Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

CCR Focus

Genome-Wide Association Study: A Useful Tool to Identify Common Genetic Variants Associated with Drug Toxicity and Efficacy in Cancer Pharmacogenomics

Siew-Kee Low, Atsushi Takahashi, Taisei Mushiroda and Michiaki Kubo
Siew-Kee Low
1Laboratory for Statistical Analysis, Core for Genomic Medicine; 2Laboratory for Pharmacogenomics; and 3Laboratory for Genotyping Development, Core for Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Atsushi Takahashi
1Laboratory for Statistical Analysis, Core for Genomic Medicine; 2Laboratory for Pharmacogenomics; and 3Laboratory for Genotyping Development, Core for Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Taisei Mushiroda
1Laboratory for Statistical Analysis, Core for Genomic Medicine; 2Laboratory for Pharmacogenomics; and 3Laboratory for Genotyping Development, Core for Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michiaki Kubo
1Laboratory for Statistical Analysis, Core for Genomic Medicine; 2Laboratory for Pharmacogenomics; and 3Laboratory for Genotyping Development, Core for Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-13-2755 Published May 2014
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

In recent years, the utilization of genome-wide association study (GWAS) has proved to be a beneficial method to identify novel common genetic variations not only for disease susceptibility but also for drug efficacy and drug-induced toxicity, creating a field of pharmacogenomics studies. In addition, the findings from GWAS also generate new biologic hypotheses that could improve the understanding of pathophysiology for disease or the mechanism of drug-induced toxicity. This review highlights the implications of GWAS that have been published to date and discusses the successes as well as challenges of using GWAS in cancer pharmacogenomics. The aim of pharmacogenomics is to realize the vision of personalized medicine; it is hoped that through GWAS, novel common genetic variations could be identified to predict clinical outcome and/or toxicity in cancer therapies that subsequently could be implemented to improve the quality of lives of patients with cancer. Nevertheless, given the complexity of cancer therapies, underpowered studies, and large heterogeneity of study designs, collaborative efforts are needed to validate these findings and overcome the limitations of GWA studies before clinical implementation.

See all articles in this CCR Focus section, “Progress in Pharmacodynamic Endpoints.”

Clin Cancer Res; 20(10); 2541–52. ©2014 AACR.

Introduction

Pharmacogenomics is the study to uncover the role of human genetic variations that affect drug efficacy and toxicity (Fig. 1). Genetic variations that include common genetic variants, represented mostly by the single-nucleotide polymorphism (SNP), and rare variants in genes that are involved in drug pharmacokinetics and pharmacodynamics could explain part of this inter-individual variability. Genetic polymorphisms in genes encoding drug metabolism and drug transporter affect drug availability at the target site (drug pharmacokinetics), whereas variants in drug target proteins, such as receptors, enzymes, and intracellular signaling proteins, affect a patient's sensitivity to a drug (drug pharmacodynamics). In recent years, great efforts have been made to identify genetic variations associated with drug-induced phenotypes, with two of the reputable examples being the association of TPMT variants with 6-mercaptopurine–induced myelosuppression for the treatment of pediatric acute lymphoblastic leukemia and the association of UGT1A1 variants with camptothecin-related neutropenia and diarrhea for the treatment of colorectal and lung cancers. Both of these genes were identified through a candidate gene approach. The U.S. Food and Drug Administration (FDA) has revised the drug labels of 6-mercaptopurine and camptothecin to include genotypes of TPMT and UGT1A1 as one of the risk factors for toxicity and stated the genotypes of these genes would be helpful to predict the occurrence of severe adverse events before treatment (1–5).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Promise of pharmacogenomics for the realization of personalized medicine. Patients with similar diagnoses are prescribed the same treatment. The identification of genetic variants associated with drug response, which include drug efficacy and toxicity, could help to predict and reduce the occurrence of events before drug treatment.

During the past decade, pharmacogenomics studies have been mostly carried out through candidate gene approaches in which genes are selected a priori based on their relevance to the drug's mechanism of action. These specific genes may be involved in drug metabolisms, transports, or toxicities that mostly affect drug pharmacokinetics. In the context of adverse drug reactions, besides these genes encoding drug-related proteins, other genes involved in immune-mediated responses may also be relevant, such as human leukocyte antigens and their association with drug hypersensitivity. The establishment of the HapMap database, development of tagSNP selection algorithms, and improvements in genotyping technology have made the concept of genome-wide association studies a reality for linking specific genetic variants across the entire genome with the phenotype of interest (e.g., human disease and individual's traits; Fig. 2). Genome-wide association studies (GWAS) have successfully identified germline variants that are associated with the genetic susceptibility of many common diseases (6). This approach has recently become one of the most beneficial methods in pharmacogenomics research. Unlike the candidate gene approach, GWAS provides a hypothesis-free approach that facilitates the possibility of identifying genetic variants in novel genes that might be involved in both drug pharmacokinetics and pharmacodynamics as well as mechanisms underlying gene–phenotype interactions. As most of the reported candidate-gene approach studies focus only on genes that influence drug pharmacokinetics, GWAS might have the capacity to discover markers that will have a direct or indirect effect on drug pharmacodynamics and will expand our understanding in the interpretation of inter-individual variability of drug response. The pharmacogenomics database pharmGKB (7) is a very useful and important resource that provides substantial information to evaluate the level of evidence of genetic variants identified from GWAS and candidate gene approaches in pharmacogenomics studies up to the present.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Summary workflow of GWAS. GWAS starts with the determination of phenotypes. In pharmacogenomics studies, cases are often the patients who do not respond or who develop severe adverse reactions, whereas controls are patients who respond to the treatment or who do not develop any adverse events after exposure to drug(s) treatment. All the samples were genotypes with chips that contained up to hundreds of thousands of SNPs. Quality control (QC) is a crucial step to ensure the association studies were performed with a good-quality sample and SNP set. Sample quality control usually includes (1) sample quality to exclude poorly genotyped samples, (2) identity-by-state analysis to exclude close relatedness samples, and (3) principal component analysis to evaluate population stratification of the sample sets to obtain a homogeneous sample set before performing the association study. SNPs will be excluded if (1) they are of low genotype quality, (2) if SNPs deviated from normal distribution by evaluating Hardy–Weinberg equilibrium in control samples, and (3) if they contain nonpolymorphic SNPs (minor allele frequency = 0). To evaluate the association distribution, quantile–quantile plots (Q–Q plot) of observed P value versus expected P value and genomic inflation factor (λ value) were evaluated to eliminate the possibility of population substructure. Manhattan plots of P value (−log10) versus chromosome loci were utilized to depict an overview of the GWAS, with each dot representing a SNP and each color representing a chromosome. The post-GWAS included (1) a meta-analysis that combined multiple studies to identify significantly associated SNPs, and (2) functional analysis. Two of the most common functional analyses of the identified variants are (A) electrophoretic mobility shift assay (EMSA) to check the existence of proteins, mainly transcription factors, binding to SNP-contained DNA fragments and (B) luciferase reporter assay (comparison of relative luciferase activity) to assess the associated SNPs that could affect differential gene expression (as shown in figure). (3) Other analyses, including gene-based analysis, pathway analysis, polygenic risk estimation, SNP–SNP interaction, SNP–environment interaction, etc., could be carried out after GWAS.

Successful Pharmacogenomics Studies Using GWAS

GWAS not only successfully verified previously reported loci but also identified a number of novel associations between drug responses or adverse reactions with genetic variants that may affect clinical practice and also provide novel insights into biologic mechanisms of drug-related pathways (Table 1; refs. 8–22).

View this table:
  • View inline
  • View popup
Table 1.

Successful pharmacogenomics studies using GWAS

For cardiovascular diseases, simvastatin is a drug that is used to control elevated cholesterol levels; GWAS identified rs4363657 in SLCO1B1 (a solute carrier organic anion transporter that transports a number of drugs, including statins), to be significantly associated with simvastatin-induced myopathy (8). rs4363657 is in strong linkage disequilibrium with rs4149056 (V174A), a nonsynonymous SNP, that is related to statin pharmacokinetics (23). The Clinical Pharmacogenetics Implementation Consortium recommends use of the SNP rs4149056 to manage the risk of simvastatin-induced myopathy (24). GWAS also successfully validated variants on VKORC1, CYP2C9, and CYP4F2 as significantly influencing the maintenance dose of warfarin, an anticoagulant for reducing thromboembolic events (9, 10). The incorporation of genetic variants has provided an additional predictor for better warfarin dose assessments.

For the treatment of infectious disease, GWAS successfully identified HLA-B*57:01 as a major determinant of drug-induced liver injury caused by the administration of flucloxacilin, a drug used in the treatment of staphylococcal infection (13). Second, two GWAS studies from Japan and Australia uncovered genetic variants in IL28B that are associated with response to pegylated IFN-α and ribavirin therapy for patients with chronic hepatitis C (17, 18). Diagnostic tests based on host IL28B genotypes might help to identify nonresponders to pegylated IFN-α and ribavirin therapy who can opt for adjunctive or alternative therapies and thus avoid potential adverse drug reactions that might otherwise occur.

Two GWA studies from Japanese and European populations validated HLA-A*31:01 to be significantly associated with cutaneous adverse drug reactions, including hypersensitivity reaction, Steven–Johnson syndrome, and toxic epidermal necrolysis caused by carbamazepine, a drug that is frequently prescribed for the treatment of epilepsy, trigeminal neuralgia, and bipolar disease (Fig. 3; refs. 19 and 20).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Manhattan plot derived from GWAS of carbamazepine-induced cutaneous adverse drug reactions in Japanese population. In this plot, SNPs on HLA-A in chromosome 6 are significantly associated (P < 5.0E−08) with cutaneous adverse drug reactions. The associations of these SNPs were successfully validated in an independent sample set. Reprinted from Ozeki et al. (19) with permission.

It is important to collect uniform phenotypes to ensure the success of GWAS in pharmacogenomics studies. Some of the studies, although using relatively small sample sizes, showed significant associations of variants that have large effect sizes; for example, genotypes in the HLA region were observed. These variants may prove to be a useful diagnostic tests to predict individual's drug response.

Use of GWAS in Pharmacogenomics Studies for Individual Cancer Types

Cancer pharmacogenomics are well recognized for their complexity, as both germline genetic variants and the somatic mutations in the individual patient's tumor play important roles in determining response to anticancer agents. Activating mutations in the tyrosine kinase domain of EGF receptor (EGFR) are one of the prominent examples of somatic mutations that increase the sensitivity to gefitinib and erlotinib in non–small cell lung cancers (NSCLC; refs. 25 and 26). However, germline genetic variants identified through GWAS, which is the focus of this review, are variants that affect drug response, efficacy, and toxicity independent from tumor somatic mutation status or disease type (Table 2; refs. 27–39).

View this table:
  • View inline
  • View popup
Table 2.

Cancer pharmacogenomics studies utilizing GWAS

Breast cancer

GWAS identified a promising locus on the T-cell leukemia 1A (TCL1A) gene to be associated with musculoskeletal adverse events (MS-AE) caused by the “third-generation” aromatase inhibitors that include anastrozole, exemestane, and letrozole, which are the effective endocrine therapies for estrogen receptor (ER)-positive early breast cancer (27). The associated variant, rs11849538 (P = 6.67E−07, OR = 2.21), closest to the 3′ end of TCL1A, was predicted to create an estrogen-response element and this prediction was subsequently confirmed with a chromatin immunoprecipitation assay using ERα-transfected lymphoblastoid cell lines (LCL) with the known genotype. In addition, LCLs expressing the variant SNP sequence showed significantly greater TCL1A expression compared with the wild-type sequence (27). Correlations between SNP-dependent differential TCL1A expression and various cytokine receptor genes, including interleukin (IL)-17, IL-17RA, IL-12, IL-12RB2, and IL-1R2 expression as well as NF-κB transcriptional activity were observed. These findings further suggested that the MS-AE occurrence is related to the inflammatory response (40). The findings of the GWAS implied an entirely new biologic hypothesis and improved the understanding of the pathophysiology of aromatase inhibitor therapy induced MS-AE.

Kiyotani and colleagues conducted a GWAS with 240 breast cancer patients who received tamoxifen monotherapy and identified a SNP, rs10509373 (log-rank P = 1.26E−10, multivariate HR = 4.53), in C10orf11 (unknown function) to be significantly associated with recurrence-free survival (RFS) after validation in two independent replication sample sets of 105 and 117 cases (28). Combined analysis revealed that there are cumulative effects of the associated genotypes from C10orf11, CYP2D6, and ABCC2 (previously identified to be associated with RFS in the same dataset; ref. 41), which indicated that the HRs for patients who carry three or more risk alleles increased the risk of recurrence from 6.51-fold (three risk alleles) to 119.51-fold (five risk alleles) compared with patients who carry only one risk allele (28).

Baldwin and colleagues performed a GWAS of paclitaxel chemotherapy-induced sensory neuropathy and identified a SNP in FGD4, rs10771973 (P = 2.6E−06, HR = 1.57) to be associated with the onset of sensory peripheral neuropathy; this finding was subsequently validated in a European and African-American replication cohort (29). FGD4 encodes a Rho-GTPase guanine nucleotide exchange factor previously reported to play a role in congenital peripheral neuropathies (42). In addition, they suggested that SNPs in EPHA5 (rs7349683) and FZD3 (rs10771973) could be associated with the onset or severity of the paclitaxel-induced sensory peripheral neuropathy based on marginal significance levels, biologic relevance, and estimated effect size (29).

Although alopecia is not a life-threatening event, it induces psychological stress, influences physical appearance, and often affects the patient's quality of life (43). Chung and colleagues reported a GWAS of monotherapy or combination chemotherapy-induced alopecia (hair loss) of patients with breast cancer and identified a SNP, rs3820706 (P = 1.85E−09, OR = 2.38) in CACNB4, a calcium channel voltage-dependent subunit β4, to be significantly associated with drug-induced alopecia (30). A potassium channel opener, minoxidil, was approved by the FDA for the treatment of alopecia (44), and the results of the current GWAS further suggest that ion channels might be involved in the pathogenesis of alopecia.

Lung cancer

Wu and colleagues reported a GWAS with advanced-stage NSCLC patients who received platinum-based chemotherapy and identified a SNP, rs1878022, in CMKLR1 (P = 5.13E−07, HR = 1.33) to be statistically significantly associated with poor overall survival (OS; ref. 31). The mean survival time (MST) for patients who possessed a non-risk homozygous genotype (16.05 months) was significantly longer than that for patients who carried the risk genotypes (10.72 months, P = 6.76E−05; ref. 31). CMKLR1 encodes for a 7 transmembrane G-protein–coupled receptor, which has been reported to be highly expressed in lung tissues (45). The binding of ligands, chimerin and resolving E1, to the receptor has been shown to activate various pathways such as mitogen-activated protein kinase (MAPK), extracellular signal-related kinase 1 and 2, and angiogenesis (46, 47).

