Pharmacogenomics and Personalized Medicine:
A Revolution in Drug Therapy
Tushar R Chandan1*, Chandrashekhar D. Patil1, Vitthal B Kundgir1, Kavita Chaudhari1
Rushikesh L. Bachhav2, Mayur S. Bhamare2, Sharwari K. Sonawane3, Sunil K. Mahajan4.
1Department of Pharmacology, SSS’s Divine College Pharmacy, Nampur Road, Satana,
Nashik, Maharashtra, India - 423301.
2Department of Quality Assurance, SSS’s Divine College Pharmacy, Nampur Road, Satana,
Nashik, Maharashtra, India - 423301.
3Department of Pharmaceutics, SSS’s Divine College Pharmacy, Nampur Road, Satana,
Nashik, Maharashtra, India - 423301.
4Department of Chemistry, SSS’s Divine College Pharmacy, Nampur Road, Satana,
Nashik, Maharashtra, India - 423301.
*Corresponding Author E-mail: tusharchandan510@gmail.com
ABSTRACT:
The field of pharmacogenomics, which examines how an individual's genetic makeup influences their response to medications, is at the forefront of a paradigm shift in modern healthcare. As part of the broader movement toward personalized medicine, pharmacogenomics enables the development and implementation of tailored therapeutic strategies aimed at optimizing efficacy and minimizing adverse drug reactions (ADRs). Traditional "one-size-fits-all" drug therapies often fail to account for interindividual variability in drug metabolism, efficacy, and toxicity factors now known to be significantly influenced by genetic differences. This review explores the scientific foundations, clinical applications, and ongoing challenges of pharmacogenomics in contemporary medical practice. We discuss the most significant pharmacogenetic markers, including polymorphisms in cytochrome P450 enzymes (such as CYP2D6, CYP2C19, and CYP2C9), drug transporter genes (e.g., SLCO1B1), and genes involved in drug targets and immune responses (e.g., HLA-B, VKORC1). Applications of pharmacogenomics are particularly advanced in oncology, cardiology, psychiatry, and infectious diseases, where genetic testing informs the use of drugs like warfarin, clopidogrel, trastuzumab, and abacavir. The integration of genetic data into clinical workflows holds the promise of not only enhancing therapeutic outcomes but also reducing healthcare costs associated with ineffective treatment and adverse effects. Despite these benefits, widespread adoption of pharmacogenomics is hindered by several challenges, including limited clinician awareness, high testing costs, insufficient regulatory frameworks, and ethical concerns related to privacy and data security. Emerging technologies such as next-generation sequencing (NGS), electronic health record (EHR) integration, and artificial intelligence (AI)-driven decision support are expected to accelerate clinical implementation. Ultimately, pharmacogenomics represents a cornerstone of precision medicine and a revolutionary step toward more individualized, effective, and safe drug therapy. This review highlights current evidence, future prospects, and the systemic changes needed to fully realize the potential of pharmacogenomics in routine clinical practice.
KEYWORDS: Pharmacogenomics, Drug Metabolism, CYP450, Genetic Polymorphism, Precision Medicine, Genomic Testing, Drug Efficacy.
Variability in drug response among individuals is a well-documented clinical phenomenon. While one patient may respond favourably to a medication, another might experience no therapeutic benefit or suffer severe adverse reactions1. Traditionally, such variability has been attributed to factors like age, weight, sex, organ function, diet, and co-administered drugs. However, in recent decades, it has become increasingly evident that genetic differences play a crucial role in shaping individual responses to drug therapy. This realization has given rise to the field of pharmacogenomics the study of how an individual’s genetic makeup influences their response to medications. Pharmacogenomics is a foundational pillar of personalized medicine, an emerging approach to healthcare that seeks to customize medical treatment to the individual characteristics of each patient2. Rather than relying on generalized treatment protocols, personalized medicine incorporates genetic, environmental, and lifestyle factors into clinical decision-making. Pharmacogenomics focuses specifically on how genetic polymorphisms particularly those affecting drug-metabolizing enzymes, transporters, receptors, and targets influence pharmacokinetics and pharmacodynamics3.
The integration of pharmacogenomic insights into clinical practice has the potential to revolutionize drug therapy. By identifying genetic variants that affect drug metabolism (e.g., CYP450 enzyme activity), efficacy (e.g., target receptor mutations), or toxicity (e.g., immune system sensitivity), clinicians can make more informed choices about drug selection and dosing. This approach not only improves treatment efficacy but also reduces the incidence of adverse drug reactions (ADRs), a major cause of morbidity, mortality, and healthcare costs worldwide. Several pharmacogenomic applications are already in use. For instance, genetic testing for CYP2C19 polymorphisms can guide antiplatelet therapy with clopidogrel, while HLA-B57:01 screening helps avoid life-threatening hypersensitivity reactions to the antiretroviral drug abacavir. In oncology, tumor genotyping for HER2, EGFR, or KRAS mutations allows for targeted therapy that significantly improves patient outcomes.
This review aims to provide a comprehensive overview of pharmacogenomics and its role in advancing personalized medicine. We explore the underlying genetic mechanisms, key clinical applications, benefits, challenges, and future directions of this rapidly evolving field4.
Pharmacogenomics is the study of how genetic variation influences an individual’s response to drugs, encompassing both pharmacokinetics (PK)how the body affects a drug and pharmacodynamics (PD) how the drug affects the body. These genetic differences can significantly impact drug absorption, distribution, metabolism, excretion (ADME), and interaction with biological targets, thereby determining the efficacy and safety of a given therapy5.
Figure 1: Principles of Pharmacogenomics
One of the most well-established areas of pharmacogenomics involves genetic polymorphisms in drug-metabolizing enzymes, particularly the cytochrome P450 (CYP450) family. These enzymes are responsible for the oxidative metabolism of approximately 75% of all prescription drugs.
Key polymorphic enzymes include:
· CYP2D6: Responsible for the metabolism of many antidepressants, opioids, and beta-blockers. Genetic variants result in poor, intermediate, extensive, or ultra-rapid metabolizer phenotypes.
· CYP2C9 and CYP2C19: Variants affect metabolism of warfarin, phenytoin, and proton pump inhibitors.
· CYP3A4 and CYP3A5: Influence the metabolism of a broad range of drugs including immunosuppressants and statins.
Individuals with reduced enzyme activity may experience drug accumulation and toxicity, while ultra-rapid metabolizers may have subtherapeutic levels, resulting in treatment failure6.
Genetic variants in drug transporter proteins also play a critical role in drug bioavailability and tissue distribution.
· ABCB1 (MDR1): Encodes P-glycoprotein, a transporter involved in drug efflux across the blood-brain barrier and gastrointestinal tract. Variations can affect drug absorption and central nervous system penetration.
· SLCO1B1: Encodes a liver-specific uptake transporter that influences the hepatic clearance of statins. Certain variants are associated with an increased risk of statin-induced myopathy7.
Beyond metabolism and transport, genetic polymorphisms in drug targets such as receptors, enzymes, and ion channels - can alter drug sensitivity and therapeutic outcomes.
Examples include:
· VKORC1: Variants affect the sensitivity to warfarin by altering the target enzyme of vitamin K antagonism.
· HER2, EGFR, KRAS, BRAF: Somatic mutations in these genes in cancer cells guide the use of targeted therapies in oncology.
· HLA alleles: Variants such as HLA-B57:01 and HLA-B15:02 are associated with severe hypersensitivity reactions to abacavir and carbamazepine, respectively8.
The clinical utility of pharmacogenomics depends on translating genotypic data into actionable phenotypes. This is often expressed as a "metabolizer status" (e.g., poor vs. ultra-rapid metabolizer) or as risk categories for drug toxicity. Guidelines from organizations such as the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) provide evidence-based recommendations for modifying drug selection and dosing based on genotype9.
· Single nucleotide polymorphisms (SNPs): The most common type of variation; often responsible for altered protein function or expression.
· Insertions/deletions (indels): Can lead to frameshift mutations or altered protein structure.
· Copy number variations (CNVs): Duplication or deletion of entire gene regions, such as multiple copies of the CYP2D6 gene.
· Epigenetic modifications: Though not changes in DNA sequence, these can affect gene expression and are emerging areas of interest in pharmacoepigenomics.10
The integration of pharmacogenomics into clinical practice is transforming how medications are prescribed, dosed, and monitored. By identifying genetic variations that affect drug metabolism, efficacy, and safety, clinicians can personalize treatment plans to optimize outcomes and minimize adverse drug reactions (ADRs). The most established and impactful applications are seen in oncology, cardiology, psychiatry, and infectious diseases11.
Figure 2: Clinical Applications of Pharmacogenomics
Cancer therapy has been at the forefront of pharmacogenomics, with genomic profiling guiding targeted treatment strategies. Somatic mutations in tumor DNA, as well as inherited genetic variants, influence drug response and toxicity.
· HER2 and Trastuzumab: HER2-positive breast cancers respond to trastuzumab, a monoclonal antibody targeting the HER2 receptor. HER2 testing is now standard before initiating therapy.
· EGFR and ALK Mutations: In non-small cell lung cancer (NSCLC), mutations in EGFR or rearrangements in ALK predict responsiveness to tyrosine kinase inhibitors (e.g., erlotinib, crizotinib).
· KRAS and BRAF Mutations: Colorectal cancers with KRAS mutations do not respond to anti-EGFR therapies (e.g., cetuximab), while BRAF mutations can guide the use of specific kinase inhibitors.
· TPMT and 6-Mercaptopurine: In acute lymphoblastic leukemia, TPMT polymorphisms affect thiopurine metabolism. Testing prevents life-threatening myelosuppression12.
Pharmacogenomics improves the safety and efficacy of commonly used cardiovascular drugs by informing dosing and drug choice.
· Warfarin: Dosing is influenced by CYP2C9 (metabolism) and VKORC1 (target enzyme). Genotype-guided dosing reduces the risk of bleeding and improves therapeutic control.
· Clopidogrel: This antiplatelet drug requires activation by CYP2C19. Carriers of loss-of-function alleles have reduced drug activity, increasing the risk of thrombotic events. Alternatives such as prasugrel or ticagrelor may be used.
· Statins and SLCO1B1: Variants in SLCO1B1 increase the risk of statin-induced myopathy, especially with simvastatin. Alternative statins or lower doses may be recommended13.
Psychotropic drugs exhibit significant interpatient variability, and pharmacogenomic testing is increasingly used to guide treatment in psychiatry.
· CYP2D6 and CYP2C19: These enzymes metabolize many antidepressants and antipsychotics. Variants can influence plasma drug levels, therapeutic response, and side effect risk. For example, poor metabolizers may require dose reductions or alternative agents.
· HLA-B*15:02: Associated with severe cutaneous adverse reactions (e.g., Stevens-Johnson syndrome) in response to carbamazepine and other anticonvulsants, particularly in individuals of Asian ancestry.
· Serotonin Transporter (SLC6A4) and COMT genes: Although their clinical utility is still being evaluated, these genes may affect response to SSRIs and other antidepressants [14].
Genetic testing helps identify patients at risk of serious ADRs or nonresponse to certain antimicrobial agents.
· Abacavir and HLA-B*57:01: Strongly associated with potentially fatal hypersensitivity reactions to the antiretroviral abacavir. Genetic screening is now standard before initiating treatment.
· IFNL3 (IL28B): Genotype may predict response to interferon-based therapy in hepatitis C, although this is less relevant in the era of direct-acting antivirals.
