Skip to main content
Log in

Upcoming Agents for the Treatment of Schizophrenia

Mechanism of Action, Efficacy and Tolerability

  • Leading Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Since the introduction of a group of atypical antipsychotics in the 1990s, there has been a decline in the rate of new antipsychotics being introduced into clinical practice. However, with increasing safety and efficacy concerns over currently available drugs and a dearth of options available for atypical depot formulations, there is a considerable need for the development of new formulations and agents. This review examines the profile of seven antipsychotic drugs currently in the premarketing stage of development and summarizes their mechanism of action, clinical potential and safety.

Asenapine is an antipsychotic with activity for multiple receptors and has potential to improve negative and cognitive symptoms of schizophrenia. Bifeprunox is a partial dopamine D2 and serotonin 5-HT1A receptor agonist showing a less than convincing efficacy profile, but which may offer safety advantages over available agents by means of a reduced risk of metabolic complications. Iloperidone is a D2 and 5-HT2A receptor antagonist requiring further studies to establish its effectiveness. It has a high affinity for α1-adrenoceptors, which can lead to associated haemodynamic adverse effects. Nemonapride is essentially a typical antipsychotic drug, similar in structure to sulpiride, which has been available for some time in Japan. It has efficacy against positive symptoms and has shown some antidepressant and anxiolytic properties, although efficacy data for it are somewhat limited. Norclozapine (N-desmethylclozapine) is a major metabolite of clozapine formed by its demethylation. Its partial agonist activity at D2 receptors has raised interest in it as an antipsychotic in its own right. In addition, it appears to have muscarinic agonist activity, which is believed to be responsible for the observed positive effects it has on cognition. It was envisaged to be effective as an adjunct to other agents or at high doses in the treatment of refractory schizophrenia, although a recent randomized, controlled study showed that it was no more effective than placebo in patients with schizophrenia experiencing an acute psychotic episode. Olanzapine pamoate depot injection has shown comparable efficacy to oral olanzapine in several studies. However, it has provoked considerable safety concerns by its association with inadvertent intravascular injection events in numerous patients. This accidental intravascular administration of olanzapine pamoate leads to excessive sedation, confusion, dizziness and altered speech. Post-injection observation periods and postmarketing surveillance are planned following the introduction of the depot. Paliperidone palmitate is the palmitate ester of paliperidone, the major metabolite of risperidone, and is formulated as a long-acting injection for intramuscular use. Its pharmacology is comparable to risperidone, having D2 and 5-HT2A receptor antagonist activity. Efficacy studies have shown positive results, and because paliperidone has no antagonistic activity at cholinergic receptors, it has low potential for anticholinergic adverse effects, including cognitive dysfunction. However, with higher doses, the frequency of extrapyramidal side effects and orthostatic hypotension have been shown to be greater than with placebo.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Table I
Fig. 1
Table II
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Table III
Fig. 8
Fig. 9
Table IV
Table V

Similar content being viewed by others

References

  1. Knapp M, Mangalore R, Simon J. The global costs of schizophrenia. Schizophr Bull 2004; 30(2): 279–93

    Article  PubMed  Google Scholar 

  2. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 4th ed. Washington, DC: American Psychiatric Association, 1994

    Google Scholar 

  3. Gervin M, Barnes TRE. Assessment of drug-related movement disorders in schizophrenia. Adv Psychiatr Treat 2000; 6: 332–41

    Article  Google Scholar 

  4. Haddad PM, Wieck A. Antipsychotic-induced hyperprolactinaemia: mechanisms, clinical features and management. Drugs 2004; 64(20): 2291–314

    Article  PubMed  CAS  Google Scholar 

  5. Cavallaro R, Smeraldi E. Antipsychotic-induced tardive dyskinesia: recognition, prevention and management. CNS Drugs 1995; 4(4): 278–93

    Article  Google Scholar 

  6. Nasrallah HA. Atypical antipsychotic-induced metabolic side effects: insights from receptor-binding profiles. Mol Psychiatry 2008; 13(1): 27–35

    Article  PubMed  CAS  Google Scholar 

  7. Taylor DM, McAskill R. Atypical antipsychotics and weight gain: a systematic review. Acta Psychiatr Scand 2000; 101(6): 416–32

    Article  PubMed  CAS  Google Scholar 

  8. Nasrallah H. A review of the effect of atypical antipsychotics on weight. Psychoneuroendocrinology 2003; 28 Suppl. 1: 83–96

    Article  Google Scholar 

  9. Haddad PM. Antipsychotics and diabetes: review of non-prospective data. Br J Psychiatry Suppl 2004; 47: S80–6

    Article  PubMed  Google Scholar 

  10. Kane J, Honigfeld G, Singer J, et al. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlor-promazine. Arch Gen Psychiatry 1988; 45: 789–96

    Article  PubMed  CAS  Google Scholar 

  11. Meltzer HY, Alphs L, Green AI, et al. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry 2003; 60: 82–91

    Article  PubMed  CAS  Google Scholar 

  12. Meltzer HY, Bobo WV, Roy A, et al. A randomized, double-blind comparison of clozapine and high-dose olanzapine in treatment-resistant patients with schizophrenia. J Clin Psychiatry 2008; 69(2): 274–85

    Article  PubMed  CAS  Google Scholar 

  13. Gray JA, Roth BL. The pipeline and future of drug development in schizophrenia. Mol Psychiatry 2007; 12(10): 904–22

    Article  PubMed  CAS  Google Scholar 

  14. Carlsson A, Lindqvist M. Effect of chlorpromazine or haloperidol on formation of 3-methoxytramine and normetanephrine in mouse brain. Acta Pharmacol Toxicol (Copenh) 1963; 20: 140–4

    Article  CAS  Google Scholar 

  15. Seeman P, Lee T, Chau-Wong M, et al. Antipsychotic drug doses and neuroleptic/dopamine receptors. Nature 1976; 261(5562): 717–9

    Article  PubMed  CAS  Google Scholar 

  16. Laruelle M, bi-Dargham A, Gil R, et al. Increased dopamine transmission in schizophrenia: relationship to illness phases. Biol Psychiatry 1999; 46(1): 56–72

    Article  PubMed  CAS  Google Scholar 

  17. Kapur S, Remington G. Serotonin-dopamine interaction and its relevance to schizophrenia. Am J Psychiatry 1996; 153(4): 466–76

    PubMed  CAS  Google Scholar 

  18. Castner SA, Williams GV, Goldman-Rakic PS. Reversal of antipsychotic-induced working memory deficits by short-term dopamine D1 receptor stimulation. Science 2000; 287(5460): 2020–2

    Article  PubMed  CAS  Google Scholar 

  19. Lieberman JA. Dopamine partial agonists: a new class of anti-psychotic. CNS Drugs 2004; 18(4): 251–67

    Article  PubMed  CAS  Google Scholar 

  20. Dickson RA, Glazer WM. Neuroleptic-induced hyperprolactinemia. Schizophr Res 1999; 35 Suppl.: S75–86

    Article  PubMed  Google Scholar 

  21. Scatton B, Sanger DJ. Pharmacological and molecular targets in the search for novel antipsychotics. Behav Pharmacol 2000; 11(3–4): 243–56

    Article  PubMed  CAS  Google Scholar 

  22. Ashby Jr CR, Wang RY. Pharmacological actions of the atypical antipsychotic drug clozapine: a review. Synapse 1996; 24(4): 349–94

    Article  PubMed  CAS  Google Scholar 

  23. Di Matteo V, Cacchio M, Di Giulio C, et al. Biochemical evidence that the atypical antipsychotic drugs clozapine and risperidone block 5-HT2C receptors in vivo. Pharmacol Biochem Behav 2002; 71(4): 607–13

    Article  PubMed  Google Scholar 

  24. Alvir JMJ, Lieberman JA, Safferman AZ, et al. Clozapine-induced agranulocytosis: incidence and risk factors in the United States. N Engl J Med 1993; 3(329): 162–7

    Article  Google Scholar 

  25. Meltzer HY. Clinical studies on the mechanism of action of clozapine: the dopamine-serotonin hypothesis of schizophrenia. Psychopharmacology (Berl) 1989; 99 Suppl.: S18–27

    Article  Google Scholar 

  26. Meltzer HY. The role of serotonin in antipsychotic drug action. Neuropsychopharmacology 1999; 21(2): 106–15S

    Article  Google Scholar 

  27. Bressan RA, Erlandsson K, Jones HM, et al. Is regionally selective D2/D3 dopamine occupancy sufficient for atypical antipsychotic effect? An in vivo quantitative [123I]epidepride SPET study of amisulpride-treated patients. Am J Psychiatry 2003; 160(8): 1413–20

    Article  PubMed  Google Scholar 

  28. Peuskens J, Bech P, Moller HJ, et al. Amisulpride vs risperidone in the treatment of acute exacerbations of schizophrenia. Amisulpride Study Group. Psychiatry Res 1999; 88(2): 107–17

    CAS  Google Scholar 

  29. Seeman P. Atypical antipsychotics: mechanism of action. Can J Psychiatry 2002; 47(1): 27–38

    PubMed  Google Scholar 

  30. Kapur S, Seeman P. Does fast dissociation from the dopamine D2 receptor explain the action of atypical antipsychotics? A new hypothesis. Am J Psychiatry 2001; 158(3): 360–9

    Article  PubMed  CAS  Google Scholar 

  31. 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(2): 161–6

    PubMed  CAS  Google Scholar 

  32. Pilowsky LS, Ell PJ. Clozapine and dopamine D2 blockade. Am J Psychiatry 2002; 159(2): 324–5

    Article  PubMed  Google Scholar 

  33. Pilowsky LS, Costa DC, Ell PJ, et al. Clozapine, single photon emission tomography, and the D2 dopamine receptor blockade hypothesis of schizophrenia. Lancet 1992; 340(8813): 199–202

    Article  PubMed  CAS  Google Scholar 

  34. Tauscher-Wisniewski S, Kapur S, Tauscher J, et al. Quetiapine: an effective antipsychotic in first-episode schizophrenia despite only transiently high dopamine-2 receptor blockade. J Clin Psychiatry 2002; 63(11): 992–7

    Article  PubMed  CAS  Google Scholar 

  35. Pilowsky LS, Busatto GF, Taylor M, et al. Dopamine D2 receptor occupancy in vivo by the novel atypical antipsychotic olanzapine: a 123I IBZM single photon emission tomography (SPET) study. Psychopharmacology 1996; 124(1–2): 148–53

    Article  PubMed  CAS  Google Scholar 

  36. Pilowsky LS, Mulligan RS, Acton PD, et al. Limbic selectivity of clozapine. Lancet 1997; 350(9076): 490–1

    Article  PubMed  CAS  Google Scholar 

  37. Burris KD, Molski TF, Xu C, et al. Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther 2002; 302(1): 381–9

    Article  PubMed  CAS  Google Scholar 

  38. Jordan S, Koprivica V, Chen R, et al. The antipsychotic aripiprazole is a potent, partial agonist at the human 5-HT1A receptor. Eur J Pharmacol 2002; 441(3): 137–40

    Article  PubMed  CAS  Google Scholar 

  39. Shahid M, Walker G, Zorn S, et al. Asenapine: a novel psychopharmacologic agent with a unique human receptor binding signature. J Psychopharmacol. Epub 2008 Feb 28

  40. Bymaster FP, Calligaro DO, Falcone JF, et al. Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology 1996; 14(2): 87–96

    Article  PubMed  CAS  Google Scholar 

  41. Daniel DG, Zimbroff DL, Potkin SG, et al. Ziprasidone 80 mg/day and 160 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 6-week placebo-controlled trial. Ziprasidone Study Group. Neuropsychopharmacology 1999; 20(5): 491–505

    CAS  Google Scholar 

  42. Kongsamut S, Roehr J, Cai J, et al. Iloperidone binding to human and rat dopamine and 5-HT receptors. Eur J Pharmacol 1996; 317: 417–23

    Article  PubMed  CAS  Google Scholar 

  43. Arnt J, Skarsfeldt T. Do novel antipsychotics have similar pharmacological characteristics? A review of the evidence. Neuropsychopharmacology 1998; 18(2): 63–101

    Article  PubMed  CAS  Google Scholar 

  44. US Food and Drag Administration. Full prescribing information sheet: aripiprazole (marketed as Abilify) [online]. Available from URL: http://www.fda.gov/[Accessed 2008 Sep 6]

  45. Lieberman JA. Dopamine partial agonists: a new class of antipsychotic. CNS Drugs 2004; 18(4): 251–67

    Article  PubMed  CAS  Google Scholar 

  46. Marquis KL, Hertel P, Reindeers JH, et al. Bifeprunox: a novel atypical antipsychotic sharing dopamine D2 receptor partial agonism and serotonin 5-HT1A receptor agonism [abstract]. Schizophr Bull 2005; 31: 305

    Google Scholar 

  47. Kalkman HO, Subramanian N, Hoyer D. Extended radioligand binding profile of iloperidone: a broad spectrum dopamine/serotonin/norepinephrine receptor antagonist for the management of psychotic disorders. Neuropsychopharmacology 2001; 25(6): 904–14

    Article  PubMed  CAS  Google Scholar 

  48. Schotte A, Bonaventure P, Janssen PF, et al. In vitro receptor binding and in vivo receptor occupancy in rat and guinea pig brain: risperidone compared with antipsychotics hitherto used. Jpn J Pharmacol 1995; 69(4): 399–412

    Article  PubMed  CAS  Google Scholar 

  49. Schotte A, Janssen PFM, Gommeren W. Risperidone compared with new reference antipsychotic drugs: in vitro and in vivo receptor binding. Psychopharmacology 1996; 124: 57–73

    Article  PubMed  CAS  Google Scholar 

  50. Kyoto N, Matsumoto M, Hidaka K, et al. Dopamine D4-like binding sites labelled by [H] nemonapride include substantial serotonin 5-HT2A receptors in primate cerebral cortex. Biochem Biophys Res Commun 1999; 255: 367–70

    Article  Google Scholar 

  51. Chou JC-Y. Targeting novel therapies in schizophrenia: considerations for physicians and patients. Clinical applications of antipsychotic pharmacodynamics and pharmacokinetics. Advances in Psychiatric Medicine 2008 May (Supplement to Psychiatric Times): 1–4

  52. Svensson TH. Alpha-adrenoceptor modulation hypothesis of antipsychotic atypicality. Prog Neuropsychopharmacol Biol Psychiatry 2003; 27(7): 1145–58

    Article  PubMed  CAS  Google Scholar 

  53. Wadenberg ML, Soliman A, VanderSpek SC, et al. Dopamine D(2) receptor occupancy is a common mechanism underlying animal models of antipsychotics and their clinical effects. Neuropsychopharmacology 2001; 25(5): 633–41

    Article  PubMed  CAS  Google Scholar 

  54. Franberg O, Wiker C, Marcus MM, et al. Asenapine, a novel psychopharmacologic agent: preclinical evidence for clinical effects in schizophrenia. Psychopharmacology (Berl) 2008; 196(3): 417–29

    Article  CAS  Google Scholar 

  55. Wadenberg ML, Kapur S, Soliman A, et al. Dopamine D2 receptor occupancy predicts catalepsy and the suppression of conditioned avoidance response behavior in rats. Psychopharmacology (Berl) 2000; 150(4): 422–9

    Article  CAS  Google Scholar 

  56. Potkin SG, Cohen M, Panagides J. Efficacy and tolerability of asenapine in acute schizophrenia: a placebo- and risperidone-controlled trial. J Clin Psychiatry 2007; 68(10): 1492–500

    Article  PubMed  CAS  Google Scholar 

  57. Kay SR, Fiszbein A, Opler LA. The positive and negative syndrome scale (PANSS) for schizophrenia. Schizophr Bull 1987; 13(2): 261–76

    Article  PubMed  CAS  Google Scholar 

  58. Chouinard G, Jones B, Remington G, et al. A Canadian multi-center placebo-controlled study of fixed doses of risperidone and haloperidol in the treatment of chronic schizophrenic patients. J Clin Psychopharmacol 1993; 13(1): 25–40

    Article  PubMed  CAS  Google Scholar 

  59. Marder SR, Meibach RC. Risperidone in the treatment of schizophrenia. Am J Psychiatry 1994; 151(6): 825–35

    PubMed  CAS  Google Scholar 

  60. Allison D, Mentore J, Heo M, et al. Antipsychotic-induced weight gain: a comprehensive research synthesis. Am J Psychiatry 1999; 156: 1686–96

    PubMed  CAS  Google Scholar 

  61. McIntyre RS, Trakas K, Lin D, et al. Risk of weight gain associated with antipsychotic treatment: results from the Canadian National Outcomes Measurement Study in Schizophrenia. Can J Psychiatry 2003; 48(10): 689–94

    PubMed  Google Scholar 

  62. McCreary A. Binding characteristics of bifeprunox to dopamine and serotonin receptors [abstract]. Eur Psychiatry 2005; 20 Suppl. 1: S67

    Google Scholar 

  63. De Vries TW, Grahnen A. Bifeprunox: dopamine D2 receptor occupancy [poster]. 61st Annual Meeting of the Society of Biological Psychiatry (SOBP); 2006 May 18–20; Toronto (ON)

  64. Hertel P, Brennum L, Helboe L, et al. Bifeprunox: relationship between antipsychotic potential, EPS liability and dopamine D2 receptor occupancy in rats [abstract]. Neuropsychopharmacology 2005; 31 Suppl. 1:S256

    Google Scholar 

  65. Casey DE, Sands EE, Heisterberg J, et al. Efficacy and safety of bifeprunox in patients with acute exacerbations of schizophrenia: results of a randomized, double blind, placebo-controlled, multicentre, dose-finding study. Psychopharmacology. Epub 2008 Jul 4

  66. Rapaport M, Barbato LM, Heisterberg J, et al. Efficacy and safety of bifeprunox versus placebo in the treatment of patients with acute exacerbations of schizophrenia [abstract]. Neuropsychopharmacology 2006; 31 (Suppl. 1): S184

    Google Scholar 

  67. Barbato LM, Potkin SG, Heisterberg J, et al. A randomized, double-blind, placebo-controlled study of bifeprunox, a partial dopamine D2 receptor agonist, in patients with acute exacerbations of schizophrenia [abstract]. Neuropsychopharmacology 2006; 31 Suppl. 1: S251–2

    Google Scholar 

  68. Bourin M, Debelle M, Heisterberg J, et al. Long-term efficacy and safety of bifeprunox in patients with schizophrenia [abstract]. Neuropsychopharmacology 2006; 31 (Suppl. 1): S187–8

    Google Scholar 

  69. Haro JM, Kamath SA, Ochoa S, et al. tThe Clinical Global Impression-Schizophrenia scale: a simple instrument to measure the diversity of symptoms present in schizophrenia. Acta Psychiatr Scand 2003; (416 Suppl.): 16–23

  70. Barnes TRE. The Barnes akathisia scale: revisited. J Psychopharmacol 2003; 17: 365–70

    Article  PubMed  Google Scholar 

  71. Simpson GM, Angus JW. A rating scale for extrapyramidal side effects. Acta Psychiatr Scand 1970; 212: 11–9

    Article  CAS  Google Scholar 

  72. National Institute of Mental Health. Abnormal Involuntary Movement Scale (AIMS): 1974. US Public Health Service publication no. MH-9-17. Washington, DC: US Government Printing Office, 1974

    Google Scholar 

  73. Shapira NA, Newcomer JW. The metabolic profile of bifeprunox in the treatment of patients with schizophrenia [abstract]. Neuropsychopharmacology 2006; 31 (Suppl. 1): S175–6

    Google Scholar 

  74. Bjorklund M, Sirvio J, Puolivali J, et al. Alpha2C-adrenoceptor-overexpressing mice are impaired in executing nonspatial and spatial escape strategies. Mol Pharmacol 1998; 54(3): 569–76

    PubMed  CAS  Google Scholar 

  75. Bjorklund M, Sirvio J, Sallinen J, et al. Alpha2C-adrenoceptor overexpression disrupts execution of spatial and non-spatial search patterns. Neuroscience 1999; 88(4): 1187–98

    Article  PubMed  CAS  Google Scholar 

  76. Jackson HC, Dickinson SL, Nutt DJ. Exploring the pharmacology of the pro-convulsant effects of alpha 2-adrenoceptor antagonists in mice. Psychopharmacology (Berl) 1991; 105(4): 558–62

    Article  CAS  Google Scholar 

  77. Janumpalli S, Butler LS, MacMillan LB, et al. A point mutation (D79N) of the alpha2A adrenergic receptor abolishes the antiepileptogenic action of endogenous norepinephrine. J Neurosci 1998; 18(6): 2004–8

    PubMed  CAS  Google Scholar 

  78. Corbett R, Griffiths L, Shipley JE, et al. Iloperidone: preclinical profile and early clinical evaluation. CNS Drag Rev 1997; 3(2): 120–47

    Article  CAS  Google Scholar 

  79. Weiden P, Cutler A, Polymeropoulos M, et al. Safety profile of iloperidone. J Clin Psychopharmacol 2008; 28: S12–19

    Article  PubMed  CAS  Google Scholar 

  80. Yamamoto M, Usuda S, Tachikawa S, et al. Pharmacological studies on a new benzamide derivative, YM-09151-2, with potential neuroleptic properties. Neuropharmacology 1982; 21(10): 945–51

    Article  PubMed  CAS  Google Scholar 

  81. Terai M, Hidaka K, Nakamura Y. Comparison of [3H]YM-09151-2 with [3H]spiperone and [3H]raclopride for dopamine d-2 receptor binding to rat striatum. Eur J Pharmacol 1989; 173(2-3): 177–82

    Article  PubMed  CAS  Google Scholar 

  82. Seeman P, Guan HC, Van Tol HH. Dopamine D4 receptors elevated in schizophrenia. Nature 1993; 365(6445): 441–5

    Article  PubMed  CAS  Google Scholar 

  83. Assie MB, Cosi C, Koek W. 5-HT1A receptor agonist properties of the antipsychotic, nemonapride: comparison with bromerguride and clozapine. Eur J Pharmacol 1997; 334(2–3): 141–7

    Article  PubMed  CAS  Google Scholar 

  84. Ishiwata K, Onoguchi K, Toyama H, et al. Effects of reserpine treatment on the dopamine receptor binding of [3H/11C]nemonapride in the mouse and rat brain. Ann Nucl Med 1997; 11(1): 21–6

    Article  PubMed  CAS  Google Scholar 

  85. Usuda S, Nishikori K, Noshiro O, et al. Neuroleptic properties of cis-N-(l-benzyl-2-methylpyrrolidin-3-yl)-5-chloro-2-methoxy-4-methylaminobenzamid e (YM-09151-2) with selective antidopaminergic activity. Psychopharmacology (Berl) 1981; 73(2): 103–9

    Article  CAS  Google Scholar 

  86. Mori O, Kazamatsuri H, Kaneno S, et al. A double-blind comparison of a new benzamide compound of YM-09151 with haloperidol in the treatment of schizophrenia (Japanese). Clin Eval 1989; 17: 349–77

    Google Scholar 

  87. Kondo T, Otani K, Tokinaga N, et al. Characteristics and risk factors of acute dystonia in schizophrenic patients treated with nemonapride, a selective dopamine antagonist. J Clin Psychopharmacol 1999; 19(1): 45–50

    Article  PubMed  CAS  Google Scholar 

  88. Aguilar EJ, Keshavan MS, Martinez-Quiles MD, et al. Predictors of acute dystonia in first-episode psychotic patients. Am J Psychiatry 1994; 151(12): 1819–21

    PubMed  CAS  Google Scholar 

  89. Singh H, Levinson DF, Simpson GM, et al. Acute dystonia during fixed-dose neuroleptic treatment. J Clin Psychopharmacol 1990; 10(6): 389–96

    Article  PubMed  CAS  Google Scholar 

  90. Ishiwata K, Senda M. In vivo binding of 11C]nemonapride to sigma receptors in the cortex and cerebellum. Nucl Med Biol 1999; 26(6): 627–31

    Article  PubMed  CAS  Google Scholar 

  91. Ujike H, Akiyama K, Kuroda S. [3H]YM-09151-2 (nemonapride), a potent radioligand for both sigma 1 and sigma 2 receptor subtypes. Neuroreport 1996; 7(5): 1057–61

    Article  PubMed  CAS  Google Scholar 

  92. Walker JM, Matsumoto RR, Bowen WD, et al. Evidence for a role of haloperidol-sensitive sigma-‘opiate’ receptors in the motor effects of antipsychotic drugs. Neurology 1988; 38(6): 961–5

    Article  PubMed  CAS  Google Scholar 

  93. Kondo T, Mihara K, Yasui N, et al. Therapeutic spectrum of nemonapride and its relationship with plasma concentrations of the drug and prolactin. J Clin Psychopharmacol 2000; 20(4): 404–9

    Article  PubMed  CAS  Google Scholar 

  94. Rhoades HM, Overall JE. The semistructured BPRS interview and rating guide. Psychopharmacol Bull 1988; 24(1): 101–4

    PubMed  CAS  Google Scholar 

  95. Van Putten T, Marder SR, Mintz J, et al. Haloperidol plasma levels and clinical response: a therapeutic window relationship. Am J Psychiatry 1992; 149(4): 500–5

    PubMed  Google Scholar 

  96. Hansen LB, Larsen NE, Gulmann N. Dose-response relationships of perphenazine in the treatment of acute psychoses. Psychopharmacology (Berl) 1982; 78(2): 112–5

    Article  CAS  Google Scholar 

  97. Dahl SG. Plasma level monitoring of antipsychotic drugs: clinical utility. Clin Pharmacokinet 1986; 11(1): 36–61

    Article  PubMed  CAS  Google Scholar 

  98. Kondo T, Ishida M, Tokinaga N, et al. Associations between side effects of nemonapride and plasma concentrations of the drug and prolactin. Prog Neuropsychopharmacol Biol Psychiatry 2002; 26(2): 287–91

    Article  PubMed  CAS  Google Scholar 

  99. Pirmohamed M, Williams D, Madden S, et al. Metabolism and bioactivation of clozapine by human liver in vitro. J Pharmacol Exp Ther 1995; 272(3): 984–90

    PubMed  CAS  Google Scholar 

  100. Olesen OV, Linnet K. Contributions of five human cytochrome P450 isoforms to the N-demethylation of clozapine in vitro at low and high concentrations. J Clin Pharmacol 2001; 41(8): 823–32

    Article  PubMed  CAS  Google Scholar 

  101. Kuoppamaki M, Syvalahti E, Hietala J. Clozapine and N-desmethylclozapine are potent 5-HT1C receptor antagonists. Eur J Pharmacol 1993; 245(2): 179–82

    Article  PubMed  CAS  Google Scholar 

  102. Sur C, Mallorga PJ, Wittmann M, et al. N-Desmethylclozapine, an allosteric agonist at muscarinic 1 receptor, potentiates N-methyl-D-aspartate receptor activity. Proc Natl Acad Sci U S A 2003; 100(23): 13674–9

    Article  PubMed  CAS  Google Scholar 

  103. Burstein ES, Ma J, Wong S, et al. Intrinsic efficacy of antipsychotics at human D2, D3, and D4 dopamine receptors: identification of the clozapine metabolite N-desmethylclozapine as a D2/D3 partial agonist. J Pharmacol Exp Ther 2005; 315(3): 1278–87

    Article  PubMed  CAS  Google Scholar 

  104. Weiner DM, Meltzer HY, Veinbergs I, et al. The role of Ml muscarinic receptor agonism of N-desmethylclozapine in the unique clinical effects of clozapine. Psychopharmacology (Berl) 2004; 177(1–2): 207–16

    Article  CAS  Google Scholar 

  105. Potkin SG, Saha AR, Kujawa MJ, et al. Aripiprazole, an antipsychotic with a novel mechanism of action, and risperidone vs placebo in patients with schizophrenia and schizoaffective disorder. Arch Gen Psychiatry 2003; 60(7): 681–90

    Article  PubMed  CAS  Google Scholar 

  106. Dettling M, Sachse C, Brockmoller J, et al. Long-term therapeutic drug monitoring of clozapine and metabolites in psychiatric in- and outpatients. Psychopharmacology (Berl) 2000; 152(1): 80–6

    Article  CAS  Google Scholar 

  107. Mauri M, Volonteri LS, Fiorentini A, et al. Clinical outcome and plasma levels of clozapine and norclozapine in drug-resistant schizophrenic patients. Schizophr Res 2004; 66(2–3): 197–8

    Article  PubMed  Google Scholar 

  108. Natesan S, Reckless GE, Barlow KB, et al. Evaluation of N-desmethylclozapine as a potential antipsychotic: preclinical studies. Neuropsychopharmacology 2007; 32(7): 1540–9

    Article  PubMed  CAS  Google Scholar 

  109. ACADIA Pharmaceuticals. ACADIA Pharmaceuticals announces results from ACP-104 IIb schizophrenia trial [online]. Available from URL: http://news.acadia-pharm.com[Accessed 2008 Sep 7]

  110. Beasley CM. Efficacy of olanzapine: an overview of pivotal clinical trials. J Clin Psychiatr Monogr 1997; 15(2): 16–8

    Google Scholar 

  111. Lieberman JA, McEvoy JP, Swartz MS, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 2005; 353: 1209–23

    Article  PubMed  CAS  Google Scholar 

  112. Beasley Jr CM, Stauffer VL, Liu-Seifert H, et al. All-cause treatment discontinuation in schizophrenia during treatment with olanzapine relative to other antipsychotics: an integrated analysis. J Clin Psychopharmacol 2007; 27(3): 252–8

    Article  PubMed  CAS  Google Scholar 

  113. Eli Lilly and Company Limited. Psychopharmacologic drugs advisory committee briefing document: zyprexa olanzapine pamoate (op) depot; olanzapine long-acting injection; schizophrenia [online]. Available from URL: http://www.fda.gov/[Accessed 2008 Sep 5]

  114. Kapur S, Zipursky RB, Remington G, et al. 5-HT2 and D2 receptor occupancy of olanzapine in schizophrenia: a PET investigation. Am J Psychiatry 1998; 155(7): 921–8

    PubMed  CAS  Google Scholar 

  115. Mamo D, Kapur S, Keshavan M, et al. D(2) receptor occupancy of olanzapine pamoate depot using positron emission tomography: an open-label study in patients with schizophrenia. Neuropsychopharmacology 2008; 33: 298–304

    Article  PubMed  CAS  Google Scholar 

  116. Beasley Jr CM, Sanger T, Satterlee W, et al. Olanzapine versus placebo: results of a double-blind, fixed-dose olanzapine trial. Psychopharmacology 1996; 124: 159–67

    Article  PubMed  CAS  Google Scholar 

  117. Beasley Jr CM, Tollefson G, Tran P, et al. Olanzapine versus placebo and haloperidol: acute phase results of the North American double-blind olanzapine trial. Neuropsychopharmacology 1996; 14(2): 111–23

    Article  PubMed  CAS  Google Scholar 

  118. Beasley Jr CM, Sutton VK, Hamilton SH, et al. A double-blind, randomized, placebo-controlled trial of olanzapine in the prevention of psychotic relapse. J Clin Psychopharmacol 2003; 23(6): 582–94

    Article  PubMed  CAS  Google Scholar 

  119. Taylor DM, Young C, Patel MX. Prospective 6-month follow-up of patients prescribed risperidone long-acting injection: factors predicting favourable outcome. Int J Neuropsycho-pharmacol 2006; 9(6): 685–94

    Article  CAS  Google Scholar 

  120. Young CL, Taylor DM. Health resource utilization associated with switching to risperidone long-acting injection. Acta Psychiatr Scand 2006; 114: 14–20

    Article  PubMed  CAS  Google Scholar 

  121. van Beijsterveldt LE, Geerts RJ, Leysen JE, et al. Regional brain distribution of risperidone and its active metabolite 9-hydroxy-risperidone in the rat. Psychopharmacology (Berl) 1994; 114(1): 53–62

    Article  Google Scholar 

  122. Leysen JE, Janssen PM, Megens AA, et al. Risperidone: a novel antipsychotic with balanced serotonin-dopamine antagonism, receptor occupancy profile, and pharmacologic activity. J Clin Psychiatry 1994; 55 Suppl.: 5–12

    PubMed  Google Scholar 

  123. Citrome L. Paliperidone: quo vadis? Int J Clin Pract 2007; 61(4): 653–62

    Article  PubMed  CAS  Google Scholar 

  124. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of immediate-release paliperidone [poster]. 46th Annual Meeting of the NCEDU; 2006 Jun 12–15; Boca Raton (FL), 186

  125. Aravagiri M, Marder SR, Nuechterlein KH, et al. Intra- and interindividual variations in steady-state plasma concentrations of risperidone and 9-hydroxyrisperidone in schizophrenic patients treated chronically with various doses of risperidone. Ther Drug Monit 2003; 25(6): 657–64

    Article  PubMed  CAS  Google Scholar 

  126. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000; 22(4): 481–5

    Article  PubMed  CAS  Google Scholar 

  127. Kramer M, Lim P, Eerdekens M, et al. Efficacy/tolerability of paliperidone palmitate: 9-week, placebo-controlled study in schizophrenia patients [abstract]. Schizophr Res 2008; 98 (Suppl. 1): 165–6

    Article  Google Scholar 

  128. Rosenheck R, Leslie D, Keefe R, et al. Barriers to employment for people with schizophrenia. Am J Psychiatry 2006; 163(3): 411–7

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Dr James C-Y Chou for his contribution to the article. No sources of funding were used to assist in the preparation of this manuscript. Miss Bishara has no conflicts of interest that are directly relevant to the content of this review. Dr Taylor has acted as a consultant for Servier and Bristol-Myers Squibb, and has received honoraria or grants from Bristol-Myers Squibb, Novartis, AstraZeneca, Janssen-Cilag, Wyeth and Sanofi-Aventis.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David Taylor.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bishara, D., Taylor, D. Upcoming Agents for the Treatment of Schizophrenia. Drugs 68, 2269–2292 (2008). https://doi.org/10.2165/0003495-200868160-00002

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/0003495-200868160-00002

Keywords

Navigation