Purpose: There are no established genetic markers for prediction of outcomes after cyclophosphamide (CP)-containing adjuvant therapy for breast cancer. In an ancillary study to a SWOG (Southwest Oncology Group) trial (S8897), we investigated functional polymorphisms in 4 genes in CP pharmacokinetic pathways in relation to hematologic toxicity and disease-free survival (DFS).

Experimental Design: Germline DNA was available from 458 women who were at high risk of relapse and was randomized to CAF (CP, intravenous doxorubicin, and 5-fluorouracil) versus CMF (CP, intravenous methotrexate, and 5-fluorouracil) ± tamoxifen, and from 874 women who had a presumed favorable prognosis and did not receive adjuvant therapy. Odds ratios for grade 3 and 4 hematologic toxicity in the treated group and hazard ratios for DFS associated with selected functional polymorphisms in CYP2B6CYP3A4GSTA1and GSTP1were estimated by logistic regression and Cox proportional hazard regression.

Results: Compared with women with AA genotypes, those with at least 1 GSTP1 variant G allele had reduced risk of grade 3 and 4 neutropenia [odds ratios (OR) = 0.63, 95% CI = 0.41–0.97] and leucopenia (OR = 0.59, 95% CI = 0.39–0.89). No other associations between single nucleotide polymorphisms and toxicity or survival were found in the treated or untreated group.

Conclusion: Known genetic variants in genes involved in CP pharmacokinetics may not have major effects on DFS in breast cancer patients. The lower risk of developing high-grade hematologic toxicity among women with variant GSTP1alleles suggests that genetic markers in combination with clinical factors may be useful in defining a subgroup of women who are less susceptible to adverse hematologic toxicities with CP-containing therapies.

Translational Relevance

In the context of a clinical trial of cyclophosphamide-containing chemotherapy for breast cancer, there were significant associations between germline GSTP1 variants and reduced risk of acute neutropenia and leucopenia after treatment. As a prophylactic measure for febrile neutropenia and infection, hematopoietic colony-stimulating factor (CSF) is commonly administered during standard chemotherapy; however, for some patients, the cost-effectiveness may not be well justified. Side effects including bone loss and bone pain may also arise from CSF use. Our findings on GSTP1 indicate that genetic variations may contribute to chemotherapy-induced neutropenia. If validated, integration of genetic factors with clinical and molecular characteristics could be used for a composite algorithm to predict risk of neutropenia prior to treatment and to facilitate clinical decision making on regimen and dosing selection of chemotherapy, as well as CSF support.

Adjuvant chemotherapy for early stage breast cancer patients has greatly improved both disease-free and overall survival by eliminating residual tumor cells after surgery (1, 2). However, because of lack of target specificity, systemically administered cytotoxic agents also cause damage to normal tissues, leading to severe and sometimes life-threatening toxicities (3). Treatment outcomes, including drug toxicities and disease-free survival (DFS), may vary greatly even among women receiving the same dose of the same drug. This is likely due, in part, to metabolic differences resulting in variability in effective drug dose delivery, as well as cell sensitivity to and defense against drug cytotoxicity.

Cyclophosphamide (CP) is a commonly used alkylating drug for breast cancer patients. There are large variations in pharmacokinetics of CP, with interindividual variability estimated at 52% in the central volume of distribution, 36% in peripheral volume of distribution, and 22% in clearance of the active metabolites (4). The variation may be partially due to patient characteristics such as weight and age but may also be due to genetic background (5). A number of genes including those encoding for phase I activation enzymes and phase II detoxification enzymes form complex pharmacokinetic networks that determine the effective dose delivered to target cells (6, 7). These key genes are highly polymorphic among individuals, and functions of some common polymorphisms have been characterized, making them excellent candidates for pharmacogenetic studies.

CYP2B6 and CYP3A4 are 2 major hepatic enzymes that convert CP prodrug to its active 4-hydroxylated metabolites. A neutral amino acid replacement in exon 4 of CYP2B6 resulting from a single nucleotide polymorphism (SNP) rs3745274 (Gln172His) is associated with increased catalytic activity in both cell cultures and human liver specimens (8, 9). For CYP3A4,SNP rs2740574 in the 5' promoter region is most commonly studied. Although it has little impact on constitutive expression (10, 11), it may result in lower enzymatic activity toward CP as shown recently in breast cancer patients (12). GSTA1 and GSTP1 are the 2 major detoxification enzymes for CP, conjugating reduced glutathione to CP metabolites. SNP rs3957356 in the 5' promoter of GSTA1gene was reported to alter binding affinity of transcription factors and thus result in lower expression (13). A nonsynonymous coding SNP rs977894 (Ile105Val) in exon 5 of GSTP1 decreases its glutathione-conjugating affinity, resulting in compromised catalytic detoxification functions (14–16).

In the Southwest Oncology Group (SWOG) S8897 trial, a clinical trial with CP-containing regimens, we conducted an ancillary pharmacogenetic study to examine 4 common SNPs of CYP2B6CYP3A4GSTA1, and GSTP1 in relation to acute hematologic toxicity and DFS after adjuvant chemotherapy.

Patient population

This correlative study was nested in a completed clinical trial led by the SWOG for The Breast Cancer Intergroup of North America (S8897, INT0102). Details regarding trial design, patient characteristics, treatment plan, and outcomes were published previously (17–20). Briefly, women with stage T1 to T3a node-negative invasive breast cancer were eligible to participate. On the basis of tumor prognostic characteristics, including tumor size and hormone (estrogen and progesterone) receptor status, they were assigned to groups depending upon risk of recurrence (high risk, low risk, or indeterminate risk). Those in the indeterminate-risk group were reassigned to either the high- or low-risk group, based on S-phase fraction of their primary cancers as determined by flow cytometry. Women in the high-risk group (either initially or after flow cytometry) were randomly assigned to an anthracycline-based regimen of 6 cycles of oral CP, intravenous doxorubicin, 5-fluorouracil (CAF; ref. 21), or a nonanthracycline-containing regimen of 6 cycles of oral CP, intravenous methotrexate, and 5-fluorouracil (CMF; ref. 22). After chemotherapy, patients in the treated group were subsequently randomized to tamoxifen for 5 years or not. Women in the low-risk group served as an observational cohort and did not receive adjuvant chemotherapy or endocrine therapy. Prophylactic hematopoietic colony-stimulating factors (CSF) were not administered at the time of the study.

Between 1989 and 1993, a total of 3,965 eligible patients were registered onto the study. Of these, 2,690 patients were assigned to the high-risk group (either initially or after flow cytometry) and received adjuvant therapy and 1,206 were assigned to the low-risk group and followed without adjuvant therapy, as shown in Figure 1. An additional 69 patients were not classifiable as at either low or high risk and were excluded. Patient characteristics of age, race, and type of primary surgery were similar among all treatment groups. After a median follow-up time of 10.8 years, the study showed slightly better overall survival but not DFS in the CAF arm than in the CMF arm, at an expense of higher toxicity (17).

Fig. 1.

Schema of S8897 trial design and tissues available for genotyping. TAM, tamoxifen.

Fig. 1.

Schema of S8897 trial design and tissues available for genotyping. TAM, tamoxifen.

Close modal

For the present ancillary pharmacogenetic study, only women with archived, uninvolved lymph node tissue available for DNA extraction in the SWOG tissue bank were included. This group consisted of women assigned to the low-risk group (initially and after flow cytometry, n = 1,206) and women initially in the indeterminate-risk group who were subsequently reassigned to the high-risk group (n = 527). Because all women with hormone receptor–negative breast cancer were classified as at high risk initially, nodal tissue was not collected. Therefore, this analysis is restricted to women who were estrogen receptor positive and/or progesterone receptor positive. The study also predates routine testing of HER2/neu status, so HER2/neu status is also unknown. Written consent was obtained from patients for use of their tissues for ancillary research studies, and this study was approved by the Institutional Review Board at Roswell Park Cancer Institute. The number of specimens collected and the number of patients for whom DNA was available in each of the cohorts are included in Figure 1, according to REMARK criteria (23). These specimens have been used for previously reported studies of the association of germline variations of other genes and outcomes (18–20).

Biospecimens, DNA, and genotyping

Two 5-μm paraffin-embedded, formalin-fixed slides of normal lymph node tissue procured from the SWOG tissue bank in San Antonio were deparaffinized and removed from slides. DNA was extracted with Qiagen DNeasy Tissue Kit (Qiagen Inc.) as previously described (24), using a modified method of Goelz et al. (25). Adequate DNA for genotyping was obtained from 458 patients in the high-risk group who received adjuvant therapy and 874 patients in the low-risk group who were followed without adjuvant therapy (Fig. 1).

Potentially functional SNPs in the 4 selected key genes involved in activation and detoxification of CP, including rs3745274 in CYP2B6, rs2740574 in CYP3A4, rs3957356 in GSTA1, and rs1695 in GSTP1, were genotyped by Sequenom matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry. Successful genotyping call rates varied between 88.4% and 99.6%, related to adequacy of DNA, with 100% concordant rates between 5% repetitive samples blindly included in the analyses.

Statistical analysis

Statistical analyses were prospectively planned in a formal, written protocol and conducted by the SWOG statistical center. For the analysis, we separated treated and untreated groups. Primary endpoints assessed in the study were high-grade hematologic toxicity and DFS. Per the protocol, DFS was defined as the time from the date of registration to the date of a second breast primary tumor, first recurrence, or death due to any cause, but not a new non–breast primary cancer (26). S8897 was conducted prior to the development of the NCI (National Cancer Institute) Common Toxicity scales and therefore standard SWOG toxicity criteria applied per protocol at the time were used (27). In brief, grade 2 to 4 neutropenia was defined by a neutrophil count of 1,000–1,500, 500–1,000, and <500/mm3, respectively; and grade 2 to 4 leucopenia was defined by a leukocyte count of 2,000–3,000, 1,000–2,000, and <1,000/mm3, respectively. The highest grade toxicity that occurred during the treatment was recorded and used in this analysis. Prevalence of acute toxicities within 1 year of registration by treatment group in the trial has been previously reported (17) and was similar among the subset of patients included in this ancillary study. Hematologic toxicities, mainly neutropenia and leucopenia, were the most common acute toxicities in all treatment groups; grade 4 nonhematologic toxicity, including severe cardiac toxicity, was rare (n = 2/458). Thus, primary endpoints in toxicity analysis were grade 3 and 4 neutropenia, grade 3 and 4 leucopenia, and combined grade 4 hematologic toxicity.

For toxicity analysis in women who received adjuvant therapy, chi-square test or Fisher's exact test was first done to examine genotype distribution by toxicity grade. Considering the incomplete knowledge of exact functions of the genes and the genotypes, if homozygous and heterozygous genotypes had similar frequency distributions by toxicity grade, the categories were combined under an assumption of dominant or recessive genetic models. Similarly, rare homozygous genotypes were collapsed with heterozygous genotypes for power considerations. Unconditional logistic regression was used to estimate associations between each SNP and risk of high-grade toxicity. For neutropenia and leucopenia, grade 3 and 4 toxicities were first combined and tested together and then tested in an ordinal model under an assumption that there is a linear relationship between toxicity grades. In addition to a binary outcome of grade 4 hematologic toxicity (yes vs. no), discrete count of grade 4 toxicity based on neutropenia and leucopenia (0, 1, and 2) was also used as an ordinal outcome in logistic regression models. Unadjusted odds ratios (OR) and 95% CIs were first derived and then were adjusted for age, menopausal status, chemotherapy arms (CAF vs. CMF), tamoxifen treatment, and time from surgery to randomization. Additional control for race or excluding nonwhite race from the analyses did not substantially change the results. According to the preplanned analytic protocol, for a SNP with a minor allele frequency in the range of 0.05 to 0.50, we would have at least 80% power to detect a minimum OR in the range of 0.56 to 0.64 for grade 4 toxicity. Four SNPs were tested in the analyses; for each SNP, 2 genetic models including dominant and ordinal models and 3 toxicity outcomes including neutropenia, leucopenia, and grade 4 hematologic toxicity were tested, yielding a total of 24 tests. To address the concern of false positivity due to multiple testing, we calculated the false-positive report possibility (FPRP), using the method by Wacholder et al. (28), assuming a prior probability for associations with toxicities of 0.1, 80% statistical power, and FPRPs below 0.5 and below 0.2 for suggestive associations and more definitive associations, respectively.

Potential modifying effects of genotypes on DFS were examined by contrasting event-free time between those carrying variant alleles and those with homozygous common alleles. Kaplan–Meier survival curves were generated and the differences were tested by log-rank test. To control for potential confounders, Cox proportion hazard models were used to adjust for age, menopausal status, and for the treated group only, also time from surgery to randomization, chemotherapy arms (CAF vs. CMF), and tamoxifen treatment.

To test interacting effects from chemotherapy regimens and tamoxifen treatment, we fitted in the regression models a productive term between treatment regimen and genotypes. The significance of the interactions was assessed by the likelihood ratio test. To test synergistic and/or additive effects among multiple SNPs, the number of “at-risk” alleles that increases CP activation or decreases detoxification was tallied (range = 0–8) and categorized into ranks. Toxicities and DFS were then compared across these ranks.

Characteristics of patients included in this ancillary study are summarized in Table 1; there were no differences between this subset of patients and those in the entire trial (17). As expected, women in the treated group tended to be younger, premenopausal, and have larger tumors than those in the untreated group who were at lower risk of recurrence. During a median 10.8 years of follow-up, there were 129 events in the treated group and 257 in the untreated group. S8897 was conducted prior to the era of CSF support. Of the patients who received adjuvant therapy, there were few reports of thrombocytopenia (2%), but 67% had grade 3 and 4 neutropenia and 62% had grade 3 and 4 leucopenia. Thirty-five percent of patients had 1 grade 4 hematologic toxicity, and 12% experienced 2 grade 4 events. Genotype frequencies are summarized in Table 2 and were similar to those in the SNP500Cancer database.

Table 1.

Characteristics of patients with adequate DNA samples in treated and untreated groups in SWOG 8897

Characteristics% treated group(n = 458)% untreated group (n = 874)
Age 
 <40 15 
 40–49 37 31 
 50–59 23 25 
 60–69 19 24 
 ≥70 12 
 Median (range), y 49 (27–85) 54 (24–89) 
Race 
 Non-Hispanic white 90 94 
 Other 10 
Menopausal status 
 Premenopausal 51 38 
 Postmenopausal 49 62 
Postmenopausal estrogen 
 Yes 14 18 
 No 86 82 
Primary treatment 
 BCS, delayed RT 18 23 
 BCS, RT prior to registration 12 12 
 Mastectomy 70 64 
Tumor size, cm 
 ≤1 50 
 1.1–1.9 100 50 
Neutropenia 
 ≤2 33 – 
 3 22 – 
 4 45 – 
Leucopenia 
 ≤2 38 – 
 3 48 – 
 4 14 – 
Thrombocytopenia 
 ≤2 98 – 
 3 – 
 4 – 
Grade 4 hematologic toxicity count 
 0 53 – 
 1 35 – 
 2 12 – 
No. of events (recurrence or death) 129 257 
Characteristics% treated group(n = 458)% untreated group (n = 874)
Age 
 <40 15 
 40–49 37 31 
 50–59 23 25 
 60–69 19 24 
 ≥70 12 
 Median (range), y 49 (27–85) 54 (24–89) 
Race 
 Non-Hispanic white 90 94 
 Other 10 
Menopausal status 
 Premenopausal 51 38 
 Postmenopausal 49 62 
Postmenopausal estrogen 
 Yes 14 18 
 No 86 82 
Primary treatment 
 BCS, delayed RT 18 23 
 BCS, RT prior to registration 12 12 
 Mastectomy 70 64 
Tumor size, cm 
 ≤1 50 
 1.1–1.9 100 50 
Neutropenia 
 ≤2 33 – 
 3 22 – 
 4 45 – 
Leucopenia 
 ≤2 38 – 
 3 48 – 
 4 14 – 
Thrombocytopenia 
 ≤2 98 – 
 3 – 
 4 – 
Grade 4 hematologic toxicity count 
 0 53 – 
 1 35 – 
 2 12 – 
No. of events (recurrence or death) 129 257 

Abbreviations: BCS, breast-conserving surgery; RT, radiation therapy.

Table 2.

Genotype frequencies of genetic polymorphisms in CP metabolism pathways in patients enrolled in SWOG 8897

Gene (SNP)Proposed function of the variant allele% treated group% untreated group
CYP2B6 (rs3745274) Increased activity N = 449 N = 858 
 GG  56 56 
 GT  35 36 
 TT  
CYP3A4 (rs2740574) Decreased activity N = 456 N = 865 
 AA  87 91 
 AG  10 
 GG  
GSTA1 (rs3957356) Decreased expression N = 414 N = 815 
 GG  40 36 
 GA  44 46 
 AA  17 18 
GSTP1 (rs1695) Decreased activity N = 405 N = 837 
 AA  47 45 
 AG  43 43 
 GG  10 12 
Gene (SNP)Proposed function of the variant allele% treated group% untreated group
CYP2B6 (rs3745274) Increased activity N = 449 N = 858 
 GG  56 56 
 GT  35 36 
 TT  
CYP3A4 (rs2740574) Decreased activity N = 456 N = 865 
 AA  87 91 
 AG  10 
 GG  
GSTA1 (rs3957356) Decreased expression N = 414 N = 815 
 GG  40 36 
 GA  44 46 
 AA  17 18 
GSTP1 (rs1695) Decreased activity N = 405 N = 837 
 AA  47 45 
 AG  43 43 
 GG  10 12 

As shown in Table 3, there were no significant associations between the polymorphisms in CYP2B6, CYP3A4, or GSTA1 and the risk of grade 4 hematologic toxicity, or with the risk of high-grade neutropenia or leucopenia (data not shown). However, there were notable associations between the GSTP1 polymorphism and reduced risk of high-grade hematologic toxicity, with similar ORs in unadjusted and adjusted models (Table 4). Compared with women homozygous for the common allele, those carrying the less active variant G allele had significantly lower risk of grade 3 and 4 neutropenia (adjusted OR = 0.63, 95% CI = 0.41–0.97, P = 0.04), and grade 3 and 4 leucopenia (adjusted OR = 0.59, 95% CI = 0.39–0.89, P = 0.01). Associations with risk of any grade 4 hematologic toxicity were of borderline significance (adjusted OR = 0.72, 95% CI = 0.48–1.07, P = 0.11). Results from the models with the ordinal outcomes (grade ≤2, 3, and 4; or count of grade 4 of hematologic toxicity 0, 1, and 2) were consistent with these models with binary outcomes. FPRP calculation indicates that the strength of the observed associations between the GSTP1 polymorphism and hematologic toxicity was between suggestive (FPRP < 0.5) to more definitive (FPRP < 0.2).

Table 3.

Estimated ORs and 95% CIs of grade 4 hematologic toxicity associated with CYP2B6, CYP3A4, and GSTA1 genotypes in SWOG 8897

GenotypeNo. with grade 4 vs. without grade 4 toxicityAdjusted OR (95% CI)aAdjusted Pa
CYP2B6 (rs3745274) 
 GG 115/135 1.00  
 TT/GT 94/105 1.05 (0.72–1.53) 0.81 
CYP3A4 (rs2740574) 
 AA 186/213 1.00  
 GG/AG 27/30 1.03 (0.59–1.82) 0.91 
GSTA1 (rs3957356) 
 GG 71/93 1.00  
 GA/AA 121/129 1.29 (0.86–1.94) 0.21 
GenotypeNo. with grade 4 vs. without grade 4 toxicityAdjusted OR (95% CI)aAdjusted Pa
CYP2B6 (rs3745274) 
 GG 115/135 1.00  
 TT/GT 94/105 1.05 (0.72–1.53) 0.81 
CYP3A4 (rs2740574) 
 AA 186/213 1.00  
 GG/AG 27/30 1.03 (0.59–1.82) 0.91 
GSTA1 (rs3957356) 
 GG 71/93 1.00  
 GA/AA 121/129 1.29 (0.86–1.94) 0.21 

aFactors adjusted in the multivariate models include age, menopausal status, chemotherapy (CAF vs. CMF), tamoxifen treatment, and time from surgery to randomization.

Table 4.

Estimated ORs and 95% CIs of high-grade hematologic toxicities associated with GSTP1 genotypes in SWOG 8897

GenotypeNo. with high-grade vs. with low-grade toxicityUnadjusted OR (95% CI)Unadjusted PAdjusted OR (95% CI)aAdjusted PaFPRPb
Neutropenia (grades 3 and 4 vs. grade ≤2) 
 AA 138/51 1.00  1.00   
 AG/GG 136/80 0.63 (0.41–0.96) 0.03 0.63 (0.41–0.97) 0.04 0.31 
 Ordinal modelc – 0.72 (0.50–1.04) 0.08 0.72 (0.50–1.05) 0.08 0.47 
Leucopenia (grades 3 and 4 vs. grade ≤2) 
 AA 130/59 1.00  1.00   
 AG/GG 123/93 0.60 (0.40–0.90) 0.01 0.59 (0.39–0.89) 0.01 0.10 
 Ordinal modelb – 0.59 (0.41–0.86) 0.006 0.58 (0.39–0.84) 0.004 0.04 
Grade 4 hematologic toxicity (yes vs. no) 
 AA 97/92 1.00  1.00   
 AG/GG 94/122 0.73 (0.49–1.08) 0.12 0.72 (0.48–1.07) 0.11 0.55 
 Ordinal modelb – 0.71 (0.49–1.04) 0.08 0.70 (0.48–1.03) 0.07 0.44 
GenotypeNo. with high-grade vs. with low-grade toxicityUnadjusted OR (95% CI)Unadjusted PAdjusted OR (95% CI)aAdjusted PaFPRPb
Neutropenia (grades 3 and 4 vs. grade ≤2) 
 AA 138/51 1.00  1.00   
 AG/GG 136/80 0.63 (0.41–0.96) 0.03 0.63 (0.41–0.97) 0.04 0.31 
 Ordinal modelc – 0.72 (0.50–1.04) 0.08 0.72 (0.50–1.05) 0.08 0.47 
Leucopenia (grades 3 and 4 vs. grade ≤2) 
 AA 130/59 1.00  1.00   
 AG/GG 123/93 0.60 (0.40–0.90) 0.01 0.59 (0.39–0.89) 0.01 0.10 
 Ordinal modelb – 0.59 (0.41–0.86) 0.006 0.58 (0.39–0.84) 0.004 0.04 
Grade 4 hematologic toxicity (yes vs. no) 
 AA 97/92 1.00  1.00   
 AG/GG 94/122 0.73 (0.49–1.08) 0.12 0.72 (0.48–1.07) 0.11 0.55 
 Ordinal modelb – 0.71 (0.49–1.04) 0.08 0.70 (0.48–1.03) 0.07 0.44 

aFactors adjusted in the multivariable models include age, menopausal status, chemotherapy (CAF vs. CMF), tamoxifen treatment, and time from surgery to randomization.

bIn ordinal models, it was assumed that there is a linear relationship between toxicity grades as well as between counts of grade 4 toxicity.

cFRRP was calculated for an assumed prior probability of associations with toxicities of 0.1. An FPRP below 0.5 was deemed as a suggestive association and an FPRP below 0.2 was deemed as a more definitive association.

Associations between each individual SNP and DFS are shown in Table 5. There was no statistically significant association of any SNP in any of the evaluated genes with DFS in women receiving or not receiving adjuvant therapy. No significant interactions between genotypes and treatment randomization (CMF vs. CAF, tamoxifen yes vs. no). In analyses to examine synergistic and/or additive effects of the 4 selected SNPs, there were also no associations of hematologic toxicities or DFS with total number of at-risk alleles (data not shown).

Table 5.

Estimated HRs and 95% CIs of DFS associated with polymorphism or CP-metabolizing genes in SWOG 8897

Adjusted HR (95% CI)
GenotypeTreated groupaUntreated groupa
CYP2B6 (rs3745274) 
 GG 1.00 1.00 
 GT 1.19 (0.82–1.73) 0.95 (0.73–1.24) 
 TT 1.14 (0.60–2.17) 1.06 (0.67–1.67) 
CYP3A4 (rs2740574) 
 AA 1.00 1.00 
 AG 0.98 (0.54–1.78) 0.69 (0.41–1.14) 
 GG 1.33 (0.49–3.64) 1.12 (0.36–3.51) 
GSTA1 (rs3957356) 
 GG 1.00 1.00 
 GA 1.02 (0.68–1.53) 1.10 (0.82–1.47) 
 AA 0.93 (0.54–1.58) 1.05 (0.72–1.54) 
GSTP1 (rs947894) 
 AA 1.00 1.00 
 AG 0.89 (0.60–1.32) 1.07 (0.82–1.40) 
 GG 1.19 (0.66–2.13) 1.11 (0.74–1.67) 
Adjusted HR (95% CI)
GenotypeTreated groupaUntreated groupa
CYP2B6 (rs3745274) 
 GG 1.00 1.00 
 GT 1.19 (0.82–1.73) 0.95 (0.73–1.24) 
 TT 1.14 (0.60–2.17) 1.06 (0.67–1.67) 
CYP3A4 (rs2740574) 
 AA 1.00 1.00 
 AG 0.98 (0.54–1.78) 0.69 (0.41–1.14) 
 GG 1.33 (0.49–3.64) 1.12 (0.36–3.51) 
GSTA1 (rs3957356) 
 GG 1.00 1.00 
 GA 1.02 (0.68–1.53) 1.10 (0.82–1.47) 
 AA 0.93 (0.54–1.58) 1.05 (0.72–1.54) 
GSTP1 (rs947894) 
 AA 1.00 1.00 
 AG 0.89 (0.60–1.32) 1.07 (0.82–1.40) 
 GG 1.19 (0.66–2.13) 1.11 (0.74–1.67) 

aFactors adjusted in the Cox proportional hazard regression models include age, menopausal status, and for treated group only, also time from surgery to randomization, chemotherapy (CAF vs. CMF), and tamoxifen treatment.

In this pharmacogenetic study, there were significant associations between a common genetic polymorphism in GSTP1 and acute hematologic toxicity but no associations with DFS. Selected polymorphisms in 3 other key genes involved in CP metabolism (CYP2B6, CYP3A4, and GSTA1) were not associated with hematologic toxicity or DFS following adjuvant therapy. Because CSFs for neutropenia prophylaxis are now commonly used in patients receiving breast cancer adjuvant chemotherapy, our findings of significant associations between the GSTP1 polymorphism and hematologic toxicity suggest that a subgroup of women may be at low risk. Thus, CSF support may not be necessary when considering the cost and potential side effects from growth factors, including bone pain and decreased bone mineral density. However, further confirmation in other data sets using archived specimens, or in a prospective trial, from patients who received CP without CSF support is required before such a strategy would be recommended

The variant G allele of GSTP1 polymorphism has been associated with increased risk of toxicity in colorectal cancer patients (29) and increased risk of neutropenia in patients with lupus erythematosus treated with CP (30). However, in women with breast cancer, this SNP was not associated with adverse drug reactions after CP-containing chemotherapy in 2 previous studies (31, 32). In a small group of 94 women receiving 6 cycles of anthracycline-based cyclophosphamide, epirubicin, and 5-fluorouracil (CEF) regimen for breast cancer, GG genotypes of GSTP1 were associated with increased risk of grade 3 and 4 hematologic toxicity (OR = 6.4, 95% CI = 1.05–39.0; ref. 33). Surprisingly, we found in our study an inverse association, which cannot be explained by direct drug detoxification function of GSTP1. However, there are other reports in the literature showing similar trends. Compared with GG genotypes, the more active AA genotype has been associated with increased risk of oxaliplatin-induced neuropathy (34), docetaxel-induced neuropathy (35), and cisplatin-induced hearing impairment (36). A possible alternative mechanism to explain these unexpected findings in drug-related toxicity is through the novel roles of GSTP1 in cellular stress response signaling as an inhibitor of c-Jun N-terminal kinase (JNK; ref. 37), a key cellular stress mediator that can activate downstream transcription factors to upregulate a number of antioxidant cellular response genes. It was speculated that less active G alleles result in increased JNK activity and elevated expression of downstream cellular stress defense genes, which may protect cells from drug cytotoxicity (34). Slower consumption of reduced glutathione by the less active GSTP1 variant may also facilitate the defense against cellular stress and may prevent cell apoptosis (38). In addition, it is possible that the functional alteration of the G allele may be drug-specific, as supported by findings in cultured cells that the “less active” GG genotype showed enhanced cisplatin detoxification compared with the AA genotype (39). Our study, along with others, calls for further drug- and tissue-specific functional characterization of this SNP.

Despite significant associations with reduced risk of toxicity, GG genotypes of GSTP1 were not associated with poorer DFS [hazard ratio (HR) = 1.19, 95% CI = 0.66–2.13] among women receiving chemotherapy (Table 5). The lack of concordance between survival and toxicity may be due to differential capacities of normal and malignant cells in dealing with drug cytotoxicity, which may be further attributed to somatic changes incurred during tumorigenesis in cancer cells. This null result, however, is unexpected on the basis of our previous findings in a smaller study, in which the HR for overall breast cancer survival was 0.3 (95% CI = 0.1–1.0) for GG genotypes compared with AA genotypes (40). Similar results were also reported in a study with Chinese breast cancer patients (41). Heterogeneity in patient characteristics and treatment regimens among those studies may partially account for the inconsistent findings.

There were no significant associations between treatment outcomes and polymorphisms in CYP2B6, CYP3A4, or GSTA1 genes. The null findings in our study could be due to redundancy in drug metabolism genes. Although we selected 4 key genes in CP metabolism pathway, a number of other genes in the same pathway may neutralize any functional changes due to the selected genetic variations.

In conclusion, in a cooperative group clinical trial for breast cancer, we found that genetic polymorphisms in key genes in CP metabolism were not associated with DFS following adjuvant chemotherapy. Of the genes evaluated, only GSTP1 polymorphism was significantly associated with risk of acute hematologic toxicity. Our results indicate that genetic variations in CP pharmacokinetic pathways may not be candidate markers for predicting treatment outcomes. Women carrying GSTP1 variants may be at lower risk of neutropenia, suggesting that genetic markers in combination with other clinical and molecular factors may be useful in predicting risk of neutropenia, and for those at low risk, prophylactic CSF may not be necessary. These results need to be further validated in independent studies before clinical recommendations can be made.

No potential conflicts of interest were disclosed.

This investigation was supported in part by the following R01 and PHS Cooperative Agreement grant numbers awarded by the National Cancer Institute, DHHS: CA095222, CA32102, CA38926, CA02599, CA13612, CA22433, CA27057, CA37981, CA46282, CA20319, CA35431, CA76447, CA45560, CA12644, CA14028, CA58416, CA04919, CA35090, CA35176, CA58686, CA58861, CA46113, CA58882, CA35128, CA74647, CA46136, CA45450, CA35261, CA35192, CA12213, CA16385, CA58658, CA46441, CA58723, CA45377, CA35119, CA42777, CA73590, CA114558–02, CA35178, and CA35262. C.B. Ambrosone, G.N. Hortobagyi, and J.M. Rae are recipients of funding from the Breast Cancer Research Foundation. S. Yao was partially supported by a Department of Defense award DAMD W81XWH-08-1-0223.

1.
Early Breast Cancer Trialists' Collaborative Group (EBCTCG)
. 
Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials
.
Lancet
2005
;
365
:
1687
717
.
2.
Eifel
P
,
Axelson
JA
,
Costa
J
, et al
National Institutes of Health Consensus Development Conference statement: adjuvant therapy for breast cancer, November 1–3, 2000
.
J Natl Cancer Inst
2001
;
93
:
979
89
.
3.
Hassett
MJ
,
O'Malley
AJ
,
Pakes
JR
,
Newhouse
JP
,
Earle
CC
. 
Frequency and cost of chemotherapy-related serious adverse effects in a population sample of women with breast cancer
.
J Natl Cancer Inst
2006
;
98
:
1108
17
.
4.
Hassan
M
,
Svensson
US
,
Ljungman
P
, et al
A mechanism-based pharmacokinetic-enzyme model for cyclophosphamide autoinduction in breast cancer patients
.
Br J Clin Pharmacol
1999
;
48
:
669
77
.
5.
Marsh
S
,
Liu
G
. 
Pharmacokinetics and pharmacogenomics in breast cancer chemotherapy
.
Adv Drug Deliv Rev
2009
;
61
:
381
7
.
6.
Choi
JY
,
Nowell
SA
,
Blanco
JG
,
Ambrosone
CB
. 
The role of genetic variability in drug metabolism pathways in breast cancer prognosis
.
Pharmacogenomics
2006
;
7
:
613
24
.
7.
Ekhart
C
,
Rodenhuis
S
,
Smits
PH
,
Beijnen
JH
,
Huitema
AD
. 
An overview of the relations between polymorphisms in drug metabolising enzymes and drug transporters and survival after cancer drug treatment
.
Cancer Treat Rev
2009
;
35
:
18
31
.
8.
Ariyoshi
N
,
Miyazaki
M
,
Toide
K
,
Sawamura
Y
,
Kamataki
T
. 
A single nucleotide polymorphism of CYP2b6 found in Japanese enhances catalytic activity by autoactivation
.
Biochem Biophys Res Commun
2001
;
281
:
1256
60
.
9.
Xie
HJ
,
Yasar
U
,
Lundgren
S
, et al
Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation
.
Pharmacogenomics J
2003
;
3
:
53
61
.
10.
Ball
SE
,
Scatina
J
,
Kao
J
, et al
Population distribution and effects on drug metabolism of a genetic variant in the 5′ promoter region of CYP3A4
.
Clin Pharmacol Ther
1999
;
66
:
288
94
.
11.
Westlind
A
,
Lofberg
L
,
Tindberg
N
,
Andersson
TB
,
Ingelman-Sundberg
M
. 
Interindividual differences in hepatic expression of CYP3A4: relationship to genetic polymorphism in the 5′-upstream regulatory region
.
Biochem Biophys Res Commun
1999
;
259
:
201
5
.
12.
Petros
WP
,
Hopkins
PJ
,
Spruill
S
, et al
Associations between drug metabolism genotype, chemotherapy pharmacokinetics, and overall survival in patients with breast cancer
.
J Clin Oncol
2005
;
23
:
6117
25
.
13.
Morel
F
,
Rauch
C
,
Coles
B
,
Le Ferrec
E
,
Guillouzo
A
. 
The human glutathione transferase alpha locus: genomic organization of the gene cluster and functional characterization of the genetic polymorphism in the hGSTA1 promoter
.
Pharmacogenetics
2002
;
12
:
277
86
.
14.
Ali-Osman
F
,
Akande
O
,
Antoun
G
,
Mao
JX
,
Buolamwini
J
. 
Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins
.
J Biol Chem
1997
;
272
:
10004
12
.
15.
Hu
X
,
Ji
X
,
Srivastava
SK
, et al
Mechanism of differential catalytic efficiency of two polymorphic forms of human glutathione S-transferase P1-1 in the glutathione conjugation of carcinogenic diol epoxide of chrysene
.
Arch Biochem Biophys
1997
;
345
:
32
8
.
16.
Sundberg
K
,
Johansson
AS
,
Stenberg
G
, et al
Differences in the catalytic efficiencies of allelic variants of glutathione transferase P1-1 towards carcinogenic diol epoxides of polycyclic aromatic hydrocarbons
.
Carcinogenesis
1998
;
19
:
433
6
.
17.
Hutchins
LF
,
Green
SJ
,
Ravdin
PM
, et al
Randomized, controlled trial of cyclophosphamide, methotrexate, and fluorouracil versus cyclophosphamide, doxorubicin, and fluorouracil with and without tamoxifen for high-risk, node-negative breast cancer: treatment results of Intergroup Protocol INT-0102
.
J Clin Oncol
2005
;
23
:
8313
21
.
18.
Ambrosone
CB
,
Barlow
WE
,
Reynolds
W
, et al
Myeloperoxidase genotypes and enhanced efficacy of chemotherapy for early-stage breast cancer in SWOG-8897
.
J Clin Oncol
2009
;
27
:
4973
9
.
19.
Choi
JY
,
Barlow
WE
,
Albain
KS
, et al
Nitric oxide synthase variants and disease-free survival among treated and untreated breast cancer patients in a Southwest Oncology Group clinical trial
.
Clin Cancer Res
2009
;
15
:
5258
66
.
20.
Yao
S
,
Barlow
WE
,
Albain
KS
, et al
Manganese superoxide dismutase polymorphism, treatment-related toxicity and disease-free survival in SWOG 8897 clinical trial for breast cancer
.
Breast Cancer Res Treat
2010;124:433–9.
21.
Bull
JM
,
Tormey
DC
,
Li
SH
, et al
A randomized comparative trial of adriamycin versus methotrexate in combination drug therapy
.
Cancer
1978
;
41
:
1649
57
.
22.
Bonadonna
G
,
Brusamolino
E
,
Valagussa
P
, et al
Combination chemotherapy as an adjuvant treatment in operable breast cancer
.
N Engl J Med
1976
;
294
:
405
10
.
23.
McShane
LM
,
Altman
DG
,
Sauerbrei
W
,
Taube
SE
,
Gion
M
,
Clark
GM
. 
REporting recommendations for tumor MARKer prognostic studies (REMARK)
.
Breast Cancer Res Treat
2006
;
100
:
229
35
.
24.
Rae
JM
,
Cordero
KE
,
Scheys
JO
,
Lippman
ME
,
Flockhart
DA
,
Johnson
MD
. 
Genotyping for polymorphic drug metabolizing enzymes from paraffin-embedded and immunohistochemically stained tumor samples
.
Pharmacogenetics
2003
;
13
:
501
7
.
25.
Goelz
SE
,
Hamilton
SR
,
Vogelstein
B
. 
Purification of DNA from formaldehyde fixed and paraffin embedded human tissue
.
Biochem Biophys Res Commun
1985
;
130
:
118
26
.
26.
Hudis
CA
,
Barlow
WE
,
Costantino
JP
, et al
Proposal for standardized definitions for efficacy end points in adjuvant breast cancer trials: the STEEP system
.
J Clin Oncol
2007
;
25
:
2127
32
.
27.
Green
S
,
Weiss
GR
. 
Southwest Oncology Group standard response criteria, endpoint definitions and toxicity criteria
.
Invest New Drugs
1992
;
10
:
239
53
.
28.
Wacholder
S
,
Chanock
S
,
Garcia-Closas
M
,
El Ghormli
L
,
Rothman
N
. 
Assessing the probability that a positive report is false: an approach for molecular epidemiology studies
.
J Natl Cancer Inst
2004
;
96
:
434
42
.
29.
Braun
MS
,
Richman
SD
,
Thompson
L
, et al
Association of molecular markers with toxicity outcomes in a randomized trial of chemotherapy for advanced colorectal cancer: the FOCUS trial
.
J Clin Oncol
2009
;
27
:
5519
28
.
30.
Zhong
S
,
Huang
M
,
Yang
X
, 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
2006
;
62
:
457
72
.
31.
Low
SK
,
Kiyotani
K
,
Mushiroda
T
,
Daigo
Y
,
Nakamura
Y
,
Zembutsu
H
. 
Association study of genetic polymorphism in ABCC4 with cyclophosphamide-induced adverse drug reactions in breast cancer patients
.
J Hum Genet
2009
;
54
:
564
71
.
32.
Ekhart
C
,
Rodenhuis
S
,
Smits
PH
,
Beijnen
JH
,
Huitema
AD
. 
Relations between polymorphisms in drug-metabolising enzymes and toxicity of chemotherapy with cyclophosphamide, thiotepa and carboplatin
.
Pharmacogenet Genomics
2008
;
18
:
1009
15
.
33.
Zarate
R
,
Gonzalez-Santigo
S
,
de la Haba
J
, et al
GSTP1 and MTHFR polymorphisms are related with toxicity in breast cancer adjuvant anthracycline-based treatment
.
Curr Drug Metab
2007
;
8
:
481
6
.
34.
Lecomte
T
,
Landi
B
,
Beaune
P
,
Laurent-Puig
P
,
Loriot
MA
. 
Glutathione S -transferase P1 polymorphism (Ile105Val) predicts cumulative neuropathy in patients receiving oxaliplatin-based chemotherapy
.
Clin Cancer Res
2006
;
12
:
3050
6
.
35.
Mir
O
,
Alexandre
J
,
Tran
A
, et al
Relationship between GSTP1 Ile(105)Val polymorphism and docetaxel-induced peripheral neuropathy: clinical evidence of a role of oxidative stress in taxane toxicity
.
Ann Oncol
2009
;
20
:
736
40
.
36.
Oldenburg
J
,
Kraggerud
SM
,
Cvancarova
M
,
Lothe
RA
,
Fossa
SD
. 
Cisplatin-induced long-term hearing impairment is associated with specific glutathione S-transferase genotypes in testicular cancer survivors
.
J Clin Oncol
2007
;
25
:
708
14
.
37.
Adler
V
,
Yin
Z
,
Fuchs
SY
, et al
Regulation of JNK signaling by GSTp
.
EMBO J
1999
;
18
:
1321
34
.
38.
Henderson
CJ
,
Wolf
CR
,
Kitteringham
N
,
Powell
H
,
Otto
D
,
Park
BK
. 
Increased resistance to acetaminophen hepatotoxicity in mice lacking glutathione S-transferase Pi
.
Proc Natl Acad Sci USA
2000
;
97
:
12741
5
.
39.
Ishimoto
TM
,
Ali-Osman
F
. 
Allelic variants of the human glutathione S-transferase P1 gene confer differential cytoprotection against anticancer agents in Escherichia coli
.
Pharmacogenetics
2002
;
12
:
543
53
.
40.
Sweeney
C
,
McClure
GY
,
Fares
MY
, et al
Association between survival after treatment for breast cancer and glutathione S-transferase P1 Ile105Val polymorphism
.
Cancer Res
2000
;
60
:
5621
4
.
41.
Yang
G
,
Shu
XO
,
Ruan
ZX
, et al
Genetic polymorphisms in glutathione-S-transferase genes (GSTM1, GSTT1, GSTP1) and survival after chemotherapy for invasive breast carcinoma
.
Cancer
2005
;
103
:
52
8
.