Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes

G Garcia-Manero, S D Gore, S Kambhampati, B Scott, A Tefferi, C R Cogle, W J Edenfield, J Hetzer, K Kumar, E Laille, T Shi, K J MacBeth, B Skikne, G Garcia-Manero, S D Gore, S Kambhampati, B Scott, A Tefferi, C R Cogle, W J Edenfield, J Hetzer, K Kumar, E Laille, T Shi, K J MacBeth, B Skikne

Abstract

CC-486, the oral formulation of azacitidine (AZA), is an epigenetic modifier and DNA methyltransferase inhibitor in clinical development for treatment of hematologic malignancies. CC-486 administered for 7 days per 28-day treatment cycle was evaluated in a phase 1 dose-finding study. AZA has a short plasma half-life and DNA incorporation is S-phase-restricted; extending CC-486 exposure may increase the number of AZA-affected diseased target cells and maximize therapeutic effects. Patients with lower-risk myelodysplastic syndromes (MDS) received 300 mg CC-486 once daily for 14 days (n=28) or 21 days (n=27) of repeated 28-day cycles. Median patient age was 72 years (range 31-87) and 75% of patients had International Prognostic Scoring System Intermediate-1 risk MDS. Median number of CC-486 treatment cycles was 7 (range 2-24) for the 14-day dosing schedule and 6 (1-24) for the 21-day schedule. Overall response (complete or partial remission, red blood cell (RBC) or platelet transfusion independence (TI), or hematologic improvement) (International Working Group 2006) was attained by 36% of patients receiving 14-day dosing and 41% receiving 21-day dosing. RBC TI rates were similar with both dosing schedules (31% and 38%, respectively). CC-486 was generally well-tolerated. Extended dosing schedules of oral CC-486 may provide effective long-term treatment for patients with lower-risk MDS.

Conflict of interest statement

GG-M has received research support from Celgene Corporation, and is a consultant for Celgene; SDG receives research support from and consults for the Celgene; SK declares no relevant conflicts of interest; B Scott has received research support from Celgene, and is a consultant and speaker for Celgene; AT declares no relevant conflicts of interest; CRC serves on the Scientific Advisory Board for the Connect MDS/AML Disease Registry study, which is sponsored by Celgene; WJE declares no relevant conflicts of interest; JH, KK, EL, TS, KJM and B Skikne are employees of, and own stock in, Celgene Corporation, Summit, NJ, USA.

Figures

Figure 1
Figure 1
Duration of CC-486 treatment and response. Gray bars indicate no response and green bars indicate a response.
Figure 2
Figure 2
(a) Mean (+s.d.) plasma concentration-vs-time profiles following SC azacitidine administration on days 1 and 7, and CC-486 300 mg once daily on days 1 and 14; and (b) Cumulative azacitidine exposure per cycle with extended CC-486 dosing regimens relative to azacitidine exposure with subcutaneous (SC) azacitidine 75 mg/m2 administered for 7 days. CC-486 480 mg/day was identified as the maximally tolerated dose (MTD).
Figure 3
Figure 3
(a) Kernel density plots of averaged genome-wide DNA methylation profiles across patients throughout the first treatment cycle. Shifts to the left from baseline (black line) represent overall reduction in methylation level. The x axis shows the percent of methylation on a locus and the y axis shows the density of loci at the different methylation levels; and (b) changes in global DNA methylation scores (GDMS) from baseline to day 28 (cycle end) in treatment cycle 1 vs clinical response. *Each dashed line represents one patient whereas the solid lines are the mean profiles of GDMS changes. The error bars represent one standard error away from the means. NR=no response; R=response. Response categories include overall response (complete remission (CR), any hematologic improvement (HI), RBC transfusion independence (TI) and platelet TI) and marrow CR (mCR). (*at any cycle of CC-486).

