2,279
Views
0
CrossRef citations to date
0
Altmetric
News & Views

DRUG FOCUS: Pharmacogenetic Studies Related to Cyclophosphamide-Based Therapy

, &
Pages 1897-1903 | Published online: 03 Dec 2009

Abstract

Cyclophosphamide is a cornerstone in the treatment of many pediatric and adult malignancies, as well as in the treatment of refractory autoimmune conditions. Genetic factors are thought to play a role in the interindividual variation in both response and toxicities associated with cyclophosphamide-based therapies. This drug focus reviews the most compelling studies conducted on the pharmacogenetics of cyclophosphamide-based therapies. Broader pharmacogenomic studies are needed and may reveal additional factors important in susceptibility to toxicity and/or response to therapy.

Cyclophosphamide (CP), an oxazophosphorine, bifunctional DNA alkylating agent, is incorporated into the treatment of most pediatric and adult malignancies and was one of the first nonhormonal agents to show anti-tumor activity in humans Citation[1]. At lower doses, CP exerts a potent immunomodulatory effect and is used as second-line therapy in many autoimmune disorders Citation[1]. Like all chemotherapies, variation in the efficacy and toxicity associated with CP exist. A better understanding of the pharmacogenetic factors influencing the variation in response and toxicity to CP offers the ability to individualize treatment. To date, most, if not all, studies have taken a candidate gene approach, focusing on known targets involved in CP bioactivation and/or detoxification; however, a whole-genome approach may allow for a more comprehensive, unbiased approach to identify factors important in CP clinical activity.

Cyclophosphamide is a prodrug that enters the liver and is metabolized by the hepatic P450 system to both active and inactive compounds. N-dechloroethylation of CP, mediated primarily by CYP3A4/3A5, gives 2-dechloroethylcyclophosphamide, which is generally believed to have no cytotoxic effects, and the neurotoxic chloroacetaldehyde Citation[2]. Oxidation at the C-4 position of CP generates 4-hydroxycyclophosphamide; this reaction is mediated by various isoforms including CYP2A6, 2B6, 2C8, 2C9, 2C19, 3A4 and 3A5 Citation[2–6]. 4-hydroxycyclophosphamide interconverts with aldophosphamide, which undergoes further chemical decomposition by fragmentation to phosphoramide mustard, the active anti-tumor metabolite, and acrolein, a metabolite responsible for urotoxicity Citation[3]. Detoxification of the metabolites of CP occurs mainly through NADPH-mediated oxidation by various aldehyde dehydrogenases (ALDH1A1 and ALDH3A1) Citation[4]. Another detoxification pathway includes the conjugation of CP with glutathione by various glutathione S-transferases (GSTs; GSTA1, GSTM1, GSTP1 and GSTT1) Citation[5]. GST-mediated conjugations of various CP metabolites with glutathione have been reported, but the significance of enzyme catalysis in these reactions is unclear as they readily occur spontaneously (in the absence of enzymatic intervention) Citation[6–8]. Pharmacogenetic studies in both the ALDH and GST genes have discovered polymorphisms important to response and/or toxicity associated with CP-based therapies (see ). DNA repair proteins including MGMT and ERCC Citation[9–10], and efflux of CP and its metabolites out of the cell (MRP) Citation[11] have been shown to be important; however, the role of genetic variants within these genes has not been extensively evaluated.

Cyclophosphamide has a relatively narrow therapeutic index, and adverse effects include cardiotoxicity, nephrotoxicity, neurotoxicity, infertility, bladder toxicity, myelosuppression and leukemogenesis. Both toxicity and response to CP is quite variable. Pharmacogenetics offers clinicians the ability to individualize therapy based on a patient‘s risk of untoward effects as well as their likelihood of response. This brief review aims to highlight the clinically significant pharmacogenetic discoveries pertinent to CP-based treatment regimens.

▪ Cytochrome P450 polymorphisms & cyclophosphamide effect

Bioactivation of CP requires the activity of cytochrome P450 (CYP) enzymes in the liver. CYP is a superfamily of hemoproteins that exert their enzymatic activity on a variety of endogenous and exogenous compounds. The specific CYPs involved in CP metabolism all have variant alleles, and some have been shown to be associated with varied levels of protein expression and/or metabolic activity of the expressed proteins Citation[12–18]. To varying degrees, CYP2A6, 2B6, 2C8, 2C9, 2C19, 3A4 and 3A5 have all been implicated in the 4-hydroxylation and/or dechloroethylation of CP. To our knowledge, only selected variants in CYP2B6, 2C19, 3A4 and 3A5 have been studied for their impact on CP clinical response and/or toxicity.

CYP2B6 accounts for approximately 3–5% of total microsomal CYP protein in the liver, and is also expressed in small amounts in other tissues including intestine, kidney, brain, lung and skin Citation[19]. Isoform 2B6, which is thought to be one of the major contributors to C-4 oxidation in CP Citation[12], is highly polymorphic and extensive interindividual variation in enzymatic activity exists Citation[13]. Decreased enzymatic activity should theoretically lead to decreased active metabolites and, therefore, fewer toxic side effects and poorer response to therapy. Takada et al. demonstrated in patients receiving pulse CP for the treatment of lupus nephritis who were homozygous for CYP2B6*5, a haplotype with decreased enzymatic function, there was a higher probability of developing end-stage renal disease. This suggests that the haplotype results in a reduced exposure to the active drug Citation[14]. Rocha et al. demonstrated in 107 allogeneic stem cell transplant recipients, most of whom received busulfan and CP conditioning, that carriers of at least one mutant allele in CYP2B6*2A and CYP2B6*4 had a statistically significantly higher incidence of hemorrhagic cystitis and oral mucositis, respectively Citation[15]. The effects of these mutations on 2B6 activity have not been well described.

In vitro studies with human liver cells reveal that CYP2C19 accounts for approximately 12% of CP 4-hydroxylation Citation[12]. The variant 2C19*2 (618G>A), creates an alternate splice site and a protein lacking the ability to activate CP. Takada et al. demonstrated that carriers of at least one 2C19*2 allele were protected against premature ovarian failure and that 2C19*2 homozygotes had a higher probability of developing end-stage renal disease when treated with CP alone for their lupus nephritis Citation[14].

Involved in the metabolism of over 50% of all drugs, CYP3A4 and CYP3A5 are among the most important and most abundant CYP isozymes in humans Citation[16]. In a study of 85 women with metastatic or inflammatory breast cancer treated with standard-dose chemotherapy followed by high-dose CP, cisplatin and carmustine, patients with CYP3A4*1B or CYP3A5*1 variants had higher area under the curve values for (parent) CP at the second and third doses (suggesting reduced activation) and a decreased median survival. To date, altered protein expression has not been demonstrated as a result of these variant alleles, and the functional significance of these variants remains controversial Citation[17,18]. In fact, a recent study of 127 premenopausal women with breast cancer exposed to CP-based therapies demonstrated that young women who were carriers of the CYP3A4*1B variant haplotype had an increased risk of developing premature ovarian failure Citation[20]. These data correspond to in vitro cell-based assays that suggest CYP3A4*1B, a SNP in the promoter region of the gene, is associated with increased 3A4 expression as compared with wild-type, which should lead to increased exposure to the active drug Citation[21–22]. These findings highlight that response to therapy and risk of toxicity are likely to depend on complex interactions between multiple genetic and nongenetic factors rather than a single SNP.

Glutathione S-transferase polymorphisms & cyclophosphamide

Glutathione S-transferases are Phase II enzymes that detoxify several chemotherapeutic agents and/or their metabolites by conjugating them with glutathione. Polymorphisms in any of the GST genes produce significant alterations in the metabolism of chemotherapeutic agents and carcinogens Citation[23–28]. As a result, these polymorphisms have been implicated as having a role in susceptibility to cancer as well as resistance to treatment Citation[29–35]. The GSTs involved in CP detoxification include GSTM1, GSTT1, GSTA1 and GSTP1 Citation[36]. Reduction in the enzymatic activity of these enzymes leads to decreased detoxification of active CP and prolonged exposure to the drug. This should, in theory, result in increased risk of adverse drug effects but, also on the other hand, lead to the possibility of improved survival.

GSTA1, the primary hepatic GST, is also expressed in the human breast and is important in detoxification of CP Citation[37,38]. In 106 newly diagnosed patients with non-Hodgkin‘s lymphoma treated with CP-containing combination chemotherapy (rituximab, CP, doxorubicin, vincristine and prednisone [R-CHOP]), Rossi et al. demonstrated that patients with the GSTA1rs3957357 CT/TT genotype had significantly improved event-free survival when compared with wild-type (CC) patients. Virtually all patients with wild-type alleles and advanced disease did not respond to therapy Citation[39]. The GSTA1 rs3957357T minor allele, one of many linked SNPs comprising the GSTA1*B haplotype, is located in the promoter region of the gene and has been associated with reduced levels of GSTA1 enzyme in healthy individuals, predicting for reduced detoxification of alkylating agents and increased exposure to the active drug Citation[40,41]. Improved outcome was also observed in breast cancer patients receiving CP-based therapy who were homozygous for the GSTA1*B allele Citation[42].

Polymorphic deletions in two GSTs, GSTT1 and GSTM1, are quite common in the general population Citation[43,44]. Homozygous deletions cause no detectable enzymatic function, and have been identified as important risk factors in the development of solid tumors Citation[45]. Several studies in patients with a variety of malignancies treated with CP-containing regimens have shown that deletions in one or both of these enzymes confer improved disease-free survival, decreased risk of disease relapse and an increased incidence of chemotherapy-related side effects; all consistent with decreased detoxification of and increased exposure to the drug Citation[29,46,47].

GSTP1, which encodes for an enzyme with significant affinity for CP metabolites, is also subject to polymorphic variations; the single-nucleotide substitution A313G results in an amino acid change at codon 105 (Ile>Val) that is associated with lower substrate specific catalytic activity and lower thermal stability for the encoded protein (GSTp) Citation[26–27,48]. Several studies have demonstrated improved clinical outcomes, including improved survival and decreased rates of relapse, for both heterozygotes and homozygotes for the 105Val polymorphism in cancer patients exposed to CP in combination chemotherapy Citation[29,49–52]. Carriers of at least one 105Val polymorphism also experienced increased toxicity when exposed to CP. Patients with at least one variant allele treated with pulse CP for lupus nephritis had increased incidence of myelotoxicity and gastrointestinal toxicity in a series of studies by Zhong et al.Citation[53]. Even more concerning was a report by Allan et al., where carriers of at least one 105Val polymorphism were overrepresented in a group of patients with therapy-related acute myeloid leukemia (AML) when compared with patients with de novo AML. This association was even stronger for therapy-related AML patients exposed to chemotherapies that were GSTP1 substrates, suggesting that increased exposure to the active drug played a role in leukemogenesis. In addition, patients with therapy-related AML previously exposed only to radiotherapy did not show the same overrepresentation of 105Val carriers Citation[54].

Aldehyde dehydrogenase polymorphisms & cyclophosphamide effect

Oxidations of both active (aldophosphamide) and inactive but toxic (acrolein) metabolites of CP are mediated by aldehyde dehydrogenases, primarily ALDH1A1 and ALDH3A1 Citation[48]. Introduction of these enzymes as vectors into human and hamster ovary cells reduced their sensitivity to CP, and tumor cells resistant to CP exhibited upregulation of ALDHs Citation[55]. These enzymes are known to have genetic polymorphisms, and although the expression of ALDH1A1 and 3A1 is not affected by their most common polymorphisms (1A1*2 and 3A1*2, respectively), the functional effects of these polymorphisms have not yet been elucidated Citation[56,57].

In a series of studies by Ekhart et al., of 113 patients with various malignancies treated with high dose CP, thiotepa and carboplatin followed by autologous stem cell rescue, important toxicities were observed in patients with common variants in ALDH1A1 and 3A1. In patients heterozygous for the 1A1*2 polymorphism, an increased incidence of liver toxicity was observed compared with wild-type patients. In patients heterozygous for the 3A1*2 polymorphism, an increased incidence of hemorrhagic cystitis was observed compared with wild-type patients Citation[58]. Interestingly, ALDH1A1 is expressed in the liver but not the bladder, and 3A1 is expressed in the bladder, but not the liver Citation[57,59].

Future perspective

Cyclophosphamide is the cornerstone in the treatment of most pediatric and adult malignancies. Pharmacogenetics offers the ability to better predict response and/or toxicity based on a patient‘s genotype and, therefore, the potential to personalize therapy. Several important pharmacogenetic discoveries have been made with respect to CP‘s effect using a candidate gene approach. These findings, with a few exceptions, have also been relatively reproducible across patient populations as well as across a diverse set of diseases. However, only certain polymorphisms in important pharmacogenes have been examined, and other genes involved in CP activation, detoxification and efflux have not yet been studied for their effect. Unfortunately, most of these studies have been conducted in patients receiving combination chemotherapies with overlapping mechanisms of metabolism and detoxification, making detection of variants specific to CP effects difficult. Genes important in the pharmacokinetic and pharmacodynamic effects of CP require discovery and validation in larger and more diverse patient populations.

One consideration is the movement beyond candidate gene-driven studies into genome-wide association studies of CP in an effort to reveal, in an unbiased manner, unknown genetic variants contributing to variation in individual response and toxicity associated with CP. These whole-genome approaches should yield more robust profiles that may better guide future genetics-based treatment. Furthermore, mechanisms beyond those that act through their effect on gene expression need to be elucidated, perhaps using proteomics technologies. Future discoveries in these areas could lead to personalized therapies that enhance therapeutic response to and minimize the adverse effects of this very important and widely used chemotherapeutic drug.

Table 1. Pharmacogenetic associations of genetic polymorphisms with cyclophosphamide.

Highlights

Cyclophosphamide is used in the treatment of most pediatric and adult malignancies as well as refractory autoimmune conditions.

Genetic polymorphisms in the CYP, GST and ALDH families of enzymes have been found to impact response and/or toxicity associated with cyclophosphamide-based therapies.

Polymorphisms that reduce the ability of GSTP1 to detoxify cyclophosphamide and/or its active metabolites appear to confer superior survival outcomes as well as increased risk of toxicity, including secondary malignancies.

To date, systematic, genome-wide studies of cyclophosphamide-based therapies are lacking. Prospective, well-powered and unbiased pharmacogenomic studies are needed to verify current observations as well as to identify additional genetic markers responsible for variation in response and toxicities associated with cyclophosphamide.

Financial & competing interests disclosure

The authors of this review are supported by the Pharmacogenetics of Anticancer Agents Research (PAAR) Group (http://pharmacogenetics.org) NIH/NIGMS grant U01GM61393, University of Chicago Breast Cancer SPORE grant P50 CA125183, RO1 CA81485 and RO1 CA16783. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Additional information

Funding

The authors of this review are supported by the Pharmacogenetics of Anticancer Agents Research (PAAR) Group (http://pharmacogenetics.org) NIH/NIGMS grant U01GM61393, University of Chicago Breast Cancer SPORE grant P50 CA125183, RO1 CA81485 and RO1 CA16783. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

Bibliography

  • Colvin OM : An overview of cyclophosphamide development and clinical applications.Curr. Pharm. Des.5(8) , 555–560 (1999).
  • Ludeman SM : The chemistry of the metabolites of cyclophosphamide.Curr. Pharm. Des.5(8) , 627–643 (1999).
  • Cox PJ : Cyclophosphamide cystitis – identification of acrolein as the causative agent.Biochem. Pharmacol.28(13) , 2045–2049 (1979).
  • Parekh HK , SladekNE: NADPH-dependent enzyme-catalyzed reduction of aldophosphamide, the pivotal metabolite of cyclophosphamide.Biochem. Pharmacol.46(6) , 1043–1052 (1993).
  • Hayes JD , PulfordDJ: The glutathione S-transferase supergene family: regulation of gst and the contribution of the isoenzymes to cancer chemoprotection and drug resistance.Crit. Rev. Biochem. Mol. Biol.30(6) , 445–600 (1995).
  • Dirven HA , VenekampJC, van Ommen B, van Bladeren PJ: The interaction of glutathione with 4-hydroxycyclophosphamide and phosphoramide mustard, studied by 31p nuclear magnetic resonance spectroscopy. Chem. Biol. Interact.93(3) , 185–196 (1994).
  • Gamcsik MP , DolanME, AnderssonBS, MurrayD: Mechanisms of resistance to the toxicity of cyclophosphamide.Curr. Pharm. Des.5(8) , 587–605 (1999).
  • Shulman-Roskes EM , NoeDA, GamcsikMP et al.: The partitioning of phosphoramide mustard and its aziridinium ions among alkylation and p–n bond hydrolysis reactions.J. Med. Chem.41(4) , 515–529 (1998).
  • Cai Y , WuMH, LudemanSM, GrdinaDJ, DolanME: Role of O6-alkylguanine-DNA alkyltransferase in protecting against cyclophosphamide-induced toxicity and mutagenicity.Cancer Res.59(13) , 3059–3063 (1999).
  • Andersson BS , SadeghiT, SicilianoMJ, LegerskiR, MurrayD: Nucleotide excision repair genes as determinants of cellular sensitivity to cyclophosphamide analogs.Cancer Chemother. Pharmacol.38(5) , 406–416 (1996).
  • Qiu R , KalhornTF, SlatteryJT: ABCC2-mediated biliary transport of 4-glutathionylcyclophosphamide and its contribution to elimination of 4-hydroxycyclophosphamide in rat.J. Pharmacol. Exp. Ther.308(3) , 1204–1212 (2004).
  • Huang Z , RoyP, WaxmanDJ: Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide.Biochem. Pharmacol.59(8) , 961–972 (2000).
  • Lang T , KleinK, FischerJ et al.: Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver.Pharmacogenetics11(5) , 399–415 (2001).
  • Takada K , ArefayeneM, DestaZ et al.: Cytochrome P450 pharmacogenetics as a predictor of toxicity and clinical response to pulse cyclophosphamide in lupus nephritis.Arthritis Rheum.50(7) , 2202–2210 (2004).
  • Rocha V , PorcherR, FernandesJF et al.: Association of drug metabolism gene polymorphisms with toxicities, graft-versus-host disease and survival after hla-identical sibling hematopoietic stem cell transplantation for patients with leukemia.Leukemia23(3) , 545–556 (2009).
  • Flockhart DA , RaeJM: Cytochrome P450 3A pharmacogenetics: the road that needs traveled.Pharmacogenomics. J.3(1) , 3–5 (2003).
  • Lamba JK , LinYS, ThummelK et al.: Common allelic variants of cytochrome p4503a4 and their prevalence in different populations.Pharmacogenetics12(2) , 121–132 (2002).
  • Spurdle AB , GoodwinB, HodgsonE et al.: The CYP3A4*1B polymorphism has no functional significance and is not associated with risk of breast or ovarian cancer.Pharmacogenetics12(5) , 355–366 (2002).
  • Gervot L , RochatB, GautierJC et al.: Human CYP2B6: expression, inducibility and catalytic activities.Pharmacogenetics9(3) , 295–306 (1999).
  • Su HI , SammelMD, VeldersL et al.: Association of cyclophosphamide drug-metabolizing enzyme polymorphisms and chemotherapy-related ovarian failure in breast cancer survivors.Fertil. Steril. (2009) (Epub ahead of print).
  • Amirimani B , NingB, DeitzAC, WeberBL, KadlubarFF, RebbeckTR: Increased transcriptional activity of the CYP3A4*1B promoter variant.Environ. Mol. Mutagen.42(4) , 299–305 (2003).
  • Amirimani B , WalkerAH, WeberBL, RebbeckTR: Response: re: modification of clinical presentation of prostate tumors by a novel genetic variant in CYP3A4.J. Natl Cancer Inst.91(18) , 1588–1590 (1999).
  • Ali-Osman F , AkandeO, AntounG, MaoJX, BuolamwiniJ: Molecular cloning, characterization, and expression in Escherichia coli of full-length cdnas of three human glutathione S-transferase π gene variants. Evidence for differential catalytic activity of the encoded proteins.J. Biol. Chem.272(15) , 10004–10012 (1997).
  • Hu X , HerzogC, ZimniakP, SinghSV: Differential protection against benzo[a]pyrene-7,8-dihydrodiol-9,10-epoxide-induced DNA damage in HEPG2 cells stably transfected with allelic variants of π class human glutathione S-transferase.Cancer Res.59(10) , 2358–2362 (1999).
  • Hu X , XiaH, SrivastavaSK et al.: Activity of four allelic forms of glutathione s-transferase hgstp1–1 for diol epoxides of polycyclic aromatic hydrocarbons.Biochem. Biophys. Res. Commun.238(2) , 397–402 (1997).
  • Pandya U , SrivastavaSK, SinghalSS et al.: Activity of allelic variants of π class human glutathione s-transferase toward chlorambucil.Biochem. Biophys. Res. Commun.278(1) , 258–262 (2000).
  • Srivastava SK , SinghalSS, HuX, AwasthiYC, ZimniakP, SinghSV: Differential catalytic efficiency of allelic variants of human glutathione s-transferase π in catalyzing the glutathione conjugation of thiotepa.Arch. Biochem. Biophys.366(1) , 89–94 (1999).
  • Watson MA , StewartRK, SmithGB, MasseyTE, BellDA: Human glutathione s-transferase p1 polymorphisms: relationship to lung tissue enzyme activity and population frequency distribution.Carcinogenesis19(2) , 275–280 (1998).
  • Stanulla M , SchrappeM, BrechlinAM, ZimmermannM, WelteK: Polymorphisms within glutathione s-transferase genes (gstm1, gstt1, gstp1) and risk of relapse in childhood b-cell precursor acute lymphoblastic leukemia: a case–control study.Blood95(4) , 1222–1228 (2000).
  • Perera FP : Molecular epidemiology: insights into cancer susceptibility, risk assessment, and prevention.J. Natl Cancer Inst.88(8) , 496–509 (1996).
  • Perera FP : Molecular epidemiology: on the path to prevention?J. Natl Cancer Inst.92(8) , 602–612 (2000).
  • Wilson JF , WealeME, SmithAC et al.: Population genetic structure of variable drug response.Nat. Genet.29(3) , 265–269 (2001).
  • Tew KD : Glutathione-associated enzymes in anticancer drug resistance.Cancer Res.54(16) , 4313–4320 (1994).
  • Hall AG , AutzenP, CattanAR et al.: Expression of µ class glutathione S-transferase correlates with event-free survival in childhood acute lymphoblastic leukemia.Cancer Res.54(20) , 5251–5254 (1994).
  • Anderer G , SchrappeM, BrechlinAM et al.: Polymorphisms within glutathione s-transferase genes and initial response to glucocorticoids in childhood acute lymphoblastic leukaemia.Pharmacogenetics10(8) , 715–726 (2000).
  • Choi JY , NowellSA, BlancoJG, AmbrosoneCB: The role of genetic variability in drug metabolism pathways in breast cancer prognosis.Pharmacogenomics7(4) , 613–624 (2006).
  • Forrester LM , HayesJD, MillisR et al.: Expression of glutathione S-transferases and cytochrome p450 in normal and tumor breast tissue.Carcinogenesis11(12) , 2163–2170 (1990).
  • Dirven HA , van Ommen B, van Bladeren PJ: Involvement of human glutathione s-transferase isoenzymes in the conjugation of cyclophosphamide metabolites with glutathione. Cancer Res.54(23) , 6215–6220 (1994).
  • Rossi D , RasiS, FranceschettiS et al.: Analysis of the host pharmacogenetic background for prediction of outcome and toxicity in diffuse large b-cell lymphoma treated with R-CHOP21.Leukemia23(6) , 1118–1126 (2009).
  • Guy CA , HoogendoornB, SmithSK, ColemanS, O‘DonovanMC, BucklandPR: Promoter polymorphisms in glutathione-s-transferase genes affect transcription.Pharmacogenetics14(1) , 45–51 (2004).
  • Kusama M , KubotaT, MatsukuraY et al.: Influence of glutathione S-transferase a1 polymorphism on the pharmacokinetics of busulfan.Clin. Chim. Acta368(1–2) , 93–98 (2006).
  • Sweeney C , AmbrosoneCB, JosephL et al.: Association between a glutathione s-transferase a1 promoter polymorphism and survival after breast cancer treatment.Int. J. Cancer103(6) , 810–814 (2003).
  • Seidegard J , VorachekWR, PeroRW, PearsonWR: Hereditary differences in the expression of the human glutathione transferase active on trans-stilbene oxide are due to a gene deletion.Proc. Natl Acad. Sci. USA85(19) , 7293–7297 (1988).
  • Pemble S , SchroederKR, SpencerSR et al.: Human glutathione s-transferase theta (gstt1): cdna cloning and the characterization of a genetic polymorphism.Biochem. J.300(Pt 1) , 271–276 (1994).
  • Rebbeck TR : Molecular epidemiology of the human glutathione S-transferase genotypes gstm1 and gstt1 in cancer susceptibility.Cancer Epidemiol. Biomarkers Prev.6(9) , 733–743 (1997).
  • Hohaus S , MassiniG, D‘aloF et al.: Association between glutathione S-transferase genotypes and hodgkin‘s lymphoma risk and prognosis.Clin. Cancer Res.9(9) , 3435–3440 (2003).
  • Barahmani N , CarpentieriS, LiXN et al.: Glutathione S-transferase M1 and T1 polymorphisms may predict adverse effects after therapy in children with medulloblastoma.Neuro. Oncol.11(3) , 292–300 (2009).
  • De Jonge ME , HuitemaAD, RodenhuisS, BeijnenJH: Clinical pharmacokinetics of cyclophosphamide.Clin. Pharmacokinet.44(11) , 1135–1164 (2005).
  • Yang G , ShuXO, RuanZX et al.: Genetic polymorphisms in glutathione-S-transferase genes (gstm1, gstt1, gstp1) and survival after chemotherapy for invasive breast carcinoma.Cancer103(1) , 52–58 (2005).
  • Sweeney C , McclureGY, FaresMY et al.: Association between survival after treatment for breast cancer and glutathione S-transferase p1 Ile105Val polymorphism.Cancer Res.60(20) , 5621–5624 (2000).
  • Hohaus S , Di Ruscio A, Di Febo A et al.: Glutathione S-transferase p1 genotype and prognosis in hodgkin‘s lymphoma. Clin. Cancer Res.11(6) , 2175–2179 (2005).
  • Dasgupta RK , AdamsonPJ, DaviesFE et al.: Polymorphic variation in GSTP1 modulates outcome following therapy for multiple myeloma.Blood102(7) , 2345–2350 (2003).
  • Zhong S , HuangM, YangX et al.: Relationship of glutathione S-transferase genotypes with side-effects of pulsed cyclophosphamide therapy in patients with systemic lupus erythematosus.Br. J. Clin. Pharmacol.62(4) , 457–472 (2006).
  • Allan JM , WildCP, RollinsonS et al.: Polymorphism in glutathione S-transferase P1 is associated with susceptibility to chemotherapy-induced leukemia.Proc. Natl Acad. Sci. USA98(20) , 11592–11597 (2001).
  • Sladek NE : Aldehyde dehydrogenase-mediated cellular relative insensitivity to the oxazaphosphorines.Curr. Pharm. Des.5(8) , 607–625 (1999).
  • Spence JP , LiangT, ErikssonCJ et al.: Evaluation of aldehyde dehydrogenase 1 promoter polymorphisms identified in human populations.Alcohol Clin. Exp. Res.27(9) , 1389–1394 (2003).
  • Vasiliou V , PappaA: Polymorphisms of human aldehyde dehydrogenases. Consequences for drug metabolism and disease.Pharmacology61(3) , 192–198 (2000).
  • Ekhart C , RodenhuisS, SmitsPH, BeijnenJH, HuitemaAD: Relations between polymorphisms in drug-metabolising enzymes and toxicity of chemotherapy with cyclophosphamide, thiotepa and carboplatin.Pharmacogenet. Genomics18(11) , 1009–1015 (2008).
  • Vasiliou V , PappaA, PetersenDR: Role of aldehyde dehydrogenases in endogenous and xenobiotic metabolism.Chem. Biol. Interact.129(1–2) , 1–19 (2000).
  • Petros WP , HopkinsPJ, SpruillS et al.: Associations between drug metabolism genotype, chemotherapy pharmacokinetics, and overall survival in patients with breast cancer.J. Clin. Oncol.23(25) , 6117–6125 (2005).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.