Antipsychotic Development: Comparison
Please note this is a comparison between Version 1 by Mujeeb Shad and Version 2 by Camila Xu.

While pharmacotherapy of depression appears to have moved from the monoamine hypothesis to more fertile grounds of glutamatergic and GABAergic mechanisms, it has been challenging to shift the antipsychotic paradigm beyond the dopaminergic hypothesis of schizophrenia. Efforts to develop non-dopaminergic antipsychotic medications (APMs) have produced negative results; thus, there has been no effective APM without dopamine involvement during 70 years of antipsychotic drug development.

  • seventy
  • years
  • antipsychotic
  • development

1. Introduction

While pharmacotherapy of depression appears to have moved from the monoamine hypothesis to more fertile grounds of glutamatergic and GABAergic mechanisms, it has been challenging to shift the antipsychotic paradigm beyond the dopaminergic hypothesis of schizophrenia. Efforts to develop non-dopaminergic antipsychotic medications (APMs) have produced negative results; thus, there has been no effective APM without dopamine involvement during 70 years of antipsychotic drug development. The only exception has been the approval of pimavanserin, a selective 5HT2A receptor blocker, to treat Parkinson’s psychosis but not schizophrenic psychosis.
Although Federal Drug Agency (FDA) has approved multiple APMs in the last 70 years, most have been with modest variations in molecular targets to qualify for “me too” drugs. The lack of significant differences in mechanisms of action explains why current APMs are primarily effective for positive symptoms without any measurable improvements in negative and cognitive symptoms. The antipsychotic response has been defined as only a 20% reduction in total scores on the most commonly used scale to assess schizophrenia symptoms, the Positive and Negative Syndrome Scale (PANSS) [1]. This reduction in response criterion accommodated the limited efficacy of current APMs for negative and general schizophrenia symptoms, including some cognitive symptoms, such as orientation, attention, insight, and judgment. However, a 20% reduction in total PANSS scores may be mediated by a completely different set of schizophrenia symptoms from one to the other APM, making it difficult to interpret and compare antipsychotic efficacy with precision.
Major psychiatric disorders are diagnosed at a syndromic level, with various symptom domains having potentially different neurobiological underpinnings. Still, most psychiatric studies have used atheoretical diagnoses based on the Diagnostic Statistical Manual [2][3] or the International Statistical Classification of Diseases and Related Health Problems (https://icd.who.int, accessed on 6 December 2022), which could explain the failure to develop comprehensive treatments. In addition, inadequate sample sizes have failed to account for heterogeneity across schizophrenia patients, not allowing for sub-group analyses to generate testable hypotheses and guide future research. These diagnostic limitations were addressed by restricting federal funding for prespecified Research Domain Criteria (RDoC) [4], which were vague, poorly defined, and not scientifically rigorous. Unfortunately, this measure was also counterproductive in promoting neurobiological research that could help advance psychiatric treatments.
Furthermore, researchers have failed to develop early predictors or intermediate phenotypes for antipsychotic response or tolerability, which could guide treatment options for individual patients. In addition, there is almost no research on finding the maintenance dose and length of antipsychotic therapy in schizophrenia patients recovering from acute psychosis. Similarly, there is a lack of laboratory measures to optimize antipsychotic response or tolerability, except for hyperprolactinemia (indicating excessive D2 receptor occupancy), therapeutic drug monitoring (TDM) for clozapine, and pharmacogenetic testing for refractory patients. Even though these measures can enhance antipsychotic efficacy and tolerability, particularly in treatment-refractory schizophrenia, they are not frequently employed, resulting in suboptimal use of APMs [5]. In addition, some providers use high antipsychotic doses, especially in severely and persistently ill patients in state hospitals and prison systems, compromising tolerability and medication adherence and resulting in repeated hospitalizations [5]. Furthermore, the antipsychotic doses required to manage acute psychosis and behavioral dyscontrol are not reduced even after the patient stabilizes. Several clinicians do not employ therapeutic drug monitoring for clozapine before labeling patients as clozapine non-responders [6].
Since NIMH does not fund clinical trials, developing new APMs is primarily owned by the pharmaceutical industry. Although this process involves billions of dollars and may not be fundable by the NIMH, post-marketing and repurposing trials for already approved agents can be affordable, providing a less biased approach than industry-sponsored trials. Another limitation is that the results from the preclinical trials for drug approval cannot be applied to the general population as they are completed in a near-ideal patient population without comorbidities. Thus, it takes years to gather post-marketing effectiveness data applicable to the general patient population. Furthermore, prescriptions of newly approved APMs are too expensive to be covered by Medicaid to be used in the severely and persistently ill, homeless, and forensic population that needs these medications the most.
Although a detailed discussion of the statistical limitations of clinical trials is beyond the scope of this resviearchw, the statistics using p < 0.05 has failed to translate into clinically relevant results [7]. However, this trend is changing to more clinically applicable measures, such as confidence interval, effect size, number needed to treat (NNT) [8], and number needed to harm (NNH) [9]. In addition, an increasing level of placebo response may have also failed several promising psychotropics, including APMs [10]. In this context, a higher placebo response can lower the chances of finding a significant difference (i.e., signal) between placebo and active treatment. Several strategies have been proposed to reduce placebo response, but none have been foolproof [11].
However, despite all these obstacles in developing more effective APMs, currently available treatments have made some progress in tolerability, if not efficacy. Thus, relatively higher dose thresholds for adverse effects are observed with second-generation APMs, partial agonists, and especially the recently approved multimodal antipsychotic lumateperone. It is also worth mentioning that long-acting injectables (LAIs) have significantly impacted long-term clinical and functional outcomes in schizophrenia patients worldwide. This resviearchw provides a synopsis of different mechanism-based classes of antipsychotic medications approved to manage schizophrenia and other psychotic disorders over the last 70 years.
While pharmacotherapy of depression appears to have moved from the monoamine hypothesis to more fertile grounds of glutamatergic and GABAergic mechanisms, it has been challenging to shift the antipsychotic paradigm beyond the dopaminergic hypothesis of schizophrenia. Efforts to develop non-dopaminergic antipsychotic medications (APMs) have been disappointing, with none of the currently available APMs moving beyond the dopamine system during 70 years of antipsychotic drug development. The only exception has been the approval of pimavanserin, a selective 5HT2A receptor blocker, to treat Parkinson’s psychosis but not schizophrenic psychosis.
Although Federal Drug Agency (FDA) has approved multiple APMs over the last seven decades, most have been with modest variations in molecular targets to qualify for “me too” drugs. The lack of significant differences in mechanisms of action explains why current APMs are primarily effective for positive symptoms without any clinically meaningful improvements in negative and cognitive symptoms. Therefore, the antipsychotic response is defined as only a 20% reduction in total scores on the Positive and Negative Syndrome Scale (PANSS) [1] instead of a 50% reduction required for an antidepressant response. This reduction in response criterion accommodated the limited efficacy of current APMs for negative and some of the cognitive symptoms assessed with PANSS, such as orientation, attention, insight, and judgment. A 20% reduction in PANSS scores suggests an antipsychotic response may be defined by a completely different set of symptoms from one patient to the other, making it difficult to interpret and compare antipsychotic efficacy with precision.
Major psychiatric disorders are diagnosed at a syndromic level, with various symptom domains having potentially different neurobiological underpinnings. However, most diagnoses in psychiatric research are based on atheoretical diagnostic tools such as the Diagnostic Statistical Manual [2][3] or the International Statistical Classification of Diseases and Related Health Problems (https://icd.who.int, accessed on 6 December 2022). The lack of neurobiologically-oriented diagnoses could explain the failure to develop more effective and comprehensive treatments. In addition, relatively small sample sizes have failed to account for heterogeneity across schizophrenia patients, not allowing for sub-group analyses to generate testable hypotheses and guide future research. The relative failure of DSM-based research was addressed by restricting federal funding to prespecified Research Domain Criteria (RDoC) [4], which were also vague, poorly defined, and not scientifically rigorous. Unfortunately, this measure was also counterproductive in promoting neurobiological research that could help advance psychiatric treatments.
Thus, second-generation APMs, including partial agonists, may have higher dosing thresholds to cause adverse effects, such as extrapyramidal symptoms (EPS) and hyperprolactinemia. It is also worth mentioning that long-acting injectables (LAIs) have significantly impacted long-term clinical and functional outcomes in schizophrenia patients worldwide.

2. Older Antipsychotic Medications

Despite limited efficacy and frequent adverse effects, older or conventional APMs provided the first effective treatment for schizophrenia patients, who did not have any chance to live outside asylums and the prison systems. These APMs are further subclassified into high potency and low potency. The low-potency conventional APMs are generally less tolerable than the high-potency conventional APMs, due to multiple sites of action representing a “shotgun” approach [12]. These actions include muscarinic, histaminic, and alpha-1 adrenergic receptor blockade. Antimuscarinic effects include dry mouth, blurred vision, urinary retention, constipation, tachycardia, loss of sweating, confusion, and worsening of closed-angle glaucoma and cognitive function. Antihistaminic effects translate into sedation and short-term weight gain, and the alpha-1 adrenergic blockade mediates postural hypotension, dizziness, and sedation. In contrast, high-potency APMs are relatively more selective for blocking dopamine-2 (D2) receptors, and the main adverse effects associated with them are due to D2 blockade, especially at high doses. These adverse effects include extrapyramidal symptoms (EPS), hyperprolactinemia (Figure 1 and Figure 2), and treatment-induced, or worsening of, pre-existing negative, affective or cognitive symptoms.
Figure 1.
Extrapyramidal Symptoms (EPS) with antipsychotic medications.
Figure 2.
Hyperprolactinemia with antipsychotic medications.
However, the most clinically-serious adverse effect of low-potency APMs is the effects on QTc prolongation, which can result in sudden cardiac death [13]. Although the high-potency APMs also prolong QTc interval, the most clinically serious effects include tardive dyskinesia [14] and neuroleptic malignant syndrome [15]. These adverse effects are generally reported with long-term use of high-dose antipsychotic pharmacotherapy [15]. However, some of the adverse effects of low-potency conventional APMs may be beneficial in few patients, including sedation and weight gain with the histamine-1 receptor blockade, reduction in pre-existing hypertension with the alpha-1 receptor blockade, and protection from EPS with the built-in anticholinergic effects. These adverse effects are also witnessed with newer APMs, such as olanzapine and clozapine [16]. Of note, none of the APMs is indicted for managing dementia-related psychosis due to increased risk for mortality in older adults [17]. There is one APM, loxapine, that is not only effective in schizophrenia but also in major depressive disorder because loxapine is demethylated to a tetracyclic metabolite, amoxapine, which has antidepressant properties [18]. Another worth-mentioning APM, molindone, has a moderate affinity for D2 receptors, with potential benefits in patients not responding to high-or low-affinity APMs [19]. It is also interesting that one of the older high-potency APM, pimozide is the only FDA-approved treatment for Tourette’s syndrome [20]. One of the most clinically significant advances in antipsychotic treatments has been the development of long-acting injectables (LAI) with high-potency APMs, haloperidol, and fluphenazine, followed by newer APMs, risperidone, paliperidone, and aripiprazole. Since medication nonadherence is commonly observed in schizophrenia subjects, the LAIs have improved the maintenance of antipsychotic response and prevention of psychotic relapse and rehospitalizations [21]. In addition, these LAIs are now also available with some of the newer generation APMs as discussed below. In summary, serotonin and dopamine antagonism (SDA) with SGAPMs may have provided some cushion from dose-related adverse effects but without any significant benefits in efficacy. The only exception among SGAPMs is clozapine, which remains the gold standard for managing treatment-refractory schizophrenia (TRS). However, despite its unique efficacy, to ouresearchers' knowledge, clozapine has not been compared with other SGAMs or studied in post-marketing effectiveness or efficacy trials because it is only approved for patients with TRS.

3. Second-Generation Antipsychotic Medications (SGAPMs)

This class includes every APM developed after the conventional APMs despite significant differences in the mechanisms of action (Table 1), which has created confusion and misperceptions.
Table 1.
Molecular targets and binding affinities (Ki, nM) for antipsychotic medications.

References

  1. Kay, S.R.; Fiszbein, A.; Opler, L.A. The positive and negative syndrome scale (PANSS) for schizophrenia. Schizophr. Bull. 1987, 13, 261–276.
  2. Allsopp, K.; Read, J.; Corcoran, R.; Kinderman, P. Heterogeneity in psychiatric diagnostic classification. Psychiatry Res. 2019, 279, 15–22.
  3. Escobar, J.I. An insider’s view of the new diagnostic and statistical manual of North American psychiatry (DSM-5). Colomb. Médica 2013, 44, 129–131.
  4. Markowitz, J.C.; Milrod, B.L. Lost in Translation: The Value of Psychiatric Clinical Trials. J. Clin. Psychiatry 2022, 83, 43385.
  5. Shad, M.U. High-Dose Therapy in Treatment-Refractory Psychosis: A Retrospective Study. Prim. Care Companion CNS Disord. 2022, 24, 44191.
  6. Shad, M.U.; Felzien, E.; Roy, K.; Sethi, S. How to identify and manage non-response to clozapine? Asian J. Psychiatr. 2019, 45, 50–52.
  7. Grabowski, B. “P < 0.05” Might Not Mean What You Think: American Statistical Association Clarifies P Values. JNCI J. Natl. Cancer Inst. 2016, 108, djw194.
  8. Citrome, L. Number needed to treat: What it is and what it isn’t, and why every clinician should know how to calculate it. J. Clin. Psychiatry 2011, 72, 412–413.
  9. Andrade, C. The numbers needed to treat and harm (NNT, NNH) statistics: What they tell us and what they do not. J. Clin. Psychiatry 2015, 76, e330–e333.
  10. Kasper, S.; Dold, M. Factors contributing to the increasing placebo response in antidepressant trials. World Psychiatry 2015, 14, 304–306.
  11. Evans, K.; Colloca, L.; Pecina, M.; Katz, N. What can be done to control the placebo response in clinical trials? A narrative review. Contemp. Clin. Trials 2021, 107, 106503.
  12. Gardner, D.M.; Baldessarini, R.J.; Waraich, P. Modern antipsychotic drugs: A critical overview. Cmaj 2005, 172, 1703–1711.
  13. Preskorn, S.H. The evolution of antipsychotic drug therapy: Reserpine, chlorpromazine, and haloperidol. J. Psychiatr. Pract. 2007, 13, 253–257.
  14. Vardar, M.K.; Ceylan, M.E.; Unsalver, B.O. Assesment of Risk Factors for Tardive Dyskinesia. Psychopharmacol. Bull. 2020, 50, 36–46.
  15. Sarkar, S.; Gupta, N. Drug information update. Atypical antipsychotics and neuroleptic malignant syndrome: Nuances and pragmatics of the association. BJPsych Bull. 2017, 41, 211–216.
  16. Chew, M.L.; Mulsant, B.H.; Pollock, B.G.; Lehman, M.E.; Greenspan, A.; Kirshner, M.A.; Bies, R.R.; Kapur, S.; Gharabawi, G. A model of anticholinergic activity of atypical antipsychotic medications. Schizophr. Res. 2006, 88, 63–72.
  17. Jeste, D.V.; Blazer, D.; Casey, D.; Meeks, T.; Salzman, C.; Schneider, L.; Tariot, P.; Yaffe, K. ACNP White Paper: Update on use of antipsychotic drugs in elderly persons with dementia. Neuropsychopharmacology 2008, 33, 957–970.
  18. Burch, E.A., Jr.; Goldschmidt, T.J. Loxapine in the treatment of psychotic-depressive disorders: Measurement of antidepressant metabolites. South. Med. J. 1983, 76, 991–995.
  19. Waugaman, R.M. Potential lower efficacy of molindone among first-generation antipsychotics. Am. J. Psychiatry 2009, 166, 491.
  20. Pringsheim, T.; Marras, C. Pimozide for tics in Tourette’s syndrome. Cochrane Database Syst. Rev. 2009, 2009, CD006996.
  21. Kane, J.M.; Leucht, S.; Carpenter, D.; Docherty, J.P. Expert Consensus Panel for Optimizing Pharmacologic Treatment of Psychotic D. The expert consensus guideline series. Optimizing pharmacologic treatment of psychotic disorders. Introduction: Methods, commentary, and summary. J. Clin. Psychiatry 2003, 64 (Suppl. 12), 5–19.
  22. Arnt, J.; Skarsfeldt, T. Do novel antipsychotics have similar pharmacological characteristics? A review of the evidence. Neuropsychopharmacology 1998, 18, 63–101.
  23. Bymaster, F.P.; Calligaro, D.O.; Falcone, J.F.; Marsh, R.D.; Moore, N.A.; Tye, N.C.; Seeman, P.; Wong, D.T. Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology 1996, 14, 87–96.
  24. Seeger, T.F.; Seymour, P.A.; Schmidt, A.W.; Zorn, S.H.; Schulz, D.W.; Lebel, L.A.; McLean, S.; Guanowsky, V.; Howard, H.R.; Lowe, J.A. Ziprasidone (CP-88,059): A new antipsychotic with combined dopamine and serotonin receptor antagonist activity. J. Pharmacol. Exp. Ther. 1995, 275, 101–113.
  25. Schotte, A.; Janssen, P.F.M.; Gommeren, W.; Luyten, W.H.M.L.; Van Gompel, P.; Lesage, A.S.; De Loore, K.; Leysen, J.E. Risperidone compared with new and reference antipsychotic drugs: In vitro and in vivo receptor binding. Psychopharmacology 1996, 124, 57–73.
  26. 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.
  27. Herman, A.; El Mansari, M.; Adham, N.; Kiss, B.; Farkas, B.; Blier, P. Involvement of 5-HT(1A) and 5-HT(2A) Receptors but Not alpha (2)-Adrenoceptors in the Acute Electrophysiological Effects of Cariprazine in the Rat Brain In Vivo. Mol. Pharmacol. 2018, 94, 1363–1370.
  28. Citrome, L. Iloperidone: Chemistry, pharmacodynamics, pharmacokinetics and metabolism, clinical efficacy, safety and tolerability, regulatory affairs, and an opinion. Expert Opin. Drug Metab. Toxicol. 2010, 6, 1551–1564.
  29. Citrome, L. A review of the pharmacology, efficacy and tolerability of recently approved and upcoming oral antipsychotics: An evidence-based medicine approach. CNS Drugs 2013, 27, 879–911.
  30. Corena-McLeod, M. Comparative Pharmacology of Risperidone and Paliperidone. Drugs R D 2015, 15, 163–174.
  31. Wenthur, C.J.; Lindsley, C.W. Classics in chemical neuroscience: Clozapine. ACS Chem. Neurosci. 2013, 4, 1018–1025.
  32. Popovic, D.; Nuss, P.; Vieta, E. Revisiting loxapine: A systematic review. Ann. Gen. Psychiatry 2015, 14, 15.
  33. Farah, A. Atypicality of atypical antipsychotics. Prim. Care Companion CNS Disord. 2005, 7, 268–274.
  34. David, S.R.; Taylor, C.C.; Kinon, B.J.; Breier, A. The effects of olanzapine, risperidone, and haloperidol on plasma prolactin levels in patients with schizophrenia. Clin. Ther. 2000, 22, 1085–1096.
  35. Kerwin, R.W.; Bolonna, A.A. Is clozapine antisuicidal? Expert Rev. Neurother. 2004, 4, 187–190.
  36. Joober, R.; Boksa, P. Clozapine: A distinct, poorly understood and under-used molecule. J. Psychiatry Neurosci. 2010, 35, 147–149.
  37. Schulte, P. Risk of clozapine-associated agranulocytosis and mandatory white blood cell monitoring. Ann. Pharmacother. 2006, 40, 683–688.
  38. Patel, N.C.; Dorson, P.G.; Bettinger, T.L. Sudden late onset of clozapine-induced agranulocytosis. Ann. Pharmacother. 2002, 36, 1012–1015.
  39. Higgins, J.M.; San, C.; Lagnado, G.; Chua, D.; Mihic, T. Incidence and Management of Clozapine-Induced Myocarditis in a Large Tertiary Hospital. Can. J. Psychiatry 2019, 64, 561–567.
  40. De Fazio, P.; Gaetano, R.; Caroleo, M.; Cerminara, G.; Maida, F.; Bruno, A.; Muscatello, M.R.; Moreno, M.J.J.; Russo, E.; Segura-García, C. Rare and very rare adverse effects of clozapine. Neuropsychiatr. Dis. Treat. 2015, 11, 1995–2003.
  41. Maher, S.; Cunningham, A.; O’Callaghan, N.; Byrne, F.; Mc Donald, C.; McInerney, S.; Hallahan, B. Clozapine-induced hypersalivation: An estimate of prevalence, severity and impact on quality of life. Ther. Adv. Psychopharmacol. 2016, 6, 178–184.
  42. Seeman, P. Clozapine, a fast-off-D2 antipsychotic. ACS Chem. Neurosci. 2014, 5, 24–29.
  43. Kapur, S.; Zipursky, R.; Jones, C.; Shammi, C.S.; Remington, G.; Seeman, P. A positron emission tomography study of quetiapine in schizophrenia: A preliminary finding of an antipsychotic effect with only transiently high dopamine D2 receptor occupancy. Arch. Gen. Psychiatry 2000, 57, 553–559.
  44. 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.
  45. Shad, M.U. Clozapine toxicity: A discussion of pharmacokinetic factors. Asian J. Psychiatry 2008, 1, 47–49.
  46. Prior, T.I.; Baker, G.B. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J. Psychiatry Neurosci. 2003, 28, 99–112.
  47. Costa-Dookhan, K.A.; Agarwal, S.M.; Chintoh, A.; Tran, V.N.; Stogios, N.; Ebdrup, B.H.; Sockalingam, S.; Rajji, T.K.; Remington, G.J.; Siskind, D.; et al. The clozapine to norclozapine ratio: A narrative review of the clinical utility to minimize metabolic risk and enhance clozapine efficacy. Expert Opin. Drug Saf. 2020, 19, 43–57.
  48. Kroon, L.A. Drug interactions with smoking. Am. J. Health Pharm. 2007, 64, 1917–1921.
  49. Stieffenhofer, V.; Saglam, H.; Schmidtmann, I.; Silver, H.; Hiemke, C.; Konrad, A. Clozapine plasma level monitoring for prediction of rehospitalization schizophrenic outpatients. Pharmacopsychiatry 2011, 44, 55–59.
  50. Dettling, M.; Sachse, C.; Brockmöller, J.; Schley, J.; Müller-Oerlinghausen, B.; Pickersgill, I.; Rolfs, A.; Schaub, R.T.; Schmider, J. Long-term therapeutic drug monitoring of clozapine and metabolites in psychiatric in- and outpatients. Psychopharmacology 2000, 152, 80–86.
  51. Greenwood-Smith, C.; Lubman, D.I.; Castle, D.J. Serum clozapine levels: A review of their clinical utility. J. Psychopharmacol. 2003, 17, 234–238.
  52. Olesen, O.V.; Thomsen, K.; Jensen, P.N.; Wulff, C.H.; Rasmussen, N.-A.; Refshammer, C.; Sørensen, J.; Bysted, M.; Christensen, J.; Rosenberg, R. Clozapine serum levels and side effects during steady state treatment of schizophrenic patients: A cross-sectional study. Psychopharmacology 1995, 117, 371–378.
  53. Tiihonen, J.; Wahlbeck, K.; Kiviniemi, V. The efficacy of lamotrigine in clozapine-resistant schizophrenia: A systematic review and meta-analysis. Schizophr. Res. 2009, 109, 10–14.
  54. Ma, X.; Maimaitirexiati, T.; Zhang, R.; Gui, X.; Zhang, W.; Xu, G.; Hu, G. HTR2C polymorphisms, olanzapine-induced weight gain and antipsychotic-induced metabolic syndrome in schizophrenia patients: A meta-analysis. Int. J. Psychiatry Clin. Pract. 2014, 18, 229–242.
  55. Grover, S.; Nebhinani, N.; Chakrabarti, S.; Avasthi, A.; Kulhara, P. Metabolic syndrome among patients receiving clozapine: A preliminary estimate. Indian J. Pharmacol. 2011, 43, 591–595.
  56. Lieberman, J.A.; Stroup, T.S.; McEvoy, J.P.; Swartz, M.S.; Rosenheck, R.A.; Perkins, D.O.; Keefe, R.S.E.; Davis, S.M.; Davis, C.E.; Lebowitz, B.D.; et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N. Engl. J. Med. 2005, 353, 1209–1223.
  57. Meyer, J.M.; Nasrallah, H.A.; McEvoy, J.P.; Goff, D.C.; Davis, S.M.; Chakos, M.; Patel, J.K.; Keefe, R.S.; Stroup, T.S.; Lieberman, J.A. The Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Schizophrenia Trial: Clinical comparison of subgroups with and without the metabolic syndrome. Schizophr. Res. 2005, 80, 9–18.
  58. Lindenmayer, J.P. Long-acting injectable antipsychotics: Focus on olanzapine pamoate. Neuropsychiatr. Dis. Treat. 2010, 6, 261–267.
  59. De Silva, V.A.; Suraweera, C.; Ratnatunga, S.S.; Dayabandara, M.; Wanniarachchi, N.; Hanwella, R. Metformin in prevention and treatment of antipsychotic induced weight gain: A systematic review and meta-analysis. BMC Psychiatry 2016, 16, 341.
  60. Correll, C.U.; Newcomer, J.W.; Silverman, B.; DiPetrillo, L.; Graham, C.; Jiang, Y.; Du, Y.; Simmons, A.; Hopkinson, C.; McDonnell, D.; et al. Effects of Olanzapine Combined with Samidorphan on Weight Gain in Schizophrenia: A 24-Week Phase 3 Study. Am. J. Psychiatry 2020, 177, 1168–1178.
  61. Brunette, M.F.; Correll, C.U.; O’Malley, S.S.; McDonnell, D.; DiPetrillo, L.; Jiang, Y.; Simmons, A.; Silverman, B.L.; Citrome, L.; Green, A.I. Olanzapine Plus Samidorphan (ALKS 3831) in Schizophrenia and Comorbid Alcohol Use Disorder: A Phase 2, Randomized Clinical Trial. J. Clin. Psychiatry 2020, 81, 13176.
  62. Hamner, M. The effects of atypical antipsychotics on serum prolactin levels. Ann. Clin. Psychiatry 2002, 14, 163–173.
  63. Sparshatt, A.; Jones, S.; Taylor, D. Quetiapine: Dose-response relationship in schizophrenia. CNS Drugs 2008, 22, 49–68; discussion 69–72.
  64. Shotbolt, P.; Samuel, M.; David, A. Quetiapine in the treatment of psychosis in Parkinson’s disease. Ther. Adv. Neurol. Disord. 2010, 3, 339–350.
  65. Zhong, K.X.; Tariot, P.N.; Mintzer, J.; Minkwitz, M.C.; Devine, N.A. Quetiapine to treat agitation in dementia: A randomized, double-blind, placebo-controlled study. Curr. Alzheimer Res. 2007, 4, 81–93.
  66. Patel, R.S.; Bhela, J.; Tahir, M.; Pisati, S.R.; Hossain, S. Pimavanserin in Parkinson’s Disease-induced Psychosis: A Literature Review. Cureus 2019, 11, e5257.
  67. Ohman, K.L.; Schultheis, J.M.; Kram, S.J.; Cox, C.E.; Gilstrap, D.L.; Yang, Z.; Kram, B.L. Effectiveness of Quetiapine as a Sedative Adjunct in Mechanically Ventilated Adults without Delirium. Ann. Pharmacother. 2021, 55, 149–156.
  68. Park, S.; Yi, K.K.; Kim, M.S.; Hong, J.P. Effects of ziprasidone and olanzapine on body composition and metabolic parameters: An open-label comparative pilot study. Behav. Brain Funct. 2013, 9, 27.
  69. Camm, A.J.; Karayal, O.N.; Meltzer, H.; Kolluri, S.; O’Gorman, C.; Miceli, J.; Tensfeldt, T.; Kane, J.M. Ziprasidone and the corrected QT interval: A comprehensive summary of clinical data. CNS Drugs 2012, 26, 351–365.
  70. Zimbroff, D.L.; Allen, M.H.; Battaglia, J.; Citrome, L.; Fishkind, A.; Francis, A.; Herr, D.L.; Hughes, D.; Martel, M.; Preval, H.; et al. Best clinical practice with ziprasidone IM: Update after 2 years of experience. CNS Spectr. 2005, 10, 1–15.
  71. Miller, D.D. Atypical antipsychotics: Sleep, sedation, and efficacy. Prim. Care Companion J. Clin. Psychiatry 2004, 6 (Suppl. 2), 3–7.
  72. Keating, A.M.; Aoun, S.L.; Dean, C.E. Ziprasidone-associated mania: A review and report of 2 additional cases. Clin. Neuropharmacol. 2005, 28, 83–86.
  73. Masand, P.S.; Nemeroff, C.B.; Newcomer, J.W.; Lieberman, J.A.; Schatzberg, A.F.; Weiden, P.J.; Kilts, C.D.; Harvey, P.D.; Daniel, D.G. From clinical research to clinical practice: A 4-year review of ziprasidone. CNS Spectr. 2005, 10 (Suppl. 17), 1–20.
  74. Carnahan, R.M.; Lund, B.C.; Perry, P.J. Ziprasidone, a new atypical antipsychotic drug. Pharmacotherapy 2001, 21, 717–730.
  75. Green, B. Focus on ziprasidone. Curr. Med. Res. Opin. 2001, 17, 146–150.
  76. Papakostas, G.I.; Fava, M.; Baer, L.; Swee, M.B.; Jaeger, A.; Bobo, W.V.; Shelton, R.C. Ziprasidone Augmentation of Escitalopram for Major Depressive Disorder: Efficacy Results from a Randomized, Double-Blind, Placebo-Controlled Study. Am. J. Psychiatry 2015, 172, 1251–1258.
  77. Shad, M.; Preskorn, S.; Miceli, J.; Wilner, K. Use of population pharmacokinetic modeling to characterize intramuscular pharmacokinetics for ziprasidone in schizophrenic patients. Clin. Pharmacol. Ther. 1999, 65, 171.
  78. Spina, E.; de Leon, J. Metabolic drug interactions with newer antipsychotics: A comparative review. Basic Clin. Pharmacol. Toxicol. 2007, 100, 4–22.
  79. Citrome, L. Using oral ziprasidone effectively: The food effect and dose-response. Adv. Ther. 2009, 26, 739–748.
  80. Urichuk, L.; Prior, T.I.; Dursun, S.; Baker, G. Metabolism of atypical antipsychotics: Involvement of cytochrome p450 enzymes and relevance for drug-drug interactions. Curr. Drug Metab. 2008, 9, 410–418.
  81. Amatniek, J.; Canuso, C.M.; Deutsch, S.I.; Henderson, D.C.; Mao, L.; Mikesell, C.; Rodriguez, S.; Sheehan, J.; Alphs, L. Safety of paliperidone extended-release in patients with schizophrenia or schizoaffective disorder and hepatic disease. Clin. Schizophr. Relat. Psychoses 2014, 8, 8–20.
  82. Chan, H.W.; Huang, C.Y.; Yen, Y.C. Clinical outcomes of paliperidone long-acting injection in patients with schizophrenia: A 1-year retrospective cohort study. BMC Psychiatry 2021, 21, 507.
  83. Caccia, S.; Pasina, L.; Nobili, A. New atypical antipsychotics for schizophrenia: Iloperidone. Drug Des. Dev. Ther. 2010, 4, 33–48.
  84. Citrome, L. Iloperidone: A clinical overview. J. Clin. Psychiatry 2011, 72 (Suppl. 1), 19–23.
  85. Shuman, M.D.; McGrane, I.R. Rationale for iloperidone in the treatment of posttraumatic stress disorder. Innov. Clin. Neurosci. 2014, 11, 23–25.
  86. Joshi, S.V.; Patel, E.P.; Vyas, B.A.; Lodha, S.R.; Kalyankar, G.G. Repurposing of Iloperidone: Antihypertensive and ocular hypotensive activity in animals. Eur. J. Pharm. Sci. 2020, 143, 105173.
  87. Llerena, A.; Berecz, R.; Dorado, P.; de la Rubia, A. QTc interval, CYP2D6 and CYP2C9 genotypes and risperidone plasma concentrations. J. Psychopharmacol. 2004, 18, 189–193.
  88. Pratts, M.; Citrome, L.; Grant, W.; Leso, L.; Opler, L.A. A single-dose, randomized, double-blind, placebo-controlled trial of sublingual asenapine for acute agitation. Acta Psychiatr. Scand. 2014, 130, 61–68.
  89. Citrome, L. Iloperidone, asenapine, and lurasidone: A brief overview of 3 new second-generation antipsychotics. Postgrad. Med. 2011, 123, 153–162.
  90. McIntyre, R.S.; Wong, R. Asenapine: A synthesis of efficacy data in bipolar mania and schizophrenia. Clin. Schizophr. Relat. Psychoses 2012, 5, 217–220.
  91. Tarazi, F.I.; Neill, J.C. The preclinical profile of asenapine: Clinical relevance for the treatment of schizophrenia and bipolar mania. Expert Opin. Drug Discov. 2013, 8, 93–103.
  92. Ballaz, S.J.; Akil, H.; Watson, S.J. The 5-HT7 receptor: Role in novel object discrimination and relation to novelty-seeking behavior. Neuroscience 2007, 149, 192–202.
  93. Shayegan, D.K.; Stahl, S.M. Atypical antipsychotics: Matching receptor profile to individual patient’s clinical profile. CNS Spectr. 2004, 9 (Suppl. 11), 6–14.
  94. Kroeze, W.K.; Hufeisen, S.J.; Popadak, B.A.; Renock, S.M.; Steinberg, S.; Ernsberger, P.; Jayathilake, K.; Meltzer, H.Y.; Roth, B.L. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology 2003, 28, 519–526.
  95. Potkin, S.G.; Keator, D.B.; Kesler-West, M.L.; Nguyen, D.D.; van Erp, T.G.M.; Mukherjee, J.; Shah, N.; Preda, A. D2 receptor occupancy following lurasidone treatment in patients with schizophrenia or schizoaffective disorder. CNS Spectr. 2014, 19, 176–181.
  96. Meyer, J.M.; Loebel, A.D.; Schweizer, E. Lurasidone: A new drug in development for schizophrenia. Expert Opin. Investig. Drugs 2009, 18, 1715–1726.
  97. Grunder, G.; Carlsson, A.; Wong, D.F. Mechanism of new antipsychotic medications: Occupancy is not just antagonism. Arch. Gen. Psychiatry 2003, 60, 974–977.
  98. Stahl, S.M. Dopamine system stabilizers, aripiprazole, and the next generation of antipsychotics, part 2: Illustrating their mechanism of action. J. Clin. Psychiatry 2001, 62, 923–924.
  99. Stahl, S.M. Dopamine system stabilizers, aripiprazole, and the next generation of antipsychotics, part 1, “Goldilocks” actions at dopamine receptors. J. Clin. Psychiatry 2001, 62, 841–842.
  100. Nasrallah, H.A.; Mulvihill, T. Iatrogenic disorders associated with conventional vs. atypical antipsychotics. Ann. Clin. Psychiatry 2001, 13, 215–227.
  101. Kim, D.D.; Barr, A.M.; Lian, L.; Yuen, J.W.Y.; Fredrikson, D.; Honer, W.G.; Thornton, A.E.; Procyshyn, R.M. Efficacy and tolerability of aripiprazole versus D(2) antagonists in the early course of schizophrenia: A systematic review and meta-analysis. Npj Schizophr. 2021, 7, 29.
  102. Singh, T. Aripiprazole-induced weight gain. Psychiatry 2005, 2, 19.
  103. Kelly, D.L.; Powell, M.M.; Wehring, H.J.; Sayer, M.A.; Kearns, A.M.; Hackman, A.L.; Buchanan, R.; Nichols, R.B.; McEvoy, J.P.; Adams, H.A.; et al. Adjunct Aripiprazole Reduces Prolactin and Prolactin-Related Adverse Effects in Premenopausal Women with Psychosis: Results from the DAAMSEL Clinical Trial. J. Clin. Psychopharmacol. 2018, 38, 317–326.
  104. Yeager, A.; Shad, M.U. Aripiprazole for the Management of Antipsychotic-Induced Hyperprolactinemia: A Retrospective Case Series. Prim. Care Companion CNS Disord. 2020, 22, 26648.
  105. Nguyen, C.T.; Rosen, J.A.; Bota, R.G. Aripiprazole partial agonism at 5-HT2C: A comparison of weight gain associated with aripiprazole adjunctive to antidepressants with high versus low serotonergic activities. Prim. Care Companion CNS Disord. 2012, 14, 26654.
  106. Citrome, L. A review of aripiprazole in the treatment of patients with schizophrenia or bipolar I disorder. Neuropsychiatr. Dis. Treat. 2006, 2, 427–443.
  107. Fryefield, K.; Shad, M.U. Can low-dose aripiprazole reverse some of the adverse effects from a long-acting injectable? Schizophr. Res. 2019, 204, 417–418.
  108. Potkin, S.G.; Preda, A. Aripiprazole once-monthly long-acting injectable for the treatment of schizophrenia. Expert Opin. Pharmacother. 2016, 17, 395–407.
  109. Cruz, M.P. Aripiprazole Lauroxil (Aristada): An Extended-Release, Long-Acting Injection For the Treatment of Schizophrenia. Pharm. Ther. 2016, 41, 556–559.
  110. Ehret, M.J.; Davis, E.; Luttrell, S.E.; Clark, C. Aripiprazole Lauroxil NanoCrystal® Dispersion Technology (Aristada Initio®). Clin. Schizophr. Relat. Psychoses 2018, 12, 92–96.
  111. Di Lorenzo, R.; Ferri, P.; Cameli, M.; Rovesti, S.; Piemonte, C. Effectiveness of 1-year treatment with long-acting formulation of aripiprazole, haloperidol, or paliperidone in patients with schizophrenia: Retrospective study in a real-world clinical setting. Neuropsychiatr. Dis. Treat. 2019, 15, 183–198.
  112. Mason, K.; Barnett, J.; Pappa, S. Effectiveness of 2-year treatment with aripiprazole long-acting injectable and comparison with paliperidone palmitate. Ther. Adv. Psychopharmacol. 2021, 11, 20451253211029490.
  113. Kishimoto, T.; Hagi, K.; Nitta, M.; Leucht, S.; Olfson, M.; Kane, J.M.; Correll, C.U. Effectiveness of Long-Acting Injectable vs Oral Antipsychotics in Patients with Schizophrenia: A Meta-analysis of Prospective and Retrospective Cohort Studies. Schizophr. Bull. 2018, 44, 603–619.
  114. Tiihonen, J.; Mittendorfer-Rutz, E.; Majak, M.; Mehtälä, 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 29,823 Patients with Schizophrenia. JAMA Psychiatry 2017, 74, 686–693.
  115. Garnock-Jones, K.P. Cariprazine: A Review in Schizophrenia. CNS Drugs 2017, 31, 513–525.
  116. Campbell, R.H.; Diduch, M.; Gardner, K.N.; Thomas, C. Review of cariprazine in management of psychiatric illness. Ment. Health Clin. 2017, 7, 221–229.
  117. McCormack, P.L. Cariprazine: First Global Approval. Drugs 2015, 75, 2035–2043.
  118. Durgam, S.; Earley, W.; Guo, H.; Li, D.; Németh, G.; Laszlovszky, I.; Fava, M.; Montgomery, S.A. Efficacy and safety of adjunctive cariprazine in inadequate responders to antidepressants: A randomized, double-blind, placebo-controlled study in adult patients with major depressive disorder. J. Clin. Psychiatry 2016, 77, 371–378.
  119. Stahl, S.M. Mechanism of action of cariprazine. CNS Spectr. 2016, 21, 123–127.
  120. Thase, M.E.; Youakim, J.M.; Skuban, A.; Hobart, M.; Zhang, P.; McQuade, R.D.; Nyilas, M.; Carson, W.H.; Sanchez, R.; Eriksson, H. Adjunctive brexpiprazole 1 and 3 mg for patients with major depressive disorder following inadequate response to antidepressants: A phase 3, randomized, double-blind study. J. Clin. Psychiatry 2015, 76, 1232–1240.
  121. 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.
  122. Citrome, L. Brexpiprazole for schizophrenia and as adjunct for major depressive disorder: A systematic review of the efficacy and safety profile for this newly approved antipsychotic-what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int. J. Clin. Pract. 2015, 69, 978–997.
  123. Stahl, S.M. Mechanism of action of brexpiprazole: Comparison with aripiprazole. CNS Spectr. 2016, 21, 1–6.
  124. Correll, C.U.; Skuban, A.; Ouyang, J.; Hobart, M.; Pfister, S.; McQuade, R.D.; Nyilas, M.; Carson, W.H.; Sanchez, R.; Eriksson, H. Efficacy and Safety of Brexpiprazole for the Treatment of Acute Schizophrenia: A 6-Week Randomized, Double-Blind, Placebo-Controlled Trial. Am. J. Psychiatry 2015, 172, 870–880.
  125. Ward, K.; Citrome, L. Brexpiprazole for the maintenance treatment of adults with schizophrenia: An evidence-based review and place in therapy. Neuropsychiatr. Dis. Treat. 2019, 15, 247–257.
More