Abstract
The 18th World Congress of Basic and Clinical Pharmacology (WCP2018), coordinated by IUPHAR and hosted by the Japanese Pharmacological Society and the Japanese Society of Clinical Pharmacology and Therapeutics, was held in July 2018 at the Kyoto International Conference Center, in Kyoto, Japan. Having as its main theme ‘Pharmacology for the Future: Science, Drug Development and Therapeutics’, WCP2018 was attended by over 4500 delegates, representing 78 countries. The present report is an overview of a symposium at WCP2018, entitled Pharmacogenomics in Special Populations, organized by IUPHAR´s Pharmacogenetics/Genomics (PGx) section. The PGx section congregates distinguished scientists from different continents, covering expertise from basic research, to clinical implementation and ethical aspects of PGx, and one of its major activities is the coordination of symposia and workshops to foster exchange of PGx knowledge (https://iuphar.org/sections‐subcoms/pharmacogenetics‐genomics/). The symposium attracted a large audience to listen to presentations covering various areas of research and clinical adoption of PGx in Oceania, Africa, Latin America and Asia.
Keywords: Africa, Asia, Latin America, Oceania, Pharmacogenomics diversity
Introduction
The 18th World Congress of Basic and Clinical Pharmacology (WCP2018), coordinated by IUPHAR and hosted by the Japanese Pharmacological Society and the Japanese Society of Clinical Pharmacology and Therapeutics, was held in July 2018 at the Kyoto International Conference Center, in Kyoto, Japan. Having as its main theme ʻPharmacology for the Future: Science, Drug Development and Therapeuticsʼ, WCP2018 was attended by over 4500 delegates, representing 78 countries.
The present report is an overview of a symposium at WCP2018, entitled Pharmacogenomics in Special Populations, organized by IUPHAR's Pharmacogenetics/Genomics (PGx) section. The PGx section congregates distinguished scientists from different continents, covering expertise from basic research, to clinical implementation and ethical aspects of PGx, and one of its major activities is the coordination of symposia and workshops to foster exchange of PGx knowledge (https://iuphar.org/sections‐subcoms/pharmacogenetics‐genomics/). The symposium attracted a large audience to listen to presentations covering various areas of research and clinical adoption of PGx in Oceania, Africa, Latin America and Asia.
Pharmacogenetics in the Oceania region: precision medicine challenges
Pharmacogenetics in the Oceania region: precision medicine challenges was presented by Professor Andrew Somogyi (University of Adelaide, Adelaide, Australia). Oceania, comprising the large islands of Australia and New Zealand (Australasia), and Papua New Guinea (PNG), and a very large number of relatively small islands of Melanesia, Micronesia and Polynesia, covers over 8.5 million square kilometres, but only contains about 40 million inhabitants. PGx studies of the inhabitants of PNG have been recently reviewed 1, whereas for Aboriginal Australians and Melanesian and Polynesian peoples, only the genes encoding cytochrome P450 (CYP) CYP2D6 and CYP2C19 have been investigated to a certain extent 2, 3. In general, results cannot be readily predicted from one region to another. For example, the frequency of CYP2C19*2 varies from 45% in PNG to 24% in Aboriginal Australian and Maori peoples, whereas CYP2C19*3, another nonfunctional allele, ranges from 2% in Maori individuals to about 20% in PNG and Australian Aborigines. The CYP2C19*17 allele frequency is much lower in the latter population, resulting in a predicted 50% lower frequency of increased enzyme function compared with Caucasians.
The CYP2D6 genotype and predicted phenotype are dependent on copy number and sequence variation detection platforms used; nevertheless, it appears that poor metabolizers (PMs) comprise only about 2% over all of Oceania. This may have implications for CYP2D6‐catalysed primaquine dosing for Plasmodium vivax malaria. Indeed, the impact of CYP2D6 polymorphism on the effectiveness of primaquine to prevent P. vivax malaria relapses was discussed in another presentation at the symposium (see below).
In PNG, most PGx studies have focused on infectious diseases, and results pertinent to the antiretroviral agent efavirenz in HIV‐infected patients were presented. Efavirenz is mainly metabolized by CYP2B6, and poor metabolizer status is associated with central nervous system (CNS)/psychiatric effects. The frequency of the major CYP2B6 variant *6 is about 60% in PNG, compared with less than 20% in Caucasian and South Asian populations. Data from 52 PNG subjects, most of whom had CNS/psychiatric adverse effects, revealed, however, that only drowsiness was related to CYP2B6*6 carrier status.
Regarding N‐acetyltransferase 2 (NAT2) and acetylator status, no genomic studies have been conducted in PNG but almost all individuals are rapid acetylators, and therefore the incidence of isoniazid‐induced hepatotoxicity is rare, although patients might be being underdosed. In Australian Aborigines, about one‐third are slow acetylators and have a relatively high frequency of the NAT2*7 allele, at 40% compared with 1% in Europeans 4.
Minimal data are available on drug transporters in Oceania; however, the frequency of the gene encoding ATP‐binding cassette subfamily B member 1 (ABCB1) 3435C > T variant was 67% in PNG subjects, and even higher, at 71%, in Australian Aborigines. These are substantially above the 40–50% range found in Europeans and South and East Asians. Such high allele variant frequencies in peoples from Oceania may have major implications for the efficacy of, and adverse effects associated with, many medicines which are ABCB1 substrates.
The gene encoding human leukocyte antigen (HLA) B*13:01 is associated with severe hypersensitivity reactions [Stevens–Johnson syndrome (SJS); toxic epidermal necrolysis (TEN); and drug reaction with eosinophilia and systemic symptoms (DRESS) to phenytoin, the frequency of which is relatively high in several South Asian countries]. In PNG and in Aboriginal Australians from Northern Australia, the HLA‐B*13:01 frequency is almost 25%. Another variant, HLA‐B*56:02, has been associated with phenytoin‐induced DRESS and several case reports of phenytoin‐associated morbidity and mortality. The frequency of this allele can be over 5% in Aboriginal Australians, but is essentially absent in Europeans.
Although the frequencies of some important pharmacogenes are markedly different in Oceania (especially in PNG and in Aboriginal Australians) compared with Caucasian and some Asian populations, these frequencies can be fairly divergent across the region. Many important genes and genotype–phenotype correlations have not been assessed, with clinical relevance and translation assessment faced by limited local resources. Caution should be exercised when interpreting the genotype with respect to the phenotype, with the vexing issue that alleles rarely found in Europeans may be common in Oceania.
