1
|
Dung PT, Su HX, Tue NC, Ben NH, Phuong NM, Tran TN, Nghia PB, Van DT, Dung NTT, Vinh HT, Rostaing L, Toan PQ. Predictive value of tacrolimus concentration/dose ratio in first post-transplant week for CYP3A5-polymorphism in kidney-transplant recipients. World J Transplant 2025; 15:103247. [DOI: 10.5500/wjt.v15.i2.103247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Tacrolimus (TAC) is metabolized primarily by the CYP3A-encoded enzyme family (CYP3A4, CYP3A5, and CYP3A7). Individuals expressing the CYP3A51 allele are considered fast metabolizers and generally require higher TAC doses to reach therapeutic levels.
AIM To evaluate the predictive value of the TAC concentration-to-dose (C0/D) ratio for identifying CYP3A5 polymorphisms in renal transplant recipients.
METHODS Eighty-six de novo kidney transplant recipients with TAC-based immunosuppression from the Department of Nephrology and Dialysis at Military Hospital 103 (Hanoi, Vietnam) were included in this retrospective study. Blood samples were collected within the first week post-transplantation to monitor TAC levels and to perform genotyping for CYP3A5 genetic polymorphisms.
RESULTS The CYP3A53/3 genotype was identified in 37 patients (43%), CYP3A51/3 in 40 patients (46.5%), and CYP3A51/1 in 9 patients (10.5%). Patients carrying the CYP3A51/3 or CYP3A51/1 genotype, classified as fast metabolizers (CYP3A5 expressers), had significantly lower TAC C0 concentrations and C0/D ratios compared to slow metabolizers (CYP3A53/3 genotype) at multiple time points during follow-up (all P < 0.001). Notably, the TAC C0/D ratio obtained on day 1 (0.91) was shown to predict CYP3A5 polymorphism with a sensitivity of 84.6% and a specificity of 84.6%.
CONCLUSION This study demonstrates that the TAC C0/D ratio provides a reliable predictive value for CYP3A5 polymorphisms, which can be used to individualize TAC dosing in renal transplant recipients in Vietnam and other low-income countries.
Collapse
Affiliation(s)
- Pham-Thai Dung
- Center of Critical Care Medicine, Emergency and Clinical Toxicology, Military Hospital, Hanoï 100000, Viet Nam
| | - Hoang-Xuan Su
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoï 100000, Viet Nam
| | - Nguyen-Chi Tue
- Department of Post-Transplant Intensive Care and Treatment, Center of Organ Transplantation - Military Hospital, Hanoï 100000, Viet Nam
| | - Nguyen-Huu Ben
- Department of Occupational Medicine, Vietnam Military Medical University, Hanoï 100000, Viet Nam
| | - Nguyen-Minh Phuong
- Department of Occupational Medicine, Vietnam Military Medical University, Hanoï 100000, Viet Nam
| | - Tuan-Ngoc Tran
- Department of Military Medical Command and Organization, Vietnam Military Medical University, Hanoï 100000, Viet Nam
| | - Phan-Ba Nghia
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
| | - Diem-Thi Van
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
| | - Nguyen Thi-Thuy Dung
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
| | - Hoang-Trung Vinh
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
| | - Lionel Rostaing
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
- Department of Nephrology, Hemodialysis, Apheresis, and Kidney Transplantation, Grenoble University Hospital, Grenoble 38043, Auvergne-Rhone-Alpes, France
| | - Pham-Quoc Toan
- Department of Nephrology and Dialysis, Military Hospital, Hanoï 100000, Viet Nam
| |
Collapse
|
2
|
Xajil-Ramos LY, Gándara-Mireles JA, Vargas Rosales RJ, Sánchez García OK, Ruano Toledo AM, Aldana de la Cruz AK, Lares-Asseff I, Patrón-Romero L, Almanza-Reyes H, Lou-Meda R. Impact of CYP3A5 1* and 3* single nucleotide variants on tacrolimus pharmacokinetics and graft rejection risk in pediatric kidney transplant patients. Front Pharmacol 2025; 16:1592134. [PMID: 40432883 PMCID: PMC12107630 DOI: 10.3389/fphar.2025.1592134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Tacrolimus, a calcineurin inhibitor, is widely used to prevent allograft rejection in kidney transplant recipients. Its metabolism is predominantly mediated by the cytochrome P450 3A5 (CYP3A5) enzyme, and single nucleotide variants (SNVs) within intron 3 of the CYP3A5 gene are strongly associated with interindividual variability in enzyme expression and activity. These SNVs can generate a cryptic splice site, resulting in either preserved enzymatic function classified as expressers (CYP3A5 *1/*1 and *1/*3) or loss of function, classified as non-expressers (CYP3A5 *3/*3). Differential expression of CYP3A5 contributes to variability in tacrolimus pharmacokinetics and clinical outcomes, including graft rejection and therapeutic efficacy. In this study, we evaluated three pharmacokinetic parameters: trough concentration (TAC-C0), weight-adjusted daily dose (TAC-D, mg/kg), and dose-normalized trough concentration (TAC-C0/D). One-way ANOVA was used to assess differences in these parameters between CYP3A5 expressers and non-expressers. Additionally, Poisson regression was performed to examine associations between clinical/genetic variables and the incidence rate of acute rejection events. Genotyping was conducted in 45 pediatric kidney transplant recipients. The CYP3A5 *3/*3 genotype was most prevalent (66.7%), followed by *1/*3 (26.7%) and *1/*1 (6.7%). During the 6-month post-transplant period, CYP3A5 expressers required significantly higher tacrolimus doses to achieve target trough levels. Increased drug exposure was associated with a higher incidence of rejection events, whereas CYP3A5 expression correlated with a reduced rate of rejection. These findings underscore the clinical utility of CYP3A5 genotyping for optimizing tacrolimus dosing strategies. Carriers of functional CYP3A5 alleles (*1/*3 or *1/*1) benefit from individualized dose adjustments to achieve therapeutic concentrations and reduce the risk of graft rejection.
Collapse
Affiliation(s)
- Lesly Yanira Xajil-Ramos
- Pharmacogenetics and Pharmacogenomics Research Unit, Faculty of Chemical Sciences and Pharmacy, University of San Carlos de Guatemala, Guatemala, Guatemala
- Pediatric Kidney Disease Research Center at FUNDANIER, Guatemala, Guatemala
- Latin American Network for the Implementation and Validation of Pharmacogenomics Clinical Guidelines (RELIVAF-CYTED), Santiago, Chile
- Doctorate Program in Biomedical Sciences, Faculty of Medical Sciences, University of San Carlos de Guatemala, Guatemala, Guatemala
| | - Jesús Alonso Gándara-Mireles
- Latin American Network for the Implementation and Validation of Pharmacogenomics Clinical Guidelines (RELIVAF-CYTED), Santiago, Chile
- Department of Genomics, Interdisciplinary Research Center for Regional Comprehensive Development Durango Unit, National Polytechnic Institute (IPN), Durango, Mexico
| | - Rodrigo José Vargas Rosales
- Pediatric Kidney Disease Research Center at FUNDANIER, Guatemala, Guatemala
- Latin American Network for the Implementation and Validation of Pharmacogenomics Clinical Guidelines (RELIVAF-CYTED), Santiago, Chile
- Galileo University, Guatemala, Guatemala
| | | | - Andrea Mariela Ruano Toledo
- Pharmacogenetics and Pharmacogenomics Research Unit, Faculty of Chemical Sciences and Pharmacy, University of San Carlos de Guatemala, Guatemala, Guatemala
| | - Amy Kateleen Aldana de la Cruz
- Pharmacogenetics and Pharmacogenomics Research Unit, Faculty of Chemical Sciences and Pharmacy, University of San Carlos de Guatemala, Guatemala, Guatemala
| | - Ismael Lares-Asseff
- Latin American Network for the Implementation and Validation of Pharmacogenomics Clinical Guidelines (RELIVAF-CYTED), Santiago, Chile
- Latin American Society for Pharmacogenomics and Personalized Medicine (SOLFAGEM), Santiago, Chile
| | - Leslie Patrón-Romero
- Faculty of Medicine and Psychology of the Autonomous University of Baja California, Tijuana, Mexico
| | - Horacio Almanza-Reyes
- Faculty of Medicine and Psychology of the Autonomous University of Baja California, Tijuana, Mexico
| | - Randall Lou-Meda
- Pediatric Kidney Disease Research Center at FUNDANIER, Guatemala, Guatemala
| |
Collapse
|
3
|
Lan T, Wu G, Zuo B, Yang J, He P, Zhang Y. Regulation of the immune microenvironment and immunotherapy after liver transplantation. Front Immunol 2025; 16:1602877. [PMID: 40421010 PMCID: PMC12104065 DOI: 10.3389/fimmu.2025.1602877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Liver transplantation (LT) is a primary treatment option for patients with end-stage liver disease. However, post-transplantation immune regulation is critical to graft survival and long-term patient outcomes. Following liver transplantation, the recipient's immune system mounts a response against the graft, while the graft promotes anti-rejection immune reactions and the establishment of immune tolerance. In recent years, advances in the study of the immune microenvironment have provided new insights into post-transplantation immune regulation. Meanwhile, immunotherapy strategies have opened new possibilities for improving transplantation success rates and long-term survival. This review summarizes recent progress in understanding the immune microenvironment and immunotherapy following liver transplantation, focusing on key components of the transplant immune microenvironment, their regulatory networks and mechanisms, major immunosuppressive strategies, emerging immunotherapeutic approaches, and current challenges. The aim was to provide a theoretical foundation for optimizing clinical practice.
Collapse
Affiliation(s)
- Tianyi Lan
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Gang Wu
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bangyou Zuo
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingming Yang
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Southwest Medical University, Luzhou, Sichuan, China
| | - Pan He
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Bernsen EC, Verwiel ETP, van der Lee M, Swen JJ, Santoso M, Brigitha LJ, Admiraal R, Tops BBJ, Huitema ADR, Kemmeren P, Hehir-Kwa JY, Hanff LM, Diekstra MHM. Implementing Pre-Emptive Pharmacogenetics: Impact of Early Pharmacogenetic Screening in a Pediatric Oncology Cohort of 1,151 Subjects. Clin Pharmacol Ther 2025. [PMID: 40331624 DOI: 10.1002/cpt.3685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
In pediatric oncology, pharmacogenetic guidelines are underutilized and the potential impact of pre-emptive pharmacogenetic screening remains largely unexplored despite this field's need for individualized approaches. While comprehensive pharmacogenetic guidelines are not yet available for all anticancer drugs, evidence-based recommendations exist for a subset of supportive care drugs and anticancer drugs, including thiopurines, irinotecan, capecitabine, and 5-fluorouracil. In this study, we evaluate the potential impact of pre-emptive pharmacogenetic screening by retrospectively identifying opportunities for dose or treatment adjustments within a national pediatric oncology cohort. Our analysis focused on ten genes and 28 drugs relevant to pediatric oncology, which are included in the Clinical Pharmacogenetics Implementation Consortium and the Dutch Pharmacogenetics Working Group guidelines. In a cohort of 1,151 pediatric oncology subjects, we identified that 16% of individuals could have benefited from altered drug dosing or treatment. These include dose and treatment recommendations for allopurinol, nonsteroidal anti-inflammatory drugs, phenytoin, amitriptyline, proton pump inhibitors, voriconazole, tramadol, codeine, paroxetine, tacrolimus, rasburicase, and 6-mercaptopurine. As genetic data increasingly becomes available through molecular diagnostics in pediatric oncology, there is a unique opportunity to re-utilize this data for pre-emptive pharmacogenetic screening. Leveraging genetic profiles to guide clinicians in drug selection and dose optimization can improve patient outcomes by enhancing the safety and efficacy of treatments. We therefore recommend incorporating pharmacogenetic screening into clinical workflows to advance personalized medicine in pediatric oncology.
Collapse
Affiliation(s)
- Emma C Bernsen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Maaike van der Lee
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel Santoso
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leiah J Brigitha
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rick Admiraal
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Bastiaan B J Tops
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Patrick Kemmeren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jayne Y Hehir-Kwa
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Lidwien M Hanff
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Meta H M Diekstra
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
5
|
Radwan A, Deininger KM, Ambardekar AV, Anderson HD, Rafaels N, Saba LM, The Colorado Center For Personalized Medicine, Aquilante CL. Prevalence of Actionable Exposures to Pharmacogenetic Medications Among Solid Organ Transplant Recipients in a Population-Scale Biobank. J Pers Med 2025; 15:185. [PMID: 40423057 DOI: 10.3390/jpm15050185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/21/2025] [Accepted: 04/26/2025] [Indexed: 05/28/2025] Open
Abstract
Background/Objectives: Solid organ transplant (SOT) recipients are exposed to multiple medications, many of which have pharmacogenetic (PGx) prescribing recommendations. This study leveraged data from a population-scale biobank and an enterprise data warehouse to determine the prevalence of actionable exposures to PGx medications among kidney, heart, and lung transplant recipients during the first six months post-transplant. Methods: We conducted a retrospective analysis of adult SOT patients with genetic data available from the Colorado Center for Personalized Medicine (CCPM) biobank and clinical data from Health Data Compass (HDC). We evaluated 29 variants in 13 pharmacogenes and 42 Clinical Pharmacogenetics Implementation Consortium (CPIC) level A or B medications (i.e., sufficient evidence to recommend at least one prescribing action based on genetics). The primary outcome was actionable exposure to a PGx medication (i.e., actionable phenotype and a prescription for an affected PGx medication). Results: The study included 358 patients. All patients were prescribed at least one PGx medication, and 49.4% had at least one actionable exposure to a PGx medication during the first six months post-transplant. The frequency of actionable exposure was highest for tacrolimus (15.4%), followed by proton pump inhibitors (PPIs) (15.1%) and statins (12.8%). Statin actionable exposures significantly differed by transplant type, likely due to variations in prescribing patterns and actionable phenotypes for individual statins. Conclusions: Our findings highlight the potential clinical utility of PGx testing among SOT patients. Further studies are needed to address the impact on clinical outcomes and the optimal timing of PGx testing in the SOT population.
Collapse
Affiliation(s)
- Alaa Radwan
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kimberly M Deininger
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Heather D Anderson
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laura M Saba
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA
| | | | - Christina L Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Kongphan O, Lert-itthiporn W, Cha’on U, Anutrakulchai S, Nahok K, Artkaew N, Sriphan C, Jusakul A. Alleles of CYP3A5 and their association with renal function in chronic kidney disease. PeerJ 2025; 13:e19424. [PMID: 40330700 PMCID: PMC12051937 DOI: 10.7717/peerj.19424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Background The cytochrome P450 family 3 subfamily A polypeptide 5 (CYP3A5) gene plays an important role in renal function through its product's involvement in metabolizing endogenous substances and drugs, including immunosuppressants used following kidney transplantation. A single-nucleotide polymorphism, CYP3A5*3 (rs776746), produces a non-functional variant that may influence progression of chronic kidney disease (CKD) by impairing renal filtration. However, the frequency of the CYP3A5*3 allele in the Thai population and its association with renal parameters remain underexplored. This study aimed to determine the prevalence of CYP3A5 polymorphisms and their association with renal function. Methods We investigated the distribution of CYP3A5 polymorphisms in 329 northeastern Thai participants, including 205 CKD patients and 124 healthy controls. Genotyping was performed using the TaqMan allelic discrimination assay. Renal function parameters were assessed and compared between CYP3A5*1 and CYP3A5*3 allele carriers. Results In the entire cohort, the allele frequency of CYP3A5*3 was 63.2%, with genotype frequencies of CYP3A5*1/*1 (16.7%), CYP3A5*1/*3 (40.1%), and CYP3A5*3/*3 (43.2%). There was no significant difference in the CYP3A5 allele frequencies between CKD and control groups. CYP3A5*3 carriers exhibited significantly lower eGFR, urine creatinine and serum creatinine clearance and higher UACR compared to CYP3A5*1 carriers. After adjusting for confounders, CYP3A5*3 remained significantly associated with reduced urine creatinine. Conclusion This study highlights a high prevalence of CYP3A5 polymorphisms in the northeastern Thai population. The association of the CYP3A5*3 allele with renal function parameters underscores the need for further research into the mechanisms by which CYP3A5 affects kidney function, which could inform personalized CKD management strategies.
Collapse
Affiliation(s)
- Onnapa Kongphan
- Department of Biomedical Science, Faculty of Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Worachart Lert-itthiporn
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ubon Cha’on
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sirirat Anutrakulchai
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanokwan Nahok
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nadthanicha Artkaew
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chanpen Sriphan
- Srinagarind Hospital, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Chronic Kidney Disease Prevention in the Northeast of Thailand (CKDNET) Project, Khon Kaen University, Khon Kaen, Thailand
- The Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
7
|
Duan B, Gao J, Ge B, Wu S, Yu J. Development and Validation of a Nomogram for Predicting Subtherapeutic Tacrolimus Blood Levels in Renal Transplant Recipients: A Multivariate Logistic Regression Analysis. Transplant Proc 2025; 57:529-537. [PMID: 40082170 DOI: 10.1016/j.transproceed.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
This study constructs a nomogram risk prediction model to identify factors affecting subtherapeutic tacrolimus (FK506) blood concentrations in postrenal transplant patients, enhancing clinical management. Data from renal transplant patients treated with tacrolimus from January to December 2023 were analyzed using multivariate logistic regression to identify risk factors. A nomogram model was constructed and validated through cross-validation and bootstrapping. Predictive performance was assessed via receiver operating characteristic curve and Hosmer- Lemeshow test. Among 340 patients, 224 achieved target FK506 concentrations (5-15 ng/mL). Independent risk factors for subtherapeutic levels included white blood cell count ≤4 × 10^9/L, total bilirubin >20 μmol/L, creatinine >73 μmol/L, and blood urea nitrogen ≤7.1 mmol/L. The model's receiver operating characteristic area under the curve was 0.84, with a Hosmer- Lemeshow test P-value of .386, indicating high predictive accuracy and good calibration. The nomogram effectively predicts subtherapeutic FK506 levels, providing a valuable tool for personalized patient management. Future research should refine and externally validate the model.
Collapse
Affiliation(s)
- Bowen Duan
- Department of Pharmacy, Gansu Provincial Hospital, Lan Zhou, China
| | - Jinxian Gao
- Department of Pharmacy, Gansu Provincial Hospital, Lan Zhou, China
| | - Bin Ge
- Department of Pharmacy, Gansu Provincial Hospital, Lan Zhou, China
| | - Shujin Wu
- Department of Pharmacy, Gansu Provincial Hospital, Lan Zhou, China
| | - Jing Yu
- Department of Pharmacy, Gansu Provincial Hospital, Lan Zhou, China.
| |
Collapse
|
8
|
Gloor YS, Mouterde M, Terrier J, Lenoir C, Gosselin P, Rollason V, Reny JL, Boukouvala S, Al-Yahyaee S, Yimer G, Černý V, Poloni ES, Samer CF, Daali Y. Cytochrome P450 phenotyping using the Geneva cocktail improves metabolic capacity prediction in a hospitalized patient population. Br J Clin Pharmacol 2025; 91:1382-1395. [PMID: 39701086 DOI: 10.1111/bcp.16368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/29/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
AIMS Liver cytochromes (CYPs) play an important role in drug metabolism but display a large interindividual variability resulting both from genetic and environmental factors. Most drug dose adjustment guidelines are based on genetics performed in healthy volunteers. However, hospitalized patients are not only more likely to be the target of new prescriptions and drug treatment modifications than healthy volunteers, but will also be more subject to polypharmacy, drug-drug interactions, or to suffer from disease or inflammation affecting CYP activities. METHODS We compared predicted phenotype based on genetic data and measured phenotype using the Geneva cocktail to determine the extent of drug metabolizing enzyme variability in a large population of hospitalized patients (>500) and healthy young volunteers (>300). We aimed to assess the correlation between predicted and measured phenotype in the two populations. RESULTS We found that, even in cases where the genetically predicted metabolizer group correlates well with measured CYP activity at group level, this prediction lacks accuracy for the determination of individual metabolizer capacities. Drugs can have a profound impact on CYP activity, but even after combining genetic and drug treatment information, the activity of a significant proportion of extreme metabolizers could not be explained. CONCLUSIONS Our results support the use of measured metabolic ratios in addition to genotyping for accurate determination of individual metabolic capacities to guide personalized drug prescription.
Collapse
Affiliation(s)
- Yvonne S Gloor
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Médéric Mouterde
- Department of Genetics and Evolution, Laboratory of Anthropology, Genetics and Peopling history, University of Geneva, Geneva, Switzerland
| | - Jean Terrier
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Camille Lenoir
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Pauline Gosselin
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Victoria Rollason
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Jean-Luc Reny
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sotiria Boukouvala
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Said Al-Yahyaee
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Getnet Yimer
- Center for Global Genomics & Health Equity, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Viktor Černý
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Czech Republic
| | - Estella S Poloni
- Department of Genetics and Evolution, Laboratory of Anthropology, Genetics and Peopling history, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Caroline F Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency, Geneva University Hospitals (HUG), Geneva, Switzerland
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Clinical Pharmacology and Toxicology, Department of Anaesthetics Pharmacology and Intensive Care, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Geneva, Switzerland
| |
Collapse
|
9
|
Moore C, Halman A, Stenta T, Khatri D, Williams E, Dyas R, Stolper J, Elliott DA, Conyers R. Frequency and Implications of High-Risk Pharmacogenomic Phenotypes Identified in a Diverse Australian Pediatric Oncology Cohort. Clin Transl Sci 2025; 18:e70246. [PMID: 40347484 PMCID: PMC12065477 DOI: 10.1111/cts.70246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
Pharmacogenomics remains underutilized in pediatric oncology, despite the existence of evidence-based guidelines. Implementation of pharmacogenomics-informed prescribing could improve medication safety and efficacy in pediatric oncology patients, who are at high risk of adverse drug reactions. This study examines the prevalence of high-risk pharmacogenomic phenotypes and the prescription of relevant medications in a diverse Australian pediatric oncology cohort, highlighting the potential impact of pharmacogenomic testing in this unique population. Whole genome sequencing data from 180 patients were analyzed to assess 14 genes with evidence-based pharmacogenomic guidelines relevant to pediatric oncology. Over 90% of patients had at least one high-risk phenotype, with 20% presenting four or more. Ondansetron, mercaptopurine, omeprazole, pantoprazole, and voriconazole were commonly prescribed medications that have pharmacogenomic prescribing recommendations, with the latter three showing the highest actionability rates. High-risk phenotypes were most frequently observed for CYP2C19 and CYP2D6, with 30% of patients having a high-risk phenotype for both genes. This study underscores the potential utility of pharmacogenomics in pediatric oncology patients across a range of pharmacogenes and commonly prescribed medications. The findings support advocacy for implementing broad, pre-emptive pharmacogenomic testing in oncology patients to improve treatment safety and efficacy.
Collapse
Affiliation(s)
- Claire Moore
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
| | - Andreas Halman
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Victorian Clinical Genetics ServicesMurdoch Children's Research InstituteMelbourneVictoriaAustralia
| | - Tayla Stenta
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
| | - Dhrita Khatri
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
| | - Elizabeth Williams
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
| | - Roxanne Dyas
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
| | - Julian Stolper
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Centre for Stem Cell Medicine, ReNEW, Melbourne NodeParkvilleVictoriaAustralia
| | - David A. Elliott
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Centre for Stem Cell Medicine, ReNEW, Melbourne NodeParkvilleVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | - Rachel Conyers
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneParkvilleVictoriaAustralia
- Children's Cancer CentreThe Royal Children's HospitalParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Centre for Stem Cell Medicine, ReNEW, Melbourne NodeParkvilleVictoriaAustralia
| |
Collapse
|
10
|
Wang J, Nithianantham S, Chai SC, Jung YH, Yang L, Ong HW, Li Y, Zhang Y, Miller DJ, Chen T. Decoding the selective chemical modulation of CYP3A4. Nat Commun 2025; 16:3423. [PMID: 40210880 PMCID: PMC11985932 DOI: 10.1038/s41467-025-58749-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Drug-drug interactions associate with concurrent uses of multiple medications. Cytochrome P450 (CYP) 3A4 metabolizes a large portion of marketed drugs. To maintain the efficacy of drugs metabolized by CYP3A4, pan-CYP3A inhibitors such as ritonavir are often co-administered. Although selective CYP3A4 inhibitors have greater therapeutic benefits as they avoid inhibiting unintended CYPs and undesirable clinical consequences, the high homology between CYP3A4 and CYP3A5 has hampered the development of such selective inhibitors. Here, we report a series of selective CYP3A4 inhibitors with scaffolds identified by high-throughput screening. Structural, functional, and computational analyses reveal that the differential C-terminal loop conformations and two distinct ligand binding surfaces disfavor the binding of selective CYP3A4 inhibitors to CYP3A5. Structure-guided design of compounds validates the model and yields analogs that are selective for CYP3A4 versus other major CYPs. These findings demonstrate the feasibility to selectively inhibit CYP3A4 and provide guidance for designing better CYP3A4 selective inhibitors.
Collapse
Affiliation(s)
- Jingheng Wang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Nithianantham
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Young-Hwan Jung
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Han Wee Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yifan Zhang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
11
|
Lubomirov R, Pérez‐Ramos L, Fudio S, Asín‐Prieto E, Ibarra‐Gómez L, Zubiaur P. CYP3A Genotype Is Associated With Variability in the Exposure and Clearance of the Novel Oncogenic Transcription Inhibitor Lurbinectedin. Clin Transl Sci 2025; 18:e70173. [PMID: 40146606 PMCID: PMC11949125 DOI: 10.1111/cts.70173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
Lurbinectedin is an oncogenic transcription inhibitor indicated for the treatment of small cell lung cancer (SCLC), which has also shown activity against other malignancies. In this work, two independent cohorts of 180 (discovery cohort) and 719 (validation cohort) cancer patients receiving lurbinectedin in Phases I, II, or III clinical trials were enrolled. Using a population pharmacokinetic (popPK) model of the discovery cohort, patients with extremely high (n = 10, cohort 1) and low (n = 10, cohort 2) etaCL values (i.e., a variable used as a surrogate of unexplained CL interindividual variability) were identified. They were sequenced for 42 candidate genes involved in lurbinectedin pharmacokinetics. A total of 34 variants located in 20 genes were significantly associated with lurbinectedin etaCL; the best nine hits (located in CYP3A5, CYP3A4, ABCB1, ARNT, NR5A2, NR1H4, and FOXA3) were subsequently genotyped in the validation cohort. A strong additive association between CYP3A4 and CYP3A5 genotypes (informed as a CYP3A activity score [AS] variable) and lurbinectedin clearance (CL) and exposure was confirmed, for example, patients with an AS of 3, 2, or 1 showed a 2.3-, 1.6-, and 1.5-fold higher total lurbinectedin CL compared to those with an AS of 0 and 2.3-, 1.8-, and 1.6-fold higher unbound lurbinectedin CL. In conclusion, preemptive CYP3A genotyping may offer a valuable approach for personalizing treatment with lurbinectedin in cancer patients.
Collapse
Affiliation(s)
- Rubin Lubomirov
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| | - Laura Pérez‐Ramos
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| | - Salvador Fudio
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| | - Eduardo Asín‐Prieto
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| | - Laura Ibarra‐Gómez
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| | - Pablo Zubiaur
- PharmaMar S.A., Clinical Pharmacology Department, Clinical DevelopmentColmenar ViejoMadridSpain
| |
Collapse
|
12
|
Raval CU, Makwana A, Patel S, Hemani R, Pandey SN. Optimizing tacrolimus dosage in post-renal transplantation using DoseOptimal framework: profiling CYP3A5 genetic variants for interpretability. Int J Clin Pharm 2025:10.1007/s11096-025-01899-y. [PMID: 40117041 DOI: 10.1007/s11096-025-01899-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Achieving optimal tacrolimus dosing is vital for effectively balancing therapeutic efficacy and safety, as CYP3A5 genetic variants and inter-patient variability emphasize the need for precision strategies. AIM This study aimed to optimize tacrolimus dosage prediction for renal transplant recipients by incorporating genetic polymorphisms, specifically profiling CYP3A5 genetic variants, within the DoseOptimal framework to enhance interpretability and accuracy of dosing decisions. METHOD The dataset comprised clinical, demographic, and CYP3A5 genetic variants information from 1045 stable tacrolimus-treated patients. The DoseOptimal framework was developed by integrating the strengths of the most effective algorithms from fifteen machine learning models. SHapley Additive exPlanations (SHAP) and decision tree insights were incorporated to enhance the framework's interpretability. The framework's performance was assessed using mean absolute error (MAE) and the coefficient of determination (R2 score). The F-statistic and p value were calculated to validate the framework's statistical significance. RESULTS The DoseOptimal framework demonstrated robust performance with an R2 score of 0.884 in the training set and 0.830 in the testing set. The MAE was 0.40 mg/day (95% CI 0.38-0.43) in the training set and 0.41 mg/day (95% CI 0.38-0.45) in the testing set. The framework predicted the ideal tacrolimus dosage in 87.6% (n = 275) of the test cohort, with 3.2% (n = 10) underestimation and 9.2% (n = 29) overestimation. The framework's statistical significance was confirmed with an F-statistic of 266.095 and a p value < 0.001. CONCLUSION The framework provides precision medicine-based dosing solutions tailored to individual genetic profiles, minimizing dosing errors and enhancing patient outcomes.
Collapse
Affiliation(s)
- Chintal Upendra Raval
- U & P U Patel Department of Computer Engineering, Chandubhai S. Patel Institute of Technology, Charotar University of Science and Technology, CHARUSAT - Campus, Changa, Anand, Gujarat, 388421, India
| | - Ashwin Makwana
- U & P U Patel Department of Computer Engineering, Chandubhai S. Patel Institute of Technology, Charotar University of Science and Technology, CHARUSAT - Campus, Changa, Anand, Gujarat, 388421, India
| | - Samir Patel
- Department of Pharmaceutical Chemistry and Analysis, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT - Campus, Changa, Anand, Gujarat, 388421, India
| | - Rashmi Hemani
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT - Campus, Changa, Anand, Gujarat, 388421, India
| | - Sachchida Nand Pandey
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, Gujarat, 387001, India.
| |
Collapse
|
13
|
Tsironi A, Lazaros K, Mendrinou E, Papasotiriou M, Siamoglou S, Kydonopoulou K, John A, Gerou A, Gerou S, Ali BR, Vrahatis AG, Patrinos GP. Impact of CYP3A4 and ABCB1 genetic variants on tacrolimus dosing in Greek kidney transplant recipients. Front Pharmacol 2025; 16:1538432. [PMID: 40176889 PMCID: PMC11962430 DOI: 10.3389/fphar.2025.1538432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Background Tacrolimus, an approved first-line calcineurin inhibitor, is widely prescribed in organ transplantation to prevent allograft rejection. Its narrow therapeutic index requires precise management to achieve optimal dosing and to minimize adverse drug events (ADEs) while ensuring its therapeutic efficacy. Among several factors, genetic differences contribute significantly to the inter-individual and inter-ethnic variability in pharmacokinetics (PK) of tacrolimus in kidney transplant recipients. As a result, investigating the role of genetic variation in Greek transplant recipients becomes crucial to optimizing therapeutic strategies and enhancing the efficacy of immunosuppressive treatment. Hypothesis Genetic variants which are known to influence the activity of enzymes or drug-transporters critical to tacrolimus pharmacokinetics, may significantly affect the required kidney post-transplant tacrolimus daily dose. Aim To assess the correlation of ABCB1 genetic variants (rs1128503, rs2229109) and CYP3A4 (rs2242480, rs4986910) with tacrolimus dose-adjusted trough concentration (C0/D), in Greek kidney transplant recipients. Methods Ninety-four unrelated Greek kidney transplant recipients were included in this study from the Department of Nephrology and Kidney Transplantation of the University General Hospital of Patras. Patients' dose-adjusted trough levels were measured at five distinct time points after transplantation and analyzed in relation to the possible influence of CYP3A4 and correlated with the abovementioned ABCB1 genetic variants using standard genotyping analysis and Sanger sequencing. Results The genetic variants rs1128503, rs2229109, rs2242480, rs4986910 did not show any significant association with the daily dosing requirements of tacrolimus for at least 1 year, in Greek patients who have undergone kidney transplant. Conclusion It remains uncertain whether these genetic variants influence the assessment of the appropriate tacrolimus dosing 1 year after transplantation in Greek kidney transplant recipients.
Collapse
Affiliation(s)
- Anna Tsironi
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | | | - Effrosyni Mendrinou
- Molecular Diagnostics Laboratory, INRASTES, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - Marios Papasotiriou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, Greece
| | - Stavroula Siamoglou
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | | | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | - Spyridon Gerou
- ANALYSI Biomedical Laboratories S.A., Thessaloniki, Greece
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Research Institute, Al-Ain, United Arab Emirates
| | | | - George P. Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Research Institute, Al-Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Clinical Bioinformatics Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
14
|
Phommavong V, Burger C, Shamseddin MK, Mansell H. Characterizing the Use of Prolonged-Release Once-Daily Tacrolimus (LCPT) across Canada. Can J Hosp Pharm 2025; 78:e3624. [PMID: 40078496 PMCID: PMC11879362 DOI: 10.4212/cjhp.3624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/05/2024] [Indexed: 03/14/2025]
Abstract
Background Tacrolimus is the most common calcineurin inhibitor given to kidney and liver transplant recipients. Prolonged-release once-daily tacrolimus (LCPT) is the newest formulation of this drug, but prescribing practices for tacrolimus across Canada are unknown. Objectives To investigate the use of tacrolimus across Canada, by determining coverage for the drug, exploring prescribing practices and factors related to decision-making, and identifying management methods for patients with rapid metabolism of tacrolimus. Methods A mixed-methods, descriptive study using survey-based data collection and qualitative interviews was undertaken. The medical director and a pharmacist from each adult kidney and liver transplant centre in Canada were invited to complete an electronic questionnaire consisting of 8 open-ended questions concerning their respective transplant programs' coverage for and use of tacrolimus. Interested participants completed a one-on-one virtual follow-up interview to explore experiences. Results A total of 28 health care providers participated in the survey, of whom 18 completed an interview, achieving representation from 15 (79%) of 19 kidney transplant programs and 3 (38%) of 8 liver transplant programs. Prescribing habits varied, with immediate-release tacrolimus (IR-Tac) being the most commonly preferred formulation (due to provider experience), followed by extended-release tacrolimus (ER-Tac) and LCPT. Most survey respondents (26/28) indicated that their centres used LCPT for maintenance but not de novo immunosuppression. The most common reason for conversion to LCPT was to reduce tremors or to address suspected rapid metabolism; barriers to uptake of LCPT included perceived disadvantages related to cost and coverage. Conclusions Prescribing practices for tacrolimus varied across Canada. IR-Tac was the most commonly used formulation, followed by ER-Tac. LCPT was used primarily in the maintenance phase for people with neurotoxicity or rapid metabolism, but there was a lack of consistency in how rapid metabolism was defined.
Collapse
Affiliation(s)
- Vanessa Phommavong
- , BSc, is a student in the College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan
| | - Cathy Burger
- , BScPhm, ACPR, is with the Department of Pharmacy, St Joseph's Healthcare Hamilton, Hamilton, Ontario
| | - M Khaled Shamseddin
- , MD, is with the Division of Nephrology, Department of Medicine, Queen's University, Kingston, Ontario
| | - Holly Mansell
- , BSP, PharmD, PhD, is with the College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan
| |
Collapse
|
15
|
Pérez-Duval E, Calderón B, Izquierdo M, Herrera-Isidrón JA, Reyes-Reyes E, Herrera A, Soto M, Beltré A, Rodeiro-Guerra I. Allele and genotype frequencies of variants in P450 cytochromes, transports, and DNA repair enzymes in the Dominican Republic population. Front Pharmacol 2025; 15:1494482. [PMID: 40109368 PMCID: PMC11919896 DOI: 10.3389/fphar.2024.1494482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/23/2024] [Indexed: 03/22/2025] Open
Abstract
Introduction Single-nucleotide variants (SNVs) give rise to important inter-individual and inter-ethnic variabilities in the metabolism and disposition of several therapeutic agents and may cause differences in the treatment response to clinically important drugs like antiarrhythmics, antidepressants, antihistamines, and antipsychotics, among others. Information about the prevalence of variants in the Dominican Republic population is still limited. The aim of this study was to describe the frequency distribution of 32 SNVs from 14 genes with pharmacogenetic interest within a sample of 150 unrelated healthy individuals. Methods Genotype and allele frequencies were determined, and pairwise Wright's FST statistic was evaluated. Results Hardy-Weinberg equilibrium deviations were found in seven loci from CYP2D6 (rs16947, rs3892097, rs1058164, rs1135840, and rs28371725) and CYP2C19 (rs12769205 and rs4244285) genes. The minor allele frequencies ranged from 0.01 to 0.50 values in the xenobiotic biotransformation enzymes and transporter genes. The average admixture estimates were 51.6%, 39.5%, and 8.9% for European, African, and Amerindian ancestries, respectively. Pairwise FST analysis revealed that Dominicans displayed genetic similarity to Latin American populations, especially those with Afro-Caribbean ancestry, given the selected variants. Higher differences were identified from East and South Asians, Europeans, and Africans, in which several values above the FST threshold for moderate differentiation were identified within variants in CYP2C, CYP3A, CYP1A1, ABCB1, SLC45A2, XRCC1, and XRCC3 genes. Conclusions These results should allow establishing the clinical relevance of pharmacogenetic testing in variant alleles related to drug transport and metabolism genes in this population.
Collapse
Affiliation(s)
- Elizabeth Pérez-Duval
- School of Medicine, Instituto Tecnológico de Santo Domingo (INTEC), Santo Domingo, Dominican Republic
| | - Berniza Calderón
- School of Medicine, Instituto Tecnológico de Santo Domingo (INTEC), Santo Domingo, Dominican Republic
- Research Unit, Centro Médico de Diabetes, Obesidad y Especialidades (CEMDOE), Santo Domingo, Dominican Republic
- Research Committee, Sociedad Dominicana de Endocrinología y Nutrición (SODENN), Santo Domingo, Dominican Republic
| | - Marlen Izquierdo
- School of Medicine, Instituto Tecnológico de Santo Domingo (INTEC), Santo Domingo, Dominican Republic
| | | | | | - Alejandro Herrera
- Cuban National Center of Biodiversity, Institute of Ecology and Sistematic of Cuba, Havana, Cuba
| | - Manuel Soto
- Research Unit, Centro Médico de Diabetes, Obesidad y Especialidades (CEMDOE), Santo Domingo, Dominican Republic
| | - Alba Beltré
- School of Medicine, Instituto Tecnológico de Santo Domingo (INTEC), Santo Domingo, Dominican Republic
- Research Unit, Centro Médico de Diabetes, Obesidad y Especialidades (CEMDOE), Santo Domingo, Dominican Republic
| | | |
Collapse
|
16
|
Bayanova M, Bolatov A, Malik D, Zhenissova A, Abdikadirova A, Sapargaliyeva M, Nazarova L, Myrzakhmetova G, Novikova S, Turganbekova A, Pya Y. Whole-Exome Sequencing Followed by dPCR-Based Personalized Genetic Approach in Solid Organ Transplantation: A Study Protocol and Preliminary Results. Methods Protoc 2025; 8:27. [PMID: 40126245 PMCID: PMC11932258 DOI: 10.3390/mps8020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Genetic profiling and molecular biology methods have made it possible to study the etiology of the end-stage organ disease that led to transplantation, the genetic factors of compatibility and tolerance of the transplant, and the pharmacogenetics of immunosuppressive drugs and allowed for the development of monitoring methods for the early assessment of allograft rejection. This study aims to report the design and baseline characteristics of an integrated personalized genetic approach in solid organ transplantation, including whole-exome sequencing (WES) and the monitoring of dd-cfDNA by dPCR. Preliminary results reported female recipients with male donors undergoing two pediatric and five adult kidney and three heart transplantations. WES revealed a pathogenic mutation in RBM20 and VUS in TTN and PKP2 in heart recipients, while kidney donors presented mutations in UMOD and APOL1 associated with autosomal-dominant kidney diseases, highlighting the risks requiring the long-term monitoring of recipients, donors, and their family members. %dd-cfDNA levels were generally stable but elevated in cadaveric kidney recipient and one pediatric patient with infectious complications and genetic variants in the ABCB1 and ABCC2 genes. These findings highlight the potential of combining genetic and molecular biomarker-based approaches to improve donor-recipient matching, predict complications, and personalize post-transplant care, paving the way for precision medicine in transplantation.
Collapse
Affiliation(s)
- Mirgul Bayanova
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Aidos Bolatov
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
- School of Medicine, Shenzhen University, Shenzhen 518060, China
- School of Medicine, Astana Medical University, Astana 010000, Kazakhstan
| | - Dias Malik
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Aida Zhenissova
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Aizhan Abdikadirova
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Malika Sapargaliyeva
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Lyazzat Nazarova
- Genetic Unit, Department of Laboratory Medicine, Pathology and Genetics, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (M.B.); (D.M.); (A.Z.); (A.A.); (M.S.); (L.N.)
| | - Gulzhan Myrzakhmetova
- Clinical Academic Department of Cardiology, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan;
| | - Svetlana Novikova
- Clinical Academic Department of Cardiac Surgery, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (S.N.); (Y.P.)
| | - Aida Turganbekova
- HLA-Laboratory, Scientific-Production Center of Transfusiology, Astana 010000, Kazakhstan;
| | - Yuriy Pya
- Clinical Academic Department of Cardiac Surgery, “University Medical Center” Corporate Fund, Astana 010000, Kazakhstan; (S.N.); (Y.P.)
| |
Collapse
|
17
|
Adamczyk K, Zuzda K, Jankowski M, Świerczyński R, Chudziński K, Czapski B, Szułdrzyński K. Effects of Opioids in Cancer Pain: An Interplay Among Genetic Factors, Immune Response, and Clinical Outcomes-A Scoping Review. Cancers (Basel) 2025; 17:863. [PMID: 40075716 PMCID: PMC11899605 DOI: 10.3390/cancers17050863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/10/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Managing cancer-related pain presents complex challenges involving the interplay between analgesic efficacy, immune system responses, and patient outcomes. Methods: Following the Scale for the Assessment of Narrative Review Articles (SANRA) criteria, we conducted a comprehensive literature search in Medline, Scopus, and Web of Science databases. The review synthesized evidence regarding opioid pain management modalities, genetic variations affecting pain perception, and associated drug metabolism. Results: The literature reveals significant associations between opioid administration and immune function, with potential implications for cancer progression and survival. Genetic polymorphisms in key genes influence individual responses to pain opioid metabolism and, finally, pain management strategies. The immunosuppressive effects of opioids emerge as a critical consideration in cancer pain management, potentially influencing disease progression and treatment outcomes. Conclusions: Genetic variants influence analgesic efficacy, while the interaction between opioid-induced immunosuppression and genetic factors impacts both pain control and survival outcomes. This emphasizes the need for personalized treatment approaches considering individual genetic profiles and immune function.
Collapse
Affiliation(s)
- Kamil Adamczyk
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Konrad Zuzda
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Miłosz Jankowski
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Rafał Świerczyński
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Kamil Chudziński
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Bartosz Czapski
- Department of Neurosurgery, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Konstanty Szułdrzyński
- Department of Anesthesiology and Intensive Care, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| |
Collapse
|
18
|
Knezevic CE, Stevenson JM, Merran J, Snyder I, Restorick G, Waters C, Marzinke MA. Implementation of Integrated Clinical Pharmacogenomics Testing at an Academic Medical Center. J Appl Lab Med 2025; 10:259-273. [PMID: 39657156 DOI: 10.1093/jalm/jfae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/04/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Pharmacogenomics has demonstrated benefits for clinical care, including a reduction in adverse events and cost savings. However, barriers in expanded implementation of pharmacogenomics testing include prolonged turnaround times and integration of results into the electronic health record with clinical decision support. A clinical workflow was developed and implemented to facilitate in-house result generation and incorporation into the electronic health record at a large academic medical center. METHODS An 11-gene actionable pharmacogenomics panel was developed and validated using a QuantStudio 12K Flex platform. Allelic results were exported to a custom driver and rules engine, and result messages, which included a diplotype and predicted metabolic phenotype, were sent to the electronic health record; an electronic consultation (eConsult) service was integrated into the workflow. Postimplementation monitoring was performed to evaluate the frequency of actionable results and turnaround times. RESULTS The actionable pharmacogenomics panel covered 39 alleles across 11 genes. Metabolic phenotypes were resulted alongside gene diplotypes, and clinician-facing phenotype summaries (Genomic Indicators) were presented in the electronic health record. Postimplementation, 8 clinical areas have utilized pharmacogenomics testing, with 56% of orders occurring in the outpatient setting; 22.1% of requests included at least one actionable pharmacogene, and 67% of orders were associated with a pre- or postresult electronic consultation. Mean turnaround time from sample collection to result was 4.6 days. CONCLUSIONS A pharmacogenomics pipeline was successfully operationalized at a quaternary academic medical center, with direct integration of results into the electronic health record, clinical decision support, and eConsult services.
Collapse
Affiliation(s)
- Claire E Knezevic
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - James M Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan Merran
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Isabel Snyder
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | | | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
McDermott JH, Burke K, Fullerton N, O’Sullivan J, Alex A, Ingham A, Sharma V, Godfrey N, Odudu A, Syed T, Stevens A, Beynon R, Greaves N, Akam D, Mirza S, Wilson P, Wright S, Payne K, Newman WG. Pre-emptive pharmacogenetic testing in the acute hospital setting: a cross-sectional study. QJM 2025; 118:154-160. [PMID: 39418209 PMCID: PMC12051386 DOI: 10.1093/qjmed/hcae200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Pharmacogenetic-guided prescribing can be used to improve the safety and effectiveness of medicines. There are several approaches by which this intervention might be implemented in clinical practice, which will vary depending on the health system and clinical context. AIM To understand the clinical utility of panel-based pharmacogenetic testing in patients admitted acutely to hospital and to establish variables that predict if an individual might benefit from the intervention. DESIGN A cross-sectional study recruiting patients admitted acutely to hospital. METHODS Participants underwent panel-based pharmacogenetic testing, and their genetic results were analysed in their context of the medicines they had been exposed to as an inpatient. The primary outcome was the proportion of patients with clinically actionable gene-drug interactions. Individual variables that predict the clinical utility of pharmacogenetic testing were established via logistic regression. RESULTS Genetic and prescribing data were available for 482 inpatients (55% male; median age 61.2 years; range: 18-96), 97.9% of whom carried a pharmacogenetic result of interest. During their admission, 79.5% of patients were exposed to a medicine for which there is pharmacogenetic prescribing guidance available. Just under one in seven individuals (13.7%) had a clinically actionable gene-drug interaction. Increasing age (>50 years) was positively correlated with the likelihood (2.7-fold increased risk) of having a clinically actionable interaction. CONCLUSIONS These findings demonstrate the potential scale, and potential clinical utility, of pharmacogenetic testing as an intervention, highlighting the need to develop infrastructure to support healthcare professionals make use of this emerging tool.
Collapse
Affiliation(s)
- John H McDermott
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Kerry Burke
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Vascular Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Neil Fullerton
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - James O’Sullivan
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Aleina Alex
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Amy Ingham
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Videha Sharma
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Nicola Godfrey
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Aghogho Odudu
- Acute Medical Unit, Manchester Royal Infirmary, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Tania Syed
- Acute Medical Unit, Manchester Royal Infirmary, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Andrew Stevens
- Acute Medical Unit, Manchester Royal Infirmary, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Rhys Beynon
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicholas Greaves
- Manchester Vascular Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Daniel Akam
- Acute Medical Unit, Manchester Royal Infirmary, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Selman Mirza
- Biostatistics Collaboration Unit, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Paul Wilson
- Centre for Primary Care and Health Services Research, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Stuart Wright
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Katherine Payne
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, The University of Manchester, Manchester, UK
| | - William G Newman
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Rim JH, Kim YG, Kim S, Choi R, Lee JS, Park S, Lee W, Song EY, Lee SY, Chun S. Clinical Pharmacogenetic Testing and Application: 2024 Updated Guidelines by the Korean Society for Laboratory Medicine. Ann Lab Med 2025; 45:121-132. [PMID: 39681357 PMCID: PMC11788703 DOI: 10.3343/alm.2024.0572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 10/25/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
In the era of precision medicine, pharmacogenetics has substantial potential for addressing inter-individual variability in drug responses. Although pharmacogenetics has been a research focus for many years, resulting in the establishment of several formal guidelines, its clinical implementation remains limited to several gene-drug combinations in most countries, including Korea. The main causes of delayed implementation are technical challenges in genotyping and knowledge gaps among healthcare providers; therefore, clinical laboratories play a critical role in the timely implementation of pharmacogenetics. This paper presents an update of the Clinical Pharmacogenetic Testing and Application guidelines issued by the Korean Society for Laboratory Medicine and aims to provide the necessary information for clinical laboratories planning to implement or expand their pharmacogenetic testing. Current knowledge regarding nomenclature, gene-drug relationships, genotyping technologies, testing strategies, methods for clinically relevant information delivery, QC, and reimbursements has been curated and described in this guideline.
Collapse
Affiliation(s)
- John Hoon Rim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young-gon Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sollip Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Rihwa Choi
- Department of Laboratory Medicine, GC Labs, Yongin, Korea
- Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jee-Soo Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seungman Park
- Department of Laboratory Medicine, National Cancer Center, Goyang, Korea
| | - Woochang Lee
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Soo-Youn Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sail Chun
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Jackson RL, Heyrend C, Bucher B, Brewer A, Peterson C, May LJ, Bonkowsky JL. Impact of Pharmacogenomic Testing in Pediatric Heart and Kidney Transplant. Pediatr Transplant 2025; 29:e70044. [PMID: 39924350 DOI: 10.1111/petr.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Pediatric solid organ transplantation is a complex process including a tightly orchestrated medication regimen, essential for prevention of infection, rejection, graft failure, and mortality. Pharmacogenomic (PGx) testing tailors medication therapy to the individual patient, focusing on safety, efficacy, and avoidance of adverse effects. Implementation of PGx panel results into clinical practice for pediatric transplant patients has not been evaluated. METHODS Pediatric patients evaluated for heart, kidney, or combined heart-kidney transplant at a tertiary children's hospital from October 2021 to October 2023 received PGx panel testing. PRIMARY OUTCOME MEASURE Report the prevalence of actionable PGx variants for key genes impacting pharmacotherapy in pre- and post-heart and kidney transplant populations. RESULTS A total of 73 patients were included, predominately white (84.9%) and male (64.4%), with a mean age of 8.8 ± 6.4 years. Indications for PGx testing included evaluation for heart transplant (38.4%), kidney transplant (38.4%), combined heart-kidney transplant (4.1%), or to inform posttransplant care (19.2%). All patients had at least one actionable phenotype identified. 37 of 73 patients (50.7%) had at least one actionable phenotype for the transplant-specific genes captured including CYP3A5, SLCO1B1, G6PD, TPMT, prothrombin (Factor 2), and Factor V Leiden. 16 of 73 patients (21.9%) had actionable CYP3A5 phenotypes. 15 of 73 (20.5%) had actionable SLCO1B1 phenotypes. 9 of 73 patients (12.3%) had actionable TPMT phenotypes. 5 of 73 (6.8%) had Prothrombin or Factor V Leiden variants. CONCLUSIONS Routine pretransplant PGx testing provided information that was actionable and could be utilized to optimize posttransplant medications for all patients.
Collapse
Affiliation(s)
- Rachel L Jackson
- Department of Pharmacy, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Caroline Heyrend
- Department of Pharmacy, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Bridget Bucher
- Department of Pharmacy, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Ashlie Brewer
- Department of Pharmacy, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Caitlin Peterson
- Division of Nephrology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Lindsay J May
- Division of Cardiology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
- Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, Utah, USA
| |
Collapse
|
22
|
Zhou X, Tang R, Zhang D, He X, Liu Z, Gao Y, Liu H. Effect of CYP 3A4*1B and CYP3A5*3 Gene Polymorphisms in Antirejection of Tacrolimus in Liver Transplant Patients. Transplant Proc 2025; 57:298-304. [PMID: 39643490 DOI: 10.1016/j.transproceed.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/16/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Tacrolimus is a substrate of CYP 3A5; to reduce the rate of liver injury and rejection in liver transplant (LT) recipients, it is feasible to optimize the administration of tacrolimus by adding CYP gene polymorphism. METHODS We divided 151 LT recipients randomly into an optimization group and a control group. All were tested routinely for clinical indicators such as FK506 trough concentration and biochemistry, and their complications and survival were observed. The optimization group additionally detected single nucleotide polymorphisms in the CYP 3A4*1B and CYP 3A5*3 genes. RESULTS There were no significant differences in tacrolimus dosage, FK506 trough concentrations, and concentration/dose value between the 2 groups. In the optimization group, all patients tested CYP 3A4*1B as wild type. CYP 3A5*3 detection classification included 35 with the G/G mutation (45.5%) and 36 A/G wild-type individuals (46.8%). The concentration/dose values of G/G mutant patients were significantly higher than those of A/G wild-type and A/A mutant patients (G/G vs. A/G; P < .05), and no significant difference in FK506. CONCLUSION The CYP 3A4*1B genotype has less influence on tacrolimus metabolism. The genetic polymorphism of CYP 3A5*3 is obvious and largely affects tacrolimus metabolism, and the variant patients need lower doses of tacrolimus to reach the target concentration.
Collapse
Affiliation(s)
- Xia Zhou
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Rujia Tang
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Dali Zhang
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xi He
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhenwen Liu
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yinjie Gao
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongling Liu
- From the Senior Department of Hepatology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
23
|
Xu J, Guo G, Zhou S, Wang H, Chen Y, Lin R, Huang P, Lin C. Physiologically-based pharmacokinetic modeling to predict the exposure and provide dosage regimens of tacrolimus in pregnant women with infection disease. Eur J Pharm Sci 2025; 206:107003. [PMID: 39788164 DOI: 10.1016/j.ejps.2025.107003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Tacrolimus is extensively used for the prevention of graft rejection following solid organ transplantation in pregnant women. However, knowledge gaps in the dosage of tacrolimus for pregnant patients with different CYP3A5 genotypes and infection conditions have been identified. This study aimed to develop a pregnant physiologically based pharmacokinetic (PBPK) model to characterize the maternal and fetal pharmacokinetics of tacrolimus during pregnancy and explore and provide dosage adjustments. We developed PBPK models for nonpregnant patients and validated them via data from previous clinical studies using PK-Sim and Mobi software. To extrapolate to pregnancy, we considered anatomical, physiological, and metabolic alterations and simulated tacrolimus by adding six groups of IL-6 concentrations (0, 5, 25, 50, 500, and 5000 pg/mL). Models were verified by assessing goodness-of-fit plots and ratios of predicted-to-observed pharmacokinetic parameters. The developed PBPK models adequately describe the available clinical data; the fold errors of the predicted and observed values of the area under the curve and peak plasma concentration were between 0.59 and 1.64, and the average folding error and the absolute average folding error values for all concentration-time data points were 1.15 and 1.36, respectively. The simulation results indicated that the area under the steady-state concentration‒time curve and trough concentrations decreased from the first to the third trimester of pregnancy. The trough concentrations were not within the therapeutic range (4-11 ng/mL) in pregnant patients with the CYP3A5 genotype for most of the infection conditions and exceeded its effective concentration in all the CYP3A5 nonexpressers. Based on the model-derived dosing regimen, the tacrolimus trough concentration in pregnant patients with different CYP3A5 genotypes could fall into the therapeutic window, which provided a clinical practice reference for dosage adjustments during pregnancy.
Collapse
Affiliation(s)
- Jianwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Guimu Guo
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shuifang Zhou
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Han Wang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuewen Chen
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Rongfang Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Cuihong Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
24
|
Kovalenko K, Bubalo J, Saultz J, Malla P. Retrospective Analysis of Tacrolimus Levels: The First 56 Days Following Allogeneic Hematopoietic Stem Cell Transplant and Patient Outcomes. Ann Pharmacother 2025:10600280251321324. [PMID: 40012142 DOI: 10.1177/10600280251321324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Despite prophylaxis, acute graft-versus-host disease (aGVHD) occurs in up to 40% to 60% of patients undergoing an allogeneic hematopoietic stem cell transplantation (alloHSCT). Tacrolimus remains a common GVHD prophylactic medication used in combination with mycophenolate or methotrexate. OBJECTIVE The purpose of this study was to compare tacrolimus levels up to day +56 to clinical outcomes in patients who underwent alloHSCT. METHODS This was a retrospective cohort study of adult patients who underwent alloHSCT between January 2009 and April 2019 at Oregon Health and Science University (OHSU) Hospital. A logistic regression analysis was performed using SAS software to evaluate the association between tacrolimus concentration range and the GVHD grade outcome. RESULTS There were 295 patients included in the study. The median patient age was 53 years (range 18-72), the majority were males (55%), with a median comorbidity index of 2 (range 0-9). Most patients received peripheral blood stem cell transplant (95%). The median tacrolimus levels were divided into 4 groups: (1) between 3.8 and 4.9 ng/mL, (2) 5.0 and 7.9 ng/mL, (3) 8.0 and 9.9 ng/mL, and (4) 10.0 and 10.7 ng/mL in 8 (2.7%), 206 (69.8%), 71 (24.1%), and 10 (3.4%) of patients, respectively. The odds ratio of 0.193 (95% confidence interval [CI]: 0.045-0.836) suggested that patients in the tacrolimus 8 to 12 ng/mL range were approximately 80.5% less likely to have grade 3 to 4 aGVHD compared to those in the 5 to 8 ng/mL range. CONCLUSION AND RELEVANCE Overall, we found that higher levels of tacrolimus (range 8-12 ng/mL) in the first 8 weeks post-transplant were associated with improved outcomes without increased rate of relapse.
Collapse
Affiliation(s)
| | - Joseph Bubalo
- Oregon Health & Science University, Portland, OR, USA
| | | | - Pavani Malla
- Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
25
|
Naldi GDAR, Minari AB, Pereira TDM, Fossaluza V, Eugenio NW, Ferreira MA, Gregório GH, Nacif L, D Albuquerque LAC, di Lazzaro Filho R, Cançado ELR, Ono SK. CYP3A5 and POR gene polymorphisms as predictors of infection and graft rejection in post-liver transplant patients treated with tacrolimus - a cohort study. THE PHARMACOGENOMICS JOURNAL 2025; 25:4. [PMID: 39994182 DOI: 10.1038/s41397-025-00363-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/30/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Liver transplantation is the only curative option for patients with advanced stages of liver disease, with tacrolimus used as the immunosuppressive drug of choice. Genetic variability can interfere with drug response, potentially leading to overexposure or underexposure. This study aims to investigate the association of CYP3A4 (rs2740574, rs2242480, rs35599367), CYP3A5 (rs776746, rs10264272), POR (rs1057868) and ABCB1 (rs1128503, rs2229109, rs9282564) gene polymorphisms with infection, acute rejection, and renal failure. The logistic regression model found an influence of CYP3A5 (rs776746) and POR28 (rs1057868) on the development of acute rejection after liver transplantation (p = 0.028). It also found an association between carriers of the variant allele of the POR*28 gene and infection.
Collapse
Affiliation(s)
| | - Ariane Boccoli Minari
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Thales D M Pereira
- Institute of Mathematics and Statistics of the University of São Paulo, São Paulo, Brazil
| | - Victor Fossaluza
- Institute of Mathematics and Statistics of the University of São Paulo, São Paulo, Brazil
| | | | | | | | - Lucas Nacif
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | | | | | - Suzane Kioko Ono
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
26
|
So GC, Lu JBL, Cheng Y, Gisch DL, Koyama S, Melo Ferreira R, Beamon TR, Desta Z, Eadon MT. Inhibition of Tacrolimus Metabolism by Cannabidiol and Its Metabolites In Vitro. Clin Transl Sci 2025; 18:e70152. [PMID: 39921332 PMCID: PMC11806196 DOI: 10.1111/cts.70152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/27/2024] [Accepted: 01/21/2025] [Indexed: 02/10/2025] Open
Abstract
Drug interactions are major causes of interindividual variability in tacrolimus exposure and effect. Tacrolimus, a widely used drug in transplant patients, is metabolized by CYP3A4 and CYP3A5. Cannabidiol (CBD) use after transplant is common. Clinical cases suggest CBD may alter tacrolimus exposure, but the mechanism of this interaction is unknown. We hypothesize that cannabidiol will inhibit tacrolimus metabolism in vitro mainly through CYP3A5 inhibition. In pooled human liver microsomes (HLMs) and recombinant (r) CYP3A4 and CYP3A5 enzymes, tacrolimus (1 μM) metabolism was determined using substrate depletion method in the absence (control) and the presence of 10 μM CBD, 7-hydroxyCBD, and 7-carboxyCBD. Ketoconazole (1 μM) served as a positive control for the inhibition of CYP3A. Linear regression analyses were performed to obtain kinetic parameters of the depletion. Tacrolimus depletion half-life was 2.54, 0.922, and 0.351 min with pooled HLMs, rCYP3A4, and rCYP3A5, respectively. In pooled HLMs, CBD and 7-hydroxyCBD increased tacrolimus half-life by 0.8- and 2.3-fold (both p < 0.0001), respectively. In rCYP3A4, CBD, 7-hydroxyCBD, and ketoconazole prolonged tacrolimus half-life by 5.8-, 14-, and 7.7-fold, respectively. In rCYP3A5, CBD, 7-hydroxyCBD, and ketoconazole prolonged half-life by 29.3-, 19.7-, and 0.1-fold, respectively. In all experiments, 7-carboxyCBD had minimal effect on tacrolimus depletion. CBD and 7-hydroxyCBD inhibited tacrolimus metabolism in vitro. CBD showed stronger inhibition in rCYP3A5 than rCYP3A4. The demonstrated CYP3A5 selectivity of cannabidiol may contribute to the in vitro identification of CYP3A5 substrates in new drug development. Our results support the potential of a clinical drug-drug interaction between CBD and tacrolimus.
Collapse
Affiliation(s)
- Gerald C. So
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jessica Bo Li Lu
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ying‐Hua Cheng
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Debora L. Gisch
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sachiko Koyama
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Travis R. Beamon
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Zeruesenay Desta
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Michael T. Eadon
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
27
|
Wang J, Pasternak AL, Maggo S, Mindanao R, Nguyen JQ, Gong CL. CYP3A5 pharmacogenetic testing for tacrolimus in pediatric heart transplant patients: a budget impact analysis. Pharmacogenet Genomics 2025; 35:81-86. [PMID: 39470413 DOI: 10.1097/fpc.0000000000000549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
BACKGROUND Pharmacogenomic testing can optimize drug efficacy and minimize adverse effects. CYP3A5 polymorphisms affect the metabolism of tacrolimus. We sought to estimate the budget impact of preemptive pharmacogenomic testing for CYP3A5 in pediatric heart transplantation patients from an institutional perspective. METHODS A decision tree was constructed to estimate the budget impact of pediatric heart transplant patients (age ≤18 years) initiated on tacrolimus with and without CYP3A5 pharmacogenomic testing. The budget impact of preemptive pharmacogenomic testing versus no pharmacogenomic testing was calculated. One-way sensitivity analysis and alternative analyses were conducted to assess the robustness of results to changes in model parameters. RESULTS CYP3A5 genotype-guided dosing provided savings of up to $17 225 per patient compared to standard dosing. These savings decreased to $11 759 when using another institution's data for the standard-dosing group. The time to achieve therapeutic concentration in the poor metabolizer genotype-guided dosing group had the largest impact on cost savings while the cost of the pharmacogenetic test had the smallest impact on cost savings. CONCLUSION Implementing CYP3A5 testing could save $17 225 per pediatric heart transplant patient receiving tacrolimus. As pharmacogenomic testing becomes more widespread, institutions should track resource requirements and outcomes to determine the best implementation policies going forward.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Pharmacy, Alfred E. Mann School of Pharmacy, University of Southern California, Los Angeles, California
| | - Amy L Pasternak
- Department of Clinical Pharmacy, University of Michigan, College of Pharmacy, Ann Arbor, Michigan
| | - Simran Maggo
- Department of Pharmacy, Bernard J. Dunn School of Pharmacy, Shenandoah University, Winchester, Virginia
| | | | - Jenny Q Nguyen
- Personalized Care Program, Department of Pathology, Children's Hospital Los Angeles
| | - Cynthia L Gong
- Fetal and Neonatal Institute, Division of Neonatology, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
28
|
Lebreton L, Boyer J, Lafay‐Chebassier C, Hennart B, Baklouti S, Cunat S, Vilquin P, Medard Y, Gautier‐Veyret E, Laffitte‐Redondo C, Verstuyft C, Ait Tayeb AEK, Haufroid V, Wils J, Lamoureux F, Evrard A, Davaze‐Schneider J, Ben‐Sassi M, Picard N, Quaranta S, Ayme‐Dietrich E, the French‐Speaking Network of Pharmacogenetics (RNPGx). French-Speaking Network of Pharmacogenetics (RNPGx) Recommendations for Clinical Use of Mavacamten. Clin Pharmacol Ther 2025; 117:387-397. [PMID: 39584620 PMCID: PMC11739748 DOI: 10.1002/cpt.3502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024]
Abstract
Mavacamten, the first drug in the class of β-cardiac myosin modulator, is used for the treatment of patients with hypertrophic cardiomyopathy. This orally administered drug demonstrates wide interpatient variability in pharmacokinetics parameters, due in part to variant CYP2C19 alleles. Individuals who are CYP2C19 poor metabolizers have increased exposure and are at increased risk of reduced cardiac hypercontractility. To ensure the safety of all patients, European Medicines Agency recommends CYP2C19 preemptive genotyping, and consecutively, to adapt maintenance and initial mavacamten doses, and to manage drug-drug interactions, according to CYP2C19 phenotype. In this article, we summarize evidence from the literature supporting the association between CYP2C19 phenotype and pharmacological features of mavacamten and provide, beyond biologic guidelines, therapeutic recommendations for the use of mavacamten based on CYP2C19 and CYP3A4/CYP3A5 genotype.
Collapse
Affiliation(s)
- Louis Lebreton
- Département de Biochimie, Hôpital PellegrinCentre Hospitalier Universitaire de BordeauxBordeauxFrance
| | - Jean‐Christophe Boyer
- Laboratoire de Biochimie et Biologie MoléculaireCarémeau University HospitalNîmesFrance
| | | | | | - Sarah Baklouti
- Laboratoire de Pharmacocinétique et Toxicologie, Institut Fédératif de BiologieCHU de ToulouseToulouseFrance
- INTHERES, Inrae, ENVT, Université de ToulouseToulouseFrance
| | - Séverine Cunat
- Service d'Hématologie BiologiqueCHU de MontpellierMontpellierFrance
| | - Paul Vilquin
- Department of Tumor Genomics and Pharmacology, Université Paris‐Cité, INSERM UMR‐S 976Saint‐Louis Hospital, AP‐HP ParisParisFrance
| | - Yves Medard
- Department of Tumor Genomics and Pharmacology, Université Paris‐Cité, INSERM UMR‐S 976Saint‐Louis Hospital, AP‐HP ParisParisFrance
| | | | - Clara Laffitte‐Redondo
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris‐Saclay, Assistance Publique‐Hôpitaux de Paris, Hôpital de BicêtreLe Kremlin BicêtreFrance
- MOODS Team, INSERM UMR 1018, CESP, Faculté de MédecineUniv Paris‐SaclayLe Kremlin BicêtreFrance
| | - Céline Verstuyft
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris‐Saclay, Assistance Publique‐Hôpitaux de Paris, Hôpital de BicêtreLe Kremlin BicêtreFrance
- MOODS Team, INSERM UMR 1018, CESP, Faculté de MédecineUniv Paris‐SaclayLe Kremlin BicêtreFrance
| | - Abd El Kader Ait Tayeb
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris‐Saclay, Assistance Publique‐Hôpitaux de Paris, Hôpital de BicêtreLe Kremlin BicêtreFrance
- INSERM UMR‐S U1185, Faculté de MédecineUniv Paris‐SaclayLe Kremlin BicêtreFrance
| | - Vincent Haufroid
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP)Institut de Recherche Expérimentale et Clinique, UClouvainBrusselsBelgium
- Clinical Chemistry DepartmentCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Julien Wils
- Department of Pharmacology, UNIROUEN, INSERM U1096, CHU RouenNormandie UniversityRouenFrance
| | - Fabien Lamoureux
- Department of Pharmacology, UNIROUEN, INSERM U1096, CHU RouenNormandie UniversityRouenFrance
| | - Alexandre Evrard
- Institut du Cancer de Montpellier, ICM, Université de Montpellier, IRCM, Inserm U1194MontpellierFrance
- Laboratoire de Biochimie et Biologie MoléculaireCHU Nîmes‐CarémeauNîmesFrance
| | - Julie Davaze‐Schneider
- Département de Biochimie, Hôpital PellegrinCentre Hospitalier Universitaire de BordeauxBordeauxFrance
| | - Mouna Ben‐Sassi
- Department of Clinical PharmacologyNational Centre Chalbi Belkahia of PharmacovigilanceTunisTunisia
- Faculty of Medicine of TunisUniversity of Tunis El ManarTunisTunisia
| | - Nicolas Picard
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, Centre de Biologie et de Recherche en Santé (CBRS)CHU de LimogesLimogesFrance
| | - Sylvie Quaranta
- Laboratoire de Biologie Moléculaire GENOPé, M2GM/Laboratoire de Pharmacocinétique et Toxicologie, PRISMHôpital de la Timone, AP‐HMMarseilleFrance
| | - Estelle Ayme‐Dietrich
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire, UR7296Hopitaux Universitaires de Strasbourg, Université de StrasbourgStrasbourgFrance
| | | |
Collapse
|
29
|
Lloberas N, Vidal-Alabró A, Colom H. Customizing Tacrolimus Dosing in Kidney Transplantation: Focus on Pharmacogenetics. Ther Drug Monit 2025; 47:141-151. [PMID: 39774592 DOI: 10.1097/ftd.0000000000001289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025]
Abstract
ABSTRACT Different polymorphisms in genes encoding metabolizing enzymes and drug transporters have been associated with tacrolimus pharmacokinetics. In particular, studies on CYP3A4 and CYP3A5, and their combined cluster have demonstrated their significance in adjusting tacrolimus dosing to minimize under- and overexposure thereby increasing the proportion of patients who achieve tacrolimus therapeutic target. Many factors influence the pharmacokinetics of tacrolimus, contributing to inter-patient variability affecting individual dosing requirements. On the other hand, the growing use of population pharmacokinetic models in solid organ transplantation, including different tacrolimus formulations, has facilitated the integration of pharmacogenetic data and other variables into algorithms to easier implement the personalized dose adjustment in transplant centers. The future of personalized medicine in transplantation lies in implementing these models in clinical practice, with pharmacogenetics as a key factor to account for the high inter-patient variability in tacrolimus exposure. To date, three clinical trials have validated the clinical application of these approaches. The aim of this review is to provide an overview of the current studies regarding the different population pharmacokinetic including pharmacogenetics and those translated to the clinical practice for individualizing tacrolimus dose adjustment in kidney transplantation.
Collapse
Affiliation(s)
- Nuria Lloberas
- Nephrology Department, Hospital Universitari de Bellvitge-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL); and
| | - Anna Vidal-Alabró
- Nephrology Department, Hospital Universitari de Bellvitge-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL); and
| | - Helena Colom
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
30
|
Strother A, Park JM, Pasternak AL. Survey of the utilization of genotype-guided tacrolimus management in United States solid organ transplant centers. Pharmacogenomics 2025; 26:89-94. [PMID: 40205800 PMCID: PMC12164375 DOI: 10.1080/14622416.2025.2489920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025] Open
Abstract
INTRODUCTION Genotype-guided tacrolimus management is not routine in clinical practice despite the availability of Clinical Pharmacogenetics Implementation Consortium dosing guidelines. Prior surveys have evaluated patient and provider perspectives of pharmacogenetics (PGx) in transplant, but limited recent data exists on tacrolimus PGx implementation across United States transplant centers. METHODS An electronic survey was distributed to transplant pharmacists regarding utilization of tacrolimus PGx, methods of implementing PGx, and barriers to clinical implementation. A survey response was requested for each organ program within the transplant center. RESULTS A total of 90 programs from 69 transplant centers (28.1% of active U.S. transplant centers) responded to the survey. Tacrolimus PGx was utilized for patient care in 14 programs (15.6%). There was substantial variability in the implementation methods and application of tacrolimus PGx results among transplant programs. In programs that had not implemented tacrolimus PGx, common barriers for implementation included PGx testing cost and availability and lack of evidence for clinical utility. CONCLUSION Implementation of PGx guided tacrolimus in solid organ transplant centers remains limited with heterogeneity in the implementation approach. Additional research is needed to establish the clinical utility of PGx guided tacrolimus and education on reimbursement and testing resources may help to increase uptake.
Collapse
Affiliation(s)
- Ashton Strother
- Department of Pharmacy, University of Michigan Health, Ann Arbor, MI, USA
| | - Jeong M. Park
- Department of Pharmacy, University of Michigan Health, Ann Arbor, MI, USA
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Amy L. Pasternak
- Department of Pharmacy, University of Michigan Health, Ann Arbor, MI, USA
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
31
|
Calleja S, Rodríguez-López A, Ochoa D, Luquero S, Navares-Gómez M, Román M, Mejia-Abril G, Martín-Vilchez S, Abad-Santos F, Zubiaur P. Impact of Genetic Variants on Pregabalin Pharmacokinetics and Safety. Pharmaceuticals (Basel) 2025; 18:151. [PMID: 40005966 PMCID: PMC11860030 DOI: 10.3390/ph18020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Pregabalin is a useful therapeutic option for patients with anxiety or neuropathic pain. Genetic variants in certain genes encoding for transporters related to absorption and distribution could have an impact on the efficacy and safety of the drug. Furthermore, extreme phenotypes in metabolic enzymes could alter pregabalin-limited metabolism. Methods: In this study, we included 24 healthy volunteers participating in a bioequivalence clinical trial and administered pregabalin 300 mg orally; 23 subjects were genotyped for 114 variants in 31 candidate genes, and we explored their impact on pregabalin pharmacokinetics and safety. Results: The uncorrected mean (±SD) of AUC∞ and Cmax were 61,097 ± 14,762 ng*h/mL and 7802 ± 1659 ng/mL, respectively, which were significantly higher in females than in males (p = 0.002 and p = 0.001, respectively), with no differences in dose/weight (DW)- corrected exposure metrics. NAT2 slow acetylators (SAs) showed a 16-18% increase in exposure compared to intermediate (IAs) and normal (NAs) acetylators; NAT2 SAs exhibited a 25% higher t1/2 as compared with NAT2 IAs and 58% higher compared to NAT2 NAs. In contrast, neither the NAT2 phenotype nor other genetic variants were related to pregabalin adverse drug reaction (ADR) occurrence. On the contrary, sex and sex-related exposure differences (i.e., females and their higher exposure compared to males) were the main predictors of ADR occurrence. Conclusions: Our findings suggest that NAT2 could be partially responsible for the minor proportion of pregabalin metabolism, but the effect of NAT2 phenotype does not seem clinically relevant. Therefore, pharmacogenetic biomarkers appear to play a restrained role in pregabalin pharmacotherapy.
Collapse
Affiliation(s)
- Sofía Calleja
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Andrea Rodríguez-López
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Dolores Ochoa
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Sergio Luquero
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Marcos Navares-Gómez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Manuel Román
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Gina Mejia-Abril
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Samuel Martín-Vilchez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain
| |
Collapse
|
32
|
Tremmel R, Hübschmann D, Schaeffeler E, Pirmann S, Fröhling S, Schwab M. Innovation in cancer pharmacotherapy through integrative consideration of germline and tumor genomes. Pharmacol Rev 2025; 77:100014. [PMID: 39952686 DOI: 10.1124/pharmrev.124.001049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 01/22/2025] Open
Abstract
Precision cancer medicine is widely established, and numerous molecularly targeted drugs for various tumor entities are approved or are in development. Personalized pharmacotherapy in oncology has so far been based primarily on tumor characteristics, for example, somatic mutations. However, the response to drug treatment also depends on pharmacological processes summarized under the term ADME (absorption, distribution, metabolism, and excretion). Variations in ADME genes have been the subject of intensive research for >5 decades, considering individual patients' genetic makeup, referred to as pharmacogenomics (PGx). The combined impact of a patient's tumor and germline genome is only partially understood and often not adequately considered in cancer therapy. This may be attributed, in part, to the lack of methods for combined analysis of both data layers. Optimized personalized cancer therapies should, therefore, aim to integrate molecular information, which derives from both the tumor and the germline genome, and taking into account existing PGx guidelines for drug therapy. Moreover, such strategies should provide the opportunity to consider genetic variants of previously unknown functional significance. Bioinformatic analysis methods and corresponding algorithms for data interpretation need to be developed to integrate PGx data in cancer therapy with a special meaning for interdisciplinary molecular tumor boards, in which cancer patients are discussed to provide evidence-based recommendations for clinical management based on individual tumor profiles. SIGNIFICANCE STATEMENT: The era of personalized oncology has seen the emergence of drugs tailored to genetic variants associated with cancer biology. However, the full potential of targeted therapy remains untapped owing to the predominant focus on acquired tumor-specific alterations. Optimized cancer care must integrate tumor and patient genomes, guided by pharmacogenomic principles. An essential prerequisite for realizing truly personalized drug treatment of cancer patients is the development of bioinformatic tools for comprehensive analysis of all data layers generated in modern precision oncology programs.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Daniel Hübschmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between the German Cancer Research Center (DKFZ) and Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Innovation and Service Unit for Bioinformatics and Precision Medicine, DKFZ, Heidelberg, Germany; Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
| | - Sebastian Pirmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between the German Cancer Research Center (DKFZ) and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany; Departments of Clinical Pharmacology, and Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany; DKTK, DKFZ, Partner Site Tuebingen, Tuebingen, Germany; NCT SouthWest, a partnership between DKFZ and University Hospital Tuebingen, Tuebingen, Germany.
| |
Collapse
|
33
|
Litonius K, Kulla N, Falkenbach P, Kristiansson K, Tarkiainen EK, Ukkola-Vuoti L, Cajanus K, Korhonen M, Khan S, Sistonen J, Orpana A, Lindstedt M, Nyrönen T, Perola M, Turpeinen M, Kytö V, Tornio A, Niemi M. Value of Pharmacogenetic Testing Assessed with Real-World Drug Utilization and Genotype Data. Clin Pharmacol Ther 2025; 117:278-288. [PMID: 39365028 DOI: 10.1002/cpt.3458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024]
Abstract
Implementation of pharmacogenetic testing in clinical care has been slow and with few exceptions is hindered by the lack of real-world evidence on how to best target testing. In this retrospective register-based study, we analyzed a nationwide cohort of 1,425,000 patients discharged from internal medicine or surgical wards and a cohort of 2,178 university hospital patients for purchases and prescriptions of pharmacogenetically actionable drugs. Pharmacogenetic variants were obtained from whole genome genotype data for a subset (n = 930) of the university hospital patients. We investigated factors associated with receiving pharmacogenetically actionable drugs and developed a literature-based cost-benefit model for pre-emptive pharmacogenetic panel testing. In a 2-year follow-up, 60.4% of the patients in the nationwide cohort purchased at least one pharmacogenetically actionable drug, most commonly ibuprofen (25.0%) and codeine (19.4%). Of the genotyped subset, 98.8% carried at least one actionable pharmacogenetic genotype and 23.3% had at least one actionable gene-drug pair. Patients suffering from musculoskeletal or cardiovascular diseases were more prone to receive pharmacogenetically actionable drugs during inpatient episode. The cost-benefit model included frequently dispensed drugs in the university hospital cohort, comprising ondansetron (19.4%), simvastatin (7.4%), clopidogrel (5.0%), warfarin (5.1%), (es)citalopram (5.3%), and azathioprine (0.5%). For untargeted pre-emptive pharmacogenetic testing of all university hospital patients, the model indicated saving €17.49 in direct healthcare system costs per patient in 2 years without accounting for the cost of the test itself. Therefore, it might be reasonable to target pre-emptive pharmacogenetic testing to patient groups most likely to receive pharmacogenetically actionable drugs.
Collapse
Affiliation(s)
- Kaisa Litonius
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Noora Kulla
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Petra Falkenbach
- Finnish Coordinating Center for Health Technology Assessment, Oulu University Hospital, University of Oulu, Oulu, Finland
| | | | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Kristiina Cajanus
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mari Korhonen
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Khan
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Johanna Sistonen
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Arto Orpana
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | | | | | - Markus Perola
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Clinical and Molecular Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Miia Turpeinen
- Finnish Coordinating Center for Health Technology Assessment, Oulu University Hospital, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ville Kytö
- Heart Center, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Research Center, Turku University Hospital, Turku, Finland
| | - Aleksi Tornio
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Dash K, Mishra M. The tradeoff between the efficacy of calcineurin inhibitors: prevention of allograft rejection vs. post-transplant renal and cardiovascular complications. Crit Rev Toxicol 2025; 55:63-79. [PMID: 39807635 DOI: 10.1080/10408444.2024.2433631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Solid organ transplantation has emerged as a crucial intervention in the field of medicine. During transplantation, our human body perceives the organ as an exogenous entity or graft, initiating an immune reaction to eliminate it. This immune response ultimately culminates in the rejection of the graft. So, to mitigate the possibility of graft rejection, implementing immune suppression is imperative. In this context, the utilization of calcineurin inhibitors (CNIs) assumes a pivotal role. Calcineurin inhibitors significantly preserve immunosuppression following solid organ transplantation. Calcineurin inhibitors have considerably improved short-term results in renal transplantation by reducing acute rejection rates. Concerning the limited therapeutic window of these medications, careful monitoring of pharmacological treatment and individual doses is required. However, a significant number of patients do experience CNI toxicity. Side effects of CNIs include renal failure, hypertension, respiratory disorders, gastrointestinal damage, gingivitis, and so on. Higher trough level of the drug causes acute nephrotoxicity, which is of three types: functional toxicity, tubular toxicity, and vascular toxicity. Acute nephrotoxicity, if untreated, leads to irreversible, progressive deterioration of allograft function, leading to chronic nephrotoxicity. Cardiovascular toxicity of CNIs includes atrial hypertension caused by vasoconstriction of the afferent arteriole, vascular remodeling, hypertrophy, dyslipidemia, and also the onset of diabetes. Such clinical complications further affect the patient's survivability and subjective well-being, possibly leading to graft loss. This review focuses on the most severe side effects of CNIs: renal and cardiovascular toxicity.
Collapse
Affiliation(s)
- Kalpanarani Dash
- Department of Life Sciences, Neural Developmental Biology Lab, National Institute of Technology, Rourkela, India
| | - Monalisa Mishra
- Department of Life Sciences, Neural Developmental Biology Lab, National Institute of Technology, Rourkela, India
| |
Collapse
|
35
|
González-Iglesias E, Méndez-Ponce C, Ochoa D, Román M, Mejía-Abril G, Martín-Vilchez S, de Miguel A, Gómez-Fernández A, Rodríguez-Lopez A, Soria-Chacartegui P, Abad-Santos F, Novalbos J. Effect of Genetic Variants on Rosuvastatin Pharmacokinetics in Healthy Volunteers: Involvement of ABCG2, SLCO1B1 and NAT2. Int J Mol Sci 2024; 26:260. [PMID: 39796117 PMCID: PMC11720188 DOI: 10.3390/ijms26010260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Statins are the primary drugs used to prevent cardiovascular disease by inhibiting the HMG-CoA reductase, an enzyme crucial for the synthesis of LDL cholesterol in the liver. A significant number of patients experience adverse drug reactions (ADRs), particularly musculoskeletal problems, which can affect adherence to treatment. Recent clinical guidelines, such as those from the Clinical Pharmacogenetics Implementation Consortium (CPIC) in 2022, recommend adjusting rosuvastatin doses based on genetic variations in the ABCG2 and SLCO1B1 genes to minimize ADRs and improve treatment efficacy. Despite these adjustments, some patients still experience ADRs. So, we performed a candidate gene study to better understand the pharmacogenetics of rosuvastatin. This study included 119 healthy volunteers who participated in three bioequivalence trials of rosuvastatin alone or in combination with ezetimibe at the Clinical Trials Unit of the Hospital Universitario de La Princesa (UECHUP). Participants were genotyped using a custom OpenArray from ThermoFisher that assessed 124 variants in 38 genes associated with drug metabolism and transport. No significant differences were observed according to sex or biogeographic origin. A significant increase in t1/2 (pmultivariate(pmv) = 0.013) was observed in the rosuvastatin plus ezetimibe trial compared with the rosuvastatin alone trials. Genetic analysis showed that decreased (DF) and poor function (PF) volunteers for the ABCG2 transporter had higher AUC∞/DW (adjusted dose/weight), AUC72h/DW and Cmax/DW compared to normal function (NF) volunteers (pmv< 0.001). DF and PF volunteers for SLCO1B1 showed an increase in AUC72h/DW (pmv = 0.020) compared to increased (IF) and NF individuals. Results for ABCG2 and SLCO1B1 were consistent with the existing literature. In addition, AUC∞/DW, AUC72h/DW and Cmax/DW were increased in intermediate (IA) and poor (PA) NAT2 acetylators (pmv = 0.001, pmv< 0.001, pmv< 0.001, respectively) compared to rapid acetylators (RA), which could be associated through a secondary pathway that was previously unknown.
Collapse
Affiliation(s)
- Eva González-Iglesias
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Clara Méndez-Ponce
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| | - Dolores Ochoa
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Manuel Román
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| | - Gina Mejía-Abril
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| | - Samuel Martín-Vilchez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| | - Alejandro de Miguel
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| | - Antía Gómez-Fernández
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Andrea Rodríguez-Lopez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesús Novalbos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), 28006 Madrid, Spain; (E.G.-I.)
| |
Collapse
|
36
|
Piriyapongsa J, Chumnumwat S, Kaewprommal P, Triparn K, Suvichapanich S, Udomsinprasert W, Jittikoon J, Shaw PJ, Nakhonsri V, Ngamphiw C, Wangkumhang P, Pithukpakorn M, Roothumnong E, Wiboonthanasarn S, Kuptanon C, Jinawath N, Porntaveetus T, Suriyaphol P, Viprakasit V, Pisitkun P, Kantaputra P, Tim-Aroon T, Wattanasirichaigoon D, Sura T, Suphapeetiporn K, Sripichai O, Khongphatthanayothin A, Fucharoen S, Ngamphaiboon N, Shotelersuk V, Mahasirimongkol S, Tongsima S. Pharmacogenomic landscape of the Thai population from genome sequencing of 949 individuals. Sci Rep 2024; 14:30683. [PMID: 39730427 DOI: 10.1038/s41598-024-79018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/04/2024] [Indexed: 12/29/2024] Open
Abstract
Inter-individual variability in drug responses is significantly influenced by genetic factors, underscoring the importance of population-specific pharmacogenomic studies to optimize clinical outcomes. In this study, we analyzed whole genome sequencing data from 949 unrelated Thai individuals and conducted an in-depth analysis of 3239 genes involved in drug pharmacokinetics, pharmacodynamics, or immune-mediated adverse drug reactions. We identified 43 single nucleotide polymorphisms (SNPs), 134 diplotypes, and 15 human leukocyte antigen (HLA) alleles, all with moderate to high clinical significance. On average, each Thai individual carried 14 SNPs, one to two HLA alleles, and six diplotypes with actionable phenotypic associations. Clinically important diplotypes were present in over 20% of individuals for seven genes (CYP2A6, CYP2B6, CYP2C19, CYP3A5, NAT2, SLCO1B1, and VKORC1). In addition, clinically significant SNPs with allele frequencies exceeding 20% were identified among 15 genes, including VKORC1, CYP4F2, and ABCG2. We also identified 21,211 potentially deleterious variants among 3239 genes. Of these variants, 3746 were novel. The comprehensive dataset from this study serves as a valuable resource of pharmacogenomic variants in the Thai population, which will facilitate the development of personalized drug therapies and enhance patient care in Thailand.
Collapse
Affiliation(s)
- Jittima Piriyapongsa
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Supatat Chumnumwat
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Pavita Kaewprommal
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Kwankom Triparn
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | | | | | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Philip J Shaw
- Medical Molecular Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Vorthunju Nakhonsri
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Chumpol Ngamphiw
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Pongsakorn Wangkumhang
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Manop Pithukpakorn
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ekkapong Roothumnong
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supakit Wiboonthanasarn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chulaluck Kuptanon
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
- Department of Pediatrics, College of Medicine, Rangsit University, Pathum Thani, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
- Integrative Computational BioScience Center (ICBS), Mahidol University, Nakhon Pathom, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prapat Suriyaphol
- Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vip Viprakasit
- Division of Hematology & Oncology, Department of Pediatrics & Siriraj Thalassemia Center, Siriraj Research Hospital, Mahidol University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Piranit Kantaputra
- Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Center of Excellence in Medical Genetics Research, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Thipwimol Tim-Aroon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangrurdee Wattanasirichaigoon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyachai Sura
- Medical Genetics and Molecular Medicine Unit, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Orapan Sripichai
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Apichai Khongphatthanayothin
- Division of Cardiology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Nuttapong Ngamphaiboon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Surakameth Mahasirimongkol
- Information and Communication Technology Center, Office of Permanent Secretary, Ministry of Public Health, Nonthaburi, Thailand.
| | - Sissades Tongsima
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand.
| |
Collapse
|
37
|
Han L, Cui Y, Pan Y, Chen R, Jiao Z. External evaluation of tacrolimus population pharmacokinetic models in adult lung transplant patients: How to enhance the predictive ability of the model? Int Immunopharmacol 2024; 143:113225. [PMID: 39353393 DOI: 10.1016/j.intimp.2024.113225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Tacrolimus is the cornerstone of current immunosuppressive strategies after lung transplantation. However, its narrow therapeutic range and considerable pharmacokinetic variability pose challenges for individualized treatment. Several tacrolimus population pharmacokinetic (popPK) models have been developed for precision dosing in adult lung transplant patients. However, their applicability across different clinical settings remains uncertain. The aim of this study was to evaluate the external predictability of these models and identify influential factors. METHODS Published models were systematically retrieved and assessed based on an external dataset of 39 patients (1240 tacrolimus trough concentrations) using three approaches: (1) prediction-based diagnosis using dosing records and patient characteristics; (2) simulation-based diagnosis, with prediction- and variability-corrected visual predictive checks (pvcVPC) and normalized prediction distribution error tests (NPDE); and (3) Bayesian forecasting using one to four observations for posterior predictions. We also investigated the impact of model structure and covariates on predictability. RESULTS The predictive performance of six published models was externally evaluated, but none demonstrated satisfactory accuracy in prediction- and simulation-based diagnosis. Bayesian forecasting yielded satisfactory results with only one prior observation and optimal predictive performance with 2-3 priors for all included models. The structural model parameterized on plasma tacrolimus concentration outperformed others. Significant correlations were observed between prediction-error and daily tacrolimus dose, postoperative day, and voriconazole co-administration. CONCLUSIONS The overall predictive performance of all published models was unsatisfactory, making direct extrapolation inappropriate. However, Bayesian forecasting significantly improves predictive performance. Utilizing plasma tacrolimus concentration for parameter estimation can improve the predictive ability of tacrolimus popPK models.
Collapse
Affiliation(s)
- Lu Han
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yifan Cui
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yan Pan
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rui Chen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Zhang Y, Wang Z, Wang Y, Jin W, Zhang Z, Jin L, Qian J, Zheng L. CYP3A4 and CYP3A5: the crucial roles in clinical drug metabolism and the significant implications of genetic polymorphisms. PeerJ 2024; 12:e18636. [PMID: 39650550 PMCID: PMC11625447 DOI: 10.7717/peerj.18636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024] Open
Abstract
CYP3A, a key member of the cytochrome P450 (CYP450) superfamily, is integral to drug metabolism, processing a substantial portion of medications. Their role in drug metabolism is particularly prominent, as CYP3A4 and CYP3A5 metabolize approximately 30-50% of known drugs. The genetic polymorphism of CYP3A4/5 is significant inter-individual variability in enzymatic activity, which can result in different pharmacokinetic profiles in response to the same drug among individuals. These polymorphisms can lead to either increased drug toxicity or reduced therapeutic effects, requiring dosage adjustments based on genetic profiles. Consequently, the study of the enzymatic activity of CYP3A4/5 gene variants is of great importance for the formulation of personalized treatment regimens. This article first reviews the role of CYP3A4/5 in drug metabolism in the human body, including inhibitors and inducers of CYP3A4/5 and drug-drug interactions. In terms of genetic polymorphism, it discusses the detection methods, enzymatic kinetic characteristics, and clinical guidelines for CYP3A5. Finally, the article summarizes the importance of CYP3A4/5 in clinical applications, including personalized therapy, management of drug-drug interactions, and adjustment of drug doses. This review contributes to the understanding of the functions and genetic characteristics of CYP3A4/5, allowing for more effective clinical outcomes through optimized drug therapy.
Collapse
Affiliation(s)
- Yuqing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziying Wang
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuchao Wang
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weikai Jin
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheyan Zhang
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lehao Jin
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianchang Qian
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Long Zheng
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
39
|
Minichmayr IK, Dreesen E, Centanni M, Wang Z, Hoffert Y, Friberg LE, Wicha SG. Model-informed precision dosing: State of the art and future perspectives. Adv Drug Deliv Rev 2024; 215:115421. [PMID: 39159868 DOI: 10.1016/j.addr.2024.115421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
Model-informed precision dosing (MIPD) stands as a significant development in personalized medicine to tailor drug dosing to individual patient characteristics. MIPD moves beyond traditional therapeutic drug monitoring (TDM) by integrating mathematical predictions of dosing and considering patient-specific factors (patient characteristics, drug measurements) as well as different sources of variability. For this purpose, rigorous model qualification is required for the application of MIPD in patients. This review delves into new methods in model selection and validation, also highlighting the role of machine learning in improving MIPD, the utilization of biosensors for real-time monitoring, as well as the potential of models integrating biomarkers for efficacy or toxicity for precision dosing. The clinical evidence of TDM and MIPD is discussed for various medical fields including infection medicine, oncology, transplant medicine, and inflammatory bowel diseases, thereby underscoring the role of pharmacokinetics/pharmacodynamics and specific biomarkers. Further research, particularly randomized clinical trials, is warranted to corroborate the value of MIPD in enhancing patient outcomes and advancing personalized medicine.
Collapse
Affiliation(s)
- I K Minichmayr
- Dept. of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - E Dreesen
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - M Centanni
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Z Wang
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Y Hoffert
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - L E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - S G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
40
|
Hemani R, Chauhan PM, Srivastava R, Shete NB, Jojera AS, Soni SM, Gang SD, Konnur AM, Hegde UN, Patel HB, Mukhopadhyay BN, Raval MA, Pandey SN. Synergistic Effect of Cytochrome P450 Family 3 Subfamily A Member 5 ( CYP3A5) Genetic Variants in Tacrolimus Dose Determination in Indian Renal Transplant Patients. ACS Pharmacol Transl Sci 2024; 7:3429-3438. [PMID: 39539273 PMCID: PMC11555502 DOI: 10.1021/acsptsci.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/15/2024] [Accepted: 07/30/2024] [Indexed: 11/16/2024]
Abstract
Tacrolimus (TAC) has a narrow therapeutic index and shows interindividual variabilities in its blood concentration. Although guidelines recommend a genetic variant (rs776746) to determine the optimized TAC dose, discrepancies in accuracy have been noted. Therefore, studying other variants of CYP3A5 may improve the accuracy of the TAC dose. Clinical exome sequencing (CES) was performed in 219 renal transplant patients. The SNPs of CYP3A5 covered by CES were recorded. The TAC blood trough concentration/dose (C 0/D) was calculated on day 7 and months 1, 3, 6, and 12 of post-transplantation, and association with CYP3A5 genotypes was studied. Further, biopsy-proven rejection and pathological events were analyzed for their association with CYP3A5 genotypes. Out of 35 variants of CYP3A5 covered in CES, rs776746, rs15524, rs4646449, and rs464645 were significantly associated with the TAC C 0/D on day 7 and months 1, 3, and 6. Further analysis showed that the slow-metabolizing genotypes of all four SNPs synergistically associated with the TAC C 0/D on day 7 and months 1, 3, 6, and 12. The "CC" genotype of rs776746 showed a significant association (RR = 1.613; p = 0.035) with allograft rejection. In addition, cox regression analysis showed that the presence of the "CA" genotype of rs4646453 increased (HR = 7.258; 95% CI = 1.354-38.904) the risk of development of pathological events, respectively. Four variants of CYP3A5 were synergistically associated with the TAC dose determination. In addition, rs776746 and rs4646453 may be associated with allograft rejection and pathological events, respectively.
Collapse
Affiliation(s)
- Rashmi
J. Hemani
- Department
of Pathology, Muljibhai Patel Urological
Hospital, Dr. V. V. Desai Road, Nadiad 387001, Gujarat, India
- Department
of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, Gujarat, India
| | - Priyal M. Chauhan
- Department
of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, Gujarat, India
| | - Ratika Srivastava
- School
of Life Sciences, Department of Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow 226025, Uttar Pradesh, India
| | - Nitiraj B. Shete
- Department
of Biostatistics, Muljibhai Patel Urological
Hospital, Nadiad 387001, Gujarat, India
| | - Amit S. Jojera
- Department
of Pathology, Muljibhai Patel Urological
Hospital, Dr. V. V. Desai Road, Nadiad 387001, Gujarat, India
| | - Shailesh M. Soni
- Department
of Pathology, Muljibhai Patel Urological
Hospital, Dr. V. V. Desai Road, Nadiad 387001, Gujarat, India
| | - Sishir D. Gang
- Department
of Nephrology, Muljibhai Patel Urological
Hospital, Nadiad 387001, Gujarat, India
| | - Abhijit M. Konnur
- Department
of Nephrology, Muljibhai Patel Urological
Hospital, Nadiad 387001, Gujarat, India
| | - Umapati N. Hegde
- Department
of Nephrology, Muljibhai Patel Urological
Hospital, Nadiad 387001, Gujarat, India
| | - Hardik B. Patel
- Department
of Nephrology, Muljibhai Patel Urological
Hospital, Nadiad 387001, Gujarat, India
| | - Banibrata N. Mukhopadhyay
- Department
of Pathology, Muljibhai Patel Urological
Hospital, Dr. V. V. Desai Road, Nadiad 387001, Gujarat, India
| | - Manan A. Raval
- Department
of Pharmacognosy, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, Gujarat, India
| | - Sachchida Nand Pandey
- Department
of Pathology, Muljibhai Patel Urological
Hospital, Dr. V. V. Desai Road, Nadiad 387001, Gujarat, India
| |
Collapse
|
41
|
Khatri D, Felmingham B, Moore C, Lazaraki S, Stenta T, Collier L, Elliott DA, Metz D, Conyers R. Evaluating the evidence for genotype-informed Bayesian dosing of tacrolimus in children undergoing solid organ transplantation: A systematic literature review. Br J Clin Pharmacol 2024; 90:2724-2741. [PMID: 39147586 DOI: 10.1111/bcp.16203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/04/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Tacrolimus, a calcineurin inhibitor, is a highly effective immunosuppressant used in solid organ transplantation (SOT). However, it is characterized by a narrow therapeutic range and high inter-patient variability in pharmacokinetics. Standard weight-based dosing followed by empiric dose titration is suboptimal in controlling drug concentrations, increasing risk of rejection or toxicity, particularly in the initial months post transplantation. This review explores the potential of combined pre-transplant genotyping and pharmacokinetic (PK) modelling to improve tacrolimus dosing in paediatric SOT recipients. A systematic search of Medline, Embase and Cochrane databases identified studies published between March 2013 and March 2023 that investigated genotype- and PK model-informed tacrolimus dosing in children post-SOT. The Newcastle-Ottawa Scale assessed study quality. Seven studies encompassing paediatric kidney, heart, liver and lung transplants reported using genotype and model-informed dosing. A combination of clinical and genetic factors significantly impacts tacrolimus clearance and thus initial dose recommendation. Body size, transplant organ and co-medications were consistently important, while either time post-transplant or haematocrit emerged in some studies. Several models were identified, however, with limitations evident in some and with absence of evidence for their effectiveness in optimizing initial and subsequent dosing. This review highlights the development of PK models in paediatric SOT that integrate genotype and clinical covariates to personalize early tacrolimus dosing. While promising, prospective studies are needed to validate and confirm their effectiveness in improving time to therapeutic concentrations and reducing under- or overexposure. This approach has the potential to optimize tacrolimus therapy in paediatric SOT, thereby improving outcomes.
Collapse
Affiliation(s)
- Dhrita Khatri
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
| | - Ben Felmingham
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
| | - Claire Moore
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Melbourne, VIC, Australia
| | - Smaro Lazaraki
- Health Sciences Library, Royal Melbourne Hospital, Melbourne Health, Australia
| | - Tayla Stenta
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
| | - Lane Collier
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
| | - David A Elliott
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Melbourne, VIC, Australia
| | - David Metz
- Department of Nephrology, The Royal Children's Hospital, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Rachel Conyers
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Melbourne, VIC, Australia
- Children's Cancer Centre, The Royal Children's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Saqr A, Al-Kofahi M, Mohamed M, Dorr C, Remmel RP, Onyeaghala G, Oetting WS, Guan W, Mannon RB, Matas AJ, Israni A, Jacobson PA. Steroid-tacrolimus drug-drug interaction and the effect of CYP3A genotypes. Br J Clin Pharmacol 2024; 90:2837-2848. [PMID: 38994750 DOI: 10.1111/bcp.16172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
AIMS Tacrolimus, metabolized by CYP3A4 and CYP3A5 enzymes, is susceptible to drug-drug interactions (DDI). Steroids induce CYP3A genes to increase tacrolimus clearance, but the effect is variable. We hypothesized that the extent of the steroid-tacrolimus DDI differs by CYP3A4/5 genotypes. METHODS Kidney transplant recipients (n = 2462) were classified by the number of loss of function alleles (LOF) (CYP3A5*3, *6 and *7 and CYP3A4*22) and steroid use at each tacrolimus trough in the first 6 months post-transplant. A population pharmacokinetic analysis was performed by nonlinear mixed-effect modelling (NONMEM) and stepwise covariate modelling to define significant covariates affecting tacrolimus clearance. A stochastic simulation was performed and translated into a Shiny application with the mrgsolve and Shiny packages in R. RESULTS Steroids were associated with modestly higher (3%-11.8%) tacrolimus clearance. Patients with 0-LOF alleles receiving steroids showed the greatest increase (11.8%) in clearance compared to no steroids, whereas those with 2-LOFs had a negligible increase (2.6%) in the presence of steroids. Steroid use increased tacrolimus clearance by 5% and 10.3% in patients with 1-LOF and 3/4-LOFs, respectively. CONCLUSIONS Steroids increase the clearance of tacrolimus but vary slightly by CYP3A genotype. This is important in individuals of African ancestry who are more likely to carry no LOF alleles, may more commonly receive steroid treatment, and will need higher tacrolimus doses.
Collapse
Affiliation(s)
- Abdelrahman Saqr
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Mahmoud Al-Kofahi
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
- Gilead Sciences, Inc., Foster City, California, USA
| | - Moataz Mohamed
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Casey Dorr
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rory P Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guillaume Onyeaghala
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - William S Oetting
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Roslyn B Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska, Omaha, Nebraska, USA
| | - Arthur J Matas
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ajay Israni
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Pamala A Jacobson
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| |
Collapse
|
43
|
Miano TA, Zuppa AF, Feng R, Griffiths S, Kalman L, Oyster M, Cantu E, Yang W, Diamond JM, Christie JD, Scheetz MH, Shashaty MG. Development and validation of a population pharmacokinetic model to guide perioperative tacrolimus dosing after lung transplantation. JHLT OPEN 2024; 6:100134. [PMID: 40145052 PMCID: PMC11935331 DOI: 10.1016/j.jhlto.2024.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background Tacrolimus therapy is standard of care for immunosuppression after lung transplantation. However, tacrolimus exposure variability during the early postoperative period may contribute to poor outcomes in this population. Few studies have examined tacrolimus pharmacokinetics (PK) during this high-risk period. Methods We conducted a retrospective pharmacokinetic study in lung transplant recipients at the University of Pennsylvania who were enrolled in the Lung Transplant Outcomes Group cohort. We used nonlinear mixed-effects regression to derive a population PK model in 270 patients and examined validity in a separate cohort of 114 patients. Covariates were examined with univariate analysis and a multivariable model was developed using forward and backward stepwise selection. The performance of the final model in the validation cohort was examined with calculation of prediction error (PE). Results We developed a 1-compartment base model with a fixed rate absorption constant. Covariates improving model fit were postoperative day, hematocrit, transplant type, CYP3A5 genotype, weight, and exposure to cytochrome p450 enzyme (CYP) inhibitor drugs. The strongest predictor of tacrolimus clearance was postoperative day, with median predicted clearance increasing more than 3-fold over the 14-day study period. In the validation cohort, the final model showed a mean PE of 36.4% (95% confidence interval 30.8%-41.9%) and a median PE of 7.2% (interquartile range -29.3% to 70.53%). Conclusions Tacrolimus clearance is highly dynamic during the early postlung transplant period. Population PK models that include lung-transplant-specific covariates may enable precision dosing algorithms that account for this highly dynamic clearance. Future multicenters studies including a broader set of covariates are warranted.
Collapse
Affiliation(s)
- Todd A. Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Real-world Effectiveness and Safety of Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Rui Feng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen Griffiths
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laurel Kalman
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Oyster
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wei Yang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Real-world Effectiveness and Safety of Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joshua M. Diamond
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D. Christie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marc H. Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University, Downers Grove, Illinois
| | - Michael G.S. Shashaty
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Li WJ, Lu Y, Zhong ZB, Gao SY, Xue CB, Hu W, Liu ZZ, Wang XX, Deng ZG, Ye SJ, Cheng H. Impact of Paxlovid on Tacrolimus Concentration in Kidney Transplant Patients: A Retrospective Observational Study. Transplant Proc 2024; 56:1954-1960. [PMID: 39500638 DOI: 10.1016/j.transproceed.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/04/2024] [Indexed: 12/10/2024]
Abstract
Kidney transplant recipients, reliant on lifelong immunosuppressive therapy, face potential drug interactions with emerging treatments such as paxlovid. This study aims to provide guidance for safe administration by examining the impact of paxlovid on tacrolimus levels in kidney transplant recipients. Seven kidney transplant patients who received paxlovid between December 2022 and August 2023 were included for retrospective analysis. Tacrolimus concentration changes were investigated both during and after the administration of paxlovid. Genetic testing for CYP3A5 polymorphisms assessed individual responses. The patient's treatment process was divided into four phases according to the paxlovid administration and the Tacrolimus trough level. The variation of tacrolimus valley concentration, concentration-to-dose ratios (C/D), and creatinine values in different periods were subsequently described and analyzed. The results indicate that tacrolimus levels increased significantly after receiving paxlovid, peaking on day two with a median trough level of 21.8 ng/mL. Prior to the administration of paxlovid, the median C/D value was 6.8 times higher (903.1 to 132.5). Once the paxlovid was stopped, the C/D value and trough level progressively returned to their preadministration levels. Importantly, no graft rejections, adverse events, or neurotoxicity were noted. The levels of creatinine remained stable. During paxlovid treatment, patients adhered to a modified tacrolimus regimen and progressively resumed baseline dosage. In summary, this is the first study to indicate a significant influence of paxlovid on tacrolimus levels in Chinese patients undergoing kidney transplantation. During paxlovid treatment, careful observation and tailored tacrolimus management are crucial to guarantee safe administration and circumvent negative consequences.
Collapse
Affiliation(s)
- Wen-Jing Li
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yun Lu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zi-Biao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Su-Yu Gao
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Cheng-Biao Xue
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhong-Zhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Xuan-Xuan Wang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gao Deng
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Shao-Jun Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
45
|
Peña-Martín MC, Marcos-Vadillo E, García-Berrocal B, Heredero-Jung DH, García-Salgado MJ, Lorenzo-Hernández SM, Larrue R, Lenski M, Drevin G, Sanz C, Isidoro-García M. A Comparison of Molecular Techniques for Improving the Methodology in the Laboratory of Pharmacogenetics. Int J Mol Sci 2024; 25:11505. [PMID: 39519058 PMCID: PMC11546559 DOI: 10.3390/ijms252111505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
One of the most critical goals in healthcare is safe and effective drug therapy, which is directly related to an individual's response to treatment. Precision medicine can improve drug safety in many scenarios, including polypharmacy, and it requires the development of new genetic characterization methods. In this report, we use real-time PCR, microarray techniques, and mass spectrometry (MALDI-TOF), which allows us to compare them and identify the potential benefits of technological improvements, leading to better quality medical care. These comparative studies, as part of our pharmacogenetic Five-Step Precision Medicine (5SPM) approach, reveal the superiority of mass spectrometry over the other methods analyzed and highlight the importance of updating the laboratory's pharmacogenetic methodology to identify new variants with clinical impact.
Collapse
Affiliation(s)
- María Celsa Peña-Martín
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Pharmacology-Toxicology and Pharmacovigilance Department, Angers University Hospital, F-49100 Angers, France;
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Elena Marcos-Vadillo
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Belén García-Berrocal
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - David Hansoe Heredero-Jung
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - María Jesús García-Salgado
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Sandra Milagros Lorenzo-Hernández
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Romain Larrue
- CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, University of Lille, F-59000 Lille, France;
| | - Marie Lenski
- CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPECS-IMPact of the Chemical Environment on Health, University of Lille, F-59000 Lille, France;
| | - Guillaume Drevin
- Pharmacology-Toxicology and Pharmacovigilance Department, Angers University Hospital, F-49100 Angers, France;
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
| | - María Isidoro-García
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
46
|
Concha J, Sangüesa E, Ribate MP, García CB. CYP3A4*1B but Not CYP3A5*3 as Determinant of Long-Term Tacrolimus Dose Requirements in Spanish Solid Organ Transplant Patients. Int J Mol Sci 2024; 25:11327. [PMID: 39457109 PMCID: PMC11508189 DOI: 10.3390/ijms252011327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Tacrolimus (TAC) is a commonly used immunosuppressive drug in solid organ transplantation. Pharmacogenetics has been demonstrated before to be decisive in TAC pharmacotherapy. The CYP3A5*3 variant has been reported to be the main determinant of TAC dose requirements; however, other polymorphisms have also proven to be influential, especially in CYP3A5 non-expressor patients. The aim of this study is to evaluate the influence of genetic polymorphisms in TAC therapy in a cohort of Spanish transplant recipients. Genetic analysis including ten polymorphic variants was performed, and demographic and clinical data and pharmacotherapy of 26 patients were analyzed. No significant differences were found in weight-adjusted dose between CYP3A5 expressors and non-expressors (0.047 mg/kg vs. 0.044 mg/kg), while they were found for carriers of the CYP3A4*1B allele (0.101 mg/kg; p < 0.05). The results showed that patients with at least one CYP3A4*1B allele had a higher TAC dose and lower blood concentration. Dose-adjusted TAC blood levels were also lower in CYP3A4*1B carriers compared to non-carriers (0.72 ng/mL/mg vs. 2.88 ng/mL/mg). These results support the independence of CYP3A5*3 and CYP3A4*1B variants as determinants of dose requirements despite the linkage disequilibrium present between the two. The variability in genotype frequency between ethnicities may be responsible for the discrepancy found between studies.
Collapse
Affiliation(s)
| | | | - María Pilar Ribate
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, E-50830 Villanueva de Gállego, Zaragoza, Spain; (J.C.); (E.S.); (C.B.G.)
| | | |
Collapse
|
47
|
González-Iglesias E, Ochoa D, Navares-Gómez M, Zubiaur P, Aldama M, de la Torre T, Ríos-Rodríguez MDL, Soria-Chacartegui P, Rodríguez-Lopez A, Abad-Santos F, Novalbos J. Evaluation of the role of metabolizing enzymes and transporter variants in ezetimibe pharmacokinetics. Front Pharmacol 2024; 15:1414059. [PMID: 39484171 PMCID: PMC11524821 DOI: 10.3389/fphar.2024.1414059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Ezetimibe inhibits cholesterol uptake by modulation of intestinal sterol absorption. Currently, although some studies have shown alterations in ezetimibe levels caused by alterations in the ABCG5, ABCG8, NPC1L1 or UGT1A1 genes, there are no pharmacogenetic guidelines to confirm these biomarkers. The aim of this work was to evaluate the effect of 49 variants in 22 pharmacogenes related to metabolism and transport. Methods A total of 96 healthy volunteers from four bioequivalence clinical trials of ezetimibe as monotherapy or in combination with simvastatin were studied. Blood samples were extracted for unconjugated ezetimibe plasma quantification and genotyping. Results and Discussion No association of metabolizing enzyme variants with ezetimibe pharmacokinetic parameters was found. The results show some trends in the univariate analysis for ABCB1 rs2032582 or ABCC2 rs2273697 and Cmax (p univariate (p uv ) = 0.056 and 0.087, respectively), which finally reach significance in the multivariate analysis (p multivariate (p mv ) = 0.049 and 0.048, respectively). Nevertheless, these results need to be validated in future studies.
Collapse
Affiliation(s)
- Eva González-Iglesias
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Dolores Ochoa
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Marcos Navares-Gómez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Marina Aldama
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
| | - Tamara de la Torre
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
| | - Marta de los Ríos-Rodríguez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
| | - Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Andrea Rodríguez-Lopez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
- Pharmacology Department, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Novalbos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Madrid, Spain
| |
Collapse
|
48
|
Prasad B, Al-Majdoub ZM, Wegler C, Rostami-Hodjegan A, Achour B. Quantitative Proteomics for Translational Pharmacology and Precision Medicine: State of The Art and Future Outlook. Drug Metab Dispos 2024; 52:1208-1216. [PMID: 38821856 DOI: 10.1124/dmd.124.001600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Over the past 20 years, quantitative proteomics has contributed a wealth of protein expression data, which are currently used for a variety of systems pharmacology applications, as a complement or a surrogate for activity of the corresponding proteins. A symposium at the 25th North American International Society for the Study of Xenobiotics meeting, in Boston, in September 2023, was held to explore current and emerging applications of quantitative proteomics in translational pharmacology and strategies for improved integration into model-informed drug development based on practical experience of each of the presenters. A summary of the talks and discussions is presented in this perspective alongside future outlook that was outlined for future meetings. SIGNIFICANCE STATEMENT: This perspective explores current and emerging applications of quantitative proteomics in translational pharmacology and precision medicine and outlines the outlook for improved integration into model-informed drug development.
Collapse
Affiliation(s)
- Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Zubida M Al-Majdoub
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Christine Wegler
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Amin Rostami-Hodjegan
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Brahim Achour
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| |
Collapse
|
49
|
Parra-Astorgano L. [Pharmaceutical Indication Service in a case of palmar-plantar erythema after amoxicillin and ibuprofen treatment]. FARMACEUTICOS COMUNITARIOS 2024; 16:83-87. [PMID: 39439871 PMCID: PMC11491920 DOI: 10.33620/fc.2173-9218.(2024).22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 10/25/2024]
Abstract
Case description Patient (29 years old) with palmo-plantar erythema, goes to the community pharmacy (FC) requesting a cream to treat atopy. Evaluation The patient accessed the Pharmaceutical Indication Service (SPIF), showing that the manifestations appeared 24 hours after the start of dental treatment with amoxicillin 1g/12h and ibuprofen 600 mg/8h without any concomitant medication. Intervention After explaining the possible relationship of the symptoms with their medication, patient was derived to the doctor with the referral report completed by SEFAC-eXPERT. Results The patient went to the emergency where she was treated with intravenous corticosteroid and a prescription for cetirizine 10 mg. The dentist changed the beta-lactam to a macrolide (azithromycin) and the ibuprofen to paracetamol. From the FC, the evolution of the symptoms was monitored, which took 72 hours to disappear. Allergy tests suggested avoiding beta-lactams, cephalosporins, and arylpropionics without being conclusive. Months later, the patient suffered similar symptoms after inhaling a disinfectant spray and the allergy diagnosis was confirmed. Conclusions The FC identified and immediately referred using SPIF a case of hypersensitivity in a patient susceptible to RNM and the SPIF helps to record the intervention and follow-up, increasing patient safety.
Collapse
Affiliation(s)
- Lola Parra-Astorgano
- Farmacéutica Comunitaria. Profesora Asociada CC Salud, Facultad de Farmacia, USAL. Miembro del grupo de trabajo en Indicación SEFACEspaña
| |
Collapse
|
50
|
Montero-Gómez A, Sánchez Pozo A. [Selection of pharmacogenomic variants and methodology for their use in community pharmacy]. FARMACEUTICOS COMUNITARIOS 2024; 16:61-82. [PMID: 39439868 PMCID: PMC11491914 DOI: 10.33620/fc.2173-9218.(2024).27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Regulatory agencies such as the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) recognize pharmacogenetics as a key tool in their pharmacological guidelines for pharmaceutical counseling. In this context, community pharmacies play a crucial role in addressing this healthcare need, which could lead to a significant improvement in patients' quality of life by preventing ineffective or contraindicated treatments.In this work, we conducted a systematic review of the available scientific evidence regarding druggene interactions relevant to community pharmacy. We identified the main genes and polymorphisms associated with treatment response and adverse effects in primary care. Finally, we propose a model for implementing pharmacogenetic services in community pharmacies.
Collapse
Affiliation(s)
- A Montero-Gómez
- Farmacéutica Comunitaria y Máster en Atención Farmacéutica. Granada.España
| | - A Sánchez Pozo
- Catedrático de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Granada.España
| |
Collapse
|