Pediatric Acute and Surgical Pain Management: Recent Advances and Future Perspectives

Pediatric Acute and Surgical Pain Management: Recent Advances and Future Perspectives Vidya Chidambaran, MD Senthilkumar Sadhasivam, MD, MPH Departme...
2 downloads 1 Views 154KB Size
Pediatric Acute and Surgical Pain Management: Recent Advances and Future Perspectives

Vidya Chidambaran, MD Senthilkumar Sadhasivam, MD, MPH Departments of Anesthesia and Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio



Objectives

1. To review limitations of currently used analgesic modalities, surgeryspecific analgesia protocols, and available alternative drugs in children. 2. To discuss the effects of developmental pharmacology and neurobiology on pain management. 3. To introduce fundamentals of pharmacogenomics and personalized analgesia.



Introduction

Children suffer pain similar to adults. Besides developmental differences, they exhibit exaggerated emotional and behavioral components of pain including higher anxiety and altered coping mechanisms compared with adults.1 Although the practice of pediatric pain management has made great progress in the last decade with the development and validation of pain assessment tools specific to children,2 acute pain remains underrecognized and undertreated even in hospitalized children.3 Parental behavior and misconceptions regarding pain medications, including fear of opioid addiction, fear of serious adverse drug effects, difficulties in assessment of pain, and lack of advocacy constitute reasons for this suboptimal alleviation of pain among this population. A national survey of 252 institutions found that REPRINTS: VIDYA CHIDAMBARAN, MD, CINCINNATI CHILDREN’S HOSPITAL MEDICAL CENTER, 3333 BURNET AVE, MLC 2001, CINCINNATI, OH 45229. E-MAIL: [email protected] INTERNATIONAL ANESTHESIOLOGY CLINICS Volume 50, Number 4, 66–82 r 2012, Lippincott Williams & Wilkins

66 | www.anesthesiaclinics.com

Pediatric Surgical Pain Management



67

only 51% had a pediatric pain management service, the practices and supervision of which were influenced by patient characteristics, institutional demographics, and published national guidelines.4 At our institution, a pain management service initiated in the early 1990s has over the years matured into full-fledged divisions encompassing specific inpatient and outpatient services for perioperative pain, medical pain, chronic pain, and palliative care in addition to education of pain in the core pediatric anesthesia fellowship as well as advanced fellowships in chronic medical pain and palliative care. Our perioperative pain service provides 24/7 coverage with an advanced practice nurse and 1 rotating pediatric anesthesia fellow during the day and 1 nurse trained in pain and a cross-covering anesthesia fellow during the night, under the overall supervision of a faculty member. In fiscal year 2011, we performed >4000 consultations including 750 regional analgesia procedures. In this chapter, we will discuss limitations of currently popular modalities including patient-controlled analgesia (PCA), protocols for the management of pain after pectus excavatum, and scoliosis surgeries and the pharmacology of newly available analgesics like intravenous (IV) acetaminophen in children. We will also review current literature regarding developmental neurobiology and pharmacology in relation to neonatal/fetal pain and neurotoxicity of analgesia to the developing brain. Finally, we will introduce concepts related to pain genetics and the effects of genetic variations on pharmacokinetics and pharmacodynamics of analgesics, especially opioids. Limitations of Currently Used Analgesic Modalities

Although there has been a steady increase in regional analgesic techniques including neuraxial and peripheral nerve blocks for pediatric pain management, PCA remains the most common method of providing opioid analgesia in children.5 PCA is controlled by the patient or a proxy (usually the guardian or the nurse). Two main controversies currently exist related to PCA use in children. 1. PCA by proxy: The Joint Commission issued a sentinel alert in 2004 cautioning against this practice, in which the nurse or parent pushes the PCA button to keep the child comfortable (http://www.jointcom mission.org/assets/1/18/SEA_33.PDF), bypassing the safety feature of the PCA, wherein an oversedated patient cannot push the button. Keeping this in mind, it is essential to have criteria for selecting appropriate patients to receive PCA by proxy. Most children are able to play computer games (by age 6 y) and can understand the PCA/ “pain relief button.” At our institution, children less than 6 years of age and those with mental or disability (preventing pushing of the button) often receive PCA by proxy (nurse and/or a responsible www.anesthesiaclinics.com

68



Chidambaran and Sadhasivam

parent). The pain management team, recovery room, and inpatient floor nurses educate family members and provide written instructions regarding the proper use of PCA by proxy emphasizing the danger of pressing the button when the child is not awake. Proxycontrolled analgesia has been found to provide effective pain relief in children less than 6 years of age, although 1.7% incidence of significant apnea/desaturation reinforces the need for patient monitoring.6 With the use of modified Ramsay sedation scale monitoring and use of IV acetaminophen and ketorolac in eligible patients, at our institution, the incidence of severe respiratory depression is T) were not associated with racial differences in morphine’s clearance even though the wild-type of the UGT2B7 isozyme is more prevalent in the African Americans.48 Nonopioid Analgesics and Developmental Pharmacogenetics

Acetaminophen (APAP): APAP is bioactivated to the reactive intermediate, N-acetyl-p-benzoquinoneimine (NAPQI) by 3 CYP 450 isoforms: CYP3A4, CYP2E1, and CYP1A2.49 NAPQI then undergoes conjugation with glutathione to form a nontoxic conjugate. Depletion of glutathione stores causes NAPQI to bind with thiols leading to hepatocellular necrosis.50 Developmental pharmacology studies indicate that CYP3A4 and CYP1A2 activity in 12- to 48-month-old children exceed younger and older children, making the 12 to 48 month olds prone to APAP hepatotoxicity. This is offset by an increased capacity to conjugate the drug with sulfates, providing “relative protection” against acute APAP intoxication. Nevertheless, true polymorphisms of glutathione transferase exist and individuals with the poor metabolizer phenotype are at increased risk of APAP-induced hepatotoxicity51 NSAID: Ketorolac does appear to significantly penetrate cerebrospinal fluid in children, unlike adults. The MDR-1 gene has been suggested as the transport factor for ketorolac to cross the blood brain barrier.52 www.anesthesiaclinics.com

76



Chidambaran and Sadhasivam

Animal studies have shown that developmental differences in COX-1 expression in the spinal cord explain the lack of efficacy of COX-1 inhibitors in 3 day-old rat pups compared with 21-day-old rat pups.53 Should similar developmental expression of COX-1 occur in humans, COX-1 inhibitors such as NSAID and APAP may exhibit reduced efficacy in neonates.54 The role of pharmacogenetics in drug response has been rapidly evolving. Genetic factors account for 20% to 95% of drug response variability; 59% of drugs having adverse reactions are metabolized by at least 1 enzyme with a variant allele associated with poor metabolism.55 Presently, the FDA has approved pharmacogenetic warnings in the labels of over 100 drugs, including analgesics like codeine, tramadol/ acetaminophen combinations, and celecoxib.56 Table 1 depicts the pharmacological effects of genetic variations on opioid analgesia. OPRM1 118G variations reduces opioid signaling and increases morphine dose requirement. The presence of the V158M variant of the catechol-O-methyl transferase (COMT) coded by the 472G > A SNP leads to a low-function COMT enzyme that fails to degrade dopamine, leading to a depletion of encephalin and compensatory upregulation of opioid receptor expression. Besides, presence of Z2 nonfunctional Melanocortin Receptor (MC1R) variants resulted in impaired G-protein coupling and decreased function of MC1R receptors leading to a sex nonspecific increase in potency of m-opioid agonists. Lotsch and Geisslinger have suggested simple genotype tailored morphine dosing ranging from 0.4 to 2 times the dose of morphine required for equivalent analgesia in those without the gene variant ( 2 for OPRM1 118G,  0.67 for COMT 472A,  0.67 for 2 MC1R nonfunctional SNPs and multiplicative model for multiple SNPs),74 and this has not been validated in different pain phenotypes and populations. Hence, it remains a futuristic challenge to validate genotype-phenotype associations and incorporate them into the decision-making process of drug and dose selection, to make personalized analgesia a practicable reality and not just a distant dream. ’

Summary

Acute surgical pain management in children is best addressed by a dedicated pain management team. Although PCA with opioids forms the main modality of analgesia, regional techniques have gained popularity. PCA by proxy and PCA basal infusions enhance analgesia but carry a risk for respiratory depression and sedation. Efficient prevention of opioid-induced respiratory depression requires the use of appropriate monitoring including pulsoximetry and respiratory rate, clinical sedation scoring system, repeated assessment by the pain team, early www.anesthesiaclinics.com

*6(516G > T, 85A > G) *16, *5 (172H-262K-487C)

Polymorphisms/Alleles

Interleukin 1 Receptor antagonist (IL1Ra) gene (IL1RN)

IL1Ra 86 bp variable number of tandem repeats (VNTR) of 86 base pair (bp) in intron 2

CYP2D6 (phase I metabolism) PM have 2 nonfunctional alleles, IM have Z1 reduced functional allele or 1 active allele (IM), extensive metabolizers have Z1 or 2 functional alleles and UM have multiple functional alleles.*3 -*8, *11-*16, *19-*21, *38, *40, *42 (inactive); *1, *2, *35 (functional/wild-type); *9, *10, *17, *29, *36, *41 (decreased activity) A118G (asparagine to aspartic acid m-opioid receptor leading to reduced receptor affinity) (OPRM1): Gprotein-coupled opioid receptor

CYP2B6 (phase I metabolism)

Gene

Affects inflammatory reaction and hence nociception

(continued)

Postoperative (after knee, abdominal, orthognathic, abdominal surgery) morphine and fentanyl requirement has been found to be higher in patients with the GG genotype, and lower pain scores in the AA genotype64–67 IL1Ra SNP may contribute to variation in postoperative morphine consumption after hysterectomy68; patients homozygous for the IL1RN*1 allele have lower concentrations of IL1Ra and require higher doses of opioids after nephrectomy than patients carrying at least 1 IL1RN*2 allele69

All opioids

Codeine Tramadol Hydrocodone Oxycodone Dihydrocodeine

Increased risk of prolonged QT interval after methadone use in *6/*6 variant because of reduced metabolism57 *6 variant associated with methadonerelated mortality58 Reports of death after codeine therapy in a breast-fed infant of UM phenotype mother and a 2-y-old boy after tonsillectomy59–61 Tramadol requirement after abdominal surgery higher in PM phenotype62,63

Clinical Consequences of Genetic Variations on Opioid Effects

Methadone Meperidine Tramadol Hydromorphone

Opioids

Table 1. Genetic Polymorphisms Affecting the Pharmacokinetics and Pharmacodynamics of Opioids

Pediatric Surgical Pain Management ’

77

www.anesthesiaclinics.com

www.anesthesiaclinics.com

rs6269, rs4633, rs4818, rs4680: decrease in COMT activity is associated with higher pain sensitivity

R151C R160W D294H C1236T, C3435T, G2677T/A: reduced pglycoprotein activity results in reduced opioid efflux across blood brain barrier and increased brain concentrations

COMT

Melanocortin receptor (MC1R) Morphine Fentanyl Methadone Sufentanil Alfentanil Morphine-6glucuronide

k-agonists like pentazocine

Affects pain perception

Opioids Low pain, average pain, and high pain sensitivity haplotypes were identified based on the SNPs. rs4680 (Val 158Met) variant was associated with increased pain sensitivity70 Red-haired women with 2 nonfunctional variants had greater analgesia from kagonist, pentazocine71 The diplotype at 2677 and 3435 in ABCB1 was associated with the frequency of vomiting72; patients with ineffective efflux pump (ABCB1/MDR-1 homozygous T/T) and a functional receptor (OPRM1 homozygous A/A) were the best responders to morphine73

Clinical Consequences of Genetic Variations on Opioid Effects

ABCB1 indicates ATP-binding cassette transporter; COMT, catechol-O-methyl transferase; IM, intermediate metabolizers; IL, interleukin; PM, poor metabolizers; UM, ultrarapid metabolizer.

ABCB1 codes for p-glycoprotein

Polymorphisms/Alleles



Gene

Table 1. (continued)

78 Chidambaran and Sadhasivam

Pediatric Surgical Pain Management



79

intervention protocols, and use of nonopioid adjuncts like IV or oral acetaminophen and ketorolac/NSAIDs. Thalamocortical connections underpinning the neuroanatomy of pain appear between 20 and 30 weeks of gestational age, and the physiological mechanisms for pain perception become established by early second trimester. There are long-lasting effects of pain experienced in early life underscoring the need to treat surgical pain in fetuses, premature infants, and neonates. In contrast, there is a growing body of evidence in animal models implicating opioids in adversely altering neuronal proliferation in the developing brain and clinical studies wherein morphine sedation in the neonatal period was found to decrease visual motor integration in childhood, suggesting a potential for neurocognitive sequelae. Ongoing research provides hope that future integration of pharmacogenetics, metabolomics, and proteomics in clinical decision and analgesic selection/dosing processes will maximize analgesia and minimize adverse effects.

The authors have no conflicts of interest to disclose.



References

1. Tripp DA, Stanish WD, Reardon G, et al. Comparing postoperative pain experiences of the adolescent and adult athlete after anterior cruciate ligament surgery. J Athl Train. 2003;38:154–157. 2. Verghese ST, Hannallah RS. Acute pain management in children. J Pain Res. 2010;3:105–123. 3. Taylor EM, Boyer K, Campbell FA. Pain in hospitalized children: a prospective crosssectional survey of pain prevalence, intensity, assessment and management in a Canadian pediatric teaching hospital. Pain Res Manag. 2008;13:25–32. 4. Nelson KL, Yaster M, Kost-Byerly S, et al. A national survey of American Pediatric Anesthesiologists: patient-controlled analgesia and other intravenous opioid therapies in pediatric acute pain management. Anesth Analg. 2010;110:754–760. 5. McDonald AJ, Cooper MG. Patient-controlled analgesia: an appropriate method of pain control in children. Paediatr Drugs. 2001;3:273–284. 6. Monitto CL, Greenberg RS, Kost-Byerly S, et al. The safety and efficacy of parent-/ nurse-controlled analgesia in patients less than six years of age. Anesth Analg. 2000;91:573–579. 7. Yildiz K, Tercan E, Dogru K, et al. Comparison of patient-controlled analgesia with and without a background infusion after appendicectomy in children. Paediatr Anaesth. 2003;13:427–431. 8. Peters JW, Bandell Hoekstra IE, Huijer Abu-Saad H, et al. Patient controlled analgesia in children and adolescents: a randomized controlled trial. Paediatr Anaesth. 1999;9:235–241. 9. Stubhaug A, Breivik H. Prevention and treatment of hyperalgesia and persistent pain after surgery. Pain, Best Practice and Research Compendium, Elsevier 2007;281–288. 10. Senturk M, Ozcan PE, Talu GK, et al. The effects of three different analgesia techniques on long-term postthoracotomy pain. Anesth Analg. 2002;94:11–15. www.anesthesiaclinics.com

80



Chidambaran and Sadhasivam

11. Wong GT, Yuen VM, Chow BF, et al. Persistent pain in patients following scoliosis surgery. Eur Spine J. 2007;16:1551–1556. 12. Butkovic D, Kralik S, Matolic M, et al. Postoperative analgesia with intravenous fentanyl PCA vs epidural block after thoracoscopic pectus excavatum repair in children. Br J Anaesth. 2007;98:677–681. 13. Soliman IE, Apuya JS, Fertal KM, et al. Intravenous versus epidural analgesia after surgical repair of pectus excavatum. Am J Ther. 2009;16:398–403. 14. Izwaryn U. Bilateral intrapleural analgesia with 0.375% bupivacaine as a routine therapy after Nuss procedure for pecus excavatum in children. Rev Pediatr Surg. 2006;1:145–152. 15. Taenzer AH, Clark C. Efficacy of postoperative epidural analgesia in adolescent scoliosis surgery: a meta-analysis. Paediatr Anaesth. 2010;20:135–143. 16. Sharma S, Balireddy RK, Vorenkamp KE, et al. Beyond opioid patient-controlled analgesia: a systematic review of analgesia after major spine surgery. Reg Anesth Pain Med. 2012;37:79–98. 17. Maxwell LG, Kaufmann SC, Bitzer S, et al. The effects of a small-dose naloxone infusion on opioid-induced side effects and analgesia in children and adolescents treated with intravenous patient-controlled analgesia: a double-blind, prospective, randomized, controlled study. Anesth Analg. 2005;100:953–958. 18. Licup N, Baumrucker SJ. Methylnaltrexone: treatment for opioid-induced constipation. Am J Hosp Palliat Care. 2011;28:59–61. 19. Buchler MW, Seiler CM, Monson JR, et al. Clinical trial: alvimopan for the management of postoperative ileus after abdominal surgery: results of an international randomised, double-blind, multicentre, placebo-controlled clinical study. Aliment Pharmacol Ther. 2008;28:312–325. 20. Moffett BS, Wann TI, Carberry KE, et al. Safety of ketorolac in neonates and infants after cardiac surgery. Paediatr Anaesth. 2006;16:424–428. 21. Aldrink JH, Ma M, Wang W, et al. Safety of ketorolac in surgical neonates and infants 0 to 3 months old. J Pediatr Surg. 2011;46:1081–1085. 22. Hong JY, Won Han S, Kim WO, et al. Fentanyl sparing effects of combined ketorolac and acetaminophen for outpatient inguinal hernia repair in children. J Urol. 2010;183:1551–1555. 23. Dart RC, Rumack BH. Intravenous acetaminophen in the United States: iatrogenic dosing errors. Pediatrics. 2012;129:349–353. 24. Marret E, Rolin M, Beaussier M, et al. Meta-analysis of intravenous lidocaine and postoperative recovery after abdominal surgery. Br J Surg. 2008;95:1331–1338. 25. Massey GV, Pedigo S, Dunn NL, et al. Continuous lidocaine infusion for the relief of refractory malignant pain in a terminally ill pediatric cancer patient. J Pediatr Hematol Oncol. 2002;24:566–568. 26. Giannakoulopoulos X, Teixeira J, Fisk N, et al. Human fetal and maternal noradrenaline responses to invasive procedures. Pediatr Res. 1999;45:494–499. 27. Fisk NM, Gitau R, Teixeira JM, et al. Effect of direct fetal opioid analgesia on fetal hormonal and hemodynamic stress response to intrauterine needling. Anesthesiology. 2001;95:828–835. 28. Lee SJ, Ralston HJ, Drey EA, et al. Fetal pain: a systematic multidisciplinary review of the evidence. JAMA. 2005;294:947–954. 29. Brusseau R. Developmental perspectives: is the fetus conscious? Int Anesthesiol Clin. 2008;46:11–23. 30. Anand KJ, Aranda JV, Berde CB, et al. Summary proceedings from the neonatal pain-control group. Pediatrics. 2006;117:S9–S22. 31. Fitzgerald M, Walker SM. Infant pain management: a developmental neurobiological approach. Nature Clin Pract Neurol. 2009;5:35–50. 32. Beggs S, Currie G, Salter MW, et al. Priming of adult pain responses by neonatal pain experience: maintenance by central neuroimmune activity. Brain. 2012;135:404–417. www.anesthesiaclinics.com

Pediatric Surgical Pain Management



81

33. Anand KJ, Sippell WG, Aynsley-Green A. Randomised trial of fentanyl anaesthesia in preterm babies undergoing surgery: effects on the stress response. Lancet. 1987;1:243–248. 34. Taddio A, Katz J, Ilersich AL, et al. Effect of neonatal circumcision on pain response during subsequent routine vaccination. Lancet. 1997;349:599–603. 35. Boat AC, Sadhasivam S, Loepke AW, et al. Outcome for the extremely premature neonate: how far do we push the edge? Paediatr Anaesth. 2011;21:765–770. 36. Handelmann GE, Dow-Edwards D. Modulation of brain development by morphine: effects on central motor systems and behavior. Peptides. 1985;6(suppl 2):29–34. 37. Sargeant TJ, Day DJ, Miller JH, et al. Acute in utero morphine exposure slows G2/M phase transition in radial glial and basal progenitor cells in the dorsal telencephalon of the E15.5 embryonic mouse. Eur J Neurosci. 2008;28:1060–1067. 38. Hu S, Sheng WS, Lokensgard JR, et al. Morphine induces apoptosis of human microglia and neurons. Neuropharmacology. 2002;42:829–836. 39. Mess B, Ruzsas C, Hayashi S. Impaired thyroid function provoked by neonatal treatment with drugs affecting the maturation of monoaminergic and opioidergic neurons. Exp Clin Endocrinol. 1989;94:73–81. 40. MacGregor R, Evans D, Sugden D, et al. Outcome at 5-6 years of prematurely born children who received morphine as neonates. Arch Dis Child Fetal Neonatal Ed. 1998;79:F40–F43. 41. de Graaf J, van Lingen RA, Simons SH, et al. Long-term effects of routine morphine infusion in mechanically ventilated neonates on children’s functioning: five-year follow-up of a randomized controlled trial. Pain. 2011;152:1391–1397. 42. Sanders RD, Sun P, Patel S, et al. Dexmedetomidine provides cortical neuroprotection: impact on anaesthetic-induced neuroapoptosis in the rat developing brain. Acta Anaesthesiol Scand. 2010;54:710–716. 43. Broussard CS, Rasmussen SA, Reefhuis J, et al. Maternal treatment with opioid analgesics and risk for birth defects. Am J Obstet Gynecol. 2011;204:e1–e11. 44. Holliday R. Mechanisms for the control of gene activity during development. Biol Rev Camb Philos Soc. 1990;65:431–471. 45. Szyf M. Epigenetics, DNA methylation, and chromatin modifying drugs. Annu Rev Pharmacol Toxicol. 2009;49:243–263. 46. Chidambaran V, Ngamprasertwong P, Vinks AA, et al. Pharmacogenetics and anesthetic drugs. Curr Clin Pharmacol. 2012;7:78–101. 47. Sadhasivam S, Chidambaran V, Ngamprasertwong P, et al. Race and unequal burden of perioperative pain and opioid related adverse effects in children. Pediatrics. 2012;129:832–838. 48. Sadhasivam S, Krekels E, Chidambaran V, et al. Morphine clearance in children: does race or genetics matter? J Opioid Manag. 2012;8:217–226. 49. Patten CJ, Thomas PE, Guy RL, et al. Cytochrome P450 enzymes involved in acetaminophen activation by rat and human liver microsomes and their kinetics. Chem Res Toxicol. 1993;6:511–518. 50. Murray GI, Foster CO, Barnes TS, et al. Cytochrome P450IA expression in adult and fetal human liver. Carcinogenesis. 1992;13:165–169. 51. Kearns GL, Leeder JS, Wasserman GS. Acetaminophen intoxication during treatment: what you don’t know can hurt you. Clin Pediatr (Phila). 2000;39:133–144. 52. Kumpulainen E, Kokki H, Laisalmi M, et al. How readily does ketorolac penetrate cerebrospinal fluid in children? J Clin Pharmacol. 2008;48:495–501. 53. Gupta A, Cheng J, Wang S, et al. Analgesic efficacy of ketorolac and morphine in neonatal rats. Pharmacol Biochem Behav. 2001;68:635–640. 54. Ririe DG, Prout HD, Barclay D, et al. Developmental differences in spinal cyclooxygenase 1 expression after surgical incision. Anesthesiology. 2006;104: 426–431. www.anesthesiaclinics.com

82



Chidambaran and Sadhasivam

55. Girard H, Court MH, Bernard O, et al. Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics. 2004;14:501–515. 56. US Food and Drug Administration. Table on Pharmacogenomic Biomarkers in Drug Labels. http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ ucm083378.htm. US Food and Drug Administration; Accessed September 3, 2012. 57. Eap CB, Bochud M, Elston RC, et al. CYP3A5 and ABCB1 genes influence blood pressure and response to treatment, and their effect is modified by salt. Hypertension. 2007;49:1007–1014. 58. Bunten H, Liang WJ, Pounder DJ, et al. OPRM1 and CYP2B6 gene variants as risk factors in methadone-related deaths. Clin Pharmacol Ther. 2010;88:383–389. 59. Koren G, Cairns J, Chitayat D, et al. Pharmacogenetics of morphine poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet. 2006;368:704. 60. Ciszkowski C, Madadi P, Phillips MS, et al. Codeine, ultrarapid-metabolism genotype, and postoperative death. N Engl J Med. 2009;361:827–828. 61. Kelly LE, Rieder M, van den Anker J, et al. More codeine fatalities after tonsillectomy in North American children. Pediatrics. 2012;129:e1343–e1347. 62. Stamer UM, Lehnen K, Hothker F, et al. Impact of CYP2D6 genotype on postoperative tramadol analgesia. Pain. 2003;105:231–238. 63. Wang G, Zhang H, He F, et al. Effect of the CYP2D6*10 C188T polymorphism on postoperative tramadol analgesia in a Chinese population. Eur J Clin Pharmacol. 2006;62:927–931. 64. Chou WY, Wang CH, Liu PH, et al. Human opioid receptor A118G polymorphism affects intravenous patient-controlled analgesia morphine consumption after total abdominal hysterectomy. Anesthesiology. 2006;105:334–337. 65. 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;103:1011–1017. 66. Sia AT, Lim Y, Lim EC, et al. A118G single nucleotide polymorphism of human muopioid receptor gene influences pain perception and patient-controlled intravenous morphine consumption after intrathecal morphine for postcesarean analgesia. Anesthesiology. 2008;109:520–526. 67. Fukuda K, Hayashida M, Ide S, et al. Association between OPRM1 gene polymorphisms and fentanyl sensitivity in patients undergoing painful cosmetic surgery. Pain. 2009;147:194–201. 68. Bessler H, Shavit Y, Mayburd E, et al. Postoperative pain, morphine consumption, and genetic polymorphism of IL-1beta and IL-1 receptor antagonist. Neurosci Lett. 2006;404:154–158. 69. Candiotti KA, Yang Z, Morris R, et al. Polymorphism in the interleukin-1 receptor antagonist gene is associated with serum interleukin-1 receptor antagonist concentrations and postoperative opioid consumption. Anesthesiology. 2011;114:1162–1168. 70. Diatchenko L, Nackley AG, Slade GD, et al. Catechol-O-methyltransferase gene polymorphisms are associated with multiple pain-evoking stimuli. Pain. 2006;125:216–224. 71. Mogil JS, Wilson SG, Chesler EJ, et al. The melanocortin-1 receptor gene mediates female-specific mechanisms of analgesia in mice and humans. Proc Natl Acad Sci USA. 2003;100:4867–4872. 72. Fujita KI, Ando Y, Yamamoto W, et al. Association of UGT2B7 and ABCB1 genotypes with morphine-induced adverse drug reactions in Japanese patients with cancer. Cancer Chemother Pharmacol. 2010;65:251–288. 73. Campa D, Gioia A, Tomei A, et al. Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther. 2008;83:559–566. 74. Lotsch J, Geisslinger G. Current evidence for a genetic modulation of the response to analgesics. Pain. 2006;121:1–5. www.anesthesiaclinics.com

Suggest Documents