Abstract
There is established clinical evidence for differences in drug response, cure rates and survival outcomes between different ethnic populations, but the causes are poorly understood. Differences in frequencies of functional genetic variants in key drug response and metabolism genes may significantly influence drug response differences in different populations. To assess this, we genotyped 1330 individuals of African (n = 372) and European (n = 958) descent for 4535 single-nucleotide polymorphisms in 350 key drug absorption, distribution, metabolism, elimination and toxicity genes. Important and remarkable differences in the distribution of genetic variants were observed between Africans and Europeans and among the African populations. These could translate into significant differences in drug efficacy and safety profiles, and also in the required dose to achieve the desired therapeutic effect in different populations. Our data points to the need for population-specific genetic variation in personalizing medicine and care.
Keywords: pharmacogenomic diversity, genetic ancestry, drug response, ADRs
INTRODUCTION
Drug response, cure rates and survival outcomes for many diseases have improved significantly over the last few decades, but not all populations have benefited equally from this progress. In particular, there is growing clinical evidence concerning differences in the incidence of adverse drug reactions (ADRs) by population.1 ADRs represent the fourth leading cause of morbidity and mortality in developed countries, with direct medical costs of US$137–177 billion annually in the USA alone.2–4 The cost and contribution of ADRs to patient morbidity, hospitalization and mortality in African countries are not known.
Several observations have pointed to the fact that African and Hispanic populations are generally at increased susceptibility to ADRs, and have poorer survival and response rates for many diseases and medications when compared with European and Asian populations.5,6 For example, the rate of cisplatin-related toxicity has been shown to be significantly higher amongst African Americans compared with western Europeans (47.6% vs 8.3%, P=0.007)7 The maximum tolerable dose of cisplatin is 40% lower in the South African Black population compared with western countries (25 vs 40 mg m−2 per week).8 The risk and frequency of cardiotoxicity and congestive heart failure (CHF) after anthracycline treatment has been reported to be significantly higher in African compared with European populations (1.7-fold greater relative risk and 7% vs 2.4% frequency, P<0.027, odds ratio = 2.93).9,10 African ancestry has also been associated with increased likelihood of anthracycline dose reduction, early termination of treatment and decreased survival rates when compared with Europeans.11 African populations also have poorer survival and response rates,12 and experience significantly more frequent hematologic toxicities of leukopenia and anemia when compared with Europeans (P<0.006) after treatment with 5-fluorouracil (5-FU).13
The causes of these differences are poorly understood but could be due to genetic, clinical and/or environmental factors. The recent identification of the genetic basis of drug response and ADRs for many agents now makes it possible to identify the genetic causes of drug response differences at a population level. In the current study, we tested the hypothesis that the differences in frequencies of genetic variants in key genes involved in drug biotransformation may underlie these differences in ADR rates and response to therapy by population. Studies on pharmacogenomic diversity in African populations are underrepresented in published genomic and pharmacogenomic research and this study begins to address this deficiency.14–15
MATERIALS AND METHODS
Study population
Our study population (n = 1330) consisted of individuals of African (n = 372) and European (n = 958) descent. Individuals of African ancestry were drawn from indigenous ethnically, geographically, linguistically and culturally diverse African populations originating from Eastern (Kenya and Tanzania), Central (Cameroon and Chad), Western (Nigeria), Saharan (Sudan),14 and Southern (South Africa) Africa, recruited via field expeditions (Table 1). Europeans were from North America (Canada) recruited via the Canadian Pharmacogenomics Network for Drug Safety, a multicenter active drug surveillance and pharmcogenomics consortium.17 All genetic ancestries were self-reported and verified by principal component analysis. All participants were verified for cryptic relatedness using identity by descent estimation, and no duplicates (>99% identity) or related individuals (86–98% identity) were found.
Table 1.
Population | Country | Ethnic group | Subsistence | Latitude | Longitude | Language family | Language major subgrouping |
Sample size |
---|---|---|---|---|---|---|---|---|
North America | Canada | Europeans | — | — | — | English | Canadian English | 958 |
Central Africa | Cameroon | Fulani | Herder | 9 | 13.5 | Niger-Kordofanian | Senegambian | 19 |
Lemande | Farmer | 4.5 | 11 | Niger-Kordofanian | Bantoid | 19 | ||
Mada | Farmer | 10.8 | 14.1 | Afro-Asiatic | Chadic | 19 | ||
Bakola Pygmy | Hunter-gatherer | 2.8 | 10 | Niger-Kordofanian | Bantoid | 19 | ||
Chad | Bulala | Farmer (with fishing) |
13 | 18 | Nilo-Saharan | Central Sudanic | 16 | |
Eastern Africa | Kenya | Boni | Hunter-gatherer | — | — | — | — | 19 |
Borana | Herder | 3 | 38 | Afro-Asiatic | Cushitic | 19 | ||
Luo | Herder | −0.5 | 34.5 | Nilo-Saharan | Eastern Sudanic | 19 | ||
Sengwar | Hunter-gatherer | 1 | 35 | Nilo-Saharan | Eastern Sudanic | 19 | ||
Tanzania | Datog | Herder | −4.5 | 35.5 | Nilo-Saharan | Eastern Sudanic | 19 | |
Hazda | Hunter-gatherer | −3.5 | 35.3 | Khoesan | Hazda | 19 | ||
Iraqw | Mixed farmer | −4 | 35.5 | Afro-Asiatic | Cushitic | 19 | ||
Sandawe | Hunter-gatherer | −5.5 | 35.5 | Khoesan | Sendawe | 18 | ||
Western Africa | Nigeria | Yoruba | Farmer | 8 | 4 | Niger-Kordofanian | Defoid | 19 |
Saharan Africa | Sudan | Beja | Herder | 21 | 36 | Afro-Asiatic | Cushitic | 19 |
Southern African |
South Africa | Tswana/Xhosa/ Venda |
Farmers/Miners | — | — | Afrikaans | Afrikaans | 91 |
Study Sample Size |
1330 |
This study was approved by the ethics committees of the universities and institutions participating in the scientific projects of the Canadian Pharmacogenomics Network for Drug Safety and the ethics committee of the University of Pennsylvania. Written informed consent was obtained from all participants or from the parents or legal guardians in case of minors in accordance with the Helsinki Declaration as revised in 2008 (http://www.wma.net/en/30publications/10policies/b3/index.html, accessed 4 March 2013).
Pharmacogenomic assay design and variant clinical annotation
Our assay was designed to genotype for 4535 single-nucleotide polymorphisms (SNPs) in 350 key genes involved in drug biotransformation, drug response and ADRs, and included transporters, enzymes, receptors, ion channels, transcription factors and drug targets. The candidate genes were selected based on their physiological roles in drug absorption, distribution, metabolism, elimination and toxicity. Genetic variations in the absorption, distribution, metabolism, elimination and toxicity genotyping panel were selected to have the maximum set of informative markers to assay the candidate genes. Tagging SNPs were selected by IdSelect algorithm,18 using data from the International HapMap consortium.19 Functional SNPs were identified via publicly available databases (PharmGKB, HuGENet, ALFRED) and also from published information about their role in protein structure and/or function. Clinical annotations were curated from PharmGKB,20 Hiv-pharmacogenomics.org,21 and published studies22–26
Genotyping and quality control
Genomic DNA was extracted from blood, saliva or buccal swab samples using the QIAamp DNA purification system (Qiagen, Toronto, Ontario, Canada) and quantified by Quant-iT PicoGreen assay (Invitrogen, Eugene, OR, USA), according to the manufacturer’s protocols. A total of 1330 DNA samples were genotyped using a customized Illumina GoldenGate SNP genotyping assay (Illumina, San Diego, CA, USA). Genotypes were called with the Illumina Genome Studio software package (Illumina). All samples in the study cohort were used to determine cluster boundaries in order to maximize clustering accuracy. The clusters were then evaluated using automated scripts, while the ambiguous ones were evaluated manually. Twenty samples and 326 SNPs with call rates below 90% were excluded from the analyses (average call rate for included samples = 95.0% and SNPs = 99.8%). Quality control analysis was performed by date of genotyping and by plate to check for systematic errors in the generated data set. No systematic errors were found. After quality control, 1310 samples and 4209 SNPs remained for further analyses.
Statistical analyses
Statistical analyses were performed using SVS/HelixTree 7.6.8 (Golden Helix, Bozeman, MT, USA). Fisher’s exact test was used for comparing allele frequencies among populations and an ANOVA F-test was used to explore the pharmacogenomic diversity in African populations. The type-I error rate of 0.05 was used as a significance threshold after Bonferroni correction for 4209 tests.
RESULTS
Pharmacogenomic diversity and population ancestry
Distribution of pharmacogenomic polymorphisms
We compared allele frequencies between African and European populations (Supplementary Table 1). Several variants in the cytochrome P4503A (CYP3A) family showed the most significant differences and were more frequent in African compared with European populations: CYP3A rs4646450 −90.34% vs 16.77%, P=6.93E-274; CYP3A4 rs2740574 −66.25% vs 3.25%, P=3.95E-257; CYP3A4 rs2242480 −77.59% vs 11.14%, P=2.92E-231; CYP3A4 rs4646437 − 77.73% vs 11.45%, P=1.43E-229; CYP3A5 rs776746 − 74.37% vs 10.13%, P=3.60E-221; and CYP3A rs6945984 − 76.89% vs 12.72%, P = 3.52E-212 (Table 2). Striking differences were also observed for other cytochrome P450 variants and variants in solute carrier and ATP-binding cassette transporters.
Table 2.
Gene | Variant | CHR | Position | Functional annotation | Allelesa | MAF—African ancestry |
MAF—European ancestry |
Bonferroni P-valueb,c |
---|---|---|---|---|---|---|---|---|
CYP3A | rs4646450 | 7 | 99104254 | Intron | A/G | 0.903 | 0.168 | 6.93E-274 |
CYP2B6 | rs34097093 | 19 | 46210210 | Coding NONSYN R378* (CYP2B6*28) |
G/A | 0.562 | 0.002 | 2.68E-262 |
CYP3A4 | rs2740574 | 7 | 99220032 | Flanking_5UTR | G/A | 0.662 | 0.033 | 3.95E-257 |
GSTA1/2/3/4/5 | rs6577 | 6 | 52723374 | Coding NONSYN E210A | C/A | 0.709 | 0.057 | 2.51E-250 |
ABCC1/6 | rs246227 | 16 | 16043648 | Intron | G/A | 0.749 | 0.083 | 1.06E-244 |
CYP3A4 | rs2242480 | 7 | 99199402 | Intron | A/G | 0.776 | 0.111 | 2.92E-231 |
CYP3A4 | rs4646437 | 7 | 99203019 | Intron | A/G | 0.777 | 0.114 | 1.43E-229 |
CYP3A5 | rs776746 | 7 | 99108475 | Intron | A/G | 0.744 | 0.101 | 3.60E-221 |
TBXAS1 | rs4529 | 7 | 139308433 | Coding NONSYN L357V | C/G | 0.478 | 0.001 | 1.49E-219 |
ABCG2 | rs2622610 | 4 | 89246566 | Intron | A/G | 0.689 | 0.077 | 2.60E-213 |
CYP3A | rs6945984 | 7 | 99186264 | Flanking_3UTR | G/A | 0.769 | 0.127 | 3.52E-212 |
SLCO1B3 | rs7311358 | 12 | 20907027 | Coding NONSYN M233I | G/A | 0.846 | 0.193 | 3.39E-208 |
CYB5R3 | rs137124 | 22 | 41345660 | Flanking_3UTR | G/A | 0.843 | 0.194 | 1.29E-206 |
SLC28A1 | rs16974622 | 15 | 83265515 | Intron | G/A | 0.620 | 0.050 | 6.25E-203 |
PPARD | rs6901410 | 6 | 35438008 | Intron | G/A | 0.655 | 0.073 | 8.79E-198 |
ALDH7A1 | rs3736171 | 5 | 125959275 | Flanking_5UTR | A/C | 0.768 | 0.145 | 9.80E-196 |
ABCA4 | rs3789375 | 1 | 94237720 | Intron | C/A | 0.724 | 0.116 | 1.80E-194 |
PPARD | rs6457816 | 6 | 35470826 | Intron | G/A | 0.657 | 0.077 | 2.08E-194 |
ALDH2 | rs2238151 | 12 | 110696216 | Intron | A/G | 0.050 | 0.658 | 5.15E-193 |
CYP27A1 | rs6436094 | 2 | 219395841 | Flanking_3UTR | G/A | 0.890 | 0.262 | 6.52E-191 |
Abbreviations: ADME, absorption, distribution, metabolism, elimination and toxicity; CHR, chromosome; MAF, minor allele frequency; SNP, single-nucleotide polymorphism; UTR, untranslated region.
Minor/major.
Corrected P-value (4209 SNPs).
Statistics—Fisher’s exact test.
Most ADR-associated risk variants or variants associated with poor response or shortened survival rates were more frequent in African populations compared with Europeans (Supplementary Table 2). Specific examples of clinically important differences between African and European populations focusing on cancer, antiretroviral and antimicrobial pharmacogenomics are provided here, while other examples can be found in the Supplementary Material of this manuscript (Supplementary Tables 1–2).
Pharmacogenomic diversity in cancer therapy
Anthracyclines and related substances
UGT1A6 rs17863783, ABCC2 rs8187694, ABCC2 rs8187710, ABCB1 rs2235047 and ABCC1 rs4148350 have been associated with the risk of anthracycline-induced cardiotoxicity (ACT) and CHF,22,23 while RAC2 rs13058338, SULT2B1 rs10426377, CBR1 rs9024 and HNMT rs17645700 have been shown to have a protective effect.22,23 Also, ABCB1 rs2032582 has been clinically linked to improved survival and response rates,26 while CYP2B6 rs3745274 has been associated with decreased tolerance and survival.27 The distributions of these variants were significantly different between African and European populations (Table 3). UGT1A6 rs17863783 (12.04% vs 2.78%, P=9.75E-15), ABCC2 rs8187694 (14.29% vs 5.37%, P=2.54E-09), ABCC2 rs8187710 (22.99% vs 5.47%, P = 8.87E-31), ABCB1 rs2235047 (18.21% vs 2.52%, P=3.84E-36), ABCC1 rs4148350 (13.48% vs 5.43%, P=2.09E-07), RAC2 rs13058338 (10.92% vs 24.97%, P=.09E-12), SULT2B1 rs10426377 (17.51% vs 27.39%, P= 4.66E-04), CBR1 rs9024 (1.40% vs 12.46%, P=1.73E-19), HNMT rs17645700 (4.76 vs 20.17, P=4.57E-22), ABCB1 rs2032582 (3.78% vs 45.10%, P=1.87E-106) and CYP2B6 rs3745274 (77.73% vs 24.97%, P=4.34E-131). Variants associated with increased risk of ACT and CHF and decreased tolerance and survival were more frequent in African populations, while those associated with protection against ACT and CHF and improved survival and response rates were less frequent.
Table 3.
Drug responsea | Gene | Variant | CHR | Position | Functional annotation | Allelesb | African ancestryc |
European ancestryc |
Bonferroni P-valued,e |
---|---|---|---|---|---|---|---|---|---|
TOXICITY/ADR Increased risk of cardiotoxicity and heart failure |
UGT1A6 ABCC2 ABCC2 ABCB1 ABCC1/6 |
rs17863783 rs8187694 rs8187710 rs2235047 rs4148350 |
2 10 10 7 16 |
234267016 101585986 101601284 86976468 16077978 |
Coding SYNON V209V Coding NONSYNON Coding NONSYN (C1515Y) Intron Intron |
A/C T/A A/G C/A A/C |
0.120 0.143 0.230 0.182 0.135 |
0.028 0.054 0.055 0.025 0.054 |
9.75E-15 2.54E-09 8.87E-31 3.84E-36 2.09E-07 |
TOXICITY/ADR Decreased risk of cardiotoxicity and heart failure |
RAC2 SULT2B1 CBR1 HNMT |
rs13058338 rs10426377 rs9024 rs17645700 |
22 19 21 2 |
35962716 53784046 36367183 138497402 |
Intron Intron Flanking_3UTR Flanking_3UTR |
T/A A/C A/G G/A |
0.109 0.175 0.014 0.048 |
0.250 0.274 0.125 0.202 |
1.09E-12 4.66E-04 1.73E-19 4.57E-22 |
DOSAGE Increased likelihood of dose reduction |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 |
EFFICACY increased response |
ABCB1 | rs2032582 | 7 | 86998554 | Coding NONSYNON (ABCB1*13 and ABCB1*2) |
A/C | 0.038 | 0.451 | 1.87E-106 |
Abbreviations: ADR, adverse drug reaction; CH, chromosome; SNP, single-nucleotide polymorphism; UTR, untranslated region.
Clinical information for relevant pharmacogenomics variants were curated from PharmGKB,20 and published studies.22–26
Alleles = minor allele/major allele.
Variant minor allele frequency (MAF).
Corrected P-value (corrected for 4209 SNPs).
Statistical test—Fisher’s exact test.
Cisplatin and platinum compounds
COMT rs9332377 has been associated with increased risk of cisplatin-induced hearing loss,28 MTR rs1805087 associated with reduced risk of cisplatin-induced toxicity, XRCC1 rs25487 associated with decreased risk of severe neutropenia when treated with platinum compounds (cisplatin, carboplatin, oxaliplatin and platinum)29 and MTHFR rs1801133 clinically linked to increased response to platinum compounds.30 The allele frequencies of these pharmacogenomic variants were significantly different between African and European populations (Table 4). COMT rs9332377 (32.68% vs 16.82%, P=5.40E-14), XRCC1 rs25487 (14.85% vs 35.07%, P=.37E-22), MTR rs1805087 (0.27.17 vs 18.68, P=.0142) and MTHFR rs1801133 (6.86% vs 34.77%, P=.57E-51). Pharmacogenomic variants associated with increased risk of toxicity related to platinum compounds were more common in African populations, while variants associated with decreased risk were less common. Also, variants associated with improved response to platinum compounds were less frequent in African populations.
Table 4.
Drug | Drug responsea | Gene | Variant | CH | Position | Functional annotation |
Allelesb | African ancestryc |
European ancestryc |
Bonferroni P-valued,e |
---|---|---|---|---|---|---|---|---|---|---|
Cisplatin | TOXICITY/ADR Increased risk of hearing loss (Ototoxicity) |
COMT | rs9332377 | 22 | 18335692 | Intron | A/G | 0.327 | 0.168 | 5.40E-14 |
TOXICITY/ADR Decreased risk of severe neutropenia |
XRCC1 | rs25487 | 19 | 48747566 | Coding NONSYN Q399R |
A/G | 0.148 | 0.351 | 3.37E-22 | |
TOXICITY/ADR Increased likelihood of drug toxicity |
MTR | rs1805087 | 1 | 235115123 | Coding NONSYN D919G |
G/A | 0.272 | 0.187 | 0.0142 | |
EFFICACY Increased response |
MTHFR | rs1801133 | 1 | 11778965 | Coding NONSYN A222V |
A/G | 0.069 | 0.348 | 2.57E-51 | |
Carboplatin | TOXICITY/ADR Decreased risk of severe neutropenia |
XRCC1 | rs25487 | 19 | 48747566 | Coding NONSYN Q399R |
A/G | 0.148 | 0.351 | 3.37E-22 |
EFFICACY Increased response |
MTHFR | rs1801133 | 1 | 11778965 | Coding NONSYN A222V |
A/G | 0.069 | 0.348 | 2.57E-51 | |
Oxaliplatin | TOXICITY/ADR Decreased risk of severe neutropenia |
XRCC1 | rs25487 | 19 | 48747566 | Coding NONSYN Q399R |
A/G | 0.148 | 0.351 | 3.37E-22 |
EFFICACY Increased response |
MTHFR | rs1801133 | 1 | 11778965 | Coding NONSYN A222V |
A/G | 0.069 | 0.348 | 2.57E-51 |
Abbreviations: ADR, adverse drug reaction; CH, chromosome; SNP, single-nucleotide polymorphism.
The clinical annotations of the relevant pharmacogenomics variants were curated from PharmGKB20 and published studies.28–30,33
Minor allele/Major allele.
Variant minor allele frequency (MAF).
Corrected P-value (corrected for 4209 SNPs).
Statistical test—Fisher’s exact test.
Fluororuracil
The majority of 5-FU-induced toxicity are related to the deficiency of dihydropyrimidine dehydrogenase, an enzyme, which metabolizes 5-FU. The common mutations in the dihydropyrimidine dehydrogenase gene—DPYD (DPYD rs1801265 and DPYD rs2297595) have been associated with fluoropyrimidine-related toxicity in cancer patients,31,32 while MTHFR rs1801133 has been linked to improved survival and response rates.33 The distributions of these mutations were different in African and European populations (Table 5). DPYD rs1801265 −48.88% vs 22.19%, P=4.46E-35; DPYD rs2297595 −19.75% vs 10.19%, P=1.30E-06; MTHFR rs1801133 −6.86% vs 34.77%, P=2.57E-51. Pharmacogenomic variants associated with increased risk of 5-FU-induced toxicity were more frequent in African populations, while MTHFR rs1801133 associated with improved response was less frequent.
Table 5.
Drugs | Drug responsea | Gene | Variant | CH | Position | Function | Allelesb | African ancestryc |
European ancestryc |
Bonferroni P-valued,e |
---|---|---|---|---|---|---|---|---|---|---|
PYRIMIDINE COMPOUNDS Fluorouracil |
TOXICITY/ADR Increased risk of middle-severe nausea and vomiting |
DPYD | rs1801265 | 1 | 98121473 | Coding NONSYN r29c (DPYD*9A) |
G/A | 0.489 | 0.222 | 4.46E-35 |
TOXICITY/ADR Increased risk of severe toxicity |
DPYD | rs2297595 | 1 | 97937679 | Coding NONSYN M166V |
G/A | 0.197 | 0.102 | 1.32E-06 | |
EFFICACY Increased response |
MTHFR | rs1801133 | 1 | 11778965 | Coding NONSYN A222V |
A/G | 0.069 | 0.348 | 2.57E-51 |
Abbreviations: ADR, adverse drug reaction; CH, chromosome; SNP, single-nucleotide polymorphism.
The clinical annotations of the relevant pharmacogenomics variants were curated from PharmGKB20 and published studies.31–33,56,57
Minor allele/Major allele.
Variant minor allele frequency (MAF).
Corrected P-value (corrected for 4209 SNPs).
Statistical test—Fisher’s exact test.
Vincristine
Vincristine is a metabolic substrate for CYP3A5. It has been shown that increased risk of vincristine-induced neurotoxicity is associated with low CYP3A5 expression.34 Therefore, mutations in CYP3A5 may influence the efficacy and toxicity of vincristine. We explored the distribution of CYP3A5 mutations in African and European populations. Strikingly significant differences were observed for the following CYP3A5 polymorphisms: CYP3A5 rs776746 −74.37% vs 10.13%, P=3.60E-221; CYP3A5 rs10224569 − 28%.37% vs 0.0%, P=2.39E-121; CYP3A5 rs10264272 −22.33% vs 0.32%, P=2.44E-83; CYP3A5 rs10249369 − 21.91% vs 0.58%, P=1.02E-75; CYP3A5 rs41303343 − 7.56% vs 0.10% P=2.59E-25; CYP3A5 rs6956305 −9.10% vs 4.41%, P=0.0443)—Table 6. Also, significant differences were observed for MTHFR rs1801133 (34.77% vs 6.86%, P=2.57E-51) and NOS rs1799983 (33.80% vs 4.95%, P=8.47E-012) that have been associated with other vincristine-related toxicities.35,36 CYP3A5 polymorphisms were more frequent in African compared with European populations. In contrast, ADR-associated variants and ABCB1*13 (3.78% vs 45.10% P=1.87E-106) that has been associated with efficacy26 were less frequent in African populations.
Table 6.
Drug responsea | Gene | Variant | CH | Position | Function | Allelesb | African ancestryc |
European ancestryc |
Bonferroni P-valued,e |
---|---|---|---|---|---|---|---|---|---|
TOXICITY/ADR Increased risk of drug toxicity |
MTHFR | rs1801133 | 1 | 11778965 | Coding NONSYN A222V | A/G | 0.069 | 0.348 | 2.57E-51 |
TOXICITY/ADR Decreased IQ |
NOS | rs1799983 | 7 | 150327044 | Coding NONSYN D298E | A/C | 0.049 | 0.338 | 8.47E-012 |
EFFICACY Improved Response |
ABCB1 | rs2032582 | 7 | 86998554 |
Coding NONSYNON (ABCB1*13 and ABCB1*2) |
A/C | 0.038 | 0.451 | 1.87E-106 |
DRUG RESPONSE PATHWAY Vincristine main metabolic substrate |
CYP3A5 CYP3A5 CYP3A5 |
rs776746 rs10224569 rs10264272 |
7 7 7 |
99108475 99086240 99100771 |
Intron (CYP3A5*3) Intron Coding SYNON K208K (CYP3A5*6) |
A/G A/G A/G |
0.744 0.284 0.223 |
0.101 0 0.003 |
3.60E-221 2.39E-121 2.44E-83 |
CYP3A5 CYP3A5 CYP3A5 |
rs10249369 rs41303343 rs6956305 |
7 7 7 |
99084928 99088358 99079246 |
Intron Coding FRAMESHIFT Flanking_3UTR |
G/A A/T G/A |
0.219 0.076 0.091 |
0.006 0.001 0.044 |
1.02E-75 2.59E-25 0.0443 |
Abbreviations: ADR, adverse drug reaction; CH, chromosome; IQ, intelligence quotient; SNP, single-nucleotide polymorphism; UTR, untranslated region.
The clinical annotations of the relevant pharmacogenomics variants were curated from PharmGKB20 and published studies.26,35,36,49–51
Minor allele/major allele.
Variant minor allele frequency (MAF).
Corrected P-value (corrected for 4209 SNPs).
Statistical test—Fisher’s exact test.
Pharmacogenomic diversity in antiretroviral and antimycobacterial therapy
CYP2B6 rs28399499 is associated with Nevirapine-induced hepatotoxicity.21 CYP2B6 rs3745274 is associated with rifampicin-induced liver injury and with efavirenz-induced lowered HDL levels, hepatotoxicity, neurotoxicity, fatigue and sleep disorders and early termination of treatment.21 Also, ABCC2 rs717620, ABCC2 rs17222723 and ABCC4 rs1751034 are associated with Tenofovir-induced proximal tubulopathy and kidney tubular dysfunction and APOE rs429358 associated with extreme hypertriglyceridemia.20,21 The allele frequencies of these variants were significantly higher in the African compared with European populations (CYP2B6 rs28399499 − 5.20% vs 0.052%, P=7.04E-17; CYP2B6 rs3745274 −77.73% vs 24.97%, P=4.34E-131; ABCC2 rs717620 −19.77% vs 3.24%, P=4.54E-28; ABCC2 rs17222723 − 14.29% vs 5.46%, P=5.94E-09; ABCC4 rs1751034 −30.95% vs 17.79%, P=4.77E-09; APOE rs429358 −24.86% vs 13.27%, P=5.75E-08 −Table 7), which would be compatible with increased frequency of ADRs with these drugs.
Table 7.
Drug | Drug responsea | Gene | Variant | C | Position | Functional annotation |
Sqb | African ancestryc |
European ancestryc |
Bonferroni P-valued |
---|---|---|---|---|---|---|---|---|---|---|
Nucleoside reverse-transcriptase inhibitors | ||||||||||
Tenofovir | Increased risk of proximal tubulopathy and risk of kidney tubular dysfunction |
ABCC2 ABCC2 |
rs717620 rs17222723 |
10 10 |
101532568 101585986 |
Flanking_5UTR Coding NONSYNON |
G/A T/A |
0.968 0.143 |
0.802 0.055 |
4.54E-28 5.94E-09 |
Increased risk of proximal tubulopathy |
ABCC4 | rs1751034 | 13 | 94512977 | Coding FRAMESHIFT | G/A | 0.310 | 0.178 | 4.77E-09 | |
Nonnucleoside reverse-transcriptase inhibitors | ||||||||||
Nevirapine | Increased risk of hepatotoxicity |
CYP2B6 | rs28399499 | 19 | 46210061 | Coding NONSYN I328T (CYP2B6* 16/*18) |
G/A | 0.052 | 0.0005 | 7.04E-17 |
Efavirenz | Increase in HDL- cholesterol levels |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 |
Increased risk of neurotoxicity, CNS depression and neuropsychiatric disorders |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 | |
Increased risk of fatigue and sleep disorder |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 | |
Increased risk of hepatotoxicity and drug-induced liver injury |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 | |
Protease inhibitors | ||||||||||
Ritonavir | Increased risk of extreme hypertriglyceridemia |
APOE | rs429358 | 19 | 50103781 | Coding NONSYN C130R |
G/A | 0.249 | 0.133 | 5.75E-08 |
Antituberculosis therapy | ||||||||||
Rifampicin | Increased risk of hepatotoxicity and drug-induced liver injury (DILI) |
CYP2B6 | rs3745274 | 19 | 46204681 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.777 | 0.250 | 4.34E-131 |
Abbreviations: C, chromosome; CNS, central nervous system; DILI, drug-induced liver injury; HDL, high-density lipoprotein; MAF, minor allele frequency
Clinical annotation curated from PharmGKB20 and http://www.hiv-pharmacogenomics.org.21
Sq, sequence (minor/major).
MAF.
P-value corrected for 4209.
Pharmacogenomic diversity among African populations
We characterized and compared allele frequencies in five different African populations (Supplementary Tables 3–5). The F-statistics and P-values revealed high pharmacogenomic diversity among African populations. The most clinically relevant diversity was observed for VKORC1 rs7294 (P=8.93E-21), which is associated with warfarin dosage requirement37 (Table 8). Other top clinically annotated variations include VKORC1 rs8050894 (P=8.92E-07) for warfarin dosage38 and several cytochrome P450 variants associated with response to immunosuppressive drugs (CYP2B6 rs2279343 - P=.46E-20 for cyclophosphamide-induced mucositis,39 CYP2B6 rs8192709 - P=.0167 for cyclophosphamide-induced hemorr-hagic cystitis39 and CYP3A5 rs776746 - P=.14E-09 for cyclospo-rine dosage requirements20).
Table 8.
Associated drugsb | Associatedb response |
Gene | Variant | CH | Position | Function | F-statisticc | Bonferroni P-valuea |
---|---|---|---|---|---|---|---|---|
Warfarin | Dosage | VKORC1 | rs7294 | 16 | 31009822 | flanking_3UTR | 34.655 | 8.93E-21 |
Cyclophosphamide | Toxicity/ADR | CYP2B6 | rs2279343 | 19 | 46207103 | Coding NONSYN K262R (CYP2B6*6) | 34.374 | 1.46E-20 |
Cyclosporine | Dosage | CYP3A5 | rs776746 | 7 | 99108475 | Intron | 17.760 | 1.14E-09 |
Warfarin | Dosage | VKORC1 | rs8050894 | 16 | 31012010 | Intron | 13.714 | 8.92E-07 |
Thiotepa | Drug clearance | GSTP1 | rs1138272 | 11 | 67110155 | Coding NONSYNON A114V | 14.343 | 3.49E-05 |
Cisplatin, | Toxicity/ADR | ERCC1 | rs11615 | 19 | 50615493 | Coding SYNON N118N | 10.730 | 0.0040 |
Cyclophosphamide | ||||||||
Repaglinide | Plasma concentration |
SLCO1B1 | rs2306283 | 12 | 21221005 | Coding NONSYNON | 8.034 | 0.0139 |
Cyclophosphamide | Toxicity/ADR | CYP2B6 | rs8192709 | 19 | 46189114 | Coding NONSYNON R22C (CYP2B6*2 and *10) |
9.654 | 0.0167 |
Abbreviations: ADR, adverse drug reactions; ANOVA, analysis of variance; CH, chromosome; SNP, single-nucleotide polymorphism.
The clinical annotations of the relevant pharmacogenomics variants were curated from PharmGKB,20 and published studies.
Statistical test = ANOVA F-test.
Corrected P-value (corrected for 4209 SNPs).
Pharmacogenetic variant frequencies including the frequencies of the CYP enzymes were observed to be remarkably variable within African populations as demonstrated by the following examples (Table 9 and Supplementary Table 3):
Table 9.
Drug | Drug responsea | Gene | Variant | Functional annotation | Sqb | Central Africans |
Eastern Africans |
Saharan Africans |
Southern Africans |
Western Africans |
---|---|---|---|---|---|---|---|---|---|---|
Anthracyclines and related substances | ||||||||||
Anthracycline | Increased risk of cardiotoxicity and heart failure |
UGT1A6 ABCC2 ABCB1 ABCC1/6 |
rs17863783 rs8187710 rs2235047 rs4148350 |
Coding SYNON V209V Coding NONSYN (C1515Y) Intron Intron |
A/C A/G C/A A/C |
0.090 0.213 0.139 0.169 |
0.128 0.257 0.178 0.122 |
0.184 0.237 0.211 0.053 |
0.129 0.200 0.213 0.135 |
0.088 0.219 0.235 0.176 |
Decreased risk of cardiotoxicity and heart failure |
RAC2 SULT2B1 CBR1 HNMT |
rs13058338 rs10426377 rs9024 rs17645700 |
Intron Intron Flanking_3UTR Flanking_3UTR |
T/A A/C A/G G/A |
0.108 0.223 0.000 0.066 |
0.134 0.158 0.013 0.044 |
0.132 0.211 0.053 0.000 |
0.079 0.146 0.017 0.056 |
0.029 0.206 0.029 0.000 |
|
Increased likelihood of dose reduction |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 | |
Increased response | ABCB1 | rs2032582 | Coding NONSYNON (ABCB1*13 and ABCB1*2) |
A/C | 0.036 | 0.034 | 0.026 | 0.045 | 0.059 | |
Platinum compounds | ||||||||||
Cisplatin | Increased risk of hearing loss (ototoxicity) |
COMT | rs9332377 | Intron | A/G | 0.323 | 0.352 | 0.237 | 0.330 | 0.206 |
Decreased risk of severe neutropenia |
XRCC1 | rs25487 | Coding NONSYN Q399R | A/G | 0.193 | 0.128 | 0.211 | 0.135 | 0.118 | |
Increased likelihood of drug toxicity |
MTR | rs1805087 | Coding NONSYN D919G | G/A | 0.295 | 0.262 | 0.132 | 0.287 | 0.324 | |
Increased response | MTHFR | rs1801133 | Coding NONSYN A222V | A/G | 0.048 | 0.060 | 0.132 | 0.090 | 0.059 | |
Carboplatin | Decreased risk of severe neutropenia |
XRCC1 | rs25487 | Coding NONSYN Q399R | A/G | 0.193 | 0.128 | 0.211 | 0.135 | 0.118 |
Increased response | MTHFR | rs1801133 | Coding NONSYN A222V | A/G | 0.048 | 0.060 | 0.132 | 0.090 | 0.059 | |
Oxaliplatin | Decreased risk of severe neutropenia Increased response |
XRCC1 MTHFR |
rs25487 rs1801133 |
Coding NONSYN Q399R Coding NONSYN A222V |
A/G A/G |
0.193 0.048 |
0.128 0.060 |
0.211 0.132 |
0.135 0.090 |
0.118 0.059 |
Antimetabolites | ||||||||||
PYRIMIDINE COMPOUNDS Fluorouracil |
Increased risk of toxicity of fluoropyrimidine- based chemotherapy |
DPYD DPYD |
rs1801265 rs2297595 |
Coding NONSYN R29C (DPYD*9A,) Coding NONSYN M166V |
G/A G/A |
0.452 0.253 |
0.550 0.141 |
0.579 0.079 |
0.381 0.275 |
0.529 0.147 |
Increased response | MTHFR | rs1801133 | Coding NONSYN A222V | A/G | 0.048 | 0.060 | 0.132 | 0.090 | 0.059 | |
Vincristine | ||||||||||
Vincristine | Increased risk of drug toxicity | MTHFR | rs1801133 | Coding NONSYN A222V | A/G | 0.048 | 0.060 | 0.132 | 0.090 | 0.059 |
Improved response | ABCB1 | rs2032582 | Coding NONSYNON (ABCB1*13 and ABCB1*2) | A/C | 0.036 | 0.034 | 0.026 | 0.045 | 0.059 | |
Vincristine main metabolic substrate | CYP3A5 | rs776746 | Intron (CYP3A5*3) | A/G | 0.771 | 0.799 | 0.684 | 0.685 | 0.500 | |
CYP3A5 CYP3A5 |
rs10224569 rs10264272 |
Intron Coding SYNON K208K (CYP3A5*6) |
A/G A/G |
0.367 0.283 |
0.260 0.209 |
0.316 0.289 |
0.264 0.191 |
0.147 0.147 |
||
CYP3A5 CYP3A5 CYP3A5 |
rs10249369 rs41303343 rs6956305 |
Intron Coding FRAMESHIFT Flanking_3UTR |
G/A A/T G/A |
0.277 0.036 0.066 |
0.205 0.111 0.117 |
0.263 0.079 0.079 |
0.193 0.062 0.079 |
0.147 0.029 0.059 |
||
Nucleoside reverse-transcriptase inhibitors | ||||||||||
Tenofovir | Increased risk of proximal tubulopathy and risk of kidney tubular dysfunction |
ABCC2 ABCC2 |
rs717620 rs17222723 |
Flanking_5UTR Coding NONSYNON |
G/A T/A |
0.957 0.151 |
0.990 0.138 |
0.974 0.211 |
0.944 0.135 |
0.941 0.118 |
Increased risk of proximal tubulopathy |
ABCC4 | rs1751034 | Coding FRAMESHIFT | G/A | 0.325 | 0.305 | 0.237 | 0.292 | 0.441 | |
Nonnucleoside reverse-transcriptase inhibitors | ||||||||||
Nevirapine | Increased risk of hepatotoxicity |
CYP2B6 | rs28399499 | Coding NONSYN I328T (CYP2B6*16/*18) |
G/A | 0.012 | 0.067 | 0.000 | 0.080 | 0.029 |
Efavirenz | Increase in HDL- cholesterol levels |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 |
Increased risk of neurotoxicity, CNS depression and neuropsychiatric disorders |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 | |
Increased risk of fatigue and sleep disorder |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 | |
Increased risk of hepatotoxicity and drug- induced liver injury |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 | |
Protease Inhibitors | ||||||||||
Ritonavir | Increased risk of extreme hypertriglyceridemia |
APOE | rs429358 | Coding NONSYN C130R | G/A | 0.179 | 0.284 | 0.211 | 0.247 | 0.324 |
Antituberculosis therapy | ||||||||||
Rifampicin | Increased risk of hepatotoxicity and drug- induced liver injury |
CYP2B6 | rs3745274 | Coding NONSYN Q172H (CYP2B6*6) |
A/C | 0.783 | 0.748 | 0.658 | 0.837 | 0.824 |
Abbreviations: CNS, central nervous system; HDL, high-density lipoprotein; UTR, untranslated region.
Clinical annotation curated from PharmGKB,20 http://www.hiv-pharmacogenomics.org and published studies.
Sq, sequence (minor/major).
Antharcycline
The UGT1A6*4 haplotype associated with the risk of ACT and clinical heart failure was more frequent among Saharan (18.4%), Southern (12.9%) and Eastern (12.8%) Africans compared with Central (9.9%) and Western (8.8%) Africans.
Cisplatin
COMT rs9332377, which is associated with increased risk of cisplatin-induced hearing loss, was less common among Western (20.6%) and Saharan (23.7%) Africans compared with Eastern (35.2%), Southern (33.0%) and Central (32.3%) Africans.
Antiretroviral therapy
ABCC4 rs1751034 is associated with Tenofovir-induced proximal tubulopathy and kidney tubular dysfunction. This variant is very frequent among Western Africans (44.1%), intermediate among Central (32.5%), Eastern (30.5%) and Southern (29.2%) Africans, and comparatively less frequent in Saharan Africans (23.7%). Also, CYP2B6 rs28399499, which is associated with Nevirapine-induced hepatotoxicity is rare among Saharan (0.0%), Central (1.2%) and Western (2.9%) Africans, and relatively more common in Southern (8.0%) and Eastern (6.7%) Africans.
DISCUSSION
In this manuscript, we have chosen to focus on antineoplastic, antiretroviral and antimycobacterial commonly used drugs from the WHO Model List of Essential Medicines. Cancer, HIV/AIDS and tuberculosis and their associated ADRs are major public health problems and have become the primary focus in health-care. In general, a higher frequency of genetic variants conferring increased risk to ADRs for different and commonly used antineoplastic, antiretroviral and antituberculosis drugs was evident in African populations.
Anthracyclines are a group of very efficacious chemotherapeutic agents and have been part of the backbone of therapy worldwide including Africa for the treatment of many cancers including leukemia, lymphoma, sarcomas, Wilms’ tumor, hepato-blastoma, and uterine, ovarian, lung and breast cancers. They are used to treat over 70% of all childhood malignancies, as well as 50–90% of breast cancer patients each year.40 Their clinical utility is primarily limited by a highly individually variable, cumulative dose-dependent, cardiac toxicity, manifesting as asymptomatic cardiac dysfunction in up to 57% of treated patients41,42 and restrictive or dilated cardiomyopathy resulting in CHF in up to 16% of treated patients.43 African populations are more sensitive to ACT and CHF when compared with Europeans (1.7-fold greater relative risk; frequency −7% vs 2.4%, P<0.027, odds ratio = 2.93).9,10 The enrichment of genetic risk factors for ACT and CHF such as UGT1A6 rs17863783, ABCC2 rs8187694, ABCC2 rs8187710, ABCB1 rs2235047 and ABCC1 rs4148350, in African populations suggests that these genetic differences may partially account for the increased sensitivity to ACT and CHF in African populations. Also, the enrichment of pharmacogenetic factors such as CYP2B6 rs3745274 associated with poor drug tolerance and poor survival rates27 could contribute to the reported increased likelihood of dose reduction, early termination of treatment and decreased survival rates in African populations.11
Cisplatin, carboplatin and oxaliplatin are all platinum compounds that have excellent antineoplastic properties. Cisplatin is the drug of choice for solid tumors including hepatoblastoma, osteosarcoma, neuroblastoma and ovarian, central nervous system, testicular, cervical, lung, bladder, head and neck tumors.44 Major complications include ototoxicity, nephrotoxicity, neurotoxicity and myelotoxicity. Irreversible hearing loss (ototoxicity) occurs in 10–25% of treated adults, 50% of patients treated with high doses (>400mgm−2) and 41–61% of treated children45–47 Cisplatin-related toxicity is more frequent in African populations when compared with the European population (47.6% vs 8.3%, P = 0.007)7 Also, the maximum tolerable dose is 40% lower in African populations (25 vs 40 mg m−2 per week).8 In the current study, an overrepresentation of COMT rs9332377 (32.58% vs 16.82%) variants associated with increased risk of hearing loss28 and a depletion of XRCC1 rs25487 (14.85% vs 35.07%) associated with protection against cisplatin-induced neutropenia,29 in African populations, was evident. This correlates with the reported increased frequency of cisplatin-induced toxicity in these populations and suggests that these differences in allele frequency may contribute to the increased ADR rates in these populations.
5-FU is a very effective drug commonly used in the treatment of advanced stage colon cancer and several other types of cancer including, breast, esophageal and stomach cancers. About 30% of 5-FU-treated patients suffer from severe and sometimes deadly toxicity including hematologic toxicities of leukopenia and anemia, myelosuppression, diarrhea, nausea, vomiting, mucositis and dermatitis.1 Poorer survival and decreased response rates,12 and more frequent toxicities have been reported in African Americans when compared with Europeans (P<0.006).13 An enrichment of risk factors for dihydropyrimidine dehydrogenase deficiency such as the DPYD variants (rs1801265 −48.88% vs 22.19% and rs2297595 − 19.75% vs 10.19%) and a decrease in the frequency of MTHFR rs1801133 (6.9% vs 34.8%) linked to increased therapeutic response was evident33 in Africans compared with Europeans. These genetic findings could at least partially explain differences in ADR rates and drug responsiveness between these two populations.
