Research paper
Cyclophosphamide: Review of its mutagenicity for an assessment of potential germ cell risks

https://doi.org/10.1016/0027-5107(95)00039-LGet rights and content

Abstract

Cyclophosphamide (CP) is used to treat a wide range of neoplastic diseases as well as some non-malignant ones such as rheumatoid arthritis. It is also used as an immunosuppressive agent prior to organ transplantation. CP is, however, a known carcinogen in humans and produces secondary tumors. There is little absorption either orally or intravenously and 10% of the drug is excreted unchanged. CP is activated by hepatic mixed function oxidases and metabolites are delivered to neoplastic cells via the bloodstream. Phosphoramide mustard is thought to be the major anti-neoplastic metabolite of CP while acrolein, which is highly toxic and is produced in equimolar amounts, is thought to be responsible for most of the toxic side effects.

DNA adducts have been formed after CP treatment in a variety of in vitro systems as well as in rats and mice using 3H-labeled CP. 32P-postlabeling techniques have also been used in mice. However, monitoring of adducts in humans has not yet been carried out. CP has also been shown to induce unscheduled DNA synthesis in a human cell line.

CP has produced mutations in base-pair substituting strains of Salmonella typhimurium in the presence of metabolic activation, but it has been shown to be negative in the E. coli chromotest. It has also been shown to be positive in Saccharomyces cerevisiae in D7 strain for many endpoints but negative in D62.M for aneuploidy/malsegregation. It has produced positive responses in Drosophila melanogaster for various endpoints and in Anopheles stephensi.

In somatic cells, CP has been shown to produce gene mutations, chromosome aberrations, micronuclei and sister chromatid exchanges in a variety of cultured cells in the presence of metabolic activation as well as sister chromatid exchanges without metabolic activation. It has also produced chromosome damage and micronuclei in rats, mice and Chinese hamsters, and gene mutations in the mouse spot test and in the transgenic lacZ construct of MutaMouse.

Increases in chromosome damage and gene mutations have been found in the peripheral blood lymphocytes of nurses, pharmacists and female workers occupationally exposured to CP during its production or distribution. Chromosome aberrations, sister chromatid exchanges and gene mutations have been observed in somatic cells of patients treated therapeutically with CP. In general, there is a maximum dose and an optimum time for the detection of genetic effects because the toxicity associated with high doses of CP will affect cell division.

In germ cells, CP has been shown to induce genetic damage in mice, rats and hamsters although the vast majority of such studies have used male mice. In males, the germ cell stages determined to be most sensitive to the induction of damage are the post-meiotic stages, which are sampled in the mouse by matings 1–3 weeks after treatment, and particularly the mid- to late-step spermatids sampled by matings 2–3 weeks after treatment. Tests by which CP has been shown to induce germ cell damage through assessment of effects in F1 progeny include those for dominant lethality, heritable translocations, specific locus mutations and malformations. Positive responses have also been reported in tests analyzing for structural chromosome damage in both spermatogonial and meiotic metaphases following exposures of various meiotic and pre-meiotic germ cell stages. CP exposures of meiotic cells have been shown to produce structural aberrations of the synaptonemal complex and to increase the frequency of spermatid micronuclei. Further, CP has been shown to induce structural, as well as some numerical, chromosome damage through analyses of second meiotic metaphases and first cleavage metaphases as well as through karyotypic analysis of F1 embryos. However, CP does not appear to be an effective aneugen. Other germ cell tests through which CP has been shown to produce genotoxic damage include those for sister chromatid exchanges and abnormal sperm head morphology. CP has also been shown to produce primary DNA damage during spermatogenesis as indicated by positive alkaline elution data from a study with rats, and by induction of unscheduled DNA synthesis in the testes of mice, rats and rabbits.

Genetic risk estimates for the male have been made for 1 mg/kg of exposure to CP under acute conditions, on the expectation that the additional risk for an individual's offspring could be obtained by multiplying the risk estimates by the total mg/kg to which an individual is exposed. The types of mutational data available following maternal exposures do not justify attempting even an approximation of the estimate of genetic risk for female germ cells. Examples are provided of how the risk estimates could be applied to practical situations based upon male exposures.

Three types of risk estimates for human health effects were derived for first generation progeny following paternal exposures. The indirect method of genetic risk estimation was used to estimate the risk from gene mutations and small deficiencies on the basis of data from the mouse morphological specific locus test. Data from the heritable translocation assay were applied to estimate risk from the induction of reciprocal translocations. In addition, an overall estimate of genetic risk was made using a modification of the direct method applied to data on F1 fetal malformations and growth retardation. These latter data are especially important because they show definitively that some mutations induced by CP actually cause serious organismic damage in first generation progeny. However, their use in estimating genetic risk warrants a special caution. Besides the unavoidable complication of making an interspecies extrapolation of phenotypic damage that is inherent in the direct method, the overall estimate of genetic risk has a further uncertainty in that the estimate of induced genetic effects manifest before the age of 25 in humans is based upon induced anomalies in the rat that are observed in late fetal stages.

The parallelogram approach was applied to our overall estimate of genetic risk to yield the estimate that, for every mg/kg of paternal exposure, there would be an increase of 600 individuals with serious induced genetic disorders by early adulthood per million liveborn offspring derived from sperm sampled within 5 weeks after treatment (i.e., for all post-meiotic stages). The available data provide no reason to expect any elevation in risk for conceptions more than 90 days after treatment if exposures are to CP alone; the risk between 5 weeks and 90 days after treatment would be relatively slight, and possibly zero. We emphasize that our risk estimates are far from precise. However, they are the best that can presently be given for hereditary damage from CP. The explanations provided for the steps needed to convert mutational information into risk estimates may lead to improvements in estimates of hereditary risk for CP and other chemicals.

There is a very extensive data base for CP which extends from the molecular level (except for adducts in humans) through the microbial and insect systems to somatic mammalian cells in various cell lines and primary cells in vitro, somatic cells in vivo, and germ cells in rodents. CP has been used as a positive control substance in many assay systems. In this review, extrapolation has been made from rodents to humans with risk estimates attempted at both individual and population levels.

References (299)

  • K.S. Bentley et al.

    Activity of germ-cell mutagens and nonmutagens in the rat spermatocyte UDS assay

    Mutation Res.

    (1988)
  • W.L. Bigbee et al.

    The effect of chemotherapy on the in vivo frequency of glycophorin A ‘null’ variant erythrocytes

    Mutation Res.

    (1990)
  • N.P. Bochkov et al.

    Cytogenetic effects of cyclophosphamide on human lymphocytes in vivo and in vitro

    Mutation Res.

    (1986)
  • J. Cannon et al.

    Cyclophosphamide-associated carcinoma of urothelium: modalities for prevention

    Urology

    (1991)
  • S. Cholerton et al.

    The role of individual cytochromes P450 in drug metabolism and clinical response

    Trends Pharmacol. Sci.

    (1992)
  • C.L. Chrisman et al.

    Cytogenetic effects of diethylstilbestrol-diphosphate (Des-dp) on mouse bone marrow monitored by the micronucleus test

    Mutation Res.

    (1979)
  • C.L. Chrisman et al.

    Micronuclei in bone-marrow cells of mice subjected to hyperthermia

    Mutation Res.

    (1980)
  • R.J. Cole et al.

    Short-term tests for transplacentally active carcinogens. A comparison of sister-chromatid exchange and the micronucleus test in mouse foetal liver crythroblasts

    Mutation Res.

    (1983)
  • P.J. Cox

    Cyclophosphamide cystitis — identification of acrolein as the causative agent

    Biochem. Pharmacol.

    (1979)
  • K. Dearfield et al.

    Evaluation of a human hepatoma cell line as a target cell in genetic toxicology

    Mutation Res.

    (1983)
  • K.L. Dearfield et al.

    In vitro assays of in vivo exposure to cyclophosphamide: Induction of sister-chromatid exchanges in peripheral lymphocytes, bone-marrow cells and in cultured cells exposed to plasma

    Mutation Res.

    (1985)
  • G.H. Deknudt et al.

    Mutagenicity tests with nickel salts in the male mouse

    Toxicol.

    (1982)
  • G. Deknudt et al.

    Influence of various mitogens on the yield of sister-chromatid exchanges, induced by chemicals, in human lymphocytes

    Mutation Res.

    (1983)
  • M. D'Incalci et al.

    Decreased half-life of cyclophosphamide in patients under continual treatment

    Eur. J. Cancer

    (1979)
  • U. Ehling et al.

    Induction of specific-locus and dominant-lethal mutations by cyclophosphamide and combined cyclophosphamide-radiation treatments

    Mutation Res.

    (1988)
  • U.H. Ehling et al.

    Dominant cataract mutations and specific locus mutations in mice induced by radiation or ethylnitrosourea

    Mutation Res.

    (1982)
  • S.S. Epstein et al.

    Detection of chemical mutagens by the dominant lethal assay in the mouse

    Toxicol. Appl. Pharmacol.

    (1972)
  • J.D. Fabricant et al.

    Postmeiotic cell mediation of behaviour in progeny of male rats treated with cyclophosphamide

    Mutation Res.

    (1983)
  • W.V. Fairchild et al.

    The incidence of bladder cancer after cyclophosphamide therapy

    J. Urol.

    (1979)
  • P.B. Farmer

    Enantiomers of cyclophosphamide amd iphosphamide

    Biochem. Pharmacol.

    (1988)
  • A.J. Francis et al.

    Tumours and malformations in the adult offspring of cyclophosphamide-treated and control male rats — Preliminary communication

    Mutation Res.

    (1990)
  • M. Furukawa et al.

    Sister chromatid exchanges induced by cyclophosphamide in V79 cells cultured in difusion chambers in mice

    Mutation Res.

    (1978)
  • M. Gad-El-Karim et al.

    Modifications in the myeloclastogenic effect of benzene in mice with toluene, phenobarbital, 3-methylcholanthrene, Aroclor 1254 and SKF-525A

    Mutation Res.

    (1984)
  • E. Gebhart et al.

    The action of anticlastogens in human lymphocyte cultures and their modification by rat-liver S9 mix

    Mutation Res.

    (1985)
  • P. Goetz et al.

    Relationship between experimental results in mammals and man. I. Cytogenetic analysis of bone marrow injury induced by a single dose of CO

    Mutation Res.

    (1975)
  • M.T. Goldberg et al.

    Comparison of the effects of 1,2-dimethylhydrazine and cyclophosphamide on micronucleus incidence in bone marrow and colon

    Mutation Res.

    (1983)
  • U. Graf et al.

    Thirty compounds tested in the Drosophila wing spot test

    Mutation Res.

    (1989)
  • M.A. Graham et al.

    Drug metabolism in carcinogenesis and cancer chemotherapy

    Pharmac. Ther.

    (1991)
  • T. Abe et al.

    Activation of cyclophosphamide using perfused rat liver system and induction of sister chromatid exchanges in vitro

    Hum Genet.

    (1981)
  • R. Albanese

    Mammalian male germ cell cytogenetics

    Mutagenesis

    (1987)
  • R. Albanese

    Induction and transmission of chemically induced chromosome aberrations in female germ cells

    Environ. Mol. Mutagen.

    (1987)
  • G. Alivizitos et al.

    Bladder cancer in a young girl with systemic lupus erythematous treated with cyclophosphamide

    Acta Urol. Belg.

    (1991)
  • J.W. Allen et al.

    In vivo BrdU-33258 Hoechst analysis of DNA replication kinetics and sister chromatid exchange formation in mouse somatic and meiotic cells

    Chromosoma

    (1976)
  • J.W. Allen et al.

    Synaptonemal complex damage as a measure of genotoxicity at meiosis

    Cell Biol. Toxicol.

    (1988)
  • M. Anderson et al.

    Vinyl chloride: dominant lethal studies in male CD-1 mice

    Mutation Res.

    (1976)
  • L.E. Appelgren et al.

    Testing of ethylene oxide for mutagenicity using the micronucleus test in mice and rats

    Acta Pharmacol. Toxicol.

    (1978)
  • F.E. Arrighi et al.

    Chromosome damage in murine and human cells following cytoxan therapy

    Tex. Rep. Biol. Med.

    (1962)
  • K. Ataya et al.

    The uptake and metabolism of cyclophosphamide by the ovary

    Select. Cancer Ther.

    (1990)
  • W.W. Au et al.

    Short-term cytogenetic assays of nine cancer chemotherapeutic drugs with metabolic activation

    Environ. Mutagen.

    (1980)
  • C.M. Bagley et al.

    Clinical pharmacology of cyclophosphamide

    Cancer Res.

    (1973)
  • Cited by (268)

    • Cytotoxic responses of the anticancer drug cyclophosphamide in the mussel Mytilus galloprovincialis and comparative sensitivity with human cells lines

      2020, Chemosphere
      Citation Excerpt :

      In humans, CP activation occurs mostly in the liver (Weber and Waxman, 1993) and the resulting metabolites are delivered to neoplastic cells via the bloodstream (Graham et al., 1991). The first step is oxidation of the oxazaphosphorine ring by cytochrome P-450 oxidases to form the unstable intermediate 4-hydroxy-CP (4-OHCP) which coexists with its tautomer, aldophosphamide (Anderson et al., 1995; Hales, 1982). This unstable transport precursor freely diffuses into the cells, where they undergo spontaneous β-deletion and give rise to acrolein and phosphoramide mustard, respectively responsible for producing high levels of oxidative stress and DNA crosslinks (Emadi et al., 2009; Tripathi etal, 2009).

    • Genotoxic potential of a novel PDE-4B inhibitor Apremilast by chromosomal aberration and micronucleus assay in mice

      2020, Saudi Pharmaceutical Journal
      Citation Excerpt :

      However, it is also known as a carcinogen and produce tumors in human (Alshahrani et al., 2019). It is also associated with chromosome aberrations, genetic mutations, sister chromatid and micronuclei exchanges (Anderson et al., 1995). In our study, cyclophosphamide at a dose of 40 mg/kg shows genotoxic effect significantly (p < 0.001) in comparison to normal control animals which indicate chromosomal aberrations were successfully induced in disease control animals.

    • Paternal safety of anti-rheumatic medications

      2020, Best Practice and Research: Clinical Obstetrics and Gynaecology
    View all citing articles on Scopus

    The authors are members of the Working Group on Cyclophosphamide assembled for the EC/Us Workshop on Risk Assessment. This Working Group wishes to extend its special appreciation to Drs. M.N. Routledge and I.R. McConnell for their assistance in preparation of the ‘Chemistry, Pharmacokinetics and Metabolism’ and ‘DNA Adducts’, and Dr. Regina Montero for assistance in preparation of the ‘Mammalian Somatic Cells’ sections of this paper.

    View full text