Pharmacogenetic markers of chemotherapy toxicity in gastrointestinal tumors: a preliminary analysis

Cover Page

Cite item

Full Text

Abstract

Aim. To assess the association between the carriage of minor allelic variants of 8 genes that encode key enzymes involved in the metabolism of anticancer drugs (DPYD, GSTP1, MTHFR, UGT1A1) and cell repair (XPC, ERCC1, TYMP, NQO1) and the severity of adverse drug events in patients with common gastrointestinal tumors.

Tasks. To study the frequency of minor allelic variants of the DPYD, GSTP1, MTHFR, UGT1A1, XPC, ERCC1, TYMP, NQO1 genes; to assess the frequency and severity of adverse drug events of chemotherapy treatment in the study population.

Materials and methods. For the period from October 2020 to April 2021, 56 patients (women – 29, men – 27) with verified malignant tumors of the gastrointestinal tract were included in a prospective clinical study as a part of the RSF grant No. 20-75-10158. The mean age was 62.3±11.4 years. Colon cancer was detected in 24 patients, tumors of the esophagus and stomach – in 19 patients, tumors of pancreas and biliary tract – in 13 patients. First-line palliative chemotherapy was given to 27 patients, adjuvant – 19 patients, neoadjuvant – 10 patients. All patients had not previously received cytotoxic or radiation treatment. Point nucleotide variants of genes DPYD, XPC, GSTP1, MTHFR, ERCC1, UGT1A1, TYMPS, NQO1 were determined by hybridization analysis on biological microchips. Differences in the tolerance of cytotoxic therapy (5-fluorouracil, platinum preparations, irinotecan) depending on the genotype were assessed using Fisher’s exact test.

Results. The average number of chemotherapy courses received was 4.2±2.6 (1–12). There was a statistically significant difference in the tolerability of chemotherapy in patients with minor allelic variants of the GSTP1 rs1695 (p=0.03), ERCC1 rs11615 (p=0.01), and UGT1A1 rs8175347 (p=0.003) genes.

Conclusion. The use of hybridization analysis on biological microchips to assess allelic variants responsible for the tolerability of cytotoxic therapy is reasonable and requires further prospective assessment.

About the authors

Denis S. Fedorinov

Russian Medical Academy of Continuous Professional Education; City Clinical Cancer Hospital №1

Author for correspondence.
Email: Fedorinov.denis@gmail.com
ORCID iD: 0000-0001-5516-7367

Senior laboratory assistant

Russian Federation, Moscow; Moscow

Rustam N. Geidarov

Engelhardt Institute of Molecular Biology

Email: Fedorinov.denis@gmail.com
ORCID iD: 0000-0002-4971-2629

Senior laboratory assistant

Russian Federation, Moscow

Igor A. Shashkov

Engelhardt Institute of Molecular Biology

Email: Fedorinov.denis@gmail.com
ORCID iD: 0000-0001-6037-730X

Cand. Sci. (Chem.)

Russian Federation, Moscow

Vladimir M. Mikhailovich

Engelhardt Institute of Molecular Biology

Email: Fedorinov.denis@gmail.com
ORCID iD: 0000-0003-4894-1304

D. Sci. (Biol.)

Russian Federation, Moscow

Marina A. Lyadova

City Clinical Cancer Hospital №1

Email: dr.lyadova@gmail.com
ORCID iD: 0000-0002-9558-5579

Cand. Sci. (Med.)

Russian Federation, Moscow

Ilya A. Pokataev

City Clinical Cancer Hospital №1

Email: Fedorinov.denis@gmail.com
ORCID iD: 0000-0001-9864-3837

D. Sci. (Med.)

Russian Federation, Moscow

Vladimir K. Lyadov

Russian Medical Academy of Continuous Professional Education; City Clinical Cancer Hospital №1; Novokuznetsk State Institute of Postgraduate Medical Education – branch of Russian Medical Academy of Continuous Professional Education

