Challenges of Clinical Pharmacogenomics Implementation in the Era of Precision Medicine

Main Article Content

Sherin Shaaban Yuan Ji

Abstract

While pharmacogenomics (PGx) presented to many as the poster child of personalized medicine, in the context of moving away from the “one size fits all” model of pharmacological therapy of diseases to a more tailored approach addressing the individuality of each person, the application of PGx has been hindered by numerous challenges. These challenges range from issues with study designs in scientific research, both clinical and for discovery, policy and regulatory hurdles affecting insurance coverage, to the very fundamental need for adequate training for laboratorians, physicians, and pharmacists. Moreover, access to services and addressing health disparities in personalized medicine generally and PGx specifically remain a complicated endeavor. PGx-related ethical, legal, and social issues continue to be a point of concern for those looking to implement PGx in clinical practice. Additionally on the technical side, the speed with which next generation sequencing (NGS) technologies have evolved, generating tens of thousands of rare PGx variants adds a new layer of challenges requiring accurate interpretation and assessment of functional role of novel variants to determine their impacts on drug response and possible toxicities. Without consensus and standardized approaches to testing and interpretation, integration of PGx into routine clinical care becomes an unattainable task. In this article we aim to address some of the challenges that impede broad adoption of clinical PGx testing, and to shed the light on needed steps towards a successful implementation of PGx, with the goal of improving health outcomes individually and for the general population.

Article Details

How to Cite
SHAABAN, Sherin; JI, Yuan. Challenges of Clinical Pharmacogenomics Implementation in the Era of Precision Medicine. Medical Research Archives, [S.l.], v. 12, n. 11, nov. 2024. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/5955>. Date accessed: 12 dec. 2024. doi: https://doi.org/10.18103/mra.v12i11.5955.
Section
Research Articles

References

1. U.S. Food & Drug Adminstration. [cited 2024 9/25/2024]; Available from:
https://www.fda.gov/

2. National Human Genome Research Institute. [cited 2024 09/25/2024]; Available from:
https://www.genome.gov/genetics-glossary/Precision-Medicine.

3. Roses, A.D., Pharmacogenetics and drug development: the path to safer and more effective drugs. Nat Rev Genet, 2004. 5(9): p. 645-56.

4. Evans, W.E. and H.L. McLeod, Pharmacogenomics--drug disposition, drug targets, and side effects. N Engl J Med, 2003. 348(6): p. 538-49.

5. Klein, K., et al., A New Panel-Based Next-Generation Sequencing Method for ADME Genes Reveals Novel Associations of Common and Rare Variants With Expression in a Human Liver Cohort. Front Genet, 2019. 10: p. 7.

6. Hovelson, D.H., et al., Characterization of ADME gene variation in 21 populations by exome sequencing. Pharmacogenet Genomics, 2017. 27 (3): p. 89-100.

7. Zanger, U.M., Pharmacogenetics - challenges and opportunities ahead. Front Pharmacol, 2010. 1: p. 112.

8. Bienfait, K., et al., Current challenges and opportunities for pharmacogenomics: perspective of the Industry Pharmacogenomics Working Group (I-PWG). Hum Genet, 2022. 141(6): p. 1165-1173.

9. Gershon, E.S., N. Alliey-Rodriguez, and K. Grennan, Ethical and public policy challenges for pharmacogenomics. Dialogues Clin Neurosci, 2014. 16(4): p. 567-74.

10. Caudle, K.E., et al., Standardization can accelerate the adoption of pharmacogenomics: current status and the path forward. Pharmacogenomics, 2018. 19(10): p. 847-860.

11. Das, T., et al., Leveraging multi-source to resolve inconsistency across pharmacogenomic datasets in drug sensitivity prediction. medRxiv, 2023.

12. Cilluffo, G., et al., Machine Learning: An Overview and Applications in Pharmacogenetics. Genes (Basel), 2021. 12(10).

13. Silva, P., et al., Implementation of Pharmacogenomics and Artificial Intelligence Tools for Chronic Disease Management in Primary Care Setting. J Pers Med, 2021. 11(6).

14. Wisler, J.R., et al., Challenges and opportunities in implementing pharmacogenomics testing in the clinics. Per Med, 2012. 9(6): p. 609-619.

15. Tong, H., et al., Review on Databases and Bioinformatic Approaches on Pharmacogenomics of Adverse Drug Reactions. Pharmgenomics Pers Med, 2021. 14: p. 61-75.

16. Katsila, T. and G.P. Patrinos, Whole genome sequencing in pharmacogenomics. Front Pharmacol, 2015. 6: p. 61.

17. Whirl-Carrillo, M., et al., An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin Pharmacol Ther, 2021. 110(3): p. 563-572.

18. Whirl-Carrillo, M., et al., Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther, 2012. 92(4): p. 414-7.

19. Relling, M.V. and T.E. Klein, CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther, 2011. 89(3): p. 464-7.

20. Gaedigk, A., et al., PharmVar and the Landscape of Pharmacogenetic Resources. Clin Pharmacol Ther, 2020. 107(1): p. 43-46.

21. Gaedigk, A., et al., The Pharmacogene Variation (PharmVar) Consortium: Incorporation of the Human Cytochrome P450 (CYP) Allele Nomenclature Database. Clin Pharmacol Ther, 2018. 103(3): p. 399-401.

22. Morris, S.A., et al., Cost Effectiveness of Pharmacogenetic Testing for Drugs with Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines: A Systematic Review. Clin Pharmacol Ther, 2022. 112(6): p. 1318-1328.

23. Saldivar, J.S., et al., Initial assessment of the benefits of implementing pharmacogenetics into the medical management of patients in a long-term care facility. Pharmgenomics Pers Med, 2016. 9: p. 1-6.

24. Verbelen, M., M.E. Weale, and C.M. Lewis, Cost-effectiveness of pharmacogenetic-guided treatment: are we there yet? Pharmacogenomics J, 2017. 17(5): p. 395-402.

25. Skokou, M., et al., Clinical implementation of preemptive pharmacogenomics in psychiatry. EBioMedicine, 2024. 101: p. 105009.

26. Huang, Q., et al., A retrospective analysis of preemptive pharmacogenomic testing in 22,918 individuals from China. J Clin Lab Anal, 2023. 37(5): p. e24855.

27. Lu, M., C.M. Lewis, and M. Traylor, Pharmacogenetic testing through the direct-to-consumer genetic testing company 23andMe. BMC Med Genomics, 2017. 10(1): p. 47.

28. Haga, S.B., Challenges of development and implementation of point of care pharmacogenetic testing. Expert Rev Mol Diagn, 2016. 16(9): p. 949-60.

29. Breaux, S., et al., Pharmacogenomics at the Point of Care: A Community Pharmacy Project in British Columbia. J Pers Med, 2020. 11(1).

30. Magavern, E.F., et al., Health equality, race and pharmacogenomics. Br J Clin Pharmacol, 2022. 88(1): p. 27-33.

31. Venkatakrishnan, K. and L.J. Benincosa, Diversity and Inclusion in Drug Development: Rethinking Intrinsic and Extrinsic Factors with Patient Centricity. Clin Pharmacol Ther, 2022. 112 (2): p. 204-207.

32. Braveman, P. and L. Gottlieb, The social determinants of health: it's time to consider the causes of the causes. Public Health Rep, 2014. 129 Suppl 2(Suppl 2): p. 19-31.

33. Ginsburg, G.S. and K.A. Phillips, Precision Medicine: From Science To Value. Health Aff (Millwood), 2018. 37(5): p. 694-701.

34. Sisodiya, S.M., Precision medicine and therapies of the future. Epilepsia, 2021. 62 Suppl 2(Suppl 2): p. S90-S105.

35. Peterson-Iyer, K., Pharmacogenomics, ethics, and public policy. Kennedy Inst Ethics J, 2008. 18(1): p. 35-56.

36. Stratton, T.P. and A.W. Olson, Personalizing Personalized Medicine: The Confluence of Pharmacogenomics, a Person's Medication Experience and Ethics. Pharmacy (Basel), 2023. 11(3).

37. Katara, P. and A. Yadav, Pharmacogenes (PGx-genes): Current understanding and future directions. Gene, 2019. 718: p. 144050.

38. Mostafa, S., et al., An analysis of allele, genotype and phenotype frequencies, actionable pharmacogenomic (PGx) variants and phenoconversion in 5408 Australian patients genotyped for CYP2D6, CYP2C19, CYP2C9 and VKORC1 genes. J Neural Transm (Vienna), 2019. 126(1): p. 5-18.

39. Jithesh, P.V., et al., A population study of clinically actionable genetic variation affecting drug response from the Middle East. NPJ Genom Med, 2022. 7(1): p. 10.

40. Van Driest, S.L., et al., Clinically actionable genotypes among 10,000 patients with preemptive pharmacogenomic testing. Clin Pharmacol Ther, 2014. 95(4): p. 423-31.

41. Matthaei, J., et al., Heritability of metoprolol and torsemide pharmacokinetics. Clin Pharmacol Ther, 2015. 98(6): p. 611-21.

42. Ingelman-Sundberg, M., et al., Integrating rare genetic variants into pharmacogenetic drug response predictions. Hum Genomics, 2018. 12(1): p. 26.

43. Muhammad, A., et al., Genome-Wide Approach to Measure Variant-Based Heritability of Drug Outcome Phenotypes. Clin Pharmacol Ther, 2021. 110(3): p. 714-722.

44. Kozyra, M., M. Ingelman-Sundberg, and V.M. Lauschke, Rare genetic variants in cellular transporters, metabolic enzymes, and nuclear receptors can be important determinants of interindividual differences in drug response. Genet Med, 2017. 19(1): p. 20-29.

45. Markianos, K., et al., Pharmacogenetic allele variant frequencies: An analysis of the VA's Million Veteran Program (MVP) as a representation of the diversity in US population. PLoS One, 2023. 18(2): p. e0274339.

46. Gordon, A.S., et al., Quantifying rare, deleterious variation in 12 human cytochrome P450 drug-metabolism genes in a large-scale exome dataset. Hum Mol Genet, 2014. 23(8): p. 1957-63.

47. Zhou, Y., et al., A paradigm shift in pharmacogenomics: From candidate polymorphisms to comprehensive sequencing. Basic Clin Pharmacol Toxicol, 2022. 131(6): p. 452-464.

48. Ng, P.C. and S. Henikoff, Predicting deleterious amino acid substitutions. Genome Res, 2001. 11(5): p. 863-74.

49. Adzhubei, I.A., et al., A method and server for predicting damaging missense mutations. Nat Methods, 2010. 7(4): p. 248-9.

50. Peterson, T.A., E. Doughty, and M.G. Kann, Towards precision medicine: advances in computational approaches for the analysis of human variants. J Mol Biol, 2013. 425(21): p. 4047-63.

51. Pandi, M.T., et al., A novel machine learning-based approach for the computational functional assessment of pharmacogenomic variants. Hum Genomics, 2021. 15(1): p. 51.

52. Shrestha, S., et al., Gene-Specific Variant Classifier (DPYD-Varifier) to Identify Deleterious Alleles of Dihydropyrimidine Dehydrogenase. Clin Pharmacol Ther, 2018. 104(4): p. 709-718.

53. van der Lee, M., et al., Toward predicting CYP2D6-mediated variable drug response from CYP2D6 gene sequencing data. Sci Transl Med, 2021. 13(603).

54. Sadee, W., The relevance of "missing heritability" in pharmacogenomics. Clin Pharmacol Ther, 2012. 92(4): p. 428-30.

55. Simona, A., et al., Polygenic risk scores in pharmacogenomics: opportunities and challenges-a mini review. Front Genet, 2023. 14: p. 1217049.

56. Pirmohamed, M., Pharmacogenomics: current status and future perspectives. Nat Rev Genet, 2023. 24(6): p. 350-362.

57. Kumuthini, J., et al., The clinical utility of polygenic risk scores in genomic medicine practices: a systematic review. Hum Genet, 2022. 141(11): p. 1697-1704.

58. Schwarz, U.I., M. Gulilat, and R.B. Kim, The Role of Next-Generation Sequencing in Pharmacogenetics and Pharmacogenomics. Cold Spring Harb Perspect Med, 2019. 9(2).

59. Cascorbi, I. and M. Schwab, Epigenetics in Drug Response. Clin Pharmacol Ther, 2016. 99(5): p. 468-70.

60. Cascorbi, I., Overlapping effects of genetic variation and epigenetics on drug response: challenges of pharmacoepigenomics. Pharmacogenomics, 2013. 14(15): p. 1807-9.

61. Lotsch, J., et al., Common non-epigenetic drugs as epigenetic modulators. Trends Mol Med, 2013. 19(12): p. 742-53.

62. Karazniewicz-Lada, M., D. Danielak, and F. Glowka, Genetic and non-genetic factors affecting the response to clopidogrel therapy. Expert Opin Pharmacother, 2012. 13(5): p. 663-83.

63. Biswas, M., N. Vanwong, and C. Sukasem, Pharmacogenomics and non-genetic factors affecting drug response in autism spectrum disorder in Thai and other populations: current evidence and future implications. Front Pharmacol, 2023. 14: p. 1285 967.

64. Michal Sadowski, M.T., Joel Mefford, Tanushree Haldar, Akinyemi Oni-Orisan, Richard Border, Ali Pazokitoroudi, Julien F. Ayroles, Sriram Sankararaman, Andy Dahl, Noah Zaitlen, Characterizing the genetic architecture of drug response using gene-context interaction methods. medRxiv, 2024.

65. Neavin, D., R. Kaddurah-Daouk, and R. Weinshilboum, Pharmacometabolomics informs Pharmacogenomics. Metabolomics, 2016. 12(7).

66. Balashova, E.E., D.L. Maslov, and P.G. Lokhov, A Metabolomics Approach to Pharmacotherapy Personalization. J Pers Med, 2018. 8(3).

67. Hertz, D.L., et al., Recommendations for pharmacogenetic testing in clinical practice guidelines in the US. Am J Health Syst Pharm, 2024. 81(16): p. 672-683.

68. Abdullah-Koolmees, H., et al., Pharmacogenetics Guidelines: Overview and Comparison of the DPWG, CPIC, CPNDS, and RNPGx Guidelines. Front Pharmacol, 2020. 11: p. 595219.

69. Cavallari, L.H. and V.M. Pratt, Building Evidence for Clinical Use of Pharmacogenomics and Reimbursement for Testing. Clin Lab Med, 2022. 42(4): p. 533-546.

70. Janssens, A.C. and P.A. Deverka, Useless until proven effective: the clinical utility of preemptive pharmacogenetic testing. Clin Pharmacol Ther, 2014. 96(6): p. 652-4.

71. Luzum, J.A., et al., Moving Pharmacogenetics Into Practice: It's All About the Evidence! Clin Pharmacol Ther, 2021. 110(3): p. 649-661.

72. Gillis, N.K. and F. Innocenti, Evidence required to demonstrate clinical utility of pharmacogenetic testing: the debate continues. Clin Pharmacol Ther, 2014. 96(6): p. 655-7.

73. Rogers, S.L., et al., A collaborative force for precision medicine progress: the STRIPE pharmacogenomics conference series. Pharmacogenomics J, 2024. 24(5): p. 27.

74. Maeda, H., et al., Assessment of Surrogate End Point Trends in Clinical Trials to Approve Oncology Drugs From 2001 to 2020 in Japan. JAMA Netw Open, 2023. 6(4): p. e238875.

75. Blumenthal, G.M., et al., Oncology Drug Approvals: Evaluating Endpoints and Evidence in an Era of Breakthrough Therapies. Oncologist, 2017. 22(7): p. 762-767.

76. Duarte, J.D., et al., Multisite investigation of strategies for the clinical implementation of pre-emptive pharmacogenetic testing. Genet Med, 2021. 23(12): p. 2335-2341.

77. Patel, J.N., et al., North Carolina's multi-institutional pharmacogenomics efforts with the North Carolina Precision Health Collaborative. Pharmacogenomics, 2021. 22(2): p. 73-80.

78. All of Us Research Program Genomics, I., Genomic data in the All of Us Research Program. Nature, 2024. 627(8003): p. 340-346.

79. McInnes, G., et al., Pharmacogenetics at Scale: An Analysis of the UK Biobank. Clin Pharmacol Ther, 2021. 109(6): p. 1528-1537.

80. Fan, J. and H. Liu, Statistical analysis of big data on pharmacogenomics. Adv Drug Deliv Rev, 2013. 65(7): p. 987-1000.

81. Barrot, C.C., J.B. Woillard, and N. Picard, Big data in pharmacogenomics: current applications, perspectives and pitfalls. Pharmacogenomics, 2019. 20(8): p. 609-620.

82. Prosperi, M., et al., Big data hurdles in precision medicine and precision public health. BMC Med Inform Decis Mak, 2018. 18(1): p. 139.

83. Milano, M., G. Agapito, and M. Cannataro, An Exploratory Application of Multilayer Networks and Pathway Analysis in Pharmacogenomics. Genes (Basel), 2023. 14(10).

84. Poo, D.C., S. Cai, and J.T. Mah, UASIS: Universal Automatic SNP Identification System. BMC Genomics, 2011. 12 Suppl 3(Suppl 3): p. S9.

85. Richards, S., et al., Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med, 2015. 17(5): p. 405-24.

86. Kim, K., et al., Clinical Utility of Pharmacogenetic Testing and a Clinical Decision Support Tool to Enhance the Identification of Drug Therapy Problems Through Medication Therapy Management in Polypharmacy Patients. J Manag Care Spec Pharm, 2018. 24(12): p. 1250-1259.

87. Swen, J.J., et al., A 12-gene pharmacogenetic panel to prevent adverse drug reactions: an open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet, 2023. 401(10374): p. 347-356.

88. Bohlen, K.N., et al., Clinical utility of pharmacogenetics in a psychiatric and primary care population. Pharmacogenomics J, 2023. 23(1): p. 21-27.

89. Tiwari, A.K., et al., Clinical utility of combinatorial pharmacogenomic testing in depression: A Canadian patient- and rater-blinded, randomized, controlled trial. Transl Psychiatry, 2022. 12(1): p. 101.

90. Elliott, L.S., et al., Clinical impact of pharmacogenetic profiling with a clinical decision support tool in polypharmacy home health patients: A prospective pilot randomized controlled trial. PLoS One, 2017. 12(2): p. e0170905.

91. Brixner, D., et al., The effect of pharmacogenetic profiling with a clinical decision support tool on healthcare resource utilization and estimated costs in the elderly exposed to polypharmacy. J Med Econ, 2016. 19(3): p. 213-28.

92. Kogan, J.N., et al., Delivering on the value proposition of precision medicine: the view from healthcare payers. Am J Manag Care, 2018. 24(4): p. 177-179.

93. Lazarou, J., B.H. Pomeranz, and P.N. Corey, Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA, 1998. 279(15): p. 1200-5.

