892
Views
26
CrossRef citations to date
0
Altmetric
Drug Discovery Case History

The discovery and development of romidepsin for the treatment of T-cell lymphoma

&
Pages 859-873 | Received 03 Mar 2017, Accepted 08 Jun 2017, Published online: 22 Jun 2017

References

  • Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127:2375–2390.
  • Jiang M, Bennani NN, Feldman AL. Lymphoma classification update: T-cell lymphomas, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms. Expert Rev Hematol. 2017 Jan 29:1–11. Epub ahead of print. DOI:10.1080/17474086.2017.1281122
  • Bellei M, Chiattone CS, Luminari S, et al. T-Cell Lymphomas in South America and Europe. Rev Bras Hematol Hemoter. 2012;34:42–47.
  • Hsi ED, Horwitz SM, Carson KR, et al. Analysis of peripheral T-cell lymphoma diagnostic workup in the United States. Clin Lymphoma Myeloma Leuk. 2017;17:113–200.
  • Piekarz R, Bates S. A review of depsipeptide and other histone deacetylase inhibitors in clinical trials. Curr Pharm Des. 2004;10:2289–2298.
  • Richmond TJ, Davey CA. The structure of DNA in the nucleosome core. Nature. 2003;423:145–150.
  • Polo SE, Almouzni G. Cancer lett histone metabolic pathways and chromatin assembly factors as proliferation markers. Cancer Lett. 2005;220:1–9.
  • Litvinov IV, Jones DA, Sasseville D, et al. Transcriptional profiles predict disease outcome in patients with cutaneous T-cell lymphoma. Clin Cancer Res. 2010;16:2106–2114.
  • Prince HM, Dickinson M. Romidepsin for cutaneous T-cell lymphoma. Clin Cancer Res. 2012;18:3509–35015.
  • Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768–3785.
  • Whittaker SJ, Marsden JR, Spittle M, et al. Joint British Association of Dermatologists and U.K. Cutaneous Lymphoma Group guidelines for the management of primary cutaneous T-cell lymphomas. Br J Dermatol. 2003;149:1095–1107.
  • Gu W, Roeder RG. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell. 1997;90:595–606.
  • Boyes J, Byfield P, Nakatani Y, et al. Regulation of activity of the transcription factor GATA-1 by acetylation. Nature. 1998;396:594–598.
  • Kim EJ, Hess S, Richardson SK, et al. Immunopathogenesis and therapy of cutaneous T cell lymphoma. J Clin Invest. 2005;115:798–812.
  • Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080.
  • Zinzani PL, Bonthapally V, Huebner D, et al. Panoptic clinical review of the current and future treatment of relapsed/refractory T-cell lymphomas: peripheral T-cell lymphomas. Crit Rev Oncol Hematol. 2016;99:214–227.
  • Ellin F, Landström J, Jerkeman M, et al. Real-world data on prognostic factors and treatment in peripheral T-cell lymphomas: a study from the Swedish Lymphoma Registry. Blood. 2014;124:1570–1577.
  • d’Amore F, Relander T, Lauritzsen GF, et al. Up-front autologous stem-cell transplantation in peripheral T-cell lymphoma: NLG-T-01. J Clin Oncol. 2012;30:3093–3099.
  • O’Connor OA, Pro B, Pinter-Brown L, et al. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol. 2011;29:1182–1189.
  • Coiffier B, Pro B, Prince HM, et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol. 2012;30:631–636.
  • O’Connor OA, Horwitz S, Masszi T, et al. Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the Pivotal Phase II BELIEF (CLN-19) Study. J Clin Oncol. 2015;33:2492–2499.
  • Pro B, Advani R, Brice P, et al. Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J Clin Oncol. 2012;30:2190–2196.
  • Ogura M, Ishida T, Hatake K, et al. Multicenter phase II study of mogamulizumab (KW-0761), a defucosylated anti-cc chemokine receptor 4 antibody, in patients with relapsed peripheral T-cell lymphoma and cutaneous T-cell lymphoma. J Clin Oncol. 2014;32:1157–1163.
  • Kuo MH, Allis CD. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays. 1998;20:615–626.
  • Ueda H, Manda T, Matsumoto S, et al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. III. Antitumor activities on experimental tumors in mice. J Antibiot (Tokyo). 1994;47:315–323.
  • Rosato RR, Grant S. Histone deacetylase inhibitors in clinical development. Expert Opin Investig Drugs. 2004;13:21–38.
  • Huang HC, Liu YC, Liu SH, et al. Geldanamycin inhibits trichostatin A-induced cell death and histone H4 hyperacetylation in COS-7 cells. Life Sci. 2002;70:1763–1775.
  • FDA Approval for Romidepsin. Available from: https://www.cancer.gov/about-cancer/treatment/drugs/fda-romidepsin. Updated: 2013 July 3 .Cited: 2017 June 6.
  • Chung CG, Poligone B. Cutaneous T cell lymphoma: an update on pathogenesis and systemic therapy. Curr Hematol Malig Rep. 2015;10:468–746.
  • Emanuele S, Lauricella M, Tesoriere G. Histone deacetylase inhibitors: apoptotic effects and clinical implications. Int J Oncol. 2008;33:637–646.
  • Glaser KB. HDAC inhibitors: clinical update and mechanism-based potential. Biochem Pharmacol. 2007;74:659–671.
  • Fukuda H, Sano N, Muto S, et al. Simple histone acetylation plays a complex role in the regulation of gene expression. Brief Funct Genomic Proteomic. 2006;5:190–208.
  • Mitsiades CS, Mitsiades NS, McMullan CJ, et al. Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci U S A. 2004;101:540–545.
  • Mahlknecht U, Hoelzer D. Histone acetylation modifiers in the pathogenesis of malignant disease. Mol Med. 2000;6:623–644.
  • Murugan K, Sangeetha S, Ranjitha S, et al. HDACiDB: a database for histone deacetylase inhibitors. Drug Des Devel Ther. 2015;9:2257–2264.
  • FDA approves Farydak for treatment of multiple myeloma. U.S. Drug&Food Administration.  Available from: https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm435296.htm. Retrieved 20 March 2017.
  • FDA approves Beleodaq to treat rare., aggressive form of non-Hodgkin lymphoma. GoodPill Primary Medical Supply. Available from: http://www.goodpillpharmacy.com/health-news/fda-approves-beleodaq-to-treat-rare-aggressive-form-of-non-hodgkin-lymphoma. Retrieved 20 March 2017.
  • Li KW, Wu J, Xing W, et al. Total synthesis of the antitumor depsipeptide FR-901,228. J Am Chem Soc. 1996;118:7237–7238.
  • Nakajima H, Kim YB, Terano H, et al. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Exp Cell Res. 1998;241:126–133.
  • Thiagalingam S, Cheng KH, Lee HJ, et al. Histone deacetylases: unique players in shaping the epigenetic histone code. Ann N Y Acad Sci. 2003;983:84–100.
  • Sandor V, Robbins AR, Robey R, et al. FR901228 causes mitotic arrest but does not alter microtubule polymerization. Anticancer Drugs. 2000;11:445–454.
  • Manoharan TH, Burgess JA, Ho D, et al. Integration of a mutant c-Ha-ras oncogene into C3H/10T1/2 cells and its relationship to tumorigenic transformation. Carcinogenesis. 1985;6:1295–1301.
  • Lech-Maranda E, Robak E, Robak T. Novel systemic drugs for cutaneous T-cell, lymphoma. Recent Pat Anticancer Drug Discov. 2011;6:70–93.
  • Rajgolikar G, Chan KK, Wang HC. Effects of a novel antitumor depsipeptide, FR901228, on human breast cancer cells. Breast Cancer Res Treat. 1998;51:29–38.
  • Yoshida M, Furumai R, Nishiyama M, et al. Histone deacetylase as a new target for cancer chemotherapy. Cancer Chem Pharm. 2001;48(Suppl 1):S20–S26.
  • Shigematsu N, Ueda H, Takase S, et al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. II. Structure determination. J Antibiot. 1994;47:311–314.
  • Barbarotta L, Hurley K. Romidepsin for the treatment of peripheral T-cell lymphoma. J Adv Pract Oncol. 2015;6:22–36.
  • Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res. 2003;63:3637–3645.
  • Ueda H, Nakajima H, Hori Y, et al. Action of FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum no. 968, on Ha-ras transformed NIH3T3 cells-+. Biosci Biotechnol Biochem. 1994;58:1579–1583.
  • Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002;108:153–164.
  • Xiao JJ, Byrd J, Marcucci G, et al. Identification of thiols and glutathione conjugates of depsipeptide FK228 (FR901228), a novel histone protein deacetylase inhibitor, in the blood. Rapid Commun Mass Spectrom. 2003;17:757–766.
  • Marks PA, Richon VM, Breslow R, et al. Histone deacetylase inhibitors as new cancer drugs. Curr Opin Oncol. 2001;13:477–483.
  • Klisovic MI, Maghraby EA, Parthun MR, et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cells. Leukemia. 2003;17:350–358.
  • Furumai R, Matsuyama A, Kobashi N, et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res. 2002;62:4916–4921.
  • Archer SY, Meng S, Shei A, et al. RA. p21WAF1 is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc Natl Acad Sci USA. 1998;95:6791–6796.
  • Ueda H, Nakajima H, Hori Y, et al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. I. Taxonomy, fermentation, isolation, physicochemical and biological properties, and antitumor activity. J Antibiot. 1994;47:301–310.
  • Sandor V, Senderowicz A, Mertins S, et al. P21-dependent G1 arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer. 2000;83:817–825.
  • Peart MJ, Tainton KM, Ruefli AA, et al. Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res. 2003;63:4460–4471.
  • Burgess AJ, Pavey S, Warrener R, et al. Up-regulation of p21WAF1/CIP1 by histone deacetylase inhibitors reduces their cytotoxicity. Mol Pharmacol. 2001;60:828–837.
  • Insinga A, Monestiroli S, Ronzoni S, et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat Med. 2005;11:71–76.
  • Pelletier G, Stefanovsky VY, Faubladier M, et al. Competitive recruitment of CBP and Rb-HDAC regulates UBF acetylation and ribosomal transcription. Mol Cell. 2000;6:1059–1066.
  • Nebbioso A, Clarke N, Voltz E, et al. Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med. 2005;11:77–84.
  • Lech-Maranda E, Robak E, Korycka A, et al. Depsipeptide (FK228) as a novel histone deacetylase mechanism of action and anticancer activity. Mini Rev Med Chem. 2007;7:1062–1069.
  • Shao Y, Gao Z, Marks PA, et al. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci USA. 2004;101:18030–18035.
  • Yu X, Guo ZS, Marcu MG, et al. Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228. Natl Cancer Inst. 2002;94:504–513.
  • Bali P, Pranpat M, Bradner J, et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem. 2005;280:26729–26734.
  • Sutheesophon K, Kobayashi Y, Takatoku MA, et al. Histone deacetylase inhibitor depsipeptide (FK228) induces apoptosis in leukemic cells by facilitating mitochondrial translocation of Bax, which is enhanced by the proteasome inhibitor bortezomib. Acta Haematol. 2006;115:78–90.
  • Bates SE, Eisch R, Ling A, et al. Romidepsin in peripheral and cutaneous T-cell lymphoma: mechanistic implications from clinical and correlative data. Br J Haematol. 2015;170:96–109.
  • Kwon HJ, Kim MS, Kim MJ, et al. Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int J Cancer. 2002;97:290–296.
  • Sasakawa Y, Naoe Y, Noto T, et al. Antitumor efficacy of FK228, a novel histone deacetylase inhibitor, depends on the effect on expression of angiogenesis factors. Biochem Pharmacol. 2003;66:897–906.
  • Woo S, Gardner ER, Chen X, et al. Population pharmacokinetics of romidepsin in patients with cutaneous T-cell lymphoma and relapsed peripheral T-cell lymphoma. Clin Cancer Res. 2009;15:1496–1503.
  • Sandor V, Bakke S, Robey RW, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res. 2002;8:718–728.
  • Marshall JL, Rizvi N, Kauh J, et al. A phase I trial of romidepsin (FR901228) in patients with advanced cancer. J Exp Ther Oncol. 2002;2:325–332.
  • Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood. 2005;105:959–967.
  • Fouladi M, Furman WL, Chin T, et al. Phase I study of depsipeptide in pediatric patients with refractory solid tumors: a Children’s Oncology Group report. J Clin Oncol. 2006;24:3678–3685.
  • Poligone B, Lin J, Chung C. Romidepsin: evidence for its potential use to manage previously treated cutaneous T cell lymphoma. Core Evid. 2011;6:1–12.
  • Bates SE, Zhan Z, Steadman K, et al. Laboratory correlates for a phase II trial of romidepsin in cutaneous and peripheral T-cell lymphoma. Br J Haematol. 2010;148:256–267.
  • Grant C, Rahman F, Piekarz R, et al. Romidepsin: a new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev Anticancer Ther. 2010;10:997–1008.
  • Fronsdal K, Saatcioglu F. Histone deacetylase inhibitors differentially mediate apoptosis in prostate cancer cells. Prostate. 2005;62:299–306.
  • Ito T, Ouchida M, Morimoto Y, et al. Significant growth suppression of synovial sarcomas by the histone deacetylase inhibitor FK228 in vitro and in vivo. Cancer Lett. 2005;224:311–319.
  • Piekarz RL, Robey RW, Zhan Z, et al. T-cell lymphoma as a model for the use of histone deacetylase inhibitors in cancer therapy: impact of depsipeptide on molecular markers, therapeutic targets, and mechanisms of resistance. Blood. 2004;103:4636–4643.
  • Wei DG, Chiang V, Fyne E, et al. Histone deacetylase inhibitor romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral therapy at concentrations achieved by clinical dosing. PLoS Pathog. 2014;10:e1004071.
  • Sun WJ, Huang H, He B, et al. Romidepsin induces G2/M phase arrest via Erk/cdc25C/cdc2/cyclinB pathway and apoptosis induction through JNK/c-Jun/caspase3 pathway in hepatocellular carcinoma cells. Biochem Pharmacol. 2016;127:90–100.
  • Tiffon C, Adams J, Van Der Fits L, et al. The histone deacetylase inhibitors vorinostat and romidepsin downmodulate IL-10 expression in cutaneous T-cell lymphoma cells. Br J Pharmacol. 2011;162:1590–1602.
  • Akilov OE, Grant C, Frye R, et al. Low-dose electron beam radiation and romidepsin therapy for symptomatic cutaneous T-cell lymphoma lesions. Br J Dermatol. 2012;167:194–197.
  • Kano Y, Akutsu M, Tsunoda S, et al. Cytotoxic effects of histone deacetylase inhibitor FK228 (depsipeptide, formally named FR901228) in combination with conventional anti-leukemia/lymphoma agents against human leukemia/lymphoma cell lines. Invest New Drugs. 2007;25:31–40.
  • Zheng H, Zhao W, Yan C, et al. HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2016;22:4119–4132.
  • Chakraborty AR, Robey RW, Luchenko VL, et al. MAPK pathway activation leads to Bim loss and histone deacetylase inhibitor resistance: rationale to combine romidepsin with an MEK inhibitor. Blood. 2013;121:4115–4125.
  • Valdez BC, Brammer JE, Li Y, et al. Romidepsin enhances the cytotoxicity of fludarabine, clofarabine and busulfan combination in malignant T-cells. Leuk Res. 2016;47:100–118.
  • Rozati S, Cheng PF, Widmer DS, et al. Romidepsin and azacitidine synergize in their epigenetic modulatory effects to induce apoptosis in CTCL. Clin Cancer Res. 2016;22:2020–2031.
  • Cosenza M, Civallero M, Fiorcari S, et al. The histone deacetylase inhibitor romidepsin synergizes with lenalidomide and enhances tumor cell death in T-cell lymphoma cell lines. Cancer Biol Ther. 2016;12:1–13.
  • Beg AA, Gray JE. HDAC inhibitors with PD-1 blockade: a promising strategy for treatment of multiple cancer types? Epigenomics. 2016;8:1015–1017.
  • Makena MR, Koneru B, Nguyen TH, et al. Reactive oxygen species-mediated synergism of fenretinide and romidepsin in preclinical models of T-cell lymphoid malignancies. Mol Cancer Ther. 2017 Jan 23;pii: molcanther.0749. Epub ahead of print. Doi: 10.1158/1535-7163.MCT-16-0749
  • Reddy SA. Romidepsin for the treatment of relapsed/refractory cutaneous T-cell lymphoma (mycosis fungoides/Sézary syndrome): use in a community setting. Crit Rev Oncol Hematol. 2016;106:99–107.
  • Kim M, Thompson LA, Wenger SD, et al. Romidepsin: a histone deacetylase inhibitor for refractory cutaneous T-cell lymphoma. Ann Pharmacother. 2012;46:1340–1348.
  • Kim EJ, Kim YH, Rook AH, et al. Clinically significant responses achieved with romidepsin across disease compartments in patients with cutaneous T-cell lymphoma. Leuk Lymphoma. 2015;56:2847–2854.
  • Foss F, Duvic M, Lerner A, et al. Clinical efficacy of romidepsin in tumor stage and folliculotropic mycosis fungoides. Clin Lymphoma Myeloma Leuk. 2016;16:637–643.
  • Foss F, Pro B, Miles Prince H, et al. Responses to romidepsin by line of therapy in patients with relapsed or refractory peripheral T-cell lymphoma. Cancer Med. 2017;6:36–44.
  • Piekarz RL, Robey R, Sandor V, et al. Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood. 2001;98:2865–2868.
  • Piekarz RL, Frye R, Turner M, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol. 2009;27:5410–5417.
  • Kim YH, Demierre MF, Kim EJ, et al. Clinically meaningful reduction in pruritus in patients with cutaneous T-cell lymphoma treated with romidepsin. Leuk Lymphoma. 2013;54:284–289.
  • Whittaker SJ, Demierre MF, Kim EJ, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol. 2010;28:4485–4491.
  • Martinez-Escala ME, Kuzel TM, Kaplan JB, et al. Durable responses with maintenance dose-sparing regimens of romidepsin in cutaneous T-cell lymphoma. JAMA Oncol. 2016;2:790–793.
  • Lemchak DM, Akilov OE. Romidepsin controls chronic lymphocytic leukemia in a patient with mycosis fungoides. Hematol Rep. 2016;8:6840.
  • Piekarz RL, Frye R, Prince HM, et al. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood. 2011 Jun 2;117:5827–5834.
  • Dupuis J, Morschhauser F, Ghesquières H, et al. Combination of romidepsin with cyclophosphamide, doxorubicin, vincristine, and prednisone in previously untreated patients with peripheral T-cell lymphoma: a non-randomised, phase 1b/2 study. Lancet Haematol. 2015;2:e160–165.
  • Pellegrini C, Dodero A, Chiappella A, et al. Italian Lymphoma Foundation (Fondazione Italiana Linfomi Onlus, FIL). A phase II study on the role of gemcitabine plus romidepsin (GEMRO regimen) in the treatment of relapsed/refractory peripheral T-cell lymphoma patients. J Hematol Oncol. 2016;9:38.
  • Foss F, Horwitz S, Pro B, et al. Romidepsin for the treatment of relapsed/refractory peripheral T cell lymphoma: prolonged stable disease provides clinical benefits for patients in the pivotal trial. J Hematol Oncol. 2016;9:22.
  • Chan KL, Van Der Weyden C, Khoo C, et al. Durable clinical remission induced by romidepsin for chemotherapy-refractory peripheral T-cell lymphoma with central nervous system involvement. Leuk Lymphoma. 2017;58:996–998.
  • Chihara D, Fanale MA, Miranda RN, et al. The survival outcome of patients with relapsed/refractory peripheral T-cell lymphoma-not otherwise specified and angioimmunoblastic T-cell lymphoma. Br J Haematol. 2016;176:750–758.
  • Fanale MA, Fredrick B, Hagemeister FB, et al. A phase I trial of alisertib plus romidepsin for relapsed/refractory aggressive B- and T-cell lymphomas. Blood. 2014;124(supplement):abstract1744.
  • Holkova B, Kmieciak M, Bose P, et al. Phase 1 study of bortezomib and romidepsin in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, indolent B-cell lymphoma, peripheral T-cell lymphoma, or cutaneous T-cell lymphoma: updated results. Blood. 2014;124(supplement):abstract3050.
  • Northwestern University; Amgen. Dose-escalation trial of carfilzomib with and without romidepsin in cutaneous T-Cell Lymphoma. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2012. cited 2017June 6. Available from: https://clinicaltrials.gov/ct2/show/NCT01738594. NML Identifier: NCT01738594.
  • Lunning MA, Ruan J, Nair S, et al. A phase I/II trial of the combination of romidepsin and lenalidomide in patients with relapsed/refractory lymphoma and myeloma: phase 1 results. J Clin Oncol. 2014;32(supplement):abstract8582.
  • Mehta-Shah N, Lunning MA, Ruan J, et al. A phase I/II trial of the combination of romidepsin and lenalidomide in patients with relapsed/refractory lymphoma and myeloma. Hematol Oncol. 2015;33(abstract):016.
  • Buckstein R, Fraser G, Cheung M, et al. Alemtuzumab and CHOP chemotherapy for the treatment of aggressive histology peripheral T cell lymphomas: a multi-center phase I study. Clin Lymphoma Myeloma Leuk. 2016;16:18–28.
  • Johnston PB, Cashen AF, Nikolinakos PG, et al. Safe and effective treatment of patients with peripheral T-cell lymphoma (PTCL) with the novel HDAC inhibitor, belinostat, in combination with CHOP: results of the Bel-CHOP Phase 1 Trial. Blood. 2015;124(supplement):abstract253.
  • Advani RH, Ansell SM, Lechowicz MJ, et al. A phase II study of cyclophosphamide, etoposide, vincristine and prednisone (CEOP) alternating with pralatrexate (P) as front line therapy for patients with peripheral T-cell lymphoma (PTCL): final results from the T- cell consortium trial. Br J Haematol. 2016;172:535–544.
  • Molife LR, Attard G, Fong PC, et al. Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC). Ann Oncol. 2010;21:109–113.
  • Nettersheim D, Jostes S, Fabry M, et al. A signaling cascade including ARID1A, GADD45B and DUSP1 induces apoptosis and affects the cell cycle of germ cell cancers after romidepsin treatment. Oncotarget. 2016;7:74931–74946.
  • Amiri-Kordestani L, Luchenko V, Peer CJ, et al. Phase I trial of a new schedule of romidepsin in patients with advanced cancers. Clin Cancer Res. 2013;19:4499–4507.
  • Holkova B, Yazbeck V, Kmieciak M, et al. A phase 1 study of bortezomib and romidepsin in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, indolent B-cell lymphoma, peripheral T-cell lymphoma, or cutaneous T-cell lymphoma. Leuk Lymphoma. 2017 Jan;19:1–9. Epub ahead of print. Doi:10.1080/10428194.2016.1276287
  • Piekarz RL, Frye AR, Wright JJ, et al. Cardiac studies in patients treated with romidepsin, FK228, in a phase II trial for T-cell lymphoma. Clin Cancer Res. 2006;12:3762–3773.
  • Rivers ZT, Oostra DR, Westholder JS, et al. Romidepsin-associated cardiac toxicity and ECG changes: a case report and review of the literature. J Oncol Pharm Pract. 2016 Oct 5;pii: 1078155216673229. Epub ahead of print.
  • Noonan AM, Eisch RA, Liewehr DJ, et al. Electrocardiographic studies of romidepsin demonstrate its safety and identify a potential role for K(ATP) channel. Clin Cancer Res. 2013;19:3095–3104.
  • Moskowitz AJ, Horwitz SM. Targeting histone deacetylases in T-cell lymphoma. Leuk Lymphoma. 2017;58:1306–1319.
  • Cooper MR, Brewer S, Broketa G. Reversible ageusia associated with romidepsin therapy. Am J Health Syst Pharm. 2013;70:1502–1505.
  • Brian Scott B, Wickless H. Romidepsin-induced neutrophilic urticaria. JAAD Case Rep. 2016;2:261–263.
  • Battistuzzi G, Giannini G. Synthesis of ST7612AA1, a novel oral HDAC inhibitor, via radical thioacetic acid addition. Curr Bioact Compd. 2016;12:282–288.
  • Rajski SR, Wassarman DA, Restituyo JA FK228 analogs and methods of making and using the same. U.S. 20060128660 A1 (2006). Available from: https://www.google.com/patents/US20060128660. Retrieved 21 March 2017

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.