Improving Perscriptions Through The P450 Superfamily

Jaya Sra
34 min readNov 30, 2023

Introduction

A 76-year-old female presented with MDD(major depressive disorder) had an inadequate response to antidepressants, post-traumatic stress disorder, obsessive-compulsive disorder, generalized anxiety disorder, pseudobulbar affect, insomnia, restless leg syndrome, constipation, back pain, gastroesophageal reflux disease, hyperlipidemia, and glaucoma. The patient's main complaint was the fact that she still had uncontrolled depression despite her multiple attempts with various antidepressants.

Symptoms of major depressive disorder

The medications she was taking included bupropion, escitalopram, and venlafaxine. All of these resulted in the patient experiencing ADEs(adverse drug reactions) and inadequate depression control. The patient was additionally treated with other medications such as quetiapine. But even after following her prescribed treatment plan nothing was working.

Recognizing the complexity of the case, the patient had enrolled in the Program of All-Inclusive Care for the Elderly (PACE), a model that brings together pharmacists and other healthcare professionals to address medication-related issues comprehensively. In this context, pharmacogenomics testing was employed to shed light on the genetic factors influencing the patient's response to medications.

After performing PGx(pharmacogenomic) testing the clinical pharmacist discovered three clinically significant drug-drug interactions (DDIs) involving CYP26, which is a member of the P450 superfamily. Additional interactions and genetic components influencing the patient's response to medication were discovered through further investigation. This finding highlighted the value of customization of prescriptions and demonstrated that patients with complex medical histories might not benefit from a one-size-fits-all approach to pharmacotherapy.

Beyond just the particular patient, this case is significant because it illustrates the wider applications of personalized medicine in mental health and the need for a more complex knowledge of drug interactions based on genetic factors. The PACE model's inclusion of PGx testing highlights the value of interdisciplinary teamwork in improving treatment regimens. In this article, I will discuss the aspects of prescription personalization and focus on the P450 superfamily which has a massive effect on drug metabolism.

Prescription Medication Impact

Prescription drugs are fundamental to the structure of contemporary healthcare, and they have a significant and complex impact on public health that goes well beyond the parameters of specific treatment regimens.

The central function that prescription drugs play in the management and treatment of disease is at the heart of this extensive influence. These pharmaceuticals are essential components of the medical arsenal because they provide symptom relief, slow the course of diseases, and prevent complications. Medications that treat chronic conditions like diabetes and hypertension or fight infectious diseases like antibiotics are essential for improving health outcomes and lowering the cost of illness in society.

Prescription medication is a major component of many public health interventions, particularly those focused on disease prevention. Keystones in the management and avoidance of infectious diseases include vaccinations, antiviral medications, and antibiotics. Their use in vaccination drives and epidemic containment programs emphasizes how crucial they are to maintaining public health and stopping the spread of infectious illnesses.

The management of chronic diseases is yet another important aspect of how prescription drugs affect public health. Customized drugs for ailments such as autoimmune diseases, respiratory ailments, and cardiovascular diseases play a major role in preserving health and averting worsening of symptoms. In addition to improving the quality of life for those who are afflicted, efficient management of chronic diseases also relieves the burden on healthcare systems that must deal with the long-term effects of untreated or inadequately managed conditions.

Prescription drugs play a crucial role in health promotion and education campaigns, as well as public health initiatives that go beyond clinical settings. They offer practical resources for illness awareness, health screenings, and preventive care. By integrating drugs into more comprehensive public health initiatives, societies enable people to make knowledgeable health decisions and promote a proactive well-being culture.

Prescription drug effects are especially noticeable in emergency and critical care situations. These drugs are essential for the quick response to life-threatening situations; they range from epinephrine for severe allergic reactions to clot-busting drugs for strokes. Their vital role in emergency medicine and critical care is highlighted by the fact that their prompt administration can mean the difference between life and death

Another area where prescription drugs have a major impact on public health is maternal and child health. Immunizations, antibiotics, and prenatal vitamins are crucial parts of a complete healthcare package for expectant mothers and their unborn children. These drugs improve the health of entire generations by tackling health problems early in life and laying the groundwork for a healthy future.

Prescription drugs have a beneficial effect on mental health, an area that is frequently neglected. Psychiatric drugs, such as mood stabilizers, antipsychotics, and antidepressants, are essential for treating mental health conditions. These drugs help to improve general societal well-being and de-stigmatize mental health conditions by offering efficient treatment options.

The therapeutic landscape is expanded by ongoing advancements in treatment, which are fueled by research and development in the pharmaceutical industry. The positive impact of prescription medicine on public health is further enhanced by the availability of new and more effective medications, which give hope to patients with previously incurable conditions.

Furthermore, the effects of prescription drugs are closely linked to economic factors. Stability in the economy is facilitated by a workforce that is healthy and productive and is kept that way through efficient treatment and disease management. The economic benefits that flow through communities and countries when people have access to essential medications highlight the comprehensive role that prescription drugs play in promoting the well-being of individuals and society as a whole.

History of Pharmaceuticals/Perscriptions

The history of pharmaceutical interventions in public health is a captivating narrative that mirrors the evolution of medical science, societal needs, and the relentless pursuit of healthier communities.

Ancient and Classical Periods

In antiquity, pharmaceutical interventions were rooted in herbal remedies, with civilizations like Egypt, Mesopotamia, China, and Greece harnessing the healing properties of plants. The foundations of pharmacology were laid as ancient societies developed a nuanced understanding of the medicinal properties of natural substances.

Middle Ages to Renaissance:

The Middle Ages witnessed the preservation and expansion of pharmaceutical knowledge in the Islamic world. Alchemical traditions paved the way for the systematic study of pharmacology, with alchemists contributing to the refinement of techniques for extracting medicinal compounds.

17th to 19th Centuries

The Scientific Revolution brought about the emergence of modern pharmacology, challenging traditional beliefs about medicine. The isolation of medicinal compounds from plants gained momentum. The discovery of quinine and its application in malaria treatment marked a pivotal moment, addressing a disease with significant public health implications.

20th Century

The 20th century heralded transformative breakthroughs in pharmaceutical interventions. The discovery of antibiotics, starting with penicillin, revolutionized the treatment of bacterial infections. Vaccination programs became widespread, leading to the eradication of smallpox and significant control of diseases like polio and measles.

