1
|
de Moraes FCA, de Almeida Barbosa AB, Sano VKT, Kelly FA, Burbano RMR. Pharmacogenetics of DPYD and treatment-related mortality on fluoropyrimidine chemotherapy for cancer patients: a meta-analysis and trial sequential analysis. BMC Cancer 2024; 24:1210. [PMID: 39350200 PMCID: PMC11441158 DOI: 10.1186/s12885-024-12981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Fluoropyrimidines are chemotherapy drugs utilized to treat a variety of solid tumors. These drugs predominantly rely on the enzyme dihydropyrimidine dehydrogenase (DPD), which is encoded by the DPYD gene, for their metabolism. Genetic mutations affecting this gene can cause DPYD deficiency, disrupting pyrimidine metabolism and increasing the risk of toxicity in cancer patients treated with 5-fluorouracil. The severity and type of toxic reactions are influenced by genetic and demographic factors and, in certain instances, can result in patient mortality. Among the more than 50 identified variants of DPYD, only a subset has clinical significance, leading to the production of enzymes that are either non-functional or impaired. The study aims to examine treatment-related mortality in cancer patients undergoing fluoropyrimidine chemotherapy, comparing those with and without DPD deficiency. METHODS The meta-analysis selected and evaluated 9685 studies from Pubmed, Cochrane, Embase and Web of Science databases. Only studies examining the main DPYD variants (DPYD*2A, DPYD p.D949V, DPYD*13 and DPYD HapB3) were included. Statistical Analysis was performed using R, version 4.2.3. Data were examined using the Mantel-Haenszel method and 95% CIs. Heterogeneity was assessed with I2 statistics. RESULTS There were 36 prospective and retrospective studies included, accounting for 16,005 patients. Most studies assessed colorectal cancer, representing 86.49% of patients. Other gastrointestinal cancers were evaluated by 11 studies, breast cancer by nine studies and head and neck cancers by five studies. Four DPYD variants were identified as predictors of severe fluoropyrimidines toxicity in literature review: DPYD*2A (rs3918290), DPYD p.D949V (rs67376798), DPYD*13 (rs55886062) and DPYD Hap23 (rs56038477). All 36 studies assessed the DPYD*2A variant, while 20 assessed DPYD p.D949V, 7 assessed DPYD*13, and 9 assessed DPYDHap23. Among the 587 patients who tested positive for at least one DPYD variant, 13 died from fluoropyrimidine toxicity. Conversely, in the non-carrier group there were 14 treatment-related deaths. Carriers of DPYD variants was found to be significantly correlated with treatment-related mortality (OR = 34.86, 95% CI 13.96-87.05; p < 0.05). CONCLUSIONS This study improves our comprehension of how the DPYD gene impacts cancer patients receiving fluoropyrimidine chemotherapy. Identifying mutations associated with dihydropyrimidine dehydrogenase deficiency may help predict the likelihood of serious side effects and fatalities. This knowledge can be applied to adjust medication doses before starting treatment, thus reducing the occurrence of these critical outcomes.
Collapse
|
2
|
Chan TH, Zhang JE, Pirmohamed M. DPYD genetic polymorphisms in non-European patients with severe fluoropyrimidine-related toxicity: a systematic review. Br J Cancer 2024; 131:498-514. [PMID: 38886557 PMCID: PMC11300675 DOI: 10.1038/s41416-024-02754-z] [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: 12/08/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Pre-treatment DPYD screening is mandated in the UK and EU to reduce the risk of severe and potentially fatal fluoropyrimidine-related toxicity. Four DPYD gene variants which are more prominently found in Europeans are tested. METHODS Our systematic review in patients of non-European ancestry followed PRISMA guidelines to identify relevant articles up to April 2023. Published in silico functional predictions and in vitro functional data were also extracted. We also undertook in silico prediction for all DPYD variants identified. RESULTS In 32 studies, published between 1998 and 2022, 53 DPYD variants were evaluated in patients from 12 countries encompassing 5 ethnic groups: African American, East Asian, Latin American, Middle Eastern, and South Asian. One of the 4 common European DPYD variants, c.1905+1G>A, is also present in South Asian, East Asian and Middle Eastern patients with severe fluoropyrimidine-related toxicity. There seems to be relatively strong evidence for the c.557A>G variant, which is found in individuals of African ancestry, but is not currently included in the UK genotyping panel. CONCLUSION Extending UK pre-treatment DPYD screening to include variants that are present in some non-European ancestry groups will improve patient safety and reduce race and health inequalities in ethnically diverse societies.
Collapse
Affiliation(s)
- Tsun Ho Chan
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - J Eunice Zhang
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Munir Pirmohamed
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, 1-5 Brownlow Street, Liverpool, L69 3GL, UK.
| |
Collapse
|
3
|
Martin JH, Galettis P, Flynn A, Schneider J. Phenotype versus genotype to optimize cancer dosing in the clinical setting-focus on 5-fluorouracil and tyrosine kinase inhibitors. Pharmacol Res Perspect 2024; 12:e1182. [PMID: 38429945 PMCID: PMC10907881 DOI: 10.1002/prp2.1182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
Cancer medicines often have narrow therapeutic windows; toxicity can be severe and sometimes fatal, but inadequate dose intensity reduces efficacy and survival. Determining the optimal dose for each patient is difficult, with body-surface area used most commonly for chemotherapy and flat dosing for tyrosine kinase inhibitors, despite accumulating evidence of a wide range of exposures in individual patients with many receiving a suboptimal dose with these strategies. Therapeutic drug monitoring (measuring the drug concentration in a biological fluid, usually plasma) (TDM) is an accepted and well validated method to guide dose adjustments for individual patients to improve this. However, implementing TDM in routine care has been difficult outside a research context. The development of genotyping of various proteins involved in drug elimination and activity has gained prominence, with several but not all Guideline groups recommending dose reductions for particular variant genotypes. However, there is increasing concern that dosing recommendations are based on limited data sets and may lead to unnecessary underdosing and increased cancer mortality. This Review discusses the evidence surrounding genotyping and TDM to guide decisions around best practice.
Collapse
Affiliation(s)
- Jennifer H. Martin
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Peter Galettis
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alex Flynn
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Jennifer Schneider
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| |
Collapse
|
4
|
Alkhatib O, Miles T, Jones RP, Mair R, Palmer R, Winter H, McDermott FD. Current and future genomic applications for surgeons. Ann R Coll Surg Engl 2024; 106:321-328. [PMID: 38555869 PMCID: PMC10981988 DOI: 10.1308/rcsann.2024.0031] [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] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Genomics is a crucial part of managing surgical disease. This review focuses on some of the genomic advances that are available now and looks to the future of their application in surgical practice. Whole-genome sequencing enables unbiased coverage across the entire human genome of approximately three billion base pairs. Newer technologies, such as those that permit long-read sequence analysis, provide additional information in longer phased fragment and base pair epigenomic (methylomic) data. Whole-genome sequencing is currently available in England for cancers in children, teenagers and young adults, central nervous system tumours, sarcoma and haematological malignancies. Circulating tumour DNA (ctDNA), immunotherapy and pharmacogenomics have emerged as groundbreaking approaches in the field of cancer treatment. These are now revolutionising the way oncologists and surgeons approach curative cancer surgery. Cancer vaccines offer an innovative approach to reducing recurrence after surgery by priming the immune system to trigger an immune response. The Cancer Vaccine Launch Pad project facilitates cancer vaccine studies in England. The BNT122-01 trial is recruiting patients with ctDNA-positive high-risk colorectal cancer after surgery to assess the impact of cancer vaccines. The evolving landscape of cancer treatment demands a dynamic and integrated approach from the surgical multidisciplinary team. Immunotherapy, ctDNA, pharmacogenomics, vaccines, mainstreaming and whole-genome sequencing are just some of the innovations that have the potential to redefine the standards of care. The continued exploration of these innovative diagnostics and therapies, the genomic pathway evolution and their application in diverse cancer types highlights the transformative impact of precision medicine in surgery.
Collapse
Affiliation(s)
- O Alkhatib
- Liverpool University Teaching Hospitals NHS Foundation Trust, UK
| | - T Miles
- Southwest Genomics Medicine Service Alliance, UK
| | | | | | | | - H Winter
- University Hospitals Bristol and Weston NHS Foundation Trust, UK
| | | |
Collapse
|
5
|
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
|
6
|
Mondello A, Dal Bo M, Toffoli G, Polano M. Machine learning in onco-pharmacogenomics: a path to precision medicine with many challenges. Front Pharmacol 2024; 14:1260276. [PMID: 38264526 PMCID: PMC10803549 DOI: 10.3389/fphar.2023.1260276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Over the past two decades, Next-Generation Sequencing (NGS) has revolutionized the approach to cancer research. Applications of NGS include the identification of tumor specific alterations that can influence tumor pathobiology and also impact diagnosis, prognosis and therapeutic options. Pharmacogenomics (PGx) studies the role of inheritance of individual genetic patterns in drug response and has taken advantage of NGS technology as it provides access to high-throughput data that can, however, be difficult to manage. Machine learning (ML) has recently been used in the life sciences to discover hidden patterns from complex NGS data and to solve various PGx problems. In this review, we provide a comprehensive overview of the NGS approaches that can be employed and the different PGx studies implicating the use of NGS data. We also provide an excursus of the ML algorithms that can exert a role as fundamental strategies in the PGx field to improve personalized medicine in cancer.
Collapse
Affiliation(s)
| | | | | | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
7
|
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
|
8
|
Wang Y, Hu H, Yu L, Zeng S. Physiologically Based Pharmacokinetic Modeling for Prediction of 5-FU Pharmacokinetics in Cancer Patients with Hepatic Impairment After 5-FU and Capecitabine Administration. Pharm Res 2023; 40:2177-2194. [PMID: 37610618 DOI: 10.1007/s11095-023-03585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE 5-fluorouracil (5-FU) and its prodrug capecitabine are commonly prescribed anti-tumor medications. We aimed to establish physiologically based pharmacokinetic (PBPK) models of capecitabine-metabolites and 5-FU-metabolites to describe their pharmacokinetics in tumor and plasma of cancer patients with liver impairment. METHODS Models including the cancer compartment were developed in PK-Sim® and MoBi® and evaluated by R programming language with 25 oral capecitabine and 18 intravenous 5-FU studies for cancer patients with and without liver impairment. RESULTS The PBPK models were constructed successfully as most simulated Cmax and AUClast were within two-fold error of observed values. The simulated alterations of tumor 5-FU Cmax and AUClast in cancer patients with severe liver injury compared with normal liver function were 1.956 and 3.676 after oral administration of capecitabine, but no significant alteration was observed after intravenous injection of 5-FU. Besides, 5-FU concentration in tumor tissue increases with higher tumor blood flow but not tumor size. Sensitivity analysis revealed that dihydropyrimidine dehydrogenase (DPD) and other metabolic enzymes' activity, capecitabine intestinal permeability and plasma protein scale factor played a vital role in tumor and plasma 5-FU pharmacokinetics. CONCLUSIONS PBPK model prediction suggests no dosage adaption of capecitabine or 5-FU is required for cancer patients with hepatic impairment but it would be reduced when the toxic reaction is observed. Furthermore, tumor blood flow rate rather than tumor size is critical for 5-FU concentration in tumor. In summary, these models could predict pharmacokinetics of 5-FU in tumor in cancer patients with varying characteristics in different scenarios.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310006, China
| | - Haihong Hu
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310006, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310006, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
9
|
Baker SD, Bates SE, Brooks GA, Dahut WL, Diasio RB, El-Deiry WS, Evans WE, Figg WD, Hertz DL, Hicks JK, Kamath S, Kasi PM, Knepper TC, McLeod HL, O'Donnell PH, Relling MV, Rudek MA, Sissung TM, Smith DM, Sparreboom A, Swain SM, Walko CM. DPYD Testing: Time to Put Patient Safety First. J Clin Oncol 2023; 41:2701-2705. [PMID: 36821823 PMCID: PMC10414691 DOI: 10.1200/jco.22.02364] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 02/25/2023] Open
Affiliation(s)
- Sharyn D. Baker
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Susan E. Bates
- Herbert Irving Comprehensive Cancer Center, Columbia University, Irving Medical Center, New York, NY
| | | | | | | | | | | | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD
| | - Dan L. Hertz
- College of Pharmacy, University of Michigan, Ann Arbor, MI
| | - J. Kevin Hicks
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL
| | - Suneel Kamath
- Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| | | | - Todd C. Knepper
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL
| | | | | | | | | | | | - D. Max Smith
- Georgetown Lombardi Comprehensive Cancer Center and MedStar Health, Georgetown University, Washington, DC
| | - Alex Sparreboom
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Sandra M. Swain
- Georgetown Lombardi Comprehensive Cancer Center and MedStar Health, Georgetown University, Washington, DC
| | - Christine M. Walko
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
10
|
Miarons M, Manzaneque Gordón A, Riera P, Gutiérrez Nicolás F. Allelic Frequency of DPYD Genetic Variants in Patients With Cancer in Spain: The PhotoDPYD Study. Oncologist 2023; 28:e304-e308. [PMID: 37014829 PMCID: PMC10166167 DOI: 10.1093/oncolo/oyad077] [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: 09/28/2022] [Accepted: 02/21/2023] [Indexed: 04/05/2023] Open
Abstract
INTRODUCTION Identifying polymorphisms in the dihydropyrimidine dehydrogenase (DPYD) gene is gaining importance to be able to predict fluoropyrimidine-associated toxicity. The aim of this project was to describe the frequency of the DPYD variants DPYD*2A (rs3918290); c.1679T>G (rs55886062); c.2846A>T (rs67376798) and c.1129-5923C>G (rs75017182; HapB3) in the Spanish oncological patients. MATERIAL AND METHODS Cross-sectional and multicentric study (PhotoDPYD study) conducted in hospitals located in Spain designed to register the frequency of the most relevant DPYD genetic variants in oncological patients. All oncological patients with DPYD genotype were recruited in the participant hospitals. The measures determined where the presence or not of the 4 DPYD previously described variants. RESULTS Blood samples from 8054 patients with cancer from 40 different hospitals were used to determine the prevalence of the 4 variants located in the DPYD gene. The frequency of carriers of one defective DPYD variant was 4.9%. The most frequently identified variant was c.1129-5923C>G (rs75017182) (HapB3), in 2.9%, followed by c.2846A>T (rs67376798) in 1.4%, c.1905 + 1G>A (rs3918290, DPYD*2A) in 0.7% and c.1679T>G (rs55886062) in 0.2% of the patients. Only 7 patients (0.08%) were carrying the c.1129-5923C>G (rs75017182) (HapB3) variant, 3 (0.04%) the c.1905 + 1G>A (rs3918290, DPYD*2A) and one (0.01%) the DPYD c.2846A>T (rs67376798, p.D949V) variant in homozygosis. Moreover, 0.07% were compound heterozygous patients, 3 carrying the DPYD variants DPYD*2A + c.2846A>T, 2 the DPYD c.1129-5923C>G + c.2846A>T and one the DPYD*2A + c.1129-5923C>G variants. CONCLUSIONS Our results demonstrate the relatively high frequency of DPYD genetic variants in the Spanish patient with cancer population, which highlights the relevance of their determination before initiating a fluoropirimidine-containing regimen.
