Skip to main content

Pharmacogenomic and Statistical Analysis

  • Protocol
  • First Online:
Statistical Genomics

Part of the book series: Methods in Molecular Biology ((MIMB,volume 2629))

  • 2076 Accesses

Abstract

Genetic variants can alter response to drugs and other therapeutic interventions. The study of this phenomenon, called pharmacogenomics, is similar in many ways to other types of genetic studies but has distinct methodological and statistical considerations. Genetic variants involved in the processing of exogenous compounds exhibit great diversity and complexity, and the phenotypes studied in pharmacogenomics are also more complex than typical genetic studies. In this chapter, we review basic concepts in pharmacogenomic study designs, data generation techniques, statistical analysis approaches, and commonly used methods and briefly discuss the ultimate translation of findings to clinical care.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 189.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Martin CB, Hales CM, Gu Q, Ogden CL (2019) Prescription drug use in the United States, 2015–2019

    Google Scholar 

  2. Jonas DE, McLeod HL (2009) Genetic and clinical factors relating to warfarin dosing. Trends Pharmacol Sci 30:375–386. https://doi.org/10.1016/J.TIPS.2009.05.001

    Article  CAS  PubMed  Google Scholar 

  3. Savi P, Combalbert J, Gaich C et al (1994) The antiaggregating activity of clopidogrel is due to a metabolic activation by the hepatic cytochrome P450-1A. Thromb Haemost 72:313–317

    Article  CAS  PubMed  Google Scholar 

  4. Kee PS, Chin PKL, Kennedy MA, Maggo SDS (2020) Pharmacogenetics of statin-induced myotoxicity. Front Genet 11:575678. https://doi.org/10.3389/fgene.2020.575678

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Marciante KD, Durda JP, Heckbert SR et al (2011) Cerivastatin, genetic variants, and the risk of rhabdomyolysis. Pharmacogenet Genomics 21:280–288. https://doi.org/10.1097/FPC.0b013e328343dd7d

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mirošević Skvrce N, Macolić Šarinić V, Šimić I et al (2015) ABCG2 gene polymorphisms as risk factors for atorvastatin adverse reactions: a case-control study. Pharmacogenomics 16:803–815. https://doi.org/10.2217/pgs.15.47

    Article  CAS  PubMed  Google Scholar 

  7. Joffe E, Iasonos A, Younes A (2017) Clinical trials in the genomic era. J Clin Oncol 35:1011–1017. https://doi.org/10.1200/JCO.2016.70.8891

    Article  PubMed  PubMed Central  Google Scholar 

  8. Waarts MR, Stonestrom AJ, Park YC, Levine RL (2022) Targeting mutations in cancer. J Clin Invest 132. https://doi.org/10.1172/JCI154943

  9. Alexanderson B, Evans DAP, Sjöqvist F (1969) Steady-state plasma levels of nortriptyline in twins: influence of genetic factors and drug therapy. Br Med J 4:764–768. https://doi.org/10.1136/BMJ.4.5686.764

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mahgoub A, Dring LG, Idle JR et al (1977) Polymorphic hydroxylation of debrisoquine in man. Lancet 310:584–586. https://doi.org/10.1016/S0140-6736(77)91430-1

    Article  Google Scholar 

  11. Eichelbaum M, Spannbrucker N, Dengler HJ (1975) Proceedings: N-oxidation of sparteine in man and its interindividual differences. Naunyn Schmiedeberg's Arch Pharmacol 287 Suppl:R94

    CAS  Google Scholar 

  12. Robarge JD, Li L, Desta Z et al (2007) The star-allele nomenclature: retooling for translational genomics. Clin Pharmacol Ther 82:244–248. https://doi.org/10.1038/SJ.CLPT.6100284

    Article  CAS  PubMed  Google Scholar 

  13. Gonzalez FJ, Skodat RC, Kimura S et al (1988) Characterization of the common genetic defect in humans deficient in debrisoquine metabolism. Nature 331:442–446. https://doi.org/10.1038/331442A0

    Article  CAS  PubMed  Google Scholar 

  14. Bush WS, Moore JH (2012) Chapter 11: Genome-wide association studies. PLoS Comput Biol 8:e1002822. https://doi.org/10.1371/journal.pcbi.1002822

