Skip to main content
Cancer Science logoLink to Cancer Science
. 2013 Jun 10;104(8):1074–1082. doi: 10.1111/cas.12186

Genome‐wide association study of chemotherapeutic agent‐induced severe neutropenia/leucopenia for patients in Biobank Japan

Siew‐Kee Low 1,4, Suyoun Chung 4,5, Atsushi Takahashi 1, Hitoshi Zembutsu 4, Taisei Mushiroda 2, Michiaki Kubo 3, Yusuke Nakamura 4,5,
PMCID: PMC7657179  PMID: 23648065

Abstract

Chemotherapeutic agents are notoriously known to have a narrow therapeutic range that often results in life‐threatening toxicity. Hence, it is clinically important to identify the patients who are at high risk for severe toxicity to certain chemotherapy through a pharmacogenomics approach. In this study, we carried out multiple genome‐wide association studies (GWAS) of 13 122 cancer patients who received different chemotherapy regimens, including cyclophosphamide‐ and platinum‐based (cisplatin and carboplatin), anthracycline‐based (doxorubicin and epirubicin), and antimetabolite‐based (5‐fluorouracil and gemcitabine) treatment, antimicrotubule agents (paclitaxel and docetaxel), and topoisomerase inhibitors (camptothecin and etoposide), as well as combination therapy with paclitaxel and carboplatin, to identify genetic variants that are associated with the risk of severe neutropenia/leucopenia in the Japanese population. In addition, we used a weighted genetic risk scoring system to evaluate the cumulative effects of the suggestive genetic variants identified from GWAS in order to predict the risk levels of individuals who carry multiple risk alleles. Although we failed to identify genetic variants that surpassed the genome‐wide significance level (P < 5.0 × 10−8) through GWAS, probably due to insufficient statistical power and complex clinical features, we were able to shortlist some of the suggestive associated loci. The current study is at the relatively preliminary stage, but does highlight the complexity and problematic issues associated with retrospective pharmacogenomics studies. However, we hope that verification of these genetic variants through local and international collaborations could improve the clinical outcome for cancer patients.


It is now widely and well recognized that medication can cause distinct heterogeneity in terms of its efficacy and toxicity among individuals. These inter‐individual differences could be explained in part by the common and/or rare genetic variants in the human genome. Pharmacogenomics aims to discover how genetic variations in the human genome can affect a drug's efficacy or toxicity, and thus brings great promise for personalized medicine in which genetic information can be used to predict the safety, toxicity, and/or efficacy of drugs.1 Pharmacogenomics study for chemotherapeutic therapies is particularly important because these drugs are known to have a narrow therapeutic window; in general, a higher concentration causes toxicity and a lower concentration reduces the efficacy of the drug. Two of the well‐described examples are the association of genetic variants in TPMT with 6‐mercaptopurine‐induced myelosuppression in treatment of pediatric acute lymphoblastic leukemia and that of UGT1A1 variants with camptothecin‐related neutropenia and diarrhea in treatment of colorectal and lung cancers. The US Food and Drug Administration have recommended that variants on these two genes should be helpful for the prediction of severe adverse reactions prior to use of the drugs.2, 3, 4, 5, 6, 7

With advances in various technologies in the life sciences, it is now possible to accurately genotype more than a million common genetic variations by genome‐wide high‐density SNP array or to characterize all genetic variants in our genome by the next generation DNA sequencing methods. Although one of the greatest drawbacks of GWAS is the requirement of the large number of samples to achieve high statistical power,8 this issue could be overcome by the establishment of Biobank Japan in 2003 (http://biobankjp.org/).9 Biobank Japan collected approximately 330 000 disease cases (200 000 individuals) that had either one or multiples of 47 different diseases including cancers from a collaborative network of 66 hospitals throughout Japan, with the major aim to identify genetic variants associated with susceptibility to complex diseases or those related to drug toxicity. By using the samples from Biobank Japan, a significant number of insightful findings have been published in recent years for identification of common genetic variants associated with complex diseases including cancer.10, 11, 12, 13, 14, 15, 16, 17, 18, 19 With a reasonable number of samples, it is also feasible to carry out pharmacogenomics studies on chemotherapy‐induced toxicity.

Neutropenia and/or leucopenia are two of the most common drug adverse events after treatment with chemotherapeutic agents, which often cause life‐threatening infections and the delay of treatment schedule that subsequently affect the treatment outcome. Although prophylactic granulocyte colony‐stimulating factor has been given to the patients as a preventive measure,20 the underlying mechanism and susceptible risk factors that cause neutropenia have not been fully elucidated. In this study, we carried out a total of 17 sets of GWAS using 13 122 cancer patients, who received various drug regimens, to identify genetic variants associated with the risk of chemotherapeutic agent‐induced severe neutropenia/leucopenia in the Japanese population.

Subjects and Method

Study subjects

A total of 13 122 DNA samples from cancer patients, who received various chemotherapeutic agents, stored in Biobank Japan (University of Tokyo, Tokyo, Japan), were used in this study. Among them, 805 patients developed severe neutropenia and/or leucopenia (≥grade 3), and 4804 patients were not reported to develop any adverse reactions after being given chemotherapeutic agents. The samples could be classified into subgroups according to the drugs used: an alkylating agent (cyclophosphamide); platinum‐based (cisplatin and carboplatin), anthracycline‐based (doxorubicin and epirubicin); antimetabolite‐based (5‐fluorouracil and gemcitabine), antimicrotubule‐based (paclitaxel and docetaxel); and topoisomerase inhibitor‐based (camptothecin and etoposide). The grade of toxicity was classified in accordance with the US National Cancer Institute's Common Toxicity Criteria version 2.0. The adverse event description is based on the medical records collected by the medical coordinator. The patients' demographic details are summarized in Table 1. Participants of this study provided written inform consent and this project was approved by the ethical committee from the Institute of Medical Sciences, University of Tokyo and the RIKEN Center for Genomic Medicine (Yokohama, Japan).

Table 1.

Demographic details of cancer patients treated with chemotherapeutic agents, whose DNA samples are stored in Biobank Japan (The University of Tokyo, Tokyo, Japan)

Category Controlsa Grade 1/2 Grade 3/4 Category Controlsa Grade 1/2 Grade 3/4
All 4804 1253 805 Drug subtype
Age, years (mean) 62.9 58.7 59.6 Alkylating agent 346 266 176
Gender Cyclophosphamide 335 255 168
Male 2604 424 318 Platinum‐based 743 429 428
Female 2200 829 487 Cisplatin 471 191 176
Cancer subtype Carboplatin 262 207 261
Lung cancer 587 259 266 Anthracycline 459 240 184
Breast cancer 876 388 204 Doxorubicin 66 85 83
Ovarian cancer 140 124 74 Epirubicin 370 132 83
Gastric cancer 827 100 56 Antimetabolite 2249 512 294
Esophageal cancer 208 65 53 5‐Fluorouracil 952 331 177
Colorectal cancer 1573 161 50 Gemcitabine 226 111 80
Endometrial cancer 78 72 45 Antimicrotubule agent 825 468 371
Cervical cancer 129 57 35 Paclitaxel 364 321 218
Prostate cancer 91 13 21 Docetaxel 233 143 147
Pancreatic cancer 83 36 20 Topoisomerase inhibitor 187 123 106
Liver cancer 366 16 9 Camptothecin 155 106 59
Gallbladder cancer 56 9 1 Etoposide 39 19 54
Paclitaxel + carboplatin 166 161 150
a

Individuals who did not develop any adverse drug reactions after chemotherapy.