The challenges in conducting PGx studies are, firstly, ethical, in terms of demonstrating that PGx testing will help and not hinder the health of indigenous peoples in Oceania; and, secondly, presenting evidence that the efficacy and toxicity of some drugs can be different, as now shown with phenytoin in Aboriginal Australians. Having indigenous precision medicine champions with community support who can drive the research direction is critical for drug therapy optimization. In PNG, logistics are a major challenge, as biological sampling is often conducted in remote communities; thus, sample collection, processing and transport are problematic. The results to date and the above challenges result in research being needed to address cost‐effective and nondiscriminatory precision medicine for the understudied indigenous peoples of Oceania.
African Pharmacogenomics Research Consortium: Focus on HIV, tuberculosis and malaria treatment
African Pharmacogenomics Research Consortium: Focus on HIV, tuberculosis and malaria treatment was presented by Professor Eleni Aklillu (Karolinska Institutet, Stockholm, Sweden). Populations of sub‐Saharan Africa (SSA) are the most genetically and ethnically diverse in the world, displaying extensive population substructure and less linkage disequilibrium between loci compared with peoples of non‐African ancestry 5. This wide genetic heterogeneity in African populations offers the opportunity to identify rare alleles and haplotypes that play a role in determining susceptibility to diseases and adverse drug responses. The African Pharmacogenomics Consortium was established to prompt PGx research and clinical implementation in African populations 6. Through regional and international research collaborations, PGx research in Africa is progressing to genomics and environmental determinants of common diseases, with the goal of improving the health of African populations 7. Policy makers in Africa have started using PGx data to recommend population‐specific treatment guidelines. For instance, CYP2D6 metabolizes codeine into its active metabolite, morphine, and CYP2D6 ultrarapid metabolizers may experience the symptoms of morphine overdose, which include life‐threatening or fatal respiratory depression 8. The US Federal Drug Administration (FDA) issued a safety alert on the use of codeine for pain in children, particularly ultrarapid metabolizers 9, which is fairly common (29% functional CYP2D6 gene duplication frequency) in those with Ethiopian origin 10. Later, the Ethiopian Food, Medicine and Health Care Administration and Control Authority banned codeine use in the country.
SSA carries a high burden of HIV/AIDS, tuberculosis (TB) and malaria. Coinfection is common, and cotreatment is challenging owing to drug interactions and overlapping toxicities. Genetic and environmental factors, nutrition, coinfection, comorbidity and use of herbal medicines may result in different drug–response profiles. Specific variant alleles such as CYP2D6*17, CYP3A5*6, CYP3A*7 or the occurrence of decreased function alleles such as CYP2B6*6 at a higher frequency may account for population‐specific phenotype variations in peoples with black African ancestry 11, 12. Key PGx studies in different African populations, with a focus on drug interactions between first‐line antiretroviral, anti‐TB and antimalarial drugs, and their impact on treatment outcomes, were presented at the symposium. The need for studies in different African populations was highlighted as the extensive genetic diversity of SSA restricts extrapolation of PGx data among countries 13, 14.
Efavirenz, a potent non‐nucleoside reverse transcriptase inhibitor and a cornerstone first‐line antiretroviral drug in Africa, is metabolized mainly by CYP2B6. CYP2B6*6, a decreased‐function variant allele causing higher plasma efavirenz exposure, is implicated in drug‐induced hepatotoxicity 15, 16, 17 and neuropsychiatric toxicities 18, 19. CYP2B6*6 occurs at a higher frequency in people of African origin, reaching up to 50% 20, 21. CYP2B6*18, another decreased‐function variant allele associated with high efavirenz plasma concentrations, occurs with an allele frequency of up to 17% in Black Africans but is absent in Asians and Caucasians.
PGx studies in genes coding for HLA revealed that HLA‐B*57 alleles (B*57:03 and B*57:02) confer susceptibility to anti‐TB and antiretroviral drug‐induced liver toxicity 22. HLA‐B*57:03 is the most prevalent HLA‐B*57 subtype in black African populations, whereas HLA‐B*57:01, which is strongly associated with abacavir‐induced hypersensitivity reaction, is absent in black Africans 22. HLA‐B*57:01 occurs with frequencies up to 5% in Asians and Caucasians, and prior screening for this allele is highly recommended in these populations.
The importance of pharmacogenetic variations in modulating interactions between antiretroviral and anti‐TB drugs was noted. A distinct population‐specific efavirenz–rifampicin drug interaction profile required varying efavirenz dose recommendations for TB‐HIV patients during rifampicin‐based anti‐TB cotreatment. Rifampicin, a potent inducer of efavirenz‐metabolizing enzymes (CYP2B6, CYP2A6 and CYP3A4), significantly reduces plasma efavirenz concentration in white individuals 23, 24. Accordingly, the FDA and the British HIV Association recommend that the dose of efavirenz is increased (to 800 mg day–1) during rifampicin cotherapy 25, 26, 27. However, studies in African patients reported no significant effect of rifampicin‐based anti‐TB therapy on efavirenz plasma concentration 28, 29, 30, 31, and the World Health organization recommends the standard 600 mg day–1 efavirenz dose during rifampicin–efavirenz cotreatment 32. Recent studies reported that even the standard 600 mg daily efavirenz dose is unnecessarily high for SSA populations, and suggested lowering the efavirenz dose based on CYP2B6 genotype 33, 34. Future randomized, clinical trials are required to confirm that the proposed CYP2B6 genotype‐based efavirenz dosing strategy is safe and effective for clinical application in Africa.
Few recent studies have investigated the pharmacogenetics of antimalarial drugs 35 and its relevance for antiretroviral and antimalarial drug interaction in HIV–malaria coinfected patients receiving dual therapy in Africa 36, 37. Lumefantrine, a long‐acting antimalarial drug, is metabolized by CYP3A4, an enzyme inducible by efavirenz 38. Efavirenz‐based antiretroviral cotreatment significantly reduces lumefantrine plasma exposure, leading to a poor malaria treatment response, particularly in CYP2B6 slow metabolizers 36.
Overall, many pharmacogenetic studies in African patients are done using a candidate gene approach, and genome‐wide association studies (GWAS) in well‐characterized matched case–control cohorts are scarce. The available few GWAS revealed potential novel genetic biomarkers for antiretroviral and anti‐TB drug‐induced liver toxicities 39.The HLA‐C*04:01 allele is associated with nevirapine‐induced SJS/TEN in sub‐Saharan Africans 40. Currently, most genome‐wide single nucleotide polymorphism (SNP) arrays are designed for European ancestry, and the tag SNPs are less efficient for non‐Europeans, particularly for SSA populations 41, 42. The lack of cost‐effective, high‐throughput genotyping technologies and microarrays with good SNP density coverage specific for African ancestry remains a challenge for the progress of PGx in Africa 42, 43. Over the past two decades, PGx research in Africa has generated ample knowledge on variant alleles that determine patient variability in the plasma exposure and treatment outcome of drugs used to treat HIV, TB and malaria. There is a serious need to reduce the burden of HIV, TB and malaria on the continent, and wide‐scale pharmacogenetic research in different African countries will significantly contribute to improving patient care and optimize treatment outcomes. The development of a point‐of‐care genotype facility to guide drug dosing is imperative to promote the translation of PGx research into clinical practice in Africa.
Pharmacogenomic implications of population admixture in the Americas
The Pharmacogenomic implications of population admixture in the Americas, especially Latin America, were examined by Professor Guilherme Suarez‐Kurtz (Brazilian Pharmacogenetics Network and Brazilian National Cancer Institute, Rio de Janeiro, Brazil). The first part of the talk expanded on the notion that PGx research and implementation in Latin America must take into account the extensive genetic heterogeneity of the population. Although Native American, European and sub‐Saharan African biogeographical ancestries are common to all Latin American countries, the relative proportions of each ancestral root vary across nations, and, most importantly, within each nation. This was illustrated with data from Brazil and Mexico, the two most populous Latin American countries, with a remarkably distinct population structure: European and sub‐Saharan ancestry predominate largely over Native American ancestry in Brazilians 44, 45, whereas in Mexico the main ancestral contributions are European and Native American, with a relatively small African contribution 46, 47. However, the individual proportions of biogeographical ancestry vary widely and, most importantly, in a continuous pattern, irrespective of self‐reported race/colour in Brazilians or geographical origin in Mexicans 48. Corollaries to this pattern are: (i) there are no ʻcutoff pointsʼ for race/colour discrimination in Brazilians or associated with geographical origin in Mexicans; (ii) average proportions of biogeographical ancestry are not predictive of the corresponding proportions at the individual level in Brazilians and Mexicans, and most likely other Latin American peoples; and (iii) combining Latin Americans into a single population entity (e.g. Latinos or Hispanics in the US) is inappropriate in view of their diversity and heterogeneity.
Professor Suarez‐Kurtz then moved on to the more formal PGx arena, and discussed the clinical implications of population admixture, using data from his studies in Brazilians. The first example related to the association of CYP3A5 polymorphisms and tacrolimus dose requirement in renal transplant patients. Santoro et al. 49 confirmed the impact of CYP3A5*3 on tacrolimus dose requirement, previously described in other populations, but disclosed two novel and important findings: first, the association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing. Second, the inclusion of Brazilian patients with the CYP3A5*1/*1 genotype, which is rare or absent in populations of European or Asian descent, allowed demonstration of a gene–dose effect of CYP3A5 polymorphisms on tacrolimus pharmacokinetics. This study provided the basis for inclusion of CYP3A5*6 and *7, in addition to the *3 allele, in the Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for tacrolimus 50.
The second example presented at the symposium pertained to warfarin. Prior to the original CPIC guideline for warfarin, published in 2011 51, Brazilian researchers 52, 53 developed and validated PGx algorithms for warfarin dosing, which explained 51–60% of the variability in stable dose requirement. Importantly, these algorithms performed equally well in self‐reported White and Black individuals, which is in marked contrast to the considerably poorer performance of several warfarin algorithms in Black Africans and African Americans vs. Europeans and White Americans 54, 55. This discrepancy was explained by the considerably higher allele frequency of the major PGx variables used in most algorithms – namely, the gene encoding vitamin K epoxide reductase complex subunit‐1 (VKORC1), rs9923231 and decreased‐function CYP2C9*2 and *3 alleles – in the extensively admixed Black Brazilians compared with Black Africans or African‐Americans 52, 56, 57. Of note, the 2017 updated CPIC guideline for warfarin 58 incorporated dosing recommendations for warfarin ʻthat are specific for continental ancestryʼ. Nevertheless, continental ancestry in admixed populations is not a categorical, but rather a continuous variable, even within self‐reported race/colour categories 56, 57. This is a caveat to the implementation of guidelines based on continental ancestry – as well as ʻraceʼ or ethnicity – in Brazil and most likely other Latin American nations.
The next topic in the presentation was the distribution of the nudix hydrolase 15 gene (NUDT15) rs116855232 (g.415C > T) SNP among Latin American populations. The variant T allele, associated with intolerance to thiopurines, is reported to be most common in East Asians and ʻHispanicsʼ, but appears to be rare or absent in Europeans and Africans 59, 60, with average frequencies of 10%, 4%, 0% and 0%, respectively, in the corresponding East Asian, admixed American, European and African groups of the 1000 Genomes Project. Suarez‐Kurtz and colleagues used the 1000 Genome Project markers to estimate the individual ancestry of the admixed American group and observed that the frequency of the rs116855232 T allele increases in parallel with the increase in the average proportion of Native American ancestry in the four subpopulations (Puerto Ricans, Colombians, Mexicans and Peruvians) comprising the admixed American group. This pattern is consistent with the notion that Asia is the likely origin of the first migrants into the American continent, 25 000–15 000 years ago.
In the ensuing discussion, the association of CYP2D6 polymorphism with the antimalarial effect of primaquine mentioned earlier by Professor Somogyi prompted reference to the recent demonstration of an increased risk of relapse of P. vivax malaria after chloroquine–primaquine combined therapy in patients from the Brazilian Amazon who were carriers of reduced‐function CYP2D6 alleles 61. The relapses were ascribed to a failure of primaquine hypnozoiticidal activity, which requires CYP2D6‐catalysed conversion of primaquine, a prodrug, into redox‐active metabolite(s).
Pharmacogenomics research and its implementation in Asia
Pharmacogenomics research and its implementation in Asia was reviewed by Dr Yoshiro Saito (National Institute of Health Sciences, Kawasaki, Japan). PGx research has been performed extensively in the Asian region, as summarized by Ang et al. 62. Publications focused on PGx are increasing in number in Asian countries/regions, particularly in relation to the CNS, cancer and blood. The majority of studies are national (local), with only a small number of collaborations across Asian countries/regions. Although most Asian publications were replication studies, several studies, primarily from Japanese and Korean groups, have reported novel findings. At the symposium, two examples were presented in which Asian researchers contributed significantly.
