Recently, a number of novel genetic alterations have been identified that predispose individuals to pituitary adenomas. Clinically relevant pituitary adenomas are relatively common, present in 0.1% of the general population. They are mostly benign monoclonal neoplasms that arise from any of the five hormone-secreting cell types of the anterior lobe of the pituitary gland, and cause disease due to hormonal alterations and local space-occupying effects. The pathomechanism of pituitary adenomas includes alterations in cell-cycle regulation and growth factor signaling, which are mostly due to epigenetic changes; somatic and especially germline mutations occur more rarely. A significant proportion of growth hormone- and adrenocorticotrophin-secreting adenomas have activating somatic mutations in the GNAS and USP8 genes, respectively. Rarely, germline mutations predispose to pituitary tumorigenesis, often in a familial setting. Classical tumor predisposition syndromes include multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4) syndromes, Carney complex, and McCune-Albright syndrome. Pituitary tumors have also been described in association with neurofibromatosis type 1, DICER1 syndrome, and SDHx mutations. Pituitary adenomas with no other associated tumors have been described as familial isolated pituitary adenomas. Patients with AIP or GPR101 mutations often present with pituitary gigantism either in a familial or simplex setting. GNAS and GPR101 mutations that arise in early embryonic age can lead to somatic mosaicism involving the pituitary gland and resulting in growth hormone excess. Senescence has been suggested as the key mechanism protecting pituitary adenomas turning malignant in the overwhelming majority of cases. Here we briefly summarize the genetic background of pituitary adenomas, with an emphasis on the recent developments in this field. Clin Cancer Res; 22(20); 5030–42. ©2016 AACR.

See all articles in this CCR Focus section, “Endocrine Cancers: Revising Paradigms.”

The pituitary gland consists of an anterior lobe of epithelial origin and a posterior lobe of neuronal origin. The main cell types of the anterior lobe are the hormone-secreting cells [growth hormone (GH), prolactin, adrenocorticotrophin (ACTH), thyrotropin (TSH), or gonadotrophin (LH and FSH)] and the folliculostellate cells. The term “pituitary adenoma” is attributed to the usually benign tumors arising from the hormone-secreting cells of the anterior pituitary. Typically, pituitary adenomas are classified as either functioning pituitary adenomas with characteristic clinical symptoms, such as acromegaly or Cushing disease, or clinically nonfunctioning pituitary adenomas (NFPA), usually arising from cells secreting LH and FSH. These adenomas generally present as slowly growing lesions with low mitotic rate and Ki-67 labeling index (1). Symptoms are present due to hormonal disturbances, hypersecretion or lack of pituitary hormones, and compression symptoms that are secondary to local invasion and lead to hypopituitarism and visual field defects.

Data derived from autopsies and radiologic imaging studies have shown that pituitary adenomas are relatively common, estimated to be present overall in 17% of the general population (2, 3). Although most of these small lesions are incidental findings, with no obvious clinical impact (3), clinically relevant pituitary adenomas are present in 0.1% of the general population, and they represent the third most-frequent intracranial tumor type after meningiomas and gliomas (4).

Pituitary adenomas are monoclonal neoplasms in origin (5). A number of different molecular mechanisms that lead to pituitary adenomas have been identified, although in the majority of the sporadic cases, the exact molecular pathogenesis remains unknown. Factors hypothesized to contribute to pituitary neoplasia initiation and proliferation include altered growth factors and cell-cycle regulators that are the result of epigenetic changes (6), abnormal hormonal milieu, abnormal intrapituitary microenvironment (7), and inherited or somatic mutations (Fig. 1). The role of environmental factors remains questionable (8–10). In the following brief overview of the underlying pathomechanisms, we will concentrate on germline and somatic mutations that lead to pituitary adenomas.

Figure 1.

Germline, somatic, and mosaic mutations identified in familial and sporadic pituitary adenomas. Mosaic mutations denote the presence of two or more populations of cells with different genotypes in one individual who has developed from a single fertilized egg. 3P association, paraganglioma/pheochromocytome/pituitary adenoma; FIPA, familial isolated pituitary adenoma; MEN, multiple endocrine neoplasia; NF1, neurofibromatosis type 1.

Figure 1.

Germline, somatic, and mosaic mutations identified in familial and sporadic pituitary adenomas. Mosaic mutations denote the presence of two or more populations of cells with different genotypes in one individual who has developed from a single fertilized egg. 3P association, paraganglioma/pheochromocytome/pituitary adenoma; FIPA, familial isolated pituitary adenoma; MEN, multiple endocrine neoplasia; NF1, neurofibromatosis type 1.

Close modal

Familial pituitary adenomas can be divided into (i) an isolated group, in which no other organs are involved in addition to the pituitary gland, and (ii) a syndromic group, which includes multiple endocrine neoplasia type 1 (MEN1), MEN4, Carney complex, DICER1 syndrome, SDHx gene–associated syndromes, and neurofibromatosis type 1 syndrome (Fig. 1; Table 1). Familial isolated pituitary adenoma (FIPA) is the most common type followed by MEN1, which together represent 5% to 7% of patients with pituitary adenomas (11).

Table 1.

Germline and mosaic mutations predisposing to pituitary adenomas

SyndromeGenetic alteration (inheritance pattern)FunctionLocationPenetrancePrevalencecMain clinical characteristics
Carney complex PRKAR1A (AD) TSG 17q24.2 >95% overall, 80% for GH excess Unknown Skin pigmentation; myxomas; thyroid, testis and adrenal tumors, as well as somatotroph hyperplasia or adenomas 
 2p16 locus (unknown gene) — 2p16 Unknown Unknown Less severe Carney complex phenotype, mostly in sporadic cases 
 PRKACB Oncogene 1p31.1 Unknown One case described Described in one case with Carney complex (31) 
DICER1 DICER1 (AD) TSG 14q32.13 Unknown, <1% for pituitary Unknown Pituitary ACTH-secreting blastomas 
FIPA AIP (AD) TSG 11q13.2 30% 2.5% Young-onset GH adenomas and prolactinomas, rarely other pituitary adenoma type. Twenty percent of FIPA and 4%–20% of sporadic pituitary adenomas have AIP mutations 
 GPR101 (X-chromosome linked)a Oncogene Xq26.3 100% Unknown (very low, 10% of pituitary gigantism cases) Gigantism due to pituitary hyperplasia or adenoma 
McCune-Albrightb GNAS (mosaic postzygotic mutation) Oncogene 20q13.32 10–20% for pituitary 1:100,000–1:1,000,000 Polyostotic fibrous dysplasia, café-au-lait spots, and precocious puberty with GH/prolactin excess in 10%–20% 
MEN1 MEN1 (AD) TSG 11q13.1 >95% overall, 30–40% for pituitary 1:30,000 Pancreatic, pituitary (typically prolactinomas), and parathyroid gland tumors with other tumors as well 
MEN4 CDKN1B (AD) TSG 12p13.1 Unknown overall, high for pituitary Unknown (very low) MEN1 like, typically somatotroph adenomas 
Neurofibromatosis type 1 NF1 (AD) TSG 17q11.2 >95% overall, very low for pituitary 1:4,000 Café-au-lait spots, Lisch nodules, neurofibromas, optic pathway gliomas, and pheochromocytoma. Unclear whether pituitary adenomas caused by NF1 mutations 
Paraganglioma/phaeochromocytoma and pituitary adenomas—3P association SDHA (AD) TSG 5p15.33 Low penetrance Unknown Familial paraganglioma type 5 (PGL and PHEO) 
 SDHB (AD) TSG 1p36.13 ∼50% overall, very low for pituitary Unknown Familial paraganglioma type 4 (PGL with increased malignant potential) 
 SDHC (AD) TSG 1q23.3 Low penetrance overall, very low for pituitary Unknown Familial paraganglioma type 3 (head and neck PGL predominance, no pheochromocytoma, associated with GIST) 
 SDHD (AD) TSG 11q23.1 Up to 80% overall, very low for pituitary Unknown Familial paraganglioma type 1 (head and neck PGL but also pheochromocytoma) 
 SDHAF2 (AD) TSG 11q12.2 Low penetrance, very low for pituitary Unknown Familial paraganglioma type 2 (head and neck PGL) 
SyndromeGenetic alteration (inheritance pattern)FunctionLocationPenetrancePrevalencecMain clinical characteristics
Carney complex PRKAR1A (AD) TSG 17q24.2 >95% overall, 80% for GH excess Unknown Skin pigmentation; myxomas; thyroid, testis and adrenal tumors, as well as somatotroph hyperplasia or adenomas 
 2p16 locus (unknown gene) — 2p16 Unknown Unknown Less severe Carney complex phenotype, mostly in sporadic cases 
 PRKACB Oncogene 1p31.1 Unknown One case described Described in one case with Carney complex (31) 
DICER1 DICER1 (AD) TSG 14q32.13 Unknown, <1% for pituitary Unknown Pituitary ACTH-secreting blastomas 
FIPA AIP (AD) TSG 11q13.2 30% 2.5% Young-onset GH adenomas and prolactinomas, rarely other pituitary adenoma type. Twenty percent of FIPA and 4%–20% of sporadic pituitary adenomas have AIP mutations 
 GPR101 (X-chromosome linked)a Oncogene Xq26.3 100% Unknown (very low, 10% of pituitary gigantism cases) Gigantism due to pituitary hyperplasia or adenoma 
McCune-Albrightb GNAS (mosaic postzygotic mutation) Oncogene 20q13.32 10–20% for pituitary 1:100,000–1:1,000,000 Polyostotic fibrous dysplasia, café-au-lait spots, and precocious puberty with GH/prolactin excess in 10%–20% 
MEN1 MEN1 (AD) TSG 11q13.1 >95% overall, 30–40% for pituitary 1:30,000 Pancreatic, pituitary (typically prolactinomas), and parathyroid gland tumors with other tumors as well 
MEN4 CDKN1B (AD) TSG 12p13.1 Unknown overall, high for pituitary Unknown (very low) MEN1 like, typically somatotroph adenomas 
Neurofibromatosis type 1 NF1 (AD) TSG 17q11.2 >95% overall, very low for pituitary 1:4,000 Café-au-lait spots, Lisch nodules, neurofibromas, optic pathway gliomas, and pheochromocytoma. Unclear whether pituitary adenomas caused by NF1 mutations 
Paraganglioma/phaeochromocytoma and pituitary adenomas—3P association SDHA (AD) TSG 5p15.33 Low penetrance Unknown Familial paraganglioma type 5 (PGL and PHEO) 
 SDHB (AD) TSG 1p36.13 ∼50% overall, very low for pituitary Unknown Familial paraganglioma type 4 (PGL with increased malignant potential) 
 SDHC (AD) TSG 1q23.3 Low penetrance overall, very low for pituitary Unknown Familial paraganglioma type 3 (head and neck PGL predominance, no pheochromocytoma, associated with GIST) 
 SDHD (AD) TSG 11q23.1 Up to 80% overall, very low for pituitary Unknown Familial paraganglioma type 1 (head and neck PGL but also pheochromocytoma) 
 SDHAF2 (AD) TSG 11q12.2 Low penetrance, very low for pituitary Unknown Familial paraganglioma type 2 (head and neck PGL) 

Abbreviations: AD, autosomal dominant; GIST, gastrointestinal stromal tumor; PGL, paraganglioma; PHEO, pheochromocytoma; TSG, tumor suppressor gene.

aGermline or somatic mosaicism.

bSomatic mosaicism.

cIn patients with pituitary adenomas.

Isolated pituitary adenomas

FIPA is defined by the presence of pituitary adenomas in two or more family members with no other syndromic features present (12). FIPA is a heterogeneous condition that includes patients with mutations in the aryl hydrocarbon receptor–interacting protein (AIP) gene (13), patients with X-linked acrogigantism (XLAG) due to duplication of GPR101 (14), and patients with a family history of pituitary adenomas with no known genetic cause. Patients with AIP or GPR101 mutations or with AIP and GPR101-negative FIPA do not present with other types of tumors, hence the name “isolated” pituitary adenomas. Not all cases grouped under this category have a known family history, either due to low penetrance (such as in AIP mutation–positive simplex cases) or due to de novo mutations (most cases of XLAG).

AIP mutations.

The prevalence of AIP mutations in FIPA families is 17% to 20% (15, 16), whereas in sporadic cases, it ranges between 3.6% (unselected pituitary adenoma patients) and 10% to 20% (pediatric pituitary adenoma cases; refs. 17, 18). About 50% of AIP mutation–positive probands have a positive family history (16), whereas mutations in the other half are found as a germline mutation in sporadically diagnosed pituitary adenoma patients, so-called “simplex cases.” The lack of apparent family history in the latter group is due to low penetrance, as de novo mutations have only been found in two cases (19, 20). Penetrance in AIP-mutated families is incomplete (Fig. 2A): only every fifth mutation carrier manifests the disease. The age of onset is also characteristic; the disease usually manifests in the second decade of life and almost all cases are diagnosed before the age of 30 years (15, 16, 21, 22).

Figure 2.

Clinical and biological aspects of AIP mutations. A, family tree of patient (arrow) with heterozygous AIP mutation showing four other family members affected by pituitary adenomas, representing various phenotypes and incomplete penetrance (16). B, MRI of a 6-year-old female AIP mutation–positive patient (p.R304*) presenting with acute severe headache and ptosis on the right side due to pituitary apoplexy of a large pituitary adenoma (courtesy of Dr. Miles Levy, Leicester, United Kingdom). C, patients with truncating AIP mutations show significantly earlier disease onset than those with nontruncating mutations. There is a significantly higher number of pediatric cases among truncating AIP mutation–positive patients compared with nontruncating cases, suggesting a genotype–phenotype correlation (16).

Figure 2.

Clinical and biological aspects of AIP mutations. A, family tree of patient (arrow) with heterozygous AIP mutation showing four other family members affected by pituitary adenomas, representing various phenotypes and incomplete penetrance (16). B, MRI of a 6-year-old female AIP mutation–positive patient (p.R304*) presenting with acute severe headache and ptosis on the right side due to pituitary apoplexy of a large pituitary adenoma (courtesy of Dr. Miles Levy, Leicester, United Kingdom). C, patients with truncating AIP mutations show significantly earlier disease onset than those with nontruncating mutations. There is a significantly higher number of pediatric cases among truncating AIP mutation–positive patients compared with nontruncating cases, suggesting a genotype–phenotype correlation (16).

Close modal

AIP encodes a 330 amino acid protein acting as a tumor suppressor. It has a wide tissue distribution; in the normal human pituitary, it is expressed in GH cells (somatotroph cells) and prolactin-secreting cells (23). Lack of interaction with cell type–specific molecular partners might explain the specific clinical phenotype of AIP deficiency. AIP has numerous interacting partners (12). It is co-chaperone to several heat shock proteins and nuclear receptors including the aryl hydrocarbon receptor, and it interacts with phosphodiesterase 4A4/5, which regulates the cAMP pathway (24–26); however, lack of AIP leads to reduced inhibitory G protein Gαi-2 expression. The latter two aspects could be important to the tumorigenic role of AIP (27), as activation of the cAMP pathway is important in somatotroph tumorigenesis (Fig. 3): (i) activating mutations of the stimulatory G protein GNAS are present in approximately 30% of sporadic somatotroph (GH-secreting) adenomas and in McCune-Albright syndrome (28, 29), (ii) mutations in PRKAR1A and PRKACB lead to increased protein kinase A activity in Carney complex (30, 31), and (iii) the duplication of the cAMP-coupled orphan receptor GPR101 causes XLAG (14).

Figure 3.

cAMP pathway alterations in somatotroph adenomas. Several mechanisms leading to somatotroph adenomas involve the cAMP pathway. AIP, aryl hydrocarbon receptor interacting protein; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; Carney, site of mutations in Carney complex; CREB, cAMP response element; Gαs, G protein stimulatory alpha subunit; Gαi-2, G protein inhibitory alpha subunit type 2; GHRH, growth hormone–releasing hormone; GHRH-R, GHRH receptor; gsp, mutated Gαs; GTP, guanine triphosphate; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; SSTR, somatostatin receptor; ZAC1, zinc finger protein PLAGL1; PDE4A4, phosphodiesterase type 4A4; PKA, protein kinase A; R, PKA, regulatory subunit; C, PKA, catalytic subunit; XLAG, X-linked acrogigantism.

Figure 3.

cAMP pathway alterations in somatotroph adenomas. Several mechanisms leading to somatotroph adenomas involve the cAMP pathway. AIP, aryl hydrocarbon receptor interacting protein; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; Carney, site of mutations in Carney complex; CREB, cAMP response element; Gαs, G protein stimulatory alpha subunit; Gαi-2, G protein inhibitory alpha subunit type 2; GHRH, growth hormone–releasing hormone; GHRH-R, GHRH receptor; gsp, mutated Gαs; GTP, guanine triphosphate; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; SSTR, somatostatin receptor; ZAC1, zinc finger protein PLAGL1; PDE4A4, phosphodiesterase type 4A4; PKA, protein kinase A; R, PKA, regulatory subunit; C, PKA, catalytic subunit; XLAG, X-linked acrogigantism.

Close modal

AIP mutations predispose individuals to childhood or young adult-onset disease with large, aggressive, poorly responsive, mostly GH or prolactin-secreting tumors, leading to gigantism in 40% of the cases (Fig. 2B and C; refs. 15, 16, 23). There is a phenotype–genotype correlation, as patients with truncating mutations are significantly younger at disease onset in comparison with patients with nontruncating AIP mutations (Fig. 2C; ref. 16). Patients with AIP mutations have an increased risk for pituitary apoplexy (Fig. 2B) compared with AIP mutation–negative young GH excess patients (16, 23, 32). Classical pituitary apoplexy refers to a clinical syndrome that is characterized by sudden onset of headache, vomiting, visual impairment, and decreased consciousness that evolves over hours or days and is caused by hemorrhage and/or infarction of a pituitary adenoma. Patients with AIP mutations often need repeated surgery and external pituitary irradiation, and they are relatively resistant to treatment with somatostatin analogues [(SSA) i.e., poor reduction in hormone levels and tumor size (15)]. The mechanism of somatostatin resistance could involve the somatostatin receptor type 2 (SSTR2)–ZAC1 pathway (33). AIP is upregulated by SSA (34, 35) and, in turn, AIP can upregulate ZAC1 mRNA expression (35, 36). Another possible mechanism explaining the somatostatin resistance of these patients is the reduced expression of the inhibitory G protein subtype Gαi-2. SSTR2 is known to regulate cAMP levels via inhibitory G proteins (37), and deficiency of Gαi-2 could play a role in the SSA resistance of AIP mutation–positive samples (ref. 35; Fig. 3).

In sporadic adenomas that do not harbor AIP mutations, low AIP protein expression is associated with reduced SSTR2 expression, which predicts a reduced responsiveness to SSA therapy (38). Tumors pretreated with SSA before surgery show an increase in AIP expression (34, 35). It has been suggested that AIP might be a better marker of invasiveness in somatotrophinomas (GH-secreting adenomas) than Ki-67 and p53, even in AIP mutation–negative adenomas (39).

Genetic screening and clinical follow-up of family members of AIP mutation–positive probands revealed numerous cases of prospectively identified patients, where the early diagnosis led to earlier treatment and potential avoidance of significant complications (16). This suggests that patients with gigantism or young-onset acromegaly and their families could benefit from genetic screening and clinical follow-up.

GPR101–X-linked acrogigantism.

XLAG is a novel genetic cause of GH excess. It usually presents at a very early age as a sporadic disease due to a de novo microduplication on the X chromosome involving the GPR101 gene in patients with gigantism (40–42). The majority of the cases are females with germline microduplication (14, 40, 42). Two familial cases have been described with transmission from affected mother to an affected son and show full penetrance (14). Somatic mosaic mutation cases have also been described in males where the mutation was identified in the pituitary tissue and/or at low level in germline (18, 41, 42). Although the originally identified Xq26.3 duplicated area involves four genes (14), only one of these, the GPR101 gene, has been found upregulated at the mRNA level in pituitary tissue. We have recently identified a patient with XLAG whose duplicated area includes only the GPR101 gene, but not the other three genes, indicating the pathogenic role of GPR101 (14, 42). Activation of GPR101, an orphan Gs protein–coupled receptor, leads to an increase in cAMP levels (refs. 43, 44; Fig. 3). GPR101 is expressed in the caudate putamen, nucleus accumbens, and hypothalamus (45). The endogenous ligand and the exact function of GPR101 remain unknown. It has been suggested, in a uterine cancer cell line, that a fragment of the gonadotrophin-releasing hormone [GnRH-(1–5)] indirectly stimulated GPR101 to induce the release of EGF and subsequent phosphorylation of the EGFR (46), leading to enhanced cellular migration. However, (i) GnRH-(1–5) does not change cAMP levels, which are important for somatotroph tumorigenesis, and (ii) an upregulated EGFR pathway seems to lead to corticotroph (i.e., ACTH-secreting) adenomas (see section on USP8), but not somatotroph adenomas. Therefore, the mechanism by which mutant GPR101 contributes to increased GH secretion is still unclear.

Patients with XLAG have significant GH excess leading invariably to gigantism due to their particularly early age of onset, even in comparison with other pituitary gigantism cases, occurring before the age of 5 years, with growth curves significantly exceeding the 97th percentile (14, 40–42, 47). Most of these patients secrete increased amounts of both GH and prolactin due to a somatomammotroph adenoma and mixed cell hyperplasia. In the adenomas, the Ki-67 index is low or moderate (except one case; ref. 48), SSTR expression is preserved, and AIP expression is moderate or high; GHRH is not expressed, whereas there is increased expression of GPR101 and the GHRH receptor (40, 42). Patients with XLAG provide a particular challenge in management due to the very young age of onset, the fact that some have hyperplasia rather than an adenoma, and due to partial or complete resistance to SSA and dopamine agonist therapy. Radical pituitary surgery or radiotherapy could be effective but often leads to hypopituitarism (40). The GH receptor antagonist pegvisomant in combination with somatostatin and dopamine agonists has been shown to control IGF-1 and excessive growth (40, 41).

Familial isolated pituitary adenoma with no known gene mutation.

Approximately 80% of FIPA cases do not have an identified gene mutation. While AIP- and GPR101-related disease starts in young or very young patients, in this group, the age of diagnosis is often (>60%) after the third decade of life and only 2% of the cases have gigantism (16). In our cohort, the most frequent diagnosis was acromegaly followed by prolactinoma, while in another large cohort, macroprolactinomas were the most common (49). Fifty-eight percent of our families presented with the same pituitary adenoma type (homogeneous FIPA families), whereas different adenoma types were present in the same family in 42% of the cases. The penetrance seems to be lower than in AIP mutation–positive families, as approximately 80% of the families have only two affected family members. We cannot exclude the possibility that, given the prevalence of pituitary adenomas in the general population (1:1,000), some of these adenomas coexist in a family as a coincidence rather than due to an existing underlying germline mutation.

Syndromic pituitary adenomas

MEN1 syndrome.

MEN1 is characterized by the presence of the classical triad of hyperparathyroidism (in almost all patients by the age of 50 years), pituitary adenomas (in about 30%–40% of cases), and neuroendocrine tumors [in about 60% of cases; further data on neuroendocrine tumors can be found in the CCR Focus article by Maxwell and colleagues (50)]. In addition, other tumor types have also been associated with this syndrome, such as facial angiofibromas (85%), collagenomas (72%), adrenal cortical adenomas (40%), lipomata (30%), meningiomas (8%), pheochromocytomas (rarely). Breast cancer (6%) has also been suggested more recently.

The MEN1 gene encodes a ubiquitously expressed transcriptional cofactor that regulates cell-cycle proteins such as p27 (51) and cyclin-dependent kinase subunit 4 (CDK4; ref. 52), factors also found to be altered in sporadic pituitary adenomas (see the following section). The interaction with p27 is especially interesting, as mutations in the gene encoding p27 (CDKN1B) are also associated with a MEN1 syndrome–like phenotype (see the following section). MENIN also has a role in G1–S checkpoint regulation, response to DNA damage and apoptosis, regulation of histone deacetylation and methyl transferase complexes, interaction with transcription factors and nuclear receptors, as well as transport of β-catenin (53).

MEN1-related pituitary disease is dominated by prolactinoma cases; however, systematic screening reveals similar numbers of small nonfunctioning pituitary adenomas (42%), followed by somatotrophinomas (7%) and a smaller percentage of the other pituitary adenoma types (54). Nonfunctioning adenomas were more often microadenomas detected during screening. Pituitary adenomas could be the first manifestation of MEN1 syndrome in 15% to 20% of the cases, and the current guidelines suggest that pituitary screening should start at the age of 5 years in mutation carriers (55).

MEN4 syndrome.

Not all patients with MEN1 syndrome harbor a mutation in the MEN1 gene. Loss-of-function mutations have been identified in CDKN1B, coding for p27, in a subset of these patients, and the phenotype was named MEN4 syndrome (56). The most common pituitary tumor type is somatotrophinoma, and a childhood-onset case has also been described (57). Other tumors include parathyroid adenomas, adrenal tumors, renal angiomyolipomas, uterine fibroids, gastrinomas, neuroendocrine cervical carcinomas, bronchial carcinoids, papillary thyroid carcinomas, and gastric carcinomas. Interestingly, in addition to the usual mutation types (nonsense, frameshift, and missense), alterations in the 5′ open reading frame were also shown to disrupt the function of the p27 protein (57, 58). In a few MEN1-like syndrome cases, mutations in other cell-cycle inhibitors, such as p15 (CDKN2B), p18 (CDKN2C), and p21 (CDKN1A), have also been described (59). These cases, together with MEN1-related pituitary adenomas and with the gene expression data of sporadic pituitary adenomas (see below), support the hypothesis that cell-cycle dysregulation is an important factor in pituitary adenoma development.

Carney complex.

Carney complex is characterized by the presence of endocrine and nonendocrine tumors with spotty skin pigmentation, as well as cardiac and cutaneous myxomas (60). More than two-thirds of patients present asymptomatic elevation of IGF-1, GH, and prolactin due to pituitary hyperplasia, and 10% of the patients present with adenomas and symptomatic acromegaly (61). The age of onset is usually after the third decade; however, a few cases of gigantism have also been described. Carney complex is caused in 70% of the cases by inactivating mutations in the regulatory subunit of protein kinase A (PRKAR1A), which leads to excessive cAMP signaling (ref. 62; Fig. 3). Large deletions within the gene lead to a more severe phenotype. Another genetic locus at 2p16 has also been shown to be associated with Carney complex (63). More recently, a single case was described of an activating mutation (gene duplication) of the catalytic subunit of protein kinase A (PRKACB; ref. 31). It has been shown that PRKAR1A haploinsufficiency leads to a dysregulated WNT signaling pathway, in addition to cell-cycle abnormalities (64). Interestingly, embryonic mutations in β-catenin (CTNNB1), a crucial element of the classical WNT pathway, leads to craniopharyngiomas, a nonhormone–secreting pituitary tumor (65).

DICER1 syndrome.

Pituitary blastoma is a novel aspect of the DICER1 syndrome. Germline mutations in DICER1 lead to infant pleuropulmonary blastoma, differentiated thyroid carcinoma, multinodular goiter, nasal chondromesenchymal hamartoma, ovarian sex cord stromal tumor, Sertoli-Leydig cell tumor, pineoblastoma, and pituitary blastoma. Pituitary blastomas develop before the age of 2 years and secrete ACTH, causing severe Cushing disease (66–68). The penetrance of pituitary blastoma is low (∼1%; ref. 66). Approximately half of the children with confirmed pituitary blastoma die of the disease within months of diagnosis (66–68).

DICER1 is a cytoplasmic endoribonuclease that processes hairpin precursor miRNAs into short, functional miRNAs that downregulate targeted mRNAs, thereby modulating cellular protein production (69). In the more than 50 reported DICER1 mutation kindreds, germline mutations usually led to truncated proteins (67), whereas the “second hit” somatic mutations were typically in the metal-binding sites of the catalytic RNase IIIa and b domains. The exact mechanism explaining this special scenario or why DICER1 mutations in general lead to tumorigenesis remains unclear.

Succinate dehydrogenase mutations (SDHx): paraganglioma, pheochromocytoma, and pituitary adenoma association.

Pheochromocytomas and/or paragangliomas (PGL) have rarely been associated with pituitary adenomas (70–74). Patient with these tumors harbor mutations in the SDHA-D or SDHA2F genes. Loss of heterozygosity at the SDH locus in the pituitary adenomas, as well as data from animal studies, supports the causality between these genes and pituitary adenoma tumorigenesis (71, 73, 75).

SDH, a multimeric enzyme bound to the inner membrane of mitochondria, has two essential roles: (i) it is an important member of the Krebs cycle, and (ii) it plays a role in oxidative phosphorylation and controls activation of hypoxia-inducible factor 1a (HIF1α), leading to VEGF upregulation (75). SDH protein complex is formed by the subunits A, B, C, and D with its associated assembly factor (SDHAF2). Several mechanisms have been described to explain the tumorigenic effect of SDH mutations: (i) succinate accumulation that inhibits HIFα-prolyl hydroxylases, leading to activation of HIF1α and resulting in a state of tissue pseudohypoxia (76) and reactive oxygen species accumulation, and (ii) inhibition of histone demethylases that leads to epigenetic changes [ref. 77; see further details in the article by Jochmanova and Pacak in this CCR Focus (78)]. Interestingly, the HIF1α–VEGF pathway is upregulated by RSUME, a gene with increased expression in sporadic pituitary adenomas (79). The upregulated VEGF pathway in SDH-related pituitary adenomas may play a role in their invasive and aggressive behavior (80).

The majority of the described SDH-related pituitary adenoma cases are prolactinomas, followed by NFPA- and GH-secreting adenomas. The age of onset of pituitary adenomas is comparable with that of sporadic tumors without germline mutations (∼40–50 years), and the vast majority of them present as invasive macroadenomas with a unique vacuolated histologic phenotype (71). The penetrance of pituitary adenomas in patients with SDH mutations is low; whether imaging of the pituitary fossa should be added to at least the first MRI screening of the neck and skull base area, especially in SDHB mutation–positive cases, remains to be determined.

Sporadic pituitary adenomas harbor a lower somatic mutation rate in comparison with malignant tumors (81, 82), which is consistent with their typically low proliferation rate and benign phenotype.

GNAS

The most frequently observed (up to 40%) genetic change in somatotroph adenomas is the somatic heterozygous gain-of-function mutation of the GNAS gene coding for the Gsα subunit (11, 83, 84). The mutations, known as gsp mutations, affecting codon 201 or 227 destroy the GTPase activity of the protein (Fig. 3). Prolonged adenylyl cyclase stimulation and increased cAMP synthesis result in increased cell proliferation and GH secretion. GNAS is an imprinted gene in the pituitary; mutations are always located on the maternal allele (85). Some, but not all, studies found gsp-positive somatothropinomas to be more responsive to SSA treatment and mostly to be densely granulated somatotroph adenomas (6, 36). No other specific recurrent somatic mutations have been identified in somatotroph adenomas (82).

USP8

In ACTH-secreting adenomas, novel somatic gain-of-function mutations have recently been identified in the USP8 gene (86, 87). Although USP8 is expressed in all anterior pituitary cell types (88), USP8 mutations have only been identified in corticotroph adenomas (87). The pathomechanism leading to corticotroph adenomas has been linked to EGFR. USP8 encodes a deubiquitinase enzyme that can protect EGFR from degradation by removing lysosome targeting ubiquitin tags, allowing recycling of the receptor to the cell surface (ref. 89; Fig. 4). EGFR is known to be expressed in corticotrophinomas, where it leads to increased POMC levels, ACTH synthesis, and corticotroph cell proliferation, and its presence correlates with invasiveness (86, 87, 90–92). All the identified USP8 mutations are located in the 14-3-3 protein-binding motif of the protein, a domain highly conserved across species, suggesting that lack of 14-3-3 binding leads to increased cleavage and, therefore, increased catalytic activity of the shortened USP8 protein. Gain-of-function mutations in USP8 increase deubiquitination of EGFR, which inhibits its degradations and leads to activation of EGFR signaling. Corticotroph adenomas with mutated USP8 are more frequently found in females (67% vs. 38%; ref. 87), are smaller tumors, have higher ACTH production, and show better prognosis (87, 90). Not all studies found a correlation between EGFR expression and USP8 mutation status (93). USP8-mutated tumors express significantly higher levels of proopiomelanocortin (the gene encoding ACTH) and SSTR5. An important clinical question is whether these adenomas, treated preoperatively or after unsuccessful surgery, respond better to multiligand somatostatin analogue pasireotide than USP8 mutation–negative adenomas. The USP8-related tumorigenesis also opens up the possibility of EGFR-directed therapy for corticotrophinomas; indeed, this concept has already been tested in various animal models using EGFR inhibitor gefitinib (94).

Figure 4.

Mechanism of USP8 induced tumorigenesis. In the normal pituitary, there is a balance between EGFR ubiquitination and degradation and deubiquitination by USP8 leading to EGFR recycling. Mutation in the 14-3-3 protein–binding site of USP8 (yellow star) results in excessive cleavage and higher deubiquitination activity, leading to increased recycling of EGFR.

Figure 4.

Mechanism of USP8 induced tumorigenesis. In the normal pituitary, there is a balance between EGFR ubiquitination and degradation and deubiquitination by USP8 leading to EGFR recycling. Mutation in the 14-3-3 protein–binding site of USP8 (yellow star) results in excessive cleavage and higher deubiquitination activity, leading to increased recycling of EGFR.

Close modal

Prolactinomas and NFPAs

Some prolactinomas show loss of chromosome 11 (95) and trisomy of chromosomes 5, 8, and 12, whereas no specific recurrent single-nucleotide mutations have been identified to date.

No recurrent specific somatic mutations have been identified in NFPAs using exome sequencing (81); however, intronic mutations, copy-number variations, or other genetic mechanism, not detected with this method, could play a role.

All pituitary adenoma types

Point mutations and increased copy number of the gene coding for the catalytic subunit of phosphoinositide 3-kinase catalytic subunit PI3K (phosphoinositide 3-kinase catalytic subunit PIK3CA) have been identified in 20% to 40% of various types of pituitary adenomas (96–98), but patients with Cowden syndrome who harbor germline mutations in the PI3K–PTEN–AKT pathway genes do not present with pituitary adenomas (99), suggesting that PIK3CA amplification could be a permissive phenomenon.

Sixty percent of oncocytic pituitary adenomas show mutations in mitochondrial DNA genes coding for various respiratory complex I components (MTND1,2,4,5, MTTL2, MTTM, MTCYB, and MTRNR2) leading to the disruption of respiratory complex I (100). It has been suggested that this leads to lack of HIF1α stabilization and, therefore, the relatively benign nature of these adenomas (101).

GNAS

Patients with somatic mosaicism with GNAS mutations have McCune-Albright syndrome (Fig. 3). One of the characteristic manifestations of this disease is pituitary hyperplasia or tumor resulting in increased GH and prolactin levels in addition to the classical triad of polyostotic fibrous dysplasia, café-au-lait spots, and precocious puberty (102). Other endocrine dysfunctions include testicular lesions, hyperthyroidism, phosphate wasting, and hypercortisolism. GH-secreting tumors (20%–30% of patients) usually present before 20 years of age, although the diagnosis can be delayed due to craniofacial fibrous dysplasia resembling features of acromegaly (103–106). This is one of very few diseases that can result in both abnormally short stature, due to precocious puberty, or gigantism, due to young-onset GH excess. Patients with GH excess and craniofacial fibrous dysplasia present a surgical challenge due to skull-base thickening and obliteration of the sphenoid sinus, and treatment often requires total hypophysectomy due to diffuse involvement of the pituitary (104). Somatostatin analogues, especially in combination with GH receptor antagonist treatment, can effectively reduce IGF-1 levels (103, 105).

GPR101

In addition to germline mutations, GPR101 duplication can occur as somatic mosaicism (41, 107). The clinical phenotype of patients with mosaic GPR101 duplication, only described in male patients until now, does not differ from the germline cases. GPR101 sequence variants, apart from gene duplication, do not play a role in pituitary tumorigenesis (42, 108).

The pathogenesis of sporadic pituitary adenomas is influenced by multiple factors (Fig. 5), and we refer to other reviews for detailed discussion of these elements (84, 109–111). The number of somatic mutations identified are small compared with other neoplastic conditions, and no data are available for simultaneous multiple mutated gene sets that are so characteristic of many other tumor types (112, 113). Abnormalities in cell-cycle regulation are considered a crucial event in the formation of pituitary adenomas. Loss of CDK inhibitors, such as p16 or p27 (114, 115), or overexpression of CDKs, such as cyclin D (116), are involved in tumor development (109). It has been estimated that 80% of human pituitary adenomas display alterations in at least one cell-cycle regulator (110). A systematic review of epigenetic regulation of pituitary adenomas identified altered expression of tumor suppressor genes [including genes coding for p16, p21, p27, p14, death associated protein kinase (DAPK), growth arrest, and DNA damage-inducible protein (GADD45γ), p73, retinoblastoma protein, BMP-4], oncogenes (PTTG and MAGEA3), imprinted genes (GNAS, NNAT, MEG3), epigenome modifiers (DNMT3b), and transcription regulators (HMGA2). More recent examples of proteins involved in pituitary tumorigenesis are the mammalian sterile-20-like kinase (MST4) and CABLES1. MST4, which was found to be upregulated in NFPAs, stimulates p38, AKT, and HIF1—known factors in pituitary tumorigenesis—whereas an MST4 inhibitor showed promising results in in vitro studies (117, 118). Another recently identified protein is CABLES1, which was lost in 55% of human corticotroph adenomas, and its levels correlated with loss of p27. The feedback effect of glucocorticoids following upregulation of CABLES1 might link the tumorigenic process with glucocorticoid-regulated cell-cycle progression (119). Hormonal factors could also have a role as prolactinomas with lower expression of estrogen receptor α have a higher tumor grade, greater resistance to treatment, and worse prognosis (120).

Figure 5.

Multiple pathways to pituitary tumorigenesis. Pathways, with representative examples, suggested to be involved in pituitary adenoma tumorigenesis either due to mutations (blue boxes; bold gene names represent tumor suppressor genes, gene names that are not bold represent oncogenes) or altered gene expression (orange boxes). Pituitary adenomas have significantly lower somatic mutation burden compared with other neoplasms, and simultaneous mutational events with representative mutational gene sets have not been described. Altered gene expression, however, has been identified in various pathways—the most important being the regulation of the cell cycle [80% of pituitary adenomas have altered gene expression resulting in a dysregulated cell cycle (110)].

Figure 5.

Multiple pathways to pituitary tumorigenesis. Pathways, with representative examples, suggested to be involved in pituitary adenoma tumorigenesis either due to mutations (blue boxes; bold gene names represent tumor suppressor genes, gene names that are not bold represent oncogenes) or altered gene expression (orange boxes). Pituitary adenomas have significantly lower somatic mutation burden compared with other neoplasms, and simultaneous mutational events with representative mutational gene sets have not been described. Altered gene expression, however, has been identified in various pathways—the most important being the regulation of the cell cycle [80% of pituitary adenomas have altered gene expression resulting in a dysregulated cell cycle (110)].

Close modal

Pituitary carcinomas are extremely rare. They are diagnosed when distant metastases are detected, and survival is usually less than 2 years after diagnosis, which is similar to some other malignancies such as adrenal carcinoma [see article by Payabyab and colleagues in this CCR Focus (121)]. The majority of the cases arise from prolactin- and ACTH-secreting cells, and more rarely GH- or LH/FSH-secreting cells. It is currently unclear whether they develop as de novo carcinomas or pituitary adenomas that gradually gain malignant features. Somatic changes in tumor suppressor genes (e.g., TP53 and RB1) and oncogenes (e.g., HRAS and MYC) commonly present in other neoplasia, have been identified in only a few cases of pituitary carcinoma (122). A few MEN1-related pituitary carcinomas (123, 124) and one SDH-related (80) pituitary carcinoma have been described in patients with pituitary tumor–predisposing syndromes.

The precise mechanism of why pituitary adenomas do not turn cancerous, even after many years of disease, remains unknown; however, it has been suggested that oncogene-induced senescence plays an important role (125, 126). Senescence is characterized by a signal transduction program leading to irreversible cell-cycle arrest and represents an important protective mechanism against malignancy. Senescence restrains proliferation, but allows the cell to remain viable and perform its physiologic function. This process can be activated by DNA damage, telomere shortening, lysosomal or oxidative stress, chromosomal instability, aneuploidy, loss of tumor-suppressive signaling, or oncogenic activity (110, 127). These events trigger the activation of different cell-cycle regulators, such as p53 and pRb, to upregulate the senescence pathways. PTTG, an oncogene often overexpressed in pituitary adenomas, promotes chromosomal instability and aneuploidy, and lack of PTTG results in pituitary-specific senescent features (127). IL6 is a cytokine involved in pituitary tumor progression, but it is also required for induction and maintenance of oncogene-induced senescence via an autocrine mechanism (128). Senescence markers have been shown to be present in human adenomas where subtype-specific senescence induction pathways could play an important role (125, 129).

In summary, several new pituitary adenoma–predisposing genes have been identified in the last few years: germline mutations, causing isolated pituitary adenomas or syndromic disease, as well as somatic or mosaic mutations. The clinical characterization of patients with sporadic disease or genetic predisposition for pituitary adenomas, and the elucidation of the exact molecular mechanisms will contribute to the identification of novel physiologic pathways, leading to better understanding of tumorigenesis as well as novel therapies.

M. Korbonits reports receiving commercial research grants from Novartis and Pfizer, and is a consultant/advisory board member for Pfizer. No potential conflicts of interest were disclosed by the other author.

Conception and design: F. Caimari, M. Korbonits

Writing, review, and/or revision of the manuscript: F. Caimari, M. Korbonits

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): F. Caimari, M. Korbonits

Study supervision: M. Korbonits

The authors are grateful to Professor Ashley Grossman (Oxford) and Julia Heckenast (Bristol) for their review of the article.

F. Caimari was supported by the Fundación Alfonso Martin Escudero fellowship.

1.
Levy
A
,
Lightman
S
. 
Molecular defects in the pathogenesis of pituitary tumours
.
Front Neuroendocrinol
2003
;
24
:
94
127
.
2.
Daly
AF
,
Tichomirowa
MA
,
Beckers
A
. 
The epidemiology and genetics of pituitary adenomas
.
Best Pract Res Clin Endocrinol Metab
2009
;
23
:
543
54
.
3.
Ezzat
S
,
Asa
SL
,
Couldwell
WT
,
Barr
CE
,
Dodge
WE
,
Vance
ML
, et al
The prevalence of pituitary adenomas: a systematic review
.
Cancer
2004
;
101
:
613
9
.
4.
Fernandez
A
,
Karavitaki
N
,
Wass
JA
. 
Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK)
.
Clin Endocrinol
2010
;
72
:
377
82
.
5.
Herman
V
,
Fagin
J
,
Gonsky
R
,
Kovacs
K
,
Melmed
S
. 
Clonal origin of pituitary adenomas
.
J Clin Endocrinol Metab
1990
;
71
:
1427
33
.
6.
Zhou
Y
,
Zhang
X
,
Klibanski
A
. 
Genetic and epigenetic mutations of tumor suppressive genes in sporadic pituitary adenoma
.
Mol Cell Endocrinol
2014
;
386
:
16
33
.
7.
Yoshida
S
,
Kato
T
,
Kato
Y
. 
EMT involved in migration of stem/progenitor cells for pituitary development and regeneration
.
J Clin Med
2016
;
5
:
pii
:
E43
.
8.
Cannavò
S
,
Ferraù
F
,
Ragonese
M
,
Curtò
L
,
Torre
ML
,
Magistri
M
, et al
Increased prevalence of acromegaly in a highly polluted area
.
Eur J Endocrinol
2010
;
163
:
509
13
.
9.
Cannavo
S
,
Ragonese
M
,
Puglisi
S
,
Romeo
PD
,
Torre
ML
,
Alibrandi
A
, et al
Acromegaly is more severe in patients with AHR or AIP gene variants living in highly polluted areas
.
J Clin Endocrinol Metab
2016
;
101
:
1872
9
.
10.
Pesatori
AC
,
Baccarelli
A
,
Consonni
D
,
Lania
A
,
Beck-Peccoz
P
,
Bertazzi
PA
, et al
Aryl hydrocarbon receptor-interacting protein and pituitary adenomas: a population-based study on subjects exposed to dioxin after the Seveso, Italy, accident
.
Eur J Endocrinol
2008
;
159
:
699
703
.
11.
Aflorei
ED
,
Korbonits
M
. 
Epidemiology and etiopathogenesis of pituitary adenomas
.
J Neurooncol
2014
;
117
:
379
94
.
12.
Beckers
A
,
Aaltonen
LA
,
Daly
AF
,
Karhu
A
. 
Familial isolated pituitary adenomas (FIPA) and the pituitary adenoma predisposition due to mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene
.
Endocr Rev
2013
;
34
:
239
77
.
13.
Vierimaa
O
,
Georgitsi
M
,
Lehtonen
R
,
Vahteristo
P
,
Kokko
A
,
Raitila
A
, et al
Pituitary adenoma predisposition caused by germline mutations in the AIP gene
.
Science
2006
;
312
:
1228
30
.
14.
Trivellin
G
,
Daly
AF
,
Faucz
FR
,
Yuan
B
,
Rostomyan
L
,
Larco
DO
, et al
Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation
.
N Engl J Med
2014
;
371
:
2363
74
.
15.
Daly
AF
,
Tichomirowa
MA
,
Petrossians
P
,
Heliövaara
E
,
Jaffrain-Rea
M-L
,
Barlier
A
, et al
Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study
.
J Clin Endocrinol Metab
2010
;
95
:
E373
83
.
16.
Hernández-Ramírez
LC
,
Gabrovska
P
,
Dénes
J
,
Stals
K
,
Trivellin
G
,
Tilley
D
, et al
Landscape of familial isolated and young-onset pituitary adenomas: prospective diagnosis in AIP mutation carriers
.
J Clin Endocrinol Metab
2015
;
100
:
E1242
54
.
17.
Cazabat
L
,
Bouligand
J
,
Salenave
S
,
Bernier
M
,
Gaillard
S
,
Parker
F
, et al
Germline AIP mutations in apparently sporadic pituitary adenomas: prevalence in a prospective single-center cohort of 443 patients
.
J Clin Endocrinol Metab
2012
;
97
:
663
70
.
18.
Cuny
T
,
Pertuit
M
,
Sahnoun-Fathallah
M
,
Daly
A
,
Occhi
G
,
Odou
MF
, et al
Genetic analysis in young patients with sporadic pituitary macroadenomas: besides AIP don't forget MEN1 genetic analysis
.
Eur J Endocrinol
2013
;
168
:
533
41
.
19.
Stratakis
CA
,
Tichomirowa
MA
,
Boikos
S
,
Azevedo
MF
,
Lodish
M
,
Martari
M
, et al
The role of germline AIP, MEN1, PRKAR1A, CDKN1B and CDKN2C mutations in causing pituitary adenomas in a large cohort of children, adolescents, and patients with genetic syndromes
.
Clin Genet
2010
;
78
:
457
63
.
20.
Ramírez-Rentería
C
,
Hernández-Ramírez
LC
,
Portocarrero-Ortiz
L
,
Vargas
G
,
Melgar
V
,
Espinosa
E
, et al
AIP mutations in young patients with acromegaly and the Tampico Giant: the Mexican experience
.
Endocrine
2016
;
53
:
402
11
.
21.
Williams
F
,
Hunter
S
,
Bradley
L
,
Chahal
HS
,
Storr
HL
,
Akker
SA
, et al
Clinical experience in the screening and management of a large kindred with familial isolated pituitary adenoma due to an aryl hydrocarbon receptor interacting protein (AIP) mutation
.
J Clin Endocrinol Metab
2014
;
99
:
1122
31
.
22.
Korbonits
M
,
Storr
H
,
Kumar
AV
. 
Familial pituitary adenomas - Who should be tested for AIP mutations?
Clin Endocrinol
2012
;
77
:
351
6
.
23.
Leontiou
CA
,
Gueorguiev
M
,
van der Spuy
J
,
Quinton
R
,
Lolli
F
,
Hassan
S
, et al
The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas
.
J Clin Endocrinol Metab
2008
;
93
:
2390
401
.
24.
Trivellin
G
,
Korbonits
M
. 
AIP and its interacting partners
.
J Endocrinol
2011
;
210
:
137
55
.
25.
Igreja
S
,
Chahal
HS
,
King
P
,
Bolger
GB
,
Srirangalingam
U
,
Guasti
L
, et al
Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families
.
Hum Mutat
2010
;
31
:
950
60
.
26.
Bolger
GB
,
Bizzi
MF
,
Pinheiro
SV
,
Trivellin
G
,
Smoot
L
,
Accavitti
M-A
, et al
cAMP-specific PDE4 phosphodiesterases and AIP in the pathogenesis of pituitary tumors
.
Endocr Relat Cancer
2016
;
23
:
419
31
.
27.
Formosa
R
,
Xuereb-Anastasi
A
,
Vassallo
J
. 
Aip regulates cAMP signalling and GH secretion in GH3 cells
.
Endocr Relat Cancer
2013
;
20
:
495
505
.
28.
Landis
CA
,
Masters
SB
,
Spada
A
,
Pace
AM
,
Bourne
HR
,
Vallar
L
. 
GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours
.
Nature
1989
;
340
:
692
6
.
29.
Weinstein
LS
,
Shenker
A
,
Gejman
PV
,
Merino
MJ
,
Friedman
E
,
Spiegel
AM
. 
Activating mutations of the stimulatory G protein in the McCune-Albright syndrome
.
N Engl J Med
1991
;
325
:
1688
95
.
30.
Kirschner
LS
,
Carney
JA
,
Pack
SD
,
Taymans
SE
,
Giatzakis
C
,
Cho
YS
, et al
Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex
.
Nat Genet
2000
;
26
:
89
92
.
31.
Forlino
A
,
Vetro
A
,
Garavelli
L
,
Ciccone
R
,
London
E
,
Stratakis
CA
. 
PRKACB and Carney Complex
.
N Engl J Med
2014
;
370
:
1065
7
.
32.
Xekouki
P
,
Mastroyiannis
SA
,
Avgeropoulos
D
,
de la Luz Sierra
M
,
Trivellin
G
,
Gourgari
EA
, et al
Familial pituitary apoplexy as the only presentation of a novel AIP mutation
.
Endocr Relat Cancer
2013
;
20
:
L11
4
.
33.
Theodoropoulou
M
,
Zhang
J
,
Laupheimer
S
,
Paez-Pereda
M
,
Erneux
C
,
Florio
T
, et al
Octreotide, a somatostatin analogue, mediates its antiproliferative action in pituitary tumor cells by altering phosphatidylinositol 3-kinase signaling and inducing Zac1 expression
.
Cancer Res
2006
;
66
:
1576
82
.
34.
Jaffrain-Rea
M-L
,
Rotondi
S
,
Turchi
A
,
Occhi
G
,
Barlier
A
,
Peverelli
E
, et al
Somatostatin analogues increase AIP expression in somatotropinomas, irrespective of Gsp mutations
.
Endocr Relat Cancer
2013
;
20
:
753
66
.
35.
Chahal
HS
,
Trivellin
G
,
Leontiou
CA
,
Alband
N
,
Fowkes
RC
,
Tahir
A
, et al
Somatostatin analogs modulate AIP in somatotroph adenomas: the role of the ZAC1 pathway
.
J Clin Endocrinol Metab
2012
;
97
:
1411
20
.
36.
Gadelha
MR
,
Kasuki
L
,
Korbonits
M
. 
Novel pathway for somatostatin analogs in patients with acromegaly
.
Trends Endocrinol Metab
2013
;
24
:
238
46
.
37.
Tuominen
I
,
Heliövaara
E
,
Raitila
A
,
Rautiainen
M-R
,
Mehine
M
,
Katainen
R
, et al
AIP inactivation leads to pituitary tumorigenesis through defective Gαi-cAMP signaling
.
Oncogene
2015
;
34
:
1174
84
.
38.
Kasuki
L
,
Vieira Neto
L
,
Wildemberg
LEA
,
Colli
LM
,
de Castro
M
,
Takiya
CM
, et al
AIP expression in sporadic somatotropinomas is a predictor of the response to octreotide LAR therapy independent of SSTR2 expression
.
Endocr Relat Cancer
2012
;
19
:
L25
9
.
39.
Kasuki
L
,
Vieira Neto
L
,
Wildemberg
LEA
,
Gasparetto
EL
,
Marcondes
J
,
de Almeida Nunes
B
, et al
Low aryl hydrocarbon receptor-interacting protein expression is a better marker of invasiveness in somatotropinomas than Ki-67 and p53
.
Neuroendocrinology
2011
;
94
:
39
48
.
40.
Beckers
A
,
Lodish
MB
,
Trivellin
G
,
Rostomyan
L
,
Lee
M
,
Faucz
FR
, et al
X-linked acrogigantism syndrome: clinical profile and therapeutic responses
.
Endocr Relat Cancer
2015
;
22
:
353
67
.
41.
Rodd
C
,
Millette
M
,
Iacovazzo
D
,
Stiles
C
,
Barry
S
,
Evanson
J
, et al
Somatic GPR101 duplication causing X-linked acrogigantism (X-LAG) - diagnosis and management
.
J Clin Endocrinol Metab
2016
;
101
:
1927
30
.
42.
Iacovazzo
D
,
Caswell
R
,
Bunce
B
,
Jose
S
,
Yuan
B
,
Hernández-Ramírez
LC
, et al
Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study
.
Acta Neuropathol Commun
2016
;
4
:
56
.
43.
Bates
B
,
Zhang
L
,
Nawoschik
S
,
Kodangattil
S
,
Tseng
E
,
Kopsco
D
, et al
Characterization of Gpr101 expression and G-protein coupling selectivity
.
Brain Res
2006
;
1087
:
1
14
.
44.
Kober
P
,
Bujko
M
,
Oledzki
J
,
Tysarowski
A
,
Siedlecki
JA
. 
Methyl-CpG binding column-based identification of nine genes hypermethylated in colorectal cancer
.
Mol Carcinog
2011
;
50
:
846
56
.
45.
Lee
DK
,
Nguyen
T
,
Lynch
KR
,
Cheng
R
,
Vanti
WB
,
Arkhitko
O
, et al
Discovery and mapping of ten novel G protein-coupled receptor genes
.
Gene
2001
;
275
:
83
91
.
46.
Cho-Clark
M
,
Larco
DO
,
Semsarzadeh
NN
,
Vasta
F
,
Mani
SK
,
Wu
TJ
. 
GnRH-(1-5) transactivates EGFR in Ishikawa human endometrial cells via an orphan G protein-coupled receptor
.
Mol Endocrinol
2014
;
28
:
80
98
.
47.
Rostomyan
L
,
Daly
AF
,
Petrossians
P
,
Natchev
E
,
Lila
AR
,
Lecoq
A-L
, et al
Clinical and genetic characterization of pituitary gigantism: an international study in 208 patients
.
Endocr Relat Cancer
2015
;
22
:
745
57
.
48.
Naves
LA
,
Daly
AF
,
Dias
LA
,
Yuan
B
,
Zakir
JCO
,
Barra
GB
, et al
Aggressive tumor growth and clinical evolution in a patient with X-linked acro-gigantism syndrome
.
Endocrine
2016
;
51
:
236
44
.
49.
Daly
AF
,
Vanbellinghen
J-F
,
Khoo
SK
,
Jaffrain-Rea
M-L
,
Naves
LA
,
Guitelman
MA
, et al
Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families
.
J Clin Endocrinol Metab
2007
;
92
:
1891
6
.
50.
Maxwell
JE
,
Sherman
SK
,
Howe
JR
. 
Translational diagnostics and therapeutics in pancreatic neuroendocrine tumors
.
Clin Cancer Res
2016
;
22
:
5022
9
.
51.
Milne
TA
,
Hughes
CM
,
Lloyd
R
,
Yang
Z
,
Rozenblatt-Rosen
O
,
Dou
Y
, et al
Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors
.
Proc Natl Acad Sci U S A
2005
;
102
:
749
54
.
52.
Gillam
MP
,
Nimbalkar
D
,
Sun
L
,
Christov
K
,
Ray
D
,
Kaldis
P
, et al
MEN1 tumorigenesis in the pituitary and pancreatic islet requires Cdk4 but not Cdk2
.
Oncogene
2015
;
34
:
932
8
.
53.
Balogh
K
,
Patocs
A
,
Hunyady
L
,
Raacz
K
. 
Menin dynamics and functional insight: take your partners
.
Mol Cell Endocrinol
2010
;
326
:
80
4
.
54.
De Laat
JM
,
Dekkers
OM
,
Pieterman
CRC
,
Kluijfhout
WP
,
Hermus
AR
,
Pereira
AM
, et al
Long-term natural course of pituitary tumors in patients with MEN1: results from the DutchMEN1 Study Group (DMSG)
.
J Clin Endocrinol Metab
2015
;
100
:
3288
96
.
55.
Thakker
RV
,
Newey
PJ
,
Walls
GV
,
Bilezikian
J
,
Dralle
H
,
Ebeling
PR
, et al
Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1)
.
J Clin Endocrinol Metab
2012
;
97
:
2990
3011
.
56.
Pellegata
NS
,
Quintanilla-Martinez
L
,
Siggelkow
H
,
Samson
E
,
Bink
K
,
Höfler
H
, et al
Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans
.
Proc Natl Acad Sci U S A
2006
;
103
:
15558
63
.
57.
Sambugaro
S
,
Di Ruvo
M
,
Ambrosio
MR
,
Pellegata
NS
,
Bellio
M
,
Guerra
A
, et al
Early onset acromegaly associated with a novel deletion in CDKN1B 5′UTR region
.
Endocrine
2015
;
49
:
58
64
.
58.
Occhi
G
,
Regazzo
D
,
Trivellin
G
,
Boaretto
F
,
Ciato
D
,
Bobisse
S
, et al
A novel mutation in the upstream open reading frame of the CDKN1B gene causes a MEN4 phenotype
.
PLoS Genet
2013
;
9
:
e1003350
.
59.
Agarwal
SK
,
Mateo
CM
,
Marx
SJ
. 
Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states
.
J Clin Endocrinol Metab
2009
;
94
:
1826
34
.
60.
Carney
JA
,
Gordon
H
,
Carpenter
PC
,
Shenoy
BV
,
Go
VL
. 
The complex of myxomas, spotty pigmentation, and endocrine overactivity
.
Medicine
1985
;
64
:
270
83
.
61.
Correa
R
,
Salpea
P
,
Stratakis
CA
. 
Carney complex: an update
.
Eur J Endocrinol
2015
;
173
:
M85
97
.
62.
Salpea
P
,
Stratakis
CA
. 
Carney complex and McCune Albright syndrome: an overview of clinical manifestations and human molecular genetics
.
Mol Cell Endocrinol
2014
;
386
:
85
91
.
63.
Matyakhina
L
,
Pack
S
,
Kirschner
LS
,
Pak
E
,
Mannan
P
,
Jaikumar
J
, et al
Chromosome 2 (2p16) abnormalities in Carney complex tumours
.
J Med Genet
2003
;
40
:
268
77
.
64.
Almeida
MQ
,
Muchow
M
,
Boikos
S
,
Bauer
AJ
,
Griffin
KJ
,
Tsang
KM
, et al
Mouse Prkar1a haploinsufficiency leads to an increase in tumors in the Trp53+/− or Rb1+/− backgrounds and chemically induced skin papillomas by dysregulation of the cell cycle and Wnt signaling
.
Hum Mol Genet
2010
;
19
:
1387
98
.
65.
Sekine
S
,
Shibata
T
,
Kokubu
A
,
Morishita
Y
,
Noguchi
M
,
Nakanishi
Y
, et al
Craniopharyngiomas of adamantinomatous type harbor beta-catenin gene mutations
.
Am J Pathol
2002
;
161
:
1997
2001
.
66.
De Kock
L
,
Sabbaghian
N
,
Plourde
F
,
Srivastava
A
,
Weber
E
,
Bouron-Dal Soglio
D
, et al
Pituitary blastoma: a pathognomonic feature of germ-line DICER1 mutations
.
Acta Neuropathol
2014
;
128
:
111
22
.
67.
Rio Frio
T
,
Bahubeshi
A
,
Kanellopoulou
C
,
Hamel
N
,
Niedziela
M
,
Sabbaghian
N
, et al
DICER1 mutations in familial multinodular goiter with and without ovarian Sertoli-Leydig cell tumors
.
JAMA
2011
;
305
:
68
77
.
68.
Scheithauer
BW
,
Kovacs
K
,
Horvath
E
,
Kim
DS
,
Osamura
RY
,
Ketterling
RP
, et al
Pituitary blastoma
.
Acta Neuropathol
2008
;
116
:
657
66
.
69.
Choong
CS
,
Priest
JR
,
Foulkes
WD
. 
Exploring the endocrine manifestations of DICER1 mutations
.
Trends Mol Med
2012
;
18
:
503
5
.
70.
O'Toole
SM
,
Dénes
J
,
Robledo
M
,
Stratakis
CA
,
Korbonits
M
. 
The association of pituitary adenomas and phaeochromocytomas or paragangliomas
.
Endocr Relat Cancer
2015
;
22
:
T105
22
.
71.
Dénes
J
,
Swords
F
,
Rattenberry
E
,
Stals
K
,
Owens
M
,
Cranston
T
, 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
:
E531
41
.
72.
Xekouki
P
,
Pacak
K
,
Almeida
M
,
Wassif
CA
,
Rustin
P
,
Nesterova
M
, et al
Succinate dehydrogenase (SDH) D subunit (SDHD) inactivation in a growth-hormone-producing pituitary tumor: a new association for SDH?
J Clin Endocrinol Metab
2012
;
97
:
E357
66
.
73.
Xekouki
P
,
Szarek
E
,
Bullova
P
,
Giubellino
A
,
Quezado
M
,
Mastroyannis
SA
, et al
Pituitary adenoma with paraganglioma/pheochromocytoma (3PAs) and succinate dehydrogenase defects in humans and mice
.
J Clin Endocrinol Metab
2015
;
100
:
E710
9
.
74.
Gill
AJ
,
Toon
CW
,
Clarkson
A
,
Sioson
L
,
Chou
A
,
Winship
I
, et al
Succinate dehydrogenase deficiency is rare in pituitary adenomas
.
Am J Surg Pathol
2014
;
38
:
560
6
.
75.
Xekouki
P
,
Stratakis
CA
. 
Succinate dehydrogenase (SDHx) mutations in pituitary tumors: could this be a new role for mitochondrial complex II and/or Krebs cycle defects?
Endocr Relat Cancer
2012
;
19
:
C33
40
.
76.
Selak
MA
,
Armour
SM
,
MacKenzie
ED
,
Boulahbel
H
,
Watson
DG
,
Mansfield
KD
, et al
Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-a prolyl hydroxylase
.
Cancer Cell
2005
;
7
:
77
85
.
77.
Bardella
C
,
Pollard
PJ
,
Tomlinson
I
. 
SDH mutations in cancer
.
Biochim Biophys Acta
2011
;
1807
:
1432
43
.
78.
Jochmanova
I
,
Pacak
K
. 
Pheochromocytoma: the first metabolic endocrine cancer
.
Clin Cancer Res
2016
;
22
:
5001
11
.
79.
Shan
B
,
Gerez
J
,
Haedo
M
,
Fuertes
M
,
Theodoropoulou
M
,
Buchfelder
M
, et al
RSUME is implicated in HIF-1-induced VEGF-A production in pituitary tumour cells
.
Endocr Relat Cancer
2012
;
19
:
13
27
.
80.
Tufton
N
,
Roncaroli
F
,
Hadjidemetriou
I
,
Dang
MD
,
Dénes
J
,
Guasti
L
, et al
Pituitary carcinoma in a patient with SDHB mutation (abstract)
.
In:
Proceedings of the 15th International Workshop on Multiple Endocrine Neoplasia (WorldMEN 2016)
; 
2016
Sep 29–Oct 1;
Utrecht, the Netherlands. Abstract nr P9.5
.
81.
Newey
PJ
,
Nesbit
MA
,
Rimmer
AJ
,
Head
RA
,
Gorvin
CM
,
Attar
M
, et al
Whole-exome sequencing studies of nonfunctioning pituitary adenomas
.
J Clin Endocrinol Metab
2013
;
98
:
796
800
.
82.
Ronchi
CL
,
Peverelli
E
,
Herterich
S
,
Weigand
I
,
Mantovani
G
,
Schwarzmayr
T
, et al
Landscape of somatic mutations in sporadic GH-secreting pituitary adenomas
.
Eur J Endocrinol
2016
;
174
:
363
72
.
83.
Spada
A
,
Vallar
L
. 
G-protein oncogenes in acromegaly
.
Horm Res
1992
;
38
:
90
3
.
84.
Gadelha
MR
,
Trivellin
G
,
Hernández Ramírez
LC
,
Korbonits
M
. 
Genetics of pituitary adenomas
.
Front Horm Res
2013
;
41
:
111
40
.
85.
Hayward
BE
,
Barlier
A
,
Korbonits
M
,
Grossman
AB
,
Jacquet
P
,
Enjalbert
A
, et al
Imprinting of the G(s)alpha gene GNAS1 in the pathogenesis of acromegaly
.
J Clin Invest
2001
;
107
:
R31
6
.
86.
Reincke
M
,
Sbiera
S
,
Hayakawa
A
,
Theodoropoulou
M
,
Osswald
A
,
Beuschlein
F
, et al
Mutations in the deubiquitinase gene USP8 cause Cushing's disease
.
Nat Genet
2015
;
47
:
31
8
.
87.
Ma
Z-Y
,
Song
Z-J
,
Chen
J-H
,
Wang
Y-F
,
Li
S-Q
,
Zhou
L-F
, et al
Recurrent gain-of-function USP8 mutations in Cushing's disease
.
Cell Res
2015
;
25
:
306
17
.
88.
Theodoropoulou
M
,
Arzberger
T
,
Gruebler
Y
,
Jaffrain-Rea
ML
,
Schlegel
J
,
Schaaf
L
, et al
Expression of epidermal growth factor receptor in neoplastic pituitary cells: evidence for a role in corticotropinoma cells
.
J Endocrinol
2004
;
183
:
385
94
.
89.
Mizuno
E
,
Iura
T
,
Mukai
A
,
Yoshimori
T
,
Kitamura
N
,
Komada
M
. 
Regulation of epidermal growth factor receptor down-regulation by UBPY-mediated deubiquitination at endosomes
.
Mol Biol Cell
2005
;
16
:
5163
74
.
90.
Theodoropoulou
M
,
Reincke
M
,
Fassnacht
M
,
Komada
M
. 
Decoding the genetic basis of cushing's disease: Usp8 in the spotlight
.
Eur J Endocrinol
2015
;
173
:
M73
83
.
91.
Lubke
D
,
Saeger
W
,
Ludecke
DK
. 
Proliferation markers and EGF in ACTH-secreting adenomas and carcinomas of the pituitary
.
Endocr Pathol
1995
;
6
:
45
55
.
92.
Cooper
O
,
Vlotides
G
,
Fukuoka
H
,
Greene
MI
,
Melmed
S
. 
Expression and function of ErbB receptors and ligands in the pituitary
.
Endocr Relat Cancer
2011
;
18
:
R197
211
.
93.
Hayashi
K
,
Inoshita
N
,
Kawaguchi
K
,
Ibrahim Ardisasmita
A
,
Suzuki
H
,
Fukuhara
N
, et al
The USP8 mutational status may predict drug susceptibility in corticotroph adenomas of Cushing's disease
.
Eur J Endocrinol
2016
;
174
:
213
26
.
94.
Fukuoka
H
,
Cooper
O
,
Ben-Shlomo
A
,
Mamelak
A
,
Ren
SG
,
Bruyette
D
, et al
EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas
.
J Clin Invest
2011
;
121
:
4712
21
.
95.
Wierinckx
A
,
Roche
M
,
Raverot
G
,
Legras-Lachuer
C
,
Croze
S
,
Nazaret
N
, et al
Integrated genomic profiling identifies loss of chromosome 11p impacting transcriptomic activity in aggressive pituitary PRL tumors
.
Brain Pathol
2011
;
21
:
533
43
.
96.
Lin
Y
,
Jiang
X
,
Shen
Y
,
Li
M
,
Ma
H
,
Xing
M
, et al
Frequent mutations and amplifications of the PIK3CA gene in pituitary tumors
.
Endocr Relat Cancer
2009
;
16
:
301
10
.
97.
Finelli
P
,
Giardino
D
,
Rizzi
N
,
Buiatiotis
S
,
Virduci
T
,
Franzin
A
, et al
Non-random trisomies of chromosomes 5, 8 and 12 in the prolactinoma sub-type of pituitary adenomas: conventional cytogenetics and interphase FISH study
.
Int J Cancer
2000
;
86
:
344
50
.
98.
Murat
CB
,
Braga
PBS
,
Fortes
MAHZ
,
Bronstein
MD
,
Corrêa-Giannella
MLC
,
Giorgi
RR
. 
Mutation and genomic amplification of the PIK3CA proto-oncogene in pituitary adenomas
.
Brazilian J Med Biol Res
2012
;
45
:
851
5
.
99.
Orloff
MS
,
He
X
,
Peterson
C
,
Chen
F
,
Chen
JL
,
Mester
JL
, et al
Germline PIK3CA and AKT1 mutations in cowden and cowden-like syndromes
.
Am J Hum Genet
2013
;
92
:
76
80
.
100.
Kurelac
I
,
MacKay
A
,
Lambros
MBK
,
Di Cesare
E
,
Cenacchi
G
,
Ceccarelli
C
, et al
Somatic complex I disruptive mitochondrial DNA mutations are modifiers of tumorigenesis that correlate with low genomic instability in pituitary adenomas
.
Hum Mol Genet
2013
;
22
:
226
38
.
101.
Porcelli
AM
,
Ghelli
A
,
Ceccarelli
C
,
Lang
M
,
Cenacchi
G
,
Capristo
M
, et al
The genetic and metabolic signature of oncocytic transformation implicates HIF1alpha destabilization
.
Hum Mol Genet
2010
;
19
:
1019
32
.
102.
Albright
F
,
Butler
AM
,
Hampton
AO
,
Smith
P
. 
Syndrome characterized by osteitis fibrosa disseminata, areas of pigmentation and endocrine dysfunction, with precocious puberty in females
.
N Engl J Med
1937
;
216
:
727
46
.
103.
Chanson
P
,
Salenave
S
,
Orcel
P
. 
McCune-Albright syndrome in adulthood
.
Pediatr Endocrinol Rev
2007
;
4
:
453
62
.
104.
Salenave
S
,
Boyce
AM
,
Collins
MT
,
Chanson
P
. 
Acromegaly and McCune-Albright syndrome
.
J Clin Endocrinol Metab
2014
;
99
:
1955
69
.
105.
Akintoye
SO
,
Kelly
MH
,
Brillante
B
,
Cherman
N
,
Turner
S
,
Butman
JA
, et al
Pegvisomant for the treatment of gsp-mediated growth hormone excess in patients with McCune-Albright syndrome
.
J Clin Endocrinol Metab
2006
;
91
:
2960
6
.
106.
Boyce
AM
,
Glover
M
,
Kelly
MH
,
Brillante
BA
,
Butman
JA
,
Fitzgibbon
EJ
, et al
Optic neuropathy in McCune-Albright syndrome: effects of early diagnosis and treatment of growth hormone excess
.
J Clin Endocrinol Metab
2013
;
98
:
E126
34
.
107.
Daly
AF
,
Yuan
B
,
Fina
F
,
Caberg
J-H
,
Trivellin
G
,
Rostomyan
L
, et al
Somatic mosaicism underlies X-linked acrogigantism (XLAG) syndrome in sporadic male subjects
.
Endocr Relat Cancer
2016
;
23
:
221
33
.
108.
Ferraù
F
,
Romeo
PD
,
Puglisi
S
,
Ragonese
M
,
Torre
ML
,
Scaroni
C
, et al
Analysis of GPR101 and AIP genes mutations in acromegaly: a multicentric study
.
Endocrine.
2016
Jan 27.
[Epub ahead of print]
.
109.
Melmed
S
. 
Pathogenesis of pituitary tumors
.
Nat Rev Endocrinol
2011
;
7
:
257
66
.
110.
Sapochnik
M
,
Nieto
LE
,
Fuertes
M
,
Arzt
E
. 
Molecular mechanisms underlying pituitary pathogenesis
.
Biochem Genet
2016
;
54
:
107
19
.
111.
Robertson
AM
,
Heaney
AP
. 
Molecular markers in pituitary tumors
.
Curr Opin Endocrinol Diabetes Obes
2016
;
23
:
324
30
.
112.
Merid
SK
,
Goranskaya
D
,
Alexeyenko
A
. 
Distinguishing between driver and passenger mutations in individual cancer genomes by network enrichment analysis
.
BMC Bioinformatics
2014
;
15
:
308
.
113.
Raue
F
,
Frank-Raue
K
. 
Thyroid cancer: risk-stratified management and individualized therapy
.
Clin Cancer Res
2016
;
22
:
5012
21
.
114.
Woloschak
M
,
Yu
A
,
Post
KD
. 
Frequent inactivation of the p16 gene in human pituitary tumors by gene methylation
.
Mol Carcinog
1997
;
19
:
221
4
.
115.
Lloyd
RV
,
Jin
L
,
Qian
X
,
Kulig
E
. 
Aberrant p27kip1 expression in endocrine and other tumors
.
Am J Pathol
1997
;
150
:
401
7
.
116.
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
6
.
117.
Xiong
W
,
Knox
AJ
,
Xu
M
,
Kiseljak-Vassiliades
K
,
Colgan
SP
,
Brodsky
KS
, et al
Mammalian Ste20-like kinase 4 promotes pituitary cell proliferation and survival under hypoxia
.
Mol Endocrinol
2015
;
29
:
460
72
.
118.
Xiong
W
,
Matheson
CJ
,
Xu
M
,
Backos
DS
,
Mills
TS
,
Salian-Mehta
S
, et al
Structure-based screen identification of a mammalian Ste20-like kinase 4 (MST4) inhibitor with therapeutic potential for pituitary tumors
.
Mol Cancer Ther
2016
;
15
:
412
20
.
119.
Roussel-Gervais
A
,
Couture
C
,
Langlais
D
,
Takayasu
S
,
Balsalobre
A
,
Rueda
BR
, et al
The Cables1 gene in glucocorticoid regulation of pituitary corticotrope growth and Cushing disease
.
J Clin Endocrinol Metab
2016
;
101
:
513
22
.
120.
Delgrange
E
,
Vasiljevic
A
,
Wierinckx
A
,
François
P
,
Jouanneau
E
,
Raverot
G
, et al
Expression of estrogen receptor alpha is associated with prolactin pituitary tumor prognosis and supports the sex-related difference in tumor growth
.
Eur J Endocrinol
2015
;
172
:
791
801
.
121.
Payabyab
EC
,
Balasubramaniam
S
,
Edgerly
M
,
Velarde
M
,
Merino
MJ
,
Venkatesan
AM
, et al
Adrenocortical cancer: a molecularly complex disease where surgery matters
.
Clin Cancer Res
2016
;
22
:
4989
5000
.
122.
Heaney
AP
. 
Clinical review: pituitary carcinoma: difficult diagnosis and treatment
.
J Clin Endocrinol Metab
2011
;
96
:
3649
60
.
123.
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
:
270
8
.
124.
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
2007
;
66
:
150
2
.
125.
Chesnokova
V
,
Zonis
S
,
Kovacs
K
,
Ben-Shlomo
A
,
Wawrowsky
K
,
Bannykh
S
, et al
p21(Cip1) restrains pituitary tumor growth
.
Proc Natl Acad Sci U S A
2008
;
105
:
17498
503
.
126.
Arzt
E
,
Chesnokova
V
,
Stalla
GK
,
Melmed
S
. 
Pituitary adenoma growth: a model for cellular senescence and cytokine action
.
Cell Cycle
2009
;
8
:
677
8
.
127.
Chesnokova
V
,
Melmed
S
. 
Pituitary senescence: the evolving role of Pttg
.
Mol Cell Endocrinol
2010
;
326
:
55
9
.
128.
Kuilman
T
,
Michaloglou
C
,
Vredeveld
LC
,
Douma
S
,
van Doorn
R
,
Desmet
CJ
, et al
Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network
.
Cell
2008
;
133
:
1019
31
.
129.
Alexandraki
KI
,
Khan
MM
,
Chahal
HS
,
Dalantaeva
NS
,
Trivellin
G
,
Berney
DM
, et al
Oncogene-induced senescence in pituitary adenomas and carcinomas
.
Hormones
2012
;
11
:
297
307
.