Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Population impact of pharmacogenetic tests in admixed populations across the Americas

Abstract

The current population of the Americas (~1 billion people) is highly heterogeneous as a result of five centuries of admixture between three major parental populations, namely Native Amerindians, Europeans, and subSaharan Africans. This heterogeneity has important pharmacogenetic (PGx) implications. The present study explores this issue with respect to the population impact of PGx tests listed in the CPIC and DWPG guidelines for the cancer chemotherapeutic agents fluoropyrimidines, irinotecan, and thiopurines. Population impact was assessed by the number of individuals needed to genotype (NNG) and to treat [NNT] in order to prevent one additional adverse effect. The 1000 Genomes Project database was accessed to obtain genotypes for the relevant PGx markers and to estimate individual proportions of Native, European, and African ancestry for six recently admixed populations across the Americas. The ancestry proportions were then employed to devise three sub-cohorts, denoted NAT, EUR, and AFR, in which one of the three major ancestral roots predominates largely (8 to 30-fold) over the other two. Minor allele frequency (MAF) of NUDT15 rs116855232, TPMT rs1800460, and UGT1A1 rs887829 differed significantly across the three sub-cohorts, whereas no difference was observed for TPMT rs1142345 and 1800462, and the four DPYD variants interrogated. The frequency of combined TPMT/NUDT and UGT1A1, but not DPD, metabolic phenotypes differed significantly among the sub-cohorts. The NNGs for the drug/sub-cohorts pairs, ranged from 12 (irinotecan/UGT1A1 in EUR) to 360 (fluoropyrimidines/DPYD in NAT). Differences in MAF of the interrogated variants and consequently in the distribution of metabolic phenotypes, plus the variable individual proportions of biogeographical ancestry concur to the 30-fold range of NNGs for the PGx tests, across the sub-cohorts. This large variation is likely to influence the perceived benefits and the clinical adoption of PGx screening for the chemotherapeutic agents investigated. This is of especial concern for fluoropyrimidines, in view of the large NNGs observed in the study sub-cohorts (NNG = 176–360) and confirmed in cancer patients from Brazil (NNG = 312).

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Distribution of metabolizer phenotypes and activity scores (AS) across the EUR, NAT, and AFR sub-cohorts.
Fig. 2: Population impact of PGx tests.

Similar content being viewed by others

References

  1. Cavalli-Sforza LL, Menozzi PA. The history and geography of human genes. Chichester: Princeton University Press; 1994.

  2. Suarez-Kurtz G, Pena SD, Struchiner CJ, Hutz MH. Pharmacogenomic diversity among Brazilians: influence of ancestry, self-reported color, and geographical origin. Front Pharmacol. 2012;3:191.

    Article  Google Scholar 

  3. Bonifaz-Peña V, Contreras AV, Struchiner CJ, Roela RA, Furuya-Mazzotti T, Chammas R, et al. Exploring the distribution of genetic markers of pharmacogenomics relevance in Brazilian and Mexican populations. PLoS ONE. 2014;9:e112640.

    Article  Google Scholar 

  4. Suarez-Kurtz G, Parra EJ. Population diversity in pharmacogenetics: a Latin American perspective. Adv Pharmacol. 2018;83:133–54.

    Article  CAS  Google Scholar 

  5. 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;48:367–73.

    Article  CAS  Google Scholar 

  6. Yang JJ, Whirl-Carrillo M, Scott SA, Turner AJ, Schwab M, Tanaka Y, et al. Pharmacogene variation consortium gene introduction: NUDT15. Clin Pharmacol Ther. 2019;105:1091–4.

    Article  Google Scholar 

  7. Suarez-Kurtz G, Brisson GD, Hutz MH, Petzl-Erler ML, Salzano FM. NUDT15 Polymorphism in Native American Populations of Brazil. Clin Pharmacol Ther. 2019;105:1321–2.

    Article  Google Scholar 

  8. Suarez-Kurtz G, Araujo GS, de Souza SJ. Pharmacogenomic implications of population diversity in Latin America: TPMT and NUDT15 polymorphisms and thiopurine dosing. Pharmacogenet Genom. 2020;30:1–4.

    Article  CAS  Google Scholar 

  9. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens J, Swen JJ, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing: 2017 update. Clin Pharmacol Ther. 2018;103:210–6.

    Article  CAS  Google Scholar 

  10. Relling M, Schwab M, Whirl-Carrillo M, Suarez-Kurtz G, Pui CH, Stein CM, et al. Clinical Pharmacogenetics Implementation Consortium guideline for thiopurine dosing based on TPMT and NUDT15 genotypes: 2018 update. Clin Pharmacol Ther. 2019;105:1095–105.

    Article  CAS  Google Scholar 

  11. Royal Dutch Pharmacists Association (KNMP). Dutch Pharmacogenetics Working Group (DPWG). Pharmacogenetic Guidelines. Irinotecan – UGT1A1. Netherlands: Royal Dutch Pharmacists Association (KNMP). https://www.g-standaard.nl/risicoanalyse/B0001694.PDF.

  12. Tonk ECM, Gurwitz D, Maitland-van der Zee AH, Janssens ACJW. Assessment of pharmacogenetic tests: presenting measures of clinical validity and potential population impact in association studies. Pharmacogenomics J. 2017;17:386–92.

    Article  CAS  Google Scholar 

  13. Debortoli G, Araujo GS, Fortes-Lima C, Parra EJ, Suarez-Kurtz G. Identification of ancestry proportions in admixed groups across the Americas using clincial pharmacogenetic panels. Manuscript in preparation.

  14. Salzano FM, Bortolini MC The Evolution and genetics of Latin American populations. Cambridge: Cambridge University Press; 2002.

  15. Ruiz-Linares A, Adhikari K, Acuña-Alonzo V, Quinto-Sanchez M, Jaramillo C, Arias W, et al. Admixture in Latin America: geographic structure, phenotypic diversity and self-perception of ancestry based on 7,342 individuals. PLoS Genet. 2014;10:e1004572.

    Article  Google Scholar 

  16. Gammal RS, Court MH, Haidar CE, Iwuchukwu OF, Gaur AH, Alvarellos M, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for UGT1A1 and Atazanavir Prescribing. Clin Pharmacol Ther. 2016;99:363–9.

    Article  CAS  Google Scholar 

  17. Ferreira GMA, Elias ABR, Nascimento J, Monteiro WM, Melo GC, Baia-da-Silva DC, et al. Pharmacogenomics of thiopurines: distribution of TPMT and NUDT15 polymorphisms in the Brazilian Amazon. Pharmacogenet Genom. 2020;30:184–9.

    Article  CAS  Google Scholar 

  18. Hoskins JM, Goldberg RM, Qu P, Ibrahim JG, McLeod HL. UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst. 2007;99:1290–5.

    Article  CAS  Google Scholar 

  19. 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;33:1235–42.

    Article  CAS  Google Scholar 

  20. Fratte CD, Polesel J, Roncato R, De Mattia E, Ecca F, Bignucolo A, et al. DPYD gene activity score predicts dose-limiting toxicity in fluoropyrimidine-treated colorectal cancer patients. J Mol Clin Med. 2018;1:143–50. https://doi.org/10.1038/tpj.2016.34

    Article  CAS  Google Scholar 

  21. Suarez-Kurtz G, Kovaleski G, Elias AB, Motta VL, Wolch K, Emerenciano M, et al. Implementation of a pharmacogenomic program in a Brazilian public institution. Pharmacogenomics 2020;21:549–57.

    Article  CAS  Google Scholar 

  22. Veenstra DL. The value of routine pharmacogenomic screening- are we there yet? A perspective on the costs and benefits of routine screening- shouldn’t everyone have this done? Clin Pharmacol Ther. 2016;99:164–6.

    Article  CAS  Google Scholar 

  23. Hughes DA. Economics of pharmacogenetic-guided treatments: Underwhelming or overstated? Clin Pharmacol Ther. 2018;103:749–51.

    Article  Google Scholar 

Download references

Acknowledgements

GS-K is supported by the Brazilian agencies Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Guilherme Suarez-Kurtz.

Ethics declarations

Conflict of interest

The author declares that he has no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Suarez-Kurtz, G. Population impact of pharmacogenetic tests in admixed populations across the Americas. Pharmacogenomics J 21, 216–221 (2021). https://doi.org/10.1038/s41397-020-00200-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41397-020-00200-w

Search

Quick links