Genotyping and quality controls

DNAs obtained from the patients' blood were genotyped using Illumina OmniExpress BeadChip (San Diego, CA, USA) that contained 733 202 SNPs. Sample quality control was carried out by methods including identity‐by‐state to evaluate cryptic relatedness for each sample and population stratification by the use of principal component analysis to exclude genetically heterogeneous samples from further analysis.21, 22 Then, our standard SNP quality control was carried out by excluding SNPs deviating from the Hardy–Weinberg equilibrium (P ≤ 1.0 × 10−6), non‐polymorphic SNPs, SNPs with a call rate of <0.99, and those on the X chromosome.21, 22 Q–Q plot and lambda values, which were calculated between observed P‐values from Fisher's test allelic model against expected P‐values, were used to further evaluate population substructure.

Statistical analysis

Genome‐wide case–control association analyses were evaluated using Fisher's exact method considering allelic, dominant, and recessive genetic models. Manhattan plots of the study were generated using the minimum P‐value among the three genetic models for each SNP.

Scoring system using wGRS

The scoring analysis was carried out using SNPs with Pmin of <1.0 × 10−5 after exclusion of SNPs that are in strong linkage disequilibrium (r 2 > 0.8) in each GWAS. The wGRS were calculated according to De Jager et al.23 Briefly, we first calculated the weight of each SNP that is the natural log of the odds ratio for each allele/genotype, considering the associated genetic model. For an additive model, we assigned a score of 2 to an individual with two risk alleles, 1 to that with one risk allele, and 0 to that with no risk allele. For a dominant model, we assigned a score of 1 to an individual with one or two risk alleles, and 0 to that with no risk allele. For a recessive model, we assigned a score of 1 to an individual with two risk alleles, and 0 to that with no or one risk allele. Then the cumulative genetic risk scores were determined by multiplying the number of risk alleles/genotype of each SNP by its corresponding weight, and subsequently took the sum across the total number of SNPs that were taken into consideration of each GWAS set. We classified the genetics risk score into four different groups created from the mean and SD: group 1, <mean − 1SD; group 2, mean − 1SD to mean; group 3, mean to mean + 1SD; and group 4, >mean + 1SD. Odds ratio, 95% confidence interval, P‐value, sensitivity, and specificity were calculated using group 1 as a reference. To calculate the OR in which one of the cells in the contingency table is zero, we applied the Haldane correction, used to avoid error in the calculation by adding 0.5 to all of the cells of a contingency table.

Results

After subdividing the patients by administered drugs/major drug subgroups, as previously mentioned, a total of 17 GWAS analyses were carried out by comparing the allele/genotype frequency between the patients who had developed severe neutropenia/leucopenia (grade 3/4) to those who had not developed any adverse drug reactions. The Q–Q plots of each GWAS and the calculated lambda value of below 1.00 indicated no significant population stratification in each of these GWAS analyses (Fig. S1). From this study, although we could not identify any SNPs that surpassed the genome‐wide significant threshold (P‐value < 5 × 10−8) for showing association with the risk of neutropenia/leucopenia induced by the certain type of drug or regimen, several possible candidate loci were identified. The results of the GWAS are summarized in Table 2, Table S1, and Figure S2; the results of wGRS are summarized in Table S2.

Table 2.

Association analysis of single nucleotide polymorphisms (SNPs) with different chemotherapeutic drugs/drug subgroups known to induce severe neutropenia/leucopenia

