56
Views
0
CrossRef citations to date
0
Altmetric
Drug Evaluation

Pasireotide for treating acromegaly

&
Pages 531-540 | Received 15 Dec 2015, Accepted 15 Mar 2016, Published online: 12 Apr 2016

References

  • 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.
  • 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(12):4769–4775.
  • Melmed S:. Medical progress: acromegaly. N Engl J Med. 2006;355:2558–2573.
  • Jallad RS, Musolino NR, Salgado LR, et al. Treatment of acromegaly: is there still a place for radiotherapy? Pituitary. 2007;10:53–59.
  • Duarte FH, Jallad RS, Bronstein MD. Clomiphene citrate for treatment of acromegaly not controlled by conventional therapies. J Clin Endocrinol Metab. 2015;100(5):1863–1869.
  • Puig DM. Treatment of acromegaly in the era of personalized and predictive medicine. Clin Endocrinol (Oxf). 2015;83(1):3–14.
  • Mercado M, Borges F, Bouterfa H, et al., 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;66:859–868.
  • Colao A, Cappabianca P, Caron P, et al., et al. Octreotide LAR vs. surgery in newly diagnosed patients with acromegaly: A randomized, open-label, multicentre study. Clin Endocrinol (Oxf). 2009;70:757–768.
  • Gadelha MR, Bronstein MD, Brue T, et al. Pasireotide C2402 study group. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomized, phase 3 trial. Lancet Diab Endocrinol. 2014;2:875–884.
  • Patel YC, Murthy KK, Escher EE, et al. Mechanism of action of somatostatin: an overview of receptor function and studies of the molecular characterization and purification of somatostatin receptor proteins. Metabolism. 1990;39(9 Suppl 2):63–69.
  • Shimon I, Yan X, Taylor JE, et al. Somatostatin receptor (SSTR) subtype-selective analogues differentially suppress in vitro growth hormone and prolactin in human pituitary adenomas. Novel potential therapy for functional pituitary tumors. J Clin Invest. 1997;100:2386–2392.
  • Patel YC. Somatostatin and its receptor family. Front Neuroendocrinol. 1999;20(3):157–198.
  • Jaquet P, Saveanu A, Gunz G, et al. Human somatostatin receptor subtypes in acromegaly: distinct patterns of messenger ribonucleic acid expression and hormone suppression identify different tumoral phenotypes. J Clin Endocrinol Metab. 2000;85:781–792.
  • Hofland LJ, Lamberts SW, Hofland LJ, et al. Somatostatin receptor subtypes in human tumors. Ann Oncol. 2001;12(suppl 2):S31–S36.
  • Nielsen S, Mellemkjaer S, Rasmussen LM, et al. Expression of somatostatin receptors on human pituitary adenomas in vivo and ex vivo. J Endocrinol Invest. 2001;24:430–437.
  • Ben-Shlomo A, Melmed S. Pituitary somatostatin receptor signaling. Trends Endocrinol Metab. 2010 Mar;21(3):123–133.
  • Eigler T, Ben-Shlomo A. Somatostatin system: molecular mechanisms regulating anterior pituitary hormones. J Mol Endocrinol. 2014;53(1):R1–19.
  • Durán-Prado M, Gahete MD, Martínez-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 Jul;94(7):2634–2643.
  • 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(2):299–306.
  • Buscail L, Esteve JP, Saint-Laurent N, et al. Inhibition of cell proliferation by the somatostatin analogue RC-160 is mediated by somatostatin receptor subtypes SSTR2 and SSTR5 through different mechanisms. Proc Natl Acad Sci U S A. 1995;92(5):1580–1584.
  • Sharma K, Patel YC, Srikant CB. Subtype-selective induction of wild-type p53 and apoptosis, but not cell cycle arrest, by human somatostatin receptor 3. Mol Endocrinol. 1996;10(12):1688–1696.
  • Patel YC, Greenwood M, Panetta R, et al. Molecular biology of somatostatin receptor subtypes. Expression of mRNA for all five human somatostatin receptors (hSSTR1–5) in pituitary tumors. Metabolism. 1996;45(8 Suppl 1):31–38.
  • 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.
  • Casarini AP, Jallad RS, Pinto EM, et al. Acromegaly: correlation between expression of somatostatin receptor subtypes and response to octreotide-lar treatment. Pituitary. 2009;12(4):297–303.
  • Hornick CA, Anthony CT, Hughey S, et al. Progressive nuclear translocation of somatostatin analogs. J Nucl Med. 2000;41(7):1256–1263.
  • Reubi JC, Waser B, Schaer JC, et al. Somatostatin receptor sst1-sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands. Eur J Nucl Med. 2001;28(7):836–846.
  • Schulz S, Lehmann A, Kliewer A, et al. Fine-tuning somatostatin receptor signalling by agonist-selective phosphorylation and dephosphorylation: IUPHAR review 5. Br J Pharmacol. 2014;171(7):1591–1599.
  • Tulipano G, Stumm R, Pfeiffer M, et al. Differential beta-arrestin trafficking and endosomal sorting of somatostatin receptor subtypes. J Biol Chem. 2004;279(20):21374–21382.
  • Liu Q, Cescato R, Dewi DA, et al. Receptor signaling and endocytosis are differentially regulated by somatostatin analogs. Mol Pharmacol. 2005;68(1):90–101.
  • Cescato R, Schulz S, Waser B, et al. Internalization of sst2, sst3, and sst5 receptors: effects of somatostatin agonists and antagonists. J Nucl Med. 2006;47(3):502–511.
  • Stroh T, Jackson AC, Sarret P, et al. Intracellular dynamics of sst5 receptors in transfected COS-7 cells: maintenance of cell surface receptors during ligand-induced endocytosis. Endocrinology. 2000;141(1):354–365.
  • Tanaka T, Tani S. Inhibitory action of somatostatin on cAMP dependent pepsinogen secretion from rat gastric chief cells: involvement of pertussis toxin-sensitive G-protein. Biol Pharm Bull. 1994;17(3):415–418.
  • Reubi JC, Waser B, Cescato R, et al. Internalized somatostatin receptor subtype 2 in neuroendocrine tumors of octreotide-treated patients. J Clin Endocrinol Metab. 2010;95(5):2343–2350.
  • Ferjoux G, Bousquet C, Cordelier P, et al. Signal transduction of somatostatin receptors negatively controlling cell proliferation. J Physiol. 2000;94:205–210.
  • Hubina E, et al. Somatostatin analogues stimulate p27 expression and inhibit the MAP kinase pathway in pituitary tumors. Eur J Endocrinol. 2006;155:371–379.
  • Lahlou H, Guillermet J, Hortala M, et al. Molecular signaling of somatostatin receptors. Ann N Y Acad Sci. 2004;1014:121–131.
  • Bevan JS. Clinical review: the antitumoral effects of somatostatin analog therapy in acromegaly. J Clin Endocrinol Metab. 2005;90:1856–1863.
  • Kumar U. Cross-talk and modulation of signaling between somatostatin and growth factor receptors. Endocrine. 2011;40(2):168–180.
  • Susini C, Buscail L. Rationale for the use of somatostatin analogs as antitumor agents. Ann Oncol. 2006;17(12):1733–1742.
  • Pyronnet S, Bousquet C, Najib S, et al. Antitumor effects of somatostatin. Mol Cell Endocrinol. 2008;286(1–2):230–237.
  • Florio T, Barbieri F, Spaziante R, et al. Efficacy of a dopamine-somatostatin chimeric molecule, BIM-23A760, in the control of cell growth from primary cultures of human non-functioning pituitary adenomas: a multi-center study. Endocr Relat Cancer. 2008;15(2):583–596.
  • Oberg K. Future aspects of somatostatin-receptor-mediated therapy. Neuroendocrinology. 2004;80(Suppl 1):57–61.
  • Bronstein M, Musolino N, Jallad R, et al. Pharmacokinetic profile of lanreotide Autogel in patients with acromegaly after four deep subcutaneous injections of 60, 90 or 120 mg every 28 days. Clin Endocrinol (Oxf). 2005;63(5):514–519.
  • Neggers SJ, Pronin V, Balcere I, LEAD Study Group, et al.. Lanreotide Autogel 120 mg at extended dosing intervals in patients with acromegaly biochemically controlled with octreotide LAR: the LEAD study. Eur J Endocrinol. 2015;173(3):313–323.
  • Carmichael JD, Bonert VS, Nuño M, et al. 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.
  • Murray RD, Kim K, Ren S-G, et al. Central and peripheral actions of somatostatin on the growth hormone-IGF-I axis. J Clin Invest. 2004;114:349–356.
  • Chanson P, Timsit J, Harris AG. Clinical pharmacokinetics of octreotide. Therapeutic applications in patients with pituitary tumours. Clin Pharmacokinet. 1993;25:375–391.
  • Lancranjan I, Bruns C, Grass P, et al., et al. Sandostatin LAR®: pharmacokinetics, pharmacodynamics, efficacy, and tolerability in acromegalic patients. Metabolism. 1995;44(Suppl 1):18–26.
  • Stewart PM, Kane KF, Stewart SE, et al. Depot long-acting somatostatin analog (Sandostatin LAR) is an effective treatment for acromegaly. J Clin Endocrinol Metab. 1995;80:3267–3272.
  • Jenkins PJ, Akker S, Chew SL, et al. Optimal dosage interval for depot somatostatin analogue therapy in acromegaly requires individual titration. Clin Endocrinol. 2000;53:719–724.
  • Carmichael JD. Lanreotide depot deep subcutaneous injection: a new method of delivery and its associated benefits. Patient Prefer Adherence. 2012;6:73–82.
  • Melmed S, Cook D, Schopohl J, et al. 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;13:18–28.
  • Attanasio R, Baldelli R, Pivonello R, et al. Lanreotide 60 mg, a new long-acting formulation: effectiveness in the chronic treatment of acromegaly. J Clin Endocrinol Metab. 2003 Nov;88(11):5258–5265.
  • Lucas T, Astorga R, Catala M. Preoperative lanreotide treatment for GH-secreting pituitary adenomas: effect on tumour volume and predictive factors of significant tumour shrinkage. Clin Endocrinol. 2003;58(4):471–481.
  • Ayuk J, Stewart SE, Stewart PM, et al. Efficacy of SandostatinR LARR (long-acting somatostatin analogue) is similar in patients with untreated acromegaly and in those previously treated with surgery and/or radiotherapy. Clin Endocrinol. 2004;60(3):375–381.
  • Jallad RS, Musolino NR, Salgado LR, et al. Treatment of acromegaly with octreotide-LAR: extensive experience in a Brazilian institution. Clin Endocrinol (Oxf). 2005;63(2):168–175.
  • Cozzi R, Montini M, Attanasio R, et al. Primary treatment of acromegaly with octreotide LAR: a long-term (up to nine years) prospective study of its efficacy in the control of disease activity and tumor shrinkage. J Clin Endocrinol Metab. 2006;91(4):1397–1403.
  • 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 Apr 25;8(4):e61523.
  • Grasso LF, Auriemma RS, Pivonello R, et al. Adverse events associated with somatostatin analogs in acromegaly. Expert Opin Drug Saf. 2015;14(8):1213–1226.
  • Jallad RS, Bronstein MD. Optimizing medical therapy of acromegaly: beneficial effects of cabergoline in patients uncontrolled with long-acting release octreotide. Neuroendocrinology. 2009;90(1):82–92.
  • Suda K, Inoshita N, Iguchi G, et al. Efficacy of combined octreotide and cabergoline treatment in patients with acromegaly: a retrospective clinical study and review of the literature. Endocr J. 2013;60(4):507–515.
  • Bruns C, Lewis I, Briner U, et al. SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol. 2002;146(5):707–716.
  • Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.
  • Ma P, Wang Y, Van Der Hoek J, et al. Pharmacokinetic-pharmacodynamic comparison of a novel multiligand somatostatin analog, SOM230, with octreotide in patients with acromegaly. Clin Pharmacol Ther. 2005;78:69–80.
  • Hipkin RW, Friedman J, Clark RB, et al. Agonist induced desensitization, internalization and phosphorylation of the sst2A somatostatin receptor. J Biol Chem. 1997;272:13869–13876.
  • Pöll 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.
  • Waser B, Cescato R, Liu Q, et al. Phosphorylation of sst2 receptors in neuroendocrine tumors after octreotide treatment of patients. Am J Pathol. 2012;180(5):1942–1949.
  • Lewis I, Bauer W, Albert R, et al. A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential. J Med Chem. 2003;46(12):2334–2344.
  • Kao YJ, Ghosh M, Schonbrunn A. Ligand-dependent mechanisms of sst2A receptor trafficking: role of site-specific phosphorylation and receptor activation in the actions of biased somatostatin agonists. Mol Endocrinol. 2011;25(6):1040–1054.
  • Liu Q, Dewi DA, Liu W, et al. Distinct phosphorylation sites in the sst2A somatostatin receptor control internalization, desensitization, and arrestin binding. Mol Pharmacol. 2008;73(2):292–304.
  • Hofland LJ, Van Der Hoek J, Van Koetsveld PM, et al., 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.
  • FDA Website. How to apply for orphan product designation [Internet]. 2014. Available from: http://www.fda.gov/ForIndustry/DevelopingProductsforRareDiseasesConditions/HowtoapplyforOrphanProductDesignation/ucm364750.htm
  • FDA Website. Pasireotide orphan designation [Internet]. 2014. Available from: http://www.accessdata.fda.gov/scripts/opdlisting/oopd/OOPD_Results_2.cfm?Index_Number=288609
  • Van Der Hoek J, De Herder WW, Feelders RA, et al., et al A single-dose comparison of the acute effects between the new somatostatin analog SOM230 and octreotide in acromegalic patients. J Clin Endocrinol Metab. 2004;89(2):638–645.
  • Petersenn S, Schopohl J, Barkan A, et al., et al Pasireotide (SOM230) demonstrates efficacy and safety in patients with acromegaly: a randomized, multicenter, phase II trial. J Clin Endocrinol Metab. 2010;95(6):2781–2789.
  • Petersenn S, Farrall AJ, De Block C, et al. Long-term efficacy and safety of subcutaneous pasireotide in acromegaly: results from an open-ended, multicenter, phase II extension study. Pituitary. 2014;17(2):132–140.
  • Petersenn S, Bollerslev J, Arafat AM, et al. Pharmacokinetics, pharmacodynamics, and safety of pasireotide LAR in patients with acromegaly: a randomized, multicenter, open-label, phase I study. J Clin Pharmacol. 2014;54(11):1308–1317.
  • Colao A, Bronstein MD, Freda P, Pasireotide C2305 Study Group, et al. Pasireotide versus octreotide in acromegaly: a head-to-head superiority study. J Clin Endocrinol Metab. 2014;99:791–799.
  • Sheppard M, Bronstein MD, Freda P, et al. Erratum to: pasireotide LAR maintains inhibition of GH and IGF-1 in patients with acromegaly for up to 25 months: results from the blinded extension phase of a randomized, double-blind, multicenter, phase III study. Pituitary. 2015;18(3):395–396.
  • 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(2):241–250.
  • Zambre Y, Ling Z, Chen MC. Inhibition of human pancreatic islet insulin release by receptor-selective somatostatin analogs directed to somatostatin receptor subtype 5. Biochem Pharmacol. 1999;57:1159–1164.
  • Singh V, Brendel MD, Zacharias S. Characterization of somatostatin receptor subtype-specific regulation of insulin and glucagon secretion: an in vitro study on isolated human pancreatic islets. J Clin Endocrinol Metab. 2007;92:673–680.
  • Braun M. The somatostatin receptor in human pancreatic β-cells. Vitam Horm. 2014;95:165–193.
  • Henry RR, Ciaraldi TP, Armstrong D, et al. Hyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteers. J Clin Endocrinol Metab. 2013;98(8):3446–3453.
  • American Diabetes Association. Approaches to glycemic treatment. Sec. 7. In standards of medical care in diabetes—2015. Diab Care. 2015;38(Suppl. 1):S41–S48.
  • Murray B, Gordon MD, Spiller KL. MS management of hyperclycemia in a patient with acromegaly treated with pasireotide LAR: a case study AACE Clinical Case Reports. Rapid Electronic Articles in Press; 2015.
  • Breitschaft A, Hu K, Darstein C, et al. Effects of subcutaneous pasireotide on cardiac repolarization in healthy volunteers: a single‐center, phase i, randomized, four‐way crossover study. J Clin Pharmacol. 2014;54(1):75–86.

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.