References

ACVAA. American College of Veterinary Anesthesiologists' position paper on the treatment of pain in animals. 2006. http://www.acvaa.org/docs/Pain_Treatment (accessed: 15 June 2020)

Aragon CL, Read MR, Gaynor JS, Barnhart MD, Wilson D, Papich MG. Pharmacokinetics of an immediate and extended release oral morphine formulation utilizing the spheroidal oral drug absorption system in dogs. Journal of veterinary pharmacology and therapeutics. 2009; 32:(2)129-136 https://doi.org/10.1111/j.1365-2885.2008.01011.x

Boothe DM. Control of pain in small animals: opioid agonists and antagonists and other locally and centrally acting analgesics, 2nd edn. In: Boothe DM (eds.). Missouri: Elsevier; 2012

Dohoo S, Tasker RAR, Donald A. Pharmacokinetics of parenteral and oral sustained-release morphine sulphate in dogs. Journal of Veterinary Pharmacology and Therapeutics. 1994; 17:(6)426-433

Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest. 2010; 120:(11)3760-3772 https://doi.org/10.1172/JCI42843

Dumas EO, Pollack GM. Opioid tolerance development: a pharmacokinetic/pharmacodynamic perspective. AAPS J. 2008; 10:(4)537-551 https://doi.org/10.1208/s12248-008-9056-1

Egger CM, Glerum LE, Allen SW, Haag M. Plasma fentanyl concentrations in awake cats and cats undergoing anesthesia and ovariohysterectomy using transdermal administration. Vet Anaesth Analg. 2003; 30:(4)229-236 https://doi.org/10.1046/j.1467-2995.2003.00109.x

Feierman DE. Identification of cytochrome P450 3A1/2 as the major P450 isoform responsible for the metabolism of fentanyl by rat liver microsomes. Anesth Analg. 1996; 82:(5)936-941

Ferrari A, Coccia CP, Bertolini A, Sternieri E. Methadone—metabolism, pharmacokinetics and interactions. Pharmacol Res. 2004; 50:(6)551-9 https://doi.org/10.1016/j.phrs.2004.05.002

Garrett ER, Derendorf H, Mattha AG. Pharmacokinetics of Morphine and its surrogates VII: High-performance liquid chromatographic analyses and pharmacokinetics of methadone and its derived metabolites in dogs. Journal of Pharmaceutical Sciences. 1985; 74:(11)1203-1214

Holmquist GL. Opioid metabolism and effects of cytochrome P450. Pain Medicine. 2009; 10:(S1)S20-S29 https://doi.org/10.1111/j.1526-4637.2009.00596.x

Ingvast-Larsson C, Holgersson A, Bondesson U, Lagerstedt AS, Olsson K. Clinical pharmacology of methadone in dogs. Vet Anaesth Analg. 2010; 37:(1)48-56 https://doi.org/10.1111/j.1467-2995.2009.00476.x

Jacqz E, Ward S, Johnson R, Schenker S, Gerkens J, Branch R. Extrahepatic glucuronidation of morphine in the dog. Drug Metabolism and Disposition. 1986; 14:(6)627-630

Kerr CL. Pain Management I: systemic analgesics, 3rd edn. In: Duke-Novakovski T, Vries MD, Seymour C (eds.). Gloucester: British Small Animal Veterinary Association; 2016

KuKanich B, Borum SL. The disposition and behavioral effects of methadone in Greyhounds. Vet Anaesth Analg. 2008; 35:(3)242-248 https://doi.org/10.1111/j.1467-2995.2007.00369.x

KuKanich B, Hubin M. The pharmacokinetics of ketoconazole and its effects on the pharmacokinetics of midazolam and fentanyl in dogs. J Vet Pharmacol Ther. 2010; 33:(1)42-49 https://doi.org/10.1111/j.1365-2885.2009.01105.x

KuKanich B, Wiese AJ. Opioids, 5th edn. In: Grimm KA, Lamont LA, Tranquilli WJ, Greene SA, Robertson SA (eds.). : John-Wiley & Sons, Iowa; 2015

Kukanich B, Lascelles BDX, Aman AM, Mealey KL, Papich MG. The effects of inhibiting cytochrome P450 3A, p-glycoprotein, and gastric acid secretion on the oral bioavailability of methadone in dogs. J Vet Pharmacol Ther. 2005a; 28:(5)461-466 https://doi.org/10.1111/j.1365-2885.2005.00681.x

KuKanich B, Lascelles BDX, Papich MG. Use of a von Frey device for evaluation of pharmacokinetics and pharmacodynamics of morphine after intravenous administration as an infusion or multiple doses in dogs. Am J Vet Res. 2005b; 66:(11)1968-1974 https://doi.org/10.2460/ajvr.2005.66.1968

Kukanich B, Kukanich KS, Rodriguez JR. The effects of concurrent administration of cytochrome P-450 inhibitors on the pharmacokinetics of oral methadone in healthy dogs. Vet Anaesth Analg. 2011; 38:(3)224-30 https://doi.org/10.1111/j.1467-2995.2011.00602.x

Kyles AE, Papich M, Hardie EM. Disposition of transdermally administered fentanyl in dogs. Am J Vet Res. 1996; 57:(5)715-719

Labroo RB, Paine MF, Thummel KE, Kharasch ED. Fentanyl metabolism by human hepatic and intestinal cytochrome P450 3A4: implications for interindividual variability in disposition, efficacy, and drug interactions. Drug Metab Dispos. 1997; 25:(9)1072-1080

Lambert DG. Drugs and receptors. Continuing Education in Anaesthesia Critical Care & Pain. 2004; 4:(6)181-184 https://doi.org/10.1093/bja-ceaccp/mkh049

Maxwell SRJ. Pharmacodynamics for the prescriber. Medicine (Baltimore). 2012; 40:(7)351-356 https://doi.org/10.1016/j.mpmed.2012.05.002

Mazoit JX, Butscher K, Samii K. Morphine in postoperative patients: pharmacokinetics and pharmacodynamics of metabolites. Anesth Analg. 2007; 105:(1)70-8 https://doi.org/10.1213/01.ane.0000265557.73688.32

McKune CM, Murrell JC, Nolan AM, White KL, Wright BD. Nociception and pain, 5th edn. In: Grimm KA, Lamont LA, Tranquilli WJ, Greene SA, Robertson SA (eds.). : John-Wiley & Sons, Iowa; 2015

Monteiro ER, Junior AR, Assis HMQ, Campagnol D, Quitzan JG. Comparative study on the sedative effects of morphine, methadone, butorphanol or tramadol, in combination with acepromazine, in dogs. Vet Anaesth Analg. 2009; 36:(1)25-33 https://doi.org/10.1111/j.1467-2995.2008.00424.x

Peck TE, Hill SA. Drug Action, 4th edn. In: Peck TE, Hill SA. Cambridge: Cambridge University Press; 2014a

Peck TE, Hill SA. Analgesics, 4th edn. In: Peck T.E., Hill S.A. Cambridge: Cambridge University Press; 2014b

Ramsey I. BSAVA Small Animal Formulary, 8th edn. Gloucester: BSAVA; 2014

Reddi D, Curran N, Stephens R. An introduction to pain pathways and mechanisms. British Journal of Hospital Medicine. 2013; 74:(12)C188-C191 https://doi.org/10.12968/hmed.2013.74.Sup12.C188

Riviere JE, Qiao GL. Heptic biotransformation and biliary excretion. In: Riviere JE (ed.). Ames, Iowa: Iowa State University Press; 1999

Robinson TM, Kruse-Elliott KT, Markel MD, Pluhar GE, Massa K, Bjorling DE. A comparison of transdermal fentanyl versus epidural morphine for analgesia in dogs undergoing major orthopedic surgery. J Am Anim Hosp Assoc. 1999; 35:(2)95-100 https://doi.org/10.5326/15473317-35-2-95

Romagnoli N, Barbarossa A, Cunto M Evaluation of methadone concentrations in bitches and in umbilical cords after epidural or systemic administration for caesarean section: A randomized trial. Vet Anaesth Analg. 2019; 46:(3)375-383 https://doi.org/10.1016/j.vaa.2018.10.005

Sams RA, Muir WW, Detra RL, Robinson EP. Comparative pharmacokinetics and anesthetic effects of methohexital, pentobarbital, thiamylal, and thiopental in Greyhound dogs and non-Greyhound, mixed-breed dogs. Am J Vet Res. 1985; 46:(8)1677-1683

Steagall PVM, Simon BT, Teixeira Neto FJ, Luna SPL. An Update on Drugs Used for Lumbosacral Epidural Anesthesia and Analgesia in Dogs. Front Vet Sci. 2017; 4 https://doi.org/10.3389/fvets.2017.00068

Taylor PM, Robertson SA, Dixon MJ Morphine, pethidine and buprenorphine disposition in the cat. J Vet Pharmacol Ther. 2001; 24:(6)391-8 https://doi.org/10.1046/j.1365-2885.2001.368goblin.x

Torske KE, Dyson DH. Epidural analgesia and anesthesia. Vet Clin North Am Small Anim Pract. 2000; 30:(4)859-74 https://doi.org/10.1016/s0195-5616(08)70011-1

Pharmacology of opioids-part 1 anaesthesia tutorial of the week 64. 2007. https://www.frca.co.uk/article.aspx?articleid=100933 (accessed: 9 February 2021)

Troncy E, Junot S, Keroack S Results of preemptive epidural administration of morphine with or without bupivacaine in dogs and cats undergoing surgery: 265 cases (1997-1999). J Am Vet Med Assoc. 2002; 221:(5)666-72 https://doi.org/10.2460/javma.2002.221.666

Veterinary Medicines Directorate. Product Information Database. 2018. https://www.vmd.defra.gov.uk/ProductInformationDatabase/ (accessed: 15 June 2020)

White DM, Mair AR, Martinez-Taboada F. Opioid-free anaesthesia in three dogs. Open Vet J. 2017; 7:(2)104-110 https://doi.org/10.4314/ovj.v7i2.5

Yamada H, Ishii K, Ishii Y Formation of highly analgesic morphine-6-glucuronide following physiologic concentration of morphine in human brain. J Toxicol Sci. 2003; 28:(5)395-401 https://doi.org/10.2131/jts.28.395

Zoran DL, Riedesel DH, Dyer DC. Pharmacokinetics of propofol in mixed-breed dogs and greyhounds. Am J Vet Res. 1993; 54:(5)755-760

Pharmacology of opioids and application to practice

02 February 2021
12 mins read
Volume 12 · Issue 1
Figure 1. Primary afferent neurons.

Abstract

This article analyses the pharmacological considerations when administering opioids in practice. Beginning with a basic review of pain pathways, followed by a more in-depth analysis of opioid pharmacology. The focus will be on the three most commonly used full mu agonist opioids (methadone, morphine, fentanyl). The aim of this article is to show the importance of applying pharmacology to a clinical situation, promoting individual pain management assessment. This involves the application of the most appropriate opioid, as part of a more extensive pain management plan to improve analgesic efficacy and patient outcomes.

The inhibition of nociceptive pathways reduces the sensory and emotional aspects of pain. Understanding these pathways, allows the selection of appropriate agents and the ethical treatment of a variety of species in veterinary practice. This article will concentrate on the pain pathways, the pharmacology of opioids and their role in administration of analgesia. As veterinary literature can be limited this analysis will use a combination of human and veterinary studies to provide a more comprehensive understanding.

Pain is an unpleasant experience providing sensory and emotional components to actual or potential tissue damage (McKune et al, 2015).

Physiologic pain is detected and interpreted by a nociceptive sensory system, responding to pain through transduction, transmission, modulation, and perception (McKune et al, 2015).

Peripheral nociceptors detect noxious (thermal, mechanical, and chemical) stimuli and an action potential is transmitted by two types of primary afferent neuron: C-fibres and A-δ fibres (Dubin and Patapoutian, 2010). A-δ fibres are lightly myelinated fibres with a diameter of 2–5 µm (Figure 1), and have a low threshold for noxious stimuli (thermal and mechanical), providing rapid delivery of action potentials in response to acute pain. C-fibres are unmyelinated and have a diameter of <2 µm (Figure 1), and provide slow delivery of action potentials. C-fibres have a high threshold and are polymodal; therefore, they respond to chemical, mechanical, and thermal noxious stimulation (Reddi et al, 2013).

Register now to continue reading

Thank you for visiting The Veterinary Nurse and reading some of our peer-reviewed content for veterinary professionals. To continue reading this article, please register today.