Abstract
Objective:
Second-generation antipsychotic (SGA) effects in youth were monitored to quantify extrapyramidal side effects (EPS) and to identify risk profiles for treatment-emergent EPS.
Method:
Data were analyzed for the nonrandomized, prospective Second-generation Antipsychotic Treatment Indications, Effectiveness and Tolerability in Youth (SATIETY) inception cohort study. EPS were assessed at baseline and 4, 8, and 12 weeks after naturalistic SGA initiation for schizophrenia, mood, disruptive behavior, and autism spectrum disorders using the Simpson–Angus Scale (SAS), Barnes Akathisia Scale, Abnormal Involuntary Movement Scale (AIMS), and Treatment Emergent Side Effect Scale. Drug-induced parkinsonism was defined by incident mean SAS score >0.33, anticholinergic initiation, or increasing total SAS score ≥2 in patients with baseline EPS.
Results:
In 342 youth aged 13.6 ± 3.5 years (male = 58.2%, antipsychotic-naive = 65.8%), 15.2% developed drug-induced parkinsonism. Raw SGA-grouped drug-induced parkinsonism rates were as follows: quetiapine = 1.5%, olanzapine = 13.8%, risperidone = 16.1%, ziprasidone = 20.0%, and aripiprazole = 27.3%. SGA type, dose, higher age, and lower baseline functioning were jointly associated with drug-induced parkinsonism (R2 = 0.18; p < .0001). Controlling for these factors, drug-induced parkinsonism rates were significantly lower only for quetiapine and olanzapine. Subjectively reported EPS (5%), EPS-related treatment discontinuation (3.3%), and anticholinergic initiation (3%) were infrequent. Anticholinergic initiation was most frequent with risperidone (10.2%; p = .0004). Treatment-emergent dyskinesia ranged from 4.5% (aripiprazole) to 15.5% (olanzapine). SGA type, younger age, white race/ethnicity, and baseline AIMS were jointly associated with treatment-emergent dyskinesia (R2 = 0.31; p < .0001). Controlling for these factors, treatment-emergent dyskinesia rates differed among SGA subgroups, with higher rates with olanzapine and ziprasidone. At baseline, psychostimulant use was associated with dyskinesia, and number of psychotropic comedications was associated with subjective EPS.
Conclusion:
In youth, SGA-related EPS rates did not generally exceed those reported in adults, with particularly low rates with quetiapine and olanzapine.
Keywords: extrapyramidal, parkinsonism, dyskinesia, second-generation antipsychotic, youth
Second-generation antipsychotics (SGAs) have initially been greeted with enthusiasm due to their reduced risk of causing extrapyramidal side effects (EPS). Subsequently, the recognition of cardiometabolic risks has rightfully shifted safety concerns.1 Nonetheless, EPS contribute to tolerability profile, treatment nonadherence,2 and poor outcome.3
Incidence rates of EPS in youth are expected to exceed those in adults.4 Meta-analyses reported a 22% EPS rate for adolescents receiving SGAs for psychosis,5 or 23%/25% for risperidone/aripiprazole,6 but reported rates vary largely depending on the SGA, study design, population, and methodology used to assess EPS.7 For aripiprazole, subject-/investigator-reported rates in randomized controlled trials (RCTs) ranged from 15%8 to 39%.9,10 For olanzapine, Simpson–Angus Scale (SAS)–based EPS rates varied from 0%11to 58%.12 Similar divergence exists for patient-/investigator-reported rates with quetiapine (0%13–46%14). Dose-dependent EPS rates have been demonstrated for risperidone (patient/investigator report: 0% at ≤0.6 mg15 to 25% at ≥6 mg,15,16 or SAS-based 0%17–51%12). Few data have been published on EPS rates for ziprasidone (11%18–18%19). Rates of anticholinergic prescriptions vary between 12% for olanzapine20 or across SGAs21 to 32% for risperidone.20 These neuromotor side effect rates were gathered as part of RCTs or in cross-sectional studies, but prospective rates in psychiatric routine care with different SGAs in a contemporary cohort are lacking.
In this study, we assessed SGA use in routine child and adolescent psychiatry treatment to prospectively quantify prevalence and incidence rates of drug-induced parkinsonism, dyskinesia, and akathisia during the first 3 months of naturalistic use of SGAs; and to identify risk profiles for neuromotor side effects in this population.
METHOD
Data were collected from December 2001 to September 2007 during the nonrandomized Second-generation Antipsychotic Treatment Indications, Effectiveness and Tolerability in Youth (SATIETY) study, an observational inception cohort study of antipsychotic medication use in routine clinical care. Inclusion/exclusion criteria and data collection strategies have been published previously,1 but the current study was not restricted to antipsychotic-naive patients. In short, children and adolescents aged 4 to 19 years who were initiating antipsychotic treatment based on clinician/family choice were included provided that baseline assessments occurred within 7 days of the current antipsychotic treatment. The following additional exclusion criteria were used: baseline use of anticholinergic medication, Tourette syndrome/tic disorder, catatonia, and antipsychotic switch explicitly because of EPS. Data from patients with sequential antipsychotic treatment trials were restricted to the first one. Participant data were recorded at baseline and at 4-, 8-, and 12-week follow-up.
Participants were recruited through the Zucker–Hillside Hospital, Queens, NY, and informed consent was obtained from all participants or guardians under protocols approved by the North Shore–Long Island Jewish Health System Institutional Review Board. All procedures were conducted in accordance with the ethical standards on human experimentation (institutional and national) and with the Declaration of Helsinki of 1975/2000.
Assessments
In addition to data collection as detailed earlier,1 investigators used the following assessments for neuromotor adverse effects: Simpson–Angus Scale (SAS) for parkinsonism (threshold at mean SAS ≥0.33 for “parkinsonism”22); the Abnormal Involuntary Movement Scale (AIMS; threshold for “dyskinesia” per Schooler and Kane criteria23); and the Barnes Akathisia Rating scale (BARS; threshold at BARS total ≥1 for “akathisia”24). For comparison with the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) trial, we also calculated rates using ≥1 for mean SAS scores, as in the CATIE trial primary analysis.25
Participant reports of adverse events were recorded by the investigators with a modified version of the Treatment Emergent Symptoms Scale (TESS).26
Data Analysis
Primary outcomes were the occurrence of drug-induced parkinsonism and treatment-emergent dyskinesia. Secondary outcomes were all other EPS-related outcomes.
To fulfill the definition of “drug-induced parkinsonism,” patients had to fulfill 1 or more of the following criteria: mean SAS >0.33 in patients with baseline mean SAS ≤0.33; start of anticholinergic medication; or marked increase of total SAS ratings of ≥2 in patients with baseline positive rating for EPS.
Dyskinesia was considered as treatment-emergent dyskinesia in patients without dyskinesia in a prior rating, or as an increase of ≥2 on AIMS total rating in patients with dyskinesia at baseline. Akathisia was defined by a rating of >1 on the BARS. A TESS sum score of neuromotor complaints was created, summing ratings for muscle pain/stiffness, joint pain/stiffness, dystonia, dyskinesia, rigidity, tremor, cramps, and myoclonus (range, 8–32). Primary psychiatric diagnoses were grouped into hierarchical categories to reduce the number of factors for statistical analyses.1
Group comparisons were performed using χ2 tests, nominal logistic fits and analysis of variance (ANOVA), t tests, Fisher’s exact test, or Wilcoxon rank-sum tests as per data type and distribution. Two-sided tests with α = 0.05 were used without correction for multiple comparisons because of the descriptive nature. To construct models of demographic and clinical variables associated with neuromotor variables, we conducted stepwise multivariate regression analyses. We used univariate analyses to screen for associations of neuromotor variables with age, sex, race/ethnicity, main psychiatric diagnosis, comorbidities, Children’s Global Assessment Scale (CGAS) scores, various aspects of medication information, and subsequent neuromotor ratings. Variables with marginal effects in these analyses (p ≤ .10) were entered into the stepwise forward regression analysis process.
Mixed-models analysis was used to explore effects of time, medication class, and their interactions, followed by post hoc t tests. Statistical calculations used JMP 5.0.1 (SAS Institute Inc., Cary, NC).
RESULTS
Data from 1,105 clinical SGA treatment initiations were collected. Of these, 4-week visits were performed in 660 trials (59.7%) from 498 patients. A total of 162 repeat trials were excluded. SAS baseline ratings were available in 373 patients (74.9%). Of the patients, 32 (8.6%) were excluded because of Tourette/tic disorder (n = 12), catatonia (n = 1), and explicit switch due to EPS or baseline anticholinergic treatment (n = 17). One patient on clozapine was excluded, resulting in a total study cohort of 342 individuals.
All 342 patients participated in the baseline and 4-week visits, and 258 and 242 patients participated in the 8- and 12-week visits, respectively (see Table S1, available online, for breakdown per SGA group).
Based on clinical decisions by their treating psychiatrists unrelated to the study team, 342 patients (mean age = 13.6 ± 3.5 years; 58.2% male) were started on SGAs (aripiprazole: n = 66, olanzapine: n = 58, quetiapine: n = 66, risperidone: n = 137, ziprasidone: n = 15; Table 1). Because of the naturalistic nature of the study, demographic, clinical, and treatment characteristics of SGA groups differed. In particular, high proportions of antipsychotic-naive patients were found in the olanzapine and risperidone groups, which also included more acutely and severely ill inpatients. Conversely, the ziprasidone and aripiprazole groups contained more chronically ill, antipsychotic-switching patients. In addition, high rates of non-antipsychotic comedications characterized the quetiapine and ziprasidone groups.
TABLE 1.
Total n = 342 | Aripiprazole n = 66 (19.30%) | Olanzapine n = 58 (16.96%) | Quetiapine n = 66 (19.30%) | Risperidone n = 137 (40. 06%) | Ziprasidone n = 15 (4.39%) | p Value | |
---|---|---|---|---|---|---|---|
Age, y, mean ± SD | 13.6 ± 3.5 | 12.97 ± 3.41 | 14.21 ± 3.26 | 13.30 ± 3.15 | 13.71 ± 3.74 | 14.45 ± 3.11 | .24 |
Sex, male, n (%) | 199 (58.19) | 44 (66.67) | 39 (67.24) | 28 (42.42) | 82 (59.85) | 6 (40.00) | .01* |
Race, nonwhite, n (%) | 186 (54.39) | 33 (50.00) | 34 (58.62) | 39 (59.09) | 74 (54.01) | 6 (40.00) | .60 |
Primary Diagnosis, n (%) | .14 | ||||||
Mood spectrum disorders | 158 (46.20) | 27 (40.91) | 29 (50.00) | 40 (60.61) | 58 (42.34) | 4 (26.67) | |
MDD | 40 (11.67) | 9 (13.64) | 8 (13.79) | 7 (10.60) | 16 (11.67) | 0 (0.00) | |
BD | 75 (21.92) | 11 (16.67) | 15 (25.86) | 21 (31.82) | 24 (17.59) | 4 (26.67) | |
Mood disorder NOS | 43 (12.57) | 7 (10.60) | 6 (10.34) | 12 (18.18) | 18 (13.14) | 0 (0.00) | |
Schizophrenia spectrum disorders | 91 (26.60) | 17 (25.76) | 16 (27.59) | 10 (15.15) | 42 (30.66) | 6 (40.00) | |
Schizophrenia, schizophreniform, schizoaffective disorder | 36 (10.53) | 7 (10.61) | 11 (18.97) | 2 (3.03) | 12 (8.76) | 4 (26.67) | |
Psychotic disorder NOS | 55 (16.08) | 10 (15.15) | 5 (8.62) | 8 (12.12) | 30 (21.90) | 2 (13.13) | |
Aggression spectrum disorder | 93 (27.20) | 22 (33.33) | 13 (22.41) | 16 (24.24) | 37 (27.01) | 5 (33.33) | |
ASD | 34 (9.94) | 12 (18.18) | 3 (5.17) | 5 (7.85) | 11 (8.03) | 3 (20.00) | |
ODD, CD | 59 (17.25) | 10 (15.15) | 10 (17.24) | 11 (16.67) | 26 (18.98) | 2 (13.33) | |
Comorbid ADHD, n (%) | 138 (43.28) | 27 (40.91) | 19 (43.94) | 29 (43.94) | 57 (41.61) | 6 (40.00) | .77 |
Comorbid OCD, n (%) | 19 (5.56) | 3 (4.55) | 3 (5.17) | 4 (6.06) | 7 (5.11) | 2 (13.33) | .75 |
Comorbid SUD, n (%) | 52 (15.21) | 6 (9.09) | 13 (22.41) | 8 (12.12) | 25 (18.25) | 0 (0.0) | .08 |
Antipsychotic Exposure Before Trial | <.0001* | ||||||
Antipsychotic history (≥1 mo off SGA) | 57 (16.67) | 13 (19.70) | 12 (20.69) | 12 (18.18) | 17 (12.41) | 3 (20.00) | |
Antipsychotic switch | 60 (17.54) | 17 (25.76) | 5 (8.62) | 19 (28.79) | 8 (5.84) | 11 (73.33) | |
Antipsychotic naive (≤1 wk lifetime) | 225 (65.79) | 36 (55.55) | 41 (70.69) | 35 (53.03) | 112 (81.75) | 1 (6.67) | |
Prior SGA treatment, mo, median (25th, 75th percentiles) | 0 (0, 2.5) | 0 (0, 6.24) | 0 (0, 1.25) | 0 (0, 6.75) | 0 (0, 0) | 7.75 (2, 35.6) | <.0001* |
CGAS/baseline, mean ± SD | 36.7 ± 8.95 | 38.68 ± 7.88 | 32.07 ± 9.25 | 38.75 ± 8.31 | 36.52 ± 8.93 | 39.67 ± 9.05 | <.0001* |
Inpatients, n (%) | 230 (67.25) | 34 (51.52) | 50 (86.21) | 45 (68.18) | 93 (67.88) | 8 (53.33) | .001* |
Non-SGA psychotropic comedication, n (%) | 237 (62.30) | 37 (56.06) | 38 (65.52) | 54 (81.82) | 97 (70.80) | 11 (73.33) | .03* |
Total comedications, median (25th, 75th percentiles) | 1 (0, 1.25) | 1 (0, 1.25) | 1 (0, 1) | 1 (1, 2) | 1 (0, 1) | 1 (0, 2) | .06 |
Comedications, n (%) | |||||||
Psychostimulants | 75 (21.99) | 14 (21.21) | 9 (15.52) | 19 (28.79) | 29 (21.32) | 4 (26.67) | .50 |
Antidepressants | 92 (26.90) | 17 (25.76) | 10 (17.24) | 22 (33.33) | 42 (30.66) | 1 (1.09) | .08 |
Mood stabilizer (antiepileptic or lithium) | 112 (32.72) | 14 (21.21) | 25 (43.10) | 31 (46.97) | 35 (25.55) | 7 (46.67) | .002* |
Lithium | 50 (14.62) | 5 (7.58) | 11 (18.97) | 17 (25.76) | 12 (8.76) | 5 (33.33) | .001* |
Valproate | 62 (18.13) | 10 (15.15) | 15 (25.86) | 13 (19.70) | 22 (16.06) | 2 (13.33) | .50 |
SGA start before baseline assessment, n (%) | 189 (55.26) | 25 (37.88) | 37 (63.79) | 39 (59.09) | 81 (59.12) | 7 (46.67) | .02* |
No. of days on new SGA before baseline assessment, mean ± SD | 1.95 ± 2.88 | 1.25 ± 2.38 | 2.64 ± 3.67 | 1.86 ± 2.35 | 1.96 ± 2.76 | 2.73 ± 4.06 | .07 |
Maximum CPZ equivalent dose, mean ± SD | 194.5 ± 187.7 | 179.05 ± 112.14 | 215.52 ± 133.43 | 404.03 ± 277.97 | 95.84 ± 75.77 | 160.32 ± 78.02 | <.0001* |
No. of wk completed, mean ± SD | 10.28 ± 3.46 | 10.40 ± 3.57 | 10.80 ± 2.63 | 10.26 ± 3.80 | 10.29 ± 3.41 | 7.69 ± 3.99 | .04* |
% CGAS Change, mean ± SE | 45.82 ± 2.84 | 28.60 ± 6.07 | 78.32 ± 6.34 | 29.80 ± 6.18 | 51.53 ± 4.14 | 6.79 ± 6.80 | .0001* |
Continuation of SGA at 3 mo, n (%) | 232 (68.44) | 48 (73.8) | 46 (79.31) | 44 (67.69) | 89 (64.96) | 5 (35.71) | .01* |
Note: ADHD = attention-deficit/hyperactivity disorder; ASD = autism spectrum disorder; BD = bipolar disorder; CD = conduct disorder; CGAS = Children’s Global Assessment Scale; CPZ = chlorpromazine; MDD = major depressive disorder; NOS = not otherwise specified; OCD = obsessive-compulsive disorder; ODD = oppositional defiant disorder; SGA = second-generation antipsychotic; SUD = substance use disorder.
Significant between-group differences (p < .05).
Baseline Neuromotor Characteristics
Detailed neuromotor characteristics for each single time point per SGA are listed in Table S1, available online. Baseline parkinsonism was present in 4.97% of patients and differed between SGA subgroups (from 0% in olanzapine to 20% in ziprasidone, p = .01; Table 2). SAS total baseline score was associated with higher age, antipsychotic switch, allocation to ziprasidone, and higher baseline CGAS score (full model R2 = 0.11, p < .0001; see Table S2, available online).
TABLE 2.
Baseline | Total N = 342 | Aripiprazole n = 66 | Olanzapine n = 58 | Quetiapine n = 66 | Risperidone n = 137 | Ziprasidone n = 15 | p Value |
---|---|---|---|---|---|---|---|
Parkinsonism (mean SAS >0.33), n (%) | 17 (4.97) | 6 (9.09) | 0 (0.00) | 3 (4.55) | 5 (3.65) | 3 (20.00) | .01* |
SAS total, mean ± SD | 0.70 ± 1.66 | 0.85 ± 1.63 | 0.31 ± 0.68 | 0.69 ± 1.71 | 0.60 ± 1.35 | 2.40 ± 4.29 | .0004* |
Dyskinesia, n (%) | 24 (7.10) | 7 (10.77) | 4 (6.90) | 6 (9.38) | 6 (9.38) | 1 (6.67) | .50 |
AIMSa total, mean ± SD | 1.23 ± 2.12 | 1.45 ± 1.92 | 1.13 ± 2.81 | 1.23 ± 2.21 | 1.19 ± 1.84 | 1.13 ± 1.92 | .92 |
Akathisia, n (%) | 2 (0.58) | 0 | 0 | 0 | 2 (1.46) | 0 | .36 |
Wk 12b | n = 259 | n = 49 | n = 51 | n = 50 | n = 101 | n = 8 | |
Antipsychotic dose, mean ± SD | N/A | 11.76 ± 7.39 | 9.68 ± 6.85 | 231.9 ± 184.3 | 1.38 ± 1.17 | 63.3 ± 52.8 | N/A |
| |||||||
Drug-induced parkinsonism, n (%) | 27 (10.42) | 13 (26.53) | 4 (7.85 ) | 1 (2.00) | 7 (6.93) | 2 (25.00) | .0003* |
SAS total, mean ± SD | 1.29 ± 2.25 | 2.22 ± 2.74 | 1.02 ± 1.54 | 0.62 ± 1.14 | 1.11 ± 2.42 | 2.13 ± 3.87 | .003* |
Treatment-emergent dyskinesia, n (%)b | 12 (4.63) | 0 (0.00) | 3 (5.88) | 2 (4.00) | 1 (0.98) | 2 (25.00) | .05* |
AIMS total, mean ± SD | 1.62 ± 2.21 | 1.49 ± 1.41 | 1.67 ± 2.85 | 2.04 ± 2.79 | 1.43 ± 1.67 | 2.13 ± 3.44 | .65 |
Akathisia, n (%) | 7 (2.78) | 3 (6.25) | 2 (4.00) | 0 (0.00) | 2 (2.06) | 0 (0.00) | .38 |
3-mo Frequencies (LOCF) | N = 342 | n = 66 | n = 58 | n = 66 | n = 137 | n = 15 | |
| |||||||
Drug-induced parkinsonism, n (%) | 52 (15.20) | 18 (27.27) | 8 (13.79) | 1 (1.52) | 22 (16.06) | 3 (20.00) | .002* |
Anticholinergic medication, n (%) | 17 (5.03) | 3 (4.76) | 0 (0.0) | 0 (0.0) | 14 (10.22) | 0 (0.0) | .0004* |
Highest single SAS item score, mean ± SD | 0.88 ± 1.00 | 1.14 ± 0.99 | 0.91 ± 0.85 | 0.55 ± 0.66 | 0.86 ± 1.08 | 1.13 ± 1.50 | .01* |
Significant, treatment-emergent dyskinesia, n (%) | 28 (8.28) | 3 (4.55) | 9 (15.52) | 6 (9.5) | 6 (4.41) | 4 (26.67) | .005* |
Highest AIMS item during 3 mo, mean ± SD | 0.91 ± 0.91 | 1.19 ± 0.84 | 0.80 ± 1.04 | 0.96 ± 0.98 | 0.84 ± 0.83 | 1.0 ± 1.0 | .11 |
Akathisia, n (%) | 16 (4.83) | 5 (8.06) | 3 (5.36) | 1 (1.59) | 7 (5.15) | 0 (0.0) | .45 |
Neuroleptic malignant syndrome, n (%) | 0 (0.0) | 0 (0.0) | 0 (0.0) | 0 (0.0) | 0 (0.0) | 0 (0.0) | - |
Discontinuation due to extrapyramidal side effect, n (%) | 11 (3.27) | 4 (6.15) | 1 (1.72) | 0 (0.0) | 6 (4.48) | 0 (0) | .008* |
Note: AIMS = Abnormal Involuntary Movement Scale; LOCF = last observation carried forward; N/A = not applicable; SAS = Simpson–Angus Scale.
Significant between-group differences shown as *p < .05.
Group comparisons remain insignificant after exclusion of previously antipsychotic-exposed participants with baseline dyskinesia.
See Table S1, available online, for wk 4 and wk 8 data.
Baseline dyskinesia was present in 7.1% (Table 2) without differences by SGA subgroup, but more frequent in switchers (13.33%) and in patients with prior antipsychotic exposure (12.28%; antipsychotic-naive: 3.62%; p = .006, see Table S3, available online). Higher baseline AIMS scores were associated with younger age, comedication with psychostimulants, and higher number of prior antipsychotic trials (full model R2 = 0.16, p < .0001; see Table S2, available online). Notably, psychostimulant use was not a proxy for attention-deficit/hyperactivity comorbidity, which did not enter the multivariate model.
Tremor was the most frequently reported neuromotor complaint at baseline (11.38%, see Table S3, available online); any moderate neuromotor complaint (>2 on TESS) was recorded for 35 participants (10.9%; without significant SGA subgroup difference, data not shown). Presence of a moderate neuromotor complaint at baseline was associated with the number of non-antipsychotic comedications, medication history (increased in antipsychotic-naive participants) and diagnosis of mood spectrum or schizophrenia spectrum disorders (full model R2 = 0.13, p < .0001; see Table S2, available online).
Baseline rates for parkinsonism did not differ (p = .96) by primary psychiatric diagnosis in a subgroup analysis of antipsychotic-naive, medication-free participants (n = 102; see Table S3, available online). Dyskinesia was even significantly less frequent in schizophrenia spectrum disorders in this subgroup analysis (0% versus 11.36% and 12% in schizophrenia spectrum, aggression, and mood disorders, respectively; p = .04; see Table S3, available online).
Follow-Up Neuromotor Characteristics
Drug-Induced Parkinsonism.
The overall 3-month incidence of drug-induced parkinsonism was low (15.2%; Table 2) and its severity was mild (SAS total score = 6.30 ± 4.13 in affected individuals [n = 27; at week 4]). Similarly, anticholinergic use (5.03%; table 2) and discontinuation due to EPS (3.27%, n = 11) were low.
The raw 3-month incidence of drug-induced parkinsonism differed between SGA subgroups (p = .002; Table 2), with highest rates in aripiprazole (27.27%) and lowest in quetiapine (1.52%). Similarly, prevalence rates at each time point and anticholinergic use differed (p < .05 for all comparisons; see Table S1, available online). Controlling for the factors age, SGA peak dose, and baseline CGAS score, significant SGA subgroup differences were identified, with the lowest rates of drug-induced parkinsonism in quetiapine and in olanzapine (full model R2 = 0.18; p < .0001; see Table S2, available online). At week 12, significant within-group increases in SAS total ratings relative to baseline (p < .001) were present with aripiprazole, olanzapine, and risperidone, but not with quetiapine (p = .30). Because of the high dropout in the ziprasidone subgroup, an LOCF analysis was calculated for this group, which did not show a significant change in this group (p = .79).
Anticholinergic comedication was most frequent with risperidone (10.22%; Table 2), in inpatients (7.02% versus 0.09% for outpatients; p = .006), in patients with schizophrenia spectrum disorder (12.5% versus 3.16% in mood disorders and 1.08% in aggression; p = .001) and with lower baseline CGAS scores (mean ± SE, 28.24 ± 1.74 versus 37.22 ± 0.49; p < .0001).
To facilitate comparison with the CATIE trial, we calculated rates of parkinsonism according to their primary threshold.25 Using this definition, SGA subgroup comparisons were significant only for baseline (aripiprazole = 0%, olanzapine = 0%, quetiapine = 1.52%, risperidone = 0%, ziprasidone = 13.33%; p = .001; see Table S1, available online) and for week 8 (aripiprazole = 7.55%, olanzapine = 0%, quetiapine = 0%, risperidone = 2.02%, ziprasidone = 14.29%; p = .03; see Table S1, available online).
Significant dose effects of the antipsychotic medication were present in all agents but aripiprazole (correlation of SAS total score: risperidone [week 4: R = 0.36; p < .0001; week 12: R = 0.28; p = .004], olanzapine [week 12: R = 0.34; p = .01], quetiapine [week 4: R = 0.30; p = .01], ziprasidone [week 12: R = 0.87; p = .004]).
Treatment-Emergent Dyskinesia.
The raw overall 3-month incidence of treatment-emergent dyskinesia was 8.28% and differed between SGA subgroups, ranging from 4.41% in the risperidone group to 26.67% in the ziprasidone group (p = .005; Table 2). Controlling for the factors baseline AIMS score, age, and race/ethnicity, SGA subgroups significantly differed, with higher rates of treatment-emergent dyskinesia in olanzapine and ziprasidone (full model R2 = 0.31; p < .0001; see Table S2, available online). Dose effects on dyskinesia ratings were significant within SGA group only for risperidone at week 4 (highest single AIMS item [R = −0.31; p = .0004], AIMS total score [R = −0.27; p = .0015]).
AIMS sum scores did not differ between switchers and the remaining cohort at week 4 (p = .97), week 8 (p = .09), and week 12 (p = .49). Similarly, these groups showed comparable rates of treatment-emergent dyskinesia (at week 4, 8, or 12; p > .5 respectively), showing no confounding effect of withdrawal dyskinesia.
Akathisia.
Observer-rated akathisia was rare throughout the study, with a cumulative rate of 4.83% for the entire cohort (Table 2), without dose effect or SGA group differences and with constant monthly rates of 2.3% to 2.7% (see Table S1, available online).
Participant-Reported Neuromotor Side Effects.
Participant-reported akathisia rates were slightly higher than those assessed by observers (see Table S3, available online), with increased early rates for aripiprazole (week 4: 11.67%) and ziprasidone (week 4: 7.14%; versus rates of <2% in the other SGAs; p = .006; see Table S1, available online).
Frequencies for all other single neuromotor TESS items are listed in Table S3, available online (for the entire cohort, as no SGA group differences were found; all p > .05). Tremor was the most frequently reported complaint across SGAs, with 18% of participants reporting this at week 4. Higher tremor ratings at week 4 were associated with mood spectrum disorders, higher age, number of psychotropic medications, and, controlling for these factors, were lower with aripiprazole (full model R2 = 0.14; p < .0001, see Table S2, available online).
DISCUSSION
This is the largest cohort of youth formally assessed for EPS, and it is the first prospective direct comparison of neuromotor side effects of the 5 most used SGAs in a naturalistic setting. As SGAs are widely used off label, populations that are not included in RCTs are well represented in our study. A comparison of comedication use is highly illustrative of how little RCT cohorts compare to our routine care cohort: 69% of participants received additional psychotropic medication, most frequently stimulants, antidepressants, or mood stabilizers. By contrast, these rates range below 10% in the majority of RCTs.8,9,16,18,27
Our study showed an overall rate of drug-induced parkinsonism of 15.2%, ranging from 1% for quetiapine to 27.7% with aripiprazole for the raw, observed rates. However, the clinical severity of parkinsonism was generally low, indicated by overall low rates of the following: subjective perception of any sort of muscle rigidity (mild in 5%); anticholinergic comedication use (3%); or treatment termination due to extrapyramidal symptoms (3%). Notably, in addition to SGA type and dose, higher age and lower baseline CGAS score were significant predictors of drug-induced parkinsonism.
The relative ranking of drug-induced parkinsonism rates across the examined 5 SGAs in our study is generally in accordance with earlier studies of SGAs in youth that examined only 1 SGA or, at most, 1 other SGA in the same study. However, the absolute rates and risk estimates differ to some degree. Controlling for the confounding factors (inherently associated with the naturalistic design), significant, independent SGA effects existed only for quetiapine (low EPS liability), olanzapine (low EPS liability), and ziprasidone (high EPS liability). Data from registration trials suggest that olanzapine (with rates <5%28,29) followed by quetiapine (with rates of <5%30 and 13%31) would be the least EPS-prone antipsychotics, roughly similar to our data. However, our data suggest an even lower EPS risk with quetiapine. Higher EPS rates than in our data were reported for aripiprazole (14%–39%8,10,32,33). Roughly comparable rates were reported for risperidone (9-16%15,16,34) and for ziprasidone (11%–24%18,35).
In contrast to the relative ranking of EPS liability of individual SGAs, our EPS rates also differ from those obtained in SGA registration trials: participant-based EPS reports were higher throughout all registration trials, whereas rating scale–based frequencies were seemingly lower. However, anticholinergic use, likely in response to higher participant complaints of parkinsonism, was far higher in all registration trials compared to those in our study. This is particularly true for fixed-dose studies with rapid titration schemes:16,36 Thus, it is possible that liberal or preventive anticholinergic comedication may have led to low/absent parkinsonism rates in some cases and, in particular, may have masked rating scale–based changes in EPS measures in these studies. Accordingly, SAS ratings were unchanged from baseline to endpoint in most registration trials.9,18,27–29,33 Similarly, reported parkinsonism rates in the risperidone registration trials were partly lower (4%–16%) than the raw cumulative rates in our study (16%), but anticholinergic use was very common and up to 10-fold higher, with 56%/38% in the high-/low-dose subgroups.34 Likewise, anticholinergic use was also higher in the Treatment of Early-Onset Schizophrenia Spectrum disorders (TEOSS) trial,20 with rates of 12% for olanzapine (0% in our study) and 33% for risperidone (10% in our study). Despite this difference, SAS-based rates of parkinsonism were also higher in the TEOSS trial, ranging above 20% for both olanzapine and risperidone. With mean doses of 9 mg of olanzapine and 1.5 mg of risperidone at week 4, the titration schedule in the majority of our study participants was somewhat slower than in the TEOSS study, which aimed at similar dose ranges by day. This difference in titration speed could explain the lower parkinsonism rate in our naturalistic study.
Participant-based reports of side effects were higher throughout all registration trials, except for tremor reporting, which was higher for all treatment subgroups in our study. Interestingly, tremor was also linked to psychotropic polypharmacy in our study, which was far less prevalent in the registration trials.
Apart from registration trials, there is also substantial variability of reported rates of EPS in other clinical trials. The low EPS rates with quetiapine observed in our study have not consistently been reported previously (0%13–46%14) but match smaller case series in youth.13,37 A similar variability in reported drug-induced parkinsonism rates exists for risperidone, with an absence of any drug-induced parkinsonism despite SAS-based ratings in a sizable cohort17 and contrasting high rates (~50%) in smaller cohorts using both rating scale–based assessments38 and participant reports.37 Finally, our SAS-based data, but not the anticholinergic use data, match the reported rates of drug-induced parkinsonism determined by use of anticholinergic comedication.19
Compared to studies in adults, our data do not support the idea that the risk of SGA-induced parkinsonism is generally elevated in youth,39,40 an idea generalized from observations of first-generation antipsychotics.4 This observation is in line with a previous indirect comparison of EPS and akathisia rates reported in RCTs of pediatric and adult patients with bipolar disorder.41 Similarly, compared to data from placebo-controlled RCTs in adults, lower rates of parkinsonism were found in our study (16%) for risperidone (adults: any significant EPS = 23%, anticholinergic use = 27%42) and particularly for quetiapine (1.5% in our study; adults: significant EPS = 11%, anticholinergic use = 7%43). Comparable rates were found for olanzapine (anticholinergic use = 13%44). Similarly, rates of drug-induced parkinsonism for aripiprazole in our study (27%) roughly match those reported in adults (adults: significant EPS = 31%, anticholinergic use = 17%45). Nevertheless, these data from usual care of youth are not suitable to refute a higher general sensitivity in antipsychotic-treated youth for EPS, as clinicians seem to have used a more cautious titration schedule and lower maximum dose in the pediatric population. Anticholinergics were barely used in our naturalistic cohort (3%, very likely reflective of the mild severity of the EPS), possibly to avoid adverse cognitive effects.
The comparison of EPS frequencies among studies is largely hampered by an inconsistent use of measures for parkinsonism. Although the use of anticholinergics as a proxy for EPS seems rational,46 these rates are typically far lower (disregarding the rate of underdiagnosed EPS) than those derived from either participant report or scale-based ratings. Moreover, even when scale-based ratings are used, there is a wide variability of SAS thresholds used, despite a clear original definition.22 Several current pediatric RCTs reported relative risks for SAS-measured EPS, without specifying thresholds.11,13,17,19,20,28,47 Most surprisingly, however, EPS rates reported in the CATIE study,25 ranging from 4%–8% only, were obtained with a 6-item SAS,48 whereas the original SAS contains 10 items, and parkinsonism was defined at a threshold of a mean SAS of ≥1 (original threshold = >0.3).22 Defining parkinsonism at this threshold would have resulted in parkinsonism rates <3% in our data and no discrimination of SGA classes with regard to neuromotor side effects. Although it may have been more appropriate to use a validated threshold in a landmark trial,22 there have also been rightful concerns regarding the originally defined SAS threshold. Testing the SAS against the Unified Parkinson’s Disease Rating Scale49,50 showed high sensitivity but low specificity.51,52 An actimetric study,52 in line with an imaging study,53 suggested that raising SAS thresholds to 0.65 doubled the specificity without reducing sensitivity. While inappropriately high thresholds may result in missed diagnoses of drug-induced parkinsonism,54 inappropriately low thresholds may result in a problematic overlap of negative symptoms with EPS. Conversely, an all-too-quick attribution of any movement symptom during antipsychotic treatment as a drug-induced movement disorder carries the risk of missing underlying organic conditions such as Wilson’s, systemic lupus, neuroakanthocytosis, spinocerebellar ataxia or adrenoleukodystrophy, all of which may begin in youth.
Our study found relatively high rates of baseline dyskinesia, mostly but not exclusively in participants with prior antipsychotic use. In contrast to the suggestion of a genuinely schizophrenia-inherent motor syndrome,55 dyskinesia rates were relatively lower in antipsychotic-naive participants with schizophrenia spectrum disorders compared to mood and aggression spectrum disorder, and parkinsonism rates did not differ across diagnostic groups. Importantly, stimulant use was associated with baseline dyskinesia, a phenomenon that is recognized as a rare side effect,56 but may be more prevalent when the stimulant is used as part of polypharmacy. We refrain from discussing our 3-month data in the context of literature on tardive dyskinesia, as its formal definition requires persistence past this time frame.57
Being naturalistic, this study is limited in multiple ways. Baseline differences of SGA subgroups reflect clinician treatment choices, with olanzapine and risperidone as first-line treatments in antipsychotic-naive and more severely ill participants during the initial SATIETY study period. Thus, raw EPS rates in our study should not be interpreted without considering the context of confounding factors that have been controlled for in the multivariate analyses. The observation period of 12 weeks was sufficient to address the occurrence of parkinsonism, but dyskinesia needs to be analyzed in a cohort followed for a longer time. The sample size of 342 starters and 250 full completers is insufficient to discover rare side effects; pharmacovigilance systems are more suitable for this purpose, but often lack detailed information and rating scale–based assessments. As this is not an RCT, no causations can be inferred. The observations are merely associational, yet clinically relevant. Not all baseline assessments were performed before SGA initiation; the mean duration before assessments was only 2 days. Raters were not blinded. The neuromotor rating scales have not been validated in children. We could include only a small number of participants with ziprasidone. Although this subgroup differed in multiple ways from the majority, we kept this group to add to the scarce database for ziprasidone in youth.
In summary, we observed the lowest rates of parkinsonism with quetiapine and olanzapine, and the highest anticholinergic use with risperidone, yet low treatment discontinuations for EPS. A surprisingly large number of youth had dyskinesia at baseline, which was associated with psychostimulant use. Incident dyskinesia was highest with olanzapine. Our data support that SGAs used in children and adolescents have, in general, relatively few neuromotor side effects, yet these differ among individual agents and are likely affected by initial dosing and titration strategies. These individual patterns need to be considered in the context of medication- and patient-related tolerability and efficacy profiles of SGAs,58 as well as patient and family preferences and choices, when making treatment decisions.
Supplementary Material
Clinical Guidance.
Overall rates of drug-induced parkinsonism during clinically adapted second-generation antipsychotic (SGA) treatment were low, indicating that the short-term neuromotor adverse effects of SGAs during routine care are not a major clinical problem.
During naturalistic treatment, particularly low rates of drug-induced parkinsonism were found with quetiapine, indicating that youth prone to extrapyramidal adverse effects might benefit from this drug choice.
Beyond SGA choice, drug-induced parkinsonism was associated with higher dose, higher age, and lower baseline functioning:
Cautious dosing seems advisable not only in young children but also in adolescents.
Cautious dosing seems advisable also in low-functioning youth.
Psychotropic polypharmacy was associated with increased rates of tremor and dyskinesia, highlighting the need for carefully weighing the indications of each and every psychotropic medication in the therapeutic management plan.
Acknowledgments
This work is supported in parts by The Zucker Hillside Hospital National Institute of Mental Health (NIMH) Advanced Center for Intervention and Services Research for the Study of Schizophrenia (P30MH090590; Dr. Kane), and by The Feinstein Institute for Medical Research North Shore-Long Island Jewish Health System General Clinical Research Center, Grant #M01RR018535 from the National Center for Research Resources (NCRR), a component of the National Institutes of Health (NIH).
The article’s content is solely the responsibility of the authors and does not necessarily represent the official view of NCRR, NIH, or NIMH. None of these non-commercial funding organizations had any role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript.
Dr. Carbon served as the statistical expert for this research.
The authors wish to thank the patients and their families for participation in this project.
Disclosure:
Dr. Carbon has the same financial disclosure as Dr. Correll due to their family relationship. Dr. Kane has served as a consultant to Alkermes, Amgen, AstraZeneca, Janssen, Pfizer, Eli Lilly and Co., Bristol-Myers Squibb, Dainippon Sumitomo/Sepracor/Sunovion, Johnson and Johnson, Otsuka, Pierre Fabre, Vanda, Proteus, Takeda, Targacept, IntraCellular Therapies, Merck, Lundbeck, Novartis, Roche, and Rules Based Medicine, and has received honoraria for lectures from Otsuka, Eli Lilly and Co., Esai, Boehringer-Ingelheim, Bristol-Myers Squibb, Merck, and Janssen. He is a shareholder of MedAvante. Dr. Correll has received grant or research support from the National Institute of Mental Health, the American Academy of Child and Adolescent Psychiatry, Bristol-Myers Squibb, Janssen/Johnson and Johnson, Novo Nordisk A/S, Otsuka, Takeda, and the Thrasher Foundation. He has served as a consultant to AbbVie, Acadia, Actavis, Alkermes, Eli Lilly and Co., Genentech, the Gerson Lehrman Group, IntraCellular Therapies, Janssen/Johnson and Johnson, Lundbeck, Medavante, Medscape, National Institute of Mental Health, Otsuka, Pfizer, ProPhase, Reviva, Roche, Sunovion, Supernus, and Takeda. He has presented expert testimony for Janssen. He has served as a member of the Data Safety Monitoring Boards for Eli Lilly and Co., Janssen, Lundbeck, and Pfizer. Drs. Kapoor, Sheridan, Al-Jadiri, Azzo, Sarkaria, and Saito report no biomedical financial interests or potential conflicts of interest.
Contributor Information
Maren Carbon, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY; Charité Berlin, Berlin.
Sandeep Kapoor, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY; Feinstein Institute for Medical Research, Manhasset, NY.
Eva Sheridan, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY.
Aseel Al-Jadiri, New York Methodist Hospital, Brooklyn, NY.
Sally Azzo, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY.
Tania Sarkaria, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY.
John M. Kane, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY; Feinstein Institute for Medical Research, Manhasset, NY; Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, NY.
Ema Saito, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY; Feinstein Institute for Medical Research, Manhasset, NY.
Christoph U. Correll, The Zucker Hillside Hospital, Psychiatry, Glen Oaks, NY; Feinstein Institute for Medical Research, Manhasset, NY; Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, NY.
REFERENCES
- 1.Correll CU, Manu P, Olshansky V, Napolitano B, Kane JM, Malhotra AK. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302:1765–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kane JM. Addressing side effects from antipsychotic treatment in schizophrenia. The J Clin Psychiatry. 2011;72:e07. [DOI] [PubMed] [Google Scholar]
- 3.Stentebjerg-Olesen M, Jeppesen P, Pagsberg AK, et al. Early nonresponse determined by the clinical global impressions scale predicts poorer outcomes in youth with schizophrenia spectrum disorders naturalistically treated with second-generation antipsychotics. J Child Adolesc Psychopharmacol. 2013;23:665–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lewis R Typical and atypical antipsychotics in adolescent schizophrenia: efficacy, tolerability, and differential sensitivity to extrapyramidal symptoms. Can J Psychiatry. 1998;43:596–604. [DOI] [PubMed] [Google Scholar]
- 5.Kumar A, Datta SS, Wright SD, Furtado VA, Russell PS. Atypical antipsychotics for psychosis in adolescents. Cochrane Database System Rev. 2013;10:CD009582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pringsheim T, Lam D, Ching H, Patten S. 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]
- 7.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;21:621–645. [DOI] [PubMed] [Google Scholar]
- 8.Owen R, Sikich L, Marcus RN, et al. Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics. 2009;124:1533–1540. [DOI] [PubMed] [Google Scholar]
- 9.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;165:1432–1441. [DOI] [PubMed] [Google Scholar]
- 10.Findling RL, Kauffman R, Sallee FR, et al. An open-label study of aripiprazole: pharmacokinetics, tolerability, and effectiveness in children and adolescents with conduct disorder. J Child Adolesc Psychopharmacol. 2009;19:431–439. [DOI] [PubMed] [Google Scholar]
- 11.Frazier JA, Biederman J, Tohen M, et al. A prospective open-label treatment trial of olanzapine monotherapy in children and adolescents with bipolar disorder. J Child Adolesc Psychopharmacol. 2001;11:239–250. [DOI] [PubMed] [Google Scholar]
- 12.Mozes T, Ebert T, Michal SE, Spivak B, Weizman A. An open-label randomized comparison of olanzapine versus risperidone in the treatment of childhood-onset schizophrenia. J Child Adolesc Psychopharmacol. 2006;16:393–403. [DOI] [PubMed] [Google Scholar]
- 13.McConville B, Carrero L, Sweitzer D, et al. Long-term safety, tolerability, and clinical efficacy of quetiapine in adolescents: an open-label extension trial. J Child Adolesc Psychopharmacol. 2003;13:75–82. [DOI] [PubMed] [Google Scholar]
- 14.Arango C, Robles O, Parellada M, et al. Olanzapine compared to quetiapine in adolescents with a first psychotic episode. Eur Child Adolesc Psychiatry. 2009;18:418–428. [DOI] [PubMed] [Google Scholar]
- 15.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 disorders. 2009;11:687–700. [DOI] [PubMed] [Google Scholar]
- 16.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;194:158–164. [DOI] [PubMed] [Google Scholar]
- 17.Aman MG, Arnold LE, McDougle CJ, et al. Acute and long-term safety and tolerability of risperidone in children with autism. J Child Adolesc Psychopharmacol. 2005;15:869–884. [DOI] [PubMed] [Google Scholar]
- 18.Findling RL, Cavus I, Pappadopulos E, et al. Ziprasidone in adolescents with schizophrenia: results from a placebo-controlled efficacy and longterm open-extension study. J Child Adolesc Psychopharmacol. 2013;23:531–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.DelBello MP, Versavel M, Ice K, Keller D, Miceli J. Tolerability of oral ziprasidone in children and adolescents with bipolar mania, schizophrenia, or schizoaffective disorder. J Child Adolesc Psychopharmacol. 2008;18:491–499. [DOI] [PubMed] [Google Scholar]
- 20.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;165:1420–1431. [DOI] [PubMed] [Google Scholar]
- 21.Hong IS, Bishop JR. Anticholinergic use in children and adolescents after initiation of antipsychotic therapy. Ann Pharmacother. 2010;44:1171–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Simpson G, Angus JW. A rating scale for extrapyramidal side effects. Acta Psychiatr Scand Suppl. 1970;212:11–19. [DOI] [PubMed] [Google Scholar]
- 23.Schooler NR, Kane JM. Research diagnoses for tardive dyskinesia. Arch Gen Psychiatry. 1982;39:486–487. [DOI] [PubMed] [Google Scholar]
- 24.Barnes T A rating scale for drug-induced akathisia. Br J Psychiatry. 1989;154:672–676. [DOI] [PubMed] [Google Scholar]
- 25.Lieberman JA, Stroup TS, McEvoy JP, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353:1209–1223. [DOI] [PubMed] [Google Scholar]
- 26.Guy W ECDEU Assessment Manual for Psychopharmacology (Revised). Rockville, MD: National Institutes of Health; 1976. [Google Scholar]
- 27.Findling RL, McKenna K, Earley WR, Stankowski J, Pathak S. Efficacy and safety of quetiapine in adolescents with schizophrenia investigated in a 6-week, double-blind, placebo-controlled trial. J Child Adolesc Psychopharmacol. 2012;22:327–342. [DOI] [PubMed] [Google Scholar]
- 28.Kryzhanovskaya LA, Robertson-Plouch CK, Xu W, Carlson JL, Merida KM, Dittmann RW. The safety of olanzapine in adolescents with schizophrenia or bipolar I disorder: a pooled analysis of 4 clinical trials. J Clin Psychiatry. 2009;70:247–258. [DOI] [PubMed] [Google Scholar]
- 29.Tohen M, Kryzhanovskaya L, Carlson G, et al. Olanzapine versus placebo in the treatment of adolescents with bipolar mania. Am J Psychiatry. 2007;164:1547–1556. [DOI] [PubMed] [Google Scholar]
- 30.Pathak S, Findling RL, Earley WR, Acevedo LD, Stankowski J, Delbello MP. 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:e100–e109. [DOI] [PubMed] [Google Scholar]
- 31.Findling RL, Pathak S, Earley WR, Liu S, DelBello M. Safety, tolerability, and efficacy of quetiapine in youth with schizophrenia or bipolar I disorder: a 26-week, open-label, continuation study. J Child Adolesc Psychopharmacology. 2013;23:490–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Findling RL, Bukstein OG, Melmed RD, et al. A randomized, double-blind, placebo-controlled, parallel-group study of methylphenidate transdermal system in pediatric patients with attention-deficit/hyperactivity disorder. J Clin Psychiatry. 2008;69:149–159. [DOI] [PubMed] [Google Scholar]
- 33.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;48:1110–1119. [DOI] [PubMed] [Google Scholar]
- 34.Haas M, Unis AS, Armenteros J, Copenhaver MD, Quiroz JA, Kushner SF. 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;19:611–621. [DOI] [PubMed] [Google Scholar]
- 35.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;23:545–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Findling RL, Correll CU, Nyilas M, et al. Aripiprazole for the treatment of pediatric bipolar I disorder: a 30-week, randomized, placebo-controlled study. Bipolar Disord. 2013;15:138–149. [DOI] [PubMed] [Google Scholar]
- 37.Castro-Fornieles J, Parellada M, Soutullo CA, et al. Antipsychotic treatment in child and adolescent first-episode psychosis: a longitudinal naturalistic approach. J Child Adolesc Psychopharmacol. 2008;18:327–336. [DOI] [PubMed] [Google Scholar]
- 38.Swadi HS, Craig BJ, Pirwani NZ, Black VC, Buchan JC, Bobier CM. 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;25:1–6. [DOI] [PubMed] [Google Scholar]
- 39.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 North Am. 2006;15:177–206. [DOI] [PubMed] [Google Scholar]
- 40.Correll CU. Antipsychotic use in children and adolescents: minimizing adverse effects to maximize outcomes. J Am Acad Child Adolesc Psychiatry. 2008;47:9–20. [DOI] [PubMed] [Google Scholar]
- 41.Correll CU, Sheridan EM, DelBello MP. Antipsychotic and mood stabilizer efficacy and tolerability in pediatric and adult patients with bipolar I mania: a comparative analysis of acute, randomized, placebo-controlled trials. Bipolar Disord. 2010;12:116–141. [DOI] [PubMed] [Google Scholar]
- 42.Rattehalli RD, Jayaram MB, Smith M. Risperidone versus placebo for schizophrenia. Cochrane Database Syst Rev. 2010;(1):CD006918. [PubMed] [Google Scholar]
- 43.Srisurapanont M, Maneeton B, Maneeton N. Quetiapine for schizophrenia. Cochrane Database Syst Rev 2004;(2):CD000967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Duggan L, Fenton M, Rathbone J, Dardennes R, El-Dosoky A, Indran S. Olanzapine for schizophrenia. Cochrane Database Syst Rev 2005;(2):CD001359. [DOI] [PubMed] [Google Scholar]
- 45.Potkin SG, Saha AR, Kujawa MJ, et al. Aripiprazole, an antipsychotic with a novel mechanism of action, and risperidone vs placebo in patients with schizophrenia and schizoaffective disorder. Arch Gen Psychiatry. 2003;60:681–690. [DOI] [PubMed] [Google Scholar]
- 46.Leucht S, Cipriani A, Spineli L, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments metaanalysis. Lancet. 2013;382:951–962. [DOI] [PubMed] [Google Scholar]
- 47.Zalsman G, Carmon E, Martin A, Bensason D, Weizman A, Tyano S. Effectiveness, safety, and tolerability of risperidone in adolescents with schizophrenia: an open-label study. J Child Adolesc Psychopharmacol. 2003;13:319–327. [DOI] [PubMed] [Google Scholar]
- 48.Miller DD, Caroff SN, Davis SM, et al. Extrapyramidal side-effects of antipsychotics in a randomised trial. Br J Psychiatry. 2008;193:279–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Goetz CG, Tilley BC, Shaftman SR, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Move Disord. 2008;23:2129–2170. [DOI] [PubMed] [Google Scholar]
- 50.Ishikawa T, Dhawan V, Kazumata K, et al. Comparative nigrostriatal dopaminergic imaging with iodine-123-beta CIT-FP/SPECT and fluorine-18-FDOPA/PET. J Nucl Med. 1996;37:1760–1765. [PubMed] [Google Scholar]
- 51.Blanchet PJ, Rompre PH. Clinimetric evaluation of the Simpson-Angus Scale in older adults with schizophrenia. J Clin Psychopharmacol. 2014;34:36–39. [DOI] [PubMed] [Google Scholar]
- 52.Janno S, Holi MM, Tuisku K, Wahlbeck K. Validity of Simpson-Angus Scale (SAS) in a naturalistic schizophrenia population. BMC Neurol. 2005;5:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tauscher J, Kufferle B, Asenbaum S, Tauscher-Wisniewski S, Kasper S. Striatal dopamine-2 receptor occupancy as measured with [123I]iodobenzamide and SPECT predicted the occurrence of EPS in patients treated with atypical antipsychotics and haloperidol. Psychopharmacology. 2002;162:42–49. [DOI] [PubMed] [Google Scholar]
- 54.Esper CD, Factor SA. Failure of recognition of drug-induced parkinsonism in the elderly. Move Disord. 2008;23:401–404. [DOI] [PubMed] [Google Scholar]
- 55.Koning JP, Tenback DE, van Os J, Aleman A, Kahn RS, van Harten PN. Dyskinesia and parkinsonism in antipsychotic-naive patients with schizophrenia, first-degree relatives and healthy controls: a metaanalysis. Schizophr Bull. 2010;36:723–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Balazs J, Besnyo M, Gadoros J. Methylphenidate-induced orofacial and extremity dyskinesia. J Child Adolesc Psychopharmacol. 2007;17:378–381. [DOI] [PubMed] [Google Scholar]
- 57.Correll CU, Kane JM. One-year incidence rates of tardive dyskinesia in children and adolescents treated with second-generation antipsychotics: a systematic review. J Child Adolesc Psychopharmacol. 2007;17:647–656. [DOI] [PubMed] [Google Scholar]
- 58.Weiden PJ. EPS profiles: the atypical antipsychotics are not all the same. J Psychiatr Pract. 2007;13:13–24. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.