Making Personalized Prescriptions More Accessible

Jaya Sra
38 min readNov 12, 2023

Pharmacogenomics is an innovative concept that promises tailored treatment plans and improved patient outcomes. As the twenty-first century progresses, the key to customizing medication regimens to each patient’s unique genetic profile and maximizing therapeutic advantages will be found in the use of genetics into clinical practice. The start of this new era presents previously unheard-of chances to transform healthcare, but in order to guarantee access to personalized prescriptions we need to first assure universal access to genetic testing.

Introduction

Pharmacogenomics, which is the study of how a person’s genetic composition affects how they respond to medications, has the potential to revolutionize healthcare by assisting with treatment decisions, averting negative drug responses, and maximizing the efficacy of medications. Every year over 100,000 people in America alone die because of a lack of personalization in their prescription drugs.

Our knowledge of the interaction between genetics and drug metabolism has expanded dramatically since the introduction of ground-breaking technologies like genome-wide association studies and next-generation sequencing. But even though science is making great strides in this area, there is still a huge gap between pharmacogenomics’ potential and its actual use in medical contexts. This divide is typified by the ongoing inaccessibility of genetic testing, which is an immovable obstacle for a large number of patients and medical professionals.

This accessibility deficiency has significant implications for patient outcomes, healthcare equity, and the realization of pharmacogenomics’ full promise. In order to ensure that every patient, regardless of their socioeconomic level or geographic location, can benefit from the insights that pharmacogenomics has to offer, it is imperative that we investigate this matter and look for creative ways to bridge the genetic gap.

The benefits of genetic testing accessibility in pharmacogenomics are enormous, but the journey there is not to be taken lightly. We have the ability to lessen the suffering brought on by adverse drug reactions, lower healthcare expenses, and improve the standard of care overall. In this article I will take you on a thorough exploration of the genetic landscape, breaking down the complexities, looking into the potential fix, and imagining a time when pharmacogenomics revolutionizes healthcare worldwide.

Problem

With new and innovative technologies and treatments being developed on a regular basis, the practice of medicine has experienced tremendous advancements over the years. Still, one enduring issue in the healthcare system is the lack of prescription medication personalization. The one-size-fits-all approach to medicine, in which individual characteristics are not taken into account and treatments are frequently prescribed based on data from the general population, is the issue at hand. The increasing use of generic drugs is one of the main problems causing prescription medicine to lack personalization. These medications take into account individual patient variations in drug metabolism, genetics, or disease characteristics, despite being widely available and cost-effective. Because generic drugs are usually created using population averages, many patients do not receive the best possible outcomes.

The fact that doctors frequently use a trial-and-error method when writing prescriptions is another example of how personalization is lacking. Until a suitable treatment is found, patients may experience numerous medication changes and dosage adjustments, which could put them at unnecessary risk and result in prolonged suffering. This strategy not only increases healthcare expenses but also postpones necessary treatment.

Patients often develop drug resistance in the context of chronic conditions, cancer, and infectious diseases. Part of the issue is that drug regimens are not personalized enough, meaning that factors unique to each patient that could affect the effectiveness of the drug are not taken into consideration. This can have negative effects on patients by necessitating the use of stronger drugs, larger dosages, or alternative treatment modalities.

The way that people react to drugs is largely influenced by their genetic makeup. However, these individual differences are ignored by a lack of genetic testing and customized prescription methods. Pharmacogenomics, which adapts medication regimens to a person’s genetic composition, is still underapplied, which can result in less-than-ideal treatment results and possibly harmful side effects.

Prescription medications are less individualized, which increases the risk of side effects and intolerances. When drugs that might not be suitable for a patient are prescribed, they put them at needless risk. The management of these unfavourable events raises healthcare costs in addition to having an impact on patient well-being.

One important effect of the issue is subpar treatment outcomes. Patients may endure protracted suffering and possibly even worsening of their condition if there is a delay in receiving symptom relief or managing their conditions. For illnesses like cancer or some infectious diseases that have limited treatment windows, this could have disastrous effects.

Prescription drug personalization is lacking, which drives up the expense of healthcare. Medication errors or poorly tolerated drugs may result in patients needing more doctor visits, hospital stays, and treatments. Moreover, the need for continuous research, development, and testing of substitute medications or treatment approaches increases the financial burden.

Total Retail Prescription Drug Spending in 2017

To allow for customized therapies, the conventional blockbuster model — which depends on mass-market pharmaceuticals — might need to be modified. The profitability of the industry may be impacted by the significant research and development costs associated with the creation of targeted pharmaceuticals.

A widespread issue in healthcare is the lack of personalization in prescription drugs. It results in less-than-ideal patient outcomes, higher healthcare expenses, and difficulties for the pharmaceutical sector. In order to solve this issue, personalized medicine strategies that take into account each patient’s unique traits, genetics, and preferences must be adopted. By doing this, we can raise patient satisfaction, lower costs, and change the healthcare system to be more effective and patient-centred.

Pharmacogenomics

The study of how a person’s particular genetic composition affects how they react to drugs is known as pharmacogenomics, a cutting-edge field at the nexus of genetics and pharmacology. It includes a thorough examination of genetic variations, including single nucleotide polymorphisms (SNPs) and other genetic markers, to clarify the ways in which these genetic factors influence drug metabolism, interactions with particular molecular targets in the body, and treatment outcomes.

Pharmacogenomics has a wide range of applications that cut across many medical specialties, giving it a broad scope. It explores the genetic variations that control how drugs behave, such as those that control how drugs are absorbed, distributed, metabolized, and excreted (pharmacokinetics) and how they affect how drugs behave pharmacologically in the body (pharmacodynamics).

Pharmacogenomics is closely related to applications that are specific to individual diseases. For example, it is essential in oncology to customize cancer therapies according to the genetic makeup of tumours, in cardiology to optimize treatments for cardiovascular disease, and in psychiatry to provide individualized psychotropic medications.

Furthermore, it plays a crucial role in the battle against infectious diseases by facilitating the estimation of the effectiveness of antibiotic and antiviral therapies and resolving issues with drug resistance.