Collapse
Affiliation(s)
- Marta Miarons
- Pharmacy Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital, Campus, Barcelona, Spain
- ReDPyD group from the Spanish Society of Hospital Pharmacy (SEFH), Tenerife, Canarias, Spain
| | - Alba Manzaneque Gordón
- Pharmacy Department, Hospital Mutua de Terrassa, Terrassa, Barcelona, Spain
- ReDPyD group from the Spanish Society of Hospital Pharmacy (SEFH), Tenerife, Canarias, Spain
| | - Pau Riera
- Pharmacy Department, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, Carrer Sant Quintí, Barcelona, Spain
- U705, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
- ReDPyD group from the Spanish Society of Hospital Pharmacy (SEFH), Tenerife, Canarias, Spain
| | - Fernando Gutiérrez Nicolás
- Pharmacy Department, Research Unit Hospital Universitario de Canarias (CHUC), Tenerife, Canarias, Spain
- ReDPyD group from the Spanish Society of Hospital Pharmacy (SEFH), Tenerife, Canarias, Spain
| |
Collapse
|
11
|
Etienne-Grimaldi MC, Pallet N, Boige V, Ciccolini J, Chouchana L, Barin-Le Guellec C, Zaanan A, Narjoz C, Taieb J, Thomas F, Loriot MA. Current diagnostic and clinical issues of screening for dihydropyrimidine dehydrogenase deficiency. Eur J Cancer 2023; 181:3-17. [PMID: 36621118 DOI: 10.1016/j.ejca.2022.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
Fluoropyrimidine drugs (FP) are the backbone of many chemotherapy protocols for treating solid tumours. The rate-limiting step of fluoropyrimidine catabolism is dihydropyrimidine dehydrogenase (DPD), and deficiency in DPD activity can result in severe and even fatal toxicity. In this review, we survey the evidence-based pharmacogenetics and therapeutic recommendations regarding DPYD (the gene encoding DPD) genotyping and DPD phenotyping to prevent toxicity and optimize dosing adaptation before FP administration. The French experience of mandatory DPD-deficiency screening prior to initiating FP is discussed.
Collapse
Affiliation(s)
| | - Nicolas Pallet
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France
| | - Valérie Boige
- Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France; Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Joseph Ciccolini
- SMARTc, CRCM INSERM U1068, Université Aix-Marseille, Marseille, France; Laboratory of Pharmacokinetics and Toxicology, Hôpital Universitaire La Timone, F-13385 Marseille, France; COMPO, CRCM INSERM U1068-Inria, Université Aix-Marseille, Marseille, France
| | - Laurent Chouchana
- Regional Center of Pharmacovigilance, Department of Pharmacology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, Université de Paris, Paris, France; French Pharmacovigilance Network, France
| | - Chantal Barin-Le Guellec
- Laboratory of Biochemistry and Molecular Biology, Centre Hospitalo-uinversitaire de Tours, Tours, France; INSERM U1248, IPPRITT, University of Limoges, Limoges, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris University; Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Céline Narjoz
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France
| | - Julien Taieb
- SIRIC CARPEM, Université de Paris; Fédération Francophone de Cancérologie Digestive (FFCD), Assistance Publique-Hôpitaux de Paris, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Fabienne Thomas
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole and CRCT, INSERM UMR1037, Université Paul Sabatier, Toulouse, France
| | - Marie-Anne Loriot
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France.
| | | |
Collapse
|
12
|
Medwid S, Wigle TJ, Kim RB. Fluoropyrimidine-associated toxicity and DPYD variants c.85T>C, c.496A>G, and c.1236G>A: impact of haplotype. Cancer Chemother Pharmacol 2023; 91:97-102. [PMID: 36357798 DOI: 10.1007/s00280-022-04491-7] [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: 08/19/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022]
Abstract
Dihydropyrimidine dehydrogenase (DPYD) is the rate-limiting step in fluoropyrimidines metabolism. Currently, genotype-guided fluoropyrimidine dosing is recommended for four DPYD single nucleotide variants (SNVs). However, the clinical impact of additional DPYD SNVs on fluoropyrimidine-related toxicity remains controversial. We assessed common DPYD SNVs c.85T>C, and c.496A>G which are often in linkage disequilibrium with c.1236G>A, a variant currently recommended for DPYD genotyping, in a retrospective cohort of cancer patients who had received fluoropyrimidines (N = 1371). When assessing individual SNVs, during the total chemotherapy treatment period, a significant increased risk of severe grade ≥ 3 toxicity was seen in carriers of c.496A>G (OR = 1.38, 95% CI 1.01-1.88, p = 0.0405) after adjusting for age, sex and treatment drug (capecitabine or 5-Fluorouracil). No association with fluoropyrimidine-related toxicity was seen in patients given standard dosing among those carrying one allele of DPYD c.1236G>A (OR = 1.19, 95% CI 0.59-2.27, p = 0.6147) or c.85T>C (OR = 1.04, 95% CI 0.80-1.62, p = 0.7536). Haplotype analysis confirmed a high linkage disequilibrium of these three variants. Toxicity was not significantly increased in haplotypes containing only one of c.85T>C or c.496A>G or c.1236G>A alleles. However, the haplotype containing both c.85T>C and c.496A>G alleles, which had a predicted frequency of 7.1%, was associated with an increased risk of fluoropyrimidine toxicity (OR = 1.57, 95% CI 1.15-2.13, p = 0.0041). This study suggests DPYD haplotype structure may help explain previous conflicting studies concerning the impact of these variants. Our findings suggest patients with both DPYD c.85T>C and c.496A>G variants have a significant increased risk for toxicity and may potentially benefit from genotype-guided fluoropyrimidine dosing.
Collapse
Affiliation(s)
- Samantha Medwid
- London Health Sciences Centre, London, ON, Canada.,Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Theodore J Wigle
- London Health Sciences Centre, London, ON, Canada.,Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Richard B Kim
- London Health Sciences Centre, London, ON, Canada. .,Department of Medicine, University of Western Ontario, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
13
|
Lombardi P, Aimar G, Peraldo-Neia C, Bonzano A, Depetris I, Fenocchio E, Filippi R, Quarà V, Milanesio M, Cavalloni G, Gammaitoni L, Basiricò M, Cagnazzo C, Ostano P, Chiorino G, Aglietta M, Leone F. Fluoropyrimidine‑induced cardiotoxicity in colorectal cancer patients: a prospective observational trial (CHECKPOINT). Oncol Rep 2022; 49:31. [PMID: 36562382 PMCID: PMC9827273 DOI: 10.3892/or.2022.8468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Fluoropyrimidines (FP) are the backbone chemotherapy in colorectal cancer (CRC) treatment; however, their use is associated with cardiotoxicity, which is underreported. In the present study, it was aimed to prospectively determine the incidence rates and related risk factors of FP‑induced cardiotoxicity (FIC) in CRC patients and at identifying predictive biomarkers. A total of 129 consecutive previously untreated CRC patients underwent active cardiological monitoring, including 5‑items simplified questionnaire on symptoms, electrocardiogram (ECG) and plasma sample collection during FP chemotherapy. FIC was defined as the presence of ECG alterations and/or the arising of at least one symptom of chest pain, dyspnoea, palpitations or syncope. The primary objective was the evaluation of FIC incidence. Secondary objectives were the correlation of FIC with well‑known cardiological risk factors and the identification of circulating biomarkers (serum levels of troponin I, pro hormone BNP; miRNA analysis) as predictors of FIC. A total of 20 out of 129 (15.5%) patients experienced FIC. The most common symptoms were dyspnoea (60%) and chest pain (40%), while only 15% of patients presented ECG alterations, including one acute myocardial infarction. Retreatment with FP was attempted in 90% of patients with a favourable outcome. Despite 48% of patients having cardiological comorbidities, an increased FIC was not observed in this subgroup. Only the subgroup of females with the habit of alcohol consumption showed an increased risk of FIC. None of the circulating biomarkers evaluated demonstrated a clinical utility as FIC predictors. FIC can be an unexpected, life‑threatening adverse event that can limit the subsequent treatment choices in patients with CRC. In this prospective study, well‑known cardiological comorbidities were not related to higher FIC risk and circulating biomarkers predictive of toxicity could not be found. With careful monitoring, mainly based on symptoms, almost all patients completed the FP treatment.
Collapse
Affiliation(s)
- Pasquale Lombardi
- Department of Oncology, University of Turin, I-10124 Torino, Italy,Phase 1 Unit, Agostino Gemelli Foundation University Hospital IRCCS, I-00168 Roma, Italy
| | - Giacomo Aimar
- Department of Oncology, University of Turin, I-10124 Torino, Italy,Department of Oncology, S. Croce and Carle Hospital, I-12100 Cuneo, Italy
| | | | | | - Ilaria Depetris
- Division of Medical Oncology 1, AOU City of Health and Science of Turin, I-12126 Turin, Italy
| | - Elisabetta Fenocchio
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, I-10060 Candiolo, Italy
| | - Roberto Filippi
- Department of Oncology, University of Turin, I-10124 Torino, Italy,Division of Medical Oncology 1, AOU City of Health and Science of Turin, I-12126 Turin, Italy
| | - Virginia Quarà
- Department of Oncology, University of Turin, I-10124 Torino, Italy,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, I-10060 Candiolo, Italy
| | | | - Giuliana Cavalloni
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, I-10060 Candiolo, Italy
| | | | - Marco Basiricò
- Department of Public Health and Pediatric Sciences, AOU City of Health and Science of Turin, Regina Margherita Hospital, I-10126 Torino, Italy
| | - Celeste Cagnazzo
- Department of Public Health and Pediatric Sciences, AOU City of Health and Science of Turin, Regina Margherita Hospital, I-10126 Torino, Italy
| | - Paola Ostano
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, I-13900 Biella, Italy
| | - Giovanna Chiorino
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, I-13900 Biella, Italy
| | - Massimo Aglietta
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, I-10060 Candiolo, Italy
| | - Francesco Leone
- Department of Medical Oncology, Infermi Hospital of Biella, Ponderano, I-13875 Biella, Italy,Correspondence to: Dr Francesco Leone, Department of Medical Oncology, Infermi Hospital of Biella, 2 Via dei Ponderanesi, Ponderano, I-13875 Biella, Italy, E-mail:
| |
Collapse
|
14
|
Khalij Y, Belaid I, Chouchane S, Amor D, Omezzine A, Ben Rejeb N, Ben Ahmed S, Bouslama A. DPYD and TYMS polymorphisms as predictors of 5 fluorouracil toxicity in colorectal cancer patients. J Chemother 2022:1-10. [PMID: 36137946 DOI: 10.1080/1120009x.2022.2125736] [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: 11/09/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth leading cause of cancer death. 5-Fluorouracil (5-FU) is an essential component of systemic chemotherapy for CRC. Our objective was to determine the genotypic frequency of polymorphisms affecting dihydropyrimidine dehydrogenase (DPYD) and thymidylate synthetase (TYMS) genes and to correlate the genetic profile with the toxicity due to 5-FU, also considering nongenetic factors. This is a prospective study that involved 66 patients. We extracted DNA by salting out methods. We carried out the genotyping of the different polymorphisms by simple PCR for the TYMS 5'UTR and by PCR-RFLP for DPYD: 1905 + 1 G > A, 85 T > C, 496 A > G, 1679 T > G, c.483 + 18G > A and the TYMS: 5'UTR VNTR, 5'UTR G > C and 3'UTR. The study of the association of DPYD and TYMS polymorphisms with the various signs of toxicity under 5-FU revealed that the polymorphisms 496 A > G were significantly associated with hepatotoxicity: OR = 3.85 (p = 0.04). In addition, 85 T > C was significantly associated with mucositis and neurotoxicity: OR = 4.35 (p = 0.03), OR = 3.79 (p = 0.02). For TYMS, the only significant association we observed for 5'UTR with vomiting: OR = 3.34 (p = 0.04). The incidence of adverse reactions related to 5-FU appears to be influenced in patients with CRC by the identified DPYD and TYMS gene polymorphisms in the Tunisian population.
Collapse
Affiliation(s)
- Yassine Khalij
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia.,University of Monastir Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Imtinen Belaid
- Carcinology Department, Farhat Hached University Hospital, Sousse, Tunisia
| | - Sana Chouchane
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia.,University of Monastir Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Dorra Amor
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia.,University of Monastir Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Asma Omezzine
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia.,University of Monastir Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Nabila Ben Rejeb
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia.,University of Monastir Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Slim Ben Ahmed
- Carcinology Department, Farhat Hached University Hospital, Sousse, Tunisia
| | - Ali Bouslama
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia
| |
Collapse
|
15
|
Farinango C, Gallardo-Cóndor J, Freire-Paspuel B, Flores-Espinoza R, Jaramillo-Koupermann G, López-Cortés A, Burgos G, Tejera E, Cabrera-Andrade A. Genetic Variations of the DPYD Gene and Its Relationship with Ancestry Proportions in Different Ecuadorian Trihybrid Populations. J Pers Med 2022; 12:jpm12060950. [PMID: 35743735 PMCID: PMC9225136 DOI: 10.3390/jpm12060950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Dihydropyrimidine dehydrogenase is one of the main pharmacological metabolizers of fluoropyrimidines, a group of drugs widely used in clinical oncology. Around 20 to 30% of patients treated with fluoropyrimidines experience severe toxicity caused by a partial or total decrease in enzymatic activity. This decrease is due to molecular variants in the DPYD gene. Their prevalence and allelic frequencies vary considerably worldwide, so their description in heterogeneous groups such as the Ecuadorian population will allow for the description of pharmacogenetic variants and proper characterization of this population. Thus, we genotyped all the molecular variants with a predictive value for DPYD in a total of 410 Ecuadorian individuals belonging to Mestizo, Afro-Ecuadorian, and Indigenous ethnic groups. Moreover, we developed a genetic ancestry analysis using 46 autosomal ancestry informative markers. We determined 20 genetic variations in 5 amplified regions, including 3 novel single nucleotide variants. The allele frequencies for DPYD variants c.1627G>A (*5, rs1801159), c.1129-15T>C (rs56293913), c.1218G>A (rs61622928), rs1337752, rs141050810, rs2786783, rs2811178, and g.97450142G>A (chr1, GRCh38.p13) are significantly related to Native American and African ancestry proportions. In addition, the FST calculated from these variants demonstrates the closeness between Indigenous and Mestizo populations, and evidences genetic divergence between Afro-Ecuadorian groups when compared with Mestizo and Indigenous ethnic groups. In conclusion, the genetic variability in the DPYD gene is related to the genetic component of ancestral populations in different Ecuadorian ethnic groups. The absence and low frequency of variants with predictive value for fluoropyrimidine toxicity such as DPYD *2A, HapB3, and c.2846A>T (prevalent in populations with European ancestry) is consistent with the genetic background found.
Collapse
Affiliation(s)
- Camila Farinango
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito 170125, Ecuador; (C.F.); (J.G.-C.); (E.T.)
| | - Jennifer Gallardo-Cóndor
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito 170125, Ecuador; (C.F.); (J.G.-C.); (E.T.)
| | - Byron Freire-Paspuel
- Laboratorios de Investigación, Universidad de Las Américas, Quito 170125, Ecuador; (B.F.-P.); (R.F.-E.)
- Vall d’Hebron Research Institute, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain
| | - Rodrigo Flores-Espinoza
- Laboratorios de Investigación, Universidad de Las Américas, Quito 170125, Ecuador; (B.F.-P.); (R.F.-E.)
- Laboratório de Diagnóstico por DNA (LDD), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
| | - Gabriela Jaramillo-Koupermann
- Laboratorio de Biología Molecular, Subproceso de Anatomía Patológica, Hospital de Especialidades Eugenio Espejo, Quito 170403, Ecuador;
| | - Andrés López-Cortés
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador; (A.L.-C.); (G.B.)
- Programa de Investigación en Salud Global, Facultad de Ciencias de la Salud, Universidad Internacional SEK, Quito 170302, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), 28001 Madrid, Spain
| | - Germán Burgos
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador; (A.L.-C.); (G.B.)
| | - Eduardo Tejera
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito 170125, Ecuador; (C.F.); (J.G.-C.); (E.T.)
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador
| | - Alejandro Cabrera-Andrade
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador
- Correspondence:
| |
Collapse
|
16
|
Franczyk B, Rysz J, Gluba-Brzózka A. Pharmacogenetics of Drugs Used in the Treatment of Cancers. Genes (Basel) 2022; 13:311. [PMID: 35205356 PMCID: PMC8871547 DOI: 10.3390/genes13020311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmacogenomics is based on the understanding of the individual differences in drug use, the response to drug therapy (efficacy and toxicity), and the mechanisms underlying variable drug responses. The identification of DNA variants which markedly contribute to inter-individual variations in drug responses would improve the efficacy of treatments and decrease the rate of the adverse side effects of drugs. This review focuses only on the impact of polymorphisms within drug-metabolizing enzymes on drug responses. Anticancer drugs usually have a very narrow therapeutic index; therefore, it is very important to use appropriate doses in order to achieve the maximum benefits without putting the patient at risk of life-threatening toxicities. However, the adjustment of the appropriate dose is not so easy, due to the inheritance of specific polymorphisms in the genes encoding the target proteins and drug-metabolizing enzymes. This review presents just a few examples of such polymorphisms and their impact on the response to therapy.
Collapse
Affiliation(s)
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
17
|
Li Y, Wu X, Zhong W, Tang X. Reversible Toxic Encephalopathy Involving the Cerebellum and Subcortical White Matter Attributed to Capecitabine. Am J Med Sci 2022; 363:364-370. [PMID: 35114182 DOI: 10.1016/j.amjms.2021.10.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 05/20/2021] [Accepted: 10/21/2021] [Indexed: 12/01/2022]
Abstract
Capecitabine is an anticancer drug related to 5-fluorouracil (5-FU) that is used to treat multiple cancers. Little is known about the central nervous system toxicity of capecitabine owing to the low frequency of occurrence. In this report we describe a rare case of capecitabine-related toxic encephalopathy involving the cerebellum and subcortical white matter. A review of the literature showed that most reported cases have shown excellent recovery within a few days of capecitabine termination. Whether uridine triacetate is a reasonable treatment choice for patients with life-threatening toxic encephalopathy depends on the availability of reliable clinical data. Prescreening for dihydropyrimidine dehydrogenase genotype variants and detection of 5-FU degradation rate prior to capecitabine treatment may become an effective way to avoid toxic reactions by regulating the therapeutic dose for each patient, which remains to be investigated and needs more clinical data to support.
Collapse
Affiliation(s)
- Yongchang Li
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaomei Wu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
18
|
Gmeiner WH. A narrative review of genetic factors affecting fluoropyrimidine toxicity. PRECISION CANCER MEDICINE 2021; 4:38. [PMID: 34901834 PMCID: PMC8664072 DOI: 10.21037/pcm-21-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Our objective is to document progress in developing personalized therapy with fluoropyrimidine drugs (FPs) to improve outcomes for cancer patients and to identify areas requiring further investigation. BACKGROUND FPs including 5-fluorouracil (5-FU), are among the most widely used drugs for treating colorectal cancer (CRC) and other gastrointestinal (GI) malignancies. While FPs confer a survival benefit for CRC patients, serious systemic toxicities, including neutropenia, occur in ~30% of patients with lethality in 0.5-1% of patients. While serious systemic toxicities may occur in any patient, patients with polymorphisms in DPYD, which encodes the rate-limiting enzyme for pyrimidine degradation are at very high risk. Other genetic factors affecting risk for 5-FU toxicity, including miR-27a, are under investigation. METHODS Literature used to inform the text of this article was selected from PubMed.gov from the National Library of Medicine while regulatory documents were identified via Google search. CONCLUSIONS Clinical studies to date have validated four DPYD polymorphisms (DPYD*2A, DPYD*13, c.2846A>T, HapB3) associated with serious toxicities in patients treated with 5-FU. Genetic screening for these is being implemented in the Netherlands and the UK and has been shown to be a cost-effective way to improve outcomes. Factors other than DPYD polymorphisms (e.g., miR-27a, TYMS, ENOSF1, p53) also affect 5-FU toxicity. Functional testing for deficient pyrimidine catabolism {defined as [U] >16 ng/mL or [UH2]:[U] <10} is being implemented in France and has demonstrated utility in identifying patients with elevated risk for 5-FU toxicity. Therapeutic drug monitoring (TDM) from plasma levels of 5-FU during first cycle treatment also is being used to improve outcomes and pharmacokinetic-based dosing is being used to increase the percent of patients within optimal area under the curve (AUC) (18-28 mg*h/L) values. Patients maintained in the optimal AUC range experienced significantly reduced systemic toxicities. As understanding the genetic basis for increased risk of 5-FU toxicity becomes more refined, the development of functional-based methods to optimize treatment is likely to become more widespread.
Collapse
Affiliation(s)
- William H Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
19
|
García-Alfonso P, Saiz-Rodríguez M, Mondéjar R, Salazar J, Páez D, Borobia AM, Safont MJ, García-García I, Colomer R, García-González X, Herrero MJ, López-Fernández LA, Abad-Santos F. Consensus of experts from the Spanish Pharmacogenetics and Pharmacogenomics Society and the Spanish Society of Medical Oncology for the genotyping of DPYD in cancer patients who are candidates for treatment with fluoropyrimidines. Clin Transl Oncol 2021; 24:483-494. [PMID: 34773566 PMCID: PMC8885558 DOI: 10.1007/s12094-021-02708-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/11/2021] [Indexed: 11/29/2022]
Abstract
5-Fluorouracil (5-FU) and oral fluoropyrimidines, such as capecitabine, are widely used in the treatment of cancer, especially gastrointestinal tumors and breast cancer, but their administration can produce serious and even lethal toxicity. This toxicity is often related to the partial or complete deficiency of the dihydropyrimidine dehydrogenase (DPD) enzyme, which causes a reduction in clearance and a longer half-life of 5-FU. It is advisable to determine if a DPD deficiency exists before administering these drugs by genotyping DPYD gene polymorphisms. The objective of this consensus of experts, in which representatives from the Spanish Pharmacogenetics and Pharmacogenomics Society and the Spanish Society of Medical Oncology participated, is to establish clear recommendations for the implementation of genotype and/or phenotype testing for DPD deficiency in patients who are candidates to receive fluoropyrimidines. The genotyping of DPYD previous to treatment classifies individuals as normal, intermediate, or poor metabolizers. Normal metabolizers do not require changes in the initial dose, intermediate metabolizers should start treatment with fluoropyrimidines at doses reduced to 50%, and poor metabolizers are contraindicated for fluoropyrimidines.
Collapse
Affiliation(s)
- P García-Alfonso
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Sociedad Española de Oncología Médica (SEOM), C/Doctor Esquerdo, 46, 28007, Madrid, Spain.
| | - M Saiz-Rodríguez
- Research Unit, Fundación Burgos por la Investigación de la Salud (FBIS), Hospital Universitario de Burgos, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Burgos, Spain
| | - R Mondéjar
- Medical Oncology Service, Hospital Universitario de la Princesa, Sociedad Española de Oncología Médica (SEOM), Madrid, Spain
| | - J Salazar
- Research Institute of Hospital de la Santa Creu I Sant Pau, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Barcelona, Spain
| | - D Páez
- Medical Oncology Department, Hospital de la Santa Creu I Sant Pau, Sociedad Española de Oncología Médica (SEOM), Barcelona, España
| | - A M Borobia
- Clinical Pharmacology Service, Hospital Universitario La Paz, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Madrid, Spain
| | - M J Safont
- Medical Oncology Service, Consorcio Hospital General Universitario de Valencia, Universidad de Valencia, CIBERONC, Sociedad Española de Oncología Médica (SEOM), Valencia, Spain
| | - I García-García
- Clinical Pharmacology Service, Hospital Universitario La Paz, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Madrid, Spain
| | - R Colomer
- Medical Oncology Service, Hospital Universitario de La Princesa y Cátedra de Medicina Personalizada de Precisión de la Universidad Autónoma de Madrid (UAM), Sociedad Española de Oncología Médica (SEOM), Madrid, Spain
| | - X García-González
- Hospital Pharmacy Service, Hospital General Universitario Gregorio Marañón, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Madrid, Spain
| | - M J Herrero
- Pharmacogenetics Platform, IIS La Fe-Hospital La Fe and Pharmacology Department, Universidad de Valencia, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Valencia, Spain
| | - L A López-Fernández
- Hospital Pharmacy Service, Hospital General Universitario Gregorio Marañón, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), Madrid, Spain
| | - F Abad-Santos
- Clinical Pharmacology Service, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Sociedad Española de Farmacogenética y Farmacogenómica (SEFF), C/Diego de León, 62., 28006, Madrid, Spain.
| |
Collapse
|
20
|
Sharma BB, Rai K, Blunt H, Zhao W, Tosteson TD, Brooks GA. Pathogenic DPYD Variants and Treatment-Related Mortality in Patients Receiving Fluoropyrimidine Chemotherapy: A Systematic Review and Meta-Analysis. Oncologist 2021; 26:1008-1016. [PMID: 34506675 DOI: 10.1002/onco.13967] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pathogenic variants of the DPYD gene are strongly associated with grade ≥3 toxicity during fluoropyrimidine chemotherapy. We conducted a systematic review and meta-analysis to estimate the risk of treatment-related death associated with DPYD gene variants. MATERIALS AND METHODS We searched for reports published prior to September 17, 2020, that described patients receiving standard-dose fluoropyrimidine chemotherapy (5-fluorouracil or capecitabine) who had baseline testing for at least one of four pathogenic DPYD variants (c.1129-5923C>G [HapB3], c.1679T>G [*13], c.1905+1G>A [*2A], and c.2846A>T) and were assessed for toxicity. Two reviewers assessed studies for inclusion and extracted study-level data. The primary outcome was the relative risk of treatment-related mortality for DPYD variant carriers versus noncarriers; we performed data synthesis using a Mantel-Haenszel fixed effects model. RESULTS Of the 2,923 references screened, 35 studies involving 13,929 patients were included. DPYD variants (heterozygous or homozygous) were identified in 566 patients (4.1%). There were 14 treatment-related deaths in 13,363 patients without identified DPYD variants (treatment-related mortality, 0.1%; 95% confidence interval [CI], 0.1-0.2) and 13 treatment-related deaths in 566 patients with any of the four DPYD variants (treatment-related mortality, 2.3%; 95% CI, 1.3%-3.9%). Carriers of pathogenic DPYD gene variants had a 25.6 times increased risk of treatment-related death (95% CI, 12.1-53.9; p < .001). After excluding carriers of the more common but less deleterious c.1129-5923C>G variant, carriers of c.1679T>G, c.1905+1G>A, and/or c.2846A>T had treatment-related mortality of 3.7%. CONCLUSION Patients with pathogenic DPYD gene variants who receive standard-dose fluoropyrimidine chemotherapy have greatly increased risk for treatment-related death. IMPLICATIONS FOR PRACTICE The syndrome of dihydropyrimidine dehydrogenase (DPD) deficiency is an uncommon but well-described cause of severe toxicity related to fluoropyrimidine chemotherapy agents (5-fluorouracil and capecitabine). Patients with latent DPD deficiency can be identified preemptively with genotyping of the DPYD gene, or with measurement of the plasma uracil concentration. In this systematic review and meta-analysis, the authors study the rare outcome of treatment-related death after fluoropyrimidine chemotherapy. DPYD gene variants associated with DPD deficiency were linked to a 25.6 times increased risk of fluoropyrimidine-related mortality. These findings support the clinical utility of DPYD genotyping as a screening test for DPD deficiency.
Collapse
Affiliation(s)
| | - Karan Rai
- Geisel School of Medicine at Dartmouth, Lebanon, New Hamphsire, USA
| | - Heather Blunt
- Biomedical Libraries, Dartmouth College, Hanover, New Hampshire, USA
| | - Wenyan Zhao
- Department of Biomedical Data Science, Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Tor D Tosteson
- Department of Biomedical Data Science, Geisel School of Medicine, Lebanon, New Hampshire, USA.,The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Gabriel A Brooks
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA.,The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Lebanon, New Hampshire, USA
| |
Collapse
|
21
|
Deligonul A, Aksoy S, Tezcan G, Tunca B, Kanat O, Cubukcu E, Yilmazlar T, Ozturk E, Egeli U, Cecener G, Alemdar A, Evrensel T. DPYD c.1905 + 1G>A Promotes Fluoropyrimidine-Induced Anemia, a Prognostic Factor in Disease-Free Survival, in Colorectal Cancer. Genet Test Mol Biomarkers 2021; 25:276-283. [PMID: 33877893 DOI: 10.1089/gtmb.2020.0285] [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: 11/12/2022] Open
Abstract
Background and Aim: In 10-30% of colorectal cancer (CRC) patients, toxic reactions occur after fluoropyrimidine-based chemotherapy. A dihydropyridine dehydrogenase (DPYD) gene variant, c.1905 + 1G>A, leads to intolerance to fluoropyrimidines. Due to the low frequency of this variant in many populations, the prevalence of fluoropyrimidine-induced hematologic side effects in CRC patients with the c.1905 + 1G>A variant is unclear. In this study, we investigated the prevalence of the DPYD c.1905 + 1 variants in a Turkish CRC cohort and the potential effects of these variants on fluoropyrimidine-induced hematologic side effects. Materials and Methods: The DPYD c.1905 + 1 variant was genotyped using polymerase chain reaction-restriction fragment length polymorphism analysis and confirmed by Sanger sequencing in peripheral blood samples of 100 CRC patients who received fluoropyrimidine-based chemotherapy and 60 healthy volunteers. The association of c.1905 + 1 variants with susceptibility to hematologic side effects was evaluated. Results: The DPYD c.1905 + 1G>A variant was more common in the CRC group than in the healthy control group (p = 0.001). The presence of the c.1905 + 1G>A variant was associated with thrombocytopenia (p = 0.039) and anemia (p = 0.035). CRC patients with fluoropyrimidine-induced anemia had shorter disease-free survival than CRC patients without fluoropyrimidine-induced anemia (p = 0.0009). Conclusions: Before administering fluoropyrimidine-based chemotherapy, genetic screening for the DPYD c.1905 + 1G>A variant should be performed with the aim of preventing anemia and anemia-induced complications in CRC patients.
Collapse
Affiliation(s)
- Adem Deligonul
- Department of Medical Oncology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Secil Aksoy
- First and Emergency Aid Program, Vocational School of Inegol, Bursa Uludag University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - Berrin Tunca
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozkan Kanat
- Department of Medical Oncology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Erdem Cubukcu
- Department of Medical Oncology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Tuncay Yilmazlar
- Department of General Surgery, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ersin Ozturk
- Department of General Surgery, Faculty of Medicine, Karatay University, Konya, Turkey.,Department of General Surgery, Medicana Hospital Bursa, Bursa, Turkey
| | - Unal Egeli
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Gulsah Cecener
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Adem Alemdar
- Institution of Health Sciences, Bursa Uludag University, Bursa, Turkey
| | - Turkkan Evrensel
- Department of Medical Oncology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
22
|
Furukawa T, Sasaki T, Ono Y, Kawano F, Takamatsu M, Mori C, Mie T, Yamada Y, Okamoto T, Takeda T, Kasuga A, Matsuyama M, Ozaka M, Takahashi Y, Sasahira N. A case of necrotic enteritis during neoadjuvant chemotherapy with gemcitabine and S-1 for resectable pancreatic ductal adenocarcinoma. Clin J Gastroenterol 2021; 14:1571-1577. [PMID: 34255287 DOI: 10.1007/s12328-021-01480-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/09/2021] [Indexed: 11/25/2022]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death in Japan. Pancreatic cancer is categorized as resectable, borderline resectable, or unresectable based on the degree of adjacent vascular invasion and the presence of distant metastases. Neoadjuvant chemotherapy with gemcitabine and S-1 (NAC-GS) has recently become a standard option for resectable pancreatic cancer in Japanese patients. According to previous reports, GS is considered to be relatively safe and feasible treatment for Japanese patients, including the elderly. However, NAC-GS is occasionally associated with severe adverse events which may ultimately render the patient unfit for surgery. A 60-year-old man with resectable pancreatic cancer suffered from severe necrotic enteritis during NAC-GS, which required surgical resection. Considering the time course and histological findings of the resected bowel, S-1 was believed to be the causative agent. The low urinary dihydrouracil to uracil ratio also suggested possible dihydropyrimidine dehydrogenase deficiency, which may have hindered the metabolism of S-1 and contributed to the development of necrotic enteritis. Life-threatening enteritis occurs in approximately 0.3% of all patients who receive S-1. As initial symptoms are non-specific, patients should be instructed to lower the hurdle for contacting the hospital during NAC-GS.
Collapse
Affiliation(s)
- Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan.
| | - Yoshihiro Ono
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Fumihiro Kawano
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Chinatsu Mori
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takafumi Mie
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Yuto Yamada
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takeshi Okamoto
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Akiyoshi Kasuga
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Masato Matsuyama
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Yu Takahashi
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| |
Collapse
|
23
|
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
|
24
|
da Rocha JEB, Lombard Z, Ramsay M. Potential Impact of DPYD Variation on Fluoropyrimidine Drug Response in sub-Saharan African Populations. Front Genet 2021; 12:626954. [PMID: 33767731 PMCID: PMC7985174 DOI: 10.3389/fgene.2021.626954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer is a critical health burden in Africa, and mortality rates are rising rapidly. Treatments are expensive and often cause adverse drug reactions (ADRs). Fluoropyrimidine treatments can lead to severe toxicity events which have been linked to variants within the dihydropyrimidine dehydrogenase (DPYD) gene. There are clinical guidelines to improve safety outcomes of treatment, but these are primarily based on variants assessed in non-African populations. Whole genome sequencing data from the 1000 Genomes Project and the African Genome Variation Project were mined to assess variation in DPYD in eight sub-Saharan African populations. Variant functional annotation was performed with a series of bioinformatics tools to assess potential likelihood of deleterious impact. There were 29 DPYD coding variants identified in the datasets assessed, of which 25 are rare, and some of which are known to be deleterious. One African-specific variant (rs115232898-C), is common in sub-Saharan Africans (1-4%) and known to reduce the function of the dihydropyrimidine dehydrogenase enzyme (DPD), having been linked to cases of severe toxicity. This variant, once validated in clinical trials, should be considered for inclusion in clinical guidelines for use in sub-Saharan African populations. The rs2297595-C variant is less well-characterized in terms of effect, but shows significant allele frequency differences between sub-Saharan African populations (0.5-11.5%; p = 1.5 × 10-4), and is more common in East African populations. This study highlights the relevance of African-data informed guidelines for fluorouracil drug safety in sub-Saharan Africans, and the need for region-specific data to ensure that Africans may benefit optimally from a precision medicine approach.
Collapse
Affiliation(s)
- Jorge E B da Rocha
- Faculty of Health Sciences, Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa.,Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Zané Lombard
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Faculty of Health Sciences, Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa.,Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
25
|
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Farkas L, Garrido-Laguna I, Grem JL, Gunn A, Hecht JR, Hoffe S, Hubbard J, Hunt S, Johung KL, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Gregory KM, Gurski LA. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:329-359. [PMID: 33724754 DOI: 10.6004/jnccn.2021.0012] [Citation(s) in RCA: 765] [Impact Index Per Article: 255.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Colon Cancer focuses on systemic therapy options for the treatment of metastatic colorectal cancer (mCRC), because important updates have recently been made to this section. These updates include recommendations for first-line use of checkpoint inhibitors for mCRC, that is deficient mismatch repair/microsatellite instability-high, recommendations related to the use of biosimilars, and expanded recommendations for biomarker testing. The systemic therapy recommendations now include targeted therapy options for patients with mCRC that is HER2-amplified, or BRAF V600E mutation-positive. Treatment and management of nonmetastatic or resectable/ablatable metastatic disease are discussed in the complete version of the NCCN Guidelines for Colon Cancer available at NCCN.org. Additional topics covered in the complete version include risk assessment, staging, pathology, posttreatment surveillance, and survivorship.
Collapse
Affiliation(s)
- Al B Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Alan P Venook
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | | | | | - Stacey Cohen
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - Linda Farkas
- UT Southwestern Simmons Comprehensive Cancer Center
| | | | | | | | | | | | | | - Steven Hunt
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Smitha Krishnamurthi
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | - Eric D Miller
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mary F Mulcahy
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Katrina Pedersen
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
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
|
27
|
Jolivet C, Nassabein R, Soulières D, Weng X, Amireault C, Ayoub JP, Beauregard P, Blais N, Carrier C, Cloutier AS, Desnoyers A, Lemay AS, Lemay F, Loungnarath R, Jolivet J, Letendre F, Tehfé M, Vadnais C, Viens D, Aubin F. Implementing DPYD*2A Genotyping in Clinical Practice: The Quebec, Canada, Experience. Oncologist 2020; 26:e597-e602. [PMID: 33274825 DOI: 10.1002/onco.13626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fluoropyrimidines are used in chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. DPYD*2A polymorphism is one of the most studied variants. The study objective was to document the impact of implementing this test in routine clinical practice. METHODS We retrospectively performed chart reviews of all patients who tested positive for a heterozygous or homozygous DPYD*2A mutation in samples obtained from patients throughout the province of Quebec, Canada. RESULTS During a period of 17 months, 2,617 patients were tested: 25 patients tested positive. All were White. Twenty-four of the 25 patients were heterozygous (0.92%), and one was homozygous (0.038%). Data were available for 20 patients: 15 were tested upfront, whereas five were identified after severe toxicities. Of the five patients confirmed after toxicities, all had grade 4 cytopenias, 80% grade ≥3 mucositis, 20% grade 3 rash, and 20% grade 3 diarrhea. Eight patients identified with DPYD*2A mutation prior to treatment received fluoropyrimidine-based chemotherapy at reduced initial doses. The average fluoropyrimidine dose intensity during chemotherapy was 50%. No grade ≥3 toxicities were observed. DPYD*2A test results were available in an average of 6 days, causing no significant delays in treatment initiation. CONCLUSION Upfront genotyping before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. The administration of chemotherapy at reduced doses appears to be safe in patients heterozygous for DPYD*2A. IMPLICATIONS FOR PRACTICE Fluoropyrimidines are part of chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. This retrospective analysis demonstrates that upfront genotyping of DPYD before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. This approach was reported previously, but insufficient data concerning its application in real practice are available. This is likely the first reported experience of systematic DPYD genotyping all over Canada and North America as well.
Collapse
Affiliation(s)
- Catherine Jolivet
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Rami Nassabein
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Denis Soulières
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Xiaoduan Weng
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | | | - Jean-Pierre Ayoub
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Patrice Beauregard
- Centre Hospitalier de l'Université Sherbrooke, Sherbrooke, Quebec, Canada
| | - Normand Blais
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Christian Carrier
- Centre Hospitalier Régional Trois-Rivières, Trois-Rivières, Quebec, Canada
| | | | | | - Anne-Sophie Lemay
- Centre Hospitalier Régional Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Frédéric Lemay
- Centre Hospitalier de l'Université Sherbrooke, Sherbrooke, Quebec, Canada
| | - Rasmy Loungnarath
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Jacques Jolivet
- Centre Intégré de Santé et de Services Sociaux (CISSS) des Laurentides, Saint-Jérôme, Quebec, Canada
| | | | - Mustapha Tehfé
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Charles Vadnais
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Daniel Viens
- Hôpital Sainte-Croix, Drummondville, Quebec, Canada
| | - Francine Aubin
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| |
Collapse
|
28
|
Simões AR, Fernández-Rozadilla C, Maroñas O, Carracedo Á. The Road so Far in Colorectal Cancer Pharmacogenomics: Are We Closer to Individualised Treatment? J Pers Med 2020; 10:E237. [PMID: 33228198 PMCID: PMC7711884 DOI: 10.3390/jpm10040237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
In recent decades, survival rates in colorectal cancer have improved greatly due to pharmacological treatment. However, many patients end up developing adverse drug reactions that can be severe or even life threatening, and that affect their quality of life. These remain a limitation, as they may force dose reduction or treatment discontinuation, diminishing treatment efficacy. From candidate gene approaches to genome-wide analysis, pharmacogenomic knowledge has advanced greatly, yet there is still huge and unexploited potential in the use of novel technologies such as next-generation sequencing strategies. This review summarises the road of colorectal cancer pharmacogenomics so far, presents considerations and directions to be taken for further works and discusses the path towards implementation into clinical practice.
Collapse
Affiliation(s)
- Ana Rita Simões
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Ceres Fernández-Rozadilla
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Olalla Maroñas
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica; SERGAS, 15706 Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Raras—CIBERER, 28029 Madrid, Spain
| |
Collapse
|
29
|
Zhou Y, Dagli Hernandez C, Lauschke VM. Population-scale predictions of DPD and TPMT phenotypes using a quantitative pharmacogene-specific ensemble classifier. Br J Cancer 2020; 123:1782-1789. [PMID: 32973300 PMCID: PMC7722893 DOI: 10.1038/s41416-020-01084-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Inter-individual differences in dihydropyrimidine dehydrogenase (DPYD encoding DPD) and thiopurine S-methyltransferase (TPMT) activity are important predictors for fluoropyrimidine and thiopurine toxicity. While several variants in these genes are known to decrease enzyme activities, many additional genetic variations with unclear functional consequences have been identified, complicating informed clinical decision-making in the respective carriers. METHODS We used a novel pharmacogenetically trained ensemble classifier to analyse DPYD and TPMT genetic variability based on sequencing data from 138,842 individuals across eight populations. RESULTS The algorithm accurately predicted in vivo consequences of DPYD and TPMT variants (accuracy 91.4% compared to 95.3% in vitro). Further analysis showed high genetic complexity of DPD deficiency, advocating for sequencing-based DPYD profiling, whereas genotyping of four variants in TPMT was sufficient to explain >95% of phenotypic TPMT variability. Lastly, we provided population-scale profiles of ethnogeographic variability in DPD and TPMT phenotypes, and revealed striking interethnic differences in frequency and genetic constitution of DPD and TPMT deficiency. CONCLUSION These results provide the most comprehensive data set of DPYD and TPMT variability published to date with important implications for population-adjusted genetic profiling strategies of fluoropyrimidine and thiopurine risk factors and precision public health.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Carolina Dagli Hernandez
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden.,Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, 05508-000, Sao Paulo, Brazil
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden.
| |
Collapse
|
30
|
Capitain O, Seegers V, Metges JP, Faroux R, Stampfli C, Ferec M, Budnik TM, Senellart H, Rossi V, Blouin N, Dauvé J, Campone M. Comparison of 4 Screening Methods for Detecting Fluoropyrimidine Toxicity Risk: Identification of the Most Effective, Cost-Efficient Method to Save Lives. Dose Response 2020; 18:1559325820951367. [PMID: 32973417 PMCID: PMC7493257 DOI: 10.1177/1559325820951367] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Fluoropyrimidines (FPs) carry around 20% risk of G3-5 toxicity and 0.2-1% risk of death, due to dihydropyrimidine dehydrogenase (DPD) deficiency. Several screening approaches exist for predicting toxicity, however there is ongoing debate over which method is best. This study compares 4 screening approaches. Method 472 patients treated for colorectal, head-and-neck, breast, or pancreatic cancers, who had not been tested pre-treatment for FP toxicity risk, were screened using: DPYD genotyping (G); phenotyping via plasma Uracil (U); phenotyping via plasma-dihydrouracil/uracil ratio (UH2/U); and a Multi-Parametric Method (MPM) using genotype, phenotype, and epigenetic data. Performance was compared, particularly the inability to detect at-risk patients (false negatives). Results False negative rates for detecting G5 toxicity risk were 51.2%, 19.5%, 9.8% and 2.4%, for G, U, UH2/U and MPM, respectively. False negative rates for detecting G4-5 toxicity risk were 59.8%, 36.1%, 21.3% and 4.7%, respectively. MPM demonstrated significantly (p < 0.001) better prediction performance. Conclusion MPM is the most effective method for limiting G4-5 toxicity. Its systematic implementation is cost-effective and significantly improves the risk-benefit ratio of FP-treatment. The use of MPM, rather than G or U testing, would avoid nearly 8,000 FP-related deaths per year globally (500 in France), and spare hundreds of thousands from G4 toxicity.
Collapse
Affiliation(s)
| | | | | | - Roger Faroux
- CH Départemental Vendée La Roche sur Yon, France
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Duran G, Cruz R, Simoes AR, Barros F, Giráldez JM, Bernárdez B, Anido U, Candamio S, López-López R, Carracedo Á, Lamas MJ. Efficacy and toxicity of adjuvant chemotherapy on colorectal cancer patients: how much influence from the genetics? J Chemother 2020; 32:310-322. [PMID: 32441565 DOI: 10.1080/1120009x.2020.1764281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We studied the predictive value for response and toxicity of functional polymorphisms in genes involved in the oxaliplatin/fluorouracil pathway in colorectal cancer patients. One hundred and twenty-seven (127) patients were treated with curative intended surgery followed by adjuvant chemotherapy with FOLFOX (fluorouracil, leucovorin and oxaliplatin) regimen. The median age was 65.53 (27-80) years (66.9% male, 59.1% rectum). The median follow-up was 8.5 years (IQR, 4.1-9.4). At the end of follow-up, 59 patients (46.5%) had relapsed or died in the whole study population. We did find that XRCC1GG genotype is associated with a higher risk of developing haematologic toxicity. Furthermore, we report a significant association of the TS 3'UTR 6 bp/6 bp polymorphism and the XRCC1 rs25487 with a higher risk of developing anaemia and diarrhoea, respectively. On the other hand, none of the studied polymorphisms showed clinically relevant association with disease-free survival and overall survival or early failure to adjuvant FOLFOX therapy.
Collapse
Affiliation(s)
- Goretti Duran
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Raquel Cruz
- Center for Biomedical Research on Rare Diseases (CIBERER), Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Rita Simoes
- Fundación Instituto de Investigación Sanitaria de Santiago (FIDIS), Santiago de Compostela, Spain.,Departamento de Ciencias Forenses, Anatomía Patolóxica, Xinecoloxía, Obstetricia e Pediatría, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain.,Genomics Medicine Group, Galician Public Foundation of Genomic Medicine (FPGMX), Santiago de Compostela, Spain
| | - Francisco Barros
- Genomics Medicine Group, Galician Public Foundation of Genomic Medicine (FPGMX), Santiago de Compostela, Spain
| | - José María Giráldez
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Beatriz Bernárdez
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Urbano Anido
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Sonia Candamio
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Rafael López-López
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain.,Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Ángel Carracedo
- Genomics Medicine Group, CIBERER, Galician Public Foundation of Genomic Medicine (FPGMX), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Jesús Lamas
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
32
|
Ferrari D, Ghi MG, Franzese C, Codecà C, Gau M, Fayette J. The Slippery Role of Induction Chemotherapy in Head and Neck Cancer: Myth and Reality. Front Oncol 2020; 10:7. [PMID: 32038985 PMCID: PMC6989487 DOI: 10.3389/fonc.2020.00007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Chemoradiotherapy as an alternative to surgery can be offered to patients affected by loco-regionally advanced head and neck cancer (HNC). Induction chemotherapy is a valid option, supported by few positive trials, but its real efficacy is still a matter of debate. The standard regimen for induction chemotherapy in Europe is a combination of docetaxel (75 mg/m2) and reduced dose doses of cisplatin (75 mg/m2) and 5-fluorouracil (750 mg/m2 day, for five consecutive days) (TPF). It is less toxic and more effective than the historical therapy PF (cisplatin 100 mg/m2 and fluorouracil 1,000 mg/m2/day for five consecutive days). However, in some studies treatment-related mortality has been reported to be as high as 6%. Therefore, some less toxic combinations, such as a modified TPF regimen and the combination of carboplatin plus paclitaxel have been studied. These regimens are showing promising results but deserve further validation in comparative trials. Furthermore, several trials are underway in order to enhance TPF with immune checkpoints inhibitors. Compared to chemoradiotherapy, induction chemotherapy followed by chemoradiation was shown to be non-inferior, and it could decrease the distant metastatic progression, especially in high-risk populations. For selected patients, induction chemotherapy could be a strong option. The chemoselective process that leads to immediate surgery for non-responders, the high response rate (complete responses are sometimes observed), and the survival data, are all arguments in favor of induction chemotherapy, if performed in experienced centers involving health professionals in the context of a skilled multidisciplinary team.
