Phase I dose-escalation study of long-acting pasireotide in patients with neuroendocrine tumors

James C Yao, Jennifer A Chan, Alain C Mita, Madan G Kundu, Karina Hermosillo Reséndiz, Ke Hu, Shoba Ravichandran, Jonathan R Strosberg, Edward M Wolin, James C Yao, Jennifer A Chan, Alain C Mita, Madan G Kundu, Karina Hermosillo Reséndiz, Ke Hu, Shoba Ravichandran, Jonathan R Strosberg, Edward M Wolin

Abstract

This phase I study aimed at determining the maximum tolerated dose (MTD) and characterizing the safety, tolerability, pharmacokinetics (PKs), and efficacy of pasireotide in patients with advanced neuroendocrine tumors (NETs). Patients were enrolled in two phases: dose-escalation phase (to determine the MTD) at a starting dose of 80 mg pasireotide long-acting release (LAR) i.m. followed by a dose-expansion phase (to evaluate safety and prelimi-nary efficacy). Associations between PK/pharmacodynamic parameters and clinical outcomes were evaluated using linear regression analysis. A total of 29 patients were treated with 80 mg (n=13) and 120 mg (n=16) doses. Most common primary tumor sites included small intestine (44.8%), pancreas (24.1%), and lung (17.2%). No protocol-defined dose-limiting toxicities were observed in the study; however, in post hoc analysis, a higher incidence of bradycardia (heart rate [HR] <40 beats per minute [bpm]) was observed with 120 mg (31.3%) vs 80 mg (0%). Two partial responses (PRs) were observed, both in the 120 mg dose cohort. Pasireotide concentrations correlated with tumor shrinkage, although the association was not statistically significant (P=0.08). Among the biomarkers analyzed, insulin-like growth factor 1 (IGF-1) showed a decreasing trend with increasing pasireotide concentration, while chromogranin A (CgA) and neuron-specific enolase (NSE) levels did not show any dose-response relationship. The most common adverse events in any dose group were hyperglycemia, fatigue, and nausea. MTD was defined at 120 mg for pasireotide LAR in patients with advanced NETs. Although objective radiographic responses were rarely observed with somatostatin analogs, two PRs were observed among 16 patients in the 120 mg cohort. Bradycardia (HR <40 bpm) appears to be a dose-limiting effect; however, the mechanism and clinical significance are uncertain. This study was registered with clinicaltrials.gov (NCT01364415).

Keywords: Bayesian logistic regression model; MTD; dose escalation with overdose control; pharmacodynamics; pharmacokinetics.

Conflict of interest statement

Disclosure JCY has received consulting or advisory fees from Ipsen, Lexicon, and Novartis and research funding from Novartis. MGK, KHR, KH, and SR are employees of Novartis. JRS has received honoraria from Novartis; consulting or advisory fees from Ipsen, Lexicon, and Novartis; and research funding from Novartis and Pfizer and is on the speaker’s bureau for Bayer and Genentech. EMW has received consulting or advisory fees from Celgene, Ipsen, and Novartis. ACM is a member of the Speaker Bureau for Genentech. JAC has received consulting or advisory fees from Ipsen, Lexicon, Novartis, and Oxigene and has stock in Merck. The authors report no other conflicts of interest in this work.

Figures

Figure 1
Figure 1
Postulated mechanism of action of pasireotide.
Figure 2
Figure 2
Kaplan–Meier estimates of PFS by local radiological review. Abbreviations: CI, confidence interval; NE, not estimable; PFS, progression-free survival; LAR, long-acting release.
Figure 3
Figure 3
Results of the PK/pharmacodynamic analysis. Notes: (A) Mean PK–concentration–time profiles from the PK analysis set. CxDy: Cycle x, Day y. Negative (mean − SD) values were truncated to zero. Below the LLOQ values (<0.15 ng/mL) were set to zero. Day 1 pre-dose concentrations on C2 through C12 represent Cmin concentrations; day 22 concentration from the C1 represents the Cmax concentration. (B) Change from baseline tumor size vs pasireotide trough concentrations. (C) Change from baseline tumor size vs pasireotide trough concentrations. (D) IGF-1 vs pasireotide trough concentrations. (E) CgA vs pasireotide trough concentrations. (F) NSE vs pasireotide trough concentrations. Abbreviations: PK, pharmacokinetic; LLOQ, lower limit of quantitation; C2, Cycle 2; C12, Cycle 12; C1, Cycle 1; CgA, chromogranin A; NSE, neuron-specific enolase; LAR, long-acting release.
Figure 4
Figure 4
Predicted probability of bradycardia vs pasireotide LAR concentration. Abbreviations: CI, confidence interval; LAR, long-acting release; HR, heart rate; bpm, beats per minute.

References

    1. Gardner-Roehnelt NM. Update on the management of neuroendocrine tumors: focus on somatostatin antitumor effects. Clin J Oncol Nurs. 2012;16(1):56–64.
    1. di Bartolomeo M, Bajetta E, Buzzoni R, et al. Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian Trials in Medical Oncology Group. Cancer. 1996;77(2):402–408.
    1. Oberg K. Management of neuroendocrine tumours. Ann Oncol. 2004;15(suppl 4):iv293–iv298.
    1. Kvols LK, Moertel CG, O’Connell MJ, Schutt AJ, Rubin J, Hahn RG. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med. 1986;315(11):663–666.
    1. Rubin J, Ajani J, Schirmer W, et al. Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol. 1999;17(2):600–606.
    1. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27(28):4656–4663.
    1. Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–233.
    1. Berardi R, Rinaldi S, Torniai M, et al. Gastrointestinal neuroendocrine tumors: searching the optimal treatment strategy – a literature review. Crit Rev Oncol Hematol. 2016;98:264–274.
    1. Schmid HA, Schoeffter P. Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology. 2004;80(suppl 1):47–50.
    1. de Herder WW, Hofland LJ, van der Lely AJ, Lamberts SW. Somatostatin receptors in gastroentero-pancreatic neuroendocrine tumours. Endocr Relat Cancer. 2003;10(4):451–458.
    1. Villaume K, Blanc M, Gouysse G, et al. VEGF secretion by neuroendocrine tumor cells is inhibited by octreotide and by inhibitors of the PI3K/AKT/mTOR pathway. Neuroendocrinology. 2010;91(3):268–278.
    1. Tulipano G, Stumm R, Pfeiffer M, Kreienkamp HJ, Höllt V, Schulz S. Differential beta-arrestin trafficking and endosomal sorting of somatostatin receptor subtypes. J Biol Chem. 2004;279(20):21374–21382.
    1. 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.
    1. Li M, Li W, Kim HJ, Yao Q, Chen C, Fisher WE. Characterization of somatostatin receptor expression in human pancreatic cancer using real-time RT-PCR. J Surg Res. 2004;119(2):130–137.
    1. Ronga G, Salerno G, Procaccini E, et al. 111In-octreotide scintigraphy in metastatic medullary thyroid carcinoma before and after octreotide therapy: in vivo evidence of the possible down-regulation of somatostatin receptors. Q J Nucl Med. 1995;39(4 suppl 1):134–136.
    1. Plockinger U, Wiedenmann B. Treatment of gastroenteropancreatic neuroendocrine tumors. Virchows Arch. 2007;451(suppl 1):S71–S80.
    1. Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.
    1. Chen X, Shen G, Jiang J, et al. Pharmacokinetics and safety of subcutaneous pasireotide and intramuscular pasireotide long-acting release in Chinese male healthy volunteers: a phase I, single-center, open-label, randomized study. Clin Ther. 2014;36(8):1196–1210.
    1. Wolin EM, Jarzab B, Eriksson B, et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Devel Ther. 2015;9:5075–5086.
    1. Cives M, Kunz PL, Morse B, et al. Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer. 2015;22(1):1–9.
    1. Kulke M, Ruszniewski P, Van Cutsem E, et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol. 2017 Mar 6; Epub.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med. 2008;27(13):2420–2439.
    1. Love JN, Howell JM. Glucagon therapy in the treatment of symptomatic bradycardia. Ann Emerg Med. 1997;29(1):181–183.
    1. Bindon MJ, Barlotta K. Glucagon treatment for bradycardia and a junctional rhythm caused by excessive beta-blockade. Resuscitation. 2009;80(11):1327.
    1. Singh B, Smith JA, Axelrod DM, et al. Insulin-like growth factor-I inhibition with pasireotide decreases cell proliferation and increases apoptosis in pre-malignant lesions of the breast: a phase 1 proof of principle trial. Breast Cancer Res. 2014;16(6):463.
    1. Gut P, Czarnywojtek A, Fischbach J, et al. Chromogranin A – unspecific neuroendocrine marker. Clinical utility and potential diagnostic pitfalls. Arch Med Sci. 2016;12(1):1–9.
    1. Baudin E, Gigliotti A, Ducreux M, et al. Neuron-specific enolase and chromogranin A as markers of neuroendocrine tumours. Br J Cancer. 1998;78(8):1102–1107.

Source: PubMed

3
Předplatit