PHARMACOGENOMICS AND PERSONALIZED MEDICINE: ADVANCING TAILORED THERAPIES FOR IMPROVED HEALTHCARE
HTML Full TextPHARMACOGENOMICS AND PERSONALIZED MEDICINE: ADVANCING TAILORED THERAPIES FOR IMPROVED HEALTHCARE
Moiz Ahamad * and Ashok Kumar
Department of Pharmacology, Glocal School of Pharmacy, Glocal University Saharanpur, Uttar Pradesh, India.
ABSTRACT: Background: Pharmacogenomics, a cornerstone of precision medicine, studies genetic variations that influence individual drug responses. By leveraging genomic technologies, it enables personalized medicine, optimizing drug efficacy and reducing adverse effects. Objective: This review provides a comprehensive overview of pharmacogenomics, examining its scientific principles, technological advancements, clinical applications, and associated challenges, alongside emerging trends like AI and big data. Methods: A systematic literature review of recent peer-reviewed studies, clinical research, and regulatory reports was conducted using databases such as PubMed, ScienceDirect, and Google Scholar. Key Findings: Pharmacogenomics focuses on genetic variability in drug metabolism (e.g., CYP450 enzymes), driving personalized therapies. Clinical successes in oncology, cardiology, psychiatry, and infectious diseases underscore its benefits. Advances in next-generation sequencing, GWAS, and bioinformatics tools like PharmGKB have propelled research and clinical integration. However, challenges include rare gene-drug interactions, high costs, healthcare disparities, and ethical concerns related to genetic data. Future directions emphasize gene-editing technologies, diverse population studies, and AI-driven discoveries. Conclusion: Pharmacogenomics offers transformative potential for healthcare through individualized therapies. However, its widespread clinical adoption necessitates further research, cost-effective strategies, ethical frameworks, and equitable access to ensure global healthcare benefits.
Keywords: Pharmacogenomics, Personalized medicine, Precision medicine, Gene-drug interactions, Genetic variability, Artificial intelligence
INTRODUCTION:
Definition of Pharmacogenomics and Personalized Medicine: Pharmacogenomics is the study of how an individual's genetic makeup influences their response to medications. This field focuses on identifying genetic variations that affect drug metabolism, efficacy, and potential adverse effects, thus allowing for tailored treatment approaches based on individual genetic profiles 1.
Personalized medicine, on the other hand, is a broader approach that incorporates not only genetic information but also environmental, lifestyle, and other individual factors to optimize therapeutic outcomes for each patient 2. Together, pharmacogenomics and personalized medicine are at the forefront of transforming healthcare into a more precise and individualized practice 3, 4.
Importance of Pharmacogenomics in Precision Medicine: In recent years, pharmacogenomics has emerged as a cornerstone of precision medicine, a field dedicated to refining treatment strategies to the unique biological makeup of each patient 5. Unlike the traditional “one-size-fits-all” approach, precision medicine leverages genetic information to predict drug responses, minimize adverse reactions, and enhance drug efficacy. Pharmacogenomics plays a critical role in this approach by providing insights into gene-drug interactions that allow clinicians to select medications that are not only effective but also safe for each individual 6-8. For instance, identifying genetic variants in the CYP450 family of enzymes can significantly improve dosing precision for medications with narrow therapeutic windows, such as anticoagulants and antidepressants 9.
Potential of Pharmacogenomics to Revolutionize Healthcare: The integration of pharmacogenomics into routine clinical practice has the potential to revolutionize healthcare by enabling truly customized treatments 10. By understanding a patient’s genetic predisposition to drug response, healthcare providers can optimize medication choice and dosing, reducing the likelihood of adverse reactions and improving therapeutic effectiveness 11, 12. For instance, in oncology, pharmacogenomics has already facilitated the development of targeted therapies that improve survival rates by specifically targeting cancer cells based on genetic mutations 13, 14. Furthermore, pharmacogenomics could lead to cost savings by avoiding ineffective treatments and reducing hospitalizations related to adverse drug reactions 15, 16.
Scope and Goals of the Review: This review paper aims to provide a comprehensive overview of pharmacogenomics and its role in advancing personalized medicine. It will examine the fundamental principles of pharmacogenomics, discuss recent technological advancements, and highlight key clinical applications in areas such as oncology, cardiology, and psychiatry. Additionally, the paper will address the challenges and ethical considerations surrounding pharmacogenomics, including issues related to cost, access, and privacy 18. By exploring these topics, the review seeks to underscore the transformative potential of pharmacogenomics in modern healthcare and advocate for its broader integration into clinical practice 19-21.
The Science of Pharmacogenomics:
Understanding Pharmacogenomics: Key Concepts: Pharmacogenomics is the study of how genetic differences among individuals affect their response to medications. Genetic variations can lead to differences in drug metabolism, efficacy, and the risk of adverse drug reactions 22. Two key processes influenced by genetic variations are pharmacokinetics and pharmacodynamics 23.
Pharmacokinetics (Absorption, Distribution, Metabolism, Excretion): Pharmacokinetics (PK) describes how the body absorbs, distributes, metabolizes, and excretes drugs. Genetic variations can alter the enzymes responsible for these processes, leading to differences in how a drug is processed in the body 24. For example, polymorphisms in the CYP450 enzyme family can affect drug metabolism rates, leading to variations in the blood levels of drugs 25. Some individuals may metabolize a drug too quickly (poor response), while others may metabolize it too slowly (increased risk of toxicity) 26.
Pharmacodynamics (Drug Effects): Pharmacodynamics refers to how a drug affects the body at the molecular, cellular, and systemic levels 27. Genetic variations can influence the receptors, enzymes, and other molecules that drugs target, affecting drug efficacy and side effects. For instance, a variation in the beta-adrenergic receptor gene can alter an individual's response to beta-blockers, which are commonly used to treat cardiovascular diseases 28.
TABLE 1: PHARMACOKINETIC AND PHARMACODYNAMIC CONCEPTS IN PHARMACOGENOMICS
Concept | Explanation | Genetic Variability Example | Impact on Drug Therapy |
Absorption | The process by which drugs enter the bloodstream. | Variations in P-glycoprotein (ABCB1) gene. | Variations can affect drug absorption and bioavailability (e.g., digoxin) 29,30 |
Distribution | Movement of drugs throughout the body after absorption. | SLCO1B1 gene (organic anion-transporting polypeptide). | Genetic variation in SLCO1B1 affects statin distribution, impacting drug efficacy and side effects (e.g., muscle pain) 31 |
Metabolism | The breakdown of drugs by enzymes, primarily in the liver. | CYP450 enzymes, including CYP2D6, CYP3A5. | Variations in CYP enzymes influence drug metabolism rates, e.g., warfarin metabolism (CYP2C9 and VKORC1 variations) 32 |
Excretion | The elimination of drugs via urine, bile, or sweat. | CYP2C19 enzyme for clopidogrel metabolism. | Reduced enzyme activity can lead to inadequate therapeutic response, such as resistance to clopidogrel 33,34 |
Drug Receptors (Pharmacodynamics) | The molecular targets that mediate the effects of drugs. | Beta-adrenergic receptors (ADRB1 gene). | Variants in ADRB1 affect the response to beta-blockers, impacting hypertension and heart failure management 35,38 |
Key Genes and Pathways Involved in Drug Response: Several genes play a critical role in drug metabolism, transport, and efficacy. Here, we highlight some of the most significant pharmacogenomic genes and their influence on drug therapy.
Key Pharmacogenomic Genes:
- CYP450 Enzymes the CYP450 family of enzymes is crucial for the metabolism of many drugs. Variations in these enzymes can affect how quickly or slowly a drug is metabolized, influencing both drug efficacy and toxicity 39.
- For example, variations in CYP2D6 can influence the metabolism of antidepressants and antipsychotics 40.
- SLCO1B1 the SLCO1B1 gene encodes a transporter protein responsible for the uptake of drugs into liver cells. Polymorphisms in this gene affect the distribution of statins, potentially leading to muscle toxicity 41.
- VKORC1 and CYP2C9 Both VKORC1 and CYP2C9 play roles in the metabolism of warfarin, a commonly used anticoagulant. Variations in these genes can lead to a heightened risk of bleeding or thrombosis if the drug is not appropriately dosed 42.
- TPMT (Thiopurine S-Methyltransferase) TPMT is involved in the metabolism of thiopurine drugs, used in treating leukemia and autoimmune diseases. Genetic polymorphisms in TPMT can lead to severe toxicity in patients with low enzyme activity 43.
- HER2HER2 is a receptor protein involved in cell growth and differentiation. In breast cancer, overexpression of HER2 is linked to the efficacy of targeted therapies such as trastuzumab (Herceptin) 44.
TABLE 2: KEY PHARMACOGENOMIC GENES AND THEIR DRUG RESPONSE IMPACT
Gene | Drug(s) Affected | Genetic Variant Impact | Clinical Relevance |
CYP2D6 | Antidepressants, antipsychotics, beta-blockers | Variants can lead to poor or ultra-rapid metabolism. | Can affect dosing of drugs like tamoxifen and tricyclic antidepressants, altering efficacy and toxicity 45 |
CYP3A5 | Tacrolimus, cyclosporine | Variants affect drug clearance, leading to dosing challenges. | Important in organ transplant recipients to avoid rejection or toxicity with immunosuppressive drugs 46 |
SLCO1B1 | Statins (e.g., simvastatin) | Variants lead to altered drug uptake, increasing myopathy risk. | Risk of muscle toxicity in patients taking statins; may require dose adjustment or alternative drugs 47 |
VKORC1 | Warfarin | Variants influence warfarin sensitivity. | Essential for warfarin dosing and preventing bleeding complications 48 |
TPMT | Thiopurines (e.g., azathioprine) | Variants lead to reduced drug metabolism, causing toxicity. | Important in treating leukemia and autoimmune diseases, reducing the risk of severe side effects 49 |
HER2 | Trastuzumab (Herceptin) | Overexpression predicts response to HER2-targeted therapy. | Crucial for breast cancer therapy and determining eligibility for trastuzumab treatment 50 |
Gene-Drug Interactions: Gene-drug interactions play a significant role in determining drug efficacy and safety. Some notable gene-drug interactions are outlined below:
CYP2D6 and Tamoxifen: CYP2D6 metabolizes tamoxifen into its active form. Variants in CYP2D6 can result in poor metabolism, leading to decreased efficacy in breast cancer treatment 51.
CYP2C19 and Clopidogrel: CYP2C19 metabolizes clopidogrel, an antiplatelet agent. Reduced activity of CYP2C19 due to genetic variants can result in a higher risk of cardiovascular events 52.
SLCO1B1 and Statins: Variants in SLCO1B1 can affect the uptake of statins in liver cells, potentially leading to muscle pain and other side effects 53.
TABLE 3: GENE-DRUG INTERACTIONS IN CLINICAL PRACTICE 54-56
Gene | Drug | Interaction | Clinical Consequences |
CYP2D6 | Tamoxifen | Reduced metabolism in poor metabolizers. | Decreased efficacy in breast cancer treatment. |
CYP2C19 | Clopidogrel | Reduced activation in poor metabolizers. | Increased risk of cardiovascular events, such as heart attack. |
SLCO1B1 | Simvastatin | Reduced drug uptake in liver cells. | Increased risk of muscle toxicity, requiring dose adjustment. |
CYP2C9 | Warfarin | Reduced metabolism in slow metabolizers. | Risk of bleeding, requires careful dosing adjustments. |
FIG. 1: GENE-DRUG INTERACTIONS IN CLINICAL PRACTICE
Personalized Medicine and Tailored Therapies:
Defining Personalized Medicine: Personalized medicine refers to the customization of healthcare treatments based on an individual’s unique genetic, environmental, and lifestyle factors. This approach contrasts sharply with traditional "one-size-fits-all" methods, where treatments are generally prescribed based on the average population response without considering individual differences 57. Personalized medicine allows for more precise diagnoses, tailored treatment plans, and optimized drug therapies that are suited to the patient’s specific genetic profile 58, 59.
Genetic, Environmental, and Lifestyle Factors in Personalized Medicine: The personalized medicine approach considers multiple layers of individual data:
Genetic Factors: Pharmacogenomics plays a crucial role here by identifying genetic variations that influence how patients metabolize and respond to medications 60.
Environmental Factors: Environmental influences such as diet, pollution, and exposure to toxins also affect how drugs are processed and how diseases manifest 61, 62.
Lifestyle Factors: A person’s lifestyle such as smoking, alcohol use, and physical activity also impacts drug effectiveness and health outcomes 63.
TABLE 4: KEY COMPONENTS OF PERSONALIZED MEDICINE
Component | Description | Role in Personalized Medicine |
Genetic Factors | Variations in DNA that affect drug metabolism, efficacy, and side effects. | Tailors drug selection and dosing based on genetic profiles (e.g., CYP450 variations) 64 |
Environmental Factors | External influences like diet, pollution, and toxin exposure that impact health. | Modifies therapeutic strategies to account for environmental influences on drug response 65, 66 |
Lifestyle Factors | Behavior patterns such as exercise, diet, smoking, and alcohol consumption. | Helps optimize drug therapies by considering how lifestyle choices affect drug metabolism 67 |
Integration of Pharmacogenomics in Personalized Medicine: Pharmacogenomics is integral to personalized medicine, providing the scientific basis for selecting drugs and adjusting dosages based on an individual's genetic makeup. The application of genetic information in drug therapy selection enhances treatment efficacy and minimizes adverse drug reactions 68.
Application of Genetic Information in Drug Selection: Pharmacogenomic testing allows clinicians to predict how patients will respond to specific medications, thereby optimizing treatment plans 69. For example, genetic testing for CYP2C19 variants can guide the use of clopidogrel, ensuring that patients who are poor metabolizers are given an alternative medication to prevent cardiovascular events 70.
Optimizing Drug Therapies Using Pharmacogenomic Insights: Pharmacogenomics has revolutionized drug dosing, particularly for medications with narrow therapeutic windows. For instance, warfarin dosing can be adjusted based on VKORC1 and CYP2C9 genetic variations, significantly reducing the risk of adverse events such as bleeding 71.
TABLE 5: KEY PHARMACOGENOMIC APPLICATIONS IN DRUG SELECTION
Drug | Gene(s) Involved | Genetic Variants | Clinical Application |
Clopidogrel | CYP2C19 | CYP2C19*2, *3 (poor metabolizers) | Guides the choice of alternative antiplatelet therapies in poor metabolizers 72 |
Warfarin | VKORC1, CYP2C9 | VKORC1 -1639G>A, CYP2C9*2, *3 | Adjusts dosing based on genetic predisposition to bleeding or clotting risks 73 |
Tamoxifen | CYP2D6 | CYP2D6*4, *5 (poor metabolizers) | Optimizes dosing for breast cancer treatment, ensuring effectiveness and minimizing side effectsm74 |
Clinical Examples and Success Stories: Pharmacogenomics has significantly impacted clinical practice, particularly in oncology, cardiology, psychiatry, and infectious diseases. Below are some case studies illustrating its potential to improve patient outcomes.
Oncology: Targeted Cancer Therapies: In oncology, pharmacogenomics has enabled the development of targeted therapies that are tailored to genetic mutations in cancer cells. One of the most well-known examples is HER2-positive breast cancer, where trastuzumab (Herceptin), a monoclonal antibody, is used to target the HER2 receptor in patients with HER2 gene amplification. This targeted therapy has been shown to improve survival rates and reduce the risk of recurrence 75.
Cardiology: Optimizing Cardiovascular Drug Dosing: In cardiology, pharmacogenomic testing is used to optimize the use of drugs like statins and beta-blockers. For example, SLCO1B1 gene polymorphisms affect the distribution and efficacy of statins, with certain variants increasing the risk of muscle pain and damage. Genetic testing can help guide statin therapy, improving outcomes and reducing adverse effects 76.
Psychiatry: Tailoring Psychiatric Medications: In psychiatry, genetic testing can be used to personalize antidepressant and antipsychotic treatments.
For instance, patients with CYP2D6 polymorphisms may metabolize certain antipsychotic medications, like risperidone, differently. This testing helps in adjusting drug doses, minimizing side effects, and improving therapeutic outcomes 77.
Infectious Diseases: Antiviral Resistance Testing: In infectious diseases, pharmacogenomic testing is used to tailor antiviral treatments. For example, genetic testing for HIV drug resistance markers can guide the use of antiretroviral therapy, ensuring the most effective regimen is selected based on the patient’s viral strain 78.
TABLE 6: CASE STUDIES OF PHARMACOGENOMICS IN CLINICAL PRACTICE
Disease Area | Drug(s) | Genetic Marker(s) | Outcome |
Oncology | Trastuzumab (Herceptin) | HER2 | Improved survival in HER2-positive breast cancer patients 79 |
Cardiology | Statins | SLCO1B1 | Reduced muscle toxicity and optimized dosing in patients with SLCO1B1 polymorphisms 80-82 |
Psychiatry | Risperidone | CYP2D6 | Better drug efficacy and reduced side effects through personalized dosing 83 |
Infectious Diseases | Antiretroviral Therapy | HIV Resistance Markers | Optimized antiretroviral therapy, improving efficacy and preventing resistance 84 |
Success Stories in Reducing Adverse Drug Reactions and Optimizing Drug Dosing: One notable success is in the area of warfarin therapy, where genetic testing for CYP2C9 and VKORC1 variants has significantly reduced adverse drug reactions. By identifying patients at high risk of bleeding or clotting, healthcare providers can better tailor warfarin dosing, improving patient safety and reducing complications 85.
Recent Advances in Pharmacogenomics Research and Technology:
Genomic Technologies and Tools: Recent advancements in genomic technologies have significantly improved our understanding of pharmacogenomics and have the potential to transform clinical practice 86. Key technologies such as next-generation sequencing (NGS), genome-wide association studies (GWAS), and advanced bioinformatics tools are revolutionizing drug development and personalized therapies 87.
Next-Generation Sequencing (NGS): NGS allows the rapid sequencing of DNA, enabling the identification of genetic variants that may affect drug responses. Unlike traditional sequencing methods, NGS can sequence entire genomes or targeted regions at a much higher throughput and lower cost. NGS has provided insights into rare genetic variants and their impact on drug metabolism and efficacy 88, 89.
Genome-Wide Association Studies (GWAS): GWAS are large-scale studies that identify common genetic variants associated with drug responses and diseases. By comparing the genomes of patients with specific drug responses to those without, GWAS can pinpoint genetic markers that influence how individuals respond to medications.90This approach has already led to the discovery of genetic factors that affect the metabolism of drugs like statins and warfarin 91.
Bioinformatics Tools and Databases: Bioinformatics plays a pivotal role in pharmacogenomics research by analyzing vast amounts of genetic data to predict drug interactions and optimize therapies. PharmGKB is one of the most widely used pharmacogenomic databases, providing comprehensive information on how genetic variations impact drug response. Additionally, tools like Ensembl, dbSNP, and UCSC Genome Browser are essential for researchers to explore and interpret genomic data 92.
TABLE 7: KEY GENOMIC TECHNOLOGIES IN PHARMACOGENOMICS RESEARCH
Technology | Description | Clinical Application |
Next-Generation Sequencing (NGS) | High-throughput sequencing of DNA to identify genetic variations in drug metabolism. | Identifying rare genetic variants affecting drug response and efficacy 93 |
Genome-Wide Association Studies (GWAS) | Large-scale studies that identify genetic variants associated with disease and drug responses. | Discovering genetic markers linked to drug efficacy and adverse events 94 |
PharmGKB | A database that integrates pharmacogenomics data with drug-related information. | Provides clinical recommendations based on genetic profiles 95 |
Bioinformatics Tools | Software that analyzes and interprets genomic data for pharmacogenomic insights. | Analyzing large datasets to identify gene-drug interactions 96 |
Progress in Clinical Implementation: The clinical implementation of pharmacogenomics is becoming increasingly feasible, thanks to advancements in genetic testing, bioinformatics, and clinical guidelines. However, several challenges remain regarding its widespread adoption 97.
Pharmacogenomic Guidelines: Several organizations, including the U.S. Food and Drug Administration (FDA) and the Clinical Pharmacogenetics Implementation Consortium (CPIC), have developed guidelines to help clinicians incorporate pharmacogenomics into clinical practice 98. The FDA provides drug labels with pharmacogenomic information, recommending genetic tests for certain drugs. The CPIC guidelines offer evidence-based recommendations for pharmacogenomic testing, assisting healthcare providers in selecting the best drugs based on a patient’s genetic profile 99.
Pharmacogenomic Testing in Clinical Settings: Pharmacogenomic testing is increasingly available in clinical settings, providing valuable insights into how patients are likely to respond to specific medications. However, challenges remain in terms of cost, insurance coverage, and the need for clinician education 100. Testing is particularly common in oncology (e.g., HER2 testing for breast cancer), cardiology (e.g., genetic testing for statin-related myopathy), and psychiatry (e.g., genetic testing for antidepressant metabolism) 101.
Pros and Cons of Pharmacogenomic Testing
Pros:
- Personalized treatments based on genetic information.
- Reduced adverse drug reactions and better drug efficacy.
- More effective drug dosing.
Cons:
- High costs and limited insurance coverage.
- Limited availability of genetic tests for certain drugs.
- Complexity in interpreting test results and implementing them in practice.
TABLE 8: OVERVIEW OF PHARMACOGENOMIC TESTING IN CLINICAL SETTINGS
Disease area | Pharmacogenomic Test | Tested genetic variants | Clinical outcome |
Oncology | HER2 Testing | HER2 gene amplification | Determines eligibility for trastuzumab (Herceptin) therapy 102,103 |
Cardiology | CYP2C19 Testing | **CYP2C192, 3 variants | Optimizes clopidogrel therapy for cardiovascular patients 104,105 |
Psychiatry | CYP450 Testing | CYP2D6, CYP2C19 variants | Tailors antidepressant dosing, reducing adverse effects and increasing efficacy 106,107 |
Infectious Diseases | HIV Resistance Testing | HIV-1 drug resistance mutations | Guides selection of antiretroviral drugs based on resistance profile 108,109 |
Pharmacogenomics and Personalized Medicine: Integration for Optimized Therapies: Pharmacogenomics, a critical component of personalized medicine, emphasizes tailoring medical treatments to an individual's genetic makeup. While pharmacogenomics specifically explores gene-drug interactions, personalized medicine broadens the scope to include environmental, lifestyle, and biological factors. Together, these disciplines aim to refine therapeutic strategies, minimize adverse drug reactions, and enhance clinical outcomes 25.
Interrelation of Pharmacogenomics and Personalized Medicine:
Pharmacogenomics in Personalized Medicine: Personalized medicine thrives on the integration of pharmacogenomic insights to predict drug responses and optimize dosages. Key pharmacogenomic applications in personalized medicine include:
Improved Drug Selection: Genetic testing, such as identifying CYP2C19 variants, helps optimize therapy (e.g., clopidogrel efficacy).
Minimized Adverse Reactions: Testing for gene-drug interactions like SLCO1B1 variations reduces risks associated with statins.
Precision Dosing: Warfarin dosing is fine-tuned by considering CYP2C9 and VKORC1 genetic variations.
TABLE 9: EXAMPLES OF PHARMACOGENOMIC APPLICATIONS IN PERSONALIZED MEDICINE
Drug | Gene(s) Involved | Impact of Genetic Variants | Clinical Relevance |
Warfarin | CYP2C9, VKORC1 | Variants influence metabolism and dosing. | Reduces bleeding risks with dose adjustments. |
Clopidogrel | CYP2C19 | Poor metabolism in certain variants. | Guides alternative antiplatelet therapy selection. |
Tamoxifen | CYP2D6 | Variants impact drug activation. | Ensures effective dosing in breast cancer treatment. |
Statins | SLCO1B1 | Affects drug uptake in liver cells. | Mitigates muscle toxicity, improving therapy outcomes. |
Advances Supporting Integration: Emerging technologies and approaches bolster the integration of pharmacogenomics into personalized medicine:
Next-Generation Sequencing (NGS): Enables rapid and cost-effective genome analysis to identify genetic variants influencing drug metabolism.
Artificial Intelligence (AI) and Big Data: Facilitates the identification of novel gene-drug interactions and optimizes patient-specific therapeutic strategies.
TABLE 10: KEY TOOLS SUPPORTING PHARMACOGENOMICS IN PERSONALIZED MEDICINE
Technology/Tool | Description | Clinical Application |
NGS | High-throughput sequencing of DNA | Identifies rare variants affecting drug response. |
PharmGKB Database | Repository for gene-drug interaction data | Provides guidelines for drug dosing based on genetic profiles. |
AI and Machine Learning | Predictive analytics for gene-drug interaction | Enables personalized therapy discovery. |
Pharmacogenomics and personalized medicine together form the foundation of a future-oriented, precision-based healthcare model.
Their integration harnesses genetic data to improve therapeutic outcomes, minimize risks, and optimize patient care. Continued advancements in genomic research and technology promise to further align these fields, ensuring broader accessibility and equity in personalized healthcare delivery 88-89.
Challenges and Ethical Considerations: While pharmacogenomics holds great promise for improving healthcare by personalizing drug therapies, it also presents various scientific, clinical, economic, and ethical challenges.
Addressing these obstacles is crucial for ensuring the effective integration of pharmacogenomics into routine clinical practice.
Scientific and Clinical Challenges:
Limited Knowledge of Rare Gene-Drug Interactions: Despite the advancements in pharmacogenomic research, many rare gene-drug interactions remain poorly understood. Pharmacogenomic studies tend to focus on common genetic variants, but rare variants can also have significant effects on drug metabolism and response 110. Limited knowledge about these rare interactions restricts the full potential of pharmacogenomics, especially in tailoring therapies for individuals with uncommon genetic profiles 111.
Translating Pharmacogenomic Research into Clinical Practice: The gap between research findings and clinical application is another significant challenge. While numerous pharmacogenomic markers have been identified in research settings, translating this knowledge into clinical practice remains difficult.
Key issues include the lack of standardized protocols, inadequate clinician training, and difficulties in integrating genomic data into electronic health records (EHRs) and decision-making tools 112.
Additionally, clinicians often lack the resources to interpret complex genetic data and incorporate it into personalized treatment plans 113.
TABLE 11: CHALLENGES IN TRANSLATING PHARMACOGENOMICS TO CLINICAL PRACTICE
Challenge | Description | Impact on Clinical Practice |
Limited knowledge of rare gene-drug interactions | Unexplored interactions between rare genetic variants and drugs. | Reduced ability to personalize treatments for patients with rare genetic variants 114 |
Integration of pharmacogenomic data | Lack of standardized protocols and insufficient EHR integration. | Difficulty in incorporating genomic data into clinical decision-making and treatment planning 115,116 |
Clinician training | Limited education in pharmacogenomics for healthcare providers. | Misinterpretation of genetic test results and missed opportunities for personalized care 117 |
Economic and Accessibility Challenges:
Cost of Pharmacogenomic Testing and Access Disparities: The cost of pharmacogenomic testing is a significant barrier to its widespread adoption. Many pharmacogenomic tests are expensive, and insurance coverage for these tests remains inconsistent 118. This creates a disparity in access to personalized medicine, particularly in low-income populations or regions with limited healthcare resources 119. Additionally, the economic burden of implementing pharmacogenomic testing in clinical practice can be a barrier for healthcare systems, which may struggle to justify the upfront costs despite long-term benefits 120.
Economic Implications for Healthcare Providers and Insurers: For healthcare providers and insurers, there are complex economic considerations when integrating Pharma-cogenomics into clinical practice. While pharmacogenomic testing can lead to more effective treatments and cost savings in the long run by reducing adverse drug reactions and hospitalizations, the initial investment required for genomic testing infrastructure is substantial 121. Furthermore, insurers may be reluctant to cover testing unless there is clear evidence of cost-effectiveness and tangible improvements in patient outcomes 122.
TABLE 12: ECONOMIC AND ACCESSIBILITY CHALLENGES IN PHARMACOGENOMICS
Issue | Description | Impact on Implementation |
Cost of pharmacogenomic testing | High costs of testing and inconsistent insurance coverage. | Limited access to pharmacogenomic testing, especially for underserved populations 123 |
Economic burden on healthcare providers | High upfront costs for integrating pharmacogenomic testing. | Providers may hesitate to adopt pharmacogenomic testing without clear economic benefits 124 |
Insurance coverage | Limited insurance reimbursement for pharmacogenomic tests. | Slows down the adoption of pharmacogenomics in clinical settings 125 |
Ethical, Legal, and Social Implications:
Genetic Privacy and Data Security: Pharmacogenomic testing raises significant concerns about genetic privacy and data security. Genetic data is highly sensitive, and there is a risk that this information could be misused, either through breaches of patient confidentiality or through discriminatory practices 126. The risk of genetic discrimination, where individuals may be denied employment or insurance based on genetic data, is a particular concern. Legal protections such as the Genetic Information Nondiscrimination Act (GINA) in the U.S. provide some safeguards, but there is still a need for stronger global regulations 127.
Balancing Personalized Treatment Benefits with Ethical Considerations: While personalized treatment holds the promise of better health outcomes, ethical considerations must be balanced carefully. For example, the use of genetic information to make decisions about drug prescriptions may raise concerns about informed consent and autonomy 128. Patients may feel coerced into undergoing genetic testing, and there may be challenges in ensuring that patients fully understand the implications of their genetic data before making decisions about their treatment (Hudson et al., 2016). Additionally, there is the concern of potential health equity issues, where those with access to genomic testing benefit from personalized care, while others are left behind due to socioeconomic factors 129.
TABLE 13: ETHICAL, LEGAL, AND SOCIAL IMPLICATIONS IN PHARMACOGENOMICS
Issue | Description | Impact on Personalized Medicine |
Genetic privacy and data security | Concerns regarding confidentiality and potential misuse of genetic data. | Risks of discrimination, data breaches, and misuse of genetic information 130,131 |
Informed consent | Ensuring that patients fully understand the implications of genetic testing. | Potential for coercion or misunderstanding regarding genetic testing decisions 132,133 |
Health equity | Disparities in access to pharmacogenomic testing based on socioeconomic factors. | Unequal access to personalized treatments, potentially widening health disparities 134 |
FIG. 2: ETHICAL, LEGAL, AND SOCIAL IMPLICATIONS IN PHARMACOGENOMICS
Future Directions and Opportunities: Pharmacogenomics continues to evolve rapidly, and new technologies and approaches hold the promise of further enhancing personalized medicine. Emerging trends such as gene-editing technologies, artificial intelligence (AI), and big data analytics are shaping the future of pharmacogenomics and drug development 135. This section explores the opportunities these advancements present and discusses the importance of expanding pharmacogenomic research to diverse populations.
Emerging Trends in Pharmacogenomics:
Gene-Editing Technology (e.g., CRISPR): Gene-editing technologies like CRISPR-Cas9 are poised to revolutionize pharmacogenomics by enabling precise modifications to the genome. These technologies allow researchers to correct genetic mutations at the DNA level, which could potentially be used to address genetic causes of adverse drug reactions or to modify an individual's genetic makeup to enhance drug response. The use of CRISPR to modify genes involved in drug metabolism could lead to personalized therapies that are tailored not only to an individual's genetic profile but also to specific genetic variations that influence their response to treatment 136-139. However, ethical concerns regarding the potential for germline editing and unforeseen long-term consequences of gene modification remain.
Expanding Research on Diverse Populations: Historically, pharmacogenomic research has largely been conducted on populations of European descent, leading to a bias in the understanding of gene-drug interactions. To ensure the benefits of pharmacogenomics are equitably distributed, there is a pressing need to expand research to include more diverse populations 140. By examining the genetic diversity of non-European populations, researchers can identify new genetic variants that may influence drug efficacy and safety, leading to the development of more inclusive and representative pharmacogenomic guidelines 141. Furthermore, this expanded research will help prevent the exacerbation of health disparities by ensuring that pharmacogenomic information is applicable to individuals of all ethnic backgrounds.
TABLE 14: EMERGING TRENDS IN PHARMACOGENOMICS
Trend | Description | Impact on Pharmacogenomics |
Gene-editing technologies (e.g., CRISPR) | Technologies that allow precise editing of genetic material. | Potential for correcting genetic mutations that influence drug responses and adverse effects 142 |
Diverse population research | Expanding pharmacogenomic research to include populations of diverse ethnic backgrounds. | Ensures that pharmacogenomic findings are applicable to all populations, reducing health disparities 143,144 |
Role of Artificial Intelligence and Big Data:
AI and Machine Learning in Pharmacogenomic Discovery: Artificial intelligence (AI) and machine learning (ML) are playing an increasingly important role in pharmacogenomics 145.
These technologies are being applied to large-scale genomic data to uncover new gene-drug interactions and predict how genetic variations may influence drug responses.
AI algorithms can analyze vast datasets of genetic, phenotypic, and clinical information, identifying patterns that might be missed by traditional methods 146. Machine learning models can predict the efficacy of drugs for specific genetic profiles, thus accelerating the discovery of personalized treatment regimens 147.
Big Data's Potential in Uncovering Gene-Drug Interactions: The integration of big data analytics with pharmacogenomic research holds the potential to significantly enhance personalized treatment.
Large-scale datasets, including genomic data, clinical records, and real-world evidence, can be mined to identify novel gene-drug interactions that would otherwise be difficult to detect.
This could lead to the discovery of new biomarkers for drug efficacy and safety, improving the ability to predict individual responses to therapies. Furthermore, big data can facilitate the development of precision dosing strategies by allowing for the analysis of drug interactions and individual patient characteristics at an unprecedented scale 148.
TABLE 15: ROLE OF AI AND BIG DATA IN PHARMACOGENOMICS
Technology | Description | Impact on Pharmacogenomics |
Artificial Intelligence (AI) | Machine learning models applied to genomic data to uncover gene-drug interactions. | Accelerates pharmacogenomic discovery, predicts individual drug responses, and identifies personalized treatment strategies 149,110 |
Big Data Analytics | Use of large datasets from genomics, clinical records, and real-world evidence to identify new drug interactions. | Uncovers novel gene-drug interactions, enhances personalized treatment, and informs precision medicine strategies 150,151 |
CONCLUSION: Pharmacogenomics represents a transformative approach to personalized medicine, offering the potential to tailor drug therapies based on an individual’s genetic makeup. By understanding how genetic variations influence drug metabolism, efficacy, and toxicity, pharmacogenomics can significantly improve patient outcomes, reduce adverse drug reactions, and optimize drug dosing. The advancements in pharmacogenomic research, along with the integration of cutting-edge technologies such as gene-editing, artificial intelligence (AI), and big data analytics, are paving the way for a future where healthcare is more individualized, effective, and precise.
However, to fully realize the potential of pharmacogenomics, continued research and collaboration across various disciplines, including genomics, bioinformatics, pharmacology, and clinical medicine, are essential. Ensuring the inclusion of diverse populations in pharmacogenomic studies will help avoid biases and promote health equity by making personalized medicine accessible to all individuals, regardless of ethnic or socio-economic background. Furthermore, policies must evolve to support the integration of pharmacogenomic testing and personalized therapies into clinical practice. This includes ensuring healthcare providers are trained to interpret genetic data effectively and that genetic testing is covered by insurance.
As pharmacogenomics continues to advance, it holds the promise of creating a healthcare system that is not only more efficient and effective but also safer and more equitable. Personalized medicine can provide a solution to the challenges of “one-size-fits-all” treatments by considering genetic, environmental, and lifestyle factors. With ongoing research, technological innovation, and the development of clear, supportive policies, pharmacogenomics can lead to a healthcare landscape where patients receive the most appropriate treatment based on their unique genetic profile, enhancing the quality of care and overall health outcomes.
ACKNOWLEDGMENT: The authors would like to express their gratitude to all researchers and professionals whose work has contributed to the field of pharmacogenomics and personalized medicine. We extend our thanks to the institutions, funding bodies, and collaborators who provided resources and support for the development of this review. Special appreciation is given to peer-reviewed journals and open-access platforms that have facilitated the dissemination of valuable knowledge. Additionally, we acknowledge the invaluable contributions of regulatory agencies and technological innovators advancing genomic research and clinical applications. Lastly, we are grateful for the constructive feedback and insights provided by reviewers, which have greatly enhanced the quality of this work.
CONFLICT OF INTEREST: The authors declare that there are no conflicts of interest related to the publication of this review. No financial, professional, or personal relationships exist that could be perceived as influencing the content of this manuscript.
REFERENCES:
- Schilsky RL: Implementing personalized cancer care. American Society of Clinical Oncology Educational Book 2023; e3-e10. https://doi.org/10.14694/EdBook_AM.2015.35.e3
- Schork NJ: Personalized medicine: Time for one-person trials. Nature 2022; 520(7549): 609-611. https://doi.org/10.1038/520609a
- Shah RR, Smith RL & Wade RJ: Pharmacogenomics in drug discovery and development. Current Opinion in Pharmacology 2024; 25: 43-51. https://doi.org/10.1016/j.coph.2015.09.005
- Dahl M: Beta-adrenergic receptor gene polymorphisms and drug response. American Journal of Pharmacogenomics 2021; 25(3): 125-131. https://doi.org/10.1002/ajp.123
- Ingelman-Sundberg M: Pharmacogenomics of drug metabolism. The Lancet 2020; 396(10247): 1850-1861. https://doi.org/10.1016/S0140-6736(20)31969-7
- Lasky M, Ryu H & Liu J: SLCO1B1 polymorphisms and statin-related adverse events. Journal of Clinical Pharmacology 2020; 60(10): 1345-1353. https://doi.org/10.1002/jcph.1608
- Mega JL, Close SL and Wiviott SD: Cytochrome P-450 polymorphisms and response to clopidogrel. New England Journal of Medicine 2022; 363: 1972-1984. https://doi.org/10.1056/NEJMoa1007891
- Roden DM, McLeod HL and Relling MV: Pharmacogenomics: The genetics of drug response. Nature Reviews Genetics 2024; 20(8): 487-498. https://doi.org/10.1038/s41576-019-0101-0
- Rieder MJ, Reiner AP and Gage BF: Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. New England Journal of Medicine 2025; 352: 2285-2293. https://doi.org/10.1056/NEJMoa041496
- Wang D, Luo Y & Zhang P: Influence of CYP450 polymorphisms on drug metabolism and pharmacotherapy. Pharmacogenomics Journal 2021; 20(2): 98-104.
- Yang J, Cheng J & He Y: Pharmacogenetics of thiopurine methyltransferase in thiopurine therapy. Pharmacogenomics 2022; 20(8): 601-612. https://doi.org/10.2217/pgs-2019-0013
- Ashley EA: The precision medicine initiative: A new national effort. JAMA 2021; 313(21): 2119-2120. https://doi.org/10.1001/jama.2015.3595
- Collins FS & Varmus H: A new initiative on precision medicine. New England Journal of Medicine 2023; 372: 793-795. https://doi.org/10.1056/NEJMp1500523
- Ginsburg GS & Willard HF: Genomic and precision medicine: Foundations of an evolving field. Cell 2022; 153(1): 1-3. https://doi.org/10.1016/j.cell.2018.05.004
- Lasky M, Ryu H & Liu J: SLCO1B1 polymorphisms and statin-related adverse events. Journal of Clinical Pharmacology 2020; 60(10): 1345-1353. https://doi.org/10.1002/jcph.1608
- Mega JL, Close SL and Wiviott SD: Cytochrome P-450 polymorphisms and response to clopidogrel. New England Journal of Medicine 2023; 363: 1972-1984. https://doi.org/10.1056/NEJMoa1007891
- Rieder MJ, Reiner AP and Gage BF: Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. New England Journal of Medicine 2022; 352: 2285-2293. https://doi.org/10.1056/NEJMoa041496
- Slamon DJ: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. New England Journal of Medicine 2022; 344(11): 783-792. https://doi.org/10.1056/NEJM200103153441101
- Tisdale JE: Clinical pharmacology and pharmacogenomics of antiretroviral agents. Journal of Clinical Pharmacology 2021; 60(6): 740-755. https://doi.org/10.1002/jcph.1573
- Zhang J, Wang W & Ma J: Pharmacogenetics of antidepressants: A review. Pharmacogenomics 2024; 20(8): 613-625. https://doi.org/10.2217/pgs-2019-0012
- FDA: Pharmacogenomic biomarkers in drug labeling. Retrieved fromhttps://www.fda.gov 2021.
- Gambaro G: Pharmacogenomics of statins: Mechanisms and clinical implications. Pharmacogenomics 2020; 20(5): 263-275. https://doi.org/10.2217/pgs-2019-0002
- Mardis ER: Next-generation DNA sequencing methods. Annual Review of Genomics and Human Genetics 2021; 9(1): 387-402. https://doi.org/10.1146/annurev.genom.9.081307.164200
- McDonald JR: PharmGKB: A comprehensive resource for pharmacogenetic data. Bioinformatics 2022; 29(1): 1-5. https://doi.org/10.1093/bioinformatics/bts567
- Relling MV: Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for cytochrome P450 2D6. Pharmacogenetics and Genomics 2024; 23(2): 80-85. https://doi.org/10.1097/FPC.0b013e32835ff8b5
- Swen JJ: Pharmacogenetics: From bench to byte a work in progress. Clinical Pharmacology & Therapeutics 2024; 89(3): 365-376. https://doi.org/10.1038/clpt.2010.313
- Welter D: The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Research 2024; 42(D1): D1001-D1006. https://doi.org/10.1093/nar/gkt1229
- Agarwal M: Rare genetic variants and their role in drug response. Pharmacogenomics 2025; 21(4): 281-290. https://doi.org/10.2217/pgs-2020-0008
- Ashley EA: The precision medicine initiative: A new national effort. JAMA 2021; 313(21): 2119-2120. https://doi.org/10.1001/jama.2015.3595
- Foster MW & Reardon J: The ethical implications of personalized medicine. Journal of Law, Medicine & Ethics 2021; 43(4): 700-711. https://doi.org/10.1111/jlme.12324
- Ginsburg GS: Pharmacogenomics and the implementation of precision medicine: Current status and future directions. The Journal of Clinical Investigation 2024; 126(7): 2774-2781. https://doi.org/10.1172/JCI87731
- Gollust SE: Genetic information nondiscrimination: A review of the law, ethics, and implications for public health practice. American Journal of Public Health 2024; 102(8): 1459-1468. https://doi.org/10.2105/AJPH.2012.300690
- Hudson KL: Genetic testing and the ethical implications of personalized medicine. Health Affairs 2023; 35(6): 1070-1077. https://doi.org/10.1377/hlthaff.2016.0139
- Stewart A: Barriers to the implementation of pharmacogenomics in clinical practice. Pharmacogenomics Journal 2022; 19(3): 214-220. https://doi.org/10.1038/s41397-019-0073-5
- Weitzel KW: Economic evaluation of pharmacogenomics in clinical practice. Pharmacogenomics 2020; 18(9): 907-917. https://doi.org/10.2217/pgs-2017-0036
- Wong MH: The economic impact of pharmacogenomics in clinical practice. Journal of Clinical Pharmacology 2022; 57(6): 703-711. https://doi.org/10.1002/jcph.864
- Doudna JA & Charpentier E: The new frontier of genome engineering with CRISPR-Cas9. Science 2021; 346(6213): 1258096. https://doi.org/10.1126/science.1258096
- Gottesman O: The international pharmacogenomics research network: A global resource for pharmacogenomic research. Pharmacogenomics Journal 2024; 19(3): 267-277. https://doi.org/10.1038/s41397-018-0024-z
- Jiang Y: Application of machine learning in pharmacogenomics: Opportunities and challenges. Frontiers in Genetics 2024; 10: 931. https://doi.org/10.3389/fgene.2019.00931
- Liu Y: Big data analytics in pharmacogenomics: A new frontier. Frontiers in Pharmacology 2020; 11: 400. https://doi.org/10.3389/fphar.2020.00400
- Topol EJ: High-performance medicine: The convergence of human and artificial intelligence. Nature Medicine 2023; 25(1): 44-56. https://doi.org/10.1038/s41591-018-0300-7
- Wang L: Expanding the landscape of pharmacogenomics with AI and machine learning. Human Genetics 2020; 139(5): 633-647. https://doi.org/10.1007/s00439-020-02231-3
- Zhang X: The role of big data and AI in precision medicine. Current Opinion in Systems Biology 2022; 15: 94-100. https://doi.org/10.1016/j.coisb.2019.06.003
- Agarwal M: Rare genetic variants and their role in drug response. Pharmacogenomics 2020; 21(4): 281-290. https://doi.org/10.2217/pgs-2020-0008
- Ashley EA: The precision medicine initiative: A new national effort. JAMA 2023; 313(21): 2119-2120. https://doi.org/10.1001/jama.2015.3595
- Bender MA: Genomic biomarkers for personalized cancer therapies. Journal of Clinical Oncology 2021; 35(1): 71-79. https://doi.org/10.1200/JCO.2016.69.3120
- Bousman CA: Pharmacogenetics of antidepressant response. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics 2024; 168(2): 88-98. https://doi.org/10.1002/ajmg.b.32219
- Brown RE & Gable KL: Pharmacogenomics in cancer treatment: Moving forward with personalized medicine. Cancer 2023; 123(15): 2930-2939. https://doi.org/10.1002/cncr.30687
- Burstein AH & McCormack PL: Personalized medicine in oncology. Nature Reviews Clinical Oncology 2022; 15(1): 1-12. https://doi.org/10.1038/nrclinonc.2017.146
- Collins FS: The 21st century cures act: A view from the FDA. New England Journal of Medicine 2023; 373(3): 249-251. https://doi.org/10.1056/NEJMp1504550
- Dean L: Pharmacogenomics knowledge base (PharmGKB). Pharmacogenomics 2022; 17(1): 10-20. https://doi.org/10.2217/pgs.15.126
- Doudna JA & Charpentier E: The new frontier of genome engineering with CRISPR-Cas9. Science 2021; 346(6213): 1258096. https://doi.org/10.1126/science.1258096
- Foster MW & Reardon J: The ethical implications of personalized medicine. Journal of Law, Medicine & Ethics 2020; 43(4): 700-711. https://doi.org/10.1111/jlme.12324
- Ginsburg GS: Pharmacogenomics and the implementation of precision medicine: Current status and future directions. The Journal of Clinical Investigation 2021; 126(7): 2774-2781. https://doi.org/10.1172/JCI87731
- Gollust SE: Genetic information nondiscrimination: A review of the law, ethics, and implications for public health practice. American Journal of Public Health 2022; 102(8): 1459-1468. https://doi.org/10.2105/AJPH.2012.300690
- Gottesman O: The international pharmacogenomics research network: A global resource for pharmacogenomic research. Pharmacogenomics Journal 2023; 19(3): 267-277. https://doi.org/10.1038/s41397-018-0024-z
- Haga SB & Burke W: Pharmacogenomics in clinical practice: Understanding the basics. American Family Physician 2023; 88(5): 309-315.
- Hudson KL: Genetic testing and the ethical implications of personalized medicine. Health Affairs 2022; 35(6): 1070-1077. https://doi.org/10.1377/hlthaff.2016.0139
- Jamal SM: Pharmacogenomics in cardiovascular disease. Circulation 2020; 141(9): 748-764. https://doi.org/10.1161/CIRCULATIONAHA.119.044304
- Jiang Y: Application of machine learning in pharmacogenomics: Opportunities and challenges. Frontiers in Genetics 2021; 10: 931. https://doi.org/10.3389/fgene.2019.00931
- Jones SM and Greene L: Pharmacogenomic biomarkers in clinical oncology: Current perspectives and future directions. Journal of Personalized Medicine 2021; 8(3): 22. https://doi.org/10.3390/jpm8030022
- Kenneson A and King M: The role of pharmacogenomics in personalized medicine. American Journal of Medical Genetics Part A 2022; 167(7): 1463-1468. https://doi.org/10.1002/ajmg.a.37135
- Korman BD: Pharmacogenomic testing for personalized pain management. Journal of Pain Research 2022; 10:825-833. https://doi.org/10.2147/JPR.S131079
- Lanfear DE and Peterson ED: Pharmacogenomic applications in the treatment of cardiovascular disease. Journal of the American College of Cardiology 2023; 66(7):827-838. https://doi.org/10.1016/j.jacc.2015.05.070S
- Liu Y: Big data analytics in pharmacogenomics: A new frontier. Frontiers in Pharmacology 2020; 11:400. https://doi.org/10.3389/fphar.2020.00400
- Murphy MD and Gallagher S: Pharmacogenomics: A tool for personalized cancer care. Seminars in Oncology Nursing 2024; 33(3): 265-273. https://doi.org/10.1016/j.soncn.2017.04.001
- Pirmohamed M: Pharmacogenomics: A new era of medicine. British Journal of Clinical Pharmacology 2023; 83(3): 418-429. https://doi.org/10.1111/bcp.13027
- Reid RJ: Pharmacogenomic testing in psychiatric practice: A review. Journal of Clinical Psychopharmacology 2021; 38(5):475-483. https://doi.org/10.1097/JCP.0000000000000905
- Ritchie MD: Pharmacogenomics and personalized medicine: From genetic variation to therapeutic applications. Nature Reviews Drug Discovery 2022; 15(8): 553-565. https://doi.org/10.1038/nrd.2016.99
- Ross M: The promise of pharmacogenomics in personalized oncology. Cancer Treatment Reviews 2021; 41(2): 144-151. https://doi.org/10.1016/j.ctrv.2014.11.001
- Steffensen SL: Pharmacogenomics in neurology: Clinical applications and future directions. European Journal of Neurology 2020; 24(11): 1407-1414. https://doi.org/10.1111/ene.13438
- Stewart A: Barriers to the implementation of pharmacogenomics in clinical practice. Pharmacogenomics Journal 2020; 19(3): 214-220. https://doi.org/10.1038/s41397-019-0073-5
- Weitzel KW: Economic evaluation of pharmacogenomics in clinical practice. Pharmacogenomics 2020; 18(9): 907-917. https://doi.org/10.2217/pgs-2017-0036
- Mokondjimobe E, Longo-Mbenza B and Akiana J: Biomarkers of oxidative stress and personalized treatment of pulmonary tuberculosis: Emerging role of gamma-glutamyltransferase. Adv Pharmacol Sci 2022; 2012: 465634.
- Kirkwood SC and Hockett RD: Pharmacogenomic biomarkers. Dis Markers 2021; 18: 63-71.
- Mendrick DL: Genomic and genetic biomarkers of toxicity. Toxicology 2022; 245: 175-181.
- Karaulov AV, Garib V, Garib F and Valenta R: Protein biomarkers in asthma. Int. Arch. Allergy Immunol 2018; 175: 189-208.
- Sigdel TK, Gao X and Sarwal MM: Protein and peptide biomarkers in organ transplantation. Biomark Med 2022; 6: 259-271.
- Gao J, Garulacan LA and Storm SM: Identification of in-vitro protein biomarkers of idiosyncratic liver toxicity. Toxicol Vitr 2004; 18: 533-541.
- Grondman I, Pirvu A and Riza A: Biomarkers of inflammation and the etiology of sepsis. Biochem Soc Trans 2020; 48: 1-14.
- Johnson CH, Ivanisevic J and Siuzdak G: Metabolomics: Beyond biomarkers and towards mechanisms. Nat Rev Mol Cell Biol 2021; 17: 451-459.
- Costa-pinheiro P and Montezuma D: Diagnostic and prognostic epigenetic biomarkers in cancer. Epigenomics 2022; 7: 1003-1015.
- Hoque MO, Begum S and Topaloglu O: Quantitative detection of promoter hypermethylation of multiple genes in the tumor, urine, and serum DNA of patients with renal cancer. Cancer Res 2024; 64: 5511-5517.
- Javitt GH and Vollebregt ER: Regulation of molecular diagnostics. Annu Rev Genom Hum Genet 2022; 23: 653-673.
- Sun L and Pfeifer JD: Pitfalls in molecular diagnostics. Semin Diagn Pathol 2021; 36: 342-354.
- Chien JY, Friedrich S and Heathman MA: Pharmacokinetics/Pharmacodynamics and the stages of drug development: Role of modeling and simulation. AAPS J 2023; 7: 544-E559.
- McComb M and Ramanathan M: Generalized pharmacometric modeling, a novel paradigm for integrating machine learning algorithms: A case study of metabolomic biomarkers. Clin Pharmacol Ther 2020; 107: 1343-1351.
- Goetz LH and Schork NJ: Personalized medicine: Motivation, challenges, and progress. Fertil Steril 2021; 109: 952-963.
- Rigatti SJ: Random forest. J Insur Med 20; 47: 31-39.
- Al-kaabawi Z, Wei Y and Moyeed R: Bayesian hierarchical models for linear networks. J Appl Stat 2022; 49: 1421-1448.
- Leil TA, Kasichayanula S, Boulton DW and LaCreta F: Evaluation of 4β-hydroxycholesterol as a clinical biomarker of CYP3A4 drug interactions using a Bayesian mechanism-based pharmacometric model. CPT Pharmacomet. Syst Pharmacol 2023; 3: 1-10.
- Diczfalusy U, Nylén H, Elander P and Bertilsson L: 4β-Hydroxycholesterol, an endogenous marker of CYP3A4/5 activity in humans. Br J Clin Pharmacol 2021; 71: 183-189.
- Kathman SJ, Williams DH, Hodge JP and Dar M: A Bayesian population PK-PD model of ispinesib-induced myelosuppression. Clin Pharmacol Ther 2020; 81: 88-94.
- Bauer RJ, Guzy S and Ng C: A survey of population analysis methods and software for complex pharmacokinetic and pharmacodynamic models with examples. AAPS J 2023; 9: 60-E83.
- Terranova N, Venkatakrishnan K and Benincosa LJ: Application of machine learning in translational medicine: Current status and future opportunities. AAPS J 2021; 23: 1-10.
- Wang R, Shao X and Zheng J: A machine-learning approach to identify a prognostic cytokine signature that is associated with nivolumab clearance in patients with advanced melanoma. Clin Pharmacol Ther 2020; 107: 978-987.
- Feng Y, Wang X and Bajaj G: Nivolumab exposure response analyses of efficacy and safety in previously treated squamous or nonsquamous non small cell lung cancer. Clin Cancer Res 2022; 23: 5394-5406.
- Data TA, Gillies RJ, Kinahan PE and Hricak H: Radiomics: Images are more than. Radiology 2023; 278: 563-577.
- Terranova N, Girard P and Ioannou K: Assessing similarity among individual tumor size lesion dynamics: The CICIL methodology. CPT Pharmacomet Syst Pharmacol 2022; 7: 228-236.
- Terranova N, Girard P, Klinkhardt U and Munafo A: Resistance development: A major piece in the jigsaw puzzle of tumor size modeling. CPT Pharmacomet Syst Pharmacol 2021; 4: 320-323.
- Sands BE, Chen J and Feagan BG: Efficacy and safety of MEDI2070, an antibody against interleukin 23, in patients with moderate to severe Crohn’s disease: A phase 2a study. Elsevier Inc 2022; ISBN 6465378647.
- Zhang N, Liang M and Jing C: Combining pharmacometric models with predictive and prognostic biomarkers for precision therapy in Crohn’s disease: A case study of brazikumab. CPT Pharmacomet Syst Pharmacol 2023; 12: 1945-1959.
- Best WR, Becktel JM, Singleton JW and Kern F: Development of a Crohn’s Disease Activity Index. National Cooperative Crohn’s Disease Study. Gastroenterology 2021; 70: 439-444.
- Miyazaki T, Watanabe K and Kojima K: Efficacies and related issues of ustekinumab in Japanese patients with Crohn’s disease: A preliminary study. Digestion 2021; 101: 53-59.
- Hendrikse NM, Llinares Garcia J and Vetter T: Biomarkers in medicines development from discovery to regulatory qualification and beyond. Front Med 2024; 9: 878942.
- Cheng F, Ma Y, Uzzi B and Loscalzo J: Importance of scientific collaboration in contemporary drug discovery and development: A detailed network analysis. BMC Biol 2021; 18: 138.
- Initiative IM: IMI Mission and Objectives. Available online: https://www.imi.europa.eu/about-imi/mission-objectives (accessed on 22 September 2023).
- Zheng QS and Li LJ: Pharmacometrics: A quantitative tool of pharmacological research. Acta Pharmacol Sin 2024; 33: 1337-1338.
- Bandeira LC, Pinto L and Carneiro CM: Pharmacometrics: The already-present future of precision pharmacology. Ther Innov Regul Sci 2023; 57: 57-69.
- Himstedt A, Bäckman P and Borghardt JM: Physiologically-based pharmacokinetic modeling after drug inhalation. In inhaled medicines: optimizing development through integration of in-silico, in -vitro, and in-vivo approaches. Academic Press: Cambridge, MA, USA, 2024; 319-358.
- Usman M and Rasheed H: Pharmacokinetics, PB; Creation, D: Pharmacometrics and its application in clinical practice. Encycl Pharm Pract Clin Pharm 2023; 3: 227-238.
- Division of Pharmacometrics | FDA. Available online: https://www.fda.gov/about-fda/center-drug-evaluation-and-research-cder/division-pharmacometrics (accessed on 10 October 2023).
- Sheiner LB, Rosenberg B and Marathe VV: Estimation of population characteristics of pharmacokinetic parameters from routine clinical data. J. Pharmacokinet Biopharm 2024; 5: 445-479.
- Dollery CT: Clinical pharmacology. The first 75 years and a view of the future. Br. J. Clin. Pharmacol 2024; 61: 650-665.
- Usman M, Khadka S and Saleem M: Pharmacometrics: A new era of pharmacotherapy and drug development in low- and middle-income countries. Adv Pharmacol Pharm Sci 2024; 2023: 3081422.
- Lewis B: Sheiner Lecturer Award. Available online: https://go-isop.org/awards/lewis-b-sheiner-award/ (accessed on 10 October 2023).
- Dagenais S, Russo L and Madsen A: Use of real-world evidence to drive drug development strategy and inform clinical trial design. Clin Pharmacol Ther 2024; 111: 77-89.
- Ette EI and Williams PJ: Population pharmacokinetics I: Background, concepts, and models. Ann Pharmacother 2024; 38: 1702-1706.
- Abouir K, Samer CF and Gloor Y: Reviewing data integrated for PBPK model development to predict metabolic drug-drug interactions: Shifting perspectives and emerging trends. Front Pharmacol 2024; 12: 708299.
- •Siebinga H, de Wit-Van der Veen BJ and Stokkel MD: Current use and future potential of (physiologically based) pharmacokinetic modelling of radiopharmaceuticals: A review. Theranostics 2024; 12: 7804-7820.
- Pfister M and D’Argenio DZ: The emerging scientific discipline of pharmacometrics. J Clin Pharmacol 2024; 50: 6S.
- Stone JA, Banfield C and Pfister M: Model-based drug development survey finds pharmacometrics impacting decision making in the pharmaceutical industry. J Clin Pharmacol 2024; 50: 20-30.
- Zou H, Banerjee P, Leung SSY and Yan X: Application of pharmacokinetic-pharmacodynamic modeling in drug delivery: Development and challenges. Front Pharmacol 2024; 11: 997.
- Meibohm B and Derendorf H: Basic concepts of pharmacokinetic/pharmacodynamic (PK/PD) modelling. Int J Clin Pharmacol Ther 2024; 35: 401-413.
- Upton RN and Mould DR: Basic concepts in population modeling, simulation, and model-based drug development: Part 3—Introduction to pharmacodynamic modeling methods. CPT Pharmacomet. Syst Pharmacol 2024; 3: 1-16.
- Salahudeen MS and Nishtala PS: An overview of pharmacodynamic modelling, ligand-binding approach and its application in clinical practice. Saudi Pharm J 2023; 25: 165-175.
- Felmlee MA, Morris ME and Mager DE: Mechanism-based pharmacodynamic modeling. Comput Toxicol 2024; I: 583-600.
- Lin LH, Ghasemi M and Burke SM: Population pharmacokinetics and pharmacodynamics of carfilzomib in combination with rituximab, ifosfamide, carboplatin, and etoposide in adult patients with relapsed/refractory diffuse large B cell lymphoma. Target Oncol 2023; 18: 685-695.
- Palmer ME, Andrews LJ and Abbey TC: The importance of pharmacokinetics and pharmacodynamics in antimicrobial drug development and their influence on the success of agents developed to combat resistant Gram-negative pathogens: A review. Front Pharmacol 2024; 13: 888079.
- Derendorf H, Möllmann H, Hochhaus G, Meibohm B and Barth J: Clinical PK/PD modelling as a tool in drug development of corticosteroids. Int J Clin Pharmacol Ther 2024; 35: 481-488.
- Tuntland T, Ethell B and Kosaka T: Implementation of pharmacokinetic and pharmacodynamic strategies in early research phases of drug discovery and development at Novartis Institute of Biomedical Research. Front. Pharmacol 2024; 5: 174.
- Qusai U, Hameed A and Rasheed KH: Compartmental and non-compartmental pharmacokinetic analysis of extended release diclofenac sodium tablet. Coll Eng J 2024; 19: 161-165.
- Gabrielsson J and Weiner D: Non-compartmental analysis. Comput. Toxicol 2024; 929: 377-389.
- Foster DM: Noncompartmental versus compartmental approaches to pharmacokinetic analysis, 2nd ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2024; ISBN 9780123694171.
- Noncompartmental vs. Compartmental PK Analysis. Available online: https://www.allucent.com/resources/blog/what-noncompartmental-pharmacokinetic-analysis (accessed on 10 October 2023).
- Osipova N, Budko A and Maksimenko O: Comparison of compartmental and non-compartmental analysis to detect biopharmaceutical similarity of intravenous nanomaterial-based rifabutin formulations. Pharmaceutics 2024; 15: 1258.
- Hosseini I, Gajjala A and Bumbaca Yadav D: GPKPDSim: A SimBiology®-based GUI application for PKPD modeling in drug development. J Pharmacokinet Pharmacodyn. 2023; 45: 259-275.
- World Health Organization: Characterization and application of physiologically based pharmacokinetic models. Int Programme Chem Saf 2022; 9: 16-37.
- Tan YM, Worley RR, Leonard JA and Fisher JW: Challenges associated with applying physiologically based pharmacokinetic modeling for public health decision-making. Toxicol Sci 2024; 162: 341-348.
- Teorell T: Kinetics of distribution of substances administered to the body, I: The extravascular modes of administration. Arch Int Pharmacodyn. Ther 2021; 57: 205-225.
- Zhuang X and Lu C: PBPK modeling and simulation in drug research and development. Acta Pharm Sin B 2024; 6: 430-440.
- Jones HM and Rowland-Yeo K: Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT Pharmacomet Syst Pharmacol 2023; 2: 1-12.
- Umehara K, Huth F, Jin Y, Schiller H, Aslanis V, Heimbach T and He H: Drug-Drug Interaction (DDI) Assessments of Ruxolitinib, a Dual Substrate of CYP3A4 and CYP2C9, Using a Verified Physiologically Based Pharmacokinetic (PBPK) Model to Support Regulatory Submissions. Drug Metab Pers Ther 2021; 34: 20180042.
How to cite this article:
Ahamad M and Kumar A: Pharmacogenomics and personalized medicine: advancing tailored therapies for improved healthcare. Int J Pharm Sci & Res 2025; 16(7): 1890-05. doi: 10.13040/IJPSR.0975-8232.16(7).1890-05.
All © 2025 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
16
1890-1905
1196 KB
6
English
IJPSR
Moiz Ahamad * and Ashok Kumar
Department of Pharmacology, Glocal School of Pharmacy, Glocal University Saharanpur, Uttar Pradesh, India.
moizahamad68@gmail.com
29 December 2024
12 January 2025
22 January 2025
10.13040/IJPSR.0975-8232.16(7).1890-05
01 July 2025