A boy looking down at colored wooden blocks.

  • Copy/Paste Link Link Copied

Autism Research Activities and Advances

NICHD's autism spectrum disorder (ASD) research portfolio is spread throughout the Institute and includes extramural components that support research on ASD and other  intellectual and developmental disabilities (IDDs) . NICHD also conducts some autism-related research projects through its intramural program.

In addition, several extramural and intramural entities within NICHD sponsor or conduct research that is not autism-focused but that can inform our understanding of the developmental and molecular processes involved in autism pathophysiology. Some of these efforts are described below.

Institute Activities and Advances

Recent findings.

  • A longitudinal study of parent-reported sensory responsiveness in toddlers at-risk for autism (PMID: 30350375 )
  • Potential for digital behavioral measurement tools to transform the detection and diagnosis of autism spectrum disorder (PMID: 30715131 )
  • Restricted and repetitive behavior and brain functional connectivity in infants at risk for developing autism spectrum disorder (PMID: 30446435 )
  • Language delay aggregates in toddler siblings of children with autism spectrum disorder (PMID: 30348077 )
  • Automatic emotion and attention analysis of young children at home: a ResearchKit autism feasibility study  
  • Parent support of preschool peer relationships in younger siblings of children with autism spectrum disorder (PMID: 28634707 )
  • Walking, gross motor development, and brain functional connectivity in infants and toddlers (PMID: 29186388 )

As one of the participants in the government-wide Interagency Autism Coordinating Committee (IACC) , NICHD's support for autism research is structured around the seven question areas of IACC's strategic plan for autism research :

  • Question 1: When Should I Be Concerned?
  • Question 2: How Can I Understand What Is Happening?
  • Question 3: What Caused This to Happen and Can It Be Prevented?
  • Question 4: Which Treatments and Interventions Will Help?
  • Question 5: Where Can I Turn for Services?
  • Question 6: What Does the Future Hold, Particularly for Adults?
  • Question 7: What Other Infrastructure and Surveillance Needs Must Be Met?

NICHD supports and conducts research in all seven areas, with particular support for research relevant to questions 1 and 2.

Much of NICHD's autism research is conducted through the trans-NIH Autism Centers of Excellence (ACE) Program . The ACE project, established in 2007, was a consolidation of two previous research efforts—the NICHD-led Collaborative Programs of Excellence in Autism and the Studies to Advance Autism Research and Treatment . ACE was intended to better coordinate autism research across the NIH.

IACC Question 1: Diagnosis of ASD

NICHD-supported research related to IACC Question 1 aims to develop and improve screening and diagnostic tools for ASD. The  Intellectual and Developmental Disabilities Branch (IDDB)  supports extramural research exploring ways to validate and improve screening and diagnosis tools for ASD, such as the Modified Checklist for Autism in Toddlers (M-CHAT), an effective screening tool for children aged 16 months to 2½ years. The Branch also supports the development of new screening tools, especially those for children younger than age 24 months, and the development of instruments for assessing symptoms and daily function of people with ASD.

The IDDB also supports studies that may inform the development of new screening tools in the future. IDDB-funded research tracks the anatomical, functional, emotional, communicative, and behavioral characteristics of infants at high risk for ASD over time in order to develop and improve the long-term accuracy of diagnostic and prognostic tools for ASD. The Branch also supports systematic efforts to identify genetic variants associated with autism, with the eventual goal of developing a new early diagnosis and classification system. IDDB-supported research studies also address the development of the linguistic and sensory symptoms of ASD throughout childhood, which may also inform screening tools.

  • IDDB-supported findings: Researchers worked with health care providers to screen more than 15,000 low-risk toddlers using an updated version of MCHAT, the Modified Checklist for Autism in Toddlers—Revised, with Follow-Up (M-CHAT–R/F) and found it to be more accurate than earlier versions at identifying children who could benefit from further evaluation . ( PMID: 24366990 )

The IDDB's research support is complemented by support from the Child Development and Behavior Branch (CDBB) for research on the processes of normal development. Data on the development of joint attention, social orientation, and emotional function and communications provide important benchmarks for understanding how early deficits in these skills develop in ASD.

NICHD's intramural scientists also conduct research relevant to this IACC question. Through its Epidemiology Branch , within the Division of Population Health Research (DiPHR) , the Institute is active in the assessment of the M-CHAT for ASD and other developmental screening algorithms. The DiPHR has also conducted research on the patterns of growth, physical development, and hormone levels throughout childhood in autism.

IACC Question 2: Biology of ASD

Several extramural branches of NICHD support research on disorders of neurologic and behavioral development, such as autism, by characterizing the developmental processes, cognitive processes, sensory and motor systems, and molecular and neural mechanisms that are relevant in the biology of the condition and its symptoms.

For instance, the IDDB supports research on the biology of ASD, including studies of the developmental processes underlying ASD biology throughout childhood. This research aims to characterize the cognitive and sensory/motor deficits in ASD, such as difficulties in recognizing emotion in faces and speech and the dysfunction in perceiving time or the differences between sounds. The Branch also supports research on the molecular and neurological underpinnings of ASD in humans as well as in model organisms. Funded research also delineates the function of genes and risk for ASD in brain development and function and maps the altered biochemical pathways and neural networks in brains of people with ASD to determine how these biological characteristics are correlated with behaviors or symptoms.

The IDDB is also interested in research on the biological processes that ASD has in common between ASD with comorbid or causative genetic conditions, such as Fragile X syndrome , t syndrome , Angelman syndrome, and Prader-Willi syndrome . The Branch also funds research to find or characterize subtypes of autism, by identifying new genes related to ASD risk and correlating known risk genes with brain structure and function and symptoms.

In addition, the CDBB's  Developmental Cognitive Psychology, Behavioral Neuroscience, and Psychobiology Program funds studies to identify and characterize the pathways involved in brain development and behavior, including those in the sensory, motor, linguistic, cognitive, and social behavioral domains, all of which are disrupted in ASD. The Branch's studies of typically developing children serve as an important benchmark for understanding the differences found in children with ASD.

The  /about/org/der/branches/dbcab  also supports research on normal and abnormal development relating to the causes and prevention of congenital and genetic defects, as well as research training in relevant academic and medical areas, with an emphasis on the biochemical, genetic, and cellular mechanisms of early development that can be disrupted in disorders like ASD.

The  Section on Cellular and Synaptic Physiology , within the Division of Intramural Research (DIR)   Neurosciences Affinity Group , focuses on the development and regulation of synapses in the cortex and hippocampus. Networks in these areas are disrupted in ASD and other brain disorders.

IACC Question 3: Causes and Preventions of ASD

The IDDB is a major supporter of human and animal studies on the causes of ASD, including investigation of the processes and pathways associated with ASD, autism symptoms, common co-morbidities, and protective factors for ASD. One large area of IDDB support is genetics and epigenetics. The Branch funds studies of the identification, expression, regulation, and interactions of gene variants linked to ASD and autism-related behaviors and symptoms. The IDDB also supports research on potential environmental risk factors and biomarkers for ASD, including gene-environment interactions.

In addition, two laboratories within the DIR conduct research relevant to the biology of ASD:

  • The Section on Molecular y conducts research on a potential new endophenotype of ASD related to hypocholesterolemia.
  • The Section on Clinical Genomics uses a cell-culture model to study neuronal networks in autism. Its research also examines the expression of non-coding RNA in the brain in autism.

IACC Question 4: Interventions for ASD

The IDDB supports research on the development and evaluation of therapies and treatments for ASD, ASD symptoms, and related disorders, such as Fragile X syndrome , as well as the long-term effects of these interventions. Potential treatment targets include repetitive behavior, joint attention, social skills, emotional sharing, symbolic understanding, language and communication, irritability and anxiety, and insistence on sameness. Researchers working in human subjects and animal models consider a range of treatment types, from behavioral and educational interventions to pharmaceutical treatments, including comprehensive treatments that combine behavior and medication.

  • IDDB-supported findings: A recent ACE network study found that directing the attention of preschool-aged children with ASD increased the children's vocabularies and language skills by the time they were age 8, compared to a control. In the intervention, adults actively engaged the children's attention by pointing to toys and using other gestures.

IACC Question 5: Services for People with ASD

As a research agency, NICHD focuses its efforts on evaluating services—how they are delivered or how effective they are, for example—rather than on providing services. For instance, the IDDB supports a few studies of methods to develop or improve services for people with ASD, including services related to teaching life skills and ensuring physical safety of people with ASD.

IACC Question 6: Health Over the Lifespan with ASD

Most NICHD research addresses the early biological origins of ASD, meaning that efforts related to this question are handled by other agencies. However, through the IDDB, NICHD supports one study related to this question, focused on teaching social skills to adolescents with high-functioning ASD.

IACC Question 7: Infrastructure for ASD Research

Much of the Institute's work within this area is related to support of the ACE program. In 2012, NIH awarded $100 million to continue support of the program. The Institute also supports other projects related to ASD research infrastructure, including the National Database for Autism Research, Brain and Tissue Bank, and NeuroBioBank resources that are described in the Other Activities and Advances section below.

Other Activities and Advances

To achieve its goals for autism research, NICHD supports a variety of other activities related to autism. Some of these activities are managed through the components listed above; others are part of NIH-wide or collaborative efforts in which NICHD participates. Some of these are listed below:

  • The  Autism Centers of Excellence (ACE) Program  is the trans-NIH research effort on ASD.
  • The Collaborative Programs of Excellence in Autism (CPEAs)/Studies to Advance Autism Research & Treatment (STAART) Centers conducted and supported studies on the causes, diagnosis, prevention, detection, and treatment of ASD. These Networks were consolidated in 2007 into the ACE Program to enable pooling of resources and maximum coordination and efficiency for autism research across the NIH.
  • NICHD's Eunice Kennedy Shriver Intellectual and Developmental Disabilities Research Centers are located at 15 universities and children's hospitals throughout the country and aim to advance understanding of a variety of conditions and topics related to IDDs.
  • The Fragile X Syndrome Research Center Program , funded by the IDDB, supports research to improve the diagnosis and treatment of Fragile X syndrome and related conditions.
  • The government-wide  Interagency Autism Coordinating Committee (IACC)  includes representatives from NICHD.
  • The  National Database for Autism Research  includes relevant data at all levels of biological and behavioral organization (i.e., molecules, genes, neural tissues, social and environmental interactions) and for all data types (e.g., text, numeric, image, time series).
  • The NIH NeuroBio Bank is a network of brain and tissue banks in the United States that collect, examine, and store tissues; the banks also make the tissues available to scientists for research on brain disorders.

External Web Site Policy

College of Education and Human Development

Department of Educational Psychology

Research topics: Autism

Identifying, preventing, and developing interventions related to autism spectrum disorder.

Autism spectrum disorder (ASD) and autism are both general terms for a group of complex disorders of brain development. These disorders are characterized, in varying degrees, by difficulties in social interaction, verbal and nonverbal communication and repetitive behaviors. Research in the Department of Educational Psychology focuses on early identification, prevention measures, and interventions related to ASD.

LeAnne Johnson

Johnson (special education) researches interventions to improve outcomes for a range of preschool and elementary school-aged children who are at high risk given social, emotional, behavioral, and communication needs. Johnson is focused on creating the next generation of intervention studies that support high fidelity implementation of evidence-based interventions within tiered intervention and prevention models. This includes research projects that are designed to test the efficacy of social-communication interventions for children with autism.

Jason Wolff

Wolff (special education) runs a lab funded funded in-part by the National Institute of Mental Health with two goals -- to leverage brain imaging data to characterize factors associated with the early emergence of behavioral excesses and deficits in autism spectrum disorder, and to identify potential neurodevelopmental moderators of response to early intervention. The ultimate goal of this work is to determine how brain and behavioral data may be used to inform the timing and content of early or even preventative interventions.

Panayiota Kendeou

Kendeou (psychological foundations of education) investigates how people learn new knowledge and revise pre-existing incorrect knowledge or misinformation during their reading experiences. She is currently investigating how misinformation that resists correction influences reasoning and decision making in health issues pertaining to ASD (e.g., reliance on ineffective treatments, withholding vaccinations), and explore ways for effective revision.

M.Y. Savana Bak

Bak's research focuses on measurement and analysis of language in children with ASD using language samples collected from the children’s natural environment. She strives to develop practical interventions and identify environmental factors that facilitate language development and increase social interaction in children with ASD.

Related degrees

Phd in special education.

Interested in conducting research in autism spectrum disorder? Learn more about earning your doctorate in special education .

Related labs and projects

  • ALAB: A Lab for Autism Research
  • Reading + Learning Lab
  • Research lab: Jason Wolff
  • See us on facebook
  • See us on instagram
  • See us on twitter

ADDRP

Autism and Developmental Disorders Research Program

Welcome to the website of the  Autism and Developmental Disorders Research Program (ADDRP) , Lucile Packard Children's Hospital at Stanford University.  This Stanford autism research program is based in the  Department of Psychiatry and Behavioral Sciences  at the Stanford University School of Medicine.

ADDRP focuses on improving the quality of life of individuals with autism spectrum disorder and/or intellectual disabilities. Through research methods that range from clinical trials, neuroimaging investigations, behavioral analysis to basic science methods, the researchers at ADDRP are committed to developing effective treatment strategies and identifying the causes of these conditions.

Our main research aim is to better understand the basic neurobiology of autism and other developmental disorders while furthering our understanding of how genetic and environmental factors may contribute to the onset and progression of these disorders. With this aim in mind, we conduct a variety of research studies and clinical trials of novel behavioral and biological therapies in hopes of developing effective interventions for the treatment of core features of these disorders.

Acknowledgements

The Stanford Autism and Developmental Disorders Research Program would like to thank the children, as well as their parents and families, for contributing to research. The joint effort to better understand and provide therapies for developmental disorders is not possible without their past and continued involvement.

Stanford ADDRP would also like to ackowledge financial support from the following organizations:

  • National Institutes of Health
  • Autism Speaks
  • Simons Foundation
  • John and Marcia Goldman Foundation
  • Stanford Bio-X
  • Child Health Research Institute
  • The Teresa and Charles Michael Endowed Fund for Autism Research and Education
  • The Mosbacher Family Fund for Autism Research
  • PTEN Research Foundation
  • The Bernard/Fung Family Fund for Autism Research at Stanford

In the News

2/2/22  Stanford Team Finds Benefits to Online Autism Treatment

7/16/21  Program improves resilience for parents of kids with autism

8/6/19 Stanford Trial Shows Parents Can Learn Therapy to Help Their Children With Autism Learn to Speak

8/5/19 One therapy bests others at motivating kids with autism to speak

5/1/19  Hormone reduces social impairment in kids with autism

3/6/19  Nature versus nurture in autism

2/2/18: Mechanical forces being studied by Stanford researchers may underlie brain's development and some diseases

7/12/17: Oxytocin improves social abilities in some kids with autism

4/10/17: Autism researchers seek teens, young adults for drug trial

9/21/2016: The seekers: Why parents try fringe therapies for autism

8/16/2016:  Automating genetic analysis helps keep up with rapid discovery of new diseases

7/22/2015 : Low levels of hormone linked to social deficit in autism

10/27/2014 : Group classes teach parents effective autism therapy, study finds

8/4/2014 : Blood-oxytocin levels in normal range in children with autism, study finds

11/14/2013 : Stanford drug trial seeks participants with autism spectrum disorder

8/13/2012 : Stanford researchers investigate the emotional side of autism

5/29/2012 : Antioxidant Shows Promise as Treatment for Certain Features of Autism, Study Finds (reprinted in ScienceDaily)

Spring 2012 : Autism Answers - Parents run experiments to see what works

9/2/2011 : Spotting autism's unique shape in the brain

7/30/2011 : Autism Risks: Genes May Not Play Biggest Role

1/25/2010 : Stanford/Packard autism researchers seek twins for brain-imaging study  

Related Pages & Events

Upcoming events.

16th Annual Pivotal Response Treatment (PRT) Conference  UPDATE: Conference has been postponed.

SAVE THE DATE! 

The Stanford Neurodiversity Summit

9/22/24-9/24/24 at the Li Ka Shing Conference Center, Stanford, CA

2024 Bay Area Adult Autism/DD Conference

12/7/24 at the Li Ka Shing Conference Center, Stanford, CA. Registration coming soon! 

18th Annual Autism Update

3/22/25  at Li Ka Shing Conference Center, Stanford,CA 

Autism Parent Support Group

Meets on second Mondays from 7-8:30pm (Sept.-June).  Please  email us  to be added to the our to the monthly email with participant zoom information.

  • Stanford Autism Center
  • Lucile Packard Children’s Hospital Stanford
  • Early Support Program for Autism
  • Child Psychiatry
  • Stanford Clinical Trials Directory
  • Center for Interdisciplinary Brain Sciences Research
  • Stanford Brain Development Project
  • Stanford Program for Psychometrics and Measurement-Based Care

autism research project ideas

  • About Autism
  • Is It Autism?
  • Starting Intervention
  • Prenatal Factors
  • Autism Assessment Tools
  • Diagnostic Checklist
  • Autism Treatment Evaluation Checklist (ATEC)
  • Understanding and Treating Self-Injurious Behavior Tool
  • Autism Support
  • Expert Webinars
  • Newly Diagnosed
  • Studies Seeking Participants
  • ARI-Funded Research Studies 2023
  • ARI Think Tanks
  • Participate in Studies
  • ARI-Funded Research By Year
  • Mission Statement
  • Board of Directors
  • Scientific Advisory Board
  • National Autism History Museum Hours
  • ARI's Latest Accomplishments
  • Annual Reports
  • Financials - Audit Reports/990s
  • Donate Cryptocurrency
  • Donate Stock/Mutual Funds

Autism Research Institute Logo

ARI-Funded Research Studies 2022

In 2022, ARI awarded more than $450,000 in grants to fund innovative research that holds realistic promise in impacting the lives of autistic people. At ARI we understand what it means to be different because ARI has always been unique. We have learned a lot over the past 55 years. We continue to focus on education while supporting research on genetics, neurology, co-occurring medical conditions, nutrition, sensory processing, severe and challenging behaviors, and adult and senior issues. Connecting investigators, professionals, parents, and those on the spectrum worldwide is essential for effective advocacy. In order to provide parents and professionals with an independent, unbiased assessment of causal and treatment efficacy issues, ARI seeks no financial support from government agencies or drug manufacturers.

More Research Studies

Mirna as a potential mediator in maternal/fetal interaction in neurodevelopmental disorders.

David Beversdorf, MD University of Missouri

A significant increase in prenatal stressors is observed in late in pregnancies in children subsequently diagnosed with autism. In animal models, atypical social behavior is observed in offspring of rodent mothers exposed to stress during pregnancy, associated with epigenetic changes in the offspring brains. Prenatal stress in cases of clinical autism is associated with maternal stress susceptibility genes, and also associated with epigenetic changes in the maternal blood, overlapping with epigenetic changes observed in mice. We now wish to determine the mechanistic role of the micro-RNA (miRNA, small pieces of RNA with major regulatory effects) in the neurodevelopmental changes associated with prenatal stress. Our pilot data revealed the epigenetic changes in maternal blood in the prenatal stress mouse model, identifying the miRNA profile at the time of pregnancy. We now wish to determine whether the miRNA associated with prenatal stress, administered to pregnant mice, recapitulate behavioral (Aim 1), neurochemical (Aim 2), and other epigenetic effects (Aim 3) of prenatal stress exposure, to begin to understand the mechanism. This will allow better understanding of mediators of the effect of stress on development, identifying multiple points of intervention for a novel mechanism in the clinical setting for a subset of cases of autism.

Impact of environmental enrichment on the resolution of inflammation in a mouse model of Autism Spectrum Disorder

Paola Bonsi, PhD Fondazione Santa Lucia

Neuroinflammation and aberrant activation of microglia, the brain immune cells, can drive derangements in the development and function of the brain, and are thought to be involved in the pathogenesis of diverse neurological conditions, among which Autism Spectrum Disorder (ASD). In recent years, neuroinflammation is emerging as a tightly regulated bidirectional process, involving both pro-inflammatory and pro-resolution (anti-inflammatory) signaling pathways. Dysregulation of these mechanisms may therefore underlie the persistence of a neuroinflammatory state, leading to pathological manifestations. Clinical, genetic, and experimental evidence indicates structural and functional alterations in a brain region called striatum in both individuals with autism and ASD mouse models. Our preliminary findings indicate that neuronal abnormalities in the striatum of an ASD mouse model showing autistic-like behaviors are accompanied by microglia-associated neuroinflammation and reduction of the pro-resolution activity. The goal of our project is to find out whether boosting the physiological process of resolution of neuroinflammation is able to revert the neuronal and behavioral dysfunctions observed in the ASD mouse model. To this aim, we will utilize an innovative approach based on a non-pharmacological intervention providing social, cognitive and motor stimulation, defined “environmental enrichment.” The results of this study will provide crucial information on the role of alterations in the pro-inflammatory and pro-resolution pathways in ASD pathophysiology, and on the capability of the multimodal stimulation provided by environmental enrichment to revert such alterations and the ensuing neuronal and behavioral dysfunction.

Understanding Role of Peroxynitrite Signaling and Developing Therapeutic Intervention for Autism Spectrum Disorder

Adrien Eshraghi, MD, MSc, FACS University of Miami Hearing Research and Communication Laboratory

Despite advances in medical field, there are still no effective medical treatments available for autism spectrum disorder (ASD) that can be attributed to the incomplete understanding about the pathophysiology of this neurological disorder. Intriguingly, we found the new avenues of developing effective therapeutic modalities for ASD from an entirely novel approach based on targeting protein tyrosine (Tyr) nitration by the oxidant peroxynitrite. The objective of this pilot study is to understand the role of peroxynitrite signaling in the pathophysiology of ASD. We will determine whether targeting peroxynitrite signaling will help in rescuing ASD associated behavior and social deficits in our preclinical animal model of ASD. This study will provide a novel catalogue of candidates that can be targeted to develop effective therapeutic interventions for ASD based on deep understanding regarding the role of peroxynitrite signaling in the molecular underpinnings of autism.

Placental Trophoblast Inclusions as Autism Risk Markers Among Preterm Infants

William Fifer, PhD, Principal Investigator Morgan Firestein, PhD, Co-Investigator Columbia University Irving Medical Center

The rising prevalence of autism spectrum disorder (ASD) and the effectiveness of early intervention places critical emphasis on the need to identify early emerging biological markers of risk. Trophoblast inclusions (TIs) in the placenta are histologically visible abnormalities within chorionic villi resulting from excessive cell proliferation of the inner cytotrophoblast layer. TIs are observed in only 2-8% of placentas from full-term uncomplicated pregnancies, however, a 3-fold increase in the number of TIs in placentas of children with ASD has been reported and TIs in placentas from children with familial risk for ASD were five times more prevalent. Moreover, TIs have been observed in 30-50% of placentas from preterm infants, a population at increased risk for ASD. Little is known about biological mechanisms underlying the association between preterm birth and increased risk for ASD and few tools reliably identify which infants are at greatest risk for ASD, potentially impeding earlier diagnosis and access to early intervention. The proposed research aims to determine if TIs are more prevalent in placentas of prematurely born infants determined to be at-risk for ASD at 18 months of age compared to prematurely born infants at low risk.

Zinc nutrition as key regulator of inflammatory gut-brain signaling in Autism

Andreas Grabrucker, PhD University of Limerick 

The microbiota-gut-brain axis and its role in healthy brain development and function have recently moved into the focus of research aiming at understanding the etiology of disorders of the Central Nervous System, including Autism Spectrum Disorders (ASDs). Increased inflammation due to a pathology in the gastrointestinal (GI) tract is considered a major contributor to abnormal gut-brain signaling. Accordingly, altered microbiota profiles, impaired intestinal barrier tightness, and the resulting activation of pro-inflammatory pathways have been reported in ASDs.

To better understand cause and consequence relationships, we will uncouple the effects of microbiota and inflammation on GI physiology, using 3D stem cell-derived intestinal organoids. Using these model systems, we can either induce bacterial components, or induce inflammation, or both. Moreover, we can investigate whether the presence of probiotic bacterial components or nutritional factors discussed in ASD prevent or normalize an observed pathology. For example, based on previous results we hypothesize that the pro-inflammatory processes in the GI tract are zinc-dependent. Our results will reveal whether zinc supplementation may be beneficial in conditions with compromised GI barrier tightness such as ASD and explore novel zinc supplements that we have characterized in the past.

Discovery of ASD related pathways in a novel preterm Rhesus macaque maternal immune activation model

Suhas Kallapur, MD University of California Los Angeles

Epidemiological data demonstrate a strong association between intrauterine infection/inflammation (IUI) causing preterm birth (PTB) and neurodevelopmental/neurobehavioral defects during infancy. IUI, especially in the 3rd trimester, is an understudied maternal immune activation in ASD research but is important because recent data show an association between IUI and prematurity with ASD. We have modeled IUI in Rhesus macaques by giving intraamniotic (IA) live E. coli injection followed 24h later by antibiotics (E. coli + Abx) that results in neuroinflammation and preterm birth. Recently we demonstrated that maternal anti-IL1 (IL1 receptor antagonist, Anakinra) therapy significantly decreases IA E. coli + Abx-induced IUI. To better understand the link between neuroinflammation caused by IUI and ASD, we will comprehensively determine snRNA-seq changes in different brain regions of the fetal Rhesus macaque brain exposed to IA E. coli in the acute (24h-72h) phase of neuroinflammation. We will determine if maternal anti-inflammatory therapy with Anakinra can reverse neuroinflammation and reverse ASD related gene expression changes in different brain regions. This fetal brain transcriptomic study will reveal mechanistic insights in the pathogenesis of ASD.

Rescue of a neurodevelopmental disorder associated with ASD by induction of the heat shock response

Andrew Levy, PhD Technion Israel Institute of Technology

Fever has been associated with the abatement of many of the social behavioral abnormalities seen in autism spectrum disorder. However, the lack of mechanistic studies exploring the mechanism for fever protection, due in large part to the lack of appropriate human disease models, has limited interest and use of this mode of therapy. A mutation in the IQSEC2 gene (A350V) was found to cause drug resistant epilepsy, autism spectrum disorder and intellectual disability. Fever in a child with this mutation has been associated with a cessation of seizures and improved social interactions [4]. Our working hypothesis, which we will test in this grant proposal, is that fever induces heat shock proteins which directly act to reduce activated Arf6. The specific aims of this proposal are based on published data underlying the key role of Arf6 activation in the pathophysiology of IQSEC2 mutations [8-10] and other ASD associated mutations [6] and our recent discovery that Arf6 activation is dramatically downregulated by the heat shock response. The long-term goals of this research are to determine the basic mechanism underlying the benefit from fever seen with A350V and other IQSEC2 mutations as well as in children with other ASD associated mutations, and to translate this into a treatment which will enhance social interactions, reduce the seizure burden and overall improve the quality of life of children afflicted with these disorders.

Characterizing Auditory Sensory Stability in Autism

Adam Naples, PhD Yale University

Up to seventy percent of autistic people experience sensitivity to sounds. Autistic adults report that these symptoms worsen with stress and anxiety and can interfere with school, work, and other activities. However, despite the common report of these symptoms, there is no understanding of the mechanisms, nor are there effective ways to measure these symptoms.

Importantly, most measurement of these symptoms in autism relies on retrospective questionnaires. These measures require participants to “average” their symptoms over some time period in the past, possibly their entire lives. Such measures are well known to have “peak and end” biases in which people recall the most memorable and distressing experiences and the experiences that were most recent. This means that these questionnaires are not able to accurately capture the day-to-day lived experience of people with autism.

In this study we take the first step towards measuring the personal timing of auditory sensitivities, and their relationship with symptom report using an innovative approach. We measure auditory sensitivity using daily symptom self-reports and brief experimental auditory tasks delivered remotely over the internet. Participants will complete established self-report measures of sensory sensitivity and then will receive daily text-message or email reminders that will link to individualized questionnaires assessing sensory symptoms for that specific day. Additionally, participants will complete a brief tone detection task delivered via headphones on their computer or mobile device that will measure in-the-moment auditory perception.

The long-term goals of this study are to gain an understanding of the stability of auditory sensitivities to support subsequent mechanistic research. Currently there are no mechanistic biomarkers for auditory sensitivities in autism despite many successes in identifying group-level differences. Most research as assumed that auditory sensitivity symptoms are stable, over time, within an individual. However, if this assumption is invalid, then research that seeks to understand biological mechanisms will need to measure those symptoms at just the right time to find a brain-behavior linkage. This problem is exacerbated in autism because increased sensory sensitives are associated with avoidance of work and school. Consequently, autistic people may be less likely to participate in a research protocol on days when their symptoms are particularly distressing.

Autistic adults often report that these symptoms vary in intensity and frequency, however, there is no research that investigates if, how, or when these symptoms might vary. In this study, by determining how these sensitivities fluctuate over time, we gain a better understanding of the psychometric properties of auditory sensitivities, which provides insight into potential mechanisms. Furthermore, understanding the variability of symptom expression and auditory perception is critical information for developing and implementing successful in-person research studies.

Randomized placebo-controlled double-blind cross-over study of Coenzyme Q10, Vitamin E and polyvitamin B in a cohort of individuals with Autism Spectrum Disorder

Antonio M. Persico, M.D. University of Modena and Reggio-Emilia (Modena, Italy)

Mitochondrial dysfunction and enhanced oxidative stress have been consistently detected in Autism Spectrum Disorder (ASD). Randomized controlled trials (RCTs) involving cocktails with multiple antioxidants have provided promising results, but cannot identify truly active compounds and maximize their efficacy. We recently published a retrospective chart review of 59 patients with neurodevelopmental disorders, demonstrating that approximately 60% of ASD cases and higher percentages of patients with Intellectual Disability display small-to-moderate improvements with rare and manageable side effects. Moreover, an exploratory RCT involving 31 patients with Phelan-McDermid syndrome (PMS) was recently completed (NCT04312152). This retrospective chart review and exploratory RCT allow us now to perform a precisely-designed, targeted RCT of a “metabolic support therapy” including Coenzyme Q10+Vit.E+Vit.B in ASD. Each patient will receive Coenzyme Q10+Vit.E+ polyvit. B for 6 months and placebo for 6 months separated by a one-week wash-out period, according to a double-blind cross-over design. Primary outcome measure of efficacy will be the CGI-I; secondary outcome measures of autism signs/symptoms, adaptive behaviors and parental quality of life will include VAS, VABS and WHOQOL, respectively. Tolerability and adverse events will be monitored. The study will involve 50 ASD patients, 25 children 2-7 y.o and 25 adolescents 9-17 y.o. Behavior, neuropsychological testing, and multiple oxidative stress parameters will be assessed at T0, T1 (6 mo) and T2 (12 mo). This study uses the most sensitive measures of clinical and biochemical change, and should be sufficiently powered to conclusively support or disconfirm the prescription of this metabolic support therapy to children with ASD.

Testing cannabinoid type 2 receptor (CB2R) as a potential therapeutic target in ASD: a pilot study on a mouse model of maternal immune activation

Anna Maria Tartaglione, PhD Istituto Superiore di Sanità (ISS), Italy

Although Autism Spectrum Disorder (ASD) is identified by its social and communicative deficits, immune system impairments as well as signs of neuroinflammation have been frequently reported. The involvement of immune dysregulation in ASD is also supported by epidemiological evidence suggesting that maternal infection (and consequent maternal immune activation, MIA) increases the risk of ASD. In rodents MIA increases pro-inflammatory cytokines that cross the placenta causing long-lasting neuroinflammatory and behavioral responses in offspring, recapitulating hallmarks of ASD. However, mechanisms through which MIA leads to ASD-like phenotype are not yet fully understood. In this scenario, it is worthy of note that the endocannabinoid system (eCB) is a well-recognized modulator of both innate and adaptive immune responses, representing a bridge between the immune and central nervous systems. Interestingly, pharmacological activation of cannabinoid type 2 receptors (CB2R) recently gained attention, because of its ability to mitigate neuroinflammation in several neuropsychiatric disorders. Notwithstanding this evidence, the therapeutic potential of CB2R in ASD has not yet been investigated. To fill this gap, my research proposal aims to verify whether the CB2R stimulation (through JWH-133, a potent CB2R agonist) in prenatal and postnatal stages could prevent or revert the ASD-like behavioral phenotype and neuroinflammatory profile in mice prenatally exposed to MIA.

Home-based measurement of autonomic and endocrine system function and relations to sensory processing in children with and without autism

Jennifer Wagner, PhD CUNY College of Staten Island

Autism spectrum disorder (ASD) is characterized by social-communicative challenges and restricted interests and repetitive behaviors (RRB), and research has suggested that difficulty regulating stress might underlie some of these associated characteristics. In line with this, studies have examined two key stress systems in ASD, the autonomic nervous system (ANS) and the hypothalamic-pituitary-adrenal axis (HPAA), and results show that social communication and RRB can be predicted by these systems. No known studies, however, have looked at the interacting patterns of ANS and HPAA activity as they align with sensory processing difficulties that are common in ASD and also vary widely in the general population.  The current study will establish a novel protocol for home-based measurement of biomarkers related to the ANS and HPAA stress systems in children with and without ASD, utilizing a) pupillometry to collect markers of both parasympathetic and sympathetic autonomic function (through the constriction phase and the re-dilation phase of the pupil light reflex, respectively) and b) diurnal cortisol to examine HPAA axis integrity. These biomarkers will be examined alongside measures of autistic traits and sensory processing, and patterns of ANS and HPAA function will be used to explore variations in behavioral characteristics.

Is there a relationship between the gut mucosal-associated microbiome, intestinal neurotransmitters and behavior in individuals on the autism spectrum?

Harland Winter, MD Massachusetts General Hospital

The relationship between behavioral disturbances and gastrointestinal (GI) dysfunction in individuals on the autism spectrum is well established, but the mechanism is not well understood.  Intestinal microbiome imbalance (dysbiosis) has been described, but most studies have evaluated the microbiota in the stool.  Clinical observations have reported that fecal microbiota transplants improved both behavioral and GI phenotypes in individuals with ASD. Additionally, gut microbiome modification through antibiotic (vancomycin) treatment resulted in transient improvement in measured behavior and communication. Although these findings implicate gut microbiome imbalance with behavioral alterations in ASD, exactly which signals are involved and how they communicate along the brain-gut axis are poorly understood. Mechanistic pathways of communication are starting to emerge, with accumulating evidence from clinical studies and animal models that GABAeric signaling is often inhibited in the brain in individuals with ASD. However, a major knowledge gap exists in our understanding of whether GABA signals are similarly affected in the intestine of ASD individuals, and, if so, whether the gut microbiome contributes to neurotransmitter imbalance and behavioral alterations.

The aim of this proposal is to test the hypothesis that a novel gut-brain axis signal is functionally coupled to specific microbiome activity in the colonic mucosa, and that behavioral phenotypes in ASD individuals correlate with intestinal neurotransmitter levels. We will test this hypothesis by:

  • Characterizing the microbiome in paired colonic mucosa and stool of individuals who are on and not on the autism spectrum.
  • Measuring neurotransmitter levels in paired mucosa and stool to determine if imbalance is related to uptake from microbial-produced GABA in the gut.
  • Correlating the behavioral phenotype of all subjects with dysbiosis and neurotransmitter levels.

We expect this information will generate new knowledge about intestinal dysbiosis and the role of intestinal neurotransmitters in affecting behavior.

Privacy Overview

National Autism Center at May Institute Logo

Research Projects

The National Autism Center at May Institute engages in research to identify, evaluate, and disseminate effective practices to better support people with autism.

Students with Autism Accessing General Education (SAAGE)

This is a federally funded research project (U.S. Department of Education, R324A150032-16) focused on school-based interventions for students with autism spectrum disorder (ASD). The project is a multi-site project; work is being done in Massachusetts through the National Autism Center at May Institute, in New York through University of Rochester Medical Center, and in Florida through University of South Florida.

SAAGE is a modular intervention that is evidence-based but designed flexibly such that it is applicable for all students with ASD and can be implemented by educators in public schools with little to no additional resources. We are currently evaluating the feasibility and effectiveness of SAAGE in several schools in Massachusetts, New York, and Florida.

Assessing the Evidence Base for Interventions for School-Aged Children

In this project we are expanding upon the National Standards Project to identify parameters of effective interventions and to better understand (a) behavioral targets addressed by a given intervention, (b) implementers of interventions, and (c) the settings in which interventions have been found to be demonstrably effective.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • HHS Author Manuscripts

Logo of nihpa

Future Directions for Research in Autism Spectrum Disorders

Cara r. damiano.

A Department of Psychology, University of North Carolina, Chapel Hill, NC

E Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC

Carla A. Mazefsky

B Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA

Susan W. White

C Department of Psychology, Virginia Polytechnic Institute and State University, Blacksburg, VA

Gabriel S. Dichter

D Department of Psychiatry, University of North Carolina, Chapel Hill, NC

This article suggests future directions for research aimed at improved understanding of the etiology and pathophysiology of autism spectrum disorder (ASD) as well as pharmacologic and psychosocial interventions for ASD across the lifespan. The past few years have witnessed unprecedented transformations in the understanding of ASD neurobiology, genetics, early identification, and early intervention. However, recent increases in ASD prevalence estimates highlight the urgent need for continued efforts to translate novel ASD discoveries into effective interventions for all individuals with ASD. In this article we highlight promising areas for ongoing and new research expected to quicken the pace of scientific discovery and ultimately the translation of research findings into accessible and empirically supported interventions for those with ASD. We highlight emerging research in the following domains as particularly promising and pressing: (1) preclinical models; (2) experimental therapeutics; (3) early identification and intervention; (4) psychiatric comorbidities and the Research Domain Criteria (RDoC) initiative; (5) ecological momentary assessment; (6) neurotechnologies; and (7) the needs of adults with ASD. Increased research emphasis in these areas has the potential to hasten the translation of knowledge on the etiological mechanisms of ASD to psychosocial and biological interventions to reduce the burden of ASD on affected individuals and their families.

The pace of autism spectrum disorder (ASD) research has increased dramatically in recent years. In 2003, approximately 800 peer-reviewed journal articles were published on the topic of ASD. In 2013, this number had increased to over 3400 articles published in a 12-month period. This remarkable increase has paralleled rapidly rising ASD prevalence estimates, which have escalated from approximately 30/10,000 to 60/10,000 a decade ago ( Fombonne, 2003 ) to the most current Center for Disease Control (CDC) estimates of 1 in 68 children in the United States ( Frieden, Jaffe, Cono, Richards, & Iademarco, 2014 ). The financial toll of ASD is extraordinary, with the lifetime economic costs of ASD estimated to be up to $2.4 million per affected individual and the national costs for the United States estimated to be $66 billion per year for children with ASD and $175 billion for adults with ASD ( Buescher, Cidav, Knapp, & Mandell, 2014 ; Knapp & Buescher, 2014 ).

ASD is currently defined on the basis of core deficits in social communication and repetitive and stereotyped behaviors and sensory symptoms, but deficits are far-reaching and pervasive, including impairments in emotional functioning, irritability, aggression, self-injury, anxiety, and impulsivity ( Lecavalier, 2006 ). Present from very early in development, ASD presents as a complex array of psychological and biomedical symptoms. As such, ASD research requires a multidisciplinary perspective, including clinical psychology, developmental pediatrics, translational psychiatry, basic developmental neuroscience, cognitive neuroscience, and genetics. Similarly, the comprehensive treatment of individuals with ASD requires interventions for not only the core social communicative symptoms associated with ASD, but also a number of related impairments, including behavior and emotion regulation, gastrointestinal problems, sleep difficulties, and epilepsy ( Coury et al., 2014 ). ASD is also a highly heterogeneous disorder, including individuals with a wide range of symptom severity and intellectual and adaptive functioning. Finally, in more than 50% of cases, ASD presents in the context of a comorbid psychiatric condition, including internalizing disorders (e.g., anxiety, depression, bipolar disorder, and obsessive compulsive disorder) and externalizing disorders (e.g., ADHD, oppositional defiant disorder) ( Mazefsky et al., 2012 ). The breadth and depth of the challenges associated with ASD have impeded progress towards the development of research-informed and person-specific novel interventions.

Despite the seemingly intractable nature of ASD etiology, the past two decades have witnessed remarkable progress towards understanding the pathophysiology of ASD. Progress has been rapid in the area of neuroimaging in particular (see Anagnostou & Taylor, 2011 ; Ecker & Murphy, 2014 ; Minshew & Keller, 2010 , for reviews). Although neuroimaging findings in ASD are somewhat inconsistent due to different study methodologies, heterogeneity of participant samples, and other confounding factors ( Salmond, Vargha-Khadem, Gadian, de Haan, & Baldeweg, 2007 ; Toal et al., 2010 ), a number of consistent patterns have emerged. Major findings in structural brain imaging have included a pattern of early brain overgrowth in ASD ( Hazlett et al., 2011 ; Redcay & Courchesne, 2005 ), volumetric differences in frontal, limbic, and cerebellar regions ( Amaral, Schumann, & Nordahl, 2008 ), reduced white matter volume ( Ecker et al., 2012 ), and atypical development and greater disorganization of white matter tracts ( Aoki, Abe, Nippashi, & Yamasue, 2013 ; Wolff et al., 2012 ). Neuropathological studies have found atypicalities in cortical organization in ASD, particularly decreased pruning of prefrontal neurons ( Courchesne et al., 2011 ), abnormal structure and organization of cortical mini-columns ( Casanova, Buxhoeveden, Switala, & Roy, 2002 ), attenuated differentiation of temporal and frontal cortical cells ( Voineagu et al., 2011 ), and atypical axonal development ( Zikopoulos & Barbas, 2010 ). Functional neuroimaging studies have revealed decreased neural specialization for social information processing (e.g., processing of faces, biological motion, and theory of mind tasks) ( McPartland, Coffman, & Pelphrey, 2011 ; Pelphrey, Shultz, Hudac, & Vander Wyk, 2011 ), attenuated prefrontal activation during executive function tasks ( Philip et al., 2012 ), aberrant processing of auditory and language stimuli ( Gomot, Belmonte, Bullmore, Bernard, & Baron-Cohen, 2008 ; Redcay & Courchesne, 2008 ) including reduced left-lateralization for the processing of language ( Kleinhans, Müller, Cohen, & Courchesne, 2008 ; Knaus, Silver, Lindgren, Hadjikhani, & Tager-Flusberg, 2008 ; Redcay & Courchesne, 2008 ) and underconnectivity among brain systems both at rest and across a range of functional tasks ( Kennedy & Courchesne, 2008 ; Minshew & Williams, 2007 ). More generally, functional neuroimaging studies have revealed a pattern of enhanced activation in lower-order motor and sensory brain regions and attenuated activation of higher-order regions related to social cognition and executive function during complex tasks ( Di Martino et al., 2009 ) and more unreliable or variable cortical responses ( Dinstein et al., 2012 ; Müller, Kleinhans, Kemmotsu, Pierce, & Courchesne, 2003 ).

Research in ASD genetics has identified a number of genes that confer increased ASD risk (see Geschwind, 2011 ; State & Levitt, 2011 , for review ). The genetics of several Mendelian syndromes associated with ASD (including fragile X syndrome, Rett syndrome, and tuberous sclerosis) have been identified and genes contributing to the etiology of ASD can now be detected in up to 25% of cases ( Abrahams & Geschwind, 2008 ; Jeste & Geschwind, 2014 ; Miles, 2011 ). Molecular pathways upon which these ASD risk genes converge have been identified, including proteins involved in cell-cell interaction (e.g., NRXN1, CNTNAP2), proteins with activity-dependent expression (e.g., MET, PTEN), and proteins modulating neuronal activity (e.g., UBE3A, SCN2A) ( Berg & Geschwind, 2012 ).

Clinical research has aided in the refinement of effective tools for the phenotypic characterization of ASD. ASD is now generally conceptualized as a dimensional rather than a categorical disorder ( Lord & Jones, 2012 ) with two major symptom domains -social/communication and repetitive behaviors - rather than three ( Gotham, Risi, Pickles, & Lord, 2007 ; Lord et al., 2006 ). In addition, developmental trajectories of ASD traits from infancy into adulthood in ASD have been identified ( Anderson, Liang, & Lord, 2013 ; Landa, Gross, Stuart, & Bauman, 2012 ; McGovern & Sigman, 2005 ), and recent work has begun to characterize the characteristics of individuals who lose their ASD diagnosis over time (often referred to as ‘optimal outcomes’) ( Anderson et al., 2013 ; Fein et al., 2013 ). A number of effective psychosocial interventions have been developed and empirically validated to treat core and associated symptoms of ASD throughout the lifespan, including early behavioral intervention programs ( Dawson et al., 2010 ; Warren et al., 2011 ), social skills training groups ( Reichow, Steiner, & Volkmar, 2013 ), vocational intervention ( Taylor et al., 2012 ), parent training programs ( Kaminski, Valle, Filene, & Boyle, 2008 ; McConachie & Diggle, 2007 ; Virues-Ortega, Julio, & Pastor-Barriuso, 2013 ), and applied behavioral analysis (ABA) ( Lovaas, 1987 ; Virués-Ortega, 2010 ).

Collectively, this research has informed our understanding ASD as a genetically and biologically based neurodevelopmental disorder. However, there remains a great unmet need for interventions that reliably and robustly address the core symptoms of ASD and translational work linking more basic research findings with clinical practice remains somewhat limited. In the following sections, we will discuss avenues for future research in the domains of preclinical models, experimental therapeutics, early identification and intervention, psychiatric comorbidities and dimensional phenotypes, ecological momentary assessment, neurotechnology, and the needs of adults with ASD. We highlight these specific emerging novel directions for ASD research because we believe that they hold particular promise for improved understanding of ASD etiology and ultimately improved day-to-day functioning of individuals with ASD.

Preclinical Models

Preclinical models of ASD will continue to be a critical tool for providing insight into ASD etiology and to identify mechanistic targets for future experimental medicine agents. Animal models provide key information about neurobiological mediators relevant to ASD, thereby providing new insights into ASD etiology and suggesting targets for novel treatments. Because ASD is a polygenetic disorder, comparisons between different lines of knockout mice may provide insights into ASD-relevant dysfunctional brain systems and may identify converging molecular pathways from diverse genetic etiologies. The animal models that have been developed for single-gene disorders associated with ASD (e.g., Rett syndrome, Fragile X syndrome) ( Bakker et al., 1994 ; Chen, Akbarian, Tudor, & Jaenisch, 2001 ) may also help elucidate the mechanisms through which these genes contribute to ASD. Understanding when and how genetic risk factors are associated with particular molecular mechanisms will provide insight into the use and timing of novel therapeutics. Given the homology between mouse and human genomes, advances in the field of mouse genetics, including panels of genetically divergent strains and new strategies for controlled gene expression and engineering of mutant lines, support the use of murine models to investigate complex heritable factors in ASD ( Moy & Nadler, 2008 ; Moy, Nadler, Magnuson, & Crawley, 2006 ). As human neuroimaging advances in terms of resolution and analytic methodology, it will be important for animal research to investigate neurobiological markers that have been identified or could potentially be studied through human neuroimaging so that findings from animal model studies could be more easily translated as in vivo markers of ASD. However, in order to accomplish these goals, future preclinical research will need to address the methodological and practical factors that limit the potential to translate animal models into the clinical setting, carefully control for any factors that might impact translatability, and foster collaboration among preclinical and clinical scientists (see Lazic & Essioux, 2013 ; Markou, Chiamulera, Geyer, Tricklebank, & Steckler, 2008 ; Nestler & Hyman, 2010 , for further discussion of these topics).

Animal models are also vitally important for understanding the epigenetics of ASD, or how the environment affects the expression of ASD risk genes. Although ASD has a prominent genetic component ( Ronald & Hoekstra, 2011 ) with hundreds of putative contributing loci ( Geschwind, 2011 ), the environment also plays a key role in the etiology of ASD, likely via epigenetic modifications ( Miyake, Hirasawa, Koide, & Kubota, 2012 ). This complexity of causal factors has spurred preclinical research as a tool to clarify the roles of specific genes as well as environmental influences on ASD pathogenesis ( Oddi, Crusio, D'Amato, & Pietropaolo, 2013 ). Although several environmental and genetic factors that individually influence ASD have been identified, the future of ASD research may involve a better understanding of the interaction of genetic and environmental processes. This interaction is particularly difficult to characterize in neurodevelopmental disorders because both genetic and environmental factors may operate dynamically over the course of development. One example of the role of epigenetics in ASD comes from emerging evidence that gut microbiota may exert an epigenetic influence on brain function in ASD ( Stilling, Dinan, & Cryan, 2014 ) because altered gut microbiota has been linked to impaired social behaviors and repetitive behaviors in animal models ( Desbonnet, Clarke, Shanahan, Dinan, & Cryan, 2013 ) and ASD has been associated with altered expression of gut microbiota ( Adams, Johansen, Powell, Quig, & Rubin, 2011 ; Mulle, Sharp, & Cubells, 2013 ; Parracho, Bingham, Gibson, & McCartney, 2005 ). These findings raise the possibility that probiotics may be a potential treatment for ASD ( Dinan & Cryan, 2013 ). It is important to note, however, that our current understanding of the influence of gut microbiota and probiotics on brain function remains rudimentary as only correlational studies have been conducted in humans thus far. In addition, consistent patterns of microbiota profiles in ASD have not been identified: some studies have found both higher and lower concentrations of different microbiota whereas others have reported no differences in ASD ( Gondalia et al., 2012 ; Louis, 2012 ). Despite these inconsistencies, this area of research emphasizes the need to examine epigenetic influences beyond discrete processes within the brain alone.

An emerging area of research designed to complement preclinical animal model studies is the study of human neural stem cells ( Cocks et al., 2013 ; Vaccarino et al., 2011 ). This line of research involves collecting skin cells from individuals with ASD and then reprogramming them into induced pluripotent stem cells that are then stimulated to develop into neurons ( Takahashi & Yamanaka, 2006 ). Because these neurons maintain the same unique genetic makeup as the cells of the individual from which they were derived, this method allows scientists to examine the downstream effects of particular genetic mutations in vitro and understand atypical neuronal development in ASD. This approach is particularly relevant for neurodevelopmental disorders like ASD with different genetic etiologies and complex polygenic mechanisms that operate via a developmental cascade of events, as researchers can directly observe the molecular impact of particular genetic risk factors for different individuals. This approach may also be useful in addressing the heterogeneity observed in response to treatment in ASD, as it may be used to test how individuals with different genetic mutations respond to pharmacological treatments ( Eglen & Reisine, 2011 ), ultimately working towards individualized medicine for individuals with ASD.

Experimental Therapeutics

Over the past several decades, treatment research has successfully identified many evidence-based interventions for ASD that have resulted in improved cognitive functioning, social ability, communication skills, and emotion regulation ( Legg et al., 2007 ; Reichow, 2012 ; Seida et al., 2009 ). This work is complimented by evidence that these interventions result in significant changes in brain functioning in individuals with ASD ( Dawson et al., 2012 ; Van Hecke et al., 2013 ; Voos et al., 2013 ). However, despite this considerable progress, treatment response to these interventions is variable and reliable predictors of clinical outcome remain limited in ASD ( National Research Council, 2001 ; Sherer & Schreibman, 2005 ; Stahmer, Schreibman, & Cunningham, 2011 ). The fact that only 50% of individuals with ASD demonstrate substantial positive gains as a result of evidence-based interventions ( Stahmer et al., 2011 ) underscores the need to fractionate ASD in order to personalize ASD treatments.

Similarly, the success of psychopharmacological treatment of ASD remains limited. Despite the promise of novel pharmacological interventions such as oxytocin ( Gordon, 2014 ) and other agents targeting synaptic functioning ( Delorme et al., 2013 ) in ASD, there are currently no FDA-approved medications to treat the core impairments of ASD. Only two medications (risperidone and aripiprazole) are FDA-approved for use in ASD, and both are approved to treat symptoms of irritability often associated with ASD. Other pharmacotherapies are used off-label to treat co-morbid and co-occurring symptoms, such as agitation, anxiety, epilepsy, and untoward behaviors ( Dove et al., 2012 ; Doyle, McDougle, & Stigler, 2014 ). Across all psychiatric disorders including ASD, the inherent challenges associated with Phase III clinical trials have made it exceedingly difficult to identify potential new pharmacological agents. These challenges include large placebo effects ( King et al., 2009 ), the inability to stratify subgroups of individuals who are most likely to respond to a particular agent ( Scahill et al., 2012 ), and the prohibitive costs associated with bringing new pharmacologic treatments to market (over $800 million) ( DiMasi, Hansen, & Grabowski, 2003 ). Traditional models of drug development in psychiatry have resulted in only 4-8% of new agents receiving FDA approval ( Brady & Insel, 2012 ; Insel, 2012 ). Such challenges are amplified for disorders involving pediatric populations such as ASD because evaluations of drug engagement on brain molecular targets may not be feasible in pediatric samples and because of reliance on caretaker reports as measures of clinical outcomes. Additionally, because relevant molecular targets are parts of complex developmental pathways in pediatric disorders, demonstrating an agent's interaction with a specific receptor does not ensure an effect on relevant network level processing or clinical endpoints ( Javitt et al., 2011 ).

The slow pace of novel psychosocial and pharmacological treatment development in ASD may be attributable to a number of factors including: (1) the phenotypic and etiological heterogeneity of ASD that makes it exceedingly unlikely that a single treatment will be effective for all, or even most, individuals with ASD; (2) a diagnosis based on social communication which is inevitably context-dependent and requires extensively trained clinicians to evaluate; (3) a relatively limited understanding of the pathophysiology of ASD and clear relationships between potential etiologies and clinical symptoms; and (4) a lack of well defined self-report or caregiver-report outcome measures.

Recent changes in the funding priorities and initiatives of the National Institute of Mental Health (NIMH) provide some direction for the future of intervention research in this field ( Insel & Gogtay, 2014 ). In 2012, the NIMH released a series of initiatives (“Fast-Fail Trials”) to speed the testing of new or repurposed compounds. This initiative is particularly relevant in the context of ASD where clinical endpoints in traditional randomized controlled trials have been difficult to define. The aim of this initiative is to rapidly identify promising agents and to identify brain targets for the development of additional candidate agents. As such, Fast-Fail trials are designed to evaluate whether a compound engages a particular neurobiological target (i.e., a specific receptor or neurotransmitter) and whether this target engagement then alters clinical functioning (e.g., improves social attention) ( Borsook, Hargreaves, & Becerra, 2011 ; Insel, 2012 ; Paul et al., 2010 ; Wagner, 2008 ). These trials are designed to be far smaller in scope than traditional clinical trials and, once safety has been established, will not rely on preclinical studies prior to testing in human patients. A multi-institutional Fast-Fail trial for ASD is currently underway [Fast-Fail Trials in Autism Spectrum Disorders (FAST-AS), HHSN271201200005I] focused on compounds that enhance gamma-aminobutyric acid (GABA) functioning using neuroimaging tools to index changes in neurobiological targets. Studies modeled after these initial Fast-Fail trials are likely to become more prominent in ASD research given their alignment with NIMH funding priorities and their cost efficiency.

More recently, the NIMH has built upon this model and announced broad new directives for intervention research that are referred to as “experimental medicine” or “experimental therapeutics” ( Insel, 2014 ). This approach emphasizes the need to identify genetic and neurobiological mechanisms of action associated with interventions. Future ASD intervention research conducted under this framework will need to evaluate the extent to which both pharmacologic and psychosocial interventions engage biological targets relevant to core ASD deficits. In other words, putative interventions should also serve as probes measuring engagement of relevant targets (e.g., a pertinent neural circuit). This directive implies that although randomized clinical trials will continue to evaluate efficacy in terms of traditional clinical endpoints, such trials will need to incorporate quantifiable measures of target engagement. Such an initiative will ensure that, even if a novel ASD therapeutic fails in terms of primary clinical endpoints, the trial will nonetheless yield valuable insights into ASD mechanisms and novel targets to evaluate in future treatments. In addition, the extent to which the target is activated should presumably inform our understanding of individual differences in response to treatment and the optimal dose and duration of an intervention.

Undoubtedly, this new direction for clinical trials raises important challenges for ASD research, including selection of appropriate treatment targets and difficulty obtaining certain target engagement metrics (e.g., functional neuroimaging) from less cognitively able individuals. However, despite these challenges, this paradigm shift in clinical trials may be particularly valuable for ASD research given the limited success in identifying novel ASD therapeutics to date and the emphasis on quantifiable, objective mechanistic targets. These changes may ultimately speed treatment development for ASD in the future. For example, whereas a traditional phase III randomized clinical trial of a novel psychosocial treatment for social communication impairments in ASD would necessitate a multisite endeavor to ascertain a sufficiently large and diverse sample (e.g., 100+ participants) and evaluate outcomes such as caregiver report metrics of social functioning, an experimental medicine approach could entail first a preliminary evaluation of whether an intervention modulates objective quantitative measures of social communication (e.g., gaze patterns to critical regions of the face in an eye-tracking paradigm or increased brain activation in regions related to social information processing when viewing faces) in a relatively small number of individuals with ASD. If the intervention successfully engaged the target measure of social communication, the intervention would then be evaluated in larger samples. This approach sheds light on the mechanisms of action involved in an intervention (and perhaps even the mechanisms involved in the etiology of ASD to some extent) and prevents wasteful spending on large clinical trials for ineffective interventions.

To adopt an experimental medicine approach in evaluating novel ASD treatments, interventionists will need to consider how to adapt their research programs in light of this new framework while maintaining continuity with traditional clinical trials. This may be accomplished by focusing on the following areas. First, an improved mechanistic understanding of core ASD symptom domains (e.g., social communication) and how to optimally measure (e.g., functional neuroimaging, electrophysiology, eyetracking) these domains as intervention targets will ensure that assays of target engagement are optimally sensitive. Next, it will be critical to leverage such mechanistic understanding to develop novel interventions with relevant hypothesized mechanisms of action. Another aspect of this line of research will be the translation of basic science findings to develop ideas for novel interventions and their possible mechanistic targets. Finally, it will be important to understand relations between the dose and duration of an intervention and its sustained impact on target engagement, consistent with the emphasis in managed healthcare in the United States on evidence for optimal treatment dose and duration ( Hansen, Lambert, & Forman, 2002 ).

Early Identification and Intervention

The experimental medicine perspective outlined above clearly compels scientists to develop a better understanding of the etiological bases of ASD. Given the complex neurodevelopmental nature of ASD, any theoretical model developed to explain its etiology will necessarily depend on further study of early brain development and the neurodevelopmental sequelae that result in ASD symptoms. A better understanding of early neurobiological ASD mechanisms will be critical for advancing early identification of ASD. ASD is not typically diagnosed until around four years of age in the United States ( Rice, 2009 ). Whereas age of first diagnosis is likely constrained to some extent by service availability and the quality of pediatric care ( Mandell, Novak, & Zubritsky, 2005 ), clearly discernible behavioral symptoms of ASD may not emerge until at least 12 months of age in most children with ASD ( Ozonoff et al., 2010 ). On the other hand, neurobiological or endophenotypic atypicalities may be evident in infants at risk for ASD as young as 6 months of age ( Elsabbagh et al., 2012 ; Shen et al., 2013 ; Wolff et al., 2012 ), and a recent study even reported that children diagnosed with ASD were characterized by abnormalities in prenatally determined cortical laminar neurons ( Stoner et al., 2014 ). This latter finding is also supported by studies demonstrating placental abnormalities (i.e., trophoblast inclusions) associated with risk for ASD ( Anderson, Jacobs-Stannard, Chawarska, Volkmar, & Kliman, 2007 ; Walker et al., 2013 ).

As markers for ASD are identified earlier in life, an important question will be how to diagnose and characterize ASD in infancy. While there is some evidence that a reliable and stable diagnosis can be made as early as 14 months in children with ASD ( Chawarska, Klin, Paul, Macari, & Volkmar, 2009 ; Chawarska, Klin, Paul, & Volkmar, 2007 ), other studies suggest that ASD diagnoses before three years may be relatively unstable, particularly in siblings of children with ASD ( Kleinman et al., 2008 ; Lord et al., 2006 ; Sutera et al., 2007 ; Turner & Stone, 2007 ). Practice guidelines in the United States, such as those developed by the American Academy of Child and Adolescent Psychiatry (AACP) suggest that any early developmental assessment include several questions related to ASD symptoms ( Volkmar, Cook Jr, Pomeroy, Realmuto, & Tanguay, 1999 ). Similarly, the Scottish Intercollegiate Guidelines Network (SIGN) indicates that the minimum age for a reliable ASD diagnosis is currently unknown, yet suggests that ASD be considered in any differential diagnosis in which development is disrupted even if the child is not yet demonstrating behaviors typical of ASD ( McClure, 2014 ; Scottish Intercollegiate Guidelines Network (SIGN), 2007 ). The National Institute for Health and Care (NICE) guidelines in England suggest that any child under 3 years of age with regression in language, social skills, or motor abilities should be referred for an ASD diagnosis, yet do not state a minimum age for diagnostic assessments ( National Institute for Health and Clinical Excellence (NICE), 2011 ).

Given this potential diagnostic instability of ASD in infancy, it will be important to develop a system for categorizing infants at high-risk for ASD who do not yet meet criteria for a diagnosis since earlier intervention is associated with better clinical outcomes in ASD ( National Research Council, 2001 ) and the “wait and see” approach could ultimately be detrimental to a child's development. Indeed, parents of children with ASD report that the “wait and see” approach is commonly adopted by pediatricians as well as mental health clinicians, much to their frustration ( Goin-Kochel, Mackintosh, & Myers, 2006 ). Accordingly, it may ultimately be useful to institute a diagnosis of “pre-ASD” or “prodromal ASD” that could be given before an infant meets full ASD criteria but warrants early intervention or prevention. This may help to alleviate clinicians’ and parents’ concerns of labeling or stigmatizing children at an early age, while also providing access to intervention. Similar high-risk labels have been implemented in other fields of medicine to describe pre-diabetes and pre-hypertension ( American Diabetes Association, 2010 ; Chobanian et al., 2003 ). These classifications are associated with specific risk markers and indicate that, without intervention, the disease will likely progress into its full expression. Another approach would be to categorize infants at risk for a range of neurodevelopmental disorders (e.g., ASD, developmental disability, attention-deficit/hyperactivity disorder), since many of these disorders share overlapping risk factors ( Gillberg, 2010 ). Recent large-scale studies have also revealed that approximately one fifth of siblings of children with ASD who do not go on to receive a formal diagnosis of ASD nevertheless go on to exhibit higher levels of ASD symptoms or lower levels of developmental functioning at three years of age, suggesting the importance of monitoring services availability for children with siblings with ASD ( Messinger et al., 2013 ).

A recent eye-tracking study raised the possibility that social developmental trajectories could be quantified through “growth charts” that plot changes in social reciprocity across infancy ( Jones & Klin, 2013 ). These growth charts could then be monitored and compared to normative standards for social development, similar to how height, weight, and head circumference are typically monitored in young children. Children who deviate significantly from the normative trajectory would then be referred for a comprehensive evaluation. This approach, which is undoubtedly an important direction for research in early identification, emphasizes the importance of individual developmental trajectories. Future research could build upon this study by tracking changes in behavioral manifestations of ASD alongside developmental changes in gene expression and brain growth. These growth charts derived on the basis of behavioral and brain phenotypes may allow for not only improved identification of at-risk infants, but also perhaps optimal matching of biopsychosocial and psychopharmacologic treatment for specific patients at particular periods during development. However, further research is needed in order to support the utility of these growth charts in ASD.

Future research should also move towards identifying risk factors for ASD in both high-risk and low-risk populations. The majority of research on early identification thus far has involved the unique population of infant siblings of children with ASD, although some studies include infants identified as high risk through specific screening tools (e.g., Wetherby et al., 2004 ) or pre-term infants who are known to have a higher risk for ASD (e.g., Limperopoulos et al., 2008 ). These high-risk samples may not be representative of the general population of infants who are later diagnosed with ASD and risk factors could vary greatly between high-risk and low-risk groups. For example, greater fixation on the eyes versus the mouth in early infancy for high-risk groups may in fact be detrimental to the development of language whereas greater fixation on the eyes in typically developing groups is not correlated with a poor outcome ( Young, Merin, Rogers, & Ozonoff, 2009 ).

Future research should also place greater emphasis on protective factors versus risk factors associated with ASD by studying the individuals at risk for ASD (e.g., infant siblings, individuals with genetic risk variants associated with ASD, premature infants) who do not go on to meet criteria for an ASD diagnosis. Identifying ASD protective factors will provide important clues for novel prevention and/or invention approaches. For example, hypotheses may be derived about protective factors related to optimal outcomes from research on factors that influence how a child responds to environmental influences and/or early intervention. Social engagement, family factors, temperament, and visual attention as well as severity of early verbal deficits and nonverbal functioning may all be candidate protective factors.

Psychiatric Comorbidities and Research Domain Criteria (RDoC)

An appreciation of comorbidity in ASD is a relatively recent phenomenon. Historically, clinicians often subsumed secondary symptoms (e.g., excessive fears) under the diagnosis of ASD. In this way, difficulties and symptoms were attributed to the more prominent (primary) diagnosis of ASD ( Mason & Scior, 2004 ). Alternatively, the identification and treatment of comorbid disorders and secondary symptoms may be productive clinically, providing much needed symptom relief, motivating the client for further treatment, and increasing quality of life and daily adaptive functioning. However, it is also equally important that comorbid conditions do not take clinical attention away from core ASD symptoms in need of intervention (e.g., treatment for a child with ASD and comorbid social anxiety may include only anxiety reduction rather than also efforts to improve social communication skills).

Most individuals with ASD have at least one comorbid psychiatric disorder ( e.g., Mazefsky et al., 2012 ). Indeed, comorbidity is more the rule than the exception in ASD as well as most neurodevelopmental disorders. This high level of comorbidity could be attributable to similar or associated risk factors, the occurrence of one disorder increasing the risk of another disorder (i.e., sequential comorbidity), misdiagnosis, or the inadequacy of our diagnostic systems to reflect the true nature of psychiatric disorders ( Caron & Rutter, 1991 ). Comorbidity in young people with ASD tends to persist well into adolescence ( Simonoff et al., 2013 ) and is associated with more impaired social functioning ( Chang, Quan, & Wood, 2012 ). Although our understanding of the processes that contribute to the high rates of comorbidity in ASD remains limited, this line of research may be particularly important as it may provide important clues to the causal mechanisms and the potential risk and protective factors involved in ASD ( Rutter, 1997 ). In addition, relative to the amount of research to date on the treatment of comorbid problems in other disorders, there has been almost no research on interventions involving comorbid presentations in ASD. Clinical outcome studies have demonstrated, for instance, that CBT is an effective treatment for anxiety disorders in children and adolescents with ASD (e.g., Reaven, Blakeley□ Smith, Culhane□ Shelburne, & Hepburn, 2012 ; White et al., 2013 ), however most of the work to date on mechanisms underlying anxiety in the context of ASD has been theoretical. Treatment outcome research has consisted mostly of studies using interventions that target mechanisms known to contribute to maintenance of anxiety in typically developing children (e.g., distorted thoughts). Although similar mechanisms operate in ASD, this has not been tested empirically. In light of the high level of variability seen in treatment response in ASD relative to treatment outcomes in typically developing samples (e.g., Lickel, MacLean, Blakeley-Smith, & Hepburn, 2012 ), it is possible that there are different, or additional, mechanisms that must be considered in the context of ASD.

Targeting two broad classes of processes likely involved in high rates of comorbidity in ASD may be particularly productive. The first class is core developmental processes directly linked to the etiology of ASD (e.g., impaired joint attention and social attention); the second class includes broader, transdiagnostic risk processes. It is possible (and perhaps likely) that as developing social neural systems increasingly depart from ‘normal’ trajectories in a child with ASD, other processes related to mental health may be affected as well. In this vein, we can consider core processes such as social aloofness and atypical social information processing in the possible pathogenesis of comorbid conditions. As a concrete example, decreased hedonic responses to the social-emotional bids of others may be involved in the development of oppositional problems or aggression. The second class is transdiagnostic processes that are not necessarily causally linked to ASD core impairments. Rather, they are ‘fundamental’ in the sense that they are central to many forms of psychopathology. There are many transdiagnostic processes, such as attentional avoidance, persistent negative affect, and rumination (e.g., ( e.g., Harvey, Watkins, Mansell, & Shafran, 2004 ). Poor emotion regulation, for example, is a transdiagnostic process that has been linked theoretically to the high rates of anxiety disorders seen in people with ASD ( Mazefsky et al., 2013 ; White, Schry, Miyazaki, Ollendick, & Scahill, 2014 ). These processes occur over the course of development and thus it will be important for future research to consider the longitudinal course of comorbidity and the possibility of sequential comorbidities over the course of a lifetime ( Rutter, Kim Cohen, & Maughan, 2006 ).

How these two types of processes relate to psychiatric comorbidity in people with ASD is under-explored. If a given process were to contribute to the development of comorbid conditions in ASD, that process could be a treatment target. This approach to translational medicine may be effective and may contribute to more sustained and generalized treatment effects given that transdiagnostic processes are thought to underlie a range of expressions of pathology. Perhaps the most prominent pragmatic challenge associated with comorbidity research is how to define the target population and ascertain the study sample. Ideally, recruitment should be based on the target mechanism (e.g., impaired emotion regulation) rather than behavioral criteria.

An alternative framework to conceptualize symptoms that commonly co-occur in ASD is NIMH's recently developed Research Domain Criteria (RDoC) initiative. This novel conceptualization of psychopathology eschews traditional DSM diagnoses defined on the basis of groupings of observable symptoms and rather focuses on dimensional constructs with linkages to tractable neurobiological mechanisms ( Casey et al., 2013 ). This framework is predicated on the inevitable conclusion that because psychiatric disorders are currently diagnosed on the basis of symptom presentation rather than biology-based etiology, no disorder can be expected to be associated with a unitary, underlying pathology, and, conversely, no single genetic variant could produce the wide array of behavioral manifestations observed in a given disorder ( Licinio & Wong, 2013 ). A host of evidence suggests that ASD comprises a heterogeneous grouping of patients characterized by non-overlapping etiologies and presentations ( Geschwind & Levitt, 2007 ). Given the heterogeneous nature of ASD, it is not surprising that a wide array of candidate brain circuits and molecular targets have been implicated in ASD. The near-impossibility of finding a unifying neurobiological account of ASD has led some to suggest that improved intervention approaches will only be achieved if future research focuses on individual variation and stratification of individuals with ASD ( McCray, Trevvett, & Frost, 2013 ; Waterhouse & Gillberg, 2014 )

The Research Domain Criteria (RDoC) initiative argues that psychiatric conditions are brain disorders characterized by dysregulated neural circuits regulating critical dimensional constructs, including processing of positive and negatively stimuli, cognition and memory, and social communication ( Cuthbert & Insel, 2013 ). Under the RDoC framework, the current definition of ASD is a somewhat arbitrary and ill-defined clustering of symptoms that are not necessarily closely related in terms of biology. A number of RDoC constructs are relevant to impairments that are common in ASD, including social processing (e.g., social communication and perception and understanding of others), negative valence systems (e.g., fear, anxiety, and frustrative non-reward), positive valence systems (e.g., initial and sustained response to rewards), and cognitive control (including response selection, inhibition, and suppression). Although development and environmental factors are not currently part of the RDoC framework, clearly any comprehensive program of ASD research will need to include these factors in their explanatory models. The long-term goal of the RDoC initiative is to establish a research database that will allow for multi-modal dimensional classification of traits related to ASD neurobiology and to foster research into the development of novel therapeutic agents that target these dimensional traits.

Ecological Momentary Assessment (EMA) in ASD

Research addressing social functioning in ASD has predominantly used retrospective questionnaires administered to probands or their caregivers to assess symptoms over a period of weeks to months. However, recall of emotions and experiences are often biased, and it is not uncommon for clinical neuroscience studies to report only modest correlations with symptom expression on self-report or caregiver-report instruments. Given that impairments in understanding and recollection of internal socio-affective states are central to ASD ( Schwartz, Neale, Marco, Shiffman, & Stone, 1999 ), innovative methods that capture emotions in an ecologically valid way are critical to advance our understanding of emotional functioning in ASD. Ecological momentary assessment (EMA) offers the promise of novel clinical endpoints for trials of interventions designed to improve social communicative functioning in ASD.

Ecological momentary assessment is a method for obtaining subjective information from respondents in a natural setting and is particularly useful for gathering information about context-dependent states ( Stone et al., 1998 ). EMA has been used to capture mood, stressful events, and coping strategies and has been shown to be less susceptible to the memory decay ( Coyne & Gottlieb, 1996 ) and systematic recall bias that is characteristic of standard questionnaires ( Schwartz et al., 1999 ; Whalen, Jamner, Henker, & Delfino, 2001 ). EMA has been successfully used in psychiatric contexts ( Shiffman, Stone, & Hufford, 2008 ) as well as with pediatric samples and severely mentally ill samples ( aan het Rot, Hogenelst, & Schoevers, 2012 ; Granholm, Ben-Zeev, Fulford, & Swendsen, 2013 ; Marhe, Waters, van de Wetering, & Franken, 2013 ; Tan et al., 2012 ) and EMA offers relatively increased ecological validity relative to laboratory measures and is thus a natural compliment to laboratory-based studies ( Myin-Germeys et al., 2009 ; Shiffman et al., 2008 ; Stone & Shiffman, 2002 ). The accessibility and mobility of technology such as smartphones and freely-available survey software has made EMA a highly attractive method to collect self-report data in naturalistic contexts. EMA via smartphones may be especially well suited for adolescents with ASD given this population's strengths in using technology ( Klin, McPartland, & Volkmar, 2005 ) and preference for electronics over other leisure activities ( Shane & Albert, 2008 ). A small qualitative study found that adults with ASD enjoyed an EMA procedure ( Hurlburt, Happe, & Frith, 1994 ) and Khor and colleagues ( 2014 ) reported excellent feasibility of using EMA in high functioning 12-18 year olds with ASD and that rates of EMA adherence were not correlated with ASD symptom severity, age, or gender.

To illustrate the potential impact of incorporating EMA into translational research, consider the example of a functional brain imaging study designed to explore activation in brain regions that process social information by presenting social stimuli in the scanner environment. The framework of this design is to assess brain function in the context of a social “press” (i.e., viewing images of faces) as a proxy for brain activation in real-world social contexts. However, in most such studies, potential brain–behavior relations are evaluated via correlations between neuroimaging data and a dimensional measure of social functioning completed either by the caregiver, who may have limited insight into aspects of their child's response to social experiences, or the research participant with ASD, who, by definition, has limited insight into internal states. It is little wonder that such correlations may be modest or non-significant, and such correlations should be interpreted with caution even in the context of significant associations. An alternative approach would be to query the participant a given number of times during the preceding days by smartphone about their feeling states via a brief (likely picture-based) questionnaire, and, most importantly, about their social context when completing the questionnaires. For example, the participant may report low anxiety when alone or when engaged in a preferred activity but may report higher anxiety in the presence of peers. In addition to the context-specificity of such reports, the repeated nature of data collection (e.g., repeated administration over a period of days to weeks) would address questions about variability and diurnal variation, providing richness to symptom data to compliment the complexity of the laboratory-based neuroimaging data. A finding that activation in social brain regions while viewing faces is correlated with symptom severity in the context of peers but not in the context of family or when alone would provide a deeper context for the neural data than a simple correlation with a retrospective self-report measure. Thus, EMA is an underused, but potentially powerful tool in ASD research.

Neurotechnologies

Neurotechnology refers to any technology that interacts with the human central nervous system. At the core of artificial intelligence, neurotechnology involves the use of technology to influence human thought or perception. What is being termed the ‘neurotechnology revolution’ has officially arrived ( Scott, 2013 ), and there is increased merging of human and computer such that we have fully thought-powered robots and virtual avatars. This integration of thought and machines is at the heart of brain-computer interface (BCI) devices. Often used in digital gaming, BCI devices have clinical utility as well, and have been used to assist in recovery and symptom management in stroke, paralysis, and degenerative conditions such as amyotrophic lateral sclerosis (e.g., ( Moghimi, Kushki, Marie Guerguerian, & Chau, 2013 ). Research on clinical applications of such neurotechnologies to mental health issues is emerging, and many are excited by the possibility that BCI and other such approaches may be useful in helping individuals with ASD in the areas of communication and social impairment.

Neurotechnologies are typically portable, easily adopted, and fun to use. They also do not present a side effect profile, and there are rarely ‘dosage’ limitations. These qualities make it likely that clinically effective technologies will be highly translational and well-disseminated. Although primarily anecdotal, there is some empirical research to support the assertion that people with ASD have, in general, an affinity for technology and computers ( e.g., Faja, Aylward, Bernier, & Dawson, 2008 ). Chen and Bernard-Opitz ( 1993 ) found that most students with ASD were more motivated to learn when using computer-based instruction relative to traditional, in-person instruction.

Available technologies have grown exponentially in terms of sophistication and accessibility to end-users over the past five years. This is perhaps most evident in the growing popularity of multi-user, virtual reality games. Likewise, there are inexpensive commodity BCI devices (e.g., NeuroSky, MindSet). Such technologies provide the user with the opportunity to interact with others (virtual bots, sometimes controlled by other people) in virtual social interactions. The social interaction deficits of ASD have proven difficult to rectify in meaningful, durable ways using traditional clinical approaches. It is possible that clinical impact and sustainability is limited by physiological over-arousal and anxiety in the context of other people in ASD, and social interaction may be less stressful and more predictable (and controllable) in virtual social interaction than in live, human-human interactions. However, the effectiveness of these virtual reality approaches has yet to be tested in randomized controlled trials that directly compare virtual reality interventions to placebo, much less to other, evidence-based approaches. It also remains unclear the extent to which social skills developed in a virtual reality context translate into a naturalistic social environment.

The diffuse etiology and pervasive impairments seen in ASD may be the primary reason why neurotechnological approaches, such as BCI, may be especially applicable to this population. Psychosocial treatments that target specific behaviors may prove less effective in the long-term than approaches that target more central and proximal processes from which multiple symptoms may emerge ( Lerner, White, & McPartland, 2012 ). Consider deficits in facial emotion recognition (FER) as an example. Impairments in FER are commonly reported in ASD ( e.g., Harms, Martin, & Wallace, 2010 ), yet it is not known how these deficits may contribute to impairments in social functioning. If FER deficits could be rehabilitated, this could contribute to improvements in a range of behaviors, such as expressed empathy, emotion regulation, and daily social competence. Neurotechnologies are promising in this regard since they allow tighter experimental control in efforts to intervene at the process (i.e., mediator) level of the deficit. Neurotechnologies should be further explored as we seek to translate mediators of ASD symptoms to clinical interventions. Developmental and applied research in this area should complement research on more traditional, less interactive technology-based interventions. Such approaches may allow for a theoretically grounded, client-responsive intervention that can more directly target key mechanisms than existing psychosocial and pharmacological treatments.

Needs of Adults with ASD

In comparison to children with ASD, adults with ASD have been markedly understudied ( Piven & Rabins, 2011 ). In fact, translational treatment research for adults with ASD is probably the least developed area of ASD research. A recent review of interventions for adults with ASD ( Bishop-Fitzpatrick, Minshew, & Eack, 2013 ) found only 13 studies that could be considered randomized controlled trials of interventions for adults with ASD. This lack of evidence-based treatments for adults combined with federal mandates that cease special education services once an individual reaches the age of 21 indicates an urgent need to develop assessment, treatment, and support services for adults with ASD.

Core symptoms of ASD and secondary behavioral problems often improve throughout adolescence but then improvement halts or even reverses during young adulthood ( Smith, Maenner, & Seltzer, 2012 ; Taylor & Seltzer, 2010 ). It is unclear to what extent this trend is due to a biologically-determined developmental progression or the cessation of supportive services. Prospective, longitudinal studies that consider moderators and mediators of successful outcomes will be essential to understanding these developmental trajectories given the urgent need to develop and disseminate effective interventions and support programs for adults with ASD.

Addressing the challenges faced by adults with ASD will require more than upward extension of effective services for children with ASD. For those with limited verbal and cognitive abilities who are unable to care for themselves, long-term dependence on their parents or other caregivers is common ( Billstedt, Gillberg, & Gillberg, 2011 ). Caregivers of children with ASD are typically their greatest advocates, and many of the treatment foci of childhood, such as communication, daily living skills, and social interaction, may still require attention into adulthood. In addition to these treatment foci, independence in tasks of daily living is often a primary concern. Finding a suitable arrangement that continues to foster positive gains after caregivers are no longer able to play this role may require rethinking group home and structured employment programs that keep adults stimulated and progressing.

In general, however, treatments for adults with ASD will likely have a number of different goals than those for children with ASD. Adult-specific targets include vocational training, supporting the transition from the structure of secondary education to work or school, and sexuality ( see Mazefsky & White, 2014 , for review ) Whereas treatment for children involves parents and providers making treatment decisions, intervention goals for adults with ASD may be more patient-driven and may require person-specific quality-of-life decisions that include self-acceptance and symptom management.

Additionally, treatment of adults with ASD may align less with the medical model and more with the “neurodiversity” model, a framework largely spearheaded by adults with ASD that argues that neurological differences in ASD are natural variations that should be accepted and celebrated rather than conceptualized as a disease to be cured ( Kapp, Gillespie-Lynch, Sherman, & Hutman, 2013 ). The neurodiversity movement is not opposed to treatment but acknowledges the need to maximize positive outcomes, suggesting possible directions for the future of adult ASD research, including a greater emphasis on acceptance and self-advocacy. One implication of this perspective is the need to advocate for increased tolerance, understanding, and respect for persons with ASD. The success of peer training approaches, which involve teaching typically developing children about ASD, is a testament to the potential of this approach. Adults with ASD could play a large role in this regard by engaging in advocacy efforts, with some well-known adults with ASD already having transformative effects in this area (e.g. Temple Grandin, John Elder Robison, Ari Ne’eman). Efforts towards teaching self-advocacy as a form of treatment also imply an emphasis on strength-building and awareness, concepts that may apply across levels of cognitive ability. Future efforts in the area should focus on developing coping strategies, strength-building, and societal adaptation and acceptance.

Conclusions

In the over seventy years since Leo Kanner first described autism ( Kanner, 1943 ), there has been remarkable progress in the areas of improved understanding of ASD neurobiology, genetics, early identification, and early intervention. However, recent increases in ASD prevalence estimates suggest the pressing need to translate these gains into access to effective interventions for all individuals with ASD. Here we have highlighted promising areas for future research to increase the pace of scientific discovery and ultimately the translation of research findings into accessible and empirically supported interventions for those with ASD across the lifespan. Future research in the areas described in this paper will need to address the factors that have constrained treatment development thus far by shifting focus to the following: (1) the study of individual differences within the ASD population in order to better account for etiological and phenotypic heterogeneity; (2) a greater emphasis on mechanistic processes and longitudinal developmental trajectories rather than outcomes or endpoints; (3) understanding the high level of psychiatric comorbidities and overlapping features shared with other neurodevelopmental disorders; (4) integration of different research methodologies (e.g., behavioral and brain imaging measures); and, (5) the development of ASD interventions that match the needs and desires of individuals with ASD and their families, including improving the functioning of individuals with ASD while preserving the positive and unique attributes of each individual with ASD.

Acknowledgments

The writing of this manuscript was supported by the following NIH grants: HD079124, HD060601, MH073402, and MH081285. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

The authors report no conflicts of interest.

  • aan het Rot M, Hogenelst K, Schoevers RA. Mood disorders in everyday life: a systematic review of experience sampling and ecological momentary assessment studies. Clinical Psychology Review. 2012; 32 (6):510–523. [ PubMed ] [ Google Scholar ]
  • Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nature Reviews Genetics. 2008; 9 (5):341–355. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Adams JB, Johansen LJ, Powell LD, Quig D, Rubin RA. Gastrointestinal flora and gastrointestinal status in children with autism--comparisons to typical children and correlation with autism severity. BMC Gastroenterology. 2011; 11 (1):22. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Amaral DG, Schumann CM, Nordahl CW. Neuroanatomy of autism. Trends in Neurosciences. 2008; 31 (3):137–145. [ PubMed ] [ Google Scholar ]
  • American Diabetes Association Diagnosis and classification of diabetes mellitus. Diabetes Care. 2010; 33 (Supplement 1):S62–S69. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Anagnostou E, Taylor MJ. Review of neuroimaging in autism spectrum disorders: what have we learned and where we go from here. Molecular Autism. 2011; 2 (4):1–9. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Anderson DK, Liang JW, Lord C. Predicting young adult outcome among more and less cognitively able individuals with autism spectrum disorders. Journal of Child Psychology and Psychiatry. 2013; 55 (5):485–494. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Anderson GM, Jacobs-Stannard A, Chawarska K, Volkmar FR, Kliman HJ. Placental trophoblast inclusions in autism spectrum disorder. Biological Psychiatry. 2007; 61 (4):487–491. [ PubMed ] [ Google Scholar ]
  • Aoki Y, Abe O, Nippashi Y, Yamasue H. Comparison of white matter integrity between autism spectrum disorder subjects and typically developing individuals: a meta-analysis of diffusion tensor imaging tractography studies. Molecular Autism. 2013; 22 (4):25. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Bakker CE, Verheij C, Willemsen R, Van der Helm R, Oerlemans F, Vermey M, Reyniers E. Fmr1 knockout mice: a model to study fragile X mental retardation. Cell. 1994; 78 (1):23–33. [ PubMed ] [ Google Scholar ]
  • Berg JM, Geschwind DH. Autism genetics: searching for specificity and convergence. Genome Biology. 2012; 13 (7):247. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Billstedt E, Gillberg IC, Gillberg C. Aspects of quality of life in adults diagnosed with autism in childhood: a population-based study. Autism. 2011; 15 (1):7–20. [ PubMed ] [ Google Scholar ]
  • Bishop-Fitzpatrick L, Minshew NJ, Eack SM. A systematic review of psychosocial interventions for adults with autism spectrum disorders. Journal of Autism and Developmental Disorders. 2013; 43 (3):687–694. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Borsook D, Hargreaves R, Becerra L. Can functional magnetic resonance imaging improve success rates in CNS drug discovery? Expert Opinion on Drug Discovery. 2011; 6 (6):597–617. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Brady LS, Insel TR. Translating discoveries into medicine: psychiatric drug development in 2011. Neuropsychopharmacology. 2012; 37 (1):281–283. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Buescher AV, Cidav Z, Knapp M, Mandell DS. Costs of Autism Spectrum Disorders in the United Kingdom and the United States. JAMA Pediatrics. 2014 [ PubMed ] [ Google Scholar ]
  • Caron C, Rutter M. Comorbidity in child psychopathology: Concepts, issues and research strategies. Journal of Child Psychology and Psychiatry. 1991; 32 (7):1063–1080. [ PubMed ] [ Google Scholar ]
  • Casanova MF, Buxhoeveden DP, Switala AE, Roy E. Minicolumnar pathology in autism. Neurology. 2002; 58 (3):428–432. [ PubMed ] [ Google Scholar ]
  • Casey B, Craddock N, Cuthbert BN, Hyman SE, Lee FS, Ressler KJ. DSM-5 and RDoC: progress in psychiatry research? Nature Reviews Neuroscience. 2013; 14 (11):810–814. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Chang Y-C, Quan J, Wood JJ. Effects of anxiety disorder severity on social functioning in children with autism spectrum disorders. Journal of Developmental and Physical Disabilities. 2012; 24 (3):235–245. [ Google Scholar ]
  • Chawarska K, Klin A, Paul R, Macari S, Volkmar F. A prospective study of toddlers with ASD: short-term diagnostic and cognitive outcomes. Journal of Child Psychology and Psychiatry. 2009; 50 (10):1235–1245. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Chawarska K, Klin A, Paul R, Volkmar F. Autism spectrum disorder in the second year: Stability and change in syndrome expression. Journal of Child Psychology and Psychiatry. 2007; 48 (2):128–138. [ PubMed ] [ Google Scholar ]
  • Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nature Genetics. 2001; 27 (3):327–331. [ PubMed ] [ Google Scholar ]
  • Chen SHA, Bernard-Opitz V. Comparison of personal and computer-assisted instruction for children with autism. Mental Retardation. 1993; 31 (6):368–376. [ PubMed ] [ Google Scholar ]
  • Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL, Jr, Wright JT., Jr The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report. Journal of the American Medical Association (JAMA) 2003; 289 (19):2560–2571. [ PubMed ] [ Google Scholar ]
  • Cocks G, Curran S, Gami P, Uwanogho D, Jeffries AR, Kathuria A, Ogilvie C. The utility of patient specific induced pluripotent stem cells for the modelling of Autistic Spectrum Disorders. Psychopharmacology. 2013:1–10. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Courchesne E, Mouton PR, Calhoun ME, Semendeferi K, Ahrens-Barbeau C, Hallet MJ, Pierce K. Neuron number and size in prefrontal cortex of children with autism. Journal of the American Medical Association (JAMA) 2011; 306 (18):2001–2010. [ PubMed ] [ Google Scholar ]
  • Coury DL, Swedo SE, Thurm AE, Miller DT, Veenstra-Vanderweele JM, Carbone PS, Taylor JL. Treating the whole person with autism: the proceedings of the autism speaks national autism conference. Current Problems in Pediatric and Adolescent Health Care. 2014; 44 (2):26–47. [ PubMed ] [ Google Scholar ]
  • Coyne JC, Gottlieb BH. The mismeasure of coping by checklist. Journal of Personality. 1996; 64 (4):959–991. [ PubMed ] [ Google Scholar ]
  • Cuthbert BN, Insel TR. Toward the future of psychiatric diagnosis: the seven pillars of RDoC. BMC Medicine. 2013; 11 (1):126. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Dawson G, Jones EJ, Merkle K, Venema K, Lowy R, Faja S, Winter J. Early behavioral intervention is associated with normalized brain activity in young children with autism. Journal of the American Academy of Child & Adolescent Psychiatry. 2012; 51 (11):1150–1159. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Dawson G, Rogers S, Munson J, Smith M, Winter J, Greenson J, Varley J. Randomized, controlled trial of an intervention for toddlers with autism: the Early Start Denver Model. Pediatrics. 2010; 125 (1):e17–e23. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Delorme R, Ey E, Toro R, Leboyer M, Gillberg C, Bourgeron T. Progress toward treatments for synaptic defects in autism. Nature Medicine. 2013; 19 (6):685–694. [ PubMed ] [ Google Scholar ]
  • Desbonnet L, Clarke G, Shanahan F, Dinan T, Cryan J. Microbiota is essential for social development in the mouse. Molecular Psychiatry. 2013; 19 :146–148. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Di Martino A, Ross K, Uddin LQ, Sklar AB, Castellanos FX, Milham MP. Functional brain correlates of social and nonsocial processes in autism spectrum disorders: an activation likelihood estimation meta-analysis. Biological Psychiatry. 2009; 65 (1):63–74. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug development costs. Journal of Health Economics. 2003; 22 (2):151–185. [ PubMed ] [ Google Scholar ]
  • Dinan T, Cryan J. Melancholic microbes: a link between gut microbiota and depression? Neurogastroenterology & Motility. 2013; 25 (9):713–719. [ PubMed ] [ Google Scholar ]
  • Dinstein I, Heeger DJ, Lorenzi L, Minshew NJ, Malach R, Behrmann M. Unreliable evoked responses in autism. Neuron. 2012; 75 (6):981–991. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Dove D, Warren Z, McPheeters ML, Taylor JL, Sathe NA, Veenstra-VanderWeele J. Medications for adolescents and young adults with autism spectrum disorders: a systematic review. Pediatrics. 2012; 130 (4):717–726. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Doyle CA, McDougle CJ, Stigler KA. Adolescents and Adults with Autism Spectrum Disorders. Springer; 2014. Pharmacotherapy of behavioral symptoms and psychiatric comorbidities in adolescents and adults with autism spectrum disorders; pp. 161–191. [ Google Scholar ]
  • Ecker C, Murphy D. Neuroimaging in autism-from basic science to translational research. Nature Reviews Neurology. 2014; 10 :82–91. [ PubMed ] [ Google Scholar ]
  • Ecker C, Suckling J, Deoni SC, Lombardo MV, Bullmore ET, Baron-Cohen S, Bailey AJ. Brain anatomy and its relationship to behavior in adults with autism spectrum disorder: a multicenter magnetic resonance imaging study. Archives of General Psychiatry. 2012; 69 (2):195–209. [ PubMed ] [ Google Scholar ]
  • Eglen R, Reisine T. Primary cells and stem cells in drug discovery: emerging tools for high-throughput screening. Assay and Drug Development Technologies. 2011; 9 (2):108–124. [ PubMed ] [ Google Scholar ]
  • Elsabbagh M, Mercure E, Hudry K, Chandler S, Pasco G, Charman T, Johnson MH. Infant neural sensitivity to dynamic eye gaze is associated with later emerging autism. Current Biology. 2012; 22 (4):338–342. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Faja S, Aylward E, Bernier R, Dawson G. Becoming a face expert: a computerized face-training program for high-functioning individuals with autism spectrum disorders. Developmental Neuropsychology. 2008; 33 (1):1–24. [ PubMed ] [ Google Scholar ]
  • Fein D, Barton M, Eigsti IM, Kelley E, Naigles L, Schultz RT, Rosenthal M. Optimal outcome in individuals with a history of autism. Journal of Child Psychology and Psychiatry. 2013; 54 (2):195–205. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Fombonne E. Epidemiological surveys of autism and other pervasive developmental disorders: an update. Journal of Autism and Developmental Disorders. 2003; 33 (4):365–382. [ PubMed ] [ Google Scholar ]
  • Frieden TR, Jaffe HW, Cono J, Richards CL, Iademarco MF. Prevalence of autism spectrum disorder among children aged 8 years — Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2010. Morbidity and Mortality Weekly Report (MMWR) 2014; 63 :1–21. [ PubMed ] [ Google Scholar ]
  • Geschwind DH. Genetics of autism spectrum disorders. Trends in Cognitive Sciences. 2011; 15 (9):409–416. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Geschwind DH, Levitt P. Autism spectrum disorders: developmental disconnection syndromes. Current Opinion in Neurobiology. 2007; 17 (1):103–111. [ PubMed ] [ Google Scholar ]
  • Gillberg C. The ESSENCE in child psychiatry: early symptomatic syndromes eliciting neurodevelopmental clinical examinations. Research in Developmental Disabilities. 2010; 31 (6):1543–1551. [ PubMed ] [ Google Scholar ]
  • Goin-Kochel RP, Mackintosh VH, Myers BJ. How many doctors does it take to make an autism spectrum diagnosis? Autism. 2006; 10 (5):439–451. [ PubMed ] [ Google Scholar ]
  • Gomot M, Belmonte MK, Bullmore ET, Bernard FA, Baron-Cohen S. Brain hyper-reactivity to auditory novel targets in children with high-functioning autism. Brain. 2008; 131 (9):2479–2488. [ PubMed ] [ Google Scholar ]
  • Gondalia SV, Palombo EA, Knowles SR, Cox SB, Meyer D, Austin DW. Molecular characterisation of gastrointestinal microbiota of children with autism (with and without gastrointestinal dysfunction) and their neurotypical siblings. Autism Research. 2012; 5 (6):419–427. [ PubMed ] [ Google Scholar ]
  • Gordon I. Can oxytocin improve core brain and behavioral features of autism spectrum disorders in children? New Frontiers in Social Neuroscience. 2014:189–199. [ Google Scholar ]
  • Gotham K, Risi S, Pickles A, Lord C. The Autism Diagnostic Observation Schedule: revised algorithms for improved diagnostic validity. Journal of Autism and Developmental Disorders. 2007; 37 (4):613–627. [ PubMed ] [ Google Scholar ]
  • Granholm E, Ben-Zeev D, Fulford D, Swendsen J. Ecological momentary assessment of social functioning in schizophrenia: impact of performance appraisals and affect on social interactions. Schizophrenia Research. 2013; 145 (1-3):120–124. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Hansen NB, Lambert MJ, Forman EM. The psychotherapy dose-response effect and its implications for treatment delivery services. Clinical Psychology: Science and Practice. 2002; 9 (3):329–343. [ Google Scholar ]
  • Harms MB, Martin A, Wallace GL. Facial emotion recognition in autism spectrum disorders: a review of behavioral and neuroimaging studies. Neuropsychology Review. 2010; 20 (3):290–322. [ PubMed ] [ Google Scholar ]
  • Harvey AG, Watkins E, Mansell W, Shafran R. Cognitive behavioural processes across psychological disorders. Oxford University Press; New York: 2004. [ Google Scholar ]
  • Hazlett HC, Poe MD, Gerig G, Styner M, Chappell C, Smith RG, Piven J. Early brain overgrowth in autism associated with an increase in cortical surface area before age 2 years. Archives of General Psychiatry. 2011; 68 (5):467–476. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Hurlburt RT, Happe F, Frith U. Sampling the form of inner experience in three adults with Asperger syndrome. Psychological Medicine, s. 1994;(2):385–395. [ PubMed ] [ Google Scholar ]
  • Insel TR. Next-generation treatments for mental disorders. Science Translational Medicine. 2012; 4 (155):155ps119–155ps119. [ PubMed ] [ Google Scholar ]
  • Insel TR. “A New Approach to Clinical Trials” (Vol. February 27) 2014 Director's Blog: http://www.nimh.nih.gov/about/director/2014/a-new-approach-to-clinical-trials.shtml .
  • Insel TR, Gogtay N. National Institute of Mental Health Clinical Trials: New Opportunities, New Expectations. JAMA Psychiatry. 2014 [ PubMed ] [ Google Scholar ]
  • Javitt DC, Schoepp D, Kalivas PW, Volkow ND, Zarate C, Merchant K, Lee CM. Translating glutamate: from pathophysiology to treatment. Science Translational Medicine. 2011; 3 (102):102. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Jeste SS, Geschwind DH. Disentangling the heterogeneity of autism spectrum disorder through genetic findings. Nature Reviews Neurology. 2014; 10 :74–81. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Jones W, Klin A. Attention to eyes is present but in decline in 2-6-month-old infants later diagnosed with autism. Nature. 2013; 504 (7480):427–431. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Kaminski JW, Valle LA, Filene JH, Boyle CL. A meta-analytic review of components associated with parent training program effectiveness. Journal of Abnormal Child Psychology. 2008; 36 (4):567–589. [ PubMed ] [ Google Scholar ]
  • Kanner L. Autistic disturbances of affective contact. Nervous Child. 1943; 2 (2):217–230. [ PubMed ] [ Google Scholar ]
  • Kapp SK, Gillespie-Lynch K, Sherman LE, Hutman T. Deficit, difference, or both? Autism and neurodiversity. Developmental Psychology. 2013; 49 (1):59. [ PubMed ] [ Google Scholar ]
  • Kennedy DP, Courchesne E. Functional abnormalities of the default network during self-and other-reflection in autism. Social Cognitive and Affective Neuroscience. 2008; 3 (2):177–190. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Khor AS, Gray KM, Reid SC, Melvin GA. Feasibility and validity of ecological momentary assessment in adolescents with high-functioning autism and Asperger's disorder. Journal of Adolescence. 2014; 37 (1):37–46. [ PubMed ] [ Google Scholar ]
  • King BH, Hollander E, Sikich L, McCracken JT, Scahill L, Bregman JD, Ritz L. Lack of efficacy of citalopram in children with autism spectrum disorders and high levels of repetitive behavior: citalopram ineffective in children with autism. Archives of General Psychiatry. 2009; 66 (6):583–590. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Kleinhans NM, Müller R-A, Cohen DN, Courchesne E. Atypical functional lateralization of language in autism spectrum disorders. Brain Research. 2008; 1221 :115–125. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Kleinman JM, Ventola PE, Pandey J, Verbalis AD, Barton M, Hodgson S, Fein D. Diagnostic stability in very young children with autism spectrum disorders. Journal of Autism and Developmental Disorders. 2008; 38 (4):606–615. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Klin A, McPartland J, Volkmar FR. Asperger syndrome. In: Volkmar FR, Paul LK, Klin A, Cohen D, editors. Handbook of Autism and Pervasive Developmental Disorders: Diagnosis, Development, Neurobiology, and Behavior. John Wiley & Sons Inc.; Hoboken, NJ: 2005. pp. 88–125. [ Google Scholar ]
  • Knapp M, Buescher A. Economic aspects of autism. In: Volkmar FR, Paul R, Rogers SJ, Pelphrey KA, editors. Handbook of Autism and Pervasive Developmental Disorders, Diagnosis, Development, and Brain Mechanisms. 4th ed. Vol. 1. John Wiley & Sons, Inc.; Hoboken, NJ: 2014. pp. 1089–1106. [ Google Scholar ]
  • Knaus TA, Silver AM, Lindgren KA, Hadjikhani N, Tager-Flusberg H. fMRI activation during a language task in adolescents with ASD. Journal of the International Neuropsychological Society. 2008; 14 (06):967–979. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Landa RJ, Gross AL, Stuart EA, Bauman M. Latent class analysis of early developmental trajectory in baby siblings of children with autism. Journal of Child Psychology and Psychiatry. 2012; 53 (9):986–996. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Lazic SE, Essioux L. Improving basic and translational science by accounting for litter-to-litter variation in animal models. BMC Neuroscience. 2013; 14 (1):37. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Lecavalier L. Behavioral and emotional problems in young people with pervasive developmental disorders: relative prevalence, effects of subject characteristics, and empirical classification. Journal of Autism and Developmental Disorders. 2006; 36 (8):1101–1114. [ PubMed ] [ Google Scholar ]
  • Legg L, Drummond A, Leonardi-Bee J, Gladman J, Corr S, Donkervoort M, Logan P. Occupational therapy for patients with problems in personal activities of daily living after stroke: systematic review of randomised trials. British Medical Journal. 2007; 335 (7626):922. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Lerner MD, White SW, McPartland JC. Mechanisms of change in psychosocial interventions for autism spectrum disorders. Dialogues in Clinical Neuroscience. 2012; 14 (3):307–318. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Licinio J, Wong M. A novel conceptual framework for psychiatry: vertically and horizontally integrated approaches to redundancy and pleiotropism that co-exist with a classification of symptom clusters based on DSM-5. Molecular Psychiatry. 2013; 18 (8):846–848. [ PubMed ] [ Google Scholar ]
  • Lickel A, MacLean WE, Blakeley-Smith A, Hepburn S. Assessment of the prerequisite skills for cognitive behavioral therapy in children with and without autism spectrum disorders. Journal of Autism and Developmental Disorders. 2012; 42 (6):992–1000. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Limperopoulos C, Bassan H, Sullivan NR, Soul JS, Robertson RL, Moore M, du Plessis AJ. Positive screening for autism in ex-preterm infants: prevalence and risk factors. Pediatrics. 2008; 121 (4):758–765. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Lord C, Jones RM. Annual Research Review: Re-thinking the classification of autism spectrum disorders. Journal of Child Psychology and Psychiatry. 2012; 53 (5):490–509. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Lord C, Risi S, DiLavore PS, Shulman C, Thurm A, Pickles A. Autism from 2 to 9 years of age. Archives of General Psychiatry. 2006; 63 (6):694–701. [ PubMed ] [ Google Scholar ]
  • Louis P. Does the human gut microbiota contribute to the etiology of autism spectrum disorders? Digestive Diseases and Sciences. 2012:1–3. [ PubMed ] [ Google Scholar ]
  • Lovaas OI. Behavioral treatment and normal educational and intellectual functioning in young autistic children. Journal of Consulting and Clinical Psychology. 1987; 55 (1):3. [ PubMed ] [ Google Scholar ]
  • Mandell DS, Novak MM, Zubritsky CD. Factors associated with age of diagnosis among children with autism spectrum disorders. Pediatrics. 2005; 116 (6):1480–1486. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Marhe R, Waters AJ, van de Wetering BJ, Franken IH. Implicit and explicit drug-related cognitions during detoxification treatment are associated with drug relapse: an ecological momentary assessment study. Journal of Consulting and Clinical Psychology. 2013; 81 (1):1–12. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Markou A, Chiamulera C, Geyer MA, Tricklebank M, Steckler T. Removing obstacles in neuroscience drug discovery: the future path for animal models. Neuropsychopharmacology. 2008; 34 (1):74–89. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Mason J, Scior K. ‘Diagnostic overshadowing'amongst clinicians working with people with intellectual disabilities in the UK. Journal of Applied Research in Intellectual Disabilities. 2004; 17 (2):85–90. [ Google Scholar ]
  • Mazefsky CA, Herrington J, Siegel M, Scarpa A, Maddox BB, Scahill L, White SW. The role of emotion regulation in autism spectrum disorder. Journal of the American Academy of Child & Adolescent Psychiatry. 2013; 52 (7):679–688. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Mazefsky CA, Oswald DP, Day TN, Eack SM, Minshew NJ, Lainhart JE. ASD, a psychiatric disorder, or both? Psychiatric diagnoses in adolescents with high-functioning ASD. Journal of Clinical Child and Adolescent Psychology. 2012; 41 (4):516–523. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Mazefsky CA, White SW. Adults with autism. In: Volkmar FR, Paul R, Rogers SJ, Pelphrey KA, editors. Handbook of Autism and Pervasive Developmental Disorders: Diagnosis Development, and Brain Mechanisms. 4th ed. Vol. 1. John Wiley & Sons, Inc.; Hoboken, NJ: 2014. pp. 191–211. [ Google Scholar ]
  • McClure I. Developing and implementing practice guidelines. In: Volkmar FR, Paul R, Rogers SJ, Pelphrey KA, editors. Handbook of Autism and Pervasive Developmental Disorders: Diagnosis, Development, and Brain Mechanisms. 4th ed. Vol. 1. John Wiley & Sons, Inc.; Hoboken, NJ: 2014. pp. 1014–1035. [ Google Scholar ]
  • McConachie H, Diggle T. Parent implemented early intervention for young children with autism spectrum disorder: a systematic review. Journal of Evaluation in Clinical Practice. 2007; 13 (1):120–129. [ PubMed ] [ Google Scholar ]
  • McCray AT, Trevvett P, Frost HR. Modeling the autism spectrum disorder phenotype. Neuroinformatics. 2013; 12 (2):291–305. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • McGovern CW, Sigman M. Continuity and change from early childhood to adolescence in autism. Journal of Child Psychology and Psychiatry. 2005; 46 (4):401–408. [ PubMed ] [ Google Scholar ]
  • McPartland JC, Coffman M, Pelphrey KA. Recent advances in understanding the neural bases of autism spectrum disorder. Current Opinion in Pediatrics. 2011; 23 (6):628–632. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Messinger D, Young GS, Ozonoff S, Dobkins K, Carter A, Zwaigenbaum L, Constantino JN. Beyond autism: a baby siblings research consortium study of high-risk children at three years of age. Journal of the American Academy of Child & Adolescent Psychiatry. 2013; 52 (3):300–308. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Miles JH. Autism spectrum disorders—a genetics review. Genetics in Medicine. 2011; 13 (4):278–294. [ PubMed ] [ Google Scholar ]
  • Minshew NJ, Keller TA. The nature of brain dysfunction in autism: functional brain imaging studies. Current Opinion in Neurology. 2010; 23 (2):124. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Minshew NJ, Williams DL. The new neurobiology of autism: cortex, connectivity, and neuronal organization. Archives of Neurology. 2007; 64 (7):945–950. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Miyake K, Hirasawa T, Koide T, Kubota T. Epigenetics in autism and other neurodevelopmental diseases. Advances in Experimental Medicine and Biology. 2012; 724 :91–98. [ PubMed ] [ Google Scholar ]
  • Moghimi S, Kushki A, Marie Guerguerian A, Chau T. A review of EEG-based brain-computer interfaces as access pathways for individuals with severe disabilities. Assistive Technology. 2013; 25 (2):99–110. [ PubMed ] [ Google Scholar ]
  • Moy SS, Nadler JJ. Advances in behavioral genetics: mouse models of autism. Molecular Psychiatry. 2008; 13 (1):4–26. [ PubMed ] [ Google Scholar ]
  • Moy SS, Nadler JJ, Magnuson TR, Crawley JN. Mouse models of autism spectrum disorders: the challenge for behavioral genetics. American Journal of Medical Genetics Part C Seminars in Medical Genetics. 2006; 142C (1):40–51. doi: 10.1002/ajmg.c.30081. [ PubMed ] [ Google Scholar ]
  • Mulle JG, Sharp WG, Cubells JF. The gut microbiome: a new frontier in autism research. Current Psychiatry Reports. 2013; 15 (2):1–9. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Müller R-A, Kleinhans N, Kemmotsu N, Pierce K, Courchesne E. Abnormal variability and distribution of functional maps in autism: an FMRI study of visuomotor learning. American Journal of Psychiatry. 2003; 160 (10):1847–1862. [ PubMed ] [ Google Scholar ]
  • Myin-Germeys I, Oorschot M, Collip D, Lataster J, Delespaul P, van Os J. Experience sampling research in psychopathology: opening the black box of daily life. Psychological Medicine. 2009; 39 (9):1533–1547. [ PubMed ] [ Google Scholar ]
  • National Institute for Health and Clinical Excellence (NICE) [June 30, 2014]; Autism: recognition, referral and diagnosis of children and young people on the autism spectrum. 2011 from http://www.nice.org.uk/guidance/CG128 .
  • National Research Council . Educating Children with Autism. Committee on Educational Interventions for Children with Autism. Division of Behavioral and Social Sciences and Education. National Academy Press; Washington, DC: 2001. [ Google Scholar ]
  • Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nature Neuroscience. 2010; 13 (10):1161–1169. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Oddi D, Crusio WE, D'Amato FR, Pietropaolo S. Monogenic mouse models of social dysfunction: implications for autism. Behavioural Brain Research. 2013; 251 :75–84. [ PubMed ] [ Google Scholar ]
  • Ozonoff S, Iosif A-M, Baguio F, Cook IC, Hill MM, Hutman T, Sigman M. A prospective study of the emergence of early behavioral signs of autism. Journal of the American Academy of Child & Adolescent Psychiatry. 2010; 49 (3):256–266. e252. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Parracho HM, Bingham MO, Gibson GR, McCartney AL. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. Journal of Medical Microbiology. 2005; 54 (10):987–991. [ PubMed ] [ Google Scholar ]
  • Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR, Schacht AL. How to improve R&D productivity: the pharmaceutical industry's grand challenge. Nature Reviews Drug Discovery. 2010; 9 (3):203–214. [ PubMed ] [ Google Scholar ]
  • Pelphrey KA, Shultz S, Hudac CM, Vander Wyk BC. Research review: constraining heterogeneity: the social brain and its development in autism spectrum disorder. Journal of Child Psychology and Psychiatry. 2011; 52 (6):631–644. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Philip R, Dauvermann MR, Whalley HC, Baynham K, Lawrie SM, Stanfield AC. A systematic review and meta-analysis of the fMRI investigation of autism spectrum disorders. Neuroscience & Biobehavioral Reviews. 2012; 36 (2):901–942. [ PubMed ] [ Google Scholar ]
  • Piven J, Rabins P. Autism spectrum disorders in older adults: toward defining a research agenda. Journal of the American Geriatrics Society. 2011; 59 (11):2151–2155. [ PubMed ] [ Google Scholar ]
  • Reaven J, Blakeley-Smith A, Culhane-Shelburne K, Hepburn S. Group cognitive behavior therapy for children with high-functioning autism spectrum disorders and anxiety: A randomized trial. Journal of Child Psychology and Psychiatry. 2012; 53 (4):410–419. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Redcay E, Courchesne E. When is the brain enlarged in autism? A meta-analysis of all brain size reports. Biological Psychiatry. 2005; 58 (1):1–9. [ PubMed ] [ Google Scholar ]
  • Redcay E, Courchesne E. Deviant functional magnetic resonance imaging patterns of brain activity to speech in 2–3-year-old children with autism spectrum disorder. Biological Psychiatry. 2008; 64 (7):589–598. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Reichow B. Overview of meta-analyses on early intensive behavioral intervention for young children with autism spectrum disorders. Journal of autism and developmental disorders. 2012; 42 (4):512–520. [ PubMed ] [ Google Scholar ]
  • Reichow B, Steiner AM, Volkmar F. Cochrane Review: Social skills groups for people aged 6 to 21 with autism spectrum disorders (ASD). Evidence-Based Child Health: A Cochrane Review Journal. 2013; 8 (2):266–315. [ PubMed ] [ Google Scholar ]
  • Rice C. Prevalence of autism spectrum disorders: Autism and Developmental Disabilities Monitoring Network, United States, 2006. Morbidity and Mortality Weekly Report. Surveillance Summaries. Volume 58, Number SS-10. Centers for Disease Control and Prevention. 2009 [ PubMed ] [ Google Scholar ]
  • Ronald A, Hoekstra RA. Autism spectrum disorders and autistic traits: a decade of new twin studies. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics. 2011; 156B (3):255–274. [ PubMed ] [ Google Scholar ]
  • Rutter M. Comorbidity: concepts, claims and choices. Criminal Behaviour and Mental Health. 1997; 7 (4):265–285. [ Google Scholar ]
  • Rutter M, Kim-Cohen J, Maughan B. Continuities and discontinuities in psychopathology between childhood and adult life. Journal of Child Psychology and Psychiatry. 2006; 47 (3-4):276–295. [ PubMed ] [ Google Scholar ]
  • Salmond CH, Vargha-Khadem F, Gadian DG, de Haan M, Baldeweg T. Heterogeneity in the patterns of neural abnormality in autistic spectrum disorders: evidence from ERP and MRI. Cortex. 2007; 43 (6):686–699. [ PubMed ] [ Google Scholar ]
  • Scahill L, McCracken JT, Bearss K, Robinson F, Hollander E, King B, Wagner A. Design and subject characteristics in the federally-funded citalopram trial in children with pervasive developmental disorders. Journal of Autism and Developmental Disorders. 2012; 42 (3):432–440. [ PubMed ] [ Google Scholar ]
  • Schwartz JE, Neale J, Marco C, Shiffman SS, Stone AA. Does trait coping exist? A momentary assessment approach to the evaluation of traits. Journal of Personality and Social Psychology. 1999; 77 (2):360–369. [ PubMed ] [ Google Scholar ]
  • Scott G. The Neurotechnology Revolution Has Arrived. The Futurist. 2013; 47 (6-7) [ Google Scholar ]
  • Scottish Intercollegiate Guidelines Network (SIGN) [June 30, 2014]; Assessment, Diagnosis and Clinical Interventions for Children and Young People with Autism Spectrum Disorders. 2007 from http://www.sign.ac.uk .
  • Seida JK, Ospina MB, Karkhaneh M, Hartling L, Smith V, Clark B. Systematic reviews of psychosocial interventions for autism: an umbrella review. Developmental Medicine & Child Neurology. 2009; 51 (2):95–104. [ PubMed ] [ Google Scholar ]
  • Shane HC, Albert PD. Electronic screen media for persons with autism spectrum disorders: results of a survey. Journal of Autism and Developmental Disorders. 2008; 38 (8):1499–1508. [ PubMed ] [ Google Scholar ]
  • Shen MD, Nordahl CW, Young GS, Wootton-Gorges SL, Lee A, Liston SE, Amaral DG. Early brain enlargement and elevated extra-axial fluid in infants who develop autism spectrum disorder. Brain. 2013; 136 (9):2825–2835. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Sherer MR, Schreibman L. Individual behavioral profiles and predictors of treatment effectiveness for children with autism. Journal of Consulting and Clinical Psychology. 2005; 73 (3):525–525. [ PubMed ] [ Google Scholar ]
  • Shiffman S, Stone AA, Hufford MR. Ecological momentary assessment. Annual Review of Clinical Psychology. 2008; 4 :1–32. [ PubMed ] [ Google Scholar ]
  • Simonoff E, Jones CR, Baird G, Pickles A, Happé F, Charman T. The persistence and stability of psychiatric problems in adolescents with autism spectrum disorders. Journal of Child Psychology and Psychiatry. 2013; 54 (2):186–194. [ PubMed ] [ Google Scholar ]
  • Smith LE, Maenner MJ, Seltzer MM. Developmental trajectories in adolescents and adults with autism: The case of daily living skills. Journal of the American Academy of Child & Adolescent Psychiatry. 2012; 51 (6):622–631. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Stahmer AC, Schreibman L, Cunningham AB. Toward a technology of treatment individualization for young children with autism spectrum disorders. Brain Research. 2011; 1380 :229–239. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • State MW, Levitt P. The conundrums of understanding genetic risks for autism spectrum disorders. Nature Neuroscience. 2011; 14 (12):1499–1506. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour–epigenetic regulation of the gut–brain axis. Genes, Brain and Behavior. 2014; 13 (1):69–86. [ PubMed ] [ Google Scholar ]
  • Stone AA, Schwartz JE, Neale JM, Shiffman S, Marco CA, Hickcox M, Cruise LJ. A comparison of coping assessed by ecological momentary assessment and retrospective recall. Journal of Personality and Social Psychology. 1998; 74 (6):1670–1680. [ PubMed ] [ Google Scholar ]
  • Stone AA, Shiffman S. Capturing momentary, self-report data: a proposal for reporting guidelines. Annals of Behavioral Medicine. 2002; 24 (3):236–243. [ PubMed ] [ Google Scholar ]
  • Stoner R, Chow ML, Boyle MP, Sunkin SM, Mouton PR, Roy S, Courchesne E. Patches of disorganization in the neocortex of children with autism. New England Journal of Medicine. 2014; 370 (13):1209–1219. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Sutera S, Pandey J, Esser EL, Rosenthal MA, Wilson LB, Barton M, Dumont-Mathieu T. Predictors of optimal outcome in toddlers diagnosed with autism spectrum disorders. Journal of Autism and Developmental Disorders. 2007; 37 (1):98–107. [ PubMed ] [ Google Scholar ]
  • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126 (4):663–676. [ PubMed ] [ Google Scholar ]
  • Tan PZ, Forbes EE, Dahl RE, Ryan ND, Siegle GJ, Ladouceur CD, Silk JS. Emotional reactivity and regulation in anxious and nonanxious youth: a cell-phone ecological momentary assessment study. Journal of Child Psychology and Psychiatry. 2012; 53 (2):197–206. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Taylor JL, McPheeters ML, Sathe NA, Dove D, Veenstra-VanderWeele J, Warren Z. A systematic review of vocational interventions for young adults with autism spectrum disorders. Pediatrics. 2012; 130 (3):531–538. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Taylor JL, Seltzer MM. Changes in the autism behavioral phenotype during the transition to adulthood. Journal of Autism and Developmental Disorders. 2010; 40 (12):1431–1446. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Toal F, Daly E, Page L, Deeley Q, Hallahan B, Bloemen O, Robertson D. Clinical and anatomical heterogeneity in autistic spectrum disorder: a structural MRI study. Psychological Medicine. 2010; 40 (07):1171–1181. [ PubMed ] [ Google Scholar ]
  • Turner LM, Stone WL. Variability in outcome for children with an ASD diagnosis at age 2. Journal of Child Psychology and Psychiatry. 2007; 48 (8):793–802. [ PubMed ] [ Google Scholar ]
  • Vaccarino FM, Urban AE, Stevens HE, Szekely A, Abyzov A, Grigorenko EL, Weissman S. Annual Research Review: The promise of stem cell research for neuropsychiatric disorders. Journal of Child Psychology and Psychiatry. 2011; 52 (4):504–516. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Van Hecke AV, Stevens S, Carson AM, Karst JS, Dolan B, Schohl K, Brockman S. Measuring the plasticity of social approach: A randomized controlled trial of the effects of the PEERS intervention on EEG asymmetry in adolescents with autism spectrum disorders. Journal of Autism and Developmental Disorders. 2013:1–20. [ PubMed ] [ Google Scholar ]
  • Virués-Ortega J. Applied behavior analytic intervention for autism in early childhood: Meta-analysis, meta-regression and dose–response meta-analysis of multiple outcomes. Clinical Psychology Review. 2010; 30 (4):387–399. [ PubMed ] [ Google Scholar ]
  • Virues-Ortega J, Julio FM, Pastor-Barriuso R. The TEACCH program for children and adults with autism: A meta-analysis of intervention studies. Clinical Psychology Review. 2013; 33 (8):940–953. [ PubMed ] [ Google Scholar ]
  • Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, Geschwind DH. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 2011; 474 (7351):380–384. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Volkmar F, Cook E, Jr, Pomeroy J, Realmuto G, Tanguay P. Practice parameters for the assessment and treatment of children, adolescents, and adults with autism and other pervasive developmental disorders. American Academy of Child and Adolescent Psychiatry Working Group on Quality Issues. Journal of the American Academy of Child and Adolescent Psychiatry. 1999; 38 (12 Suppl):32S. [ PubMed ] [ Google Scholar ]
  • Voos AC, Pelphrey KA, Tirrell J, Bolling DZ, Vander Wyk B, Kaiser MD, Ventola P. Neural mechanisms of improvements in social motivation after pivotal response treatment: two case studies. Journal of autism and developmental disorders. 2013; 43 (1):1–10. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Wagner JA. Strategic approach to fit-for-purpose biomarkers in drug development. Annual Review of Pharmacology and Toxicology. 2008; 48 :631–651. doi: 10.1146/annurev.pharmtox.48.113006.094611. [ PubMed ] [ Google Scholar ]
  • Walker CK, Anderson KW, Milano KM, Ye S, Tancredi DJ, Pessah IN, Kliman HJ. Trophoblast inclusions are significantly increased in the placentas of children in families at risk for autism. Biological Psychiatry. 2013; 74 (3):204. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Warren Z, McPheeters ML, Sathe N, Foss-Feig JH, Glasser A, Veenstra-VanderWeele J. A systematic review of early intensive intervention for autism spectrum disorders. Pediatrics. 2011; 127 (5):e1303–e1311. [ PubMed ] [ Google Scholar ]
  • Waterhouse L, Gillberg C. Why autism must be taken apart. Journal of Autism and Developmental Disorders. 2014 doi: 10.1007/s10803-013-2030-5. [ PubMed ] [ Google Scholar ]
  • Wetherby AM, Woods J, Allen L, Cleary J, Dickinson H, Lord C. Early indicators of autism spectrum disorders in the second year of life. Journal of autism and developmental disorders. 2004; 34 (5):473–493. [ PubMed ] [ Google Scholar ]
  • Whalen CK, Jamner LD, Henker B, Delfino RJ. Smoking and moods in adolescents with depressive and aggressive dispositions: evidence from surveys and electronic diaries. Health Psychology. 2001; 20 (2):99–111. [ PubMed ] [ Google Scholar ]
  • White SW, Ollendick T, Albano AM, Oswald D, Johnson C, Southam-Gerow MA, Scahill L. Randomized controlled trial: multimodal anxiety and social skill intervention for adolescents with autism spectrum disorder. Journal of Autism and Developmental Disorders. 2013; 43 (2):382–394. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • White SW, Schry AR, Miyazaki Y, Ollendick TH, Scahill L. Effects of verbal ability and severity of autism on anxiety in adolescents with ASD: one-year follow-up after cognitive behavioral therapy. Journal of Clinical Child & Adolescent Psychology. 2014 [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Wolff JJ, Gu H, Gerig G, Elison JT, Styner M, Gouttard S, Estes AM. Differences in white matter fiber tract development present from 6 to 24 months in infants with autism. American Journal of Psychiatry. 2012; 169 (6):589–600. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Young GS, Merin N, Rogers SJ, Ozonoff S. Gaze behavior and affect at 6 months: predicting clinical outcomes and language development in typically developing infants and infants at risk for autism. Developmental Science. 2009; 12 (5):798–814. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Zikopoulos B, Barbas H. Changes in prefrontal axons may disrupt the network in autism. The Journal of Neuroscience. 2010; 30 (44):14595–14609. [ PMC free article ] [ PubMed ] [ Google Scholar ]

Master Thesis Project Fills Gap in Autism Research

graphic of scattered letters, with six in the middle spelling autism

By Molly Loughman

As a graduate student in Communication Sciences & Disorders , Kaya LeGrand, MS ’20 spearheaded a research project to help answer one of many questions surrounding Autism Spectrum Disorder (ASD): Which features of autistic children’s language are most useful for their adult language ability?

“What is essential there? Is it the ability to combine words? To use specific types of words? To use social language?” LeGrand asks, through her master’s thesis project. “To our knowledge, we are the first people to have looked into this question.”

Kaya LeGrand head shot

Now a speech language pathologist, LeGrand reflects back on her final year at Emerson, a tireless pursuit that ultimately shifted her career trajectory toward research and teaching. “I knew I wanted to do something about language and autism because that was clinically what I was becoming more and more interested in throughout the program,” says LeGrand. 

She received inspiration, advice, and research contributions from Associate Professor Rhiannon Luyster and Lisa Wisman Weil , CSD’s senior scholar-in-residence and Undergraduate Program director. The collaborative effort was based out of the Lab for Infant + Toddler Language at Emerson ( LI+TLE ), where researchers study early language and communication in children. Launched in March 2019, the project came to fruition in June 2021 with the publication of “Identifying Childhood Expressive Language Features That Best Predict Adult Language and Communication Outcome in Individuals with Autism Spectrum Disorder ,” co-authored by LeGrand, Luyster, Wisman Weil and UCLA’s Catherine Lord, in the Journal of Speech, Language and Hearing Research .  

THE RESEARCH

LeGrand’s project addressed vague findings first reported in the 1950s— that “the presence of useful speech by age five in kids with autism is very important for later language outcomes.” 

“We didn’t know what ‘useful speech’ meant; it wasn’t very well defined initially, and then every researcher who replicated that finding either defined it a different way or also kind of left it vague,” says LeGrand. 

To clarify the field’s understanding, the team examined a longitudinal data set collected by Lord, who has been studying nearly 200 individuals with autism from age 2, starting in the mid-90s until present day. LeGrand studied  hours of footage from a sample of 29 participants, analyzing their noun and verb diversity (the number of different nouns and verbs a child used), the length of spoken sentences, the ability to combine words, and the proportion of speech motivated by social interaction—all to understand which features best predicted adult language.

LeGrand transcribed and coded footage with help from other graduate and undergraduate students. Lord reviewed the final study; Luyster, Wisman-Weil, and LeGrand devised the coding scheme. Wisman-Weil mentored LeGrand on transcribing and language sampling analysis. And as research supervisor, Luyster acted as a sounding board for any of LeGrand’s thesis problems or ideas.

“I really couldn’t have done this without the help of everyone who was involved,” says LeGrand. “ I really respect Lisa and Rhiannon and the whole faculty. I was able to get close to them during the process. They both have done so much amazing research on  language and autism, so it’s very cool to learn from them. And it was also very cool to work with Cathy, one of the foremost researchers in the field.”

THE FINDING: LeGrand found that the number of different verbs produced by children with ASD is the best predictor of adult language outcomes, followed by the length of sentences they form.

“It’s hard to draw strong conclusions just from one study. Basically, we think that verbs are important and it’s also important to use a variety of verbs — not just to have a large vocabulary or combine words,” says LeGrand. “You need a verb to make a sentence, so technically, they are very important for overall language outcomes.” 

LeGrand’s master thesis was completed in April 2020 and submitted for publication that fall. “I really enjoy organizing data, so this worked out for me. I realized that I much prefer doing research compared to clinical work.​​ It was so helpful to have all the lab community support me throughout the project. It was a nice collaborative experience.” 

LeGrand will begin a PhD program at the University of Connecticut this August. She plans to eventually become a professor researching minimally verbal and nonverbal individuals with autism.

“It is a very understudied group. I think it might be challenging to get into given there is not much research done already in that area.” 

News & Stories

Alumni , Communication Disorders , School of Communication

' src=

Molly Loughman View All

Leave a reply cancel reply.

You must be logged in to post a comment.

IMAGES

  1. Autism Awareness Day

    autism research project ideas

  2. The Autism Helper

    autism research project ideas

  3. 8 Creative Sensory Crafts for Children With Autism · The Inspiration Edit

    autism research project ideas

  4. Introducing your autistic child to art can help them in many ways

    autism research project ideas

  5. 25 Autism Awareness ideas in 2021

    autism research project ideas

  6. Our Research

    autism research project ideas

VIDEO

  1. New Autism Research

  2. Autism Research Project

  3. BBA Project Ideas: Unique & Creative Topics for Final Year Students

  4. Research project ideas for high school students! #researchproject #highschooltips #highschooladvice

COMMENTS

  1. Autism Research Activities and Advances | NICHD - Eunice ...

    NICHD's autism spectrum disorder (ASD) research portfolio is spread throughout the Institute and includes extramural components that support research on ASD and other intellectual and developmental disabilities (IDDs).

  2. Guidelines for conducting research studies with the autism ...

    Our research builds on this emerging body of work by providing succinct and practical recommendations that researchers should take into account when conducting research with the autism community, in order to foster an ethos guided more by participatory research principles.

  3. Research topics: Autism | Department of Educational Psychology

    These disorders are characterized, in varying degrees, by difficulties in social interaction, verbal and nonverbal communication and repetitive behaviors. Research in the Department of Educational Psychology focuses on early identification, prevention measures, and interventions related to ASD.

  4. Making the future together: Shaping autism research through ...

    In the context of a large and growing body of autism research, with continued poor implementation, and some evidence of community dissatisfaction, there is a powerful case for participatory autism research.

  5. Autism and Developmental Disorders Research Program

    Our main research aim is to better understand the basic neurobiology of autism and other developmental disorders while furthering our understanding of how genetic and environmental factors may contribute to the onset and progression of these disorders.

  6. ARI-Funded Research Studies 2022 - Autism Research Institute

    Research has the potential to improve the health and well-being of autistic people. Learn about ARI's 2022 research grant recipients. In 2022, ARI awarded more than $450,000 in grants to fund innovative research that holds realistic promise in impacting the lives of autistic people.

  7. Research, Clinical, and Sociological Aspects of Autism

    As the definition of autism has subtly changed over the years, so ideas and trends in autism research have waxed and waned. The focus on psychology and behaviour as core descriptive features of autism has, in many respects, guided research and clinical views and opinions about the condition.

  8. Research Projects - National Autism Center at May Institute

    The National Autism Center at May Institute engages in research to identify, evaluate, and disseminate effective practices to better support people with autism.

  9. Future Directions for Research in Autism Spectrum Disorders

    This article suggests future directions for research aimed at improved understanding of the etiology and pathophysiology of autism spectrum disorder (ASD) as well as pharmacologic and psychosocial interventions for ASD across the lifespan.

  10. Master Thesis Project Fills Gap in Autism Research

    As a graduate student in Communication Sciences & Disorders, Kaya LeGrand, MS ’20 spearheaded a research project to help answer one of many questions surrounding Autism Spectrum Disorder (ASD): Which features of autistic childrens language are most useful for their adult language ability?