Comprehensive

Review

Profiling Dihydropyrimidine Dehydrogenase Deficiency in Patients with Cancer Undergoing 5-Fluorouracil/Capecitabine Therapy Cédric Mercier,1 Joseph Ciccolini2 Abstract Fluoropyrimidine drugs such as 5-fluorouracil (5-FU) and capecitabine are a mainstay in the treatment of numerous solid tumors, including colorectal cancers, alone or as part of combination therapies. Cytotoxic drugs such as 5-FU and oral capecitabine display narrow therapeutic indexes combined with high interpatient pharmacokinetic variability. As a result, severe toxicities often limit or delay the administration of successive, optimal chemotherapeutic courses, leading to unfavorable clinical outcome in patients with cancer. Catabolism and deactivation of fluoropyrimidine drugs depend on a single and exclusive enzymatic step driven by dihydropyrimidine dehydrogenase (DPD). Dihydropyrimidine dehydrogenase is prone to marked circadian rhythms, drug-drug interactions, and genetic polymorphisms; influence of its erratic activity on 5-FU pharmacokinetics and toxicity profile has been extensively investigated, and it is now well known that DPD deficiency leads to severe toxicities with 5-FU or possibly capecitabine exposure. With the everincreasing number of patients with cancer likely to be treated with fluoropyrimidines, predicting and preventing the occurrence of such toxicities is now a major issue in clinical oncology. Early determination of DPD status in patients with cancer would allow identification of those at risk and help in subsequent dose adjustment or selection of other treatment modalities. Numerous methods, either genotypic or phenotypic, have been proposed to achieve this goal.This review covers a wide range of techniques available to establish DPD status in patients with cancer.

Clinical Colorectal Cancer, Vol. 6, No. 4, 288-296, 2006 Key words: Fluoropyrimidines, Genetic polymorphisms, Pharmacokinetics, Phenotypic polymorphisms

Detoxifying Fluoropyrimidine Drugs: The Dihydropyrimidine Dehydrogenase Gateway 5-fluorouracil (5-FU) metabolism has been extensively studied for decades. As a pyrimidine derivative, 5-FU is converted within tumor cells to active nucleotides, following 2 different routes known as the RNA and the DNA pathways. The role of orotate phosphoribosyl transferase,1,2 uridine phosphorylase,3,4 thymidine phosphorylase,5-8 and thymidine kinase9 in the activation process of fluoropyrimidine drugs has been extensively investigated, and so far, the exact way 5-FU is metabolized after tumoral 1EA3286,

Medical Oncology Unit, La Timone University Hospital Pharmacokinetics Laboratory, Université de la Méditerranée Marseille, France 2EA3286,

Submitted: Sep 6, 2006; Revised: Oct 27, 2006; Accepted: Nov 1, 2006 Address for correspondence: Cédric Mercier, MD, Service d’Oncologie Médicale, CHU Timone Adultes, 264, rue Saint Pierre, 13385 Marseille Cedex 05, France Fax: 33-491-835-667; e-mail: [email protected]

uptake remains far from being easily predictable.10 In contrast, the 5-FU elimination pattern is univocal. The drug displays an extensive first-pass metabolism, because > 95% of an administered dose of 5-FU is quickly dehydrogenated in the liver by dihydropyrimidine dehydrogenase (DPD) to dihydrofluorouracil (5-FU-H2). Dihydrofluorouracil is subsequently converted to β-fluoro-β-ureido-propionic acid, then fluoro-β alanine (FβAL) by dihydropyrimidinase and β-ureidopropionase, respectively, and conjugated FβAL derivatives are eventually eliminated in urine.11-14 As a result, terminal half-life of 5-FU rarely exceeds 15 minutes in most patients. The rate-limiting role of DPD in the catabolism of 5-FU has been extensively studied. It has been shown that DPD activity was slightly lower in women (< 15%) than in men,15 an observation fully consistent with the fact that 5-FU clearance is lower in women.16 Besides intersex variations, DPD displays marked circadian rhythms, with interpatient and intrapatient variabilities.17,18 Controversial evidence has emerged from the studies of ethnic influence on DPD activity. Variations in DPD activities ranged from 1 to 5 in white patients, 1 to 8 in Kenyan patients,

Electronic forwarding or copying is a violation of US and International Copyright Laws. Authorization to photocopy items for internal or personal use, or the internal or personal use of specific clients, is granted by CIG Media Group, LP, ISSN #1533-0028, provided the appropriate fee is paid directly to Copyright Clearance Center, 222 Rosewood Drive, Danvers, MA 01923 USA 978-750-8400.

288 • Clinical Colorectal Cancer November 2006

Mercier CR.indd 2

12/19/06 1:35:26 PM

and 1 to 13 in Asian patients but with nonsignificant different medians between the groups.19-22 As a result, population studies of DPD activity revealed extremely heterogeneous values,15 ranging from 65 pmol/min/mg to 559 pmol/min/mg (ratio, 9; n = 185), with unimodal or multimodal distributions.23-25 Besides circadian variations, genetic factors might partly explain this interpatient variability. The DPYD gene is highly polymorphic with > 40 mutations being identified so far,26 plus a possible methylation of the promoter leading to reduced activity,27 coupled with epigenetic regulations.28 Admittedly, respective frequencies of partial and total DPD deficiencies linked with a gene polymorphism syndrome are 3%-5% and 0.5%.15 Recent studies have suggested that black patients would be more prone to DPYD gene polymorphism than white patients,29 an observation that requires further investigations to be confirmed. Finally, intrapatient and interpatient differences in DPD activity can be caused by drug interactions. Any pyrimidine derivative that acts as a substrate for DPD can interact with fluoropyrimidine drugs in a competitive manner, such as the antiviral sorivudine.30 Conversely, anticancer drugs such as raltitrexed proved to upregulate DPD in several experimental models, with an increase in enzymatic activity and messenger RNA (mRNA) levels.31 Because of its pivotal role in 5-FU and capecitabine elimination patterns, it is not surprising that any change in DPD activity has dramatic impact on clinical outcome of patients with cancer. Admittedly, 30%-40% of the severe toxicities upon 5-FU exposure could be attributed to a diminished DPD activity32; however, recent studies suggest that DPD impairment is responsible for 60%-70% of the treatmentrelated toxicities.33

Dihydropyrimidine Dehydrogenase Deficiency and 5-Fluorouracil Pharmacokinetics: The Less You Have, the More You Get Regarding the major role played by fluoropyrimidine drugs in the treatment of numerous cancer types, alone or in combination, extensive pharmacokinetic studies of 5-FU and capecitabine have been undertaken for years, and all showed marked variations between individuals.34 Doses, routes of administration,35,36 and demographic data, such as sex or age,15,16,37,38 are the most commonly reported factors influencing 5-FU pharmacokinetic profile, although clinical studies sometimes have conflicting data. Because 5-FU elimination follows a single enzymatic pattern driven by liver DPD, many studies have focused on the impact of DPD erratic activity on 5-FU pharmacokinetic parameters.39-41 Although no relation was established between overall hepatic function and the pharmacokinetic profile of 5-FU,42,43 data have shown that DPD impairment leads to profound alteration of 5-FU pharmacokinetic profile, with reduced clearance and increased half-life.24,44,45 Consequently, patients with deficient DPD activity given the standard dosage are likely to be dramatically overexposed. This hypothesis has been fully confirmed by some case reports showing 5-FU area under the curve increasing from 9.2 (control population) to 24.7 hours per µg/mL in a patient with deficient activity46 and by a recent study showing that patients with impaired DPD displayed 5-FU plasma

concentrations up to 15-times higher than usually observed.47 Besides 5-FU, influence of DPD activity on the pharmacokinetics of oral capecitabine has been investigated and confirmed using physiologically based pharmacokinetic models.48

Dihydropyrimidine Dehydrogenase Activity and Clinical Outcome: The Lower the Activity, the Deeper the Impact Since the late 1980s, numerous studies have investigated the clinical consequences of 5-FU and capecitabine administration in patients with impaired DPD activity.44-47,49 It is well known now that reduced activity leads to sharp overexposure to 5-FU when standard dosages are administered, with subsequent sharp increase in the occurrence of drug-related toxicities. Admittedly, more than one third of the severe toxicities reported with 5-FU could be attributed to partial or total alteration in DPD activity.32 Recent studies have reconsidered this scoring on the rise, and impaired DPD function could be finally suspected in > 60%-70% of 5-FU unanticipated toxicities.33,50,51 In the most extreme cases, total DPD deficiency can lead to toxic death,52 although some studies have shown that fatal outcome can also be observed in only partially deficient patients.53 Similarly, such a dramatic outcome has been reported in 18 Japanese patients with abrogated DPD activity after the intake of sorivudine, an anti–herpes zoster drug that interacted with tegafur, one of the oral 5-FU prodrugs, in a competitive manner.30,54 Digestive and hematologic toxicities are the most frequently reported side effects observed in DPD-deficient patients treated with standard dosages of 5-FU.17,46,47,55-57 Such severe toxicities not only greatly affect patient quality of life, in some instances when not directly life-threatening, they also coerce patients to postpone or cancel the forthcoming administrations, with subsequent loss of successive and optimal courses in a row. The impact of DPD deficiency on the clinical outcome of patients with cancer upon capecitabine treatment is highly controversial. As a triple oral prodrug, capecitabine has been normally designed to yield specifically 5-FU in tumors, and in this respect, its pharmacokinetic profile should not be heavily affected by erratic DPD activity in the liver. As the final activating enzymes of capecitabine, the cytidine deaminase thymidine phosphorylase is expressed in hepatocytes, and unscheduled synthesis of 5-FU can occur in the liver, thus putting DPD-deficient patients at risk.10 This has been suggested by clinical reports describing toxicities with capecitabine in patients with impaired DPD activity.58 This has finally been confirmed by the first toxic death case reported in a patient with DPYD gene polymorphism and severe deficiency undergoing a capecitabine/oxaliplatin protocol.49

Determining Dihydropyrimidine Dehydrogenase Status in Patients with Cancer: To Be or Not to Be Deficient, That is the Question Regarding the dramatic impact of DPD impairment on 5-FU pharmacokinetic profile and subsequent occurrence of treatmentrelated toxicities, DPD status is unanimously considered a critical Clinical Colorectal Cancer November 2006 • 289

Mercier CR.indd 3

12/19/06 1:35:27 PM

Profiling Dihydropyrimidine Dehydrogenase Deficiency determinant for predicting clinical outcome in patients treated with fluoropyrimidine drugs.26 Because millions of patients with cancer receive fluoropyrimidines each year, detecting DPD deficiency has been an increasing concern since the early 1990s. Several methods (either genotypic or phenotypic, direct or indirect, or focusing on DPD protein expression levels) have been proposed to achieve this goal. As of now, none of them have stood out as a standard because of a wide range of bias, technical limitations, or inadequacies with routine clinical practice requiring simple, rapid, and inexpensive detection methods.

Methods for Gene Polymorphism of Dihydropyrimidine Dehydrogenase Gene polymorphism of DPD has been well characterized since the mid-1990s as an autosomal recessive disease, with 0.5% and 5% of the white patients being subsequently affected by total and partial deficiencies, respectively.15,59 Inherited complete DPD deficiency at birth causes a condition called thymine uraciluria that can be asymptomatic or associated with several neurologic disorders.60-62 The 150 kb DPYD gene is located on chromosome 1p22 and consists of 23 exons, ranging in size from 69 pb to 1404 pb, with 1-20 kb introns.63,64 The DPYD gene is highly polymorphic, with > 40 mutations described so far.26,65,66 The IVS14+1G>A splice-site mutation is reported as being the most common mutation in Europeans.32,50,55,59,67-70 Beside this canonical polymorphism, several deletions (298delTCAT, 812delT, 1897delC) and other missense mutations (85 T>C, 496 A>G, 703 T>C, 775 A>G, 1601 G>A, 1627 A>G, 1679 T>G, 2194 G>A, and 2846 A>T, to name but a few) have been described as well, with possible decreased expression and low DPD activity in patients.67,68,71-74 Some point mutations at positions 62 (G>A), 74 (A>G), 1003 (G>T), 1156 (G>T), and 1714 (C>G) and deletion (1812delT) have been found more specifically in Asian patients, such as Taiwanese and Japanese.19,75,76 More recently, methylation of the DPYD promoter has been related to decreased activity27 but with a probable tissue specificity.77 In addition to promoter hypermethylation, epigenetic gene silencing through histone modification could play a role in DPD impairment, at least in some tumor tissues.28 Several direct sequencing and polymerase chain reaction (PCR)–based genotypic methods59,71,78-81 and denaturing high performance liquid chromatography (HPLC) techniques74,82,83 have been published to detect the most frequent mutations on the DPYD gene. Extracting DNA from formalin-fixed paraffin-embedded tissue or blood sample is a standard, simple, and automatable procedure. The most frequently used methods for detecting single nucleotide polymorphisms are based upon real-time (RT)–PCR approaches or PCR–restriction fragment length polymorphism assays using appropriate restriction enzymes. Real time–PCR is based on different melting temperatures of fluorescent-labeled oligonucleotide hybridization probes using a single-step assay that combines fluorescence PCR and melting curve analysis. Although more complex, denaturing HPLC is capable of detecting all known sequence variations but can also identify unknown polymorphisms and promoter

hypermethylations.27,83 This method discriminates different sequence variations in the same DNA fragment because of specific elution patterns under the optimized buffer gradient conditions and calculated melting temperatures of different melting domains inherent in a given DNA sequence. Whatever method is used, the prevalence of most commonly reported mutations such as the exon-14 skipping remains controversial, with allele frequencies ranging from 0 to 2.7% according to the population studied.19,55,71,76,84-86

Methods for Phenotypic Gene Polymorphism of Dihydropyrimidine Dehydrogenase Based on enzymatic nature, developing functional testing of DPD has been proposed for years for directly or indirectly establishing deficient/nondeficient status in patients with cancer. Dihydropyrimidine dehydrogenase activity can be evaluated after 2 alternative approaches: determining enzymatic activity through ex vivo DPD direct assays or indirect evaluations performed in patients based on the monitoring of endogenous or exogenous substrates of DPD as surrogate markers. Finally, a test dose of 5-FU has been proposed as well as a possible strategy to determine DPD status in patients with cancer.

Dihydropyrimidine Dehydrogenase Direct Assays Although most markedly expressed in the liver, DPD is a ubiquitous enzyme, and several reports have shown that DPD activity measured in surrogate tissues such as lymphocytes or fibroblasts could be used as a marker for the actual liver activity.87,88 Precise measurement of DPD activity ex vivo from blood mononuclear cells was the first method proposed in the late 1980s to evaluate overall DPD functionality.23 This method was derived from the radiometric assay reported by Naguib et al for tumor biopsies.89 Dihydropyrimidine dehydrogenase activity was assessed using a radio-HPLC method measuring the conversion of radiolabeled 5-FU into 5-FU-H2. Adapted and optimized methods used tritium-labeled 5-FU, radioactive carbon–labeled (14C) 5-FU, 14C thymine, or 14C uracil as substrates, with limit of detection as low as 5 pmol/min/mg protein, down to 0.4 pmol.17,87,90-94 In addition to HPLC, a thin layer chromatography method has been developed based on the use of radiolabeled uracil.95 More recently, HPLC methods without the use of radiolabeled substrates have been proposed, thus rendering this approach cheaper and easier to adapt in most laboratories.96,97 Admittedly, peripheral blood mononuclear cell DPD levels < 100-150 pmol/min/mg protein are associated with a partial deficiency syndrome, whereas patients displaying DPD < 50-80 pmol/min/mg protein are considered profoundly deficient.24,91,98-100

Indirect Evaluations An estimation of the DPD functionality can be achieved by monitoring physiologic substrates used as probes. Plasma uracil level was proposed as a surrogate marker but showed poor correlation with DPD activity or 5-FU clearance, thereby showing limited relevance.24 Expression of levels of DPD substrate and its catabolite as a ratio was proposed to yield

290 • Clinical Colorectal Cancer November 2006

Mercier CR.indd 4

12/19/06 1:35:27 PM

Cédric Mercier, Joseph Ciccolini more relevant information regarding the enzymatic activity. Sumi et al first theorized the utility of determining uracil-dihydrouracil ratios as a potential surrogate for DPD status in patients with cancer.101 Numerous methods have monitored uracil or, more frequently, uracil and dihydrouracil, sometimes along with thymine and dihydrothymine102 in urine103,104 or, more frequently, plasma,33,105-111 using UVHPLC105,108,109 and gas chromatography–mass spectrometry102,103,111 or liquid chromatography–mass spectrometry/mass spectometry104,112-114 systems. Clear correlations have been demonstrated between uracil-dihydrouracil values and 5-FU pharmacokinetic parameters108,113 or between ratio values and the occurrence of severe/lethal toxicities of 5-FU or capecitabine.33,49,51 More recently, a method known as the Uracil Breath Test has been proposed; this test is based on the ingestion of a stable isotope of uracil (13C uracil) and monitoring of expired 13C carbon dioxide.98,115,116 Following the same principle of exogenous uracil intake before monitoring in vivo catabolism, a simpler method that does not require labeled uracil or a specific analytic device gave promising preliminary results.117

5-Fluorouracil Test Dose Reduced test dose strategies with subsequent sampling for pharmacokinetic evaluation could provide valuable information on patients with impaired DPD and increased risk of iatrogeny upon 5-FU administration. Monitoring of 5-FU and 5-FU catabolites such as dihydrofluorouracil97,118 or FβAL119 in plasma has been proposed as a marker for DPD function. Precisely, it has been shown that FβAL plasma levels demonstrated good correlation with DPD activity in lymphocytes, thus suggesting it could be used to detect patients with deficient activity; however, this approach requires ≥ 1 dose of 5-FU to be administered before determining in real time the DPD status. A recently demonstrated pharmacokinetic-based test to prevent severe toxicities upon 5-FU administration showed that using a reduced 5-FU test dose with 5-FU/5-FU-H2 monitoring permitted one to detect approximately 2% of patients with marked alterations in 5-FU pharmacokinetic profiles. These patients were subsequently selected for alternative treatments without fluoropyrimidine drugs, thus preventing life-threatening toxicities.120 Another simpler approach consists of administration of low-dose 5-FU without pharmacokinetic support, with careful monitoring of toxicities during the first cycle of treatment and a possible dose escalation in the subsequent courses. Although rather empirical, this would ensure some safety for patients with possible DPD impairment, provided the patient remains hospitalized for monitoring.

Other Methods: Determining Dihydropyrimidine Dehydrogenase Genetic/Protein Expression Several methods for genetic or protein DPD measurement have been reported, essentially to determine DPD levels in tumors as a putative marker of response to 5-FU. Intratumoral thymidylate synthase and DPD expression has been extensively investigated and was a predictive marker of response

to 5-FU in many cancer types.121-123 Most of these methods evaluate the DPD expression through its mRNA with RT-PCR assays59,81,95,124 or directly by Western blot13,91,95,125 or immunohistochemistry methods.126 Importantly, all these methods have been developed to predict the sensitivity to 5-FU, with little application for detecting DPD-deficient patients at risk from fluoropyrimidine drugs. Although most of these methods provide good correlation between Western blot, DPD activity, and mRNA expression, they only provide semiquantitative data.91,95 More recently, fully quantitative enzyme-linked immunosorbent assays for DPD have been reported.127-131 This approach displays excellent correlations with enzyme activity,128,129 but to date, this approach has been developed and validated only in tumor biopsies of surrounding tissues as a marker of response to 5-FU.

Dihydropyrimidine Dehydrogenase Profiling: Pros and Cons Preliminary determination of DPD status in patients with cancer eligible for treatment with 5-FU or capecitabine should reduce the occurrence of severe/lethal toxicities. This would lead to optimizing clinical outcome through appropriate dosing, using DPD status as a covariate to be taken into account to customize dosage.132 In an ideal world, every patient should benefit from this approach. To achieve such a routine, largescale, systematic prescreening, one should use a rapid, simple, and inexpensive method that yields unambiguous data regarding the deficient/nondeficient status of patients with cancer. There are now a wide range of methods and strategies available to determine DPD status in patients with cancer likely to be given fluoropyrimidine drugs. Because mutations affecting the DPYD gene have been extensively investigated, genotypic studies might look attractive, because most of them are based upon simple, rapid, and widely used methods, such as RT-PCR technology, using µg quantities of patients’ genomic DNA. In addition, DPYD is highly polymorphic, and the fact that all mutations are probably not yet documented might yield false-negative results. Use of new DNA-binding dye such as LCGreen® should overcome this drawback in the near future through the generation of high-resolution melting curves for the detection of subtle alterations indicative of change in the hybridization status in the PCR product, but to date, no such methods are available for DPD genotyping. Besides, of the 40 mutations described, only the canonical IVS14+1A>G and the less frequent 1679 T>G polymorphisms have been frequently associated with decreased DPD activity,52,133-135 yet the actual clinical relevance of the detected single nucleotide polymorphisms remains unclear. For instance, the mutation reported as the most critical is the IVS14+1G>A polymorphism located on the exon 14, which results in a truncated protein.50 This has often been linked to the most severe toxicities of 5-FU treatment.26,52 However, recent studies have suggested that the implication of this exon-14 skipping might be less frequent than initially presumed in the occurrence of treatment-related side effects.86 In a retrospective 10-year Clinical Colorectal Cancer November 2006 • 291

Mercier CR.indd 5

12/19/06 1:35:27 PM

Profiling Dihydropyrimidine Dehydrogenase Deficiency study, 142 of 144 patients (98.6%) with severe toxicities with 5-FU did not display the IVS14+1G>A mutation,57 leading to serious concerns about its relevance as a predictive marker of severe toxicities with fluoropyrimidine drugs. In another study screening for exon 14 skipping in 105 patients with severe toxicities after 5-FU or capecitabine intake, none of them showed this mutation, including the 6 patients with fatal outcome.33 However, in the first case of toxic death reported after capecitabine intake, retrospective phenotypic determination of the DPD status showed marked DPD deficiency but without being associated with the IVS14+1G>A polymorphism. Further investigations instead revealed a heterozygosity for the 1896 C>T mutation located in exon 14 of the DPYD gene affecting the codon for F632, which could alter DPYD mRNA and, subsequently, protein expression.49 This strongly suggests that the canonical IVS14+1G>A polymorphism might not be a reliable, predictive marker of DPD deficiency and might be involved in subsequent increased risk of toxicities after 5-FU or capecitabine-based chemotherapies. Similar concerns have emerged from the study of other mutations such as the 85 T>C or 74 A>G polymorphisms on DPD activity and clinical outcome.76 The allelic regulations of the DPYD gene could ensure a proper enzymatic activity despite heterozygosity for sequence variations, with a risk of wrongly diagnosing deficiency syndrome in patients with restored DPD function.86 Finally, patients at risk after 5-FU or 5-FU–derivative intake from DPD deficiency caused by exogenous factors such as drug interactions30,54,136 will not be identified as deficient using any genotypic methods. Taken together, this strongly suggests that a limited number of patients with cancer could benefit from such genotypic screening, with an increased risk of precluding the proper diagnosis in routine clinical settings. In many respects, considering DPD activity as an ultimate endpoint to detect patients with impaired DPD, regardless of causes, might look safer. Several direct enzymatic assays in peripheral blood mononuclear cells (PBMCs) or fibroblasts have been proposed for 20 years now, but all require large (up to 60 mL) volumes of blood and time-consuming multistep sample treatment, and most are based on the use of radiolabeled substrates, even if a couple of recent methods are based on nonradioactive material.96 Besides these technical limitations, several biases hinder these methods. Isolating PBMCs is a long procedure that can hardly be automated, DPD activity can vary according to the subfraction of monocytes,72,90 and only a weak link was found between DPD activity in PBMCs and 5-FU clearances.24,100,118,120 Besides significant relationships between liver and PBMCs, DPD activity showed mild correlation (r² < 0.3),87 thus hampering proper diagnosis, especially in patients with intermediate deficiencies.90 Simpler and cheaper methods using surrogate markers as an index for DPD status is the most attractive approach for detecting DPD deficiencies in patients with cancer. Monitoring physiologic uracil and dihydrouracil in plasma as a ratio is a well-established strategy that combines simple sample treatment and analytical procedures such as basic UV-HPLC. Retrospective

studies have proven that 70% of the severe toxicities and 80% of the toxic deaths caused by 5-FU/capecitabine treatment would have been detected using this approach,33,47,51 yet interpretation of the ratios can vary from one institute to another based on the methods used, and most laboratories have performed local reference-population studies before setting their own cutoff indicative of a DPD deficiency. Indeed, uracil-dihydrouracil ratios are generated from calibration curves built from the original biologic matrix,106,109,113 water,105,107,108 or from a simple signal/signal basis,33,47,49 with subsequent large variations in the values reported between institutes. This is illustrated by the fact that mean molar ratios observed for dihydrouracil/uracil levels in urine ranged from 0.42 to 4.8 according to the populations studied.112 Finally, it is still not clear which of the ratios measured in plasma or urine would give the most relevant information on DPD status in patients with cancer. Higher concentrations of uracil and dihydrouracil found in urine would theoretically allow a better analytic determination while being noninvasive, but urine levels of uracil are more likely to be influenced by food intake, thus leading to a possible bias in the determination of the ratio. Recently, the Uracil Breath Test has been proposed as an alternative test to detect DPD deficiency in patients. The advantage of this noninvasive test is that it is expedited by the use of exogenous 2-13C uracil as a probe. However, subjects are asked to spend up to 3 hours blowing into dozens of bags every 5-10 minutes,98 a “patient-unfriendly” requirement compared with a single 3-mL blood sample to perform most uracil-dihydrouracil ratio determinations in plasma. Besides, this test requires specific equipment such as an infrared spectrophotometer breath analyzer, whereas dihydrouracil/uracil monitoring can be performed with basic HPLC already widely available in most hospitals. Finally, a major drawback of this method is the amount of 2-13C uracil to be ingested by the patients (6 mg/kg), which renders this procedure extremely costly, especially for large-scale systematic screening. The oral uracil challenge test requiring a nonlabeled probe, single blood sample, and UV-HPLC procedure could be an attractive alternative, although only preliminary results are available so far.137 Still, whatever is the phenotypic method chosen, one will have to deal with the issue of circadian regulation of DPD activity. Most tests are performed empirically in the morning as an attempt to limit the influence of intrapatient variability, although no data support this hypothesis, and a more significant circadian pattern has been evidenced when DPD function is expressed as uracil-dihydrouracil ratios, compared with the actual activity in PBMCs.114 Based on these drawbacks, administering test doses of fluoropyrimidine drugs, with pharmacokinetic support, would appear as the less biased approach for detecting DPD impairment in patients with cancer. The major advantage of this strategy is that actual drug administration is monitored in real time, without the experimental bias encountered in prescreening methods. Still, the reduced dose administered must be high enough to allow pharmacokinetic monitoring and, therefore, would lead to life-threatening toxicities in patients with totally abrogated

292 • Clinical Colorectal Cancer November 2006

Mercier CR.indd 6

12/19/06 1:35:27 PM

Cédric Mercier, Joseph Ciccolini DPD activity. Furthermore, even if no pharmacokinetics are required, patients must be hospitalized for careful monitoring of toxicities, thus excluding the ones treated with ambulatory therapies such as capecitabine.

Conclusion Choosing a method for identifying DPD-deficient patients at risk with fluoropyrimidine drugs remains an uneasy task. No method has stood out as a standard that would meet all the requirements (eg, time- and cost-effectiveness, availability, and relevance) of large-scale screening. However, whatever method is eventually chosen, it is still a better option than sticking with the blind administration of standardized dosages of 5-FU performed regardless of the DPD status of patients with cancer. Because of the countless reports demonstrating the relationships between DPD deficiency and severe toxicities with 5-FU/capecitabine, specific sensitive screening methods for detecting DPD expression are warranted.

16. 17. 18. 19. 20. 21. 22. 23. 24.

References 1. 2.

3. 4. 5.

6.

7.

8.

9. 10. 11. 12. 13. 14. 15.

Peters GJ, van Groeningen CJ, Laurensse EJ, et al. A comparison of 5-fluorouracil metabolism in human colorectal cancer and colon mucosa. Cancer 1991; 68:1903-1909. Peters GJ, van Groeningen CJ, Laurensse EJ, et al. Thymidylate synthase from untreated human colorectal cancer and colonic mucosa: enzyme activity and inhibition by 5-fluoro-2’-deoxy-uridine-5’monophosphate. Eur J Cancer 1991; 27:263-267. Cuq P, Rouquet C, Evrard A, et al. Fluoropyrimidine sensitivity of human MCF-7 breast cancer cells stably transfected with human uridine phosphorylase. Br J Cancer 2001; 84:1677-1680. Cao D, Pizzorno G. Uridine phosophorylase: an important enzyme in pyrimidine metabolism and fluoropyrimidine activation. Drugs Today (Barc) 2004; 40:431-443. Evrard A, Cuq P, Ciccolini J, et al. Increased cytotoxicity and bystander effect of 5-fluorouracil and 5-deoxy-5-fluorouridine in human colorectal cancer cells transfected with thymidine phosphorylase. Br J Cancer 1999; 80:1726-1733. Ciccolini J, Peillard L, Evrard A, et al. Enhanced antitumor activity of 5-fluorouracil in combination with 2’-deoxyinosine in human colorectal cell lines and human colon tumor xenografts. Clin Cancer Res 2000; 6:1529-1535. Ciccolini J, Peillard L, Aubert C, et al. Monitoring of the intracellular activation of 5-fluorouracil to deoxyribonucleotides in HT29 human colon cell line: application to modulation of metabolism and cytotoxicity study. Fundam Clin Pharmacol 2000; 14:147-154. Ciccolini J, Cuq P, Evrard A, et al. Combination of thymidine phosphorylase gene transfer and deoxyinosine treatment greatly enhances 5-fluorouracil antitumor activity in vitro and in vivo. Mol Cancer Ther 2001; 1:133-139. Fanciullino R, Evrard A, Cuq P, et al. Genetic and biochemical modulation of 5-fluorouracil through the overexpression of thymidine kinase: an in-vitro study. Anticancer Drugs 2006; 17:463-470. Ciccolini J, Evrard A, Cuq P. Thymidine phosphorylase and fluoropyrimidines efficacy: a Jekyll and Hyde story. Curr Med Chem Anticancer Agents 2004; 4:71-81. Sommadossi JP, Gewirtz DA, Diasio RB, et al. Rapid catabolism of 5-fluorouracil in freshly isolated rat hepatocytes as analyzed by high performance liquid chromatography. J Biol Chem 1982; 257:8171-8176. Mentre F, Steimer JL, Sommadossi JP, et al. A mathematical model of the kinetics of 5-fluorouracil and its catabolites in freshly isolated rat hepatocytes. Biochem Pharmacol 1984; 33:2727-2732. Lu ZH, Zhang R, Diasio RB. Purification and characterization of dihydropyrimidine dehydrogenase from human liver. J Biol Chem 1992; 267:17102-17109. Aubert C, Sommadossi JP, Coassolo P, et al. Quantitative analysis of 5fluorouracil and 5,6-dihydrofluorouracil in plasma by gas chromatography mass spectrometry. Biomed Mass Spectrom 1982; 9:336-339. Etienne MC, Lagrange JL, Dassonville O, et al. Population study of

25. 26. 27. 28.

29.

30. 31. 32. 33. 34. 35. 36. 37.

38. 39.

dihydropyrimidine dehydrogenase in cancer patients. J Clin Oncol 1994; 12:2248-2253. Milano G, Etienne MC, Cassuto-Viguier E, et al. Influence of sex and age on fluorouracil clearance. J Clin Oncol 1992; 10:1171-1175. Harris BE, Carpenter JT, Diasio RB. Severe 5-fluorouracil toxicity secondary to dihydropyrimidine dehydrogenase deficiency. A potentially more common pharmacogenetic syndrome. Cancer 1991; 68:499-501. Grem JL, Yee LK, Venzon DJ, et al. Inter- and intraindividual variation in dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells. Cancer Chemother Pharmacol 1997; 40:117-125. Yamaguchi K, Arai Y, Kanda Y, et al. Germline mutation of dihydropyrimidine dehydrogenese gene among a Japanese population in relation to toxicity to 5-fluorouracil. Jpn J Cancer Res 2001; 92:337-342. Sohn DR, Cho MS, Chung PJ. Dihydropyrimidine dehydrogenase activity in a Korean population. Ther Drug Monit 1999; 21:152-154. Ridge SA, Sludden J, Brown O, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in Caucasian subjects. Br J Clin Pharmacol 1998; 46:151-156. Morsman JM, Sludden J, Ameyaw MM, et al. Evaluation of dihydropyrimidine dehydrogenase activity in South-west Asian, Kenyan and Ghanaian populations. Br J Clin Pharmacol 2000; 50:269-272. Tuchman M, Roemeling RV, Hrushesky WA, et al. Dihydropyrimidine dehydrogenase activity in human blood mononuclear cells. Enzyme 1989; 42:15-24. Fleming RA, Milano G, Thyss A, et al. Correlation between dihydropyrimidine dehydrogenase activity in peripheral mononuclear cells and systemic clearance of fluorouracil in cancer patients. Cancer Res 1992; 52:2899-2902. Fleming RA, Milano G, Gaspard MH, et al. Dihydropyrimidine dehydrogenase activity in cancer patients. Eur J Cancer 1993; 29A:740-744. van Kuilenburg AB. Dihydropyrimidine dehydrogenase and the efficacy and toxicity of 5-fluorouracil. Eur J Cancer 2004; 40:939-950. Ezzeldin HH, Lee AM, Mattison LK, et al. Methylation of the DPYD promoter: an alternative mechanism for dihydropyrimidine dehydrogenase deficiency in cancer patients. Clin Cancer Res 2005; 11:8699-8705. Sato K, Kitajima Y, Miyoshi A, et al. Deficient expression of the DPD gene is caused by epigenetic modification in biliary tract cancer cells, and induces high sensitivity to 5-FU treatment. Int J Oncol 2006; 29:429-435. Mattison LK, Fourie J, Carpenter M, et al. Evidence for increased incidence of dihydropyrimidine dehydrogenase (DPD) deficiency in African Americans compared to Caucasians. J Clin Oncol 2005; 23:136s (Abstract #2004). Okuda H, Nishiyama T, Ogura K, et al. Lethal drug interactions of sorivudine, a new antiviral drug, with oral 5-fluorouracil prodrugs. Drug Metab Dispos 1997; 25:270-273. Nozoe Y, Ogata Y, Araki Y, et al. Up-regulation in dihydropyrimidine dehydrogenase activity by raltitrexed causes antagonism in combination with 5-fluorouracil. Anticancer Res 2003; 23:4663-4669. Milano G, Etienne MC, Pierrefite V, et al. Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity. Br J Cancer 1999; 79:627-630. Ciccolini J, Mercier C, Evrard A, et al. A rapid and inexpensive method for anticipating severe toxicity to fluorouracil and fluorouracil-based chemotherapy. Ther Drug Monit 2006; 28:678-685. Jodrell DI, Stewart M, Aird R, et al. 5-fluorouracil steady state pharmacokinetics and outcome in patients receiving protracted venous infusion for advanced colorectal cancer. Br J Cancer 2001; 84:600-603. Terret C, Erdociain E, Guimbaud R, et al. Dose and time dependencies of 5-fluorouracil pharmacokinetics. Clin Pharmacol Ther 2000; 68:270-279. Port RE, Daniel B, Ding RW, et al. Relative importance of dose, body surface area, sex, and age for 5-fluorouracil clearance. Oncology 1991; 48:277-281. Lu Z, Zhang R, Diasio RB. Population characteristics of hepatic dihydropyrimidine dehydrogenase activity, a key metabolite enzyme in 5-fluorouracil chemotherapy. Clin Pharmacol Ther 1995; 58:512-522. Fleming GF, Schilsky RL, Schumm LP, et al. Phase I and pharmacokinetic study of 24-hour infusion 5-fluorouracil and leucovorin in patients with organ dysfunction. Ann Oncol 2003; 14:1142-1147. van Kuilenburg AB, Haasjes J, Richel DJ, et al. Clinical implications of dihydropyrimidine dehydrogenase (DPD) deficiency in patients with severe 5-fluorouracil-associated toxicity: identification of new

Clinical Colorectal Cancer November 2006 • 293

Mercier CR.indd 7

12/19/06 1:35:28 PM

Profiling Dihydropyrimidine Dehydrogenase Deficiency 40. 41. 42. 43. 44. 45.

46.

47. 48.

49.

50.

51.

52. 53.

54.

55.

56.

57.

58.

59.

mutations in the DPD gene. Clin Cancer Res 2000; 6:4705-4712. Santini J, Milano G, Thyss A, et al. 5-FU therapeutic monitoring with dose adjustement leads to an improved therapeutic index in head and neck cancer. Br J Cancer 1989; 59:287-290. Diasio RB. The role of dihydropyrimidine dehydrogenase (DPD) modulation in 5-FU pharmacology. Oncology (Williston Park) 1998; 12:23-27. Maring JG, Piersma H, van Dalen A, et al. Extensive hepatic replacement due to liver metastases has no effect on 5-fluorouracil pharmacokinetics. Cancer Chemother Pharmacol 2003; 51:167-173. Fleming RA, Milano GA, Etienne MC, et al. No effect of dose, hepatic function, or nutritional status on 5-FU clearance following continuous (5-day), 5-FU infusion. Br J Cancer 1992; 66:668-672. Heggie GD, Sommadossi JP, Cross DS, et al. Clinical pharmacokinetics of 5-fluorouracil and its metabolites in plasma, urine, and bile. Cancer Res 1987; 47:2203-2206. Diasio RB, Beavers TL, Carpenter JT. Familial deficiency of dihydropyrimidine dehydrogenase. Biochemical basis for familial pyrimidinemia and severe 5-fluorouracil-induced toxicity. J Clin Invest 1988; 81:47-51. Bocci G, Danesi R, Allegrini G, et al. Severe 5-fluorouracil toxicity associated with a marked alteration of pharmacokinetics of 5-fluorouracil and its catabolite 5-fluoro-5,6-dihydrouracil: a case report. Eur J Clin Pharmacol 2002; 58:593-595. Ciccolini J, Mercier C, Dahan L, et al. Implication of DPD deficiency in severe or lethal toxicities after 5-fluorouracil and capecitabine-based chemotherapies. Proc Assoc Amer Cancer Res 2005; 46:942 (Abstract #3998). Tsukamoto Y, Kato Y, Ura M, et al. A physiologically based pharmacokinetic analysis of capecitabine, a triple prodrug of 5-FU, in humans: the mechanism for tumor-selective accumulation of 5-FU. Pharm Res 2001; 18:1190-1202. Ciccolini J, Mercier C, Dahan L, et al. Toxic death-case after capecitabine + oxaliplatin (XELOX) administration: probable implication of dihydropyrimidine deshydrogenase deficiency. Cancer Chemother Pharmacol 2006; 58:272-275. van Kuilenburg AB, Meinsma R, Zoetekouw L, et al. High prevalence of the IVS14 + 1G>A mutation in the dihydropyrimidine dehydrogenase gene of patients with severe 5-fluorouracil-associated toxicity. Pharmacogenetics 2002; 12:555-558. Mercier C, Yang C, Ciccolini J, et al. Determination of uracil/UH2 ratio as potential surrogate for DPD status in cancer patients presenting with severe toxicities during fluoropyrimidine treatment. J Clin Oncol 2006; 24(18 suppl):84s (Abstract #2020). Johnson MR, Hageboutros A, Wang K, et al. Life-threatening toxicity in a dihydropyrimidine dehydrogenase-deficient patient after treatment with topical 5-fluorouracil. Clin Cancer Res 1999; 5:2006-2011. Ezzeldin H, Johnson MR, Okamoto Y, et al. Denaturing high performance liquid chromatography analysis of the DPYD gene in patients with lethal 5-fluorouracil toxicity. Clin Cancer Res 2003; 9:3021-3028. Okuda H, Ogura K, Kato A, et al. A possible mechanism of eighteen patient deaths caused by interactions of sorivudine, a new antiviral drug, with oral 5-fluorouracil prodrugs. J Pharmacol Exp Ther 1998; 287:791-799. Raida M, Schwabe W, Hausler P, et al. Prevalence of a common point mutation in the dihydropyrimidine dehydrogenase (DPD) gene within the 5'-splice donor site of intron 14 in patients with severe 5-fluorouracil (5-FU)- related toxicity compared with controls. Clin Cancer Res 2001; 7:2832-2839. Lyss AP, Lilenbaum RC, Harris BE, et al. Severe 5-fluorouracil toxicity in a patient with decreased dihydropyrimidine dehydrogenase activity. Cancer Invest 1993; 11:239-240. Magne N, Renee N, Formento J, et al. Prospective study of dihydropyrimidine dehydrogenase (DPD) activity and DPYD IVS14+1G>A mutation in patients developing FU-related toxicities: an updated analysis based on ten-year recruitment across multiple French institutions. J Clin Oncol 2005; 23(16 suppl):135s (Abstract #2003). Hooiveld EA, van Kuilenburg AB, Haanen JB, et al. Severe toxicity after treatment with capecitabine and fluorouracil due to partial dihydropyrimidine dehydrogenase deficiency. Ned Tijdschr Geneeskd 2004; 148:626-628. Meinsma R, Fernandez-Salguero P, van Kuilenburg AB, et al. Human polymorphism in drug metabolism: mutation in the dihydropyrimidine dehydrogenase gene results in exon skipping and thymine

60. 61. 62. 63.

64. 65. 66.

67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78.

79.

80. 81. 82. 83.

uracilurea. DNA Cell Biol 1995; 14:1-6. Wadman SK, Berger R, Duran M, et al. Dihydropyrimidine dehydrogenase deficiency leading to thymine-uraciluria. An inborn error of pyrimidine metabolism. J Inherit Metab Dis 1985; 8(suppl 2):113-114. Au KM, Lai CK, Yuen YP, et al. Diagnosis of dihydropyrimidine dehydrogenase deficiency in a neonate with thymine-uraciluria. Hong Kong Med J 2003; 9:130-132. Al Sanna’a NA, van Kuilenburg AB, Atrak TM, et al. Dihydropyrimidine dehydrogenase deficiency presenting at birth. J Inherit Metab Dis 2005; 28:793-796. Yokota H, Fernandez-Salguero P, Furuya H, et al. cDNA cloning and chromosome mapping of human dihydropyrimidine dehydrogenase, an enzyme associated with 5-fluorouracil toxicity and congenital thymine uraciluria. J Biol Chem 1994; 269:23192-23196. Johnson MR, Wang K, Tillmanns S, et al. Structural organization of the human dihydropyrimidine dehydrogenase gene. Cancer Res 1997; 57:1660-1663. van Kuilenburg AB, De Abreu RA, Van Gennip AH. Pharmacogenetic and clinical aspects of dihydropyrimidine dehydrogenase deficiency. Ann Clin Biochem 2003; 40:41-45. Hasegawa T, Kim HS, Fukushima M, et al. Sequence analysis of the 5'-flanking regions of human dihydropyrimidine dehydrogenase gene: identification of a new polymorphism related with effects of 5-fluorouracil. Nucleosides Nucleotides Nucleic Acids 2005; 24:233-242. Ridge SA, Sludden J, Wei X, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in patients with colorectal cancer. Br J Cancer 1998; 77:497-500. McLeod HL, Sludden J, Murray GI, et al. Characterization of dihydropyrimidine dehydrogenase in human colorectal tumours. Br J Cancer 1998; 77:461-465. Innocenti F, Iyer L, Ratain MJ. Pharmacogenetics: a tool for individualizing antineoplastic therapy. Clin Pharmacokinet 2000; 39:315-325. Allegra CJ. Dihydropyrimidine dehydrogenase activity: prognostic partner of 5-fluorouracil? Clin Cancer Res 1999; 5:1947-1949. Wei X, McLeod HL, McMurrough J, et al. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J Clin Invest 1996; 98:610-615. van Kuilenburg AB, Vreken P, Abeling NG, et al. Genotype and phenotype in patients with dihydropyrimidine dehydrogenase deficiency. Hum Genet 1999; 104:1-9. Johnson MR, Wang K, Diasio RB. Profound dihydropyrimidine dehydrogenase deficiency resulting from a novel compound heterozygote genotype. Clin Cancer Res 2002; 8:768-774. Gross E, Seck K, Neubauer S, et al. High-throughput genotyping by DHPLC of the dihydropyrimidine dehydrogenase gene implicated in (fluoro)pyrimidine catabolism. Int J Oncol 2003; 22:325-332. Kouwaki M, Hamajima N, Sumi S, et al. Identification of novel mutations in the dihydropyrimidine dehydrogenase gene in a Japanese patient with 5-fluorouracil toxicity. Clin Cancer Res 1998; 4:2999-3004. Hsiao HH, Yang MY, Chang JG, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in the Taiwanese population. Cancer Chemother Pharmacol 2004; 53:445-451. Yu J, McLeod HL, Ezzeldin HH, et al. Methylation of the DPYD promoter and dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res 2006; 12:3864. Vreken P, van Kuilenburg AB, Meinsma R, et al. Dihydropyrimidine dehydrogenase (DPD) deficiency: identification and expression of missense mutations C29R, R886H and R235W. Hum Genet 1997; 101:333-338. Nauck M, Gierens H, Marz W, et al. Rapid detection of a common dihydropyrimidine dehydrogenase mutation associated with 5-fluorouracil toxicity and congenital thymine uraciluria using fluorogenic hybridization probes. Clin Biochem 2001; 34:103-105. Johnston SJ, Ridge SA, Cassidy J, et al. Regulation of dihydropyrimidine dehydrogenase in colorectal cancer. Clin Cancer Res 1999; 5:2566-2570. Johnson MR, Wang K, Smith JB, et al. Quantitation of dihydropyrimidine dehydrogenase expression by real-time reverse transcription polymerase chain reaction. Ann Biochem 2000; 278:175-184. Fischer J, Schwab M, Eichelbaum M, et al. Mutational analysis of the human dihydropyrimidine dehydrogenase gene by denaturing highperformance liquid chromatography. Genet Test 2003; 7:97-105. Ezzeldin H, Okamoto Y, Johnson MR, et al. A high-throughput denaturing high-performance liquid chromatography method for the

294 • Clinical Colorectal Cancer November 2006

Mercier CR.indd 8

12/19/06 1:35:28 PM

Cédric Mercier, Joseph Ciccolini

84.

85. 86. 87. 88. 89. 90. 91.

92.

93.

94. 95.

96.

97.

98. 99.

100. 101.

102.

103.

identification of variant alleles associated with dihydropyrimidine dehydrogenase deficiency. Ann Biochem 2002; 306:63-73. van Kuilenburg AB, Muller EW, Haasjes J, et al. Lethal outcome of a patient with a complete dihydropyrimidine dehydrogenase (DPD) deficiency after administration of 5-fluorouracil: frequency of the common IVS14+1G>A mutation causing DPD deficiency. Clin Cancer Res 2001; 7:1149-1153. Celik I, Kars A, Guc D, et al. Dihydropyrimidine dehydrogenase enzyme deficiency: clinical and genetic assessment of prevalence in Turkish cancer patients. Cancer Invest 2002; 20:333-339. Collie-Duguid ES, Etienne MC, Milano G, et al. Known variant DPYD alleles do not explain DPD deficiency in cancer patients. Pharmacogenetics 2000; 10:217-223. Chazal M, Etienne MC, Renee N, et al. Link between dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells and liver. Clin Cancer Res 1996; 2:507-510. Milano G, Etienne MC. Dihydropyrimidine dehydrogenase (DPD) and clinical pharmacology of 5-fluorouracil (review). Anticancer Res 1994; 14:2295-2297. Naguib FN, el Kouni MH, Cha S. Enzymes of uracil catabolism in normal and neoplastic human tissues. Cancer Res 1985; 45:5405-5412. van Kuilenburg AB, Van Lenthe H, Tromp A, et al. Pitfalls in the diagnosis of patients with a partial dihydropyrimidine dehydrogenase deficiency. Clin Chem 2000; 46:9-17. Lu Z, Zhang R, Diasio RB. Dihydropyrimidine dehydrogenase activity in human peripheral blood mononuclear cells and liver: population characteristics, newly identified deficient patients, and clinical implication in 5-fluorouracil chemotherapy. Cancer Res 1993; 53:5433-5438. Katsumata K, Tomioka H, Sumi T, et al. Correlation between clinicopathologic factors and kinetics of metabolic enzymes for 5-fluorouracil given to patients with colon carcinoma by two different dosage regimens. Cancer Chemother Pharmacol 2003; 51:155-160. Johnson MR, Yan J, Shao L, et al. Semi-automated radioassay for determination of dihydropyrimidine dehydrogenase (DPD) activity. Screening cancer patients for DPD deficiency, a condition associated with 5-fluorouracil toxicity. J Chromatogr B Biomed Sci Appl 1997; 696:183-191. Etienne MC, Milano G, Fleming RA, et al. Dihydropyrimidine dehydrogenase activity in lymphocytes: predictive factor for 5-fluorouracil clearance. Bull Cancer 1992; 79:1159-1163. Fernandez-Salguero P, Gonzalez FJ, Etienne MC, et al. Correlation between catalytic activity and protein content for the polymorphically expressed dihydropyrimidine dehydrogenase in human lymphocytes. Biochem Pharmacol 1995; 50:1015-1020. Di Paolo A, Danesi R, Ciofi L, et al. Improved analysis of 5-fluorouracil and 5,6-dihydro-5-fluorouracil by HPLC with diode array detection for determination of cellular dihydropyrimidine dehydrogenase activity and pharmacokinetic profiling. Ther Drug Monit 2005; 27:362-368. Deporte-Fety R, Picot M, Amiand M, et al. High-performance liquid chromatographic assay with ultraviolet detection for quantification of dihydrofluorouracil in human lymphocytes: application to measurement of dihydropyrimidine dehydrogenase activity. J Chromatogr B Biomed Sci Appl 2001; 762:203-209. Saif MW, Mattison L, Carollo T, et al. Dihydropyrimidine dehydrogenase deficiency in an Indian population. Cancer Chemother Pharmacol 2006; 58:396-401. Milano G, Etienne MC. Potential importance of dihydropyrimidine dehydrogenase (DPD) in cancer chemotherapy. Pharmacogenetics 1994; 4:301-306. Milano G, Etienne MC. Individualizing therapy with 5-fluorouracil related to dihydropyrimidine dehydrogenase: theory and limits. Ther Drug Monit 1996; 18:335-340. Sumi S, Kidouchi K, Ohba S, et al. Automated screening system for purine and pyrimidine metabolism disorders using high performance liquid chromatography. J Chromatogr B Biomed Sci Appl 1995; 672:233-239. Kuhara T, Ohdoi C, Ohse M, et al. Rapid gas chromatographic-mass spectrometric diagnosis of dihydropyrimidine dehydrogenase deficiency and dihydropyrimidinase deficiency. J Chromatogr B Analyt Technol Biomed Life Sci 2003; 792:107-115. Kuhara T, Ohdoi C, Ohse M. Simple gas chromatographic-mass spectrometric procedure for diagnosing pyrimidine degradation defects for prevention of severe anticancer side effects. J Chromatogr B Biomed Sci

Appl 2001; 758:61-74. 104. Jiang H, Jiang J, Hu P, et al. Measurement of endogenous uracil and dihydrouracil in plasma and urine of normal subjects by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 769:169-176. 105. Remaud G, Boisdron-Celle M, Hameline C, et al. An accurate dihydrouracil/uracil determination using improved high performance liquid chromatography method for preventing fluoropyrimidines-related toxicity in clinical practice. J Chromatogr B Analyt Technol Biomed Life Sci 2005; 823:98-107. 106. Garg M, Sevester JC, Sakoff JA, et al. Simple liquid chromatographic method for the determination of uracil and dihydrouracil plasma levels: a potential pretreatment predictor of 5-fluorouracil toxicity. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 774:223-230. 107. Gamelin E, Boisdron-Celle M, Larra F, et al. A simple chromatographic method for the analysis of pyrimidines and their dihydrogenated metabolites. J Liq Chromatogr Relat Technol 1997; 20:3155-3172. 108. Gamelin E, Boisdron-Celle M, Guerin-Meyer V, et al. Correlation between uracil and dihydrouracil plasma ratio, fluorouracil (5-FU) pharmacokinetic parameters, and tolerance in patients with advanced colorectal cancer: a potential interest for predicting 5-FU toxicity and determining optimal 5-FU dosage. J Clin Oncol 1999; 17:1105. 109. Deporte R, Amiand M, Moreau A, et al. High performance liquid chromatographic assay with UV detection for measurement of dihydrouracil/uracil ratio in plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 834:170-177. 110. Ciccolini J, Mercier C, Blachon MF, et al. A simple and rapid highperformance liquid chromatographic (HPLC) method for 5-fluorouracil (5-FU) assay in plasma and possible detection of patients with impaired dihydropyrimidine dehydrogenase (DPD) activity. J Clin Pharm Ther 2004; 29:307-315. 111. Bi D, Anderson LW, Shapiro J, et al. Measurement of plasma uracil using gas chromatography-mass spectrometry in normal individuals and in patients receiving inhibitors of dihydropyrimidine dehydrogenase. J Chromatogr B Biomed Sci Appl 2000; 738:249-258. 112. Sparidans RW, Bosch TM, Jorger M, et al. Liquid chromatographytandem mass spectrometric assay for the analysis of uracil, 5,6-dihydrouracil and beta-ureidopropionic acid in urine for the measurement of the activities of the pyrimidine catabolic enzymes. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 839:45-53. 113. Jiang H, Lu J, Jiang J, et al. Important role of the dihydrouracil/uracil ratio in marked interpatient variations of fluoropyrimidine pharmacokinetics and pharmacodynamics. J Clin Pharmacol 2004; 44:1260-1272. 114. Jiang H, Lu J, Ji J. Circadian rhythm of dihydrouracil/uracil ratios in biological fluids: a potential biomarker for dihydropyrimidine dehydrogenase levels. Br J Pharmacol 2004; 141:616-623. 115. Mattison LK, Ezzeldin H, Carpenter M, et al. Rapid identification of dihydropyrimidine dehydrogenase deficiency by using a novel 2-13Curacil breath test. Clin Cancer Res 2004; 10:2652-2658. 116. Mattison LK, Fourie J, Hirao Y, et al. The uracil breath test in the assessment of dihydropyrimidine dehydrogenase activity: pharmacokinetic relationship between expired 13CO2 and plasma [213C]dihydrouracil. Clin Cancer Res 2006; 12:549-555. 117. Maring JG. DPD Phenotyping in Human Volunteers and a DPD Deficient Patient by Assessing Uracil Pharmacokinetics After an Oral Uracil Test Dose. A Preliminary Report, in “New Insight of Pyrimidine Antagonists Chemotherapy” [master’s thesis]. Gronigen, NL: University of Gronigen; 2005:102-111. 118. Bocci G, Danesi R, Di Paolo AD, et al. Comparative pharmacokinetic analysis of 5-fluorouracil and its major metabolite 5-fluoro-5,6-dihydrouracil after conventional and reduced test dose in cancer patients. Clin Cancer Res 2000; 6:3032-3037. 119. Furuhata T, Kawakami M, Okita K, et al. Plasma level of a 5-fluorouracil metabolite, fluoro-beta-alanine correlates with dihydropyrimidine dehydrogenase activity of peripheral blood mononuclear cells in 5fluorouracil treated patients. J Exp Clin Cancer Res 2006; 25:79-82. 120. Bocci G, Barbara C, Vannozzi F, et al. A pharmacokinetic-based test to prevent severe 5-fluorouracil toxicity. Clin Pharmacol Ther 2006; 80:384-395. 121. Shintani Y, Ohta M, Hirabayashi H, et al. Thymidylate synthase and dihydropyrimidine dehydrogenase mRNA levels in tumor tissues and the efficacy of 5-fluorouracil in patients with non-small-cell lung cancer. Lung Cancer 2004; 45:189-196. 122. Matsuyama R, Togo S, Shimizu D, et al. Predicting 5-fluorouracil

Clinical Colorectal Cancer November 2006 • 295

Mercier CR.indd 9

12/19/06 1:35:28 PM

Profiling Dihydropyrimidine Dehydrogenase Deficiency

123.

124.

125.

126.

127.

128. 129.

chemosensitivity of liver metastases from colorectal cancer using primary tumor specimens: three-gene expression model predicts clinical response. Int J Cancer 2006; 119:406-413. Huang CL, Yokomise H, Kobayashi S, et al. Intratumoral expression of thymidylate synthase and dihydropyrimidine dehydrogenase in non-small cell lung cancer patients treated with 5-FU-based chemotherapy. Int J Oncol 2000; 17:47-54. Okumura K, Shiomi H, Mekata E, et al. Correlation between chemosensitivity and mRNA expression level of 5-fluorouracil-related metabolic enzymes during liver metastasis of colorectal cancer. Oncol Rep 2006; 15:875-882. Takechi T, Okabe H, Fujioka A, et al. Relationship between protein levels and gene expression of dihydropyrimidine dehydrogenase in human tumor cells during growth in culture and in nude mice. Jpn J Cancer Res 1998; 89:1144-1153. Takenoue T, Kitayama J, Takei Y, et al. Characterization of dihydropyrimidine dehydrogenase on immunohistochemistry in colon carcinoma, and correlation between immunohistochemical score and protein level or messenger RNA expression. Ann Oncol 2000; 11:273-279. Okita A, Tsukuda K, Murakami M, et al. Thymidine phosphorylase and dihydropyrimidine dehydrogenase expression levels in tumor and normal tissue specimens of T3 human colorectal carcinoma. Surg Today 2006; 36:348-353. Oi K, Makino M, Ozaki M, et al. Immunohistochemical dihydropyrimidine dehydrogenase expression is a good prognostic indicator for patients with Dukes’ C colorectal cancer. Anticancer Res 2004; 24:273-279. Kurebayashi J, Yamamoto Y, Udagawa K, et al. Establishment of enzyme-linked immunosorbent assays for thymidylate synthase and

130. 131. 132. 133. 134. 135. 136.

137.

dihydropyriminide dehydrogenase in cancer tissues. Oncol Rep 2004; 11:973-979. Ikeguchi M, Hirooka Y, Makino M, et al. Dihydropyrimidine dehydrogenase activity of cancerous and non-cancerous tissues in liver and large intestine. Oncol Rep 2001; 8:621-625. Hotta T, Takifuji K, Taniguchi K, et al. The relationship between survival and the expression of dihydropyrimidine dehydrogenase in patients with colorectal cancer. Oncol Rep 2006; 16:177-182. van Kuilenburg AB. Screening for dihydropyrimidine dehydrogenase deficiency: to do or not to do, that’s the question? Cancer Invest 2006; 24:215-217. Mattison LK, Soong R, Diasio RB. Implications of dihydropyrimidine dehydrogenase on 5-fluorouracil pharmacogenetics and pharmacogenomics. Pharmacogenomics 2002; 3:485-492. Johnson MR, Diasio RB. Importance of dihydropyrimidine dehydrogenase (DPD) deficiency in patients exhibiting toxicity following treatment with 5-fluorouracil. Adv Enzyme Regul 2001; 41:151-157. Gardiner SJ, Begg EJ, Robinson BA. The effect of dihydropyrimidine dehydrogenase deficiency on outcomes with fluorouracil. Adverse Drug React Toxicol Rev 2002; 21:1-16. Kanamitsu SI, Ito K, Okuda H, et al. Prediction of in vivo drugdrug interactions based on mechanism-based inhibition from in vitro data: inhibition of 5-fluorouracil metabolism by (E)-5-(2Bromovinyl)uracil. Drug Metab Dispos 2000; 28:467-474. Maring JG, Oonq BN, de Vries EG. Plasma pharmacokinetics of uracil after an oral uracil challenge dose for dihydropyrimidine dehydrogenase (DPD) phenotyping. Proc Am Soc Clin Oncol 2004; 23:157 (Abstract #2120).

296 • Clinical Colorectal Cancer November 2006

Mercier CR.indd 10

12/19/06 1:35:28 PM

CCC 6.4 Cover.indd

Nov 1, 2006 - Address for correspondence: Cédric Mercier, MD, Service d'Oncologie. Médicale, CHU ... Fax: 33-491-835-667; e-mail: [email protected]. Profiling ..... a long procedure that can hardly be automated, DPD activity can vary ...

152KB Sizes 2 Downloads 242 Views

Recommend Documents

CCC / CCC+ EXAMINATION REGISTRATION FORM
NOTE : ALL INFORMATION SHOULD BE FILL IN ENGLISH CAPITAL LETTERS ONLY. 1 NAME OF SECRETARIAT. : 2 NAME OF DEPARTMENT. : 3 NAME OF INSTITUTE / OFFICE. : 4 OFFICE ADDRESS. : PHONE NUMBER. 5 NAME AND DESIGNATION OF HEAD OF. INSTITUTE/OFFICE. CONTACT NUM

CCC / CCC+ EXAMINATION REGISTRATION FORM
NAME OF EMPLOYEE. : SEAT NUMBER : : TRIAL NUMBER. : 1 (One) / 2 (Two) / 3 (Three). SIGNATURE OF HEAD OF EXAM CENTRE. SIGNATURE OF HEAD ...

CCC Materials.pdf
Sign in. Loading… Page 1. Whoops! There was a problem loading more pages. Retrying... CCC Materials.pdf. CCC Materials.pdf. Open. Extract. Open with.

ccc-plus.pdf
2o±© »– ÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚÚ Ô ... ̧ø ̈ · ø2 —°aÆø ̈·21 Õß ̈a3 ·. ‹·ooaÆa2 ̈ ̈ß°a ±o ø2 —°aÆø ̈·21 Õß ̈a3 ·. ÿ±© ̈± Õ ̈øÆ ̈ ›±3° ́ ̈aÆ ·. ÃaÆ3 ±o ̈a2 ́ao o±Æ ...·2o±© »–. ÿ±© ̈± ø1⁄21⁄2a ›

CCC'17.pdf
Cambridge College. Cameo Modeling and Talent ... St. John's University. Stonehill College. Stony Brook University ... CCC'17.pdf. CCC'17.pdf. Open. Extract.

Whipple - CCC Support Letter.pdf
Download. Connect more apps... Try one of the apps below to open or edit this item. Whipple - CCC Support Letter.pdf. Whipple - CCC Support Letter.pdf. Open.

64.pdf
Los computadores, el Internet y la. multimedia permiten acceder a cantidades abrumadoras de información,. procesar y enviar textos, datos, imágenes, sonido, ...

64.pdf
BBFuels of PR, with Biotechnology Development and Training Complex (BDTC), ... modified auto-hydrolysis pretreatment followed with two round steam ...

CCC CAD Operator Job Description.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. CCC CAD ...

CCC Change in Name Result.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. CCC Change in ...

Reclamation Project USACE and CCC jurisdictional delineaiton.pdf
North was pro- mised 25 new army hats and a year's supply of legal advice. from the law firm, .... U.S. Attorney Rudolf Giulliano has ordered a six- month postponement of the Center's construction in order. to facilitate a trace on the .... Retrying.

64-SBIS_Ju'alah.pdf
amanat kepadamu dan jangan kamu mengkhianati orang. Page 3 of 6. 64-SBIS_Ju'alah.pdf. 64-SBIS_Ju'alah.pdf. Open. Extract. Open with. Sign In. Main menu.

Dossier Officiel CCC 2K17.pdf
M. José Luis LOPEZ CERRON (ESP) représentant l'UEC. • M. Toni KIRSH (GER) représentant l'UEC. • M. Tom VAN DAMME (BEL) ... Public Relations Officer Michel Marie Sainte Martinique. Trustee Perry Gibson Belize. Trustee Henney Bonafacio Curacao.

CCC v2.0 040915.pdf
CLOVER VETCH SUNNHEMP CLUSTER. BEAN QUINOA PROSO. MILLET. CEREAL. RYE RADISH FLAX LENTIL CRIMSON. CLOVER. BIRDSFOOT.

Boletín_N°64.pdf
Una Asamblea Nacional Constituyente, originaria, plenipo- tenciaria, constitucional, para enfrentar y encarar esta situa- ción, y encausar una solución verdadera, democrática, libre y. definitiva”. NICOLÁS MADURO MOROS. Presidente de la República Bol

nintendo-64.pdf
Sign in. Page. 1. /. 24. Loading… Page 1 of 24. Page 1 of 24. Page 2 of 24. Page 2 of 24. Page 3 of 24. Page 3 of 24. nintendo-64.pdf. nintendo-64.pdf. Open. Extract. Open with. Sign In. Main menu. Displaying nintendo-64.pdf. Page 1 of 24.Missing:

Windows 8 64
Adobe pro pdf.Theflash 1x3. Mannequinmagic baby got boobs.Megamind 1080p 5.1.Decisive byChip and DanHeath.Windows 8 64.The Assaultmovie.One man ...

project 64 rom.pdf
There was a problem loading more pages. project 64 rom.pdf. project 64 rom.pdf. Open. Extract. Open with. Sign In. Main menu. Displaying project 64 rom.pdf.

GTA RN Integrated Acute to Inpt Rehab CCC Referral Form.pdf ...
Page 3 of 11. GTA RN Integrated Acute to Inpt Rehab CCC Referral Form.pdf. GTA RN Integrated Acute to Inpt Rehab CCC Referral Form.pdf. Open. Extract.

+47% +25% 64% Services
Post-implementation, the Bacaterus team was delighted that AMP pages load 4 seconds faster than its standard mobile pages. “AMP improves mobile user ...

64-17.pdf
(Cantonese,實為繁體中文)、中文(Chinese Mandarin,實為簡體中文)、法語、印度 .... 感性地認為,隨著獨立,蘇格蘭可以建立一個社會民族:一個. Page 3 of 5. 64-17.pdf.

+47% +25% 64% services
Bacaterus.com, a lifestyle website for travel, beauty, film and food segment, began publishing Accelerated Mobile Pages (AMP) six months ago. Bacaterus ...

Descargar fate extra ccc psp iso
descargar mp3 hecho con tus sueños.6783661349837110.descargar la ... libros delasaga decrepusculo para descargar gratis pdf.descargar imagenes d carros ...