Pub Date : 2025-05-01Epub Date: 2025-08-03DOI: 10.1080/14622416.2025.2541402
Emytis Tavakoli, Leen Magarbeh, Samar Elsheikh, Amy Y Zhang, Arun K Tiwari, Clement C Zai, Martin Kronenbuerger, Heike Weber, Maike Scherf-Clavel, Stefan Unterecker, Jürgen Deckert, Daniel J Müller
Rationale: Preemptive pharmacogenetic (PGx) testing offers a promising approach to personalized antidepressant treatment by identifying genetic variations influencing drug metabolism. By focusing on CYP2D6 and CYP2C19 genes, this strategy aims to improve treatment response, minimize adverse effects, and optimize dosing in patients with depression.
Objectives and methods: This systematic review evaluates the effectiveness of preemptive PGx testing, primarily for CYP2D6 and CYP2C19, in enhancing antidepressant treatment outcomes. A comprehensive search of databases, including PubMed and Embase, was conducted to identify relevant studies. The review included randomized controlled trials and meta-analyses that assessed PGx testing in relation to treatment response and remission. Data on clinical outcomes were extracted and analyzed.
Results: PGx testing led to improved antidepressant response rates and remission at 8- and 12-week follow-ups compared to treatment-as-usual (TAU). However, where data were available, benefits were less pronounced after six months of follow-up. The findings suggest that PGx testing plays an important role in achieving earlier remission, while TAU requires a longer time to achieve remission.
Conclusion: Preemptive pharmacogenetic testing for CYP2D6 and CYP2C19 could enhance early antidepressant treatment outcomes, offering a valuable tool for personalized medicine. Further research is required to explore implementation challenges in diverse clinical settings.
{"title":"Evidence level for pharmacogenetic testing in antidepressant treatment: a systematic review.","authors":"Emytis Tavakoli, Leen Magarbeh, Samar Elsheikh, Amy Y Zhang, Arun K Tiwari, Clement C Zai, Martin Kronenbuerger, Heike Weber, Maike Scherf-Clavel, Stefan Unterecker, Jürgen Deckert, Daniel J Müller","doi":"10.1080/14622416.2025.2541402","DOIUrl":"10.1080/14622416.2025.2541402","url":null,"abstract":"<p><strong>Rationale: </strong>Preemptive pharmacogenetic (PGx) testing offers a promising approach to personalized antidepressant treatment by identifying genetic variations influencing drug metabolism. By focusing on CYP2D6 and CYP2C19 genes, this strategy aims to improve treatment response, minimize adverse effects, and optimize dosing in patients with depression.</p><p><strong>Objectives and methods: </strong>This systematic review evaluates the effectiveness of preemptive PGx testing, primarily for CYP2D6 and CYP2C19, in enhancing antidepressant treatment outcomes. A comprehensive search of databases, including PubMed and Embase, was conducted to identify relevant studies. The review included randomized controlled trials and meta-analyses that assessed PGx testing in relation to treatment response and remission. Data on clinical outcomes were extracted and analyzed.</p><p><strong>Results: </strong>PGx testing led to improved antidepressant response rates and remission at 8- and 12-week follow-ups compared to treatment-as-usual (TAU). However, where data were available, benefits were less pronounced after six months of follow-up. The findings suggest that PGx testing plays an important role in achieving earlier remission, while TAU requires a longer time to achieve remission.</p><p><strong>Conclusion: </strong>Preemptive pharmacogenetic testing for CYP2D6 and CYP2C19 could enhance early antidepressant treatment outcomes, offering a valuable tool for personalized medicine. Further research is required to explore implementation challenges in diverse clinical settings.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"295-309"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427529/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144776000","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-08-16DOI: 10.1080/14622416.2025.2547561
Thinagary Jegathesan, Syaratul Dalina Yusoff, Nor Asyikin Mohd Tahir
Inflammatory bowel disease (IBD) poses a major therapeutic challenge due to its chronic course and variable treatment responses. Genetic polymorphisms significantly influence drug efficacy, prompting this review to analyze their role in treatment non-response. A systematic literature search (PubMed/Scopus) using terms like "genetic polymorphism," "non-response," and "IBD" identified 25 relevant studies. Key findings linked TNFα (-308 G > A), TNFRSF1A, and TNFRSF1B polymorphisms to reduced anti-TNFα therapy effectiveness. TPMT variants correlated with thiopurine toxicity, while CYP3A4 and CYP3A5 polymorphisms altered tacrolimus metabolism. These genetic markers could serve as predictive tools for personalized IBD treatment, emphasizing the potential of genetic screening in clinical practice. Future research should integrate multi-omics approaches to refine predictive models and advance precision medicine, ultimately improving patient outcomes through tailored therapeutic strategies.
炎症性肠病(IBD)是一个主要的治疗挑战,由于其慢性病程和多变的治疗反应。遗传多态性显著影响药物疗效,促使本综述分析其在治疗无反应中的作用。系统的文献检索(PubMed/Scopus)使用“基因多态性”、“无反应”和“IBD”等术语确定了25项相关研究。关键发现将TNFα (-308 G > A)、TNFRSF1A和TNFRSF1B多态性与降低抗TNFα治疗效果联系起来。TPMT变异与硫嘌呤毒性相关,而CYP3A4和CYP3A5多态性改变了他克莫司的代谢。这些遗传标记可以作为IBD个性化治疗的预测工具,强调了遗传筛查在临床实践中的潜力。未来的研究应该整合多组学方法来完善预测模型和推进精准医学,最终通过量身定制的治疗策略改善患者的治疗效果。
{"title":"Genetic polymorphism associated with non-response to therapy in inflammatory bowel disease patients: a review.","authors":"Thinagary Jegathesan, Syaratul Dalina Yusoff, Nor Asyikin Mohd Tahir","doi":"10.1080/14622416.2025.2547561","DOIUrl":"10.1080/14622416.2025.2547561","url":null,"abstract":"<p><p>Inflammatory bowel disease (IBD) poses a major therapeutic challenge due to its chronic course and variable treatment responses. Genetic polymorphisms significantly influence drug efficacy, prompting this review to analyze their role in treatment non-response. A systematic literature search (PubMed/Scopus) using terms like \"genetic polymorphism,\" \"non-response,\" and \"IBD\" identified 25 relevant studies. Key findings linked TNFα (-308 G > A), TNFRSF1A, and TNFRSF1B polymorphisms to reduced anti-TNFα therapy effectiveness. TPMT variants correlated with thiopurine toxicity, while CYP3A4 and CYP3A5 polymorphisms altered tacrolimus metabolism. These genetic markers could serve as predictive tools for personalized IBD treatment, emphasizing the potential of genetic screening in clinical practice. Future research should integrate multi-omics approaches to refine predictive models and advance precision medicine, ultimately improving patient outcomes through tailored therapeutic strategies.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"271-283"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427478/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144859537","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-08-21DOI: 10.1080/14622416.2025.2547565
Eva González-Iglesias, Jesús Novalbos, Francisco Abad-Santos
Introduction: Rosuvastatin has become a good choice in the statin group because it has shown greater efficacy in reducing lipid levels than other statins, allowing patients to reach their therapeutic goal more quickly. To date, research has shown a broad relationship between the kinetics and efficacy of this drug and the phenotype of two transporters, OATP1B1 and BCRP, encoded by SLCO1B1 and ABCG2 genes. However, there are many other genes whose variation may also affect the treatment.
Objective: To conduct a systematic review including all information on the kinetics, measured by AUC and Cmax parameters; efficacy, by reduction in lipid levels and carotid intima-media thickness; and safety of rosuvastatin, by the occurrence of adverse events.
Methods: A search of the published literature was conducted in PubMed using the term "rosuvastatin AND pharmacogenetics" (PROSPERO code: CRD420251041953).
Results: A total of 37 articles were included, investigating 40 genes.
Conclusions: The importance of ABCG2 in drug kinetics and efficacy and of SLCO1B1 in kinetics is confirmed, as is the possible association of CYP3A5 with efficacy and safety and of APOC1 with efficacy. There are also positive results for other genes that should be studied further to confirm their association with rosuvastatin.
{"title":"The pharmacogenetics of rosuvastatin and implications for treatment: a systematic review.","authors":"Eva González-Iglesias, Jesús Novalbos, Francisco Abad-Santos","doi":"10.1080/14622416.2025.2547565","DOIUrl":"10.1080/14622416.2025.2547565","url":null,"abstract":"<p><strong>Introduction: </strong>Rosuvastatin has become a good choice in the statin group because it has shown greater efficacy in reducing lipid levels than other statins, allowing patients to reach their therapeutic goal more quickly. To date, research has shown a broad relationship between the kinetics and efficacy of this drug and the phenotype of two transporters, OATP1B1 and BCRP, encoded by <i>SLCO1B1</i> and <i>ABCG2</i> genes. However, there are many other genes whose variation may also affect the treatment.</p><p><strong>Objective: </strong>To conduct a systematic review including all information on the kinetics, measured by AUC and C<sub>max</sub> parameters; efficacy, by reduction in lipid levels and carotid intima-media thickness; and safety of rosuvastatin, by the occurrence of adverse events.</p><p><strong>Methods: </strong>A search of the published literature was conducted in PubMed using the term \"rosuvastatin AND pharmacogenetics\" (PROSPERO code: CRD420251041953).</p><p><strong>Results: </strong>A total of 37 articles were included, investigating 40 genes.</p><p><strong>Conclusions: </strong>The importance of <i>ABCG2</i> in drug kinetics and efficacy and of <i>SLCO1B1</i> in kinetics is confirmed, as is the possible association of <i>CYP3A5</i> with efficacy and safety and of <i>APOC1</i> with efficacy. There are also positive results for other genes that should be studied further to confirm their association with rosuvastatin.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"311-329"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427528/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144964808","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Colorectal cancer is one of the most prevalent cancers worldwide. We have thus focused on studies on the association of genetic polymorphisms with treatment response and adverse effects in colorectal cancer patients. This review highlights the inter-individual and inter-population genetic variations in ABC transporters and their association with variability in drug response. Several studies report the association of specific ABC transporter gene polymorphisms with the risk of developing colorectal carcinoma and the clinical outcomes in patients undergoing chemotherapy. It is important to understand genetic polymorphisms and genetic profiling for colorectal cancer patients in the context of personalized medication. This review is built on a comprehensive knowledge of the pharmacogenetics of colorectal carcinoma with a special focus on ABC polymorphism.
{"title":"Variation in the <i>ABC</i> transporter genes and association with clinical outcomes in colorectal cancer: a brief review.","authors":"Zari Salahud Din, Maryam Saqib, Sehrish Zafar, Nyla Munawar, Sagheer Ahmed","doi":"10.1080/14622416.2025.2534319","DOIUrl":"10.1080/14622416.2025.2534319","url":null,"abstract":"<p><p>Colorectal cancer is one of the most prevalent cancers worldwide. We have thus focused on studies on the association of genetic polymorphisms with treatment response and adverse effects in colorectal cancer patients. This review highlights the inter-individual and inter-population genetic variations in <i>ABC</i> transporters and their association with variability in drug response. Several studies report the association of specific <i>ABC</i> transporter gene polymorphisms with the risk of developing colorectal carcinoma and the clinical outcomes in patients undergoing chemotherapy. It is important to understand genetic polymorphisms and genetic profiling for colorectal cancer patients in the context of personalized medication. This review is built on a comprehensive knowledge of the pharmacogenetics of colorectal carcinoma with a special focus on <i>ABC</i> polymorphism.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"285-293"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427436/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144659875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aims: Dihydropyrimidine dehydrogenase (DPYD) plays a critical role in the metabolism of fluoropyrimidine-based chemotherapies such as 5-fluorouracil (5-FU), capecitabine, and tegafur. Genetic variants in DPYD can lead to partial or complete enzyme deficiency, resulting in toxic accumulation of these drugs and severe, sometimes fatal, adverse reactions.
Materials & methods: Whole‑exome sequencing data from 1,612 individuals were analyzed for DPYD variants. Variants were classified as decreased, no function, or potentially deleterious. Comparative allele frequency analysis was performed using global population datasets to identify inter-population differences.
