We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short Communication

UGT1A1 regulatory variant with potential effect on efficacy of HIV and cancer drugs commonly prescribed in South Africa

    Jenny Mary Mathew

    Division of Human Genetics, National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa

    Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa

    ,
    Phelelani Thokozani Mpangase

    Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa

    ,
    Dhriti Sengupta

    Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa

    ,
    Stanford Kwenda

    Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, 2192, South Africa

    ,
    Demetra Mavri-Damelin

    School of Molecular & Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, 2050, South Africa

    &
    Michèle Ramsay

    *Author for correspondence:

    E-mail Address: Michele.Ramsay@wits.ac.za

    Division of Human Genetics, National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa

    Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa

    Published Online:https://doi.org/10.2217/pgs-2021-0062

    Aim: Despite the high disease burden of human immunodeficiency virus (HIV) infection and colorectal cancer (CRC) in South Africa (SA), treatment-relevant pharmacogenetic variants are understudied. Materials & methods: Using publicly available genotype and gene expression data, a bioinformatic pipeline was developed to identify liver expression quantitative trait loci (eQTLs). Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Conclusion: As increased UGT1A1 expression could affect the clinical outcome of dolutegravir and irinotecan treatment by increasing drug clearance, patients with the rs28967009A variant may require increased drug doses to reach therapeutic levels or should be prescribed alternative drugs.

    References

    • 1. Nica AC, Dermitzakis ET. Expression quantitative trait loci: present and future. Philos. Trans. RSoc. B Biol. Sci. 368(1620), (2013).
    • 2. Shabalin AA. Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics 28(10), 1353–1358 (2012).
    • 3. Hovelson DH, Xue Z, Zawistowski M et al. Characterization of ADME gene variation in 21 populations by exome sequencing. Pharmacogenet. Genomics 27(3), 89–100 (2017).
    • 4. Ramos E, Doumatey A, Elkahloun AG et al. Pharmacogenomics, ancestry and clinical decision making for global populations. Pharmacogenomics J. 14(3), 217–222 (2014).
    • 5. Sengupta D, Choudhury A, Fortes-Lima C et al. Genetic-substructure and complex demographic history of South African Bantu speakers. Nat. Comm. 12, 2080 (2021).
    • 6. Lee H-K, Hu M, Lui SS, Ho C-S, Wong C-K, Tomlinson B. Effects of polymorphisms in ABCG2, SLCO1B1, SLC10A1 and CYP2C9/19 on plasma concentrations of rosuvastatin and lipid response in Chinese patients. Pharmacogenomics 14(11), 1283–1294 (2013).
    • 7. Birmingham BK, Bujac SR, Elsby R et al. Rosuvastatin pharmacokinetics and pharmacogenetics in Caucasian and Asian subjects residing in the United States. Eur. J. Clin. Pharmacol. 71(3), 329–340 (2015).
    • 8. Iskakova AN, Romanova AA, Aitkulova AM, Sikhayeva NS, Zholdybayeva EV, Ramanculov EM. Polymorphisms in genes involved in the absorption, distribution, metabolism, and excretion of drugs in the Kazakhs of Kazakhstan. BMC Genet. 17(1), (2016).
    • 9. Hrdlickova B, de Almeida RC, Borek Z, Withoff S. Genetic variation in the non-coding genome: involvement of micro-RNAs and long non-coding RNAs in disease. Biochim. Biophys. Acta – Mol. Basis Dis. 1842(10), 1910–1922 (2014).
    • 10. Gona PN, Gona CM, Ballout S et al. Burden and changes in HIV/AIDS morbidity and mortality in Southern Africa Development Community Countries, 1990–2017. BMC Public Health 20(1), 867 (2020).
    • 11. Satoh S, Boyer E. HIV in South Africa. Lancet 394(10197), 467 (2019).
    • 12. Brand M, Gaylard P, Ramos J. Colorectal cancer in South Africa: an assessment of disease presentation, treatment pathways and 5-year survival. South African Med. J. 108(2), 118 (2018).
    • 13. Herbst C, Miot JK, Moch SL, Ruff P. Colorectal Cancer (CRC) treatment and associated costs in the public sector compared to the private sector in Johannesburg, South Africa. BMC Health Serv. Res. 20(1), 290 (2020).
    • 14. Schröder A, Klein K, Winter S et al. Genomics of ADME gene expression: mapping expression quantitative trait loci relevant for absorption, distribution, metabolism and excretion of drugs in human liver. Pharmacogenomics J. 13(1), 12–20 (2013).
    • 15. Schulz C, Boeck S, Heinemann V, Stemmler H-J. UGT1A1 genotyping: a predictor of irinotecan-associated side effects and drug efficacy? Anticancer Drugs 20(10), 867–879 (2009).
    • 16. Barbarino JM, Haidar CE, Klein TE, Altman RB. PharmGKB summary: very important pharmacogene information for UGT1A1. Pharmacogenet. Genomics 24(3), 177–183 (2014).
    • 17. Ramesh M, Ahlawat P, Srinivas NR. Irinotecan and its active metabolite, SN-38: review of bioanalytical methods and recent update from clinical pharmacology perspectives. Biomed. Chromatogr. 24(1), 104–123 (2010).
    • 18. Mathijssen RHJ, van Alphen RJ, Verweij J et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin. Cancer Res. 7(8), 2182 LP–2194 (2001).
    • 19. Dias MM, McKinnon RA, Sorich MJ. Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Pharmacogenomics 13(8), 889–899 (2012).
    • 20. Lankisch TO, Schulz C, Zwingers T et al. Gilbert's syndrome and irinotecan toxicity: combination with UDP-glucuronosyltransferase 1A7 variants increases risk. Cancer Epidemiol. Biomarkers Prev. 17(3), 695 LP–701 (2008).
    • 21. Tam VC, Rask S, Koru-Sengul T, Dhesy-Thind S. Generalizability of toxicity data from oncology clinical trials to clinical practice: toxicity of irinotecan-based regimens in patients with metastatic colorectal cancer. Curr. Oncol. 16(6), 13–20 (2009).
    • 22. Obradovic M, Mrhar A, Kos M. Cost–effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Pharmacogenomics 9(5), 539–549 (2008).
    • 23. Liu X, Cheng D, Kuang Q, Liu G, Xu W. Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Pharmacogenomics J. 14(2), 120–129 (2014.)
    • 24. Castellino S, Moss L, Wagner D et al. Metabolism, excretion, and mass balance of the HIV-1 integrase inhibitor dolutegravir in humans. Antimicrob. Agents Chemother. 57(8), 3536–3546 (2013).
    • 25. Yagura H, Watanabe D, Kushida H et al. Impact of UGT1A1 gene polymorphisms on plasma dolutegravir trough concentrations and neuropsychiatric adverse events in Japanese individuals infected with HIV-1. BMC Infect. Dis. 17(1), 622 (2017).
    • 26. Walmsley SL, Antela A, Clumeck N et al. Dolutegravir plus abacavir–lamivudine for the treatment of HIV-1 infection. N. Engl. J. Med. 369(19), 1807–1818 (2013).
    • 27. Moorhouse MA, Carmona S, Davies N et al. Southern African HIV Clinicians Society Guidance on the use of dolutegravir in first-line antiretroviral therapy. South African J. HIV Med. 19(1), (2018).
    • 28. Venter WDF, Moorhouse M, Sokhela S et al. Dolutegravir plus two different prodrugs of tenofovir to treat HIV. N. Engl. J. Med. 381(9), 803–815 (2019).
    • 29. Spano J-P, Poizot-Martin I, Costagliola D et al. Non-AIDS-related malignancies: expert consensus review and practical applications from the multidisciplinary CANCERVIH Working Group. Ann. Oncol. 27(3), 397–408 (2016).
    • 30. Elzi L, Erb S, Furrer H et al. Adverse events of raltegravir and dolutegravir. AIDS 31(13), 1853–1858 (2017).
    • 31. Lepik KJ, Yip B, Ulloa AC et al. Adverse drug reactions to integrase strand transfer inhibitors. AIDS 32(7), (2018).
    • 32. Cuzin L, Pugliese P, Katlama C et al. Integrase strand transfer inhibitors and neuropsychiatric adverse events in a large prospective cohort. J. Antimicrob. Chemother. 74(3), 754–760 (2019).
    • 33. Llibre JM, Montoliu A, Miró JM et al. Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort. HIV Med. 20(3), 237–247 (2019).
    • 34. Gammal RS, Court MH, Haidar CE et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and atazanavir prescribing. Clin. Pharmacol. Ther. 99(4), 363–369 (2016).
    • 35. Teh LK, Hashim H, Zakaria ZA, Salleh MZ. Polymorphisms of UGT1A1*6, UGT1A1*27 & UGT1A1*28 in three major ethnic groups from Malaysia. Indian J. Med. Res. 136(2), 249–259 (2012).
    • 36. Dai X, Wu C, He Y et al. A genome-wide association study for serum bilirubin levels and gene-environment interaction in a Chinese Population. Genet. Epidemiol. 37(3), 293–300 (2013).
    • 37. Liu JY, Qu K, Sferruzza AD, Bender RA. Distribution of the UGT1A1*28 polymorphism in Caucasian and Asian populations in the US: a genomic analysis of 138 healthy individuals. Anticancer Drugs 18(6), 693–696 (2007).
    • 38. Palomaki GE, Bradley LA, Douglas MP, Kolor K, Dotson WD. Can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? An evidence-based review. Genet. Med. 11(1), 21–34 (2009).
    • 39. Dean L. Irinotecan therapy and UGT1A1 genotype drug. In: Medical Genetics Summaries [Internet]. Pratt VM, Scott SA, Pirmohamed M et al. (Eds). National Center for Biotechnology Information, Bethseda, MA, USA (2018).
    • 40. Fukuda M, Suetsugu T, Shimada M et al. Prospective study of the UGT1A1*27 gene polymorphism during irinotecan therapy in patients with lung cancer: results of Lung Oncology Group in Kyusyu (LOGIK1004B). Thorac. Cancer 7(4), 467–472 (2016).
    • 41. Sukasem C, Atasilp C, Chansriwong P, Chamnanphon M, Puangpetch A, Sirachainan E. Development of pyrosequencing method for detection of UGT1A1 polymorphisms in Thai colorectal cancers. J. Clin. Lab. Anal. 30(1), 84–89 (2016).
    • 42. Marques SC, Ikediobi ON. The clinical application of UGT1A1 pharmacogenetic testing: gene–environment interactions. Hum. Genomics 4(4), 238–249 (2010).
    • 43. Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc. Natl Acad. Sci. USA 95(14), 8170–8174 (1998).
    • 44. Achenbach CJ, Darin KM, Murphy RL, Katlama C. Atazanavir/ritonavir-based combination antiretroviral therapy for treatment of HIV-1 infection in adults. Future Virol. 6(2), 157–177 (2011).
    • 45. Turatti L, Sprinz E, Lazzaretti RK et al. Short Communication: UGT1A1*28 variant allele is a predictor of severe hyperbilirubinemia in HIV-infected patients on HAART in Southern Brazil. AIDS Res. Hum. Retroviruses 28(9), 1015–1018 (2011).
    • 46. Johnson DH, Venuto C, Ritchie MD et al. Genomewide association study of atazanavir pharmacokinetics and hyperbilirubinemia in AIDS Clinical Trials Group protocol A5202. Pharmacogenet. Genomics 24(4), 195–203 (2014).
    • 47. Nishijima T, Tsuchiya K, Tanaka N et al. Single-nucleotide polymorphisms in the UDP-glucuronosyltransferase 1A-3′ untranslated region are associated with atazanavir-induced nephrolithiasis in patients with HIV-1 infection: a pharmacogenetic study. J. Antimicrob. Chemother. 69(12), 3320–3328 (2014).
    • 48. Vardhanabhuti S, Ribaudo HJ, Landovitz RJ et al. Screening for UGT1A1 genotype in study A5257 would have markedly reduced premature discontinuation of atazanavir for hyperbilirubinemia. Open forum Infect. Dis. 2(3), ofv085–ofv085 (2015).
    • 49. Turner RM, Fontana V, Zhang JE et al. A genome-wide association study of circulating levels of atorvastatin and its major metabolites. Clin. Pharmacol. Ther. 108(2), 287–297 (2020).
    • 50. Han J-Y, Lim H-S, Shin ES et al. Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. J. Clin. Oncol. 24(15), 2237–2244 (2006).
    • 51. Jada SR, Lim R, Wong CI et al. Role of UGT1A1*6, UGT1A1*28 and ABCG2 c.421C>A polymorphisms in irinotecan-induced neutropenia in Asian cancer patients. Cancer Sci. 98(9), 1461–1467 (2007).
    • 52. Takano M, Kato M, Yoshikawa T et al. Clinical significance of UDP-Glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers. Oncology 76(5), 315–321 (2009).
    • 53. Danoff TM, Campbell DA, McCarthy LC et al. A Gilbert's syndrome UGT1A1 variant confers susceptibility to tranilast-induced hyperbilirubinemia. Pharmacogenomics J. 4(1), 49–53 (2004).
    • 54. Trontelj J, Marc J, Zavratnik A, Bogataj M, Mrhar A. Effects of UGT1A1*28 polymorphism on raloxifene pharmacokinetics and pharmacodynamics. Br. J. Clin. Pharmacol. 67(4), 437–444 (2009).
    • 55. Kishi S, Yang W, Boureau B et al. Effects of prednisone and genetic polymorphisms on etoposide disposition in children with acute lymphoblastic leukemia. Blood 103(1), 67–72 (2004).