View Article

Abstract

The pharmacogenomics aims to unravel the way that human genetic variation affects drug efficacy and toxicity. Pain is a common symptom that can be complex to treat. Analgesic medications are the mainstay treatment, but there is wide interindividual variability in analgesic response and adverse effects. Pharmacogenomics is the study of inherited genetic traits that result in these individual responses to drugs. This narrative review will attempt to cover the current understanding of the pharmacogenomics of pain, examining common genes affecting metabolism of analgesic medications, their distribution throughout the body, and end organ effects.

Keywords

Pharmacogenetics, Pain. Medication, Pharmacokinetics, Pharmacodynamics, Inherited traits, Plan of Work, Method and Procedure.

Introduction

1] Pharmacogenetics: -

The Pharmacogenetics (PGx) refers to how genetic differences between individuals influence patient drug responses and disposition. Generally, genes affecting the treatment outcome can be divided into two broad categories. On the one hand, genes affecting pharmacodynamics, are based on variations in drug target receptors and downstream signal transduction (i.e. ?opioid receptor, OPRM1; enzyme catecholamine methyltransferase, COMT, etc.). On the other hand, genes affecting pharmacokinetics (PK) that affect drug metabolism and/or elimination (i.e. cytochrome P450 family of enzymes, enzymes responsible for glucuronidation, drug transporter proteins, COX enzymes, etc.) altering the relationship between drug dose and steady-state serum drug concentrations.

Pharmacodynamics:

The Some candidate genes are implied either directly (opioid receptors) or indirectly into the opioid transduction pathways when the signal is transmitted to a variety of effectors (e.g. adenylate cyclase or calcium and potassium ion channels named Kir3.2, KCNJ6).

       
            Pharmacogenetics testing for pain management.png
       

Figure.0.1 Pharmacogenetics testing for pain management.

2]Pain Medication:-

The Most clinical pain management options involve pharmacological interventions. Pain therapy has evolved over the years into a large specialty field. Ideal pain management approaches must provide adequate analgesia without excessive adverse effects. However, there are large interindividual differences in response to pain medications, concerning efficacy and the development of severe ADRs. Current pain management strategies are devised using the World Health Organization pain ladder, which begins with nonopioid medications, such as nonsteroidal anti-inflammatory drugs (NSAIDs), progresses to weak opioids, and culminates with strong opioids. Other pain medications include anticonvulsant drugs for neuralgia. Additionally, adjuvant therapies using antidepressant medications can aid in reducing chronic pain-associated anxiety. In Taiwan, based on the National Health Insurance Research Database, we found the most frequently used drugs for pain management. The field of pain medications is a valuable one to study germline variants using a pharmacogenomic approach, because of the considerable repercussions of these medications on the biological, psychological, sociological, and economic welfare of patients. Genetic studies have identified several polymorphic loci that govern the pharmacodynamics and kinetics of analgesic drugs. This review aims to highlight recent advances in the pharmacogenomics of pain medicine.

3]Pharmacogenomics in neuropathic pain treatment:- The Neuropathic pain is characterized by a complicated combination of positive (e.g. hyperalgesia and allodynia) and negative (e.g. hypoesthesia and hypoalgesia) symptoms and is refractory to conventional pharmacological agents, including morphine. Recently, Finnerup et al. proposed a revision of the NeuPSIG (Special Interest Group on Neuropathic Pain, from the International im Association for the Study of Pain) recommendations for the pharmacotherapy of neuropathic pain as follows: first-line treatment for tricyclic antidepressants, serotoninnoradrenalin reuptake inhibitors, pregabalin and gabapentin; second line for lidocaine patches, capsaicin high concentration patches and tramadol; third line for strong opioids and botulinum toxin A. They also stated that topical agents and botulinum toxin A are recommended only for peripheral neuropathic pain. In recent years, it has begun to be appreciated that the pathobiology of various neuropathic pain subtypes may differ. As an example, chemotherapy-induced peripheral neuropathy (CIPN) is a common secondary toxicity to neurotoxic anticancer drugs. The type of anticancer drug and the cumulative dose may impact in the incidence (possibly until 90% of patients for oxaliplatin), the symptoms, and the severity/grade of neuropathy. Recent metaanalysis demonstrated that among 31 studies (4179 patients), 68% (57.7–78.4) of patients suffered of CIPN 1 month after chemotherapy, 60% (36.4–81.6) after 3 months and 30% (6.4–

53.5) at 6 months or more. In the case of oxaliplatin, which is probably the most neurotoxic anticancer drug, neuropathy symptoms can last until several years after the end of the chemotherapy cycles (2 years in the study by André et al. and 8 years in the study of Yothers et al. Neurotoxicity mechanisms of anticancer drugs are not fully understood, but they may result from interactions with DNA, mitochondria, ion channels, glutamate neurotransmission and/or kinases, at various levels such as DRG (sensory neurons, Schwann cells, satellite cells), and spinal cord (neurons, glial cells) . CIPN is thus relatively distinct from other forms of neuropathic pain, including pathophysiology and symptomatology. After a systematic literature search identifying randomized controlled trials for the treatment of CIPN, it has been concluded the poor efficacy of these drugs in CIPN. Genetic factors may be important in predisposing patients to this adverse effect.