Vincristine is a commonly prescribed vinca alkaloid and is used in the treatment of both hematological and solid malignancies. In the US alone, vincristine is used to treat over 50% of all childhood cancers and ~30 000 adults cancer patients.34 Vincristine-induced neurotoxicity has been found in 34.8% of Europeans vs 4.8% of African Americans (P = 0.007). Europeans have been shown to have a higher average grade of neurotoxicity (2.72 vs 1, P<0.0001) and require dose reduction (4% vs 0.1%, P<0.0001) and dose omission (1.2% vs 0.1%, P<0.01) when compared with African populations.48 The biotransformation of vincristine is CYP3A5-dependent.1 The current study found an increased frequency of CYP3A5 polymorphisms in African populations, which correlates with the reported increased in the expression of CYP3A5 in these populations when compared with Europeans (10–30% vs 60–70%).1,49–51 Increased CYP3A5 expression would increase the clearance of vincristine, thus lowering the concentration of the drug in the body and decrease the risk of vincristine-related toxicity in African populations. This could possibly be a mechanism by which African populations are protected from vincristine-related toxicity.
African populations experience more frequent antiretroviral and antimycobacterial drug-induced toxicities compared with Europeans. The incidence of nevirapine-induced hepatotoxicity is 17% among South Africans Blacks compared with 1–10% in Europeans, while 10% of Africans discontinue efavirenz therapy because of persistent toxicity compared with only 3% of Europeans.52 Also, 69% of Africans compared with 50% of European patients experience neurotoxicity after initiation of efavirenz therapy.52 This correlates with an overrepresentation of pharmacogenetic risk variants such as ABCC2 rs717620, ABCC2 rs17222723, ABCC4 rs1751034, CYP2B6 rs28399499, CYP2B6 rs3745274, APOE rs429358 and CYP2B6 rs3745274 in Africans compared with Europeans as observed in the current study. CYP2B6 rs3745274 is also associated with rifampicin-induced liver injury, an important drug used in antituberculous therapy.20,21 Even though differences in the incidence between Africans and Europeans are currently not known, this result indicates the possibility of such differences, which should be investigated in the future.
Overall, CYP3A variants showed the most significant differences and were more frequent in Africans compared with Europeans. CYP3A enzymes are involved in the metabolism of ~40–60% of all drugs.53 Their expression varies significantly by population, with increased expression of CYP3A5 in particular reported in African compared with European populations (10%–30% of Europeans vs 60%–70% of Africans).49–51 This observation is consistent with the observed dramatic enrichment of CYP3A5 variants in African compared with European populations in the current study. Renbarger has postulated that CYP3A5 gene region could explain most of the drug response differences by population.48 Other striking differences were observed with ATP-binding cassette and solute carrier transporters, indicating the additional contribution of other genes. In general, we observed an enrichment of genetic variants, which could underlie an enhanced predisposition to several ADRs and poor response rates in Africans compared with Europeans.
The current study also observed important differences among the African populations, which could translate to significant differences in drug efficacy and safety profiles, and also in the dose required to achieve the desired therapeutic effect in different African populations. This is consistent with the reported highly variable distribution of ABCB1, VKORC1 and CYP enzymes among African populations.54,55 This pharmacogenomic heterogeneity across different ethnic groups and geographical regions within African populations highlights the challenge faced by regulatory agencies in African countries when assessing new drug applications especially when there is minimal or no data from local clinical trials. Therefore, an important area of focus for improving drug distribution and access in African populations is the development and effective use of pharmacovigilance systems to monitor drug response in treated patients in order to avoid, in particular, serious and permanently disabling ADRs. Also, local trials to assess the frequency of ADRs will be important.
The current study places emphasis on the importance of including different populations in the development of biomarkers for pharmacogenetic testing, clinical practice guidelines, and clinical trials. To the best of our knowledge, it is the largest study of pharmacokinetic and pharmcodynamic genetic markers to dissect the pharmacogenomic variation at the level of individual populations and the first to have included such a large number of individuals recruited from different indigenous African populations. This study clearly demonstrates that clinical trials and safety studies, which are typically done in European populations, cannot be extrapolated to African populations. Furthermore, it also highlights the compounded challenge of population heterogeneity with respect to the delivery of health-care services in Canada and the USA. This type of study can inform clinical practice and clinical trials and is imperative for tailoring therapy towards individual populations. Our study population is not the complete representative sample of all African and European populations, but points to the need for local studies of genetic variants contributing to ADRs within all populations.
We have shown that there are important differences in the distribution of genetic variants in key drug biotransformation genes by population. These could translate to significant differences in drug response and toxicity rates. African populations have a pharmacogenetic enrichment to ADR susceptibility when compared with Europeans, which could explain the increased susceptibility and conferring poorer survival and response rates in these populations. This observation highlights the need for further investment in active drug surveillance systems, which should be central to all health-care systems to ensure each patient achieves maximal therapeutic benefit and minimal toxicity. Even though studies of pharmacogenomic differences among populations predicts the existence of drug response differences by populations, some of which have already being elucidated, a prospective evaluation of the relationship between pharmacogenomic diversity and drug response variability will be warranted to validate these findings. As population diversity, especially in Canada and the USA continues to increase, the need for information on population-specific genetic variation for the implementation of personalized medicine will become more important.
Supplementary Material
ACKNOWLEDGEMENTS
We acknowledged the participation of all those who took part in this study from the different Africa countries and Canada. We also acknowledge the contributions of Bill Beggs and other members of the Tishkoff Lab (University of Pennsylvania) and the Canadian Pharmacogenomics Network for Drug Safety consortium (University of British Columbia) for the enrolment of the study participants and for the handling of the samples, assays and records. This work was supported by the Canadian Institutes of Health Research, Child and Family Research Institute (Bertram Hoffmeister Postdoctoral Fellowship Award for Folefac Aminkeng), Canada Foundation for Innovation, Genome British Columbia. Additional funding was provided by Child & Family Research Institute (Vancouver, BC), Faculty of Medicine of the University of British Columbia, The Canadian Gene Cure Foundation and C17 Research Network and Childhood Cancer Foundation-Candlelighters Canada, as well as NSF (BCS-0827436) and NIH (R01GM076637, 8 DP1 ES022577-04) grants to S.A.T.
APPENDIX
THE CANADIAN PHARMACOGENOMICS NETWORK FOR DRUG SAFETY CONSORTIUM
The Canadian Pharmacogenomics Network for Drug Safety Consortium (Participants are arranged geographically by institution across Canada)—Vancouver, BC, Children’s Hospital, Child & Family Research Institute, CMMT, POPi: Michael Hayden, Bruce Carleton, Colin Ross, Stuart MacLeod, Wyeth Wasserman, Craig Mitton, Anne Smith, Claudette Hildebrand, Lucila Castro Pastrana, Reza Ghannadan, Rod Rassekh, Jonathan Lim, Fudan Miao, Henk Visscher, Kusala Pussegoda, Folefac Aminkeng, Michelle Higginson, Nasim Massah,Mojgan Yazdanpanah, Johanne Sistonen, Ricardo Jimenez, Adrienne Borrie, Ursula Amstutz, Shevaun Hughes, Kaitlyn Shaw; Calgary, Alberta Children’s Hospital: Cheri Nijssen-Jordan, David Johnson, Linda Verbeek, Rick Kaczowka, Patti Stevenson, Andrea Hurton; Edmonton, Stollery Children’s Hospital: Paul Grundy, Kent Stobart, Bev Wilson, Sunil Desai, Maria Spavor, Linda Churcher, Terence Chow; Winnipeg, Winnipeg Children’s Hospital: Kevin Hall, Nick Honcharik, Sara Israels, Shanna Chan, Byron Garnham, Michelle Staub; London, London Health Sciences Centre: Michael Rieder, Becky Malkin; Hamilton, McMaster Children’s Hospital: Carol Portwine, Amy Cranston; Toronto, Hospital for Sick Children: Gideon Koren, Shinya Ito, Paul Nathan, Mark Greenberg, Facundo Garcia Bournissen, Miho Inoue, Sachi Sakaguchi, Toshihiro Tanaka, Hisaki Fujii, Mina Ogawa, Ryoko Ingram, Taro Kamiya & Smita Karande; Kingston, Kingston General Hospital: Mariana Silva, Stephanie Willing; Ottawa, Children’s Hospital of Eastern Ontario: Régis Vaillancourt, Pat Elliott-Miller, Donna Johnston, Herpreet Mankoo, Elaine Wong, Brenda Wilson, Lauren O’Connor; Health Canada: Maurica Maher; Montreal, Hospital Sainte-Justine: Jean-Francois Bussie`res, Denis Lebel, Pierre Barret, Aure´lie Closon, Eve Coulson; Montreal Heart Institute: Marie-Pierre Dube´, Michael Phillips; McGill University Health Centre-Montreal Children’s Hospital: Nada Jabado, Anelise Espirito Santo, Martine Nagy; McGill University: Denise Avard; Halifax, IWK Health Centre: Margaret Murray, Darlene Boliver, Marilyn Tiller and Carolanne Osborne.
Footnotes
CONFLICT OF INTEREST
The authors declare no conflict of interest.
Supplementary Information accompanies the paper on the The Pharmacogenomics Journal website (http://www.nature.com/tpj)
REFERENCES
- 1.O’Donnell PH, Dolan ME. Cancer pharmacoethnicity: ethnic differences in susceptibility to the effects of chemotherapy. Clin Cancer Res. 2009;15:4806–4814. doi: 10.1158/1078-0432.CCR-09-0344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Phillips KA, Veenstra D, Van Bebber S, Sakowski J. An introduction to cost-effectiveness and cost-benefit analysis of pharmacogenomics. Pharmacogenomics. 2003;4:231–239. doi: 10.1517/phgs.4.3.231.22691. [DOI] [PubMed] [Google Scholar]
- 3.Ernst FR, Grizzle AJ. Drug-related morbidity and mortality: updating the cost-of-illness model. J Am Pharm Assoc (Wash) 2001;41:192–199. doi: 10.1016/s1086-5802(16)31229-3. [DOI] [PubMed] [Google Scholar]
- 4.Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998;279:1200–1205. doi: 10.1001/jama.279.15.1200. [DOI] [PubMed] [Google Scholar]
- 5.Yang JJ, Cheng C, Devidas M, Cao X, Fan Y, Campana D, et al. Ancestry and pharmacogenomics of relapse in acute lymphoblastic leukemia. Nat Genet. 2011;43:237–241. doi: 10.1038/ng.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kadan-Lottick NS, Ness KK, Bhatia S, Gurney JG. Survival variability by race and ethnicity in childhood acute lymphoblastic leukemia. JAMA. 2003;290:2008–2014. doi: 10.1001/jama.290.15.2008. [DOI] [PubMed] [Google Scholar]
- 7.Shord SS, Thompson DM, Krempl GA, Hanigan MH. Effect of concurrent medications on cisplatin-induced nephrotoxicity in patients with head and neck cancer. Anti-cancer drugs. 2006;17:207–215. doi: 10.1097/00001813-200602000-00013. [DOI] [PubMed] [Google Scholar]
- 8.Nyongesa C, Ruff P, Donde B, Kotzen J. A phase I study of concurrent cisplatin chemotherapy in patients with carcinoma of the cervix receiving pelvic radiotherapy. Int J Gynecol Cancer. 2006;16:1614–1619. doi: 10.1111/j.1525-1438.2006.00635.x. [DOI] [PubMed] [Google Scholar]
- 9.Krischer JP, Epstein S, Cuthbertson DD, Goorin AM, Epstein ML, Lipshultz SE. Clinical cardiotoxicity following anthracycline treatment for childhood cancer: the Pediatric Oncology Group experience. J Clin Oncol. 1997;15:1544–1552. doi: 10.1200/JCO.1997.15.4.1544. [DOI] [PubMed] [Google Scholar]
- 10.Hasan S, Dinh K, Lombardo F, Kark J. Doxorubicin cardiotoxicity in African Americans. J Natl Med Assoc. 2004;96:196–199. [PMC free article] [PubMed] [Google Scholar]
- 11.Hershman D, McBride R, Jacobson JS, Lamerato L, Roberts K, Grann VR, et al. Racial disparities in treatment and survival among women with early-stage breast cancer. J Clin Oncol. 2005;23:6639–6646. doi: 10.1200/JCO.2005.12.633. [DOI] [PubMed] [Google Scholar]
- 12.Polite BN, Dignam JJ, Olopade OI. Colorectal cancer model of health disparities: understanding mortality differences in minority populations. J Clin Oncol. 2006;24:2179–2187. doi: 10.1200/JCO.2005.05.4775. [DOI] [PubMed] [Google Scholar]
- 13.McCollum AD, Catalano PJ, Haller DG, Mayer RJ, Macdonald JS, Benson AB, 3rd, et al. Outcomes and toxicity in african-american and caucasian patients in a randomized adjuvant chemotherapy trial for colon cancer. J Natl Cancer Inst. 2002;94:1160–1167. doi: 10.1093/jnci/94.15.1160. [DOI] [PubMed] [Google Scholar]
- 14.Tishkoff SA, Reed FA, Friedlaender FR, Ehret C, Ranciaro A, Froment A, et al. The genetic structure and history of Africans and African Americans. Science. 2009;324:1035–1044. doi: 10.1126/science.1172257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Campbell MC, Tishkoff SA. African genetic diversity: implications for human demographic history, modern human origins, and complex disease mapping. Ann Rev Genomics Hum Genet. 2008;9:403–433. doi: 10.1146/annurev.genom.9.081307.164258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lachance J, Vernot B, Elbers CC, Ferwerda B, Froment A, Bodo JM, et al. Evolutionary history and adaptation from high-coverage whole-genome sequences of diverse african hunter-gatherers. Cell. 2012;150:457–469. doi: 10.1016/j.cell.2012.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Carleton B, Poole R, Smith M, Leeder J, Ghannadan R, Ross C, et al. Adverse drug reaction active surveillance: developing a national network in Canada’s children’s hospitals. Pharmacoepidemiol Drug Saf. 2009;18:713–721. doi: 10.1002/pds.1772. [DOI] [PubMed] [Google Scholar]
- 18.Carlson CS, Eberle MA, Rieder MJ, Yi Q, Kruglyak L, Nickerson DA. Selecting a maximally informative set of single-nucleotide polymorphisms for association analyses using linkage disequilibrium. Am J Hum Genet. 2004;74:106–120. doi: 10.1086/381000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.International HapMap Consortium. A haplotype map of the human genome. Nature. 2005;437:1299–1320. doi: 10.1038/nature04226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.PharmGKB. [Accessed 10th of December 2012];The Pharmacogenomics Knowledgebase (PharmGKB) is a comprehensive resource that curates knowledge about the impact of genetic variation on drug response for clinicians and researchers. http://www.pharmgkborg.
- 21. [Accessed 10 December, 2012];HIV-pharmacogenomics.org. http://www.hiv-pharmacogenomics.org.
- 22.Visscher H, Ross CJ, Rassekh SR, Barhdadi A, Dube MP, Al-Saloos H, et al. Phar-macogenomic prediction of anthracycline-induced cardiotoxicity in children. J Clin Oncol. 2012;30:1422–1428. doi: 10.1200/JCO.2010.34.3467. [DOI] [PubMed] [Google Scholar]
- 23.Wojnowski L, Kulle B, Schirmer M, Schluter G, Schmidt A, Rosenberger A, et al. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation. 2005;112:3754–3762. doi: 10.1161/CIRCULATIONAHA.105.576850. [DOI] [PubMed] [Google Scholar]
- 24.Blanco JG, Sun CL, Landier W, Chen L, Esparza-Duran D, Leisenring W, et al. Anthracycline-related cardiomyopathy after childhood cancer: role of polymorphisms in carbonyl reductase genes--a report from the Children’s Oncology Group. J Clin Oncol. 2012;30:1415–1421. doi: 10.1200/JCO.2011.34.8987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Blanco JG, Leisenring WM, Gonzalez-Covarrubias VM, Kawashima TI, Davies SM, Relling MV, et al. Genetic polymorphisms in the carbonyl reductase 3 gene CBR3 and the NAD(P)H:quinone oxidoreductase 1 gene NQO1 in patients who developed anthracycline-related congestive heart failure after childhood cancer. Cancer. 2008;112:2789–2795. doi: 10.1002/cncr.23534. [DOI] [PubMed] [Google Scholar]
- 26.Maggini V, Buda G, Martino A, Presciuttini S, Galimberti S, Orciuolo E, et al. MDR1 diplotypes as prognostic markers in multiple myeloma. Pharmacogenet Genomics. 2008;18:383–389. doi: 10.1097/FPC.0b013e3282f82297. [DOI] [PubMed] [Google Scholar]
- 27.Bray J, Sludden J, Griffin MJ, Cole M, Verrill M, Jamieson D, et al. Influence of pharmacogenetics on response and toxicity in breast cancer patients treated with doxorubicin and cyclophosphamide. British J Cancer. 2010;102:1003–1009. doi: 10.1038/sj.bjc.6605587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ross CJ, Katzov-Eckert H, Dube MP, Brooks B, Rassekh SR, Barhdadi A, et al. Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet. 2009;41:1345–1349. doi: 10.1038/ng.478. [DOI] [PubMed] [Google Scholar]
- 29.Khrunin AV, Moisseev A, Gorbunova V, Limborska S. Genetic polymorphisms and the efficacy and toxicity of cisplatin-based chemotherapy in ovarian cancer patients. Pharmacogenomics J. 2010;10:54–61. doi: 10.1038/tpj.2009.45. [DOI] [PubMed] [Google Scholar]
