DOI QR코드

DOI QR Code

Gene Polymorphisms of OPRM1 A118G and ABCB1 C3435T May Influence Opioid Requirements in Chinese Patients with Cancer Pain

  • Gong, Xiao-Di (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Wang, Jiong-Yi (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Liu, Feng (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Yuan, Hai-Hua (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Zhang, Wen-Ying (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Guo, Yue-Hui (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University) ;
  • Jiang, Bin (Department of Oncology, Shanghai Third People's Hospital, School of Medicine, Shanghai Jiao Tong University)
  • Published : 2013.05.30

Abstract

Backgrounds: Polymorphisms of OPRM1 A118G and ABCB1 C3435T have been suggested to contribute to inter-individual variability regarding pain sensitivity, opioid usage, tolerance and dependence and incidence of adverse effects in patients with chronic pain. This study aimed to investigate the association of both two polymorphisms with opioid requirements in Chinese patients with cancer pain. Methods: The genotypes of rs1799971 (OPRM1) and rs1045642 (ABCB1) were determined by PCR-RFLP and direct sequencing methods respectively in 112 patients with cancer-related pain. Comparisons between the different genotype or allele groups were performed with t-tests or one-way ANOVA tests, as appropriate. The potential relationship of allele number with opioid response was performed with a trend Jonckheere-Terpstra test. Results: In the 112 subjects, the frequencies of variant 118 G and 3435T allele were 38.4% and 37.9%, respectively. Significant higher 24h-opioid doses were observed in patients with GG (P=0.0004) and AG + GG (P=0.005) genotypes than the AA carriers. The dominant mutant 118G allele tended to be associated with progressively increasing 24h-opioiddoses (P=0.001). Compared with CC/CT, patients with ABCB1 TT genotype received higher 24h- and weight-surface area-adjusted-24h- opioids doses (P=0.057 and 0.028, respectively). Conclusions: The OPRM1 A118G single nucleotide polymorphism (SNP) is a key contributor for the inter-individual variability in opioidrequirements in Chinese cancer pain patients. This may possibly extend to the ABCB1 C3435T SNP.

Keywords

References

  1. Ambudkar SV, Kimchi-Sarfaty C, Sauna ZE, et al (2003). P-glycoprotein: from genomics to mechanism. Oncogene, 22, 7468-85. https://doi.org/10.1038/sj.onc.1206948
  2. Baldissera VD, de Mattos AA, Coral GP, et al (2012). Evaluation of the C3435T polymorphism in the MDR1 gene in patients with hepatocellular carcinoma. Ann Hepatol, 11, 899-906.
  3. Bayerer B, Stamer U, Hoeft A, et al (2007). Genomic Variations and Transcriptional Regulation of the Human Mu-Opioid Receptor Gene. Eur J Pain, 11, 421-7. https://doi.org/10.1016/j.ejpain.2006.06.004
  4. Befort K, Filliol D, Decaillot FM, et al (2001). A Single Nucleotide Polymorphic Mutation in the Human Mu-Opooid Receptor Severely Impairs Receptor Signaling. J Biol Chem, 276, 3130-7. https://doi.org/10.1074/jbc.M006352200
  5. Beyer A, Koch T, Schroder H, et al (2004). Effect of the A118G polymorphism on binding affinity, potency and agonistmediated endocytosis, desensitization, and resensitization of the human mu-opioid receptor. J Neurochem, 89, 553-60. https://doi.org/10.1111/j.1471-4159.2004.02340.x
  6. Bond C, LaForge KS, Tian M, et al (1998). Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A, 95, 9608-13. https://doi.org/10.1073/pnas.95.16.9608
  7. Campa D, Gioia A, Tomei A, et al (2008). Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther, 83, 559-66. https://doi.org/10.1038/sj.clpt.6100385
  8. Chen CJ, Chin JE, Ueda K, et al (1986). Internal duplication and homology with bacterial transport proteins in the mdr1 (P-glycoprotein) gene from multidrug-resistant human cells. Cell, 47, 381-9. https://doi.org/10.1016/0092-8674(86)90595-7
  9. Elliott AM, Smith BH, Penny KI, et al (1999). The epidemiology of chronic pain in the community. Lancet, 354, 1248-52. https://doi.org/10.1016/S0140-6736(99)03057-3
  10. Fernandez Robles CR, Degnan M, Candiotti KA(2012). Pain and genetics. Curr Opin Anaesthesiol, 25, 444-9. https://doi.org/10.1097/ACO.0b013e3283556228
  11. Fujita K, Ando Y, Yamamoto W, et al (2010). Association of UGT2B7 and ABCB1 genotypes with morphine-induced adverse drug reactions in Japanese patients with cancer. Cancer Chemother Pharmacol, 65, 251-8. https://doi.org/10.1007/s00280-009-1029-2
  12. Haerian BS, Haerian MS (2013). OPRM1 rs1799971 polymorphism and opioid dependence: evidence from a meta-analysis. Pharmacogenomics, 14, 813-24. https://doi.org/10.2217/pgs.13.57
  13. Hamabe W, Maeda T, Fukazawa Y, et al (2006). P-glycoprotein ATPase activating effect of opioid analgesics and their P-glycoprotein-dependent antinociception in mice. Pharmacol Biochem Behav, 85, 629-36. https://doi.org/10.1016/j.pbb.2006.10.018
  14. Hirota T, Ieiri I, Takane H, et al (2003). Sequence variability and candidate gene analysis in two cancer patients with complex clinical outcomes during morphine therapy. Drug Metab Dispos, 31, 677-80 https://doi.org/10.1124/dmd.31.5.677
  15. Hoehe MR, Kopke K, Wendel B, et al (2000). Sequence variability and candidate gene analysis in complex disease: association of mu opioid receptor gene variation with substance dependence. Hum Mol Genet, 9, 2895-908. https://doi.org/10.1093/hmg/9.19.2895
  16. Hoffmeyer S, Burk O, von Richter O, et al (2000). Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A, 97, 3473-8. https://doi.org/10.1073/pnas.97.7.3473
  17. Huang P, Chen C, Mague SD, et al (2012). A common single nucleotide polymorphism A118G of the $\mu$ opioid receptor alters its N-glycosylation and protein stability. Biochem J, 441, 379-86. https://doi.org/10.1042/BJ20111050
  18. Hung CC, Chen CC, Lin CJ, et al (2008). Functional evaluation of polymorphisms in the human ABCB1 gene and the impact on clinical responses of antiepileptic drugs. Pharmacogenet Genomics, 18, 390-402. https://doi.org/10.1097/FPC.0b013e3282f85e36
  19. Ikeda K, Ide S, Han W, et al (2005). How individual sensitivity to opiates can be predicted by gene analyses. Trends Pharmacol Sci, 26, 311-7. https://doi.org/10.1016/j.tips.2005.04.001
  20. Kasai S, Hayashida M, Sora I, et al (2008). Candidate gene polymorphisms predicting individual sensitivity to opioids. Naunyn Schmiedebergs Arch Pharmacol, 377, 269-81. https://doi.org/10.1007/s00210-007-0205-3
  21. Kasai S, Ikeda K (2011). Pharmacogenomics of the human $\mu$-opioid receptor. Pharmacogenomics, 12, 1305-20. https://doi.org/10.2217/pgs.11.68
  22. Kimchi-Sarfaty C, Oh JM, Kim IW, et al (2007). A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science, 315, 525-8. https://doi.org/10.1126/science.1135308
  23. Klepstad P, Fladvad T, Skorpen F, et al (2011). Influence from genetic variability on opioid use for cancer pain: a European genetic association study of 2294 cancer pain patients. Pain, 152, 1139-45. https://doi.org/10.1016/j.pain.2011.01.040
  24. Kosarac B, Fox AA, Collard CD (2009). Effect of genetic factors on opioid action. Curr Opin Anaesthesiol, 22, 476-82. https://doi.org/10.1097/ACO.0b013e32832e34c9
  25. Landau R, Kern C, Columb MO, et al (2008). Genetic variability of the mu-opioid receptor influences intrathecal fentanyl analgesia requirements in laboring women. Pain, 139, 5-14. https://doi.org/10.1016/j.pain.2008.02.023
  26. Leschziner G, Zabaneh D, Pirmohamed M, et al (2006). Exon sequencing and high resolution haplotype analysis of ABC transporter genes implicated in drug resistance. Pharmacogenet Genomics, 16, 439-50. https://doi.org/10.1097/01.fpc.0000197467.21964.67
  27. Li D, Zhang GL, Lou YQ, et al (2007). Genetic polymorphisms in MDR1 and CYP3A5 and MDR1 haplotype in mainland Chinese Han, Uygur and Kazakh ethnic groups. J Clin Pharm Ther, 32, 89-95. https://doi.org/10.1111/j.1365-2710.2007.00791.x
  28. Lloret Linares C, Hajj A, Poitou C, et al (2011). Pilot study examining the frequency of several gene polymorphisms involved in morphine pharmacodynamics and pharmacokinetics in a morbidly obese population. Obes Surg, 21, 1257-64. https://doi.org/10.1007/s11695-010-0143-x
  29. Lotsch J, Geisslinger G (2006). Relevance of frequent muopioid receptor polymorphisms for opioid activity in healthy volunteers. Pharmacogenomics J, 6, 200-10. https://doi.org/10.1038/sj.tpj.6500362
  30. Lotsch J, Skarke C, Wieting J, et al (2006). Modulation of the central nervous effects of levomethadone by genetic polymorphisms potentially affecting its metabolism, distribution, and drug action. Clin Pharmacol Ther, 79, 72-89. https://doi.org/10.1016/j.clpt.2005.09.010
  31. Mealey KL (2008). Canine ABCB1 and macrocyclic lactones: heartworm prevention and pharmacogenetics. Vet Parasitol, 158, 215-22. https://doi.org/10.1016/j.vetpar.2008.09.009
  32. Oertel BG, Schmidt R, Schneider A, et al (2006). The mu-opioid receptor gene polymorphism 118A>G depletes alfentanilinduced analgesia and protects against respiratory depression in homozygous carriers. Pharmacogenet Genomics, 16, 625-36. https://doi.org/10.1097/01.fpc.0000220566.90466.a2
  33. Reyes-Gibby CC, Shete S, Rakvåg T, et al (2007). Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: OPRM1 and COMT gene. Pain, 130, 25-30. https://doi.org/10.1016/j.pain.2006.10.023
  34. Salama NN, Yang Z, Bui T, et al (2006). MDR1 haplotypes significantly minimize intracellular uptake and transcellular P-gp substrate transport in recombinant LLC-PK1 cells. J Pharm Sci, 95, 2293-308. https://doi.org/10.1002/jps.20717
  35. Shabalina SA, Zaykin DV, Gris P, et al (2009). Expansion of the human mu-opioid receptor gene architecture: novel functional variants. Hum Mol Genet, 18, 1037-51. https://doi.org/10.1093/hmg/ddn439
  36. Sia AT, Lim Y, Lim EC, et al (2008). A118G single nucleotide polymorphism of human mu-opioid receptor gene influences pain perception and patient-controlled intravenous morphine consumption after intrathecal morphine for postcesarean analgesia. Anesthesiology, 109, 520-6. https://doi.org/10.1097/ALN.0b013e318182af21
  37. Sia AT, Sng BL, Lim EC, et al (2010). The influence of ATPbinding cassette sub-family B member -1 (ABCB1) genetic polymorphisms on acute and chronic pain after intrathecal morphine for caesarean section: a prospective cohort study. Int J Obstet Anesth, 19, 254-60. https://doi.org/10.1016/j.ijoa.2010.03.001
  38. Smith K, Hopp M, Mundin G, et al (2012). Low absolute bioavailability of oral naloxone in healthy subjects. J Clin Oncol, 19, 2542-54.
  39. Tan EC, Lim EC, Teo YY, et al (2009). Ethnicity and OPRM variant independently predict pain perception and patientcontrolled analgesia usage for post-operative pain. Mol Pain, 5, 32. https://doi.org/10.1186/1744-8069-5-32
  40. Tan EC, Tan CH, Karupathivan U, et al (2003). Mu opioid receptor gene polymorphisms and heroin dependence in Asian populations. Neuroreport, 14, 569-72. https://doi.org/10.1097/00001756-200303240-00008
  41. Tang K, Wong LP, Lee EJ, et al (2004). Genomic evidence for recent positive selection at the human MDR1 gene locus. Hum Mol Genet, 13, 783-97. https://doi.org/10.1093/hmg/ddh099
  42. Uhl GR, Sora I, Wang Z (1999). The mu opiate receptor as a candidate gene for pain: polymorphisms, variations in expression, nociception, and opiate responses. Proc Natl Acad Sci U S A, 96, 7752-55. https://doi.org/10.1073/pnas.96.14.7752
  43. Wang D, Johnson AD, Papp AC, et al (2005). Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T affects mRNA stability. Pharmacogenet Genomics, 15, 693-704. https://doi.org/10.1097/01.fpc.0000178311.02878.83
  44. Wang D, Quillan JM, Winans K, et al (2001). Single nucleotide polymorphisms in the human mu opioid receptor gene alter basal G protein coupling and calmodulin binding. J Biol Chem, 276, 34624-30. https://doi.org/10.1074/jbc.M104083200
  45. Wang JB, Johnson PS, Persico AM, et al (1994). Human mu opiate receptor: cDNA and genomic clones, pharmacologic characterization and chromosomal assignment. FEBS Lett, 338, 217-22. https://doi.org/10.1016/0014-5793(94)80368-4
  46. Zhou B, Wang J, Yan Z, et al (2012). Liver cancer: effects, safety, and cost-effectiveness of controlled-release oxycodone for pain control after TACE. Radiology, 262, 1014-21. https://doi.org/10.1148/radiol.11110552
  47. Zhou SF, Zhou ZW, Li CG, et al (2007). Identification of drugs that interact with herbs in drug development. Drug Discov Today, 12, 664-73. https://doi.org/10.1016/j.drudis.2007.06.004

Cited by

  1. Maternal Medication Use, Fetal 3435 C>T Polymorphism of the ABCB1 Gene, and Risk of Isolated Septal Defects in a Han Chinese Population vol.35, pp.7, 2014, https://doi.org/10.1007/s00246-014-0906-6
  2. Association of single nucleotide polymorphisms of ABCB1, OPRM1 and COMT with pain perception in cancer patients vol.35, pp.5, 2015, https://doi.org/10.1007/s11596-015-1502-6
  3. The pharmacogenetics of opioid therapy in the management of postpartum pain: a systematic review vol.17, pp.1, 2016, https://doi.org/10.2217/pgs.15.157
  4. Opioid treatment failure in cancer patients: the role of clinical and genetic factors vol.17, pp.13, 2016, https://doi.org/10.2217/pgs-2016-0082
  5. Polymorphisms and Cold Pain Sensitivity Among Healthy Opioid-naive Malay Males vol.17, pp.7, 2017, https://doi.org/10.1111/papr.12546
  6. Pharmacogenomics and Patient Treatment Parameters to Opioid Treatment in Chronic Pain: A Focus on Morphine, Oxycodone, Tramadol, and Fentanyl vol.18, pp.12, 2017, https://doi.org/10.1093/pm/pnw317
  7. OPRM1 c.118A>G Polymorphism and Duration of Morphine Treatment Associated with Morphine Doses and Quality-of-Life in Palliative Cancer Pain Settings vol.18, pp.5, 2017, https://doi.org/10.3390/ijms18040669
  8. Review of Opioid Pharmacogenetics and Considerations for Pain Management vol.37, pp.9, 2017, https://doi.org/10.1002/phar.1986
  9. ABCB1 genotype is associated with fentanyl requirements in critically ill children vol.82, pp.1, 2017, https://doi.org/10.1038/pr.2017.103
  10. genotypes influence pharmacokinetics of morphine in children vol.15, pp.10, 2014, https://doi.org/10.2217/pgs.14.99
  11. Polymorphism of Opioid Receptors μ1 in Highly Hypnotizable Subjects vol.66, pp.1, 2018, https://doi.org/10.1080/00207144.2018.1396128
  12. Complex Role of Hypnotizability in the Cognitive Control of Pain vol.9, pp.1664-1078, 2018, https://doi.org/10.3389/fpsyg.2018.02272
  13. Clinical and pharmacogenetics associated with recovery time from general anesthesia vol.19, pp.14, 2018, https://doi.org/10.2217/pgs-2018-0085
  14. influence on and effectiveness of an individualized treatment plan for prescription opioid use disorder patients vol.1425, pp.1, 2018, https://doi.org/10.1111/nyas.13735