Hostname: page-component-7c8c6479df-r7xzm Total loading time: 0 Render date: 2024-03-28T03:19:27.879Z Has data issue: false hasContentIssue false

Drug information update. Unconventional treatment strategies for schizophrenia: Polypharmacy and heroic dosing

Published online by Cambridge University Press:  02 January 2018

Bret A. Moore
Affiliation:
U.S. Army Regional Health Command-Central, San Antonio, Texas, USA
Debbi A. Morrissette
Affiliation:
Neuroscience Education Institute, Carlsbad, California, USA
Jonathan M. Meyer
Affiliation:
California Department of State Hospitals, Sacramento, California, USA
Stephen M. Stahl*
Affiliation:
Neuroscience Education Institute, Carlsbad, California, USA Department of Psychiatry, University of Cambridge, Cambridge, UK
*
Correspondence to Stephen M. Stahl (smstahl@neiglobal.com)
Rights & Permissions [Opens in a new window]

Summary

The majority of patients respond to antipsychotic monotherapy at standard doses, but a subset of patients will require more heroic measures that include antipsychotic polypharmacy and high-dose monotherapy. Indeed, research has shown that roughly 30% of patients with psychosis are prescribed multiple antipsychotic medications. We discuss the potential benefits and challenges of these approaches and provide a rationale for why and when they should be utilised.

Type
Special Articles
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an open-access article published by the Royal College of Psychiatrists and distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © 2017 The Author

Schizophrenia is one of the most challenging psychiatric disorders faced by clinicians today, affecting roughly 1% of the population. Reference Perälä, Suvisaari, Saarni, Kuoppasalmi, Isometsä and Pirkola1 Many out-patient clinics and virtually all in-patient facilities manage these often complex clinical cases. Since the introduction of the first-generation antipsychotics (FGA) and subsequent development of the dopamine hypothesis, blockade of dopamine 2 (D2) receptors has been the primary goal of pharmacological intervention. Even with the development and proliferation of the second-generation antipsychotics (SGA), D2 blockade remains central to effective control of positive symptoms whereas serotonin 2A (5-HT2A) plays an important but secondary role in the modulation of dopamine, theoretically improving negative symptoms while reducing the risk of extrapyramidal symptoms. Reference Schwartz and Stahl2

The term ‘dopamine hypothesis’ of schizophrenia is slowly being phased out, as more sophisticated imaging studies have identified the nature of the dopamine dysfunction and its locus within the striatum. The striatum is a major component of the basal ganglia, and is divided functionally into the dorsal sensorimotor portion, the central associative striatum, and the ventral limbic striatum. Recently it has been found that the best correlation between positive symptoms of psychosis in schizophrenia and levels of D2 activity is in the rostral caudate, a portion of the central associative striatum. Reference Kegels, Abi-Dargham, Frankle, Gil, Cooper and Slifstein3,Reference Thompson, Urban, Slifstein, Xu, Kegeles and Girgis4 We thus have in vivo confirmation of excessive D2 neurotransmission and the relationship to the positive symptoms of schizophrenia. In the limbic striatum low dopamine release is directly related to the severity of negative symptoms; the lower the dopamine levels, the greater the negative symptom severity. Reference Kegels, Abi-Dargham, Frankle, Gil, Cooper and Slifstein3

While the goal of treatment is normalisation of D2 neurotransmission in the striatum, a subset of patients do not respond to traditional treatment algorithms which include antipsychotic monotherapy at dosing levels defined in registrational trials. Consequently, many patients continue to exhibit uncontrolled positive and negative symptoms as well as manifesting aggressive and impulsive behaviours. Reference Stahl5 It has been argued that ‘unconventional’ strategies for better management of treatment-resistant psychosis should be employed. Reference Morrissette and Stahl6Reference Stahl8 The two primary methods we discuss in the following pages include antipsychotic polypharmacy and high-dose monotherapy.

Why do standard treatments sometimes fail?

To effectively treat the positive symptoms of schizophrenia it is important to achieve at least 60% striatal D2 blockade with antipsychotics. Reference Uchida, Takeuchi, Graff-Guerrero, Suzuki, Watanabe and Mamo9 The exception is clozapine, which has been shown to have some antipsychotic effect at as little as 20% blockade. Reference Uchida, Takeuchi, Graff-Guerrero, Suzuki, Watanabe and Mamo10 Beyond 80% D2 occupancy, risk of extrapyramidal side-effects (EPS) increases, leading to non-adherence and treatment failure. Therefore, the ‘sweet spot’ of D2 receptor blockade for most patients is between 60 and 80%. Reference Morrissette and Stahl6 However, in some treatment-resistant cases, particularly when aggression and impulsivity are of concern, moving beyond 80% may be necessary. Reference Morrissette and Stahl6,Reference Stahl8,Reference Stahl, Morrissette and Ritsner11Reference Remington, Mamo, Labelle, Reiss, Shammi and Mannaert14 Moreover, there is significant evidence from the literature that a substantial portion of patients tolerate plasma antipsychotic levels consistent with >80% D2 occupancy.

One of the early imaging studies from 1997 noted that a large fraction of patients stabilised on low-dose oral haloperidol had plasma levels associated with >80% D2 occupancy; Reference Kapur, Zipursky, Roy, Jones, Remington and Reed12 however, prior clinical studies found that even at a plasma haloperidol level of 6 ng/ml (predicted 90% D2 occupancy), only 30% experienced adverse effects. Reference Midha, Hubbard, Marder, Marshall and Van Putten13 A similar pattern is seen with risperidone. The pivotal trials studied doses of 10 mg and 16 mg, which correlate with a plasma active moiety level of 70 ng/ml and 112 ng/ml, respectively. Reference de Leon, Sandson and Cozza15 The plasma level of 70 ng/ml would correspond to a D2 occupancy of 85%; Reference Remington, Mamo, Labelle, Reiss, Shammi and Mannaert14 however, despite this high level of predicted D2 occupancy, the proportion of patients on 10 mg who required anti-Parkinsonian treatment was only 31%. Reference Marder and Meibach16

Pharmacokinetic failure

Pharmacokinetics is the umbrella term that covers drug absorption, bioavailability, distribution, metabolism and excretion. The role of pharmacokinetics in antipsychotic treatment failure is relatively simple. Through abnormal pharmacokinetic processes such as poor absorption, rapid metabolism and enzymatic polymorphisms, antipsychotic plasma levels do not reach the threshold associated with 60% striatal D2 occupancy. This leads to continued psychotic symptoms. It also leads to frustration and confusion for clinicians in that the patient does not sufficiently respond to standard treatment algorithms.

Pharmacodynamic failure

Pharmacodynamics encompasses receptor binding and sensitivity, postreceptor effects, and chemical communication. Pharmacodynamic treatment failure with regard to antipsychotics is the inability to provide significant amelioration of psychotic symptoms in spite of achieving plasma levels associated with at least 60–80% D2 occupancy. Reference Correll17 Although adequate drug plasma levels are achieved, patients with treatment-resistant psychosis present with continued positive, cognitive and aggressive symptoms. The treatment failure associated with pharmacodynamic influences is hypothesised to be related to lack of D2 receptor sensitivity or hypersensitivity. When patients manifest a lack of extrapyramidal adverse effects or akathisia, increasing drug doses to achieve plasma levels that are associated with >80% D2 blockade may be necessary to provide symptom control. Reference Remington and Kapur18Reference Seeman20 The overriding principle is that there are a subset of patients who both tolerate and require high levels of D2 antagonism for symptomatic relief.

The importance of being patient

While recent studies have demonstrated that minimal response after 2 weeks on a particular antipsychotic dose portends a low likelihood of week 6 response on that dose, the full therapeutic effects of adequate D2 receptor blockade in schizophrenia may not be apparent until many weeks or months later. Reference Stahl7 Therefore, patience in pharmacological treatment of psychosis is critical when a patient exhibits partial response. Reference Stahl7,Reference Stahl, Morrissette and Ritsner11 For example, Robinson and colleagues found that in a sample of 118 first-episode patients with schizophrenia or schizoaffective disorder only 20% responded to treatment at 4 weeks. The picture was quite different at 52 weeks; roughly 87% responded to treatment. Reference Robinson, Woerner, Alvir, Geisler, Koreen and Sheitman21 Other studies of ziprasidone, risperidone and olanzapine have shown continued improvement over several months of treatment. Reference Stahl, Morrissette and Ritsner11

Strategies to use prior to heroic measures

We believe polypharmacy and high dosing should not be the initial approach to treating schizophrenia. However, considering that roughly 30% of patients with psychosis are on multiple antipsychotics, the practice is far from rare. Reference Längle, Steinert, Weiser, Schepp, Jaeger and Pfiffner22,Reference Essock, Schooler, Stroup, McEvoy, Rojas and Jackson23 In an effort to address the growing practice of antipsychotic polypharmacy and high dosing of antipsychotics in spite of little support in the literature, Stahl provides 12 case-based recommendations. Reference Stahl7 We review several below.

Utilise monotherapy first to include clozapine

Sequential trials of at least two SGAs are recommended. If both trials fail, consideration of an FGA is appropriate. Also, it is important to not overlook clozapine as monotherapy. The efficacy of clozapine in treatment-resistant schizophrenia, particularly with regard to aggression and violence, is well documented. Reference Volavka and Citrome24Reference Volavka and Citrome26 However, some clinicians may be hesitant to initiate a trial of clozapine owing to fear of side-effects such as agranulocytosis.

Monitor blood levels

Securing drug plasma levels is the only way to know whether treatment failure is due to a pharmacokinetic issue such as rapid metabolism or a cytochrome P450 polymorphism, or simply poor adherence with oral therapy. Likewise, blood levels can alert you to pharmacodynamic abnormalities which occur when treatment response does not correlate with adequate dosing. Blood level monitoring of both FGAs and SGAs can provide the clinician with important information which can guide the treatment plan for patients with treatment-resistant psychosis. This is supported by the recent work of Lopez & Kane as relevant to haloperidol, fluphenazine, perphenazine, risperidone, olanzapine and clozapine. Reference Lopez and Kane27

Time may not be on your side

As noted above, it takes some patients longer than others to respond to antipsychotic treatments. Granted, it may not be possible to wait several weeks (and certainly not months) in acute settings or when a patient's behaviour is potentially harmful to self or others, but when possible, allowing adequate time for full response may be all that is needed when a patient has exhibited a partial response. The result of impatience is that a second antipsychotic may be prescribed or a single medication may be dosed in an unnecessarily aggressive manner.

Double-check the diagnosis

It is common practice to rethink the primary diagnosis if the treatment plan appears ineffective. Once pharmacokinetic, pharmacodynamic or time-course failures have been ruled out, the presence of substance misuse or a personality disorder or neurological illness should be considered.

Antipsychotic polypharmacy

Although a number of published treatment guidelines for schizophrenia are available, some of which conflict with each other, it is clear that clinicians should utilise a monotherapy approach to antipsychotic medication use. Reference Stahl, Morrissette, Citrome, Saklad, Cummings and Meyer28 Multiple trials of antipsychotic medications, generally SGAs to include clozapine, are recommended. In fact, divergence from this sequential clinical progression has historically been met with scepticism, caution and outright criticism. Reference Stahl29Reference Stahl, Freudenreich and Goff36 It is certainly understandable why this is the case. The literature is replete with evidence supporting the efficacy of monotherapy for schizophrenia. Furthermore, the pitfalls associated with combining antipsychotics are well documented. Increased side-effects, higher medication costs, scant information supporting efficacy, and suboptimal outcomes are all problematic with regard to antipsychotic polypharmacy. Reference Stahl29Reference Stahl, Freudenreich and Goff36 So, why the need to even review the topic? The reality is that patients included in research studies are generally those who are able to give consent, exhibit less violence and less impulsivity, have lower rates of chemical dependency, and are less likely to have histories of sequential trials of antipsychotics at documented therapeutic levels. Reference Stahl7,Reference Stahl8 In other words, consistent with much of psychiatry research, they are healthier and not mirror images of the patients seen in clinical practice. Therefore, we believe a strict adherence to a treatment guideline based on highly selective samples does not necessarily translate well to community-based out-patient clinics and in-patient facilities.

We acknowledge that antipsychotic monotherapy is sufficient for the majority of patients with schizophrenia and that adherence to established guidelines should generally occur. Indeed, recent studies support this position. A 2004 study by Suzuki and colleagues revealed that when patients with schizophrenia were switched from multiple antipsychotics to monotherapy, roughly half maintained gains whereas a quarter showed improvements. Another quarter of the sample decompensated. Reference Suzuki, Uchida, Tanaka, Nomura, Takano and Tanabe37 In a similar study by Essock and colleagues it was found that patients switched to monotherapy maintained gains, but also showed improvement in metabolic effects assumed to be caused by antipsychotic polypharmacy. It should be noted that approximately a third of patients required multiple antipsychotics. Reference Essock, Schooler, Stroup, McEvoy, Rojas and Jackson23 However, some evidence supports the use of antipsychotic polypharmacy. A recent meta-analysis of randomised controlled trials comparing antipsychotic monotherapy and polypharmacy highlighted that polypharmacy may be superior to monotherapy in certain clinical cases. Reference Correll, Rummel-Kluge, Corves, Kane and Leucht38

In addition to achieving adequate D2 occupancy, antipsychotic polypharmacy also exploits other receptor-binding properties that could lead to improvement in other schizophrenia symptom clusters. For example, serotonergic, noradrenergic and histaminergic binding theoretically ameliorate depression, anxiety, insomnia, impulsivity and aggression. On the flip side, however, the patient is potentially exposed to adverse side-effects from multiple receptor binding or excessive binding via similar properties shared by antipsychotics (e.g. excessive histaminergic binding leading to daytime sedation or appetite stimulation and weight gain). Consequently, combining antipsychotics should be done rationally based on their binding profiles. One clear example is the need to avoid combining the partial D2 agonism of aripiprazole with antipsychotics with full D2 antagonism. The binding interference may lead to a worsening of symptoms due to aripiprazole's high affinity for the D2 receptor, and the fact that even low doses such as 10 mg achieve 83% D2 occupancy, and thus may displace full antagonists. Reference Morrissette and Stahl6

High dosing of antipsychotics

Antipsychotic polypharmacy is not the only means of addressing the more complex and treatment-resistant cases of schizophrenia. High-dose monotherapy is a viable option as well. In fact, it has been argued that if the goal is to occupy a greater degree of D2 receptors in order to address treatment-resistant positive and aggressive symptoms, high-dose monotherapy is the preferred option when compared with polypharmacy. High-dose monotherapy does, however, come at a greater financial expense and the risk of increased metabolic and other potential treatment-limiting side-effects. Reference Stahl, Morrissette and Ritsner11

It is impossible to know what dose of a particular antipsychotic is required to achieve the intended outcome. Therefore, the prudent action is to start low within the US Food and Drug Administration (FDA)- and British National Formulary (BNF)-approved guidelines for the particular medication. The medication can be gradually increased outside the FDA-approved dosing window until therapeutic response occurs or the patient develops intolerable side-effects. It is important that informed consent is obtained and treatment rationale is well documented when this occurs. Below we discuss the typical dosing ranges and special considerations for high dosing of the antipsychotics. A more detailed analysis can be found in Stahl & Morrissette's review of the topic. Reference Stahl, Morrissette and Ritsner11

Clozapine

Clozapine is typically only recommended after subsequent trials of other antipsychotics have failed. This is primarily owing to its side-effect profile. At typical dosing of 300–450 mg/day, clozapine binds to less than 50% of D2 receptors, but as noted earlier, the antipsychotic benefits with this medication can be seen at as low as 20% occupancy. Reference Uchida, Takeuchi, Graff-Guerrero, Suzuki, Watanabe and Mamo10 A meta-analysis by Davis & Chen revealed that patients on high levels of clozapine responded more frequently than those on low levels. Reference Davis and Chen39 Clozapine can be dosed as high as 900 mg/day, but seizure risk does increase with higher plasma levels, so titration to this dose should be done slowly. Furthermore, due to the diverse binding profile of clozapine, improvement in multiple symptoms clusters is possible.

Quetiapine

Quetiapine has a relatively weak affinity for D2 receptors and often requires high dosing to achieve intended outcomes. Only at the upper range of 400–800 mg/day are the antipsychotic properties of the medication seen. It is generally believed that a dose of 1200 mg/day is no more effective than the typical dosing range and carries greater incidence of metabolic effects; however, clinical practice has shown that 1800 mg/day may be useful in treating violent patients. Reference Schwartz and Stahl2,Reference Stahl5,Reference Davis and Chen39

Olanzapine

Doses of olanzapine between 10 and 20 mg/day equate to 60–80% D2 occupancy. Higher doses of 40–60 mg daily appear to be more effective, particularly with aggressive patients and in some forensic settings. Reference Schwartz and Stahl2,Reference Stahl and Grady32,Reference Mauri, Volonteri, Colasanti, Fiorentini, De Gaspari and Bareggi40,Reference Barnes and Paton41 A note of caution is that as plasma levels increase the risks of anticholinergic and metabolic effects also increase. Reference Stahl5,Reference Uchida, Takeuchi, Graff-Guerrero, Suzuki, Watanabe and Mamo10

Risperidone/paliperidone

Risperidone reaches 70–80% of D2 occupancy at doses between 2 and 6 mg/day. The risk of EPS is positively correlated with dose. Doses above 8 mg/day are generally not considered beneficial for most patients, but in some, the side-effects may not appear until higher dosages. Reference Stahl5 As noted previously, even at 10 mg/day only 31% of patients required anti-Parkinsonian medication in the pivotal trials, again providing evidence that a subgroup may both require and tolerate higher dosages and plasma levels. Reference Marder and Meibach16 Risperidone's active metabolite paliperidone has less chance of drug–drug interactions as it is not metabolised by the liver. Similar to risperidone, paliperidone carries increased risk of EPS as the dose increases. Reference Stahl, Morrissette and Ritsner11

Ziprasidone

Data support the use of high doses of ziprasidone, particularly in forensic settings at 360 mg/day. Reference Schwartz and Stahl2Reference Stahl5,Reference Mauri, Volonteri, Colasanti, Fiorentini, De Gaspari and Bareggi40,Reference Barnes and Paton41 It can be difficult to achieve adequate plasma levels with ziprasidone in out-patient settings as food is required to increase absorption. It has been reported that ziprasidone has historically been under-dosed due to concern about increased agitation and QTc prolongation.

Aripiprazole

Aripiprazole has a different mechanism of action compared with the ‘first wave’ of SGAs. Contrary to its predecessors, high doses of aripiprazole may not result in increased efficacy in schizophrenia. This is due to its partial agonist properties and high affinity for D2 receptors. Reference Stahl, Morrissette and Ritsner11 Doses of 40 mg/day are associated with 96.8% D2 occupancy, so further increases will not have an impact on D2 neurotransmission to any considerable extent.

Asenapine, iloperidone, and lurasidone

Asenapine, iloperidone, and lurasidone are newer atypical antipsychotics. Consequently, there is limited information that supports their use in high doses. Although doses of asenapine of 30–40 mg/day may be effective for some treatment-resistant cases, there are virtually no data supporting use at these higher doses, and the buccal absorption of asenapine declines significantly for each 5 mg increase in the dose. As with asenapine, there are limited to no data supporting the use of iloperidone at high doses. One treatment-limiting issue with iloperidone is orthostatic hypotension. Lurasidone is approved up to 160 mg/day for schizophrenia, but higher dosages have not been studied for efficacy, only for safety (e.g. thorough QT studies up to 600 mg). Similar to ziprasidone, lurasidone should be taken with food to increase absorption. Reference Stahl, Morrissette and Ritsner11

Summary

Schizophrenia is a relatively common psychiatric disorder but it is often difficult to treat. Although antipsychotic monotherapy at standard dosing levels is sufficient for the majority of patients, a subset will require ‘unconventional’ approaches such as antipsychotic polypharmacy and higher than normal dosing. If done cautiously and rationally, these approaches can provide much-needed benefit for those most in need of relief.

Footnotes

Declaration of interest

J.M.M. reports speaking or advising fees from Acadia Pharmaceuticals, Alkermes, Forum Pharmaceuticals, Merck, Otsuka America, Inc., Sunovion Pharmaceuticals and Teva Pharmaceutical Industries. S.M.S. has served as a consultant for Acadia, BioMarin, EnVivo Pharmaceuticals, Forum Pharmaceuticals, Jazz Pharmaceuticals, Orexigen Therapeutics, Otsuka, Pamlab, Servier, Shire, Sprout, Taisho Pharmaceutical, Takeda and Trius Therapeutics; as an Advisory Board Member for BioMarin, EnVivo, Forum, Genomind, Lundbeck, Otsuka, RCT Logic and Shire; on the Speakers Bureau for Forum, Takeda, Servier and Sunovion UK; and he has received research and/or grant support from Alkermes, Clintara, Eli Lilly, Forest Laboratories, Forum, Genomind, JayMac Pharmaceuticals, Jazz, Merck, Novartis, Otsuka, Pamlab, Pfizer, Servier, Shire, Sprout, Sunovion, Sunovion UK, Takeda, Teva and Tonix.

References

1 Perälä, J, Suvisaari, J, Saarni, SI, Kuoppasalmi, K, Isometsä, E, Pirkola, S, et al. Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch Gen Psychiatry 2007; 64: 1928.Google Scholar
2 Schwartz, T, Stahl, S. Treatment strategies for dosing the second generation antipsychotics. CNS Neurosci Ther 2011; 17: 110–7.Google Scholar
3 Kegels, LS, Abi-Dargham, A, Frankle, WG, Gil, R, Cooper, TB, Slifstein, M, et al. Increased synaptic dopamine function in associative regions of the striatum in schizophrenia. Arch Gen Psych 2010; 67: 231–9.Google Scholar
4 Thompson, JL, Urban, N, Slifstein, M, Xu, X, Kegeles, LS, Girgis, RR, et al. Striatal dopamine release in schizophrenia comorbid with substance dependence. Mol Psychiatry 2013; 18: 909–15.Google Scholar
5 Stahl, SM. Stahl's Essential Psychopharmacology: Neuroscientific Basis and Practical Applications, 4th edn. Cambridge University Press, 2013.Google Scholar
6 Morrissette, DA, Stahl, SM. Treating the violent patient with psychosis or impulsivity utilizing antipsychotic polypharmacy and high-dose monotherapy. CNS Spectr 2014; 19: 439–48.Google Scholar
7 Stahl, SM. Emerging guidelines for the use of antipsychotic polypharmacy. Rev Psiquiatr Salud Ment 2013; 6: 97100.Google Scholar
8 Stahl, SM. Antipsychotic polypharmacy: never say never, but never say always. Acta Psychiatr Scand 2012; 125: 349–51.Google Scholar
9 Uchida, H, Takeuchi, H, Graff-Guerrero, A, Suzuki, T, Watanabe, K, Mamo, DC. Dopamine D2 receptor occupancy and clinical effects: a systematic review and pooled analysis. J Clin Psychopharmacol 2011; 31: 497502.Google Scholar
10 Uchida, H, Takeuchi, H, Graff-Guerrero, A, Suzuki, T, Watanabe, K, Mamo, DC. Predicting dopamine D receptor occupancy from plasma levels of antipsychotic drugs: a systematic review and pooled analysis. J Clin Psychopharmacol 2011; 31: 318–25.Google Scholar
11 Stahl, S, Morrissette, D. Should high dose or very long-term antipsychotic monotherapy be considered before antipsychotic polypharmacy? In Polypharmacy in Psychiatry Practice. Vol. I: Multiple Medication Use Strategies (ed Ritsner, MS): pp. 107125. Springer Netherlands, 2013.Google Scholar
12 Kapur, S, Zipursky, R, Roy, P, Jones, C, Remington, G, Reed, K, et al. The relationship between D2 receptor occupancy and plasma levels on low dose oral haloperidol: a PET study. Psychopharmacology 1997; 131: 148–52.Google Scholar
13 Midha, KK, Hubbard, JW, Marder, SR, Marshall, BD, Van Putten, T. Impact of clinical pharmacokinetics on neuroleptic therapy in patients with schizophrenia. J Psychiatry Neurosci 1994; 19: 254–64.Google Scholar
14 Remington, G, Mamo, D, Labelle, A, Reiss, J, Shammi, C, Mannaert, E, et al. A PET study evaluating dopamine D2 receptor occupancy for long-acting injectable risperidone. Am J Psychiatry 2006; 163: 396401.Google Scholar
15 de Leon, J, Sandson, NB, Cozza, KL. A preliminary attempt to personalize risperidone dosing using drug-drug interactions and genetics, Part II. Psychosomatics 2008; 48: 347–61.Google Scholar
16 Marder, SR, Meibach, RC. Risperidone in the treatment of schizophrenia. Am J Psychiatry 1994; 151: 825–35.Google Scholar
17 Correll, CU. From receptor pharmacology to improved outcomes: individualising the selection, dosing, and switching of antipsychotics. Eur Psychiatry 2010; 25: S1221.Google Scholar
18 Remington, G, Kapur, S. Antipsychotic dosing: how much but also how often? Schizophr Bull 2010; 36: 900–3.Google Scholar
19 Samaha, AN, Seeman, P, Stewart, J, Rajabi, H, Kapur, S. ‘Breakthrough’ dopamine supersensitivity during ongoing antipsychotic treatment leads to treatment failure over time. J Neurosci 2007; 27: 2979–86.Google Scholar
20 Seeman, P. Dopamine D2 receptors as treatment targets in schizophrenia. Clin Schizophr Relat Psychoses 2010; 4: 5673.Google Scholar
21 Robinson, DG, Woerner, MG, Alvir, JM, Geisler, S, Koreen, A, Sheitman, B, et al. Predictors of treatment response from a first episode of schizophrenia or schizoaffective disorder. Am J Psychiatry 1999; 156: 544–9.Google Scholar
22 Längle, G, Steinert, T, Weiser, P, Schepp, W, Jaeger, S, Pfiffner, C, et al. Effects of polypharmacy on outcome in patients with schizophrenia in routine psychiatric treatment. Acta Psychiatr Scand 2012; 125: 372–81.Google Scholar
23 Essock, SM, Schooler, NR, Stroup, TS, McEvoy, JP, Rojas, I, Jackson, C, et al. Effectiveness of switching from antipsychotic polypharmacy to monotherapy. Am J Psychiatry 2011; 168: 702–8.Google Scholar
24 Volavka, J, Citrome, L. Heterogeneity of violence in schizophrenia and implications for long-term treatment. Int J Clin Pract 2008; 62: 1237–45.Google Scholar
25 Volavka, J, Czobor, P, Nolan, K, Sheitman, B, Lindenmayer, JP, Citrome, L, et al. Overt aggression and psychotic symptoms in patients with schizophrenia treated with clozapine, olanzapine, risperidone, or haloperidol. J Clin Psychopharmacol 2004; 24: 225–8.Google Scholar
26 Volavka, J, Citrome, L. Pathways to aggression in schizophrenia affect results of treatment. Schizophr Bull 2011; 37: 921–9.Google Scholar
27 Lopez, L, Kane, J. Recommendations for the monitoring of serum concentrations of antipsychotic drugs in the treatment of schizophrenia. J Clin Psychiatry 2015; 76: 1249–50.Google Scholar
28 Stahl, SM, Morrissette, DA, Citrome, L, Saklad, SR, Cummings, MA, Meyer, JM, et al. ‘Meta-guidelines’ for the management of patients with schizophrenia. CNS Spectr 2013; 18: 150–62.Google Scholar
29 Stahl, SM. Antipsychotic polypharmacy, part I: therapeutic option or dirty little secret? J Clin Psychiatry 1999; 60: 425–6.Google Scholar
30 Stahl, SM. Antipsychotic polypharmacy, part II: tips on use and misuse. J Clin Psychiatry 1999; 60: 506–7.Google Scholar
31 Stahl, SM. Antipsychotic polypharmacy: squandering precious resources? J Clin Psychiatry 2002; 63: 93–4.Google Scholar
32 Stahl, SM, Grady, MM. A critical review of atypical antipsychotic utilization: comparing monotherapy with polypharmacy and augmentation. Cur Med Chem 2004; 11: 313–26.Google Scholar
33 Stahl, SM. Focus: antipsychotic polypharmacy – evidence based prescribing or prescribing based evidence? Int J Neuropsychopharmacol 2004; 7: 113–6.Google Scholar
34 Stahl, SM, Grady, M. High cost use of second-generation antipsychotics under California's Medicaid program. Psychiatr Serv 2004; 57: 127–9.Google Scholar
35 Stahl, SM. Antipsychotic polypharmacy: evidence based or eminence based? Acta Psychiatr Scand 2002; 106: 321–2.Google Scholar
36 Stahl, SM, Freudenreich, O, Goff, D. A successful antipsychotic combination trial. Quo Vadis? Acta Psychiatr Scand 2004; 110: 241–2.Google Scholar
37 Suzuki, T, Uchida, H, Tanaka, KF, Nomura, K, Takano, H, Tanabe, A, et al. Revising polypharmacy to a single antipsychotic regimen for patients with chronic schizophrenia. Int J Neuropsychopharmacol 2004; 7: 133–42.Google Scholar
38 Correll, CU, Rummel-Kluge, C, Corves, C, Kane, JM, Leucht, S. Antipsychotic combinations vs monotherapy in schizophrenia: a meta-analysis of randomized controlled trials. Schizophr Bull 2009; 35: 443–57.Google Scholar
39 Davis, JM, Chen, N. Dose response and dose equivalence of antipsychotics. J Clin Psychopharmacol 2004; 24: 192208.Google Scholar
40 Mauri, MC, Volonteri, LS, Colasanti, A, Fiorentini, A, De Gaspari, IF, Bareggi, SR. Clinical pharmacokinetics of atypical antipsychotics: a critical review of the relationship between plasma concentrations and clinical response. Clin Pharmacokinet 2007; 46: 359–88.Google Scholar
41 Barnes, TR, Paton, C. Antipsychotic polypharmacy in schizophrenia: benefits and risks. CNS Drugs 2011; 25: 383–99.Google Scholar
Submit a response

eLetters

No eLetters have been published for this article.