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Takeuchi F, McGinnis R, Bourgeois S et al (2009) A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet 5. https://doi.org/10.1371/journal.pgen.1000433

  16. Pulley JM, Denny JC, Peterson JF et al (2012) Operational implementation of prospective genotyping for personalized medicine: the design of the Vanderbilt PREDICT project. Clin Pharmacol Ther 92:87–95. https://doi.org/10.1038/clpt.2011.371

    Article  CAS  PubMed  Google Scholar 

  17. Schildcrout JS, Denny JC, Bowton E et al (2012) Optimizing drug outcomes through pharmacogenetics: a case for preemptive genotyping. Clin Pharmacol Ther 92:235–242. https://doi.org/10.1038/clpt.2012.66

    Article  CAS  PubMed  Google Scholar 

  18. Relling MV, Klein TE (2011) CPIC: Clinical pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther 89:464–467. https://doi.org/10.1038/clpt.2010.279

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Relling MV, Klein TE, Gammal RS et al (2020) The clinical pharmacogenetics implementation consortium: 10 years later. Clin Pharmacol Ther 107:171–175. https://doi.org/10.1002/cpt.1651

    Article  PubMed  Google Scholar 

  20. Klein TE, Chang JT, Cho MK et al (2001) Integrating genotype and phenotype information: an overview of the PharmGKB project. Pharmacogenetics Research Network and Knowledge Base. Pharmacogenomics J 1:167–170

    Article  CAS  PubMed  Google Scholar 

  21. Whirl-Carrillo M, McDonagh EM, Hebert JM et al (2012) Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther 92:414–417. https://doi.org/10.1038/clpt.2012.96

    Article  CAS  PubMed  Google Scholar 

  22. Van Booven D, Marsh S, McLeod H et al (2010) Cytochrome P450 2C9-CYP2C9. Pharmacogenet Genomics 20:277–281. https://doi.org/10.1097/FPC.0b013e3283349e84

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Thorn CF, Klein TE, Altman RB (2010) PharmGKB summary: very important pharmacogene information for angiotensin-converting enzyme. Pharmacogenet Genomics 20:143–146. https://doi.org/10.1097/FPC.0b013e3283339bf3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang L, Pelleymounter L, Weinshilboum R et al (2010) Very important pharmacogene summary: thiopurine S-methyltransferase. Pharmacogenet Genomics 20:401–405. https://doi.org/10.1097/FPC.0b013e3283352860

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mapes B, El Charif O, Al-Sawwaf S, Dolan ME (2017) Genome-wide association studies of chemotherapeutic toxicities: genomics of inequality. Clin Cancer Res 23:4010–4019. https://doi.org/10.1158/1078-0432.CCR-17-0429

    Article  PubMed  PubMed Central  Google Scholar 

  26. Maranville JC, Cox NJ (2016) Pharmacogenomic variants have larger effect sizes than genetic variants associated with other dichotomous complex traits. Pharmacogenomics J 16:388–392. https://doi.org/10.1038/tpj.2015.47

    Article  CAS  PubMed  Google Scholar 

  27. Zazuli Z, Otten LS, Drögemöller BI et al (2019) Outcome definition influences the relationship between genetic polymorphisms of ERCC1, ERCC2, SLC22A2 and cisplatin nephrotoxicity in adult testicular cancer patients. Genes (Basel) 10. https://doi.org/10.3390/genes10050364

  28. Higashi MK, Veenstra DL, Midori Kondo L et al (2002) Association between CYP2C9 genetic variants and anticoagulation-related outcomes during warfarin therapy. JAMA 287:1690–1698. https://doi.org/10.1001/JAMA.287.13.1690

    Article  CAS  PubMed  Google Scholar 

  29. Vuorinen A-L, Lehto M, Niemi M et al (2021) Pharmacogenetics of anticoagulation and clinical events in warfarin-treated patients: a register-based cohort study with biobank data and national health registries in Finland. Clin Epidemiol 13:183–195. https://doi.org/10.2147/CLEP.S289031

    Article  PubMed  PubMed Central  Google Scholar 

  30. Wheeler HE, Dolan ME (2012) Lymphoblastoid cell lines in pharmacogenomic discovery and clinical translation. Pharmacogenomics 13:55–70. https://doi.org/10.2217/pgs.11.121

    Article  CAS  PubMed  Google Scholar 

  31. Moen EL, Godley LA, Zhang W, Dolan ME (2012) Pharmacogenomics of chemotherapeutic susceptibility and toxicity. Genome Med 4. https://doi.org/10.1186/gm391

  32. Belmont JW, Hardenbol P, Willis TD et al (2003) The international HapMap project. Nature 426:789–796. https://doi.org/10.1038/NATURE02168

    Article  Google Scholar 

  33. International HapMap Consortium (2005) A haplotype map of the human genome. Nature 437:1299–1320. https://doi.org/10.1038/nature04226

    Article  CAS  Google Scholar 

  34. Abecasis GR, Auton A, Brooks LD et al (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491:56–65. https://doi.org/10.1038/nature11632

    Article  CAS  PubMed  Google Scholar 

  35. Lappalainen T, Sammeth M, Friedländer MR et al (2013) Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501:506–511. https://doi.org/10.1038/nature12531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bell JT, Pai AA, Pickrell JK et al (2011) DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol 12. https://doi.org/10.1186/gb-2011-12-1-r10

  37. Gamazon ER, Im H-K, Duan S et al (2010) Exprtarget: an integrative approach to predicting human microRNA targets. PLoS One 5:e13534. https://doi.org/10.1371/journal.pone.0013534

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Relling MV, Krauss RM, Roden DM et al (2017) New pharmacogenomics research network: an open community catalyzing research and translation in precision medicine. Clin Pharmacol Ther 102:897–902. https://doi.org/10.1002/cpt.755

    Article  CAS  PubMed  Google Scholar 

  39. Huang RS, Duan S, Bleibel WK et al (2007) A genome-wide approach to identify genetic variants that contribute to etoposide-induced cytotoxicity. Proc Natl Acad Sci U S A 104:9758–9763. https://doi.org/10.1073/PNAS.0703736104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Komatsu M, Wheeler HE, Chung S et al (2015) Pharmacoethnicity in paclitaxel-induced sensory peripheral neuropathy. Clin Cancer Res 21:4337–4346. https://doi.org/10.1158/1078-0432.CCR-15-0133

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mulford AJ, Wing C, Dolan ME, Wheeler HE (2021) Genetically regulated expression underlies cellular sensitivity to chemotherapy in diverse populations. Hum Mol Genet 30:305–317. https://doi.org/10.1093/hmg/ddab029

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Njiaju UO, Gamazon ER, Gorsic LK et al (2012) Whole-genome studies identify solute carrier transporters in cellular susceptibility to paclitaxel. Pharmacogenet Genomics 22:498–507. https://doi.org/10.1097/FPC.0b013e328352f436

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Turner S, Armstrong LL, Bradford Y et al (2011) Quality control procedures for genome-wide association studies. Curr Protoc Hum Genet. https://doi.org/10.1002/0471142905.hg0119s68

  44. Gordon AS, Fulton RS, Qin X et al (2016) PGRNseq: a targeted capture sequencing panel for pharmacogenetic research and implementation. Pharmacogenet Genomics 26:161–168. https://doi.org/10.1097/FPC.0000000000000202

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Grada A, Weinbrecht K (2013) Next-generation sequencing: methodology and application. J Invest Dermatol 133. https://doi.org/10.1038/JID.2013.248

  46. Bolger AM, Lohse M, Usadel B (2014) Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120. https://doi.org/10.1093/BIOINFORMATICS/BTU170

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. GitHub – FelixKrueger/TrimGalore: a wrapper around Cutadapt and FastQC to consistently apply adapter and quality trimming to FastQ files, with extra functionality for RRBS data. https://github.com/FelixKrueger/TrimGalore. Accessed 4 Jun 2020

  48. Langmead B, Trapnell C, Pop M, Salzberg SL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10:R25. https://doi.org/10.1186/gb-2009-10-3-r25

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li H, Durbin R (2009) Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25:1754–1760. https://doi.org/10.1093/bioinformatics/btp324

