroid hormone modulation of 25-hydroxyvitamin D3 metabolism by cultured chick kidney cells is mimicked and enhanced by forskolin. Endocrinology. 1985;116(2):503-510.
  56. Armbrecht HJ, Forte LR, Wongsurawat N, Zenser TV, Davis BB. Forskolin increases 1,25-dihydroxyvitamin D3 production by rat renal slices in vitro. Endocrinology. 1984;114(2):644-649.
  57. Janulis M, Tembe V, Favus MJ. Role of protein kinase C in parathyroid hormone stimulation of renal 1,25-dihydroxyvitamin D3 secretion. The Journal of clinical investigation. 1992;90(6):2278-2283.
  58. Janulis M, Wong MS, Favus MJ. Structure-function requirements of parathyroid hormone for stimulation of 1,25-dihydroxyvitamin D3 production by rat renal proximal tubules. Endocrinology. 1993;133(2):713-719.
  59. Brenza HL, Kimmel-Jehan C, Jehan F, Shinki T, Wakino S, Anazawa H, Suda T, DeLuca HF. Parathyroid hormone activation of the 25-hydroxyvitamin D3-1alpha- hydroxylase gene promoter. Proc Natl Acad Sci U S A. 1998;95(4):1387-1391.
  60. Zierold C, Nehring JA, DeLuca HF. Nuclear receptor 4A2 and C/EBPbeta regulate the parathyroid hormone-mediated transcriptional regulation of the 25-hydroxyvitamin D3-1alpha-hydroxylase. Archives of biochemistry and biophysics. 2007;460(2):233-239.
  61. Takeyama K, Kato S. The vitamin D3 1alpha-hydroxylase gene and its regulation by active vitamin D3. Biosci Biotechnol Biochem. 2011;75(2):208-213.
  62. Bikle DD, Murphy EW, Rasmussen H. The ionic control of 1,25-dihydroxyvitamin D3 synthesis in isolated chick renal mitochondria. The role of calcium as influenced by inorganic phosphate and hydrogen-ion. The Journal of clinical investigation. 1975;55(2):299-304.
  63. Bushinsky DA, Riera GS, Favus MJ, Coe FL. Evidence that blood ionized calcium can regulate serum 1,25(OH)2D3 independently of parathyroid hormone and phosphorus in the rat. The Journal of clinical investigation. 1985;76(4):1599-1604.
  64. Hulter HN, Halloran BP, Toto RD, Peterson JC. Long-term control of plasma calcitriol concentration in dogs and humans. Dominant role of plasma calcium concentration in experimental hyperparathyroidism. The Journal of clinical investigation. 1985;76(2):695-702.
  65. Hughes MR, Brumbaugh PF, Hussler MR, Wergedal JE, Baylink DJ. Regulation of serum 1alpha,25-dihydroxyvitamin D3 by calcium and phosphate in the rat. Science. 1975;190(4214):578-580.
  66. Portale AA, Halloran BP, Morris RC, Jr. Dietary intake of phosphorus modulates the circadian rhythm in serum concentration of phosphorus. Implications for the renal production of 1,25-dihydroxyvitamin D. The Journal of clinical investigation. 1987;80(4):1147-1154.
  67. Condamine L, Menaa C, Vrtovsnik F, Vztovsnik F, Friedlander G, Garabedian M. Local action of phosphate depletion and insulin-like growth factor 1 on in vitro production of 1,25-dihydroxyvitamin D by cultured mammalian kidney cells. The Journal of clinical investigation. 1994;94(4):1673-1679.
  68. Gray RW. Control of plasma 1,25-(OH)2-vitamin D concentrations by calcium and phosphorus in the rat: effects of hypophysectomy. Calcif Tissue Int. 1981;33(5):485-488.
  69. Yoshida T, Yoshida N, Monkawa T, Hayashi M, Saruta T. Dietary phosphorus deprivation induces 25-hydroxyvitamin D(3) 1alpha- hydroxylase gene expression. Endocrinology. 2001;142(5):1720-1726.
  70. Gray RW, Garthwaite TL, Phillips LS. Growth hormone and triiodothyronine permit an increase in plasma 1,25(OH)2D concentrations in response to dietary phosphate deprivation in hypophysectomized rats. Calcif Tissue Int. 1983;35(1):100-106.
  71. Halloran BP, Spencer EM. Dietary phosphorus and 1,25-dihydroxyvitamin D metabolism: influence of insulin-like growth factor I. Endocrinology. 1988;123(3):1225-1229.
  72. Saito H, Kusano K, Kinosaki M, Ito H, Hirata M, Segawa H, Miyamoto K, Fukushima N. Human fibroblast growth factor-23 mutants suppress Na+-dependent phosphate co-transport activity and 1alpha,25-dihydroxyvitamin D3 production. The Journal of biological chemistry. 2003;278(4):2206-2211.
  73. White KE, Evans WE, O'Riordan JLH, Speer MC, Econs MJ, Lorenz-Depiereux B, Grabowski M, Meitinger T, Strom TM. Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nature Genetics. 2000;26:345.
  74. Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(11):6500-6505.
  75. Blau JE, Collins MT. The PTH-Vitamin D-FGF23 axis. Reviews in endocrine & metabolic disorders. 2015;16(2):165-174.
  76. Meyer MB, Benkusky NA, Kaufmann M, Lee SM, Redfield RR, Jones G, Pike JW. Targeted genomic deletions identify diverse enhancer functions and generate a kidney-specific, endocrine-deficient Cyp27b1 pseudo-null mouse. The Journal of biological chemistry. 2019;294(24):9518-9535.
  77. Schuster I, Egger H, Astecker N, Herzig G, Schussler M, Vorisek G. Selective inhibitors of CYP24: mechanistic tools to explore vitamin D metabolism in human keratinocytes. Steroids. 2001;66(3-5):451-462.
  78. Xie Z, Munson S, Huang N, Schuster I, Portale AA, Miller WL, Bikle DD. The mechanism of 1,25-Dihydroxyvitamin D3 auto-regulation in keratinocytes. j bone min Res (Program & abstracts). 2001;16 suppl 1:S556.
  79. Kato S, Fujiki R, Kim MS, Kitagawa H. Ligand-induced transrepressive function of VDR requires a chromatin remodeling complex, WINAC. The Journal of steroid biochemistry and molecular biology. 2007;103(3-5):372-380.
  80. Akiyoshi-Shibata M, Sakaki T, Ohyama Y, Noshiro M, Okuda K, Yabusaki Y. Further oxidation of hydroxycalcidiol by calcidiol 24-hydroxylase. A study with the mature enzyme expressed in Escherichia coli. Eur J Biochem. 1994;224(2):335-343.
  81. Jones G, Prosser DE, Kaufmann M. 25-Hydroxyvitamin D-24-hydroxylase (CYP24A1): its important role in the degradation of vitamin D. Arch Biochem Biophys. 2012;523(1):9-18.
  82. Schlingmann KP, Kaufmann M, Weber S, Irwin A, Goos C, John U, Misselwitz J, Klaus G, Kuwertz-Broking E, Fehrenbach H, Wingen AM, Guran T, Hoenderop JG, Bindels RJ, Prosser DE, Jones G, Konrad M. Mutations in CYP24A1 and idiopathic infantile hypercalcemia. N Engl J Med. 2011;365(5):410-421.
  83. Shah AD, Hsiao EC, O'Donnell B, Salmeen K, Nussbaum R, Krebs M, Baumgartner-Parzer S, Kaufmann M, Jones G, Bikle DD, Wang Y, Mathew AS, Shoback D, Block-Kurbisch I. Maternal Hypercalcemia Due to Failure of 1,25-Dihydroxyvitamin-D3 Catabolism in a Patient With CYP24A1 Mutations. The Journal of clinical endocrinology and metabolism. 2015;100(8):2832-2836.
  84. St-Arnaud R, Arabian A, Travers R, Barletta F, Raval-Pandya M, Chapin K, Depovere J, Mathieu C, Christakos S, Demay MB, Glorieux FH. Deficient mineralization of intramembranous bone in vitamin D-24- hydroxylase-ablated mice is due to elevated 1,25-dihydroxyvitamin D and not to the absence of 24,25-dihydroxyvitamin D. Endocrinology. 2000;141(7):2658-2666.
  85. Hahn CN, Kerry DM, Omdahl JL, May BK. Identification of a vitamin D responsive element in the promoter of the rat cytochrome P450(24) gene. Nucleic Acids Res. 1994;22(12):2410-2416.
  86. Chen KS, DeLuca HF. Cloning of the human 1 alpha,25-dihydroxyvitamin D-3 24-hydroxylase gene promoter and identification of two vitamin D-responsive elements. Biochim Biophys Acta. 1995;1263(1):1-9.
  87. Ohyama Y, Ozono K, Uchida M, Shinki T, Kato S, Suda T, Yamamoto O, Noshiro M, Kato Y. Identification of a vitamin D-responsive element in the 5'-flanking region of the rat 25-hydroxyvitamin D3 24-hydroxylase gene. The Journal of biological chemistry. 1994;269(14):10545-10550.
  88. Chen ML, Boltz MA, Armbrecht HJ. Effects of 1,25-dihydroxyvitamin D3 and phorbol ester on 25- hydroxyvitamin D3 24-hydroxylase cytochrome P450 messenger ribonucleic acid levels in primary cultures of rat renal cells. Endocrinology. 1993;132(4):1782-1788.
  89. Pike JW, Kerner SA, Jin CH, Allegretto EA, Elgort M. Direct activation of the human 25-(OH)2D3 and PMA: Identification of cis elements and transactivators. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1995;10:S144.
  90. Garabedian M, Holick MF, Deluca HF, Boyle IT. Control of 25-hydroxycholecalciferol metabolism by parathyroid glands. Proc Natl Acad Sci U S A. 1972;69(7):1673-1676.
  91. Shigematsu T, Horiuchi N, Ogura Y, Miyahara T, Suda T. Human parathyroid hormone inhibits renal 24-hydroxylase activity of 25- hydroxyvitamin D3 by a mechanism involving adenosine 3',5'- monophosphate in rats. Endocrinology. 1986;118(4):1583-1589.
  92. Armbrecht HJ, Wongsurawat N, Zenser TV, Davis BB. Effect of PTH and 1,25(OH)2D3 on renal 25(OH)D3 metabolism, adenylate cyclase, and protein kinase. Am J Physiol. 1984;246(1 Pt 1):E102-107.
  93. Armbrecht HJ, Hodam TL, Boltz MA, Partridge NC, Brown AJ, Kumar VB. Induction of the vitamin D 24-hydroxylase (CYP24) by 1,25-dihydroxyvitamin D3 is regulated by parathyroid hormone in UMR106 osteoblastic cells. Endocrinology. 1998;139(8):3375-3381.
  94. Armbrecht HJ, Wongsurawat VJ, Hodam TL, Wongsurawat N. Insulin markedly potentiates the capacity of parathyroid hormone to increase expression of 25-hydroxyvitamin D3-24-hydroxylase in rat osteoblastic cells in the presence of 1,25-dihydroxyvitamin D3. FEBS Lett. 1996;393(1):77-80.
  95. Meyer MB, Lee SM, Carlson AH, Benkusky NA, Kaufmann M, Jones G, Pike JW. A chromatin-based mechanism controls differential regulation of the cytochrome P450 gene Cyp24a1 in renal and non-renal tissues. The Journal of biological chemistry. 2019;294(39):14467-14481.
  96. Inoue Y, Segawa H, Kaneko I, Yamanaka S, Kusano K, Kawakami E, Furutani J, Ito M, Kuwahata M, Saito H, Fukushima N, Kato S, Kanayama HO, Miyamoto K. Role of the vitamin D receptor in FGF23 action on phosphate metabolism. Biochem J. 2005;390(Pt 1):325-331.
  97. Wu S, Grieff M, Brown AJ. Regulation of renal vitamin D-24-hydroxylase by phosphate: effects of hypophysectomy, growth hormone and insulin-like growth factor I. Biochem Biophys Res Commun. 1997;233(3):813-817.
  98. Cooke NE, Haddad JG. Vitamin D binding protein (Gc-globulin). Endocrine reviews. 1989;10(3):294-307.
  99. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D levels in serum from normal subjects, pregnant subjects, and subjects with liver disease. The Journal of clinical investigation. 1984;74(6):1966-1971.
  100. Bikle DD, Siiteri PK, Ryzen E, Haddad JG. Serum protein binding of 1,25-dihydroxyvitamin D: a reevaluation by direct measurement of free metabolite levels. The Journal of clinical endocrinology and metabolism. 1985;61(5):969-975.
  101. Bikle DD, Gee E, Halloran B, Kowalski MA, Ryzen E, Haddad JG. Assessment of the free fraction of 25-hydroxyvitamin D in serum and its regulation by albumin and the vitamin D-binding protein. The Journal of clinical endocrinology and metabolism. 1986;63(4):954-959.
  102. Bikle DD, Halloran BP, Gee E, Ryzen E, Haddad JG. Free 25-hydroxyvitamin D levels are normal in subjects with liver disease and reduced total 25-hydroxyvitamin D levels. The Journal of clinical investigation. 1986;78(3):748-752.
  103. Madden K, Feldman HA, Chun RF, Smith EM, Sullivan RM, Agan AA, Keisling SM, Panoskaltsis-Mortari A, Randolph AG. Critically Ill Children Have Low Vitamin D-Binding Protein, Influencing Bioavailability of Vitamin D. Ann Am Thorac Soc. 2015;12(11):1654-1661.
  104. Nielson CM, Jones KS, Chun RF, Jacobs JM, Wang Y, Hewison M, Adams JS, Swanson CM, Lee CG, Vanderschueren D, Pauwels S, Prentice A, Smith RD, Shi T, Gao Y, Schepmoes AA, Zmuda JM, Lapidus J, Cauley JA, Bouillon R, Schoenmakers I, Orwoll ES, Osteoporotic Fractures in Men Research G. Free 25-Hydroxyvitamin D: Impact of Vitamin D Binding Protein Assays on Racial-Genotypic Associations. The Journal of clinical endocrinology and metabolism. 2016;101(5):2226-2234.
  105. Pettifor JM, Bikle DD, Cavaleros M, Zachen D, Kamdar MC, Ross FP. Serum levels of free 1,25-dihydroxyvitamin D in vitamin D toxicity. Ann Intern Med. 1995;122(7):511-513.
  106. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, Liebhaber SA, Cooke NE. Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D binding protein. The Journal of clinical investigation. 1999;103(2):239-251.
  107. Zella LA, Shevde NK, Hollis BW, Cooke NE, Pike JW. Vitamin D-binding protein influences total circulating levels of 1,25-dihydroxyvitamin D3 but does not directly modulate the bioactive levels of the hormone in vivo. Endocrinology. 2008;149(7):3656-3667.
  108. Henderson CM, Fink SL, Bassyouni H, Argiropoulos B, Brown L, Laha TJ, Jackson KJ, Lewkonia R, Ferreira P, Hoofnagle AN, Marcadier JL. Vitamin D-Binding Protein Deficiency and Homozygous Deletion of the GC Gene. The New England journal of medicine. 2019;380(12):1150-1157.
  109. Bikle D, Bouillon R, Thadhani R, Schoenmakers I. Vitamin D metabolites in captivity? Should we measure free or total 25(OH)D to assess vitamin D status? The Journal of steroid biochemistry and molecular biology 2017;173:105-116.
  110. Malmstroem S, Rejnmark L, Imboden JB, Shoback DM, Bikle DD. Current Assays to Determine Free 25-Hydroxyvitamin D in Serum. J AOAC Int 2017;100:1323-1327.
  111. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz J, Melsen F, Christensen EI, Willnow TE. An endocytic pathway essential for renal uptake and activation of the steroid 25-(OH) vitamin D3. Cell. 1999;96(4):507-515.
  112. Arnaud J, Constans J. Affinity differences for vitamin D metabolites associated with the genetic isoforms of the human serum carrier protein (DBP). Hum Genet. 1993;92(2):183-188.
  113. Cooke NE, David EV. Serum vitamin D-binding protein is a third member of the albumin and alpha fetoprotein gene family. The Journal of clinical investigation. 1985;76(6):2420-2424.
  114. Cooke NE, Levan G, Szpirer J. The rat vitamin D binding protein (Gc-globulin) gene is syntenic with the rat albumin and alpha-fetoprotein genes on chromosome 14. Cytogenet Cell Genet. 1987;44(2-3):98-100.
  115. Guha C, Osawa M, Werner PA, Galbraith RM, Paddock GV. Regulation of human Gc (vitamin D--binding) protein levels: hormonal and cytokine control of gene expression in vitro. Hepatology. 1995;21(6):1675-1681.
  116. Lees A, Haddad JG, Lin S. Brevin and DBP comparison of the effects of two serum protein on actin assembly and disassembly. Biochemistry. 1984;23:3038-3047.
  117. Yamamoto N, Homma S, Haddad JG, Kowalski MA. Vitamin D3 binding protein required for in vitro activation of macrophages after alkylglycerol treatment of mouse peritoneal cells. Immunology. 1991;74(3):420-424.
  118. Schneider GB, Benis KA, Flay NW, Ireland RA, Popoff SN. Effects of vitamin D binding protein-macrophage activating factor (DBP- MAF) infusion on bone resorption in two osteopetrotic mutations. Bone. 1995;16(6):657-662.
  119. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, Liebhaber SA, Cooke NE. Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D binding protein. The Journal of clinical investigation. 1999;103(2):239-251.
  120. Haussler MR, Norman AW. Chromosomal receptor for a vitamin D metabolite. Proc Natl Acad Sci U S A. 1969;62(1):155-162.
  121. McDonnell DP, Mangelsdorf DJ, Pike JW, Haussler MR, O'Malley BW. Molecular cloning of complementary DNA encoding the avian receptor for vitamin D. Science. 1987;235(4793):1214-1217.
  122. Baker AR, McDonnell DP, Hughes M, Crisp TM, Mangelsdorf DJ, Haussler MR, Pike JW, Shine J, O'Malley BW. Cloning and expression of full-length cDNA encoding human vitamin D receptor. Proc Natl Acad Sci U S A. 1988;85(10):3294-3298.
  123. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA, Pike JW, Feldman D, O'Malley BW. Point mutations in the human vitamin D receptor gene associated with hypocalcemic rickets. Science. 1988;242(4886):1702-1705.
  124. Yoshizawa T, Handa Y, Uematsu Y, Takeda S, Sekine K, Yoshihara Y, Kawakami T, Arioka K, Sato H, Uchiyama Y, Masushige S, Fukamizu A, Matsumoto T, Kato S. Mice lacking the vitamin D receptor exhibit impaired bone formation, uterine hypoplasia and growth retardation after weaning. Nat Genet. 1997;16(4):391-396.
  125. Li YC, Pirro AE, Amling M, Delling G, Baron R, Bronson R, Demay MB. Targeted ablation of the vitamin D receptor: an animal model of vitamin D-dependent rickets type II with alopecia. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(18):9831-9835.
  126. Walters MR. Newly identified actions of the vitamin D endocrine system. Endocr Rev. 1992;13(4):719-764.
  127. Harris SS, Eccleshall TR, Gross C, Dawson-Hughes B, Feldman D. The vitamin D receptor start codon polymorphism (FokI) and bone mineral density in premenopausal American black and white women. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1997;12(7):1043-1048.
  128. Rachez C, Gamble M, Chang CP, Atkins GB, Lazar MA, Freedman LP. The DRIP complex and SRC-1/p160 coactivators share similar nuclear receptor binding determinants but constitute functionally distinct complexes. Mol Cell Biol. 2000;20(8):2718-2726.
  129. Makowski A, Brzostek S, Cohen RN, Hollenberg AN. Determination of nuclear receptor corepressor interactions with the thyroid hormone receptor. Molecular endocrinology (Baltimore, Md). 2003;17(2):273-286.
  130. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D. The crystal structure of the nuclear receptor for vitamin D bound to its natural ligand. Mol Cell. 2000;5(1):173-179.
  131. Orlov I, Rochel N, Moras D, Klaholz BP. Structure of the full human RXR/VDR nuclear receptor heterodimer complex with its DR3 target DNA. The EMBO journal. 2012;31(2):291-300.
  132. Wurtz JM, Bourguet W, Renaud JP, Vivat V, Chambon P, Moras D, Gronemeyer H. A canonical structure for the ligand-binding domain of nuclear receptors. Nat Struct Biol. 1996;3(1):87-94.
  133. Chen S, Cui J, Nakamura K, Ribeiro RC, West BL, Gardner DG. Coactivator-vitamin D receptor interactions mediate inhibition of the atrial natriuretic peptide promoter. The Journal of biological chemistry.275(20):15039-15048.
  134. McKenna NJ, Lanz RB, O'Malley BW. Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev. 1999;20(3):321-344.
  135. Polly P, Herdick M, Moehren U, Baniahmad A, Heinzel T, Carlberg C. VDR-Alien: a novel, DNA-selective vitamin D(3) receptor-corepressor partnership. Faseb J. 2000;14(10):1455-1463.
  136. Xie Z, Chang S, Oda Y, Bikle DD. Hairless suppresses vitamin D receptor transactivation in human keratinocytes. Endocrinology. 2006;147(1):314-323.
  137. Hsieh JC, Sisk JM, Jurutka PW, Haussler CA, Slater SA, Haussler MR, Thompson CC. Physical and functional interaction between the vitamin D receptor and hairless corepressor, two proteins required for hair cycling. The Journal of biological chemistry. 2003;278(40):38665-38674.
  138. Bikle DD, Elalieh H, Chang S, Xie Z, Sundberg JP. Development and progression of alopecia in the vitamin D receptor null mouse. J Cell Physiol. 2006;207:340-353.
  139. Leo C, Chen JD. The SRC family of nuclear receptor coactivators. Genes. 2000;245:1-11.
  140. Rachez C, Lemon BD, Suldan Z, Bromleigh V, Gamble M, Naar AM, Erdjument-Bromage H, Tempst P, Freedman LP. Ligand-dependent transcription activation by nuclear receptors requires the DRIP complex. Nature. 1999;398(6730):824-828.
  141. Teichert A, Arnold LA, Otieno S, Oda Y, Augustinaite I, Geistlinger TR, Kriwacki RW, Guy RK, Bikle DD. Quantification of the vitamin D receptor-coregulator interaction. Biochemistry. 2009;48(7):1454-1461.
  142. Oda Y. Abstract 809. Journal of Investigative Dermatology. 2001;116.
  143. Hawker NP, Pennypacker SD, Chang SM, Bikle DD. Regulation of Human Epidermal Keratinocyte Differentiation by the Vitamin D Receptor and its Coactivators DRIP205, SRC2, and SRC3. The Journal of investigative dermatology. 2007;127:874.
  144. Schauber J, Oda Y, Buchau AS, Yun QC, Steinmeyer A, Zugel U, Bikle DD, Gallo RL. Histone acetylation in keratinocytes enables control of the expression of cathelicidin and CD14 by 1,25-dihydroxyvitamin D3. J Invest Dermatol. 2008;128(4):816-824.
  145. Oda Y, Uchida Y, Moradian S, Crumrine D, Elias P, Bikle D. Vitamin D receptor and coactivators SRC 2 and 3 regulate epidermis-specific sphingolipid production and permeability barrier formation. J Invest Dermatol. 2009;129(6):1367-1378.
  146. Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI, Young RA. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell. 2013;153(2):307-319.
  147. Yanagisawa J, Yanagi Y, Masuhiro Y, Suzawa M, Watanabe M, Kashiwagi K, Toriyabe T, Kawabata M, Miyazono K, Kato S. Convergence of transforming growth factor-beta and vitamin D signaling pathways on SMAD transcriptional coactivators. Science. 1999;283(5406):1317-1321.
  148. Hsieh JC, Jurutka PW, Nakajima S, Galligan MA, Haussler CA, Shimizu Y, Shimizu N, Whitfield GK, Haussler MR. Phosphorylation of the human vitamin D receptor by protein kinase C. Biochemical and functional evaluation of the serine 51 recognition site. The Journal of biological chemistry. 1993;268(20):15118-15126.
  149. Palmer HG, Gonzalez-Sancho JM, Espada J, Berciano MT, Puig I, Baulida J, Quintanilla M, Cano A, de Herreros AG, Lafarga M, Munoz A. Vitamin D(3) promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of beta-catenin signaling. The Journal of cell biology. 2001;154(2):369-387.
  150. Palmer HG, Anjos-Afonso F, Carmeliet G, Takeda H, Watt FM. The Vitamin D Receptor Is a Wnt Effector that Controls Hair Follicle Differentiation and Specifies Tumor Type in Adult Epidermis. PloS one. 2008;3(1):e1483.
  151. Kurokawa R, Yu VC, Naar A, Kyakumoto S, Han Z, Silverman S, Rosenfeld MG, Glass CK. Differential orientations of the DNA-binding domain and carboxy- terminal dimerization interface regulate binding site selection by nuclear receptor heterodimers. Genes Dev. 1993;7(7B):1423-1435.
  152. Prufer K, Racz A, Lin GC, Barsony J. Dimerization with retinoid X receptors promotes nuclear localization and subnuclear targeting of vitamin D receptors. The Journal of biological chemistry. 2000;275(52):41114-41123.
  153. Xie Z, Bikle DD. Cloning of the human phospholipase C-gamma1 promoter and identification of a DR6-type vitamin D-responsive element. The Journal of biological chemistry. 1997;272(10):6573-6577.
  154. Schrader M, Muller KM, Nayeri S, Kahlen JP, Carlberg C. Vitamin D3-thyroid hormone receptor heterodimer polarity directs ligand sensitivity of transactivation. Nature. 1994;370(6488):382-386.
  155. Lemon BD, Freedman LP. Selective effects of ligands on vitamin D3 receptor- and retinoid X receptor-mediated gene activation in vivo. Mol Cell Biol. 1996;16(3):1006-1016.
  156. Pike JW, Meyer MB. The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D3. Rheumatic diseases clinics of North America. 2012;38(1):13-27.
  157. Carlberg C, Seuter S, Heikkinen S. The first genome-wide view of vitamin D receptor locations and their mechanistic implications. Anticancer research. 2012;32(1):271-282.
  158. MacDonald PN, Dowd DR, Nakajima S, Galligan MA, Reeder MC, Haussler CA, Ozato K, Haussler MR. Retinoid X receptors stimulate and 9-cis retinoic acid inhibits 1,25- dihydroxyvitamin D3-activated expression of the rat osteocalcin gene. Mol Cell Biol. 1993;13(9):5907-5917.
  159. Kang S, Li XY, Duell EA, Voorhees JJ. The retinoid X receptor agonist 9-cis-retinoic acid and the 24- hydroxylase inhibitor ketoconazole increase activity of 1,25- dihydroxyvitamin D3 in human skin in vivo. The Journal of investigative dermatology. 1997;108(4):513-518.
  160. Gill RK, Christakos S. Identification of sequence elements in mouse calbindin-D28k gene that confer 1,25-dihydroxyvitamin D3- and butyrate-inducible responses. Proc Natl Acad Sci U S A. 1993;90(7):2984-2988.
  161. Liu SM, Koszewski N, Lupez M, Malluche HH, Olivera A, Russell J. Characterization of a response element in the 5'-flanking region of the avian (chicken) PTH gene that mediates negative regulation of gene transcription by 1,25-dihydroxyvitamin D3 and binds the vitamin D3 receptor. Mol Endocrinol. 1996;10(2):206-215.
  162. Kremer R, Sebag M, Champigny C, Meerovitch K, Hendy GN, White J, Goltzman D. Identification and characterization of 1,25-dihydroxyvitamin D3- responsive repressor sequences in the rat parathyroid hormone-related peptide gene. The Journal of biological chemistry. 1996;271(27):16310-16316.
  163. Alroy I, Towers TL, Freedman LP. Transcriptional repression of the interleukin-2 gene by vitamin D3: direct inhibition of NFATp/AP-1 complex formation by a nuclear hormone receptor. Mol Cell Biol. 1995;15(10):5789-5799.
  164. Morley P, Whitfield JF, Vanderhyden BC, Tsang BK, Schwartz JL. A new, nongenomic estrogen action: the rapid release of intracellular calcium. Endocrinology. 1992;131(3):1305-1312.
  165. Wistrom CA, Meizel S. Evidence suggesting involvement of a unique human sperm steroid receptor/Cl- channel complex in the progesterone-initiated acrosome reaction. Dev Biol. 1993;159(2):679-690.
  166. Koenig H, Fan CC, Goldstone AD, Lu CY, Trout JJ. Polyamines mediate androgenic stimulation of calcium fluxes and membrane transport in rat heart myocytes. Circ Res. 1989;64(3):415-426.
  167. Orchinik M, Murray TF, Moore FL. A corticosteroid receptor in neuronal membranes. Science. 1991;252(5014):1848-1851.
  168. Segal J. Thyroid hormone action at the level of the plasma membrane. Thyroid. 1990;1(1):83-87.
  169. Caffrey JM, Farach-Carson MC. Vitamin D3 metabolites modulate dihydropyridine-sensitive calcium currents in clonal rat osteosarcoma cells. The Journal of biological chemistry. 1989;264(34):20265-20274.
  170. Baran DT, Sorensen AM, Honeyman TW, Ray R, Holick MF. 1 alpha,25-dihydroxyvitamin D3-induced increments in hepatocyte cytosolic calcium and lysophosphatidylinositol: inhibition by pertussis toxin and 1 beta,25-dihydroxyvitamin D3. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1990;5(5):517-524.
  171. Morelli S, de Boland AR, Boland RL. Generation of inositol phosphates, diacylglycerol and calcium fluxes in myoblasts treated with 1,25-dihydroxyvitamin D3. Biochem J. 1993;289(Pt 3):675-679.
  172. Wali RK, Baum CL, Sitrin MD, Brasitus TA. 1,25(OH)2 vitamin D3 stimulates membrane phosphoinositide turnover, activates protein kinase C, and increases cytosolic calcium in rat colonic epithelium. The Journal of clinical investigation. 1990;85(4):1296-1303.
  173. Khare S, Bolt MJ, Wali RK, Skarosi SF, Roy HK, Niedziela S, Scaglione-Sewell B, Aquino B, Abraham C, Sitrin MD, Brasitus TA, Bissonnette M. 1,25 dihydroxyvitamin D3 stimulates phospholipase C-gamma in rat colonocytes: role of c-Src in PLC-gamma activation. The Journal of clinical investigation. 1997;99(8):1831-1841.
  174. Nemere I, Yoshimoto Y, Norman AW. Calcium transport in perfused duodena from normal chicks: enhancement within fourteen minutes of exposure to 1,25-dihydroxyvitamin D3. Endocrinology. 1984;115(4):1476-1483.
  175. Nemere I, Norman AW. Rapid action of 1,25-dihydroxyvitamin D3 on calcium transport in perfused chick duodenum: effect of inhibitors. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1987;2(2):99-107.
  176. de Boland AR, Norman AW. Influx of extracellular calcium mediates 1,25-dihydroxyvitamin D3- dependent transcaltachia (the rapid stimulation of duodenal Ca2+ transport). Endocrinology. 1990;127(5):2475-2480.
  177. de Boland AR, Norman A. Evidence for involvement of protein kinase C and cyclic adenosine 3',5' monophosphate-dependent protein kinase in the 1,25-dihydroxy-vitamin D3- mediated rapid stimulation of intestinal calcium transport, (transcaltachia). Endocrinology. 1990;127(1):39-45.
  178. de Boland AR, Nemere I, Norman AW. Ca2(+)-channel agonist BAY K8644 mimics 1,25(OH)2-vitamin D3 rapid enhancement of Ca2+ transport in chick perfused duodenum. Biochem Biophys Res Commun. 1990;166(1):217-222.
  179. Nemere I, Dormanen MC, Hammond MW, Okamura WH, Norman AW. Identification of a specific binding protein for 1 alpha,25- dihydroxyvitamin D3 in basal-lateral membranes of chick intestinal epithelium and relationship to transcaltachia. The Journal of biological chemistry. 1994;269(38):23750-23756.
  180. Nemere I, Safford SE, Rohe B, DeSouza MM, Farach-Carson MC. Identification and characterization of 1,25D3-membrane-associated rapid response, steroid (1,25D3-MARRS) binding protein. The Journal of steroid biochemistry and molecular biology. 2004;89-90(1-5):281-285.
  181. Pedrozo HA, Schwartz Z, Rimes S, Sylvia VL, Nemere I, Posner GH, Dean DD, Boyan BD. Physiological importance of the 1,25(OH)2D3 membrane receptor and evidence for a membrane receptor specific for 24,25(OH)2D3. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1999;14(6):856-867.
  182. Khanal RC, Peters TM, Smith NM, Nemere I. Membrane receptor-initiated signaling in 1,25(OH)2D3-stimulated calcium uptake in intestinal epithelial cells. Journal of cellular biochemistry. 2008;105(4):1109-1116.
  183. Norman AW, Okamura WH, Farach-Carson MC, Allewaert K, Branisteanu D, Nemere I, Muralidharan KR, Bouillon R. Structure-function studies of 1,25-dihydroxyvitamin D3 and the vitamin D endocrine system. 1,25-dihydroxy-pentadeuterio-previtamin D3 (as a 6- s-cis analog) stimulates nongenomic but not genomic biological responses. The Journal of biological chemistry. 1993;268(19):13811-13819.
  184. Sequeira VB, Rybchyn MS, Tongkao-On W, Gordon-Thomson C, Malloy PJ, Nemere I, Norman AW, Reeve VE, Halliday GM, Feldman D, Mason RS. The role of the vitamin D receptor and ERp57 in photoprotection by 1alpha,25-dihydroxyvitamin D3. Molecular endocrinology (Baltimore, Md). 2012;26(4):574-582.
  185. Mizwicki MT, Keidel D, Bula CM, Bishop JE, Zanello LP, Wurtz JM, Moras D, Norman AW. Identification of an alternative ligand-binding pocket in the nuclear vitamin D receptor and its functional importance in 1alpha,25(OH)2-vitamin D3 signaling. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(35):12876-12881.
  186. Schachter D, Rosen S. Active transport of Ca45 by the small intestine and its dependence on vitamin D. Am J Physiol. 1959;196:357-362.
  187. Wasserman RH, Kallfelz FA, Comar CL. Active transport of calcium by rat duodenum in vivo. Science. 1961;133:883-884.
  188. Bikle DD. Regulation of intestinal calcium transport by vitamin D [1,25(OH)2]:role of membrane structure. In: Membrane transport and information storage. New York: Wiley-Liss. 1990, pp 191-219
  189. Wasserman RH, Fullmer CS. Vitamin D and intestinal calcium transport: facts, speculations and hypotheses. J Nutr. 1995;125(7 Suppl):1971S-1979S.
  190. Wongdee K, Chanpaisaeng K, Teerapornpuntakit J, Charoenphandhu N. Intestinal Calcium Absorption. Compr Physiol. 2021;11(3):2047-2073.
  191. Bikle DD, Shoback DM, Munson S. 1,25-dihydroxyvitamin D increases the intracellular free calcium concentration of duodenal epithelial cells. In: Vitamin D: Chemial, biochemical and clinical update. New York: Walter de Gruyter, 1985, p 416
  192. Morrissey RL, Zolock DT, Mellick PW, Bikle DD. Influence of cycloheximide and 1,25-dihydroxyvitamin D3 on mitochondrial and vesicle mineralization in the intestine. Cell Calcium. 1980;1:69-79.
  193. Davis WL, Hagler HK, Jones RG, Farmer GR, Cooper OJ, Martin JH, Bridges GE, Goodman DB. Cryofixation, ultracryomicrotomy, and X-ray microanalysis of enterocytes from chick duodenum: vitamin-D-induced formation of an apical tubulovesicular system. Anat Rec. 1991;229(2):227-239.
  194. Nemere I, Leathers V, Norman AW. 1,25-Dihydroxyvitamin D3-mediated intestinal calcium transport. Biochemical identification of lysosomes containing calcium and calcium- binding protein (calbindin-D28K). The Journal of biological chemistry. 1986;261(34):16106-16114.
