101
|
Lunenburg CATC, Henricks LM, van Kuilenburg ABP, Mathijssen RHJ, Schellens JHM, Gelderblom H, Guchelaar HJ, Swen JJ. Diagnostic and Therapeutic Strategies for Fluoropyrimidine Treatment of Patients Carrying Multiple DPYD Variants. Genes (Basel) 2018; 9:E585. [PMID: 30487465 PMCID: PMC6316498 DOI: 10.3390/genes9120585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/16/2023] Open
Abstract
DPYD genotyping prior to fluoropyrimidine treatment is increasingly implemented in clinical care. Without phasing information (i.e., allelic location of variants), current genotype-based dosing guidelines cannot be applied to patients carrying multiple DPYD variants. The primary aim of this study is to examine diagnostic and therapeutic strategies for fluoropyrimidine treatment of patients carrying multiple DPYD variants. A case series of patients carrying multiple DPYD variants is presented. Different genotyping techniques were used to determine phasing information. Phenotyping was performed by dihydropyrimidine dehydrogenase (DPD) enzyme activity measurements. Publicly available databases were queried to explore the frequency and phasing of variants of patients carrying multiple DPYD variants. Four out of seven patients carrying multiple DPYD variants received a full dose of fluoropyrimidines and experienced severe toxicity. Phasing information could be retrieved for four patients. In three patients, variants were located on two different alleles, i.e., in trans. Recommended dose reductions based on the phased genotype differed from the phenotype-derived dose reductions in three out of four cases. Data from publicly available databases show that the frequency of patients carrying multiple DPYD variants is low (< 0.2%), but higher than the frequency of the commonly tested DPYD*13 variant (0.1%). Patients carrying multiple DPYD variants are at high risk of developing severe toxicity. Additional analyses are required to determine the correct dose of fluoropyrimidine treatment. In patients carrying multiple DPYD variants, we recommend that a DPD phenotyping assay be carried out to determine a safe starting dose.
Collapse
Affiliation(s)
- Carin A T C Lunenburg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Linda M Henricks
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - André B P van Kuilenburg
- Department of Clinical Chemistry, Amsterdam University Medical Centre, 1105 AZ Amsterdam, The Netherlands.
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands.
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
102
|
Goirand F, Lemaitre F, Launay M, Tron C, Chatelut E, Boyer JC, Bardou M, Schmitt A. How can we best monitor 5-FU administration to maximize benefit to risk ratio? Expert Opin Drug Metab Toxicol 2018; 14:1303-1313. [PMID: 30451549 DOI: 10.1080/17425255.2018.1550484] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION 5-Fluorouracil (5-FU) is currently used as a chemotherapy in several cancers such as head-and-neck (H&N) and colorectal cancers. 5-FU dosing is traditionally based on body surface area (BSA), but this strategy is usually associated with severe toxicities. 5-FU is mainly catabolized by dihydropyrimidine dehydrogenase (DPD), and 5-FU dosage adaptation according to DPD status at the first cycle of treatment is now recommended. To further optimize 5-FU-based chemotherapy, a body of evidences justifies therapeutic drug monitoring (TDM). Areas covered: 5-FU pharmacokinetics, relationships between pharmacokinetics and efficacy or toxicity of 5-FU, proofs of interest of 5-FU TDM and its practical considerations are discussed. Expert opinion: BSA-adjusted 5-FU administration is associated with a large inter-individual variability, and according to this strategy, many patients experience under- or overexposure. Moreover, relationships between 5-FU area under the curve (AUC) and its toxicity or efficacy have been demonstrated, at least in patients with colorectal or H&N cancers. 5-FU therapeutic index has been validated and algorithms of 5-FU dosage adaptation according to its AUC are now available. Advances in pre-analytical and analytical steps of 5-FU TDM make its use feasible in clinical practice. Thus, there are consistent evidences to recommend 5-FU TDM in patients with advanced colorectal or H&N cancers.
Collapse
Affiliation(s)
- Françoise Goirand
- a Centre Hospitalo-Universitaire de Dijon-Bourgogne , Dijon , France
| | - Florian Lemaitre
- b Service de Pharmacologie Clinique , Laboratoire de Pharmacologie Expérimentale et Clinique, Centre d'Investigation Clinique INSERM 1414, CHU de Rennes, Université Rennes 1 , Rennes , France
| | - Manon Launay
- c Service de Pharmacologie, Hôpital Européen Georges Pompidou , Laboratoire de Pharmacologie et de Toxicologie , Paris , France
| | - Camille Tron
- b Service de Pharmacologie Clinique , Laboratoire de Pharmacologie Expérimentale et Clinique, Centre d'Investigation Clinique INSERM 1414, CHU de Rennes, Université Rennes 1 , Rennes , France
| | - Etienne Chatelut
- d Institut Claudius-Regaud et Centre de Recherches en Cancérologie de Toulouse , IUCT - Oncopole , Toulouse , France
| | - Jean-Christophe Boyer
- e Laboratoire de Biochimie et Biologie Moléculaire , CHU de Nîmes Carémeau , Nîmes , France
| | - Marc Bardou
- f Centre d'Investigations Cliniques 1432, Module Plurithématique , CHU Dijon-Bourgogne, Hôpital François Mitterrand , Dijon , France
| | - Antonin Schmitt
- g Service Pharmacie , Centre Georges-François Leclerc , Dijon , France
| |
Collapse
|
103
|
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: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
104
|
Dihydropyrimidine Dehydrogenase Deficiency: Homozygosity for an Extremely Rare Variant in DPYD due to Uniparental Isodisomy of Chromosome 1. JIMD Rep 2018. [PMID: 30349988 DOI: 10.1007/8904_2018_138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Dihydropyrimidine dehydrogenase (DPD) deficiency is a rare autosomal recessive disorder of the pyrimidine degradation pathway and can lead to intellectual disability, motor retardation, and seizures. Genetic variations in DPYD have also emerged as predictive risk factors for severe toxicity in cancer patients treated with fluoropyrimidines. We recently observed a child born to non-consanguineous parents, who demonstrated seizures, cognitive impairment, language delay, and MRI abnormalities and was found to have marked thymine-uraciluria. No residual DPD activity could be detected in peripheral blood mononuclear cells. Molecular analysis showed that the child was homozygous for the very rare c.257C > T (p.Pro86Leu) variant in DPYD. Functional analysis of the recombinantly expressed DPD mutant showed that the DPD mutant carrying the p.Pro86Leu did not possess any residual DPD activity. Carrier testing in parents revealed that the father was heterozygous for the variant but unexpectedly the mother did not carry the variant. Microsatellite repeat testing with markers covering chromosome 1 showed that the DPD deficiency in the child is due to paternal uniparental isodisomy. Our report thus extends the genetic spectrum underlying DPYD deficiency.
Collapse
|
105
|
Function and crystal structure of the dimeric P-loop ATPase CFD1 coordinating an exposed [4Fe-4S] cluster for transfer to apoproteins. Proc Natl Acad Sci U S A 2018; 115:E9085-E9094. [PMID: 30201724 DOI: 10.1073/pnas.1807762115] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Maturation of iron-sulfur (Fe-S) proteins in eukaryotes requires complex machineries in mitochondria and cytosol. Initially, Fe-S clusters are assembled on dedicated scaffold proteins and then are trafficked to target apoproteins. Within the cytosolic Fe-S protein assembly (CIA) machinery, the conserved P-loop nucleoside triphosphatase Nbp35 performs a scaffold function. In yeast, Nbp35 cooperates with the related Cfd1, which is evolutionary less conserved and is absent in plants. Here, we investigated the potential scaffold function of human CFD1 (NUBP2) in CFD1-depleted HeLa cells by measuring Fe-S enzyme activities or 55Fe incorporation into Fe-S target proteins. We show that CFD1, in complex with NBP35 (NUBP1), performs a crucial role in the maturation of all tested cytosolic and nuclear Fe-S proteins, including essential ones involved in protein translation and DNA maintenance. CFD1 also matures iron regulatory protein 1 and thus is critical for cellular iron homeostasis. To better understand the scaffold function of CFD1-NBP35, we resolved the crystal structure of Chaetomium thermophilum holo-Cfd1 (ctCfd1) at 2.6-Å resolution as a model Cfd1 protein. Importantly, two ctCfd1 monomers coordinate a bridging [4Fe-4S] cluster via two conserved cysteine residues. The surface-exposed topology of the cluster is ideally suited for both de novo assembly and facile transfer to Fe-S apoproteins mediated by other CIA factors. ctCfd1 specifically interacted with ATP, which presumably associates with a pocket near the Cfd1 dimer interface formed by the conserved Walker motif. In contrast, ctNbp35 preferentially bound GTP, implying differential regulation of the two fungal scaffold components during Fe-S cluster assembly and/or release.
Collapse
|
106
|
Feasibility of 5-fluorouracil pharmacokinetic monitoring using the My-5FU PCM™ system in a quaternary oncology centre. Cancer Chemother Pharmacol 2018; 82:865-876. [DOI: 10.1007/s00280-018-3679-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/27/2018] [Indexed: 12/16/2022]
|
107
|
Hishinuma E, Narita Y, Saito S, Maekawa M, Akai F, Nakanishi Y, Yasuda J, Nagasaki M, Yamamoto M, Yamaguchi H, Mano N, Hirasawa N, Hiratsuka M. Functional Characterization of 21 Allelic Variants of Dihydropyrimidine Dehydrogenase Identified in 1070 Japanese Individuals. Drug Metab Dispos 2018; 46:1083-1090. [PMID: 29769267 DOI: 10.1124/dmd.118.081737] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/11/2018] [Indexed: 11/22/2022] Open
Abstract
Dihydropyrimidine dehydrogenase (DPD, EC 1.3.1.2), encoded by the DPYD gene, is the rate-limiting enzyme in the degradation pathway of endogenous pyrimidine and fluoropyrimidine drugs such as 5-fluorouracil (5-FU). DPD catalyzes the reduction of uracil, thymine, and 5-FU. In Caucasians, DPYD mutations, including DPYD*2A, DPYD*13, c.2846A>T, and c.1129-5923C>G/hapB3, are known to contribute to interindividual variations in the toxicity of 5-FU; however, none of these DPYD polymorphisms has been identified in the Asian population. Recently, 21 DPYD allelic variants, including some novel single-nucleotide variants (SNVs), were identified in 1070 healthy Japanese individuals by analyzing their whole-genome sequences (WGSs), but the functional alterations caused by these variants remain unknown. In this study, in vitro analysis was performed on 22 DPD allelic variants by transiently expressing wild-type DPD and 21 DPD variants in 293FT cells and characterizing their enzymatic activities using 5-FU as a substrate. DPD expression levels and dimeric forms were determined using immunoblotting and blue-native PAGE, respectively. Additionally, the values of three kinetic parameters-the Michaelis constant (Km ), maximum velocity (Vmax ), and intrinsic clearance (CLint = Vmax/Km )-were determined for the reduction of 5-FU. Eleven variants exhibited significantly decreased intrinsic clearance compared with wild-type DPD. Moreover, the band patterns observed in the immunoblots of blue-native gels indicated that DPD dimerization is required for enzymatic activity in DPD. Thus, the detection of rare DPYD variants might facilitate severe adverse effect prediction of 5-FU-based chemotherapy in the Japanese population.
Collapse
Affiliation(s)
- Eiji Hishinuma
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Yoko Narita
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Sakae Saito
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masamitsu Maekawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Fumika Akai
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Yuya Nakanishi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Jun Yasuda
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masao Nagasaki
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masayuki Yamamoto
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Hiroaki Yamaguchi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Nariyasu Mano
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| |
Collapse
|
108
|
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: 2.1] [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
|
109
|
Pires AS, Marques CR, Encarnação JC, Abrantes AM, Marques IA, Laranjo M, Oliveira R, Casalta-Lopes JE, Gonçalves AC, Sarmento-Ribeiro AB, Botelho MF. Ascorbic Acid Chemosensitizes Colorectal Cancer Cells and Synergistically Inhibits Tumor Growth. Front Physiol 2018; 9:911. [PMID: 30083105 PMCID: PMC6064950 DOI: 10.3389/fphys.2018.00911] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is continuously classified as one of the most incidental and mortal types of cancer worldwide. The positive outcomes of the conventional chemotherapy are frequently associated with high toxicity, which often leads to the suspension of the treatment. Growing evidences consider the use of pharmacological concentrations of ascorbic acid (AA), better known as vitamin C, in the treatment of cancer. The use of AA in a clinical context is essentially related to the adoption of new therapeutic strategies based on combination regimens, where AA plays a chemosensitizing role. The reduced sensitivity of some tumors to chemotherapy and the highly associated adverse effects continue to be some of the major obstacles in the effective treatment of CRC. So, this paper aimed to study the potential of a new therapeutic approach against this neoplasia with diminished side effects for the patient. This approach was based on the study of the combination of high concentrations of AA with reduced concentrations of drugs conventionally used in CRC patients and eligible for first and second line chemotherapeutic regimens, namely 5-fluorouracilo (5-FU), oxaliplatin (Oxa) or irinotecan (Iri). The evaluation of the potential synergy between the compounds was first assessed in vitro in three CRC cell lines with different genetic background and later in vivo using one xenograft animal model of CRC. AA and 5-FU act synergistically in vitro just for longer incubation times, however, in vivo showed no benefit compared to 5-FU alone. In contrast to the lack of synergy seen in in vitro studies with the combination of AA with irinotecan, the animal model revealed the therapeutic potential of this combination. AA also potentiated the effect of Oxa, since a synergistic effect was demonstrated, in almost all conditions and in the three cell lines. Moreover, this combined therapy (CT) caused a stagnation of the tumor growth rate, being the most promising tested combination. Pharmacological concentrations of AA increased the efficacy of Iri and Oxa against CRC, with promising results in cell lines with more aggressive phenotypes, namely, tumors with mutant or null P53 expression and tumors resistant to chemotherapy.
Collapse
Affiliation(s)
- Ana S Pires
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia R Marques
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - João C Encarnação
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana M Abrantes
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês A Marques
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Mafalda Laranjo
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rui Oliveira
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Department of Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - João E Casalta-Lopes
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana C Gonçalves
- Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Oncobiology and Hematology Laboratory, Applied Molecular Biology and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana B Sarmento-Ribeiro
- Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Oncobiology and Hematology Laboratory, Applied Molecular Biology and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Department of Hematology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria F Botelho
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
110
|
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: 2.1] [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]
|
111
|
Genome sequencing reveals a novel genetic mechanism underlying dihydropyrimidine dehydrogenase deficiency: A novel missense variant c.1700G>A and a large intragenic inversion in DPYD
spanning intron 8 to intron 12. Hum Mutat 2018; 39:947-953. [DOI: 10.1002/humu.23538] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/03/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022]
|
112
|
Tecza K, Pamula-Pilat J, Lanuszewska J, Butkiewicz D, Grzybowska E. Pharmacogenetics of toxicity of 5-fluorouracil, doxorubicin and cyclophosphamide chemotherapy in breast cancer patients. Oncotarget 2018; 9:9114-9136. [PMID: 29507678 PMCID: PMC5823653 DOI: 10.18632/oncotarget.24148] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/02/2018] [Indexed: 01/28/2023] Open
Abstract
The differences in patients' response to the same medication, toxicity included, are one of the major problems in breast cancer treatment. Chemotherapy toxicity makes a significant clinical problem due to decreased quality of life, prolongation of treatment and reinforcement of negative emotions associated with therapy. In this study we evaluated the genetic and clinical risk factors of FAC chemotherapy-related toxicities in the group of 324 breast cancer patients. Selected genes and their polymorphisms were involved in FAC drugs transport (ABCB1, ABCC2, ABCG2,SLC22A16), metabolism (ALDH3A1, CBR1, CYP1B1, CYP2C19, DPYD, GSTM1, GSTP1, GSTT1, MTHFR,TYMS), DNA damage recognition, repair and cell cycle control (ATM, ERCC1, ERCC2, TP53, XRCC1). The multifactorial risk models that combine genetic risk modifiers and clinical characteristics were constructed for 12 toxic symptoms. The majority of toxicities was dependent on the modifications in components of more than one pathway of FAC drugs, while the impact level of clinical factors was comparable to the genetic ones. For the carriers of multiple high risk factors the chance of developing given symptom was significantly elevated which proved the factor-dosage effect. We found the strongest associations between concurrent presence of clinical factors - overall and recurrent anemia, nephrotoxicity and early nausea and genetic polymorphisms in genes responsible for DNA repair, drugs metabolism and transport pathways. These results indicate the possibility of selection of the patients with expected high tolerance to FAC treatment and consequently with high chance of chemotherapy completion without the dose reduction, treatment delays and decline in the quality of life.
Collapse
Affiliation(s)
- Karolina Tecza
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Jolanta Pamula-Pilat
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Joanna Lanuszewska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Dorota Butkiewicz
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Ewa Grzybowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| |
Collapse
|
113
|
Lau MF, Vellasamy S, Chua KH, Sabaratnam V, Kuppusamy UR. Rosiglitazone diminishes the high-glucose-induced modulation of 5-fluorouracil cytotoxicity in colorectal cancer cells. EXCLI JOURNAL 2018; 17:186-199. [PMID: 29743857 PMCID: PMC5938530 DOI: 10.17179/excli2018-1011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the third most leading cause of morbidity and mortality throughout the world. 5-fluorouracil (5-FU), which is often administrated to disrupt carcinogenesis, was found to elevate blood glucose level among CRC patients. Thus, this study was conducted to evaluate the influence of rosiglitazone on antiproliferative effect of 5-FU using cellular model. Two human colonic carcinoma cell lines (HCT 116 and HT 29) were cultured in the presence of 5-FU, rosiglitazone or in combination under normal and high glucose concentration. The drug cytotoxicity was evaluated using the MTT assay whereas the assessment of cell cycle was carried out using the flow cytometry technique. Combination index (CI) method was used to determine the drug interaction between rosiglitazone and 5-FU. High glucose diminished the cytotoxic effect of 5-FU but at a high drug dosage, this effect could be overcome. Cell cycle analysis demonstrated that 5-FU and rosiglitazone caused G1-phase arrest and S-phase arrest, respectively. CI values indicated that rosiglitazone exerted synergistic effect on 5-FU regardless of glucose levels. This study is the first to demonstrate the influence of rosiglitazone on cytotoxicity of 5-FU under normal or high glucose level. Rosiglitazone may be a promising drug for enhancing the efficacy of 5-FU in the treatment of CRC associated with hyperglycemia.
Collapse
Affiliation(s)
- Meng-Fei Lau
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shalini Vellasamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Kek-Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Vikineswary Sabaratnam
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
114
|
Shrestha S, Zhang C, Jerde CR, Nie Q, Li H, Offer SM, Diasio RB. Gene-Specific Variant Classifier (DPYD-Varifier) to Identify Deleterious Alleles of Dihydropyrimidine Dehydrogenase. Clin Pharmacol Ther 2018; 104:709-718. [PMID: 29327356 DOI: 10.1002/cpt.1020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 11/10/2022]
Abstract
Deleterious variants in dihydropyrimidine dehydrogenase (DPD, DPYD gene) can be highly predictive of clinical toxicity to the widely prescribed chemotherapeutic 5-fluorouracil (5-FU). However, there are very limited data pertaining to the functional consequences of the >450 reported no-synonymous DPYD variants. We developed a DPYD-specific variant classifier (DPYD-Varifier) using machine learning and in vitro functional data for 156 missense DPYD variants. The developed model showed 85% accuracy and outperformed other in silico prediction tools. An examination of feature importance within the model provided additional insight into functional aspects of the DPD protein relevant to 5-FU toxicity. In the absence of clinical data for unstudied variants, prediction tools like DPYD-Varifier have great potential to individualize medicine and improve the clinical decision-making process.
Collapse
Affiliation(s)
- Shikshya Shrestha
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Calvin R Jerde
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Qian Nie
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic College of Medicine, Rochester, Minnesota, USA.,Mayo Clinic Center for Individualized Medicine, Minnesota, USA
| | - Steven M Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Robert B Diasio
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic College of Medicine, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Rochester, Minnesota, USA
| |
Collapse
|
115
|
Deenen MJ, Meulendijks D. Recommendation on testing for dihydropyrimidine dehydrogenase deficiency in the ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2018; 28:184. [PMID: 27701067 DOI: 10.1093/annonc/mdw533] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- M J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, The Netherlands.,Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - D Meulendijks
- Dutch Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands
| |
Collapse
|
116
|
Hydrogel-Based Stimuli-Responsive Functionalized Graft Copolymers for the Controlled Delivery of 5-Fluorouracil, an Anticancer Drug. GELS HORIZONS: FROM SCIENCE TO SMART MATERIALS 2018. [DOI: 10.1007/978-981-10-6077-9_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
117
|
Elander N, Aughton K, Greenhalf W. Development of Novel Therapeutic Response Biomarkers. PANCREATIC CANCER 2018:1273-1304. [DOI: 10.1007/978-1-4939-7193-0_59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
118
|
DPYD and UGT1A1 genotyping to predict adverse events during first-line FOLFIRI or FOLFOXIRI plus bevacizumab in metastatic colorectal cancer. Oncotarget 2017; 9:7859-7866. [PMID: 29487697 PMCID: PMC5814264 DOI: 10.18632/oncotarget.23559] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022] Open
Abstract
Our study addresses the issue of the clinical reliability of three candidate DPYD and one UGT single nucleotide polymorphisms in predicting 5-fluorouracil- and irinotecan-related adverse events. To this purpose, we took advantage of a large cohort of metastatic colorectal cancer patients treated with first-line 5-fluorouracil- and irinotecan-based chemotherapy regimens (i.e., FOLFIRI or FOLFOXIRI) plus bevacizumab in the randomized clinical trial TRIBE by GONO (clinicaltrials.gov: NCT00719797), in which adverse events were carefully and prospectively collected at each treatment cycle. Here we show that patients bearing DPYD c.1905+1G/A and c.2846A/T genotypes, together with UGT1A1*28 variant carriers, have an increased risk of experiencing clinically relevant toxicities, including hematological AEs and stomatitis. No carrier of the DPYD c.1679T>G minor allele was identified. Present results support the preemptive screening of mentioned DPYD and UGT1A1 variants to identify patients at risk of clinically relevant 5-fluoruracil- and irinotecan-related AEs, in order to improve treatments’ safety through a “genotype-guided” approach.
