The Pharmacogenomics Journal (2013) 13, 389–395 & 2013 Macmillan Publishers Limited All rights reserved 1470-269X/13 www.nature.com/tpj

REVIEW Evaluation of predictive tests for screening for dihydropyrimidine dehydrogenase deficiency

MC van Staveren1, H Jan Guchelaar2, ABP van Kuilenburg3, H Gelderblom4 and JG Maring5

5-Fluorouracil (5-FU) is rapidly degraded by dihyropyrimidine dehydrogenase (DPD). Therefore, DPD deficiency can lead to severe toxicity or even death following treatment with 5-FU or capecitabine. Different tests based on assessing DPD enzyme activity, genetic variants in DPYD and mRNA variants have been studied for screening for DPD deficiency, but none of these are implemented broadly into clinical practice. We give an overview of the tests that can be used to detect DPD deficiency and discuss the advantages and disadvantages of these tests.

The Pharmacogenomics Journal (2013) 13, 389–395; doi:10.1038/tpj.2013.25; published online 16 July 2013 Keywords: dihyropyrimidine dehydrogenase; 5-fluorouracil; genotyping; pharmacokinetics; phenotyping;

INTRODUCTION Pyrimidinemia’, ‘uracil’, ‘capecitabine’, ‘dihydrouracil dehydrogenase The fluoropyrimidine 5-fluorouracil (5-FU) and its prodrug (nadp)’, ‘dihydrouracil dehydrogenase’, ‘dihydropyrimidine dehydrogen- ase’, ‘dihydrouracil dehydrogenase (nad þ )’ and ‘dpd’. The search resulted capecitabine, are the cornerstone of treatment of numerous types in 397, 378, 568 and 28 hits for MEDLINE, EMBASE, Web of Science and of cancer. The use of fluoropyrimidines is associated with Cochrane, respectively. The unique hits collected from these databases numerous side effects, such as myelosuppression, hand-foot were selected and further limited to English language papers from 1980 to syndrome, mucositis, diarrhea and occasionally cardiac toxicity. 1 January 2012. Papers describing studies aimed at testing DPD activity After parenteral administration of 5-FU, 70–90% of the parent and/or DPD deficiency in volunteers or patients were selected. Cross drug is degraded by dihydropyrimidine dehydrogenase (DPD).1–5 references were identified from bibliographies from the selected studies. A small proportion of patients develop extreme toxicity after Only articles that describe the complete performance of the test were administration of a fluoropyrimidine due to a partial or complete included; reviews and papers describing in vitro studies including DPD DPD deficiency and hence a strongly reduced capacity to degrade activity in cancer cells or studies in animals were neglected. The collected publications were divided into three categories, describing tests aimed 5-FU.6–10 In case of complete DPD deficiency, 5FU treatment may 11 at assessing DPD enzyme activity (38 articles), genetic variants in DPYD even result in a lethal outcome. It was initially estimated that in (20 articles) and mRNA variants (3 articles). 3–5% of Caucasians the activity of DPD is strongly reduced due to (epi)genetic variations in the gene encoding DPD.4,12 However, this percentage has been disputed as there is still no consensus Tests aimed at assessing (surrogates for) DPD enzyme activity on the definition of DPD deficiency, and therefore the incidence Several tests have been described to assess the activity of the enzyme of DPD deficiency reported in numerous studies is strongly DPD. dependent on the method used to assess DPD deficiency13 and the cutoff level chosen to define DPD deficiency.5 DPD activity in peripheral blood mononuclear cells (PBMCs). The majority of DPD is reported to be in the liver,1,14 but DPD activity in other tissues such Prospective testing for DPD deficiency in patients might as lymphocytes contribute to metabolism of fluoropyrimidines as well.15 prevent DPD-deficient patients from severe toxicity or even death. The liver DPD activity in patients revealed a strong correlation with DPD In this review, we describe current methods for determination of activity in PBMCs.16 The mean DPD activity in PBMCs of patients with a DPD deficiency. We discuss the potential and limitations of these partial DPD deficiency is approximately 48% to that observed in the tests for routine clinical use. In addition, we have defined normal population due to heterozygosity for a pathological mutation.17 recommendations that can help successful implementation of a The methodology of the test includes incubating isolated lymphocytes pre-emptive testing strategy to predict fluoropyrimidine-related with radioactive labeled 5-FU or after which the degradation toxicity. products are measured by high-performance liquid chromatography (HPLC) with a radioisotope flow detector.2,18–20 A HPLC-electrospray tandem mass spectrometry (HPLC MS/MS) method has also been developed.17,21 The use of a porous graphitic carbon column22 results in METHODS a HPLC process that is highly pH stable compared with the reversed-phase To identify studies describing diagnostic tests to detect DPD deficiency, C-18 column, and the detection limit was at least similar to the C-18 a systematic MEDLINE, EMBASE, Web of Science and Cochrane search columns with considerably shorter analysis time. This method was was conducted using the following combination of MESH terms:, ‘dihydro- validated (Table 1). Evaluation of the stability of DPD in PBMCs indicated pyrimidine dehydrogenase deficiency’, ‘dpd deficiency’, ‘Familial that the DPD activity decreased approximately 50% upon freezing but was

1Department of Pharmacy, Scheper Hospital Emmen and Ro¨pcke Zweers Hospital Hardenberg, Emmen, The Netherlands; 2Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands; 3Laboratory Genetic Metabolic Diseases, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands; 4Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands and 5Department of Pharmacy, Diaconessen Hospital Meppel and Bethesda Hospital Hoogeveen, Meppel, The Netherlands. Correspondence: MC van Staveren, Department of Pharmacy, Scheper Hospital Emmen and Ro¨pcke Zweers Hospital Hardenberg, Boermarkeweg 60, Emmen 7824 AA, The Netherlands. E-mail: [email protected] Received 7 November 2012; revised 22 May 2013; accepted 29 May 2013; published online 16 July 2013 390 h hraoeoisJunl(03,39–395 – 389 (2013), Journal Pharmacogenomics The

Table 1. Validation parameters of the diagnostic tests at the enzyme level to screen for DPD deficiency

Inter-assay Study n Reference method Recovery Sensitivity Specificity variability Linear Intra-assay variability

Mattison et al.24 58 volunteers DPD in PBMC ND 100% 96% ‘Reproducible’ ND o5% Genotyping with DHPLC Mattison et al.25 23 volunteers, 8 cancer patients DPD in PBMC ND ND ND ND ND ND Mattison et al.26 258 volunteers DPD in PBMC ND 86% 99% ND ND ND Johnson et al.19 100 volunteers, 80 cancer NA 495% ND ND o8% CV Yes o6.5% CV patients Liem et al.22 132 volunteers NA 99±2%, 3% CV ND ND o4% CV Yes o3% Lostia et al.21 39 cancer patients HPLC-UV ND ND ND 0.7–5.6% CVa Yes 8.3–1.4% CVa b c van Kuilenburg 28 cancer patients, 1 patient Radioactive 94.4–102.4 99.4–101.6% ND ND 1.7–4.7% CV Yes 1.0–3.2% CV DPD for tests Predictive et al.17 with verified DPYD mutation thymine by HPLC Staveren van MC MS/MS van Kuilenburg 2 cancer patients, unknown NA ND ND ND 9% CV Yes 5% CV et al.18 number healthy volunteers Bi et al.29 4 patients participating in a NA (Relative recovery ND ND 6.73, 6.95, 5.96% ND 7.04, 5.40, 1.24% (RSD phase 1 trial compared with water) (RSD 0.27, 25 and 0.27, 25 and 250 mM) 95.8±2.0 1 mM 95.5±10.9% 250 mM) tal et 100 mM Garg et al.34 23 cancer patients, blank plasma NA 93–100 Uracil d, 95–99% ND ND 0.2–7% RSD Yes 0.8–7% RSD uracile, healthy volunteers UH2 Uracil 1.2–9.9% 2.6–9.7% RSD UH2 RSD UH2 Kristensen et al.37 68 CRC patients, 100 healthy IVS þ 1G4A ND 87% 93% ND ND ND controls mutation 5-FU plasma levels Ciccolini et al.28 30 blank plasma samples, one U/UH2 ratio 90% ND ND 10.6, 1212.8% Yes 12, 2.4, 3.3% case report 20 ng ml À 1 , 20 ng ml À 1 , 75 ng ml À 1 , 75 ng ml À 1 , 375 ng ml À 1 5-FU 375 ng ml À 1 5-FU Beumer et al.42 156 plasma samples obtained LC-MS/MS 93–104% ND ND o2% CV Yes 2.1–3.9% CV from patients; Plasma pool samples di Paolo et al.39 25 CRC patients DPD in PBMC 85 5-FU; 81% 5-FDHU ND ND 5.28–9.44 5-FU; Yes 4.91–6.05% 5-FU; 5.00–9.03 5-FDHU 4.55–6.99% 5-FDHU Remaud et al.36 8 volunteers, blank plasma NA 73±2% Uracil 67±2% ND ND 0.9–2.3% U; Yes 1.3–5.3% U; 1.3–7.1% & UH2; 82±3FUH2% 0.7–5.6% UH2 UH2 03McilnPbihr Limited Publishers Macmillan 2013 van Kuilenburg 30 cancer patients, 18 controls 5-FU loading dose ND 100%f 90%f ND Yes ND et al.8 De´ porte-Fe´ty 1 volunteer, 29 cancer patients DPD in PBMC with ND ND ND 2.5–8.7% Yes 9.5–3.2% et al.40 radiolabeled 5-FU Abbreviations: CRC, colorectal cancer; CV, coefficient of variation; DHPLC, denaturing high-performance liquid chromatography; DPD, dihydropyrimidine dehydrogenase; 5-FDHU, 5-fluoro-5,6-dihydrouracil; FUH2, dihydrofluorouracil; 5-FU, 5-fluorouracil; HPLC, high-performance liquid chromatography; LC-MS/MS, liquid chromatography-tandem mass spectrometry; NA, not applicable; ND, not determinated; PBMC, peripheral blood mononuclear cell; RSD, relative standard variation; U, uracil; UH2, dihydrouracil; UV, ultraviolet. a5-FU concentration range of 0.156, 0.195, 078 and 1.95 mgmlÀ 1 . bInter-assay recovery at low, normal and high concentration of . cIntra-assay recovery at low, normal and high concentration of d e dihydrothymine. Concentration range of 0.05, 0.1, 0.2, 0.5, 1 and 2 mM of uracil and dihydrouracil respectively. Concentration range of 0.02, 0.05, 0.1, 0.2, 0.5, 1 and 2 mM of uracil and dihydrouracil, respectively. f À 1 À 2 Multiple variables Vmax (mg h h) and t1/2 (h) after a single dose of 300 mg m 5-FU. Predictive tests for DPD MC van Staveren et al 391 stable for at least 1 month.16 The stability of radioactive 5-FU and validated (Table 1). A strong correlation was detected between the DHU/U its metabolite dihydrofluorouracil (DHFU) in the reaction mixture was ratio in plasma with 5-FU half-life, clearance and plasma levels.30,32,33 found to be stable for 3 months and not affected by at least three However, it was reported that in some individuals with a normal U/DHU freeze-thawing cycles.19 The use of an on-line radioisotope flow detector ratio strongly elevated 5-FU levels were measured, indicating that makes this method very useful as a semi-automated radioassay, but the determination of the U/DHU ratio may not always correctly reflect 5-FU implementation on a broad scale might be hampered by the specific levels.37 The stability of uracil in plasma is at least 2 months at À 80 oC.29 equipment that is necessary. The correlation between DPD activity in A significant circadian rhythm in plasma and urine DHU/U ratio and DPD PBMCs and systemic 5-FU clearance has been studied, but the results are activity in mononuclear cells was observed in healthy subjects, but this was contradictive. One study found a good correlation,2 but two other studies disrupted in patients who were continuously infused with a high dose of found a much poorer relation.4,23 Instead of radiolabeled substrates, the 5-FU.38 The biological significance of the DPD circadian pattern to use of non-radiolabeled thymine as substrate was investigated.17 fluoropyrimidine drug treatment is obvious, as the variations in DPD Nevertheless, because of the superior sensitivity of radiochemical assays, between individuals can result in 5-FU concentration fluctuations, which the presence of a complete DPD deficiency can be established only with can be directly correlated with treatment toxicity. The circadian rhythm in the DPD assay that use radiolabeled substrates.18,19,22 The distinction a population of patients and volunteers should be investigated to between a partial and complete DPD deficiency is important to make, determine threshold values for those patients who are prone to toxicity. because in the case of a partial deficiency the oncologist might consider The good stability of uracil in plasma and the fact that the HPLC dose reduction. Complete DPD deficiency may lead to switching to a non- equipment needed for this assay is available in most clinical laboratories fluoropyrimidine-containing regimen. In general, the assays to measure make