REFERENCES

1. Relling MV, Evans WE. Pharmacogenomics in the clinic. Nature 2015;526:343-50.

2. Filipski KK, Mechanic LE, Long R, Freedman AN. Pharmacogenomics in oncology care. Front Genet 2014;5:73.

3. Gillis NK, Patel JN, Innocenti F. Clinical implementation of germ line cancer pharmacogenetic variants during the next-generation sequencing era. Clin Pharmacol Ther 2014;95:269-80.

4. Oscarson M. Pharmacogenetics of drug metabolising enzymes: importance for personalised medicine. Clin Chem Lab Med 2003;41:573-80.

5. Caudle KE, Thorn CF, Klein TE, Swen JJ, McLeod HL, et al. Clinical pharmacogenetics implementation consortium guidelines for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing. Clin Pharmacol Ther 2013;94:640-5.

6. Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin adjuvant (HERA) trial. Lancet 2017;389:1195-205.

7. O’Dwyer ME, Druker BJ. STI571: an inhibitor of the BCR-ABL tyrosine kinase for the treatment of chronic myelogenous leukaemia. Lancet Oncol 2000;1:207-11.

8. Carr DF, Pirmohamed M. Biomarkers of adverse drug reactions. Exp Biol Med (Maywood) 2018;243:291-9.

9. Abaji R, Krajinovic M. Thiopurine S-methyltransferase polymorphisms in acute lymphoblastic leukemia, inflammatory bowel disease and autoimmune disorders: influence on treatment response. Pharmgenomics Pers Med 2017;10:143-56.

10. Lee SHR, Yang JJ. Pharmacogenomics in acute lymphoblastic leukemia. Best Pract Res Clin Haematol 2017;30:229-36.

11. Paugh SW, Stocco G, McCorkle JR, Diouf B, Crews KR, et al. Cancer pharmacogenomics. Clin Pharmacol Ther 2011;90:461-6.

12. Schwab M, Schaffeler E, Marx C, Fischer C, Lang T, et al. Azathioprine therapy and adverse drug reactions in patients with inflammatory bowel disease: impact of thiopurine S-methyltransferase polymorphism. Pharmacogenetics 2002;12:429-36.

13. Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, et al. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther 2011;89:387-91.

14. Schaeffeler E, Fischer C, Brockmeier D, Wernet D, Moerike K, et al. Comprehensive analysis of thiopurine S-methyltransferase phenotype-genotype correlation in a large population of German-Caucasians and identification of novel TPMT variants. Pharmacogenetics 2004;14:407-17.

15. Wellmann R, Borden BA, Danahey K, Nanda R, Polite BN, et al. Analyzing the clinical actionability of germline pharmacogenomic findings in oncology. Cancer 2018;124:3052-65.

16. Kim JH, Cheon JH, Hong SS, Eun CS, Byeon JS, et al. Influences of thiopurine methyltransferase genotype and activity on thiopurine-induced leukopenia in Korean patients with inflammatory bowel disease: a retrospective cohort study. J Clin Gastroenterol 2010;44:e242-8.

17. Jung YS, Cheon JH, Park JJ, Moon CM, Kim ES, et al. Correlation of genotypes for thiopurine methyltransferase and inosine triphosphate pyrophosphatase with long-term clinical outcomes in Korean patients with inflammatory bowel diseases during treatment with thiopurine drugs. J Hum Genet 2010;55:121-3.

18. Yang SK, Hong M, Baek J, Choi H, Zhao W, et al. A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 2014;46:1017-20.

19. Moriyama T, Nishii R, Lin TN, Kihira K, Toyoda H, et al. The effects of inherited NUDT15 polymorphisms on thiopurine active metabolites in Japanese children with acute lymphoblastic leukemia. Pharmacogenet Genomics 2017;27:236-9.

20. Moriyama T, Yang YL, Nishii R, Ariffin H, Liu C, et al. Novel variants in NUDT15 and thiopurine intolerance in children with acute lymphoblastic leukemia from diverse ancestry. Blood 2017;130:1209-12.

21. Yang JJ, Landier W, Yang W, Liu C, Hageman L, et al. Inherited NUDT15 variant is a genetic determinant of mercaptopurine intolerance in children with acute lymphoblastic leukemia. J Clin Oncol 2015;33:1235-42.

22. Yang JJ, Whirl-Carrillo M, Scott SA, Turner AJ, Schwab M, et al. Pharmacogene variation consortium gene introduction: NUDT15. Clin Pharmacol Ther 2018; doi: 10.1002/cpt.1268.

23. Relling MV, Schwab M, Whirl-Carrillo M, Suarez-Kurtz G, Pui CH, et al. Clinical pharmacogenetics implementation consortium (CPIC) guideline for thiopurine dosing based on TPMT and NUDT15 genotypes: 2018 update. Clin Pharmacol Ther 2018; doi: 10.1002/cpt.1304.

24. Bosma P, Chowdhury JR, Jansen PH. Genetic inheritance of Gilbert’s syndrome. Lancet 1995;346:314-5.

25. Bosma PJ, Chowdhury JR, Bakker C, Gantla S, de Boer A, et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert’s syndrome. N Engl J Med 1995;333:1171-5.

26. Dean L. Irinotecan therapy and UGT1A1 genotype. Medical Genetics Summaries. MD, USA: Bethesda; 2012.

27. Barbarino JM, Haidar CE, Klein TE, Altman RB. PharmGKB summary: very important pharmacogene information for UGT1A1. Pharmacogenet Genomics 2014;24:177-83.

28. Hoskins JM, Goldberg RM, Qu P, Ibrahim JG, McLeod HL. UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst 2007;99:1290-5.

29. Etienne-Grimaldi MC, Boyer JC, Thomas F, Quaranta S, Picard N, et al. UGT1A1 genotype and irinotecan therapy: general review and implementation in routine practice. Fundam Clin Pharmacol 2015;29:219-37.

30. Fujii H, Yamada Y, Watanabe D, Matsuhashi N, Takahashi T, et al. Dose adjustment of irinotecan based on UGT1A1 polymorphisms in patients with colorectal cancer. Cancer Chemother Pharmacol 2019;83:123-9.

31. Tejpar S, Yan P, Piessevaux H, Dietrich D, Brauchli P, et al. Clinical and pharmacogenetic determinants of 5-fluorouracyl/leucovorin/irinotecan toxicity: results of the PETACC-3 trial. Eur J Cancer 2018;99:66-77.

32. Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018;39:18-40.

33. Meinsma R, Fernandez-Salguero P, Van Kuilenburg AB, Van Gennip AH, Gonzalez FJ. Human polymorphism in drug metabolism: mutation in the dihydropyrimidine dehydrogenase gene results in exon skipping and thymine uracilurea. DNA Cell Biol 1995;14:1-6.

34. Saif MW, Ezzeldin H, Vance K, Sellers S, Diasio RB. DPYD*2A mutation: the most common mutation associated with DPD deficiency. Cancer Chemother Pharmacol 2007;60:503-7.

35. Offer SM, Lee AM, Mattison LK, Fossum C, Wegner NJ, et al. A DPYD variant (Y186C) in individuals of african ancestry is associated with reduced DPD enzyme activity. Clin Pharmacol Ther 2013;94:158-66.

36. Khushman M, Patel GK, Hosein PJ, Laurini JA, Cameron D, et al. Germline pharmacogenomics of DPYD*9A (c.85T>C) variant in patients with gastrointestinal malignancies treated with fluoropyrimidines. J Gastrointest Oncol 2018;9:416-24.

37. Morel A, Boisdron-Celle M, Fey L, Soulie P, Craipeau MC, et al. Clinical relevance of different dihydropyrimidine dehydrogenase gene single nucleotide polymorphisms on 5-fluorouracil tolerance. Mol Cancer Ther 2006;5:2895-904.

38. Offer SM, Wegner NJ, Fossum C, Wang K, Diasio RB. Phenotypic profiling of DPYD variations relevant to 5-fluorouracil sensitivity using real-time cellular analysis and in vitro measurement of enzyme activity. Cancer Res 2013;73:1958-68.

39. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JHM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther 2018;103:210-6.

40. Toffoli G, Innocenti F, Polesel J, De Mattia E, Sartor F, et al. The genotype for DPYD risk variants in patients with colorectal cancer and the related toxicity management costs in clinical practice. Clin Pharmacol Ther 2018; doi: 10.1002/cpt.1257.

41. Henricks LM, Lunenburg C, de Man FM, Meulendijks D, Frederix GWJ, et al. DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 2018;19:1459-67.

42. Schroth W, Antoniadou L, Fritz P, Schwab M, Muerdter T, et al. Breast cancer treatment outcome with adjuvant tamoxifen relative to patient CYP2D6 and CYP2C19 genotypes. J Clin Oncol 2007;25:5187-93.

43. Goetz MP, Sangkuhl K, Guchelaar HJ, Schwab M, Province M, et al. Clinical pharmacogenetics implementation consortium (CPIC) guideline for CYP2D6 and tamoxifen therapy. Clin Pharmacol Ther 2018;103:770-7.

44. Brauch H, Schroth W, Goetz MP, Murdter TE, Winter S, et al. Tamoxifen use in postmenopausal breast cancer: CYP2D6 matters. J Clin Oncol 2013;31:176-80.

45. Li J, Czene K, Brauch H, Schroth W, Saladores P, et al. Association of CYP2D6 metabolizer status with mammographic density change in response to tamoxifen treatment. Breast Cancer Res 2013;15:R93.

46. Province MA, Goetz MP, Brauch H, Flockhart DA, Hebert JM, et al. CYP2D6 genotype and adjuvant tamoxifen: meta-analysis of heterogeneous study populations. Clin Pharmacol Ther 2014;95:216-27.

47. Pelizzari G, Arpino G, Biganzoli L, Cinieri S, De Laurentiis M, et al. An Italian delphi study to evaluate consensus on adjuvant endocrine therapy in premenopausal patients with breast cancer: the ERA project. BMC Cancer 2018;18:932.

48. Francis PA, Pagani O, Fleming GF, Walley BA, Colleoni M, et al. Tailoring adjuvant endocrine therapy for premenopausal breast cancer. N Engl J Med 2018;379:122-37.

49. Fabian CJ. The what, why and how of aromatase inhibitors: hormonal agents for treatment and prevention of breast cancer. Int J Clin Pract 2007;61:2051-63.

50. Morales L, Neven P, Paridaens R. Choosing between an aromatase inhibitor and tamoxifen in the adjuvant setting. Curr Opin Oncol 2005;17:559-65.

51. Drogemoller BI, Wright GEB, Shih J, Monzon JG, Gelmon KA, et al. CYP2D6 as a treatment decision aid for ER-positive non-metastatic breast cancer patients: a systematic review with accompanying clinical practice guidelines. Breast Cancer Res Treat 2018; doi: 10.1007/s10549-018-5027-0.

52. Mayer SE, Weiss NS, Chubak J, Doody DR, Carlson CS, et al. CYP2D6-inhibiting medication use and inherited CYP2D6 variation in relation to adverse breast cancer outcomes after tamoxifen therapy. Cancer Causes Control 2019;30:103-12.

53. Sanchez-Spitman A, Dezentje V, Swen J, Moes D, Bohringer S, et al. Tamoxifen pharmacogenetics and metabolism: results from the prospective CYPTAM study. J Clin Oncol 2019; doi: 10.1200/JCO.18.00307.

54. Aminkeng F, Ross CJ, Rassekh SR, Hwang S, Rieder MJ, et al. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol 2016;82:683-95.

55. Nagar S, Zalatoris JJ, Blanchard RL. 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.

56. Krishnaswamy S, Hao Q, Al-Rohaimi A, Hesse LM, von Moltke LL, et al. UDP glucuronosyltransferase (UGT) 1A6 pharmacogenetics: II. Functional impact of the three most common nonsynonymous UGT1A6 polymorphisms (S7A, T181A, and R184S). J Pharmacol Exp Ther 2005;313:1340-6.

57. Visscher H, Ross CJ, Rassekh SR, Sandor GS, Caron HN, et al. Validation of variants in SLC28A3 and UGT1A6 as genetic markers predictive of anthracycline-induced cardiotoxicity in children. Pediatr Blood Cancer 2013;60:1375-81.

58. Aminkeng F, Bhavsar AP, Visscher H, Rassekh SR, Li Y, et al. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet 2015;47:1079-84.

59. Wojnowski L, Kulle B, Schirmer M, Schluter G, Schmidt A, et al. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation 2005;112:3754-62.

60. Chen SH, Pei D, Yang W, Cheng C, Jeha S, et al. Genetic variations in GRIA1 on chromosome 5q33 related to asparaginase hypersensitivity. Clin Pharmacol Ther 2010;88:191-6.

61. Lopez-Santillan M, Iparraguirre L, Martin-Guerrero I, Gutierrez-Camino A, Garcia-Orad A. Review of pharmacogenetics studies of L-asparaginase hypersensitivity in acute lymphoblastic leukemia points to variants in the GRIA1 gene. Drug Metab Pers Ther 2017;32:1-9.

62. Lee JJ, Swain SM. Peripheral neuropathy induced by microtubule-stabilizing agents. J Clin Oncol 2006;24:1633-42.

63. Zhang Y, Zolov SN, Chow CY, Slutsky SG, Richardson SC, et al. Loss of Vac14, a regulator of the signaling lipid phosphatidylinositol 3,5-bisphosphate, results in neurodegeneration in mice. Proc Natl Acad Sci U S A 2007;104:17518-23.

64. Sucheston-Campbell LE, Clay-Gilmour AI, Barlow WE, Budd GT, Stram DO, et al. Genome-wide meta-analyses identifies novel taxane-induced peripheral neuropathy-associated loci. Pharmacogenet Genomics 2018;28:49-55.

65. Li W, Zhang H, Assaraf YG, Zhao K, Xu X, et al. Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies. Drug Resist Updat 2016;27:14-29.

66. Bruhn O, Cascorbi I. Polymorphisms of the drug transporters ABCB1, ABCG2, ABCC2 and ABCC3 and their impact on drug bioavailability and clinical relevance. Expert Opin Drug Metab Toxicol 2014;10:1337-54.

67. Cascorbi I, Gerloff T, Johne A, Meisel C, Hoffmeyer S, et al. Frequency of single nucleotide polymorphisms in the P-glycoprotein drug transporter MDR1 gene in white subjects. Clin Pharmacol Ther 2001;69:169-74.

68. Haenisch S, Zimmermann U, Dazert E, Wruck CJ, Dazert P, et al. Influence of polymorphisms of ABCB1 and ABCC2 on mRNA and protein expression in normal and cancerous kidney cortex. Pharmacogenomics J 2007;7:56-65.

69. Plasschaert SL, Groninger E, Boezen M, Kema I, de Vries EG, et al. Influence of functional polymorphisms of the MDR1 gene on vincristine pharmacokinetics in childhood acute lymphoblastic leukemia. Clin Pharmacol Ther 2004;76:220-9.

70. Zheng Q, Wu H, Yu Q, Kim DH, Lipton JH, et al. ABCB1 polymorphisms predict imatinib response in chronic myeloid leukemia patients: a systematic review and meta-analysis. Pharmacogenomics J 2015;15:127-34.

71. Zu B, Li Y, Wang X, He D, Huang Z, et al. MDR1 gene polymorphisms and imatinib response in chronic myeloid leukemia: a meta-analysis. Pharmacogenomics 2014;15:667-77.

72. Jeong H, Herskowitz I, Kroetz DL, Rine J. Function-altering SNPs in the human multidrug transporter gene ABCB1 identified using a Saccharomyces-based assay. PLoS Genet 2007;3:e39.

73. Sakurai A, Tamura A, Onishi Y, Ishikawa T. Genetic polymorphisms of ATP-binding cassette transporters ABCB1 and ABCG2: therapeutic implications. Expert Opin Pharmacother 2005;6:2455-73.

74. Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood 2002;99:507-12.

75. Jordanides NE, Jorgensen HG, Holyoake TL, Mountford JC. Functional ABCG2 is overexpressed on primary CML CD34+ cells and is inhibited by imatinib mesylate. Blood 2006;108:1370-3.

76. Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist 2003;8:411-24.

77. Litman T, Brangi M, Hudson E, Fetsch P, Abati A, et al. The multidrug-resistant phenotype associated with overexpression of the new ABC half-transporter, MXR (ABCG2). J Cell Sci 2000;113:2011-21.

78. Kim DH, Sriharsha L, Xu W, Kamel-Reid S, Liu X, et al. Clinical relevance of a pharmacogenetic approach using multiple candidate genes to predict response and resistance to imatinib therapy in chronic myeloid leukemia. Clin Cancer Res 2009;15:4750-8.

79. van der Veldt AA, Eechoute K, Gelderblom H, Gietema J, Guchelaar HJ, et al. Genetic polymorphisms associated with a prolonged progression-free survival in patients with metastatic renal cell cancer treated with sunitinib. Clin Cancer Res 2011;17:620-9.

80. Rudin CM, Liu W, Desai A, Karrison T, Jiang X, et al. Pharmacogenomic and pharmacokinetic determinants of erlotinib toxicity. J Clin Oncol 2008;26:1119-27.

81. Sparreboom A, Loos WJ, Burger H, Sissung TM, Verweij J, et al. Effect of ABCG2 genotype on the oral bioavailability of topotecan. Cancer Biol Ther 2005;4:650-8.

82. Takahashi N, Miura M, Scott SA, Kagaya H, Kameoka Y, et al. Influence of CYP3A5 and drug transporter polymorphisms on imatinib trough concentration and clinical response among patients with chronic phase chronic myeloid leukemia. J Hum Genet 2010;55:731-7.

83. Dohse M, Scharenberg C, Shukla S, Robey RW, Volkmann T, et al. Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib. Drug Metab Dispos 2010;38:1371-80.

84. Ripperger A, Benndorf RA. The C421A (Q141K) polymorphism enhances the 3’-untranslated region (3’-UTR)-dependent regulation of ATP-binding cassette transporter ABCG2. Biochem Pharmacol 2016;104:139-47.

85. Imai Y, Nakane M, Kage K, Tsukahara S, Ishikawa E, et al. C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141K protein and low-level drug resistance. Mol Cancer Ther 2002;1:611-6.

86. Poonkuzhali B, Lamba J, Strom S, Sparreboom A, Thummel K, et al. Association of breast cancer resistance protein/ABCG2 phenotypes and novel promoter and intron 1 single nucleotide polymorphisms. Drug Metab Dispos 2008;36:780-95.

87. Cascorbi I, Werk AN. Advances and challenges in hereditary cancer pharmacogenetics. Expert Opin Drug Metab Toxicol 2017;13:73-82.

88. Rau T, Erney B, Gores R, Eschenhagen T, Beck J, et al. High-dose methotrexate in pediatric acute lymphoblastic leukemia: impact of ABCC2 polymorphisms on plasma concentrations. Clin Pharmacol Ther 2006;80:468-76.

89. Au A, Baba AA, Azlan H, Norsa’adah B, Ankathil R. Clinical impact of ABCC1 and ABCC2 genotypes and haplotypes in mediating imatinib resistance among chronic myeloid leukaemia patients. J Clin Pharm Ther 2014;39:685-90.

90. Cuffe S, Azad AK, Qiu X, Qiu X, Brhane Y, et al. ABCC2 polymorphisms and survival in the Princess Margaret cohort study and the NCIC clinical trials group BR.24 trial of platinum-treated advanced stage non-small cell lung cancer patients. Cancer Epidemiol 2016;41:50-6.

91. Kaehler M, Ruemenapp J, Gonnermann D, Nagel I, Bruhn O, et al. MicroRNA-212/ABCG2-axis contributes to development of imatinib-resistance in leukemic cells. Oncotarget 2017;8:92018-31.

92. Oberstadt MC, Bien-Moller S, Weitmann K, Herzog S, Hentschel K, et al. Epigenetic modulation of the drug resistance genes MGMT, ABCB1 and ABCG2 in glioblastoma multiforme. BMC cancer 2013;13:617.

93. Bram EE, Stark M, Raz S, Assaraf YG. Chemotherapeutic drug-induced ABCG2 promoter demethylation as a novel mechanism of acquired multidrug resistance. Neoplasia 2009;11:1359-70.

94. Reed K, Hembruff SL, Sprowl JA, Parissenti AM. The temporal relationship between ABCB1 promoter hypomethylation, ABCB1 expression and acquisition of drug resistance. Pharmacogenomics J 2010;10:489-504.

95. To KK, Zhan Z, Bates SE. Aberrant promoter methylation of the ABCG2 gene in renal carcinoma. Mol Cell Biol 2006;26:8572-85.

96. Turner JG, Gump JL, Zhang C, Cook JM, Marchion D, et al. ABCG2 expression, function, and promoter methylation in human multiple myeloma. Blood 2006;108:3881-9.

97. Habano W, Kawamura K, Iizuka N, Terashima J, Sugai T, et al. Analysis of DNA methylation landscape reveals the roles of DNA methylation in the regulation of drug metabolizing enzymes. Clin Epigenetics 2015;7:105.

98. Peng L, Zhong X. Epigenetic regulation of drug metabolism and transport. Acta Pharm Sin B 2015;5:106-12.

99. Zhao H, Wang D, Du W, Gu D, Yang R. MicroRNA and leukemia: tiny molecule, great function. Crit Rev Oncol Hematol 2010;74:149-55.

100. Shang Y, Zhang Z, Liu Z, Feng B, Ren G, et al. miR-508-5p regulates multidrug resistance of gastric cancer by targeting ABCB1 and ZNRD1. Oncogene 2014;33:3267-76.

101. Haenisch S, Laechelt S, Bruckmueller H, Werk A, Noack A, et al. Down-regulation of ATP-binding cassette C2 protein expression in HepG2 cells after rifampicin treatment is mediated by microRNA-379. Mol Pharmacol 2011;80:314-20.

102. Werk AN, Bruckmueller H, Haenisch S, Cascorbi I. Genetic variants may play an important role in mRNA-miRNA interaction: evidence for haplotype-dependent downregulation of ABCC2 (MRP2) by miRNA-379. Pharmacogenet Genomics 2014;24:283-91.

103. To KK, Zhan Z, Litman T, Bates SE. Regulation of ABCG2 expression at the 3’ untranslated region of its mRNA through modulation of transcript stability and protein translation by a putative microRNA in the S1 colon cancer cell line. Mol Cell Biol 2008;28:5147-61.

104. Bruhn O, Drerup K, Kaehler M, Haenisch S, Roder C, et al. Length variants of the ABCB1 3’-UTR and loss of miRNA binding sites: possible consequences in regulation and pharmacotherapy resistance. Pharmacogenomics 2016;17:327-40.

105. He Y, Chevillet JR, Liu G, Kim TK, Wang K. The effects of microRNA on the absorption, distribution, metabolism and excretion of drugs. Br J Pharmacol 2015;172:2733-47.

106. Zanger UM, Klein K, Kugler N, Petrikat T, Ryu CS. Epigenetics and MicroRNAs in Pharmacogenetics. Adv Pharmacol 2018;83:33-64.

107. Zheng T, Wang J, Chen X, Liu L. Role of microRNA in anticancer drug resistance. Int J Cancer 2010;126:2-10.

108. Polkowska M, Ekk-Cierniakowski P, Czepielewska E, Kozlowska-Wojciechowska M. Efficacy and safety of BRAF inhibitors and anti-CTLA4 antibody in melanoma patients-real-world data. Eur J Clin Pharmacol 2019;75:329-34.

109. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015;161:205-14.

110. Xu C, Chen YP, Du XJ, Liu JQ, Huang CL, et al. Comparative safety of immune checkpoint inhibitors in cancer: systematic review and network meta-analysis. BMJ 2018;363:k4226.

Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/