Collapse
Affiliation(s)
- Daris Ferrari
- Department of Oncology, San Paolo Hospital, Milan, Italy
| | | | - Ciro Franzese
- Department of Radiotherapy and Radiosurgery, Humanitas Cancer Center, Rozzano, Italy
| | - Carla Codecà
- Department of Oncology, San Paolo Hospital, Milan, Italy
| | - Max Gau
- Rhône-Alpes, Centre de Lutte Contre le Cancer Léon Bérard, Lyon, France
| | - Jerome Fayette
- Rhône-Alpes, Centre de Lutte Contre le Cancer Léon Bérard, Lyon, France
| |
Collapse
|
33
|
Barin-Le Guellec C, Lafay-Chebassier C, Ingrand I, Tournamille JF, Boudet A, Lanoue MC, Defossez G, Ingrand P, Perault-Pochat MC, Etienne-Grimaldi MC. Toxicities associated with chemotherapy regimens containing a fluoropyrimidine: A real-life evaluation in France. Eur J Cancer 2020; 124:37-46. [PMID: 31715555 DOI: 10.1016/j.ejca.2019.09.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
AIMS Despite fluoropyrimidines (FPs) constituting the main component of the chemotherapy combination protocols in 50% of chemotherapies for solid tumour treatments, incidence data for FP-related toxicity are poorly documented in real life. This study evaluated the number of patients receiving FP-based chemotherapies in France, along with the true incidence of FP-related serious adverse effects (SAEs) before the recent mandatory dihydropyrimidine dehydrogenase (DPD)-screening was introduced by French health authorities, DPD being the rate-limiting enzyme of 5-fluorouracil (5-FU) catabolism. METHODS Exhaustive data on the number of patients treated with FP-based chemotherapy in 2013-2014 were collected in the Centre-Val de Loire region of France. True incidence of SAEs was extracted from a cohort of 513 patients with incident solid tumours receiving first-line FP-based chemotherapy. RESULTS After extrapolation at national level, we estimated that 76,200 patients are currently treated annually with 5FU (53,100 patients, 62% digestive system-related versus 26% breast cancers versus 12% head and neck cancers) or capecitabine (23,100 patients, 45% digestive system-related versus 37% breast cancers versus 18% non-documented). Earlier (in the first two cycles) the SAE incidence rate was 19.3% (95% confidence interval (CI) 16-23%) including one toxic death (0.2%, 95%CI 0-1%). SAE incidence rate was 32.2% (95%CI 28-36%) over the first 6 months of treatment. Incidence of death, life-threatening prognosis or incapacity/disability was 1.4% (95%CI 0.4-2.4%) and 1.6% (95%CI 0.5-2.6%) during first two cycles and first 6 months, respectively. CONCLUSION These data highlight the significant public health issue related to FP toxicity, with around 1200 patients developing FP-related life-threatening prognosis or incapacity/disability annually in France, including 150 toxic deaths. It is hoped that DPD-deficiency screening will reduce such iatrogenic events and eradicate toxic deaths.
Collapse
Affiliation(s)
- Chantal Barin-Le Guellec
- Laboratoire de Biochimie et Biologie Moléculaire, Unité de Pharmacogénétique, CHU de Tours, France; INSERM U1248-IPPRIT, Université, CHU de Limoges, France.
| | - Claire Lafay-Chebassier
- Service de Pharmacologie Clinique et Vigilances, Université, CHU de Poitiers, France; INSERM U1084-LNEC/ INSERM CIC 1402, Université, CHU de Poitiers, France
| | - Isabelle Ingrand
- Service de Pharmacologie Clinique et Vigilances, Université, CHU de Poitiers, France; Unité d'Epidémiologie et Biostatistique, Registre Général des Cancers Poitou-Charentes, INSERM CIC 1402, Université, CHU de Poitiers, France
| | | | | | | | - Gautier Defossez
- Unité d'Epidémiologie et Biostatistique, Registre Général des Cancers Poitou-Charentes, INSERM CIC 1402, Université, CHU de Poitiers, France
| | - Pierre Ingrand
- Unité d'Epidémiologie et Biostatistique, Registre Général des Cancers Poitou-Charentes, INSERM CIC 1402, Université, CHU de Poitiers, France
| | - Marie-Christine Perault-Pochat
- Service de Pharmacologie Clinique et Vigilances, Université, CHU de Poitiers, France; INSERM U1084-LNEC/ INSERM CIC 1402, Université, CHU de Poitiers, France
| | | |
Collapse
|
34
|
Merloni F, Ranallo N, Scortichini L, Giampieri R, Berardi R. Tailored therapy in patients treated with fluoropyrimidines: focus on the role of dihydropyrimidine dehydrogenase. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:787-802. [PMID: 35582578 PMCID: PMC8992529 DOI: 10.20517/cdr.2018.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/01/2019] [Accepted: 06/04/2019] [Indexed: 01/19/2023]
Abstract
Fluoropyrimidines are widely used in the treatment of solid tumors, mainly gastrointestinal, head and neck and breast cancer. Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme for catabolism of 5-FU and it is encoded by DPYD gene. To date, many known polymorphisms cause DPD deficiency and subsequent increase of 5-FU toxicity. In addition, reduced inactivation of 5-FU could lead to increased 5-FU intracellular concentration and augmented efficacy of this drugs. Therefore DPD expression, particularly intratumoral, has been investigated as predictive and prognostic marker in 5-FU treated patients. There also seems to be a tendency to support the correlation between DPD expression and response/survival in patients treated with fluoropyrimidine even if definitive conclusions cannot be drawn considering that some studies are conflicting. Therefore, the debate on intratumoral DPD expression as a potential predictor and prognostic marker in patients treated with fluoropyrimidines is still open. Four DPD-polymorphisms are the most relevant for their frequency in population and clinical relevance. Many studies demonstrate that treating a carrier of one of these polymorphisms with a full dose of fluoropyrimidine can expose patient to a severe, even life-threatening, toxicity. Severe toxicity is reduced if this kind of patients received a dose-adjustment after being genotyped. CPIC (Clinical Pharmacogenetics Implementation Consortium) is an International Consortium creating guidelines for facilitating use of pharmacogenetic tests for patient care and helps clinicians ensuring a safer drug delivery to the patient. Using predictive DPD deficiency tests in patients receiving 5FU-based chemotherapy, in particular for colorectal cancer, has proven to be a cost-effective strategy.
Collapse
Affiliation(s)
- Filippo Merloni
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Nicoletta Ranallo
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Laura Scortichini
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Riccardo Giampieri
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona 60126, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona 60126, Italy
| |
Collapse
|
35
|
The Prevalence of DPYD*9A(c.85T>C) Genotype and the Genotype-Phenotype Correlation in Patients with Gastrointestinal Malignancies Treated With Fluoropyrimidines: Updated Analysis. Clin Colorectal Cancer 2019; 18:e280-e286. [PMID: 31160238 DOI: 10.1016/j.clcc.2019.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/08/2019] [Accepted: 04/26/2019] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The dihydropyrimidine dehydrogenase gene (DPYD)*9A (c.85T>C) genotype is relatively common. The correlation between DPYD*9A genotype and dihydropyrimidine dehydrogenase (DPD) deficiency phenotype is controversial. In a cohort of 28 patients, DPYD*9A was the most commonly diagnosed variant (13 patients [46%]) and there was a noticeable genotype-phenotype correlation. In this study we genotyped a larger cohort of a mixed racial background to explore the prevalence of DPYD*9A variant and to confirm the genotype-phenotype correlation. PATIENTS AND METHODS Between 2011 and 2018, in addition to genotyping for high-risk DPYD variants (DPYD*2A, DPYD*13 and DPYD*9B), genotyping for DPYD*9A variant was performed on 113 patients with gastrointestinal malignancies treated with fluoropyrimidines. Fluoropyrimidines-associated toxicity was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (version 5.0). Fisher exact test was used for statistical analysis. RESULTS Heterozygous and homozygous DPYD*9A genotypes were identified in 46 (41%) and 11 (10%) patients, respectively. Among patients with DPYD*9A genotypes (n = 57), men and women represented 30 (53%) and 27 (47%) patients, respectively. Caucasian, African American, and other ethnicities represented 29 (50.9%), 26 (45.6%), and 2 (3.5%) patients, respectively. Grade 3/4 toxicities were experienced in 26 patients with DPYD*9A genotype (3 patients had homozygous status) and in 20 patients with wild type DPYD*9A (P = .4405). In patients who received full-dose fluoropyrimidines (n = 85), Grade 3/4 toxicities were experienced in 22 patients with DPYD*9A genotype (2 patients had homozygous status), and in 17 patients with wild type DPYD (P = .8275). CONCLUSION In our updated analysis, the prevalence of heterozygous and homozygous DPYD*9A genotypes were 41% and 10%, respectively. The correlation between DPYD*9A genotype and DPD clinical phenotype was not reproduced. The noticeable correlation that we previously reported is likely because of small sample size and selection bias.
Collapse
|
36
|
De Falco V, Natalicchio MI, Napolitano S, Coppola N, Conzo G, Martinelli E, Zanaletti N, Vitale P, Giunta EF, Vietri MT, Vitiello PP, Ciardiello D, Marinaccio A, De Vita F, Ciardiello F, Troiani T. A case report of a severe fluoropyrimidine-related toxicity due to an uncommon DPYD variant. Medicine (Baltimore) 2019; 98:e15759. [PMID: 31124962 PMCID: PMC6571425 DOI: 10.1097/md.0000000000015759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Fluoropyrimidines such as 5-fluorouracil (5-FU) and its orally active prodrug, capecitabine, are widely used in the treatment of gastrointestinal cancer, including colorectal cancer. Dihydropyrimidine dehydrogenase (DPD) plays an important role in the 5-FU metabolism. Dihydropyrimidine dehydrogenase gene (DPYD) is a highly polymorphic gene with several hundreds of reported genetic variants and DPD activity levels vary considerably among individuals, with different 5-FU-related efficacy and toxicity. About 5% of the population is deficient in DPD enzyme activity. The most well studied DPYD variant is the IVS14+1G>A, also known as DPYD *2A. In this report, we present a case of a patient with a double heterozygote DPYD variant (DPYD activity score: 0,5 according to Clinical Pharmacogenetics Implementation Consortium) who experienced a severe fluoropyrimidine-related toxicity resolved without any consequence. PATIENT CONCERNS A 46-years-old Caucasian man with diagnosis of left colon adenocarcinoma underwent left hemicolectomy on July 2017: pT3 G3 N1c M0. According to the disease stage, he started an adjuvant therapy with XELOX using capecitabine at 50% of total dose, because of his DPYD IVS14+1G>A variant, detected before the treatment. DIAGNOSIS After few days, despite of this dose reduction, he experienced life-threatening adverse events such as mucositis G3, diarrhea G3, neutropenia G4, thrombocytopenia G4, and hyperbilirubinemia G3 according to Common Terminology Criteria for Adverse Events v 5.0. INTERVENTIONS As first, we set up an intensive rehydration therapy, antibiotic and antifungal prophylaxis, Granulocyte-Colony Stimulating Factors, and supportive blood transfusions. Additional genetic tests revealed a double heterozygote variant of DPYD gene (DPYD IVS14+1G>A and 2846A>T) which is a very rare situation and only 3 cases are described in literature, all of them concluded with patient's death. OUTCOMES After 3 weeks of intensive therapy, the patient was fully recovered. Furthermore, all the whole-body CT scans performed since discharge from the hospital until now, have confirmed no evidence of disease. CONCLUSIONS Recent studies demonstrated that screening strategy for the most common DPYD variants allowed for avoiding toxicities and saving money. This report underlines the importance of genotyping DPYD before treatment and emphasizes the role of genotype-guided dose individualization.
Collapse
Affiliation(s)
- Vincenzo De Falco
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | | | - Stefania Napolitano
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicola Coppola
- Malattie Infettive, Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania Luigi Vanvitelli
| | - Giovanni Conzo
- UOC di Chirurgia Generale ed Oncologica, Università degli Studi della Campania Luigi Vanvitelli
| | - Erika Martinelli
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Nicoletta Zanaletti
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Pasquale Vitale
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Emilio Francesco Giunta
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Maria Teresa Vietri
- UOC Patologia clinica e molecolare, Dipartimento di Medicina di Precisione, DAI dei Sevizi di laboratorio e Sanità pubblica, Università degli Studi della Campania Luigi Vanvitelli, Complesso di S. Andrea delle Dame, Napoli, Italia
| | - Pietro Paolo Vitiello
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Davide Ciardiello
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Anna Marinaccio
- Laboratorio di Biologia Molecolare, Ospedale Riuniti, Foggia, Italia
| | - Ferdinando De Vita
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Fortunato Ciardiello
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| | - Teresa Troiani
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Napoli
| |
Collapse
|
37
|
Soveri LM, Lamminmäki A, Hänninen UA, Karhunen M, Bono P, Osterlund P. Long-term neuropathy and quality of life in colorectal cancer patients treated with oxaliplatin containing adjuvant chemotherapy. Acta Oncol 2019; 58:398-406. [PMID: 30638100 DOI: 10.1080/0284186x.2018.1556804] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Oxaliplatin, combined with capecitabine (CAPOX) or infused 5-fluorouracil (FOLFOX), is standard of care in the adjuvant treatment of colorectal cancer (CRC). Prospective data on prevalence of oxaliplatin induced acute and long-term neuropathy in a real-life patient population and its effects on quality of life (QOL) and survival is limited, and scarce in CAPOX versus FOLFOX treated, especially in a subarctic climate. METHODS One hundred forty-four adjuvant CRC patients (all 72 CAPOX cases and 72 matched FOLFOX controls) were analyzed regarding oxaliplatin induced sensory neuropathy, which was graded according to NCI-CTCAEv3.0. Ninety-two long-term survivors responded to the QOL (EORTC QLQ-C30) and Chemotherapy-Induced Peripheral Neuropathy (EORTC CIPN20) questionnaires and were interviewed regarding long-term neuropathy. RESULTS Acute neurotoxicity was present in 94% (136/144) during adjuvant therapy and there was a significant association between acute neurotoxicity and long-term neuropathy (p < .001). Long-term neuropathy was present in 69% (grade 1/2/3/4 in 36/24/8/1%) at median 4.2 years. Neuropathy grades 2-4 did not influence global health status, but it was associated with decreased physical functioning (p = .031), decreased role functioning (p = .040), and more diarrhea (p = .021) in QLQ-C30 items. There were no differences in acute neurotoxicity, long-term neuropathy, or in QOL between CAPOX and FOLFOX treated. Neuropathy showed no pattern of variation according to starting and stopping month or treatment during winter. CONCLUSIONS Neuropathy following oxaliplatin containing adjuvant chemotherapy is present in two-thirds, years after cessation, and impairs some QOL scales. There is no difference in severity of acute or long-term neuropathy between CAPOX and FOLFOX treated and QOL is similar. No seasonal variation in neuropathy was noted.
Collapse
Affiliation(s)
- L. M. Soveri
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Oncology at Clinicum, University of Helsinki, Helsinki, Finland
| | - A. Lamminmäki
- Department of Oncology, Kuopio University Central Hospital, Helsinki, Finland
| | - U. A. Hänninen
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - M. Karhunen
- Medical Bioinformatics Centre, Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - P. Bono
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Oncology at Clinicum, University of Helsinki, Helsinki, Finland
| | - P. Osterlund
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Oncology at Clinicum, University of Helsinki, Helsinki, Finland
- Department of Oncology, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| |
Collapse
|
38
|
Henricks LM, van Merendonk LN, Meulendijks D, Deenen MJ, Beijnen JH, de Boer A, Cats A, Schellens JHM. Effectiveness and safety of reduced-dose fluoropyrimidine therapy in patients carrying the DPYD*2A variant: A matched pair analysis. Int J Cancer 2019; 144:2347-2354. [PMID: 30485432 DOI: 10.1002/ijc.32022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/06/2018] [Indexed: 12/27/2022]
Abstract
Carriers of the genetic DPYD*2A variant, resulting in dihydropyrimidine dehydrogenase deficiency, are at significantly increased risk of developing severe fluoropyrimidine-associated toxicity. Upfront DPYD*2A genotype-based dose reductions improve patient safety, but uncertainty exists whether this has a negative impact on treatment effectiveness. Therefore, our study investigated effectiveness and safety of DPYD*2A genotype-guided dosing. A cohort of 40 prospectively identified heterozygous DPYD*2A carriers, treated with a ~50% reduced fluoropyrimidine dose, was identified. For effectiveness analysis, a matched pair-analysis was performed in which for each DPYD*2A carrier a matched DPYD*2A wild-type patient was identified. Overall survival and progression-free survival were compared between the matched groups. The frequency of severe (grade ≥ 3) treatment-related toxicity was compared to 1] a cohort of 1606 wild-type patients treated with full dose and 2] a cohort of historical controls derived from literature, i.e. 86 DPYD*2A variant carriers who received a full fluoropyrimidine dose. For 37 out of 40 DPYD*2A carriers, a matched control could be identified. Compared to matched controls, reduced doses did not negatively affect overall survival (median 27 months versus 24 months, p = 0.47) nor progression-free survival (median 14 months versus 10 months, p = 0.54). Risk of severe fluoropyrimidine-related toxicity in DPYD*2A carriers treated with reduced dose was 18%, comparable to wild-type patients (23%, p = 0.57) and significantly lower than the risk of 77% in DPYD*2A carriers treated with full dose (p < 0.001). Our study is the first to show that DPYD*2A genotype-guided dosing appears to have no negative effect on effectiveness of fluoropyrimidine-based chemotherapy, while resulting in significantly improved patient safety.
Collapse
Affiliation(s)
- Linda M Henricks
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lisanne N van Merendonk
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Didier Meulendijks
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Dutch Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, The Netherlands.,Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Anthonius de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Annemieke Cats
- Department of Gastrointestinal Oncology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan H M Schellens
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
39
|
Lam YWF. Principles of Pharmacogenomics. Pharmacogenomics 2019. [DOI: 10.1016/b978-0-12-812626-4.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
40
|
Henricks LM, Jacobs BAW, Meulendijks D, Pluim D, van den Broek D, de Vries N, Rosing H, Beijnen JH, Huitema ADR, Guchelaar H, Cats A, Schellens JHM. Food-effect study on uracil and dihydrouracil plasma levels as marker for dihydropyrimidine dehydrogenase activity in human volunteers. Br J Clin Pharmacol 2018; 84:2761-2769. [PMID: 30047584 PMCID: PMC6256055 DOI: 10.1111/bcp.13719] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/05/2018] [Accepted: 07/14/2018] [Indexed: 12/31/2022] Open
Abstract
AIMS This study aimed to determine the effect of food intake on uracil and dihydrouracil plasma levels. These levels are a promising marker for dihydropyrimidine dehydrogenase activity and for individualizing fluoropyrimidine anticancer therapy. METHODS A randomized, cross-over study in 16 healthy volunteers was performed, in which subjects were examined in fasted and fed state on two separate days. In fed condition, a high-fat, high-caloric breakfast was consumed between 8:00 h and 8:30 h. Whole blood for determination of uracil, dihydrouracil and uridine plasma levels was drawn on both test days at predefined time points between 8:00 h and 13:00 h. RESULTS Uracil levels were statistically significantly different between fasting and fed state. At 13:00 h, the mean uracil level in fasting state was 12.6 ± 3.7 ng ml-1 and after a test meal 9.4 ± 2.6 ng ml-1 (P < 0.001). Dihydrouracil levels were influenced by food intake as well (mean dihydrouracil level at 13:00 h in fasting state 147.0 ± 36.4 ng ml-1 and in fed state 85.7 ± 22.1 ng ml-1 , P < 0.001). Uridine plasma levels showed curves with similar patterns as for uracil. CONCLUSIONS It was shown that both uracil and dihydrouracil levels were higher in fasting state than in fed state. This is hypothesized to be an direct effect of uridine plasma levels, which were previously shown to be elevated in fasting state and reduced after intake of food. These findings show that, when assessing plasma uracil and dihydrouracil levels for adaptive fluoropyrimidine dosing in clinical practice, sampling should be done between 8:00 h and 9:00 h after overnight fasting to avoid bias caused by circadian rhythm and food effects.
Collapse
Affiliation(s)
- Linda M. Henricks
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical Pharmacology, Division of Medical OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Bart A. W. Jacobs
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Didier Meulendijks
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical Pharmacology, Division of Medical OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Dutch Medicines Evaluation Board (CBG‐MEB)UtrechtThe Netherlands
| | - Dick Pluim
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical Pharmacology, Division of Medical OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daan van den Broek
- Department of Clinical ChemistryThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Niels de Vries
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Hilde Rosing
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos H. Beijnen
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Alwin D. R. Huitema
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical PharmacyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Henk‐Jan Guchelaar
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | - Annemieke Cats
- Department of Gastrointestinal Oncology, Division of Medical OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jan H. M. Schellens
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical Pharmacology, Division of Medical OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
41
|
Korver SK, Gibson RJ, Bowen JM, Coller JK. Toll-like receptor/interleukin-1 domain innate immune signalling pathway genetic variants are candidate predictors for severe gastrointestinal toxicity risk following 5-fluorouracil-based chemotherapy. Cancer Chemother Pharmacol 2018; 83:217-236. [PMID: 30474704 DOI: 10.1007/s00280-018-3729-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Severe gastrointestinal (GI) toxicity is a common adverse effect following 5-fluorouracil (5-FU)-based chemotherapy treatment. The presence of severe GI toxicity leads to treatment revisions, sub-optimal therapy outcomes, and decreases to patients' quality of life. There are no adequate predictors for 5-FU-induced severe GI toxicity risk. The Toll-like receptor/interleukin-1 (TIR) domain innate immune signalling pathway is known to be a mediating pathway in the development of GI toxicity. Hence, genetic variability in this signalling pathway may alter the pathophysiology of GI toxicity and, therefore, be predictive of risk. However, little research has investigated the effects of TIR domain innate immune signalling pathway single nucleotide polymorphism (SNPs) on the risk and development of severe GI toxicity. METHODS This critical review surveyed the literature and reported on the in vitro, ex vivo and in vivo effects, as well as the genetic association, of selected TIR domain innate immune signalling pathway SNPs on disease susceptibility and gene functioning. RESULTS Of the TIR domain innate immune signalling pathway SNPs reviewed, evidence suggests interleukin-1 beta (IL1B) and tumour necrosis factor alpha (TNF) SNPs have the greatest potential as predictors for severe GI toxicity risk. These results warrant further research into the effect of IL1B and TNF SNPs on the risk and development of severe GI toxicity. CONCLUSIONS SNPs of the TIR domain innate immune signalling pathway have profound effects on disease susceptibility and gene functioning, making them candidate predictors for severe GI toxicity risk. The identification of a predictor for 5-FU-induced severe GI toxicity will allow the personalization of supportive care measures.
Collapse
Affiliation(s)
- Samantha K Korver
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia.
| | - Rachel J Gibson
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia.,Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Joanne M Bowen
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia
| | - Janet K Coller
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia
| |
Collapse
|
42
|
Tariq Z, Mudge R, Phillips S. Capecitabine Toxicity and Dihydropyrimidine Dehydrogenase Deficiency. Am J Ther 2018; 25:e742-e743. [DOI: 10.1097/mjt.0000000000000753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Madi A, Fisher D, Maughan TS, Colley JP, Meade AM, Maynard J, Humphreys V, Wasan H, Adams RA, Idziaszczyk S, Harris R, Kaplan RS, Cheadle JP. Pharmacogenetic analyses of 2183 patients with advanced colorectal cancer; potential role for common dihydropyrimidine dehydrogenase variants in toxicity to chemotherapy. Eur J Cancer 2018; 102:31-39. [PMID: 30114658 DOI: 10.1016/j.ejca.2018.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/29/2018] [Accepted: 07/08/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inherited genetic variants may influence response to, and side-effects from, chemotherapy. We sought to generate a comprehensive inherited pharmacogenetic profile for oxaliplatin and 5FU/capecitabine therapy in advanced colorectal cancer (aCRC). METHODS We analysed more than 200 potentially functional, common, inherited variants in genes within the 5FU, capecitabine, oxaliplatin and DNA repair pathways, together with four rare dihydropyrimidine dehydrogenase (DPYD) variants, in 2183 aCRC patients treated with oxaliplatin-fluoropyrimidine chemotherapy with, or without, cetuximab (from MRC COIN and COIN-B trials). Primary end-points were response, any toxicity and peripheral neuropathy. We had >85% power to detect odds ratios (ORs) = 1.3 for variants with minor allele frequencies >20%. RESULTS Variants in DNA repair genes (Asn279Ser in EXO1 and Arg399Gln in XRCC1) were most associated with response (OR 1.9, 95% confidence interval [CI] 1.2-2.9, P = 0.004, and OR 0.7, 95% CI 0.5-0.9, P = 0.003, respectively). Common variants in DPYD (Cys29Arg and Val732Ile) were most associated with toxicity (OR 0.8, 95% CI 0.7-1.0, P = 0.008, and OR 1.6, 95% CI 1.1-2.1, P = 0.006, respectively). Two rare DPYD variants were associated with increased toxicity (Asp949Val with neutropenia, nausea and vomiting, diarrhoea and infection; IVS14+1G>A with lethargy, diarrhoea, stomatitis, hand-foot syndrome and infection; all ORs > 3). Asp317His in DCLRE1A was most associated with peripheral neuropathy (OR 1.3, 95% CI 1.1-1.6, P = 0.003). No common variant associations remained significant after Bonferroni correction. CONCLUSIONS DNA repair genes may play a significant role in the pharmacogenetics of aCRC. Our data suggest that both common and rare DPYD variants may be associated with toxicity to fluoropyrimidine-based chemotherapy.
Collapse
Affiliation(s)
- Ayman Madi
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - David Fisher
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Timothy S Maughan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - James P Colley
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Angela M Meade
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Julie Maynard
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Vikki Humphreys
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Harpreet Wasan
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK
| | - Richard A Adams
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Shelley Idziaszczyk
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Rebecca Harris
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Richard S Kaplan
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Jeremy P Cheadle
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
44
|
Castro-Rojas CA, Esparza-Mota AR, Hernandez-Cabrera F, Romero-Diaz VJ, Gonzalez-Guerrero JF, Maldonado-Garza H, Garcia-Gonzalez IS, Buenaventura-Cisneros S, Sanchez-Lopez JY, Ortiz-Lopez R, Camacho-Morales A, Barboza-Quintana O, Rojas-Martinez A. Thymidylate synthase gene variants as predictors of clinical response and toxicity to fluoropyrimidine-based chemotherapy for colorectal cancer. Drug Metab Pers Ther 2018; 32:209-218. [PMID: 29257755 DOI: 10.1515/dmpt-2017-0028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/15/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Fluoropyrimidines form the chemotherapy backbone of advanced and metastatic colorectal cancer (CRC). These drugs are frequently associated with toxicity events that result in dose adjustments and even suspension of the treatment. The thymidylate synthase (TYMS) gene is a potential marker of response and toxicity to fluoropyirimidines as this enzyme is the molecular target of these drugs. Our aim was to assess the association between variants of TYMS with response and toxicity to fluoropyrimidines in patients with CRC in independent retrospective and prospective studies. METHODS Variants namely rs45445694, rs183205964, rs2853542 and rs151264360 of TYMS were genotyped in 105 CRC patients and were evaluated to define their association with clinical response and toxicity to fluoropyrimidines. Additionally, the relationship between genotypes and tumor gene expression was analyzed by quantitative polymerase chain reaction. RESULTS The 2R/2R (rs45445694) was associated with clinical response (p=0.05, odds ratio (OR)=3.45) and severe toxicity (p=0.0014, OR=5.21, from pooled data). Expression analysis in tumor tissues suggested a correlation between the 2R/2R genotype and low TYMS expression. CONCLUSIONS The allele 2R (rs45445694) predicts severe toxicity and objective response in advanced CRC patients. In addition, the alleles G(rs2853542) and 6bp-(rs151264360) are independent predictors of response failure to chemotherapy. This is the first study made on a Latin American population that points out TYMS gene variants have predictive values for response and toxicity in patients with CRC treated with fluoropyrimidine-based chemotherapy.
