Central nervous system-related safety and tolerability of add-on ketamine to antidepressant medication in treatment-resistant depression: focus on the unique safety profile of bipolar depression

Adam Włodarczyk, Wiesław J Cubała, Maria Gałuszko-Węgielnik, Joanna Szarmach, Adam Włodarczyk, Wiesław J Cubała, Maria Gałuszko-Węgielnik, Joanna Szarmach

Abstract

Background: There is evidence supporting the use of ketamine in treatment-resistant depression (TRD). However, there are some safety and tolerability concerns associated with ketamine. This study aimed to investigate ketamine's safety and tolerability to the central nervous system and to assess the relationship between dissociative symptomology and psychometric outcomes during and after intravenous ketamine treatment concurrent with treatment by varying psychotropic medications in treatment-refractory inpatients with major depressive disorder (MDD) and bipolar disorder (BP).

Methods: A total of 49 patients with MDD and BP were included in this study. The subjects were administered ketamine and were assessed for changes using an observational protocol.

Results: No antidepressants were associated with psychomimetic symptomatology except for citalopram (p = 0.019). Patients treated with citalopram showed a higher intensity of psychomimetic symptomatology. The use of classic mood-stabilizers was significantly associated with an increase in psychomimetic symptomatology according to the Brief Psychiatric Rating Scale (BPRS; lamotrigine p = 0.009, valproate p = 0.048, lithium p = 0.012). No sequelae were observed.

Conclusions: Despite the limitations that this study may be underpowered due to the small sample size, the sample consisted of a heterogeneous TRD population in a single site, and there no blinding of who underwent only acute ketamine administration, our observations indicate ketamine use requires close safety and tolerability monitoring with regards to psychomimetic and dissociative symptoms in TRD-BP and careful management for MDD patients.ClinicalTrials.gov identifier: NCT04226963.

Keywords: dissociation; ketamine; psychosis; safety; tolerability; treatment-resistant depression.

Conflict of interest statement

Conflict of interest statement: W has received research support from Actavis, Eli Lilly, Minerva Neurosciences, Sunovion Pharmaceuticals, KCR, Janssen, Otsuka, Apodemus, Cortexyme, Acadia. WJC has received research support from Actavis, Alkermes, Allergan, Auspex, Biogen, Bristol-Myers Squibb, Cephalon, Eli Lilly, Ferrier, Forest Laboratories, Gedeon Richter, GW Pharmaceuticals, Janssen, KCR, Lundbeck, Orion, Otsuka, Sanofi, and Servier; he has served on speakers bureaus for Adamed, Angelini, AstraZeneca, Bristol-Myers Squibb, Celon, GlaxoSmithKline, Janssen, Krka, Lekam, Lundbeck, Novartis, Orion, Pfizer, Polfa Tarchomin, Sanofi, Servier, and Zentiva; and he has served as a consultant for GW Pharmaceuticals, Janssen, KCR, Quintiles, and Roche. MG-W has received research support from Janssen, Servier, Alkermes, KCR, Lilly, Biogen, Celon; JS has received research support from Actavis, Eli Lilly, Minerva Neurosciences, Sunovion Pharmaceuticals, KCR, Janssen, Otsuka, Apodemus, Cortexyme, Acadia.

© The Author(s), 2021.

Figures

Figure 1.
Figure 1.
Means and standard errors for the BPRS scores were measured after subsequent ketamine infusions in groups with and without lamotrigine coexisting treatment. *Significantly higher BPRS scores were observed between patients who were being treated with lamotrigine (n = 7) and those who were not (n = 42), with regards to BPRS score after the eighth infusion. A detailed analysis of the results showed that the BPRS score was higher after the fifth infusion compared with that after the seventh infusion. The differences between the remaining measurements were not significant (p > 0.05). BPRS, Brief Psychiatric Rating Scale.
Figure 2.
Figure 2.
Means and standard errors for the BPRS scores were measured after subsequent ketamine infusions in groups with and without citalopram coexisting treatment. *Significantly higher scores in BPRS were observed, after the second infusion between patients who were taking citalopram (n = 4) and those who were not (n = 45). BPRS, Brief Psychiatric Rating Scale.
Figure 3.
Figure 3.
Means and standard errors for the BPRS scores were measured after subsequent ketamine infusions in groups with and without coexisting lithium treatment. *Significantly higher BPRS scores were observed between patients being treated with lithium (n = 5) and those who were not (n = 44), after the sixth infusion, and after the eighth infusion. BPRS, Brief Psychiatric Rating Scale.
Figure 4.
Figure 4.
Means and standard errors for the BPRS scores were measured after subsequent ketamine infusions in groups with and without valproate coexisting treatment. *Significantly higher BPRS scores were observed between patients being treated with valproate (n = 9) and those who were not (n = 40) following the sixth infusion, and after the eighth infusion. BPRS, Brief Psychiatric Rating Scale.

References

    1. Berman RM, Cappiello A, Anand A, et al.. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000; 47: 351–354.
    1. Correia-Melo FS, Argolo FC, Araújo-de-Freitas L, et al.. Rapid infusion of esketamine for unipolar and bipolar depression: a retrospective chart review. Neuropsychiatr Dis Treat 2017; 13: 1627–1632.
    1. Diazgranados N, Ibrahim L, Brutsche NE, et al.. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment- resistant bipolar depression. Arch Gen Psychiatry 2010; 67: 793–802.
    1. McCloud TL, Caddy C, Jochim J, et al.. Ketamine and other glutamate receptor modulators for depression in bipolar disorder in adults. Cochrane Database Syst Rev 2015; 9: CD011611.
    1. Short B, Fong J, Galvez V, et al.. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatry 2018; 5: 65–78.
    1. Wan LB, Levitch CF, Perez AM, et al.. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry 2015; 76: 247–252.
    1. Włodarczyk A, Cubała WJ, Szarmach J, et al.. Short-term ketamine administration in treatment-resistant depression patients: focus on adverse effects on the central nervous system. Psychiatr Danub 2019; 31: 530–533.
    1. McAllister-Williams RH, Arangoc C, Blierd P, et al.. The identification, assessment and management of difficult-to-treat depression: an international consensus statement. J Affect Disord 2020; 267: 264–282.
    1. Słupski J, Cubała WJ, Górska N, et al.. Copper and anti-anhedonic effect of ketamine in treatment-resistant depression. Med Hypotheses 2020; 144: 110268.
    1. Szarmach J, Cubała WJ, Włodarczyk A, et al.. Metabolic risk factors and cardiovascular safety in ketamine use for treatment resistant depression. Neuropsychiatr Dis Treat 2020; 16: 2539–2551.
    1. Poon SH, Sim K, Baldessarini RJ. Pharmacological approaches for treatment-resistant bipolar disorder. Curr Neuropharmacol 2015; 13: 592–604.
    1. Bremner JD, Krystal JH, Putnam FW, et al.. Measurement of dissociative states with the Clinician-Administered Dissociative States Scale (CADSS). J Trauma Stress 1998; 11: 125–136.
    1. Włodarczyk A, Cubała WJ. Safety and tolerability of ketamine use in treatment-resistant bipolar depression patients with regard to central nervous system symptomatology: literature review and analysis. Medicina 2020; 56: 67.
    1. Trivedi MH, Corey-Lisle PK, Guo Z, et al.. Remission, response without remission, and nonresponse in major depressive disorder: impact on functioning. Int Clin Psychopharmacol 2009; 24: 133–138.
    1. Young T. What exactly is a mood stabilizer? J Psychiatry Neurosci 2004; 29: 87–88.
    1. Fava M, Freeman MP, Flynn M, et al.. Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD). Mol Psychiatry 2018; 25: 1592–1603.
    1. Lapidus KAB, Levitch CF, Perez AM, et al.. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry 2014; 76: 970–976.
    1. Luckenbaugh DA, Niciu MJ, Ionescu DF, et al.. Do the dissociative side effects of ketamine mediate its antidepressant effects? J Affect Disord 2014; 159: 56–61.
    1. Phillips JL, Norris S, Talbot J, et al.. Single, repeated, and maintenance ketamine infusions for treatment-resistant depression: a randomized controlled trial. Am J Psychiatry 2019; 176: 401–409.
    1. Sos P, Klirova M, Novak T, et al.. Relationship of ketamine’s antidepressant and psychotomimetic effects in unipolar depression. Neuro Endocrinol Lett 2013; 34: 287–293.
    1. Valentine GW, Mason GF, Gomez R, et al.. The antidepressant effect of ketamine is not associated with changes in occipital amino acid neurotransmitter content as measured by [(1)H]-MRS. Psychiatry Res 2011; 191: 122–127.
    1. Vidal S, Gex-Fabry M, Bancila V, et al.. Efficacy and safety of a rapid intravenous injection of ketamine 0.5mg/kg in treatment-resistant major depression: an open 4-Week longitudinal study. J Clin Psychopharmacol 2018; 38: 590–597.
    1. Daly EJ, Trivedi MH, Janik A, et al.. Efficacy of esketamine nasal spray plus oral antidepressant treatment for relapse prevention in patients with treatment-resistant depression. JAMA Psychiatry 2019; 76: 893–903.
    1. Popova V, Daly EJ, Trivedi M, et al.. 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 2019; 176: 428–438.
    1. Schmechtig A, Lees J, Perkins A, et al.. The effects of ketamine and risperidone on eye movement control in healthy volunteers. Transl Psychiatry 2013; 3: e334.
    1. Aalto S, Ihalainen J, Hirvonen J, et al.. Cortical glutamate–dopamine interaction and ketamine-induced psychotic symptoms in man. Psychopharmacology (Berl) 2005; 182: 375–383.
    1. Kegeles LS, Martinez D, Kochan LD, et al.. NMDA antagonist effects on striatal dopamine release: positron emission tomography studies in humans. Synapse 2002; 43: 19–29.
    1. Vernaleken I, Klomp M, Moeller O, et al.. Vulnerability to psychotogenic effects of ketamine is associated with elevated D2/3-receptor availability. Int J Neuropsychopharmacol 2013; 16: 745–754.
    1. Dwyer JM, Duman RS. Activation of mammalian target of rapamycin and synaptogenesis: role in the actions of rapid-acting antidepressants. Biol Psychiatry 2013; 73: 1189–1198.
    1. Niciu MJ, Shovestul BJ, Jaso BA, et al.. Features of dissociation differentially predict antidepressant response to ketamine in treatment-resistant depression. J Affect Disord 2018; 232: 310–315.

Source: PubMed

3
Prenumerera