166
Views
3
CrossRef citations to date
0
Altmetric
Review

Potential role of pharmacogenomics testing in the setting of enhanced recovery pathways after surgery

, , , &
Pages 145-154 | Published online: 26 Jul 2019

References

  • Dort JC, Farwell DG, Findlay M, et al. Optimal perioperative care in major head and neck cancer surgery with free flap reconstruction: a consensus review and recommendations from the enhanced recovery after surgery society. JAMA Otolaryngol Head Neck Surg. 2017;143:292–303. doi:10.1001/jamaoto.2016.298127737447
  • Porter ME. What is value in health care? N Engl J Med. 2010;363:2477–2481. doi:10.1056/NEJMp101102421142528
  • Ljungqvist O, Young-Fadok T, Demartines N. The history of enhanced recovery after surgery and the ERAS society. J Laparoendosc Adv Surg Tech. 2017;27:860–862. doi:10.1089/lap.2017.0350
  • Lyon A, Payne CJ, MacKay GJ. Enhanced recovery programme in colorectal surgery: does one size fit all? World J Gastroenterol. 2012;18:5661. doi:10.3748/wjg.v18.i40.566123155304
  • Wilkinson GR. Drug metabolism and variability among patients in drug response. N Engl J Med. 2005;352:2211–2221. doi:10.1056/NEJMra03242415917386
  • CYP2C8 CC, CYP2C19 CD, CYP2E1 CA, CYP3A7 O, OPRK O. A review of the role of genetic testing in pain medicine. Pain Physician. 2014;17:425–445.25247900
  • Carmichael JC, Keller DS, Baldini G, et al. Clinical practice guidelines for enhanced recovery after colon and rectal surgery from the American Society of Colon and Rectal Surgeons and Society of American Gastrointestinal and Endoscopic Surgeons. Dis Colon Rectum. 2017;60:761–784. doi:10.1097/DCR.000000000000088328682962
  • Kalow W, Tang B, Endrenyi L. Hypothesis: comparisons of inter- and intra-individual variations can substitute for twin studies in drug research. Pharmacogenetics. 1998;8:283–290.9731714
  • Jaeger M, Stappers MH, Joosten LA, Gyssens IC, Netea MG. Genetic variation in pattern recognition receptors: functional consequences and susceptibility to infectious disease. Future Microbiol. 2015;10:989–1008. doi:10.2217/fmb.15.3726059622
  • Crews KR, Hicks JK, Pui CH, Relling MV, Evans WE. Pharmacogenomics and individualized medicine: translating science into practice. Clin Pharmacol Ther. 2012;92:467–475.22948889
  • Verbelen M, Weale ME, Lewis CM. Cost-effectiveness of pharmacogenetic-guided treatment: are we there yet? Pharmacogenomics J. 2017;17:395. doi:10.1038/tpj.2017.2128607506
  • Kain ZN, Sevarino F, Pincus S, et al. Attenuation of the preoperative stress response with midazolameffects on postoperative outcomes. Anesthesiology. 2000;93:141–147. doi:10.1097/00000542-200007000-0002410861157
  • Ljungqvist O, Scott M, Fearon KC. Enhanced recovery after surgery: a review. JAMA Surg. 2017;152:292–298. doi:10.1001/jamasurg.2016.495228097305
  • Thompson T, Keogh E, French CC, Davis R. Anxiety sensitivity and pain: generalisability across noxious stimuli. Pain. 2008;134:187–196. doi:10.1016/j.pain.2007.04.01817532572
  • Scott MJ, McEvoy MD, Gordon DB, et al. American Society for Enhanced Recovery (ASER) and perioperative quality initiative (POQI) joint consensus statement on optimal analgesia within an enhanced recovery pathway for colorectal surgery: part 2 – from PACU to the transition home. Perioper Med. 2017;6:7. doi:10.1186/s13741-017-0063-6
  • Kearney M, Jennrich MK, Lyons S, Robinson R, Berger B. Effects of preoperative education on patient outcomes after joint replacement surgery. Orthop Nurs. 2011;30:391–396. doi:10.1097/NOR.0b013e31823710ea22124192
  • Gustafsson U, Scott M, Schwenk W, et al. Guidelines for perioperative care in elective colonic surgery: Enhanced Recovery After Surgery (ERAS®) Society recommendations. Clin Nutr. 2012;31:783–800. doi:10.1016/j.clnu.2012.08.01323099039
  • Gonsalves SG, Dirksen RT, Sangkuhl K, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for the use of potent volatile anesthetic agents and succinylcholine in the context of RYR 1 or CACNA 1S genotypes. Clin Pharmacol Ther. 2019;105:1338–1344. doi:10.1002/cpt.131930499100
  • Cohen M, Sadhasivam S, Vinks AA. Pharmacogenetics in perioperative medicine. Curr Opin Anesthesiol. 2012;25:419–427. doi:10.1097/ACO.0b013e3283556129
  • Palmer SN, Giesecke NM, Body SC, Shernan SK, Fox AA, Collard CD. Pharmacogenetics of anesthetic and analgesic agents. Anesthesiology. 2005;102:663–671. doi:10.1097/00000542-200503000-0002815731608
  • Feldheiser A, Aziz O, Baldini G, et al. Enhanced Recovery After Surgery (ERAS) for gastrointestinal surgery, part 2: consensus statement for anaesthesia practice. Acta Anaesthesiol Scand. 2016;60:289–334. doi:10.1111/aas.1265126514824
  • Beverly A, Kaye AD, Ljungqvist O, Urman RD. Essential elements of multimodal analgesia in enhanced recovery after surgery (ERAS) guidelines. Anesthesiol Clin. 2017;35:e115–e43. doi:10.1016/j.anclin.2017.01.01828526156
  • Hurley RW, Cohen SP, Williams KA, Rowlingson AJ, Wu CL. The analgesic effects of perioperative gabapentin on postoperative pain: a meta-analysis. Reg Anesth Pain Med. 2006;31:237–247. doi:10.1016/j.rapm.2006.01.00516701190
  • Khan JS, Yousuf M, Victor JC, Sharma A, Siddiqui N. An estimation for an appropriate end time for an intraoperative intravenous lidocaine infusion in bowel surgery: a comparative meta-analysis. J Clin Anesth. 2016;28:95–104. doi:10.1016/j.jclinane.2015.07.00726342631
  • Bao-Lin Guo YL, Wei H, Zhen C-X, et al. Effects of systemic magnesium on post-operative analgesia: is the current evidence strong enough. Pain Physician. 2015;18:405–417.26431120
  • Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anesth. 2011;58:911. doi:10.1007/s12630-011-9560-021773855
  • Wang L, Johnston B, Kaushal A, Cheng D, Zhu F, Martin J. Ketamine added to morphine or hydromorphone patient-controlled analgesia for acute postoperative pain in adults: a systematic review and meta-analysis of randomized trials. Can J Anesth. 2016;63:311–325. doi:10.1007/s12630-015-0551-426659198
  • Kapur BM, Lala PK, Shaw JL. Pharmacogenetics of chronic pain management. Clin Biochem. 2014;47:1169–1187. doi:10.1016/j.clinbiochem.2014.05.06524912048
  • Thorell A, MacCormick A, Awad S, et al. Guidelines for perioperative care in bariatric surgery: enhanced recovery after surgery (ERAS) society recommendations. World J Surg. 2016;40:2065–2083. doi:10.1007/s00268-016-3492-326943657
  • Nelson G, Altman A, Nick A, et al. Guidelines for postoperative care in gynecologic/oncology surgery: Enhanced Recovery After Surgery (ERAS®) Society recommendations – part II. Gynecol Oncol. 2016;140:323. doi:10.1016/j.ygyno.2015.12.01926757238
  • Melloul E, Hübner M, Scott M, et al. Guidelines for perioperative care for liver surgery: enhanced recovery after surgery (ERAS) society recommendations. World J Surg. 2016;40:2425–2440. doi:10.1007/s00268-016-3700-127549599
  • Nygren J, Thacker J, Carli F, et al. Guidelines for perioperative care in elective rectal/pelvic surgery: Enhanced Recovery After Surgery (ERAS®) Society recommendations. Clin Nutr. 2012;31:801–816. doi:10.1016/j.clnu.2012.08.01223062720
  • Lassen K, Coolsen MM, Slim K, et al. Guidelines for perioperative care for pancreaticoduodenectomy: Enhanced Recovery After Surgery (ERAS®) Society recommendations. World J Surg. 2013;37:240–258. doi:10.1007/s00268-012-1771-122956014
  • Harirforoosh S, Asghar W, Jamali F. Adverse effects of nonsteroidal antiinflammatory drugs: an update of gastrointestinal, cardiovascular and renal complications. J Pharm Pharm Sci. 2014;16:821–847. doi:10.18433/J3VW2F