Collapse
Affiliation(s)
- Carlos A Castro-Rojas
- Universidad Autonoma de Nuevo Leon, Center for Research and Development in Health Sciences, Monterrey, Mexico
| | - Antonio R Esparza-Mota
- Universidad Autonoma de Nuevo Leon, Center for Research and Development in Health Sciences, Monterrey, Mexico
| | | | - Viktor J Romero-Diaz
- Universidad Autonoma de Nuevo Leon, Center for Research and Development in Health Sciences, Monterrey, Mexico
| | | | - Hector Maldonado-Garza
- Universidad Autonoma de Nuevo Leon, Service of Gastroenterology, University Hospital, Monterrey, Mexico
| | - Irma S Garcia-Gonzalez
- Mexican Institute for Social Security (IMSS), High Specialties Unit No. 25, Monterrey, Mexico
| | | | - Josefina Y Sanchez-Lopez
- Mexican Institute for Social Security (IMSS), Western Center for Biomedical Research, Guadalajara, Mexico
| | - Rocio Ortiz-Lopez
- Universidad Autonoma de Nuevo Leon, Center for Research and Development in Health Sciences, Monterrey, Mexico
| | | | | | - Augusto Rojas-Martinez
- Universidad Autonoma de Nuevo Leon, Center for Research and Development in Health Sciences, Monterrey, Mexico
| |
Collapse
|
45
|
Tong CC, Lam CW, Lam KO, Lee VHF, Luk MY. A Novel DPYD Variant Associated With Severe Toxicity of Fluoropyrimidines: Role of Pre-emptive DPYD Genotype Screening. Front Oncol 2018; 8:279. [PMID: 30087856 PMCID: PMC6066555 DOI: 10.3389/fonc.2018.00279] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 07/04/2018] [Indexed: 12/27/2022] Open
Abstract
Background: The fluoropyrimidine anticancer drug, especially 5- fluorouracil (5-FU) and its prodrug capecitabine are still being the backbone of chemotherapeutic regimens for colorectal cancer. Dihydropyrimidine dehydrogenase (DPD) is the crucial enzyme in the catabolism of 5-FU. Over the past 30 years, there is substantial clinical evidence showing that DPD deficiency is strongly associated with severe and fatal fluoropyrimidine-induced toxicity. Patients and methods: A 49-year-old lady with resected stage III carcinoma of sigmoid colon was scheduled to have a course of 5-FU based adjuvant chemotherapy. She developed unexpected acute severe (grade 4) toxicity after the first cycle of chemotherapy. Genomic DNA was isolated from 3 ml peripheral blood cells for full sequencing of DPYD (the gene encoding DPD). Results: Exome sequencing confirmed that she is heterozygous for NM_000110.3: c.321+2T>C of the DPYD gene. To the best of our knowledge, this variant is a novel pathogenic splicing variant of the DPYD gene resulting in a non-functional allele. As she has a heterozygous genotype and considered having decreased DPD activity, we followed the international recommendation and restart chemotherapy with at least 50% reduction for 5-FU dose. We then titrated the 5-FU dose, and she tolerated the subsequent cycles of chemotherapy and completed the whole course of adjuvant chemotherapy. Conclusions: With a pre-emptive test on DPD deficiency before the administration of the fluoropyrimidine drugs, the aforementioned patient's life-threatening event could be avoided. This clinical utility has been confirmed by two recent large-scale studies and called for a drug label update.
Collapse
Affiliation(s)
- Chi C Tong
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Ching W Lam
- Department of Pathology, University of Hong Kong, Pokfulam, Hong Kong
| | - Ka O Lam
- Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong
| | - Victor H F Lee
- Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong
| | - Mai-Yee Luk
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, Hong Kong
| |
Collapse
|
46
|
Khushman M, Patel GK, Hosein PJ, Laurini JA, Cameron D, Clarkson DR, Butler TW, Norden CW, Baliem W, Jones V, Bhadkamkar S, Nelson C, Lee F, Singh AP, Taylor WR. Germline pharmacogenomics of DPYD*9A (c.85T>C) variant in patients with gastrointestinal malignancies treated with fluoropyrimidines. J Gastrointest Oncol 2018; 9:416-424. [PMID: 29998006 DOI: 10.21037/jgo.2018.02.03] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background The correlation between DPYD*9A (c.85T>C) genotype and dihydropyrimidine dehydrogenase (DPD) deficiency clinical phenotype is controversial. Reference laboratories either did not perform DPYD*9A genotyping or have stopped DPYD*9A genotyping and limited genotyping to high-risk variants (DPYD*2A, DPYD*13 and DPYD*9B) only. This study explored DPYD*9A genotype and clinical phenotype correlation in patients with gastrointestinal (GI) malignancies treated with fluoropyrimidines. Methods Between 2011 and 2017, 67 patients with GI malignancies were genotyped for DPYD variants. Fluoropyrimidines-associated toxicity was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (version 3.0). Fisher's exact test was used for statistical analysis. Results DPYD variants were identified in 17 out of 67 (25%) patients. One patient was homozygous for DPYD*9A variant and one patient was double heterozygous for DPYD*9A and DPYD*9B variants. In patients with identified DPYD variants, 13/17 (76%) patients had DPYD*9A variant, 3/17 (18%) patients had DPYD*2A variant and 2/17 (12%) patient had DPYD*9B variant. Only patients genotyped prior to 2015 were genotyped for DPYD*9A variant (N=28). Of those, 13/28 patients (46%) had DPYD*9A variant. Grade 3-4 diarrhea was associated with DPYD*9A variant in patients treated with full dose fluoropyrimidines (P=0.0055). Conclusions In our cohort, DPYD*9A variant was the most common diagnosed variant. The correlation between DPYD*9A genotype and DPD deficiency in clinical phenotype was noticeable in patients who received full dose fluoropyrimidines as they all experienced grade 3-4 toxicities (diarrhea).
Collapse
Affiliation(s)
- Moh'd Khushman
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Girijesh Kumar Patel
- Department of Oncologic Sciences, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Peter Joel Hosein
- Medical Oncology, The University of Miami, Sylvester Cancer Center, Miami, Florida, USA
| | | | - Daniel Cameron
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - David Roland Clarkson
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Thomas Wayne Butler
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Carole Wiseman Norden
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Wilma Baliem
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Vanessa Jones
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Sanjyot Bhadkamkar
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Cindy Nelson
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Frances Lee
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | - William R Taylor
- Medical Oncology, The University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| |
Collapse
|
47
|
DPD functional tests in plasma, fresh saliva and dried saliva samples as predictors of 5-fluorouracil exposure and occurrence of drug-related severe toxicity. Clin Biochem 2018; 56:18-25. [DOI: 10.1016/j.clinbiochem.2018.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 01/07/2023]
|
48
|
Battaglin F, Puccini A, Naseem M, Schirripa M, Berger MD, Tokunaga R, McSkane M, Khoukaz T, Soni S, Zhang W, Lenz HJ. Pharmacogenomics in colorectal cancer: current role in clinical practice and future perspectives. JOURNAL OF CANCER METASTASIS AND TREATMENT 2018; 4:12. [PMID: 34532592 PMCID: PMC8442855 DOI: 10.20517/2394-4722.2018.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment scenario of colorectal cancer (CRC) has been evolving in recent years with the introduction of novel targeted agents and new therapeutic strategies for the metastatic disease. An extensive effort has been directed to the identification of predictive biomarkers to aid patients selection and guide therapeutic choices. Pharmacogenomics represents an irreplaceable tool to individualize patients treatment based on germline and tumor acquired somatic genetic variations able to predict drugs response and risk of toxicities. The growing knowledge of CRC molecular characteristics and complex genomic makeup has played a crucial role in identifying predictive pharmacogenomic biomarkers, while supporting the rationale for the development of new drugs and treatment combinations. Clinical validation of promising biomarkers, however, is often an issue. More recently, a deeper understanding of resistance mechanisms and tumor escape dynamics under treatment pressure and the availability of novel technologies are opening new perspectives in this field. This review aims to present an overview of current pharmacogenomic biomarkers and future perspectives of pharmacogenomics in CRC, in an evolving scenario moving from a single drug-gene interactions approach to a more comprehensive genome-wide approach, comprising genomics and epigenetics.
Collapse
Affiliation(s)
- Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV - IRCCS, Padua 35128, Italy
| | - Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Marta Schirripa
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV - IRCCS, Padua 35128, Italy
| | - Martin D. Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Medical Oncology, University Hospital of Bern, Bern 3010, Switzerland
| | - Ryuma Tokunaga
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michelle McSkane
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Taline Khoukaz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
49
|
DMET™ (Drug Metabolism Enzymes and Transporters): a pharmacogenomic platform for precision medicine. Oncotarget 2018; 7:54028-54050. [PMID: 27304055 PMCID: PMC5288240 DOI: 10.18632/oncotarget.9927] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/29/2016] [Indexed: 02/07/2023] Open
Abstract
In the era of personalized medicine, high-throughput technologies have allowed the investigation of genetic variations underlying the inter-individual variability in drug pharmacokinetics/pharmacodynamics. Several studies have recently moved from a candidate gene-based pharmacogenetic approach to genome-wide pharmacogenomic analyses to identify biomarkers for selection of patient-tailored therapies. In this aim, the identification of genetic variants affecting the individual drug metabolism is relevant for the definition of more active and less toxic treatments. This review focuses on the potentiality, reliability and limitations of the DMET™ (Drug Metabolism Enzymes and Transporters) Plus as pharmacogenomic drug metabolism multi-gene panel platform for selecting biomarkers in the final aim to optimize drugs use and characterize the individual genetic background.
Collapse
|
50
|
Henricks LM, Opdam FL, Beijnen JH, Cats A, Schellens JHM. DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update. Ann Oncol 2017; 28:2915-2922. [PMID: 29045513 DOI: 10.1093/annonc/mdx411] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The fluoropyrimidine anticancer drugs, especially 5-fluorouracil (5-FU) and capecitabine, are frequently prescribed for several types of cancer, including breast, colorectal, head and neck and gastric cancer. In the current drug labels of 5-FU and capecitabine in the European Union and the United States, no adaptive dosing strategies are incorporated for polymorphic metabolism of 5-FU. Although treatment with fluoropyrimidines is generally well tolerated, a major clinical limitation is that a proportion of the treated population experiences severe, sometimes life-threatening, fluoropyrimidine-related toxicity. This toxicity is strongly affected by interindividual variability in activity of dihydropyrimidine dehydrogenase (DPD), the main metabolic enzyme for inactivation of fluoropyrimidines, with an estimated 3%-8% of the population being partially DPD deficient. A reduced functional or abrogated DPD enzyme is often caused by genetic polymorphisms in DPYD, the gene encoding for DPD, and heterozygous carriers of such DPYD polymorphisms have a partial DPD deficiency. When these partially DPD deficient patients are treated with a full dose of fluoropyrimidines, they are generally exposed to toxic levels of 5-FU and its metabolites, and the risk of developing severe treatment-related toxicity is therefore significantly increased.Currently, functional and clinical validity is well established for four DPYD variants (DPYD*2A, c.2846A>T, c.1679T>G and c.1236G>A), as those variants have retrospectively and in a large population study prospectively been shown to be associated with increased risk of fluoropyrimidine-associated toxicity. Patient safety of fluoropyrimidine treatment can be significantly improved by pre-emptive screening for DPYD genotype variants and dose reductions in heterozygous DPYD variant allele carriers, thereby normalizing 5-FU exposure. Based on the critical appraisal of currently available data, adjusting the labels of capecitabine and 5-FU by including recommendations on pre-emptive screening for DPYD variants and DPYD genotype-guided dose adjustments should be the new standard of care.
Collapse
Affiliation(s)
- L M Henricks
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - F L Opdam
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| | - A Cats
- Department of Gastroenterology and Hepatology, Division of Medical Oncology
| | - J H M Schellens
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| |
Collapse
|