3.1] Pharmacodynamics:

The Chemotherapy-induced peripheral neuropathy and druginduced peripheral neuropathies (DIPNs) are encountered, including small-fiber involvement. The introduction of new diagnostic techniques, such as excitability studies, skin laser Doppler flowmetry, and PGx, holds promise for early detection and elucidation of underlying mechanisms. New approaches to improve functions and quality of life in CIPN patients are discussed. Apart from developing less neurotoxic anticancer therapies, there is still hope to identify chemoprotective agents, such as erythropoietin and substances involved in the endocannabinoid system, able to prevent or correct painful CIPNs.

Increased susceptibility to peripheral neurotoxicity after exposure to offending agents has been associated with polymorphisms in genes involved in the following pathways: chemotherapyinduced DNA   adduct repair, immune            function (cytotoxic Tlymphocyteassociated protein 4), also known as CD152 (cluster of differentiation 152), CTLA4 and compatible time-sharing system, reflexive coupling within Schwann cells (Gap Junction Protein, Epsilon 1, GJE1), drug binding (proteasome subunit beta 1, PSMB1) and neuron function (Transcription Factor, 4TCF4 and dynein cytoplasmic 1 intermediate chain 1, DYNC1I1), apoptosis, mitochondrial dysfunction, inflammation and oxidative stress scavengers such as glutathione S-transferase 1                    (GST1).

3.2] Pharmacokinetics

This Chemotherapy-induced peripheral neuropathy has been associated with variations in genes encoding for drug transporters, detoxification enzymes, genes involved in DNA repair mechanisms, and integrin B3 Leu33Pro polymorphism.           For instance, polymorphisms of the gene encoding ABCB1, have been suggested to partially explain the variability of taxaneinduced DIP. It should be noted that several other studies have been unable to identify relevant associations. Similarly, genetic variants of proteins involved in the metabolism of xenobiotics, for example, cytochrome 3A5, have been linked to increased risk of DIPN in children receiving vincristine. A range of polymorphisms has also been  identified  with  GWAS  in  association  with  oxaliplatin, paclitaxel, bortezomib, thalidomide, and vincristine. Other genes are implicated in the cellular transport of the molecules (such as ABCB1) or in their metabolism, which aims to convert lipophilic chemical compounds into more readily excreted hydrophilic products (mainly cytochrome isoforms CYP2D6, CYP3A4, and CYP2B6, and a glucuronosyltransferase implied in morphine metabolism, UGT2B7)

4] Translating pharmacogenomics discoveries into the clinic :- In general, PGx studies thus far in pain management have failed to yield evidence of improved clinical outcomes associated with knowledge of patient genotypes when prescribing pain medications. Genetic factors are thought to be responsible for approximately 12%–60% of response variance in opioid treatment, as evaluated in twin studies. Many genes have been studied to identify PGx markers in opioid treatment, including genes implied in opioids’ pharmacodynamics and PK. In this review, we focus on those genes, although numerous genes have been implicated in nociception and inflammation, and can participate in the analgesic dose requirement. On an individual level, there is a difference in the analgesic response to a given opioid. Various factors such as gender, age, and genetic variation can affect the analgesic response. Opioid analgesia can be predicted from the activity of reward-responsive brain regions during pain as well as subjects' trait-reward responsiveness ratings. Thus, studies have shown promising results regarding PGx as a diagnostic tool for predicting the individual response to a given opioid in experimental settings; however, in the clinic, it is a more complicated task to accomplish.

5] Factors that result in individual differences:-

It is Individual differences have been attributed to environmental, psychological, and genetic factors, including age, gender, patient weight ethnic origin, hepatic or renal function, type of surgery, surgical methods, duration of surgery, anxiety, and psychological distress. In this section, we describe each of these factors.

5.1] | Environmental factors:

It’s Various environmental factors have been reported. Among these, advancing age has been shown to reduce morphine requirements because age has been suggested to blunt peripheral nociceptive function. Additionally, fentanyl use for pain management is more in females than in males.

5.2] Psychological factors: The Many studies have reported associations between depression and pain. The prevalence of pain in depressed cohorts and the prevalence of depression in pain cohorts are higher than the prevalence when these conditions are examined individually. Indeed, the prevalence of pain in depressed patients was approximately 60%, and the intensity of depression was generally higher in patients with greater pain. Thus psychological distress, such as depressed mood and negative affect, can increase postoperative analgesic consumption. Moreover, patients who undergo emergency surgery may have less preoperative information and time for psychological preparation, resulting in increased requirements for postoperative analgesia.

AIM And Objective :-

The Pharmagenomics, the study of how genes affect a person’s response to drugs, plays a crucial role in the development of personalized medicine, particularly in pain management.

The aim of pharmagenomics in this context includes:

1. The Tailored Treatments: By analyzing an individual's genetic makeup, healthcare providers can select medications that are more likely to be effective for specific patients, reducing trialand-error prescribing.

2. Minimized Side Effects: Understanding genetic variations helps predict which patients may experience adverse reactions to certain medications, allowing for safer prescribing practices.

3. Optimized Dosage: Genetic insights can guide dose adjustments, ensuring that patients receive the right amount of medication for their unique metabolic profiles.

4. Targeted Therapies: Identifying specific genetic markers associated with pain pathways can lead to the development of targeted therapies that address the underlying mechanisms of pain.

5. Improved Outcomes: Personalized approaches can enhance overall treatment efficacy and patient satisfaction, leading to better management of chronic pain conditions.

6. Cost-effectiveness: By reducing the likelihood of ineffective treatments and hospitalizations due to side effects, pharmagenomics can contribute to more cost-effective pain management strategies.

7. Research Advancements: Continued research in this field can lead to new insights into the genetic basis of pain and the development of novel therapeutic agents. Overall, the integration of pharmagenomics into pain management aims to create a more precise, efficient, and patient-centered approach to treating pain.

Literature And Review:-

  1. Introduction to Pharmagenomics in Pain Management.

The Pharmagenomics combines pharmacology and genomics to develop personalized treatment strategies based on individual genetic profiles. Pain management, a complex and multifactorial area of medicine, benefits significantly from this approach. Variability in pain perception and response to analgesics can be attributed to genetic differences, making personalized medicine a promising avenue.

2. Genetic Variability and Pain Perception

Research has identified several genetic polymorphisms that influence pain sensitivity and response to analgesics. Key areas include:

• Opioid Receptor Genes: Variants in the OPRM1 gene, which encodes the mu-opioid receptor, have been linked to differences in opioid efficacy and risk of addiction.

• Cytochrome P450 Enzymes: Genetic variations in CYP450 enzymes affect drug metabolism, influencing how patients respond to medications like NSAIDs and antidepressants used for pain management.

• Other Pain-Related Genes: Genes such as COMT (involved in pain modulation) and TRPV1 (related to nociception) have been studied for their roles in pain pathways.

3. Personalized Analgesic Strategies

It’s Pharmagenomics data can guide clinicians in selecting the most appropriate analgesic regimen. For example:

• Opioids: Genetic testing for OPRM1 variants can inform decisions about which opioid to prescribe and at what dosage, potentially improving pain control while minimizing the risk of side effects.

• Non-Opioid Therapies: Understanding genetic factors influencing the effectiveness of non-opioid analgesics can help tailor treatments, such as anticonvulsants and antidepressants, for chronic pain conditions.

4. Clinical Implementation

The While the potential of pharmagenomics is clear, its integration into clinical practice faces challenges:

• Awareness and Education: Clinicians need training on interpreting genetic tests and applying results to clinical decision-making.

• Cost and Accessibility: Genetic testing can be expensive, and not all patients have access to these services.

• Regulatory Issues: The evolving landscape of genetic testing regulations poses hurdles for widespread adoption.

5. Future Directions

This Ongoing research is essential to further validate the clinical utility of pharmacogenomics testing in pain management. Future studies should focus on:

• Large-Scale Genomic Studies: Identifying additional genetic markers associated with pain and treatment responses.

• Integration into Clinical Guidelines: Developing standardized protocols for incorporating pharmacogenomics data into pain management strategies.

• Patient-Centered Approaches: Engaging patients in their treatment plans by providing information about how genetic factors can influence their pain management.

 Plan of Work:-

It is Pharmacogenomics in Personalized Medicine for Pain Management To investigate and implement pharmacogenomic approaches to enhance personalized pain management, improving efficacy and reducing adverse effects of pain medications.

Timeline:1 month

Activities:

- Review current literature on pharmacogenomics and its application in pain management. - Identify key genes associated with pain response and drug metabolism (e.g., CYP450 enzymes, OPRM1).

- Summarize findings on how genetic variations influence pain perception and treatment outcomes.

2. Stakeholder Engagement

- Timeline: 2 weeks - Activities:

- Engage healthcare professionals (physicians, pharmacists, genetic counsellors) to discuss the relevance of pharmacogenomics in pain management.

- Conduct surveys or interviews to assess current practices and perceptions about genetic testing in pain treatment.

3. Data Collection and Analysis

- Timeline: 2-3 months - Activities:

- Collect      genetic    samples           from patients  undergoing      pain management therapy.

- Analyze samples for relevant genetic variants.

- Correlate genetic data with patient responses to pain medications (efficacy, side effects).

4. Development

of Pharmacogenomic Guidelines - Timeline: 1 month - Activities:

- Develop    guidelines for      clinicians        on interpreting pharmacogenomic data in pain management.

- Create       a  decision-making         framework for selecting         pain medications based on genetic profiles.

 5. Implementation of Pharmacogenomic Testing

- Timeline: 3-6 months - Activities:

- Pilot a pharmacogenomic testing program in a clinical setting.

- Train         healthcare           staff     on        the use and      interpretation  of pharmacogenomic data.

- Monitor patient outcomes before and after the implementation of testing.

6. Evaluation and Feedback

- Timeline: 2 months - Activities:

- Evaluate the impact of pharmacogenomic testing on pain management outcomes

(pain relief, side effects, patient satisfaction).

- Gather       feedback from    patients           and clinicians to         assess usability and effectiveness of the guidelines.

7. Dissemination of Findings

-Timeline: 2 months - Activities:

-Prepare a report detailing the study’s findings and implications for clinical practice.

-Present       results     at         relevant conferences   and      publish in peerreviewed journals.

- Create       educational         materials         for patients      about pharmacogenomics and personalized pain management.

8. Future Directions

- Timeline: Ongoing - Activities:

- Explore additional genetic markers and their potential role in pain management. - Investigate the long-term effects of pharmacogenomic-guided therapies on pain management outcomes.

- Develop partnerships for broader implementation in different healthcare settings

Method and Procedure:-

1. Study Design

• Type: Observational cohort study.

• Setting: Clinical pain management centers.

• Participants: Patients with chronic pain receiving pharmacological treatment.

  1. Informed Consent

• Obtain informed consent from participants, explaining the purpose, procedures, potential risks, and benefits of genetic testing.

  1. Baseline Assessment

• Demographic Data: Collect information on age, sex, ethnicity, medical history, and pain characteristics.

• Pain Assessment: Use validated scales (e.g., Numeric Rating Scale, McGill Pain Questionnaire) to quantify pain levels and impact on quality of life.

• Medication History: Document current and past pain management medications, dosages, and treatment outcomes.

  1. Genetic Testing

• Sample Collection:

O Collect saliva or blood samples for DNA analysis.

O Use          appropriate         kits      for sample collection  and preservation.

• Genetic Analysis:

O Utilize next-generation sequencing (NGS) or polymerase chain reaction (PCR) to analyze specific genes associated with pain response (e.g., CYP450 variants,

OPRM1, COMT).

O Identify genetic variants that may influence drug metabolism, efficacy, and side effects.

  1. Data Integration and Interpretation

• Pharmacogenomic Profiling:

O Integrate genetic test results with patient demographics and clinical data. o Use bioinformatics tools to interpret genetic variants and predict responses to pain medications.

• Clinical Decision Support:

O Develop algorithms or decision-support tools for clinicians to guide medication selection based on pharmacogenomic profiles.

  1. Implementation of Pharmacogenomic Recommendations

• Medication Adjustment:

O Based on genetic findings, adjust medication regimens (e.g., select alternative medications, modify dosages).

• Monitoring:

O Regularly monitor patient responses to medication changes, including pain levels, side effects, and overall satisfaction.

  1. Follow-Up Assessment

• Short-Term Follow-Up: Assess patient outcomes 1-3 months after implementing pharmacogenomic-guided therapy. o Use pain assessment tools to evaluate changes in pain levels and quality of life. o Collect feedback on patient satisfaction and any adverse effects.

• Long-Term Follow-Up: Conduct assessments at 6 months and 12 months to evaluate sustained efficacy and safety.

  1. Data Analysis

• Statistical Methods:

O Use appropriate statistical analyses (e.g., regression analysis, ANOVA) to compare pain management outcomes before and after genetic testing. o Assess the correlation between genetic variants and clinical outcomes.

• Outcome Measures:

• Primary: Changes in pain intensity and quality of life.

O Secondary: Frequency of adverse drug reactions, patient satisfaction ratings.

  1. Reporting and Dissemination

• Compile findings into a comprehensive report.

• Present      results     at         medical conferences   and      submit for publication in peer-reviewed journals.

• Create       educational         materials         for patients      and      clinicians regarding the role of pharmacogenomics in pain management.

  1. Ethical Consider actions

• Ensure confidentiality of genetic information.

• Address potential ethical concerns related to genetic testing and its implications for treatment.

 

CONCLUSION

the clinically useful pharmacogenomics tests currently available for pain therapy are more direct toward at predicting drug toxicity or dose adjustment. More studies are needed to identify genetic variants that determine drug efficacy of pain medications. This area of research is likely rapidly accelerating with the reducing cost of nextgeneration sequencing and well-established bio banking system

ACKNOWLEDGEMENT: -

Authors are thankful to the “first and foremost, I would like to praise and thank God, the Almighty, who has granted countless blessings, knowledge, and opportunity to the writer. The Authors are thankful to the Management & Principal of Satyajeet College Of Pharmacy, Mehkar for Providing facilities to carry out the work. The authors are thankful to Project Guide Prof. Satish G. Lodhe. The authors are also thankful to Prof. Tejas J. Sharma Providing the ideas about the publication

REFERENCES

        1. Ana M. Peiró*, Beatriz Planelles, Gabriella Juhasz, György Bagdy, Frédéric Libert, Alain Eschalier, Jérôme Busserolles, Beata Sperlagh and Adrián Llerena Pharmacogenomics in pain treatmen
        2. Pharmacogenomics for personalized pain medicine Tai-Ming Ko 1, 2 , Chih-Shung Wong 3 , Jer-Yuarn Wu 1, 4 , Yuan-Tsong Chen 1, 5
        3. Nielsen LM, Olesen AE, Branford R, Christrup LL, Sato H, Drewes AM. Association between human pain-related genotypes and variability in opioid analgesia: an updated review. Pain Pract.2015;15(6):580–594.
        4. American Pain Society. Pain research funding inadequate in the face of soaring incidence and treatment costs [press release]. Chicago, IL: American Pain Society; 2013[May 10]. Available from: http://americanpainsociety.org/about- us/pressroom/painresearchfundinginadequate. Accessed May2, 2015.
        5. National Pain Summit Initiative. National Pain Strategy:Pain Management for All Australians. National Pain Summit Initiative; 2010. Available from: http://www.painaustralia.org.au/images/pain_australia/NPS/National Pain Strategy 2011.pdf. Accessed May 16, 2015.
        6. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999;286(5439):487–491.
        7. Pirmohamed M. Pharmacogenetics and pharmacogenomics. Br J Clin Pharmacol. 2001;52(4):345–347.
        8. Rollason V, Samer C, Piguet V, Dayer P, Desmeules J. Pharmacogenetics of analgesics: toward the individualization of prescription. Pharmacogenomics. 2008;9(7):905–933. 9]. Browne EA. The inheritance of an intrinsic abnormality of the red blood cell predisposing to drug-induced hemolytic anemia. Johns Hopkins Med J. 1957;101:115–118.
        9. Stamer UM, Zhang L, Stüber F. Personalized therapy in pain management: where do we stand? Pharmacogenomics. 2010;11(6):843–864.
        10. Kapur BM, Lala PK, Shaw JL. Pharmacogenetics of chronic pain management. Clin Biochem. 2014;47(13–14):1169–1187.
        11. Trescot AM, Faynboym S. A review of the role of genetic testing in pain medicine. Pain Physician. 2014;17(5):425–445.
        12. Foster A, Mobley E, Wang Z. Complicated pain management in a CYP450 2D6 poor metabolizer. Pain Pract. 2007;7(4):352–356.
        13. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev. 2009;41(2): 89–295.
        14. Armstrong SC, Cozza KL. Pharmacokinetic drug interactions of morphine, codeine, and their derivatives: theory and clinical reality, Part II. Psychosomatics. 2003;44(6):515–520. 16]. Bernard S, Neville KA, Nguyen AT, Flockhart DA. Interethnic differences in genetic polymorphisms of CYP2D6 in the US population: clinical implications. Oncologist. 2006;11(2):126–135.
        15. Madadi P, Ross CJ, Hayden MR, et al. Pharmacogenetics of neonatal opioid toxicity following maternal use of codeine during breastfeeding: a case-control study. Clin Pharmacol Ther. 2009;85(1):31–35.
        16. Orliaguet G, Hamza J, Couloigner V, et al. A case of respiratory depression in a child with ultrarapid CYP2D6 metabolism after tramadol. Pediatrics. 2015;135(3):e753–e755.
        17. Kirchheiner J, Nickchen K, Bauer M, et al. Pharmacogenetics of antidepressants and antipsychotics: the contribution of allelic variations to the phenotype of drug response. Mol Psychiatry. 2004;9(5):442–473
        18. Visser LE, van Schaik RH, van Vliet M, et al. Allelic variants of cytochrome P450 2C9 modify the interaction between nonsteroidal anti-inflammatory drugs and coumarin anticoagulants. Clin Pharmacol Ther. 2005;77(6):479–485.
        19. García-Martín E, Martínez C, Ladero JM, Agúndez JA. Interethnic and intraethnic variability of CYP2C8 and CYP2C9 polymorphisms in healthy individuals. Mol Diagn Ther. 2006;10(1):29–40.
        20. Rosemary J, Adithan C. The pharmacogenetics of CYP2C9 and CYP2C19: ethnic variation and clinical significance. Curr Clin Pharmacol. 2007;2(1):93–109.
        21. Pilotto A, Seripa D, Franceschi M, et al. Genetic susceptibility to nonsteroidal antiinflammatory drug-related gastroduodenal bleeding: role of cytochrome P450 2C9 polymorphisms. Gastroenterology.2007;133(2):465–471.
        22. Lundblad MS, Ohlsson S, Johansson P, Lafolie P, Eliasson E. Accumulation of celecoxib with a 7-fold higher drug exposure in individuals homozygous for CYP2C9*3. Clin Pharmacol Ther.2006;79(3):287–288.
        23. Stempak D, Bukaveckas BL, Linder M, Koren G, Baruchel S. Cytochrome P450 2C9 genotype: impact on celecoxib safety and pharmacokinetics in a pediatric patient. Clin Pharmacol Ther.2005;78(3):309–310.
        24. Carbonell N, Verstuyft C, Massard J, et al. CYP2C9*3 loss-of-function allele is associated with acute upper gastrointestinal bleeding related to the use of NSAIDs other than aspirin. Clin Pharmacol Ther.2010;87(6):693–698.
        25. Trescot AM. Genetics and implications in perioperative analgesia. Best Pract Res Clin Anaesthesiol. 2014;28(2):153–166.
        26. Wadelius M, Chen LY, Lindh JD, et al. The largest prospective warfarintreated cohort supports genetic forecasting. Blood. 2009;113(4):784–792.
        27. Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther. 2008;83(4):559–566.
        28. Cohen M, Sadhasivam S, Vinks AA. Pharmacogenetics in perioperative medicine. Curr Opin Anaesthesiol. 2012;25(4):419–427.31. Meineke I, Freudenthaler S, Hofmann U, et al. Pharmacokinetic modelling of morphine, morphine-3-glucuronide and morphine6glucuronide in plasma and cerebrospinal fluid of neurosurgical patients after shortterm infusion of morphine. Br J Clin Pharmacol. 2002;54(6):592–603.
        29. Zwisler ST, Enggaard TP, Noehr-Jensen L, et al. The antinociceptive effect and adverse drug reactions of oxycodone in human experimental pain in relation to genetic variations in the OPRM1 and ABCB1 genes. Fundam Clin Pharmacol. 2010;24(4):517–524.

Reference

  1. Ana M. Peiró*, Beatriz Planelles, Gabriella Juhasz, György Bagdy, Frédéric Libert, Alain Eschalier, Jérôme Busserolles, Beata Sperlagh and Adrián Llerena Pharmacogenomics in pain treatmen
  2. Pharmacogenomics for personalized pain medicine Tai-Ming Ko 1, 2 , Chih-Shung Wong 3 , Jer-Yuarn Wu 1, 4 , Yuan-Tsong Chen 1, 5
  3. Nielsen LM, Olesen AE, Branford R, Christrup LL, Sato H, Drewes AM. Association between human pain-related genotypes and variability in opioid analgesia: an updated review. Pain Pract.2015;15(6):580–594.
  4. American Pain Society. Pain research funding inadequate in the face of soaring incidence and treatment costs [press release]. Chicago, IL: American Pain Society; 2013[May 10]. Available from: http://americanpainsociety.org/about- us/pressroom/painresearchfundinginadequate. Accessed May2, 2015.
  5. National Pain Summit Initiative. National Pain Strategy:Pain Management for All Australians. National Pain Summit Initiative; 2010. Available from: http://www.painaustralia.org.au/images/pain_australia/NPS/National Pain Strategy 2011.pdf. Accessed May 16, 2015.
  6. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999;286(5439):487–491.
  7. Pirmohamed M. Pharmacogenetics and pharmacogenomics. Br J Clin Pharmacol. 2001;52(4):345–347.
  8. Rollason V, Samer C, Piguet V, Dayer P, Desmeules J. Pharmacogenetics of analgesics: toward the individualization of prescription. Pharmacogenomics. 2008;9(7):905–933. 9]. Browne EA. The inheritance of an intrinsic abnormality of the red blood cell predisposing to drug-induced hemolytic anemia. Johns Hopkins Med J. 1957;101:115–118.
  9. Stamer UM, Zhang L, Stüber F. Personalized therapy in pain management: where do we stand? Pharmacogenomics. 2010;11(6):843–864.
  10. Kapur BM, Lala PK, Shaw JL. Pharmacogenetics of chronic pain management. Clin Biochem. 2014;47(13–14):1169–1187.
  11. Trescot AM, Faynboym S. A review of the role of genetic testing in pain medicine. Pain Physician. 2014;17(5):425–445.
  12. Foster A, Mobley E, Wang Z. Complicated pain management in a CYP450 2D6 poor metabolizer. Pain Pract. 2007;7(4):352–356.
  13. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev. 2009;41(2): 89–295.
  14. Armstrong SC, Cozza KL. Pharmacokinetic drug interactions of morphine, codeine, and their derivatives: theory and clinical reality, Part II. Psychosomatics. 2003;44(6):515–520. 16]. Bernard S, Neville KA, Nguyen AT, Flockhart DA. Interethnic differences in genetic polymorphisms of CYP2D6 in the US population: clinical implications. Oncologist. 2006;11(2):126–135.
  15. Madadi P, Ross CJ, Hayden MR, et al. Pharmacogenetics of neonatal opioid toxicity following maternal use of codeine during breastfeeding: a case-control study. Clin Pharmacol Ther. 2009;85(1):31–35.
  16. Orliaguet G, Hamza J, Couloigner V, et al. A case of respiratory depression in a child with ultrarapid CYP2D6 metabolism after tramadol. Pediatrics. 2015;135(3):e753–e755.
  17. Kirchheiner J, Nickchen K, Bauer M, et al. Pharmacogenetics of antidepressants and antipsychotics: the contribution of allelic variations to the phenotype of drug response. Mol Psychiatry. 2004;9(5):442–473
  18. Visser LE, van Schaik RH, van Vliet M, et al. Allelic variants of cytochrome P450 2C9 modify the interaction between nonsteroidal anti-inflammatory drugs and coumarin anticoagulants. Clin Pharmacol Ther. 2005;77(6):479–485.
  19. García-Martín E, Martínez C, Ladero JM, Agúndez JA. Interethnic and intraethnic variability of CYP2C8 and CYP2C9 polymorphisms in healthy individuals. Mol Diagn Ther. 2006;10(1):29–40.
  20. Rosemary J, Adithan C. The pharmacogenetics of CYP2C9 and CYP2C19: ethnic variation and clinical significance. Curr Clin Pharmacol. 2007;2(1):93–109.
  21. Pilotto A, Seripa D, Franceschi M, et al. Genetic susceptibility to nonsteroidal antiinflammatory drug-related gastroduodenal bleeding: role of cytochrome P450 2C9 polymorphisms. Gastroenterology.2007;133(2):465–471.
  22. Lundblad MS, Ohlsson S, Johansson P, Lafolie P, Eliasson E. Accumulation of celecoxib with a 7-fold higher drug exposure in individuals homozygous for CYP2C9*3. Clin Pharmacol Ther.2006;79(3):287–288.
  23. Stempak D, Bukaveckas BL, Linder M, Koren G, Baruchel S. Cytochrome P450 2C9 genotype: impact on celecoxib safety and pharmacokinetics in a pediatric patient. Clin Pharmacol Ther.2005;78(3):309–310.
  24. Carbonell N, Verstuyft C, Massard J, et al. CYP2C9*3 loss-of-function allele is associated with acute upper gastrointestinal bleeding related to the use of NSAIDs other than aspirin. Clin Pharmacol Ther.2010;87(6):693–698.
  25. Trescot AM. Genetics and implications in perioperative analgesia. Best Pract Res Clin Anaesthesiol. 2014;28(2):153–166.
  26. Wadelius M, Chen LY, Lindh JD, et al. The largest prospective warfarintreated cohort supports genetic forecasting. Blood. 2009;113(4):784–792.
  27. Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther. 2008;83(4):559–566.
  28. Cohen M, Sadhasivam S, Vinks AA. Pharmacogenetics in perioperative medicine. Curr Opin Anaesthesiol. 2012;25(4):419–427.31. Meineke I, Freudenthaler S, Hofmann U, et al. Pharmacokinetic modelling of morphine, morphine-3-glucuronide and morphine6glucuronide in plasma and cerebrospinal fluid of neurosurgical patients after shortterm infusion of morphine. Br J Clin Pharmacol. 2002;54(6):592–603.
  29. Zwisler ST, Enggaard TP, Noehr-Jensen L, et al. The antinociceptive effect and adverse drug reactions of oxycodone in human experimental pain in relation to genetic variations in the OPRM1 and ABCB1 genes. Fundam Clin Pharmacol. 2010;24(4):517–524.

Photo
Farhan Khan
Corresponding author

Satyajeet College Of Pharmacy, Mehkar

Photo
Kasim Bhuriwale
Co-author

Satyajeet College of pharmacy, Mehkar

Photo
Adil Shah Bad Shah
Co-author

Satyajeet College of pharmacy, Mehkar

Photo
Dr. Shivshankar Mhaske
Co-author

Satyajeet College of pharmacy, Mehkar

Photo
Satish Lodhe
Co-author

Satyajeet College of pharmacy, Mehkar

Farhan Khan*, Kasim Bhuriwale, Adil Shah Bad Shah, Dr. Shivshankar Mhaske, Satish Lodhe, Pharmacogenomics: Personalized Medicine In Pain Management, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 1, 517-526. https://doi.org/10.5281/zenodo.14615790

More related articles
Formulation And Evaluation Of Skin Infection Care ...
Shashank Tiwari, Sushil Kumar Pal, Vaibhav Raj Pandey, ...
Role of lifestyle Interventions in Diabetes Preven...
Manobalan M, Selvabalaji S , Rashvin CV , Haribaskar R, NaveenKum...
Extraction, Phytochemical Investigation and Antimi...
Ganesh D. Barkade, Akshata B. Sinare, Mayur G. Tandale, Rutuja B....
Hemophilia Gene Therapy: The Dawn Of A New Era In Treatment ...
Kanchan Thorat , Ajit B. Tuwar, Dr. Megha Salve, ...
Water Quality Monitoring In SPMVV By IOT Tools ...
K. Swathi, U. Madhavi, D. Kalyani, P. Sai Pratyusha, ...
Related Articles
A Review On Preparation Of Herbal Shampoo, Uses And Products Available In Market...
Shital Rohidas kamble , Sameera M. Shaikh, Rauf Shaikh, Pradnya S. Shingare, Vaishnavi V. Shisodiya,...
Terbinafine Tablet Quality Control Test Report and Pharmacology: Compliance with...
Aditya Mane, Anirudha Patil, Tejashri Kamble, Nilesh chougule, ...
Anti Ulcer Activity Of Medicinal Plants ...
Gauri S Salunke , Gayatri B Gaware , Rupali N Tarde , Payal A Pangude , Sayyed Afnan kirmani, ...
Formulation And Evaluation Of Skin Infection Care Cream...
Shashank Tiwari, Sushil Kumar Pal, Vaibhav Raj Pandey, ...
More related articles
Formulation And Evaluation Of Skin Infection Care Cream...
Shashank Tiwari, Sushil Kumar Pal, Vaibhav Raj Pandey, ...
Role of lifestyle Interventions in Diabetes Prevention and Management ...
Manobalan M, Selvabalaji S , Rashvin CV , Haribaskar R, NaveenKumar Mannar, ...
Extraction, Phytochemical Investigation and Antimicrobial Activity of Plumeria o...
Ganesh D. Barkade, Akshata B. Sinare, Mayur G. Tandale, Rutuja B. Sonawane, Rutuja R. Sumbe, ...
Formulation And Evaluation Of Skin Infection Care Cream...
Shashank Tiwari, Sushil Kumar Pal, Vaibhav Raj Pandey, ...
Role of lifestyle Interventions in Diabetes Prevention and Management ...
Manobalan M, Selvabalaji S , Rashvin CV , Haribaskar R, NaveenKumar Mannar, ...
Extraction, Phytochemical Investigation and Antimicrobial Activity of Plumeria o...
Ganesh D. Barkade, Akshata B. Sinare, Mayur G. Tandale, Rutuja B. Sonawane, Rutuja R. Sumbe, ...