Hari College Of Pharmacy.
Pharmacogenomics, which examines the influence of genetic variations on drug reactions, has transformed personalized medicine. By customizing treatment to a person's distinct genetic profile, and pharmacogenetic data to fingerprint the pharmacological treatment of neuropsychiatric late-life conditions throughout the analysis of metabolizing enzymes and transporters of psychotropic drugs, mainly those of the cytochrome P450 (CYP) family. Pharmacodynamic response measures as treatment effects mediated through targets (i.e., receptors in the brain) may also contribute to this image. Drug treatment outcome represents a complex phenotype, encoded by dozens, if not hundreds, of genes, and affected by many environmental factors; therefore, we will almost always see a gradient of response. Phenotyping assays of blood enzyme activities (if feasible) are generally more successful than DNA genotyping for predicting unequivocal outcomes of drug therapy in each and every patient. Genotyping to predict drug disposition, efficacy, toxicity, and clinical outcome has been proposed, but the success of genotyping in individualized drug therapy currently appears unlikely because of the many shortcomings (frequency of DNA variant sites, ethnic difference differences, admixture), Genomics is an important tool in basic research; yet, it is unrealistic to include genotyping within the realm of tests available to the practicing clinician in the foreseeable future. Personalized medicine involves the selection of the safest and most effective pharmacological treatment based on the molecular characteristics of the patient. In the case of anticancer drugs, tumour cell alterations can have a great impact on drug activity and, in fact, most Pharmacogenomics studies how genetic affect drug responses and has revolutionized personalized medicine. Pharmacogenomics into clinical practice allows healthcare professionals to make better informed treatment choices, resulting in enhanced patient outcomes. according to a person's specific genetic makeup, pharmacogenomic targeted therapy seeks to enhance treatment effectiveness and reduce negative side effects. This review offers an extensive summary of the pharmacological results related to pharmacogenomic targeted therapy, emphasizing its advantages, obstacles, and future prospects.
History:
Pharmacogenomics has become an essential resource for enhancing drug treatments. By discovering genetic variations that affect drug responses, healthcare professionals can choose the best treatment approach for each patient. Pharmacogenomic directed therapy has demonstrated potential in numerous therapeutic fields, such as oncology, psychiatry, and cardiovascular medicine. Both lack of efficacy and toxicity of therapeutic agents are among the major obstacles to improving survival and quality of life of cancer patients. These are critical problems for all therapies but particularly for cancer treatment because most anticancer drugs are only effective in a minority of patients and have a narrow therapeutic index that frequently leads to severe toxicities and even death. By providing customized treatment plans based on each patient's own genetic profile, pharmacogenomics is transforming the medical industry. The development of targeted medicines for a number of illnesses, most notably cancer, has resulted from this strategy. Here are a few instances of recently developed targeted medications and illness treatments. Everything from human behavior to health is influenced by genetics. Variation in genetics is another factor contributing to the observed disparities among humans. Individual differences in the drug's pharmacokinetic and pharmacodynamic characteristics can also be attributed to genetic variances. In certain people, these genetic differences may be the cause of negative medication responses. By linking a medicine's toxicity or efficacy to gene expression or polymorphism, pharmacogenomics examines how genetic diversity affects pharmacological response.
<a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250530171324-1.png" target="_blank">
<img alt="Progress in Pharmacogenomics and pharmacogenetics.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250530171324-1.png" width="150">
</a>
Fig. 1 Progress in Pharmacogenomics and pharmacogenetics
Gene Variability in Drug Transporters:
Drug Transporter Genetic Polymorphism Drug transporters are membrane-spanning proteins that let drugs pass through the gastrointestinal tract, be excreted into bile and urine, and pass across the blood-brain barrier. Drug concentrations at the site of action and drug dispersion are impacted by genetic variations in the drug transport proteins. One transport protein that demonstrates genetic variation is P-GP (P-glycoprotein).
Function:
Genetic Variability and Drug Metabolizer Mutations
Different subgroups within the population are created by genetic variations in drug-metabolizing enzymes, which vary in their capacity to carry out certain drug biotransformation activities. Genetic variations in drug metabolism are caused by variations in alleles for genes encoding the enzymes involved in drug metabolism. For instance, CYP2D6 genetic variation results in reduced metoprolol excretion. ADR results from this.
Phenotype
A phenotype is any of an organism's physical, physiological, biochemical, or behavioral traits. Any specific characteristic seen following medication administration is referred to as a phenotype in clinical pharmacology (Fig-I). But how simple is it to detect a phenotypic with certainty Phenocopy, or the same phenotype occurring in two people due to distinct genes or environmental variables contributing to that feature, is one issue. Many pharmacogenomics researchers are looking for phenotype-genotype associations in order to prevent the majority of serious adverse drug reactions (ADRs). If a phenotype (trait) is consistently linked to a particular genotype (an individual's genetic sequence), then a DNA test conducted prior to drug administration should prevent nearly all serious ADRs.