Hu and colleagues performed three-stage GWAS studies in advanced-stage NSCLC patients receiving platinum-based chemotherapy in the Chinese population and identified five SNPs, rs7629386, rs969088, rs3850370, rs41997, and rs12000445 (meta-analysis P ranging from 3.63E−05 to 4.19E−07) that were suggestively associated with the survival of patients with NSCLC. Among these five SNPs, two SNPs, rs7629386 in CTNNB1 and rs3850370 in SNW1-ALKBH1-NRXN3, were further replicated in a Caucasian population (32). One of the strengths of this study is that the suggestive associated SNPs were not only validated in an independent sample from the Chinese population but also in another sample set from the Caucasian population. Assuming the underlying mechanism that caused the adverse drug event occurrence is the same, it is important to evaluate the associated genetic loci across different populations even though the genetic architecture (different allele frequency and linkage disequilibrium structure) might be a complex issue to resolve.

Sato and colleagues reported a GWAS of advanced NSCLC in which they found that three SNPs, rs1656402 in EIF4E2 (P = 8.4E−08, HR = 4.22, MST for AG+AA and GG were 18.0 and 7.7 months, respectively), rs1209950 in ETS2 (P = 2.8E−07, HR = 4.96, MST for CC and CT+TT were 17.7 and 7.4 months, respectively), and rs9981861 in DSCAM (P = 3.5E−06, HR = 16.1, MST for GG+AG and AA were 17.1 and 3.8 months, respectively), were suggestively associated with OS of patients with NSCLC after they received combined therapy of carboplatin and paclitaxel (33).

In addition to drug efficacy studies, Han and colleagues conducted a GWAS for irinotecan-induced diarrhea (grade 3) and neutropenia (grade 4) in patients with NSCLC and reported that rs1517114 in C8orf34, rs1661167 in FLJ41856, and rs2745761 in PLCB1 showed strong association with severe diarrhea, whereas rs11128347 in PDZRN3 and rs11979430 on chromosome 7 were suggestively associated with irinotecan-induced severe neutropenia (34). The authors also demonstrated that associated variants identified from candidate gene approaches, such as UGT1A1*6 and SLCO1B1 521T>C, were significantly associated with severe neutropenia and ABCC2 3972C>T was associated with diarrhea (34).

Pancreatic cancer (gemcitabine)

Gemcitabine is a deoxycytidine analogue that is used in treating patients with various solid tumors, including pancreatic cancer and NSCLC. A GWAS in patients with pancreatic cancer who received gemcitabine identified a nonsynonymous coding variant, rs763780 (H161R), in IL17F (P = 2.61E−08, HR = 3.3) to be associated with OS. Patients who were rs763780 heterozygous had reduced median OS (3.1 months) compared with patients without the variant (6.8 months; ref. 35). IL17F encodes for IL-17F, which is involved in angiogenesis and plays a role in the growth and metastatic spread of pancreatic cancer (48, 49).

Kiyotani and colleagues conducted a GWAS to identify gemcitabine monotherapy–induced severe leukopenia/neutropenia with patients recruited from the Biobank Japan and identified two possibly associated loci, rs11141915 in DAPK1 (P = 1.27E−06, OR = 4.10) and rs1901440 between NCKAP5 and MIR3679 (P = 3.11E−06, OR = 34.00; ref. 36). As an expansion of this report, a large-scale pharmacogenomics study involving patients recruited from Biobank Japan who received therapy with various chemotherapeutic agents identified six suggestive loci (P < 1.0E−05), including rs9961113 between BDP1P and SALL3, rs2547917 in PDE4D, rs12900463 in ALPK3, rs9609078 in OSBP2, rs6863418 near to HMP19 and rs6037430 near to NRSN2, to be associated with gemcitabine-induced grade 3 or 4 leukopenia/neutropenia (37).

Acute lymphoblastic leukemia

Methotrexate is an antimetabolite agent that is used to treat malignancies such as acute lymphoblastic leukemia (ALL) and autoimmune diseases. A GWAS with ALL patients from various ancestry groups (African, European, and Asian) identified multiple SNPs on the SLCO1B1 gene, encoding for the organic anion transporter (OATP1B1), to be significantly associated with methotrexate clearance and gastrointestinal toxicity (38). OATP1B1 was localized to the sinusoidal membrane of hepatocytes, and was shown to transport methotrexate in vitro. The association was further validated in another GWAS study with a larger cohort of patients with ALL (n = 1,279) treated with high-dose methotrexate (50). Variants in SLCO1B1 could be clinically useful in identifying patients who are at risk of low methotrexate clearance so that methotrexate clearance could be improved by possible interventions, such as increased intravenous hydration and/or alkalinization (50).

Lessons, Challenges, and Future Directions in Cancer Pharmacogenomics Studies Using GWAS

The identification of associated genetic variants through GWAS in cancer pharmacogenomics has been challenging and seemingly still requires great efforts to realize the goal of personalized medicine. The two greatest challenges commonly encountered while performing GWAS are (i) statistically underpowered case–control studies and (ii) stringent significant threshold because of multiple testing.

It is important to evaluate the statistical power, which correlates among sample size, effect size, causal allele frequency, and association of the causal variant to the study phenotype, beforehand. To identify an association of a modest effect SNP with an OR of 1.2 and causal allele frequency of 0.25, a sample set of approximately 5,000 individuals is necessary to identify significant association; on the other hand, in a high-effect SNP with an OR of 2.0 and causal allele frequency at 0.25, only 300 individuals are necessary (51). This estimation reflects the fact that to achieve sufficient statistical power in pharmacogenomics studies to identify truly associated variants with low/moderate effect size, a large number of patients who are treated with uniform therapy is required. This issue is particularly difficult to achieve in the study of cancer pharmacogenomics because of varied drug combinations, different dosing regimens, and treatment durations, as well as dosage adjustment based on the patient's condition that often contribute to the heterogeneity in treatment effects. Rapid advances in the development of molecular-targeted drugs coupled with changing treatment paradigms add to the complexity of pharmacogenomics analysis. Furthermore, the incidence of adverse drug events is expectedly low, which subsequently contributes to the difficulties in collecting an adequate number of cases given a specific therapy. Hence, to overcome this issue, collecting DNA samples with identical treatment protocols and robust phenotype/clinical data from large, collaborative clinical trials, both local and international, could further advance the field of cancer pharmacogenomics. One of the ways this could happen is through the establishment of an international alliance, such as the Global Alliance for Pharmacogenomics (52) to facilitate international collaboration by bringing together resources to advance the understanding of drug responses.

Because GWAS studies are carried out by testing approximately a million SNPs simultaneously, and multiple testing is applied to avoid the identification of false-positive associated SNPs that are due to chance. Currently, the genome-wide significance threshold is set at P < 5.0 × 10−8 based on Bonferroni correction, which divides 0.05 by a million independent SNPs. As most of the identified variants from the GWAS of cancer pharmacogenomics did not surpass this stringent and conservative threshold, verification with an independent set of samples would be useful to support the findings from GWAS. One reasonable way to compromise the GWAS stringent significant threshold is the use of pharmacogene panels (this topic is addressed extensively in this CCR Focus section), which screened SNPs in genes with known relevance to drug pharmacokinetics and pharmacodynamics (53). To provide additional supportive evidence of variants identified from GWAS, functional analyses, such as electrophoretic mobility shift assay and luciferase reporter assay, could be carried out to investigate the effects of the variants affecting the phenotypes. HapMap lymphoblastoid cell lines (LCL), which are known to have ample genetic information, could be utilized to identify or validate variants that correlate with cytotoxicity after specific drug exposure (Fig. 4; ref. 54). This method could enhance our understanding of genotype–phenotype correlation in pharmacogenomics studies.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Cell-based genome-wide approach by using EBV-transformed B lymphoblastoid cell lines (LCL) derived from blood donor of the HapMap/1000 Genomes project. One of the greatest advantages of using the cell-based model is the readily available SNP information from the HapMap and/or 1000 Genomes database. It is now feasible to treat the cell lines with the drug of interest to evaluate the association of genotype and drug response and genotype and gene expression as well as gene expression and drug response. The identification of candidate genes or SNPs that are associated with drug response in the cell model could be validated in an independent study by using a clinical sample.

Markers that have been identified through GWAS of pharmacogenomics studies should be validated in an independent sample set before being incorporated into a prediction algorithm. Subsequently, the validity of the prediction algorithm could be evaluated in a prospective study with patients who received the same treatment protocol. An automated SNP genotyping system could be provided to speed up the process of SNP genotyping before treatment. Application of GWAS could be useful in the drug discovery phase in the future by incorporating SNP genotypes that could distinguish/predict individuals who might develop adverse drug reactions beforehand.

Conclusions

Although the use of GWAS to identify susceptible loci with complex diseases has reached a bottleneck in the genomic era, GWAS of cancer pharmacogenomics could still contribute in identifying novel loci because of the constant development of new drugs or new protocols from clinical trials. In fact, the applications of GWAS in cancer pharmacogenomics have identified a handful of candidate genetic loci associated with drug-induced toxicities and efficacies as well as suggested novel biologic pathways to further improve our understanding of the underlying mechanisms. Nevertheless, most of these studies are underpowered and require additional validation in multiple independent sample sets or functional analyses to further elucidate the gene–phenotype relationship. To ensure the quality and success of pharmacogenomics studies, local and international collaborative efforts are essential to collection of sufficient and homogeneous sample sets. It is hoped that genetic markers identified from pharmacogenomics studies could be used in clinical practice in the near future to predict drug efficacy and toxicity, and thus represent the promise of personalized medicine to improve patients' quality of life.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Authors' Contributions

Conception and design: S.-K. Low, A. Takahashi, M. Kubo

Development of methodology: S.-K. Low, A. Takahashi

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.-K. Low, A. Takahashi, M. Kubo

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.-K. Low, A. Takahashi

Writing, review, and or revision of the manuscript: S.-K. Low, A. Takahashi, T. Mushiroda, M. Kubo

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.-K. Low, A. Takahashi, M. Kubo

Study supervision: A. Takahashi, M. Kubo

  • Received December 19, 2013.
  • Revision received March 14, 2014.
  • Accepted March 27, 2014.
  • ©2014 American Association for Cancer Research.

References

  1. 1.↵
    1. Relling MV,
    2. Hancock ML,
    3. Rivera GK,
    4. Sandlund JT,
    5. Ribeiro RC,
    6. Krynetski EY,
    7. et al.
    Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus. J Natl Cancer Inst 1999;91:2001–8.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Relling MV,
    2. Klein TE
    . CPIC: clinical pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther 2011;89:464–7.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Innocenti F,
    2. Undevia SD,
    3. Iyer L,
    4. Chen PX,
    5. Das S,
    6. Kocherginsky M,
    7. et al.
    Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 2004;22:1382–8.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Iyer L,
    2. Das S,
    3. Janisch L,
    4. Wen M,
    5. Ramirez J,
    6. Karrison T,
    7. et al.
    UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2002;2:43–7.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Hoskins JM,
    2. Goldberg RM,
    3. Qu P,
    4. Ibrahim JG,
    5. McLeod HL
    . UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst 2007;99:1290–5.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    National Human Genome Research Institute. Available from: www.genome.gov/gwastudies/.
  7. 7.↵
    www.pharmgkb.org
  8. 8.↵
    1. Group SC,
    2. Link E,
    3. Parish S,
    4. Armitage J,
    5. Bowman L,
    6. Heath S,
    7. et al.
    SLCO1B1 variants and statin-induced myopathy—a genomewide study. N Engl J Med 2008;359:789–99.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Takeuchi F,
    2. McGinnis R,
    3. Bourgeois S,
    4. Barnes C,
    5. Eriksson N,
    6. Soranzo N,
    7. et al.
    A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet 2009;5:e1000433.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Cha PC,
    2. Mushiroda T,
    3. Takahashi A,
    4. Kubo M,
    5. Minami S,
    6. Kamatani N,
    7. et al.
    Genome-wide association study identifies genetic determinants of warfarin responsiveness for Japanese. Hum Mol Genet 2010;19:4735–44.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    1. Shuldiner AR,
    2. O'Connell JR,
    3. Bliden KP,
    4. Gandhi A,
    5. Ryan K,
    6. Horenstein RB,
    7. et al.
    Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA 2009;302:849–57.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Turner ST,
    2. Bailey KR,
    3. Fridley BL,
    4. Chapman AB,
    5. Schwartz GL,
    6. Chai HS,
    7. et al.
    Genomic association analysis suggests chromosome 12 locus influencing antihypertensive response to thiazide diuretic. Hypertension 2008;52:359–65.
    OpenUrlCrossRef
  13. 13.↵
    1. Daly AK,
    2. Donaldson PT,
    3. Bhatnagar P,
    4. Shen Y,
    5. Pe'er I,
    6. Floratos A,
    7. et al.
    HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet 2009;41:816–9.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Lucena MI,
    2. Molokhia M,
    3. Shen Y,
    4. Urban TJ,
    5. Aithal GP,
    6. Andrade RJ,
    7. et al.
    Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011;141:338–47.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Ochi H,
    2. Maekawa T,
    3. Abe H,
    4. Hayashida Y,
    5. Nakano R,
    6. Kubo M,
    7. et al.
    ITPA polymorphism affects ribavirin-induced anemia and outcomes of therapy—a genome-wide study of Japanese HCV virus patients. Gastroenterology 2010;139:1190–7.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Fellay J,
    2. Thompson AJ,
    3. Ge D,
    4. Gumbs CE,
    5. Urban TJ,
    6. Shianna KV,
    7. et al.
    ITPA gene variants protect against anaemia in patients treated for chronic hepatitis C. Nature 2010;464:405–8.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Tanaka Y,
    2. Nishida N,
    3. Sugiyama M,
    4. Kurosaki M,
    5. Matsuura K,
    6. Sakamoto N,
    7. et al.
    Genome-wide association of IL28B with response to pegylated interferon-α and ribavirin therapy for chronic hepatitis C. Nat Genet 2009;41:1105–9.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Suppiah V,
    2. Moldovan M,
    3. Ahlenstiel G,
    4. Berg T,
    5. Weltman M,
    6. Abate ML,
    7. et al.
    IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet 2009;41:1100–4.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Ozeki T,
    2. Mushiroda T,
    3. Yowang A,
    4. Takahashi A,
    5. Kubo M,
    6. Shirakata Y,
    7. et al.
    Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population. Hum Mol Genet 2011;20:1034–41.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. McCormack M,
    2. Alfirevic A,
    3. Bourgeois S,
    4. Farrell JJ,
    5. Kasperaviciute D,
    6. Carrington M,
    7. et al.
    HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med 2011;364:1134–43.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. GoDarts, Group UDPS, Wellcome Trust Case Control C,
    2. Zhou K,
    3. Bellenguez C,
    4. Spencer CC,
    5. Bennett AJ,
    6. Coleman RL,
    7. Tavendale R,
    8. et al.
    Common variants near ATM are associated with glycemic response to metformin in type 2 diabetes. Nat Genet 2011;43:117–20.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Singer JB,
    2. Lewitzky S,
    3. Leroy E,
    4. Yang F,
    5. Zhao X,
    6. Klickstein L,
    7. et al.
    A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat Genet 2010;42:711–4.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Konig J,
    2. Seithel A,
    3. Gradhand U,
    4. Fromm MF
    . Pharmacogenomics of human OATP transporters. Naunyn Schmiedebergs Arch Pharmacol 2006;372:432–43.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Wilke RA,
    2. Ramsey LB,
    3. Johnson SG,
    4. Maxwell WD,
    5. McLeod HL,
    6. Voora D,
    7. et al.
    The clinical pharmacogenomics implementation consortium: CPIC guideline for SLCO1B1 and simvastatin-induced myopathy. Clin Pharmacol Ther 2012;92:112–7.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Paez JG,
    2. Janne PA,
    3. Lee JC,
    4. Tracy S,
    5. Greulich H,
    6. Gabriel S,
    7. et al.
    EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Lynch TJ,
    2. Bell DW,
    3. Sordella R,
    4. Gurubhagavatula S,
    5. Okimoto RA,
    6. Brannigan BW,
    7. et al.
    Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129–39.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Ingle JN,
    2. Schaid DJ,
    3. Goss PE,
    4. Liu M,
    5. Mushiroda T,
    6. Chapman JA,
    7. et al.
    Genome-wide associations and functional genomic studies of musculoskeletal adverse events in women receiving aromatase inhibitors. J Clin Oncol 2010;28:4674–82.
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    1. Kiyotani K,
    2. Mushiroda T,
    3. Tsunoda T,
    4. Morizono T,
    5. Hosono N,
    6. Kubo M,
    7. et al.
    A genome-wide association study identifies locus at 10q22 associated with clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients in Japanese. Hum Mol Genet 2012;21:1665–72.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Baldwin RM,
    2. Owzar K,
    3. Zembutsu H,
    4. Chhibber A,
    5. Kubo M,
    6. Jiang C,
    7. et al.
    A genome-wide association study identifies novel loci for paclitaxel-induced sensory peripheral neuropathy in CALGB 40101. Clin Cancer Res 2012;18:5099–109.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Chung S,
    2. Low SK,
    3. Zembutsu H,
    4. Takahashi A,
    5. Kubo M,
    6. Sasa M,
    7. et al.
    A genome-wide association study of chemotherapy-induced alopecia in breast cancer patients. Breast Cancer Res 2013;15:R81.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Wu X,
    2. Ye Y,
    3. Rosell R,
    4. Amos CI,
    5. Stewart DJ,
    6. Hildebrandt MA,
    7. et al.
    Genome-wide association study of survival in non-small cell lung cancer patients receiving platinum-based chemotherapy. J Natl Cancer Inst 2011;103:817–25.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    1. Hu L,
    2. Wu C,
    3. Zhao X,
    4. Heist R,
    5. Su L,
    6. Zhao Y,
    7. et al.
    Genome-wide association study of prognosis in advanced non-small cell lung cancer patients receiving platinum-based chemotherapy. Clin Cancer Res 2012;18:5507–14.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Sato Y,
    2. Yamamoto N,
    3. Kunitoh H,
    4. Ohe Y,
    5. Minami H,
    6. Laird NM,
    7. et al.
    Genome-wide association study on overall survival of advanced non-small cell lung cancer patients treated with carboplatin and paclitaxel. J Thorac Oncol 2011;6:132–8.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Han JY,
    2. Shin ES,
    3. Lee YS,
    4. Ghang HY,
    5. Kim SY,
    6. Hwang JA,
    7. et al.
    A genome-wide association study for irinotecan-related severe toxicities in patients with advanced non-small-cell lung cancer. Pharmacogenomics J 2013;13:417–22.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Innocenti F,
    2. Owzar K,
    3. Cox NL,
    4. Evans P,
    5. Kubo M,
    6. Zembutsu H,
    7. et al.
    A genome-wide association study of overall survival in pancreatic cancer patients treated with gemcitabine in CALGB 80303. Clin Cancer Res 2012;18:577–84.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Kiyotani K,
    2. Uno S,
    3. Mushiroda T,
    4. Takahashi A,
    5. Kubo M,
    6. Mitsuhata N,
    7. et al.
    A genome-wide association study identifies four genetic markers for hematological toxicities in cancer patients receiving gemcitabine therapy. Pharmacogenet Genomics 2012;22:229–35.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Low SK,
    2. Chung S,
    3. Takahashi A,
    4. Zembutsu H,
    5. Mushiroda T,
    6. Kubo M,
    7. et al.
    Genome-wide association study of chemotherapeutic agent-induced severe neutropenia/leucopenia for patients in Biobank Japan. Cancer Sci 2013;104:1074–82.
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. Trevino LR,
    2. Shimasaki N,
    3. Yang W,
    4. Panetta JC,
    5. Cheng C,
    6. Pei D,
    7. et al.
    Germline genetic variation in an organic anion transporter polypeptide associated with methotrexate pharmacokinetics and clinical effects. J Clin Oncol 2009;27:5972–8.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Sarasquete ME,
    2. Garcia-Sanz R,
    3. Marin L,
    4. Alcoceba M,
    5. Chillon MC,
    6. Balanzategui A,
    7. et al.
    Bisphosphonate-related osteonecrosis of the jaw is associated with polymorphisms of the cytochrome P450 CYP2C8 in multiple myeloma: a genome-wide single nucleotide polymorphism analysis. Blood 2008;112:2709–12.
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    1. Liu M,
    2. Wang L,
    3. Bongartz T,
    4. Hawse JR,
    5. Markovic SN,
    6. Schaid DJ,
    7. et al.
    Aromatase inhibitors, estrogens and musculoskeletal pain: estrogen-dependent T-cell leukemia 1A (TCL1A) gene-mediated regulation of cytokine expression. Breast Cancer Res 2012;14:R41.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Kiyotani K,
    2. Mushiroda T,
    3. Imamura CK,
    4. Hosono N,
    5. Tsunoda T,
    6. Kubo M,
    7. et al.
    Significant effect of polymorphisms in CYP2D6 and ABCC2 on clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients. J Clin Oncol 2010;28:1287–93.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Stendel C,
    2. Roos A,
    3. Deconinck T,
    4. Pereira J,
    5. Castagner F,
    6. Niemann A,
    7. et al.
    Peripheral nerve demyelination caused by a mutant Rho GTPase guanine nucleotide exchange factor, frabin/FGD4. Am J Hum Genet 2007;81:158–64.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. McGarvey EL,
    2. Baum LD,
    3. Pinkerton RC,
    4. Rogers LM
    . Psychological sequelae and alopecia among women with cancer. Cancer Pract 2001;9:283–9.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Price VH
    . Treatment of hair loss. N Engl J Med 1999;341:964–73.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Wittamer V,
    2. Franssen JD,
    3. Vulcano M,
    4. Mirjolet JF,
    5. Le Poul E,
    6. Migeotte I,
    7. et al.
    Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med 2003;198:977–85.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Ohira T,
    2. Arita M,
    3. Omori K,
    4. Recchiuti A,
    5. Van Dyke TE,
    6. Serhan CN
    . Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J Biol Chem 2010;285:3451–61.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Kaur J,
    2. Adya R,
    3. Tan BK,
    4. Chen J,
    5. Randeva HS
    . Identification of chemerin receptor (ChemR23) in human endothelial cells: chemerin-induced endothelial angiogenesis. Biochem Biophys Res Commun 2010;391:1762–8.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Starnes T,
    2. Robertson MJ,
    3. Sledge G,
    4. Kelich S,
    5. Nakshatri H,
    6. Broxmeyer HE,
    7. et al.
    Cutting edge: IL-17F, a novel cytokine selectively expressed in activated T cells and monocytes, regulates angiogenesis and endothelial cell cytokine production. J Immunol 2001;167:4137–40.
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    1. Korc M
    . Pathways for aberrant angiogenesis in pancreatic cancer. Mol Cancer 2003;2:8.
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. Ramsey LB,
    2. Panetta JC,
    3. Smith C,
    4. Yang W,
    5. Fan Y,
    6. Winick NJ,
    7. et al.
    Genome-wide study of methotrexate clearance replicates SLCO1B1. Blood 2013;121:898–904.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    1. Wang WY,
    2. Barratt BJ,
    3. Clayton DG,
    4. Todd JA
    . Genome-wide association studies: theoretical and practical concerns. Nat Rev Genet 2005;6:109–18.
    OpenUrlCrossRefPubMed
  52. 52.↵
    PGRN-RIKEN International Collaborative Studies. Available from: http://bts.ucsf.edu/pgrn-cgm/.
  53. 53.↵
    1. Hertz DL,
    2. McLeod HL
    . Using pharmacogene polymorphism panels to detect germline pharmacodynamic markers in oncology. Clin Cancer Res 2014;20:2530–40.
    OpenUrlAbstract/FREE Full Text
  54. 54.↵
    1. Wheeler HE,
    2. Gamazon ER,
    3. Wing C,
    4. Njiaju UO,
    5. Njoku C,
    6. Baldwin RM,
    7. et al.
    Integration of cell line and clinical trial genome-wide analyses supports a polygenic architecture of paclitaxel-induced sensory peripheral neuropathy. Clin Cancer Res 2013;19:491–9.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Clinical Cancer Research: 20 (10)
May 2014
Volume 20, Issue 10
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Genome-Wide Association Study: A Useful Tool to Identify Common Genetic Variants Associated with Drug Toxicity and Efficacy in Cancer Pharmacogenomics
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Genome-Wide Association Study: A Useful Tool to Identify Common Genetic Variants Associated with Drug Toxicity and Efficacy in Cancer Pharmacogenomics
Siew-Kee Low, Atsushi Takahashi, Taisei Mushiroda and Michiaki Kubo
Clin Cancer Res May 15 2014 (20) (10) 2541-2552; DOI: 10.1158/1078-0432.CCR-13-2755

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Genome-Wide Association Study: A Useful Tool to Identify Common Genetic Variants Associated with Drug Toxicity and Efficacy in Cancer Pharmacogenomics
Siew-Kee Low, Atsushi Takahashi, Taisei Mushiroda and Michiaki Kubo
Clin Cancer Res May 15 2014 (20) (10) 2541-2552; DOI: 10.1158/1078-0432.CCR-13-2755
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Successful Pharmacogenomics Studies Using GWAS
    • Use of GWAS in Pharmacogenomics Studies for Individual Cancer Types
    • Lessons, Challenges, and Future Directions in Cancer Pharmacogenomics Studies Using GWAS
    • Conclusions
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Endpoints for Immuno-oncology Trials
  • Limitations and Challenges in Immuno-oncology Trials
  • Developing Early-Phase Combination Immunotherapy Trials
Show more CCR Focus
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement