Journal of Pharmacokinetics & Experimental Therapeutics
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • J Pharmacokinet Exp Ther, Vol 8(6)

Personalized Drug Treatment: The Future of Medicine Through Pharmacogenomics

Hina Shahid*
Department of Microbiology, University of Health Sciences, Pakistan
*Corresponding Author: Hina Shahid, Department of Microbiology, University of Health Sciences, Pakistan, Email: hina684@gmail.com

Received: 02-Dec-2024 / Manuscript No. jpet-25-160244 / Editor assigned: 06-Dec-2024 / PreQC No. jpet-25-160244 / Reviewed: 20-Dec-2024 / QC No. jpet-25-160244 / Revised: 27-Dec-2024 / Manuscript No. jpet-25-160244 / Published Date: 31-Dec-2024

Introduction

Pharmacogenomics is an interdisciplinary field that combines pharmacology and genomics to study how an individual’s genetic makeup affects their response to drugs. By understanding the genetic factors that influence drug metabolism, efficacy, and safety, pharmacogenomics aims to provide personalized medicine, optimizing drug therapy and minimizing adverse drug reactions (ADRs). Unlike traditional pharmacology, which uses a one-size-fits-all approach to drug prescriptions, pharmacogenomics recognizes that people are genetically diverse, and this genetic diversity plays a significant role in determining how they respond to treatments. Human genetic variation can influence various aspects of drug action, including how drugs are absorbed, distributed, metabolized, and eliminated by the body. For instance, variations in genes that code for drug-metabolizing enzymes, such as the CYP450 enzyme family, can cause patients to metabolize drugs at different rates, leading to either therapeutic failure or toxicity [1]. Additionally, genetic differences in drug receptors can make individuals more or less sensitive to certain medications.

Methodology

The methodology of pharmacogenomics involves a series of approaches to identify and analyze the genetic variations that influence an individual's response to medications. The process typically involves genetic testing, data analysis, and the application of findings to clinical decision-making. Key methods include:

Genetic testing: The first step in pharmacogenomic analysis involves obtaining a biological sample (commonly blood, saliva, or cheek swabs) from the patient. These samples are analyzed to identify genetic variations that can impact drug metabolism, efficacy, and safety. Common genetic variations of interest include single nucleotide polymorphisms (SNPs) in genes encoding drug-metabolizing enzymes (e.g., CYP450 enzymes), drug receptors, and transporters [2]. For example, testing for variations in the CYP2C9 or VKORC1 genes can help guide the dosing of warfarin, an anticoagulant.

Pharmacogenomic databases: Once genetic variants are identified, the data is compared to pharmacogenomic databases such as the PharmGKB (Pharmacogenomics Knowledge Base) or ClinVar to understand the clinical significance of these variations [3]. These databases provide information on how specific genetic variants impact drug response, offering insights into dosing recommendations, potential drug interactions, and adverse effects.

Data analysis and interpretation: Bioinformatics tools and algorithms are used to analyze the genetic data and interpret how the identified variations will affect drug response. This may involve assessing the patient’s genotype (the specific genetic sequence) against established pharmacogenomic guidelines, helping healthcare providers make evidence-based decisions about drug selection and dosing [4, 5].

Clinical application: The final step involves using the pharmacogenomic information to tailor drug therapy for the individual. This can include adjusting the dosage, choosing alternative drugs, or preventing potential adverse drug reactions. Pharmacogenomic testing is especially important for drugs with a narrow therapeutic index or those known to have significant variability in response across different genetic profiles, such as chemotherapy drugs, anticoagulants, and psychotropic medications.

Key applications of pharmacogenomics

Personalized Drug Selection: By analyzing a patient's genetic profile, healthcare providers can select drugs that are more likely to be effective for that individual. This helps avoid the trial-and-error approach that is often used in prescribing medications. For example, pharmacogenomic testing can determine which antidepressant is likely to work best based on the patient's genetic predisposition to metabolize certain drugs [6].

Optimizing drug dosing: Genetic variations can affect how quickly or slowly a drug is metabolized, which influences its concentration in the bloodstream. Pharmacogenomics can help determine the optimal dose for a patient, reducing the risk of side effects or therapeutic failure. For instance, the dosing of warfarin, an anticoagulant, is strongly influenced by genetic variations in the CYP2C9 and VKORC1 genes [7, 8]. Testing for these variants helps tailor the right warfarin dose, minimizing the risk of bleeding or clotting.

Reducing adverse drug reactions (ADRs): Adverse drug reactions are a significant concern in healthcare and often result in hospitalizations, morbidity, and even death. By identifying genetic factors that increase the risk of ADRs, pharmacogenomics can help prevent these reactions before they occur. For example, individuals with a variation in the HLA-B*1502 allele are at a higher risk for severe skin reactions when treated with carbamazepine, an anticonvulsant. Genetic testing before prescribing the drug can prevent these life-threatening reactions.

Improving drug efficacy: Pharmacogenomics also plays a role in increasing the effectiveness of drugs. Some medications work well for certain individuals but fail to have the desired effect in others due to genetic variations. For example, certain cancer treatments, such as trastuzumab (Herceptin), are effective only in patients whose tumors overexpress the HER2 protein, which can be identified through genetic testing. This ensures that only patients who are likely to benefit from the treatment receive it [9].

Future of pharmacogenomics

Pharmacogenomics holds immense potential to revolutionize healthcare by enabling personalized medicine that tailors drug treatment to an individual’s genetic profile. As the field advances, more genetic variations linked to drug responses will be identified, leading to more targeted therapies and a deeper understanding of how genetics influence disease. Furthermore, with the decreasing cost of genomic sequencing and the increasing availability of pharmacogenomic tests, personalized medicine will become more accessible to patients worldwide.

In the future, pharmacogenomics could become a routine part of clinical practice, allowing for the prevention of adverse drug reactions, improved efficacy of treatments, and reduced healthcare costs by ensuring patients receive the most appropriate therapy from the start [10]. The integration of pharmacogenomics into clinical decision-making represents a significant leap toward more precise and effective healthcare.

Conclusion

Pharmacogenomics is transforming the way drugs are prescribed, improving the precision and safety of medical treatments. By understanding how genetic variations influence drug metabolism and response, pharmacogenomics enables the development of personalized therapies tailored to individual needs. Although challenges remain in terms of accessibility and cost, the potential benefits of pharmacogenomics are profound, offering a future where drug therapy is optimized for every patient based on their unique genetic profile.

References

  1. Miranda Furtado CL, Silva Santos RD, Furtado GP (2019) Epidrugs: targeting epigenetic marks in cancer treatment. Epigenetics 14:1164-1176.
  2. Indexed at, Google Scholar, Crossref

  3. Currie GM (2018) Pharmacology, Part 2: Introduction to Pharmacokinetics  J Nucl Med Technol 46-3:221-230.
  4. Indexed at, Google Scholar, Crossref

  5. Whirl-Carrillo M, Mc-Donagh EM, Hebert JM, Gong L, Sangkuhl K, et al. (2012) Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther 92:414-417.
  6. Indexed at, Google Scholar, Crossref

  7. Tao D, Wang Y, Bao XQ, Yang BB, Gao F, et al. (2019) Discovery of coumarin Mannich base derivatives as multifunctional agents against monoamine oxidase B and neuroinflammation for the treatment of Parkinson's disease. Eur J Med Chem 173:203-212.
  8. Indexed at, Google Scholar, Crossref

  9. Johnson P, Loganathan C, Iruthayaraj A, Poomani K, Thayumanavan P (2018) S-allyl cysteine as potent anti-gout drug: insight into the xanthine oxidase inhibition and anti-inflammatory activity. Biochimie 154:1-9.
  10. Indexed at, Google Scholar, Crossref

  11. Zhang HF, Li ZH, Liu JY, Liu TT, Wang P, et al. (2016) Correlation of cytochrome P450 oxidoreductase expression with the expression of 10 isoforms of cytochrome P450 in human liver. Drug Metab Dispos 44:1193-1200.
  12. Indexed at, Google Scholar, Crossref

  13. Stone NR, Bicanic T, Salim R, Hope W (2016) Liposomal Amphotericin B (AmBisome (®)): A Review of the Pharmacokinetics, Pharmacodynamics, Clinical Experience and Future Directions. Drugs 76:485-500.
  14. Indexed at, Google Scholar, Crossref

  15. Roden DM, McLeod HL, Relling MV, Williams MS, Mensah GA, et al. (2019) Pharmacogenomics. Lancet 394:521-532.
  16. Indexed at, Google Scholar, Crossref

  17. Mazerska Z, Mróz A, Pawlowska M, Augustin E (2016) The role of glucuronidation in drug resistance. Pharmacol Ther 159:35-55.
  18. Indexed at, Google Scholar, Crossref

  19. Qi C, Fu J, Zhao H, Xing H, Dong D, et al. (2019) Identification of UGTs and BCRP as potential pharmacokinetic determinants of the natural flavonoid alpinetin. Xenobiotica 49:276-283.
  20. Indexed at, Google Scholar, Crossref

Citation: Hina S (2024) Personalized Drug Treatment: The Future of Medicine Through Pharmacogenomics. J Pharmacokinet Exp Ther 8: 280

Copyright: © 2024 Hina S. 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.

Top