Third-Generation Antipsychotics and Lurasidone in Substance-Induced Psychoses Treatment: Comparison
Please note this is a comparison between Version 2 by Lindsay Dong and Version 1 by Valerio Ricci.

Substance-induced psychosis (SIP) is a psychiatric condition triggered by substance misuse or withdrawal, characterized by unique features distinct from those of primary psychotic disorders. These distinctive features include a heightened prevalence of positive symptoms, such as hallucinations and delusions, in addition to a spectrum of mood and cognitive disturbances. 

  • substance-induced psychosis
  • psychostimulants
  • schizophrenia
  • third-generation antipsychotics

1. Introduction

Numerous research findings indicate that illicit substances, like cannabinoids, cocaine, amphetamines, and hallucinogens, exhibit psychotomimetic properties [1,2][1][2]. This implies that their usage not only triggers temporary psychotic symptoms during acute intoxication but also may result in a syndrome closely resembling a primary psychotic disorder. In recent decades, a diverse range of novel psychoactive substances has emerged, encompassing synthetic cannabinoids, cathinone derivatives, psychedelic phenethylamines, new stimulants, synthetic opioids, tryptamine derivatives, phencyclidine-like dissociatives, piperazines, and GABAA/B receptor agonists. These substances are increasingly prevalent in the landscape of substance abuse [3].

A complex clinical challenge revolves around accurately differentiating substance-induced psychosis from a primary psychotic disorder or a psychotic disorder co-occurring with substance use. This poses a nuanced dilemma and an opportunity for thorough investigation, gaining significance when determining the optimal therapeutic approach for patients. The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) [4] more accurately describes the substance/medication-induced psychotic disorder as a psychiatric condition characterized by delusions and/or hallucinations that arise during or shortly following the intoxication or withdrawal from a substance.
Additionally, the symptoms of a non-substance-induced psychotic disorder are not yet fully understood. Nevertheless, in this context, a particular definition has been formulated utilizing the diagnostic classification of “substance-related exogenous psychosis (SREP)”. This concept refers to a range of psychotic symptoms that are temporary or enduring and are associated with substance use. These include changes in consciousness; feelings of being persecuted; disorders affecting sensory perception, like visual and bodily hallucinations; impulsive behavior, self- or other-directed aggression, and psychomotor restlessness; fluctuations in mood; negative affects, such as indifference, lack of motivation, and inability to feel pleasure; an overwhelming feeling of being detached from reality; and maintained self-awareness [1].
The occurrence of drug-induced psychosis appears to be associated with various pathogenetic mechanisms: (a) elevated levels of central dopamine, particularly for hallucinogens or psychedelic substances, stimulants, and cathinone derivatives; (b) activity as an agonist at cannabinoid CB1 receptors, particularly in substances related to cannabis; (c) agonist activity at 5-HT2A receptors in hallucinogenic plants, newer phenethylamines, and tryptamine derivatives; (d) activity as an antagonist at NMDA receptors (n-methyl-D-aspartate receptors), seen in substances like ketamine and methoxetamine; and (e) activation of k-opioid receptors in plants, such as Salvia divinorum [6][5].

2. Third-Generation Antipsychotics and Lurasidone in Substance-Induced Psychoses Treatment 

2.1. Aripiprazole

Aripiprazole, classified as a partial agonist within the antipsychotic drug category, exhibits lower intrinsic activity at receptors compared to full agonists [24][6]. Its partial agonism at dopamine D2 receptors leads to (1) functional antagonism in the mesolimbic dopamine pathway, reducing positive symptoms caused by excessive dopamine activity, and (2) agonist activity in the mesocortical pathway, addressing negative symptoms and cognitive impairment due to reduced dopamine activity [25,26,27][7][8][9]. This dual effect contributes to significant improvements in both positive and negative psychotic symptoms. Moreover, aripiprazole avoids the complete blockade of the nigrostriatal or tuberoinfundibular pathways, thereby preserving the avoidance of extrapyramidal symptoms and hyperprolactinemia [24][6]. It also demonstrates a favorable cardiac safety profile, preventing QTc prolongation and causing minimal weight gain or sedation. Focusing on substance use, aripiprazole shows promise in animal models, where acute administration prevented increased locomotion induced by stimulants, like amphetamine, cocaine, and methylphenidate, and attenuated their reinforcing properties without interfering with spontaneous motor activity [26][8]. It reversed amphetamine-associated anhedonia and prevented the reinstatement of cocaine-seeking behavior [27][9], suggesting its potential in alleviating withdrawal symptoms linked to dopamine depletion and maintaining balanced dopamine neurotransmission in drug-dependent behavior [28,29,30][10][11][12].

2.2. Cariprazine

Cariprazine, a third-generation antipsychotic drug approved for schizophrenia [39][13], is notable for its partial agonism toward D2 receptors. Distinguishing itself from other second-generation antipsychotics (SGAs), it demonstrates a lower intrinsic activity at the D2 receptor compared to aripiprazole and brexpiprazole, approximately 0.15. This positioning suggests a minimized capacity to activate the D2 receptor, offering an intermediary profile between aripiprazole and traditional receptor antagonists with zero intrinsic activity [39,40,41,42][13][14][15][16]. The reduced intrinsic activity of cariprazine is associated with a decreased likelihood of side effects, such as restlessness and akathisia, commonly observed with aripiprazole [40][14]. Cariprazine’s primary distinction lies in its high affinity for the dopaminergic D3 receptor, exceeding that of endogenous dopamine. This high D3 affinity is significant, considering that most antipsychotics have a lower affinity for this receptor, leading to inadequate D3 receptor occupancy in the brain. This unique affinity is crucial for cariprazine’s clinical efficacy, as indicated by PET studies on schizophrenic patients, demonstrating a stronger effect on the D3 receptor compared to the D2 receptor [41,42,43,44,45,46,47][15][16][17][18][19][20][21]. Cariprazine also exhibits significant serotonergic activity, with high affinity for the 5-HT2B receptor and moderate affinity for the 5-HT2A and 5-HT1A receptors. This receptor profile is somewhat distinct from those of many SGAs, which tend to have high affinities for the 5-HT2A receptor. The 5-HT2A/D2 affinity ratio of cariprazine is lower than those of other antipsychotic drugs. Additionally, it has low affinity for the 5-HT7 and 5-HT2C receptors as well as the noradrenaline α1A and α1C receptors. Its interactions with the 5-HT6, α1a, and α2b receptors and other potential targets are minimal in therapeutic activity [42,43][16][17]. When comparing cariprazine with other partial agonists, such as blonanserin [48,49][22][23], an antagonist of dopaminergic D2 and D3 receptors, its D3/D2 affinity ratio is notably higher. This suggests a potential influence on negative symptoms, a theory supported by its unique receptor action. In contrast, brexpiprazole shows higher 5-HT2A/D2 and 5-HT1A/D2 ratios. The combined dopaminergic and serotonergic receptor activities of these drugs modulate specific brain circuits and neurotransmitter release, as observed in increased dopamine release in the prefrontal cortex with olanzapine and lurasidone, enhancing cognitive functions [50][24]. In conclusion, the receptor profile of cariprazine provides significant insights into cariprazine’s mechanism of action and ability to modulate various neurotransmitter systems. Unlike other antipsychotics, cariprazine’s efficacy is based on not only its receptor profile but also its capacity to modulate intracellular mechanisms downstream of these receptors [51,52][25][26]. Its dual action as a partial agonist at both D2 and D3 receptors, with a stronger effect on the latter, is particularly effective in improving negative symptoms, setting it apart from other SGAs [53][27]. Another intriguing facet of cariprazine pertains to its activity in the context of substance abuse. Experimental studies have illustrated cariprazine’s capacity to mitigate the stimulating effects of cocaine and forestall relapses associated with the abused substance. This observed activity seems to correlate with its partial agonism for dopamine receptors D2 and D3 [54][28].

2.3. Brexpiprazole

Brexpiprazole, an atypical antipsychotic, received FDA approval in July 2015 for treating schizophrenia and as an adjunct therapy for managing major depressive disorder (MDD). Acting as a partial agonist at 5-HT1A and D2 neuroreceptors, brexpiprazole also engages with noradrenergic receptors, although the clinical significance of this interaction is not fully understood. The safety and efficacy of brexpiprazole were evaluated in four finished placebo-controlled Phase III trials—two targeting major depressive disorder (MDD) as an add-on to antidepressants and two for schizophrenia. These studies showed that brexpiprazole was more effective than a placebo at certain dosages for both disorders [62,63,64][29][30][31]. For schizophrenia, the recommended initiation is 1 mg once daily, titrating to a target dose from 2 mg to 4 mg daily. As an adjunct therapy for MDD, the initiation dose is advised at 0.5 mg or 1 mg once daily, with a weekly increase to a target dose of 2 mg. Contraindications include prior hypersensitivity reactions to similar medications. Common adverse reactions involve weight gain and akathisia, with additional associations with metabolic changes, like dyslipidemia and hyperglycemia. Precautions and warnings for this medication include cerebrovascular adverse reactions in elderly patients with dementia-related psychosis, the risk of neuroleptic malignant syndrome, tardive dyskinesia, leukopenia, orthostatic hypotension, and seizures. A prominent black box warning highlights the increased risk of mortality in elderly patients with dementia-related psychosis, as well as the potential for suicidal thoughts and behaviors in children, adolescents, and young adults. Additionally, its use during pregnancy is cautioned against owing to the risk of extrapyramidal and/or withdrawal symptoms in neonates exposed during the third trimester. Brexpiprazole is metabolized by hepatic enzymes, and dose adjustments are necessary if the CYP2D6 or CYP3A4 superfamilies of enzymes are impacted [65][32]. Brexpiprazole functions as a partial agonist at dopamine D2 and serotonin 5-HT1A receptors and a potent antagonist at serotonin 5-HT2A, α1B, and α2C adrenergic receptors. In contrast to aripiprazole, brexpiprazole exhibits significantly greater potency at these three receptors: 5-HT2A, 5HT1A, and α1B. Despite commonly reported side effects, such as EPS and akathisia, the strong affinity of brexpiprazole to these receptors may contribute to a reduction in the occurrence of these symptoms. Brexpiprazole exhibits a higher intrinsic activity at the serotonin 5-HT2A receptor and a lower intrinsic activity at the dopamine D2 receptor along with a stronger affinity for the norepinephrine transporter [66][33]. Hyperprolactinemia, an often-undesirable side effect of many antipsychotics, is largely due to the blockade of dopamine D2 receptors, which interrupts the dopaminergic inhibition of the prolactin release. In a detailed evaluation, brexpiprazole shows moderate antagonist activity at dopamine D3 and serotonin 5-HT2B and 5-HT7 receptors, as well as α1A and α1D receptors. It also has moderate affinity for histamine H1 receptors and low affinity for muscarinic cholinergic M1 receptors. This pharmacodynamic profile not only enhances brexpiprazole’s efficacy but also positions brexpiprazole as a potentially preferred alternative based on patient tolerability and therapy goals [67][34]. Brexpiprazole undergoes primary metabolism by the enzymes CYP3A4 and CYP2D6. The major metabolite, DM-3411, constitutes between 23% and 48% of brexpiprazole’s exposure at a steady state, although it has not been demonstrated to contribute to any antipsychotic effects. Various factors influence the rate of brexpiprazole metabolism, subsequently impacting its overall exposure (AUC). Patients who use strong CYP3A4 inhibitors (like erythromycin or itraconazole) or potent CYP2D6 inhibitors (such as bupropion, fluoxetine, or paroxetine) may experience increased exposure to the drug. This heightened exposure is also seen in individuals who are poor metabolizers of CYP2D6. On the other hand, using potent CYP2D6 inducers (for instance, rifampicin or glucocorticoids) is likely to decrease the exposure to brexpiprazole. Brexpiprazole demonstrates efficacy in acute schizophrenia according to two studies [70,71][35][36]. Although the 2 mg dose showed inconsistent results, the recommended 4 mg dosage exhibited more consistent benefits. Higher brexpiprazole dosages, especially at 4 mg, significantly improved the PANSS-EC score and PANSS scores for negative symptoms, disorganized thought, and uncontrolled hostility/excitement. In a 52-week maintenance study, brexpiprazole outperformed the placebo, prolonging the time to exacerbation and showing significant benefits in psychosocial, occupational, and cognitive functioning, including attention/vigilance and visual learning [63][30]. Brexpiprazole also demonstrated cognitive improvement in animal models, distinguishing itself from aripiprazole in this regard. Short-term trials showed a good safety profile, with weight gain being the only common adverse event, and long-term studies indicated a decrease in the mean bodyweight change. Akathisia was dose-dependent but generally mild, with no treatment discontinuations [72][37]. Other adverse effects were comparable to those of the placebo, and minimal impacts on glucose, lipids, prolactin, and the QTc interval were observed.

2.4. Lurasidone

Lurasidone, like other second-generation antipsychotics, functions as a complete antagonist at dopamine D2 and serotonin 5-HT2A receptors, with binding affinities (Ki) of 1 nM and 0.5 nM, respectively. A distinctive feature of lurasidone is its high affinity for serotonin 5-HT7 receptors (0.5 nM, on par with its affinity for dopamine D2 and 5-HT2A receptors) and its partial agonist activity at 5-HT1A receptors (Ki, 6.4 nM). The serotonin 5-HT7 receptor is of significant interest as it is linked to potential procognitive and antidepressant effects. The 5-HT1A receptor is considered important in the treatment of major depressive disorder and schizophrenia. Notably, lurasidone lacks affinity for histamine H1 and muscarinic M1 receptors, contributing to its characteristics of low sedation, minimal weight gain, and limited interference with cognitive and functional assessments [75,76][38][39]. The pharmacokinetic characteristics of lurasidone support its suitability for once-daily administration, as it has an elimination half-life of 18 h [77,78][40][41]. When lurasidone was administered with food, both the mean Cmax and the area under the curve were approximately threefold and twofold greater, respectively, compared to those for administering lurasidone while fasting [79][42]. Based on these findings and clinical trial results, it is recommended to take lurasidone once daily in the evening, either with a meal or within 30 min after eating. Notably, lurasidone absorption remains unaffected by the fat content of the ingested food [79][42]. Lurasidone is primarily metabolized by the CYP3A4 enzyme system. Consequently, its use is contraindicated when there are strong inducers or inhibitors of CYP3A4 present. Within the category of psychotropic medications, notable examples of strong inhibitors of CYP3A4 include fluvoxamine and fluoxetine. On the other hand, a well-known strong inducer of CYP3A4 is carbamazepine. When moderate inhibitors of CYP3A4 are present, the suggested initial dose of lurasidone is 20 mg/day instead of 40 mg/day, and the highest recommended dose is 80 mg/day rather than 160 mg/day. Lurasidone’s pharmacokinetics do not interfere with those of other drugs, including lithium, valproate, or those metabolized by the CYP3A4 pathway [80][43]. In patients who have moderate or severe renal or hepatic impairment, the advised initial dosage of this medication is set at 20 mg per day. For those with moderate-to-severe renal impairment or moderate hepatic impairment, the maximum dosage should not surpass 80 mg per day. In cases of severe hepatic impairment, the dosage should be restricted to a maximum of 40 mg per day. Lurasidone demonstrated good tolerability, with consistent side effects observed in both short-term and long-term use. In trials spanning six weeks, the most frequently reported adverse reactions to lurasidone included drowsiness, restlessness, nausea, Parkinson-like symptoms, and sleeplessness [81][44]. Finally, lurasidone was associated with less weight gain and fewer metabolic disturbances than brexpiprazole [82,83][45][46].

3. Summary

Third-generation antipsychotics and lurasidone emerge as promising therapeutic strategies in the treatment of substance-induced psychoses. Aripiprazole has been effective in improving a wide range of psychotic symptoms, including both positive and negative aspects, as well as impacting substance use disorders positively. Its mode of action is unique; it is a partial agonist, meaning it does not stimulate receptors as strongly as full agonists. Its effectiveness lies in its dual role: it diminishes positive symptoms by antagonizing the mesolimbic dopamine pathway and improves negative symptoms and cognitive deficits by activating the mesocortical pathway. This selective mechanism helps aripiprazole to avoid severe side effects, like motor disorders and elevated prolactin levels, that are common for other antipsychotics. Additionally, it is known for its cardiac safety, causing negligible QTc prolongation and having a low risk of weight gain or sedation. In research with animals, aripiprazole has been observed to curb the heightened activity caused by stimulants, such as amphetamine, cocaine, and methylphenidate, and reduce their addictive qualities without hampering normal motor functions. It also reverses the lack of pleasure associated with amphetamine use and hinders the recurrence of cocaine-seeking behaviors. These findings indicate that aripiprazole might be effective in easing withdrawal symptoms associated with dopamine deficiency and, owing to its broad receptor activity, could represent a new strategy for achieving balanced dopamine levels in the treatment of drug addiction. Cariprazine’s receptor profile, particularly its high affinity for the D3 receptor and reduced intrinsic activity at D2 receptors, makes it an effective treatment for schizophrenia, improving both positive and negative symptoms. Its ability to modulate different neurotransmitter systems further highlights its potential as a distinct and effective antipsychotic medication. Cariprazine’s role in substance abuse treatment is noteworthy. Its effectiveness emerges in reducing the stimulating effects of substances, like cocaine, and mitigating cravings and relapses. This effect is possibly due to its partial agonism at D2 and D3 receptors. Case studies show cariprazine’s beneficial impact on schizophrenic patients with a history of substance abuse and its effectiveness in treating psychosis induced by substances, like methamphetamine. Brexpiprazole exhibits potential efficacy in the domain of substance abuse therapy. Primarily indicated for the management of schizophrenia and as an adjunctive treatment in major depressive disorder (MDD), its unique pharmacodynamic properties extend to the mitigation of substance-induced psychotic disorders. Operating as a partial agonist at the 5-HT1A and D2 neuroreceptors, brexpiprazole also engages with noradrenergic receptors. Consequently, albeit preliminary and limited in scope, research indicates its utility in addressing psychotic sequelae associated with cannabis consumption and in modulating dopaminergic activity in heroin-exposed rodents. Lurasidone has shown potential in treating psychopathological conditions related to substance abuse, although research in this area is limited. Known for its antagonistic action at dopamine D2 and serotonin 5-HT2A receptors and strong affinity for serotonin 5-HT7 receptors, lurasidone is unique in its partial agonism at 5-HT1A receptors. This receptor profile contributes to its low sedative effects and minimal impact on weight and cognitive functions, making it an appealing option in treating substance-induced psychoses. In the context of substance abuse, recent studies have shown lurasidone to be effective in treating young individuals with substance-induced psychosis, particularly from cannabis, improving various symptoms, including mood. It has also been beneficial for a complex case involving a young person with alcohol, cannabis, and LSD abuse along with behavioral and psychotic symptoms. It is important to consider that lurasidone is an approved medication for treating schizophrenia in individuals as young as 13, an age group particularly susceptible to substance use. Consequently, this could make it a feasible option for treatment in this younger demographic in the future.

References

  1. Martinotti, G.; De Risio, L.; Vannini, C.; Schifano, F.; Pettorruso, M.; Di Giannantonio, M. Substance-related exogenous psychosis: A postmodern syndrome. CNS Spectr. 2021, 26, 84–91.
  2. Orsolini, L.; Chiappini, S.; Papanti, D.; De Berardis, D.; Corkery, J.M.; Schifano, F. The Bridge Between Classical and “Synthetic”/Chemical Psychoses: Towards a Clinical, Psychopathological, and Therapeutic Perspective. Front. Psychiatry 2019, 10, 851.
  3. Schifano, F.; Napoletano, F.; Chiappini, S.; Guirguis, A.; Corkery, J.M.; Bonaccorso, S.; Ricciardi, A.; Scherbaum, N.; Vento, A. New/emerging psychoactive substances and associated psychopathological consequences. Psychol. Med. 2021, 51, 30–42.
  4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; Text Rev.; American Psychiatric Publishing: Washington, DC, USA, 2022.
  5. Schifano, F. Recent Changes in Drug Abuse Scenarios: The New/Novel Psychoactive Substances (NPS) Phenomenon. Brain Sci. 2018, 8, 221.
  6. Lieberman, J.A. Dopamine partial agonists: A new class of antipsychotic. CNS Drugs 2004, 18, 251–267.
  7. Orsolini, L.; Tomasetti, C.; Valchera, A.; Vecchiotti, R.; Matarazzo, I.; Vellante, F.; Iasevoli, F.; Buonaguro, E.F.; Fornaro, M.; Fiengo, A.L.; et al. An update of safety of clinically used atypical antipsychotics. Expert Opin. Drug Saf. 2016, 15, 1329–1347.
  8. Leite, J.V.; Guimarães, F.S.; Moreira, F.A. Aripiprazole, an atypical antipsychotic, prevents the motor hyperactivity induced by psychotomimetics and psychostimulants in mice. Eur. J. Pharmacol. 2008, 578, 222–227.
  9. Sørensen, G.; Sager, T.N.; Petersen, J.H.; Brennum, L.T.; Thøgersen, P.; Hee Bengtsen, C.; Thomsen, M.; Wörtwein, G.; Fink-Jensen, A.; Woldbye, D.P. Aripiprazole blocks acute self-administration of cocaine and is not self-administered in mice. Psychopharmacology 2008, 199, 37–46.
  10. Natesan, S.; Reckless, G.E.; Nobrega, J.N.; Fletcher, P.J.; Kapur, S. Dissociation between in vivo occupancy and functional antagonism of dopamine D2 receptors: Comparing aripiprazole to other antipsychotics in animal models. Neuropsychopharmacology 2006, 31, 1854–1863.
  11. Feltenstein, M.W.; Altar, C.A.; See, R.E. Aripiprazole blocks reinstatement of cocaine seeking in an animal model of relapse. Biol. Psychiatry 2007, 61, 582–590.
  12. Futamura, T.; Akiyama, S.; Sugino, H.; Forbes, A.; McQuade, R.D.; Kikuchi, T. Aripiprazole attenuates established behavioral sensitization induced by methamphetamine. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2010, 34, 1115–1119.
  13. Edinoff, A.; Ruoff, M.T.; Ghaffar, Y.T.; Rezayev, A.; Jani, D.; Kaye, A.M.; Cornett, E.M.; Kaye, A.D.; Viswanath, O.; Urits, I. Cariprazine to Treat Schizophrenia and Bipolar Disorder in Adults. Psychopharmacol. Bull. 2020, 50, 83–117.
  14. Stahl, S.M. Drugs for psychosis and mood: Unique actions at D3, D2, and D1 dopamine receptor subtypes. CNS Spectr. 2017, 22, 375–384.
  15. Calabrese, F.; Tarazi, F.I.; Racagni, G.; Riva, M.A. The role of dopamine D3 receptors in the mechanism of action of cariprazine. CNS Spectr. 2020, 25, 343–351.
  16. Kiss, B.; Horváth, A.; Némethy, Z.; Schmidt, E.; Laszlovszky, I.; Bugovics, G.; Fazekas, K.; Hornok, K.; Orosz, S.; Gyertyán, I.; et al. Cariprazine (RGH-188), a dopamine D3 receptor-preferring, D3/D2 dopamine receptor antagonist-partial agonist antipsychotic candidate: In vitro and neurochemical profile. J. Pharmacol. Exp. Ther. 2010, 333, 328–340.
  17. Kiss, B.; Krámos, B.; Laszlovszky, I. Potential Mechanisms for Why Not All Antipsychotics Are Able to Occupy Dopamine D3 Receptors in the Brain in vivo. Front. Psychiatry 2022, 13, 785592.
  18. Wesołowska, A.; Partyka, A.; Jastrzębska-Więsek, M.; Kołaczkowski, M. The preclinical discovery and development of cariprazine for the treatment of schizophrenia. Expert Opin. Drug Discov. 2018, 13, 779–790.
  19. Legros, C.; Rojas, A.; Dupré, C.; Brasseur, C.; Riest-Fery, I.; Muller, O.; Ortuno, J.C.; Nosjean, O.; Guenin, S.P.; Ferry, G.; et al. Approach to the specificity and selectivity between D2 and D3 receptors by mutagenesis and binding experiments part I: Expression and characterization of D2 and D3 receptor mutants. Protein Sci. A Publ. Protein Soc. 2022, 31, e4459.
  20. Legg, D.R.K.; Dubroff, J.G.; Labban, K.J.; Mach, R.H. Selectivity of probes for PET imaging of dopamine D3 receptors. Neurosci. Lett. 2019, 691, 18–25.
  21. Weerasinghe, D.K.; Hodge, J.M.; Pasco, J.A.; Samarasinghe, R.M.; Azimi Manavi, B.; Williams, L.J. Antipsychotic-induced bone loss: The role of dopamine, serotonin and adrenergic receptor signalling. Front. Cell Dev. Biol. 2023, 11, 1184550.
  22. Huang, M.; Panos, J.J.; Kwon, S.; Oyamada, Y.; Rajagopal, L.; Meltzer, H.Y. Comparative effect of lurasidone and blonanserin on cortical glutamate, dopamine, and acetylcholine efflux: Role of relative serotonin (5-HT)2A and DA D2 antagonism and 5-HT1A partial agonism. J. Neurochem. 2014, 128, 938–949.
  23. Girgis, R.R.; Slifstein, M.; D’Souza, D.; Lee, Y.; Periclou, A.; Ghahramani, P.; Laszlovszky, I.; Durgam, S.; Adham, N.; Nabulsi, N.; et al. Preferential binding to dopamine D3 over D2 receptors by cariprazine in patients with schizophrenia using PET with the D3/D2 receptor ligand -(+)-PHNO. Psychopharmacology 2016, 233, 3503–3512.
  24. Huang, M.; He, W.; Kiss, B.; Farkas, B.; Adham, N.; Meltzer, H.Y. The Role of Dopamine D3 Receptor Partial Agonism in Cariprazine-Induced Neurotransmitter Efflux in Rat Hippocampus and Nucleus Accumbens. J. Pharmacol. Exp. Ther. 2019, 371, 517–525.
  25. Yun, S.; Yang, B.; Anair, J.D.; Martin, M.M.; Fleps, S.W.; Pamukcu, A.; Yeh, N.H.; Contractor, A.; Kennedy, A.; Parker, J.G. Antipsychotic drug efficacy correlates with the modulation of D1 rather than D2 receptor-expressing striatal projection neurons. Nat. Neurosci. 2023, 26, 1417–1428.
  26. Sanson, A.; Riva, M.A. Anti-Stress Properties of Atypical Antipsychotics. Pharmaceuticals 2020, 13, 322.
  27. Németh, G.; Laszlovszky, I.; Czobor, P.; Szalai, E.; Szatmári, B.; Harsányi, J.; Barabássy, Á.; Debelle, M.; Durgam, S.; Bitter, I.; et al. Cariprazine versus risperidone monotherapy for treatment of predominant negative symptoms in patients with schizophrenia: A randomised, double-blind, controlled trial. Lancet 2017, 389, 1103–1113.
  28. Román, V.; Gyertyán, I.; Sághy, K.; Kiss, B.; Szombathelyi, Z. Cariprazine (RGH-188), a D₃-preferring dopamine D3/D2 receptor partial agonist antipsychotic candidate demonstrates anti-abuse potential in rats. Psychopharmacology 2013, 226, 285–293.
  29. Maeda, K.; Sugino, H.; Akazawa, H.; Amada, N.; Shimada, J.; Futamura, T.; Yamashita, H.; Ito, N.; McQuade, R.D.; Mørk, A.; et al. Brexpiprazole I: In vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. J. Pharmacol. Exp. Ther. 2014, 350, 589–604.
  30. Maeda, K.; Lerdrup, L.; Sugino, H.; Akazawa, H.; Amada, N.; McQuade, R.D.; Stensbøl, T.B.; Bundgaard, C.; Arnt, J.; Kikuchi, T. Brexpiprazole II: Antipsychotic-like and procognitive effects of a novel serotonin-dopamine activity modulator. J. Pharmacol. Exp. Ther. 2014, 350, 605–614.
  31. Stahl, S.M. Mechanism of action of brexpiprazole: Comparison with aripiprazole. CNS Spectr. 2016, 21, 1–6.
  32. Edinoff, A.N.; Wu, N.W.; Maxey, B.S.; Ren, A.L.; Leethy, K.N.; Girma, B.; Odisho, A.; Kaye, J.S.; Kaye, A.J.; Kaye, A.M.; et al. Brexpiprazole for the Treatment of Schizophrenia and Major Depressive Disorder: A Comprehensive Review of Pharmacological Considerations in Clinical Practice. Psychopharmacol. Bull. 2021, 51, 69–95.
  33. Brand, B.A.; Willemse, E.J.; Hamers, I.M.; Sommer, I.E. Evidence-Based Recommendations for the Pharmacological Treatment of Women with Schizophrenia Spectrum Disorders. Curr. Psychiatry Rep. 2023, 25, 723–733.
  34. Mauri, M.C.; Paletta, S.; Di Pace, C.; Reggiori, A.; Cirnigliaro, G.; Valli, I.; Altamura, A.C. Clinical Pharmacokinetics of Atypical Antipsychotics: An Update. Clin. Pharmacokinet. 2018, 57, 1493–1528.
  35. Kane, J.M.; Skuban, A.; Ouyang, J.; Hobart, M.; Pfister, S.; McQuade, R.D.; Nyilas, M.; Carson, W.H.; Sanchez, R.; Eriksson, H. A multicenter, randomized, double-blind, controlled phase 3 trial of fixed-dose brexpiprazole for the treatment of adults with acute schizophrenia. Schizophr. Res. 2015, 164, 127–135.
  36. Fleischhacker, W.W.; Hobart, M.; Ouyang, J.; Forbes, A.; Pfister, S.; McQuade, R.D.; Carson, W.H.; Sanchez, R.; Nyilas, M.; Weiller, E. Efficacy and safety of brexpiprazole (OPC-34712) as maintenance treatment in adults with schizophrenia: A randomized, double-blind, placebo-controlled study. Int. J. Neuropsychopharmacol. 2017, 20, 11–21.
  37. Siwek, M.; Wojtasik-Bakalarz, K.; Krupa, A.J.; Chrobak, A.A. Brexpiprazole-Pharmacologic Properties and Use in Schizophrenia and Mood Disorders. Brain Sci. 2023, 13, 397.
  38. Ishibashi, T.; Horisawa, T.; Tokuda, K.; Ishiyama, T.; Ogasa, M.; Tagashira, R.; Matsumoto, K.; Nishikawa, H.; Ueda, Y.; Toma, S.; et al. Pharmacological profile of lurasidone, a novel antipsychotic agent with potent 5-hydroxytryptamine 7 (5-HT7) and 5-HT1A receptor activity. J. Pharmacol. Exp. Ther. 2010, 334, 171–181.
  39. Miura, I.; Horikoshi, S.; Ichinose, M.; Suzuki, Y.; Watanabe, K. Lurasidone for the Treatment of Schizophrenia: Design, Development, and Place in Therapy. Drug Des. Dev. Ther. 2023, 17, 3023–3031.
  40. Sunovion. Latuda (Lurasidone HCl) Tablets: Highlights of Prescribing Information. Sunovion. 2013. Available online: http://www.latuda.com/LatudaPrescribingInformation.pdf (accessed on 20 January 2024).
  41. European Medicines Agency. Latuda: EPAR—Product Information. Annex I: Summary of Product Characteristics. EMA. 2014. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/latuda (accessed on 20 January 2024).
  42. Preskorn, S.; Ereshefsky, L.; Chiu, Y.Y.; Poola, N.; Loebel, A. Effect of food on the pharmacokinetics of lurasidone: Results of two randomized, open-label, crossover studies. Hum. Psychopharmacol. 2013, 28, 495–505.
  43. Chiu, Y.Y.; Ereshefsky, L.; Preskorn, S.H.; Poola, N.; Loebel, A. Lurasidone drug-drug interaction studies: A comprehensive review. Drug Metab. Drug Interact. 2014, 29, 191–202.
  44. Sanford, M. Lurasidone: In the Treatment of Schizophrenia. CNS Drugs 2013, 27, 67–80.
  45. Ng-Mak, D.; Tongbram, V.; Ndirangu, K.; Rajagopalan, K.; Loebel, A. Efficacy and Metabolic Effects of Lurasidone Versus Brexpiprazole in Schizophrenia: A Network Meta-Analysis. J. Comp. Eff. Res. 2018, 7, 737–748.
  46. Corponi, F.; Fabbri, C.; Bitter, I.; Montgomery, S.; Vieta, E.; Kasper, S.; Pallanti, S.; Serretti, A. Novel Antipsychotics Specificity Profile: A Clinically Oriented Review of Lurasidone, Brexpiprazole, Cariprazine, and Lumateperone. Eur. Neuropsychopharmacol. 2019, 29, 971–985.
More
Video Production Service