FFCD-1004 Clinical Trial: Impact of Cytidine Deaminase Activity on Clinical Outcome in Gemcitabine-Monotherapy Treated Patients

Cindy Serdjebi, Johan Gagnière, Jérôme Desramé, Francine Fein, Rosine Guimbaud, Eric François, Thierry André, Jean-François Seitz, Carole Montérymard, Dominique Arsene, Julien Volet, Abakar Abakar-Mahamat, Thierry Lecomte, Véronique Guerin-Meyer, Jean-Louis Legoux, Gaël Deplanque, Pierre Guillet, Joseph Ciccolini, Côme Lepage, Laetitia Dahan, Cindy Serdjebi, Johan Gagnière, Jérôme Desramé, Francine Fein, Rosine Guimbaud, Eric François, Thierry André, Jean-François Seitz, Carole Montérymard, Dominique Arsene, Julien Volet, Abakar Abakar-Mahamat, Thierry Lecomte, Véronique Guerin-Meyer, Jean-Louis Legoux, Gaël Deplanque, Pierre Guillet, Joseph Ciccolini, Côme Lepage, Laetitia Dahan

Abstract

Purpose: Because cytidine deaminase (CDA) is the key enzyme in gemcitabine metabolism, numerous studies have attempted to investigate impact of CDA status (i.e. genotype or phenotype) on clinical outcome. To date, data are still controversial because none of these studies has fully investigated genotype-phenotype CDA status, pharmacokinetics and clinical outcome relationships in gemcitabine-treated patients. Besides, most patients were treated with gemcitabine associated with other drugs, thus adding a confounding factor. We performed a multicenter prospective clinical trial in gemcitabine-treated patients which aimed at investigating the link between CDA deficiency on the occurrence of severe toxicities and on pharmacokinetics, and studying CDA genotype-phenotype relationships.

Experimental design: One hundred twenty patients with resected pancreatic adenocarcinoma eligible for adjuvant gemcitabine monotherapy were enrolled in this study promoted and managed by the Fédération Francophone de Cancérologie Digestive. Toxicities were graded according to National Cancer Institute's Common Terminology Criteria for Adverse Events Version 4. They were considered severe for grade ≥ 3, and early when occurring during the first eight weeks of treatment. CDA status was evaluated using a double approach: genotyping for 79A>C and functional testing. Therapeutic drug monitoring of gemcitabine and its metabolite were performed on the first course of gemcitabine.

Results: Five patients out of 120 (i.e., 4.6%) were found to be CDA deficient (i.e., CDA activity <1.3 U/mg), and only one among them experienced early severe hematological toxicity. There was no statistically significant difference in CDA activity between patients experiencing hematological severe toxicities (28.44%) and patients who tolerated the treatment (71.56%). CDA genetic analysis failed in evidencing an impact in terms of toxicities or in CDA activity. Regarding pharmacokinetics, a wide inter-individual variability has been observed in patients.

Conclusion: This study, which included only 4.6% of CDA-deficient patients, failed in identifying CDA status as a predictive marker of toxicities with gemcitabine. A lack of statistical power because of smoothing effect of CDA variability as compared with real life conditions could explain this absence of impact.

Trial registration: ClinicalTrials.gov NCT01416662.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Consort flowchart of the primary…
Fig 1. Consort flowchart of the primary objective in the FFCD-1004 clinical trial.
Fig 2. Pharmacokinetics profiles of gemcitabine (A)…
Fig 2. Pharmacokinetics profiles of gemcitabine (A) and its metabolite (B) overtime during the first round of gemcitabine administration in pancreatic adenocarcinoma cancer patients.
Patients have been sampled before infusion, at the end, and 90 and 120 minutes after ending the infusion. A wide inter-individual variability is observed, either for gemcitabine or for dFdU.
Fig 3. Boxplots of CDA activity in…
Fig 3. Boxplots of CDA activity in prospective compared to retrospective study.
CDAretro stands for CDA activity in gemcitabine-treated patients for various types of cancers in the retrospective study. The boxplot in the middle represents CDA activity in the subgroup of gemcitabine-monotherapy patients in the retrospective study. CDA activities of patients from the prospective study are represented in the boxplot on the right. Isolating gemcitabine-monotherapy treated patients leads to a smoothing effect in CDA activity variability.

References

    1. Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15(6):2403–13. Epub 1997/06/01.
    1. Jones OP, Melling JD, Ghaneh P. Adjuvant therapy in pancreatic cancer. World journal of gastroenterology: WJG. 2014;20(40):14733–46. Epub 2014/10/31. 10.3748/wjg.v20.i40.14733
    1. Kroep JR, Giaccone G, Tolis C, Voorn DA, Loves WJ, Groeningen CJ, et al. Sequence dependent effect of paclitaxel on gemcitabine metabolism in relation to cell cycle and cytotoxicity in non-small-cell lung cancer cell lines. British journal of cancer. 2000;83(8):1069–76. Epub 2000/09/20.
    1. Thota R, Pauff JM, Berlin JD. Treatment of metastatic pancreatic adenocarcinoma: a review. Oncology (Williston Park). 2014;28(1):70–4. Epub 2014/04/02.
    1. Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, et al. nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst. 2015;107(2). Epub 2015/02/02.
    1. Bouffard DY, Laliberte J, Momparler RL. Kinetic studies on 2',2'-difluorodeoxycytidine (Gemcitabine) with purified human deoxycytidine kinase and cytidine deaminase. Biochemical pharmacology. 1993;45(9):1857–61. Epub 1993/05/05.
    1. Serdjebi C, Milano G, Ciccolini J. Role of cytidine deaminase in toxicity and efficacy of nucleosidic analogs, in press. Expert Opin Drug Metab Toxicol. 2014.
    1. Ciccolini J, Dahan L, Andre N, Evrard A, Duluc M, Blesius A, et al. Cytidine deaminase residual activity in serum is a predictive marker of early severe toxicities in adults after gemcitabine-based chemotherapies. J Clin Oncol. 2010;28(1):160–5. Epub 2009/11/26. 10.1200/JCO.2009.24.4491
    1. Collins AR. Molecular epidemiology in cancer research. Molecular aspects of medicine. 1998;19(6):359–432. Epub 1999/07/01.
    1. . 2014.
    1. Baker JA, Wickremsinhe ER, Li CH, Oluyedun OA, Dantzig AH, Hall SD, et al. Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase. Drug metabolism and disposition: the biological fate of chemicals. 2013;41(3):541–5. Epub 2012/12/12.
    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. Epub 2013/11/05. 10.1016/j.ijbiomac.2013.10.029
    1. Tibaldi C, Giovannetti E, Vasile E, Mey V, Laan AC, Nannizzi S, et al. Correlation of CDA, ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small cell lung cancer patients. Clinical cancer research: an official journal of the American Association for Cancer Research. 2008;14(6):1797–803. Epub 2008/03/19.
    1. Giovannetti E, Laan AC, Vasile E, Tibaldi C, Nannizzi S, Ricciardi S, et al. Correlation between cytidine deaminase genotype and gemcitabine deamination in blood samples. Nucleosides Nucleotides Nucleic Acids. 2008;27(6):720–5. Epub 2008/07/05. 10.1080/15257770802145447
    1. Li H, Wang X. The impact of CDA A79C gene polymorphisms on the response and hematologic toxicity in gemcitabine-treated patients: A meta-analysis. The International journal of biological markers. 2014;29(3):224–32. Epub 2014/02/22.
    1. Sugiyama E, Kaniwa N, Kim SR, Kikura-Hanajiri R, Hasegawa R, Maekawa K, et al. Pharmacokinetics of gemcitabine in Japanese cancer patients: the impact of a cytidine deaminase polymorphism. J Clin Oncol. 2007;25(1):32–42. Epub 2006/12/30.
    1. Deenen MJ, Cats A, Beijnen JH, Schellens JH. Part 2: pharmacogenetic variability in drug transport and phase I anticancer drug metabolism. The oncologist. 2011;16(6):820–34. Epub 2011/06/03. 10.1634/theoncologist.2010-0259
    1. Serdjebi C, Seitz JF, Ciccolini J, Duluc M, Norguet E, Fina F, et al. Rapid deaminator status is associated with poor clinical outcome in pancreatic cancer patients treated with a gemcitabine-based regimen. Pharmacogenomics. 2013;14(9):1047–51. Epub 2013/07/11. 10.2217/pgs.13.93
    1. Zhou J, Gao S, Zhang F, Jiang B, Zhan Q, Cai F, et al. Liquid chromatography-tandem mass spectrometry method for simultaneous determination of seven commonly used anticancer drugs in human plasma. Journal of chromatography B, Analytical technologies in the biomedical and life sciences. 2012;906:1–8. Epub 2012/09/11. 10.1016/j.jchromb.2012.07.033
    1. Jiang X, Galettis P, Links M, Mitchell PL, McLachlan AJ. Population pharmacokinetics of gemcitabine and its metabolite in patients with cancer: effect of oxaliplatin and infusion rate. British journal of clinical pharmacology. 2008;65(3):326–33.
    1. . 2014
    1. Maring JG, Wachters FM, Slijfer M, Maurer JM, Boezen HM, Uges DR, et al. Pharmacokinetics of gemcitabine in non-small-cell lung cancer patients: impact of the 79A>C cytidine deaminase polymorphism. Eur J Clin Pharmacol. 2010;66(6):611–7. Epub 2010/03/10. 10.1007/s00228-010-0799-0
    1. Ludovini V, Floriani I, Pistola L, Minotti V, Meacci M, Chiari R, et al. Association of cytidine deaminase and xeroderma pigmentosum group D polymorphisms with response, toxicity, and survival in cisplatin/gemcitabine-treated advanced non-small cell lung cancer patients. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer. 2011;6(12):2018–26. Epub 2011/11/05.
    1. Fitzgerald SM, Goyal RK, Osborne WR, Roy JD, Wilson JW, Ferrell RE. Identification of functional single nucleotide polymorphism haplotypes in the cytidine deaminase promoter. Hum Genet. 2006;119(3):276–83. Epub 2006/02/01.
    1. Sugiyama E, Kaniwa N, Kim SR, Hasegawa R, Saito Y, Ueno H, et al. Population pharmacokinetics of gemcitabine and its metabolite in Japanese cancer patients: impact of genetic polymorphisms. Clin Pharmacokinet. 2010;49(8):549–58. Epub 2010/07/09. 10.2165/11532970-000000000-00000

Source: PubMed

3
購読する