Pharmacogenomics is also changing the way that pain is managed by providing customized options for painkillers that reduce side effects and maximize control over pain. It is noteworthy that it now covers pediatrics and neonatology, guaranteeing that even the youngest patients are given the safest and most appropriate medications. The promise of pharmacogenomics to usher in a personalized era of medicine, where treatments are precisely tailored to the genetic profile of each patient, maximizing treatment efficacy and reducing side effects, makes it significant. This area of study has the potential to transform healthcare by lowering the frequency of adverse drug reactions, increasing drug efficacy, and ultimately producing more affordable and high-quality medical treatments.

Pharmacogenomics has evolved historically, with notable turning points and discoveries that have molded the discipline into what it is today. Its foundations are found in the larger fields of genetics and pharmacology. With Karl Landsteiner’s invention of blood typing and the realization that some people might react negatively to particular medications, the first knowledge of how genetic factors affect drug responses emerged in the early 1900s.

Karl Landsteiner’s

But it wasn’t until the middle of the 20th century that the field started to take shape thanks to the work of researchers like Linus Pauling, who discovered the genetic basis for sickle cell anemia, and others who tried to figure out how drug metabolism traits, like glucose-6-phosphate dehydrogenase deficiency, are inherited. American geneticist Friedrich Vogel first used the term “pharmacogenetics” in the late 1950s. Over the next few decades, a growing amount of information about genetic polymorphisms that impact drug metabolism was discovered, most notably the function of the cytochrome P450 system in drug processing. However, the field did not really take off until the late 20th century with the introduction of sophisticated genotyping and sequencing technologies. After the Human Genome Project was completed in 2003, a thorough map of the human genome was produced, providing pharmacogenomic researchers with a plethora of information and research opportunities.

Timeline of the pharmacogenomic industry

With multiple pharmacogenomic studies, applications in various medical specialties, and an ever-expanding understanding of the genetic basis of drug responses, the field has experienced explosive growth since then. Pharmacogenomics’ historical development highlights how it has progressed from an academic idea to a crucial part of contemporary healthcare, with the potential to completely change the way that drugs are prescribed and given to patients based on their individual genetic profiles.

Personalized medicine is a cutting-edge approach to healthcare that is revolutionizing the way that medical treatments are provided. It is important because it can significantly improve patient outcomes by customizing medical interventions to each patient’s unique genetic and physiological makeup. Personalized medicine makes sure that patients receive treatments that are not only more effective but also less likely to cause adverse reactions or suboptimal responses by utilizing cutting-edge technologies like pharmacogenomics and precision diagnostics.

In the field of oncology, where targeted therapies have raised survival rates and enhanced the quality of life for cancer patients, this strategy has shown to be particularly revolutionary. Better management of cardiovascular diseases has also resulted from the ability to choose the right medications and dosages based on a patient’s individual genetic profile in cardiology. Personalized medicine maximizes drug choice, dosage, and treatment plans by reducing the need for trial-and-error methods. Beyond these specific fields, personalized medicine’s main significance rests in its ability to improve the management of chronic conditions, lessen adverse drug reactions, and address healthcare disparities by guaranteeing that each patient receives a treatment plan customized to meet their unique needs. All of these benefits will ultimately lead to better patient outcomes, lower healthcare costs, and a more patient-centered healthcare system.

It is impossible to overestimate the importance of using pharmacogenomics to reduce Adverse Drug Reactions (ADRs). ADRs have a significant negative impact on patient health as well as the overall healthcare system. These responses can range from minor discomfort to serious, potentially fatal events, which can result in extended suffering, hospital stays, and higher medical expenses. Healthcare providers can proactively reduce the risks of adverse drug reactions (ADRs) by utilizing pharmacogenomic approaches to identify genetic factors that influence drug response. This leads to significant cost savings by averting unfavourable events that require further medical interventions and extended hospital stays, in addition to improving patient safety and care quality. Furthermore, since patients are more likely to follow treatment plans that do not result in uncomfortable or dangerous side effects, a decrease in ADRs also improves patient compliance with regimens. To put it succinctly, pharmacogenomics presents a revolutionary approach to the long-standing issue of adverse drug reactions (ADRs), highlighting its pivotal role in the advancement of healthcare toward personalized medicine, in which individualized treatments are administered to maximize therapeutic outcomes and reduce the likelihood of adverse drug reactions.

Pharmacogenomics-assisted drug efficacy optimization is a revolutionary change in how we think about medicine. Pharmacogenomics ensures that the right drug is given at the right dose by customizing drug therapies to a person’s genetic profile, greatly improving treatment outcomes.

By using a precision approach, medical professionals can choose drugs that have a higher chance of working for the patient and reducing the possibility of side effects and ineffective treatment. Pharmacogenomics has enabled the development of targeted therapies that specifically target the distinct genetic features of a patient’s tumour, a development that has proven beneficial in domains like oncology, where genetic variations are critical to the progression of the disease. Patients with cancer now have better quality of life in addition to higher survival rates.

Pharmacogenomics has proven useful not only in oncology but also in cardiology, psychiatry, pediatrics, and pain management. Patients benefit from therapies that are more in line with their genetic makeup, which improves disease management and increases the chance of effective therapy. In addition to improving patient outcomes, this strategy may lower healthcare expenses by lowering the need for expensive tests, hospital stays, and medication adjustments brought on by ineffective therapies. Personalized medicine aims to provide patients with treatments that are as unique as them, giving them the best chance for a full recovery while minimizing the risks associated with traditional trial-and-error approaches to drug therapy. Optimizing drug efficacy with pharmacogenomics is essentially a critical step toward achieving these goals.

Genetic polymorphisms, or naturally occurring variations in DNA sequences, are central to this field of study. These genetic changes, which include copy number variations, insertions, deletions, and single nucleotide polymorphisms (SNPs), can have a big impact on how people metabolize and react to medications. The Cytochrome P450 enzymes, a family of heme-containing proteins that mediate the metabolism of various pharmaceutical compounds, are important players in this landscape. Cytochrome P450 gene polymorphisms can result in extensive or poor metabolizer phenotypes, which can affect the safety and effectiveness of different medications.

Furthermore, genetic variations affect drug transporters, which are in charge of moving drugs across cell membranes. Drug response and possible toxicity are ultimately influenced by genetic variations in drug transporter genes, such as SLCO1B1 and ABCB1 (P-glycoprotein), which have profound effects on drug absorption, distribution, and excretion. In pharmacogenomics, genetic polymorphisms, Cytochrome P450 enzymes, and drug transporters combine to highlight the complex relationship between genetics and drug responses. This holds promise for precision medicine in that it allows for the customization of drug therapies to a patient’s specific genetic profile, maximizing therapeutic efficacy, and minimizing side effects.

The two fundamental pillars of pharmacogenomics is pharmacokinetics and pharmacodynamics, together determine how drugs interact with the human body. Pharmacokinetics is the study of drug metabolism, clearance, absorption, and distribution as well as how the body absorbs and distributes medications (ADME). Conversely, pharmacodynamics studies how drugs act at the molecular, cellular, and physiological levels to produce both therapeutic and toxic effects. They serve as the cornerstone for comprehending drug actions and enhancing drug therapy when combined.

The main focus of pharmacokinetics is how a drug moves through the body. The four main steps in this journey are elimination, distribution, metabolism, and absorption (ADME). When a medication is given, the absorption phase starts, figuring out how it gets into the bloodstream. This process is influenced by various factors, including the drug formulation, the route of administration, and physiological barriers such as the blood-brain barrier and the gastrointestinal lining.

After being absorbed, the medication travels throughout the body’s different tissues and spaces, where it disperses. During this stage, blood flow, tissue binding, and drug characteristics like lipophilicity and molecular size are all very important. Drugs are converted into metabolites through enzymatic reactions during metabolism, which frequently take place in the liver. This stage can affect a medication’s overall efficacy and safety by changing its activity, bioavailability, and half-life.

Lastly, the drug and its metabolites are eliminated from the body by the elimination process, which mostly involves renal excretion. To optimize drug therapy, healthcare professionals must have a thorough understanding of these pharmacokinetic processes. This will allow them to determine the right dosages, administration routes, and dosing frequencies, all while ensuring therapeutic efficacy and reducing the risk of side effects.

The pharmacodynamics principle is how medications act at the cellular and molecular levels. The idea of drug-receptor interactions, in which a drug binds to particular target receptors to start a series of cellular reactions, is fundamental to this field.

Because medications typically bind to receptors with high affinity and produce the desired pharmacological effects, these interactions are very selective. The relationship between the amount of medication given and the strength of the medication’s effect is explained by dose-response relationships. Dose-response curves, which show the threshold for attaining the maximum therapeutic effect and highlight the possibility of side effects at higher doses, are frequently used to illustrate these relationships. We distinguish between a drug’s capacity to produce a specific effect at a given concentration (potency) and its capacity to produce a maximal effect (efficacy) when evaluating drug potency. Potency indicates how well a drug can produce an effect at lower concentrations, whereas efficacy indicates the drug’s maximum limit in eliciting a response.

Comprehending these parameters is essential for choosing the right medication and figuring out the best dosage schedule. Pharmacodynamics also makes it possible to distinguish between toxic and therapeutic effects, highlighting the delicate balancing act between preventing negative side effects and achieving a desired therapeutic result. This information helps medical practitioners select the best medication for a given ailment, determine the best dosage, and identify any possible drug interactions or side effects, all of which contribute to the safety and efficacy of drug therapies.

Genetic differences can affect the permeability and activity of cellular transporters and drug-metabolizing enzymes in the gastrointestinal tract, which can change the rate and degree of drug absorption in the context of absorption. Variations in genetic polymorphisms can also impact drug transporter expression and function, resulting in differences in drug distribution across the body. Genetic variables also play a role in the variability of drug metabolism, especially when it comes to Cytochrome P450 enzymes, which are in charge of biotransforming a large variety of medications.

These enzyme polymorphisms can result in extensive or poor metabolizer phenotypes, which impact the rate of drug metabolism and, in turn, the therapeutic efficacy or potential toxicity of the drug. Genetic variations can also affect the expression and function of drug targets or signalling molecules, which can affect drug-receptor binding and cellular responses. This may have an impact on a medication’s affinity for its target receptor, the strength of the cellular reaction, and the treatment’s overall effectiveness. In order to optimize drug efficacy, minimize the risk of side effects, and ultimately advance the field of personalized medicine, it is imperative to comprehend these genetic variations and their impact on ADME processes, drug-receptor interactions, and cellular responses.

In this field, genotyping and sequencing are essential tools that enable us to decipher the genetic variations underlying drug responses. The identification of particular genetic markers, such as single nucleotide polymorphisms (SNPs), insertions, deletions, or copy number variations within an individual’s genome, is made possible by genotyping techniques like polymerase chain reaction (PCR) and microarrays.

These markers may provide insight into a patient’s propensity to react to particular drugs by being linked to the activity of drug-metabolizing enzymes, drug transporters, or drug target receptors. However, next-generation sequencing (NGS) methods, such as whole exome and genome sequencing, offer a thorough understanding of a patient’s genetic makeup. Novel genetic variants can be found using NGS, and the complex interactions between various genes and drug responses can be better understood.

Because they enable medical professionals to customize drug therapies to each patient’s specific genetic profile, genotyping and sequencing play a critical role in laying the groundwork for personalized medicine by optimizing therapeutic efficacy and reducing side effects. Genotyping and sequencing methods are essential for deciphering the genetic code that drives personalized medication therapy as pharmacogenomics keeps transforming the healthcare industry.

Pharmacogenomics relies heavily on data analysis and bioinformatics, which are essential for utilizing the abundance of genetic data to predict drug responses and enable clinical applications. Single nucleotide polymorphisms (SNPs) and other genetic markers are analyzed in detail to find correlations between particular genetic variants and drug efficacy or side effects in order to mine genetic data for drug response predictions. Large datasets can be explored using bioinformatics tools and algorithms, allowing for the discovery of genetic signatures that direct personalized drug therapy.

Moreover, the creation of pharmacogenomic databases offers a centralized location for the storage and retrieval of genetic data associated with medication reactions. These databases aggregate information from multiple sources, such as research studies and clinical trials, and provide insightful information about the connections between drug outcomes and genetic variants. Pharmacogenomics has numerous clinical applications in a wide range of medical specialties, including cardiology, psychiatry, pediatrics, and oncology.

Applications/different fields of pharmacogenomics

Oncology

In the field of oncology, pharmacogenomics has become a revolutionary force, having paved the way for the creation of tailored cancer treatments and novel targeted drug delivery strategies. Pharmacogenomics has transformed cancer treatment methods by focusing on the genetic properties of a patient’s tumour. Cancer specialists are able to customize treatment to specifically target the molecular causes of a patient’s cancer by identifying distinct genetic markers, such as mutations or amplifications in genes linked to cancer. This method reduces the toxicity and side effects that can come from conventional chemotherapy while also improving treatment effectiveness. Furthermore, by delivering anticancer agents directly to tumour sites while sparing healthy tissues, targeted drug delivery systems aim to increase the therapeutic index of anticancer medications. This was made possible by advances in pharmacogenomics. With the promise of more potent and less toxic treatments, this personalized approach to oncology, powered by pharmacogenomics, offers hope to cancer patients and signifies a dramatic change in the way cancer is treated.

Cardio

In the field of cardiology, pharmacogenomics has become a game-changing instrument, providing customized antiplatelet and anticoagulant treatments that could completely alter how cardiovascular disease is managed. The genetic foundations of individual drug responses are important in cardiology, and pharmacogenomic insights have made medication regimens more individualized and precise. Variations in the genes CYP2C19 and VKORC1, which are important for drug metabolism and response, have a substantial impact on how patients react to antiplatelet and anticoagulant drugs.

Genetic profiling can be used to identify patients who are either extensive or poor metabolizers of the antiplatelet drug clopidogrel, which is frequently prescribed. While extensive metabolizers may be more susceptible to bleeding complications as a result of an overactive response to the medication, poor metabolizers may not experience the desired antiplatelet effect, placing them at risk of cardiovascular events. Healthcare professionals can optimize therapeutic efficacy and reduce the risk of adverse events by customizing antiplatelet therapy based on a patient’s genetic profile. This improves outcomes for patients with coronary artery disease or those who have had stent placement.

Pharmacogenomics is a key component in anticoagulant therapies, helping to determine how best to use drugs such as warfarin. The ideal warfarin dosage for each patient is influenced by genetic differences in the CYP2C9 and VKORC1 genes. In order to obtain the desired anticoagulant effect, patients with particular genetic variants may need to take lower or higher doses of warfarin, which lowers the risk of bleeding or insufficient anticoagulation. Warfarin dosing guided by pharmacogenomic factors may improve the safety and effectiveness of this vital anticoagulant medication, especially for patients suffering from diseases like deep vein thrombosis or atrial fibrillation.

Overall, the field of cardiology’s application of pharmacogenomics has changed the paradigm in cardiovascular care by providing individualized treatment plans that maximize the advantages of antiplatelet and anticoagulant medications while lowering the risks connected with universally applied approaches. This strategy not only enhances patient outcomes but also highlights how pharmacogenomics can greatly improve the treatment of cardiovascular diseases, which are a major global source of morbidity and mortality.

Psychiatry

In psychiatry, pharmacogenomics has become a game-changer, transforming the way that mental health disorders are treated by customizing psychotropic drugs to each patient’s unique genetic profile. Because mental health conditions are frequently complicated, patients’ reactions to psychotropic drugs can differ greatly. A major contributor to this variability is genetic variation. Healthcare professionals can determine particular genetic polymorphisms that affect drug metabolism, receptor binding, and neurotransmitter pathways — all of which affect how people react to psychotropic medications — by examining a patient’s genetic composition. With this information, clinicians can choose and dose drugs more intelligently, improving the accuracy and efficacy of mental health treatments. It lessens the tiresome process of trying and failing to find the best medication for every patient, lowering the risk of side effects and raising the standard of care as a whole. Pharmacogenomics’ ability to enable personalized treatment in psychiatry gives people with mental health issues fresh hope and represents a revolutionary change in the way we view mental healthcare.

Infectious diseases

Pharmacogenomics has great potential for treating infectious diseases, especially when it comes to creating and refining antibiotic and antiviral treatments and dealing with the urgent problem of drug resistance. Pharmacogenomics takes a personalized approach to treating infectious diseases by taking a person’s genetic composition into account. Genetic variations affect the effectiveness of antiviral therapies, which are medications used to treat viruses such as hepatitis C and HIV. By knowing a patient’s genetic profile, medical professionals can choose the best antiviral drugs and adjust the dosage to successfully suppress the virus while lowering the possibility of side effects.

Pharmacogenomics plays a key role in addressing the growing issue of antibiotic resistance in the context of antibiotic therapies. An individual’s reaction to antibiotics is significantly influenced by genetic factors, which also have an impact on drug-target interactions and drug metabolism. Healthcare professionals can customize antibiotic treatments for specific patients and guarantee that the most suitable antibiotics are prescribed at the appropriate dosages by identifying genetic variants that affect antibiotic efficacy and the risk of resistance.

Furthermore, pharmacogenomics can support the creation of plans to lessen medication resistance, a problem for world health. Researchers can create combination therapies that target multiple pathways or more effective drugs that are less likely to develop resistance by understanding the genetic basis of resistance mechanisms. This multimodal strategy prolongs the useful life of currently available antibiotics and antiviral drugs while also improving treatment efficacy.

Pain Management

In the field of pain management, pharmacogenomics has become a ground-breaking paradigm, especially when it comes to opioid responsiveness and the impact of genetic factors. Although the effectiveness and possibility of side effects of opioid medications can differ greatly from person to person, they are frequently prescribed for the treatment of both acute and chronic pain. Genetic variations are important factors in determining an individual’s response to opioids. Genes encoding opioid receptors, drug-metabolizing enzymes, and neurotransmitter systems can have polymorphisms that affect downstream cellular responses, opioid metabolism, and receptor binding. Pharmacogenomics enables doctors to customize painkillers to each patient’s specific genetic profile by identifying these genetic variables. The goal of this individualized approach to pain management is to maximize control over pain while lowering the possibility of side effects like opioid dependence, respiratory depression, or deaths from opioid overuse. Pharmacogenomics in pain management offers a way to strike a balance between efficient pain relief and the safe use of opioid medications, ultimately improving patient care and safety, as the opioid epidemic continues to pose a serious public health challenge.

The Gaps

Equitable access

  • Racial and ethnic differences, geographic location, and socioeconomic differences are a few of the numerous variables causing unequal access. Healthcare delivery disparities, lack of awareness, limited healthcare infrastructure, and affordability are some of the obstacles that patients from marginalized communities may encounter. These discrepancies highlight a larger issue of healthcare inequality, where those with more access and resources may benefit disproportionately from innovative therapies like pharmacogenomics. One of the most important ethical and policy challenges is addressing the unequal access to pharmacogenomic testing. In order to democratize the advantages of personalized medicine and enhance healthcare outcomes for all, it is necessary to make efforts in the areas of financial support, awareness-raising, training healthcare providers, infrastructure establishment, and ensuring that healthcare systems prioritize equitable access.

Integration into clinical practices

  • The incorporation of pharmacogenomics into clinical practice is a significant advancement in customized medicine. Healthcare professionals can optimize treatment outcomes while minimizing side effects by customizing drug therapies to each patient’s unique genetic profile, thanks to the abundance of genetic information available in this dynamic field. Nevertheless, there are obstacles to overcome before pharmacogenomics can be successfully incorporated into clinical care. The requirement for healthcare professionals to receive education and training in order to ensure that they can efficiently interpret and use genomic data is one of the main challenges. To further expedite the integration process, standardized guidelines and clinical decision support systems must be developed. To seamlessly integrate genomic data into patient care, healthcare systems must invest in the requisite infrastructure, such as genetic testing platforms and electronic health records. In addition, it is imperative that geneticists, pharmacists, and clinicians collaborate across disciplinary boundaries to guarantee the efficient conversion of genomic data into practical treatment choices. Pharmacogenomics is gaining momentum and its integration into clinical practice has the potential to provide patients with safer, more effective, and more precise drug therapies — ultimately changing the way we approach healthcare and enhancing patient outcomes and care.

Economic Challenges

  • Pharmacogenomic testing presents a variety of complex financial issues that have a big impact on patients, providers, and healthcare systems. The price of genomic testing itself is one of the main obstacles to economic growth. The cost of sequencing and interpreting a person’s genetic data can be high, placing a strain on healthcare systems and possibly preventing its general adoption. Moreover, there are problems with insurance coverage and reimbursement guidelines concerning pharmacogenomic testing. The cost of testing may not always be fully covered by insurance companies, leaving patients to pick up the tab. The fact that these tests are not reimbursed can discourage medical professionals from regularly providing them, especially when patients may find it difficult to pay for them. There is also an economic challenge in that cost-effectiveness studies are required. Pharmacogenomic testing can result in more effective and efficient treatment plans, but in order to support its inclusion in routine care, it is imperative to show the testing’s financial benefit. The time and resources needed to conduct these studies can be a barrier to funding and support from the healthcare system. Navigating the complicated world of healthcare financing, insurance policies, and resource allocation is necessary to address the financial challenges associated with pharmacogenomic testing. At the same time, it is important to highlight the long-term financial advantages of more individualized and efficient treatment approaches.

Genomic testing

  • The complexity and variability of genomic tests are one of the main obstacles. Targeted gene panels, whole-genome sequencing, whole-exome sequencing, and other methods are all included in the broad category of genomic testing; each has advantages and disadvantages of its own. Standardization and quality control may be hampered by this variability, which can result in variations in the calibre of testing, interpretation, and result communication. Moreover, the process of interpreting genomic data can be quite complex. It is a difficult task to sort through the vast amount of genetic variations in an individual’s genome and find variants that are clinically relevant. Since the clinical significance of many genetic variations is unknown, guidelines need to be updated and revised frequently. These variants’ clinical implications are frequently context-dependent and subject to change as new information becomes available. Furthermore, incorporating genomic data into clinical practice is a very difficult task. It’s possible that medical professionals lack the necessary training to comprehend and use genomic data in patient care. To close this knowledge gap and enable efficient decision-making based on genomic data, specialized genetic counsellors, multidisciplinary teams, and educational initiatives are required. The additional layer of complexity associated with genomic testing is the possibility of ethical and legal quandaries. As the field develops, ethical and legal considerations surrounding informed consent, data privacy, and the return of incidental findings will inevitably change. Another big problem is the issue of unequal access to genomic testing. Inequalities may emerge as a result of socioeconomic variables, geographic location, or low awareness, impeding fair access to the advantages of genomic testing. These differences could make already-existing healthcare disparities worse.

The most pressing problem is genomic testing and it will take cooperation from a range of stakeholders, including researchers, healthcare professionals, legislators, and regulatory agencies, to overcome these challenges in genomic testing. To guarantee that genomic testing develops into a useful and accessible tool for enhancing healthcare outcomes, standardization, quality control, the creation of guidelines, and investments in education and training are crucial procedures. The field’s continued advancements may be able to overcome these obstacles and fully realize the advantages of genomic testing in clinical practice and personalized medicine.

New Testing Process

There are several subcategories of genetic testing, each with a distinct purpose and application. The various kinds of genetic testing, such as pharmacogenomic, diagnostic, and prenatal testing, are covered in this section. It looks at the various roles that each type plays in healthcare and the conditions that each type seeks to treat. The first step in genetic testing is sample collection, which is essential to guarantee the precision and dependability of the results. Samples of blood, saliva, tissue, buccal swabs, urine, and other types are used. The sample to use depends on the particular test being run and the genetic data being sought. Due to their ease of use and patient acceptance, non-invasive sample collection techniques like buccal swabs and saliva have grown in favour in recent years. These samples can be used for a variety of purposes because they are rich sources of DNA and don’t require invasive procedures. The process of isolating DNA from a sample and keeping it apart from proteins, other biological materials, and components of cells is known as DNA extraction. A number of methods are used to extract DNA, such as:

  1. Chemical lysis: To release DNA from samples and break down cell membranes, a series of chemical reagents, including salt and detergents, are applied to the samples. This method’s efficiency and simplicity make it a popular choice in lab settings.
  2. Physical Lysis: In physical lysis procedures, membranes and cell walls are broken down mechanically to liberate DNA. Physical lysis techniques include mechanical grinding and high-speed homogenization.
  3. Heat and Enzymatic Lysis: These methods disassemble cell structures and liberate DNA by using high temperatures and enzymes like proteinase K. Tissues with paraffin-embedded in formalin are especially well suited for this technique.
  4. Spin Column-Based Extraction: Commercial kits bind DNA while extracting impurities using spin columns filled with specialty resins or membranes. In molecular biology, this approach is popular because it provides excellent purity.
  5. Magnetic Bead-Based Extraction: This method of extracting DNA selectively uses magnetic particles coated in agents that bind DNA. This method is ideal for high-throughput applications and is very effective.
  6. Polymerase Chain Reaction (PCR): PCR is a commonly used method for amplifying particular DNA segments, facilitating their detection and investigation. It is used for forensic testing, mutation detection, and targeted DNA analysis.
  7. Microarray-Based Testing: This technique allows for the simultaneous analysis of multiple genetic variants by using DNA probes affixed to a solid surface to analyze DNA segments. It is frequently used in studies involving genome-wide associations and gene expression analysis.
  8. Southern Blotting: This age-old method is used to identify particular DNA sequences in a complicated mixture. It can be used for gene mapping and restriction fragment length polymorphism (RFLP) analysis.

These methods for genetic testing and pharmacogenomic studies are lengthy and costly. This is the main reason why pharmacogenomics isn't mainstream yet. But scientists have now discovered a new way that we can personalize prescriptions cheaper and faster than ever, assuring that anyone around the world can have access to this technology. One recent discovery is an evolution of CRISPR technology, now being used to detect the unique genetic fingerprints encoded in virtually any DNA or RNA sequence in any organism or pathogen. This method turrets single molecule detection of nucleic acid targets. It works by programming special CRISPR-Cas enzymes to detect the presence of a specific nucleic acid signature in a sample through smart amplicon detection. Once researchers take a sample from a patient they then simplify the levels of RNA in it and add reports that are sensitive to cas13. Then an engineered crispr cas13 is added to the sample. This cas13 is programmed with an RNA that is designed to detect and bind to things. When this happens cas13 activates its cleaving mechanism and begins randomly slicing nearby RNA, including the reporters. Since each end of the reporter carries a different label, cas13 separates these two signatures creating a unique signa; within the sample. The sample is then applied to a commercial flow detection system. If the sample is negative the reporter remains intact and collects at the first detection line, if the sample is positive it collects at a different location making the line/diagnosis easy to spot.

Although this technology/solution is rapid, portable, flexible, and multiplexable it still isn't as convenient as doctors may like. Because they still need to perform amplifications on things such as the RNA or DNA. And it isn't ideal for testing certain genes.

So what's the solution how can we make genetic testing for pharmacogenomics as easy as possible? Well, we don't know for sure but I learned as much as I could and came up with a theoretical test idea that could have the potential to forever change the world of prescriptions.

My Spin On It

For scientists and medical professionals, urine — often regarded as a waste product of the body — contains a wealth of data. Urine analysis has been a vital diagnostic tool for centuries, offering important information about a person’s health, metabolism, and level of hydration, among other things. Here are some of the things this frequently disregarded bodily fluids can tell us:

  1. Through urine analysis, scientists can learn a great deal about a person’s metabolic processes. Urine contains several metabolic byproducts, one of which is urea, which is produced when proteins break down. The body’s overall health and the effectiveness of its protein metabolism can both be determined by measuring urea levels.
  2. Hormones are essential for controlling a number of body processes. Certain hormones, like cortisol and adrenaline, which represent the body’s reaction to stress, can be found in urine and are detected by scientists. Furthermore, hCG (human chorionic gonadotropin) is a crucial marker in pregnancy tests that helps doctors determine whether a woman is pregnant.
  3. Urine also includes extra salt and water-soluble vitamins and minerals. Elevations in specific waste products may suggest the presence of underlying medical conditions like kidney failure or dehydration.
  4. Urine testing for specific drug metabolites and toxins can reveal information about a person’s exposure to hazardous substances and medication compliance. This is especially crucial for people who take prescription drugs or have been exposed to pollutants in the environment.
  5. Proteins and blood cells can also be found in urine. Red blood cells in the urine can be a sign of urinary tract problems or kidney damage. Furthermore, excess proteins in the urine, or proteinuria, is a crucial indicator of kidney disease and may indicate other health issues.
  6. The levels of different electrolytes, including calcium, potassium, and sodium, which support the body’s electrolyte balance, can be determined by analyzing urine. Urine’s pH level can reveal information about the body’s acid-base equilibrium and be useful in the diagnosis of diseases like kidney stones.
  7. Urinary glucose levels may be a sign of uncontrolled diabetes. Although the kidneys normally reabsorb glucose, high blood sugar levels can cause glucose to spill over into the urine. Urine glucose monitoring is an essential diagnostic tool for diabetes management.
  8. White blood cells and bacteria can be found in urine, suggesting the possibility of kidney or urinary tract infections. Urine pathogen identification is essential for the diagnosis and management of infectious diseases.

As you see urine can tell us many things about the human body, but the one indicator this test is going to focus on is hormones. In a pregnancy test, the hCG hormone is present in the urine, it migrates towards and binds to the trace antibody. This complex will then continue to flow to the immobilized capture antibody, which will glow to indicate a positive test for pregnancy. This efficient and cheap testing process is the type we are aiming for, for a fast and convenient gene test.

Hormones are signalling molecules that act as messengers, passing information from one tissue to another and from cell to cell. Hormones have the ability to travel to distant tissues within the body, in contrast to neurotransmitters, which are limited to travelling across the tiny synaptic cleft.

But how do your hormones relate to your genes and medicine? Well, not only do genes code for hormones, but hormones also regulate genes. So with this in mind, although hormones don’t directly contribute to drug metabolism, some hormones have the ability to affect how active cytochrome P450 enzymes are. For instance:

  1. Hormones related to sex: Certain cytochrome P450 enzymes can be affected by the action of sex hormones, including testosterone and estrogen. Drug metabolism can be impacted by hormonal changes, such as those that take place during pregnancy or the menstrual cycle.
  2. Thyroid hormones: Thyroid hormones may have an impact on drug metabolism by influencing the expression of certain cytochrome P450 enzymes.
  3. Adrenal hormones: The adrenal glands’ production of cortisol has the ability to affect the way some cytochrome P450 enzymes function.

It’s crucial to remember that hormones have a complex effect on drug metabolism that varies based on the particular drug and enzyme involved. Additionally, a person’s capacity to metabolize specific drugs is largely determined by genetic factors.

These hormones and other similar ones would be tested the same way your pregnancy hormones are. The vision here is to essentially understand how much a patient can metabolize drugs and at what rate. So although creating a test that tests for certain hormones that have some relation to drug metabolism won't be as accurate, it still is something that can be done today.

Testing hormones through urine is possible, but the long-term goal is to somehow find a way to test genes, within minutes through easy tests such as urine. Although this is still being researched here are the genes that have the biggest effect on your medicine. And this is what genes would be tested.

CYP2D6

Among the P450 enzymes, CYP2D6 exhibits the greatest phenotypic variation. Polymorphisms in this gene have now been connected to several of the initial reports of differences in drug metabolism. Researchers studying the metabolism of two novel medications in the 1970s, debrisoquine and sparteine, discovered that a sizeable minority of people could not metabolize these substances. Subsequent research was able to demonstrate that the incapacity to metabolize these medications was a recessive trait [24], that it affected roughly 5–10% of Europeans, and that the incapacity to hydroxylate debrisoquine was linked to the incapacity to oxidize sparteine, indicating that the same enzyme was responsible for the metabolism of these two medications.

The metabolism of other medications was found to be impacted by this deficiency as well [26], and it was found that genetics had an impact on nortryptyline plasma steady-state concentrations in the past. The development of molecular techniques in the 1990s made it possible to sequence patients who exhibited the 2D6 poor metabolizer phenotype, identify multiple variations in the CYP2D6 sequence, and determine the functional effects of these variants on gene expression.

The current allelic dosage model of CYP2D6 metabolism resulted from further identification of patients with an ultrarapid metabolism of CYP2D6 substrates due to duplicated or multiple extra copies of a functional CYP2D6 gene and higher levels of enzyme expression: In contrast, extensive metabolizers have an increase in 2D6 function because they have duplicated or multiple extra copies of a functional CYP2D6 gene, while poor metabolizers are homozygous or compound heterozygotes for various loss-of-function alleles, intermediate metabolizers carry one defective allele, and normal metabolizers carry zero defective alleles.

25–30% of prescription drugs are metabolized by CYP2D6, which accounts for 2–5% of the liver’s total CYP content. Additionally, the brain, gastrointestinal system, and lungs contain trace amounts of it. Since 2D6 is believed to be the only non-inducible P450 in humans, genetics may account for a significant portion of inter-individual variation in its metabolism [38]. CYP2D6 has more than 80 allelic variants, the majority of which are extremely uncommon. Significant variations include CYP2D6*3 through CYP2D6*8 and CYP2D6*36, which have no functional activity (poor metabolizer phenotype); duplications of CYP2D6*1xN, CYP2D6*2, or CYP2D6*35; and CYP2D6*9, CYP2D6*10, CYP2D6*17, CYP2D6*29, and CYP2D6*41, which have decreased catalytic activity (intermediate metabolizer phenotype). Which results in the ultrarapid metabolizer phenotype and increased functional capacity [39].

Different ethnic groups have different distributions of extensive, ultrarapid, intermediate, and poor CYP2D6 metabolizers. For instance, only about 3% of Caucasians and 6% of Africans have the reduced-function *10 allele, whereas it is present in roughly 38% to 50% of East Asians. Similarly, the *17 allele with reduced function is almost exclusively found in Africans (roughly 21%). In contrast, only 10% to 12% of East Asians have the normal function *2 allele (extensive metabolizer phenotype), which is found in roughly 25% of Caucasians and 31% of Africans. Some of the ethnic variation in response to CYP2D6 substrate medications is probably explained by these differences.

CYP2C19

The same methods utilized to identify polymorphisms in CYP2D6 were also applied to the study of people who could not metabolize the anticonvulsant S-mephenytoin. Since sparteine and debrisoquine did not co-segregate, it was assumed that poor metabolizers of S-mephenytoin had a distinct defect. After being cloned in 1994, the gene encoding this enzyme is presently known as CYP2C19. Using S-mephenytoin as a metabolic probe of CYP2C19 activity, a significant ethnic variation in the poor metabolizer phenotype has been noted. Although only 2–6% of Caucasians exhibit the poor metabolizer phenotype, up to 20% of Asians do.

Numerous studies and a meta-analysis in patients with coronary artery disease receiving clopidogrel as an antiplatelet agent have brought attention to the impact of 2C19 metabolic variation. Prodrug clopidogrel is mostly oxidized by CYP2C19 to its active metabolite. The CYP2C19*2 allele causes a truncated protein and a splicing defect at position 681, making it difficult for patients to activate clopidogrel and increasing their risk of serious cardiovascular events. Nevertheless, the impact of CYP2C19 genotype on variability to clopidogrel response seems to be minimal, even in a genetically homogenous population.

Patients with one or more CYP2C19*17 alleles experienced a markedly higher incidence of bleeding complications during percutaneous coronary stent implantation, a phenotype consistent with extensive metabolism, according to a separate analysis involving over 1500 patients. Although the variability in clopidogrel response is only partially predicted by the CYP2C19*2 genotype and other non-genetic factors, the US Food and Drug Administration modified the clopidogrel label in 2009 to emphasize the influence of the CYP2C19 genotype on clopidogrel pharmacokinetics and clinical response. Prospective investigations are being conducted on the effects of clopidogrel at a higher dose or other antiplatelet agents like prasugrel in 2C19 poor metabolizers with coronary artery disease.

According to recent in vitro metabolomic studies, the majority of clopidogrel activation was attributed to the enzyme paraoxonase-1 (PON1), and the majority of the variability in drug biotransformation was explained by a variant within the gene called Q192R. These results were validated in a clinical cohort, in which QQ192 homozygotes exhibited lower concentrations of the active metabolite of clopidogrel and a significantly higher rate of coronary stent thrombosis in comparison to RR192 homozygotes. This illustration demonstrates one of the primary challenges with pharmacogenetic and genomic research: variable associations may result from variations in the phenotype endpoints and/or populations under investigation.

CYP2C9

Phenytoin and warfarin are just two examples of medications that are metabolized by CYP2C9, a human P450 that has numerous functionally significant polymorphisms. A wide range of other medications can also both induce and inhibit it. One to two percent of Caucasians exhibit the phenotype of a poor metabolizer.

Differences in 2C9 metabolism have played a significant role in explaining the significant interpatient variability in warfarin dosage requirements, one of the most often prescribed anticoagulants. Warfarin clearance has been known to be impacted by pharmacologic inhibition of 2C9 by medications such as fluconazole. Patients who needed a lower warfarin maintenance dose to achieve therapeutic anticoagulation were six times more likely to carry one or more CYP2C9 variant alleles. Poor metabolizers were also found to have a significantly higher risk of suffering a significant bleeding event while taking warfarin.

Subsequent research determined that the CYP2C9 genotype accounted for 10–20% of the variability in the warfarin dose requirement, while variations in the VKORC1 gene, which is involved in the pharmacodynamic pathway of the drug, could explain an additional 20–30% of the variability. In addition to other clinical factors (age, race, smoking, concomitant medications), two sizable prospective studies of pharmacogenetic-based warfarin dosing based on CYP2C9 and VKORC1 genotype are currently underway. The drug label has already included language regarding the impact of variation in CYP2C9 and VKORC1 on warfarin dosing.

CYP2A4/CYP3A5

About half of all drug metabolism, including that of immunosuppressives, chemotherapeutics, macrolide antibiotics, antidepressants, anxiolytics, antipsychotics, opiates, calcium channel blockers, and statins, is mediated by CYP3A4, which makes up about 30% of the hepatic P450 content. Although there is a significant range in activity levels between people, CYP3A4 does not seem to have any polymorphisms that lead to a lack of functional protein. The gene contains a number of polymorphisms, some of which do change the catalytic activity of the enzyme, but these changes have not yet had an effect on clinical treatment.

One reason for the wide range of CYP3A4 activity is the abundance of substrates that can either induce or inhibit the enzyme. CYP3A5, a related enzyme that shares a broad substrate specificity overlap with CYP3A4, exhibits variable expression, which contributes to the wide interpatient variability in 3A4 activity. Because of the CYP3A5*3 variant, which is an A to G polymorphism at position 6986 that results in the formation of a new splice site and a truncated protein, only 10% of Europeans express CYP3A5.Due in large part to the rare distribution of the CYP3A5*3 allele in African Americans, the proportion of CYP3A5 expressers in this population is significantly higher. The majority of polymorphisms in African Americans who do not express 3A5 are CYP3A5*6 and CYP3A5*7, which both cause aberrant splicing.

Status Quo

To understand what today's status quo for prescription drugs is, let's assume that you have restless leg syndrome and you book an appointment with your doctor.

For context, Willis-Ekbom disease, sometimes referred to as restless legs syndrome (RLS), is a neurological condition marked by an overwhelming desire to move the legs. RLS patients frequently have tingling, uncomfortable, or crawling sensations in their legs, which go away when they move. These feelings usually manifest when the individual is at rest, particularly in the evening and at night or during periods of inactivity, which makes it difficult to get to sleep or stay asleep. Although the precise cause of RLS is not always known, a mix of environmental and genetic factors are thought to be involved.

Peripheral neuropathy, pregnancy, kidney failure, iron deficiency, and certain medications are a few traits and conditions that can cause or worsen RLS. RLS can have a major influence on sleep and general quality of life, and its symptoms can range in intensity. Medication, managing underlying conditions, and lifestyle modifications are some possible treatment modalities. It’s critical to speak with a healthcare provider if you think you may have restless legs syndrome in order to receive an accurate diagnosis and suitable treatment.

So in this case let's assume that your treatment method for RLS is through medicine. And your doctor prescribes you medicine based on your weight, age, and symptom severity. So you take your prescriptions as your doctor prescribes them. Little do you know, you have a very high metabolism rate, and this affects the speed at which you process drugs, but you nor your doctor have any idea of this. Individuals with a high metabolism may process drugs more quickly, resulting in the drug being cleared from the body faster than intended. But since your prescriptions aren't personalized to your genes it's impossible to know of this.

The different processes affected by metabolism

But if we were able to make testing for metabolism rates more accessible by creating hormone testing strip tests or even one-day genetic testing strip tests. The next time you get your RLS prescription your dosage amount will be altered by the doctors through an understanding of your body and how it processes drugs. And not just your weight and age.

Optimizing drug efficacy through a better understanding and consideration of individual metabolism holds the potential to revolutionize healthcare outcomes on multiple fronts. By tailoring medication regimens to individuals based on their specific metabolism rates, we can not only enhance the effectiveness of everyday prescriptions but also usher in a new era of precision medicine with profound implications for patient well-being.

The customization of drug dosages according to metabolism can lead to more accurate therapeutic interventions. This individualized approach ensures that patients receive the right amount of medication needed to achieve the desired therapeutic effect. Consequently, it minimizes the risk of under-dosing, where medications may fall short in addressing health conditions, and over-dosing, which can lead to adverse reactions and toxicity.

Moreover, this personalized approach has the potential to save lives by reducing the likelihood of treatment failures and complications. In situations where time-sensitive interventions are crucial, such as in acute medical conditions or during emergency scenarios, ensuring the optimal effectiveness of prescribed medications becomes paramount. Precision dosing based on metabolism can contribute significantly to the rapid and efficient management of health crises.

Beyond the immediate benefits to individual patients, a more nuanced understanding of metabolism and its impact on drug response has broader public health implications. Tailoring prescriptions to individual metabolism may help mitigate the development of medication resistance, a growing concern in the medical community. By optimizing drug dosages for each patient, we can potentially reduce the selective pressure on pathogens or conditions to evolve resistance, preserving the effectiveness of existing medications and extending their lifespan.

I hope this article has provided you insight into the field of prescription drugs, and how much there is a need for assessable genetic testing to assure personalization. Although a simple strip test to test for genes isn't possible right now, one day it will be. But for now, why not create strip tests that test for more than just pregnancy hormones? If you enjoyed this article and would love to talk more about it feel free to email me @srajaya02@gmail.com

--

--