Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cancer Therapy: Clinical

UGT1A7 and UGT1A9 Polymorphisms Predict Response and Toxicity in Colorectal Cancer Patients Treated with Capecitabine/Irinotecan

Leslie E. Carlini, Neal J. Meropol, John Bever, Michael L. Andria, Todd Hill, Philip Gold, Andre Rogatko, Hao Wang and Rebecca L. Blanchard
Leslie E. Carlini
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Neal J. Meropol
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
John Bever
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael L. Andria
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Todd Hill
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Philip Gold
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andre Rogatko
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hao Wang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rebecca L. Blanchard
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published February 2005
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: Capecitabine and irinotecan are commonly used in the treatment of metastatic colorectal cancer (CRC). We hypothesized that germline polymorphisms within genes related to drug target (thymidylate synthase) or metabolizing enzymes (UDP-glucuronosyltransferase, UGT) would impact response and toxicity to the combination of capecitabine plus irinotecan (CPT-11).

Experimental Design: Sixty-seven patients with measurable CRC were treated with irinotecan i.v. (100 or 125 mg/m2) on days 1 and 8 and capecitabine orally (900 or 1,000 mg/m2, twice daily) on days 2 through 15 of each 3-week cycle. Genomic DNA was extracted from peripheral blood and genotyped using Pyrosequencing, GeneScan, and direct sequencing (Big Dye terminator) technologies.

Results: The overall objective response rate was 45% with 21 patients (31%) exhibiting grade 3 or 4 diarrhea and 3 patients (4.5%) demonstrating grade 3 or 4 neutropenia in the first two cycles. Low enzyme activity UGT1A7 genotypes, UGT1A7*2/*2 (six patients) and UGT1A7*3/*3 (seven patients), were significantly associated with antitumor response (p = 0.013) and lack of severe gastrointestinal toxicity (p = 0.003). In addition, the UGT1A9 −118 (dT)9/9 genotype was significantly associated with reduced toxicity (p = 0.002) and increased response (p = 0.047). There were no statistically significant associations between UGT1A1, UGT1A6, or thymidylate synthase genotypes and toxicity or tumor response.

Conclusions: These data strongly suggest that UGT1A7 and/or UGT1A9 genotypes may be predictors of response and toxicity in CRC patients treated with capecitabine plus irinotecan. Specifically, patients with genotypes conferring low UGT1A7 activity and/or the UGT1A9 (dT)9/9 genotype may be particularly likely to exhibit greater antitumor response with little toxicity.

INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer in both men and women and the third most prevalent cause of cancer-related death (1). A common approach to the systemic management of metastatic colorectal cancer is a combination of fluoropyrimidine (e.g., 5-fluorouracil or capecitabine) and irinotecan (reviewed in ref. 2). Fluoropyrimidines act primarily through the inhibition of thymidylate synthase (TS), resulting in impaired DNA synthesis and cell death. Irinotecan is a camptothecin compound that inhibits DNA topoisomerase I, resulting in an accumulation of DNA damage and cell death. Irinotecan is a prodrug that undergoes conversion by liver carboxylesterases to form the active compound SN-38, 7-ethyl-10-hydroxy-camptothecin (2, 3). SN-38 is largely metabolized to the inactive glucuronide, SN-38G, via the action of several UDP-glucuronosyltransferase (UGT) including hepatic UGT1A1, UGT1A6, and UGT1A9 and extrahepatic UGT1A7 (4–7).

The existence of significant patient variability in response to fluoropyrimidines and irinotecan, coupled with knowledge of common genetic polymorphisms within genes important to the pharmacology of these drugs, suggests that pharmacogenetic studies could identify individuals likely to benefit from treatment or develop severe toxicity. Genetic variation within the promoter and the 3′-untranslated region (UTR) of the human TS gene has been previously linked to the efficacy of 5-fluorouracil-related drugs (reviewed in ref. 8). The polymorphic UGT1A family members may govern variability in patient response to irinotecan (reviewed in ref. 9). A common TA repeat within the promoter of the human UGT1A1 gene has been associated with CPT-11-related toxicity, most predominantly neutropenia (10–12). Functionally significant genetic variation has also been described for human UGT1A6, UGT1A7, and UGT1A9 (6, 13–17). In this study, we correlate the efficacy and toxicity of combined capecitabine/CPT-11 therapy with genetic variation in genes important in the metabolism of CPT-11 (UGT1A1, UGT1A6, UGT1A7, and UGT1A9). Given the narrow therapeutic index, frequent resistance, and expanding options for treatment of CRC, the need for predictive markers of response and toxicity has assumed increased importance.

MATERIALS AND METHODS

Patient Eligibility. This pharmacogenetic analysis was a secondary objective of a multicenter phase II trial of capecitabine/CPT-11 combination therapy in patients with metastatic CRC. The primary clinical objective was determination of objective response. Eligible patients were at least 18 years old with histologically confirmed metastatic colorectal adenocarcinoma. Previous cytotoxic chemotherapy was not permitted except for neoadjuvant or adjuvant treatment completed 12 months before study enrollment. Patients were required to be ambulatory with a Karnofsky performance status of ≥70%. Required laboratory values for inclusion included neutrophil count ≥1.5 × 109, platelet count ≥100 × 109/L, serum creatinine ≤1.5 × upper limit normal, estimated creatinine clearance ≥50 ml/min, serum bilirubin ≤1.25 × upper limit normal, ALAT and ASAT ≤2.5 × upper limit normal (<5 with liver metastasis or <10 with bone metastasis), or alkaline phosphatase ≤2.5 × upper normal limit (<5 with liver metastasis or <10 with bone metastasis). Exclusion criteria included known Gilbert's disease, pregnancy, central nervous system metastasis, active cardiac disease, or myocardial infarction within the previous 12 months, active infections, physical disorders of the gastrointestinal tract, or problems with malabsorption. Written informed consent was required, and the study was approved by the institutional review boards at all of the participating sites in accord with an assurance filed with and approved by the U.S. Department of Health and Human Services.

Drug Administration. Capecitabine (Xeloda, Roche Laboratories, Inc., Nutley, NJ) was given orally at a dosage of 1,000 mg/m2 (cohort 1, 15 patients) or 900 mg/m2 (cohort 2, 52 patients) twice daily on days 2 through 15 of 3-week cycles. Irinotecan (Camptosar, Pfizer, Inc., New York, NY) was given at a dosage of 125 mg/m2 (cohort 1, 15 patients) or 100 mg/m2 (cohort 2, 52 patients) as a 90-minute i.v. infusion on days 1 and 8 of each cycle. The doses of capecitabine and irinotecan were decreased as described for cohort 2 after the first 15 patients (cohort 1) because of unacceptable toxicity. The maximum number of cycles given was 12.

Evaluation of Response and Toxicities. Baseline tumor measurements were obtained within 21 days before initial treatment. Response assessments were obtained every two cycles (6 weeks) with confirmation of response after 4 weeks according to RECIST criteria (18). Toxicity was assessed weekly during the first two cycles of treatment according to the National Cancer Institute Common Toxicity Criteria, version 2.0 (National Cancer Institute Common Toxicity Criteria, http://ctep.cancer.gov). All toxicities evaluated in this study occurred during the first two cycles of treatment.

Genomic DNA Preparation and Genotyping Assays. Blood samples were collected for isolation of genomic DNA during venipuncture for other diagnostic labs at least 1 week before starting treatment. Genomic DNA was prepared from peripheral WBCs and suspended in 10 mmol/L Tris with 50 mmol/L EDTA (TE; Covance, Princeton, NJ). DNA was isolated from 66 of the 67 subjects and was used to identify UGT1A1, UGT1A6, UGT1A7, UGT1A9, and thymidylate synthase polymorphisms. For each batch of assays, appropriate positive and negative controls of established genotype were assayed.

UGT1A1. The UGT1A1 variable length (TA)n repeat polymorphism (n = 5-8) was evaluated using Genescan technology (Applied Biosystems, Foster City, CA; refs. 19, 20). A 254-bp region of the UGT1A1 gene was amplified using a hexachloro-6-carboxyfluorescein fluorescently tagged antisense primer (5′-ATCAACAGTATCTTCCCAG-3′) and a nonfluorescent sense primer (5′-TATCTCTGAAAGTGAACTC-3′). The PCR reaction mixture included 20 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 1.5 mmol/L MgCl2, 0.2 mmol/L each deoxynucleotide triphosphate (dNTP), 0.2 μmol/L each primer, 1.25 units Platinum Taq polymerase (Invitrogen, Carlsbad, CA), and 20 ng genomic DNA in a 50 μL reaction volume. The PCR profile included 1-minute denaturation at 94°C, 34 cycles of 30 seconds at 94°C, 30 seconds at 52°C, and 30 seconds at 72°C, with a final 5-minute extension at 72°C in a Perkin-Elmer 9700 Thermal Cycler (Perkin-Elmer, Boston, MA). Fluorescently labeled products were electrophoretically separated on a 4% polyacrylamide gel on an ABI 377 DNA Sequencer. Fluorescent bands were analyzed using GENESCAN 2.1 software to determine fragment length (Applied Biosystems). Genotypes were assigned as UGT1A1*1 (reference), UGT1A1*36, UGT1A7*28, and UGT1A7*37 for TA repeat numbers of 6, 5, 7, and 8, respectively.

UGT1A6.UGT1A6 genotype was determined with a PCR-based assay that uses Pyrosequencing technology (Pyrosequencing, Westborough, MA). UGT1A6 alleles are defined by permutations of three single-nucleotide polymorphisms (SNPs) that alter the encoded amino acid sequence at nucleotide positions 19T > G, 541A > G, and 552A > C (for amino acids S7A, T181A, and R184S, respectively; ref. 15). For detection of the 19T > G SNP, a 238-bp fragment was amplified using primer F-53 (5′-GATTTGGAGAGTGAAAACTCTTT-3′) and R184 (5′-biotin-CAGGCACCACCACTA-CAATCTC-3′). For the remaining two SNPs, a 215-bp fragment was amplified using primer F414 (5′-biotin-CTTTAAGGAGAGCAAGTTTGATG-3′) and R628 (5′-CCACTCGTTG-GGAAAAAGTC-3′). Approximately 25 to 50 ng of genomic DNA was amplified in a reaction mixture containing 20 mmol/L Tris-HCl (pH 8.4), 50 mmol/L KCl, 1.5 mmol/L MgCl2, 0.2 mmol/L dNTPs, 0.2 μmol/L primers, and 2.5 units of Platinum Taq polymerase in a 50 μL volume. Reactions were performed in a Perkin-Elmer 9700 Thermal Cycler with 45-second denaturation at 94°C, followed by 35 cycles of 94°C for 30 seconds, 56°C for 30 seconds, and 72°C for 50 seconds. A final 3-minute extension at 72°C completed the amplification.

Amplicons were prepared for automatic Pyrosequencing SNP analysis on the PSQ 96 system using reagents from the PSQ96 SNP Reagent Kit (Pyrosequencing). A 25-μL volume of double-stranded biotinylated amplicon was incubated with 100 μg of streptavidin-coated M280 DynaBeads (Dynal, Brown Deer, WI) in binding buffer [5 mmol/L Tris (pH 7.6), 1 mol/L NaCl, 0.5 mmol/L EDTA, 0.05% Tween 20] and incubated at 65°C for 15 minute followed by denaturation in 0.5 mol/L NaOH. Single-stranded biotinylated DNA was transferred to annealing buffer [20 mmol/L Tris Acetate (pH 7.6) and 5 mmol/L Mg(OAc)2] for 1 minute then transferred to sequencing primer solution containing annealing buffer and 10 pmol of the appropriate sequencing primer. The 19T > G primer was 5′-GATGGCCTGCCTCCTT-3′, whereas the 541A > G and 552A > C primer was 5′-AGGACACAGGGTCTG-3′. The nucleotide dispensation sequence for the 19T > G SNP was TCGCTGACA with the underlined nucleotide representing the negative control and those in bold representing the polymorphic site. The nucleotide dispensation sequence for the remaining SNPs was CGCTGACTGCTGATGTCAT. Genotypes were assigned as UGT1A6*1 (S7, T181, and R184 reference), UGT1A6*2 (A7, A181, and S184), UGT1A6*3 (A7) and UGT1A6*4 (A7 and S184).

