Effects of sedative psychotropic drugs combined with oxycodone on respiratory depression in the rat

Lin Xu, Ashok Krishna, Sharron Stewart, Katherine Shea, Rebecca Racz, James L Weaver, Donna A Volpe, Nageswara R Pilli, Suresh Narayanasamy, Jeffry Florian, Vikram Patel, Murali K Matta, Marc B Stone, Hao Zhu, Michael C Davis, David G Strauss, Rodney Rouse, Lin Xu, Ashok Krishna, Sharron Stewart, Katherine Shea, Rebecca Racz, James L Weaver, Donna A Volpe, Nageswara R Pilli, Suresh Narayanasamy, Jeffry Florian, Vikram Patel, Murali K Matta, Marc B Stone, Hao Zhu, Michael C Davis, David G Strauss, Rodney Rouse

Abstract

Following a decision to require label warnings for concurrent use of opioids and benzodiazepines and increased risk of respiratory depression and death, the US Food and Drug Administratioin (FDA) recognized that other sedative psychotropic drugs may be substituted for benzodiazepines and be used concurrently with opioids. In some cases, data on the ability of these alternatives to depress respiration alone or in conjunction with an opioid are lacking. A nonclinical in vivo model was developed that could detect worsening respiratory depression when a benzodiazepine (diazepam) was used in combination with an opioid (oxycodone) compared to the opioid alone based on an increased arterial partial pressure of carbon dioxide (pCO2 ). The current study used that model to assess the impact on respiration of non-benzodiazepine sedative psychotropic drugs representative of different drug classes (clozapine, quetiapine, risperidone, zolpidem, trazodone, carisoprodol, cyclobenzaprine, mirtazapine, topiramate, paroxetine, duloxetine, ramelteon, and suvorexant) administered alone and with oxycodone. At clinically relevant exposures, paroxetine, trazodone, and quetiapine given with oxycodone significantly increased pCO2 above the oxycodone effect. Analyses indicated that most pCO2 interaction effects were due to pharmacokinetic interactions resulting in increased oxycodone exposure. Increased pCO2 recorded with oxycodone-paroxetine co-administration exceeded expected effects from only drug exposure suggesting another mechanism for the increased pharmacodynamic response. This study identified drug-drug interaction effects depressing respiration in an animal model when quetiapine or paroxetine were co-administered with oxycodone. Clinical pharmacodynamic drug interaction studies are being conducted with these drugs to assess translatability of these findings.

Conflict of interest statement

The authors declared no competing interests for this work.

Published 2021. This article is a U.S. Government work and is in the public domain in the USA. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

FIGURE 1
FIGURE 1
Comparison of arterial partial pressure of carbon dioxide (pCO2). Changes between oxycodone alone and paroxetine (a) or Quetiapine (b) co‐administered with oxycodone. Plots of mean pCO2 at each timepoint following administration shows a significant increase in pCO2 with co‐administration of oxycodone (150 mg/kg) with (a) paroxetine (50 mg/kg) and (b) quetiapine (250 mg/kg) compared to oxycodone alone (150 mg/kg). Values are mean ± SD; significant difference (p < 0.05) was determined by analysis of variance across the entire experimental times series for both paroxetine and quetiapine; n = 6 per experimental group. Paroxetine was given 3 h prior to time zero when oxycodone was administered. Quetiapine and oxycodone were given concurrently
FIGURE 2
FIGURE 2
Effects of oxycodone (Oxy) alone (a) versus combined effects of oxycodone with diazepam (b), paroxetine (c), quetiapine (d), ramelteon (e), and trazodone (f) on change from baseline arterial partial pressure of carbon dioxide (pCO2). Shown are the univariate (a) and multivariate (b–f) linear regression results using data from the animal experiments with oxycodone alone, the sedative psychotropic drug (SPD) alone, and the SPD combined with oxycodone. The univariate linear regression for oxycodone (dark gray with 95% confidence interval [light gray]) is shown to display model‐predicted effects on change from baseline arterial pCO2 from oxycodone alone. Multivariate linear regression models were developed for each combination of oxycodone and SPD. The resulting oxycodone relationship from the multivariate linear regression is shown on each plot for comparison with the effects from oxycodone combined with the SPD. Shown in blue are the mean change from baseline arterial pCO2 with 95% confidence for the SPD and oxycodone combination arms (amount administered is labeled on each figure). Each point is represented on the x‐axis based on the geometric maximum concentration of all rats at that dose and/or combination. If the combination treatment is less than or overlaps with the mean effect of oxycodone alone (dark gray), this would suggest oxycodone alone could explain the observed effects on change from baseline arterial pCO2. Likewise, if the combination effect is greater, this would suggest the drug combination is having an additional effect, which could be due to the SPD alone or synergy

References

    1. SAMSA: key substance use and mental health indicators in the United States: results from the 2018 National Survey on Drug Use and Health. 2020. . Accessed March 17, 2020.
    1. Hedegaard H, Miniño AM, Warner M. Drug overdose deaths in the United States, 1999–2018. NCHS Data Brief, no 356. Hyattsville, MD: National Center for Health Statistics; 2020.
    1. Boudreau D, Von Korff M, Rutter CM, et al. Trends in long‐term opioid therapy for chronic non‐cancer pain. Pharmacoepidemiol Drug Saf. 2009;18:1166‐1175.
    1. NIDA: overdose death rates (attached supplemental document: drug overdoses data document), revised March 2020. National Institute of Drug Abuse. 2020. . Accessed March 17, 2020.
    1. Baltimore Commissioner of Health , Citizens petition. 2016. . Accessed March 12, 2020.
    1. FDA 2016, New safety measures announced for opioid analgesics, prescription opioid cough products, and benzodiazepines. . Accessed March 18, 2020.
    1. Xu L, Chockalingam A, Stewart S, et al. Developing an animal model to detect drug‐drug interactions impacting drug‐induced respiratory depression. Toxicol Rep. 2020;7:188‐197.
    1. National Institutes of Health (NIH), US National Library of Medicine . . Clinical study to investigate the effect of the combination of psychotropic drugs and an opioid on ventilation. . Accessed May 15, 2020.
    1. Guide for the Care and Use of Laboratory Animals, 8th ed. 2011. . Accessed March 12, 2020.
    1. FDA, Guidance for Industry . Estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. 2005. . Accessed March 17, 2020.
    1. Batra VR, Schrott LM. Acute oxycodone induces the pro‐emetic pica response in rats. J Pharmacol Exp Ther. 2011;339(3):738‐745.
    1. Merchant KM, Dobner PR, Dorsa DM. Differential effects of haloperidol and clozapine on neurotensin gene transcription in rat neostriatum. J Neurosci. 1992;12(2):652‐663.
    1. Gao XM, Hashimoto T, Cooper TB, Tamminga CA. The dose‐response characteristics of rat oral dyskinesias with chronic haloperidol or clozapine administration. J Neural Transm. 1997;104:97‐104.
    1. Sun L, Lau CE. Intravenous and oral clozapine pharmacokinetics, pharmacodynamics, and concentration‐effect relations: acute tolerance. Eur J Pharmacol. 2000;398(2):225‐238.
    1. Invernizzi R, Morali F, Pozzi L, Samanin R. Effects of acute and chronic clozapine on dopamine release and metabolism in the striatum and nucleus accumbens of conscious rats. Br J Pharmacol. 1990;100(4):774‐778.
    1. Ezzeldin E, Asiri YA, Iqbal M. Effects of green tea extracts on the pharmacokinetics of quetiapine in rats. Evid Based Complement Alternat Med. 2015;2015:615285.
    1. Aravagiri M, Marder SR. Brain, plasma and tissue pharmacokinetics of risperidone and 9‐hydroxyrisperidone after separate oral administration to rats. Psychopharmacology. 2002;159(4):424‐431.
    1. Diaz‐Garcia JM, Oliver‐Botana J, Galve DF. Pharmacokinetics of diazepam in the rat: influence of an experimentally induced hepatic injury. Eur J Drug Metab Pharmacokinet. 1991;3:94‐101.
    1. Brisbare‐Roch C, Dingemanse J, Koberstein R, et al. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med. 2007;13:150‐155.
    1. Me Abdel Salam O, Sleem AA, Shafee N. Effect of trazodone and nefazodone on hepatic injury induced by carbon tetrachloride. Drug Discov Ther. 2010;4(4):285‐297.
    1. Gonzalez LA, Gatch MB, Taylor CM, Bell‐Horner CL, Forster MJ, Dillon GH. Carisoprodol‐mediated modulation of GABAA receptors. In vitro and in vivo studies. J Pharmacol Exp Ther. 2009;329(2):827‐837.
    1. Flexeril® (Cyclobenzaprine HCl) Tablets – FDA NDA 17‐821/S‐045. . Accessed May 15, 2020.
    1. Lightowler S, Kennett GA, Williamson IJ, Blackburn TP, Tulloch IF. Anxiolytic‐like effect of paroxetine in a rat social interaction test. Pharmacol Biochem Behav. 1994;49(2):281‐285.
    1. Rouini MR, Lavasani H, Sheikholeslami B, Owen H, Giorgi M. Pharmacokinetics of mirtazapine and its main metabolites after single intravenous and oral administrations in rats at two dose rates. DARU J Pharmaceut Sci. 2014;22(1):13.
    1. Wauquier A, Zhou S. Topiramate: a potent anticonvulsant in the amygdala‐kindled rat. Epilepsy Res. 1996;24(2):73‐77.
    1. Iyengar S, Webster AA, Hemrick‐Luecke SK, Xu JY, Simmons RMA. Efficacy of duloxetine, a potent and balanced serotonin‐norepinephrine reuptake inhibitor in persistent pain models in rats. J Pharmacol Exp Ther. 2004;311(2):576‐584.
    1. Hirai K, Kita M, Ohta H, et al. Ramelteon (TAK‐375) accelerates reentrainment of circadian rhythm after a phase advance of the light‐dark cycle in rats. J Biol Rhythms. 2005;20(1):27‐37.
    1. Winrow CJ, Gotter AL, Cox CD, et al. Promotion of sleep by suvorexant‐a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1–2):52‐61.
    1. Pilli NR, Narayanasamy S, Lin XU, et al. A high‐throughput bioanalytical assay to support pharmacokinetic interaction study of oxycodone and diazepam in Sprague Dawley rats. RSC Adv. 2020;10:886‐896.
    1. Narayanasamy S, Pilli NR, Lin XU, et al. An alternating polarity switching assay for quantification of oxycodone and topiramate: An application of LC‐MS/MS method in support to PK/PD study in rodents. J Chromatogr B. 2019;1119:93‐100.
    1. Pandya NK, Sharma S. Capnography and pulse oximetry. NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health. Treasure Island, FL: StatPearls Publishing; 2020.
    1. Jaffer KY, Chang T, Vanle B, et al. Tradozone for insomnia: a systematic review. Innov Clin Neurosci. 2017;14(7–8):24‐34.
    1. Ciranni MA, Kearney TE, Olson KR. Comparing acute toxicity of first‐ and second‐generation antipsychotic drugs: a 10‐year, retrospective study. J Clin Psychiat. 2009;70:122‐129.
    1. Henderson DR, Konkle DM, Mitchell GS. Effects of serotonin re‐uptake inhibition on ventilator control in goats. Resp Physiol. 1999;115:1‐10.
    1. Erjavec MK, Coda BA, Nguyen Q, Donaldson G, Risler L, Shen DD. Morphine‐fluoxetine interactions in healthy volunteers: analgesia and side effects. J Clin Pharmacol. 2000;40:1286‐1295.
    1. Lalovic B, Phillips B, Risler LL, Howald W, Shen DD. Quantitative contribution of CYP2D6 and CYP3A to oxycodone metabolism in human liver and intestinal microsomes. Drug Metab Dispos. 2004;32(4):447‐454.
    1. Andreassen TN, Klepstad P, Davies A, et al. Influences on the pharmacokinetics of oxycodone: a multicentre cross‐sectional study in 439 adult cancer patients. Eur J Clin Pharmacol. 2011;67(5):493‐506.
    1. Saari TI, Grönlund J, Hagelberg NM, et al. Effects of itraconazole on the pharmacokinetics and pharmacodynamics of intravenously and orally administered oxycodone. Eur J Clin Pharmacol. 2010;66(4):387‐397.
    1. Heiskanen T, Olkkola KT, Kalso E. Effects of blocking CYP2D6 on the pharmacokinetics and pharmacodynamics of oxycodone. Clin Pharmacol Ther. 1998;64(6):603‐611.
    1. Jornil J, Jensen KG, Larsen K, Linnet K. Identification of cytochrome P450 isoforms involved in the metabolism of paroxetine and estimation of their importance for human paroxetine metabolism using a population‐based simulator. Drug Metab Dispos. 2010;38(3):376‐385.
    1. VandenBrink BM, Foti RS, Rock DA, Wienkers LC, Wahlstrom JL. Prediction of CYP2D6 Drug interactions from in vitro data: evidence for substrate‐dependent inhibition. Drug Metab Dispos. 2012;40(1):47‐53.
    1. Lemberg KK, Heiskanen TE, Neuvonen M, et al. Does co‐administration of paroxetine change oxycodone analgesia: an interaction study in chronic pain patients. Scand J Pain. 2010;1(1):24‐33.
    1. Grönlund J, Saari TI, Hagelberg NM, Neuvonen PJ, Olkkola KT, Laine K. Exposure to oral oxycodone is increased by concomitant inhibition of CYP2D6 and 3A4 pathways, but not by inhibition of CYP2D6 alone. Br J Clin Pharmacol. 2010;70(1):78‐87.
    1. Kummer O, Hammann F, Moser C, Schaller O, Drewe J, Krähenbühl S. Effect of the inhibition of CYP3A4 or CYP2D6 on the pharmacokinetics and pharmacodynamics of oxycodone. Eur J Clin Pharmacol. 2011;67:63‐71.
    1. Martignoni M, Groothuis GMM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP‐mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol. 2006;2(6):875‐894.
    1. Read DJ. A clinical method for assessing the ventilatory response to carbon dioxide. Australas Ann Med. 1967;16(1):20‐32.
    1. NDA 22272 Clinical Pharmacology/Biopharmaceutics Review, Table 2.3 (page 28). . Accessed May 15, 2020.
    1. Glue P, Gale C, Menkes DB, Hung N. Evaluation of bioequivalence between clozapine suspension and tablet formulations. Clin Drug Invest. 2012;32(11):723‐727.
    1. NDA 21777 clinical pharmacology/biopharmaceutics review, Table 1 (page 8). . Accessed May 15, 2020.
    1. NDA 21427 clinical pharmacology/biopharmaceutics review, Table 39 (page 2). . Accessed May 15, 2020.
    1. Spaans E, van den Heuvel MW, Schnabel PG, et al. Concomitant use of mirtazapine and phenytoin: a drug–drug interaction study in healthy male subjects. Eur J Clin Pharmacol. 2002;58(6):423‐429.
    1. NDA 21299 clinical pharmacology/biopharmaceutics review, Table 5 (page 22). . Accessed May 15, 2020.
    1. NDA 21299 clinical pharmacology/biopharmaceutics review, Table 5 (page 22). . Accessed May 15, 2020.
    1. Thyrum PT, Wong YW, Yeh C. Single‐dose pharmacokinetics of quetiapine in subjects with renal or hepatic impairment. Prog Neuropsychopharmacol Biol Psychiatry. 2000;24(4):521‐533.
    1. NDA 20639 S016 and S017 clinical pharmacology/biopharmaceutics review, Table 1 (page 313). . Accessed May 15, 2020.
    1. Karim A, Tolbert D, Cao C. Disposition kinetics and tolerance of escalating single doses of ramelteon, a high‐affinity MT1 and MT2 melatonin receptor agonist indicated for treatment of insomnia. J Clin Pharmacol. 2006;46(2):140‐148.
    1. NDA 20272 Clinical Pharmacology/Biopharmaceutics Review, Table 9 (page 11). . Accessed May 15, 2020.
    1. NDA 204569 Clinical Pharmacology/Biopharmaceutics Review, page 58. . Accessed May 15, 2020.
    1. Bialer M, Shekh‐Ahmad T, Braun TL, Halvorsen MB. Comparative steady‐state pharmacokinetic evaluation of immediate‐release topiramate and USL 255, a once‐daily extended‐release topiramate formulation. Epilepsia. 2013;54(8):1444‐1452.
    1. Gammans RE, Mackenthun AV, Russell JW. Comparative bioavailability of trazodone formulations using stable isotope methodology. Br J Clin Pharmacol. 1984;18(3):431‐437.
    1. Oleptro (trazodone hydrochloride) extended‐release tablets. Full Prescribing Information, Section 12.3 (Pharmacokinetics). . Accessed May 15, 2020.
    1. NDA 19908 clinical pharmacology/biopharmaceutics review, page 11. . Accessed May 15, 2020.
    1. NDA 11792 Clinical Pharmacology Review, Table 1 (page 3; page 337 of PDF), . Accessed May 15, 2020.

Source: PubMed

3
Iratkozz fel