Hostname: page-component-7bb8b95d7b-cx56b Total loading time: 0 Render date: 2024-09-29T19:07:18.313Z Has data issue: false hasContentIssue false

Randomised controlled trials of antipsychotics for people with autism spectrum disorder: a systematic review and a meta-analysis

Published online by Cambridge University Press:  04 August 2023

Shoumitro Deb*
Affiliation:
Department of Brain Sciences, Faculty of Medicine, Imperial College London, 2nd Floor Commonwealth Building, Du Cane Road, London W12 0NN, UK
Meera Roy
Affiliation:
Hereford and Worcestershire Health and Care Trust, Kings Court, 2 Charles Hastings Way, WR5 1JR, UK
Bharati Limbu
Affiliation:
Department of Brain Sciences, Faculty of Medicine, Imperial College London, 2nd Floor Commonwealth Building, Du Cane Road, London W12 0NN, UK
Basma Akrout Brizard
Affiliation:
Université de Paris, Laboratory of Psychopathology and Health Processes, F-92100 Boulogne Billancourt, France
Meena Murugan
Affiliation:
Specialty Registrar in Psychiatry of Intellectual Disabilities, Coventry and Warwickshire Partnership NHS Foundation Trust, Brooklands Hospital, Coleshill Road, Birmingham, B37 7HL, UK
Ashok Roy
Affiliation:
Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
Jacopo Santambrogio
Affiliation:
Department of Medicine and Surgery, University Milano-Bicocca, Via della Misericordia 51, Vedano al Lambro (MB), 20854, Italy
*
Corresponding author: Shoumitro Deb; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Background

Despite unclear evidence to support the long-term use of antipsychotics to treat challenging (problem) behaviours in people with autism in the absence of a psychiatric disorder, this practice is common.

Methods

We conducted a systematic review and meta-analysis of all randomised controlled trials (RCTs) involving antipsychotics for people with autism of all ages, irrespective of the outcomes assessed. We searched seven databases and hand-searched ten relevant journals. Two authors independently screened titles, abstracts and full papers and extracted data using the Cochrane Handbook template. We conducted meta-analyses of outcomes and the rate of adverse events.

Results

We included 39 papers based on 21 primary RCTs that recruited 1482 people with autism. No RCT assessed any psychiatric disorder outcome, such as psychoses or bipolar disorder. A meta-analysis of ten placebo-controlled RCTs showed a significantly improved Aberrant Behaviour Checklist-Irritability score in the antipsychotic group with an effect size of −6.45 [95% confidence interval (CI) −8.13 to −4.77] (low certainty). Pooled Clinical Global Impression data on 11 placebo-controlled RCTs showed an overall effect size of 0.84 (95% CI 0.48 to 1.21) (moderate certainty). There was a significantly higher risk of overall adverse effects (p = 0.003) and also weight gain (p < 0.00001), sedation (p < 0.00001) and increased appetite (p = 0.001) in the antipsychotic group.

Conclusions

There is some evidence for risperidone and preliminary evidence for aripiprazole to significantly improve scores on some outcome measures among children with autism but not adults or for any other antipsychotics. There is a definite increased risk of antipsychotic-related different adverse effects.

Type
Original Article
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) that starts in early childhood and often continues into adulthood (American Psychiatric Association, 2013). ASD is characterised by (a) persistent deficits in social communication and social interaction across multiple contexts and (b) restricted, repetitive patterns of behaviour, interests, or activities (American Psychiatric Association, 2013). The reported rate varies between 1 in 160 (American Psychiatric Association, 2013) to 44 (Maenner et al., Reference Maenner, Shaw, Bakian, Bilder, Durkin, Esler and Cogswell2021) children. Comorbidities (overall 55–70%) such as other NDDs like intellectual developmental disabilities (IDD) (38%) and attention deficit hyperactivity disorder (25–28%), and psychiatric disorders such as psychosis (4–12%), anxiety (18–20%), and depression (11–19%) and also problem (challenging) behaviours (10–15%) are more common in ASD compared with the general population who do not have ASD (Deb et al., Reference Deb, Perera, Krysta, Ozer, Bertelli, Novell and Sappok2022). Similarly, the use of psychotropic medications (41.9–61.5%), particularly antipsychotics (11.7%), psychostimulants (12.5%), and antidepressants (3.8%), is widespread in this population, which seems to have increased over time (57% in 1998 v. 64% in 2002; p < 0.05) (Bachmann, Manthey, Kamp-Becker, Glaeske, & Hoffmann, Reference Bachmann, Manthey, Kamp-Becker, Glaeske and Hoffmann2013; Coury et al., Reference Coury, Anagnostou, Manning-Courtney, Reynolds, Cole, McCoy and Perrin2012; Deb, Roy, & Limbu, Reference Deb, Roy and Limbu2022; Jobski, Höfer, Hoffmann, & Bachmann, Reference Jobski, Höfer, Hoffmann and Bachmann2017). Antipsychotics are commonly used for challenging behaviour in people with ASD (Deb et al., Reference Deb, Roy and Limbu2022).

Recent meta-analyses showed no definitive evidence of antidepressants, anti-anxiety medication (Deb et al., Reference Deb, Roy, Lee, Majid, Limbu, Santambrogio and Bertelli2021), and mood stabiliser medications’ efficacy (Limbu et al., Reference Limbu, Deb, Roy, Lee, Roy and Taiwo2022) on the core (such as restrictive and repetitive behaviour, RRB and impaired communication skills) or associated symptoms (such as aggression, irritability and agitation) of ASD. The evidence for the efficacy of antipsychotic medications for people with ASD without a psychiatric diagnosis is unclear (Deb et al., Reference Deb, Roy and Limbu2022; Unwin & Deb, Reference Unwin and Deb2011). The quality of evidence varies, and different methodologies were used to gather evidence in different studies. For example, a recent systematic review and meta-analysis of randomised controlled trials (RCTs) of antipsychotics for people with ASD (D'Alò et al., Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) only included studies on children and combined withdrawal studies with efficacy studies in the same meta-analysis and did not include non-inferiority RCTs. Other studies included RCTs on a small number of new-generation antipsychotics (Fallah et al., Reference Fallah, Shaikh, Neupane, Rusiecki, Bennett and Beyene2019; Linden et al., Reference Linden, Best, Elise, Roberts, Branagan, Tay and Gurusamy2023; Zhou et al., Reference Zhou, Nasir, Farhat, Kook, Artukoglu and Bloch2021) or excluded non-peer-reviewed publications (Fallah et al., Reference Fallah, Shaikh, Neupane, Rusiecki, Bennett and Beyene2019). One meta-analysis included RCTs only involving side effects, and no efficacy data were presented (Alfageh et al., Reference Alfageh, Wang, Mongkhon, Besag, Alhawassi, Brauer and Wong2019). One meta-analysis included youths, some of whom had autism, but no separate data were presented for youths with ASD (Park et al., Reference Park, Cervesi, Galling, Molteni, Walyzada, Ameis and Correll2016).

Therefore, we updated the previous meta-analysis by including RCTs of all antipsychotics involving children, adolescents and adults and non-inferiority head-to-head comparison RCTs. We have described how our systematic review and meta-analysis differ from other recent meta-analyses in this field (Alfageh et al., Reference Alfageh, Wang, Mongkhon, Besag, Alhawassi, Brauer and Wong2019; D'Alò et al., Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021; Fallah et al., Reference Fallah, Shaikh, Neupane, Rusiecki, Bennett and Beyene2019; Linden et al., Reference Linden, Best, Elise, Roberts, Branagan, Tay and Gurusamy2023; Park et al., Reference Park, Cervesi, Galling, Molteni, Walyzada, Ameis and Correll2016; Zhou et al., Reference Zhou, Nasir, Farhat, Kook, Artukoglu and Bloch2021) (online Supplementary Appendix 1).

Methods

We followed PROSPERO guidelines (crd.york.ac.uk/prospero) and the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocol (PRISMA-P) checklist (Moher et al., Reference Moher, Shamseer, Clarke, Ghersi, Liberati, Petticrew and Stewart2015) for this review and search strategy. The study was registered with the PROSPERO database (registration number: CRD42022343669, 4th July 2022). We searched the following databases for English language publications EMBASE, MEDLINE, Cochrane database, clinicaltrials.gov, PsycINFO, ERIC, and CINAHL from their inception till 30th May 2022. We also hand-searched four relevant journals in the field of ASD (Autism, Journal of Autism and Developmental Disorders, Autism Research, Journal of Autism Spectrum Disorder) and six in psychopharmacology (Psychopharmacology, Neuropsychopharmacology, International Journal of Neuropsychopharmacology, Journal of Clinical Psychopharmacology, Human Psychopharmacology, Journal of Child and Adolescent Psychopharmacology) for relevant articles between 2000 and 30th May 2022. Search terms using descriptors for ASD, antipsychotics, outcomes (ASD core symptoms, associated symptoms such as challenging behaviour and psychiatric disorders such as psychoses, schizophrenia and mania) and RCTs (online Supplementary Appendix 2) were developed after a scoping search and based on our previous systematic reviews on psychotropic medications in ASD (Deb et al., Reference Deb, Roy, Lee, Majid, Limbu, Santambrogio and Bertelli2021; Limbu et al., Reference Limbu, Deb, Roy, Lee, Roy and Taiwo2022). Two authors (MR and JS) independently screened titles, abstracts and full papers using the eligibility criteria (online Supplementary Appendix 2). Bibliographies of identified articles were also searched. Grey literature including conference abstracts and unpublished data available on clinicaltrials.gov site were included. Two authors (AR and MM) independently assessed the quality of papers using the Cochrane Risk of Bias scale (Higgins, Savović, Page, Elbers, & Sterne, Reference Higgins, Savović, Page, Elbers, Sterne, Higgins, Thomas, Chandler, Cumpston, Li, Page and Welch2020) (online Supplementary Appendix 2). Two authors (AR and MM) independently extracted data using a data extraction form based on the Cochrane Handbook template (Lefebvre et al., Reference Lefebvre, Glanville, Briscoe, Littlewood, Marshall and Metzendorf2019) (online Supplementary Appendix 2). A third author (SD) arbitrated any disagreement between the authors. Where necessary, the authors of the original articles were contacted for more information.

Eligibility criteria included all RCTs on people with ASD (defined using a standardised method) of all ages involving any antipsychotics, irrespective of the outcome measures (any repeatable measure) used (e.g. psychiatric disorder, challenging behaviour and ASD core symptoms). The control arm included a placebo or another medication or non-pharmacological intervention. RCTs with both matched and unmatched control groups were included. Crossover trials were included only if data were available from Phase I, as it was impossible to exclude any bias caused by the carryover effect on Phase II.

Apart from presenting summary information through a narrative synthesis, where possible, we pooled data for meta-analysis using RevMan 5.4 for Windows software and created forest plots. We did meta-analysis only on the primary RCTs and not on data derived from secondary publications from the main RCTs. We used a random effects odds ratio (OR) for dichotomous or standardised mean difference (SMD) with a 95% confidence interval (CI) for continuous data. We conducted a sensitivity analysis where heterogeneity was high (I2 > 50%). We assessed each meta-analysis's certainty level as either high or moderate or low or very low based on the five domains using the Grading of Recommendations Assessment, Development and Assessment (GRADE) criteria (Guyatt et al., Reference Guyatt, Oxman, Santesso, Helfand, Vist, Kunz and Schünemann2013a, Reference Guyatt, Thorlund, Oxman, Walter, Patrick, Furukawa and Schünemann2013b). We assessed publication bias using a funnel plot and calculated Egger's test (Egger, Smith, Schneider, & Minder, Reference Egger, Smith, Schneider and Minder1997). We used AMSTAR2 scoring (Shea et al., Reference Shea, Reeves, Wells, Thuku, Hamel, Moran and Henry2017) to assess the overall quality of our systematic review.

We contacted the relevant authors for missing data but received no response from most. If the data were still missing, we either excluded those data or, using RevMan 5.4 calculator, converted CI or standard error (s.e.) data to standard deviation (s.d.) as per the Cochrane Handbook formulae (Li, Higgins, & Deeks, Reference Li, Higgins, Deeks, Higgins, Thomas, Chandler, Cumpston, Li, Page and Welch2019). We used the mean endpoint score if data on mean change from baseline were unavailable. For consistency, as per the Cochrane Handbook guideline (Li et al., Reference Li, Higgins, Deeks, Higgins, Thomas, Chandler, Cumpston, Li, Page and Welch2019), we converted pooled dichotomous data (OD) to continuous data (MSD), where most studies presented continuous data.

Results

Study characteristics

Our search identified 2340 citations from seven databases, from which 523 duplicates were removed. From the remaining 1817 citations, 1392 were excluded at the title and 355 and 31, respectively, at the abstract and full paper screening stage (see PRISMA flow chart in online Supplementary Appendix 3). We provided reasons for the exclusion in online Supplementary Appendix 1. We included 39 papers based on 21 primary RCTs and 18 secondary papers originating from these 21 primary RCTs. Altogether 1482 people with ASD were recruited in these primary studies, of whom 1262 completed the study (85.2%). Of the 21 primary RCTs, only one study included adults aged 18 or above (n = 31) and the rest were on children and adolescents (2–17.5 years). Of 1396 participants (one study of n = 86 did not state the gender ratio), 1177 were male (84.3%). Of the 21 primary RCTs, only five were on aripiprazole and one each on olanzapine and lurasidone. The rest were on risperidone (n = 14). All RCTs on aripiprazole, olanzapine and lurasidone were placebo-controlled. All these studies used a pure placebo (i.e. identical sugar pills not vitamins etc.) (Fent, Rosemann, Fässler, Senn, & Huber, Reference Fent, Rosemann, Fässler, Senn and Huber2011). Of the 14 RCTs involving risperidone, seven were placebo-controlled. Two RCTs compared risperidone with aripiprazole and one each haloperidol, divalproex sodium and memantine. One RCT compared the combination of risperidone with parent training and another behavioural intervention in the form of virtual reality respectively with the risperidone-only group. Nine studies included participants with IDD, and the rest did not specify the IQ of the participants. In one of the nine studies, all children had low to moderate IDD (n = 45), but the rest did not present separate data on participants with IDD. The dose of risperidone ranged between 0.125–10 mg/day, but most used 0.25–2.5 mg/day dose. The dose of aripiprazole varied between 5–20 mg/day. The dose of lurasidone was 20–60 mg/day, and olanzapine up to 20 mg/day. We excluded five crossover trials involving haloperidol because no data were available from Phase I.

ASD was diagnosed either clinically or using standardised diagnostic criteria such as the Diagnostic and Statistical Manual (DSM) (American Psychiatric Association, 2013) or International Classification of Diseases (ICD) (World Health Organization, 2019), or standardised diagnostic tools such as Autism Diagnostic Interview-Revised (ADI-R) (Lord, Rutter, & Le Couteur, Reference Lord, Rutter and Le Couteur1994) and Autism Diagnostic Observation Schedule (ADOS) (Lord et al., Reference Lord, Rutter, Goode, Heemsbergen, Jordan, Mawhood and Schopler1989). Most studies assessed the effect of antipsychotics on associated behaviours such as irritability, aggression and agitation using measures like the Aberrant Behavior Checklist-Irritability subscale (ABC-I) (Aman, Burrow, & Wolford, Reference Aman, Burrow and Wolford1995) and Clinical Global Intervention-Improvement (CGI-I) scale (National Institute of Mental Health, 1985) etc. A few studies assessed compulsive behaviour using Children-Yale Brown Obsessive Compulsive Scale (C-YBOCS) (Scahill et al., Reference Scahill, Riddle, McSwiggin-Hardin, Ort, King, Goodman and Leckman1997). No RCT assessed the effect of medications on psychiatric disorders such as psychoses or bipolar disorder.

Narrative synthesis

A narrative synthesis of data is presented in online Supplementary Appendix 1. Most RCTs are on risperidone (n = 14), followed by aripiprazole (n = 5). There is only one large-scale RCT on lurasidone involving 150 children and a very small RCT on olanzapine involving only eight children. There are no published RCTs on other antipsychotics for people with autism apart from the crossover trials on haloperidol that we excluded.

Risperidone

Seven of the 14 RCTs compared risperidone with a placebo, all showing a statistically significant improvement in the risperidone over the placebo group according to the various outcome measures. However, different studies used different outcome measures. Therefore, it is difficult to point out one specific clinical symptom/behaviour that was improved by risperidone. Two RCTs compared risperidone with aripiprazole. Both studies showed post-intervention improvement in the primary outcome measures in both arms, but in one study, risperidone was better than aripiprazole when compared with the placebo, but the other did not show any statistically significant intergroup difference in the outcome. One non-inferiority RCT found a statistically significant better outcome from risperidone than haloperidol. Another non-inferiority RCT found no significant intergroup difference between risperidone and memantine. However, in another small head-to-head comparison, risperidone was found to be significantly better than divalproex sodium. One RCT compared risperidone with a combination of risperidone and parent training, and another a combination with a behavioural intervention using a virtual reality (VR) game. Both studies showed a statistically significant improvement in the combination than the risperidone alone group. Most RCTs were from the USA, and apart from one major RCT, most studies were supported by pharma companies. Two studies were not published in any peer-reviewed journal, but the data were collected from a conference presentation in one case and the Clinical Trial web page in the other.

Aripiprazole

Five RCTs compared the efficacy of aripiprazole with a placebo. Four found significantly better outcomes in the aripiprazole group, but one did not. The two large-scale RCTs were conducted by the pharma company that manufactures aripiprazole. Two of the five RCTs were not published in any peer-reviewed journal, but the data were obtained from the Clinical Trial website.

Lurasidone

Only one RCT has been published on the efficacy of lurasidone. This large-scale (n = 150) multi-centre placebo-controlled study showed no statistically significant intergroup difference in the outcome.

Olanzapine

Only one very small (n = 8) placebo-controlled RCT on the efficacy of olanzapine showed significantly better improvement in the intervention group.

Adverse effects

Most studies reported significant weight gain, increased appetite, and somnolence in the intervention group. Other adverse effects included raised prolactin levels with and without galactorrhoea, drooling, constipation, and extrapyramidal symptoms (online Supplementary Appendix 1).

Meta-analysis of efficacy

It was possible to pool data on ABC-I on ten placebo-controlled RCTs (five on aripiprazole, four on risperidone, and one on lurasidone) (see Fig. 1). The overall pooled data, including all three antipsychotics, showed a significant ABC-I score improvement in the intervention group (effect size: −6.45; 95% CI −1.83 to −4.77; p < 0.00001), with moderate heterogeneity, I2 = 58%, and low certainty. As for individual antipsychotics, lurasidone did not show any statistically significant intergroup difference (effect size: −1.9, 95% CI −5.92 to 2.12; p = 0.35; no heterogeneity, I2 = 0%). Both aripiprazole (effect size = −5.23, 95% CI −6.22 to −4.25; p < 0.00001; no heterogeneity, I2 = 0%) and risperidone (effect size = −8.25, 95% CI −10.93 to −5.56; p < 0.00001; moderate heterogeneity, I2 = 52%) showed a statistically significant improvement in ABC-I score in the intervention group.

Figure 1. ABC-I forest plot.

Initially, we pooled CGI data on 11 placebo-controlled RCTs (four on aripiprazole, five on risperidone, and one each on lurasidone and olanzapine) (see Fig. 2). The overall effect size involving all antipsychotics was 0.84 (95% CI 0.48 to 1.21; p < 0.00001). As for the individual antipsychotics, aripiprazole showed an effect size of 0.83 (95% CI 0.14 to 1.51), risperidone 1.03 (95% CI 0.58 to 1.49), olanzapine 0.99 (95% CI −1.27 to 3.25), and lurasidone 0.31 (95% CI −0.03 to 0.65). However, the heterogeneity was high (I2 = 75%), and the certainty level was moderate as we combined the continuous variable, such as SMD, with dichotomous data, such as OR. After a sensitivity analysis and removing the dichotomous data, we pooled data from six RCTs (three on aripiprazole and one each on risperidone, olanzapine and lurasidone). This lowered the heterogeneity (I2 = 57%). After the sensitivity analysis, the overall effect size involving all antipsychotics was −0.77 (95% CI −1.14 to −0.04; p < 0.00001) (online Supplementary Appendix 3). A forest plot of C-YBOCS data is presented in online Supplementary Appendix 3.

Figure 2. CGI-I forest plot.

Adverse effects

We pooled data on the overall rate of adverse effects for eight RCTs (four for aripiprazole, three for risperidone and one for lurasidone). Overall, the antipsychotic group showed a statistically significant (p = 0.003) increased odd (OR 2.25, 95% CI 1.33–3.83) with moderate heterogeneity (I2: 41%) (moderate certainty) (see Fig. 3). We could pool data on weight gain for 12 RCTs (seven for risperidone, three for aripiprazole and one each for lurasidone and olanzapine). Weight gain was highly significantly worse (p < 0.00001) in the overall antipsychotic than the placebo group (OR 3.9; 95% CI 2.84–5.36), showing no heterogeneity (low certainty) (see Fig. 4). We pooled data on the sedation rate from 12 RCTs (six for risperidone, four for aripiprazole, and one each for lurasidone and olanzapine). Sedation was highly significantly worse (p < 0.00001) in the overall antipsychotic than the placebo group (OR 6.66; 95% CI 3.94–11.26), showing very low heterogeneity (I2: 18%) (moderate certainty) (see Fig. 5). We pooled data on the rate of increased appetite for nine RCTs (six for risperidone, two for aripiprazole and one for olanzapine). Increased appetite was significantly worse (p = 0.001) in the overall antipsychotic than the placebo group (OR 4.15; 95% CI 1.75–9.87), showing moderate heterogeneity (I2: 56) and low certainty (online Supplementary Appendix 3). There was no increased risk for the dropout rates due to any cause but an increased risk for dropouts for adverse effects only (p = 0.003) (low certainty) (online Supplementary Appendix 3).

Figure 3. Overall rate of the adverse events forest plot.

Figure 4. Weight gain adverse effect forest plot.

Figure 5. Sedation adverse effect forest plot.

Quality assessment

Funnel plot

Funnel plots (online Supplementary Appendix 3) did not reveal any publication bias (Egger's test: z = 0.0886, p = 0.9294).

Risk of bias

Six of the 21 (29%) included RCTs showed at least one high risk of bias according to the Cochrane risk of bias scores, but most showed uncertain risks in most assessment areas (online Supplementary Appendix 2).

AMSTAR2

AMSTAR 2 rating showed correct response to all areas (online Supplementary Appendix 2). We have presented a PRISMA-P checklist in online Supplementary Appendix 2.

Discussion

In this systematic review, we included 21 primary RCTs and 18 secondary papers (n = 39 papers) involving 1482 participants with ASD of all ages compared with D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) recent meta-analysis that included 21 RCTs involving overall 1309 children only. We excluded nine of the 21 RCTs in D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) review for the following reasons. Five were on haloperidol which we excluded as they were all crossover trials. Another RCT on risperidone used a crossover design which we excluded as data were unavailable from Phase I. We excluded three more studies as they were placebo-controlled withdrawal RCTs which may bias analysis if mixed with prospective efficacy studies. Instead, we added one RCT on adults with ASD and five head-to-head comparison RCTs with risperidone not included in the D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) review. Additionally, we included two RCTs that combined risperidone and non-pharmacological interventions and another placebo-controlled study involving aripiprazole, not included in D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) study.

In our review, according to the ABC-I and the CGI-I scores, the antipsychotic group showed a statistically significant improvement compared with the placebo. It is difficult to compare our findings directly with D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) as they conducted meta-analyses based on symptoms and behaviours rather than a specific outcome measure. D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) may have combined scores from different outcome measures for the same meta-analysis, which we did not do. This is problematic because the outcome measures used in the RCTs are not directly equivalent to specific symptoms or behaviours.

For example, the most commonly used outcome measure in these studies, ABC-I, although often presented as a measure of irritability and agitation, combines 15 symptoms and behaviour measures. These items include very different symptoms, such as aggression toward others, property and self, irritability, depressed mood, crying, immediate meeting of demands, temper tantrums, labile mood, agitation, shouting and inappropriate screaming etc. Therefore, it is difficult to know which of these symptoms or a combination of symptoms showed improvement when the total score of ABC-I improved. It is unclear which outcome measure or measures D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) used for data analysis on ‘hyperactivity, inattention, oppositeness, disruptive behaviour.’ However, their findings in this meta-analysis are similar to our findings of the analysis of ABC-I data. Similarly, D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) did not state which measures were used in their meta-analysis of the ‘global functioning, global improvement’, but their finding is similar to our findings based on the CGI-I score.

We have not presented the forest plot on the C-YBOCS scale score in the main text for the following reasons. First, it appears that in most studies, this scale was not used to measure obsessive-compulsive symptoms per se but was used as a proxy rather than a direct measure of ASD core symptoms, such as restrictive and repetitive behaviour. Second, C-YBOCS data were available on only a few RCTs and none involving risperidone. Also, autistic people do not like to receive intervention to change their core symptoms, such as repetitive behaviour, unless that improves their mental health or quality of life (Linden et al., Reference Linden, Best, Elise, Roberts, Branagan, Tay and Gurusamy2023).

No RCT has presented any measure for a psychiatric disorder such as psychoses or bipolar disorder, or quality of life. Measuring the quality of life of people with ASD or IDD could be problematic as these are mostly patient-rated, and many autistic people may find communicating their feelings and views difficult or express them differently than non-autistic people (Bertelli et al., Reference Bertelli, Azeem, Underwood, Scattoni, Persico, Ricciardello, Munir, Bertelli, Deb, Munir, Hassiotis and Salvador-Carulla2022). This may affect the validity of these measures. Most quality-of-life measures are not health-related (Unwin & Deb, Reference Unwin and Deb2014). In that respect, CGI-I may be the closest measure to an overall improvement in functioning as a proxy for the quality of life.

Our findings of the increased risk of the overall rate of adverse events and specific medication-related adverse effects, such as increased appetite, weight gain and sedation, contrast D'Alò et al. (Reference D'Alò, De Crescenzo, Amato, Cruciani, Davoli and Fulceri2021) findings but are consistent with findings from another meta-analysis (Fallah et al., Reference Fallah, Shaikh, Neupane, Rusiecki, Bennett and Beyene2019). However, not much data were available to analyse the adverse metabolic effects of the new-generation antipsychotics. Aripiprazole does not raise serum prolactin levels to the same extent as risperidone and can even decrease the prolactin level raised by other medications such as risperidone (Deb et al., Reference Deb, Farmah, Arshad, Deb, Roy and Unwin2014). Therefore, our findings showed a definite increased risk of adverse events associated with antipsychotics when used for people with ASD of any age in the context of a significant improvement in scores according to two measures.

Although the funnel plots did not show any significant publication bias, it is noteworthy that most RCTs were from the USA and were supported by pharma companies. In the case of aripiprazole RCTs, the pharma company that manufactures the medication conducted two main large studies. In other studies, it is difficult to determine any unconscious cognitive bias caused by pharma company support.

Several cofounders may have produced bias in the outcome of the RCTs. For example, most studies did not present separate data on participants with IDD, a common comorbidity of ASD (Bertelli et al., Reference Bertelli, Azeem, Underwood, Scattoni, Persico, Ricciardello, Munir, Bertelli, Deb, Munir, Hassiotis and Salvador-Carulla2022; Deb et al., Reference Deb, Perera, Krysta, Ozer, Bertelli, Novell and Sappok2022). Most RCTs showed a strong placebo effect, and both placebo and medication effects were most pronounced within the first 1–3 weeks of the trial. After that, the effect tended to plateau from 5–7 weeks onward, risking an increase in the dose in real-life practice (McCracken et al., Reference McCracken, McGough, Shah, Cronin, Hong and Aman2002; Owen et al., Reference Owen, Sikich, Marcus, Corey-Lisle, Manos, McQuade and Findling2009; Shea et al., Reference Shea, Turgay, Carroll, Schulz, Orlik, Smith and Dunbar2004). As most studies were add-on trials of antipsychotics, the confounding effect of concomitant medication use is unknown. Similarly, the bias caused by the confounding effect of concomitant non-pharmacological psychosocial and behavioural interventions is unknown. Given that associated behaviours like irritability and agitation are often long-standing, the short follow-up period of around eight weeks may not provide enough time to assess the long-term effects of these medications. Although some post-RCT open-label data are available for long-term efficacy and tolerability (Deb et al., Reference Deb, Roy and Limbu2022), without an RCT design, the long-term effects of placebo could not be assessed. Most studies did not consider the participants' baseline level of challenging behaviour, thus difficult to determine the optimum severity of challenging behaviour for which the medication may be effective. Similarly, a different baseline severity level of challenging behaviour in the intervention and the placebo arm may affect the outcome at follow-up in each arm. Apart from one small study, all other studies are on children thus, the effect of antipsychotics on adults with ASD remains unknown.

Also, it is not easy to configure the optimum dose of medication for treating challenging behaviour on which the outcomes of the included RCTs are primarily based. A low dose may not be effective, whereas a high dose may cause adverse effects. Most studies included a small number of participants risking a Type II error, and a high proportion of studies showed either high or uncertain risk according to the Cochrane risk of bias score. Psychiatric disorders such as psychoses, common in ASD (Bertelli et al., Reference Bertelli, Piva Merli, Bradley, Keller, Varrucciu, Del Furia and Panocchia2015, Reference Bertelli, Azeem, Underwood, Scattoni, Persico, Ricciardello, Munir, Bertelli, Deb, Munir, Hassiotis and Salvador-Carulla2022), may lead to challenging behaviour. Therefore, without assessing the medication's effect on them, which is the case in the included RCTs, it is impossible to ascertain their confounding effect on the outcome of challenging behaviour, which is the common outcome measure used in the included studies.

Apart from risperidone and aripiprazole (and one large study of lurasidone with a negative finding), very little RCT-based evidence is available for other antipsychotics. In the future, RCTs involving antipsychotics should specify specific mental health or behavioural outcomes, include a valid quality-of-life measure, and consider the impact of relevant confounding factors in their design.

Strengths

Our study included the highest number of RCTs involving the highest number of participants among all the published meta-analyses of new-generation antipsychotics among people with ASD of all ages. We followed stringent criteria for the systematic review and meta-analysis, such as PROSPERO and Cochrane guidelines, and also the risk of bias assessment. We conducted funnel plot analysis and Egger's test to assess publication bias. We included papers published in peer-reviewed journals, conference abstracts, and web-based data. The overall quality of our systematic review, assessed by AMSTAR 2 criteria, is very high.

Limitations

We have included only English literature publications. To conduct a meta-analysis, it was only possible to pool data from 71.4–78.6% of 14 placebo-controlled RCTs. The heterogeneity of the meta-analysis was mostly moderate but sometimes high before the sensitivity analysis. According to the GRADE assessment, the certainty levels were low to moderate.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S003329172300212X.

Data

Data availability does not apply to this article as no new data were created or analysed in this study.

Acknowledgements

The Imperial Biomedical Research Centre Facility, funded by the National Institute of Health Research (NIHR), UK, supported the study.

Author contributions

All authors were involved in the conceptualisation and design of the review, contributed to the draft and approved the final version of the paper.

Financial support

There was no specific funding associated with this study.

Competing interest

None.

Ethical standard

As this is a literature review, no ethics approval and consent were required.

References

Alfageh, B. H., Wang, Z., Mongkhon, P., Besag, F. M., Alhawassi, T. M., Brauer, R., & Wong, I. C. K. (2019). Safety and tolerability of antipsychotic medication in individuals with autism spectrum disorder: A systematic review and meta-analysis. Pediatric Drugs, 21, 153167.CrossRefGoogle ScholarPubMed
Aman, M. G., Burrow, W. H., & Wolford, P. L. (1995). The aberrant behaviour checklist community: Factor validity and effect of subject variables for adults in group homes. American Journal of Mental Retardation, 100, 283292.Google ScholarPubMed
American Psychiatric Association (2013). Diagnostic and statistical manual of mental disorders (5th ed.). Washington, DC: American Psychiatric Publishing.Google Scholar
Bachmann, C. J., Manthey, T., Kamp-Becker, I., Glaeske, G., & Hoffmann, F. (2013). Psychopharmacological treatment in children and adolescents with autism spectrum disorders in Germany. Research in Developmental Disabilities, 34(9), 25512563.CrossRefGoogle ScholarPubMed
Bertelli, M. O., Azeem, M. W., Underwood, L., Scattoni, M. L., Persico, A. M., Ricciardello, A., … Munir, K. (2022). Autism spectrum disorder. In Bertelli, M. O., Deb, S., Munir, K., Hassiotis, A., & Salvador-Carulla, L. (Eds.), Textbook of psychiatry for intellectual disability and autism spectrum disorder (pp. 369455). Switzerland, AG: Springer Nature.CrossRefGoogle Scholar
Bertelli, M. O., Piva Merli, M., Bradley, E., Keller, R., Varrucciu, N., Del Furia, C., & Panocchia, N. (2015). The diagnostic boundary between autism spectrum disorder, intellectual developmental disorder and schizophrenia spectrum disorders. Advances Mental Health Intellect Disabilities, 9, 243264.CrossRefGoogle Scholar
Coury, D. L., Anagnostou, E., Manning-Courtney, P., Reynolds, A., Cole, L., McCoy, R., … Perrin, J. M. (2012). Use of psychotropic medication in children and adolescents with autism spectrum disorders. Pediatrics, 130(Suppl.2), S69S76. doi:10.1542/peds.2012-0900D.CrossRefGoogle ScholarPubMed
D'Alò, G. L., De Crescenzo, F., Amato, L., Cruciani, F., Davoli, M., & Fulceri, F., … the ISACA Guideline Working Group (2021). Impact of antipsychotics in children and adolescents with autism spectrum disorder: A systematic review and meta-analysis. Health Quality and Life Outcomes, 19(1), 119.Google ScholarPubMed
Deb, S., Farmah, B. K., Arshad, E., Deb, T., Roy, M., & Unwin, G. L. (2014). The effectiveness of aripiprazole in the management of problem behaviour in people with intellectual disabilities, developmental disabilities and/or autistic spectrum disorder: A systematic review. Research in Developmental Disabilities, 35, 711725. doi:10.1016/j.ridd.2013.12.004.CrossRefGoogle ScholarPubMed
Deb, S., Perera, B., Krysta, K., Ozer, M., Bertelli, M., Novell, R., … Sappok, T. (2022). The European guideline on the assessment and diagnosis of psychiatric disorders in adults with intellectual disabilities. European Journal of Psychiatry, 36, 1125.CrossRefGoogle Scholar
Deb, S., Roy, M., Lee, R., Majid, M., Limbu, B., Santambrogio, J., … Bertelli, M. O. (2021). Randomised controlled trials of antidepressant and anti-anxiety medications for people with autism spectrum disorder: Systematic review and meta-analysis. British Journal of Psychiatry Open, 7, e179, 1–15. doi:10.1192/bjo.2021.1003.CrossRefGoogle ScholarPubMed
Deb, S., Roy, M., & Limbu, B. (2022). Psychopharmacological treatments for psychopathology in people with intellectual disabilities and/or autism spectrum disorder. British Journal of Psychiatry Advances, 112 (FirstView). https://doi.org/10.1192/bja.2022.61.Google Scholar
Egger, M., Smith, G. D., Schneider, M., & Minder, C. (1997). Bias in meta-analysis detected by a simple, graphical test. British Medical Journal, 315, 629634.CrossRefGoogle ScholarPubMed
Fallah, M. S., Shaikh, M. R., Neupane, B., Rusiecki, D., Bennett, T., & Beyene, J. (2019). Atypical antipsychotics for irritability in pediatric autism: A systematic review and network meta-analysis. Journal of Child and Adolescent Psychopharmacology, 29(3), 168180.CrossRefGoogle ScholarPubMed
Fent, R., Rosemann, T., Fässler, M., Senn, O., & Huber, C. A. (2011). The use of pure and impure placebo interventions in primary care a qualitative approach. BMC Family Practice, 12(1), 17.CrossRefGoogle ScholarPubMed
Guyatt, G. H., Oxman, A. D., Santesso, N., Helfand, M., Vist, G., Kunz, R., … Schünemann, H. J. (2013a). GRADE guidelines: 12. Preparing summary of findings tables-binary outcomes. Journal of Clinical Epidemiology, 66(2), 158172.CrossRefGoogle ScholarPubMed
Guyatt, G. H., Thorlund, K., Oxman, A. D., Walter, S. D., Patrick, D., Furukawa, T. A., … Schünemann, H. J. (2013b). GRADE guidelines: 13. Preparing summary of findings tables-continuous outcomes. Journal of Clinical Epidemiology, 66(2), 173183.CrossRefGoogle ScholarPubMed
Higgins, J. P. T., Savović, J., Page, M. J., Elbers, R. G., & Sterne, J. A. C. (2020). Chapter 8: Assessing risk of bias in a randomized trial. In Higgins, J. P. T., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M. J., & Welch, V. A. (Eds.), Cochrane handbook for systematic reviews of interventions version 6.1 (updated September 2020) (pp. 205–228). Chichester (UK): John Wiley & Sons. Available from www.training.cochrane.org/handbook. Accessed 09 March 2023.Google Scholar
Jobski, K., Höfer, J., Hoffmann, F., & Bachmann, C. (2017). Use of psychotropic drugs in patients with autism spectrum disorders: A systematic review. Acta Psychiatrica Scandinavica, 135, 828.CrossRefGoogle ScholarPubMed
Lefebvre, C., Glanville, J., Briscoe, S., Littlewood, A., Marshall, C., & Metzendorf, M-I., … Cochrane Information Retrieval Methods Group (2019). Searching for and selecting studies. Cochrane Handbook for systematic reviews of interventions, 67–107. (www.training.cochrane.org/handbook).CrossRefGoogle Scholar
Li, T., Higgins, J. P. T., & Deeks, J. J. (2019). Collecting data. In Higgins, J. P. T., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M. J., & Welch, V. A. (Eds.), Cochrane handbook for systematic reviews of interventions version 6.0 (updated July 2019) (pp. 109–142). Chichester (UK): John Wiley & Sons. Available from www.training.cochrane.org/handbook. Accessed 03 December 2022.Google Scholar
Limbu, B., Deb, S., Roy, M., Lee, R., Roy, A., & Taiwo, O. (2022). Randomised controlled trials of mood stabilisers for people with autism spectrum disorder: A systematic review and a meta-analysis. British Journal of Psychiatry Open, 8, e52, 1–12. https://doi.org/10.1192/bjo.2022.18.CrossRefGoogle Scholar
Linden, A., Best, L., Elise, F., Roberts, D., Branagan, A., Tay, Y. B. E., … Gurusamy, K. (2023). Benefits and harms of interventions to improve anxiety, depression, and other mental health outcomes for autistic people: A systematic review and network meta-analysis of randomised controlled trials. Autism, 27(1), 730. doi:10.1177/13623613221117931.CrossRefGoogle ScholarPubMed
Lord, C., Rutter, M., Goode, S., Heemsbergen, J., Jordan, H., Mawhood, L., & Schopler, E. (1989). Autism Diagnostic Observation Schedule: A standardized observation of communicative and social behaviour. Journal of Autism and Developmental Disorder, 19(2), 185212.CrossRefGoogle Scholar
Lord, C., Rutter, M., & Le Couteur, A. (1994). Autism diagnostic interview-revised: A revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. Journal of Autism and Developmental Disorder, 24(5), 659685.CrossRefGoogle ScholarPubMed
Maenner, M. J., Shaw, K. A., Bakian, A. V., Bilder, D. A., Durkin, M. S., Esler, A., … Cogswell, M. E. (2021). Prevalence and characteristics of autism spectrum disorder among children aged 8 years-autism and developmental disabilities monitoring network, 11 sites, United States, 2018. Centers for Disease Control and Prevention, Morbidity and Mortality Weekly Report, December 3, 2021, 70(11), 116. doi:10.15585/mmwr.ss7011a1externalicon.CrossRefGoogle Scholar
McCracken, J. T., McGough, J., Shah, B., Cronin, P., Hong, D., & Aman, M. G. Research units on pediatric psychopharmacology autism network (2002). Risperidone in children with autism and serious behavioural problems. New England Journal of Medicine, 347, 314321.CrossRefGoogle Scholar
Moher, D., Shamseer, L., Clarke, M., Ghersi, D., Liberati, A., Petticrew, M., … Stewart, L. A. (2015). Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Systematic Review, 4(1), 1.CrossRefGoogle ScholarPubMed
National Institute of Mental Health. (1985). CGI Clinical global impression scale-NIMH. Psychopharmacology Bulletin, 21, 839844.Google Scholar
Owen, R., Sikich, L., Marcus, R. N., Corey-Lisle, P., Manos, G., McQuade, R. D., … Findling, R. L. (2009). Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics, 124(6), 15331540.CrossRefGoogle ScholarPubMed
Park, S. Y., Cervesi, C., Galling, B., Molteni, S., Walyzada, F., Ameis, S. H., … Correll, C. U. (2016). Antipsychotic use trends in youth with autism spectrum disorder and/or Intellectual Disability: A meta-analysis. Journal of American Academy of Child and Adolescent Psychiatry, 55(6), 456468.CrossRefGoogle ScholarPubMed
Scahill, L., Riddle, M. A., McSwiggin-Hardin, M., Ort, S. I., King, R. A., Goodman, W. K., … Leckman, J. F. (1997). Children's Yale-Brown obsessive compulsive scale: Reliability and validity. Journal of American Academy of Child and Adolescent Psychiatry, 36(6), 844852.CrossRefGoogle ScholarPubMed
Shea, B. J., Reeves, B. C., Wells, G., Thuku, M., Hamel, C., Moran, J., … Henry, D. A. (2017). AMSTAR 2: A critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. British Medical Journal, 358, j4008.CrossRefGoogle ScholarPubMed
Shea, S., Turgay, A., Carroll, A., Schulz, M., Orlik, H., Smith, I., … Dunbar, F. (2004). Risperidone in the treatment of disruptive behavioural symptoms in children with autistic and other pervasive developmental disorders. Pediatrics, 114(5), e634e641. https://doi.org/10.1542/peds.2003-0264-F.CrossRefGoogle ScholarPubMed
Unwin, G., & Deb, S. (2014). Caregiver's Concerns-Quality of Life Scale (CC-QoLS): Development and evaluation of psychometric properties. Research in Developmental Disabilities, 35, 23292340. http://dx.doi.org/10.1016/j.ridd.2014.05.018.CrossRefGoogle ScholarPubMed
Unwin, G. L., & Deb, S. (2011). Efficacy of atypical antipsychotic medication in the management of behaviour problems in children with intellectual disabilities and borderline intelligence: A systematic review. Research in Developmental Disabilities, 32, 21212133.CrossRefGoogle ScholarPubMed
World Health Organization. (2019). International classification of diseases for mortality and morbidity statistics (11th Revision). Retrieved 11 March 2021, from https://icd.who.int/browse11/l-m/en.Google Scholar
Zhou, M. S., Nasir, M., Farhat, L. C., Kook, C. M., Artukoglu, B. B., & Bloch, M. H. (2021). Meta-analysis: Pharmacologic treatment of restricted and repetitive behaviours in autism spectrum disorders. Journal of American Academy of Child and Adolescent Psychiatry, 60(1), 3545.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. ABC-I forest plot.

Figure 1

Figure 2. CGI-I forest plot.

Figure 2

Figure 3. Overall rate of the adverse events forest plot.

Figure 3

Figure 4. Weight gain adverse effect forest plot.

Figure 4

Figure 5. Sedation adverse effect forest plot.

Supplementary material: File

Deb et al. supplementary material 1

Deb et al. supplementary material
Download Deb et al. supplementary material 1(File)
File 117.6 KB
Supplementary material: File

Deb et al. supplementary material 2

Deb et al. supplementary material
Download Deb et al. supplementary material 2(File)
File 624.3 KB
Supplementary material: File

Deb et al. supplementary material 3

Deb et al. supplementary material
Download Deb et al. supplementary material 3(File)
File 4.6 MB