201
|
Standard fluoropyrimidine dosages in chemoradiation therapy result in an increased risk of severe toxicity in DPYD variant allele carriers. Eur J Cancer 2018; 104:210-218. [PMID: 30361102 DOI: 10.1016/j.ejca.2018.07.138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prospective DPYD genotyping prevents severe fluoropyrimidine (FP)-induced toxicity by decreasing dosages in DPYD variant allele carriers. FP dosages in chemoradiation therapy (CRT) are lower than those in other FP-containing regimens. Pharmacogenetic guidelines do not distinguish between regimens, leaving physicians in doubt to apply dose reductions. Our aim was to investigate severe toxicity in DPYD variant allele carriers receiving CRT. METHODS Medical records of 828 patients who received FP-based CRT were reviewed from three centres. Severe (grade ≥III) toxicity in DPYD variant allele carriers receiving upfront FP dose reductions according to pharmacogenetic dosing guidelines and DPYD variant allele carriers not receiving FP dose reductions was compared with DPYD wild-type patients receiving standard dose of FPs in CRT. RESULTS DPYD variant allele carriers treated with standard dosages (N = 34) showed an increased risk of severe gastrointestinal (adjusted OR = 2.58, confidence interval [CI] = 1.02-6.53, P = 0.045) or severe haematological (adjusted OR = 4.19, CI = 1.32-13.25, P = 0.015) toxicity compared with wild-type patients (N = 771). DPYD variant allele carriers who received dose reductions (N = 22) showed a comparable frequency of severe gastrointestinal toxicity compared with wild-type patients, but more (not statistically significant) severe haematological toxicity. Hospitalisations for all DPYD variant allele carriers were comparable, independent of dose adjustments; however, the mean duration of hospitalisation was significantly shorter in the dose reduction group (P = 0.010). CONCLUSIONS Standard FP dosages in CRT resulted in an increased risk of severe toxicity in DPYD variant allele carriers. We advise to apply FP dose reductions according to current guidelines in DPYD variant allele carriers starting CRT.
Collapse
|
202
|
Amstutz U, Largiadèr CR. Genotype-guided fluoropyrimidine dosing: ready for implementation. Lancet Oncol 2018; 19:1421-1422. [PMID: 30348539 DOI: 10.1016/s1470-2045(18)30744-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Ursula Amstutz
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Bern 3010, Switzerland.
| | - Carlo R Largiadèr
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Bern 3010, Switzerland
| |
Collapse
|
203
|
Henricks LM, Lunenburg CATC, de Man FM, Meulendijks D, Frederix GWJ, Kienhuis E, Creemers GJ, Baars A, Dezentjé VO, Imholz ALT, Jeurissen FJF, Portielje JEA, Jansen RLH, Hamberg P, Ten Tije AJ, Droogendijk HJ, Koopman M, Nieboer P, van de Poel MHW, Mandigers CMPW, Rosing H, Beijnen JH, Werkhoven EV, van Kuilenburg ABP, van Schaik RHN, Mathijssen RHJ, Swen JJ, Gelderblom H, Cats A, Guchelaar HJ, Schellens JHM. DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 2018; 19:1459-1467. [PMID: 30348537 DOI: 10.1016/s1470-2045(18)30686-7] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Fluoropyrimidine treatment can result in severe toxicity in up to 30% of patients and is often the result of reduced activity of the key metabolic enzyme dihydropyrimidine dehydrogenase (DPD), mostly caused by genetic variants in the gene encoding DPD (DPYD). We assessed the effect of prospective screening for the four most relevant DPYD variants (DPYD*2A [rs3918290, c.1905+1G>A, IVS14+1G>A], c.2846A>T [rs67376798, D949V], c.1679T>G [rs55886062, DPYD*13, I560S], and c.1236G>A [rs56038477, E412E, in haplotype B3]) on patient safety and subsequent DPYD genotype-guided dose individualisation in daily clinical care. METHODS In this prospective, multicentre, safety analysis in 17 hospitals in the Netherlands, the study population consisted of adult patients (≥18 years) with cancer who were intended to start on a fluoropyrimidine-based anticancer therapy (capecitabine or fluorouracil as single agent or in combination with other chemotherapeutic agents or radiotherapy). Patients with all tumour types for which fluoropyrimidine-based therapy was considered in their best interest were eligible. We did prospective genotyping for DPYD*2A, c.2846A>T, c.1679T>G, and c.1236G>A. Heterozygous DPYD variant allele carriers received an initial dose reduction of 25% (c.2846A>T and c.1236G>A) or 50% (DPYD*2A and c.1679T>G), and DPYD wild-type patients were treated according to the current standard of care. The primary endpoint of the study was the frequency of severe (National Cancer Institute Common Terminology Criteria for Adverse Events version 4.03 grade ≥3) overall fluoropyrimidine-related toxicity across the entire treatment duration. We compared toxicity incidence between DPYD variant allele carriers and DPYD wild-type patients on an intention-to-treat basis, and relative risks (RRs) for severe toxicity were compared between the current study and a historical cohort of DPYD variant allele carriers treated with full dose fluoropyrimidine-based therapy (derived from a previously published meta-analysis). This trial is registered with ClinicalTrials.gov, number NCT02324452, and is complete. FINDINGS Between April 30, 2015, and Dec 21, 2017, we enrolled 1181 patients. 78 patients were considered non-evaluable, because they were retrospectively identified as not meeting inclusion criteria, did not start fluoropyrimidine-based treatment, or were homozygous or compound heterozygous DPYD variant allele carriers. Of 1103 evaluable patients, 85 (8%) were heterozygous DPYD variant allele carriers, and 1018 (92%) were DPYD wild-type patients. Overall, fluoropyrimidine-related severe toxicity was higher in DPYD variant carriers (33 [39%] of 85 patients) than in wild-type patients (231 [23%] of 1018 patients; p=0·0013). The RR for severe fluoropyrimidine-related toxicity was 1·31 (95% CI 0·63-2·73) for genotype-guided dosing compared with 2·87 (2·14-3·86) in the historical cohort for DPYD*2A carriers, no toxicity compared with 4·30 (2·10-8·80) in c.1679T>G carriers, 2·00 (1·19-3·34) compared with 3·11 (2·25-4·28) for c.2846A>T carriers, and 1·69 (1·18-2·42) compared with 1·72 (1·22-2·42) for c.1236G>A carriers. INTERPRETATION Prospective DPYD genotyping was feasible in routine clinical practice, and DPYD genotype-based dose reductions improved patient safety of fluoropyrimidine treatment. For DPYD*2A and c.1679T>G carriers, a 50% initial dose reduction was adequate. For c.1236G>A and c.2846A>T carriers, a larger dose reduction of 50% (instead of 25%) requires investigation. Since fluoropyrimidines are among the most commonly used anticancer agents, these findings suggest that implementation of DPYD genotype-guided individualised dosing should be a new standard of care. FUNDING Dutch Cancer Society.
Collapse
Affiliation(s)
- Linda M Henricks
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Carin A T C Lunenburg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Femke M de Man
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Didier Meulendijks
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Dutch Medicines Evaluation Board (CBG-MEB), Utrecht, Netherlands
| | - Geert W J Frederix
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emma Kienhuis
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, Netherlands
| | - Arnold Baars
- Department of Internal Medicine, Hospital Gelderse Vallei, Ede, Netherlands
| | - Vincent O Dezentjé
- Department of Internal Medicine, Reinier de Graaf Hospital, Delft, Netherlands; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Frank J F Jeurissen
- Department of Internal Medicine, Haaglanden Medical Center, The Hague, Netherlands
| | - Johanna E A Portielje
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands; Department of Internal Medicine, Haga Hospital, The Hague, Netherlands
| | - Rob L H Jansen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, Netherlands
| | - Albert J Ten Tije
- Department of Internal Medicine, Amphia Hospital, Breda, Netherlands
| | | | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Peter Nieboer
- Department of Internal Medicine, Wilhelmina Hospital Assen, Assen, Netherlands
| | | | | | - Hilde Rosing
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Erik van Werkhoven
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - André B P van Kuilenburg
- Laboratory of Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Annemieke Cats
- Department of Gastrointestinal Oncology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Jan H M Schellens
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
204
|
Abstract
Pharmacogenetics, a major component of individualized or precision medicine, relies on human genetic diversity. The remarkable developments in sequencing technologies have revealed that the number of genetic variants modulating drug action is much higher than previously thought and that a true personalized prediction of drug response requires attention to rare mutations (minor allele frequency, MAF<1%) in addition to polymorphisms (MAF>1%) in pharmacogenes. This has major implications for the conceptual development and clinical implementation of pharmacogenetics. Drugs used in cancer treatment have been major targets of pharmacogenetics studies, encompassing both germline polymorphisms and somatic variants in the tumor genome. The present overview, however, has a narrower scope and is focused on germline cancer pharmacogenetics, more specifically, on drug/gene pairs for which pharmacogenetics-informed prescription guidelines have been published by the Clinical Pharmacogenetics Implementation Consortium and/or the Dutch Pharmacogenetic Working Group, namely, thiopurines/TPMT, fluoropyrimidines/UGT1A1, irinotecan/UGT1A1 and tamoxifen/CYP2D6. I begin by reviewing the general principles of pharmacogenetics-informed prescription, pharmacogenetics testing and the perceived barriers to the adoption of routine pharmacogenetics testing in clinical practice. Then, I highlight aspects of the pharmacogenetics testing of the selected drug-gene pairs and finally present pharmacogenetics data from Brazilian studies pertinent to these drug-gene pairs. I conclude with the notion that pharmacogenetics testing has the potential to greatly benefit patients by enabling precision medicine applied to drug therapy, ensuring better efficacy and reducing the risk of adverse effects.
Collapse
Affiliation(s)
- Guilherme Suarez-Kurtz
- Instituto Nacional de Cancer, Rio de Janeiro, RJ, BR
- Rede Nacional de Farmacogenetica, Rio de Janeiro, RJ, BR
- *Corresponding author. E-mail:
| |
Collapse
|
205
|
Jacobs BAW, Pluim D, van der Laan P, Tzani A, Beijnen JH, Schellens JHM. Development and validation of a quantitative method for thymidine phosphorylase activity in peripheral blood mononuclear cells. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2018; 37:436-454. [PMID: 30285552 DOI: 10.1080/15257770.2018.1498270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The enzyme thymidine phosphorylase (TP) is important for activation of capecitabine and 5-fluorouracil. Assessment of TP phenotype might be suitable for identification of patients at risk of fluoropyrimidine-induced toxicity. In this paper, we describe the development and validation an assay for TP activity in peripheral blood mononuclear cells (PBMCs). The assay was based on ex vivo conversion of the TP substrate thymidine to thymine. The amount of thymine formed was determined by high-performance liquid chromatography - ultraviolet detection (HPLC-UV) with 5-bromouracil as internal standard. Lymphocytes and monocytes were purified from isolated PBMCs to examine cell-specific TP activity. TP activity in PBMCs demonstrated Michaelis-Menten kinetics. The lower limit of quantification was 2.3 µg PBMC protein and assay linearity was demonstrated up to 22.7 µg PBMC protein. Within-day and between-day precisions were ≤9.2% and ≤6.0%, respectively. Adequate stability TP activity was demonstrated after long-term storage of PBMC dry pellets and lysates at -80 °C. In monocytes, TP activity was approximately 3 times higher than in lymphocytes. Clinical applicability was demonstrated in samples that were collected from five cancer patients. A simple, precise and sensitive HPLC-UV assay for quantification of TP activity in PBMCs was developed that can be applied for clinical research.
Collapse
Affiliation(s)
- Bart A W Jacobs
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands.,b Department of Pharmacy & Pharmacology , The Netherlands Cancer Institute and MC Slotervaart , Amsterdam , The Netherlands
| | - Dick Pluim
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | - Pia van der Laan
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | - Anna Tzani
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | - Jos H Beijnen
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands.,b Department of Pharmacy & Pharmacology , The Netherlands Cancer Institute and MC Slotervaart , Amsterdam , The Netherlands.,c Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University , Utrecht , The Netherlands
| | - Jan H M Schellens
- a Department of Clinical Pharmacology , The Netherlands Cancer Institute , Amsterdam , The Netherlands.,c Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University , Utrecht , The Netherlands
| |
Collapse
|
206
|
Madi A, Fisher D, Maughan TS, Colley JP, Meade AM, Maynard J, Humphreys V, Wasan H, Adams RA, Idziaszczyk S, Harris R, Kaplan RS, Cheadle JP. Pharmacogenetic analyses of 2183 patients with advanced colorectal cancer; potential role for common dihydropyrimidine dehydrogenase variants in toxicity to chemotherapy. Eur J Cancer 2018; 102:31-39. [PMID: 30114658 DOI: 10.1016/j.ejca.2018.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/29/2018] [Accepted: 07/08/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inherited genetic variants may influence response to, and side-effects from, chemotherapy. We sought to generate a comprehensive inherited pharmacogenetic profile for oxaliplatin and 5FU/capecitabine therapy in advanced colorectal cancer (aCRC). METHODS We analysed more than 200 potentially functional, common, inherited variants in genes within the 5FU, capecitabine, oxaliplatin and DNA repair pathways, together with four rare dihydropyrimidine dehydrogenase (DPYD) variants, in 2183 aCRC patients treated with oxaliplatin-fluoropyrimidine chemotherapy with, or without, cetuximab (from MRC COIN and COIN-B trials). Primary end-points were response, any toxicity and peripheral neuropathy. We had >85% power to detect odds ratios (ORs) = 1.3 for variants with minor allele frequencies >20%. RESULTS Variants in DNA repair genes (Asn279Ser in EXO1 and Arg399Gln in XRCC1) were most associated with response (OR 1.9, 95% confidence interval [CI] 1.2-2.9, P = 0.004, and OR 0.7, 95% CI 0.5-0.9, P = 0.003, respectively). Common variants in DPYD (Cys29Arg and Val732Ile) were most associated with toxicity (OR 0.8, 95% CI 0.7-1.0, P = 0.008, and OR 1.6, 95% CI 1.1-2.1, P = 0.006, respectively). Two rare DPYD variants were associated with increased toxicity (Asp949Val with neutropenia, nausea and vomiting, diarrhoea and infection; IVS14+1G>A with lethargy, diarrhoea, stomatitis, hand-foot syndrome and infection; all ORs > 3). Asp317His in DCLRE1A was most associated with peripheral neuropathy (OR 1.3, 95% CI 1.1-1.6, P = 0.003). No common variant associations remained significant after Bonferroni correction. CONCLUSIONS DNA repair genes may play a significant role in the pharmacogenetics of aCRC. Our data suggest that both common and rare DPYD variants may be associated with toxicity to fluoropyrimidine-based chemotherapy.
Collapse
Affiliation(s)
- Ayman Madi
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - David Fisher
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Timothy S Maughan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - James P Colley
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Angela M Meade
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Julie Maynard
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Vikki Humphreys
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Harpreet Wasan
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK
| | - Richard A Adams
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Shelley Idziaszczyk
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Rebecca Harris
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Richard S Kaplan
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Jeremy P Cheadle
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
207
|
Patel JN. Lessons in practicing cancer genomics and precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1526081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Jai N. Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
208
|
Economic burden of adverse drug reactions and potential for pharmacogenomic testing in Singaporean adults. THE PHARMACOGENOMICS JOURNAL 2018; 19:401-410. [PMID: 30250149 DOI: 10.1038/s41397-018-0053-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 07/03/2018] [Accepted: 08/10/2018] [Indexed: 01/12/2023]
Abstract
Adverse drug reactions (ADRs) contribute to hospitalization but data on its economic burden is scant. Pre-emptive pharmacogenetic (PGx) testing can potentially reduce ADRs and its associated costs. The objectives of this study were to quantify the economic burden of ADRs and to estimate the breakeven cost of pre-emptive PGx testing in Singapore. We collected itemized costs for 1000 random non-elective hospitalizations of adults admitted to a tertiary-care general hospital in Singapore. The presence of ADRs at admission and their clinical characteristics were reported previously. The economic burden of ADRs was assessed from two perspectives: (1) Total cost and (2) incremental costs. The breakeven cost of PGx testing was estimated by dividing avoidable hospitalization costs for ADRs due to selected drugs by the number of patients taking those drugs. The total cost of 81 admissions caused by ADRs was US$570,404. Costs were significantly higher for bleeding/elevated international normalized ratio (US$9906 vs. US$2251, p = 6.58 × 10-3) compared to other ADRs, and for drugs acting on the blood coagulation system (US$9884 vs. US$2229, p = 4.41 × 10-3) compared to other drug classes. There were higher incremental laboratory costs due to ADRs causing or being present at admission. The estimated breakeven cost of a pre-emptive PGx test for patients taking warfarin, clopidogrel, chemotherapeutic and neuropsychiatric drugs was US$114 per patient. These results suggest that future studies designed to directly measure the clinical and cost impact of a pre-emptive genotyping program will help inform clinical practice and health policy decisions.
Collapse
|
209
|
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]
|
210
|
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
|
211
|
Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018; 39:18-40. [PMID: 30075835 DOI: 10.1016/j.drup.2018.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Adverse events affect the pharmacological treatment of approximately 90% of colorectal cancer (CRC) patients at any stage of the disease. Chemotherapy including fluoropyrimidines, irinotecan, and oxaliplatin is the cornerstone of the pharmacological treatment of CRC. The introduction of novel targeted agents, as anti-EGFR (i.e. cetuximab, panitumumab) and antiangiogenic (i.e. bevacizumab, ziv-aflibercept, regorafenib, and ramucirumab) molecules, into the oncologist's toolbox has led to significant improvements in the life expectancy of advanced CRC patients, but with a substantial increase in toxicity burden. In this respect, pharmacogenomics has largely been applied to the personalization of CRC chemotherapy, focusing mainly on the study of inhered polymorphisms in genes encoding phase I and II enzymes, ATP-binding cassette (ABC)/solute carrier (SLC) membrane transporters, proteins involved in DNA repair, folate pathway and immune response. These research efforts have led to the identification of some validated genetic markers of chemotherapy toxicity, for fluoropyrimidines and irinotecan. No validated genetic determinants of oxaliplatin-specific toxicity, as peripheral neuropathy, has thus far been established. The contribution of host genetic markers in predicting the toxicity associated with novel targeted agents' administration is still controversial due to the heterogeneity of published data. Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. However, these recommendations are still poorly applied at the patient's bedside. Several ongoing projects in the U.S. and Europe are currently evaluating how pharmacogenomics can be implemented successfully in daily clinical practice. The majority of drug-related adverse events are still unexplained, and a great deal of ongoing research is aimed at improving knowledge of the role of pharmacogenomics in increasing treatment safety. In this review, the issue of pre-treatment identification of CRC patients at risk of toxicity via the analysis of patients' genetic profiles is addressed. Available pharmacogenomics guidelines with ongoing efforts to implement them in clinical practice and new exploratory markers for clinical validation are described.
Collapse
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
212
|
Hamzic S, Amstutz U, Largiadèr CR. Come a long way, still a ways to go: from predicting and preventing fluoropyrimidine toxicity to increased efficacy? Pharmacogenomics 2018; 19:689-692. [PMID: 29783877 DOI: 10.2217/pgs-2018-0040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Seid Hamzic
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.,Graduate School for Cellular & Biomedical Sciences, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Ursula Amstutz
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| | - Carlo R Largiadèr
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| |
Collapse
|
213
|
Battaglin F, Puccini A, Naseem M, Schirripa M, Berger MD, Tokunaga R, McSkane M, Khoukaz T, Soni S, Zhang W, Lenz HJ. Pharmacogenomics in colorectal cancer: current role in clinical practice and future perspectives. JOURNAL OF CANCER METASTASIS AND TREATMENT 2018; 4:12. [PMID: 34532592 PMCID: PMC8442855 DOI: 10.20517/2394-4722.2018.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment scenario of colorectal cancer (CRC) has been evolving in recent years with the introduction of novel targeted agents and new therapeutic strategies for the metastatic disease. An extensive effort has been directed to the identification of predictive biomarkers to aid patients selection and guide therapeutic choices. Pharmacogenomics represents an irreplaceable tool to individualize patients treatment based on germline and tumor acquired somatic genetic variations able to predict drugs response and risk of toxicities. The growing knowledge of CRC molecular characteristics and complex genomic makeup has played a crucial role in identifying predictive pharmacogenomic biomarkers, while supporting the rationale for the development of new drugs and treatment combinations. Clinical validation of promising biomarkers, however, is often an issue. More recently, a deeper understanding of resistance mechanisms and tumor escape dynamics under treatment pressure and the availability of novel technologies are opening new perspectives in this field. This review aims to present an overview of current pharmacogenomic biomarkers and future perspectives of pharmacogenomics in CRC, in an evolving scenario moving from a single drug-gene interactions approach to a more comprehensive genome-wide approach, comprising genomics and epigenetics.
Collapse
Affiliation(s)
- Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV - IRCCS, Padua 35128, Italy
| | - Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Marta Schirripa
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV - IRCCS, Padua 35128, Italy
| | - Martin D. Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Medical Oncology, University Hospital of Bern, Bern 3010, Switzerland
| | - Ryuma Tokunaga
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michelle McSkane
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Taline Khoukaz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
214
|
Roncato R, Cecchin E, Toffoli G. Improving decision making on DPYD and UGT1A1*28 patients’ profiling with an innovative reimbursement strategy. Pharmacogenomics 2018; 19:301-304. [DOI: 10.2217/pgs-2017-0303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Rossana Roncato
- Experimental & Clinical Pharmacology, Centro Di Riferimento Oncologico - National Cancer Center, Aviano (PN), Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology, Centro Di Riferimento Oncologico - National Cancer Center, Aviano (PN), Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology, Centro Di Riferimento Oncologico - National Cancer Center, Aviano (PN), Italy
| |
Collapse
|
215
|
[Dihydropyrimidine déhydrogenase (DPD) deficiency screening and securing of fluoropyrimidine-based chemotherapies: Update and recommendations of the French GPCO-Unicancer and RNPGx networks]. Bull Cancer 2018; 105:397-407. [PMID: 29486921 DOI: 10.1016/j.bulcan.2018.02.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/04/2018] [Accepted: 02/06/2018] [Indexed: 11/21/2022]
Abstract
Fluoropyrimidines (FU) are still the most prescribed anticancer drugs for the treatment of solid cancers. However, fluoropyrimidines cause severe toxicities in 10 to 40% of patients and toxic deaths in 0.2 to 0.8% of patients, resulting in a real public health problem. The main origin of FU-related toxicities is a deficiency of dihydropyrimidine dehydrogenase (DPD), the rate-limiting enzyme of 5-FU catabolism. DPD deficiency may be identified through pharmacogenetics testing including phenotyping (direct or indirect measurement of enzyme activity) or genotyping (detection of inactivating polymorphisms on the DPYD gene). Approximately 3 to 15% of patients exhibit a partial deficiency and 0.1 to 0.5% a complete DPD deficiency. Currently, there is no regulatory obligation for DPD deficiency screening in patients scheduled to receive a fluoropyrimidine-based chemotherapy. Based on the levels of evidence from the literature data and considering current French practices, the Group of Clinical Pharmacology in Oncology (GPCO)-UNICANCER and the French Network of Pharmacogenetics (RNPGx) recommend the following: (1) to screen DPD deficiency before initiating any chemotherapy containing 5-FU or capecitabine; (2) to perform DPD phenotyping by measuring plasma uracil (U) concentrations (possibly associated with dihydrouracil/U ratio), and DPYD genotyping (variants *2A, *13, p.D949V, HapB3); (3) to reduce the initial FU dose (first cycle) according to DPD status, if needed, and further, to consider increasing the dose at subsequent cycles according to treatment tolerance. In France, 17 public laboratories currently undertake routine screening of DPD deficiency.
Collapse
|
216
|
The impact of liver resection on the dihydrouracil:uracil plasma ratio in patients with colorectal liver metastases. Eur J Clin Pharmacol 2018; 74:737-744. [PMID: 29430582 DOI: 10.1007/s00228-018-2426-4] [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] [Received: 07/13/2017] [Accepted: 02/01/2018] [Indexed: 12/30/2022]
Abstract
PURPOSE The dihydrouracil (DHU):uracil (U) plasma ratio is a promising marker for identification of dihydropyrimidine dehydrogenase (DPD)-deficient patients. The objective of this study was to determine the effect of liver resection on the DHU:U plasma ratio in patients with colorectal liver metastases (CRLM). METHODS An observational study was performed in which DHU:U plasma ratios in patients with CRLM were analyzed prior to and 1 day after liver resection. In addition, the DHU:U plasma ratio was quantified in six additional patients 4-8 weeks after liver resection to explore long-term effects on the DHU:U plasma ratio. Quantification of U and DHU plasma levels was performed using a validated ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) assay. RESULTS The median (range) DHU:U plasma ratio in 15 patients prior to liver resection was 10.7 (2.6-14.4) and was significantly reduced to 5.5 (< quantification limit (LLOQ-10.5) 1 day after resection (p = 0.0026). This reduction was caused by a decrease in DHU plasma levels from 112.0 (79.8-153) ng/mL to 41.2 (< LLOQ-160) ng/mL 1 day after resection (p = 0.0004). Recovery of the DHU:U plasma ratio occurred 4-8 weeks after liver resection, which was shown by a median (range) DHU:U plasma ratio in six patients of 9.1 (6.9-14.5). CONCLUSION Liver resection leads to very low DHU:U plasma ratios 1 day after liver resection, which is possibly caused by a reduction in DPD activity. Quantification of the DHU:U plasma ratios directly after liver resection could lead to false-positive identification of DPD deficiency and is therefore not advised.
Collapse
|
217
|
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
|
218
|
Danesi R, Del Re M, Ciccolini J, Schellens JHM, Schwab M, van Schaik RHN, van Kuilenburg ABP. Prevention of fluoropyrimidine toxicity: do we still have to try our patient's luck? Ann Oncol 2018; 28:183. [PMID: 27687313 DOI: 10.1093/annonc/mdw448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- R Danesi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M Del Re
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - J Ciccolini
- Clinical Pharmacokinetics, La Timone University Hospital, Marseille, and GPCO-Unicancer, Paris, France
| | - J H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Schwab
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and Department of Clinical Pharmacology, University Hospital, Tuebingen, Germany
| | - R H N van Schaik
- IFCC Reference center - Pharmacogenetics, Department of Clinical Chemistry, Erasmus University, Rotterdam
| | - A B P van Kuilenburg
- Department of Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
219
|
Milano G. DPD testing must remain a recommended option, but not a recommended routine test. Ann Oncol 2018; 28:1399. [PMID: 28327971 DOI: 10.1093/annonc/mdx118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- G Milano
- Centre Antoine-Lacassagne, Oncopharmacology Unit, UNS EA 3836, Nice, France
| |
Collapse
|
220
|
Bank PCD, Swen JJ, Guchelaar HJ. Implementation of Pharmacogenomics in Everyday Clinical Settings. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 83:219-246. [PMID: 29801576 DOI: 10.1016/bs.apha.2018.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Currently, germline pharmacogenomics (PGx) is successfully implemented within certain specialties in clinical care. With the integration of PGx in pharmacotherapy multiple stakeholders are involved, which are identified in this chapter. Clinically relevant pharmacogenes with their related PGx test are discussed, along with diagnostic test criteria to guide clinicians and policy makers in PGx test selection. The chapter further reviews the similarities and the differences between the guidelines of the Dutch Pharmacogenetics Working Group and the Clinical Pharmacogenetics Implementation Consortium which both support healthcare professionals in understanding PGx test results and help guiding pharmacotherapy by providing evidence-based dosing recommendations. Finally, clinical studies which provide scientific evidence and information on cost-effectiveness supporting clinical implementation of PGx in clinical care are discussed along with the remaining barriers for adoption of PGx testing by healthcare professionals.
Collapse
Affiliation(s)
- Paul C D Bank
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
221
|
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]
|
222
|
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
|
223
|
Smyth E, Zhang S, Cunningham D, Wotherspoon A, Soong R, Peckitt C, Valeri N, Fassan M, Rugge M, Okines A, Allum W, Stenning S, Nankivell M, Langley R, Tan P. Pharmacogenetic Analysis of the UK MRC (Medical Research Council) MAGIC Trial: Association of Polymorphisms with Toxicity and Survival in Patients Treated with Perioperative Epirubicin, Cisplatin, and 5-fluorouracil (ECF) Chemotherapy. Clin Cancer Res 2017; 23:7543-7549. [PMID: 28972045 PMCID: PMC6175041 DOI: 10.1158/1078-0432.ccr-16-3142] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/21/2017] [Accepted: 09/26/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Germline polymorphisms may affect chemotherapy efficacy and toxicity. We examined the effect of polymorphisms in drug metabolism and DNA repair genes on pathologic response rates, survival, and toxicity for patients randomized to surgery alone or perioperative ECF chemotherapy in the MRC MAGIC trial.Experimental Design: DNA was extracted from nontumor resection formalin-fixed paraffin-embedded (FFPE) blocks. ERCC1, ERCC2, XRCC1, DYPD, and OPRT SNPs were evaluated using Sequenom, GSTP1, GSTT1 deletion, and TYMS (TS) 5' 2R/3R using multiplex PCR. Post PCR amplification, TS 2R/3R and GSTT1 samples underwent gel electrophoresis.Results: Polymorphism data were available for 289 of 456 (63.4%) operated patients. No polymorphism was statistically significantly associated with pathologic response to chemotherapy. Median overall survival (OS) for patients treated with surgery alone with any TS genotype was not different (1.76 years 2R/2R, 1.68 years 2R/3R, 2.09 years 3R/3R). Median OS for patients with a TS 2R/2R genotype treated with chemotherapy was not reached, whereas median OS for 2R/3R and 3R/3R patients were 1.44 and 1.60 years, respectively (log rank P value = 0.0053). The P value for the interaction between treatment arm and genotype (3R/3R and 3R/2R vs. 2R/2R) was 0.029. No polymorphism was statistically significantly associated with chemotherapy toxicity.Conclusions: In MAGIC, patients with a TS 2R/2R genotype appeared to derive a larger benefit from perioperative ECF chemotherapy than patients with 3R containing genotypes. Further exploration of this potential predictive biomarker in this patient population is warranted. Clin Cancer Res; 23(24); 7543-9. ©2017 AACR.
Collapse
Affiliation(s)
- Elizabeth Smyth
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Shenli Zhang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - David Cunningham
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom.
| | - Andrew Wotherspoon
- Department of Pathology, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Clare Peckitt
- Department of Clinical Research and Development, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Nicola Valeri
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
- Department of Molecular Pathology, The Institute of Cancer Research London & Sutton, United Kingdom
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Massimo Rugge
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Alicia Okines
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - William Allum
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Sally Stenning
- Department of Surgery, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Matthew Nankivell
- Department of Surgery, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Ruth Langley
- Medical Research Council Clinical Trials Unit at UCL, London, United Kingdom
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Cancer Therapeutics and Stratified Oncology Group, Genome Institute of Singapore, Singapore, Singapore
| |
Collapse
|
224
|
Hariprakash JM, Vellarikkal SK, Keechilat P, Verma A, Jayarajan R, Dixit V, Ravi R, Senthivel V, Kumar A, Sehgal P, Sonakar AK, Ambawat S, Giri AK, Philip A, Sivadas A, Faruq M, Bharadwaj D, Sivasubbu S, Scaria V. Pharmacogenetic landscape of DPYD variants in south Asian populations by integration of genome-scale data. Pharmacogenomics 2017; 19:227-241. [PMID: 29239269 DOI: 10.2217/pgs-2017-0101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Adverse drug reactions to 5-Fluorouracil(5-FU) is frequent and largely attributable to genetic variations in the DPYD gene, a rate limiting enzyme that clears 5-FU. The study aims at understanding the pharmacogenetic landscape of DPYD variants in south Asian populations. MATERIALS & METHODS Systematic analysis of population scale genome wide datasets of over 3000 south Asians was performed. Independent evaluation was performed in a small cohort of patients. RESULTS Our analysis revealed significant differences in the the allelic distribution of variants in different ethnicities. CONCLUSIONS This is the first and largest genetic map the DPYD variants associated with adverse drug reaction to 5-FU in south Asian population. Our study highlights ethnic differences in allelic frequencies.
Collapse
Affiliation(s)
- Judith M Hariprakash
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Shamsudheen K Vellarikkal
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Pavithran Keechilat
- Department of Medical Oncology, Amrita Institute of Medical Sciences & Research Centre, Amrita University, Kochi-682041, India
| | - Ankit Verma
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Rijith Jayarajan
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Vishal Dixit
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Rowmika Ravi
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Vigneshwar Senthivel
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Anoop Kumar
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Paras Sehgal
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Akhilesh K Sonakar
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Sakshi Ambawat
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Anil K Giri
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Arun Philip
- Department of Medical Oncology, Amrita Institute of Medical Sciences & Research Centre, Amrita University, Kochi-682041, India
| | - Akhila Sivadas
- Department of Medical Oncology, Amrita Institute of Medical Sciences & Research Centre, Amrita University, Kochi-682041, India
| | - Mohammed Faruq
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Dwaipayan Bharadwaj
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Sridhar Sivasubbu
- Genomics & Molecular Medicine, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| | - Vinod Scaria
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics & Integrative Biology, Mathura Road, Delhi 110025, India
| |
Collapse
|
225
|
Henricks LM, Opdam FL, Beijnen JH, Cats A, Schellens JHM. DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update. Ann Oncol 2017; 28:2915-2922. [PMID: 29045513 DOI: 10.1093/annonc/mdx411] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The fluoropyrimidine anticancer drugs, especially 5-fluorouracil (5-FU) and capecitabine, are frequently prescribed for several types of cancer, including breast, colorectal, head and neck and gastric cancer. In the current drug labels of 5-FU and capecitabine in the European Union and the United States, no adaptive dosing strategies are incorporated for polymorphic metabolism of 5-FU. Although treatment with fluoropyrimidines is generally well tolerated, a major clinical limitation is that a proportion of the treated population experiences severe, sometimes life-threatening, fluoropyrimidine-related toxicity. This toxicity is strongly affected by interindividual variability in activity of dihydropyrimidine dehydrogenase (DPD), the main metabolic enzyme for inactivation of fluoropyrimidines, with an estimated 3%-8% of the population being partially DPD deficient. A reduced functional or abrogated DPD enzyme is often caused by genetic polymorphisms in DPYD, the gene encoding for DPD, and heterozygous carriers of such DPYD polymorphisms have a partial DPD deficiency. When these partially DPD deficient patients are treated with a full dose of fluoropyrimidines, they are generally exposed to toxic levels of 5-FU and its metabolites, and the risk of developing severe treatment-related toxicity is therefore significantly increased.Currently, functional and clinical validity is well established for four DPYD variants (DPYD*2A, c.2846A>T, c.1679T>G and c.1236G>A), as those variants have retrospectively and in a large population study prospectively been shown to be associated with increased risk of fluoropyrimidine-associated toxicity. Patient safety of fluoropyrimidine treatment can be significantly improved by pre-emptive screening for DPYD genotype variants and dose reductions in heterozygous DPYD variant allele carriers, thereby normalizing 5-FU exposure. Based on the critical appraisal of currently available data, adjusting the labels of capecitabine and 5-FU by including recommendations on pre-emptive screening for DPYD variants and DPYD genotype-guided dose adjustments should be the new standard of care.
Collapse
Affiliation(s)
- L M Henricks
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - F L Opdam
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| | - A Cats
- Department of Gastroenterology and Hepatology, Division of Medical Oncology
| | - J H M Schellens
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| |
Collapse
|
226
|
Páez D, Salazar R, Tabernero J. DPYD genotype-guided fluoropyrimidines dose: is it ready for prime time? Ann Oncol 2017; 28:2913-2914. [PMID: 29045550 DOI: 10.1093/annonc/mdx632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- D Páez
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - R Salazar
- Medical Oncology Department, Catalan Institute of Oncology (ICO), CIBERONC, L'Hospitalet de Llobregat, Barcelona
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
227
|
Lauschke VM, Milani L, Ingelman-Sundberg M. Pharmacogenomic Biomarkers for Improved Drug Therapy—Recent Progress and Future Developments. AAPS JOURNAL 2017; 20:4. [DOI: 10.1208/s12248-017-0161-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/06/2017] [Indexed: 12/13/2022]
|
228
|
Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JHM, Swen JJ, Klein TE, McLeod HL, Caudle KE, Diasio RB, Schwab M. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther 2017; 103:210-216. [PMID: 29152729 DOI: 10.1002/cpt.911] [Citation(s) in RCA: 427] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/19/2017] [Accepted: 10/11/2017] [Indexed: 12/13/2022]
Abstract
The purpose of this guideline is to provide information for the interpretation of clinical dihydropyrimidine dehydrogenase (DPYD) genotype tests so that the results can be used to guide dosing of fluoropyrimidines (5-fluorouracil and capecitabine). Detailed guidelines for the use of fluoropyrimidines, their clinical pharmacology, as well as analyses of cost-effectiveness are beyond the scope of this document. The Clinical Pharmacogenetics Implementation Consortium (CPIC® ) guidelines consider the situation of patients for which genotype data are already available (updates available at https://cpicpgx.org/guidelines/guideline-for-fluoropyrimidines-and-dpyd/).
Collapse
Affiliation(s)
- Ursula Amstutz
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Linda M Henricks
- Department of Clinical Pharmacology, Division of Medical Oncology and Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Steven M Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Julia Barbarino
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Division of Medical Oncology and Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute and the Department of Population Sciences, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Kelly E Caudle
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Robert B Diasio
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Clinical Pharmacology, University Hospital, Tuebingen, Germany.,Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
229
|
Henricks LM, Kienhuis E, de Man FM, van der Veldt AA, Hamberg P, van Kuilenburg AB, van Schaik RH, Lunenburg CA, Guchelaar HJ, Schellens JH, Mathijssen RH. Treatment Algorithm for Homozygous or Compound Heterozygous DPYD Variant Allele Carriers With Low-Dose Capecitabine. JCO Precis Oncol 2017; 1:1-10. [DOI: 10.1200/po.17.00118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Linda M. Henricks
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Emma Kienhuis
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Femke M. de Man
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Astrid A.M. van der Veldt
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Paul Hamberg
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - André B.P. van Kuilenburg
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Ron H.N. van Schaik
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Carin A.T.C. Lunenburg
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Henk-Jan Guchelaar
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Jan H.M. Schellens
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| | - Ron H.J. Mathijssen
- Linda M. Henricks, Astrid A.M. van der Veldt, and Jan H.M. Schellens, the Netherlands Cancer Institute; André B.P. van Kuilenburg, Academic Medical Center, Amsterdam; Emma Kienhuis, Femke M. de Man, Astrid A.M. van der Veldt, Ron H.N. van Schaik, and Ron H.J. Mathijssen, Erasmus Medical Center; Paul Hamberg, Franciscus Gasthuis & Vlietland, Rotterdam; Carin A.T.C. Lunenburg and Henk-Jan Guchelaar, Leiden University Medical Center, Leiden; and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
230
|
Ab Mutalib NS, Md Yusof NF, Abdul SN, Jamal R. Pharmacogenomics DNA Biomarkers in Colorectal Cancer: Current Update. Front Pharmacol 2017; 8:736. [PMID: 29075194 PMCID: PMC5644034 DOI: 10.3389/fphar.2017.00736] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/29/2017] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) remains as one of the most common cause of worldwide cancer morbidity and mortality. Improvements in surgical modalities and adjuvant chemotherapy have increased the cure rates in early stage disease, but a significant portion of the patients will develop recurrence or advanced disease. The efficacy of chemotherapy of recurrence and advanced CRC has improved significantly over the last decade. Previously, the historical drug 5-fluorouracil was used as single chemotherapeutic agent. Now with the addition of other drugs such as capecitabine, irinotecan, oxaliplatin, bevacizumab, cetuximab, panitumumab, vemurafenib, and dabrafenib, the median survival of patients with advanced CRC has significantly improved from less than a year to the current standard of almost 2 years. However, the side effects of systemic therapy such as toxicity may cause fatal complications and have a major consequences on the patients' quality of life. Hence, there is an urgent need for key biomarkers which will enable the selection of optimal drug singly or in combination for an individual patient. The application of personalized therapy based on DNA testing could aid the clinicians in providing the most effective chemotherapy agents and dose modifications for each patient. Yet, some of the current findings are controversial and the evidences are conflicting. This review aims at summarizing the current state of knowledge about germline pharmacogenomics DNA variants that are currently used to guide therapeutic decisions and variants that have the potential to be clinically useful in the future. In addition, current updates on germline variants conferring treatment sensitivity, drug resistance to existing chemotherapy agents and variants affecting prognosis and survival will also be emphasized. Different alteration in the same gene might confer resistance or enhanced sensitivity; and while most of other published reviews generally stated only the gene name and codon location, we will specifically discuss the exact variants to offer more accurate information in this mini review.
Collapse
Affiliation(s)
- Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Najwa F Md Yusof
- UKM Medical Molecular Biology Institute, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shafina-Nadiawati Abdul
- UKM Medical Molecular Biology Institute, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute, UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
231
|
Henricks LM, Siemerink EJ, Rosing H, Meijer J, Goorden SM, Polstra AM, Zoetekouw L, Cats A, Schellens JH, van Kuilenburg AB. Capecitabine-based treatment of a patient with a novelDPYDgenotype and complete dihydropyrimidine dehydrogenase deficiency. Int J Cancer 2017; 142:424-430. [DOI: 10.1002/ijc.31065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/16/2017] [Accepted: 09/08/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Linda M. Henricks
- Division of Pharmacology and Division of Clinical Pharmacology, Department of Medical Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Ester J.M. Siemerink
- Department of Internal Medicine; Ziekenhuis Groep Twente (ZGT); Hengelo The Netherlands
| | - Hilde Rosing
- Department of Pharmacy and Pharmacology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Judith Meijer
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Clinical Genetics; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Susan M.I. Goorden
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Clinical Genetics; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Abeltje M. Polstra
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Clinical Genetics; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
- Department of Clinical Genetics; VU University Medical Center; Amsterdam The Netherlands
| | - Lida Zoetekouw
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Clinical Genetics; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| | - Annemieke Cats
- Department of Gastrointestinal Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Jan H.M. Schellens
- Division of Pharmacology and Division of Clinical Pharmacology, Department of Medical Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University; Utrecht The Netherlands
| | - André B.P. van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Clinical Genetics; Academic Medical Center, University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
232
|
Pre-treatment assay of 5-fluorouracil degradation rate (5-FUDR) to improve prediction of 5-fluorouracil toxicity in gastro-esophageal cancer. Oncotarget 2017; 8:14050-14057. [PMID: 27738344 PMCID: PMC5355161 DOI: 10.18632/oncotarget.12571] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/05/2016] [Indexed: 01/25/2023] Open
Abstract
Background 5-fluorouracil (5-FU) based chemotherapy is the most common first line regimen used in gastric and gastroesophageal junction cancer, but development of severe toxicity is a main concern in the treatment. The present study is aimed to evaluate a novel pre-treatment assay, known as the 5-FU degradation rate (5-FUDR), as a predictive factor for 5-FU toxicity. Methods Pre-treatment 5-FUDR and gene polymorphisms related to 5-FU metabolism (DPYDIVS14+1G>A, MTHFRA1298T or C677T, TMYS TSER) were characterized in gastro-esophageal cancer patients. Association with toxicities was retrospectively evaluated, using multivariate logistic regression analysis. Results 107 gastro-esophageal cancer patients were retrospectively analyzed. No relation between gene polymorphisms and toxicity were detected, while low (< 5th centile) and high (> 95th centile) 5-FUDRs were associated with development of grade 3-4 toxicity (OR 11.14, 95% CI 1.09-113.77 and OR 9.63, 95% CI 1.70-54.55, p = 0.002). Conclusions Compared to currently used genetic tests, the pre-treatment 5-FUDR seems useful in identifying patients at risk of developing toxicity.
Collapse
|
233
|
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
|
234
|
Dihydropyrimidine dehydrogenase pharmacogenetics for predicting fluoropyrimidine-related toxicity in the randomised, phase III adjuvant TOSCA trial in high-risk colon cancer patients. Br J Cancer 2017; 117:1269-1277. [PMID: 29065426 PMCID: PMC5709672 DOI: 10.1038/bjc.2017.289] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/28/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Dihydropyrimidine dehydrogenase (DPD) catabolises ∼85% of the administered dose of fluoropyrimidines. Functional DPYD gene variants cause reduced/abrogated DPD activity. DPYD variants analysis may help for defining individual patients’ risk of fluoropyrimidine-related severe toxicity. Methods: The TOSCA Italian randomised trial enrolled colon cancer patients for 3 or 6 months of either FOLFOX-4 or XELOX adjuvant chemotherapy. In an ancillary pharmacogenetic study, 10 DPYD variants (*2A rs3918290 G>A, *13 rs55886062 T>G, rs67376798 A>T, *4 rs1801158 G>A, *5 rs1801159 A>G, *6 rs1801160 G>A, *9A rs1801265 T>C, rs2297595 A>G, rs17376848 T>C, and rs75017182 C>G), were retrospectively tested for associations with ⩾grade 3 fluoropyrimidine-related adverse events (FAEs). An association analysis and a time-to-toxicity (TTT) analysis were planned. To adjust for multiple testing, the Benjamini and Hochberg’s False Discovery Rate (FDR) procedure was used. Results: FAEs occurred in 194 out of 508 assessable patients (38.2%). In the association analysis, FAEs occurred more frequently in *6 rs1801160 A allele carriers (FDR=0.0083). At multivariate TTT analysis, significant associations were found for *6 rs1801160 A allele carriers (FDR<0.0001), *2A rs3918290 A allele carriers (FDR<0.0001), and rs2297595 GG genotype carriers (FDR=0.0014). Neutropenia was the most common FAEs (28.5%). *6 rs1801160 (FDR<0.0001), and *2A rs3918290 (FDR=0.0004) variant alleles were significantly associated with time to neutropenia. Conclusions: This study adds evidence on the role of DPYD pharmacogenetics for safety of patients undergoing fluoropyrimidine-based chemotherapy.
Collapse
|
235
|
García-González X, López-Fernández LA. Using pharmacogenetics to prevent severe adverse reactions to capecitabine. Pharmacogenomics 2017; 18:1199-1213. [DOI: 10.2217/pgs-2017-0102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Xandra García-González
- Servicio de Farmacia, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Luis A López-Fernández
- Servicio de Farmacia, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
236
|
Geenen JW, Baranova EV, Asselbergs FW, de Boer A, Vreman RA, Palmer CNA, Maitland-van der Zee AH, Hövels AM. Early health technology assessments in pharmacogenomics: a case example in cardiovascular drugs. Pharmacogenomics 2017; 18:1143-1153. [DOI: 10.2217/pgs-2017-0063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the required characteristics (cost, sensitivity and specificity) of a pharmacogenomic test for being a cost-effective prevention of angiotensin-converting enzyme inhibitors induced angioedema. Furthermore, we assessed the influence of only testing high-risk populations. Materials & methods: A decision tree was used. Results: With a willingness-to-pay threshold of €20,000 and €80,000 per quality adjusted life year, a 100% sensitive and specific test may have a maximum cost of €1.30 and €1.95, respectively. When only genotyping high-risk populations, the maximum test price would be €5.03 and €7.55, respectively. Conclusion: This theoretical pharmacogenomic test is only cost-effective at high specificity, high sensitivity and a low price. Only testing high-risk populations yields more realistic maximum test prices for cost–effectiveness of the intervention.
Collapse
Affiliation(s)
- Joost W Geenen
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| | - Ekaterina V Baranova
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, UMC Utrecht, 3508 GA Utrecht, The Netherlands
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, 3501 DG Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, WC1E 6BT London, UK
| | - Anthonius de Boer
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| | - Rick A Vreman
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| | - Colin NA Palmer
- Population Pharmacogenetics Group, Biomedical Research Centre, University of Dundee, Ninewells Hospital & Medical School, DD2 1GZ Dundee, UK
| | - Anke H Maitland-van der Zee
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
- Department of Respiratory Disease, Academic Medical Center, PO Box 22660,1100 DD, Amsterdam, The Netherlands
| | - Anke M Hövels
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
237
|
Abstract
Pharmacogenomics (PGx), a substantial component of "personalized medicine", seeks to understand each individual's genetic composition to optimize drug therapy -- maximizing beneficial drug response, while minimizing adverse drug reactions (ADRs). Drug responses are highly variable because innumerable factors contribute to ultimate phenotypic outcomes. Recent genome-wide PGx studies have provided some insight into genetic basis of variability in drug response. These can be grouped into three categories. [a] Monogenic (Mendelian) traits include early examples mostly of inherited disorders, and some severe (idiosyncratic) ADRs typically influenced by single rare coding variants. [b] Predominantly oligogenic traits represent variation largely influenced by a small number of major pharmacokinetic or pharmacodynamic genes. [c] Complex PGx traits resemble most multifactorial quantitative traits -- influenced by numerous small-effect variants, together with epigenetic effects and environmental factors. Prediction of monogenic drug responses is relatively simple, involving detection of underlying mutations; due to rarity of these events and incomplete penetrance, however, prospective tests based on genotype will have high false-positive rates, plus pharmacoeconomics will require justification. Prediction of predominantly oligogenic traits is slowly improving. Although a substantial fraction of variation can be explained by limited numbers of large-effect genetic variants, uncertainty in successful predictions and overall cost-benefit ratios will make such tests elusive for everyday clinical use. Prediction of complex PGx traits is almost impossible in the foreseeable future. Genome-wide association studies of large cohorts will continue to discover relevant genetic variants; however, these small-effect variants, combined, explain only a small fraction of phenotypic variance -- thus having limited predictive power and clinical utility.
Collapse
Affiliation(s)
- Ge Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States.
| | - Daniel W Nebert
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States; Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati School of Medicine, Cincinnati, OH 45267-0056, United States.
| |
Collapse
|
238
|
Liu D, Li J, Gao J, Li Y, Yang R, Shen L. Examination of multiple UGT1A and DPYD polymorphisms has limited ability to predict the toxicity and efficacy of metastatic colorectal cancer treated with irinotecan-based chemotherapy: a retrospective analysis. BMC Cancer 2017. [PMID: 28637434 PMCID: PMC5480170 DOI: 10.1186/s12885-017-3406-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background To evaluate a new UGT1A and DPYD polymorphism panel to better predict irinotecan-induced toxicity and the clinical response in Chinese patients with metastatic colorectal cancer (mCRC). Methods The genotypes of UGT1A (UGT1A1*6, UGT1A1*27, UGT1A1*28, UGT1A7*2, UGT1A7*3, UGT1A7*4 and UGT1A9*22) and DPYD (DPYD*5, DPYD c.1896 T > C, and DPYD*2A) were examined by direct sequencing in 661 mCRC patients receiving irinotecan-based chemotherapy. The influences of UGT1A and DPYD polymorphisms on severe irinotecan-induced toxicities and clinical outcomes were assessed. Results In the cohort studied here, the incidence of UGT1A1*6, UGT1A1*28, UGT1A7*2, UGT1A7*3, UGT1A9*22, DPYD*5, and DPYD c.1896 T > C variants were 34.8%, 24.2%, 34.3%, 39.4%, 81.8%, 48.4% and 20.4%, respectively. UGT1A1*27 and DPYD*2A had low frequencies and UGT1A7*4 was not found. A total of 59 patients (8.9%) suffered severe diarrhea and 136 patients (20.6%) suffered severe neutropenia. UGT1A1*28 heterozygotes (OR = 2.263, 95%CI 1.395–3.670), UGT1A1*28 homozygotes (OR = 5.910, 95%CI 1.138–30.672) and UGT1A1*6 homozygotes (OR = 4.737, 95%CI 1.946–11.533) were independent risk factors for severe neutropenia. UGT1A polymorphisms were not found to relate to severe diarrhea. DPYD*5 was determined to be an independent risk factor for severe diarrhea (OR = 2.143, 95%CI 1.136–4.041). Neither DPYD*5 nor DPYD c.1896 T > C was found to relate to severe neutropenia. In the first-line irinotecan-based treatment, UGT1A1*28 and DPYD*5 contributed to higher response rates (P = 0.043 and P = 0.019, respectively), while DPYD*5 was found to associate with better progression-free survival (P = 0.015). UGT1A1*27 contributed to worse overall survival (P < 0.001). Conclusion Results still showed UGT1A1*6 and UGT1A1*28 to be partially associated with irinotecan-induced toxicity and clinical response. An examination of more UGT1A loci, except for UGT1A1*6 and UGT1A1*28, was not helpful to improve the predictive value of irinotecan-based toxicity and efficacy. An examination of DPYD*5 assisted in the prediction of severe diarrhea. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3406-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jian Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jing Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yanyan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Rui Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
239
|
|
240
|
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
|
241
|
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
|
242
|
Quaranta S, Thomas F. Pharmacogénétique des médicaments anticancéreux : état des connaissances et des pratiques – recommandations du Réseau national de pharmacogénétique (RNPGx). Therapie 2017; 72:193-204. [DOI: 10.1016/j.therap.2016.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 11/26/2022]
|
243
|
A phase II study of preoperative chemoradiation with tegafur-uracil plus leucovorin for locally advanced rectal cancer with pharmacogenetic analysis. Radiat Oncol 2017; 12:62. [PMID: 28347333 PMCID: PMC5369180 DOI: 10.1186/s13014-017-0800-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 03/14/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the efficacy of a high dose of oral tegafur-uracil (400 mg/m2) plus leucovorin with preoperative chemoradiation of locally advanced rectal cancer and to explore the impact of polymorphisms of cytochrome P 2A6 (CYP2A6), uridine monophosphate synthetase (UMPS), and ATP-binding cassette B1 (ABCB1) on clinical outcome. METHODS Patients with cT3 or cT4 rectal cancer were enrolled and were given tegafur-uracil 400 mg/m2/day and leucovorin 90 mg/m2/day for 7 days a week during preoperative chemoradiation (50.4 Gy/28 fractions) in this phase II trial. Primary endpoint was pathologic complete response rate, and the secondary endpoint was to explore the association between clinical outcomes and genetic polymorphisms CYP2A6 (*4, *7, *9 and *10), UMPS G638C, and three ABCB1 genotypes (C1236T, C3435T, and G2677T). RESULTS Ninety-one patients were given study treatment, and 90 underwent surgery. Pathologic complete response was noted in 10 patients (11.1%). There was no grade 4 or 5 toxicity; 20 (22.0%) experienced grade 3 toxicities, including diarrhea (10, 11.0%), abdominal pain (2, 2.2%), and anemia (2, 2.2%). Relapse-free survival and overall survival at 5 years were 88.6% and 94.2%, respectively. Patients with the UMPS 638 CC genotype experienced significantly more frequent grade 2 or 3 diarrhea (p for trend = 0.018). CONCLUSIONS Preoperative chemoradiation with tegafur-uracil 400 mg/m2/day with leucovorin was feasible, but did not meet the expected pathologic complete response rate. The UMPS 638 CC genotype might be a candidate biomarker predicting toxicity in patients receiving tegafur-uracil/leucovorin-based preoperative chemoradiation for locally advanced rectal cancer. TRIAL REGISTRATION ISRCTN11812525 , registered on 25 July 2016. Retrospectively registered.
Collapse
|
244
|
5-Fluorouracil degradation rate could predict toxicity in stages II–III colorectal cancer patients undergoing adjuvant FOLFOX. Anticancer Drugs 2017; 28:322-326. [DOI: 10.1097/cad.0000000000000453] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
245
|
Schirripa M, Procaccio L, Lonardi S, Loupakis F. The role of pharmacogenetics in the new ESMO colorectal cancer guidelines. Pharmacogenomics 2017; 18:197-200. [DOI: 10.2217/pgs-2016-0191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Marta Schirripa
- Oncologia Medica 1, Istituto Oncologico Veneto, IRCCS Padova, Italy
| | | | - Sara Lonardi
- Oncologia Medica 1, Istituto Oncologico Veneto, IRCCS Padova, Italy
| | - Fotios Loupakis
- Oncologia Medica 1, Istituto Oncologico Veneto, IRCCS Padova, Italy
| |
Collapse
|
246
|
Quaranta S, Thomas F. Pharmacogenetics of anti-cancer drugs: State of the art and implementation - recommendations of the French National Network of Pharmacogenetics. Therapie 2017; 72:205-215. [PMID: 28262261 DOI: 10.1016/j.therap.2017.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 12/27/2022]
Abstract
Individualized treatment is of special importance in oncology because the drugs used for chemotherapy have a very narrow therapeutic index. Pharmacogenetics may contribute substantially to clinical routine for optimizing cancer treatment to limit toxic effects while maintaining efficacy. This review presents the usefulness of pharmacogenetic tests for some key applications: dihydropyrimidine dehydrogenase (DPYD) genotyping for fluoropyrimidine (5-fluorouracil, capecitabine), UDP glucuronosylstransferase (UGT1A1) for irinotecan and thiopurine S-methyltransferase (TPMT) for thiopurine drugs. Depending on the level of evidence, the French National Network of Pharmacogenetics (RNPGx) has issued three levels of recommendations for these pharmacogenetic tests: essential, advisable, and potentially useful. Other applications, for which the level of evidence is still discussed, will be evoked in the final section of this review.
Collapse
Affiliation(s)
- Sylvie Quaranta
- Service de pharmacocinétique et toxicologie, laboratoire de biologie médicale, hôpital de la Timone, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Fabienne Thomas
- Institut Claudius-Regaud, CRCT, Université de Toulouse, Inserm, UPS, 31059 Toulouse, France; GPCO-Unicancer, 101, rue de Tolbiac, 75013 Paris, France.
| |
Collapse
|
247
|
Glaire MA, Brown M, Church DN, Tomlinson I. Cancer predisposition syndromes: lessons for truly precision medicine. J Pathol 2017; 241:226-235. [PMID: 27859368 DOI: 10.1002/path.4842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 02/11/2024]
Abstract
Cancer predisposition syndromes are typically uncommon, monogenic, high-penetrance disorders. Despite their rarity, they have proven to be highly clinically relevant in directing cancer prevention strategies. As such, they share notable similarities with an expanding class of low-frequency somatic mutations that are associated with a striking prognostic or predictive effect in the tumours in which they occur. In this review, we highlight these commonalities, with particular reference to mutations in the proofreading domain of replicative DNA polymerases. These molecular phenotypes may occur as either germline or somatic events, and in the latter case, have been shown to confer a favourable prognosis and potential increased benefit from immune checkpoint inhibition. We note that incorporation of these variants into clinical management algorithms will help refine patient management, and that this will be further improved by the inclusion of other germline variants, such as those that determine the likelihood of benefit or toxicity from anti-neoplastic therapy. Finally, we propose that such integrated patient and tumour profiling will be essential if we are to deliver truly precision medicine for cancer patients, but in a similar way to rare germline mutations, we must ensure that we identify and utilize rare somatic mutations with strong predictive and prognostic effects. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mark A Glaire
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Matthew Brown
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - David N Church
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| |
Collapse
|
248
|
Lunenburg C, Swen JJ, Guchelaar HJ, Gelderblom H. Capecitabine-Induced Severe Toxicity Secondary to DPD Deficiency and Successful Treatment with Low Dose 5-Fluorouracil. J Gastrointest Cancer 2016; 48:117-118. [PMID: 28025815 DOI: 10.1007/s12029-016-9908-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Catc Lunenburg
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, Post zone C7-P, 2300 RC, Leiden, The Netherlands
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - H-J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, Post zone C7-P, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
249
|
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
|
250
|
Botticelli A, Borro M, Onesti CE, Strigari L, Gentile G, Cerbelli B, Romiti A, Occhipinti M, Sebastiani C, Lionetto L, Marchetti L, Simmaco M, Marchetti P, Mazzuca F. Degradation Rate of 5-Fluorouracil in Metastatic Colorectal Cancer: A New Predictive Outcome Biomarker? PLoS One 2016; 11:e0163105. [PMID: 27656891 PMCID: PMC5033390 DOI: 10.1371/journal.pone.0163105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/04/2016] [Indexed: 12/12/2022] Open
Abstract
Background 5-FU based chemotherapy is the most common first line regimen used for metastatic colorectal cancer (mCRC). Identification of predictive markers of response to chemotherapy is a challenging approach for drug selection. The present study analyzes the predictive role of 5-FU degradation rate (5-FUDR) and genetic polymorphisms (MTHFR, TSER, DPYD) on survival. Materials and Methods Genetic polymorphisms of MTHFR, TSER and DPYD, and the 5-FUDR of homogenous patients with mCRC were retrospectively studied. Genetic markers and the 5-FUDR were correlated with clinical outcome. Results 133 patients affected by mCRC, treated with fluoropyrimidine-based chemotherapy from 2009 to 2014, were evaluated. Patients were classified into three metabolic classes, according to normal distribution of 5-FUDR in more than 1000 patients, as previously published: poor-metabolizer (PM) with 5-FU-DR ≤ 0,85 ng/ml/106 cells/min (8 pts); normal metabolizer with 0,85 < 5-FU-DR < 2,2 ng/ml/106 cells/min (119 pts); ultra-rapid metabolizer (UM) with 5-FU-DR ≥ 2,2 ng/ml/106 cells/min (6 pts). PM and UM groups showed a longer PFS respect to normal metabolizer group (14.5 and 11 months respectively vs 8 months; p = 0.029). A higher G3-4 toxicity rate was observed in PM and UM, respect to normal metabolizer (50% in both PM and UM vs 18%; p = 0.019). No significant associations between genes polymorphisms and outcomes or toxicities were observed. Conclusion 5-FUDR seems to be significantly involved in predicting survival of patients who underwent 5-FU based CHT for mCRC. Although our findings require confirmation in large prospective studies, they reinforce the concept that individual genetic variation may allow personalized selection of chemotherapy to optimize clinical outcomes.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), “Sapienza” University of Rome, Rome, Italy
| | | | - Lidia Strigari
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy
| | | | - Bruna Cerbelli
- Department of Radiological Oncological and Pathological Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Adriana Romiti
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
| | | | | | | | - Luca Marchetti
- Department of Medical Oncology, Policlinico Umberto I, Rome, Italy
| | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), “Sapienza” University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Medical Oncology Unit, Sant’Andrea Hospital, Rome, Italy
- Istituto Dermopatico dell’Immacolata-IRCCS, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Medical Oncology Unit, Sant’Andrea Hospital, Rome, Italy
| |
Collapse
|