www.journalononcology.org

Research Article

Open Access, Volume 3

Fluoropyrimidine Dose Based on Uracil Plasma Concentration Impairs Survival in Patients

Khalil Hodroj1; Thomas Walter2,7; Sophie Testu8; Céline Verot8; David Barthelemy1,2; Jean-Christophe Lega3,9; Guillaume Grenet5,9; Marielle Guillet10; Marion Chauvenet11; Jerome Guitton1,6; Léa Payen-Gay1,2,4*

1Laboratory of Biochemistry, Molecular Biology and Pharmacotoxicology, Groupe Hospitalier Sud, Hospices Civils de Lyon, 69495 Pierre- Benite, France.
2Hospices Civils de Lyon Cancer institute, Circulating Cancer (CIRCAN) Program, 69495 Pierre-Benite, France.
3Hospices Civils de Lyon, Internal and Vascular Medicine Department, Lyon Sud Hospital, 69310 Pierre-Benite, France.
4Therapeutic Targeting in Oncology, Faculty of Medicine Lyon-Sud, University Lyon 1, University of Lyon, 69600 Oullins, France.
5Hospices Civils de Lyon, Pole Sante Publique, University Hospital Department of Pharmacotoxicology, 69002 Lyon, France.
6Lyon Cancer Research Center - Ribosome, Translation and Cancer; UMR INSERM 1052 CNRS 5286, 69008 Lyons, France.
7Hospices Civils de Lyon, Medical Oncology Department, Edouard Herriot Hospital, 69003, Lyon, France.
8North-West Hospital of Villefranche-sur-Soane, 69400, Gleize, France.
9Universite de Lyon, University Lyon 1, Team Evaluation and Modeling of Therapeutic Effects, Laboratory of Biology and Evolutionary Biometry, UMR CNRS 5558, 69008 Lyon, France.
10Hospices Civils de Lyon, Hepatogastroenterology Department, Croix-Rousse Hospital, 69004 Lyon, France.
11Hospices Civils de Lyon, Hepatogastroenterology Department, Lyon Sud Hospital, 69310 Pierre-Benite, France.

Abstract

Fluoropyrimidine drugs (5-flurouracil (5-FU) / capecitabine) are the backbone of therapeutic regimens for digestive carcinomas and can lead to severe toxicities. To lower that risk, the French health authorities recommend dose adaptation based on plasma uracil concentration. Since dihydro pyrimidine dehydrogenase (DPD) catabolises more than 80% of 5-FU, a uracil threshold of ³16 ng/mL has been considered as partial DPD deficiency. 5-FU displays a dose-response relationship regarding both its efficacy and its toxicity we retrospectively assessed how this guideline has been applied in routine practice by evaluating fluoropyrimidine dosage and survival rate according to the 16 ng/mL uracil concentration threshold. Patients were included in this multicentre retrospective study if they had digestive cancer and a plasma uracil quantification performed between February 2018 and January 2020, and if they received at least one cycle of fluoropyrimidine-based chemotherapy in one of the four participating oncology departments. Among 302 patients included, 71 (23.5%) had a plasma uracil concentration ≥16 ng/mL. For the latter, the fluoropyrimidine was 0-50% of the theoretical dose in 60.5% of patients, 51-75% in 15.5%, and 76-100% in 24% at cycle 1 of treatment and the dose was increased after a well-tolerated first cycle for 7/69 (10.1%) patients at cycle 2 and for 13/69 (18.8%) patients from baseline at cycle 4. Median time to failure was 8.6 months in those with uracil ³16 ng/ mL and 24.3 months in the others; hazard ratio (HR) 0.38, 95% confidence interval (CI) [0.27; 0.53], p<0.0001. Overall survival was, respectively, 15.8 and 39.9 months; HR 0.46, 95%CI [0.29; 0.74], p=0.001). Tailored fluoropyrimidine dose impaired survival in patients with uracil ³16 ng/mL and we should consider to increase more frequently the fluoropyrimidine dose administered after a well-tolerated first cycle.

Keywords: Digestive carcinomas; Fluoropyrimidine dosage; Uracil; Toxicity; Predictive biomarker; 5-FU (5-fluorouracil).

Abbreviations: DPD: Dihydro Pyrimidine Dehydrogenase; EMA: European Medicines Agency; HPLC: High Performance Liquid Chromatography.

Manuscript Information: Received: Mar 22, 2023; Accepted: Apr 26, 2023; Published: May 03, 2023

Correspondance: Léa Payen-Gay, Laboratory of Biochemistry, Molecular Biology and Pharmacotoxicology, Groupe Hospitalier Sud, Hospices Civils de Lyon, 69495 Pierre-Benite, France. Tel: + 33-478864401; Email: Lea.payen-gay@chu-lyon.fr

Citation: Hodroj K, Walter T, Testu S, Verot C, Barthelemy D, et al. Fluoropyrimidine Dose Based on Uracil Plasma Concentration Im- pairs Survival in Patients. J Oncology. 2023; 3(1): 1087.

Copyright: © Payen-Gay L 2023. Content published in the journal follows creative common attribution license.

Introduction

Digestive carcinoma chemotherapy regimens are mostly based on fluoropyrimidine drugs (5-fluorouracil [5-FU] or capecitabine) [1-5]. However, 5-FU is mainly catabolised by dihydro pyrimidine dehydrogenase (DPD) [6], and partial or complete DPD activity deficiency can cause severe adverse reactions including death [7].

Different strategies have been proposed to predict DPD activity deficiency; the two main approaches are phenotyping the enzyme activity (directly or indirectly), or genotyping the four main polymorphisms of DPYD gene associated with 5-FU toxicity [8-12]. In February 2018, the French medicines agency (Agence nationale de sécuritédu médicament et des produits de santé) recommended DPYD genotyping for all patients receiving a fluoropyrimidine-based treatment to improve its safety as did later the European Medicines Agency (EMA) [13] and other pharmacogenetics working group. In contrast, the US Food and Drug Administration chose not to require any regulatory review of laboratory or genetic tests for use of 5-FU [14]. In December 2018 a new guideline from the French cancer institute (Institut National Du Cancer, InCA) and the French health authority (Haute Autorité de Santé, HAS) recommended the measurement of the plasma uracil concentration, and, based on a consensus, dose adaptation is required if this uracil level is between 16 and 150 ng/mL while another drug should be considered if it is greater than 150 ng/ mL [15]. The aim was that phenotyping DPD activity could avoid severe adverse reactions due to unknown DPYD variants that impair DPD activity [16,17].

To our knowledge, no evaluation of this guideline in real-life practice has been reported, which is of importance since 5-FU displays a dose-response relationship regarding both its efficacy and its toxicity [18,19]. To address that matter, we conducted a retrospective study to evaluate how fluoropyrimidine dosage was adapted to uracil concentration and its impact on patient outcomes.

Materials and methods

Patients and study design

Patients were included in this multicentre retrospective study if they had digestive cancer and an plasma uracil quantification performed between February 2018 and January 2020, and if they received at least one cycle of fluoropyrimidine-based chemotherapy in one of the four participating oncology departments (Hôpital Edouard Heriot [Lyon], Centre Hospitalier de Lyon Sud [Lyon], Hôpital de la Croix Rousse [Lyon], Hôpital Nord-Ouest de Villefranche-sur-Saone [Gleize]). The objective was to compare time to failure (TTF) and Overall Survival (OS) among those with uracil <16 ng/ml to those with uracil ≥16 ng/ml. The following characteristics were collected from the patient medical files: histology, stage (localised vs metastatic disease), chemotherapy regimen, proportion of fluoropyrimidine dose administered, fluoropyrimidine induced-toxicity, date of progression and that of death (or last follow-up). The proportion of fluoropyrimidine dose administered and adverse reactions of fluoropyrimidine were assessed at cycle 1, 2 and 4 for those with uracil ³16 ng/mL to characterise early (1st and 2nd cycles) and longterm (4th cycle) dose adaptation. Last active search for vital status was March 30th 2021.

This is a non-interventional study and conducted according to the guidelines of the Declaration of Helsinki, and registered by the national data protection committee (Commission nationale de l’informatique et des libertés [CNIL] in March 2021, number 21_5368).

