167
Views
13
CrossRef citations to date
0
Altmetric
Review

Octreotide-Resistant Acromegaly: Challenges and Solutions

, , , , , , , , ORCID Icon & ORCID Icon show all
Pages 379-391 | Published online: 05 May 2020

References

  • Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest. 2009;119(11):3189–3202. doi:10.1172/JCI39375
  • Pita-Gutierrez F, Pertega-Diaz S, Pita-Fernandez S, et al. Place of preoperative treatment of acromegaly with somatostatin analog on surgical outcome: a systematic review and meta-analysis. PLoS One. 2013;8(4):e61523. doi:10.1371/journal.pone.0061523
  • Holdaway IM, Rajasoorya C. Epidemiology of acromegaly. Pituitary. 1999;2(1):29–41. doi:10.1023/A:1009965803750
  • Melmed S. Medical progress: acromegaly. N Engl J Med. 2006;355(24):2558–2573. doi:10.1056/NEJMra062453
  • Christofides EA. Clinical importance of achieving biochemical control with medical therapy in adult patients with acromegaly. Patient Prefer Adherence. 2016;10:1217–1225. doi:10.2147/PPA.S102302
  • Gatto F, Trifiro G, Lapi F, et al. Epidemiology of acromegaly in Italy: analysis from a large longitudinal primary care database. Endocrine. 2018;61(3):533–541. doi:10.1007/s12020-018-1630-4
  • Ben-Shlomo A, Sheppard MC, Stephens JM, Pulgar S, Melmed S. Clinical, quality of life, and economic value of acromegaly disease control. Pituitary. 2011;14(3):284–294. doi:10.1007/s11102-011-0310-7
  • Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf). 2010;72(3):377–382.
  • Abreu A, Tovar AP, Castellanos R, et al. Challenges in the diagnosis and management of acromegaly: a focus on comorbidities. Pituitary. 2016;19(4):448–457.
  • Holdaway IM, Bolland MJ, Gamble GD. A meta-analysis of the effect of lowering serum levels of GH and IGF-I on mortality in acromegaly. Eur J Endocrinol. 2008;159(2):89–95. doi:10.1530/EJE-08-0267
  • Reid TJ, Post KD, Bruce JN, Nabi Kanibir M, Reyes-Vidal CM, Freda PU. Features at diagnosis of 324 patients with acromegaly did not change from 1981 to 2006: acromegaly remains under-recognized and under-diagnosed. Clin Endocrinol (Oxf). 2010;72(2):203–208. doi:10.1111/cen.2010.72.issue-2
  • Katznelson L, Laws ER Jr., Melmed S, et al. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99(11):3933–3951. doi:10.1210/jc.2014-2700
  • Colao A, Auriemma RS, Lombardi G, Pivonello R. Resistance to somatostatin analogs in acromegaly. Endocr Rev. 2011;32(2):247–271. doi:10.1210/er.2010-0002
  • Melmed S, Bronstein MD, Chanson P, et al. A consensus statement on acromegaly therapeutic outcomes. Nat Rev Endocrinol. 2018;14(9):552–561. doi:10.1038/s41574-018-0058-5
  • McKeage K. Pasireotide in acromegaly: a review. Drugs. 2015;75(9):1039–1048. doi:10.1007/s40265-015-0413-y
  • Bacigaluppi S, Gatto F, Anania P, et al. Impact of pre-treatment with somatostatin analogs on surgical management of acromegalic patients referred to a single center. Endocrine. 2016;51(3):524–533. doi:10.1007/s12020-015-0619-5
  • Abu Dabrh AM, Asi N, Farah WH, et al. Radiotherapy versus radiosurgery in treating patients with acromegaly: a systematic review and meta-analysis. Endocr Pract. 2015;21(8):943–956. doi:10.4158/EP14574.OR