Late 20th Century to Present

The latter part of the 20th century and the beginning of the 21st century brought about groundbreaking developments. Antiretroviral drugs transformed HIV/AIDS from a fatal condition to a manageable chronic one. Genomic medicine and personalized therapies emerged, allowing for tailored drug treatments based on individual genetic makeup.

The ongoing battle against emerging infectious diseases, exemplified by outbreaks like SARS, H1N1, and COVID-19, showcases the adaptability of pharmaceutical interventions. Rapid vaccine development, notably with mRNA COVID-19 vaccines, underscores the agility of the pharmaceutical industry in responding to global health crises.

Global Health Initiatives and Challenges

International efforts, such as the World Health Organization’s Essential Medicines List and global health partnerships, have aimed to improve access to essential pharmaceutical interventions, particularly in low-income countries. However, challenges persist, including the complexity of drug development, concerns about access and affordability, and ethical considerations in clinical trials.

The Roles Of Pharmaceuticals In Managing And Treating A Diverse Range Of Health Conditions

Infectious diseases, long-standing adversaries to human health, have faced formidable challenges through the development of pharmaceutical interventions. Antibiotics, antivirals, and antifungals have become indispensable tools in the fight against pathogens. From the groundbreaking discovery of penicillin to the development of antiretroviral drugs for managing HIV/AIDS, pharmaceuticals have played a vital role in controlling the spread of infectious diseases and improving outcomes for affected individuals.

The management of chronic conditions has been revolutionized by pharmaceuticals, offering a diverse array of medications tailored to specific diseases. Cardiovascular diseases, diabetes, and autoimmune disorders are among the myriad conditions benefitting from medications such as statins, antihypertensives, insulin, and immunosuppressants. These pharmaceutical interventions not only control symptoms but also contribute to preventing complications, enhancing the quality of life for those living with chronic health conditions.

Pain management, a critical aspect of healthcare, relies heavily on pharmaceuticals. Analgesics and anti-inflammatory medications, both over-the-counter and prescription, provide relief for individuals experiencing acute or chronic pain. The responsible use of opioids for severe pain underscores the delicate balance between effective pain management and mitigating the risks of substance abuse.

In the realm of mental health, psychiatric medications are instrumental in managing a spectrum of disorders. Antidepressants, antipsychotics, mood stabilizers, and anxiolytics regulate neurotransmitter imbalances, offering effective treatment for conditions such as depression, schizophrenia, bipolar disorder, and anxiety disorders. These medications not only alleviate symptoms but also facilitate the recovery and well-being of individuals grappling with mental health challenges.

Cancer treatment has witnessed groundbreaking advancements through pharmaceutical interventions. Chemotherapy targeted therapies, and immunotherapies are tailored to inhibit the growth of cancer cells, induce apoptosis, or modulate the immune system to recognize and attack cancer cells. These treatments represent a paradigm shift in the approach to cancer care, offering more targeted and effective options with reduced side effects.

Respiratory conditions, including asthma and chronic obstructive pulmonary disease (COPD), benefit significantly from pharmaceutical interventions. Bronchodilators, corticosteroids, and anti-leukotriene agents help control symptoms, improve lung function, and enhance overall respiratory health. These medications empower individuals to manage their conditions effectively and lead more active lives.

Vaccination stands as a cornerstone of preventive medicine, demonstrating the proactive role of pharmaceuticals in averting disease outbreaks. Immunizations against diseases like measles, polio, influenza, and most recently, COVID-19, showcase the industry’s commitment to preventing the onset and spread of illnesses, safeguarding communities worldwide.

In the era of precision medicine, pharmaceuticals are increasingly tailored to specific genetic and rare diseases. Orphan drugs, designed for conditions with a limited patient population, exemplify the commitment to addressing niche health challenges and providing targeted therapies. These medications offer hope to individuals facing conditions that were once deemed untreatable.

Pharmaceutical interventions have also played a crucial role in responding to emerging infectious diseases. The rapid development of vaccines, particularly highlighted by the mRNA COVID-19 vaccines, exemplifies the industry’s ability to innovate and adapt to evolving health challenges. These advancements underscore the critical importance of pharmaceuticals in global health initiatives, addressing diseases that disproportionately affect low-income countries and striving towards health equity on a global scale.

Tailoring medical treatments for optimal health outcomes

In the dynamic landscape of modern healthcare, the significance of tailoring medical treatments for optimal health outcomes has emerged as a transformative approach, reshaping the way we perceive and deliver healthcare interventions.

At the heart of the significance lies the acknowledgment of the inherent diversity among individuals, both in terms of genetic makeup and biological variability. Tailoring medical treatments recognizes that each person is unique, possessing a distinct genetic profile that influences responses to medications and susceptibility to diseases. This individualized approach enhances the efficacy of treatments, aligning interventions with the specific needs and characteristics of the patient.

Optimizing safety profiles is another critical dimension of tailoring medical treatments. By understanding genetic predispositions and potential responses to medications, healthcare providers can minimize the risk of adverse reactions and side effects. This not only enhances patient safety but also contributes to a more favourable risk-benefit profile, ensuring that the chosen interventions align with the patient’s individual health profile.

Precision in disease management is a key outcome of tailoring medical treatments. The approach allows for a more accurate identification of the underlying causes of a disease, leading to targeted interventions that address the individual factors contributing to the condition. This precision in disease management translates to more effective treatment strategies, minimizing the guesswork and providing tailored solutions for optimal health outcomes.

Patient-centred care is a fundamental principle that tailoring medical treatments promotes. By actively involving patients in decisions about their healthcare, personalized medicine fosters a collaborative relationship between healthcare providers and individuals. This engagement empowers patients to take an active role in their treatment plans, contributing to improved adherence, satisfaction, and overall health outcomes.

Optimal resource utilization is a practical benefit of tailoring medical treatments. Moving away from the traditional trial-and-error approaches, personalized medicine reduces the need for multiple interventions before finding an effective treatment. This not only saves valuable time but also optimizes the utilization of healthcare resources, preventing unnecessary costs and potential complications.

Chronic disease management stands to gain substantial benefits from personalized medical treatments. The long-term health advantages are evident as interventions are fine-tuned based on individual responses over time. This approach minimizes the progression of diseases, prevents complications, and contributes to the overall well-being of individuals managing chronic conditions.

The field of pharmacogenomics represents a tangible manifestation of tailoring medical treatments based on genetic factors. By analyzing genetic variations influencing drug metabolism and response, healthcare providers can make informed decisions about the most suitable medications for an individual. This targeted approach improves treatment outcomes and minimizes adverse effects, exemplifying the potential of personalized medicine in achieving optimal health outcomes.

Innovations in cancer therapies, particularly in precision oncology, underscore the transformative impact of tailoring medical treatments. Targeted therapies and immunotherapies, designed to address specific molecular and genetic characteristics of tumours, have revolutionized cancer care. These advancements offer more effective and less toxic treatments, promising improved survival rates and enhanced quality of life for cancer patients.

Beyond individual care, the significance of tailoring medical treatments extends to inform public health strategies. Understanding population-specific genetic and environmental factors enables the development of customized public health interventions. This tailored approach optimizes the impact of health initiatives on diverse communities, addressing health disparities and promoting equitable access to healthcare resources.

Introduction To The Field Of Pharmacogenomics And Its Focus On Genetic Influences On Drug Response

Pharmacogenomics represents the cutting-edge intersection of pharmacology and genomics, embodying a revolutionary paradigm shift in the realm of personalized medicine. At its core, this field delves into the intricate and nuanced relationship between an individual’s genetic makeup and their response to pharmaceutical agents. Pharmacogenomics is not merely a scientific discipline; it is a transformative approach that seeks to unravel the genetic underpinnings of drug efficacy, toxicity, and overall therapeutic outcomes.

The primary focus of pharmacogenomics lies in deciphering how genetic variations among individuals contribute to the variability in drug response. This intricate interplay involves a meticulous examination of the diverse array of genes that influence drug metabolism, transport, and target interactions. By scrutinizing the genetic landscape, pharmacogenomic research aims to unveil the molecular intricacies that dictate whether a drug will be a therapeutic boon or an adverse event waiting to unfold.

One of the key tenets of pharmacogenomics is the recognition that individuals differ not only in their physiological characteristics but also in their genetic makeup, which can profoundly impact the pharmacokinetics and pharmacodynamics of drugs. This understanding has paved the way for the development of tailored therapeutic regimens that consider an individual’s genetic profile, allowing for more precise and efficacious treatment strategies.

The significance of pharmacogenomics is underscored by its potential to mitigate the unpredictable nature of drug responses, reducing adverse reactions and optimizing therapeutic outcomes. As researchers delve deeper into the vast expanse of the human genome, uncovering novel genetic markers and biomarkers, the field continues to evolve, presenting a wealth of opportunities to revolutionize the landscape of healthcare.

In essence, pharmacogenomics stands as a beacon of precision medicine, ushering in an era where therapeutic interventions are finely tuned to the unique genetic fabric of each patient. The promise of tailoring drug regimens based on an individual’s genetic blueprint holds immense potential for enhancing the safety, efficacy, and overall success of medical treatments, thereby heralding a new era in the trajectory of medicine.

Evolution Of Pharmacogenomics

Pharmacogenomics (PGx) has undergone a remarkable evolution over the years, representing a fascinating intersection of genetics, medicine, and technology. The journey of PGx can be traced back to the early days of genomic research when the Human Genome Project laid the foundation for understanding the intricacies of our genetic code. Since then, PGx has evolved through distinct phases, each marked by breakthroughs, challenges, and paradigm shifts.

Genomic Era (1990s-early 2000s)

The roots of PGx can be found in the completion of the Human Genome Project in 2003, a monumental achievement that provided a comprehensive map of the human genome. This milestone paved the way for researchers to identify genetic variations that influence drug response. Early PGx studies focused on candidate genes, such as those encoding drug-metabolizing enzymes, to understand how genetic variations could affect drug metabolism and efficacy.

Technological Advancements (mid-2000s)

The mid-2000s saw a surge in technological advancements, particularly in DNA sequencing technologies. This allowed for a shift from candidate gene studies to genome-wide approaches, enabling the identification of previously unknown genetic factors influencing drug response. High-throughput sequencing and genotyping technologies became instrumental in large-scale PGx studies, providing a more comprehensive understanding of the genetic landscape.

Pharmacogenomics in Clinical Practice (late 2000s-2010s)

As the understanding of pharmacogenomics deepened, efforts were made to translate these findings into clinical practice. This era witnessed the integration of PGx into drug development processes and clinical guidelines. Regulatory bodies started to recognize the importance of incorporating genetic information into drug labels, emphasizing the need for personalized medicine approaches. Institutions and healthcare providers began to adopt PGx testing to optimize drug selection and dosing.

Rise of Direct-to-Consumer Testing and Personal Genomics (2010s)

With the advent of direct-to-consumer genetic testing services, individuals gained unprecedented access to their genetic information, including pharmacogenetic data. This democratization of genetic testing raised awareness about PGx among the general public. However, it also brought about challenges related to the interpretation and communication of complex genetic information, as well as concerns about privacy and data security.

Integration of Informatics and Artificial Intelligence (2010s-present)

The current era is characterized by the integration of informatics, bioinformatics, and artificial intelligence (AI) into PGx research and implementation. Advanced computational methods are employed to analyze vast datasets, identify complex gene-drug interactions, and predict individual responses to treatment. AI algorithms contribute to the development of more precise and personalized treatment regimens, taking into account a patient’s unique genetic profile along with other clinical factors.

Challenges and Future Directions

Despite significant progress, challenges persist, such as the need for standardized guidelines, addressing ethical considerations, and ensuring the equitable distribution of PGx benefits. The future of PGx holds promise with ongoing research in epigenomics, transcriptomics, and systems biology, providing a more comprehensive understanding of the factors influencing drug response. The integration of PGx into electronic health records and routine clinical practice is expected to become more widespread, further realizing the vision of personalized medicine.

How genetic information can guide drug selection

This personalized approach to medicine takes into account the inherent genetic variability among individuals, aiming to optimize drug therapy for better efficacy and reduced adverse effects. Two key concepts in this field are pharmacokinetics and pharmacodynamics, which intricately intertwine to shape an individual’s response to medications.

Pharmacokinetics (PK)

Pharmacokinetics involves the study of how the body processes drugs, encompassing absorption, distribution, metabolism, and excretion (ADME). Genetic variations can significantly impact each of these processes, thereby influencing the concentration of a drug in the body over time.

  • Absorption: Genetic factors can affect the absorption of drugs in the gastrointestinal tract. For instance, variations in genes encoding drug transporters or enzymes involved in drug metabolism can influence how quickly or efficiently a drug is absorbed.
  • Distribution: Genetic variations may influence the distribution of drugs within the body, affecting factors such as protein binding and tissue penetration. This can result in variations in drug concentrations at the target site, impacting therapeutic outcomes.
  • Metabolism: Cytochrome P450 enzymes, a family of drug-metabolizing enzymes, exhibit substantial genetic polymorphisms. These polymorphisms can lead to variations in the rate at which drugs are metabolized, influencing both the therapeutic effect and the risk of adverse reactions.
  • Excretion: Genetic factors also play a role in the renal and hepatic excretion of drugs. Variations in genes associated with drug transporters or renal function can impact the elimination half-life of a drug, affecting its overall pharmacokinetic profile.
Metabolism and Excretion

Pharmacodynamics (PD)

Pharmacodynamics involves the study of how drugs exert their effects on the body, focusing on the relationship between drug concentration and its therapeutic or toxic effects. Genetic variations can influence drug receptors, signal transduction pathways, and downstream cellular responses.

  • Receptor Variability: Receptor variability pertains to the variations in the composition, capabilities, and quantity of receptors within the body, which can greatly impact a person’s reaction to medications. Environmental factors, genetic factors, or a combination of the two may be responsible for this variability. Receptor variability is significantly influenced by genetic factors. Genetic variations can affect drug response by modulating protein function, which includes drug receptor function. This is a fundamental component of pharmacogenomics, the study of how people react to drugs based on their genetic makeup. Variable drug responses may be caused by differences in the disease pathways or drug target molecules. Due to the fact that variations in several genes have occasionally been linked, the term “combinatorial pharmacogenetics” was created. Genetic differences can impact how well drug-metabolizing enzymes work, which can change how the body reacts to drugs. It has been demonstrated that common variations in the coding sequence and/or regulatory regions of genes encoding drug transporters, drug receptors, and drug-metabolizing enzymes affect toxicity and treatment response 2.
    Although there are variations in receptor sensitivity, few of these variations — whether inherited or acquired — have clear clinical implications. Drug response variations can be pharmacodynamic, meaning that different individuals respond differently to the same concentrations of the drug, or pharmacokinetic, meaning that different people receiving the same dose of the drug will have different concentrations of the drug in different bodily fluids. A person’s response to any given substance can change depending on changes in the number or function of their receptors. The control of both the quantity and activity of receptors can result in a number of phenomena that influence the variability of drug response. Pharmacogenomic variability refers to variation within a population that may have a genetic basis rooted in the polymorphism of enzymes or receptor molecules among members of the same species 6.
    To summarize, receptor variability is a multifaceted phenomenon that is impacted by a wide range of variables, such as genetic variants, environmental cues, and the interactions between these variables. Comprehending this variability is essential for forecasting individual drug responses and for formulating strategies for personalized medicine.
  • Signal Transduction Pathways: A set of molecular processes known as signal transduction pathways carry signals from the outside of a cell into its interior, where they are eventually translated into a cellular response. These pathways entail the binding of ligands, or signalling molecules, to cell surface receptors. A cellular reaction is triggered by the binding, which sets off a series of intracellular events, many of which involve protein phosphorylation mediated by protein kinases. Changes in cellular location, protein activity, or gene expression are examples of the response. There are various kinds of signal transduction pathways, such as the G protein-coupled receptor (GPCR), JAK-STAT, and receptor tyrosine kinase (RTK) pathways. These pathways are essential for many cellular functions, including apoptosis, differentiation, and cell growth. The body needs signal transduction pathways to remain in a normal physiological balance. They enable intercellular communication and the response of cells to environmental cues, growth factors, and hormones, among other external stimuli. It is essential to comprehend these pathways in order to develop personalized medicine techniques and targeted therapies.
  • Drug Metabolism and Targets: The term “drug metabolism” describes how the body changes drugs, usually by using specific enzyme systems. Drugs are substances that are not normally present in an organism’s biochemistry, and this process also referred to as biotransformation, alters their chemical structure. Drug metabolism aims to increase the drug’s solubility in water in order to facilitate its excretion. Drugs are primarily metabolized in the liver, where enzymes change them into active or inactive forms. The duration and potency of a drug’s pharmacologic action are determined by its rate of metabolism. Drug metabolism can be impacted by a number of factors, including genetic polymorphisms, drug-drug interactions, and organ dysfunction. Conversely, the specific biological entities within the body that drugs interact with to produce a therapeutic effect are known as drug targets. Proteins such as enzymes, protein kinases, and G-protein coupled receptors (GPCRs) are the majority of drug targets. A drug’s ability to modulate a target’s function through binding can alter the physiological state of a cell. One of the most important steps in the discovery and development of new drugs is choosing a target. The degree to which a medication interacts with its target determines how effective it is in most cases. For mechanism-based drug discovery to be successful, the definition of a drug target is essential. Additionally, efforts to comprehend stratified clinical efficacy and safety, predict drug utility in patient subgroups, rationalize the differences between drugs in the same therapeutic class, and link drug response to genetic variation are becoming more and more important.
    For the creation of safe and efficient drugs, it is imperative to comprehend both drug targets and drug metabolism. It makes it possible to anticipate drug interactions, identify possible side effects, and tailor drug therapy to a patient’s genetic composition.

How Personalized Medicine Through Pharmacogenomics Can Enhance Treatment Efficiency

One of the primary advantages of personalized medicine through PGx lies in its ability to unravel the intricate genetic variations that underlie an individual’s response to medications. The human genome is a mosaic of diverse genetic compositions, and subtle variations in genes encoding drug-metabolizing enzymes, drug transporters, and drug targets can profoundly influence how an individual processes and responds to a particular medication. By deciphering these genetic nuances, PGx empowers clinicians to prescribe medications that are not only more effective but also safer for the patient.

Preventing adverse drug reactions is a paramount goal in medicine, as ADRs not only compromise patient well-being but also contribute to increased healthcare costs.

Adverse Drug Reactions (ADRs) refer to unintended and harmful responses to medications that occur at doses used for treatment, prevention, or diagnosis. These reactions can range from mild to severe and may manifest in various ways, including side effects, allergic reactions, and toxicity.

Adverse drug reactions are a significant concern in healthcare, as they can impact patient well-being, lead to increased healthcare costs, and sometimes result in the discontinuation of a potentially beneficial treatment. There are several types of adverse drug reactions:

  1. Side Effects: These are unintended, usually predictable effects of a medication that occur at therapeutic doses. Side effects can vary widely and may include symptoms such as drowsiness, nausea, headache, or dizziness. While side effects are often mild and tolerable, they can influence a patient’s adherence to a treatment plan.
  2. Allergic Reactions: Some individuals may develop an allergic response to a medication. This can range from mild allergic reactions, such as skin rashes, to severe and life-threatening reactions, like anaphylaxis. Allergic reactions are often unpredictable and can occur even after the patient has taken the medication without incident.
  3. Toxic Reactions: Toxic reactions result from an excessive dose of a medication, leading to harmful effects on the body. The toxicity can affect specific organs or systems and may manifest as symptoms such as liver damage, kidney dysfunction, or cardiovascular complications.
  4. Idiosyncratic Reactions: These are unusual and unexpected responses to a medication that are not related to the pharmacological action of the drug. Idiosyncratic reactions are often rare and can be influenced by individual genetic factors or other unknown predispositions.
  5. Drug Interactions: Adverse drug reactions can also occur when two or more medications interact in ways that enhance or diminish their effects. Drug interactions may lead to unexpected side effects or altered therapeutic outcomes.

Identifying and managing adverse drug reactions is a critical aspect of patient care. Healthcare professionals must consider factors such as the patient’s medical history, concurrent medications, and individual variations in drug metabolism to minimize the risk of adverse reactions. Monitoring for side effects during treatment and promptly addressing any concerns or symptoms reported by the patient are essential components of a comprehensive approach to medication safety. In the era of personalized medicine, pharmacogenomics plays a crucial role in predicting and preventing adverse drug reactions by tailoring treatment based on an individual’s genetic makeup. PGx achieves this by identifying genetic markers associated with an increased risk of ADRs. For example, certain variations in genes responsible for drug metabolism, such as those encoding cytochrome P450 enzymes, can predispose individuals to poor metabolizer status, leading to elevated drug levels and an augmented risk of toxicity. Armed with this genetic information, clinicians can tailor drug dosages or select alternative medications to circumvent potential adverse reactions, thereby fostering a safer therapeutic environment.

Furthermore, personalized medicine through PGx serves as a potent tool in preventing undesirable drug responses. Genetic variations can influence drug efficacy by altering the drug’s pharmacokinetics or its interaction with specific targets. For instance, a polymorphism in a receptor gene might result in diminished drug binding affinity, rendering the treatment less effective. By identifying such variations, PGx facilitates the selection of medications that are not only more likely to be efficacious but also aligned with the individual’s genetic predispositions, thereby optimizing treatment outcomes.

The integration of PGx into clinical decision-making is not without its challenges. Implementation requires a concerted effort to establish robust infrastructure for genetic testing, interpret complex genetic data, and educate healthcare professionals about the implications of genetic information on drug response. Moreover, ethical considerations surrounding privacy and the potential for genetic discrimination must be carefully addressed to ensure the responsible and equitable application of PGx.

P450

Cryochrom P450 (CYP) is essential for the metabolism of medicine. Given the potential severity of drug-drug interactions, advanced practitioners (APs) must have a thorough understanding of the CYP system. Before understanding what P450 is, let’s discuss what drug metabolism is and genetic polymorphisms.

Genetic Polymorphisms And Drug Metabolism

Genetic polymorphisms, the natural variations in the DNA sequence among individuals, form the intricate tapestry of human diversity. These variations, occurring at specific loci in the genome, are the building blocks of our genetic makeup, contributing to the unique traits, physical characteristics, and susceptibilities that distinguish one individual from another. This essay delves into the nature, inheritance, functional impact, disease associations, and broader implications of genetic polymorphisms, exploring their role in shaping the rich mosaic of the human population.

Nature of Genetic Polymorphisms:

Genetic polymorphisms are ubiquitous and fundamental to the genetic architecture of human populations. Among the various types, Single Nucleotide Polymorphisms (SNPs) stand out as the most prevalent, involving the substitution of a single nucleotide base. These variations occur naturally and contribute to the remarkable diversity observed within and across different populations.

Inheritance and Genetic Makeup:

Each individual inherits a unique combination of genetic polymorphisms from their parents. The combination of these variations shapes an individual’s genetic makeup, influencing traits ranging from eye colour and height to complex physiological processes. The interplay of polymorphisms inherited from both parents results in the myriad genetic profiles that define the human population.

Functional Impact of Genetic Polymorphisms:

Certain genetic polymorphisms exert functional consequences, impacting gene expression, protein function, or other molecular processes. These functional variations contribute to the vast array of individual differences observed in traits, behaviours, and responses to environmental factors. Some polymorphisms may confer adaptive advantages, allowing populations to thrive in specific environments.

Disease Associations:

Genetic polymorphisms play a crucial role in influencing susceptibility to various diseases. Certain variations are associated with an increased risk of developing specific conditions, such as diabetes, cardiovascular diseases, or certain types of cancer. These associations highlight the complex interplay between genetic factors and disease pathogenesis.

Evolutionary Significance:

The prevalence of genetic polymorphisms reflects the dynamic nature of human evolution. Over time, populations have adapted to diverse environments, leading to the enrichment of specific polymorphisms that confer advantages in the face of selective pressures. The diversity observed in different regions around the world is a testament to the ongoing evolutionary journey of the human species.

Population Diversity:

Different populations exhibit varying frequencies of specific genetic polymorphisms, reflecting the historical and geographical context of their evolution. This diversity is not only a testament to the complex history of human migration but also emphasizes the need for inclusive genetic research and medical practices that consider the unique genetic makeup of diverse populations.

Genetic polymorphisms, variations in DNA sequences among individuals, play a crucial role in drug metabolism, influencing how medications are absorbed, distributed, metabolized, and eliminated in the body.

Drug Metabolism

Drug metabolism, a fundamental aspect of pharmacology, orchestrates the intricate processes by which the human body transforms and eliminates pharmacologically active compounds. This essay provides an in-depth exploration of the multifaceted nature of drug metabolism, encompassing its phases, key enzymatic players, factors influencing its dynamics, and the clinical significance of these processes.

1)Phases of Drug Metabolism:

1.a)Phase I Metabolism: Navigating Oxidation, Reduction, and Hydrolysis

1.a.a)Enzymatic Landscape: Cytochrome P450 (CYP) Orchestration

In Phase I metabolism, the spotlight falls on the formidable Cytochrome P450 (CYP) enzymes, stationed predominantly in the liver. These enzymes are the maestros orchestrating a complex arrangement of oxidation, reduction, and hydrolysis reactions. Their versatility allows for the introduction of reactive functional groups onto drug molecules, setting the stage for subsequent metamorphoses.

1.a.b)Functional Goal: Enhancing Reactivity for Transformation

The primary objective of Phase I metabolism is to elevate drug reactivity. By introducing functional groups, CYP enzymes prepare the groundwork for subsequent transformations in the drug molecule. This enhancement in reactivity serves as a crucial prelude to the subsequent phases, paving the way for the nuanced alterations that define the continuum of drug metabolism.

1.b)Phase II Metabolism: Conjugation and Water Solubility Enhancement

1.b.a)Enzymatic Harmonization: The Role of Conjugative Enzymes

As the curtain rises on Phase II metabolism, a cohort of conjugative enzymes takes center stage. Notable participants in this enzymatic harmonization include UDP-glucuronosyltransferases (UGTs) and sulfotransferases. These enzymes engage in intricate conjugation reactions, forming alliances with drug molecules to shape their destiny.

1.b.b)Water Solubility Augmentation: A Symphony of Conjugation

The thematic resonance of Phase II metabolism lies in its pursuit of increased water solubility. Through the meticulous dance of conjugation reactions, drugs undergo transformations that render them more amenable to efficient excretion. The water-soluble derivatives emerging from this phase mark the crescendo of drug metabolism, setting the stage for their exit from the biological stage.

2)Key Enzymes in Drug Metabolism:

2.a)Cytochrome P450 (CYP) Enzymes: Liver Predominance and Genetic Variability

2.a.a)Liver Predominance: The Hepatic Command Center

In the intricate dance of drug metabolism, CYP enzymes take center stage, predominantly residing in the liver. With a primary role in Phase I reactions, these hepatic virtuosos exert direct influence over drug bioavailability, shaping the initial acts of drug transformation within the body.

2.a.b)Genetic Variability: The Interplay of Polymorphisms

Adding a layer of complexity to this biochemical ballet is the genetic variability within CYP genes. Polymorphisms contribute to interindividual diversity in drug response, necessitating nuanced therapeutic considerations. The variations in CYP genes underscore the need for personalized approaches to drug administration.

2.b) UDP-Glucuronosyltransferases (UGTs): Conjugation Virtuosos with Versatility

2.b.a)Phase II Conjugation: UGTs Taking the Center Stage

As the molecular ballet progresses to Phase II, UGTs emerge as the protagonists in conjugation reactions. Catalyzing the coupling of drugs with glucuronic acid, UGTs enhance water solubility, preparing drugs for their graceful exit from the body. Their role is pivotal in the finale of drug metabolism.

2.b.b)Versatility: A Diverse Substrate Spectrum

UGTs showcase remarkable versatility, embracing a diverse array of drugs within their substrate spectrum. This expansive repertoire underscores their centrality in the grand narrative of drug metabolism, ensuring a comprehensive approach to the conjugation of varied pharmacological entities.

2.c) Sulfotransferases: Water Solubility Augmentation and Clinical Relevance

2.c.a)Water Solubility Augmentation: Executing Sulfation Reactions

Sulfotransferases, another cohort of enzymatic artists, contribute significantly to Phase II metabolism. Their mastery lies in executing sulfation reactions that augment water solubility, marking a crucial phase in the transformation of drugs into metabolites ready for efficient excretion.

2.c.b) Clinical Relevance: Essential Players in Clearance and Elimination

Beyond their role in augmenting water solubility, sulfotransferases emerge as essential players in drug clearance and elimination. Their clinical relevance extends to the intricate balance required for maintaining optimal drug levels within the body, influencing therapeutic outcomes.

2.c.c)Glutathione S-Transferases: Detoxification Prowess and Multifaceted Influence

2.c.d)Detoxification Prowess: Conjugation with Glutathione

In the final act of this enzymatic ballet, Glutathione S-Transferases showcase their detoxification prowess. Conjugation with glutathione serves as a cellular defence mechanism, facilitating the removal of potentially harmful entities and contributing to the body’s overall detoxification processes.

2.c.e)Beyond Drug Metabolism: Multifaceted Influence

The influence of Glutathione S-Transferases extends beyond the realm of drug metabolism. Their involvement in cellular homeostasis underscores a multifaceted role that goes beyond the biochemical transformations of pharmacological compounds, emphasizing their importance in broader physiological processes.

3. Factors Influencing Drug Metabolism:

3.a) Genetic Factors: Unraveling the Code of Individualized Medicine

3.a.a)Polymorphic Variations: A Genetic Tapestry of Diversity

Genetic polymorphisms in drug-metabolizing enzymes weave a tapestry of diversity in the intricate code of individualized medicine. The variations introduce complexity, demanding a nuanced understanding to tailor therapeutic strategies to the unique genetic landscape of each patient.

3.a.b) Clinical Relevance: Paving the Way for Personalized Therapeutics

Understanding the genetic landscape is not merely an academic pursuit; it is the key to unlocking personalized therapeutic strategies. This knowledge becomes the compass guiding clinicians toward optimized drug selection and dosing, ensuring that treatment aligns harmoniously with the individual’s genetic makeup.

3.b)Age and Gender: Dynamic Influences on Drug Metabolism

3.b.a)Physiological Dynamics: Tailored Considerations Across the Lifespan

Age-related variations in drug metabolism necessitate a tailored approach in pediatric and geriatric populations. The physiological dynamics that evolve with age demand a nuanced understanding to ensure that therapeutic interventions are calibrated to the unique needs of each age group.

3.b.bHormonal Influences: Gender-Specific Pharmacology

In the intricate dance of drug metabolism, gender emerges as a significant partner. Influenced by hormonal fluctuations, gender-related disparities in drug metabolism underscore the importance of gender-specific pharmacology. Recognizing these influences becomes paramount in crafting precision medicine for diverse patient populations.