CHR SNP BP RA NRA RAF_Case RAF_Ctrl P_allelic P_dom P_rec Pmin OR L95 U95 Gene rel.loci
Cyclophosphamide
16 rs2519974a 22889186 T C 0.503 0.381 2.52E‐04 4.35E‐06 2.77E‐01 4.35E‐06 1.647 1.264 2.146 HS3ST2 0
1 rs10922438a 198469162 T C 0.214 0.106 6.01E‐06 1.71E‐05 7.10E‐02 6.01E‐06 2.301 1.608 3.293 ATP6V1G3 23190
19 rs3745571a 6475613 T C 0.778 0.670 4.05E‐04 7.72E‐06 1.00E+00 7.72E‐06 1.730 1.276 2.345 DENND1C 0
All platinum‐based drugs
15 rs4886670a 75449674 A C 0.320 0.227 9.86E‐07 1.43E‐05 8.14E‐04 9.86E‐07 1.605 1.330 1.937 RPL36AP45 29318
19 rs33428a 30937843 G A 0.481 0.403 2.71E‐04 2.78E‐06 3.11E‐01 2.78E‐06 1.375 1.160 1.629 ZNF536 0
14 rs12589282a 22937656 G T 0.535 0.437 6.30E‐06 5.42E‐03 4.11E‐06 4.11E‐06 1.480 1.250 1.752 TRA@ 0
3 rs3845905a 66525963 G A 0.915 0.850 4.12E‐06 7.45E‐05 3.65E‐04 4.12E‐06 1.894 1.433 2.503 LRIG1 0
5 rs1895302a 169542600 C T 0.551 0.478 6.95E‐04 7.41E‐06 4.11E‐01 7.41E‐06 1.340 1.132 1.587 FOXI1 5871
1 rs16825455a 21837755 T C 0.686 0.605 8.85E‐05 8.62E‐06 1.90E‐01 8.62E‐06 1.425 1.193 1.702 ALPL 0
Cisplatin
7 rs10253216a 16861849 T C 0.565 0.468 2.18E‐03 1.68E‐07 1.00E+00 1.68E‐07 1.478 1.155 1.891 AGR2 −17111
4 rs11944965a 63424089 T C 0.807 0.678 3.45E‐06 1.68E‐06 6.65E‐02 1.68E‐06 1.986 1.475 2.676 LOC644534 47600
7 rs7797977a 16862235 C A 0.668 0.580 4.06E‐03 5.23E‐01 2.17E‐06 2.17E‐06 1.457 1.127 1.883 AGR2 −17497
18 rs2406342a 74488280 T G 0.605 0.475 3.59E‐05 2.48E‐06 6.71E‐02 2.48E‐06 1.697 1.323 2.177 ZNF236 −47836
20 rs6077251a 7752366 T C 0.271 0.153 2.50E‐06 3.64E‐06 3.06E‐02 2.50E‐06 2.065 1.537 2.773 SFRS13AP2 59982
8 rs11774576a 27740417 A G 0.702 0.581 6.78E‐05 2.82E‐06 2.62E‐01 2.82E‐06 1.699 1.307 2.208 SCARA5 0
11 rs4627050a 18822037 G A 0.781 0.649 4.43E‐06 9.01E‐06 2.17E‐02 4.43E‐06 1.932 1.452 2.572 PTPN5 −8648
1 rs12142335a 108302922 A G 0.040 0.004 9.91E‐06 8.45E‐06 1.00E+00 8.45E‐06 9.713 3.175 29.710 VAV3 0
Carboplatin
15 rs11071200a 55950082 T G 0.060 0.008 1.25E‐06 8.51E‐07 1.00E+00 8.51E‐07 8.241 2.888 23.520 PRTG 0
5 rs3822735a 35799994 G A 0.862 0.752 7.24E‐06 1.68E‐06 3.50E‐01 1.68E‐06 2.062 1.500 2.834 SPEF2 0
3 rs1623879a 58027197 G A 0.441 0.321 7.69E‐05 1.89E‐02 3.75E‐06 3.75E‐06 1.669 1.297 2.148 FLNB 0
15 rs936229a 75132319 G A 0.713 0.595 7.27E‐05 4.41E‐06 3.01E‐01 4.41E‐06 1.685 1.302 2.180 ULK3 0
13 rs7989332a 21050575 G T 0.853 0.738 5.47E‐06 1.16E‐04 1.74E‐03 5.47E‐06 2.056 1.507 2.806 CRYL1 0
3 rs3845905a 66525963 G A 0.921 0.828 5.99E‐06 2.20E‐05 1.50E‐02 5.99E‐06 2.433 1.645 3.598 LRIG1 0
8 rs1714746a 4105147 G A 0.554 0.435 1.59E‐04 6.63E‐02 7.44E‐06 7.44E‐06 1.610 1.261 2.056 CSMD1 0
16 rs12446319a 81774798 A G 0.253 0.143 8.97E‐06 1.27E‐04 3.16E‐04 8.97E‐06 2.026 1.480 2.774 CMIP 29431
1 rs1277203a 109392837 A G 0.730 0.626 3.50E‐04 3.51E‐02 9.38E‐06 9.38E‐06 1.615 1.243 2.098 AKNAD1 0
All anthracycline‐based drugs
5 rs10040979a 158424391 G A 0.701 0.618 4.68E‐03 5.35E‐01 4.60E‐07 4.60E‐07 1.452 1.120 1.883 EBF1 0
2 rs12615435a 200638509 T G 0.883 0.773 3.95E‐06 4.09E‐06 9.17E‐02 3.95E‐06 2.214 1.555 3.154 LOC348751 0
5 rs7720283a 158459721 C T 0.775 0.706 1.29E‐02 3.37E‐01 4.15E‐06 4.15E‐06 1.431 1.078 1.898 EBF1 0
1 rs1367448a 68633924 C T 0.633 0.526 5.02E‐04 5.32E‐06 5.12E‐01 5.32E‐06 1.554 1.212 1.993 LOC100289178 0
6 rs2505059a 98495952 G A 0.538 0.398 5.41E‐06 2.29E‐04 2.04E‐04 5.41E‐06 1.765 1.383 2.252 MIR2113 23455
12 rs4149639a 6442001 C T 0.120 0.047 7.39E‐06 1.52E‐05 8.16E‐02 7.39E‐06 2.763 1.781 4.287 TNFRSF1A 0
19 rs1654260a 20329111 A G 0.625 0.488 8.49E‐06 3.03E‐03 2.18E‐05 8.49E‐06 1.749 1.365 2.240 LOC100421704 −3576
Doxorubicin
15 rs11857176a 78164706 A G 0.657 0.515 1.74E‐02 1.00E+00 8.08E‐07 8.08E‐07 1.800 1.127 2.874 LOC100302666 −6274
2 rs4380275a 773278 G A 0.392 0.152 4.99E‐06 1.54E‐05 2.05E‐02 4.99E‐06 3.604 2.041 6.365 LOC339822 6559
11 rs2512987a 86414282 T C 0.681 0.417 7.02E‐06 6.04E‐03 3.42E‐05 7.02E‐06 2.985 1.855 4.803 ME3 −30604
Epirubicin
12 rs4149639a 6442001 C T 0.163 0.042 2.89E‐07 8.31E‐07 3.32E‐02 2.89E‐07 4.443 2.571 7.677 TNFRSF1A 0
5 rs2964475a 5407814 C A 0.615 0.415 4.13E‐06 3.95E‐04 1.10E‐04 4.13E‐06 2.248 1.592 3.174 KIAA0947 −14993
13 rs1923834a 28360487 G A 0.916 0.770 9.40E‐06 4.61E‐06 3.33E‐01 4.61E‐06 3.236 1.823 5.744 GSX1 −6293
10 rs908366a 126144839 A G 0.518 0.328 7.04E‐06 1.08E‐04 8.69E‐04 7.04E‐06 2.199 1.564 3.092 LHPP −5502
3 rs1553091a 187716886 G A 0.452 0.358 2.65E‐02 8.04E‐01 7.46E‐06 7.46E‐06 1.480 1.053 2.080 LOC100505844 −22691
All antimetabolite drugs
18 rs7228133a 4539085 C A 0.733 0.686 2.26E‐02 6.64E‐01 1.70E‐06 1.70E‐06 1.255 1.034 1.522 LOC284215 243085
21 rs8127977a 26826514 A G 0.804 0.722 1.30E‐05 2.11E‐06 2.67E‐01 2.11E‐06 1.587 1.282 1.966 NCRNA00158 −22501
12 rs894734a 54319727 G A 0.849 0.776 3.84E‐05 3.97E‐06 6.63E‐01 3.97E‐06 1.619 1.279 2.050 HOXC13 −12849
13 rs9580312a 22754093 G A 0.480 0.409 1.35E‐03 8.09E‐06 6.25E‐01 8.09E‐06 1.330 1.120 1.581 LOC100506622 −30331
21 rs2055011a 19481354 C T 0.184 0.143 1.12E‐02 2.12E‐01 8.82E‐06 8.82E‐06 1.347 1.075 1.686 CHODL −135796
12 rs12582168a 124894184 C T 0.333 0.256 8.50E‐05 9.31E‐06 2.84E‐01 9.31E‐06 1.454 1.210 1.748 NCOR2 0
5‐Fluorouracil
7 rs10488226a 12713070 A C 0.195 0.107 1.09E‐05 3.54E‐06 2.98E‐01 3.54E‐06 2.026 1.500 2.737 LOC100505995 −12175
2 rs6740660a 224943685 G A 0.966 0.894 4.10E‐06 8.83E‐06 2.40E‐01 4.10E‐06 3.386 1.870 6.131 SERPINE2 −39649
4 rs1567482a 36026747 G A 0.952 0.875 6.26E‐06 1.44E‐05 9.14E‐02 6.26E‐06 2.846 1.716 4.719 LOC651644 39948
2 rs6706693a 192465598 A G 0.328 0.219 1.62E‐05 2.12E‐03 9.45E‐06 9.45E‐06 1.743 1.362 2.232 OBFC2A −77200
Gemcitabine
18 rs9961113a 75605399 C T 0.625 0.403 1.43E‐06 3.83E‐04 3.73E‐05 1.43E‐06 2.473 1.706 3.584 LOC100421527 −260017
5 rs2547917a 58713680 A G 0.350 0.212 9.06E‐04 8.79E‐02 3.33E‐06 3.33E‐06 1.997 1.345 2.965 PDE4D 0
15 rs12900463a 85415386 C T 0.219 0.115 2.24E‐03 1.02E‐01 4.03E‐06 4.03E‐06 2.154 1.342 3.457 ALPK3 0
22 rs9609078a 31153276 T C 0.089 0.009 4.32E‐06 9.97E‐06 2.59E‐01 4.32E‐06 10.890 3.528 33.610 OSBP2 0
5 rs6863418a 173625154 A G 0.175 0.055 1.37E‐05 6.98E‐06 4.55E‐01 6.98E‐06 3.623 2.042 6.429 HMP19 88972
20 rs6037430a 344079 G A 0.894 0.730 9.74E‐06 1.75E‐05 7.92E‐02 9.74E‐06 3.109 1.805 5.359 NRSN2 8567
All antimicrotubule drugs
17 rs11651483a 12777402 C T 0.729 0.665 1.69E‐03 2.60E‐01 3.37E‐07 3.37E‐07 1.357 1.120 1.643 RICH2 0
6 rs4235898a 77266188 A G 0.830 0.740 1.05E‐06 3.34E‐06 6.50E‐03 1.05E‐06 1.718 1.377 2.142 LOC100131680 −103976
13 rs4771859a 93088651 G A 0.764 0.709 5.49E‐03 2.60E‐01 1.47E‐06 1.47E‐06 1.328 1.088 1.623 GPC5 0
1 rs12145418a 216716320 T G 0.334 0.274 3.04E‐03 3.17E‐01 2.35E‐06 2.35E‐06 1.331 1.104 1.604 ESRRG 0
6 rs9386485a 106329055 T C 0.596 0.492 2.65E‐06 2.85E‐05 8.59E‐04 2.65E‐06 1.524 1.279 1.817 PRDM1 −205140
16 rs12935229a 77328895 A G 0.249 0.182 1.83E‐04 2.23E‐02 4.40E‐06 4.40E‐06 1.495 1.214 1.840 ADAMTS18 0
7 rs6961860a 17085321 G A 0.557 0.495 5.33E‐03 8.87E‐01 4.67E‐06 4.67E‐06 1.283 1.078 1.527 LOC100131425 156806
14 rs12882718a 86902054 T C 0.737 0.643 5.91E‐06 5.55E‐06 2.03E‐02 5.55E‐06 1.555 1.283 1.884 LOC100421119 −42891
12 rs1043763a 122630909 T C 0.668 0.574 1.36E‐05 6.51E‐06 3.10E‐02 6.51E‐06 1.496 1.248 1.795 MLXIP 1920
2 rs4591358a 196365890 C T 0.302 0.215 6.60E‐06 6.89E‐04 1.89E‐04 6.60E‐06 1.578 1.297 1.920 LOC391470 81627
14 rs8022296a 97987857 G A 0.663 0.608 1.06E‐02 6.54E‐01 7.29E‐06 7.29E‐06 1.269 1.058 1.521 LOC100129345 111127
4 rs6817170a 154374984 G A 0.377 0.286 9.29E‐06 4.61E‐05 3.27E‐03 9.29E‐06 1.517 1.264 1.822 KIAA0922 −12514
Paclitaxel
1 rs922106a 90025519 T G 0.298 0.202 2.17E‐04 1.95E‐02 9.28E‐07 9.28E‐07 1.679 1.277 2.207 LRRC8B 0
6 rs9386485a 106329055 T C 0.624 0.477 1.17E‐06 1.43E‐05 5.24E‐04 1.17E‐06 1.821 1.429 2.320 PRDM1 −205140
8 rs2444896a 99022009 T G 0.727 0.603 1.58E‐05 2.43E‐06 7.41E‐02 2.43E‐06 1.754 1.355 2.269 MATN2 0
2 rs4666360a 20335709 C T 0.216 0.114 4.62E‐06 3.24E‐06 2.27E‐01 3.24E‐06 2.136 1.546 2.950 RPS16P2 19625
9 rs3138083a 35648950 A G 0.220 0.117 3.78E‐06 1.24E‐05 1.09E‐02 3.78E‐06 2.136 1.551 2.942 SIT1 345
17 rs3786094a 9875205 C T 0.528 0.422 5.30E‐04 1.80E‐01 5.83E‐06 5.83E‐06 1.531 1.206 1.944 GAS7 0
15 rs4886670a 75449674 A C 0.353 0.229 7.26E‐06 5.38E‐05 4.26E‐03 7.26E‐06 1.835 1.412 2.383 RPL36AP45 29318
5 rs792975a 172271007 T C 0.654 0.519 7.66E‐06 1.19E‐04 6.08E‐04 7.66E‐06 1.746 1.366 2.232 ERGIC1 0
Docetaxel
9 rs3747851a 124521260 T C 0.337 0.176 5.61E‐07 1.12E‐05 5.63E‐04 5.61E‐07 2.377 1.693 3.339 DAB2IP 0
7 rs4727963a 122759980 C T 0.772 0.618 7.99E‐06 1.04E‐06 1.69E‐01 1.04E‐06 2.094 1.505 2.914 SLC13A1 0
14 rs1756650a 87741025 G A 0.211 0.162 9.95E‐02 9.10E‐01 1.74E‐06 1.74E‐06 1.386 0.954 2.014 GALC 658333
13 rs488248a 106596719 T C 0.918 0.795 3.29E‐06 3.23E‐05 1.17E‐02 3.29E‐06 2.896 1.802 4.655 LOC728192 −432192
6 rs12660691a 130008445 A C 0.935 0.819 3.62E‐06 4.99E‐06 1.62E‐01 3.62E‐06 3.199 1.899 5.391 ARHGAP18 0
18 rs4553720a 62170726 T C 0.377 0.281 7.77E‐03 4.56E‐01 6.77E‐06 6.77E‐06 1.547 1.131 2.116 LOC284294 79890
6 rs2157460a 130021128 T C 0.932 0.820 7.07E‐06 1.07E‐05 1.62E‐01 7.07E‐06 3.013 1.806 5.025 ARHGAP18 0
2 rs837841a 130034012 T G 0.714 0.662 1.49E‐01 5.26E‐01 8.34E‐06 8.34E‐06 1.279 0.930 1.759 LOC151121 −33653
All topoisomerase inhibitors
5 rs10074959a 104208013 T C 0.321 0.237 3.23E‐02 8.08E‐01 1.13E‐06 1.13E‐06 1.524 1.048 2.217 RAB9P1 −227162
7 rs1035147a 12094966 T G 0.981 0.877 4.01E‐06 4.75E‐06 5.56E‐01 4.01E‐06 7.294 2.587 20.559 TMEM106B −155882
1 rs303386a 99589379 A G 0.585 0.444 1.10E‐03 4.30E‐06 3.88E‐01 4.30E‐06 1.766 1.256 2.483 LOC100129620 0
14 rs7494275a 56231800 C A 0.543 0.406 1.86E‐03 4.26E‐01 8.50E‐06 8.50E‐06 1.732 1.232 2.433 RPL13AP3 −1163
3 rs480409a 7010081 G A 0.495 0.348 6.08E‐04 1.63E‐01 9.28E‐06 9.28E‐06 1.842 1.307 2.596 GRM7 0
Camptothecin
6 rs17318866a 3837198 G A 0.966 0.790 1.47E‐06 1.78E‐06 1.91E‐01 1.47E‐06 7.559 2.689 21.263 FAM50B −12434
2 rs17027130a 41273631 C T 0.644 0.387 2.54E‐06 8.03E‐04 3.91E‐05 2.54E‐06 2.865 1.844 4.452 LOC729984 −110074
1 rs303386a 99589379 A G 0.627 0.429 3.34E‐04 3.61E‐06 1.06E‐01 3.61E‐06 2.238 1.448 3.460 LOC100129620 0
Etoposide
20 rs6039763a 10183517 A G 0.370 0.090 1.27E‐05 1.54E‐06 4.61E‐01 1.54E‐06 5.966 2.502 14.230 LOC100131208 0
1 rs2506991a 48098406 G A 0.593 0.269 1.39E‐05 1.11E‐02 2.28E‐06 2.28E‐06 3.948 2.101 7.418 LOC388630 127794
2 rs12987465a 49715021 A G 0.593 0.359 1.87E‐03 6.26E‐01 6.61E‐06 6.61E‐06 2.597 1.424 4.737 FSHR –333355
7 rs3095008a 20255705 T C 1.000 0.846 1.74E‐05 9.39E‐06 1.00E+00 9.39E‐06 inf N/A N/A MACC1 0
Paclitaxel + carboplatin
12 rs12310399a 95490248 A G 0.708 0.567 2.60E‐04 1.08E‐01 2.46E‐07 2.46E‐07 1.852 1.329 2.580 FGD6 0
9 rs10785877a 137125501 T C 0.833 0.660 7.38E‐07 1.54E‐05 1.98E‐04 7.38E‐07 2.580 1.766 3.769 RXRA ‐92815
19 rs995834a 28866596 C T 0.580 0.425 1.28E‐04 1.20E‐06 1.51E‐01 1.20E‐06 1.871 1.364 2.566 LOC100420587 307385
1 rs922107a 90022796 G A 0.323 0.211 1.55E‐03 9.07E‐02 2.73E‐06 2.73E‐06 1.788 1.250 2.558 LRRC8B 0
7 rs1425132a 37562368 T C 0.740 0.666 4.55E‐02 7.35E‐01 4.68E‐06 4.68E‐06 1.430 1.013 2.017 LOC442668 62349
1 rs6429703a 15339960 T C 0.200 0.078 8.40E‐06 2.59E‐05 5.61E‐02 8.40E‐06 2.942 1.802 4.804 RP1‐21O18.1 0
a

SNPs used for weighted genetic risk score analyses. BP, SNP genomic location; CHR, chromosome; inf, infinity; L95, lower 95% confidence interval; N/A, not applicable; NRA, non‐risk allele; OR, odds ratio; P_allelic, P‐value from allelic model; P_dom, P‐value from dominant model; P_min, minimum P‐value among the three models; P_rec, P‐value from recessive model; RA, risk allele; RAF, risk allele frequency; rel.loci, distance of the SNP from the gene; U95, upper 95% confidence interval.

Among these datasets, GWAS carried out using samples who were given: (i) any kind of platinum‐based chemotherapy (428 cases vs 743 controls); (ii) cisplatin‐based chemotherapy (176 cases vs 471 controls); or (iii) carboplatin‐based chemotherapy (261 cases vs 262 controls) identified SNPs showing the most significant association with chemotherapy‐induced severe neutropenia/leucopenia are: rs4886670 (P min 9.86 × 10−7, OR = 1.61, 95% CI = 1.33–1.94) near RPL36AP45 for (i); rs10253216 (P min 1.68 × 10−7, OR = 1.48, 95% CI = 1.16–1.89) near AGR2 for (ii); and rs11071200 (P min 8.51 × 10−7, OR = 8.24, 95% CI = 2.89–23.5) on PRTG for (iii) (Table 2, Table S1, Fig. S2b). For the anthracycline‐based regimen, we carried out GWAS with individuals given all anthracycline‐based (184 cases vs 459 controls), doxorubicin‐based (83 cases vs 66 controls), and epirubicin‐based (83 cases vs 370 controls) chemotherapy, and identified three SNPs, rs10040979 (P min 4.60 × 10−7, OR = 1.45, 95% CI = 1.12–1.88) in EBF1, rs11857176 (P min 8.08 × 10−7, OR = 1.80, 95% CI = 1.13–2.87) near a hypothetical gene LOC100302666, and rs4149639 (P min 2.89 × 10−7, OR = 4.44, 95% CI = 2.57–7.68) in TNFRSF1A, to be most significantly associated with the risk of high‐grade neutropenia/leucopenia, respectively (Table 2, Table S1, Fig. S2c). In the case of antimicrotubule agents, we carried out three different GWAS with individuals who were treated with antimicrotubule (371 cases vs 825 controls), paclitaxel‐based (218 cases vs 364 controls), or docetaxel‐based (147 cases vs 233 controls) regimens. We identified three SNPs, rs11651483 (P min 3.37 × 10−7, OR = 1.36, 95% CI = 1.12–1.64) in RICH2, rs922106 (P min 9.28 × 10−7, OR = 1.68, 95% CI = 1.28–2.21) in LRRC8B and rs3747851 (P min 5.61 × 10−7, OR = 2.38, 95% CI = 1.69–3.34) in DAB2IP, to be those most significantly associated with the increased risk of severe neutropenia/leucopenia, respectively (Table 2, Table S1, Fig. S2e). Our previous report by Kiyotani et al.24 identified four SNPs to be associated with gemcitabine‐induced hematological toxicities. Three of the four SNPs were included in the current study with suggestive association, rs12046844 (P min 5.84 × 10−4, OR = 2.53, 95% CI = 1.45–4.43), rs6430443 (P min 8.61 × 10−4, OR = 6.33, 95% CI = 1.90–22.2; r 2 = 0.895 with rs1901440) and rs11719165 (P min 1.16 × 10−2, OR = 2.36, 95% CI = 1.18–4.70) (Table S4). However, it is noted that some of the samples used in this study overlapped with those in the study reported by Kiyotani et al., as both sourced samples from Biobank Japan.