Results and conclusion: A total of 95 individuals (5.3%) carried at least one decreased or no function DPYD variant, indicating a significant prevalence of clinically actionable genotypes in this Indian cohort. The most frequent variant, c.1236 G > A (HapB3), was found in 53 individuals (3.28%), supporting its relevance in the Indian population. Comparative analysis revealed distinct population patterns and novel variants not captured in current guidelines. These results support the urgency in implementing preemptive DPYD genotyping to avoid adverse drug reactions and further studies to gather evidence on rare and novel variants in the Indian population. To the best of our knowledge, this is the largest population analysis of DPYD variants from India.
目的:二氢嘧啶脱氢酶(DPYD)在以氟嘧啶为基础的化疗药物如5-氟尿嘧啶(5-FU)、卡培他滨和替加氟的代谢中起关键作用。DPYD的遗传变异可导致部分或完全酶缺乏,导致这些药物的毒性积累和严重的,有时是致命的不良反应。材料与方法:对来自1,612名个体的全外显子组测序数据进行DPYD变异分析。变异被分类为减少、无功能或潜在有害。使用全球种群数据集进行比较等位基因频率分析,以确定种群间差异。结果和结论:共有95人(5.3%)携带至少一种DPYD减少或无功能变异,表明该印度队列中临床可操作基因型的显著流行。最常见的变异是c.1236 G > A (HapB3),在53个个体(3.28%)中被发现,支持其在印度人群中的相关性。比较分析揭示了不同的种群模式和新的变异没有在当前的指南中被捕获。这些结果支持实施预防性DPYD基因分型以避免药物不良反应的紧迫性,并支持进一步研究以收集印度人群中罕见和新型变异的证据。据我们所知,这是对印度DPYD变体进行的最大规模的人口分析。
{"title":"Opportunistic analysis of clinically actionable DPYD gene variants in a germline testing cohort in India.","authors":"Rajdeep Raha, Rahul C Bhoyar, Ranendra Pratap Biswal, Radhika Venkatakrishnan, Prashant Rai, Eshaa Umashankar, Pooja Mahesh Kulkarni, Sridhar Sivasubbu, Vinod Scaria, Bani Jolly","doi":"10.1080/14622416.2025.2547563","DOIUrl":"10.1080/14622416.2025.2547563","url":null,"abstract":"<p><strong>Aims: </strong>Dihydropyrimidine dehydrogenase (DPYD) plays a critical role in the metabolism of fluoropyrimidine-based chemotherapies such as 5-fluorouracil (5-FU), capecitabine, and tegafur. Genetic variants in DPYD can lead to partial or complete enzyme deficiency, resulting in toxic accumulation of these drugs and severe, sometimes fatal, adverse reactions.</p><p><strong>Materials & methods: </strong>Whole‑exome sequencing data from 1,612 individuals were analyzed for DPYD variants. Variants were classified as decreased, no function, or potentially deleterious. Comparative allele frequency analysis was performed using global population datasets to identify inter-population differences.</p><p><strong>Results and conclusion: </strong>A total of 95 individuals (5.3%) carried at least one decreased or no function DPYD variant, indicating a significant prevalence of clinically actionable genotypes in this Indian cohort. The most frequent variant, c.1236 G > A (HapB3), was found in 53 individuals (3.28%), supporting its relevance in the Indian population. Comparative analysis revealed distinct population patterns and novel variants not captured in current guidelines. These results support the urgency in implementing preemptive DPYD genotyping to avoid adverse drug reactions and further studies to gather evidence on rare and novel variants in the Indian population. To the best of our knowledge, this is the largest population analysis of DPYD variants from India.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"231-236"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427532/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144855970","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-09-05DOI: 10.1080/14622416.2025.2546770
Lakshmi Aravindan, Sanjana Velu, Inesh Sivam, Sahana Sivam, Pooja S Tallapaneni, Ruthie Ressler, Michael Marks, Raman Venkataramanan, Rupa Radhakrishnan, Senthilkumar Sadhasivam
Buprenorphine, a semi-synthetic opioid, is used to treat Opioid Use Disorder (OUD) and as an analgesic. Buprenorphine's benefits over other opioids include longer duration of action, lower abuse potential, and a ceiling effect to serious adverse effects such as respiratory depression. This is a literature review of gene variants affecting the pharmacokinetics and pharmacodynamics of buprenorphine from databases, such as PubMed. Genetic polymorphisms related to metabolism and receptor binding of buprenorphine can alter the pharmacokinetics and response to buprenorphine. Specifically, genetic variants in CYP3A4, UGT2B7, OPRM1, PDYN, and SLC6A3 may affect metabolism and clinical response to buprenorphine. There is strong evidence linking polymorphism in Cytochrome P450 3A4 (CYP3A4) and UDP-Glucuronosyltransferase-2B7 (UGT2B7), enzymes involved in buprenorphine metabolism, with dosing requirements, treatment of OUD, and pain relief. Response to buprenorphine, an effective treatment for opioid use disorder and pain management, differs significantly based on several human genetic variations. However, there is currently insufficient evidence for the clinical significance of individualized treatment of buprenorphine based on genetic variants. Therefore, it is crucial that future research should prioritize evaluating the feasibility and clinical significance of individualized risk predictions and personalized dosing of buprenorphine.
{"title":"Pharmacogenomics of buprenorphine - a narrative review.","authors":"Lakshmi Aravindan, Sanjana Velu, Inesh Sivam, Sahana Sivam, Pooja S Tallapaneni, Ruthie Ressler, Michael Marks, Raman Venkataramanan, Rupa Radhakrishnan, Senthilkumar Sadhasivam","doi":"10.1080/14622416.2025.2546770","DOIUrl":"10.1080/14622416.2025.2546770","url":null,"abstract":"<p><p>Buprenorphine, a semi-synthetic opioid, is used to treat Opioid Use Disorder (OUD) and as an analgesic. Buprenorphine's benefits over other opioids include longer duration of action, lower abuse potential, and a ceiling effect to serious adverse effects such as respiratory depression. This is a literature review of gene variants affecting the pharmacokinetics and pharmacodynamics of buprenorphine from databases, such as PubMed. Genetic polymorphisms related to metabolism and receptor binding of buprenorphine can alter the pharmacokinetics and response to buprenorphine. Specifically, genetic variants in <i>CYP3A4, UGT2B7, OPRM1, PDYN</i>, and <i>SLC6A3</i> may affect metabolism and clinical response to buprenorphine. There is strong evidence linking polymorphism in Cytochrome P450 3A4 (<i>CYP3A4</i>) and UDP-Glucuronosyltransferase-2B7 (<i>UGT2B7</i>), enzymes involved in buprenorphine metabolism, with dosing requirements, treatment of OUD, and pain relief. Response to buprenorphine, an effective treatment for opioid use disorder and pain management, differs significantly based on several human genetic variations. However, there is currently insufficient evidence for the clinical significance of individualized treatment of buprenorphine based on genetic variants. Therefore, it is crucial that future research should prioritize evaluating the feasibility and clinical significance of individualized risk predictions and personalized dosing of buprenorphine.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"263-270"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427432/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145001239","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-08-07DOI: 10.1080/14622416.2025.2541404
Piotr Łacina, Jagoda Siemaszko, Katarzyna Bogunia-Kubik
Graft-versus-host disease (GvHD) remains a significant complication of allogeneic hematopoietic stem cell transplantation (HSCT), contributing to increased morbidity and mortality. Thus, continuous development of novel prophylactic and therapeutic approaches is crucial for GvHD prevention and management. With the current development of personalized medicine and a more patient-oriented approach, pharmacogenetics has the potential to become a critical factor in optimizing the prophylaxis and treatment of GvHD. This review explores the role of pharmacogenetic variants in prophylaxis and management of GvHD, including drugs such as calcineurin inhibitors, methotrexate, mycophenolate mofetil, cyclophosphamide, and corticosteroids. A deeper understanding of these genetic factors could help in developing a more personalized approach to GvHD management, improving clinical outcomes and minimizing adverse effects. This review underscores the need for more pharmacogenetic association studies, as well as for incorporating pharmacogenetic testing into clinical practice to refine drug selection and dosing strategies in GvHD treatment.
{"title":"Pharmacogenetics of graft-versus-host disease: a path to personalized medicine.","authors":"Piotr Łacina, Jagoda Siemaszko, Katarzyna Bogunia-Kubik","doi":"10.1080/14622416.2025.2541404","DOIUrl":"10.1080/14622416.2025.2541404","url":null,"abstract":"<p><p>Graft-versus-host disease (GvHD) remains a significant complication of allogeneic hematopoietic stem cell transplantation (HSCT), contributing to increased morbidity and mortality. Thus, continuous development of novel prophylactic and therapeutic approaches is crucial for GvHD prevention and management. With the current development of personalized medicine and a more patient-oriented approach, pharmacogenetics has the potential to become a critical factor in optimizing the prophylaxis and treatment of GvHD. This review explores the role of pharmacogenetic variants in prophylaxis and management of GvHD, including drugs such as calcineurin inhibitors, methotrexate, mycophenolate mofetil, cyclophosphamide, and corticosteroids. A deeper understanding of these genetic factors could help in developing a more personalized approach to GvHD management, improving clinical outcomes and minimizing adverse effects. This review underscores the need for more pharmacogenetic association studies, as well as for incorporating pharmacogenetic testing into clinical practice to refine drug selection and dosing strategies in GvHD treatment.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"247-262"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12558389/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144795074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-07-29DOI: 10.1080/14622416.2025.2539061
Kelsey J Cook, Nathan D Seligson, Benjamin Q Duong, Vicky L Funanage, Susan M Kirwin, Edward B Mougey, Stephen Lawless, David West, Pamela H Arn, Kathryn V Blake
Nemours Children's Health (NCH) is a large, multi-state pediatric health system in the United States with hospitals and outpatient locations across Delaware, New Jersey, Pennsylvania, and Florida. NCH has maintained consistent effort in pharmacogenomic (PGx) research through the Center for Pharmacogenomics and Translational Research since 2003. In 2018, NCH funded the development of a dedicated Clinical Pharmacogenomics Service (CPGxS) to support PGx testing and return of results across the NCH enterprise. The CPGxS consultant-based service provides clinical PGx testing and supports PGx-focused research. Over the past seven years, the CPGxS has developed an in-house PGx testing platform, integrated a PGx clinical decision support (CDS) system into the electronic medical record (EHR), established a comprehensive consultant clinic, and provided a broad variety of education opportunities for providers, patients, and trainees. In this institutional profile, we highlight the development of a PGx program and related efforts across a multi-state pediatric-specific healthcare system.
Nemours Children's Health (NCH)是美国一个大型的多州儿科卫生系统,在特拉华州、新泽西州、宾夕法尼亚州和佛罗里达州设有医院和门诊点。自2003年以来,NCH通过药物基因组学和转化研究中心一直致力于药物基因组学(PGx)研究。2018年,NCH资助了一个专门的临床药物基因组学服务(CPGxS)的开发,以支持整个NCH企业的PGx测试和结果返回。CPGxS基于顾问的服务提供临床PGx测试并支持以PGx为重点的研究。在过去的七年中,CPGxS开发了一个内部PGx测试平台,将PGx临床决策支持(CDS)系统集成到电子病历(EHR)中,建立了一个综合咨询诊所,并为提供者、患者和学员提供了广泛的教育机会。在这个机构简介中,我们强调了PGx计划的发展以及跨多州儿科特定医疗保健系统的相关努力。
{"title":"From bench to bedside: a spotlight on pharmacogenomics at Nemours Children's Health.","authors":"Kelsey J Cook, Nathan D Seligson, Benjamin Q Duong, Vicky L Funanage, Susan M Kirwin, Edward B Mougey, Stephen Lawless, David West, Pamela H Arn, Kathryn V Blake","doi":"10.1080/14622416.2025.2539061","DOIUrl":"10.1080/14622416.2025.2539061","url":null,"abstract":"<p><p>Nemours Children's Health (NCH) is a large, multi-state pediatric health system in the United States with hospitals and outpatient locations across Delaware, New Jersey, Pennsylvania, and Florida. NCH has maintained consistent effort in pharmacogenomic (PGx) research through the Center for Pharmacogenomics and Translational Research since 2003. In 2018, NCH funded the development of a dedicated Clinical Pharmacogenomics Service (CPGxS) to support PGx testing and return of results across the NCH enterprise. The CPGxS consultant-based service provides clinical PGx testing and supports PGx-focused research. Over the past seven years, the CPGxS has developed an in-house PGx testing platform, integrated a PGx clinical decision support (CDS) system into the electronic medical record (EHR), established a comprehensive consultant clinic, and provided a broad variety of education opportunities for providers, patients, and trainees. In this institutional profile, we highlight the development of a PGx program and related efforts across a multi-state pediatric-specific healthcare system.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"223-229"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427439/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144732745","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-05-01Epub Date: 2025-08-01DOI: 10.1080/14622416.2025.2541397
Jungsun Kim, Ji Min Han, Jeong Yee, Young Ju Kim, Suk Joo Choi, Hye Sun Gwak
Background: Nifedipine, a substrate of ATP-binding cassette superfamily G member 2 (ABCG2), is a tocolytic agent. This study aimed to construct a scoring system by exploring the association between ABCG2 polymorphisms and the treatment response of nifedipine in pregnant women.
Methods: This study was conducted at a tertiary medical center. Eight single-nucleotide polymorphisms from the ABCG2 gene were selected. The adjusted OR (AOR) were calculated using multivariable analysis. A risk scoring system was constructed by dividing the AOR of independent risk factors by the minimum AOR to predict treatment failure.
Results: A total of 69 patients were analyzed for treatment failure. Maternal age ≥33 years, contraction interval <4 minutes, and contraction intensity ≥20 mmHg were clinical risk factors associated with treatment failure. Patients with the ABCG2 rs2622604 and rs4148157 experienced an 8.6-fold and 4.3-fold increased risk of treatment failure, respectively. The predicted risks of treatment failure of patients who scored 0, 1-2, 3-4, 5-6, 7-8, and 9 points were 2.8%, 9.1%, 26.0%, 55.4%, 81.4%, and 93.9%, respectively.
Conclusion: This novel risk scoring system may aid clinicians in identifying patients less likely to respond to nifedipine, improving individualized care in preterm labor management.
{"title":"Predicting nifedipine responses in preterm labor: a risk score incorporating ABCG2 polymorphisms and clinical factors.","authors":"Jungsun Kim, Ji Min Han, Jeong Yee, Young Ju Kim, Suk Joo Choi, Hye Sun Gwak","doi":"10.1080/14622416.2025.2541397","DOIUrl":"10.1080/14622416.2025.2541397","url":null,"abstract":"<p><strong>Background: </strong>Nifedipine, a substrate of ATP-binding cassette superfamily G member 2 (ABCG2), is a tocolytic agent. This study aimed to construct a scoring system by exploring the association between <i>ABCG2</i> polymorphisms and the treatment response of nifedipine in pregnant women.</p><p><strong>Methods: </strong>This study was conducted at a tertiary medical center. Eight single-nucleotide polymorphisms from the <i>ABCG2</i> gene were selected. The adjusted OR (AOR) were calculated using multivariable analysis. A risk scoring system was constructed by dividing the AOR of independent risk factors by the minimum AOR to predict treatment failure.</p><p><strong>Results: </strong>A total of 69 patients were analyzed for treatment failure. Maternal age ≥33 years, contraction interval <4 minutes, and contraction intensity ≥20 mmHg were clinical risk factors associated with treatment failure. Patients with the <i>ABCG2</i> rs2622604 and rs4148157 experienced an 8.6-fold and 4.3-fold increased risk of treatment failure, respectively. The predicted risks of treatment failure of patients who scored 0, 1-2, 3-4, 5-6, 7-8, and 9 points were 2.8%, 9.1%, 26.0%, 55.4%, 81.4%, and 93.9%, respectively.</p><p><strong>Conclusion: </strong>This novel risk scoring system may aid clinicians in identifying patients less likely to respond to nifedipine, improving individualized care in preterm labor management.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"237-246"},"PeriodicalIF":1.9,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427512/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144760793","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2025-05-14DOI: 10.1080/14622416.2025.2504864
Yan Wang, XiaoLi Wang, Jun Xu, Didi Zhang, YongFeng Cao
Objective: To explore the impact of low miR-369-3p expression on the resistance of PC-9 cells to osimertinib.
Methods: The PC-9/AZD9291 cell line was established with osimertinib. Real-time quantitative PCR was employed to measure the expression levels of miR-369-3p in both PC-9 and PC-9/AZD9291 cells, and Western blotting was utilized to detect PTPN12 protein expression. A dual-luciferase reporter assay was conducted to investigate the target relationship between miR-369-3p and PTPN12. CCK8 assays were performed to evaluate the impact of miR-369-3p inhibition on drug resistance.
Results: In comparison to PC-9 cells, there was a significant upregulation of miR-369-3p and downregulation of PTPN12 protein in PC-9/AZD9291 cells (p < 0.05). Transfection with the miR-369-3p inhibitor resulted in decreased levels of miR-369-3p and increased expression of PTPN12 protein in PC-9/AZD9291 cells (p < 0.05). Conversely, transfection with miR-369 mimics led to an increase in miR-369-3p levels accompanied by a decrease in PTPN12 protein (p < 0.05). Notably, treatment with the miR-369-3p inhibitor lowered the IC50 value for PC-9/AZD9291 cells; however, following downregulation of PTPN12 using PTPN12-siRNA, sensitivity due to low expression of miR-369-3p was significantly diminished (p < 0.05).
Conclusion: miR-369-3p plays a crucial role in modulating drug resistance in PC-9/AZD9291 cells against osimertinib through regulation of PTPN12.
目的:探讨miR-369-3p低表达对PC-9细胞对奥希替尼耐药的影响。方法:用奥西替尼建立PC-9/AZD9291细胞系。Real-time定量PCR检测PC-9和PC-9/AZD9291细胞中miR-369-3p的表达水平,Western blotting检测PTPN12蛋白的表达。通过双荧光素酶报告基因检测来研究miR-369-3p与PTPN12之间的靶标关系。CCK8检测评估miR-369-3p抑制对耐药的影响。结果:与PC-9细胞相比,PC-9/AZD9291细胞中miR-369-3p显著上调,PTPN12蛋白下调(p p p p)。结论:miR-369-3p通过调控PTPN12在PC-9/AZD9291细胞对奥西替尼的耐药中起关键作用。
{"title":"miR-369-3p regulates the drug resistance of lung cancer cells by targeting PTPN12.","authors":"Yan Wang, XiaoLi Wang, Jun Xu, Didi Zhang, YongFeng Cao","doi":"10.1080/14622416.2025.2504864","DOIUrl":"10.1080/14622416.2025.2504864","url":null,"abstract":"<p><strong>Objective: </strong>To explore the impact of low miR-369-3p expression on the resistance of PC-9 cells to osimertinib.</p><p><strong>Methods: </strong>The PC-9/AZD9291 cell line was established with osimertinib. Real-time quantitative PCR was employed to measure the expression levels of miR-369-3p in both PC-9 and PC-9/AZD9291 cells, and Western blotting was utilized to detect PTPN12 protein expression. A dual-luciferase reporter assay was conducted to investigate the target relationship between miR-369-3p and PTPN12. CCK8 assays were performed to evaluate the impact of miR-369-3p inhibition on drug resistance.</p><p><strong>Results: </strong>In comparison to PC-9 cells, there was a significant upregulation of miR-369-3p and downregulation of PTPN12 protein in PC-9/AZD9291 cells (<i>p</i> < 0.05). Transfection with the miR-369-3p inhibitor resulted in decreased levels of miR-369-3p and increased expression of PTPN12 protein in PC-9/AZD9291 cells (<i>p</i> < 0.05). Conversely, transfection with miR-369 mimics led to an increase in miR-369-3p levels accompanied by a decrease in PTPN12 protein (<i>p</i> < 0.05). Notably, treatment with the miR-369-3p inhibitor lowered the IC50 value for PC-9/AZD9291 cells; however, following downregulation of PTPN12 using PTPN12-siRNA, sensitivity due to low expression of miR-369-3p was significantly diminished (<i>p</i> < 0.05).</p><p><strong>Conclusion: </strong>miR-369-3p plays a crucial role in modulating drug resistance in PC-9/AZD9291 cells against osimertinib through regulation of PTPN12.</p>","PeriodicalId":20018,"journal":{"name":"Pharmacogenomics","volume":" ","pages":"165-170"},"PeriodicalIF":1.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12203845/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144027339","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}