3.c) Disease States: Navigating Altered Hepatic and Renal Landscapes

3.c.a)Hepatic and Renal Impact: Profound Influence on Metabolic Pathways

Altered hepatic or renal function casts a profound influence on drug metabolism, transforming the metabolic landscape. Therapeutic approaches must adapt to these changes, acknowledging the shifting terrain shaped by compromised organ function.

3.c.a)Clinical Monitoring Imperative: Vigilance in the Face of Organ Compromise

Patients navigating disease states with compromised organ function become focal points of clinical vigilance. Vigilant clinical monitoring becomes imperative to ensure optimal drug dosing, balancing the delicate equilibrium between therapeutic efficacy and avoiding adverse effects.

3.d)Drug-Drug Interactions: The Intricacies of Therapeutic Navigation

3.d.a)Enzyme Modulation: Reshaping the Metabolic Landscape

Co-administration of drugs introduces a dynamic element — enzyme modulation. This interaction can induce or inhibit drug-metabolizing enzymes, reshaping the metabolic landscape and altering the fate of concurrently administered therapeutic agents.

3.d.b)Therapeutic Navigation: A Comprehensive Understanding is Paramount

In the intricate ballet of drug-drug interactions, therapeutic navigation becomes an art. A comprehensive understanding of these interactions is paramount for optimizing treatment regimens, steering clear of undesirable outcomes, and ensuring the harmonious coexistence of multiple therapeutic agents.

4. Clinical Significance:

4.a)Individualized Drug Therapy: Precision in Practice

4.a.a)Precision in Practice: Harnessing Genetic and Physiological Insights

The cornerstone of individualized drug therapy lies in precision, achieved through pharmacokinetic profiling based on genetic and physiological factors. This nuanced understanding allows clinicians to unravel the unique metabolic tapestry of each patient, guiding therapeutic decisions with unparalleled accuracy.

4.a.b)Clinical Implementation: Tailoring Regimens for Optimal Outcomes

Beyond theoretical precision, individualized medicine finds its essence in clinical implementation. Armed with insights into individual metabolic profiles, clinicians tailor drug regimens to optimize clinical outcomes. This approach ensures that each patient receives a personalized prescription, finely tuned to their distinct physiological nuances.

4.b)Drug Safety and Efficacy: A Dual Imperative

4.b.a)Adverse Reaction Mitigation: Predicting and Preventing Unwanted Consequences

The depth of knowledge in drug metabolism becomes a shield against adverse reactions. In-depth insights aid in the prediction and mitigation of adverse reactions, ensuring that the therapeutic journey is free from unexpected hurdles. This proactive stance enhances the safety profile of drug regimens.

4.b.b)Efficacy Optimization: A Precise Path to Therapeutic Success

The quest for optimal treatment outcomes hinges on precise comprehension of drug metabolism. This knowledge not only mitigates the risk of suboptimal treatment outcomes but actively contributes to therapeutic efficacy. The result is a therapeutic journey sculpted for success, with each drug fulfilling its intended purpose with maximal impact.

The intricate interplay between genetics and drug metabolism is a captivating symphony that shapes the fate of pharmaceutical agents within the human body. Genetic variations, or polymorphisms, hold the key to understanding how enzymes responsible for drug clearance operate with diverse efficiency among individuals. And now let's assess the P450 chronotype (CYP), and how CYP and the genes affect medicine.

What is P450

The cytochrome P450 (CYP) enzymes are a protein superfamily involved in the synthesis and metabolism of drugs, toxins and normal cellular components.

Catalytic Functions of Individual CYPs:

Within the vast realm of the CYP superfamily, each member predominantly functions as a monooxygenase, intricately adding polar hydroxyl groups to substrates. This enzymatic modification serves a dual purpose: it enhances the reactivity of the substrate while rendering it more hydrophilic. The result is a transformed molecule with heightened chemical reactivity and water solubility, poised for further biological interactions.

Substrate Specificity and Structure-Activity Relationship:

Individual CYPs exhibit distinct affinities for specific compounds, showcasing a nuanced substrate specificity dictated by the molecular architecture of their active sites. The interaction between CYPs and substrates follows a structure-activity relationship, where the chemical structure of a substrate dictates its compatibility with a particular CYP enzyme. This tailored specificity contributes to the diverse catalytic functions exhibited by different members of the CYP superfamily.

Diversity in Compound Interaction:

The collective impact of the CYP superfamily extends beyond endogenous compounds, reaching out to interact with a plethora of exogenous substances. This expansive reach underscores the versatility of CYP enzymes, allowing them to engage with compounds of highly diverse chemical structures. The ability to metabolize both endogenous and exogenous compounds highlights the significance of CYPs in maintaining biochemical homeostasis.

Physiological Processes Governed by CYP Superfamilies:

The influence of CYP enzymes extends to critical physiological processes, with specific families playing instrumental roles. Families such as CYP11, CYP17, CYP19, CYP21, and CYP51 are integral in catalyzing key steps in hormone synthesis. This involvement in hormonal pathways underscores the profound impact of CYP superfamily members on the regulation of essential physiological functions.

Nomenclature and Genomic Sequencing:

The nomenclature of CYP genes adheres to standardized conventions, primarily based on sequence similarity. Families, subfamilies, and individual genes are assigned numerical identifiers, facilitating a systematic classification. The advent of genomic sequencing technologies has propelled the identification of the complete CYP gene component in various metazoan species. This includes meticulous exploration within major metazoan phyla, offering a comprehensive understanding of the genetic landscape of the CYP superfamily.

Gene Numbering and Sequence Identity:

CYP genes are assigned numerical identifiers based on the order of their discovery. A critical aspect of classification is the determination of sequence identity, with CYPs sharing greater than 40% amino acid sequence belonging to the same family and those with greater than 55% sequence identity designated to the same subfamily. This systematic approach ensures a precise categorization reflective of genetic relationships.

Evolution of CYP Genes and Genomic Diversity:

Advancements in genomic sequencing have facilitated the identification of CYP genes across various metazoan species, revealing a wide spectrum of genomic diversity. The number of CYP genes tends to vary, with most vertebrate genomes containing between 50–100 CYP genes. This genomic diversity highlights the evolutionary significance of the CYP superfamily in adapting to the metabolic needs of different species.