- 30.Cui LH, Yu Z, Zhang TT, Shin MH, Kim HN, Choi JS. Influence of polymorphisms in MTHFR 677 C-->T, TYMS 3R-->2R and MTR 2756 A-->G on NSCLC risk and response to platinum-based chemotherapy in advanced NSCLC. Pharmaco-genomics. 2011;12:797–808. doi: 10.2217/pgs.11.27. [DOI] [PubMed] [Google Scholar]
- 31.Van Kuilenburg AB, Vreken P, Abeling NG, Bakker HD, Meinsma R, Van Lenthe H, et al. Genotype and phenotype in patients with dihydropyrimidine dehy-drogenase deficiency. Human Genetics. 1999;104:1–9. doi: 10.1007/pl00008711. [DOI] [PubMed] [Google Scholar]
- 32.Gross E, Busse B, Riemenschneider M, Neubauer S, Seck K, Klein HG, et al. Strong association of a common dihydropyrimidine dehydrogenase gene polymorphism with fluoropyrimidine-related toxicity in cancer patients. PLoS One. 2008;3:e4003. doi: 10.1371/journal.pone.0004003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Etienne-Grimaldi MC, Milano G, Maindrault-Goebel F, Chibaudel B, Formento JL, Francoual M, et al. Methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms and FOLFOX response in colorectal cancer patients. Br J Clin Pharmacol. 2010;69:58–66. doi: 10.1111/j.1365-2125.2009.03556.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Egbelakin A, Ferguson MJ, MacGill EA, Lehmann AS, Topletz AR, Quinney SK, et al. Increased risk of vincristine neurotoxicity associated with low CYP3A5 expression genotype in children with acute lymphoblastic leukemia. Pediatr Blood Cancer. 2011;56:361–367. doi: 10.1002/pbc.22845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Patino-Garcia A, Zalacain M, Marrodan L, San-Julian M, Sierrasesumaga L. Methotrexate in pediatric osteosarcoma: response and toxicity in relation to genetic polymorphisms and dihydrofolate reductase and reduced folate carrier 1 expression. J Pediatr. 2009;154:688–693. doi: 10.1016/j.jpeds.2008.11.030. [DOI] [PubMed] [Google Scholar]
- 36.Krajinovic M, Robaey P, Chiasson S, Lemieux-Blanchard E, Rouillard M, Primeau M, et al. Polymorphisms of genes controlling homocysteine levels and IQ score following the treatment for childhood ALL. Pharmacogenomics. 2005;6:293–302. doi: 10.1517/14622416.6.3.293. [DOI] [PubMed] [Google Scholar]
- 37.Shrif NE, Won HH, Lee ST, Park JH, Kim KK, Kim MJ, et al. Evaluation of the effects of VKORC1 polymorphisms and haplotypes, CYP2C9 genotypes, and clinical factors on warfarin response in Sudanese patients. Eur J Clin Pharmacol. 2011;67:1119–1130. doi: 10.1007/s00228-011-1060-1. [DOI] [PubMed] [Google Scholar]
- 38.Geisen C, Watzka M, Sittinger K, Steffens M, Daugela L, Seifried E, et al. VKORC1 haplotypes and their impact on the inter-individual and inter-ethnical variability of oral anticoagulation. Thromb Haemost. 2005;94:773–779. doi: 10.1160/TH05-04-0290. [DOI] [PubMed] [Google Scholar]
- 39.Rocha V, Porcher R, Fernandes JF, Filion A, Bittencourt H, Silva W, Jr, et al. Association of drug metabolism gene polymorphisms with toxicities, graft-versus-host disease and survival after HLA-identical sibling hematopoietic stem cell transplantation for patients with leukemia. Leukemia. 2009;23:545–556. doi: 10.1038/leu.2008.323. [DOI] [PubMed] [Google Scholar]
- 40.Kremer LC, Caron HN. Anthracycline cardiotoxicity in children. N Engl J Med. 2004;351:120–121. doi: 10.1056/NEJMp048113. [DOI] [PubMed] [Google Scholar]
- 41.Kremer LC, van der Pal HJ, Offringa M, van Dalen EC, Voute PA. Frequency and risk factors of subclinical cardiotoxicity after anthracycline therapy in children: a systematic review. Ann Oncol. 2002;13:819–829. doi: 10.1093/annonc/mdf167. [DOI] [PubMed] [Google Scholar]
- 42.van der Pal HJ, van Dalen EC, Hauptmann M, Kok WE, Caron HN, van den Bos C, et al. Cardiac function in 5-year survivors of childhood cancer: a long-term follow-up study. Arch Intern Med. 170:1247–1255. doi: 10.1001/archinternmed.2010.233. [DOI] [PubMed] [Google Scholar]
- 43.Kremer LC, van Dalen EC, Offringa M, Voute PA. Frequency and risk factors of anthracycline-induced clinical heart failure in children: a systematic review. Ann Oncol. 2002;13:503–512. doi: 10.1093/annonc/mdf118. [DOI] [PubMed] [Google Scholar]
- 44.McKeage MJ. Comparative adverse effect profiles of platinum drugs. Drug Saf. 1995;13:228–244. doi: 10.2165/00002018-199513040-00003. [DOI] [PubMed] [Google Scholar]
- 45.Coradini PP, Cigana L, Selistre SG, Rosito LS, Brunetto AL. Ototoxicity from cisplatin therapy in childhood cancer. J Pediatr Hematol Oncol. 2007;29:355–360. doi: 10.1097/MPH.0b013e318059c220. [DOI] [PubMed] [Google Scholar]
- 46.Kushner BH, Budnick A, Kramer K, Modak S, Cheung NK. Ototoxicity from high-dose use of platinum compounds in patients with neuroblastoma. Cancer. 2006;107:417–422. doi: 10.1002/cncr.22004. [DOI] [PubMed] [Google Scholar]
- 47.Knight KR, Kraemer DF, Neuwelt EA. Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. J Clin Oncol. 2005;23:8588–8596. doi: 10.1200/JCO.2004.00.5355. [DOI] [PubMed] [Google Scholar]
- 48.Renbarger JL, McCammack KC, Rouse CE, Hall SD. Effect of race on vincristine-associated neurotoxicity in pediatric acute lymphoblastic leukemia patients. Pediatr Blood Cancer. 2008;50:769–771. doi: 10.1002/pbc.21435. [DOI] [PubMed] [Google Scholar]
- 49.Wrighton SA, Brian WR, Sari MA, Iwasaki M, Guengerich FP, Raucy JL, et al. Studies on the expression and metabolic capabilities of human liver cytochrome P450IIIA5 (HLp3) Mol Pharmacol. 1990;38:207–213. [PubMed] [Google Scholar]
- 50.Kuehl P, Zhang J, Lin Y, Lamba J, Assem M, Schuetz J, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27:383–391. doi: 10.1038/86882. [DOI] [PubMed] [Google Scholar]
- 51.Lee SJ, Usmani KA, Chanas B, Ghanayem B, Xi T, Hodgson E, et al. Genetic findings and functional studies of human CYP3A5 single nucleotide polymorphisms in different ethnic groups. Pharmacogenetics. 2003;13:461–472. doi: 10.1097/00008571-200308000-00004. [DOI] [PubMed] [Google Scholar]
- 52.Subbaraman R, Chaguturu SK, Mayer KH, Flanigan TP, Kumarasamy N. Adverse effects of highly active antiretroviral therapy in developing countries. Clin Infect Dis. 2007;45:1093–1101. doi: 10.1086/521150. [DOI] [PubMed] [Google Scholar]
- 53.Vazquez MA. Southwestern Internal Medicine Conference. New advances in immunosuppression therapy for renal transplantation. Am J Med Sci. 1997;314:415–435. doi: 10.1097/00000441-199712000-00012. [DOI] [PubMed] [Google Scholar]
- 54.Dandara C, Lombard Z, Du Plooy I, McLellan T, Norris SA, Ramsay M. Genetic variants in CYP (−1A2, −2C9, −2C19, −3A4 and −3A5), VKORC1 and ABCB1 genes in a black South African population: a window into diversity. Pharmacogenomics. 2011;12:1663–1670. doi: 10.2217/pgs.11.106. [DOI] [PubMed] [Google Scholar]
- 55.Sistonen J, Fuselli S, Palo JU, Chauhan N, Padh H, Sajantila A. Pharmacogenetic variation at CYP2C9, CYP2C19, and CYP2D6 at global and microgeographic scales. Pharmacogenet Genomics. 2009;19:170–179. doi: 10.1097/FPC.0b013e32831ebb30. [DOI] [PubMed] [Google Scholar]
- 56.Evans WE. Pharmacogenetics of thiopurine S-methyltransferase and thiopurine therapy. Ther Drug Monit. 2004;26:186–191. doi: 10.1097/00007691-200404000-00018. [DOI] [PubMed] [Google Scholar]
- 57.Krynetski EY, Schuetz JD, Galpin AJ, Pui CH, Relling MV, Evans WE. A single point mutation leading to loss of catalytic activity in human thiopurine S-methyl-transferase. Proc Natl Acad Sci USA. 1995;92:949–953. doi: 10.1073/pnas.92.4.949. [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.