UGT1A7.UGT1A7 genotype was determined by direct sequencing of a PCR product that spans all of the polymorphic sites. The UGT1A7 alleles are defined by permutations of six SNPs (342G > A, 387T > G, 391C > A and 392G > A, 417G > C, and 622T > C) that alter the encoded amino acid sequences (G115S, N129K, R131K, E139D, and W208R, respectively; ref. 14). A 415-bp fragment from UGT1A7 was amplified using primer F277 (5′-TTTGCCGATGCTCGCTGGACG-3′) and R692 (5′-GCTAT-TTCTAAGACATTTTTGAAAAAATAGGG-3′). The PCR reaction mixture included JumpStart REDTaq ReadyMix PCR reaction mix (Sigma, St. Louis, MO) containing 10 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 2 mmol/L MgCl2, 0.2 mmol/L each dNTP, and 0.03 units/Taq polymerase with the addition of 0.2 μmol/L each primer and 40 ng genomic DNA in a 50-μL reaction volume. The PCR conditions included 1-minute denaturation at 94°C, 35 cycles of 30 seconds at 94°C, 30 seconds at 56°C, and 30 seconds at 72°C, with a final 5-minute extension at 72°C in a Perkin-Elmer 9700 Thermal Cycler. Amplified products were sequenced using an ABI Prism 377 DNA Sequencer (Applied Biosystems) and primers F292 (5′-TGGACGGCACCATTG-3′) and R675 (5′-TTTGAAAAAATAGGGGCAA-3′). Genotypes were assigned as UGT1A7*1 (G115, N129, R131, and W208 reference), UGT1A7*2 (K129 and K131), UGT1A7*3 (K129, K131, and R208), UGT1A7*4 (R208).

UGT1A9. Five UGT1A9 polymorphisms were evaluated by direct DNA sequencing of a PCR amplicon spanning all of the polymorphic sites. Polymorphisms of interest included a −118 (dT)n repeat (n = 9 or 10), C3Y (8G > A), M33T (98T > C), Y242X (726T > G), and D256N (766G > A; refs.14, 21, 22). We also detected the following promoter: and intronic SNPs: −87G > A, and intron 1 I143C > T, I152G > A, I201A > C, I219T > A, and I313A > C. The −118 (dT)9/10 polymorphism has been previously localized to nucleotides −98 through −109 (16, 17). However, we believe it is most accurate to assign the localization of this poly d(T) tract as spanning nucleotides −118 through −129 (based on assigning the “A” in the “ATG” translation start codon as +1) and we refer to the polymorphisms as −118 (dT)9 or −118 (dT)10.

A 1.4-kb fragment from UGT1A9 was amplified using primer F-273 (5′-AAACTTAACATTGCAGCACAGGGC-3′) and RI337 (intron 1, 5′-CTAAGACCATTT-CCTCTGGGGC-3′). The PCR reaction mixture included 10 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 2 mmol/L MgCl2, 0.2 mmol/L each dNTP, 0.2 μmol/L each primer, 5% DMSO, 1.5 units Platinum Taq polymerase (Invitrogen), and 40 ng genomic DNA in a 50-μL reaction volume. The PCR conditions included 1-minute denaturation at 94°C, 35 cycles of 94°C for 30 seconds, 55°C for 30 seconds, and 72°C for 1.5 minutes, with a final 5-minute extension at 72°C in a Perkin-Elmer 9700 Thermal Cycler. Polymorphisms were detected by direct sequencing (ABI 377 DNA Sequencer, Applied Biosystems) of amplified sequences using primers F-273, R361 (5′-AAAAATAAGTCAAAAATGTC-ATTGT-3′), Fsp (5′-GGAGGAACATTTATTATGCCACCG-3′), and RI337 primers.

Observed haplotypes were designated UGT1A9HI (10GCGAC), UGT1A9HII (9GCGTA), UGT1A9HIII (9GCATA), UGT1A9HIII (9ACGTA), and UGT1A9HIV (9GTGTA) for the following polymorphisms detected in 66 subjects: −118 (dT)n repeat, −87G > A, and the intron 1 SNPs I143C > T, I152G > A, I219A > T, and I313C > A.

Thymidylate Synthase Promoter.TS genotypes were determined using a combination of gel electrophoresis–based and direct DNA sequencing techniques. Common functional polymorphisms in the TS gene include a 28-bp variable tandem repeat (n = 2, 3, 4, 5, or 9) within an enhancer of the promoter and a SNP within the 5′-UTR of the three repeat (3R) allele (−58G > C; refs.23, 24). The variable tandem repeat was detected by PCR amplification with F-220 (5′-GTGGCTCCTGCGTTTCCCCC-3′) and R + 1 primers (5′-TCCGAGCCGGCCACAGGCAT-3′) that flank the repeat region. The PCR reaction mixture included 10 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 2 mmol/L MgCl2, 5% DMSO, 0.2 mmol/L each dNTP, 0.2 μmol/L each primer, 1.5 units Platinum Taq polymerase (Invitrogen), and 40 ng genomic DNA in a 50-μL reaction volume (25). The PCR conditions included 1-minute denaturation at 94°C, 34 cycles of 94°C for 40 seconds, 70°C for 40 seconds and 72°C for 40 seconds, with a final 5-minute extension at 72°C in a Perkin-Elmer 9700 Thermal Cycler. After separating the gene fragments via gel electrophoresis, fragment lengths were determined relative to known molecular markers. Direct sequencing of the amplified products allowed identification of the 5′-UTR SNP. Genotypes were assigned according to repeat lengths and the existence of the SNP: one repeat (1R), two repeats (2R reference), three repeats (3RC or 3RG), four repeats (4R), five repeats (5R), or nine repeats (9R).

Thymidylate Synthase 3′-UTR. The presence or absence of the 6-bp sequence TTAAAG at position 1494 of the TS mRNA was detected by PCR amplification with F3′utrTS (5′-CAAATCTGAGGGAGCTGAGT-3′) and R3′utrTS (5′-CAGATAAGTGGCAGTACAGA-3′) to produce either 148- or 142-bp fragments (26). The PCR reaction mixture included 10 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 2.5 mmol/L MgCl2, 0.2 mmol/L each dNTP, 0.2 μmol/L each primer, 1.5 units Platinum Taq polymerase (Invitrogen), and 40 ng genomic DNA in a 50 μL reaction volume (26, 27). The PCR profile included 1-minute denaturation at 94°C, 35 cycles at 94°C for 30 seconds, 58°C for 45 seconds, and 72°C for 45 seconds, and a final 5-minute extension at 72°C in a Perkin-Elmer 9700 Thermal Cycler. Amplified products were separated via gel electrophoresis on 3.5% Metaphor agarose and fragment lengths were determined relative to known molecular markers. Genotypes were assigned according to the presence or absence of the 6-bp sequence as determined by fragment lengths: 6 bp (148 bp, reference) or 0 bp (142 bp, deletion).

