1
|
Leong IUS, 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 SEA, 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 2024:JCO2302761. [PMID: 39481076 DOI: 10.1200/jco.23.02761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
| | - Eleanor M Williams
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | - Louise C Daugherty
- Genomics England Ltd, Level 21 One Canada Square, London, United Kingdom
| | | | | | | | | | - 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
|
2
|
Malekkou A, Tomazou M, Mavrikiou G, Dionysiou M, Georgiou T, Papaevripidou I, Alexandrou A, Sismani C, Drousiotou A, Grafakou O, Petrou PP. A novel large intragenic DPYD deletion causing dihydropyrimidine dehydrogenase deficiency: a case report. BMC Med Genomics 2024; 17:78. [PMID: 38528593 PMCID: PMC10962175 DOI: 10.1186/s12920-024-01846-2] [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: 10/31/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Dihydropyrimidine dehydrogenase (DPD), is the initial and rate-limiting enzyme in the catabolic pathway of pyrimidines. Deleterious variants in the DPYD gene cause DPD deficiency, a rare autosomal recessive disorder. The clinical spectrum of affected individuals is wide ranging from asymptomatic to severely affected patients presenting with intellectual disability, motor retardation, developmental delay and seizures. DPD is also important as the main enzyme in the catabolism of 5-fluorouracil (5-FU) which is extensively used as a chemotherapeutic agent. Even in the absence of clinical symptoms, individuals with either complete or partial DPD deficiency face a high risk of severe and even fatal fluoropyrimidine-associated toxicity. The identification of causative genetic variants in DPYD is therefore gaining increasing attention due to their potential use as predictive markers of fluoropyrimidine toxicity. METHODS A male infant patient displaying biochemical features of DPD deficiency was investigated by clinical exome sequencing. Bioinformatics tools were used for data analysis and results were confirmed by MLPA and Sanger sequencing. RESULTS A novel intragenic deletion of 71.2 kb in the DPYD gene was identified in homozygosity. The deletion, DPYD(NM_000110.4):c.850 + 23455_1128 + 8811del, eliminates exons 9 and 10 and may have resulted from a non-homologous end-joining event, as suggested by in silico analysis. CONCLUSIONS The study expands the spectrum of DPYD variants associated with DPD deficiency. Furthermore, it raises the concern that patients at risk for fluoropyrimidine toxicity due to DPYD deletions could be missed during pre-treatment genetic testing for the currently recommended single nucleotide polymorphisms.
Collapse
Affiliation(s)
- Anna Malekkou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Marios Tomazou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Gavriella Mavrikiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Maria Dionysiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Theodoros Georgiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Ioannis Papaevripidou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Angelos Alexandrou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Carolina Sismani
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Anthi Drousiotou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus
| | - Olga Grafakou
- Department of Pediatrics, Inborn Errors of Metabolism Clinic, Archbishop Makarios III Hospital, Korytsas 6, 2012, Nicosia, Cyprus
| | - Petros P Petrou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, P. O. Box 23462, 1683, Nicosia, Cyprus.
| |
Collapse
|
3
|
Pinheiro M, Peixoto A, Rocha P, Santos C, Escudeiro C, Veiga I, Porto M, Guerra J, Barbosa A, Pinto C, Arinto P, Resende A, Teixeira MR. Implementation of upfront DPYD genotyping with a low-cost and high-throughput assay to guide fluoropyrimidine treatment in cancer patients. Pharmacogenet Genomics 2023; 33:165-171. [PMID: 37611150 DOI: 10.1097/fpc.0000000000000505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
OBJECTIVES Genetic variants in the dihydropyrimidine dehydrogenase (DPYD ) gene are associated with reduced dihydropyrimidine dehydrogenase enzyme activity and can cause severe fluoropyrimidine-related toxicity. We assessed the frequency of the four most common and well-established DPYD variants associated with fluoropyrimidine toxicity and implemented a relatively low-cost and high-throughput genotyping assay for their detection. METHODS This study includes 457 patients that were genotyped for the DPYD c.1129-5923C>G, c.1679T>G, c.1905 + 1G>A and c.2846A>T variants, either by Sanger sequencing or kompetitive allele specific PCR (KASP) technology. Of these, 172 patients presented toxicity during treatment with fluoropyrimidines (post-treatment group), and 285 were tested before treatment (pretreatment group). RESULTS Heterozygous DPYD variants were identified in 7.4% of the entire series of 457 patients, being the c.2846A>T the most frequent variant. In the post-treatment group, 15.7% of the patients presented DPYD variants, whereas only 2.5% of the patients in the pretreatment group presented a variant. The KASP assays designed in this study presented 100% genotype concordance with the results obtained by Sanger sequencing. CONCLUSIONS The combined assessment of the four DPYD variants in our population increases the identification of patients at high risk for developing fluoropyrimidine toxicity, supporting the upfront routine implementation of DPYD variant genotyping. Furthermore, the KASP genotyping assay described in this study presents a rapid turnaround time and relatively low cost, making upfront DPYD screening feasible in clinical practice.
Collapse
Affiliation(s)
- Manuela Pinheiro
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
| | - Ana Peixoto
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Patrícia Rocha
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Catarina Santos
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Carla Escudeiro
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Isabel Veiga
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Miguel Porto
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
| | - Joana Guerra
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
| | - Ana Barbosa
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Carla Pinto
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Patrícia Arinto
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
| | - Adriana Resende
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
| | - Manuel R Teixeira
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
4
|
White C, Scott RJ, Paul C, Ziolkowski A, Mossman D, Ackland S. Ethnic Diversity of DPD Activity and the DPYD Gene: Review of the Literature. Pharmgenomics Pers Med 2021; 14:1603-1617. [PMID: 34916829 PMCID: PMC8668257 DOI: 10.2147/pgpm.s337147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/10/2021] [Indexed: 12/31/2022] Open
Abstract
Pharmacogenomic screening can identify patients with gene variants that predispose them to the development of severe toxicity from fluoropyrimidine (FP) chemotherapy. Deficiency of the critical metabolic enzyme dihydropyrimidine dehydrogenase (DPD) leads to excessive toxicity on exposure to fluoropyrimidine chemotherapy. This can result in hospitalisation, intensive care admissions and even death. Upfront screening of the gene that encodes for DPD (DPYD) has recently been implemented in regions throughout Europe and the United Kingdom. Current screening evaluates DPYD variants that are well described within Caucasian patient populations and provides genotyped-guided dose adjustment recommendations based upon the presence of these variants. This article reviews the differences in DPYD gene variants within non-Caucasian populations compared to Caucasian populations, with regard to the implications for clinical tolerance of fluoropyrimidine chemotherapies and genotype guided dose adjustment guidelines.
