117
Views
2
CrossRef citations to date
0
Altmetric
Review

Potential markers of disease behavior in acromegaly and gigantism

ORCID Icon
Pages 171-183 | Received 06 Dec 2019, Accepted 26 Mar 2020, Published online: 06 May 2020

References

  • Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest. 2009;119:3189–3202.
  • Lavrentaki A, Paluzzi A, Wass JA, et al. Epidemiology of acromegaly: review of population studies. Pituitary. 2017;20:4–9.
  • Colao A, Grasso LFS, Giustina A, et al. Acromegaly. Nat Rev Dis Primers. 2019;5:20.
  • Gadelha MR, Kasuki L, Lim DST, et al. Systemic complications of acromegaly and the impact of the current treatment landscape: an update. Endocr Rev. 2019;40:268–332.
  • Katznelson L, Laws ER Jr., Melmed S, et al. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99:3933–3951.
  • Kasuki L, Wildemberg LE, Gadelha MR. Management of endocrine disease: personalized medicine in the treatment of acromegaly. Eur J Endocrinol. 2018;178:R89–R100.
  • Puig Domingo M. Treatment of acromegaly in the era of personalized and predictive medicine. Clin Endocrinol (Oxf). 2015;83:3–14.
  • Fernandez-Rodriguez E, Casanueva FF, Bernabeu I. Update on prognostic factors in acromegaly: is a risk score possible? Pituitary. 2015;18:431–440.
  • Caimari F, Korbonits M. Novel genetic causes of pituitary adenomas. Clin Cancer Res. 2016;22:5030–5042.
  • Stratakis CA, Tichomirowa MA, Boikos S, 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;78:457–463.
  • Tichomirowa MA, Barlier A, Daly AF, et al. High prevalence of AIP gene mutations following focused screening in young patients with sporadic pituitary macroadenomas. Eur J Endocrinol. 2011;165:509–515.
  • Rostomyan L, Daly AF, Petrossians P, et al. Clinical and genetic characterization of pituitary gigantism: an international collaborative study in 208 patients. Endocr Relat Cancer. 2015;22:745–757.
  • Iacovazzo D, Caswell R, Bunce B, et al. Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study. Acta Neuropathol Commun. 2016;4:56.
  • Gadelha MR, Kasuki L, Korbonits M. The genetic background of acromegaly. Pituitary. 2017;20:10–21.
  • Valdes-Socin H, Poncin J, Stevens V, et al. Familial isolated pituitary adenomas unrelated to MEN1 mutations: A follow-up of 27 patients. 10th meeting of the Belgian Endocrine Society. Ann Endocrinol (Paris). 2000;61:343.
  • Daly AF, Rixhon M, Adam C, et al. High prevalence of pituitary adenomas: a cross-sectional study in the province of Liege, Belgium. J Clin Endocrinol Metab. 2006;91:4769–4775.
  • Daly AF, Tichomirowa MA, Petrossians P, 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.
  • Igreja S, Chahal HS, King P, et al. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Hum Mutat. 2010;31:950–960.
  • Beckers A, Aaltonen LA, Daly AF, et al. 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;34:239–277.
  • Daly AF, Vanbellinghen JF, Khoo SK, et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92:1891–1896.
  • Denes J, Swords F, Rattenberry E, 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.
  • Xekouki P, Szarek E, Bullova P, et al. Pituitary adenoma with paraganglioma/pheochromocytoma (3PAs) and succinate dehydrogenase defects in humans and mice. J Clin Endocrinol Metab. 2015;100:E710–E719.
  • De Sousa SM, McCabe MJ, Wu K, et al. Germline variants in familial pituitary tumour syndrome genes are common in young patients and families with additional endocrine tumours. Eur J Endocrinol. 2017;176:635–644.
  • Iacovazzo D, Hernández-Ramírez LC, Korbonits M. Sporadic pituitary adenomas: the role of germline mutations and recommendations for genetic screening. Expert Rev Endocrinol Metab. 2017;12:143–153.
  • Hernández-Ramírez LC, Gabrovska P, Dénes J, 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.
  • Marques P, Caimari F, Hernandez-Ramirez LC, et al. Significant benefits of AIP testing and clinical screening in familial isolated and young-onset pituitary tumors. J Clin Endocrinol Metab. 2020. DOI:10.1210/clinem/dgaa040.
  • Xekouki P, Mastroyiannis SA, Avgeropoulos D, et al. Familial pituitary apoplexy as the only presentation of a novel AIP mutation. Endocr Relat Cancer. 2013;20:L11–L14.
  • Naves LA, Daly AF, Vanbellinghen JF, et al. 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;157:383–391.
  • Leontiou CA, Gueorguiev M, van der Spuy J, et al. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J Clin Endocrinol Metab. 2008;93:2390–2401.
  • Daly A, Rostomyan L, Betea D, et al. AIP-mutated acromegaly resistant to first-generation somatostatin analogs: long-term control with pasireotide LAR in two patients. Endocr Connect. 2019;8(4):367–377.
  • Williams F, Hunter S, Bradley L, 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;99:1122–1131.
  • Trivellin G, Daly AF, Faucz FR, et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med. 2014;371:2363–2374.
  • Iacovazzo D, Korbonits M. Gigantism: X-linked acrogigantism and GPR101 mutations. Growth Horm IGF Res. 2016;30-31:64–69.
  • Beckers A, Lodish M, Giampaolo T, et al. X-linked acrogigantism (X-LAG) syndrome: clinical profile and therapeutic responses. Endocr Relat Cancer. 2015;22:353–367.
  • Gordon RJ, Bell J, Chung WK, et al. Childhood acromegaly due to X-linked acrogigantism: long term follow-up. Pituitary. 2016;19:560–564.
  • Trivellin G, Hernandez-Ramirez LC, Swan J, et al. An orphan G-protein-coupled receptor causes human gigantism and/or acromegaly: molecular biology and clinical correlations. Best Pract Res Clin Endocrinol Metab. 2018;32:125–140.
  • Wise-Oringer BK, Zanazzi GJ, Gordon RJ, et al. Familial X-linked acrogigantism: postnatal outcomes and tumor pathology in a prenatally diagnosed infant and his mother. J Clin Endocrinol Metab. 2019;104:4667–4675.
  • Daly AF, Yuan B, Fina F, et al. Somatic mosaicism underlies X-linked acrogigantism syndrome in sporadic male subjects. Endocr Relat Cancer. 2016;23:221–233.
  • Daly AF, Lysy PA, Desfilles C, et al. GHRH excess and blockade in X-LAG syndrome. Endocr Relat Cancer. 2016;23:161–170.
  • Rodd C, Millette M, Iacovazzo D, et al. Somatic GPR101 duplication causing X-Linked Acrogigantism (XLAG)-diagnosis and management. J Clin Endocrinol Metab. 2016;101:1927–1930.
  • Wermer P. Genetic aspects of adenomatosis of endocrine glands. Am J Med. 1954;16:363–371.
  • Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4). Mol Cell Endocrinol. 2014;386:2–15.
  • Machens A, Schaaf L, Karges W, et al. Age-related penetrance of endocrine tumours in multiple endocrine neoplasia type 1 (MEN1): a multicentre study of 258 gene carriers. Clin Endocrinol (Oxf). 2007;67:613–622.
  • Thakker RV, Newey PJ, Walls GV, et al. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012;97:2990–3011.
  • Goudet P, Dalac A, Le Bras M, et al. MEN1 disease occurring before 21 years old: a 160-patient cohort study from the groupe d’etude des tumeurs endocrines. J Clin Endocrinol Metab. 2015;100:1568–1577.
  • Verges B, Boureille F, Goudet P, et al. Pituitary disease in MEN type 1 (MEN1): data from the France-Belgium MEN1 multicenter study. J Clin Endocrinol Metab. 2002;87:457–465.
  • de Laat JM, Dekkers OM, Pieterman CR, 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;100:3288–3296.
  • Trouillas J, Labat-Moleur F, Sturm N, et al. Pituitary tumors and hyperplasia in multiple endocrine neoplasia type 1 syndrome (MEN1): a case-control study in a series of 77 patients versus 2509 non-MEN1 patients. Am J Surg Pathol. 2008;32:534–543.
  • Pellegata NS, Quintanilla-Martinez L, Siggelkow H, et al. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci USA. 2006;103:15558–15563.
  • 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;94:1826–1834.
  • Georgitsi M. MEN-4 and other multiple endocrine neoplasias due to cyclin-dependent kinase inhibitors (p27(Kip1) and p18(INK4C)) mutations. Best Pract Res Clin Endocrinol Metab. 2010;24:425–437.
  • Frederiksen A, Rossing M, Hermann P, et al. Clinical features of multiple endocrine neoplasia type 4: novel pathogenic variant and review of published cases. J Clin Endocrinol Metab. 2019;104:3637–3646.
  • Xekouki P, Pacak K, Almeida 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.
  • O’Toole SM, Denes J, Robledo M, et al. 15 YEARS OF PARAGANGLIOMA: the association of pituitary adenomas and phaeochromocytomas or paragangliomas. Endocr Relat Cancer. 2015;22:T105–22.
  • Niemeijer ND, Papathomas TG, Korpershoek E, et al. Succinate Dehydrogenase (SDH)-deficient pancreatic neuroendocrine tumor expands the SDH-related tumor spectrum. J Clin Endocrinol Metab. 2015;100:E1386–93.
  • Niemeijer ND, Rijken JA, Eijkelenkamp K, et al. The phenotype of SDHB germline mutation carriers: a nationwide study. Eur J Endocrinol. 2017;177:115–125.
  • Dumitrescu CE, Collins MT. McCune-Albright syndrome. Orphanet J Rare Dis. 2008;3:12.
  • Weinstein LS, Shenker A, Gejman PV, et al. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med. 1991;325:1688–1695.
  • Akintoye SO, Chebli C, Booher S, et al. Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. J Clin Endocrinol Metab. 2002;87:5104–5112.
  • Kovacs K, Horvath E, Thorner MO, et al. Mammosomatotroph hyperplasia associated with acromegaly and hyperprolactinemia in a patient with the McCune-Albright syndrome. A histologic, immunocytologic and ultrastructural study of the surgically-removed adenohypophysis. Virchows Arch A Pathol Anat Histopathol. 1984;403:77–86.
  • Vortmeyer AO, Glasker S, Mehta GU, et al. Somatic GNAS mutation causes widespread and diffuse pituitary disease in acromegalic patients with McCune-Albright syndrome. J Clin Endocrinol Metab. 2012;97:2404–2413.
  • Galland F, Kamenicky P, Affres H, et al. McCune-Albright syndrome and acromegaly: effects of hypothalamopituitary radiotherapy and/or pegvisomant in somatostatin analog-resistant patients. J Clin Endocrinol Metab. 2006;91:4957–4961.
  • Stratakis CA, Kirschner LS, Carney JA. Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. J Clin Endocrinol Metab. 2001;86:4041–4046.
  • Horvath A, Bertherat J, Groussin L, et al. Mutations and polymorphisms in the gene encoding regulatory subunit type 1-alpha of protein kinase A (PRKAR1A): an update. Hum Mutat. 2010;31:369–379.
  • Carney JA, Gordon H, Carpenter PC, et al. The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine (Baltimore). 1985;64:270–283.
  • Stratakis CA, Carney JA, Lin JP, et al. Carney complex, a familial multiple neoplasia and lentiginosis syndrome. Analysis of 11 kindreds and linkage to the short arm of chromosome 2. J Clin Invest. 1996;97:699–705.
  • Kirschner LS, Carney JA, Pack SD, 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.
  • Forlino A, Vetro A, Garavelli L, et al. PRKACB and Carney complex. N Engl J Med. 2014;370:1065–1067.
  • Salpea P, Horvath A, London E, et al. Deletions of the PRKAR1A Locus at 17q24.2-q24.3 in Carney complex: genotype-phenotype correlations and implications for genetic testing. J Clin Endocrinol Metab. 2014;99:E183–E188.
  • Bertherat J, Horvath A, Groussin L, et al. Mutations in regulatory subunit type 1A of cyclic adenosine 5ʹ-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients and 80 different genotypes. J Clin Endocrinol Metab. 2009;94:2085–2091.
  • Pack SD, Kirschner LS, Pak E, et al. Genetic and histologic studies of somatomammotropic pituitary tumors in patients with the “complex of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas” (Carney complex). J Clin Endocrinol Metab. 2000;85:3860–3865.
  • Ly KI, Blakeley JO. The diagnosis and management of neurofibromatosis type 1. Med Clin North Am. 2019;103:1035–1054.
  • Cambiaso P, Galassi S, Palmiero M, et al. Growth hormone excess in children with neurofibromatosis type-1 and optic glioma. Am J Med Genet A. 2017;173:2353–2358.
  • Hannah-Shmouni F, Demidowich AP, Rowell J, et al. Large pituitary gland with an expanding lesion in the context of neurofibromatosis 1. BMJ Case Rep. 2017.
  • Hozumi K, Fukuoka H, Odake Y, et al. Acromegaly caused by a somatotroph adenoma in patient with neurofibromatosis type 1. Endocr J. 2019;66:853–857.
  • Bizzarri C, Bottaro G. Endocrine implications of neurofibromatosis 1 in childhood. Horm Res Paediatr. 2015;83:232–241.
  • Caimari F, Hernández-Ramírez LC, Dang MN, et al. Risk category system to identify pituitary adenoma patients with AIP mutations. J Med Genet. 2018;55:254–260.
  • Mete OK,M, Osamura RY, Trouillas J, et al. Somatotroph adenoma. In: Lloyd RVO,RY, Klöppel G, Rosai J, editors. WHO classification of tumours of endocrine organs. 4th ed., Vol. 10. World Health Organization International Agency for Research on Cancer; Lyon, France. 2017. p. 19–23.
  • Larkin S, Reddy R, Karavitaki N, et al. Granulation pattern, but not GSP or GHR mutation, is associated with clinical characteristics in somatostatin-naive patients with somatotroph adenomas. Eur J Endocrinol. 2013;168:491–499.
  • Kiseljak-Vassiliades K, Carlson NE, Borges MT, et al. Growth hormone tumor histological subtypes predict response to surgical and medical therapy. Endocrine. 2015;49:231–241.
  • Iacovazzo D, Carlsen E, Lugli F, et al. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol. 2016;174:241–250.
  • Mazal PR, Czech T, Sedivy R, et al. Prognostic relevance of intracytoplasmic cytokeratin pattern, hormone expression profile, and cell proliferation in pituitary adenomas of akromegalic patients. Clin Neuropathol. 2001;20:163–171.
  • Obari A, Sano T, Ohyama K, et al. Clinicopathological features of growth hormone-producing pituitary adenomas: difference among various types defined by cytokeratin distribution pattern including a transitional form. Endocr Pathol. 2008;19:82–91.
  • Bakhtiar Y, Hirano H, Arita K, et al. Relationship between cytokeratin staining patterns and clinico-pathological features in somatotropinomae. Eur J Endocrinol. 2010;163:531–539.
  • Heck A, Ringstad G, Fougner SL, et al. Intensity of pituitary adenoma on T2-weighted magnetic resonance imaging predicts the response to octreotide treatment in newly diagnosed acromegaly. Clin Endocrinol (Oxf). 2012;77:72–78.
  • Kato M, Inoshita N, Sugiyama T, et al. Differential expression of genes related to drug responsiveness between sparsely and densely granulated somatotroph adenomas. Endocr J. 2012;59:221–228.
  • Brzana J, Yedinak CG, Gultekin SH, et al. Growth hormone granulation pattern and somatostatin receptor subtype 2A correlate with postoperative somatostatin receptor ligand response in acromegaly: a large single center experience. Pituitary. 2013;16:490–498.
  • Kiseljak-Vassiliades K, Xu M, Mills TS, et al. Differential somatostatin receptor (SSTR) 1-5 expression and downstream effectors in histologic subtypes of growth hormone pituitary tumors. Mol Cell Endocrinol. 2015;417:73–83.
  • Fougner SL, Lekva T, Borota OC, et al. The expression of E-cadherin in somatotroph pituitary adenomas is related to tumor size, invasiveness, and somatostatin analog response. J Clin Endocrinol Metab. 2010;95:2334–2342.
  • Lekva T, Berg JP, Fougner SL, et al. Gene expression profiling identifies ESRP1 as a potential regulator of epithelial mesenchymal transition in somatotroph adenomas from a large cohort of patients with acromegaly. J Clin Endocrinol Metab. 2012;97:E1506–E1514.
  • Corbetta S, Ballare E, Mantovani G, et al. Somatostatin receptor subtype 2 and 5 in human GH-secreting pituitary adenomas: analysis of gene sequence and mRNA expression. Eur J Clin Invest. 2001;31:208–214.
  • Taboada GF, Luque RM, Bastos W, et al. Quantitative analysis of somatostatin receptor subtype (SSTR1-5) gene expression levels in somatotropinomas and non-functioning pituitary adenomas. Eur J Endocrinol. 2007;156:65–74.
  • Taboada GF, Luque RM, Neto LV, et al. Quantitative analysis of somatostatin receptor subtypes (1-5) gene expression levels in somatotropinomas and correlation to in vivo hormonal and tumor volume responses to treatment with octreotide LAR. Eur J Endocrinol. 2008;158:295–303.
  • Casar-Borota O, Heck A, Schulz S, et al. Expression of SSTR2a, but not of SSTRs 1, 3, or 5 in somatotroph adenomas assessed by monoclonal antibodies was reduced by octreotide and correlated with the acute and long-term effects of octreotide. J Clin Endocrinol Metab. 2013;98:E1730–9.
  • Venegas-Moreno E, Vazquez-Borrego MC, Dios E, et al. Association between dopamine and somatostatin receptor expression and pharmacological response to somatostatin analogues in acromegaly. J Cell Mol Med. 2018;22:1640–1649.
  • Fischer T, Doll C, Jacobs S, et al. Reassessment of sst2 somatostatin receptor expression in human normal and neoplastic tissues using the novel rabbit monoclonal antibody UMB-1. J Clin Endocrinol Metab. 2008;93:4519–4524.
  • Gatto F, Feelders RA, van der Pas R, et al. Immunoreactivity score using an anti-sst2A receptor monoclonal antibody strongly predicts the biochemical response to adjuvant treatment with somatostatin analogs in acromegaly. J Clin Endocrinol Metab. 2013;98:E66–71.
  • Liu W, Xie L, He M, et al. Expression of somatostatin receptor 2 in somatotropinoma correlated with the short-term efficacy of somatostatin analogues. Int J Endocrinol. 2017;2017:9606985.
  • Gatto F, Barbieri F, Arvigo M, et al. Biological and biochemical basis of the differential efficacy of first and second generation somatostatin receptor ligands in neuroendocrine neoplasms. Int J Mol Sci. 2019;20:3940.
  • Gatto F, Feelders RA, Franck SE, et al. In vitro head-to-head comparison between octreotide and pasireotide in GH-secreting pituitary adenomas. J Clin Endocrinol Metab. 2017;102:2009–2018.
  • Muhammad A, Coopmans EC, Gatto F, et al. Pasireotide responsiveness in acromegaly is mainly driven by somatostatin receptor subtype 2 expression. J Clin Endocrinol Metab. 2019;104:915–924.
  • Hofland LJ, van der Hoek J, van Koetsveld PM, et al. The novel somatostatin analog SOM230 is a potent inhibitor of hormone release by growth hormone- and prolactin-secreting pituitary adenomas in vitro. J Clin Endocrinol Metab. 2004;89:1577–1585.
  • Duran-Prado M, Saveanu A, Luque RM, et al. A potential inhibitory role for the new truncated variant of somatostatin receptor 5, sst5TMD4, in pituitary adenomas poorly responsive to somatostatin analogs. J Clin Endocrinol Metab. 2010;95:2497–2502.
  • Luque RM, Ibáñez-Costa A, Neto LV, et al. Truncated somatostatin receptor variant sst5TMD4 confers aggressive features (proliferation, invasion and reduced octreotide response) to somatotropinomas. Cancer Lett. 2015;359:299–306.
  • Ballare E, Persani L, Lania AG, et al. Mutation of somatostatin receptor type 5 in an acromegalic patient resistant to somatostatin analog treatment. J Clin Endocrinol Metab. 2001;86:3809–3814.
  • Filopanti M, Ballare E, Lania AG, et al. Loss of heterozygosity at the SS receptor type 5 locus in human GH- and TSH-secreting pituitary adenomas. J Endocrinol Invest. 2004;27:937–942.
  • Mantovani G, Treppiedi D, Giardino E, et al. Cytoskeleton actin-binding proteins in clinical behavior of pituitary tumors. Endocr Relat Cancer. 2019;26:R95–R108.
  • Coelho MCA, Vasquez ML, Wildemberg LE, et al. Molecular evidence and clinical importance of beta-arrestins expression in patients with acromegaly. J Cell Mol Med. 2018;22:2110–2116.
  • Peverelli E, Giardino E, Treppiedi D, et al. Filamin A (FLNA) plays an essential role in somatostatin receptor 2 (SST2) signaling and stabilization after agonist stimulation in human and rat somatotroph tumor cells. Endocrinology. 2014;155:2932–2941.
  • Gatto F, Biermasz NR, Feelders RA, et al. Low beta-arrestin expression correlates with the responsiveness to long-term somatostatin analog treatment in acromegaly. Eur J Endocrinol. 2016;174:651–662.
  • Gatto F, Feelders R, van der Pas R, et al. beta-Arrestin 1 and 2 and G protein-coupled receptor kinase 2 expression in pituitary adenomas: role in the regulation of response to somatostatin analogue treatment in patients with acromegaly. Endocrinology. 2013;154:4715–4725.
  • Theodoropoulou M, Zhang J, Laupheimer S, 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;66:1576–1582.
  • Pagotto U, Arzberger T, Theodoropoulou M, et al. The expression of the antiproliferative gene ZAC is lost or highly reduced in nonfunctioning pituitary adenomas. Cancer Res. 2000;60:6794–6799.
  • Jaffrain-Rea ML, Angelini M, Gargano D, et al. Expression of aryl hydrocarbon receptor (AHR) and AHR-interacting protein in pituitary adenomas: pathological and clinical implications. Endocr Relat Cancer. 2009;16:1029–1043.
  • Kasuki Jomori de PL, Vieira NL, Armondi Wildemberg LE, 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.
  • Kasuki L, Colli LM, Elias PC, et al. Resistance to octreotide LAR in acromegalic patients with high SSTR2 expression: analysis of AIP expression. Arq Bras Endocrinol Metabol. 2012;56:501–506. .
  • Kasuki L, Neto L, Wildemberg LEA, 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–L29.
  • Chahal HS, Trivellin G, Leontiou CA, et al. Somatostatin analogs modulate AIP in somatotroph adenomas: the role of the ZAC1 pathway. J Clin Endocrinol Metab. 2012;97:E1411–20.
  • Umahara M, Okada S, Ohshima K, et al. Glucose-dependent insulinotropic polypeptide induced growth hormone secretion in acromegaly. Endocr J. 2003;50:643–650.
  • Occhi G, Losa M, Albiger N, et al. The glucose-dependent insulinotropic polypeptide receptor is overexpressed amongst GNAS1 mutation-negative somatotropinomas and drives growth hormone (GH)-promoter activity in GH3 cells. J Neuroendocrinol. 2011;23:641–649.
