GET THE APP

Assessment of Genetic Variants Associated with Drug Metabolism and Response: Pharmacogenomic Insights into Personalized Medicine Strategies
..

Journal of Bioanalysis & Biomedicine

ISSN: 1948-593X

Open Access

Commentary - (2024) Volume 16, Issue 2

Assessment of Genetic Variants Associated with Drug Metabolism and Response: Pharmacogenomic Insights into Personalized Medicine Strategies

Alexouda Zissis*
*Correspondence: Alexouda Zissis, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece, Email:
Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece

Received: 29-Mar-2024, Manuscript No. jbabm-24-139005; Editor assigned: 01-Apr-2024, Pre QC No. P-139005; Reviewed: 15-Apr-2024, QC No. Q-139005; Revised: 20-Apr-2024, Manuscript No. R-139005; Published: 29-Apr-2024 , DOI: 10.37421/1948-593X.2024.16.436
Citation: Zissis, Alexouda. “Assessment of Genetic Variants Associated with Drug Metabolism and Response: Pharmacogenomic Insights into Personalized Medicine Strategies.” J Bioanal Biomed 16 (2024): 436.
Copyright: © 2024 Zissis A. 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.

Introduction

Pharmacogenomics, the study of how genetic variations influence an individual's response to drugs, has revolutionized the field of personalized medicine. This discipline seeks to tailor medical treatment to the individual characteristics of each patient by integrating genetic information with drug therapy decisions. The primary goal is to enhance drug efficacy, minimize adverse drug reactions, and optimize therapeutic outcomes. Genetic variants, particularly those affecting drug metabolism and response, play a crucial role in this personalized approach [1].

Description

Drug metabolism is primarily mediated by enzymes, many of which are encoded by highly polymorphic genes. The cytochrome P450 (CYP) enzyme family, for instance, is responsible for the metabolism of a large proportion of clinically used drugs. Variants in CYP genes, such as CYP2D6, CYP2C19, and CYP3A4, can significantly alter enzyme activity, leading to variations in drug plasma levels and therapeutic effects [2]. For example, CYP2D6 polymorphisms can categorize individuals into poor, intermediate, extensive, or ultra-rapid metabolizers, influencing their response to drugs like antidepressants, antipsychotics, and opioids. Beyond metabolism, genetic variants can also affect drug transporters, receptors, and targets. Variants in the SLCO1B1 gene, which encodes the OATP1B1 transporter, can impact the pharmacokinetics of statins, leading to differences in drug efficacy and the risk of adverse effects like myopathy. Similarly, polymorphisms in the VKORC1 gene, which encodes the target of warfarin, influence dosing requirements and risk of bleeding complications [3].

Advances in genomic technologies, such as next-generation sequencing and genome-wide association studies, have facilitated the identification of numerous genetic variants associated with drug metabolism and response. These insights have paved the way for pharmacogenetic testing, which involves analyzing specific genes or genetic markers to predict an individual’s response to particular medications [4]. The implementation of pharmacogenetic testing in clinical practice holds the promise of personalized medicine, where drug therapies are tailored to an individual's genetic makeup, thereby improving treatment outcomes and reducing adverse drug reactions. This review aims to provide a comprehensive overview of genetic variants associated with drug metabolism and response. By highlighting key genes and polymorphisms that influence drug efficacy and safety, we seek to underscore the importance of pharmacogenomics in developing personalized medicine strategies. Understanding these genetic variations can guide clinicians in selecting appropriate medications and dosing regimens, ultimately enhancing patient care [5].

Conclusion

The implementation of pharmacogenetic testing in clinical settings offers several benefits. It enhances the precision of prescribing, ensuring that patients receive medications that are more likely to be effective and less likely to cause harm. This is particularly important for drugs with narrow therapeutic windows or significant variability in response among individuals. By reducing trial-and-error prescribing, pharmacogenomics can also improve patient adherence to treatment and overall satisfaction with care. In conclusion, the assessment of genetic variants associated with drug metabolism and response is integral to the advancement of personalized medicine. By leveraging pharmacogenomic insights, healthcare providers can make more informed decisions about drug therapy, ultimately improving patient outcomes and enhancing the quality of care. As research in this field continues to evolve, the integration of pharmacogenomics into routine clinical practice will further refine our ability to deliver personalized, effective, and safe treatments to patients.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Wang, Ching-Ya, Chuang-Wei Wang, Chun-Bing Chen and Wei-Ti Chen, et al. "Pharmacogenomics on the treatment response in patients with psoriasis: An updated review." Int J Mol Sci 24 (2023): 7329.
  2. Google Scholar, Crossref, Indexed at

  3. Ovejero-Benito, Maria C., Ester Munoz-Aceituno, Alejandra Reolid and Miriam Saiz-Rodriguez, et al. "Pharmacogenetics and pharmacogenomics in moderate-to-severe psoriasis." Am J Clin Dermatol 19 (2018): 209-222.
  4. Google Scholar, Crossref, Indexed at

  5. Owczarczyk‐Saczonek, Agnieszka, Witold Owczarek, Agnieszka Osmola‐Mańkowska and Zygmunt Adamski, et al. "Secondary failure of TNF‐α inhibitors in clinical practice." Dermatol Ther 32 (2019): e12760.
  6. Google Scholar, Crossref, Indexed at

  7. Elberdín, Laida, Rosa M. Fernández-Torres, Sabela Paradela and María Mateos, et al. "Biologic therapy for moderate to severe psoriasis. Real-world follow-up of patients who initiated biologic therapy at least 10 years ago." Dermatol Ther 12 (2022): 761-770.
  8. Google Scholar, Crossref, Indexed at

  9. Nassim, David, Ali Alajmi, Abdulhadi Jfri and Kevin Pehr. "Apremilast in dermatology: A review of literature." Dermatol Ther 33 (2020): e14261.
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3099

Journal of Bioanalysis & Biomedicine received 3099 citations as per Google Scholar report

Journal of Bioanalysis & Biomedicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward
https://www.olimpbase.org/1937/