Lastly, we also attempted to identify genetic variants associated with combined treatment of paclitaxel and carboplatin‐induced severe neutropenia/leucopenia (150 cases vs 166 controls), as this combined treatment is commonly used as the standard therapy for both ovarian and lung cancers. We found the most significant association with the SNP rs12310399 (P min 2.46 × 10−7, OR = 1.85, 95% CI = 1.33–2.58) near the FGD6 gene (Table 2, Table S1, Fig. S2a), which is suggested to activate CDC42, a member of the Ras‐like family of Rho and Rac proteins, and has a critical role in regulating the actin cytoskeleton. The second strongest association was observed at the locus encoding RXRA (P min 7.38 × 10−7, OR = 2.58, 95% CI = 1.77–3.77), an important transcriptional factor. We also calculated the cumulative genetic scores using SNPs on six loci and identified that individuals in group 4 could have 188 times (95% CI = 36.1–979) higher risk of developing severe neutropenia/leucopenia than those belonging to group 1 with the sensitivity of 95.9% and the specificity of 88.9% (Table S2). Because this drug combination is of clinical importance, we further investigated the association of these six selected loci using 161 individuals who developed grade 1/2 neutropenia/leucopenia, using cases registered in the Biobank Japan. Interestingly, the association results for the six loci were moderate for grade 1/2 neutropenia/leucopenia, with intermediate allele frequency and OR between individuals without any adverse reactions and those with neutropenia/leucopenia of ≥grade 3 (Table S3). In addition, as shown in Table 3 and Figure 1, the higher the calculated score becomes, the higher the proportion and grade of neutropenia/leucopenia. The intermediate scores for patients with grade 1/2 neutropenia/leucopenia could imply the possible usefulness of this scoring system for the prediction.

Table 3.

Weighted genetic risk score (wGRS) analysis of cancer patients who received combination treatment with paclitaxel and carboplatin

wGRS group Score G3G4 G1G2 G0 %_G3G4 %_G1G2 %_G0 G3/4 versus G0 G1/2 versus G0
OR 95%_CI P‐value OR 95%_CI P‐value
1 <5.802 2 21 48 0.03 0.29 0.68 REF REF
2 5.802–7.665 36 58 77 0.21 0.34 0.45 11.2 2.58–48.70 8.69E‐05 1.72 0.93–3.19 9.55E‐02
3 7.665–9.528 64 62 33 0.40 0.39 0.21 46.5 10.60–204.00 2.36E‐13 4.29 2.21–8.35 1.61E‐05
4 >9.528 47 20 6 0.64 0.28 0.08 188.0 36.10–979.00 4.78E‐20 7.62 2.68–21.70 6.08E‐05
Total 149 161 164

95%_CI, 95% confidence interval; G0, individuals who did not develop any adverse drug reaction; G1G2, grade 1 and grade 2 neutropenia (mild); G3G4, grade 3 and grade 4 neutropenia (severe); OR, odds ratio; REF, reference.

Figure 1.

Figure 1

Proportions of cancer patients who developed no adverse reaction (G0), mild neutropenia/leucopenia (G1/2), or severe neutropenia/leucopenia (G3/4) in each of the weighted genetic risk score (wGRS) score groups. All patients received combined treatment with paclitaxel and carboplatin and were registered with Biobank Japan. The total numbers of patients in scores 1, 2, 3, and 4 are 71, 171, 159, and 73, respectively.

Furthermore, we used simulation to estimate how many samples are required to validate this scoring result. We started off by estimating the incidence of neutropenia/leucopenia by the combined treatment of paclitaxel and carboplatin. In Biobank Japan, a total of 477 individuals received this combined treatment; among them, 166 individuals (35%) did not develop any adverse drug reactions, 161 (35%) developed mild neutropenia/leucopenia (grade 1 or 2) and 150 (30%) developed severe neutropenia/leucopenia (grade 3 or higher). The frequency of developing severe neutropenia/leucopenia is in agreement with a multicenter study reported by Guastalla et al.25 When we assume that 100 patients who receive this combination therapy are prospectively registered, the incidences of the adverse drug reactions are estimated as shown in Table 4. If we categorize the patients by wGRS according to the proportions indicated in Table 3 (and our hypothesis is right), the statistical power should be enough to validate by this small subset of patients. Even if two individuals in both group 1 and group 4 are incorrectly predicted, the calculated P‐value is still 0.03 by Fisher's exact test.

Table 4.

Simulation of weighted genetic risk score (wGRS) analysis for a prospective study of 100 patients who received combination treatment with paclitaxel and carboplatin

Estimated verification samples (= 100; 35 expected to have grade 1/2 neutropenia)
wGRS group G3G4 G0 OR 95%_CI P‐value
1 0 10
2 7 17 9.0 0.47–174.00 7.82E‐02
3 13 7 37.8 1.93–740.00 1.06E‐03
4 10 1 147.0 5.35–4040.00 3.40E‐05
Total 30 35

95%_CI, 95% confidence interval; G0, individuals without any adverse drug reaction; G3G4, grade 3 and 4 neutropenia (severe); OR, odds ratio.

Discussion

In this study, we carried out GWAS analyses for a total of 17 subsets of chemotherapies to identify genetic variants that might be associated with chemotherapeutic‐induced neutropenia/leucopenia with grades 3 and 4, however, we could not identify any SNPs that surpassed the genome‐wide significant threshold (P‐value < 5 × 10−8). Through this study, we encountered several important issues, which are now common problems in pharmacogenomics studies using retrospective clinical data, including confounding factors and heterogeneous treatments for individual patients (often given different combinations of drugs, different dosage of drugs, and different time‐periods of treatment), that increase the complexity of studies and generate various noises in the analyses, and diminished the statistical power in the case–control association studies. We understand that our current approach was not an ideal study design, but it is not easy to perfectly standardize therapy in the daily clinical practice of cancer treatment. There are several factors contributing to the variability in treatments: (i) there is some preference by doctors or by hospitals to select a particular regimen among the various recommended standard treatments; (ii) the modifications (adjustments) of the dosage or schedule according to the patient's conditions (performance status, results of laboratory tests, etc.); and (iii) although we have been collecting the clinical information, it is not perfect to collect complete clinical information in some hospitals, particularly those that do not use electronic medical records. One can say that this kind of study should be performed as a prospective design, however, due to the very rapid advances in the development of novel molecular‐targeted drugs and new regimens in the oncology area, the protocols have been and will be modified or improved. Hence, spending many years and a huge budget on a prospective study may result in a clinically useless outcome, because the results are unable to be applied due to the replacement of the study protocol with a new protocol, when the results of association studies are available. Nevertheless, retrospective pharmacogenomic studies could be improved by implementing electronic medical record systems that could include detailed descriptions of patients' conditions and their responses to various drugs.

Although we understand the pitfalls in study designs like our present study, we need to seek possible ways to identify candidate genetic variants that might contribute to improvement in the clinical management of cancer patients, including chemotherapy‐induced severe neutropenia/leucopenia. Nevertheless, some of the candidate genes that we identified are of interest, considering their known functions as well as their relations with drug actions. For example, the proto‐oncogene AGR2, whose genetic variants were suggested to associate with cisplatin‐induced neutropenia/leucopenia, encodes an anterior gradient 2 homolog (Xenopus laevis) that is known to play a critical role in cell migration, cell differentiation, and cell growth.26 Cells stably expressing AGR2 confer resistance to cisplatin in vivo, compared with control cells (empty vector) in a xenograft animal model.27 The second example is TNFRSF1A, suggested to be associated with anthracycline‐based and epirubicin‐induced neutropenia/leucopenia. This gene encodes TNFRSF1A, which is a major receptor for TNF‐α. The soluble TNFRSF1A level was found to be elevated after 1 month of anthracycline‐based chemotherapy.28 Additionally, both TNF‐α and TNFRSF1A are known to play a critical role in doxorubicin‐induced cardiotoxicity, in which doxorubicin stimulates an increase in circulating TNF and upregulates TNFRSF1A.29, 30 Furthermore, genetic variants on PDE4D, which encodes for phosphodiesterase 4D, cAMP‐specific, showed suggestive association with gemcitabine‐induced severe neutropenia/leucopenia. Ablation of PDE4D has been reported to impair the neutrophil function with altered chemotaxis ability and adhesion capability as well as to reduce neutrophil recruitment to the site of inflammation.31 Besides, genetic variants on RXRA identified to be associated with combined treatment of paclitaxel and carboplatin‐induced severe neutropenia/leucopenia, encodes retinoid X receptor alpha. Disruption of this gene in mouse models moderately alters lymphocyte proliferation and survival, and affects the T helper type1/type 2 balances.32 All of these genes might provide some important insights into the mechanism of various chemotherapy‐induced severe neutropenia/leucopenia, however, further validations are definitely essential.

As already described, the GWAS approach could provide a list of genetic variants that might be associated with complex phenotypes (drug responsiveness or drug‐induced adverse reactions) in pharmacogenomics studies. One of the clinically important aims for identification of the associated genetic variants is to establish a prediction model to identify individuals who are at risk of adverse reactions with certain drugs or protocols. In this study, we have applied the wGRS system, by which we could distinguish high‐risk patients from low‐risk individuals by counting the number of risk alleles of the suggestively‐associated SNPs in combination with estimating the effect size of each SNP. One of the best examples from this study was indicated by a scoring system using six candidate SNP loci that were identified through the GWAS of severe neutropenia/leucopenia caused by combination treatment of paclitaxel and carboplatin; among 53 individuals in the high‐risk group (group 4) by this scoring method, 47 (89%) revealed high‐grade neutropenia/leucopenia. In contrast, among 50 individuals in the low‐risk group (group 1), only 2 (4%) revealed high‐grade neutropenia/leucopenia, and the odds ratio to have the severe adverse reaction in individuals belonging to group 4 was calculated to be 188 times higher than those categorized to group 1 (Table 3). Interestingly, individuals who developed grade 1/2 (mild neutropenia/leucopenia) were found to show intermediate risk scores between patients with severe neutropenia/leucopenia and those without any adverse reactions. Hence, we suggest that wGRS is an applicable method to evaluate the clinical utility of possible variants with specific phenotypes. However, the data are preliminary and require verification by an independent test sample(s) before any definitive conclusions can be drawn. But, considering that the OR of the high‐risk group is very high, the number of samples required for the verification (if our hypothesis is right) is not so large. In fact, we have tried to simulate a prospective study design using a model of 100 patients according to the assumption that 35% individuals will not develop any adverse drug reactions, 35% individuals will develop mild neutropenia/leucopenia (grade 1/2), and 30% will develop severe neutropenia/leucopenia (grade 3/4). As shown in Table 4, the study of 100 patients should have very strong statistical power to verify. If this is verified, as we expect, it should improve the quality of lives of cancer patients and also contribute to reducing medical care costs by avoiding unnecessary adverse events. However, to achieve success in pharmacogenomics and personalized medicine, both local and international collaborative efforts are essential.

Disclosure Statement

The authors have no conflict of interest.

Abbreviations

GWAS

genome‐wide association study

OR

odds ratio

SD

standard deviation

SNP

single nucleotide polymorphism

TNFRSF1A

tumor necrosis factor receptor superfamily, member 1A

TNF‐α

tumor necrosis factor‐α

wGRS

weighted genetic risk score

Supporting information

Fig. S1. Quantile–quantile plots of 17 genome‐wide association studies of drug‐induced or drug subgroup‐induced severe neutropenia and their corresponding lambda (λ) value.

Fig. S2. (a) Manhattan plot for genome‐wide association study of cyclophosphamide and paclitaxel + carboplatin induced severe neutropenia/leucopenia. (b) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of platinum‐based agents, cisplatin, or carboplatin. (c) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of anthracycline‐based agents, doxorubicin, or epirubicin. (d) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of antimetabolite agents, 5‐fluorouracil, or gemcitabine. (e) Manhattan plot for genome‐wide association study of severe neutropenia induced by all types of antimicrotubule agents, paclitaxel, or docetaxel. (f) Manhattan plot for genome‐wide association study severe neutropenia/leucopenia induced by all types of topoisomerase inhibitors, camptothecin, or etoposide.

Table S1. Genome‐wide association study of each chemotherapy regimen with P < 1 × 10−4.

Table S2. Weighted genetic risk score of each genome‐wide association study of specific chemotherapeutic‐based induced severe neutropenia/leucopenia.

Table S3. Association study of cancer patients who do not develop any adverse drug reaction and those who developed neutropenia/leucopenia after being given combination treatment with paclitaxel and carboplatin.

Table S4. Association of previously reported SNPs that associated with gemcitabine induced hematological toxicity.

Acknowledgments

We would like to express our heartfelt gratitude to all the patients who participated in this study. We convey our sincere appreciation to Dr Teruhiko Yoshida and Dr Hiromi Sakamoto from the National Cancer Center Research Institute for their kind support. Our thanks also goes to the members of the laboratory for statistical analysis and the laboratory for the genotyping development from the Center for Genomic Medicine for their kind support and fruitful discussions. We would like to extend our gratitude to the staff of Biobank Japan for their outstanding assistance. This work was carried out as part of the Biobank Japan Project, supported by the Ministry of Education, Culture, Sports, Sciences and Technology, Japan. In addition, this project was supported by the JSPS postdoctoral fellowship.

(Cancer Sci 2013; 104: 1074–1082)

References

  • 1. Wheeler HE, Maitland ML, Dolan ME, Cox NJ, Ratain MJ. Cancer pharmacogenomics: strategies and challenges. Nat Rev Genet 2013; 14: 23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Relling MV, Hancock ML, Rivera GK et al Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S‐methyltransferase gene locus. J Natl Cancer Inst 1999; 91: 2001–8. [DOI] [PubMed] [Google Scholar]
  • 3. Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet 1980; 32: 651–62. [PMC free article] [PubMed] [Google Scholar]
  • 4. Relling MV, Klein TE. CPIC: clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther 2011; 89: 464–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Innocenti F, Undevia SD, Iyer L 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. [DOI] [PubMed] [Google Scholar]
  • 6. Iyer L, Das S, Janisch L et al UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2002; 2: 43–7. [DOI] [PubMed] [Google Scholar]
  • 7. Hoskins JM, Goldberg RM, Qu P, Ibrahim JG, McLeod HL. UGT1A1*28 genotype and irinotecan‐induced neutropenia: dose matters. J Natl Cancer Inst 2007; 99: 1290–5. [DOI] [PubMed] [Google Scholar]
  • 8. McCarthy MI, Abecasis GR, Cardon LR et al Genome‐wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet 2008; 9: 356–69. [DOI] [PubMed] [Google Scholar]
  • 9. Triendl R. Japan launches controversial Biobank project. Nat Med 2003; 9: 982. [DOI] [PubMed] [Google Scholar]
  • 10. Takata R, Akamatsu S, Kubo M et al Genome‐wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet 2010; 42: 751–4. [DOI] [PubMed] [Google Scholar]
  • 11. Akamatsu S, Takata R, Haiman CA et al Common variants at 11q12, 10q26 and 3p11.2 are associated with prostate cancer susceptibility in Japanese. Nat Genet 2012; 44: 426–9, S1. [DOI] [PubMed] [Google Scholar]
  • 12. Cui R, Kamatani Y, Takahashi A et al Functional variants in ADH1B and ALDH2 coupled with alcohol and smoking synergistically enhance esophageal cancer risk. Gastroenterology 2009; 137: 1768–75. [DOI] [PubMed] [Google Scholar]
  • 13. Cui R, Okada Y, Jang SG et al Common variant in 6q26‐q27 is associated with distal colon cancer in an Asian population. Gut 2011; 60: 799–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kumar V, Kato N, Urabe Y et al Genome‐wide association study identifies a susceptibility locus for HCV‐induced hepatocellular carcinoma. Nat Genet 2011; 43: 455–8. [DOI] [PubMed] [Google Scholar]
  • 15. Low SK, Kuchiba A, Zembutsu H et al Genome‐wide association study of pancreatic cancer in Japanese population. PLoS ONE 2010; 5: e11824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Elgazzar S, Zembutsu H, Takahashi A et al A genome‐wide association study identifies a genetic variant in the SIAH2 locus associated with hormonal receptor‐positive breast cancer in Japanese. J Hum Genet 2012; 57: 766–71. [DOI] [PubMed] [Google Scholar]
  • 17. Miki D, Ochi H, Hayes CN et al Variation in the DEPDC5 locus is associated with progression to hepatocellular carcinoma in chronic hepatitis C virus carriers. Nat Genet 2011; 43: 797–800. [DOI] [PubMed] [Google Scholar]
  • 18. Miki D, Kubo M, Takahashi A et al Variation in TP63 is associated with lung adenocarcinoma susceptibility in Japanese and Korean populations. Nat Genet 2010; 42: 893–6. [DOI] [PubMed] [Google Scholar]
  • 19. Cha PC, Zembutsu H, Takahashi A, Kubo M, Kamatani N, Nakamura Y. A genome‐wide association study identifies SNP in DCC is associated with gallbladder cancer in the Japanese population. J Hum Genet 2012; 57: 235–7. [DOI] [PubMed] [Google Scholar]
  • 20. Silvestris N, Del Re M, Azzariti A et al Optimized granulocyte colony‐stimulating factor prophylaxis in adult cancer patients: from biological principles to clinical guidelines. Expert Opin Ther Targets 2012; 16(Suppl 2): S111–7. [DOI] [PubMed] [Google Scholar]
  • 21. Yamaguchi‐Kabata Y, Nakazono K, Takahashi A et al Japanese population structure, based on SNP genotypes from 7003 individuals compared to other ethnic groups: effects on population‐based association studies. Am J Hum Genet 2008; 83: 445–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Low SK, Takahashi A, Cha PC et al Genome‐wide association study for intracranial aneurysm in the Japanese population identifies three candidate susceptible loci and a functional genetic variant at EDNRA. Hum Mol Genet 2012; 21: 2102–10. [DOI] [PubMed] [Google Scholar]
  • 23. De Jager PL, Chibnik LB, Cui J et al Integration of genetic risk factors into a clinical algorithm for multiple sclerosis susceptibility: a weighted genetic risk score. Lancet Neurol 2009; 8: 1111–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kiyotani K, Uno S, Mushiroda T 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. [DOI] [PubMed] [Google Scholar]
  • 25. Guastalla JP, Pujade‐Lauraine E, Weber B et al Efficacy and safety of the paclitaxel and carboplatin combination in patients with previously treated advanced ovarian carcinoma. A multicenter GINECO (Group d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens) phase II study. Ann Oncol 1998; 9: 37–43. [DOI] [PubMed] [Google Scholar]
  • 26. Wang Z, Hao Y, Lowe AW. The adenocarcinoma‐associated antigen, AGR2, promotes tumor growth, cell migration, and cellular transformation. Cancer Res 2008; 68: 492–7. [DOI] [PubMed] [Google Scholar]
  • 27. Hengel SM, Murray E, Langdon S et al Data‐independent proteomic screen identifies novel tamoxifen agonist that mediates drug resistance. J Proteome Res 2011; 10: 4567–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Perik PJ, De Vries EG, Boomsma F et al The relation between soluble apoptotic proteins and subclinical cardiotoxicity in adjuvant‐treated breast cancer patients. Anticancer Res 2006; 26: 3803–11. [PubMed] [Google Scholar]
  • 29. Mukherjee S, Banerjee SK, Maulik M, Dinda AK, Talwar KK, Maulik SK. Protection against acute adriamycin‐induced cardiotoxicity by garlic: role of endogenous antioxidants and inhibition of TNF‐alpha expression. BMC Pharmacol 2003; 3: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Gilliam LA, St Clair DK. Chemotherapy‐induced weakness and fatigue in skeletal muscle: the role of oxidative stress. Antioxid Redox Signal 2011; 15: 2543–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Ariga M, Neitzert B, Nakae S et al Nonredundant function of phosphodiesterases 4D and 4B in neutrophil recruitment to the site of inflammation. J Immunol 2004; 173: 7531–8. [DOI] [PubMed] [Google Scholar]
  • 32. Stephensen CB, Borowsky AD, Lloyd KC. Disruption of Rxra gene in thymocytes and T lymphocytes modestly alters lymphocyte frequencies, proliferation, survival and T helper type 1/type 2 balance. Immunology 2007; 121: 484–98. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Fig. S1. Quantile–quantile plots of 17 genome‐wide association studies of drug‐induced or drug subgroup‐induced severe neutropenia and their corresponding lambda (λ) value.

Fig. S2. (a) Manhattan plot for genome‐wide association study of cyclophosphamide and paclitaxel + carboplatin induced severe neutropenia/leucopenia. (b) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of platinum‐based agents, cisplatin, or carboplatin. (c) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of anthracycline‐based agents, doxorubicin, or epirubicin. (d) Manhattan plot for genome‐wide association study of severe neutropenia/leucopenia induced by all types of antimetabolite agents, 5‐fluorouracil, or gemcitabine. (e) Manhattan plot for genome‐wide association study of severe neutropenia induced by all types of antimicrotubule agents, paclitaxel, or docetaxel. (f) Manhattan plot for genome‐wide association study severe neutropenia/leucopenia induced by all types of topoisomerase inhibitors, camptothecin, or etoposide.

Table S1. Genome‐wide association study of each chemotherapy regimen with P < 1 × 10−4.

Table S2. Weighted genetic risk score of each genome‐wide association study of specific chemotherapeutic‐based induced severe neutropenia/leucopenia.

Table S3. Association study of cancer patients who do not develop any adverse drug reaction and those who developed neutropenia/leucopenia after being given combination treatment with paclitaxel and carboplatin.

Table S4. Association of previously reported SNPs that associated with gemcitabine induced hematological toxicity.


Articles from Cancer Science are provided here courtesy of Wiley

RESOURCES