Hostname: page-component-76fb5796d-9pm4c Total loading time: 0 Render date: 2024-04-26T23:06:50.128Z Has data issue: false hasContentIssue false

Short-term efficacy and safety of lurasidone versus placebo in antipsychotic-naïve versus previously treated adolescents with an acute exacerbation of schizophrenia

Published online by Cambridge University Press:  24 March 2022

Christoph U. Correll*
Affiliation:
Department of Psychiatry, Northwell Health, The Zucker Hillside Hospital, Glen Oaks, New York, USA Department of Psychiatry and Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
Michael Tocco
Affiliation:
Global Medical Affairs, Sunovion Pharmaceuticals Inc., Fort Lee, New Jersey, USA Sunovion Pharmaceuticals Inc., Marlborough, Massachusetts, USA
Jay Hsu
Affiliation:
Global Medical Affairs, Sunovion Pharmaceuticals Inc., Fort Lee, New Jersey, USA Sunovion Pharmaceuticals Inc., Marlborough, Massachusetts, USA
Robert Goldman
Affiliation:
Global Medical Affairs, Sunovion Pharmaceuticals Inc., Fort Lee, New Jersey, USA Sunovion Pharmaceuticals Inc., Marlborough, Massachusetts, USA
Andrei Pikalov
Affiliation:
Global Medical Affairs, Sunovion Pharmaceuticals Inc., Fort Lee, New Jersey, USA Sunovion Pharmaceuticals Inc., Marlborough, Massachusetts, USA
*
*Author for correspondence: Christoph U. Correll, E-mail: ccorrell@northwell.edu

Abstract

Background

To evaluate the efficacy of short-term lurasidone in antipsychotic treatment-naïve (TN) adolescents with schizophrenia versus those treated previously (TP) with antipsychotics.

Methods

Patients aged 13–17 with schizophrenia, and a Positive and Negative Symptom Scale (PANSS) score ≥ 70 and < 120, were randomized to 6 weeks of double-blind treatment with lurasidone (40 or 80 mg/day) or placebo. In a post-hoc, pooled-dose analysis, efficacy was evaluated for TN (criteria: never received antipsychotic treatment) versus TP at the time of the study. Treatment response criteria: ≥20% reduction in PANSS total score.

Results

Altogether, 57 TN and 269 TP patients enrolled in the 6-week DB study. Mean endpoint change in PANSS total score was significantly greater for lurasidone versus placebo in both the TN group (−25.0 vs. -14.4; p < 0.02; effect size = 0.75), and in the TP group (−17.3 vs. -10.0; p < 0.001; effect size = 0.45); and responder rates were higher for lurasidone versus placebo in both the TN group 84.6% versus 38.9%; number needed to treat [NNT] = 3 and in the TP group (60% vs. 42%; NNT = 6). Rates of treatment-emergent adverse events, and mean changes in body weight and metabolic parameters were similar for the TN and TP groups.

Conclusions

In a 6-week, placebo-controlled trial, lurasidone demonstrated significant efficacy in adolescents with schizophrenia regardless of previous antipsychotic therapy status; however, the effect size was notably larger in the TN patient group. In both the TN and TP groups, minimal effects were noted on weight, metabolic parameters, or prolactin.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of the European Psychiatric Association

Introduction

Schizophrenia is a debilitating neurodevelopmental disorder that typically emerges in late adolescence and early adulthood [Reference Kahn, Sommer, Murray, Meyer-Lindenberg, Weinberger and Cannon1]. The disorder is estimated to affect approximately 21 million people worldwide and is characterized by substantial ongoing disability [2, 3]. The lifespan of individuals with schizophrenia is estimated to be 14.5 years shorter on average compared to the general population due to various factors, including notably higher rates of tobacco smoking, alcohol/drug use, cardiovascular disease, diabetes, and poor dietary habits [Reference Lasser, Boyd, Woolhandler, Himmelstein, McCormick and Bor4Reference Hjorthoj, Sturup, McGrath and Nordentoft10].

Meta-analytic evidence indicates that a younger age at onset of schizophrenia is associated with more hospitalizations, more negative symptoms, more relapses, and poorer social/occupational functioning and global outcome [Reference Immonen, Jääskeläinen, Korpela and Miettunen11]. However, there is also some conflicting evidence indicating that factors other than earlier age of onset of schizophrenia determine poorer outcomes [Reference Vernal, Boldsen, Lauritsen, Correll and Nielsen12]. Outcome predictors aside, it is especially important to establish the clinical efficacy and safety of antipsychotic therapies during the developmentally sensitive biopsychosocial phase of adolescence. Although atypical antipsychotics are recommended first-line treatments for schizophrenia, concerns about weight gain, risk of diabetes, and metabolic problems associated with many of the approved atypical antipsychotics is an important public health concern [Reference De Hert, Detraux, van Winkel, Yu and Correll13Reference Perez Rodriguez, Tajima-Pozo, Lewczuk and Montañes-Rada16]. These concerns are particularly relevant for children and adolescents where weight gain and the risk of developing hyperglycemia, hyperlipidemia, hyperprolactinemia, and diabetes with atypical antipsychotics has been extensively documented [Reference Galling, Roldán, Nielsen, Nielsen, Gerhard and Carbon17Reference De Hert, Dobbelaere, Sheridan, Cohen and Correll21].

Given the typical onset of illness, many adolescents with a diagnosis of schizophrenia may be experiencing their first episode and have never received antipsychotic medication. Not receiving antipsychotic medication for an extended period after diagnosis of psychosis has been associated with structural changes in the brain and relatively poor clinical outcomes [Reference Loebel, Lieberman, Alvir, Mayerhoff, Geisler and Szymanski22Reference Zhu, Krause, Huhn, Rothe, Schneider-Thoma and Chaimani27]. Several studies have examined the efficacy of different antipsychotics among first-episode and treatment-naïve (TN) adult patients [Reference Sanger, Lieberman, Tohen, Grundy, Beasley and Tollefson28Reference Sanz-Fuentenebro, Taboada, Palomo, Aragües, Ovejero and Del Alamo35]. However, whether or not these findings generalize to an adolescent population is uncertain. For example, there is some evidence that TN adolescent onset schizophrenia may have a different pattern of cortical gray matter deficits compared with TN adult onset schizophrenia [Reference Torres, Duran, Schaufelberger, Crippa, Louzã and Sallet36,Reference Zhang, Wang, Ni, Deng, Li and Zhao37]. Drug response of TN adolescents with schizophrenia (or previously treated) may therefore differ from TN adults with schizophrenia.

The atypical antipsychotic agent lurasidone shows high binding affinity for D2, 5-HT2A, and 5-HT7 receptors (antagonist); moderate affinity for 5-HT1A receptors (partial agonist); and no appreciable affinity for H1 and M1 receptors [Reference Ishibashi, Horisawa, Tokuda, Ishiyama, Ogasa and Tagashira38]. Efficacy and safety of lurasidone in the acute and long-term treatment of adults with schizophrenia in the dose range of 40–160 mg/day has been demonstrated across multiple studies [Reference Nakamura, Ogasa, Guarino, Phillips, Severs and Cucchiaro39Reference Tandon, Cucchiaro, Phillips, Hernandez, Mao and Pikalov46]. A low propensity for weight gain and metabolic disturbance was consistently found across these lurasidone trials [Reference Loebel and Citrome47,Reference Tocco, Newcomer, Mao and Pikalov48]. The absence of activity for lurasidone at 5HT2C and histamine H1 receptors is thought to be responsible for the minimal observed effect on weight [Reference Kroeze, Hufeisen, Popadak, Renock, Steinberg and Ernsberger49Reference Lord, Wyler, Wan, Castorena, Ahmed and Mathew51]. In a previously reported 6-week, double-blind, placebo-controlled, fixed-dose trial, treatment with 40 and 80 mg/day doses of lurasidone were found to be safe and generally well-tolerated, and to have significant efficacy in adolescents with schizophrenia [Reference Goldman, Loebel, Cucchiaro, Deng and Findling52]. Based on the efficacy and safety results from this short-term study, the results of a pharmacokinetic study [Reference Findling, Goldman, Chiu, Silva, Jin and Pikalov53], and a subsequent long-term study in adolescents [Reference Correll, Findling, Tocco, Pikalov, Deng and Goldman54], lurasidone has been approved, in doses of 40–80 mg/day, by the U.S. Food and Drug Administration for the treatment of schizophrenia in youth 13–17 years old.

Few studies have examined treatment response in adolescents with schizophrenia who are TN compared to previously treated adolescents; and no study (to the best of our knowledge) has examined this question in the context of a placebo-controlled study, either in adolescents or in adults. The objective of the current post-hoc analysis was to evaluate the short-term efficacy and safety of lurasidone in TN adolescent patients with a diagnosis of schizophrenia compared with a previously treated group.

Based on previous data indicating that first-episode and TN patients generally respond better to antipsychotics than patients who have been ill for longer [Reference Carbon and Correll26, Reference Zhu, Krause, Huhn, Rothe, Schneider-Thoma and Chaimani27, Reference Correll55], but are also more sensitive to adverse effects of antipsychotics [Reference Correll, Manu, Olshanskiy, Napolitano, Kane and Malhotra19Reference De Hert, Dobbelaere, Sheridan, Cohen and Correll21], we hypothesized that short-term treatment with lurasidone in antipsychotic-naïve patients would be associated with greater efficacy and adverse effect burden (relative to placebo) than in previously treated adolescents.

Methods

The study used in the current post-hoc analysis was a double-blind (DB), parallel-group, placebo-controlled, multicenter trial that randomized patients (1:1:1) to 6 weeks of fixed-dose treatment with lurasidone (40 or 80 mg/day) or placebo. Eligible patients were aged 13–17 years with a Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (Text Revision; DSM-IV-TR) diagnosis of schizophrenia who were experiencing an acute exacerbation (≤2 months in duration) of symptoms defined by a Positive and Negative Syndrome Scale (PANSS) [Reference Kay, Fiszbein and Opler56] total score ≥70 and a Clinical Global Impression-Severity (CGI-S) [Reference Guy57] score ≥ 4 (at least moderately ill). Patients were excluded if they had a history of intellectual disability or any neurologic disorder; or an alcohol or substance use disorder diagnosis in the previous 6 months. Additional details on study design and study entry criteria may be found in the primary publication [Reference Goldman, Loebel, Cucchiaro, Deng and Findling52].

The study protocol and any amendments were reviewed and approved by institutional review boards at each investigational site. Written informed consent was obtained from a parent or legal guardian, and assent was obtained from each adolescent patient prior to the conduct of any study procedures.

Concomitant medication

Concomitant treatment with antidepressants and stimulants (for ADHD) was permitted. Concomitant use of lorazepam or equivalent benzodiazepine was permitted at the discretion of the investigator (≤6 mg/day or equivalent dose) for intolerable anxiety/agitation. Benzodiazepine and nonbenzodiazepine sedative-hypnotic agents were also permitted on an as-needed basis for insomnia. Treatment with benztropine (≤6 mg/day) or alternative medications was permitted as needed for movement disorders and treatment with propranolol (≤120 mg/day) was permitted as needed for akathisia. However, prophylactic use of medications to treat movement disorders was not permitted.

Study assessments

Efficacy

Efficacy measures included the PANSS total score and the PANSS Positive and Negative subscales [Reference Kay, Fiszbein and Opler56]; the CGI-S; the clinician-rated Children’s Global Assessment Scale (CGAS) [Reference Shaffer, Gould, Brasic, Ambrosini, Fisher and Bird58]; and the Pediatric Quality of Life Enjoyment and Satisfaction Questionnaire (PQ-LES-Q) [Reference Endicott, Nee, Yang and Wohlberg59]. The CGAS is a clinician-administered measure that evaluates global impairment on a scale of 0–100, with higher scores indicating better outcomes. The PQ-LES-Q is a quality of life measure that has demonstrated reliability and validity in youth [Reference Endicott, Nee, Yang and Wohlberg59]. Efficacy and safety assessments were performed by trained site-based raters. Prior treatment status was obtained from self-report by caregiver and patient in response to standard questions during the screening visit for the DB study.

Safety and tolerability assessments

Adverse events were recorded based on spontaneous report and also by administration of the Udvalg for Kliniske Undersogelser (UKU) Side Effect Rating Scale, a clinician-rated scale consisting of 48 adverse events divided into four categories (psychic, neurologic, autonomic, and other) [Reference Lingjaerde, Ahlfors, Bech, Dencker and Elgen60]. Mean severity scores (1—mild to 4—severe) were calculated for the total score and each side effect category. Movement disorders were assessed by three scales: the Simpson–Angus Scale (SAS) [Reference Simpson and Angus61], the Barnes Akathisia Rating Scale (BARS) [Reference Barnes62], and the Abnormal Involuntary Movement Scale (AIMS) [Reference Guy57, Reference Munetz and Benjamin63]. Suicidal ideation and behavior were measured with the Columbia Suicide Severity Rating Scale (C-SSRS) [Reference Posner, Brown, Stanley, Brent, Yershova and Oquendo64]. Additional safety evaluations included vital signs, weight, laboratory tests (metabolic parameters and other blood chemistry and hematology parameters), 12-lead electrocardiogram (ECG), and physical examination.

Statistical analyses

The efficacy population was defined as all patients enrolled who received at least one dose of study medication in the trial and had at least one postbaseline efficacy assessment. Efficacy measures were examined in terms of least squared mean (SD) change from DB study baseline to week 6 or last observation carried forward (LOCF) endpoint, depending on statistical methodology used. Efficacy at week 6 (or LOCF endpoint) was compared for lurasidone (both doses combined) versus placebo in two patient subgroups based on prior treatment status: TN versus previously treated. TN was defined as never having been treated with an antipsychotic medication prior to study entry; and patients in the previously treated subgroup had been treated with an antipsychotic medication prior to study entry. Note that the data on the number and adequacy of previous antipsychotic medication trials was not available.

Change scores were analyzed using a mixed model for repeated measurement (MMRM) analysis or analysis of covariance (ANCOVA) using an LOCF approach. Effect sizes (ES) were calculated as the least squares mean difference in week 6 (or endpoint) change score for lurasidone versus placebo divided by the pooled standard deviation. Standardized mean difference in endpoint change scores (lurasidone vs. placebo) were also calculated for the TN versus previously treated groups. Treatment response was calculated, based on LOCF-endpoint data, using both ≥20% and ≥50% improvement from double-blind baseline in PANSS total score. The number-needed-to-treat (NNT) was calculated as the reciprocal of the difference in response rates for lurasidone versus placebo at LOCF-endpoint. All significance tests were two-tailed with alpha = 0.05. Baseline differences in the demographic and clinical characteristics of the TN and previously treated groups (for the combined study treatments) were analyzed using Fishers’ exact test (for sex and race), two-sample t-tests (age, age of onset, duration of current episode, PANSS total score, CGI-S score, CGAS score, and Q-LES-Q score), and chi-square (prior hospitalization).

The safety population was defined as all patients who were enrolled who received at least one dose of lurasidone in that study. Safety analyses were descriptive and included the number (%) of treatment-emergent adverse events, discontinuations due to adverse events, and use of medications for acute extrapyramidal symptoms. Means with 95% confidence intervals (CIs), or NNH were reported as appropriate. Observed case analyses were calculated for change from double-blind baseline for safety variables, including weight, laboratory tests, ECG parameters (including QTcF, Fridericia’s formula), and movement disorder scale scores (SAS, BARS, AIMS).

Results

Patient disposition and characteristics

At DB baseline, there were 57 TN patients and 269 previously treated patients (Figure 1). Of these, 50 (87.7%) TN and 221 (82.2%) previously treated patients completed the 6-week DB study.

Figure 1. Change from double-blind baseline in PANSS total score.

Table 1 presents demographic and clinical characteristics of patients at DB baseline, separately for the TN group and previously treated group. In general, the baseline characteristics of the TN and previously treated groups were similar, with the following notable exceptions: (a) the proportion of patients with prior hospitalizations for schizophrenia was 58% in the previous treatment group and 30% in the TN group (p < 0.001); (b) the CGAS score was statistically significantly lower (p < 0.001; more functional impairment) in the previous treatment group compared to the TN group; and (c) the CGI-Severity score was higher in the previous treatment group compared to the TN group. It should be noted that the between-group differences in the CGAS and CGI-S, while statistically significant were of modest clinical significance.

Table 1. Patient characteristics at double-blind baseline.

Note: Significance testing of between-group difference in baseline characteristics for previously treated versus previously treated groups.

Abbreviations: CGAS, clinical global assessment scale; CGI-S, clinical global impression-severity; PANSS, positive and negative syndrome scale; PQ-LES-Q, pediatric quality of life, enjoyment and satisfaction questionnaire; SD, standard deviation.

*P < 0.05: CGI-S; **P < 0.001. Prior hospitalization and CGAS.

a Mean percentage of maximum possible score.

Efficacy for TN and previously treated patients

Compared to placebo, treatment with lurasidone was associated with significantly greater improvement from DB baseline to week 6 (MMRM) in the PANSS total score for both the TN (p = 0.016; ES = 0.75) and previously treated groups (p = 0.0008; ES = 0.45; Table 2). Within the TN group, there was significantly greater improvement for lurasidone, compared to placebo on the PANSS Positive Symptom score (p = 0.0045; ES = 0.89), but not the PANSS Negative Symptom score (p = 0.18; ES = 0.40; Table 2). Within the previously treated group, treatment with lurasidone was associated with greater improvement versus placebo in the PANSS Positive Symptom score (p < 0.0001; ES = 0.57) and in the Negative Symptoms score (p = 0.017; ES = 0.32) at week 6 (MMRM; Table 2). Among TN patients, the proportion of lurasidone versus placebo responders was 84.6% versus 38.9% (NNT = 3) based on the ≥ 20% PANNS improvement criterion, and 30.8% versus 8.5% (NNT = 7) based on the ≥ 50% PANNS improvement criterion. Among previously treated patients, the proportion of lurasidone versus placebo responders was 60.0% versus 42.6% (NNT = 6) based on the ≥20% PANNS improvement criterion, and 18.9% versus 8.5% (NNT = 10) based on the ≥50% PANNS improvement criterion.

Table 2. Mean (SE) change from DB baseline on efficacy measures.

Note: For PANSS scales and CGI-S scores are estimated least square mean change to week 6 derived from mixed model for repeated measurements. For CGAS and PQ-LES-Q, scores are change from baseline to Week 6 LOCF endpoint derived from ANCOVA analyses.

Abbreviations: CGAS, clinical global assessment scale; CGI-S, clinical global impression-severity; PANSS, positive and negative syndrome scale; PQ-LES-Q, pediatric quality of life, enjoyment and satisfaction questionnaire; SD, standard deviation; SE, standard error.

*p < 0.05; **p < 0.01; ***p < 0.001, in comparison to placebo.

a Mean percentage of maximum possible score.

Significantly greater improvement on the CGI-Severity scale was also observed for lurasidone-treated patients compared to placebo in both the TN group (p = 0.0023; ES = 0.97) and previously treated group (p = 0.005; ES = 0.38; Table 2). Lurasidone treatment (vs. placebo) was associated with slightly larger endpoint effect sizes in the TN group compared to the previously treated group on the Children’s Global Assessment Scale (ES = 0.55 vs. 0.43) and on the PQ-LES-Q (ES = 0.54 vs. 0.47), though significant differences from placebo were not obtained for the TN group due to reduced statistical power as a result of smaller sample size (Table 2). With the larger sample size in the previously treated group, significantly greater improvement for lurasidone, compared to placebo, was obtained on both the CGAS (p = 0.0012) and PQ-LES-Q (p = 0.0004).

Safety and tolerability for TN and previously treated patients

The rates of TEAEs in lurasidone-treated patients were similar for the TN group and the previously treated groups (Table 3). Within the TN group, lurasidone-treated patients reported nausea, anxiety, akathisia, vomiting, and somnolence at 10% or greater incidence and rates twice (or greater) than that found in the placebo group. In the previously treated group, lurasidone-treated patients reported nausea, akathisia, and vomiting at 10% incidence and rates twice (or greater) than that found among placebo-treated patients. Within the TN group, the incidence of extrapyramidal symptom-related adverse events (excluding akathisia) was 5.1% (n = 2) in the lurasidone group and 0% in the placebo group (Table 3). Among those that were not TN, 9 (5.1%) lurasidone-treated patients and 2 (2.1%) placebo-treated patients reported extrapyramidal symptom-related adverse events (excluding akathisia).

Table 3. Adverse events during 6-weeks of double-blind, placebo-controlled treatment.

Treatment-emergent adverse events (≥ 5% on lurasidone and greater than placebo).

Abbreviations: NNH, number-needed-to-harm; TEAE, treatment-emergent adverse event.

On the UKU, change from baseline to endpoint in the four side effect categories (psychic, neurologic, autonomic, and other) was generally similar between the pooled lurasidone dose groups and the placebo group for the TN group, and also for the previously treated group (Table 4). Within the TN group, the proportion of patients with treatment-emergent suicidal ideation, measured by the C-SSRS, was 7.7% (n = 3) in the lurasidone group and 5.6% (n = 1) in the placebo group. For the previously treated group, 2.3% (n = 4) of patients were found to have treatment-emergent suicidal ideation on the CSSR in the lurasidone group compared to 4.3% (n = 4) in the placebo group. No occurrence of suicidal behavior or completed suicide was evident in either the TN or previously treated groups.

Table 4. Udvalgfor Kliniske Undersogelser Side Effect Rating Scale Scores:Mean (SD) Baseline-to-EndpointChange.

Note: Higher scores indicate greater severity; range of 0–30 for psychic, 0–24 for neurologic, 0–33 f for autonomic, and 0–48 for other. Udvalg for Kliniske Undersogelser side effect rating scale scores: mean (SD) baseline-to-endpoint change.

Abbreviations: SD, standard deviation; UKU, Udvalg for Kliniske Undersogelser.

Among TN patients, the proportion who received anticholinergic medications for acute extrapyramidal symptoms in the lurasidone versus placebo groups were 2.6% versus 0%; and the proportion who received benzodiazepines were 17.9% versus 16.7%. Among previously treated patients, the proportion who received anticholinergic medications for acute extrapyramidal symptoms in the lurasidone versus placebo groups were 4.0% versus 2.1%; and the proportion who received benzodiazepines were 21.1% versus 24.5%).

No clinically meaningful mean changes from baseline to endpoint were evident in the TN group for lurasidone or placebo, respectively, for the SAS (0.03 vs. 0.02), BARS (0.4 vs. 0.1), and AIMS (0.3 vs. 0.1). Similarly, no clinically meaningful mean changes from baseline to endpoint were evident in the previously treated group for lurasidone or placebo for the SAS (0.01 vs. 0.01), BARS (0.0 vs. 0.0), and AIMS (0.0 vs. 0.0).

Laboratory measures of lipid parameters, glycemic indices, and prolactin levels also showed no clinically meaningful differences between lurasidone and placebo for the TN and for the previously treatment group (Table 5). Within the TN group, there was one patient in the lurasidone group, and none in the placebo group, that experienced a clinically meaningful (≥ 7%) increase in body weight. Within the previously treated group, there were 8 (4.6%) patients in the lurasidone group and 9 (9.6%) in the placebo group with clinically meaningful weight change. No patients had a QTcF ≥ 460 milliseconds or a postbaseline change in QTcF ≥ 60 ms in either the TN group or the previously treated group.

Table 5. Change from double-blind baseline in laboratory values and weight/BMI.

Discussion

To the best of our knowledge, this is the first placebo-controlled study that has examined the efficacy and safety of an atypical antipsychotic in TN adolescents (or adults) with schizophrenia. The results of this post-hoc analysis suggest that lurasidone is an effective treatment option for both antipsychotic-naïve adolescents diagnosed with schizophrenia and adolescents previously treated with antipsychotic medication. The magnitude of the treatment effect (lurasidone vs. placebo), measured by effect sizes at endpoint, was greater for TN versus previously treated patients on the PANSS total score (ES = 0.75 vs. 0.45). Similarly, the standardized mean difference score was also greater for TN versus previously treated patients on the PANSS total score. These results compare favorably to results of a previous meta-analysis of acute antipsychotic treatment of adolescents with schizophrenia-spectrum disorders, where the standard mean difference (SMD) scores ranged from −0.34/−0.38 (aripiprazole/asenapine) up to −0.57/−0.59 (risperidone/olanzapine) [Reference Pagsberg, Tarp, Glintborg, Stenstrøm, Fink-Jensen and Correll65]. In addition, in a network meta-analysis comparing atypical antipsychotics in the treatment of adolescent schizophrenia [Reference Arango, Ng-Mak, Finn, Byrne and Loebel66], treatment with lurasidone had comparable efficacy to other atypical antipsychotics, but lower risk of all-cause discontinuation than aripiprazole and paliperidone and lower risk of weight gain than all other included atypical antipsychotics, except for aripiprazole and ziprasidone.

Currently, regulatory approval of lurasidone in the United States for adolescents with schizophrenia is limited to doses of 40–80 mg/day. However, the results of a recent analysis [Reference Costamagna, Calisti, Cattaneo, Hsu, Tocco and Pikalov67] of young adults (ages 18–25) with schizophrenia reported that use of higher doses (120–160 mg/day) is associated with a notable increase in effect size. For example, the effect size of lurasidone 80 mg/day (vs. placebo) on the PANSS total score increased from 0.57 to 0.90 for lurasidone 120–180 mg/day. It is possible that lower effect sizes observed in the current study in the previously treated adolescent patient group may be largely dose-related and respond to use of higher doses.

Lurasidone was generally well-tolerated in both TN and previously treated adolescents with schizophrenia. Notably, in the TN group, only two AEs occurred with an NNH < 10 (anxiety, NNH = 8; akathisia, NNH = 8), and only one AE in the previously treatment group (nausea, NNH = 9). This relatively favorable tolerability profile in TN patients, who are generally more sensitive to adverse effects, especially weight gain [Reference Arango, Giráldez, Merchán-Naranjo, Baeza, Castro-Fornieles and Alda18–20], confirms the recent finding from a large systematic review that found lurasidone to be the most well-tolerated antipsychotic when comparing adverse effects versus placebo in pediatric patients [Reference Solmi, Fornaro, Ostinelli, Zangani, Croatto and Monaco68].

Given that the current results are based on post-hoc analyses with limited sample sizes, additional data from prospective studies are needed before any firm conclusions may be made. However, the finding that the efficacy of lurasidone among TN patients is at least as strong, if not stronger, than that found for previously treated patients has potentially important implications. For one, these results suggest that antipsychotic efficacy among TN adults [Reference Sanger, Lieberman, Tohen, Grundy, Beasley and Tollefson28Reference Sanz-Fuentenebro, Taboada, Palomo, Aragües, Ovejero and Del Alamo35, Reference Robinson, Gallego, John, Petrides, Hassoun and Zhang69] may extend to TN adolescents, despite research indicating that the extent and pattern of gray matter deficits in early onset schizophrenia may be different from the deficits observed in adult-onset schizophrenia [Reference Torres, Duran, Schaufelberger, Crippa, Louzã and Sallet36, Reference Zhang, Wang, Ni, Deng, Li and Zhao37]. Further prospective placebo-controlled studies of lurasidone and other antipsychotics are needed to confirm efficacy among TN adolescents with schizophrenia.

A second potential implication of the current findings is that TN adolescent patients appear to respond especially well to early treatment intervention with lurasidone, while not receiving early treatment may result in a relatively poor clinical outcome that may include a greater degree of brain structural changes [Reference Loebel, Lieberman, Alvir, Mayerhoff, Geisler and Szymanski22Reference Guo, Li, Wei, Fan, Kennedy and Shen25]. Shortening the duration of untreated psychosis by earlier antipsychotic intervention is likely to improve outcomes in youth with early-onset schizophrenia [Reference Harrigan, McGorry and Krstev70, Reference Souaiby, Gaillard and Krebs71].

Alternatively, one might hypothesize that the relatively greater treatment effect observed for the TN versus previously treated group (effect size: 0.75 vs. 0.45) is largely attributable to a reduced response rate in the previously treated group. This relative treatment resistance might be due to the progressive nature of the schizophrenic illness since chronicity is a well-established predictor of reduced antipsychotic response [Reference Clemmensen, Vernal and Steinhausen72]. It is also possible that the potency of antipsychotic drugs is diminished across repeated courses of treatment, though this treatment effect might be difficult to disentangle from the effects of chronicity.

In the current study, lurasidone was found to have minimal effects on weight, prolactin, and metabolic measures, even in the TN subgroup, although with other atypical antipsychotics treatment-naïveté has previously been shown to be a strong risk factor for enhanced cardiovascular risk [Reference Correll, Manu, Olshanskiy, Napolitano, Kane and Malhotra19]. Since adolescents with a diagnosis of schizophrenia (vs. non-schizophrenia controls) are an especially high-risk population, with a long-term outcome characterized by increased cardiovascular morbidity and mortality, and a significantly shorter life span (~20 years) [Reference Firth, Siddiqi, Koyanagi, Siskind, Rosenbaum and Galletly15, Reference Correll, Solmi, Veronese, Bortolato, Rosson and Santonastaso73], choice of antipsychotic should include consideration of metabolic adverse effects [Reference De Hert, Dobbelaere, Sheridan, Cohen and Correll21, Reference Laursen, Nordentoft and Mortensen74, Reference Solmi, Murru, Pacchiarotti, Undurraga, Veronese and Fornaro75].

Several limitations of this study should be noted. First, the comparisons of TN and previously treated patients were based on post-hoc analyses, and thus should be considered exploratory. Second, the inclusion and exclusion criteria may limit the generalizability of these results. Third, the 6-week duration of the study does not allow for a comparison versus placebo over longer periods if time when, for example, body weight changes might accumulate. However, antipsychotic-related weight gain generally occurs early and predicts later weight gain [Reference Correll, Manu, Olshanskiy, Napolitano, Kane and Malhotra19, Reference Vandenberghe, Najar-Giroud, Holzer, Conus, Eap and Ambresin76]. Furthermore, longer exposure to placebo is ethically questionable. Also, it should be noted that data in 271 patients who entered the 2-year open-label extension study of the current randomized trial found that efficacy advantages in the TN (vs. previously treated) population extended into the long-term treatment phase and that the tolerability was also maintained in both patient groups [Reference Correll, Findling, Tocco, Pikalov, Deng and Goldman54]. Fourth, the sample size of the antipsychotic-naïve adolescent group was relatively modest; however, to the best of our knowledge, this is the first sample of antipsychotic-naïve patients with schizophrenia, adolescent or adult, that has been part of a placebo in a double blind, placebo-controlled trial of an antipsychotic. A final limitation of the current post-hoc analysis was that medical records were not obtained for patients entering the current clinical trial. Consequently, we do not have reliable information on adequacy of previous treatment (dosing or duration) or response to previous treatment. Additional studies that more fully characterize the previous treatment history are needed to evaluate the efficacy and safety of lurasidone and other antipsychotics among TN and previously treated adolescents with schizophrenia.

In conclusion, the current post-hoc analysis found lurasidone (40 and 80 mg/day) to be a safe, well-tolerated, and effective short-term treatment in adolescents with schizophrenia, regardless of prior treatment history. However, notably larger treatment effect sizes were observed in the subgroup of patients who had never received antipsychotic therapy compared to patients with previous antipsychotic exposure. Consistent with previously reported safety findings in both adult and pediatric populations, no clinically meaningful changes in weight, metabolic parameters, and prolactin were evident with lurasidone treatment. However, in contrast to our hypothesis based on results from studies of other antipsychotics, favorable tolerability was also observed for lurasidone in the TN subgroup, despite previous findings that this subgroup is generally more sensitive and adverse effect prone. This favorable short-term benefit–risk profile suggests that lurasidone should be considered as a potential first-line treatment for the acute management of adolescents with schizophrenia. Complementary and consistent data from the long-term open-label extension study of this trial indicate that the first-line status for lurasidone extends from the acute into the longer-term management of adolescents with schizophrenia.

Data Availability Statement

Data are available upon request from Dr. Michael Tocco ().

Acknowledgments

The sponsor was involved in the design of the original study, and the collection and analysis of the data. The authors of these post-hoc analyses interpreted the post-hoc data and made the decision to submit the article for publication. Medical writing support was provided by Edward Schweizer, MD, of Paladin Consulting Group, Inc., Princeton, NJ and was funded by Sunovion Pharmaceuticals, Inc.

Author Contributions

Conceptualization: C.U.C., M.T.; Data interpretation: C.U.C., M.T., R.G., A.P.; Manuscript development and editing: C.U.C., M.T., J.H., R.G., A.P.; Statistical analysis: J.H.

Financial Support

This work was supported by funding from Sunovion Pharmaceuticals Inc.

Conflicts of Interest

Dr. Correll has served as a consultant and/or adviser to or has received honoraria from Alkermes, Allergan, Angelini, Boehringer Ingelheim, Bristol-Myers Squibb, Gerson Lehrman Group, Indivior, Intra-Cellular Therapies, Janssen/Johnson & Johnson, LB Pharma, Lundbeck, Medavante-ProPhase, Medscape, Merck, Neurocrine, Noven, Otsuka, Pfizer, Rovi, Servier, Sunovion, Supernus, Takeda, and Teva. He has provided expert testimony for Bristol-Myers Squibb, Janssen, and Otsuka. He has served on data safety monitoring boards for Boehringer Ingelheim, Lundbeck, Pfizer, Rovi, Supernus, and Teva. He has received grant support from Janssen and Takeda. And, he is a shareholder in LB Pharma. Drs. Goldman, Tocco, Pikalov, Hsu, and Loebel are employees of Sunovion Pharmaceuticals Inc.

References

Kahn, RS, Sommer, IE, Murray, RM, Meyer-Lindenberg, A, Weinberger, DR, Cannon, TD, et al. Schizophrenia. Nat Rev Dis Primers. 2015;1:15067. doi:10.1038/nrdp.2015.67.CrossRefGoogle ScholarPubMed
World Health Organization. Schizophrenia fact sheet, http://www.who.int/mediacentre/factsheets/fs397/en/; 2016.Google Scholar
GBD. Disease and injury incidence and prevalence collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2017;392:1789–858. doi:10.1016/S0140-6736(18)32279-7.Google Scholar
Lasser, K, Boyd, JW, Woolhandler, S, Himmelstein, DU, McCormick, D, Bor, DH. Smoking and mental illness: a population-based prevalence study. JAMA. 2000;284:2606–10. doi:10.1001/jama.284.20.2606.CrossRefGoogle ScholarPubMed
Dipasquale, S, Pariante, CM, Dazzan, P, Aguglia, E, McGuire, P, Mondelli, V. The dietary pattern of patients with schizophrenia: a systematic review. J Psychiatr Res. 2013;47:197207. doi:10.1017/S0033291711002947.CrossRefGoogle ScholarPubMed
Hjorthøj, C, Østergaard, ML, Benros, ME, Toftdahl, NG, Erlangsen, A, Andersen, JT, et al. Association between alcohol and substance use disorders and all-cause and cause-specific mortality in schizophrenia, bipolar disorder, and unipolar depression: a nationwide, prospective, register-based study. Lancet Psychiatry. 2015;2:801–8. 10.1016/S2215-0366(15)00207-2.CrossRefGoogle ScholarPubMed
Stubbs, B, Vancampfort, D, De Hert, M, Mitchell, AJ. The prevalence and predictors of type two diabetes mellitus in people with schizophrenia: a systematic review and comparative meta-analysis. Acta Psychiatr Scand. 2015;132:144–57. doi: 10.1111/acps.12439.CrossRefGoogle ScholarPubMed
Azad, MC, Shoesmith, WD, Al Mamun, M, Abdullah, AF, Naing, DKS, Phanindranath, M, et al. Cardiovascular diseases among patients with schizophrenia. Asian J Psychiatr. 2016;19:2836. doi:10.1016/j.ajp.2015.11.012.CrossRefGoogle ScholarPubMed
Toftdahl, NG, Nordentoft, M, Hjorthøj, C. Prevalence of substance use disorders in psychiatric patients: a nationwide Danish population-based study. Soc Psychiatry Psychiatr Epidemiol. 2016;51:129–40. doi:10.1007/s00127-015-1104-4.CrossRefGoogle ScholarPubMed
Hjorthoj, C, Sturup, AE, McGrath, JJ, Nordentoft, M. Years of potential life lost and life expectancy in schizophrenia: a systematic review and meta-analysis. Lancet Psychiatry. 2017;4:295301. doi:10.1016/S2215-0366(17)30078-0.CrossRefGoogle ScholarPubMed
Immonen, J, Jääskeläinen, E, Korpela, H, Miettunen, J. Age at onset and the outcomes of schizophrenia: a systematic review and meta-analysis. Early Interv Psychiatry. 2017;11:453–60. doi:10.1111/eip.12412.CrossRefGoogle ScholarPubMed
Vernal, DL, Boldsen, SK, Lauritsen, MB, Correll, CU, Nielsen, RE. Long-term outcome of early-onset compared to adult-onset schizophrenia: a nationwide Danish register study. Schizophr Res. 2020;220:123–9. doi:10.1016/j.schres.2020.03.045.CrossRefGoogle ScholarPubMed
De Hert, M, Detraux, J, van Winkel, R, Yu, W, Correll, CU. Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nat Rev Endocrinol. 2011;8:114–26. doi:10.1038/nrendo.2011.156.CrossRefGoogle ScholarPubMed
Correll, CU, Lencz, T, Malhotra, AK. Antipsychotic drugs and obesity. Trends Mol Med. 2011;17:97107. doi:10.1016/j.molmed.2010.10.010.CrossRefGoogle ScholarPubMed
Firth, J, Siddiqi, N, Koyanagi, A, Siskind, D, Rosenbaum, S, Galletly, C, et al. The lancet psychiatry commission: a blueprint for protecting physical health in people with mental illness. Lancet Psychiatry. 2019;6:675712. doi:10.1016/S2215-0366(19)30132-4.CrossRefGoogle ScholarPubMed
Perez Rodriguez, A, Tajima-Pozo, K, Lewczuk, A, Montañes-Rada, F. Atypical antipsychotics and metabolic syndrome. Cardiovasc Endocrinol Metab. 2015;4:132–7. doi:10.1097/XCE.0000000000000063.CrossRefGoogle Scholar
Galling, B, Roldán, A, Nielsen, RE, Nielsen, J, Gerhard, T, Carbon, M, et al. Type 2 diabetes mellitus in youth exposed to antipsychotics: a systematic review and meta-analysis. JAMA Psychiatry. 2016;73:247–59. doi:10.1001/jamapsychiatry.2015.2923.CrossRefGoogle ScholarPubMed
Arango, C, Giráldez, M, Merchán-Naranjo, J, Baeza, I, Castro-Fornieles, J, Alda, JA, et al. Second-generation antipsychotic use in children and adolescents: a six-month prospective cohort study in drug-naïve patients. J Am Acad Child Adolesc Psychiatry. 2014;53:1179–90.e1–4. doi:10.1016/j.jaac.2014.08.009.CrossRefGoogle ScholarPubMed
Correll, CU, Manu, P, Olshanskiy, V, Napolitano, B, Kane, JM, Malhotra, AK. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302:1765–73. doi:10.1001/jama.2009.1549.CrossRefGoogle ScholarPubMed
Jensen, KG, Correll, CU, Rudå, D, Klauber, DG, Stentebjerg-Olesen, M, Fagerlund, B, et al. Pretreatment cardiometabolic status in youth with early-onset psychosis: baseline results from the TEA trial. J Clin Psychiatry. 2017;78:e1035–46. doi:10.4088/JCP.15m10479.CrossRefGoogle ScholarPubMed
De Hert, M, Dobbelaere, M, Sheridan, EM, Cohen, D, Correll, CU. Metabolic and endocrine adverse effects of second-generation antipsychotics in children and adolescents: a systematic review of randomized, placebo-controlled trials and guidelines for clinical practice. Eur Psychiatry. 2011;26:144–58. doi:10.1016/j.eurpsy.2010.09.011.CrossRefGoogle Scholar
Loebel, AD, Lieberman, JA, Alvir, JM, Mayerhoff, DI, Geisler, SH, Szymanski, SR. Duration of psychosis and outcome in first-episode schizophrenia. Am J Psychiatry. 1982;149:1183–8. doi:10.1176/ajp.149.9.1183.Google Scholar
Crumlish, N, Whitty, P, Clarke, M, Browne, S, Kamali, M, Gervin, M, et al. Beyond the critical period: Longitudinal study of 8-year outcome in first-episode non-affective psychosis. Br J Psychiatry. 2009;194:1824. doi:10.1192/bjp.bp.107.048942.CrossRefGoogle ScholarPubMed
Owens, DC, Johnstone, EC, Miller, P, Macmillan, JF, Crow, TJ. Duration of untreated illness and outcome in schizophrenia: test of predictions in relation to relapse risk. Br J Psychiatry. 2010;196:296301. doi:10.1192/bjp.bp.109.067694.CrossRefGoogle ScholarPubMed
Guo, X, Li, J, Wei, Q, Fan, X, Kennedy, DN, Shen, Y, et al. Duration of untreated psychosis is associated with temporal and occipitotemporal gray matter volume decrease in treatment naive schizophrenia. PLoS One. 2013;8:e83679. doi:10.1371/journal.pone.0083679.CrossRefGoogle ScholarPubMed
Carbon, M, Correll, CU. Clinical predictors of therapeutic response to antipsychotics in schizophrenia. Dialogues Clin Neurosci. 2014;16:505–24. doi:10.31887/DCNS.2014.16.4/mcarbon.CrossRefGoogle Scholar
Zhu, Y, Krause, M, Huhn, M, Rothe, P, Schneider-Thoma, J, Chaimani, A, et al. Antipsychotic drugs for the acute treatment of patients with a first episode of schizophrenia: a systematic review with pairwise and network meta-analyses. Lancet Psychiatry. 2017;4:694705. doi:10.1016/S2215-0366(17)30270-5.CrossRefGoogle ScholarPubMed
Sanger, TM, Lieberman, JA, Tohen, M, Grundy, S, Beasley, C Jr, Tollefson, GD. Olanzapine versus haloperidol treatment in first-episode psychosis. Am J Psychiatry. 1999;156:7987. doi:10.1176/ajp.156.1.79.CrossRefGoogle ScholarPubMed
Lieberman, JA, Phillips, M, Gu, H, Stroup, S, Zhang, P, Kong, L, et al. Atypical and conventional antipsychotic drugs in treatment-naive first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology. 2003;28:9951003. doi:10.1038/sj.npp.1300157.CrossRefGoogle ScholarPubMed
Schooler, N, Rabinowitz, J, Davidson, M, Emsley, R, Harvey, PD, Kopala, L, et al. Risperidone and haloperidol in first-episode psychosis: a long-term randomized trial. Am J Psychiatry. 2005;162:947–53. doi:10.1176/appi.ajp.162.5.947.CrossRefGoogle ScholarPubMed
Green, AI, Lieberman, JA, Hamer, RM, Glick, ID, Gur, RE, Kahn, RS, et al. Olanzapine and haloperidol in first episode psychosis: two-year data. Schizophr Res. 2006;86:234–43. doi:10.1016/j.schres.2006.06.021.CrossRefGoogle ScholarPubMed
Gaebel, W, Riesbeck, M, Wolwer, W, Klimke, A, Eickhoff, M, von Wilmsdorff, M, et al. Maintenance treatment with risperidone or low-dose haloperidol in first-episode schizophrenia: 1-year results of a randomized controlled trial within the German research network on schizophrenia. J Clin Psychiatry. 2007;68:1763–74. doi:10.4088/jcp.v68n1116.CrossRefGoogle ScholarPubMed
Kahn, RS, Fleischhacker, WW, Boter, H, Davidson, M, Vergouwe, Y, Keet, IP, et al. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371:1085–97. doi:10.1016/S0140-6736(08)60486-9.CrossRefGoogle ScholarPubMed
San, L, Arranz, B, Perez, V, Safont, G, Corripio, I, Ramirez, N, et al. One-year, randomized, open trial comparing olanzapine, quetiapine, risperidone and ziprasidone effectiveness in antipsychotic-naive patients with a first-episode psychosis. Psychiatry Res. 2012;200:693701. doi:10.1016/j.psychres.2012.07.005.CrossRefGoogle ScholarPubMed
Sanz-Fuentenebro, J, Taboada, D, Palomo, T, Aragües, M, Ovejero, S, Del Alamo, C, et al. Randomized trial of clozapine vs. risperidone in treatment-naïve first-episode schizophrenia: results after one year. Schizophr Res. 2013;149:156–61. doi:10.1016/j.schres.2013.07.003.CrossRefGoogle ScholarPubMed
Torres, US, Duran, FL, Schaufelberger, MS, Crippa, JA, Louzã, MR, Sallet, PC, et al. Patterns of regional gray matter loss at different stages of schizophrenia: a multisite, cross-sectional VBM study in first-episode and chronic illness. NeuroImage. 2016;12:115. doi:10.1016/j.nicl.2016.06.002.CrossRefGoogle ScholarPubMed
Zhang, C, Wang, Q, Ni, P, Deng, W, Li, Y, Zhao, L, et al. Differential cortical gray matter deficits in adolescent and adult onset first episode treatment naïve patients with schizophrenia. Sci Rep. 2017;7:10267 doi:10.1038/s41598-017-10688-1.CrossRefGoogle ScholarPubMed
Ishibashi, T, Horisawa, T, Tokuda, K, Ishiyama, T, Ogasa, M, Tagashira, R, et al. Pharmacological profile of lurasidone, a novel antipsychotic agent with potent 5-hydroxytryptamine 7 (5-HT7) and 5-HT1A receptor activity. J Pharmacol Exp Ther. 2010;334:171–81. doi:10.1124/jpet.110.167346.CrossRefGoogle ScholarPubMed
Nakamura, M, Ogasa, M, Guarino, J, Phillips, D, Severs, J, Cucchiaro, J, et al. Lurasidone in the treatment of acute schizophrenia: a double-blind, placebo-controlled trial. J Clin Psychiatry. 2009;70:829–36. doi:10.4088/JCP.08m04905.CrossRefGoogle ScholarPubMed
Meltzer, HY, Cucchiaro, J, Silva, R, Ogasa, M, Phillips, D, Xu, J, et al. Lurasidone in the treatment of schizophrenia: a randomized, double-blind, placebo- and olanzapine-controlled study. Am J Psychiatry. 2011;168:957–67. doi:10.1176/appi.ajp.2011.10060907.CrossRefGoogle ScholarPubMed
Citrome, L, Cucchiaro, J, Sarma, K, Phillips, D, Silva, R, Tsuchiya, S, et al. Long-term safety and tolerability of lurasidone in schizophrenia: a 12-month, double-blind, active-controlled study. Int Clin Psychopharmacol. 2012;27:165–76. doi:10.1097/YIC.0b013e32835281ef.CrossRefGoogle ScholarPubMed
Loebel, A, Cucchiaro, J, Sarma, K, Xu, L, Hsu, C, Kalali, AH, et al. Efficacy and safety of lurasidone 80 mg/day and 160 mg/day in the treatment of schizophrenia: a randomized, double-blind, placebo and active-controlled trial. Schizophr Res. 2013;145:101–9. doi:10.1016/j.schres.2013.01.009.CrossRefGoogle ScholarPubMed
Loebel, A, Cucchiaro, J, Xu, J, Sarma, K, Pikalov, A, Kane, JM. Effectiveness of lurasidone vs. quetiapine XR for relapse prevention in schizophrenia: a 12-month, double-blind, noninferiority study. Schizophr Res. 2013;147:95102. doi:10.1016/j.schres.2013.03.013.CrossRefGoogle ScholarPubMed
Nasrallah, HA, Silva, R, Phillips, D, Cucchiaro, J, Hsu, J, Xu, J, et al. Lurasidone for the treatment of acutely psychotic patients with schizophrenia: a 6-week, randomized, placebo-controlled study. J Psychiatr Res. 2013;47:670–7. doi:10.1016/j.jpsychires.2013.01.020.CrossRefGoogle ScholarPubMed
Ogasa, M, Kimura, T, Nakamura, M, Guarino, J. Lurasidone in the treatment of schizophrenia: a 6-week, placebo-controlled study. Psychopharmacology. 2013;225:519–30. doi:10.1007/s00213-012-2838-2.CrossRefGoogle ScholarPubMed
Tandon, R, Cucchiaro, J, Phillips, D, Hernandez, D, Mao, Y, Pikalov, A, et al. A double-blind, placebo-controlled, randomized withdrawal study of lurasidone for the maintenance of efficacy in patients with schizophrenia. J Psychopharmacol. 2016;30:6977. doi:10.1177/0269881115620460.CrossRefGoogle ScholarPubMed
Loebel, A, Citrome, L. Lurasidone: a novel antipsychotic agent for the treatment of schizophrenia and bipolar depression. B J Psych Bull. 2015;39:237–41. doi:10.1192/pb.bp.114.048793.Google ScholarPubMed
Tocco, M, Newcomer, JW, Mao, Y, Pikalov, A. Lurasidone and metabolic syndrome: results from short- and long-term clinical studies in patients with bipolar depression. CNS Spectr. 2020;25:302–3. doi:10.1017/S1092852920001698.CrossRefGoogle Scholar
Kroeze, WK, Hufeisen, SJ, Popadak, BA, Renock, SM, Steinberg, S, Ernsberger, P, et al. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology. 2003;28:519–26. doi:10.1038/sj.npp.1300027.CrossRefGoogle ScholarPubMed
Reynolds, GP, Kirk, SL. Metabolic side effects of antipsychotic drug treatment-pharmacological mechanisms. Pharmacol Ther. 2010;125:169–79. doi:10.1016/j.pharmthera.2009.10.010.CrossRefGoogle ScholarPubMed
Lord, CC, Wyler, SC, Wan, R, Castorena, CM, Ahmed, N, Mathew, D, et al. The atypical antipsychotic olanzapine causes weight gain by targeting serotonin receptor 2C. J Clin Invest. 2017;127:3402–6. doi:10.1172/JCI93362.CrossRefGoogle ScholarPubMed
Goldman, R, Loebel, A, Cucchiaro, J, Deng, L, Findling, RL. Efficacy and safety of lurasidone in adolescents with schizophrenia: a 6-week, randomized placebo-controlled study. J Child Adolesc Psychopharmacol. 2017;27:516–25. doi:10.1089/cap.2016.0189.CrossRefGoogle ScholarPubMed
Findling, RL, Goldman, R, Chiu, YY, Silva, R, Jin, F, Pikalov, A, et al. Pharmacokinetics and tolerability of Lurasidone in children and adolescents with psychiatric disorders. Clin Ther. 2015;37:2788–97. doi:10.1016/j.clinthera.2015.11.001.CrossRefGoogle ScholarPubMed
Correll, CU, Findling, RL, Tocco, M, Pikalov, A, Deng, L, Goldman, R. Safety and effectiveness of lurasidone in adolescents with schizophrenia: results of a 2-year, open-label extension study. CNS Spectr. 2020;20:111. doi:10.1017/S1092852920001893.com/watch?v=RaAG-SwEa7k.Google Scholar
Correll, CU. Safety and tolerability of antipsychotic treatment in young patients with schizophrenia. J Clin Psychiatry. 2011;72:e26. doi:10.4088/JCP.9101tx5c.CrossRefGoogle ScholarPubMed
Kay, SR, Fiszbein, A, Opler, LA. The positive and negative syndrome scale (PANSS) for schizophrenia. Schizophr Bull. 1987;13:261–76. doi:10.1093/schbul/13.2.261.CrossRefGoogle Scholar
Guy, W. Clinical global impressions. In: ECDEU assessment manual for psychopharmacology, Rockville, MD: US Department of Health, Education, and Welfare; 1976, pp. 217222.Google Scholar
Shaffer, D, Gould, MS, Brasic, J, Ambrosini, P, Fisher, P, Bird, H, et al. A children’s global assessment scale (CGAS). Arch Gen Psychiatry. 1983;40:1228–31. doi:10.1001/archpsyc.1983.01790100074010.CrossRefGoogle Scholar
Endicott, J, Nee, J, Yang, R, Wohlberg, C. Pediatric quality of life enjoyment and satisfaction questionnaire (PQ-LES-Q): reliability and validity. J Am Acad Child Adolesc Psychiatry. 2006;45:401–7. doi:10.1097/01.chi.0000198590.38325.81.CrossRefGoogle ScholarPubMed
Lingjaerde, O, Ahlfors, UG, Bech, P, Dencker, SJ, Elgen, K. The UKU side effect rating scale. A new comprehensive rating scale for psychotropic drugs and a cross-sectional study of side effects in neuroleptic-treated patients. Acta Psychiatr Scand Suppl. 1987;334:1100. doi:10.1111/j.1600-0447.1987.tb10566.x.CrossRefGoogle Scholar
Simpson, GM, Angus, JW. A rating scale for extrapyramidal side effects. Acta Psychiatr Scand Suppl. 1970;212:11–9. doi:10.1111/j.1600-0447.1970.tb02066.x.CrossRefGoogle ScholarPubMed
Barnes, TR. A rating scale for drug-induced akathisia. Br J Psychiatry. 1989;154:672–6. doi:10.1192/bjp.154.5.672.CrossRefGoogle ScholarPubMed
Munetz, MR, Benjamin, S. How to examine patients using the abnormal involuntary movement scale. Hosp Community Psychiatry. 1988;39:1172–7. doi:10.1176/ps.39.11.1172.Google ScholarPubMed
Posner, K, Brown, GK, Stanley, B, Brent, DA, Yershova, KV, Oquendo, MA, et al. The Columbia-suicide severity rating scale: Initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168:1266–77. doi:10.1176/appi.ajp.2011.10111704.CrossRefGoogle ScholarPubMed
Pagsberg, AK, Tarp, S, Glintborg, D, Stenstrøm, AD, Fink-Jensen, A, Correll, CU, et al. Acute antipsychotic treatment of children and adolescents with schizophrenia-Spectrum disorders: a systematic review and network meta-analysis. J Am Acad Child Adolesc Psychiatry. 2017;56:191202. doi:10.1016/j.jaac.2016.12.013.CrossRefGoogle ScholarPubMed
Arango, C, Ng-Mak, D, Finn, E, Byrne, A, Loebel, A. Lurasidone compared to other atypical antipsychotic monotherapies for adolescent schizophrenia: a systematic literature review and network meta-analysis. Eur Child Adolesc Psychiatry. 2020;29(9):1195–205. doi:10.1007/s00787-019-01425-2.CrossRefGoogle ScholarPubMed
Costamagna, I, Calisti, F, Cattaneo, A, Hsu, J, Tocco, M, Pikalov, A, et al. Efficacy and safety of lurasidone in adolescents and young adults with schizophrenia: a pooled post hoc analysis of double-blind, placebo-controlled 6-week studies. Eur Psychiatry. 2021;64(1):e35. doi:10.1192/j.eurpsy.2021.30.CrossRefGoogle Scholar
Solmi, M, Fornaro, M, Ostinelli, EG, Zangani, C, Croatto, G, Monaco, F, et al. Safety of 80 antidepressants, antipsychotics, anti-attention-deficit/hyperactivity medications and mood stabilizers in children and adolescents with psychiatric disorders: a large scale systematic meta-review of 78 adverse effects. World Psychiatry. 2020;19:214–32. doi:10.1002/wps.20765.CrossRefGoogle ScholarPubMed
Robinson, DG, Gallego, JA, John, M, Petrides, G, Hassoun, Y, Zhang, JP, et al. A randomized comparison of aripiprazole and risperidone for the acute treatment of first-episode schizophrenia and related disorders: 3-month outcomes. Schizophr Bull. 2015;41:1227–36. doi:10.1093/schbul/sbv125.CrossRefGoogle ScholarPubMed
Harrigan, SM, McGorry, PD, Krstev, H. Does treatment delay in first-episode psychosis really matter? Psychol Med. 2003;33(1):97110. doi:10.1017/s003329170200675x.CrossRefGoogle ScholarPubMed
Souaiby, L, Gaillard, R, Krebs, M-O. Duration of untreated psychosis: a state-of-the-art review and critical analysis. Encéphale. 2016;42(4):361–6. doi:10.1016/j.encep.2015.09.007.CrossRefGoogle ScholarPubMed
Clemmensen, L, Vernal, DL, Steinhausen, HC. A systematic review of the long-term outcome of early onset schizophrenia. BMC Psychiatry. 2012;12:150 doi:10.1186/1471-244X-12-150.CrossRefGoogle ScholarPubMed
Correll, CU, Solmi, M, Veronese, N, Bortolato, B, Rosson, S, Santonastaso, P, et al. Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: a large-scale meta-analysis of 3,211,768 patients and 113,383,368 controls. World Psychiatry. 2017;16:163–80. doi:10.1002/wps.20420.CrossRefGoogle ScholarPubMed
Laursen, TM, Nordentoft, M, Mortensen, PB. Excess early mortality in schizophrenia. Annu Rev Clin Psychol. 2014;10:425–48. doi:10.1146/annurev-clinpsy-032813-153657.CrossRefGoogle Scholar
Solmi, M, Murru, A, Pacchiarotti, I, Undurraga, J, Veronese, N, Fornaro, M, et al. Safety, tolerability, and risks associated with first- and second-generation antipsychotics: a state-of-the-art clinical review. Ther Clin Risk Manag. 2017;13:757–77. doi:10.2147/TCRM.S117321.CrossRefGoogle ScholarPubMed
Vandenberghe, F, Najar-Giroud, A, Holzer, L, Conus, P, Eap, CB, Ambresin, AE. Second-generation antipsychotics in adolescent psychiatric patients: metabolic effects and impact of an early weight change to predict longer term weight gain. J Child Adolesc Psychopharmacol. 2018;28:258–65. doi:10.1089/cap.2017.0038.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Change from double-blind baseline in PANSS total score.

Figure 1

Table 1. Patient characteristics at double-blind baseline.

Figure 2

Table 2. Mean (SE) change from DB baseline on efficacy measures.

Figure 3

Table 3. Adverse events during 6-weeks of double-blind, placebo-controlled treatment.

Figure 4

Table 4. Udvalgfor Kliniske Undersogelser Side Effect Rating Scale Scores:Mean (SD) Baseline-to-EndpointChange.

Figure 5

Table 5. Change from double-blind baseline in laboratory values and weight/BMI.

Submit a response

Comments

No Comments have been published for this article.