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Donato L, Scimone C, Rinaldi C et al (2021) New evaluation methods of read mapping by 17 aligners on simulated and empirical NGS data: an updated comparison of DNA- and RNA-Seq data from Illumina and Ion Torrent technologies. Neural Comput Appl 33:15669–15692. https://doi.org/10.1007/s00521-021-06188-z

    Article  PubMed  PubMed Central  Google Scholar 

  51. Li H, Wren J (2014) Toward better understanding of artifacts in variant calling from high-coverage samples. Bioinformatics 30:2843–2851. https://doi.org/10.1093/bioinformatics/btu356

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. McKenna A, Hanna M, Banks E et al (2010) The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20:1297–1303. https://doi.org/10.1101/gr.107524.110

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Challis D, Yu J, Evani US et al (2012) An integrative variant analysis suite for whole exome next-generation sequencing data. BMC Bioinform 13. https://doi.org/10.1186/1471-2105-13-8

  54. Miller NA, Farrow EG, Gibson M et al (2015) A 26-hour system of highly sensitive whole genome sequencing for emergency management of genetic diseases. Genome Med 7. https://doi.org/10.1186/s13073-015-0221-8

  55. Poplin R, Chang PC, Alexander D et al (2018) A universal snp and small-indel variant caller using deep neural networks. Nat Biotechnol 36:983. https://doi.org/10.1038/nbt.4235

    Article  CAS  PubMed  Google Scholar 

  56. Zhao S, Agafonov O, Azab A et al (2020) Accuracy and efficiency of germline variant calling pipelines for human genome data. Sci Rep 10. https://doi.org/10.1038/s41598-020-77218-4

  57. Naj AC, Lin H, Vardarajan BN et al (2018) Quality control and integration of genotypes from two calling pipelines for whole genome sequence data in the Alzheimer’s disease sequencing project. Genomics. https://doi.org/10.1016/j.ygeno.2018.05.004

  58. Van der Auwera GA, Carneiro MO, Hartl C et al (2002) Current protocols in bioinformatics. Wiley, Hoboken, NJ

    Google Scholar 

  59. Anderson CA, Pettersson FH, Clarke GM et al (2010) Data quality control in genetic case-control association studies. Nat Protoc 5:1564–1573. https://doi.org/10.1038/nprot.2010.116

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wittke-Thompson JK, Pluzhnikov A, Cox NJ (2005) Rational inferences about departures from Hardy-Weinberg equilibrium. Am J Hum Genet 76:967–986. https://doi.org/10.1086/430507

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang MH, Cordell HJ, Van Steen K (2019) Statistical methods for genome-wide association studies. Semin Cancer Biol 55:53–60. https://doi.org/10.1016/j.semcancer.2018.04.008

    Article  PubMed  Google Scholar 

  62. Price AL, Patterson NJ, Plenge RM et al (2006) Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet 38:904–909. https://doi.org/10.1038/ng1847

    Article  CAS  PubMed  Google Scholar 

  63. Hall JB, Dumitrescu L, Dilks HH et al (2014) Accuracy of administratively-assigned ancestry for diverse populations in an electronic medical record-linked biobank. PLoS One 9:e99161. https://doi.org/10.1371/journal.pone.0099161

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Dumitrescu L, Ritchie MD, Brown-Gentry K et al (2010) Assessing the accuracy of observer-reported ancestry in a biorepository linked to electronic medical records. Genet Med 12:648–650. https://doi.org/10.1097/GIM.0b013e3181efe2df

    Article  PubMed  PubMed Central  Google Scholar 

  65. Lettre G, Lange C, Hirschhorn JN (2007) Genetic model testing and statistical power in population-based association studies of quantitative traits. Genet Epidemiol 31:358–362. https://doi.org/10.1002/gepi.20217

    Article  PubMed  Google Scholar 

  66. Chang CC, Chow CC, Tellier LC et al (2015) Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4:7. https://doi.org/10.1186/s13742-015-0047-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Carroll RJ, Bastarache L, Denny JC (2014) R PheWAS: data analysis and plotting tools for phenome-wide association studies in the R environment. Bioinformatics 30(16):2375–2376. https://doi.org/10.1093/bioinformatics/btu197

  68. Willer CJ, Li Y, Abecasis GR (2010) METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26(17):2190–2191. https://doi.org/10.1093/bioinformatics/btq340

  69. Fraser AG, Orchard TR, Jewell DP (2002) The efficacy of azathioprine for the treatment of inflammatory bowel disease: a 30 year review. Gut 50:485–489. https://doi.org/10.1136/GUT.50.4.485

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yang SK, Hong M, Baek J et al (2014) A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 46:1017–1020. https://doi.org/10.1038/NG.3060

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Liu N, Zhang K, Zhao H (2008) Haplotype-association analysis. Adv Genet 60:335–405. https://doi.org/10.1016/S0065-2660(07)00414-2

    Article  PubMed  Google Scholar 

  72. Lee CR, Goldstein JA, Pieper JA (2002) Cytochrome P450 2C9 polymorphisms: a comprehensive review of the in-vitro and human data. Pharmacogenetics 12:251–263. https://doi.org/10.1097/00008571-200204000-00010

    Article  CAS  PubMed  Google Scholar 

  73. Excoffier L, Slatkin M (1995) Maximum-likelihood estimation of molecular haplotype frequencies in a diploid population. Mol Biol Evol 12:921–927. https://doi.org/10.1093/oxfordjournals.molbev.a040269

    Article  CAS  PubMed  Google Scholar 

  74. Stephens M, Smith NJ, Donnelly P (2001) A new statistical method for haplotype reconstruction from population data. Am J Hum Genet 68:978–989. https://doi.org/10.1086/319501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Li N, Stephens M (2003) Modeling linkage disequilibrium and identifying recombination hotspots using single-nucleotide polymorphism data. Genetics 165:2213–2233. https://doi.org/10.1093/genetics/165.4.2213

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Delaneau O, Marchini J, Zagury JF (2012) A linear complexity phasing method for thousands of genomes. Nat Methods 9:179–181. https://doi.org/10.1038/nmeth.1785

    Article  CAS  Google Scholar 

  77. Shi S, Yuan N, Yang M et al (2018) Comprehensive assessment of genotype imputation performance. Hum Hered 83:107–116. https://doi.org/10.1159/000489758

    Article  CAS  PubMed  Google Scholar 

  78. Clarke L, Fairley S, Zheng-Bradley X et al (2017) The international Genome sample resource (IGSR): a worldwide collection of genome variation incorporating the 1000 Genomes Project data. Nucleic Acids Res 45:D854–D859. https://doi.org/10.1093/nar/gkw829

    Article  CAS  PubMed  Google Scholar 

  79. McCarthy S, Das S, Kretzschmar W et al (2016) A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet 48:1279–1283. https://doi.org/10.1038/ng.3643

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Das S, Forer L, Schönherr S et al (2016) Next-generation genotype imputation service and methods. Nat Genet 48:1284–1287. https://doi.org/10.1038/ng.3656

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Taliun D, Harris DN, Kessler MD et al (2021) Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature 590:290–299. https://doi.org/10.1038/S41586-021-03205-Y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Vergara C, Parker MM, Franco L et al (2018) Genotype imputation performance of three reference panels using African ancestry individuals. Hum Genet 137:281–292. https://doi.org/10.1007/S00439-018-1881-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Del Tredici AL, Malhotra A, Dedek M et al (2018) Frequency of CYP2D6 alleles including structural variants in the United States. Front Pharmacol 9:305. https://doi.org/10.3389/fphar.2018.00305

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Molden E, Jukić MM (2021) CYP2D6 reduced function variants and genotype/phenotype translations of CYP2D6 intermediate metabolizers: implications for personalized drug dosing in psychiatry. Front Pharmacol 12:650750. https://doi.org/10.3389/fphar.2021.650750

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Häkkinen K, Kiiski JI, Lähteenvuo M et al (2022) Implementation of CYP2D6 copy-number imputation panel and frequency of key pharmacogenetic variants in Finnish individuals with a psychotic disorder. Pharmacogenomics J. https://doi.org/10.1038/s41397-022-00270-y

  86. Ammar R, Paton TA, Torti D et al (2015) Long read nanopore sequencing for detection of HLA and CYP2D6 variants and haplotypes. F1000Research 4:17. https://doi.org/10.12688/f1000research.6037.2

    Article  PubMed  PubMed Central  Google Scholar 

  87. Laver TW, Caswell RC, Moore KA et al (2016) Pitfalls of haplotype phasing from amplicon-based long-read sequencing. Sci Rep 6. https://doi.org/10.1038/srep21746

  88. Mantere T, Kersten S, Hoischen A (2019) Long-read sequencing emerging in medical genetics. Front Genet 10:426. https://doi.org/10.3389/fgene.2019.00426

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Limdi NA, Wadelius M, Cavallari L et al (2010) Warfarin pharmacogenetics: a single VKORC1 polymorphism is predictive of dose across 3 racial groups. Blood 115:3827–3834. https://doi.org/10.1182/BLOOD-2009-12-255992

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Diaz FJ, Yeh H-W, de Leon J (2012) Role of statistical random-effects linear models in personalized medicine. Curr Pharmacogenomics Person Med 10:22–32. https://doi.org/10.2174/1875692111201010022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Kaur H, Crawford DC, Liang J et al (2021) Replication of European hypertension associations in a case-control study of 9,534 African Americans. PLoS One 16. https://doi.org/10.1371/journal.pone.0259962

  92. Wilke RA, Berg RL, Linneman JG et al (2008) Characterization of low-density lipoprotein cholesterol-lowering efficacy for atorvastatin in a population-based DNA biorepository. Basic Clin Pharmacol Toxicol 103:354–359. https://doi.org/10.1111/j.1742-7843.2008.00291.x

    Article  CAS  PubMed  Google Scholar 

  93. Peissig P, Sirohi E, Berg RL et al (2007) Construction of atorvastatin dose-response relationships using data from a large population-based DNA Biobank. Basic Clin Pharmacol Toxicol 100:286–288. https://doi.org/10.1111/j.1742-7843.2006.00035.x

    Article  CAS  PubMed  Google Scholar 

  94. Vis DJ, Bombardelli L, Lightfoot H et al (2016) Multilevel models improve precision and speed of IC50 estimates. Pharmacogenomics 17:691–700. https://doi.org/10.2217/pgs.16.15

    Article  CAS  PubMed  Google Scholar 

  95. Wei WQ, Feng Q, Jiang L et al (2014) Characterization of statin dose response in electronic medical records. Clin Pharmacol Ther 95:331–338. https://doi.org/10.1038/CLPT.2013.202

    Article  CAS  PubMed  Google Scholar 

  96. Abbas-Aghababazadeh F, Lu P, Fridley BL (2019) Nonlinear mixed-effects models for modeling in vitro drug response data to determine problematic cancer cell lines. Sci Rep 9. https://doi.org/10.1038/S41598-019-50936-0

  97. Yang J, Lee SH, Goddard ME, Visscher PM (2011) GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet 88:76–82. https://doi.org/10.1016/j.ajhg.2010.11.011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Hall JB, Bush WS (2016) Analysis of heritability using genome-wide data. Curr Protoc Hum Genet 91:1.30.1–1.30.10. https://doi.org/10.1002/cphg.25

    Article  PubMed  Google Scholar 

  99. Yang J, Benyamin B, McEvoy BP et al (2010) Common SNPs explain a large proportion of the heritability for human height. Nat Genet 42:565–569. https://doi.org/10.1038/ng.608

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Zhao JH (2005) Mixed-effects Cox models of alcohol dependence in extended families. BMC Genet 6. https://doi.org/10.1186/1471-2156-6-S1-S127

  101. Yang CC, Zhang B, Wang S et al (2019) Gene-based association analysis of survival traits via functional regression-based mixed effect cox models for related samples. Genet Epidemiol 43:952–965. https://doi.org/10.1002/gepi.22254

    Article  CAS  Google Scholar 

  102. Mcgeachie MJ, Stahl EA, Himes BE et al (2013) Polygenic heritability estimates in pharmacogenetics: focus on asthma and related phenotypes. Pharmacogenet Genomics 23:324–328. https://doi.org/10.1097/FPC.0b013e3283607acf

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Cecchin E, Stocco G (2020) Pharmacogenomics and personalized medicine. Genes (Basel) 11:1–5. https://doi.org/10.3390/genes11060679

    Article  CAS  Google Scholar 

  104. Butkiewicz M, Blue EE et al (2018) Functional annotation of genomic variants in studies of late-onset Alzheimer’s disease. Bioinformatics. https://doi.org/10.1093/bioinformatics/bty177

  105. Bush WS, Crosslin DR, Obeng AO et al (2016) Genetic variation among 82 pharmacogenes: the PGRN-Seq data from the eMERGE network. Clin Pharmacol Ther. https://doi.org/10.1002/cpt.350

  106. Rasmussen-Torvik LJ, Stallings SC, Gordon AS et al (2014) Design and anticipated outcomes of the eMERGE-PGx project: a multicenter pilot for preemptive pharmacogenomics in electronic health record systems. Clin Pharmacol Ther 96:482–489. https://doi.org/10.1038/clpt.2014.137

    Article  CAS  PubMed  Google Scholar 

  107. Ingelman-Sundberg M, Mkrtchian S, Zhou Y, Lauschke VM (2018) Integrating rare genetic variants into pharmacogenetic drug response predictions. Hum Genomics 12. https://doi.org/10.1186/s40246-018-0157-3

  108. Zhou Y, Mkrtchian S, Kumondai M et al (2019) An optimized prediction framework to assess the functional impact of pharmacogenetic variants. Pharmacogenomics J 19:115–126. https://doi.org/10.1038/s41397-018-0044-2

    Article  CAS  PubMed  Google Scholar 

  109. Li B, Leal SM (2008) Methods for detecting associations with rare variants for common diseases: application to analysis of sequence data. Am J Hum Genet 83:311–321. https://doi.org/10.1016/j.ajhg.2008.06.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Wu MC, Lee S, Cai T et al (2011) Rare-variant association testing for sequencing data with the sequence kernel association test. Am J Hum Genet 89:82–93. https://doi.org/10.1016/j.ajhg.2011.05.029

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lee S, Abecasis GR, Boehnke M, Lin X (2014) Rare-variant association analysis: study designs and statistical tests. Am J Hum Genet 95:5–23. https://doi.org/10.1016/j.ajhg.2014.06.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. West RM, Lu W, Rotroff DM et al (2019) Identifying individual risk rare variants using protein structure guided local tests (POINT). PLoS Comput Biol 15. https://doi.org/10.1371/JOURNAL.PCBI.1006722

  113. Tang ZZ, Sliwoski GR, Chen G et al (2020) PSCAN: spatial scan tests guided by protein structures improve complex disease gene discovery and signal variant detection. Genome Biol 21. https://doi.org/10.1186/s13059-020-02121-0

  114. Jin B, Capra JA, Benchek P et al (2022) An association test of the spatial distribution of rare missense variants within protein structures identifies Alzheimer’s disease-related patterns. Genome Res 32:778–790. https://doi.org/10.1101/gr.276069.121

    Article  PubMed  PubMed Central  Google Scholar 

  115. Pasta A, Cremonini AL, Pisciotta L et al (2020) PCSK9 inhibitors for treating hypercholesterolemia. Expert Opin Pharmacother 21:353–363. https://doi.org/10.1080/14656566.2019.1702970

    Article  CAS  PubMed  Google Scholar 

  116. Maleki F, Ovens K, Hogan DJ, Kusalik AJ (2020) Gene set analysis: challenges, opportunities, and future research. Front Genet 11:654. https://doi.org/10.3389/fgene.2020.00654

    Article  PubMed  PubMed Central  Google Scholar 

  117. Subramanian A, Tamayo P, Mootha VK et al (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102:15545–15550. https://doi.org/10.1073/pnas.0506580102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ashburner M, Ball CA, Blake JA et al (2000) Gene ontology: tool for the unification of biology. Gene Ontol Consortium Nat Genet 25:25–29. https://doi.org/10.1038/75556

    Article  CAS  Google Scholar 

  119. Carbon S, Douglass E, Good BM et al (2021) The gene ontology resource: enriching a GOld mine. Nucleic Acids Res 49:D325–D334. https://doi.org/10.1093/nar/gkaa1113

    Article  CAS  Google Scholar 

  120. Kanehisa M, Goto S (2000) KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res 28:27–30. https://doi.org/10.1093/nar/28.1.27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Kanehisa M, Furumichi M, Sato Y et al (2021) KEGG: integrating viruses and cellular organisms. Nucleic Acids Res 49:D545–D551. https://doi.org/10.1093/nar/gkaa970

    Article  CAS  PubMed  Google Scholar 

  122. Whirl-Carrillo M, Huddart R, Gong L et al (2021) An evidence-based framework for evaluating pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther 110:563–572. https://doi.org/10.1002/cpt.2350

    Article  PubMed  PubMed Central  Google Scholar 

  123. Jiao X, Sherman BT, Huang DW et al (2012) DAVID-WS: a stateful web service to facilitate gene/protein list analysis. Bioinformatics 28:1805–1806. https://doi.org/10.1093/BIOINFORMATICS/BTS251

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Liao Y, Wang J, Jaehnig EJ et al (2019) WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res 47:W199–W205. https://doi.org/10.1093/NAR/GKZ401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Collins KS, Pratt VM, Stansberry WM et al (2019) Analytical validity of a genotyping assay for use with personalized antihypertensive and chronic kidney disease therapy. Pharmacogenet Genomics 29:18–22. https://doi.org/10.1097/FPC.0000000000000361

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lin CH, Yeakley JM, McDaniel TK, Shen R (2009) Medium- to high-throughput SNP genotyping using VeraCode microbeads. Methods Mol Biol 496:129–142. https://doi.org/10.1007/978-1-59745-553-4_10

    Article  CAS  PubMed  Google Scholar 

  127. Goldspiel BR, Flegel WA, DiPatrizio G et al (2014) Integrating pharmacogenetic information and clinical decision support into the electronic health record. J Am Med Inform Assoc 21:522–528. https://doi.org/10.1136/AMIAJNL-2013-001873

    Article  PubMed  Google Scholar 

  128. Dunnenberger HM, Crews KR, Hoffman JM et al (2015) Preemptive clinical pharmacogenetics implementation: current programs in five US medical centers. Annu Rev Pharmacol Toxicol 55:89–106. https://doi.org/10.1146/ANNUREV-PHARMTOX-010814-124835

    Article  CAS  PubMed  Google Scholar 

  129. Schildcrout JS, Shi Y, Danciu I et al (2016) A prognostic model based on readily available clinical data enriched a pre-emptive pharmacogenetic testing program. J Clin Epidemiol 72:107–115. https://doi.org/10.1016/J.JCLINEPI.2015.08.028

    Article  PubMed  Google Scholar 

  130. Rogers SL, Keeling NJ, Giri J et al (2020) PARC report: a health-systems focus on reimbursement and patient access to pharmacogenomics testing. Pharmacogenomics 21:785–796. https://doi.org/10.2217/PGS-2019-0192

    Article  Google Scholar 

  131. Jain KK (2016) Role of proteomics in the development of personalized medicine. Adv Protein Chem Struct Biol 102:41–52. https://doi.org/10.1016/BS.APCSB.2015.09.002

    Article  CAS  PubMed  Google Scholar 

  132. Clayton TA, Lindon JC, Cloarec O et al (2006) Pharmaco-metabonomic phenotyping and personalized drug treatment. Nature 440:1073–1077. https://doi.org/10.1038/NATURE04648

    Article  CAS  PubMed  Google Scholar 

  133. Pavlovic S, Kotur N, Stankovic B et al (2019) Pharmacogenomic and pharmacotranscriptomic profiling of childhood acute lymphoblastic Leukemia: paving the way to personalized treatment. Genes (Basel) 10. https://doi.org/10.3390/GENES10030191

  134. Mullard A (2022) 2021 FDA approvals. Nat Rev Drug Discov 21:83–88. https://doi.org/10.1038/D41573-022-00001-9

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to William S. Bush .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2023 The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Bai, H., Zhang, X., Bush, W.S. (2023). Pharmacogenomic and Statistical Analysis. In: Fridley, B., Wang, X. (eds) Statistical Genomics. Methods in Molecular Biology, vol 2629. Humana, New York, NY. https://doi.org/10.1007/978-1-0716-2986-4_14

Download citation

  • DOI: https://doi.org/10.1007/978-1-0716-2986-4_14

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-0716-2985-7

  • Online ISBN: 978-1-0716-2986-4

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics