The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Full Access

Appendix C.  Review of Research Evidence Supporting Guideline Statements

Appendix C.

Assessment and Determination of Treatment Plan

Statement 1: Assessment of Possible Schizophrenia

APA recommends (1C) that the initial assessment of a patient with a possible psychotic disorder include the reason the individual is presenting for evaluation; the patient’s goals and preferences for treatment; a review of psychiatric symptoms and trauma history; an assessment of tobacco use and other substance use; a psychiatric treatment history; an assessment of physical health; an assessment of psychosocial and cultural factors; a mental status examination, including cognitive assessment; and an assessment of risk of suicide and aggressive behaviors, as outlined in APA’s Practice Guidelines for the Psychiatric Evaluation of Adults (3rd edition).

Evidence for this statement comes from general principles of assessment and clinical care in psychiatric practice. Expert opinion suggests that conducting such assessments as part of the initial psychiatric evaluation improves diagnostic accuracy, appropriateness of treatment selection, and treatment safety. For additional details, see Guideline I, “Review of Psychiatric Symptoms, Trauma History, and Psychiatric Treatment History,” Guideline II, “Substance Use Assessment,” Guideline III, “Assessment of Suicide Risk,” Guideline IV, “Assessment of Risk for Aggressive Behaviors,” Guideline V, “Assessment of Cultural Factors,” and Guideline VI, “Assessment of Medical Health,” in the APA Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd Edition (American Psychiatric Association 2016a). A detailed systematic review to support this statement is outside the scope of this guideline; however, less comprehensive searches of the literature did not yield any studies related to this recommendation in the context of schizophrenia treatment. Consequently, the strength of research evidence is rated as low.

Grading of the Overall Supporting Body of Research Evidence for Assessment of Possible Schizophrenia

On the basis of the limitations of the evidence for assessment of possible schizophrenia, no grading of the body of research evidence is possible.

Statement 2: Use of Quantitative Measures

APA recommends (1C) that the initial psychiatric evaluation of a patient with a possible psychotic disorder include a quantitative measure to identify and determine the severity of symptoms and impairments of functioning that may be a focus of treatment.

Evidence for this statement comes from general principles of assessment and clinical care in psychiatric practice. Consequently, the strength of research evidence is rated as low. Expert opinion suggests that conducting such assessments as part of the initial psychiatric evaluation improves diagnostic accuracy, appropriateness of treatment selection, and longitudinal assessment of patient symptoms and treatment effects. This recommendation is also consistent with Guideline VII, “Quantitative Assessment,” as part of the APA Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd Edition (American Psychiatric Association 2016a).

Grading of the Overall Supporting Body of Research Evidence for Use of Quantitative Measures

On the basis of the limitations of the evidence for use of quantitative measures, no grading of the body of research evidence is possible.

Statement 3: Evidence-Based Treatment Planning

APA recommends (1C) that patients with schizophrenia have a documented, comprehensive, and person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments.

Evidence for this statement comes from general principles of assessment and clinical care in psychiatric practice. A detailed systematic review to support this statement was outside the scope of this guideline; however, less comprehensive searches of the literature did not yield any studies that directly related to this recommendation in the context of schizophrenia treatment. Consequently, the strength of research evidence is rated as low. Nevertheless, in the bulk of the literature reviewed in the Agency for Healthcare Research and Quality (AHRQ) report (McDonagh et al. 2017), pharmacotherapy was included in all treatment arms in the studies of psychosocial interventions. Invariably, in studies of pharmacotherapies, some additional form of clinical intervention is incorporated into treatment and can include elements of patient education, supportive psychotherapy, and other brief interventions.

Grading of the Overall Supporting Body of Research Evidence for Evidence-Based Treatment Planning

On the basis of the limitations of the evidence for evidence-based treatment planning, no grading of the body of research evidence is possible.

Pharmacotherapy

Statement 4: Antipsychotic Medications

APA recommends (1A) that patients with schizophrenia be treated with an antipsychotic medication and monitored for effectiveness and side effects.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence for this statement comes from the AHRQ review (McDonagh et al. 2017) as well as from other high-quality meta-analyses that examined findings from randomized controlled trials (RCTs) of antipsychotic medications in schizophrenia (Huhn et al. 2019; Leucht et al. 2017). The data from placebo-controlled trials are essential in making an initial determination of whether the benefits of antipsychotic medications outweigh the harms of antipsychotic medications. Placebo-controlled trial data as well as findings from head-to-head comparison studies and network analyses provide additional information on whether the benefits and harms of specific antipsychotic medications suggest preferential use (or nonuse) as compared with other antipsychotic medications. The strength of the research evidence is rated as high in demonstrating that the benefits of treatment with an antipsychotic medication outweigh the harms, although harms are clearly present and must be taken into consideration.

Primary evidence for placebo-controlled antipsychotic trial data came from the systematic review, Bayesian meta-analysis, and meta-regression conducted by Leucht et al. (2017), which included 167 studies (total N  =  28,102) published from 1955 to 2016 that were randomized and double-blinded with placebo control groups. The authors excluded studies of acute treatment with short-acting intramuscular antipsychotic medications and relapse prevention (including studies of long-acting injectable [LAI] antipsychotic agents). Studies of clozapine were excluded because of possible superior efficacy, and studies conducted in China were excluded because of concerns about study quality. Studies were also excluded if subjects had primarily negative symptoms or significant comorbidity in either psychiatric or physical health conditions. The median study duration was 6 weeks, with almost all studies lasting 12 weeks or less in terms of primary study outcomes. None of the studies was focused on first-episode or treatment-resistant samples of subjects, and the mean illness duration was 13.4 years (standard deviation [SD] 4.7), with a mean subject age of 38.7 (SD 5.5). The number of studies available on each drug was highly variable, with chlorpromazine, haloperidol, olanzapine, and risperidone being most often studied, and limited information was available on some antipsychotic medications. Results are provided in Table C–1.

Results of meta-analysis on placebo-controlled trials of antipsychotic treatment

Number of studies

Number of subjects

Measure

95% CI

I2

Comments

All studies

167

28,102

Drug effect size

105

22,741

Mean effect size = 0.47

0.42, 0.51

52%

Any response with drug vs. placebo

97

20,690

Response ratio = 1.93

1.72, 2.19

NNT = 6

Good response

30

8,408

Response ratio = 1.96

1.65, 2.44

NNT = 8; 23% good response with antipsychotic vs. 14% with placebo

At least minimal response

46

8,918

Response ratio = 1.75

1.59, 1.07

NNT = 5; 51% minimal response with antipsychotic vs. 30% with placebo

Discontinuation for any reason

105

22,851

Risk ratio = 1.25

1.20, 1.31

NNT = 11; 38% discontinuation with antipsychotic vs. 56% with placebo

Discontinuation for inefficacy

94

23,017

Risk ratio = 2.09

1.90, 2.32

NNT = 7; 13% discontinuation with antipsychotic vs. 26% with placebo

Positive symptoms

64

18,174

SMD = 0.45

0.40, 0.50

56%

Negative symptoms

69

18,632

SMD = 0.35

0.31, 0.40

42%

Depression

33

9,658

SMD = 0.27

0.20, 0.34

50%

Quality of life

6

1,900

SMD = 0.35

0.16, 0.51

43%

Social functioning

10

3,077

SMD = 0.34

0.21, 0.47

46%

Use of antiparkinsonian medications

63

14,942

Risk ratio = 1.93

1.65, 2.29

NNH = 12; 19% with antipsychotic vs. 10% with placebo

Sedation

86

18,574

Risk ratio = 2.80

2.30, 3.55

54%

14% with antipsychotic vs. 6% with placebo

Weight gain

59

15,219

SMD = -0.43

–0.55, –0.30

73%

Prolactin increase

51

15,219

SMD = -0.43

–0.55, –0.30

91%

QTc prolongation

29

9,833

SMD = -0.19

–0.29, –0.08

80%

Abbreviations. CI = confidence interval; NNH = number needed to harm; NNT = number needed to treat; SMD = standardized mean difference.

Source. Data from Leucht et al. 2017.

Results of meta-analysis on placebo-controlled trials of antipsychotic treatment

Enlarge table

The authors found a moderate benefit of antipsychotic medications, with positive symptoms improving the most but improvements in negative symptoms, depression, quality of life, and social functioning also noted with treatment (Leucht et al. 2017). Side effects were also present but differed substantially among medications. The authors also found, however, that effect sizes for antipsychotic medications have decreased with time over the past 60 years. This seems to result from increasing placebo response rates rather than decreasing medication response, although the benefit of haloperidol as compared with placebo has decreased with time. Not surprisingly, these trends are likely to confound comparisons of newer versus older medications. Although industry sponsorship was associated with a lower effect size as compared with studies funded by other mechanisms, publication bias was observed because of the tendency to avoid publishing studies with no effect of treatment.

In the AHRQ review (McDonagh et al. 2017), few head-to-head comparison studies were available for most of the antipsychotic medications. In terms of functioning, the strength of evidence (SOE) was low. Older second-generation antipsychotics (SGAs; risperidone, olanzapine, quetiapine, ziprasidone) and paliperidone did not differ in terms of global functioning or employment rates, although social functioning with risperidone in an LAI formulation was better than with quetiapine in a single study (Rouillon et al. 2013). Measures of quality of life also showed no difference among older SGAs or between older SGAs and FGAs (specifically, haloperidol and perphenazine) on the basis of a low to moderate SOE.

In terms of response rates (McDonagh et al. 2017), there was no difference between haloperidol and risperidone (16 RCTs, N = 3,452; relative risk [RR] 0.94, 95% confidence interval [CI] 0.87–1.02; moderate SOE), aripiprazole (5 RCTs, N = 2,185; RR 1.01, 95% CI 0.76–1.34; low SOE), quetiapine (6 RCTs, N = 1,421; RR 0.99, 95% CI 0.76–1.30; low SOE), and ziprasidone (6 RCTs, N = 1,283; RR 0.98, 95% CI 0.74–1.30; low SOE). However, response with olanzapine was significantly better than with haloperidol (14 RCTs, N = 4,099; RR 0.86, 95% CI 0.78–0.96; low SOE). In addition, a network meta-analysis of 46 head-to-head RCTs showed a significantly greater likelihood of response with olanzapine (odds ratio [OR] 1.71, 95% CI 1.11–2.68) and risperidone (OR 1.41, 95% CI 1.01–2.00) than quetiapine (low SOE). Olanzapine was also associated with higher remission rates as compared with haloperidol (3 RCTs; pooled RR 0.65, 95% CI 0.45–0.94; I2 = 54%; low SOE), but there was no difference in remission rates between haloperidol and ziprasidone (3 RCTs; RR 0.89, 95% CI 0.71–1.12; low SOE).

In terms of core illness symptoms (e.g., delusions, hallucinations, disorganized thinking), all SGAs that were studied were superior to placebo (standardized mean difference [SMD] –0.33 to –0.88; low SOE; McDonagh et al. 2017). Risperidone (21 RCTs, N = 4,020; mean difference [MD] 3.24, 95% CI 1.62–4.86) and olanzapine (15 RCTs, N = 4,209; MD 2.31, 95% CI 0.44–4.18) were associated with greater improvements in total Positive and Negative Syndrome Scale (PANSS) score as compared with haloperidol (moderate SOE), but no differences were noted in other comparisons of FGAs and SGAs (low SOE). With comparisons among SGAs, clozapine improved core illness symptoms more than other SGAs except for olanzapine (network meta-analysis of 212 RCTs; SMDs on PANSS or Brief Psychiatric Rating Scale [BPRS] –0.32 to –0.55; low SOE); olanzapine and risperidone improved core illness symptoms more than the other SGAs except for each other and paliperidone (SMDs –0.13 to –0.26; low SOE); and paliperidone improved core illness symptoms more than lurasidone and iloperidone (SMDs –0.17; low SOE).

For negative symptoms (McDonagh et al. 2017), haloperidol was less effective than olanzapine (5 RCTs, N = 535; MD based on the Scale for the Assessment of Negative Symptoms scores 2.56, 95% CI 0.94–4.18; moderate SOE), aripiprazole (3 RCTs, N = 1,701; MD 0.80, 95% CI 0.14–1.46), olanzapine (14 RCTs, N = 3,742; MD 1.06, 95% CI 0.46–1.67), and risperidone (22 RCTs, N = 4,142; MD 0.80, 95% CI 0.14–1.46), with the latter findings based on negative symptom scores of the PANSS and having a low SOE. Other comparisons of FGAs versus SGAs showed no effects on negative symptoms (low SOE).

In an additional network meta-analysis of 32 antipsychotic medications, Huhn et al. (2019) included 402 placebo-controlled and head-to-head randomized controlled trials that included a total of 53,463 adult participants with acute symptoms and a diagnosis of schizophrenia or a related disorder. Not included were studies that focused on individuals with a first episode of psychosis or treatment resistance and studies in which individuals had concomitant medical illnesses or a predominance of negative or depressive symptoms. For the majority of antipsychotic medications, treatment was associated with a statistically significant reduction in overall symptoms as compared with placebo, and there were few significant differences between individual drugs. With antipsychotic medications that did not differ significantly from placebo, there were numerical differences favoring the antipsychotic medication, and the number of subjects in the network meta-analysis was small, yielding a wide credible interval (CrI). Only clozapine, amisulpride, zotepine, olanzapine, and risperidone exhibited greater efficacy than many other antipsychotic medications for overall symptoms, with the greatest benefit noted with clozapine (SMD –0.89, 95% CrI –1.08 to –0.71). Discontinuation rates for inefficacy paralleled the findings for treatment efficacy (Huhn et al. 2019). In terms of positive symptoms, negative symptoms, and depressive symptoms, the majority of the medications showed a statistically significant difference from placebo, with the exception of several antipsychotic agents for which sample sizes were small and CrIs were wide. Few studies had assessed effects of antipsychotic medications on social functioning. As in the Leucht et al. (2017) meta-analysis, side-effect profiles differed considerably among the antipsychotic medications.

Few studies assessed effects of antipsychotic medications on self-harm, but among patients at high risk, the International Suicide Prevention Trial (InterSePT; Meltzer et al. 2003) found that clozapine was superior to olanzapine in preventing significant suicide attempts or hospitalization to prevent suicide (hazard ratio [HR] 0.76, 95% CI 0.58–0.97; low SOE).

In terms of dose-response effects on antipsychotic medication effectiveness, standard doses of antipsychotic medications are superior to low or very low dose treatment in reducing the risk of relapse (Uchida et al. 2011a). In addition, there is evidence of a dose-response relationship for many antipsychotic medications in short-term trials of acute efficacy (Davis and Chen 2004).

Overall discontinuation rates and time to discontinuation reflect whether a treatment is effective but also whether it is tolerable. In this regard, a network meta-analysis of 111 studies (McDonagh et al. 2017) found that rates of discontinuation were lower with the following medications:

  • Olanzapine and clozapine as compared with asenapine, cariprazine, iloperidone, lurasidone, olanzapine LAI, quetiapine, risperidone, and ziprasidone (ORs range from 0.42 for clozapine vs. iloperidone to 0.69 for clozapine vs. risperidone)

  • Clozapine as compared with monthly paliperidone palmitate LAI (OR 0.56, 95% CI 0.33–0.96)

  • Olanzapine as compared with paliperidone (OR 0.67, 95% CI 0.50–0.89)

  • Quetiapine extended release (XR) as compared with iloperidone, olanzapine LAI, or quetiapine (ORs 0.26–0.35)

  • Risperidone and aripiprazole as compared with iloperidone or quetiapine (ORs 0.61–0.77).

  • Risperidone and monthly aripiprazole LAI as compared with iloperidone (ORs 0.52 and 0.62, respectively)

Findings on time to discontinuation are more limited and need replication (low SOE), but they suggest that olanzapine may have a longer time to discontinuation than quetiapine, risperidone, and ziprasidone (4 months on the basis of trial data; 1.5–2.2 months shorter on the basis of observational data); clozapine may have a longer time to discontinuation than olanzapine, risperidone, or quetiapine (7.2–7.8 months in Phase 2E of the Clinical Antipsychotic Trials of Intervention Effectiveness [CATIE] study); and risperidone LAI may have a longer time to discontinuation than aripiprazole, clozapine, olanzapine, quetiapine, or ziprasidone (2.6–4 months).

A network meta-analysis (McDonagh et al. 2017), which used data from 90 head-to-head trials of greater than 6 weeks’ duration, found the risk of withdrawals due to adverse events was less with the following medications:

  • Risperidone LAI as compared with clozapine (OR 0.27, 95% CI 0.10–0.71), lurasidone (OR 0.39, 95% CI 0.18–0.84), quetiapine XR (OR 0.43, 95% CI 0.22–0.81), risperidone (OR 0.50, 95% CI 0.25–0.99), and ziprasidone (OR 0.40, 95% CI 0.20–0.82)

  • Olanzapine as compared with clozapine (OR 0.39, 95% CI 0.19–0.79), lurasidone (OR 0.57, 95% CI 0.34–0.94), quetiapine (OR 0.62, 95% CI 0.44–0.87), risperidone (OR 0.72, 95% CI 0.55–0.96), and ziprasidone (OR 0.58, 95% CI 0.41–0.82)

  • Aripiprazole as compared with clozapine (OR 0.43, 95% CI 0.21–0.88) and ziprasidone (OR 0.64, 95% CI 0.44–0.94)

  • Cariprazine as compared with clozapine (OR 0.40, 95% CI 0.17–0.95)

  • Iloperidone as compared with clozapine (OR 0.34, 95% CI 0.13–0.91)

These findings had a low SOE, and head-to-head comparison data were not available for all available antipsychotic medications. For haloperidol, withdrawals due to adverse events were significantly higher than with SGAs (moderate SOE), specifically, aripiprazole (8 RCTs, N = 3,232; RR 1.25, 95% CI 1.07–1.47; I2 = 0%), olanzapine (24 RCTs, N = 5,708; RR 1.89, 95% CI 1.57–2.27; I2 = 0%), risperidone (25 RCTs, N = 4,581; RR 1.32, 95% CI 1.09–1.60; I2 = 0%), and ziprasidone (7 RCTs, N = 1,597; RR 1.68, 95% CI 1.26–2.23; I2 = 0%).

Overall adverse event rates also favored SGAs as compared with haloperidol (moderate SOE), specifically aripiprazole (3 RCTs, N = 1,713; RR 1.11, 95% CI 1.06–1.17; I2 = 0%), risperidone (8 RCTs, N = 1,313; RR 1.20, 95% CI 1.01–1.42; I2 = 84%), and ziprasidone (6 RCTs, N = 1,448; RR 1.13, 95% CI 1.03–1.23; I2 = 31%). Among comparisons between SGAs, no differences in overall adverse events were noted (low to moderate SOE).

In terms of mortality, comparisons were difficult because of the short duration of most studies and the small number of reported events in these clinical trials (incidence rates 0%–1.17%). Nevertheless, there were no significant mortality differences found between asenapine versus olanzapine (2 RCTs; RR 2.49, 95% CI 0.54–11.5; low SOE), quetiapine versus risperidone (2 RCTs; RR 3.24, 95% CI 0.72–14.6; low SOE), and paliperidone palmitate LAI (monthly) versus risperidone LAI (2 RCTs; RR 1.26, 95% CI 0.21–7.49; low SOE). Additional findings from retrospective cohort studies found no significant difference in the risk of all-cause (1 study, N = 48,595) or cardiovascular (2 studies, N = 55,582) mortality between risperidone, olanzapine, and quetiapine (low SOE).

For the additional harms data described in the AHRQ report (McDonagh et al. 2017), evidence was relatively limited and did not adjust for known factors that confound risk. Data on cardiac disease are mixed. A large, good-quality retrospective cohort study found no significant differences in the risk of cardiovascular death, acute coronary syndrome, or ischemic stroke between risperidone and olanzapine or quetiapine in patients ages 18–64 years within the first year of starting the drug. However, a large adverse event database study found that clozapine was significantly associated with myocarditis or cardiomyopathy, whereas olanzapine, quetiapine, and risperidone were not. In contrast, other limited evidence suggested an increased risk of cardiac arrest and arrhythmia with risperidone compared with clozapine, and data from CATIE suggested a higher estimated 10-year risk of coronary heart disease with olanzapine compared with risperidone. As compared with FGAs, the SGA aripiprazole showed a lower likelihood of cardiomyopathy or coronary heart disease.

Findings on neurological side effects such as akathisia and parkinsonism also showed significant variability among the head-to-head comparison studies, which makes it difficult to draw overall conclusions about side-effect rates or risk. For new-onset tardive dyskinesia, overall rates were low (3% of subjects treated with risperidone as compared with 1%–2% for other medications). Nevertheless, findings from observational trials suggested a significant increase in risk with risperidone as compared with olanzapine (OR 1.70, 95% CI 1.35–2.14).

Metabolic effects varied with study duration, but clinically important weight gain (defined as a 7% or more increase from baseline) was greater with olanzapine than with aripiprazole (RR 2.31), asenapine (RR 2.59), clozapine (RR 1.71), quetiapine (RR 1.82), risperidone (RR 1.81), and ziprasidone (RR 5.76) across 3.7–24 months. Olanzapine had a significantly greater risk of metabolic syndrome than risperidone (pooled OR 1.60, 95% CI 1.10–2.21; I2 = 0%; follow-up of 6 weeks to 3 months) or aripiprazole (pooled OR 2.50, 95% CI 1.32–4.76; I2 = 0%; follow-up of 3.5–12 months). In adults, observational evidence indicated an increased risk of new-onset diabetes with olanzapine compared with risperidone (OR 1.16, 95% CI 1.03–1.31). A single study found diabetic ketoacidosis to be increased with olanzapine compared with risperidone (OR 3.5, 95% CI 1.7–7.9), but a second study found no difference in diabetic ketoacidosis, hyperglycemia, or hyperglycemic hyperosmolar state between risperidone and olanzapine, regardless of age group, but a significantly lower risk with quetiapine compared with risperidone in older patients (adjusted HR 0.69, 95% CI 0.53–0.90).

Taken together, the findings of the AHRQ review (McDonagh et al. 2017) complement the meta-analyses of Leucht et al. (2017) and Huhn et al. (2019) in showing efficacy of antipsychotic medications, particularly for core illness symptoms but also for other outcomes. Furthermore, research evidence demonstrates no clear and consistent superiority of one antipsychotic medication as compared with other antipsychotic medications, with the exception of clozapine. In addition, the systematic reviews suggest considerable variability in side-effect profiles among antipsychotic medications, without a clear continuum of risk for individual medications when all side effects are considered.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Antipsychotic Medications

  • Magnitude of effect: Moderate. The magnitude of effect varies among individual antipsychotic medications but is moderate overall on the basis of findings from meta-analyses of placebo-controlled trials.

  • Risk of bias: Medium. Studies are RCTs that are summarized in multiple good-quality meta-analyses. Although the risk of bias of individual RCTs varies, most have some limitations, and, in older trials, reporting of study design features is often incomplete. Among head-to-head comparison trials, some studies are observational trials and are associated with a higher risk of bias.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The studies include subjects from countries around the world, with the exception of China. The doses of medication used are representative of usual clinical practice.

  • Directness: Direct. Studies measure functioning, quality of life, core illness symptoms, negative symptoms, and response to treatment.

  • Consistency: Consistent. When multiple studies that included a given comparison are available, results are generally consistent. In addition, the overall direction of effects is generally consistent among antipsychotic medications in placebo-controlled trials.

  • Precision: Variable. For many comparisons, particularly when multiple RCTs are available, findings are precise. However, for other comparisons, imprecision is present because of wide confidence intervals that often cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Present. There is evidence of a dose-response relationship in acute treatment trials as well as in studies of antipsychotic medications for relapse prevention.

  • Confounding factors: Present. In placebo-controlled trials, effect sizes have decreased over the past 60 years, apparently due to increases in placebo response rates; these trends are likely to confound comparisons of older and newer medications.

  • Publication bias: Suspected. Among placebo-controlled trials, studies with no effect of treatment appear to have had lower rates of publication.

  • Overall strength of research evidence: High. There are a large number of randomized, double-blind, placebo-controlled trials of antipsychotic medication as well as a smaller number of head-to-head comparison RCTs. Although many studies have a medium risk of bias and publication bias appears to be present, there is also consistency in overall study findings and a dose-response relationship is present, strengthening confidence in the conclusions.

Grading of the Overall Supporting Body of Research Evidence or Harms of Antipsychotic Medications

  • Magnitude of effect: Small to moderate. The magnitude of effect for harms of antipsychotic medication differs by drug and by side effect but is small to moderate overall.

  • Risk of bias: Medium to high. Studies are RCTs that are summarized in multiple good-quality meta-analyses. Particularly in older clinical trials, side effects tend not to be assessed or reported as systematically as efficacy and effectiveness-related outcomes.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The studies include subjects from around the world, with the exception of China. The doses of medication used are representative of usual clinical practice.

  • Directness: Variable. Most studies measure overall adverse events, and some measure specific adverse effects, each of which is a direct measure. Other studies measure study withdrawal rates due to adverse effects, which is an indirect measure.

  • Consistency: Consistent. In studies that compare the same medication with placebo, side effect–related outcomes are generally consistent in their direction and relative degree. Head-to-head comparison data are less consistent.

  • Precision: Precise. Confidence intervals are narrow for comparisons in which multiple studies with good sample sizes are available. For comparisons with a small number of studies or small samples, imprecision is present because of wide confidence intervals. Head-to-head comparisons also have imprecision due to outcomes that cross the threshold for clinically significant harms of the intervention.

  • Dose-response relationship: Suspected. There is less systematic information available on dose-response relationships for side effects of antipsychotic medication; however, the available evidence suggests that greater doses are associated with a greater degree of medication-related side effects.

  • Confounding factors: Present. Cohort effects that are present in efficacy and effectiveness studies of antipsychotic medication are also likely to be relevant when assessing harms of antipsychotic medication.

  • Publication bias: Suspected. Among placebo-controlled trials, studies with no effect of treatment appear to have had lower rates of publication.

  • Overall strength of research evidence: Moderate. Available studies are RCTs that are generally of moderate quality and have good sample sizes. Findings are consistent, with narrow confidence intervals for many comparisons, and are likely to exhibit a dose-response relationship.

Antipsychotic Medications in First-Episode Schizophrenia

In subgroup analyses, the AHRQ review (McDonagh et al. 2017) found that patients experiencing a first episode of schizophrenia did not show significant differences in response or remission when treated with olanzapine, quetiapine, risperidone, ziprasidone, aripiprazole, or paliperidone. Another systematic review (Zhu et al. 2017) in individuals with a first episode of schizophrenia found that amisulpride, olanzapine, ziprasidone, and risperidone reduced overall symptoms more than haloperidol, but the evidence was noted as being of very low to moderate quality, and only 13 studies were available to address this clinical question.

Treatment Approaches to Partial Response or Nonresponse

High Doses of Antipsychotic Medication

A limited amount of evidence suggests no benefit from high doses of an antipsychotic medication in individuals who have not responded to typical doses of the medication. A systematic review and meta-analysis by Dold et al. (2015) found 5 trials, which included a total of 348 patients and studied this question with FGAs or SGAs. Dose escalation was not found to confer any benefits in terms of study attrition, response rates, or symptoms (as measured by PANSS or BPRS). A subsequent systematic review and meta-analysis by Samara et al. (2018) found 10 relevant RCTs, which included a total of 675 participants. Although no clear differences in response were noted between subjects who received the same dose of medication as compared with those who received a higher dose, many of the studies had a medium to high risk of bias. There were also no differences in other outcomes, including the proportion of individuals who left the study early because of adverse effects or for any reason.

Augmentation Pharmacotherapy

A number of pharmacotherapies have been studied as augmentation strategies in individuals with treatment-resistant schizophrenia. Evidence has been primarily from small short-term, open-label studies that have yielded mixed findings. Correll et al. (2017b) conducted a systematic search for meta-analyses that addressed the effects of combining an antipsychotic medication with another pharmacotherapy in individuals with schizophrenia. They found 29 meta-analyses that together encompassed 19,833 subjects in 381 trials and that evaluated 42 augmentation strategies. Although 14 of these augmentation therapies showed better outcomes than comparison treatment, the meta-analyses with the highest effect sizes had the lowest quality of included studies, undermining confidence in the benefits of augmentation.

In terms of augmentation of clozapine, Siskind et al. (2018) conducted a systematic review and meta-analysis of augmentation strategies for individuals with clozapine-refractory schizophrenia and found 46 studies of 25 interventions. They noted possible benefits of memantine for negative symptoms and aripiprazole, fluoxetine, and sodium valproate for overall psychotic symptoms but found that many of the studies had a poor study quality and short periods of follow-up, which limited the ability to draw conclusions. Wagner et al. (2019a) conducted a systematic meta-review of 21 meta-analyses that examined strategies for augmenting treatment with clozapine. Although the best evidence was available for combination treatment of clozapine with FGAs or SGAs for psychotic symptoms and with antidepressants for persistent negative symptoms, these authors also concluded that additional high-quality clinical trials are essential before making definitive statements about clozapine augmentation. Furthermore, their findings are consistent with those of Correll et al. (2017b), who did not identify any combination medication strategies with clozapine that led to better outcomes than comparator treatments and found that available studies were of low quality.

Other meta-analyses have also examined the effects of using more than one antipsychotic medication as compared with antipsychotic monotherapy. Galling et al. (2017) found a possible benefit of aripiprazole augmentation in terms of greater improvement in negative symptoms and reductions in prolactin levels and body weight. However, they noted that the apparent benefits of antipsychotic augmentation in reducing total symptoms were no longer seen when the analysis was restricted to double-blind trials of higher quality. A Cochrane review of antipsychotic combination treatments for schizophrenia (Ortiz-Orendain et al. 2017) also found that evidence on combinations of antipsychotic medications was of very low quality. Nevertheless, data from a large nationwide cohort study in Finland suggested that use of two different antipsychotic medications may have some benefits as compared with monotherapy. Tiihonen et al. (2019) studied 62,250 patients with a diagnosis of schizophrenia and compared hospitalization rates within the same individual during periods of antipsychotic monotherapy and periods with use of more than one antipsychotic medication. They found that rehospitalization rates with clozapine were lower than with other monotherapies and that individuals receiving more than one antipsychotic medication had a 7%–13% lower risk of psychiatric rehospitalization than individuals treated with monotherapy (P < 0.001). Use of multiple antipsychotic medications was also associated with a reduction in secondary outcomes (e.g., all-cause hospitalization, nonpsychiatric hospitalization, mortality). Thus, there is weak and inconsistent evidence suggesting possible benefits of combined treatment with more than one antipsychotic medication, but more research is needed.

On the other hand, augmentation of antipsychotic therapy with an antidepressant medication may be helpful, particularly for patients with negative symptoms or depression. Stroup et al. (2019) used U.S. Medicaid data on 81,921 adult outpatients ages 18–64 years who had a diagnosis of schizophrenia. The authors employed propensity score matching and weighted Cox proportional hazards regression models to examine the effect of adding an antidepressant, a benzodiazepine, a mood stabilizer, or another antipsychotic medication to existing treatment with an antipsychotic medication. These authors found that the addition of an antidepressant medication was associated with a reduced risk for psychiatric hospitalization or emergency visits. In addition, Helfer et al. (2016) conducted a systematic review and meta-analysis of the addition of antidepressant medication to antipsychotic treatment. Data from 82 RCTs that included 3,608 subjects indicated that antidepressant augmentation was associated with improvements in quality of life (SMD –0.32, 95% CI –0.57 to –0.06) and rates of response (risk ratio 1.52, 95% CI 1.29–1.78; NNT = 5, 95% CI 4–7) as well as greater reductions in depressive symptoms (SMD –0.25, 95% CI –0.38 to –0.12), positive symptoms (SMD –0.17, 95% CI –0.33 to –0.01), negative symptoms (SMD –0.30, 95% CI –0.44 to –0.16), and overall symptoms (SMD –0.24, 95% CI –0.39 to –0.09).

Statement 5: Continuing Medications

APA recommends (1A) that patients with schizophrenia whose symptoms have improved with an antipsychotic medication continue to be treated with an antipsychotic medication.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence in support of this statement is based primarily on the evidence for antipsychotic efficacy in improving symptoms and quality of life as well as promoting functioning (see Statement 4 earlier in the appendix). Thus, the strength of research evidence is rated as high.

Additional evidence supporting this statement comes from registry database studies and from discontinuation studies. For example, in a nationwide prospective registry study (N = 6,987) with a 5-year follow-up of individuals with first-onset schizophrenia (Kiviniemi et al. 2013), there was a significant decrease in all-cause mortality in individuals taking SGAs as compared with individuals who were not taking antipsychotic medication (OR 0.69; P = 0.005).

Another nationwide study (N = 8,719) using prospectively collected registry data found that the lowest rates of rehospitalization or death occurred in individuals who received continuing treatment with an antipsychotic medication for up to 16.4 years (Tiihonen et al. 2018). Individuals who discontinued antipsychotic medication had a risk of death that was 174% higher than that in continuous users of antipsychotic medications (HR 2.74, 95% CI 1.09–6.89), whereas the risk of death was 214% higher (HR 3.14, 95% CI 1.29–7.68) in nonusers of antipsychotic medications as compared with continuous users. Rates of treatment failure, which included rehospitalization as well as death, were also lower in individuals who received continuous treatment with an antipsychotic medication. More specifically, 38% of those who discontinued treatment experienced treatment failure as compared with a matched group of continuous users of an antipsychotic medication, in which the rate of treatment failure was 29.3%. For nonusers of antipsychotic medication, treatment failure occurred in 56.5% as compared with 34.3% of a matched group of continuous antipsychotic medication users.

Several meta-analyses have examined mortality-related data with antipsychotic treatment. A meta-analysis of studies with follow-up periods of at least 1 year found that mortality was increased in individuals who did not receive antipsychotic medication as compared with those who were treated with an antipsychotic medication (pooled risk ratio 0.57, 95% CI 0.46–0.76; P < 0.001 based on 22,141 deaths in 715,904 patient years in 4 cohort studies) (Vermeulen et al. 2017). With continuous treatment with clozapine, mortality was found to be lower in long-term follow-up (median 5.4 years) as compared with treatment with other antipsychotic medications (mortality rate ratio 0.56, 95% CI 0.36–0.85, P = 0.007 based on 1,327 deaths in 217,691 patient years in 24 studies) (Vermeulen et al. 2019).

On the basis of 10 RCTs (total N = 776) with mean study duration of 18.6 ± 5.97 months, a meta-analysis of discontinuation studies (Kishi et al. 2019) concluded that relapse rates were lower in individuals with schizophrenia who continued treatment with an antipsychotic medication as compared with those who discontinued treatment (RR 0.47, 95% CI 0.35–0.62; P < 0.00001; I2 = 31%; NNT = 3). An additional meta-analysis (Thompson et al. 2018), using somewhat different inclusion and exclusion criteria for studies, also found that relapse rates were lower in individuals who received maintenance treatment (N = 230; 19%; 95% CI 0.05%–37%) as compared with those who stopped the antipsychotic medication (N = 290; 53%; 95% CI 39%–68%). Although caution may be needed in interpreting these results because of methodological considerations (Moncrieff and Steingard 2019), the findings align with expert opinion on the benefits of maintenance treatment with an antipsychotic medication (Goff et al. 2017).

Grading of the Overall Supporting Body of Research Evidence for the Efficacy of Continuing Treatment With an Antipsychotic Medication

  • Magnitude of effect: Large. The magnitude of effect is large in terms of lower relapse rates and lower mortality for individuals who received maintenance treatment with antipsychotic medications as compared with discontinuation of antipsychotic medication.

  • Risk of bias: Medium. Studies include RCTs of antipsychotic discontinuation and observational studies using registry data. Although the registry studies have a greater risk of bias than RCTs, they use prospectively collected data and have good observational study designs.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The doses of medication used are representative of usual clinical practice. The observational studies include data from a nationwide registry and have broad generalizability, in contrast to RCTs with more restrictive inclusion and exclusion criteria. However, the applicability of registry data from Nordic countries may be reduced by differences in the health care delivery system as compared with that of the United States.

  • Directness: Direct. Studies measured relapse rates and mortality.

  • Consistency: Consistent. Findings showing benefits of maintenance antipsychotic treatment are consistent among the different studies and study designs.

  • Precision: Variable. Most meta-analyses have narrow confidence intervals that do not cross the threshold for clinically significant benefit of treatment; however, some studies have wider confidence intervals.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Unclear. It is possible that missing data or cohort-related effects may influence the results from multiyear registry databases. For long-term follow-up studies, which are needed to assess long-term effects of antipsychotic medication, loss of individuals to follow-up and changes in treatment over time may also confound data interpretation.

  • Publication bias: Not assessed.

  • Overall strength of research evidence: High. Available studies include RCTs with a medium risk of bias. These RCTs are complemented by prospective registry studies with very large sample sizes. Confidence intervals for most outcomes are relatively narrow, and findings are consistent in showing substantial benefit for continued antipsychotic medication treatment.

Grading of the Overall Supporting Body of Research Evidence for the Harms of Continuing Treatment With an Antipsychotic Medication

See Statement 4, subsection “Grading of the Overall Supporting Body of Research Evidence for Harms of Antipsychotic Medications,” earlier in the appendix.

Statement 6: Continuing the Same Medications

APA suggests (2B) that patients with schizophrenia whose symptoms have improved with an antipsychotic medication continue to be treated with the same antipsychotic medication.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence in support of this statement includes the evidence described for antipsychotic efficacy (see Statement 4 earlier in the appendix) and the evidence for continuing with antipsychotic treatment (see Statement 5 earlier in the appendix). Additional evidence that specifically addresses this guideline statement comes from randomized trials of a change in antipsychotic medication. On the basis of these studies, the strength of research evidence is rated as moderate.

The CATIE study provided important findings on medication changes (Essock et al. 2006). At the time of randomization, some individuals happened to be randomly assigned to a medication that they were already taking, whereas other individuals were assigned to a different antipsychotic medication. Individuals who were assigned to change to a different antipsychotic medication (N = 269) had an earlier time to all-cause treatment discontinuation than those assigned to continue taking the same antipsychotic medication (N = 129; Cox proportional HR 0.69; P = 0.007). Although a change from olanzapine to a different antipsychotic medication was beneficial in terms of weight gain, there were no other differences in outcome measures for individuals who switched medications as compared with those who continued with the same treatment (Rosenheck et al. 2009).

Additional evidence comes from an RCT aimed at reducing the metabolic risk of antipsychotic treatment by changing medication from olanzapine, quetiapine, or risperidone to aripiprazole (Stroup et al. 2011). Individuals were followed for 24 weeks after being assigned to continue taking their current medication (N = 106) or to switch to aripiprazole (N = 109). Although the two groups did not differ in the proportion of individuals with medication efficacy (as measured by the PANSS total score or change in Clinical Global Impression [CGI] severity score), individuals who switched medication were more likely to stop medication (43.9% vs. 24.5%; P = 0.0019), and treatment discontinuation occurred earlier in those who switched medication as compared with those who did not (HR 0.456, 95% CI 0.285–0.728; P = 0.0010). However, modest but statistically significant changes did occur in weight, serum non-high-density lipoprotein cholesterol, and serum triglycerides in individuals who switched to aripiprazole as compared with those who continued with olanzapine, quetiapine, or risperidone.

Together, these findings suggest that changes in antipsychotic medications may be appropriate for addressing significant side effects such as weight or metabolic considerations, but switching medications may also confer an increased risk of medication discontinuation, with associated risks of increased relapse and increased mortality.

Grading of the Overall Supporting Body of Research Evidence for the Efficacy of Continuing the Same Antipsychotic Medication

  • Magnitude of effect: Moderate. Evidence from two RCTs suggests that a change in medication is associated with a moderate risk of earlier treatment discontinuation compared with continuing the same medication.

  • Risk of bias: Medium. Studies are RCTs with a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The studies were conducted in the United States. Doses of medication used are representative of usual clinical practice. The available RCTs examine changes in medication aimed at reducing metabolic effects of treatment, and a change from a high metabolic risk medication to a low metabolic risk medication may not be representative of other medication changes.

  • Directness: Indirect. Studies measure all-cause treatment discontinuation, which combines effects due to inefficacy and lack of tolerability.

  • Consistency: Consistent. The two studies are consistent in showing benefits of continuing with the same antipsychotic medication.

  • Precision: Precise. Confidence intervals are narrow and do not cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Absent.

  • Publication bias: Unable to be assessed.

  • Overall strength of research evidence: Moderate. The two RCTs that assess changing from one antipsychotic to another have good sample sizes and a medium risk of bias. Their findings are consistent with each other and with the results of studies discussed for Statements 4 and 5 on the benefits of antipsychotic medication treatment.

Grading of the Overall Supporting Body of Research Evidence for the Harms of Continuing the Same Antipsychotic Medication

See Statement 4, subsection “Grading of the Overall Supporting Body of Research Evidence for Harms of Antipsychotic Medications,” earlier in the appendix.

Statement 7: Clozapine in Treatment-Resistant Schizophrenia

APA recommends (1B) that patients with treatment-resistant schizophrenia be treated with clozapine.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence on clozapine comes from multiple RCTs, observational studies (including clinical trials and studies using administrative databases), and meta-analyses. In some instances, the studies were limited to individuals with treatment-resistant schizophrenia, whereas in other studies a formal determination of treatment resistance was not reported or possible. Nevertheless, most information about clozapine will be of relevance to patients with treatment-resistant schizophrenia because, in current practice, most individuals receive clozapine only after a lack of response to other treatments.

In comparisons of SGAs, the AHRQ report (McDonagh et al. 2017) found that, independent of prior treatment history, clozapine improved core illness symptoms more than other SGAs (except for olanzapine) and was associated with a lower risk of suicide or suicide attempts than olanzapine, quetiapine, and ziprasidone (low SOE). In addition, in treatment-resistant patients, clozapine treatment was associated with a lower rate of treatment discontinuation due to lack of efficacy than the other SGAs that were studied. It is not clear whether rates of overall treatment discontinuation with clozapine may be influenced by the increased frequency of clinical interactions related to the more intensive monitoring with clozapine as compared with other antipsychotic medications.

The AHRQ review drew on several meta-analyses related to treatment-resistant schizophrenia (Ranasinghe and Sin 2014; Samara et al. 2016; Souza et al. 2013); however, some additional studies are also relevant to this guideline statement. A meta-analysis by Siskind et al. (2016b) had considerable overlap with the meta-analysis of Samara et al. (2016) in terms of the included studies. Despite this, the findings of the two meta-analyses were somewhat different, likely due to differences in the inclusion criteria and analytic approach (Samara and Leucht 2017). Samara et al. (2016) found few significant differences in outcomes and did not find clozapine to be significantly better than most other drugs in treatment-resistant schizophrenia. Siskind et al. (2016b) found no difference for clozapine compared with other antipsychotic medications in long-term studies but did find clozapine to be superior to other medications in short-term studies and across all studies in reducing total psychotic symptoms (24 studies, N = 1,858; P < 0.005). Similarly, in terms of response to treatment (as reflected by a 20%–30% reduction in symptoms), clozapine showed higher rates of response than comparators in short-term studies of treatment-resistant schizophrenia (8 studies, total N = 598 for clozapine, 620 for comparators; RR 1.17, 95% CI 1.07–2.7; P = 0.03; absolute risk reduction 12.48%, 95% CI 7.52–17.43; NNT = 9). Again, however, studies that assessed long-term response showed no difference between clozapine and comparators. A greater benefit of clozapine than comparators was also seen when the analysis was limited to non-industry-funded trials (6 studies, N =  208; RR 1.68, 95% CI 1.20–2.35; P = 0.002). In a subsequent meta-analysis using data from the same studies, Siskind et al. (2017b) found that 40.1% of treatment-resistant individuals who received clozapine had a response, with a reduction of PANSS scores of 25.8% (22 points on the PANSS) from baseline.

In an additional network meta-analysis of 32 antipsychotic medications, Huhn et al. (2019) analyzed 402 placebo-controlled and head-to-head RCTs that included a total of 53,463 adult participants with acute symptoms and a diagnosis of schizophrenia or a related disorder. Studies that focused on individuals with a first episode of psychosis or treatment resistance were excluded, as were studies in which individuals had concomitant medical illnesses or a predominance of negative or depressive symptoms. Only clozapine, amisulpride, zotepine, olanzapine, and risperidone exhibited greater efficacy than many other antipsychotic medications for overall symptoms, with the greatest benefit noted with clozapine (SMD –0.89, 95% CrI –1.08 to –0.71). Clozapine also was statistically better than placebo and the majority of the other antipsychotic medications in terms of all-cause discontinuation (SMD 0.76, 95% CrI 0.59–0.92) and its effects on positive symptoms (SMD –0.64, 95% CrI –1.09 to –0.19), negative symptoms (SMD 0.62, 95% CrI –0.84 to –0.39), and depressive symptoms (SMD –0.52, 95% CrI –0.82 to –0.23).

Findings from studies using administrative databases also suggest benefits of treatment with clozapine. For example, a prospective nationwide study conducted over a 7.5-year period in Sweden (Tiihonen et al. 2017) found significantly reduced rates of rehospitalization with the use of clozapine as compared with no antipsychotic treatment (HR 0.53, 95% CI 0.48–0.58). In addition, the reduction in rehospitalization with clozapine was comparable to reductions in rehospitalization with LAI antipsychotic medications, whereas other oral formulations of antipsychotic medications had higher risks of rehospitalization. In comparison with oral olanzapine, clozapine had a lower rate of treatment failure (HR 0.58, 95% CI 0.53–0.63) that was comparable to the rate of treatment failure with LAI antipsychotic medications (range of HRs 0.65–0.80).

Similar benefits of clozapine were found in analysis of prospective registry data from Finland obtained for all persons with schizophrenia who received inpatient care from 1972 to 2014 (Taipale et al. 2018a). Of the 62,250 individuals in the prevalent cohort, 59% were readmitted during follow-up time of up to 20 years (median follow-up duration 14.1 years). Among oral antipsychotic medications, clozapine was associated with the lowest risk for psychiatric readmission compared with no antipsychotic use (HR 0.51, 95% CI 0.49–0.53) and for all-cause readmission (HR 0.60, 95% CI 0.58–0.61). For the 8,719 individuals with a first episode of schizophrenia, risks of psychiatric readmission and all-cause readmission were also reduced (HR 0.45, 95% CI 0.40–0.50 and HR 0.51, 95% CI 0.47–0.56, respectively).

A meta-analysis that examined effects of clozapine on hospital use also found benefits for clozapine (Land et al. 2017). Although the vast majority of studies in the meta-analysis were observational studies, use of clozapine as compared with other antipsychotic medications was associated with a significant decrease in the proportion of individuals who were hospitalized (22 studies, N = 44,718; RR 0.74, 95% CI 0.69–0.80; P < 0.001), although the time to rehospitalization did not differ.

In terms of suicide risk, subjects in the InterSePT study (Meltzer et al. 2003) who met criteria for treatment-resistant schizophrenia showed benefits of clozapine that were comparable to the benefits seen in the overall sample. For the sample as a whole, clozapine was superior to olanzapine in preventing significant suicide attempts or hospitalization to prevent suicide in high-risk patients (HR 0.76, 95% CI 0.58–0.97). Fewer clozapine-treated patients in the InterSePT study attempted suicide (P = 0.03); required hospitalizations (P = 0.05) or rescue interventions (P = 0.01) to prevent suicide; or required concomitant treatment with antidepressants (P = 0.01) or with anxiolytics or soporifics (P = 0.03). Subjects treated with clozapine were also less likely to have CGI severity of suicidality scale ratings of “much worse” or “very much worse” (HR 0.78, 95% CI 0.61–0.99) than subjects treated with olanzapine.

In terms of mortality risk, a population-based cohort study of 2,370 patients with treatment-resistant schizophrenia found a higher rate of self-harm in individuals treated with non-clozapine antipsychotic medications than in those treated with clozapine (HR 1.36, 95% CI 1.04–1.78) (Wimberley et al. 2017). There was also a higher rate of all-cause mortality in patients not receiving clozapine than in those treated with clozapine (HR 1.88, 95% CI 1.16–3.05); however, the comparator group included individuals who were not taking any antipsychotic medication. When the study subjects were limited to those who were adhering to treatment, the higher mortality during treatment with other antipsychotic medications did not reach statistical significance. In the year after clozapine discontinuation, an increase in mortality was observed (HR 2.65, 95% CI 1.47–4.78), consistent with benefits of clozapine treatment in reducing overall mortality. Another cohort study also found significant benefits of clozapine on all-cause mortality in individuals with treatment-resistant schizophrenia (adjusted HR 0.61, 95% CI 0.38–0.97; P = 0.04) (Cho et al. 2019). These findings are also consistent with results of a meta-analysis that showed significantly lower rates of long-term crude mortality in patients who received continuous treatment with clozapine as compared with patients treated with other antipsychotic medications (mortality rate ratio 0.56, 95% CI 0.36–0.85; P = 0.007) (Vermeulen et al. 2019).

In terms of side effects with clozapine, a network meta-analysis conducted as part of the AHRQ report (McDonagh et al. 2017) showed that clozapine had a higher risk of study withdrawal due to adverse events than some other SGAs (low SOE) but did not show differences in overall rates of adverse events as compared with risperidone (low SOE). In the meta-analysis by Siskind et al. (2016b), individuals treated with clozapine had a higher likelihood of experiencing sialorrhea (P < 0.001; number needed to harm [NNH] = 4), seizures (P < 0.05; NNH = 17), tachycardia (P < 0.01; NNH = 7), fever (P < 0.01; NNH = 19), dizziness (P < 0.01; NNH = 11), sedation (P < 0.001; NNH = 7), constipation (P < 0.05; NNH = 12), and nausea or vomiting (P < 0.05; NNH = 19) than individuals treated with comparator antipsychotic medications. In the meta-analysis by Leucht et al. (2013), all-cause treatment discontinuation was less likely with clozapine than placebo (OR 0.46, 95% CI 0.32–0.65), as were extrapyramidal side effects (OR 0.3, 95% CI 0.17–0.62). In contrast, weight gain (SMD 0.65, 95% CI 0.31–0.99) and sedation (OR 8.82, 95% CI 4.72–15.06) were more likely with clozapine than placebo.

In an Australian national survey of 1,049 people with a diagnosis of schizophrenia or schizoaffective disorder who reported taking any antipsychotic medication (Siskind et al. 2017a), the proportion of individuals with diabetes, obesity, and metabolic syndrome was higher in individuals taking clozapine as compared with other antipsychotic medications (adjusted ORs 1.744, 1.899, and 2.300, respectively; P < 0.001). In addition, clozapine was associated with a greater proportion of individuals with dry or watery mouth (adjusted OR 2.721; P < 0.001), difficulty swallowing (adjusted OR 1.754; P < 0.01), constipation (adjusted OR 1.996; P < 0.001), dizziness/vertigo (adjusted OR 1.571; P < 0.01), and palpitations (adjusted OR 1.543; P < 0.05). The proportion of individuals who reported trembling or shaking was significantly less in those treated with clozapine as compared with other antipsychotic agents (adjusted OR 0.581; P < 0.01).

In the network meta-analysis by Huhn et al. (2019), individuals treated with clozapine were less likely to require use of an antiparkinsonian medication (SMD 0.46, 95% CrI 0.19–0.88) than those treated with other antipsychotic agents or placebo. However, clozapine was associated with a greater degree of weight gain (SMD 2.37, 95% CrI 1.43–3.32), sedation (SMD 3.02, 95% CrI 2.52–3.37), and experiencing at least one anticholinergic side effect (SMD 2.21, 95% CrI 1.26–3.47) than placebo.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Clozapine in Treatment-Resistant Schizophrenia

  • Magnitude of effect: Moderate. The magnitude of clozapine’s effect varies with the study design and inclusion criteria. Some meta-analyses of RCTs show no difference for clozapine, but most studies show significant benefit, at least in the short term. Observational studies also show a magnitude of effect that is at least moderate.

  • Risk of bias: Medium. Studies include RCTs and observational studies, primarily registry studies. Most studies have some limitations based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. Most individuals who receive treatment with clozapine have had at least one trial of another antipsychotic medication, and most would meet usual clinical criteria for treatment-resistant schizophrenia, even when this is not well specified in the study description. The doses of medication used are representative of usual clinical practice.

  • Directness: Variable. Studies measure psychotic symptoms, response to treatment, all-cause treatment discontinuation, psychiatric hospitalization, all-cause hospitalization, depression, and mortality. Some of these outcomes are directly related to the review questions, and some are indirectly related.

  • Consistency: Inconsistent. Although most meta-analyses and observational studies show benefits for clozapine, not all meta-analyses show superiority of clozapine to other antipsychotic medications in individuals with treatment-resistant schizophrenia.

  • Precision: Variable. Some confidence intervals are narrow without overlapping the threshold for clinically significant benefits, whereas other confidence intervals are wide or overlapping.

  • Dose-response relationship: Present. Increases in dose and corresponding increases in blood levels of clozapine appear to be related to improved clinical efficacy in nontoxic ranges of dosing.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Additional monitoring and an increased frequency of clinical contacts with clozapine may enhance the effects of the medication relative to other antipsychotic medications, at least in observational studies.

  • Publication bias: Unclear. Although publication bias for clozapine-specific studies was not tested, publication bias is relatively common in studies of psychopharmacology because of nonpublication of negative studies.

  • Overall strength of research evidence: Moderate. The available studies include RCTs of moderate quality and good sample sizes. The effect sizes for clozapine vary among meta-analyses and outcomes. However, most studies, including RCTs and prospective observational studies, show benefits of clozapine as compared with other antipsychotic medications.

Grading of the Overall Supporting Body of Research Evidence for Harms of Clozapine

  • Magnitude of effect: Moderate. The magnitude of effect is moderate overall but varies with the specific side effect. As compared with other antipsychotic medications, clozapine is associated with a greater risk of weight gain, sialorrhea, sedation, metabolic effects, seizures, constipation, anticholinergic side effects, tachycardia, and dizziness but a lower risk of all-cause treatment discontinuation, extrapyramidal side effects, or need for anticholinergic medication.

  • Risk of bias: Medium. Studies include RCTs and a large observational study of patient-reported side effects. RCTs are of low to medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts, whereas the observational study has a high risk of bias.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The doses of medication used are representative of usual clinical practice.

  • Directness: Variable. Studies measure observed and reported side effects of clozapine, as well as treatment discontinuation (all cause and due to adverse effects).

  • Consistency: Consistent. Study findings are consistent in the relative magnitude and direction of effects for specific side effects and for treatment discontinuation.

  • Precision: Precise. Confidence intervals are narrow and do not cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed. However, clinical observations suggest that many side effects do increase in occurrence or severity with the dose of clozapine.

  • Confounding factors: Unclear. Not all studies assess side effects in a systematic fashion, and patients may be less likely to report some side effects if they are not directly assessed.

  • Publication bias: Not assessed. Nevertheless, publication bias is relatively common in studies of psychopharmacology because of nonpublication of negative studies.

  • Overall strength of research evidence: Low to moderate. Available studies include RCTs and an observational study. Data from several meta-analyses suggest a moderate strength of research evidence for outcomes related to clozapine harms, but the AHRQ review found a low strength of research evidence in a network meta-analysis, and the observational study also has a high risk of bias.

Other Interventions for Treatment-Resistant Schizophrenia

Use of Antipsychotic Medications Other Than Clozapine

The network analysis conducted as part of the AHRQ review (McDonagh et al. 2017) found that treatment-resistant patients had a small benefit with olanzapine over other older SGAs in core illness symptom improvement and negative symptoms, whereas response rates and all-cause treatment discontinuations were not different. Negative symptoms were also significantly reduced with olanzapine as compared with haloperidol (N = 2,207; MD 1.28, 95% CI 0.11–2.44), and patients treated with ziprasidone showed better response than those treated with haloperidol (N = 120; RR 1.54, 95% CI 1.19–2.00).

Electroconvulsive Therapy

Some studies have shown evidence for benefits of electroconvulsive therapy (ECT) in combination with antipsychotic medications. Pompili et al. (2013) conducted a systematic review that included RCTs and observational studies, including case-control studies, and concluded that ECT in combination with antipsychotic medications may be helpful for a subgroup of individuals who have treatment resistance, catatonia, aggression, or suicidal behavior, particularly when rapid improvement is needed.

Zheng et al. (2016) conducted a systematic review and meta-analysis of RCTs comparing antipsychotic medications other than clozapine with antipsychotic medication in combination with ECT in patients with treatment-resistant schizophrenia. In the 11 studies, which included 818 patients, the addition of ECT was associated with greater improvements in symptoms (SMD –0.67; P < 0.00001) and greater rates of study-defined response (RR 1.48; P < 0.0001; NNT = 6) and remission (RR 2.18; P = 0.0002; NNT = 8) as well as greater rates of headache (P = 0.02; NNH = 6) and memory impairment (P = 0.001; NNH = 3).

In terms of ECT augmentation of clozapine in treatment-resistant schizophrenia, Petrides et al. (2015) conducted a randomized, single-blind, 8-week trial in which patients who had not responded to clozapine alone received a constant dose of clozapine or clozapine plus bilateral ECT (three times per week for 4 weeks and then twice weekly for 4 weeks for 20 total treatments). Fifty percent of the 20 patients treated with ECT plus clozapine experienced a reduction in psychotic symptoms of at least 40% and also achieved a CGI improvement rating of “much improved” and a CGI severity rating of “borderline mentally ill” or “not at all ill.” This contrasts with 19 patients who received clozapine but not ECT in the randomized phase of the trial, none of whom showed response by these criteria. When the latter group of patients received ECT in the unblinded crossover phase of the trial, the rate of response was 47%. Global cognitive outcomes did not differ for the two randomized groups. In contrast, in another randomized trial of 23 patients who received 12 sessions of ECT as compared with sham ECT, no differences were found in PANSS score reductions, although both groups showed improvement during the study (Melzer-Ribeiro et al. 2017).

Lally et al. (2016b) conducted a systematic review and found 5 trials (4 open-label studies plus the study by Petrides and colleagues, with a total of 71 subjects) in which the pooled response rate to clozapine plus ECT was 54%. When cohort studies, nonblinded randomized trials, case series, and case reports were considered, the overall response rate for clozapine plus ECT was 76% (83 of 126 patients), even though clozapine doses and serum levels were relatively high (mean serum clozapine level of 772.6 ng/mL at a mean daily dose of 506.9 mg for the 52 patients with an available clozapine level; mean daily dose 412.3 mg for the sample as a whole).

G. Wang et al. (2018) conducted a systematic review and meta-analysis of RCTs of ECT augmentation of clozapine for clozapine-resistant schizophrenia that included Chinese and non-Chinese studies. Findings from 18 RCTs that included 1,769 subjects showed benefits of adjunctive ECT compared with clozapine alone for symptomatic improvement at post-ECT and endpoint assessments (SMD –0.88, P = 0.0001 and SMD –1.44, P < 0.00001, respectively). Significant benefits of adjunctive ECT were also seen in study-defined response rates and in remission rates at both assessments (P < 0.00001, NNT = 3 and NNT = 4, respectively, for response and P 0.0001, NNT = 13 and NNT = 14, respectively, for remission); however, subjective memory issues and headache were more frequent in the group that received adjunctive ECT (P < 0.0001, NNH = 4 and P = 0.005, NNH  = 8, respectively).

These studies and meta-analyses suggest a beneficial effect of ECT in combination with antipsychotic medication in individuals with treatment-resistant schizophrenia and clozapine-resistant schizophrenia despite the small number of studies and low quality of observational trials. The increases in reported rates of headache and memory impairment, however, suggest a need to weigh the potential benefits and risks of ECT for the individual patient as compared with the risks of treatment-resistant schizophrenia.

Transcranial Magnetic Stimulation in Treatment-Resistant Schizophrenia

Studies have also been done with transcranial magnetic stimulation (TMS) for treatment of hallucinations and for treatment of negative symptoms in individuals with schizophrenia. He et al. (2017) conducted a meta-analysis of studies published in English or Chinese that studied low (1 Hz) or high (10 Hz) frequency TMS in individuals with schizophrenia. In 13 studies of 1 Hz TMS, auditory hallucinations showed greater improvement with active TMS as compared with sham treatment, but publication bias was noted, and sensitivity analysis also indicated that the meta-analytic finding was unstable and likely to change with additional research. In 7 studies of 10 Hz TMS, there was no effect of active treatment on negative symptoms as compared with sham TMS.

Aleman et al. (2018) conducted a meta-analysis of studies of TMS applied to the dorsolateral prefrontal cortex as compared with sham TMS for treatment of negative symptoms and found a mean weighted effect size of 0.64 (0.32–0.96, total N = 827); however, sham TMS showed a significant improvement of negative symptoms from baseline to posttreatment, with a mean weighted effect size of 0.31 (0.09–0.52, total N = 333). Interpretation of the findings was also complicated by the use of several different coil placements (i.e., right, left, bilateral) and variability in other stimulation parameters (e.g., frequency, intensity, number of stimuli per session, duration of treatment). A meta-analysis by Dollfus et al. (2016) of 13 parallel design trials of TMS for treatment of auditory hallucinations in schizophrenia also showed a significant placebo effect, which was greatest with the 45° position coil and was viewed as introducing substantial bias in determining TMS efficacy.

In terms of addition of TMS to clozapine, Wagner et al. (2019b) used data from the TMS for the Treatment of Negative Symptoms in Schizophrenia (RESIS) trial and examined a subgroup of patients who received treatment with clozapine with the addition of active (N = 12) or sham (N = 14) TMS applied to the left dorsolateral prefrontal cortex for 3 weeks, with five treatment sessions per week. There was no effect of active TMS on negative symptoms, although there was significant benefit of TMS on secondary outcomes (i.e., PANSS positive symptom and general subscales; total PANSS).

These findings on benefits of TMS may change with further research using larger samples and rigorous study designs; however, at present, there is limited evidence for benefits of TMS in reducing either auditory hallucinations or negative symptoms, and findings are confounded by significant placebo effects and publication biases.

Statement 8: Clozapine in Suicide Risk

APA recommends (1B) that patients with schizophrenia be treated with clozapine if the risk for suicide attempts or suicide remains substantial despite other treatments.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

For individuals with schizophrenia who are at substantial risk for suicide, evidence on the use of clozapine comes from retrospective cohort studies and a large pragmatic, open-label RCT (N = 980). Consequently, the strength of research evidence is rated as moderate.

On the basis of findings from the InterSePT study (Meltzer et al. 2003), the AHRQ report (McDonagh et al. 2017) concluded that clozapine was superior to olanzapine in preventing significant suicide attempts or hospitalization to prevent suicide in high-risk patients (HR 0.76, 95% CI 0.58–0.97; moderate SOE). Fewer clozapine-treated patients in the InterSePT study attempted suicide (P = 0.03); required hospitalizations (P = 0.05) or rescue interventions (P = 0.01) to prevent suicide; or required concomitant treatment with antidepressants (P =  0.01) or with anxiolytics or soporifics (P = 0.03). Although there was not a significant difference in suicide deaths (5 for clozapine and 3 for olanzapine), Kaplan-Meier life table estimates indicated a significant reduction in the 2-year event rate in the clozapine group (P = 0.02), with an NNT of 12. Data from other RCTs, in which suicide-related outcomes were reported as adverse events, showed very low event rates and no differences among antipsychotic medications.

One large retrospective study (Kiviniemi et al. 2013) used a nationwide registry to follow up with patients presenting with a first episode of schizophrenia (N = 6,987). At 5 years, the risk of suicide in patients treated with clozapine was significantly reduced (OR 0.29, 95% CI 0.14–0.63), whereas suicide risk in those treated with risperidone, olanzapine, or quetiapine was comparable to the risk with no antipsychotic treatment. Another large nationwide study (N = 9,567) of patients newly starting treatment with SGAs found lower rates of suicide attempts in those beginning with clozapine as compared with other drugs studied (Bitter et al. 2013). Suicide attempt rates were 1.1% at 1 year in those treated with clozapine in contrast to suicide attempt rates that ranged from 2.1% to 3.7% for other SGAs at 1 year. The suicide attempt rate with clozapine treatment was also reduced as compared with the 6 months prior to clozapine initiation (2.2% prior to clozapine as compared with 1.1% after clozapine initiation).

For a discussion of the evidence related to the side effects of clozapine, see Statement 7 earlier in the appendix.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Clozapine in Individuals With Substantial Risk Factors for Suicide Attempts or Suicide

  • Magnitude of effect: Moderate to large. In the randomized controlled InterSePT study, moderate effects are present for clozapine as compared with olanzapine in reducing suicide attempts and hospitalizations to prevent suicide. As compared with other antipsychotic medications, larger effects of clozapine on suicide attempts and suicide are found in observational registry studies with longer periods of follow-up and larger sample sizes.

  • Risk of bias: Medium. Studies include an RCT and observational studies. There is low risk of bias in the RCT on most outcomes but a medium to high risk of bias for the observational studies because of their lack of randomization, lack of blinding, and retrospective study design.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. Doses of clozapine used in the RCT are representative of usual clinical practice. In addition, the RCT includes individuals with an increased risk of suicide, whereas the observational studies assessed suicide-related outcomes without preselecting for high-risk individuals. Nevertheless, rates of suicide are increased among individuals with schizophrenia, making the observational study findings of relevance to routine clinical practice.

  • Directness: Variable. In the RCT, studies measured suicide attempts and deaths due to suicide, but mortality was infrequent, making statistical comparisons invalid. For the observational studies, suicide attempts and deaths from suicide were also studied. Nevertheless, observational study findings are indirect because of the lack of selection of patients at high risk of suicide.

  • Consistency: Consistent. Reductions in suicide attempts are consistent in the RCT and in observational studies. The reduction in suicide deaths in larger samples with longer follow-up periods is consistent with the reduction in suicide attempts.

  • Precision: Precise. Confidence intervals are narrow and do not cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Present. In the RCT, effects on suicide deaths may be reduced by the need to intervene with increased monitoring, hospitalization, or study withdrawal if suicidal risk is significant. Additional monitoring and an increased frequency of clinical contacts with clozapine may enhance the effects of the medication relative to other antipsychotic medications, at least in observational studies.

  • Publication bias: Unable to be assessed. The small number of relevant studies makes assessment of publication bias impossible.

  • Overall strength of research evidence: Moderate. In terms of clozapine effects on suicidal behaviors and suicide, available studies include an RCT and several observational studies with large samples and long periods of follow-up. Confidence intervals are relatively narrow, and the findings are consistent.

Grading of the Overall Supporting Body of Research Evidence for Harms of Clozapine in Individuals With Substantial Risk Factors for Suicide Attempts or Suicide

See Statement 7, subsection “Grading of the Overall Supporting Body of Research Evidence for Harms of Clozapine,” earlier in the appendix.

Statement 9: Clozapine in Aggressive Behavior

APA suggests (2C) that patients with schizophrenia be treated with clozapine if the risk for aggressive behavior remains substantial despite other treatments.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence for the use of clozapine for individuals with substantial aggressive behavior is limited, and the strength of research evidence is rated as low. A systematic review on pharmacological management of persistent hostility and aggression in persons with schizophrenia spectrum disorders found 92 articles with sufficient methodological information to evaluate, although none were at low risk of bias (Victoroff et al. 2014). The authors found two studies (1 RCT, N = 157; 1 open-label, N = 44) showing that in inpatients with schizophrenia spectrum disorders, clozapine was superior to haloperidol in reducing scores on the Overt Aggression Scale (Conley et al. 2003; Ratey et al. 1993). Another RCT conducted in physically assaultive inpatients (N = 100) also found clozapine to be superior to haloperidol or olanzapine in reducing scores on the Overt Aggression Scale (Krakowski et al. 2006, 2008). In reducing hostility (as measured by PANSS or BPRS hostility items), 4 RCTs (3 in inpatients, 1 in outpatients) reported superiority of clozapine as compared with FGAs. Two of these studies (N = 48 and N = 151) compared clozapine with chlorpromazine (Claghorn et al. 1987; Niskanen et al. 1974), and the other 2 studies (N = 167 and N = 71) compared clozapine with haloperidol (Citrome et al. 2001; Kane et al. 2001).

These findings support the opinions of many experts in viewing clozapine as beneficial in patients at substantial risk of aggressive behaviors. Nevertheless, additional evidence from well-designed clinical trials is needed. For a discussion of the evidence related to the side effects of clozapine, see Statement 7 earlier in the appendix.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Clozapine in Individuals With Substantial Risk Factors for Aggressive Behaviors

  • Magnitude of effect: Unclear. Available studies report statistical superiority, but there are no good estimates of the magnitude effect either within or among studies.

  • Risk of bias: High. Most of the available studies, including RCTs and open-label studies, have a significant risk of bias and a lack of reported details about randomization, blinding, and other features of study design.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. Most studies are focused on inpatients, including forensic psychiatry populations, who exhibit physically assaultive behavior. The doses of medication used are within normal to high dose ranges for usual clinical practice.

  • Directness: Variable. Studies measure multiple different outcomes, including hostility items on PANSS or BPRS, time in restraint, episodes of restraint, and episodes of assaultive behavior.

  • Consistency: Consistent. Studies generally report reductions in hostility or aggressive behavior.

  • Precision: Unknown. Confidence intervals are not reported in all studies or in the available meta-analysis. Nevertheless, a lack of precision is likely due to the small samples in most studies.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Present. In observational outpatient studies, additional monitoring and an increased frequency of clinical contacts with clozapine may enhance medication effects relative to other antipsychotic medications. The high risk of bias in many of these studies suggests that confounding factors may be present but unrecognized.

  • Publication bias: Unable to be assessed. The relatively small number of studies and the heterogeneity of study designs make it difficult to assess publication bias. However, publication bias seems possible because of the tendency for negative clinical trial results to go unpublished.

  • Overall strength of research evidence: Low. The available studies include RCTs and open-label studies with a high risk of bias. Although the findings are consistent, the applicability to typical clinical practice is limited. Other sources of possible bias were unable to be assessed but are likely to be present.

Grading of the Overall Supporting Body of Research Evidence for Harms of Clozapine in Individuals With Substantial Risk Factors for Aggressive Behaviors

See Statement 7, subsection “Grading of the Overall Supporting Body of Research Evidence for Harms of Clozapine,” earlier in the appendix.

Statement 10: Long-Acting Injectable Antipsychotic Medications

APA suggests (2B) that patients receive treatment with a long-acting injectable antipsychotic medication if they prefer such treatment or if they have a history of poor or uncertain adherence.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence for this guideline statement comes from the AHRQ review (McDonagh et al. 2017) as well as from other RCTs, registry database studies, cohort studies, “mirror image” studies, and meta-analyses of such trials. The findings from these studies are mixed because RCTs show few differences in outcomes between LAI antipsychotic medications and oral antipsychotic agents, whereas observational trials show consistent benefits of LAI formulations. There are a number of possible explanations for these apparent disparities related to the design of the studies and differences in study populations (Correll et al. 2016; Fagiolini et al. 2017). Individuals who are agreeable to participating in a randomized clinical trial are more likely to be adherent to treatment than a broader population of individuals with a particular diagnosis. A greater focus on adherence-related questions and a greater frequency of visits may occur in an RCT as compared with treatment as usual, which may also influence adherence or outcomes. Consequently, the possible advantages of LAIs over oral formulations in promoting or assuring adherence may be less salient in RCTs as compared with observational trials. Although each type of study has advantages and disadvantages, observational trials that use registry databases are also able to examine outcomes among large numbers of individuals over many years of follow-up in contrast to the smaller numbers and shorter follow-up periods of RCTs.

In the AHRQ review (McDonagh et al. 2017), the ability to draw conclusions about the comparative effectiveness of LAI antipsychotic medications is limited by the relatively small number of head-to-head comparison studies among LAI antipsychotic medications or for LAI formulations as compared with oral agents. Few studies assessed differences in symptoms with treatment, but clozapine was noted to be superior to aripiprazole LAI (monthly or every 6 weeks), olanzapine LAI, paliperidone LAI (monthly and every 3 months), and risperidone LAI, whereas paliperidone LAI every 3 months was superior to oral lurasidone (low SOE). For the few comparisons where data on response were available, no differences were found (low SOE). Risperidone LAI was significantly better than quetiapine in social function over 24 months but did not differ from quetiapine on measures of quality of life (low SOE). No difference in social function was found between monthly paliperidone palmitate LAI and biweekly risperidone LAI (low SOE). In terms of quality of life, oral aripiprazole and monthly aripiprazole LAI did not differ from one another with up to 2 years of follow-up (low SOE).

In terms of findings on harms, a network meta-analysis of 90 head-to-head RCTs showed that risperidone LAI had a significantly lower risk of withdrawal due to adverse events than clozapine (OR 0.27, 95% CI 0.10–0.71), lurasidone (OR 0.39, 95% CI 0.18–0.84), quetiapine XR (OR 0.43, 95% CI 0.22–0.81), risperidone (OR 0.50, 95% CI 0.25–0.99), or ziprasidone (OR 0.40, 95% CI 0.20–0.82). No differences in overall adverse events were found between aripiprazole as compared with monthly aripiprazole LAI or between paliperidone or monthly paliperidone palmitate LAI as compared with risperidone LAI (low SOE). In addition, no differences in extrapyramidal side effects were seen in a 28-week trial of aripiprazole and monthly paliperidone LAI or a network meta-analysis comparing monthly and 4- to 6-week aripiprazole LAI. However, monthly aripiprazole LAI had a greater incidence of extrapyramidal side effects (RR 1.88) and worse akathisia than oral aripiprazole in the short term but not at 1 year. For mortality, no significant difference was found between monthly paliperidone palmitate LAI versus risperidone LAI (RR 1.26, 95% CI 0.21–7.49) on the basis of 2 RCTs of 4–24 months’ duration (low SOE).

A number of other meta-analyses of RCTs are also available and provide complementary information to the findings in the AHRQ review. Ostuzzi et al. (2017) conducted a systematic review and meta-analysis of RCTs that compared the oral and LAI formulations of the same antipsychotic medication and included risperidone (6 studies), olanzapine (2 studies), aripiprazole (3 studies), zuclopenthixol (1 study), fluphenazine (7 studies), and haloperidol (2 studies). There was a small benefit for aripiprazole LAI (2 studies, N = 986; RR 0.78, 95% CI 0.64–0.95; high SOE) as compared with oral aripiprazole for all-cause discontinuation, a small benefit for oral olanzapine as compared with olanzapine LAI for discontinuation due to inefficacy (2 studies, N = 1,445; RR 1.52, 95% CI 1.12–2.07; low SOE), and a small benefit for risperidone LAI as compared with oral risperidone for hyperprolactinemia (5 studies, N = 891; RR 0.81, 95% CI 0.68–0.98; moderate SOE). The other comparisons showed no differences for these outcomes, and there were also no differences noted for nonresponse rate, relapse rate, dropouts for adverse events, extrapyramidal symptoms, or weight gain.

Kishimoto et al. (2014) examined the relative efficacy of LAI antipsychotic medications as compared with oral antipsychotic medications in relapse prevention and found that LAIs were similar to oral agents in outpatient studies lasting  1 year (12 studies; RR 0.93, 95% CI 0.71–1.07; P = 0.31) and at the longest study time point across all settings (21 studies, N = 4,950; RR 0.93, 95% CI 0.80–1.08; P = 0.35). When analyzed by drug, fluphenazine LAI showed greater benefit than oral antipsychotic agents in preventing relapse (RR 0.79, 95% CI 0.65–0.96; P = 0.02); however, the authors note that this may be mediated by a cohort effect rather than a drug-specific effect because all of the fluphenazine studies predated 1992. For drug inefficacy, calculated as the sum of relapses plus discontinuations due to inefficacy, fluphenazine LAI was again superior to oral antipsychotic agents (8 studies, N = 826; RR 0.78, 95% CI 0.66–0.91; P = 0.002), whereas olanzapine LAI was inferior to oral antipsychotic agents (N = 1,445; RR 1.52, 95% CI 1.12–2.07; P = 0.007). In preventing hospitalization, fluphenazine LAI was also superior to oral antipsychotic agents (4 studies, N = 197; RR 0.82, 95% CI 0.67–0.99, P = 0.04); however, pooled data for LAIs as compared with oral antipsychotic agents showed no statistically significant effects (10 studies, N = 2,296; RR 0.88, 95% CI 0.75–1.03; P = 0.09). No differences in all-cause discontinuation were noted in pooled analyses or for individual LAIs (fluphenazine, haloperidol, zuclopenthixol, risperidone, olanzapine).

A separate meta-analysis (Kishi et al. 2016b) examined the efficacy of paliperidone LAI or risperidone LAI as compared with oral antipsychotic agents in patients with a recent-onset psychotic disorder. Although there was significant heterogeneity in the study findings, LAIs and oral agents were comparable overall in relapse prevention (3 studies; N = 875). The LAIs did have fewer study discontinuations because of inefficacy (RR 0.34, NNT = –50) or nonadherence (RR 0.30, NNT = –33), but LAIs also had a higher incidence of tremor (RR 2.38) or at least one adverse effect (RR 1.13). In terms of mortality with LAIs as compared with oral antipsychotic agents, another meta-analysis of 52 RCTs (Kishi et al. 2016a) found no difference between LAIs and placebo or oral antipsychotics in all-cause death or death due to suicide (total N = 17,416; LAI antipsychotics = 11,360, oral antipsychotics = 3,910, and placebo = 2,146; LAI antipsychotics vs. placebo = 28.9, LAI antipsychotics vs. oral antipsychotics = 64.5).

Another RCT, the Preventing Relapse Oral Antipsychotics Compared to Injectables Evaluating Efficacy (PROACTIVE) study (P. F. Buckley et al. 2015), was a multisite trial conducted at 8 academic centers in the United States. The researchers randomly assigned patients with schizophrenia or schizoaffective disorder to risperidone LAI or the physician’s choice of an oral SGA for up to 30 months. Subjects were outpatients who were neither resistant to treatment nor experiencing a first episode of psychosis. Approximately half of the subjects (161 of 305) discontinued treatment before the end of the trial. There was no significant difference noted in the proportion with a relapse (42% for risperidone LAI vs. 32% for oral SGA; P = 0.08), time to first relapse (P = 0.13), or time to first hospitalization (P = 0.30). In addition, no significant differences between risperidone LAI and oral SGAs were noted for the bulk of symptom ratings (anxiety-depression, negative symptoms, excitement, affective flattening, avolition, asociality-anhedonia, CGI severity and CGI improvement). However, changes in symptom scores did differ between the treatment arms, with lower Scale for the Assessment of Negative Symptoms (SANS) alogia scores with oral SGAs and greater improvement in psychotic symptoms and BRPS total scores with risperidone LAI. In patients followed after an initial relapse, 32 (11%) had two relapses, and 13 (4%) had three relapses, with no significant differences in the rate or time to successive relapse between those treated with risperidone LAI and those treated with oral SGAs (Buckley et al. 2016).

In contrast to the findings from RCTs, observational studies often find benefits of LAI antipsychotic formulations as compared with oral antipsychotic formulations. Tiihonen et al. (2017) used a prospective national database in Sweden (with individuals as their own controls) to examine the risk of treatment failure, which was defined as psychiatric rehospitalization, admissions due to a suicide attempt, discontinuation of antipsychotic medication or switch to another antipsychotic medication, or death. Of the 29,823 patients, 43.7% were rehospitalized, and 71.7% met criteria for treatment failure. The LAI formulations of antipsychotic medications were associated with a 20%–30% lower risk of rehospitalization as compared with oral formulations of an antipsychotic (HR 0.78, 95% CI 0.72–0.84 for the total cohort; HR 0.68, 95% CI 0.53–0.86 for the incident cohort). For specific LAI antipsychotic medications as compared with no use of antipsychotic medication, rehospitalization risk was lowest with once-monthly paliperidone LAI (HR 0.51, 95% CI 0.41–0.64), zuclopenthixol LAI (HR 0.53, 95% CI 0.48–0.57), perphenazine LAI (HR 0.58, 95% CI 0.52–0.65), and olanzapine LAI (HR 0.58, 95% CI 0.44–0.77). Of the oral medications, rehospitalization rates were lowest with clozapine (HR 0.53, 95% CI 0.48–0.58). Rates of treatment failure were also lowest with clozapine (HR 0.58, 95% CI 0.53–0.63) and with LAI antipsychotic formulations as compared with other oral formulations (HR values for LAI formulations: perphenazine LAI 0.65, haloperidol LAI 0.67, zuclopenthixol LAI 0.69, paliperidone LAI 0.72, flupentixol LAI 0.75, olanzapine LAI 0.77, and risperidone LAI 0.80).

Tiihonen et al. (2011) also compared LAI antipsychotics with their equivalent oral formulation in a nationwide cohort of 2,588 consecutive patients in Finland who had an initial admission with a diagnosis of schizophrenia. Of those individuals, only 58.2% used an antipsychotic medication after discharge, and 45.7% of the cohort continued to take an antipsychotic for at least 30 days. For rehospitalization as well as for all-cause discontinuation, LAI antipsychotic had a lower adjusted hazard ratio (aHR) than the equivalent oral formulation (aHR 0.36, 95% CI 0.17–0.75; P = 0.007 and aHR 0.41, 95% CI 0.27–0.61; P = <0.0001, respectively). For each LAI antipsychotic as compared with its oral equivalent, rehospitalization was lower with haloperidol LAI (aHR 0.12, 95% CI 0.01–1.13; P = 0.06) but not perphenazine LAI (aHR 0.53, 95% CI 0.22–1.28; P = 0.16) or risperidone LAI (aHR 0.57, 95% CI 0.30–1.08; P = 0.09). Use of an LAI antipsychotic was also associated with lower rates of all-cause discontinuation for haloperidol LAI, perphenazine LAI, and risperidone LAI as compared with their oral equivalents (aHR 0.27, 95% CI 0.08–0.88; P = 0.03; aHR 0.32, 95% CI 0.19–0.53; P = <0.0001; and aHR 0.44, 95% CI 0.31–0.62; P = <0.0001, respectively). Zuclopenthixol LAI showed no difference from its equivalent oral formulation in either rehospitalization or all-cause discontinuation.

Taipale et al. (2018a) used the same nationwide Finnish health care registry to assess the long-term effectiveness of antipsychotic medications on the risk of psychiatry rehospitalization over follow-up periods of up to 20 years (median of 14.1 years). The sample included a prevalence cohort of 62,250 individuals as well as 8,719 individuals who were followed prospectively after a first episode of psychosis. The risk of psychiatric rehospitalization was lower with LAI antipsychotic medications than with oral antipsychotic formulations (LAI FGAs HR 0.46, 95% CI 0.40–0.54; LAI SGAs HR 0.45, 95% CI 0.39–0.52; oral FGAs HR 0.67, 95% CI 0.60–0.74; oral SGAs HR 0.57, 95% CI 0.53–0.61) in first-episode patients, as was the risk of all-cause hospitalization (LAI FGAs HR 0.58, 95% CI 0.51–0.66; LAI SGAs HR 0.56, 95% CI 0.50–0.63; oral FGAs HR 0.80, 95% CI 0.74–0.87; oral SGAs HR 0.69, 95% CI 0.66–0.73), with similar patterns noted in the prevalence cohort.

A nationwide registry was also used by Taipale et al. (2018b) to examine all-cause mortality and its relationship to medication treatment among patients with schizophrenia in Sweden. Information was available on 29,823 individuals between 2006 and 2013, of which 4,603 patients were in the incident cohort. For LAI SGAs, the cumulative mortality rate was about one-third lower than the mortality rate for equivalent oral antipsychotics in pairwise analyses (aHRs 0.67, 95% CI 0.56–0.80). Those taking an LAI formulation of an SGA had the lowest cumulative mortality (7.5%), with median follow-up of 6.9 years. Corresponding rates of cumulative mortality were 8.5% for oral SGAs, 12.2% for oral FGAs, 12.3% for LAI FGAs, and 15.2% in those who were not taking an antipsychotic medication. As compared with LAI SGAs, corresponding aHRs were 1.52 (95% CI 1.13–2.05) for oral SGAs, 1.37 (95% CI 1.01–1.86) for LAI FGAs, 1.83 (95% CI 1.33–2.50) for oral FGAs, and 3.39 (95% CI 2.53–4.56) in those who were not taking an antipsychotic medication.

MacEwan et al. (2016b) used a multistate database of U.S. Medicaid patients to examine the probability of rehospitalization after an index admission with LAI antipsychotic treatment as compared with oral antipsychotic treatment. Using multivariate logistic regression with propensity score matching for 1,450 patients with a diagnosis of schizophrenia, an LAI antipsychotic medication was associated with a lower probability of readmission at 60 days postdischarge (adjusted OR 0.60, 95% CI 0.41–0.90) but not at 30 days postdischarge.

Kishimoto and colleagues conducted meta-analyses of cohort studies and mirror image studies to compare the effectiveness of LAIs versus oral antipsychotic agents in terms of hospitalization and treatment discontinuation (Kishimoto et al. 2013, 2018). On the basis of 42 prospective and retrospective cohort studies (total N = 101,624; mean follow-up 18.6 ± 10.0 months), LAIs were found to be superior to oral antipsychotics in terms of all-cause discontinuations (10 studies, N =  37,293; risk ratio 0.78, 95% CI 0.67–0.91; P = 0.001) and hospitalization rates (15 studies, 68,009 person-years; rate ratio 0.85, 95% CI 0.78–0.93; P < 0.001) but not hospitalization risk or days of hospitalization (Kishimoto et al. 2018). However, the patients treated with an LAI antipsychotic medication had longer illness durations than those treated with oral formulations of antipsychotic medication, which may have influenced the findings. In 25 mirror image studies that followed patients for at least 6 months before and after a transition between medication formulations, LAI antipsychotic medications were superior to oral antipsychotic medications in preventing hospitalization (16 studies, N = 4,066; risk ratio 0.43, 95% CI 0.35–0.53; P < 0.001) and in decreasing the number of hospitalizations (15 studies, 6,342 person-years; rate ratio 0.38, 95% CI 0.28–0.51; P < 0.001) (Kishimoto et al. 2013).

Grading of the Overall Supporting Body of Research Evidence for the Efficacy of LAI Antipsychotic Medications

  • Magnitude of effect: Variable. In RCTs, there are few differences in outcomes between LAI and oral formulations of antipsychotic medications. However, significant benefits with a moderate magnitude of effect are noted in observational studies, including prospective registry database studies and mirror image studies.

  • Risk of bias: Medium. Studies include RCTs that have some limitations in study design or reporting of features such as randomization or blinding. Observational studies based on prospective registry data are well designed but have at least a medium risk of bias because of a lack of randomization or blinding.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with schizoaffective disorder. The doses of medication used are not always stated but appear to be representative of usual clinical practice. The observational studies include data from a nationwide registry and have broad generalizability, in contrast to RCTs with more restrictive inclusion and exclusion criteria. However, the applicability of registry data from Nordic countries may be reduced by differences in the health care delivery system as compared with that of the United States.

  • Directness: Variable. Most studies measure direct outcomes, including differences in symptoms, quality of life, functioning, relapse prevention, and rehospitalization. However, some studies assess indirect outcomes, including all-cause treatment discontinuation.

  • Consistency: Inconsistent. RCTs generally show little or no benefit of LAI as compared with oral formulations of antipsychotic medications, whereas observational studies show moderate benefits. However, findings are consistent for different types of observational studies, including prospective registry database studies and mirror image analyses.

  • Precision: Imprecise. For RCTs, confidence intervals cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Unclear. Confounding factors may be present for the observational studies because of the lack of randomization. Individuals with poor adherence or more severe symptoms may be more likely to receive an LAI, which would give LAI-treated patients a greater risk of relapse or rehospitalization.

  • Publication bias: Not suspected. Publication bias was not detected in the meta-analyses that specifically examined this question.

  • Overall strength of research evidence: Moderate. Available evidence includes data from several types of observational studies, each of which shows consistent benefits for LAI as compared with oral formulations of antipsychotic medication. The potential benefit of LAI formulations in assuring adherence may not be observable in RCTs in which patients are already selected for high adherence. Although trials are of varying quality, most have good sample sizes. When beneficial effects are noted, most confidence intervals are narrow. There is some variation from drug to drug, but registry data show better outcomes with LAI formulations as a group as compared with oral formulations of antipsychotic medication as a group.

Grading of the Overall Supporting Body of Research Evidence for the Harms of LAI Antipsychotic Medications

  • Magnitude of effect: Variable. In general, there appear to be few differences between harms of LAI antipsychotic medications and oral formulations of antipsychotic medications, particularly when LAI and oral formulations of the same drug are compared. When differences are noted in rates of specific side effects, the magnitude of those effects is small.

  • Risk of bias: Medium. In RCTs, some limitations in study design are present. In other studies, harms of treatment were not systematically assessed.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. The doses of medication used are not always stated but appear to be representative of usual clinical practice. The observational studies include data from a nationwide registry and have broad generalizability, in contrast to RCTs with more restrictive inclusion and exclusion criteria. However, the applicability of registry data from Nordic countries may be reduced by differences in the health care delivery system as compared with that of the United States.

  • Directness: Variable. When assessments of adverse effects are conducted, studies measure specific side effects. However, other studies measure study withdrawals due to adverse effects.

  • Consistency: Inconsistent. Some comparisons show differences between LAI and oral formulations on specific side effects, but these are not consistent among medications or meta-analyses.

  • Precision: Imprecise. Confidence intervals cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed. Data from studies of oral medications suggest that increases in dose are likely to be associated with increases in medication side effects.

  • Confounding factors: Unclear. Adverse effects are not always assessed in a systematic fashion, and reporting biases may be present.

  • Publication bias: Not suspected. Publication bias was not detected in the meta-analyses that specifically examined this question.

  • Overall strength of research evidence: Low. Available studies include RCTs that assess side effects of LAI and oral formulations of antipsychotic medications. Meta-analyses and network meta-analyses are also available that include head-to-head comparison trials. In terms of ascertainment and reporting of information on side effects, studies have at least a medium risk of bias, and there is significant inconsistency in the findings among the available studies, making it difficult to draw conclusions with any degree of confidence.

Statement 11: Anticholinergic Medications for Acute Dystonia

APA recommends (1C) that patients who have acute dystonia associated with antipsychotic therapy be treated with an anticholinergic medication.

This recommendation is based on expert opinion and is supported by studies of the prophylactic use of anticholinergic medications to reduce the risk of acute dystonia in the initial phases of antipsychotic therapy. The strength of research evidence for this guideline statement is rated as low. No studies were found that specifically examined the treatment of acute dystonia with anticholinergic medications in a randomized or controlled manner, although intramuscular administration of an anticholinergic agent is widely viewed as the treatment of choice for acute dystonia associated with antipsychotic therapy (Stanilla and Simpson 2017).

Information on the use of anticholinergic medications to prevent acute dystonia associated with antipsychotic therapy comes from a review of 9 studies (Arana et al. 1988), of which 4 were randomized, blinded trials (total N = 232); 2 were open trials (total N = 856); and 3 were retrospective studies (total N = 278). On the basis of data from all of these studies, prophylactic use of an anticholinergic medication was associated with 1.9-fold reduction in risk of acute dystonia (14.8% without prophylaxis vs. 7.7% with prophylaxis). In patients who received a high-potency antipsychotic agent (e.g., haloperidol), the benefits of prophylactic anticholinergic medication were even more pronounced (5.4-fold reduction in risk; 46.8% without prophylaxis vs. 8.7% with prophylaxis).

A subsequent study of consecutive psychiatric admissions (N = 646) showed a lower rate of acute dystonia in patients who received anticholinergic prophylaxis (8.5% without anticholinergic prophylaxis vs. 2.8% with anticholinergic prophylaxis), and rates of acute dystonia were greater in individuals treated with a high-potency antipsychotic agent (Spina et al. 1993). A small double-blind RCT (N = 29) showed a decrease in acute dystonia associated with antipsychotic therapy in patients who received benztropine as compared with placebo, but the results did not reach statistical significance (Goff et al. 1991). These studies suggest therapeutic effects of anticholinergic medications in acute dystonia associated with antipsychotic therapy, and although the studies were conducted in patients who received FGAs, they likely would also apply to acute dystonia when it occurs with use of SGAs.

Grading of the Overall Supporting Body of Research Evidence for Anticholinergic Medications for Acute Dystonia

On the basis of the limitations of the evidence for anticholinergic medications for acute dystonia, no grading of the body of research evidence is possible.

Statement 12: Treatments for Parkinsonism

APA suggests (2C) the following options for patients who have parkinsonism associated with antipsychotic therapy: lowering the dosage of the antipsychotic medication, switching to another antipsychotic medication, or treating with an anticholinergic medication.

This statement is based on expert opinion, and, consequently, the strength of research evidence is rated as low. Knowledge of pharmacology and pharmacokinetics suggests that side effects such as parkinsonism may be diminished by reducing the dose of a medication or changing to a medication with a different side-effect profile and a lesser propensity for treatment-related parkinsonism. Clinical experience also suggests that an anticholinergic medication can be used to treat antipsychotic-associated parkinsonism (Stanilla and Simpson 2017). A good-quality systematic review assessed the use of anticholinergic medication compared with placebo for parkinsonism associated with antipsychotic therapy (Dickenson et al. 2017). Although many studies of anticholinergic treatment for parkinsonism were conducted decades ago and suggested benefits of anticholinergics, few of these studies met the systematic review’s inclusion criteria. In addition, sample sizes in the two included studies were small, and no definitive conclusions could be drawn from the systematic review.

Grading of the Overall Supporting Body of Research Evidence for Treatments for Parkinsonism

On the basis of the limitations of the evidence for treatments for parkinsonism, no grading of the body of research evidence is possible.

Statement 13: Treatments for Akathisia

APA suggests (2C) the following options for patients who have akathisia associated with antipsychotic therapy: lowering the dosage of the antipsychotic medication, switching to another antipsychotic medication, adding a benzodiazepine medication, or adding a beta-adrenergic blocking agent.

This statement is based on expert opinion, and, consequently, the strength of research evidence is rated as low. Knowledge of pharmacology and pharmacokinetics suggests that side effects such as akathisia may be diminished by reducing the dose of a medication or changing to a medication with a different side-effect profile and a lesser propensity for treatment-related akathisia. A good-quality systematic review identified some benefits of benzodiazepines for akathisia associated with antipsychotic therapy (Lima et al. 2002), but only 2 studies (total N = 27) met the inclusion criteria. Another good-quality systematic review assessed the use of β-adrenergic blocking agents in akathisia and also found insufficient evidence to draw conclusions about therapeutic benefits (3 RCTs, total N = 51; Lima et al. 2004). In addition, no reliable evidence was found to support or refute the use of anticholinergic agents as compared with placebo for akathisia associated with antipsychotic therapy (Rathbone and Soares-Weiser 2006). The literature search did not identify well-designed trials published after these systematic reviews that shed additional light on any of these treatment approaches.

Grading of the Overall Supporting Body of Research Evidence for Treatments for Akathisia

On the basis of the limitations of the evidence for treatments for akathisia, no grading of the body of research evidence is possible.

Statement 14: VMAT2 Medications for Tardive Dyskinesia

APA recommends (1B) that patients who have moderate to severe or disabling tardive dyskinesia associated with antipsychotic therapy be treated with a reversible inhibitor of the vesicular monoamine transporter 2 (VMAT2).

This statement is based on information from a good-quality systematic review (Solmi et al. 2018b) on deutetrabenazine and valbenazine treatment; information on tetrabenazine comes from less robust clinical trials. The strength of research evidence for this guideline statement is rated as moderate.

For deutetrabenazine, data were available from 2 double-blind, placebo-controlled RCTs (Anderson et al. 2017; Fernandez et al. 2017) that enrolled subjects with moderate to severe tardive dyskinesia. Each trial lasted 12 weeks, and the dosage of deutetrabenazine was 12–48 mg/day. Treatment with deutetrabenazine was associated with a significant decrease in total Abnormal Involuntary Movement Scale (AIMS) scores (N = 413; SMD –0.40, 95% CI –0.19 to –0.62, P < 0.001; weighted mean difference [WMD] –1.44, 95% CI –0.67 to –2.19, P < 0.001) and significantly greater rates of response (defined as an AIMS score reduction of at least 50%; RR 2.13, 95% CI 1.10–4.12, P = 0.024; NNT = 7, 95% CI 3–333, P = 0.046) (Solmi et al. 2018b). The rate of treatment response increased with treatment duration during the open-label extension phase of the study (Hauser et al. 2019). Deutetrabenazine was well tolerated, with trial completion rates and rates of adverse effects similar to rates with placebo (Solmi et al. 2018b).

For valbenazine, data were available from 4 double-blind, placebo-controlled trials (total N = 488) of 4–6 weeks each using a valbenazine dosage of 12.5–100 mg/day in individuals with moderate to severe tardive dyskinesia (Citrome 2017b; Correll et al. 2017a; Factor et al. 2017; Hauser et al. 2017; Josiassen et al. 2017; Kane et al. 2017; O’Brien et al. 2015). Treatment with valbenazine was associated with a significant decrease in total AIMS scores (N = 421; SMD –0.58, 95% CI –0.26 to –0.91, P < 0.001; WMD –2.07, 95% CI –1.08 to –3.05, P < 0.001) and significantly greater rates of response (RR 3.05, 95% CI 1.81–5.11, P < 0.001; NNT = 4, 95% CI 3–6, P < 0.001; Solmi et al. 2018b). With valbenazine, as with deutetrabenazine, the rate of treatment response increased with treatment duration during the open-label extension phase of the study (Factor et al. 2017). Furthermore, in the randomized KINECT 3 study, a dose-response relationship was observed, with greater benefit at dosages of 80 mg/day as compared with 40 mg/day (Hauser et al. 2017). Valbenazine was well tolerated, with trial completion rates and rates of adverse effects that were similar to rates with placebo (Solmi et al. 2018b).

For tetrabenazine, prospective placebo-controlled data are more limited and include a single-blind trial of 20 subjects (Ondo et al. 1999), a double-blind crossover trial of 6 subjects (Godwin-Austen and Clark 1971), and another double-blind crossover trial of 24 subjects (Kazamatsuri et al. 1972). Although benefits of tetrabenazine were seen at dosages of up to 150 mg/day, the quality of evidence is not sufficient to draw robust conclusions or conduct meta-analyses (Leung and Breden 2011; Solmi et al. 2018b). Adverse effects that were more frequent with tetrabenazine than placebo included drowsiness, sedation or somnolence, parkinsonism, insomnia, anxiety, depression, and akathisia.

Although this statement specifically relates to the use of a reversible inhibitor of the vesicular monoamine transporter 2 (VMAT2) (e.g., deutetrabenazine, tetrabenazine, valbenazine), the guideline writing group also reviewed evidence for other possible treatments for tardive dyskinesia. Amantadine has been mentioned in the literature as a treatment for tardive dyskinesia, but evidence for its use is extremely limited. One randomized double-blind crossover trial (Angus et al. 1997) included only 16 patients and had significant attrition. Another randomized double-blind crossover trial (Pappa et al. 2010) also had a small sample (N = 22), and the period of treatment was only 2 weeks. Thus, data from these trials are insufficient to support use of amantadine for treatment of tardive dyskinesia. Other studies of treatments for tardive dyskinesia have been discussed in systematic reviews, as summarized in Table C–2. On the basis of these findings, there is insufficient evidence to support a guideline statement on use of these treatments in individuals with tardive dyskinesia.

Other systematic reviews of treatments for tardive dyskinesia

Intervention

Reference

Comments

Anticholinergic agents

Bergman and Soares-Weiser 2018

2 trials (total N = 30) had very low-quality evidence

Benzodiazepines

Bergman et al. 2018a

4 trials (total N = 75) of very low-quality evidence showed no clinically significant difference relative to placebo

Calcium channel blockers

Soares-Weiser and Rathbone 2011

No studies met inclusion criteria

Cessation or reduction of antipsychotic

Bergman et al. 2018b

2 trials (total N = 17) had very low-quality evidence

Change to clozapine

Mentzel et al. 2018

In 4 trials (total N = 48), subjects who had clinically significant tardive dyskinesia showed improvement with a change to clozapine (standardized mean change –2.56, 95% CI –4.85 to –0.28; P = 0.02), which is consistent with observational data (Lieberman et al. 1991; Naber et al. 1989; Pinninti et al. 2015)

Cholinergic medication

Tammenmaa-Aho et al. 2018

Low-quality evidence showed no clinically important improvement in tardive dyskinesia symptoms (4 trials; N = 27) or effect on deterioration of tardive dyskinesia symptoms (8 trials; N = 147) when compared with placebo

Gamma-aminobutyric acid agonists

Alabed et al. 2018

Low-quality evidence showed no clinically important improvement in tardive dyskinesia symptoms (5 trials; N = 36), and a greater rate of side effects and attrition was suggested

Ginkgo biloba extract

Soares-Weiser et al. 2018b

1 trial (N = 157) showed benefit compared with placebo (RR 0.88, 95% CI 0.81–0.96) in a moderate-quality study, but this requires replication

Non-antipsychotic catecholaminergic drugs

El-Sayeh et al. 2018

10 trials (N = 261) showed very low-quality evidence, and there were only 1–2 trials per therapeutic comparison

Vitamin B6 (pyridoxal 5’-phosphate)

Adelufosi et al. 2015

In 3 trials (total N = 80), inpatients followed for 9–26 weeks showed significant improvement in tardive dyskinesia symptoms when compared with placebo, but evidence was of low quality, with wide CIs

Vitamin E

Soares-Weiser et al. 2018a

13 trials (total N = 478) of low quality showed possible blunting of additional deterioration but no clear difference when compared with placebo in terms of clinically important improvement

Miscellaneous agents, including branched-chain amino acids, buspirone, dihydrogenated ergot alkaloids, estrogen, γ-linolenic acid, insulin, isocarboxazid, lithium, melatonin, pemoline, promethazine, ritanserin, and selegiline

Soares-Weiser et al. 2018b

In 1–2 short-term trials (total N = 10–52 for each medication), low- to very low-quality evidence was inconclusive

Abbreviations. CI = confidence interval; RR = relative risk.

Other systematic reviews of treatments for tardive dyskinesia

Enlarge table

Grading of the Overall Supporting Body of Research Evidence for Efficacy of VMAT2 Inhibitors

  • Magnitude of effect: Moderate. Effects are at least moderate in terms of the proportion of individuals who are much improved or very much improved with valbenazine or deutetrabenazine as compared with placebo. Statistically significant differences are also found for rates of response and for decreases in total AIMS scores. Fewer data are available for tetrabenazine, and its magnitude of effect is unclear.

  • Risk of bias: Low to medium. Studies are RCTs with a low to medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with moderate to severe tardive dyskinesia, although some studies include individuals with diagnoses other than schizophrenia. The doses of medication used are representative of usual clinical practice.

  • Directness: Direct. Studies measure changes in signs of tardive dyskinesia on the AIMS and proportions of individuals who showed at least 50% response or who were much improved or very much improved on a global measure of benefit.

  • Consistency: Consistent. Studies are consistent in showing benefits of valbenazine and deutetrabenazine. Studies of tetrabenazine are more limited in number, sample size, and design but also show benefit in individuals with tardive dyskinesia.

  • Precision: Imprecise. Confidence intervals are wide but do not cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Present. Valbenazine and deutetrabenazine show an increase in clinical benefit with an increase in dose.

  • Confounding factors: Unclear. No specific confounding factors are noted for valbenazine or deutetrabenazine, but confounding factors may be present for tetrabenazine because of weaknesses in study methodologies.

  • Publication bias: Unable to be assessed. The small number of available studies precludes assessment of publication bias.

  • Overall strength of research evidence: Moderate. The available studies of valbenazine and deutetrabenazine are of good quality with good sample sizes. However, not all confidence intervals are narrow. In addition, the duration of the randomized phase of the trials was relatively short, as little as 4–6 weeks in some studies. The long-term follow-up data are based only on open-label extension phases of these RCTs. Data on tetrabenazine have a higher risk of bias, smaller samples sizes, and inadequate blinding, yielding a low strength of research evidence.

Grading of the Overall Supporting Body of Research Evidence for Harms of VMAT2 Inhibitors

  • Magnitude of effect: Minimal to small. For deutetrabenazine and valbenazine, there are no significant differences from placebo in rates of adverse effects. For tetrabenazine, some adverse effects are more frequent than with placebo, but the magnitude of the difference is still relatively small.

  • Risk of bias: Medium to high. Studies of valbenazine and deutetrabenazine determine adverse events in a systematic fashion, but the duration of the randomized phase of the clinical trials is relatively short, and the open-label extension phases have a greater risk of bias. Studies of tetrabenazine have a greater number of limitations in study design, with a high risk of bias overall.

  • Applicability: The included trials all involve individuals with moderate to severe tardive dyskinesia, although some studies include individuals with diagnoses other than schizophrenia. The doses of medication used are representative of usual clinical practice.

  • Directness: Direct. Studies measure occurrence of specific side effects.

  • Consistency: Variable. Studies of deutetrabenazine and valbenazine are consistent in showing negligible side effects as compared with placebo. Studies of tetrabenazine generally show greater rates of some side effects than placebo.

  • Precision: Imprecise. Confidence intervals cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not assessed. Although effects of dose on side effects were not evaluated, dose-response relationships are noted for efficacy of valbenazine and deutetrabenazine.

  • Confounding factors: Unclear. No specific confounding factors are noted for valbenazine or deutetrabenazine, but confounding factors may be present for tetrabenazine because of weaknesses in the study methodologies.

  • Publication bias: Unable to be assessed. The small number of available studies precludes assessments of publication bias.

  • Overall strength of research evidence: Low. Available studies of valbenazine and deutetrabenazine include RCTs of short duration, as well as longer-term open-label follow-up phases. Findings for these medications are consistent in showing no apparent difference in side effects relative to placebo. For tetrabenazine, studies have significant limitations and a high risk of bias but show more frequent side effects than placebo. Overall, studies are generally applicable to individuals with moderate to severe tardive dyskinesia, including individuals with a diagnosis of schizophrenia.

Psychosocial Interventions

Statement 15: Coordinated Specialty Care Programs

APA recommends (1B) that patients with schizophrenia who are experiencing a first episode of psychosis be treated in a coordinated specialty care program.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This recommendation is based on evidence from four clinical trials as presented in the AHRQ review (McDonagh et al. 2017) as well as an additional study (Anderson et al. 2018) that showed reduced mortality at 2 years for those who had participated in an early intervention program. The strength of research evidence for this statement is rated as moderate on the basis of the moderate SOE found for multiple key outcomes.

Many studies of coordinated specialty care programs were not included in the AHRQ review, although there is a long history of using such programs worldwide for early identification and treatment of attenuated psychosis syndrome, related syndromes of high psychosis risk, or first-episode psychosis (J. Addington et al. 2017; Cotton et al. 2016; Malla and McGorry 2019); however, these programs were not within the scope of this guideline recommendation because they include individuals who do not have a psychiatric diagnosis or who have diagnoses other than schizophrenia at later follow-up times (Fusar-Poli et al. 2016; Iorfino et al. 2019). Only one of the studies in a meta-analysis of early intervention programs (Marshall and Rathbone 2011) was included in the AHRQ analysis; all other studies were excluded because of such factors as small sample size, enrollment of inpatients, lack of applicability to the U.S. population, and/or use of a one-off intervention.

Pooled results from studies of individuals with a first episode of psychosis (McDonagh et al. 2017) found that up to 2 years of treatment with a coordinated specialty care program was associated with higher global functioning based on Global Assessment of Functioning (GAF) and Global Assessment Scale scores (3 RCTs; WMD 3.88, 95% CI 0.91–6.85; moderate SOE), significantly more people working or in school (3 RCTs; RR 1.22, 95% CI 1.01–1.47; moderate SOE), significantly higher ratings of quality of life (2 RCTs; effect size 0.84, 95% CI 0.14–1.55; moderate SOE), and a greater rate of retention in treatment (RR 1.27, 95% CI 1.16–1.38; Cochran’s Q = 0.03, degrees of freedom = 1; high SOE) as compared with usual care. Coordinated specialty care program participants were also less likely to relapse compared with those in usual care (2 RCTs; RR 0.64, 95% CI 0.52–0.79; moderate SOE). These treatment effects were not sustained and had generally dissipated by 5 years after treatment discontinuation. In addition, as compared with usual care, there were no significant effects of coordinated specialty care programs on housing status (2 RCTs; low SOE), self-harm (N = 506; RR 0.93, 95% CI 0.06–14.81), or total PANSS scores (3 RCTs; WMD –2.53, 95% CI –5.45 to 0.39; low SOE).

One study (Bertelsen et al. 2007; Nordentoft et al. 2002; Secher et al. 2015) found no differences in rates of accidental death (RR 0.31, 95% CI 0.01–7.59) or unexplained death (RR 0.31, 95% CI 0.01–7.56) at 2 years and no difference in mortality at 10-year follow-up (RR 0.92, 95% CI 0.45–1.88) between individuals who received a coordinated specialty care intervention as compared with usual care. In contrast, in an early intervention program for psychosis in Ontario, Canada (Anderson et al. 2018), rates of self-harm behavior (HR 0.86, 95% CI 0.18–4.24) and suicide (HR 0.73, 95% CI 0.29–1.80) did not differ during the initial 2 years after enrollment as compared with usual care, but rates of all-cause mortality were lower in the coordinated specialty care intervention group (HR 0.24, 95% CI 0.11–0.53; absolute risk reduction 2.5%; NNT = 40). As compared with those who received usual care, individuals in the coordinated specialty care program also saw a psychiatrist more rapidly (user median days = 13 compared with nonuser median days = 78), were more likely to have contact with a psychiatrist (HR 6.05, 95% CI 5.30–6.91), and were more likely to be hospitalized (HR 1.42, 95% CI 1.18–1.71) but were less likely to have emergency department visits (HR 0.71, 95% CI 0.60–0.83) or primary care contacts (HR 0.46, 95% CI 0.41–0.52).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Coordinated Specialty Care Programs

  • Magnitude of effect: Moderate. CSC programs are associated with significant benefits in terms of retention in treatment, with smaller benefits on quality of life, functioning, and relapse reduction.

  • Risk of bias: Medium. Studies are RCTs with a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia as well as individuals with a first episode of psychosis in the context of other diagnoses. In usual practice, coordinated specialty care programs follow the same model as research programs and would be expected to be comparable.

  • Directness: Direct. Studies measure core illness symptoms, functioning, quality of life, relapse, and treatment retention.

  • Consistency: Consistent. Findings are generally consistent among the available RCTs.

  • Precision: Variable. Confidence intervals are generally narrow and do not cross the threshold for clinically significant benefit of the intervention for the majority of outcomes. For some outcomes, however, imprecision was noted because of wide confidence intervals.

  • Dose-response relationship: Not assessed. Studies do not examine effects of varying treatment duration or visit frequency.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Unable to be assessed. A small number of studies met inclusion criteria for the AHRQ review, which precludes assessment of publication bias.

  • Overall strength of research evidence: Moderate. The strength of research evidence is moderate for most outcomes, although a high strength of evidence is noted for rates of treatment retention, and a low strength of research evidence is noted for housing status and core illness symptoms as measured by the PANSS. Neither of the latter outcomes showed an effect of coordinated specialty care programs. Trials vary in sample size, but several had large samples and findings were generally consistent among the available RCTs.

Grading of the Overall Supporting Body of Research Evidence for Harms of Coordinated Specialty Care Programs

Harms of coordinated specialty care programs were not systematically studied, and no grading of the evidence for harms is possible.

Statement 16: Cognitive-Behavioral Therapy

APA recommends (1B) that patients with schizophrenia be treated with cognitive-behavioral therapy for psychosis (CBTp).*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence in support of this statement comes from multiple RCTs and meta-analyses as described in the AHRQ review (McDonagh et al. 2017). The strength of the research evidence is rated as moderate on the basis of the evidence of CBTp benefits for core illness symptoms and short-term functioning.

In terms of overall symptoms, the AHRQ report relied primarily on a systematic review of 34 RCTs (Jauhar et al. 2014) that found CBTp to be more effective than usual care at improving overall symptoms based on symptom-based rating scales such as the PANSS and BPRS (SMD –0.33, 95% CI –0.47 to –0.19; I2 = 68%; moderate SOE). The effect was less pronounced but remained significant (95% CI –0.27 to –0.03) when the analysis was restricted to trials with blinded outcome assessments. Because this review did not conduct stratified analysis by format, it is not possible to tell whether distinctions in outcome exist for individual as compared with group CBTp. Although the included studies ranged in duration from 8 weeks to 5 years, analysis of shorter as compared with longer durations of treatment was not conducted, limiting the ability to determine whether more prolonged treatment is able to maintain shorter-term treatment gains. For negative symptoms, there was no meaningful difference noted between CBTp and usual care based on data from two systematic reviews (Jauhar et al. 2014; Velthorst et al. 2015) (low SOE).

The AHRQ report (McDonagh et al. 2017) also found CBTp to be associated with improvements in global function in the short term ( 6 months since CBTp initiation) as measured using the GAF scale (6 trials: MD 5.35, 95% CI 1.05–9.65; I2 = 77%). Removing the one study that used group CBTp from the analysis strengthened the effect and eliminated statistical heterogeneity. In one study that focused on global function (van der Gaag et al. 2011), a higher proportion of CBTp patients had normal functioning after 6 months of treatment as compared with patients who received usual care (28% vs. 14%; RR 2.21, 95% CI 1.25–3.93). Another study (Zimmer et al. 2007) found short-term benefits of an integrated cognitive-behavioral intervention on social and/or occupational function (as measured by the Social and Occupational Functioning Assessment Scale and overall functioning (GAF mean difference 5.69, 95% CI 2.05–10.97; p = 0.000). Benefits of CBTp on global, social, and occupational function were not maintained for more than 6 months after treatment cessation (low SOE). CBTp also improved quality of life more than usual care in the short term but not with longer periods of follow-up (low SOE).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Cognitive-Behavioral Therapy for Psychosis

  • Magnitude of effect: Moderate. With outcomes for which an effect is observed (such as core illness symptoms and short-term functioning improvements), there is a moderate magnitude of benefit. For other outcomes, either no benefit is seen or evidence is inconclusive.

  • Risk of bias: Medium. Studies in meta-analyses and other RCTs have a moderate degree of study limitations based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. Some studies also include individuals with other diagnoses such as schizoaffective disorder. When delivered in clinical practice, CBTp may be conducted with a lesser degree of fidelity than when delivered in research contexts.

  • Directness: Direct. Studies measure core illness symptoms; negative symptoms; and social, occupational, and global function.

  • Consistency: Variable. For outcomes with an observed effect, study findings are consistent. For outcomes with no observed effect, study findings are less consistent.

  • Precision: Precise. Confidence intervals are narrow and do not cross the threshold for clinically significant benefit of the intervention for outcomes with an observed effect. For outcomes with no observed effect, findings are imprecise for most outcomes.

  • Dose-response relationship: Not assessed. The impact of differences in the numbers or frequency of treatment sessions is unclear.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Unclear. At least one meta-analysis of CBTp suggests that publication bias may be present, but analysis for possible publication bias based on all of the included studies was not available.

  • Overall strength of research evidence: Moderate. For outcomes that show an effect of CBTp, there is a moderate strength of research evidence. For other outcomes that did not show an effect, the strength of research evidence is low or insufficient.

Grading of the Overall Supporting Body of Research Evidence for Harms of Cognitive-Behavioral Therapy for Psychosis

Harms of the treatment were rarely reported in studies of CBTp and were not systematically studied. Information was available from only a single RCT, and the AHRQ report notes that the evidence is insufficient to draw any conclusions (McDonagh et al. 2017). Thus, no grading of the evidence for harms is possible.

Statement 17: Psychoeducation

APA recommends (1B) that patients with schizophrenia receive psychoeducation.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence in support of this statement comes from a good-quality systematic review as described in the AHRQ report (McDonagh et al. 2017). The strength of the research evidence is rated as moderate on the basis of the evidence of psychoeducation benefits on relapse rates.

The 10 RCTs (total N = 1,125) of psychoeducation included in the systematic review (Pekkala and Merinder 2002) varied in length (with duration of follow-up as long as 5 years), included diverse interventions, and used individual and group techniques. Although most of the studies included stabilized outpatients and were conducted in North America and northern Europe, many studies contained some individuals with a diagnosis other than schizophrenia. On the basis of the data from 6 trials, psychoeducation had a greater effect than usual care on relapse rates (with or without readmission) at 9–18 months of follow-up (RR 0.80, 95% CI 0.70–0.92; moderate SOE). Psychoeducation was also superior to usual care in terms of global functional outcomes at 1 year of follow-up (3 RCTs; MD –5.23, 95% CI –8.76 to –1.71; low SOE).

In terms of potential harms, few studies reported adverse outcomes. Nevertheless, for psychoeducation as compared with usual care, no differences were observed in the number of deaths, which were small in both groups, and rates of all-cause study dropout were also comparable between study groups (McDonagh et al. 2017).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Psychoeducation

  • Magnitude of effect: Small. A modest benefit is seen for psychoeducation in terms of relapse rates and global functional outcomes. Effects on other outcomes are less robust or are inconclusive.

  • Risk of bias: Medium. Studies are RCTs, including RCTs as part of a meta-analysis, and have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. There is considerable variability in the content and format of interventions; however, variability is also present in the delivery of psychoeducation in usual clinical practice.

  • Directness: Direct. Studies measure relapse rates, symptoms, functioning, and quality of life.

  • Consistency: Consistent. When outcome data are available from multiple studies, findings are consistent.

  • Precision: Variable. The confidence interval is narrow for relapse at 9–18 months, but other outcomes have wide confidence intervals.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Unclear. The most recent meta-analysis of psychoeducation in schizophrenia did not assess whether publication bias was present.

  • Overall strength of research evidence: Moderate. Available RCTs on effects of psychoeducation have reasonable sample sizes and precise, consistent findings on relapse. For functioning, the overall strength of research evidence is low.

Grading of the Overall Supporting Body of Research Evidence for Harms of Psychoeducation

Harms of psychoeducation were reported in only a few studies but appeared comparable to usual care; no grading of the evidence for harms is possible because of the small amount of available evidence.

Statement 18: Supported Employment Services

APA recommends (1B) that patients with schizophrenia receive supported employment services.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Evidence in support of this statement comes from one study comparing supported employment with usual care and an RCT and meta-analysis comparing supported employment with other vocational interventions as described in the AHRQ report (McDonagh et al. 2017). The strength of the research evidence is rated as moderate on the basis of the evidence of benefits for supported employment on obtaining competitive work.

The AHRQ review (McDonagh et al. 2017) found that supported employment, using the individual placement and support (IPS) model, results in better employment outcomes than usual care with 2 years of follow-up. Patients receiving IPS in one fair-quality trial (N = 204) were significantly more likely to obtain competitive work than those receiving usual care (75% vs. 27.5%, P = 0.001; Mueser et al. 2004). They were also more likely to obtain any form of paid work than those receiving usual care (73.9% vs. 53.6%). A large RCT (N = 1,273), with both usual care and vocational training comparisons, showed similar benefits of IPS (55% vs. 34%, P < 0.001 overall and 22% vs. 12%, P < 0.001 in the subgroup of study subjects with schizophrenia) (Cook et al. 2005). These findings are consistent with findings of a good-quality systematic review of 14 RCTs (N = 2,265) in which other vocational training interventions were used as controls (Kinoshita et al. 2013). Together, these studies provide a moderate SOE for benefits of supported employment using IPS. Although associated with a lower SOE, supported employment also showed benefits in terms of working more than 20 hours per week (13% vs. 34%, P = 0.00), having more weeks of employment overall (24 more weeks competitive and 11 more weeks any employment, P < 0.001), and longer tenure per individual job (4 weeks, P = 0.048) than those in either usual care, other vocational interventions, or both. Patients receiving IPS also reported earning more money than those in usual care ($2,078/month vs. $617.59/month, P < 0.001).

Several other meta-analyses of supported employment using somewhat different analytic methods and different inclusion and exclusion criteria than the AHRQ review found similar benefits of supported employment using the IPS approach (Frederick and VanderWeele 2019; Suijkerbuijk et al. 2017). In terms of outcomes unrelated to work, findings are less consistent but suggest potential advantages to supported employment in reducing symptoms and hospitalization risk (Burns et al. 2007; Hoffmann et al. 2014; Luciano et al. 2014).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Supported Employment Services

  • Magnitude of effect: Moderate. Moderate effects are present for the proportion of individuals attaining competitive employment or any form of paid employment as compared with usual care or vocational training comparisons.

  • Risk of bias: Medium. Studies are RCTs (including meta-analyses of RCTs) and have a medium risk of bias based on descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. Supported employment interventions, particularly those using the IPS model, appear to be representative of usual clinical practice.

  • Directness: Direct. Studies measure the duration of competitive employment and the proportion of individuals attaining paid or competitive employment, as well as less direct outcomes such as wages earned.

  • Consistency: Consistent. When outcome data are available from more than one study, findings are consistent. In addition, several meta-analyses using somewhat different inclusion and exclusion criteria reached similar findings.

  • Precision: Precise. Narrow confidence intervals are present for the proportion of individuals in competitive employment or working at least 20 hours per week, as well as for mean weeks worked and mean wages earned.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Suspected. There appears to be a lack of reporting of smaller trials with negative results based on network meta-analysis and associated funnel plots to identify publication bias (Suijkerbuijk et al. 2017).

  • Overall strength of research evidence: Moderate. There is a moderate overall strength of research evidence for the proportion of individuals attaining competitive employment, any form of paid employment, mean weeks worked, or monthly wages earned as compared with usual care or vocational training comparisons. Trials are of varying quality, but many have a good sample size; large numbers of study subjects are included in meta-analyses.

Grading of the Overall Supporting Body of Research Evidence for Harms of Supported Employment Services

Harms of supported employment services were not systematically studied, and no grading of the evidence for harms is possible.

Statement 19: Assertive Community Treatment

APA recommends (1B) that patients with schizophrenia receive assertive community treatment if there is a history of poor engagement with services leading to frequent relapse or social disruption (e.g., homelessness; legal difficulties, including imprisonment).*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This recommendation is based on information from the AHRQ review (McDonagh et al. 2017), which used a good-quality systematic review (14 RCTs; N = 2,281) as a primary source (Marshall and Lockwood 2000) and also considered one additional RCT (N = 118; Sytema et al. 2007). The strength of research evidence for this guideline statement is rated as moderate on the basis of the moderate SOE found for multiple key outcomes.

The AHRQ review (McDonagh et al. 2017) focused on assertive community treatment (ACT) alone as compared with usual care and did not include a recent review in which evidence for ACT was combined with evidence for intensive case management. Significant variability was noted in study populations, with a range of ages, demographic characteristics, diagnoses, and eligibility criteria (e.g., frequent or recent hospitalization, other risk factors for reduced continuity of care). In addition, the degree of fidelity to ACT principles was often unclear, which may influence effectiveness.

Individuals who received ACT were less likely to discontinue treatment and be lost to follow-up than individuals who received usual care (12 trials; OR 0.51, 95% CI 0.41–0.63; moderate SOE). They were also less likely to be admitted to a hospital compared with individuals who received usual care (6 RCTs; OR 0.59, 95% CI 0.41–0.85; I2 = 73%), and many of the reported studies also showed a decrease in the number of days in the hospital. Furthermore, individuals who received ACT were less likely to be unemployed (3 trials; OR 0.46, 95% CI 0.21–0.99; I2 = 34%), homeless (4 trials; OR 0.20, 95% CI 0.09–0.47; I2 = 52%), or living nonindependently (4 trials; OR 0.52, 95% CI 0.35–0.79; I2 = 0%) compared with usual care (moderate SOE). Core illness symptoms also improved with ACT, but the degree of improvement was comparable to that in the usual care group (3 trials, N = 255; MD –0.14, 95% CI –0.36 to 0.08; moderate SOE). As compared with usual care, there was no significant difference in social function (pooled analysis of 3 studies; MD 0.03, 95% CI –0.28 to 0.34; low SOE) and no significant differences in arrests (2 trials, total N = 604; OR 1.17, 95% CI 0.60–2.29; I2 = 0%), imprisonment (4 trials, total N = 471; OR 1.19, 95% CI 0.70–2.01; I2 = 27%), or police contacts (2 trials, total N = 149; OR 0.76, 95% CI 0.32–1.79; I2 = 84%) with ACT (low SOE). Findings from the additional RCT were generally consistent with the meta-analytic results. Only 2 trials reported information on quality of life, with one finding a small but statistically significant difference and the other showing no difference.

In individuals with co-occurring schizophrenia and a substance use disorder, one good-quality systematic review of 32 trials (N = 3,165) examined differences between integrated ACT and usual care (Hunt et al. 2013). For most outcomes of interest, only one or two of the studies from the systematic review contributed relevant data; however, these limited data showed no differences between integrated ACT and usual care for substance use, treatment discontinuation, function, or mortality through follow-up durations of up to 36 months.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Assertive Community Treatment

  • Magnitude of effect: Moderate. On several important outcomes, there is at least a moderate effect. These outcomes include treatment discontinuation and likelihood of hospitalization, unemployment, or homelessness. On other outcomes, however, there is no difference between ACT and treatment as usual.

  • Risk of bias: Medium. Evidence comes from an RCT and a meta-analysis of RCTs, but individual studies have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. The delivery of ACT in the trials is likely to be representative of usual clinical practice, although program fidelity may be diminished in usual practice.

  • Directness: Direct. Studies measured core illness symptoms, functioning, quality of life, employment, housing status, and interactions with the criminal justice system.

  • Consistency: Variable. Findings of benefits for having housing or employment are consistent, whereas other outcomes show inconsistencies among individual studies within the meta-analysis.

  • Precision: Imprecise. Confidence intervals for most outcomes are wide, and many cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Not applicable.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Not assessed.

  • Overall strength of research evidence: Moderate. A significant number of RCTs of ACT are available and, compared with usual care, show that ACT is associated with a lower likelihood of being unemployed and a lower likelihood of not living independently or being homeless. Many trials are moderate in quality, although there is some variability.

Grading of the Overall Supporting Body of Research Evidence for Harms of Assertive Community Treatment

Harms of assertive community treatment were not systematically studied, and no grading of the evidence for harms is possible.

Statement 20: Family Interventions

APA suggests (2B) that patients with schizophrenia who have ongoing contact with family receive family interventions.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This guideline statement is based on 1 fair-quality systematic review (27 non-Chinese studies, total N = 2,297; Pharoah et al. 2010) and 6 additional studies (total N = 562; Barrowclough et al. 1999; Dyck et al. 2000; Garety et al. 2008; Kopelowicz et al. 2012; Mayoral et al. 2015; Sellwood et al. 2001, 2007; Valencia et al. 2007) as described in the AHRQ review (McDonagh et al. 2017). Because most family interventions are aimed at reducing relapse, the strength of research evidence for this statement is rated as moderate on the basis of the moderate SOE found for relapse in the AHRQ review with medium-term follow-up, although other outcomes had a low SOE.

The studies of family intervention described in the AHRQ review (McDonagh et al. 2017) had significant variation in content and methods of the delivered intervention (e.g., psychoeducation, motivational interviewing, behavioral family therapy, support groups, 24-hour support, communication training, stress management, role-play, homework, goal setting, development of social networks), measured outcomes, study design (e.g., setting, treatment duration, treatment frequency, follow-up duration, single-family versus multiple-family format, family members only vs. family members plus patient), and demographics of the study population (e.g., age, illness duration, symptom severity at baseline). Most studies had small sample sizes, and most had some risk of bias due to lack of reporting of randomization procedures or outcome assessment blinding. Studies conducted in China were excluded because of concerns about their applicability to Canadian and U.S. populations.

In the AHRQ review (McDonagh et al. 2017), family interventions resulted in significantly lower relapse rates than usual care when measured at 0–6 months (3 RCTs; N = 244; 23% vs. 37%; RR 0.62, 95% CI 0.41–0.92; low SOE), 7–12 months (19 RCTs; N = 1,118; 30% vs. 44%; RR 0.67, 95% CI 0.54–0.83; moderate SOE), 13–24 months (9 RCTs; N = 517; 49% vs. 61%; RR 0.75, 95% CI 0.58–0.99; low SOE), and 5 years posttreatment (2 RCTs; N = 140; 78% vs. 94%; RR 0.82, 95% CI 0.72–0.94; low SOE) but not at 25–36 months. The strongest evidence for effects of family interventions on relapse occurred in studies that included at least 10 treatment sessions over 7–12 months.

Improvements in core illness symptoms (4 RCTs; N = 223; SMD –0.46, 95% CI –0.73 to –0.20; low SOE) and negative symptoms (3 trials; N = 163; SMD –0.38, 95% CI –0.69 to –0.07; low SOE) were also found with family intervention compared with usual care. Unemployment (4 trials; N = 230; 75% vs. 66% after 6–12 months of follow-up; RR 1.09, 95% CI 0.91–1.29; I2 = 0%; low SOE), independent living (3 RCTs; N = 164; 57% vs. 63% at 1 year; RR 0.83, 95% CI 0.66–1.03; low SOE), and reduction in self-harm (6 trials; N = 314; 4% vs. 6%; RR 0.85, 95% CI 0.24–3.02; I2 = 23%; low SOE) were not found to be different between family intervention and usual care groups (low SOE). For social functioning, quality of life, family burden, and nonsuicide mortality, there was insufficient evidence to draw any conclusions from the available studies. Rates of treatment discontinuation varied with time in the study, but family interventions either did not differ from usual care or had fewer treatment dropouts than usual care (McDonagh et al. 2017; low SOE).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Family Interventions

  • Magnitude of effect: Moderate. For outcomes that show an effect of family interventions, such as effects on relapse rate and illness symptoms, the magnitude of the effect is generally moderate.

  • Risk of bias: Medium. Evidence comes from RCTs, including RCTs in a fair-quality systematic review. These studies have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. Studies from China are excluded from the analysis because of concerns about their applicability to Canadian and U.S. populations. There is considerable variability in the content and methods of the family interventions that are studied; however, there is also considerable variability in interventions that are delivered in clinical practice.

  • Directness: Direct. Studies measure relapse, core illness symptoms, and outcomes related to functioning.

  • Consistency: Consistent. For outcomes with data from more than one study, findings among the studies are consistent.

  • Precision: Imprecise. Confidence intervals for all outcomes are wide or cross the threshold for clinically significant benefit of the intervention.

  • Dose-response relationship: Present. The strongest evidence for effects of family interventions on relapse occurs in studies that include at least 10 treatment sessions over 7–12 months.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Not assessed. The most recent meta-analysis of family interventions did not assess whether publication bias was present.

  • Overall strength of research evidence: Moderate. For relapse at 7–12 months, there is a moderate strength of research evidence, but for most other outcomes, the strength of research evidence is low. The strength of research evidence is influenced by a lack of precision, as well as by the small sample sizes for some of the outcomes.

Grading of the Overall Supporting Body of Research Evidence for Harms of Family Interventions

Harms of family interventions were not systematically studied, and no grading of the evidence for harms is possible.

Statement 21: Self-Management Skills and Recovery-Focused Interventions

APA suggests (2C) that patients with schizophrenia receive interventions aimed at developing self-management skills and enhancing person-oriented recovery.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This guideline statement is based on evidence provided by a fair-quality systematic review (13 studies; total N = 1,404; Zou et al. 2013) and 1 additional fair-quality study (N = 210; Hasson-Ohayon et al. 2007) as described in the AHRQ review (McDonagh et al. 2017), as well as a meta-analysis of person-oriented recovery approaches (7 RCTs, N = 1,739; Thomas et al. 2018). The strength of research evidence for this statement is rated as low on the basis of the low SOE found for the majority of outcomes in the AHRQ review and a significant risk of bias (consistent with a low SOE) for most of the studies in the meta-analysis of person-oriented recovery approaches.

For illness self-management training and for recovery-focused interventions, interpretation of the evidence can be challenging because of the degree of heterogeneity in the content and format of the interventions. For example, illness self-management training programs are designed to improve knowledge, management of symptoms, and social and occupational functioning, with a primary goal of reducing the risk of relapse by focusing on medication management, recognizing signs of relapse, and developing a relapse prevention plan and coping skills for persistent symptoms (McDonagh et al. 2017; Substance Abuse and Mental Health Services Administration 2010a). Recovery-focused interventions can include similar approaches but are focused primarily on supporting a recovery-oriented vision that strives for community integration in the context of individual goals, needs, and strengths (Le Boutillier et al. 2011; Thomas et al. 2018). Activities of recovery-focused interventions incorporate opportunities for participants to share experiences and receive support as well as practicing strategies for success in illness self-management. With illness self-management, the interventions were typically administered in a group format, whereas recovery-focused interventions included a mix of group and individual formats as well as a mix of peer- and professional-led activities. Both illness self-management and recovery-focused interventions had significant variations in session content, duration, and number.

In terms of outcomes with illness self-management, the AHRQ review (McDonagh et al. 2017) noted a reduction in core illness symptom severity based on the BPRS (5 RCTs; pooled WMD –4.19, 95% CI –5.84 to –2.54; moderate SOE) and a reduced likelihood of relapse with completion of 10 or more self-management sessions (OR 0.41, 95% CI 0.21–0.79; low SOE). Effects of intervention were reduced if low-fidelity treatment was given or if fewer self-management sessions were completed. No significant effect of illness self-management was noted for negative symptoms (low SOE).

With recovery-focused interventions (Thomas et al. 2018), individuals in the intervention group showed a modest improvement in person-oriented recovery, empowerment, and hope immediately after the intervention (effect size 0.24, 95% CI 0.04–0.44) and at follow-up (effect size 0.21, 95% CI 0.06–0.35). Moderator analysis suggested that the greatest improvement was seen when mental health professionals and peer providers collaborated in treatment delivery.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Self-Management Skills and Recovery-Focused Interventions

  • Magnitude of effect: Small. Modest changes were noted in core illness symptom severity, likelihood of relapse, measures of person-oriented recovery, empowerment, and hope.

  • Risk of bias: Medium. Studies are RCTs, including RCTs as part of a meta-analysis, and have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. There is considerable variability in the content and format of delivered interventions; however, there is also wide variation in illness self-management and recovery-focused interventions in clinical practice.

  • Directness: Direct. Studies measure functioning, symptoms, and relapse.

  • Consistency: Variable. Consistent for symptoms and relapse but inconsistent for functioning.

  • Precision: Variable. Precise for symptoms as measured by the BPRS but imprecise for other outcomes because of wide confidence intervals.

  • Dose-response relationship: Present. Effects of illness self-management were less prominent if fewer self-management sessions were completed.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Not assessed.

  • Overall strength of research evidence: Low. Studies are RCTs, including a meta-analysis of RCTs, and have a medium risk of bias. The strength of research evidence is moderate for effects on symptoms; it is low or insufficient for other outcomes. Although a dose-response effect seems to be present, increasing confidence in the findings, this is offset by the lack of precision for most outcomes.

Grading of the Overall Supporting Body of Research Evidence for Harms of Self-Management Skills and Recovery-Focused Interventions

Harms of self-management skills and recovery-focused interventions were not systematically studied, and no grading of the evidence for harms is possible.

Statement 22: Cognitive Remediation

APA suggests (2C) that patients with schizophrenia receive cognitive remediation.*

This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This guideline statement is based on evidence provided by 2 good-quality systematic reviews (57 studies, total N = 2,885; Cella et al. 2017; Wykes et al. 2011), 1 good-quality trial (N = 90; Deste et al. 2015; Vita et al. 2011), and 3 fair-quality trials (N = 56–156; Farreny et al. 2012; Mueller et al. 2015; Twamley et al. 2012). The strength of research evidence for this statement is rated as low on the basis of the low SOE found for the majority of outcomes.

Studies included in the AHRQ review (McDonagh et al. 2017) used standard cognitive remediation principles (Saperstein and Medalia 2012) and usual care control comparisons, but other population and study characteristics varied (e.g., population demographics, treatment setting, individual vs. group format, drill and practice vs. drill plus strategy methodology, sessions per week, treatment duration, follow-up duration, extent of treatment fidelity, baseline symptom severity, computerized vs. noncomputerized content delivery, presence of active comparator condition).

Overall, as compared with usual care, use of cognitive remediation for 15–16 weeks of treatment was associated with small positive effects on core illness symptoms (2 trials, N = 153; SMD –0.62, 95% CI –1.01 to –0.24; low SOE), but effects were inconsistent among the studies, and symptom improvement was not sustained following treatment removal (8 RCTs; effect size 0.17, 95% CI –0.03 to 0.48). Cognitive remediation as compared with usual care was also associated with improvements in negative symptoms (1 systematic review of 18 RCTs; effect size –0.36, 95% CI –0.52 to –0.20; moderate SOE) as well as small positive effects on social, occupational, and global function (6 RCTs; effect sizes of 0.16–0.40; low SOE). Effects of intervention on cognitive functioning were outside the scope of the AHRQ review, but some evidence suggests that improved cognitive function can result from treatment with cognitive remediation, with indirect benefits for global function (Harvey et al. 2018). Only 1 study reported on health-related quality of life, and study limitations preclude drawing conclusions on this outcome. Treatment with cognitive remediation did not differ from usual care in terms of rates of treatment discontinuation (McDonagh et al. 2017). Cognitive remediation also seems to be acceptable to individuals who receive treatment in clinical settings as compared with research settings (Medalia et al. 2019).

Although the AHRQ review did not specifically assess cognitive outcomes with cognitive remediation, this has been a major focus of study and the primary target of cognitive remediation as an intervention. Results from meta-analyses (Revell et al. 2015; Wykes et al. 2011) and more recent randomized trials (D’Amato et al. 2011; Donohoe et al. 2018; Gomar et al. 2015; Keefe et al. 2012; McGurk et al. 2016; Mueller et al. 2015; Reeder et al. 2017) have been mixed, with significant heterogeneity in the degree of cognitive improvement (if any), the domains of cognitive improvement, and the persistence and generalizability of improvements. This may not be surprising given the wide variety of cognitive remediation approaches and formats that have been used in an effort to enhance cognitive processes such as attention, memory, executive function, social cognition, or meta-cognition (Delahunty and Morice 1996; Medalia et al. 2018; Pentaraki et al. 2017; Reeder et al. 2016; Wykes et al. 2011). There are also no clear-cut factors that are predictive of whether cognitive improvement will occur (Reser et al. 2019), which makes it difficult to determine how to target the intervention to individuals who are most likely to respond. Nevertheless, cognitive remediation does seem to result in improvements in cognition in individuals with schizophrenia, at least on a short-term basis (Harvey et al. 2018; Revell et al. 2015).

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Cognitive Remediation

  • Magnitude of effect: Small. Small but significant effects are seen for core illness symptoms and negative symptoms as well as for cognitive processes in some domains. However, significant heterogeneity is present in the degree of benefit as well as the persistence and generalizability of benefits.

  • Risk of bias: Medium. RCTs, including those in systematic reviews, have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. Multiple different approaches to delivering cognitive remediation are used in the clinical trials. In addition, the use of cognitive remediation remains limited outside research settings, which makes it difficult to compare the study methods with current practice.

  • Directness: Direct. Studies measure core illness symptoms, functioning, quality of life, and treatment discontinuation as well as cognitive effects.

  • Consistency: Consistent. Within each outcome, study findings were consistent.

  • Precision: Imprecise. Confidence intervals are relatively wide for many outcomes.

  • Dose-response relationship: Not assessed. It is not clear whether using a different frequency or duration of cognitive remediation sessions will affect outcomes.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Not suspected. On the basis of analyses conducted as part of meta-analyses on cognitive remediation, there is no evidence of publication bias.

  • Overall strength of research evidence: Low. Ratings of the strength of evidence are low for global, social, and occupational function and for core illness symptoms and moderate for negative symptoms. There is significant variability in the findings, perhaps related to the many differences in the study populations and treatment-related characteristics. Nevertheless, this reduces confidence in conclusions related to cognitive remediation.

Grading of the Overall Supporting Body of Research Evidence for Harms of Cognitive Remediation

Harms of cognitive remediation were not systematically studied, and no grading of the evidence for harms is possible.

Statement 23: Social Skills Training

APA suggests (2C) that patients with schizophrenia who have a therapeutic goal of enhanced social functioning receive social skills training.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This guideline statement is based on 3 fair-quality RCTs (total N = 384; Bartels et al. 2014; Mueser et al. 2010; Valencia et al. 2007, 2013) as described in the AHRQ review (McDonagh et al. 2017). The strength of research evidence for this statement is rated as low on the basis of the low SOE found for outcomes in the AHRQ review with social skills training.

In the trials of social skills training that were included in the AHRQ review (McDonagh et al. 2017), sessions were held weekly for 24–52 weeks and included specific, progressive intervention modules on such topics as management of symptoms and medication, improving social and family relationships, and increasing functional skills such as money management. Goals of social skills training included enhanced psychosocial function and reductions in relapse and need for hospitalization. Demographic parameters, diagnoses of participants, and outcome measures varied among the trials.

Social function was noted to be significantly improved with social skills training as compared with usual care (SMD on GAF at 6 months 1.60, 95% CI 1.19–2.02; SMD on GAF at 1 year 2.02, 95% CI 1.53–2.52; SMD on Multnomah Community Ability Scale at 2 years 0.65, 95% CI 0.36–0.95; low SOE), but it was not clear whether gains were maintained after treatment discontinuation. Core illness symptoms, as measured by the PANSS, also showed more improvement with social skills training as compared with usual care (SMD at 6 months –1.50, 95% CI –1.92 to –1.09; SMD at 2 years –0.81, 95% CI –1.22 to –0.40; low SOE). Negative symptoms also improved with social skills training as compared with usual care (SMD at 6 months –1.30, 95% CI –1.70 to –0.90; SMD at 1 year –0.82, 95% CI –1.23 to –1.40; SMD at 2 years –0.45, 95% CI –0.74 to –0.15; low SOE), and in one study gains were maintained 1 year after treatment had ended. It was unclear whether relapse rates were affected by social skills training because of a small number of studies, small sample sizes, and small numbers of individuals who experienced relapse. In terms of treatment discontinuation, individuals who received social skills training did not differ from those in the usual care group (RR 1.10 at 1 year, 95% CI 0.92–1.31; RR 1.01 at 2 years, 95% CI 0.88–1.16; low SOE), with high rates of treatment retention in both groups.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Social Skills Training

  • Magnitude of effect: Small. A modest effect of social skills training was noted on social function, core illness symptoms, and negative symptoms.

  • Risk of bias: Medium. Studies are RCTs with a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, many of the trials also include individuals with other diagnoses. Typically, individuals also had decreases in social functioning.

  • Directness: Direct. Studies measure social functioning, core illness symptoms, negative symptoms, relapse, and ability to maintain treatment.

  • Consistency: Variable. Findings in the 3 included studies are consistent for negative symptom improvements but inconsistent for improvements in functioning and core illness symptoms.

  • Precision: Imprecise. Confidence intervals for some outcomes cross the threshold for clinically significant benefit of the intervention in some studies.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Present. Confounding factors may increase the observed effect. Subjects and treating clinicians are aware of the treatment arm to which subjects were assigned. This may cause confounding of effects due to expectancies.

  • Publication bias: Not suspected. A meta-analysis conducted subsequent to the AHRQ review showed no significant publication bias for studies of social skills training (Turner et al. 2018).

  • Overall strength of research evidence: Low. Available RCTs have medium risk of bias, have variable consistency, and lack precision, reducing the strength of the evidence.

Grading of the Overall Supporting Body of Research Evidence for Harms of Social Skills Training

Harms of social skills training were not systematically studied, and no grading of the evidence for harms is possible.

Statement 24: Supportive Psychotherapy

APA suggests (2C) that patients with schizophrenia be treated with supportive psychotherapy.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

This guideline statement is based on studies that compared supportive psychotherapy with usual care (total N = 822) in 1 good-quality systematic review (L. A. Buckley et al. 2015) as described in the AHRQ review (McDonagh et al. 2017). The strength of research evidence for this statement is rated as low on the basis of the low SOE found for study outcomes in the AHRQ review.

The studies in the systematic review (L. A. Buckley et al. 2015) were aimed primarily at helping patients with coping abilities and maintaining levels of functioning. In other respects, there was significant variation in measured outcomes, study design (e.g., setting, treatment duration, treatment frequency, follow-up duration), and demographics of the study population (e.g., age, illness duration, symptom severity at baseline). In addition, most of the included studies had some risk of bias.

The AHRQ review (McDonagh et al. 2017) found no difference in global or social function on the basis of 2 studies, but study results were not able to be pooled for analysis. Four RCTs reported information on study attrition, and no significant difference was noted between supportive psychotherapy and usual care (N = 354; RR 0.86, 95% CI 0.53–1.40; low SOE). For other outcomes, evidence was available from only a single study and sample sizes were small, making it difficult to draw reliable conclusions.

Grading of the Overall Supporting Body of Research Evidence for Efficacy of Supportive Psychotherapy

  • Magnitude of effect: Not present. On the basis of a small number of studies, there is no difference from usual care in global functioning or treatment discontinuation.

  • Risk of bias: Medium. Studies are RCTs that have a medium risk of bias based on their descriptions of randomization, blinding procedures, and study dropouts.

  • Applicability: The included trials all involve individuals with schizophrenia. However, some trials also include individuals with other diagnoses. There is significant variation in the duration and frequency of treatment; however, variability in the delivery of supportive psychotherapy is also common in usual clinical practice.

  • Directness: Direct. Studies measure functioning, core symptoms, negative symptoms, relapse, quality of life, and treatment discontinuation.

  • Consistency: Consistent. For outcomes that are studied in more than one trial, findings are generally consistent.

  • Precision: Imprecise. Confidence intervals are wide and cross the threshold for clinically significant benefit of the intervention for many outcomes.

  • Dose-response relationship: Not assessed.

  • Confounding factors: Unclear. Supportive therapy is similar to the type of therapy that is commonly delivered in usual care, so expectancy effects of receiving a novel intervention are likely to be minimal.

  • Publication bias: Unable to be assessed. The number of studies on supportive therapy is too small to be able to assess for the presence or absence of publication bias.

  • Overall strength of research evidence: Low. The overall strength of evidence is low for global functioning and study discontinuation. There is insufficient evidence to draw conclusions about other outcomes. The available trials vary in their sample sizes. Study designs also differ, which makes comparison of findings difficult.

Grading of the Overall Supporting Body of Research Evidence for Harms of Supportive Psychotherapy

Harms of supportive psychotherapy were not systematically studied, and no grading of the evidence for harms is possible.