Skip to main content
Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie logoLink to Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie
. 2018 Jun 4;63(10):661–678. doi: 10.1177/0706743718779950

Harms of Antipsychotics in Children and Young Adults: A Systematic Review Update

Avantages et inconvénients des antipsychotiques chez les enfants et les jeunes adultes : mise à jour d’une revue systématique

Jennifer Pillay 1,, Khrista Boylan 2, Amanda Newton 1,3, Lisa Hartling 1,3, Ben Vandermeer 1, Megan Nuspl 1, Tara MacGregor 1, Robin Featherstone 1, Normand Carrey 4,5
PMCID: PMC6187435  PMID: 29865900

Abstract

Objective:

To update and extend our previous systematic review on first- (FGAs) and second-generation antipsychotics (SGAs) for treatment of psychiatric and behavioral conditions in children, adolescents, and young adults (aged ≤24 years). This article focuses on the evidence for harms.

Method:

We searched (to April 2016) 8 databases, gray literature, trial registries, Food and Drug Administration reports, and reference lists. Two reviewers conducted study screening and selection independently, with consensus for selection. One reviewer extracted and another verified all data; 2 reviewers independently assessed risk of bias. We conducted meta-analyses when appropriate and network meta-analysis across conditions for changes in body composition. Two reviewers reached consensus for ratings on the strength of evidence for prespecified outcomes.

Results:

A total of 135 studies (95 trials and 40 observational) were included, and 126 reported on harms. FGAs caused slightly less weight gain and more extrapyramidal symptoms than SGAs. SGAs as a class caused adverse effects, including weight gain, high triglyceride levels, extrapyramidal symptoms, sedation, and somnolence. They appeared to increase the risk for high cholesterol levels and type 2 diabetes. Many outcomes for individual drug comparisons were of low or insufficient strength of evidence. Olanzapine caused more short-term gains in weight and body mass index than several other SGAs. The dose of SGAs may not make a difference over the short term for some outcomes.

Conclusions:

Clinicians need to weigh carefully the benefit-to-harm ratio when using antipsychotics, especially when treatment alternatives exist. More evidence is needed on the comparative harms between antipsychotics over the longer term.

Keywords: children, pediatric, young adult, antipsychotic agents, systematic review


In Canada, prescribing of antipsychotics for children, adolescents, and young adults increased dramatically (upwards of 4-fold) between 1996 and 2011,14 with 12% of prescriptions provided to Canadian children younger than 9 years.5 In 2013, second-generation antipsychotics (SGAs) constituted 97% of all antipsychotic prescriptions.6 SGAs were frequently prescribed to augment stimulants.1,7 Most antipsychotic prescriptions for children are off-label in Canada and in some cases are being used as first-line treatment despite recommendations for their use in second- or beyond-line therapies.8

A shift away from first-generation antipsychotics (FGAs) toward SGAs has been driven by perceptions of their more favorable harm profile (e.g., less sedation and fewer extrapyramidal symptoms [EPS] and other adverse effects [AEs] caused by the persistent dopamine receptor blockade by FGAs). However, SGAs have been associated with AEs such as weight gain, elevated prolactin levels, and metabolic syndrome.9,10 This risk profile is concerning because of known associations between obesity with type 2 diabetes, dyslipidemia, and hypertension.11 While Canadian guidance exists for monitoring the safety of antipsychotics in children and youth,12 ongoing systematic review on the efficacy and harms of SGAs and FGAs is needed to best inform treatment decisions. Compilation of evidence on harms across multiple conditions may also help inform situations in which harms data are insufficient for reviews underpinning condition-specific guidance.13

In this systematic review, we examined the benefits and harms from the use of FGAs and SGAs for various psychiatric and behavioral conditions in children, adolescents, and young adults ≤24 years of age. Here, we report on the results for harms across all conditions; we will report on benefits within conditions in subsequent articles. This is an update and extension of a review commissioned by the Agency for Healthcare Research and Quality (AHRQ) and published in 2012.14

Methods

Methods for this review are based on the AHRQ Methods Guide15 and an a priori protocol (available at http://www.crd.york.ac.uk; CRD No.42016032943 and https://effectivehealthcare.ahrq.gov/topics/antipsychotics-children-update/research-2017). Panels of stakeholders, including a patient representative and clinical experts, as well as public commentary informed our methods, and full details on the methods and results are in the full report on AHRQ’s Effective Healtcare website (www.effectivehealthcare.ahrq.gov/reports/final.cfm).16

Identification and Selection of Studies

A research librarian ran searches to April 2016 for studies published after 1987 in MEDLINE (Suppl. Material Appendix A), EMBASE, PsycINFO, CINAHL, ProQuest® Dissertations and Theses Global, and TOXLINE. We did not just update the previous search conducted in 201114 because of our broader scope including additional conditions (e.g., anxiety, substance use disorders) and medications (e.g., molindone). We scanned reference lists of included studies and relevant systematic reviews, searched clinical trial registries, and hand searched relevant journals and Food and Drug Administration (FDA) reports. Studies were included based on predefined eligibility criteria; Table 1 includes all criteria for both benefits and harms. The antipsychotic had to be approved by the FDA but not necessarily for the condition(s) studied in the articles. Additional outcomes were considered (see full report16), although we focus here on the ones considered by our stakeholders to be the most important for decision making. For example, we focused on reports of abnormal serum lipids rather than mean changes in serum levels, which may not reflect a clinically relevant degree of harm. Based on clinical input, we assessed harms across all conditions rather than within each condition because of a lack of strong evidence of a difference in AEs based on indication.

Table 1.

Eligibility Criteria for the Review.

Category Criteria
Population Children and young adults (≤24 years)a with 1 or more of the following conditions/issues: anxiety disorder, ADHD/DICD, ASD, bipolar disorder, depression, eating disorders, OCD, PTSD, substance use disorders, schizophrenia, tic disorders, or behavioral issues outside the context of a disorder (e.g., insomnia) For benefits: data must be provided for groups having ≥90% of patients diagnosed with the specific condition(s) For harms: studies could focus on single or mixed conditions; >90% of the mixed study population must include patients with the conditions of interest Subgroup variables: sex, age, race/ethnicity, comorbidities/co-conditions, history of treatment, phase and features of disorder (e.g., acute mania vs. maintenance in bipolar disorder, first-episode or prodromal schizophrenia); treatment condition (for harms across conditions)
Interventions Any FDA-approved FGA (chlorpromazine, droperidol, fluphenazine, haloperidol, loxapine, molindone, perphenazine, pimozide, prochlorperazine, thiothixene, thioridazine, trifluoperazine) Any FDA-approved SGA (aripiprazole, asenapine, brexpiprazole, cariprazine, clozapine, iloperidone, lurasidone, olanzapine, paliperidone, quetiapine, risperidone, ziprasidone) All formulations and doses eligible Subgroup variables: co-treatments (e.g., other medication, nonpharmacological therapy, as reported), medication dose, duration of treatment
Comparators Placebo/no treatment, any other antipsychotic, or same antipsychotic at different dose Exclusion of nonantipsychotic medications as comparator
Outcomesb Benefits: condition-specific core symptoms (e.g., negative symptoms in schizophrenia, hyperactivity and aggression in ADHD or DICD), response and remission rates, global clinical impressions, quality of life, cognitive and emotional development and functioning, suicide-related ideations and behaviors, legal or justice system interactions Harms: major (e.g., mortality, development of diabetes, tardive dyskinesia, agranulocytosis, cardiac myopathies) and general (i.e., neuromotor effects, metabolic effects including body composition and dyslipidemia, somnolence and sedation, prolactin-related effects)
Timing Publication date: 1987 onwards Follow-up duration: no minimum; categories: <6 months, ≥6 months to <12 months, ≥12 months
Setting Any setting
Study design Clinical trials (RCTs and CCTs), controlled cohort studies (prospective or retrospective), controlled before-after studies (e.g., open-label extensions with comparator group), pooled analyses of individual patient-level data from one or a combination of similar trials
Language English

Abbreviations: ADHD, attention-deficit/hyperactivity disorder; ASD, autism spectrum disorders; CCT, controlled clinical trial; DICD, disruptive, impulse-control, or conduct disorders; FDA, Food and Drug Administration; FGA, first-generation antipsychotic; OCD, obsessive-compulsive disorder; PTSD, posttraumatic stress disorder; RCT, randomized controlled trial; SGA, second-generation antipsychotic

a Studies in adult populations were included if they provided data specific to younger ages (mean age + 1 standard deviation ≤24).

b Outcomes considered most important by our stakeholders. Additional outcomes were included, and results are in the full report (www.effectivehealthcare.ahrq.com).

For the database searches, 2 reviewers independently screened the titles and abstracts of all citations, and any deemed relevant by either reviewer based on broad criteria were reviewed in full text in duplicate using a standardized form. One reviewer conducted all other searches outlined in the above section. Disagreements on final inclusion of all studies were resolved through consensus or a third reviewer.

Data Extraction and Analysis

A standardized data extraction form was created and piloted with all reviewers. Thereafter, one reviewer extracted data and another verified data. We reference the primary publication of each study; all associated literature is tabulated in the full report. We recorded intention-to-treat results, and for continuous outcomes, we extracted change scores where possible. If standard deviations were not reported, we computed these. For dichotomous outcomes, we reported counts or proportions and sample size by study arm. We used the longest follow-up duration when data were available for more than 1 time point within each of our follow-up strata (e.g., results for both 1- and 3-month follow-up were within our 0- to <6-month stratum). Only numerical data for harms were extracted; that is, we made no assumptions about lack or presence of an AE if it was not reported. We extracted data (taking care to avoid duplication with other study reports) on harms from trial registries and regulatory agency reports of pediatric trials.

We synthesized data in the following order based on type of comparison: all comparisons combined (for select outcomes, see description of network meta-analysis), aggregate (across drug class) data for FGAs versus SGAs, individual FGAs versus SGAs, within-class comparisons between individual FGAs and individual SGAs (other drug or dose), and then aggregate and individual data for FGAs versus placebo/no treatment and SGAs versus placebo/no treatment. For harms, we combined data from all study designs; empirical evidence has found no difference in estimates of harms between meta-analyses of randomized controlled trial (RCT) and cohort study designs,17 and cohort studies are commonly recognized as contributing relatively high-quality evidence applicable to real-world settings.

Various approaches to synthesizing the evidence are available, including direct pairwise meta-analysis and methods that combine direct and indirect evidence (i.e., network meta-analysis or mixed-treatment comparisons).18,19 The summary effect from direct comparisons (e.g., an SGA vs. placebo, one SGA or FGA vs. another SGA or FGA) for one outcome at a similar time point is meaningful. However, such an approach does not allow for comparisons between drugs that may not have much direct evidence (e.g., drug A was compared with drug B and C, but drugs B and C were not directly compared). Where feasible, we conducted network meta-analyses. Detailed methods for our Bayesian pairwise and network meta-analyses are reported in the online Supplemental Material. We considered pooling studies when 3 or more studies reported on the same outcome.

We report mean differences (MD) for continuous outcomes including weight gain; for dichotomous outcomes, we report the relative risk (RR). Variance in meta-analysis results are reported with a 95% credible interval (CrI; used for Bayesian analysis), which can be interpreted similarly to a 95% confidence interval (CI). When the upper or lower limit of the interval crosses 0.0 (for MD) or 1.0 (for RR), the results are not statistically significant.

For prespecified population and intervention subgroups (Table 1), we extracted information on analyses conducted by study authors and also performed our own subgroup analyses based on study-level data. We employed univariate Bayesian meta-regression analyses for our outcomes with the most data (weight, weight gain of 7% or greater, somnolence, incidence of EPS in SGA vs. placebo/no-treatment comparisons) in terms of patient age, sex, antipsychotic treatment history (i.e., % naïve), and treatment duration.

Quality Assessment and Grading the Body of Evidence

Two reviewers independently assessed the risk of bias of all studies based on study design.15,20,21 We also assessed the strength of the body of evidence, using 5 domains22: risk of bias, consistency, directness, precision, and reporting biases. For rare outcomes (i.e., <5%), findings were considered precise when the sample size (across studies) was at least 2000 patients, to estimate that adequate prognostic balance was achieved to detect at least a small difference.23 Our overall assessments of high, moderate, low, or insufficient strength of evidence (SOE) indicate the degree of certainty we had in the effect estimate. Discrepancies between reviewers were resolved based on consensus. Because of limited guidance from the literature, we did not grade the SOE for our network meta-analysis findings.

Results

We identified 135 studies (57 new studies after the 2012 report14) meeting our review criteria; of these, 126 (93%) reported on harms (Figure 1).24149 Eighty-four (67%) were RCTs, 6 (5%) were controlled clinical trials (experimental studies without randomization), and 36 (28%) were observational studies. Most (about 70%) studies reported only on follow-up durations of less than 6 months. The risk of bias was high for 55% of trials. The main contributor to risk of bias was incomplete outcome data (i.e., <90% of patients included in analysis). Overall, the observational studies were of good quality; of 36 studies, 4 (11%) were rated as having high risk of bias, 12 (33%) as having medium risk of bias, and 20 (56%) as low risk of bias. The Supplemental Material provides a table of study characteristics including findings for risk of bias of each study (Appendix B).

Figure 1.

Figure 1.

Flow of literature.

We present findings first from the network meta-analyses in which data across any study could be pooled to examine the impact of FGAs and SGAs on increases in weight and body mass index (BMI). Then, head-to-head comparisons between FGAs and SGAs were followed by head-to-head and placebo/no-treatment comparisons for FGAs and then SGAs. Tables 2 and 3 contain details for general AEs having moderate and low SOE, respectively. Results for major AEs are presented within the text only.

Table 2.

Findings with Moderate Strength of Evidencea: General Adverse Effects.

Comparison (G1 vs. G2) Outcome n Studies/Comparisons; n Patients GI G2 Relative Effectsb Conclusionsc
Events n Events n
Short-term findings
 FGAs vs SGAs Weight (kg) 14; 506 190 316 MD, –2.62; 95% CrI, –4.35 to –0.8638,44,52,72,91,96,104,117,131,132,149 FGAs were better
BMI (kg·m−2) 7; 236 73 163 MD, –1.57; 95% CrI, –2.49 to –0.5373,117,131,132 FGAs were better
 Olanzapine vs. risperidone Weight (kg) 13; 936 311 583 MD, 2.18; 95% CrI, 1.13 to 3.2531,36,45,46,66,67,81,105,116,117,131,132,142 Risperidone was better
BMI (kg·m−2) 9; 737 224 471 MD, 0.94; 95% CrI, 0.64 to 1.3031,45,46,66,67,87,117,131,132 Risperidone was slightly better
 Quetiapine vs. risperidone BMI (kg·m−2) 3; 463 116 347 MD, 0.04; 95% CrI, –1.34 to 1.2031,45,67 No significant difference
≥7% weight increase 4; 417 55 104 176 313 RR: 0.91; 95% CI, 0.56 to 1.4431,45,82,137 No significant difference
 Aripiprazole high dose (15/30 mg/d)  vs. low dose (10 mg/d) Weight (kg) 3; 463 229 234 MD, 0.22; 95% CrI, –0.64 to 1.0962,64,98 No significant difference
 Asenapine high dose (10 mg/d)  vs. low dose (5 mg/d) ≥7% weight increase 2; 376 10 8 99 90 9 11 95 92 RR, 1.07; 95% CI, 0.45 to 2.5158 RR, 0.74; 95% CI, 0.31 to 1.7657 No significant difference
Somnolence 2; 407 31 52 106 99 24 49 98 104 RR, 1.19; 95% CI, 0.76 to 1.8958 RR, 1.11; 95% CI, 0.85 to 1.4757 No significant difference
 Quetiapine high dose (600/800 mg/d)  vs. low dose (400 mg/d) ≥7% weight increase 2; 340 14 10 74 98 17 14 73 95 RR, 0.81; 95% CI, 0.43 to 1.5260 RR, 0.69; 95% CI, 0.32 to 1.48114 No significant difference
Somnolence 2; 340 22 31 74 98 20 27 73 95 RR, 1.09; 95% CI, 0.65 to 1.8160 RR, 1.11; 95% CI, 0.72 to 1.71114 No significant difference
Sedation 2; 340 22 31 74 98 20 27 73 95 RR, 0.99; 95% CI, 0.26 to 3.8060 RR, 1.10; 95% CI, 0.67 to 1.81114 No significant difference
 All SGAs vs. placebo/no treatment Any EPS 15; 2730 2; 32 233 0 1757 33 40 0 973 33 RR, 2.94; 95% CI, 2.02 to 4.2727,53,59,60,62,64,75,77,98,114,127,130,134,139,148 Not estimable49,127 SGAs increased risk
Weight (kg) 37; 3919 2384 1535 MD, 1.53; 95% CI, 1.11 to 1.982628,33,39,43,49,50,53,54,5964,75,79,80,86,89,90,94,98,102,113,114,124,130,133,134,138,139,141,145,148 SGAs increased slightly
BMI (kg·m−2) 16; 2462 1582 880 MD, 0.66; 95% CI, 0.44 to 0.9133,49,57,62,64,75,86,89,90,94,98,113,120,134,138,148 SGAs increased slightly
≥7% weight increase 17; 3057 337 2023 42 1034 RR, 3.53; 95% CrI, 2.49 to 5.235760,6264,75,80,90,98,113,114,133,138,141,145 SGAs increased risk
Increased triglycerides 10; 1383 130 897 38 486 RR, 1.64; 95% CrI, 1.09 to 2.6326,49,52,60,62,90,98,113,114,138 SGAs increased risk
Sedation 21; 2710 288 1696 79 1014 RR, 2.19; 95% CrI, 1.50 to 3.4126,43,49,50,54,58,60-63,75,80,85,86,91,94,98,113,114,124,148 SGAs increased risk
Somnolence 26; 3942 56 2481 119 1461 RR, 2.91; 95% CrI, 2.27 to 3.8627,33,39,53,54,58,60,62-64,75,77,86,90,94,98,102,113,114,124,130,133,134,139,145,148 SGAs increased risk
 Aripiprazole vs. placebo Weight (kg) 7; 1042 647 395 MD, 0.98; 95% CrI, 0.54 to 1.4859,62,64,98,113,139,148 Aripiprazole increased slightly
BMI (kg·m−2) 5; 881 587 294 MD, 0.33; 95% CI, 0.07 to 0.6762,64,98,113,148 Aripiprazole increased slightly
 Olanzapine vs. placebo Weight (kg) 4; 337 215 122 MD, 3.96; 95% CI, 2.31 to 6.3480,90,138,145 Olanzapine increased
BMI (kg·m−2) 2; 267 — — 107 72 — — 54 34 MD, 1.16; 95% CI, 0.93 to 1.39138 MD, 1.50; 95% CI, 1.06 to 1.9490 Olanzapine increased
 Quetiapine vs. placebo Weight (kg) 6; 778 473 305 MD, 1.44; 95% CI, 0.60 to 2.3143,49,50,60,63,114 Quetiapine increased slightly
 Risperidone vs. placebo Weight (kg) 14; 929 522 475 MD, 1.52; 95% CI, 0.78 to 2.292628,33,39,61,75,79,86,89,102,130,134,141 Risperidone increased slightly
BMI (kg·m−2) 6; 730 397 333 MD, 0.68; 95% CI, 0.27 to 1.1833,75,86,89,120,134 Risperidone increased slightly
Somnolence 9; 862 163 473 43 389 RR, 3.25; 95% CrI, 1.96 to 5.9427,33,39,75,77,86,102,130,134 Risperidone increased risk
 Ziprasidone vs. placebo Weight (kg) 3; 360 246 114 MD, –0.10; 95% CI, –1.34 to 1.1353,54,124 No significant difference
Long-term findings
 Olanzapine vs. quetiapine Weight (kg), 6 to <12 months 3; 185 90 95 MD, 7.91; 95% CrI, 3.65 to 12.2931,32,67 Quetiapine was better
BMI (kg·m−2), 6 to <12 months 4; 203 99 104 MD, 2.68; 95% CrI, 0.96 to 4.2731,32,41,67 Quetiapine was better
 Olanzapine vs. risperidone BMI (kg·m−2), 6 to <12 months 5; 328 94 234 MD, 1.66; 95% CrI, 0.19 to 3.4231,41,66,67,131 Risperidone was slightly better

Abbreviations: BMI, body mass index; CI, confidence interval; CrI, credible interval; EPS, extrapyramidal symptoms; FGA, first-generation antipsychotic; G1, group 1; G2, group 2; MD, mean difference; RR, risk ratio; SGA, second-generation antipsychotic; SOA, strength of evidence.

aMost outcomes were downgraded from high to moderate SOE for risk of bias; outcomes for different doses of asenapine were downgraded instead for imprecision, because 95% CIs include possibility for clinically relevant benefit for the low-dose group.

bRRs greater than 1.0 and MDs greater than 0 favor group 2. We did not pool data from 1 or 2 studies, so these results are always presented separately.

cFor weight and BMI, the effects were considered small when gains were approximately <1.5 kg and <1 kg/m2, respectively.

Table 3.

Findings with Low Strength of Evidencea: General Adverse Effects.

