Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-04-30T22:21:08.539Z Has data issue: false hasContentIssue false

Chapter 5 - Genomics and personalized medicine in postoperative nausea and vomiting management

Published online by Cambridge University Press:  05 March 2016

Tong Joo Gan
Affiliation:
Duke University Medical Center, Durham
Ashraf S. Habib
Affiliation:
Duke University Medical Center, Durham
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Postoperative Nausea and Vomiting
A Practical Guide
, pp. 41 - 52
Publisher: Cambridge University Press
Print publication year: 2016

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

de Leon, J. Pharmacogenomics: the promise of personalized medicine for CNS disorders. Neuropsychopharmacology 2009; 34:159–72.CrossRefGoogle ScholarPubMed
Dawood, S, Leyland-Jones, B. Pharmacology and pharmacogenetics of chemotherapeutic agents. Cancer Invest 2009; 27:482–8.CrossRefGoogle ScholarPubMed
Janicki, PK, Sugino, S. Genetic factors associated with pharmacotherapy and background sensitivity to postoperative and chemotherapy‑induced nausea and vomiting. Exp Brain Res 2014; 232(8):2613–25.CrossRefGoogle ScholarPubMed
Raeder, J. History of postoperative nausea and vomiting. Int Anesthesiol Clin 2003; 41:112.CrossRefGoogle ScholarPubMed
Palazzo, M, Evans, R. Logistic regression analysis of fixed patient factors for postoperative sickness: a model for risk assessment. Br J Anaesth 1993; 70:135–40.CrossRefGoogle Scholar
Cohen, MM, Duncan, PG, DeBoer, DP, et al. The postoperative interview: assessing risk factors for nausea and vomiting. Anesth Analg 1994; 78:716.CrossRefGoogle ScholarPubMed
Sinclair, DR, Chung, F, Mezei, G. Can postoperative nausea and vomiting be predicted? Anesthesiology 1999; 91:109–18.CrossRefGoogle ScholarPubMed
Koivuranta, M, Laara, E, Snare, L, et al. A survey of postoperative nausea and vomiting. Anaesthesia 1997; 52:443449.CrossRefGoogle ScholarPubMed
Eberhart, LH, Hogel, J, Seeling, W, et al. Evaluation of three risk scores to predict postoperative nausea and vomiting. Acta Anaesthesiol Scand 2000; 44:480–8.CrossRefGoogle ScholarPubMed
Apfel, CC, Kranke, P, Eberhart, LH, et al. Comparison of predictive models for postoperative nausea and vomiting. Br J Anaesth 2002; 88:234–40.CrossRefGoogle ScholarPubMed
Gan, TJ. Risk factors for postoperative nausea and vomiting. Anesth Analg 2006; 102:1884–98.CrossRefGoogle ScholarPubMed
Ikeda, K, Ide, S, Han, W, et al. How individual sensitivity to opiates can be predicted by gene analyses. Trends Pharmacol Sci 2005; 26:311–7.CrossRefGoogle ScholarPubMed
Manolio, TA, Brooks, LD, Collins, FS. A HapMap harvest of insights into the genetics of common disease. J Clin Invest 2008; 118:1590–605.CrossRefGoogle ScholarPubMed
Feero, WG, Guttmacher, AE, Collins, FS. Genomic medicine – an updated primer. N Engl J Med 2010; 362:2001–11.CrossRefGoogle ScholarPubMed
Gan, TJ, Meyer, T, Apfel, CC, et al. Consensus guidelines for managing postoperative nausea and vomiting. Anesth Analg 2003; 97:6271.CrossRefGoogle ScholarPubMed
Habib, AS, Gan, TJ. Evidence-based management of postoperative nausea and vomiting: a review. Can J Anesth 2004; 51:326–41.CrossRefGoogle ScholarPubMed
Cohen, M, Sadhasivam, S, Vinks, AA. Pharmacogenetics in perioperative medicine. Curr Opin Anesthesiol 2012; 25:419–27.CrossRefGoogle ScholarPubMed
Ho, KY, Gan, TJ. Pharmacology, pharmacogenetics, and clinical efficacy of 5-hydroxytryptamine type 3 receptor antagonists for postoperative nausea and vomiting. Curr Opin Anaesthesiol 2006; 19:606–11.CrossRefGoogle ScholarPubMed
Andrews, PL. Physiology of nausea and vomiting. Br J Anaesth 1992; 69(1):219.CrossRefGoogle ScholarPubMed
Miller, AD, Leslie, RA. The area postrema and vomiting. Front Neuroendocrinol 1994; 15:301–20.CrossRefGoogle ScholarPubMed
Miyake, A, Mochizuki, S, Takemoto, Y, et al. Molecular cloning of human 5-hydroxytryptamine3 receptor: heterogeneity in distribution and function among species. Mol Pharmacol 1995; 48:407–16.Google ScholarPubMed
Belelli, D, Balcarek, JM, Hope, AG, et al. Cloning and functional expression of a human 5-hydroxytryptamine type 3AS receptor subunit. Mol Pharmacol 1995; 48:1054–62.Google ScholarPubMed
Davies, PA, Pistis, M, Hanna, MC, et al. The 5-HT3B subunit is a major determinant of serotonin-receptor function. Nature 1999; 397:359–63.CrossRefGoogle Scholar
Dubin, AE, Huvar, R, D’Andrea, MR, et al. The pharmacological and functional characteristics of the serotonin 5-HT(3A) receptor are specifically modified by a 5-HT(3B) receptor subunit. J Biol Chem 1999; 274:30799–810.CrossRefGoogle ScholarPubMed
Bruss, M, Barann, M, Hayer-Zillgen, M, et al. Modified 5-HT3A receptor function by co-expression of alternatively spliced human 5-HT3A receptor isoforms. Naunyn Schmiedebergs Arch Pharmacol 2000; 362:392401.CrossRefGoogle ScholarPubMed
Gothert, M, Propping, P, Bonisch, H, et al. Genetic variation in human 5-HT receptors: potential pathogenetic and pharmacological role. Ann NY Acad Sci 1998; 861:2630.CrossRefGoogle ScholarPubMed
Bruss, M, Eucker, T, Gothert, M, et al. Exon–intron organization of the human 5-HT3A receptor gene. Neuropharmacology 2000; 39:308–15.CrossRefGoogle ScholarPubMed
Kaiser, R, Tremblay, PB, Sezer, O, et al. Investigation of the association between 5-HT3A receptor gene polymorphisms and efficiency of antiemetic treatment with 5-HT3 receptor antagonists. Pharmacogenetics 2004; 14:271–8.CrossRefGoogle Scholar
Tremblay, PB, Kaiser, R, Sezer, O, et al. Variations in the 5-hydroxytryptamine type 3B receptor gene as predictors of the efficacy of antiemetic treatment in cancer patients. J Clin Oncol 2003; 21:2147–55.CrossRefGoogle ScholarPubMed
Rueffert, H, Thieme, V, Wallenborn, J, et al. Do variations in the 5-HT3A and 5-HT3B serotonin receptor genes (HTR3A and HTR3B) influence the occurrence of postoperative vomiting? Anesth Analg 2009; 109:1442–7.CrossRefGoogle ScholarPubMed
Sugai, T, Suzuki, Y, Sawamura, K, et al. The effect of 5-hydroxytryptamine 3A and 3B receptor genes on nausea induced by paroxetine. Pharmacogenomics J 2006; 6:351–6.CrossRefGoogle ScholarPubMed
Ma, XX, Chen, QX, Wu, SJ, et al. Polymorphisms of the HTR3B gene are associated with post-surgery emesis in a Chinese Han population. J Clin Pharm Ther 2013; 38:150–5.CrossRefGoogle Scholar
Fischer, V, Vickers, AE, Heitz, F, et al. The polymorphic cytochrome P-450 2D6 is involved in the metabolism of both 5-hydroxytryptamine antagonists, tropisetron and ondansetron. Drug Metab Dispos 1994; 22:269–74.Google Scholar
Dixon, CM, Colthup, PV, Serabjit-Singh, CJ, et al. Multiple forms of cytochrome P450 are involved in the metabolism of ondansetron in humans. Drug Metab Dispos 1995; 23:1225–30.Google Scholar
Bloomer, JC, Baldwin, SJ, Smith, GJ, et al. Characterization of the cytochrome P450 enzymes involved in the in vitro metabolism of granisetron. Br J Clin Pharmacol 1994; 38:557–66.CrossRefGoogle ScholarPubMed
Candiotti, KA. Anesthesia and pharmacogenomics: not ready for prime time. Anesth Analg 2009; 109:1377–8.CrossRefGoogle Scholar
Candiotti, KA, Birnbach, DJ, Lubarsky, DA, et al. The impact of pharmacogenomics on postoperative nausea and vomiting: do CYP2D6 allele copy number and polymorphisms affect the success or failure of ondansetron prophylaxis? Anesthesiology 2005; 102:543–9.CrossRefGoogle ScholarPubMed
Kaiser, R, Sezer, O, Papies, A, et al. Patient-tailored antiemetic treatment with 5-hydroxytryptamine type 3 receptor antagonists according to cytochrome P-450 genotypes. J Clin Oncol 2002; 20,12:2805–11.CrossRefGoogle ScholarPubMed
McKenzie, R, Kovac, A, O’Connor, T, et al. Comparison of ondansetron versus placebo to prevent postoperative nausea and vomiting in women undergoing ambulatory gynecologic surgery. Anesthesiology 1993; 78:21–8.CrossRefGoogle ScholarPubMed
Janicki, PK, Schuler, HG, Jarzembowski, TM, et al. Prevention of postoperative nausea and vomiting with granisetron and dolasetron in relation to CYP2D6 genotype. Anesth Analg 2006; 102:1127–33.CrossRefGoogle ScholarPubMed
Janicki, PK, Schuler, G, Francis, D, et al. A genetic association study of the functional A118G polymorphism of the human mu-opioid receptor gene in patients with acute and chronic pain. Anesth Analg 2006; 103:1011–7.CrossRefGoogle ScholarPubMed
Stamer, U, Rauers, N, Eun-Hae, L, et al. Ondansetron for the treatment of opioid induced nausea and vomiting: impact of cytochrome polymorphisms. Eur J Pain 2009; 13:S193/667.CrossRefGoogle Scholar
Wesmiller, SW, Henker, RA, Sereika, SM, et al. The association of CYP2D6 genotype and postoperative nausea and vomiting in orthopedic trauma patients. Biol Res Nurs 2013; 15:382–9.CrossRefGoogle ScholarPubMed
Wesmiller, SW, Bender, CM, Sereika, SM, et al. Association between serotonin transport polymorphisms and postdischarge nausea and vomiting in women following breast cancer surgery. Oncol Nurs Forum 2014; 41(2):195202.CrossRefGoogle Scholar
Sweeney, BP. Why does smoking protect against PONV? Br J Anaesth 2002; 89:810–3.Google ScholarPubMed
Janicki, PK. Cytochrome P450 2D6 metabolism and 5-hydroxytryptamine type 3 receptor antagonists for postoperative nausea and vomiting. Med Sci Monit 2005; 11:RA322328.Google ScholarPubMed
Sanger, GJ, Andrews, PL. Treatment of nausea and vomiting: gaps in our knowledge. Auton Neurosci 2006; 129:316.CrossRefGoogle ScholarPubMed
Pergolizzi, JV Jr, Philip, BK, Leslie, JB, et al. Perspectives on transdermal scopolamine for the treatment of postoperative nausea and vomiting. J Clin Anesth 2012; 24:334–5.CrossRefGoogle ScholarPubMed
Janicki, PK, Vealey, R, Liu, J, et al. Genome-wide association study using pooled DNA to identify candidate markers mediating susceptibility to postoperative nausea and vomiting. Anesthesiology 2011; 115:5464.CrossRefGoogle ScholarPubMed
Hayashi, H, Sugino Hayase, T, Nawa, Y, et al. CHRM3 gene polymorphysm is associated with postoperative nausea and vomiting in the Japanese population. Annual Meeting of the American Society of Anesthesiology Online Abstracts 2012; A1006. http://www.asaabstracts.com/ (accessed December 23, 2015).Google Scholar
Hyde, TM, Knable, MB, Murray, AM. Distribution of dopamine D1–D4 receptor subtypes in human dorsal vagal complex. Synapse 1996; 24:224–32.3.0.CO;2-G>CrossRefGoogle ScholarPubMed
Nakagawa, M, Kuri, M, Kambara, N, et al. Dopamine D2 receptor Taq IA polymorphism is associated with postoperative nausea and vomiting. J Anesth 2008; 22:397403.CrossRefGoogle ScholarPubMed
Chou, W, Yang, L, Lu, H, et al. Association of μ-opioid receptor gene polymorphism (AI18G) with variations in morphine consumption for analgesia after total knee arthroplasty. Acta Anaestesiol Scand 2006; 50:787–92.CrossRefGoogle ScholarPubMed
Sia, A, Lim, Y, Lim, E, et al. A118G single nucleotide polymorphism of human mu-opioid receptor gene influences pain perception and patient-controlled intravenous morphine consumption after intrathecal morphine for post cesarean analgesia. Anesthesiology 2008; 109:520–6.CrossRefGoogle Scholar
Zhang, W, Yuan, JJ, Kan, QC, et al. Study of the OPRM1 A118G genetic polymorphism associated with postoperative nausea and vomiting induced by fentanyl intravenous analgesia. Minerva Anestesiol 2011; 77:33–9.Google ScholarPubMed
Chen, LK, Chen, SS, Huang, CH, et al. Polymorphism of μ-opioid receptor gene (OPRM1:c.118A>G) might not protect against or enhance morphine-induced nausea or vomiting. Pain Res Treat 2013; 259306. http://dx.doi.org/10.1155/2013/259306G)+might+not+protect+against+or+enhance+morphine-induced+nausea+or+vomiting.+Pain+Res+Treat+2013;+259306.+http://dx.doi.org/10.1155/2013/259306>Google Scholar
Song, Z, Du, B, Wang, K, et al. Effects of OPRM1 A118G polymorphism on epidural analgesia with fentanyl during labor: a meta-analysis. Genet Test Mol Biomarkers 2013; 17:743–9.CrossRefGoogle ScholarPubMed
Sugino, S, Hayase, T, Higuchi, M, et al. Associations of μ-opioid receptor gene (OPRM1) haplotypes with postoperative nausea and vomiting (PONV) during intravenous patient-controlled analgesia. Biology and Control of Nausea and Vomiting Online Abstracts 2013; 60. www.researchgate.net/publication/262609924_Association_of_m-opioid_receptor_gene_OPRM1_haplotypes_with_postoperative_nausea_and_vomiting (accessed December 18, 2015).Google Scholar
Marzolini, C, Paus, E, Buclin, T, et al. Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance. Clin Pharmacol Ther 2004; 75:1333.CrossRefGoogle ScholarPubMed
Choi, EM, Lee, MG, Lee, SH, et al. Association of ABCB1 polymorphisms with the efficacy of ondansetron for postoperative nausea and vomiting. Anaesthesia 2010; 65:9961000.CrossRefGoogle ScholarPubMed
Coulbault, L, Beaussier, M, Verstuyft, C, et al. Environmental and genetic factors associated with morphine response in the postoperative period. Clin Pharmacol Ther 2006; 79:316–24.CrossRefGoogle ScholarPubMed
Zwisler, S, Enggaard, T, Noehr-Jensen, L, et al. The antinociceptive effect and adverse drug reactions of oxycodone in human experimental pain in relation to genetic variations in the OPRM1 and ABCB1 genes. Fundam Clin Pharmacol 2009; 10:18.Google Scholar
Perwitasari, DA, Wessels, JA, van der Straaten, RJ, et al. Association of ABCB1, 5-HT3B receptor and CYP2D6 genetic polymorphisms with ondansetron and metoclopramide antiemetic response in Indonesian cancer patients treated with highly emetogenic chemotherapy. Jpn J Clin Oncol 2011; 41(10):1168–76.CrossRefGoogle ScholarPubMed
Babaoglu, MO, Bayar, B, Aynacioglu, AS, et al. Association of the ABCB1 3435C>T polymorphism with antiemetic efficacy of 5-hydroxytryptamine type 3 antagonists. Clin Pharmacol Ther 2005; 78:619–26.CrossRefGoogle Scholar
Andersen, S, Skorpen, F. Variation in the COMT gene: implications for pain perception and pain treatment. Pharmacogenomics 2009; 10:669–84.CrossRefGoogle ScholarPubMed
Park, JW, Lee, KS, Kim, JS, et al. Genetic contribution of catechol-O-methyltransferase polymorphism in patients with migraine without aura. J Clin Neurol 2007; 3:2430.CrossRefGoogle ScholarPubMed
Kolesnikov, Y, Gabovits, B, Levin, A, et al. Combined catechol-O-methyltransferase and mu-opioid receptor gene polymorphisms affect morphine postoperative analgesia and central side effects. Anesth Analg 2011; 112:448–53.CrossRefGoogle ScholarPubMed
Steinhoff, MS, von Mentzer, B, Geppetti, P, et al. Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease. Physiol Rev 2014; 94:265301.CrossRefGoogle ScholarPubMed
Hargreaves, R, Ferreira, JC, Hughes, D, et al. Development of aprepitant, the first neurokinin-1 receptor antagonist for the prevention of chemotherapy-induced nausea and vomiting. Ann NY Acad Sci 2011; 1222:4048.CrossRefGoogle ScholarPubMed
Gan, T, Gu, J, Singla, N, et al. Rolapitant for the prevention of postoperative nausea and vomiting: a prospective, double-blinded, placebo-controlled randomized trial. Anesthes Analg 2011; 112: 804–12.CrossRefGoogle ScholarPubMed
Hayase, T, Sugino, S, Tachibana, S, et al. Haplotypes in human TACR1 gene are associated with postoperative nausea and vomiting. Annual Meeting of the American Society of Anesthesiology Online Abstracts 2012. http://www.asaabstracts.com/A1003 (accessed December 23, 2015).Google Scholar
Dos Santos, LV, Souza, FH, Brunetto, AT, et al. Neurokinin-1 receptor antagonists for chemotherapy-induced nausea and vomiting: a systematic review. J Natl Cancer Inst 2012; 104:1280–92.CrossRefGoogle ScholarPubMed
Walter, C, Lotsch, J. Meta-analysis of the relevance of the OPRM1 118A>G genetic variant for pain treatment. Pain 2009; 146:270275.CrossRefGoogle Scholar
Wong, CA, McCarthy, RJ, Blouin, J, et al. Observational study of the effect of muopioid receptor genetic polymorphism on intrathecal opioid labor analgesia and postcesarean delivery analgesia. Int J Obstetr Anesth 2010; 19:246–53.Google Scholar
Jannetto, PJ, Bratanow, NC. Pain management in the 21st century: utilization of pharmacogenomics and therapeutic drug monitoring. Expert Opin Drug Metab Toxicol 2011; 7:745752.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×