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, the Pharmacogenomics Global Research Network Publication Committee. 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
|
Kapoor VS, Ciccolini J, Kapoor S. A Big Problem With a Feasible Solution, Not a Small Problem With a Complex Solution. JCO Oncol Pract 2025; 21:262-264. [PMID: 39626134 DOI: 10.1200/op-24-00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 02/13/2025] Open
Affiliation(s)
- Vimal Scott Kapoor
- Vimal Scott Kapoor, MD, BSc, MSc, CCFP-EM, FRCPC(PHPM), FRCPC(Occ Med), University of Toronto, Toronto, Canada; Joseph Ciccolini, PharmD, PhD, Aix Marseille Univ, Marseille, France; and Sunil Kapoor, MD, FRCPC, University of Toronto, Toronto, Canada
| | - Joseph Ciccolini
- Vimal Scott Kapoor, MD, BSc, MSc, CCFP-EM, FRCPC(PHPM), FRCPC(Occ Med), University of Toronto, Toronto, Canada; Joseph Ciccolini, PharmD, PhD, Aix Marseille Univ, Marseille, France; and Sunil Kapoor, MD, FRCPC, University of Toronto, Toronto, Canada
| | - Sunil Kapoor
- Vimal Scott Kapoor, MD, BSc, MSc, CCFP-EM, FRCPC(PHPM), FRCPC(Occ Med), University of Toronto, Toronto, Canada; Joseph Ciccolini, PharmD, PhD, Aix Marseille Univ, Marseille, France; and Sunil Kapoor, MD, FRCPC, University of Toronto, Toronto, Canada
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Mosch R, van der Lee M, Guchelaar HJ, Swen JJ. Pharmacogenetic Panel Testing: A Review of Current Practice and Potential for Clinical Implementation. Annu Rev Pharmacol Toxicol 2025; 65:91-109. [PMID: 39348848 DOI: 10.1146/annurev-pharmtox-061724-080935] [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] [Indexed: 10/02/2024]
Abstract
Pharmacogenetics (PGx) aims to optimize drug treatment outcomes by using a patient's genetic profile for individualized drug and dose selection. Currently, reactive and pretherapeutic single-gene PGx tests are increasingly applied in clinical practice in several countries and institutions. With over 95% of the population carrying at least one actionable PGx variant, and with drugs impacted by these genetic variants being in common use, pretherapeutic or preemptive PGx panel testing appears to be an attractive option for better-informed drug prescribing. Here, we discuss the current state of PGx panel testing and explore the potential for clinical implementation. We conclude that available evidence supports the implementation of pretherapeutic PGx panel testing for drugs covered in the PGx guidelines, yet identification of specific patient populations that benefit most and cost-effectiveness data are necessary to support large-scale implementation.
Collapse
Affiliation(s)
- R Mosch
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - M van der Lee
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - H J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Ardizzone A, Bulzomì M, De Luca F, Silvestris N, Esposito E, Capra AP. Dihydropyrimidine Dehydrogenase Polymorphism c.2194G>A Screening Is a Useful Tool for Decreasing Gastrointestinal and Hematological Adverse Drug Reaction Risk in Fluoropyrimidine-Treated Patients. Curr Issues Mol Biol 2024; 46:9831-9843. [PMID: 39329936 PMCID: PMC11430620 DOI: 10.3390/cimb46090584] [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: 07/18/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Although the risk of fluoropyrimidine toxicity may be decreased by identifying poor metabolizers with a preemptive dihydropyrimidine dehydrogenase (DPYD) test, following international standards, many patients with wild-type (WT) genotypes for classic variations may still exhibit adverse drug reactions (ADRs). Therefore, the safety of fluoropyrimidine therapy could be improved by identifying new DPYD polymorphisms associated with ADRs. This study was carried out to assess whether testing for the underestimated c.2194G>A (DPYD*6 polymorphism, rs1801160) is useful, in addition to other well-known variants, in reducing the risk of ADRs in patients undergoing chemotherapy treatment. This retrospective study included 132 patients treated with fluoropyrimidine-containing regimens who experienced ADRs such as gastrointestinal, dermatological, hematological, and neurological. All subjects were screened for DPYD variants DPYD2A (IVS14+1G>A, c.1905+1G>A, rs3918290), DPYD13 (c.1679T>G, rs55886062), c.2846A>T (rs67376798), c.1236G>A (rs56038477), and c.2194G>A by real-time polymerase chain reaction (RT-PCR). In this cohort, the heterozygous c.2194G>A variant was present in 26 patients, while 106 individuals were WT; both subgroups were compared for the incidence of ADRs. This assessment revealed a high incidence of gastrointestinal and hematological ADRs in DPYD6 carriers compared to WT. Moreover, we have shown a higher prevalence of ADRs in females compared to males when stratifying c.2194G>A carrier individuals. Considering that c.2194G>A was linked to clinically relevant ADRs, we suggest that this variant should also be assessed preventively to reduce the risk of fluoropyrimidine-related ADRs.
Collapse
Affiliation(s)
- Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.A.); (M.B.); (F.D.L.); (A.P.C.)
| | - Maria Bulzomì
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.A.); (M.B.); (F.D.L.); (A.P.C.)
| | - Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.A.); (M.B.); (F.D.L.); (A.P.C.)
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy;
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.A.); (M.B.); (F.D.L.); (A.P.C.)
- Genetics and Pharmacogenetics Unit, “Gaetano Martino” University Hospital, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.A.); (M.B.); (F.D.L.); (A.P.C.)
| |
Collapse
|
13
|
Ikonnikova A, Fedorinov D, Gryadunov D, Heydarov R, Lyadova M, Moskalenko A, Mikhailovich V, Emelyanova M, Lyadov V. MIR27A Gene Polymorphism Modifies the Effect of Common DPYD Gene Variants on Severe Toxicity in Patients with Gastrointestinal Tumors Treated with Fluoropyrimidine-Based Anticancer Therapy. Int J Mol Sci 2024; 25:8503. [PMID: 39126072 PMCID: PMC11313059 DOI: 10.3390/ijms25158503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
To reduce severe fluoropyrimidine-related toxicity, pharmacogenetic guidelines recommend a dose reduction for carriers of four high-risk variants in the DPYD gene (*2A, *13, c.2846A>T, HapB3). The polymorphism in the MIR27A gene has been shown to enhance the predictive value of these variants. Our study aimed to explore whether rs895819 in the MIR27A gene modifies the effect of five common DPYD variants: c.1129-5923C>G (rs75017182, HapB3), c.2194G>A (rs1801160, *6), c.1601G>A (rs1801158, *4), c.496A>G (rs2297595), and c.85T>C (rs1801265, *9A). The study included 370 Caucasian patients with gastrointestinal tumors who received fluoropyrimidine-containing chemotherapy. Genotyping was performed using high-resolution melting analysis. The DPYD*6 allele was associated with overall severe toxicity and neutropenia with an increased risk particularly pronounced in patients carrying the MIR27A variant. All carriers of DPYD*6 exhibited an association with asthenia regardless of their MIR27A status. The increased risk of neutropenia in patients with c.496G was only evident in those co-carrying the MIR27A variant. DPYD*4 was also significantly linked to neutropenia risk in co-carriers of the MIR27A variant. Thus, we have demonstrated the predictive value of the *6, *4, and c.496G alleles of the DPYD gene, considering the modifying effect of the MIR27A polymorphism.
Collapse
Affiliation(s)
- Anna Ikonnikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (M.E.)
| | - Denis Fedorinov
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, 117152 Moscow, Russia; (D.F.); (M.L.); (A.M.); (V.L.)
- Department of Oncology and Palliative Medicine Named after Academician A.I. Savitsky, Russian Medical Academy of Continuous Professional Education, 123242 Moscow, Russia
| | - Dmitry Gryadunov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (M.E.)
| | - Rustam Heydarov
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (R.H.); (V.M.)
| | - Marina Lyadova
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, 117152 Moscow, Russia; (D.F.); (M.L.); (A.M.); (V.L.)
- Department of Oncology, Novokuznetsk State Institute for Postgraduate Medical Education, 654005 Novokuznetsk, Russia
| | - Alexey Moskalenko
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, 117152 Moscow, Russia; (D.F.); (M.L.); (A.M.); (V.L.)
| | - Vladimir Mikhailovich
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (R.H.); (V.M.)
| | - Marina Emelyanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (M.E.)