Statistical Methods. The sample size for this trial (ultimately cohort 2) was governed by the primary objective of response rate (complete or partial) and powered to reject the null hypothesis of 30% with a one-sided significance level of 5% (α = 0.05). This provided at least 80% power to reject the null hypothesis at a response rate of 50% with a sample of 45 patients. The overall significance level was 0.028 and the overall power was 0.814. A total of 50 patients allowed for an approximate dropout or nonevaluable rate of 10%.

Two-sided tests of statistical significance were used to determine statistically significant P values (P < 0.05) using SAS software. A permutation procedure based on an exact test was applied to check Hardy-Weinberg equilibrium of each marker, and P < 0.05 indicated a lack of agreement with Hardy-Weinberg equilibrium (28). Haplotype frequencies and their corresponding variance were estimated by expectation-maximization algorithm and jackknife method, respectively. Toxicity was dichotomized with toxicity grades 3 or 4 as one category and grade ≤2 as another. Statistical associations between genotypes and drug response or toxicity were examined using a two-sided Fisher's exact test for each individual marker. The exact test of Cochran-Armitage trend test was done to detect the direction of the relationship between toxicity or response rate and genotype activity bins. Associations between genotype groups and clinical measurements were examined using the nonparametric Kruskal-Wallis test and the Jonckheere-Terpstra test.

For the purposes of genotype/response analyses, we binned genotypes into functional categories based on published descriptions of the alleles or genotypes of interest. UGT1A1 genotypes were grouped into high (UGT1A1 5/6 and UGT1A1 6/6), moderate (UGT1A1 6/7), and low (UGT1A1 7/7 and UGT1A1 7/8) activities (19, 20). UGT1A6 was defined by high (UGT1A6*2/*2), moderate (UGT1A6*1/*1), low (UGT1A6*1/*2), and undefined/unknown activities (UGT1A6*1/*3 and UGT1A6*1/*4; 15). UGT1A7 genotypes were categorized into high (UGT1A7*1/*1), moderate (UGT1A7*1/*2 and UGT1A7*1/*3), low (UGT1A7*2/*2 and UGT1A7*3/*3), and undefined/unknown (UGT1A7*2/*3) activity (14, 29). For UGT1A9, genotypes and polymorphisms were analyzed individually for associations with toxicity and response due to the lack of functional data regarding these variations. However, a recent publication reported 2.6-fold higher transcriptional activity associated with the −118 (dT)10 allele compared with the −118 (dT)9 allele based on in vitro transcriptional reporter assays (16). The TS promoter activities were defined as high (3G/3G), moderate (2/3G and 3G/3C), and low activity (2/2, 2/3C, and 3C/3C). A separate analysis of high (3G/3G, 2/3G, and 3G/3C) and low activity (2/2, 2/3C, and 3C/3C) was also completed due to discrepancies in the literature (23, 24), . Finally, the 3′-UTR TS binning included high (6/6), moderate (0/6), and low (0/0) activities (26, 30).

RESULTS

A total of 67 patients with metastatic colorectal cancer were enrolled in this prospective phase II trial, and germline DNA was available for 66 of these patients. Of those 66 subjects, response and toxicity data were collected from 56 and 66 subjects, respectively. Fifty-five subjects (83%) were Caucasian, nine (14%) were African American, one was Samoan, and one was Hispanic (Table 1). The overall objective response rate (partial or complete response) was 45% (30 of 67 patients). The incidence of severe or life-threatening diarrhea or neutropenia (grade 3 or 4, National Cancer Institute Common Toxicity Criteria version 2.0) was 33% (22 of 66 subjects). Nineteen of 66 subjects (29%) developed grade 3 or 4 diarrhea alone, one subject (1%) exhibited grade 3 or 4 neutropenia alone, and two subjects (3%) experienced both diarrhea and neutropenia within the first two cycles of treatment.

View this table:
  • View inline
  • View popup
Table 1

Demographics, overall response and toxicity of study subjects

Allele and Genotype Frequencies and Hardy-Weinberg Equilibrium. We evaluated the predictive value of genotypes within the UDP-glucuronosyltransferases (UGT1A1, UGT1A6, UGT1A7, and UGT1A9) as well thymidylate synthase. Table 2 lists the observed allele frequencies, whereas Table 3 lists genotype frequencies. There were no significant differences in allele frequencies between cohorts 1 and 2. Therefore, these cohorts were combined for genotype/phenotype analyses. With the exception of UGT1A9 and TS promoter polymorphisms, all genotypes were observed in Hardy-Weinberg equilibrium (Table 2). The fairly rare UGT1A1 TA5 and TA8 alleles were observed only in African American patients at frequencies consistent with published values (19, 31). The genotype frequencies observed for UGT1A1 6/6, UGT1A1 6/7, and UGT1A1 7/7 (Table 3) agreed with that reported in a random population (13, 20) despite the exclusion of patients with elevated bilirubin, a phenotype associated with the UGT1A1 TA7 allele (10, 32). Hence, the distribution of UGT1A1 alleles followed Hardy-Weinberg equilibrium (Table 2). Allele and genotype frequencies for UGT1A6 (Tables 2 and 3) agreed with our previous study (15). UGT1A7 allele and genotype frequencies were as expected from the literature (13, 14), although we did not observe UGT1A7*4 (Table 3).

View this table:
  • View inline
  • View popup
Table 2

Observed allele frequencies

View this table:
  • View inline
  • View popup
Table 3

Observed genotype frequencies

We did not detect the C3Y, M33T, Y242X, or D256N polymorphisms described for UGT1A9 (14, 21, 22). However, we did identify the −118 (dT)n polymorphism as well as four SNPs within intron sequences of the UGT1A9 gene (Tables 2 and 3) that defined a total of five haplotypes (Table 4). The −118 (dT)n repeat and intron 1 SNPs I219 and I313 were completely linked in this study population. UGT1A9 genotypes were in agreement with Hardy-Weinberg equilibrium (Table 4), although the haplotype frequencies were not (Table 2). Although this study was completed in a diseased population, haplotype frequencies closely resembled those observed within a healthy population (n = 53) of mixed ethnicity, mainly comprised of Caucasians (59.4% for n = 9 and 40.6% for n = 10 repeats).4

View this table:
  • View inline
  • View popup
Table 4

UGT1A9 haplotype analysis

We identified two novel alleles within the TS promoter, defined by one repeat (1R) from a Caucasian subject and a 2R allele with a novel SNP (−30C > T) in the second repeat from a patient of undefined ethnicity (Tables 2 and 3). The rare 4R and 9R alleles were detected in two subjects of non-Caucasian ethnicity (31). Allele (Table 2) and genotype (Table 3) frequencies for the TS 3′-UTR polymorphism were within the ranges reported by previous studies (26, 27). However, the linkage disequilibrium reported between the 3R TS promoter and 0 bp 3′-UTR polymorphisms (26, 30) was not observed in this study.

Genotype Correlations with Tumor Response. We observed an association between the low activity UGT1A7*2/*2 and UGT1A7*3/*3 genotypes and the UGT1A9 (dT)9 polymorphism and increased tumor response (complete or partial, Tables 5 and 6). Among individuals with low activity UGT1A7*2/*2 and UGT1A7*3/*3 genotypes, the response rate was 85% (P = 0.013, 11 of 13 patients responded compared with 44% or 19 of 43 patients with other genotypes, Table 5). Furthermore, a statistically significant trend was observed for increased response across UGT1A7 genotypes (p for trend = 0.017; Table 5). A statistically significant trend was also observed between the UGT1A9 −118 (dT)n repeat and tumor response (p for trend = 0.033, Table 6). The −118 (dT)9/9 genotype was significantly associated with efficacious tumor response when compared with all other genotypes (p = 0.047; 74% or 14 of 19 patients with −118 (dT)9/9 versus 43% or 16 of 37 patients with other genotypes).

View this table:
  • View inline
  • View popup
Table 5

UGT1A7 genotypes with reduced enzyme activity (UGT1A7*2/*2 and UGT1A7*3/*3) are associated with improved efficacy and reduced toxicity

View this table:
  • View inline
  • View popup
Table 6

UGT1A9 −118 (dT)9/9 genotype is associated with improved efficacy and reduced toxicity

We found no significant association between UGT1A1 genotype and tumor response. Although not statistically significant, subjects with low UGT1A1 enzyme activity responded better to treatment (83% for UGT1A1 7/7 or UGT1A1 7/8 genotype relative to 46% for high activity UGT1A1 5/6 or UGT1A1 6/6, Table 7). No significant associations or trends were observed between UGT1A6 genotype (Table 8), TS polymorphisms, or overall UGT1A9 haplotypes and tumor response.

View this table:
  • View inline
  • View popup
Table 7

UGT1A1 genotypes did not significantly associate with efficacy and toxicity

View this table:
  • View inline
  • View popup
Table 8

UGT1A6 genotypes did not significantly associate with efficacy and toxicity

Genotype Correlation with Toxicity. Twenty-two subjects developed grade 3 or 4 diarrhea or neutropenia (33%, Table 1). The predominant toxicity observed was diarrhea (21 subjects) as opposed to neutropenia (three subjects). We observed a striking correlation between the low activity UGT1A7 genotypes (UGT1A7*2/*2 and UGT1A7*3/*3) and lack of drug toxicity (p = 0.003, Table 5). The UGT1A9 −118 (dT)9/9 genotype was also associated with a lower incidence of toxicity (p = 0.002, Table 6). We found no statistically significant association between UGT1A1 genotype and toxic events. However, none of the six subjects with low activity UGT1A1 genotypes (UGT1A1 7/7 or UGT1A1 7/8) experienced toxicity (Table 7). There was no significant association between UGT1A6 genotype and incidence of toxicity, but none of the five patients with high activity UGT1A6*2/*2 genotype experienced toxicity (Table 8). There was no association between TS polymorphisms and incidence of toxicity.

Because the UGT1A1 promoter polymorphism has been associated with neutropenia, it is of interest to report the genotypes specifically of the individuals that experienced neutropenia. Three subjects (4.5%) developed grade 3 neutropenia. Two subjects possessed the UGT1A1 6/7 genotype coupled with either the UGT1A7*1/*2 or UGT1A7*1/*3 genotype, whereas the other subject possessed UGT1A1 6/8 and UGT1A7*1/*2. All three were heterozygous for the UGT1A9 −118 (dT)n repeat. Two of the subjects with neutropenia also exhibited grade 3 diarrhea; both patients possessed the UGT1A7*1/*2 genotype.

Linkage Disequilibrium/Haplotyping Analysis. We observed UGT1A haplotype frequencies similar to those obtained by Kohle et al. (13) for UGT1A1, UGT1A6, and UGT1A7 (Table 9). Additionally, we expanded our haplotype analysis to include the UGT1A9 −118 (dT)n repeat. We considered only this polymorphism for UGT1A9 because the observed UGT1A9 haplotypes were not in Hardy-Weinberg equilibrium (Tables 2 and 4). We discovered that the low activity UGT1A7*2 and UGT1A7*3 alleles were completely associated with the UGT1A9 −118 (dT)9 allele, whereas the high activity UGT1A7*1 allele was linked with the UGT1A9 −118 (dT)10 allele (Table 9, likelihood ratio test of association p < 0.0001). Such clearly defined associations across haplotypes were not observed for UGT1A1 or UGT1A6. Finally, a statistically significant association was identified between haplotype I and patient response and toxicity (p = 0.04 and p = 0.035, respectively, Table 9).

View this table:
  • View inline
  • View popup
Table 9

UGT1A haplotype analysis

DISCUSSION

This study was significant for two major observations. First, genetic variation in the human UGT1A7 and UGT1A9 genes predicted for tumor response as well as development of diarrhea during combination irinotecan/capecitabine therapy and second, low activity UGT1A1 alleles did not predict for the development of diarrhea. We observed associations between UGT1A7 and UGT1A9 genotypes with both tumor response and toxic events (Tables 5 and 6). We also found that the UGT1A9 −118 (dT)9/9 genotype was predictive for efficacious tumor response with lower incidence of toxicity (Table 6), whereas the UGT1A9 −118 (dT)10/10 genotype predicted for poor tumor response. The functional consequences of the UGT1A9 promoter polymorphism are not well established. However, the −118 (dT)10 allele has recently been associated with 2.6-fold greater transcriptional activity than the −118 (dT)9 allele (16). If these in vitro studies are predictive of in vivo function (which remains to be proven), then we would predict that alleles containing the UGT1A9 (dT)9 polymorphism might confer low activity relative to alleles defined by the UGT1A9 (dT)10 polymorphism.

Only one previous study has examined the association of UGT1A7 pharmacogenetics in patients receiving irinotecan and those investigators reported no significant association between UGT1A7 genotype and the occurrence of toxicity (33). However, it is important to note that the frequencies of the UGT1A7*2 and UGT1A7*3 alleles are significantly lower in the Japanese population, so that the power to detect associations in that study may have been compromised. To our knowledge, no previous studies have addressed the association between UGT1A9 polymorphisms and the incidence of irinotecan toxicity or tumor response.

The present study allowed us to evaluate UGT1A haplotypes (Table 9). This is an important component of this study because the human UGT1A genes comprise a unique nested gene structure on human chromosome 2 allowing for linkage disequilibrium among these genes (13). This raises the possibility that association between UGT1A alleles and specific phenotypes may be due to linkage disequilibrium. We observed complete linkage between the UGT1A7 low activity alleles (UGT1A7*2 and UGT1A7*3) and the UGT1A9 (dT)9 polymorphism (Table 9). Both enzymes have been reported to catalyze the glucuronidation of SN-38 with significant efficiency (6, 7, 34). UGT1A9 is expressed in the colon and liver and might be predicted to affect plasma levels of SN-38 (35, 36). UGT1A7 is not expressed in the liver, but is expressed in the proximal gastrointestinal tract and may influence the disposition of SN-38 within the gut (35, 36). Based on our current knowledge of tissue expression patterns, one might speculate that UGT1A9 is more likely to affect irinotecan disposition than is UGT1A7; however, the UGT1A7 alleles are clearly functionally significant and it is possible that UGT1A7 is expressed (either constitutively or inducibly) in as yet undetermined tissues that might influence SN-38 disposition. Therefore, it is premature at this time to predict which of the two genes is more likely the biological connection to the observed responses to irinotecan. Overall, we determined that low activity UGT genotypes were associated with better tumor response (Tables 5 and 6). This is consistent with the notion that low UGT activity might predict for higher concentrations of plasma SN-38 and increased tumor concentration of SN-38.

Previous studies have described an association between the UGT1A1 TA7 promoter polymorphism and the occurrence of irinotecan-induced toxicity (10–12). We were surprised that we did not observe the expected association between UGT1A1 low activity genotypes (UGT1A1 7/7 or UGT1A1 7/8) and increased incidence of toxicity (Table 7). Indeed, although not statistically significant, we observed the opposite trend. Of the six subjects with low activity genotypes, none experienced toxicity. The distribution of UGT1A1 genotypes in this study was reflective of a random population although patients were excluded with high serum bilirubin levels. A careful study of the primary literature regarding UGT1A1 pharmacogenetics suggests a clear link between low activity alleles and plasma ratio of SN-38/SN-38G as well as the development of neutropenia (10–12, 33), but the association between these alleles and the development of diarrhea has been much weaker. That is true, in part, because neutropenia was the predominant dose-limiting toxicity observed in these studies.

We suggest that different dosing regimens of irinotecan predispose patients to different toxicities. Our study differed from the previous studies in that subjects received combination irinotecan (100 or 125 mg/m2 on days 1 and 8) with oral capecitabine therapy. With this regimen, we observed diarrhea to be the predominant toxicity (22 subjects with grade 3 or 4 toxicity: 19 with diarrhea alone, two with diarrhea and neutropenia, and one with neutropenia alone). This provided us with a unique opportunity to study the association between UGT alleles and specifically irinotecan-induced diarrhea. We observed that low activity alleles protected subjects from developing diarrhea, rather than predisposing for toxicity as has been observed for irinotecan-associated neutropenia.

This apparent dichotomous set of observations is plausible when one considers the complex pharmacology of irinotecan. Glucuronidation represents an elimination pathway from the plasma. Hence, low capacity to glucuronidate results in increased plasma levels of SN-38 (11) and increased susceptibility to systemic toxicities such as neutropenia (10–12, 33). However, glucuronidation also represents a route of delivery of SN-38 to the gastrointestinal tract. In humans, renal excretion of irinotecan and its metabolites accounts for <20% of the dose with most of the remaining elimination, particularly for SN-38G, occurring via the biliary route (37–39). It is well documented that bacterial glucuronidases within the gut catalyze the deglucuronidation of many drugs, including SN-38G (reviewed in ref. 40). Therefore, glucuronidation of SN-38 directs its delivery to the gut where the toxic SN-38 molecule can be regenerated to promote lesions within the mucosa of the gastrointestinal tract leading to diarrhea. Thus, glucuronidation detoxifies SN-38 in the liver, contributing to lower systemic circulation of SN-38 and protects against systemic toxicity including neutropenia; but the glucuronidation pathway, in directing SN-38G to the gut, may predispose for irinotecan-induced diarrhea.

Clinical and animal studies seem to support this hypothesis. At least two clinical pharmacokinetic studies suggested that neutropenia, but not diarrhea, was significantly associated with the AUC for CPT-11 and/or SN-38 (11, 41). In rats, the activity of bacterial β-glucuronidase, the enzyme that generates SN-38 from SN-38G in the gut, correlated with irinotecan-induced cecal damage and was attenuated by administration of antibiotics (42) or of a specific β-glucuronidase inhibitor (43). Furthermore, human studies have shown that administration of antibiotics before irinotecan therapy significantly diminishes the incidence of severe diarrhea (44). Thus, glucuronidation may well promote one toxicity (diarrhea) yet protect for the other (neutropenia). Hence, genetic variation in specific UGT genes may predict for either high incidence of toxicity or low incidence depending on the dosing regimen.

These data have important public health implications. It has been suggested that clinical testing for the UGT1A1 promoter polymorphism should be implemented as a predictor of toxicity in patients receiving irinotecan (45, 46). It possible that for dosing regimens in which neutropenia is clearly the dose-limiting toxicity this practice might benefit patients. However, we observed that the dose limiting toxicity of combination capecitabine/irinotecan was diarrhea and that low capacity to glucuronidate may be protective for (rather than a risk factor for) occurrence of diarrhea and also predicts for efficacious tumor response. Although our proposed model regarding the role of glucuronidation in predicting different toxicities remains to be proven, we suggest that caution be applied in implementing clinical and regulatory precedence regarding low activity UGT alleles as general predictors for toxicity.

Acknowledgments

We thank Yin-Miao Chen of Roche Laboratories for helpful discussions regarding statistical analyses; the contributions of the Fox Chase Cancer Center Core Facilities, including the Biosample Repository, Tumor Bank, oligonucleotide synthesis, DNA sequencing, cell culture, and genotyping; and the participation of the following clinical personnel at multiple centers: Fakniuddin Ahmed HemOnCare, PC Research, New York, NY; Miklos Auber Robert Byrd Health Science Center, Morgantown, WV; Hoo Chun New York Medical College, Valhalla, NY; Philip Desimone University of Kentucky, Lexington, KY; Mandeep Dhami Eastern CT Hematology and Oncology, Norwich, CT; George Giels Charleston Hematology and Oncology, Charleston, SC; Thomas Godfrey Loma Linda University Cancer Institute, Loma Linda, CA; Stephen Kahanic Siouxland Hematology/Oncology, Sioux City, IA; Kirk Lund Rockwood Clinic-Oncology Department, Spokane, WA; John Marshall Georgetown University Medical Center, Washington, DC; Edith Mitchell Thomas Jefferson University, Philadelphia, PA; Muhammad Saif Wallace Tumor Institute, Birmingham, AL; and Robert Shepard University of Virginia Health System, Charlottesville, VA.

Footnotes

  • ↵4 Unpublished data.

  • Grant support: Roche Laboratories, Inc.

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • Note: Presented in part at the 40th Annual Meeting of the American Society of Clinical Oncology, June 5-8, 2004, New Orleans, LA.

    • Accepted November 9, 2004.
    • Received September 2, 2004.
    • Revision received November 3, 2004.

References

  1. ↵
    Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer J Clin 2004;54:8–29.
    OpenUrlCrossRefPubMed
  2. ↵
    Garcia-Carbonero R, Supko JG. Current perspectives on the clinical experience, pharmacology, and continued development of the camptothecins. Clin Cancer Res 2002;8:641–61.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    Ulukan H, Swaan PW. Camptothecins: a review of their chemotherapeutic potential. Drugs 2002;62:2039–57.
    OpenUrlCrossRefPubMed
  4. ↵
    Iyer L, King CD, Whitington PF, et al. Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J Clin Invest 1998;101:847–54.
    OpenUrlCrossRefPubMed
  5. ↵
    Ciotti M, Basu N, Brangi M, Owens IS. Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Biochem Biophys Res Commun 1999;260:199–202.
    OpenUrlCrossRefPubMed
  6. ↵
    Gagne JF, Montminy V, Belanger P, Journault K, Gaucher G, Guillemette C. Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Mol Pharmacol 2002;62:608–17.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Hanioka N, Ozawa S, Jinno H, Ando M, Saito Y, Sawada J. Human liver UDP-glucuronosyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin. Xenobiotica 2001;31:687–99.
    OpenUrlCrossRefPubMed
  8. ↵
    Marsh S, McLeod HL. Thymidylate synthase pharmacogenetics in colorectal cancer. Clin Colorectal Cancer 2001;1:175–8; discussion 179–81.
    OpenUrlPubMed
  9. ↵
    Guillemette C. Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J 2003;3:136–58.
    OpenUrlCrossRefPubMed
  10. ↵
    Ando Y, Saka H, Ando M, et al. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res 2000;60:6921–6.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Iyer L, Das S, Janisch L, et al. UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2002;2:43–7.
    OpenUrlCrossRefPubMed
  12. ↵
    Innocenti F, Undevia SD, Iyer L, et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 2004;22:1382–8.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Kohle C, Mohrle B, Munzel PA, et al. Frequent co-occurrence of the TATA box mutation associated with Gilbert's syndrome (UGT1A1*28) with other polymorphisms of the UDP-glucuronosyltransferase-1 locus (UGT1A6*2 and UGT1A7*3) in Caucasians and Egyptians. Biochem Pharmacol 2003;65:1521–7.
    OpenUrlCrossRefPubMed
  14. ↵
    Villeneuve L, Girard H, Fortier LC, Gagne JF, Guillemette C. Novel functional polymorphisms in the UGT1A7 and UGT1A9 glucuronidating enzymes in Caucasian and African-American subjects and their impact on the metabolism of 7-ethyl-10-hydroxycamptothecin and flavopiridol anticancer drugs. J Pharmacol Exp Ther 2003;307:117–28.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Nagar S, Zalatoris J, Blanchard R. Human UGT1A6 Pharmacogenetics: identification of a novel SNP, characterization of allele frequencies and functional analysis of recombinant allozymes in human liver tissue and in cultured cells. Pharmacogenetics 2004;14:487–99.
    OpenUrlCrossRefPubMed
  16. ↵
    Yamanaka H, Nakajima M, Katoh M, et al. A novel polymorphism in the promoter region of human UGT1A9 gene (UGT1A9*22) and its effects on the transcriptional activity. Pharmacogenetics 2004;14:329–32.
    OpenUrlCrossRefPubMed
  17. ↵
    Girard H, Court MH, Bernard O, et al. Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics 2004;14:501–15.
    OpenUrlCrossRefPubMed
  18. ↵
    Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000;92:205–16.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci U S A 1998;95:8170–4.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Innocenti F, Grimsley C, Das S, et al. Haplotype structure of the UDP-glucuronosyltransferase 1A1 promoter in different ethnic groups. Pharmacogenetics 2002;12:725–33.
    OpenUrlCrossRefPubMed
  21. ↵
    Jinno H, Saeki M, Saito Y, et al. Functional characterization of human UDP-glucuronosyltransferase 1A9 variant, D256N, found in Japanese cancer patients. J Pharmacol Exp Ther 2003;306:688–93.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Mackenzie P. Glucuronosyltransferases Home Page: Human UGT Allele Tables. 2004.
  23. ↵
    Kawakami K, Watanabe G. Identification and functional analysis of single nucleotide polymorphism in the tandem repeat sequence of thymidylate synthase gene. Cancer Res 2003;63:6004–7.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Mandola MV, Stoehlmacher J, Muller-Weeks S, et al. A novel single nucleotide polymorphism within the 5′ tandem repeat polymorphism of the thymidylate synthase gene abolishes USF-1 binding and alters transcriptional activity. Cancer Res 2003;63:2898–904.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Kawakami K, Omura K, Kanehira E, Watanabe Y. Polymorphic tandem repeats in the thymidylate synthase gene is associated with its protein expression in human gastrointestinal cancers. Anticancer Res 1999;19:3249–52.
    OpenUrlPubMed
  26. ↵
    Mandola MV, Stoehlmacher J, Zhang W, et al. A 6 bp polymorphism in the thymidylate synthase gene causes message instability and is associated with decreased intratumoral TS mRNA levels. Pharmacogenetics 2004;14:319–27.
    OpenUrlCrossRefPubMed
  27. ↵
    Ulrich CM, Bigler J, Velicer CM, Greene EA, Farin FM, Potter JD. Searching expressed sequence tag databases: discovery and confirmation of a common polymorphism in the thymidylate synthase gene. Cancer Epidemiol Biomarkers Prev 2000;9:1381–5.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Guo S, Thompson E. Performing the exact test of Hardy-Weinberg Proportion for multiple alleles. Biometrics 1992;48:361–72.
    OpenUrlCrossRefPubMed
  29. ↵
    Strassburg CP, Vogel A, Kneip S, Tukey RH, Manns MP. Polymorphisms of the human UDP-glucuronosyltransferase (UGT) 1A7 gene in colorectal cancer. Gut 2002;50:851–6.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Kumagai K, Hiyama K, Oyama T, Maeda H, Kohno N. Polymorphisms in the thymidylate synthase and methylenetetrahydrofolate reductase genes and sensitivity to the low-dose methotrexate therapy in patients with rheumatoid arthritis. Int J Mol Med 2003;11:593–600.
    OpenUrlPubMed
  31. ↵
    Marsh S, Ameyaw MM, Githang'a J, Indalo A, Ofori-Adjei D, McLeod HL. Novel thymidylate synthase enhancer region alleles in African populations. Hum Mutat 2000;16:528.
    OpenUrlPubMed
  32. ↵
    Lampe JW, Bigler J, Horner NK, Potter JD. UDP-glucuronosyltransferase (UGT1A1*28 and UGT1A6*2) polymorphisms in Caucasians and Asians: relationships to serum bilirubin concentrations. Pharmacogenetics 1999;9:341–9.
    OpenUrlCrossRefPubMed
  33. ↵
    Ando M, Ando Y, Sekido Y, Shimokata K, Hasegawa Y. Genetic polymorphisms of the UDP-glucuronosyltransferase 1A7 gene and irinotecan toxicity in Japanese cancer patients. Jpn J Cancer Res 2002;93:591–7.
    OpenUrlCrossRefPubMed
  34. ↵
    Brangi M, Litman T, Ciotti M, et al. Camptothecin resistance: role of the ATP-binding cassette (ABC), mitoxantrone-resistance half-transporter (MXR), and potential for glucuronidation in MXR-expressing cells. Cancer Res 1999;59:5938–46.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Strassburg CP, Manns MP, Tukey RH. Expression of the UDP-glucuronosyltransferase 1A locus in human colon. Identification and characterization of the novel extrahepatic UGT1A8. J Biol Chem 1998;273:8719–26.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Strassburg CP, Oldhafer K, Manns MP, Tukey RH. Differential expression of the UGT1A locus in human liver, biliary, and gastric tissue: identification of UGT1A7 and UGT1A10 transcripts in extrahepatic tissue. Mol Pharmacol 1997;52:212–20.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Gupta E, Wang X, Ramirez J, Ratain MJ. Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother Pharmacol 1997;39:440–4.
    OpenUrlCrossRefPubMed
  38. ↵
    Atsumi R, Suzuki W, Hakusui H. Identification of the metabolites of irinotecan, a new derivative of camptothecin, in rat bile and its biliary excretion. Xenobiotica 1991;21:1159–69.
    OpenUrlPubMed
  39. ↵
    Kaneda N, Yokokura T. Nonlinear pharmacokinetics of CPT-11 in rats. Cancer Res 1990;50:1721–5.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Roberts MS, Magnusson BM, Burczynski FJ, Weiss M. Enterohepatic circulation: physiological, pharmacokinetic and clinical implications. Clin Pharmacokinet 2002;41:751–90.
    OpenUrlCrossRefPubMed
  41. ↵
    de Jonge MJ, Verweij J, de Bruijn P, et al. Pharmacokinetic, metabolic, and pharmacodynamic profiles in a dose-escalating study of irinotecan and cisplatin. J Clin Oncol 2000;18:195–203.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Takasuna K, Hagiwara T, Hirohashi M, et al. Involvement of β-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res 1996;56:3752–7.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Horikawa M, Kato Y, Sugiyama Y. Reduced gastrointestinal toxicity following inhibition of the biliary excretion of irinotecan and its metabolites by probenecid in rats. Pharm Res 2002;19:1345–53.
    OpenUrlCrossRefPubMed
  44. ↵
    Alimonti A, Gelibter A, Pavese I. et al. New approaches to prevent intestinal toxicity of irinotecan-based regimens. Cancer Treat Rev 2004;30:555–62.
    OpenUrlCrossRefPubMed
  45. ↵
    McLeod HL, Watters JW. Irinotecan pharmacogenetics: is it time to intervene? J Clin Oncol 2004;22:1356–9.
    OpenUrlFREE Full Text
  46. ↵
    Desai AA, Innocenti F, Ratain MJ. Pharmacogenomics: road to anticancer therapeutics nirvana? Oncogene 2003;22:6621–8.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Clinical Cancer Research: 11 (3)
February 2005
Volume 11, Issue 3
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
UGT1A7 and UGT1A9 Polymorphisms Predict Response and Toxicity in Colorectal Cancer Patients Treated with Capecitabine/Irinotecan
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
UGT1A7 and UGT1A9 Polymorphisms Predict Response and Toxicity in Colorectal Cancer Patients Treated with Capecitabine/Irinotecan
Leslie E. Carlini, Neal J. Meropol, John Bever, Michael L. Andria, Todd Hill, Philip Gold, Andre Rogatko, Hao Wang and Rebecca L. Blanchard
Clin Cancer Res February 1 2005 (11) (3) 1226-1236;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
UGT1A7 and UGT1A9 Polymorphisms Predict Response and Toxicity in Colorectal Cancer Patients Treated with Capecitabine/Irinotecan
Leslie E. Carlini, Neal J. Meropol, John Bever, Michael L. Andria, Todd Hill, Philip Gold, Andre Rogatko, Hao Wang and Rebecca L. Blanchard
Clin Cancer Res February 1 2005 (11) (3) 1226-1236;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Radiation and TGFβ Blockade in Metastatic Breast Cancer
  • Biomarker Analysis from the BERIL-1 Study
  • Novel Intermediate Endpoint in Immunotherapy Studies
Show more Cancer Therapy: Clinical
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement