1932

Abstract

A long-standing recognition that information from human genetics studies has the potential to accelerate drug discovery has led to decades of research on how to leverage genetic and phenotypic information for drug discovery. Established simple and advanced statistical methods that allow the simultaneous analysis of genotype and clinical phenotype data by genome- and phenome-wide analyses, colocalization analyses with quantitative trait loci data from transcriptomics and proteomics data sets from different tissues, and Mendelian randomization are essential tools for drug development in the postgenomic era. Numerous studies have demonstrated how genomic data provide opportunities for the identification of new drug targets, the repurposing of drugs, and drug safety analyses. With an increase in the number of biobanks that enable linking in-depth omics data with rich repositories of phenotypic traits via electronic health records, more powerful ways for the evaluation and validation of drug targets will continue to expand across different disciplines of clinical research.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-051421-111324
2023-01-20
2024-05-10
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/63/1/annurev-pharmtox-051421-111324.html?itemId=/content/journals/10.1146/annurev-pharmtox-051421-111324&mimeType=html&fmt=ahah

Literature Cited

  1. 1.
    Dowden H, Munro J. 2019. Trends in clinical success rates and therapeutic focus. Nat. Rev. Drug Discov. 18:7495–96
    [Google Scholar]
  2. 2.
    Wouters OJ, McKee M, Luyten J. 2020. Estimated research and development investment needed to bring a new medicine to market, 2009–2018. JAMA 323:9844–53
    [Google Scholar]
  3. 3.
    Nelson MR, Tipney H, Painter JL, Shen J, Nicoletti P et al. 2015. The support of human genetic evidence for approved drug indications. Nat. Genet. 47:8856–60
    [Google Scholar]
  4. 4.
    King EA, Davis JW, Degner JF. 2019. Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval. PLOS Genet. 15:12e1008489
    [Google Scholar]
  5. 5.
    Jensen PB, Jensen LJ, Brunak S. 2012. Mining electronic health records: towards better research applications and clinical care. Nat. Rev. Genet. 13:395–405
    [Google Scholar]
  6. 6.
    Zhou W, Kanai M, Wu K-HH, Humaira R, Tsuo K et al. 2021. Global Biobank Meta-analysis Initiative: powering genetic discovery across human diseases. medRxiv 2021.11.19.21266436. https://doi.org/10.1101/2021.11.19.21266436
    [Crossref]
  7. 7.
    Sanseau P, Agarwal P, Barnes MR, Pastinen T, Richards JB et al. 2012. Use of genome-wide association studies for drug repositioning. Nat. Biotechnol. 30:4317–20
    [Google Scholar]
  8. 8.
    Namba S, Konuma T, Wu K-H, Zhou W, Biobank G et al. 2021. A practical guideline of genomics-driven drug discovery in the era of global biobank meta-analysis. medRxiv 2021.12.03.21267280. https://doi.org/10.1101/2021.12.03.21267280
    [Crossref] [Google Scholar]
  9. 9.
    Yarmolinsky J, Díez-Obrero V, Richardson TG, Pigeyre M, Sjaarda J et al. 2022. Genetically proxied therapeutic inhibition of antihypertensive drug targets and risk of common cancers: a mendelian randomization analysis. PLOS Med. 19:2e1003897
    [Google Scholar]
  10. 10.
    Swerdlow DI, Holmes MV, Kuchenbaecker KB, Engmann JEL, Shah T et al. 2012. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet 379:98221214–24
    [Google Scholar]
  11. 11.
    Storm CS, Kia DA, Almramhi MM, Bandres-Ciga S, Finan C et al. 2021. Finding genetically-supported drug targets for Parkinson's disease using Mendelian randomization of the druggable genome. Nat. Commun. 12:7342
    [Google Scholar]
  12. 12.
    Suhre K, McCarthy MI, Schwenk JM. 2021. Genetics meets proteomics: perspectives for large population-based studies. Nat. Rev. Genet. 22:119–37
    [Google Scholar]
  13. 13.
    Folkersen L, Gustafsson S, Wang Q, Hansen DH, Hedman ÅK et al. 2020. Genomic and drug target evaluation of 90 cardiovascular proteins in 30,931 individuals. Nat. Metab. 2:101135–48
    [Google Scholar]
  14. 14.
    Ferkingstad E, Sulem P, Atlason BA, Sveinbjornsson G, Magnusson MI et al. 2021. Large-scale integration of the plasma proteome with genetics and disease. Nat. Genet. 53:121712–21
    [Google Scholar]
  15. 15.
    Richardson TG, Leyden GM, Wang Q, Bell JA, Elsworth B et al. 2022. Characterising metabolomic signatures of lipid-modifying therapies through drug target mendelian randomisation. PLOS Biol. 20:2e3001547
    [Google Scholar]
  16. 16.
    Nielsen JB, Rom O, Surakka I, Graham SE, Zhou W et al. 2020. Loss-of-function genomic variants highlight potential therapeutic targets for cardiovascular disease. Nat. Commun. 11:6417
    [Google Scholar]
  17. 17.
    Szustakowski JD, Balasubramanian S, Kvikstad E, Khalid S, Bronson PG et al. 2021. Advancing human genetics research and drug discovery through exome sequencing of the UK Biobank. Nat. Genet. 53:7942–48
    [Google Scholar]
  18. 18.
    Minikel EV, Karczewski KJ, Martin HC, Cummings BB, Whiffin N et al. 2020. Evaluating drug targets through human loss-of-function genetic variation. Nature 581:7809459–64
    [Google Scholar]
  19. 19.
    Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E et al. 2016. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536:7616285–91
    [Google Scholar]
  20. 20.
    Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J et al. 2020. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581:7809434–43
    [Google Scholar]
  21. 21.
    Finan C, Gaulton A, Kruger FA, Lumbers RT, Shah T et al. 2017. The druggable genome and support for target identification and validation in drug development. Sci. Transl. Med. 9:383eaag1166
    [Google Scholar]
  22. 22.
    Abul-Husn NS, Kenny EE. 2019. Personalized medicine and the power of electronic health records. Cell 177:158–69
    [Google Scholar]
  23. 23.
    Pendergrass SA, Crawford DC. 2019. Using electronic health records to generate phenotypes for research. Curr. Protoc. Hum. Genet. 100:1e80
    [Google Scholar]
  24. 24.
    Hripcsak G, Duke JD, Shah NH, Reich CG, Huser V et al. 2015. Observational Health Data Sciences and Informatics (OHDSI): opportunities for observational researchers. Stud. Health Technol. Inform. 216:574–78
    [Google Scholar]
  25. 25.
    Robinson JR, Denny JC, Roden DM, Van Driest SL. 2018. Genome-wide and phenome-wide approaches to understand variable drug actions in electronic health records. Clin. Transl. Sci. 11:2112–22
    [Google Scholar]
  26. 26.
    Denny JC, Ritchie MD, Basford MA, Pulley JM, Bastarache L et al. 2010. PheWAS: demonstrating the feasibility of a phenome-wide scan to discover gene-disease associations. Bioinformatics 26:91205–10
    [Google Scholar]
  27. 27.
    Denny JC, Bastarache L, Ritchie MD, Carroll RJ, Zink R et al. 2013. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat. Biotechnol. 31:121102–10
    [Google Scholar]
  28. 28.
    Rastegar-Mojarad M, Ye Z, Kolesar JM, Hebbring SJ, Lin SM. 2015. Opportunities for drug repositioning from phenome-wide association studies. Nat. Biotechnol. 33:4342–45
    [Google Scholar]
  29. 29.
    Diogo D, Tian C, Franklin CS, Alanne-Kinnunen M, March M et al. 2018. Phenome-wide association studies across large population cohorts support drug target validation. Nat. Commun. 9:4285
    [Google Scholar]
  30. 30.
    Jerome RN, Joly MM, Kennedy N, Shirey-Rice JK, Roden DM et al. 2020. Leveraging human genetics to identify safety signals prior to drug marketing approval and clinical use. Drug Saf. 43:6567–82
    [Google Scholar]
  31. 31.
    Millwood IY, Bennett DA, Walters RG, Clarke R, Waterworth D et al. 2016. A phenome-wide association study of a lipoprotein-associated phospholipase A2 loss-of-function variant in 90 000 Chinese adults. Int. J. Epidemiol. 45:51588–99
    [Google Scholar]
  32. 32.
    Roitmann E, Eriksson R, Brunak S. 2014. Patient stratification and identification of adverse event correlations in the space of 1190 drug related adverse events. Front. Physiol. 5:332
    [Google Scholar]
  33. 33.
    Warrer P, Hansen EH, Juhl-Jensen L, Aagaard L. 2012. Using text-mining techniques in electronic patient records to identify ADRs from medicine use. Br. J. Clin. Pharmacol. 73:5674–84
    [Google Scholar]
  34. 34.
    Wasylewicz A, van de Burgt B, Weterings A, Jessurun N, Korsten E et al. 2022. Identifying adverse drug reactions from free-text electronic hospital health record notes. Br. J. Clin. Pharmacol. 88:31235–45
    [Google Scholar]
  35. 35.
    Krebs K, Bovijn J, Zheng N, Lepamets M, Censin JC et al. 2020. Genome-wide study identifies association between HLA-B*55:01 and self-reported penicillin allergy. Am. J. Hum. Genet. 107:4612–21
    [Google Scholar]
  36. 36.
    Hebbring SJ, Rastegar-Mojarad M, Ye Z, Mayer J, Jacobson C, Lin S 2015. Application of clinical text data for phenome-wide association studies (PheWASs). Bioinformatics 31:121981–87
    [Google Scholar]
  37. 37.
    Denny JC, Ritchie MD, Basford MA, Pulley JM, Bastarache L et al. 2010. PheWAS: demonstrating the feasibility of a phenome-wide scan to discover gene-disease associations. Bioinformatics 26:91205–10
    [Google Scholar]
  38. 38.
    Wei W-Q, Bastarache LA, Carroll RJ, Marlo JE, Osterman TJ et al. 2017. Evaluating phecodes, clinical classification software, and ICD-9-CM codes for phenome-wide association studies in the electronic health record. PLOS ONE 12:7e0175508
    [Google Scholar]
  39. 39.
    Wu P, Gifford A, Meng X, Li X, Campbell H et al. 2019. Mapping ICD-10 and ICD-10-CM codes to phecodes: workflow development and initial evaluation. JMIR Med. Inform. 7:4e14325
    [Google Scholar]
  40. 40.
    Abifadel M, Varret M, Rabès JP, Allard D, Ouguerram K et al. 2003. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat. Genet. 34:2154–56
    [Google Scholar]
  41. 41.
    Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs HH. 2005. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat. Genet. 37:2161–65
    [Google Scholar]
  42. 42.
    Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. 2006. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N. Engl. J. Med. 354:121264–72
    [Google Scholar]
  43. 43.
    Sanna S, Li B, Mulas A, Sidore C, Kang HM et al. 2011. Fine mapping of five loci associated with low-density lipoprotein cholesterol detects variants that double the explained heritability. PLOS Genet. 7:7e1002198
    [Google Scholar]
  44. 44.
    Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G et al. 2015. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N. Engl. J. Med. 372:161489–99
    [Google Scholar]
  45. 45.
    Brunkow ME, Gardner JC, Van Ness J, Paeper BW, Kovacevich BR et al. 2001. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am. J. Hum. Genet. 68:3577–89
    [Google Scholar]
  46. 46.
    McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP et al. 2014. Romosozumab in postmenopausal women with low bone mineral density. N. Engl. J. Med. 370:5248–59
    [Google Scholar]
  47. 47.
    Amgen 2019. FDA approves EVENITYTM (romosozumab-aqqg) for the treatment of osteoporosis in postmenopausal women at high risk for fracture News Release Apr. 9. https://www.prnewswire.com/news-releases/fda-approves-evenity-romosozumab-aqqg-for-the-treatment-of-osteoporosis-in-postmenopausal-women-at-high-risk-for-fracture-300828376.html
  48. 48.
    Amgen 2019. European Commission approves EVENITY® (romosozumab) for the treatment of severe osteoporosis in postmenopausal women at high risk of fracture Press Release Dec. 11. https://www.amgen.com/newsroom/press-releases/2019/12/european-commission-approves-evenity-romosozumab-for-the-treatment-of-severe-osteoporosis-in-postmenopausal-women-at-high-risk-of-fracture
  49. 49.
    Harper AR, Nayee S, Topol EJ. 2015. Protective alleles and modifier variants in human health and disease. Nat. Rev. Genet. 16:12689–701
    [Google Scholar]
  50. 50.
    Kathiresan S, Melander O, Guiducci C, Surti A, Burtt NP et al. 2008. Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans. Nat. Genet. 40:2189–97
    [Google Scholar]
  51. 51.
    Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS et al. 2006. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314:58041461–63
    [Google Scholar]
  52. 52.
    Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR et al. 2008. A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology 135:41130–41
    [Google Scholar]
  53. 53.
    Wu P, Feng QP, Kerchberger VE, Nelson SD, Chen Q et al. 2022. Integrating gene expression and clinical data to identify drug repurposing candidates for hyperlipidemia and hypertension. Nat. Commun. 13:46
    [Google Scholar]
  54. 54.
    Bovijn J, Krebs K, Chen C-Y, Boxall R, Censin JC et al. 2020. Evaluating the cardiovascular safety of sclerostin inhibition using evidence from meta-analysis of clinical trials and human genetics. Sci. Transl. Med. 12:549eaay6570
    [Google Scholar]
  55. 55.
    Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM et al. 2010. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 375:9716735–42
    [Google Scholar]
  56. 56.
    Liu G, Shi M, Mosley JD, Weng C, Zhang Y et al. 2021. A Mendelian randomization approach using 3-HMG-coenzyme-A reductase gene variation to evaluate the association of statin-induced low-density lipoprotein cholesterol lowering with noncardiovascular disease phenotypes. JAMA Netw. Open 4:6e2112820
    [Google Scholar]
  57. 57.
    Chong M, Sjaarda J, Pigeyre M, Mohammadi-Shemirani P, Lali R et al. 2019. Novel drug targets for ischemic stroke identified through Mendelian randomization analysis of the blood proteome. Circulation 140:10819–30
    [Google Scholar]
  58. 58.
    Nguyen PA, Born DA, Deaton AM, Nioi P, Ward LD. 2019. Phenotypes associated with genes encoding drug targets are predictive of clinical trial side effects. Nat. Commun. 10:11579
    [Google Scholar]
  59. 59.
    COVID-19 Host Genet. Initiative 2020. The COVID-19 Host Genetics Initiative, a global initiative to elucidate the role of host genetic factors in susceptibility and severity of the SARS-CoV-2 virus pandemic. 2020. Eur. J. Hum. Genet. 28:6715–18
    [Google Scholar]
  60. 60.
    Satterfield BA, Dikilitas O, Kullo IJ. 2021. Leveraging the electronic health record to address the COVID-19 pandemic. Mayo Clin. Proc. 96:61592–608
    [Google Scholar]
  61. 61.
    Pairo-Castineira E, Clohisey S, Klaric L, Bretherick AD, Rawlik K et al. 2021. Genetic mechanisms of critical illness in COVID-19. Nature 591:784892–98
    [Google Scholar]
  62. 62.
    Severe Covid-19 GWAS Group 2020. Genomewide association study of severe Covid-19 with respiratory failure.. N. Engl. J. Med. 383:161522–34
    [Google Scholar]
  63. 63.
    Niemi MEK, Karjalainen J, Liao RG, Neale BM, Daly M et al. 2021. Mapping the human genetic architecture of COVID-19. Nature 600:7889472–77
    [Google Scholar]
  64. 64.
    Shelton JF, Shastri AJ, Ye C, Weldon CH, Filshtein-Sonmez T et al. 2021. Trans-ancestry analysis reveals genetic and nongenetic associations with COVID-19 susceptibility and severity. Nat. Genet. 53:6801–8
    [Google Scholar]
  65. 65.
    Huffman JE, Butler-Laporte G, Khan A, Pairo-Castineira E, Drivas TG et al. 2022. Multi-ancestry fine mapping implicates OAS1 splicing in risk of severe COVID-19. Nat. Genet. 54:2125–27
    [Google Scholar]
  66. 66.
    Daniloski Z, Jordan TX, Wessels HH, Hoagland DA, Kasela S et al. 2021. Identification of required host factors for SARS-CoV-2 infection in human cells. Cell 184:192–105.e16
    [Google Scholar]
  67. 67.
    Aguet F, Barbeira AN, Bonazzola R, Brown A, Castel SE et al. 2020. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369:65091318–30
    [Google Scholar]
  68. 68.
    Hao K, Bossé Y, Nickle DC, Paré PD, Postma DS et al. 2012. Lung eQTLs to help reveal the molecular underpinnings of asthma. PLOS Genet. 8:11e1003029
    [Google Scholar]
  69. 69.
    Palmos AB, Millischer V, Menon DK, Nicholson TR, Taams LS et al. 2022. Proteome-wide Mendelian randomization identifies causal links between blood proteins and severe COVID-19. PLOS Genet. 18:3e1010042
    [Google Scholar]
  70. 70.
    Gaziano L, Giambartolomei C, Pereira AC, Gaulton A, Posner DC et al. 2021. Actionable druggable genome-wide Mendelian randomization identifies repurposing opportunities for COVID-19. Nat. Med. 27:4668–76
    [Google Scholar]
  71. 71.
    Horowitz JE, Kosmicki JA, Damask A, Sharma D, Roberts GHL et al. 2022. Genome-wide analysis provides genetic evidence that ACE2 influences COVID-19 risk and yields risk scores associated with severe disease. Nat. Genet. 54:382–92
    [Google Scholar]
  72. 72.
    Vela D, Vela-Gaxha Z, Rexhepi M, Olloni R, Hyseni V, Nallbani R. 2022. Efficacy and safety of tocilizumab versus standard care/placebo in patients with COVID-19: a systematic review and meta-analysis of randomized clinical trials. Br. J. Clin. Pharmacol. 88:51955–63
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-051421-111324
Loading
/content/journals/10.1146/annurev-pharmtox-051421-111324
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error