U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2024.

Cover of GeneReviews®

GeneReviews® [Internet].

Show details

AIP Familial Isolated Pituitary Adenomas

, MD, PhD and , MD.

Author Information and Affiliations

Initial Posting: ; Last Update: April 16, 2020.

Estimated reading time: 27 minutes

Summary

Clinical characteristics.

AIP familial isolated pituitary adenoma (AIP-FIPA) is defined as the presence of an AIP germline pathogenic variant in an individual with a pituitary adenoma (regardless of family history).

The most commonly occurring pituitary adenomas in this disorder are growth hormone-secreting adenomas (somatotropinoma), followed by prolactin-secreting adenomas (prolactinoma), growth hormone and prolactin co-secreting adenomas (somatomammotropinoma), and nonfunctioning pituitary adenomas (NFPA). Rarely TSH-secreting adenomas (thyrotropinomas) are observed. Clinical findings result from excess hormone secretion, lack of hormone secretion, and/or mass effects (e.g., headaches, visual field loss). Within the same family, pituitary adenomas can be of the same or different type. Age of onset in AIP-FIPA is usually in the second or third decade.

Diagnosis/testing.

The diagnosis of AIP-FIPA is established in a proband with a pituitary adenoma by identification of a heterozygous germline pathogenic variant in AIP by molecular genetic testing.

Management.

Treatment of manifestations: Pituitary adenomas identified in those with AIP-FIPA are generally treated in the same manner as pituitary adenomas of unknown cause: they can be treated by medical therapy (somatostatin analogs, growth hormone receptor antagonists, and dopamine agonists), surgery, and/or radiotherapy. Although surgery is usually performed in persons with AIP-FIPA, it often does not fully control the tumor; thus, medical therapy and radiotherapy following surgery may be required to control hormone output and tumor growth. AIP-FIPA adenomas often do not respond well to first-generation somatostatin analog, while data suggest that they may respond better to second-generation multi-ligand agonists. Prolactinomas are treated with dopamine agonist therapy or surgery and can be aggressive and difficult to treat. NFPA is treated with surgery and if necessary radiotherapy.

Prevention of secondary complications: Expert management for hypopituitarism which can be due to tumor size, surgery, or radiotherapy. Persons on glucocorticoid replacement therapy need to increase their steroid dose when ill or stressed.

Surveillance: In asymptomatic individuals: annual growth assessment and evaluation for signs/symptoms of pituitary adenoma and pubertal development from age four years until adulthood. Continue annual evaluation for signs and symptoms of pituitary adenoma until age 30 years and then every five years between ages 30 and 50 years. Annual pituitary function tests (serum IGF-1, prolactin, estradiol/testosterone, LH, FSH, TSH, free T4) beginning at age four years until age 30; pituitary MRI at age ten years and repeated (every 5 years has been suggested) or as necessary based on clinical and biochemical parameters until age 30 years. Starting at age 30 to 50 years surveillance can be relaxed.

In symptomatic individuals: annual clinical assessment and pituitary function tests (serum IGF-1, spot growth hormone, prolactin, estradiol/testosterone, LH, FSH, TSH, free T4, and morning cortisol); if indicated annual dynamic testing to evaluate for hormone excess or deficiency (e.g., glucose tolerance test, insulin tolerance test); pituitary MRI with frequency depending on clinical status, previous extent of the tumor, and treatment modality. Clinical monitoring of secondary complications of the tumor and/or its treatment (e.g., diabetes mellitus, hypertension, osteoarthritis, hypogonadism, osteoporosis); in those with acromegaly, colonoscopy at age 40 years and repeated every three to ten years depending on the number of colorectal lesions and IGF-1 levels.

Evaluation of relatives at risk: Family members at risk for AIP-FIPA warrant molecular genetic testing for the family-specific pathogenic variant to identify those who harbor the variant and thus require surveillance for pituitary adenomas.

Genetic counseling.

AIP-FIPA is inherited in an autosomal dominant manner. Each child of an individual with AIP-FIPA has a 50% chance of inheriting the pathogenic variant. Prenatal testing for a pregnancy at increased risk is possible if the AIP pathogenic variant of an affected family member has been identified; however, because AIP-FIPA demonstrates reduced penetrance, the finding of an AIP pathogenic variant prenatally does not allow accurate prediction of whether a tumor will develop, or the type of adenoma, age of onset, prognosis, or availability and/or outcome of treatment.

Diagnosis

Suggestive Findings

AIP familial isolated pituitary adenoma (AIP-FIPA) should be suspected in individuals with the following:

  • A pituitary adenoma diagnosed before age 18 years, especially a growth hormone-secreting pituitary adenoma, regardless of family history
  • A pituitary macroadenoma (tumor >10 mm in diameter) diagnosed before age 30 years, especially a growth hormone-secreting pituitary adenoma, regardless of family history
  • A prolactin-secreting pituitary macroadenoma (tumor >10 mm in diameter) diagnosed before age 30 years, regardless of family history
    Note: (1) A germline AIP pathogenic variant is identified in approximately 20% of simplex cases of childhood-onset growth hormone-secreting pituitary adenomas [Chahal et al 2010, Cazabat et al 2011, Tichomirowa et al 2011]. (2) A germline AIP pathogenic variant is identified in 11% of simplex cases of young-onset (age <30 years) pituitary macroadenomas [Tichomirowa et al 2011].
  • A family history of more than one individual with a pituitary adenoma
    Note: (1) A germline AIP pathogenic variant is identified in approximately 20% of families with FIPA [Chahal et al 2010] and in 40% of families in which somatotropinomas are the only tumor type observed. (2) To date, AIP pathogenic variants have not been identified in families with two adults with microprolactinomas (prolactin secreting tumors <10 mm in diameter); therefore, the probability of identifying an AIP pathogenic variant in such a family is low.
  • Absence of clinical features of other disorders associated with pituitary adenomas such as multiple endocrine neoplasia type 1 or type 4 (MEN1 or MEN4) or Carney complex

The pituitary adenomas in individuals with AIP-FIPA can include:

  • Growth hormone-secreting (somatotropinoma)
    Note: Somatotroph (growth hormone-secreting) cell hyperplasia has also been described in individuals with AIP-FIPA, although it is extremely rare.
  • Prolactin-secreting (prolactinoma)
  • Growth hormone and prolactin co-secreting (somatomammotropinoma)
  • Nonfunctioning pituitary adenoma (NFPA)
    Note: Most AIP-related NFPAs show growth hormone and/or prolactin immunostaining in tumor tissue.
  • Thyrotropinoma (TSH-secreting) (rare; 1 thyrotropinoma described)
  • Multihormonal (i.e., secreting >1 pituitary hormone) (extremely rare apart from tumors secreting growth hormone and prolactin)

No unequivocal cases of corticotropinomas have been described.

Establishing the Diagnosis

The diagnosis of AIP-FIPA is established in a proband with a pituitary adenoma(s) by identification of a heterozygous germline pathogenic variant in AIP by molecular genetic testing (see Table 1).

Molecular genetic testing approaches can include a combination of gene-targeted testing (single-gene testing and multigene panel) and comprehensive genomic testing (exome sequencing, exome array, genome sequencing) depending on the phenotype.

Gene-targeted testing requires that the clinician determine which gene(s) are likely involved, whereas genomic testing does not. Because the phenotype of AIP-FIPA can occur with other disorders, individuals with the distinctive findings described in Suggestive Findings are likely to be diagnosed using gene-targeted testing (see Option 1), whereas those with a phenotype indistinguishable from other inherited disorders with pituitary tumors are more likely to be diagnosed using comprehensive genomic testing (see Option 2).

Option 1

When the phenotypic and laboratory findings suggest the diagnosis of AIP-FIPA, molecular genetic testing approaches can include single-gene testing (or concurrent or serial single-gene testing) or use of a multigene panel:

  • Single-gene testing. Sequence analysis of AIP detects small intragenic deletions/insertions and missense, nonsense, and splice site variants; typically, exon or whole-gene deletions/duplications are not detected. Perform sequence analysis first. If no pathogenic variant is found perform gene-targeted deletion/duplication analysis to detect intragenic deletions or duplications.
  • A multigene panel that includes AIP and other genes of interest (see Differential Diagnosis) is most likely to identify the genetic cause of the condition at the most reasonable cost while limiting identification of variants of uncertain significance and pathogenic variants in genes that do not explain the underlying phenotype. Note: (1) The genes included in the panel and the diagnostic sensitivity of the testing used for each gene vary by laboratory and are likely to change over time. (2) Some multigene panels may include genes not associated with the condition discussed in this GeneReview. (3) In some laboratories, panel options may include a custom laboratory-designed panel and/or custom phenotype-focused exome analysis that includes genes specified by the clinician. (4) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests. For this disorder a multigene panel that also includes deletion/duplication analysis is recommended (see Table 1).
    For an introduction to multigene panels click here. More detailed information for clinicians ordering genetic tests can be found here.

Option 2

When the phenotype is indistinguishable from other inherited disorders characterized by pituitary tumors, comprehensive genomic testing (which does not require the clinician to determine which gene[s] are likely involved) is a possible option. Exome sequencing is most commonly used; genome sequencing is also possible.

If exome sequencing is not diagnostic – and particularly when evidence supports autosomal dominant inheritance – exome array (when clinically available) may be considered to detect (multi)exon deletions or duplications that cannot be detected by sequence analysis.

For an introduction to comprehensive genomic testing click here. More detailed information for clinicians ordering genomic testing can be found here.

Table 1.

Molecular Genetic Testing Used in AIP-FIPA

Gene 1MethodProportion of Probands with a Pathogenic Variant 2 Detectable by Method
AIP Sequence analysis 3~95% 4, 5
Gene-targeted deletion/duplication analysis 6~5% 7
1.
2.

See Molecular Genetics for information on allelic variants detected in this gene.

3.

Sequence analysis detects variants that are benign, likely benign, of uncertain significance, likely pathogenic, or pathogenic. Variants may include small intragenic deletions/insertions and missense, nonsense, and splice site variants; typically, exon or whole-gene deletions/duplications are not detected. For issues to consider in interpretation of sequence analysis results, click here.

4.
5.

One promoter variant has been reported (see Molecular Genetics).

6.

Gene-targeted deletion/duplication analysis detects intragenic deletions or duplications. Methods used may include quantitative PCR, long-range PCR, multiplex ligation-dependent probe amplification (MLPA), and a gene-targeted microarray designed to detect single-exon deletions or duplications.

7.

To date, four individuals/families with exon or multiexon deletions and two families with whole-gene deletions have been identified [Georgitsi et al 2008, Igreja et al 2010, Hernández-Ramírez et al 2015, Marques et al 2020].

Clinical Characteristics

Clinical Description

To date, more than 300 individuals with a germline pathogenic variant in AIP have been identified [Beckers et al 2013, Hernández-Ramírez et al 2015]. The following description of the phenotypic features associated with this condition is based on these reports.

Table 2.

Pituitary Adenomas in Individuals with AIP-FIPA

Type of Pituitary Adenoma% of Persons w/Type of AdenomaReference
Somatotropinoma (growth hormone-secreting)70% Hernández-Ramírez et al [2015]
Somatomammotropinoma (growth hormone- and prolactin-secreting)10%
Prolactinoma 10%
Nonfunctioning pituitary adenoma 8%
Thyrotropinoma (TSH-secreting)Rare

The median age of diagnosis of AIP familial isolated pituitary adenoma (AIP-FIPA) is 23 years. The earliest age of diagnosis of a pituitary adenoma in a person with an AIP pathogenic variant is four years [Dutta et al 2019].

Hormone Dysfunction

Somatotropinoma (growth hormone-secreting pituitary adenoma)

  • Acromegaly. Approximately 80% of persons with AIP-FIPA have acromegaly. Persons with acromegaly have excess growth hormone secretion resulting in enlargement of hands and feet, and coarse facial appearance with prognathism and malocclusion of teeth. They may have headaches, joint pain, carpal tunnel syndrome, sleeping difficulties, excessive sweating, hypertension, diabetes mellitus, and muscle weakness. Individuals with longstanding acromegaly often have cardiovascular and rheumatologic/orthopedic complications, which need to be treated accordingly. Individuals with acromegaly of any cause are at increased risk for colon cancer.
    If acromegaly starts in childhood/adolescence it can lead to pituitary gigantism.
  • Pituitary gigantism. Excessive growth hormone secretion before fusion of the growth plates results in pituitary gigantism. Exceptionally tall stature results from a combination of high growth hormone levels and delayed onset of puberty due to suppression of LH/FSH secretion by mass effect of the tumor and/or, when present, the direct effect of high prolactin levels.
    One third of all individuals with a germline AIP pathogenic variant and 40%-50% of individuals with AIP-FIPA with a somatotropinoma have pituitary gigantism [Daly et al 2010].

Prolactinomas. Approximately 10% of persons with an AIP pathogenic variant have a prolactinoma [Daly et al 2010, Igreja et al 2010]. Prolactinomas result in signs and symptoms of prolactin excess (i.e., amenorrhea, sexual problems, galactorrhea, and infertility) and can also cause mass effects (e.g., visual field defects, headaches).

Almost all AIP-related prolactinomas are macroadenomas with male predominance [Daly et al 2010, Igreja et al 2010].

Nonfunctioning pituitary adenomas (NFPAs). NFPAs are seen in 4%-7% of persons with an AIP pathogenic variant.

NFPAs are usually diagnosed due to the local effects of the tumor, such as bitemporal hemianopia or hypogonadism. It is unclear why these silent adenomas do not release hormones at a clinically recognizable level; however, there is likely to be a continuum between fully functional and completely silent adenomas [Drummond et al 2019]. Distinguishing NFPA from prolactinomas can occasionally be difficult due to the stalk effect (pituitary stalk compression resulting in increased prolactin levels in the absence of a prolactin-secreting adenoma).

In AIP-FIPA, NFPAs that have been resected are often (but not always) silent somatotropinoma or lactotroph adenomas [Igreja et al 2010, Villa et al 2011]. In families with AIP-FIPA, NFPAs are identified at a younger age than NFPAs in persons without a germline pathogenic variant [Daly et al 2010]. Screening of clinically unaffected AIP heterozygotes can identify small nonfunctioning pituitary lesions, equivalent to incidentalomas in the general population [Caimari et al 2018].

Thyrotropinomas (TSH-secreting adenomas causing hyperthyroidism) are rarely seen in AIP-FIPA.

A single individual with AIP-FIPA and a thyrotropinoma has been described [Daly et al 2007].

AIP-FIPA does not appear to increase the risk of corticotropinoma. The individuals with FIPA previously reported with Cushing disease were subsequently found to have likely benign variants in AIP, variants of uncertain significance [Beckers et al 2013], or no loss of heterozygosity identified in the tumor [Cazabat et al 2012].

Subfertility is common in persons with pituitary tumors. No data are available specifically regarding subfertility in AIP-FIPA.

Mass effects. Large pituitary adenomas can be associated with deficiencies of other pituitary hormones that result in subfertility, hypothyroidism, hypoadrenalism, low levels of growth hormone, and panhypopituitarism.

Macroadenomas (>10 mm in diameter) may also press on the optic chiasm and optic tracts, causing bitemporal hemianopia. The tumor may invade the adjacent cavernous sinus. Headache can be present in any type of adenoma but is especially common in acromegaly; the mechanism for the increased frequency is unknown.

Larger pituitary tumors may autoinfarct, resulting in pituitary apoplexy (sudden-onset severe headache, visual disturbance, cranial nerve palsies, hypoglycemia, and hypotensive shock). Pituitary apoplexy has been described in individuals with AIP-FIPA [Chahal et al 2011].

Pituitary carcinoma. To date pituitary carcinoma has not been described in an individual with AIP-FIPA.

Other, non-pituitary tumors have been observed in some families with AIP-FIPA; however, because the background population risk for tumors is fairly high and because no consistent pattern has been observed, at present there is no conclusive evidence that an AIP germline pathogenic variant increases the risk for any other tumors. In addition, non-pituitary tumors from AIP heterozygotes have been analyzed for loss of heterozygosity at the AIP locus, and no abnormality was found [Hernández-Ramírez et al 2015].

Genotype-Phenotype Correlations

Individuals with AIP truncating pathogenic variants may have a slightly earlier age of onset and diagnosis compared to those with non-truncating pathogenic variants [Hernández-Ramírez et al 2015].

Penetrance

Studies on large families with AIP pathogenic variants show a clinical penetrance of pituitary adenomas of approximately 23% (range 15%-30%) [Vierimaa et al 2006, Naves et al 2007, Williams et al 2014, Hernández-Ramírez et al 2015]. Although some families with AIP-FIPA can show high penetrance, the higher levels of penetrance initially reported in some families is probably ascertainment bias due to insufficient information on all at-risk family members (e.g., lack of medical records, information on pituitary hormone testing, and/or imaging studies) [Daly et al 2007, Leontiou et al 2008].

The factors influencing penetrance are not known; the possibility of a second locus has been investigated but not confirmed [Khoo et al 2009, Hernández-Ramírez et al 2015].

Nomenclature

Previously, pituitary adenoma predisposition (PAP) syndrome was used to refer to individuals who had an AIP pathogenic variant; the term is not used widely.

Prevalence

The exact prevalence of AIP-FIPA is not known. To date, about 150 families and about 150 simplex cases (i.e., a single occurrence in a family) of AIP-FIPA have been identified [Daly et al 2010, Hernández-Ramírez et al 2015, Caimari et al 2018].

Differential Diagnosis

In children more often than in adults, pituitary adenomas may be a manifestation of a genetic condition. Pituitary adenomas of genetic origin can be divided into isolated and syndromic categories.

Familial Isolated Pituitary Adenomas (FIPA)

FIPA is defined as a hereditary condition associated with pituitary adenomas and no other features of a syndrome known to be associated with pituitary adenomas.

X-linked acrogigantism (XLAG), a second genetically characterized type of FIPA, is caused by duplication of GPR101. XLAG, a highly penetrant disorder, is associated with pituitary hyperplasia or adenoma resulting in growth hormone excess with onset in infancy, usually with associated hyperprolactinemia [Trivellin et al 2014]. Most individuals with XLAG have a de novo somatic mosaic genetic alteration not inherited from a parent.

Families with FIPA of known or unknown cause can have homogeneous pituitary adenoma phenotypes (i.e., pituitary tumors of the same type) or heterogeneous phenotypes (i.e., pituitary tumors of different types).

Aspects of FIPA that tend to differ between families in which a germline AIP pathogenic variant has been identified and those in which no germline AIP pathogenic variant has been identified include: age of onset, number of persons affected in the family, male-to-female ratio, and typical adenoma types. Tumor variables may also include: size, aggressiveness, and response to treatment [Hernández-Ramírez et al 2015] (see Table 3).

Table 3.

Comparison of Findings in Persons with Isolated Pituitary Adenomas by Family History and Presence/Absence of a Germline AIP Pathogenic Variant

CharacteristicsFamilial Isolated Pituitary AdenomaSimplex Somatotropinoma 1
AIP-FIPAXLAGFIPA of unknown cause
Clinical features Age of onset 24-24 yrsInfancy / early childhood40 yrs43 yrs
Average # of affected family members 33-412-3NA
Male-to-female ratio 41:1 to 2:11:21:11:1
Adenoma features Somatotropinomas / somatomammotropinomas70%-80%~100%~50%NA
SizeMacroadenomas in vast majorityPituitary hyperplasia-macroadenomaMajority macroadenoma 5Smaller
AggressivenessMoreVariableMoreLess
Response to treatmentPoorerPoorerPoorerBetter

Syndromes Associated with Pituitary Tumors

Table 4.

Syndromes Associated with Pituitary Tumors

Gene(s)DisorderMOIPituitary Tumor FeaturesOther Features
MEN1 Multiple endocrine neoplasia type 1 (MEN1)ADPituitary tumors occur in ~40% of affected individuals, most often prolactinomas
  • Gastro-entero-pancreatic tract tumors
  • Parathyroid adenoma w/hypercalcemia
  • Other manifestations
CDKN1A
CDKN1B
CDKN2B
CDKN2C
MEN1-like syndrome 1ADPituitary tumors occur in ~40% of affected individuals, most often somatotropinomas
  • Rare disorder
  • Clinical findings similar to those of MEN1
PRKAR1A
PRKACB 2
Carney complex AD~80% of affected individuals have somatotroph cell hyperplasia or small pituitary adenoma
  • Skin pigmentary abnormalities
  • Myxomas of the organs
  • Schwannomas
  • Primary pigmented nodular adrenocortical disease
  • Large-cell calcifying Sertoli cell tumors
  • Thyroid nodules
  • Acromegaly
GNAS McCune-Albright syndrome NA (somatic)
  • ~30% of affected individuals have pituitary disease
  • Pituitary adenomas w/↑ secretion of growth hormone
  • Hyperplasia of somatomammotroph cells w/prolactinemia
  • Polyostotic fibrous dysplasia
  • Café au lait patches
  • Multiple endocrine disorders (e.g., multinodular goiters, multinodular adrenal hyperplasia, & precocious puberty)
MAX
SDHA
SDHB
SDHC
SDHD
RET
Hereditary paraganglioma-pheochromocytoma syndromes AD
  • Low penetrance of pituitary disease
  • Pituitary carcinoma described, vacuolated histology picture
  • Paragangliomas
  • Pheochromocytoma
  • GIST
  • Kidney tumors
DICER1 DICER1 syndrome AD
  • Low penetrance of pituitary disease
  • ACTH-secreting pituitary blastoma
Manifests before age 2 yrs
MSH2
MSH6
MLH1
?PMS2
Lynch syndrome AD
  • Low penetrance of pituitary disease
  • ACTH-secreting macroadenomas
Colorectal, endometrial, ovarian, & other carcinomas
USP8 Pituitary adenoma 4 (OMIM 219090)AD1 individual w/pituitary adenomaDevelopmental delay

AD = autosomal dominant; GIST = gastrointestinal stromal tumor; MEN1 = multiple endocrine neoplasia type 1; MOI = mode of inheritance

1.
2.

One individual with Carney complex (<1% of families with Carney complex) had a germline rearrangement resulting in four copies of PRKACB [Forlino et al 2014]. PRKACB encodes the catalytic subunit Cβ of the cyclic AMP-dependent protein kinase A (PKA). Levels of Cβ and PKA activity were increased in the individual's lymphoblasts and fibroblasts; the authors propose that this is a Carney complex-causing gain-of-function variant.

Note: Autopsy and radiologic studies suggest that 14%-22% of the population may harbor a pituitary adenoma, most of these being asymptomatic [Ezzat et al 2004]. Thus, it is possible for two pituitary adenomas, especially prolactinomas, to occur sporadically in a family by chance.

Other Space-Occupying Lesions

In addition to pituitary adenomas, numerous space-occupying lesions can occur in the pituitary fossa [Saeger et al 2007]. The most common space-occupying lesions after pituitary adenomas are craniopharyngiomas, which cause symptoms by compressing the normal pituitary, resulting in hormonal deficiencies and mass effects on the surrounding tissues and brain [Zacharia et al 2012].

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs of an individual with AIP familial isolated pituitary adenoma (AIP-FIPA), the evaluations in Table 5 are recommended (for details see Katznelson et al [2011]).

Table 5.

Recommended Evaluations Following Initial Diagnosis in Individuals with AIP-FIPA

System/ConcernEvaluationComment
Pituitary adenoma
  • LH, FSH, testosterone/estradiol
  • Visual field eval
  • Consultation w/endocrinologist
  • If adenoma suspected: pituitary MRI to detect size & extent
  • DXA scan in those w/hypogonadism
Somatotropinoma
(growth hormone-
secreting)
  • Evaluate for signs/symptoms (e.g., stature, change in facial appearance, change in shoe size, problems w/ring sizes, headache, excessive sweating, joint pains, carpal tunnel syndrome).
  • Spot serum growth hormone, IGF-1
Include:
  • Review of serial photographs for acromegalic changes
  • Measurement of parental heights
  • GTT in persons w/findings of acromegaly
  • Assess ACTH reserve if needed.
Prolactinoma
  • Evaluate for signs/symptoms (e.g., menstrual history, galactorrhea, infertility, low libido, impotence).
  • Serum prolactin
Note: (1) High prolactin levels can be due to stalk effect; (2) There are many non-pituitary causes of hyperprolactinemia.
NFPAs Evaluate for signs/symptoms (e.g., headache, lack of other pituitary hormones, visual field problems).Many nonfunctioning adenomas are identified incidentally w/no clinical or biochemical associations.
Thyrotropinoma TSH, free T4Consider other causes of thyroid hormone resistance.
Other Consultation w/clinical geneticist &/or genetic counselor

GTT = glucose tolerance test; NFPAs = nonfunctioning pituitary adenomas

Treatment of Manifestations

While there is experience with treating pituitary adenomas in symptomatic persons with FIPA, the experience of management and treatment of persons identified prospectively through clinical screening due to family history of FIPA and/or presence of a heterozygous germline AIP pathogenic variant is relatively recent. However, the age of detection in the AIP-FIPA is earlier and the prognosis generally better. Given the prevalence of incidental pituitary adenomas, it is important to remember that such a tumor may arise in an individual with an AIP pathogenic variant, completely by chance. The following recommendations are based on those of Katznelson et al [2011], Melmed et al [2011], Williams et al [2014], and Hernández-Ramírez et al [2015].

Table 6.

Treatment of Manifestations in Individuals with AIP-FIPA

Manifestation/ConcernTreatmentConsiderations/Other
Pituitary microadenomas Medical therapy (e.g., somatostatin analogs, growth hormone receptor antagonists, & dopamine agonists), surgery &/or radiotherapyMonitor microadenomas w/normal clinical & biochemistry findings closely.
Pituitary macroadenomas Transsphenoidal surgery, medical therapy, &/or radiotherapySurgery often does not fully control the tumor; large recurring tumors may require radiotherapy if tumor invades neighboring anatomic structures (e.g., cavernous sinus).
Somatotropinomas
  • Radiotherapy (conventional or radiosurgery) for growing adenomas, for which repeat surgery is unlikely to control hormone levels
  • Standard treatment of cardiovascular & rheumatologic/orthopedic complications for those w/acromegaly
Tumors often do not respond to medical therapy w/1st-generation somatostatin analogs; 1 2nd-generation may have more effect. 2
Prolactinomas
  • Dopamine agonist therapy (e.g., cabergoline)
  • Surgical treatment often used for macroprolactinoma (diameter >10 mm)
Prolactinomas in AIP-FIPA can be aggressive & difficult to treat. 1
Nonfunctioning pituitary adenomas Surgery & (if necessary) radiotherapyUsually do not respond to traditional somatostatin analogs

Prevention of Secondary Complications

Tumor size, surgery, and/or radiotherapy can cause hypopituitarism, which requires careful expert follow up.

Persons on glucocorticoid replacement therapy need to increase their steroid dose when ill or stressed.

Surveillance

No formal guidelines regarding surveillance of persons with AIP-FIPA have been established. The following recommendations are based on expert opinion from the literature and on the authors' personal experience with more than 200 persons with symptomatic or asymptomatic AIP-FIPA.

Table 7.

Recommended Surveillance for Individuals with AIP-FIPA

System/ConcernEvaluationFrequency
Pituitary adenoma
  • Measure height & weight; calculate height velocity.
  • Evaluate for signs/symptoms & evaluate pubertal development.
Annually beginning at age 4 yrs until adulthood 1
Evaluate for signs/symptoms.Annually until age 30 yrs & every 5 yrs between ages 30 & 50 yrs, earlier if symptomatic
Serum IGF-1, prolactin, estradiol/testosterone, LH, FSH, TSH, free T4Annually from age 4 yrs to 30 yrs; to date, no secreting pituitary adenomas have developed after age 30 in those w/normal findings at age 30 yrs. Blood tests if symptomatic after age 30 yrs.
Pituitary MRI
  • Baseline at age 10 yrs unless indicated earlier due to clinical findings
  • Repeat MRI every 5 yrs until age 30 yrs (if clinical & pituitary function tests remain normal)
  • If clinical or biochemical abnormality between ages 30 & 50 yrs
1.

In children between ages four and ten years, it may be difficult to get annual blood samples. In these cases, monitoring symptoms and growth may be an acceptable alternative, as non-growth hormone-secreting adenomas before age ten years are rare.

Table 8.

Recommended Additional Surveillance for Symptomatic Individuals with AIP-FIPA

System/ConcernEvaluationFrequency
History of pituitary
adenoma
  • Clinical assessment
  • Serum IGF-1, spot growth hormone, prolactin, estradiol/testosterone, LH, FSH, TSH, free T4, morning cortisol
  • If necessary dynamic testing (e.g., glucose tolerance test, insulin tolerance test) to evaluate for hormone excess or deficiency
Annually
Pituitary MRIFrequency depends on clinical status, previous extent of tumor, & treatment modality.
Osteoporosis in those
w/hypogonadism
DXA evalPer established guidelines
Complications of
acromegaly
Monitor for diabetes mellitus, hypertension, hypogonadism, & osteoarthritis.Per established guidelines
Colonoscopy
  • At age 40 yrs
  • Repeat every 3 to 10 yrs depending on # of colorectal lesions on initial colonoscopy & IGF-1 levels. 1

Evaluation of Relatives at Risk

It is appropriate to clarify the genetic status of apparently asymptomatic at-risk relatives of an affected individual by molecular genetic testing for the familial AIP pathogenic variant so that morbidity and mortality can be reduced by early diagnosis and treatment. Identification of at-risk family members may also reduce the need for costly screening procedures in those family members who have not inherited a pathogenic variant.

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.

Pregnancy Management

Pregnancy may increase the size of a growth hormone-secreting adenoma or a prolactin-secreting adenoma (especially macroadenomas); thus, a pregnant woman with pituitary macroadenoma is at risk of developing visual field defects. In each trimester it is appropriate to inquire about headaches and perform visual field testing. Medical therapies are stopped during pregnancy.

See MotherToBaby for further information on medication use during pregnancy.

Therapies Under Investigation

A study that is actively ascertaining individuals with FIPA or childhood-onset pituitary adenoma is listed in ClinicalTrials.gov.

Growth hormone receptor antagonists block the action of endogenous growth hormone, thereby controlling disease manifestations such as headaches, soft tissue enlargement, diabetes mellitus, hypertension, and high IGF-1 levels. A growth hormone receptor antagonist has been used successfully in persons with AIP-FIPA and acromegaly and pituitary gigantism [Goldenberg et al 2008]. In two individuals with AIP-related pituitary tumors resistant to treatment with first-generation somatostatin analogs, pasireotide, a second-generation multi-ligand somatostatin analog with affinity to multiple somatostatin receptors, was shown to achieve long-term control of disease [Daly et al 2019]. Although growth hormone receptor antagonists are currently not licensed for pediatric use, several case reports have shown their effectiveness, especially when IGF-1 levels need to be reduced immediately to prevent abnormally rapid growth [Higham et al 2010, Dutta et al 2019].

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for access to information on clinical studies for a wide range of diseases and conditions.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, mode(s) of inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members; it is not meant to address all personal, cultural, or ethical issues that may arise or to substitute for consultation with a genetics professional. —ED.

Mode of Inheritance

AIP familial isolated pituitary adenoma (AIP-FIPA) is inherited in an autosomal dominant manner with reduced penetrance.

Risk to Family Members

Parents of a proband

  • Almost all individuals reported to date with AIP-FIPA have a parent who is also heterozygous for the AIP pathogenic variant [Hernández-Ramírez et al 2015]. Because of reduced penetrance, the parent may or may not be affected.
  • Reports of unequivocally de novo pathogenic variants are rare [Ramírez-Rentería et al 2016]. As not all simplex AIP-FIPA cases (i.e., a single occurrence in a family) have been evaluated sufficiently to determine if the pathogenic variant occurred de novo in the proband, the proportion of AIP-FIPA resulting from a de novo AIP pathogenic variant is unknown.
  • Molecular genetic testing is recommended for the parents of a proband with an apparently negative family history of the disease and a pathogenic AIP variant.
  • If the AIP pathogenic variant found in the proband cannot be detected in the leukocyte DNA of either parent, possible explanations include a de novo pathogenic variant in the proband or germline mosaicism in a parent.* Although no instances of germline mosaicism have been reported, it remains a possibility.
    * Misattributed parentage can also be explored as an alternative explanation for an apparent de novo pathogenic variant.
  • The family history of some individuals diagnosed with AIP-FIPA may appear to be negative because of failure to recognize the disorder in family members due to a milder phenotypic presentation or reduced penetrance. Therefore, an apparently negative family history cannot be confirmed unless molecular genetic testing has been performed on the parents of the proband.

Sibs of a proband. The risk to the sibs of the proband depends on the genetic status of the proband's parents:

Offspring of a proband. Each child of an individual heterozygous for an AIP pathogenic variant has a 50% chance of inheriting the AIP pathogenic variant.

Other family members. The risk to other family members depends on the status of the proband's parents; if a parent is heterozygous for an AIP pathogenic variant, his or her family members may be at risk for AIP-FIPA.

Related Genetic Counseling Issues

See Management, Evaluation of Relatives at Risk for information on evaluating at-risk relatives for the purpose of early diagnosis and treatment.

Family planning

  • The optimal time for determination of genetic risk and discussion of the availability of prenatal/preimplantation genetic testing is before pregnancy.
  • It is appropriate to offer genetic counseling (including discussion of potential risks to offspring and reproductive options) to young adults who are affected or at risk.

DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. Because it is likely that testing methodology and our understanding of genes, allelic variants, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals.

Prenatal Testing and Preimplantation Genetic Testing

Once the AIP pathogenic variant has been identified in the family, prenatal and preimplantation genetic testing for AIP-FIPA are possible. As AIP-FIPA demonstrates reduced penetrance, the finding of a pathogenic variant in AIP prenatally does not allow accurate prediction of a tumor, the adenoma type, age of onset, prognosis, or availability and/or outcome of treatment.

Differences in perspective may exist among medical professionals and within families regarding the use of prenatal testing, particularly if the testing is being considered for the purpose of pregnancy termination rather than early diagnosis. While use of prenatal testing is a personal decision, discussion of these issues may be helpful.

Resources

GeneReviews staff has selected the following disease-specific and/or umbrella support organizations and/or registries for the benefit of individuals with this disorder and their families. GeneReviews is not responsible for the information provided by other organizations. For information on selection criteria, click here.

Molecular Genetics

Information in the Molecular Genetics and OMIM tables may differ from that elsewhere in the GeneReview: tables may contain more recent information. —ED.

Table A.

AIP Familial Isolated Pituitary Adenomas : Genes and Databases

GeneChromosome LocusProteinLocus-Specific DatabasesHGMDClinVar
AIP 11q13​.2 AH receptor-interacting protein AIP database
AIP Gene Mutations
AIP AIP

Data are compiled from the following standard references: gene from HGNC; chromosome locus from OMIM; protein from UniProt. For a description of databases (Locus Specific, HGMD, ClinVar) to which links are provided, click here.

Table B.

OMIM Entries for AIP Familial Isolated Pituitary Adenomas (View All in OMIM)

102200PITUITARY ADENOMA 1, MULTIPLE TYPES; PITA1
605555ARYL HYDROCARBON RECEPTOR-INTERACTING PROTEIN; AIP

Molecular Pathogenesis

AIP encodes AIP, a co-chaperone with several interacting partners. In the context of pituitary adenomas, AIP behaves as a tumor suppressor gene [Leontiou et al 2008]. AIP, previously known as hepatitis B virus (HBV) X-associated protein (XAP2) or aryl hydrocarbon receptor (AhR)-associated protein (ARA9), is a 330-amino acid 37-kd protein [Kuzhandaivelu et al 1996, Carver & Bradfield 1997]. The C-terminal end of the protein has three tetratricopeptide repeats (TPRs) and a final alpha-helix. The three TPR domains are degenerate sequences of 34 amino acids comprising two antiparallel helices that play a crucial role in mediating the protein-protein interactions of AIP [Kazlauskas et al 2002].

Mechanism of disease causation. The majority (75%) of AIP pathogenic variants result in, or predict, a truncated protein. Many of the pathogenic missense variants affect structurally important conserved amino acids of the TPR structure [Vargiolu et al 2009, Igreja et al 2010, Cai et al 2011]. Clinical data and functional studies both indicate a tumor suppressor role for AIP.

Of note, truncating variants have been reported throughout the protein. Some are predicted to cause nonsense-mediated decay, while others lose the functionally important C-terminal alpha helix. In addition, truncating variants may result in protein with a shortened half-life. Shortened protein half-life has also been shown for many of the pathogenic missense variants [Hernández-Ramírez et al 2016].

Table 9.

Notable AIP Pathogenic Variants

Reference SequencesDNA Nucleotide ChangePredicted Protein ChangeComment [Reference]
NM_003977​.3
NP_003968​.3
c.40C>Tp.Gln14TerFinnish founder variant [Vierimaa et al 2006]
c.241C>Tp.Arg81TerMutational hot spot identified in apparently independent families from Brazil, USA, India, & UK [Chahal et al 2010, Beckers et al 2013]
c.805_825dupp.Phe269_His275dupEnglish/European founder variant [Salvatori et al 2017]
c.910C>Tp.Arg304TerThe most common mutational hot spot; Irish, Romanian, English, Italian, Indian & Mexican families described; a founder effect in some regions (e.g., Ireland) [Chahal et al 2010, Beckers et al 2013]

Variants listed in the table have been provided by the authors. GeneReviews staff have not independently verified the classification of variants.

GeneReviews follows the standard naming conventions of the Human Genome Variation Society (varnomen​.hgvs.org). See Quick Reference for an explanation of nomenclature.

References

Published Guidelines / Consensus Statements

  • Katznelson L, Atkinson JL, Cook DM, Ezzat SZ, Hamrahian AH, Miller KK, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the diagnosis and treatment of acromegaly--2011 update. Endocr Pract. 2011;17 Suppl 4:1–44. [PubMed: 21846616]

Literature Cited

  • 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. [PMC free article: PMC2684477] [PubMed: 19141585]
  • 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. [PMC free article: PMC3610678] [PubMed: 23371967]
  • Cai W, Kramarova TV, Berg P, Korbonits M, Pongratz I. The immunophilin-like protein XAP2 is a negative regulator of estrogen signaling through interaction with estrogen receptor α. PLoS One. 2011;6:e25201. [PMC free article: PMC3184960] [PubMed: 21984905]
  • Cairns SR, Scholefield JH, Steele RJ, Dunlop MG, Thomas HJ, Evans GD, Eaden JA, Rutter MD, Atkin WP, Saunders BP, Lucassen A, Jenkins P, Fairclough PD, Woodhouse CR, et al. Guidelines for colorectal cancer screening and surveillance in moderate and high risk groups (update from 2002). Gut. 2010;59:666–89. [PubMed: 20427401]
  • Caimari F, Hernández-Ramírez LC, Dang MN, Gabrovska P, Iacovazzo D, Stals K, Ellard S, Korbonits M. Risk category system to identify pituitary adenoma patients with AIP mutations. J Med Genet. 2018;55:254–60. [PMC free article: PMC5869708] [PubMed: 29440248]
  • Carver LA, Bradfield CA. Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J Biol Chem. 1997;272:11452–6. [PubMed: 9111057]
  • Cazabat L, Bouligand J, Chanson P. AIP mutation in pituitary adenomas. N Engl J Med. 2011;364:1973–4. [PubMed: 21591954]
  • Cazabat L, Guillaud-Bataille M, Bertherat J, Raffin-Sanson ML. Mutations of the gene for the aryl hydrocarbon receptor-interacting protein in pituitary adenomas. Horm Res. 2009;71:132–41. [PubMed: 19188737]
  • Cazabat L, Bouligand J, Salenave S, Bernier M, Gaillard S, Parker F, Young J, Guiochon-Mantel A, Chanson P. Germline AIP mutations in apparently sporadic pituitary adenomas: prevalence in a prospective single-center cohort of 443 patients. J Clin Endocrinol Metab. 2012;97:E663–70. [PubMed: 22319033]
  • Chahal HS, Chapple JP, Frohman LA, Grossman AB, Korbonits M. Clinical, genetic and molecular characterization of patients with familial isolated pituitary adenomas (FIPA). Trends Endocrinol Metab. 2010;21:419–27. [PubMed: 20570174]
  • Chahal HS, Stals K, Unterländer M, Balding DJ, Thomas MG, Kumar AV, Besser GM, Atkinson AB, Morrison PJ, Howlett TA, Levy MJ, Orme SM, Akker SA, Abel RL, Grossman AB, Burger J, Ellard S, Korbonits M. AIP mutation in pituitary adenomas in the 18th century and today. N Engl J Med. 2011;364:43–50. [PubMed: 21208107]
  • Daly AF, Tichomirowa MA, Petrossians P, Heliövaara E, Jaffrain-Rea ML, Barlier A, Naves LA, Ebeling T, Karhu A, Raappana A, Cazabat L, De Menis E, Montañana CF, Raverot G, Weil RJ, Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E, Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo RA, Sabaté MI, Villa C, Popelier M, Salvatori R, Jennings J, Longás AF, Labarta Aizpún JI, Georgitsi M, Paschke R, Ronchi C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT, Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A, Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen LA, Beckers A. Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab. 2010;95:E373–83. [PubMed: 20685857]
  • Daly AF, Vanbellinghen JF, Khoo SK, Jaffrain-Rea ML, Naves LA, Guitelman MA, Murat A, Emy P, Gimenez-Roqueplo AP, Tamburrano G, Raverot G, Barlier A, De Herder W, Penfornis A, Ciccarelli E, Estour B, Lecomte P, Gatta B, Chabre O, Sabaté MI, Bertagna X, Garcia Basavilbaso N, Stalldecker G, Colao A, Ferolla P, Wémeau JL, Caron P, Sadoul JL, Oneto A, Archambeaud F, Calender A, Sinilnikova O, Montañana CF, Cavagnini F, Hana V, Solano A, Delettieres D, Luccio-Camelo DC, Basso A, Rohmer V, Brue T, Bours V, Teh BT, Beckers A. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92:1891–6. [PubMed: 17244780]
  • Daly AF, Yuan B, Fina F, Caberg JH, Trivellin G, Rostomyan L, de Herder WW, Naves LA, Metzger D, Cuny T, Rabl W, Shah N, Jaffrain-Rea ML, Zatelli MC, Faucz FR, Castermans E, Nanni-Metellus I, Lodish M, Muhammad A, Palmeira L, Potorac I, Mantovani G, Neggers SJ, Klein M, Barlier A, Liu P, Ouafik L, Bours V, Lupski JR, Stratakis CA, Beckers A. Somatic mosaicism underlies X-linked acrogigantism syndrome in sporadic male subjects. Endocr Relat Cancer. 2016;23:221–33. [PMC free article: PMC4877443] [PubMed: 26935837]
  • Daly AF, Rostomyan L, Betea D, Bonneville JF, Villa C, Pellegata NS, Waser B, Reubi JC, Waeber Stephan C, Christ E, Beckers A. AIP-mutated acromegaly resistant to first-generation somatostatin analogs: long-term control with pasireotide LAR in two patients. Endocr Connect. 2019;8:367–77. [PMC free article: PMC6454377] [PubMed: 30851160]
  • Drummond J, Roncaroli F, Grossman AB, Korbonits M. Clinical and pathological aspects of silent pituitary adenomas. J Clin Endocrinol Metab. 2019;104:2473–89. [PMC free article: PMC6517166] [PubMed: 30020466]
  • Dutta P, Reddy KS, Rai A, Madugundu AK, Solanki HS, Bhansali A, Radotra BD, Kumar N, Collier D, Iacovazzo D, Gupta P, Raja R, Gowda H, Pandey A, Devgun JS, Korbonits M. Surgery, octreotide, temozolomide, bevacizumab, radiotherapy, and pegvisomant treatment of an AIP mutation positive child. J Clin Endocrinol Metab. 2019;104:3539–44. [PMC free article: PMC6619489] [PubMed: 31125088]
  • Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML, McCutcheon IE. The prevalence of pituitary adenomas: a systematic review. Cancer. 2004;101:613–9. [PubMed: 15274075]
  • Forlino A, Vetro A, Garavelli L, Ciccone R, London E, Stratakis CA, Zuffardi O. PRKACB and Carney complex. N Engl J Med. 2014;370:1065–7. [PubMed: 24571725]
  • Georgitsi M, Heliövaara E, Paschke R, Kumar AV, Tischkowitz M, Vierimaa O, Salmela P, Sane T, De Menis E, Cannavò S, Gündogdu S, Lucassen A, Izatt L, Aylwin S, Bano G, Hodgson S, Koch CA, Karhu A, Aaltonen LA. Large genomic deletions in AIP in pituitary adenoma predisposition. J Clin Endocrinol Metab. 2008;93:4146–51. [PubMed: 18628514]
  • Goldenberg N, Racine MS, Thomas P, Degnan B, Chandler W, Barkan A. Treatment of pituitary gigantism with the growth hormone receptor antagonist pegvisomant. J Clin Endocrinol Metab. 2008;93:2953–6. [PMC free article: PMC2515082] [PubMed: 18492755]
  • Hernández-Ramírez LC, Gabrovska P, Denes J, Stals K, Trivellin G, Tilley D, Ferrau F, Evanson J, Ellard S, Grossman AB, Roncaroli F, Gadelha MR, Korbonits M, International FC. Landscape of familial isolated and young-onset pituitary adenomas: prospective diagnosis in AIP mutation carriers. J Clin Endocrinol Metab. 2015;100:E1242–54. [PMC free article: PMC4570169] [PubMed: 26186299]
  • Hernández-Ramírez LC, Martucci F, Morgan RM, Trivellin G, Tilley D, Ramos-Guajardo N, Iacovazzo D, D'Acquisto F, Prodromou C, Korbonits M. Rapid proteasomal degradation of mutant proteins is the primary mechanism leading to tumorigenesis in patients with missense AIP mutations. J Clin Endocrinol Metab. 2016;101:3144–54. [PMC free article: PMC4971335] [PubMed: 27253664]
  • Higham CE, Emy P, Ferone D, Finke R, Laurberg P, Main K, Maffei P, Martini C, Minuto F, Rainegard I, Salzgeber K, Vogel C, Koltowska-Haggstrom M, Trainer PJ. Treatment experience in 11 patients with gigantism. Endocrine Abstracts. 2010;21:280.
  • Igreja S, Chahal HS, King P, Bolger GB, Srirangalingam U, Guasti L, Chapple JP, Trivellin G, Gueorguiev M, Guegan K, Stals K, Khoo B, Kumar AV, Ellard S, Grossman AB, Korbonits M, et al. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Hum Mutat. 2010;31:950–60. [PMC free article: PMC3065644] [PubMed: 20506337]
  • Katznelson L, Atkinson JL, Cook DM, Ezzat SZ, Hamrahian AH, Miller KK, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the diagnosis and treatment of acromegaly--2011 update. Endocr Pract. 2011;17 Suppl 4:1–44. [PubMed: 21846616]
  • Kazlauskas A, Poellinger L, Pongratz I. Two distinct regions of the immunophilin-like protein XAP2 regulate dioxin receptor function and interaction with hsp90. J Biol Chem. 2002;277:11795–801. [PubMed: 11805120]
  • Khoo SK, Pendek R, Nickolov R, Luccio-Camelo DC, Newton TL, Massie A, Petillo D, Menon J, Cameron D, Teh BT, Chan SP. Genome-wide scan identifies novel modifier loci of acromegalic phenotypes for isolated familial somatotropinoma. Endocr Relat Cancer. 2009;16:1057–63. [PubMed: 19443539]
  • Kuzhandaivelu N, Cong YS, Inouye C, Yang WM, Seto E. XAP2, a novel hepatitis B virus X-associated protein that inhibits X transactivation. Nucleic Acids Res. 1996;24:4741–50. [PMC free article: PMC146319] [PubMed: 8972861]
  • Leontiou CA, Gueorguiev M, van der Spuy J, Quinton R, Lolli F, Hassan S, Chahal HS, Igreja SC, Jordan S, Rowe J, Stolbrink M, Christian HC, Wray J, Bishop-Bailey D, Berney DM, Wass JA, Popovic V, Ribeiro-Oliveira A Jr, Gadelha MR, Monson JP, Akker SA, Davis JR, Clayton RN, Yoshimoto K, Iwata T, Matsuno A, Eguchi K, Musat M, Flanagan D, Peters G, Bolger GB, Chapple JP, Frohman LA, Grossman AB, Korbonits M. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J Clin Endocrinol Metab. 2008;93:2390–401. [PubMed: 18381572]
  • Marques P, Caimari F, Hernández-Ramírez LC, Collier D, Iacovazzo D, Ronaldson A, Magid K, Lim CT, Stals K, Ellard S, Grossman AB, Korbonits M, et al. Significant benefits of AIP testing and clinical screening in familial isolated and young-onset pituitary tumors. J Clin Endocrinol Metab. 2020:105. Epub ahead of print. [PMC free article: PMC7137887] [PubMed: 31996917]
  • Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, Wass JA, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96:273–88. [PubMed: 21296991]
  • Naves LA, Daly AF, Vanbellinghen JF, Casulari LA, Spilioti C, Magalhães AV, Azevedo MF, Giacomini LA, Nascimento PP, Nunes RO, Rosa JW, Jaffrain-Rea ML, Bours V, Beckers A. Variable pathological and clinical features of a large Brazilian family harboring a mutation in the aryl hydrocarbon receptor-interacting protein gene. Eur J Endocrinol. 2007;157:383–91. [PubMed: 17893251]
  • Rahbari R, Wuster A, Lindsay SJ, Hardwick RJ, Alexandrov LB, Turki SA, Dominiczak A, Morris A, Porteous D, Smith B, Stratton MR, Hurles ME, et al. Timing, rates and spectra of human germline mutation. Nat Genet. 2016;48:126–33. [PMC free article: PMC4731925] [PubMed: 26656846]
  • Ramírez-Rentería C, Hernández-Ramírez LC, Portocarrero-Ortiz L, Vargas G, Melgar V, Espinosa E, Espinosa-de-Los-Monteros AL, Sosa E, Gonzalez B, Zuniga S, Unterlander M, Burger J, Stals K, Bussell AM, Ellard S, Dang M, Iacovazzo D, Kapur S, Gabrovska P, Radian S, Roncaroli F, Korbonits M, Mercado M. AIP mutations in young patients with acromegaly and the Tampico Giant: the Mexican experience. Endocrine. 2016;53:402–11. [PubMed: 27033541]
  • Salvatori R, Radian S, Diekmann Y, Iacovazzo D, David A, Gabrovska P, Grassi G, Bussell AM, Stals K, Weber A, Quinton R, Crowne EC, Corazzini V, Metherell L, Kearney T, Du Plessis D, Sinha AK, Baborie A, Lecoq AL, Chanson P, Ansorge O, Ellard S, Trainer PJ, Balding D, Thomas MG, Korbonits M. In-frame seven amino-acid duplication in AIP arose over the last 3000 years, disrupts protein interaction and stability and is associated with gigantism. Eur J Endocrinol. 2017;177:257–66. [PMC free article: PMC5510572] [PubMed: 28634279]
  • Saeger W, Lüdecke 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;156:203–16. [PubMed: 17287410]
  • Tichomirowa MA, Barlier A, Daly AF, Jaffrain-Rea ML, Ronchi C, Yaneva M, Urban JD, Petrossians P, Elenkova A, Tabarin A, Desailloud R, Maiter D, Schürmeyer T, Cozzi R, Theodoropoulou M, Sievers C, Bernabeu I, Naves LA, Chabre O, Montañana CF, Hana V, Halaby G, Delemer B, Aizpún JI, Sonnet E, Longás AF, Hagelstein MT, Caron P, Stalla GK, Bours V, Zacharieva S, Spada A, Brue T, Beckers A. High prevalence of AIP gene mutations following focused screening in young patients with sporadic pituitary macroadenomas. Eur J Endocrinol. 2011;165:509–15. [PubMed: 21753072]
  • Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, Schernthaner-Reiter MH, Szarek E, Leal LF, Caberg JH, Castermans E, Villa C, Dimopoulos A, Chittiboina P, Xekouki P, Shah N, Metzger D, Lysy PA, Ferrante E, Strebkova N, Mazerkina N, Zatelli MC, Lodish M, Horvath A, de Alexandre RB, Manning AD, Levy I, Keil MF, Sierra MD, Palmeira L, Coppieters W, Georges M, Naves LA, Jamar M, Bours V, Wu TJ, Choong CS, Bertherat J, Chanson P, Kamenicky P, Farrell WE, Barlier A, Quezado M, Bjelobaba I, Stojilkovic SS, Wess J, Costanzi S, Liu P, Lupski JR, Beckers A, Stratakis CA. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med. 2014;371:2363–74. [PMC free article: PMC4291174] [PubMed: 25470569]
  • Vargiolu M, Fusco D, Kurelac I, Dirnberger D, Baumeister R, Morra I, Melcarne A, Rimondini R, Romeo G, Bonora E. The tyrosine kinase receptor RET interacts in vivo with aryl hydrocarbon receptor-interacting protein to alter survivin availability. J Clin Endocrinol Metab. 2009;94:2571–8. [PubMed: 19366855]
  • Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, Gündogdu S, De Menis E, Mäkinen MJ, Launonen V, Karhu A, Aaltonen LA. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science. 2006;312:1228–30. [PubMed: 16728643]
  • Villa C, Lagonigro MS, Magri F, Koziak M, Jaffrain-Rea ML, Brauner R, Bouligand J, Junier MP, Di Rocco F, Sainte-Rose C, Beckers A, Roux FX, Daly AF, Chiovato L. Hyperplasia-adenoma sequence in pituitary tumorigenesis related to aryl hydrocarbon receptor interacting protein gene mutation. Endocr Relat Cancer. 2011;18:347–56. [PubMed: 21450940]
  • Williams F, Hunter S, Bradley L, Chahal HS, Storr H, Akker SA, Kumar AV, Orme SM, Evanson J, Morrison PJ, Korbonits M, Atkinson AB. 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. [PubMed: 24423289]
  • Zacharia BE, Bruce SS, Goldstein H, Malone HR, Neugut AI, Bruce JN. Incidence, treatment and survival of patients with craniopharyngioma in the surveillance, epidemiology and end results program. Neuro Oncol. 2012;14:1070–8. [PMC free article: PMC3408265] [PubMed: 22735773]

Chapter Notes

Author Notes

Website: www.fipapatients.org

The FIPA Patients website, established by Dr Korbonits in collaboration with the FIPA Consortium, is an information resource for patients and families with familial isolated pituitary adenoma. It also provides general information for medical professionals on research in the field of FIPA, including links to relevant publications.

The authors welcome comments and inquiries: gro.stneitapapif@ofni.

Acknowledgments

We are grateful to referring colleagues and patients for providing information on this disease.

Revision History

  • 16 April 2020 (sw) Comprehensive update posted live
  • 21 June 2012 (me) Review posted live
  • 3 February 2011 (mk) Original submission
Copyright © 1993-2024, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

GeneReviews® chapters are owned by the University of Washington. Permission is hereby granted to reproduce, distribute, and translate copies of content materials for noncommercial research purposes only, provided that (i) credit for source (http://www.genereviews.org/) and copyright (© 1993-2024 University of Washington) are included with each copy; (ii) a link to the original material is provided whenever the material is published elsewhere on the Web; and (iii) reproducers, distributors, and/or translators comply with the GeneReviews® Copyright Notice and Usage Disclaimer. No further modifications are allowed. For clarity, excerpts of GeneReviews chapters for use in lab reports and clinic notes are a permitted use.

For more information, see the GeneReviews® Copyright Notice and Usage Disclaimer.

For questions regarding permissions or whether a specified use is allowed, contact: ude.wu@tssamda.

Bookshelf ID: NBK97965PMID: 22720333

Views

Tests in GTR by Gene

Related information

  • OMIM
    Related OMIM records
  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed
  • Gene
    Locus Links

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...