  195. Max EE, Goodman DB, Rasmussen H. Purification and characterization of chick intestine brush border membrane. Effects of 1alpha(OH) vitamin D3 treatment. Biochimica et biophysica acta. 1978;511(2):224-239.
  196. Brasitus TA, Dudeja PK, Eby B, Lau K. Correction by 1,25(OH)2D3 of the abnormal fluidity and lipid composition of enterocyte brush border membranes in vitamin D-deprived rats. Journal of Biological Chemistry. 1981;256:3354-3360.
  197. Matsumoto T, Fontaine O, Rasmussen H. Effect of 1,25-dihydroxyvitamin D3 on phospholipid metabolism in chick duodenal mucosal cell. Relationship to its mechanism of action. The Journal of biological chemistry. 1981;256(7):3354-3360.
  198. Bikle DD, Whitney J, Munson S. The relationship of membrane fluidity to calcium flux in chick intestinal brush border membranes. Endocrinology. 1984;114(1):260-267.
  199. Peng JB, Chen XZ, Berger UV, Vassilev PM, Tsukaguchi H, Brown EM, Hediger MA. Molecular cloning and characterization of a channel-like transporter mediating intestinal calcium absorption. The Journal of biological chemistry. 1999;274(32):22739-22746.
  200. Hoenderop JG, van der Kemp AW, Hartog A, van de Graaf SF, van Os CH, Willems PH, Bindels RJ. Molecular identification of the apical Ca2+ channel in 1, 25- dihydroxyvitamin D3-responsive epithelia. The Journal of biological chemistry. 1999;274(13):8375-8378.
  201. Peng JB, Chen XZ, Berger UV, Vassilev PM, Brown EM, Hediger MA. A rat kidney-specific calcium transporter in the distal nephron. The Journal of biological chemistry. 2000;275(36):28186-28194.
  202. Muller D, Hoenderop JG, Meij IC, van den Heuvel LP, Knoers NV, den Hollander AI, Eggert P, Garcia-Nieto V, Claverie-Martin F, Bindels RJ. Molecular cloning, tissue distribution, and chromosomal mapping of the human epithelial Ca2+ channel (ECAC1). Genomics. 2000;67(1):48-53.
  203. Peng JB, Brown EM, Hediger MA. Structural conservation of the genes encoding cat1, cat2, and related cation channels. Genomics. 2001;76(1-3):99-109.
  204. Song Y, Peng X, Porta A, Takanaga H, Peng JB, Hediger MA, Fleet JC, Christakos S. Calcium transporter 1 and epithelial calcium channel messenger ribonucleic acid are differentially regulated by 1,25 dihydroxyvitamin D3 in the intestine and kidney of mice. Endocrinology. 2003;144(9):3885-3894.
  205. Bianco SD, Peng JB, Takanaga H, Suzuki Y, Crescenzi A, Kos CH, Zhuang L, Freeman MR, Gouveia CH, Wu J, Luo H, Mauro T, Brown EM, Hediger MA. Marked disturbance of calcium homeostasis in mice with targeted disruption of the Trpv6 calcium channel gene. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2007;22(2):274-285.
  206. Sampson HW, Matthews JL, Martin JH, Kunin AS. An electron microscopic localization of calcium in the small intestine of normal, rachitic, and vitamin-D-treated rats. Calcif Tissue Res. 1970;5(4):305-316.
  207. Schaefer HJ. Ultrastructure and ion distribution of the intestinal cell during experimental vitamin D deficiency rickets in rats. Virchows Archiv. 1973;359:111-123.
  208. Chandra S, Fullmer CS, Smith CA, Wasserman RH, Morrison GH. Ion microscopic imaging of calcium transport in the intestinal tissue of vitamin D-deficient and vitamin D-replete chickens: a 44Ca stable isotope study. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(15):5715-5719.
  209. Bikle DD, Zolock DT, Morrissey RL, Herman RH. Independence of 1,25-dihydroxyvitamin D3-mediated calcium transport from de novo RNA and protein synthesis. The Journal of biological chemistry. 1978;253(2):484-488.
  210. Bikle DD, Morrissey RL, Zolock DT. The mechanism of action of vitamin D in the intestine. Am J Clin Nutr. 1979;32(11):2322-2328.
  211. Glenney JR, Jr., Glenney P. Comparison of Ca++-regulated events in the intestinal brush border. J Cell Biol. 1985;100(3):754-763.
  212. Bikle DD, Gee E. Free, and not total, 1,25-dihydroxyvitamin D regulates 25- hydroxyvitamin D metabolism by keratinocytes. Endocrinology. 1989;124(2):649-654.
  213. Bikle DD, Munson S, Chafouleas J. Calmodulin may mediate 1,25-dihydroxyvitamin D-stimulated intestinal calcium transport. FEBS Lett. 1984;174(1):30-33.
  214. Howe CL, Keller TC, 3rd, Mooseker MS, Wasserman RH. Analysis of cytoskeletal proteins and Ca2+-dependent regulation of structure in intestinal brush borders from rachitic chicks. Proc Natl Acad Sci U S A. 1982;79(4):1134-1138.
  215. Bikle DD, Munson S. The villus gradient of brush border membrane calmodulin and the calcium-independent calmodulin-binding protein parallels that of calcium-accumulating ability. Endocrinology. 1986;118(2):727-732.
  216. Drenckhahn D, Dermietzel R. Organization of the actin filament cytoskeleton in the intestinal brush border: a quantitative and qualitative immunoelectron microscope study. J Cell Biol. 1988;107(3):1037-1048.
  217. Munson S, Wang Y, Chang W, Bikle DD. Myosin 1a Regulates Osteoblast Differentiation Independent of Intestinal Calcium Transport. J Endocr Soc. 2019;3(11):1993-2011.
  218. Wasserman RH, Taylor AN. Vitamin D-dependent calcium-binding protein. Response to some physiological and nutritional variables. The Journal of biological chemistry. 1968;243(14):3987-3993.
  219. Lee GS, Lee KY, Choi KC, Ryu YH, Paik SG, Oh GT, Jeung EB. Phenotype of a calbindin-D9k gene knockout is compensated for by the induction of other calcium transporter genes in a mouse model. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2007;22(12):1968-1978.
  220. Ghijsen WE, De Jong MD, Van Os CH. ATP-dependent calcium transport and its correlation with Ca2+ -ATPase activity in basolateral plasma membranes of rat duodenum. Biochim Biophys Acta. 1982;689(2):327-336.
  221. Cai Q, Chandler JS, Wasserman RH, Kumar R, Penniston JT. Vitamin D and adaptation to dietary calcium and phosphate deficiencies increase intestinal plasma membrane calcium pump gene expression. Proc Natl Acad Sci U S A. 1993;90(4):1345-1349.
  222. Wasserman RH, Chandler JS, Meyer SA, Smith CA, Brindak ME, Fullmer CS, Penniston JT, Kumar R. Intestinal calcium transport and calcium extrusion processes at the basolateral membrane. J Nutr. 1992;122(3 Suppl):662-671.
  223. Ryan ZC, Craig TA, Filoteo AG, Westendorf JJ, Cartwright EJ, Neyses L, Strehler EE, Kumar R. Deletion of the intestinal plasma membrane calcium pump, isoform 1, Atp2b1, in mice is associated with decreased bone mineral density and impaired responsiveness to 1, 25-dihydroxyvitamin D3. Biochem Biophys Res Commun. 2015;467(1):152-156.
  224. Liu C, Weng H, Chen L, Yang S, Wang H, Debnath G, Guo X, Wu L, Mohandas N, An X. Impaired intestinal calcium absorption in protein 4.1R-deficient mice due to altered expression of plasma membrane calcium ATPase 1b (PMCA1b). The Journal of biological chemistry. 2013;288(16):11407-11415.
  225. Hoenderop JG, Nilius B, Bindels RJ. Calcium absorption across epithelia. Physiol Rev. 2005;85(1):373-422.
  226. Charoenphandhu N, Krishnamra N. Prolactin is an important regulator of intestinal calcium transport. Can J Physiol Pharmacol. 2007;85(6):569-581.
  227. Turner JR, Rill BK, Carlson SL, Carnes D, Kerner R, Mrsny RJ, Madara JL. Physiological regulation of epithelial tight junctions is associated with myosin light-chain phosphorylation. Am J Physiol. 1997;273(4):C1378-1385.
  228. Fujita H, Chiba H, Yokozaki H, Sakai N, Sugimoto K, Wada T, Kojima T, Yamashita T, Sawada N. Differential expression and subcellular localization of claudin-7, -8, -12, -13, and -15 along the mouse intestine. J Histochem Cytochem. 2006;54(8):933-944.
  229. Van Itallie CM, Fanning AS, Anderson JM. Reversal of charge selectivity in cation or anion-selective epithelial lines by expression of different claudins. Am J Physiol Renal Physiol. 2003;285(6):F1078-1084.
  230. Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, Uchiyama Y, Yamamoto Y, Wada T, Kojima T, Yokozaki H, Yamashita T, Kato S, Sawada N, Chiba H. Tight junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2+ absorption between enterocytes. Mol Biol Cell. 2008;19(5):1912-1921.
  231. Charoenphandhu N, Nakkrasae LI, Kraidith K, Teerapornpuntakit J, Thongchote K, Thongon N, Krishnamra N. Two-step stimulation of intestinal Ca(2+) absorption during lactation by long-term prolactin exposure and suckling-induced prolactin surge. Am J Physiol Endocrinol Metab. 2009;297(3):E609-619.
  232. Harrison HE, Harrison HC. Intestinal transport of phosphate: Action of vitamin D, calcium, and potassium. American Journal of Physiology. 1961;201:1007-1012.
  233. Peterlik M, Wasserman RH. Regulation by vitamin D of intestinal phosphate absorption. Horm Metab Res. 1980;12(5):216-219.
  234. Xu H, Bai L, Collins JF, Ghishan FK. Molecular cloning, functional characterization, tissue distribution, and chromosomal localization of a human, small intestinal sodium-phosphate (Na+-Pi) transporter (SLC34A2). Genomics. 1999;62(2):281-284.
  235. Xu H, Bai L, Collins JF, Ghishan FK. Age-dependent regulation of rat intestinal type IIb sodium-phosphate cotransporter by 1,25-(OH)(2) vitamin D(3). American journal of physiology Cell physiology. 2002;282(3):C487-493.
  236. Fuchs R, Peterlik M. Vitamin D-induced transepithelial phosphate and calcium transport by chick jejunum. Effect of microfilamentous and microtubular inhibitors. FEBS Lett. 1979;100(2):357-359.
  237. Narbaitz R, Stumpf WE, Sar M, Huang S, DeLuca HF. Autoradiographic localization of target cells for 1 alpha, 25- dihydroxyvitamin D3 in bones from fetal rats. Calcif Tissue Int. 1983;35(2):177-182.
  238. Boivin G, Mesguich P, Pike JW, Bouillon R, Meunier PJ, Haussler MR, Dubois PM, Morel G. Ultrastructural immunocytochemical localization of endogenous 1,25- dihydroxyvitamin D3 and its receptors in osteoblasts and osteocytes from neonatal mouse and rat calvaria. Bone Miner. 1987;3(2):125-136.
  239. Nakamichi Y, Udagawa N, Horibe K, Mizoguchi T, Yamamoto Y, Nakamura T, Hosoya A, Kato S, Suda T, Takahashi N. VDR in Osteoblast-Lineage Cells Primarily Mediates Vitamin D Treatment-Induced Increase in Bone Mass by Suppressing Bone Resorption. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2017;32(6):1297-1308.
  240. Panda DK, Miao D, Bolivar I, Li J, Huo R, Hendy GN, Goltzman D. Inactivation of the 25-hydroxyvitamin D 1alpha-hydroxylase and vitamin D receptor demonstrates independent and interdependent effects of calcium and vitamin D on skeletal and mineral homeostasis. The Journal of biological chemistry. 2004;279(16):16754-16766.
  241. Chenu C, Valentin-Opran A, Chavassieux P, Saez S, Meunier PJ, Delmas PD. Insulin like growth factor I hormonal regulation by growth hormone and by 1,25(OH)2D3 and activity on human osteoblast-like cells in short- term cultures. Bone. 1990;11(2):81-86.
  242. Kurose H, Yamaoka K, Okada S, Nakajima S, Seino Y. 1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] increases insulin-like growth factor I (IGF-I) receptors in clonal osteoblastic cells. Study on interaction of IGF-I and 1,25-(OH)2D3. Endocrinology. 1990;126(4):2088-2094.
  243. Scharla SH, Strong DD, Mohan S, Baylink DJ, Linkhart TA. 1,25-Dihydroxyvitamin D3 differentially regulates the production of insulin-like growth factor I (IGF-I) and IGF-binding protein-4 in mouse osteoblasts. Endocrinology. 1991;129(6):3139-3146.
  244. Moriwake T, Tanaka H, Kanzaki S, Higuchi J, Seino Y. 1,25-Dihydroxyvitamin D3 stimulates the secretion of insulin-like growth factor binding protein 3 (IGFBP-3) by cultured human osteosarcoma cells. Endocrinology. 1992;130(2):1071-1073.
  245. Sato T, Ono T, Tuan RS. 1,25-Dihydroxy vitamin D3 stimulation of TGF-beta expression in chick embryonic calvarial bone. Differentiation. 1993;52(2):139-150.
  246. Wang DS, Yamazaki K, Nohtomi K, Shizume K, Ohsumi K, Shibuya M, Demura H, Sato K. Increase of vascular endothelial growth factor mRNA expression by 1,25- dihydroxyvitamin D3 in human osteoblast-like cells. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1996;11(4):472-479.
  247. Lacey DL, Grosso LE, Moser SA, Erdmann J, Tan HL, Pacifici R, Villareal DT. IL-1-induced murine osteoblast IL-6 production is mediated by the type 1 IL-1 receptor and is increased by 1,25 dihydroxyvitamin D3. The Journal of clinical investigation. 1993;91(4):1731-1742.
  248. Lacey DL, Erdmann JM, Tan HL, Ohara J. Murine osteoblast interleukin 4 receptor expression: upregulation by 1,25 dihydroxyvitamin D3. Journal of cellular biochemistry. 1993;53(2):122-134.
  249. Nambi P, Wu HL, Lipshutz D, Prabhakar U. Identification and characterization of endothelin receptors on rat osteoblastic osteosarcoma cells: down-regulation by 1,25-dihydroxy- vitamin D3. Mol Pharmacol. 1995;47(2):266-271.
  250. Zhou X, von der Mark K, Henry S, Norton W, Adams H, de Crombrugghe B. Chondrocytes transdifferentiate into osteoblasts in endochondral bone during development, postnatal growth and fracture healing in mice. PLoS Genet. 2014;10(12):e1004820.
  251. Johnson JA, Grande JP, Roche PC, Kumar R. Ontogeny of the 1,25-dihydroxyvitamin D3 receptor in fetal rat bone. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1996;11(1):56-61.
  252. Miller SC, Halloran BP, DeLuca HF, Lee WSS. Studies on the role of vitamin D in early skeletal development, mineralization, and growth in rats. Calcif Tissue Int. 1983;35:455-460.
  253. Kyeyune-Nyombi E, Lau KH, Baylink DJ, Strong DD. 1,25-Dihydroxyvitamin D3 stimulates both alkaline phosphatase gene transcription and mRNA stability in human bone cells. Arch Biochem Biophys. 1991;291(2):316-325.
  254. Irving JT, Wuthier RE. Histochemistry and biochemistry of calcification with special reference to the role of lipids. Clin Orthop. 1968;56:237-260.
  255. Howell DS, Marquez JF, Pita JC. The nature of phospholipids in normal and rachitic costochondral plates. Arthritis Rheum. 1965;8(6):1039-1046.
  256. Dean DD, Boyan BD, Muniz OE, Howell DS, Schwartz Z. Vitamin D metabolites regulate matrix vesicle metalloproteinase content in a cell maturation-dependent manner. Calcif Tissue Int. 1996;59(2):109-116.
  257. Roughley PJ, Dickson IR. A comparison of proteoglycan from chick cartilage of different types and a study of the effect of vitamin D on proteoglycan structure. Connect Tissue Res. 1986;14(3):187-197.
  258. Plachot JJ, Du Bois MB, Halpern S, Cournot-Witmer G, Garabedian M, Balsan S. In vitro action of 1,25-dihydroxycholecalciferol and 24,25- dihydroxycholecalciferol on matrix organization and mineral distribution in rabbit growth plate. Metab Bone Dis Relat Res. 1982;4(2):135-142.
  259. Boyan BD, Schwartz Z, Carnes DL, Jr., Ramirez V. The effects of vitamin D metabolites on the plasma and matrix vesicle membranes of growth and resting cartilage cells in vitro. Endocrinology. 1988;122(6):2851-2860.
  260. Schwartz Z, Boyan B. The effects of vitamin D metabolites on phospholipase A2 activity of growth zone and resting zone cartilage cells in vitro. Endocrinology. 1988;122(5):2191-2198.
  261. Swain LD, Schwartz Z, Caulfield K, Brooks BP, Boyan BD. Nongenomic regulation of chondrocyte membrane fluidity by 1,25-(OH)2D3 and 24,25-(OH)2D3 is dependent on cell maturation. Bone. 1993;14(4):609-617.
  262. Sylvia VL, Schwartz Z, Schuman L, Morgan RT, Mackey S, Gomez R, Boyan BD. Maturation-dependent regulation of protein kinase C activity by vitamin D3 metabolites in chondrocyte cultures. J Cell Physiol. 1993;157(2):271-278.
  263. Boyan BD, Chen J, Schwartz Z. Mechanism of Pdia3-dependent 1alpha,25-dihydroxy vitamin D3 signaling in musculoskeletal cells. Steroids. 2012;77(10):892-896.
  264. Owen TA, Aronow MS, Barone LM, Bettencourt B, Stein GS, Lian JB. Pleiotropic effects of vitamin D on osteoblast gene expression are related to the proliferative and differentiated state of the bone cell phenotype: dependency upon basal levels of gene expression, duration of exposure, and bone matrix competency in normal rat osteoblast cultures. Endocrinology. 1991;128(3):1496-1504.
  265. Lian J, Stewart C, Puchacz E, Mackowiak S, Shalhoub V, Collart D, Zambetti G, Stein G. Structure of the rat osteocalcin gene and regulation of vitamin D- dependent expression. Proc Natl Acad Sci U S A. 1989;86(4):1143-1147.
  266. Manolagas SC, Burton DW, Deftos LJ. 1,25-Dihydroxyvitamin D3 stimulates the alkaline phosphatase activity of osteoblast-like cells. The Journal of biological chemistry. 1981;256(14):7115-7117.
  267. Rowe DW, Kream BE. Regulation of collagen synthesis in fetal rat calvaria by 1,25- dihydroxyvitamin D3. The Journal of biological chemistry. 1982;257(14):8009-8015.
  268. Prince CW, Butler WT. 1,25-Dihydroxyvitamin D3 regulates the biosynthesis of osteopontin, a bone-derived cell attachment protein, in clonal osteoblast-like osteosarcoma cells. Coll Relat Res. 1987;7(4):305-313.
  269. Demay MB, Gerardi JM, DeLuca HF, Kronenberg HM. DNA sequences in the rat osteocalcin gene that bind the 1,25- dihydroxyvitamin D3 receptor and confer responsiveness to 1,25- dihydroxyvitamin D3. Proc Natl Acad Sci U S A. 1990;87(1):369-373.
  270. Kerner SA, Scott RA, Pike JW. Sequence elements in the human osteocalcin gene confer basal activation and inducible response to hormonal vitamin D3. Proceedings of the National Academy of Sciences of the United States of America. 1989;86(12):4455-4459.
  271. Noda M, Vogel RL, Craig AM, Prahl J, DeLuca HF, Denhardt DT. Identification of a DNA sequence responsible for binding of the 1,25- dihydroxyvitamin D3 receptor and 1,25-dihydroxyvitamin D3 enhancement of mouse secreted phosphoprotein 1 (SPP-1 or osteopontin) gene expression. Proc Natl Acad Sci U S A. 1990;87(24):9995-9999.
  272. Zhang R, Ducy P, Karsenty G. 1,25-dihydroxyvitamin D3 inhibits Osteocalcin expression in mouse through an indirect mechanism. The Journal of biological chemistry. 1997;272(1):110-116.
  273. Wronski TJ, Halloran BP, Bikle DD, Globus RK, Morey-Holton ER. Chronic administration of 1,25-dihydroxyvitamin D3: increased bone but impaired mineralization. Endocrinology. 1986;119(6):2580-2585.
  274. Hock JM, Gunness-Hey M, Poser J, Olson H, Bell NH, Raisz LG. Stimulation of undermineralized matrix formation by 1,25 dihydroxyvitamin D3 in long bones of rats. Calcif Tissue Int. 1986;38(2):79-86.
  275. Suda T, Takahashi N, Abe E. Role of vitamin D in bone resorption. Journal of cellular biochemistry. 1992;49(1):53-58.
  276. Merke J, Klaus G, Hugel U, Waldherr R, Ritz E. No 1,25-dihydroxyvitamin D3 receptors on osteoclasts of calcium- deficient chicken despite demonstrable receptors on circulating monocytes. The Journal of clinical investigation. 1986;77(1):312-314.
  277. Mee AP, Hoyland JA, Braidman IP, Freemont AJ, Davies M, Mawer EB. Demonstration of vitamin D receptor transcripts in actively resorbing osteoclasts in bone sections. Bone. 1996;18(4):295-299.
  278. Rodan GA, Martin TJ. Role of osteoblasts in hormonal control of bone resorption--a hypothesis. Calcif Tissue Int. 1981;33(4):349-351.
  279. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin TJ. Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev. 1999;20(3):345-357.
  280. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, Mochizuki S, Tomoyasu A, Yano K, Goto M, Murakami A, Tsuda E, Morinaga T, Higashio K, Udagawa N, Takahashi N, Suda T. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci U S A. 1998;95(7):3597-3602.
  281. Takeda S, Yoshizawa T, Nagai Y, Yamato H, Fukumoto S, Sekine K, Kato S, Matsumoto T, Fujita T. Stimulation of osteoclast formation by 1,25-dihydroxyvitamin D requires its binding to vitamin D receptor (VDR) in osteoblastic cells: studies using VDR knockout mice. Endocrinology. 1999;140(2):1005-1008.
  282. Friedman PA, Gesek FA. Cellular calcium transport in renal epithelia: measurement, mechanisms, and regulation. Physiol Rev. 1995;75(3):429-471.
  283. Winaver J, Sylk DB, Robertson JS, Chen TC, Puschett JB. Micropuncture study of the acute renal tubular transport effects of 25-hydroxyvitamin D3 in the dog. Mineral and electrolyte metabolism. 1980;4:178-188.
  284. Tenenhouse HS. Cellular and molecular mechanisms of renal phosphate transport. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1997;12(2):159-164.
  285. Puschett JB, Beck WS, Jr., Jelonek A, Fernandez PC. Study of the renal tubular interactions of thyrocalcitonin, cyclic adenosine 3',5'-monophosphate, 25-hydroxycholecalciferol, and calcium ion. The Journal of clinical investigation. 1974;53(3):756-767.
  286. Puschett JB, Fernandez PC, Boyle IT, Gray RW, Omdahl JL, DeLuca HF. The acute renal tubular effects of 1,25-dihydroxycholecalciferol. Proc Soc Exp Biol Med. 1972;141(1):379-384.
  287. Puschett JB, Moranz J, Kurnick WS. Evidence for a direct action of cholecalciferol and 25- hydroxycholecalciferol on the renal transport of phosphate, sodium, and calcium. The Journal of clinical investigation. 1972;51(2):373-385.
  288. Popovtzer MM, Robinette JB, DeLuca HF, Holick MF. The acute effect of 25-hydroxycholecalciferol on renal handling of phosphorus. Evidence for a parathyroid hormone-dependent mechanism. The Journal of clinical investigation. 1974;53(3):913-921.
  289. Yamamoto M, Kawanobe Y, Takahashi H, Shimazawa E, Kimura S, Ogata E. Vitamin D deficiency and renal calcium transport in the rat. The Journal of clinical investigation. 1984;74(2):507-513.
  290. Kumar R, Schaefer J, Grande JP, Roche PC. Immunolocalization of calcitriol receptor, 24-hydroxylase cytochrome P- 450, and calbindin D28k in human kidney. Am J Physiol. 1994;266(3 Pt 2):F477-485.
  291. Borke JL, Minami J, Verma AK, Penniston JT, Kumar R. Co-localization of erythrocyte Ca++-Mg++ ATPase and vitamin D-dependent 28-kDa-calcium binding protein. Kidney Int. 1988;34(2):262-267.
  292. Christakos S, Brunette MG, Norman AW. Localization of immunoreactive vitamin D-dependent calcium binding protein in chick nephron. Endocrinology. 1981;109(1):322-324.
  293. Roth J, Thorens B, Hunziker W, Norman AW, Orci L. Vitamin D--dependent calcium binding protein: immunocytochemical localization in chick kidney. Science. 1981;214(4517):197-200.
  294. Bouhtiauy I, Lajeunesse D, Christakos S, Brunette MG. Two vitamin D3-dependent calcium binding proteins increase calcium reabsorption by different mechanisms. I. Effect of CaBP 28K. Kidney Int. 1994;45(2):461-468.
  295. Biber J, Hernando N, Forster I. Phosphate transporters and their function. Annu Rev Physiol. 2013;75:535-550.
  296. Silver J, Russell J, Sherwood LM. Regulation by vitamin D metabolites of messenger ribonucleic acid for preproparathyroid hormone in isolated bovine parathyroid cells. Proc Natl Acad Sci U S A. 1985;82(12):4270-4273.
  297. Cantley LK, Russell J, Lettieri D, Sherwood LM. 1,25-Dihydroxyvitamin D3 suppresses parathyroid hormone secretion from bovine parathyroid cells in tissue culture. Endocrinology. 1985;117(5):2114-2119.
  298. Demay MB, Kiernan MS, DeLuca HF, Kronenberg HM. Sequences in the human parathyroid hormone gene that bind the 1,25- dihydroxyvitamin D3 receptor and mediate transcriptional repression in response to 1,25-dihydroxyvitamin D3. Proc Natl Acad Sci U S A. 1992;89(17):8097-8101.
  299. Russell J, Ashok S, Koszewski NJ. Vitamin D receptor interactions with the rat parathyroid hormone gene: synergistic effects between two negative vitamin D response elements. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1999;14(11):1828-1837.
  300. Nishishita T, Okazaki T, Ishikawa T, Igarashi T, Hata K, Ogata E, Fujita T. A negative vitamin D response DNA element in the human parathyroid hormone-related peptide gene binds to vitamin D receptor along with Ku antigen to mediate negative gene regulation by vitamin D. The Journal of biological chemistry. 1998;273(18):10901-10907.
  301. Hawa NS, O'Riordan JL, Farrow SM. Functional analysis of vitamin D response elements in the parathyroid hormone gene and a comparison with the osteocalcin gene. Biochem Biophys Res Commun. 1996;228(2):352-357.
  302. Mackey SL, Heymont JL, Kronenberg HM, Demay MB. Vitamin D receptor binding to the negative human parathyroid hormone vitamin D response element does not require the retinoid x receptor. Mol Endocrinol. 1996;10(3):298-305.
  303. Canaff L, Hendy GN. Human calcium-sensing receptor gene. Vitamin D response elements in promoters P1 and P2 confer transcriptional responsiveness to 1,25-dihydroxyvitamin D. The Journal of biological chemistry. 2002;277(33):30337-30350.
  304. Naveh-Many T, Marx R, Keshet E, Pike JW, Silver J. Regulation of 1,25-dihydroxyvitamin D3 receptor gene expression by 1,25- dihydroxyvitamin D3 in the parathyroid in vivo. The Journal of clinical investigation. 1990;86(6):1968-1975.
  305. Russell J, Bar A, Sherwood LM, Hurwitz S. Interaction between calcium and 1,25-dihydroxyvitamin D3 in the regulation of preproparathyroid hormone and vitamin D receptor messenger ribonucleic acid in avian parathyroids. Endocrinology. 1993;132(6):2639-2644.
  306. Dedhar S, Rennie PS, Shago M, Hagesteijn CY, Yang H, Filmus J, Hawley RG, Bruchovsky N, Cheng H, Matusik RJ, et al. Inhibition of nuclear hormone receptor activity by calreticulin. Nature. 1994;367(6462):480-483.
  307. Wheeler DG, Horsford J, Michalak M, White JH, Hendy GN. Calreticulin inhibits vitamin D3 signal transduction. Nucleic Acids Res. 1995;23(16):3268-3274.
  308. Sela A, Silver J, Naveh-Many T. Chronic hypocalcemia increases PTH mRNA despite high 1,25(OH)2D levels:Roles of calretivulin and the vitamin D receptor. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 1996;11: (abstract).
  309. Ritter CS, Haughey BH, Armbrecht HJ, Brown AJ. Distribution and regulation of the 25-hydroxyvitamin D3 1alpha-hydroxylase in human parathyroid glands. J Steroid Biochem Mol Biol. 2012;130(1-2):73-80.
  310. Kadowaki S, Norman AW. Demonstration that the vitamin D metabolite 1,25(OH)2-vitamin D3 and not 24R,25(OH)2-vitamin D3 is essential for normal insulin secretion in the perfused rat pancreas. Diabetes. 1985;34(4):315-320.
  311. Lee S, Clark SA, Gill RK, Christakos S. 1,25-Dihydroxyvitamin D3 and pancreatic beta-cell function: vitamin D receptors, gene expression, and insulin secretion. Endocrinology. 1994;134(4):1602-1610.
  312. Clark SA, Stumpf WE, Sar M, DeLuca HF, Tanaka Y. Target cells for 1,25 dihydroxyvitamin D3 in the pancreas. Cell and tissue research. 1980;209(3):515-520.
  313. Morrissey RL, Bucci TJ, Richard B, Empson N, Lufkin EG. Calcium-binding protein: its cellular localization in jejunum, kidney and pancreas. Proc Soc Exp Biol Med. 1975;149(1):56-60.
  314. Bland R, Markovic D, Hills CE, Hughes SV, Chan SL, Squires PE, Hewison M. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in pancreatic islets. The Journal of steroid biochemistry and molecular biology. 2004;89-90(1-5):121-125.
  315. Sooy K, Schermerhorn T, Noda M, Surana M, Rhoten WB, Meyer M, Fleischer N, Sharp GW, Christakos S. Calbindin-D(28k) controls [Ca(2+)](i) and insulin release. Evidence obtained from calbindin-d(28k) knockout mice and beta cell lines. The Journal of biological chemistry. 1999;274(48):34343-34349.
  316. Rabinovitch A, Suarez-Pinzon WL, Sooy K, Strynadka K, Christakos S. Expression of calbindin-D(28k) in a pancreatic islet beta-cell line protects against cytokine-induced apoptosis and necrosis. Endocrinology. 2001;142(8):3649-3655.
  317. Pittas AG, Lau J, Hu FB, Dawson-Hughes B. The role of vitamin D and calcium in type 2 diabetes. A systematic review and meta-analysis. The Journal of clinical endocrinology and metabolism. 2007;92(6):2017-2029.
  318. Pittas AG, Jorde R, Kawahara T, Dawson-Hughes B. Vitamin D Supplementation for Prevention of Type 2 Diabetes Mellitus: To D or Not to D? The Journal of clinical endocrinology and metabolism. 2020;105(12).
  319. Pittas A, Dawson-Hughes B, Staten M. Vitamin D Supplementation and Prevention of Type 2 Diabetes. Reply. N Engl J Med. 2019;381(18):1785-1786.
  320. Harris SS, Pittas AG, Palermo NJ. A randomized, placebo-controlled trial of vitamin D supplementation to improve glycaemia in overweight and obese African Americans. Diabetes Obes Metab. 2012;14(9):789-794.
  321. Kolek OI, Hines ER, Jones MD, LeSueur LK, Lipko MA, Kiela PR, Collins JF, Haussler MR, Ghishan FK. 1alpha,25-Dihydroxyvitamin D3 upregulates FGF23 gene expression in bone: the final link in a renal-gastrointestinal-skeletal axis that controls phosphate transport. American journal of physiology Gastrointestinal and liver physiology. 2005;289(6):G1036-1042.
  322. Fukumoto S, Yamashita T. FGF23 is a hormone-regulating phosphate metabolism--unique biological characteristics of FGF23. Bone. 2007;40(5):1190-1195.
  323. Eisman JA, Martin TJ, MacIntyre I, Moseley JM. 1,25-dihydroxyvitamin-D-receptor in breast cancer cells. Lancet. 1979;2(8156-8157):1335-1336.
  324. Fleet JC, DeSmet M, Johnson R, Li Y. Vitamin D and cancer: a review of molecular mechanisms. Biochem J. 2012;441(1):61-76.
  325. Ingraham BA, Bragdon B, Nohe A. Molecular basis of the potential of vitamin D to prevent cancer. Current medical research and opinion. 2008;24(1):139-149.
  326. Shah S, Hecht A, Pestell R, Byers SW. Trans-repression of beta-catenin activity by nuclear receptors. The Journal of biological chemistry. 2003;278(48):48137-48145.
  327. Shah S, Islam MN, Dakshanamurthy S, Rizvi I, Rao M, Herrell R, Zinser G, Valrance M, Aranda A, Moras D, Norman A, Welsh J, Byers SW. The molecular basis of vitamin D receptor and beta-catenin crossregulation. Molecular cell. 2006;21(6):799-809.
  328. Dixon KM, Deo SS, Wong G, Slater M, Norman AW, Bishop JE, Posner GH, Ishizuka S, Halliday GM, Reeve VE, Mason RS. Skin cancer prevention: a possible role of 1,25dihydroxyvitamin D3 and its analogs. The Journal of steroid biochemistry and molecular biology. 2005;97(1-2):137-143.
  329. Demetriou SK, Ona-Vu K, Teichert AE, Cleaver JE, Bikle DD, Oh DH. Vitamin D receptor mediates DNA repair and is UV inducible in intact epidermis but not in cultured keratinocytes. The Journal of investigative dermatology. 2012;132(8):2097-2100.
  330. De Haes P, Garmyn M, Degreef H, Vantieghem K, Bouillon R, Segaert S. 1,25-Dihydroxyvitamin D3 inhibits ultraviolet B-induced apoptosis, Jun kinase activation, and interleukin-6 production in primary human keratinocytes. J Cell Biochem. 2003;89(4):663-673.
  331. Gupta R, Dixon KM, Deo SS, Holliday CJ, Slater M, Halliday GM, Reeve VE, Mason RS. Photoprotection by 1,25 dihydroxyvitamin D3 is associated with an increase in p53 and a decrease in nitric oxide products. The Journal of investigative dermatology. 2007;127(3):707-715.
  332. Garland C, Shekelle RB, Barrett-Connor E, Criqui MH, Rossof AH, Paul O. Dietary vitamin D and calcium and risk of colorectal cancer: a 19-year prospective study in men. Lancet. 1985;1(8424):307-309.
  333. Bostick RM, Potter JD, Sellers TA, McKenzie DR, Kushi LH, Folsom AR. Relation of calcium, vitamin D, and dairy food intake to incidence of colon cancer among older women. The Iowa Women's Health Study. Am J Epidemiol. 1993;137(12):1302-1317.
  334. Kearney J, Giovannucci E, Rimm EB, Ascherio A, Stampfer MJ, Colditz GA, Wing A, Kampman E, Willett WC. Calcium, vitamin D, and dairy foods and the occurrence of colon cancer in men. Am J Epidemiol. 1996;143(9):907-917.
  335. Garland FC, Garland CF, Gorham ED, Young JF. Geographic variation in breast cancer mortality in the United States: a hypothesis involving exposure to solar radiation. Prev Med. 1990;19(6):614-622.
  336. Hanchette CL, Schwartz GG. Geographic patterns of prostate cancer mortality. Evidence for a protective effect of ultraviolet radiation. Cancer. 1992;70(12):2861-2869.
  337. Boscoe FP, Schymura MJ. Solar ultraviolet-B exposure and cancer incidence and mortality in the United States, 1993-2002. BMC Cancer. 2006;6:264.
  338. Diercke K, Kohl A, Lux CJ, Erber R. Strain-dependent up-regulation of ephrin-B2 protein in periodontal ligament fibroblasts contributes to osteogenesis during tooth movement. The Journal of biological chemistry. 2011;286(43):37651-37664.
  339. Lappe JM, Travers-Gustafson D, Davies KM, Recker RR, Heaney RP. Vitamin D and calcium supplementation reduces cancer risk: results of a randomized trial. The American journal of clinical nutrition. 2007;85(6):1586-1591.
  340. Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, Gibson H, Gordon D, Copeland T, D'Agostino D, Friedenberg G, Ridge C, Bubes V, Giovannucci EL, Willett WC, Buring JE, Group VR. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. N Engl J Med. 2019;380(1):33-44.
  341. Gross C, Stamey T, Hancock S, Feldman D. Treatment of early recurrent prostate cancer with 1,25-dihydroxyvitamin D3 (calcitriol). J Urol. 1998;159(6):2035-2039; discussion 2039-2040.
  342. Beer TM, Ryan CW, Venner PM, Petrylak DP, Chatta GS, Ruether JD, Redfern CH, Fehrenbacher L, Saleh MN, Waterhouse DM, Carducci MA, Vicario D, Dreicer R, Higano CS, Ahmann FR, Chi KN, Henner WD, Arroyo A, Clow FW. Double-blinded randomized study of high-dose calcitriol plus docetaxel compared with placebo plus docetaxel in androgen-independent prostate cancer: a report from the ASCENT Investigators. J Clin Oncol. 2007;25(6):669-674.
  343. Barnett CM, Beer TM. Prostate cancer and vitamin D: what does the evidence really suggest? Urol Clin North Am. 2011;38(3):333-342.
  344. Lehmann B, Tiebel O, Meurer M. Expression of vitamin D3 25-hydroxylase (CYP27) mRNA after induction by vitamin D3 or UVB radiation in keratinocytes of human skin equivalents-- a preliminary study. Arch Dermatol Res. 1999;291(9):507-510.
  345. Bikle DD, Nemanic MK, Gee E, Elias P. 1,25-Dihydroxyvitamin D3 production by human keratinocytes. Kinetics and regulation. The Journal of clinical investigation. 1986;78(2):557-566.
  346. Bikle DD, Pillai S, Gee E, Hincenbergs M. Tumor necrosis factor-alpha regulation of 1,25-dihydroxyvitamin D production by human keratinocytes. Endocrinology. 1991;129(1):33-38.
  347. Bikle DD, Pillai S, Gee E, Hincenbergs M. Regulation of 1,25-dihydroxyvitamin D production in human keratinocytes by interferon-gamma. Endocrinology. 1989;124(2):655-660.
  348. Pillai S, Bikle DD, Elias PM. 1,25-Dihydroxyvitamin D production and receptor binding in human keratinocytes varies with differentiation. The Journal of biological chemistry. 1988;263(11):5390-5395.
  349. Bikle DD. Vitamin D and the skin: Physiology and pathophysiology. Reviews in endocrine & metabolic disorders. 2012;13(1):3-19.
  350. Hennings H, Michael D, Cheng C, Steinert P, Holbrook K, Yuspa SH. Calcium regulation of growth and differentiation of mouse epidermal cells in culture. Cell. 1980;19(1):245-254.
  351. Hennings H, Holbrook KA. Calcium regulation of cell-cell contact and differentiation of epidermal cells in culture. An ultrastructural study. Experimental cell research. 1983;143(1):127-142.
  352. Hennings H, Holbrook KA, Yuspa SH. Factors influencing calcium-induced terminal differentiation in cultured mouse epidermal cells. Journal of cellular physiology. 1983;116(3):265-281.
  353. Praeger FC, Stanulis-Praeger BM, Gilchrest BA. Use of strontium to separate calcium-dependent pathways for proliferation and differentiation in human keratinocytes. J Cell Physiol. 1987;132(1):81-89.
  354. Pillai S, Bikle DD, Mancianti ML, Cline P, Hincenbergs M. Calcium regulation of growth and differentiation of normal human keratinocytes: modulation of differentiation competence by stages of growth and extracellular calcium. Journal of Cellular Physiology. 1990;143(2):294-302.
  355. Yuspa SH, Kilkenny AE, Steinert PM, Roop DR. Expression of murine epidermal differentiation markers is tightly regulated by restricted extracellular calcium concentrations in vitro. The Journal of cell biology. 1989;109(3):1207-1217.
  356. Bikle DD, Xie Z, Tu CL. Calcium regulation of keratinocyte differentiation. Expert review of endocrinology & metabolism. 2012;7(4):461-472.
  357. Sheu HM, Kitajima Y, Yaoita H. Involvement of protein kinase C in translocation of desmoplakins from cytosol to plasma membrane during desmosome formation in human squamous cell carcinoma cells grown in low to normal calcium concentration. Exp Cell Res. 1989;185(1):176-190.
  358. Denning MF, Dlugosz AA, Williams EK, Szallasi Z, Blumberg PM, Yuspa SH. Specific protein kinase C isozymes mediate the induction of keratinocyte differentiation markers by calcium. Cell Growth Differ. 1995;6(2):149-157.
  359. Filvaroff E, Calautti E, McCormick F, Dotto GP. Specific changes of Ras GTPase-activating protein (GAP) and a GAP- associated p62 protein during calcium-induced keratinocyte differentiation. Mol Cell Biol. 1992;12(12):5319-5328.
  360. Filvaroff E, Calautti E, Reiss M, Dotto GP. Functional evidence for an extracellular calcium receptor mechanism triggering tyrosine kinase activation associated with mouse keratinocyte differentiation. The Journal of biological chemistry. 1994;269(34):21735-21740.
  361. Rice RH, Green H. Presence in human epidermal cells of a soluble protein precursor of the cross-linked envelope: activation of the cross-linking by calcium ions. Cell. 1979;18(3):681-694.
  362. Hohl D, Lichti U, Breitkreutz D, Steinert PM, Roop DR. Transcription of the human loricrin gene in vitro is induced by calcium and cell density and suppressed by retinoic acid. The Journal of investigative dermatology. 1991;96(4):414-418.
  363. Simon M, Green H. Participation of membrane-associated proteins in the formation of the cross-linked envelope of the keratinocyte. Cell. 1984;36(4):827-834.
  364. Hohl D. Cornified cell envelope. Dermatologica. 1990;180(4):201-211.
  365. Thacher SM, Rice RH. Keratinocyte-specific transglutaminase of cultured human epidermal cells: relation to cross-linked envelope formation and terminal differentiation. Cell. 1985;40(3):685-695.
  366. Hennings H, Steinert P, Buxman MM. Calcium induction of transglutaminase and the formation of epsilon(gamma-glutamyl) lysine cross-links in cultured mouse epidermal cells. Biochemical and biophysical research communications. 1981;102(2):739-745.
  367. Hennings H, Kruszewski FH, Yuspa SH, Tucker RW. Intracellular calcium alterations in response to increased external calcium in normal and neoplastic keratinocytes. Carcinogenesis. 1989;10(4):777-780.
  368. Gibson DF, Ratnam AV, Bikle DD. Evidence for separate control mechanisms at the message, protein, and enzyme activation levels for transglutaminase during calcium-induced differentiation of normal and transformed human keratinocytes. Journal of Investigative Dermatology. 1996;106(1):154-161.
  369. Su MJ, Bikle DD, Mancianti ML, Pillai S. 1,25-Dihydroxyvitamin D3 potentiates the keratinocyte response to calcium. The Journal of biological chemistry. 1994;269(20):14723-14729.
  370. Menon GK, Grayson S, Elias PM. Ionic calcium reservoirs in mammalian epidermis: ultrastructural localization by ion-capture cytochemistry. Journal of Investigative Dermatology. 1985;84(6):508-512.
  371. Mauro T, Bench G, Sidderas-Haddad E, Feingold K, Elias P, Cullander C. Acute barrier perturbation abolishes the Ca2+ and K+ gradients in murine epidermis: quantitative measurement using PIXE. Journal of Investigative Dermatology. 1998;111(6):1198-1201.
  372. Lee SH, Elias PM, Feingold KR, Mauro T. A role for ions in barrier recovery after acute perturbation. The Journal of investigative dermatology. 1994;102(6):976-979.
  373. Menon GK, Price LF, Bommannan B, Elias PM, Feingold KR. Selective obliteration of the epidermal calcium gradient leads to enhanced lamellar body secretion. The Journal of investigative dermatology. 1994;102(5):789-795.
  374. Lee SH, Elias PM, Proksch E, Menon GK, Mao-Quiang M, Feingold KR. Calcium and potassium are important regulators of barrier homeostasis in murine epidermis. The Journal of clinical investigation. 1992;89(2):530-538.
  375. Menon GK, Elias PM, Lee SH, Feingold KR. Localization of calcium in murine epidermis following disruption and repair of the permeability barrier. Cell and Tissue Research. 1992;270(3):503-512.
  376. Bikle DD, Ratnam A, Mauro T, Harris J, Pillai S. Changes in calcium responsiveness and handling during keratinocyte differentiation. Potential role of the calcium receptor. J Clin Invest. 1996;97(4):1085-1093.
  377. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC. Cloning and characterization of an extracellular Ca-2+-sensing receptor from bovine parathyroid. Nature. 1993;366(6455):575-580.
  378. Garrett JE, Capuano IV, Hammerland LG, Hung BC, Brown EM, Hebert SC, Nemeth EF, Fuller F. Molecular cloning and functional expression of human parathyroid calcium receptor cDNAs. Journal of Biological Chemistry. 1995;270(21):12919-12925.
  379. Tu CL, Chang W, Bikle DD. The extracellular calcium-sensing receptor Is Required for calcium- induced differentiation in human keratinocytes. The Journal of biological chemistry. 2001;276(44):41079-41085.
  380. Tu C, Chang W, Xie Z, Bikle D. Inactivation of the Calcium Sensing Receptor Inhibits E-cadherin-mediated Cell-Cell Adhesion and Calcium-induced Differentiation in Human Epidermal Keratinocytes. J Biol Chem. 2008;283:3519-3528.
  381. Tu CL, Crumrine DA, Man MQ, Chang W, Elalieh H, You M, Elias PM, Bikle DD. Ablation of the calcium-sensing receptor in keratinocytes impairs epidermal differentiation and barrier function. The Journal of investigative dermatology. 2012;132(10):2350-2359.
  382. Oda Y, Tu CL, Pillai S, Bikle DD. The calcium sensing receptor and its alternatively spliced form in keratinocyte differentiation. Journal of Biological Chemistry. 1998;273(36):23344-23352.
  383. Chang W, Tu C, Chen TH, Bikle D, Shoback D. The extracellular calcium-sensing receptor (CaSR) is a critical modulator of skeletal development. Sci Signal. 2008;1(35):ra1.
  384. Jaken S, Yuspa SH. Early signals for keratinocyte differentiation: role of Ca2+-mediated inositol lipid metabolism in normal and neoplastic epidermal cells. Carcinogenesis. 1988;9(6):1033-1038.
  385. Punnonen K, Denning M, Lee E, Li L, Rhee SG, Yuspa SH. Keratinocyte differentiation is associated with changes in the expression and regulation of phospholipase C isoenzymes. The Journal of investigative dermatology. 1993;101(5):719-726.
  386. Xie Z, Bikle DD. Phospholipase C-gamma1 is required for calcium-induced keratinocyte differentiation. The Journal of biological chemistry. 1999;274(29):20421-20424.
  387. Xie Z, Bikle DD. The recruitment of phosphatidylinositol 3-kinase to the E-cadherin-catenin complex at the plasma membrane is required for calcium-induced phospholipase C-gamma1 activation and human keratinocyte differentiation. The Journal of biological chemistry. 2007;282(12):8695-8703.
  388. Xie Z, Singleton PA, Bourguignon LY, Bikle DD. Calcium-induced human keratinocyte differentiation requires src- and fyn-mediated phosphatidylinositol 3-kinase-dependent activation of phospholipase C-gamma1. Molecular biology of the cell. 2005;16(7):3236-3246.
  389. Yuspa SH, Ben T, Hennings H, Lichti U. Divergent responses in epidermal basal cells exposed to the tumor promoter 12-O-tetradecanoylphorbol-13-acetate. Cancer Res. 1982;42(6):2344-2349.
  390. Dlugosz AA, Yuspa SH. Protein kinase C regulates keratinocyte transglutaminase (TG-K) gene expression in cultured primary mouse epidermal keratinocytes induced to terminally differentiate by calcium. Journal of Investigative Dermatology. 1994;102(4):409-414.
  391. Lu B, Rothnagel JA, Longley MA, Tsai SY, Roop DR. Differentiation-specific expression of human keratin 1 is mediated by a composite AP-1/steroid hormone element. The Journal of biological chemistry. 1994;269(10):7443-7449.
  392. Ng DC, Shafaee S, Lee D, Bikle DD. Requirement of an AP-1 site in the calcium response region of the involucrin promoter. The Journal of biological chemistry. 2000;275(31):24080-24088.
  393. Bikle DD, Ng D, Oda Y, Hanley K, Feingold K, Xie Z. The vitamin D response element of the involucrin gene mediates its regulation by 1,25-dihydroxyvitamin D3. The Journal of investigative dermatology. 2002;119(5):1109-1113.
  394. Hosomi J, Hosoi J, Abe E, Suda T, Kuroki T. Regulation of terminal differentiation of cultured mouse epidermal cells by 1 alpha,25-dihydroxyvitamin D3. Endocrinology. 1983;113(6):1950-1957.
  395. Stumpf WE, Sar M, Reid FA, Tanaka Y, DeLuca HF. Target cells for 1,25-dihydroxyvitamin D3 in intestinal tract, stomach, kidney, skin, pituitary, and parathyroid. Science. 1979;206(4423):1188-1190.
  396. Smith EL, Walworth NC, Holick MF. Effect of 1 alpha,25-dihydroxyvitamin D3 on the morphologic and biochemical differentiation of cultured human epidermal keratinocytes grown in serum-free conditions. The Journal of investigative dermatology. 1986;86(6):709-714.
  397. Pillai S, Bikle DD. Role of intracellular-free calcium in the cornified envelope formation of keratinocytes: differences in the mode of action of extracellular calcium and 1,25 dihydroxyvitamin D3. Journal of cellular physiology. 1991;146(1):94-100.
  398. McLane JA, Katz M, Abdelkader N. Effect of 1,25-dihydroxyvitamin D3 on human keratinocytes grown under different culture conditions. In Vitro Cell Dev Biol. 1990;26(4):379-387.
  399. McLaughlin JA, Cantley LC, Holick MF. 1,25(OH)2D3 increased calcium and phosphatidylinositol metabolism in differentiating cultured human keratinocytes. J Nutr Biochem. 1990;1:81-87.
  400. Ratnam AV, Bikle DD, Cho JK. 1,25 dihydroxyvitamin D3 enhances the calcium response of keratinocytes. Journal of Cellular Physiology. 1999;178(2):188-196.
  401. Pillai S, Bikle DD, Su MJ, Ratnam A, Abe J. 1,25-Dihydroxyvitamin D3 upregulates the phosphatidylinositol signaling pathway in human keratinocytes by increasing phospholipase C levels. The Journal of clinical investigation. 1995;96(1):602-609.
  402. Xie Z, Bikle DD. Inhibition of 1,25-dihydroxyvitamin-D-induced keratinocyte differentiation by blocking the expression of phospholipase C-gamma1. The Journal of investigative dermatology. 2001;117(5):1250-1254.
  403. Vandenberghe M, Raphael M, Lehen'kyi V, Gordienko D, Hastie R, Oddos T, Rao A, Hogan PG, Skryma R, Prevarskaya N. ORAI1 calcium channel orchestrates skin homeostasis. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(50):E4839-4848.
  404. Tu CL, Chang W, Bikle DD. The calcium-sensing receptor-dependent regulation of cell-cell adhesion and keratinocyte differentiation requires Rho and filamin A. The Journal of investigative dermatology. 2011;131(5):1119-1128.
  405. Tu CL, Chang W, Bikle DD. Phospholipase cgamma1 is required for activation of store-operated channels in human keratinocytes. The Journal of investigative dermatology. 2005;124(1):187-197.
  406. Malloy PJ, Pike JW, Feldman D. The vitamin D receptor and the syndrome of hereditary 1,25-dihydroxyvitamin D-resistant rickets. Endocrine reviews. 1999;20(2):156-188.
  407. Bikle DD, Chang S, Crumrine D, Elalieh H, Man MQ, Choi EH, Dardenne O, Xie Z, Arnaud RS, Feingold K, Elias PM. 25 Hydroxyvitamin D 1 alpha-hydroxylase is required for optimal epidermal differentiation and permeability barrier homeostasis. The Journal of investigative dermatology. 2004;122(4):984-992.
  408. Cianferotti L, Cox M, Skorija K, Demay MB. Vitamin D receptor is essential for normal keratinocyte stem cell function. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(22):9428-9433.
  409. Palmer HG, Martinez D, Carmeliet G, Watt FM. The vitamin D receptor is required for mouse hair cycle progression but not for maintenance of the epidermal stem cell compartment. The Journal of investigative dermatology. 2008;128(8):2113-2117.
  410. Plikus MV, Gay DL, Treffeisen E, Wang A, Supapannachart RJ, Cotsarelis G. Epithelial stem cells and implications for wound repair. Seminars in cell & developmental biology. 2012;23(9):946-953.
  411. Mascre G, Dekoninck S, Drogat B, Youssef KK, Brohee S, Sotiropoulou PA, Simons BD, Blanpain C. Distinct contribution of stem and progenitor cells to epidermal maintenance. Nature. 2012;489(7415):257-262.
  412. Tian XQ, Chen TC, Holick MF. 1,25-dihydroxyvitamin D3: a novel agent for enhancing wound healing. Journal of cellular biochemistry. 1995;59(1):53-56.
  413. Luderer HF, Nazarian RM, Zhu ED, Demay MB. Ligand-dependent actions of the vitamin D receptor are required for activation of TGF-beta signaling during the inflammatory response to cutaneous injury. Endocrinology. 2013;154(1):16-24.
  414. Song L, Papaioannou G, Zhao H, Luderer HF, Miller C, Dall'Osso C, Nazarian RM, Wagers AJ, Demay MB. The Vitamin D Receptor Regulates Tissue Resident Macrophage Response to Injury. Endocrinology. 2016;157(10):4066-4075.
  415. Oda Y, Tu CL, Menendez A, Nguyen T, Bikle DD. Vitamin D and calcium regulation of epidermal wound healing. The Journal of steroid biochemistry and molecular biology. 2015.
  416. Chen S, Lewallen M, Xie T. Adhesion in the stem cell niche: biological roles and regulation. Development. 2013;140(2):255-265.
  417. Lechler T, Fuchs E. Asymmetric cell divisions promote stratification and differentiation of mammalian skin. Nature. 2005;437(7056):275-280.
  418. Li L, Hartley R, Reiss B, Sun Y, Pu J, Wu D, Lin F, Hoang T, Yamada S, Jiang J, Zhao M. E-cadherin plays an essential role in collective directional migration of large epithelial sheets. Cellular and molecular life sciences : CMLS. 2012;69(16):2779-2789.
  419. Bikle DD, Ng D, Tu CL, Oda Y, Xie Z. Calcium- and vitamin D-regulated keratinocyte differentiation. Mol Cell Endocrinol. 2001;177(1-2):161-171.
  420. van Etten E, Mathieu C. Immunoregulation by 1,25-dihydroxyvitamin D3: basic concepts. The Journal of steroid biochemistry and molecular biology. 2005;97(1-2):93-101.
  421. Daniel C, Sartory NA, Zahn N, Radeke HH, Stein JM. Immune modulatory treatment of trinitrobenzene sulfonic acid colitis with calcitriol is associated with a change of a T helper (Th) 1/Th17 to a Th2 and regulatory T cell profile. The Journal of pharmacology and experimental therapeutics. 2008;324(1):23-33.
  422. Gregori S, Casorati M, Amuchastegui S, Smiroldo S, Davalli AM, Adorini L. Regulatory T cells induced by 1 alpha,25-dihydroxyvitamin D3 and mycophenolate mofetil treatment mediate transplantation tolerance. Journal of immunology. 2001;167(4):1945-1953.
  423. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775-787.
  424. Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A, Soler D, Butcher EC. DCs metabolize sunlight-induced vitamin D3 to 'program' T cell attraction to the epidermal chemokine CCL27. Nat Immunol. 2007;8(3):285-293.
  425. Chen S, Sims GP, Chen XX, Gu YY, Chen S, Lipsky PE. Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation. Journal of immunology. 2007;179(3):1634-1647.
  426. Bouillon R, Carmeliet G, Verlinden L, van Etten E, Verstuyf A, Luderer HF, Lieben L, Mathieu C, Demay M. Vitamin D and human health: lessons from vitamin D receptor null mice. Endocrine reviews. 2008;29(6):726-776.
  427. Adorini L, Penna G. Control of autoimmune diseases by the vitamin D endocrine system. Nat Clin Pract Rheumatol. 2008;4(8):404-412.
  428. Adamopoulos IE, Bowman EP. Immune regulation of bone loss by Th17 cells. Arthritis Res Ther. 2008;10(5):225.
  429. Limketkai BN, Mullin GE, Limsui D, Parian AM. Role of Vitamin D in Inflammatory Bowel Disease. Nutr Clin Pract. 2017;32(3):337-345.
  430. Mathieu C, Van Etten E, Gysemans C, Decallonne B, Kato S, Laureys J, Depovere J, Valckx D, Verstuyf A, Bouillon R. In vitro and in vivo analysis of the immune system of vitamin D receptor knockout mice. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2001;16(11):2057-2065.
  431. O'Kelly J, Hisatake J, Hisatake Y, Bishop J, Norman A, Koeffler HP. Normal myelopoiesis but abnormal T lymphocyte responses in vitamin D receptor knockout mice. The Journal of clinical investigation. 2002;109(8):1091-1099.
  432. Josien R, Li HL, Ingulli E, Sarma S, Wong BR, Vologodskaia M, Steinman RM, Choi Y. TRANCE, a tumor necrosis factor family member, enhances the longevity and adjuvant properties of dendritic cells in vivo. The Journal of experimental medicine. 2000;191(3):495-502.
  433. Baroni E, Biffi M, Benigni F, Monno A, Carlucci D, Carmeliet G, Bouillon R, D'Ambrosio D. VDR-dependent regulation of mast cell maturation mediated by 1,25-dihydroxyvitamin D3. J Leukoc Biol. 2007;81(1):250-262.
  434. Froicu M, Weaver V, Wynn TA, McDowell MA, Welsh JE, Cantorna MT. A crucial role for the vitamin D receptor in experimental inflammatory bowel diseases. Molecular endocrinology (Baltimore, Md). 2003;17(12):2386-2392.
  435. Gysemans C, van Etten E, Overbergh L, Giulietti A, Eelen G, Waer M, Verstuyf A, Bouillon R, Mathieu C. Unaltered diabetes presentation in NOD mice lacking the vitamin D receptor. Diabetes. 2008;57(1):269-275.
  436. Topilski I, Flaishon L, Naveh Y, Harmelin A, Levo Y, Shachar I. The anti-inflammatory effects of 1,25-dihydroxyvitamin D3 on Th2 cells in vivo are due in part to the control of integrin-mediated T lymphocyte homing. European journal of immunology. 2004;34(4):1068-1076.
  437. Wittke A, Weaver V, Mahon BD, August A, Cantorna MT. Vitamin D receptor-deficient mice fail to develop experimental allergic asthma. Journal of immunology. 2004;173(5):3432-3436.
  438. Wittke A, Chang A, Froicu M, Harandi OF, Weaver V, August A, Paulson RF, Cantorna MT. Vitamin D receptor expression by the lung micro-environment is required for maximal induction of lung inflammation. Archives of biochemistry and biophysics. 2007;460(2):306-313.
  439. Adorini L. Intervention in autoimmunity: the potential of vitamin D receptor agonists. Cellular immunology. 2005;233(2):115-124.
  440. Ehrchen J, Helming L, Varga G, Pasche B, Loser K, Gunzer M, Sunderkotter C, Sorg C, Roth J, Lengeling A. Vitamin D receptor signaling contributes to susceptibility to infection with Leishmania major. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2007;21(12):3208-3218.
  441. Rajapakse R, Mousli M, Pfaff AW, Uring-Lambert B, Marcellin L, Bronner C, Jeanblanc M, Villard O, Letscher-Bru V, Klein JP, Candolfi E. 1,25-Dihydroxyvitamin D3 induces splenocyte apoptosis and enhances BALB/c mice sensitivity to toxoplasmosis. The Journal of steroid biochemistry and molecular biology. 2005;96(2):179-185.
  442. Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, Gilliet M, Ho S, Antonenko S, Lauerma A, Smith K, Gorman D, Zurawski S, Abrams J, Menon S, McClanahan T, de Waal-Malefyt Rd R, Bazan F, Kastelein RA, Liu YJ. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3(7):673-680.
  443. Liu PT, Krutzik SR, Modlin RL. Therapeutic implications of the TLR and VDR partnership. Trends Mol Med. 2007;13(3):117-124.
  444. Schauber J, Gallo RL. The vitamin D pathway: a new target for control of the skin's immune response? Experimental dermatology. 2008;17(8):633-639.
  445. Gombart AF, Borregaard N, Koeffler HP. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2005;19(9):1067-1077.
  446. Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, Tavera-Mendoza L, Lin R, Hanrahan JW, Mader S, White JH. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. Journal of immunology. 2004;173(5):2909-2912.
  447. Schauber J, Dorschner RA, Coda AB, Buchau AS, Liu PT, Kiken D, Helfrich YR, Kang S, Elalieh HZ, Steinmeyer A, Zugel U, Bikle DD, Modlin RL, Gallo RL. Injury enhances TLR2 function and antimicrobial peptide expression through a vitamin D-dependent mechanism. The Journal of clinical investigation. 2007;117(3):803-811.
  448. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, Ochoa MT, Schauber J, Wu K, Meinken C, Kamen DL, Wagner M, Bals R, Steinmeyer A, Zugel U, Gallo RL, Eisenberg D, Hewison M, Hollis BW, Adams JS, Bloom BR, Modlin RL. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770-1773.
  449. Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, Gallo RL, Leung DY. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. The New England journal of medicine. 2002;347(15):1151-1160.
  450. Howell MD, Gallo RL, Boguniewicz M, Jones JF, Wong C, Streib JE, Leung DY. Cytokine milieu of atopic dermatitis skin subverts the innate immune response to vaccinia virus. Immunity. 2006;24(3):341-348.
  451. Bilezikian JP, Bikle D, Hewison M, Lazaretti-Castro M, Formenti AM, Gupta A, Madhavan MV, Nair N, Babalyan V, Hutchings N, Napoli N, Accili D, Binkley N, Landry DW, Giustina A. MECHANISMS IN ENDOCRINOLOGY: Vitamin D and COVID-19. Eur J Endocrinol. 2020;183(5):R133-R147.
  452. Nogues X, Ovejero D, Pineda-Moncusi M, Bouillon R, Arenas D, Pascual J, Ribes A, Guerri-Fernandez R, Villar-Garcia J, Rial A, Gimenez-Argente C, Cos ML, Rodriguez-Morera J, Campodarve I, Quesada-Gomez JM, Garcia-Giralt N. Calcifediol Treatment and COVID-19-Related Outcomes. The Journal of clinical endocrinology and metabolism. 2021;106(10):e4017-e4027.
  453. Ustianowski A, Shaffer R, Collin S, Wilkinson RJ, Davidson RN. Prevalence and associations of vitamin D deficiency in foreign-born persons with tuberculosis in London. J Infect. 2005;50(5):432-437.
  454. Rook GA, Steele J, Fraher L, Barker S, Karmali R, O'Riordan J, Stanford J. Vitamin D3, gamma interferon, and control of proliferation of Mycobacterium tuberculosis by human monocytes. Immunology. 1986;57(1):159-163.
  455. Liu PT, Stenger S, Tang DH, Modlin RL. Cutting edge: vitamin D-mediated human antimicrobial activity against Mycobacterium tuberculosis is dependent on the induction of cathelicidin. Journal of immunology. 2007;179(4):2060-2063.
  456. Sly LM, Lopez M, Nauseef WM, Reiner NE. 1alpha,25-Dihydroxyvitamin D3-induced monocyte antimycobacterial activity is regulated by phosphatidylinositol 3-kinase and mediated by the NADPH-dependent phagocyte oxidase. The Journal of biological chemistry. 2001;276(38):35482-35493.
  457. Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, Bleharski JR, Maitland M, Norgard MV, Plevy SE, Smale ST, Brennan PJ, Bloom BR, Godowski PJ, Modlin RL. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science. 1999;285(5428):732-736.
  458. Salahuddin N, Ali F, Hasan Z, Rao N, Aqeel M, Mahmood F. Vitamin D accelerates clinical recovery from tuberculosis: results of the SUCCINCT Study [Supplementary Cholecalciferol in recovery from tuberculosis]. A randomized, placebo-controlled, clinical trial of vitamin D supplementation in patients with pulmonary tuberculosis'. BMC Infect Dis. 2013;13:22.
  459. Martineau AR, Timms PM, Bothamley GH, Hanifa Y, Islam K, Claxton AP, Packe GE, Moore-Gillon JC, Darmalingam M, Davidson RN, Milburn HJ, Baker LV, Barker RD, Woodward NJ, Venton TR, Barnes KE, Mullett CJ, Coussens AK, Rutterford CM, Mein CA, Davies GR, Wilkinson RJ, Nikolayevskyy V, Drobniewski FA, Eldridge SM, Griffiths CJ. High-dose vitamin D(3) during intensive-phase antimicrobial treatment of pulmonary tuberculosis: a double-blind randomised controlled trial. Lancet. 2011;377(9761):242-250.
  460. Schauber J, Oda Y, Buchau AS, Steinmeyer A, Zugel U, Bikle DD, Gallo RL. Histone acetylation in keratinocytes enables control of the expression of cathelicidin and CD14 by 1,25-dihydroxyvitamin D3. The Journal of investigative dermatology. 2008;128(4):816-824.
  461. Norman AW, Roth J, Orci L. The vitamin D endocrine system: steroid metabolism, hormone receptors, and biological response (calcium binding proteins). Endocr Rev. 1982;3(4):331-366.
  462. Weishaar RE, Kim SN, Saunders DE, Simpson RU. Involvement of vitamin D3 with cardiovascular function. III. Effects on physical and morphological properties. Am J Physiol. 1990;258(1 Pt 1):E134-142.
  463. Walters MR, Ilenchuk TT, Claycomb WC. 1,25-Dihydroxyvitamin D3 stimulates 45Ca2+ uptake by cultured adult rat ventricular cardiac muscle cells. The Journal of biological chemistry. 1987;262(6):2536-2541.
  464. Selles J, Boland R. Rapid stimulation of calcium uptake and protein phosphorylation in isolated cardiac muscle by 1,25-dihydroxyvitamin D3. Mol Cell Endocrinol. 1991;77(1-3):67-73.
  465. Wu J, Garami M, Cao L, Li Q, Gardner DG. 1,25(OH)2D3 suppresses expression and secretion of atrial natriuretic peptide from cardiac myocytes. Am J Physiol. 1995;268(6 Pt 1):E1108-1113.
  466. Gardner DG, Chen S, Glenn DJ. Vitamin D and the heart. Am J Physiol Regul Integr Comp Physiol. 2013;305(9):R969-977.
  467. Boland R. Role of vitamin D in skeletal muscle function. Endocr Rev. 1986;7(4):434-448.
  468. Girgis CM, Clifton-Bligh RJ, Hamrick MW, Holick MF, Gunton JE. The roles of vitamin D in skeletal muscle: form, function, and metabolism. Endocrine reviews. 2013;34(1):33-83.
  469. Boland RL. VDR activation of intracellular signaling pathways in skeletal muscle. Mol Cell Endocrinol. 2011;347(1-2):11-16.
  470. Gelbard HA, Stern PH, U'Prichard DC. 1 alpha, 25-Dihydroxyvitamin D3 nuclear receptors in pituitary. Science. 1980;209(4462):1247-1249.
  471. Haug E, Gautvik KM. Demonstration and characterization of a 1 alpha,25-(OH)2D3 receptor- like macromolecule in cultured rat pituitary cells. J Steroid Biochem. 1985;23(5A):625-635.
  472. Tornquist K. Pretreatment with 1,25-dihydroxycholecalciferol enhances thyrotropin- releasing hormone- and inositol 1,4,5-trisphosphate-induced release of sequestered Ca2+ in permeabilized GH4C1 pituitary cells. Endocrinology. 1992;131(4):1677-1681.
  473. Tornquist K, Lamberg-Allardt C. The effect of 1,25-dihydroxy-cholecalciferol on the TRH induced TSH release in rats. Acta Endocrinol (Copenh). 1987;114(1):55-59.
  474. Narbaitz R, Sar M, Stumpf WE, Huang S, DeLuca HF. 1,25-Dihydroxyvitamin D3 target cells in rat mammary gland. Horm Res. 1981;15(4):263-269.
  475. Lopes N, Paredes J, Costa JL, Ylstra B, Schmitt F. Vitamin D and the mammary gland: a review on its role in normal development and breast cancer. Breast Cancer Res. 2012;14(3):211.
  476. Eisman JA, Suva LJ, Martin TJ. Significance of 1,25-dihydroxyvitamin D3 receptor in primary breast cancers. Cancer Res. 1986;46(10):5406-5408.
  477. Eisman JA, Sutherland RL, McMenemy ML, Fragonas JC, Musgrove EA, Pang GY. Effects of 1,25-dihydroxyvitamin D3 on cell-cycle kinetics of T 47D human breast cancer cells. J Cell Physiol. 1989;138(3):611-616.
  478. Colston KW, Berger U, Coombes RC. Possible role for vitamin D in controlling breast cancer cell proliferation. Lancet. 1989;1(8631):188-191.
  479. Duncan WE, Whitehead D, Wray HL. A 1,25-dihydroxyvitamin D3 receptor-like protein in mammalian and avian liver nuclei. Endocrinology. 1988;122(6):2584-2589.
  480. Cao Y, Shu XB, Yao Z, Ji G, Zhang L. Is vitamin D receptor a druggable target for non-alcoholic steatohepatitis? World J Gastroenterol. 2020;26(38):5812-5821.
  481. Rixon RH, MacManus JP, Whitfield JF. The control of liver regeneration by calcitonin, parathyroid hormone and 1 alpha,25-dihydroxycholecalciferol. Mol Cell Endocrinol. 1979;15(2):79-89.
  482. Nguyen TM, Guillozo H, Marin L, Dufour ME, Tordet C, Pike JW, Garabedian M. 1,25-dihydroxyvitamin D3 receptors in rat lung during the perinatal period: regulation and immunohistochemical localization. Endocrinology. 1990;127(4):1755-1762.
  483. Gaultier C, Harf A, Balmain N, Cuisinier-Gleizes P, Mathieu H. Lung mechanics in rachitic rats. Am Rev Respir Dis. 1984;130(6):1108-1110.
  484. Herscovitch K, Dauletbaev N, Lands LC. Vitamin D as an anti-microbial and anti-inflammatory therapy for Cystic Fibrosis. Paediatr Respir Rev. 2014;15(2):154-162.

 

Diabetes Mellitus in People with Cancer

ABSTRACT

 

There is increasing evidence of an association between cancer and diabetes mellitus. Patients with type II diabetes are at increased risk of malignancy due to shared risk factors between the two conditions, and people with a diagnosis of cancer may develop new onset diabetes or impaired glycemic control, partly as a result of the systemic anti-cancer treatments (SACT) they receive. Many newer targeted anti-cancer treatments can have off-target metabolic toxicities not seen with conventional chemotherapy agents. Early recognition of diabetes or hyperglycemia in people with cancer can improve outcomes. This chapter aims to summarize these associations, provide an overview of how different SACT modalities can impact on glycemic control, and highlight key recommendations for the management of this complex patient group.

 

INTRODUCTION

 

Diabetes mellitus (DM) is a rising global public health emergency, with recent estimates suggesting that over 780 million people globally will be affected by 2045 (1). DM is typically classified into broad categories including type 1 (T1DM), type 2 (T2DM), gestational, monogenic, pharmacologically-induced, endocrinopathy-driven and DM due to pancreatic disease/deficiency (sometimes referred to as type 3c) (2, 3). T2DM is regarded as the most common subtype and is reported to account for over 85% of cases (1). All types of DM can lead to multisystem microvascular (nephropathy, retinopathy, neuropathy) and macrovascular (ischemic heart disease, stroke and peripheral vascular disease) complications, with management of these complications placing a strain upon many health services.

 

People with a diagnosis of DM are also at higher risk for developing several cancers (4), with reasons for this in part due to shared risk factors between the two, including age, obesity, sedentary lifestyle, and diet (5,6). A recent umbrella review of meta-analyses found that risks of developing most cancers were higher in people with DM compared to those without, with the most convincing evidence seen in breast cancer, intrahepatic cholangiocarcinoma, colorectal cancer, and endometrial cancer. One exception in this study was prostate cancer, where the risk appeared lower in individuals with DM (4). In view of this increased cancer risk in people with DM, some groups even advocate that regular screening for underlying cancer should be part of routine DM assessments (7).

 

It is estimated that approximately 20% of people with cancer have concurrent diabetes (8). Individuals with cancer are also at an increased risk of developing new onset DM or hyperglycemia, independent of an underlying diagnosis of diabetes, whilst cancer patients with concurrent DM often experience worsening glycemic control (9). Reasons for poor glycemic control in these individuals include complications from systemic anticancer treatments (SACT) along with supportive medications to treat treatment side effects, and symptoms of the underlying malignancy. This chapter aims to summarize the complex relationship between malignancy and DM, particularly the effects of SACT on glycemic control and risk of DM, as well as outlining management guidelines for DM in people with cancer.

 

DIABETES/HYPERGLYCEMIA AND CANCER OUTCOMES                       

 

A number of observational studies have demonstrated that hyperglycemia is associated with poorer overall survival (OS) and increased risk of disease recurrence in a number of malignancies, solid and hematological (10-17), with a number of individual studies, and larger meta-analyses supporting this. One meta-analysis reviewed 12 studies comprising 9,872 people with a diagnosis of cancer without known diabetes. Individuals with hyperglycemia were found to have significantly worse disease-free survival (DFS) (hazard ratio (HR) 1.98, 95% confidence interval (CI) 1.20-3.27) compared to those without, as well as worse OS (HR 2.05, 95% CI 1.67-2.551) (18). A further meta-analysis of 4,241 patients with pancreatic cancer suggested that those individuals with concurrent DM (1,034) have poorer OS (HR 1.16, 95% CI 1.08-1.25) and a higher risk of on-treatment death than those without concurrent DM (19). Furthermore, in a meta-analysis of 8 studies in breast cancer, concurrent DM was found to confer a greater risk of death, and a later stage at presentation, as well as impact on the treatment given (20). People with DM also have a higher prevalence of oral cancers, as well as a higher mortality from these cancers (21).

 

In addition to this, a number of preclinical studies have suggested that hyperglycemia may specifically attenuate the efficacy of chemotherapy in people with cancer with or without diabetes, which could in part account for these observations (22). For example, hyperglycemia may attenuate chemotherapy-induced reactive oxygen species (ROS) production, which in-turn can diminish the efficacy of treatment (23). In vivo, there are some small series that have demonstrated an association between hyperglycemia and resistance to chemotherapy. A clinical study of 88 people with estrogen-receptor positive breast cancer demonstrated impaired glucose tolerance significantly correlated with disease progression in those patients receiving chemotherapy (24). Furthermore, high blood glucose levels irrespective of an underlying DM diagnosis, were shown to significantly enhance oxaliplatin resistance in individuals with stage III colorectal cancer receiving adjuvant chemotherapy (22). Studies such as these highlight the importance of adequate glycemic control during treatment for cancer to potentially improve outcomes, although these data are mainly from observational studies, with interventional studies lacking.

 

EFFECT OF DIABETES OR HYPERGLYCEMIA ON QUALITY OF LIFE IN PEOPLE WITH CANCER

 

Cancer-related symptoms and SACT side effects, such as fatigue, nausea, anorexia and pain can be debilitating to patients. When confounded by symptoms of hyperglycemia, the impact upon an individuals’ quality of life can be significant (25). Furthermore, the impact of a cancer diagnosis, as well as treatment and cancer-related symptoms can have major negative impacts on diabetes self-care (26), with data suggesting that adherence to glucose lowering drugs often decreases in individuals following a cancer diagnosis (27). A cancer diagnosis can also have financial and social impacts upon individuals, affecting access to healthy food and outpatient diabetes services, resulting in lower quality of life and a higher symptom burden (28). A systematic review of 10 studies, demonstrated poorer patient reported outcomes (PROs) in those diagnosed with both cancer and DM compared to having either one of these diseases alone (29).

 

 

People with DM are known to be at higher risk from infections, and undergoing SACT can exacerbate this, resulting in higher rates of infection and hospitalization observed in those with cancer and DM (30, 31). This in turn leads to higher rates of chemotherapy dose reductions and early treatment cessation (28, 32-34). A meta-analysis of 10 observational studies involving 8,688 cases found that the likelihood of developing chemotherapy-induced neutropenia was higher amongst individuals with DM/hyperglycemia than those without (odds ratio (OR) 1.32, 95% CI 1.06-1.64) (31). Chemotherapy-induced neutropenia poses a significant risk for infection and hospitalization in all people with cancer, with an associated rate of morbidity and mortality which is higher in those with raised blood glucose levels (30). In addition to severe hematological toxicity, more severe rates of non-hematological toxicity have also been associated with hyperglycemia during chemotherapy in people with prostate cancer and lymphoma (35). A single-center retrospective analysis found that individuals with cancer and DM who had good glycemic control had no increased risk of treatment-related complications compared with individuals without DM (36), suggesting that optimal glycemic control during SACT could improve tolerability, thereby reducing rates of admission and dose-limiting toxicity.

 

Conceivably, people with DM may be more prone to neuro- and nephrotoxic agents due to their underlying predisposition conferred by the DM. Indeed, a previous report suggested that taxane-based chemotherapy regimens resulted in a significantly higher rates of peripheral neuropathy in those with DM compared to those without (74.4% vs. 58.5%) (37). There are no convincing data to suggest that a concurrent cancer diagnosis accelerates the risk of diabetic nephropathy or retinopathy.

 

EFFECTS OF SYSTEMIC ANTICANCER THERAPIES ON GLYCEMIA  

 

Systemic anti-cancer therapies (SACT) encompass a wide range of treatments including cytotoxic chemotherapy, hormone therapy, targeted therapy, and immunotherapy, many of which can impact upon glycemic control directly or as a result of toxicity management or supportive medications which are given alongside treatment. Several anti-cancer agents have been demonstrated to increase the risk of hyperglycemia as summarized in Table 1, and many can do this even in those without a known diagnosis of DM. People receiving SACT are also at risk of developing a new diagnosis of diabetes. One study demonstrated that 11% of people (15/134) undergoing routine chemotherapy met the criteria for a new diagnosis of diabetes (using the diagnostic criteria as per guidelines from the UK National Institute for Clinical and Healthcare Excellence (NICE) and without a previous known diagnosis) based upon HbA1cmeasurements). The majority of these individuals (73%) had been receiving short course steroids with chemotherapy, and 40% were being treated in the curative/adjuvant setting (38). A second prospective cohort study in 90 people taking glucocorticoids as part of therapy protocols for primary brain tumor or metastases, lymphoma, or for bone marrow transplant, found non-DM range hyperglycemia in 58% and DM-range hyperglycemia in 18.9% (39). These individuals with hyperglycemia are also more likely to present with an emergency admission during cancer therapy than those with normoglycemia (40).

 

Table 1. SACT used in the Treatment of Cancer Demonstrated to be Associated with Worsening Glycemic Control

Type of SACT

Drug Examples

Risk of Diabetes/Hyperglycemia (Range of any grade)

Type of diabetes most likely to develop

Targeted therapy

 

mTOR inhibitors

Everolimus (41, 42)

12-50%

T2DM

Temsirolimus (42)

26%

PI3K inhibitors

Alpelisib (43)

37%

T2DM

Idelialisib (44)

28/30%

EGFR inhibitor

Osimertinib (45)

2%

T2DM

Panitumumab (46, 47)

1-10%

Multikinase inhibitor

Sunitinib (48-50)

0-8%

Risk of hypoglycemia

Reverses T1/T2DM, but also causes hyperglycemia

Pazopanib (50)

Tyrosine kinase inhibitor (TKI)

Nilotinib (51)

6%

T2DM

Ponatinib (52)

3%

ALK Inhibitor

Ceritinib (53)

49%

T2DM

FLT3 inhibitor

Midostaurin (54, 55)

7-20%

T2DM

Gilteritinib (56)

13%

Monoclonal antibody

Gemtuzumab (anti-CD33) *inpatient use (57)

10%

T2DM

Somatostatin Analogues

Octreotide, Lanreotide (58)

Up to 30%

T2DM, but risk of hypoglycemia

Chemotherapy

 

Anti-metabolite

5-fluorouracil (59, 60)

Up to 10%

T2DM

Pemetrexed (61, 62)

4%

Decitadine/Azacitidine (63)

6-33%

Alkylating agents

Busulfan (64)

66-67%

Platinum based

Oxaliplatin (65, 66)

4%

Anthracyclines

Doxorubicin (60, 67)

Up to 10%

Other

Arsenic trioxide (ATO) (68)

45%

 

Immune Checkpoint Inhibitors

 

PD-1

Nivolumab (69)

<1%

T1DM

Pembrolizumab (70)

1-2.2%

CTLA-4

Ipilumumab (69)

<1%

 

Combination ICP (71)

4%

Hormone Therapy

 

Hormone Treatment

ADT (44, 72)

Risk ratio 1.39 (95% CI 1.27-1.53) n=65,595 cases

T2DM

Tamoxifen (73)

Diabetes risk adj. odds ratio 1.24 (95% CI 1.08-1.42)

Abbreviations: ADT = androgen deprivation therapy; ALK – anaplastic lymphoma kinase; ATO – arsenic trioxide; CTLA-4 – cytotoxic T-lymphocyte protein-4; EGFR – epidermal growth factor receptor; FLT3 – FMS-like tyrosine kinase-3; ICP – immune checkpoint inhibitor; TKI – tyrosine kinase inhibitor; mTOR – mechanistic target of rapamycin; PI3K – phosphoinositide-3 kinase; PD-1 – programmed cell death protein-1; T1DM – type 1 diabetes mellitus; T2DM – type 2 diabetes mellitus

 

Cytotoxic Chemotherapy

 

Hyperglycemia occurs in between 10 and 30% of people undergoing cytotoxic chemotherapy for malignancy (74), and although often transient during treatment, can persist, or even lead to DM in some people. Poor glycemic control can increase the risk of infections and hospitalization (28, 34), as previously discussed, leading to treatment interruptions and dose reductions, as well as significant morbidity, and even mortality (33). A number of cytotoxic chemotherapy regimens are reported to cause hyperglycemia in people without diabetes, including commonly used drugs such as 5-fluorouracil (5-FU), platinum-based drugs (oxaliplatin, carboplatin, cisplatin) and anthracyclines (doxorubicin, epirubicin) (75). In one cohort study of 422 people receiving 5FU-based chemotherapy regimens for the treatment of early or advanced colorectal cancer, 11.6% (42 people) developed diabetes and a further 11.3% developed impaired fasting blood glucose (FBG) levels. Of the 42 people who developed diabetes, 7 required no treatment, 13 received diet control and physiotherapy only, and 22 received antidiabetic medication (75). In a second cohort of 185 people with head and neck cancer treated with platinum-containing regimens, 3.8% developed type 2 DM, with 3 presenting with hyperglycemic crises (DKA, HHS) (65). One possible contributing factor for developing impaired FBG levels and/or type 2 DM is the concurrent use of corticosteroids in highly emetogenic chemotherapy regimens, but an analysis of type 2 DM following anthracycline use in 3,147 lymphoma patients suggested that the use of these drugs independently increases the risk of T2DM, when data was adjusted for corticosteroid use, comorbidities, age, and gender. A threshold doxorubicin dose of 253mg was identified, below which there was no increased risk of developing T2DM (76). Risk of diabetes from cytotoxic chemotherapy may also increase with age, with one pediatric study suggesting that the risk was higher in acute lymphoblastic leukemia (ALL) patients aged > 10, compared with those < 10 years old (77).

 

Exact mechanisms of how and why some cytotoxic chemotherapies can lead to hyperglycemia or T2DM remain unclear. Proposed mechanisms include the induction of an inflammatory state which predisposes to hyperglycemia (78) or direct metabolic effects on tissues vital to glucose homeostasis such as skeletal muscles (79).

 

Oral Targeted Anticancer Agents

 

Many new targeted cancer therapies inhibit various points in the insulin receptor signaling pathway including the commonly used class of tyrosine kinase inhibitors (TKIs) (80). Reported effects of targeted TKIs on blood glucose metabolism range from the development of metabolic syndrome and diabetes via the blocking of insulin signaling (80), as well as erratic glycemic control and even hypoglycemia in those with pre-existing type 1 or type 2 DM49, (81, 82). In contrast some TKIs may improve glycemic control suggesting that management of these individuals needs to be individualized with no one-size-fits-all management algorithm. Reversibility of these effects is also unclear, with reported improvements in glycemic control and HbA1c levels following dose reductions or treatment termination (83).

 

Inhibitors of mTOR (everolimus, temsirolimus or ridaforolimus) have also been shown to impact glycemic control since mTOR is a protein kinase that plays a key role in regulating cell growth as well as lipid and glucose metabolism (84, 85). Meta-analyses looking into these effects have demonstrated significantly higher rates of hyperglycemia, hypercholesterolemia, and hypertriglyceridemia compared with controls (86, 87) In isolated cases, the effects have been severe enough to precipitate DKA (88). To date, studies have not demonstrated either positive or negative associations between treatment response rates and incidence of metabolic complications (89).

 

As novel targeted agents continue to be introduced to manage a range of cancers, it is expected that metabolic toxicities continue to be reported given the homeostatic function of many of these druggable targets. Whilst some of these agents will provide meaningful benefit in terms of survival for people with advanced cancers, such as the PI3Kainhibitor alpelisib for PI3KA-mutated metastatic breast cancer (43), glycemic control needs to be at the forefront of the prescriber’s mind at initiation, to ensure adequate management of toxicities.

 

Hormone Therapy

 

ANDROGEN DEPRIVATION THERAPY

 

Androgen deprivation therapy (ADT) is recognized as a risk factor for development of diabetes, metabolic syndrome, and cardiovascular disease (72, 90, 91). In a large observational study of over 35,000 men treated for prostate cancer, ADT in the form of gonadotropin-releasing hormone (GnRH) agonists, oral antiandrogens, a combination of the two, or orchiectomy was associated with a significantly increased risk of diabetes, coronary heart disease, myocardial infarction, and sudden cardiac death (90). These findings are supported by other studies, including a meta-analysis of over 150,000 men with prostate cancer receiving ADT (72), with association observed with all forms of ADT, with the weakest association with anti-androgen therapy alone.

 

ESTROGEN TARGETED THERAPY

 

Studies examining the effect of estrogen-targeted therapies on the development of diabetes in women with breast cancer are less clear cut. Whilst one retrospective cohort analysis failed to demonstrate a link between tamoxifen use and the development of DM (92), two large population-based studies demonstrated a significant association between tamoxifen use and the development of diabetes in women diagnosed with breast cancer (73, 93); The first of these studies included almost 15,000 Canadian women aged 65 years or older diagnosed with early breast cancer, whilst the second included over 22,000 women in Taiwan aged 20 years and over. Whilst tamoxifen appears to increase the risk of developing DM, aromatase inhibitor therapy does not, with no link found in any of these three studies.

 

Immune Checkpoint Inhibitors

 

Immune checkpoint inhibitors (ICPi), including cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PDL-1) inhibitors are a sub-class of monoclonal antibody treatments that have revolutionized cancer treatment over the last decade. First approved for use in the treatment of melanoma, ICPi are now recognized as providing a survival benefit across a number of cancers, and are increasingly used in early-stage cancers in the adjuvant setting and also in combination with chemotherapy (94). Whilst clinically effective, ICPi can lead to a spectrum of immune-related adverse events (IRAEs). Endocrine IRAEs include hypophysitis, thyroiditis, adrenalitis and de novo diabetes. The risk of developing de novo diabetes is low, occurring in 0.2-4% of ICPi treated individuals depending on the immunotherapy given (69). The immune checkpoint PD-1 and its ligand PD-L1 have been shown to have an important immune homeostatic function in the pancreas by promoting beta cell maturation and preventing immune-mediated beta cell destruction (95). To date, there is no convincing evidence for a physiological role for CTLA-4 within the pancreas. PD-1 inhibitors, PD-L1 inhibitors, and combination CTLA-4/PD-1 therapy have been demonstrated to precipitate diabetes more commonly than CTLA-4 inhibitors alone. The underlying clinical presentation is akin to type 1 diabetes (70) and believed to be precipitated by inappropriate activation of self-reactive T-cells and destruction of insulin-producing pancreatic islet β-cells. ICP-induced insulin deficiency may lead to new-onset insulin-dependent diabetes or worsening pre-existing type 2 diabetes. Up to 75% of people who develop ICP-induced hyperglycemia/diabetes present with diabetic ketoacidosis (DKA) (96-98). Presentations are frequently acute with a precipitous increase in blood glucose (99). Therefore ICP-induced diabetes can be discriminated from ‘standard’ type 1 diabetes mellitus, by its tendency towards a faster onset, apparently fulminant course, and high degree of antibody negativity (99). The nomenclature of the condition in the published literature varies mainly between ‘type 1 like’ to ‘fulminant’ with there being differences between the presentation of ICPi-induced diabetes and type 1 and fulminant diabetes. Kyriacou and colleagues compared the characteristics of 75 published cases and concluded that there is some overlap with type 1 DM and fulminant DM. However, this was felt to be insufficient overlap for ICPi diabetes to be wholly classified as either type 1 like or fulminant (100). Nevertheless, the recognition that these agents can precipitate rapid beta cell destruction which results in an unusually high number of emergency presentations is key. Treatment of non-endocrine IRAEs is typically with high dose steroids, often for prolonged periods of time. At present, steroids are used in up to a third of people receiving ICPs, further increasing the risk of hyperglycemia, and steroid induced T2DM.

 

An analysis of the World Health Organizations (WHO) pharmacovigilance database over a 4-year period detected 283 cases of ICP-induced diabetes mellitus, 50.2% of which presented with DKA, and 6% of whom were on concurrent steroids at diagnosis (101). There was a wide variability in duration of ICP treatment, and timing of DM onset, occurring even up to 8 months after cessation of ICP treatment. A systematic review of 90 cases, demonstrated a diagnosis of DM on average after 4.5 cycles of ICP (102). C-peptide levels were usually low or undetectable at diagnosis, islet autoantibodies were positive in 53%, with a predominance of glutamic acid decarboxylase antibodies, and susceptible HLA genotypes present in 65% (102). HbA1c levels were relatively low, consistent with the observed rapid onset of beta cell inflammation. Importantly, an elegant albeit small single-center study, used radiological and biochemical phenotyping to demonstrate that ICPi DM is irreversible (103). This has important clinical implications such that any individual diagnosed with ICPi-induced DM should be counselled around an expected life-long requirement of insulin.

 

Glucocorticoid (Steroid) Treatment

 

Glucocorticoids (GC) increase insulin resistance and glucose production and inhibit the production and secretion of insulin by pancreatic beta cells, as well as acting centrally to counteract the appetite-reducing effects of insulin (104). As such they are commonly associated with the development of hyperglycemia and diabetes. GCs have a direct hyperglycemic effect which starts very early after ingestion (105, 106). They typically cause an increase in blood glucose levels 4-8 hours after ingestion leading to a peak blood glucose level between midday meal and evening meal (106, 107). One in ten people not known to have diabetes develop GC-induced diabetes (108) an effect which is dose dependent (109). The incidence of glucocorticoid-induced hyperglycemia has been shown to occur in up to 30% of individuals without diabetes (110), but could be as high as 50%. The consequences of missing it can lead to significant harm, including the development of Hyperosmolar Hyperglycemic State (HHS), hospitalization, and in extreme circumstances, death. In a single center UK prevalence study 12.8% (120/940) of inpatients were found to be on glucocorticoids, however only 20.5% of these individuals (25/120) had their blood glucose levels measured during admission, demonstrating how infrequently glucose is measured in hospital (111). It is important to ensure that if glucocorticoid (steroid) induced hyperglycemia does occur, it is picked up early.

 

The use of GCs, is common in advanced cancer, to reduce peri-lesional edema, relieve pain, control nausea, combat fatigue, or boost appetite. For oncological emergencies such as cerebral metastases, superior vena-cava obstruction (SVCO), or metastatic spinal cord compression (MSCC), high dose GC treatment is integral to patient management. Furthermore, GC treatments are the backbone of many hematological cancer treatment regimens, and are often used as supportive anti-emetic medications, or to prevent allergic reactions, in many solid tumor regimens (105), and, as discussed above, the main first-line treatment for the management of ICP toxicity. In one study, the incidence of glucocorticoid-induced diabetes was 20% in those with newly diagnosed gastrointestinal cancer following at least 3 cycles of highly or moderately emetogenic chemotherapy, including dexamethasone as a supportive medication. Furthermore, almost 60% of people in the study exhibited signs of insulin resistance and multivariate analysis showed a significant association between the cumulative dose of dexamethasone and the incidence of corticosteroid-induced diabetes (112). In a separate smaller study of 16 women without diabetes with ovarian or endometrial cancer receiving carboplatin/paclitaxel chemotherapy with dexamethasone as supportive care, almost all experienced elevated interstitial glucose levels with diurnal variation during the first five days of treatment (113). For those who receive prednisolone as part of a treatment regimen for hematological malignancies, rates of steroid-induced diabetes and hyperglycemia have been reported to be as high as 32.5% and 47% respectively, highlighting the scale of this issue (114, 115).

 

Supra-physiological doses of glucocorticoids approximate to a dose of prednisolone greater than 5mg per day – or an equivalent dose of the alternative synthetic GC (Table 2). With increasing dose of GC, the risk of potential hyperglycemia increases, and in people without pre-existing diabetes, a glucocorticoid dose equivalent of >12mg dexamethasone and longer acting steroids are associated with a greater degree of hyperglycemia (116). As duration of GC treatment increases, it becomes increasingly likely that hyperglycemia may not resolve once the GCs are withdrawn, with those groups at particular risk of developing glucocorticoid induced diabetes, shown in Table 3.

 

Table 2. Glucocorticoid Dose Equivalent

Glucocorticoid (steroid)

Potency (equivalent doses)

Duration of action (half-life, in hours)

Hydrocortisone

20 mg

8

Prednisolone

5 mg

16-36

Methylprednisolone

4 mg

18-40

Dexamethasone

0.8 mg

36-54

Betamethasone

0.8 mg

26-54

 

Table 3. Risk Factors for Glucocorticoid-Inducted Diabetes

Pre-existing type 1 or type 2 diabetes

Family history of diabetes

Increasing age

Obesity

Ethnic minorities

Impaired fasting glucose or impaired glucose tolerance

Polycystic ovarian syndrome

Previous gestational diabetes

Previous development of hyperglycemia on glucocorticoid therapy

Concurrent cytotoxic therapy known to cause hyperglycemia

 

HYPOGYCEMIA IN PEOPLE ON SACT

 

Although anti-cancer therapies and glucocorticoid use lead predominantly to hyperglycemia, there are risks of hypoglycemia that require consideration. People at risk of hypoglycemia should be counselled on the signs and symptoms to be aware of, and of the requirement to inform the driver and vehicle licensing agency should they experience any episodes of hypoglycemia requiring third party assistance.

 

Poor oral intake and nausea/vomiting from the underlying cancer or treatments put individuals at increased risk of hypoglycemia. Poor glycemic control can cause weight loss and precipitate nutrition impact symptoms (NIS) such as nausea, poor appetite, and altered bowel movements, further increasing the risks of hypoglycemia, particularly when dietary intake has been poor for some time. People with diabetes on an insulin secretagogue (sulfonylureas or meglitinides), or those on insulin, are also at higher risk of hypoglycemia.

 

In patients with end-stage metastatic disease, and shortened life expectancy, tight glucose control is not indicated, potentially placing individuals at unnecessary risk for hypoglycemia, particularly in those with a poor performance status >2. Individual risk for hypoglycemia and prognosis should be considered and recommended glycemic measurement targets are between 6.0 mmol/L – 15 mmol/L (108 – 225 mg/dl) (117).

 

People with new onset ICPi-induced insulin deficiency often have labile glucose control (99). More relaxed glucose targets may be required to avoid hypoglycemia wherever possible. Immune checkpoint inhibitors can also induce hypopituitarism leading to secondary adrenal insufficiency. This may lead to hypoglycemia (together with any of the following - hyponatremia, hyperkalemia and hypotension). Adrenalitis leading to primary adrenal insufficiency is very rare. Presentation of adrenal insufficiency ranges from asymptomatic laboratory alterations to the acutely unwell, with management depending on the severity (118). Other causes of adrenal or pituitary deficiency leading to hypoglycemia include metastases at these sites, surgery, irradiation, azole class of anti-fungal medication, and inappropriate abrupt cessation of glucocorticoid medication.

 

In oncology patients being weaned from long-term steroids, glucose monitoring will need to be continued after glucocorticoid cessation, with doses of anti-diabetic treatments adjusted accordingly, and individuals advised on risks of hypoglycemia. Caution is also required whilst using certain hematological anti-cancer therapies, including lenalidomide (119) and bortezomib (120), which can precipitate hypoglycemia, particularly in people with an underlying diagnosis of diabetes.

 

All cancer patients at risk from hypoglycemia should receive advice regarding appropriate treatment with 15–20 g of fast-acting carbohydrate, taken immediately (121). Comprehensive guidelines from the Joint British Diabetes Societies for Inpatient Care on the management of hypoglycemia can be found at this reference (122).

 

MANAGEMENT RECOMMENDATIONS  

 

Despite the effects of hyper- and hypoglycemia in people with diabetes (PWD) and those without known diabetes in cancer, there is a sparsity of guidance on the specific management considerations of these individuals. To address this, collaborative guidelines have recently been produced by the UK Chemotherapy Board (UKCB) and Joint British Diabetes Society for Inpatient Care (JBDS) (123, 124). The scope of these guidelines are to provide advice for the oncology/hemato-oncology and diabetes multidisciplinary teams to manage people with diabetes, commencing anti-cancer/ steroid therapy, as well as identifying individuals without a known diagnosis of diabetes who are at risk of developing hyperglycemia and new onset diabetes. These guidelines are intended for the outpatient management of people with cancer, particularly in the setting of the oncology/hemato-oncology clinic, and provision of advice for individuals at home, but where necessary, may be applied to inpatients as well. Whilst covering these guidelines in detail is beyond the scope of this chapter, key management considerations are summarized in tables 4-9.

 

Table 4. At Baseline

·       HbA1c and venous plasma glucose should be checked in all people with cancer at baseline clinic visit

·       Provide high risk individuals with capillary blood glucose (CBG) meter and glucose testing strips, or if baseline plasma glucose is ≥12 mmol/L (216 mg/dl)

·       Individuals with raised baseline HbA1c (>47 mmol/mol [6.5%]) should be referred to primary care for management of hyperglycemia prior to next follow up visit

·       When initiating SACT/glucocorticoids individuals must be informed of the risk of developing hyperglycemia/diabetes and potential symptoms to expect

·       The recommended glucose target level is 6.0-10.0 mmol/L (108 – 180 mg/dl), allowing a range of 6.0-12.0 mmol/L (108 – 216 mg/dl)

·       There are differences in opinion at where the threshold for intervention should be drawn - 12.0 mmol/L (216 mg/dl) is a pragmatic threshold

 

Table 5. Commencing Glucocorticoids (GC) /Systemic Anti-Cancer Therapy

·       Check baseline HbA1c and random venous plasma glucose before starting therapy

·       Monitor random plasma glucose at each treatment visit

·       Educate patients in symptoms of hyperglycemia

·       Consider commencing gliclazide 40mg if raised blood glucose ≥12mmol/L (216 mg/dl) on two occasions

·       Gliclazide may require frequent and significant increases in dose to reduce glucose levels, particularly on high dose steroids

·       Inform diabetes care provider if persistently raised blood glucose

·       If blood glucose is ≥20mmol/L (360 mg/dl), rule out DKA/HHS

 

Table 6. Commencing Immune Checkpoint Inhibitors (ICP)

·       Educate patients to be aware of symptoms of hyperglycemia

·       Rule out DKA or HHS which often occurs precipitously

·       Withhold ICP if evidence of ICP-induced diabetes emergency. Once patient has been regulated with insulin substitution, consider restarting ICP

·       Almost all patients require insulin therapy – refer urgently to diabetes team

 

Table 7. Managing Nausea and Vomiting

·       People with diabetes should be made aware of likely exacerbation of hyperglycemia whilst on anti-emetic therapy

·       People with diabetes receiving emetogenic chemotherapy should be offered an NK1 antagonist (e.g., aprepitant) with a long acting 5HT3 inhibitor (e.g., ondansetron)

·       Consider the use of a GC in the first cycle and reduce doses or withdraw completely based on the PWD’s emetic control and on blood glucose management

 

Table 8. For Non-Insulin-Treated Individuals with Type 2 Diabetes

·       Check baseline HbA1c and random venous plasma glucose before starting therapy

·       Monitor random plasma glucose at each treatment visit

·       Educate patients in symptoms of hyperglycemia

·       If plasma glucose is ≥12 mmol/L (216 mg/dl) on two occasions, screen for symptoms of hyperglycemia and ketonuria/ketonemia

·       In individuals already on a sulphonyurea such as gliclazide or meglitinides, up-titrate morning dose of gliclazide to a maximum doses of 240 mg. Evening dose of gliclazide may be initiated to achieve a maximum daily dose of 320 mg

·       Insulin therapy may be required

·       In individuals on a diet-controlled regimen, or on other non-sulfonylurea treatments (e.g., metformin, DPP4 inhibitors, pioglitazone, SGLT2 inhibitors) commence gliclazide 40 mg, and up-titrate

 

Table 9. For Insulin-Treated Individuals with Type 2 Diabetes

·       Check baseline HbA1c and random venous plasma glucose before starting therapy

·       Monitor random plasma glucose at each treatment visit

·       If plasma glucose is ≥12 mmol/L (216 mg/dl) on two occasions, screen for symptoms of hyperglycemia and ketonuria/ketonemia

·       Contact usual diabetes team for support in titrating insulin

·       Consider titrating insulin by 10-20% of the original dose daily

·       Individuals should be made aware of ‘sick day rules’ with insulin administration

 

Full management guidelines can be found at the UK Chemotherapy Board (UKCB) and Joint British Diabetes Society for Inpatient Care (JBDS) websites.

 

ADDITIONAL MANAGEMENT CONSIDERATIONS: CHOICE OF DIABETES THERAPEUTIC AGENT

 

Special consideration should also be given to the non-glycemic effects of hypoglycemic agents, including specific side effects and the impact on weight. Although weight reduction is associated with improvement in glycemic and metabolic profile in people with type 2 diabetes and is a key consideration in the choice of therapy, significant weight loss would usually be an unwanted effect in the oncology population. Indeed, weight gain is often used as a metric of improving nutritional state, especially in cancer related cachexia. This also has implications when counselling people with cancer about dietary choices when there is an additional cancer diagnosis. It is imperative that personalized advice is offered by healthcare professionals considering the global impact on the individual of any dietary or even lifestyle advice. SGLT2 inhibitors and GLP-1 agonists, for their potential weight reduction effects, are therefore less attractive options in the oncology setting. Insulin and sulfonylureas, on the other hand, offer an anabolic effect and therefore may be more desirable. Gastrointestinal side effects are common among hypoglycemic agents including metformin, DPP4 inhibitors, and GLP-1 agonists, and have the potential to complicate issues with nausea, vomiting, and oral intake from the underlying cancer and its treatment. Similarly, poor oral intake and nephrotoxic effects of certain SACT, added to a potential osmotic diuretic effect of SGLT2

inhibitors, could also increase the risk of acute kidney injury. The associated risk of genital tract infections with SGLT2 inhibitors would also be an additional consideration especially within an immunocompromised population (125). The impact and significance of these non-glycemic effects in the oncology population clearly differ to that of the general population, therefore highlighting the importance of a personalized approach with regular review of patients’ diabetes treatment through their oncology journey. 

 

CONCLUSIONS

 

It is common practice in oncology to initiate systemic anti-cancer therapy (including chemotherapy, targeted treatment, immunotherapy and steroids) in people with pre-existing diabetes. Diabetes, or risk of developing diabetes are by no means a contraindication to treatment but treating clinicians should be aware of the risks to patients, and counsel them appropriately. As more sophisticated anti-cancer treatments become licensed for use, the metabolic effects of these treatments will become better understood, and oncology teams should utilize and collaborate with endocrinology and primary care services to minimize the risks to individuals from poor glycemic control and diabetes. The recent publication of specific guidelines should act as a reference aid for clinicians and wider healthcare professionals to aid in risk recognition, diagnostic and screening for treatment induced diabetes, and provide the tools to appropriately manage these individuals and reduce the risks of complications.

 

REFERENCES

 

  1. International Diabetes Federation. IDF Diabetes Atlas 2021. Available at https://diabetesatlas.org/atlas/tenth-edition/. Accessed Dec 2021.
  2. American Diabetes Association. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2021. Diabetes Care [Internet]. 2021; 44(Supplement 1): S15-S33.https://doi.org/10.2337/dc21-S002
  3. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2010;33 Suppl 1(Suppl 1):S62-9. doi:10.2337/dc10-S062
  4. Tsilidis KK, Kasimis JC, Lopez DS, Ntzani EE, Ioannidis JPA. Type 2 diabetes and cancer: umbrella review of meta-analyses of observational studies. BMJ : British Medical Journal. 2015;350:g7607. doi:10.1136/bmj.g7607
  5. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60(4):207-21. doi:10.3322/caac.20078
  6. Yan P, Wang Y, Yu X, Liu Y, Zhang ZJ. Type 2 diabetes mellitus and risk of head and neck cancer subtypes: a systematic review and meta-analysis of observational studies. Acta Diabetol. 2021;58(5):549-565. doi:10.1007/s00592-020-01643-0
  7. Suh S, Kim KW. Diabetes and Cancer: Cancer should be screened in routine diabetes assessment. Diabetes Metab J. 2019;43(6):733-743. doi:10.4093/dmj.2019.0177
  8. Diabetes UK. Diabetes and Cancer. https://wwwdiabetesorguk/diabetes-the-basics/related-conditions/diabetes-and-cancer. Accessed 28th February 2020
  9. Barone BB, Yeh HC, Snyder CF, et al. Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: a systematic review and meta-analysis. JAMA. 2008;300(23):2754-64. doi:10.1001/jama.2008.824
  10. Calderillo-Ruiz G, Lopez H, Herrera M, et al. P-149 - Obesity and hyperglycemia as a bad prognosis factor for recurrence and survival in colon cancer. Annals of Oncology. 2019;30:iv40-iv41. doi:https://doi.org/10.1093/annonc/mdz155.148
  11. Villarreal-Garza C, Shaw-Dulin R, Lara-Medina F, et al. Impact of diabetes and hyperglycemia on survival in advanced breast cancer patients. Exp Diabetes Res. 2012;2012:732027. doi:10.1155/2012/732027
  12. Hosokawa T, Kurosaki M, Tsuchiya K, et al. Hyperglycemia is a significant prognostic factor of hepatocellular carcinoma after curative therapy. World journal of gastroenterology. 2013;19(2):249-57. doi:10.3748/wjg.v19.i2.249
  13. Wright JL, Plymate SR, Porter MP, et al. Hyperglycemia and prostate cancer recurrence in men treated for localized prostate cancer. Prostate cancer and prostatic diseases. 2013;16(2):204-8. doi:10.1038/pcan.2013.5
  14. Barba M, Sperati F, Stranges S, et al. Fasting glucose and treatment outcome in breast and colorectal cancer patients treated with targeted agents: results from a historic cohort. Ann Oncol. 2012;23(7):1838-45. doi:10.1093/annonc/mdr540
  15. Chen S, Tao M, Zhao L, Zhang X. The association between diabetes/hyperglycemia and the prognosis of cervical cancer patients: A systematic review and meta-analysis. Medicine (Baltimore). 2017;96(40):e7981. doi:10.1097/md.0000000000007981
  16. Liu H, Liu Z, Jiang B, et al. Prognostic significance of hyperglycemia in patients with brain tumors: a meta-analysis. Mol Neurobiol. 2016;53(3):1654-1660. doi:10.1007/s12035-015-9115-4
  17. Ali NA, O'Brien Jr JM, Blum W, et al. Hyperglycemia in patients with acute myeloid leukemia is associated with increased hospital mortality. https://doi.org/10.1002/cncr.22777. Cancer. 2007/07/01 2007;110(1):96-102. doi:https://doi.org/10.1002/cncr.22777
  18. Barua R, Templeton AJ, Seruga B, Ocana A, Amir E, Ethier JL. Hyperglycaemia and survival in solid tumours: A systematic review and meta-analysis. Clin Oncol (R Coll Radiol). 2018;30(4):215-224. doi:10.1016/j.clon.2018.01.003
  19. Ma J, Wang J, Ge L, Long B, Zhang J. The impact of diabetes mellitus on clinical outcomes following chemotherapy for the patients with pancreatic cancer: a meta-analysis. Acta Diabetol. 2019;56(10):1103-1111. doi:10.1007/s00592-019-01337-2
  20. Peairs KS, Barone BB, Snyder CF, et al. Diabetes mellitus and breast cancer outcomes: a systematic review and meta-analysis. J Clin Oncol. 2011;29(1):40-6. doi:10.1200/jco.2009.27.3011
  21. Ramos-Garcia P, Roca-Rodriguez MDM, Aguilar-Diosdado M, Gonzalez-Moles MA. Diabetes mellitus and oral cancer/oral potentially malignant disorders: A systematic review and meta-analysis. Oral Dis. 2021;27(3):404-421. doi:10.1111/odi.13289
  22. Gerards MC, van der Velden DL, Baars JW, et al. Impact of hyperglycemia on the efficacy of chemotherapy—A systematic review of preclinical studies. Critical reviews in oncology/hematology. 2017;113:235-241. doi:https://doi.org/10.1016/j.critrevonc.2017.03.007
  23. Garufi A, Traversi G, Gilardini Montani MS, et al. Reduced chemotherapeutic sensitivity in high glucose condition: implication of antioxidant response. Oncotarget. 2019;10(45):4691-4702. doi:10.18632/oncotarget.27087
  24. Stebbing J, Sharma A, North B, et al. A metabolic phenotyping approach to understanding relationships between metabolic syndrome and breast tumour responses to chemotherapy. Ann Oncol. 2012;23(4):860-6. doi:10.1093/annonc/mdr347
  25. Hershey DS, Given B, Given C, Von Eye A, You M. Diabetes and cancer: impact on health-related quality of life. Oncol Nurs Forum. 2012;39(5):449-57. doi:10.1188/12.Onf.449-457
  26. Hershey DS, Tipton J, Given B, Davis E. Perceived impact of cancer treatment on diabetes self-management. Diabetes Educ. 2012;38(6):779-90. doi:10.1177/0145721712458835
  27. Pettit S, Cresta E, Winkley K, Purssell E, Armes J. Glycaemic control in people with type 2 diabetes mellitus during and after cancer treatment: A systematic review and meta-analysis. PLoS One. 2017;12(5):e0176941. doi:10.1371/journal.pone.0176941
  28. Hershey DS. Importance of glycemic control in cancer patients with diabetes: treatment through end of life. Asia-Pacific journal of oncology nursing. 2017;4(4):313-318. doi:10.4103/apjon.apjon_40_17
  29. Vissers PAJ, Falzon L, van de Poll-Franse LV, Pouwer F, Thong MSY. The impact of having both cancer and diabetes on patient-reported outcomes: a systematic review and directions for future research. J Cancer Surviv. 2016;10(2):406-415. doi:10.1007/s11764-015-0486-3
  30. Alenzi EO, Kelley GA. The association of hyperglycemia and diabetes mellitus and the risk of chemotherapy-induced neutropenia among cancer patients: A systematic review with meta-analysis. J Diabetes Complications. Jan 2017;31(1):267-272. doi:10.1016/j.jdiacomp.2016.09.006
  31. Chao C, Page JH, Yang SJ, Rodriguez R, Huynh J, Chia VM. History of chronic comorbidity and risk of chemotherapy-induced febrile neutropenia in cancer patients not receiving G-CSF prophylaxis. Annals of Oncology. 2014;25(9):1821-1829. doi:10.1093/annonc/mdu203
  32. Yang IP, Miao ZF, Huang CW, et al. High blood sugar levels but not diabetes mellitus significantly enhance oxaliplatin chemoresistance in patients with stage III colorectal cancer receiving adjuvant FOLFOX6 chemotherapy. Ther Adv Med Oncol. 2019;11:1758835919866964. doi:10.1177/1758835919866964
  33. Liu X, Ji J, Sundquist K, Sundquist J, Hemminki K. The impact of type 2 diabetes mellitus on cancer-specific survival: a follow-up study in Sweden. Cancer. 2012;118(5):1353-61. doi:10.1002/cncr.26420
  34. Park JH, Kim H-Y, Lee H, Yun EK. A retrospective analysis to identify the factors affecting infection in patients undergoing chemotherapy. European Journal of Oncology Nursing. 2015;19(6):597-603. doi:https://doi.org/10.1016/j.ejon.2015.03.006
  35. Brunello A, Kapoor R, Extermann M. Hyperglycemia during chemotherapy for hematologic and solid tumors is correlated with increased toxicity. Am J Clin Oncol. 2011;34(3):292-6. doi:10.1097/COC.0b013e3181e1d0c0
  36. Attili V, Bapsy P, Dadhich HK, Batra U, Lokanatha D, Babu KG. Impact of diabetes on cancer chemotherapy outcome: A retrospective analysis. International Journal of Diabetes in Developing Countries. 2007;27(4)122-128
  37. de la Morena Barrio P, Conesa M, González-Billalabeitia E, et al. Delayed recovery and increased severity of Paclitaxel-induced peripheral neuropathy in patients with diabetes. J Natl Compr Canc Netw. 2015;13(4):417-23. doi:10.6004/jnccn.2015.0057
  38. Kim S, Garg A, Raja F. Cancer patients with undiagnosed and poorly managed diabetes mellitus. Journal of Clinical Oncology. 2017;35(15_suppl):e18232-e18232. doi:10.1200/JCO.2017.35.15_suppl.e18232
  39. Hamilton A, Todd A, Wallace H, et al. Improving identification and management of steroid-induced hyperglycaemia in day-case chemotherapy patients: Results from a quality improvement project in a regional oncology centre. 2018. (Abstract) Diabetic Medicine 2018;35 (Suppl 1)
  40. Zylla D, Gilmore G, Eklund J, Richter S, Carlson A. Impact of diabetes and hyperglycemia on health care utilization, infection risk, and survival in patients with cancer receiving glucocorticoids with chemotherapy. J Diabetes Complications. 2019;33(4):335-339. doi:10.1016/j.jdiacomp.2018.12.012
  41. Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: A meta-analysis. Acta Oncol. 2016;55(9-10):1196-1203. doi:10.3109/0284186x.2016.1168939
  42. Verges B, Walter T, Cariou B. Endocrine side effects of anti-cancer drugs: effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol. 2014;170(2):R43-55. doi:10.1530/EJE-13-0586
  43. André F, Ciruelos E, Rubovszky G, et al. Alpelisib for PIK3CA-mutated, hormone receptor–positive advanced breast cancer. New England Journal of Medicine. 2019;380(20):1929-1940. doi:10.1056/NEJMoa1813904
  44. Shariff AI, Syed S, Shelby RA, et al. Novel cancer therapies and their association with diabetes. 2019;62(2):R187. doi:10.1530/jme-18-0002
  45. Sullivan I, Planchard D. Osimertinib in the treatment of patients with epidermal growth factor receptor T790M mutation-positive metastatic non-small cell lung cancer: clinical trial evidence and experience. Therapeutic Advances in Respiratory Disease. 2016;10(6):549-565. doi:10.1177/1753465816670498
  46. Wadlow RC, Hezel AF, Abrams TA, et al. Panitumumab in patients with KRAS wild-type colorectal cancer after progression on cetuximab. Oncologist. 2012;17(1):14. doi:10.1634/theoncologist.2011-0452
  47. Joint Formulary Committee. British National Formulary. Available at: https://bnf.nice.org.uk/drug/panitumumabhtml. 2020;Accessed 3rd March 2020
  48. Armstrong AJ, Halabi S, Eisen T, et al. Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (ASPEN): a multicentre, open-label, randomised phase 2 trial. The Lancet Oncology. 2016;17(3):378-388. doi:10.1016/S1470-2045(15)00515-X
  49. Agostino NM, Chinchilli VM, Lynch CJ, et al. Effect of the tyrosine kinase inhibitors (sunitinib, sorafenib, dasatinib, and imatinib) on blood glucose levels in diabetic and nondiabetic patients in general clinical practice. J Oncol Pharm Pract. 2011;17(3):197-202. doi:10.1177/1078155210378913
  50. Chang L, An Y, Yang S, Zhang X. Meta-analysis of therapeutic effects and the risks of hypertension and hyperglycemia in patients with renal cell carcinoma who were receiving antiangiogenic drugs. J Cancer Res Ther. 2016;12(Supplement):96-103. doi:10.4103/0973-1482.191614
  51. Saglio G, Larson RA, Hughes TP, et al. Efficacy and Safety of Nilotinib In Chronic Phase (CP) Chronic Myeloid Leukemia (CML) Patients (Pts) with Type 2 Diabetes In the ENESTnd Trial. Blood. 2010;116(21):3430-3430. doi:10.1182/blood.V116.21.3430.3430
  52. Chan O, Talati C, Isenalumhe L, et al. Side-effects profile and outcomes of ponatinib in the treatment of chronic myeloid leukemia. Blood advances. 2020;4(3):530-538. doi:10.1182/bloodadvances.2019000268
  53. Villadolid J, Ersek JL, Fong MK, Sirianno L, Story ES. Management of hyperglycemia from epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) targeting T790M-mediated resistance. Transl Lung Cancer Res. 2015;4(5):576-583. doi:10.3978/j.issn.2218-6751.2015.10.01
  54. Schlenk RF, Weber D, Fiedler W, et al. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood. 2019;133(8):840-851. doi:10.1182/blood-2018-08-869453
  55. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. New England Journal of Medicine. 2017;377(5):454-464. doi:10.1056/NEJMoa1614359
  56. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. New England Journal of Medicine. 2019;381(18):1728-1740. doi:10.1056/NEJMoa1902688
  57. US FDA. Mylotarg (gemtuzumab ozogamicin for injection). Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2006/021174s020lblpdf. Accessed January 2021;
  58. Wolin EM. The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointest Cancer Res. 2012;5(5):161-168.
  59. Yang J, Jia B, Qiao Y, Chen W, Qi X. Variations of blood glucose in cancer patients during chemotherapy. Original Article. Nigerian Journal of Clinical Practice. 2016 2016;19(6):704-708. doi:10.4103/1119-3077.187323
  60. Yang J, Jia B, Yan J, He J. Glycaemic adverse drug reactions from anti-neoplastics used in treating pancreatic cancer. Niger J Clin Pract. 2017;20(11):1422-1427. doi:10.4103/njcp.njcp_444_16
  61. Paz-Ares L, de Marinis F, Dediu M, et al. Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non-squamous non-small-cell lung cancer (PARAMOUNT): a double-blind, phase 3, randomised controlled trial. Lancet Oncol. 2012;13(3):247-55. doi:10.1016/s1470-2045(12)70063-3
  62. Clinical Trials. Pemetrexed and best supportive care versus placebo and best supportive care in non-small cell lung cancer (NSCLC). Available at: https://clinicaltrialsgov/ct2/show/results/NCT00102804. 2014;Accessed 3rd March 2020
  63. Ma YY, Zhao M, Liu Y, et al. Use of decitabine for patients with refractory or relapsed acute myeloid leukemia: a systematic review and meta-analysis. Hematology. 2019;24(1):507-515. doi:10.1080/16078454.2019.1632407
  64. Electronic Medicines Compendium (emc). Busulfan concentrate for infusion. Available at : https://www.medicines.org.uk/emc/product/2160/smpc#gref. Accessed January 2021;
  65. Huang CY, Lin YS, Liu YH, Lin SC, Kang BH. Hyperglycemia crisis in head and neck cancer patients with platinum-based chemotherapy. J Chin Med Assoc. 2018;81(12):1060-1064. doi:10.1016/j.jcma.2018.05.008
  66. Nan DN, Fernandez-Ayala M, Vega Villegas ME, et al. Diabetes mellitus following cisplatin treatment. Acta Oncol. 2003;42(1):75-8. doi:10.1080/0891060310002276
  67. Yang J, Wang Y, Liu K, Yang W, Zhang J. Risk factors for doxorubicin-induced serious hyperglycaemia-related adverse drug reactions. Diabetes Ther. 2019;10(5):1949-1957. doi:10.1007/s13300-019-00677-0
  68. Kritharis A, Bradley TP, Budman DR. Association of diabetes mellitus with arsenic trioxide (ATO) evaluated in the treatment of acute promyelocytic leukemia (APL). Journal of Clinical Oncology. 2011;29(15_suppl):e19724-e19724. doi:10.1200/jco.2011.29.15_suppl.e19724
  69. Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Annals of Oncology. 2017;28:iv119-iv142. doi:https://doi.org/10.1093/annonc/mdx225
  70. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Research & Care. 2019;7(1):e000591. doi:10.1136/bmjdrc-2018-000591
  71. Lu J, Yang J, Liang Y, Meng H, Zhao J, Zhang X. Incidence of immune checkpoint inhibitor-associated diabetes: A meta-analysis of randomized controlled studies. Systematic Review. Frontiers in Pharmacology. 2019;10(1453)doi:10.3389/fphar.2019.01453
  72. Wang H, Sun X, Zhao L, Chen X, Zhao J. Androgen deprivation therapy is associated with diabetes: Evidence from meta-analysis. J Diabetes Investig. 2016;7(4):629-636. doi:10.1111/jdi.12472
  73. Lipscombe LL, Fischer HD, Yun L, et al. Association between tamoxifen treatment and diabetes: a population-based study. Cancer. 2012;118(10):2615-22. doi:10.1002/cncr.26559
  74. Hwangbo Y, Lee EK. Acute hyperglycemia associated with anti-cancer medication. Endocrinol Metab (Seoul). 2017;32(1):23-29. doi:10.3803/EnM.2017.32.1.23
  75. Feng JP, Yuan XL, Li M, et al. Secondary diabetes associated with 5-fluorouracil-based chemotherapy regimens in non-diabetic patients with colorectal cancer: results from a single-centre cohort study. Colorectal Dis. 2013;15(1):27-33. doi:10.1111/j.1463-1318.2012.03097.x
  76. Teng CJ, Chang KH, Tsai IJ, Hwang WL, Hsu CY, Sheu WH. Impact of anthracyclines on diabetes mellitus development in B-cell lymphoma patients: A nationwide population-based study. Clin Drug Investig. 2018;38(7):603-610. doi:10.1007/s40261-018-0645-1
  77. Koltin D, Sung L, Naqvi A, Urbach SL. Medication induced diabetes during induction in pediatric acute lymphoblastic leukemia: prevalence, risk factors and characteristics. Support Care Cancer. 2012;20(9):2009-15. doi:10.1007/s00520-011-1307-5
  78. van Niekerk G, Engelbrecht AM. Inflammation-induced metabolic derangements or adaptation: An immunometabolic perspective. Cytokine Growth Factor Rev. 2018;43:47-53. doi:10.1016/j.cytogfr.2018.06.003
  79. de Lima Junior EA, Yamashita AS, Pimentel GD, et al. Doxorubicin caused severe hyperglycaemia and insulin resistance, mediated by inhibition in AMPk signalling in skeletal muscle. J Cachexia Sarcopenia Muscle. 2016;7(5):615-625. doi:10.1002/jcsm.12104
  80. Giovannucci E. The critical need for guidance in managing glycaemic control in patients with cancer. https://doi.org/10.1111/dme.14624. Diabetic Medicine. 2021;e14624. doi:https://doi.org/10.1111/dme.14624
  81. Buffier P, Bouillet B, Smati S, Archambeaud F, Cariou B, Verges B. Expert opinion on the metabolic complications of new anticancer therapies: Tyrosine kinase inhibitors. Ann Endocrinol (Paris). 2018;79(5):574-582. doi:10.1016/j.ando.2018.07.011
  82. Romero-Ventosa EY, Otero-Millán L, González-Costas S, et al. Effect of tyrosine kinase inhibitors on the glucose levels in diabetic and nondiabetic patients. Indian journal of cancer. 2017;54(1):136-143. doi:10.4103/ijc.IJC_190_17
  83. Sakuma I, Nagano H, Yoshino I, Yokote K, Tanaka T. Ceritinib aggravates glycemic control in insulin-treated patients with diabetes and metastatic ALK-positive lung cancer. Intern Med. 2019;58(6):817-820. doi:10.2169/internalmedicine.1870-18
  84. Bouillet B, Buffier P, Smati S, Archambeaud F, Cariou B, Vergès B. Expert opinion on the metabolic complications of mTOR inhibitors. Ann Endocrinol (Paris). 2018;79(5):583-590. doi:10.1016/j.ando.2018.07.010
  85. Vergès B. mTOR and cardiovascular diseases: diabetes mellitus. Transplantation. 2018;102(2S Suppl 1):S47-s49. doi:10.1097/tp.0000000000001722
  86. Martel S, Bruzzone M, Ceppi M, et al. Risk of adverse events with the addition of targeted agents to endocrine therapy in patients with hormone receptor-positive metastatic breast cancer: A systematic review and meta-analysis. Cancer treatment reviews. 2018;62:123-132. doi:10.1016/j.ctrv.2017.09.009
  87. Sivendran S, Agarwal N, Gartrell B, et al. Metabolic complications with the use of mTOR inhibitors for cancer therapy. Cancer treatment reviews. 2014;40(1):190-6. doi:10.1016/j.ctrv.2013.04.005
  88. Acharya GK, Hita AG, Yeung SJ, Yeung SJ. Diabetic ketoacidosis and acute pancreatitis: serious adverse effects of everolimus. Ann Emerg Med. 2017;69(5):666-667. doi:10.1016/j.annemergmed.2017.01.002
  89. Bono P, Oudard S, Bodrogi I, et al. Outcomes in patients with metastatic renal cell carcinoma who develop everolimus-related hyperglycemia and hypercholesterolemia: combined subgroup analyses of the RECORD-1 and REACT Trials. Clin Genitourin Cancer. 2016;14(5):406-414. doi:10.1016/j.clgc.2016.04.011
  90. Keating NL, O'Malley AJ, Freedland SJ, Smith MR. Diabetes and cardiovascular disease during androgen deprivation therapy: observational study of veterans with prostate cancer. J Natl Cancer Inst. 2010;102(1):39-46. doi:10.1093/jnci/djp404
  91. Ng HS, Koczwara B, Roder D, Vitry A. Development of comorbidities in men with prostate cancer treated with androgen deprivation therapy: an Australian population-based cohort study. Prostate cancer and prostatic diseases. 2018;21(3):403-410. doi:10.1038/s41391-018-0036-y
  92. Santorelli ML, Hirshfield KM, Steinberg MB, Rhoads GG, Lin Y, Demissie K. Hormonal therapy for breast cancer and diabetes incidence among postmenopausal women. Ann Epidemiol. 2016;26(6):436-40. doi:10.1016/j.annepidem.2016.04.004
  93. Sun LM, Chen HJ, Liang JA, Li TC, Kao CH. Association of tamoxifen use and increased diabetes among Asian women diagnosed with breast cancer. Br J Cancer. 2014;111(9):1836-42. doi:10.1038/bjc.2014.488
  94. Quandt Z, Young A, Anderson M. Immune checkpoint inhibitor diabetes mellitus: a novel form of autoimmune diabetes. https://doi.org/10.1111/cei.13424. Clinical & Experimental Immunology. 2020;200(2):131-140. doi:https://doi.org/10.1111/cei.13424
  95. Falcone M, Fousteri G. Role of the PD-1/PD-L1 dyad in the maintenance of pancreatic immune tolerance for prevention of type 1 diabetes. Review. Frontiers in Endocrinology. 2020;11(569)doi:10.3389/fendo.2020.00569
  96. Clotman K, Janssens K, Specenier P, Weets I, De Block CEM. Programmed cell death-1 inhibitor–induced type 1 diabetes mellitus. The Journal of Clinical Endocrinology & Metabolism. 2018;103(9):3144-3154. doi:10.1210/jc.2018-00728
  97. Stamatouli AM, Quandt Z, Perdigoto AL, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018;67(8):1471-1480. doi:10.2337/dbi18-0002
  98. Akturk HK, Kahramangil D, Sarwal A, Hoffecker L, Murad MH, Michels AW. Immune checkpoint inhibitor-induced Type 1 diabetes: a systematic review and meta-analysis. https://doi.org/10.1111/dme.14050. Diabetic Medicine. 2019;36(9):1075-1081. doi:https://doi.org/10.1111/dme.14050
  99. Perdigoto AL, Quandt Z, Anderson M, Herold KC. Checkpoint inhibitor-induced insulin-dependent diabetes: an emerging syndrome. The Lancet Diabetes & Endocrinology. 2019;7(6):421-423. doi:10.1016/S2213-8587(19)30072-5
  100. Kyriacou A, Melson E, Chen W, Kempegowda P. Is immune checkpoint inhibitor-associated diabetes the same as fulminant type 1 diabetes mellitus? Clinical Medicine. 2020;20(4):417-423. doi:10.7861/clinmed.2020-0054
  101. Wright JJ, Salem J-E, Johnson DB, et al. Increased reporting of immune checkpoint inhibitor–associated diabetes. Diabetes Care. 2018;41(12):e150. doi:10.2337/dc18-1465
  102. de Filette JMK, Pen JJ, Decoster L, et al. Immune checkpoint inhibitors and type 1 diabetes mellitus: a case report and systematic review. Eur J Endocrinol. Sep 2019;181(3):363-374. doi:10.1530/eje-19-0291
  103. Byun DJ, Braunstein R, Flynn J, et al. Immune checkpoint inhibitor-associated diabetes: A single-institution experience. Diabetes Care. 2020;43(12):3106-3109. doi:10.2337/dc20-0609
  104. Perez A, Jansen-Chaparro S, Saigi I, Bernal-Lopez MR, Miñambres I, Gomez-Huelgas R. Glucocorticoid-induced hyperglycemia. J Diabetes. 2014;6(1):9-20. doi:10.1111/1753-0407.12090
  105. Dunning T, Martin P. Palliative and end of life care of people with diabetes: Issues, challenges and strategies. Diabetes Res Clin Pract. 2018;143:454-463. doi:10.1016/j.diabres.2017.09.018
  106. Joint British Diabetes Society (JBDS) for Inpatient Care Group. Steroid use for inpatients with diabetes. Available at: https://abcdcare/resource/steroid-use-inpatients-diabetes. May 202
  107. Tamez-Pérez HE, Quintanilla-Flores DL, Rodríguez-Gutiérrez R, González-González JG, Tamez-Peña AL. Steroid hyperglycemia: Prevalence, early detection and therapeutic recommendations: A narrative review. World J Diabetes. 2015;6(8):1073-1081. doi:10.4239/wjd.v6.i8.1073
  108. Pilkey J, Streeter L, Beel A, Hiebert T, Li X. Corticosteroid-induced diabetes in palliative care. J Palliat Med. 2012;15(6):681-9. doi:10.1089/jpm.2011.0513
  109. Gannon C, Dando N. Dose-sensitive steroid-induced hyperglycaemia. Palliat Med. 2010;24(7):737-9. doi:10.1177/0269216310377816
  110. Liu XX, Zhu XM, Miao Q, Ye HY, Zhang ZY, Li YM. Hyperglycemia induced by glucocorticoids in nondiabetic patients: a meta-analysis. Ann Nutr Metab. 2014;65(4):324-32. doi:10.1159/000365892
  111. Narwani V, Swafe L, Stavraka C, Dhatariya K. How frequently are bedside glucose levels measured in hospital inpatients on glucocorticoid treatment? Clin Med (Lond). 2014;14(3):327-328. doi:10.7861/clinmedicine.14-3-326a
  112. Jeong Y, Han HS, Lee HD, et al. A pilot study evaluating steroid-induced diabetes after antiemetic dexamethasone therapy in chemotherapy-treated cancer patients. Cancer Res Treat. 2016;48(4):1429-1437. doi:10.4143/crt.2015.464
  113. Lyall MJ, Thethy I, Steven L, et al. Diurnal profile of interstitial glucose following dexamethasone prophylaxis for chemotherapy treatment of gynaecological cancer. Diabet Med. 2018;35(11):1508-1514. doi:10.1111/dme.13770
  114. Lamar ZS, Dothard A, Kennedy L, et al. Hyperglycemia during first-line R-CHOP or dose adjusted R-EPOCH chemotherapy for non-Hodgkin lymphoma is prevalent and associated with chemotherapy alteration - a retrospective study. Leuk Lymphoma. 2018;59(8):1871-1877. doi:10.1080/10428194.2017.1410889
  115. Lee S-y, Kurita N, Yokoyama Y, et al. Glucocorticoid-induced diabetes mellitus in patients with lymphoma treated with CHOP chemotherapy. Supportive Care in Cancer. 2014;22(5):1385-1390. doi:10.1007/s00520-013-2097-8
  116. Healy SJ, Nagaraja HN, Alwan D, Dungan KM. Prevalence, predictors, and outcomes of steroid-induced hyperglycemia in hospitalized patients with hematologic malignancies. Endocrine. 2017;56(1):90-97. doi:10.1007/s12020-016-1220-2
  117. Roberts A, James J, Dhatariya K. Management of hyperglycaemia and steroid (glucocorticoid) therapy: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabet Med. 2018;35(8):1011-1017. doi:10.1111/dme.13675
  118. Higham CE, Olsson-Brown A, Carroll P, et al. Society for Endocrinology emergency guidance: Acute management of the endocrine complications of checkpoint inhibitor therapy. Endocrine connections. 2018;7(7):G1-G7. doi:10.1530/EC-18-0068
  119. Przybylski DJ, Birhiray R, Reeves DJ. Severe Hypoglycemia Caused by Lenalidomide. Pharmacotherapy. 2018;38(1):e1-e6. doi:10.1002/phar.2061
  120. Electronic Medicines Compendium (emc). Bortezomib. Available from: https://wwwmedicinesorguk/emc/product/10201/smpc#gref. Accessed September 2021;
  121. Joint British Diabetes Society (JBDS) for Inpatient Care Group. The Hospital Management of Hypoglycaemia in Adults with Diabetes Mellitus 3rd edition.

         Available from: https://abcdcare/joint-british-diabetes-societies-jbds-inpatient-care-group. 2018;

  1. Joint British Diabetes Society for Inpatient Care. The Hospital Management of Hypoglycaemia in Adults with Diabetes Mellitus 4th Edition. Available at: https://abcdcare/sites/abcdcare/files/site_uploads/JBDS_HypoGuideline_4th_edition_FINALpdf. 2020;
  2. Joharatnam-Hogan N, Chambers P, Dhatariya K, Board R. A guideline for the outpatient management of glycaemic control in people with cancer. Diabet Med. Jul 9 2021:e14636. doi:10.1111/dme.14636
  3. The UK Chemotherapy Board (UKCB) and Joint British Diabetes Society (JBDS). Management of Glycaemic Control in Patients with Cancer. Available from: https://wwwukchemotherapyboardorg/publications. Accessed September 2021;
  4. Liu J, Li L, Li S, et al. Effects of SGLT2 inhibitors on UTIs and genital infections in type 2 diabetes mellitus: a systematic review and meta-analysis. Sci Rep. Jun 6 2017;7(1):2824. doi:10.1038/s41598-017-02733-w

Clinical Management of Dyslipidemia in Youth with Chronic Kidney Disease

ABSTRACT

 

Chronic kidney disease (CKD) is commonly associated with abnormal lipid metabolism which may contribute to the morbidity and premature mortality associated with impaired renal function.  Dyslipidemia often occurs in the early phases and becomes progressively worse with disease severity and progression to end stage renal disease (ESRD). In this review, we discuss the clinical features, diagnosis, and management of dyslipidemia in children with renal disease, focusing primarily on nephrotic syndrome (NS) and ESRD.  There are limited data on treatment of dyslipidemia, outcomes, and prevention of CVD in youth with these conditions to help inform clinical decision-making and define best practices. 

 

INTRODUCTION

 

Chronic kidney disease (CKD) is characterized by a progressive decrease in renal function, divided into five stages (Table 1).  The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) defines CKD as a glomerular filtration rate (GFR) persistently < 60 mL/min (1). Further decline in kidney function results in ESRD, with permanent and complete loss of renal function, necessitating either dialysis or renal transplantation.  Due to a wide variety of common causes, including diabetes and hypertension, the prevalence of CKD is increasing (2).  In individuals with CKD, CVD is the leading cause of death and dyslipidemia is recognized as a major risk factor (3).  Mortality in up to half of individuals with CKD is the result of CVD (4).

 

Table 1. CKD Stages

Stage

GFR

1

> 90

mL/min

2

60-90

mL/min

3A

45-59

mL/min

3B

30-44

mL/min

4

15-29

mL/min

5

< 15

mL/min

Modified from Table 1 Hager et al. (1).

 

Nephrotic Syndrome (NS), clinically characterized by proteinuria, hypoalbuminemia, and edema, is one of the most common kidney diseases affecting youth. Injury to podocytes and glomeruli in NS is well described. Complications include acute injury of the kidney, systemic infection, and thromboembolism. Dyslipidemia is common and corresponds to the severity of proteinuria with or without CKD. In adults, risk of fatal and non-fatal MI is based upon the degree of proteinuria and the GFR. Despite the presence of dyslipidemia similar to that in adults, youth with CKD are often undertreated. This, in part, may reflect a lack of data regarding CVD risk in this population (5).

 

In addition to traditional risk factors, CVD disease in youth with ESRD appears to be directly related to the effects of renal impairment, exacerbated, in part, by medications necessary for treatment. Because mono and polygenic disorders affecting lipid and lipoprotein metabolism are common, some youth may also have an underlying predisposition to atherosclerosis in addition to the risk associated with renal failure – the impact of these on the risk of coronary artery disease and of congestive heart failure may well be substantially larger among younger patients (6,7).

 

Early onset of risk factors in children with kidney disease provides a longer period of exposure, significantly increasing risk of premature CVD. Although unproven, it is likely that optimum management of risk factors, such as elevated cholesterol and blood pressure, especially when implemented at an early age, may result in substantial reduction in subsequent incidence of CVD-related events. It is important, therefore, that youth with ESRD undergo global risk factor assessment and that all risk factors be optimally managed.  Based on randomized trials, data suggests that reduction of LDL-C concentrations may decrease the risk of CVD among individuals with renal failure who have average (or even below average) LDL-C concentrations (8-11).

 

ETIOLOGY AND PATHOGENESIS OF DYSLIPIDEMIA IN KIDNEY DISEASE

 

The pattern of dyslipidemia seen in CKD is typically characterized by hypertriglyceridemia (HTG), decreased HDL-C, variable changes in LDL-C, increase in non-HDL-C, and an increase in small dense LDL-C (12), as well as an increase in the apoB to apoA-I ratio. Elevations in Lp (a) are also common in CKD.  However, elevations in Lp (a) generally occur in subgroups of individuals who express larger Lp (a) isoforms (13).  HTG is present in early stages of renal disease and its origin is multifactorial, including impaired catabolism of VLDL and chylomicrons secondary to decreased lipoprotein lipase (LPL) activity. With the onset of uremia, inhibitors of LPL are increased, including apoC-III and pre-beta-HDL. A decrease in lecithin:cholesterol acyltransferase (LCAT), important for the maturation of HDL, and reduced expression of the apoA-I gene APOA1, the main apoprotein of HDL, have also been reported. These changes in gene expression and protein availability lead to alterations in two key HDL functions: 1) reverse cholesterol transport; and 2) anti-oxidation (1).

 

Individuals with NS often have elevated triglycerides (TG) and other atherogenic apoB-containing lipoproteins, including VLDL, IDL, LDL and lipoprotein(a). A decrease in oncotic pressure may be contributory to increased production of lipoproteins by stimulating the synthesis of apoB.  However, the mechanisms are not well understood. HDL-C levels are similar to those of healthy individuals.  Despite normal levels, however, it is likely the efficiency of the HDL-related reverse cholesterol transport in NS is decreased. The elevation in TG is likely due to decreased LPL activity. There is also downregulation of glycosylphosphatidylinositol-anchored HDL-binding protein 1 (GPIHBP1), which serves to anchor LPL to heparin sulfate proteoglycans on endothelial cells. The increased levels of LDL-C seen in NS is believed to be the result of increased production through increased acyl-CoA cholesterol acyltransferase (ACAT) and HMG-CoA reductase activity and decreased clearance through decreased LDL-receptor activity. Increased activity of proprotein convertase subtilisin/kexin type 9 (PCSK9) has also been reported, leading to a decrease in the number of LDL receptors and a reduction in hepatic uptake (5).

 

Table 2. Lipid Patterns in Kidney Disease

Lipid / Lipoprotein

CKD 1-5

Nephrotic Syndrome

Hemodialysis

Peritoneal Dialysis

Total Cholesterol

Progressive increase

↑↑

↔ ↓

TG

Progressive increase

↑↑

HDL-C

LDL-C

Progressive increase

↑↑

↔ ↓

Non-HDL cholesterol

Progressive increase

↑↑

↔ ↓

Lipoprotein (a)

Progressive increase

↑↑

↑↑

Modified from Mikolasevic et al (14)

 

CLINICAL FEATURES

 

Nephrotic Syndrome

 

Elevated fractions of apoB-containing particles in NS can increase the risk for formation of atherosclerotic plaque, leading to CVD-related events, such as MI and stroke, and may contribute to the increased risk for thrombosis and other adverse events (Table 3).  Progressive loss of renal function and development of CKD further increases the risk of morbidity (5).

 

Dyslipidemia may also contribute to glomerulosclerosis (15), further damaging the kidney (i.e., the lipid nephrotoxicity hypothesis). Excess accumulation of lipids, particularly in the interstitium and glomeruli (16), is accompanied by a pronounced inflammatory response, which appears to injure glomerular podocytes and mesangial cells. Such changes contribute to renal injury and impaired function. 

 

Table 3. Nephrotic Syndrome and CKD. Estimates of Clinical Consequences of Dyslipidemia

 

NS

CKD

NS and CKD

CVD

 

 

 

·       Atherosclerosis

++

+

+++

·       Myocardial infarction

++

+

+++

·       Stroke

++

+

+++

Progressive Kidney Disease

 

 

 

·       Glomerulosclerosis

++

+

+++

·       Mesangial Proliferation

+

+/-

+

·       Podocyte injury

+

+

++

·       Tubuloinsterstitial disease

++

+

+++

·       Proximal tubular cell injury

+

+

++

NS=nephrotic syndrome; CKD=chronic kidney disease. Adapted from Table 2 Agrawal, et al. (5)

 

Chronic Kidney Disease  

 

Dyslipidemia in CKD is characterized by increased serum levels of TG, decreased HDL-C, variable levels of LDL-C and an increase in apoB to apoA-I ratio. HTG is often present even in the early stages of CKD and is one of the most common lipid abnormalities encountered in this population. A large cross-sectional analysis of 391 children ((236 male, 154 female aged 1-16 years (median age 12 years), 71% Caucasian) with moderate CKD (median GFR 43 mL/min/1.73m2) enrolled in the Chronic Kidney Disease in Children (CKiD) Study noted 32% of children with HTG, 21% low HDL-C and 16% elevated non-HDL-C (17). Overall, 45% children with CKD had dyslipidemia, and of those 179 children, 45% had two or more lipid abnormalities.

 

There was a higher prevalence of HTG in children with nephrotic-range proteinuria (61%), as compared to 21%, 30%, and 24% in children with normal, mild, and moderate proteinuria, respectively. Twenty-one percent (21%) had a total cholesterol (TC) >200 mg/dl.  However, no relationship was observed between TC and GFR. Twenty-one percent (21%) had HDL-C <40 mg/d, and obese children had an average HDL-C 14% lower than children with normal BMI. Changes in LDL-C levels were not discussed in this study (17).

 

Longitudinal data of 508 children (76% non-glomerular CKD, 24% glomerular CKD) from the CKiD study, representing 1,514 person-visits and a median follow-up of 4 years (interquartile range, 2.1–6.0), showed that non-HDL-C and TG worsened in proportion to declining GFR, increasing BMI and worsening proteinuria (18).  A waist to height ratio of >0.49 has also been shown to be associated with lower HDL-C, higher left ventricular mass index, TGs, and non-HDL cholesterol compared to lean controls (19).

 

The prevalence of dyslipidemia was 61.5% among 356 East Asian pediatric patients < 20 years of age (median age 10.8 years; 246 males, 110 females) with CKD who participated in the KoreaN cohort study for Outcomes in patients With Pediatric Chronic Kidney Disease (KNOW-PedCKD) (20). Twenty-five percent (25%) had elevated TC, 19% elevated LDL-C, 15.2% low HDL-C, and 15.2% elevated TG. The authors demonstrated that children with glomerulonephropathy and nephrotic range proteinuria exhibited increased risk for high TC; whereas increased BMI z-score, elevated proteinuria, hypocalcemia, and 1,25-dihydroxyvitamin D deficiency were associated with low HDL-C. Glomerular filtration rate stage 3b or higher and hyperphosphatemia were associated with increased the risk for HTG (20).

 

Dialysis

 

While dyslipidemia is common in ESRD, the need for chronic dialysis, either hemodialysis (HD) or continuous ambulatory peritoneal dialysis (CAPD), often results in further alteration in lipids and lipoproteins. (21,22). Some studies demonstrate no significant differences in TC, LDL-C, HDL-C, TG, ApoA, ApoB, or Lp(a) serum levels between individuals receiving HD when compared to PD (23).  However other studies reported important differences in lipoprotein concentrations and their composition in adults undergoing HD and CAPD. A more atherogenic profile was observed in the latter group, consisting mainly of lower concentrations of HDL-C with higher levels of TC, TG, LDL-C, ApoB and ApoE. CAPD patients showed significantly higher TG and LDL-C levels, with a different pattern of apoprotein profile characterized by lower ApoA-I levels and higher ApoE levels than controls. Similar differences in ApoA-I and ApoE were also seen between controls and HD patients, whereas in the hemodialysis group a significant increase in ApoB was also observed (24).

 

There have been studies demonstrating different lipid patterns in children receiving dialysis.  Children (aged 12.6 +/- 4.7 years) undergoing treatment with continuous ambulatory peritoneal dialysis/continuous cycling peritoneal dialysis (CAPD/CCPD) were found to have fasting mean levels of TGs (90%) and cholesterol (69%) above the 95th percentile of published normal values prior to the start of dialysis. The authors found a high prevalence of hyperlipidemia at baseline with no significant change of serum lipid levels during 2 years of treatment with CAPD/CCPD. (25).

 

Renal Transplantation

 

Second only to infection, cardiovascular disease is a significant cause of mortality in pediatric renal transplant patients (26). Analysis of retrospective data from the CERTAIN registry (386-transplant recipients aged 0.5-25 years) showed the prevalence of dyslipidemia to be 95% before engraftment and 88% at 1-year following transplant (27). TC and LDL-C levels are considerably higher post-renal transplant compared to children undergoing hemodialysis (27). Risk factors include elevated pre-transplant serum cholesterol, years since renal transplant (28), and use of certain immunosuppressive medications (28).

 

Immunosuppressive drugs, including prednisone, cyclosporine, and sirolimus, have been shown to be associated with dyslipidemia, whereas the use of tacrolimus and mycophenolic acid is associated with lower lipid parameters (27,28,29). TC and LDL-C in these children have not been shown to have direct association with age, sex, ethnicity, duration of ESRD, stage of chronic kidney disease, diabetes mellitus, or BMI (27,28). Reduced GFR is a risk factor for elevated TGs in this population (27,30). 

 

DIAGNOSIS

 

Dyslipidemia in children with renal disease is the result of complex interactions of a variety of factors, including the primary disease process, use of medications such as corticosteroids, the presence of malnutrition or obesity, diet, and genetics.  When present in NS or those who have undergone renal transplantation, dyslipidemia it is easily recognized; while often less obvious in those with chronic renal insufficiency or ESRD.  Detection of dyslipidemia in the latter requires more careful analysis and knowledge of normal laboratory ranges for children. Current KDIGO clinical practice guidelines recommend an initial lipid profile in all newly diagnosed children with CKD, including those who require chronic dialysis therapy or kidney transplant therapy. After the initial lipid profile, annual testing is recommended (31).

 

MANAGEMENT

 

Since publication of the 2003 National Kidney Foundation-Kidney Disease Outcomes Quality Initiative (NKF-KDOQI) clinical guidelines for management of dyslipidemia in CKD, data from randomized controlled trials on statin therapy in adults with CKD KDIGO (Kidney Disease: Improving Global Outcomes or KDIGO) have helped inform management guidelines.  Recommendations for treatment (Table 4 and Table 5) are based on risk for coronary heart disease (32, 33, 34). It should be recognized, however, that clinical recommendations (Table 5) differ for adults as well as children. While all guidelines target CVD prevention, none specifically address treatment of lipid abnormalities to prevent deterioration of kidney function, especially in youth with NS (15,16).

 

Table 4.  Assessment of Lipid Status and Treatment in Children (< 18 years-of-age) with Chronic Kidney Disease (CKD).

Clinical Scenario

Lipid Profile

Statins ± Ezetimibe

Management of HTG

CKD, including those treated with chronic dialysis or kidney transplantation.

Recommended at initial diagnosis and repeated annually.

Not recommended.

Therapeutic lifestyle changes are recommended.

Lipid profile=TC, TG, HDL-C and LDL-C. Modified from: Wanner (32).

 

Table 5. Lipid Management Guidelines for CKD in Children (< 18 years-of-age)

KDIGO

ACC/AHA

2014 ADA

AACE

Do not initiate.

Prior AHA statement for high-risk patients (including CKD) recommends therapeutic lifestyle intervention; if >10 years-of-age and LDL-C remains >100 mg/dL despite therapeutic lifestyle recommendations, treat with statin.

If patient has DM, consider statin use ≥10 years-of-age if, following changes in diet and lifestyle, LDL-C >160 or >130 mg/dL with multiple risk factors.

Recommend pharmacotherapy for > 8 years-of-age if no response to therapeutic lifestyle, especially if LDL-C ≥ 190 or ≥ 160 mg/dL with additional risk factors.

Adapted from Table 1; Sarnak (33).  ACC/AHA, 2014 ADA, and AACE guidelines not CKD specific.

 

Nephrotic Syndrome

 

Treatment options for dyslipidemia in children with NS include lifestyle changes and pharmacologic agents.  Little evidence exists on optimal lifestyle management in children with NS, and the majority of studies have included adult populations. Studies of soy-based vegetarian diets have shown promising results, but include limited subjects and these findings have not been confirmed (35,36). The addition of omega-3 fatty acids has demonstrated a small decrease in TG and postprandial chylomicron levels (37,38).

 

There is also very limited data on pharmacologic treatment in youth compared with the adult population. Medications commonly used in adults include statins, bile acid sequestrants, and fibric acid derivatives.  Most of the studies in youth have been limited to statins. These studies have shown reductions in TG and LDL-C levels but tend to be small in number and often lack a control group (5).

 

The utility of lipid apheresis, a technique used to lower cholesterol in patients with homozygous familial hypercholesterolemia, has been assessed in youth with NS.  A study of children who underwent lipid apheresis in combination with prednisone found reductions in both cholesterol and TG.  Of the study group, 7/11 youth achieved a partial or complete remission of NS; and all remained in remission at their 10-year follow-up (39).  However, as discussed in other chapters of Endotext, lipid apheresis is currently not preformed specifically for lipid management. At present, apheresis is only FDA approved for new onset focal segmental glomerulosclerosis in pediatric patients who are resistant to standard forms of treatment (40).

 

Chronic Kidney Disease

 

The American Heart Association (AHA) classifies youth with ESRD in the highest risk group and those with pre-dialysis CKD at moderate risk for development of CVD and its sequelae. It recommends therapeutic lifestyle changes (TLC) as the initial management strategy, with the goal of lowering LDL to ≤130 mg/dL and TG <400; with addition of pharmacological therapy if these goals are not met (41).  

 

In contrast, the KDIGO 2013 clinical practice guidelines recommend assessment of fasting lipids annually, while discouraging the use of statin or statin/ezetimibe combination in youth <18 years with CKD (33,42).  Boys >10 years-of-age and post-menarche girls with severely elevated LDL-C in the setting of a family history of premature coronary disease, diabetes, hypertension, smoking, and ESRD might be candidates for low dose statin use. However multi-drug regimens, even in youth with severely elevated LDL-C (42), is not recommended.

 

In youth with fasting TG >500 mg/dL, KDIGO recommendations a very low-fat diet (<15% total calories), use of medium-chain triglycerides, and fish oil. Pharmacologic treatment can be considered in those with TG >1000 mg/dL, however, the safety or efficacy of fibric acid or niacin for this population is unknown (43).

 

In 2011, the NHLBI, although focused primarily on youth with FH, recommended pharmacologic management for children >10 years-of-age with LDL-C >190 mg/dL alone, >160 mg/dL with one high-risk condition, or >130 mg/dL with two high-risk conditions despite lifestyle modifications. High-risk conditions include high blood pressure (treated with antihypertensive medication), BMI >97th percentile, smoking, and chronic kidney disease (44).

 

Dialysis

 

CVD-related events are the leading cause of death among adults with ESRD receiving maintenance dialysis.  It accounts for 45% of deaths, a rate 10-30 times higher than that in the general population (45-49).

 

The relationship between serum cholesterol and CVD is more complex in individuals with CKD, particularly those receiving maintenance hemodialysis. A history of coronary heart disease, coronary artery bypass surgery, coronary angioplasty or an abnormal coronary angiogram was present in 36% (peritoneal dialysis) and 42% (hemodialysis dialysis) (50).

 

In contrast, comparable data in youth receiving maintenance dialysis are limited in regarding to the prevalence of CVD-related risk factors, clinical management of modifiable risk factors, and the incidence of morbidity and mortality.  Despite the common occurrence of hyperlipidemia in youth with ESRD, monitoring is rarely performed (51,52). The relative risk of CVD, however, appears to be even greater in younger dialysis patients (8).

 

A study by Blanche and colleagues suggests CVD is also common amongst children who require chronic dialysis. (Table 6) The type of cardiac-related events differed significantly among ethnic groups, being highest among Black youth (53).

 

Table 6.  Adjusted Annual Cardiac Events/1000 Patient-Years in Children Receiving Chronic Dialysis.

Event

1991

1992

1993

1994

1995

1996

Trend

Arrhythmia

90.9

115

138.9

145.0

141.3

128.6

P= NS

Cardiac Arrest

19.1

18.0

10.0

19.5

11.6

22.0

P= NS

Valvular disease

59.3

66.3

55.4

91.2

79.6

68.1

P= NS

Cardiomyopathy

42.0

44.9

50.9

60.7

73.8

84.8

P= 0.003

All-cause death

56.9

30.7

31.1

48.1

32.2

31.4

P=NS

Cardiac death

14.4

10.4

12.6

18.1

12.4

4.5

P=NS

Adapted from Table 2, Chavers (53).

 

Despite the increased prevalence of CVD, randomized controlled trials have not provided definitive evidence that lipid-lowering therapies are effective in reducing risk in adults with ESRD who require chronic dialysis. This lack of benefit may be the result of 1) a significant difference in the pathophysiology and spectrum of CVD in adults who require chronic dialysis compared to the general population; and 2) although affected by atherosclerosis, the majority of deaths in dialysis patients are not related to coronary artery disease and, therefore, would not be expected to respond favorably to lipid-lowering therapy.

 

As noted in the Table 6, coronary disease in youth with ESRD receiving chronic dialysis is also rare.  Most of the CVD involves cardiomyopathy and/or dysrhythmia. Therefore, as in adults, lipid-lowering therapy may be of limited benefit in this population.

 

In its 2014 clinical practice guidelines, the KDIGO Work Group noted that the magnitude of any relative risk reduction in individuals who require chronic dialysis appears to be substantially smaller than in earlier stages of CKD. Therefore, the KDIGO Work Group does not recommend initiation of statin treatment for most adults and children undergoing chronic dialysis. Previous guidelines in this population suggested the use of targets for LDL-C, with treatment escalation to higher doses of statin when LDL-C targets are not achieved with lower dose therapy. Current recommendations, however, do not support this strategy since higher doses of statins have not been proven to be safe in the setting of CKD. Furthermore, since LDL-C levels do not necessarily suggest the need to increase statin doses, follow-up measurement of lipid levels is not recommended (54).

 

While there is little evidence that lifestyle changes will reduce serum TG levels and/or improve clinical outcomes in adults, the KDIGO Work Group recommend advising youth with high fasting levels of serum TGs (>5.65 mmol/l or >500 mg/dl) to adopt lifestyle changes (54). Dietary modification should be used judiciously, if at all, in youth who are malnourished. The safety and efficacy of fibric acid and niacin have not been established in youth nor FDA approved for use in this population. Prescription omega-3-fatty acids appear to lower serum TGs in adults. The benefits, harms, and tolerability of such treatment in children is unproven, nor are there data to suggest preferential use of EPA vs combination EPA/DHA productions.

 

Transplantation

 

TLC remains the first-line intervention for treatment of dyslipidemia in youth who undergo renal transplantation. The KDOQI clinical practice guidelines for nutrition in youth recommend that families receive intensive nutrition guidance to promote a heart-healthy diet and ≥60 minutes of active play time daily, along with limiting screen time (television + computer + video games) to ≤2 hours per day (55). KDIGO guidelines recommends against the use of statin or statin/ezetimibe combination in youth <18 years, although low dose statin should be considered in boys >10 years and post-menarche girls with severely elevated LDL-C in the setting of a family history of premature coronary disease, diabetes, hypertension, smoking, and ESRD (42).

 

NHLBI and AHA both recommended considering pharmacologic therapy if LDL-C goals are not met with TLC alone. If statins are considered, caution needs to be exercised and low doses given concommitentlly with medications that utilize the CYP3A4 pathway, like cyclosporine, as they may increase serum concentration of the statin and risk of statin-induced rhabdomyolysis. There are no randomized trials for use of ezetimibe or bile acid sequestrants in pediatric renal transplant patients; and KDIGO does not recommend multi-drug regimens even in those with severely elevated LDL-C. The use of fibrates in adult renal transplant recipients with HTG has been accompanied by elevations in serum creatinine and also with reduced cyclosporine concentrations when used concomitantly (56).  It should be noted that the effect of cyclosporine is more complex than CYP3A4 inhibition alone (see chapter 18 of Endotext on medications which states, in part, “ Most statins are transported into the liver and other tissues by organic anion transporting polypeptides, particularly OATP1B1. Drugs, such as clarithromycin, ritonavir, indinavir, saquinavir, and cyclosporine that inhibit OATP1B1 can increase serum statin levels thereby increasing the risk of statin muscle toxicity. Fluvastatin is the statin that is least affected by OATP1B1 inhibitors. In fact, fluvastatin 40mg per day has been studied in adults receiving renal transplants concomitantly treated with cyclosporine and over a five year study period the risk of myopathy or rhabdomyolysis was not increased in the fluvastatin treated patients compared to those treated with placebo.”)

 

Several studies have shown an impact of cyclosporine, mTOR inhibitors, and prednisone immunosuppressive regimen on post-transplant dyslipidemia and may contribute to CVD morbidity and mortality in pediatric renal transplant recipients (27,28,57).  One study noted the prevalence of post-transplant dyslipidemia may be decreasing with the use of newer immunosuppressive regimens that include tacrolimus and lower doses of prednisone (28). Thus, improvement of the CVD risk profile may be accomplished by alteration of the immunosuppressive regimen.

 

CONCLUSIONS

 

Chronic kidney disease and nephrotic syndrome are often accompanied by dyslipidemia, contributing to disease-related morbidity and increasing risk of premature CVD. Given the lack of randomized controlled trials in youth and long-term clinical outcomes, such as CVD-related events and mortality, optimum management is unknown. Further research is needed to demonstrate the benefit of strategies to improve health and wellbeing in this vulnerable population, including use of lipid-lowering medications, with the aim of decreasing CVD-related events. In addition, given the role of dyslipidemia in potentially contributing to deterioration of renal function in youth with NS, aggressive lipid-lowering therapy may be beneficial. Further studies, however, are needed.

 

ACKNOWLEDGEMENTS

 

The authors would like to acknowledge Luke Hamilton, Suzanne Beckett, Dena Hanson, and Ashley Brock for their assistance in preparing and editing this manuscript.

 

REFERENCES

 

  1. Hager MR, Narla AD, Tannock LR. Dyslipidemia in patients with chronic kidney disease. Rev Endocr Metab Disord. 2017;18(1):29-40.
  2. Reiss AB, Voloshyna I, De Leon J, et al. Cholesterol metabolism in CKD. Am J Kidney Dis. 2015;66(6):1071-1082.
  3. Vaziri ND, Norris K. Lipid disorders and their relevance to outcomes in chronic kidney disease. Blood Purif. 2011;31(1-3):189-196.
  4. Matsushita K, Ballew SH, Coresh J. Cardiovascular risk prediction in people with chronic kidney disease. Curr Opin Nephrol Hypertens. 2016;25(6):518-523.
  5. Agrawal S, Zaritsky JJ, Fornoni A, Smoyer WE. Dyslipidaemia in nephrotic syndrome: mechanisms and treatment. Nat Rev Nephrol. 2018;14(1):57-70.
  6. Prospective Studies Collaboration. Cholesterol, diastolic blood pressure, and stroke: 13,000 strokes in 450,000 people in 45 prospective cohorts. Lancet. 1995;346(8991-8992):1647-1653.
  7. Law MR, Wald NJ, Thompson SG. By how much and how quickly does reduction in serum cholesterol concentration lower risk of ischaemic heart disease? BMJ. 1994;308(6925):367-372.
  8. Baigent C, Burbury K, Wheeler D. Premature cardiovascular disease in chronic renal failure. Lancet. 2000;356(9224):147-152.
  9. Baigent C, Armitage J. Cholesterol reduction among patients at increased risk of coronary heart disease. Proceedings-Royal College of Physicians of Edinburgh. 1999;29(Suppl 5):10-15.
  10. Baigent C, Wheeler DC. Should we reduce blood cholesterol to prevent cardiovascular disease among patients with chronic renal failure.Nephrol Dial Transplant. 2000;15(8):1118-1119.
  11. Saland JM, Ginsberg H, Fisher EA. Dyslipidemia in pediatric renal disease: epidemiology, pathophysiology, and management. Curr Opin Pediatr. 2002;14(2):197-204.
  12. Chu M, Wang AYM, Chan IHS, Chui SH, Lam CWK.Serum small-dense LDL abnormalities in chronic renal disease patients. Br J Biomed Sci.2012(69):99-102.
  13. Hopewell JC, Haynes R, Baigent C. The role of lipoprotein (a) in chronic kidney disease. Journal of Lipid Research. 2018; (59):577-585.
  14. Mikolasevic I, Zutelija M, Mavrinac V, Orlic L. Dyslipidemia in patients with chronic kidney disease: etiology and management. International Journal of Nephrology and Renovascular Disease. 2017; (10):35-45
  15. Moorhead JF, Chan MK, El-Nahas M, Varghese Z. Lipid nephrotoxicity in chronic progressive glomerular and tubulo-interstitial disease. Lancet. 1982;2(8311):1309-1311.
  16. Gyebi L, Soltani Z, Reisin E. Lipid nephrotoxicity: new concept for an old disease. Curr Hypertens Rep. 2012;14(2):177-181.
  17. Saland JM, Pierce CB, Mitsnefes MM, et al. Dyslipidemia in children with chronic kidney disease. Kidney Int. 2010;78(11):1154-1163.
  18. Saland JM, Kupferman JC, Pierce CB, et al. Change in dyslipidemia with declining glomerular filtration rate and increasing proteinuria in children with CKD. Clin J Am Soc Nephrol. 2019;14(12):1711-1718.
  19. Sgambat K, Roem J, Mitsnefes M, et al. Waist-to-height ratio, body mass index, and cardiovascular risk profile in children with chronic kidney disease. Pediatr Nephrol. 2018;33(9):1577-1583.
  20. Baek HS, Kim SH, Kang HG, et al. Dyslipidemia in pediatric CKD patients: results from KNOW-PedCKD (KoreaN cohort study for Outcomes in patients wwth Pediatric CKD). Pediatr Nephrol. 2020;35(8):1455-1461.
  21. Sentí M, Romero R, Pedro-Botet J, et al. Lipoprotein abnormalities in hyperlipidemic and normolipidemic men on hemodialysis with chronic renal failure. Kidney Int. 1992;41(5):1394-1399.
  22. Lacour B, Roullet JB, Beyne P, et al. Comparison of several atherogenicity indices by the analysis of serum lipoprotein composition in patients with chronic renal failure with or without haemodialysis, and in renal transplant patients. J Clin Chem Clin Biochem. 1985;23(12):805-810.
  23. Kanbay M, Delibasi T, Kaya A, et al. Effect of dialysis type on serum lipids, apolipoproteins, and lipoproteins. Ren Fail. 2006;28(7):567-571.
  24. Fytili CI, Progia EG, Panagoutsos SA, et al. Lipoprotein abnormalities in hemodialysis and continuous ambulatory peritoneal dialysis patients. Ren Fail. 2002;24(5):623-630.
  25. Querfeld U, Salusky IB, Nelson Pet al. Hyperlipidemia in pediatric patients undergoing peritoneal dialysis. Pediatr Nephrol. 1988;2(4):447-452.
  26. Mitsnefes MM. Cardiovascular disease in children with chronic kidney disease. J Am Soc Nephrol. 2012;23(4):578-585.
  27. Habbig S, Volland R, Krupka K, et al. Dyslipidemia after pediatric renal transplantation-the impact of immunosuppressive regimens. Pediatr Transplant. 2017;21(3): e12914.
  28. Sgambat K, He J, McCarter RJ, Moudgil A. Lipoprotein profile changes in children after renal transplantation in the modern immunosuppression era. Pediatr Transplant. 2008;12(7):796-803.

29     Herink M, Ito MK.  Medication Induced Changes in Lipids and Lipoproteins. Endotext [Internet]. May 8, 2018.

  1. Silverstein DM, Palmer J, Polinsky MS, et al. Risk factors for hyperlipidemia in long-term pediatric renal transplant recipients. Pediatr Nephrol. 2000;14(2):105-110.
  2. Chapter 3: Assessment of lipid status in children with CKD. Kidney Int Suppl (2011). 2013;3(3):280-281.
  3. Wanner C, Tonelli M, Kidney Disease: Improving Global Outcomes Lipid Guideline Development Work Group Members. KDIGO Clinical Practice Guideline for Lipid Management in CKD: summary of recommendation statements and clinical approach to the patient. Kidney Int. 2014;85(6):1303-1309.
  4. Sarnak MJ, Bloom R, Muntner P, et al. KDOQI US commentary on the 2013 KDIGO Clinical Practice Guideline for Lipid Management in CKD. Am J Kidney Dis. 2015;65(3):354-366.
  5. Tannock L.Dyslipidemia in Chronic Kidney Disease. Endotext [Internet]. January 22, 2018.
  6. D'Amico G, Gentile MG, Manna G, et al. Effect of vegetarian soy diet on hyperlipidaemia in nephrotic syndrome. Lancet. 1992;339(8802):1131-1134.
  7. Gentile MG, Fellin G, Cofano F, et al. Treatment of proteinuric patients with a vegetarian soy diet and fish oil. Clin Nephrol. 1993;40(6):315-320.
  8. Bell S, Cooney J, Packard CJ, et al. The effect of omega-3 fatty acids on the atherogenic lipoprotein phenotype in patients with nephrotic range proteinuria. Clin Nephrol. 2012;77(6):445-453.
  9. Hall AV, Parbtani A, Clark WF, et al. Omega-3 fatty acid supplementation in primary nephrotic syndrome: effects on plasma lipids and coagulopathy. J Am Soc Nephrol. 1992;3(6):1321-1329.
  10. Hattori M, Chikamoto H, Akioka Y, et al. A combined low-density lipoprotein apheresis and prednisone therapy for steroid-resistant primary focal segmental glomerulosclerosis in children. Am J Kidney Dis. 2003;42(6):1121-1130.
  11. Feingold KR, Grunfeld C.Lipoprotein Apheresis.  Endotext [Internet]. January 18,2020.
  12. de Ferranti SD, Steinberger J, Ameduri R, et al. Cardiovascular risk reduction in high-risk pediatric patients: a scientific statement from the American Heart Association. Circulation. 2019;139(13):e603-e634.
  13. Chapter 4: Pharmacological cholesterol-lowering treatment in children. Kidney Int Suppl (2011). 2013;3(3):282-283.
  14. Chapter 6: Triglyceride-lowering treatment in children. Kidney Int Suppl (2011). 2013;3(3):286.
  15. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents, National Heart, Lung, and Blood Institute. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. 2011;128 Suppl 5(Suppl 5):213.
  16. United States Renal Data System Coordinating Center. USRDS 2013 annual data report: atlas of chronic kidney disease and end-stage renal disease in the United States. Bethesda, MD, United States: National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 2013.
  17. Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis. 1998;32(5 Suppl 3):112.
  18. Foley RN, Collins AJ. End-stage renal disease in the United States: an update from the United States Renal Data System. J Am Soc Nephrol. 2007;18(10):2644-2648.
  19. Herzog CA, Asinger RW, Berger AK, et al. Cardiovascular disease in chronic kidney disease. A clinical update from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2011;80(6):572-586.
  20. Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation.2003;108(17):2154-2169.
  21. The USRDS Dialysis Morbidity and Mortality Study: Wave 2. United States Renal Data System. Am J Kidney Dis. 1997;30(2):S67-S85.
  22. Querfeld U, Lang M, Friedrich JB, et al. Lipoprotein(a) serum levels and apolipoprotein(a) phenotypes in children with chronic renal disease. Pediatr Res. 1993;34(6):772-776.
  23. Ma KW, Greene EL, Raij L. Cardiovascular risk factors in chronic renal failure and hemodialysis populations. Am J Kidney Dis. 1992;19(6):505-513.
  24. Chavers BM, Li S, Collins AJ, Herzog CA. Cardiovascular disease in pediatric chronic dialysis patients. Kidney Int. 2002;62(2):648-653.
  25. Wanner C, Tonelli M.KDIGO Clinical Practice Guideline for Lipid Management in CDK: Summary of Recommendation statements and clinical approach to the Patient. Kidney Int. 2014:85(6):1303-09.
  26. KDOQI Work Group. KDOQI Clinical Practice Guideline for Nutrition in Children with CKD: 2008 update. Executive summary. Am J Kidney Dis. 2009;53(3 Suppl 2):S11-S104.
  27. Devuyst O, Goffin E, Pirson Y, et al. Creatinine rise after fibrate therapy in renal graft recipients. Lancet. 1993;341(8848):840.
  28. Filler G, Medeiros M. Improving long-term outcomes after pediatric renal transplantation by addressing dyslipidemia. Pediatr Transplant. 2017;21(3):e12880.

Monogenic Disorders Causing Hypobetalipoproteinemia – copy

ABSTRACT

 

Monogenic mutations leading to hypobetalipoproteinemia are rare. The monogenic causes of hypobetalipoproteinemia include familial hypobetalipoproteinemia, abetalipoproteinemia, chylomicron retention disease, loss of function mutations in PCSK9, and loss of function mutations in angiopoietin-like protein 3 (ANGPTL3) (Familiar Combined Hypolipidemia). This chapter describes the etiology, pathogenesis, clinical and laboratory findings, and the treatment of these rare monogenic disorders.

 

INTRODUCTION

 

Monogenic mutations leading to hypobetalipoproteinemia are rare. The monogenic causes of hypobetalipoproteinemia include familial hypobetalipoproteinemia (FHBL), abetalipoproteinemia (ABL), chylomicron retention disease (CMRD), loss of function mutations in PCSK9, and loss of function mutations in angiopoietin-like protein 3 (ANGPTL3) (Familial Combined Hypolipidemia, FCH). Increased understanding of the genetic and the molecular underpinnings of these disorders has allowed a focused prioritization of therapeutic targets for drug development. Table 1 summarizes genetic, lipid, and clinical features of the major hypobetalipoproteinemia syndromes. Of note the parental lipid profile is normal in abetalipoproteinemia and chylomicron retention disease.

 

It should be recognized that secondary, non-familial, forms of hypobetalipoproteinemia occur and include strict vegan diet, malnutrition, hyperthyroidism, malignancy, and chronic liver disease. In addition, hypobetalipoproteinemia can also be due to polymorphisms in multiple genes that together result in hypobetalipoproteinemia (polygenic etiology) (1-3). In a study of 111 patients with LDL-C levels below the fifth percentile 36% had monogenic hypobetalipoproteinemia, 34% had polygenic hypobetalipoproteinemia, and 30% had hypobetalipoproteinemia from an unknown cause (1). In a study of women with an LDL-C ≤1st percentile (≤50 mg/dL) 15.7% carried mutations causing monogenic hypocholesterolemia and 49.6% were genetically predisposed to a low LDL-C on the basis of an extremely low weighted polygenetic risk score (3).

 

Table 1. Characteristics of the Hypobetalipoproteinemia Syndromes

 

Inheritance

Effected gene

Prevalence

Lipids

Clinical features

FHBL

ACD

Truncation mutations in Apo B

1:1000 – 1:3000

Apo B <5th percentile,

LDL-C 20- 50 mg/dL

Hepatic steatosis

Mild elevation of transaminases. Lower prevalence of ASCVD

ABL

AR

MTTP

<1:1,000,000

Triglycerides < 30 mg/dl,

Cholesterol < 30 mg/dl),

LDL and Apo B undetectable

Hepatic steatosis

Malabsorption, steatorrhea, diarrhea, and failure to thrive.

Deficiency of fat-soluble vitamins.

PCSK9

ACD

Loss of function mutations in PCSK9

 

Heterozygous – mild to moderate reduction in LDL-C

Homozygous – LDL-C ~15 mg/dl

Normal health; significantly lower prevalence of ASCVD

FCH

ACD

Loss of function mutations in ANGPTL3

Very rare

Panhypolipidemia

Normal health; significantly lower prevalence of ASCVD

CMRD

AR

SAR1B

Very rare

LDL-C and HDL-C -decreased by 50%,

Triglycerides - normal

hypocholesterolemia associated with failure to thrive, diarrhea, steatorrhea, and abdominal distension

ACD- autosomal co-dominant; AR- autosomal recessive; FHBL- familial hypobetalipoproteinemia; ABL- abetalipoproteinemia; FCH- Familiar Combined Hypolipidemia; CMRD- chylomicron retention disease, MTTP- microsomal triglyceride transfer protein; ANGPTL3- angiopoietin-like protein 3; ASCVD- atherosclerotic cardiovascular disease

 

FAMILIAL HYPOBETALIPOPROTEINEMIA  

 

Familial Hypobetalipoproteinemia (FHBL) is most commonly due to truncation mutations in the gene coding for Apo B (4-6). Variants that lead to truncated proteins that are 30% in length or shorter have more severe signs and symptoms than those with longer truncated proteins (4,5). The truncated forms of Apo B found in FHBL are generally non-functional (truncation decreases lipidation and secretion) and are catabolized quickly, resulting in markedly reduced levels in the plasma (Apo B <5th percentile and LDL-C typically between 20- 50 mg/dL) (5,6). Although there is one normal allele in heterozygous FHBL, plasma Apo B levels are approximately 25% of normal rather than the predicted 50% (6). These lower than expected levels result from a lower secretion rate of VLDL Apo B from the liver, decreased production of LDL Apo B, increased catabolism of VLDL, and extremely low secretion of the truncated Apo B (4-6). Given the reduced substrate (Apo B) for lipid (predominantly triglyceride) loading, fatty liver develops in these patients (4,7). Hepatic steatosis and mild elevation of liver enzymes are common in heterozygous FHBL (4,7). Interestingly, individuals with monogenic hypobetalipoproteinemia had a much greater prevalence of hepatic dysfunction than individuals with polygenic hypobetalipoproteinemia (1). In contrast to non-alcoholic fatty liver disease, FHBL is not associated with hepatic or peripheral insulin resistance (7). This observation, however, does not imply that hepatic steatosis associated with FHBL is benign. There are several reports of steatohepatitis, cirrhosis, and hepatocellular carcinoma in patients with FHBL and it is estimated that 5-10% of individuals with FHBL develop relatively more severe nonalcoholic steatohepatitis (4). Because of the risk of developing liver disease liver function tests should be checked every 1-2 years and a hepatic ultrasound in those with elevated liver transaminases (4). While hepatic fat accumulation is the rule, there is generally sufficient chylomicron production to handle dietary fat. However, oral fat intolerance and intestinal fat malabsorption have been reported (4). On the positive side the decrease in proatherogenic lipoproteins has been associated with a reduced risk of cardiovascular disease (8).

 

Given the association of FHBL and low LDL-C, Apo B has been an attractive target for drug development. Indeed, unraveling the genetic and molecular mechanisms of FHBL provided the motivation to pharmacologically antagonize Apo B synthesis for therapeutic gains. This culminated in the production of mipomersen, a synthetic single strand anti-sense oligonucleotide to Apo B (9,10). Essentially, anti-sense oligonucleotides contain approximately ~20 deoxyribonucleic acid (DNA) base pairs complementary to a unique messenger ribonucleic acid (mRNA) sequence. The hybridization of the anti-sense oligonucleotide to the mRNA of interest leads to its catabolism via RNase H1, with markedly reduced mRNA levels and ultimately reduced target protein levels. In this case, mipomersen binds to Apo B mRNA leading to reduced production of the protein, and mimicking (albeit to a lesser extent) FHBL. Mipomersen is the first anti-sense oligonucleotide approved by the United States Food and Drug Administration (FDA) and was commercialized in 2013 with a limited indication for adjunctive LDL-C lowering in patients with homozygous familial hypercholesterolemia (HoFH) (10). It is an injectable agent administered subcutaneously once a week. In the clinical trials, mipomersen was associated with a reduction of LDL-C by 21% in subjects with HoFH and 33% in subjects with heterozygous familial hypercholesterolemia (HeFH) (10). Interestingly, it was also found to lower Lp(a) by 21- 23% (10). While it is highly efficacious in LDL-C lowering, it has side effects, many of which can be predicted based on the experience with FHBL (e.g., hepatic steatosis, elevated liver enzymes) (10). It is also associated with injection site reactions in a considerable number of subjects (10). In May 2018 sales were discontinued due to safety concerns related to increased liver transaminases and fatty liver.

 

Homozygous hypobetalipoproteinemia (HHBL) is extremely rare (4). These patients are homozygous or compound heterozygous for mutations in the Apo B gene. The clinical manifestations mimic ABL (see below) (4).

 

ABETALIPOPROTEINEMIA  

 

Abetalipoproteinemia (ABL) is a rare disorder characterized by very low plasma concentrations of triglyceride and cholesterol (under 30 mg/dl) and undetectable levels of LDL and Apo B (5,11,12). HDL-C levels are usually normal or modestly reduced. It is due to mutations in the gene that codes for microsomal triglyceride transfer protein (MTTP) (5,11-13). MTTP lipidates nascent Apo B in the endoplasmic reticulum to produce VLDL and chylomicrons in the liver and small intestine, respectively (13,14). Unlipidated Apo B is targeted for proteasomal degradation leading to the absence of Apo B containing lipoproteins in the plasma (and thus markedly reduced levels of LDL-C and triglycerides) (13,14). Similar to FHBL, VLDL production is inhibited (12). The reduced triglyceride export from the liver leads to hepatic steatosis, which rarely may progress to steatohepatitis, fibrosis, and cirrhosis (7,11). Additionally, lack of MTTP facilitated lipidation of chylomicrons in the small intestine results in lipid accumulation in enterocytes with associated malabsorption, steatorrhea, and diarrhea (5,11). The malabsorption and diarrhea lead to failure to thrive during infancy (5,11). Acanthocytosis may encompass 50% of circulating red blood cells (red blood cells with spiked cell membranes, due to thorny projections) (11,12). An additional issue of importance related to ABL is deficiency of fat-soluble vitamins (11). Early diagnosis of ABL and homozygous hypobetalipoproteinemia is extremely important as vitamin E deficiency culminates in atypical retinitis pigmentosa, spinocerebellar degeneration with ataxia, and vitamin K deficiency can lead to a significant bleeding diathesis (11). High dose supplementation with fat soluble vitamins early in life can prevent these devastating complications (5,11). Additional treatment measures include a low-fat diet and supplementation with essential fatty acids (5,11).

 

Given the very low level of atherogenic lipoproteins and lipids associated with ABL, there was interest in inhibiting MTTP therapeutically. Lomitapide is an oral MTP inhibitor that has been developed over the course of many years (10,15). In early trials, it was tested at a relatively high dose and the side effect profile was prohibitive (nausea, flatulence, and diarrhea). The more recent clinical trial program tested lower doses with drug titration in subjects with HoFH (10,15). On an intention to treat basis, LDL-C was decreased by 40% and apolipoprotein B by 39% (10). In patients who were actually taking lomitapide, LDL-C levels were reduced by 50% (10). In addition to decreasing LDL-C levels, non-HDL-C levels were decreased by 50%, Lp(a) by 15%, and triglycerides by 45% (10). Lomitapide received the same limited indication as mipomersen for adjunctive treatment of patients with HoFH (10). Besides the gastrointestinal issues already alluded to, its side effect profile includes hepatic steatosis (10). Its long-term safety has not been established.

 

PROPROTEIN CONVERTASE SUBTILISIN/KEXIN TYPE 9 (PCSK9)

 

Proprotein convertase subtilisin/ kexin type 9 (PCSK9) belongs to the proprotein convertase class of serine proteases (16-18). After synthesis, PCSK9 undergoes autocatalytic cleavage. This step is required for secretion, most likely because the prodomain functions as a chaperone and facilitates folding (16,17). PCSK9 is associated with LDL particles and the LDL-receptor (LDLR) (18). In 2003, Abifadel reported the seminal work that mapped PCSK9 as the third locus for autosomal dominant hypercholesterolemia (Familial Hypercholesterolemia- FH) (19). This finding revealed a previously unknown actor involved in cholesterol homeostasis and served to launch a series of investigations into PCSK9 biology. As it turns out, PCSK9 functions as a central regulator of plasma LDL-C concentration (16-18). It binds to the LDLR and targets it for destruction in the lysosome (16-18). Overactivity of PCSK9 results in a decrease in LDLR and an increase in LDL-C levels while decreased activity of PCSK9 results in an increase in LDLR and a decrease in LDL-C.

 

Since the discovery of gain-of-function mutations in PCSK9 as a cause of FH, investigators have also uncovered loss of function mutations of PCSK9. Loss-of-function mutations in PCSK9 are associated with low LDL-C levels and markedly reduced ASCVD (16,17). In African Americans 2.6 percent had nonsense mutations in PCSK9 that resulted in a 28 percent reduction in LDL-C and an 88 percent reduction in the risk of coronary heart disease (20). The hypolipidemia is not associated with liver abnormalities or other disorders. Interestingly, rare individuals homozygous or compound heterozygotes for loss of function mutations in PCSK9 have been reported with extremely low levels of LDL-C (~15 mg/dl), normal health and reproductive capacity, and no evidence of neurologic or cognitive dysfunction (18,21,22). Collectively, these observations served as further motivation to pursue antagonism of PCSK9 as a therapeutic target. Antagonizing PCSK9 would prolong the lifespan of LDLR, leading to significant reductions in plasma LDL-C levels.

 

There are numerous approaches to inhibiting PCSK9 including humanized monoclonal antibodies (mAbs), gene silencing, and use of small inhibitory peptides (18). Thus far, approaches utilizing mAbs are FDA approved (10). The two fully human monoclonal antibodies (alirocumab and evolocumab) targeting PCSK9 became commercially available in 2015. Clinical trials of mAbs targeted to PCSK9 have demonstrated remarkable efficacy in LDL-C reduction (~50% reduction in LDL-C as monotherapy and ~65% reduction in LDL-C in combination with a statin) with an excellent short-term safety and tolerability profile (10). Moreover, a large randomized controlled trial (FOURIER) demonstrated incremental improvement with a 15% reduction in the composite primary endpoint of major adverse cardiovascular outcome with addition of evolocumab on top of standard of care in patients with stable vascular disease (23). Additionally, the ODYSSEY OUTCOMES trial also demonstrated a similar reduction in major adverse cardiovascular events with alirocumab vs. placebo in patients with recent acute coronary syndromes (24). Finally, inclisiran, a small interfering RNA that inhibits translation of PCSK9, is approved in Europe but not yet in the US (10).

 

FAMILIAL COMBINED HYPOLIPIDEMIA

 

Familial combined hypolipidemia (FCH) is due to loss of function mutations in the gene encoding angiopoietin-like protein 3 (ANGPTL3) (25,26). ANGPTL3 inhibits various lipases, such as lipoprotein lipase and endothelial lipase (25,26). Therefore, loss of function mutations in ANGPTL3 relinquishes this inhibition resulting in more efficient metabolism of VLDL and HDL particles (25,26). In addition, to increasing VLDL clearance the secretion of VLDL is also decreased due to a decrease in free fatty acid flux to the liver (25). LDL clearance is increased but the mechanism remains to be fully elucidated (25). Studies have suggested that ANGPTL3 inhibition lowers LDL-C by limiting LDL particle production due to ANGPTL3 inhibition and increased endothelial lipase activity reducing VLDL-lipid content and size, generating remnant particles that are efficiently removed from the circulation rather than being further metabolized to LDL (27). Clinically, FCH manifests as panhypolipidemia (decreased triglycerides, LDL-C, and HDL-C) (25,26). Interestingly, heterozygotes for certain nonsense mutations in the first exon of ANGPTL3 have moderately reduced LDL-C and triglyceride levels while compound heterozygotes have significant reductions in HDL-C as well (25,26).  Homozygosity or compound heterozygosity for other loss-of-function mutations in exon 1 of ANGPTL3 have no detectable ANGPTL3 in plasma and striking reductions of atherogenic lipoproteins with HDL particles containing only apo A-I and preß-HDL. Individuals who are heterozygous for the loss of function mutations in ANGPTL3 have normal HDL-C levels and significantly reduced LDL-C (<25th percentile) (25,26).

 

A pooled analysis of cases of familial combined hypolipidemia was published 2013 (28). One hundred fifteen individuals carrying 13 different mutations in the ANGPTL3 gene (14 homozygotes, 8 compound heterozygotes, and 93 heterozygotes) and 402 controls were evaluated. Homozygotes and compound heterozygotes (two mutant alleles) had no measurable ANGPTL3 protein. In heterozygotes, ANGPTL3 was reduced by 34-88%, according to genotype. All cases (homozygotes and heterozygotes) demonstrated significantly lower concentrations of all plasma lipoproteins (except for Lp(a)) as compared to controls. Familial combined hypolipidemia is not associated with any comorbidity. In fact, the prevalence of fatty liver was the same as controls. However, ANGPTL3 deficiency is associated with a reduced risk of cardiovascular disease (25,29).

 

Recently, evinacumab, a human monoclonal antibody against ANGPTL3, was approved for the treatment of Homozygous Familial Hypercholesterolemia (10). Evinacumab decreases LDL-C levels by mechanisms independent of LDL receptor activity (10).

 

CHYLOMICRON RETENTION DISEASE

 

Chylomicron retention disease (CMRD), known also as Anderson’s disease for the individual who first described the condition in 1961, is a rare inherited lipid malabsorption syndrome (30,31). It is due to mutations in the SAR1B gene which codes for the protein SAR1b, a small GTPase, involved in intracellular protein trafficking (30). Mutations in SAR1b result in the failure of pre-chylomicrons to move from the endoplasmic reticulum to the golgi (30). This disorder usually presents in young infants with diarrhea, steatorrhea, abdominal distention, and failure to thrive (30,31). Patients with CMRD demonstrate a specific autosomal recessive hypocholesterolemia that differs from other familial hypocholesterolemias. CMRD is associated with a 50% reduction in both plasma LDL-C and HDL-C with normal fasting triglyceride levels (30,31). Mutations in SAR1B do not affect VLDL secretion by the liver. The decrease in HDL-C is postulated to be due to a decrease in Apo A-I secretion and cholesterol efflux by the small intestine (30). The mechanism accounting for the decrease in LDL-C is not clear. The usual increase in triglycerides and chylomicron levels following a fat meal is blocked (30). The duodenal mucosa is white on endoscopy and intestinal biopsy reveals cytosolic lipid droplets and lipoprotein-sized particles in enterocytes (30). As one would expect the absorption of fat-soluble vitamins (A, D, K, and E) and essential fatty acids is impaired (30,31).

 

Treatment for individuals with CMRD is similar to that described above for individuals with ABL (31).

 

REFERENCES

 

  1. Rimbert A, Vanhoye X, Coulibaly D, Marrec M, Pichelin M, Charriere S, Peretti N, Valero R, Wargny M, Carrie A, Lindenbaum P, Deleuze JF, Genin E, Redon R, Rollat-Farnier PA, Goxe D, Degraef G, Marmontel O, Divry E, Bigot-Corbel E, Moulin P, Cariou B, Di Filippo M. Phenotypic Differences Between Polygenic and Monogenic Hypobetalipoproteinemia. Arterioscler Thromb Vasc Biol 2021; 41:e63-e71
  2. Blanco-Vaca F, Martin-Campos JM, Beteta-Vicente A, Canyelles M, Martinez S, Roig R, Farre N, Julve J, Tondo M. Molecular analysis of APOB, SAR1B, ANGPTL3, and MTTP in patients with primary hypocholesterolemia in a clinical laboratory setting: Evidence supporting polygenicity in mutation-negative patients. Atherosclerosis2019; 283:52-60
  3. Balder JW, Rimbert A, Zhang X, Viel M, Kanninga R, van Dijk F, Lansberg P, Sinke R, Kuivenhoven JA. Genetics, Lifestyle, and Low-Density Lipoprotein Cholesterol in Young and Apparently Healthy Women. Circulation 2018; 137:820-831
  4. Burnett JR, Hooper AJ, Hegele RA. APOB-Related Familial Hypobetalipoproteinemia. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, eds. GeneReviews((R)). Seattle (WA) 2021.
  5. Hooper AJ, Burnett JR. Update on primary hypobetalipoproteinemia. Curr Atheroscler Rep 2014; 16:423
  6. Hooper AJ, van Bockxmeer FM, Burnett JR. Monogenic hypocholesterolaemic lipid disorders and apolipoprotein B metabolism. Crit Rev Clin Lab Sci 2005; 42:515-545
  7. Welty FK. Hypobetalipoproteinemia and abetalipoproteinemia: liver disease and cardiovascular disease. Curr Opin Lipidol 2020; 31:49-55
  8. Peloso GM, Nomura A, Khera AV, Chaffin M, Won HH, Ardissino D, Danesh J, Schunkert H, Wilson JG, Samani N, Erdmann J, McPherson R, Watkins H, Saleheen D, McCarthy S, Teslovich TM, Leader JB, Lester Kirchner H, Marrugat J, Nohara A, Kawashiri MA, Tada H, Dewey FE, Carey DJ, Baras A, Kathiresan S. Rare Protein-Truncating Variants in APOB, Lower Low-Density Lipoprotein Cholesterol, and Protection Against Coronary Heart Disease. Circ Genom Precis Med 2019; 12:e002376
  9. Crooke ST, Geary RS. Clinical pharmacological properties of mipomersen (Kynamro), a second generation antisense inhibitor of apolipoprotein B. Br J Clin Pharmacol 2013; 76:269-276
  10. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Grossman A, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  11. Burnett JR, Hooper AJ, Hegele RA. Abetalipoproteinemia. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, eds. GeneReviews((R)). Seattle (WA) 2018.
  12. Lee J, Hegele RA. Abetalipoproteinemia and homozygous hypobetalipoproteinemia: a framework for diagnosis and management. J Inherit Metab Dis 2014; 37:333-339
  13. Wetterau JR, Lin MC, Jamil H. Microsomal triglyceride transfer protein. Biochim Biophys Acta 1997; 1345:136-150
  14. Hussain MM, Shi J, Dreizen P. Microsomal triglyceride transfer protein and its role in apoB-lipoprotein assembly. J Lipid Res 2003; 44:22-32
  15. Cuchel M, Rader DJ. Microsomal transfer protein inhibition in humans. Curr Opin Lipidol 2013; 24:246-250
  16. Debose-Boyd RA, Horton JD. Opening up new fronts in the fight against cholesterol. Elife 2013; 2:e00663
  17. Seidah NG, Awan Z, Chretien M, Mbikay M. PCSK9: a key modulator of cardiovascular health. Circ Res 2014; 114:1022-1036
  18. Shapiro MD, Tavori H, Fazio S. PCSK9: From Basic Science Discoveries to Clinical Trials. Circ Res 2018; 122:1420-1438
  19. Abifadel M, Varret M, Rabes JP, Allard D, Ouguerram K, Devillers M, Cruaud C, Benjannet S, Wickham L, Erlich D, Derre A, Villeger L, Farnier M, Beucler I, Bruckert E, Chambaz J, Chanu B, Lecerf JM, Luc G, Moulin P, Weissenbach J, Prat A, Krempf M, Junien C, Seidah NG, Boileau C. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet 2003; 34:154-156
  20. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006; 354:1264-1272
  21. Zhao Z, Tuakli-Wosornu Y, Lagace TA, Kinch L, Grishin NV, Horton JD, Cohen JC, Hobbs HH. Molecular characterization of loss-of-function mutations in PCSK9 and identification of a compound heterozygote. Am J Hum Genet 2006; 79:514-523
  22. Cariou B, Ouguerram K, Zair Y, Guerois R, Langhi C, Kourimate S, Benoit I, Le May C, Gayet C, Belabbas K, Dufernez F, Chetiveaux M, Tarugi P, Krempf M, Benlian P, Costet P. PCSK9 dominant negative mutant results in increased LDL catabolic rate and familial hypobetalipoproteinemia. Arterioscler Thromb Vasc Biol 2009; 29:2191-2197
  23. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR, FOURIER Steering Committee and Investigators. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722
  24. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM, ODYSSEY OUTCOMES Committees and Investigators. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  25. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  26. Kersten S. Angiopoietin-like 3 in lipoprotein metabolism. Nat Rev Endocrinol 2017; 13:731-739
  27. Adam RC, Mintah IJ, Alexa-Braun CA, Shihanian LM, Lee JS, Banerjee P, Hamon SC, Kim HI, Cohen JC, Hobbs HH, Van Hout C, Gromada J, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res2020; 61:1271-1286
  28. Minicocci I, Santini S, Cantisani V, Stitziel N, Kathiresan S, Arroyo JA, Marti G, Pisciotta L, Noto D, Cefalu AB, Maranghi M, Labbadia G, Pigna G, Pannozzo F, Ceci F, Ciociola E, Bertolini S, Calandra S, Tarugi P, Averna M, Arca M. Clinical characteristics and plasma lipids in subjects with familial combined hypolipidemia: a pooled analysis. J Lipid Res 2013; 54:3481-3490
  29. Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, Leader JB, Murray MF, Ritchie MD, Kirchner HL, Habegger L, Lopez A, Penn J, Zhao A, Shao W, Stahl N, Murphy AJ, Hamon S, Bouzelmat A, Zhang R, Shumel B, Pordy R, Gipe D, Herman GA, Sheu WHH, Lee IT, Liang KW, Guo X, Rotter JI, Chen YI, Kraus WE, Shah SH, Damrauer S, Small A, Rader DJ, Wulff AB, Nordestgaard BG, Tybjaerg-Hansen A, van den Hoek AM, Princen HMG, Ledbetter DH, Carey DJ, Overton JD, Reid JG, Sasiela WJ, Banerjee P, Shuldiner AR, Borecki IB, Teslovich TM, Yancopoulos GD, Mellis SJ, Gromada J, Baras A. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease. N Engl J Med 2017; 377:211-221
  30. Levy E, Poinsot P, Spahis S. Chylomicron retention disease: genetics, biochemistry, and clinical spectrum. Curr Opin Lipidol 2019; 30:134-139
  31. Sissaoui S, Cochet M, Poinsot P, Bordat C, Collardeau-Frachon S, Lachaux A, Lacaille F, Peretti N. Lipids Responsible for Intestinal or Hepatic Disorder: When to Suspect a Familial Intestinal Hypocholesterolemia? J Pediatr Gastroenterol Nutr 2021; 73:4-8

 

Adolescent Bariatric Surgery

ABSTRACT

 

The prevalence of adolescent obesity has rapidly increased over the past several decades. With this increase, there has also been a rise in the prevalence of complications of obesity leading to premature mortality. While lifestyle and medical management remain a part of the initial management of obesity, these therapies have been shown to be inferior when compared to metabolic and bariatric surgery (MBS) for adolescents with severe obesity. A multidisciplinary approach is recommended to evaluate medically eligible candidates for MBS, prepare patients for surgery, and guide postoperative management. Laparoscopic sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) are the most common MBS procedures performed in both adolescent and adult patients. Postoperative hospital stays are generally short and long-term routine follow-up with the MBS team is recommended to monitor weight loss, resolution of complications of obesity, and to monitor for postoperative complications. Most adolescent MBS studies demonstrate average percent body mass index loss between 25-29% after surgery. This is also associated with resolution or improvement of most complications of obesity at rates that are similar or superior to adult studies. Resolution and prevention of type 2 diabetes mellitus (T2DM) after MBS is a particularly compelling reason to pursue surgical treatment due to the complications from T2DM that occur over a patient’s lifetime as well as the overall burden of health-related costs. These adverse consequences of T2DM can be mitigated by early use of MBS. MBS is generally well tolerated. Complication rates are similar to adult patients therefore it is recommended to refer patients for MBS whenever they are medically qualified. Most common short-term (<30 days) complications include leak, bleeding, and surgical site infections. Most common long-term (>30 days) complications are nutritional deficiencies.

 

INTRODUCTION

 

The prevalence of worldwide overweight and obesity in adolescents has more than quadrupled since 1975. Currently, it is estimated that over 14 million children age 2-19 years suffer from obesity in the United States alone (1, 2). Adolescents with obesity are at risk for developing significant comorbidities including insulin resistance, type 2 diabetes mellitus (T2DM), hypertension, dyslipidemia, obstructive sleep apnea, nonalcoholic fatty liver disease, depression, polycystic ovarian syndrome, impaired quality of life, cardiovascular disease, and longer term, certain malignancies (3-9). Similar to obesity, the prevalence of T2DM has been increasing dramatically (3). Obesity is a major risk factor the development of T2DM with overweight adolescents having close to a three times greater risk of developing T2DM when compared to adolescents with normal weight (10-12). Additionally, obesity in adolescence is associated with persistent obesity into adulthood, increased risk for obesity related comorbidities, and premature mortality in adulthood (13-15). Lifestyle and medical management remain the first-line treatment for adolescent obesity; however, current evidence suggests that pharmacotherapy, dietary, and behavioral modifications rarely lead to long-term weight loss in adolescents with severe obesity (16-18). The use of metabolic and bariatric surgery (MBS) in adolescents with severe obesity and complications of obesity has been shown to have superior results in both efficacy and durability (19).

 

PREOPERATIVE EVALUATION

 

Multidisciplinary Program

 

A multidisciplinary approach is recommended when considering MBS for an adolescent (20, 21). At a minimum, this includes a bariatric surgeon with adolescent experience, pediatrician, dietitian, nurse, and pediatric psychologist. It is also important the core providers have access to additional pediatric specialists including anesthesiologists, radiologists, and appropriate specialists to aid the management of complications of obesity (e.g., pulmonology, endocrinology, gastroenterology/hepatology). Adolescents undergoing preoperative work-up should be evaluated for the presence and severity of complications of obesity. Additionally, it is important for the multidisciplinary team to determine a potential patient and caregivers’ ability to assess the risks and benefits of surgery as well as to adhere to postoperative requirements including daily vitamin regimens and attending postoperative visits.

 

Patient Selection

 

BODY MASS INDEX (BMI)

 

The following criteria have been recommended by multiple panels of experts for consideration of weight loss surgery in adolescents under 18 years old: (4, 19)

  • BMI ≥ 120 percent of the 95th percentile for BMI for age or BMI ≥ 35kg/m2, whichever is lower, with complications of obesity that has a significant effect on health (Table 1).
  • OR -
  • BMI ≥ 140 percent of the 95th percentile of BMI for age or BMI ≥ 40 kg/m2, whichever is lower

 

Table 1. Qualifying Comorbidities for Consideration of MBS in Adolescents (4).

Obstructive sleep apnea (apnea-hypoxia index > 5)

Type 2 diabetes mellitus

Idiopathic intracranial hypertension

Nonalcoholic steatohepatitis

Blount’s disease

Slipped capital femoral epiphysis

Gastroesophageal Reflux Disease

Hypertension

 

CONTRAINDICATIONS

 

Contraindications to adolescent MBS are listed in Table 2.

 

Table 2. Contraindications to Adolescent MBS

Medically correctable cause of obesity

Ongoing substance abuse problem (within the preceding year)

Medical, psychiatric, psychosocial, or cognitive condition that prevents adherence to postoperative dietary and medication regimens or impairs decisional capacity

Current or planned pregnancy within 18 months of the procedure

Inability for patient or caregivers to comprehend risks and benefits of surgical weight loss procedure

 

AGE

 

A recent retrospective review of the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) data registry from 2015 to 2018 demonstrated that adolescents and young adults only represented 3.7% of total MBS cases performed suggesting significant underutilization within this population (22). Multiple studies have evaluated the safety and efficacy of MBS in younger adolescents. Current evidence suggests there are no significant clinical differences in outcomes between MBS in younger (e.g., <16 years) versus older adolescents (e.g., ≥16 years)(23-27). It is therefore not recommended to limit access to MBS based on patient’s age, physical maturity (e.g., bone age), or pubertal status. These findings have prompted increase advocacy for the use of MBS in the adolescent population by the American Academy of Pediatrics (19).

 

TYPES OF SURGERY

 

Sleeve Gastrectomy

 

A laparoscopic sleeve gastrectomy (SG) results in the removal of the greater curvature of the stomach resulting in a smaller, tubular stomach that has a reduced capacity (Figure 1). Given the procedure is less complex than the Roux-en-Y gastric bypass (RYGB) and has less risk for micronutrient deficiencies, it is an appealing option for adolescents. SG currently accounts for approximately 80% of bariatric procedures in adolescents (22, 28-30). A sleeve gastrectomy may also be converted to RYGB in the event additional MBS is indicated or in the setting of postoperative medically refractory gastroesophageal reflux disease (GERD).

Figure 1. Sleeve Gastrectomy

Roux-en-Y Gastric Bypass

 

Laparoscopic Roux-en-Y gastric bypass involves creating a small, proximal gastric pouch which is separated from the remnant stomach and anastomosed to a Roux-limb of small bowel 70-150cm distally (Figure 2). The RYGB results in similar weight loss when compared to SG and dramatically improves glycemic control (29, 31). The incidence of postoperative GERD is significantly less following RYGB compared to SG, making the procedure an attractive option for adolescents with GERD at baseline (32).

Figure 2. Roux-en-Y Gastric Bypass

 

Others

 

Additional procedures including intragastric balloons are not currently approved by the United States Food and Drug Administration (FDA) for use in adolescents. Adjustable gastric bands have been previously used in the adolescent population; however, they have fallen out of favor due inferior efficacy compared to SG and RYGB (33).

 

POSTOPERATIVE MANAGEMENT

 

Inpatient  

 

Average inpatient stay is typically 1-3 days following both a SG and RYGB (34, 35). Patients are monitored for immediate postoperative complications including a leak, bleeding, and venous thromboembolism (VTE). Following discharge, patients are seen at regular postoperative visits to monitor body weight, nutritional status, and to manage complications of obesity.

 

Diet

 

Following a SG or RYGB, patients are gradually progressed from a high protein liquid diet to incorporating small volumes of regular food. Patients are encouraged to eat three to four protein-rich meals a day while avoiding carbohydrate rich foods. Supplemental sugar-free fluids between meals are also essential following surgery in order to avoid dehydration. Patients are typically encouraged to avoid excessive fluids with meals in order to minimize nausea and maximize nutritional intake with meals due to the restrictive component of both procedures.

 

Postoperative nausea is not uncommon following surgery but typically self resolves. Meals high in carbohydrates or sugar can result in dumping syndrome or weight regain following surgery. Some providers recommend limiting carbonated or caffeinated beverages following MBS based on theoretical concerns, however there is minimal evidence to support this apprehension. Similar to non-operative weight loss recommendations, general recommendations including exercising for 30 to 60 minutes daily, drinking sugar-free fluids, and portion-controlled protein rich meals are the same. Overall, it is recommended that patient and caregiver meet with a dietitian prior to discharge to develop a plan tailored to patient’s specific nutritional needs. Regular follow-up visits with a dietitian are also recommended to assist with postoperative weight management and to monitor for nutritional deficiencies.

 

Nutritional Supplements and Monitoring

 

Although SG may be associated with a decreased risk of nutritional deficiencies when compared to RYGB, lifelong supplementation with vitamins and minerals is recommended following both operations (Table 3). Patients are particularly at risk for deficiencies in iron, vitamin B12, and vitamin D. Additionally, lifelong annual monitoring of nutritional and micronutrient status is recommended with annual laboratory testing (Table 3). Adjustments in supplements may need to be made over time as specific deficiencies emerge. 

 

Table 3. Nutritional Supplementation and Monitoring Recommendations (36)

Nutritional Supplements

Standard multivitamin with folate or iron, or prenatal vitamin if female (once or twice daily)

Vitamin B12, 500mcg sublingually daily, or 1000mcg intramuscularly month

Calcium, 1200 to 1500mg daily (measured as elemental calcium) with 800 to 1000 international units of vitamin D.

Annual Nutritional Monitoring

Complete blood cell count with differential

Serum iron and ferritin

Red blood cell folate, serum vitamin B12, and serum homocysteine

Serum thiamin (vitamin B1)

Hepatic panel (including albumin, total protein, serum aminotransferase levels, gamma-glutamyl transpeptidase, and alkaline phosphatase

Calcium, 25-hydroxyvitamin D, and parathyroid hormone

Dual-energy x-ray absorptiometry (DXA) scan to monitor bone density (optimal frequency not yet established)

 

Pregnancy Prevention

 

Pregnancy should be avoided for 12 to 18 months following MBS to allow patients to achieve weight maintenance and to avoid potential micronutrient deficiencies which may affect both patient and fetus (37). Obesity can result in decreased fertility secondary to irregular menstruation and ovulatory dysfunction (38, 39). Weight loss after MBS has been shown to result in more regular ovulation and improved fertility (40, 41). In a retrospective review of 47 adolescents who underwent MBS surgery, seven pregnancies occurred, six of them within 10 to 22 months following surgery (42). While all six deliveries were healthy and at term, the twofold higher than anticipated pregnancy rate highlights the need for contraception counseling following MBS.

 

Multiple studies have evaluated the efficacy of hormonal contraceptive methods in patients with elevated BMIs and no definitive association was found between higher BMI and effectiveness of hormonal contraceptives (43). Due to concern for malabsorption after intestinal bypass procedures and the subsequent potential for decreased oral contraceptive efficacy, the American College of Obstetricians and Gynecologist recommend using non-oral forms of hormonal contraception in patients who have undergone malabsorptive MBS (44). Additionally, oral contraceptives are associated with increased risk of venous thromboembolism (VTE) which may be worrisome for adolescents with elevated BMIs who already have a higher predisposition for VTE (45, 46).

 

Intrauterine devices (IUDs) are an appealing option following MBS in adolescent patients as they are one of the most effective contraception methods, do not increase risk of VTE, and can be placed at the time of surgery (47). Levonorgestrel-releasing IUDs have the added benefit of promoting amenorrhea which could help reduce the risk of iron deficiency anemia following surgery (48). Regardless of the form of contraception selection, adolescents should be counseled on safe sex practices including the use of barrier protection against sexually transmitted infections.

 

Adolescent patients who become pregnant following MBS should be counseled on adequate nutritional intake with close monitoring of iron, folate, and vitamin B12 levels. Additionally, one must be cautions when screening for gestational diabetes in pregnant patients who have undergone MBS. In a study of a 119 post-bariatric surgery pregnant patients, oral glucose tolerance test resulted in hypoglycemia in 83% of patients with history of RYGB and 55% of patients with history of SG (49). Alternative methods for screening such as capillary blood glucose measurements are therefore recommended (50, 51).

 

Comorbidity Reassessment

 

Regular reassessment of complications of obesity should occur at routine intervals in the postoperative phase to monitor for resolution or need for continued management. Patients with T2DM should be evaluated by their endocrinologist every three months. Repeat polysomnography are generally obtained between three to six months after surgery for patients previously on continuous positive airway pressure therapy (52, 53). Twenty-four-hour blood pressure monitoring can also be repeated three months after surgery to demonstrate resolution or persistence of hypertension. Medication may be restarted if blood pressure is consistently ≥120 mmHg systolic or ≥80 mmHg diastolic. Patients with biopsy proven nonalcoholic fatty liver disease may be re-biopsied 12 months after surgery to document regression. Finally, patients’ mental health needs should be re-evaluated by a pediatric psychologist at 6 and 12 months after surgery.

 

In the setting of weight regain, patients should be monitored for complications of obesity. There is emerging evidence however, that some complications of obesity may be weight dependent and others non-weight dependent (54). Some surgeons will routinely obtain an upper gastrointestinal contrast study at 12 months after surgery or as needed to assess anatomy which may lead to weight regain. Anatomical abnormalities that may contribute to weight regain include a dilated gastric sleeve or gastrogastric fistula.

 

Follow Up

 

Close follow up with the multidisciplinary team including the bariatric surgeon, pediatrician, dietitian, and pediatric psychologist is strongly recommended. Patients are typically followed by a pediatrician to ensure ongoing continuity of care. It is important for the core providers to have access to pediatric specialists including endocrinology, gastroenterology/hepatology, and pulmonology as needed in those with complications of obesity that require ongoing monitoring or management. Additionally, a gynecologist for contraception counseling may be required for female patients. The transition from pediatric to adult medicine can be challenging in patients with chronic medical conditions and frequently requires assistance from multiple members of the team for transition care coordination and preparation as well as to ensure adequate communication, support, and education (55-57). 

 

OUTCOMES

 

Percent BMI Loss

 

Both SG and RYGB have resulted in clinically significant weight loss in adolescents. The efficacy of both procedures appears to be similar in the adolescent population with potentially slightly greater weight loss following RYBG. In a large, multicenter analysis of 177 adolescents who underwent RYGB and 306 adolescents who underwent SG, there was a three-year postoperative average percent BMI loss of -29% (95% CI, -26 to -33) and -25% (95% CI, -22 to -28) for RYGB and SG, respectively (29). Similar results were seen in the largest prospective study to date of 228 adolescents undergoing either RYGB or SG with an average 28% reduction in BMI at three years following RYGB compared to 26% reduction following SG (53). Smaller, long-term studies demonstrate the durability of weight loss with MBS with an average percent BMI loss of 29% in adolescents undergoing RYGB up to 12 years after surgery(58, 59). 

 

Complications of Obesity

 

TYPE 2 DIABETES MELLITUS  

 

Multiple studies have demonstrated improved glycemic control, even remission as well as prevention of T2DM following MBS, making a compelling case of MBS as a treatment for T2DM (31, 59-63). The Teen-Longitudinal Assessment of Bariatric Surgery (Teen-LABS) is a prospective, observational study of pediatric patients undergoing MBS at 5 children’s hospitals in the United States with 3 and 5 year of follow-up data published to date. Of the 242 adolescents with obesity who underwent MBS, 29 had T2DM. By 3 years after the procedure, remission of T2DM occurred in 95% (95% CI, 85-100) of participates who had diabetes at baseline with no new cases of T2DM in those without the condition at baseline (53). Additionally, 19 participants had prediabetes at baseline with a 76% (95% CI, 56-97) rate of remission at 3 years (53). These remission rates in Teen-LABS were compared to adults who underwent MBS. Among those who underwent RYGB, adolescents were more likely to have remission of T2DM at 5 years with a remission rate of 86% compared to 53% in adults (64).

 

Similar findings were demonstrated in another study of 226 adolescents undergoing SG, of which 23% of patients were found to have T2DM. Eighty-five percent of patients with T2DM were on medication for diabetes prior to surgery and 89% achieved normal fasting plasma glucose and hemoglobin A1c levels without the use of medication postoperatively (52).

 

In an effort to compare surgical versus medical therapy for T2DM in adolescents with severe obesity, data from participants with T2DM enrolled in the Teen-LABS study were compared to participants of similar age and racial distribution from the Treatment Options of Type 2 Diabetes in Adolescents and Youth (TODAY) studies. Teen-LABS participants underwent MBS. TODAY participants were randomized to metformin alone or in combination with rosiglitazone or intensive lifestyle intervention, with insulin therapy given for glycemic progression. At two years, mean hemoglobin A1c concentration decreased from 6.8% to 5.5% in patients who underwent MBS compared to an increase from 6.4% to 7.8% in those enrolled in the TODAY study. Compared to baseline, average BMI decreased by 29% in Teen-LABS participants while the average BMI increased by 3.7% in TODAY participants (65). Cardiovascular disease (CVD) risk reduction was also explored in a secondary analysis of this study and despite higher pretreatment risk for CVD, treatment with MBS resulted in reduction of estimated CVD that were sustained at 5-year follow-up where medical therapy was associated with an increase in risk of CVD in adolescents with T2DM and severe obesity (66).

 

While these initial results are promising of the beneficial effects of MBS for the treatment of T2DM, no studies have prospectively compared the efficacy of MBS with that of medical therapy for the treatment of T2DM in adolescents with obesity. Additionally, the majority of initial MBS data in adolescents were from those who underwent RYGB which is no longer the primary MBS procedure performed in adolescents due to its inferior safety profile. In 2019, the National Institute of Health funded the Surgical or Medical Treatment for Pediatric T2DM (ST2OMP) trial which will compare SG to advanced medical therapy (67, 68).

 

OTHER COMORBIDITIES

 

In the Teen-LABS study described above, a mean 74% (95% CI, 64 to 84) remission of hypertension (HTN), 66% (95% CI 57 to 74) remission of dyslipidemia, and 86% (95% CI 72 to 100) resolution of abnormal kidney function was found at 3 years (53). In a secondary analysis of Teen-LABS and TODAY data, medical management of adolescents with obesity was associated with higher odds of diabetic kidney disease when compared to MBS (69). Greater weight loss after MBS in adolescents has also been associated with greater remission of T2DM, HTN, and dyslipidemia (54, 70). In a comparison of adolescents and adults who underwent RYGB, adolescents were more likely to have remission of HTN at 5 years compared to adults (68% vs 41%) (64).

 

Additional studies have demonstrated a 66% to 84% remission of obstructive sleep apnea as well as improvements in liver disease and polycystic ovarian syndrome (8, 52, 71). Improvements in functional mobility as well as reduction in musculoskeletal pain have also been well described (72, 73).

 

Mental Health

 

Multiple studies have reported higher rates of depression, emotional and behavioral disorders, and suicidal ideation in adolescents with obesity (74-77). Additionally, binge and loss of control eating is prevalent among more than one quarter of adolescents with overweight and obesity (78, 79). A recent prospective study demonstrated that undergoing MBS in adolescence did not heighten or lower the risk of suicidal thoughts or behaviors following the initial 4 years after surgery (80). While still unclear whether obesity leads to psychopathology, or vice versa, the association highlights the need for appropriate psychological services in the pre- and postoperative period (74).

 

MBS can lead to improvements in psychosocial outcomes, although the improvements appear too often be transient. In the TEEN-Labs study, quality of life measured by the Impact of Weight on Quality of Life and Short Form 36 Health Survey improved after MBS (53, 73). Several studies have demonstrated improved depressive and anxiety symptoms in the months following MBS, although the results were not maintained after the first postoperative year (81, 82). In a multisite study assessing two year follow up of psychopathology prevalence in adolescents undergoing MBS, most patients retained their symptomatic or non-symptomatic psychopathology status at two years, although remission of symptoms was more prevalent than the development of new symptoms (83). These results emphasize the need for long-term psychosocial monitoring following MBS as well as early treatment in those with psychopathology

 

Short-Term Complications

 

Short-term complications (<30 days after surgery) in adolescents undergoing MBS are similar to those seen in adults. Early postoperative complications, though rare, include surgical site infections, bleeding, leak, strictures, and pulmonary embolism. In a retrospective review of 21,592 adolescents and young adults who underwent SG or RYGB between 2015 and 2018, 3.7% of patients required readmission, 1.1% of patients required reoperation, and 3.3% required percutaneous, endoscopic, or other intervention (22). Major complications were rare; the most common complication was bleeding (0.4%), followed by leak (0.4%), and deep surgical site infections (0.2%). RYGB was associated with higher rates of reoperation (2.1% vs. 0.8%), readmission (6.3% vs. 3.0%), and serious complications (5.5% vs. 1.8%) compared to SG. Mortality occurred in 0.05% of patients and there were no differences in mortality noted between groups (22). In an additional retrospective review of 483 adolescents (SG n=306, RYGB n=177) no perioperative deaths occurred and the rate of major adverse events were too rare for statistical comparison. VTEs occurred in only 0.4% of patients and failure to discharge in 30 days was observed in 0.7% of patients (29).

 

Multiple studies have also suggested that MBS may be safer in adolescents when compared with adults. In a large study evaluating perioperative outcomes of MBS between 309 adolescents and 55,192 adults, the overall 30-day complication rate was significantly lower in adolescents (5.5%) as compared with adults (9.8%). No in-hospital mortalities occurred in the adolescent group compared to 0.2% the adult group. The 30-day morbidity for adolescents following SG was zero compared to 4.3% following RYGB (84). In an additional study evaluating 1047 adolescents,10,429 college-aged individuals, and 24,841 young adults who underwent SG or RYGB, there were no differences in 30-day complication rates between age groups (85).

 

Long-Term Complications

 

NUTRITIONAL DEFICIENCIES  

 

Long-term complications after MBS in adolescents are primarily nutritional. Patients are particularly at risk for deficiencies in iron, vitamin B12, and vitamin D. Iron deficiency is common in premenopausal females due to menstruation. Some patients may require iron infusion if oral supplementation is not adequate. Symptomatic thiamine deficiency following MBS is rare, however can have serious consequences (86-88).These risks are higher for patients who undergo RYGB compared to SG due to potential malabsorption. In a Teen-LABS study evaluating nutritional deficiencies at 5 years postoperatively, low serum ferritin levels were seen in 71% of patients who underwent RYGB compared to 45% following a SG indicating iron deficiency (86). Iron deficiency anemia can occasionally be severe in adolescent women following MBS which can be compounded by menstruation and challenges in recognizing symptoms therefore daily supplementation and routine nutritional monitoring is essential following MBS.

 

Vitamin B12 deficiency was seen in approximately 12% of patients after either procedure. Approximately 40% of patients had low vitamin D levels at baseline with no significant change at follow up. Parathyroid hormone concentrations increased in patients undergoing RYGB from an average baseline concentration of 44 pg/ml to 59 pg/ml at 5 years with the risk of abnormal parathyroid hormone nearly sixfold higher after RYGB compared with SG (86). Elevated parathyroid hormone is utilized as a surrogate for calcium deficiency and raises concerns about long-term bone health. In adolescents, reduced bone mass has been noted two years after MBS although the bone mass remains appropriate for the patients’ age and new body weight (89). Concerns of growth retardation after MBS have been refuted and the most recent adolescent ASMBS guidelines have removed the recommendation of patients reaching physical maturity prior to MBS (4, 24).

 

The risk of nutritional deficiencies decreases with adherence to prescribed micronutrient supplements and increases with pregnancy (86). Given the high prevalence of nutritional deficiencies, lifelong micronutrient supplementation is required following surgery. One concern emphasized in the adolescent population is adherence to regular multivitamin use. In a prospective study of 41 adolescents who underwent MBS, multivitamin adherence was only 29.8%  23.9 (90).

 

WEIGHT REGAIN

 

Current data demonstrates adequate maintenance of weight loss at 5-9 years (29, 91). Several studies have suggested a long-term weight loss advantage in adolescents undergoing RYGB compared to SG, although there is still insufficient evidence to directly compare long-term outcomes of both procedures (29, 53, 92, 93). More research is needed to fully understand the mechanisms behind long-term weight maintenance after MBS.

 

OTHER COMPLICATIONS  

 

Cholelithiasis is a common complication due to rapid weight loss following MBS in both adolescents and adults. In the Teen-LABS study, cholecystectomy was required within three years in 9.9% of adolescents who underwent RYGB and 5.1% who underwent SG (53). Five percent of Teen-LABS participants required other abdominal operations including lysis of adhesions, gastrostomy, ventral hernia repair, or internal hernia repair (53). Symptoms of GERD, nausea, bloating, and diarrhea can also increase following MBS. During five years of follow up, the incidence of GERD increased from 2% to 8% in adolescents who underwent RYGB and from 11% to 24% in those who underwent SG. At five years postoperatively, the SG group had more than fourfold greater odds of having gastrointestinal distress symptoms when compared to RYGB (32). Dumping syndrome can been seen after both procedures, however it is much more common after RYGB compared to SG (94, 95). The incidence of dumping syndrome (~12%) in adolescents after RYGB was similar to adult patients two years after surgery(96).

 

There are no current established guidelines for surveillance of Barrett’s esophagus after SG for adolescent patients, however routine screening is recommended for adult patients after SG, therefore it would be prudent for adolescent patients to undergo intermittent surveillance also as the length of possible GERD exposure is theoretically longer (97). Similarly, there are no established guidelines for monitoring of bone density following use of MBS in adolescence, but due to inadequate vitamin D levels and rising PTH at least in those who underwent RYGB, periodic monitoring with DEXA may be prudent.

 

Emerging Evidence

 

Current evidence evaluating the outcomes and efficacy of adolescent MBS is generally limited to ≤10 years of follow up. Smaller, long-term studies with data available for 7 to 12 post-operatively in patients who primary underwent RYGB demonstrate the durability of weight loss and similar rates of complications, although inference is limited due to small sample sizes with reduced attrition rates (59, 60, 98, 99). Characteristics including study size, length of follow up, and attrition rate of available studies on MBS published from 2012 to present are available in Table 4. As SG is now the most predominate MBS procedure performed in the Unites States long-term data with this procedure is required. While some longitudinal studies are ongoing (Table 4), there remains a paucity of long-term data in the adolescent population.

 

Table 4. Characteristics of Studies on MBS, 2012 – Present

Author; Year

Study Design

Sample Size (N)

Type of MBS

 

 

RYBG     SG

Longest follow up

N

(1 yr)

N

(3 yr)

N

(5 yr)

Comments

Inge;

2018 (29)

RO 

483

177

306

5 yr

466 (96%)

153 (32%)

41 (8%)

The PCORnet bariatric study (2005 – 2015)

Olbers; 2017 (58)

CC

81

81

0

5 yr

81

(100%)

n/a

81 (100%)

Adolescent Morbid Obesity Surgery (AMOS) study

Inge; 2017(59)

PO

74

74

0

12.5 yr

n/a

n/a

58 (81%)

Adolescent Bariatric Surgery at 5 Plus Years (FABS-5+) study (2001-2007); mean follow up 8.0 yr

Inge; 2016 (53)

PO 

228

161

67

3 yr

205 (90%)

194 (85%)

n/a

Teen-Longitudinal Assessment of Bariatric Surgery (Teen-LABS) study; (2007-2012)

Vilallonga; 2016 (60)

RO

19

19

0

10.2 yr

n/a

n/a

n/a

Mean follow up 7.2 years; (2003-2008)

Al-Sabah; 2015 (61)

RO

125

0

135

4 yr

54 (40%)

n/a

n/a

2 yr follow up: 46 (34%); (2008-2012)

Cozacov; 2014 (98)

RO

18

8

10

7 yr

15 (83%)

10 (56%)

n/a

7 yr follow up: 3 (17%); (2002 – 2011)

Messiah; 2013 (71)

PO

454

454

0

1 yr

108 (24%)

n/a

n/a

Bariatric Outcomes Longitudinal Database (BOLD) (2004-2010)

Alqahtani; 2012 (25)

RO

108

0

108

2 yr

41 (38%)

n/a

n/a

2 yr follow-up: 8 (7%); (2008 – 2011)

Nijhawan; 2012 (99)

RO

25

25

0

9 yr

n/a

n/a

20 (80%)

Study dates not provided

RO- Retrospective observational; CC- Case-control; PO- Prospective observational

 

DISCUSSION

 

Surgical weight loss is an appropriate consideration for adolescents with severe obesity and/or complications of obesity who have failed to lose weight through conservative management. It is essential that adolescents undergoing evaluation for MBS due so in the context of a multidisciplinary program with specific expertise in adolescent medicine and MBS. SG and RYGB are safe and effective treatment options in adolescents. Weight loss outcomes are comparable between SG and RYGB. Both procedures also result in substantial improvement in complications of obesity, including T2DM. SG appears to have an improved safety profile when compared to RYGB and is now the most common adolescent bariatric procedure performed in the United States. Emerging evidence demonstrates advantages of earlier surgical intervention in those with obesity including improved weight loss, increased resolution of comorbidities, and decreased adverse events when compared to adults (64, 100). Perioperative complications in adolescents undergoing MBS are similar to those in adults but occur less frequently (84, 85). Long-term complications are primarily nutritional and life-long vitamin and mineral supplementation is recommended. Regular follow up is required following MBS and it is important for patients to have access to appropriate medical, dietary, and psychological care.

 

REFERENCES

 

  1. Obesity and overweight World Health Organization2020 [Available from: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight.
  2. Childhood Obesity Facts Centers for Disease Control and Prevention: Centers for Disease Control and Prevention; 2021 [Available from: https://www.cdc.gov/obesity/data/childhood.html.
  3. Rosenbaum M. Special Considerations Relevant to Pediatric Obesity. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA)2000.
  4. Pratt JSA, Browne A, Browne NT, Bruzoni M, Cohen M, Desai A, et al. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg Obes Relat Dis. 2018;14(7):882-901.
  5. Xanthakos SA, Jenkins TM, Kleiner DE, Boyce TW, Mourya R, Karns R, et al. High Prevalence of Nonalcoholic Fatty Liver Disease in Adolescents Undergoing Bariatric Surgery. Gastroenterology. 2015;149(3):623-34.e8.
  6. Zeller MH, Roehrig HR, Modi AC, Daniels SR, Inge TH. Health-related quality of life and depressive symptoms in adolescents with extreme obesity presenting for bariatric surgery. Pediatrics. 2006;117(4):1155-61.
  7. Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, et al. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2013;98(12):4565-92.
  8. Kaar JL, Morelli N, Russell SP, Talker I, Moore JM, Inge TH, et al. Obstructive sleep apnea and early weight loss among adolescents undergoing bariatric surgery. Surg Obes Relat Dis. 2021;17(4):711-7.
  9. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 2019;92:121-35.
  10. Tirosh A, Shai I, Afek A, Dubnov-Raz G, Ayalon N, Gordon B, et al. Adolescent BMI trajectory and risk of diabetes versus coronary disease. N Engl J Med. 2011;364(14):1315-25.
  11. Abdullah A, Stoelwinder J, Shortreed S, Wolfe R, Stevenson C, Walls H, et al. The duration of obesity and the risk of type 2 diabetes. Public Health Nutr. 2011;14(1):119-26.
  12. Lee JM, Gebremariam A, Vijan S, Gurney JG. Excess body mass index-years, a measure of degree and duration of excess weight, and risk for incident diabetes. Arch Pediatr Adolesc Med. 2012;166(1):42-8.
  13. Fontaine KR, Redden DT, Wang C, Westfall AO, Allison DB. Years of life lost due to obesity. Jama. 2003;289(2):187-93.
  14. Greenberg JA. Obesity and early mortality in the United States. Obesity (Silver Spring). 2013;21(2):405-12.
  15. Inge TH, King WC, Jenkins TM, Courcoulas AP, Mitsnefes M, Flum DR, et al. The effect of obesity in adolescence on adult health status. Pediatrics. 2013;132(6):1098-104.
  16. Berkowitz RI, Wadden TA, Gehrman CA, Bishop-Gilyard CT, Moore RH, Womble LG, et al. Meal replacements in the treatment of adolescent obesity: a randomized controlled trial. Obesity (Silver Spring). 2011;19(6):1193-9.
  17. Andela S, Burrows TL, Baur LA, Coyle DH, Collins CE, Gow ML. Efficacy of very low-energy diet programs for weight loss: A systematic review with meta-analysis of intervention studies in children and adolescents with obesity. Obes Rev. 2019;20(6):871-82.
  18. Srivastava G, Fox CK, Kelly AS, Jastreboff AM, Browne AF, Browne NT, et al. Clinical Considerations Regarding the Use of Obesity Pharmacotherapy in Adolescents with Obesity. Obesity (Silver Spring). 2019;27(2):190-204.
  19. Armstrong SC, Bolling CF, Michalsky MP, Reichard KW. Pediatric Metabolic and Bariatric Surgery: Evidence, Barriers, and Best Practices. Pediatrics. 2019;144(6).
  20. Inge TH, Krebs NF, Garcia VF, Skelton JA, Guice KS, Strauss RS, et al. Bariatric surgery for severely overweight adolescents: concerns and recommendations. Pediatrics. 2004;114(1):217-23.
  21. Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, et al. Pediatric Obesity-Assessment, Treatment, and Prevention: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017;102(3):709-57.
  22. Mocanu V, Lai K, Dang JT, Switzer NJ, Birch DW, Ball GDC, et al. Evaluation of the Trends, Characteristics, and Outcomes in North American Youth Undergoing Elective Bariatric Surgery. Obes Surg. 2021;31(5):2180-7.
  23. Ogle SB, Dewberry LC, Jenkins TM, Inge TH, Kelsey M, Bruzoni M, et al. Outcomes of Bariatric Surgery in Older Versus Younger Adolescents. Pediatrics. 2021;147(3).
  24. Alqahtani A, Elahmedi M, Qahtani AR. Laparoscopic Sleeve Gastrectomy in Children Younger Than 14 Years: Refuting the Concerns. Ann Surg. 2016;263(2):312-9.
  25. Alqahtani AR, Antonisamy B, Alamri H, Elahmedi M, Zimmerman VA. Laparoscopic sleeve gastrectomy in 108 obese children and adolescents aged 5 to 21 years. Ann Surg. 2012;256(2):266-73.
  26. El Chaar M, King K, Al-Mardini A, Galvez A, Claros L, Stoltzfus J. Thirty-Day Outcomes of Bariatric Surgery in Adolescents: a First Look at the MBSAQIP Database. Obes Surg. 2021;31(1):194-9.
  27. Poliakin L, Roberts A, Thompson KJ, Raheem E, McKillop IH, Nimeri A. Outcomes of adolescents compared with young adults after bariatric surgery: an analysis of 227,671 patients using the MBSAQIP data registry. Surg Obes Relat Dis. 2020;16(10):1463-73.
  28. Alqahtani A, Alamri H, Elahmedi M, Mohammed R. Laparoscopic sleeve gastrectomy in adult and pediatric obese patients: a comparative study. Surg Endosc. 2012;26(11):3094-100.
  29. Inge TH, Coley RY, Bazzano LA, Xanthakos SA, McTigue K, Arterburn D, et al. Comparative effectiveness of bariatric procedures among adolescents: the PCORnet bariatric study. Surg Obes Relat Dis. 2018;14(9):1374-86.
  30. Jackson WL, Lewis SR, Bagby JP, Hilton LR, Milad M, Bledsoe SE. Laparoscopic sleeve gastrectomy versus laparoscopic Roux-en-Y gastric bypass in the pediatric population: a MBSAQIP analysis. Surg Obes Relat Dis. 2020;16(2):254-60.
  31. Inge TH, Miyano G, Bean J, Helmrath M, Courcoulas A, Harmon CM, et al. Reversal of type 2 diabetes mellitus and improvements in cardiovascular risk factors after surgical weight loss in adolescents. Pediatrics. 2009;123(1):214-22.
  32. Dewberry LC, Khoury JC, Ehrlich S, Jenkins TM, Beamish AJ, Kalkwarf HJ, et al. Change in gastrointestinal symptoms over the first 5 years after bariatric surgery in a multicenter cohort of adolescents. J Pediatr Surg. 2019;54(6):1220-5.
  33. Dewberry LC, Jalivand A, Gupta R, Jenkins TM, Beamish A, Inge TH, et al. Weight Loss and Health Status 5 Years After Adjustable Gastric Banding in Adolescents. Obes Surg. 2020;30(6):2388-94.
  34. Cummins CB, Nunez Lopez O, Hughes BD, Adhikari D, Guidry CA, Stubbs S, et al. Adolescent Bariatric Surgery: Effects of Socioeconomic, Demographic, and Hospital Characteristics on Cost, Length of Stay, and Type of Procedure Performed. Obes Surg. 2019;29(3):757-64.
  35. Kyler KE, Bettenhausen JL, Hall M, Fraser JD, Sweeney B. Trends in Volume and Utilization Outcomes in Adolescent Metabolic and Bariatric Surgery at Children's Hospitals. J Adolesc Health. 2019;65(3):331-6.
  36. Mechanick JI, Apovian C, Brethauer S, Timothy Garvey W, Joffe AM, Kim J, et al. Clinical Practice Guidelines for the Perioperative Nutrition, Metabolic, and Nonsurgical Support of Patients Undergoing Bariatric Procedures - 2019 Update: Cosponsored by American Association of Clinical Endocrinologists/American College of Endocrinology, The Obesity Society, American Society for Metabolic and Bariatric Surgery, Obesity Medicine Association, and American Society of Anesthesiologists. Obesity (Silver Spring). 2020;28(4):O1-o58.
  37. Mechanick JI, Youdim A, Jones DB, Garvey WT, Hurley DL, McMahon MM, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient--2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring). 2013;21 Suppl 1(0 1):S1-27.
  38. Broughton DE, Moley KH. Obesity and female infertility: potential mediators of obesity's impact. Fertil Steril. 2017;107(4):840-7.
  39. Silvestris E, de Pergola G, Rosania R, Loverro G. Obesity as disruptor of the female fertility. Reprod Biol Endocrinol. 2018;16(1):22.
  40. Teitelman M, Grotegut CA, Williams NN, Lewis JD. The impact of bariatric surgery on menstrual patterns. Obes Surg. 2006;16(11):1457-63.
  41. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, Botella Carretero JI. Prevalence of 'obesity-associated gonadal dysfunction' in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update. 2017;23(4):390-408.
  42. Roehrig HR, Xanthakos SA, Sweeney J, Zeller MH, Inge TH. Pregnancy after gastric bypass surgery in adolescents. Obes Surg. 2007;17(7):873-7.
  43. Lopez LM, Bernholc A, Chen M, Grey TW, Otterness C, Westhoff C, et al. Hormonal contraceptives for contraception in overweight or obese women. Cochrane Database Syst Rev. 2016(8):Cd008452.
  44. ACOG practice bulletin no. 105: bariatric surgery and pregnancy. Obstet Gynecol. 2009;113(6):1405-13.
  45. de Bastos M, Stegeman BH, Rosendaal FR, Van Hylckama Vlieg A, Helmerhorst FM, Stijnen T, et al. Combined oral contraceptives: venous thrombosis. Cochrane Database Syst Rev. 2014(3):Cd010813.
  46. Lassandro G, Palmieri VV, Palladino V, Amoruso A, Faienza MF, Giordano P. Venous Thromboembolism in Children: From Diagnosis to Management. Int J Environ Res Public Health. 2020;17(14).
  47. ACOG Committee Opinion No. 735: Adolescents and Long-Acting Reversible Contraception: Implants and Intrauterine Devices. Obstet Gynecol. 2018;131(5):e130-e9.
  48. Apter D. International Perspectives: IUDs and Adolescents. J Pediatr Adolesc Gynecol. 2019;32(5s):S36-s42.
  49. Rottenstreich A, Elazary R, Ezra Y, Kleinstern G, Beglaibter N, Elchalal U. Hypoglycemia during oral glucose tolerance test among post-bariatric surgery pregnant patients: incidence and perinatal significance. Surg Obes Relat Dis. 2018;14(3):347-53.
  50. Benhalima K, Minschart C, Ceulemans D, Bogaerts A, Van Der Schueren B, Mathieu C, et al. Screening and Management of Gestational Diabetes Mellitus after Bariatric Surgery. Nutrients. 2018;10(10).
  51. Minschart C, Beunen K, Benhalima K. An Update on Screening Strategies for Gestational Diabetes Mellitus: A Narrative Review. Diabetes Metab Syndr Obes. 2021;14:3047-76.
  52. Alqahtani AR, Elahmedi MO, Al Qahtani A. Co-morbidity resolution in morbidly obese children and adolescents undergoing sleeve gastrectomy. Surg Obes Relat Dis. 2014;10(5):842-50.
  53. Inge TH, Courcoulas AP, Jenkins TM, Michalsky MP, Helmrath MA, Brandt ML, et al. Weight Loss and Health Status 3 Years after Bariatric Surgery in Adolescents. N Engl J Med. 2016;374(2):113-23.
  54. Derderian SC, Patten L, Kaizer AM, Moore JM, Ogle S, Jenkins TM, et al. Influence of Weight Loss on Obesity-Associated Complications After Metabolic and Bariatric Surgery in Adolescents. Obesity (Silver Spring). 2020;28(12):2397-404.
  55. Wafa S, Nakhla M. Improving the Transition from Pediatric to Adult Diabetes Healthcare: A Literature Review. Can J Diabetes. 2015;39(6):520-8.
  56. Coyne I, Sheehan A, Heery E, While AE. Healthcare transition for adolescents and young adults with long-term conditions: Qualitative study of patients, parents and healthcare professionals' experiences. J Clin Nurs. 2019;28(21-22):4062-76.
  57. Butalia S, McGuire KA, Dyjur D, Mercer J, Pacaud D. Youth with diabetes and their parents' perspectives on transition care from pediatric to adult diabetes care services: A qualitative study. Health Sci Rep. 2020;3(3):e181.
  58. Olbers T, Beamish AJ, Gronowitz E, Flodmark CE, Dahlgren J, Bruze G, et al. Laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity (AMOS): a prospective, 5-year, Swedish nationwide study. Lancet Diabetes Endocrinol. 2017;5(3):174-83.
  59. Inge TH, Jenkins TM, Xanthakos SA, Dixon JB, Daniels SR, Zeller MH, et al. Long-term outcomes of bariatric surgery in adolescents with severe obesity (FABS-5+): a prospective follow-up analysis. Lancet Diabetes Endocrinol. 2017;5(3):165-73.
  60. Vilallonga R, Himpens J, van de Vrande S. Long-Term (7 Years) Follow-Up of Roux-en-Y Gastric Bypass on Obese Adolescent Patients (<18 Years). Obes Facts. 2016;9(2):91-100.
  61. Al-Sabah SK, Almazeedi SM, Dashti SA, Al-Mulla AY, Ali DA, Jumaa TH. The efficacy of laparoscopic sleeve gastrectomy in treating adolescent obesity. Obes Surg. 2015;25(1):50-4.
  62. Syn NL, Cummings DE, Wang LZ, Lin DJ, Zhao JJ, Loh M, et al. Association of metabolic-bariatric surgery with long-term survival in adults with and without diabetes: a one-stage meta-analysis of matched cohort and prospective controlled studies with 174 772 participants. Lancet. 2021;397(10287):1830-41.
  63. Stefater MA, Inge TH. Bariatric Surgery for Adolescents with Type 2 Diabetes: an Emerging Therapeutic Strategy. Curr Diab Rep. 2017;17(8):62.
  64. Inge TH, Courcoulas AP, Jenkins TM, Michalsky MP, Brandt ML, Xanthakos SA, et al. Five-Year Outcomes of Gastric Bypass in Adolescents as Compared with Adults. N Engl J Med. 2019;380(22):2136-45.
  65. Inge TH, Laffel LM, Jenkins TM, Marcus MD, Leibel NI, Brandt ML, et al. Comparison of Surgical and Medical Therapy for Type 2 Diabetes in Severely Obese Adolescents. JAMA Pediatr. 2018;172(5):452-60.
  66. Ryder JR, Xu P, Nadeau KJ, Kelsey MM, Xie C, Jenkins T, et al. Effect of surgical versus medical therapy on estimated cardiovascular event risk among adolescents with type 2 diabetes and severe obesity. Surg Obes Relat Dis. 2021;17(1):23-33.
  67. Shah AS, Nadeau KJ, Helmrath MA, Inge TH, Xanthakos SA, Kelsey MM. Metabolic outcomes of surgery in youth with type 2 diabetes. Semin Pediatr Surg. 2020;29(1):150893.
  68. Shah AS, Helmrath MA, Inge TH, Xanthakos SA, Kelsey MM, Jenkins T, et al. Study protocol: a prospective controlled clinical trial to assess surgical or medical treatment for paediatric type 2 diabetes (ST(2)OMP). BMJ Open. 2021;11(8):e047766.
  69. Bjornstad P, Hughan K, Kelsey MM, Shah AS, Lynch J, Nehus E, et al. Effect of Surgical Versus Medical Therapy on Diabetic Kidney Disease Over 5 Years in Severely Obese Adolescents With Type 2 Diabetes. Diabetes Care. 2020;43(1):187-95.
  70. Michalsky MP, Inge TH, Jenkins TM, Xie C, Courcoulas A, Helmrath M, et al. Cardiovascular Risk Factors After Adolescent Bariatric Surgery. Pediatrics. 2018;141(2).
  71. Messiah SE, Lopez-Mitnik G, Winegar D, Sherif B, Arheart KL, Reichard KW, et al. Changes in weight and co-morbidities among adolescents undergoing bariatric surgery: 1-year results from the Bariatric Outcomes Longitudinal Database. Surg Obes Relat Dis. 2013;9(4):503-13.
  72. Ryder JR, Edwards NM, Gupta R, Khoury J, Jenkins TM, Bout-Tabaku S, et al. Changes in Functional Mobility and Musculoskeletal Pain After Bariatric Surgery in Teens With Severe Obesity: Teen-Longitudinal Assessment of Bariatric Surgery (LABS) Study. JAMA Pediatr. 2016;170(9):871-7.
  73. Bout-Tabaku S, Gupta R, Jenkins TM, Ryder JR, Baughcum AE, Jackson RD, et al. Musculoskeletal Pain, Physical Function, and Quality of Life After Bariatric Surgery. Pediatrics. 2019;144(6).
  74. Burton ET, Mackey ER, Reynolds K, Cadieux A, Gaffka BJ, Shaffer LA. Psychopathology and Adolescent Bariatric Surgery: A Topical Review to Support Psychologists in Assessment and Treatment Considerations. J Clin Psychol Med Settings. 2020;27(2):235-46.
  75. Kalarchian MA, Marcus MD. Psychiatric comorbidity of childhood obesity. Int Rev Psychiatry. 2012;24(3):241-6.
  76. Rankin J, Matthews L, Cobley S, Han A, Sanders R, Wiltshire HD, et al. Psychological consequences of childhood obesity: psychiatric comorbidity and prevention. Adolesc Health Med Ther. 2016;7:125-46.
  77. Zeller MH, Reiter-Purtill J, Jenkins TM, Ratcliff MB. Adolescent suicidal behavior across the excess weight status spectrum. Obesity (Silver Spring). 2013;21(5):1039-45.
  78. He J, Cai Z, Fan X. Prevalence of binge and loss of control eating among children and adolescents with overweight and obesity: An exploratory meta-analysis. Int J Eat Disord. 2017;50(2):91-103.
  79. Goldschmidt AB, Khoury JC, Mitchell JE, Jenkins TM, Bond DS, Zeller MH, et al. Loss of Control Eating and Health Indicators Over 6 Years in Adolescents Undergoing Metabolic and Bariatric Surgery. Obesity (Silver Spring). 2021;29(4):740-7.
  80. Zeller MH, Reiter-Purtill J, Jenkins TM, Kidwell KM, Bensman HE, Mitchell JE, et al. Suicidal thoughts and behaviors in adolescents who underwent bariatric surgery. Surg Obes Relat Dis. 2020;16(4):568-80.
  81. Järvholm K, Olbers T, Marcus C, Mårild S, Gronowitz E, Friberg P, et al. Short-term psychological outcomes in severely obese adolescents after bariatric surgery. Obesity (Silver Spring). 2012;20(2):318-23.
  82. Zeller MH, Reiter-Purtill J, Ratcliff MB, Inge TH, Noll JG. Two-year trends in psychosocial functioning after adolescent Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2011;7(6):727-32.
  83. Hunsaker SL, Garland BH, Rofey D, Reiter-Purtill J, Mitchell J, Courcoulas A, et al. A Multisite 2-Year Follow Up of Psychopathology Prevalence, Predictors, and Correlates Among Adolescents Who Did or Did Not Undergo Weight Loss Surgery. J Adolesc Health. 2018;63(2):142-50.
  84. Varela JE, Hinojosa MW, Nguyen NT. Perioperative outcomes of bariatric surgery in adolescents compared with adults at academic medical centers. Surg Obes Relat Dis. 2007;3(5):537-40; discussion 41-2.
  85. Grant HM, Perez-Caraballo A, Romanelli JR, Tirabassi MV. Metabolic and bariatric surgery is likely safe, but underutilized in adolescents aged 13-17 years. Surg Obes Relat Dis. 2021;17(6):1146-51.
  86. Xanthakos SA, Khoury JC, Inge TH, Jenkins TM, Modi AC, Michalsky MP, et al. Nutritional Risks in Adolescents After Bariatric Surgery. Clin Gastroenterol Hepatol. 2020;18(5):1070-81.e5.
  87. Armstrong-Javors A, Pratt J, Kharasch S. Wernicke Encephalopathy in Adolescents After Bariatric Surgery: Case Report and Review. Pediatrics. 2016;138(6).
  88. Towbin A, Inge TH, Garcia VF, Roehrig HR, Clements RH, Harmon CM, et al. Beriberi after gastric bypass surgery in adolescence. J Pediatr. 2004;145(2):263-7.
  89. Kaulfers AM, Bean JA, Inge TH, Dolan LM, Kalkwarf HJ. Bone loss in adolescents after bariatric surgery. Pediatrics. 2011;127(4):e956-61.
  90. Modi AC, Zeller MH, Xanthakos SA, Jenkins TM, Inge TH. Adherence to vitamin supplementation following adolescent bariatric surgery. Obesity (Silver Spring). 2013;21(3):E190-5.
  91. Elhag W, El Ansari W. Durability of Cardiometabolic Outcomes Among Adolescents After Sleeve Gastrectomy: First Study with 9-Year Follow-up. Obes Surg. 2021;31(7):2869-77.
  92. Pedroso FE, Angriman F, Endo A, Dasenbrock H, Storino A, Castillo R, et al. Weight loss after bariatric surgery in obese adolescents: a systematic review and meta-analysis. Surg Obes Relat Dis. 2018;14(3):413-22.
  93. Shoar S, Mahmoudzadeh H, Naderan M, Bagheri-Hariri S, Wong C, Parizi AS, et al. Long-Term Outcome of Bariatric Surgery in Morbidly Obese Adolescents: a Systematic Review and Meta-Analysis of 950 Patients with a Minimum of 3 years Follow-Up. Obes Surg. 2017;27(12):3110-7.
  94. Ramadan M, Loureiro M, Laughlan K, Caiazzo R, Iannelli A, Brunaud L, et al. Risk of Dumping Syndrome after Sleeve Gastrectomy and Roux-en-Y Gastric Bypass: Early Results of a Multicentre Prospective Study. Gastroenterol Res Pract. 2016;2016:2570237.
  95. Ahmad A, Kornrich DB, Krasner H, Eckardt S, Ahmad Z, Braslow A, et al. Prevalence of Dumping Syndrome After Laparoscopic Sleeve Gastrectomy and Comparison with Laparoscopic Roux-en-Y Gastric Bypass. Obes Surg. 2019;29(5):1506-13.
  96. Laurenius A, Olbers T, Naslund I, Karlsson J. Dumping syndrome following gastric bypass: validation of the dumping symptom rating scale. Obes Surg. 2013;23(6):740-55.
  97. Campos GM, Mazzini GS, Altieri MS, Docimo S, Jr., DeMaria EJ, Rogers AM. ASMBS position statement on the rationale for performance of upper gastrointestinal endoscopy before and after metabolic and bariatric surgery. Surg Obes Relat Dis. 2021;17(5):837-47.
  98. Cozacov Y, Roy M, Moon S, Marin P, Lo Menzo E, Szomstein S, et al. Mid-term results of laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass in adolescent patients. Obes Surg. 2014;24(5):747-52.
  99. Nijhawan S, Martinez T, Wittgrove AC. Laparoscopic gastric bypass for the adolescent patient: long-term results. Obes Surg. 2012;22(9):1445-9.
  100. Benedix F, Krause T, Adolf D, Wolff S, Lippert H, Manger T, et al. Perioperative Course, Weight Loss and Resolution of Comorbidities After Primary Sleeve Gastrectomy for Morbid Obesity: Are There Differences Between Adolescents and Adults? Obes Surg. 2017;27(9):2388-97.