  • Bjarnason I. Gastrointestinal safety of NSAIDs and over‐the‐counter analgesics. Int J Clin Pract. 2013;67:37–42. doi:10.1111/ijcp.2012.67.issue-s178
  • Saba R, Kaye AD, Urman RD. Pharmacogenomics in pain management. Anesthesiol Clin. 2017;35:295–304. doi:10.1016/j.anclin.2017.01.01528526150
  • Ko T-M, Wong C-S, Wu J-Y, Chen Y-T. Pharmacogenomics for personalized pain medicine. Acta Anaesthesiol Taiwan. 2016;54:24–30. doi:10.1016/j.aat.2016.02.00126976339
  • Martínez C, García‐Martín E, Blanco G, Gamito FJ, Ladero JM, Agúndez JA. The effect of the cytochrome P450 CYP2C8 polymorphism on the disposition of (R)‐ibuprofen enantiomer in healthy subjects. Br J Clin Pharmacol. 2005;59:62–68. doi:10.1111/j.1365-2125.2004.02183.x15606441
  • Kumar S, Samuel K, Subramanian R, et al. Extrapolation of diclofenac clearance from in vitro microsomal metabolism data: role of acyl glucuronidation and sequential oxidative metabolism of the acyl glucuronide. J Pharmacol Exp Ther. 2002;303:969–978. doi:10.1124/jpet.102.03899212438516
  • Blanco G, Martínez C, Ladero JM, et al. Interaction of CYP2C8 and CYP2C9 genotypes modifies the risk for nonsteroidal anti-inflammatory drugs-related acute gastrointestinal bleeding. Pharmacogenet Genomics. 2008;18:37–43. doi:10.1097/FPC.0b013e3282f305a918216720
  • Graham GG, Davies MJ, Day RO, Mohamudally A, Scott KF. The modern pharmacology of paracetamol: therapeutic actions, mechanism of action, metabolism, toxicity and recent pharmacological findings. Inflammopharmacology. 2013;21:201–232. doi:10.1007/s10787-013-0172-x23719833
  • Ong CK, Seymour RA, Lirk P, Merry AF. Combining paracetamol (acetaminophen) with nonsteroidal antiinflammatory drugs: a qualitative systematic review of analgesic efficacy for acute postoperative pain. Anesth Analg. 2010;110:1170–1179.20142348
  • Ziemann-Gimmel P, Hensel P, Koppman J, Marema R. Multimodal analgesia reduces narcotic requirements and antiemetic rescue medication in laparoscopic Roux-en-Y gastric bypass surgery. Surg Obes Relat Dis. 2013;9:975–980. doi:10.1016/j.soard.2013.02.00323499469
  • Maund E, McDaid C, Rice S, Wright K, Jenkins B, Woolacott N. Paracetamol and selective and non-selective non-steroidal anti-inflammatory drugs for the reduction in morphine-related side-effects after major surgery: a systematic review. Br J Anaesth. 2011;106:292–297. doi:10.1093/bja/aeq40621285082
  • Remy C, Marret E, Bonnet F. Effects of acetaminophen on morphine side-effects and consumption after major surgery: meta-analysis of randomized controlled trials. Br J Anaesth. 2005;94:505–513. doi:10.1093/bja/aei08515681586
  • Mazaleuskaya LL, Sangkuhl K, Thorn CF, FitzGerald GA, Altman RB, Klein TE. PharmGKB summary: pathways of acetaminophen metabolism at the therapeutic versus toxic doses. Pharmacogenet Genomics. 2015;25:416. doi:10.1097/FPC.000000000000015026049587
  • Nakagawa T, Mure T, Yusoff S, et al. Acetaminophen administration in a patient with Gilbert’s syndrome. Pediatr Int. 2012;54:934–936. doi:10.1111/j.1442-200X.2012.03602.x23279026
  • Tankanitlert J, Morales NP, Howard TA, et al. Effects of combined UDP-glucuronosyltransferase (UGT) 1A1* 28 and 1A6* 2 on paracetamol pharmacokinetics in β-thalassemia/HbE. Pharmacology. 2007;79:97–103. doi:10.1159/00009790817164591
  • Buchard A, Eefsen M, Semb S, et al. The role of the glutathione S-transferase genes GSTT1, GSTM1, and GSTP1 in acetaminophen-poisoned patients. Clin Toxicol. 2012;50:27–33. doi:10.3109/15563650.2011.639713
  • Nagar S, Walther S, Blanchard RL. Sulfotransferase (SULT) 1A1 polymorphic variants* 1,* 2, and* 3 are associated with altered enzymatic activity, cellular phenotype, and protein degradation. Mol Pharmacol. 2006;69:2084–2092. doi:10.1124/mol.105.01924016517757
  • Ueshima Y, Tsutsumi M, Takase S, Matsuda Y, Kawahara H. Acetaminophen metabolism in patients with different cytochrome P‐4502E1 genotypes. Alcoholism. 1996;20:25a–8a. doi:10.1111/acer.1996.20.issue-s18651456
  • Freytsis M, Wang X, Peter I, et al.; Group ALFS. The UDP-glucuronosyltransferase (UGT) 1A polymorphism c. 2042C> G (rs8330) is associated with increased human liver acetaminophen glucuronidation, increased UGT1A exon 5a/5b splice variant mRNA ratio, and decreased risk of unintentional acetaminophen-induced acute liver failure. J Pharmacol Exp Ther. 2013;345:297–307. doi:10.1124/jpet.112.20201023408116
  • Navarro SL, Chen Y, Li L, et al. UGT1A6 and UGT2B15 polymorphisms and acetaminophen conjugation in response to a randomized, controlled diet of select fruits and vegetables. Drug Metab Dispos. 2011;39:1650–1657. doi:10.1124/dmd.111.03914921666065
  • Siddiqui KM, Khan FA. Effect of preinduction low-dose ketamine bolus on intra operative and immediate postoperative analgesia requirement in day care surgery: a randomized controlled trial. Saudi J Anaesth. 2015;9:422. doi:10.4103/1658-354X.15946826543461
  • Temple-Oberle C, Shea-Budgell MA, Tan M, et al. Consensus review of optimal perioperative care in breast reconstruction: enhanced recovery after surgery (ERAS) society recommendations. Plast Reconstr Surg. 2017;139:1056e–71e. doi:10.1097/PRS.0000000000003242
  • Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms toN-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–858. doi:10.1124/dmd.30.7.85312065445
  • Aroke EN, Dungan JR. Pharmacogenetics of anesthesia: an integrative review. Nurs Res. 2016;65:318–330. doi:10.1097/NNR.000000000000016427362518
  • Li Y, Jackson KA, Slon B, et al. CYP2B6* 6 allele and age substantially reduce steady‐state ketamine clearance in chronic pain patients: impact on adverse effects. Br J Clin Pharmacol. 2015;80:276–284. doi:10.1111/bcp.1261425702819
  • McEvoy MD, Scott MJ, Gordon DB, et al. American Society for Enhanced Recovery (ASER) and Perioperative Quality Initiative (POQI) joint consensus statement on optimal analgesia within an enhanced recovery pathway for colorectal surgery: part 1 – from the preoperative period to PACU. Perioper Med. 2017;6:8. doi:10.1186/s13741-017-0064-5
  • Garimella V, Cellini C. Postoperative pain control. Clin Colon Rectal Surg. 2013;26:191–196. doi:10.1055/s-0033-135113824436674
  • Grant MC, Sommer PM, He C, et al. Preserved analgesia with reduction in opioids through the use of an acute pain protocol in enhanced recovery after surgery for open hepatectomy. Reg Anesth Pain Med. 2017;42:451–457. doi:10.1097/AAP.000000000000061528525409
  • Williams D, Patel A, Howard R. Pharmacogenetics of codeine metabolism in an urban population of children and its implications for analgesic reliability. Br J Anaesth. 2002;89:839–845. doi:10.1093/bja/aef28412453926
  • Smith DM, Weitzel KW, Cavallari LH, Elsey AR, Schmidt SO. Clinical application of pharmacogenetics in pain management. Per Med. 2018;15:117–126. doi:10.2217/pme-2017-003229714124
  • Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3:229–243. doi:10.1517/14622416.3.2.22911972444
  • Punjasawadwong Y, Phongchiewboon A, Bunchungmongkol N. Bispectral index for improving anaesthetic delivery and postoperative recovery. Cochrane Database Syst Rev. 2014. doi:10.1002/14651858.CD003843.pub3
  • Mikstacki A, Skrzypczak-Zielinska M, Tamowicz B, Zakerska-Banaszak O, Szalata M, Slomski R. The impact of genetic factors on response to anaesthetics. Adv Med Sci. 2013;58:9–14. doi:10.2478/v10039-012-0065-z23640947
  • Mastrogianni O, Gbandi E, Orphanidis A, et al. Association of the CYP2B6 c. 516G> T polymorphism with high blood propofol concentrations in women from northern Greece. Drug Metab Pharmacokinet. 2014;29(2):215–8.
  • Eugene AR. CYP2B6 genotype guided dosing of propofol anesthesia in the elderly based on nonparametric population pharmacokinetic modeling and simulations. Int J Clin Pharmacol Toxicol. 2017;6:242.28154789
  • Mourão AL, de Abreu FG, Fiegenbaum M. Impact of the cytochrome P450 2B6 (CYP2B6) gene polymorphism c. 516G> T (rs3745274) on propofol dose variability. Eur J Drug Metab Pharmacokinet. 2016;41:511–515. doi:10.1007/s13318-015-0289-y26141406
  • Iohom G, Chonghaile MN, O’brien J, Cunningham A, Fitzgerald D, Shields D. An investigation of potential genetic determinants of propofol requirements and recovery from anaesthesia. Eur J Anaesthesiol. 2007;24:912–919. doi:10.1017/S026502150700047617555608
  • Zhong Q, Chen X, Zhao Y, Liu R, Yao S. Association of polymorphisms in pharmacogenetic candidate genes with propofol susceptibility. Sci Rep. 2017;7:3343. doi:10.1038/s41598-017-03229-328611364
  • Garcia DF, Oliveira TG, Molfetta GA, et al. Biochemical and genetic analysis of butyrylcholinesterase (BChE) in a family, due to prolonged neuromuscular blockade after the use of succinylcholine. Genet Mol Biol. 2011;34:40–44. doi:10.1590/S1415-4757201100010000821637541
  • Delacour H, Lushchekina S, Mabboux I, et al. Characterization of a novel BCHE “silent” allele: point mutation (p. Val204Asp) causes loss of activity and prolonged apnea with suxamethonium. PLoS One. 2014;9:e101552. doi:10.1371/journal.pone.010155225054547
  • Gätke MR, Bundgaard JR, Viby-Mogensen J. Two novel mutations in the BCHE gene in patients with prolonged duration of action of mivacurium or succinylcholine during anaesthesia. Pharmacogenet Genomics. 2007;17:995–999. doi:10.1097/FPC.0b013e3282f0664618075469
  • Jensen F, Viby‐Mogensen J. Plasma cholinesterase and abnormal reaction to succinylcholine: twenty years’ experience with the Danish Cholinesterase Research Unit. Acta Anaesthesiol Scand. 1995;39:150–156.7793179
  • Alvarellos ML, McDonagh EM, Patel S, McLeod HL, Altman RB, Klein TE. PharmGKB summary: succinylcholine pathway, pharmacokinetics/pharmacodynamics. Pharmacogenet Genomics. 2015;25:622. doi:10.1097/FPC.000000000000017026398623
  • Mei Y, Wang SY, Li Y, et al. Role of SLCO1B1, ABCB1, and CHRNA1 gene polymorphisms on the efficacy of rocuronium in Chinese patients. J Clin Pharmacol. 2015;55:261–268. doi:10.1002/jcph.40525279974
  • Rosenberg H, Pollock N, Schiemann A, Bulger T, Stowell K. Malignant hyperthermia: a review. Orphanet J Rare Dis. 2015;10:93. doi:10.1186/s13023-015-0310-126238698
  • Habib AS, Gan TJ. Postoperative nausea and vomiting: then & now. Anesth Analg. 2012;115(3):493–495..
  • Gan TJ, Diemunsch P, Habib AS, et al. Consensus guidelines for the management of postoperative nausea and vomiting. Anesth Analg. 2014;118:85–113. doi:10.1213/ANE.000000000000000224356162
  • Kim M-S, Lee J-R, Choi E-M, Kim EH, Choi SH. Association of 5-HT3B receptor gene polymorphisms with the efficacy of ondansetron for postoperative nausea and vomiting. Yonsei Med J. 2015;56:1415–1420. doi:10.3349/ymj.2015.56.5.141526256989
  • Nakagawa M, Kuri M, Kambara N, et al. Dopamine D2 receptor Taq IA polymorphism is associated with postoperative nausea and vomiting. J Anesth. 2008;22:397–403. doi:10.1007/s00540-008-0661-z19011779
  • Kolesnikov Y, Gabovits B, Levin A, Voiko E, Veske A. Combined catechol-O-methyltransferase and μ-opioid receptor gene polymorphisms affect morphine postoperative analgesia and central side effects. Anesth Analg. 2011;112:448–453. doi:10.1213/ANE.0b013e318202cc8d21127283
  • Sugino S, Hayase T, Higuchi M, et al. Association of μ-opioid receptor gene (OPRM1) haplotypes with postoperative nausea and vomiting. Exp Brain Res. 2014;232:2627–2635. doi:10.1007/s00221-014-3987-924858579
  • Iyer S, Saunders WB, Stemkowski S. Economic burden of postoperative ileus associated with colectomy in the United States. J Manag Care Pharm. 2009;15:485–494. doi:10.18553/jmcp.2009.15.6.48519610681
  • Short V, Herbert G, Perry R, et al. Chewing gum for postoperative recovery of gastrointestinal function. Cochrane Database Syst Rev. 2015. doi:10.1002/14651858.CD006506.pub3
  • Kranke P, Jokinen J, Pace NL, et al. Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery. Cochrane Database Syst Rev. 2015. doi:10.1002/14651858.CD009642.pub2
  • Desta Z, Soukhova N, Mahal SK, Flockhart DA. Interaction of cisapride with the human cytochrome P450 system: metabolism and inhibition studies. Drug Metab Dispos. 2000;28:789–800.10859153
  • Quigley EM. Cisapride: what can we learn from the rise and fall of a prokinetic? J Dig Dis. 2011;12:147–156. doi:10.1111/j.1751-2980.2011.00491.x21615867
  • Zanger UM, Fischer J, Raimundo S, et al. Comprehensive analysis of the genetic factors determining expression and function of hepatic CYP2D6. Pharmacogenet Genomics. 2001;11:573–585. doi:10.1097/00008571-200110000-00004
  • Ljungqvist O. ERAS – enhanced recovery after surgery: moving evidence‐based perioperative care to practice. J Parenter Enteral Nutr. 2014;38:559–566. doi:10.1177/0148607114523451
  • Van der Padt A, van Schaik R, Sonneveld P. Acute dystonic reaction to metoclopramide in patients carrying homozygous cytochrome P450 2D6 genetic polymorphisms. Neth J Med. 2006;64:160–162.16702617
  • Yasny JS, White J. Environmental implications of anesthetic gases. Anesth Prog. 2012;59:154–158. doi:10.2344/0003-3006-59.4.15423241038
  • Kim JH. Mechanism of emergence agitation induced by sevoflurane anesthesia. Korean J Anesthesiol. 2011;60:73–74. doi:10.4097/kjae.2011.60.2.7321390159
  • Nair AS. Pharmacogenomics of inhalational anesthetic agents. Med Gas Res. 2019;9:52–53. doi:10.4103/2045-9912.25464130950419
  • Liem EB, Lin CM, Suleman MI, et al. Anesthetic requirement is increased in redheads. Anesthesiology. 2004;101:279–283. doi:10.1097/00000542-200408000-0000615277908
  • DRH’s Division of Industry Communication and Education (DICE). The FDA Warns against the Use of Many Genetic Tests with Unapproved Claims to Predict Patient Response to Specific Medications: FDA Safety Communication. Silver Spring, MD; 2018.
  • Moyer AM, Vitek CRR, Giri J, Caraballo PJ. Challenges in ordering and interpreting pharmacogenomic tests in clinical practice. Am J Med. 2017;130:1342–1344. doi:10.1016/j.amjmed.2017.07.01228757317
  • Kehlet H. Enhanced Recovery After Surgery (ERAS): good for now, but what about the future? Can J Anesth. 2015;62:99–104. doi:10.1007/s12630-014-0261-325391731
  • Saba R, Kaye AD, Urman RD. Pharmacogenomics in anesthesia. Anesthesiol Clin. 2017;35:285–294. doi:10.1016/j.anclin.2017.01.01428526149