Skip to main content

Pharmacogenetics of DPYD and treatment-related mortality on fluoropyrimidine chemotherapy for cancer patients: a meta-analysis and trial sequential analysis

Abstract

Background

Fluoropyrimidines are chemotherapy drugs utilized to treat a variety of solid tumors. These drugs predominantly rely on the enzyme dihydropyrimidine dehydrogenase (DPD), which is encoded by the DPYD gene, for their metabolism. Genetic mutations affecting this gene can cause DPYD deficiency, disrupting pyrimidine metabolism and increasing the risk of toxicity in cancer patients treated with 5-fluorouracil. The severity and type of toxic reactions are influenced by genetic and demographic factors and, in certain instances, can result in patient mortality. Among the more than 50 identified variants of DPYD, only a subset has clinical significance, leading to the production of enzymes that are either non-functional or impaired. The study aims to examine treatment-related mortality in cancer patients undergoing fluoropyrimidine chemotherapy, comparing those with and without DPD deficiency.

Methods

The meta-analysis selected and evaluated 9685 studies from Pubmed, Cochrane, Embase and Web of Science databases. Only studies examining the main DPYD variants (DPYD*2A, DPYD p.D949V, DPYD*13 and DPYD HapB3) were included. Statistical Analysis was performed using R, version 4.2.3. Data were examined using the Mantel-Haenszel method and 95% CIs. Heterogeneity was assessed with I2 statistics.

Results

There were 36 prospective and retrospective studies included, accounting for 16,005 patients. Most studies assessed colorectal cancer, representing 86.49% of patients. Other gastrointestinal cancers were evaluated by 11 studies, breast cancer by nine studies and head and neck cancers by five studies. Four DPYD variants were identified as predictors of severe fluoropyrimidines toxicity in literature review: DPYD*2A (rs3918290), DPYD p.D949V (rs67376798), DPYD*13 (rs55886062) and DPYD Hap23 (rs56038477). All 36 studies assessed the DPYD*2A variant, while 20 assessed DPYD p.D949V, 7 assessed DPYD*13, and 9 assessed DPYDHap23. Among the 587 patients who tested positive for at least one DPYD variant, 13 died from fluoropyrimidine toxicity. Conversely, in the non-carrier group there were 14 treatment-related deaths. Carriers of DPYD variants was found to be significantly correlated with treatment-related mortality (OR = 34.86, 95% CI 13.96–87.05; p < 0.05).

Conclusions

This study improves our comprehension of how the DPYD gene impacts cancer patients receiving fluoropyrimidine chemotherapy. Identifying mutations associated with dihydropyrimidine dehydrogenase deficiency may help predict the likelihood of serious side effects and fatalities. This knowledge can be applied to adjust medication doses before starting treatment, thus reducing the occurrence of these critical outcomes.

Graphical abstract

Peer Review reports

Introduction

Fluoropyrimidines are antimetabolic agents that form the basis of cytotoxic chemotherapy for various malignancies [1, 2]. This class comprises 5-fluorouracil (5-FU) and its oral prodrug capecitabine, which are mainly used to treat gastrointestinal, breast, and head and neck tumors, either as monotherapy or in combination [3, 4]. 5-FU is metabolized by dihydropyrimidine dehydrogenase enzyme (DPD) into dihydrofluorouracil (DHFU), which is an inactive metabolite of this drug [5]. Approximately 80% of the 5-FU administered is catabolized by DPD, which is highly expressed in the liver [6, 7]. Interindividual variability in DPD enzyme activity is well established in the literature [8, 9]. Approximately 3–5% of the general population have partially reduced DPD activity, whereas 0.2% have complete DPD deficiency [10, 11]. Patients with reduced enzyme activity often exhibit severe toxicity to cancer treatment with 5-FU-based cytotoxic chemotherapy [12].

Although changes in the administration and dosages of fluoropyrimidine have had an impact on reducing treatment-related toxicities in recent years, approximately 20% of patients receiving fluoropyrimidine monotherapy will experience serious effects (grade 3 or more) during treatment [13, 14]. Grade 3 toxicities are more common among patients receiving fluoropyrimidine in combination, affecting up to 56% of these patients [15, 16]. Deaths related to the administration of this drug (grade 5 toxic events) are rare events during treatment, accounting for approximately 1% of all cases [17]. However, uncommon genetic variants in DPYD, which responsible for making the DPD enzyme through the process of transcription and translation. Previous studies identified in molecular analysis 128 polymorphisms in the DPYD that cause partial loss or total of activity of DPD [18], variants in DPYD represent factors recognized as a cause of severe or fatal fluoropyrimidine toxicity [17, 19, 20].

The DPYD gene is on chromosome 1p22, which is 843.32 kb long and has 23 exons, which are responsible for encoding DPD [21, 22]. Germline variants in DPYD are the main cause of DPD deficiency, and pathogenic variants are associated with an 8-fold increase in the risk of developing severe toxicities [23]. Four DPYD alleles are widely described as being highly associated with severe toxicities to fluoropyrimidine treatment, including rs75017182, rs55886062, rs3918290, and rs67376798 [24, 25]. The frequency of approximately 8% of these four alleles in European or North American populations has been previously described [26].

Although there has been extensive research into the risk of toxicity to fluoropyrimidine in cancer patients carrying genetic variants of DPYD, due to their rarity. A meta-analysis published in 2021 identified a significant association between allele status and treatment-related lethality, and provided estimates of lethality in carriers [27]. However, since then, numerous new studies have emerged, underscoring the need for an updated meta-analysis. Additionally, the previous meta-analysis did not address potential connections between ethnicity and DPD deficiency, an area that requires further exploration.

Therefore, we conducted a systematic review and meta-analysis to clarify the risk of death associated with the administration of a standard dose of fluoropyrimidine to patients with cancer.

Methods

Protocol and registration

This meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and registered with the Prospective International Registry of Systematic Reviews (PROSPERO) under registration number CRD42024564336.

Eligibility criteria

Studies were eligible if they met the following criteria: (1) the population consisted of solid tumor (non-hematologic) cancer patients receiving standard dose of fluoropyrimidine chemotherapy; (2) at least one patient had one of the specified DPYD variants; (3) outcomes included analysis of the risk of treatment-related mortality.

Exclusions included studies with overlapping populations, qualitative or economic reviews, opinion pieces, technical reports, guidelines, animal studies, in vitro experiments, studies lacking results, and studies not in English. Studies in which patients started treatment with reduced doses of fluoropyrimidine chemotherapy were also excluded.

Search strategy and data extraction

We systematically searched for published studies across PubMed, Cochrane Central, Embase, and Web of Science up to June 18, 2024. The search was restricted to English language papers and abstracts and conducted by two authors. Medical Subject Headings (MeSH) terms and specific syntax rules were used with Boolean connectors (OR, AND). Supplementary Table 5 provides a description of the terms used in search.

Our aim was to investigate treatment-related mortality in patients undergoing fluoropyrimidine chemotherapy, comparing those with and without DPYD deficiency. To achieve this, we identified key DPYD variants known to impact DPYD deficiency, as depicted in Supplementary Table 6.

Studies identified from databases and references were imported into Zotero (version 6.0.36) for deduplication and subsequently managed using Rayyan. Duplicate records were removed through automated and manual screening. Two authors independently assessed titles and abstracts of identified articles and extracted data based on predefined search criteria and quality assessment protocols. Any disagreements between reviewers were resolved through consensus.

Endpoints

The primary outcome of interest for a pooled analysis was the overall mortality on DPYD variants carriers attributed to fluoropyrimidines chemotherapy toxicity.

Risk of bias assessment

The quality assessment of individual observational studies utilized the Newcastle-Ottawa Scale, specifically tailored for non-randomized studies [28]. Two reviewers (A.B. and V.K.T.S) independently conducted the evaluation, resolving any discrepancies through consensus. Each study was evaluated across three main domains: selection of exposed cohorts, comparability of key factors, and assessment of outcomes including follow-up duration and adequacy. To explore potential publication bias, contour-enhanced funnel plots were visually inspected and assessed using Egger’s regression asymmetry and Begg’s rank correlation tests [29, 30].

Statistical analysis

Pertinent baseline characteristics of the sample were pooled to test the probability of their effects on outcome. Logit transformation was performed on the reported events to compute the binary outcome of interest with a 95% confidence interval (CI) using the Mantel-Haenszel method. Heterogeneity was assessed with I² and Tau². We used DerSimonian and Laird random-effect models for the primary endpoint [31]. Statistical Analysis was performed using R software, version 4.2.3.

Results

Study selection and baseline characteristics

As described in PRISMA flow diagram (Fig. 1), a total of 9,685 studies were assessed in our systematic search. After the removal of duplicates and the screening of titles or abstracts, 93 manuscripts were eligible to be thoroughly reviewed for inclusion and exclusion criteria. Finally, 36 studies, encompassing a population of 16,005 patients, formed the scope of the analysis [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67]. References for the excluded studies can be found in supplementary material Table 3.

Fig. 1
figure 1

PRISMA diagrams of included studies

The studies were divided into clinical trials and observational studies (prospectives and retrospectives), accounting for nine, 16 and 11 studies respectively. The baseline characteristics of included studies are summarized in Table 1.

Table 1 Summary characteristics of included studies. PS = prospective observational studies; RS = retrospective observational studies

The distribution by sex showed that 47% of the patients were male, 36% were female and 17% the sex was not identified, as shown in supplementary Table 4. Most studies studied groups of patients with different cancer sites, but the majority of population consisted of colorectal cancer (86.49% of the studies and 78.67% of the population), followed by other gastrointestinal cancers (29.73% of the studies and 3.09% of the population), breast cancer (24.32% of the studies and 2.55% of the population), pancreas cancer (16.22% of the studies and 1.76% of the population), head and neck cancer (13.51% of the studies and 0.35% of the population), and other types of cancer (18.92% of the studies and 0.32% of the population).

In the meta-analysis, the genetic variants of interest were identified in 587 patients, which represents 3.62% of the total population of 16,005 patients studied. Specifically, DPYD2A (rs3918290) was found in 174 patients, DPYD13 (rs55886062) in 11 patients, DPYD D949V (rs67376798) in 105 patients, and DPYD HapB3 (rs75017182) in 127 patients. The remaining patients were classified as non-carrier for these variants.

Geographically, the majority of studies in this meta-analysis were conducted in Europe (78.38%), with smaller proportions in Asia (18.92%), Americas (2.7%), and Oceania (2.7%). This European predominance posed challenges in understanding the relationship between DPYD gene variants and ethnicity, as well as their clinical implications. In the Asian population, the rs3918290 variant appeared to be five times more prevalent (4,89%) compared to the European population (0,93%). Conversely, the rs75017182 variant was more prevalent among Europeans (0.82%) than Asians (0.17%). The other variants were not identified in Asian studies, as shown in supplementary Table 5.

Overall mortality

Twenty-seven deaths attributable to fluoropyrimidine toxicity were identified across all 36 studies. Thirteen of these deaths occurred in carriers of DPYD variants of interest (Table 2), while fourteen occurred in individuals non-carrier for these variants. This represents a 36-fold higher likelihood of death among DPYD variant carriers undergoing fluoropyrimidine chemotherapy compared to the general population.

Table 2 Characteristics of grade 5 fluoropyrimidine toxicity in DPYD variants carriers. (aa) = heterozygous; (AA) = homozygous; FOLFOXIRI = folinic acid, fluorouracil (5FU), oxaliplatin and irinotecan; FOLFOX = folinic acid, fluorouracil and oxaliplatin

The most prevalent variant identified in these fatalities was rs3918290, with seven patients being heterozygous. Two of these patients also carried the rs67376798 and rs55886062 alleles. The second most common variant was rs55886062, observed in three heterozygous patients including one previously mentioned. The only patient with the rs75017182 variant was homozygous. Only one patient was provenient from an American study, being all others from europeans studies. Five of these patients used capecitabine, while all the other eight patients received combined fluoropyrimidine chemotherapy.

Statistics showed the carriers of DPYD variants was found to be significantly correlated with treatment-related mortality (OR = 34.86, 95% CI 13.96–87.05; p < 0.05)|(Fig. 2A), as shown in Fig. 2. A Z-value for a test of the null hypothesis is 7.61 with a corresponding p-value < 0,000001. Between study variation of observed effects is estimated by an I-squared value of 2% along with an absolute true effect size variance estimated by a Tau-squared value of 0,3838.

Fig. 2
figure 2

Overall mortality

Estimation of publication bias

Figure 3A shows the funnel plot of the included articles for publication bias assessment. The X-axis corresponds to the odds ratio, while the Y-axis represents the standard errors on either side of the mean effects. Each circle is representative of one study. Our results support that there seems to be a low risk of publication bias. Figure 3B shows the L’Abbé plot for comparison of studies’ effect size to index of precision for analysis of publication bias. These data suggest low variability between the confidence interval and the number of deaths counted in each study.

Fig. 3
figure 3

Funnel plot (A) and L’Abbé plot (B) for comparison of studies’ effect size to index of precision for analysis of publication bias

Quality assessment

Our main outcome showed low heterogeneity (I2 = 2%). However, when we carried out the sensitive analysis, we observed that the omission of Gross 2008, Jennings 2013, Largillier 2006, Cremolini 2017, Rosmarin 2014 resulted in a total absence of heterogeneity (I2 = 0%). In addition, the evaluation of the NOS Scale showed that most of the studies scored highly, reflecting a low risk of bias (score 8–9).

Discussion

Our systematic review and meta-analysis support that genomic alterations in the DPYD gene are associated with increased mortality among cancer patients treated with fluoropyrimidine-based cytotoxic chemotherapy. We included 36 studies, totaling 16,005 patients from clinical trials and prospective and retrospective observational studies. The gender distribution of the patients showed that 47% were men, 36% were women and in 17% the sex was not identified. Most studies focused on colorectal cancer, representing 86.49% of the studies and 78.67% of the population.

Fluoropyrimidines have a narrow therapeutic index; even at standard doses, 30% of patients are expected to experience severe toxicities such as myelosuppression, gastrointestinal effects, and hand-foot syndrome [68]. It is estimated that DPD enzyme deficiency accounts for 61% of the severe toxicities to this chemotherapy regimen, typically developing within the first 1–2 cycles of treatment [69]. The DPD enzyme comprises 23 exons on chromosome 1, and only a small number of pathogenic variants have been identified as significantly increasing toxicity. These include DPYD*2A (rs3918290), D949V (rs67376798), HapB3 (rs75017182), and DPYD*13 (rs55886062) [70].

The clinical use of identifying pathogenic variants in DPYD is based on dose adjustments to minimize toxicities, guided by the variant status in this gene [71]. Dose adjustments improve tolerance and increase the safety of prescribing fluoropyrimidines for the treatment of solid tumors [72, 73]. Two pharmacogenetics expert groups, the Clinical Pharmacogenetics Implementation Consortium (CPIC) [74] and the Dutch Pharmacogenetics Working Group (DPWG) [75], recommend clinical stratification into poor, intermediate/partial, and normal metabolizers. They suggest dose reductions of up to 50% for patients carrying any of the four described variants [76]. Dose reductions above 50% or even the omission of fluoropyrimidines are indicated for heterozygous or compound heterozygous patients [77].

The results of this meta-analysis support the guidelines by reinforcing the evidence that patients with DPD deficiency experience a higher treatment-related mortality rate when undergoing fluoropyrimidine chemotherapy. We recommend testing patients for the key variants identified in this study: DPYD2A (rs3918290), D949V (rs67376798), HapB3 (rs75017182), and DPYD13 (rs55886062). However, this meta-analysis was unable to determine whether specific populations should be tested for additional variants due to the limited number of studies involving non-Caucasian groups.

Our meta-analysis identified 587 (3.62%) patients carrying variants in DPYD2A, DPYD13, DPYD D949V, and DPYD HapB3. Additionally, most studies were conducted in Europe (78.38%), with smaller proportions in Asia (18.92%), the Americas (2.7%), and Oceania (2.7%). These epidemiological data are expected since these four variants are well-described, primarily derived from studies with Caucasian populations. The incidence of these four variants in the Caucasian population reaches 12%, and thus this European predominance may limit the understanding of genetic variants in different ethnicities [77,78,79]. Notably, the rs3918290 variant was five times more prevalent in the Asian population compared to the European one. In contrast, the rs75017182 variant was more prevalent among Europeans than Asians.

Our study identified 27 deaths attributed to fluoropyrimidine chemotherapy regimens, indicating that patients carrying variants in DPYD are at significant risk of mortality during treatment. Notably, the rs3918290 variant was the most prevalent among fatalities, followed by rs55886062 and rs75017182. Additionally, our statistical analysis suggested a significant correlation between the carriers of variants in DPYD and treatment-related mortality (OR = 34.86, 95% CI 13.96–87.05; p < 0.000001). This result reinforces that genotyping is a crucial tool for personalizing cancer treatment, especially for carriers of genetic variants in DPYD, and its use could minimize the risk of severe toxicity and mortality during cytotoxic chemotherapy treatment.

Sharma et al. (2021) conducted a meta-analysis that associated pathogenic variants in DPYD and increased the risk for treatment-related death [14]. Our meta-analysis not only reinforces these results but also provides direct evidence, through TSA, that the studies currently available are sufficient to prove the direct relationship of 4 genetic variants in DPYD and mortality from 5FU exposure, different from the previous metanalysis that didn’t use TSA, also we evaluated a different result in overall mortality compared with the previous meta-analysis and made additionally sensitivity analysis with the funnel plot and meta-regression.

This meta-analysis has some limitations. First, most of the included studies consisted of patients with colorectal cancer, which could influence the generalization of the results. Second, the majority of the studies consisted of Caucasian patients from Europe; this ethnic and geographic limitation may reflect the absence of data in other global ethnicities and also influence the generalization of our findings [80, 81]. However, the low heterogeneity (I2 = 2%) reinforces, together with trial-sequential analysis (TSA), that our meta-analysis represents convincing evidence, and the number of patients has already exceeded the required number to prove (Z-score = 5548) the association.

Conclusions

In conclusion, our systematic review and meta-analysis provide compelling evidence that genomic alterations in the DPYD gene significantly increase the risk of mortality among cancer patients undergoing fluoropyrimidine-based chemotherapy. The findings underscore the critical importance of genotyping for DPYD variants to personalize chemotherapy regimens, thereby enhancing treatment safety and efficacy. Although our analysis primarily reflects data from colorectal cancer patients and Caucasian populations, the strong correlation between DPYD variants and treatment-related mortality highlights the need for broader implementation of pharmacogenetic screening in diverse patient groups to mitigate severe toxicities and improve clinical outcomes.

Data availability

The datasets generated and/or analysed during the current study are available within the manuscript or supplementary information files.

References

  1. Scartozzi M, Maccaroni E, Giampieri R, Pistelli M, Bittoni A, Del Prete M, et al. 5-fluorouracil pharmacogenomics: still rocking after all these years? Pharmacogenomics. 2011;12:251–65.

    Article  CAS  PubMed  Google Scholar 

  2. Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer. 2003;3:330–8.

    Article  CAS  PubMed  Google Scholar 

  3. Lee JJ, Beumer JH, Chu E. Therapeutic drug monitoring of 5-fluorouracil. Cancer Chemother Pharmacol. 2016;78:447–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Miura K, Shima H, Takebe N, Rhie J, Satoh K, Kakugawa Y, et al. Drug delivery of oral anti-cancer fluoropyrimidine agents. Expert Opin Drug Deliv. 2017;14:1355–66.

    Article  CAS  PubMed  Google Scholar 

  5. Dean L, Kane M. Fluorouracil therapy and DPYD genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kattman BL, Malheiro AJ, editors. Medical Genetics Summaries. Bethesda (MD): National Center for Biotechnology Information (US); 2012 [cited 2024 Jul 8]. http://www.ncbi.nlm.nih.gov/books/NBK395610/

  6. Schmulenson E, Zimmermann N, Mikus G, Joerger M, Jaehde U. Current status and future outlooks on therapeutic drug monitoring of fluorouracil. Expert Opin Drug Metab Toxicol. 2021;17:1407–22.

    Article  CAS  PubMed  Google Scholar 

  7. Álvarez P, Marchal JA, Boulaiz H, Carrillo E, Vélez C, Rodríguez-Serrano F, et al. 5-fluorouracil derivatives: a patent review. Expert Opin Ther Pat. 2012;22:107–23.

    Article  PubMed  Google Scholar 

  8. Schneider JJ, Galettis P, Martin JH. Overcoming barriers to implementing precision dosing with 5-fluorouracil and capecitabine. Br J Clin Pharmacol. 2021;87:317–25.

    Article  CAS  PubMed  Google Scholar 

  9. van Kuilenburg ABP, Meinsma R, van Gennip AH. Pyrimidine degradation defects and severe 5-fluorouracil toxicity. Nucleosides Nucleotides Nucleic Acids. 2004;23:1371–5.