Email: vlyadov@gmail.com
ORCID iD: 0000-0002-7281-3591

D. Sci. (Med.)

Russian Federation, Moscow; Moscow; Novokuznetsk

References

  1. Состояние онкологической помощи населению России в 2018 году. Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М., 2019 [The state of cancer care for the population of Russia in 2018. Ed. AD Kaprin, VV Starinsky, GV Petrova. Moscow, 2019 (in Russian)].
  2. Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med. 2018;379(25):2395-406.
  3. Al-Batran S-E, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet Lond Engl. 2019;393(10184):1948-57.
  4. Cremolini C, Loupakis F, Antoniotti C, et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015;16(13):1306-15.
  5. De Man FM, Goey AKL, van Schaik RHN, et al. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics. Clin Pharmacokinet. 2018;57(10):1229-54.
  6. Abad A, Martínez-Balibrea E, Viéitez JM, et al. Genotype-based selection of treatment of patients with advanced colorectal cancer (SETICC): a pharmacogenetic-based randomized phase II trial. Ann Oncol. 2018;29(2):439-44.
  7. Van Kuilenburg AB, Vreken P, Abeling NG, et al. Genotype and phenotype in patients with dihydropyrimidine dehydrogenase deficiency. Hum Genet. 1999;104(1):1-9.
  8. Lamas MJ, Duran G, Balboa E, et al. Use of a comprehensive panel of biomarkers to predict response to a fluorouracil-oxaliplatin regimen in patients with metastatic colorectal cancer. Pharmacogenomics. 2011;12(3):433-42.
  9. Tran P, Leclerc D, Chan M, et al. Multiple transcription start sites and alternative splicing in the methylenetetrahydrofolate reductase gene result in two enzyme isoforms. Mamm Genome. 2002;13(9):483-92.
  10. Nahid NA, Apu MNH, Islam MR, et al. DPYD*2A and MTHFR C677T predict toxicity and efficacy, respectively, in patients on chemotherapy with 5-fluorouracil for colorectal cancer. Cancer Chemother Pharmacol. 2018;81(1):119-29.
  11. Chen Y-J, Hu F, Li C-Y, et al. The association of UGT1A1*6 and UGT1A1*28 with irinotecan-induced neutropenia in Asians: a meta-analysis. Biomarkers. 2014;19(1):56-62.
  12. Han F, Guo C, Yu D, et al. Associations between UGT1A1*6 or UGT1A1*6/*28 polymorphisms and irinotecan-induced neutropenia in Asian cancer patients. Cancer Chemother Pharmacol. 2014;73(4):779-88.
  13. Sakano S, Hinoda Y, Sasaki M, et al. Nucleotide excision repair gene polymorphisms may predict acute toxicity in patients treated with chemoradiotherapy for bladder cancer. Pharmacogenomics. 2010;11(10):1377-87.
  14. Khrunin AV, Moisseev A, Gorbunova V, Limborska S. Genetic polymorphisms and the efficacy and toxicity of cisplatin-based chemotherapy in ovarian cancer patients. Pharmacogenomics J. 2010;10(1):54-61.
  15. Giovannetti E, Pacetti P, Reni M, et al. Association between DNA-repair polymorphisms and survival in pancreatic cancer patients treated with combination chemotherapy. Pharmacogenomics. 2011;12(12):1641-52.
  16. Pérez-Ramírez C, Cañadas-Garre M, Alnatsha A, et al. Pharmacogenetic predictors of toxicity to platinum based chemotherapy in non-small cell lung cancer patients. Pharmacol Res. 2016;111:877-84.
  17. Castro-Rojas CA, Esparza-Mota AR, Hernandez-Cabrera F, et al. Thymidylate synthase gene variants as predictors of clinical response and toxicity to fluoropyrimidine-based chemotherapy for colorectal cancer. Drug Metab Pers Ther. 2017;32(4):209-18.
  18. Geng R, Chen Z, Zhao X, et al. Oxidative stress-related genetic polymorphisms are associated with the prognosis of metastatic gastric cancer patients treated with epirubicin, oxaliplatin and 5-fluorouracil combination chemotherapy. PloS One. 2014;9(12):e116027.

Copyright (c) 2021 Consilium Medicum

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
 


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies