Safety and Efficacy of Subcutaneous Pasireotide in Patients With Cushing's Disease: Results From an Open-Label, Multicenter, Single-Arm, Multinational, Expanded-Access Study

Maria Fleseriu, Chioma Iweha, Luiz Salgado, Tania Longo Mazzuco, Federico Campigotto, Ricardo Maamari, Padiporn Limumpornpetch, Maria Fleseriu, Chioma Iweha, Luiz Salgado, Tania Longo Mazzuco, Federico Campigotto, Ricardo Maamari, Padiporn Limumpornpetch

Abstract

Introduction: The efficacy and safety of subcutaneous (sc) pasireotide have been evaluated in a Phase III trial. Here, we report safety and efficacy results from a multinational, expanded-access study of pasireotide sc in patients with Cushing's disease (CD) in a real-world setting (clinicaltrials.gov, identifier: NCT01582061). Methods: Adults with active CD previously untreated with pasireotide were enrolled; pasireotide sc was initiated at 600 μg twice daily (bid; EU countries) or 900 μg bid (non-EU countries; 600 μg bid in patients with impaired glucose metabolism). Pasireotide dose could be adjusted in 300 μg increments/decrements to a maximum of 900 μg bid or minimum of 300 μg bid for sustained urinary free cortisol (UFC) normalization/tolerability issues. Primary objective: document the safety of pasireotide sc in patients with CD. Key secondary objectives: assess the proportion of patients with mean UFC (mUFC) not exceeding the upper limit of normal (ULN) and changes from baseline in clinical signs/symptoms and quality of life (QoL) to weeks 12, 24, and 48. Results: One hundred and four patients received pasireotide: female, n = 84 (80.8%); median duration of pasireotide exposure, 25.1 weeks; median (range) baseline mUFC, 321.2 nmol/24 h (142-10,920; 2.3 × ULN [1.0-79.2]). Forty (38.5%) patients completed the study. The most common reasons for premature discontinuation of pasireotide were unsatisfactory therapeutic effect (n = 26, 25.0%) and adverse events (AEs; n = 20, 19.2%). Drug-related grade 3/4 AEs or drug-related serious AEs (primary endpoint) were documented in 42 (40.4%) patients, most commonly diabetes mellitus (n = 12, 11.5%) and hyperglycemia (n = 8, 7.7%). All patients experienced ≥1 AE and most (n = 102; 98.1%) reported ≥1 drug-related AE; six (5.8%) patients discontinued treatment because of hyperglycemia-related AEs. At weeks 12, 24, and 48, respectively, 36/66 (54.5%), 22/46 (47.8%), and 9/21 (42.9%) evaluable patients had normalized mUFC levels. Clinical signs/symptoms and QoL were also improved. Conclusions: In an international, real-world, clinical-practice setting, pasireotide sc was generally well-tolerated (no new safety signals were identified), effectively reduced UFC (normalization in ~50% of evaluable patients) and improved clinical signs and QoL in patients with CD. While hyperglycemia-related AEs were common, consistent with previous studies, most were manageable, with <6% of patients discontinuing treatment because of these events.

Keywords: Cushing's disease; clinical practice; pasireotide sc; real world; somatostatin analog.

Figures

Figure 1
Figure 1
Proportion of patients with (A) normalized mUFC and (B) a decrease in mUFC of ≥50% from baseline at weeks 12, 24, and 48, overall and by dose group. Values in parentheses are two-sided 95% exact CIs.
Figure 2
Figure 2
Mean percentage change in signs and symptoms of hypercortisolism from baseline to weeks 12, 24, and 48 in the overall study population. Values in parentheses are two-sided 95% exact CIs.
Figure 3
Figure 3
Proportion of patients with improvement, no change or worsening of categorical signs of Cushing's disease from baseline to last post-baseline value in the overall study population.
Figure 4
Figure 4
Proportion of patients with improvement, no change or worsening of categorical signs of Cushing's disease from baseline to last post-baseline value by dose group.

References

    1. Lacroix A, Feelders RA, Stratakis CA, Nieman LK. Cushing's syndrome. Lancet. (2015) 386:913–27. 10.1016/S0140-6736(14)61375-1
    1. Feelders RA, Pulgar SJ, Kempel A, Pereira AM. The burden of Cushing's disease: clinical and health-related quality of life aspects. Eur J Endocrinol. (2012) 167:311–26. 10.1530/EJE-11-1095
    1. Etxabe J, Vazquez JA. Morbidity and mortality in Cushing's disease: an epidemiological approach. Clin Endocrinol. (1994) 40:479–84. 10.1111/j.1365-2265.1994.tb02486.x
    1. Pivonello R, Isidori AM, De Martino MC, Newell-Price J, Biller BMK, Colao A. Complications of Cushing's syndrome: state of the art. Lancet Diabetes Endocrinol. (2016) 4:611–29. 10.1016/S2213-8587(16)00086-3
    1. Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price J, Savage MO, et al. . Treatment of Cushing's syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. (2015) 100:2807–31. 10.1210/jc.2015-1818
    1. Dekkers OM, Horváth-Puhó E, Jørgensen JO, Cannegieter SC, Ehrenstein V, Vandenbroucke JP, et al. . Multisystem morbidity and mortality in Cushing's syndrome: a cohort study. J Clin Endocrinol Metab. (2013) 98:2277–84. 10.1210/jc.2012-3582
    1. Graversen D, Vestergaard P, Stochholm K, Gravholt CH, Jorgensen JO. Mortality in Cushing's syndrome: a systematic review and meta-analysis. Eur J Intern Med. (2012) 23:278–82. 10.1016/j.ejim.2011.10.013
    1. Pivonello R, De Leo M, Cozzolino A, Colao A. The treatment of Cushing's disease. Endocr Rev. (2015) 36:385–486. 10.1210/er.2013-1048
    1. Tritos NA, Biller BMK. Medical therapy for Cushing's syndrome in the twenty-first century. Endocrinol Metab Clin North Am. (2018) 47:427–40. 10.1016/j.ecl.2018.01.006
    1. Fleseriu M, Hamrahian AH, Hoffman AR, Kelly DF, Katznelson L. American association of clinical endocrinologists and American college of endocrinology disease state clinical review: diagnosis of recurrence in Cushing disease. Endocr Pract. (2016) 22:1436–48. 10.4158/EP161512.DSCR
    1. Biller BMK, Grossman AB, Stewart PM, Melmed S, Bertagna X, Bertherat J, et al. . Treatment of adrenocorticotropin-dependent Cushing's syndrome: a consensus statement. J Clin Endocrinol Metab. (2008) 93:2454–62. 10.1210/jc.2007-2734
    1. Fleseriu M, Castinetti F. Updates on the role of adrenal steroidogenesis inhibitors in Cushing's syndrome: a focus on novel therapies. Pituitary. (2016) 19:643–53. 10.1007/s11102-016-0742-1
    1. de Bruin C, Feelders RA, Lamberts SW, Hofland LJ. Somatostatin and dopamine receptors as targets for medical treatment of Cushing's syndrome. Rev Endocr Metab Disord. (2009) 10:91–102. 10.1007/s11154-008-9082-4
    1. Fleseriu M, Petersenn S. New avenues in the medical treatment of Cushing's disease: corticotroph tumor targeted therapy. J Neurooncol. (2013) 114:1–11. 10.1007/s11060-013-1151-1
    1. Langlois F, Chu J, Fleseriu M. Pituitary-directed therapies for Cushing's disease. Front Endocrinol. (2018) 9:164. 10.3389/fendo.2018.00164
    1. Colao A, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. . A 12-month Phase 3 study of pasireotide in Cushing's disease. N Engl J Med. (2012) 366:914–24. 10.1056/NEJMoa1105743
    1. Yedinak CG, Hopkins S, Williams J, Ibrahim A, Cetas JS, Fleseriu M. Medical therapy with pasireotide in recurrent Cushing's disease: experience of patients treated for at least 1 year at a single center. Front Endocrinol. (2017) 8:35. 10.3389/fendo.2017.00035
    1. Trementino L, Michetti G, Angeletti A, Marcelli G, Concettoni C, Cardinaletti C, et al. . A single-center 10-year experience with pasireotide in Cushing's disease: patients' characteristics and outcome. Horm Metab Res. (2016) 48:290–8. 10.1055/s-0042-101347
    1. Pivonello R, Arnaldi G, Scaroni C, Giordano C, Cannavo S, Iacuaniello D, et al. . The medical treatment with pasireotide in Cushing's disease: an Italian multicentre experience based on “real-world evidence.” Endocrine. (2019) 64:657–72. 10.1007/s12020-018-1818-7
    1. Albani A, Ferraù F, Ciresi A, Pivonello R, Scaroni C, Iacuaniello D, et al. . Pasireotide treatment reduces cardiometabolic risk in Cushing's disease patients: an Italian, multicenter study. Endocrine. (2018) 61:118–24. 10.1007/s12020-018-1524-5
    1. Novartis Pharma AG. Signifor Summary of Product Characteristics. (2016). Available online at: (accessed October 2018).
    1. National Cancer Institute. Common Terminology Criteria for Adverse Events v3.0. (2006). Available online at: (accessed June, 2019).
    1. Webb SM, Badia X, Barahona MJ, Colao A, Strasburger CJ, Tabarin A, et al. . Evaluation of health-related quality of life in patients with Cushing's syndrome with a new questionnaire. Eur J Endocrinol. (2008) 158:623–30. 10.1530/EJE-07-0762
    1. Colao A, De Block C, Gaztambide MS, Kumar S, Seufert J, Casanueva FF. Managing hyperglycemia in patients with Cushing's disease treated with pasireotide: medical expert recommendations. Pituitary. (2014) 17:180–6. 10.1007/s11102-013-0483-3
    1. Silverstein JM. Hyperglycemia induced by pasireotide in patients with Cushing's disease or acromegaly. Pituitary. (2016) 19:536–43. 10.1007/s11102-016-0734-1
    1. Petersenn S, Salgado LR, Schopohl J, Portocarrero-Ortiz L, Arnaldi G, Lacroix A, et al. . Long-term treatment of Cushing's disease with pasireotide: 5-year results from an open-label extension study of a Phase III trial. Endocrine. (2017) 57:156–65. 10.1007/s12020-017-1316-3
    1. Attanasio R, Mainolfi A, Grimaldi F, Cozzi R, Montini M, Carzaniga C, et al. . Somatostatin analogs and gallstones: a retrospective survey on a large series of acromegalic patients. J Endocrinol Invest. (2008) 31:704–10. 10.1007/BF03346419
    1. Lacroix A, Gu F, Gallardo W, Pivonello R, Yu Y, Witek P, et al. . Efficacy and safety of once-monthly pasireotide in Cushing's disease: a 12 month clinical trial. Lancet Diabetes Endocrinol. (2018) 6:17–26. 10.1016/S2213-8587(17)30326-1
    1. Pivonello R, Petersenn S, Newell-Price J, Findling J, Gu F, Maldonado M, et al. . Pasireotide treatment significantly improves clinical signs and symptoms in patients with Cushing's disease: results from a Phase III study. Clin Endocrinol. (2014) 81:408–17. 10.1111/cen.12431
    1. Colao A, Cozzolino A, Pivonello R. Quality of life in patients with Cushing's disease: a modern approach. Clin Endocrinol. (2012) 76:776–7. 10.1111/j.1365-2265.2012.04344.x
    1. Castinetti F, Guignat L, Giraud P, Muller M, Kamenicky P, Drui D, et al. . Ketoconazole in Cushing's disease: is it worth a try? J Clin Endocrinol Metab. (2014) 99:1623–30. 10.1210/jc.2013-3628
    1. Young J, Bertherat J, Vantyghem MC, Chabre O, Senoussi S, Chadarevian R, et al. . Hepatic safety of ketoconazole in Cushing's syndrome: results of a compassionate use programme in France. Eur J Endocrinol. (2018) 178:447–58. 10.1530/EJE-17-0886
    1. Daniel E, Aylwin S, Mustafa O, Ball S, Munir A, Boelaert K, et al. . Effectiveness of metyrapone in treating Cushing's syndrome: a retrospective multicenter study in 195 patients. J Clin Endocrinol Metab. (2015) 100:4146–54. 10.1210/jc.2015-2616
    1. Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart DE, Gross C. Mifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing's syndrome. J Clin Endocrinol Metab. (2012) 97:2039–49. 10.1210/jc.2011-3350
    1. Castinetti F, Conte-Devolx B, Brue T. Medical treatment of Cushing's syndrome: glucocorticoid receptor antagonists and mifepristone. Neuroendocrinology. (2010) 92(Suppl. 1):125–30. 10.1159/000314224
    1. Feelders RA, de Bruin C, Pereira AM, Romijn JA, Netea-Maier RT, Hermus AR, et al. Pasireotide alone or with cabergoline and ketoconazole in Cushing's disease. N Engl J Med. (2010) 362:1846–8. 10.1056/NEJMc1000094

Source: PubMed

3
S'abonner