Abstract
Background:
In vitro data suggest that the growth of rhabdomyosarcoma (RMS) cells is suppressed in a concentration-dependent manner by 4-hydroxycyclophosphamide (4HCY), the principal precursor to the cytotoxic metabolite of cyclophosphamide (CY). Various retrospective studies on the relationship between genes encoding proteins involved in the formation and elimination of 4HCY (i.e., 4HCY pharmacokinetics) and cyclophosphamide (CY) efficacy and toxicity have been conflicting.
Procedures:
We evaluated germline pharmacogenetics in 262 patients with newly diagnosed intermediate-risk RMS who participated in one prospective Children’s Oncology Group clinical trial, ARST0531. Patients were treated with either vincristine/actinomycin/cyclophosphamide (VAC) or VAC alternating with vincristine/irinotecan (VAC/VI). We analyzed the associations between event-free survival and 394 single-nucleotide polymorphisms (SNP) in 14 drug metabolizing enzymes or transporters involved in 4HCY pharmacokinetics.
Results:
Eight SNPs were associated (p-value < .05 by univariate analysis) with 3-year event-free survival; no SNPs survived a false discovery rate < 0.05.
Conclusions:
Our data suggest that a pharmacogenomic approach to therapy personalization of cyclophosphamide in intermediate-risk rhabdomyosarcoma is not viable. Other methods to personalize therapy should be explored.
Keywords: alklylating agents, biomarkers, cyclophosphamide, pediatric cancer, pharmacogenomics, rhabdomyosarcoma
1 |. INTRODUCTION
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children and adolescents.1 The intensity of RMS therapy is tailored to the likelihood of disease recurrence. The Children’s Oncology Group (COG) defines intermediate-risk RMS to include both embryonal RMS arising at an unfavorable primary site, unresected prior to systemic therapy, and without distant metastases and alveolar RMS without distant metastases regardless of the primary site or resection status.2 For over 40 years, the North American standard therapy for intermediate-risk RMS has been the combination chemotherapy regimen of vincristine, dactinomycin, and cyclophosphamide (VAC), with radiation and/or surgery for local control.2
In vitro, the growth of RMS cells is suppressed in a concentration-dependent manner by 4-hydroxycyclophosphamide (4HCY), the principal precursor to the cytotoxic metabolite of cyclophosphamide (CY).3 Pharmacogenetic associations reported between clinical outcomes and genes expected to affect 4HCY pharmacokinetics have predominantly been conducted in adults, who often receive different CY-based combination chemotherapy regimens than children.4 The genotype–phenotype relationships may differ with CY dose and age, as we have previously reviewed.5 The contribution of the cytochrome P450 enzymes involved in 4HCY formation varies with CY concentration. The timing of when children attain the enzyme activity of adults is enzyme-specific, with hepatic CYP2B6, CYP2C9, CYP2C19, CYP3A4/5, and GST activity (normalized for bodyweight) being greater in children between the ages of 1 and 14 years.6–12 We hypothesized that genetic polymorphisms in 4HCY-pharmacokinetic enzymes are associated with event-free survival (EFS) in children with newly diagnosed intermediate-risk RMS. As a correlative study to the most recent COG phase III trial for intermediate-risk RMS (ARST0531), we evaluated germline genetic factors associated with the efficacy of VAC. The polymorphisms of interest were those associated with 4HCY formation13–21 or 4HCY elimination.22–31 In choosing the candidate genes, there are no pharmacodynamic-based gene candidates specific to RMS sensitivity to chemotherapy.
2 |. METHODS
2.1 |. Patient population selection
A full description of the selection of the patient population and treatment arms for ARST0531 (ClinicalTrials.gov identifier: NCT01222715) has been previously published.32 Briefly, participants were diagnosed with intermediate-risk RMS and subsequently randomized to receive intravenous (IV) CY (1.2 g/m2 each course) as part of the VAC regimen (cumulative CY dose, 16.8 g/m2) or the VAC regimen alternating with vincristine/irinotecan (VI) (cumulative CY dose, 8.4 g/m2) for 14 total courses of therapy over 42 weeks (Table S1).32 All participants and/or parents in this pharmacogenetic analysis consented as an optional component of the trial. Data were collected at the COG participating institutions and entered electronically within the COG electronic remote data entry system (eRDES).
2.2 |. Candidate gene selection
We analyzed the associations between EFS and the single-nucleotide polymorphisms (SNPs) in 14 drug metabolizing enzymes or transporters involved in CY metabolism (Figure S1). These genes of interest were cytochrome P450 (CYP) 2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, myeloperoxidase (MPO), aldehyde dehydrogenase (ALDH) 1A1, ALDH3A1, glutathione S-transferase (GST) A1, GSTM1, GSTP1, GSTT1, ATP-binding cassette (ABC) ABCC2, and ABCC4 (Table S2).
2.3 |. DNA genotyping
Total genomic DNA was isolated from a 10-ml whole blood sample using the Gentra Purgene DNA purification kit according to the manufacturer’s instructions (Gentra Systems, Minneapolis, MN). DNA was genotyped using Illumina’s GoldenGate genotyping assay on the VeraCode Platform (Illumina, San Diego, CA) following manufacturer’s recommendation (VeraCode Assay Guide 11312819 rev A1).33 250 nanograms of genomic DNA was aliquoted into 96-well plates, processed accordingly, and scanned on the BeadXpress reader using GenomeStudio software (v2011.1). To ensure quality control (QC) of the data, 90 samples representing 30 parent–parent–child Centre d’Étude du Polymorphisme Humain (CEPH) trios (Utah residents with ancestry from northern and western Europe)34 were genotyped to assess the performance of the Illumina oligonucleotide pool assay (OPA) before analysis of study samples. Assay accuracy was verified by comparing genotypes to publicly available genotype data for these samples from HapMap (http://www.hapmap.org/), 1000Genomes (http://www.1000genomes.org), dbSNP (http://www.ncbi.nlm.nih.gov/projects/SNP/), and by assessing inheritance errors. Two external control samples from the HapMap project were included on each plate to confirm the reliability and reproducibility of the genotyping across the study plates. Intra- and inter-plate duplicates comprised >10% of all samples. Laboratory personnel were blinded to all research information about the samples. Other QC procedures included the use of barcodes on samples and plates, dedicated materials and working space, and a visual review of SNP cluster plots by two laboratory staff members. Samples with weak signals, discordant duplicates, and outliers were repeated at least once.
Any SNPs with less than 90% concordance with public data for CEPH controls were considered failed and excluded from the analysis. Among successfully genotyped SNPs, sample and SNP call rates were >85%. All nonancestry SNPs were in Hardy–Weinberg equilibrium (p> .001). All SNPs with allele frequencies included in the analysis are given in Table S3.
2.4 |. Statistical analysis
EFS was defined as the time from study enrollment to disease progression, disease recurrence, second malignant neoplasm, or death as a result of any cause.32 There were 105 events. Cox proportional hazards models were used to estimate the hazard ratios and p-values for the associations of each SNP and EFS. All analyses were adjusted for prognostic variables for intermediate-risk RMS, including age, histology, clinical group, regional lymph node involvement, primary tumor size, and primary site).32 No adjustment was made for the treatment arm because the EFS was similar for both the VAC and VAC/VI arms on the study.32 Our analysis did not assume that particular genotypes would affect outcomes in any particular direction or by any joint effects; rather, we took an unbiased approach of testing for difference in EFS across the three genotypes (or two, in some cases) for each SNP. Variants with less than five observations were combined with the other less common variant for SNPs with three genotypes. SNPs with only two genotypes were dropped if the variant had less than five observations. For those SNPs nominally significant at p< .05, contrast was carried out to evaluate which variant had the greatest risk or protection. The Benjamini–Hochberg algorithm was used to control the false discovery rate (FDR < 0.05).35 All analyses were conducted in Stata (v16.1, StataCorp, College Station, TX).
3 |. RESULTS
3.1 |. Patient characteristics
The demographic characteristics of the entire ARST0531 study cohort have been described previously.32 The demographics of the 262 participants for which DNA was available are shown in Table 1.
TABLE 1.
Clinical characteristics of participants (n= 262)
| Clinical characteristics | VAC (n= 128) |
VAC/VI (n= 134) |
||
|---|---|---|---|---|
| Count | % | Count | % | |
| Gender | ||||
| Male | 69 | 54 | 72 | 53 |
| Female | 59 | 46 | 62 | 46 |
| Age, years | ||||
| 0–0.99 | 6 | 5 | 9 | 7 |
| 1–9.99 | 86 | 67 | 85 | 63 |
| 10+ | 36 | 28 | 40 | 30 |
| Self-reported race | ||||
| Caucasian | 91 | 71 | 103 | 77 |
| African American | 16 | 13 | 10 | 47 |
| Asian | 2 | 2 | 6 | 11 |
| Other/unknown | 19 | 15 | 15 | |
| Self-reported ethnicity | ||||
| Hispanic or Latino | 22 | 17 | 13 | 10 |
| Non-Hispanic/Latino | 102 | 80 | 116 | 87 |
| Unknown | 4 | 3 | 5 | 4 |
| Histology | ||||
| Embryonal/botryoid/spindle cell | 67 | 52 | 80 | 60 |
| Alveolar | 565 | 444 | 50 | 37 |
| Other | 4 | 3 | ||
| Clinical group | ||||
| I | 4 | 3 | 4 | 3 |
| II | 17 | 13 | 14 | 10 |
| III | 107 | 84 | 116 | 87 |
| Stage | ||||
| 1 | 15 | 12 | 10 | 7 |
| 2 | 43 | 34 | 40 | 30 |
| 3 | 70 | 55 | 84 | 63 |
| Maximum tumor size, cm | ||||
| <5 | 61 | 48 | 60 | 45 |
| 5–9.99 | 50 | 39 | 59 | 44 |
| 10+ | 17 | 13 | 15 | 11 |
| Regional lymph node status | ||||
| N0 | 103 | 80 | 103 | 77 |
| N1 | 23 | 18 | 30 | 22 |
| Nx | 2 | 2 | 1 | 1 |
| Primary site | ||||
| Orbit | 3 | 2 | 2 | 1 |
| Head or neck | 9 | 7 | 8 | 6 |
| Parameningeal | 59 | 46 | 62 | 46 |
| GU, bladder/prostate | 15 | 12 | 22 | 16 |
| GU, non-bladder/prostate | 1 | 1 | 1 | 1 |
| Extremity | 16 | 13 | 16 | 12 |
| Retroperitoneal/perineal | 15 | 12 | 17 | 13 |
| Trunk | 9 | 7 | 4 | 3 |
Note: Percentages may not total to 100 due to rounding off.
Abbreviations: GU, genitourinary; N0, no clinical or radiographic evidence of regional lymph node involvement; N1, clinical and/or radiographic evidence of regional lymph node involvement; Nx, regional lymph nodes not evaluated; RMS, NOS, rhabdomyosarcoma not otherwise specified; VAC, vincristine, dactinomycin, and cyclophosphamide; VI, vincristine and irinotecan.
3.2 |. SNP associations with event-free survival
We analyzed the associations between the EFS and the SNPs in genetic polymorphisms encoding for one of the following 14 drug metabolizing enzymes or transporters. These genes of interest were CYP2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, MPO, ALDH1A1, ALDH3A1, GSTA1, GSTM1, GSTP1, GSTT1, ABCC2, and ABCC4. Of the 394 SNPs assayed, 363 had at least two variants for evaluation. Table S4 shows the univariate hazard ratios for the risk of EFS for all SNPs tested. Eight SNPs were nominally significant at p< .05 (Table 2). However, none survived the FDR < 0.05 threshold. Five of these were variants in the ABCC4 gene. Based on power analyses of the differences in risk between genotype groups, we were underpowered to detect a significant association in this prospective trial.
TABLE 2.
SNPs associated with EFS overall at a nominal p< .05 and within genotype
| rs# | Gene | Genotypea | N b | HR | SE | p-Valuec | Linear gene dose from HR? |
|---|---|---|---|---|---|---|---|
| rs1729747 | ABCC4 | Overall | 262 | 1.56 | 0.25 | .007 | Yes |
| CC | 183 | Ref | |||||
| CG | 70 | 1.76 | 0.37 | .007 | |||
| GG | 9 | 1.85 | 0.87 | .19 | |||
| rs931111 | ABCC4 | Overall | 260 | 1.55 | 0.25 | .008 | Yes |
| TT | 182 | Ref | |||||
| TC | 69 | 1.75 | 0.37 | .008 | |||
| CC | 9 | 1.84 | 0.87 | .008 | |||
| rs1751057 | ABCC4 | Overall | 243 | 2.15 | 0.63 | .009 | No |
| AA | 222 | Ref | |||||
| AG/GG | 19/2 | 2.15 | 0.63 | .009 | |||
| rs1629441 | ABCC4 | Overall | 262 | 1.83 | 0.53 | .02 | Yes |
| CC | 237 | Ref | |||||
| TC/TT | 23/2 | 1.93 | 0.52 | .02 | |||
| rs2304925 | Intron variant | Overall | 23 | 11.94 | 12.31 | .02 | N/A |
| TT | 16 | Ref | |||||
| TG/GG | 5/2 | 11.94 | 12.31 | .02 | |||
| rs2065982 | Ancestry information marker | Overall | 262 | 0.61 | 0.14 | .03 | Yes |
| TT | 212 | Ref | |||||
| TC | 41 | 0.79 | 0.22 | .4 | |||
| CC | 9 | 0.15 | 0.15 | .06 | |||
| rs4148418 | ANKRD40 | Overall | 259 | 0.75 | 0.1 | .04 | N/A |
| AG | 111 | Ref | |||||
| AA | 96 | 0.88 | 0.19 | .55 | |||
| GG | 52 | 0.52 | 0.16 | .03 | |||
| rs9590183 | ABCC4 | Overall | 262 | 0.49 | 0.17 | .04 | N/A |
| TT | 223 | Ref | |||||
| AT/AA | 38/1 | 0.49 | 0.17 | .04 |
Abbreviations: EFS, event-free survival; HR, hazard ratio; N/A, not applicable; Ref, reference group; SE, standard error.
SNP variants with less than five patients were combined with the other variants for the genotype.
Several assays with quality control samples were used, however, there were a small number of failures.
There were no SNPs significant at FDR <0.05 (363 SNPs evaluated). A linear gene dose–response relationship, in which the heterozygotes and homozygotes for the variant allele’s change in the EFS are in a similar direction.
4 |. DISCUSSION
Among 262 patients with intermediate-risk RMS treated with VAC or VAC/VI, we did not find an association between EFS and germline SNP that encodes an enzyme involved in 4HCY formation13–21 or 4HCY elimination.22–31 Previous data from smaller studies have been conflicting. From a clinical intervention trial testing almost 400 gene variants, our analysis provides evidence that the germline pharmacogenomics of 4HCY pharmacokinetics are not informative enough to personalize treatment for patients with intermediate-risk RMS treated with CY-containing chemotherapy.
Since the 1970s, newly diagnosed RMS patients have received VAC (with or without additional agents) in conjunction with radiation therapy.36–40 Dose escalation of vincristine or dactinomycin has not been investigated due to dose-limiting neurotoxicity and hepatotoxicity, respectively.41 CY dose intensification was investigated in a pilot study but abandoned due to excessive toxicity.42 VAC is an excellent treatment model for evaluating the pharmacogenomics of CY in children because it has clearly shown age-dependent toxicity with empiric CY dosing.43 In an attempt to reduce CY toxicity, ARST0531 used a lower CY dose per course (1.2 g/m2) and cumulative CY dose (8.4 and 16.8 g/m2) than the two prior North American clinical trials (2.2 g/m2 per course and 25.1–30.8 g/m2 cumulatively).40,44 We sought to identify germline polymorphisms associated with EFS.
CY is a pro-drug with a complex metabolic schema (Figure S1), with a wide range of parent drug area under the plasma concentration–time curve (AUC) and even greater variability in the AUCs of its metabolites, such as acrolein and phosphoramide mustard.45 Of the pharmacogenetic studies in children receiving CY (references highlighted in blue in Table S2), the majority have focused upon GST polymorphisms.46–50 Pharmacogenetic studies in adults receiving conventional-dose CY also included genetic polymorphisms of the CYP enzymes relevant to 4HCY formation (Table S2).13–20 Many of these studies were limited by their small sample size and inconsistency in CY dosing, which could affect the predominant CYP enzyme involved in 4HCY formation14,51–53 and thus, genotype-clinical outcome phenotype associations. The adult data cannot, however, be directly translated to children as children have faster CY clearance than adults.45,54 Overall, children have impaired hepatic metabolism over the first 6–12 months of life, followed by a period up to adolescence when drug metabolism is more rapid relative to adults. The timing of when children attain the enzyme activity of adults is enzyme-specific, with hepatic CYP2B6, CYP2C9, CYP2C19, CYP3A4/5, and GST activity (normalized for bodyweight) being greater in children between the ages of 1 and 14 years.6–12 Data regarding the ontogeny of the other metabolizing enzymes and transporters relevant to 4HCY elimination in vivo are not available. Thus, the predominant cytochrome P450 catalyzing 4HCY formation may differ between children and adults, potentially influencing the genotype–phenotype relationship.
Furthermore, the administered CY dose also influences the extent of 4HCY formation in children (Fig. 2b of Raccor et al.),5 which qualitatively agrees with adult data that the extent of bioactivation is greater with conventional-dose CY (0.5 g/m2) compared to myeloablative-dose CY (100 mg/kg).55,56 Thus, there are potential differences in the quantitative contribution of CY-metabolizing enzymes toward 4HCY formation based on the CY dose. There is a shift in CYP contribution based on CY dose, with CYP2C9 and CYP2C19 being the major hepatic cytochrome P450 responsible for 4HCY formation at low concentrations and CYP3A4/5 and CYP2B6 assuming greater roles at high CY concentrations.14,51–53 We did consider using archived tumor samples from the prior COG study (D9803) for this pharmacogenetic study,57,58 however, this approach is suboptimal because this prior study (D9803) used a higher CY dose of 2.2 g/m2, which would have a lower ratio of 4HCY/CY AUC.
There are limitations to these data. Given the relative rarity of RMS, patient numbers for this trial were limited and likely hampered our ability to detect meaningful differences in outcome between genotypic groups. We chose pharmacokinetic-based candidate genes, which have been associated with efficacy, toxicity, or pharmacokinetics (Table S2). Many of the studies in Table S2 are from small populations of heterogeneous CY-based combination chemotherapy regimens with incomplete candidate gene lists. We sought to overcome these limitations by systematically evaluating all of these candidate genes in a homogenous population of children. We evaluated many of the candidate genes in children receiving conventional-dose CY, as only the CYP3A4, CYP3A5, and GST polymorphisms have been evaluated in children with either immunologic conditions or acute lymphoblastic leukemia.46–50 Another potential confounder is drug–drug interactions between CY and other chemotherapy or supportive care agents.59–61 Our in vitro data suggest that 4HCY formation is not influenced by concomitant chemotherapeutics, including those concomitant with CY in the VAC regimen - vincristine and dactinomycin.5 Furthermore, the inclusion/exclusion criteria for ARTS0531 recommend avoiding strong inhibitors of CYP3A4, which is the anticipated mechanism of most drug–drug interactions.
Over the past few decades, there has been substantial enthusiasm for germline pharmacogenomics improving the treatment of patients with pediatric cancers.62 The benefits of mercaptopurine dosing based on thiopurine methyltransferase genotype are clear for children with acute lymphoblastic leukemia (ALL).63 The treatment of ALL is similar to other pediatric cancers: multiagent chemotherapy given over an extended duration. Patients diagnosed with intermediate-risk RMS receive either three (VAC) or four (VAC/VI) anticancer agents. With this relatively homogenous chemotherapy regimen and the in vitro data showing that growth of RMS cells is suppressed in a concentration-dependent manner by 4HCY,3 we took a candidate gene approach based on genes regulating proteins involved in 4HCY formation and 4HCY elimination to evaluate their association with EFS. Our analysis suggests that there are no promising germline SNPs to improve the effectiveness of the VAC or VAC/VI regimen in children with intermediate-risk RMS. These negative genotype–phenotype associations indicate that germline pharmacogenetic testing is likely not a viable approach to the personalization of chemotherapy in patients with intermediate-risk RMS treated with VAC or VAC/VI. In contrast, the recent identification of somatic MYOD1 and TP53 mutations as biomarkers for inferior outcome in RMS64 provides an opportunity to incorporate a molecular feature in risk stratification of RMS, with the long-range goal of improving EFS.
Supplementary Material
ACKNOWLEDGMENTS
This work is supported in part by grants from the National Institutes of Health (U10CA180886, U10CA180899, U10CA098543, and U10CA098413; R03CA178104; R21CA162059; R01GM129863); Alex’s Lemonade Stand, Children’s Foundation, and Fraternal Order of Eagles. We appreciate the children and their families who participated in this study. We also appreciate the contributions of their health care team. We also appreciate the contributions of the research staff involved in data generation, especially Linda Risler, BS and Karen W. Makar, Ph.D.
Funding Information
National Institutes of Health, Grant Numbers: U10CA180886, U10CA180899, U10CA098543, and U10CA098413; R03CA178104; R21CA162059; R01GM129863; Alex’s Lemonade Stand; Children’s Foundation; Fraternal Order of Eagles.
Footnotes
CONFLICT OF INTEREST
The authors declare that there is no conflict of interest.
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
SUPPORTING INFORMATION
Additional supporting information may be found online in the Supporting Information section at the end of the article.
REFERENCES
- 1.Skapek SX, Ferrari A, Gupta AA, et al. Rhabdomyosarcoma. Nat Rev Dis Primers 2019;5(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hawkins DS, Spunt SL, Skapek SX, COG Soft Tissue Sarcoma Committee. Children’s Oncology Group’s 2013 blueprint for research: soft tissue sarcomas. Pediatr Blood Cancer 2013;60(6):1001–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ozkaynak MF, Nolta J, Parkman R. In vitro purging of human rhabdomyosarcoma cells using 4-hydroperoxycyclophosphamide. Cancer Res 1990;50(5):1455–1458. [PubMed] [Google Scholar]
- 4.Pinto N, Ludeman SM, Dolan ME. Drug focus: pharmacogenetic studies related to cyclophosphamide-based therapy. Pharmacogenomics 2009;10(12):1897–1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Raccor BS, Claessens AJ, Dinh JC, et al. Potential contribution of cytochrome P450 2B6 to hepatic 4-hydroxycyclophosphamide formation in vitro and in vivo. Drug Metab Dispos 2012;40(1):54–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Stempak D, Gammon J, Klein J, Koren G, Baruchel S. Single-dose and steady-state pharmacokinetics of celecoxib in children. Clin Pharmacol Ther 2002;72(5):490–497. [DOI] [PubMed] [Google Scholar]
- 7.Chiba K, Ishizaki T, Miura H, Minagawa K. Michaelis-Menten pharmacokinetics of diphenylhydantoin and application in the pediatric age patient. J Pediatr 1980;96:479–484. [DOI] [PubMed] [Google Scholar]
- 8.Takahashi H, Ishikawa S, Nomoto S, et al. Developmental changes in pharmacokinetics and pharmacodynamics of warfarin enantiomers in Japanese children. Clin Pharmacol Ther 2000;68(5):541–555. [DOI] [PubMed] [Google Scholar]
- 9.Korpela R, Olkkola KT. Pharmacokinetics of intravenous diclofenac sodium in children. Eur J Clin Pharmacol 1990;38(3):293–295. [DOI] [PubMed] [Google Scholar]
- 10.Hughes J, Gill AM, Mulhearn H, Powell E, Choonara I. Steady-state plasma concentrations of midazolam in critically ill infants and children. Ann Pharmacother 1996;30(1):27–30. [DOI] [PubMed] [Google Scholar]
- 11.Veal GJ, Hartford CM, Stewart CF. Clinical pharmacology in the adolescent oncology patient. J Clin Oncol 2010;28(32):4790–4799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.McCarver DG, Hines RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. J Pharmacol Exp Ther 2002;300(2):361–366. [DOI] [PubMed] [Google Scholar]
- 13.Takada K, Arefayene M, Desta Z, et al. Cytochrome P450 pharmacogenetics as a predictor of toxicity and clinical response to pulse cyclophosphamide in lupus nephritis. Arthritis Rheum 2004;50(7):2202–2210. [DOI] [PubMed] [Google Scholar]
- 14.Timm R, Kaiser R, Lotsch J, et al. Association of cyclophosphamide pharmacokinetics to polymorphic cytochrome P450 2C19. Pharmacogenomics J 2005;5(6):365–373. [DOI] [PubMed] [Google Scholar]
- 15.Xie H, Griskevicius L, Stahle L, et al. Pharmacogenetics of cyclophosphamide in patients with hematological malignancies. Eur J Pharm Sci 2006;27(1):54–61. [DOI] [PubMed] [Google Scholar]
- 16.Nakajima M, Komagata S, Fujiki Y, et al. Genetic polymorphisms of CYP2B6 affect the pharmacokinetics/pharmacodynamics of cyclophosphamide in Japanese cancer patients. Pharmacogenet Genomics 2007;17(6):431–445. [DOI] [PubMed] [Google Scholar]
- 17.Singh G, Saxena N, Aggarwal A, Misra R. Cytochrome P450 polymorphism as a predictor of ovarian toxicity to pulse cyclophosphamide in systemic lupus erythematosus. J Rheumatol 2007;34(4):731–733. [PubMed] [Google Scholar]
- 18.Su HI, Sammel MD, Velders L, et al. Association of cyclophosphamide drug-metabolizing enzyme polymorphisms and chemotherapy-related ovarian failure in breast cancer survivors. Fertil Steril 2010;94(2): 645–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gor PP, Su HI, Gray RJ, et al. Cyclophosphamide-metabolizing enzyme polymorphisms and survival outcomes after adjuvant chemotherapy for node-positive breast cancer: a retrospective cohort study. Breast Cancer Res 2010;12(3):R26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bray J, Sludden J, Griffin MJ, et al. Influence of pharmacogenetics on response and toxicity in breast cancer patients treated with doxorubicin and cyclophosphamide. Br J Cancer 2010;102(6):1003–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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(30):4973–4979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Allan JM, Wild CP, Rollinson S, et al. Polymorphism in glutathione S-transferase P1 is associated with susceptibility to chemotherapy-induced leukemia. Proc Natl Acad Sci U S A 2001;98(20):11592–11597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.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(20):5621–5624. [PubMed] [Google Scholar]
- 24.Sweeney C, Ambrosone CB, Joseph L, et al. Association between a glutathione S-transferase A1 promoter polymorphism and survival after breast cancer treatment. Int J Cancer 2003;103(6):810–814. [DOI] [PubMed] [Google Scholar]
- 25.Dasgupta RK, Adamson PJ, Davies FE, et al. Polymorphic variation in GSTP1 modulates outcome following therapy for multiple myeloma. Blood 2003;102(7):2345–2350. [DOI] [PubMed] [Google Scholar]
- 26.Hohaus S, Massini G, D’Alo F, et al. Association between glutathione S-transferase genotypes and Hodgkin’s lymphoma risk and prognosis. Clin Cancer Res 2003;9(9):3435–3440. [PubMed] [Google Scholar]
- 27.Hohaus S, Di Ruscio A, Di Febo A, et al. Glutathione S-transferase P1 genotype and prognosis in Hodgkin’s lymphoma. Clin Cancer Res 2005;11(6):2175–2179. [DOI] [PubMed] [Google Scholar]
- 28.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(1): 52–58. [DOI] [PubMed] [Google Scholar]
- 29.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(4):457–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rossi D, Rasi S, Franceschetti S, et al. Analysis of the host pharmacogenetic background for prediction of outcome and toxicity in diffuse large B-cell lymphoma treated with R-CHOP21. Leukemia 2009;23(6):1118–1126. [DOI] [PubMed] [Google Scholar]
- 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(10):564–571. [DOI] [PubMed] [Google Scholar]
- 32.Hawkins DS, Chi YY, Anderson JR, et al. Addition of vincristine and irinotecan to vincristine, dactinomycin, and cyclophosphamide does not improve outcome for intermediate-risk rhabdomyosarcoma: a report from the Children’s Oncology Group. J Clin Oncol 2018;36(27):2770–2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shen R, Fan JB, Campbell D, et al. High-throughput SNP genotyping on universal bead arrays. Mutat Res 2005;573(1–2):70–82. [DOI] [PubMed] [Google Scholar]
- 34.Thorisson GA, Smith AV, Krishnan L, Stein LD. The International HapMap Project web site. Genome Res 2005;15(11):1592–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Benjamini Y, Hochberg Y. Controlling the false discovery rate - a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol 1995;57(1):289–300. [Google Scholar]
- 36.Pratt CB, Hustu HO, Fleming ID, Pinkel D. Coordinated treatment of childhood rhabdomyosarcoma with surgery, radiotherapy, and combination chemotherapy. Cancer Res 1972;32(3):606–610. [PubMed] [Google Scholar]
- 37.Maurer HM, Beltangady M, Gehan EA, et al. The Intergroup Rhabdomyosarcoma Study-I. A final report. Cancer 1988;61(2):209–220. [DOI] [PubMed] [Google Scholar]
- 38.Maurer HM, Gehan EA, Beltangady M, et al. The Intergroup Rhabdomyosarcoma Study-II. Cancer 1993;71(5):1904–1922. [DOI] [PubMed] [Google Scholar]
- 39.Crist W, Gehan EA, Ragab AH, et al. The Third Intergroup Rhabdomyosarcoma Study. J Clin Oncol 1995;13(3):610–630. [DOI] [PubMed] [Google Scholar]
- 40.Crist WM, Anderson JR, Meza JL, et al. Intergroup rhabdomyosarcoma study-IV: results for patients with nonmetastatic disease. J Clin Oncol 2001;19(12):3091–3102. [DOI] [PubMed] [Google Scholar]
- 41.Arndt C, Hawkins D, Anderson JR, Breitfeld P, Womer R, Meyer W. Age is a risk factor for chemotherapy-induced hepatopathy with vincristine, dactinomycin, and cyclophosphamide. J Clin Oncol 2004;22(10):1894–1901. [DOI] [PubMed] [Google Scholar]
- 42.Spunt SL, Smith LM, Ruymann FB, et al. Cyclophosphamide dose intensification during induction therapy for intermediate-risk pediatric rhabdomyosarcoma is feasible but does not improve outcome: a report from the soft tissue sarcoma committee of the children’s oncology group. Clin Cancer Res 2004;10(18 Pt 1):6072–6079. [DOI] [PubMed] [Google Scholar]
- 43.Gupta AA, Anderson JR, Pappo AS, et al. Patterns of chemotherapy-induced toxicities in younger children and adolescents with rhabdomyosarcoma: a report from the Children’s Oncology Group Soft Tissue Sarcoma Committee. Cancer 2012;118(4):1130–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Arndt CA, Stoner JA, Hawkins DS, et al. Vincristine, actinomycin, and cyclophosphamide compared with vincristine, actinomycin, and cyclophosphamide alternating with vincristine, topotecan, and cyclophosphamide for intermediate-risk rhabdomyosarcoma: Children’s Oncology Group Study D9803. J Clin Oncol 2009;27(31):5182–5188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.de Jonge ME, Huitema AD, Rodenhuis S, Beijnen JH. Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet 2005;44(11):1135–1164. [DOI] [PubMed] [Google Scholar]
- 46.Stanulla M, Schrappe M, Brechlin AM, Zimmermann M, Welte K. 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. Blood 2000;95(4):1222–1228. [PubMed] [Google Scholar]
- 47.Rocha JC, Cheng C, Liu W, et al. Pharmacogenetics of outcome in children with acute lymphoblastic leukemia. Blood 2005;105(12):4752–4758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Vester U, Kranz B, Zimmermann S, Buscher R, Hoyer PF. The response to cyclophosphamide in steroid-sensitive nephrotic syndrome is influenced by polymorphic expression of glutathion-S-transferases-M1 and -P1. Pediatr Nephrol 2005;20(4):478–481. [DOI] [PubMed] [Google Scholar]
- 49.Sharda SV, Gulati S, Tripathi G, et al. Do glutathione-S-transferase polymorphisms influence response to intravenous cyclophosphamide therapy in idiopathic nephrotic syndrome? Pediatr Nephrol 2008;23(11):2001–2006. [DOI] [PubMed] [Google Scholar]
- 50.Barahmani N, Carpentieri S, Li XN, et al. Glutathione S-transferase M1 and T1 polymorphisms may predict adverse effects after therapy in children with medulloblastoma. Neuro Oncol 2009;11(3):292–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ren S, Yang JS, Kalhorn TF, Slattery JT. Oxidation of cyclophosphamide to 4-hydroxycyclophosphamide and deschloroethyl-cyclophosphamide in human liver microsomes. Cancer Res 1997;57(19):4229–4235. [PubMed] [Google Scholar]
- 52.Chang TK, Weber GF, Crespi CL, Waxman DJ. Differential activation of cyclophosphamide and ifosphamide by cytochromes P-450 2B and 3A in human liver microsomes. Cancer Res 1993;53(23): 5629–5637. [PubMed] [Google Scholar]
- 53.Roy P, Yu LJ, Crespi CL, Waxman DJ. Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos 1999;27(6):655–666. [PubMed] [Google Scholar]
- 54.McCune JS, Salinger DH, Vicini P, Oglesby C, Blough DK, Park JR. Population pharmacokinetics of cyclophosphamide and metabolites in children with neuroblastoma: a report from the Children’s Oncology Group. J Clin Pharmacol 2009;49(1):88–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Busse D, Busch FW, Bohnenstengel F, et al. Dose escalation of cyclophosphamide in patients with breast cancer: consequences for pharmacokinetics and metabolism. J Clin Oncol 1997;15(5):1885–1896. [DOI] [PubMed] [Google Scholar]
- 56.Busse D, Busch FW, Schweizer E, et al. Fractionated administration of high-dose cyclophosphamide: influence on dose-dependent changes in pharmacokinetics and metabolism. Cancer Chemother Pharmacol 1999;43(3):263–268. [DOI] [PubMed] [Google Scholar]
- 57.Marsh S, Mallon MA, Goodfellow P, McLeod HL. Concordance of pharmacogenetic markers in germline and colorectal tumor DNA. Pharmacogenomics 2005;6(8):873–877. [DOI] [PubMed] [Google Scholar]
- 58.Jaremko M, Justenhoven C, Abraham BK, et al. MALDI-TOF MS and TaqMan assisted SNP genotyping of DNA isolated from formalin-fixed and paraffin-embedded tissues (FFPET). Hum Mutat 2005;25(3): 232–238. [DOI] [PubMed] [Google Scholar]
- 59.McCune JS, Hawke RL, LeCluyse EL, et al. In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther 2000;68(4):356–366. [DOI] [PubMed] [Google Scholar]
- 60.Gilbert CJ, Petros WP, Vredenburgh J, et al. Pharmacokinetic interaction between ondansetron and cyclophosphamide during high-dose chemotherapy for breast cancer. Cancer Chemother Pharmacol 1998;42(6):497–503. [DOI] [PubMed] [Google Scholar]
- 61.Cagnoni PJ, Matthes S, Day TC, Bearman SI, Shpall EJ, Jones RB. Modification of the pharmacokinetics of high-dose cyclophosphamide and cisplatin by antiemetics. Bone Marrow Transplant 1999;24(1):1–4. [DOI] [PubMed] [Google Scholar]
- 62.Evans WE, Relling MV. Moving towards individualized medicine with pharmacogenomics. Nature 2004;429(6990):464–468. [DOI] [PubMed] [Google Scholar]
- 63.Pui CH, Yang JJ, Hunger SP, et al. Childhood acute lymphoblastic leukemia: progress through collaboration. J Clin Oncol 2015;33(27):2938–2948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Shern JF, Patidar R, Song Y, et al. Targeted resequencing of pediatric rhabdomyosarcoma: report from the Children’s Oncology Group, the Children’s Cancer and Leukaemia Group, The Institute of Cancer Research UK, and the National Cancer Institute. J Clin Oncol 2018;36:10515. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
