A Phase II Trial of a Histone Deacetylase Inhibitor Panobinostat in Patients With Low-Grade Neuroendocrine Tumors

Ning Jin, Sam J Lubner, Daniel L Mulkerin, Saurabh Rajguru, Lakeesha Carmichael, Herb Chen, Kyle D Holen, Noelle K LoConte, Ning Jin, Sam J Lubner, Daniel L Mulkerin, Saurabh Rajguru, Lakeesha Carmichael, Herb Chen, Kyle D Holen, Noelle K LoConte

Abstract

Lessons learned: Pancreatic neuroendocrine tumors versus carcinoid tumors should be examined separately in clinical trials.Progression-free survival is more clinically relevant as the primary endpoint (rather than response rate) in phase II trials for low-grade neuroendocrine tumors.

Background: The most common subtypes of neuroendocrine tumors (NETs) are pancreatic islet cell tumors and carcinoids, which represent only 2% of all gastrointestinal malignancies. Histone deacetylase (HDAC) inhibitors have already been shown to suppress tumor growth and induce apoptosis in various malignancies. In NET cells, HDAC inhibitors have resulted in increased Notch1 expression and subsequent inhibition of growth. We present here a phase II study of the novel HDAC inhibitor panobinostat in patients with low-grade NET.

Methods: Adult patients with histologically confirmed, metastatic, low-grade NETs and an Eastern Cooperative Oncology Group (ECOG) performance status of ≤2 were treated with oral panobinostat 20 mg once daily three times per week. Treatment was continued until patients experienced unacceptable toxicities or disease progression. The study was stopped at planned interim analysis based on a Simon two-stage design.

Results: Fifteen patients were accrued, and 13 were evaluable for response. No responses were seen, but the stable disease rate was 100%. The median progression-free survival (PFS) was 9.9 months, and the median overall survival was 47.3 months. Fatigue (27%), thrombocytopenia (20%), diarrhea (13%), and nausea (13%) were the most common related grade 3 toxicities. There was one grade 4 thrombocytopenia (7%). These results did not meet the prespecified criteria to open the study to full accrual.

Conclusion: The HDAC inhibitor panobinostat has a high stable disease rate and reasonable PFS in low-grade NET, but has a low response rate.

Trial registration: ClinicalTrials.gov NCT00985946.

©AlphaMed Press; the data published online to support this summary is the property of the authors.

Figures

Figure 1.
Figure 1.
Kaplan-Meier curve for median progression-free survival, which is 9.90 months with 90% confidence interval (4.10–16.9 months).
Figure 2.
Figure 2.
Percentage change in tumor size based on Response Evaluation Criteria in Solid Tumors (RECIST) criteria. Waterfall plot of radiographic changes from baseline to best response of 13 evaluable patients, revealing that every patient (100%) has stable disease. Stable disease is defined as neither sufficient shrinkage to qualify for partial response (less than 30% decrease in the sum of the longest diameters of target lesions) nor sufficient increase to qualify for progressive disease (at least 20% increase in the sum of the longest diameters of target lesions).
Figure 3.
Figure 3.
Kaplan-Meier curve for overall survival, which is 47.27 months with 90% confidence interval, with a follow-up time of 5 years. Abbreviation: CI, confidence interval.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4943400/bin/theoncologist_1660CTR_f4.jpg
Kaplan-Meier curve for median progression-free survival, which is 9.90 months with 90% confidence interval (4.10–16.9 months).

References

    1. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: Epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072.
    1. Kunz PL. Carcinoid and neuroendocrine tumors: Building on success. J Clin Oncol. 2015;33:1855–1863.
    1. Bosman FT, Carneiro F, Hruban RH, Theise ND, editors. WHO Classification of Tumours of the Digestive System. 4th ed. Geneva, Switzerland: WHO Press; 2010.
    1. Rinke A, Müller 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;27:4656–4663.
    1. Toumpanakis C, Caplin ME. Update on the role of somatostatin analogs for the treatment of patients with gastroenteropancreatic neuroendocrine tumors. Semin Oncol. 2013;40:56–68.
    1. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523.
    1. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513.
    1. Nakakura EK, Sriuranpong VR, Kunnimalaiyaan M, et al. Regulation of neuroendocrine differentiation in gastrointestinal carcinoid tumor cells by notch signaling. J Clin Endocrinol Metab. 2005;90:4350–4356.
    1. Kunnimalaiyaan M, Yan S, Wong F, et al. Hairy Enhancer of Split-1 (HES-1), a Notch1 effector, inhibits the growth of carcinoid tumor cells. Surgery. 2005;138:1137–1142; discussion 1142.
    1. Sriuranpong V, Borges MW, Ravi RK, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res. 2001;61:3200–3205.
    1. Radtke F, Raj K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756–767.
    1. Miyamoto Y, Maitra A, Ghosh B, et al. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565–576.
    1. Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, et al. Valproic acid activates notch-1 signaling and regulates the neuroendocrine phenotype in carcinoid cancer cells. The Oncologist. 2007;12:942–951.
    1. Greenblatt DY, Cayo M, Ning L, et al. Suberoyl bishydroxamic acid inhibits cellular proliferation by inducing cell cycle arrest in carcinoid cancer cells. J Gastrointest Surg. 2007;11:1515–1520; discussion 1520.
    1. Mohammed TA, Holen KD, Jaskula-Sztul R, et al. A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. The Oncologist. 2011;16:835–843.
    1. Atadja P. Development of the pan-DAC inhibitor panobinostat (LBH589): Successes and challenges. Cancer Lett. 2009;280:233–241.
    1. Wagner JM, Hackanson B, Lübbert M, et al. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics. 2010;1:117–136.
    1. Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine tumors: Consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol. 2011;29:934–943.
    1. Zikusoka MN, Kidd M, Eick G, et al. The molecular genetics of gastroenteropancreatic neuroendocrine tumors. Cancer. 2005;104:2292–2309.
    1. Murray-Lyon IM, Eddleston AL, Williams R, et al. Treatment of multiple-hormone-producing malignant islet-cell tumour with streptozotocin. Lancet. 1968;2:895–898.
    1. Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117:268–275.
    1. Fine RL, Gulati AP, Krantz BA, et al. Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol. 2013;71:663–670.
    1. Yao JC, Lagunes DR, Kulke MH. Targeted therapies in neuroendocrine tumors (NET): Clinical trial challenges and lessons learned. The Oncologist. 2013;18:525–532.
    1. Younes A, Sureda A, Ben-Yehuda D, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: Results of a phase II study. J Clin Oncol. 2012;30:2197–2203.
    1. DeAngelo DJ, Mesa RA, Fiskus W, et al. Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis. Br J Haematol. 2013;162:326–335.
    1. Duvic M, Dummer R, Becker JC, et al. Panobinostat activity in both bexarotene-exposed and -naïve patients with refractory cutaneous T-cell lymphoma: Results of a phase II trial. Eur J Cancer. 2013;49:386–394.
    1. Richardson PG, Schlossman RL, Alsina M, et al. PANORAMA 2: Panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood. 2013;122:2331–2337.
    1. Rathkopf D, Wong BY, Ross RW, et al. A phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancer. Cancer Chemother Pharmacol. 2010;66:181–189.
    1. Hainsworth JD, Infante JR, Spigel DR, et al. A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma. Cancer Invest. 2011;29:451–455.
    1. Wang H, Cao Q, Dudek AZ. Phase II study of panobinostat and bortezomib in patients with pancreatic cancer progressing on gemcitabine-based therapy. Anticancer Res. 2012;32:1027–1031.
    1. Morita S, Oizumi S, Minami H, et al. Phase I dose-escalating study of panobinostat (LBH589) administered intravenously to Japanese patients with advanced solid tumors. Invest New Drugs. 2012;30:1950–1957.
    1. Kelly WK, O’Connor OA, Krug LM, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol. 2005;23:3923–3931.
    1. Bromberg J. Stat proteins and oncogenesis. J Clin Invest. 2002;109:1139–1142.
    1. Fantin VR, Loboda A, Paweletz CP, et al. Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res. 2008;68:3785–3794.
    1. Fantin VR, Richon VM. Mechanisms of resistance to histone deacetylase inhibitors and their therapeutic implications. Clin Cancer Res. 2007;13:7237–7242.
    1. Slingerland M, Guchelaar HJ, Gelderblom H. Histone deacetylase inhibitors: An overview of the clinical studies in solid tumors. Anticancer Drugs. 2014;25:140–149.

Source: PubMed

3
Se inscrever