this method suitable for broad clinical implementation. DPD activity in PBMCs are labor intensive and therefore expensive. For this reason, it is not likely that this method is suitable to use broadly in clinical 5-FU therapeutic drug monitoring. The pharmacokinetics of 5-FU and its practice. metabolite DHFU can be assessed to detect possible DPD deficiency. The determination of plasma concentrations of 5-FU and DHFU can be Uracil breath test. The principle of this test is that after ingestion of an performed by HPLC,31,39,40 LC-MS/MS41 or immunoassay.42 These methods 13 À 1 13 aqueous solution of 2- C-uracil (6 mg kg ), degradation of 2- C-uracil have been validated (Table 1)31,39,42 Based on this principle, several by DPD followed by the other two enzymes of the pyrimidine degradation investigators hypothesized that the administration of a test dose of 5-FU 13 24–26 pathway takes place resulting in the production of CO2, which is before the start of chemotherapy might enable identifying subjects at risk 8,39,40,43–46 determined in exhaled breath using IR spectroscopy (UBiT-IR300). In DPD- of severe treatment-related side effects. The test doses 5-FU 13 deficient individuals, reduced 2- C-uracil catabolism results in decreased investigated were 250, 300, 370 and 450 mgm À 2 after which 5-FU and 13 13 exhaled CO2 levels. The amount of CO2 present in breath samples is DHFU were measured in plasma. The pharmacokinetic results following a expressed as delta over baseline ratio that represents a change in the test dose of 250 mgm À 2 5-FU were significantly different from a dose of 13 12 13 À 2 44 CO2/ CO2 ratio of breath samples collected before and after 2- C-uracil 370 mgm , but there was no linear correlation between 5-FU and DHFU ingestion. The breath test was evaluated in multiple studies with pharmacokinetics and no correlation between DPD activity and 5-FU volunteers and cancer patients with or without (partial) DPD deficiency pharmacokinetics following a dose of 250 mgm À 2. In one study, test doses and has been extensively validated (Table 1). One study compared the of 300 or 450 mgm À 2 were used in subjects with and without the 13 13 46 plasma 2- C-uracil pharmacokinetics with expired CO2 in subjects with presence of the c.1905 þ 1G4A mutation. Only with the dose of 25 normal and reduced PBMC DPD activity. The results of this study showed 450 mgm À 2, the mean area under the curve and clearance of 5-FU was that Tmax and Cmax in exhaled air are not correlated to plasma Cmax and significantly different, but the terminal half-life of 5-FU measured at both Tmax. The breath test is rapid and non-invasive as it only requires exhaled test doses showed a highly significant difference. A limited sampling air from patients which is collected into sealed bags that are transported to model based on two time points was studied in patients receiving a central laboratory. The integrity of the breath collection bags was stable 370 mgm À 2 5-FU as an intravenous bolus making the test more suitable for up to 201 days after their initial examination. However, the transpor- for clinical practice and less patient intensive.45 More recently, a validated tation may delay the availability of outcome of the test result for the limited sampling PK model was presented based on a single sample after physician, possibly delaying the start of chemotherapy. A broad clinical use 300 or 450 mgm À 2 5-FU. Normal and DPD-deficient patients could be of this test is further hampered by the limited availability of the expensive discriminated at 300 mgm À 2.8 Because of the simple methodology and 13 2- C-uracil and the fact that the UBiT-IR300 spectrophotometer that is analysis, this test can be easily performed in hospital settings. A potential needed for the analysis is not commonly available in every hospital and disadvantage of this method is that the test dose of 5-FU might cause must be purchased specially for this test. toxicity in severely deficient DPD patients (that is, homozygous DPD deficient). One study showed no toxicity in 20 of 22 patients heterozygous Uracil oral loading dose. The uracil loading dose uses uracil as a marker to for the c.1905 þ 1G4A after the administration of the single doses of 5-FU determine possible DPD deficiency.27 This test is currently under investiga- 300 and 450 mgm À 2.8 However, possible toxic reactions following a 5-FU tion in two clinical trials. Uracil is administered orally to individuals after test dose in severely deficient patients should be studied further in a larger which blood samples are withdrawn at predetermined time points. The population. amount of uracil and its metabolite dihydrouracil (DHU) are measured in plasma. The doses of uracil that were tested were 500 and 1000 mgm À 2, but the higher dose had no substantial benefits compared with the lower Tests aimed at assessing genetic variants in DPYD, the gene dose. In the group that received 500 mgm À 2, the pharmacokinetic encoding DPD parameters of U and DHU in subjects with DPD deficiency differed Denaturing HPLC (DHPLC). One possible method to detect DNA mutations significantly compared with subjects with normal DPD activity. An involves DHPLC.47–49 This assay is based on temperature-dependent advantage of this test is that uracil is relatively inexpensive compared separation of DNA containing mismatched base pairs from a pool of PCR- 13 with the 2- C-uracil that is used for the breath test. Uracil is only available amplified DNA fragments. This method is highly sensitive and can detect as a pharmaceutical ingredient and not as a licensed drug substance, heterozygous variants. Homozygous wild-type vs homozygous mutated necessitating a drug formulation step before use. Uracil in plasma can be sequences are not really resolved by this method. To detect homozygous measured with HPLC, which makes it suitable for broad clinical mutations, mixing of each sample with a wild type sample is necessary. For implementation. The sampling scheme that is currently used is extensive sequencing samples with aberrant peak patterns, it is necessary to finally and for that reason less convenient for the patient. In addition, the determine the kind of mutation with the use of a secondary test, which will sensitivity and specificity of this test have not yet been determined that increase the total cost. DHPLC can be used to detect currently makes it impossible to compare with other methods. unrecognized unknown sequence variations in the DPYD gene. The DNA is isolated out of the blood obtained by a single venapuncture. It takes Endogenous U/DHU or DHU/U ratio. Besides the ex vivo measurement of approximately 250 min to screen the DPYD gene for mutations at one DPD activity in human cells, alternative assays have been developed, temperature.47 A drawback of the DHPLC method is that it is laborious and including the analysis of endogenous uracil (U) and/or DHU levels or their time consuming, and for that reason, not suitable for large numbers of ratio in plasma28–34 or urine.20,35 In case of a (partial) DPD deficiency, the patients. Secondly, the equipment that is used is not available in every breakdown of U is impaired causing elevated endogenous uracil levels and hospital laboratory. With this technique, the 23 DPYD exons were screened decreased DHU levels in biological fluids, such as plasma or urine. U and in DNA samples of randomly selected individuals, DPD-deficient patients DHU can be measured by HPLC36 and GC29 methods, which have been and patients who experienced toxicity following 5-FU treatment.47–49

& 2013 Macmillan Publishers Limited The Pharmacogenomics Journal (2013), 389 – 395 Predictive tests for DPD MC van Staveren et al 392 Missense mutations in the DPYD gene will not necessary lead to reduced for aberrant methylation of the promoter of DPYD will become current DPD activity. Gross et al.49 analyzed DNA of four individuals with symptoms practice soon. of 5-FU-related toxicity and compared the data with control samples of 157 healthy individuals. Several missense mutations were found in the DNA of the patients, but only in one patient with four mutations a lower DPD Tests aimed at assessing mRNA variants encoding DPD activity in PBMCs was found. This result displays a general problem that DNA mutations can result in altered mRNA levels or aberrant splicing of the can be found with genotyping and 5-FU-related toxicity. For a good pre-mRNA. It is hypothesized that DPD enzyme activity is possibly 68 predictive value, there has to be a clear correlation between genetic correlated with DPD mRNA expression. Using a reverse transcription- variants and its effect on DPD activity or the likelihood to develop toxicity. PCR-based assay, the exon 14 skipping mutation has been studied in 69 A solution to encounter this problem is to determine the impact of novel patients with and without grade 3–4 toxicity following 5-FU therapy. The mutations found in the DPD gene on enzyme activity.49–51 DPD enzyme activity correlated with DPD mRNA expression in biopsy-sized tissue samples, including PBMCs.70 A disadvantage of mRNA screening is 51–57 the instability of mRNA in the blood that makes it unsuitable for routinely Pyrosequencing. Pyrosequencing is based on the utilization of ATP to screening, as the equipment needed for mRNA screening is not widely produce light. The pyrosequencing reaction results in the release of a available in every hospital. For this reason, it is unlikely that this technique pyrophosphate molecule with the sequential incorporation of bases to the is suitable for prospective screening of large number of patients. DNA template. The technique is developed into a fully automated process as a result of advanced equipment that allows genotyping of multiple samples within an hour. However, the initial investment in the equipment is around Euro 50 000–80 000, and as a result, it is not available in every DISCUSSION hospital laboratory. The technique of pyrosequencing and its role Predicting toxicity in detecting mutations in the DPYD gene has been extensively studied The aim of the tests described in this review is to predict and in volunteers and cancer patients. In a population of 14 individuals with prevent extreme toxicity following 5-FU treatment by detecting a reduced DPD activity and severe 5-FU-related toxicity, 57% had a 57 DPD deficiency, which results in decreased 5-FU clearance leading molecular basis for their deficient phenotype. Three DPYD variants, the to a higher incidence of side effects. However, some patients who c.1905 þ 1G4A, c.2846A4T and c.1236G4A, were strongly associated with fluoropyrimidine-induced toxicity.58–60 In a large population of 487 suffer from severe side effects caused by 5-FU show normal 5-FU patients, one-third of the patients with one of the single- pharmacokinetics or wild-type DPYD genotype, and therefore it is polymorphisms (SNPs) had no adverse reactions, and therefore the important to realize that prospective testing for DPD deficiency presence of a SNP will not automatically imply the development of will not exclude all 5-FU-related toxicity. toxicity following 5-FU therapy. Interestingly, the absence of a SNP will also not exclude developing toxicity as in several studies patients were Sensitivity and specificity identified who had no SNP but still suffered from severe initial side effects as a result of a lowered 5-FU plasma clearance.53,54,61 The determination of Tests at the DNA and RNA level can be used to determine the a single SNP can be easily performed in hospital settings and costs, at this presence of certain mutations in the DPYD gene. Analysis of the moment, around 25 Euros per SNP. Determination of multiple clinically prevalence of the various mutations in the DPD gene (DPYD) relevant SNPs could potentially enhance sensitivity and specificity. revealed that three mutations, c.1905 þ 1G4A, c.2846A4T and Pyrosequencing is used more and more in daily practice before 5-FU c.1129–5923C4G, were most frequently associated with administration despite the lack of prospective evidence of its usefulness. toxicity.5,11,54,58–60,71 Unfortunately, fluoropyrimidine toxicity is Like all PCR-based tests, a disadvantage of pyrosequencing is that with this only partly explained by mutations in the coding region of method only SNPs and small deletions are detected. Large genomic DPYD.59 With respect to overall toxicity, the sensitivity for deletions are missed and especially those that are present in the DPD 72 gene.59 screening for only the c.1905 þ 1G4A mutation was 5.5% and 31% for c.1905 þ 1G4A, c.2846A4T (p.D949V) and c.1679T4G.54 Restriction fragment-length polymorphism (RFLP). RFLP is a technique in Until now, only coding areas and flanking intron sequences have which DNA is spliced into fragments by restriction enzymes. Following the been analyzed, though deep intron mutations might influence the 59 splicing, the DNA fragments are separated by gel electrophoresis. RFLP has splicing of DYPD pre-mRNA, as was demonstrated recently. In been used in studies that investigated the presence of the c.1905 þ 1G4A the case of a test to detect DPD deficiency, a high sensitivity is mutation in cancer patients with 5-FU-related toxicity.11,62 essential as low specificity might lead to unnecessary dose reduction and therefore suboptimal therapy. A solution for this 73 Single-strand conformation polymorphisms (SSCP). SSCP is a technique might be to develop a 5-FU dose escalation model, but this has that was also optimized to be useful for pharmacogenetic DPD studies and to be studied more intensively for different types of cancer and can be used to detect DNA sequence changes. The principle of SSCP is 5-FU treatment. based on changes in the secondary structures in single-strand DNA fragments caused by a change in sequence, which are detected as alterations in fragment mobility by gel electrophoresis. The conditions of Definition of DPD deficiency SSCP were optimized with an automated system to screen genetic Among the tests described, there is wide variation in the definition polymorphisms in the DPYD gene and its efficacy was evaluated by using of DPD deficiency. The lack of a clear definition of DPD deficiency 63 21 DNA samples with previously characterized polymorphisms. The might hamper the implementation of prospective screening on a polymorphism detection rate of this technique was 95.3%. broad scale. With phenotyping, a patient is considered to be DPD deficient if the DPD activity is below a subjective pre-set threshold Epigenetics. Since the observation in several studies that DPYD sequence value. DPD activity measured in PBMCs has the problem that its variants could not fully explain the molecular basis of DPD deficiency, it is 64,65 precision largely depends on the isolated PBMC sub-fractions of hypothesized that methylation of the DPYD promoter might be an blood cell components, which can be variable.74 With genotyping alternative mechanism for DPD deficiency in cancer patients. Methylation of CpG islands located in the 50-regulatory region of the DPYD gene has studies, DPD deficiency is often defined as the presence of one or been shown to inhibit transcription.64 The methylation status of DNA multiple SNPs, but the correlation between the genotype and fragments can be detected with the use of DHPLC or with pyrosequencing. diminished 5-FU clearance of all known polymorphisms has not One study showed a significant association between aberrant methylation been clarified. of the DPYD promoter and DPD enzyme deficiency in 80% of DPD-deficient individuals, whereas all individuals with normal DPD enzyme activity tested Cost effectiveness negative for methylation.64 In contrast, other studies did not confirm the role of DPYD promoter hypermethylation to the development of severe The cost-effectiveness aspects of methods that can be used for 5-FU toxicity.66,67 The contradictory results of the studies investigating the prospective DPD screening are sparsely described. The preva- role of methylation of the DPYD promoter make it unlikely that screening lence of partial DPD deficiency in Caucasian population is at

The Pharmacogenomics Journal (2013), 389 – 395 & 2013 Macmillan Publishers Limited Predictive tests for DPD MC van Staveren et al 393 4,12,54 least 3%. As a result, many patients need to be screened 9 Mercier C, Ciccolini J. Severe or lethal toxicities upon capecitabine intake: is pre-emptively in order to diagnose those with DPD deficiency. DPYD genetic polymorphism the ideal culprit? Trends Pharmacol Sci 2007; 28: Generally, genotyping is more simple and less patient invasive 597–598. than phenotyping tests. However, the main problem with 10 Yang CG, Ciccolini J, Blesius A, Dahan L, Bagarry-Liegey D, Brunet C et al. DPD- genotyping is the relatively low sensitivity and specificity. The based adaptive dosing of 5-FU in patients with head and neck cancer: impact on determination of multiple SNPs with high prevalence might help treatment efficacy and toxicity. Cancer Chemother Pharmacol 2011; 67: 49–56. 11 van Kuilenburg AB, Muller EW, Haasjes J, Meinsma R, Zoetekouw L, Waterham HR to enhance sensitivity. Phenotyping tests for now seem to have et al. Lethal outcome of a patient with a complete dihydropyrimidine dehydro- a better correlation with the occurrence of 5-FU toxicity, but genase (DPD) deficiency after administration of 5-fluorouracil: frequency of the as with the genotyping strategy, sensitivity and specificity have to common IVS14 þ 1G4A mutation causing DPD deficiency. Clin Cancer Res 2001; be established and improved. 7: 1149–1153. In order to improve broad clinical implication of a diagnostic 12 Lu Z, Zhang R, Carpenter JT, Diasio RB. Decreased dihydropyrimidine dehy- DPD test, we make the following considerations: drogenase activity in a population of patients with breast cancer: implication for 5-fluorouracil-based chemotherapy. Clin Cancer Res 1998; 4: 325–329. 13 Boisdron-Celle M, Remaud G, Traore S, Poirier AL, Gamelin L, Morel A et al.  To enhance sensitivity of preventing fluoropyrimidine toxicity, 5-Fluorouracil-related severe toxicity: a comparison of different methods for future studies should investigate the optimization of the the pretherapeutic detection of dihydropyrimidine dehydrogenase deficiency. genotyping and fenotyping strategy to determine which test Cancer Lett 2007; 249: 271–282. principle is in favor for detection of DPD-deficient patients. 14 van Kuilenburg AB, van Lenthe H, Van Gennip AH. Activity of pyrimidine degra-  When the effectiveness of a test is established, future studies dation enzymes in normal tissues. Nucleosides Nucleic Acids 2006; 25: regarding this test should specifically focus on the cost- 1211–1214. effectiveness. 15 Naguib FN, el Kouni MH, Cha S. Enzymes of uracil catabolism in normal and  A consensus definition of DPD deficiency has to be derived neoplastic human tissues. Cancer Res 1985; 45: 5405–5412. internationally to establish incidence of DPD deficiency and to 16 Lu Z, Zhang R, Diasio RB. Dihydropyrimidine dehydrogenase activity in human compare study results. peripheral blood mononuclear cells and liver: population characteristics, newly identified deficient patients, and clinical implication in 5-fluorouracil chemo-  Sensitivity and specificity regarding prevention of fluoropyrimi- therapy. Cancer Res 1993; 53: 5433–5438. dine-related toxicity of every test have to be determined. 17 van Kuilenburg AB, van LH, Zoetekouw L, Kulik W. HPLC-electrospray tandem mass spectrometry for rapid determination of dihydropyrimidine dehydrogenase In conclusion, several tests are available to screen patients for DPD activity. Clin Chem 2007; 53: 528–530. deficiency before their first treatment with fluoropyrimidines in 18 van Kuilenburg AB, van LH, Tromp A, Veltman PC, Van Gennip AH. Pitfalls in the a rapid and possibly low invasive way. At this moment, the diagnosis of patients with a partial dihydropyrimidine dehydrogenase deficiency. challenge is to optimize the tests and to determine which test Clin Chem 2000; 46: 9–17. strategy is most suitable to predict patients at risk of developing 19 Johnson MR, Yan J, Shao L, Albin N, Diasio RB. Semi-automated radioassay for fluoropyrimidine-related toxicity caused by DPD deficiency and to determination of dihydropyrimidine dehydrogenase (DPD) activity. Screening cancer patients for DPD deficiency, a condition associated with 5-fluorouracil incorporate pre-emptive screening broadly into daily routine. toxicity. J Chromatogr B Biomed Sci Appl 1997; 696: 183–191. 20 Dobashi K, Ohe E, Yamaguchi K, Takeshita S, Ayabe T, Miyazaki M et al. Severe 5-fluorouracil-induced toxicity due to increased dihydropyrimidine dehydro- genase activity: report of a patient survival and urinary pyrimidine and CONFLICT OF INTEREST dihydropyrimidine levels. Int J Clin Oncol 1999; 4: 241–243. The authors declare no conflict of interest. 21 Lostia AM, Lionetto L, Ialongo C, Gentile G, Viterbo A, Malaguti P et al. A liquid chromatography-tandem mass spectrometry method for the determination of 5-Fluorouracil degradation rate by intact peripheral blood mononuclear cells. Ther Drug Monit 2009; 31: 482–488. REFERENCES 22 Liem LK, Choong LH, Woo KT. Porous graphitic carbon shows promise for 1 Diasio RB, Harris BE. Clinical pharmacology of 5-fluorouracil. Clin Pharmacokinet the rapid screening partial DPD deficiency in lymphocyte dihydropyrimidine 1989; 16: 215–237. dehydrogenase in Chinese, Indian and Malay in Singapore by using semi- 2 Fleming RA, Milano G, Thyss A, Etienne MC, Renee N, Schneider M et al. Corre- automated HPLC-radioassay. Clin Biochem 2002; 35: 181–187. lation between dihydropyrimidine dehydrogenase activity in peripheral mono- 23 Di PA, Danesi R, Falcone A, Cionini L, Vannozzi F, Masi G et al. Relationship nuclear cells and systemic clearance of fluorouracil in cancer patients. Cancer Res between 5-fluorouracil disposition, toxicity and dihydropyrimidine dehydro- 1992; 52: 2899–2902. genase activity in cancer patients. Ann Oncol 2001; 12: 1301–1306. 3 Harris BE, Song R, Soong SJ, Diasio RB. Relationship between dihydropyrimidine 24 Mattison LK, Ezzeldin H, Carpenter M, Modak A, Johnson MR, Diasio RB. Rapid dehydrogenase activity and plasma 5-fluorouracil levels with evidence for circa- identification of dihydropyrimidine dehydrogenase deficiency by using a novel dian variation of enzyme activity and plasma drug levels in cancer patients 2-13C-uracil breath test. Clin Cancer Res 2004; 10: 2652–2658. receiving 5-fluorouracil by protracted continuous infusion. Cancer Res 1990; 50: 25 Mattison LK, Fourie J, Hirao Y, Koga T, Desmond RA, King JR et al. The uracil breath 197–201. test in the assessment of dihydropyrimidine dehydrogenase activity: pharmaco- 4 Etienne MC, Lagrange JL, Dassonville O, Fleming R, Thyss A, Renee N et al. kinetic relationship between expired 13CO2 and plasma [2-13C]dihydrouracil. Population study of dihydropyrimidine dehydrogenase in cancer patients. J Clin Clin Cancer Res 2006; 12: 549–555. Oncol 1994; 12: 2248–2253. 26 Mattison LK, Fourie J, Desmond RA, Modak A, Saif MW, Diasio RB. Increased 5 van Kuilenburg AB, Meinsma R, Zoetekouw L, Van Gennip AH. Increased risk of prevalence of dihydropyrimidine dehydrogenase deficiency in African-Americans grade IV neutropenia after administration of 5-fluorouracil due to a dihydro- compared with Caucasians. Clin Cancer Res 2006; 12: 5491–5495. pyrimidine dehydrogenase deficiency: high prevalence of the IVS14 þ 1g4a 27 van Staveren MC, Theeuwes-Oonk B, Guchelaar HJ, van Kuilenburg AB, Maring JG. mutation. Int J Cancer 2002; 101: 253–258. Pharmacokinetics of orally administered uracil in healthy volunteers and in 6 Ciccolini J, Mercier C, Dahan L, Evrard A, Boyer JC, Richard K et al. Toxic death-case DPD-deficient patients, a possible tool for screening of DPD deficiency. Cancer after capecitabine þ oxaliplatin (XELOX) administration: probable implication of Chemother Pharmacol 2011; 68: 1611–1617. dihydropyrimidine deshydrogenase deficiency. Cancer Chemother Pharmacol 28 Ciccolini J, Mercier C, Evrard A, Dahan L, Boyer JC, Duffaud F et al. A rapid and 2006; 58: 272–275. inexpensive method for anticipating severe toxicity to fluorouracil and fluoro- 7 Maring JG, van Kuilenburg AB, Haasjes J, Piersma H, Groen HJ, Uges DR et al. uracil-based chemotherapy. Ther Drug Monit 2006; 28: 678–685. Reduced 5-FU clearance in a patient with low DPD activity due to heterozygosity 29 Bi D, Anderson LW, Shapiro J, Shapiro A, Grem JL, Takimoto CH. Measurement of for a mutant allele of the DPYD gene. Br J Cancer 2002; 86: 1028–1033. plasma uracil using gas chromatography-mass spectrometry in normal individuals 8 van Kuilenburg AB, Hausler P, Schalhorn A, Tanck MW, Proost JH, Terborg C et al. and in patients receiving inhibitors of dihydropyrimidine dehydrogenase. Evaluation of 5-fluorouracil pharmacokinetics in cancer patients with a J Chromatogr B Biomed Sci Appl 2000; 738: 249–258. c.1905 þ 1G4A mutation in DPYD by means of a Bayesian limited sampling 30 Ben FR, Gross E, Ben AS, Hassine H, Saguem S. The dihydrouracil/uracil ratio in strategy. Clin Pharmacokinet 2012; 51: 163–174. plasma, clinical and genetic analysis for screening of dihydropyrimidine

& 2013 Macmillan Publishers Limited The Pharmacogenomics Journal (2013), 389 – 395 Predictive tests for DPD MC van Staveren et al 394 dehydrogenase deficiency in colorectal cancer patients treated with 5-fluoro- dehydrogenase gene interpreted by analysis of the three-dimensional protein uracil. Pathol Biol (Paris) 2009; 57: 470–476. structure. Biochem J 2002; 364: 157–163. 31 Ciccolini J, Mercier C, Blachon MF, Favre R, Durand A, Lacarelle B. A simple and 51 Weidensee S, Goettig P, Bertone M, Haas D, Magdolen V, Kiechle M et al. A mild rapid high-performance liquid chromatographic (HPLC) method for 5-fluorouracil phenotype of dihydropyrimidine dehydrogenase deficiency and developmental (5-FU) assay in plasma and possible detection of patients with impaired dihydro- retardation associated with a missense mutation affecting cofactor binding. pyrimidine dehydrogenase (DPD) activity. J Clin Pharm Ther 2004; 29: 307–315. Clin Biochem 2011; 44: 722–724. 32 Zhou ZW, Wang GQ, Wan dS, Lu ZH, Chen YB, Li S et al. The dihydrouracil/uracil 52 Ahluwalia R, Freimuth R, McLeod HL, Marsh S. Use of pyrosequencing to detect ratios in plasma and toxicities of 5-fluorouracil-based adjuvant chemotherapy in clinically relevant polymorphisms in dihydropyrimidine dehydrogenase. colorectal cancer patients. Chemotherapy 2007; 53: 127–131. Clin Chem 2003; 49: 1661–1664. 33 Gamelin E, Boisdron-Celle M, Guerin-Meyer V, Delva R, Lortholary A, Genevieve F 53 Amstutz U, Farese S, Aebi S, Largiader CR. Dihydropyrimidine dehydrogenase et al. Correlation between uracil and dihydrouracil plasma ratio, fluorouracil (5-FU) gene variation and severe 5-fluorouracil toxicity: a haplotype assessment. pharmacokinetic parameters, and tolerance in patients with advanced colorectal Pharmacogenomics 2009; 10: 931–944. cancer: a potential interest for predicting 5-FU toxicity and determining optimal 54 Morel A, Boisdron-Celle M, Fey L, Soulie P, Craipeau MC, Traore S et al. Clinical 5-FU dosage. J Clin Oncol 1999; 17: 1105. relevance of different dihydropyrimidine dehydrogenase gene single nucleotide 34 Garg MB, Sevester JC, Sakoff JA, Ackland SP. Simple liquid chromatographic polymorphisms on 5-fluorouracil tolerance. Mol Cancer Ther 2006; 5: method for the determination of uracil and dihydrouracil plasma levels: a 2895–2904. potential pretreatment predictor of 5-fluorouracil toxicity. J Chromatogr B Analyt 55 Nauck M, Gierens H, Marz W, Wieland H. Rapid detection of a common Technol Biomed Life Sci 2002; 774: 223–230. dihydropyrimidine dehydrogenase mutation associated with 5-fluorouracil toxi- 35 Sumi S, Kidouchi K, Ohba S, Wada Y. Automated screening system for purine and city and congenital thymine uraciluria using fluorogenic hybridization probes. pyrimidine metabolism disorders using high-performance liquid chromatography. Clin Biochem 2001; 34: 103–105. J Chromatogr B Biomed Appl 1995; 672: 233–239. 56 Salgueiro N, Veiga I, Fragoso M, Sousa O, Costa N, Pellon ML et al. Mutations in 36 Remaud G, Boisdron-Celle M, Hameline C, Morel A, Gamelin E. An accurate exon 14 of dihydropyrimidine dehydrogenase and 5-fluorouracil toxicity in dihydrouracil/uracil determination using improved high performance liquid Portuguese colorectal cancer patients. Genet Med 2004; 6: 102–107. chromatography method for preventing fluoropyrimidines-related toxicity in 57 van Kuilenburg AB, Haasjes J, Richel DJ, Zoetekouw L, van LH, clinical practice. J Chromatogr B Analyt Technol Biomed Life Sci 2005; 823: De Abreu RA et al. Clinical implications of dihydropyrimidine dehydro- 98–107. genase (DPD) deficiency in patients with severe 5-fluorouracil-associated 37 Kristensen MH, Pedersen P, Mejer J. The value of dihydrouracil/uracil plasma toxicity: identification of new mutations in the DPD gene. Clin Cancer Res 2000; 6: ratios in predicting 5-fluorouracil-related toxicity in colorectal cancer patients. 4705–4712. J Int Med Res 2010; 38: 1313–1323. 58 Deenen MJ, Tol J, Burylo AM, Doodeman VD, de Boer A, Vincent A et al. Rela- 38 Jiang H, Lu J, Ji J. Circadian rhythm of dihydrouracil/uracil ratios in biological tionship between single nucleotide polymorphisms and haplotypes in DPYD and fluids: a potential biomarker for dihydropyrimidine dehydrogenase levels. toxicity and efficacy of capecitabine in advanced colorectal cancer. Clin Cancer Res Br J Pharmacol 2004; 141: 616–623. 2011; 17: 3455–3468. 39 Di PA, Danesi R, Ciofi L, Vannozzi F, Bocci G, Lastella M et al. Improved analysis of 59 van Kuilenburg AB, Meijer J, Mul AN, Meinsma R, Schmid V, Dobritzsch D et al. 5-Fluorouracil and 5,6-dihydro-5-Fluorouracil by HPLC with diode array detection Intragenic deletions and a deep intronic mutation affecting pre-mRNA splicing for determination of cellular dihydropyrimidine dehydrogenase activity and in the dihydropyrimidine dehydrogenase gene as novel mechanisms causing pharmacokinetic profiling. Ther Drug Monit 2005; 27: 362–368. 5-fluorouracil toxicity. Hum Genet 2010; 128: 529–538. 40 Deporte-Fety R, Picot M, Amiand M, Moreau A, Campion L, Lanoe D et al. 60 van Kuilenburg AB, Vreken P, Abeling NG, Bakker HD, Meinsma R, van LH et al. High-performance liquid chromatographic assay with ultraviolet detection for Genotype and phenotype in patients with dihydropyrimidine dehydrogenase quantification of dihydrofluorouracil in human lymphocytes: application to deficiency. Hum Genet 1999; 104: 1–9. measurement of dihydropyrimidine dehydrogenase activity. J Chromatogr B 61 Giorgio E, Caroti C, Mattioli F, Uliana V, Parodi MI, D’Amico M et al. Severe Biomed Sci Appl 2001; 762: 203–209. fluoropyrimidine-related toxicity: clinical implications of DPYD analysis and UH2/U 41 Remaud G, Boisdron-Celle M, Morel A, Gamelin A. Sensitive MS/MS-liquid chro- ratio evaluation. Cancer Chemother Pharmacol 2011; 68: 1355–1361. matography assay for simultaneous determination of tegafur, 5-fluorouracil and 62 Magne N, Etienne-Grimaldi MC, Cals L, Renee N, Formento JL, Francoual M et al. 5-fluorodihydrouracil in plasma. J Chromatogr B Analyt Technol Biomed Life Sci Dihydropyrimidine dehydrogenase activity and the IVS14 þ 1G4A mutation 2005; 824: 153–160. in patients developing 5FU-related toxicity. Br J Clin Pharmacol 2007; 64: 42 Beumer JH, Boisdron-Celle M, Clarke W, Courtney JB, Egorin MJ, Gamelin E et al. 237–240. Multicenter evaluation of a novel nanoparticle immunoassay for 5-fluorouracil on 63 Okamoto Y, Ueta A, Sumi S, Ito T, Okubo Y, Jose Y et al. SSCP screening of the Olympus AU400 analyzer. Ther Drug Monit 2009; 31: 688–694. the dihydropyrimidine dehydrogenase gene polymorphisms of the Japanese 43 Bocci G, Barbara C, Vannozzi F, Di PA, Melosi A, Barsanti G et al. A pharmacoki- population using a semi-automated electrophoresis unit. Biochem Genet 2007; 45: netic-based test to prevent severe 5-fluorouracil toxicity. Clin Pharmacol Ther 713–724. 2006; 80: 384–395. 64 Ezzeldin HH, Lee AM, Mattison LK, Diasio RB. Methylation of the DPYD promoter: 44 Bocci G, Danesi R, Di Paolo AD, Innocenti F, Allegrini G, Falcone A et al. Com- an alternative mechanism for dihydropyrimidine dehydrogenase deficiency in parative pharmacokinetic analysis of 5-fluorouracil and its major metabolite cancer patients. Clin Cancer Res 2005; 11: 8699–8705. 5-fluoro-5,6-dihydrouracil after conventional and reduced test dose in cancer 65 Noguchi T, Tanimoto K, Shimokuni T, Ukon K, Tsujimoto H, Fukushima M et al. patients. Clin Cancer Res 2000; 6: 3032–3037. Aberrant methylation of DPYD promoter, DPYD expression, and cellular 45 Di PA, Danesi R, Vannozzi F, Falcone A, Mini E, Cionini L et al. Limited sampling sensitivity to 5-fluorouracil in cancer cells. Clin Cancer Res 2004; 10: model for the analysis of 5-fluorouracil pharmacokinetics in adjuvant chemo- 7100–7107. therapy for colorectal cancer. Clin Pharmacol Ther 2002; 72: 627–637. 66 Amstutz U, Farese S, Aebi S, Largiader CR. Hypermethylation of the DPYD pro- 46 van Kuilenburg AB, Maring JG, Schalhorn A, Terborg C, Schmalenberg H, Behnke moter region is not a major predictor of severe toxicity in 5-fluorouracil based D et al. Pharmacokinetics of 5-fluorouracil in patients heterozygous for the chemotherapy. J Exp Clin Cancer Res 2008; 27: 54. IVS14 þ 1G4A mutation in the dihydropyrimidine dehydrogenase gene. 67 Savva-Bordalo J, Ramalho-Carvalho J, Pinheiro M, Costa VL, Rodrigues A, Dias PC Nucleosides Nucleotides Nucleic Acids 2008; 27: 692–698. et al. Promoter methylation and large intragenic rearrangements of DPYD are not 47 Ezzeldin H, Okamoto Y, Johnson MR, Diasio RB. A high-throughput denaturing implicated in severe toxicity to 5-fluorouracil-based chemotherapy in gastro- high-performance liquid chromatography method for the identification of intestinal cancer patients. BMC Cancer 2010; 10: 470. variant alleles associated with dihydropyrimidine dehydrogenase deficiency. 68 Raida M, Schwabe W, Hausler P, van Kuilenburg AB, Van Gennip AH, Anal Biochem 2002; 306: 63–73. Behnke D et al. Prevalence of a common point mutation in the dihydropyrimidine 48 Fischer J, Schwab M, Eichelbaum M, Zanger UM. Mutational analysis of the human dehydrogenase (DPD) gene within the 50-splice donor site of intron 14 in patients dihydropyrimidine dehydrogenase gene by denaturing high-performance liquid with severe 5-fluorouracil (5-FU)- related toxicity compared with controls. chromatography. Genet Test 2003; 7: 97–105. Clin Cancer Res 2001; 7: 2832–2839. 49 Gross E, Ullrich T, Seck K, Mueller V, de Wit WM, von Schilling SC et al. Detailed 69 Seck K, Riemer S, Kates R, Ullrich T, Lutz V, Harbeck N et al. Analysis of the DPYD analysis of five mutations in dihydropyrimidine dehydrogenase detected gene implicated in 5-fluorouracil catabolism in a cohort of Caucasian individuals. in cancer patients with 5-fluorouracil-related side effects. Hum Mutat 2003; Clin Cancer Res 2005; 11: 5886–5892. 22: 498. 70 Johnson MR, Wang K, Smith JB, Heslin MJ, Diasio RB. Quantitation of dihydro- 50 van Kuilenburg AB, Dobritzsch D, Meinsma R, Haasjes J, Waterham HR, pyrimidine dehydrogenase expression by real-time reverse transcription poly- Nowaczyk MJ et al. Novel disease-causing mutations in the dihydropyrimidine merase chain reaction. Anal Biochem 2000; 278: 175–184.

The Pharmacogenomics Journal (2013), 389 – 395 & 2013 Macmillan Publishers Limited Predictive tests for DPD MC van Staveren et al 395 71 van Kuilenburg AB, Meinsma R, Zoetekouw L, Van Gennip AH. High prevalence of 73 Gamelin EC, Danquechin-Dorval EM, Dumesnil YF, Maillart PJ, Goudier MJ, Burtin the IVS14 þ 1G4A mutation in the dihydropyrimidine dehydrogenase gene of PC et al. Relationship between 5-fluorouracil (5-FU) dose intensity and therapeutic patients with severe 5-fluorouracil-associated toxicity. Pharmacogenetics 2002; 12: response in patients with advanced colorectal cancer receiving infusional therapy 555–558. containing 5-FU. Cancer 1996; 77: 441–451. 72 Schwab M, Zanger UM, Marx C, Schaeffeler E, Klein K, Dippon J et al. Role of 74 van Kuilenburg AB, van LH, Blom MJ, Mul EP, Van Gennip AH. Profound variation genetic and nongenetic factors for fluorouracil treatment-related severe toxicity: a in dihydropyrimidine dehydrogenase activity in human blood cells: major impli- prospective clinical trial by the German 5-FU Toxicity Study Group. J Clin Oncol cations for the detection of partly deficient patients. Br J Cancer 1999; 79: 2008; 26: 2131–2138. 620–626.

& 2013 Macmillan Publishers Limited The Pharmacogenomics Journal (2013), 389 – 395