  • Regazzo D, Losa M, Albiger NM, et al. The GIP/GIPR axis is functionally linked to GH-secretion increase in a significant proportion of gsp- somatotropinomas. Eur J Endocrinol. 2017;176:543–553.
  • Herman V, Fagin J, Gonsky R, et al. Clonal origin of pituitary adenomas. J Clin Endocrinol Metab. 1990;71:1427–1433.
  • Gadelha MR, Trivellin G, Hernández-Ramírez LC, et al. Genetics of pituitary adenomas. Front Horm Res. 2013;41:111–140.
  • Suhardja A, Kovacs K, Rutka J. Genetic basis of pituitary adenoma invasiveness: a review. J Neurooncol. 2001;52:195–204.
  • Ewing I, Pedder-Smith S, Franchi G, et al. A mutation and expression analysis of the oncogene BRAF in pituitary adenomas. Clin Endocrinol (Oxf). 2007;66:348–352.
  • Chamberlain CE, Scheel DW, McGlynn K, et al. Menin determines K-RAS proliferative outputs in endocrine cells. J Clin Invest. 2014;124:4093–4101.
  • Landis CA, Masters SB, Spada A, et al. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature. 1989;340:692–696.
  • Weinstein LS, Liu J, Sakamoto A, et al. Minireview: GNAS: normal and abnormal functions. Endocrinology. 2004;145:5459–5464.
  • Hayward BE, Barlier A, Korbonits M, et al. Imprinting of the G(s)alpha gene GNAS1 in the pathogenesis of acromegaly. J Clin Invest. 2001;107:R31–R36.
  • Mantovani G, Bondioni S, Lania AG, et al. Parental origin of Gsalpha mutations in the McCune-Albright syndrome and in isolated endocrine tumors. J Clin Endocrinol Metab. 2004;89:3007–3009.
  • Spada A, Arosio M, Bochicchio D, et al. Clinical, biochemical, and morphological correlates in patients bearing growth hormone-secreting pituitary-tumors with or without constitutively active adenylyl cyclase. J Clin Endocrinol Metab. 1990;71:1421–1426.
  • Buchfelder M, Fahlbusch R, Merz T, et al. Clinical correlates in acromegalic patients with pituitary tumors expressing GSP oncogenes. Pituitary. 1999;1:181–185.
  • Mayr B, Buslei R, Theodoropoulou M, et al. Molecular and functional properties of densely and sparsely granulated GH-producing pituitary adenomas. Eur J Endocrinol. 2013;169:391–400.
  • Efstathiadou ZA, Bargiota A, Chrisoulidou A, et al. Impact of gsp mutations in somatotroph pituitary adenomas on growth hormone response to somatostatin analogs: a meta-analysis. Pituitary. 2015;18:861–867.
  • Valimaki N, Demir H, Pitkanen E, et al. Whole-genome sequencing of Growth Hormone (GH)-secreting pituitary adenomas. J Clin Endocrinol Metab. 2015;100:3918–3927.
  • Ronchi CL, Peverelli E, Herterich S, et al. Landscape of somatic mutations in sporadic GH-secreting pituitary adenomas. Eur J Endocrinol. 2016;174(3):363–372.
  • Hage M, Viengchareun S, Brunet E, et al. Genomic alterations and complex subclonal architecture in sporadic GH-secreting pituitary adenomas. J Clin Endocrinol Metab. 2018;103:1929–1939.
  • Valimaki N, Schalin-Jantti C, Karppinen A, et al. Genetic and epigenetic characterization of growth hormone-secreting pituitary tumors. Mol Cancer Res. 2019;17:2432–2443.
  • Salomon MP, Wang X, Marzese DM, et al. The epigenomic landscape of pituitary adenomas reveals specific alterations and differentiates among acromegaly, cushing’s disease and endocrine-inactive subtypes. Clin Cancer Res. 2018;24:4126–4136.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.