DOI QR코드

DOI QR Code

Clinical Observations on Associations Between the UGT1A1 Genotype and Severe Toxicity of Irinotecan

  • Lu, Yan-Yan (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Huang, Xin-En (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Wu, Xue-Yan (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Cao, Jie (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Liu, Jin (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Wang, Lin (Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research) ;
  • Xiang, Jin (Department of Research, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University & Jingsu Institute of Cancer Research)
  • Published : 2014.04.01

Abstract

Background: Severe toxicity is commonly observed in cancer patients receiving irinotecan (CPT-11) UDPglucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Our study aimed to assess this point to guide clinical use of CPT-11. Materials and Methods: 89 cancer patients with advanced disease received CPT-11-based chemotherapy for at least two cycles. Toxicity, including GI and hematologic toxicity was recorded in detail and UGT1A1 variants were genotyped. Regression analysis was used to analyse relationships between these variables and tumor response. Results: The prevalence of grade III-IV diarrhea was 10.1%, this being more common in patients with the TA 6/7 genotype (5 of 22 patients, 22.7%) (p<0.05). The prevalence of grade III-IV neutropenia was 13.4%and also highest in patients with the TA 6/7 genotype (4 of 22 patients; 18.2%) but without significance (p>0.05). The retreatment total bilirubin levels were significantly higher in TA6/7 patients (mean, $12.75{\mu}mol/L$) with compared to TA6/6 (mean, $9.92{\mu}mol/L$) with p<0.05. Conclusions: Our study support the conclusion that patients with a $UGT1A1^*28$ allele (s) will suffer an increased risk of severe irinotecan-induced diarrhea, whether with mid-or low-dosage. However, the $UGT1A1^*28$ allele (s) did not increase severe neutropenia. Higher serum total bilirubin is an indication that patients UGT1A1 genotype is not wild-type, with significance for clinic usage of CPT-11.

