1
|
Ho TT, Smith DM, Aquilante CL, Cicali EJ, El Rouby N, Hertz DL, Imanirad I, Patel JN, Scott SA, Swain SM, Tuteja S, Hicks JK. A Guide for Implementing DPYD Genotyping for Systemic Fluoropyrimidines into Clinical Practice. Clin Pharmacol Ther 2025; 117:1194-1208. [PMID: 39887719 PMCID: PMC11993294 DOI: 10.1002/cpt.3567] [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: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025]
Abstract
The safety of systemic fluoropyrimidines (e.g., 5-fluorouracil, capecitabine) is impacted by germline genetic variants in DPYD, which encodes the dihydropyrimidine dehydrogenase (DPD) enzyme that functions as the rate-limiting step in the catabolism of this drug class. Genetic testing to identify those with DPD deficiency can help mitigate the risk of severe and life-threatening fluoropyrimidine-induced toxicities. Globally, the integration of DPYD genetic testing into patient care has varied greatly, ranging from being required as the standard of care in some countries to limited clinical use in others. Thus, implementation strategies have evolved differently across health systems and countries. The primary objective of this tutorial is to provide practical considerations and best practice recommendations for the implementation of DPYD-guided systemic fluoropyrimidine dosing. We adapted the Exploration, Preparation, Implementation, and Sustainment (EPIS) framework to cover topics including the clinical evidence supporting DPYD genotyping to guide fluoropyrimidine therapy, regulatory guidance for DPYD genotyping, key stakeholder engagement, logistics for DPYD genotyping, development of point-of-care clinical decision support tools, and considerations for the creation of sustainable and scalable DPYD genotype-integrated workflows. This guide also describes approaches to counseling patients about DPYD testing and result disclosure, along with examples of patient and provider educational resources. Together, DPYD testing and clinical practice integration aim to promote safe prescribing of fluoropyrimidine therapy and decrease the risk of severe and life-threatening fluoropyrimidine toxicities.
Collapse
Affiliation(s)
- Teresa T. Ho
- Department of PathologyH. Lee Moffitt Cancer Center and Research InstituteTampaFloridaUSA
| | - D. Max Smith
- Department of OncologyGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
- MedStar HealthColumbiaMarylandUSA
| | - Christina L. Aquilante
- Department of Pharmaceutical SciencesUniversity of Colorado Skaggs School of Pharmacy and Pharmaceutical SciencesAuroraColoradoUSA
- Colorado Center for Personalized MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Emily J. Cicali
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Nihal El Rouby
- Department of Pharmacy Practice and Administrative Sciences, James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOhioUSA
- St Elizabeth Health CareEdgewoodKentuckyUSA
| | - Daniel L. Hertz
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Iman Imanirad
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research InstituteTampaFloridaUSA
| | - Jai N. Patel
- Division of Cancer Pharmacology & PharmacogenomicsAtrium Health Levine Cancer InstituteCharlotteNorth CarolinaUSA
- Atrium Health Wake Forest Baptist Comprehensive Cancer CenterWinston‐SalemNorth CarolinaUSA
| | - Stuart A. Scott
- Department of PathologyStanford UniversityStanfordCaliforniaUSA
| | - Sandra M. Swain
- MedStar HealthColumbiaMarylandUSA
- Georgetown Lombardi Comprehensive Cancer CenterWashingtonDistrict of ColumbiaUSA
| | - Sony Tuteja
- Division of Translational Medicine and Human Genetics, Department of MedicinePerelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - J. Kevin Hicks
- Department of PathologyH. Lee Moffitt Cancer Center and Research InstituteTampaFloridaUSA
| | | |
Collapse
|
2
|
Bianchino G, Perrone A, Sgambato A, Sarno I, Nozza F, Omer LC, Ulivi M, Traficante A, Campisi B, Russi S, Calice G, Falco G, Tartarone A. Application of dihydropyrimidine dehydrogenase deficiency testing for the prevention of fluoropyrimidine toxicity: a real-world experience in a Southern Italy cancer center. J Chemother 2025:1-7. [PMID: 40247645 DOI: 10.1080/1120009x.2025.2489837] [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: 11/14/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fluoropyrimidines (FPs) are antineoplastic agents used for the treatment of various solid tumors, especially gastrointestinal cancers. Patients with variations in dihydropyrimidine dehydrogenase gene (DPYD), which can determine the partial or complete deficiency of the dihydropyrimidine dehydrogenase enzyme (DPD), are at an increased risk of developing severe and potentially life-threatening toxicity. Worldwide the introduction of pharmacogenetic testing into clinical practice has been a slow process and in our center the analysis of the DPYD gene has been adopted since April 2020. We evaluated the clinical application of routine DPYD screening and its ability to prevent early-onset of fluoropyrimidine-related toxicity in patients treated at the Oncology Reference Center of Basilicata (IRCCS-CROB), a recognized cancer centre in Southern Italy. From April 2020 to November 2022, 300 patients (male 137; female 163) diagnosed with various types of cancer were subjected to DPYD genotyping, before starting treatment with FPs. In accordance with the current European Medicines Agency (EMA) and the Italian Association of Medical Oncology (AIOM) guidelines patients were tested for four DPYD variants that are associated with reduced DPD activity. FPs dose adjustments in DPYD variant carriers were made following the previously mentioned guidelines. Three hundred patients underwent DPYD testing and thirteen (4.3%) patients were found to be heterozygous variant carriers; ten out of thirteen patients received FP dose reduction as indicated by the guidelines, one out of thirteen patients received alternative treatment, two of the thirteen patients received no treatment at all. The main toxicities observed in patients who received a DPYD genotype-based dose reduction were anemia, neutropenia, nausea and mucositis but events were primarily grade 1 or 2. Our experience confirms the technical feasibility and the usefulness of DPYD genotyping to reduce the risk of severe FPs toxicities.
Collapse
Affiliation(s)
- Gabriella Bianchino
- Unit of Clinical Pathology IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Alessandra Perrone
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Italo Sarno
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Filomena Nozza
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Ludmila Carmen Omer
- Experimental, Oncology Unit, RCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Massimo Ulivi
- Research and Development team TIB molbiol Syntheselabor GmbH, Berlin, Germany
| | - Antonio Traficante
- Unit of Clinical Pathology IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Biagina Campisi
- Unit of Clinical Pathology IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Sabino Russi
- Laboratory of Preclinical and Translational Research, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Alfredo Tartarone
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| |
Collapse
|
3
|
Hansson P, Blacker C, Uvdal H, Wadelius M, Green H, Ljungman G. Pharmacogenomics in pediatric oncology patients with solid tumors related to chemotherapy-induced toxicity: a systematic review. Crit Rev Oncol Hematol 2025:104720. [PMID: 40222694 DOI: 10.1016/j.critrevonc.2025.104720] [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: 01/21/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025] Open
Abstract
Chemotherapy-induced toxicities remain challenging in pediatric oncology, affecting patient outcomes, hospital stays, and quality of life. Genetic variation can partly explain these toxicities, and pharmacogenomics could potentially optimize treatment. This review provides an overview of pharmacogenomic studies in relation to chemotherapy-induced toxicity in children with solid tumors. A systematic literature search was performed in PubMed, Embase, and Web of Science following PRISMA guidelines. Two independent reviewers assessed eligibility, risk of bias using ROBINS-I, and extracted data. Out of 9000 articles screened, 279 were deemed relevant, and 59 met the inclusion criteria by focusing on children with solid tumors and pharmacogenomics in relation to chemotherapy-induced toxicity. Following risk of bias assessment, 24 articles with low to moderate risk of bias were summarized. Identifying specific SNPs associated with toxicities proved challenging due to variability across studies. For methotrexate, the genes ABCC2, MTHFR, and SXR were associated with myelosuppression and hepatotoxicity. The genes ABCC3, COMT, ERCC2, GSTP1, GSTT1, LRP2, SLC22A2, and TPMT showed associations with ototoxicity due to platinum-based drugs. Anthracycline-induced cardiotoxicity was associated with CBR2, CELF4, GSTM1, HAS3, RARG, and SLC28A3, and further with HNMT and SLC22A2 in younger children, with ABCB4 in females, and with SULT2B1 in males. A dose-dependent effect of CELF4 on cardiotoxicity was noted with anthracycline doses over 300mg/m². This review highlights the complexity and variability of pharmacogenomic associations with chemotherapy-induced toxicities in pediatric oncology. While certain genetic variants show associations with specific toxicities, larger multinational/center studies are needed to strengthen the associations and improve clinical guidelines.
Collapse
Affiliation(s)
- Paula Hansson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| | - Christopher Blacker
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Hanna Uvdal
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mia Wadelius
- Department of Medical Sciences, Clinical Pharmacogenomics and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Henrik Green
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Gustaf Ljungman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Chao CJ, Gardner I, Lin CJ, Yeh KH, Lu WC, Abduljalil K, Ho YF. Administration mode matters for 5-fluorouracil therapy: Physiologically based pharmacokinetic evidence for avoidance of myelotoxicity by continuous infusion but not intravenous bolus. Br J Clin Pharmacol 2025; 91:1031-1040. [PMID: 38627941 DOI: 10.1111/bcp.16061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 02/07/2024] [Accepted: 03/10/2024] [Indexed: 04/13/2025] Open
Abstract
AIMS Pre-emptive prediction to avoid myelosuppression and harmful sequelae is difficult given the complex interplay among patients, drugs and treatment protocols. This study aimed to model plasma and bone marrow concentrations and the likelihood of myelotoxicity following administration of 5-fluorouracil (5-FU) by diverse intravenous (IV) bolus or continuous infusion (cIF) regimens. METHODS Using physicochemical, in vitro and clinical data obtained from the literature consisting of various regimens and patient cohorts, a 5-FU physiologically based pharmacokinetic (PBPK) model was developed. The predicted and observed PK values were compared to assess model performance prior to examining myelotoxicity potential of IV bolus vs. cIF and DPYD wild type vs. genetic variant. RESULTS The established model was verified by utilizing 5-FU concentration-time profiles of adequate heterogeneity contributed by 36 regimens from 15 studies. The study provided corroborative evidence to explain why cIF (vs. IV bolus) had lower myelotoxicity risk despite much higher total doses. The PBPK model was used to estimate the optimal dosage in patients heterozygous for the DPYD c.1905 + 1G > A allele and suggested that a dose reduction of at least 25% was needed (compared to the dose in wild-type subjects). CONCLUSION A verified PBPK model was used to explain the lower myelotoxicity risk of cIF vs. IV bolus administration of 5-FU and to estimate the dose reduction needed in carriers of a DPYD variant. With appropriate data, expertise and resources, PBPK models have many potential uses in precision medicine application of oncology drugs.
Collapse
Affiliation(s)
- Chih-Jia Chao
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Iain Gardner
- Simcyp Division, Certara UK Limited, Sheffield, UK
| | - Chun-Jung Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kun-Huei Yeh
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Chen Lu
- Department of Pharmacy, MacKay Memorial Hospital, Taipei, Taiwan
| | | | - Yunn-Fang Ho
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Gil-Rodríguez A, Recarey-Rama S, Rodríguez-Viyuela A, Barros F, Carracedo A, Maroñas O. Balance of care activity after EMA recommendation for DPYD gene testing in Galicia. Front Pharmacol 2025; 16:1523536. [PMID: 40223928 PMCID: PMC11985815 DOI: 10.3389/fphar.2025.1523536] [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: 11/06/2024] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Since April 2020, pretherapeutic screening for accessing the deficiency of the DPD enzyme by genotyping the dihydropyrimidine dehydrogenase gene (DPYD) is required by the European Medicine Agency (EMA) prior to the administration of fluoropyrimidine-based chemotherapy. In May 2020, the Spanish Drug and Medical Devices Agency (AEMPS) published an informative note highlighting the importance of DPYD analysis prior fluoropyrimidines derivatives administration to prevent the development of severe adverse drug reactions (ADRs). The publication of these recommendations marked a turning point in the daily routine in many pharmacogenetics laboratories in Spain. This article aims to illustrate the current state of the DPYD testing in the reference genomic medicine center in Galicia, 4 years after the EMA's updated recommendations. Methods The Pharmacogenetics Unit in the reference genomic medicine center conducted genotyping of the four DPYD variants recommended by regulatory agencies that oncologists can adjust fluoropyrimidine treatment based on DPYD genotype results. Results Between 1 June 2020 to 1 May 2024, both included, a total of 2,798 DPYD requests were analyzed. DPYD genotyping results revealed a 3.15% prevalence of heterozygosity for at least one of the four DPYD variants, being rs56038477 the most prevalent variant (1.31%). Conclusion This study addresses the importance of the DPYD analysis implementation in clinical practice after the changes in EMA and AEMPs recommendations which has led to a significant increase in DPYD genotyping requests. This highlights the significance of preemptive genotyping for accurately adjusting fluoropyrimidines doses before initiating treatment.
Collapse
Affiliation(s)
- Almudena Gil-Rodríguez
- Pharmacogenomics and drug discovery (GenDeM), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Sheila Recarey-Rama
- Pharmacogenomics and drug discovery (GenDeM), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Rodríguez-Viyuela
- Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Genetics group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Barros
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Galician Public Foundation of Genomic Medicine (FPGMX), Galician Healthcare Service (SERGAS), Santiago de Compostela, Spain
| | - Angel Carracedo
- Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Genetics group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Galician Public Foundation of Genomic Medicine (FPGMX), Galician Healthcare Service (SERGAS), Santiago de Compostela, Spain
| | - Olalla Maroñas
- Pharmacogenomics and drug discovery (GenDeM), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Galician Public Foundation of Genomic Medicine (FPGMX), Galician Healthcare Service (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
6
|
Sánchez-Bayona R, Catalán C, Cobos MA, Bergamino M. Pharmacogenomics in Solid Tumors: A Comprehensive Review of Genetic Variability and Its Clinical Implications. Cancers (Basel) 2025; 17:913. [PMID: 40149251 PMCID: PMC11939999 DOI: 10.3390/cancers17060913] [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: 01/14/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Pharmacogenomics, the study of how genetic variations influence drug response, has become integral to cancer treatment as personalized medicine evolves. This review aims to explore key pharmacogenomic biomarkers relevant to cancer therapy and their clinical implications, providing an updated and comprehensive perspective on how genetic variations impact drug metabolism, efficacy, and toxicity in oncology. Genetic heterogeneity among oncology patients significantly impacts drug efficacy and toxicity, emphasizing the importance of incorporating pharmacogenomic testing into clinical practice. Genes such as CYP2D6, DPYD, UGT1A1, TPMT, EGFR, KRAS, and BRCA1/2 play pivotal roles in influencing the metabolism, efficacy, and adverse effects of various chemotherapeutic agents, targeted therapies, and immunotherapies. For example, CYP2D6 polymorphisms affect tamoxifen metabolism in breast cancer, while DPYD variants can result in severe toxicities in patients receiving fluoropyrimidines. Mutations in EGFR and KRAS have significant implications for the use of targeted therapies in lung and colorectal cancers, respectively. Additionally, BRCA1/2 mutations predict the efficacy of PARP inhibitors in breast and ovarian cancer. Ongoing research in polygenic risk scores, liquid biopsies, gene-drug interaction networks, and immunogenomics promises to further refine pharmacogenomic applications, improving patient outcomes and reducing treatment-related adverse events. This review also discusses the challenges and future directions in pharmacogenomics, including the integration of computational models and CRISPR-based gene editing to better understand gene-drug interactions and resistance mechanisms. The clinical implementation of pharmacogenomics has the potential to optimize cancer treatment by tailoring therapies to an individual's genetic profile, ultimately enhancing therapeutic efficacy and minimizing toxicity.
Collapse
Affiliation(s)
| | - Camila Catalán
- Medical Oncology, Universidad Finis Terrae, Santiago 7501014, Chile;
| | - Maria Angeles Cobos
- Medical Oncology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Milana Bergamino
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain;
| |
Collapse
|
7
|
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
|
8
|
Kiiski JI, Khan S, Hosio A, Neuvonen M, Niemi M. Genetic and Functional Characterization of DPYD Exon 4 Deletion Common in the Finnish Population. Basic Clin Pharmacol Toxicol 2025; 136:e70011. [PMID: 39988348 DOI: 10.1111/bcpt.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Affiliation(s)
- Johanna I Kiiski
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Sofia Khan
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Antti Hosio
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Mikko Niemi
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
9
|
Abushanab D, Mohamed S, Abdel-Latif R, Moustafa DA, Marridi W, Elazzazy S, Badji R, Al-Muftah W, Ismail SI, Bujassoum S, Al-Thani A, Al-Badriyeh D, Al Hail M. Dihydropyrimidine Dehydrogenase Deficiency (DPYD) Genotyping-Guided Fluoropyrimidine-Based Adjuvant Chemotherapy for Breast Cancer. A Cost-Effectiveness Analysis. Clin Drug Investig 2025; 45:151-163. [PMID: 39885055 DOI: 10.1007/s40261-024-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND AND OBJECTIVE While standard doses of adjuvant fluoropyrimidine-based chemotherapies are generally safe for most patients, the risk of severe adverse drug reactions (ADRs) is increased for those with dihydropyrimidine dehydrogenase deficiency (DPYD), a genetic variation that affects drug metabolism. The objective of this study was to examine the cost effectiveness of offering DPYD pharmacogenetic-guided care, where genetic testing informs personalized dosing versus the current standard of care (SoC), which involves administering fluoropyrimidine-based therapies without prior genetic screening, for local or metastatic breast cancer patients in Qatar. METHODS We developed a two-stage decision analysis, with an analytic tree model over a 6-month period, followed by a life-table Markov model over a lifetime horizon. We compared the current SoC with the alternate strategy of DPYD genetic screening in patients living in Qatar with local or metastatic breast cancer who were eligible for adjuvant fluoropyrimidine therapy. Clinical outcomes and utilities were obtained from published studies, while healthcare costs were estimated from Hamad Medical Corporation, Qatar. The short-term outcome included the incremental cost-effectiveness ratio (ICER), defined as cost per success (survival without grade III/IV ADRs) at 6 months. The long-term outcome was the ICER, defined as cost per quality-adjusted life year (QALY) gained, with a 3% annual discount rate. The study adopted a public healthcare perspective in Qatar. Sensitivity analyses were conducted to explore the impact of key input parameters on the robustness of the model. RESULTS In the short-term model, at its base case, DPYD genomic screening was dominant over SoC with a mean cost-saving of QAR84,585 (95% confidence interval [CI], 45,270-151,657). This cost saving reflects the overall economic benefits associated with the implementation of DPYD genomic screening. In the long-term model, compared to the current SoC, DPYD genetic screening would result in an ICER of QAR21,107 (95% CI -59,382-145,664) per QALY gained. CONCLUSION Based on our model, implementing DPYD genetic screening to detect DPYD mutations in breast cancer patients before therapy initiation seems to be a cost-saving and cost-effective strategy in Qatar.
Collapse
Affiliation(s)
- Dina Abushanab
- Pharmacy Department, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Shaban Mohamed
- Pharmacy Department, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Rania Abdel-Latif
- Qatar Genome Program, Qatar Precision Health Institute, Qatar Foundation, Doha, 34173, Qatar
| | | | - Wafa Marridi
- College of Medicine, QU Health, Qatar University, Doha, 2713, Qatar
| | - Shereen Elazzazy
- College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
- Pharmacy Department, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Radja Badji
- Qatar Genome Program, Qatar Precision Health Institute, Qatar Foundation, Doha, 34173, Qatar
| | - Wadha Al-Muftah
- Qatar Genome Program, Qatar Precision Health Institute, Qatar Foundation, Doha, 34173, Qatar
| | - Said I Ismail
- Qatar Genome Program, Qatar Precision Health Institute, Qatar Foundation, Doha, 34173, Qatar
| | - Salha Bujassoum
- Medical Oncology, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Asma Al-Thani
- Biomedical Research Center, QU Health, Qatar University, Doha, 2713, Qatar
| | - Daoud Al-Badriyeh
- College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar.
| | - Moza Al Hail
- Pharmacy Department, Hamad Medical Corporation, Doha, 3050, Qatar
| |
Collapse
|
10
|
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] [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
|
11
|
Thompson JT, Wood DM, Dargan PI. Review of the fluoropyrimidine antidote uridine triacetate. Br J Clin Pharmacol 2025; 91:615-627. [PMID: 39468799 DOI: 10.1111/bcp.16319] [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: 05/13/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
In 2015, the United States Food and Drug Administration (FDA) approved uridine triacetate to treat overdose and severe toxicity of the fluoropyrimidine chemotherapy agents 5-fluorouracil (5-FU) and its oral prodrug capecitabine. Uridine triacetate is as an oral prodrug of uridine that competes with cytotoxic fluoropyrimidine metabolites for incorporation into nucleotides. Two million people worldwide start fluoropyrimidine chemotherapy each year, with 20-30% developing severe or life-threatening adverse effects, often attributable to a genetic predisposition such as dihydropyrimidine dehydrogenase deficiency. Whilst genetic prescreening is recommended prior to starting fluoropyrimidine agents, this only prevents 20-30% of early-onset life-threatening toxicity and so does not obviate the need for an antidote. Initial in-human studies established that uridine triacetate more than doubles the maximum tolerated weekly 5-FU bolus dose. A lack of clinical equipoise meant a placebo-controlled phase III trial was not ethical and so the phase III trials used historical controls. These found that uridine triacetate improved survival in those with fluoropyrimidine overdose and severe toxicity from 16% to 94%, with 34% able to resume chemotherapy within 30 days. Five case reports of delayed fluoropyrimidine toxicity demonstrate improvement following uridine triacetate treatment 120-504 h after last fluoropyrimidine administration, suggesting efficacy beyond the FDA licencing indications. Mechanistically uridine triacetate would be expected to be effective for overdose and severe toxicity of tegafur (a 5-FU prodrug), but there are no published case reports describing this. Uridine triacetate is available internationally through an expanded access scheme and has been available in the UK since 2019 on a named patient basis.
Collapse
Affiliation(s)
- Jack T Thompson
- Clinical Toxicology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - David M Wood
- Clinical Toxicology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Paul I Dargan
- Clinical Toxicology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
12
|
Bedrikovetski S, Murshed I, Fitzsimmons T, Traeger L, Price TJ, Penniment M, Selva‐Nayagam S, Vather R, Sammour T. Total neoadjuvant therapy in early-onset rectal cancer: A multicentre prospective cohort study. Colorectal Dis 2025; 27:e70059. [PMID: 40123409 PMCID: PMC11931348 DOI: 10.1111/codi.70059] [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: 11/05/2024] [Revised: 01/21/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
AIM The incidence of early-onset (age <50 years) rectal cancer (EORC) is rising globally, often presenting at an advanced stage. Total neoadjuvant therapy (TNT) is increasingly utilised in the management of advanced rectal cancers due to improved response and survival rates. However, it remains unclear whether EORC in an unscreened population responds similarly to TNT compared to average or late-onset (age ≥50 years) rectal cancer (AORC). METHOD This study included consecutive patients treated with curative intent with TNT for rectal cancer at three South Australian hospitals between 2019 and 2024. Patients were divided into EORC and AORC cohorts. The primary outcome was overall complete response (oCR) rate, defined as the proportion of patients who achieved a clinical complete response (cCR) and/or pathological complete response (pCR). Secondary outcomes included compliance and treatment-related toxicity. RESULTS Of 202 eligible patients, 48 (23.8%) were in the EORC cohort and 154 (76.2%) in the AORC cohort. No significant difference in oCR rate was observed between EORC and AORC patients (43.8% vs. 37.9%, P = 0.470). cCR, pCR and complete M1 response rates were also similar between the two groups. EORC patients experienced significantly less Grade 3-4 chemotherapy-induced toxicity compared to AORC patients (2.1% vs. 25.3%, P < 0.001), but reported higher rates of patient-reported Grade 3-4 radiotherapy-induced toxicity than AORC patients (31.3% vs. 12.3%, P = 0.004). CONCLUSION EORC patients exhibit comparable overall tumour response rates to AORC patients treated with TNT. However, toxicity profiles differ, with EORC patients experiencing less chemotherapy-induced toxicity but more patient-reported radiation-induced toxicity.
Collapse
Affiliation(s)
- Sergei Bedrikovetski
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Ishraq Murshed
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Tracy Fitzsimmons
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Luke Traeger
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Timothy J. Price
- Department of Medical OncologyQueen Elizabeth HospitalAdelaideSouth AustraliaAustralia
| | - Michael Penniment
- Department of Radiation OncologyRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | | | - Ryash Vather
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
- Centre for Cancer BiologyUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Tarik Sammour
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Colorectal Unit, Department of SurgeryRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| |
Collapse
|
13
|
Leong IU, Cabrera CP, Cipriani V, Ross PJ, Turner RM, Stuckey A, Sanghvi S, Pasko D, Moutsianas L, Odhams CA, Elgar GS, Chan G, Giess A, Walker S, Foulger RE, Williams EM, Daugherty LC, Rueda-Martin A, Rhodes DJ, Niblock O, Pickard A, Marks L, Leigh SE, Welland MJ, Bleda M, Snow C, Deans Z, Murugaesu N, Scott RH, Barnes MR, Brown MA, Rendon A, Hill S, Sosinsky A, Caulfield MJ, McDonagh EM. Large-Scale Pharmacogenomics Analysis of Patients With Cancer Within the 100,000 Genomes Project Combining Whole-Genome Sequencing and Medical Records to Inform Clinical Practice. J Clin Oncol 2025; 43:682-693. [PMID: 39481076 PMCID: PMC11825504 DOI: 10.1200/jco.23.02761] [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: 12/22/2023] [Revised: 05/10/2024] [Accepted: 09/03/2024] [Indexed: 11/02/2024] Open
Abstract
PURPOSE As part of the 100,000 Genomes Project, we set out to assess the potential viability and clinical impact of reporting genetic variants associated with drug-induced toxicity for patients with cancer recruited for whole-genome sequencing (WGS) as part of a genomic medicine service. METHODS Germline WGS from 76,805 participants was analyzed for pharmacogenetic (PGx) variants in four genes (DPYD, NUDT15, TPMT, UGT1A1) associated with toxicity induced by five drugs used in cancer treatment (capecitabine, fluorouracil, mercaptopurine, thioguanine, irinotecan). Linking genomic data with prescribing and hospital incidence records, a phenome-wide association study (PheWAS) was performed to identify whether phenotypes indicative of adverse drug reactions (ADRs) were enriched in drug-exposed individuals with the relevant PGx variants. In a subset of 7,081 patients with cancer, DPYD variants were reported back to clinicians and outcomes were collected. RESULTS We identified clinically relevant PGx variants across the four genes in 62.7% of participants in our cohort. Extending this to annual prescription numbers in England for the drugs affected by these PGx variants, approximately 14,540 patients per year could potentially benefit from a reduced dose or alternative drug to reduce the risk of ADRs. Validating PGx associations in a real-world data set, we found a significant association between PGx variants in DPYD and toxicity-related phenotypes in patients treated with capecitabine or fluorouracil. Reported DPYD variants were deemed informative for clinical decision making in a majority of cases. CONCLUSION Reporting PGx variants from germline WGS relevant to patients with cancer alongside primary findings related to their cancer can be clinically informative, informing prescribing to reduce the risk of ADRs. Extending the range of actionable variants to those found in patients of non-European ancestry is important and will extend the potential clinical impact.
Collapse
Affiliation(s)
- Ivone U.S. Leong
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Claudia P. Cabrera
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Valentina Cipriani
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Paul J. Ross
- Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Richard M. Turner
- Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- GSK, Stevenage, Hertfordshire, United Kingdom
| | - Alex Stuckey
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Sonali Sanghvi
- Integrating Pharmacy & Medicines Optimisation Team, NHS North Central London Integrated Care System, UCLH NHS Foundation Trust, London
| | - Dorota Pasko
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Loukas Moutsianas
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | | | - Greg S. Elgar
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Georgia Chan
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Adam Giess
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Susan Walker
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Rebecca E. Foulger
- SciBite Limited, BioData Innovation Centre, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | | | | | | | - Lauren Marks
- NHS England and NHS Improvement, London, United Kingdom
| | - Sarah E.A. Leigh
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Matthew J. Welland
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Marta Bleda
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Catherine Snow
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Zandra Deans
- NHS England and NHS Improvement, London, United Kingdom
- GenQA, Laboratory Medicine, NHS Lothian NINE, Edinburgh, United Kingdom
| | - Nirupa Murugaesu
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
- Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Richard H. Scott
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Michael R. Barnes
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Matthew A. Brown
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Augusto Rendon
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Sue Hill
- NHS England and NHS Improvement, London, United Kingdom
| | - Alona Sosinsky
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Mark J. Caulfield
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- Faculty of Medicine and Dentistry, VP Health Office, Queen Mary University of London, London, United Kingdom
| | - Ellen M. McDonagh
- Open Targets, Wellcome Genome Campus, Hinxton, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| |
Collapse
|
14
|
Aberle MR, Coolsen MME, Wenmaekers G, Volmer L, Brecheisen R, van Dijk D, Wee L, Van Dam RM, de Vos-Geelen J, Rensen SS, Damink SWMO. Skeletal muscle is independently associated with grade 3-4 toxicity in advanced stage pancreatic ductal adenocarcinoma patients receiving chemotherapy. Clin Nutr ESPEN 2025; 65:134-143. [PMID: 39577693 DOI: 10.1016/j.clnesp.2024.11.004] [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: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Patients with advanced-stage pancreatic ductal adenocarcinoma (PDAC) are regularly treated with FOLFIRINOX, a chemotherapy regimen based on 5-fluorouracil, irinotecan and oxaliplatin, which is associated with high toxicity. Dosing of FOLFIRINOX is based on body surface area, risking under- or overdosing caused by altered pharmacokinetics due to interindividual differences in body composition. This study aimed to investigate the relationship between body composition and treatment toxicity in advanced stage PDAC patients treated with FOLFIRINOX. METHODS Data from patients treated at the Maastricht University Medical Centre + between 2012 and 2020 were collected retrospectively (n = 65). Skeletal muscle-, visceral adipose tissue, subcutaneous adipose tissue-, (SM-Index, VAT-Index, SAT-Index resp.) and Skeletal Muscle Radiation Attenuation (SM-RA) were calculated after segmentation of computed tomography (CT) images at the third lumbar level using a validated deep learning method. Lean body mass (LBM) was estimated using SM-Index. Toxicities were scored and grade 3-4 adverse events were considered dose-limiting toxicities (DLTs). RESULTS Sixty-seven DLTs were reported during the median follow-up of 51.4 (95%CI 39.2-63.7) weeks. Patients who experienced at least one DLT had significantly higher dose intensity per LBM for all separate cytotoxics of FOLFIRINOX. Independent prognostic factors for the number of DLTs per cycle were: sarcopenia (β = 0.292; 95%CI 0.013 to 0.065; p = 0.013), SM-Index change (% per 30 days, β = -0.045; 95%CI -0.079 to -0.011; p = 0.011), VAT-Index change (% per 30 days, β = -0.006; 95%CI -0.012 to 0.000; p = 0.040) between diagnosis and the first follow-up CT scan, and cumulative relative dose intensity >80 % (β = -0.315; 95 % CI -0.543 to -0.087; p = 0.008). CONCLUSION Sarcopenia and early muscle and fat wasting during FOLFIRINOX treatment were associated with treatment-related toxicity, warranting exploration of body composition guided personalized dosing of chemotherapeutics to limit DLTs.
Collapse
Affiliation(s)
- Merel R Aberle
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Mariëlle M E Coolsen
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| | - Gilles Wenmaekers
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Leroy Volmer
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Radiotherapy (MAASTRO), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Ralph Brecheisen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - David van Dijk
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Leonard Wee
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Radiotherapy (MAASTRO), Maastricht University Medical Centre+, Maastricht, the Netherlands; Clinical Data Science, Maastricht University, Maastricht, the Netherlands
| | - Ronald M Van Dam
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| | - Judith de Vos-Geelen
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Medical Oncology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Sander S Rensen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.
| | - Steven W M Olde Damink
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| |
Collapse
|
15
|
Keen J, McDermott JH, Aguilar-Martinez E, Newman WG. Pharmacogenomics: DPYD and Prevention of Toxicity. Clin Oncol (R Coll Radiol) 2025; 38:103706. [PMID: 39721301 DOI: 10.1016/j.clon.2024.103706] [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: 10/08/2023] [Revised: 10/10/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024]
Abstract
In 2020, the introduction of pre-emptive DPYD genotyping prior to the administration of systemic fluoropyrimidine-based chemotherapy represented one of the first widespread pharmacogenetic testing programmes to be applied nationally in the United Kingdom. Pharmacogenetic variants in the DPYD gene found in between 3 and 6% of the population are a recognised cause of primary DPD enzyme deficiency and associated increased risk of severe fluoropyrimidine toxicity [1]. Yet, the availability of testing globally is heterogeneous. Despite growing evidence that in addition to reducing drug-induced toxicity, DPYD-guided dosing does not negatively affect outcomes, further research on the impact of routine DPYD genotyping in the UK population is required. With mandatory testing in the UK focussed on four well-characterised variants, there is a need to address the applicability of this strategy across diverse ethnic or ancestral populations. We highlight approaches to identify and characterise rare variants in DPYD and in other genes involved in the pyrimidine metabolic pathway to reduce healthcare inequalities. Finally, we discuss the future of pharmacogenomics within cancer care, and the potential to harness innovative digital and genotyping technologies to streamline prescribing and optimise both systemic anti-cancer therapies and supportive care.
Collapse
Affiliation(s)
- J Keen
- NHS North West Genomic Medicine Service Alliance, UK.
| | - J H McDermott
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK; The Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - E Aguilar-Martinez
- The Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - W G Newman
- NHS North West Genomic Medicine Service Alliance, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK; The Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
16
|
Donnelly RS, Ayoubieh H, Kudron EL, Mills R, Weiler T, Empey PE. When Patients Present With Direct-to-Consumer Pharmacogenomic Test Results. Mayo Clin Proc 2025; 100:197-203. [PMID: 39818680 DOI: 10.1016/j.mayocp.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 09/11/2024] [Accepted: 10/25/2024] [Indexed: 01/18/2025]
Affiliation(s)
- Roseann S Donnelly
- Department of Pharmacy Practice, Massachusetts College of Pharmacy and Health Sciences, Boston, MA.
| | - Houriya Ayoubieh
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX
| | - Elizabeth L Kudron
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Rachel Mills
- Genetic Counseling Program, The University of North Carolina at Greensboro, Greensboro, NC
| | - Tracey Weiler
- Department of Medical Education, Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| |
Collapse
|
17
|
Peruzzi E, Roncato R, De Mattia E, Bignucolo A, Swen JJ, Guchelaar H, Toffoli G, Cecchin E. Implementation of pre-emptive testing of a pharmacogenomic panel in clinical practice: Where do we stand? Br J Clin Pharmacol 2025; 91:270-282. [PMID: 37926674 PMCID: PMC11773130 DOI: 10.1111/bcp.15956] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Adverse drug reactions (ADRs) account for a large proportion of hospitalizations among adults and are more common in multimorbid patients, worsening clinical outcomes and burdening healthcare resources. Over the past decade, pharmacogenomics has been developed as a practical tool for optimizing treatment outcomes by mitigating the risk of ADRs. Some single-gene reactive tests are already used in clinical practice, including the DPYD test for fluoropyrimidines, which demonstrates how integrating pharmacogenomic data into routine care can improve patient safety in a cost-effective manner. The evolution from reactive single-gene testing to comprehensive pre-emptive genotyping panels holds great potential for refining drug prescribing practices. Several implementation projects have been conducted to test the feasibility of applying different genetic panels in clinical practice. Recently, the results of a large prospective randomized trial in Europe (the PREPARE study by Ubiquitous Pharmacogenomics consortium) have provided the first evidence that prospective application of a pre-emptive pharmacogenomic test panel in clinical practice, in seven European healthcare systems, is feasible and yielded a 30% reduction in the risk of developing clinically relevant toxicities. Nevertheless, some important questions remain unanswered and will hopefully be addressed by future dedicated studies. These issues include the cost-effectiveness of applying a pre-emptive genotyping panel, the role of multiple co-medications, the transferability of currently tested pharmacogenetic guidelines among patients of non-European origin and the impact of rare pharmacogenetic variants that are not detected by currently used genotyping approaches.
Collapse
Affiliation(s)
- Elena Peruzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
| | - Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
- Department of MedicineUniversity of UdineUdineItaly
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
| | - Jesse J. Swen
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | - Henk‐Jan Guchelaar
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere ScientificoAvianoItaly
| |
Collapse
|
18
|
Ranasinghe P, Jeyapragasam H, Sirisena N, Bhagya Hendalage DP, Dissanayake VHW. Pharmacogenomic profiling of variants affecting efficacy and toxicity of anti-infective medicines in a south Asian population from Sri Lanka. BMC Infect Dis 2025; 25:153. [PMID: 39893405 PMCID: PMC11786550 DOI: 10.1186/s12879-025-10538-w] [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: 09/20/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Anti-infective medicines are crucial for treating infections, but improper dosing can cause toxicity, resistance and treatment failure. Pharmacogenomics can address genetic variations affecting drug efficacy and safety. Despite the high burden of diseases like TB and HIV in Sri Lanka and South Asia, pharmacogenomic data for these populations are limited. This study aims to fill this gap by investigating pharmacogenomic variants in a South Asian population from Sri Lankan. METHODS Pharmacogenomic data on anti-infective medicines were obtained from the PharmGKB database, selecting variants with evidence levels 1 A, 1B, 2 A, and 2B. Sri Lankan genetic data were sourced from an anonymized database of 670 Sri Lankans maintained by the Centre for Genetics and Genomics, Faculty of Medicine, University of Colombo. MAFs were compared between Sri Lankan sub-populations and global data from gnomAD, with statistical significance set at p < 0.05. RESULTS MAFs of NAT2 gene rs1041983 and rs1799931 variants were, 43.7% (95%CI:41.1-46.4), 7.3% (95%CI:6.0-8.8), respectively. The UGT1A1 rs4148323 variant had a MAF of 3.5% (95%CI:2.6-4.6). In the CYP2B6 gene, 109 individuals were homozygous for the rs3745274 (poor metaboliser) variant, with a MAF of 39.6% (95%CI:37.0-42.3), while the rs34097093 and rs28399499 variants had no individuals homozygous for the variant (MAF: 0.2% [95%CI:0-0.5] (poor/intermediate metaboliser), and 0.1% [95%CI:0-0.4] (poor/intermediate metaboliser), respectively). The MAFs of the CYP2C19 rs12769205 (poor/intermediate metaboliser), rs4244285 (poor/intermediate metaboliser), rs3758581 (poor/intermediate metaboliser), and rs4986893 (poor/intermediate metaboliser) variants were 41.9% (95%CI:39.3-44.6), 41.9% (95%CI:39.2-44.7), 9.7% (95%CI:8.2-11.4), and 0.5% [(95%CI:0.2-1.1), respectively. Most variants showed significant differences compared to global populations, with some exhibiting higher frequencies, particularly when compared to Europeans. CYP2C19 rs12769205 and rs4244285 exhibited higher MAFs in Sri Lankans compared to both other South Asians and Europeans. The NAT2 rs1041983, NAT2 rs1799931, CYP2C19 rs4986893, CYP2C19 rs3758581, and CYP2B6 rs3745274 variants demonstrated significantly higher MAFs than in Europeans but not significantly different from South Asians. CONCLUSION This preliminary study identifies variants in NAT2, UGT1A1, CYP2B6, and CYP2C19 genes relevant to the metabolism of anti-TB drugs, antiretrovirals, and voriconazole among Sri Lankans. Several variants, including CYP2C19 rs12769205 and rs4244285, showed higher MAFs, particularly in comparison to European populations, indicating potential differences in drug response. However, the nature of the study limits the ability to explore clinical correlations with the genotypes, therefore further research focusing on clinical correlation and functional validation is required.
Collapse
Affiliation(s)
- Priyanga Ranasinghe
- Department of Pharmacology, Faculty of Medicine, University of Colombo, Colombo, 08, Sri Lanka.
| | - Hajanthy Jeyapragasam
- Department of Pharmacology, Faculty of Medicine, University of Colombo, Colombo, 08, Sri Lanka
| | - Nirmala Sirisena
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, 08, Sri Lanka
| | - D P Bhagya Hendalage
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, 08, Sri Lanka
| | - Vajira H W Dissanayake
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, 08, Sri Lanka
| |
Collapse
|
19
|
Surprenant K, Murray L, Merritt K, Hopkins M, McIntyre J. Give Patients the Choice to Test for DPD Deficiency Before Fluoropyrimidine Chemotherapy. JCO Oncol Pract 2025; 21:264-265. [PMID: 39626133 DOI: 10.1200/op.24.00443] [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: 06/01/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 02/13/2025] Open
Abstract
Patients, not physicians, should decide if DPYD screening is right for them before FU/Xeloda chemotherapy.
Collapse
Affiliation(s)
- Ken Surprenant
- Ken Surprenant, MA, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; Lindsay Murray, HS, Advocates for Universal DPD/DPYD Testing, Andover, MA; Karen Merritt, BS, Advocates for Universal DPD/DPYD Testing, Chelan, WA; Maria Hopkins, BS, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; and Joanne McIntyre, AA, Advocates for Universal DPD/DPYD Testing, Portland, OR
| | - Lindsay Murray
- Ken Surprenant, MA, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; Lindsay Murray, HS, Advocates for Universal DPD/DPYD Testing, Andover, MA; Karen Merritt, BS, Advocates for Universal DPD/DPYD Testing, Chelan, WA; Maria Hopkins, BS, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; and Joanne McIntyre, AA, Advocates for Universal DPD/DPYD Testing, Portland, OR
| | - Karen Merritt
- Ken Surprenant, MA, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; Lindsay Murray, HS, Advocates for Universal DPD/DPYD Testing, Andover, MA; Karen Merritt, BS, Advocates for Universal DPD/DPYD Testing, Chelan, WA; Maria Hopkins, BS, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; and Joanne McIntyre, AA, Advocates for Universal DPD/DPYD Testing, Portland, OR
| | - Maria Hopkins
- Ken Surprenant, MA, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; Lindsay Murray, HS, Advocates for Universal DPD/DPYD Testing, Andover, MA; Karen Merritt, BS, Advocates for Universal DPD/DPYD Testing, Chelan, WA; Maria Hopkins, BS, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; and Joanne McIntyre, AA, Advocates for Universal DPD/DPYD Testing, Portland, OR
| | - Joanne McIntyre
- Ken Surprenant, MA, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; Lindsay Murray, HS, Advocates for Universal DPD/DPYD Testing, Andover, MA; Karen Merritt, BS, Advocates for Universal DPD/DPYD Testing, Chelan, WA; Maria Hopkins, BS, Advocates for Universal DPD/DPYD Testing, Battle Creek, MI; and Joanne McIntyre, AA, Advocates for Universal DPD/DPYD Testing, Portland, OR
| |
Collapse
|
20
|
Shamaei Zadeh A, Roberts D, Williams A, Pandey D, Villano JL. Dihydropyrimidine enzyme activity and its effect on chemotherapy toxicity: importance of genetic testing. Cancer Chemother Pharmacol 2025; 95:26. [PMID: 39825952 DOI: 10.1007/s00280-024-04740-x] [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: 09/23/2024] [Accepted: 12/08/2024] [Indexed: 01/30/2025]
Abstract
PURPOSE Patients with partial or complete DPD deficiency have decreased capacity to degrade fluorouracil and are at risk of developing toxicity, which can be even life-threatening. CASE A 43-year-old man with moderately differentiated rectal adenocarcinoma on capecitabine presented to the emergency department with complaints of nausea, vomiting, diarrhea, weakness, and lower abdominal pain for several days. Laboratory findings include grade 4 neutropenia (ANC 10) and thrombocytopenia (platelets 36,000). Capecitabine is used as a component of first-line adjuvant therapy by approximately 2 million patients worldwide each year. Capecitabine is metabolized to fluorouracil via the enzyme dihydropyrimidine dehydrogenase (DPD). With worsening pancytopenia and diarrhea, genetic testing for DPD deficiency was sent. Prompt treatment with uridine triacetate was initiated for presumed DPD deficiency. Unfortunately, he passed away from an infectious complication and was later confirmed to have a heterozygous DPYD*2A mutation. DISCUSSION Our case demonstrates uneven testing guidelines for DPD prior to initiating 5-FU chemotherapy, appropriateness of treating with uridine triacetate, and analysis of next-generation sequencing (NGS) on tumor samples and co-incidentally obtaining germline DPD deficiency status. Our case also highlights the severe clinical impact of having DPD deficiency even with early uridine triacetate therapy. CONCLUSION It is our recommendation to perform DPD deficiency in curative intent cancer treatment and this information can increasingly be obtained in standard tumor NGS profiling, a growing norm in medical oncology.
Collapse
Affiliation(s)
| | | | - Abby Williams
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Pharmacy Services, University of Kentucky, Lexington, KY, USA
| | - Deepali Pandey
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - John L Villano
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
- Division of Medical Oncology, University of Kentucky, Lexington, KY, USA.
- Department of Neurology, University of Kentucky, Lexington, KY, USA.
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
21
|
D'Amato M, Iengo G, Massa N, Carlomagno C. Dihydropyrimidine dehydrogenase polymorphisms in patients with gastrointestinal malignancies and their impact on fluoropyrimidine tolerability: Experience from a single Italian institution. World J Gastrointest Oncol 2025; 17:96822. [PMID: 39817118 PMCID: PMC11664602 DOI: 10.4251/wjgo.v17.i1.96822] [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: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Fluoropyrimidines are metabolized in the liver by the enzyme dihydropyrimidine dehydrogenase (DPD), encoded by the DPYD gene. About 7% of the European population is a carrier of DPYD gene polymorphisms associated with reduced DPD enzyme activity. AIM To assess the prevalence of DPYD polymorphisms and their impact on fluoropyrimidine tolerability in Italian patients with gastrointestinal malignancies. METHODS A total of 300 consecutive patients with a diagnosis of gastrointestinal malignancy and treated with a fluoropyrimidine-based regimen were included in the analysis and divided into two cohorts: (1) 149 patients who started fluoropyrimidines after DPYD testing; and (2) 151 patients treated without DPYD testing. Among the patients in cohort A, 15% tested only the DPYD2A polymorphism, 19% tested four polymorphisms (DPYD2A, HapB3, c.2846A>T, and DPYD13), and 66% tested five polymorphisms including DPYD6. RESULTS Overall, 14.8% of patients were found to be carriers of a DPYD variant, the most common being DPYD6 (12.1%). Patients in cohort A reported ≥ G3 toxicities (P = 0.00098), particularly fewer nonhematological toxicities (P = 0.0028) compared with cohort B, whereas there was no statistically significant difference between the two cohorts in hematological toxicities (P = 0.6944). Significantly fewer chemotherapy dose reductions (P = 0.00002) were observed in cohort A compared to cohort B, whereas there was no statistically significant differences in chemotherapy delay. CONCLUSION Although this study had a limited sample size, it provides additional information on the prevalence of DPYD polymorphisms in the Italian population and highlights the role of pharmacogenetic testing to prevent severe toxicity.
Collapse
Affiliation(s)
- Mariarosaria D'Amato
- Department of Oncology, Ospedale San Rocco ASL Caserta, Sessa Aurunca 81037, Campania, Italy
| | - Gennaro Iengo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Campania, Italy
| | - Nicola Massa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Campania, Italy
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Campania, Italy
| |
Collapse
|
22
|
Yuan M, Zheng Y, Wang F, Bai N, Zhang H, Bian Y, Liu H, He X. Discussion on the optimization of personalized medication using information systems based on pharmacogenomics: an example using colorectal cancer. Front Pharmacol 2025; 15:1516469. [PMID: 39877392 PMCID: PMC11772163 DOI: 10.3389/fphar.2024.1516469] [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: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025] Open
Abstract
Pharmacogenomics (PGx) is a powerful tool for clinical optimization of drug efficacy and safety. However, due to many factors affecting drugs in the real world, PGx still accounts for a small proportion of actual clinical application scenarios. Therefore, based on the information software, pharmacists use their professional advantages to integrate PGx into all aspects of pharmaceutical care, which is conducive to promoting the development of personalized medicine. In this paper, the establishment of an information software platform is summarized for the optimization of a personalized medication program based on PGx. Taking colorectal cancers (CRC) as an example, this paper also discusses the role of PGx in different working modes and participation in drug management of CRC patients by pharmacists with the help of information systems. Finally, we summarized the recommendations of different PGx guidelines to provide reference for the follow-up personalized pharmaceutical care.
Collapse
Affiliation(s)
- Mengying Yuan
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuankun Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Wang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Niuniu Bai
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Haoling Zhang
- Department of Pharmacy, Yuncheng Central Hospital, Yuncheng, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia He
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
23
|
Brlek P, Bulić L, Mršić L, Sokač M, Brenner E, Matišić V, Skelin A, Bach-Rojecky L, Primorac D. Exploring the Pharmacogenomic Map of Croatia: PGx Clustering of 522-Patient Cohort Based on UMAP + HDBSCAN Algorithm. Int J Mol Sci 2025; 26:589. [PMID: 39859305 PMCID: PMC11765990 DOI: 10.3390/ijms26020589] [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: 12/19/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Pharmacogenetics is a branch of genomic medicine aiming to personalize drug prescription guidelines based on individual genetic information. This concept might lead to a reduction in adverse drug reactions, which place a heavy burden on individual patients' health and the economy of the healthcare system. The aim of this study was to present insights gained from the pharmacogenetics-based clustering of over 500 patients from the Croatian population. The data used in this article were obtained by the pharmacogenetic testing of 522 patients from the Croatian population. The patients were clustered based on the genotypes of 28 pharmacologically relevant genes. Dimensionality reduction was employed using the UMAP algorithm, after which clusters were defined using HDBSCAN. Validation of clustering was performed by decision tree analysis and predictive modeling using the RandomForest, XGBoost, and ExtraTrees classification algorithms. The clustering algorithm defined six clusters of patients based on two UMAP components (silhouette score = 0.782). Decision tree analysis demonstrated CYP2D6 and SLCO1B1 genotypes as the main points of cluster determination. Predictive modeling demonstrated an excellent ability to discern the cluster of each patient based on all genes (avg. ROC-AUC = 0.998), CYP2D6 and SLCO1B1 (avg. ROC-AUC = 1.000), and CYP2D6 alone (avg. ROC-AUC = 0.910). Membership in each cluster provided clinically relevant information, in the context of ruling out certain favorable or unfavorable phenotypes. However, this study's main limitation is its cohort size. Through further research and investigation of a larger number of patients, more accurate and clinically applicable associations between pharmacogenetic genotypes and phenotypes might be discovered.
Collapse
Affiliation(s)
- Petar Brlek
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Science, Department of Molecular Biology, University of Zagreb, 10000 Zagreb, Croatia
| | - Luka Bulić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
| | - Leo Mršić
- Department of Information Systems and Business Analytics, Algebra University, 10000 Zagreb, Croatia
| | - Mateo Sokač
- Department of Information Systems and Business Analytics, Algebra University, 10000 Zagreb, Croatia
| | - Eva Brenner
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
| | - Vid Matišić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
| | - Andrea Skelin
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Lidija Bach-Rojecky
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Eberly College of Science, The Pennsylvania State University, State College, PA 16802, USA
- School of Medicine, University of Split, 21000 Split, Croatia
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, New Haven, CT 06516, USA
- Regiomed Kliniken, 96450 Coburg, Germany
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- National Forensic Sciences University, Gandhinagar 382007, India
| |
Collapse
|
24
|
Vilquin P, Medard Y, Thomas F, Goldwirt L, Teixeira L, Mourah S, Jacqz-Aigrain E. DPYD genotype should be extended to rare variants: report on two cases of phenotype / genotype discrepancy. Cancer Chemother Pharmacol 2025; 95:16. [PMID: 39745516 DOI: 10.1007/s00280-024-04738-5] [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: 07/22/2024] [Accepted: 12/08/2024] [Indexed: 04/04/2025]
Abstract
The enzyme dihydropyrimidine dehydrogenase (DPD) is the primary catabolic pathway of fluoropyrimidines including 5 fluorouracil (5FU) and capecitabine. Cases of lethal toxicity have been reported in cancer patients with complete DPD deficiency receiving standard dose of 5FU or capecitabine. DPD is encoded by the pharmacogene DPYD in which more than 200 variants have been identified. Different approaches have been developed for screening DPD-deficiency, including DPYD genotyping and phenotyping. Plasma uracil ([U]) and dihydrouracil ([UH2]) concentrations are routinely used as surrogate markers for systemic DPD activity: [U] ≥ 16 ng/ml and < 150 ng/ml, and [U] ≥ 150 ng/mL indicate partial and complete DPD deficient phenotype, respectively, while values of 5 or 10 for [UH2]/([U] ratio are often cited. Four clinically relevant DPYD defective variants (DPYD*13, DPYD*2A, p.Asp949Val and haplotype B3), are targeted in genetic testing via PCR. In practice, pretreatment [U], alone or combined with these 4 recommended DPYD alleles guides individual dosage selection, though this approach has limitations. This is illustrated by two cases showing discrepancy between DPD deficient phenotype and normal standard genotype. In these two cases, DPYD exome sequencing with Next Generation Sequencing identified rare inactive variants, establishing concordance between phenotype and genotype. In patient 1, [U] levels of 21.1 and 25.5 ng/mL, indicated partial deficiency though the targeted genotype was normal and 5FU dose was adjusted based on the phenotype. In patient 2, [U] levels of 16.2 and 15.2 ng/mL were near the 16 ng/ml threshold. With a normal genotype, he as considered non-deficient as targeted genotype was normal and the standard dose was administered. These two cases underscore the need to pair DPD phenotyping with whole DPYD gene sequencing, due to the frequent discrepancies between these pharmacogenetic tools, the burden of rare variants and ethnic differences in variant frequencies.
Collapse
Affiliation(s)
- Paul Vilquin
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
- INSERM UMRS 976, Paris, France
| | - Yves Medard
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Fabienne Thomas
- Oncopole Claudius Regaud, Institut Universitaire du Cancer and Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
- University of Toulouse, Inserm, Toulouse, France
| | - Lauriane Goldwirt
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Luis Teixeira
- Service de Sénologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Sénopôle Territoire Cancer Nord, Toulouse, France
- Université Paris Cité, Paris, France
| | - Samia Mourah
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
- INSERM UMRS 976, Paris, France
- Université Paris Cité, Paris, France
| | - Evelyne Jacqz-Aigrain
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France.
- Université Paris Cité, Paris, France.
- Pharmacologie biologique - Pharmacogénétique Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, 1 avenue Charles Vellefaux - Paris, Saint-Louis, 75010, France.
| |
Collapse
|
25
|
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
|
26
|
Cavallari LH, Hicks JK, Patel JN, Elchynski AL, Smith DM, Bargal SA, Fleck A, Aquilante CL, Killam SR, Lemke L, Ochi T, Ramsey LB, Haidar CE, Ho T, El Rouby N, Monte AA, Allen JD, Beitelshees AL, Bishop JR, Bousman C, Campbell R, Cicali EJ, Cook KJ, Duong B, Tsermpini EE, Girdwood ST, Gregornik DB, Grimsrud KN, Lamb N, Lee JC, Lopez RO, Mazhindu TA, Morris SA, Nagy M, Nguyen J, Pasternak AL, Petry N, van Schaik RH, Schultz A, Skaar TC, Al Alshaykh H, Stevenson JM, Stone RM, Tran NK, Tuteja S, Woodahl EL, Yuan LC, Lee CR. The Pharmacogenomics Global Research Network Implementation Working Group: global collaboration to advance pharmacogenetic implementation. Pharmacogenet Genomics 2025; 35:1-11. [PMID: 39485373 PMCID: PMC11664750 DOI: 10.1097/fpc.0000000000000547] [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] [Indexed: 11/03/2024]
Abstract
Pharmacogenetics promises to optimize treatment-related outcomes by informing optimal drug selection and dosing based on an individual's genotype in conjunction with other important clinical factors. Despite significant evidence of genetic associations with drug response, pharmacogenetic testing has not been widely implemented into clinical practice. Among the barriers to broad implementation are limited guidance for how to successfully integrate testing into clinical workflows and limited data on outcomes with pharmacogenetic implementation in clinical practice. The Pharmacogenomics Global Research Network Implementation Working Group seeks to engage institutions globally that have implemented pharmacogenetic testing into clinical practice or are in the process or planning stages of implementing testing to collectively disseminate data on implementation strategies, metrics, and health-related outcomes with the use of genotype-guided drug therapy to ultimately help advance pharmacogenetic implementation. This paper describes the goals, structure, and initial projects of the group in addition to implementation priorities across sites and future collaborative opportunities.