The first example relates to drug‐induced severe cutaneous adverse reactions (SCARs), such as SJS and TEN, important issues for drug safety. Although SJS/TEN is very rare, it requires hospitalization and, even after recovery, patients may suffer from severe sequelae such as vision loss 63. Chung and collaborators in Taiwan, Thailand, mainland China, Hong Kong, Malaysia, Singapore, Philippines and Japan (including Saito's group) assessed SJS/TEN cases using the algorithm of drug causality for epidermal necrolysis (ALDEN) score (≥4), resulting in the identification of 1028 verified cases 64. The top suspected drug classes were antiepileptic and antipsychotic agents, followed by antibiotics/antiviral drugs, and allopurinol. Lamotrigine was the leading suspected drug in Japan, whereas carbamazepine and allopurinol were the primary culprits in mainland China, Thailand, Malaysia, Singapore, Philippines and Taiwan.
In Chinese, Thai, Korean, Japanese, as well as Caucasian populations, allopurinol‐related SJS/TEN/hypersensitivity syndrome is associated with HLA‐B*58:01, which displayed relatively high frequencies in East, South‐East and South Asian populations (except for Japanese) compared with Caucasians 63, 65. The first report, from Taiwan, revealed a high odds ratio (580) for an association of SJS/TEN with HLA‐B*58:01 in the Han Chinese 66. Screening for HLA‐B*58:01 prior to starting allopurinol administration prevented allopurinol‐induced SCARs in Taiwan 67. Accordingly, pre‐emptive testing is highly recommended in the allopurinol label in Taiwan, whereas the labels in Singapore (and the UK) state that prescreening should be considered for high‐risk (high‐frequency) populations, and the Japanese label refers to studies of the causal association of HLA‐B*58:01 with the SJS/TEN/hypersensitivity syndrome.
Carbamazepine‐related SJS/TEN is associated with HLA‐A*31:01 and HLA‐B75 (such as HLA‐B*15:02 and B‐15:11). In addition, milder skin reactions are also linked to HLA‐A*31:01, which occurs in relatively high frequency in Japanese and Korean populations 65. HLA‐B75 (especially HLA‐B*15:02) is prevalent in Chinese and South‐East Asians 68. HLA‐B*15:02 was most strongly associated with carbamazepine‐induced SJS/TEN in Han Chinese in Taiwan, with an odds ratio of 1357 69. Testing for HLA‐B15:02 prior to carbamazepine administration completely prevented the onset of SJS/TEN in Taiwanese patients 70, whereas pre‐emptive testing for HLA‐A*31:01 significantly reduced the rate of carbamazepine‐related adverse skin reactions in Japanese patients 71. Table 1 compares the information on prescreening tests for HLA‐B*15:02 and HLA‐A*31:01 in the carbamazepine package inserts in different Asian countries, and, for comparison, Australia, Canada and the US. In Taiwan and Singapore, the labels state that the pre‐emptive testing is highly recommended for HLA‐B*15:02 and should be considered for HLA‐A*31:01 in patients of Asian ancestry or ancestry in a genetically at‐risk population. The Japanese label refers to studies of the association between these HLA variants and carbamazepine‐induced SCARs, but does not include recommendations for PGx testing 72. Of note, testing for HLA‐B*15:02 is covered by the national health insurance in Taiwan.
Table 1.
HLA type | Country | Description of prescreening HLA test in the package inserts (section) |
---|---|---|
B*15:02 | Taiwan | (Warning and precautions) Highly recommended in patients of Asian ancestrya |
Singapore | (Warning and precautions) Highly recommended in patients of Asian ancestry | |
Japan | (Side effects) HLA–SCAR association mentioned by referenced papers | |
USA | (Box warning) HLA test should be performed in patients with ancestry in genetically at‐risk populations | |
Canada | (Special warning and precautions) HLA test to be considered in patients with ancestry in genetically at‐risk populations | |
Australia | (Precautions) HLA test should be considered in patients with ancestry in genetically at‐risk populations | |
A*31:01 | Taiwan | (Warning and precautions) HLA test should be considered in patients with ancestry in genetically at‐risk populations |
Singapore | (Warning and precautions) HLA test should be considered in patients with ancestry in genetically at‐risk populations | |
Japan | (Side effects) HLA–SCAR association mentioned by referenced papers | |
USA | (Warning) The risks and benefits of carbamazepine therapy should be weighed | |
Canada | (Special warning and precautions) HLA test to be considered in patients with ancestry in genetically at‐risk populations | |
Australia | (Precautions) HLA test should be considered in patients with ancestry in genetically at‐risk populations |
HLA, human leukocyte antigen; SCAR, severe cutaneous adverse reaction
Pharmacogenetics/genomics test reimbursed by national health insurance
Regarding the PGx of other drug‐induced SCARs, an association of CYP2C9*3 with phenytoin‐related SCARs was first reported in Han Chinese, Japanese and Malay populations 73 and then replicated in Thailand 74. CYP2C9 is the major metabolizing enzyme of phenytoin, and CYP2C9*3 is a reduced‐function allele. High plasma concentrations after phenytoin withdrawal were observed in SCAR patients 73. The CYP2C9*3 frequency is higher in South Asian and European populations than in East and South‐East Asians 65. A dapsone‐related SCAR has been associated with HLA‐B*13:01 in Han Chinese and Tai populations 75, 76.
The second example of significant contributions of Asian researchers to PGx knowledge presented at the symposium referred to the anticancer agent, irinotecan, a prodrug which requires in vivo conversion into the active metabolite, SN‐38. SN‐38 is a substrate of UDP glucuronosyltransferase family 1 member A1 (UGT1A1), encoded by the polymorphic UGT1A1 gene. Patients with the UGT1A1*6 and UGT1A1*28 alleles have reduced glucuronidation of SN‐38 and therefore may be at increased risk of irinotecan toxicity. UGT1A1*6 is prevalent in East and South‐East Asian populations, whereas UGT1A1*28 occurs at higher frequencies in South Asian and Caucasian populations than in East and South‐East Asians 65. Both alleles were shown to associate with irinotecan‐related severe neutropenia in Japanese people 77, and a meta‐analysis of 20 irinotecan studies in East Asians confirmed this association, with an odds ratio of 3.3 78. Table 2 contrasts the UGT1A1 PGx information in the irinotecan package inserts in Japan, Singapore and Taiwan, and selected non‐Asian countries. Although pre‐emptive testing is not recommended in any of the package inserts, most provide information regarding the association of UGT1A1 genotypes with intolerance to irinotecan.
Table 2.
Country | UGT1A1 pharmacogenomic information in package insert (section) |
---|---|
Taiwan | (Warning and precautions) UGT1A1*28 homozygotes should be administered the normally indicated starting dose. The haematological toxicity of the patients should be monitored |
Singapore | (Warning and precautions) Patients with UGT1A1*6 or *28 homozygotes or compound heterozygotes may be at increased risk of serious adverse reactions, especially neutropenia, caused by reduced glucuronidation of SN‐38. Caution should be exercised when administering irinotecan to such patients. The haematological toxicity of the patients should be monitored |
Japan | (Pharmacokinetics) UGT1A1*6/*28 – neutropenia association is mentioned in referenced papersa |
USA | (Warning and precautions) UGT1A1*28 homozygotes are at increased risk of neutropenia |
(Dosage and administration) A reduction in the starting dose by at least one level should be considered for UGT1A1*28 homozygous patients | |
Canada | (Warning and precautions) A reduced starting dose should be considered for UGT1A1*28 homozygous patients |
UK | (Pharmacodynamic properties) UGT1A1*28 homozygotes should be administered the normally indicated starting dose. A reduced starting dose should be considered for patients who have experienced haematological toxicity with previous treatment |
Australia | (Precautions) Same as in the UK |
Pharmacogenetics/genomics test reimbursed by national health insurance
In summary, this presentation showed selected examples of genetic polymorphisms related to drug efficacy and adverse reactions in Asian populations. Dr Saito highlighted the increasing importance of collaborative pharmacogenomic research among Asian countries/regions, and suggested that further efforts aimed toward the construction of an Asian PGx research network should be a priority.
Collectively, the presentations at this symposium confirmed that PGx information is an important factor for population differences in the pharmacokinetics, efficacy and safety of drugs. In addition to the International Conference on Harmonization of Technical Requirements for the International Council for Harmonisation (ICH) of Technical Requirements for Pharmaceuticals for Human Use E5 guideline on ethnic factors 79, two related ICH guidelines were adopted and issued in 2017 on multiregional clinical trials (E17) 80 and on genomic sampling and management of genomic data (E18) 81. A harmonized PGx‐specific guideline describing ethnic specificity would be a necessary next step.
Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY 82, and are permanently archived in the Concise Guide to PHARMACOLOGY 2017/18 83.
Competing Interests
All authors have completed the Unified Competing Interest Form at www.icmje.org/coi_disclosure.pdf, and declare: G.S.K. receives grant support from the Brazilian agencies Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) and Departamento de Ciência e Tecnologia, Ministério da Saúde (DECIT); E.A. receives a grant from the European and Developing Countries Clinical Trial Partnership (EDCTP), Swedish Research Council and Swedish International Development Agency (SIDA); Y.S. receives grants from the Japan Agency for Medical Research and Development and JSPS KAKENHI; A.S. receives grant support from the National Health and Medical Research Council of Australia. None of the authors had any financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years, and no other relationships or activities that could appear to have influenced the submitted work.
Suarez‐Kurtz, G. , Aklillu, E. , Saito, Y. , and Somogyi, A. A. (2019) Conference report: pharmacogenomics in special populations at WCP2018. Br J Clin Pharmacol, 85: 467–475. 10.1111/bcp.13828.
References
- 1. Tucci JD, Pumuye PP, Helsby NA, Barratt DT, Pokeya PP, Hombhanje F, et al Pharmacogenomics in Papua New Guineans: unique profiles and implications for enhancing drug efficacy whilst improving drug safety. Pharmacogenet Genomics 2018; 28: 153–164. [DOI] [PubMed] [Google Scholar]
- 2. Griese E‐U, Ilett KF, Kitteringham NR, Eichelbaum M, Powell H, Spargo RM, et al Allele and genotype frequencies of polymorphic cytochromes P4502D6, 2C19 and 2E1 in Aborigines from Western Australia. Pharmacogenetics 2011; 11: 69–76. [DOI] [PubMed] [Google Scholar]
- 3. Helsby NA. CYP2C19 and CYP2D6 genotypes in Pacific peoples. Br J Clin Pharmacol 2016; 82: 1303–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ilett KF, Chiswell GM, Spargo RM, Platt E, Minchin RF. Acetylation phenotype and genotype in Aboriginal leprosy patients from the north‐west region of Western Australia. Pharmacogenetics 1993; 3: 264–269. [DOI] [PubMed] [Google Scholar]
- 5. Campbell MC, Tishkoff SA. African genetic diversity: implications for human demographic history, modern human origins, and complex disease mapping. Annu Rev Genomics Hum Genet 2008; 9: 403–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Dandara C, Suarez‐Kurtz G, Aklillu E. Conference report: First Pharmacogenetics and Precision Medicine Conference in Africa (April 7–9, 2016, Cape Town, South Africa). OMICS 2016; 20: 496–497. [DOI] [PubMed] [Google Scholar]
- 7. Dandara C, Huzair F, Borda‐Rodriguez A, Chirikure S, Okpechi I, Warnich L, et al H3Africa and the African life sciences ecosystem: building sustainable innovation. OMICS 2014; 18: 733–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Dean L. Codeine therapy and CYP2D6 genotype In: Medical Genetics Summaries [online]. eds Pratt V, McLeod H, Rubinstein W, Dean L, Kattman B, Malheiro A. Bethesda, MD, 2012. Available at http://www.ncbi.nlm.nih.gov/books/NBK100662/ (last accessed 4 October 2018). [Google Scholar]
- 9. Kuehn BM. FDA: no codeine after tonsillectomy for children. JAMA 2013; 309: 1100. [DOI] [PubMed] [Google Scholar]
- 10. Aklillu E, Persson I, Bertilsson L, Johansson I, Rodrigues F, Ingelman‐Sundberg M. Frequent distribution of ultrarapid metabolizers of debrisoquine in an Ethiopian population carrying duplicated and multiduplicated functional CYP2D6 alleles. J Pharmacol Exp Ther 1996; 278: 441–446. [PubMed] [Google Scholar]
- 11. Masimirembwa C, Persson I, Bertilsson L, Hasler J, Ingelman‐Sundberg M. A novel mutant variant of the CYP2D6 gene (CYP2D6*17) common in a black African population: association with diminished debrisoquine hydroxylase activity. Br J Clin Pharmacol 1996; 42: 713–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Masimirembwa CM, Hasler JA. Genetic polymorphism of drug metabolising enzymes in African populations: implications for the use of neuroleptics and antidepressants. Brain Res Bull 1997; 44: 561–571. [DOI] [PubMed] [Google Scholar]
- 13. Ngaimisi E, Habtewold A, Minzi O, Makonnen E, Mugusi S, Amogne W, et al Importance of ethnicity, CYP2B6 and ABCB1 genotype for efavirenz pharmacokinetics and treatment outcomes: a parallel‐group prospective cohort study in two sub‐Saharan Africa populations. PLoS One 2013; 8: e67946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Aklillu E, Habtewold A, Ngaimisi E, Yimer G, Mugusi S, Amogne W, et al SLCO1B1 gene variations among Tanzanians, Ethiopians, and Europeans: relevance for African and worldwide precision medicine. OMICS 2016; 20: 538–545. [DOI] [PubMed] [Google Scholar]
- 15. Yimer G, Amogne W, Habtewold A, Makonnen E, Ueda N, Suda A, et al High plasma efavirenz level and CYP2B6*6 are associated with efavirenz‐based HAART‐induced liver injury in the treatment of naive HIV patients from Ethiopia: a prospective cohort study. Pharmacogenomics J 2012; 12: 499–506. [DOI] [PubMed] [Google Scholar]
- 16. Yimer G, Gry M, Amogne W, Makonnen E, Habtewold A, Petros Z, et al Evaluation of patterns of liver toxicity in patients on antiretroviral and anti‐tuberculosis drugs: a prospective four arm observational study in Ethiopian patients. PLoS One 2014; 9: e94271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Mugusi S, Ngaimisi E, Janabi M, Minzi O, Bakari M, Riedel KD, et al Liver enzyme abnormalities and associated risk factors in HIV patients on efavirenz‐based HAART with or without tuberculosis co‐infection in Tanzania. PLoS One 2012; 7: e40180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Mukonzo JK, Okwera A, Nakasujja N, Luzze H, Sebuwufu D, Ogwal‐Okeng J, et al Influence of efavirenz pharmacokinetics and pharmacogenetics on neuropsychological disorders in Ugandan HIV‐positive patients with or without tuberculosis: a prospective cohort study. BMC Infect Dis 2013; 13: 261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Mugusi S, Ngaimisi E, Janabi M, Mugusi F, Minzi O, Aris E, et al Neuropsychiatric manifestations among HIV‐1 infected African patients receiving efavirenz‐based cART with or without tuberculosis treatment containing rifampicin. Eur J Clin Pharmacol 2018; 74: 1405–1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Nyakutira C, Roshammar D, Chigutsa E, Chonzi P, Ashton M, Nhachi C, et al High prevalence of the CYP2B6 516G‐‐>T(*6) variant and effect on the population pharmacokinetics of efavirenz in HIV/AIDS outpatients in Zimbabwe. Eur J Clin Pharmacol 2008; 64: 357–365. [DOI] [PubMed] [Google Scholar]
- 21. Swart M, Skelton M, Ren Y, Smith P, Takuva S, Dandara C. High predictive value of CYP2B6 SNPs for steady‐state plasma efavirenz levels in south African HIV/AIDS patients. Pharmacogenet Genomics 2013; 23: 415–427. [DOI] [PubMed] [Google Scholar]
- 22. Petros Z, Kishikawa J, Makonnen E, Yimer G, Habtewold A, Aklillu E. HLA‐B*57 allele is associated with concomitant anti‐tuberculosis and antiretroviral drugs induced liver toxicity in Ethiopians. Front Pharmacol 2017; 8: 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lopez‐Cortes LF, Ruiz‐Valderas R, Viciana P, Alarcon‐Gonzalez A, Gomez‐Mateos J, Leon‐Jimenez E, et al Pharmacokinetic interactions between efavirenz and rifampicin in HIV‐infected patients with tuberculosis. Clin Pharmacokinet 2002; 41: 681–690. [DOI] [PubMed] [Google Scholar]
- 24. Yenny, Nafrialdi, Djoerban Z, Setiabudy R. Pharmacokinetic interaction between efavirenz and rifampicin in healthy volunteers. Int J Clin Pharmacol Ther 2011; 49: 162–168. [DOI] [PubMed] [Google Scholar]
- 25. Liu J, Chan‐Tack KM, Jadhav P, Seo S, Robertson SM, Kraft J, et al Why did the FDA approve efavirenz 800 mg when co‐administered with rifampin? Int J Clin Pharmacol Ther 2014; 52: 446–453. [DOI] [PubMed] [Google Scholar]
- 26. Thompson MA, Aberg JA, Hoy JF, Telenti A, Benson C, Cahn P, et al Antiretroviral treatment of adult HIV infection: 2012 recommendations of the International Antiviral Society‐USA panel. JAMA 2012; 308: 387–402. [DOI] [PubMed] [Google Scholar]
- 27. Pozniak AL, Coyne KM, Miller RF, Lipman MC, Freedman AR, Ormerod LP, et al British HIV association guidelines for the treatment of TB/HIV coinfection 2011. HIV Med 2011; 12: 517–524. [DOI] [PubMed] [Google Scholar]
- 28. Habtewold A, Makonnen E, Amogne W, Yimer G, Aderaye G, Bertilsson L, et al Is there a need to increase the dose of efavirenz during concomitant rifampicin‐based antituberculosis therapy in sub‐Saharan Africa? The HIV‐TB pharmagene study. Pharmacogenomics 2015; 16: 1047–1064. [DOI] [PubMed] [Google Scholar]
- 29. Gengiah TN, Holford NH, Botha JH, Gray AL, Naidoo K, Abdool Karim SS. The influence of tuberculosis treatment on efavirenz clearance in patients co‐infected with HIV and tuberculosis. Eur J Clin Pharmacol 2012; 68: 689–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kwara A, Ramachandran G, Swaminathan S. Dose adjustment of the non‐nucleoside reverse transcriptase inhibitors during concurrent rifampicin‐containing tuberculosis therapy: one size does not fit all. Expert Opin Drug Metab Toxicol 2010; 6: 55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Cohen K, Grant A, Dandara C, McIlleron H, Pemba L, Fielding K, et al Effect of rifampicin‐based antitubercular therapy and the cytochrome P450 2B6 516G>T polymorphism on efavirenz concentrations in adults in South Africa. Antivir Ther 2009; 14: 687–695. [PMC free article] [PubMed] [Google Scholar]
- 32. World Health Organization. Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach. Second edition [online]. Available at http://apps.who.int/iris/bitstream/10665/208825/1/9789241549684_eng.pdf?ua=1 (last accessed 4 October 2018).
- 33. Mukonzo JK, Bisaso RK, Ogwal‐Okeng J, Gustafsson LL, Owen JS, Aklillu E. CYP2B6 genotype‐based efavirenz dose recommendations during rifampicin‐based antituberculosis cotreatment for a sub‐Saharan Africa population. Pharmacogenomics 2016; 17: 603–613. [DOI] [PubMed] [Google Scholar]
- 34. Mukonzo JK, Owen JS, Ogwal‐Okeng J, Kuteesa RB, Nanzigu S, Sewankambo N, et al Pharmacogenetic‐based efavirenz dose modification: suggestions for an African population and the different CYP2B6 genotypes. PLoS One 2014; 9: e86919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Mutagonda RF, Kamuhabwa AAR, Minzi OMS, Massawe SN, Asghar M, Homann MV, et al Effect of pharmacogenetics on plasma lumefantrine pharmacokinetics and malaria treatment outcome in pregnant women. Malar J 2017; 16: 267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Maganda BA, Minzi OM, Ngaimisi E, Kamuhabwa AA, Aklillu E. CYP2B6*6 genotype and high efavirenz plasma concentration but not nevirapine are associated with low lumefantrine plasma exposure and poor treatment response in HIV‐malaria‐coinfected patients. Pharmacogenomics J 2016; 16: 88–95. [DOI] [PubMed] [Google Scholar]
- 37. Maganda BA, Ngaimisi E, Kamuhabwa AA, Aklillu E, Minzi OM. The influence of nevirapine and efavirenz‐based anti‐retroviral therapy on the pharmacokinetics of lumefantrine and anti‐malarial dose recommendation in HIV‐malaria co‐treatment. Malar J 2015; 14: 179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Habtewold A, Amogne W, Makonnen E, Yimer G, Nylen H, Riedel KD, et al Pharmacogenetic and pharmacokinetic aspects of CYP3A induction by efavirenz in HIV patients. Pharmacogenomics J 2013; 13: 484–489. [DOI] [PubMed] [Google Scholar]
- 39. Petros Z, Lee MM, Takahashi A, Zhang Y, Yimer G, Habtewold A, et al Genome‐wide association and replication study of anti‐tuberculosis drugs‐induced liver toxicity. BMC Genomics 2016; 17: 755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Carr DF, Chaponda M, Jorgensen AL, Castro EC, van Oosterhout JJ, Khoo SH, et al Association of human leukocyte antigen alleles and nevirapine hypersensitivity in a Malawian HIV‐infected population. Clin Infect Dis 2013; 56: 1330–1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Peprah E, Xu H, Tekola‐Ayele F, Royal CD. Genome‐wide association studies in Africans and African Americans: expanding the framework of the genomics of human traits and disease. Public Health Genomics 2015; 18: 40–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Adebamowo SN, Francis V, Tambo E, Diallo SH, Landoure G, Nembaware V, et al Implementation of genomics research in Africa: challenges and recommendations. Glob Health Action 2018; 11: 1419033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Petros Z, Makonnen E, Aklillu E. Genome‐wide association studies for idiosyncratic drug‐induced hepatotoxicity: looking Back‐looking forward to next‐generation innovation. OMICS 2017; 21: 123–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Salzano FM, Bortolini MC. The Evolution and Genetics of Latin American Populations. Cambridge, UK: Cambridge University Press, 2002. [Google Scholar]
- 45. Pena SD, Di Pietro G, Fuchshuber‐Moraes M, Genro JP, Hutz MH, Kehdy Fde S, et al The genomic ancestry of individuals from different geographical regions of Brazil is more uniform than expected. PLoS One 2011; 6: e17063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Silva‐Zolezzi I, Hidalgo‐Miranda A, Estrada‐Gil J, Fernandez‐Lopez JC, Uribe‐Figueroa L, Contreras A, et al Analysis of genomic diversity in Mexican mestizo populations to develop genomic medicine in Mexico. Proc Natl Acad Sci USA 2009; 106: 8611–8616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Galanter JM, Fernandez‐Lopez JC, Gignoux CR, Barnholtz‐Sloan J, Fernandez‐Rozadilla C, Via M, et al Development of a panel of genome‐wide ancestry informative markers to study admixture throughout the Americas. PLoS Genet 2012; 8: e1002554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Bonifaz‐Peña V, Contreras AV, Struchiner CJ, Roela RA, Furuya‐Mazzotti TK, Chammas R, et al Exploring the distribution of genetic markers of pharmacogenomics relevance in Brazilian and Mexican populations. PLoS One 2014; 9: e112640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Santoro A, Felipe CR, Tedesco‐Silva H, Medina‐Pestana JO, Struchiner CJ, Ojopi EB, et al Pharmacogenetics of calcineurin inhibitors in Brazilian renal transplant patients. Pharmacogenomics 2011; 12: 1293–1303. [DOI] [PubMed] [Google Scholar]
- 50. Birdwell KA, Decker B, Barbarino JM, Peterson JF, Stein CM, Sadee W, et al Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for CYP3A5 genotype and tacrolimus dosing. Clin Pharmacol Ther 2015; 9: 19–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Johnson JA, Gong L, Whirl‐Carrillo M, Gage BF, Scott SA, Stein CM, et al Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther 2011; 90: 625–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Perini JA, Struchiner CJ, Silva‐Assunção E, Santana ISC, Rangel F, Ojopi EB, et al Pharmacogenetics of warfarin: development of a dosing algorithm for Brazilian patients. Clin Pharmacol Ther 2008; 84: 722–728. [DOI] [PubMed] [Google Scholar]
- 53. Suarez‐Kurtz G, Perini JA, Silva‐Assuncao E, Struchiner CJ. Relative contribution of VKORC1, CYP2C9, and INR response to warfarin stable dose. Blood 2009; 113: 4125–4126. [DOI] [PubMed] [Google Scholar]
- 54. Gage BF, Eby C, Johnson JA, Deych E, Rieder MJ, Ridker PM, et al Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfarin. Clin Pharmacol Ther 2008; 84: 326–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Limdi NA, Beasley TM, Crowley MR, Goldstein JA, Rieder MJ, Flockhart DA, et al VKORC1 polymorphisms, haplotypes and haplotype groups on warfarin dose among African‐Americans and European‐Americans. Pharmacogenomics 2008; 9: 1445–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Suarez‐Kurtz G, Pena SDJ, Struchiner CJ, Hutz MH. Pharmacogenomic diversity among Brazilians: influence of ancestry, self‐reported color, and geographical origin. Front Pharmacol 2012; 3: 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Suarez‐Kurtz G, Paula DP, Struchiner CJ. Pharmacogenomic implications of population admixture: Brazil as a model case. Pharmacogenomics 2014; 15: 209–219. [DOI] [PubMed] [Google Scholar]