    Article  PubMed  Google Scholar 

  10. Wei Y, Yang P, Cao S, Zhao L. The combination of curcumin and 5-fluorouracil in cancer therapy. Arch Pharm Res. 2018;41:1–13.

    Article  CAS  PubMed  Google Scholar 

  11. Diasio RB, Offer SM. Testing for dihydropyrimidine dehydrogenase deficiency to individualize 5-fluorouracil therapy. Cancers (Basel). 2022;14:3207.

    Article  CAS  PubMed  Google Scholar 

  12. Pan X, Wang C, Wang F, Li P, Hu Z, Shan Y, et al. Development of 5-fluorouracil derivatives as anticancer agents. Curr Med Chem. 2011;18:4538–56.

    Article  CAS  PubMed  Google Scholar 

  13. Brito RA, Medgyesy D, Zukowski TH, Royce ME, Ravandi-Kashani F, Hoff PM, et al. Fluoropyrimidines: a critical evaluation. Oncology. 1999;57(Suppl 1):2–8.

    Article  CAS  PubMed  Google Scholar 

  14. Sharma V, Gupta SK, Verma M. Dihydropyrimidine dehydrogenase in the metabolism of the anticancer drugs. Cancer Chemother Pharmacol. 2019;84:1157–66.

    Article  CAS  PubMed  Google Scholar 

  15. van Kuilenburg ABP, De Abreu RA, van Gennip AH. Pharmacogenetic and clinical aspects of dihydropyrimidine dehydrogenase deficiency. Ann Clin Biochem. 2003;40:41–5.

    Article  PubMed  Google Scholar 

  16. Maslarinou A, Manolopoulos VG, Ragia G. Pharmacogenomic-guided dosing of fluoropyrimidines beyond DPYD: time for a polygenic algorithm? Front Pharmacol. 2023;14:1184523.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lunenburg CATC, Henricks LM, Guchelaar H-J, Swen JJ, Deenen MJ, Schellens JHM, et al. Prospective DPYD genotyping to reduce the risk of fluoropyrimidine-induced severe toxicity: ready for prime time. Eur J Cancer. 2016;54:40–8.

    Article  PubMed  Google Scholar 

  18. Offer SM, Fossum CC, Wegner NJ, Stuflesser AJ, Butterfield GL, Diasio RB. Comparative functional analysis of DPYD variants of potential clinical relevance to dihydropyrimidine dehydrogenase activity. Cancer Res. 2014;74:2545–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Aquino de Moraes FC, Dantas Leite Pessôa FD, Duarte de Castro Ribeiro CH, Rodrigues Fernandes M, Rodríguez Burbano RM, Carneiro Dos Santos NP. Trifluridine-tipiracil plus bevacizumab versus trifluridine-tipiracil monotherapy for chemorefractory metastatic colorectal cancer: a systematic review and meta-analysis. BMC Cancer. 2024;24:674.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. de Moraes FCA, Kelly FA, Souza MEC, Burbano RMR. Impact of adjuvant chemotherapy on survival after pathological complete response in rectal cancer: a meta-analysis of 31,558 patients. Int J Colorectal Dis. 2024;39:96.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Matsusaka S, Lenz H-J. Pharmacogenomics of fluorouracil -based chemotherapy toxicity. Expert Opin Drug Metab Toxicol. 2015;11:811–21.

    Article  CAS  PubMed  Google Scholar 

  22. Houtsma D, Guchelaar HJ, Gelderblom H. Pharmacogenetics in oncology: a promising field. Curr Pharm Des. 2010;16:155–63.

    Article  CAS  PubMed  Google Scholar 

  23. Yen JL, McLeod HL. Should DPD analysis be required prior to prescribing fluoropyrimidines? Eur J Cancer. 2007;43:1011–6.

    Article  PubMed  Google Scholar 

  24. Lam SW, Guchelaar HJ, Boven E. The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev. 2016;50:9–22.

    Article  CAS  PubMed  Google Scholar 

  25. Koo K, Pasternak AL, Henry NL, Sahai V, Hertz DL. Survey of US medical oncologists’ practices and beliefs regarding DPYD testing before fluoropyrimidine chemotherapy. JCO Oncol Pract. 2022;18:e958–65.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Baker SD, Bates SE, Brooks GA, Dahut WL, Diasio RB, El-Deiry WS, et al. DPYD testing: time to put patient safety first. J Clin Oncol. 2023;41:2701–5.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sharma BB, Rai K, Blunt H, Zhao W, Tosteson TD, Brooks GA. Pathogenic DPYD variants and treatment-related mortality in patients receiving fluoropyrimidine chemotherapy: a systematic review and meta-analysis. Oncologist. 2021;26:1008–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lo CK-L, Mertz D, Loeb M. Newcastle-Ottawa scale: comparing reviewers’ to authors’ assessments. BMC Med Res Methodol. 2014;14:45.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327:557–60.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315:629–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.

    Article  CAS  PubMed  Google Scholar 

  32. Fernández MA, Izquierdo MM, Ramos JC, de Lope LR, Mdlcd R, Martin MM. 5PSQ-061 determination of genetic polymorphisms of the dihydropyrimidine dehydrogenase gene in real clinical practice: posological individualisation. Eur J Hosp Pharm. 2019;26:A229–30.

    Google Scholar 

  33. Amirfallah A, Calibasi Kocal G, Unal OU, Ellidokuz H, Oztop I, Basbinar Y. DPYD, TYMS and MTHFR genes polymorphism frequencies in a series of Turkish colorectal cancer patients. J Pers Med. 2018;8:45.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Boige V, Vincent M, Alexandre P, Tejpar S, Landolfi S, Le Malicot K, et al. DPYD genotyping to predict adverse events following treatment with fluorouracil-based adjuvant chemotherapy in patients with stage III colon cancer: a secondary analysis of the PETACC-8 randomized clinical trial. JAMA Oncol. 2016;2:655–62.

    Article  PubMed  Google Scholar 

  35. Boige V, Mendiboure J, Pignon J-P, Loriot M-A, Castaing M, Barrois M, et al. Pharmacogenetic assessment of toxicity and outcome in patients with metastatic colorectal cancer treated with LV5FU2, FOLFOX, and FOLFIRI: FFCD 2000-05. J Clin Oncol. 2010;28:2556–64.

    Article  CAS  PubMed  Google Scholar 

  36. Boisdron-Celle M, Capitain O, Faroux R, Borg C, Metges JP, Galais MP, et al. Prevention of 5-fluorouracil-induced early severe toxicity by pre-therapeutic dihydropyrimidine dehydrogenase deficiency screening: Assessment of a multiparametric approach. Semin Oncol. 2017;44:13–23.

    Article  CAS  PubMed  Google Scholar 

  37. Botticelli A, Onesti CE, Strigari L, Occhipinti M, Di Pietro FR, Cerbelli B, et al. A nomogram to predict 5-fluorouracil toxicity: when pharmacogenomics meets the patient. Anticancer Drugs. 2017;28:551–6.

    Article  CAS  PubMed  Google Scholar 

  38. Braun MS, Richman SD, Thompson L, Daly CL, Meade AM, Adlard JW, et al. Association of molecular markers with toxicity outcomes in a randomized trial of chemotherapy for advanced colorectal cancer: the FOCUS trial. J Clin Oncol. 2009;27:5519–28.

    Article  CAS  PubMed  Google Scholar 

  39. Cai X, Fang J-M, Xue P, Song W-F, Hu J, Gu H-L, et al. The role of IVS14 + 1 G > A genotype detection in the dihydropyrimidine dehydrogenase gene and pharmacokinetic monitoring of 5-fluorouracil in the individualized adjustment of 5-fluorouracil for patients with local advanced and metastatic colorectal cancer: a preliminary report. Eur Rev Med Pharmacol Sci. 2014;18:1247–58.

    CAS  PubMed  Google Scholar 

  40. Cerić T, Obralić N, Kapur-Pojskić L, Macić D, Bešlija S, Pašić A, et al. Investigation of IVS14 + 1G > A polymorphism of DPYD gene in a group of Bosnian patients treated with 5-fluorouracil and capecitabine. Bosn J Basic Med Sci. 2010;10:133–9.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Cremolini C, Del Re M, Antoniotti C, Lonardi S, Bergamo F, Loupakis F, et al. DPYD and UGT1A1 genotyping to predict adverse events during first-line FOLFIRI or FOLFOXIRI plus bevacizumab in metastatic colorectal cancer. Oncotarget. 2018;9:7859–66.

    Article  PubMed  Google Scholar 

  42. Deenen MJ, Tol J, Burylo AM, Doodeman VD, de Boer A, Vincent A, et al. Relationship between single nucleotide polymorphisms and haplotypes in DPYD and toxicity and efficacy of capecitabine in advanced colorectal cancer. Clin Cancer Res. 2011;17:3455–68.

    Article  CAS  PubMed  Google Scholar 

  43. Detailleur S, Segelov E, Re MD, Prenen H. Dihydropyrimidine dehydrogenase deficiency in patients with severe toxicity after 5-fluorouracil: a retrospective single-center study. Ann Gastroenterol. 2021;34:68–72.

    PubMed  Google Scholar 

  44. Dhawan D, Panchal H, Shukla S, Padh H. Genetic variability & chemotoxicity of 5-fluorouracil & cisplatin in head & neck cancer patients: a preliminary study. Indian J Med Res. 2013;137:125–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Etienne-Grimaldi M-C, Boyer J-C, Beroud C, Mbatchi L, van Kuilenburg A, Bobin-Dubigeon C, et al. New advances in DPYD genotype and risk of severe toxicity under capecitabine. PLoS ONE. 2017;12:e0175998.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Falvella FS, Cheli S, Martinetti A, Mazzali C, Iacovelli R, Maggi C, et al. DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Br J Clin Pharmacol. 2015;80:581–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Froehlich TK, Amstutz U, Aebi S, Joerger M, Largiadèr CR. Clinical importance of risk variants in the dihydropyrimidine dehydrogenase gene for the prediction of early-onset fluoropyrimidine toxicity. Int J Cancer. 2015;136:730–9.

    Article  CAS  PubMed  Google Scholar 

  48. Gross E, Busse B, Riemenschneider M, Neubauer S, Seck K, Klein H-G, et al. Strong association of a common dihydropyrimidine dehydrogenase gene polymorphism with fluoropyrimidine-related toxicity in cancer patients. PLoS ONE. 2008;3:e4003.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Jennings BA, Loke YK, Skinner J, Keane M, Chu GS, Turner R, et al. Evaluating predictive pharmacogenetic signatures of adverse events in colorectal cancer patients treated with fluoropyrimidines. PLoS ONE. 2013;8:e78053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Joerger M, Huitema ADR, Boot H, Cats A, Doodeman VD, Smits PHM, et al. Germline TYMS genotype is highly predictive in patients with metastatic gastrointestinal malignancies receiving capecitabine-based chemotherapy. Cancer Chemother Pharmacol. 2015;75:763–72.

    Article  CAS  PubMed  Google Scholar 

  51. Kristensen MH, Pedersen PL, Melsen GV, Ellehauge J, Mejer J. Variants in the dihydropyrimidine dehydrogenase, methylenetetrahydrofolate reductase and thymidylate synthase genes predict early toxicity of 5-fluorouracil in colorectal cancer patients. J Int Med Res. 2010;38:870–83.

    Article  CAS  PubMed  Google Scholar 

  52. Largillier R, Etienne-Grimaldi M-C, Formento J-L, Ciccolini J, Nebbia J-F, Ginot A, et al. Pharmacogenetics of capecitabine in advanced breast cancer patients. Clin Cancer Res. 2006;12:5496–502.

    Article  CAS  PubMed  Google Scholar 

  53. Loganayagam A, Arenas Hernandez M, Corrigan A, Fairbanks L, Lewis CM, Harper P, et al. Pharmacogenetic variants in the DPYD, TYMS, CDA and MTHFR genes are clinically significant predictors of fluoropyrimidine toxicity. Br J Cancer. 2013;108:2505–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Morel A, Boisdron-Celle M, Fey L, Soulie P, Craipeau MC, Traore S, et al. Clinical relevance of different dihydropyrimidine dehydrogenase gene single nucleotide polymorphisms on 5-fluorouracil tolerance. Mol Cancer Ther. 2006;5:2895–904.

    Article  CAS  PubMed  Google Scholar 

  55. Nahid NA, Apu MNH, Islam MR, Shabnaz S, Chowdhury SM, Ahmed MU, 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;81:119–29.

    Article  CAS  PubMed  Google Scholar 

  56. Negarandeh R, Salehifar E, Saghafi F, Jalali H, Janbabaei G, Abdhaghighi MJ, et al. Evaluation of adverse effects of chemotherapy regimens of 5-fluoropyrimidines derivatives and their association with DPYD polymorphisms in colorectal cancer patients. BMC Cancer. 2020;20:560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rosmarin D, Palles C, Church D, Domingo E, Jones A, Johnstone E, et al. Genetic markers of toxicity from capecitabine and other fluorouracil-based regimens: investigation in the QUASAR2 study, systematic review, and meta-analysis. J Clin Oncol. 2014;32:1031–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ruzzo A, Graziano F, Galli F, Galli F, Rulli E, Lonardi S, et al. 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;117:1269–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Salgado J, Zabalegui N, Gil C, Monreal I, Rodríguez J, García-Foncillas J. Polymorphisms in the thymidylate synthase and dihydropyrimidine dehydrogenase genes predict response and toxicity to capecitabine-raltitrexed in colorectal cancer. Oncol Rep. 2007;17:325–8.

    CAS  PubMed  Google Scholar 

  60. Salgueiro N, Veiga I, Fragoso M, Sousa O, Costa N, Pellon ML, et al. Mutations in exon 14 of dihydropyrimidine dehydrogenase and 5-fluorouracil toxicity in Portuguese colorectal cancer patients. Genet Med. 2004;6:102–7.

    Article  CAS  PubMed  Google Scholar 

  61. Schwab M, Zanger UM, Marx C, Schaeffeler E, Klein K, Dippon J, et al. Role of genetic and nongenetic factors for fluorouracil treatment-related severe toxicity: a prospective clinical trial by the German 5-FU toxicity study group. JCO. 2008;26:2131–8.

    Article  CAS  Google Scholar 

  62. Toffoli G, Innocenti F, Polesel J, De Mattia E, Sartor F, Dalle Fratte C, et al. The genotype for DPYD risk variants in patients with colorectal cancer and the related toxicity management costs in clinical practice. Clin Pharmacol Ther. 2019;105:994–1002.

    Article  CAS  PubMed  Google Scholar 

  63. Toffoli G, Giodini L, Buonadonna A, Berretta M, De Paoli A, Scalone S, et al. Clinical validity of a DPYD-based pharmacogenetic test to predict severe toxicity to fluoropyrimidines. Int J Cancer. 2015;137:2971–80.

    Article  CAS  PubMed  Google Scholar 

  64. Vivaldi C, Crucitta S, Catanese S, Cucchiara F, Arrigoni E, Pecora I, et al. Comprehensive pharmacogenetic analysis of DPYD, UGT, CDA, and ABCB1 polymorphisms in pancreatic cancer patients receiving mFOLFIRINOX or gemcitabine plus nab-paclitaxel. Pharmacogenomics J. 2021;21:233–42.

    Article  CAS  PubMed  Google Scholar 

  65. Ohnuma S, Toshima M, Miura K, Kudoh K, Ishida M, Karasawa H, et al. 38P single-nucleotide polymorphisms of DPYD predict adverse events associated with 5-fluorouracil in patients with gastrointestinal cancer. Ann Oncol. 2015;26:ix8.

    Article  Google Scholar 

  66. Ghoche A, Zadjali SA, Omar R, Osman A, Barwani HA, Mahrouqi NA, et al. P-204 DPYD gene variants and chemotherapy-induced toxicity in Omani patients with gastrointestinal tumors. Ann Oncol. 2023;34:S88.

    Article  Google Scholar 

  67. Lee AM, Shi Q, Pavey E, Alberts SR, Sargent DJ, Sinicrope FA, et al. DPYD variants as predictors of 5-fluorouracil toxicity in adjuvant colon cancer treatment (NCCTG N0147). J Natl Cancer Inst. 2014;106:dju298.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Innocenti F, Mills SC, Sanoff H, Ciccolini J, Lenz H-J, Milano G. All you need to know about DPYD genetic testing for patients treated with fluorouracil and capecitabine: a practitioner-friendly guide. JCO Oncol Pract. 2020;16:793–8.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Hertz DL, Glatz A, Pasternak AL, Lonigro RJ, Vats P, Wu Y-M et al. Integration of germline pharmacogenetics into a tumor sequencing program. JCO Precis Oncol. 2018;2:PO.18.00011.

  70. Ly RC, Schmidt RE, Kiel PJ, Pratt VM, Schneider BP, Radovich M et al. Severe capecitabine toxicity associated with a rare DPYD variant identified through whole-genome sequencing. JCO Precis Oncol. 2020;4:PO.20.00067.

  71. Paulsen NH, Vojdeman F, Andersen SE, Bergmann TK, Ewertz M, Plomgaard P, et al. DPYD genotyping and dihydropyrimidine dehydrogenase (DPD) phenotyping in clinical oncology. A clinically focused minireview. Basic Clin Pharmacol Toxicol. 2022;131:325–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Innocenti F. DPYD variants to predict 5-FU toxicity: the ultimate proof. J Natl Cancer Inst. 2014;106:dju351.

    Article  PubMed  Google Scholar 

  73. Cevik M, Namal E, Sener ND, Koksal UI, Cagatay P, Deliorman G, et al. Investigation of DPYD, MTHFR and TYMS polymorphisms on 5-fluorouracil related toxicities in colorectal cancer. Per Med. 2022;19:435–44.

    Article  CAS  PubMed  Google Scholar 

  74. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JHM, Swen JJ, et al. Clinical pharmacogenetics implementation consortium (CPIC) guideline for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing: 2017 update. Clin Pharmacol Ther. 2018;103:210–6.

    Article  CAS  PubMed  Google Scholar 

  75. Lunenburg CATC, van der Wouden CH, Nijenhuis M, Crommentuijn-van Rhenen MH, de Boer-Veger NJ, Buunk AM, et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction of DPYD and fluoropyrimidines. Eur J Hum Genet. 2020;28:508–17.

    Article  CAS  PubMed  Google Scholar 

  76. Henricks LM, Lunenburg CATC, de Man FM, Meulendijks D, Frederix GWJ, Kienhuis E, et al. DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol. 2018;19:1459–67.

    Article  CAS  PubMed  Google Scholar 

  77. Henricks LM, Lunenburg CATC, Meulendijks D, Gelderblom H, Cats A, Swen JJ, et al. Translating DPYD genotype into DPD phenotype: using the DPYD gene activity score. Pharmacogenomics. 2015;16:1277–86.

    Article  PubMed  Google Scholar 

  78. Stavraka C, Pouptsis A, Okonta L, DeSouza K, Charlton P, Kapiris M, et al. Clinical implementation of pre-treatment DPYD genotyping in capecitabine-treated metastatic breast cancer patients. Breast Cancer Res Treat. 2019;175:511–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Saif MW. Dihydropyrimidine dehydrogenase gene (DPYD) polymorphism among Caucasian and non-Caucasian patients with 5-FU- and capecitabine-related toxicity using full sequencing of DPYD. Cancer Genomics Proteom. 2013;10:89–92.

    CAS  Google Scholar 

  80. White C, Scott RJ, Paul C, Ziolkowski A, Mossman D, Ackland S. Ethnic diversity of DPD activity and the DPYD gene: review of the literature. PGPM. 2021;14:1603–17.<\/p>

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Loriot M-A, Ciccolini J, Thomas F, Barin-Le-Guellec C, Royer B, Milano G, et al. 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.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Federal University of Pará (UFPA); the Center for Research Oncology (NPO/UFPA), and thanks to the Pró-Reitoria de Pesquisa e Pós-Graduação da UFPA (PROPESP) for paying for the article.

Funding

No funding was received to assist with the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. [F.C.A.M] conceived the project, material preparation, data collection and analysis were performed by [F.C.A.M., F.A.K., and A.B.A.B.]. The figures and tables were created by [F.C.A.M., F.A.K., A.B.A.B.]. The first draft of the manuscript was written by [F.C.A.M., A.B.A.B., V.K.T.S., and R.M.R.B.] and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Francisco Cezar Aquino de Moraes.

Ethics declarations

Institutional review board statement

No ethical approval was required for this systematic review with meta-analysis, as all data were already published in peer-reviewed journals. Furthermore, no patients or animals were involved in the design, conduct, or interpretation of our study.

Informed consent

Not aplicable.

Competing interests

The authors declare no competing interests.

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Moraes, F.C.A., de Almeida Barbosa, A.B., Sano, V.K.T. et al. Pharmacogenetics of DPYD and treatment-related mortality on fluoropyrimidine chemotherapy for cancer patients: a meta-analysis and trial sequential analysis. BMC Cancer 24, 1210 (2024). https://doi.org/10.1186/s12885-024-12981-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12981-5

Keywords