94. Pirmohamed, M., et al., Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ, 2004. 329(7456): p. 15-9.

95. Sultana, J., P. Cutroneo, and G. Trifiro, Clinical and economic burden of adverse drug reactions. J Pharmacol Pharmacother, 2013. 4 (Suppl 1): p. S73-7.

96. Wong, W.B., et al., Cost effectiveness of pharmacogenomics: a critical and systematic review. Pharmacoeconomics, 2010. 28(11): p. 1001-13.

97. van der Wouden, C.H., et al., Cost-Effectiveness of Pharmacogenomics-Guided Prescribing to Prevent Gene-Drug-Related Deaths: A Decision-Analytic Model. Front Pharmacol, 2022. 13: p. 918493.

98. Kelley, E.F., et al., Economic evaluation of a pharmacogenomic multi-gene panel test to optimize anti-hypertension therapy: simulation study. J Med Econ, 2018. 21(12): p. 1246-1253.

99. Tanner, J.A., et al., Cost-effectiveness of combinatorial pharmacogenomic testing for depression from the Canadian public payer perspective. Pharmacogenomics, 2020. 21(8): p. 521-531.

100. Limdi, N.A., et al., Cost-effectiveness of CYP2C19-guided antiplatelet therapy in patients with acute coronary syndrome and percutaneous coronary intervention informed by real-world data. Pharmacogenomics J, 2020. 20(5): p. 724-735.

101. Dong, O.M., et al., Cost-Effectiveness of Multigene Pharmacogenetic Testing in Patients With Acute Coronary Syndrome After Percutaneous Coronary Intervention. Value Health, 2020. 23(1): p. 61-73.

102. Jarvis, J.P., et al., Real-World Impact of a Pharmacogenomics-Enriched Comprehensive Medication Management Program. J Pers Med, 2022. 12(3).

103. Van Heukelom, J., et al., Evolution of pharmacogenomic services and implementation of a multi-state pharmacogenomics clinic across a large rural healthcare system. Front Pharmacol, 2023. 14: p. 1274165.

104. Tamara Apted, A.H., Pharmacogenomics for Improved Outcomes and Decreased Costs in Health Care. The American Journal of Managed Care, 2023.

105. Ji, Y. and S. Shaaban, Interrogating Pharmacogenetics Using Next-Generation Sequencing. J Appl Lab Med, 2024. 9(1): p. 50-60.

106. Naidoo, N., et al., Human genetics and genomics a decade after the release of the draft sequence of the human genome. Hum Genomics, 2011. 5(6): p. 577-622.

107. Hu, T., et al., Next-generation sequencing technologies: An overview. Hum Immunol, 2021. 82(11): p. 801-811.

108. Yang, Y., et al., Sequencing the CYP2D6 gene: from variant allele discovery to clinical pharmacogenetic testing. Pharmacogenomics, 2017. 18(7): p. 673-685.

109. Bravo-Egana, V., H. Sanders, and N. Chitnis, New challenges, new opportunities: Next generation sequencing and its place in the advancement of HLA typing. Hum Immunol, 2021. 82(7): p. 478-487.

110. Lee, S.B., et al., Stargazer: a software tool for calling star alleles from next-generation sequencing data using CYP2D6 as a model. Genet Med, 2019. 21(2): p. 361-372.

111. Chen, X., et al., Cyrius: accurate CYP2D6 genotyping using whole-genome sequencing data. Pharmacogenomics J, 2021. 21(2): p. 251-261.

112. Hari, A., et al., An efficient genotyper and star-allele caller for pharmacogenomics. Genome Res, 2023. 33(1): p. 61-70.

113. Yuan, D.Y., et al., A New Cloud-Native Tool for Pharmacogenetic Analysis. Genes (Basel), 2024. 15(3).

114. Twesigomwe, D., et al., StellarPGx: A Nextflow Pipeline for Calling Star Alleles in Cytochrome P450 Genes. Clin Pharmacol Ther, 2021. 110(3): p. 741-749.

115. Twesigomwe, D., et al., A systematic comparison of pharmacogene star allele calling bioinformatics algorithms: a focus on CYP2D6 genotyping. NPJ Genom Med, 2020. 5: p. 30.

116. Graansma, L.J., et al., From gene to dose: Long-read sequencing and *-allele tools to refine phenotype predictions of CYP2C19. Front Pharmacol, 2023. 14: p. 1076574.

117. van der Lee, M., et al., Application of long-read sequencing to elucidate complex pharmacogenomic regions: a proof of principle. Pharmacogenomics J, 2022. 22(1): p. 75-81.

118. Wohlers, I., S. Garg, and J.Y. Hehir-Kwa, Editorial: Long-read sequencing-Pitfalls, benefits and success stories. Front Genet, 2022. 13: p. 1114542.

119. Hassan, M., et al., Innovations in Genomics and Big Data Analytics for Personalized Medicine and Health Care: A Review. Int J Mol Sci, 2022. 23(9).

120. Suthaharan, S., Machine Learning Models and Algorithms for Big Data Classification : Thinking with Examples for Effective Learning, in Integrated Series in Information Systems,. 2016, Springer US : Imprint: Springer,: New York, NY. p. 1 online resource (XIX, 359 pages 149 illustrations, 82 illustrations in color.

121. Tai, A.M.Y., et al., Machine learning and big data: Implications for disease modeling and therapeutic discovery in psychiatry. Artif Intell Med, 2019. 99: p. 101704.

122. Bzdok, D. and A. Meyer-Lindenberg, Machine Learning for Precision Psychiatry: Opportunities and Challenges. Biol Psychiatry Cogn Neurosci Neuroimaging, 2018. 3(3): p. 223-230.

123. Costello, J.C., et al., A community effort to assess and improve drug sensitivity prediction algorithms. Nat Biotechnol, 2014. 32(12): p. 1202-12.

124. Sakellaropoulos, T., et al., A Deep Learning Framework for Predicting Response to Therapy in Cancer. Cell Rep, 2019. 29(11): p. 3367-3373 e4.

125. Lin, E., C.H. Lin, and H.Y. Lane, Machine Learning and Deep Learning for the Pharmacogenomics of Antidepressant Treatments. Clin Psychopharmacol Neurosci, 2021. 19(4): p. 577-588.

126. Chenoweth, M.J., et al., Global Pharmacogenomics Within Precision Medicine: Challenges and Opportunities. Clin Pharmacol Ther, 2020. 107(1): p. 57-61.

127. El Shamieh, S. and N.K. Zgheib, Pharmacogenetics in developing countries and low resource environments. Hum Genet, 2022. 141(6): p. 1159-1164.

128. Abou Diwan, E., et al., Implementation and obstacles of pharmacogenetics in clinical practice: An international survey. Br J Clin Pharmacol, 2019. 85(9): p. 2076-2088.

129. Mitropoulos, K., et al., Relevance of pharmacogenomics for developing countries in Europe. Drug Metabol Drug Interact, 2011. 26(4): p. 143-6.

130. Zhou, Y. and V.M. Lauschke, Population pharmacogenomics: an update on ethnogeographic differences and opportunities for precision public health. Hum Genet, 2022. 141(6): p. 1113-1136.

131. Goljan, E., et al., Identification of pharmacogenetic variants from large scale next generation sequencing data in the Saudi population. PLoS One, 2022. 17(1): p. e0263137.

132. Volpi, S., et al., Research Directions in the Clinical Implementation of Pharmacogenomics: An Overview of US Programs and Projects. Clin Pharmacol Ther, 2018. 103(5): p. 778-786.

133. Robarge, J.D., et al., The star-allele nomenclature: retooling for translational genomics. Clin Pharmacol Ther, 2007. 82(3): p. 244-8.

134. Godoy Torso, N., P.C. Jl Santos, and P. Moriel, Challenges for the application of pharmacogenomics associated with the nomenclature of allelic variants. Pharmacogenomics, 2023. 24(15): p. 793-796.

135. Nebert, D.W., Suggestions for the nomenclature of human alleles: relevance to ecogenetics, pharmacogenetics and molecular epidemiology. Pharmacogenetics, 2000. 10(4): p. 279-90.

136. ClinPGx. 2024 [cited 2024 10/04/2024]; Available from: https://clinpgx.org/.

137. Koutsilieri, S., et al., Unveiling the guidance heterogeneity for genome-informed drug treatment interventions among regulatory bodies and research consortia. Pharmacol Res, 2020. 153: p. 104590.

138. Pratt, V.M., et al., Recommendations for Clinical CYP2D6 Genotyping Allele Selection: A Joint Consensus Recommendation of the Association for Molecular Pathology, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, and the European Society for Pharmacogenomics and Personalized Therapy. J Mol Diagn, 2021. 23(9): p. 1047-1064.

139. Pratt, V.M., et al., CYP3A4 and CYP3A5 Genotyping Recommendations: A Joint Consensus Recommendation of the Association for Molecular Pathology, Clinical Pharmacogenetics Implementation Consortium, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, European Society for Pharmacogenomics and Personalized Therapy, and Pharmacogenomics Knowledgebase. J Mol Diagn, 2023. 25(9): p. 619-629.

140. Pratt, V.M., et al., TPMT and NUDT15 Genotyping Recommendations: A Joint Consensus Recommendation of the Association for Molecular Pathology, Clinical Pharmacogenetics Implementation Consortium, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, European Society for Pharmacogenomics and Personalized Therapy, and Pharmacogenomics Knowledgebase. J Mol Diagn, 2022. 24(10): p. 1051-1063.

141. Hoffman, J.M., et al., PG4KDS: a model for the clinical implementation of pre-emptive pharmacogenetics. Am J Med Genet C Semin Med Genet, 2014. 166C(1): p. 45-55.

142. Schildcrout, J.S., et al., Optimizing drug outcomes through pharmacogenetics: a case for preemptive genotyping. Clin Pharmacol Ther, 2012. 92(2): p. 235-42.

143. Keeling, N.J., et al., Preemptive pharmacogenetic testing: exploring the knowledge and perspectives of US payers. Genet Med, 2019. 21(5): p. 1224-1232.

144. Weitzel, K.W., L.H. Cavallari, and L.J. Lesko, Preemptive Panel-Based Pharmacogenetic Testing: The Time is Now. Pharm Res, 2017. 34(8): p. 1551-1555.

145. Klein, M.D., et al., Clinical Utility of CYP2C19 Genotyping to Guide Antiplatelet Therapy in Patients With an Acute Coronary Syndrome or Undergoing Percutaneous Coronary Intervention. Arterioscler Thromb Vasc Biol, 2019. 39(4): p. 647-652.

146. Rajan, A. and H. Glorikian, Point-of-care diagnostics: market trends and growth drivers. Expert Opin Med Diagn, 2009. 3(1): p. 1-4.

147. Pirmohamed, M., et al., A randomized trial of genotype-guided dosing of warfarin. N Engl J Med, 2013. 369(24): p. 2294-303.

148. Franke, C.A., L.M. Dickerson, and P.J. Carek, Improving anticoagulation therapy using point-of-care testing and a standardized protocol. Ann Fam Med, 2008. 6 Suppl 1(Suppl 1): p. S28-32.

149. Rasmussen-Torvik, L.J., et al., Design and anticipated outcomes of the eMERGE-PGx project: a multicenter pilot for preemptive pharmacogenomics in electronic health record systems. Clin Pharmacol Ther, 2014. 96(4): p. 482-9.

150. Gottesman, O., et al., The CLIPMERGE PGx Program: clinical implementation of personalized medicine through electronic health records and genomics-pharmacogenomics. Clin Pharmacol Ther, 2013. 94(2): p. 214-7.

151. Haga, S.B., et al., Primary care physicians' knowledge of and experience with pharmacogenetic testing. Clin Genet, 2012. 82(4): p. 388-94.

152. Blagec, K., et al., Pharmacogenomics decision support in the U-PGx project: Results and advice from clinical implementation across seven European countries. PLoS One, 2022. 17(6): p. e0268534.

153. Massmann, A., et al., Evaluation of pharmacogenetic automated clinical decision support for clopidogrel. Pharmacogenomics, 2024: p. 1-9.

154. Jacob, V., et al., Cost and economic benefit of clinical decision support systems for cardiovascular disease prevention: a community guide systematic review. J Am Med Inform Assoc, 2017. 24(3): p. 669-676.

155. Lewkowicz, D., A. Wohlbrandt, and E. Boettinger, Economic impact of clinical decision support interventions based on electronic health records. BMC Health Serv Res, 2020. 20(1): p. 871.

156. Bright, T.J., et al., Effect of clinical decision-support systems: a systematic review. Ann Intern Med, 2012. 157(1): p. 29-43.

157. Chen, Z., et al., Harnessing the power of clinical decision support systems: challenges and opportunities. Open Heart, 2023. 10(2).

158. Haga, S.B., G. Tindall, and J.M. O'Daniel, Professional perspectives about pharmacogenetic testing and managing ancillary findings. Genet Test Mol Biomarkers, 2012. 16(1): p. 21-4.

159. Guy, J.W., I. Patel, and J.H. Oestreich, Clinical Application and Educational Training for Pharmacogenomics. Pharmacy (Basel), 2020. 8(3).

160. Haga, S.B., The Critical Role of Pharmacists in the Clinical Delivery of Pharmacogenetics in the U.S. Pharmacy (Basel), 2023. 11(5).

161. Dressler, L.G., et al., Implementing pharmacogenetic testing in rural primary care practices: a pilot feasibility study. Pharmacogenomics, 2019. 20(6): p. 433-446.

162. Hockings, J.K., et al., Pharmacogenomics: An evolving clinical tool for precision medicine. Cleve Clin J Med, 2020. 87(2): p. 91-99.

163. Lu, C.Y., et al., Insurance Coverage Policies for Pharmacogenomic and Multi-Gene Testing for Cancer. J Pers Med, 2018. 8(2).

164. Park, S.K., J. Thigpen, and I.J. Lee, Coverage of pharmacogenetic tests by private health insurance companies. J Am Pharm Assoc (2003), 2020. 60(2): p. 352-356 e3.

165. Anderson, H.D., et al., The landscape of pharmacogenetic testing in a US managed care population. Genet Med, 2020. 22(7): p. 1247-1253.

166. Empey, P.E., et al., Expanding evidence leads to new pharmacogenomics payer coverage. Genet Med, 2021. 23(5): p. 830-832.

167. Local Coverage Determination (LCD). Pharmacogenomics Testing. [cited 2024 10/15/2024]; Available from: https://www.cms.gov/medicare-coverage-database/view/lcd.aspx?lcdId=39063.

168. Rogers, S.L., et al., Conference report: inaugural Pharmacogenomics Access & Reimbursement Symposium. Pharmacogenomics J, 2021. 21(5): p. 622-624.

169. Chapman, C.R., et al., Genetic discrimination: emerging ethical challenges in the context of advancing technology. J Law Biosci, 2020. 7(1): p. lsz016.

170. Seaver, L.H., et al., Points to consider to avoid unfair discrimination and the misuse of genetic information: A statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med, 2022. 24(3): p. 512-520.

171. Muto, K., et al., Is legislation to prevent genetic discrimination necessary in Japan? An overview of the current policies and public attitudes. J Hum Genet, 2023. 68(9): p. 579-585.

172. Joly, Y., et al., Looking Beyond GINA: Policy Approaches to Address Genetic Discrimination. Annu Rev Genomics Hum Genet, 2020. 21: p. 491-507.

173. Wauters, A. and I. Van Hoyweghen, Global trends on fears and concerns of genetic discrimination: a systematic literature review. J Hum Genet, 2016. 61(4): p. 275-82.

174. Wertz, D.C., Ethical, social and legal issues in pharmacogenomics. Pharmacogenomics J, 2003. 3(4): p. 194-6.

175. Clayton, E.W., et al., The law of genetic privacy: applications, implications, and limitations. J Law Biosci, 2019. 6(1): p. 1-36.

176. Lucassen, A. and R. Gilbar, Alerting relatives about heritable risks: the limits of confidentiality. BMJ, 2018. 361: p. k1409.

177. Rego, S., et al., Informed Consent in the Genomics Era. Cold Spring Harb Perspect Med, 2020. 10(8).

178. Thaldar, D., The wisdom of claiming ownership of human genomic data: A cautionary tale for research institutions. Dev World Bioeth, 2024.

179. Rahnasto, J., Genetic data are not always personal-disaggregating the identifiability and sensitivity of genetic data. J Law Biosci, 2023. 10(2): p. lsad029.

180. Thaldar, D.W., et al., The multidimensional legal nature of personal genomic sequence data: A South African perspective. Front Genet, 2022. 13: p. 997595.

181. Andanda, P.A., Human-tissue-related inventions: ownership and intellectual property rights in international collaborative research in developing countries. J Med Ethics, 2008. 34(3): p. 171-9.

182. Henrikson, N.B., W. Burke, and D.L. Veenstra, Ancillary risk information and pharmacogenetic tests: social and policy implications. Pharmacogenomics J, 2008. 8(2): p. 85-9.

183. Westbrook, M.J., et al., Mapping the incidentalome: estimating incidental findings generated through clinical pharmacogenomics testing. Genet Med, 2013. 15(5): p. 325-31.

184. Haga, S.B., Revisiting Secondary Information Related to Pharmacogenetic Testing. Front Genet, 2021. 12: p. 741395.

185. Brothers, K.B., et al., Eliciting preferences on secondary findings: the Preferences Instrument for Genomic Secondary Results. Genet Med, 2017. 19(3): p. 337-344.

186. Chao, E.C., et al., Incidental detection of acquired variants in germline genetic and genomic testing: a points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med, 2021. 23(7): p. 1179-1184.

187. Shaaban, S. and Y. Ji, Pharmacogenomics and health disparities, are we helping? Front Genet, 2023. 14: p. 1099541.

188. Lee, H., et al., The concepts of health inequality, disparities and equity in the era of population health. Appl Nurs Res, 2020. 56: p. 151 367.

189. Cohn, E.G., G.E. Henderson, and P.S. Appelbaum, Distributive justice, diversity, and inclusion in precision medicine: what will success look like? Genet Med, 2017. 19(2): p. 157-159.

190. Roman, Y.M., et al., Challenges in pharmacotherapy for older adults: a framework for pharmacogenomics implementation. Pharmacogenomics, 2020. 21(9): p. 627-635.

191. Richman, L., et al., Addressing health inequalities in diverse, rural communities: An unmet need. SSM Popul Health, 2019. 7: p. 100398.

192. Neyro, V., E. Jacqz-Aigrain, and T. Adam de Beaumais, Pharmacogenetics and application in pediatrics. Therapie, 2018. 73(2): p. 157-163.

193. Dandara, C., A. Ndadza, and N. Soko, The importance of including African populations in pharmacogenetics studies of warfarin response. Pharmacogenomics, 2022. 23(1): p. 1-4.