Keywords

References

  1. Araki K (2006), Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Cancer Sci, 97, 1255-9. https://doi.org/10.1111/j.1349-7006.2006.00321.x
  2. Ando Y (2000). Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res, 60, 6921-6.
  3. Beutler E, Gelbart T, Demina A (1998), Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Aca Sci USA, 95, 8170-4. https://doi.org/10.1073/pnas.95.14.8170
  4. Bosma PJ, Chowdhury JR, Bakker C, et al (1995). The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert's syndrome. N Engl J Med, 333, 1171-5. https://doi.org/10.1056/NEJM199511023331802
  5. Bozkurt O, Karaca H, Ciltas A, et al (2014). Efficacy and Safety of Raltitrexed Combinations with Uracil- Tegafur or Mitomycin C as Salvage Treatment in Advanced Colorectal Cancer Patients: A Multicenter Study of Anatolian Society of Medical Oncology (ASMO). Asian Pac J Cancer Prev. 15, 1845-9. https://doi.org/10.7314/APJCP.2014.15.4.1845
  6. Carlini LE, Meropol NJ, Bever J (2005). UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res, 11, 1226-36.
  7. Cunningham D, Pyrhonen S, James RD, et al (1998). Randomised trial of irinotecan plus supportive care versus supportive care alone after fluorouracil failure for patients with metastatic colorectal cancer. Lancet, 352, 1413-8. https://doi.org/10.1016/S0140-6736(98)02309-5
  8. EGAPP Working Group (2009). Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Genet Med, 11, 15-20. https://doi.org/10.1097/GIM.0b013e31818efd9d
  9. Fuchs CS, Moore MR, Harker G, et al (2003). Phase III comparison of two irinotecan dosing regimens in second-line therapy of metastatic colorectal cancer. J Clin Oncol, 21, 807-14. https://doi.org/10.1200/JCO.2003.08.058
  10. Fukuoka M, Niitani H, Suzuki A, et al (1992). A phase II study of CPT-11, a new derivative of camptothecin, for previously untreated non-small-cell lung cancer. J Clin Oncol, 10, 16-20. https://doi.org/10.1200/JCO.1992.10.1.16
  11. Gupta E, Lestingi TM, Mick R, et al (1994). Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res, 54, 3723-5.
  12. Gupta E, Mick R, Ramirez J, et al (1997), Pharmacokinetic and pharmacodynamic evaluation of the topoisomerase inhibitor irinotecan in cancer patients. J Clin Oncol, 15, 1502-10. https://doi.org/10.1200/JCO.1997.15.4.1502
  13. Hoskins JM, Goldberg RM, Qu P, Ibrahim JG, McLeod HL (2007). UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst, 99, 1290-5. https://doi.org/10.1093/jnci/djm115
  14. Hu ZY, Yu Q, Zhao YS et al (2010). Dose-dependent association between UGT1A1*28 polymorphism and irinotecan-induced diarrhoea: a meta-analysis. Eur J Cancer, 46, 1856-65. https://doi.org/10.1016/j.ejca.2010.02.049
  15. Iyer L, King CD, Whitington PF, et al (1998). 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, 101, 847-54. https://doi.org/10.1172/JCI915
  16. Iyer L, Das S, Janisch L, et al (2002), UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics, 2, 43-7. https://doi.org/10.1038/sj.tpj.6500072
  17. Innocenti F, Undevia SD, Iyer L, et al (2004), Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol, 22, 1382-8. https://doi.org/10.1200/JCO.2004.07.173
  18. Ichikawa W, Araki K, Fujita K, et al (2008). UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst, 100, 224-5. https://doi.org/10.1093/jnci/djm302
  19. Kudoh S, Fukuoka M, Masuda N et al (1995). Relationship between the pharmacokinetics of irinotecan and diarrhea during combination chemotherapy with cisplatin. Jpn J Cancer Res, 86, 406-13. https://doi.org/10.1111/j.1349-7006.1995.tb03071.x
  20. Kudoh S, Fujiwara Y, Takada Y, et al (1998). Phase II study of irinotecan combined with cisplatin in patients with previously untreated small-cell lung cancer. West Japan Lung Cancer Group. J Clin Oncol, 16, 1068-74. https://doi.org/10.1200/JCO.1998.16.3.1068
  21. Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K (1991). Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res, 51, 4187-91.
  22. Negoro S, Fukuoka M, Masuda N, et al (1991). Phase I study of weekly intravenous infusions of CPT-11, a new derivative of camptothecin, in the treatment of advanced non-small-cell lung cancer. J Natl Cancer Inst, 83, 1164-8. https://doi.org/10.1093/jnci/83.16.1164
  23. Marcuello E1, Altes A, Menoyo A, et al (2004). UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer, 91, 678-82. https://doi.org/10.1038/sj.bjc.6602042
  24. Masuda N (1999). Randomized trial comparing cisplatin (CDDP) and irinotecan (CPT-11) versus CDDP and vindesine (VDS) versus CPT-11 alone in advanced non-small cell lung cancer (NSCLC), a multicenter phase III study. Proc Am Soc Clin, 18, 459.
  25. Ma D (2011). The relationship between UGT1A1*28 genotypes and toxicity and effect of CPT-11 in Chinese. SUN Yat-sen Univ (Med Sci), 32, 495-9.
  26. Marcuello E, et al. (2004). UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer, 91 (4):678-82. https://doi.org/10.1038/sj.bjc.6602042
  27. Rouits E, Boisdron-Celle M, Dumont A, et al (2004). Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res, 10, 5151-9. https://doi.org/10.1158/1078-0432.CCR-03-0548
  28. Rouits E, Charasson V, Petain A, et al (2008). Pharmacokinetic and pharmacogenetic determinants of the activity and toxicity of irinotecan in metastatic colorectal cancer patients. Br J Cancer, 99, 1239-45. https://doi.org/10.1038/sj.bjc.6604673
  29. Rougier P, Van Cutsem E, Bajetta E, et al (1998). Randomised trial of irinotecan versus fluorouracil by continuous infusion after fluorouracil failure in patients with metastatic colorectal cancer. Lancet, 352, 1407-12. https://doi.org/10.1016/S0140-6736(98)03085-2
  30. Rothenberg ML, Kuhn JG, Burris HA, et al (1993). Phase I and pharmacokinetic trial of weekly CPT-11. J Clin Oncol, 11, 2194-204. https://doi.org/10.1200/JCO.1993.11.11.2194
  31. Reilly JJ, Workman P, et al (1993). Normalisation of anti-cancer drug dosage using body weight and surface area: is it worthwhile? A review of theoretical and practical considerations. Cancer Chemother Pharmacol, 32, 411-8. https://doi.org/10.1007/BF00685883
  32. Raynal C, Pascussi JM, Leguelinel G, et al (2010). Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Mol Cancer, 9, 46.
  33. Rothenberg ML, Kuhn JG, Schaaf LJ, et al (2001). Phase I dose-finding and pharmacokinetic trial of irinotecan (CPT-11) administered every two weeks. Ann Oncol, 12, 1631-41. https://doi.org/10.1023/A:1013157727506
  34. Soepenberg O, Dumez H, Verweij J, et al (2005). Phase I pharmacokinetic, food effect, and pharmacogenetic study of oral irinotecan given as semisolid matrix capsules in patients with solid tumors. Clin Cancer Res, 11, 1504-11. https://doi.org/10.1158/1078-0432.CCR-04-1758
  35. Saltz LB, Cox JV, Blanke C, et al (2000). Irinotecan plus fluororail and leucovorin for metastatc colorectal cancer. Irinotecan Study Group. N Engl J Med, 343, 905-14.
  36. Takasuna K, Hagiwara T, Hirohashi M, et al (1996). Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res, 56, 3752-7.
  37. Toffoli G, Cecchin E, Corona G, et al (2006). The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. J Clin Oncol, 24, 3061-8. https://doi.org/10.1200/JCO.2005.05.5400
  38. Uygun K, Bilici A, Kaya S, et al (2013). XELIRI plus bevacizumab compared with FOLFIRI plus bevacizumab as first-line setting in patients with metastatic colorectal cancer: experiences at two-institutions. Asian Pac J Cancer Prev, 14, 2283-8. https://doi.org/10.7314/APJCP.2013.14.4.2283
  39. Vanhoefer U, Harstrick A, Achterrath W, et al (2001). Irinotecan in the treatment of colorectal cancer: clinical overview. J Clin Oncol, 19, 1501-18. https://doi.org/10.1200/JCO.2001.19.5.1501
  40. Wei GL, Huang XE, Huo JG, et al (2013). Phase II study on pemetrexed-based chemotherapy in treating patients with metastatic gastric cancer not responding to prior palliative chemotherapy. Asian Pac J Cancer Prev, 14, 2703-6. https://doi.org/10.7314/APJCP.2013.14.5.2703
  41. Wu XY, Huang XE, You SX, et al (2013). Phase II study of pemetrexed as second or third line combined chemotherapy in patients with colorectal cancer. Asian Pac J Cancer Prev, 14, 2019-22. https://doi.org/10.7314/APJCP.2013.14.3.2019
  42. Zhang E, Cao W, Cheng C, et al (2014). A systemic analysis of s-1 regimens for treatment of patients with colon cancer. Asian Pac J Cancer Prev, 15, 2191-4. https://doi.org/10.7314/APJCP.2014.15.5.2191

Cited by

  1. A Predictive Model for Evaluating Responsiveness to Pemetrexed Treatment in Patients with Advanced Colorectal Cancer vol.15, pp.14, 2014, https://doi.org/10.7314/APJCP.2014.15.14.5941
  2. Kinase inhibitors and monoclonal antibodies in oncology: clinical implications vol.13, pp.4, 2015, https://doi.org/10.1038/nrclinonc.2015.213
  3. Clinical Study on Safety and Efficacy of JiSaiXin (Recombinant Human Granulocyte Colony Stimulating Factor Injection Manufactured in China) for Chinese Undergoing Chemotherapy vol.16, pp.1, 2015, https://doi.org/10.7314/APJCP.2015.16.1.299
  4. Screening for Patients with Non-small Cell Lung Cancer Who Could Survive Long Term Chemotherapy vol.16, pp.2, 2015, https://doi.org/10.7314/APJCP.2015.16.2.647
  5. Clinical Application of Serum Tumor Abnormal Protein (TAP) in Colorectal Cancer Patients vol.16, pp.8, 2015, https://doi.org/10.7314/APJCP.2015.16.8.3425
  6. Safety and Efficacy of a Mouth-Rinse with Granulocyte Colony Stimulating Factor in Patients with Chemotherapy-Induced Oral Mucositis vol.17, pp.1, 2016, https://doi.org/10.7314/APJCP.2016.17.1.413
  7. A Clinical Study on Juheli (Recombinant Human Interleukin - 11) in the Second Prevention of Chemotherapy Induced Thrombocytopenia vol.17, pp.2, 2016, https://doi.org/10.7314/APJCP.2016.17.2.485
  8. Pharmacogenomics in Pediatric Oncology: Review of Gene—Drug Associations for Clinical Use vol.17, pp.9, 2016, https://doi.org/10.3390/ijms17091502