Collapse
Affiliation(s)
- Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville
| | - J. Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jai N. Patel
- Atrium Health Levine Cancer Institute, Charlotte
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | | | - D. Max Smith
- MedStar Health, Columbia, Maryland
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Salma A. Bargal
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashley Fleck
- Department of Pharmacy, Richard L. Roudebush Veterans Affairs Medical Center, Veteran Health Indiana, Indianapolis, Indiana
| | - Christina L. Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shayna R. Killam
- L.S. Skaggs Institute for Health Innovation and Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | | | - Taichi Ochi
- Department of Pharmacotherapy, Epidemiology & Economics, Groningen Research Institute of Pharmacy; and University Library, University of Groningen, Groningen, The Netherlands
| | - Laura B. Ramsey
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
| | - Cyrine E. Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Teresa Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Nihal El Rouby
- Department of Pharmacy, St. Elizabeth HealthCare, Edgewood, Kentucky
- Division of Pharmacy Practice and Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Andrew A. Monte
- Rocky Mountain Poison & Drug Safety, Denver Health & Hospital Authority, Denver, Colorado
- University of Colorado School of Medicine, Aurora, Colorado
| | - Josiah D. Allen
- Department of Pharmacy, St. Elizabeth HealthCare, Edgewood, Kentucky
| | - Amber L. Beitelshees
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jeffrey R. Bishop
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chad Bousman
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
| | - Ronald Campbell
- Allegheny General Hospital, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Emily J. Cicali
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville
| | - Kelsey J. Cook
- Department of Pharmacy Education and Practice, University of Florida College of Pharmacy
- Nemours Children’s Health, Jacksonville, Florida
| | - Benjamin Duong
- Precision Medicine Program, Nemours Children’s Health Delaware Valley, Wilmington, Delaware, USA
| | - Evangelia Eirini Tsermpini
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Sonya Tang Girdwood
- Divisions of Hospital Medicine and Translational and Clinical Pharmacology, Cincinnati Children’s Hospital
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David B. Gregornik
- Pharmacogenomics Program, Children’s Minnesota, Minneapolis/St Paul, Minnesota
| | - Kristin N. Grimsrud
- Department of Pathology and Laboratory Medicine, University of California Health, Sacramento, California
| | - Nathan Lamb
- Department of Pharmacy, Ann & Robert H. Lurie Children’s Hospital of Chicago
| | - James C. Lee
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rocio Ortiz Lopez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| | | | - Sarah A. Morris
- Atrium Health Levine Cancer Institute, Charlotte
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Mohamed Nagy
- Personalised Medication Management Unit, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Jenny Nguyen
- Personalized Care Program, Children’s Hospital Los Angeles, Los Angeles, California
| | - Amy L. Pasternak
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan
| | - Natasha Petry
- Sanford Imagenetics, Sanford Health, Sioux Falls, South Dakota
- Department of Pharmacy Practice, North Dakota State University, Fargo, North Dakota, USA
| | - Ron H.N. van Schaik
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - April Schultz
- Sanford Imagenetics, Sanford Health, Sioux Falls, South Dakota
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, South Dakota
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hana Al Alshaykh
- Pharmaceutical Care Department, King Faisal Specialist Hospital and Research Center, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
| | - James M. Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rachael M. Stone
- Department of Pharmacy, University of Virginia, Charlottesville, Virginia
| | - Nam K. Tran
- Department of Pathology and Laboratory Medicine, University of California Health, Sacramento, California
| | - Sony Tuteja
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Erica L. Woodahl
- L.S. Skaggs Institute for Health Innovation and Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Li-Chi Yuan
- Providence Health and Services, Irvine, California
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
27
|
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
|
28
|
Guevara M, de la Cruz CG, Rodrigues-Soares F, Rodríguez E, Manóchio C, Peñas-Lledó E, Dorado P, LLerena A. The Frequency of DPYD c.557A>G in the Dominican Population and Its Association with African Ancestry. Pharmaceutics 2024; 17:8. [PMID: 39861660 PMCID: PMC11768636 DOI: 10.3390/pharmaceutics17010008] [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: 11/14/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Genetic polymorphism of the dihydropyrimidine dehydrogenase gene (DPYD) is responsible for the variability found in the metabolism of fluoropyrimidines such as 5-fluorouracil (5-FU), capecitabine, or tegafur. The DPYD genotype is linked to variability in enzyme activity, 5-FU elimination, and toxicity. Approximately 10-40% of patients treated with fluoropyrimidines develop severe toxicity. The interethnic variability of DPYD gene variants in Afro-Latin Americans is poorly studied, thereby establishing a barrier to the implementation of personalized medicine in these populations. Therefore, the present study aims to analyze the frequency of DPYD variants with clinical relevance in the Dominican population and their association with genomic ancestry components. Methods: For this study, 196 healthy volunteers from the Dominican Republic were genotyped for DPYD variants by qPCR, and individual genomic ancestry analysis was performed in 178 individuals using 90 informative ancestry markers. Data from the 1000 Genomes project were also retrieved for comparison and increased statistical power. Results and Conclusions: The c.557A>G variant (decreased dihydropyrimidine dehydrogenase function) presented a frequency of 2.6% in the Dominican population. Moreover, the frequency of this variant is positively associated with African ancestry (r2 = 0.67, p = 1 × 10-7), which implies that individuals with high levels of African ancestry are more likely to present this variant. HapB3 is completely absent in Dominican, Mexican, Peruvian, Bangladeshi, and all East Asian and African populations, which probably makes its analysis dispensable in these populations. The implementation of pharmacogenetics in oncology, specifically DPYD, in populations of Afro-Latin American ancestry should include c.557A>G, to be able to carry out the safe and effective treatment of patients treated with fluoropyrimidines.
Collapse
Affiliation(s)
- Mariela Guevara
- Research and Development Department, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 10203, Dominican Republic; (M.G.); (E.R.)
| | - Carla González de la Cruz
- Personalized Medicine and Mental Health Unit, University Institute for Bio-Sanitary Research of Extremadura, 06080 Badajoz, Spain; (C.G.d.l.C.); (F.R.-S.); (E.P.-L.); (A.L.)
| | - Fernanda Rodrigues-Soares
- Personalized Medicine and Mental Health Unit, University Institute for Bio-Sanitary Research of Extremadura, 06080 Badajoz, Spain; (C.G.d.l.C.); (F.R.-S.); (E.P.-L.); (A.L.)
- Department of Pathology, Genetic and Evolution, Biological and Natural Sciences Institute, Universidade Federal do Triângulo Mineiro, Uberaba 38025-350, Brazil;
| | - Ernesto Rodríguez
- Research and Development Department, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 10203, Dominican Republic; (M.G.); (E.R.)
| | - Caíque Manóchio
- Department of Pathology, Genetic and Evolution, Biological and Natural Sciences Institute, Universidade Federal do Triângulo Mineiro, Uberaba 38025-350, Brazil;
- Department of Genetics, Ecology and Evolution, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Eva Peñas-Lledó
- Personalized Medicine and Mental Health Unit, University Institute for Bio-Sanitary Research of Extremadura, 06080 Badajoz, Spain; (C.G.d.l.C.); (F.R.-S.); (E.P.-L.); (A.L.)
| | - Pedro Dorado
- Personalized Medicine and Mental Health Unit, University Institute for Bio-Sanitary Research of Extremadura, 06080 Badajoz, Spain; (C.G.d.l.C.); (F.R.-S.); (E.P.-L.); (A.L.)
| | - Adrián LLerena
- Personalized Medicine and Mental Health Unit, University Institute for Bio-Sanitary Research of Extremadura, 06080 Badajoz, Spain; (C.G.d.l.C.); (F.R.-S.); (E.P.-L.); (A.L.)
| |
Collapse
|
29
|
Hernández-Guío A, Ángel Calleja-Hernández M, Corno-Caparrós A, Zayas-Soriano M, Bernabéu-Martínez MÁ, Gutiérrez-Nicolás F. Clinical impact of DPYD genotyping and dose adjustment in candidates for fluoropyrimidine treatment. Heliyon 2024; 10:e40808. [PMID: 39719989 PMCID: PMC11666974 DOI: 10.1016/j.heliyon.2024.e40808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/02/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ana Hernández-Guío
- Department of Pharmacy, San Juan de Alicante University Hospital, N-332, s/n, 03550, Sant Joan d'Alacant, Alicante, Spain
| | | | - Andrés Corno-Caparrós
- Miguel Hernández University, Elche, Avinguda de la Universitat d'Elx, s/n, 03202, Elche, Alicante, Spain
| | - Marta Zayas-Soriano
- Department of Pharmacy, San Juan de Alicante University Hospital, N-332, s/n, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Mª Ángeles Bernabéu-Martínez
- Department of Pharmacy, San Juan de Alicante University Hospital, N-332, s/n, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Fernando Gutiérrez-Nicolás
- Instituto de Investigación Sanitaria de Canarias, Carretera Gral. La Cuesta-Taco, n.s/n Pabellón de Gobierno del HUC Planta -1ª, 38320, San Cristobal de La Laguna, Santa Cruz de, Tenerife, Spain
| |
Collapse
|
30
|
De Mattia E, Polesel J, Scarabel L, Cecchin E. Clinical implications of a gain-of-function genetic polymorphism in DPYD (rs4294451) in colorectal cancer patients treated with fluoropyrimidines. Front Pharmacol 2024; 15:1516375. [PMID: 39703399 PMCID: PMC11655210 DOI: 10.3389/fphar.2024.1516375] [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: 10/24/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
Dihydropyrimidine dehydrogenase (DPD, encoded by the DPYD gene) is the rate-limiting enzyme for the detoxification of fluoropyrimidines (FLs). Rs4294451 is a regulatory DPYD polymorphism that has recently been functionally characterized and associated with increased DPD expression in the liver. The aim of the present study was to test the clinical implications of being a carrier of rs4294451 in a cohort of 645 FL-treated colorectal cancer patients. Carriers of at least one DPYD rs4294451-T variant allele had a lower risk of developing NCI-CTC grade 4-5 hematological [odds ratio (OR) = 0.39; 95% confidence interval (CI): 0.15-0.98; additive model] and hematological/non-hematological (OR = 0.44; 95% CI: 0.22-0.88; dominant model) FL-related toxicity. Patients with the DPYD rs4294451-T allele also had a longer time to severe toxicity development after starting FL treatment [hematological, Hazard ratio (HR) = 0.27; 95% CI: 0.09-0.79; Fine-Gray test = 0.1569; hematological/non-hematological: HR = 0.38, 95% CI: 0.17-0.85; Fine-Gray test = 0.0444]. It is worth noting that while being at lower risk of toxicity, DPYD rs4294451-T allele carriers also tend to present a shorter overall survival (HR = 1.41; 95% CI: 1.05-1.90; log-rank p = 0.0406). These findings demonstrate a clinical effect of DPYD-rs4294451 polymorphism coherent with the recently described functional effect. Further investigation is warranted to elucidate the potential clinical value to the rs4294451 polymorphism as toxicity and especially as an efficacy marker in colorectal cancer.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Province of Pordenone, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Province of Pordenone, Italy
| | - Lucia Scarabel
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Province of Pordenone, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Province of Pordenone, Italy
| |
Collapse
|
31
|
Roncato R, Bignucolo A, Peruzzi E, Montico M, De Mattia E, Foltran L, Guardascione M, D’Andrea M, Favaretto A, Puglisi F, Swen JJ, Guchelaar HJ, Toffoli G, Cecchin E. Clinical Benefits and Utility of Pretherapeutic DPYD and UGT1A1 Testing in Gastrointestinal Cancer: A Secondary Analysis of the PREPARE Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2449441. [PMID: 39641926 PMCID: PMC11624585 DOI: 10.1001/jamanetworkopen.2024.49441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/14/2024] [Indexed: 12/07/2024] Open
Abstract
Importance To date, the clinical benefit and utility of implementing a DPYD/UGT1A1 pharmacogenetic-informed therapy with fluoropyrimidines and/or irinotecan have not been prospectively investigated. Objective To examine clinically relevant toxic effects, hospitalizations, and related costs while preserving treatment intensity and efficacy outcomes in patients with gastrointestinal cancer. Design, Setting, and Participants This nonprespecified secondary analysis stems from Pre-Emptive Pharmacogenomic Testing for Preventing Adverse Drug Reactions (PREPARE), a multicenter, controlled, open, block-randomized, crossover implementation trial conducted from March 7, 2017, to June 30, 2020, and includes data from Italy according to a sequential study design. The study population included 563 patients (intervention, 252; control [standard of care], 311) with gastrointestinal cancer (age ≥18 years) who were eligible for fluoropyrimidine and/or irinotecan treatment. Data analysis for the present study was performed from May 27 to October 10, 2024. Interventions Participants with actionable variants (DPYD*2A, DPYD*13, .DPYD c.2846A>T, and DPYD c.1236G>A for fluoropyrimidines, and UGT1A1*28, UGT1A1*6, and UGT1A1*27 for irinotecan) received drug or dose adjustments based on Dutch Pharmacogenetics Working Group recommendations. Main Outcomes and Measures The primary outcome was clinically relevant toxic effects (National Cancer Institute Common Terminology Criteria for Adverse Events grade ≥4 hematologic, grade ≥3 nonhematologic, or causing hospitalization, fluoropyrimidines and/or irinotecan causally related). Secondary outcomes included hospitalization rates, toxic effect management costs, intensity of treatment, quality-adjusted life-years, and 3-year overall survival. Results Overall, 1232 patients were enrolled in Italy, with 563 included in this analysis (317 [56.3%] men; median age, 68.0 [IQR, 60.0-75.0] years). In the intervention arm, carriers of any actionable genotype exhibited a 90% lower risk of clinically relevant toxic effects compared with the control arm (odds ratio, 0.1; 95% CI, 0.0-0.8; P = .04). They also presented higher toxic effect management costs per patient ($4159; 95% CI, $1510-$6810) compared with patients in the intervention arm ($26; 95% CI, 0-$312) (P = .004) and a higher rate of hospitalization (34.8% vs 11.8%; P = .12). The differences were not significant among all patients. Three-year overall survival did not differ significantly between arms, while quality-adjusted life-years significantly improved in the intervention arm. The pharmacogenetics-informed approach did not manifest a detrimental effect on treatment intensity in actionable genotype carriers. Conclusions and Relevance In this secondary analysis of PREPARE, pretreatment application of DPYD- and UGT1A1-guided treatment appeared to increase safety and reduce hospitalizations and related costs in patients with gastrointestinal cancer. Clinical benefit did not appear to be affected. Trial Registration ClinicalTrials.gov Identifier: NCT03093818.
Collapse
Affiliation(s)
- Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elena Peruzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Luisa Foltran
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Michela Guardascione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Mario D’Andrea
- Department of Medical Oncology, Ospedale San Paolo / Ospedale Padre Pio, Civitavecchia, Rome, Italy
| | - Adolfo Favaretto
- Department of Medical Oncology, Azienda ULSS 2 Marca Trevigiana Distretto di Treviso, Treviso, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Jesse Joachim Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
32
|
McLeod HL, Nguyen DG. Pharmacogenomics in Oncology-Running Out of Excuses for Slow Adoption. JAMA Netw Open 2024; 7:e2449453. [PMID: 39641933 DOI: 10.1001/jamanetworkopen.2024.49453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Affiliation(s)
- Howard L McLeod
- Center for Precision Medicine, Utah Tech University, St George
| | - D Grace Nguyen
- Department of Cancer Pharmacology and Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, North Carolina
| |
Collapse
|
33
|
Glewis S, Lingaratnam S, Lee B, Campbell I, IJzerman M, Fagery M, Harris S, Georgiou C, Underhill C, Warren M, Campbell R, Jayawardana M, Silva SSM, Martin JH, Tie J, Alexander M, Michael M. Pharmacogenetic-guided dosing for fluoropyrimidine (DPYD) and irinotecan (UGT1A1*28) chemotherapies for patients with cancer (PACIFIC-PGx): A multicenter clinical trial. Clin Transl Sci 2024; 17:e70083. [PMID: 39614408 PMCID: PMC11606843 DOI: 10.1111/cts.70083] [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: 05/07/2024] [Revised: 09/26/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024] Open
Abstract
PACIFIC-PGx evaluated the feasibility of implementing pharmacogenetics (PGx) screening in Australia and the impact of DPYD/UGT1A1 genotype-guided dosing on severe fluoropyrimidine (FP) and irinotecan-related toxicities and hospitalizations, compared to historical controls. This prospective single arm trial enrolled patients starting FP/irinotecan for any cancer between 7 January 2021 and 25 February 2022 from four Australian hospitals (one metropolitan, three regional). During the accrual period, 462/487 (95%) consecutive patients screened for eligibility for DPYD and 50/109 (46%) for UGT1A1 were enrolled and genotyped (feasibility analysis), with 276/462 (60%) for DPYD and 30/50 (60%) for UGT1A1 received FP/irinotecan (safety analysis). DPYD genotyping identified 96% (n = 443/462) Wild-Type, 4% (n = 19/462) Intermediate Metabolizers (50% dose reduction), and 0% Poor Metabolizers. UGT1A1 genotyping identified 52% (n = 26/50) Wild-Type, 40% (n = 20/50) heterozygous, and 8% (n = 4/50) homozygous (30% dose reduction). Key demographics for the FP/irinotecan safety cohorts included: age range 23-89/34-74 years, male 56%/73%, Caucasian 83%/73%, lower gastrointestinal cancer 50%/57%. Genotype results were reported prior to cycle-1 (96%), average 5-7 days from sample collection. PGx-dosing for DPYD variant allele carriers reduced high-grade toxicities compared to historic controls (7% vs. 39%; OR = 0.11, 95% CI 0.01-0.97, p = 0.024). High-grade toxicities among Wild-Type were similar (14% vs. 14%; OR = 0.99, 95% CI 0.64-1.54, p = 0.490). PGx-dosing reduced FP-related hospitalizations (-22%) and deaths (-3.7%) compared to controls. There were no high-grade toxicities or hospitalizations for UGT1A1*28 homozygotes. PGx screening and prescribing were feasible in routine oncology care and improved patient outcomes. Findings may inform expanded PGx programs within cancer and other disease settings.
Collapse
Affiliation(s)
- Sarah Glewis
- Department of PharmacyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Senthil Lingaratnam
- Department of PharmacyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Benjamin Lee
- Department of PharmacyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Ian Campbell
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Cancer Genetics LaboratoryPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Maarten IJzerman
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Cancer Health Services Research Unit, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Cancer Health Services Research Unit, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Erasmus School of Health Policy and ManagementErasmus University RotterdamRotterdamThe Netherlands
| | - Mussab Fagery
- Cancer Health Services Research Unit, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Cancer Health Services Research Unit, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Sam Harris
- Department of Medical OncologyBendigo HealthBendigoVictoriaAustralia
| | - Chloe Georgiou
- Department of Medical OncologyBendigo HealthBendigoVictoriaAustralia
| | - Craig Underhill
- VCCC AllianceParkvilleVictoriaAustralia
- Border Medical Oncology Research UnitAlbury Wodonga Regional Cancer CentreEast AlburyNew South WalesAustralia
- UNSW Rural Medical School, Albury CampusNew South WalesEast AlburyAustralia
| | - Mark Warren
- Department of Medical OncologyBendigo HealthBendigoVictoriaAustralia
| | - Robert Campbell
- Department of Medical OncologyBendigo HealthBendigoVictoriaAustralia
| | - Madawa Jayawardana
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Office of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - S. Sandun M. Silva
- Centre for Health Systems and Safety Research, Australian Institute of Health InnovationMacquarie UniversityMacquarie ParkNew South WalesAustralia
| | - Jennifer H. Martin
- School of Medicine and Public HealthUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Jeanne Tie
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Personalised Oncology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Marliese Alexander
- Department of PharmacyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Michael Michael
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| |
Collapse
|
34
|
Sarhangi N, Rouhollah F, Niknam N, Sharifi F, Nikfar S, Larijani B, Patrinos GP, Hasanzad M. Pharmacogenetic DPYD allele variant frequencies: A comprehensive analysis across an ancestrally diverse Iranian population. Daru 2024; 32:715-727. [PMID: 39424756 PMCID: PMC11555172 DOI: 10.1007/s40199-024-00538-7] [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: 04/19/2024] [Accepted: 08/24/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Cancer treatment has improved over the past decades, but many cancer patients still experience adverse drug reactions (ADRs). Pharmacogenomics (PGx), known as personalized treatment, is a pillar of precision medicine that aims to optimize the efficacy and safety of medications by studying the germline variations. Germline variations in the DPYD lead to significant ADRs. The present cross-sectional study aims to evaluate the allele frequency of the DPYD gene variations in the Iranian population to provide insights into personalized treatment decisions in the Iranian population. METHODS The allele frequency of 51 pharmacogenetic variations in the clinically relevant DPYD was assessed in a representative sample set of 1142 unrelated Iranian individuals and subpopulations of different ethnic groups who were genotyped using the Infinium Global Screening Array-24 BeadChip. RESULTS The genotyping assay revealed eight pharmacogenetic variants including DPYD rs1801265 (c.85T > C; DPYD*9A), rs2297595 (c.496A > G), rs1801158 (c.1601G > A; DPYD*4), rs1801159 (c.1627A > G; DPYD*5), rs1801160 (c.2194G > A; DPYD*6), rs17376848 (c.1896T > C), rs56038477 (c.1236G > A; HapB3), and rs75017182 (c.1129-5923C > G; HapB3) with minor allele frequency (MAF) ≥ 1%. CONCLUSION The results of the study reveal significant genetic variations among Iranian population that could significantly influence clinical decision-making. These variants, with their potential to explain the substantial variability in drug response phenotypes among different populations, shed light on a crucial aspect of pharmacogenomics. These findings not only provide valuable insights but also inspire the design and implementation of future pharmacogenomic clinical trials, motivating further research in this crucial area.
Collapse
Affiliation(s)
- Negar Sarhangi
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, 1916893813, Iran
| | - Fatemeh Rouhollah
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, 1916893813, Iran
| | - Negar Niknam
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
- LifeandMe, Inc., Tehran, 1497719825, Iran
| | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - Shekoufeh Nikfar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - George P Patrinos
- School of Health Sciences, Department of Pharmacy, University of Patras, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al-Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, Abu Dhabi, UAE
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran.
| |
Collapse
|
35
|
Lingyan L, Linjun W, Wenjun Z. Methylenetetrahydrofolate Reductase (MTHFR) Variants and Severe Capecitabine Toxicity: A Case Report and Review of Literature. Cureus 2024; 16:e75791. [PMID: 39816293 PMCID: PMC11734037 DOI: 10.7759/cureus.75791] [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] [Accepted: 12/15/2024] [Indexed: 01/18/2025] Open
Abstract
Capecitabine is an oral prodrug metabolized into 5-fluorouracil (5-FU) and serves as a representative anticancer agent. While fluoropyrimidine treatment is usually well-tolerated, a subset of patients unfortunately experiences severe and sometimes life-threatening toxicity related to these compounds. This adverse reaction is frequently attributed to partial or complete deficiencies in the dihydropyrimidine dehydrogenase (DPD) enzyme. However, some patients may still suffer from severe toxic effects despite normal DPD screening results when treated with capecitabine. This paper presents the case of a Chinese woman with stage IIIB moderately differentiated adenocarcinoma of the lower rectum (cT3N2aM0) who exhibited severe toxicity after two weeks of neoadjuvant concurrent chemoradiotherapy in TNT mode at a low dose (825mg/m2 bid) of capecitabine. We found that this severe toxicity might be attributable to insufficient methylenetetrahydrofolate reductase (MTHFR) activity. To our knowledge, such reports are scarce in the medical literature concerning the Chinese population.
Collapse
Affiliation(s)
- Li Lingyan
- Oncology, Qiannan People's Hospital, Duyun, CHN
| | - Wang Linjun
- Oncology, Qiannan People's Hospital, Duyun, CHN
| | | |
Collapse
|
36
|
Walko CM. DPYD and fluoropyrimidines: Using the data as our guide. Pharmacotherapy 2024; 44:896-897. [PMID: 39737555 DOI: 10.1002/phar.4634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 01/01/2025]
|
37
|
Launay M, Raymond L, Guitton J, Loriot MA, Chatelut E, Haufroid V, Thomas F, Etienne-Grimaldi MC. Can we identify patients carrying targeted deleterious DPYD variants with plasma uracil and dihydrouracil? A GPCO-RNPGx retrospective analysis. Clin Chem Lab Med 2024; 62:2415-2424. [PMID: 38896022 DOI: 10.1515/cclm-2024-0317] [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: 03/08/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Dihydropyrimidine dehydrogenase (DPD) deficiency is the main cause of severe fluoropyrimidine-related toxicities. The best strategy for identifying DPD-deficient patients is still not defined. The EMA recommends targeted DPYD genotyping or uracilemia (U) testing. We analyzed the concordance between both approaches. METHODS This study included 19,376 consecutive French patients with pre-treatment plasma U, UH2 and targeted DPYD genotyping (*2A, *13, D949V, *7) analyzed at Eurofins Biomnis (2015-2022). RESULTS Mean U was 9.9 ± 10.1 ng/mL (median 8.7, range 1.6-856). According to French recommendations, 7.3 % of patients were partially deficient (U 16-150 ng/mL) and 0.02 % completely deficient (U≥150 ng/mL). DPYD variant frequencies were *2A: 0.83 %, *13: 0.17 %, D949V: 1.16 %, *7: 0.05 % (2 homozygous patients with U at 22 and 856 ng/mL). Variant carriers exhibited higher U (median 13.8 vs. 8.6 ng/mL), and lower UH2/U (median 7.2 vs. 11.8) and UH2/U2 (median 0.54 vs. 1.37) relative to wild-type patients (p<0.00001). Sixty-six% of variant carriers exhibited uracilemia <16 ng/mL, challenging correct identification of DPD deficiency based on U. The sensitivity (% patients with a deficient phenotype among variant carriers) of U threshold at 16 ng/mL was 34 %. The best discriminant marker for identifying variant carriers was UH2/U2. UH2/U2<0.942 (29.7 % of patients) showed enhanced sensitivity (81 %) in identifying deleterious genotypes across different variants compared to 16 ng/mL U. CONCLUSIONS These results reaffirm the poor concordance between DPD phenotyping and genotyping, suggesting that both approaches may be complementary and that targeted DPYD genotyping is not sufficiently reliable to identify all patients with complete deficiency.
Collapse
Affiliation(s)
- Manon Launay
- Service de médecine intensive et réanimation médicale et Centre Régional de Pharmacovigilance, CHU de Saint-Etienne, Saint-Etienne, France
- French Clinical Oncopharmacology Group (GPCO)-UNICANCER, Paris, France
| | - Laure Raymond
- Département de génétique, Laboratoire Eurofins Biomnis, Lyon, France
- Francophone Network of Pharmacogenetics (RNPGx), Paris, France
| | - Jérôme Guitton
- Laboratoire de Biochimie et Toxicologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
- Laboratoire de Toxicologie, ISPB, Faculté de Pharmacie, Université Lyon 1, Université de Lyon, Lyon, France
- French Clinical Oncopharmacology Group (GPCO)-UNICANCER, Paris, France
| | - Marie-Anne Loriot
- Department of Clinical Chemistry, Hôpital européen Georges-Pompidou, Assistance Publique Hôpitaux de Paris-Centre, Paris, France
- INSERM UMR-S1138, Université of Paris Cité, Centre de recherches des Cordeliers, Paris, France
- Francophone Network of Pharmacogenetics (RNPGx), Paris, France
| | - Etienne Chatelut
- Oncopole Claudius Regaud, Institut Universitaire du Cancer and CRCT, University of Toulouse, Inserm, Toulouse, France
- French Clinical Oncopharmacology Group (GPCO)-UNICANCER, Paris, France
| | - Vincent Haufroid
- Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de recherche expérimentale et clinique, UClouvain, Brussels and Clinical Chemistry Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Francophone Network of Pharmacogenetics (RNPGx), Paris, France
| | - Fabienne Thomas
- Oncopole Claudius Regaud, Institut Universitaire du Cancer and CRCT, University of Toulouse, Inserm, Toulouse, France
- French Clinical Oncopharmacology Group (GPCO)-UNICANCER, Paris, France
- Francophone Network of Pharmacogenetics (RNPGx), Paris, France
| | - Marie-Christine Etienne-Grimaldi
- Oncopharmacology Laboratory, Centre Antoine Lacassagne, Nice, France
- French Clinical Oncopharmacology Group (GPCO)-UNICANCER, Paris, France
- Francophone Network of Pharmacogenetics (RNPGx), Paris, France
| |
Collapse
|
38
|
Ragia G, Pallikarou M, Michou C, Manolopoulos VG. MIR27A rs895819 CC Genotype Severely Reduces miR-27a Plasma Expression Levels. Genes (Basel) 2024; 15:1491. [PMID: 39596691 PMCID: PMC11593693 DOI: 10.3390/genes15111491] [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: 10/29/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives:MIR27A rs895819 polymorphism has emerged as a potential additional pharmacogenomic marker of fluoropyrimidine response. Current evidence on its potential effect on miR-27a expression, which represses DPD activity, leading to DPD deficiency and increased fluoropyrimidine-associated toxicity risk, is scarce and inconsistent. We have analyzed the effect of MIR27A rs895819 polymorphism on miR-27a-3p plasma expression levels under different models of inheritance to contribute further evidence on its plausible biological role in miR-27a expression. Methods: A total of 59 individuals with no medical history of cancer were included in this study. MIR27A rs895819 genotyping and miR-27a-3p expression were analyzed by using predesigned TaqMan assays. Results: The frequency of TT, TC, and CC genotypes was present at a prevalence of 50.8%, 44.1%, and 5.1%, respectively. Individuals carrying the CC genotype presented with decreased miR-27a-3p expression (0.422 fold-change versus TT, p = 0.041; 0.461 fold-change versus TC, p = 0.064), whereas no differences were present between TT and TC individuals (1.092 fold-change, p = 0.718). miR-27a-3p expression was decreased in CC individuals under a recessive model of inheritance (0.440 fold-change, p = 0.047). No differences were found in dominant (TT vs. TC+CC, 0.845 fold-change, p = 0.471) or over dominant (TT+CC vs. TC, 0.990 fold-change, p = 0.996) models of inheritance. Conclusions:MIR27A rs895819CC genotype leads to severely reduced miR-27a-3p expression in plasma. Further study of this association is warranted in cancer patients to apply MIR27A genotyping in therapeutics to identify fluoropyrimidine-treated patients who are at a decreased risk of experiencing fluoropyrimidine-induced severe toxicity.
Collapse
Affiliation(s)
- Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, 68100 Alexandroupolis, Greece; (M.P.); (C.M.)
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, 68100 Alexandroupolis, Greece
| | - Myria Pallikarou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, 68100 Alexandroupolis, Greece; (M.P.); (C.M.)
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, 68100 Alexandroupolis, Greece
| | - Chrysoula Michou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, 68100 Alexandroupolis, Greece; (M.P.); (C.M.)
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, 68100 Alexandroupolis, Greece; (M.P.); (C.M.)
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, 68100 Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Dragana Campus, 68100 Alexandroupolis, Greece
| |
Collapse
|
39
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [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: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
40
|
Tęcza K, Kalinowska-Herok M, Rusinek D, Zajkowicz A, Pfeifer A, Oczko-Wojciechowska M, Pamuła-Piłat J. Are the Common Genetic 3'UTR Variants in ADME Genes Playing a Role in Tolerance of Breast Cancer Chemotherapy? Int J Mol Sci 2024; 25:12283. [PMID: 39596349 PMCID: PMC11594993 DOI: 10.3390/ijms252212283] [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: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
We studied the associations between 3'UTR genetic variants in ADME genes, clinical factors, and the risk of breast cancer chemotherapy toxicity. Those variants and factors were tested in relation to seven symptoms belonging to myelotoxicity (anemia, leukopenia, neutropenia), gastrointestinal side effects (vomiting, nausea), nephrotoxicity, and hepatotoxicity, occurring in overall, early, or recurrent settings. The cumulative risk of overall symptoms of anemia was connected with AKR1C3 rs3209896 AG, ERCC1 rs3212986 GT, and >6 cycles of chemotherapy; leukopenia was determined by ABCC1 rs129081 allele G and DPYD rs291593 allele T; neutropenia risk was correlated with accumulation of genetic variants of DPYD rs291583 allele G, ABCB1 rs17064 AT, and positive HER2 status. Risk of nephrotoxicity was determined by homozygote DPYD rs291593, homozygote AKR1C3 rs3209896, postmenopausal age, and negative ER status. Increased risk of hepatotoxicity was connected with NR1/2 rs3732359 allele G, postmenopausal age, and with present metastases. The risk of nausea and vomiting was linked to several genetic factors and premenopausal age. We concluded that chemotherapy tolerance emerges from the simultaneous interaction of many genetic and clinical factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jolanta Pamuła-Piłat
- Department of Clinical and Molecular Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (K.T.); (M.K.-H.); (D.R.); (A.Z.); (A.P.); (M.O.-W.)
| |
Collapse
|
41
|
Metts JL, Aye JM, Crane JN, Oberoi S, Balis FM, Bhatia S, Bona K, Carleton B, Dasgupta R, Dela Cruz FS, Greenzang KA, Kaufman JL, Linardic CM, Parsons SK, Robertson-Tessi M, Rudzinski ER, Soragni A, Stewart E, Weigel BJ, Wolden SL, Weiss AR, Venkatramani R, Heske CM. Roadmap for the next generation of Children's Oncology Group rhabdomyosarcoma trials. Cancer 2024; 130:3785-3796. [PMID: 38941509 PMCID: PMC11511643 DOI: 10.1002/cncr.35457] [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] [Indexed: 06/30/2024]
Abstract
Clinical trials conducted by the Intergroup Rhabdomyosarcoma (RMS) Study Group and the Children's Oncology Group have been pivotal to establishing current standards for diagnosis and therapy for RMS. Recent advancements in understanding the biology and clinical behavior of RMS have led to more nuanced approaches to diagnosis, risk stratification, and treatment. The complexities introduced by these advancements, coupled with the rarity of RMS, pose challenges to conducting large-scale phase 3 clinical trials to evaluate new treatment strategies for RMS. Given these challenges, systematic planning of future clinical trials in RMS is paramount to address pertinent questions regarding the therapeutic efficacy of drugs, biomarkers of response, treatment-related toxicity, and patient quality of life. Herein, the authors outline the proposed strategic approach of the Children's Oncology Group Soft Tissue Sarcoma Committee to the next generation of RMS clinical trials, focusing on five themes: improved novel agent identification and preclinical to clinical translation, more efficient trial development and implementation, expanded opportunities for knowledge generation during trials, therapeutic toxicity reduction and quality of life, and patient engagement.
Collapse
Affiliation(s)
- Jonathan L Metts
- Sarcoma Department, Moffitt Cancer Center, Tampa, Florida, USA
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Jamie M Aye
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jacquelyn N Crane
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sapna Oberoi
- Department of Pediatric Hematology/Oncology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank M Balis
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kira Bona
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Bruce Carleton
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roshni Dasgupta
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Katie A Greenzang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, USA
- Patient Advocacy Committee, Children's Oncology Group, Monrovia, California, USA
| | - Corinne M Linardic
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Susan K Parsons
- Institute for Clinical Research and Health Policy Studies and Division of Hematology/Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Mark Robertson-Tessi
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Erin R Rudzinski
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital and University of Washington Medical Center, Seattle, Washington, USA
| | - Alice Soragni
- Department of Orthopedic Surgery, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - Elizabeth Stewart
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Brenda J Weigel
- Division of Pediatric Hematology Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Aaron R Weiss
- Department of Pediatrics, Maine Medical Center, Portland, Maine, USA
| | | | - Christine M Heske
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
42
|
White C, Wardill H, Paul C, Price T, Karapetis C, Nalder M, Burge ME, Thomas A, Oldmeadow C, Barker D, Edney LC, Coller J, Bowen J, Ostroff C, Cheek B, Carlson M, Rankmore T, Nagrial A, Clarke S, Chantrill L, Ackland S, Scott RJ. DPYD genotype-guided dose personalisation for fluoropyrimidine-based chemotherapy prescribing in solid organ cancer patients in Australia: GeneScreen 5-FU study protocol. BMC Cancer 2024; 24:1369. [PMID: 39516829 PMCID: PMC11549825 DOI: 10.1186/s12885-024-13122-8] [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: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Fluoropyrimidine (FP) chemotherapies are commonly prescribed for upper and lower gastrointestinal, breast and head and neck malignancies. Over 16,000 people with cancer require FP chemotherapies per annum in Australia. Between 10 and 40% patients experience grade 3-4 (≥ G3) toxicities that require hospital-based management ± intensive care admission. Approximately 1% of patients die secondary to FP toxicities. Prospective screening for DPYD gene variants (encoding the key enzyme for FP catabolism) can identify patients at risk of ≥ G3 toxicity and allow for dose adjustment prior to first FP exposure. Evidence supports this as a cost-effective method of improving patient safety and reducing healthcare burden internationally; however, no Australian data confirms its feasibility on a large scale. METHOD This investigator-led, single-arm study will determine large scale feasibility of prospective DPYD genotyping, confirming patient safety and cost-effectiveness within the Australian health care system. 5000 patients aged 18 years and older with solid organ cancers requiring FP chemotherapy will be consented and genotyped prior to commencing treatment, and early toxicity (within 60 days) post-FP exposure will be determined. Toxicity data for DPYD variant carriers who have dose adjustments will be compared to the wild-type cohort and historical cohorts of carriers who did not undergo genotyping prior to FP exposure, and prospective variant carriers who do not undergo dose-adjustment. Prevalence of the four standard DPYD gene variants will be confirmed in an Australian population. Additionally, health economic analysis, implementation research via semi-structured interviews of patients and clinicians, and feasibility of UGT1A1 genotyping will be conducted. DISCUSSION This study will determine the prevalence of DPYD gene variant status in Australia and its impact on FP-induced toxicity among Australians with cancer. Feasibility and cost-effectiveness for Australian health care system will be estimated to support national roll-out of prospective DPYD genotyping prior to FP administration. Additionally, feasibility will be confirmed with the intention of including UGT1A1 in future pharmacogenomic panels to aid chemotherapy prescribing. TRIAL REGISTRATION This trial was registered with the Australian and New Zealand Cancer Trials Registry on 13th Dec 2023, ACTRN12623001301651.
Collapse
Affiliation(s)
- Cassandra White
- University of Newcastle, College of Health, Medicine and Wellbeing, School of Medicine and Public Health, Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.
| | - Hannah Wardill
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
- Supportive Oncology Research Group, Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Christine Paul
- University of Newcastle, College of Health, Medicine and Wellbeing, School of Medicine and Public Health, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Timothy Price
- The Queen Elizabeth Hospital and University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christos Karapetis
- Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Mark Nalder
- Royal Brisbane and Women's Hospital, Brisbane, QLD, 4006, Australia
- University of Queensland, Brisbane, QLD, 4006, Australia
| | - Matthew E Burge
- Royal Brisbane and Women's Hospital, Brisbane, QLD, 4006, Australia
| | - Ann Thomas
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Chris Oldmeadow
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Daniel Barker
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Laura C Edney
- Flinders University, College of Medicine and Public Health; Flinders Health and Medical Research Institute, Adelaide, SA, 5042, Australia
| | - Janet Coller
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Joanne Bowen
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Cheri Ostroff
- University of South Australia, Centre for Workplace Excellence, Adelaide, SA, 5001, Australia
| | - Bruce Cheek
- Australasian Gastrointestinal Trials Group (Consumer Panel), Camperdown, NSW, 2050, Australia
- Cancer Quality of Life Expert Service Team (Member Steering Committee), University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Cancer Voices New South Wales (Consumer Representative), Milsons Point, NSW, 1565 , Australia
| | - Mel Carlson
- University of Newcastle, College of Health, Medicine and Wellbeing, School of Medicine and Public Health, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Trumaine Rankmore
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Adnan Nagrial
- Western Sydney Local Health District, Westmead, NSW, 2148, Australia
- University of Sydney, Westmead Clinical School, Westmead, NSW, 2148, Australia
| | - Stephen Clarke
- Northern Sydney Local Health District, St. Leonards, NSW, 2065, Australia
- University of Sydney, Northern Clinical School, St. Leonards, NSW, 2065, Australia
| | - Lorraine Chantrill
- Illawarra Shoalhaven Local Health District, Wollongong, NSW, 2500, Australia
- University of Wollongong, Wollongong, NSW, 2500, Australia
| | - Stephen Ackland
- University of Newcastle, College of Health, Medicine and Wellbeing, School of Medicine and Public Health, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Rodney J Scott
- University of Newcastle, College of Health, Medicine and Wellbeing, School of Biomedical Science and Pharmacy, Callaghan, NSW, 2308, Australia
- Department of Molecular Genetics, Pathology North John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| |
Collapse
|
43
|
Steuerwald NM, Morris S, Nguyen DG, Patel JN. Understanding the Biology and Testing Techniques for Pharmacogenomics in Oncology: A Practical Guide for the Clinician. JCO Oncol Pract 2024; 20:1441-1451. [PMID: 39531848 DOI: 10.1200/op.24.00191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 11/16/2024] Open
Abstract
Pharmacogenomic (PGx) testing is a growing area of personalized medicine with demonstrated clinical utility in improving patient outcomes in oncology. PGx testing of pharmacogenes affecting drug pharmacokinetics, pharmacodynamics, and response can help inform drug selection and dosing of several anticancer therapies and supportive care medications. Several PGx testing techniques exist including polymerase chain reaction (PCR), MassARRAY, microarray, and sequencing. This review article provides a clinician-friendly guide of these techniques. Understanding the advantages, limitations, ideal use, and potential clinical applications of each platform can help clinicians choose the appropriate PGx testing platform for specific use cases.
Collapse
Affiliation(s)
- Nury M Steuerwald
- Molecular Biology and Genomics Core Laboratory, Atrium Health Levine Cancer Institute, Charlotte, NC
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Sarah Morris
- Department of Cancer Pharmacology and Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - D Grace Nguyen
- Department of Cancer Pharmacology and Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Jai N Patel
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Cancer Pharmacology and Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| |
Collapse
|
44
|
Bembenek BM, Joshy A, Offer SM. Lethal 5-Fluorouracil Toxicity in a Carrier of DPYD c.704G>A (p.R235Q). JCO Precis Oncol 2024; 8:e2400294. [PMID: 39576955 DOI: 10.1200/po.24.00294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/17/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Affiliation(s)
- Brianna M Bembenek
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
| | - Aneena Joshy
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
- Charles University, Prague, Czechia
| | - Steven M Offer
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
45
|
van den Wildenberg SAH, Genet SAAM, Streng AS, Broeren MAC, Deenen MJ, van Dongen JLJ, Brunsveld L, Scharnhorst V, van de Kerkhof D. Partial protein binding of uracil and thymine affects accurate dihydropyrimidine dehydrogenase (DPD) phenotyping. J Pharm Biomed Anal 2024; 249:116381. [PMID: 39067280 DOI: 10.1016/j.jpba.2024.116381] [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: 03/20/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Fluorouracil is among the most used antimetabolite drugs for the chemotherapeutic treatment of various types of gastrointestinal malignancies. Dihydropyrimidine dehydrogenase (DPYD) genotyping prior to fluorouracil treatment is considered standard practice in most European countries. Yet, current pre-therapeutic DPYD genotyping procedures do not identify all dihydropyrimidine dehydrogenase (DPD)-deficient patients. Alternatively, DPD activity can be estimated by determining the DPD phenotype by quantification of plasma concentrations of the endogenous uracil and thymine concentrations and their respective metabolites dihydrouracil (DHU) and dihydrothymine (DHT). Liquid chromatography - mass spectrometry (LC-MS) detection is currently considered as the most adequate method for quantification of low-molecular weight molecules, although the sample preparation method is highly critical for analytical outcome. It was hypothesized that during protein precipitation, the recovery of the molecule of interest highly depends on the choice of precipitation agent and the extent of protein binding in plasma. In this work, the effect of protein precipitation using acetonitrile (ACN) compared to strong acid perchloric acid (PCA) on the recovery of uracil, thymine, DHU and DHT is demonstrated. Upon the analysis of plasma samples, PCA precipitation showed higher concentrations of uracil and thymine as compared to ACN precipitation. Using ultrafiltration, it was shown that uracil and thymine are significantly (60-65 %) bound to proteins compared to DHU and DHT. This shows that before harmonized cut-off levels of DPD phenotyping can be applied in clinical practice, the analytical methodology requires extensive further optimization.
Collapse
Affiliation(s)
- Sebastian A H van den Wildenberg
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Clinical Laboratory, Catharina Hospital Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands
| | - Sylvia A A M Genet
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Clinical Laboratory, Catharina Hospital Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands
| | - Alexander S Streng
- Clinical Laboratory, Catharina Hospital Eindhoven, the Netherlands; Clinical Laboratory Bernhoven, Siemens Healthineers, Uden, the Netherlands
| | - Maarten A C Broeren
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands; Clinical Laboratory, Máxima Medical Center, Veldhoven, Eindhoven, the Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, the Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Joost L J van Dongen
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands
| | - Volkher Scharnhorst
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Clinical Laboratory, Catharina Hospital Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands
| | - Daan van de Kerkhof
- Laboratory of Chemical Biology, department of Biomedical Engineering, Eindhoven University of Technology, the Netherlands; Clinical Laboratory, Catharina Hospital Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, the Netherlands.
| |
Collapse
|
46
|
Pratt VM, Cavallari LH, Fulmer ML, Gaedigk A, Hachad H, Ji Y, Kalman LV, Ly RC, Moyer AM, Scott SA, Turner AJ, van Schaik RHN, Whirl-Carrillo M, Weck KE. DPYD Genotyping Recommendations: A Joint Consensus Recommendation of the Association for Molecular Pathology, American College of Medical Genetics and Genomics, Clinical Pharmacogenetics Implementation Consortium, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, European Society for Pharmacogenomics and Personalized Therapy, Pharmacogenomics Knowledgebase, and Pharmacogene Variation Consortium. J Mol Diagn 2024; 26:851-863. [PMID: 39032821 DOI: 10.1016/j.jmoldx.2024.05.015] [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: 04/09/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 07/23/2024] Open
Abstract
The goals of the Association for Molecular Pathology Clinical Practice Committee's Pharmacogenomics (PGx) Working Group are to define the key attributes of pharmacogenetic alleles recommended for clinical testing and a minimum set of variants that should be included in clinical PGx genotyping assays. This document series provides recommendations for a minimum set of variant alleles (tier 1) and an extended list of variant alleles (tier 2) that will aid clinical laboratories when designing assays for PGx testing. The Association for Molecular Pathology PGx Working Group considered the functional impact of the variant alleles, allele frequencies in multiethnic populations, the availability of reference materials, and other technical considerations for PGx testing when developing these recommendations. The goal of this Working Group is to promote standardization of PGx testing across clinical laboratories. This document will focus on clinical DPYD PGx testing that may be applied to all dihydropyrimidine dehydrogenase-related medications. These recommendations are not to be interpreted as prescriptive but to provide a reference guide.
Collapse
Affiliation(s)
- Victoria M Pratt
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Agena Bioscience, San Diego, California.
| | - Larisa H Cavallari
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, Florida
| | - Makenzie L Fulmer
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and ARUP Laboratories, University of Utah School of Medicine, Salt Lake City, Utah
| | - Andrea Gaedigk
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri; School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Houda Hachad
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Clinical Operations, AccessDx, Houston, Texas
| | - Yuan Ji
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and ARUP Laboratories, University of Utah School of Medicine, Salt Lake City, Utah
| | - Lisa V Kalman
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Division of Laboratory Systems, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Reynold C Ly
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ann M Moyer
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Stuart A Scott
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Stanford University, Stanford, California; Clinical Genomics Laboratory, Stanford Medicine, Palo Alto, California
| | - Amy J Turner
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pediatrics, Children's Research Institute, The Medical College of Wisconsin, Milwaukee, Wisconsin; RPRD Diagnostics LLC, Wauwatosa, Wisconsin
| | - Ron H N van Schaik
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Clinical Chemistry/International Federation of Clinical Chemistry and Laboratory Medicine Expert Center Pharmacogenetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Michelle Whirl-Carrillo
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Biomedical Data Science, Stanford University, Stanford, California
| | - Karen E Weck
- Pharmacogenomics Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina; Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
47
|
Shugg T, Tillman EM, Breman AM, Hodge JC, McDonald CA, Ly RC, Rowe EJ, Osei W, Smith TB, Schwartz PH, Callaghan JT, Pratt VM, Lynch S, Eadon MT, Skaar TC. Development of a Multifaceted Program for Pharmacogenetics Adoption at an Academic Medical Center: Practical Considerations and Lessons Learned. Clin Pharmacol Ther 2024; 116:914-931. [PMID: 39169556 PMCID: PMC11452286 DOI: 10.1002/cpt.3402] [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: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
In 2019, Indiana University launched the Precision Health Initiative to enhance the institutional adoption of precision medicine, including pharmacogenetics (PGx) implementation, at university-affiliated practice sites across Indiana. The overarching goal of this PGx implementation program was to facilitate the sustainable adoption of genotype-guided prescribing into routine clinical care. To accomplish this goal, we pursued the following specific objectives: (i) to integrate PGx testing into existing healthcare system processes; (ii) to implement drug-gene pairs with high-level evidence and educate providers and pharmacists on established clinical management recommendations; (iii) to engage key stakeholders, including patients to optimize the return of results for PGx testing; (iv) to reduce health disparities through the targeted inclusion of underrepresented populations; (v) and to track third-party reimbursement. This tutorial details our multifaceted PGx implementation program, including descriptions of our interventions, the critical challenges faced, and the major program successes. By describing our experience, we aim to assist other clinical teams in achieving sustainable PGx implementation in their health systems.
Collapse
Affiliation(s)
- Tyler Shugg
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Emma M. Tillman
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amy M. Breman
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jennelle C. Hodge
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christine A. McDonald
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reynold C. Ly
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth J. Rowe
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wilberforce Osei
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tayler B. Smith
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter H. Schwartz
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - John T. Callaghan
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Victoria M. Pratt
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheryl Lynch
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael T. Eadon
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
48
|
Gaedigk A, Turner AJ, Moyer AM, Zubiaur P, Boone EC, Wang WY, Broeckel U, Kalman LV. Characterization of Reference Materials for DPYD: A GeT-RM Collaborative Project. J Mol Diagn 2024; 26:864-875. [PMID: 39032822 PMCID: PMC11818935 DOI: 10.1016/j.jmoldx.2024.06.004] [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: 05/01/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/23/2024] Open
Abstract
The DPYD gene encodes dihydropyrimidine dehydrogenase (DPD), which is involved in the catalysis of uracil and thymine, as well as 5-fluorouracil (5-FU), which is used to treat solid tumors. Patients with decreased DPD activity are at risk of serious, sometimes fatal, adverse drug reactions to this important cancer drug. Pharmacogenetic testing for DPYD is increasingly provided by clinical and research laboratories; however, only a limited number of quality control and reference materials are currently available for clinical DPYD testing. To address this need, the Division of Laboratory Systems, Centers for Disease Control and Prevention-based Genetic Testing Reference Materials Coordination Program, in collaboration with members of the pharmacogenetic testing and research communities and the Coriell Institute for Medical Research, has characterized 33 DNA samples derived from Coriell cell lines for DPYD. Samples were distributed to four volunteer laboratories for genetic testing using a variety of commercially available and laboratory-developed tests. Sanger sequencing was used by one laboratory and publicly available whole-genome sequence data from the 1000 Genomes Project were used by another to inform genotype. Thirty-three distinct DPYD variants were identified among the 33 samples characterized. These publicly available and well-characterized materials can be used to support the quality assurance and quality control programs of clinical laboratories performing clinical pharmacogenetic testing.
Collapse
Affiliation(s)
- Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri
| | - Amy J Turner
- RPRD Diagnostics, Milwaukee, Wisconsin; Section on Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria de La Princesa, Madrid, Spain
| | - Erin C Boone
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri
| | - Wendy Y Wang
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri
| | - Ulrich Broeckel
- RPRD Diagnostics, Milwaukee, Wisconsin; Section on Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lisa V Kalman
- Division of Laboratory Systems, Centers for Disease Control and Prevention, Atlanta, Georgia.
| |
Collapse
|
49
|
Nogueiras-Álvarez R, Pérez Francisco I. Pharmacogenetics in Oncology: A useful tool for individualizing drug therapy. Br J Clin Pharmacol 2024; 90:2483-2508. [PMID: 39077855 DOI: 10.1111/bcp.16181] [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: 06/04/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
With the continuous development of genetics in healthcare, there has been a significant contribution to the development of precision medicine, which is ultimately aimed at improving the care of patients. Generally, drug treatments used in Oncology are characterized by a narrow therapeutic range and by their potential toxicity. Knowledge of pharmacogenomics and pharmacogenetics can be very useful in the area of Oncology, as they constitute additional tools that can help to individualize patients' treatment. This work includes a description of some genes that have been revealed to be useful in the field of Oncology, as they play a role in drug prescription and in the prediction of treatment response.
Collapse
Affiliation(s)
- Rita Nogueiras-Álvarez
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Basque Country Pharmacovigilance Unit, Galdakao, Bizkaia/Vizcaya, Spain
| | - Inés Pérez Francisco
- Breast Cancer Research Group, Bioaraba Health Research Institute, Vitoria-Gasteiz, Araba/Álava, Spain
| |
Collapse
|
50
|
Zhao R, Han M, Lin S, Lin Z, Yu M, Zhang B, Ma L, Li D, Peng L. Adverse drug events associated with fluorouracil use in patients with metastatic colorectal cancer: a real-world pharmacovigilance study based on the FDA adverse event reporting system. Expert Opin Drug Saf 2024; 23:1295-1307. [PMID: 39010662 DOI: 10.1080/14740338.2024.2380513] [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: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Fluorouracil (5-FU) is widely used to treat metastatic colorectal cancer (mCRC), but real-world safety data is limited. Our study aimed to evaluate 5-FU's safety profile in a large mCRC population using the FAERS database. RESEARCH DESIGN AND METHODS We conducted disproportionality analyses to identify adverse drug events associated with 5-FU use in mCRC patients from 2004 to 2023. Subgroup analyses, gender difference analyses, and logistic regression were also performed. RESULTS We identified 1,458 reports with 5-FU as the primary suspected drug, with males accounting for 48.8% of reports. Gastrointestinal disorders were the most common adverse event (864 cases), while pregnancy-related conditions showed the strongest signal intensity (ROR = 2.97). We found 19 preferred terms with positive signals, including ischemic hepatitis (ROR = 59.32), blood iron increased (ROR = 59.32), and stress cardiomyopathy (ROR = 51.94). Males were more susceptible to weight loss and skin toxicity. Most adverse events occurred within the first month of 5-FU administration. CONCLUSION Our study provides a comprehensive analysis of 5-FU's safety profile in mCRC patients, helping healthcare professionals mitigate risks in clinical practice.
Collapse
Affiliation(s)
- Ruiqi Zhao
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengyao Han
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Sen Lin
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Zhimei Lin
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengjiao Yu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bei Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Lanyue Ma
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Danfei Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Lisheng Peng
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|