Collapse
|
119
|
Nakajima Y, Meijer J, Dobritzsch D, Ito T, Zhang C, Wang X, Watanabe Y, Tashiro K, Meinsma R, Roelofsen J, Zoetekouw L, van Kuilenburg ABP. Dihydropyrimidinase deficiency in four East Asian patients due to novel and rare DPYS mutations affecting protein structural integrity and catalytic activity. Mol Genet Metab 2017; 122:216-222. [PMID: 29054612 DOI: 10.1016/j.ymgme.2017.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 01/07/2023]
Abstract
Dihydropyrimidinase (DHP) is the second enzyme of the pyrimidine degradation pathway and catalyzes the ring opening of 5,6-dihydrouracil and 5,6-dihydrothymine. To date, only 31 genetically confirmed patients with a DHP deficiency have been reported and the clinical, biochemical and genetic spectrum of DHP deficient patients is, therefore, still largely unknown. Here, we show that 4 newly identified DHP deficient patients presented with strongly elevated levels of 5,6-dihydrouracil and 5,6-dihydrothymine in urine and a highly variable clinical presentation, ranging from asymptomatic to infantile spasm and reduced white matter and brain atrophy. Analysis of the DHP gene (DPYS) showed the presence of 8 variants including 4 novel/rare missense variants and one novel deletion. Functional analysis of recombinantly expressed DHP mutants carrying the p.M250I, p.H295R, p.Q334R, p.T418I and the p.R490H variant showed residual DHP activities of 2.0%, 9.8%, 9.7%, 64% and 0.3%, respectively. The crystal structure of human DHP indicated that all point mutations were likely to cause rearrangements of loops shaping the active site, primarily affecting substrate binding and stability of the enzyme. The observation that the identified mutations were more prevalent in East Asians and the Japanese population indicates that DHP deficiency may be more common than anticipated in these ethnic groups.
Collapse
Affiliation(s)
- Yoko Nakajima
- Fujita Health University School of Medicine, Department of Pediatrics, Toyoake 470-1192, Japan; Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands
| | - Judith Meijer
- Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands
| | - Doreen Dobritzsch
- Uppsala University, Department of Chemistry, Biomedical Center, S-751 24 Uppsala, Sweden
| | - Tetsuya Ito
- Fujita Health University School of Medicine, Department of Pediatrics, Toyoake 470-1192, Japan
| | - Chunhua Zhang
- MILS International, Department of Research and Development, Kanazawa 921-8105, Japan
| | - Xu Wang
- Beijing Children's Hospital, Capital University of Medical Sciences, Department of Neurology, Beijing 100045, China
| | - Yoriko Watanabe
- Kurume University, School of Medicine, Department of Pediatrics, Kurume 830-0011, Japan
| | - Kyoko Tashiro
- Kurume University, School of Medicine, Research Institute of Medical Mass Spectrometry, Kurume 830-0011, Japan
| | - Rutger Meinsma
- Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands
| | - Jeroen Roelofsen
- Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands
| | - Lida Zoetekouw
- Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands
| | - André B P van Kuilenburg
- Academic Medical Center, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, 1105, AZ, Amsterdam, The Netherlands.
| |
Collapse
|
120
|
Etienne-Grimaldi MC, Le Guellec CB, Boyer JC, Chatelut E, Evrard A, Loriot MA, Paci A, Royer B, Thomas F, Ciccolini J. Prevention of 5-fluorouracil-induced early severe toxicity by pre-therapeutic dihydropyrimidine dehydrogenase deficiency screening: The multiparametric approach is not convincing. Semin Oncol 2017; 44:159-160. [PMID: 28923215 DOI: 10.1053/j.seminoncol.2017.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/19/2017] [Indexed: 11/11/2022]
Affiliation(s)
| | | | | | - Etienne Chatelut
- Laboratoire de Biologie Médicale Oncologique, Institut Claudius-Regaud, CRCT, Toulouse, France
| | | | - Marie-Anne Loriot
- Biochemistry Department, Hopital Européen Georges Pompidou, Paris, France
| | - Angelo Paci
- Department of Clinical Pharmacy, SIPAM, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Bernard Royer
- Department of Clinical Pharmacology, CHU Jean Minjoz, Besançon, France
| | - Fabienne Thomas
- Laboratoire de Biologie Médicale Oncologique, Institut Claudius-Regaud, CRCT, Toulouse, France
| | - Joseph Ciccolini
- SMARTc, Department of Pharmacokinetics, CHU Timone, Marseille, France
| |
Collapse
|
121
|
P53 represses pyrimidine catabolic gene dihydropyrimidine dehydrogenase (DPYD) expression in response to thymidylate synthase (TS) targeting. Sci Rep 2017; 7:9711. [PMID: 28851987 PMCID: PMC5575263 DOI: 10.1038/s41598-017-09859-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/12/2017] [Indexed: 12/29/2022] Open
Abstract
Nucleotide metabolism in cancer cells can influence malignant behavior and intrinsic resistance to therapy. Here we describe p53-dependent control of the rate-limiting enzyme in the pyrimidine catabolic pathway, dihydropyrimidine dehydrogenase (DPYD) and its effect on pharmacokinetics of and response to 5-fluorouracil (5-FU). Using in silico/chromatin-immunoprecipitation (ChIP) analysis we identify a conserved p53 DNA-binding site (p53BS) downstream of the DPYD gene with increased p53 occupancy following 5-FU treatment of cells. Consequently, decrease in Histone H3K9AC and increase in H3K27me3 marks at the DPYD promoter are observed concomitantly with reduced expression of DPYD mRNA and protein in a p53-dependent manner. Mechanistic studies reveal inhibition of DPYD expression by p53 is augmented following thymidylate synthase (TS) inhibition and DPYD repression by p53 is dependent on DNA-dependent protein kinase (DNA-PK) and Ataxia telangiectasia mutated (ATM) signaling. In-vivo, liver specific Tp53 loss increases the conversion of 5-FU to 5-FUH2 in plasma and elicits a diminished 5-FU therapeutic response in a syngeneic colorectal tumor model consistent with increased DPYD-activity. Our data suggest that p53 plays an important role in controlling pyrimidine catabolism through repression of DPYD expression, following metabolic stress imposed by nucleotide imbalance. These findings have implications for the toxicity and efficacy of the cancer therapeutic 5-FU.
Collapse
|
122
|
Emmerich P, Clipson L, Deming DA. Resistance Mechanisms to Colorectal Cancer Therapeutics and the Clinical Implications. CURRENT COLORECTAL CANCER REPORTS 2017. [DOI: 10.1007/s11888-017-0374-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
123
|
Ludovini V, Antognelli C, Rulli A, Foglietta J, Pistola L, Eliana R, Floriani I, Nocentini G, Tofanetti FR, Piattoni S, Minenza E, Talesa VN, Sidoni A, Tonato M, Crinò L, Gori S. Influence of chemotherapeutic drug-related gene polymorphisms on toxicity and survival of early breast cancer patients receiving adjuvant chemotherapy. BMC Cancer 2017; 17:502. [PMID: 28747156 PMCID: PMC5530465 DOI: 10.1186/s12885-017-3483-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We investigated whether GSTT1 ("null" allele), GSTM1 ("null"allele), GSTP1 (A313G), RFC1 (G80A), MTHFR (C677T), TS (2R/3R) polymorphisms were associated with toxicity and survival in patients with early breast cancer (EBC) treated with adjuvant chemotherapy (CT). METHODS This prospective trial included patients with stage I-III BC subjected to CT with CMF or FEC regimens. PCR-RFLP was performed for MTHFR, RFC1 and GSTP1, while PCR for TS, GSTT1 and GSTM1 genes. RESULTS Among the 244 patients consecutively enrolled, 48.7% were treated with FEC and 51.3% with CMF. Patients with TS2R/3R genotype showed less frequently severe neutropenia (G3/G4) than those with TS2R/2R and 3R/3R genotype (p = 0.038). Patients with MTHFRCT genotype had a higher probability of developing severe neutropenia than those with MTHFR CC genotype (p = 0.043). Patients with RFC1GG or GSTT1-null genotype or their combination (GSTT1-null/RFC1GG) were significantly associated with a shorter disease free survival (DFS) (p = 0.009, p = 0.053, p = 0.003, respectively) and overall survival (OS) (p = 0.036, p = 0.015, p = 0.005, respectively). Multivariate analysis confirmed the association of RFC1GG genotype with a shorter DFS (p = 0.018) and of GSTT1-null genotype of a worse OS (p = 0.003), as well as for the combined genotypes GSTT1-null/RFC1GG, (DFS: p = 0.004 and OS: p = 0.003). CONCLUSIONS Our data suggest that TS2R/2R and 3R/3R or MTHFR CT genotypes have a potential role in identifying patients with greater risk of toxicity to CMF/FEC and that RFC1 GG and GSTT1-null genotypes alone or in combination could be important markers in predicting clinical outcome in EBC patients.
Collapse
Affiliation(s)
- Vienna Ludovini
- Medical Oncology Division, S. Maria della Misericordia Hospital, Azienda Ospedaliera of Perugia, Perugia, Italy
| | - Cinzia Antognelli
- Department of Experimental Medicine, University of Perugia, Piazzale Menghini 8/9, 06156 Perugia, Italy
| | - Antonio Rulli
- Breast Unit, Department of Surgical, University of Perugia, Perugia, Italy
| | - Jennifer Foglietta
- Medical Oncology Division, S. Maria della Misericordia Hospital, Azienda Ospedaliera of Perugia, Perugia, Italy
| | - Lorenza Pistola
- Medical Oncology Division, S. Maria della Misericordia Hospital, Azienda Ospedaliera of Perugia, Perugia, Italy
| | - Rulli Eliana
- Oncology Department, IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy
| | - Irene Floriani
- Oncology Department, IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy
| | - Giuseppe Nocentini
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Francesca Romana Tofanetti
- Medical Oncology Division, S. Maria della Misericordia Hospital, Azienda Ospedaliera of Perugia, Perugia, Italy
| | | | - Elisa Minenza
- Medical Oncology Division, “S. Maria” Hospital, Terni, Italy
| | - Vincenzo Nicola Talesa
- Department of Experimental Medicine, University of Perugia, Piazzale Menghini 8/9, 06156 Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, Section of Anatomic and Histology, Medical School, University of Perugia, Perugia, Italy
| | | | - Lucio Crinò
- Medical Oncology, Istituto Scientifico Romagnolo per lo studio e la cura dei tumori (IRST), IRCCS, Meldola, Italy
| | - Stefania Gori
- Medical Oncology, SacroCuore-Don Calabria Hospital, Negrar, Verona Italy
| |
Collapse
|
124
|
Pan W, Li Y, Feng Y, Yang F, Liu H. A new sample preparation and separation combination for the precise, accurate, and simultaneous determination of uracil and dihydrouracil in human plasma by reversed-phase HPLC. J Sep Sci 2017; 40:3763-3770. [PMID: 28726286 DOI: 10.1002/jssc.201700279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/23/2017] [Accepted: 07/13/2017] [Indexed: 11/09/2022]
Abstract
We have developed an efficient procedure and detection method using reversed-phase high-performance liquid chromatography for the simultaneous measurement of uracil and dihydrouracil in human plasma. The procedure, including chromatographic conditions and sample preparation, was optimized and validated. Optimization of the sample preparation included deproteinization, extraction, and cleanup. A new sample preparation method which resulted in an improved extraction yield of analytes and significantly reduced interference at low-wavelength UV detection was developed. The developed method was validated for specificity, linearity, limits of detection and quantitation, precision, and accuracy. All calibration curves showed excellent linear regression (R2 > 0.9990) within the testing range. The limit of detection for uracil and dihydrouracil was 2.5 and 5.0 ng/mL, respectively. The extraction yields were >94% for uracil and 91% for dihydrouracil. Intra- and interassay precision and accuracy for uracil and dihydrouracil were lower than 8% at all tested concentrations. The proposed method was successfully applied to measure plasma concentrations of uracil and dihydrouracil in colorectal cancer patients scheduled to receive fluoropyrimidine-based chemotherapy.
Collapse
Affiliation(s)
- Wen Pan
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yuandong Li
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yan Feng
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Fan Yang
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Haizhou Liu
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, P. R. China
| |
Collapse
|
125
|
Al-Khateeb M, Awidi A, Al-Hadidi K, Battah A. Low Incidence of the DPD IVS14+1G>A Polymorphism in Jordanian Breast and Colorectal Cancer patients. Asian Pac J Cancer Prev 2017; 18:1651-1654. [PMID: 28670884 PMCID: PMC6373818 DOI: 10.22034/apjcp.2017.18.6.1651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: Dihydropyrimidine dehydrogenase (DPD) is a crucial enzyme in the catabolism of 5-fluorouracil (5-FU), a drug that is frequently used in cancer therapy. Patients with deficient DPD activity are at risk of developing severe 5-FU–associated toxicity. One possible cause of deficiency is genetic polymorphisms in the DPD gene, such as IVS14+1G>A. Aim: The present study was conducted to screen for the IVS14+1G>A polymorphism in cancer patients receiving 5-FU and a control group. Methods: A total of 40 cancer patients (30 colorectal cancer (CRC) and 10 breast cancer patients) were enrolled in this study. One hundred healthy controls were also tested using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). DNA sequence analysis was carried out to confirm the presence of the IVSI14+1G>A polymorphism. Results: Only one CRC patient showed heterozygous IVS14+1G>A polymorphism in the DPD gene. Conclusion: The results of this study demonstrated a very low frequency of the IVS14+1G>A polymorphism among Jordanian patients with colorectal and breast cancer.
Collapse
Affiliation(s)
- Mohammad Al-Khateeb
- Thrombosis Haemostasis Laboratory, University of Jordan, Amman, Jordan.,Department of Pathology and Microbiology, Forensic Medicine, University of Jordan, Amman, Jordan.
| | | | | | | |
Collapse
|
126
|
Lu M, Lewis CM, Traylor M. Pharmacogenetic testing through the direct-to-consumer genetic testing company 23andMe. BMC Med Genomics 2017. [PMID: 28629370 PMCID: PMC5477417 DOI: 10.1186/s12920-017-0283-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Rapid advances in scientific research have led to an increase in public awareness of genetic testing and pharmacogenetics. Direct-to-consumer (DTC) genetic testing companies, such as 23andMe, allow consumers to access their genetic information directly through an online service without the involvement of healthcare professionals. Here, we evaluate the clinical relevance of pharmacogenetic tests reported by 23andMe in their UK tests. Methods
The research papers listed under each 23andMe report were evaluated, extracting information on effect size, sample size and ethnicity. A wider literature search was performed to provide a fuller assessment of the pharmacogenetic test and variants were matched to FDA recommendations. Additional evidence from CPIC guidelines, PharmGKB, and Dutch Pharmacogenetics Working Group was reviewed to determine current clinical practice. The value of the tests across ethnic groups was determined, including information on linkage disequilibrium between the tested SNP and causal pharmacogenetic variant, where relevant.
Results 23andMe offers 12 pharmacogenetic tests to their UK customers, some of which are in standard clinical practice, and others which are less widely applied. The clinical validity and clinical utility varies extensively between tests. The variants tested are likely to have different degrees of sensitivity due to different risk allele frequencies and linkage disequilibrium patterns across populations. The clinical relevance depends on the ethnicity of the individual and variability of pharmacogenetic markers. Further research is required to determine causal variants and provide more complete assessment of drug response and side effects. Conclusion 23andMe reports provide some useful pharmacogenetics information, mirroring clinical tests that are in standard use. Other tests are unspecific, providing limited guidance and may not be useful for patients without professional interpretation. Nevertheless, DTC companies like 23andMe act as a powerful intermediate step to integrate pharmacogenetic testing into clinical practice.
Collapse
Affiliation(s)
- Mengfei Lu
- Department of Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8EF, UK
| | - Matthew Traylor
- Department of Medical and Molecular Genetics, King's College London, 8th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
127
|
Hishinuma E, Akai F, Narita Y, Maekawa M, Yamaguchi H, Mano N, Oda A, Hirasawa N, Hiratsuka M. Functional characterization of 21 allelic variants of dihydropyrimidinase. Biochem Pharmacol 2017. [PMID: 28642038 DOI: 10.1016/j.bcp.2017.06.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dihydropyrimidinase (DHP, EC 3.5.2.2), encoded by the gene DPYS, is the second enzyme in the catabolic pathway of pyrimidine and of fluoropyrimidine drugs such as 5-fluorouracil, which are commonly used in anticancer treatment; DHP catalyzes the hydrolytic ring opening of dihydrouracil and dihydro-5-fluorouracil. DPYS mutations are known to contribute to interindividual variations in the toxicity of fluoropyrimidine drugs, but the functional characterization of DHP allelic variants remains inadequate. In this study, in vitro analysis was performed on 22 allelic variants of DHP by transiently expressing wild-type DHP and 21 DHP variants in 293FT cells and characterizing their enzymatic activities by using dihydrouracil and dihydro-5-fluorouracil as substrates. DHP expression levels and oligomeric forms were determined using immunoblotting and blue native PAGE, respectively, and the stability of the DHP variants was assessed by examining the proteins in variant-transfected cells treated with cycloheximide or bortezomib. Moreover, three kinetic parameters, Km, Vmax, and intrinsic clearance (Vmax/Km), for the hydrolysis of dihydrouracil and dihydro-5-fluorouracil were determined. We found that 5/21 variants showed significantly decreased intrinsic clearance as compared to wild-type DHP, and that 9/21 variants were expressed at low levels and were inactive due to proteasome-mediated degradation. The band patterns observed in the immunoblotting of blue native gels corresponded to DHP activity, and, notably, 18/21 DHP variants exhibited decreased or null enzymatic activity and these variants also showed a drastically reduced ability to form large oligomers. Thus, detection of DPYS genetic polymorphisms might facilitate the prediction severe adverse effects of fluoropyrimidine-based treatments.
Collapse
Affiliation(s)
- Eiji Hishinuma
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Fumika Akai
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yoko Narita
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Akifumi Oda
- Department of Biophysical Chemistry, Faculty of Pharmacy, Meijo University, Nagoya 468-8503, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
128
|
Etienne-Grimaldi MC, Boyer JC, Beroud C, Mbatchi L, van Kuilenburg A, Bobin-Dubigeon C, Thomas F, Chatelut E, Merlin JL, Pinguet F, Ferrand C, Meijer J, Evrard A, Llorca L, Romieu G, Follana P, Bachelot T, Chaigneau L, Pivot X, Dieras V, Largillier R, Mousseau M, Goncalves A, Roché H, Bonneterre J, Servent V, Dohollou N, Château Y, Chamorey E, Desvignes JP, Salgado D, Ferrero JM, Milano G. New advances in DPYD genotype and risk of severe toxicity under capecitabine. PLoS One 2017; 12:e0175998. [PMID: 28481884 PMCID: PMC5421769 DOI: 10.1371/journal.pone.0175998] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/04/2017] [Indexed: 12/29/2022] Open
Abstract
Background Deficiency in dihydropyrimidine dehydrogenase (DPD) enzyme is the main cause of severe and lethal fluoropyrimidine-related toxicity. Various approaches have been developed for DPD-deficiency screening, including DPYD genotyping and phenotyping. The goal of this prospective observational study was to perform exhaustive exome DPYD sequencing and to examine relationships between DPYD variants and toxicity in advanced breast cancer patients receiving capecitabine. Methods Two-hundred forty-three patients were analysed (88.5% capecitabine monotherapy). Grade 3 and grade 4 capecitabine-related digestive and/or neurologic and/or hemato-toxicities were observed in 10.3% and 2.1% of patients, respectively. DPYD exome, along with flanking intronic regions 3’UTR and 5’UTR, were sequenced on MiSeq Illumina. DPD phenotype was assessed by pre-treatment plasma uracil (U) and dihydrouracil (UH2) measurement. Results Among the 48 SNPs identified, 19 were located in coding regions, including 3 novel variations, each observed in a single patient (among which, F100L and A26T, both pathogenic in silico). Combined analysis of deleterious variants *2A, I560S (*13) and D949V showed significant association with grade 3–4 toxicity (sensitivity 16.7%, positive predictive value (PPV) 71.4%, relative risk (RR) 6.7, p<0.001) but not with grade 4 toxicity. Considering additional deleterious coding variants D342G, S492L, R592W and F100L increased the sensitivity to 26.7% for grade 3–4 toxicity (PPV 72.7%, RR 7.6, p<0.001), and was significantly associated with grade 4 toxicity (sensitivity 60%, PPV 27.3%, RR 31.4, p = 0.001), suggesting the clinical relevance of extended targeted DPYD genotyping. As compared to extended genotype, combining genotyping (7 variants) and phenotyping (U>16 ng/ml) did not substantially increase the sensitivity, while impairing PPV and RR. Conclusions Exploring an extended set of deleterious DPYD variants improves the performance of DPYD genotyping for predicting both grade 3–4 and grade 4 toxicities (digestive and/or neurologic and/or hematotoxicities) related to capecitabine, as compared to conventional genotyping restricted to consensual variants *2A, *13 and D949V.
Collapse
Affiliation(s)
| | | | - Christophe Beroud
- Aix-Marseille University, INSERM UMR S910, GMGF, Marseille, France
- APHM Hôpital Timone, Laboratoire de Génétique Moléculaire, Marseille, France
| | - Litaty Mbatchi
- Faculté de Pharmacie de Montpellier, Montpellier, France
| | - André van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam,The Netherlands
| | | | - Fabienne Thomas
- Institut Claudius-Regaud, CRCT, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Etienne Chatelut
- Institut Claudius-Regaud, CRCT, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Jean-Louis Merlin
- Institut de Cancérologie de Lorraine, UMR CNRS 7039 CRAN, Université de Lorraine, Nancy, France
| | | | | | - Judith Meijer
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam,The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | - Henri Roché
- Institut Claudius-Regaud, CRCT, Université de Toulouse, Inserm, UPS, Toulouse, France
| | | | | | | | | | | | | | - David Salgado
- Aix-Marseille University, INSERM UMR S910, GMGF, Marseille, France
| | | | | |
Collapse
|
129
|
Hajj A, Ghosn M, Mourad D, Hojaiban K, Mousallem P, Khabbaz LR. Lethal hepatotoxicity following 5-fluorouracil/cisplatin chemotherapy: a relevant case report. Per Med 2017; 14:197-201. [PMID: 29767581 DOI: 10.2217/pme-2016-0085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Some articles have reported severe toxicities induced by cisplatin/5-fluorouracil regimens, nevertheless, severe and lethal liver toxicity has not been previously reported. In this article, we report the case of a 72-year-old woman, who developed fulminant hepatitis, hypoglycemia and hypotension with atrial fibrillation not responding to treatment. After ruling out all other possible causes of hepatitis, the toxicity was more likely attributed to 5-fluorouracil. Genotyping was performed and the patient was found to be a homozygote carrier of the T variant of the MTHFR gene. The patient died two days later. Several factors, including genetic factors, could explain this severe toxicity. The present case discusses the importance of personalized medicine in oncology based on pharmacogenetic analysis of polymorphisms.
Collapse
Affiliation(s)
- Aline Hajj
- Laboratoire de Pharmacologie, Pharmacie clinique et Contrôle de Qualité des Médicaments, Pôle Technologie-Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Marwan Ghosn
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Diane Mourad
- Laboratoire de Pharmacologie, Pharmacie clinique et Contrôle de Qualité des Médicaments, Pôle Technologie-Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Khalil Hojaiban
- Laboratoire de Pharmacologie, Pharmacie clinique et Contrôle de Qualité des Médicaments, Pôle Technologie-Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Perla Mousallem
- Laboratoire de Pharmacologie, Pharmacie clinique et Contrôle de Qualité des Médicaments, Pôle Technologie-Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Lydia Rabbaa Khabbaz
- Laboratoire de Pharmacologie, Pharmacie clinique et Contrôle de Qualité des Médicaments, Pôle Technologie-Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| |
Collapse
|
130
|
Pretreatment serum uracil concentration as a predictor of severe and fatal fluoropyrimidine-associated toxicity. Br J Cancer 2017; 116:1415-1424. [PMID: 28427087 PMCID: PMC5520099 DOI: 10.1038/bjc.2017.94] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
Background: We investigated the predictive value of dihydropyrimidine dehydrogenase (DPD) phenotype, measured as pretreatment serum uracil and dihydrouracil concentrations, for severe as well as fatal fluoropyrimidine-associated toxicity in 550 patients treated previously with fluoropyrimidines during a prospective multicenter study. Methods: Pretreatment serum concentrations of uracil and dihydrouracil were measured using a validated LC-MS/MS method. The primary endpoint of this analysis was global (any) severe fluoropyrimidine-associated toxicity, that is, grade ⩾3 toxicity according to the NCI CTC-AE v3.0, occurring during the first cycle of treatment. The predictive value of uracil and the uracil/dihydrouracil ratio for early severe fluoropyrimidine-associated toxicity were compared. Pharmacogenetic variants in DPYD (c.2846A>T, c.1679T>G, c.1129-5923C>G, and c.1601G>A) and TYMS (TYMS 5′-UTR VNTR and TYMS 3′-UTR 6-bp ins/del) were measured and tested for associations with severe fluoropyrimidine-associated toxicity to compare predictive value with DPD phenotype. The Benjamini-Hochberg false discovery rate method was used to control for type I errors at level q<0.050 (corresponding to P<0.010). Results: Uracil was superior to the dihydrouracil/uracil ratio as a predictor of severe toxicity. High pretreatment uracil concentrations (>16 ng ml−1) were strongly associated with global severe toxicity (OR 5.3, P=0.009), severe gastrointestinal toxicity (OR 33.7, P<0.0001), toxicity-related hospitalisation (OR 16.9, P<0.0001), as well as fatal treatment-related toxicity (OR 44.8, P=0.001). None of the DPYD variants alone, or TYMS variants alone, were associated with severe toxicity. Conclusions: High pretreatment uracil concentration was strongly predictive of severe, including fatal, fluoropyrimidine-associated toxicity, and is a highly promising phenotypic marker to identify patients at risk of severe fluoropyrimidine-associated toxicity.
Collapse
|
131
|
Abstract
Cancer treatment is becoming more and more individually based as a result of the large inter-individual differences that exist in treatment outcome and toxicity when patients are treated using population-based drug doses. Polymorphisms in genes encoding drug-metabolizing enzymes and transporters can significantly influence uptake, metabolism, and elimination of anticancer drugs. As a result, the altered pharmacokinetics can greatly influence drug efficacy and toxicity. Pharmacogenetic screening and/or drug-specific phenotyping of cancer patients eligible for treatment with chemotherapeutic drugs, prior to the start of anticancer treatment, can identify patients with tumors that are likely to be responsive or resistant to the proposed drugs. Similarly, the identification of patients with an increased risk of developing toxicity would allow either dose adaptation or the application of other targeted therapies. This review focuses on the role of genetic polymorphisms significantly altering the pharmacokinetics of anticancer drugs. Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. For other drugs, however, the association of polymorphisms with pharmacokinetics is less clear. To date, the influence of genetic variations on the pharmacokinetics of the increasingly used monoclonal antibodies has hardly been investigated. Some studies indicate that genes encoding the Fcγ-receptor family are of interest, but more research is needed to establish if screening before the start of therapy is beneficial. Considering the profound impact of polymorphisms in drug transporters and drug-metabolizing enzymes on the pharmacokinetics of chemotherapeutic drugs and hence, their toxicity and efficacy, pharmacogenetic and pharmacokinetic profiling should become the standard of care.
Collapse
Affiliation(s)
| | | | - André B P van Kuilenburg
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Emma Children's Hospital, F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
132
|
Zanello P. The competition between chemistry and biology in assembling iron–sulfur derivatives. Molecular structures and electrochemistry. Part V. {[Fe4S4](SCysγ)4} proteins. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2016.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
133
|
van Kuilenburg ABP, Meijer J, Maurer D, Dobritzsch D, Meinsma R, Los M, Knegt LC, Zoetekouw L, Jansen RLH, Dezentjé V, van Huis-Tanja LH, van Kampen RJW, Hertz JM, Hennekam RCM. Severe fluoropyrimidine toxicity due to novel and rare DPYD missense mutations, deletion and genomic amplification affecting DPD activity and mRNA splicing. Biochim Biophys Acta Mol Basis Dis 2016; 1863:721-730. [PMID: 28024938 DOI: 10.1016/j.bbadis.2016.12.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022]
Abstract
Dihydropyrimidine dehydrogenase (DPD) is the initial and rate-limiting enzyme in the catabolism of 5-fluorouracil (5FU). Genetic variations in DPD have emerged as predictive risk factors for severe fluoropyrimidine toxicity. Here, we report novel and rare genetic variants underlying DPD deficiency in 9 cancer patients presenting with severe fluoropyrimidine-associated toxicity. All patients possessed a strongly reduced DPD activity, ranging from 9 to 53% of controls. Analysis of the DPD gene (DPYD) showed the presence of 21 variable sites including 4 novel and 4 very rare aberrations: 3 missense mutations, 2 splice-site mutations, 1 intronic mutation, a deletion of 21 nucleotides and a genomic amplification of exons 9-12. Two novel/rare variants (c.2843T>C, c.321+1G>A) were present in multiple, unrelated patients. Functional analysis of recombinantly-expressed DPD mutants carrying the p.I948T and p.G284V mutation showed residual DPD activities of 30% and 0.5%, respectively. Analysis of a DPD homology model indicated that the p.I948T and p.G284V mutations may affect electron transfer and the binding of FAD, respectively. cDNA analysis showed that the c.321+1G>A mutation in DPYD leads to skipping of exon 4 immediately upstream of the mutated splice-donor site in the process of DPD pre-mRNA splicing. A lethal toxicity in two DPD patients suggests that fluoropyrimidines combined with other therapies such as radiotherapy might be particularly toxic for DPD deficient patients. Our study advocates a more comprehensive genotyping approach combined with phenotyping strategies for upfront screening for DPD deficiency to ensure the safe administration of fluoropyrimidines.
Collapse
Affiliation(s)
- André B P van Kuilenburg
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands.
| | - Judith Meijer
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Dirk Maurer
- Uppsala University, Department of Chemistry, Biomedical Center, S-751 24 Uppsala, Sweden
| | - Doreen Dobritzsch
- Uppsala University, Department of Chemistry, Biomedical Center, S-751 24 Uppsala, Sweden
| | - Rutger Meinsma
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Maartje Los
- St. Antonius Hospital, Department of Oncology, Nieuwegein, The Netherlands
| | - Lia C Knegt
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Lida Zoetekouw
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Rob L H Jansen
- Maastricht University Medical Center, Department of Oncology, Maastricht, The Netherlands
| | - Vincent Dezentjé
- Reinier de Graaf Gasthuis, Department of Clinical Oncology, Delft, The Netherlands
| | | | - Roel J W van Kampen
- Zuyderland Medical Center, Department of Oncology, Sittard-Geleen, The Netherlands
| | - Jens Michael Hertz
- Odense University Hospital, Department of Clinical Genetics, Odense C, Denmark
| | - Raoul C M Hennekam
- Academic Medical Center, University of Amsterdam, Emma Children's Hospital, Departments of Clinical Chemistry, Pediatrics and Clinical Genetics, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
134
|
Alpha- and Beta-Cyclodextrin Inclusion Complexes with 5-Fluorouracil: Characterization and Cytotoxic Activity Evaluation. Molecules 2016; 21:molecules21121644. [PMID: 27916966 PMCID: PMC6273350 DOI: 10.3390/molecules21121644] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 11/24/2016] [Accepted: 11/25/2016] [Indexed: 11/16/2022] Open
Abstract
Cyclodextrins are natural macrocyclic oligosaccharides able to form inclusion complexes with a wide variety of guests, affecting their physicochemical and pharmaceutical properties. In order to obtain an improvement of the bioavailability and solubility of 5-fluorouracil, a pyrimidine analogue used as chemotherapeutic agent in the treatment of the colon, liver, and stomac cancers, the drug was complexed with alpha- and beta-cyclodextrin. The inclusion complexes were prepared in the solid state by kneading method and characterized by Fourier transform-infrared (FT-IR) spectroscopy and X-ray powder diffractometry. In solution, the 1:1 stoichiometry for all the inclusion complexes was established by the Job plot method and the binding constants were determined at different pHs by UV-VIS titration. Furthermore, the cytotoxic activity of 5-fluorouracil and its complexation products were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay on MCF-7 (breast cancer cell line), Hep G2 (hepatocyte carcinoma cell line), Caco-2 (colon adenocarcinoma cell line), and A-549 (alveolar basal epithelial carcinoma cell line). The results showed that both inclusion complexes increased the 5-fluorouracil capability of inhibiting cell growth. In particular, 5-fluorouracil complexed with beta-cyclodextrin had the highest cytotoxic activity on MCF-7; with alpha-cyclodextrin the highest cytotoxic activity was observed on A-549. The IC50 values were equal to 31 and 73 µM at 72 h, respectively. Our results underline the possibility of using these inclusion complexes in pharmaceutical formulations for improving 5-fluorouracil therapeutic efficacy.
Collapse
|
135
|
Rantalainen M, Klevebring D, Lindberg J, Ivansson E, Rosin G, Kis L, Celebioglu F, Fredriksson I, Czene K, Frisell J, Hartman J, Bergh J, Grönberg H. Sequencing-based breast cancer diagnostics as an alternative to routine biomarkers. Sci Rep 2016; 6:38037. [PMID: 27901097 PMCID: PMC5128815 DOI: 10.1038/srep38037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/25/2016] [Indexed: 12/20/2022] Open
Abstract
Sequencing-based breast cancer diagnostics have the potential to replace routine biomarkers and provide molecular characterization that enable personalized precision medicine. Here we investigate the concordance between sequencing-based and routine diagnostic biomarkers and to what extent tumor sequencing contributes clinically actionable information. We applied DNA- and RNA-sequencing to characterize tumors from 307 breast cancer patients with replication in up to 739 patients. We developed models to predict status of routine biomarkers (ER, HER2,Ki-67, histological grade) from sequencing data. Non-routine biomarkers, including mutations in BRCA1, BRCA2 and ERBB2(HER2), and additional clinically actionable somatic alterations were also investigated. Concordance with routine diagnostic biomarkers was high for ER status (AUC = 0.95;AUC(replication) = 0.97) and HER2 status (AUC = 0.97;AUC(replication) = 0.92). The transcriptomic grade model enabled classification of histological grade 1 and histological grade 3 tumors with high accuracy (AUC = 0.98;AUC(replication) = 0.94). Clinically actionable mutations in BRCA1, BRCA2 and ERBB2(HER2) were detected in 5.5% of patients, while 53% had genomic alterations matching ongoing or concluded breast cancer studies. Sequencing-based molecular profiling can be applied as an alternative to histopathology to determine ER and HER2 status, in addition to providing improved tumor grading and clinically actionable mutations and molecular subtypes. Our results suggest that sequencing-based breast cancer diagnostics in a near future can replace routine biomarkers.
Collapse
Affiliation(s)
- Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Klevebring
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Emma Ivansson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gustaf Rosin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lorand Kis
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Pathology and Cytology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Fuat Celebioglu
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - Irma Fredriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast- and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jan Frisell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast- and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Pathology and Cytology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Pathology and Cytology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
136
|
Evaluation of 5-fluorouracil degradation rate and Pharmacogenetic profiling to predict toxicity following adjuvant Capecitabine. Eur J Clin Pharmacol 2016; 73:157-164. [DOI: 10.1007/s00228-016-2160-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/07/2016] [Indexed: 12/17/2022]
|
137
|
Chen B, Solis-Villa C, Hakenberg J, Qiao W, Srinivasan RR, Yasuda M, Balwani M, Doheny D, Peter I, Chen R, Desnick RJ. Acute Intermittent Porphyria: Predicted Pathogenicity of HMBS Variants Indicates Extremely Low Penetrance of the Autosomal Dominant Disease. Hum Mutat 2016; 37:1215-1222. [PMID: 27539938 DOI: 10.1002/humu.23067] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/12/2016] [Indexed: 12/17/2022]
Abstract
Acute intermittent porphyria results from hydroxymethylbilane synthase (HMBS) mutations that markedly decrease HMBS enzymatic activity. This dominant disease is diagnosed when heterozygotes have life-threatening acute attacks, while most heterozygotes remain asymptomatic and undiagnosed. Although >400 HMBS mutations have been reported, the prevalence of pathogenic HMBS mutations in genomic/exomic databases, and the actual disease penetrance are unknown. Thus, we interrogated genomic/exomic databases, identified non-synonymous variants (NSVs) and consensus splice-site variants (CSSVs) in various demographic/racial groups, and determined the NSV's pathogenicity by prediction algorithms and in vitro expression assays. Caucasians had the most: 58 NSVs and two CSSVs among ∼92,000 alleles, a 0.00575 combined allele frequency. In silico algorithms predicted 14 out of 58 NSVs as "likely-pathogenic." In vitro expression identified 10 out of 58 NSVs as likely-pathogenic (seven predicted in silico), which together with two CSSVs had a combined allele frequency of 0.00056. Notably, six presumably pathogenic mutations/NSVs in the Human Gene Mutation Database were benign. Compared with the recent prevalence estimate of symptomatic European heterozygotes (∼0.000005), the prevalence of likely-pathogenic HMBS mutations among Caucasians was >100 times more frequent. Thus, the estimated penetrance of acute attacks was ∼1% of heterozygotes with likely-pathogenic mutations, highlighting the importance of predisposing/protective genes and environmental modifiers that precipitate/prevent the attacks.
Collapse
Affiliation(s)
- Brenden Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Constanza Solis-Villa
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Jörg Hakenberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Wanqiong Qiao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Ramakrishnan R Srinivasan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Manisha Balwani
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Dana Doheny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Rong Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York.
| |
Collapse
|
138
|
Meulendijks D, Cats A, Beijnen JH, Schellens JHM. Improving safety of fluoropyrimidine chemotherapy by individualizing treatment based on dihydropyrimidine dehydrogenase activity - Ready for clinical practice? Cancer Treat Rev 2016; 50:23-34. [PMID: 27589829 DOI: 10.1016/j.ctrv.2016.08.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 01/05/2023]
Abstract
Fluoropyrimidines remain the cornerstone of treatment for different types of cancer, and are used by an estimated two million patients annually. The toxicity associated with fluoropyrimidine therapy is substantial, however, and affects around 30% of the patients, with 0.5-1% suffering fatal toxicity. Activity of the main 5-fluorouracil (5-FU) metabolic enzyme, dihydropyrimidine dehydrogenase (DPD), is the key determinant of 5-FU pharmacology, and accounts for around 80% of 5-FU catabolism. There is a consistent relationship between DPD activity and 5-FU exposure on the one hand, and risk of severe and potentially lethal fluoropyrimidine-associated toxicity on the other hand. Therefore, there is a sound rationale for individualizing treatment with fluoropyrimidines based on DPD status in order to improve patient safety. The field of individualized treatment with fluoropyrimidines is now rapidly developing. The main strategies that are available, are based on genotyping of the gene encoding DPD (DPYD) and measuring of pretreatment DPD phenotype. Clinical validity of additional approaches, including genotyping of MIR27A has also recently been demonstrated. Here, we critically review the evidence on clinical validity and utility of strategies available to clinicians to identify patients at risk of developing severe and potentially fatal toxicity as a result of DPD deficiency. We evaluate the advantages and limitations of these methods when used in clinical practice, and discuss for which strategies clinical implementation is currently justified based on the available evidence and, in addition, which additional data will be required before implementing other, as yet less developed strategies.
Collapse
Affiliation(s)
- Didier Meulendijks
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Dutch Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands.
| | - Annemieke Cats
- Department of Gastroenterology & Hepatology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Faculty of Science, Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Faculty of Science, Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
139
|
Latchman J, Guastella A, Tofthagen C. 5-Fluorouracil toxicity and dihydropyrimidine dehydrogenase enzyme: implications for practice. Clin J Oncol Nurs 2016; 18:581-5. [PMID: 25253112 DOI: 10.1188/14.cjon.581-585] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
5-fluorouracil (5-FU) is a fluorinated pyrimidine analog, which is commonly used in combination chemotherapy for treating solid tumors. Dihydropyrimidine dehydrogenase plays an important role in catabolism and clearance of 5-FU. Any alteration in that sequence of enzymatic activity can lead to toxicity and even death in some patients. The most common loss of a functional allele of the dihydropyrimidine gene is the splice-site mutation c.1905+1G>A, which leads to deficiency of the enzyme. However, because of the small percentage of the population in which the deficiency occurs, routine screening is not recommended, and commercial testing is costly. Treatment measures for 5-FU toxicity are mainly supportive, including palliation of symptoms. Good patient assessment and education are imperative to early treatment of 5-FU-induced toxicity. Advanced oncology practitioners and oncology nurses should thoroughly educate patients and their caregivers on both the common and adverse side effects of 5-FU-based therapy and when it may be necessary to immediately contact their healthcare provider.
Collapse
Affiliation(s)
| | - Ann Guastella
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | |
Collapse
|
140
|
Ison G, Beaver JA, McGuinn WD, Palmby TR, Dinin J, Charlab R, Marathe A, Jin R, Liu Q, Chen XH, Ysern X, Stephens O, Bai G, Wang Y, Dorff SE, Cheng J, Tang S, Sridhara R, Pierce W, McKee AE, Ibrahim A, Kim G, Pazdur R. FDA Approval: Uridine Triacetate for the Treatment of Patients Following Fluorouracil or Capecitabine Overdose or Exhibiting Early-Onset Severe Toxicities Following Administration of These Drugs. Clin Cancer Res 2016; 22:4545-9. [PMID: 27401247 DOI: 10.1158/1078-0432.ccr-16-0638] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/10/2016] [Indexed: 01/24/2023]
Abstract
On December 11, 2015, the FDA approved uridine triacetate (VISTOGARD; Wellstat Therapeutics Corporation) for the emergency treatment of adult and pediatric patients following a fluorouracil or capecitabine overdose regardless of the presence of symptoms, and of those who exhibit early-onset, severe, or life-threatening toxicity affecting the cardiac or central nervous system, and/or early onset, unusually severe adverse reactions (e.g., gastrointestinal toxicity and/or neutropenia) within 96 hours following the end of fluorouracil or capecitabine administration. Uridine triacetate is not recommended for the nonemergent treatment of adverse reactions associated with fluorouracil or capecitabine because it may diminish the efficacy of these drugs, and the safety and efficacy of uridine triacetate initiated more than 96 hours following the end of administration of these drugs has not been established. The approval is based on data from two single-arm, open-label, expanded-access trials in 135 patients receiving uridine triacetate (10 g or 6.2 g/m(2) orally every 6 hours for 20 doses) for fluorouracil or capecitabine overdose, or who exhibited severe or life-threatening toxicities within 96 hours following the end of fluorouracil or capecitabine administration. Ninety-six percent of patients met the major efficacy outcome measure, which was survival at 30 days or survival until the resumption of chemotherapy, if prior to 30 days. The most common adverse reactions were vomiting, nausea, and diarrhea. This article summarizes the FDA review of this New Drug Application, the data supporting approval of uridine triacetate, and the unique regulatory situations encountered by this approval. Clin Cancer Res; 22(18); 4545-49. ©2016 AACR.
Collapse
Affiliation(s)
- Gwynn Ison
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland.
| | - Julia A Beaver
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - W David McGuinn
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Todd R Palmby
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Jeannette Dinin
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Rosane Charlab
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Anshu Marathe
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Runyan Jin
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Qi Liu
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Xiao Hong Chen
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Xavier Ysern
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Olen Stephens
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Ge Bai
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Yaning Wang
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Sarah E Dorff
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Joyce Cheng
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Shenghui Tang
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Rajeshwari Sridhara
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - William Pierce
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Amy E McKee
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Amna Ibrahim
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Geoffrey Kim
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Richard Pazdur
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| |
Collapse
|
141
|
Negrei C, Hudita A, Ginghina O, Galateanu B, Voicu SN, Stan M, Costache M, Fenga C, Drakoulis N, Tsatsakis AM. Colon Cancer Cells Gene Expression Signature As Response to 5- Fluorouracil, Oxaliplatin, and Folinic Acid Treatment. Front Pharmacol 2016; 7:172. [PMID: 27445811 PMCID: PMC4917556 DOI: 10.3389/fphar.2016.00172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/03/2016] [Indexed: 12/05/2022] Open
Abstract
5-FU cytotoxicity mechanism has been assigned both to the miss-incorporation of fluoronucleotides into RNA and DNA and to the inhibition of thymidylate synthase. 5-FU is one of the most widely used chemotherapeutic drugs, although it has severe side effects that may vary between patients. Pharmacogenetic studies related to 5-FU have been traditionally focused on the rate-limiting catabolic enzyme, dihydropyrimidine dehydrogenase that breaks 80–85% of 5-FU into its inactive metabolite. Choosing the right dosing scheme and chemotherapy strategy for each individual patient remains challenging for personalized chemotherapy management. In the general effort toward reduction of colorectal cancer mortality, in vitro screening studies play a very important role. To accelerate translation research, increasing interest has been focused on using in vivo-like models such as three-dimensional spheroids. The development of higher throughput assays to quantify phenotypic changes in spheroids is an active research area. Consequently, in this study we used the microarray technology to reveal the HT-29 colorectal adenocarcinoma cells gene expression signature as response to 5-FU/OXP/FA treatment in a state of the art 3D culture system. We report here an increased reactive oxygen species production under treatment, correlated with a decrease in cell viability and proliferation potential. With respect to the HT-29 cells gene expression under the treatment with 5-FU/OXP/FA, we found 15.247 genes that were significantly differentially expressed (p < 0.05) with a fold change higher that two-fold. Among these, 7136 genes were upregulated and 8111 genes were downregulated under experimental conditions as compared to untreated cells. The most relevant and statistic significant (p < 0.01) pathways in the experiment are associated with the genes that displayed significant differential expression and are related to intracellular signaling, oxidative stress, apoptosis, and cancer.
Collapse
Affiliation(s)
- Carolina Negrei
- Department of Toxicology, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy Bucharest, Romania
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Octav Ginghina
- Department of Surgery, "Sf. Ioan" Clinical Emergency HospitalBucharest, Romania; Department II, Faculty of Dental Medicine, "Carol Davila" University of Medicine and PharmacyBucharest, Romania
| | - Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Sorina Nicoleta Voicu
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Miriana Stan
- Department of Toxicology, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest Bucharest, Romania
| | - Concettina Fenga
- Occupational Medicine Section, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina Messina, Italy
| | - Nikolaos Drakoulis
- Research Laboratory of Clinical Pharmacology and Pharmacogenomics, School of Health Sciences, Faculty of Pharmacy, National and Kapodistrian University of Athens Athens, Greece
| | - Aristidis M Tsatsakis
- Department of Toxicology and Forensic Sciences, Medical School, University of Crete Heraklion, Greece
| |
Collapse
|
142
|
Ciccolini J, Serdjebi C, Le Thi Thu H, Lacarelle B, Milano G, Fanciullino R. Nucleoside analogs: ready to enter the era of precision medicine? Expert Opin Drug Metab Toxicol 2016; 12:865-77. [DOI: 10.1080/17425255.2016.1192128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Joseph Ciccolini
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Cindy Serdjebi
- Assistance Publique Hôpitaux de Marseille. Multidisciplinary Oncology & Therapeutic Innovations dpt, Aix Marseille University, Marseille, France
| | - Hau Le Thi Thu
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Bruno Lacarelle
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Gerard Milano
- Oncopharmacology Unit, Centre Antoine Lacassagne, Nice, France
| | | |
Collapse
|
143
|
Pharmacokinetically guided algorithm of 5-fluorouracil dosing, a reliable strategy of precision chemotherapy for solid tumors: a meta-analysis. Sci Rep 2016; 6:25913. [PMID: 27229175 PMCID: PMC4882511 DOI: 10.1038/srep25913] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/22/2016] [Indexed: 12/17/2022] Open
Abstract
Precision medicine characterizes a new era of cancer care and provides each patient with the right drug at the right dose and time. However, the practice of precision dosing is hampered by a lack of smart dosing algorithms. A pharmacokinetically guided (PKG) dosing algorithm is considered to be the leading strategy for precision chemotherapy, although the effects of PKG dosing are not completely confirmed. Hence, we conducted a meta-analysis to evaluate the effects of the PKG algorithm of 5-fluorouracil (5-FU) dosing on patients with solid tumors. A comprehensive retrieval was performed to identify all of the prospective controlled studies that compared the body surface area (BSA)-based algorithm with the PKG algorithm of 5-FU in patients with solid tumors. Overall, four studies with 504 patients were included. The PKG algorithm significantly improved the objective response rate of 5-FU-based chemotherapy compared with the BSA-based algorithm. Furthermore, PKG dosing markedly decreased the risk of total grade 3/4 adverse drug reactions, especially those related to hematological toxicity. Overall, the PKG algorithm may serve as a reliable strategy for individualized dosing of 5-FU.
Collapse
|
144
|
Dhelens C, Bonadona A, Thomas F, Chapuis C, Potton L, Marsili S, Bedouch P, Schwebel C. Lethal 5-fluorouracil toxicity in a colorectal patient with severe dihydropyrimidine dehydrogenase (DPD) deficiency. Int J Colorectal Dis 2016; 31:699-701. [PMID: 25796495 DOI: 10.1007/s00384-015-2191-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Carole Dhelens
- Department of Pharmacy, Centre Hospitalier Universitaire de Grenoble, Grenoble, France. .,CHU Grenoble, Pôle Pharmacie, Pharmacie clinique, Pavillon Vercors, CS 10217, 38043, Grenoble cedex 9, France.
| | - Agnès Bonadona
- Department of Medical Intensive Care Unit, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Fabienne Thomas
- Department of Pharmacology, IUCT-O, Institut Claudius Regaud, Toulouse, France.,University Toulouse III Paul Sabatier/EA4553, Toulouse, France
| | - Claire Chapuis
- Department of Pharmacy, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Leila Potton
- Department of Medical Intensive Care Unit, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Sabrina Marsili
- Department of Pharmacology, IUCT-O, Institut Claudius Regaud, Toulouse, France.,University Toulouse III Paul Sabatier/EA4553, Toulouse, France
| | - Pierrick Bedouch
- Department of Pharmacy, Centre Hospitalier Universitaire de Grenoble, Grenoble, France.,Grenoble-Alpes University UJF-Grenoble 1/CNRS/TIMC-IMAG, UMR 5525/Themas, Grenoble, France
| | - Carole Schwebel
- Department of Medical Intensive Care Unit, Centre Hospitalier Universitaire de Grenoble, Grenoble, France.,Grenoble-Alpes University UJF Grenoble 1/Inserm U1039, Grenoble, France
| |
Collapse
|
145
|
Meulendijks D, van Hasselt JC, Huitema AD, van Tinteren H, Deenen MJ, Beijnen JH, Cats A, Schellens JH. Renal function, body surface area, and age are associated with risk of early-onset fluoropyrimidine-associated toxicity in patients treated with capecitabine-based anticancer regimens in daily clinical care. Eur J Cancer 2016; 54:120-130. [DOI: 10.1016/j.ejca.2015.10.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/11/2015] [Accepted: 10/18/2015] [Indexed: 12/27/2022]
|
146
|
Kuilenburg ABPV, Meijer J, Tanck MWT, Dobritzsch D, Zoetekouw L, Dekkers LL, Roelofsen J, Meinsma R, Wymenga M, Kulik W, Büchel B, Hennekam RCM, Largiadèr CR. Phenotypic and clinical implications of variants in the dihydropyrimidine dehydrogenase gene. Biochim Biophys Acta Mol Basis Dis 2016; 1862:754-762. [PMID: 26804652 DOI: 10.1016/j.bbadis.2016.01.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/21/2015] [Accepted: 01/08/2016] [Indexed: 12/22/2022]
Abstract
Dihydropyrimidine dehydrogenase (DPD) is the initial and rate-limiting enzyme in the catabolism of the pyrimidine bases uracil, thymine and the antineoplastic agent 5-fluorouracil. Genetic variations in the gene encoding DPD (DPYD) have emerged as predictive risk alleles for 5FU-associated toxicity. Here we report an in-depth analysis of genetic variants in DPYD and their consequences for DPD activity and pyrimidine metabolites in 100 Dutch healthy volunteers. 34 SNPs were detected in DPYD and 15 SNPs were associated with altered plasma concentrations of pyrimidine metabolites. DPD activity was significantly associated with the plasma concentrations of uracil, the presence of a specific DPYD mutation (c.1905+1G>A) and the combined presence of three risk variants in DPYD (c.1905+1G>A, c.1129-5923C>G, c.2846A>T), but not with an altered uracil/dihydrouracil (U/UH2) ratio. Various haplotypes were associated with different DPD activities (haplotype D3, a decreased DPD activity; haplotype F2, an increased DPD activity). Functional analysis of eight recombinant mutant DPD enzymes showed a reduced DPD activity, ranging from 35% to 84% of the wild-type enzyme. Analysis of a DPD homology model indicated that the structural effect of the novel p.G401R mutation is most likely minor. The clinical relevance of the p.D949V mutation was demonstrated in a cancer patient heterozygous for the c.2846A>T mutation and a novel nonsense mutation c.1681C>T (p.R561X), experiencing severe grade IV toxicity. Our studies showed that the endogenous levels of uracil and the U/UH2 ratio are poor predictors of an impaired DPD activity. Loading studies with uracil to identify patients with a DPD deficiency warrants further investigation.
Collapse
Affiliation(s)
- André B P van Kuilenburg
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands.
| | - Judith Meijer
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael W T Tanck
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Doreen Dobritzsch
- Department of Chemistry, Biomedical Center, Uppsala University, S-751 24 Uppsala, Sweden
| | - Lida Zoetekouw
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Jeroen Roelofsen
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Rutger Meinsma
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Machteld Wymenga
- Department of Oncology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Wim Kulik
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Barbara Büchel
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raoul C M Hennekam
- Departments of Clinical Chemistry, Pediatrics, Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlo R Largiadèr
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
147
|
Capecitabine-Induced Severe Toxicity Secondary to DPD Deficiency and Successful Treatment with Low Dose 5-Fluorouracil. J Gastrointest Cancer 2016; 48:66-69. [PMID: 26744322 DOI: 10.1007/s12029-015-9797-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
148
|
Duley JA, Ni M, Shannon C, Norris RL, Sheffield L, Harris M, van Kuilenburg AB, Mead S, Cameron A, Helsby N, George R, Charles BG. Towards a test to predict 5-fluorouracil toxicity: Pharmacokinetic data for thymine and two sequential metabolites following oral thymine administration to healthy adult males. Eur J Pharm Sci 2016; 81:36-41. [DOI: 10.1016/j.ejps.2015.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/25/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023]
|
149
|
Shimamoto Y, Nukatsuka M, Takechi T, Fukushima M. Association between mRNA expression of chemotherapy-related genes and clinicopathological features in colorectal cancer: A large-scale population analysis. Int J Mol Med 2015; 37:319-28. [PMID: 26676887 PMCID: PMC4716796 DOI: 10.3892/ijmm.2015.2427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022] Open
Abstract
To establish the individualized treatment of patients with colorectal cancer, factors associated with chemotherapeutic effects should be identified. However, to the best of our knowledge, few studies are available on this topic, although it is known that the prognosis of patients and sensitivity to chemotherapy depend on the location of the tumor and that the tumor location is important for individualized treatment. In this study, primary tumors obtained from 1,129 patients with colorectal cancer were used to measure the mRNA expression levels of the following genes associated with the effects of standard chemotherapy for colorectal cancer: 5-fluorouracil (5-FU)-related thymidylate synthase (TYMS), dihydropyrimidine dehydrogenase (DPYD) and thymidine phosphorylase (TYMP); folate-related dihydrofolate reductase (DHFR), folylpolyglutamate synthase (FPGS) and gamma-glutamyl hydrolase (GGH); irinotecan-related topoisomerase I (TOP1); oxaliplatin-related excision repair cross-complementing 1 (ERCC1); biologic agent-related vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR). Large-scale population analysis was performed to determine the association of gene expression with the clinicopathological features, in particular, the location of the colorectal cancer. From the results of our analysis of the mRNA expression of these 10 genes, we noted the strongest correlation between DPYD and TYMP, followed by TYMS and DHFR. The location of the colorectal cancer was classified into 4 regions (the right- and left-sided colon, rectosigmoid and rectum) and was compared with gene expression. A significant difference in all genes, apart from VEGF, was noted. Of the remaining 9 genes, the highest expression of TYMS and DPYD was observed in the right-sided colon; the highest expression of GGH and EGFR was noted in the left-sided colon; the highest expression of DHFR, FPGS, TOP1 and ERCC1 was noted in the rectosigmoid, whereas TYMP expression was approximately equivalent in the right-sided colon and rectum, and higher than that in other locations. The data generated from this study may prove to be useful for the development of individualized chemotherapeutic treatments for patients with colorectal cancer, and will mean that the tumor location is taken into account.
Collapse
Affiliation(s)
- Yuji Shimamoto
- Translational Research Laboratory, Taiho Pharmaceutical Co., Ltd., Uchikanda, Chiyoda-ku, Tokyo 101-0047, Japan
| | - Mamoru Nukatsuka
- Translational Research Laboratory, Taiho Pharmaceutical Co., Ltd., Kawauchi-Cho, Tokushima‑shi, Tokushima 771-0194, Japan
| | - Teiji Takechi
- Translational Research Laboratory, Taiho Pharmaceutical Co., Ltd., Uchikanda, Chiyoda-ku, Tokyo 101-0047, Japan
| | - Masakazu Fukushima
- The Japan-Multinational Trial Organization, Naka-ku, Nagoya-shi, Aichi 460-0002, Japan
| |
Collapse
|
150
|
Meulendijks D, Henricks LM, Sonke GS, Deenen MJ, Froehlich TK, Amstutz U, Largiadèr CR, Jennings BA, Marinaki AM, Sanderson JD, Kleibl Z, Kleiblova P, Schwab M, Zanger UM, Palles C, Tomlinson I, Gross E, van Kuilenburg ABP, Punt CJA, Koopman M, Beijnen JH, Cats A, Schellens JHM. Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data. Lancet Oncol 2015; 16:1639-50. [PMID: 26603945 DOI: 10.1016/s1470-2045(15)00286-7] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/26/2015] [Accepted: 08/28/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND The best-known cause of intolerance to fluoropyrimidines is dihydropyrimidine dehydrogenase (DPD) deficiency, which can result from deleterious polymorphisms in the gene encoding DPD (DPYD), including DPYD*2A and c.2846A>T. Three other variants-DPYD c.1679T>G, c.1236G>A/HapB3, and c.1601G>A-have been associated with DPD deficiency, but no definitive evidence for the clinical validity of these variants is available. The primary objective of this systematic review and meta-analysis was to assess the clinical validity of c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity. METHODS We did a systematic review of the literature published before Dec 17, 2014, to identify cohort studies investigating associations between DPYD c.1679T>G, c.1236G>A/HapB3, and c.1601G>A and severe (grade ≥3) fluoropyrimidine-associated toxicity in patients treated with fluoropyrimidines (fluorouracil, capecitabine, or tegafur-uracil as single agents, in combination with other anticancer drugs, or with radiotherapy). Individual patient data were retrieved and analysed in a multivariable analysis to obtain an adjusted relative risk (RR). Effect estimates were pooled by use of a random-effects meta-analysis. The threshold for significance was set at a p value of less than 0·0167 (Bonferroni correction). FINDINGS 7365 patients from eight studies were included in the meta-analysis. DPYD c.1679T>G was significantly associated with fluoropyrimidine-associated toxicity (adjusted RR 4·40, 95% CI 2·08-9·30, p<0·0001), as was c.1236G>A/HapB3 (1·59, 1·29-1·97, p<0·0001). The association between c.1601G>A and fluoropyrimidine-associated toxicity was not significant (adjusted RR 1·52, 95% CI 0·86-2·70, p=0·15). Analysis of individual types of toxicity showed consistent associations of c.1679T>G and c.1236G>A/HapB3 with gastrointestinal toxicity (adjusted RR 5·72, 95% CI 1·40-23·33, p=0·015; and 2·04, 1·49-2·78, p<0·0001, respectively) and haematological toxicity (adjusted RR 9·76, 95% CI 3·03-31·48, p=0·00014; and 2·07, 1·17-3·68, p=0·013, respectively), but not with hand-foot syndrome. DPYD*2A and c.2846A>T were also significantly associated with severe fluoropyrimidine-associated toxicity (adjusted RR 2·85, 95% CI 1·75-4·62, p<0·0001; and 3·02, 2·22-4·10, p<0·0001, respectively). INTERPRETATION DPYD variants c.1679T>G and c.1236G>A/HapB3 are clinically relevant predictors of fluoropyrimidine-associated toxicity. Upfront screening for these variants, in addition to the established variants DPYD*2A and c.2846A>T, is recommended to improve the safety of patients with cancer treated with fluoropyrimidines. FUNDING None.
Collapse
Affiliation(s)
- Didier Meulendijks
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Linda M Henricks
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gabe S Sonke
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tanja K Froehlich
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Ursula Amstutz
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Carlo R Largiadèr
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | | | | | | | - Zdenek Kleibl
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Petra Kleiblova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; Department of Clinical Pharmacology, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Claire Palles
- Molecular and Population Genetics Laboratory and Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory and Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Eva Gross
- Department of Gynecology and Obstetrics, Technische Universität München, Munich, Germany
| | - André B P van Kuilenburg
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Annemieke Cats
- Department of Gastroenterology and Hepatology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|