- 58. Johnson J, Caudle K, Gong L, Whirl‐Carrillo M, Stein C, Scott S, et al Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for pharmacogenetics‐guided warfarin dosing: 2017 update. Clin Pharmacol Ther 2017; 102: 397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Yang JJ, Landier W, Yang W, Liu C, Hageman L, Cheng C, et al Inherited NUDT15 variant is a genetic determinant of mercaptopurine intolerance in children with acute lymphoblastic leukemia. J Clin Oncol 2015; 33: 1235–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Moriyama T, Nishii R, Perez‐Andreu V, Yang W, Klussmann FA, Zhao X, et al NUDT15 polymorphisms alter thiopurine metabolism and hematopoietic toxicity. Nat Genet 2016; 48: 367–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Brasil LW, Rodrigues‐Soares F, Santoro AB, Almeida ACG, Kühn A, Ramasawmy R, et al CYP2D6 activity and the risk of recurrence of Plasmodium vivax malaria in the Brazilian Amazon: a prospective cohort study. Malar J 2018. Feb 1; 17: 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Ang HX, Chan SL, Sani LL, Quah CB, Brunham LR, Tan BOP, et al Pharmacogenomics in Asia: a systematic review on current trends and novel discoveries. Pharmacogenomics 2017; 18: 891–910. [DOI] [PubMed] [Google Scholar]
- 63. Chung WH, Wang CW, Dao RL. Severe cutaneous adverse drug reactions. J Dermatol 2016; 43: 758–766. [DOI] [PubMed] [Google Scholar]
- 64. Wang YH, Chen CB, Tassaneeyakul W, Saito Y, Aihara M, Choon SE, et al The medication risk of Stevens‐Johnson syndrome and toxic epidermal necrolysis in Asians: the major drug causality and comparison to the USA FDA label. Clin Pharmacol Ther 2019; 105: 112–120. [DOI] [PubMed] [Google Scholar]
- 65. Kurose K, Sugiyama E, Saito Y. Population differences in major functional polymorphisms of pharmacokinetics/pharmacodynamics‐related genes in Eastern Asians and Europeans: implications in the clinical trials for novel drug development. Drug Metab Pharmacokinet 2012; 27: 9–54. [DOI] [PubMed] [Google Scholar]
- 66. Hung SI, Chung WH, Liou LB, Chu CC, Lin M, Huang HP, et al HLA‐B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc Natl Acad Sci U S A 2005; 102: 4134–4139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Ko TM, Tsai CY, Chen SY, Chen KS, Yu KH, Chu CS, et al Use of HLA‐B*58:01 genotyping to prevent allopurinol induced severe cutaneous adverse reactions in Taiwan: national prospective cohort study. BMJ 2015; 351: h4848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Allele frequency net [online]. Available at http://www.allelefrequencies.net/default.asp (last accessed 6 September 2018).
- 69. Hung SI, Chung WH, Jee SH, Chen WC, Chang YT, Lee WR, et al Genetic susceptibility to carbamazepine‐induced cutaneous adverse drug reactions. Pharmacogenet Genomics 2006; 16: 297–306. [DOI] [PubMed] [Google Scholar]
- 70. Chen P, Lin JJ, Lu CS, Ong CT, Hsieh PF, Yang CC, et al Carbamazepine‐induced toxic effects and HLA‐B*1502 screening in Taiwan. N Engl J Med 2011; 364: 1126–1133. [DOI] [PubMed] [Google Scholar]
- 71. Mushiroda T, Takahashi Y, Onuma T, Yamamoto Y, Kamei T, Hoshida T, et al Association of HLA‐A*31:01 screening with the incidence of carbamazepine‐induced cutaneous adverse reactions in a Japanese population. JAMA Neurol 2018; 75: 842–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Imatoh T, Sai K, Saito Y. Pharmacogenomic information in the warning section of drug labels: a comparison between labels in the United States and those in five other countries/regions. J Clin Pharm Ther 2018; 43: 493–499. [DOI] [PubMed] [Google Scholar]
- 73. Chung WH, Chang WC, Lee YS, Wu YY, Yang CH, Ho HC, et al Genetic variants associated with phenytoin‐related severe cutaneous adverse reactions. JAMA 2014; 312: 525–534. [DOI] [PubMed] [Google Scholar]
- 74. Tassaneeyakul W, Prabmeechai N, Sukasem C, Kongpan T, Konyoung P, Chumworathayi P, et al Associations between HLA class I and cytochrome P450 2C9 genetic polymorphisms and phenytoin‐related severe cutaneous adverse reactions in a Thai population. Pharmacogenet Genomics 2016; 26: 225–234. [DOI] [PubMed] [Google Scholar]
- 75. Zhang FR, Liu H, Irwanto A, Fu XA, Li Y, Yu GQ, et al HLA‐B*13:01 and the dapsone hypersensitivity syndrome. N Engl J Med 2013; 369: 1620–1628. [DOI] [PubMed] [Google Scholar]
- 76. Tempark T, Satapornpong P, Rerknimitr P, Nakkam N, Saksit N, Wattanakrai P, et al Dapsone‐induced severe cutaneous adverse drug reactions are strongly linked with HLA‐B*13: 01 allele in the Thai population. Pharmacogenet Genomics 2017; 27: 429–437. [DOI] [PubMed] [Google Scholar]
- 77. Minami H, Sai K, Saeki M, Saito Y, Ozawa S, Suzuki K, et al Irinotecan pharmacokinetics/pharmacodynamics and UGT1A genetic polymorphisms in Japanese: roles of UGT1A1*6 and *28. Pharmacogenet Genomics 2007; 17: 497–504. [DOI] [PubMed] [Google Scholar]
- 78. Han FF, Guo CL, Yu D, Zhu J, Gong LL, Li GR, et al Associations between UGT1A1*6 or UGT1A1*6/*28 polymorphisms and irinotecan‐induced neutropenia in Asian cancer patients. Cancer Chemother Pharmacol 2014; 73: 779–788. [DOI] [PubMed] [Google Scholar]
- 79. ICH E5 (R1) Guideline: Ethnic factors in the acceptability of foreign clinical data [online]. Available at https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E5_R1/Step4/E5_R1__Guideline.pdf (last accessed 6 October 2018).
- 80. ICH E17 Guideline: General principles for planning and design of multi‐regional clinical trials [online]. Available at https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E17/E17EWG_Step4_2017_1116.pdf (last accessed 6 October 2018).
- 81. ICH E18 Guideline: Guideline on genomic sampling and management of genomic data [online]. Available at https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E18/E18EWG_Step4_Guideline_2017_0803.pdf (last accessed 6 October 2018).
- 82. Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S, et al The IUPHAR/BPS guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acid Res 2018: D1091–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E, et al The Concise Guide to PHARMACOLOGY 2017/18: Enzymes. Br J Pharmacol 2017; 174: S272–S359. [DOI] [PMC free article] [PubMed] [Google Scholar]