435
Views
1
CrossRef citations to date
0
Altmetric
Systematic Review

Efficacy, safety, and tolerability of ulotaront (SEP-363856, a trace amine-associated receptor 1 agonist) for the treatment of schizophrenia and other mental disorders: a systematic review of preclinical and clinical trials

, , , ORCID Icon, , , , , , , & show all
Pages 401-415 | Received 14 Sep 2022, Accepted 20 Apr 2023, Published online: 01 May 2023

References

  • James SL, Abate D, Abate KH, et al. 2018. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet. 392(10159): 1789–1858. DOI:10.1016/S0140-67361832279-7
  • World Health Organization - Schizophrenia [Internet]. WHO; 2022 [cited 2022 Jul 15]. Available from: https://www.who.int/news-room/fact-sheets/detail/schizophrenia
  • Chien WT, Leung SF, Yeung FKK, et al. Current approaches to treatments with schizophrenia spectrum disorders, part II: psychosocial interventions and patient-focused perspectives in psychiatric care. Neuropsychiatr Dis Treat. 2013;9:1463–1481.
  • Talaslahti T, Alanen HM, Hakko H, et al. Mortality and causes of death in older patients with schizophrenia. Int J Geriatr Psychiatry. 2012;27(11):1131–1137.
  • Rhee TG, Rosenheck RA. Does improvement in symptoms and quality of life in chronic schizophrenia reduce family caregiver burden. Psychiatry Res. 2019;271:402–404.
  • Chong HY, Teoh SL, DBC W, et al. Global economic burden of schizophrenia: a systematic review. Neuropsychiatr Dis Treat. 2016;12:357–373.
  • Hjorthøj C, Stürup AE, McGrath JJ, et al. Years of potential life lost and life expectancy in schizophrenia: a systematic review and meta-analysis. Lancet. 2017;4(4):295–301.
  • Hoertel N, Rotenberg L, Blanco C, et al. Psychiatric symptoms and quality of life in older adults with schizophrenia spectrum disorder: results from a multicenter study. Eur Arch Psychiatry Clin Neurosci. 2020;270(6):673–688
  • Laruelle M. Schizophrenia: from dopaminergic to glutamatergic interventions. Curr Opin Pharmacol. 2014;14:97–102.
  • Brisch R, Saniotis A, Wolf R, et al. The role of dopamine in schizophrenia from a neurobiological and evolutionary perspective: old fashioned, but still in vogue. Front Psychiatry. 2014;5:47.
  • Shen LH, Liao MH, Tseng YC. Recent advances in imaging of dopaminergic neurons for evaluation of neuropsychiatric disorders. J Biomed Biotechnol. 2012;2012:259349.
  • Stępnicki P, Kondej M, Kaczor AA. Current concepts and treatments of schizophrenia. Molecules. 2018;23(8):2087.
  • Howes OD, McCutcheon R, Owen MJ, et al. The role of genes, stress and dopamine in the development of schizophrenia. Biol Psychiatry. 2017;81(1):9–20.
  • Singam AP, Mamarde A, Behere PB. A single blind comparative clinical study of the effects of chlorpromazine and risperidone on positive and negative symptoms in patients of schizophrenia. Indian J Psychol Med. 2011;33(2):134–140
  • Uno Y, Coyle JT. Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci. 2019;73(5):204–215.
  • Heffernan MLR, Herman LW, Brown S, et al. Ulotaront: a TAAR1 agonist for the treatment of schizophrenia. ACS Med Chem Lett. 2022;13(1):92–98.
  • Cha DS, McIntyre RS. Treatment-emergent adverse events associated with atypical antipsychotics. Expert Opin Pharmacother. 2012;13(11):1587–1598.
  • Kane J, Honigfeld G, Singer J, et al. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry. 1988;45(9):789–796.
  • Leucht S, Corves C, Arbter D, et al. Second-generation versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis. Lancet. 2009;373(9657):31–41.
  • Ricci V, Paggi A, Cristofori E, et al. Efficacy of brexpiprazole for treatment persistent negative symptoms in three schizophrenic patients: a case series. Case Rep Psychiatry. 2022;1(2):100040.
  • Viegas F, Ferreira T, Campos C. Using cariprazine to ameliorate negative symptoms and metabolic side effects of clozapine and paliperidone – clinical cases. Neuropsychiatr Dis Treat. 2022;18:1145–1149.
  • Liang L, Ren X, Xu J, et al. Effects of co-treatment of olanzapine with SEP-363856 in mice models of schizophrenia. Molecules. 2022;27(8):2550
  • Hert MD, Detraux J, van Winkel R, et al. Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nat Rev Endocrinol. 2011;8(2):114–126.
  • Leucht S, Chaimani A, Leucht C, et al. 60 years of placebo-controlled antipsychotic drug trials in acute schizophrenia: meta-regression of predictors of placebo response. Schizophr Res. 2018;201:315–323.
  • Girgis RR, Zoghbi AW, Javitt DC, et al. The past and future of novel, non-dopamine-2 receptor therapeutics for schizophrenia: a critical and comprehensive review. J Psychiatr Res. 2019;108:57–83.
  • Bender AM, Jones CK, Lindsley CW. Classics in Chemical Neuroscience: xanomeline. ACS Chem Neurosci. 2017;8(3):435–443.
  • Brannan SK, Sawchak S, Miller AC, et al. Muscarinic cholinergic receptor agonist and peripheral antagonist for schizophrenia. N Engl J Med. 2021;384(8):717–726.
  • Alnefeesi Y, Tamura JK, Lui LMW, et al. Trace amine-associated receptor 1 (TAAR1): potential application in mood disorders: a systematic review. Neurosci Biobehav Rev. 2021;131:192–210.
  • Dedic N, Dworak H, Zeni C, et al. Therapeutic potential of TAAR1 agonists in schizophrenia: evidence from preclinical models and clinical studies. Int J Mol Sciences. 2021;22(24):13185. DOI:10.3390/ijms222413185
  • Leo D, Mus L, Espinoza S, et al. Taar1-mediated modulation of presynaptic dopaminergic neurotransmission: role of D2 dopamine autoreceptors. Neuropharmacology. 2014;81:283–291.
  • Bradaia A, Trube G, Stalder H, et al. The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system. Proc Natl Acad Sci, USA. 2009;106(47):20081–20086.
  • Rutigliano G, Accorroni A, Zucchi R. The case for TAAR1 As a modulator of central nervous system function. Front Pharmacol. 2018;8:987.
  • Berry MD. Mammalian central nervous system trace amines. Pharmacological amphetamines, physiological neuromodulators. J Neurochem. 2004;90(2):257–271.
  • Revel FG, Moreau JL, Gainetdinov RR, et al. Trace amine-associated receptor 1 partial agonism reveals novel paradigm for neuropsychiatric therapeutics. Biol Pscyhiatry. 2012;72(11):934–942. DOI:10.1016/j.biopsych.2012.05.014
  • Revel FG, Moreau JL, Pouzet B, et al. A new perspective for schizophrenia: tAAR1 agonists reveal antipsychotic- and antidepressant-like activity, improve cognition and control body weight. Mol Psychiatry. 2013;18(5):543–556.
  • Koblan KS, Kent J, Hopkins SC, et al. A non-D2-receptor-binding drug for the treatment of schizophrenia. N Engl J Med. 2020;382(16):1497–1506.
  • Correll CU, Koblan KS, Hopkins SC, et al. Safety and effectiveness of ulotaront (SEP-363856) in schizophrenia: results of a 6-month, open-label extension study. NPJ Schizophr. 2021;7(1):63.
  • Page MJ, Moher D, Bossuyt PM, et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;372:n160.
  • Higgins JP, Altman DG. Assessing risk of bias in included studies. In: Higgins JP Green S, editors. Cochrane handbook for systematic reviews of interventions. John Wiley and Sons; 2008. p. 187–241. DOI:10.1002/9780470712184.ch8.
  • Higgins JP, Altman DG, Gøtzsche PC, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.
  • Haddaway NR, Page MJ, Pritchard CC, et al. PRISMA2020: an R package and shiny app for producing prisma 2020-compliant flow diagrams, with interactivity for optimised digital transparency and open synthesis. Campbell Systematic Reviews. 2020;18:e1230. DOI:10.1002/cl2.1230
  • Hopkins SC, Dedic N, Koblan KS. Effect of TAAR1/5-HT1A agonist SEP-363856 on REM sleep in humans. Transl Psychiatry. 2021;11(1):228.
  • Galluppi GR, Polhamus DG, Fisher JM, et al. Population pharmacokinetic analysis of ulotaront in subjects with schizophrenia. CPT Pharmacometrics Syst Pharmacol. 2021;10(10):1245–1254.
  • Hopkins SC, Ogirala A, Worden M, et al. Depicting safety profile of TAAR1 agonist ulotaront relative to reactions anticipated for a dopamine d2-based pharmacological class in FAERS. Clin Drug Investig. 2021;41(12):1067–1073.
  • Begni V, Sanson A, Luoni A, et al. Towards novel treatments for schizophrenia: molecular and behavioral signatures of the psychotropic agent SEP-363856. Int J Mol Sci. 2021;22(8):4119
  • Dedic N, Jones PG, Hopkins SC, et al. SEP-363856, a novel psychotropic agent with a unique, Non-D2 receptor mechanism of action. J Pharmacol Exp Ther. 2019;371(1):1–14.
  • Xiao G, Chen YL, Dedic N, et al. In Vitro ADME And preclinical pharmacokinetics of ulotaront, a TAAR1/5-HT1A receptor agonist for the treatment of schizophrenia. Pharm Res. 2022;39:837–850.
  • Kokkinou M, Irvine EE, Bonsall DR, et al. Reproducing the dopamine pathophysiology of schizophrenia and approaches to ameliorate it: a translational imaging study with ketamine. Mol Psychiatry. 2021;26(6):2562–2575.
  • Nahar L, Delacroix BM, Nam HW. The role of parvalbumin interneurons in neurotransmitter balance and neurological disease. Front Psychiatry. 2021;12:679960.
  • Grace AA. Dysregulation of the dopamine system in the pathophysiology of schizophrenia and depression. Nat Rev Neurosci. 2016;17(8):524–532.
  • Hofstetter JR, Lysaker PH, Mayeda AR. Quality of sleep in patients with schizophrenia is associated with quality of life and coping. BMC Psyfchiatry. 2005;5:13.
  • Wang YQ, Li R, Zhang MQ, et al. The neurobiological mechanisms and treatments of REM sleep disturbances in depression. Curr Neuropharmacol. 2015;13(4):543–553.
  • Estrada-Prat X, Álvarez-Guerrico I, Batlle-Vila S, et al. Sleep alterations in pediatric bipolar disorder versus attention deficit disorder. Psychiatry Res. 2019;275:39–45.
  • Kurian SM, Le-Niculescu H, Patel SD, et al. Identification of blood biomarkers for psychosis using convergent functional genomics. Mol Psychiatry. 2011;16(1):37–58.
  • Cattane N, Minelli A, Milanesi E, et al. Altered gene expression in schizophrenia: findings from transcriptional signatures in fibroblasts and blood. PLoS ONE. 2015;10(2):20116686. DOI:10.1371/journal.pone.0116686
  • Gallo FT, Katche C, Morici JF, et al. Immediate early genes, memory and psychiatric disorders: focus on c-Fos, Egr1 and Arc. Front Behav Neurosci. 2018;12:79.
  • Czobor P, Sebe B, Acsai K, et al. What is the minimum clinically important change in negative symptoms of schizophrenia? PANSS based post-hoc analyses of a phase III clinical trial. Front Psychiatry. 2022;13:816339.
  • Espinoza S, Salahpour A, Masri B, et al. Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor. Mol Pharmacol. 2011;80(3):416–425.
  • Harmeier A, Obermueller S, Meyer CA, et al. Trace amine-associated receptor 1 activation silences GSK3β signaling of TAAR1 and D2R heteromers. Eur Neuropsychopharmacol. 2015;25(11):2049–2061.
  • Revel FG, Moreau J-L, Gainetdinov RR, et al. TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc Natl Acad Sci, USA. 2011;108(20):8485–8490.
  • Beaulieu J-M, Gainetdinov RR, Caron MG. Akt/GSK3 signaling in the action of psychotropic drugs. Annu Rev Pharmacol Toxicol. 2009;49:327–347.
  • Urs NM, Snyder JC, Jacobsen JPR, et al. Deletion of GSK3β in D2R-expressing neurons reveals distinct roles for β-arrestin signaling in antipsychotic and lithium action. Proc Natl Acad Sci U S A. 2012;109(50):20732–20737.
  • Terzić T, Kastelic M, Dolžan V, et al. Influence of 5-HT1A and 5-HTTLPR genetic variants on the schizophrenia symptoms and occurrence of treatment-resistant schizophrenia. Neuropsychiatr Dis Treat. 2015;11:453–459.
  • Bantick RA, Deakin JF, Grasby PM. The 5-HT1A receptor in schizophrenia: a promising target for novel atypical neuroleptics? J Psychopharmacol. 2001;15(1):37–46.
  • Puig MV, Artigas F, Celada P. Modulation of the activity of pyramidal neurons in rat prefrontal cortex by raphe stimulation in vivo: involvement of serotonin and GABA. Cereb Cortex. 2005;15(1):1–14.
  • Euston DR, Gruber AJ, McNaughton BL. The role of medial prefrontal cortex in memory and decision making. Neuron. 2012;76(6):1057–1070.
  • Deng C. Effects of antipsychotic medications on appetite, weight, and insulin resistance. Endocrinol Metab Clin North Am. 2013;42(3):545–563.
  • Burghardt KJ, Seyoum B, Mallisho A, et al. Atypical antipsychotics, insulin resistance and weight; a meta-analysis of healthy volunteer studies. Prog Neuro Psychopharmacol Biol Psychiatry. 2018;83:55–63.
  • Dayabandara M, Hanwella R, Ratnatunga S, et al. Antipsychotic-associated weight gain: management strategies and impact on patient adherence. Neuropsychiatr Dis Treat. 2017;13:2231–2241.
  • Kotorki P, Pelka P, Leotsakou C, et al. Reversal of symptomatic antipsychotic-induced hyperprolactinemia with addition of aripiprazole. Ann Gen Psychiatry. 2010;9:S164.
  • Latif Z, Jabbar F, Kelly BD. Clozapine and blood dyscrasia. The Psych. 2011;35(1):27–29.
  • Sunovion. A clinical study to investigate the effect of an investigational drug as an added medication to an antipsychotic, in adults with schizophrenia, as measured Positron Emission Tomography (PET) imaging. ClinicalTrials.Gov identifier: nCT04038957. Updated Jul 11, 2022. Accessed Aug 13, 2022. https://clinicaltrials.gov/show/NCT04038957
  • Sunovion. A clinical study to determine whether an investigational medication (sep-363856) changes how long it takes for food to move through the stomach into the small intestine in patients with schizophrenia. the study will help to understand any effect sep-363856 may have on blood sugar levels after a meal. ClinicalTrials.Gov identifier: nCT05402111. Updated Aug 2, 2022. Accessed Aug 13, 2022. https://clinicaltrials.gov/show/NCT05402111
  • Sunovion. A clinical study that will look at an investigational medication, SEP-363856 (Called “Study Medication”) in patients with schizophrenia and assess whether it changes: how the body processes (uses) glucose (blood sugar), how much insulin the pancreas can make. ClinicalTrials.gov identifier: NCT05463770. Updated Jul 19, 2022. Accessed Aug 13, 2022. https://clinicaltrials.gov/show/NCT05463770
  • Sumitomo Pharma Co., Ltd. A clinical trial to evaluate the efficacy and safety of SEP-363856 in acutely psychotic people with schizophrenia, followed by an open-label extension phase. ClinicalTrials.gov identifier: NCT04825860. Updated Apr 12, 2022. Accessed Aug 14, 2022. https://clinicaltrials.gov/show/NCT04825860
  • Sunovion. A study to evaluate the efficacy, safety and tolerability of SEP-363856 in subjects with parkinson’s disease psychosis. ClinicalTrials.Gov identifier: nCT02969369. Updated Aug 4, 2021. Accessed Aug 14, 2022. https://clinicaltrials.gov/show/NCT02969369
  • Sumitomo Pharma Co., Ltd. A clinical trial to evaluate the long-term safety and tolerability of SEP-363856 in patients with schizophrenia in Japan. ClinicalTrials.gov identifier: NCT05359081. Updated Jul 7, 2022. Accessed Aug 14, 2022. https://clinicaltrials.gov/show/NCT05359081
  • Sunovion. A clinical study to evaluate the long-term safety and tolerability of an investigational drug in people with schizophrenia. ClinicalTrials.Gov identifier: nCT04109950. Updated Aug 2, 2022. Accessed Aug 14, 2022. https://clinicaltrials.gov/show/NCT04109950
  • Sunovion. A clinical trial that will study the efficacy and safety of an investigational drug in acutely psychotic people with schizophrenia. ClinicalTrials.Gov identifier: nCT04092686. Updated Aug 2, 2022. Accessed Aug 15, 2022. https://clinicaltrials.gov/show/NCT04092686
  • Sunovion. A clinical trial to study the efficacy and safety of an investigational drug in acutely psychotic people with schizophrenia. ClinicalTrials.Gov identifier: nCT04072354. Updated Aug 2, 2022. Accessed Aug 15, 2022. https://clinicaltrials.gov/show/NCT04072354
  • Sunovion. A study of the long-term safety and tolerability of an investigational drug in people with schizophrenia. ClinicalTrials.gov identifier: NCT04115319. Updated Feb 17, 2022. Accessed Aug 15, 2022. https://clinicaltrials.gov/show/NCT04115319
  • Zhang Y, Gruber R. Can slow-wave sleep enhancement improve memory? a review of current approaches and cognitive outcomes. Yale J Biol Med. 2019;92(1):63–80.
  • Cirillo MA, Seidman LJ. Verbal declarative memory dysfunction in schizophrenia: from clinical assessment to genetics and brain mechanisms. Neuropsychol rev. 2003;13(2):43–77.
  • Iorio C, Pacitti F, Rossi A, et al. Declarative memory impairment and emotional bias in recurrent depression with a seasonal pattern: the interplay between emotion and cognition in seasonal affective disorder. Brain Sci. 2022;12(10):1352.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.