Clozapine for Treating Treatment-Resistant Schizophrenia: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Clozapine is listed as one of the most effective antipsychotics and has been approved for treating treatment-resistant schizophrenia (TRS); however, several type A and B adverse reactions, including weight gain, metabolic complications, cardiotoxicity, convulsions, and discontinuation syndromes, exist. The critical mechanisms of clinical efficacy for schizophrenia, TRS, and adverse reactions of clozapine have not been elucidated. The GABA isomer L-β-aminoisobutyric acid (L-BAIBA), a protective myokine in the peripheral organs, was identified as a candidate novel transmission modulator in the central nervous system (CNS). L-BAIBA activates adenosine monophosphate-activated protein kinase (AMPK) signalling in both the peripheral organs and CNS. Activated AMPK signalling in peripheral organs is an established major target for treating insulin-resistant diabetes, whereas activated AMPK signalling in the hypothalamus contributes to the pathophysiology of weight gain and metabolic disturbances. Clozapine increases L-BAIBA synthesis in the hypothalamus. In addition, the various functions of L-BAIBA in the CNS have been elucidated, including as an activator of GABA-B and group-III metabotropic glutamate (III-mGlu) receptors. Considering the expressions of GABA-B and III-mGlu receptors (localised in the presynaptic regions), the activation of GABA-B and III-mGlu receptors can explain the distinct therapeutic advantages of clozapine in schizophrenia or TRS associated with N-methyl-D-aspartate (NMDA) receptor disturbance compared with other atypical antipsychotics via the inhibition of the persistent tonic hyperactivation of thalamocortical glutamatergic transmission in the prefrontal cortex. L-BAIBA has also been identified as a gliotransmitter, and a detailed exploration of the function of L-BAIBA in tripartite synaptic transmission can further elucidate the pathophysiology of effectiveness for treating TRS and/or specific adverse reactions of clozapine.

  • clozapine L-β-aminoisobutyric acid
  • treatment-resistant schizophrenia
  • metabolic complication
  • thalamocortical pathway

1. Introduction

Traditionally, more than 30% of patients with schizophrenia spectrum are considered to suffer from treatment-resistant schizophrenia (TRS) [1][2][3]. Clozapine is evaluated as the most effective antipsychotic agent for TRS since 30–60% of patients with TRS respond to clozapine medication [4][5][6]. Therefore, clozapine is currently the only approved antipsychotic for TRS treatment [7]. In fact, several guidelines recommend initiating treatment with clozapine for patients with TRS [8][9][10]. Furthermore, systematic reviews and meta-analyses have demonstrated that clozapine is associated with lower hospitalisation rates, lower overall discontinuation rates, and better overall symptom outcomes compared with other atypical antipsychotics [11][12][13].
All antipsychotics approved for the treatment of schizophrenia are antagonists of the dopamine D2 receptor at therapeutically relevant concentrations [14][15]. The introduction of clozapine in the 1970s marked a significant turning point in the pharmacotherapy of schizophrenia. As an alternative, clozapine minimised the risk of extrapyramidal symptoms, such as antipsychotic-induced parkinsonism and tardive dyskinesia, while demonstrating excellent efficacy for both positive and negative symptoms of schizophrenia [16][17]. Based on these clinical advantages of clozapine, receptor-binding profile screenings have contributed to the development of several second-generation antipsychotics (atypical antipsychotics) that share pharmacological characteristics distinct from the preceding first-generation antipsychotics (typical antipsychotics) [16][18]. It is well known that olanzapine has a similar receptor-binding profile to clozapine, except for the 5-HT7 receptor [19]; however, the specific effectiveness of clozapine for treating TRS suggests the pathophysiology of clozapine may involve molecules other than monoamine receptors.
Most atypical antipsychotics had been developed by exploring molecules that have similar receptor-binding profiles to clozapine that are distinct from the preceding typical antipsychotics, such as having a relatively lower binding affinity to the dopamine D2 receptor and higher affinity to serotonin 5-HT2A receptors [20]. Therefore, the pathophysiological hypothesis proposed to distinguish between typical and atypical antipsychotics, having a relatively low affinity to the D2 receptor and relatively high affinity to the 5-HT2A receptor, cannot account for the distinct therapeutic advantages of clozapine against other atypical antipsychotics.
Clozapine (CLZ), lurasidone (LUR), aripiprazole (APZ), brexpiprazole (Brex), olanzapine (OLZ), quetiapine (QTP), risperidone (RIS), zotepine (ZTP), and haloperidol (HPD) against serotonin (5-HT) type 1A (5-HT1A), type 2A (5-HT2A), type 2C (5-HT2C), and type 7 (5-HT7) receptors, histamine H1 (H1) receptor, and dopamine receptors type 1 (D1) and 2 (D2). Data are equilibrium constant (Ki) values (nM).

2. Clozapine-Induced Metabolic Complications

Weight gain is the most prevalent adverse reaction of atypical antipsychotic medications. Weight gain induced by atypical antipsychotics usually occurs during the early stages of antipsychotic treatment (within the first year), with an increase of 7% over baseline weight observed in approximately two-thirds of antipsychotic-treated patients [21][22]. Diabetes treatment in patients treated with clozapine is manageable by following current diabetes treatment guidelines [23][24]. Thus, a history of diabetes in TRS patients does not constitute a contraindication to clozapine medication [25]. Among pharmacological interventions, metformin has an excellent safety profile and is the most effective for weight gain stabilisation [26][27][28]. Topiramate has also been demonstrated to be as effective as metformin in suppressive effects on clozapine-induced weight gain [29][30]. Glucagon-like peptide-1 (GLP1) receptor agonists have been recently shown to effectively mitigate clozapine-induced metabolic disturbances [31]. However, weight gain induced by antipsychotics other than clozapine, including olanzapine and quetiapine, reaches a plateau within the therapeutic dose range, whereas the unique features of weight gain with clozapine indicate a linear dose-dependent manner ranging from therapeutic to supratherapeutic doses [32]. This specific linear dose-dependent weight gain induced by clozapine indicates that different mechanisms might underlie the weight gain induced by other antipsychotics.
Atypical antipsychotic-induced metabolic complications have been considered to be related to the inhibition of the histamine H1 and serotonin 5-HT2A receptors, which leads to the disturbance of energy regulation systems in the hypothalamus [33][34]. The inhibition of the H1 and 5-HT2A receptors suppresses the synthesis of inositol trisphosphate (IP3), which activates the calcium-induced calcium-releasing system (CICR) via the enhancement of the IP3 receptor (Figure 1) [35][36]. The elevation in intracellular calcium ion levels activates adenosine triphosphate (ATP) synthase, leading to an increase in ATP and/or a decrease in adenosine monophosphate (AMP) levels (Figure 1) [34][36][37]. Therefore, CICR suppression induced by H1 and 5-HT2A receptor inhibition secondarily increases intracellular AMP levels, leading to the activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) (Figure 1) [33][34][37][38]. This hypothesis has been supported by the clinical findings on high-affinity H1 and 5-HT2A receptor antagonistic antipsychotics, including zotepine, quetiapine, olanzapine, and clozapine listed as being high-risk for metabolic complications [32]. However, the activation of AMPK in the peripheral organs is one of the major therapeutic targets for insulin-resistant diabetes [28][39][40], whereas the activation of AMPK signalling in the hypothalamus increases feeding and reduces energy expenditure in the body [40].
Figure 1. Schematic presentation of hypothalamic signalling associated with traditional hypothesis regarding the mechanisms of antipsychotic-induced metabolic complications and weight gain. Red and blue arrows indicate activation and inhibition, respectively. Abbreviations: H1 receptor—histamine H1 receptor, 5-HT2A receptor—serotonin 5-HT2A receptor, IP3—of inositol trisphosphate, CICR—Ca2+-induced Ca2+-releasing system, ATP—adenosine triphosphate, AMP—adenosine monophosphate, and AMPK—AMP-activated protein kinase.
Chronic administration of therapeutically relevant doses of clozapine, quetiapine, brexpiprazole, and lurasidone decreased IP3 synthesis, and increased AMP levels in the rat hypothalamus [38][41][42]. However, contrary to expectations, AMPK signallings were activated and unaffected by high-risk (clozapine and quetiapine) and low-risk (brexpiprazole and lurasidone) antipsychotics for weight gain, respectively [38][41][43][44][45]. Both clozapine and quetiapine are high-affinity antagonists of the histamine H1 receptor and the 5-HT2A receptor, whereas brexpiprazole and lurasidone are high-affinity 5-HT2A receptors but have low binding affinity to the H1 receptor [33][46]. Therefore, enhanced intra-hypothalamic AMPK signalling plays fundamental roles in antipsychotic-induced metabolic complications and weight gain, but decreasing IP3 with increasing AMP levels via inhibition of H1 and/or 5-HT2A receptors alone cannot explain the pathophysiology of antipsychotic-induced weight gain. Similar to clozapine, an H1 and 5-HT2A high-affinity atypical antipsychotic agent, olanzapine, which was established to also be a high-risk antipsychotic for weight gain, decreased IP3 synthesis [47][48]; however, olanzapine has been reported to enhance [34][49][50] and suppress [51][52] hypothalamic AMPK signalling with contradictory results.

3. Clozapine and TRS

3.1. Efficacy of Clozapine in TRS

TRS is internationally defined by the Treatment Response and Resistance in Psychosis (TRRIP) Working Group and includes the following aspects: the presence of persistent symptoms—including positive and negative symptoms, and cognitive impairment—over at least 12 weeks of at least moderate severity caused by moderate levels of functional impairments [53]. Symptom classifications and thresholds should be based on standardised and validated clinical rating scales. Insufficient response to medication with at least two different antipsychotic medications, with a minimum treatment duration of twelve weeks (six weeks for each antipsychotic agent). This corresponds to a minimum dose equivalent to 600 mg per day of chlorpromazine. Confirmation of adequate treatment adherence is defined as the patient having taken at least 80% of the prescribed dose. To achieve this, at least two methods should be employed, including counting tablets, patient and caregiver reports, and review of medical records and documentation. Additionally, plasma drug concentrations should be monitored at least once for each antipsychotic agent [53][54].
Incontrovertible evidence supports the superior efficacy of clozapine compared with other atypical antipsychotics in improving positive symptoms and global psychopathology in TRS [5][13][55][56]. Considering the lack of evidence to support using polypharmacy of antipsychotics other than clozapine that is as effective as clozapine, the efficacy of clozapine in TRS is evaluated as being more robust [57]. Furthermore, patients treated with clozapine have also shown improvements in treatment adherence, resulting in decreased rehospitalisation rates [57][58].

3.2. Candidate Pathophysiology of TRS

Some research groups have emphasised the importance of distinguishing between primary and secondary TRS: primary TRS already presents with antipsychotic-resistant clinical features at the onset of the schizophrenia spectrum, whereas secondary TRS develops at later stages of the schizophrenia spectrum after an initial adequate response to antipsychotics [59][60][61]. Dopaminergic supersensitivity induced by consecutive exposure to antipsychotics has been speculated as a candidate mechanism of secondary TRS [62]. Persistent exposure to antipsychotics upregulates postsynaptic D2 receptors, leading to further psychotic exacerbation [62]. The estimated overall response rate to antipsychotic medications ranges from 40% to 60% [63][64]. The response rate to antipsychotic medication in antipsychotic-naïve patients is estimated to be approximately 75%; however, the response rate in a second trial using antipsychotic medications other than clozapine was considerably lower, ranging from 20% to 45% [65][66]. Response rates to clozapine have been reported to be maximally up to 80% when treatment is initiated within the first 2–3 years after resistance is established [64][65][67]. With subsequent initiation of clozapine medication, the response rate might be as low as 30% [64]. The efficacy of clozapine against TRS is significant compared with other antipsychotics but decreases depending on the duration of antipsychotic exposure, which is similar to other antipsychotics. These clinical findings regarding duration-dependent resistance at least partially support the dopaminergic supersensitivity hypothesis [62].
The specific features of clozapine, such as low affinity and rapid dissociation from D2 receptors, are considered to be candidate mechanisms via which clozapine-induced D2 receptor supersensitivity is less than that of other antipsychotics [20][46][68][69][70]. However, several line studies have demonstrated that the dissociation rate of clozapine from D2 receptors is not significantly faster compared with the rates of other antipsychotics, such as quetiapine, amisulpride, remoxipride, and sulpiride [20][71][72][73]. These pharmacodynamic findings suggest that the efficacy of clozapine in secondary TRS cannot be solely explained by either its low affinity or rapid dissociation from D2 receptors, even if the pathophysiology of TRS involves D2 receptor supersensitivity.

3.3. Candidate Targets of Clozapine Other Than Monoamine Receptors

Although schizophrenia is commonly speculated to be a pathophysiologically contiguous spectrum between treatment-responsive schizophrenia and TRS, several findings suggest that TRS might be a subtype with extreme characteristics from the perspective of neurodevelopmental disorders [74][75]. In other words, there are possibly two subtypes of pathophysiology of TRS, one being secondary treatment resistance due to long-term exposure to antipsychotic drugs, and the other already developing as TRS during the onset period. Approximately 70–80% of patients with TRS have been reported to present antipsychotic-resistant clinical features from the first episode [74][75]. Furthermore, predictors of antipsychotic resistance in schizophrenia are similar to the clinical features of ‘neurodevelopmental’ schizophrenia, such as being male, being of a younger age at onset, poor premorbid adjustment, and a longer duration of untreated illness [76][77]. So far, various studies have revealed impairments in cognitive components, such as sensorimotor function, attention, working memory, visuospatial processing, verbal intelligence, and memory in TRS patients compared with treatment-responsive schizophrenia [78][79][80][81]. These cognitive impairments are more suggestive of impaired function of glutamate transmission (via thalamocortical pathways) than monoamine transmission (via the mesolimbic and mesocortical systems). These cognitive impairment features of TRS suggest it may be caused by dysfunction of glutamatergic transmission (via thalamocortical pathways) rather than monoaminergic transmission (via mesolimbic and mesocortical pathways) [15][43][68][82][83][84][85].
Quantitative reviews of mRNA and protein expression of N-methyl-D-aspartate glutamate receptor (NMDA-R) in post-mortem studies have demonstrated that both mRNA and protein expression of the NR1 subunit of NMDA-R in the prefrontal cortex decreased in patients with schizophrenia compared with healthy volunteers [86]. mGluR5 (I-mGluR) signalling in the dorsolateral prefrontal cortex decreased, indicating that NMDA-R hypofunctions [87]. In the post-mortem frontal cortex of untreated patients with schizophrenia, downregulation of group II metabotropic glutamate receptors (II-mGluR), such as mGlu2/3, was reported [88]. Conversely, III-mGlu receptor expression in schizophrenia remains unreported, whereas the activation of the III-mGlu receptor suppressed the hyperactivated transmission induced by NMDA-R impairment in wild-type and II-mGluR deficit models [83][89].
In other line studies, both post-mortem and experimental animal model studies also demonstrated that impairment of the GABA-B receptor plays an important role in the pathophysiology of schizophrenia. Decreased GABA-B receptor expression in the hippocampus, prefrontal cortex, inferior temporal cortex, and entorhinal cortex in schizophrenia has been reported [90][91]. Decreased GABA-B receptor expression in the prefrontal cortex and hippocampus of the DBA/2J schizophrenia model compared with C57BL/6J mice was also revealed [92]. Clinically, clozapine is evaluated as the most effective antipsychotic to improve sensorimotor gating dysfunction in patients with schizophrenia [93]. Maladaptive perseveration with strategies that cannot lead to the desired outcome resulting from cognitive and behavioural inflexibility via possible sensorimotor gating dysfunction in the thalamocortical pathway is considered a characteristic feature of schizophrenia [33][46][68][94][95]. Pre-pulse inhibition (PPI) has been established as an endo-phenotype of sensorimotor gating function. Clozapine improved PPI deficits in an experimental animal model, ZFP804A mutant mice, and an NMDA/glutamate receptor (ketamine)-induced model [96][97]. Baclofen has also been indicated to counter PPI disruption of the acoustic startle reflex produced by the blockading of the NMDA-R [92]. Notably, the effects of baclofen on PPI deficit were comparable to those of clozapine but more prominent than those of the typical antipsychotic, haloperidol [92]. These behavioural studies suggest that the impacts of a GABA-B deficit contribute to sensorimotor impairment in schizophrenia.
A recent study using molecular docking calculations for the X-ray crystal structure of the GABA-B receptor suggested that clozapine, like baclofen, might bind to the GABA-B receptor [98]. Both clinical and preclinical studies have suggested that clozapine enhances GABA-B receptor function, and the direct binding of clozapine to the GABA-B receptor has not been demonstrated but, rather, has been denied [32][99][100]. Considering these previous findings, the enhancement of GABA-B receptor function with clozapine may be mediated by an indirect mechanism of clozapine rather than a direct agonist action. Therefore, the hypothesis regarding the stimulatory effects of clozapine on GABA-B receptor function is intriguing for understanding the underlying pathophysiology of the clinical efficacy of clozapine in TRS.

This entry is adapted from the peer-reviewed paper 10.3390/biom13091288

References

  1. Lally, J.; MacCabe, J.H. Antipsychotic medication in schizophrenia: A review. Br. Med. Bull. 2015, 114, 169–179.
  2. Tandon, R. Schizophrenia and Other Psychotic Disorders in Diagnostic and Statistical Manual of Mental Disorders (DSM)-5: Clinical Implications of Revisions from DSM-IV. Indian J. Psychol. Med. 2014, 36, 223–225.
  3. Tan, N.; Van Os, J. Schizofreniespectrum en andere psychotische stoornissen in de DSM-5. Tijdschr. Voor Psychiatr. 2014, 56, 167–172.
  4. Hu, R.J.; Malhotra, A.K.; Pickar, D. Predicting response to clozapine. CNS Drugs 1999, 11, 317–326.
  5. Kane, J.; Honigfeld, G.; Singer, J.; Meltzer, H. Clozapine for the treatment-resistant schizophrenic: A double-blind comparison with chlorpromazine. Arch. Gen. Psychiatry 1988, 45, 789–796.
  6. Meltzer, H.Y. Clozapine: Balancing safety with superior antipsychotic efficacy. Clin. Schizophr. Relat. Psychoses 2012, 6, 134–144.
  7. Tiihonen, J.; Mittendorfer-Rutz, E.; Majak, M.; Mehtala, J.; Hoti, F.; Jedenius, E.; Enkusson, D.; Leval, A.; Sermon, J.; Tanskanen, A.; et al. Real-World Effectiveness of Antipsychotic Treatments in a Nationwide Cohort of 29823 Patients with Schizophrenia. JAMA Psychiatry 2017, 74, 686–693.
  8. Falkai, P.; Wobrock, T.; Lieberman, J.; Glenthoj, B.; Gattaz, W.F.; Moller, H.J.; WFSBP Task Force on Treatment Guidelines for Schizophrenia. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of schizophrenia, part 2: Long-term treatment of schizophrenia. World J. Biol. Psychiatry 2006, 7, 5–40.
  9. Hasan, A.; Falkai, P.; Wobrock, T.; Lieberman, J.; Glenthoj, B.; Gattaz, W.F.; Thibaut, F.; Moller, H.J.; WFSBP Task Force on Treatment Guidelines for Schizophrenia. World Federation of Societies of Biological Psychiatry (WFSBP) Guidelines for Biological Treatment of Schizophrenia, part 1: Update 2012 on the acute treatment of schizophrenia and the management of treatment resistance. World J. Biol. Psychiatry 2012, 13, 318–378.
  10. Rogers, J.P.; Oldham, M.A.; Fricchione, G.; Northoff, G.; Ellen Wilson, J.; Mann, S.C.; Francis, A.; Wieck, A.; Elizabeth Wachtel, L.; Lewis, G.; et al. Evidence-based consensus guidelines for the management of catatonia: Recommendations from the British Association for Psychopharmacology. J. Psychopharmacol. 2023, 37, 327–369.
  11. Masuda, T.; Misawa, F.; Takase, M.; Kane, J.M.; Correll, C.U. Association with Hospitalization and All-Cause Discontinuation Among Patients with Schizophrenia on Clozapine vs Other Oral Second-Generation Antipsychotics: A Systematic Review and Meta-analysis of Cohort Studies. JAMA Psychiatry 2019, 76, 1052.
  12. Huhn, M.; Nikolakopoulou, A.; Schneider-Thoma, J.; Krause, M.; Samara, M.; Peter, N.; Arndt, T.; Backers, L.; Rothe, P.; Cipriani, A.; et al. Comparative efficacy and tolerability of 32 oral antipsychotics for the acute treatment of adults with multi-episode schizophrenia: A systematic review and network meta-analysis. Lancet 2019, 394, 939–951.
  13. Mizuno, Y.; McCutcheon, R.A.; Brugger, S.P.; Howes, O.D. Heterogeneity and efficacy of antipsychotic treatment for schizophrenia with or without treatment resistance: A meta-analysis. Neuropsychopharmacology 2020, 45, 622–631.
  14. Wenthur, C.J.; Lindsley, C.W. Classics in chemical neuroscience: Clozapine. ACS Chem. Neurosci. 2013, 4, 1018–1025.
  15. Rusheen, A.E.; Gee, T.A.; Jang, D.P.; Blaha, C.D.; Bennet, K.E.; Lee, K.H.; Heien, M.L.; Oh, Y. Evaluation of electrochemical methods for tonic dopamine detection in vivo. TrAC Trends Anal. Chem. 2020, 132, 116049.
  16. Meltzer, H.Y. Update on typical and atypical antipsychotic drugs. Annu. Rev. Med. 2013, 64, 393–406.
  17. O’Connor, W.T.; O’Shea, S.D. Clozapine and GABA transmission in schizophrenia disease models: Establishing principles to guide treatments. Pharmacol. Ther. 2015, 150, 47–80.
  18. Meltzer, H.Y.; Lee, M.A.; Ranjan, R.; Mason, E.A.; Cola, P.A. Relapse following clozapine withdrawal: Effect of neuroleptic drugs and cyproheptadine. Psychopharmacology 1996, 124, 176–187.
  19. Correll, C.U. From receptor pharmacology to improved outcomes: Individualising the selection, dosing, and switching of antipsychotics. Eur. Psychiatry 2010, 25 (Suppl. S2), S12–S21.
  20. Seeman, P. Clozapine, a fast-off-D2 antipsychotic. ACS Chem. Neurosci. 2014, 5, 24–29.
  21. Kelly, A.C.; Sheitman, B.B.; Hamer, R.M.; Rhyne, D.C.; Reed, R.M.; Graham, K.A.; Rau, S.W.; Gilmore, J.H.; Perkins, D.O.; Peebles, S.S.; et al. A naturalistic comparison of the long-term metabolic adverse effects of clozapine versus other antipsychotics for patients with psychotic illnesses. J. Clin. Psychopharmacol. 2014, 34, 441–445.
  22. Lund, B.C.; Perry, P.J.; Brooks, J.M.; Arndt, S. Clozapine use in patients with schizophrenia and the risk of diabetes, hyperlipidemia, and hypertension: A claims-based approach. Arch. Gen. Psychiatry 2001, 58, 1172–1176.
  23. American Diabetes, A. Introduction: Standards of Medical Care in Diabetes-2022. Diabetes Care 2022, 45, S1–S2.
  24. Taylor, S.I.; Yazdi, Z.S.; Beitelshees, A.L. Pharmacological treatment of hyperglycemia in type 2 diabetes. J. Clin. Investig. 2021, 131, e142243.
  25. Popli, A.P.; Konicki, P.E.; Jurjus, G.J.; Fuller, M.A.; Jaskiw, G.E. Clozapine and associated diabetes mellitus. J. Clin. Psychiatry 1997, 58, 108–111.
  26. Jiang, W.L.; Cai, D.B.; Yin, F.; Zhang, L.; Zhao, X.W.; He, J.; Ng, C.H.; Ungvari, G.S.; Sim, K.; Hu, M.L.; et al. Adjunctive metformin for antipsychotic-induced dyslipidemia: A meta-analysis of randomized, double-blind, placebo-controlled trials. Transl. Psychiatry 2020, 10, 117.
  27. Zimbron, J.; Khandaker, G.M.; Toschi, C.; Jones, P.B.; Fernandez-Egea, E. A systematic review and meta-analysis of randomised controlled trials of treatments for clozapine-induced obesity and metabolic syndrome. Eur. Neuropsychopharmacol. 2016, 26, 1353–1365.
  28. Siskind, D.J.; Leung, J.; Russell, A.W.; Wysoczanski, D.; Kisely, S. Metformin for Clozapine Associated Obesity: A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0156208.
  29. Wang, C.; Shi, W.; Xu, J.; Huang, C.; Zhu, J. Outcomes and safety of concomitant topiramate or metformin for antipsychotics-induced obesity: A randomized-controlled trial. Ann. Gen. Psychiatry 2020, 19, 68.
  30. Dayabandara, M.; Hanwella, R.; Ratnatunga, S.; Seneviratne, S.; Suraweera, C.; de Silva, V.A. Antipsychotic-associated weight gain: Management strategies and impact on treatment adherence. Neuropsychiatr. Dis. Treat. 2017, 13, 2231–2241.
  31. Siskind, D.; Hahn, M.; Correll, C.U.; Fink-Jensen, A.; Russell, A.W.; Bak, N.; Broberg, B.V.; Larsen, J.; Ishoy, P.L.; Vilsboll, T.; et al. Glucagon-like peptide-1 receptor agonists for antipsychotic-associated cardio-metabolic risk factors: A systematic review and individual participant data meta-analysis. Diabetes Obes. Metab. 2019, 21, 293–302.
  32. Wu, H.; Siafis, S.; Hamza, T.; Schneider-Thoma, J.; Davis, J.M.; Salanti, G.; Leucht, S. Antipsychotic-Induced Weight Gain: Dose-Response Meta-Analysis of Randomized Controlled Trials. Schizophr. Bull. 2022, 48, 643–654.
  33. Fukuyama, K.; Motomura, E.; Okada, M. Therapeutic Potential and Limitation of Serotonin Type 7 Receptor Modulation. Int. J. Mol. Sci. 2023, 24, 2070.
  34. Carli, M.; Kolachalam, S.; Longoni, B.; Pintaudi, A.; Baldini, M.; Aringhieri, S.; Fasciani, I.; Annibale, P.; Maggio, R.; Scarselli, M. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals (Basel) 2021, 14, 238.
  35. Okada, M.; Yoshida, S.; Zhu, G.; Hirose, S.; Kaneko, S. Biphasic actions of topiramate on monoamine exocytosis associated with both soluble N-ethylmaleimide-sensitive factor attachment protein receptors and Ca(2+)-induced Ca(2+)-releasing systems. Neuroscience 2005, 134, 233–246.
  36. de Brito, O.M.; Scorrano, L. An intimate liaison: Spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J. 2010, 29, 2715–2723.
  37. Decrock, E.; De Bock, M.; Wang, N.; Gadicherla, A.K.; Bol, M.; Delvaeye, T.; Vandenabeele, P.; Vinken, M.; Bultynck, G.; Krysko, D.V.; et al. IP3, a small molecule with a powerful message. Biochim. Biophys. Acta 2013, 1833, 1772–1786.
  38. Fukuyama, K.; Motomura, E.; Okada, M. Opposing effects of clozapine and brexpiprazole on beta-aminoisobutyric acid: Pathophysiology of antipsychotics-induced weight gain. Schizophrenia 2023, 9, 8.
  39. Foretz, M.; Guigas, B.; Bertrand, L.; Pollak, M.; Viollet, B. Metformin: From mechanisms of action to therapies. Cell Metab. 2014, 20, 953–966.
  40. Lopez, M. Hypothalamic AMPK as a possible target for energy balance-related diseases. Trends Pharmacol. Sci. 2022, 43, 546–556.
  41. Fukuyama, K.; Motomura, E.; Okada, M. Enhanced L-beta-Aminoisobutyric Acid Is Involved in the Pathophysiology of Effectiveness for Treatment-Resistant Schizophrenia and Adverse Reactions of Clozapine. Biomolecules 2023, 13, 862.
  42. Fukuyama, K.; Motomura, E.; Okada, M. A Candidate Gliotransmitter, L-β-Aminoisobutyrate, Contributes to Weight Gain and Metabolic Complication Induced by Atypical Antipsychotics. Nutrients 2023, 15, 1621.
  43. Fukuyama, K.; Okada, M. Effects of Atypical Antipsychotics, Clozapine, Quetiapine and Brexpiprazole on Astroglial Transmission Associated with Connexin43. Int. J. Mol. Sci. 2021, 22, 5623.
  44. Fukuyama, K.; Motomura, E.; Shiroyama, T.; Okada, M. Impact of 5-HT7 receptor inverse agonism of lurasidone on monoaminergic tripartite synaptic transmission and pathophysiology of lower risk of weight gain. Biomed. Pharmacother. 2022, 148, 112750.
  45. Okada, M.; Fukuyama, K.; Motomura, E. Dose-Dependent Biphasic Action of Quetiapine on AMPK Signalling via 5-HT7 Receptor: Exploring Pathophysiology of Clinical and Adverse Effects of Quetiapine. Int. J. Mol. Sci. 2022, 23, 9103.
  46. Okubo, R.; Hasegawa, T.; Fukuyama, K.; Shiroyama, T.; Okada, M. Current Limitations and Candidate Potential of 5-HT7 Receptor Antagonism in Psychiatric Pharmacotherapy. Front. Psychiatry 2021, 12, 623684.
  47. Bymaster, F.P.; Nelson, D.L.; DeLapp, N.W.; Falcone, J.F.; Eckols, K.; Truex, L.L.; Foreman, M.M.; Lucaites, V.L.; Calligaro, D.O. Antagonism by olanzapine of dopamine D1, serotonin2, muscarinic, histamine H1 and alpha 1-adrenergic receptors in vitro. Schizophr. Res. 1999, 37, 107–122.
  48. Arranz, B.; Rosel, P.; San, L.; Ramirez, N.; Duenas, R.M.; Salavert, J.; Centeno, M.; del Moral, E. Low baseline serotonin-2A receptors predict clinical response to olanzapine in first-episode schizophrenia patients. Psychiatry Res. 2007, 153, 103–109.
  49. Deng, C.; Weston-Green, K.; Huang, X.F. The role of histaminergic H1 and H3 receptors in food intake: A mechanism for atypical antipsychotic-induced weight gain? Prog. Neuropsychopharmacol. Biol. Psychiatry 2010, 34, 1–4.
  50. Han, M.; Lian, J.; Su, Y.; Deng, C. Cevimeline co-treatment attenuates olanzapine-induced metabolic disorders via modulating hepatic M3 muscarinic receptor: AMPKalpha signalling pathway in female rats. J. Psychopharmacol. 2022, 36, 202–213.
  51. Castellani, L.N.; Pereira, S.; Kowalchuk, C.; Asgariroozbehani, R.; Singh, R.; Wu, S.; Hamel, L.; Alganem, K.; Ryan, W.G.; Zhang, X.; et al. Antipsychotics impair regulation of glucose metabolism by central glucose. Mol. Psychiatry 2022, 27, 4741–4753.
  52. Ferreira, V.; Folgueira, C.; Guillen, M.; Zubiaur, P.; Navares, M.; Sarsenbayeva, A.; Lopez-Larrubia, P.; Eriksson, J.W.; Pereira, M.J.; Abad-Santos, F.; et al. Modulation of hypothalamic AMPK phosphorylation by olanzapine controls energy balance and body weight. Metabolism 2022, 137, 155335.
  53. Howes, O.D.; McCutcheon, R.; Agid, O.; de Bartolomeis, A.; van Beveren, N.J.; Birnbaum, M.L.; Bloomfield, M.A.; Bressan, R.A.; Buchanan, R.W.; Carpenter, W.T.; et al. Treatment-Resistant Schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. Am. J. Psychiatry 2017, 174, 216–229.
  54. Howes, O.D.; Thase, M.E.; Pillinger, T. Treatment resistance in psychiatry: State of the art and new directions. Mol. Psychiatry 2022, 27, 58–72.
  55. Siskind, D.; McCartney, L.; Goldschlager, R.; Kisely, S. Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: Systematic review and meta-analysis. Br. J. Psychiatry 2016, 209, 385–392.
  56. Wagner, E.; Siafis, S.; Fernando, P.; Falkai, P.; Honer, W.G.; Roh, A.; Siskind, D.; Leucht, S.; Hasan, A. Efficacy and safety of clozapine in psychotic disorders-a systematic quantitative meta-review. Transl. Psychiatry 2021, 11, 487.
  57. Joo, S.W.; Kim, H.; Jo, Y.T.; Ahn, S.; Choi, Y.J.; Choi, W.; Park, S.; Lee, J. Comparative effectiveness of antipsychotic monotherapy and polypharmacy in schizophrenia patients with clozapine treatment: A nationwide, health insurance data-based study. Eur. Neuropsychopharmacol. 2022, 59, 36–44.
  58. McEvoy, J.P.; Lieberman, J.A.; Stroup, T.S.; Davis, S.M.; Meltzer, H.Y.; Rosenheck, R.A.; Swartz, M.S.; Perkins, D.O.; Keefe, R.S.; Davis, C.E.; et al. Effectiveness of clozapine versus olanzapine, quetiapine, and risperidone in patients with chronic schizophrenia who did not respond to prior atypical antipsychotic treatment. Am. J. Psychiatry 2006, 163, 600–610.
  59. Correll, C.U.; Howes, O.D. Treatment-Resistant Schizophrenia: Definition, Predictors, and Therapy Options. J. Clin. Psychiatry 2021, 82, MY20096AH1C.
  60. Mouchlianitis, E.; McCutcheon, R.; Howes, O.D. Brain-imaging studies of treatment-resistant schizophrenia: A systematic review. Lancet Psychiatry 2016, 3, 451–463.
  61. Potkin, S.G.; Kane, J.M.; Correll, C.U.; Lindenmayer, J.P.; Agid, O.; Marder, S.R.; Olfson, M.; Howes, O.D. The neurobiology of treatment-resistant schizophrenia: Paths to antipsychotic resistance and a roadmap for future research. NPJ Schizophr. 2020, 6, 1.
  62. Wada, M.; Noda, Y.; Iwata, Y.; Tsugawa, S.; Yoshida, K.; Tani, H.; Hirano, Y.; Koike, S.; Sasabayashi, D.; Katayama, H.; et al. Dopaminergic dysfunction and excitatory/inhibitory imbalance in treatment-resistant schizophrenia and novel neuromodulatory treatment. Mol. Psychiatry 2022, 27, 2950–2967.
  63. Siskind, D.; Siskind, V.; Kisely, S. Clozapine Response Rates among People with Treatment-Resistant Schizophrenia: Data from a Systematic Review and Meta-Analysis. Can. J. Psychiatry 2017, 62, 772–777.
  64. Yoshimura, B.; Yada, Y.; So, R.; Takaki, M.; Yamada, N. The critical treatment window of clozapine in treatment-resistant schizophrenia: Secondary analysis of an observational study. Psychiatry Res. 2017, 250, 65–70.
  65. Agid, O.; Arenovich, T.; Sajeev, G.; Zipursky, R.B.; Kapur, S.; Foussias, G.; Remington, G. An algorithm-based approach to first-episode schizophrenia: Response rates over 3 prospective antipsychotic trials with a retrospective data analysis. J. Clin. Psychiatry 2011, 72, 1439–1444.
  66. Kahn, R.S.; Winter van Rossum, I.; Leucht, S.; McGuire, P.; Lewis, S.W.; Leboyer, M.; Arango, C.; Dazzan, P.; Drake, R.; Heres, S.; et al. Amisulpride and olanzapine followed by open-label treatment with clozapine in first-episode schizophrenia and schizophreniform disorder (OPTiMiSE): A three-phase switching study. Lancet Psychiatry 2018, 5, 797–807.
  67. John, A.P.; Ko, E.K.F.; Dominic, A. Delayed Initiation of Clozapine Continues to Be a Substantial Clinical Concern. Can. J. Psychiatry 2018, 63, 526–531.
  68. Okada, M.; Fukuyama, K.; Shiroyama, T.; Murata, M. A Working Hypothesis Regarding Identical Pathomechanisms between Clinical Efficacy and Adverse Reaction of Clozapine via the Activation of Connexin43. Int. J. Mol. Sci. 2020, 21, 7019.
  69. Kapur, S.; Seeman, P. Does fast dissociation from the dopamine d(2) receptor explain the action of atypical antipsychotics?: A new hypothesis. Am. J. Psychiatry 2001, 158, 360–369.
  70. Stepnicki, P.; Kondej, M.; Kaczor, A.A. Current Concepts and Treatments of Schizophrenia. Molecules 2018, 23, 2087.
  71. Sykes, D.A.; Moore, H.; Stott, L.; Holliday, N.; Javitch, J.A.; Lane, J.R.; Charlton, S.J. Extrapyramidal side effects of antipsychotics are linked to their association kinetics at dopamine D(2) receptors. Nat. Commun. 2017, 8, 763.
  72. Sahlholm, K.; Zeberg, H.; Nilsson, J.; Ogren, S.O.; Fuxe, K.; Arhem, P. The fast-off hypothesis revisited: A functional kinetic study of antipsychotic antagonism of the dopamine D2 receptor. Eur. Neuropsychopharmacol. 2016, 26, 467–476.
  73. Kapur, S.; Seeman, P. Antipsychotic agents differ in how fast they come off the dopamine D2 receptors. Implications for atypical antipsychotic action. J. Psychiatry Neurosci. 2000, 25, 161–166.
  74. Demjaha, A.; Lappin, J.M.; Stahl, D.; Patel, M.X.; MacCabe, J.H.; Howes, O.D.; Heslin, M.; Reininghaus, U.A.; Donoghue, K.; Lomas, B.; et al. Antipsychotic treatment resistance in first-episode psychosis: Prevalence, subtypes and predictors. Psychol. Med. 2017, 47, 1981–1989.
  75. Lally, J.; Ajnakina, O.; Di Forti, M.; Trotta, A.; Demjaha, A.; Kolliakou, A.; Mondelli, V.; Reis Marques, T.; Pariante, C.; Dazzan, P.; et al. Two distinct patterns of treatment resistance: Clinical predictors of treatment resistance in first-episode schizophrenia spectrum psychoses. Psychol. Med. 2016, 46, 3231–3240.
  76. Carbon, M.; Correll, C.U. Clinical predictors of therapeutic response to antipsychotics in schizophrenia. Dialogues Clin. Neurosci. 2014, 16, 505–524.
  77. Murray, R.M.; O’Callaghan, E.; Castle, D.J.; Lewis, S.W. A neurodevelopmental approach to the classification of schizophrenia. Schizophr. Bull. 1992, 18, 319–332.
  78. Bourque, J.; Lakis, N.; Champagne, J.; Stip, E.; Lalonde, P.; Lipp, O.; Mendrek, A. Clozapine and visuospatial processing in treatment-resistant schizophrenia. Cogn. Neuropsychiatry 2013, 18, 615–630.
  79. de Bartolomeis, A.; Balletta, R.; Giordano, S.; Buonaguro, E.F.; Latte, G.; Iasevoli, F. Differential cognitive performances between schizophrenic responders and non-responders to antipsychotics: Correlation with course of the illness, psychopathology, attitude to the treatment and antipsychotics doses. Psychiatry Res. 2013, 210, 387–395.
  80. Huang, J.; Zhu, Y.; Fan, F.; Chen, S.; Hong, Y.; Cui, Y.; Luo, X.; Tan, S.; Wang, Z.; Shang, L.; et al. Hippocampus and cognitive domain deficits in treatment-resistant schizophrenia: A comparison with matched treatment-responsive patients and healthy controls(✰,✰✰, bigstar, bigstar bigstar). Psychiatry Res. Neuroimaging 2020, 297, 111043.
  81. Lin, A.S.; Chan, H.Y.; Peng, Y.C.; Chen, W.J. Severity in sustained attention impairment and clozapine-resistant schizophrenia: A retrospective study. BMC Psychiatry 2019, 19, 220.
  82. Tanahashi, S.; Yamamura, S.; Nakagawa, M.; Motomura, E.; Okada, M. Clozapine, but not haloperidol, enhances glial D-serine and L-glutamate release in rat frontal cortex and primary cultured astrocytes. Br. J. Pharmacol. 2012, 165, 1543–1555.
  83. Fukuyama, K.; Kato, R.; Murata, M.; Shiroyama, T.; Okada, M. Clozapine Normalizes a Glutamatergic Transmission Abnormality Induced by an Impaired NMDA Receptor in the Thalamocortical Pathway via the Activation of a Group III Metabotropic Glutamate Receptor. Biomolecules 2019, 9, 234.
  84. Fukuyama, K.; Okubo, R.; Murata, M.; Shiroyama, T.; Okada, M. Activation of Astroglial Connexin is Involved in Concentration-Dependent Double-Edged Sword Clinical Action of Clozapine. Cells 2020, 9, 414.
  85. Ito, Y.; Murata, M.; Taku, O.; Fukuyama, K.; Motomura, E.; Dohi, K.; Okada, M. Developed catatonia with rhabdomyolysis and exacerbated cardiac failure upon switching from clozapine to olanzapine owing to cardiomyopathy during clozapine medication—A case report. Asian J. Psychiatr. 2023, 80, 103376.
  86. Kruse, A.O.; Bustillo, J.R. Glutamatergic dysfunction in Schizophrenia. Transl. Psychiatry 2022, 12, 500.
  87. Wang, H.Y.; MacDonald, M.L.; Borgmann-Winter, K.E.; Banerjee, A.; Sleiman, P.; Tom, A.; Khan, A.; Lee, K.C.; Roussos, P.; Siegel, S.J.; et al. mGluR5 hypofunction is integral to glutamatergic dysregulation in schizophrenia. Mol. Psychiatry 2020, 25, 750–760.
  88. Gonzalez-Maeso, J.; Ang, R.L.; Yuen, T.; Chan, P.; Weisstaub, N.V.; Lopez-Gimenez, J.F.; Zhou, M.; Okawa, Y.; Callado, L.F.; Milligan, G.; et al. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature 2008, 452, 93–97.
  89. Fell, M.J.; Svensson, K.A.; Johnson, B.G.; Schoepp, D.D. Evidence for the role of metabotropic glutamate (mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclohexane-4,6-dicarboxylic acid (LY404039). J. Pharmacol. Exp. Ther. 2008, 326, 209–217.
  90. Mizukami, K.; Sasaki, M.; Ishikawa, M.; Iwakiri, M.; Hidaka, S.; Shiraishi, H.; Iritani, S. Immunohistochemical localization of gamma-aminobutyric acid(B) receptor in the hippocampus of subjects with schizophrenia. Neurosci. Lett. 2000, 283, 101–104.
  91. Ishikawa, M.; Mizukami, K.; Iwakiri, M.; Asada, T. Immunohistochemical and immunoblot analysis of gamma-aminobutyric acid B receptor in the prefrontal cortex of subjects with schizophrenia and bipolar disorder. Neurosci. Lett. 2005, 383, 272–277.
  92. Bortolato, M.; Frau, R.; Orru, M.; Piras, A.P.; Fa, M.; Tuveri, A.; Puligheddu, M.; Gessa, G.L.; Castelli, M.P.; Mereu, G.; et al. Activation of GABA(B) receptors reverses spontaneous gating deficits in juvenile DBA/2J mice. Psychopharmacology 2007, 194, 361–369.
  93. Micoulaud-Franchi, J.A.; Aramaki, M.; Geoffroy, P.A.; Richieri, R.; Cermolacce, M.; Faget, C.; Ystad, S.; Kronland-Martinet, R.; Lancon, C.; Vion-Dury, J. Effects of clozapine on perceptual abnormalities and sensory gating: A preliminary cross-sectional study in schizophrenia. J. Clin. Psychopharmacol. 2015, 35, 184–187.
  94. Beas, B.S.; Setlow, B.; Bizon, J.L. Effects of acute administration of the GABA(B) receptor agonist baclofen on behavioral flexibility in rats. Psychopharmacology 2016, 233, 2787–2797.
  95. Okada, M.; Kawano, Y.; Fukuyama, K.; Motomura, E.; Shiroyama, T. Candidate Strategies for Development of a Rapid-Acting Antidepressant Class That Does Not Result in Neuropsychiatric Adverse Effects: Prevention of Ketamine-Induced Neuropsychiatric Adverse Reactions. Int. J. Mol. Sci. 2020, 21, 7951.
  96. Huang, Y.; Huang, J.; Zhou, Q.X.; Yang, C.X.; Yang, C.P.; Mei, W.Y.; Zhang, L.; Zhang, Q.; Hu, L.; Hu, Y.Q.; et al. ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol. Psychiatry 2021, 26, 2514–2532.
  97. Yang, S.Y.; Hong, C.J.; Huang, Y.H.; Tsai, S.J. The effects of glycine transporter I inhibitor, N-methylglycine (sarcosine), on ketamine-induced alterations in sensorimotor gating and regional brain c-Fos expression in rats. Neurosci. Lett. 2010, 469, 127–130.
  98. Nair, P.C.; McKinnon, R.A.; Miners, J.O.; Bastiampillai, T. Binding of clozapine to the GABAB receptor: Clinical and structural insights. Mol. Psychiatry 2020, 25, 1910–1919.
  99. Roth BL, D.J. Psychoactive Drug Screening Program (PDSP). 2017. Available online: https://pdsp.unc.edu/databases/pdsp.php?knowID=0&kiKey=&receptorDD=&receptor=&speciesDD=&species=&sourcesDD=&source=&hotLigandDD=&hotLigand=&testLigandDD=&testFreeRadio=testFreeRadio&testLigand=clozapine&referenceDD=&reference=&KiGreater=&KiLess=&kiAllRadio=all&doQuery=Submit+Query (accessed on 27 July 2023).
  100. Miyazawa, A.; Kanahara, N.; Shiko, Y.; Ozawa, Y.; Kawasaki, Y.; Komatsu, H.; Masumo, Y.; Nakata, Y.; Iyo, M. The cortical silent period in schizophrenia: A systematic review and meta-analysis focusing on disease stage and antipsychotic medication. J. Psychopharmacol. 2022, 36, 479–488.
More
This entry is offline, you can click here to edit this entry!
Video Production Service