· CYP2B6: Influences efavirenz metabolism, used in HIV therapy. Poor metabolizers have increased risk of neurotoxicity and may benefit from dose adjustment [15]
Genetic variation influences both the efficacy and risk of opioid analgesics and anesthetic agents.
· CYP2D6 and Codeine: Codeine is a prodrug activated to morphine by CYP2D6. Poor metabolizers experience little pain relief, while ultra-rapid metabolizers are at risk for toxicity due to excessive morphine levels.
· OPRM1 : Polymorphisms may affect opioid sensitivity and dosing requirements 15.
· Transplantation: CYP3A5 polymorphisms affect tacrolimus metabolism. Genotype-guided dosing helps achieve target immunosuppressive levels more quickly.
· Diabetes and Metformin: Variants in the SLC22A1 and ATM genes may influence response to metformin, although clinical guidelines are still evolving.
· Asthma: Pharmacogenomics of beta-agonists and leukotriene inhibitors is under investigation to optimize asthma management15.
Table 1: Key Pharmacogenes and Their Clinical Relevance16
|
Gene |
Function |
Clinical Relevance |
Associated Drugs |
|
CYP2D6 |
Drug metabolism (Phase I) |
Ultra-rapid, extensive, intermediate, or poor metabolism |
Codeine, Tamoxifen, Antidepressants |
|
CYP2C19 |
Drug metabolism (Phase I) |
Alters antiplatelet response |
Clopidogrel, PPIs, SSRIs |
|
CYP2C9 |
Warfarin metabolism |
Affects warfarin sensitivity |
Warfarin, NSAIDs |
|
VKORC1 |
Vitamin k epoxide reductase |
Affects warfarin dosing |
Warfarin |
|
TPMT |
Thiopurine metabolism |
Risk of toxicity in low activity |
Azathioprine, 6-MP |
|
DPYD |
Pyrimidine metabolism |
Risk of severe toxicity |
5-FU, Capecitabine |
|
HLA-B*57:01 |
Immune response |
Hypersensitivity reactions |
Abacavir |
|
SLCO1B1 |
Drug transporter |
Statin-induced myopathy risk |
Simvastatin |
The application of pharmacogenomics in clinical practice represents a transformative shift in how healthcare professionals approach drug therapy. By incorporating genetic information into prescribing decisions, pharmacogenomics enhances the safety, efficacy, and overall efficiency of medical treatment. This section outlines the major benefits and broader impacts of pharmacogenomics on patients, healthcare systems, and public health16.
Figure 3: Benefits and impact pharmacogenomics and personalized medicine: a revolution in drug therapy
One of the most significant advantages of pharmacogenomics is its ability to increase the likelihood that a prescribed medication will be effective for an individual patient. By identifying genetic variants that influence drug targets or metabolic pathways, clinicians can select the most appropriate drug and dosage from the outset, reducing the trial-and-error approach that characterizes much of current prescribing practice.
Example: EGFR mutation testing in non-small cell lung cancer enables clinicians to prescribe tyrosine kinase inhibitors only to patients who are most likely to respond, thereby improving therapeutic outcomes17.
Adverse drug reactions are a major cause of hospitalizations, morbidity, and mortality. Genetic differences in drug metabolism or immune response can predispose certain individuals to severe or life-threatening reactions. Pharmacogenomic testing allows for the identification of at-risk patients before treatment begins.
Example: Screening for HLA-B57:01 prevents abacavir-induced hypersensitivity in HIV patients.
Impact: Reducing ADRs leads to safer prescribing, fewer medical complications, and improved patient adherence to therapy18.
Pharmacogenomics allows for individualized dosing based on metabolic capacity or drug sensitivity. Standard dosing regimens do not account for inter-individual differences in drug clearance or sensitivity, which can result in subtherapeutic effects or toxicity.
Example: Warfarin dosing guided by CYP2C9 and VKORC1 genotypes achieves therapeutic INR levels more quickly and safely than empirical dosing19.
Although genetic testing can incur initial costs, the long-term economic benefits of pharmacogenomics are substantial. By preventing ADRs, reducing hospital admissions, and minimizing ineffective treatments, pharmacogenomics has the potential to significantly reduce healthcare expenditures.
· Healthcare System Impact:
· Fewer emergency visits and hospital stays
· Lower medication waste
· More efficient use of healthcare resources
Several studies have demonstrated cost savings associated with genotype-guided therapy, especially in high-risk populations or with drugs known to have narrow therapeutic windows20.
Patients who receive therapies tailored to their genetic profile often experience fewer side effects and improved therapeutic outcomes, which enhances their overall treatment satisfaction and quality of life. Personalized approaches also foster greater trust in the healthcare system and promote better patient engagement21.
Pharmacogenomics contributes to the design of more efficient and targeted clinical trials by identifying genetic markers associated with drug response. This stratification leads to better-defined study populations and more robust outcomes.
Pharmaceutical Industry Impact:
· Accelerated drug development
· Reduced failure rates in clinical trials
· Expansion of companion diagnostics (e.g., tests for HER2 or KRAS status)
Regulatory bodies such as the U.S. FDA and the EMA have also begun including pharmacogenomic data in drug labeling, providing clinicians with actionable information at the point of care22.
At the population level, pharmacogenomics can be used to develop national or regional drug policies that reflect the genetic diversity of specific populations. This is particularly valuable in multi-ethnic societies where the prevalence of risk alleles can vary significantly between groups.
Example: Implementation of HLA-B15:02 screening programs in Southeast Asia to prevent carbamazepine-induced Stevens-Johnson syndrome22.
Pharmacogenomics lays the groundwork for broader precision medicine initiatives, including integration with:
· Epigenetics
· Microbiomics
· Proteomics
· Artificial intelligence (AI) and machine learning
As the field evolves, pharmacogenomics will play an increasingly central role in data-driven, predictive, and preventive healthcare23.
Despite the growing evidence supporting the clinical utility of pharmacogenomics and its transformative potential in personalized medicine, widespread implementation faces numerous challenges. These limitations span scientific, clinical, economic, ethical, and infrastructural domains. Understanding and addressing these barriers is crucial for the successful integration of pharmacogenomics into routine healthcare24.
Although several pharmacogenomic tests are available and validated, many healthcare systems have been slow to adopt them in routine practice. Reasons include:
· Lack of clinician awareness and education: Many physicians and pharmacists lack formal training in genetics or do not feel confident interpreting genetic test results.
· Unclear clinical guidelines: For many gene-drug pairs, evidence is still emerging, and standardized guidelines are lacking or underutilized.24
· Workflow integration issues: Incorporating genetic testing into busy clinical settings can be complex and time-consuming without dedicated infrastructure.
The cost of pharmacogenomic testing remains a significant barrier, particularly in resource-limited settings. While some tests are covered by insurance, many are not, especially when the clinical benefit is still under investigation.
· High upfront costs: Though long-term benefits may be substantial, healthcare systems are often hesitant to invest in testing without immediate returns.
· Lack of reimbursement policies: Inconsistent insurance coverage and unclear reimbursement pathways discourage both providers and patients from pursuing testing [24].
The use of genetic information raises complex ethical and legal concerns, particularly around privacy, consent, and potential discrimination.
· Genetic data privacy: Patients may be concerned about who has access to their genomic information and how it is stored or shared.
· Informed consent: Ensuring patients fully understand the implications of genetic testing remains a challenge.
· Discrimination risks: Despite legal protections in some countries (e.g., the Genetic Information Non-discrimination Act in the U.S.), fear of discrimination by employers or insurers persists24.
Much of the pharmacogenomic data used to guide clinical decisions has been derived from studies involving primarily individuals of European ancestry. This creates a critical gap in applicability for ethnically diverse populations.
· Underrepresentation in Genomic Research: Populations from Africa, Asia, and Latin America are underrepresented, leading to inaccurate or incomplete predictions of drug response.
· Variability in Allele Frequencies: Important pharmacogenetic variants differ in prevalence across populations, and some may be entirely population-specific.
While many pharmacogenomic markers are well understood, others lack robust evidence or have complex gene-environment interactions that make interpretation difficult.
· Incomplete understanding of gene-drug interactions: Not all clinically relevant variants have been discovered or validated.
· Polygenic influences: Drug response often results from interactions between multiple genes and environmental factors, which are challenging to model and predict.
· Dynamic influences: Epigenetic changes, comorbidities, diet, microbiome composition, and drug-drug interactions can modulate gene expression and complicate genotype-to-phenotype predictions24.
The development of pharmacogenomic tests and their integration into healthcare is hampered by inconsistent regulatory frameworks across regions.
· Lack of global standardization: Guidelines from regulatory bodies (e.g., FDA, EMA, PMDA) may vary, leading to confusion or delays in implementation.
· Approval process: Companion diagnostics often require complex, time-consuming approval processes and must be linked to specific drug labels [24].
Implementing pharmacogenomics at scale requires robust infrastructure, including:
· Electronic health record (EHR) integration: Most healthcare systems are not yet equipped to incorporate genomic data into patient records in real time.
· Clinical decision support systems (CDSS): These tools are needed to help providers interpret genetic results and make informed treatment decisions.
· Data storage and security: Managing large volumes of genomic data requires secure, interoperable systems that protect patient privacy24.
To bridge the gap between genetic science and clinical practice, education at all levels medical students, practicing clinicians, pharmacists, and patients is essential.
· Limited inclusion in medical curricula: Many healthcare professionals are not exposed to pharmacogenomics during their training.
· Continuing education needs: Ongoing professional development opportunities are necessary to keep pace with rapidly evolving knowledge24.
The field of pharmacogenomics is rapidly evolving, propelled by advances in genomic technologies, data analytics, and digital health tools. As our understanding of gene-drug interactions deepens and the cost of genomic analysis continues to decline, pharmacogenomics is transitioning from research settings into routine clinical practice. This section explores emerging technologies that are enhancing the capabilities of pharmacogenomics and outlines key trends likely to shape its future25.
Next-generation sequencing technologies have revolutionized genomic medicine by enabling rapid, high-throughput, and cost-effective analysis of the genome.
· Whole genome sequencing (WGS) and whole exome sequencing (WES) allow for the identification of both known and novel pharmacogenomic variants.
· Unlike single-gene tests, NGS platforms can analyze multiple pharmacogenes simultaneously, increasing diagnostic yield and efficiency.
· Targeted pharmacogenomic panels are becoming more accessible and are being incorporated into clinical workflows for a variety of conditions25.
Artificial intelligence (AI) and machine learning (ML) are playing an increasingly important role in the interpretation of complex genomic data and the prediction of drug responses.
· Predictive modeling can integrate genomic, clinical, and environmental data to personalize treatment plans.
· AI-driven clinical decision support systems (CDSS) are being developed to provide real-time, evidence-based recommendations at the point of care.
· ML algorithms are also aiding in the discovery of new pharmacogenomic biomarkers by analyzing large datasets from biobanks and clinical trials25.
The integration of pharmacogenomic data into electronic health records is essential for translating genomic insights into clinical action.
· EHR systems are being enhanced to store, update, and flag relevant genetic information.
· Embedded CDSS tools can alert clinicians about potential gene-drug interactions and suggest alternative therapies or dosing strategies.
· Institutions like Vanderbilt University Medical Center and St. Jude Children’s Research Hospital have pioneered such systems, demonstrating improved prescribing safety and efficiency25.
Beyond genetics, epigenetic modifications, transcriptomics, proteomics, and metabolomics offer deeper insights into interindividual variability in drug response.
· Pharmacoepigenomics studies how DNA methylation, histone modification, and non-coding RNAs affect gene expression related to drug metabolism and efficacy.
· Multi-omics integration may uncover additional predictive biomarkers and refine existing pharmacogenomic models, leading to more accurate, personalized treatments25.
The increasing availability of DTC genetic tests is raising public awareness of pharmacogenomics and empowering patients to engage in their own health decisions.
· Companies such as 23andMe offer limited pharmacogenomic insights directly to consumers, though their clinical utility may vary.
· While DTC testing democratizes access to genomic information, it also raises concerns about interpretation accuracy, regulatory oversight, and informed decision-making without medical guidance26.
Efforts are underway to expand the benefits of pharmacogenomics beyond high-income countries.
· Global pharmacogenomics initiatives aim to develop population-specific databases, recognizing that genetic variation differs significantly across ethnic groups.
· Partnerships between academic institutions, governments, and international organizations are helping to build capacity in low- and middle-income countries, ensuring more equitable access to precision medicine26.
Several trends are expected to define the future of pharmacogenomics:
· Routine pre-emptive genotyping: Proactive genetic testing at birth or during early medical visits may become part of standard care, with results stored for lifelong use.
· Pharmacogenomics in rare diseases: Genomic profiling can uncover therapeutic options for patients with rare or undiagnosed conditions by identifying actionable variants.
· Companion diagnostics: The development of FDA-approved tests paired with specific drugs will continue to grow, particularly in oncology and immunology.
· Personalized drug development: Pharmaceutical companies may design drugs for genetically defined subgroups, improving success rates in clinical trials and enabling faster regulatory approval26.
To support the future of pharmacogenomics, parallel advancements in policy, education, and ethics are critical.
· Provider training programs must evolve to include genomics education at all levels.
· Updated regulatory policies will be needed to address issues of data sharing, informed consent, and equitable access.
· Public engagement will be essential to build trust, promote genetic literacy, and encourage informed participation in genomic healthcare26.
Pharmacogenomics is poised to transform modern medicine by making drug therapy safer, more effective, and tailored to the individual. Although challenges remain, continued research, technological innovation, and systemic healthcare reforms will accelerate its adoption. Personalized medicine is not a distant future—it is an emerging reality reshaping the therapeutic landscape.
1. Morganti S, Tarantino P, Ferraro E, D’Amico P, Duso BA, Curigliano G. Next generation sequencing (NGS): a revolutionary technology in pharmacogenomics and personalized medicine in cancer. Translational Research and Onco-omics Applications in the Era of Cancer Personal Genomics. 2019:9-30.
2. Mancinelli L, Cronin M, Sadée W. Pharmacogenomics: the promise of personalized medicine. AAPS Pharmsci. 2000 Mar; 2:29-41.
3. Sadee W. Drug therapy and personalized health care: pharmacogenomics in perspective. Pharmaceutical Research. 2008 Dec; 25(12): 2713-9.
4. Singh DB. The impact of pharmacogenomics in personalized medicine. Current Applications of Pharmaceutical Biotechnology. 2020: 369-94.
5. Kelton T. Pharmacogenomics: The rediscovery of the concept of tailored drug therapy and personalized medicine. Health Law. 2006; 19:1.
6. Lam YF. Principles of pharmacogenomics: pharmacokinetic, pharmacodynamic, and clinical implications. In Pharmacogenomics 2019 Jan 1 (pp. 1-53). Academic Press.
7. Hasanzad M, Sarhangi N, Hashemian L, Sarrami B. Principles of pharmacogenomics and pharmacogenetics. In Precision Medicine in Clinical Practice 2022 Oct 1 (pp. 13-32). Singapore: Springer Nature Singapore.
8. Palumbo S, Mariotti V, Pellegrini S. A narrative review on pharmacogenomics in psychiatry: scientific definitions, principles, and practical resources. Journal of Clinical Psychopharmacology. 2024 Jan 1; 44(1): 49-56.
9. van Schaik RH, Bach-Rojecky L, Primorac D. Principles of Pharmacogenetics. InPharmacogenomics in Clinical Practice 2024 Jan 5 (pp. 1-12). Cham: Springer International Publishing.
10. Dodson CH. Pharmacogenomics: Principles and relevance to oncology nursing. Clinical Journal of Oncology Nursing. 2017 Dec 1; 21(6).
11. Ma JD, Lee KC, Kuo GM. Clinical application of pharmacogenomics. Journal of Pharmacy Practice. 2012 Aug; 25(4): 417-27.
12. Quiñones L, Roco Á, Cayún JP, Escalante P, Miranda C, Varela N, Meneses F, Gallegos B, Zaruma-Torres F, Lares-Asseff I. Clinical applications of pharmacogenomics. Revista médica de Chile. 2017 Apr 1; 145(4): 483-500.
13. Huang SM, Goodsaid F, Rahman A, Frueh F, Lesko LJ. Application of pharmacogenomics in clinical pharmacology. Toxicology mechanisms and methods. 2006 Jan 1;16(2-3):89-99.
14. Norton RM. Clinical Pharmacogenomics: applications in pharmaceutical R&D. Drug Discovery Today. 2001 Feb 15; 6(4): 180-5.
15. Rioux PP. Clinical trials in pharmacogenetics and pharmacogenomics: methods and applications. American Journal of Health-System Pharmacy. 2000 May 1; 57(9): 887-98.
16. Severino G, Chillotti C, Stochino ME, Del Zompo M. Pharmacogenomics: state of the research and perspectives in clinical application. Neurological Sciences. 2003 May; 24: s146-8.
17. Singh DB. The impact of pharmacogenomics in personalized medicine. Current Applications of Pharmaceutical Biotechnology. 2020: 369-94.
18. Mancinelli L, Cronin M, Sadée W. Pharmacogenomics: the promise of personalized medicine. Aaps Pharmsci. 2000 Mar; 2:29-41.
19. Sadee W. Drug therapy and personalized health care: pharmacogenomics in perspective. Pharmaceutical Research. 2008 Dec; 25(12): 2713-9.
20. Harrison J. Impact of the Genomics Revolution on Drug Development and Personalized Medicine in Pharmaceutics.
21. Morganti S, Tarantino P, Ferraro E, D’Amico P, Duso BA, Curigliano G. Next generation sequencing (NGS): a revolutionary technology in pharmacogenomics and personalized medicine in cancer. Translational Research and Onco-omics Applications in the Era of Cancer Personal Genomics. 2019: 9-30.
22. De Leon J. Pharmacogenomics: the promise of personalized medicine for CNS disorders. Neuropsychopharmacology. 2009 Jan; 34(1): 159-72.
23. Paul-Chima O, Ugo E, Chukwudi A. The Role of Pharmacogenomics in Personalized Medicine: Historical Context, Principles, Applications, and Future Challenges. Practice. 14:16.
24. Vizirianakis IS. Challenges in current drug delivery from the potential application of pharmacogenomics and personalized medicine in clinical practice. Current Drug Delivery. 2004 Jan 1; 1(1): 73-80.
25. Morganti S, Tarantino P, Ferraro E, D’Amico P, Duso BA, Curigliano G. Next generation sequencing (NGS): a revolutionary technology in pharmacogenomics and personalized medicine in cancer. Translational Research and Onco-omics Applications in the Era of Cancer Personal Genomics. 2019: 9-30.
26. Amir-Aslani A, Mangematin V. The future of drug discovery and development: shifting emphasis towards personalized medicine. Technological Forecasting and Social Change. 2010 Feb 1; 77(2): 203-17.
|
Received on 04.07.2025 Revised on 14.08.2025 Accepted on 16.09.2025 Published on 11.10.2025 Available online from October 25, 2025 Res.J. Pharmacology and Pharmacodynamics.2025;17(4):311-318. DOI: 10.52711/2321-5836.2025.00048 ©A and V Publications All right reserved
|
|
|
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License. |
|