Hostname: page-component-7bb8b95d7b-cx56b Total loading time: 0 Render date: 2024-09-29T21:20:55.916Z Has data issue: false hasContentIssue false

Off-label psychopharmacological interventions for autism spectrum disorders: strategic pathways for clinicians

Published online by Cambridge University Press:  04 August 2023

Nihit Gupta
Affiliation:
Dayton Children’s Hospital, Dayton, OH, USA
Mayank Gupta*
Affiliation:
Southwood Psychiatric Hospital, Pittsburgh, PA, USA
*
Corresponding author: Mayank Gupta; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

The prevalence of autism spectrum disorder (ASD) continues to see a trend upward with a noticeable increase to 1 in 36 children less than 8 years of age in the recent MMWR. There are many factors linked to the substantially increased burden of seeking mental health services, and clinically these individuals are likely to present for impairments associated with co-occurring conditions. The advances in cutting-edge research and the understanding of co-occurring conditions in addition to psychosocial interventions have provided a window of opportunity for psychopharmacological interventions given the limited availability of therapeutics for core symptomatology. The off-label psychopharmacological treatments for these co-occurring conditions are central to clinical practice. However, the scattered evidence remains an impediment for practitioners to systematically utilize these options. The review collates the crucial scientific literature to provide stepwise treatment alternatives for individuals with ASD; with an aim to lead practitioners in making informed and shared decisions. There are many questions about the safety and tolerability of off-label medications; however, it is considered the best practice to utilize the available empirical data in providing psychoeducation for patients, families, and caregivers. The review also covers experimental medications and theoretical underpinnings to enhance further experimental studies. In summary, amidst the growing clinical needs for individuals with ASD and the lack of approved clinical treatments, the review addresses these gaps with a practical guide to appraise the risk and benefits of off-label medications.

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

Introduction

With the increased prevalence in the CDC’s recent MMWR surveys, there are trends of increasing the burden of individuals with ASD in clinical settings.Reference Maenner1 There is an expectation to provide evidence-based interventions to address serious impairments associated with ASD. Mitigating the impairments associated with the symptoms remains a key clinical reason for individuals and families seeking interventions. Given there are no approved psychopharmacological interventions for the core symptoms of ASD, the primary goal of pharmacotherapy in ASD is the treatment of comorbid mental disorders.Reference Goel, Hong, Findling and Ji2 The experts overwhelmingly recommend empirically driven multimodal strategies, including primarily psychosocial treatments, psychopharmacological interventions, parenting sessions, and psychoeducation.Reference Gosling, Cartigny, Mellier, Solanes, Radua and Delorme3

In real-world clinical practice, the accessibility, higher cost, and questionable efficacies of many multimodal interventions remain a challenge, and therefore psychopharmacological treatments with parental psychoeducation on many occasions remain a mainstay of care planning.Reference Subramanyam, Mukherjee, Dave and Chavda4 Among a limited repertoire of therapeutic options, risperidone was the first medication approved by the Food and Drug Administration (FDA) in 2006 for individuals aged 5 and above with irritability associated with ASD. Subsequently, aripiprazole (2009) was approved for the treatment of individuals 6 and over for similar indications.Reference Blankenship, Erickson, Stigler, Posey and McDougle5

About 70% of individuals with ASD also have at least one comorbid psychiatric condition, and 41% also have two or more comorbidities.Reference Simonoff, Pickles, Charman, Chandler, Loucas and Baird6 Taking into account the scope of the problem, there are many psychopharmacologic agents which are used safely and effectively for co-occurring conditions associated with ASD but have not undergone regulatory approval.Reference Bonati, Jacqz-Aigrain and Choonara7 These agents are referred to as “Off-label” for unapproved indications or age groups. A low or insufficient level of evidence is usually the basis for off-label. Off-label use is common and legal, unless it violates ethical guidelines or safety regulations, but carries legal liabilities due to potential health risks.Reference Syed, Dixson, Constantino and Regan8 The dearth of evidence-based psychopharmacological interventions has made off-label use of psychotropic medications common and upholds the right to health (1948 Universal Declaration of Human Rights). About 47% of psychopharmacological agents used in the children and adolescent (CA) population are off-label.Reference Braüner, Johansen, Roesbjerg and Pagsberg9 Therefore, in this review, we appraise the empirical evidence supporting the use of off-label medications for co-occurring symptoms in ASD.

Methods

A comprehensive search of several databases including PubMed, PsychINFO, Cochrane Library, Google Scholar, Scopus, Medline, and web of science from inception to the date of the search was conducted. The search was designed using the following controlled vocabulary and keywords: “Autism*,” “SSRI” and other agents, “Oxytocin,” “Methylphenidate,” “Amphetamines,” “Aripiprazole,” “Risperidone,” “Antidepressants,” “Attention-Deficient Hyperactivity disorder,” “Propranolol,” “Valproic*,” “AcetylCysteine*,” “Amytriptiline” and other TCA’s, “Olanzapine,” Quetiapine,” “Ziprasidone,” “Clozapine,” “Lurasidone,” “Antipsychotic*,” “Lithium,” “Atomoxetine,” “Guanfacine,” “Clonidine,” Alpha-2-agonists,” “Lamotrigine,” “Benzodiazepine,” “Metformin,” “Melatonin,” “Trazodone,” “Mirtazapine,” “Buspirone,” “Amantadine,” “Memantine.” Keywords were selected by creating a list of the most commonly used psychotropic medications in the CA population in clinical practice.Reference Lopez-Leon, Lopez-Gomez, Warner and Ruiter-Lopez10, Reference Madden, Lakoma and Lynch11 The search was performed in all languages. A manual search was also conducted. We included any clinical studies or chart reviews examining the impact of psychopharmacology on the core symptoms of autism or associated comorbidities. Expert opinions and review articles were also included. “We used the following filter for age: Birth to 18 years.” 458 articles were identified after removing duplicates. Study abstracts were reviewed, and 96 of the studies meeting inclusion criteria were selected for this review. We attempted to encapsulate as many trials as possible in the review. We selected only those studies that were mutually agreed upon by the authors. 32 additional studies were added later by manual search. We also updated the material by reviewing reverse citations and added additional studies including numerous newer off-label use.

Results

We have summarized the findings in the following subsections of this narrative review.

Core symptoms

The hallmark of clinical presentations in ASD includes symptoms of restricted, repetitive behaviors and interests (RRBI) and social and communication impairment (SCI). The severity of the impairments is the rationale for multimodal interventions that include focused intervention practices (FIP) commonly used to teach specific skills to children with ASD. A few examples are behavioral strategies, pivotal response therapy, and picture exchange communication systems (PECS cards). A meta-analysis of 43 trials based on FIP found “medium positive effects” with a large effect size, especially with parent participation.Reference Bejarano-Martín, Canal-Bedia and Magán-Maganto12

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed medications for RRBI, but a 2013 Cochrane review of five RCTs concluded that SSRIs do not improve core symptoms, behavioral problems, self-injurious behaviors, or quality of life in individuals with ASD. The review further suggested behavioral activation and agitation due to SSRIs, and possible harm.Reference Williams, Brignell, Randall, Silove and Hazell13 Another meta-analysis found only modest benefits of fluvoxamine and no benefits with citalopram and fluoxetine in RRBI and, instead, found evidence supporting second-generation antipsychotics (SGA) like risperidone and aripiprazole. They concluded that SSRIs are not beneficial and that further studies are needed.Reference Zhou, Nasir, Farhat, Kook, Artukoglu and Bloch14

Among the SGA, risperidone, the most vigorously studied medication, has shown some benefit in reducing RRBI in children with high irritability based on the secondary data analysis of the Research Units on Pediatric Psychopharmacology (RUPP) study,Reference McDougle, Scahill and Aman15 but the overall evidence for RRBI remains weak. Similarly, risperidone has weak evidence for SCI and it is attributed to indirect effects due to improvement in irritability.Reference Canitano and Scandurra16

In our last section “Future directions,” we have discussed preliminary clinical trials on propranolol, divalproex sodium, oxytocin, sulforaphane, insulin growth factor 1, and acetylcysteine. Oxytocin deserves a special mention because initial studies have shown oxytocin to be promising, but recent meta-analysesReference Ooi, Weng, Kossowsky, Gerger and Sung17 have analyzed 12 RCTs on oxytocin and social cognition and on RRBI and found no significant effects of oxytocin on these two core symptoms.

Comorbidities

According to many epidemiologic studies, approximately 70% of individuals with ASD also have at least one comorbid psychiatric condition, and 41% have two or more.Reference Simonoff, Pickles, Charman, Chandler, Loucas and Baird6 Among the most associated psychiatric illnesses with ASD are ADHD, irritability, sleep disturbances, anxiety, and depression. There are many medical comorbidities that are also linked with ASD, which must be assessed and intervened in order to address confounding effects on overall outcomes.

Attention-deficient hyperactivity disorder

The symptoms of hyperactivity, impulsivity, and inattention are observed in 28% of individuals with ASD.Reference Simonoff, Pickles, Charman, Chandler, Loucas and Baird6 Before the changes in the Diagnostic and Statistical Manual of Mental Disorders Fifth Edition (DSM-5), ADHD and ASD (APA, 2013) were not mutually exclusive diagnoses. Therefore, the treatment studies prior to 2013 focused on symptoms of hyperactivity, impulsivity, and inattention which were considered intrinsic to ASD, and measured response to the interventions with proxies.

In the 1990s there were two low-powered studies on the effects of methylphenidate (MPH)Reference Handen, Johnson and Lubetsky18, Reference Quintana, Birmaher and Stedge19 that were inconclusive. MPH in variable doses (7.5 mg to 50 mg) was found to be superior to placebo for hyperactivity symptoms in individuals with ASD; however, there was marked variability in the response and a higher likelihood of adverse effects like a decrease in appetite and insomnia.20 According to a recent Cochrane review, high doses of MPH (0.43 mg/kg/d- 0.60 mg/kg/d) showed significant improvement in hyperactivity. There was a “significant but not clinically relevant” benefit in attention span, but there was no conclusion on impulsivity due to lack of data.Reference Sturman, Deckx and van Driel21 Furthermore, MPH did not affect any of the core symptoms such as social interactions, stereotypical behaviors, or impaired communication. Three out of the four trials on MPHReference Handen, Johnson and Lubetsky18-20, Reference Pearson, Santos and Aman22 were included in the quantitative analysis. Since the three included trials were short-duration trials with small sample sizes, and children intolerant to MPH were excluded during testing, the overall quality of evidence was low and inconclusive.Reference Sturman, Deckx and van Driel21 Another 6-week open-label trial (2017) found long-acting liquid MPH was beneficial for ADHD symptoms in 27 CA with ASD and comorbid ADHD.Reference Kim, Shonka, French, Strickland, Miller and Stein23

A noteworthy trial found that MPH significantly improved performance in tasks requiring sustained attention, selective attention, and inhibition/impulsivity. In general, the relationship between dose and response was linear in the dose range studied, and there were no signs of deterioration at higher MPH dose levels.Reference Pearson, Santos and Aman24 Recent literature suggests that youth with high-functioning ASD respond linearly to MPH, but that might not be the case for low-functioning youth with ASD among whom experts recommend titrating the dose cautiously to avoid undesired effects, such as behavioral activation.Reference Joshi, Wilens, Firmin, Hoskova and Biederman25 There are no published studies on amphetamine (AMP) in individuals with ASD and comorbid ADHD. A systemic review aimed at studying the effect of stimulants on irritability in ASD found that none of the trials on stimulants (MPH or AMP) studied the effect on irritability, and there is no evidence for or against the use of stimulants for irritability in ASD.Reference Ghanizadeh, Molla and Olango26

ATX is an inhibitor of the norepinephrine transporter, preventing the reuptake of norepinephrine, and is approved for use in ADHD in children 6 years and above. Atomoxetine has been studied, primarily for ADHD but secondary analyses of the data were conducted for the core symptoms of ASD. The initial studies on ATX in ASD (Troost et al.Reference Troost, Steenhuis and Tuynman-Qua27; Posey 2006Reference Posey, Wiegand, Wilkerson, Maynard, Stigler and McDougle28) have shown that ATX (1.2–1.4 mg/kg/day) is beneficial for mild to moderate ADHD symptoms. Additionally, there were some benefits to social withdrawal and stereotypical and repetitive speech. However, it is associated with adverse effects like gastrointestinal symptoms, irritability, sleep disturbances (SD), and fatigue. About 42% of participants in a studyReference Troost, Steenhuis and Tuynman-Qua27 completed the trial and the rest dropped out due to side effects. A group of Dutch researchers in an 8-week trial compared ATX (1.2 mg/day) to a placebo in individuals with ASD and ADHD and found that besides the improvement in ADHD symptoms (primary outcome measure),Reference Harfterkamp, van de Loo-Neus and Minderaa29, Reference van der Meer, Harfterkamp and van de Loo-Neus30 there were additional benefits with speech, stereotyped behaviors, and fear of change.Reference Harfterkamp, Buitelaar, Minderaa, van de Loo-Neus, van der Gaag and Hoekstra31 In a 28-week follow-up study, the authors concluded that continued treatment improved ADHD symptoms, while adverse events decreased. Evidence, however, was weak, and higher-powered studies are necessary.Reference Harfterkamp, Buitelaar, Minderaa, van de Loo-Neus, van der Gaag and Hoekstra32 Similarly, another 10-week double-blind multicenter randomized cross-over trial comparing ATX with or without Parental Therapy (PT) and a placebo of 128 CA participants with ASD and ADHD was conducted at three sites. The results suggested that ATX and PT were better than placebo for ADHD symptomsReference Handen, Aman and Arnold33 with effects sustained over a 24-week extension studyReference Smith, Aman and Arnold34 and at 1.5 years follow-up.Reference Arnold, Ober and Aman35

Another medication that is less commonly used in ASD is bupropion which has no data from clinical trials. It is used as an off-label second-line medication for ADHD, although the effect is weaker than that of stimulants.Reference Conners, Casat and Gualtieri36 It is used for depression in individuals thought to have irritability due to bipolar disorder, and this use is through extrapolation of the data in the adult population which suggests that bupropion is least likely to worsen mood.Reference Leverich, Altshuler and Frye37 This clinical decision is not evidence-based in CA,Reference Post, Altshuler and Frye38 and even amongst the adult population, the evidence is weak.

Clonidine and guanfacine both activate the alpha-2 receptors at the prefrontal cortex, preventing the secretion of norepinephrine, which helps with cognitive and emotional functioning. Clonidine binds to all three types of alpha-2 receptors A, B, and C, while guanfacine is specific for alpha-2 receptor type A. The long-acting form of both, clonidine extended-release (CXR) and guanfacine extended-release (GXR), are FDA-approved in the pediatric population both as monotherapy for ADHD and as an adjunct to stimulants. The shorter-acting clonidine and guanfacine are used off-label for insomnia associated with (hyperarousal effects of) stimulants.Reference Johnson and Malow39 A recent systematic review found limited evidence for clonidine’s ability to treat behavioral problems in ASD. They found that most data are from case reports and limited guidance is available. However, it continues to be clinically used, and given the due paucity of pharmacological options for challenging behaviors in ASD, a clonidine trial may be an appropriate and cost-effective option. Daytime sedation related to clonidine is a common adverse effect affecting its overall effectiveness.Reference Banas and Sawchuk40

In a multisite RCT,Reference Scahill, McCracken and King41 extended-release guanfacine (GXR) was compared to a placebo in children with ASD (mean age, 8.5 years) over 8weeks, and was found to be safe and effective in treating hyperactivity in this group of children. GXR was found to reduce oppositional behavior and repetitive behaviors, but not to improve anxiety or sleep, in an analysis of secondary outcomes.Reference Politte, Scahill, Figueroa, McCracken, King and McDougle42

Interestingly, SGA may also be effective in treating ADHD in ASD, and one open-label pilot RCT showed that both aripiprazole and risperidone showed improvement in ADHD symptoms after 24 weeks of treatment.Reference Lamberti, Siracusano and Italiano43

Irritability, aggression, self‐injurious behavior, and disruptive behaviors

Antipsychotics are the most prescribed medications for individuals with ASD irritability, aggression, and disruptive behaviors. Second-generation antipsychotics, which offer additional serotonin receptor blockade, are generally preferred in the CA population over typical antipsychotics due to less likelihood of extrapyramidal side effects. Risperidone and aripiprazole are FDA-approved for irritability associated with ASD.Reference Blankenship, Erickson, Stigler, Posey and McDougle5

Risperidone was FDA-approved in 2006 after two positive double-blinded, placebo-controlled, randomized controlled trials (DB-PC-RCTs) of 8 weeks in CAP with ASD.Reference McCracken, McGough and Shah44, Reference Shea, Turgay and Carroll45 Risperidone at a mean dose of 1.8 mg/ day was found to be safe and effective for irritability.Reference McDougle, Scahill and Aman15 The safety and efficacy were maintained for up to 6 months.Reference Aman, Arnold and McDougle46 Similarly, aripiprazole’s safety and efficacy for irritability associated with ASD were established by two DB-PC-RCTs for a duration of 8 weeksReference Marcus, Owen and Kamen47, Reference Owen, Sikich and Marcus48 which guided FDA approval in 2009.

In many cases of risperidone-induced hyperprolactinemia and galactorrhea, a small dose of aripiprazole in addition to risperidone is highly effective.Reference Chen, Su and Bian49

One study suggests 59.8% of antipsychotics used are off-label in CAP with ASD.Reference Chen, Barner and Cho50 Much of this use is through extrapolation of research in pediatric bipolar disorder where lurasidone and asenapine were recently approved in addition to risperidone and aripiprazole. However, other SGAs, such as olanzapine, quetiapine, asenapine, ziprasidone, and clozapine, have not been evaluated by adequately powered RCTs. Small trials with lurasidone were negative while there are no trials on asenapine.

Lurasidone has a chemical structure similar to aripiprazole but did not demonstrate statistically significant efficacy on the primary outcome measures compared to placebo. However, overall Clinical Global Impressions, (CGI–I) scores improved when fixed doses of 20 and 60 mg/day of Lurasidone were used in this study. Loebel et al.Reference Loebel, Brams and Goldman51 conducted a multicenter RCT with 150 participants and found that once-daily, fixed doses of 20 and 60 mg/day of lurasidone were not effective for treating short-term moderate-to-severe irritability associated with ASD. There are reports of response at higher doses which may be considered as an alternative when approved medications are not effective.Reference Gupta and Hoover52 In an 8-week small open-label trial of 11 adolescents with ASD-associated aggression, quetiapine was found to be effective and well tolerated with improvement in aggression and SD.Reference Golubchik, Sever and Weizman53 A 6-week head-to-head trial of olanzapine and haloperidol found that both olanzapine (mean dose 7.9 mg/day) and haloperidol (mean dose 1.4 mg/day) provided a statistically significant clinical improvement on the clinical global impression scale (CGI-S) and children’s psychiatry rating scale (CPRS) (p = 0.0008).Reference Malone, Cater, Sheikh, Choudhury and Delaney54 6 out of 8 participants in another small open-label trial on olanzapine responded to a mean dose of 7.8 mg, improving hyperactivity, social relatedness, affectual reactions, sensory responses, and language. There were no changes in repetitive behaviors, however, and sedation and weight gain were the most common side effects.Reference Potenza, Holmes, Kanes and McDougle55 In another 8-week DB-PC-RCT of 11 participants, 50% responded to olanzapine compared to 20% placebo, with weight gain (7.6 +/− 4.8 lbs. vs 1.5 +/− 1.5 lbs. on placebo) as the common side effect.Reference Hollander, Wasserman and Swanson56 In another open-label trial, 13 weeks of olanzapine treatment led to improved CGI-S scores in 30% of participants. There was an additional significant reduction in aberrant behavior checklist (ABC) sub-scores of irritability, lethargy, stereotypical behaviors, hyperactivity, and inappropriate speech.Reference Fido and Al-Saad57 There has been a lack of research on clozapine due to its potentially serious side-effect profile, and monitoring requirements and evidence are limited to case reportsReference Lambrey, Falissard and Martin-Barrero58 and retrospective chart reviews,Reference Rothärmel, Szymoniak and Pollet59 which are not generalizable. An interesting secondary analysis of an 8-week open-label trial showed no difference in response rates or tolerance between risperidone, aripiprazole, ziprasidone, olanzapine, and quetiapine in those with bipolar disorder and ASD comorbidity.Reference Joshi, Biederman and Wozniak60

Lithium and sodium valproate are effective mood stabilizers in treating persistent aggression in children, but data about ASD are scarce. While they are commonly prescribed to individuals with ASD and persistent aggression, data indicates that they may not be as effective as SGA at treating irritability.Reference Stigler and McDougle61 It may be helpful to use divalproex sodium when there are seizures and abnormal EEGs associated with ASD,Reference Hollander, Dolgoff-Kaspar, Cartwright, Rawitt and Novotny62 but it should be used with caution due to adverse effects. In a 12-week double-blind placebo-controlled RCT of 55 individuals with a mean age of 9.63 years and mean non-verbal-IQ of 63.3 points, 62% showed improvement in the ABC-I scale with depakote, compared with 9% with placebo, with higher blood valproic acid levels showing a better response. Even though the results were optimistic, the study was small, and more research is needed.Reference Hollander, Chaplin and Soorya63 In another study, the author found that divalproex sodium reduced repetitive behavior.Reference Hollander, Soorya, Wasserman, Esposito, Chaplin and Anagnostou64 However, in a DB-PC-RCT involving 30 participants with a pervasive developmental disorder (PDD) and aggression, there was no difference between valproic acid and placebo. This was thought to be due to heterogeneity, small sample size, and large placebo response.Reference Hellings, Weckbaugh and Nickel65

Mitochondrial diseases are more common in ASDReference Legido, Jethva and Goldenthal66 and divalproex sodium should be avoided in any individual with mitochondrial dysfunction due to the possibility of fulminant liver failureReference Krähenbühl, Brandner, Kleinle, Liechti and Straumann67 In addition, divalproex sodium requires monitoring and has a side-effect profile that may cause long-term harm. Likewise, Rezaei et al.Reference Rezaei, Mohammadi and Ghanizadeh68 found that risperidone plus topiramate was superior to risperidone alone. It is important to carefully evaluate the risks, benefits, and side-effect profiles before combining mood stabilizers.

Lithium may also be used for ASD, but the evidence is limited. In a retrospective chart review, 73.7% (n = 14) of patients with ASD and maladaptive behaviors showed improvement (CGI-I ratings of 3) when lithium was added to their treatment regimens.Reference Mintz and Hollenberg69 The effects of lithium on individuals with ASD and Phelan-McDermid Syndrome (SHANK3 Haploinsufficiency) will be investigated in a future pilot study.70 Monitoring lithium levels and lithium’s side-effects profile is also of concern, but at lower dosages, it is quite safe for individuals with intellectual disabilities.Reference Yuan, Song and Zhu71 A studyReference Belsito, Law, Kirk, Landa and Zimmerman72 comparing placebo with lamotrigine for 16 weeks on 27 children (mean age 5.8 years) with ASD found no benefits.

Gabapentin is another commonly prescribed medication for ASD but lacks evidence. The only evidence available includes 1 case report for disruptive and compulsive behaviorsReference Guglielmo, Ioime, Grandinetti and Janiri73 and 1 retrospective chart review for refractory insomnia in 23 CA with various neurodevelopmental disorders, 9 of whom had ASD. They found that 8 out of the 9 participants reported improvement in insomnia and was tolerated well.Reference Robinson and Malow74

Sleep disorders

ASD is associated with issues with sleep onset, sleep maintenance, and irregular sleep–wake cycles. Many causes have been discovered, including genetic abnormalities that disrupt the melatonin pathway. A meta-analysis found that, besides being very well tolerated, was beneficial for improving sleep characteristics, improving daytime behavior, and producing minimal side effects at doses ranging from 1 to 10 mg. The meta-analysis expressed concern about the small sample sizes in five of the included studies.Reference Rossignol and Frye75 Additional studies indicate that children sleep longer and faster with good tolerability on melatonin.Reference Gringras, Gamble and Jones76 Children’s PedPRM, prolonged-release melatonin, was also effective.Reference Gringras, Nir, Breddy, Frydman-Marom and Findling77 Long-term data on prolonged-release melatonin showed promise as well.Reference Maras, Schroder and Malow78 PedPRM was found to be safe and effective for children with ASD after a 3-month double-blind, placebo-controlled study. This study aimed to determine whether long-term use of PedPRM would affect sleep, growth, body mass index, and pubertal development. For the long-term treatment of CA with ASD and insomnia, PedPRM at a dose of 2, 5, or 10 mg was found to be safe and effective. In addition to the lack of adverse effects on growth and pubertal development, there was no withdrawal or safety concern related to the use or discontinuation of the drug.Reference Malow, Findling and Schroder79

The effectiveness of trazodone, a sedating antidepressant, in treating pediatric sleep disorders is lacking in both typically developing children and those with neurodevelopmental disorders; however, the drug is commonly used in clinical practice to treat insomnia. A retrospective review reported on trazodone’s use for the treatment of sleep problems associated with the opsoclonus-myoclonus syndrome, although no studies have specifically examined children with ASD or other neurodevelopmental disorders (NDDs).Reference Pranzatelli, Tate, Dukart, Flint, Hoffman and Oksa80 There is the potential for priapism with trazodoneReference Kem, Posey and McDougle81 and behavioral activation, given the similarities between trazodone and SSRIs.

Antidepressants may be beneficial for children who are anxious or depressed, but they should be used with caution as discussed above. There is also some evidence supporting mirtazapine in insomnia associated with ASD and it is discussed in the anxiety and depression section below. There is weak evidence that risperidone may help insomnia in ASD,Reference Zuddas, Di Martino, Muglia and Cianchetti82 especially in individuals with extreme irritability. However, the Canadian Academy of Child and Adolescent Psychiatry recommends against using SGA as a first-line medication for insomnia.83

Anxiety and depression disorders

Polymorphism of serotonin transporter genes like SLC6A4 has been linked to ASD.Reference Sutcliffe, Delahanty and Prasad84 Also, depressive and anxiety disorders are common in ASD. A Stockholm cohort study found that 19.8% of individuals with ASD without intellectual impairment experienced depression by age 27 compared to 6% in the general population,Reference Rai, Heuvelman and Dalman85 and 20.1% had anxiety compared to 8.7% in the controls.Reference Nimmo-Smith, Heuvelman and Dalman86 In addition to depression and anxiety, they are often prescribed for RRBI which is a core symptom of ASD and can be distressing. OCD is another frequent comorbidity with ASD and is associated with worse outcomes in individuals with ASD compared to individuals without ASD.Reference Jassi, Vidal-Ribas, Krebs, Mataix-Cols and Monzani87 OCD has a similar developmental pattern to RRBI, but it can be differentiated based on the characteristics of anxiety; obsessions may worsen anxiety while sameness and restricted communication may reduce anxiety symptoms. In a 2019 multicenter RCT among 146 individuals with ASD and obsessive-compulsive behaviors, fluoxetine (20–30 mg/day) was compared with a placebo over 16 weeks. They found a significant change in children’s Yale-Brown obsessive-compulsive scale modified for pervasive developmental disorders (CYBOCS-PDD) with fluoxetine, but it had a high dropout rate. Additionally, the study suggested the potential of confounders in the baseline measurement. They concluded that the efficacy remains uncertain.Reference Reddihough, Marraffa and Mouti88 Sample heterogeneity has been an issue in most clinical trials with SSRIs in ASD. Additionally, fewer trials are available on escitalopram, paroxetine, and sertraline, which may be more effective, and therefore there is a need for large RCTs.Reference Reiersen and Handen89

One naturalistic open-label trial of mirtazapineReference Posey, Guenin, Kohn, Swiezy and McDougle90 showed modest improvement to a mean dose of 30.3 mg. Of 26 participants, only 9 showed improvements in aggression, hyperactivity, and insomnia. The results of a DB-PC-RCT conducted over 10 weeks with 30 participants with ASD aged 5–17 found mixed results with mirtazapine for anxiety in ASD. An improvement in Pediatric Anxiety Rating Scale (PARS) was observed within the group (p = 0.001), but not when compared with the placebo group (p = 0.64).Reference McDougle, Thom and Ravichandran91

Buspirone is a partial agonist of pre-and post-synaptic 5HT1A receptors which was originally developed as an antipsychotic but was found ineffective. In subsequent studies, it was found to be mostly serotonergic with antianxiety effects due to presynaptic 5HT1A receptor agonism with some dopamine affinity. Although no dopamine affinity was found, some early research on buspirone in ASD was initially focused on hyperactivity.Reference Realmuto, August and Garfinkel92, Reference McCormick93 In 1986, buspirone got FDA approval for generalized anxiety disorder in adults, but its use in CA is still off-label. Several studies have examined the effectiveness of buspirone in treating anxiety and irritability among individuals with ASD.Reference Ceranoglu, Wozniak and Fried94-Reference Ghanizadeh and Ayoobzadehshirazi96 Low-dose buspirone may prove to be a safe alternative, especially in younger children, due to its favorable safety and side-effect profiles.Reference Chugani, Chugani and Wiznitzer95 Since SSRIs are at risk for behavioral activation and akathisia; buspirone may be a suitable alternative. No clinical trials have been conducted on hydroxyzine in individuals with ASD. Benzodiazepines should not be used to treat irritability and aggression in ASD with a risk for behavioral disinhibition, which could worsen aggression.Reference Howes, Rogdaki and Findon97 Benzodiazepines may, however, be necessary to manage acute aggression.

Avoidant/restrictive food intake disorder and other feeding difficulties

ARFID is another condition that frequently co-occurs with ASD. One study estimated a prevalence of ARFID in up to 21% of individuals with ASD.Reference Koomar, Thomas, Pottschmidt, Lutter and Michaelson98 ARFID shares many symptoms with ASD including feeding problems and sensory aversions.Reference Bourne, Mandy and Bryant-Waugh99 One meta-analysis estimates individuals with ASD are five times more likely to have feeding difficulties compared to TDY.Reference Sharp, Berry and McCracken100 The treatment of ARFID is limited to modified behavioral therapies, and there is no medication that has evidence to support its benefits.Reference Burton, Allan and Eckhardt101 Some case reports have suggested the benefits of mirtazapine but need further empirical supportReference Brigham, Manzo, Eddy and Thomas102 (Table 1).

Table 1. Key Studies Included in the Review

Discussion

ASD remains a highly heterogeneous condition, with a myriad range of genetic variations and developmental phenotypes. With a higher-than-ever prevalence of 1:36 (under the age of 8) in the United States, it is imperative to screen given in a proportion of individuals with ASD the signs and symptoms are often overshadowed by comorbidities. Late diagnostics are not uncommon and are often previously treated for co-occurring conditions without the awareness of underlying ASD and modifications wherein. Therefore, given its high comorbidity with both psychiatric and medical disorders, multiple interacting variables often challenge clinical diagnostics, and it often takes a deep understanding of the developmental aspects of ASD in deciding what, how, and when to treat (Figure 1).

Figure 1. A flowchart of a complex clinical pathway for diagnostics of ASD and assessment of co-occurring conditions. (a) Autism is a clinical diagnosis that incorporates levels of dysfunction and support needed in various contexts. (b) Testing is an aid to clinical diagnosis but is not necessary. ADOS-2 is an excellent test but is far from being the gold standard. (c) Testing should incorporate various elements including intellectual functioning, adaptive functioning, ADOS-2, ADI-R, and others. (d) ADOS-2 alone may only provide a 60-minute snapshot of symptoms and supplemental detailed parent interviews like ADI-R are helpful in improving sensitivity and specificity. (e) Given the high rates of false negatives, negative testing should be interpreted based on context and dysfunction. (f) In case of negative testing, consider diagnostic overshadowing or “Multiple Complex Developmental Disorder” which could lead to masking of symptoms, especially in HFA individuals. (g) If testing continued to be negative but clinical symptoms are suggestive of autism, consider diagnosing ASD. (h) Aggression in ASD can be complex and multimodal interventions have better outcomes. Interventions include considerations of sensory and food insensitivities along with ruling out any medical causes like constipation. (i) Although a linear dose–response was reported with MPH for higher-functioning ASD youths; however, experts suggest using caution with dose titration in lower-functioning ASD youths to avoid undesired side effects including behavioral activation. (j) Slow-dose titration of SSRI is recommended due to the risk of behavioral activation. There is a high risk of partial response and no response due to poor metabolism and precision medicine is recommended. (k) Could be secondary to sensory insensitivities or medical reasons.

Guiding principles of treatment with off-label medications

Due to these multilayered complexities, the diagnosis and treatment of ASD and its comorbidities is often a clinical conundrum requiring astute medical decision-making. It is not uncommon, and many symptoms associated with ASD are transdiagnostic and do not meet the threshold for a DSM diagnosis; however, the severity of symptoms (associated with impairments) continues to necessitate treatment despite a lack of evidence-based psychopharmacological interventions. Therefore, with marked variability in the clinical phenotypes, interacting risks, perpetuating factors, and limited resources, the onus is on synthesizing and sequencing the best available therapeutics with shared decision-making.

The off-label use of psychotropics may have better outcomes when clinicians are aware of the following guiding principles that apply to CA and more specifically to ones with ASD. Given the challenge of diagnostic uncertainty, it may be prudent to target symptoms since it takes a developmental course for the illness to develop. The knowledge of the quality, efficacy, safety, and intended use of therapeutics for unlicensed applications (“off-label” use) is critical prior to interventions. Always start low and go slow but may need a higher dose based on the pharmacokinetic profiles; also, multiple medications may be needed which requires an adequate duration of trial with optimal dosing before switching. While switching it is advised to change one drug at a time to gather information about the responses and adverse effects.

The incorporation of screening tools and standardized diagnostic tools under measurement-based care (MBC) is a recommended step to discriminate and identify co-occurring conditions. The clinical outcomes must be measured in two settings, with one preferably structured like a school, that could guide the need for titration or change. The knowledge of black box warnings, pharmacogenetics profiles, pharmacokinetics, gender difference, and so forth further advances the likelihood of response under the core strategies of precision psychiatry. Therefore, integrating psychiatric decision-making with MBC, and precision medicine approaches are the cornerstone for sequencing the psychiatric management of these highly complex clinical presentations. Although some advanced precision medicine tests might not be cost-effective or widely available, these could be considered for non-responders or partial responders. Lastly, psychoeducation about these off-label medications should be provided with NbN-2-based classification to effectively convey underlying mechanisms and rationale.Reference Sultan, Correll, Zohar, Zalsman and Veenstra-VanderWeele104

Synthesizing and sequencing treatments for multiple comorbidities

With the widely spread and scattered but rich literature on off-label therapeutics for ASD, there is a need to develop processes in how and when to target specific symptoms, diagnoses, and impairments. There are many moving targets, confounders, and caveats which we discuss in the subsequent sections.

Firstly, one of the most common presentations requiring interventions in individuals with ASD is emotional dysregulation. The term emotional dysregulation (ED) is a broad construct defined as a failure to regulate emotions appropriately with an excessive response to stimuli. The transdiagnostic symptoms of irritability (which is a proneness to anger) are a feature of several different DSM diagnoses and are difficult to discriminate from other mental disorders. It is also recommended to use broad, nonspecific, validated tools like the overt aggression scale (OAS), and the strengths and difficulties questionnaire (SDQ) to assess these symptomatologys. There are also more specific scales for ED in ASD like the affective reactivity index (ARI) and the aberrant behavior checklist (ABC). The applications of these measurement tools could assist in differentiating between persistent non-episodic irritability and frank mania (which tends to present as distinct episodes). It has been widely accepted that with the advances in MBC, it is useful to develop strategies using objective scales validated for symptoms and disorders in question. In the last decade, narrow scales have been developed, that have a propensity to detect salient features to discriminate when overlapping symptoms are present. When a decision is made to target impairing symptoms, multimodal interventions must be part of the treatment plan to maximize efficacies. While ABA and other evidence-based interventions are not widely accessible in all geographical areas, educating patients and families about the limitations of off-label interventions is necessary.

In individuals with ASD with chronic persistent irritability, the highest effect size is for risperidone and aripiprazole which are also considered the first line of treatment. Due to the difference in the half-lives, the response with risperidone is earlier as compared to aripiprazole, but they have similar effect sizes and must be considered as first-line interventions. The agents with shorter half-lives reach a steady state earlier, whereas ones with longer half-lives may take longer to achieve a response based on their pharmacokinetics profiles. Many studies have also shown that variable dose schedules have better outcomes as compared to the fixed-dose. This underscores the need for both optimal drug titration to a therapeutic dose and waiting for an adequate duration for a clinical response prior to the switch or if the agent is deemed ineffective.

In individuals with ASD with comorbid ADHD symptoms, chronic persistent non-episodic irritability could be a challenging symptom to treat. Unlike typically developing children, treatment of ADHD symptoms only after the titration of stimulant medication with or without alpha agonists and psychosocial interventions was not found to be statistically significant. In these individuals (with ASD with ADHD) who do not respond to conventional regimes (stimulants with or without alpha agonists), the addition of risperidone or divalproex sodium to the stimulants has been empirically supported.

About 50% of individuals with ADHD also have irritability or emotional dysregulation, and it is the most commonly co-occurring condition associated with ASD. It is highly challenging to differentiate the emotional dysregulation of ADHD from the irritability associated with ASD. Therefore, the decision to treat ED due to ADHD symptoms or irritability associated with ASD remains critical for overall outcomes. At the same time, the knowledge of the presence and absence of atypical sensory problems (ASP), co-occurring anxiety, tics, sleep disturbances, cognitive impairments, seizures, gastrointestinal problems, and RRBI is critical to incorporate into the choice of interventions.

The late diagnosis of ASD is common with many presenting with new onset problems after being stable for years on previous treatments for ADHD (with stimulants with or without alpha agonists). Interestingly studies of risperidone and aripiprazole targeting irritability measured on the subscale of the ABC also found improvement in the “hyperactivity/defiance and stereotyped behaviors” subscales in individuals with ASD.Reference McPheeters, Warren and Sathe105 In contrast, most of the studies on MPH in individuals with ASD failed to distinguish between hyperactivity and irritability.Reference Joshi, Wilens, Firmin, Hoskova and Biederman25 This compelling evidence provides an overarching guide to avoiding a priori assumptions. Therefore, this specific group of individuals who were chronologically diagnosed late with ASD (as compared to ADHD) needs further assessment if they require titration of ADHD medication or switch to a different class of medications.

In disorders when higher comorbidities like ASD, it is a widely accepted general principle to treat an established clinical diagnostic than nonspecific irritability or aggression. However, in the case of the treatment of ADHD in individuals with ASD, there is a caveat in dosing stimulants for specific developmental phenotypes. Emerging literature has reported a linear dose–response with MPH for higher-functioning ASD youths;Reference Sturman, Deckx and van Driel21 however, experts suggest using caution with dose titration in lower-functioning ASD youths to avoid undesired side effects, including behavioral activation. The knowledge of curve linear dose–response with MPH in lower-functioning ASD children is crucial to avoid adverse effects like iatrogenic insomnia with higher doses and therefore slow, gradual titration of MPH remains key for optimizing response in these subgroups.Reference Joshi, Wilens, Firmin, Hoskova and Biederman25 Treatment-emergent mood lability (ie, mood dysregulation and mood-related adverse event) are also associated with MPH in individuals with ASD, and caution is needed during slow titration in lower-functioning ASD individuals. When only a partial response is observed with stimulants on ADHD symptoms, in the absence of any verbal or physical outburst or aggression the addition of alpha agonists like guanfacineReference Wilens, Bukstein and Brams106 and clonidineReference Kollins, Jain and Brams107 is recommended.

In a highly significant 2014 study “Treatment of Severe Childhood Aggression” (The TOSCA Study), there were additional benefits of adding risperidone to individuals with ADHD optimized on stimulants and parent training.Reference Aman, Bukstein and Gadow108 Although ADHD medications like MPH may partially reduce irritability in some individuals with ASD and co-occurring ADHD, on many occasions, dose-optimized ADHD treatments may not be sufficient. Therefore, at that point, assessment for combination treatment with additional psychotropics like risperidone or aripiprazole may be considered, given these agents have established efficacy and comparable effect sizes.

Uncovering confounders with emerging evidence and neuroscientific context of SSRIs controversies

Counterintuitively psychosis is also common (0–60%) in individuals with ASD, but unlike schizophrenia, it presents at an earlier age and has atypical presentations like behavioral disorganization, often suggestive of psychosis in individuals with ID and ASD.Reference De Giorgi, De Crescenzo and D’Alò109, Reference Bakken, Friis, Lovoll, Smeby and Martinsen110 About one in five individuals with subclinical psychotic symptoms were found at risk of conversion to psychosis.Reference Foss-Feig, Velthorst and Smith111

SSRIs have commonly prescribed medications in CAP, although there have been many controversies related to black box warnings. Low-dose SSRI has been better tolerated; however, the risk and benefits rationale is yet to be empirically substantiated. In mouse models, the disruption of the excitatory/inhibitory (E/I) balance of cortical circuits is thought to underlie the pathophysiology of ASDReference Saitow, Takumi and Suzuki112 and the imbalance of the neuronal circuity in the prefrontal cortex (PFC). To understand the relationship between the serotonin transporter gene (SLC6A4) and the serotonin receptor 2A gene (HTR2A), the serotonin-transporter-linked polymorphic region (5HTTLPR)Reference Sugie, Sugie and Fukuda113 of SLC6A4 and SSRI response in ASD has been the area of interest.Reference Najjar, Owley and Mosconi114 Studies comparing allele 5-HTTLPR (long (l) or short(s)) and response to SSRIs found the LL and SL groups showed a greater reduction in irritability scores than did the SS group.Reference Owley, Brune and Salt115

Serotonin systems have been dysregulated in ASD individuals and a study found a statistically significant decrease in serotonin transporters (5-HTT) within the anterior cingulate cortex (ACC). A decrease in 5-HT2 receptor density in the ACC in the adult cohort was found but not in child postmortem ASD cases as compared to controls. This suggests a deficit in 5-HTT within the ACC in individuals with ASD, while reductions in 5-HT2 density are age-dependent.

These findings also correlate with the behavioral activation associated with SSRI, observed in almost a third of the individuals with ASD.Reference King116 Younger children are more affected as compared to adolescents;Reference Luft, Lamy, DelBello, McNamara and Strawn117 likewise, poor-metabolizer with regard to P450-2D6, serotonin-transporter-linked polymorphic region (5HTTLPR)Reference Sugie, Sugie and Fukuda113 variability, and increase in the plasma concentration due to rapid medication titration are also linked to the activation.Reference Luft, Lamy, DelBello, McNamara and Strawn117

Another study reviewed messenger RNA expression in the HTR and found that the expression of excitatory glutamate serotonin 2A receptors HTR2A appears earlier than inhibitory GABA interneurons serotonin 2C receptors.Reference Lambe, Fillman, Webster and Shannon Weickert118 These ontological delays could potentially contribute to the E/I imbalance.

While SSRI use is linked to some benefits in the reduction of RRBI, caution is required while using them in younger children. While there is a risk of dose-related activation, it is imperative to slow titration and discontinue if behavioral activation develops. Also, the role of precision medicine and pharmacogenetics data could clarify the nuanced individual variability. With emerging data linking higher deaths by suicide in ASD individuals it is paramount to be informed about the limitation of SSRIs and controversies about its association with black box warnings.Reference Gupta and Gupta119 Buspirone could be a non-SSRI alternative for anxiety disorders and mirtazapine for depressive illness.

Distinct sleep issues need special attention

Last, but not least, sleep disturbances have been extensively reported and studied and prevalence rates fall between 64% and 93% without much gender differences (girls may have stronger links between SD and RB). This section has been extensively covered in our earlier articles. SD is also known to be independently linked with worsening of the core symptoms and requires screening specifically for SD. The relationship is bidirectional and accounts for at least 22–32% variance in behavioral problems. The role of ASP in regulating arousal and over-responsivity to environmental stimuli has been found to be associated with SD. There is a strong neurobiological basis for SD in ASD, including genetic mutations in the circadian clock and non-circadian genes. Besides screening, it is also important to evaluate and identify if presentations and symptoms are consistent with subcategories like sleep-related breathing disorders, circadian rhythm sleep–wake disorders, sleep-related movement disorders (SLMD), and/or parasomnias. On some occasions, further gold-standard testing like polysomnography and expert evaluation with sleep specialists is required. With highly complex clinical phenotypes, the presence of comorbid disorders like ADHD could further add to the complexity, requiring knowledge of pharmacokinetics, safety, medication efficacy, available efficacy data, agent safety, dosing, and drug–drug interactions with concomitant medications. There is overwhelming empirical support for prolonged-release melatonin (PedPRM), an easy-to-use formulation that is efficacious, and a safe option for long-term treatment that improves caregivers’ quality of life. Interestingly, in individuals with ASD and SLMD, there is weak evidence for iron supplementation to augment ferritin levels to more than 50 ng/dl (Figure 2).

Figure 2. Overarching principles and multimodal interventions to mitigate impairments associated with ASD.

Conclusion

ASD is a heterogeneous neurodevelopmental disorder with varied phenotypic manifestations and co-occurring conditions. The presence of debilitating symptoms and impairments is often the reason for pharmacologic interventions. The rich but scattered literature on the off-label psychotropic agents in ASD alludes to the rationale, efficacy, and risks associated. Treatment aims to initiate the minimal effective dose while carefully considering the psychopharmacological agent’s risks, benefits, and side effects. Most medications being used for CA with mental health conditions are prescribed off-label, and therefore patient assent and parental informed consent are paramount. Although psychopharmacological interventions are no panacea, whenever available, behavioral and cognitive interventions along with parent training must be tried alone or in combination for improved efficacies.

It is recommended to start at a lower-than-usual dose and increase the dose every five half-lives of the drug, given the increased susceptibility to side effects in individuals with ASD. ASD populations are at a higher risk of experiencing side effects, including EPS and weight gain, and should be carefully monitored. An individual’s metabolic profile, waist circumference, and weight should be monitored regularly. In the event of weight gain, various interventions such as lifestyle modifications may be useful. It is important to monitor closely for side effects since there could be medical conditions that result in more severe side effects, such as GERD, and consultation with other subspecialties may be needed.

Limitations

Although we have endeavored to provide a comprehensive overview of the existing evidence, it is important to note that this is a narrative literature review. Our interpretation of the studies and conclusions drawn from them may be influenced by selection biases. To avoid any possibility of misleading conclusions, we not only reviewed clinical trials for objective data but also tried to include expert opinions or any other narrative reviews. This information must be used carefully, and clinical judgment must be exercised as the overall evidence supporting it is either low or insufficient and it continues to be off-label. There are many overlapping symptoms in the co-occurring disorders, which could be difficult to differentiate. The review also does not focus on multimodal treatments often recommended for these conditions.

Future directions

There are many ongoing trials to empirically test the scientific hypothesis that continues to provide guidance and hope for the future of therapeutics for ASD. Metformin is commonly used for the treatment of antipsychotic-induced weight gain is metforminReference de Silva, Suraweera, Ratnatunga, Dayabandara, Wanniarachchi and Hanwella120 but it is also beneficial in rodent models (AMPK activator-related neuro-protective effects) for spatial and verbal memory. The early use of postnatal metformin improved social interactions and reduced repetitive behavior in Black and Tan Brachyury (BTBR) mice.Reference Wang, Cai and Fan121 Despite its neuroprotective effects, a 2018 meta-analysis found no further improvement in mini mental status exam (MMSE) scores, and though metformin may be helpful, no data supports its use in individuals without diabetes.Reference Campbell, Stephenson, de Courten, Chapman, Bellman and Aromataris122 A 32-week, two-phase study was conducted examining the effects of metformin on spatial and verbal memory in 51 patients with atypical antipsychotics-associated weight gain; results showed no difference in spatial or verbal memory over 32 weeks, although there was nominally better spatial memory on the NSPSY-II in 16 weeks.Reference Aman, Hollway and Veenstra-VanderWeele123

Fragile X syndrome (FXS) is one of the most common genetic conditions associated with ASD. A case report of two individuals with FXS who had been treated with metformin clinically for 1 year showed significant cognitive and behavioral improvements, as well as normalizing their weight percentiles.Reference Protic, Aydin, Tassone, Tan, Hagerman and Schneider124 In a DB-RCT currently underway at the University of California, Davis (2022), for individuals with FXS ages 6–25, metformin will be evaluated for the treatment of language deficits, behavior problems, and obesity/excessive appetite.125

There are many preliminary trials of propranolol to study its effects on cognitive functioning in ASD, which includes improvement in verbal problem-solvingReference Beversdorf, Carpenter, Miller, Cios and Hillier126 verbal decision-making,Reference Narayanan, White and Saklayen127 and category fluency.Reference Beversdorf, Saklayen, Higgins, Bodner, Kanne and Christ128 A series of trials tested the short-term effect of administration of propranolol and found improved working memory,Reference Bodner, Beversdorf, Saklayen and Christ129 reduction in oral fixation,Reference Zamzow, Christ and Saklayen130 and conversational reciprocity.Reference Zamzow, Ferguson and Stichter131 Propranolol led to faster performance in verbal problem-solving, and there was an improvement in autonomic activity and anxiety, which can be potential biomarkers for response to propranolol.Reference Zamzow, Ferguson, Ragsdale, Lewis and Beversdorf132 Two studies used fMRI and found increased functional connectivity, which could be a potential image marker of cognitive effects of propranolol in ASD.Reference Narayanan, White and Saklayen127, Reference Hegarty, Ferguson and Zamzow133 London did a retrospective study on 46 adults and adolescents with ASD with adjunct propranolol and found that 85% of participants had much or very much improved CGI scale.Reference London, Yoo, Fethke and Zimmerman-Bier134

Based on mouse models, the IGF-AKT–mTOR signaling network is over-active at synapses, making neurotrophic factors like IGF-1 potentially useful for the treatment of ASD by reducing neuroinflammation. Researchers have found that IGF-1 may restore function in Retts and ASD caused by SCHANK3 gene deficiency, but these are preliminary findings that require further testing for their efficacy and safety.Reference Riikonen135

A promising agent has been oxytocin, which is responsible for the formation of monogamous bonds in prairie vole rats. Research has indicated low blood oxytocin levels in ASD children as compared with those with typically developing youths.Reference John and Jaeggi136 Ooi et al.Reference Ooi, Weng, Kossowsky, Gerger and Sung17 reviewed 12 RCTs in a meta-analysis of oxytocin and its effect on social cognition and on restricting repetitive behaviors and found no significant effects of oxytocin on these two core symptoms. One of the proposed explanations is the incapacity of oral or intranasal oxytocin to penetrate the blood–brain barrier. Newer molecules such as Melanotan II are being studied to improve oxytocin delivery to the brain.Reference Minakova, Lang and Medel-Matus137 Similarly, Horvath compared 5 RCT in his meta-analysis of omega 3 fatty acids in ASD and found no effects of omega 3 in individuals with ASD with a limited number of studies and a small sample size.Reference Horvath, Łukasik and Szajewska138

Another proposed etiology of ASD is inflammation and oxidative stress, and multiple agents have been investigated. Some initial evidence suggested low levels of carnitine, a cofactor for carnitine Acyltransferase enzyme, are associated with more severe symptoms of ASD.Reference Demarquoy and Demarquoy139 A review of clinical studies on levocarnitineReference Malaguarnera and Cauli140 found low evidence but suggested a subgroup of ASD with altered mitochondrial metabolism who responded to levocarnitine. This is noteworthy as mitochondrial diseases are more common in ASD compared to TDY.Reference Legido, Jethva and Goldenthal66

Another theory related to mitochondria is the dysfunctional cell danger response and integrated stress response. When under stress, mitochondria cannot regulate oxidative metabolism and the release of purine and pyrimidine nucleotides. Suramin, an inhibitor of purinergic signaling, was tested based on this hypothesis in a small (n = 10) 6 weeks pilot trial which indicated a small but significant change in ADOS-2 comparison scores with some improvement in core symptoms.Reference Naviaux, Curtis and Li141 NAC can modulate free radical pathways and help treat ASD but a DB-PC-RCTReference Dean, Gray and Villagonzalo142 found NAC 500 mg for 6 months ineffective.

Gluten and casein have also been implicated as potential etiologies of autism since they show excess opioid activity and permeability in the gut. Multiple reviews have examined the efficacy of GCFC diets.Reference Millward, Ferriter, Calver and Connell‐Jones143, Reference Piwowarczyk, Horvath, Łukasik, Pisula and Szajewska144 and found no evidence to support it. Quan suggests GCFC is “promising”, but evidence levels were low as the studies were not double-blinded, and most had performance bias. We need more high-quality studies.Reference Quan, Xu and Cui145

Sulforaphane, another potent antioxidant found in cruciferous vegetables, has been studied for its effect on autism.Reference McGuinness and Kim146 looked at three RCTs and two open-label trials, which may offer some benefits, but the quality of evidence is low due to the small size of studies that were poorly reported and moderately biased.

An open-label 12-week studyReference Hellings, Reed and Cain147 studied add-on loxapine (BDNF stimulator) doses of 5–15 mg in 16 participants with ASD who were on one or more psychotropics of different types. Most improved in their Clinical Global Impression-Improvement scale (CGI-I) scores and their Aberrant Behavior Checklist- irritability (ABC-I) subscale scores with minimal side effects. They measured loxapine and metabolites 7-hydroxyloxapine, 8-hydroxyloxapine, and amoxapine concentrations in addition to using the brain-derived neurotrophic factor (BDNF) as a biomarker of neuromodulation and found that the concentration of BDNF increased statistically significantly. Low-dose loxapine is a medium potency antipsychotic with D2 and 5HT2 receptor blockage, mostly resembling SGA at low dosages,Reference Li, Ichikawa and Meltzer148 and may be an option for irritability. A retrospective chart review of 50 CA with ASD and hyperactivity and impulsivity who have been prescribed amitriptyline (mean dose of 1.3 mg/kg/day). The results showed that 60% of the participants improved in their CGI-I scores.Reference Bhatti, Thome and Smith149 The use of brain-derived neurotropic factor stimulators, such as loxapine or amitriptyline, deserves further study.Reference Hellings, Arnold and Han150

NMDA receptor antagonists memantine and amantadine have also been studied. Initially, memantine showed promise,Reference Chez, Burton, Dowling, Chang, Khanna and Kramer151, Reference Ghaleiha, Asadabadi and Mohammadi152 but later trials have been negative.Reference Aman, Findling and Hardan153, Reference Hardan, Hendren and Aman154 Open-label trials in adults with ASDReference Joshi, Wozniak and Faraone155 found some benefits. A recent small study found that memantine improved verbal recognition memory as measured by Narrative Memory-Recognition (NEPSY-II) (p = .03).Reference Soorya, Fogg and Ocampo156 The tolerability of memantine was not a concern in any of the studies. As with amantadine, early studies showed promise, either as a monotherapyReference King, Wright and Handen157 or in combination with risperidone,Reference Mohammadi, Yadegari and Hassanzadeh158 but the quality of the evidence is low. Based on a retrospective cohort study conducted at Baylor University in Texas between 2014 and 2018 on 297 children ages 6–18 years with psychiatric disorders (ADHD being the most common 251/297), amantadine (median dose 200 mg daily) resulted in favorable clinical outcomes in 67.8% (40/59) of individuals with ASD, suggesting its benefits in individuals with ADHD comorbid with ASD.Reference Morrow, Choi, Young, Haidar, Boduch and Bourgeois159

Abbreviations

ABC

The Aberrant Behavior Checklist (ABC) measures psychiatric symptoms and behavioral disturbances exhibited by individuals across 5 domains: Irritability, Agitation, and Crying; Lethargy/Social Withdrawal; Stereotypic Behavior; Hyperactivity/Noncompliance

ABC-I

Aberrant Behavior Checklist (ABC) Irritability Subscale

ADHD

attention deficit hyperactivity disorder

ADOS-2

Autism Diagnostic Observation Schedule-2

AMP

amphetamines

AMPK

AMP-activated protein kinase

Anti-5HT

Anti- 5-hydroxytryptamine

ARFID

Avoidant/Restrictive Food Intake Disorder

AS

Antipsychotics

ATX

atomoxetine

ASD

autism spectrum disorder

BDNF

Brain-Derived Neurotrophic Factor

BTBR

inbred mouse strain with face validity as a preclinical model of the core autism symptom domains

CGI-S

The Clinical Global Impression – Severity scale (CGI-S). A seven-point scale that requires the clinician to rate the severity of the patient’s illness at the time of assessment

CPRS

Children’s Psychiatric Rating Scale

CXR

Clonidine Extended-Release

CYBOCS-PDD

Children’s Yale-Brown Obsessive-Compulsive Scale modified for pervasive developmental disorders

D2 receptors

Dopamine Receptor Type 2

DB-PC-RCT

Double-Blind Placebo-Controlled Randomized Control Trial

DSM

Diagnostic and Statistical Manual of Mental Disorders

EEG

Electroencephalogram

EPS

Extrapyramidal Side Effects

FDA

US. Food and Drug Administration

FIP

Focused Intervention Practices

fMRI

Functional Magnetic Resonance Imaging

FXS

Fragile X Syndrome

GCFC

Gluten Free Casein Free diet

GERD

Gastroesophageal Reflux Disease

GXR

Guanfacine Extended-Release

IGF-1

Insulin-Like Growth Factor 1

MMSE

Mini Mental Status Exam

MPH

Methylphenidate

NAC

N-Acetylcysteine

NDD

Neurodevelopmental disorder

NMDA

N-Methyl-D-aspartic acid

NSPSY-II

Developmental NEuroPSYchological Assessment

OCD

Obsessive-Compulsive Disorder

PARS

Pediatric Anxiety Rating Scale

PECS

Picture exchange communication systems

PedPRM

pediatric prolonged-release melatonin

PT

Parental Therapy

RCT

Randomized control trial

RRBI

Restricted and Repetitive Behaviors and Interests

RUPP

Research Units of Pediatric Psychopharmacology

SCI

social and communication impairment

SCHANK3 gene

SH3 And Multiple Ankyrin Repeat Domains 3

SSRI

Selective serotonin reuptake inhibitors

SGA

Second-Generation Antipsychotics

TDY

Typically Developing Youths

5HT1A receptors

serotonin 1A receptor

5HT2

Receptors a subfamily of 5-HydroxyTryptamine receptors

Author contribution

Conceptualization: N.G.; Data curation: N.G.; Investigation: M.G.; Project administration: M.G.; Writing – review & editing: M.G.

Disclosure

The authors declare none.

References

Maenner, MJ. Prevalence and characteristics of autism spectrum disorder among children aged 8 years — autism and developmental disabilities monitoring network, 11 Sites, United States. MMWR Surveill Summ. 2020;2023:72. doi:10.15585/mmwr.ss7202a1Google Scholar
Goel, R, Hong, JS, Findling, RL, Ji, NY. An update on pharmacotherapy of autism spectrum disorder in children and adolescents. Int Rev Psychiatry Abingdon Engl. 2018;30(1):7895. doi:10.1080/09540261.2018.1458706CrossRefGoogle ScholarPubMed
Gosling, CJ, Cartigny, A, Mellier, BC, Solanes, A, Radua, J, Delorme, R. Efficacy of psychosocial interventions for autism spectrum disorder: an umbrella review. Mol Psychiatry. 2022;27(9):36473656. doi:10.1038/s41380-022-01670-zCrossRefGoogle ScholarPubMed
Subramanyam, AA, Mukherjee, A, Dave, M, Chavda, K. Clinical practice guidelines for autism spectrum disorders. Indian J Psychiatry. 2019;61(Suppl 2):254. doi:10.4103/psychiatry.IndianJPsychiatry_542_18CrossRefGoogle ScholarPubMed
Blankenship, K, Erickson, CA, Stigler, KA, Posey, DJ, McDougle, CJ. Aripiprazole for irritability associated with autistic disorder in children and adolescents aged 6–17 years. Pediatr Health. 2010;4(4):375381.CrossRefGoogle ScholarPubMed
Simonoff, E, Pickles, A, Charman, T, Chandler, S, Loucas, T, Baird, G. Psychiatric disorders in children with autism spectrum disorders: prevalence, comorbidity, and associated factors in a population-derived sample. J Am Acad Child Adolesc Psychiatry. 2008;47(8):921929. doi:10.1097/CHI.0b013e318179964fCrossRefGoogle Scholar
Bonati, M, Jacqz-Aigrain, E, Choonara, I. Licensed medicines, off-label use or evidence-based. Which is most important? Arch Dis Child. 2017;102(1):5354. doi:10.1136/archdischild-2016-311527CrossRefGoogle ScholarPubMed
Syed, SA, Dixson, BA, Constantino, E, Regan, J. The law and practice of off-label prescribing and physician promotion. J Am Acad Psychiatry Law. 2021;49(1):5359. doi:10.29158/JAAPL.200049-20Google ScholarPubMed
Braüner, JV, Johansen, LM, Roesbjerg, T, Pagsberg, AK. Off-label prescription of psychopharmacological drugs in child and adolescent psychiatry. J Clin Psychopharmacol. 2016;36(5):500507. doi:10.1097/JCP.0000000000000559CrossRefGoogle ScholarPubMed
Lopez-Leon, S, Lopez-Gomez, MI, Warner, B, Ruiter-Lopez, L. Psychotropic medication in children and adolescents in the United States in the year 2004 vs 2014. Daru J Fac Pharm Tehran Univ Med Sci. 2018;26(1):510. doi:10.1007/s40199-018-0204-6CrossRefGoogle ScholarPubMed
Madden, JM, Lakoma, MD, Lynch, FL, et al. Psychotropic medication use among insured children with autism spectrum disorder. J Autism Dev Disord. 2017;47(1):144154. doi:10.1007/s10803-016-2946-7CrossRefGoogle ScholarPubMed
Bejarano-Martín, Á, Canal-Bedia, R, Magán-Maganto, M, et al. Efficacy of focused social and communication intervention practices for young children with autism spectrum disorder: A meta-analysis. Early Child Res Q. 2020;51:430445. doi:10.1016/j.ecresq.2020.01.004CrossRefGoogle Scholar
Williams, K, Brignell, A, Randall, M, Silove, N, Hazell, P. Selective serotonin reuptake inhibitors (SSRIs) for autism spectrum disorders (ASD). Cochrane Database Syst Rev. 2013;(8):CD004677. doi:10.1002/14651858.CD004677.pub3Google ScholarPubMed
Zhou, MS, Nasir, M, Farhat, LC, Kook, M, Artukoglu, BB, Bloch, MH. Meta-analysis: pharmacologic treatment of restricted and repetitive behaviors in autism spectrum disorders. J Am Acad Child Adolesc Psychiatry. 2021;60(1):3545. doi:10.1016/j.jaac.2020.03.007CrossRefGoogle ScholarPubMed
McDougle, CJ, Scahill, L, Aman, MG, et al. Risperidone for the core symptom domains of autism: results from the study by the autism network of the research units on pediatric psychopharmacology. Am J Psychiatry. 2005;162(6):11421148. doi:10.1176/appi.ajp.162.6.1142CrossRefGoogle Scholar
Canitano, R, Scandurra, V. Risperidone in the treatment of behavioral disorders associated with autism in children and adolescents. Neuropsychiatr Dis Treat. 2008;4(4):723730. doi:10.2147/ndt.s1450CrossRefGoogle ScholarPubMed
Ooi, YP, Weng, SJ, Kossowsky, J, Gerger, H, Sung, M. Oxytocin and autism spectrum disorders: a systematic review and meta-analysis of randomized controlled trials. Pharmacopsychiatry. 2017;50(1):513. doi:10.1055/s-0042-109400Google ScholarPubMed
Handen, BL, Johnson, CR, Lubetsky, M. Efficacy of methylphenidate among children with autism and symptoms of attention-deficit hyperactivity disorder. J Autism Dev Disord. 2000;30:245255. doi:10.1023/A:1005548619694CrossRefGoogle ScholarPubMed
Quintana, H, Birmaher, B, Stedge, D, et al. Use of methylphenidate in the treatment of children with autistic disorder. J Autism Dev Disord. 1995;25(3):283294. doi:10.1007/BF02179289CrossRefGoogle ScholarPubMed
Research Units on Pediatric Psychopharmacology Autism Network. Randomized, controlled, crossover trial of methylphenidate in pervasive developmental disorders with hyperactivity. Arch Gen Psychiatry. 2005;62(11):12661274. doi:10.1001/archpsyc.62.11.1266CrossRefGoogle Scholar
Sturman, N, Deckx, L, van Driel, ML. Methylphenidate for children and adolescents with autism spectrum disorder. Cochrane Database Syst Rev. 2017;11:CD011144. doi:10.1002/14651858.CD011144.pub2Google ScholarPubMed
Pearson, DA, Santos, CW, Aman, MG, et al. Effects of extended release methylphenidate treatment on ratings of attention-deficit/hyperactivity disorder (ADHD) and associated behavior in children with autism spectrum disorders and ADHD symptoms. J Child Adolesc Psychopharmacol. 2013;23(5):337351. doi:10.1089/cap.2012.0096CrossRefGoogle ScholarPubMed
Kim, SJ, Shonka, S, French, WP, Strickland, J, Miller, L, Stein, MA. Dose-response effects of long-acting liquid methylphenidate in children with attention deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD): a pilot study. J Autism Dev Disord. 2017;47(8):23072313. doi:10.1007/s10803-017-3125-1CrossRefGoogle ScholarPubMed
Pearson, DA, Santos, CW, Aman, MG, et al. Effects of extended-release methylphenidate treatment on cognitive task performance in children with autism spectrum disorder and attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol. 2020;30(7):414426. doi:10.1089/cap.2020.0004CrossRefGoogle ScholarPubMed
Joshi, G, Wilens, T, Firmin, ES, Hoskova, B, Biederman, J. Pharmacotherapy of attention deficit/hyperactivity disorder in individuals with autism spectrum disorder: a systematic review of the literature. J Psychopharmacol Oxf Engl. 2021;35(3):203210. doi:10.1177/0269881120972336CrossRefGoogle ScholarPubMed
Ghanizadeh, A, Molla, M, Olango, GJ. The effect of stimulants on irritability in autism comorbid with ADHD: a systematic review. Neuropsychiatr Dis Treat. 2019;15:1547. doi:10.2147/NDT.S194022CrossRefGoogle ScholarPubMed
Troost, PW, Steenhuis, MP, Tuynman-Qua, HG, et al. Atomoxetine for attention-deficit/hyperactivity disorder symptoms in children with pervasive developmental disorders: a pilot study. J Child Adolesc Psychopharmacol. 2006;16(5):611619. doi:10.1089/cap.2006.16.611CrossRefGoogle ScholarPubMed
Posey, D. J., Wiegand, R. E., Wilkerson, J., Maynard, M., Stigler, K. A., & McDougle, C. J. (2006). Open-label atomoxetine for attention-deficit/ hyperactivity disorder symptoms associated with high-functioning pervasive developmental disorders. Journal of child and adolescent psychopharmacology, 16(5), 599610. https://doi.org/10.1089/cap.2006.16.599.CrossRefGoogle ScholarPubMed
Harfterkamp, M, van de Loo-Neus, G, Minderaa, RB, et al. A randomized double-blind study of atomoxetine versus placebo for attention-deficit/hyperactivity disorder symptoms in children with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2012;51(7):733741. doi:10.1016/j.jaac.2012.04.011CrossRefGoogle ScholarPubMed
van der Meer, JMJ, Harfterkamp, M, van de Loo-Neus, G, et al. A randomized, double-blind comparison of atomoxetine and placebo on response inhibition and interference control in children and adolescents with autism spectrum disorder and comorbid attention-deficit/hyperactivity disorder symptoms. J Clin Psychopharmacol. 2013;33(6):824827. doi:10.1097/JCP.0b013e31829c764fCrossRefGoogle ScholarPubMed
Harfterkamp, M, Buitelaar, JK, Minderaa, RB, van de Loo-Neus, G, van der Gaag, RJ, Hoekstra, PJ. Atomoxetine in autism spectrum disorder: no effects on social functioning; some beneficial effects on stereotyped behaviors, inappropriate speech, and fear of change. J Child Adolesc Psychopharmacol. 2014;24(9):481485. doi:10.1089/cap.2014.0026CrossRefGoogle ScholarPubMed
Harfterkamp, M, Buitelaar, JK, Minderaa, RB, van de Loo-Neus, G, van der Gaag, RJ, Hoekstra, PJ. Long-term treatment with atomoxetine for attention-deficit/hyperactivity disorder symptoms in children and adolescents with autism spectrum disorder: an open-label extension study. J Child Adolesc Psychopharmacol. 2013;23(3):194199. doi:10.1089/cap.2012.0012CrossRefGoogle ScholarPubMed
Handen, BL, Aman, MG, Arnold, LE, et al. Atomoxetine, parent training, and their combination in children with autism spectrum disorder and attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2015;54(11):905915. doi:10.1016/j.jaac.2015.08.013CrossRefGoogle ScholarPubMed
Smith, T, Aman, MG, Arnold, LE, et al. Atomoxetine and parent training for children with autism and attention-deficit/hyperactivity disorder: a 24-week extension study. J Am Acad Child Adolesc Psychiatry. 2016;55(10):868876.e2. doi:10.1016/j.jaac.2016.06.015CrossRefGoogle ScholarPubMed
Arnold, LE, Ober, N, Aman, MG, et al. A 1.5-year follow-up of parent training and atomoxetine for attention-deficit/hyperactivity disorder symptoms and noncompliant/disruptive behavior in autism. J Child Adolesc Psychopharmacol. 2018;28(5):322330. doi:10.1089/cap.2017.0134CrossRefGoogle Scholar
Conners, CK, Casat, CD, Gualtieri, CT, et al. Bupropion hydrochloride in attention deficit disorder with hyperactivity. J Am Acad Child Adolesc Psychiatry. 1996;35(10):13141321. doi:10.1097/00004583-199610000-00018CrossRefGoogle ScholarPubMed
Leverich, GS, Altshuler, LL, Frye, MA, et al. Risk of switch in mood polarity to hypomania or mania in patients with bipolar depression during acute and continuation trials of venlafaxine, sertraline, and bupropion as adjuncts to mood stabilizers. Am J Psychiatry. 2006;163(2):232239. doi:10.1176/appi.ajp.163.2.232CrossRefGoogle ScholarPubMed
Post, RM, Altshuler, LL, Frye, MA, et al. New findings from the bipolar collaborative network: clinical implications for therapeutics. Curr Psychiatry Rep. 2006;8(6):489497. doi:10.1007/s11920-006-0056-5CrossRefGoogle ScholarPubMed
Johnson, KP, Malow, BA. Sleep in children with autism spectrum disorders. Curr Treat Options Neurol. 2008;10(5):350359. doi:10.1007/s11940-008-0038-5CrossRefGoogle ScholarPubMed
Banas, K, Sawchuk, B. Clonidine as a treatment of behavioural disturbances in autism spectrum disorder: a systematic literature review. J Can Acad Child Adolesc Psychiatry 2020;29(2):110120.Google ScholarPubMed
Scahill, L, McCracken, JT, King, BH, et al. Extended-release guanfacine for hyperactivity in children with autism spectrum disorder. Am J Psychiatry. 2015;172(12):11971206. doi:10.1176/appi.ajp.2015.15010055CrossRefGoogle ScholarPubMed
Politte, LC, Scahill, L, Figueroa, J, McCracken, JT, King, B, McDougle, CJ. A randomized, placebo-controlled trial of extended-release guanfacine in children with autism spectrum disorder and ADHD symptoms: an analysis of secondary outcome measures. Neuropsychopharmacol 2018;43(8):17721778. doi:10.1038/s41386-018-0039-3CrossRefGoogle ScholarPubMed
Lamberti, M, Siracusano, R, Italiano, D, et al. Head-to-head comparison of aripiprazole and risperidone in the treatment of ADHD symptoms in children with autistic spectrum disorder and ADHD: a pilot, open-label, randomized controlled study. Paediatr Drugs. 2016;18(4):319329. doi:10.1007/s40272-016-0183-3CrossRefGoogle ScholarPubMed
McCracken, JT, McGough, J, Shah, B, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med. 2002;347(5):314321. doi:10.1056/NEJMoa013171CrossRefGoogle ScholarPubMed
Shea, S, Turgay, A, Carroll, A, et al. Risperidone in the treatment of disruptive behavioral symptoms in children with autistic and other pervasive developmental disorders. Pediatrics. 2004;114(5):e634e641. doi:10.1542/peds.2003-0264-FCrossRefGoogle ScholarPubMed
Aman, MG, Arnold, LE, McDougle, CJ, et al. Acute and long-term safety and tolerability of risperidone in children with autism. J Child Adolesc Psychopharmacol. 2005;15(6):869884. doi:10.1089/cap.2005.15.869CrossRefGoogle ScholarPubMed
Marcus, RN, Owen, R, Kamen, L, et al. A placebo-controlled, fixed-dose study of aripiprazole in children and adolescents with irritability associated with autistic disorder. J Am Acad Child Adolesc Psychiatry. 2009;48(11):11101119. doi:10.1097/CHI.0b013e3181b76658CrossRefGoogle ScholarPubMed
Owen, R, Sikich, L, Marcus, RN, et al. Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics. 2009;124(6):15331540. doi:10.1542/peds.2008-3782CrossRefGoogle ScholarPubMed
Chen, JX, Su, YA, Bian, QT, et al. Adjunctive aripiprazole in the treatment of risperidone-induced hyperprolactinemia: a randomized, double-blind, placebo-controlled, dose-response study. Psychoneuroendocrinology. 2015;58:130140. doi:10.1016/j.psyneuen.2015.04.011CrossRefGoogle ScholarPubMed
Chen, S, Barner, JC, Cho, E. Trends in off-label use of antipsychotic medications among Texas Medicaid children and adolescents from 2013 to 2016. J Manag Care Spec Pharm. 2021;27(8):10351045. doi:10.18553/jmcp.2021.27.8.1035Google ScholarPubMed
Loebel, A, Brams, M, Goldman, RS, et al. Lurasidone for the treatment of irritability associated with autistic disorder. J Autism Dev Disord. 2016;46(4):11531163. doi:10.1007/s10803-015-2628-xCrossRefGoogle ScholarPubMed
Gupta, M, Hoover, G. Lurasidone an effective alternative for the treatment of irritability associated with autism spectrum disorder. Cureus. 2020;12(12):e12360. doi:10.7759/cureus.12360Google ScholarPubMed
Golubchik, P, Sever, J, Weizman, A. Low-dose quetiapine for adolescents with autistic spectrum disorder and aggressive behavior: open-label trial. Clin Neuropharmacol. 2011;34(6):216219. doi:10.1097/WNF.0b013e31823349acCrossRefGoogle ScholarPubMed
Malone, RP, Cater, J, Sheikh, RM, Choudhury, MS, Delaney, MA. Olanzapine versus haloperidol in children with autistic disorder: an open pilot study. J Am Acad Child Adolesc Psychiatry. 2001;40(8):887894. doi:10.1097/00004583-200108000-00009CrossRefGoogle ScholarPubMed
Potenza, MN, Holmes, JP, Kanes, SJ, McDougle, CJ. Olanzapine treatment of children, adolescents, and adults with pervasive developmental disorders: an open-label pilot study. J Clin Psychopharmacol. 1999;19(1):3744. doi:10.1097/00004714-199902000-00008CrossRefGoogle ScholarPubMed
Hollander, E, Wasserman, S, Swanson, EN, et al. A double-blind placebo-controlled pilot study of olanzapine in childhood/adolescent pervasive developmental disorder. J Child Adolesc Psychopharmacol. 2006;16(5):541548. doi:10.1089/cap.2006.16.541CrossRefGoogle ScholarPubMed
Fido, A, Al-Saad, S. Olanzapine in the treatment of behavioral problems associated with autism: an open-label trial in Kuwait. Med Princ Pract 2008;17(5):415418. doi:10.1159/000141508CrossRefGoogle ScholarPubMed
Lambrey, S, Falissard, B, Martin-Barrero, M, et al. Effectiveness of clozapine for the treatment of aggression in an adolescent with autistic disorder. J Child Adolesc Psychopharmacol. 2010;20(1):7980. doi:10.1089/cap.2009.0057CrossRefGoogle Scholar
Rothärmel, M, Szymoniak, F, Pollet, C, et al. Eleven years of clozapine experience in autism spectrum disorder: efficacy and tolerance. J Clin Psychopharmacol. 2018;38(6):577581. doi:10.1097/JCP.0000000000000955CrossRefGoogle Scholar
Joshi, G, Biederman, J, Wozniak, J, et al. Response to second generation antipsychotics in youth with comorbid bipolar disorder and autism spectrum disorder. CNS Neurosci Ther. 2012;18(1):2833. doi:10.1111/j.1755-5949.2010.00219.xCrossRefGoogle ScholarPubMed
Stigler, KA, McDougle, CJ. Pharmacotherapy of irritability in pervasive developmental disorders. Child Adolesc Psychiatr Clin N Am. 2008;17(4):739752, vii-viii. doi:10.1016/j.chc.2008.06.002CrossRefGoogle ScholarPubMed
Hollander, E, Dolgoff-Kaspar, R, Cartwright, C, Rawitt, R, Novotny, S. An open trial of divalproex sodium in autism spectrum disorders. J Clin Psychiatry. 2001;62(7):530534. doi:10.4088/jcp.v62n07a05CrossRefGoogle ScholarPubMed
Hollander, E, Chaplin, W, Soorya, L, et al. Divalproex sodium vs placebo for the treatment of irritability in children and adolescents with autism spectrum disorders. Neuropsychopharmacology. 2010;35(4):990998. doi:10.1038/npp.2009.202CrossRefGoogle ScholarPubMed
Hollander, E, Soorya, L, Wasserman, S, Esposito, K, Chaplin, W, Anagnostou, E. Divalproex sodium vs. placebo in the treatment of repetitive behaviours in autism spectrum disorder. Int J Neuropsychopharmacol. 2006;9(2):209213. doi:10.1017/S1461145705005791CrossRefGoogle ScholarPubMed
Hellings, JA, Weckbaugh, M, Nickel, EJ, et al. A double-blind, placebo-controlled study of valproate for aggression in youth with pervasive developmental disorders. J Child Adolesc Psychopharmacol. 2005;15(4):682692. doi:10.1089/cap.2005.15.682CrossRefGoogle ScholarPubMed
Legido, A, Jethva, R, Goldenthal, MJ. Mitochondrial dysfunction in autism. Semin Pediatr Neurol. 2013;20(3):163175. doi:10.1016/j.spen.2013.10.008CrossRefGoogle ScholarPubMed
Krähenbühl, S, Brandner, S, Kleinle, S, Liechti, S, Straumann, D. Mitochondrial diseases represent a risk factor for valproate-induced fulminant liver failure. Liver. 2000;20(4):346348. doi:10.1034/j.1600-0676.2000.020004346.xCrossRefGoogle ScholarPubMed
Rezaei, V, Mohammadi, MR, Ghanizadeh, A, et al. Double-blind, placebo-controlled trial of risperidone plus topiramate in children with autistic disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(7):12691272. doi:10.1016/j.pnpbp.2010.07.005CrossRefGoogle ScholarPubMed
Mintz, M, Hollenberg, E. Revisiting lithium: utility for behavioral stabilization in adolescents and adults with autism spectrum disorder. Psychopharmacol Bull. 2019;49(2):2840.Google ScholarPubMed
Assistance Publique - Hôpitaux de Paris. Effect of Lithium in Patients with Autism Spectrum Disorder and Phelan-McDermid Syndrome (SHANK3 Haploinsufficiency): Pilot Study. clinicaltrials.gov; 2022; https://clinicaltrials.gov/ct2/show/NCT04623398. Accessed March 10, 2022.Google Scholar
Yuan, J, Song, J, Zhu, D, et al. Lithium treatment is safe in children with intellectual disability. Front Mol Neurosci. 2018;11:425. doi:10.3389/fnmol.2018.00425CrossRefGoogle ScholarPubMed
Belsito, KM, Law, PA, Kirk, KS, Landa, RJ, Zimmerman, AW. Lamotrigine therapy for autistic disorder: a randomized, double-blind, placebo-controlled trial. J Autism Dev Disord. 2001;31(2):175181. doi:10.1023/a:1010799115457CrossRefGoogle ScholarPubMed
Guglielmo, R, Ioime, L, Grandinetti, P, Janiri, L. Managing disruptive and compulsive behaviors in adult with autistic disorder with gabapentin. J Clin Psychopharmacol. 2013;33(2):273274. doi:10.1097/JCP.0b013e318285680cCrossRefGoogle ScholarPubMed
Robinson, AA, Malow, BA. Gabapentin shows promise in treating refractory insomnia in children. J Child Neurol. 2013;28(12):16181621. doi:10.1177/0883073812463069CrossRefGoogle ScholarPubMed
Rossignol, DA, Frye, RE. Melatonin in autism spectrum disorders: a systematic review and meta-analysis. Dev Med Child Neurol. 2011;53(9):783792. doi:10.1111/j.1469-8749.2011.03980.xCrossRefGoogle ScholarPubMed
Gringras, P, Gamble, C, Jones, AP, et al. Melatonin for sleep problems in children with neurodevelopmental disorders: randomised double masked placebo controlled trial. BMJ. 2012;345:e6664. doi:10.1136/bmj.e6664CrossRefGoogle ScholarPubMed
Gringras, P, Nir, T, Breddy, J, Frydman-Marom, A, Findling, RL. Efficacy and safety of pediatric prolonged-release melatonin for insomnia in children with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2017;56(11):948957.e4. doi:10.1016/j.jaac.2017.09.414CrossRefGoogle ScholarPubMed
Maras, A, Schroder, CM, Malow, BA, et al. Long-term efficacy and safety of pediatric prolonged-release melatonin for insomnia in children with autism spectrum disorder. J Child Adolesc Psychopharmacol. 2018;28(10):699710. doi:10.1089/cap.2018.0020CrossRefGoogle ScholarPubMed
Malow, BA, Findling, RL, Schroder, CM, et al. Sleep, growth, and puberty after 2 years of prolonged-release melatonin in children with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2021;60(2):252261.e3. doi:10.1016/j.jaac.2019.12.007CrossRefGoogle ScholarPubMed
Pranzatelli, MR, Tate, ED, Dukart, WS, Flint, MJ, Hoffman, MT, Oksa, AE. Sleep disturbance and rage attacks in opsoclonus-myoclonus syndrome: response to trazodone. J Pediatr. 2005;147(3):372378. doi:10.1016/j.jpeds.2005.05.016CrossRefGoogle ScholarPubMed
Kem, DL, Posey, DJ, McDougle, CJ. Priapism associated with trazodone in an adolescent with autism. J Am Acad Child Adolesc Psychiatry. 2002;41(7):758. doi:10.1097/00004583-200207000-00004CrossRefGoogle Scholar
Zuddas, A, Di Martino, A, Muglia, P, Cianchetti, C. Long-term risperidone for pervasive developmental disorder: efficacy, tolerability, and discontinuation. J Child Adolesc Psychopharmacol. 2000;10(2):7990. doi:10.1089/cap.2000.10.79CrossRefGoogle ScholarPubMed
First-line treatment for insomnia should not include routine use of antipsychotics, say Canadian psychiatrists - Canadian Psychiatric Association - Association des psychiatres du Canada; https://www.cpa-apc.org/first-line-treatment-for-insomnia-should-not-include-routine-use-of-antipsychotics-say-canadian-psychiatrists/. Accessed January 5, 2022.Google Scholar
Sutcliffe, JS, Delahanty, RJ, Prasad, HC, et al. Allelic heterogeneity at the serotonin transporter locus (SLC6A4) confers susceptibility to autism and rigid-compulsive behaviors. Am J Hum Genet. 2005;77(2):265279. doi:10.1086/432648CrossRefGoogle ScholarPubMed
Rai, D, Heuvelman, H, Dalman, C, et al. Association between autism spectrum disorders with or without intellectual disability and depression in young adulthood. JAMA Netw Open. 2018;1(4):e181465. doi:10.1001/jamanetworkopen.2018.1465CrossRefGoogle ScholarPubMed
Nimmo-Smith, V, Heuvelman, H, Dalman, C, et al. Anxiety disorders in adults with autism spectrum disorder: a population-based study. J Autism Dev Disord. 2020;50(1):308318. doi:10.1007/s10803-019-04234-3CrossRefGoogle ScholarPubMed
Jassi, AD, Vidal-Ribas, P, Krebs, G, Mataix-Cols, D, Monzani, B. Examining clinical correlates, treatment outcomes and mediators in young people with comorbid obsessive-compulsive disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry. 2021;32(7), 12011210. doi:10.1007/s00787-021-01921-4CrossRefGoogle ScholarPubMed
Reddihough, DS, Marraffa, C, Mouti, A, et al. Effect of fluoxetine on obsessive-compulsive behaviors in children and adolescents with autism spectrum disorders: a randomized clinical trial. JAMA. 2019;322(16):15611569. doi:10.1001/jama.2019.14685CrossRefGoogle ScholarPubMed
Reiersen, AM, Handen, B. Commentary on ‘selective serotonin reuptake inhibitors (SSRIs) for autism spectrum disorders (ASD).’. Evid-Based Child Health Cochrane Rev J. 2011;6(4):10821085. doi:10.1002/ebch.786CrossRefGoogle ScholarPubMed
Posey, DJ, Guenin, KD, Kohn, AE, Swiezy, NB, McDougle, CJ. A naturalistic open-label study of mirtazapine in autistic and other pervasive developmental disorders. J Child Adolesc Psychopharmacol. 2001;11(3):267277. doi:10.1089/10445460152595586CrossRefGoogle ScholarPubMed
McDougle, CJ, Thom, RP, Ravichandran, CT, et al. A randomized double-blind, placebo-controlled pilot trial of mirtazapine for anxiety in children and adolescents with autism spectrum disorder. Neuropsychopharmacology. 2022;47(6):12631270. doi:10.1038/s41386-022-01295-4CrossRefGoogle ScholarPubMed
Realmuto, GM, August, GJ, Garfinkel, BD. Clinical effect of buspirone in autistic children. J Clin Psychopharmacol. 1989;9(2):122125. doi:10.1097/00004714-198904000-00009CrossRefGoogle ScholarPubMed
McCormick, LH. Treatment with buspirone in a patient with autism. Arch Fam Med. 1997;6(4):368370. doi:10.1001/archfami.6.4.368CrossRefGoogle Scholar
Ceranoglu, TA, Wozniak, J, Fried, R, et al. A retrospective chart review of buspirone for the treatment of anxiety in psychiatrically referred youth with high-functioning autism spectrum disorder. J Child Adolesc Psychopharmacol. 2019;29(1):2833. doi:10.1089/cap.2018.0021CrossRefGoogle ScholarPubMed
Chugani, DC, Chugani, HT, Wiznitzer, M, et al. Efficacy of low-dose buspirone for restricted and repetitive behavior in young children with autism spectrum disorder: a randomized trial. J Pediatr. 2016;170:4553.e1-4. doi:10.1016/j.jpeds.2015.11.033CrossRefGoogle ScholarPubMed
Ghanizadeh, A, Ayoobzadehshirazi, A. A randomized double-blind placebo-controlled clinical trial of adjuvant buspirone for irritability in autism. Pediatr Neurol. 2015;52(1):7781. doi:10.1016/j.pediatrneurol.2014.09.017CrossRefGoogle ScholarPubMed
Howes, OD, Rogdaki, M, Findon, JL, et al. Autism spectrum disorder: consensus guidelines on assessment, treatment and research from the British Association for psychopharmacology. J Psychopharmacol. 2018;32(1):329. doi:10.1177/0269881117741766CrossRefGoogle ScholarPubMed
Koomar, T, Thomas, TR, Pottschmidt, NR, Lutter, M, Michaelson, JJ. Estimating the prevalence and genetic risk mechanisms of ARFID in a large autism cohort. Front Psychiatry. 2021;12:668297. doi:10.3389/fpsyt.2021.668297CrossRefGoogle Scholar
Bourne, L, Mandy, W, Bryant-Waugh, R. Avoidant/restrictive food intake disorder and severe food selectivity in children and young people with autism: a scoping review. Dev Med Child Neurol. 2022;64(6):691700. doi:10.1111/dmcn.15139CrossRefGoogle Scholar
Sharp, WG, Berry, RC, McCracken, C, et al. Feeding problems and nutrient intake in children with autism spectrum disorders: a meta-analysis and comprehensive review of the literature. J Autism Dev Disord. 2013;43(9):21592173. doi:10.1007/s10803-013-1771-5CrossRefGoogle ScholarPubMed
Burton, C, Allan, E, Eckhardt, S, et al. Case presentations combining family-based treatment with the unified protocols for transdiagnostic treatment of emotional disorders in children and adolescents for comorbid avoidant restrictive food intake disorder and autism spectrum disorder. J Can Acad Child Adolesc Psychiatry. 2021;30(4):280291.Google ScholarPubMed
Brigham, KS, Manzo, LD, Eddy, KT, Thomas, JJ. Evaluation and treatment of avoidant/restrictive food intake disorder (ARFID) in adolescents. Curr Pediatr Rep. 2018;6(2):107113. doi:10.1007/s40124-018-0162-yCrossRefGoogle ScholarPubMed
Salazar de Pablo, G, Pastor Jordá, C, Vaquerizo-Serrano, J, et al. Systematic review and meta-analysis: efficacy of pharmacological interventions for irritability and emotional dysregulation in autism spectrum disorder and predictors of response. J Am Acad Child Adolesc Psychiatry. 2023;62(2):151168. doi:10.1016/j.jaac.2022.03.033CrossRefGoogle ScholarPubMed
Sultan, RS, Correll, CU, Zohar, J, Zalsman, G, Veenstra-VanderWeele, J. What’s in a Name? Moving to neuroscience-based nomenclature in pediatric psychopharmacology. J Am Acad Child Adolesc Psychiatry. 2018;57(10):719721. doi:10.1016/j.jaac.2018.05.024CrossRefGoogle Scholar
McPheeters, ML, Warren, Z, Sathe, N, et al. A systematic review of medical treatments for children with autism spectrum disorders. Pediatrics. 2011;127(5):e1312e1321. doi:10.1542/peds.2011-0427CrossRefGoogle ScholarPubMed
Wilens, TE, Bukstein, O, Brams, M, et al. A controlled trial of extended-release guanfacine and psychostimulants for attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2012;51(1):7485.e2. doi:10.1016/j.jaac.2011.10.012CrossRefGoogle ScholarPubMed
Kollins, SH, Jain, R, Brams, M, et al. Clonidine extended-release tablets as add-on therapy to psychostimulants in children and adolescents with ADHD. Pediatrics. 2011;127(6):e1406e1413. doi:10.1542/peds.2010-1260CrossRefGoogle ScholarPubMed
Aman, MG, Bukstein, OG, Gadow, KD, et al. What does risperidone add to parent training and stimulant for severe aggression in child attention-deficit/hyperactivity disorder? J Am Acad Child Adolesc Psychiatry. 2014;53(1):4760.e1. doi:10.1016/j.jaac.2013.09.022CrossRefGoogle ScholarPubMed
De Giorgi, R, De Crescenzo, F, D’Alò, GL, et al. Prevalence of non-affective psychoses in individuals with autism spectrum disorders: a systematic review. J Clin Med. 2019;8(9):1304. doi:10.3390/jcm8091304CrossRefGoogle ScholarPubMed
Bakken, TL, Friis, S, Lovoll, S, Smeby, NA, Martinsen, H. Behavioral disorganization as an indicator of psychosis in adults with intellectual disability and autism. Ment Health Asp Dev Disabil. 2007;10(2):3747.Google Scholar
Foss-Feig, JH, Velthorst, E, Smith, L, et al. Clinical profiles and conversion rates among young individuals with autism spectrum disorder who present to clinical high risk for psychosis services. J Am Acad Child Adolesc Psychiatry. 2019;58(6):582588. doi:10.1016/j.jaac.2018.09.446CrossRefGoogle ScholarPubMed
Saitow, F, Takumi, T, Suzuki, H. Change in serotonergic modulation contributes to the synaptic imbalance of neuronal circuit at the prefrontal cortex in the 15q11-13 duplication mouse model of autism. Neuropharmacology. 2020;165:107931. doi:10.1016/j.neuropharm.2019.107931CrossRefGoogle Scholar
Sugie, Y, Sugie, H, Fukuda, T, et al. Clinical efficacy of fluvoxamine and functional polymorphism in a serotonin transporter gene on childhood autism. J Autism Dev Disord. 2005;35(3):377385. doi:10.1007/s10803-005-3305-2CrossRefGoogle Scholar
Najjar, F, Owley, T, Mosconi, MW, et al. Pharmacogenetic study of serotonin transporter and 5HT2A genotypes in autism. J Child Adolesc Psychopharmacol. 2015;25(6):467474. doi:10.1089/cap.2014.0158CrossRefGoogle ScholarPubMed
Owley, T, Brune, CW, Salt, J, et al. A pharmacogenetic study of escitalopram in autism spectrum disorders. Autism Res. 2010;3(1):17. doi:10.1002/aur.109CrossRefGoogle ScholarPubMed
King, BH. Fluoxetine and repetitive behaviors in children and adolescents with autism spectrum disorder. JAMA. 2019;322(16):15571558. doi:10.1001/jama.2019.11738CrossRefGoogle ScholarPubMed
Luft, MJ, Lamy, M, DelBello, MP, McNamara, RK, Strawn, JR. Antidepressant-induced activation in children and adolescents: risk, recognition and management. Curr Probl Pediatr Adolesc Health Care. 2018;48(2):5062. doi:10.1016/j.cppeds.2017.12.001CrossRefGoogle Scholar
Lambe, EK, Fillman, SG, Webster, MJ, Shannon Weickert, C. Serotonin receptor expression in human prefrontal cortex: balancing excitation and inhibition across postnatal development. PloS One. 2011;6(7):e22799. doi:10.1371/journal.pone.0022799CrossRefGoogle ScholarPubMed
Gupta, M, Gupta, N. FDA black box warning for SSRI: reexamining the role of high-functioning autism as a confounder. Adv Neurodev Disord. 2022;26. doi:10.1007/s41252-022-00301-6Google Scholar
de Silva, VA, Suraweera, C, Ratnatunga, SS, Dayabandara, M, Wanniarachchi, N, Hanwella, R. Metformin in prevention and treatment of antipsychotic induced weight gain: a systematic review and meta-analysis. BMC Psychiatry. 2016;16(1):341. doi:10.1186/s12888-016-1049-5CrossRefGoogle ScholarPubMed
Wang, L, Cai, Y, Fan, X. Metformin administration during early postnatal life rescues autistic-like behaviors in the BTBR T+ Itpr3tf/J mouse model of autism. Front Behav Neurosci. 2018;12:290. doi:10.3389/fnbeh.2018.00290CrossRefGoogle ScholarPubMed
Campbell, JM, Stephenson, MD, de Courten, B, Chapman, I, Bellman, SM, Aromataris, E. Metformin use associated with reduced risk of dementia in patients with diabetes: a systematic review and meta-analysis. J Alzheimers Dis. 2018;65(4):12251236. doi:10.3233/JAD-180263CrossRefGoogle ScholarPubMed
Aman, MG, Hollway, JA, Veenstra-VanderWeele, J, et al. Effects of metformin on spatial and verbal memory in children with ASD and overweight associated with atypical antipsychotic use. J Child Adolesc Psychopharmacol. 2018;28(4):266273. doi:10.1089/cap.2017.0072CrossRefGoogle ScholarPubMed
Protic, D, Aydin, EY, Tassone, F, Tan, MM, Hagerman, RJ, Schneider, A. Cognitive and behavioral improvement in adults with fragile X syndrome treated with metformin-two cases. Mol Genet Genomic Med. 2019;7(7):e00745. doi:10.1002/mgg3.745CrossRefGoogle ScholarPubMed
University of California, Davis. A Double-Blind, Placebo-Controlled Trial of Metformin in Individuals with Fragile X Syndrome. clinicaltrials.gov; 2022; https://clinicaltrials.gov/ct2/show/NCT03479476. Accessed March 13, 2022.Google Scholar
Beversdorf, DQ, Carpenter, AL, Miller, RF, Cios, JS, Hillier, A. Effect of propranolol on verbal problem solving in autism spectrum disorder. Neurocase. 2008;14(4):378383. doi:10.1080/13554790802368661CrossRefGoogle ScholarPubMed
Narayanan, A, White, CA, Saklayen, S, et al. Effect of propranolol on functional connectivity in autism spectrum disorder--a pilot study. Brain Imaging Behav. 2010;4(2):189197. doi:10.1007/s11682-010-9098-8CrossRefGoogle ScholarPubMed
Beversdorf, DQ, Saklayen, S, Higgins, KF, Bodner, KE, Kanne, SM, Christ, SE. Effect of propranolol on word fluency in autism. Cogn Behav Neurol. 2011;24(1):1117. doi:10.1097/WNN.0b013e318204d20eCrossRefGoogle ScholarPubMed
Bodner, KE, Beversdorf, DQ, Saklayen, SS, Christ, SE. Noradrenergic moderation of working memory impairments in adults with autism spectrum disorder. J Int Neuropsychol Soc. 2012;18(3):556564. doi:10.1017/S1355617712000070CrossRefGoogle ScholarPubMed
Zamzow, RM, Christ, SE, Saklayen, SS, et al. Effect of propranolol on facial scanning in autism spectrum disorder: a preliminary investigation. J Clin Exp Neuropsychol. 2014;36(4):431445. doi:10.1080/13803395.2014.904844CrossRefGoogle ScholarPubMed
Zamzow, RM, Ferguson, BJ, Stichter, JP, et al. Effects of propranolol on conversational reciprocity in autism spectrum disorder: a pilot, double-blind, single-dose psychopharmacological challenge study. Psychopharmacology. 2016;233(7):11711178. doi:10.1007/s00213-015-4199-0CrossRefGoogle ScholarPubMed
Zamzow, RM, Ferguson, BJ, Ragsdale, AS, Lewis, ML, Beversdorf, DQ. Effects of acute beta-adrenergic antagonism on verbal problem solving in autism spectrum disorder and exploration of treatment response markers. J Clin Exp Neuropsychol. 2017;39(6):596606. doi:10.1080/13803395.2016.1252724CrossRefGoogle ScholarPubMed
Hegarty, JP, Ferguson, BJ, Zamzow, RM, et al. Beta-adrenergic antagonism modulates functional connectivity in the default mode network of individuals with and without autism spectrum disorder. Brain Imaging Behav. 2017;11(5):12781289. doi:10.1007/s11682-016-9604-8CrossRefGoogle ScholarPubMed
London, EB, Yoo, JH, Fethke, ED, Zimmerman-Bier, B. The safety and effectiveness of high-dose propranolol as a treatment for challenging behaviors in individuals with autism spectrum disorders. J Clin Psychopharmacol. 2020;40(2):122129. doi:10.1097/JCP.0000000000001175CrossRefGoogle ScholarPubMed
Riikonen, R. Treatment of autistic spectrum disorder with insulin-like growth factors. Eur J Paediatr Neurol. 2016;20(6):816823. doi:10.1016/j.ejpn.2016.08.005CrossRefGoogle ScholarPubMed
John, S, Jaeggi, AV. Oxytocin levels tend to be lower in autistic children: a meta-analysis of 31 studies. Autism Int J Res Pract. 2021;25(8):21522161. doi:10.1177/13623613211034375CrossRefGoogle ScholarPubMed
Minakova, E, Lang, J, Medel-Matus, JS, et al. Melanotan-II reverses autistic features in a maternal immune activation mouse model of autism. PloS One. 2019;14(1):e0210389. doi:10.1371/journal.pone.0210389CrossRefGoogle Scholar
Horvath, A, Łukasik, J, Szajewska, H. ω-3 fatty acid supplementation does not affect autism spectrum disorder in children: a systematic review and meta-analysis. J Nutr. 2017;147(3):367376. doi:10.3945/jn.116.242354CrossRefGoogle Scholar
Demarquoy, C, Demarquoy, J. Autism and carnitine: a possible link. World J Biol Chem. 2019;10(1):716. doi:10.4331/wjbc.v10.i1.7CrossRefGoogle ScholarPubMed
Malaguarnera, M, Cauli, O. Effects of l-carnitine in patients with autism spectrum disorders: review of clinical studies. Mol. 2019;24(23):E4262. doi:10.3390/molecules24234262CrossRefGoogle ScholarPubMed
Naviaux, RK, Curtis, B, Li, K, et al. Low-dose suramin in autism spectrum disorder: a small, phase I/II, randomized clinical trial. Ann Clin Transl Neurol. 2017;4(7):491505. doi:10.1002/acn3.424CrossRefGoogle ScholarPubMed
Dean, OM, Gray, KM, Villagonzalo, KA, et al. A randomised, double blind, placebo-controlled trial of a fixed dose of N-acetyl cysteine in children with autistic disorder. Aust N Z J Psychiatry. 2017;51(3):241249. doi:10.1177/0004867416652735CrossRefGoogle ScholarPubMed
Millward, C, Ferriter, M, Calver, SJ, Connell‐Jones, GG. Gluten‐ and casein‐free diets for autistic spectrum disorder. Cochrane Database Syst Rev. 2008;CD003498. doi:10.1002/14651858.CD003498.pub3Google ScholarPubMed
Piwowarczyk, A, Horvath, A, Łukasik, J, Pisula, E, Szajewska, H. Gluten- and casein-free diet and autism spectrum disorders in children: a systematic review. Eur J Nutr. 2018;57(2):433440. doi:10.1007/s00394-017-1483-2CrossRefGoogle ScholarPubMed
Quan, L, Xu, X, Cui, Y, et al. A systematic review and meta-analysis of the benefits of a gluten-free diet and/or casein-free diet for children with autism spectrum disorder. Nutr Rev. 2021;7:nuab073. doi:10.1093/nutrit/nuab073Google Scholar
McGuinness, G, Kim, Y. Sulforaphane treatment for autism spectrum disorder: a systematic review. EXCLI J. 2020;19:892903. doi:10.17179/excli2020-2487Google ScholarPubMed
Hellings, JA, Reed, G, Cain, SE, et al. Loxapine add-on for adolescents and adults with autism spectrum disorders and irritability. J Child Adolesc Psychopharmacol. 2015;25(2):150159. doi:10.1089/cap.2014.0003CrossRefGoogle ScholarPubMed
Li, Z, Ichikawa, J, Meltzer, HY. A comparison of the effects of loxapine with ziprasidone and thioridazine on the release of dopamine and acetylcholine in the prefrontal cortex and nucleus accumbens. Psychopharmacology. 2003;167(3):315323. doi:10.1007/s00213-003-1418-xCrossRefGoogle ScholarPubMed
Bhatti, I, Thome, A, Smith, PO, et al. A retrospective study of amitriptyline in youth with autism spectrum disorders. J Autism Dev Disord. 2013;43(5):10171027. doi:10.1007/s10803-012-1647-0CrossRefGoogle ScholarPubMed
Hellings, JA, Arnold, LE, Han, JC. Dopamine antagonists for treatment resistance in autism spectrum disorders: review and focus on BDNF stimulators loxapine and amitriptyline. Expert Opin Pharmacother. 2017;18(6):581588. doi:10.1080/14656566.2017.1308483CrossRefGoogle ScholarPubMed
Chez, MG, Burton, Q, Dowling, T, Chang, M, Khanna, P, Kramer, C. Memantine as adjunctive therapy in children diagnosed with autistic spectrum disorders: an observation of initial clinical response and maintenance tolerability. J Child Neurol. 2007;22(5):574579. doi:10.1177/0883073807302611CrossRefGoogle ScholarPubMed
Ghaleiha, A, Asadabadi, M, Mohammadi, MR, et al. Memantine as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Int J Neuropsychopharmacol. 2013;16(4):783789. doi:10.1017/S1461145712000880CrossRefGoogle ScholarPubMed
Aman, MG, Findling, RL, Hardan, AY, et al. Safety and efficacy of memantine in children with autism: randomized, placebo-controlled study and open-label extension. J Child Adolesc Psychopharmacol. 2017;27(5):403412. doi:10.1089/cap.2015.0146CrossRefGoogle ScholarPubMed
Hardan, AY, Hendren, RL, Aman, MG, et al. Efficacy and safety of memantine in children with autism spectrum disorder: results from three phase 2 multicenter studies. Autism Int J Res Pract. 2019;23(8):20962111. doi:10.1177/1362361318824103CrossRefGoogle ScholarPubMed
Joshi, G, Wozniak, J, Faraone, SV, et al. A prospective open-label trial of memantine hydrochloride for the treatment of social deficits in intellectually capable adults with autism spectrum disorder. J Clin Psychopharmacol. 2016;36(3):262271. doi:10.1097/JCP.0000000000000499CrossRefGoogle ScholarPubMed
Soorya, LV, Fogg, L, Ocampo, E, et al. Neurocognitive outcomes from memantine: a pilot, double-blind, placebo-controlled trial in children with autism spectrum disorder. J Child Adolesc Psychopharmacol. 2021;31(7):475484. doi:10.1089/cap.2021.0010CrossRefGoogle ScholarPubMed
King, BH, Wright, DM, Handen, BL, et al. Double-blind, placebo-controlled study of amantadine hydrochloride in the treatment of children with autistic disorder. J Am Acad Child Adolesc Psychiatry. 2001;40(6):658665. doi:10.1097/00004583-200106000-00010CrossRefGoogle ScholarPubMed
Mohammadi, MR, Yadegari, N, Hassanzadeh, E, et al. Double-blind, placebo-controlled trial of risperidone plus amantadine in children with autism: a 10-week randomized study. Clin Neuropharmacol. 2013;36(6):179184. doi:10.1097/WNF.0b013e3182a9339dCrossRefGoogle ScholarPubMed
Morrow, K, Choi, S, Young, K, Haidar, M, Boduch, C, Bourgeois, JA. Amantadine for the treatment of childhood and adolescent psychiatric symptoms. Proc Bayl Univ Med Cent. 34(5):566570. doi:10.1080/08998280.2021.1925827CrossRefGoogle Scholar
Figure 0

Table 1. Key Studies Included in the Review

Figure 1

Figure 1. A flowchart of a complex clinical pathway for diagnostics of ASD and assessment of co-occurring conditions. (a) Autism is a clinical diagnosis that incorporates levels of dysfunction and support needed in various contexts. (b) Testing is an aid to clinical diagnosis but is not necessary. ADOS-2 is an excellent test but is far from being the gold standard. (c) Testing should incorporate various elements including intellectual functioning, adaptive functioning, ADOS-2, ADI-R, and others. (d) ADOS-2 alone may only provide a 60-minute snapshot of symptoms and supplemental detailed parent interviews like ADI-R are helpful in improving sensitivity and specificity. (e) Given the high rates of false negatives, negative testing should be interpreted based on context and dysfunction. (f) In case of negative testing, consider diagnostic overshadowing or “Multiple Complex Developmental Disorder” which could lead to masking of symptoms, especially in HFA individuals. (g) If testing continued to be negative but clinical symptoms are suggestive of autism, consider diagnosing ASD. (h) Aggression in ASD can be complex and multimodal interventions have better outcomes. Interventions include considerations of sensory and food insensitivities along with ruling out any medical causes like constipation. (i) Although a linear dose–response was reported with MPH for higher-functioning ASD youths; however, experts suggest using caution with dose titration in lower-functioning ASD youths to avoid undesired side effects including behavioral activation. (j) Slow-dose titration of SSRI is recommended due to the risk of behavioral activation. There is a high risk of partial response and no response due to poor metabolism and precision medicine is recommended. (k) Could be secondary to sensory insensitivities or medical reasons.

Figure 2

Figure 2. Overarching principles and multimodal interventions to mitigate impairments associated with ASD.