GET THE APP

The Impact of Genetic Variation on Drug Response and Personalized Medicine
..

Journal of Genetics and DNA Research

ISSN: 2684-6039

Open Access

Mini Review - (2024) Volume 8, Issue 2

The Impact of Genetic Variation on Drug Response and Personalized Medicine

Li Wei*
*Correspondence: Li Wei, Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, China, Email:
Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, China

Received: 01-Feb-2024, Manuscript No. jgdr-24-136213; Editor assigned: 02-Feb-2024, Pre QC No. P-136213; Reviewed: 19-Feb-2024, QC No. Q-136213; Revised: 26-Feb-2024, Manuscript No. R-136213; Published: 04-Mar-2024 , DOI: 10.37421/2684-6039.2024.8.198
Citation: Wei, Li. “The Impact of Genetic Variation on Drug Response and Personalized Medicine.” J Genet DNA Res 8 (2024): 198.
Copyright: © 2024 Wei L. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

The impact of genetic variation on drug response is a critical aspect of personalized medicine, aiming to optimize therapeutic outcomes by tailoring treatment regimens to individual genetic profiles. Genetic variability in drug-metabolizing enzymes, drug transporters and drug targets can significantly influence pharmacokinetics and pharmacodynamics, leading to inter-individual variability in drug efficacy, toxicity and adverse reactions. This mini-review provides an overview of the role of genetic variation in drug response and its implications for personalized medicine. We discuss key genetic determinants of drug response, approaches for pharmacogenomic testing, challenges in implementation and future perspectives for integrating genetic information into clinical practice.

Keywords

Genetic variation • Drug response • Personalized medicine • Pharmacogenomics • Pharmacokinetics • Drug metabolism • Drug transporters • Pharmacogenetic testing

Introduction

Personalized medicine represents a paradigm shift in healthcare, moving away from a one-size-fits-all approach to treatment towards tailored interventions that consider individual variability in genetics, environment and lifestyle factors. Central to the realization of personalized medicine is the recognition that genetic variation plays a pivotal role in determining an individual's response to pharmacotherapy. Genetic polymorphisms in drugmetabolizing enzymes, drug transporters and drug targets can profoundly influence drug pharmacokinetics (PK) and pharmacodynamics (PD), leading to inter-individual variability in drug efficacy, safety and tolerability [1].

Pharmacogenomics, the study of how genetic variation affects drug response, holds promise for optimizing therapeutic outcomes, minimizing adverse reactions and reducing healthcare costs. By identifying genetic biomarkers associated with drug response phenotypes, clinicians can stratify patients into subgroups with differential treatment outcomes and tailor pharmacotherapy to maximize efficacy while minimizing toxicity [2]. Furthermore, pharmacogenomic testing enables the prediction of individual drug responses a priori, guiding drug selection, dosing regimens and therapeutic strategies to achieve optimal clinical outcomes.

Literature Review

The impact of genetic variation on drug response is multifaceted, encompassing diverse mechanisms that influence drug PK, PD and therapeutic outcomes. One of the primary determinants of drug response is genetic variability in drug-metabolizing enzymes, which catalyze the biotransformation of drugs into active or inactive metabolites. Polymorphisms in genes encoding phase I enzymes (e.g., cytochrome P450s) and phase II conjugation enzymes (e.g., UDP-glucuronosyltransferases) can alter drug metabolism rates, leading to variability in drug clearance, bioavailability and half-life [3].

In addition to drug metabolism, genetic variation in drug transporters plays a crucial role in modulating drug disposition and tissue distribution. Membranebound transporters, such as ATP-binding cassette (ABC) transporters and solute carrier (SLC) transporters, mediate the uptake, efflux and distribution of drugs across cellular membranes. Polymorphisms in transporter genes can affect drug absorption, distribution and excretion, influencing systemic drug exposure and tissue-specific drug concentrations [4]. For example, genetic variants in the ABCB1 gene encoding P-glycoprotein have been associated with altered drug bioavailability and central nervous system penetration, impacting the efficacy and toxicity of various drugs, including anticancer agents and psychotropic medications.

Discussion

Moreover, genetic variation can also affect drug target interactions, leading to variability in drug efficacy and therapeutic responses. Single nucleotide polymorphisms (SNPs) and structural variants in genes encoding drug targets (e.g., receptors, enzymes) can alter protein structure, function and drug binding affinity, influencing the pharmacological effects of drugs. For instance, genetic variants in the β1-adrenergic receptor gene (ADRB1) have been associated with differential responses to β-blockers in the treatment of cardiovascular diseases, with certain variants conferring increased susceptibility to adverse cardiovascular events or reduced therapeutic efficacy [5].

Despite the promise of pharmacogenomics for personalized medicine, several challenges remain in translating genetic information into clinical practice. One of the main challenges is the complexity and multifactorial nature of drug response phenotypes, which are influenced by genetic, environmental and clinical factors. Identifying clinically actionable genetic variants with robust associations with drug response requires large-scale, well-powered studies, as well as validation in diverse patient populations [6]. Moreover, integrating pharmacogenomic information into clinical decision-making poses logistical challenges, including the need for standardized testing platforms, interoperable electronic health record systems and guidelines for interpretation and implementation in clinical practice.

Conclusion

In conclusion, the impact of genetic variation on drug response represents a cornerstone of personalized medicine, offering opportunities to optimize therapeutic outcomes and minimize adverse reactions through tailored pharmacotherapy. Pharmacogenomic testing holds promise for guiding drug selection, dosing optimization and treatment strategies based on individual genetic profiles. However, the implementation of pharmacogenomics in clinical practice requires overcoming various challenges, including the identification of clinically actionable genetic variants, validation in diverse populations and integration into existing healthcare systems. Despite these challenges, pharmacogenomics represents a transformative approach to individualizing drug therapy and improving patient care in the era of precision medicine.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Chen, Thomas, Peter H. O’Donnell, Merisa Middlestadt and Gregory W. Ruhnke, et al. "Implementation of pharmacogenomics into inpatient general medicine."Pharmacogenetics Genom 33 (2023): 19-23.

    Google Scholar, Crossref, Indexed at

  2. Zhou, Yitian, Lenka Nevosadová, Erik Eliasson and Volker M. Lauschke. "Global distribution of functionally important CYP2C9 alleles and their inferred metabolic consequences."Hum Genom 17 (2023): 15.

    Google Scholar, Crossref, Indexed at

  3. Al-Metwali, Basma Zuheir, Peter Rivers and Larry Goodyer, et al. "Personalised warfarin dosing in children post-cardiac surgery."Pediatr Cardiol 40 (2019): 1735-1744.

    Google Scholar, Crossref, Indexed at

  4. Baldwin, R. Michael, Staffan Ohlsson, Rasmus Steen Pedersen and Jessica Mwinyi, et al. "Increased omeprazole metabolism in carriers of the CYP2C19* 17 allele; a pharmacokinetic study in healthy volunteers."Br J Clin Pharmacol 65 (2008): 767-774.

    Google Scholar, Crossref, Indexed at

  5. Udomuksorn, Wandee, David J. Elliot, Benjamin C. Lewis and Peter I. Mackenzie, et al. "Influence of mutations associated with Gilbert and Crigler–Najjar type II syndromes on the glucuronidation kinetics of bilirubin and other UDP-glucuronosyltransferase 1A substrates."Pharmacogenet Genom 17 (2007): 1017-1029.

    Google Scholar, Crossref, Indexed at

  6. Al-Eitan, Laith N., Ayah Y. Almasri and Rame H. Khasawneh. "Impact of CYP2C9 and VKORC1 polymorphisms on warfarin sensitivity and responsiveness in Jordanian cardiovascular patients during the initiation therapy."Genes9 (2018): 578.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3

Journal of Genetics and DNA Research received 3 citations as per Google Scholar report

Journal of Genetics and DNA Research peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward