Long-term safety of long-acting octreotide in patients with diabetic retinopathy: results of pooled data from 2 randomized, double-blind, placebo-controlled phase 3 studies

Rosario Pivonello, Giovanna Muscogiuri, Geoffrey Holder, Michaela Paul, Severine Sarp, Anastasia Lesogor, Pierre Jordaan, Johannes Eisinger, Annamaria Colao, Rosario Pivonello, Giovanna Muscogiuri, Geoffrey Holder, Michaela Paul, Severine Sarp, Anastasia Lesogor, Pierre Jordaan, Johannes Eisinger, Annamaria Colao

Abstract

Purpose: Octreotide (OCT) has been successfully used for treatment of acromegaly and neuroendocrine tumors for more than 30 years. However, long-term safety of OCT has not been documented in placebo-controlled setting. This present analysis pooled safety data from two similarly-designed, randomized, and placebo-controlled studies to evaluate long-term safety of long-acting OCT (20, 30 mg); targeted post-hoc analyzes focused on cardiac, hepatic, and renal safety.

Methods: Two studies (NCT00131144, NCT001308450) were conducted in patients with diabetic retinopathy (OCT20 = 191, OCT30 = 348, placebo = 347). In this analysis, patients were stratified based on baseline glomerular filtration rate. Hepatic, cardiac, and renal adverse events (AEs) were identified by standardized MedDRA queries.

Results: Median duration of exposure was >3.5 years. Most common AEs reported with OCT were diarrhea, cholelithiasis, hypoglycemia, nasopharyngitis, and hypertension. Incidence of cardiac events (QT prolongation and arrhythmia) with OCT20 and OCT30 were comparable to placebo (OCT20, RR = 1.11 [95% CI, 0.61-2.03]; OCT30, RR = 1.09 [95% CI, 0.70-1.68]). For ECG findings, changes in QTcF were similar in treatment groups, and outliers did not exceed 480 ms. Incidence of cardiac ischemia was lower with OCT than placebo (OCT20 = 12.6%, OCT30 = 10.6%, placebo = 15.3%). Incidence of liver-related AEs was higher with OCT30 than placebo (RR = 2.04 [95% CI, 1.28-3.26]); incidences were comparable with OCT20 and placebo (RR = 1.50 [95% CI, 0.69-3.25]). Overall incidences of renal AEs were comparable between treatment groups (OCT20 = 5.8%; OCT30 = 6.3%; placebo = 7.2%). Drug-related SAEs were reported more frequently with OCT (OCT20 = 7.9%; OCT30 = 10.1%; placebo = 3.5%); predominantly gallbladder-related, GI-related, and hypoglycemia.

Conclusions: The results from these long-term placebo-controlled studies confirm the established safety profile of long-acting OCT, in particular low risk of cardiac, hepatic and renal toxicity in a high-risk population.

Trial registration: ClinicalTrials.gov NCT00131144 NCT01308450.

Keywords: Cardiac function; Diabetic retinopathy; Hepatic function; Long-term safety; Octreotide; Renal function.

Conflict of interest statement

G.H., M.P., S.S., A.L., P.J., and J.E. are employees of Novartis. R.P. has been Principal investigator of research studies from Novartis, has received research grants from Novartis, Pfizer, Viropharma and IBSA, has been occasional consultant for Novartis, Ipsen, Pfizer, Viropharma, Ferring, and Italfarmaco, has received Fees and Honoraria for presentations from Novartis and Shire. A.C. is a member of the advisory board Novartis Europe and is recipient of unrestricted funds from Ipsen, Novartis, Pfizer, and Serono for research in neuroendocrinology. G.M. declares that she has no competing interests.

Figures

Fig. 1
Fig. 1
Forest plot of overall relative risk of incidence of hepatic, renal, and cardiac adverse events (sub-populations defined by renal function and all patients). Incidence calculated as number of subjects with any treatment-emergent AE divided by number of subjects at risk. Mantel-Haenszel estimate adjusted for study as stratification factor is used for RR and 95% confidence interval

References

    1. Battershill PE, Clissold SP. Octreotide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in conditions associated with excessive peptide secretion. Drugs. 1989;38:658–702. doi: 10.2165/00003495-198938050-00002.
    1. Grant MB, Mames RN, Fitzgerald C, Hazariwala KM, Cooper-DeHoff R, et al. The efficacy of octreotide in the therapy of severe nonproliferative and early proliferative diabetic retinopathy: a randomized controlled study. Diabetes Care. 2000;23:504–509. doi: 10.2337/diacare.23.4.504.
    1. Hyer SL, Sharp PS, Brooks RA, Burrin JM, Kohner EM. Continuous subcutaneous octreotide infusion markedly suppresses IGF-I levels whilst only partially suppressing GH secretion in diabetics with retinopathy. Acta Endocrinol. 1989;120:187–194.
    1. Marbach P, Bauer W, Bodmer D, Briner U, Bruns C, et al. Discovery and development of somatostatin agonists. Pharm Biotechnol. 1998;11:183–209. doi: 10.1007/0-306-47384-4_9.
    1. Patel YC. Somatostatin and its receptor family. Front. Neuroendocrinol. 1999;20:157–198. doi: 10.1006/frne.1999.0183.
    1. Bornschein J, Drozdov I, Malfertheiner P. Octreotide LAR: safety and tolerability issues. Expert Opin. Drug Saf. 2009;8:755–768. doi: 10.1517/14740330903379525.
    1. Giustina A, Karamouzis I, Patelli I, Mazziotti G. Octreotide for acromegaly treatment: a reappraisal. Expert Opin. Pharmacother. 2013;14:2433–2447. doi: 10.1517/14656566.2013.847090.
    1. Ludlam WH, Anthony L. Safety review: dose optimization of somatostatin analogs in patients with acromegaly and neuroendocrine tumors. Adv. Ther. 2011;28:825–841. doi: 10.1007/s12325-011-0062-9.
    1. Colao A, Auriemma RS, Galdiero M, Cappabianca P, Cavallo LM, et al. Impact of somatostatin analogs versus surgery on glucose metabolism in acromegaly: results of a 5-year observational, open, prospective study. J. Clin. Endocrinol. Metab. 2009;94:528–537. doi: 10.1210/jc.2008-1546.
    1. Mazziotti G, Porcelli T, Bogazzi F, Bugari G, Cannavo S, et al. Effects of high-dose octreotide LAR on glucose metabolism in patients with acromegaly inadequately controlled by conventional somatostatin analog therapy. Eur. J. Endocrinol. 2011;164:341–347. doi: 10.1530/EJE-10-0811.
    1. Mazziotti G, Floriani I, Bonadonna S, Torri V, Chanson P, Giustina A. Effects of somatostatin analogs on glucose homeostasis: a metaanalysis of acromegaly studies. J. Clin. Endocrinol. Metab. 2009;94:1500–1508. doi: 10.1210/jc.2008-2332.
    1. Fatti LM, Scacchi M, Lavezzi E, Pecori Giraldi F, De Martin M, et al. Effects of treatment with somatostatin analogues on QT interval duration in acromegalic patients. Clin. Endocrinol. 2006;65:626–630. doi: 10.1111/j.1365-2265.2006.02639.x.
    1. Colao A, Auriemma RS, Galdiero M, Lombardi G, Pivonello R. Effects of initial therapy for five years with somatostatin analogs for acromegaly on growth hormone and insulin-like growth factor-I levels, tumor shrinkage, and cardiovascular disease: a prospective study. J. Clin. Endocrinol. Metab. 2009;94:3746–3756. doi: 10.1210/jc.2009-0941.
    1. Warszawski L, Kasuki L, Sa R, Dos Santos Silva CM, Volschan I, et al. Low frequency of cardiac arrhythmias and lack of structural heart disease in medically-naive acromegaly patients: a prospective study at baseline and after 1 year of somatostatin analogs treatment. Pituitary. 2016;19:582–589. doi: 10.1007/s11102-016-0749-7.
    1. Stevens LA, Coresh J, Greene T, Levey AS. Assessing kidney function—measured and estimated glomerular filtration rate. N. Engl. J. Med. 2006;354:2473–2483. doi: 10.1056/NEJMra054415.
    1. Administration USFaD. Octreotide. . Accessed 30 Mar 2016
    1. Anthony L, Freda PU. From somatostatin to octreotide LAR: evolution of a somatostatin analog. Curr. Med. Res. Opin. 2009;25:2989–2999. doi: 10.1185/03007990903328959.
    1. Katznelson L, Atkinson JL, Cook DM, Ezzat SZ, Hamrahian AH, 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. doi: 10.4158/EP.17.S4.1.
    1. Pavel M, Kidd M, Modlin I. Systemic therapeutic options for carcinoid. Semin. Oncol. 2013;40:84–99. doi: 10.1053/j.seminoncol.2012.11.003.
    1. Astruc B, Marbach P, Bouterfa H, Denot C, Safari M, et al. Long-acting octreotide and prolonged-release lanreotide formulations have different pharmacokinetic profiles. J. Clin. Pharmacol. 2005;45:836–844. doi: 10.1177/0091270005277936.
    1. Barbare JC, Bouche O, Bonnetain F, Dahan L, Lombard-Bohas C, et al. Treatment of advanced hepatocellular carcinoma with long-acting octreotide: a phase III multicentre, randomised, double blind placebo-controlled study. Eur. J. Cancer. 2009;45:1788–1797. doi: 10.1016/j.ejca.2009.02.018.
    1. Bhatt HB, Smith RJ. Fatty liver disease in diabetes mellitus. Hepatobiliary Surg. Nutr. 2015;4:101–108.
    1. Hazlehurst JM, Woods C, Marjot T, Cobbold JF, Tomlinson JW. Non-alcoholic fatty liver disease and diabetes. Metabolism. 2016;65:1096–1108. doi: 10.1016/j.metabol.2016.01.001.
    1. Richard J, Lingvay I. Hepatic steatosis and type 2 diabetes: current and future treatment considerations. Expert Rev. Cardiovasc. Ther. 2011;9:321–328. doi: 10.1586/erc.11.15.
    1. Caroli A, Perico N, Perna A, Antiga L, Brambilla P, et al. Effect of long-acting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): a randomised, placebo-controlled, multicentre trial. Lancet. 2013;382:1485–1495. doi: 10.1016/S0140-6736(13)61407-5.
    1. Ottesen LH, Aagaard NK, Kiszka-Kanowitz M, Rehling M, Henriksen JH, et al. Effects of a long-acting formulation of octreotide on renal function and renal sodium handling in cirrhotic patients with portal hypertension: a randomized, double-blind, controlled trial. Hepatology. 2001;34:471–477. doi: 10.1053/jhep.2001.26754.
    1. Sun H, Zou S, Candiotti KA, Peng Y, Zhang Q, et al. Octreotide attenuates acute kidney injury after hepatic ischemia and reperfusion by enhancing autophagy. Sci. Rep. 2017;7:1–9. doi: 10.1038/s41598-016-0028-x.

Source: PubMed

3
Sottoscrivi