References

    1. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997; 89: 2079–2088.
    1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009; 10: 223–232.
    1. Lyons RM, Cosgriff TM, Modi SS, Gersh RH, Hainsworth JD, Cohn AL et al. Hematologic response to three alternative dosing schedules of azacitidine in patients with myelodysplastic syndromes. J Clin Oncol 2009; 27: 1850–1856.
    1. Musto P, Maurillo L, Spagnoli A, Gozzini A, Rivellini F, Lunghi M et al. Azacitidine for the treatment of lower risk myelodysplastic syndromes: a retrospective study of 74 patients enrolled in an Italian named patient program. Cancer 2010; 116: 1485–1494.
    1. Garcia-Manero G, Gore SD, Cogle C, Ward R, Shi T, Macbeth KJ et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol 2011; 29: 2521–2527.
    1. Li LH, Olin EJ, Buskirk HH, Reineke LM. Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia. Cancer Res 1970; 30: 2760–2769.
    1. Glover AB, Leyland-Jones B. Biochemistry of azacitidine: a review. Cancer Treat Rep 1987; 71: 959–964.
    1. Stresemann C, Bokelmann I, Mahlknecht U, Lyko F. Azacytidine causes complex DNA methylation responses in myeloid leukemia. Mol Cancer Ther 2008; 7: 2998–3005.
    1. Hollenbach PW, Nguyen AN, Brady H, Williams M, Ning Y, Richard N et al. A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. Plos One 2010; 5: e9001.
    1. Plagemann PG, Behrens M, Abraham D. Metabolism and cytotoxicity of 5-azacytidine in cultured Novikoff rat hepatoma and P388 mouse leukemia cells and their enhancement by preincubation with pyrazofurin. Cancer Res 1978; 38: 2458–2466.
    1. Juttermann R, Li E, Jaenisch R. Toxicity of 5-aza-2'-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. Proc Natl Acad Sci USA 1994; 91: 11797–11801.
    1. Li LH, Olin EJ, Fraser TJ, Bhuyan BK. Phase specificity of 5-azacytidine against mammalian cells in tissue culture. Cancer Res 1970; 30: 2770–2775.
    1. Jones PA, Taylor SM, Wilson V. DNA modification, differentiation, and transformation. J Exp Zool 1983; 228: 287–295.
    1. Leone G, Teofili L, Voso MT, Lubbert M. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica 2002; 87: 1324–1341.
    1. Stresemann C, Brueckner B, Musch T, Stopper H, Lyko F. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 2006; 66: 2794–2800.
    1. Flotho C, Claus R, Batz C, Schneider M, Sandrock I, Ihde S et al. The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia 2009; 23: 1019–1028.
    1. Momparler RL, Goodman J. In vitro cytotoxic and biochemical effects of 5-aza-2'-deoxycytidine. Cancer Res 1977; 37: 1636–1639.
    1. Ghoshal K, Datta J, Majumder S, Bai S, Kutay H, Motiwala T et al. 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol 2005; 25: 4727–4741.
    1. Santi DV, Garrett CE, Barr PJ. On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell 1983; 33: 9–10.
    1. Friedman S, Cheong LC. Effect of 5-azacytidine on deoxyribonucleic acid methylation in Escherichia coli K12. Biochem Pharmacol 1984; 33: 2675–2679.
    1. Jones PA. Effects of 5-azacytidine and its 2'-deoxyderivative on cell differentiation and DNA methylation. Pharmacol Ther 1985; 28: 17–27.
    1. Aimiuwu J, Wang H, Chen P, Xie Z, Wang J, Liu S et al. RNA-dependent inhibition of ribonucleotide reductase is a major pathway for 5-azacytidine activity in acute myeloid leukemia. Blood 2012; 119: 5229–5238.
    1. Saunthararajah Y. Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes. Hematology Am Soc Hematol Educ Program; 2013: 511–521.
    1. Prebet T, Sun Z, Figueroa ME, Ketterling R, Melnick A, Greenberg P et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup Trial E1905. J Clin Oncol 2014; 32: 1242–1248.