  • Giustina A, Chanson P, Kleinberg D, et al. Expert consensus document: a consensus on the medical treatment of acromegaly. Nat Rev Endocrinol. 2014;10(4):243–248. doi:10.1038/nrendo.2014.21
  • Murray RD, Melmed S. A critical analysis of clinically available somatostatin analog formulations for therapy of acromegaly. J Clin Endocrinol Metab. 2008;93(8):2957–2968. doi:10.1210/jc.2008-0027
  • Carmichael JD, Bonert VS, Nuno M, Ly D, Melmed S. Acromegaly clinical trial methodology impact on reported biochemical efficacy rates of somatostatin receptor ligand treatments: a meta-analysis. J Clin Endocrinol Metab. 2014;99(5):1825–1833. doi:10.1210/jc.2013-3757
  • Babu H, Ortega A, Nuno M, et al. Long-term endocrine outcomes following endoscopic endonasal transsphenoidal surgery for acromegaly and associated prognostic factors. Neurosurgery. 2017;81(2):357–366. doi:10.1093/neuros/nyx020
  • Giustina A, Arnaldi G, Bogazzi F, et al. Pegvisomant in acromegaly: an update. J Endocrinol Invest. 2017;40(6):577–589. doi:10.1007/s40618-017-0614-1
  • Pokrajac A, Wark G, Ellis AR, Wear J, Wieringa GE, Trainer PJ. Variation in GH and IGF-I assays limits the applicability of international consensus criteria to local practice. Clin Endocrinol (Oxf). 2007;67(1):65–70. doi:10.1111/cen.2007.67.issue-1
  • Arafat AM, Mohlig M, Weickert MO, et al. Growth hormone response during oral glucose tolerance test: the impact of assay method on the estimation of reference values in patients with acromegaly and in healthy controls, and the role of gender, age, and body mass index. J Clin Endocrinol Metab. 2008;93(4):1254–1262. doi:10.1210/jc.2007-2084
  • Clemmons DR. IGF-I assays: current assay methodologies and their limitations. Pituitary. 2007;10(2):121–128. doi:10.1007/s11102-007-0032-z
  • Clemmons DR. Consensus statement on the standardization and evaluation of growth hormone and insulin-like growth factor assays. Clin Chem. 2011;57(4):555–559. doi:10.1373/clinchem.2010.150631
  • Schilbach K, Strasburger CJ, Bidlingmaier M. Biochemical investigations in diagnosis and follow up of acromegaly. Pituitary. 2017;20(1):33–45. doi:10.1007/s11102-017-0792-z
  • Paragliola RM, Corsello SM, Salvatori R. Somatostatin receptor ligands in acromegaly: clinical response and factors predicting resistance. Pituitary. 2017;20(1):109–115. doi:10.1007/s11102-016-0768-4
  • Fleseriu M. Clinical efficacy and safety results for dose escalation of somatostatin receptor ligands in patients with acromegaly: a literature review. Pituitary. 2011;14(2):184–193. doi:10.1007/s11102-010-0282-z
  • Freda PU, Katznelson L, van der Lely AJ, Reyes CM, Zhao S, Rabinowitz D. Long-acting somatostatin analog therapy of acromegaly: a meta-analysis. J Clin Endocrinol Metab. 2005;90(8):4465–4473. doi:10.1210/jc.2005-0260
  • Roelfsema F, Biermasz NR, Pereira AM, Romijn JA. Therapeutic options in the management of acromegaly: focus on lanreotide Autogel. Biol Targets Ther. 2008;2(3):463–479.
  • Chanson P, Borson-Chazot F, Kuhn JM, et al. Control of IGF-I levels with titrated dosing of lanreotide Autogel over 48 weeks in patients with acromegaly. Clin Endocrinol (Oxf). 2008;69(2):299–305. doi:10.1111/j.1365-2265.2008.03208.x
  • Tutuncu Y, Berker D, Isik S, et al. Comparison of octreotide LAR and lanreotide autogel as post-operative medical treatment in acromegaly. Pituitary. 2012;15(3):398–404. doi:10.1007/s11102-011-0335-y
  • Mazziotti G, Giustina A. Effects of lanreotide SR and Autogel on tumor mass in patients with acromegaly: a systematic review. Pituitary. 2010;13(1):60–67. doi:10.1007/s11102-009-0169-z
  • Giustina A, Mazziotti G, Torri V, Spinello M, Floriani I, Melmed S. Meta-analysis on the effects of octreotide on tumor mass in acromegaly. PLoS One. 2012;7(5):e36411. doi:10.1371/journal.pone.0036411
  • Cuevas-Ramos D, Fleseriu M. Somatostatin receptor ligands and resistance to treatment in pituitary adenomas. J Mol Endocrinol. 2014;52(3):R223–R240. doi:10.1530/JME-14-0011
  • Mercado M, Borges F, Bouterfa H, et al. A prospective, multicentre study to investigate the efficacy, safety and tolerability of octreotide LAR (long-acting repeatable octreotide) in the primary therapy of patients with acromegaly. Clin Endocrinol (Oxf). 2007;66(6):859–868. doi:10.1111/cen.2007.66.issue-6
  • Melmed S, Cook D, Schopohl J, Goth MI, Lam KS, Marek J. Rapid and sustained reduction of serum growth hormone and insulin-like growth factor-1 in patients with acromegaly receiving lanreotide Autogel therapy: a randomized, placebo-controlled, multicenter study with a 52 week open extension. Pituitary. 2010;13(1):18–28. doi:10.1007/s11102-009-0191-1
  • Shimatsu A, Teramoto A, Hizuka N, Kitai K, Ramis J, Chihara K. Efficacy, safety, and pharmacokinetics of sustained-release lanreotide (lanreotide Autogel) in Japanese patients with acromegaly or pituitary gigantism. Endocr J. 2013;60(5):651–663. doi:10.1507/endocrj.EJ12-0417
  • Colao A, Auriemma RS, Pivonello R, Kasuki L, Gadelha MR. Interpreting biochemical control response rates with first-generation somatostatin analogues in acromegaly. Pituitary. 2016;19(3):235–247. doi:10.1007/s11102-015-0684-z
  • Gadelha MR, Wildemberg LE, Bronstein MD, Gatto F, Ferone D. Somatostatin receptor ligands in the treatment of acromegaly. Pituitary. 2017;20(1):100–108. doi:10.1007/s11102-017-0791-0
  • Gatto F, Campana C, Cocchiara F, et al. Current perspectives on the impact of clinical disease and biochemical control on comorbidities and quality of life in acromegaly. Rev Endocr Metab Disord. 2019;20:365–381. doi:10.1007/s11154-019-09506-y
  • Besser GM, Burman P, Daly AF. Predictors and rates of treatment-resistant tumor growth in acromegaly. Eur J Endocrinol. 2005;153(2):187–193. doi:10.1530/eje.1.01968
  • Casarini AP, Pinto EM, Jallad RS, Giorgi RR, Giannella-Neto D, Bronstein MD. Dissociation between tumor shrinkage and hormonal response during somatostatin analog treatment in an acromegalic patient: preferential expression of somatostatin receptor subtype 3. J Endocrinol Invest. 2006;29(9):826–830. doi:10.1007/BF03347378
  • 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(5):651–662. doi:10.1530/EJE-15-0391
  • Gadelha MR, Kasuki L, Korbonits M. Novel pathway for somatostatin analogs in patients with acromegaly. Trends Endocrinol Metab. 2013;24(5):238–246. doi:10.1016/j.tem.2012.11.007
  • 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(1):E66–E71. doi:10.1210/jc.2012-2609
  • Wildemberg LE, Neto LV, Costa DF, et al. Low somatostatin receptor subtype 2, but not dopamine receptor subtype 2 expression predicts the lack of biochemical response of somatotropinomas to treatment with somatostatin analogs. J Endocrinol Invest. 2013;36(1):38–43. doi:10.3275/8305
  • Ferone D, de Herder WW, Pivonello R, et al. Correlation of in vitro and in vivo somatotropic adenoma responsiveness to somatostatin analogs and dopamine agonists with immunohistochemical evaluation of somatostatin and dopamine receptors and electron microscopy. J Clin Endocrinol Metab. 2008;93(4):1412–1417. doi:10.1210/jc.2007-1358
  • 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(2):82–91. doi:10.1007/s12022-008-9029-z
  • Mayr B, Buslei R, Theodoropoulou M, Stalla GK, Buchfelder M, Schofl C. Molecular and functional properties of densely and sparsely granulated GH-producing pituitary adenomas. Eur J Endocrinol. 2013;169(4):391–400. doi:10.1530/EJE-13-0134
  • 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(3):221–228. doi:10.1507/endocrj.EJ11-0177
  • Brzana J, Yedinak CG, Gultekin SH, Delashaw JB, Fleseriu M. 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(4):490–498. doi:10.1007/s11102-012-0445-1
  • Fougner SL, Casar-Borota O, Heck A, Berg JP, Bollerslev J. Adenoma granulation pattern correlates with clinical variables and effect of somatostatin analogue treatment in a large series of patients with acromegaly. Clin Endocrinol (Oxf). 2012;76(1):96–102. doi:10.1111/cen.2011.76.issue-1
  • 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(1):72–78. doi:10.1111/j.1365-2265.2011.04286.x
  • Potorac I, Petrossians P, Daly AF, et al. T2-weighted MRI signal predicts hormone and tumor responses to somatostatin analogs in acromegaly. Endocr Relat Cancer. 2016;23(11):871–881. doi:10.1530/ERC-16-0356
  • Grant M, Alturaihi H, Jaquet P, Collier B, Kumar U. Cell growth inhibition and functioning of human somatostatin receptor type 2 are modulated by receptor heterodimerization. Mol Endocrinol. 2008;22(10):2278–2292. doi:10.1210/me.2007-0334
  • 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(3):295–303. doi:10.1530/EJE-07-0562
  • Duran-Prado M, Gahete MD, Martinez-Fuentes AJ, et al. Identification and characterization of two novel truncated but functional isoforms of the somatostatin receptor subtype 5 differentially present in pituitary tumors. J Clin Endocrinol Metab. 2009;94(7):2634–2643. doi:10.1210/jc.2008-2564
  • 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(5):2497–2502. doi:10.1210/jc.2009-2247
  • Gatto F, Hofland LJ. The role of somatostatin and dopamine D2 receptors in endocrine tumors. Endocr Relat Cancer. 2011;18(6):R233–R251. doi:10.1530/ERC-10-0334
  • Treppiedi D, Mangili F, Giardino E, et al. Cytoskeleton protein Filamin A is required for efficient somatostatin receptor type 2 internalization and recycling through Rab5 and Rab4 sorting endosomes in tumor somatotroph cells. Neuroendocrinology. 2019. doi:10.1159/000503791
  • Treppiedi D, Jobin ML, Peverelli E, et al. Single-molecule microscopy reveals dynamic FLNA interactions governing SSTR2 clustering and internalization. Endocrinology. 2018;159(8):2953–2965. doi:10.1210/en.2018-00368
  • 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(12):4715–4725. doi:10.1210/en.2013-1672
  • 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(8):2932–2941. doi:10.1210/en.2014-1063
  • Coelho MCA, Vasquez ML, Wildemberg LE, et al. Clinical significance of filamin A in patients with acromegaly and its association with somatostatin and dopamine receptor profiles. Sci Rep. 2019;9(1):1122. doi:10.1038/s41598-018-37692-3
  • 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(4):2110–2116. doi:10.1111/jcmm.13427
  • Venegas-Moreno E, Flores-Martinez A, Dios E, et al. E-cadherin expression is associated with somatostatin analogue response in acromegaly. J Cell Mol Med. 2019;23(5):3088–3096. doi:10.1111/jcmm.13851
  • Fougner SL, Lekva T, Borota OC, Hald JK, Bollerslev J, Berg JP. 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(5):2334–2342. doi:10.1210/jc.2009-2197
  • Peverelli E, Mantovani G, Lania AG, Spada A. cAMP in the pituitary: an old messenger for multiple signals. J Mol Endocrinol. 2014;52(1):R67–R77. doi:10.1530/JME-13-0172
  • Barlier A, Gunz G, Zamora AJ, et al. Pronostic and therapeutic consequences of Gs alpha mutations in somatotroph adenomas. J Clin Endocrinol Metab. 1998;83(5):1604–1610. doi:10.1210/jcem.83.5.4797
  • 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(8):E1411–E1420. doi:10.1210/jc.2012-1111
  • 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(7):419–427. doi:10.1016/j.tem.2010.02.007
  • Cazabat L, Bouligand J, Salenave S, 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(4):E663–E670. doi:10.1210/jc.2011-2291
  • Kasuki L, Vieira Neto L, Wildemberg LE, 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(3):L25–L29.
  • Astruc B, Marbach P, Bouterfa H, et al. Long-acting octreotide and prolonged-release lanreotide formulations have different pharmacokinetic profiles. J Clin Pharmacol. 2005;45(7):836–844.
  • McKeage K, Cheer S, Wagstaff AJ. Octreotide long-acting release (LAR): a review of its use in the management of acromegaly. Drugs. 2003;63(22):2473–2499.
  • Lancranjan I, Bruns C, Grass P, et al. Sandostatin LAR: a promising therapeutic tool in the management of acromegalic patients. Metabolism. 1996;45(8 Suppl 1):67–71.
  • 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(16):3940.
  • Colao A, Pivonello R, Auriemma RS, Galdiero M, Savastano S, Lombardi G. Beneficial effect of dose escalation of octreotide-LAR as first-line therapy in patients with acromegaly. Eur J Endocrinol. 2007;157(5):579–587.
  • Giustina A, Bonadonna S, Bugari G, et al. High-dose intramuscular octreotide in patients with acromegaly inadequately controlled on conventional somatostatin analogue therapy: a randomised controlled trial. Eur J Endocrinol. 2009;161(2):331–338.
  • Giustina A, Mazziotti G, Cannavo S, et al. High-dose and high-frequency lanreotide autogel in acromegaly: a randomized, multicenter study. J Clin Endocrinol Metab. 2017;102(7):2454–2464.
  • Auriemma RS, Pivonello R, Ferreri L, Priscitelli P, Colao A. Cabergoline use for pituitary tumors and valvular disorders. Endocrinol Metab Clin North Am. 2015;44(1):89–97.
  • Sandret L, Maison P, Chanson P. Place of cabergoline in acromegaly: a meta-analysis. J Clin Endocrinol Metab. 2011;96(5):1327–1335.
  • Kuhn E, Chanson P. Cabergoline in acromegaly. Pituitary. 2017;20(1):121–128.
  • Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.
  • Poll F, Lehmann D, Illing S, et al. Pasireotide and octreotide stimulate distinct patterns of sst2A somatostatin receptor phosphorylation. Mol Endocrinol. 2010;24(2):436–446.
  • Colao A, Bronstein MD, Freda P, et al. Pasireotide versus octreotide in acromegaly: a head-to-head superiority study. J Clin Endocrinol Metab. 2014;99(3):791–799.
  • 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(6):2009–2018.
  • 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(4):1577–1585.
  • Ibanez-Costa A, Rivero-Cortes E, Vazquez-Borrego MC, et al. Octreotide and pasireotide (dis)similarly inhibit pituitary tumor cells in vitro. J Endocrinol. 2016;231(2):135–145.
  • Gadelha MR, Bronstein MD, Brue T, et al. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, Phase 3 trial. Lancet Diabetes Endocrinol. 2014;2(11):875–884.
  • Shimon I, Adnan Z, Gorshtein A, et al. Efficacy and safety of long-acting pasireotide in patients with somatostatin-resistant acromegaly: a multicenter study. Endocrine. 2018;62(2):448–455.
  • Schmid HA, Brue T, Colao A, et al. Effect of pasireotide on glucose- and growth hormone-related biomarkers in patients with inadequately controlled acromegaly. Endocrine. 2016;53(1):210–219.
  • Coopmans EC, Muhammad A, van der Lely AJ, Janssen J, Neggers S. How to position pasireotide LAR treatment in acromegaly. J Clin Endocrinol Metab. 2019;104(6):1978–1988.
  • Grottoli S, Maffei P, Bogazzi F, et al. ACROSTUDY: the Italian experience. Endocrine. 2015;48(1):334–341.
  • Freda PU, Gordon MB, Kelepouris N, Jonsson P, Koltowska-Haggstrom M, van der Lely AJ. Long-term treatment with pegvisomant as monotherapy in patients with acromegaly: experience from ACROSTUDY. Endocr Pract. 2015;21(3):264–274.
  • van der Lely AJ, Biller BM, Brue T, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab. 2012;97(5):1589–1597.
  • Buchfelder M, van der Lely AJ, Biller BMK, et al. Long-term treatment with pegvisomant: observations from 2090 acromegaly patients in ACROSTUDY. Eur J Endocrinol. 2018;179(6):419–427.
  • Ragonese M, Grottoli S, Maffei P, et al. How to improve effectiveness of pegvisomant treatment in acromegalic patients. J Endocrinol Invest. 2018;41(5):575–581. doi:10.1007/s40618-017-0773-0
  • Sievers C, Baur DM, Schwanke A, et al. Prediction of therapy response in acromegalic patients under pegvisomant therapy within the German ACROSTUDY cohort. Pituitary. 2015;18(6):916–923. doi:10.1007/s11102-015-0673-2
  • Franck SE, Korevaar TIM, Petrossians P, et al. A multivariable prediction model for pegvisomant dosing: monotherapy and in combination with long-acting somatostatin analogues. Eur J Endocrinol. 2017;176(4):421–431. doi:10.1530/EJE-16-0956
  • Feola T, Cozzolino A, Simonelli I, et al. Pegvisomant improves glucose metabolism in acromegaly: a meta-analysis of prospective interventional studies. J Clin Endocrinol Metab. 2019;104(7):2892–2902. doi:10.1210/jc.2018-02281
  • Neggers SJ, Franck SE, de Rooij FW, et al. Long-term efficacy and safety of pegvisomant in combination with long-acting somatostatin analogs in acromegaly. J Clin Endocrinol Metab. 2014;99(10):3644–3652. doi:10.1210/jc.2014-2032
  • Neggers SJ, de Herder WW, Feelders RA, van der Lely AJ. Conversion of daily pegvisomant to weekly pegvisomant combined with long-acting somatostatin analogs, in controlled acromegaly patients. Pituitary. 2011;14(3):253–258.
  • Madsen M, Poulsen PL, Orskov H, Moller N, Jorgensen JO. Cotreatment with pegvisomant and a somatostatin analog (SA) in SA-responsive acromegalic patients. J Clin Endocrinol Metab. 2011;96(8):2405–2413. doi:10.1210/jc.2011-0654
  • van der Lely AJ, Bernabeu I, Cap J, et al. Coadministration of lanreotide Autogel and pegvisomant normalizes IGF1 levels and is well tolerated in patients with acromegaly partially controlled by somatostatin analogs alone. Eur J Endocrinol. 2011;164(3):325–333. doi:10.1530/EJE-10-0867
  • Trainer PJ, Ezzat S, D’Souza GA, Layton G, Strasburger CJ. A randomized, controlled, multicentre trial comparing pegvisomant alone with combination therapy of pegvisomant and long-acting octreotide in patients with acromegaly. Clin Endocrinol (Oxf). 2009;71(4):549–557. doi:10.1111/cen.2009.71.issue-4
  • Neggers SJ, van Aken MO, Janssen JA, Feelders RA, de Herder WW, van der Lely AJ. Long-term efficacy and safety of combined treatment of somatostatin analogs and pegvisomant in acromegaly. J Clin Endocrinol Metab. 2007;92(12):4598–4601. doi:10.1210/jc.2007-1234
  • Bianchi A, Valentini F, Iuorio R, et al. Long-term treatment of somatostatin analog-refractory growth hormone-secreting pituitary tumors with pegvisomant alone or combined with long-acting somatostatin analogs: a retrospective analysis of clinical practice and outcomes. J Exp Clin Cancer Res. 2013;32:40. doi:10.1186/1756-9966-32-40
  • Higham CE, Atkinson AB, Aylwin S, et al. Effective combination treatment with cabergoline and low-dose pegvisomant in active acromegaly: a prospective clinical trial. J Clin Endocrinol Metab. 2012;97(4):1187–1193. doi:10.1210/jc.2011-2603
  • Bernabeu I, Alvarez-Escola C, Paniagua AE, et al. Pegvisomant and cabergoline combination therapy in acromegaly. Pituitary. 2013;16(1):101–108. doi:10.1007/s11102-012-0382-z
  • Muhammad A, van der Lely AJ, Delhanty PJD, et al. Efficacy and safety of switching to pasireotide in patients with acromegaly controlled with pegvisomant and first-generation somatostatin analogues (PAPE Study). J Clin Endocrinol Metab. 2018;103(2):586–595. doi:10.1210/jc.2017-02017
  • Muhammad A, Coopmans EC, Delhanty PJD, et al. Efficacy and safety of switching to pasireotide in acromegaly patients controlled with pegvisomant and somatostatin analogues: PAPE extension study. Eur J Endocrinol. 2018;179(5):269–277. doi:10.1530/EJE-18-0353
  • Yamada S, Fukuhara N, Oyama K, Takeshita A, Takeuchi Y. Repeat transsphenoidal surgery for the treatment of remaining or recurring pituitary tumors in acromegaly. Neurosurgery. 2010;67(4):949–956. doi:10.1227/NEU.0b013e3181ec4379
  • Mathioudakis N, Salvatori R. Management options for persistent postoperative acromegaly. Neurosurg Clin N Am. 2012;23(4):621–638. doi:10.1016/j.nec.2012.06.005
  • Abe T, Ludecke DK. Recent results of secondary transnasal surgery for residual or recurring acromegaly. Neurosurgery. 1998;42(5):1013–1021; discussion 1021–1012. doi:10.1097/00006123-199805000-00036
  • Ludecke DK. Value of transcavernous surgery in the treatment of pituitary adenomas. Eur J Endocrinol. 1995;133(2):147–148. doi:10.1530/eje.0.1330147
  • Nomikos P, Buchfelder M, Fahlbusch R. The outcome of surgery in 668 patients with acromegaly using current criteria of biochemical ‘cure’. Eur J Endocrinol. 2005;152(3):379–387. doi:10.1530/eje.1.01863
  • Kurosaki M, Luedecke DK, Abe T. Effectiveness of secondary transnasal surgery in GH-secreting pituitary macroadenomas. Endocr J. 2003;50(5):635–642. doi:10.1507/endocrj.50.635
  • Wilson TJ, McKean EL, Barkan AL, Chandler WF, Sullivan SE. Repeat endoscopic transsphenoidal surgery for acromegaly: remission and complications. Pituitary. 2013;16(4):459–464. doi:10.1007/s11102-012-0457-x
  • Almeida JP, Ruiz-Trevino AS, Liang B, et al. Reoperation for growth hormone-secreting pituitary adenomas: report on an endonasal endoscopic series with a systematic review and meta-analysis of the literature. J Neurosurg. 2018;129(2):404–416. doi:10.3171/2017.2.JNS162673
  • Long H, Beauregard H, Somma M, Comtois R, Serri O, Hardy J. Surgical outcome after repeated transsphenoidal surgery in acromegaly. J Neurosurg. 1996;85(2):239–247. doi:10.3171/jns.1996.85.2.0239
  • Del Porto LA, Liubinas SV, Kaye AH. Treatment of persistent and recurrent acromegaly. J Clin Neurosci. 2011;18(2):181–190. doi:10.1016/j.jocn.2010.10.003
  • Shih HA, Loeffler JS. Radiation therapy in acromegaly. Rev Endocr Metab Disord. 2008;9(1):59–65. doi:10.1007/s11154-007-9065-x
  • Kim EH, Oh MC, Chang JH, et al. Postoperative gamma knife radiosurgery for cavernous sinus-invading growth hormone-secreting pituitary adenomas. World Neurosurg. 2018;110:e534–e545. doi:10.1016/j.wneu.2017.11.043
  • Ding D, GU M, MR P, et al. Stereotactic radiosurgery for acromegaly: an international multicenter retrospective cohort study. Neurosurgery. 2019;84(3):717–725. doi:10.1093/neuros/nyy178