Diffrent genes in P450

Mammalian CYP Families: A Genomic Symphony:

In the orchestration of mammalian biology, the 18 cytochrome P450 families stand as distinct movements, encoding 57 genes within the intricate composition of the human genome. This genomic symphony unfolds as a testament to the evolutionary complexity and adaptive diversity embedded within the CYP families.

Categorization of Cytochromes: Navigating the ‘Type I’ and ‘Type II’ Dichotomy:

The categorization of cytochromes into ‘type I’ and ‘type II’ delineates not only their subcellular locations but also underscores the structural and functional diversity inherent within these molecular entities. While ‘type I’ cytochromes find residence in bacteria and eukaryotic mitochondria, their ‘type II’ counterparts grace the eukaryotic endoplasmic reticulum (ER), encapsulated within microsomes.

Mitochondrial Marvels: The Exclusive Seven:

Within the cellular tapestry, seven P450 enzymes — encoded by the CYP11A1, CYP11B1, CYP11B2, CYP24A1, CYP27A1, CYP27B1, and CYP27C1 genes — emerge as exclusive inhabitants of the mitochondria, categorizing them unequivocally as ‘type I’ cytochromes. Their residence within this cellular powerhouse unveils a specialized landscape where NADH or NADPH orchestrates a dance of electrons, intricately choreographed through membrane-bound flavoprotein ferredoxin reductase (FDXR) and soluble iron–sulphur protein ferredoxin (FDX), culminating in the electron donation to these ‘type I’ P450s.

Electron Dance in the Mitochondrion: A Molecular Ballet:

The mitochondrion, a stage for molecular ballet, witnesses the graceful exchange of electrons facilitated by NADH or NADPH. The dance unfolds as FDXR passes these electrons to FDX, a soluble iron–sulphur protein, which then elegantly donates the electrons to the awaiting ‘type I’ P450s. This finely tuned choreography underscores the elegance with which cellular processes unfold within the mitochondria.

Within the superfamily, there are different P450(CYP) enzymes. Which play a crucial role in the detoxification of drugs and foreign chemicals, as well as in the synthesis and metabolism of various endogenous molecules. Here’s a break down of some of the key enzymes and their functions:

  1. CYP1A1: Involved in the metabolism of eicosanoids and foreign chemicals.
  2. CYP1A2: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  3. CYP1B1: Involved in the metabolism of eicosanoids and foreign chemicals. Associated with primary congenital glaucoma.
  4. CYP2A6, 2A13: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  5. CYP2A7: Involved in nitrosamine metabolism.
  6. CYP2B6: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  7. CYP2C8, 2C9, 2C18, 2C19: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals. CYP2C9 contributes to warfarin metabolism.
  8. CYP2D6: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  9. CYP2E1: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  10. CYP2F1: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  11. CYP2J2: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  12. CYP2R1: Acts as a vitamin D 25-hydroxylase. Associated with vitamin D 25-hydroxylase deficiency.
  13. CYP2S1: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals. Also, metabolizes retinoids.
  14. CYP2U1: Involved in the metabolism of eicosanoids, long-chain fatty acids, drugs, and foreign chemicals.
  15. CYP2W1: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  16. CYP3A4: Major enzyme in the metabolism of eicosanoids, sex steroids, and a wide range of clinically used drugs and foreign chemicals.
  17. CYP3A5, 3A7, 3A43: Involved in the metabolism of eicosanoids, sex steroids, drugs, and foreign chemicals.
  18. CYP4A11: Involved in the metabolism of eicosanoids, medium- and long-chain fatty acids, drugs, and foreign chemicals. Associated with hypertension and coronary artery disease.
  19. CYP4B1: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  20. CYP4F2: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals. Contributes to warfarin metabolism.
  21. CYP4F3, 4F8: Involved in the metabolism of eicosanoids, drugs, and foreign chemicals.
  22. CYP7A1: Associated with elevated LDL-cholesterol levels. Acts as cholesterol 7α-hydroxylase and is involved in the biosynthesis of bile acids and the metabolism of oxysterols.
  23. CYP11A1: Involved in cholesterol side-chain cleavage. Associated with congenital adrenal insufficiency with 46, XY sex reversal.
  24. CYP11B1: Acts as a steroid 11β-hydroxylase. Associated with occasional congenital adrenal hyperplasia (CAH).
  25. CYP11B2: Acts as a steroid 11β- and 18-hydroxylase and 18-oxidase. Associated with corticosterone methyloxidase deficiency types I and II, increased aldosterone-to-renin ratio.
  26. CYP17A1: Acts as a steroid 17α-hydroxylase and 17,20-lyase. Associated with steroid 17α-hydroxylase, 17,20-lyase deficiency.
  27. CYP19A1: Acts as an androgen aromatase and estrogen synthetase. Associated with aromatase deficiency and aromatase excess syndrome.
  28. CYP21A2: Acts as a steroid 21-hydroxylase. Associated with congenital adrenal hyperplasia (CAH).
  29. CYP24A1: Acts as a vitamin D 24-hydroxylase. Associated with idiopathic infantile hypercalcemia.
  30. CYP26A1, CYP26B1, CYP26C1: Involved in retinoid acid (RA) inactivation (hydroxylase). Associated with embryo lethality in mice.
  31. CYP27A1: Involved in bile acid biosynthesis, sterol 27-hydroxylase, and vitamin D 25-hydroxylase. Associated with cerebrotendinous xanthomatosis.
  32. CYP27B1: Acts as a 25-hydroxy-vitamin D 1α-hydroxylase. Associated with vitamin D-dependent rickets type 1A and infantile hypocalcemia.
  33. CYP39A1: Acts as an oxysterol 7α-hydroxylase.
  34. CYP46A1: Acts as a cholesterol 24-hydroxylase in the brain.
  35. CYP51A1: Acts as a lanosterol 14α-demethylase. Associated with embryo lethality in mice.

Thank you, and I hope you enjoyed reading and learning about the P450 superfamily, drug metabolism, and the importance of personalization of prescription drugs. If you want to connect here's my LinkedIn: https://www.linkedin.com/in/jayasra/ and my email is @srajaya02@gmail.com stay tuned for my next project on how we can acctually create a more convenient test to improve the world of pharmacogenomics.

--

--