Collapse
Affiliation(s)
- Cassandra White
- University of Newcastle, Newcastle, NSW, Australia.,Hunter Cancer Research Alliance, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Rodney J Scott
- University of Newcastle, Newcastle, NSW, Australia.,Hunter Cancer Research Alliance, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Division of Molecular Medicine, Pathology North John Hunter Hospital, Newcastle, NSW, Australia
| | - Christine Paul
- University of Newcastle, Newcastle, NSW, Australia.,Hunter Cancer Research Alliance, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Andrew Ziolkowski
- Division of Molecular Medicine, Pathology North John Hunter Hospital, Newcastle, NSW, Australia
| | - David Mossman
- Division of Molecular Medicine, Pathology North John Hunter Hospital, Newcastle, NSW, Australia
| | - Stephen Ackland
- University of Newcastle, Newcastle, NSW, Australia.,Hunter Cancer Research Alliance, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Hunter Cancer Centre, Lake Macquarie Private Hospital, Gateshead, NSW, Australia
| |
Collapse
|
5
|
Cura Y, Pérez Ramírez C, Sánchez Martín A, Martínez Martínez F, Calleja Hernández MÁ, Ramírez Tortosa MDC, Jiménez Morales A. Genetic polymorphisms on the effectiveness or safety of breast cancer treatment: Clinical relevance and future perspectives. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108391. [PMID: 34893156 DOI: 10.1016/j.mrrev.2021.108391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 06/14/2023]
Abstract
Breast cancer (BC) is the most frequent neoplasm and one of the main causes of death in women. The pharmacological treatment of BC consists of hormonal therapy, chemotherapeutic agents and targeted therapy. The response to BC therapy is highly variable in clinical practice. This variability can be explained by the presence of genetic polymorphisms in genes involved in the pharmacokinetics, pharmacodynamics or immune response of patients. The abundant evidence of associations between low-activity alleles CYP2D6*3, *4, *5, *6, *10 and *41 and poor results with tamoxifen therapy, and between DPYD gene polymorphisms rs3918290, rs55886062, rs67376798 and rs75017182 and increased risk of toxicity to fluoropyrimidine therapy, justify the existence of clinical pharmacogenetic guidelines. The NQO1 rs1800566 polymorphism is related to poorer results in BC therapy with chemotherapy agents. The polymorphism rs1695 of the GSTP1 gene has been associated with the effectiveness and toxicity of fluorouracil, cyclophosphamide and epirubicin therapy. Finally, the HLA-DQA1*02:01 allele is significantly associated with the occurrence of liver toxicity events in patients receiving lapatinib. There is moderate evidence to support the aforementioned associations and, therefore, a high probability of these being considered as future predictive genetic biomarkers of response. However, further studies are required to reinforce or clarify their clinical relevance.
Collapse
Affiliation(s)
- Yasmin Cura
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, Granada, Spain.
| | - Cristina Pérez Ramírez
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen Macarena, Seville, Spain.
| | - Almudena Sánchez Martín
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, Granada, Spain.
| | - Fernando Martínez Martínez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada, Spain.
| | | | | | - Alberto Jiménez Morales
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, Granada, Spain.
| |
Collapse
|
6
|
Saarenheimo J, Wahid N, Eigeliene N, Ravi R, Salomons GS, Ojeda MF, Vijzelaar R, Jekunen A, van Kuilenburg ABP. Preemptive screening of DPYD as part of clinical practice: high prevalence of a novel exon 4 deletion in the Finnish population. Cancer Chemother Pharmacol 2021; 87:657-663. [PMID: 33544210 DOI: 10.1007/s00280-021-04236-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/18/2021] [Indexed: 01/08/2023]
Abstract
Capecitabine is a fluoropyrimidine that is widely used as a cancer drug for the treatment of patients with a variety of cancers. Unfortunately, early onset, severe or life-threatening toxicity is observed in 19-32% of patients treated with capecitabine and 5FU. Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme in the degradation of 5FU and a DPD deficiency has been shown to be a major determinant of severe fluoropyrimidine-associated toxicity. DPD is encoded by the DPYD gene and some of the identified variants have been described to cause DPD deficiency. Preemptive screening for DPYD gene alterations enables the identification of DPD-deficient patients before administering fluoropyrimidines. In this article, we describe the application of upfront DPD screening in Finnish patients, as a part of daily clinical practice, which was based on a comprehensive DPYD gene analysis, measurements of enzyme activity and plasma uracil concentrations. Almost 8% of the patients (13 of 167 patients) presented with pathogenic DPYD variants causing DPD deficiency. The DPD deficiency in these patients was further confirmed via analysis of the DPD activity and plasma uracil levels. Interestingly, we identified a novel intragenic deletion in DPYD which includes exon 4 in four patients (31% of patients carrying a pathogenic variant). The high prevalence of the exon 4 deletion among Finnish patients highlights the importance of full-scale DPYD gene analysis. Based on the literature and our own experience, genotype preemptive screening should always be used to detect DPD-deficient patients before fluoropyrimidine therapy.
Collapse
Affiliation(s)
- Jatta Saarenheimo
- Department of Pathology, Vasa Central Hospital, Hietalahdenkatu 2-4, 65130, Vaasa, Finland.
| | - Nesna Wahid
- Department of Oncology, Vasa Central Hospital, Vaasa, Finland
| | - Natalja Eigeliene
- Department of Oncology, Vasa Central Hospital, Vaasa, Finland.,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland
| | | | - Gajja S Salomons
- Metabolic Unit, Department of Clinical Chemistry& Laboratory Genetic Metabolic Diseases & Department of Paediatric Metabolic Diseases, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Matilde Fernandez Ojeda
- Metabolic Unit, Department of Clinical Chemistry& Laboratory Genetic Metabolic Diseases & Department of Paediatric Metabolic Diseases, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Antti Jekunen
- Department of Oncology, Vasa Central Hospital, Vaasa, Finland.,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland
| | - André B P van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Stavraka C, Pouptsis A, Okonta L, DeSouza K, Charlton P, Kapiris M, Marinaki A, Karapanagiotou E, Papadatos-Pastos D, Mansi J. Clinical implementation of pre-treatment DPYD genotyping in capecitabine-treated metastatic breast cancer patients. Breast Cancer Res Treat 2019; 175:511-517. [PMID: 30746637 PMCID: PMC6533219 DOI: 10.1007/s10549-019-05144-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 01/21/2019] [Indexed: 12/26/2022]
Abstract
Purpose Metastatic breast cancer (mBC) patients with DPYD genetic variants linked to loss of dihydropyrimidine dehydrogenase (DPD) activity are at risk of severe capecitabine-associated toxicities. However, prospective DPYD genotyping has not yet been implemented in routine clinical practice. Following a previous internal review in which two patients underwent lengthy hospitalisations whilst receiving capecitabine, and were subsequently found to be DPD deficient, we initiated routine DPYD genotyping prior to starting capecitabine. This study evaluates the clinical application of routine DPYD screening at a large cancer centre in London. Methods We reviewed medical records for consecutive patients with mBC who underwent DPYD genotyping before commencing capecitabine between December 2014 and December 2017. Patients were tested for four DPYD variants associated with reduced DPD activity. Results Sixty-six patients underwent DPYD testing. Five (8.4%) patients were found to carry DPYD genetic polymorphisms associated with reduced DPD activity; of these, two received dose-reduced capecitabine. Of the 61 patients with DPYD wild-type, 14 (23%) experienced grade 3 toxicities which involved palmar–plantar erythrodysesthesia (65%), and gastrointestinal toxicities (35%); no patient was hospitalised due to toxicity. Conclusions Prospective DPYD genotyping can be successfully implemented in routine clinical practice and can reduce the risk of severe fluoropyrimidine toxicities.
Collapse
Affiliation(s)
- Chara Stavraka
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Athanasios Pouptsis
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Leroy Okonta
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Karen DeSouza
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Philip Charlton
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Matthaios Kapiris
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Anthony Marinaki
- Purine Research Laboratory, Viapath, Guy’s and St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London, SE1 7EH UK
| | - Eleni Karapanagiotou
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Dionysis Papadatos-Pastos
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| | - Janine Mansi
- Breast Unit, Guy’s and St Thomas’ NHS Foundation Trust and King’s Biomedical Centre, 4th Floor, Bermondsey Wing, Great Maze Pond, London, SE1 9RT UK
| |
Collapse
|
8
|
Murphy C, Byrne S, Ahmed G, Kenny A, Gallagher J, Harvey H, O'Farrell E, Bird B. Cost Implications of Reactive Versus Prospective Testing for Dihydropyrimidine Dehydrogenase Deficiency in Patients With Colorectal Cancer: A Single-Institution Experience. Dose Response 2018; 16:1559325818803042. [PMID: 30288154 PMCID: PMC6168732 DOI: 10.1177/1559325818803042] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/14/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Severe toxicity is experienced by a substantial minority of patients receiving
fluoropyrimidine-based chemotherapy, with approximately 20% of these severe toxicities
attributable to polymorphisms in the DPYD gene. The
DPYD codes for the enzyme dihydropyrimidine dehydrogenase (DPD)
important in the metabolism of fluoropyrimidine-based chemotherapy. We questioned
whether prospective DPYD mutation analysis in all patients commencing
such therapy would prove more cost-effective than reactive testing of patients
experiencing severe toxicity. Methods: All patients experiencing severe toxicity from fluoropyrimidine-based chemotherapy for
colorectal cancer in an Irish private hospital over a 3-year period were tested for 4
DPYD polymorphisms previously associated with toxicity. The costs
associated with an index admission for toxicity in DPD-deficient patients were examined.
A cost analysis was undertaken comparing the anticipated cost of implementing screening
for DPYD mutations versus current usual care. One-way sensitivity
analysis was conducted on known input variables. An alternative scenario analysis from
the perspective of the Irish health-care payer (responsible for public hospitals) was
also performed. Results: Of 134 patients commencing first-line fluoropyrimidine chemotherapy over 3 years, 30
(23%) patients developed grade 3/4 toxicity. Of these, 17% revealed heterozygote
DPYD mutations. The cost of hospitalization for the
DPYD-mutated patients was €232 061, while prospectively testing all
134 patients would have cost €23 718. Prospective testing would result in cost savings
across all scenarios. Conclusions: The cost of hospital admission for severe chemotherapy-related toxicity is
significantly higher than the cost of prospective DPYD testing of each
patient commencing fluoropyrimidine chemotherapy.
Collapse
Affiliation(s)
- Con Murphy
- Medical Oncology, Bon Secours Cork, University College Cork School of Medicine, Cork, Ireland
| | - Stephen Byrne
- School of Pharmacy, University College Cork National University of Ireland, Cork, Ireland
| | - Gul Ahmed
- Medical Oncology, Bons Secours Cork, Cork, Ireland
| | | | - James Gallagher
- School of Pharmacy, University College Cork National University of Ireland, Cork, Ireland
| | - Harry Harvey
- University College Cork School of Medicine, Cork, Ireland
| | - Eoin O'Farrell
- University College Cork School of Medicine, Cork, Ireland
| | - Brian Bird
- Medical Oncology, Bon Secours Cork, University College Cork School of Medicine, Cork, Ireland
| |
Collapse
|
9
|
Emami AH, Sadighi S, Shirkoohi R, Mohagheghi MA. Prediction of Response to Irinotecan and Drug Toxicity Based on Pharmacogenomics Test: A Prospective Case Study in Advanced Colorectal Cancer. Asian Pac J Cancer Prev 2017; 18:2803-2807. [PMID: 29072417 PMCID: PMC5747407 DOI: 10.22034/apjcp.2017.18.10.2803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: FOLFIRI regimen, which is composed of 5-FU, Leucovorin, and Irinotecan, is used in the first-line chemotherapy of metastatic colorectal cancer. Irinotecan life threatening toxicity is partly related to cytotoxic drug metabolite which is primarily inactivated by the UGT1A1 enzyme. The primary aim of the present research was to find the correlation between UGT1A1-genotype and clinical toxicity of irinotecan. Methods: In a prospective study from March 2011 to December 2013, all patients with metastatic colorectal cancer who had been referred to Medical Oncology Department of Iran Cancer Institute were genotyped for UGT1A1*28 before the first cycle of chemotherapy. All of the patients signed informed consent and trial approved by Ethics Committee of the Tehran University of Medical Sciences. Reduction of the standard dose of Irinotecan (180 mg/m2 body surface area) was measured based on NCI toxicity criteria after the first cycle of chemotherapy. Patients with previous treatment with Oxaliplatin and fluorouracil (5-FU) in the adjuvant setting and adequate liver, kidney, and heart function were included in the trial. Both synchronous and metachronous metastatic disease were noticeable. Results: A total of 50 patients with median age of 52 years were included. Most (70%) of the patients had more than one site of metastases in peritoneum, liver, and/or lung. Thirty-one patients had UGT1A1*1 normal genotype, 13 were in heterozygote and 6 were in homozygote state ofUGT1A1*28/*28. A clinically relevant increase in early toxicity was found in patients carrying the UGT1A1*28/*28 genotype with odds Ratio (OR) of 2.6 (95%CI 2.5-27.28). Similarly, there was a trend of lower overall survival in homozygote group with an HR (Hazardous Ratio) of 2.76 (95%CI .88-.61). No statistically significant relationship was found between UGT1A1genotypes and response to therapy. Conclusions: UGT1A1 28*/28* is strongly associated with drug’s life-threatening toxicity even in a moderate dose of Irinotecan. On the other hand, UGT1A1 genotype data was not helpful to differentiate response to treatment.
Collapse
Affiliation(s)
- A H Emami
- Hematology-Medical Oncology Department Cancer Research Center, Cancer Institute of Iran, Tehran University Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
10
|
Galarza AFA, Linden R, Antunes MV, Hahn RZ, Raymundo S, da Silva ACC, Staggemeier R, Spilki FR, Schwartsmann G. Endogenous plasma and salivary uracil to dihydrouracil ratios and DPYD genotyping as predictors of severe fluoropyrimidine toxicity in patients with gastrointestinal malignancies. Clin Biochem 2016; 49:1221-1226. [PMID: 27399164 DOI: 10.1016/j.clinbiochem.2016.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/20/2016] [Accepted: 07/06/2016] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the use of plasma and saliva uracil (U) to dihydrouracil (UH2) metabolic ratio and DPYD genotyping, as a means to identify patients with dihydropyrimidine dehydrogenase (DPD) deficiency and fluoropyrimidine toxicity. METHODS Paired plasma and saliva samples were obtained from 60 patients with gastrointestinal cancer, before fluoropyrimidine treatment. U and UH2 concentrations were measured by LC-MS/MS. DPYD was genotyped for alleles *7, *2A, *13 and Y186C. Data on toxicity included grade 1 to 4 neutropenia, mucositis, diarrhea, nausea/vomiting and cutaneous rash. RESULTS 35% of the patients had severe toxicity. There was no variant allele carrier for DPYD. The [UH2]/[U] metabolic ratios were 0.09-26.73 in plasma and 0.08-24.0 in saliva, with higher correlation with toxicity grade in saliva compared to plasma (rs=-0.515 vs rs=-0.282). Median metabolic ratios were lower in patients with severe toxicity as compared to those with absence of toxicity (0.59 vs 2.83 saliva; 1.62 vs 6.75 plasma, P<0.01). A cut-off of 1.16 for salivary ratio was set (AUC 0.842), with 86% sensitivity and 77% specificity for the identification of patients with severe toxicity. Similarly, a plasma cut-off of 4.0 (AUC 0.746), revealed a 71% sensitivity and 76% specificity. CONCLUSIONS DPYD genotyping for alleles 7, *2A, *13 and Y186C was not helpful in the identification of patients with severe DPD deficiency in this series of patients. The [UH2]/[U] metabolic ratios, however, proved to be a promising functional test to identify the majority of cases of severe DPD activity, with saliva performing better than plasma.
Collapse
Affiliation(s)
- Andrés Fernando Andrade Galarza
- Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Oncologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rafael Linden
- Instituto de Ciências da Saúde, Universidade Feevale, Novo Hamburgo, RS, Brazil
| | | | - Roberta Zilles Hahn
- Instituto de Ciências da Saúde, Universidade Feevale, Novo Hamburgo, RS, Brazil
| | - Suziane Raymundo
- Instituto de Ciências da Saúde, Universidade Feevale, Novo Hamburgo, RS, Brazil
| | | | - Rodrigo Staggemeier
- Instituto de Ciências da Saúde, Universidade Feevale, Novo Hamburgo, RS, Brazil
| | | | - Gilberto Schwartsmann
- Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Oncologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
11
|
Negrei C, Hudita A, Ginghina O, Galateanu B, Voicu SN, Stan M, Costache M, Fenga C, Drakoulis N, Tsatsakis AM. Colon Cancer Cells Gene Expression Signature As Response to 5- Fluorouracil, Oxaliplatin, and Folinic Acid Treatment. Front Pharmacol 2016; 7:172. [PMID: 27445811 PMCID: PMC4917556 DOI: 10.3389/fphar.2016.00172] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/03/2016] [Indexed: 12/05/2022] Open
Abstract
5-FU cytotoxicity mechanism has been assigned both to the miss-incorporation of fluoronucleotides into RNA and DNA and to the inhibition of thymidylate synthase. 5-FU is one of the most widely used chemotherapeutic drugs, although it has severe side effects that may vary between patients. Pharmacogenetic studies related to 5-FU have been traditionally focused on the rate-limiting catabolic enzyme, dihydropyrimidine dehydrogenase that breaks 80–85% of 5-FU into its inactive metabolite. Choosing the right dosing scheme and chemotherapy strategy for each individual patient remains challenging for personalized chemotherapy management. In the general effort toward reduction of colorectal cancer mortality, in vitro screening studies play a very important role. To accelerate translation research, increasing interest has been focused on using in vivo-like models such as three-dimensional spheroids. The development of higher throughput assays to quantify phenotypic changes in spheroids is an active research area. Consequently, in this study we used the microarray technology to reveal the HT-29 colorectal adenocarcinoma cells gene expression signature as response to 5-FU/OXP/FA treatment in a state of the art 3D culture system. We report here an increased reactive oxygen species production under treatment, correlated with a decrease in cell viability and proliferation potential. With respect to the HT-29 cells gene expression under the treatment with 5-FU/OXP/FA, we found 15.247 genes that were significantly differentially expressed (p < 0.05) with a fold change higher that two-fold. Among these, 7136 genes were upregulated and 8111 genes were downregulated under experimental conditions as compared to untreated cells. The most relevant and statistic significant (p < 0.01) pathways in the experiment are associated with the genes that displayed significant differential expression and are related to intracellular signaling, oxidative stress, apoptosis, and cancer.
Collapse
Affiliation(s)
- Carolina Negrei
- Department of Toxicology, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy Bucharest, Romania
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Octav Ginghina
- Department of Surgery, "Sf. Ioan" Clinical Emergency HospitalBucharest, Romania; Department II, Faculty of Dental Medicine, "Carol Davila" University of Medicine and PharmacyBucharest, Romania
| | - Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Sorina Nicoleta Voicu
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Miriana Stan
- Department of Toxicology, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Concettina Fenga
- Occupational Medicine Section, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina Messina, Italy
| | - Nikolaos Drakoulis
- Research Laboratory of Clinical Pharmacology and Pharmacogenomics, School of Health Sciences, Faculty of Pharmacy, National and Kapodistrian University of Athens Athens, Greece
| | - Aristidis M Tsatsakis
- Department of Toxicology and Forensic Sciences, Medical School, University of Crete Heraklion, Greece
| |
Collapse
|
12
|
DPYD gene polymorphisms are associated with risk and chemotherapy prognosis in pediatric patients with acute lymphoblastic leukemia. Tumour Biol 2016; 37:10393-402. [DOI: 10.1007/s13277-016-4908-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/22/2016] [Indexed: 01/13/2023] Open
|
13
|
Meulendijks D, Henricks LM, Sonke GS, Deenen MJ, Froehlich TK, Amstutz U, Largiadèr CR, Jennings BA, Marinaki AM, Sanderson JD, Kleibl Z, Kleiblova P, Schwab M, Zanger UM, Palles C, Tomlinson I, Gross E, van Kuilenburg ABP, Punt CJA, Koopman M, Beijnen JH, Cats A, Schellens JHM. Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data. Lancet Oncol 2015; 16:1639-50. [PMID: 26603945 DOI: 10.1016/s1470-2045(15)00286-7] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/26/2015] [Accepted: 08/28/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND The best-known cause of intolerance to fluoropyrimidines is dihydropyrimidine dehydrogenase (DPD) deficiency, which can result from deleterious polymorphisms in the gene encoding DPD (DPYD), including DPYD*2A and c.2846A>T. Three other variants-DPYD c.1679T>G, c.1236G>A/HapB3, and c.1601G>A-have been associated with DPD deficiency, but no definitive evidence for the clinical validity of these variants is available. The primary objective of this systematic review and meta-analysis was to assess the clinical validity of c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity. METHODS We did a systematic review of the literature published before Dec 17, 2014, to identify cohort studies investigating associations between DPYD c.1679T>G, c.1236G>A/HapB3, and c.1601G>A and severe (grade ≥3) fluoropyrimidine-associated toxicity in patients treated with fluoropyrimidines (fluorouracil, capecitabine, or tegafur-uracil as single agents, in combination with other anticancer drugs, or with radiotherapy). Individual patient data were retrieved and analysed in a multivariable analysis to obtain an adjusted relative risk (RR). Effect estimates were pooled by use of a random-effects meta-analysis. The threshold for significance was set at a p value of less than 0·0167 (Bonferroni correction). FINDINGS 7365 patients from eight studies were included in the meta-analysis. DPYD c.1679T>G was significantly associated with fluoropyrimidine-associated toxicity (adjusted RR 4·40, 95% CI 2·08-9·30, p<0·0001), as was c.1236G>A/HapB3 (1·59, 1·29-1·97, p<0·0001). The association between c.1601G>A and fluoropyrimidine-associated toxicity was not significant (adjusted RR 1·52, 95% CI 0·86-2·70, p=0·15). Analysis of individual types of toxicity showed consistent associations of c.1679T>G and c.1236G>A/HapB3 with gastrointestinal toxicity (adjusted RR 5·72, 95% CI 1·40-23·33, p=0·015; and 2·04, 1·49-2·78, p<0·0001, respectively) and haematological toxicity (adjusted RR 9·76, 95% CI 3·03-31·48, p=0·00014; and 2·07, 1·17-3·68, p=0·013, respectively), but not with hand-foot syndrome. DPYD*2A and c.2846A>T were also significantly associated with severe fluoropyrimidine-associated toxicity (adjusted RR 2·85, 95% CI 1·75-4·62, p<0·0001; and 3·02, 2·22-4·10, p<0·0001, respectively). INTERPRETATION DPYD variants c.1679T>G and c.1236G>A/HapB3 are clinically relevant predictors of fluoropyrimidine-associated toxicity. Upfront screening for these variants, in addition to the established variants DPYD*2A and c.2846A>T, is recommended to improve the safety of patients with cancer treated with fluoropyrimidines. FUNDING None.
Collapse
Affiliation(s)
- Didier Meulendijks
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Linda M Henricks
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gabe S Sonke
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tanja K Froehlich
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Ursula Amstutz
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Carlo R Largiadèr
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | | | | | | | - Zdenek Kleibl
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Petra Kleiblova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; Department of Clinical Pharmacology, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Claire Palles
- Molecular and Population Genetics Laboratory and Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory and Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Eva Gross
- Department of Gynecology and Obstetrics, Technische Universität München, Munich, Germany
| | - André B P van Kuilenburg
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Annemieke Cats
- Department of Gastroenterology and Hepatology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
14
|
Kleist B, Kempa M, Meurer T, Poetsch M. Correlation betweenDPYDgene variation andKRASwild type status in colorectal cancer. J Clin Pathol 2015; 69:204-8. [DOI: 10.1136/jclinpath-2015-202903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/26/2015] [Indexed: 11/03/2022]
|
15
|
Matsusaka S, Lenz HJ. Pharmacogenomics of fluorouracil -based chemotherapy toxicity. Expert Opin Drug Metab Toxicol 2015; 11:811-21. [PMID: 25800061 DOI: 10.1517/17425255.2015.1027684] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION 5- fluorouracil (5-FU), alone or in combination, is the most prevalent and effective chemotherapeutic agent for the treatment of cancers of the head and neck, breast, pancreas and gastrointestinal tract. AREAS COVERED Three rare DPYD mutations, a splice mutation in intron 14 (c.1905+1G>A) and two nonsynonymous coding variants (c.1679T>G, c.2846A>T), have consistently been associated with severe 5-FU toxicity. A relatively common haplotype, hapB3, containing three intronic polymorphisms (c.483+18G>A; c.680+139G>A; c.959-51T>C) and a synonymous mutation c.1236G>A linked to c.1129-5923C>G, is a major contributor to early onset severe toxicity. TYMS VNTR 2R and TYMS-3'-UTR 6-bp ins-del variants were associated with global toxicity in capecitabine-treated patients. A candidate gene study of capecitabine-related toxicity reported that the s12132152 were strongly associated with hand-foot syndrome (HFS), whereas rs7548189 was associated with diarrhea. The rs2612091 and rs2741171, which are downstream of TYMS and intronic for ENOSF1, were associated with increased global toxicity and HFS. EXPERT OPINION Sex-dependent differences, ethnicity, cancer types and 5-FU-based chemotherapy regimens might affect the heterogeneity of genetic variants for predictive 5-FU-related toxicity. Future approaches using genome-wide association analyses may help in identifying additional candidate genes causally involved in the path mechanisms of 5-FU-related toxicity.
Collapse
Affiliation(s)
- Satoshi Matsusaka
- University of Southern California, Keck School of Medicine, Norris Comprehensive Cancer Center, Division of Medical Oncology , Los Angeles, CA , USA
| | | |
Collapse
|
16
|
Identification of genetic variants associated with capecitabine-induced hand-foot syndrome through integration of patient and cell line genomic analyses. Pharmacogenet Genomics 2014; 24:231-7. [PMID: 24595012 DOI: 10.1097/fpc.0000000000000037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE A primary challenge in identifying replicable pharmacogenomic markers from clinical genomewide association study (GWAS) trials in oncology is the difficulty in performing a second large clinical trial with the same drugs and dosage regimen. We sought to overcome this challenge by incorporating GWAS results from cell-based studies using the same chemotherapy as a clinical cohort. METHODS In this study, we test whether the overlap between genetic variants identified in a preclinical study and a clinical study on capecitabine is more than expected by chance. A GWAS of capecitabine-induced cytotoxicity was performed in 164 lymphoblastoid cell lines derived from the CEU HapMap population and compared with a GWAS of hand-foot syndrome (HFS), the most frequent capecitabine-induced adverse drug reaction, in Spanish breast and colorectal cancer patients (n=160) treated with capecitabine. RESULTS We observed an overlap of 16 single nucleotide polymorphisms associated with capecitabine-induced cytotoxicity (P<0.001) in lymphoblastoid cell lines and HFS (P<0.05) in patients, which is a greater overlap than expected by chance (genotype-phenotype permutation empirical P=0.015). Ten tag single nucleotide polymorphisms, which cover the overlap loci, were genotyped in a second patient cohort (n=85) and one of them, rs9936750, was associated with capecitabine-induced HFS (P=0.0076). CONCLUSION The enrichment results imply that cellular models of capecitabine-induced cytotoxicity may capture components of the underlying polygenic architecture of related toxicities in patients.
Collapse
|
17
|
Lee AM, Shi Q, Pavey E, Alberts SR, Sargent DJ, Sinicrope FA, Berenberg JL, Goldberg RM, Diasio RB. DPYD variants as predictors of 5-fluorouracil toxicity in adjuvant colon cancer treatment (NCCTG N0147). J Natl Cancer Inst 2014; 106:dju298. [PMID: 25381393 DOI: 10.1093/jnci/dju298] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Previous studies have suggested the potential importance of three DPYD variants (DPYD*2A, D949V, and I560S) with increased 5-FU toxicity. Their individual associations, however, in 5-FU-based combination therapies, remain controversial and require further systematic study in a large patient population receiving comparable treatment regimens with uniform clinical data. METHODS We genotyped 2886 stage III colon cancer patients treated adjuvantly in a randomized phase III trial with FOLFOX or FOLFIRI, alone or combined with cetuximab, and tested the individual associations between functionally deleterious DPYD variants and toxicity. Logistic regressions were used to assess univariate and multivariable associations. All statistical tests were two-sided. RESULTS In 2594 patients with complete adverse event (AE) data, the incidence of grade 3 or greater 5FU-AEs in DPYD*2A, I560S, and D949V carriers were 22/25 (88.0%), 2/4 (50.0%), and 22/27 (81.5%), respectively. Statistically significant associations were identified between grade 3 or greater 5FU-AEs and both DPYD*2A (odds ratio [OR] = 15.21, 95% confidence interval [CI] = 4.54 to 50.96, P < .001) and D949V (OR = 9.10, 95% CI = 3.43 to 24.10, P < .001) variants. Statistical significance remained after adjusting for multiple variables. The DPYD*2A variant statistically significantly associated with the specific AEs nausea/vomiting (P = .007) and neutropenia (P < .001), whereas D949V statistically significantly associated with dehydration (P = .02), diarrhea (P = .003), leukopenia (P = .002), neutropenia (P < .001), and thrombocytopenia (P < .001). Although two patients with I560S had grade≥3 5FU-AEs; a statistically significant association could not be demonstrated because of its low frequency (P = .48). CONCLUSION In the largest study to date, statistically significant associations were found between DPYD variants (DPYD*2A and D949V) and increased incidence of grade 3 or greater 5FU-AEs in patients treated with adjuvant 5-FU-based combination chemotherapy.
Collapse
Affiliation(s)
- Adam M Lee
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Qian Shi
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Emily Pavey
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Steven R Alberts
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Daniel J Sargent
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Frank A Sinicrope
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Jeffrey L Berenberg
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Richard M Goldberg
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG)
| | - Robert B Diasio
- Department of Molecular Pharmacology and Experimental Therapeutics (AML, RBD), Alliance Statistics and Data Center (QS, EP, DJS), Division of Medical Oncology (SRA, FAS, RBD), Division of Gastroenterology (FAS), Mayo Clinic Cancer Center, Rochester, MN; Cancer Prevention and Control Program, University of Hawaii Cancer Center, Honolulu, HI (JLB); Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH (RMG).
| |
Collapse
|
18
|
Frequent intragenic rearrangements of DPYD in colorectal tumours. THE PHARMACOGENOMICS JOURNAL 2014; 15:211-8. [DOI: 10.1038/tpj.2014.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/31/2014] [Accepted: 09/19/2014] [Indexed: 01/14/2023]
|
19
|
Panczyk M. Pharmacogenetics research on chemotherapy resistance in colorectal cancer over the last 20 years. World J Gastroenterol 2014; 20:9775-827. [PMID: 25110414 PMCID: PMC4123365 DOI: 10.3748/wjg.v20.i29.9775] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 01/17/2014] [Accepted: 04/21/2014] [Indexed: 02/07/2023] Open
Abstract
During the past two decades the first sequencing of the human genome was performed showing its high degree of inter-individual differentiation, as a result of large international research projects (Human Genome Project, the 1000 Genomes Project International HapMap Project, and Programs for Genomic Applications NHLBI-PGA). This period was also a time of intensive development of molecular biology techniques and enormous knowledge growth in the biology of cancer. For clinical use in the treatment of patients with colorectal cancer (CRC), in addition to fluoropyrimidines, another two new cytostatic drugs were allowed: irinotecan and oxaliplatin. Intensive research into new treatment regimens and a new generation of drugs used in targeted therapy has also been conducted. The last 20 years was a time of numerous in vitro and in vivo studies on the molecular basis of drug resistance. One of the most important factors limiting the effectiveness of chemotherapy is the primary and secondary resistance of cancer cells. Understanding the genetic factors and mechanisms that contribute to the lack of or low sensitivity of tumour tissue to cytostatics is a key element in the currently developing trend of personalized medicine. Scientists hope to increase the percentage of positive treatment response in CRC patients due to practical applications of pharmacogenetics/pharmacogenomics. Over the past 20 years the clinical usability of different predictive markers has been tested among which only a few have been confirmed to have high application potential. This review is a synthetic presentation of drug resistance in the context of CRC patient chemotherapy. The multifactorial nature and volume of the issues involved do not allow the author to present a comprehensive study on this subject in one review.
Collapse
|
20
|
Bank PC, Swen JJ, Guchelaar HJ. Pharmacogenetic biomarkers for predicting drug response. Expert Rev Mol Diagn 2014; 14:723-35. [PMID: 24857685 DOI: 10.1586/14737159.2014.923759] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Drug response shows significant interpatient variability and evidence that genetics influences outcome of drug therapy has been known for more than five decades. However, the translation of this knowledge to clinical practice remains slow. Using examples from clinical practice six considerations about the implementation of pharmacogenetics (PGx) into routine care are discussed: the need for PGx biomarkers; the sources of genetic variability in drug response; the amount of variability explained by PGx; whether PGx test results are actionable; the level of evidence needed for implementation of PGx and the sources of information regarding interpretation of PGx data.
Collapse
Affiliation(s)
- Paul Christiaan Bank
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|
21
|
Alcazar-González GA, Calderón-Garcidueñas AL, Garza-Rodríguez ML, Rubio-Hernández G, Escorza-Treviño S, Olano-Martin E, Cerda-Flores RM, Castruita-Avila AL, González-Guerrero JF, le Brun S, Simon-Buela L, Barrera-Saldaña HA. Comparative study of polymorphism frequencies of the CYP2D6, CYP3A5, CYP2C8 and IL-10 genes in Mexican and Spanish women with breast cancer. Pharmacogenomics 2014; 14:1583-92. [PMID: 24088129 DOI: 10.2217/pgs.13.83] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIM Pharmacogenetic studies in breast cancer (BC) may predict the efficacy of tamoxifen and the toxicity of paclitaxel and capecitabine. We determined the frequency of polymorphisms in the CYP2D6 gene associated with activation of tamoxifen, and those of the genes CYP2C8, CYP3A5 and DPYD associated with toxicity of paclitaxel and capecitabine. We also included a IL-10 gene polymorphism associated with advanced tumor stage at diagnosis. PATIENTS & METHODS Genomic DNAs from 241 BC patients from northeast Mexico were genotyped using DNA microarray technology. RESULTS For tamoxifen processing, CYP2D6 genotyping predicted that 90.8% of patients were normal metabolizers, 4.2% ultrarapid, 2.1% intermediate and 2.9% poor metabolizers. For paclitaxel and the CYP2C8 gene, 75.3% were normal, 23.4% intermediate and 1.3% poor metabolizers. Regarding the DPYD gene, only one patient was a poor metabolizer. For the IL-10 gene, 47.1% were poor metabolizers. CONCLUSION These results contribute valuable information towards personalizing BC chemotherapy in Mexican women.
Collapse
|
22
|
Terrazzino S, Cargnin S, Del Re M, Danesi R, Canonico PL, Genazzani AA. DPYD IVS14+1G>A and 2846A>T genotyping for the prediction of severe fluoropyrimidine-related toxicity: a meta-analysis. Pharmacogenomics 2014; 14:1255-72. [PMID: 23930673 DOI: 10.2217/pgs.13.116] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
AIM In the present study we conducted a systematic review and meta-analysis of published data to quantify the impact of the DPYD IVS14+1G>A and 2846A>T variants on the risk of fluoropyrimidine-related toxicities and to determine sensitivity and specificity testing for DPYD variants. METHODS Relevant studies were identified through PubMed and Web of Knowledge databases, studies included were those published up until to May 2012. Study quality was assessed according to the HuGENET guidelines and Strengthening the Reporting of Genetic Association (STREGA) recommendations. RESULTS Random-effects meta-analysis provided evidence that carriers of DPYD IVS14+1G>A are at higher risk of ≥3 degrees of overall grade toxicity, hematological toxicity, mucositis and diarrhea. In addition, a strong association was also found between carriers of the DPYD 2846T allele and overall grade ≥3 toxicity or grade ≥3 diarrhea. An inverse linear relationship was found in prospective studies between the odds ratio of DPYD IVS14+1G>A and the incidence of overall grade ≥3 toxicity, indicating an higher impact in cohorts in which the incidence of severe toxicity was lower. CONCLUSION The results of this meta-analysis confirm clinical validity of DPYD IVS14+1G>A and 2846A>T as risk factors for the development of severe toxicities following fluoropyrimidine treatment. Furthermore, the sensitivity and specificity estimates obtained could be useful in establishing the cost-effectiveness of testing for DPYD variants.
Collapse
Affiliation(s)
- Salvatore Terrazzino
- Dipartimento di Scienze del Farmaco & Centro di Ricerca Interdipartimentale di Farmacogenetica e Farmacogenomica-CRIFF, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Jennings BA, Loke YK, Skinner J, Keane M, Chu GS, Turner R, Epurescu D, Barrett A, Willis G. Evaluating predictive pharmacogenetic signatures of adverse events in colorectal cancer patients treated with fluoropyrimidines. PLoS One 2013; 8:e78053. [PMID: 24167597 PMCID: PMC3805522 DOI: 10.1371/journal.pone.0078053] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 09/09/2013] [Indexed: 12/18/2022] Open
Abstract
The potential clinical utility of genetic markers associated with response to fluoropyrimidine treatment in colorectal cancer patients remains controversial despite extensive study. Our aim was to test the clinical validity of both novel and previously identified markers of adverse events in a broad clinical setting. We have conducted an observational pharmacogenetic study of early adverse events in a cohort study of 254 colorectal cancer patients treated with 5-fluorouracil or capecitabine. Sixteen variants of nine key folate (pharmacodynamic) and drug metabolising (pharmacokinetic) enzymes have been analysed as individual markers and/or signatures of markers. We found a significant association between TYMP S471L (rs11479) and early dose modifications and/or severe adverse events (adjusted OR = 2.02 [1.03; 4.00], p = 0.042, adjusted OR = 2.70 [1.23; 5.92], p = 0.01 respectively). There was also a significant association between these phenotypes and a signature of DPYD mutations (Adjusted OR = 3.96 [1.17; 13.33], p = 0.03, adjusted OR = 6.76 [1.99; 22.96], p = 0.002 respectively). We did not identify any significant associations between the individual candidate pharmacodynamic markers and toxicity. If a predictive test for early adverse events analysed the TYMP and DPYD variants as a signature, the sensitivity would be 45.5 %, with a positive predictive value of just 33.9 % and thus poor clinical validity. Most studies to date have been under-powered to consider multiple pharmacokinetic and pharmacodynamic variants simultaneously but this and similar individualised data sets could be pooled in meta-analyses to resolve uncertainties about the potential clinical utility of these markers.
Collapse
Affiliation(s)
- Barbara A. Jennings
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- * E-mail:
| | - Yoon K. Loke
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Jane Skinner
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Melanie Keane
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Gavin S. Chu
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Richard Turner
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Daniel Epurescu
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Ann Barrett
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Gavin Willis
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| |
Collapse
|
24
|
Kline CLB, El-Deiry WS. Personalizing colon cancer therapeutics: targeting old and new mechanisms of action. Pharmaceuticals (Basel) 2013; 6:988-1038. [PMID: 24276379 PMCID: PMC3817731 DOI: 10.3390/ph6080988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/30/2013] [Accepted: 08/16/2013] [Indexed: 12/17/2022] Open
Abstract
The use of pharmaceuticals for colon cancer treatment has been increasingly personalized, in part due to the development of new molecular tools. In this review, we discuss the old and new colon cancer chemotherapeutics, and the parameters that have been shown to be predictive of efficacy and safety of these chemotherapeutics. In addition, we discuss how alternate pharmaceuticals have been developed in light of a potential lack of response or resistance to a particular chemotherapeutic.
Collapse
Affiliation(s)
- Christina Leah B Kline
- Hematology/Oncology Division, Penn State Hershey Medical Center, Hershey, PA 17033, USA.
| | | |
Collapse
|
25
|
Pharmacogenetic variants in the DPYD, TYMS, CDA and MTHFR genes are clinically significant predictors of fluoropyrimidine toxicity. Br J Cancer 2013; 108:2505-15. [PMID: 23736036 PMCID: PMC3694243 DOI: 10.1038/bjc.2013.262] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background: Fluoropyrimidine drugs are extensively used for the treatment of solid cancers. However, adverse drug reactions are a major clinical problem, often necessitating treatment discontinuation. The aim of this study was to identify pharmacogenetic markers predicting fluoropyrimidine toxicity. Methods: Toxicity in the first four cycles of 5-fluorouracil or capecitabine-based chemotherapy were recorded for a series of 430 patients. The association between demographic variables, DPYD, DPYS, TYMS, MTHFR, CDA genotypes, and toxicity were analysed using logistic regression models. Results: Four DPYD sequence variants (c.1905+1G>A, c.2846A>T, c.1601G>A and c.1679T>G) were found in 6% of the cohort and were significantly associated with grade 3–4 toxicity (P<0.0001). The TYMS 3′-untranslated region del/del genotype substantially increased the risk of severe toxicity (P=0.0123, odds ratio (OR)=3.08, 95% confidence interval (CI): 1.38–6.87). For patients treated with capecitabine, a MTHFR c.1298CC homozygous variant genotype predicted hand–foot syndrome (P=4.1 × 10−6, OR=9.99, 95% CI: 3.84–27.8). The linked CDA c.−92A>G and CDA c.−451C>T variants predicted grade 2–4 diarrhoea (P=0.0055, OR=2.3, 95% CI: 1.3–4.2 and P=0.0082, OR=2.3, 95% CI: 1.3–4.2, respectively). Conclusion: We have identified a panel of clinically useful pharmacogenetic markers predicting toxicity to fluoropyrimidine therapy. Dose reduction should be considered in patients carrying these sequence variants.
Collapse
|
26
|
Offer SM, Wegner NJ, Fossum C, Wang K, Diasio RB. Phenotypic profiling of DPYD variations relevant to 5-fluorouracil sensitivity using real-time cellular analysis and in vitro measurement of enzyme activity. Cancer Res 2013; 73:1958-68. [PMID: 23328581 DOI: 10.1158/0008-5472.can-12-3858] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the 45 years since its development, the pyrimidine analog 5-fluorouracil (5-FU) has become an integral component of many cancer treatments, most notably for the management of colorectal cancer. An appreciable fraction of patients who receive 5-FU suffer severe adverse toxicities, which in extreme cases may result in death. Dihydropyrimidine dehydrogenase (DPD, encoded by DPYD) rapidly degrades 85% of administered 5-FU, and as such, limits the amount of drug available for conversion into active metabolites. Clinical studies have suggested that genetic variations in DPYD increase the risk for 5-FU toxicity, however, there is not a clear consensus about which variations are relevant predictors. In the present study, DPYD variants were expressed in mammalian cells, and the enzymatic activity of expressed protein was determined relative to wild-type (WT). Relative sensitivity to 5-FU for cells expressing DPYD variations was also measured. The DPYD*2A variant (exon 14 deletion caused by IVS14+1G>A) was confirmed to be catalytically inactive. Compared with WT, two variants, S534N and C29R, showed significantly higher enzymatic activity. Cells expressing S534N were more resistant to 5-FU-mediated toxicity compared with cells expressing WT DPYD. These findings support the hypothesis that selected DPYD alleles are protective against severe 5-FU toxicity, and, as a consequence, may decrease the effectiveness of 5-FU an antitumor drug in carriers. In addition, this study shows a method that may be useful for phenotyping other genetic variations in pharmacologically relevant pathways.
Collapse
Affiliation(s)
- Steven M Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Cancer Center, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
27
|
Mueller F, Büchel B, Köberle D, Schürch S, Pfister B, Krähenbühl S, Froehlich TK, Largiader CR, Joerger M. Gender-specific elimination of continuous-infusional 5-fluorouracil in patients with gastrointestinal malignancies: results from a prospective population pharmacokinetic study. Cancer Chemother Pharmacol 2012; 71:361-70. [PMID: 23139054 DOI: 10.1007/s00280-012-2018-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 10/21/2012] [Indexed: 01/02/2023]
Abstract
BACKGROUND This study was initiated to assess the quantitative impact of patient anthropometrics and dihydropyrimidine dehydrogenase (DPYD) mutations on the pharmacokinetics (PK) of 5-fluorouracil (5FU) and to explore limited sampling strategies of 5FU. PATIENTS AND METHODS We included 32 patients with gastrointestinal malignancies, receiving 46-h continuous-infusional 5FU and performed PK-sampling at baseline, 15, 30, 45 min, 1 and 2 h after the start of infusion and at the end of infusion, for 2 subsequent cycles. Plasma concentrations of 5FU, 5-fluorodihydrouracil (5FUH2), uracil (U) and 5,6-dihydrouracil (UH2) were determined using LC-MS/MS and submitted to population PK analysis using nonlinear mixed-effects modeling. Broad genotyping of DPYD was performed, and the potential impact of the DPYD genotype on the elimination of 5FU was assessed. Limited sampling strategies were evaluated for their accuracy to predict steady-state concentrations of 5FU (CSS(5FU)), using data simulations based on the final PK-model. RESULTS The area-under-the concentration-time curve of 5FU (AUC(5FU)) was found to be <20 mg h/L in 33 occasions (58 %), between 20 and 30 mg h/L in 17 occasions (30 %) and >30 mg h/L in 7 occasions (12 %). Men had a 26 % higher elimination of 5FU and a 18 % higher apparent elimination of 5FUH2. Accordingly, women had a higher AUC(5FU) compared to men (22 vs. 18 mg h/L, p = 0.04). No DPYD risk variants were found, and the DPYD variants detected (c.496A>G, c.1601G>A, c.1627A>G) were not significantly associated with the elimination of 5FU. Individual baseline UH(2)/U ratio was significantly associated with AUC(5FU) (R = -0.49, p < 0.001). Limited sampling strategies with time-points <3 h after the start of infusion were not adequate to predict CSS(5FU). Female gender was the only predictor of nausea/emesis in the multivariate model. CONCLUSIONS Gender-specific elimination of 5FU is supported by the present data and may partly explain the gender-specific association between DPYD risk variants and 5FU-specific toxicity.
Collapse
Affiliation(s)
- F Mueller
- Department of Internal Medicine, Cantonal Hospital, St Gallen, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Corrigan A, Arenas-Hernandez M, Blaker P, Sanderson J, Marinaki A. Let's get personal: predicting thiopurine and fluoropyrimidine toxicity. Per Med 2012; 9:859-870. [PMID: 29776234 DOI: 10.2217/pme.12.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The US FDA now recognizes the need to individualize treatment paradigms using biomarkers that predict response to therapy. In clinical practice the best example of this is TPMT testing, which is used to rationalize the starting dose of azathioprine and mercaptopurine. The more recent addition of drug metabolite monitoring means that thiopurine therapy can now be personalized to unprecedented levels. Of interest, parallels exist between TPMT deficiency as an explanation for thiopurine toxicity and DPD deficiency in fluoropyrimidine toxicity. For these drugs, variations in a single locus predict severe toxicity. However, while TPMT testing has translated into routine clinical practice, DPD testing has not. This article summarizes the recent research investigating interindividual differences in the metabolism of thiopurine and fluoropyrimidine drugs, and explores the attitudes which influence the uptake of pharmacogenetic testing.
Collapse
Affiliation(s)
- Adele Corrigan
- Purine Research Laboratory, GSTS Pathology, Guy's & St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Monica Arenas-Hernandez
- Purine Research Laboratory, GSTS Pathology, Guy's & St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Paul Blaker
- Department of Gastroenterology, 1st Floor College House, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Jeremy Sanderson
- Department of Gastroenterology, 1st Floor College House, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Anthony Marinaki
- Purine Research Laboratory, GSTS Pathology, Guy's & St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK.
| |
Collapse
|
29
|
Amstutz U, Froehlich TK, Largiadèr CR. Dihydropyrimidine dehydrogenase gene as a major predictor of severe 5-fluorouracil toxicity. Pharmacogenomics 2012; 12:1321-36. [PMID: 21919607 DOI: 10.2217/pgs.11.72] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of polymorphisms in the dihydropyrimidine dehydrogenase (DPD) gene (DPYD) for the prediction of severe toxicity in 5-fluorouracil (5-FU) based chemotherapy has been controversially debated. As a key enzyme in the catabolism of 5-FU, DPD is the top candidate for pharmacogenetic studies on 5-FU toxicity, since a reduced DPD activity is thought to result in an increased half-life of the drug, and thus, an increased risk of toxicity. Here, we review the current knowledge on well-known and frequently studied DPYD variants such as the c.1905+1G>A splice site variant, as well as the recent discoveries of important functional variation in the noncoding regions of DPYD. We also outline future directions that are needed to further improve the risk assessment of 5-FU toxicity, in particular with respect to metabolic profiling and in the context of different combination therapeutic regimens, in which 5-FU is used today.
Collapse
Affiliation(s)
- Ursula Amstutz
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, & University of Bern, INO F, CH-3010 Bern, Switzerland
| | | | | |
Collapse
|
30
|
Ciccolini J, Gross E, Dahan L, Lacarelle B, Mercier C. Routine dihydropyrimidine dehydrogenase testing for anticipating 5-fluorouracil-related severe toxicities: hype or hope? Clin Colorectal Cancer 2011; 9:224-8. [PMID: 20920994 DOI: 10.3816/ccc.2010.n.033] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
5-Fluorouracil (5-FU) is a mainstay for treating colorectal cancer, alone or more frequently as part of combination therapies. However, its efficacy/toxicity balance is often limited by the occurrence of severe toxicities, showing in about 15%-20% of patients. Several clinical reports have shown the deleterious effect of dihydropyrimidine dehydrogenase (DPD) genetic polymorphism, a condition that reduces the liver detoxification step of standard dosages of 5-FU, in patients undergoing fluoropyrimidine-based therapy. Admittedly, DPD deficiency accounts for 50%-75% of the severe and sometimes life-threatening toxicities associated with 5-FU (or oral 5-FU). However, technical consensus on the best way to identify patients with DPD deficiency before administrating 5-FU is far from being achieved. Consequently, no regulatory step has been undertaken yet to recommend DPD testing as part of routine clinical practice for securing the administration of 5-FU. This review covers the limits and achievements of the various strategies proposed so far for determining DPD status in patients scheduled for 5-FU therapy.
Collapse
|
31
|
|
32
|
Obi EE, McDonald A, Kemp E. A bilateral cicatricial ectropion and bilateral upper lid shortening caused by 5-fluorouracil toxicity in a patient with dihydropyrimidine dehydrogenase deficiency. Cutan Ocul Toxicol 2010; 30:157-9. [DOI: 10.3109/15569527.2010.532846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
van Kuilenburg ABP, Meijer J, Mul ANPM, Meinsma R, Schmid V, Dobritzsch D, Hennekam RCM, Mannens MMAM, Kiechle M, Etienne-Grimaldi MC, Klümpen HJ, Maring JG, Derleyn VA, Maartense E, Milano G, Vijzelaar R, Gross E. Intragenic deletions and a deep intronic mutation affecting pre-mRNA splicing in the dihydropyrimidine dehydrogenase gene as novel mechanisms causing 5-fluorouracil toxicity. Hum Genet 2010; 128:529-38. [PMID: 20803296 PMCID: PMC2955237 DOI: 10.1007/s00439-010-0879-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 08/17/2010] [Indexed: 02/06/2023]
Abstract
Dihydropyrimidine dehydrogenase (DPD) is the initial enzyme acting in the catabolism of the widely used antineoplastic agent 5-fluorouracil (5FU). DPD deficiency is known to cause a potentially lethal toxicity following administration of 5FU. Here, we report novel genetic mechanisms underlying DPD deficiency in patients presenting with grade III/IV 5FU-associated toxicity. In one patient a genomic DPYD deletion of exons 21–23 was observed. In five patients a deep intronic mutation c.1129–5923C>G was identified creating a cryptic splice donor site. As a consequence, a 44 bp fragment corresponding to nucleotides c.1129–5967 to c.1129–5924 of intron 10 was inserted in the mature DPD mRNA. The deleterious c.1129–5923C>G mutation proved to be in cis with three intronic polymorphisms (c.483 + 18G>A, c.959–51T>G, c.680 + 139G>A) and the synonymous mutation c.1236G>A of a previously identified haplotype. Retrospective analysis of 203 cancer patients showed that the c.1129–5923C>G mutation was significantly enriched in patients with severe 5FU-associated toxicity (9.1%) compared to patients without toxicity (2.2%). In addition, a high prevalence was observed for the c.1129–5923C>G mutation in the normal Dutch (2.6%) and German (3.3%) population. Our study demonstrates that a genomic deletion affecting DPYD and a deep intronic mutation affecting pre-mRNA splicing can cause severe 5FU-associated toxicity. We conclude that screening for DPD deficiency should include a search for genomic rearrangements and aberrant splicing.
Collapse
Affiliation(s)
- André B P van Kuilenburg
- Department of Clinical Chemistry, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2010. [DOI: 10.1002/pds.1852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|