Effects of Mu-Opiate Receptor Gene Polymorphism rs1799971 (A118G) on the Antidepressant and Dissociation Responses in Esketamine Nasal Spray Clinical Trials

Ziad Saad, Derrek Hibar, Maggie Fedgchin, Vanina Popova, Maura L Furey, Jaskaran B Singh, Hartmuth Kolb, Wayne C Drevets, Guang Chen, Ziad Saad, Derrek Hibar, Maggie Fedgchin, Vanina Popova, Maura L Furey, Jaskaran B Singh, Hartmuth Kolb, Wayne C Drevets, Guang Chen

Abstract

Background: At ketamine and esketamine doses at which antidepressant doses are achieved, these agents are relatively selective, noncompetitive, N-methyl-D-aspartate receptor antagonists. However, at substantially higher doses, ketamine has shown mu-opioid receptor (MOR-gene symbol: OPRM1) agonist effects. Preliminary clinical studies showed conflicting results on whether naltrexone, a MOR antagonist, blocks the antidepressant action of ketamine. We examined drug-induced or endogenous MOR involvement in the antidepressant and dissociative responses to esketamine by assessing the effects of a functional single nucleotide polymorphism rs1799971 (A118G) of OPRM1, which is known to alter MOR agonist-mediated responses.

Methods: Participants with treatment-resistant depression from 2 phase III, double-blind, controlled trials of esketamine (or placebo) nasal spray plus an oral antidepressant were genotyped for rs1799971. Participants received the experimental agents twice weekly for 4 weeks. Antidepressant responses were rated using the change in Montgomery-Åsberg Depression Rating Scale (MADRS) score on days 2 and 28 post-dose initiation, and dissociative side effects were assessed using the Clinician-Administered Dissociative-States Scale at 40 minutes post-dose on days 1 and 25.

Results: In the esketamine + antidepressant arm, no significant genotype effect of single nucleotide polymorphism rs1799971 (A118G) on MADRS score reductions was detected on either day 2 or 28. By contrast, in the antidepressant + placebo arm, there was a significant genotype effect on MADRS score reductions on day 2 and a nonsignificant trend on day 28 towards an improvement in depression symptoms in G-allele carriers. No significant genotype effects on dissociative responses were detected.

Conclusions: Variation in rs1799971 (A118G) did not affect the antidepressant response to esketamine + antidepressant. Antidepressant response to antidepressant + placebo was increased in G-allele carriers, compatible with previous reports that release of endorphins/enkephalins may play a role in mediating placebo effect.

Trial registration: NCT02417064 and NCT02418585; www.clinicaltrials.gov.

Keywords: depression; esketamine; ketamine; mu-opioid receptor; polymorphism.

© The Author(s) 2020. Published by Oxford University Press on behalf of CINP.

Figures

Figure 1.
Figure 1.
Effects of OPRM1 single nucleotide polymorphism (SNP) rs1799971 (A118G) alleles on improvements in depression severity, assessed as reductions from baseline in the Montgomery–Åsberg Depression Rating Scale (MADRS) total score on day 2. Black dot, mean; boxed annotation: mean (SD); error bars, 95% mean confidence interval.
Figure 2.
Figure 2.
Effects of OPRM1 single nucleotide polymorphism (SNP) rs1799971 (A118G) alleles on improvements in depression severity, assessed as reductions from baseline in the Montgomery–Åsberg Depression Rating Scale (MADRS) total score on day 28. Black dot, mean; boxed annotation, mean (SD); error bars, 95% mean confidence interval.

References

    1. Aalto S, Ihalainen J, Hirvonen J, Kajander J, Scheinin H, Tanila H, Någren K, Vilkman H, Gustafsson LL, Syvälahti E, Hietala J (2005) Cortical glutamate-dopamine interaction and ketamine-induced psychotic symptoms in man. Psychopharmacology (Berl) 182:375–383.
    1. Aslaksen PM, Forsberg JT, Gjerstad J (2018) The opioid receptor mu 1 (OPRM1) rs1799971 and catechol-O-methyltransferase (COMT) rs4680 as genetic markers for placebo analgesia. Pain 159:2585–2592.
    1. Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L (1998) Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A 95:9608–9613.
    1. Browne CA, Erickson RL, Blendy JA, Lucki I (2017) Genetic variation in the behavioral effects of buprenorphine in female mice derived from a murine model of the OPRM1 A118G polymorphism. Neuropharmacology 117:401–407.
    1. Browne CA, Lucki I (2019) Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 201:51–76.
    1. Chelnokova O, Laeng B, Eikemo M, Riegels J, Løseth G, Maurud H, Willoch F, Leknes S (2014) Rewards of beauty: the opioid system mediates social motivation in humans. Mol Psychiatry 19:746–747.
    1. Choi SW, Lam DMH, Wong SSC, Shiu HHC, Wang AXM, Cheung CW (2017) Effects of single nucleotide polymorphisms on surgical and postsurgical opioid requirements: a systematic review and meta-analysis. Clin J Pain 33:1117–1130.
    1. Colloca L, Wang Y, Martinez PE, Chang YC, Ryan KA, Hodgkinson C, Goldman D, Dorsey SG (2019) OPRM1 rs1799971, COMT rs4680, and FAAH rs324420 genes interact with placebo procedures to induce hypoalgesia. Pain 160:1824–1834.
    1. Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, Thase ME, Winokur A, Van Nueten L, Manji H, Drevets WC (2018) Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: a randomized clinical trial. JAMA Psychiatry 75:139–148.
    1. Daniel M, Martin AD, Carter J (1992) Opiate receptor blockade by naltrexone and mood state after acute physical activity. Br J Sports Med 26:111–115.
    1. Eikemo M, Løseth GE, Johnstone T, Gjerstad J, Willoch F, Leknes S (2016) Sweet taste pleasantness is modulated by morphine and naltrexone. Psychopharmacology (Berl) 233:3711–3723.
    1. Fava M, Freeman MP, Flynn M, Judge H, Hoeppner BB, Cusin C, Ionescu DF, Mathew SJ, Chang LC, Iosifescu DV, Murrough J, Debattista C, Schatzberg AF, Trivedi MH, Jha MK, Sanacora G, Wilkinson ST, Papakostas GI (2018) Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD). Mol Psychiatry Published online 2018 Oct 3. doi: 10.1038/s41380-018-0256-5.
    1. Fedgchin M, Trivedi M, Daly EJ, Melkote R, Lane R, Lim P, Vitagliano D, Blier P, Fava M, Liebowitz M, Ravindran A, Gaillard R, Ameele HVD, Preskorn S, Manji H, Hough D, Drevets WC, Singh JB (2019) Efficacy and safety of fixed-dose esketamine nasal spray combined with a new oral antidepressant in treatment-resistant depression: results of a randomized, double-blind, active-controlled study (TRANSFORM-1). Int J Neuropsychopharmacol 22:616–630.
    1. Garriock HA, Tanowitz M, Kraft JB, Dang VC, Peters EJ, Jenkins GD, Reinalda MS, McGrath PJ, von Zastrow M, Slager SL, Hamilton SP (2010) Association of mu-opioid receptor variants and response to citalopram treatment in major depressive disorder. Am J Psychiatry 167:565–573.
    1. Gassaway MM, Rives ML, Kruegel AC, Javitch JA, Sames D (2014) The atypical antidepressant and neurorestorative agent tianeptine is a μ-opioid receptor agonist. Transl Psychiatry 4:e411.
    1. Henderson-Redmond AN, Yuill MB, Lowe TE, Kline AM, Zee ML, Guindon J, Morgan DJ (2016) Morphine-induced antinociception and reward in “humanized” mice expressing the mu opioid receptor A118G polymorphism. Brain Res Bull 123:5–12.
    1. Hirota K, Sikand KS, Lambert DG (1999) Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells. J Anesth 13:107–109.
    1. Howie B, Fuchsberger C, Stephens M, Marchini J, Abecasis GR (2012) Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nat Genet 44:955–959.
    1. Huang P, Chen C, Mague SD, Blendy JA, Liu-Chen LY (2012) A common single nucleotide polymorphism A118G of the μ opioid receptor alters its N-glycosylation and protein stability. Biochem J 441:379–386.
    1. Hustveit O, Maurset A, Oye I (1995) Interaction of the chiral forms of ketamine with opioid, phencyclidine, sigma and muscarinic receptors. Pharmacol Toxicol 77:355–359.
    1. Inagaki TK, Ray LA, Irwin MR, Way BM, Eisenberger NI (2016) Opioids and social bonding: naltrexone reduces feelings of social connection. Soc Cogn Affect Neurosci 11:728–735.
    1. Janssen Research & Development LLC (2019) Esketamine nasal spray for patients with treatment-resistant depression. Advisory Committee Briefing Document. . Accessed 22 July 2019.
    1. Järvekülg A, Viru A (2002) Opioid receptor blockade eliminates mood effects of aerobic gymnastics. Int J Sports Med 23:155–157.
    1. Kalsi SS, Wood DM, Dargan PI (2011) The epidemiology and patterns of acute and chronic toxicity associated with recreational ketamine use. Emerg Health Threats J 4:7107.
    1. Kokkinou M, Ashok AH, Howes OD (2018) The effects of ketamine on dopaminergic function: meta-analysis and review of the implications for neuropsychiatric disorders. Mol Psychiatry 23:59–69.
    1. Lee MC, Wagner HN Jr, Tanada S, Frost JJ, Bice AN, Dannals RF (1988) Duration of occupancy of opiate receptors by naltrexone. J Nucl Med 29:1207–1211.
    1. Lek M, et al. ; Exome Aggregation Consortium (2016) Analysis of protein-coding genetic variation in 60,706 humans. Nature 536:285–291.
    1. Lutz PE, Kieffer BL (2013) Opioid receptors: distinct roles in mood disorders. Trends Neurosci 36:195–206.
    1. Mague SD, Isiegas C, Huang P, Liu-Chen LY, Lerman C, Blendy JA (2009) Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior. Proc Natl Acad Sci U S A 106:10847–10852.
    1. Mague SD, Port RG, McMullen ME, Carlson GC, Turner JR (2015) Mouse model of OPRM1 (A118G) polymorphism has altered hippocampal function. Neuropharmacology 97:426–435.
    1. Mahmoud S, Thorsell A, Sommer WH, Heilig M, Holgate JK, Bartlett SE, Ruiz-Velasco V (2011) Pharmacological consequence of the A118G μ opioid receptor polymorphism on morphine- and fentanyl-mediated modulation of Ca²⁺ channels in humanized mouse sensory neurons. Anesthesiology 115:1054–1062.
    1. Mallik A, Chanda ML, Levitin DJ (2017) Anhedonia to music and mu-opioids: evidence from the administration of naltrexone. Sci Rep 7:41952.
    1. Marton T, Barnes DE, Wallace A, Woolley JD (2019) Concurrent use of buprenorphine, methadone, or naltrexone does not inhibit ketamine’s antidepressant activity. Biol Psychiatry 85:e75–e76.
    1. Murphy MR, Checkley SA, Seckl JR, Lightman SL (1990) Naloxone inhibits oxytocin release at orgasm in man. J Clin Endocrinol Metab 71:1056–1058.
    1. Murray E, Brouwer S, McCutcheon R, Harmer CJ, Cowen PJ, McCabe C (2014) Opposing neural effects of naltrexone on food reward and aversion: implications for the treatment of obesity. Psychopharmacology (Berl) 231:4323–4335.
    1. Peciña M, Bohnert AS, Sikora M, Avery ET, Langenecker SA, Mickey BJ, Zubieta JK (2015) Association between placebo-activated neural systems and antidepressant responses: neurochemistry of placebo effects in major depression. JAMA Psychiatry 72:1087–1094.
    1. Peciña M, Love T, Stohler CS, Goldman D, Zubieta JK (2015) Effects of the Mu opioid receptor polymorphism (OPRM1 A118G) on pain regulation, placebo effects and associated personality trait measures. Neuropsychopharmacology 40:957–965.
    1. Peciña M, Zubieta JK (2015) Molecular mechanisms of placebo responses in humans. Mol Psychiatry 20:416–423.
    1. Popova V, Daly EJ, Trivedi M, Cooper K, Lane R, Lim P, Mazzucco C, Hough D, Thase ME, Shelton RC, Molero P, Vieta E, Bajbouj M, Manji H, Drevets WC, Singh JB (2019) Efficacy and safety of flexibly dosed esketamine nasal spray combined with a newly initiated oral antidepressant in treatment-resistant depression: a randomized double-blind active-controlled study. Am J Psychiatry 176:428–438.
    1. Rabiner EA, Beaver J, Makwana A, Searle G, Long C, Nathan PJ, Newbould RD, Howard J, Miller SR, Bush MA, Hill S, Reiley R, Passchier J, Gunn RN, Matthews PM, Bullmore ET (2011) Pharmacological differentiation of opioid receptor antagonists by molecular and functional imaging of target occupancy and food reward-related brain activation in humans. Mol Psychiatry 16:826–835, 785.
    1. Samuels BA, Nautiyal KM, Kruegel AC, Levinstein MR, Magalong VM, Gassaway MM, Grinnell SG, Han J, Ansonoff MA, Pintar JE, Javitch JA, Sames D, Hen R (2017) The behavioral effects of the antidepressant tianeptine require the mu-opioid receptor. Neuropsychopharmacology 42:2052–2063.
    1. Sanacora G. (2019) Caution against overinterpreting opiate receptor stimulation as mediating antidepressant effects of ketamine. Am J Psychiatry 176:249.
    1. Schak KM, Vande Voort JL, Johnson EK, Kung S, Leung JG, Rasmussen KG, Palmer BA, Frye MA (2016) Potential risks of poorly monitored ketamine use in depression treatment. Am J Psychiatry 173:215–218.
    1. Shaffer CL, Osgood SM, Smith DL, Liu J, Trapa PE (2014) Enhancing ketamine translational pharmacology via receptor occupancy normalization. Neuropharmacology 86:174–180.
    1. Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G, Tadic A, Sienaert P, Wiegand F, Manji H, Drevets WC, Van Nueten L (2016a) Intravenous esketamine in adult treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol Psychiatry 80:424–431.
    1. Singh JB, Fedgchin M, Daly EJ, De Boer P, Cooper K, Lim P, Pinter C, Murrough JW, Sanacora G, Shelton RC, Kurian B, Winokur A, Fava M, Manji H, Drevets WC, Van Nueten L (2016b) A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am J Psychiatry 173:816–826.
    1. Vollenweider FX, Vontobel P, Oye I, Hell D, Leenders KL (2000) Effects of (S)-ketamine on striatal dopamine: a [11C]raclopride PET study of a model psychosis in humans. J Psychiatr Res 34:35–43.
    1. Weerts EM, McCaul ME, Kuwabara H, Yang X, Xu X, Dannals RF, Frost JJ, Wong DF, Wand GS (2013) Influence of OPRM1 Asn40Asp variant (A118G) on [11C]carfentanil binding potential: preliminary findings in human subjects. Int J Neuropsychopharmacol 16:47–53.
    1. Williams NR, Heifets BD, Blasey C, Sudheimer K, Pannu J, Pankow H, Hawkins J, Birnbaum J, Lyons DM, Rodriguez CI, Schatzberg AF (2018) Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry 175:1205–1215.
    1. Yeomans MR, Gray RW (1996) Selective effects of naltrexone on food pleasantness and intake. Physiol Behav 60:439–446.
    1. Yeomans MR, Gray RW (1997) Effects of naltrexone on food intake and changes in subjective appetite during eating: evidence for opioid involvement in the appetizer effect. Physiol Behav 62:15–21.
    1. Yoon G, Petrakis IL, Krystal JH (2019) Association of combined naltrexone and ketamine with depressive symptoms in a case series of patients with depression and alcohol use disorder. JAMA Psychiatry 76:337–338.
    1. Zanos P, Moaddel R, Morris PJ, Riggs LM, Highland JN, Georgiou P, Pereira EFR, Albuquerque EX, Thomas CJ, Zarate CA Jr, Gould TD (2018) Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharmacol Rev 70:621–660.
    1. Zhang K, Hashimoto K (2019) Lack of opioid system in the antidepressant actions of ketamine. Biol Psychiatry 85:e25–e27.
    1. Zubieta JK, Bueller JA, Jackson LR, Scott DJ, Xu Y, Koeppe RA, Nichols TE, Stohler CS (2005) Placebo effects mediated by endogenous opioid activity on mu-opioid receptors. J Neurosci 25:7754–7762.

Source: PubMed

3
購読する