Comparison (G1 vs. G2) Outcomes n Studies/Comparisons; n Patients GI G2 Relative Effects,b Studies Conclusions
Events n Events n
Short-term findings (<6 months)
 FGAs vs. SGAs Any EPS 4; 110 16 37 13 73 RR, 2.59; 95% CrI, 1.00 to 7.0096,117,148 SGAs decreased risk
Sedation 7; 345 70 160 79 185 RR, 1.04; 95% CrI, 0.86 to 1.3744,117,131,132 No significant difference
 Clozapine vs. olanzapine Weight (kg) 5; 136 62 74 MD, –1.56; 95% CrI, –5.12 to 1.5766,81,92,93,129 No significant difference
 Olanzapine vs. risperidone ≥7% increase in weight 6; 504 107 150 188 354 RR, 1.36; 95% CrI, 0.93 to 2.0431,45,66,82,117,142 No significant difference
Sedation 7; 321 35 133 36 188 RR, 1.19; 95% CrI, 0.73 to 2.3524,36,68,82,117,131,132 No significant difference
 Olanzapine vs. quetiapine Weight (kg) 3; 232 116 116 MD, 4.00; 95% CrI, –1.67 to 10.7931,45,67 No significant difference
BMI (kg·m−2) 3; 232 116 116 MD, 1.36; 95% CrI, –0.29 to 3.4031,45,67 No significant difference
≥7% increase in weight 3; 192 72 99 47 93 RR: 1.41; 95% CI, 0.65 to 2.8331,45,82 No significant difference
 Quetiapine vs. risperidone Weight (kg) 3; 463 116 347 MD, 0.08; 95% CrI, –3.77 to 3.1431,45,67 No significant difference
Hyperprolactinemia 4; 118 4 31 45 87 RR, 0.20; 95% CrI, 0.06 to 0.7324,100,123,137 Quetiapine decreased risk
 Aripiprazole high dose (15/30mg/d) vs.   low dose (10 mg/d) Any EPS 2; 310 39 12 99 54 23 13 98 59 RR, 1.68; 95% CI, 1.09 to 2.5962 RR, 1.01; 95% CI, 0.50 to 2.0298 No significant difference
BMI (kg·m−2) 3; 456 223 233 MD, 0.14; 95% CrI, –0.47 to 5.8662,64,98 No significant difference
≥7% weight increase 3; 506 37 250 24 256 RR, 1.62; 95% CrI, 0.47 to 5.8662,64,98 No significant difference
High cholesterol 2; 242 28 0 65 54 27 0 64 59 RR, 1.02; 95% CI, 0.68 to 1.5262 Not estimable98 No significant difference
High triglycerides 2; 243 22 2 65 54 22 6 65 59 RR, 1.00; 95% CI, 0.62 to 1.6262 RR: 0.36; 95% CI, 0.08 to 1.7398 No significant difference
Somnolence 3; 512 62 255 47 257 RR, 1.31; 95% CrI, 0.46 to 3.8062,64,98 No significant difference
 Asenapine high dose (10 mg/d) vs.   low dose (5 mg/d) BMI (kg·m−2) 1; 198 99 99 MD, 0.03; 95% CI, –0.04 to 0.1058 No significant difference
Hyperprolactinemia 1; 204 20 106 23 98 RR, 1.24; 95% CI, 0.73 to 2.1258 No significant difference
 Risperidone high dose (3-6 mg/d) vs. low dose (0.5-3 mg/d) Any EPS 2; 217 20 15 51 61 18 4 55 50 RR, 1.20; 95% CI, 0.72 to 2.0077 RR, 3.07; 95% CI, 1.09 to 8.6875 No significant difference
Somnolence 2; 217 6 34 51 61 13 21 55 50 RR, 0.50; 95% CI, 0.20 to 1.2177 RR, 1.33; 95% CI, 0.89 to 1.9775 No significant difference
 Clozapine vs. olanzapine Weight (kg) 5; 136 62 74 MD, –1.56; 95% CrI, –5.12 to 1.5766,81,92,93,129 No significant difference
 Olanzapine vs. quetiapine Weight (kg) 3; 232 116 116 MD, 4.00; 95% CrI, –1.67 to 10.7931,45,67 No significant difference
BMI (kg·m−2) 3; 232 116 116 MD, 1.36; 95% CrI, –0.29 to 3.4031,45,67 No significant difference
≥7% increase in weight 3; 192 72 99 47 93 RR: 1.41; 95% CI, 0.65 to 2.8331,45,82 No significant difference
 Olanzapine vs. risperidone ≥7% increase in weight 6; 504 107 150 188 354 RR, 1.36; 95% CrI, 0.93 to 2.0431,45,66,82,117,142 No significant difference
Sedation 7; 321 35 133 36 188 RR, 1.19; 95% CrI, 0.73 to 2.3524,36,68,82,117,131,132 No significant difference
 Quetiapine vs. risperidone Weight (kg) 3; 463 116 347 MD, 0.08; 95% CrI, –3.77 to 3.1431,45,67 No significant difference
Hyperprolactinemia 4; 118 4 31 45 87 RR, 0.20; 95% CrI, 0.06 to 0.7324,100,123,137 Quetiapine decreased risk
 All SGAs vs. placebo Akathisia 21; 3638 151 2433 56 1205 RR, 1.29; 95% CrI, 0.81 to 2.2739,43,53,54,57,58,62,64,75,77,86,90,94,97,98,113,124,133,138,139,148 No significant difference
Increased total cholesterol 6; 643 1; 218 92 0 410 52 13 0 233 166 RR, 3.17; 95% CrI, 1.29 to 9.1349,52,62,113,114,138 Not estimable98 SGAs increase risk
 Aripiprazole vs. placebo Any EPS 6; 1000 117 655 17 345 RR, 3.10; 95% CrI, 1.26 to 7.0159,62,64,98,139,148 Aripiprazole increased risk
≥7% increase in weight 5; 991 93 647 15 344 RR, 3.01; 95% CrI, 1.33 to 7.1059,62,64,98,113 Aripiprazole increased risk
Somnolence 6; 1012 119 661 29 351 RR, 2.73; 95% CrI, 1.24 to 7.6562,64,98,113,139,148 Aripiprazole increased risk
 Olanzapine vs. placebo ≥7% increase in weight 4; 337 99 215 8 122 RR, 6.08; 95% CrI, 1.84 to 27.0680,90,138,145 Olanzapine increased risk
Hyperprolactinemia 2; 268 50 58 107 72 1 6 54 35 RR, 25.53; 95% CI, 3.58 to 177.76138 RR, 4.70; 95% CI, 2.25 to 9.8290 Olanzapine increased risk
 Quetiapine vs. placebo Sedation 6; 778 90 473 32 305 RR, 1.67; 95% CrI, 0.77 to 3.8743,49,50,60,63,114 No significant difference
Somnolence 3; 697 106 432 18 265 RR, 2.95; 95% CrI, 0.92 to 8.6260,63,114 No significant difference
 Risperidone vs. placebo Any EPS 5; 636 52 365 13 271 RR, 2.78; 95% CrI, 1.27 to 6.5027,75,77,130,134 Risperidone increased risk
 Ziprasidone vs. placebo Somnolence 3; 548 76 358 13 190 RR, 2.97; 95% CrI, 0.84 to 9.9653,54,124 No significant difference
Long-term findings
 Olanzapine vs. risperidone Weight (kg), 6 to <12 months 4; 295 85 210 MD, 4.40; 95% CrI, -0.54 to 9.8631,66,67,131 No significant difference
≥7% increase in weight, 6 to <12 months 3; 264 28 64 64 200 RR: 1.44; 95% CI, 0.55 to 5.5031,42,66 No significant difference
 Quetiapine vs. risperidone Weight (kg), 6 to <12 months 3; 295 93 202 MD, –1.48; 95% CrI, –4.16 to 1.1831,67,122 No significant difference
BMI (kg·m−2), 6 to <12 months 4; 328 102 226 MD, –0.32; 95% CrI, –1.56 to 1.1231,41,67,122 No significant difference
 Risperidone vs. placebo Weight (kg), 6 to <12 months 4; 467 239 228 MD, 2.86; 95% CrI, –1.22 to 7.4295,99,106,120 No significant difference
BMI (kg·m−2), 6 to <12 months 2; 405 209 196 MD, 0.70; 95% CI, 0.49 to 0.91120 MD, 1.80; 95% CI, –0.61 to 4.2199 No significant difference

Abbreviations: BMI, body mass index; CI, confidence interval; CrI, credible interval; EPS, extrapyramidal symptoms; FGA, first-generation antipsychotic; G1, group 1; G2, group 2; MD, mean difference; RR, risk ratio; SGA, second-generation antipsychotic.

aMost outcomes were downgraded for risk of bias and imprecision, because 95% CIs included possibility for clinically relevant benefit for one group reducing our confidence in no effects.

bRRs greater than 1.0 and MDs greater than 0 favor group 2. We did not pool data from 1 or 2 studies, so these results are always presented separately.

Network Meta-analyses for Body Composition Outcomes across All Antipsychotics

Network meta-analysis was conducted for the outcomes of weight gain (Figure 2; n = 71 studies) and changes to BMI (Suppl. Figure S1; n = 35 studies). Results are presented in terms of a placebo referent, to rank the drugs based on a common comparator, but data from head-to-head comparisons were incorporated in the analysis. Results showed that patients taking most antipsychotics gain more weight than do patients taking placebo or not receiving antipsychotics. The relative harm from olanzapine (22 studies, n = 611; 4.12 kg, 95% CrI, 3.43 to 4.88) was most robust compared with aripiprazole (11 studies, n = 869; 0.88 kg, 95% CrI, 0.26 to 1.50), quetiapine (12 studies, n = 655; 1.25 kg, 95% CrI, 0.51 to 1.95), and risperidone (37 studies, n = 1535; 1.85 kg, 95% CrI, 1.40 to 2.35), because of the higher precision in these estimates compared with other drugs (e.g., pimozide, molindone, and lurasidone had fewer patients contributing to the findings). For BMI, olanzapine (16 studies, n = 470; 1.51 kg/m−2, 95% CrI, 1.28 to 1.84) remained worse than all other antipsychotics except for, possibly, clozapine (Suppl. Figure S1). We used data for longest treatment duration from each study for these analyses, but 81% (weight) and 71% (BMI) of studies were short term (often 6-12 weeks).

Figure 2.

Figure 2.

Plot of weight gain by first-generation antipsychotics (FGAs) and second-generation antipsychotics (SGAs) compared with placebo/no treatment This plot shows the findings from network meta-analyses combining placebo-controlled and head-to-head comparisons of FGAs and SGAs within one analysis. The effects shown represent the mean difference and credible intervals of each drug relative to placebo, which was used as the reference standard.

FGAs versus SGAs

Nine studies reported on major (4 long-term duration)* and 16 reported on general AEs (2 long-term) when comparing FGAs with SGAs. All findings on major AEs (i.e., neuroleptic malignant syndrome, seizures, cardiac arrhythmias, agranulocytosis, diabetes mellitus) had insufficient SOE. FGAs caused lower gains in weight and BMI than did SGAs (Table 2). Compared with SGAs, FGAs increased the short-term risk for experiencing any EPS symptom (Table 3). Low SOE found no significant difference between classes for sedation (Table 3). Because of risk of bias and imprecision due to very small samples sizes, evidence was insufficient for other across-class outcomes (e.g., akathisia, dystonia, hyperprolactinemia) and for all outcomes in individual drug comparisons.

FGAs: Comparisons between FGAs and between FGAs and Placebo/No Treatment

There was insufficient SOE for all major and general AEs in comparisons between different FGAs or between different doses of the same FGA and between FGAs (aggregate or individual) compared with placebo or no treatment. Single, very small studies29,115,118,125,128,136 reported data for a small number of outcomes within the short- and long-term duration categories.

SGAs: Comparisons between SGAs

Sixteen (5 long-term) studies reported on major AEs and 37 (13 long-term) reported on general AEs.

For conditions for which SGAs were not considered primary treatment (i.e., exclusion of patients with schizophrenia, autistic spectrum disorders, tic disorders, and all patients talking clozapine), aripiprazole (hazard ratio [HR] vs. no antipsychotic, 7.72; 95% CI, 3.70 to 16.12) may increase the risk for developing diabetes more than risperidone (HR, 2.20; 95% CI, 1.14 to 4.26) in patients newly initiated and remaining on these drugs for at least a year (low SOE).37 Data for other major AEs were insufficient, mostly from single studies having inadequate sample sizes for these rare events and moderate or higher risk of bias.

For weight and BMI, findings from pairwise comparisons between individual SGAs were similar to those seen from the network meta-analysis (Figure 1; Tables 2 and 3). There was less precision in some of the findings (due to smaller samples; Table 3), although for comparison with olanzapine in which no statistical significance was found, the limits of the 95% CrIs usually suggested more harm for olanzapine. Findings for clinically meaningful gains in weight (≥7% body weight) were of lower SOE; findings were not statistically significant but imprecise, with the 95% CIs including findings for meaningful harm from olanzapine.

Other findings with low SOE for individual SGA drug comparisons included no significant difference between olanzapine and risperidone for sedation and between quetiapine and risperidone for hyperprolactinemia (Table 3).

Different doses of aripiprazole resulted in no significant difference in short-term weight gain (Table 2). There was low SOE showing no difference between doses of aripiprazole over the short term for any EPS symptoms, BMI, the proportion gaining ≥7% weight, and somnolence (Table 3); the 95% CIs included values favoring the low dose. There was no difference for hypertriglyceridemia or increased total cholesterol (low SOE). There was no significant difference in the short term between low and high doses of asenapine for the proportion of patients gaining ≥7% weight, for risk of somnolence, or for hyperprolactinemia. Low and high doses of quetiapine were not significantly different for risk of gaining ≥7% weight, somnolence, or sedation over the short term (moderate SOE). There was no significant difference between low and high doses of risperidone for any EPS or somnolence (low SOE).

SGAs: Comparisons with Placebo/No Treatment

There was no significant difference in the short term across all SGAs compared with placebo/no antipsychotic for mortality (13 studies, n = 2447; 0 events in either group) or for having a pathologically prolonged QT interval (14 studies, n = 2425; SGAs 14 of 1490 and placebo/no antipsychotic 9 of 935) (both moderate SOE). Compared with no antipsychotic treatment, patients newly initiated on SGAs may have an increased risk for developing diabetes over treatment of at least 1 year (low SOE; HR, 2.89; 95% CI, 1.64 to 5.10; 25.3 vs. 7.8 cases per 10,000 person-years follow-up).37 Moreover, across all of the antipsychotics evaluated in this study (risperidone, olanzapine, aripiprazole, quetiapine, and ziprasidone), the risk remained elevated for up to 1 year following discontinuation of the drug. Other major AEs were assessed as having insufficient SOE due to rare events occurring in samples too small (<2000) to offer adequate power.

There was moderate SOE that SGAs were worse than placebo/no treatment for 7 general AE outcomes: any EPS symptoms, changes to body composition (weight, BMI, and ≥7% weight gain), increased triglycerides, sedation, and somnolence (Figure 3; Table 2). Low SOE found no significant difference for akathisia and that SGAs may increase the risk for hypercholesterolemia (Figure 3; Table 3).

Figure 3.

Figure 3.

Results of meta-analyses for second-generation antipsychotics (SGAs) versus placebo/no treatment. EPS, extrapyramidal symptoms; RR, relative risk; SOE, strength of evidence (degree of certainty review authors had in the effect size [RR] based on study quality, consistency in effects, precision of findings, other limitations; no outcome had high SOE). Each row is from a different meta-analysis of studies comparing all SGAs versus placebo/no treatment. The number of studies and participants contributing to each meta-analysis is indicated.

For individual SGAs compared with placebo/no treatment, findings for weight and BMI were similar to those found by our network meta-analyses (Table 3). Risperidone and aripiprazole increased the risk for any EPS and for somnolence; findings for somnolence showing no significant difference for quetiapine and ziprasidone were low SOE because the possibility of meaningful harm from the SGA could not be ruled out. Although findings for hyperprolactinemia across all SGAs were insufficient, there was large heterogeneity between the SGAs, and findings specific to olanzapine were of low SOE, indicating this SGA increases the risk.

Subgroup Effects

Twenty-six studies reported on within-study subgroup analyses for harm outcomes (see full report). Findings were often inconsistent. Body composition, fasting glucose, and prolactin elevations do not appear to differ in patients taking SGAs and concomitant psychostimulants.25,45 Dose of SGAs—particularly when considering cumulative doses—was found in 2 large, high-quality observational studies to increase the risk for metabolic effects (e.g., >10 mg/d olanzapine or >1.5 mg/d risperidone)45 and development of diabetes (across all SGA doses vs. no antipsychotic: HR, 2.89 [95% CI, 1.674 to 5.10); ≥100-g dose equivalents of all SGAs: HR, 5.61 [95% CI, 2.38 to 13.24]; and ≥100 g risperidone: HR, 14.95 [95% CI, 4.60 to 48.64]).37 Quetiapine114 and risperidone44,7577,120 appear to increase serum prolactin more in females than in males. Findings for effect moderation on risk for somnolence and neuromotor effects were mainly from single studies.

Only one subgroup effect was evident based on our meta-regressions using study-level data. Slightly greater weight changes can be expected over longer treatment durations (0.043 kg [95% CrI, 0.015 to 0.072] per week of additional treatment). Results for the study-level variables of mean age, sex (% male), and percentage with previous antipsychotic exposure were not statistically significant for weight or the other outcomes we examined in the regressions (greater than 7% increase in weight, somnolence, and EPS symptoms). Based on observations of the plots of data used for our meta-regressions, we could not see any trends indicating the effects varied by condition (Suppl. Figures S2-S5). The results for conditions for which these drugs are typically used in lower doses (e.g., attention-deficit hyperactivity disorder) than for other conditions (e.g., schizophrenia) appeared to be very similar when looking across studies. These study-level observations should be interpreted with caution.

Discussion

We systematically reviewed the harms of FGAs and SGAs across a wide array of psychiatric conditions in young people. As commonly recognized, FGAs as a class cause less weight gain and more EPS than SGAs. There may be no difference between the classes for sedation, and findings for many other harms were either not reported or of insufficient quality. SGAs have a poorer harms profile than placebo or no antipsychotic treatment for weight gain, somnolence, risk of high triglycerides, EPS, and sedation. Some differences exist between SGAs, with olanzapine being worse than several SGAs for body composition outcomes, although several comparisons had low or insufficient SOE. There is some indication that SGAs may increase the risk for hypercholesterolemia and type 2 diabetes compared with placebo or no antipsychotic treatment. The dose of SGAs may not make a difference over the short term for some outcomes, but there is some indication of worse effects over the longer term.

Overall, our conclusions for some harms are fairly similar to those reported by other review authors.150154 However, our focus on clinically meaningful outcomes (e.g., hypercholesterolemia instead of mean cholesterol) and applying our criteria for SOE (in particular with respect to judgments of precision) limited our certainty about many effects, including between-drug differences about which other authors have made more definitive conclusions (see the full report16 for details).

Findings are mainly applicable to acute, rather than maintenance, treatment. Patients with a history of various adverse events, including tardive dyskinesia, suicide-related behaviors, neuroleptic malignant syndrome, or abnormal laboratory values, were often excluded from studies. The mean age across the studies was greater than 8 years, and most studies did not include patients older than 18 years; therefore, the evidence is not directly applicable to young children or young adults. The applicability of study findings outside of research settings is also limited, as monitoring practices to ensure treatment adherence and timely dose adjustments based on response and tolerability assessments are less likely to occur. Although comprehensive and individualized monitoring for AEs has been recommended for several years,155,156 evidence suggests that these practices remain inadequate157 or may be difficult to maintain.4

Limitations to this review are discussed in the Supplemental Materials. Results of the network meta-analyses should be considered exploratory in nature because of the use (i.e., modeling) of direct and indirect comparisons. Nevertheless, findings were quite consistent with those from the pairwise/direct comparisons described. This report was limited to comparisons between various antipsychotics and between antipsychotics and placebo or no antipsychotic treatment. As such, evidence on the use of other drug classes (e.g., anticonvulsants, mood stabilizers) that are frequently used in the treatment of these patient populations was not considered. We did not assess any outcome as having high SOE, although it should be recognized that attaining high SOE from trials of antipsychotics in children with psychiatric conditions is likely very difficult, and the overall evidence reviewed should not be interpreted as lacking in credibility.

Conclusion

Clinicians need to weigh carefully the benefit-to-harm ratio of antipsychotics when making prescribing decisions, especially in cases in which treatment alternatives exist and are supported by research evidence. More direct evidence on the harms of SGAs compared with FGAs, as well as from longer-term treatment studies, is required to advance clinical care. More evidence on sources of clinical heterogeneity, from gender, age, and other biological and environmental factors that may moderate the effects on harms, would be valuable.

Supplementary Material

779950_Supplementary_material - Harms of Antipsychotics in Children and Young Adults: A Systematic Review Update

779950_Supplementary_material for Harms of Antipsychotics in Children and Young Adults: A Systematic Review Update by Jennifer Pillay, Khrista Boylan, Amanda Newton, Lisa Hartling, Ben Vandermeer, Megan Nuspl, Tara MacGregor, Robin Featherstone, and Normand Carrey in The Canadian Journal of Psychiatry

Acknowledgments

We acknowledge with appreciation the staff at AHRQ, Laura Pincock and Christine Chang, for their efforts in helping us engage with key informants and the technical expert panel for this review; they also provided constructive comments on our draft full technical report (www.effectivehealthcare.ahrq.gov). In addition, we greatly value the time spent and excellent comments on our full technical report by EPC Associate Editor Marian McDonagh. We also thank the key informants and technical expert panel members (listed in the full report) who provided input into the review. Several technical expert panel members also provided peer review of the draft report submitted to AHRQ and available online. We acknowledge Syed Hamza Ahmed Jafri, BDS, MPH (University of Alberta), for his assistance with study selection, data extraction, and data analysis.

*

References 42, 44, 74, 91, 96, 104, 117, 131, 132.

References 54, 57, 59, 62, 64, 75, 77, 86, 98, 107, 113, 133, 138.

References 27, 49, 53, 57, 58, 60, 62, 63, 113, 114, 130, 133, 134, 148.

Footnotes

Declaration of Conflicting Interests: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: All authors have completed the Unified Competing Interest form at www.icmje.org/coi_disclosure.pdf (available on request from the corresponding author) and declared that they have no potential conflicts of interest with respect to research, authorship, and/or publication of this article.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This project was funded under contract No. HHSA29020150001I from the Agency for Healthcare Research and Quality (AHRQ), U.S. Department of Health and Human Services (HHS). The authors of this article are responsible for its content. Statements in the article do not necessarily represent the official views of or imply endorsement by AHRQ or HHS. Dr. Newton is supported by a New Investigator Award from the Canadian Institutes of Health Research.

Role of the Funding Source: This topic was nominated by the American Academy of Child and Adolescent Psychiatry and selected by AHRQ for systematic review by an EPC. A representative from AHRQ served as a contracting officer’s technical representative and provided technical assistance during the conduct of the full evidence report and comments on draft versions of the full evidence report. AHRQ did not directly participate in the literature search, determination of study eligibility criteria, data analysis or interpretation, or preparation, review, or approval of the manuscript for publication.

Systematic Review Registration: PROSPERO No. 42016032943

Supplemental Material: Supplemental material for this article is available online.

References

  • 1. Alessi-Severini S, Biscontri RG, Collins DM, et al. Ten years of antipsychotic prescribing to children: a Canadian population-based study. Can J Psychiatry. 2012;57(1):52–58. [DOI] [PubMed] [Google Scholar]
  • 2. Murphy AL, Gardner DM, Cooke C, et al. Prescribing trends of antipsychotics in youth receiving income assistance: results from a retrospective population database study. BMC Psychiatry. 2013;13:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Pringsheim T, Lam D, Patten SB. The pharmacoepidemiology of antipsychotic medications for Canadian children and adolescents: 2005-2009. J Child Adolesc Psychopharmacol. 2011;21(6):537–543. [DOI] [PubMed] [Google Scholar]
  • 4. Ronsley R, Scott D, Warburton WP, et al. A population-based study of antipsychotic prescription trends in children and adolescents in British Columbia, from 1996 to 2011. Can J Psychiatry. 2013;58(6):361–369. [DOI] [PubMed] [Google Scholar]
  • 5. Doey T, Handelman K, Seabrook JA, et al. Survey of atypical antipsychotic prescribing by Canadian child psychiatrists and developmental pediatricians for patients aged under 18 years. Can J Psychiatry. 2007;52(6):363–368. [DOI] [PubMed] [Google Scholar]
  • 6. Arora N, Knowles S, Gomes T, et al. Interprovincial variation in antipsychotic and antidepressant prescriptions dispensed in the Canadian pediatric population. Can J Psychiatry. 2016;61(12):758–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lachaine J, De G, Sikirica V, et al. Treatment patterns, resource use, and economic outcomes associated with atypical antipsychotic prescriptions in children and adolescents with attention-deficit hyperactivity disorder in quebec. Can J Psychiatry. 2014;59(11):597–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Gorman DA, Gardner DM, Murphy AL, et al. Canadian guidelines on pharmacotherapy for disruptive and aggressive behaviour in children and adolescents with attention-deficit hyperactivity disorder, oppositional defiant disorder, or conduct disorder. Can J Psychiatry. 2015;60(2):62–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Correll CU, Penzner JB, Parikh UH, et al. Recognizing and monitoring adverse events of second-generation antipsychotics in children and adolescents. Child Adolesc Psychiatr Clin N Am. 2006;15(1):177–206. [DOI] [PubMed] [Google Scholar]
  • 10. Pringsheim T, Lam D, Ching H, et al. Metabolic and neurological complications of second-generation antipsychotic use in children: a systematic review and meta-analysis of randomized controlled trials. Drug Safety. 2011;34(8):651–668. [DOI] [PubMed] [Google Scholar]
  • 11. Panagiotopoulos C, Ronsley R, Elbe D, et al. First do no harm: promoting an evidence-based approach to atypical antipsychotic use in children and adolescents. J Can Acad Child Adolesc Psychiatry. 2010;19(2):124–137. [PMC free article] [PubMed] [Google Scholar]
  • 12. Pringsheim T, Panagiotopoulos C, Davidson J, et al. Evidence-based recommendations for monitoring safety of second generation antipsychotics in children and youth. J Can Acad Child Adolesc Psychiatry. 2011;20(3):218–233. [PMC free article] [PubMed] [Google Scholar]
  • 13. Pringsheim T, Hirsch L, Gardner D, et al. The pharmacological management of oppositional behaviour, conduct problems, and aggression in children and adolescents with attention-deficit hyperactivity disorder, oppositional defiant disorder, and conduct disorder: a systematic review and meta-analysis. Part 2: antipsychotics and traditional mood stabilizers. Can J Psychiatry. 2015;60(2):52–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Seida JC, Schouten JR, Mousavi SS, et al. First- and second-generation antipsychotics for children and young adults. Comparative Effectiveness Review No. 39. (Prepared by the University of Alberta Evidence-based Practice Center under Contract No. 290-2007-10021.) AHRQ Publication No. 11(12)-EHC077-EF. Rockville, MD: Agency for Healthcare Research and Quality; February 2012. Available at: https://www.ncbi.nlm.nih.gov/books/NBK84643/ [Google Scholar]
  • 15. Agency for Healthcare Research and Quality. Methods guide for effectiveness and comparative effectiveness reviews. AHRQ Publication No. 10(14)-EHC063-EF. Rockville, MD: AHRQ; 2014. Available at https://www.ahrq.gov/research/findings/evidence-based-reports/technical/methodology/index.html [PubMed] [Google Scholar]
  • 16. Pillay JP, Boylan K, Carrey N, et al. First- and second-generation antipsychotics in children and young adults: systematic review update. Comparative Effectiveness Review No. 184. (Prepared by the University of Alberta Evidence-based Practice Center under Contract No. 290-2015-00001-I.) AHRQ Publication No. 17-EHC001-EF Rockville, MD: Agency for Healthcare Research and Quality; [cited March 2017] www.effectivehealthcare.ahrq.gov/reports/final/cfm. [PubMed] [Google Scholar]
  • 17. Golder S, Loke YK, Bland M. Meta-analyses of adverse effects data derived from randomised controlled trials as compared to observational studies: methodological overview. PLoS Med. 2011;8(5):e1001026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lu G, Ades AE. Combination of direct and indirect evidence in mixed treatment comparisons. Stat Med. 2004;23(20):3105–3124. [DOI] [PubMed] [Google Scholar]
  • 19. Lumley T. Network meta-analysis for indirect treatment comparisons. Stat Med. 2002;21(16):2313–2324. [DOI] [PubMed] [Google Scholar]
  • 20. Higgins JP, Green S. Section 8. Assessing the risk of bias of included studies. In: Cochrane handbook for systematic reviews of interventions: the Cochrane Collaroration; 2011. [cited 2011 March]. Available at: http://training.cochrane.org/handbook
  • 21. Wells G, Shea B, O’Connell N, et al. The Newcastle-Ottawa Scale for assessing the quality of nonrandomized studies in meta-analyses. Ottawa, ON: Department of Epidemiology and Community Medicine, University of Ottawa; 2009. Available at: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp [Google Scholar]
  • 22. Berkman ND, Lohr KN, Ansari MT, et al. Grading the strength of a body of evidence when assessing health care interventions: an EPC update. J Clin Epidemiol. 2015;68(11):1312–1324. [DOI] [PubMed] [Google Scholar]
  • 23. Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol. 2011;64(4):383–394. [DOI] [PubMed] [Google Scholar]
  • 24. Alacqua M, Trifiro G, Arcoraci V, et al. Use and tolerability of newer antipsychotics and antidepressants: a chart review in a paediatric setting. Pharm World Sci. 2008;30(1):44–50. [DOI] [PubMed] [Google Scholar]
  • 25. Aman MG, Binder C, Turgay A. Risperidone effects in the presence/absence of psychostimulant medicine in children with ADHD, other disruptive behavior disorders, and subaverage IQ. J Child Adolesc Psychopharmacology. 2004;14(2):243–524. [DOI] [PubMed] [Google Scholar]
  • 26. Aman MG, Bukstein OG, Gadow KD, et al. What does risperidone add to parent training and stimulant for severe aggression in child attention-deficit/hyperactivity disorder? J Am Acad Child Adolesc Psychiatry. 2014;53(1):47–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Aman MG, De SG, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry. 2002;159(8):1337–1346. [DOI] [PubMed] [Google Scholar]
  • 28. Aman MG, Hollway JA, Leone S, et al. Effects of risperidone on cognitive-motor performance and motor movements in chronically medicated children. Res Dev Disabil. 2009;30(2):386–396. [DOI] [PubMed] [Google Scholar]
  • 29. Aman MG, Marks RE, Turbott SH, et al. Clinical effects of methylphenidate and thioridazine in intellectually subaverage children. J Am Acad Child Adolesc Psychiatry. 1991;30(2):246–256. [DOI] [PubMed] [Google Scholar]
  • 30. Anderson LT, Campbell M, Adams P, et al. The effects of haloperidol on discrimination learning and behavioral symptoms in autistic children. J Autism Dev Disord. 1989;19(2):227–239. [DOI] [PubMed] [Google Scholar]
  • 31. Arango C, Giraldez M, Merchan-Naranjo J, et al. Second-generation antipsychotic use in children and adolescents: a six-month prospective cohort study in drug-naive patients. J Am Acad Child Adolesc Psychiatry. 2014;53(11):1179–1190. [DOI] [PubMed] [Google Scholar]
  • 32. Arango C, Robles O, Parellada M, et al. Olanzapine compared to quetiapine in adolescents with a first psychotic episode. Eur Child Adolesc Psychiatry. 2009;18(7):418–428. [DOI] [PubMed] [Google Scholar]
  • 33. Armenteros JI, Lewis JE, Davalos M. Risperidone augmentation for treatment-resistant aggression in attention-deficit/hyperactivity disorder: a placebo-controlled pilot study. J Am Acad Child Adolesc Psychiatry. 2007;46(5):558–565. [DOI] [PubMed] [Google Scholar]
  • 34. Bastiaens L. A non-randomized, open study with aripiprazole and ziprasidone for the treatment of aggressive behavior in youth in a community clinic. Community Ment Health J. 2009;45(1):73–77. [DOI] [PubMed] [Google Scholar]
  • 35. Berger GE, Proffitt TM, McConchie M, et al. Dosing quetiapine in drug-naive first-episode psychosis: a controlled, double-blind, randomized, single-center study investigating efficacy, tolerability, and safety of 200 mg/day vs. 400 mg/day of quetiapine fumarate in 141 patients aged 15 to 25 years. J Clin Psychiatry. 2008;69(11):1702–1714. [PubMed] [Google Scholar]
  • 36. Biederman J, Mick E, Hammerness P, et al. Open-label, 8-week trial of olanzapine and risperidone for the treatment of bipolar disorder in preschool-age children. Biol Psychiatry. 2005;58(7):589–594. [DOI] [PubMed] [Google Scholar]
  • 37. Bobo WV, Cooper WO, Stein CM, et al. Antipsychotics and the risk of type 2 diabetes mellitus in children and youth. JAMA Psychiatry. 2013;70(10):1067–1075. [DOI] [PubMed] [Google Scholar]
  • 38. Bruggeman R, van der LC, Buitelaar JK, et al. Risperidone versus pimozide in Tourette’s disorder: a comparative double-blind parallel-group study. J Clin Psychiatry. 2001;62(1):50–46. [DOI] [PubMed] [Google Scholar]
  • 39. Buitelaar JK, van der Gaag RJ, Cohen-Kettenis P, et al. A randomized controlled trial of risperidone in the treatment of aggression in hospitalized adolescents with subaverage cognitive abilities. J Clin Psychiatry. 2001;62(4):239–248. [DOI] [PubMed] [Google Scholar]
  • 40. Calarge CA, Nicol G, Schlechte JA, et al. Cardiometabolic outcomes in children and adolescents following discontinuation of long-term risperidone treatment. J Child Adolesc Psychopharmacol. 2014;24(3):120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Castro-Fornieles J, Parellada M, Soutullo CA, et al. Antipsychotic treatment in child and adolescent first-episode psychosis: a longitudinal naturalistic approach. J Child Adoles Psychopharmacol. 2008;18(4):327–336. [DOI] [PubMed] [Google Scholar]
  • 42. Cianchetti C, Ledda MG. Effectiveness and safety of antipsychotics in early onset psychoses: a long-term comparison. Psychiatry Res. 2011;189(3):349–356. [DOI] [PubMed] [Google Scholar]
  • 43. Connor DF, McLaughlin TJ, Jeffers-Terry M. Randomized controlled pilot study of quetiapine in the treatment of adolescent conduct disorder. J Child Adoles Psychopharmacol. 2008;18(2):140–156. [DOI] [PubMed] [Google Scholar]
  • 44. Conus P, Berk M, Cotton SM, et al. Olanzapine or chlorpromazine plus lithium in first episode psychotic mania: an 8-week randomised controlled trial. Eur Psychiatry. 2015;30(8):975–982. [DOI] [PubMed] [Google Scholar]
  • 45. Correll CU, Manu P, Olshanskiy V, et al. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302(16):1765–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Crocq MA, Guillon MS, Bailey PE, et al. Orally disintegrating olanzapine induces less weight gain in adolescents than standard oral tablets. Eur Psychiatry. 2007;22(7):453–454. [DOI] [PubMed] [Google Scholar]
  • 47. Cuerda C, Merchan-Naranjo J, Velasco C, et al. Influence of resting energy expenditure on weight gain in adolescents taking second-generation antipsychotics. Clin Nutr. 2011;30(5):616–623. [DOI] [PubMed] [Google Scholar]
  • 48. deHaan L, van Bruggen M, Lavalaye J, et al. Subjective experience and D2 receptor occupancy in patients with recent-onset schizophrenia treated with low-dose olanzapine or haloperidol: a randomized, double-blind study. Am J Psychiatry. 2003;160(2):303–309. [DOI] [PubMed] [Google Scholar]
  • 49. DelBello MP, Chang K, Welge JA, et al. A double-blind, placebo-controlled pilot study of quetiapine for depressed adolescents with bipolar disorder. Bipolar Disord. 2009;11(5):483–493. [DOI] [PubMed] [Google Scholar]
  • 50. DelBello MP, Schwiers ML, Rosenberg HL, et al. A double-blind, randomized, placebo-controlled study of quetiapine as adjunctive treatment for adolescent mania. J Am Acad Child Adoles Psychiatry. 2002;41(10):1216–1223. [DOI] [PubMed] [Google Scholar]
  • 51. DelBello MP, Versavel M, Ice K, et al. Tolerability of oral ziprasidone in children and adolescents with bipolar mania, schizophrenia, or schizoaffective disorder. J Child Adoles Psychopharmacology. 2008;18(5):491–499. [DOI] [PubMed] [Google Scholar]
  • 52. Ebert T, Midbari Y, Shmilovitz R, et al. Metabolic effects of antipsychotics in prepubertal children: a retrospective chart review. J Child Adoles Psychopharmacology. 2014;24(4):218–222. [DOI] [PubMed] [Google Scholar]
  • 53. Findling RL, Cavus I, Pappadopulos E, et al. Ziprasidone in adolescents with schizophrenia: results from a placebo-controlled efficacy and long-term open-extension study. J Child Adolesc Psychopharmacol. 2013;23(8):531–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Findling RL, Cavus I, Pappadopulos E, et al. Efficacy, long-term safety, and tolerability of ziprasidone in children and adolescents with bipolar disorder. J Child Adolesc Psychopharmacol. 2013;23(8):545–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Findling RL, Goldman R, Chiu YY, et al. Pharmacokinetics and tolerability of lurasidone in children and adolescents with psychiatric disorders. Clin Ther. 2015;37(12):2788–2797. [DOI] [PubMed] [Google Scholar]
  • 56. Findling RL, Kauffman RE, Sallee FR, et al. Tolerability and pharmacokinetics of aripiprazole in children and adolescents with psychiatric disorders: an open-label, dose-escalation study. J Clin Psychopharmacology. 2008;28(4):441–446. [DOI] [PubMed] [Google Scholar]
  • 57. Findling RL, Landbloom RL, Szegedi A, et al. Asenapine for the acute treatment of pediatric manic or mixed episode of bipolar I disorder. J Am Acad Child Adolesc Psychiatry. 2015;54(12):1032–1041. [DOI] [PubMed] [Google Scholar]
  • 58. Findling RL, Landbloom RP, Mackle M, et al. Safety and efficacy from an 8 week double-blind trial and a 26 week open-label extension of asenapine in adolescents with schizophrenia. J Child Adoles Psychopharmacology. 2015;25(5):384–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Findling RL, Mankoski R, Timko K, et al. A randomized controlled trial investigating the safety and efficacy of aripiprazole in the long-term maintenance treatment of pediatric patients with irritability associated with autistic disorder. J Clin Psychiatry. 2014;75(1):22–30. [DOI] [PubMed] [Google Scholar]
  • 60. Findling RL, McKenna K, Earley WR, et al. Efficacy and safety of quetiapine in adolescents with schizophrenia investigated in a 6-week, double-blind, placebo-controlled trial. J Child Adolesc Psychopharmacol. 2012;22(5):327–342. [DOI] [PubMed] [Google Scholar]
  • 61. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry. 2000;39(4):509–516. [DOI] [PubMed] [Google Scholar]
  • 62. Findling RL, Nyilas M, Forbes RA, et al. Acute treatment of pediatric bipolar I disorder, manic or mixed episode, with aripiprazole: a randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2009;70(10):1441–1451. [DOI] [PubMed] [Google Scholar]
  • 63. Findling RL, Pathak S, Earley WR, et al. Efficacy and safety of extended-release quetiapine fumarate in youth with bipolar depression: an 8 week, double-blind, placebo-controlled trial. J Child Adoles Psychopharmacology. 2014;24(6):325–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Findling RL, Robb A, Nyilas M, et al. A multiple-center, randomized, double-blind, placebo-controlled study of oral aripiprazole for treatment of adolescents with schizophrenia. Am J Psychiatry. 2008;165(11):1432–1441. [DOI] [PubMed] [Google Scholar]
  • 65. Findling RL, Youngstrom EA, McNamara NK, et al. Double-blind, randomized, placebo-controlled long-term maintenance study of aripiprazole in children with bipolar disorder. J Clin Psychiatry. 2012;73(1):57–63. [DOI] [PubMed] [Google Scholar]
  • 66. Fleischhaker C, Heiser P, Hennighausen K, et al. Clinical drug monitoring in child and adolescent psychiatry: side effects of atypical neuroleptics. J Child Adolesc Psychopharmacol. 2006;16(3):308–316. [DOI] [PubMed] [Google Scholar]
  • 67. Fraguas D, Merchan-Naranjo J, Laita P, et al. Metabolic and hormonal side effects in children and adolescents treated with second-generation antipsychotics. J Clin Psychiatry. 2008;69(7):1166–1175. [DOI] [PubMed] [Google Scholar]
  • 68. Friedlander R, Lazar S, Klancnik J. Atypical antipsychotic use in treating adolescents and young adults with developmental disabilities. Can J Psychiatry. 2001;46(8):741–745. [DOI] [PubMed] [Google Scholar]
  • 69. Germano E, Italiano D, Lamberti M, et al. ECG parameters in children and adolescents treated with aripiprazole and risperidone. Prog Neuropsychopharmacol Biol Psychiatry. 2014;51:23–27. [DOI] [PubMed] [Google Scholar]
  • 70. Ghanizadeh A, Haghighi A. Aripiprazole versus risperidone for treating children and adolescents with tic disorder: a randomized double blind clinical trial. Child Psychiatry Hum Dev. 2014;45(5):596–603. [DOI] [PubMed] [Google Scholar]
  • 71. Ghanizadeh A, Sahraeizadeh A, Berk M. A head-to-head comparison of aripiprazole and risperidone for safety and treating autistic disorders, a randomized double blind clinical trial. Child Psychiatry Hum Dev. 2014;45(2):185–192. [DOI] [PubMed] [Google Scholar]
  • 72. Gilbert DL, Batterson JR, Sethuraman G, et al. Tic reduction with risperidone versus pimozide in a randomized, double-blind, crossover trial. J Am Acad Child Adolesc Psychiatry. 2004;43(2):206–214. [DOI] [PubMed] [Google Scholar]
  • 73. Gothelf D, Falk B, Singer P, et al. Weight gain associated with increased food intake and low habitual activity levels in male adolescent schizophrenic inpatients treated with olanzapine. Am J Psychiatry. 2002;159(6):1055–1057. [DOI] [PubMed] [Google Scholar]
  • 74. Gulisano M, Cali PV, Cavanna AE, et al. Cardiovascular safety of aripiprazole and pimozide in young patients with Tourette syndrome. Neurol Sci. 2011;32(6):1213–1217. [DOI] [PubMed] [Google Scholar]
  • 75. Haas M, Delbello MP, Pandina G, et al. Risperidone for the treatment of acute mania in children and adolescents with bipolar disorder: a randomized, double-blind, placebo-controlled study. Bipolar Disord. 2009;11(7):687–700. [DOI] [PubMed] [Google Scholar]
  • 76. Haas M, Eerdekens M, Kushner S, et al. Efficacy, safety and tolerability of two dosing regimens in adolescent schizophrenia: double-blind study. Br J Psychiatry. 2009;194(2):158–164. [DOI] [PubMed] [Google Scholar]
  • 77. Haas M, Unis AS, Armenteros J, et al. A 6-week, randomized, double-blind, placebo-controlled study of the efficacy and safety of risperidone in adolescents with schizophrenia. J Child Adolesc Psychopharmacol. 2009;19(6):611–621. [DOI] [PubMed] [Google Scholar]
  • 78. Hagman J, Gralla J, Sigel E, et al. A double-blind, placebo-controlled study of risperidone for the treatment of adolescents and young adults with anorexia nervosa: a pilot study. J Am Acad Child Adolesc Psychiatry. 2011;50(9):915–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Hellings JA, Zarcone JR, Reese RM, et al. A crossover study of risperidone in children, adolescents and adults with mental retardation. J Autism Dev Disord. 2006;36(3):401–411. [DOI] [PubMed] [Google Scholar]
  • 80. Hollander E, Wasserman S, Swanson EN, et al. A double-blind placebo-controlled pilot study of olanzapine in childhood/adolescent pervasive developmental disorder. J Child Adolesc Psychopharmacology. 2006;16(5):541–548. [DOI] [PubMed] [Google Scholar]
  • 81. Hrdlicka M, Zedkova I, Blatny M, et al. Weight gain associated with atypical and typical antipsychotics during treatment of adolescent schizophrenic psychoses: a retrospective study. Neuro Endocrinol Lett. 2009;30(2):256–261. [PubMed] [Google Scholar]
  • 82. Jensen JB, Kumra S, Leitten W, et al. A comparative pilot study of second-generation antipsychotics in children and adolescents with schizophrenia-spectrum disorders. J Child Adolesc Psychopharmacology. 2008;18(4):317–326. [DOI] [PubMed] [Google Scholar]
  • 83. Jerrell JM, McIntyre RS. Adverse events in children and adolescents treated with antipsychotic medications. Human Psychopharmacol. 2008;23(4):283–290. [DOI] [PubMed] [Google Scholar]
  • 84. Johnson & Johnson Pharmaceutical Research & Development. Open-label study to evaluate the safety and pharmacokinetics of single- and multiple-dose extended-release paliperidone in pediatric subjects (10 to 17 years of age) with schizophrenia, schizoaffective disorder, or schizophreniform disorder. (Clinical Study Report) [cited 2007 January 29]. Available at: https://clinicaltrials.gov/ct2/show/NCT00796081
  • 85. Kafantaris V, Leigh E, Hertz S, et al. A placebo-controlled pilot study of adjunctive olanzapine for adolescents with anorexia nervosa. J Child Adolesc Psychopharmacology. 2011;21(3):207–212. [DOI] [PubMed] [Google Scholar]
  • 86. Kent JM, Kushner S, Ning X, et al. Risperidone dosing in children and adolescents with autistic disorder: a double-blind, placebo-controlled study. J Autism Dev Disord. 2013;43(8):1773–1783. [DOI] [PubMed] [Google Scholar]
  • 87. Khan RA, Mican LM, Suehs BT. Effects of olanzapine and risperidone on metabolic factors in children and adolescents: a retrospective evaluation. J Psychiatr Pract. 2009;15(4):320–328. [DOI] [PubMed] [Google Scholar]
  • 88. Khan SS, Mican LM. A naturalistic evaluation of intramuscular ziprasidone versus intramuscular olanzapine for the management of acute agitation and aggression in children and adolescents. J Child Adoles Psychopharmacol. 2006;16(6):671–677. [DOI] [PubMed] [Google Scholar]
  • 89. Kowatch RA, Scheffer RE, Monroe E, et al. Placebo-controlled trial of valproic Acid versus risperidone in children 3-7 years of age with bipolar I disorder. J Child Adoles Psychopharmacol. 2015;25(4):306–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Kryzhanovskaya L, Schulz SC, McDougle C, et al. Olanzapine versus placebo in adolescents with schizophrenia: a 6-week, randomized, double-blind, placebo-controlled trial. J Am Acad Child Adolesc Psychiatry. 2009;48(1):60–70. [DOI] [PubMed] [Google Scholar]
  • 91. Kumra S, Frazier JA, Jacobsen LK, et al. Childhood-onset schizophrenia: a double-blind clozapine-haloperidol comparison. Arch Gen Psychiatry. 1996;53(12):1090–1097. [DOI] [PubMed] [Google Scholar]
  • 92. Kumra S, Jacobsen LK, Lenane M, et al. Childhood-onset schizophrenia: an open-label study of olanzapine in adolescents. J Am Acad Child Adoles Psychiatry. 1998;37(4):377–385. [DOI] [PubMed] [Google Scholar]
  • 93. Kumra S, Kranzler H, Gerbino-Rosen G, et al. Clozapine and “high-dose” olanzapine in refractory early-onset schizophrenia: a 12-week randomized and double-blind comparison. Biol Psychiatry. 2008;63(5):524–529. [DOI] [PubMed] [Google Scholar]
  • 94. Loebel A, Brams M, Goldman RS, et al. Lurasidone for the treatment of irritability associated with autistic disorder. J Autism Dev Disord. 2016;46(4):1153–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Luby J, Mrakotsky C, Stalets MM, et al. Risperidone in preschool children with autistic spectrum disorders: an investigation of safety and efficacy. J Child Adolesc Psychopharmacol. 2006;16(5):575–587. [DOI] [PubMed] [Google Scholar]
  • 96. Malone RP, Cater J, Sheikh RM, et al. Olanzapine versus haloperidol in children with autistic disorder: an open pilot study. J Am Acad Child Adolesc Psychiatry. 2001;40(8):887–894. [DOI] [PubMed] [Google Scholar]
  • 97. Mankoski R, Stockton G, Manos G, et al. Aripiprazole treatment of irritability associated with autistic disorder and the relationship between prior antipsychotic exposure, adverse events, and weight change. J Child Adolesc Psychopharmacol. 2013;23(8):572–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Marcus RN, Owen R, Kamen L, et al. A placebo-controlled, fixed-dose study of aripiprazole in children and adolescents with irritability associated with autistic disorder. J Am Acad Child Adolesc Psychiatry. 2009;48(11):1110–1119. [DOI] [PubMed] [Google Scholar]
  • 99. Martin A, Landau J, Leebens P, et al. Risperidone-associated weight gain in children and adolescents: a retrospective chart review. J Child Adolesc Psychopharmacol. 2000;10(4):259–268. [DOI] [PubMed] [Google Scholar]
  • 100. Masi G, Milone A, Stawinoga A, et al. Efficacy and safety of risperidone and quetiapine in adolescents with bipolar II disorder comorbid with conduct disorder. J Clin Psychopharmacol. 2015;35(5):587–590. [DOI] [PubMed] [Google Scholar]
  • 101. Masi G, Pfanner C, Brovedani P. Antipsychotic augmentation of selective serotonin reuptake inhibitors in resistant tic-related obsessive-compulsive disorder in children and adolescents: a naturalistic comparative study. J Psychiatr Res. 2013;47(8):1007–1012. [DOI] [PubMed] [Google Scholar]
  • 102. McCracken JT, McGough J, Shah B, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med. 2002;347(5):314–321. [DOI] [PubMed] [Google Scholar]
  • 103. Migliardi G, Spina E, D’Arrigo C, et al. Short- and long-term effects on prolactin of risperidone and olanzapine treatments in children and adolescents. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(8):1496–1501. [DOI] [PubMed] [Google Scholar]
  • 104. Miral S, Gencer O, Inal-Emiroglu FN, et al. Risperidone versus haloperidol in children and adolescents with AD: a randomized, controlled, double-blind trial. Eur Child Adolesc Psychiatry. 2008;17(1):1–8. [DOI] [PubMed] [Google Scholar]
  • 105. Mozes T, Ebert T, Michal SE, et al. An open-label randomized comparison of olanzapine versus risperidone in the treatment of childhood-onset schizophrenia. J Child Adoles Psychopharmacol. 2006;16(4):393–403. [DOI] [PubMed] [Google Scholar]
  • 106. Nagaraj R, Singhi P, Malhi P. Risperidone in children with autism: randomized, placebo-controlled, double-blind study. J Child Neurol. 2006;21(6):450–455. [DOI] [PubMed] [Google Scholar]
  • 107. NCT00194012. Study of aripiprazole (Abilify) versus placebo in children with subsyndromal bipolar disorder [cited 2014 May 5]. Available at: https://clinicaltrials.gov/ct2/show/NCT00194012
  • 108. NCT00619190. Study of aripiprazole to treat children and adolescents with autism [cited 2014 March 26]. Available at: https://clinicaltrials.gov/ct2/show/NCT00619190
  • 109. NCT01149655. Efficacy & safety study of oral aripiprazole in adolescents with schizophrenia [cited 2015 April 2]. Available at: https://clinicaltrials.gov/ct2/show/NCT01149655
  • 110. Norris ML, Spettigue W, Buchholz A, et al. Olanzapine use for the adjunctive treatment of adolescents with anorexia nervosa. J Child Adoles Psychopharmacol. 2011;21(3):213–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. O’Donoghue B, Schafer MR, Becker J, et al. Metabolic changes in first-episode early-onset schizophrenia with second-generation antipsychotics. Early Interv Psychiatry. 2014;8(3):276–280. [DOI] [PubMed] [Google Scholar]
  • 112. Oh J, Chang JG, Lee SB, et al. Comparison of aripiprazole and other atypical antipsychotics for pediatric bipolar disorder: a retrospective chart review of efficacy and tolerability. Clin Psychopharmacol Neurosci. 2013;11(2):72–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Owen R, Sikich L, Marcus RN, et al. Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics. 2009;124(6):1533–1540. [DOI] [PubMed] [Google Scholar]
  • 114. Pathak S, Findling RL, Earley WR, et al. Efficacy and safety of quetiapine in children and adolescents with mania associated with bipolar I disorder: a 3-week, double-blind, placebo-controlled trial. J Clin Psychiatry. 2013;74(1):e100–e109. [DOI] [PubMed] [Google Scholar]
  • 115. Perry R, Campbell M, Adams P, et al. Long-term efficacy of haloperidol in autistic children: continuous versus discontinuous drug administration. J Am Acad Child Adoles Psychiatry. 1989;28(1):87–92. [DOI] [PubMed] [Google Scholar]
  • 116. Pogge DL, Singer MB, Harvey PD. Rates and predictors of adherence with atypical antipsychotic medication: a follow-up study of adolescent inpatients. J Child Adoles Psychopharmacol. 2005;15(6):901–912. [DOI] [PubMed] [Google Scholar]
  • 117. Ratzoni G, Gothelf D, Brand-Gothelf A, et al. Weight gain associated with olanzapine and risperidone in adolescent patients: a comparative prospective study. J Am Acad Child Adolesc Psychiatry. 2002;41(3):337–343. [DOI] [PubMed] [Google Scholar]
  • 118. Remington G, Sloman L, Konstantareas M, et al. Clomipramine versus haloperidol in the treatment of autistic disorder: a double-blind, placebo-controlled, crossover study. J Clin Psychopharmacol. 2001;21(4):440–444. [DOI] [PubMed] [Google Scholar]
  • 119. Research Units on Pediatric Psychopharmacology Autism Network. Risperidone treatment of autistic disorder: longer-term benefits and blinded discontinuation after 6 months. Am J Psychiatry. 2005;162(7):1361–1369. [DOI] [PubMed] [Google Scholar]
  • 120. Reyes M, Buitelaar J, Toren P, et al. A randomized, double-blind, placebo-controlled study of risperidone maintenance treatment in children and adolescents with disruptive behavior disorders. Am J Psychiatry. 2006;163(3):402–410. [DOI] [PubMed] [Google Scholar]
  • 121. Rizzo R, Eddy CM, Cali P, et al. Metabolic effects of aripiprazole and pimozide in children with Tourette syndrome. Pediatr Neurol. 2012;47(6):419–422. [DOI] [PubMed] [Google Scholar]
  • 122. Ronsley R, Nguyen D, Davidson J, et al. Increased risk of obesity and metabolic dysregulation following 12 months of second-generation antipsychotic treatment in children: a prospective cohort study. Can J Psychiatry. 2015;60(10):441–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Saito E, Correll CU, Gallelli K, et al. A prospective study of hyperprolactinemia in children and adolescents treated with atypical antipsychotic agents. J Child Adolesc Psychopharmacol. 2004;14(3):350–358. [DOI] [PubMed] [Google Scholar]
  • 124. Sallee FR, Kurlan R, Goetz CG, et al. Ziprasidone treatment of children and adolescents with Tourette’s syndrome: a pilot study. J Am Acad Child Adolesc Psychiatry. 2000;39(3):292–299. [DOI] [PubMed] [Google Scholar]
  • 125. Sallee FR, Nesbitt L, Jackson C, et al. Relative efficacy of haloperidol and pimozide in children and adolescents with Tourette’s disorder. Am J Psychiatry. 1997;154(8):1057–1062. [DOI] [PubMed] [Google Scholar]
  • 126. Savitz AJ, Lane R, Nuamah I, et al. Efficacy and safety of paliperidone extended release in adolescents with schizophrenia: a randomized, double-blind study. J Am Acad Child Adolesc Psychiatry. 2015;54(2):126–137. [DOI] [PubMed] [Google Scholar]
  • 127. Scahill L, Leckman JF, Schultz RT, et al. A placebo-controlled trial of risperidone in Tourette syndrome. Neurology. 2003;60(7):1130–1135. [DOI] [PubMed] [Google Scholar]
  • 128. Sehgal N. Short-term versus longer term pimozide therapy in Tourette’s syndrome: a preliminary study. Neurology. 1999;52(4):874–877. [DOI] [PubMed] [Google Scholar]
  • 129. Shaw P, Sporn A, Gogtay N, et al. Childhood-onset schizophrenia: a double-blind, randomized clozapine-olanzapine comparison. Arch Gen Psychiatry. 2006;63(7):721–730. [DOI] [PubMed] [Google Scholar]
  • 130. Shea S, Turgay A, Carroll A, et al. Risperidone in the treatment of disruptive behavioral symptoms in children with autistic and other pervasive developmental disorders. Pediatrics. 2004;114(5):e634–e641. [DOI] [PubMed] [Google Scholar]
  • 131. Sikich L, Frazier JA, McClellan J, et al. Double-blind comparison of first- and second-generation antipsychotics in early-onset schizophrenia and schizo-affective disorder: findings from the treatment of early-onset schizophrenia spectrum disorders (TEOSS) study. Am J Psychiatry. 2008;165(11):1420–1431. [DOI] [PubMed] [Google Scholar]
  • 132. Sikich L, Hamer RM, Bashford RA, et al. A pilot study of risperidone, olanzapine, and haloperidol in psychotic youth: a double-blind, randomized, 8-week trial. Neuropsychopharmacol. 2004;29(1):133–145. [DOI] [PubMed] [Google Scholar]
  • 133. Singh J, Robb A, Vijapurkar U, et al. A randomized, double-blind study of paliperidone extended-release in treatment of acute schizophrenia in adolescents. Biol Psychiatry. 2011;70(12):1179–1187. [DOI] [PubMed] [Google Scholar]
  • 134. Snyder R, Turgay A, Aman M, et al. Effects of risperidone on conduct and disruptive behavior disorders in children with subaverage IQs. J Am Acad Child Adolesc Psychiatry. 2002;41(9):1026–1036. [DOI] [PubMed] [Google Scholar]
  • 135. Spencer EK, Campbell M. Children with schizophrenia: diagnosis, phenomenology, and pharmacotherapy. Schizophr Bull. 1994;20(4):713–725. [DOI] [PubMed] [Google Scholar]
  • 136. Stocks JD, Taneja BK, Baroldi P, et al. A phase 2a randomized, parallel group, dose-ranging study of molindone in children with attention-deficit/hyperactivity disorder and persistent, serious conduct problems. J Child Adolesc Psychopharmacol. 2012;22(2):102–111. [DOI] [PubMed] [Google Scholar]
  • 137. Swadi HS, Craig BJ, Pirwani NZ, et al. A trial of quetiapine compared with risperidone in the treatment of first onset psychosis among 15-to 18-year-old adolescents. Int Clin Psychopharmacol. 2010;25(1):1–6. [DOI] [PubMed] [Google Scholar]
  • 138. Tohen M, Kryzhanovskaya L, Carlson G, et al. Olanzapine versus placebo in the treatment of adolescents with bipolar mania. Am J Psychiatry. 2007;164(10):1547–1556. [DOI] [PubMed] [Google Scholar]
  • 139. Tramontina S, Zeni CP, Ketzer CR, et al. Aripiprazole in children and adolescents with bipolar disorder comorbid with attention-deficit/hyperactivity disorder: a pilot randomized clinical trial. J Clin Psychiatry. 2009;70(5):756–764. [DOI] [PubMed] [Google Scholar]
  • 140. Troost PW, Lahuis BE, Steenhuis MP, et al. Long-term effects of risperidone in children with autism spectrum disorders: a placebo discontinuation study. J Am Acad Child Adolesc Psychiatry. 2005;44(11):1137–1144. [DOI] [PubMed] [Google Scholar]
  • 141. Van Bellinghen M, De Troch C. Risperidone in the treatment of behavioral disturbances in children and adolescents with borderline intellectual functioning: a double-blind, placebo-controlled pilot trial. J Child Adolesc Psychopharmacol. 2001;11(1):5–13. [DOI] [PubMed] [Google Scholar]
  • 142. van Bruggen J, Tijssen J, Dingemans P, et al. Symptom response and side-effects of olanzapine and risperidone in young adults with recent onset schizophrenia. Int Clin Psychopharmacol. 2003;18(6):341–346. [DOI] [PubMed] [Google Scholar]
  • 143. Wink LK, Early M, Schaefer T, et al. Body mass index change in autism spectrum disorders: comparison of treatment with risperidone and aripiprazole. J Child Adolesc Psychopharmacol. 2014;24(2):78–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Wonodi I, Reeves G, Carmichael D, et al. Tardive dyskinesia in children treated with atypical antipsychotic medications. Mov Disord. 2007;22(12):1777–1782. [DOI] [PubMed] [Google Scholar]
  • 145. Woods SW, Breier A, Zipursky RB, et al. Randomized trial of olanzapine versus placebo in the symptomatic acute treatment of the schizophrenic prodrome. Biol Psychiatry. 2003;54(4):453–464. [DOI] [PubMed] [Google Scholar]
  • 146. Wudarsky M, Nicolson R, Hamburger SD, et al. Elevated prolactin in pediatric patients on typical and atypical antipsychotics. J Child Adolesc Psychopharmacol. 1999(4):239–245. [DOI] [PubMed] [Google Scholar]
  • 147. Yen YC, Lung F-W, Chong MY. Adverse effects of risperidone and haloperidol treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28(2):285–290. [DOI] [PubMed] [Google Scholar]
  • 148. Yoo HK, Joung YS, Lee JS, et al. A multicenter, randomized, double-blind, placebo-controlled study of aripiprazole in children and adolescents with Tourette’s disorder. J Clin Psychiatry. 2013;74(8):e772–e780. [DOI] [PubMed] [Google Scholar]
  • 149. Yoo HK, Lee JS, Paik KW, et al. Open-label study comparing the efficacy and tolerability of aripiprazole and haloperidol in the treatment of pediatric tic disorders. Eur Child Adolesc Psychiatry. 2011;20(3):127–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Almandil NB, Liu Y, Murray ML, et al. Weight gain and other metabolic adverse effects associated with atypical antipsychotic treatment of children and adolescents: a systematic review and meta-analysis. Paediatr Drugs. 2013;15(2):139–150. [DOI] [PubMed] [Google Scholar]
  • 151. Cohen D, Bonnot O, Bodeau N, et al. Adverse effects of second-generation antipsychotics in children and adolescents: a Bayesian meta-analysis. J Clin Psychopharmacol. 2012;32(3):309–316. [DOI] [PubMed] [Google Scholar]
  • 152. Fraguas D, Correll CU, Merchan-Naranjo J, et al. Efficacy and safety of second-generation antipsychotics in children and adolescents with psychotic and bipolar spectrum disorders: comprehensive review of prospective head-to-head and placebo-controlled comparisons. Eur Neuropsychopharmacol. 2011;21(8):621–645. [DOI] [PubMed] [Google Scholar]
  • 153. Maayan L, Correll CU. Weight gain and metabolic risks associated with antipsychotic medications in children and adolescents. J Child Adolesc Psychopharmacol. 2011;21(6):517–535. [DOI] [PubMed] [Google Scholar]
  • 154. Pringsheim T, Lam D, Ching H, et al. Metabolic and neurological complications of second-generation antipsychotic use in children: a systematic review and meta-analysis of randomized controlled trials. Drug Saf. 2011;34(8):651–668. [DOI] [PubMed] [Google Scholar]
  • 155. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. J Clin Psychiatry. 2004;65(2):267–272. [DOI] [PubMed] [Google Scholar]
  • 156. Birnbaum ML, Saito E, Gerhard T, et al. Pharmacoepidemiology of antipsychotic use in youth with ADHD: trends and clinical implications. Curr Psychiatry Rep. 2013;15(8):382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Rodday AM, Parsons SK, Mankiw C, et al. Child and adolescent psychiatrists’ reported monitoring behaviors for second-generation antipsychotics. J Child Adolesc Psychopharmacol. 2015;25(4):351–361. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

779950_Supplementary_material - Harms of Antipsychotics in Children and Young Adults: A Systematic Review Update

779950_Supplementary_material for Harms of Antipsychotics in Children and Young Adults: A Systematic Review Update by Jennifer Pillay, Khrista Boylan, Amanda Newton, Lisa Hartling, Ben Vandermeer, Megan Nuspl, Tara MacGregor, Robin Featherstone, and Normand Carrey in The Canadian Journal of Psychiatry


Articles from Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie are provided here courtesy of SAGE Publications

RESOURCES