DPD phenotyping

Plasma uracil concentration was quantified by high performance liquid chromatography (HPLC) coupled with high resolution mass spectrometry detection [20]. The results were analysed by a senior biologist and the results of plasma uracil concentration were available to clinicians within 8 to 10 days from initial patient blood sample before the administration of treatment.

Statistical analysis

Data were described using median [interquartile range, IQR] and mean (standard deviation, SD) for continuous variables, and frequencies (percentage) for categorical variables. TTF and progression-free survival (PFS) was defined as the time from the first treatment with 5-FU / capecitabine to death or morphological progression according to RECIST criteria or clinical progression requiring a new anti-tumour treatment, whichever occurred first. PFS was used for metastatic disease only. OS was defined as the time from the first treatment with 5-FU / capecitabine to death or last follow-up. Patients without these events were censored at the time of last follow-up. TTF, PFS (for metastatic disease) and OS were estimated using the Kaplan-Meier method. Univariate analyses were performed using the Log-rank test for each variable of interest. Multivariate analyses using a Cox proportional hazards regression model were performed to identify factors independently associated with prognosis. All significant factors from the univariate analysis (Log-rank p<0.10) were included in the multivariate analyses; p<0.05 was considered statistically significant. The results from the survival analyses are presented with the effect estimates, hazard ratios (HR), and 95% confidence interval (CI). All statistical analyses were performed using IBM- SPSS version 21.

Results

Patient characteristics

We identified a series of 334 patients with digestive cancers with a known plasma uracil concentration; 32 were excluded for missing data or lack of treatment with fluoropyrimidine (Figure 1).

Figure 1: Study flow chart.

Patients with a plasma uracil concentration ≥16 ng/mL represented 23.5 % (71/302) of the total population. The two most frequent digestive cancers were colorectal adenocarcinoma and pancreatic adenocarcinoma; there was no significant difference between groups except for the prevalence of squamous cell carcinoma of oesophagus/anus (Table 1).

Fluoropyrimidine dose management of and toxicity evaluation Among those with plasma uracil ³16 ng/mL, at cycle 1 continuous 5-FU or capecitabine dose was 0-50% of the theoretical dose in 60.5% of patients, 51-75% in 15.5%, and 76-100% in 24%; FU bolus was administered to 13.2% (9/68) of patients. Grade 3 or 4 fluoropyrimidine toxicity was observed in 2.8% of patients (2/71) after cycle 1 (Table 2). Fluoropyrimidine increased dose after a well-tolerated first cycle was observed for 7/69 (10.1%) patients at cycle 2 and for 13/69 (18.8%) patients at cycle 4. Among patients with plasma uracill <16 ng/mL, at cycle 1 the full dose of continuous 5-FU or capecitabine was administered to 97.4% of patients and 98.1% of patients received a 5-FU bolus. Grade 3 or 4 toxicity was experienced by 17 (7.4%) patients (Table 2).

Survival analysis

The median TTF was estimated to be 8.6 months among those with plasma uracil ³16 ng/mL, and 15.8 months among those with uracil <16 ng/mL; the risk of tumour progression was significantly higher among those with plasma uracil ³16 ng/mL (HR 0.38, 95%CI [0.27; 0.53], p< 0.0001). The median OS was estimated to be 24.3 months among those with plasma uracil ³16 ng/mL, and 39.9 months among those with uracil <16 ng/mL; the risk of death was significantly higher among those with uracil ³16 ng/mL (HR 0.46, 95%CI [0.29; 0.74], p=0.001; Figure 2).

Table 1: Patient characteristics according to plasma uracil concentration.
Uracil >_16 ng/mL Uracil <16 ng/mL P value
Number of patients 71 231
Median age (range) 70.3 (49.7-86.2) 67.5 (28-91)
Male 44 (62%) 144 (62.3%) 0.96
Cancer type
Colorectal ADK 29 (40.9%) 119 (51.5%) 0.11
Pancreatic ADK 20 (28.1%) 46 (19.9%) 0.14
SCC of oesophagus / anus 11 (15.5%) 10 (4.3%) 0.001
Gastric ADK 4 (5.6%) 31 (13.4%) 0.07
Neuroendocrine tumour 4 (5.6%) 15 (6.5%) 0.79
Other 3 (4.2%) 10 (4.3%) 0.97
Cancer stage
Localised 25 (34.7%) 81 (35.1%) 0.96
Metastatic 47 (65.3%) 150 (64.9%) 0.96

ADK: Adenocarcinoma; SCC: Squamous cell carcinoma.

Table 2: Proportion of fluoropyrimidine dose administered and adverse reactions of fluoropyrimidine based on plasma uracil concentration.
Uracil <16 ng/mL Uracil >16 ng/mL
Cycle 1 (n=7) Cycle 2 (n=67) Cycle4 (n= 67) Cycle 1 (n=231)
% of continuous 5-FU or Capecitabine, n(%)
0-50% 43 (60.5) 36 (53.7) 36 (53.7) 2 (0.9)
51-75% 11 (15.5) 14 (20.4) 17 (25.4) 4 (1.7)
76-100% 17 (24) 17 (25.4) 14 (20.9) 225 (97.4)
Patients with a bolus of 5FU 9/68 (13.1) 9 (13.1) 7 (10.1) 207/211 (98.1)
G3 or G4 loxicity 2 (2.8) 0 0 17(7.4)
Patients with an increased dose after well-tolerated first cycle - 7/69(10.1) 13/69 (18.8) -
Figure 2: Kaplan-Meier curves of time to failure (A) and overall survival (B) according to plasma uracil concentration. The number of patients at risk are noted under each graph. Median TTF and median OS were estimated to be, respectively 8.6 and 24.3 months among those with uracil ³16 ng/mL and 15.8 and 39.9 months among those with uracil <16 ng/mL; the risk of failure and that of death was significantly higher among those with uracil ³16 ng/mL (PFS: HR 0.38, 95%CI [0.27; 0.53], p <0.0001; OS: HR 0.46, 95%CI [0.29-0.74], p=0.001).

For patients with metastatic colorectal adenocarcinoma, the median PFS and median OS were estimated to be, respectively 8.6 and 25.7 months among those with uracil ³16 ng/mL and 14.9 and 40.1 months among those with uracil <16 ng/mL; the risk of progression and that of death was significantly higher among those with uracil ³16 ng/mL (PFS: HR 0.26, 95%CI [0.11; 0.59], p <0.0001; OS: HR 0.29, 95%CI 0.09-0.97, p=0.02, 174 Figure 3).


Figure 3: Kaplan-Meier curves of time to failure (A) and overall survival (B) according to plasma uracil concentration. The number of patients at risk are noted under each graph. Median TTF and median OS were estimated to be, respectively 8.6 and 24.3 months among those with uracil ³16 ng/mL and 15.8 and 39.9 months among those with uracil <16 ng/mL; the risk of failure and that of death was significantly higher among those with uracil ³16 ng/mL (PFS: HR 0.38, 95%CI [0.27; 0.53], p <0.0001; OS: HR 0.46, 95%CI [0.29-0.74], p=0.001).

In multivariate analysis, the factors significantly associated with TTF and OS were colorectal adenocarcinoma vs non-colorectal adenocarcinoma (HR for TTF 0.64, 95%CI [0.48; 0.86], p<0.003 and HR for OS 0.34, 95%CI [0.21; 0.54], p<0.0001), localised vs metastatic cancer (HR for TTF 0.25, 95%CI [0.17; 0.36], p<0.0001 and HR for OS 0.26, 95%IC A B[0.14; 0.48], p<0.0001 ) and full administrated fluoropyrimidine dose vs tailored (HR for TTF 0.14, 95%CI 0.06-0.34 and HR for OS 0.24, (95%IC [0.11; 0.55], p=0.001; Tables 3 & 4).

Table 3: Factors associated with time to failure (TTF).
Univariate analysis Multivariate analysis
Hazard ratio [95% CI] P value Hazard ratio [95% CI] P value
Age, < vs ³ median 0.8 [0.60; 1.06] 0.13
Sex, female vs male 0.92 [0.69; 1.23] 0.59
colorectal adenocarcinoma vs 0.72 [0.54; 0.96] 0.03 0.64 [0.48; 0.86] 0.003
Non-colorectal adenocarcinoma
Localised vs metastatic cancer 0.26 [0.18; 0.38] <0.0001 0.25 [0.17; 0.36] <0.0001
Fluoropyrimidine dose (full dose vs tailored*) 0.23 [0.17; 0.33] <0.0001 0.14 [0.06; 0.34] <0.0001
Plasma uracil concentration (<16 vs ³ 16) 0.38 [0.27; 0.53] <0.0001 1.79 [0.74; 4.29] 0.193
Table 4: Factors associated with overall survival (OS).
Univariate analysis Multivariate analysis
Hazard ratio [95% CI] P value Hazard ratio [95% CI] P value
Age, < vs ³ median 0.63 [0.41; 0.96] 0.03 0.53 [0.34; 0.82] 0.004
Sex, female vs male 0.88 [0.57; 1.38] 0.59
Colorectal adenocarcinoma vs non-colorectal adenocarcinoma 0.38 [0.25; 0.60] <0.0001 0.34 [0.21; 0.54] <0.0001
Localised vs metastatic cancer 0.29 [0.16; 0.54] <0.0001 0.26 [0.14; 0.48] <0.0001
Fluoropyrimidine dose (full dose vs tailored* ) 0.28 [0.18; 0.45] <0.0001 0.24 [0.11; 0.55] 0.001
Plasma uracil concentration (<16 vs ³ 16) 0.46 [0.29; 0.74] 0.001 1.66 [0.73; 3.76] 0.227

Discussion

In this retrospective cohort, we found that a tailored fluoropyrimidine dose impaired OS in patients with uracil ³16 ng/mL in routine practice. Univariate analysis found that those with uracil ³16 ng/mL and those with decreased fluoropyrimidine dose had a worse survival. As in multivariate analysis plasma uracil concentration was not associated to a worse prognosis, impaired survival in the plasma uracil ³16 ng/mL group was due to decreased chemotherapy dosage in this population. By applying the French recommendations in patients with uracil ³16 ng/mL, only 2.8 % of patients herein experienced G3 or G4 toxicity at cycle 1 but from baseline treatment dose was increased for only 18.8% of patients at cycle 4. The present study highlights that we should consider to increase more frequently the fluoropyrimidine dose administered after a well-tolerated first cycle. The results of the study also emphasise that plasma uracil is not a prognostic factor but that chemotherapy treatment displays a dose-effectiveness relationship as described in the literature [18]. However, many severe toxicities induced by fluoropyrimidine can be explained by partial DPD deficiency and complete DPD deficiency can lead to death [8,11,21]. To improve the identification of patients at high risk of toxicity, a combined composite biomarker should be proposed based on both phenotyping and genotyping of the DPYD gene. The latter is another way to evaluate DPD activity [22] and the reported experience of systematic genotyping DPYD gene in reallife practice indicated that the administration of 5-FU at reduced dose in patients heterozygous for DPYD*2A is safe [23]. The limitation of this technique is that in current clinical practice only four variants are tested for 5-FU toxicity (DPYD*2A, DPYD*13, D949V and HapB3), the use of which has failed to predict all cases of DPD deficiency – possibly because other genes are implicated in 5-FU toxicity and efficacy such as MTHFR, ABCB1 or TYMS [24,25,26]. Furthermore, to our knowledge, the impact of such testing in routine practice on survival has yet to be reported; only the impact on toxicity has been published [26,27]. The main limitation of the present study is its retrospective design that is associated with a risk of confusion bias. In addition, the participating centres are located in one administrative area of France, which may limit the generalizability of the results. In the future, a model that associates phenotyping DPD and genotyping DPYD with other genes of interest may be useful to better predict fluoropyrimidine toxicity and also to better adapt chemotherapy dosage.

Conclusions

The present study highlights that tailored fluoropyrimidine dose impaired survival in patients with uracil 16 ng/mL and we should consider to increase more frequently the fluoropyrimidine dose administered after a well-tolerated first cycle. These results should be confirmed by evaluating the clinical practice in the whole French territory.

Declarations

Funding: This research received no external funding.

Institutional review board statement: This is a non-interventional study and conducted according to the guidelines of the Declaration of Helsinki, and registered by the CNIL in March 2021, number 21_5368.

Informed consent statement: Informed consent was obtained from all subjects involved in the study.

Data availability statement: To protect patient confidentiality, the data will be consider for sharing only on written requests and on case-by-case basis.

Acknowledgments: We thank Philip Robinson (DRS, Hospices Civils de Lyon, Lyon, France) for help in manuscript preparation.

Conflicts of interest: The authors declare no conflict of interest.

Highlights

- Patients with uracil 16 ng/mL are treated with reduced doses of fluoropyrimidine

- Fluoropyrimidine dose is not always increased after a well-tolerated first cycle

- Patients with a reduced fluoropyrimidine dose have worse survival

References

  1. André T, Meyerhardt J, Iveson T, Sobrero A, Yoshino T, et al. Effect of Duration of Adjuvant Chemotherapy for Patients with Stage III Colon Cancer (IDEA Collaboration): Final Results from a Prospective, Pooled Analysis of Six Randomised, Phase 3 Trials. Lancet Oncol. 2020; 21: 1620-1629.
  2. Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, et al. FOL- FIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer. N Engl J Med. 2011; 364: 1817-1825.
  3. Cremolini C, Loupakis F, Antoniotti C, Lupi C, Sensi E, et al. FOLFOXIRI plus Bevacizumab versus FOLFIRI plus Bevacizumab as First-Line Treatment of Patients with Metastatic Colorectal Cancer: Updated Overall Survival and Molecular Subgroup Analyses of the Open-Label, Phase 3 TRIBE Study. Lancet Oncol. 2015; 16: 1306-1315.
  4. Lemelin A, Barritault M, Hervieu V, Payen L, Péron J, et al. O6-Methylguanine-DNA Methyltransferase (MGMT) Status in Neuroendocrine Tumors: A Randomized Phase II Study (MGMT-NET). Digestive and Liver Disease. 2019; 51: 595-599.
  5. Al-Batran SE, Homann N, Pauligk C, Goetze TO, Meiler J, et al. Perioperative Chemotherapy with Fluorouracil plus Leucovorin, Oxaliplatin, and Docetaxel versus Fluorouracil or Capecitabine plus Cisplatin and Epirubicin for Locally Advanced, Resectable Gastric or Gastro-Oesophageal Junction Adenocarcinoma (FLOT4): A Randomised, Phase 2/3 Trial. Lancet. 2019; 393: 1948-1957.
  6. Heggie GD, Sommadossi JP, Cross DS, Huster WJ, Diasio RB. Clinical Pharmacokinetics of 5-Fluorouracil and Its Metabolites in Plasma, Urine, and Bile. Cancer Res. 1987; 47: 2203-2206.
  7. Madi A, Fisher D, Maughan TS, Colley JP, Meade AM, et al. 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.
  8. Fleming RA, Milano G, Thyss A, Demani F, Renà N. Correlation between Dihydropyrimidine Dehydrogenase Activity in Peripheral Mononuclear Cells and Systemic Clearance of Fluorouracil in Cancer Patients. Cancer Res. 1992; 52: 2899-2902.
  9. Gamelin E, Boisdron-Celle M, Guérin-Meyer V, Delva R, Lortholary A, et al. Correlation Between Uracil and Dihydrouracil Plasma Ratio, Fluorouracil (5-FU) Pharmacokinetic Parameters, and Tolerance in Patients With Advanced Colorectal Cancer: A Potential Interest for Predicting 5-FU Toxicity and Determining Optimal 5-FU Dosage. Journal of Clinical Oncology. 1999; 17: 1105.
  10. Lu Z, Zhang R, Diasio RB. Dihydropyrimidine Dehydrogenase Activity in Human Peripheral Blood Mononuclear Cells and Liver: Population Characteristics, Newly Identified Deficient Patients, and Clinical Implication in 5-Fluorouracil Chemotherapy. Cancer Res. 1993; 53: 5433-5438.
  11. Amstutz U, Farese S, Aebi S, Largiadèr CR. Dihydropyrimidine Dehydrogenase Gene Variation and Severe 5-Fluorouracil Toxicity: A Haplotype Assessment. Pharmacogenomics. 2009; 10: 931-944.
  12. Meulendijks D, Cats A, Beijnen JH, Schellens JHM. Improving Safety of Fluoropyrimidine Chemotherapy by Individualizing Treatment Based on Dihydropyrimidine Dehydrogenase Activity – Ready for Clinical Practice? Cancer Treat Rev. 2016; 50: 23-34.
  13. Fluorouracil-Fluorouracil-Related-Substances-Article-31-Referral-Assessment-Report_en.Pdf.
  14. Health, C. for D. and R. Table of Pharmacogenetic Associations. FDA 2020.
  15. Recherche de Déficit En Dihydropyrimidine Déshydrogenase EnVue de Prévenir Certaines Toxicités Sévères Survenant Sous Traitement Comportant Des Fluoropyrimidines (5-Fluorouracile), Recommandations et Référentiels, INCa, HAS, 2018.
  16. Cosmic DPYD Gene – COSMIC.
  17. Etienne-Grimaldi MC, Boyer JC, Beroud C, Mbatchi L, van Kuilenburg A, et al. New Advances in DPYD Genotype and Risk of Severe Toxicity under Capecitabine. PLoS One. 2017; 12.
  18. Kuilenburg ABPV, Lenthe H. van, Blom MJ, Mul EPJ, Gennip AHV. Profound Variation in Dihydropyrimidine Dehydrogenase Activity in Human Blood Cells: Major Implications for the Detection of Partly Deficient Patients. Br J Cancer. 1999; 79: 620-626.
  19. Hodroj K, Barthelemy D, Lega JC, Grenet G, Gagnieu MC, et al. Issues and Limitations of Available Biomarkers for Fluoropyrimidine-Based Chemotherapy Toxicity, a Narrative Review of the Literature. ESMO Open. 2021; 6: 100125.
  20. Tafzi N, Woillard JB, Fleytoux A, Picard N, Marquet P. Phenotyping of Uracil and 5-Fluorouracil Metabolism Using LC-MS/MS for Prevention of Toxicity and Dose Adjustment of Fluoropyrimidines. Ther Drug Monit. 2020; 42: 540-547.
  21. Kuilenburg ABP, van Haasjes J, Richel DJ, Zoetekouw L, Lenthe HV, Abreu, R.A.D.; Maring, J.G.; Vreken, P.; Gennip, A.H. van Clinical Implications of Dihydropyrimidine Dehydrogenase (DPD) Deficiency in Patients with Severe 5-Fluorouracil-Associated Toxicity: Identification of New Mutations in the DPD Gene. Clin Cancer Res. 2000; 6: 4705-4712.
  22. Wei X, Elizondo G, Sapone A, McLeod HL, Raunio H, et al. Characterization of the Human Dihydropyrimidine Dehydrogenase Gene. Genomics. 1998; 51: 391-400.
  23. Jolivet C, Nassabein R, Soulières D, Weng X, Amireault C, et al. Implementing DPYD*2A Genotyping in Clinical Practice: The Quebec, Canada, Experience. Oncologist. 2021; 26: e597-e602.
  24. Lecomte T, Ferraz JM, Zinzindohoué F, Loriot MA, Tregouet DA, et al. Thymidylate Synthase Gene Polymorphism Predicts Toxicity in Colorectal Cancer Patients Receiving 5-Fluorouracil-Based Chemotherapy. Clin Cancer Res. 2004; 10: 5880-5888.
  25. Gonzalez-Haba E, García MI, Cortejoso L, López-Lillo C, Barrueco N, et al. ABCB1 Gene Polymorphisms Are Associated with Adverse Reactions in Fluoropyrimidine-Treated Colorectal Cancer Patients. Pharmacogenomics. 2010; 11: 1715-1723.
  26. Nahid NA, Apu MNH, Islam R, Shabnaz S, Chowdhury SM, et al. DPYD*2A and MTHFR C677T Predict Toxicity and Efficacy, Respectively, in Patients on Chemotherapy with 5-Fluorouracil for Colorectal Cancer. Cancer Chemother Pharmacol. 2018; 81: 119-129.
  27. Hishinuma E, Narita Y, Saito S, Maekawa M, Akai F, et al. Functional Characterization of 21 Allelic Variants of Dihydropyrimidine Dehy-drogenase Identified in 1070 Japanese Individuals. Drug Metab.Dispos. 2018; 46: 1083-1090.
  28. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JHM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther. 2018; 103: 210-216.