Log in

Genotoxicity kinetics in murine normoblasts as an approach for the in vivo action of difluorodeoxycytidine

  • Original Article
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

This study analyzed the kinetics of in vivo micronucleus induction in normoblasts by determining the kinetics of difluorodeoxycytidine (dFdC)-induced micronucleated polychromatic erythrocytes (MN-PCEs) in the peripheral blood of mice. The kinetic indexes of MN-PCE induction of dFdC were correlated with the previously reported mechanisms DNA damage induction by this compound. In general, this study aimed to establish an in vivo approach for discerning the processes underlying micronucleus induction by antineoplastic agents or mutagens in general.

Methods

The frequencies of PCEs and MN-PCEs in the peripheral blood of mice were determined prior to treatment and after treatment using dFdC at doses of 95, 190, or 380 µmol/kg at 8 h intervals throughout a 72 h post-treatment.

Results

The area beneath the curve (ABC) for MN-PCE induction as a function of time, which is an index of the total effect, indicated that the dose response was directly proportional and that the effect of dFdC on micronucleus induction was reduced compared with that of aneuploidogens and monofunctional and bifunctional alkylating agents but increased compared with that of promutagens, which is consistent with our previous results. The ABC showed a single peak with a small broadness index, which indicates that dFdC has a single mechanism or concomitant mechanisms for inducing DNA breaks. The time of the relative maximal induction (T rmi) indicated that dFdC requires more time to achieve MN-PCE induction compared with aneugens and monofunctional and bifunctional alkylating agents, although it requires a similar time to achieve MN-PCE induction as azacytidine, which is consistent with evidence showing that both agents must be incorporated into DNA for their action to be realized. The timing of maximal cytotoxicity observed with the lowest dFdC dose was correlated with the timing of the main genotoxic effect. However, early and late cytotoxic effects were detected, and these effects were independent of the genotoxic response.

Conclusions

A correlation analysis indicated that dFdC appears to induce MN-PCEs through only one mechanism or mechanisms that occur concomitantly, which could be explained by the previously reported concurrent inhibitory effects of dFdC on DNA polymerase alpha, polymerase epsilon, and/or topoisomerase. The timing of maximal cytotoxicity was correlated with the timing of maximal genotoxicity; however, an early cytotoxic effect that appeared to occur prior to the incorporation of dFdC into DNA was likely related to a previously reported inhibitory effect of dFdC on thymidylate synthase and/or ribonucleotide reductase.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price includes VAT (Germany)

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Serdjebi C, Milano G, Ciccolini J (2015) Role of cytidine deaminase in toxicity and efficacy of nucleosidic analogs. Expert Opin Drug Met Toxicol 11:665–672.

  2. Lawley PD, Phillips DH (1996) DNA adducts from chemotherapeutic agents. Mutation Res 355:13–40.

    Article  PubMed  Google Scholar 

  3. Parker PW (2009) Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer. Chem Rev 109:2880–2893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Crespan E, Garbelli A, Amoroso A, Maga G (2011) Exploiting the nucleotide substrate specificity of repair DNA polymerases to develop novel anticancer agents. Molecules 16:7994–8019

    Article  CAS  PubMed  Google Scholar 

  5. Li KK, Li F, Li QS, Yang K, ** B (2013) DNA methylation as a target of epigenetic therapeutics in cancer. Anticancer Agents Med Chem 13:242–247

    Article  CAS  PubMed  Google Scholar 

  6. Ferrara R, Pilotto S, Peretti U, Caccese M, Kinspergher S, Carbognin L, Karachaliou N, Rosell R, Tortora G, Bria E (2016) Tubulin inhibitors in non-small cell lung cancer: looking back and forward. Expert Opin Pharmacother 17:1113–1129

    Article  CAS  PubMed  Google Scholar 

  7. Lee JJ, Huang J, England CG, McNally LR, Hermann B, Frieboes HB (2013) Predictive modeling of in vivo response to gemcitabine in pancreatic cancer. PLoS One 9:e1003231.

    CAS  Google Scholar 

  8. Morales-Ramírez P, Vallarino-Kelly T, Anguiano-Orozco G, Rodríguez-Reyes R (1997) Pharmacokinetic parameters of genotoxic activity inferred by the comparison of the kinetics of MN-PCE induced by chemical agents and ionizing radiation. Mutation Res 391:127–134.

    Article  PubMed  Google Scholar 

  9. Morales-Ramírez P, Vallarino-Kelly T, Cruz-Vallejo V (2014) Kinetics of micronucleus induction and cytotoxicity caused by distinct antineoplastics and alkylating agents in vivo. Toxicol Lett 224:319–325

    Article  PubMed  Google Scholar 

  10. Armstrong MJ, Galloway SM (1997) Mismatch repair provokes chromosome aberrations in hamster cells treated with methylating agents or 6-thioguanine, but not with ethylating agents. Mutation Res 373:167–178.

    Article  CAS  PubMed  Google Scholar 

  11. Aydemir N, Bilaloglu R (2003) Genotoxicity of two anticancer drugs, gemcitabine and topotecan, in mouse bone marrow in vivo. Mutation Res 537:43–51.

    Article  CAS  PubMed  Google Scholar 

  12. Classen J, Paulsen F, Hehr T, Bamberg M, Budach W (2002) Effect of gemcitabine on acute and late radiation toxicity of skin and underlying soft tissues to single-dose irradiation in a nude mice model. Int J Radiat Oncol Biol Phys 53:197–205

    Article  CAS  PubMed  Google Scholar 

  13. Grégoire V, Beauduin M, Rosier JF, De Coster B, Bruniaux M, Octave-Prignot M, Scalliet P (1997) Kinetics of mouse jejunum radiosensitization by 2′,2′-difluorodeoxycytidine (gemcitabine) and its relationship with pharmacodynamics of DNA synthesis inhibition and cell cycle redistribution in crypt cells. Br J Cancer 76:1315–1321

    Article  PubMed  Google Scholar 

  14. Viveka S, Udyavar A, Shetty B, Kuriakose S, Sudha MJ (2015) Histomorphometric effects of gemcitabine on Swiss albino mice spermatogenesis. Adv Biomed Res 4:29–34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schmid W (1975) The micronucleus. Mutation Res 31:9–15.

    Article  CAS  PubMed  Google Scholar 

  16. Morales-Ramírez P, Vallarino-Kelly T (1998) Pharmacokinetic parameters determined from the clastogenic activity of ethylnitrosourea (ENU) and dimethylnitrosamine (DMN) in mice in vivo. Mutation Res 412:315–322.

    Article  PubMed  Google Scholar 

  17. Erlichman C (1987) The pharmacology of anticancer drugs. In: Tannock IF, Hill RP (eds) The basic science of oncology. Pergamon Press, USA, pp 292–307

    Google Scholar 

  18. Braakhuis BJ, van Dongen GA, Vermorken JB, Snow GB (1991) Preclinical in vivo activity of 2,2-difluorodeoxycytidine (gemcitabine) against human head and neck cancer. Cancer Res 51:211–214

    CAS  PubMed  Google Scholar 

  19. Grunewald R, Kantarjian H, Du M, Faucher K, Tarassoff P, Plunkett W (1992) Gemcitabine in leukemia: a phase I clinical, plasma, and cellular pharmacology study. J Clin Oncol 10:406–413

    Article  CAS  PubMed  Google Scholar 

  20. Noble S, Goa KL (1997) Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lungcancer and pancreatic cancer. Drugs 54:447–472

    Article  CAS  PubMed  Google Scholar 

  21. Mackey JR, Mani RS, Seiner M, Mowles D, Young JD, Belt JA, Crawford CR, Cass CE (I998) Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 58:4349–4357

  22. Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey GB, Plunkett W (1990) Inhibition of ribonucleotide reduction in CCRF-CEM cells by 2′,2′-difluorodeoxycytidine. Mol Pharmacol 38:567–572

    CAS  PubMed  Google Scholar 

  23. Honeywell RJ, Ruiz van Haperen VW, Veerman G, Smid K, Peters GJ (2015) Inhibition of thymidylate synthase by 2′,2′-difluoro-2′-deoxycytidine (Gemcitabine) and its metabolite 2′,2′-difluoro-2′-deoxyuridine. Int J Biochem Cell Biol 60:73–81

    Article  CAS  PubMed  Google Scholar 

  24. Veltkamp SA, Pluim D, van Eijndhoven MAJ, Bolijn MJ, Ong FHG, Govindarajan R, Unadkat JD, Beijnen JH, Schellens JHM (2008) New insights into the pharmacology and cytotoxicity of gemcitabine and 2′,2′-difluorodeoxyuridine. Mol Cancer Ther 7:2415–2425

    Article  CAS  PubMed  Google Scholar 

  25. Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W (1991) Action of 2′,2′-difluorodeoxycytidine on DNA synthesis. Cancer Res 51:6110–6117

    CAS  PubMed  Google Scholar 

  26. Pourquier P, Gioffre C, Kohlhagen G, Urasaki Y, Goldwasser F, Hertel LW, Yu S, Pon RT, Gmeiner WH, Pommier Y (2002) Gemcitabine (2′,2′-difluoro-2′-deoxycytidine), an antimetabolite that poisons topoisomerase I. Clin Cancer Res 8:2499–2504

    CAS  PubMed  Google Scholar 

  27. Morales-Ramírez P, Vallarino-Kelly T, Cruz-Vallejo VL (2008) Mechanisms of DNA breaks induction in vivo by 5-azacytidine: paths of micronucleus induction by azaC. J Appl Toxicol 28:254–259

    Article  PubMed  Google Scholar 

  28. Santi DV, Norment A, Garret CE (1984) Covalent bond formation, between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine. Proc Natl Acad Sci USA 81:6993–6997

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Satoh T, Yamamoto K, Miura KF, Sofuni T (2004) Region-specific chromatin decondensation and micronucleus formation induced by 5-azacytidine in human TIG-7 cells. Cytogenet Genome Res 104:289–294

  30. Tobey RA (1972) Effects of cytosine arabinoside, daunomycin, mithramycin, azacytidine, adriamycin, and camptothecin on mammalian cell cycle traverse. Cancer Res 32:2720–2725

    CAS  PubMed  Google Scholar 

  31. Bouchard J, Momparler RL (1983) Incorporation of 5-aza-2′-deoxycytidine-5′-triphosphate into DNA. Interactions with mammalian DNA polymerase alpha and DNA methylase. Mol Pharmacol 24:109–114

    CAS  PubMed  Google Scholar 

  32. Giovannetti E, Mey V, Loni L, Nannizzi S, Barsanti G, Savarino G, Ricciardi S, Del Tacca M, Danesi R (2007) Cytotoxic activity of gemcitabine and correlation with expression profile of drug-related genes in human lymphoid cells. Pharmacol Res 55:343–349

    Article  CAS  PubMed  Google Scholar 

  33. Brenet F, Kermani P, Spektor R, Rafii S, Scandura JM (2013) TGFβ restores hematopoietic homeostasis after myelosuppressive chemotherapy. J Exp Med 210:623–639

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Auer H, Oehler R, Lindner R, Kowalski H, Sliutz G, Orel L, Kucera E, Simon MM, Glössl J (1997) Characterisation of genotoxic properties of 2′,2′-difluorodeoxycytidine. Mutation Res 393:165–173.

    Article  CAS  PubMed  Google Scholar 

  35. Donadelli M, Constanzo C, Beghelli S, Scupoli MT, Dandrea M, Bonora A, Piacentini P, Budillon A, Caraglia M, Scarpa A, Palmieri M (2007) Synergistic inhibition of pancreatic adenocarcinoma cell growth by trichostatin A and gemcitabine. Biochim Biophys Acta 1773:1095–1106.

    Article  CAS  PubMed  Google Scholar 

  36. Maehara S, Tanaka S, Shimada M, Shirabe K, Saito Y, Takahashi K, Maehara Y (2004) Selenoprotein P, as a predictor for evaluating gemcitabine resistance in human pancreatic cancer cells. Int J Cancer 112:184–189

    Article  CAS  PubMed  Google Scholar 

  37. Morales-Ramírez P, Vallarino-Kelly T, Rodríguez-Reyes R (1996) Effect of chlorophyllin on gamma ray induced micronuclei in polychromatic erythrocytes of murine peripheral blood determined by the ABC strategy. Mutation Res 367:51–56.

    Article  PubMed  Google Scholar 

  38. Perry P, Evans HJ (1975) Cytological detection of mutagen-carcinogen exposure by sister chromatid exchange. Nature 258:121–125

    Article  CAS  PubMed  Google Scholar 

  39. Abramsson-Zetterberg L, Grawé J, Zetterberg G (1995) Flow cytometric analysis of micronucleus induction in mice by internal exposure to 137Cs at very low dose rates. Int J Radiat Biol 67:29–36

    Article  CAS  PubMed  Google Scholar 

  40. Morales-Ramírez P, Vallarino-Kelly T, Cruz-Vallejo VL, López-Iturbe R, Alvaro-Delgadillo H (2004) In vivo kinetics of micronuclei induction by bifunctional alkylating antineoplastics. Mutagenesis 19:207–213

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Angel Reyes and Perfecto Aguilar for providing excellent technical assistance.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pedro Morales-Ramírez.

Ethics declarations

Animal welfare

The animals were treated and housed in accordance with the Guide for the Care and Use of Laboratory Animals, Commission on Life Sciences, Institute of Laboratory Animal Research, National Research Council (1996). The protocol was reviewed and approved by the Internal Committee of Care and Use of Laboratory Animals (CICUAL) that oversees the ethics of research involving the use and welfare of animals.

Funding

This work was supported by Project CB-507 from the Instituto Nacional de Investigaciones Nucleares (México) and Project 240116 from the Consejo Nacional de Ciencia y Tecnología (CONACYT) of México.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morales-Ramírez, P., Vallarino-Kelly, T. & Cruz-Vallejo, V. Genotoxicity kinetics in murine normoblasts as an approach for the in vivo action of difluorodeoxycytidine. Cancer Chemother Pharmacol 79, 843–853 (2017). https://doi.org/10.1007/s00280-017-3290-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-017-3290-0

Keywords

Navigation