Pharmacokinetics and Pharmacodynamics with Extended Dosing of CC-486 in Patients with Hematologic Malignancies

Eric Laille, Tao Shi, Guillermo Garcia-Manero, Christopher R Cogle, Steven D Gore, Joel Hetzer, Keshava Kumar, Barry Skikne, Kyle J MacBeth, Eric Laille, Tao Shi, Guillermo Garcia-Manero, Christopher R Cogle, Steven D Gore, Joel Hetzer, Keshava Kumar, Barry Skikne, Kyle J MacBeth

Abstract

CC-486 (oral azacitidine) is an epigenetic modifier in development for patients with myelodysplastic syndromes and acute myeloid leukemia. In part 1 of this two-part study, a 7-day CC-486 dosing schedule showed clinical activity, was generally well tolerated, and reduced DNA methylation. Extending dosing of CC-486 beyond 7 days would increase duration of azacitidine exposure. We hypothesized that extended dosing would therefore provide more sustained epigenetic activity. Reported here are the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of CC-486 extended dosing schedules in patients with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML) or acute myeloid leukemia (AML) from part 2 of this study. PK and/or PD data were available for 59 patients who were sequentially assigned to 1 of 4 extended CC-486 dosing schedules: 300mg once-daily or 200mg twice-daily for 14 or 21 days per 28-day cycle. Both 300mg once-daily schedules and the 200mg twice-daily 21-day schedule significantly (all P < .05) reduced global DNA methylation in whole blood at all measured time points (days 15, 22, and 28 of the treatment cycle), with sustained hypomethylation at cycle end compared with baseline. CC-486 exposures and reduced DNA methylation were significantly correlated. Patients who had a hematologic response had significantly greater methylation reductions than non-responding patients. These data demonstrate that extended dosing of CC-486 sustains epigenetic effects through the treatment cycle.

Trial registration: ClinicalTrials.gov NCT00528983.

Conflict of interest statement

Competing Interests: EL, TS, JH, KK, BS, and KM are employed by Celgene Corporation and own Celgene stock. GG-M has received research support from Celgene Corp, and is a consultant and speaker for Celgene. SDG receives research support from and consults for the Celgene Corporation. CRC serves on the Scientific Advisory Board for the Connect MDS/AML Disease Registry study, which is sponsored by Celgene. This study was funded by Celgene Corporation. Sheila Truten and Brian Kaiser of MC2 provided editorial support, funded by Celgene. There are no patents or marketed products to declare. This paper describes data for a product in clinical development, CC-486. This does not alter the authors' adherence to all PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Study flow-chart.
Fig 1. Study flow-chart.
Fig 2. Patient populations.
Fig 2. Patient populations.
Fig 3. Azacitidine plasma concentrations over time…
Fig 3. Azacitidine plasma concentrations over time following CC-486 administration (200 mg and 300 mg doses) on the first day (day 1) and last day (day 14/21) of cycle 1.
(A) linear and (B) semi-log scales.
Fig 4. Mean GDMS changes in cycle…
Fig 4. Mean GDMS changes in cycle 1 following CC-486 administration in extended dosing schedules.
* (*Too few samples were available to evaluate GDMS changes over time with the CC = 486 200 mg twice daily x 14-day dosing regimen).
Fig 5. Venn diagrams showing overlaps of…
Fig 5. Venn diagrams showing overlaps of significantly regulated genomic loci, by CC-486 dosing schedule.
Red numbers represent upregulated loci; green numbers represent down-regulated loci.
Fig 6. Correlation between azacitidine exposures and…
Fig 6. Correlation between azacitidine exposures and methylation changes.
(A) Azacitidine AUC at Cycle 1, Day 1 (ng*hr/mL) vs methylation change at Cycle 1, Day 15; (B) Extrapolated cumulative azacitidine AUC for Cycle 1 vs methylation change at Cycle 1, Day 22, and (C) vs methylation change at Cycle 1, Day 28 (cycle end).
Fig 7. Changes in GDMS during cycle…
Fig 7. Changes in GDMS during cycle 1 by clinical response (any CC-486 treatment cycle).
Fig 8. Changes in GDMS at cycle…
Fig 8. Changes in GDMS at cycle 2 end and cycle 3 end by clinical response (any CC-486 treatment cycle).
NR = no response; R = response. Response categories include complete remission (CR), any hematologic improvement (HI), HI-erythroid, HI-platelet, HI-neutrophil, RBC transfusion.

References

    1. Figueroa ME, Skrabanek L, Li Y, Jiemjit A, Fandy TE, Paietta E, et al. MDS and secondary AML display unique patterns and abundance of aberrant DNA methylation. Blood. 2009;114(16):3448–58. 10.1182/blood-2009-01-200519
    1. Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. NatRevDrug Discov. 2006;5(1):37–50.
    1. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. NatRevGenet. 2012;13(7):484–92.
    1. Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358(11):1148–59. 10.1056/NEJMra072067
    1. Li LH, Olin EJ, Buskirk HH, Reineke LM. Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia. Cancer Res. 1970;30(11):2760–9.
    1. Glover AB, Leyland-Jones B. Biochemistry of azacitidine: a review. Cancer treatment reports. 1987;71(10):959–64.
    1. Stresemann C, Bokelmann I, Mahlknecht U, Lyko F. Azacytidine causes complex DNA methylation responses in myeloid leukemia. MolCancer Ther. 2008;7(9):2998–3005.
    1. Robertson KD, Jones PA. DNA methylation: past, present and future directions. Carcinogenesis. 2000;21(3):461–7.
    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(11):4727–41.
    1. Santi DV, Garrett CE, Barr PJ. On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell. 1983;33(1):9–10.
    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(5):2794–800.
    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(6):1019–28. 10.1038/leu.2008.397
    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(22):5229–38. 10.1182/blood-2011-11-382226
    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(2):e9001 10.1371/journal.pone.0009001
    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 research. 1978;38(8):2458–66.
    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. ProcNatlAcadSciUSA. 1994;91(25):11797–801.
    1. Li LH, Olin EJ, Fraser TJ, Bhuyan BK. Phase specificity of 5-azacytidine against mammalian cells in tissue culture. Cancer Res. 1970;30(11):2770–5.
    1. Jones PA, Taylor SM, Wilson V. DNA modification, differentiation, and transformation. J ExpZool. 1983;228(2):287–95.
    1. Leone G, Teofili L, Voso MT, Lubbert M. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica. 2002;87(12):1324–41.
    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 ClinOncol. 2011;29(18):2521–7.
    1. Momparler RL, Goodman J. In vitro cytotoxic and biochemical effects of 5-aza-2'-deoxycytidine. Cancer Res. 1977;37(6):1636–9.
    1. Head DR, Jacobberger JW, Mosse C, Jagasia M, Dupont W, Goodman S, et al. Innovative analyses support a role for DNA damage and an aberrant cell cycle in myelodysplastic syndrome pathogenesis. Bone marrow research. 2011;2011:950934 10.1155/2011/950934
    1. Mahfouz RZ, Koh LS, Teo M, Chee CL, Toh HC, Saunthararajah Y. Gender, cytidine deaminase, and 5-aza/decitabine—response. Clin Cancer Res. 2013;19(11):3106–7. 10.1158/1078-0432.CCR-13-0872
    1. Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R. FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. Oncologist. 2005;10(3):176–82.
    1. Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117(12):2697–702. 10.1002/cncr.25774
    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(3):223–32. 10.1016/S1470-2045(09)70003-8
    1. Harris NL, Jaffe ES, Diebold J, Flandrin G, Muller-Hermelink HK, Vardiman J, et al. The World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues. Report of the Clinical Advisory Committee meeting, Airlie House, Virginia, November, 1997. Ann Oncol. 1999;10(12):1419–32.
    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. ProcNatlAcadSciUSA. 2011;108(11):4364–9.
    1. Smyth GK. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3(1). 10.2202/1544-6115.1027
    1. Kupershmidt I, Su QJ, Grewal A, Sundaresh S, Halperin I, Flynn J, et al. Ontology-based meta-analysis of global collections of high-throughput public data. PLoSOne. 2010;5(9).
    1. Cheson BD, Bennett JM, Kopecky KJ, Buchner T, Willman CL, Estey EH, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21(24):4642–9.
    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(2):419–25.
    1. Grovdal M, Khan R, Aggerholm A, Antunovic P, Astermark J, Bernell P, et al. Negative effect of DNA hypermethylation on the outcome of intensive chemotherapy in older patients with high-risk myelodysplastic syndromes and acute myeloid leukemia following myelodysplastic syndrome. Clinical cancer research: an official journal of the American Association for Cancer Research. 2007;13(23):7107–12.
    1. Aggerholm A, Holm MS, Guldberg P, Olesen LH, Hokland P. Promoter hypermethylation of p15INK4B, HIC1, CDH1, and ER is frequent in myelodysplastic syndrome and predicts poor prognosis in early-stage patients. European journal of haematology. 2006;76(1):23–32.
    1. Saunthararajah Y. Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes. ASH Education Program Book. 2013;2013(1):511–21.
    1. Fandy TE, Herman JG, Kerns P, Jiemjit A, Sugar EA, Choi SH, et al. Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood. 2009;114(13):2764–73. 10.1182/blood-2009-02-203547
    1. Marcucci G, Silverman L, Eller M, Lintz L, Beach CL. Bioavailability of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J ClinPharmacol. 2005;45(5):597–602.
    1. Rudek MA, Zhao M, He P, Hartke C, Gilbert J, Gore SD, et al. Pharmacokinetics of 5-azacitidine administered with phenylbutyrate in patients with refractory solid tumors or hematologic malignancies. J ClinOncol. 2005;23(17):3906–11.
    1. Micozzi D, Carpi FM, Pucciarelli S, Polzonetti V, Polidori P, Vilar S, et al. Human cytidine deaminase: a biochemical characterization of its naturally occurring variants. International journal of biological macromolecules. 2014;63:64–74. 10.1016/j.ijbiomac.2013.10.029
    1. Thompson PW, James IT, Wheatcroft S, Pownall R, Barnes CG. Circadian rhythm of serum cytidine deaminase in patients with rheumatoid arthritis during rest and exercise. Annals of the rheumatic diseases. 1989;48(6):502–4.
    1. Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, Carducci M, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer research. 2006;66(12):6361–9.
    1. Soriano AO, Yang H, Faderl S, Estrov Z, Giles F, Ravandi F, et al. Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood. 2007;110(7):2302–8.
    1. Bontkes HJ, Ruben JM, Alhan C, Westers TM, Ossenkoppele GJ, van de Loosdrecht AA. Azacitidine differentially affects CD4(pos) T-cell polarization in vitro and in vivo in high risk myelodysplastic syndromes. Leukemia research. 2012;36(7):921–30. 10.1016/j.leukres.2012.03.026
    1. Li H, Chiappinelli KB, Guzzetta AA, Easwaran H, Yen RW, Vatapalli R, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5(3):587–98.
    1. Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28(6):1280–8. 10.1038/leu.2013.355

Source: PubMed

3
購読する