    1. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the Cancer and Leukemia Group B. J Clin Oncol 2002; 20: 2429–2440.
    1. Cheson BD, Greenberg PL, Bennett JM, Lowenberg B, Wijermans PW, Nimer SD et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood 2006; 108: 419–425.
    1. Bennett JM. World Health Organization classification of the acute leukemias and myelodysplastic syndrome. Int J Hematol 2000; 72: 131–133.
    1. Sproul D, Nestor C, Culley J, Dickson JH, Dixon JM, Harrison DJ et al. Transcriptionally repressed genes become aberrantly methylated and distinguish tumors of different lineages in breast cancer. Proc Natl Acad Sci USA 2011; 108: 4364–4369.
    1. Sekeres MA. How to manage lower-risk myelodysplastic syndromes. Leukemia 2012; 26: 390–394.
    1. Fenaux P. Response assessments in low-risk and high-risk myelodysplastic syndromes (MDS). Semin Oncol 2005; 32: S11–S15.
    1. Garcia-Manero G. Myelodysplastic syndromes: 2014 update on diagnosis, risk-stratification, and management. Am J Hematol 2014; 89: 97–108.
    1. Garcia-Manero G, Shan J, Faderl S, Cortes J, Ravandi F, Borthakur G et al. A prognostic score for patients with lower risk myelodysplastic syndrome. Leukemia 2008; 22: 538–543.
    1. Laille E, Shi T, Garcia-Manero G, Cogle CR, Gore SD, Hetzer J et al. Pharmacokinetics and pharmacodynamics with extended dosing of CC-486 in patients with hematologic malignancies. Plos One 2015; 10: e0135520.
    1. Gonzalez-Porras JR, Cordoba I, Such E, Nomdedeu B, Vallespi T, Carbonell F et al. Prognostic impact of severe thrombocytopenia in low-risk myelodysplastic syndrome. Cancer 2011; 117: 5529–5537.
    1. Santini V, Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Silverman LR, List A et al. Management and supportive care measures for adverse events in patients with myelodysplastic syndromes treated with azacitidine. Eur J Haematol 2010; 85: 130–138.
    1. Garcia-Manero G, Jabbour E, Borthakur G, Faderl S, Estrov Z, Yang H et al. Randomized open-label phase II study of decitabine in patients with low- or intermediate-risk myelodysplastic syndromes. J Clin Oncol 2013; 31: 2548–2553.
    1. Seymour JF, Fenaux P, Silverman LR, Mufti GJ, Hellstrom-Lindberg E, Santini V et al. Effects of azacitidine compared with conventional care regimens in elderly (⩾75 years) patients with higher-risk myelodysplastic syndromes. Crit Rev Oncol Hematol 2010; 76: 218–227.
    1. Bristol-Myers Squibb Company, Princeton NJ, USA, Rev 10/2010.
    1. Laille E, Savona MR, Scott BL, Boyd TE, Dong Q, Skikne B. Pharmacokinetics of different formulations of oral azacitidine (CC-486) and the effect of food and modified gastric pH on pharmacokinetics in subjects with hematologic malignancies. J Clin Pharmacol 2014; 54: 630–639.
    1. Liu G, Franssen E, Fitch MI, Warner E. Patient preferences for oral versus intravenous palliative chemotherapy. J Clin Oncol 1997; 15: 110–115.
    1. Kang SP, Ratain MJ. Inconsistent labeling of food effect for oral agents across therapeutic areas: differences between oncology and non-oncology products. Clin Cancer Res 2010; 16: 4446–4451.
    1. Koch KM, Reddy NJ, Cohen RB, Lewis NL, Whitehead B, Mackay K et al. Effects of food on the relative bioavailability of lapatinib in cancer patients. J Clin Oncol 2009; 27: 1191–1196.
    1. Greenberg PL, Attar E, Bennett JM, Bloomfield CD, De Castro CM, Deeg HJ et al. NCCN Clinical Practice Guidelines in Oncology: myelodysplastic syndromes. J Natl Compr Canc Netw 2011; 9: 30–56.
    1. Cogle CR, Craig BM, Rollison DE, List AF. Incidence of the myelodysplastic syndromes using a novel claims-based algorithm: high number of uncaptured cases by cancer registries. Blood 2011; 117: 7121–7125.

Source: PubMed

3
購読する