Genotype
The late 1990s saw the introduction of high throughput resequencing of every given gene, which sparked the discovery of an incredibly high number of DNA variant sites As a result, our entire understanding of "mutation" has evolved. The phrase "nucleotide substitution" has mostly been superseded by the word "single nucleotide polymorphism" (SNP). SNPs make about 90 to95% of all variation sites, whereas the remaining portion is made up of insertions or deletions (indels) of any number of bases, ranging from one to more than one million. The section of DNA that codes for a functional product is called a gene; it runs from the 5′ most regulatory element to the 3′ most regulatory element that surrounds the actual area that has been transcribed. Consequently, certain genes overlap, a gene may even be found inside another gene, and one gene may have many SNPs that are different from those found in the reference (consensus) allele of the same gene.
Table I Phenotypic Issues That Impact Research of Medication Phenotype-Genotype Associations
Issues |
Example |
Drug substrate specificity overlap |
Enzymes that break down drugs, transporters of drugs, chaperones, receptors, ion channels, and transcription factors |
Drug-drug interactions, many arising from pharmacogenetic differences |
Drugs or other external substances that induce or inhibit enzymes |
Developmental And environmental factors that can affect each step in drug disposition |
Nutrition, age, sex, health and illness conditions, smoking, renal tubular excretion, exposure to chemicals, and over-the-counter natural items. |
medications with many enzymes that are processed by genes that show functional Variations |
The enzymes that bio-transform benzene [C6H6] are at least 14 and are encoded by as many genes as possible. |
Illustrations of Pharmacogenomics in Practice
Instruction For Analysis:
Absence of Pharmacogenomics Education: It might be challenging to interpret and apply PGx testing since many healthcare professionals lack sufficient pharmacogenomics training.
Algorithms for Limited Dosage: Clinical practice must be guided by precise dose recommendations based on PGx findings.
Information Complexity: Patients and medical professionals may find the intricacy of pharmacogenomics data to be daunting.
Infrastructure and Data Analysis
Pharmacogenetics
Exciting studies of gene-drug and gene-environment interactions have appeared over the past seven decades. Each of these apparent success stories represents a predominantly mono-genic trait in which the functional consequence of the gene was recognized, e.g. phenylthiol-urea nontaster, atypical serum cholinesterase, glucose-6-phosphate dehydrogenase deficiency, isoniazid slow N-acetylation, debrisoquine oxidation poor metabolizer, paraoxons low activity and thiopurine methyltransferase (TPMT) deficiency. Except for the first example (taste test), all the others represent a trait described as high versus low (to nil) drug- metabolizing enzyme (DME) activity (thereby clearing any drug substrate more slowly), with the high activity designated as the wild-type (reference, consensus) normally-occurring trait.
Pharmacogenetics And Psychotropic Medications Disorders
Almost one in five elderly people living in the community in the United States took psychotropic medicines, mostly antidepressants and then antianxiety drugs. Almost 2.5 million (7.5%) and almost 3 million (9.1%) of the elderly took antidepressants. Among these, people affected by dementia assumed greater number of CNS-active medications including anti- psychotics, anxiolytics, and antidepressants. The CYP2D6 metabolized the great number of CNS drugs including 80% of antidepressants.
Biomarkers Compound:
Biomarkers are biological molecules that indicate a normal or aberrant process, a condition, or a disease and can be discovered in blood, other bodily fluids, or tissues. They are employed to diagnose, track, and forecast the course of a disease or the effectiveness of a therapy.
Types:
Examples:
Imaging Biomarkers: Imaging methods, such PET or MRI scans, can track the course of a disease or the effectiveness of a treatment.
Uses for Biomarkers:
Necessity Of Biomarkers for Individualized Care in The Elderly Population
In modern physical medicine, personalized treatment is quickly becoming a reality. The development of pharmacogenetically tailored treatment regimens that account for interindividual genetic differences is ongoing, even in older subjects, given the substantial advancements in our understanding of the biochemical, genetic, and neurobiological processes underlying major mental disorders. The avoidance of the start or progression of sickness and the reduction of the risk of harm associated with more complicated treatment regimens are thus added to the specific goals of therapeutic intervention. In psychiatry, there are currently no biomarkers measurable biological traits that indicate pathogenic processes or treatment responses or other risk markers that can be used to develop profiles that improve therapy selection and prediction.
<a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250530171324-0.png" target="_blank">
<img alt="Fig. 2.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250530171324-0.png" width="150">
</a>
Fig. 2
Novel predictive biomarkers
The challenge of precisely determining the usefulness of the discovered markers or tactics for patients and healthcare systems is a significant constraint in pharmacogenomics. General recommendations have been proposed with a focus on omics-based markers, and the amount of evidence needed to prove that a marker is clinically valuable and should be adopted for routine usage has been examined. Drug selection and dosage for the majority of cancer treatments still adhere to standard operating procedures based on the general population, ignoring the unique traits of individual patients and tumor types. A common standard dose is administered to all patients for many targeted medications, and for many cytotoxic drugs, dose modifications are solely made by considering the patient's body surface area, which is frequently an incorrect dose indication. Finding biomarkers that predict the fate of anticancer drugs could significantly alter the situation, but there are significant obstacles to overcome and the work is not straightforward.
Personalized Medicine
Adapting Healthcare to Meet Individual Requirements Personalized medicine, referred to as precision medicine, is a healthcare approach that considers an individual's distinct traits, including genetic makeup, medical background, lifestyle choices, and environmental influences, to deliver customized treatment and prevention methods.
Key Aspects of Tailored Medicine
Research on Populations and Customized Drug Treatment
Many of the hundreds of phenotype-genotype association studies of large populations that look into pharmacogenetic disorders or other complex diseases in humans have found that a certain trait is statistically significantly correlated with one or more SNPs. The results are shown as odds ratios (ORs) and confidence intervals (CIs). Consequently, it is evident that a single nucleotide site will practically never be effectively used clinically to predict and reduce each person's risk of ADRs for the reasons already mentioned. The average individual risk in the entire population can be inferred from large research, but the exact risk estimate for a given patient cannot. These outliers have been uncovered by the field of pharmacogenetics, and they are indeed a major factor driving the demand for individualized treatment.
Beyond the Genes
The study of heritable differences in medication response is known as pharmacogenetics. Pharmacogenomics, one of the Human Genome Project's byproducts, was just acknowledged as a field of study that is marginally distinct from pharmacogenetics. Pharmacogenomics describes how medications affect biological pathways and processes by interacting with the entire expression output of the genome; it has frequently been said that this field will aid in the development of new medications. A variety of recently developed "omics" disciplines of study are included in Table II.
Research on Populations and Customized Drug Treatment
Many of the hundreds of phenotype-genotype association studies of large populations that look into pharmacogenetic disorders or other complex diseases in humans have found that a certain trait is statistically significantly correlated with one or more SNPs. The results are shown as odds ratios (ORs) and confidence intervals (CIs). Consequently, it is evident that a single nucleotide site will practically never be effectively used clinically to predict and reduce each person's risk of ADRs for the reasons already mentioned. The average individual risk in the entire population can be inferred from large research, but the exact risk estimate for a given patient cannot. These outliers have been uncovered by the field of pharmacogenetics, and they are indeed a major factor driving the demand for individualized treatment.
Table: II
New Phrase |
Method |
Research tool |
Customized treatment |
Genomicsa |
SNP analysis |
Yes |
Unlikely |
Transcriptomicsb |
cDNA, microarray |
Yes |
Limited |
Proteomicsc |
Protein analysis |
Yes |
Limited |
Metabolomics |
Metabolic profiling |
Yes |
Shows promise |
Phenomics |
Trait analysis |
Yes |
Shows promise |
Benefits of Pharmacogenomics Targeted Therapies:
Ethical Challenges:
Pharmacogenomics has a number of significant obstacles that fall into the general categories of practical execution, public policy, and ethical concerns.
Pharmacological implementation in clinical changes:
The US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have authorized 190 and 155 medications, respectively, with labels that discuss the significance of genetic diversity and the growing clinical use of pharmacogenomic biomarkers. seven clinical facilities in Europe to examine how pharmacogenomic testing affects treatment results. 8100 individuals will be recruited in total, and 40 clinically significant PGx indicators from 13 significant pharmacogenes will be examined. Pharmacokinetically guided therapy will be administered to patients in one arm of the experiment, while traditional physiological and clinical measures will be used to treat patients in the other arm.
Limitations
Result and Outcomes
a. Pharmacological Results
Pharmacogenomics Enhances Treatment Results in various ways:
CONCLUSION
Personalized medication therapy is a primary goal of pharmacogenomics, and this article has explored why it appears unlikely to be successful in assisting the practicing physician in for the foreseeable future. The doctor wants to be able to confidently predict the phenotype (efficacy, toxicity, or therapeutic failure) in each of the patients treated with medication. The best genotyping and phenotyping assays now on the market may provide two or three false-negative results and two or three false-positive results for some patients; this is insufficient for the clinician. The decision-making process for choosing drugs represents a greater level of sophistication based on more technological discoveries and improvements, including CYP and drug transporter polymorphisms. In general, phenotyping using probe medicines to identify phenotype-genotype relationships has not been successful. It has been suggested that genotyping can predict medication disposition, efficacy, toxicity, and clinical outcome. However, due to the numerous drawbacks and complications discussed here, the success of genotyping in customized drug therapy currently seems doubtful. Despite being a valuable tool in fundamental research, genotyping is not likely to be included in the range of tests that practicing clinicians can use in the near future. The domains of proteomics and transcriptomics are comparable. There may be ways to predict and reduce each patient's risk for adverse drug reactions (ADRs) as well as the beginning and course of their illness using the recently developed domains of metabonomic and phenomics.
REFERENCES
Sharif*, Indu Saini, Dr. Kshitiz Aggarwal, Review on Pharmacological Outcomes for Pharmacogenomics Targeted Therapy, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 4914-4926. https://doi.org/10.5281/zenodo.15553385