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (R.H.); (V.M.)
| | - Vladimir Lyadov
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, 117152 Moscow, Russia; (D.F.); (M.L.); (A.M.); (V.L.)
- Department of Oncology and Palliative Medicine Named after Academician A.I. Savitsky, Russian Medical Academy of Continuous Professional Education, 123242 Moscow, Russia
- Department of Oncology, Novokuznetsk State Institute for Postgraduate Medical Education, 654005 Novokuznetsk, Russia
| |
Collapse
|
14
|
Tamraz B, Venook AP. DPYD Pharmacogenetics: To Opt-in or to Opt-out. JCO Oncol Pract 2024; 20:1009-1011. [PMID: 38743915 DOI: 10.1200/op.24.00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Bani Tamraz
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA
| | - Alan P Venook
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
15
|
Jacobson JO, Rompelman G, Chen A, Morrison-Ma S, Murray L, Ferzoco M, Bunnell C, Wagner AJ, Roberts D, Chan J, Block C, Rubinson D. Design and Implementation of an Opt-Out, End-to-End, Preemptive DPYD Testing Program for Patients Planned for a Systemic Fluoropyrimidine. JCO Oncol Pract 2024; 20:1115-1122. [PMID: 38608224 DOI: 10.1200/op.23.00776] [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: 12/03/2023] [Revised: 02/11/2024] [Accepted: 02/27/2024] [Indexed: 04/14/2024] Open
Abstract
PURPOSE Several allelic variants of the gene DPYD encoding dihydropyrimidine dehydrogenase (DPD) are associated with impaired metabolism of the systemic fluoropyrimidine fluorouracil (5FU) and its oral prodrug, capecitabine, which elevates the risk for severe toxicity. Following a patient death related to capecitabine toxicity in which DPD deficiency was suspected, a multidisciplinary advisory panel was convened to develop an institution-wide approach to future patients planned for a systemic fluoropyrimidine. METHODS The panel selected an opt-out testing strategy which focused on developing reliable processes to collect and report test results and targeted education. An electronic health record-based automated reminder was designed to activate when a 5FU- or capecitabine-containing chemotherapy regimen was ordered for a patient without prior exposure to either agent and without a prior DPYD sequencing test result. DPYD testing was standardized across all sites of care, and a closed loop reporting system for abnormal test results was created. Before implementation, targeted education was provided to providers, pharmacists, and nurses, and a failure mode and effects analysis was performed. Program rollout was staged over a 6-month period. RESULTS At 10 months, the rate of preemptive testing increased from a baseline of 26% to a sustained rate of >90%. In the six network sites, the testing rate increased from 9% to 96%. A total of 1,043 patients have been tested preemptively; allelic variants have been identified in 43 (4.1%). Among 25 evaluable patients, dose reduction or change to a non-fluoropyrimidine-based regimen was accomplished in 96%. CONCLUSION Preemptive DPYD testing is feasible, and high rates of testing can be achieved using an opt-out, reminder-based program. We provide the details of the implementation and encourage others to emulate it.
Collapse
Affiliation(s)
| | | | - Angela Chen
- Children's Specialized Hospital, New Brunswick, NJ
| | | | | | | | - Craig Bunnell
- Dana-Farber Cancer, Boston, MA
- Harvard Medical School, Boston, MA
| | - Andrew J Wagner
- Dana-Farber Cancer, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Jennifer Chan
- Dana-Farber Cancer, Boston, MA
- Harvard Medical School, Boston, MA
| | - Caroline Block
- Dana-Farber Cancer, Boston, MA
- Harvard Medical School, Boston, MA
| | - Douglas Rubinson
- Dana-Farber Cancer, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
El Rouby N, Allen JD, Muldoon M, Beck M, Hesse K, Sebree N, Yoder R, Ritter S, Alqahtani Z, Grund J, Holbrook BP. Reply to "A call for reporting of tumor-specific outcomes in studies of DPYD genotyping". Clin Transl Sci 2024; 17:e70008. [PMID: 39172770 PMCID: PMC11340691 DOI: 10.1111/cts.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Affiliation(s)
- Nihal El Rouby
- St Elizabeth HealthcareEdgewoodKentuckyUSA
- Department of Pharmacy Practice and Administrative Sciences, James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOhioUSA
| | | | - Megan Muldoon
- Swedish Hospital, Part of Endeavor HealthChicagoIllinoisUSA
| | | | | | - Nichlas Sebree
- University of Cincinnati Medical CenterCincinnatiOhioUSA
| | | | | | - Zuhair Alqahtani
- Department of Pharmacy Practice and Administrative Sciences, James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOhioUSA
| | | | | |
Collapse
|
17
|
Tracksdorf T, Smith DM, Pearse S, Cicali EJ, Aquilante CL, Scott SA, Ho TT, Patel JN, Hicks JK, Hertz DL. Strategies for DPYD testing prior to fluoropyrimidine chemotherapy in the US. Support Care Cancer 2024; 32:497. [PMID: 38980476 DOI: 10.1007/s00520-024-08674-1] [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: 04/02/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE Patients with dihydropyrimidine dehydrogenase (DPD) deficiency are at high risk for severe and fatal toxicity from fluoropyrimidine (FP) chemotherapy. Pre-treatment DPYD testing is standard of care in many countries, but not the United States (US). This survey assessed pre-treatment DPYD testing approaches in the US to identify best practices for broader adoption. METHODS From August to October 2023, a 22-item QualtricsXM survey was sent to institutions and clinicians known to conduct pre-treatment DPYD testing and broadly distributed through relevant organizations and social networks. Responses were analyzed using descriptive analysis. RESULTS Responses from 24 unique US sites that have implemented pre-treatment DPYD testing or have a detailed implementation plan in place were analyzed. Only 33% of sites ordered DPYD testing for all FP-treated patients; at the remaining sites, patients were tested depending on disease characteristics or clinician preference. Almost 50% of sites depend on individual clinicians to remember to order testing without the assistance of electronic alerts or workflow reminders. DPYD testing was most often conducted by commercial laboratories that tested for at least the four or five DPYD variants considered clinically actionable. Approximately 90% of sites reported receiving results within 10 days of ordering. CONCLUSION Implementing DPYD testing into routine clinical practice is feasible and requires a coordinated effort among the healthcare team. These results will be used to develop best practices for the clinical adoption of DPYD testing to prevent severe and fatal toxicity in cancer patients receiving FP chemotherapy.
Collapse
Affiliation(s)
- Tabea Tracksdorf
- Deparment of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church St, Room 2560C, Ann Arbor, MI, 48109-1065, USA
| | - D Max Smith
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
- MedStar Health, Columbia, MD, USA
| | - Skyler Pearse
- Health Behavior and Health Education, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Emily J Cicali
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, USA
| | - Christina L Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stuart A Scott
- Department of Pathology, Stanford University, Stanford, CA, USA
- Clinical Genomics Laboratory, Stanford Medicine, Palo Alto, CA, USA
| | - Teresa T Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jai N Patel
- Department of Cancer Pharmacology and Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC, USA
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - J Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
| | - Daniel L Hertz
- Deparment of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church St, Room 2560C, Ann Arbor, MI, 48109-1065, USA.
| |
Collapse
|
18
|
Nguyen DG, Morris SA, Hamilton A, Kwange SO, Steuerwald N, Symanowski J, Moore DC, Hanson S, Mroz K, Lopes KE, Larck C, Musselwhite L, Kadakia KC, Koya B, Chai S, Osei-Boateng K, Kalapurakal S, Swift K, Hwang J, Patel JN. Real-World Impact of an In-House Dihydropyrimidine Dehydrogenase ( DPYD) Genotype Test on Fluoropyrimidine Dosing, Toxicities, and Hospitalizations at a Multisite Cancer Center. JCO Precis Oncol 2024; 8:e2300623. [PMID: 38935897 PMCID: PMC11371106 DOI: 10.1200/po.23.00623] [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: 11/09/2023] [Revised: 03/07/2024] [Accepted: 04/12/2024] [Indexed: 06/29/2024] Open
Abstract
PURPOSE Fluoropyrimidine-related toxicity and mortality risk increases significantly in patients carrying certain DPYD genetic variants with standard dosing. We implemented DPYD genotyping at a multisite cancer center and evaluated its impact on dosing, toxicity, and hospitalization. METHODS In this prospective observational study, patients receiving (reactive) or planning to receive (pretreatment) fluoropyrimidine-based chemotherapy were genotyped for five DPYD variants as standard practice per provider discretion. The primary end point was the proportion of variant carriers receiving fluoropyrimidine modifications. Secondary end points included mean relative dose intensity, fluoropyrimidine-related grade 3+ toxicities, and hospitalizations. Fisher's exact test compared toxicity and hospitalization rates between pretreatment carriers, reactive carriers, and wild-type patients. Univariable and multivariable logistic regression identified factors associated with toxicity and hospitalization risk. Kaplan-Meier methods estimated time to event of first grade 3+ toxicity and hospitalization. RESULTS Of the 757 patients who received DPYD genotyping (median age 63, 54% male, 74% White, 19% Black, 88% GI malignancy), 45 (5.9%) were heterozygous carriers. Fluoropyrimidine was modified in 93% of carriers who started treatment. In 442 patients with 3-month follow-up, 64%, 31%, and 30% of reactive carriers, pretreatment carriers, and wild-type patients had grade 3+ toxicity, respectively (P = .085); 64%, 25%, and 13% were hospitalized (P < .001). Reactive carriers had 10-fold higher odds of hospitalization compared with wild-type patients (P = .001), whereas no significant difference was noted between pretreatment carriers and wild-type patients. Time-to-event of toxicity and hospitalization were significantly different between genotype groups (P < .001), with reactive carriers having the earliest onset and highest incidence. CONCLUSION DPYD genotyping prompted fluoropyrimidine modifications in most carriers. Pretreatment testing reduced toxicities and hospitalizations compared with reactive testing, thus normalizing the risk to that of wild-type patients, and should be considered standard practice.
Collapse
Affiliation(s)
- D. Grace Nguyen
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Sarah A. Morris
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Alicia Hamilton
- Molecular Biology and Genomics Core Facility, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Simeon O. Kwange
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Nury Steuerwald
- Molecular Biology and Genomics Core Facility, Atrium Health Levine Cancer Institute, Charlotte, NC
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - James Symanowski
- Department of Biostatistics and Data Sciences, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Donald C. Moore
- Department of Pharmacy, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Sarah Hanson
- Department of Pharmacy, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Kaitlyn Mroz
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Karine E. Lopes
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Chris Larck
- Department of Pharmacy, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Laura Musselwhite
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Kunal C. Kadakia
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Brinda Koya
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Seungjean Chai
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Kwabena Osei-Boateng
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Sini Kalapurakal
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Kristen Swift
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Jimmy Hwang
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
- Department of Solid Tumor Oncology, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Jai N. Patel
- Department of Cancer Pharmacology & Pharmacogenomics, Atrium Health Levine Cancer Institute, Charlotte, NC
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| |
Collapse
|
19
|
van Kuilenburg ABP, Pleunis-van Empel MCH, Brouwer RB, Sijben AEJ, Knapen DG, Oude Munnink TH, van Zanden JJ, Janssens-Puister J, Dobritzsch D, Meinsma R, Meijer-Jansen J, van Dooren SJM, Vijzelaar R, Pop A, Salomons GS, Maring JG, Niezen-Koning KE. Lethal Capecitabine Toxicity in Patients With Complete Dihydropyrimidine Dehydrogenase Deficiency Due to Ultra-Rare DPYD Variants. JCO Precis Oncol 2024; 8:e2300599. [PMID: 38709992 DOI: 10.1200/po.23.00599] [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: 10/31/2023] [Revised: 12/14/2023] [Accepted: 03/19/2024] [Indexed: 05/08/2024] Open
Abstract
A DPD deficiency should be considered in case of severe toxicity even in the absence of common risk variants in DPYD.
Collapse
Affiliation(s)
- André B P van Kuilenburg
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | | | - Rick B Brouwer
- Department of Clinical Chemistry and Laboratory Medicine, Medisch Spectrum Twente, Medlon BV, Enschede, the Netherlands
| | | | - Daan G Knapen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Thijs H Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Jelmer J van Zanden
- Martini Hospital Groningen, Certe Department of Clinical Chemistry, the Netherlands
| | - Jenny Janssens-Puister
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Doreen Dobritzsch
- Department of Chemistry, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Rutger Meinsma
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | - Judith Meijer-Jansen
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | - Silvy J M van Dooren
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | | | - Ana Pop
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | - Gajja S Salomons
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, Amsterdam, the Netherlands
| | - Jan Gerard Maring
- Departments of Clinical Pharmacy and Medical Oncology, Isala, Zwolle, the Netherlands
| | - Klary E Niezen-Koning
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
20
|
Lingaratnam S, Shah M, Nicolazzo J, Michael M, Seymour JF, James P, Lazarakis S, Loi S, Kirkpatrick CMJ. A systematic review and meta-analysis of the impacts of germline pharmacogenomics on severe toxicity and symptom burden in adult patients with cancer. Clin Transl Sci 2024; 17:e13781. [PMID: 38700261 PMCID: PMC11067509 DOI: 10.1111/cts.13781] [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: 12/08/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 05/05/2024] Open
Abstract
The clinical application of Pharmacogenomics (PGx) has improved patient safety. However, comprehensive PGx testing has not been widely adopted in clinical practice, and significant opportunities exist to further optimize PGx in cancer care. This systematic review and meta-analysis aim to evaluate the safety outcomes of reported PGx-guided strategies (Analysis 1) and identify well-studied emerging pharmacogenomic variants that predict severe toxicity and symptom burden (Analysis 2) in patients with cancer. We searched MEDLINE, EMBASE, CENTRAL, clinicaltrials.gov, and International Clinical Trials Registry Platform from inception to January 2023 for clinical trials or comparative studies evaluating PGx strategies or unconfirmed pharmacogenomic variants. The primary outcomes were severe adverse events (SAE; ≥ grade 3) or symptom burden with pain and vomiting as defined by trial protocols and assessed by trial investigators. We calculated pooled overall relative risk (RR) and 95% confidence interval (95%CI) using random effects models. PROSPERO, registration number CRD42023421277. Of 6811 records screened, six studies were included for Analysis 1, 55 studies for Analysis 2. Meta-analysis 1 (five trials, 1892 participants) showed a lower absolute incidence of SAEs with PGx-guided strategies compared to usual therapy, 16.1% versus 34.0% (RR = 0.72, 95%CI 0.57-0.91, p = 0.006, I2 = 34%). Meta-analyses 2 identified nine medicine(class)-variant pairs of interest across the TYMS, ABCB1, UGT1A1, HLA-DRB1, and OPRM1 genes. Application of PGx significantly reduced rates of SAEs in patients with cancer. Emergent medicine-variant pairs herald further research into the expansion and optimization of PGx to improve systemic anti-cancer and supportive care medicine safety and efficacy.
Collapse
Affiliation(s)
- Senthil Lingaratnam
- Pharmacy DepartmentPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Mahek Shah
- Faculty of Pharmacy and Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Joseph Nicolazzo
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Michael Michael
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - John F. Seymour
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Clinical HaematologyPeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
| | - Paul James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
| | - Smaro Lazarakis
- Health Sciences LibraryRoyal Melbourne HospitalMelbourneVictoriaAustralia
| | - Sherene Loi
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Carl M. J. Kirkpatrick
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
21
|
Shriver SP, Adams D, McKelvey BA, McCune JS, Miles D, Pratt VM, Ashcraft K, McLeod HL, Williams H, Fleury ME. Overcoming Barriers to Discovery and Implementation of Equitable Pharmacogenomic Testing in Oncology. J Clin Oncol 2024; 42:1181-1192. [PMID: 38386947 PMCID: PMC11003514 DOI: 10.1200/jco.23.01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/08/2023] [Accepted: 12/12/2023] [Indexed: 02/24/2024] Open
Abstract
Pharmacogenomics (PGx), the study of inherited genomic variation and drug response or safety, is a vital tool in precision medicine. In oncology, testing to identify PGx variants offers patients the opportunity for customized treatments that can minimize adverse effects and maximize the therapeutic benefits of drugs used for cancer treatment and supportive care. Because individuals of shared ancestry share specific genetic variants, PGx factors may contribute to outcome disparities across racial and ethnic categories when genetic ancestry is not taken into account or mischaracterized in PGx research, discovery, and application. Here, we examine how the current scientific understanding of the role of PGx in differential oncology safety and outcomes may be biased toward a greater understanding and more complete clinical implementation of PGx for individuals of European descent compared with other genetic ancestry groups. We discuss the implications of this bias for PGx discovery, access to care, drug labeling, and patient and provider understanding and use of PGx approaches. Testing for somatic genetic variants is now the standard of care in treatment of many solid tumors, but the integration of PGx into oncology care is still lacking despite demonstrated actionable findings from PGx testing, reduction in avoidable toxicity and death, and return on investment from testing. As the field of oncology is poised to expand and integrate germline genetic variant testing, it is vital that PGx discovery and application are equitable for all populations. Recommendations are introduced to address barriers to facilitate effective and equitable PGx application in cancer care.
Collapse
Affiliation(s)
| | | | | | - Jeannine S McCune
- City of Hope/Beckman Research Institute Department of Hematologic Malignancies Translational Sciences, Duarte, CA
| | | | | | | | | | | | | |
Collapse
|
22
|
Le Teuff G, Cozic N, Boyer JC, Boige V, Diasio RB, Taieb J, Meulendijks D, Palles C, Schwab M, Deenen M, Largiadèr CR, Marinaki A, Jennings BA, Wettergren Y, Di Paolo A, Gross E, Budai B, Ackland SP, van Kuilenburg ABP, McLeod HL, Milano G, Thomas F, Loriot MA, Kerr D, Schellens JHM, Laurent-Puig P, Shi Q, Pignon JP, Etienne-Grimaldi MC. Dihydropyrimidine dehydrogenase gene variants for predicting grade 4-5 fluoropyrimidine-induced toxicity: FUSAFE individual patient data meta-analysis. Br J Cancer 2024; 130:808-818. [PMID: 38225422 PMCID: PMC10912560 DOI: 10.1038/s41416-023-02517-2] [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: 07/12/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Dihydropyrimidine dehydrogenase (DPD) deficiency is the main known cause of life-threatening fluoropyrimidine (FP)-induced toxicities. We conducted a meta-analysis on individual patient data to assess the contribution of deleterious DPYD variants *2A/D949V/*13/HapB3 (recommended by EMA) and clinical factors, for predicting G4-5 toxicity. METHODS Study eligibility criteria included recruitment of Caucasian patients without DPD-based FP-dose adjustment. Main endpoint was 12-week haematological or digestive G4-5 toxicity. The value of DPYD variants *2A/p.D949V/*13 merged, HapB3, and MIR27A rs895819 was evaluated using multivariable logistic models (AUC). RESULTS Among 25 eligible studies, complete clinical variables and primary endpoint were available in 15 studies (8733 patients). Twelve-week G4-5 toxicity prevalence was 7.3% (641 events). The clinical model included age, sex, body mass index, schedule of FP-administration, concomitant anticancer drugs. Adding *2A/p.D949V/*13 variants (at least one allele, prevalence 2.2%, OR 9.5 [95%CI 6.7-13.5]) significantly improved the model (p < 0.0001). The addition of HapB3 (prevalence 4.0%, 98.6% heterozygous), in spite of significant association with toxicity (OR 1.8 [95%CI 1.2-2.7]), did not improve the model. MIR27A rs895819 was not associated with toxicity, irrespective of DPYD variants. CONCLUSIONS FUSAFE meta-analysis highlights the major relevance of DPYD *2A/p.D949V/*13 combined with clinical variables to identify patients at risk of very severe FP-related toxicity.
Collapse
Affiliation(s)
- Gwénaël Le Teuff
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Oncostat U1018 INSERM, labeled Ligue Contre le Cancer, Université Paris-Saclay, Villejuif, France.
| | - Nathalie Cozic
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Oncostat U1018 INSERM, labeled Ligue Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | | | - Valérie Boige
- Department of cancer medicine, Gustave-Roussy Cancer Campus, Paris-Saclay and Paris-Sud Universities, Villejuif, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC, 5096, Paris, France
| | - Robert B Diasio
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Julien Taieb
- Université Paris-Cité, SIRIC CARPEM, Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP, Paris, France
| | - Didier Meulendijks
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Claire Palles
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence IFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72074, Tübingen, Germany
| | - Maarten Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, the Netherlands
| | - Carlo R Largiadèr
- Department of Clinical Chemistry, Bern University Hospital, University of Bern, Inselspital, Bern, Switzerland
| | | | | | | | - Antonello Di Paolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eva Gross
- LMU Munich, University Hospital, Campus Grosshadern, Munich, Germany
| | - Barna Budai
- National Institute of Oncology, Budapest, Hungary
| | - Stephen P Ackland
- College of Heath, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - André B P van Kuilenburg
- Amsterdam UMC, location University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Imaging and biomarkers, Amsterdam, The Netherlands
| | - Howard L McLeod
- Intermountain Precision Genomics, Intermountain Healthcare, St George, UT, USA
| | - Gérard Milano
- Oncopharmacology Laboratory, Centre Antoine Lacassagne, Nice, France
| | - Fabienne Thomas
- Institut Claudius Regaud, IUCT-Oncopôle and CRCT, University of Toulouse, Inserm, Toulouse, France
| | - Marie-Anne Loriot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC, 5096, Paris, France
- Hôpital Européen Georges Pompidou, Hôpitaux Universitaires Paris Ouest, Paris, France
| | - David Kerr
- Nuffield Division of Clinical and Laboratory Sciences and University of Oxford, Oxford, UK
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC, 5096, Paris, France
- Hôpital Européen Georges Pompidou, Hôpitaux Universitaires Paris Ouest, Paris, France
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jean-Pierre Pignon
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Oncostat U1018 INSERM, labeled Ligue Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | | |
Collapse
|
23
|
Turner AJ, Haidar CE, Yang W, Boone EC, Offer SM, Empey PE, Haddad A, Tahir S, Scharer G, Broeckel U, Gaedigk A. Updated DPYD HapB3 haplotype structure and implications for pharmacogenomic testing. Clin Transl Sci 2024; 17:e13699. [PMID: 38129972 PMCID: PMC10777430 DOI: 10.1111/cts.13699] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The DPYD gene encodes dihydropyrimidine dehydrogenase, the rate-limiting enzyme for the metabolism of fluoropyrimidines 5-fluorouracil and capecitabine. Genetic variants in DPYD have been associated with altered enzyme activity, therefore accurate detection and interpretation is critical to predict metabolizer status for individualized fluoropyrimidine therapy. The most commonly observed deleterious variation is the causal variant linked to the previously described HapB3 haplotype, c.1129-5923C>G (rs75017182) in intron 10, which introduces a cryptic splice site. A benign synonymous variant in exon 11, c.1236G>A (rs56038477) is also linked to HapB3 and is commonly used for testing. Previously, these single-nucleotide polymorphisms (SNPs) have been reported to be in perfect linkage disequilibrium (LD); therefore, c.1236G>A is often utilized as a proxy for the function-altering intronic variant. Clinical genotyping of DPYD identified a patient who had c.1236G>A, but not c.1129-5923C>G, suggesting that these two SNPs may not be in perfect LD, as previously assumed. Additional individuals with c.1236G>A, but not c.1129-5923C>G, were identified in the Children's Mercy Data Warehouse and the All of Us Research Program version 7 cohort substantiating incomplete SNP linkage. Consequently, testing only c.1236G>A can generate false-positive results in some cases and lead to suboptimal dosing that may negatively impact patient therapy and prospect of survival. Our data show that DPYD genotyping should include the functional variant c.1129-5923C>G, and not the c.1236G>A proxy, to accurately predict DPD activity.
Collapse
Affiliation(s)
| | - Cyrine E. Haidar
- Department of Pharmacy and Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Erin C. Boone
- Division of Clinical Pharmacology, Toxicology & Therapeutic InnovationChildren's Mercy Research Institute (CMRI)Kansas CityMissouriUSA
| | - Steven M. Offer
- Department of Molecular Pharmacology and Experimental TherapeuticsMayo ClinicRochesterMinnesotaUSA
| | - Philip E. Empey
- Department of Pharmacy and TherapeuticsUniversity of Pittsburgh School of PharmacyPittsburghPennsylvaniaUSA
| | - Andrew Haddad
- Department of Pharmaceutical SciencesUniversity of Pittsburgh School of PharmacyPittsburghPennsylvaniaUSA
| | - Saba Tahir
- Medical College of Wisconsin, School of PharmacyMilwaukeeWisconsinUSA
| | | | | | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic InnovationChildren's Mercy Research Institute (CMRI)Kansas CityMissouriUSA
- School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| |
Collapse
|