The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ReviewsFull Access

Biomarkers in Autism Spectrum Disorder: Challenges, Advances, and the Need for Biomarkers of Relevance to Public Health

Abstract

Although autism spectrum disorder (ASD) is the most strongly genetic of all complex neuropsychiatric disorders, it is still defined and diagnosed behaviorally. The vast genotypic and phenotypic heterogeneity of the condition necessitate a vigorous search for biological markers capable of aiding in diagnosis, identifying more homogeneous subgroups for biological study, individualizing treatment, and measuring treatment response. Many candidate biomarkers are available, spanning genetic, metabolic, electroencephalographic, magnetic resonance imaging, and neuropsychological methods. Although biomarker research has focused primarily on mechanistic etiologic hypotheses, the biomarkers more likely to result in optimized clinical outcomes in the near term are cost-effective and community-viable measures obtained through eye-tracking technology involving infants and toddlers. Although these tools are still far from being ready for widespread application, the goal is to develop objective procedures and measures for population-based screening and diagnosis to increase access to early treatment and intervention.

Autism spectrum disorder (ASD) refers to a highly complex family of conditions defined by early-onset impairments in social interaction and social communication, accompanied by a wide range of behavior rigidities (1), with lifetime consequences for language and learning skills, independent-living skills, and, potentially, the presence of severe behavior challenges (2, 3). It is one of the most highly heritable of all complex neuropsychiatric conditions (4), but no single molecular marker defines its diagnosis. Instead, current estimates suggest that hundreds of genetic and genomic disorders (5)—the majority of which are still unknown—may play a role in etiology, including rare and common variants (6, 7). No single gene has yet been associated with more than a fraction of patient cases (<1% [8]), and the extent to which any pattern or patterns of gene variants or expression can reliably indicate risk of the condition remains unclear.

Biomarkers in Autism Spectrum Disorder: The Need

There are numerous hoped-for future insights into the developmental neurobiology of ASD (9), but the condition is still diagnosed behaviorally by the presence of its defining characteristics, through direct behavioral examination and historical information (3). There is, however, vast phenotypic heterogeneity, spanning the entire range of IQ and language function, with variable profiles of strengths and deficits as well as symptom characteristics; change over time; and comorbidities with common psychiatric conditions (e.g., anxiety, mood, and attentional disorders) (10). The most robust clinical features for early diagnosis of autism include reduced interaction with and attention to others, reduced attention to others’ eyes, failure to respond to the calling of one’s own name, and inability to join in imitative games and reciprocal vocalizations (1113). These indicators are all failures in normative skills that represent milestones in the development of social interaction and social communication skills. Such failures, in turn, become causative factors in subsequent atypical developmental trajectories and in the emergence of more severe symptomatology (14).

Given these multiple layers of complexity, there has been great interest and investment in the identification of biological markers (or biomarkers) for ASD, with the hope of identifying more homogeneous groups for biological study, of aiding in diagnosis (including early detection prior to the emergence or exacerbation of symptoms), and of developing more robust and sensitive markers for individualization of treatment and for measurement of treatment response (1520). This effort is not distinct from the search for biomarkers in other neuropsychiatric conditions, in which there is an emerging consensus that clinical phenomenology, although still the primary means for classifying individuals into diagnostic categories, does not capture biologically meaningful differentiations (2123).

Although ASD is a strongly genetic condition, it is important to note that biomarkers need not necessarily be genetically based, familial, or trait dependent (attributes of a biomarker that is also a “developmental endophenotype” [15]). A biomarker candidate can be defined as such if it is a biological variable associated with the “disease” condition and measurable directly in a given patient or in the patient’s biomaterials through sensitive and reliable quantitative procedures (15). As such, candidate biomarkers also need not be measurements resulting from analysis of biosamples (e.g., urine, blood); they can be measurements of behavior, neuropsychological performance, or brain function.

Another important consideration in biomarker research is that the term is used as a tool to capture biological states that may serve a wide range of purposes; indeed, it is likely that there are different biomarkers for different purposes, in regard to ASD as in regard to most neuropsychiatric disorders. The Foundation for the National Institutes of Health has been a major scientific agent in defining evidentiary criteria to support the regulatory acceptance of biomarker use for a wide range of purposes, such as drug-development programs (24). These purposes may relate to indicators of susceptibility or risk, diagnosis or diagnostic subsets, change, prognosis, and prediction of treatment response (including pharmacodynamic response), among others. In ASD, despite major advances in biomarker research in multiple domains—involving, for example, genes and gene expression, metabolism, multimethod neuroimaging, and behavioral experiments using sophisticated technologies and paradigms—no candidate ASD biomarker for any of the referred purposes has as yet been identified that meets the Foundation for the National Institutes of Health’s evidentiary criteria for clinical or preclinical usage or that has gained a measure of clinical utility or viability (18).

Biomarkers in ASD: The Challenges

The vast genotypic heterogeneity and complexity found among individuals with ASD tempers hopes for a near-term genetic biomarker that might be used as a signature for the condition at the level of the individual patient. Researchers have attempted genotypic subsampling by deploying a genotype-first approach in the study of phenotypic variability in ASD—involving large numbers of individuals sharing recurrent genetic variants known to increase the risk of developing ASD. The results to date have been modest, however, with resultant phenotypes still varying greatly. For example, in a large study of individuals ascertained for the 16p11.2 duplication, previously found to be associated with ASD, behavioral phenotypes ranged from asymptomatic presentation to significant disability, with only a small minority meeting criteria for ASD. Neuropsychiatric symptomatology of affected individuals was predominantly within the realms of intellectual disability, motor delays, and ADHD among children and of anxiety among adults (25). Although these findings are emblematic of the challenges in this line of research, they are not unexpected given the many obstacles resulting from pleiotropy, variable penetrance, and the likely variable additive burden accruing from multiple small-effect risk alleles and interactions thereof, among many others (26).

The vast phenotypic heterogeneity of ASD also accounts for why the identification of biomarkers for the condition has proved to be so elusive. Researchers have primarily focused on mapping biomarkers onto clinically defined categories, but the categories do not capture the multidimensional and complex clinical, cognitive, and behavioral phenotype associated with ASD and its overlap with other disorders (18). Additionally, developmentally invariant biomarkers for ASD are particularly challenging, because the phenotypic manifestations unfold over time, especially in early childhood, reflecting dynamic developmental interactions among multiple risk factors (27). Phenotypic heterogeneity is also a significant hurdle in regard to replicability and generalizability of findings emerging from studies involving small sample sizes or samples of convenience. Population-based studies involving biological or neuropsychological variables, which could achieve greater coverage of the phenotypic spectrum as well as statistical power, are still quite scarce. These examples typify a wide array of challenges slowing down advances toward reproducible ASD biomarkers, which can be briefly summarized in the following general categories.

Sensitivity

A wide range of proposed biomarkers—ranging from profiles in gene expression, proteomics, metabolomics, brain size, structure and connectivity, and oculomotor measures (2834)—have been found not to be universal. They display low levels of sensitivity and have failed, in fact, to positively identify the majority of the various samples studied.

Specificity

Many of the proposed biomarkers are also associated with neuropsychiatric conditions other than ASD, which thus compromises the specificity of the given measure. Probably the most important example in this category is the oldest of all candidate biomarkers in ASD: elevated whole-blood serotonin (20, 35). This is hardly surprising given that many genetic mutations implicated in autism have been shown to play a role not only in other neuropsychiatric conditions but also in other nonpsychiatric medical conditions, such as congenital heart disease (36) and cancer (37).

Relevance to Individual Patients

For most purposes outlined for biomarker research, clinical relevance can only be considered if some criteria are met. Biomarkers need to be shown to perform well for individual patients not only in terms of accuracy (sensitivity and specificity) but also in terms of stability over time, precision, cost-effectiveness, and viability (18). In other words, biomarkers that, at the individual patient level, are too noisy (i.e., contain intraindividual measurement error or variability), are too expensive or too laborious (which makes them impractical in clinical practice), or require special technology or very high levels of technical expertise (which makes them inaccessible beyond specialized research sites) are all unlikely to attain clinical relevance until these challenges are fully addressed. Unless actionable biological mechanisms are identified that promote specific and more effective treatments, biomarkers need to be judged against current gold standards, which still are expert-clinician diagnostic, prescriptive, and prognostic procedures. Implementation variables such as cost, time, and access are of equal importance.

Collectively, these considerations raise the possibility that ASD biomarkers might be identified for some subgroups but not be applicable to others; that their utility might be established along more specific clinically or biologically meaningful dimensions; and that different biomarkers might emerge for specific age groups and for very specific purposes directly tied to a given intervention, be it behavioral or pharmacological.

Biomarkers in ASD: Examples and Progress

Until fairly recently, most biomarker research in ASD focused almost exclusively on the goal of segregating individuals with ASD from other populations—the diagnostic purpose. Nevertheless, there is now a concerted effort to generate ASD biomarkers specifically for clinical trials (38) and for mechanistic studies of biological states (39). In addition, most research has focused on cross-diagnostic group results, failing to involve large enough samples to meaningfully probe performance accuracy at the individual patient level. This situation has been changing markedly in the past decade, with some studies beginning to yield encouraging results (15, 19).

Given that ASD is one of the most strongly genetic of all neuropsychiatric conditions (40), it is little surprise that ASD research has generated a large number of candidate genetic biomarkers (41). Genomewide association studies have identified, with replication, de novo variations that are strongly implicated in ASD (42, 43). Several commercial operations are already marketing genetic testing for ASD on the basis of clusters of genes associated with ASD risk and gene-expression profiles. To be sure, genetic testing is fully established as an important element of “standard of care,” with tangible benefits to patients (44, 45) and results that may be helpful in clinical management of ASD. No genetic biomarker, however, has yet been shown to be ready for screening, diagnosis, or treatment of ASD (46) beyond the discrete examples of well-known, highly penetrant genetic disorders (e.g., Rett syndrome, fragile X syndrome, tuberous sclerosis, and neurofibromatosis) (18).

The most established search for ASD biomarkers, however, has been in biochemical measurements (19). As noted, hyperserotonemia has consistently been recorded in 25%−41% of individuals with ASD (47). The pursuit of a biochemical marker such as this has engendered a great deal of clinical excitement, because such findings could lead to targeted treatments addressing the metabolic imbalance directly. This hope, however, has not yet yielded results robust enough to affect clinical practice, and psychopharmacological treatment of ASD is still symptom based, not biomarker based (48). As a sign of what the future holds, however, metabolomics as a tool for discovery of ASD biomarkers, which uses a broad range of metabolites, has already yielded high diagnostic accuracy in some studies (49). It remains to be seen whether such panels retain their sensitivity and specificity characteristics when used with larger population-based samples and in studies involving more complex samples that include individuals with a wide range of psychiatric and medical conditions.

Another class of candidate biomarkers is emerging from electroencephalography (32). Some studies have detected abnormalities as early as 6 months of age, thus raising the possibility that such a procedure could also facilitate early detection of risk, before symptoms emerge in the second year of life (50, 51). Magnetic resonance imaging has also been a fertile ground for ASD biomarker research, with measures of brain structure, function, maturation, connectivity, and metabolism beginning to yield fruit (16). It is still difficult to judge whether findings are contributing factors to the development of ASD or are a result of another underlying abnormality. Although there have been claims of ASD neural signatures for several years (52), only recently were neuroimaging findings tested specifically as ASD biomarkers (39).

In one study (53), increased extra-axial cerebrospinal fluid successfully predicted which infants at high familial risk for autism actually developed autism at the later point of diagnostic ascertainment. In another study of at-risk infants, hyperexpansion of the cortical surface area between 6 and 12 months of age preceded brain-volume overgrowth observed between 12 and 24 months, and the latter was linked to the emergence and severity of autism-related symptomatology (54). The authors then deployed a deep-learning algorithm using brain surface-area information at 6–12 months to predict ASD diagnosis at 24 months, achieving a positive predictive value of 81% and sensitivity of 88%. These impressive findings, of course, await replication in other samples, but they clearly suggest a future in which brain biomarkers could be a tool for the early identification of children with ASD.

The examples provided here are but a small sample of the large number of brain structure and function studies of candidate neuroimaging biomarkers in ASD (16, 17). Such studies have included evoked potentials, single-wave and complex EEG patterns, structural imaging, diffusion tensor imaging, resting-state functional MRI, and magnetic resonance spectroscopy. Although the emerging body of findings is indeed very promising, it is still the case that, in clinical practice, the etiologic yield of neuroimaging methods is very low (55, 56). These procedures are not clinically indicated in diagnosis or treatment of ASD without accompanying symptoms that necessitate them (e.g., suspicion of seizures).

Across the field, despite a massive number of studies (16), consensus on replicable and individual-patient neural measures mapping onto behavioral phenotypes (clinical or experimental) is still quite low insofar as their clinical utility potential is concerned. Research has yielded very consistent brain findings at the group level, however, primarily in regard to activation of circuitries associated with social function (57). This state of affairs may change as studies become more robust, with larger sample sizes, pooled data across research sites, and more standardized analytical approaches.

Biomarkers in ASD: Relevance to the Public Health Priority of Early Detection, Diagnosis, and Treatment

A major issue in ASD biomarker research, represented by the sampling of candidate biomarkers briefly outline above, is that the more cost-effective and clinically viable methods are yet to achieve performance levels that would make them clinically relevant. In addition, a small number of biomarkers that have yielded exciting preliminary results, with high levels of accuracy and predictive power, are probably too expensive or technically complex to be considered in clinical practice anytime soon. Yet it is exactly this combination of attributes—high accuracy and viable cost-effectiveness—that the kinds of biomarkers most likely to have an impact on clinical management of individuals with ASD need to achieve.

In medicine, the most widespread biomarkers result from analyses of relatively easily obtained biosamples (e.g., blood, urine). To date, metabolomics appears to hold the greatest translational promise in ASD diagnostic biomarkers (49, 58), but studies thus far have involved very small samples and await replication. Standard radiological and neuroimaging methods have, of course, also had a profound impact on biomarker discovery research in an array of medical conditions and, as described, have also shown great promise in ASD biomarker research. However, the most robust findings have resulted from procedures that would be too costly or difficult to execute in the nonspecialized clinical centers. Examples of relevant considerations include high cost of neuroimaging and viable financial coverage (in the absence of a specific clinical justification for the procedure); standardization of data collection parameters as well as of data-analytic approaches; and viability of the procedure in the case of young patients and patients with intellectual disability (in most cases, sedation might not be an ethical alternative in the absence of a well-justified clinical indication for the procedure).

None of these challenges, however, is insurmountable if robust, reliable, sensitive, and specific biomarkers are found. From a clinical standpoint, though, such biomarkers would still have to match well to current, expert-clinician-based standards of cost, access, viability, and utility in diagnostic and clinic evaluations. Therefore, for ASD biomarkers to become clinically translatable, it is very likely that they will also have to be cost-effective and viable in more general clinical settings.

Some progress along these lines has already been achieved in biomarkers for early identification and diagnosis of very young children (17). From a public heath standpoint, few—if any—topics in ASD research have been prioritized more strongly than the need for better tools and procedures for early identification and diagnosis. There is consensus about the criticality of early diagnosis (59, 60) and intervention (61, 62) to promote optimized outcomes, as well as about the need for much greater understanding of early ASD pathogenesis in social brain and social behavior (13, 63).

The American Academy of Pediatrics (59, 64) strongly recommends universal early screening for autism in the second year of life because early intervention significantly improves outcomes (6568) and may even normalize aspects of brain function (68, 69) for children with ASD. However, with very few exceptions, early intervention typically requires the diagnosis of ASD, which, in, turn awaits the emergence of symptoms that can only be identified as early as in the second or third year of life (70, 71). Nationally, the median age of ASD diagnosis still hovers around 4.5 years (72).

ASD is also a lifetime condition associated with economic burdens of more than $2.4 million per child or family (73) and more than $120 billion per year in the United States alone, most of which is related to supports in adult life (7476). These facts have prompted the U.S. Department of Health and Human Services’ Interagency Autism Coordinating Committee to repeatedly prioritize this domain of research, to accelerate progress (77). This consensus comes from several advances in developmental neuroscience, which, in turn, translate into research imperatives.

First, there is a need to capitalize on maximal neuroplasticity. The first two years of human life represent the period of greatest brain transformation: A newborn’s brain doubles in size in the first year of life and increases again by another 35% by year 3 (78, 79). Synaptic density, a marker of experience-dependent brain specialization, quadruples within year 1 alone, and it reaches levels 200%−300% greater than the synaptic density of an adult by the end of the third year (with concurrent and subsequent pruning and strengthening) (80, 81). Of importance, longitudinal gene expression associated with synaptogenesis over the first two years of life, over a wide range of brain structures, is characterized by maximal values across the 6- to 12-month window, then decreases drastically after 15 months, or much before the time at which ASD symptoms emerge and the condition can be reliably diagnosed (82). Therefore, treatment that is conditional on ASD diagnoses misses important windows of opportunity, and identified prodromal risks need to be addressed (13, 8385).

Second, there is a need to better understand the first two years of life of children with autism to identify early opportunities for intervention. This consensus comes from findings suggesting that developmental disruption of early-emerging mechanisms of socialization drives pathogenesis and results in autism symptoms (8487). For example, mechanisms such as preferential eye fixation move social and communication development forward, because the eyes and gaze are critical for extraction of socially adaptive information (88) and are potent enhancers of social-information neural processing throughout the lifespan (89, 90).

Developmental social neuroscience work in this area has already yielded some reliable and replicated quantitative markers of prodromal ASD. These span the spectrum of social-communication symptoms (84), represent variable instantiation of genetic vulnerability that likely represents dosage and timing of disruption (63), focus on skills that are present from the first days and weeks of life (89, 9193), and are population-wide quantitative traits under stringent genetic control (85). If these findings are fully replicated in larger samples, they will strengthen the hypothesis that both symptomatology and outcome levels result from these early disruptions and, as such, might be significantly attenuated and optimized (94, 95).

There is potentially great significance to this hypothesis if fully confirmed: As noted, most cases of ASD are tied to highly complex polygenic profiles of genetic burden (7, 96). In only a small minority of cases can single gene mutations be thought of as causal in ASD (97). Thus, the notion that the vastly heterogeneous nature of ASD is well captured by a syndromewide entity (as in its current DSM formal definition [1]) and is reliably diagnosed with standardized instruments (e.g., the Autism Diagnostic Observation Schedule and Autism Diagnostic Interview [98, 99]) might be possible not because of commonalities across the hundreds of initial causes (the so called “autisms” [100]) but because of commonalities in what these causes disrupt: infant-caregiver reciprocal social engagement. This connection between infant and caregiver can be considered not only as the platform for infant survival but also as the co-opted platform for social and communication brain-behavior development (63).

In fact, disruptions in patterns of reciprocal social engagement may occur among children with unrelated medical conditions, such as prenatal and perinatal suboptimalities (e.g., extremely preterm babies [101, 102]) and diseases (e.g., congenital heart disease [103]). These children then develop ASD-related outcomes at higher than expected levels. In summary, the phenotypic heterogeneity of ASD should not surprise us given the equally baffling genotypic heterogeneity. What might be surprising is, in fact, the homogeneity found in ASD despite such etiologic complexity (63)—sufficient homogeneity to constitute one of the most robustly validated and reliable DSM diagnoses of a neuropsychiatric disorder (104).

Several developmental neuroscience research groups using cheaper and more scalable tools than electroencephalography and MRI are capitalizing on these concepts to test experimental procedures that might serve the purpose of biomarkers for early detection and diagnosis in ASD, maybe even at the population level. In one example, Pierce and colleagues (105) used eye-tracking-based measurements to show that abnormal visual preference for geometric images (relative to social images) among toddlers had both diagnostic and predictive value, in that these measurements were associated with increased symptom severity. The performance of this test, however, had very low sensitivity (21%) and displayed exceptional specificity (98%) for ASD. The authors proposed, therefore, that this low-cost procedure might be a potential biomarker for an ASD subtype with more severe symptoms.

In another example, Jones and Klin showed that eye-tracking-based measurements of social-visual engagement (the way infants visually explore, engage, and, ultimately, learn from and adapt to their surrounding world) could segregate babies who were later diagnosed with ASD from babies who were not, from as early as 2–6 months of age. These data, moreover, were predictive of both diagnostic classification and level of disability at the time of diagnostic ascertainment, at 24 and 36 months (84). Although the densely sampled design used in that study is not community viable, similar measures used cross-sectionally at the age of 21 months or so yielded similarly accurate performance (85).

Eye-tracking technologies offer several initial advantages over neuroimaging methods. Relative to EEG- and MRI-based procedures, they are cheaper to acquire, simpler to set up, and more convenient to the patient, and procedures are briefer. This allows for repeated sessions, as one might require in a clinical trial. Procedures are also more robust relative to movement or other artifacts, which makes it possible to achieve high levels of success with all age groups and levels of function, including toddlers and intellectually disabled individuals (provided there are no major impairments in sensory or motor function).

Data acquisition can be brought to acceptable standards with multiple kinds of data-acquisition technologies, with many fewer parameters requiring standardization, and validation of data can more easily be established through standard analytic pipelines (e.g., ensuring integrity of the visual system and adequacy of equipment calibration). Data-analytic approaches, however sophisticated and computation heavy, can be executed agnostic to the eye-tracking technology used (because, ultimately, eye-tracking data refer to simple space-time coordinates relative to standardized stimuli, e.g., preprogrammed static or dynamic images or naturalistic video). Although data analysis may require sophisticated approaches, including algorithms resulting from machine-learning methods, these can be automatized in pipeline fashion and thus hidden from a clinical end user who would only need access to the clinically relevant information from a patient’s data analysis relative to normative or clinical benchmarks.

In summary, although the technology, methods, and data-analytic strategies used in eye-tracking research are advanced, this procedure, at the point of “service delivery,” may amount to a noninvasive behavioral protocol administered by a technician and lasting, depending on paradigm, maybe 10–12 minutes. Current technology allows for transmission of eye-tracking data to a central location for analyses through secure web portals, where pipelines for automated data processing and analysis can be implemented. This blueprint could result in a low-cost, high-throughput biomarker.

To date, however, none of this infrastructure has been adequately validated by any of the various research groups working on this research line. Yet progress toward this goal is being made. Early intervention offers enormous potential for optimizing outcomes for children with ASD (6062, 106, 107), although the challenges in achieving universal screening that is robust, sensitive, and specific (62) are still pervasive. Moreover, early identification holds great promise for translating screen positives into access to diagnosis and for establishing effective access to treatment and intervention (72, 108, 109). Given these possibilities, the attainment of community-viable biomarkers for early identification of ASD might be one of the most critical “game changers” in clinical management of the next generations of individuals with ASD.

Conclusion

Although ASD biomarker research has yielded substantial advancements to date, with candidate biomarkers spanning multiple domains of inquiry and multiple tools of measurement—from genetics and genomics to metabolomics, brain structure and function, and neuropsychological performance—none has as yet achieved clinical acceptance. Clinical management of individuals with ASD remains the domain of expert clinicians using symptom-based approaches. Genotypic heterogeneity and complexity continue to challenge progress in the discovery of genetic and genomic signatures of the condition; similarly, phenotypic heterogeneity continues to be a formidable challenge to mechanistic biological studies dependent on more homogeneous samples. However, near-term biomarker impact on efforts to optimize clinical outcomes is a reasonable expectation given more recent, cost-effective, and community-viable tools to aid in early screening, identification, and diagnosis of ASD. These, in turn, might strongly enable policy promoting increased access to early intervention, which is known to have a favorable lifetime impact. If so, this effort provides us with a promising opportunity to optimize outcomes for the next generations of individuals with ASD.

Dr. Klin is with the Division of Autism & Related Disabilities, Department of Pediatrics, Emory University School of Medicine, Atlanta, George, and the Marcus Autism Center, Children’s Healthcare of Atlanta.
Send correspondence to Dr. Klin (e-mail: ).

This work was supported by grants 187398AK and 94924AK from the Simons Foundation, grants R01 MH083727 and P50-MH100029 from the National Institute of Mental Health, and grant R01-HD068479 from the National Institute of Child Health and Human Development. Additional support was provided by the Marcus Foundation, the J.B. Whitehead Foundation, the Cox Foundation, and the Georgia Research Alliance.

Dr. Klin reports no financial relationships with commercial interests.

References

1 Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, VA, American Psychiatric Association.Google Scholar

2 DiCicco-Bloom E, Lord C, Zwaigenbaum L, et al.: The developmental neurobiology of autism spectrum disorder. J Neurosci 2006; 26:6897–6906CrossrefGoogle Scholar

3 Volkmar FR, Lord C, Bailey A, et al.: Autism and pervasive developmental disorders. J Child Psychol Psychiatry 2004; 45:135–170CrossrefGoogle Scholar

4 Constantino JN, Todorov A, Hilton C, et al.: Autism recurrence in half siblings: strong support for genetic mechanisms of transmission in ASD. Mol Psychiatry 2013; 18:137–138CrossrefGoogle Scholar

5 Betancur C: Etiological heterogeneity in autism spectrum disorders: more than 100 genetic and genomic disorders and still counting. Brain Res 2011; 1380:42–77CrossrefGoogle Scholar

6 Sanders SJ, He X, Willsey AJ, et al.: Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 2015; 87:1215–1233CrossrefGoogle Scholar

7 Geschwind DH, State MW: Gene hunting in autism spectrum disorder: on the path to precision medicine. Lancet Neurol 2015; 14:1109–1120CrossrefGoogle Scholar

8 Chaste P, Klei L, Sanders SJ, et al.: A genome-wide association study of autism using the Simons Simplex Collection: does reducing phenotypic heterogeneity in autism increase genetic homogeneity? Biol Psychiatry 2015; 77:775–784CrossrefGoogle Scholar

9 State MW, Šestan N: The emerging biology of autism spectrum disorders. Science 2012; 337:1301–1303CrossrefGoogle Scholar

10 Tordjman S, Cohen D, Coulon N, et al.: Reframing autism as a behavioral syndrome and not a specific mental disorder: implications of genetic and phenotypic heterogeneity. Neurosci Biobehav Rev 2017; 80:210CrossrefGoogle Scholar

11 Wetherby AM, Woods J, Allen L, et al.: Early indicators of autism spectrum disorders in the second year of life. J Autism Dev Disord 2004; 34:473–493CrossrefGoogle Scholar

12 Zwaigenbaum L, Bryson S, Lord C, et al.: Clinical assessment and management of toddlers with suspected autism spectrum disorder: insights from studies of high-risk infants. Pediatrics 2009; 123:1383–1391CrossrefGoogle Scholar

13 Klin A, Shultz S, Jones W: Social visual engagement in infants and toddlers with autism: early developmental transitions and a model of pathogenesis. Neurosci Biobehav Rev 2015; 50:189–203CrossrefGoogle Scholar

14 Klin A, Jones W, Schultz R, et al.: The enactive mind, or from actions to cognition: lessons from autism. Philos Trans R Soc Lond B Biol Sci 2003; 358:345–360CrossrefGoogle Scholar

15 Ruggeri B, Sarkans U, Schumann G, et al.: Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology (Berl) 2014; 231:1201–1216CrossrefGoogle Scholar

16 Li D, Karnath H-O, Xu X: Candidate biomarkers in children with autism spectrum disorder: a review of MRI studies. Neurosci Bull 2017; 33:219–237CrossrefGoogle Scholar

17 Varcin KJ, Nelson CA 3rd: A developmental neuroscience approach to the search for biomarkers in autism spectrum disorder. Curr Opin Neurol 2016; 29:123–129CrossrefGoogle Scholar

18 Walsh P, Elsabbagh M, Bolton P, et al.: In search of biomarkers for autism: scientific, social and ethical challenges. Nat Rev Neurosci 2011; 12:603–612CrossrefGoogle Scholar

19 Goldani AAS, Downs SR, Widjaja F, et al.: Biomarkers in autism. Front Psychiatry 2014; 5:100CrossrefGoogle Scholar

20 Anderson GM: Autism biomarkers: challenges, pitfalls and possibilities. J Autism Dev Disord 2015; 45:1103–1113CrossrefGoogle Scholar

21 Clementz BA, Sweeney JA, Hamm JP, et al.: Identification of distinct psychosis biotypes using brain-based biomarkers. Am J Psychiatry 2016; 173:373–384CrossrefGoogle Scholar

22 Pearlson GD, Clementz BA, Sweeney JA, et al.: Does biology transcend the symptom-based boundaries of psychosis? Psychiatr Clin North Am 2016; 39:165–174CrossrefGoogle Scholar

23 Insel TR: The NIMH Research Domain Criteria (RDoC) Project: precision medicine for psychiatry. Am J Psychiatry 2014; 171:395–397CrossrefGoogle Scholar

24 Framework for Defining Evidentiary Criteria for Biomarker Qualification. North Bethesda, MD, Foundation for the National Institutes of Health, 2016. https://fnih.org/sites/default/files/final/pdf/Evidentiary Criteria Framework Final Version Oct 20 2016.pdf Accessed Feb 8, 2018Google Scholar

25 Green Snyder L, D’Angelo D, Chen Q, et al.: Autism spectrum disorder, developmental and psychiatric features in 16p11.2 duplication. J Autism Dev Disord 2016; 46:2734–2748CrossrefGoogle Scholar

26 Meyer-Lindenberg A, Weinberger DR: Intermediate phenotypes and genetic mechanisms of psychiatric disorders. Nat Rev Neurosci 2006; 7:818–827CrossrefGoogle Scholar

27 Elsabbagh M, Johnson MH: Getting answers from babies about autism. Trends Cogn Sci 2010; 14:81–87CrossrefGoogle Scholar

28 Scherer SW, Dawson G: Risk factors for autism: translating genomic discoveries into diagnostics. Hum Genet 2011; 130:123–148CrossrefGoogle Scholar

29 Schwarz E, Guest PC, Rahmoune H, et al.: Sex-specific serum biomarker patterns in adults with Asperger’s syndrome. Mol Psychiatry 2011; 16:1213–1220CrossrefGoogle Scholar

30 Yap IKS, Angley M, Veselkov KA, et al.: Urinary metabolic phenotyping differentiates children with autism from their unaffected siblings and age-matched controls. J Proteome Res 2010; 9:2996–3004CrossrefGoogle Scholar

31 Ecker C, Marquand A, Mourão-Miranda J, et al.: Describing the brain in autism in five dimensions—magnetic resonance imaging-assisted diagnosis of autism spectrum disorder using a multiparameter classification approach. J Neurosci 2010; 30:10612–10623CrossrefGoogle Scholar

32 Bosl W, Tierney A, Tager-Flusberg H, et al.: EEG complexity as a biomarker for autism spectrum disorder risk. BMC Med 2011; 9:18CrossrefGoogle Scholar

33 Griffin R, Westbury C: Infant EEG activity as a biomarker for autism: a promising approach or a false promise? BMC Med 2011; 9:61CrossrefGoogle Scholar

34 Freedman EG, Foxe JJ: Eye movements, sensorimotor adaptation and cerebellar-dependent learning in autism: toward potential biomarkers and subphenotypes. Eur J Neurosci (Epub June 14, 2017). doi: 10.1111/ejn.13625CrossrefGoogle Scholar

35 Muller CL, Anacker AMJ, Veenstra-VanderWeele J: The serotonin system in autism spectrum disorder: from biomarker to animal models. Neuroscience 2016; 321:24–41CrossrefGoogle Scholar

36 Homsy J, Zaidi S, Shen Y, et al.: De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science 2015; 350:1262–1266CrossrefGoogle Scholar

37 Crawley JN, Heyer W-D, LaSalle JM: Autism and cancer share risk genes, pathways, and drug targets. Trends Genet 2016; 32:139–146CrossrefGoogle Scholar

38 Autism Biomarkers Consortium for Clinical Trials. New Haven, CT, McPartland Lab. https://medicine.yale.edu/lab/mcpartland/participate/abc-ct.aspx. Accessed Feb 12, 2018Google Scholar

39 Pelphrey K: Charting a course for autism biomarkers. Biol Psychiatry 2017; 82:155–156CrossrefGoogle Scholar

40 Constantino JN, Charman T: Diagnosis of autism spectrum disorder: reconciling the syndrome, its diverse origins, and variation in expression. Lancet Neurol 2016; 15:279–291CrossrefGoogle Scholar

41 El-Fishawy P, State MW: The genetics of autism: key issues, recent findings, and clinical implications. Psychiatr Clin North Am 2010; 33:83–105CrossrefGoogle Scholar

42 State MW, Levitt P: The conundrums of understanding genetic risks for autism spectrum disorders. Nat Neurosci 2011; 14:1499–1506CrossrefGoogle Scholar

43 Murdoch JD, State MW: Recent developments in the genetics of autism spectrum disorders. Curr Opin Genet Dev 2013; 23:310–315CrossrefGoogle Scholar

44 Shen Y, Dies KA, Holm IA, et al.: Clinical genetic testing for patients with autism spectrum disorders. Pediatrics 2010; 125:e727–e735CrossrefGoogle Scholar

45 Herman GE, Henninger N, Ratliff-Schaub K, et al.: Genetic testing in autism: how much is enough? Genet Med 2007; 9:268–274CrossrefGoogle Scholar

46 Jiang Y-H, Wang Y, Xiu X, et al.: Genetic diagnosis of autism spectrum disorders: the opportunity and challenge in the genomics era. Crit Rev Clin Lab Sci 2014; 51:249–262CrossrefGoogle Scholar

47 Anderson, G. M., Horne, W. C., Chatterjee, D., et al.: The hyperserotonemia of autism. Ann NY Acad Sci 1990; 600: 331-340–2CrossrefGoogle Scholar

48 Kaplan G, McCracken JT: Psychopharmacology of autism spectrum disorders. Pediatr Clin North Am 2012; 59:175–187, xiiCrossrefGoogle Scholar

49 West PR, Amaral DG, Bais P, et al.: Metabolomics as a tool for discovery of biomarkers of autism spectrum disorder in the blood plasma of children. PLoS One 2014; 9:e112445CrossrefGoogle Scholar

50 Righi G, Tierney AL, Tager-Flusberg H, et al.: Functional connectivity in the first year of life in infants at risk for autism spectrum disorder: an EEG study. PLoS One 2014; 9:e105176CrossrefGoogle Scholar

51 Wang J, Barstein J, Ethridge LE, et al.: Resting state EEG abnormalities in autism spectrum disorders. J Neurodev Disord 2013; 5:24CrossrefGoogle Scholar

52 Kaiser MD, Hudac CM, Shultz S, et al.: Neural signatures of autism. Proc Natl Acad Sci USA 2010; 107:21223–21228CrossrefGoogle Scholar

53 Shen MD, Kim SH, McKinstry RC, et al.: Increased extra-axial cerebrospinal fluid in high-risk infants who later develop autism. Biol Psychiatry 2017; 82:186–193CrossrefGoogle Scholar

54 Hazlett HC, Gu H, Munsell BC, et al.: Early brain development in infants at high risk for autism spectrum disorder. Nature 2017; 542:348–351CrossrefGoogle Scholar

55 Tomas Vila M: Diagnostic yield in studies of autism: the contribution made by neuroimaging, metabolic tests and genetic studies. Rev Neurol 2004; 38(Suppl 1):S15–S20Google Scholar

56 Shevell MI, Majnemer A, Rosenbaum P, et al.: Etiologic yield of autistic spectrum disorders: a prospective study. J Child Neurol 2001; 16:509–512CrossrefGoogle Scholar

57 Philip RCM, Dauvermann MR, Whalley HC, et al.: A systematic review and meta-analysis of the fMRI investigation of autism spectrum disorders. Neurosci Biobehav Rev 2012; 36:901–942CrossrefGoogle Scholar

58 Noto A, Fanos V, Barberini L, et al.: The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. Mol Autism 2016; 7:47–52Google Scholar

59 Johnson CP, Myers SM: Identification and evaluation of children with autism spectrum disorders. Pediatrics 2007; 120:1183–1215CrossrefGoogle Scholar

60 Dawson G: Why it’s important to continue universal autism screening while research fully examines its impact. JAMA Pediatr 2016; 170:527–528CrossrefGoogle Scholar

61 Dawson G, Bernier R: A quarter century of progress on the early detection and treatment of autism spectrum disorder. Dev Psychopathol 2013; 25:1455–1472CrossrefGoogle Scholar

62 Daniels AM, Halladay AK, Shih A, et al.: Approaches to enhancing the early detection of autism spectrum disorders: a systematic review of the literature. J Am Acad Child Adolesc Psychiatry 2014; 53:141–152CrossrefGoogle Scholar

63 Jones W, Klin A: Heterogeneity and homogeneity across the autism spectrum: the role of development. J Am Acad Child Adolesc Psychiatry 2009; 48:471–473CrossrefGoogle Scholar

64 Adams RC, Tapia C: Early intervention, IDEA Part C services, and the medical home: collaboration for best practice and best outcomes. Pediatrics 2013; 132:e1073–e1088CrossrefGoogle Scholar

65 Dawson G, Rogers S, Munson J, et al.: Randomized, controlled trial of an intervention for toddlers with autism: the Early Start Denver Model. Pediatrics 2010; 125:e17–e23CrossrefGoogle Scholar

66 Wetherby AM, Guthrie W, Woods J, et al.: Parent-implemented social intervention for toddlers with autism: an RCT. Pediatrics 2014; 134:1084–1093CrossrefGoogle Scholar

67 Warren Z, McPheeters ML, Sathe N, et al.: A systematic review of early intensive intervention for autism spectrum disorders. Pediatrics 2011; 127:e1303–e1311CrossrefGoogle Scholar

68 Reichow B, Barton EE, Boyd BA, et al.: Early intensive behavioral intervention (EIBI) for young children with autism spectrum disorders (ASD). Cochrane Database Syst Rev; 10:CD009260, 2012. doi: 10.1002/14651858.CD009260.pub2CrossrefGoogle Scholar

69 Venkataraman A, Yang DY-J, Dvornek N, et al.: Pivotal response treatment prompts a functional rewiring of the brain among individuals with autism spectrum disorder. Neuroreport 2016; 27:1081–1085CrossrefGoogle Scholar

70 Chawarska K, Klin A, Paul R, et al.: Autism spectrum disorder in the second year: stability and change in syndrome expression. J Child Psychol Psychiatry 2007; 48:128–138CrossrefGoogle Scholar

71 Chawarska K, Klin A, Volkmar FR: Autism spectrum disorders in infants and toddlers. New York: Guilford Press, 2008Google Scholar

72 Shattuck PT, Durkin M, Maenner M, et al.: Timing of identification among children with an autism spectrum disorder: findings from a population-based surveillance study. J Am Acad Child Adolesc Psychiatry 2009; 48:474–483CrossrefGoogle Scholar

73 Buescher AV, Cidav Z, Knapp M, et al.: Costs of autism spectrum disorders in the United Kingdom and the United States of America. JAMA Pediatr 2014; 721–728. doi: 10.1001/jamapediatrics.2014.210CrossrefGoogle Scholar

74 Cidav Z, Marcus SC, Mandell DS: Implications of childhood autism for parental employment and earnings. Pediatrics 2012; 129:617–623CrossrefGoogle Scholar

75 Peacock G, Amendah D, Ouyang L, et al.: Autism spectrum disorders and health care expenditures: the effects of co-occurring conditions. J Dev Behav Pediatr 2012; 33:2–8CrossrefGoogle Scholar

76 Wang L, Mandell DS, Lawer L, et al.: Healthcare service use and costs for autism spectrum disorder: a comparison between Medicaid and private insurance. J Autism Dev Disord 2013; 43:1057–1064CrossrefGoogle Scholar

77 Interagency Autism Coordinating Committee. 2016–2017 Interagency Autism Coordinating Committee, strategic plan for autism spectrum disorder research.Rockville, MD, Department of Health and Human Services, 2017. https://iacc.hhs.gov/publications/strategic-plan/2017/strategic_plan_2017.pd. accessed Feb 12, 2018Google Scholar

78 Gilmore JH, Lin W, Prastawa MW, et al.: Regional gray matter growth, sexual dimorphism, and cerebral asymmetry in the neonatal brain. J Neurosci 2007; 27:1255–1260CrossrefGoogle Scholar

79 Pfefferbaum A, Mathalon DH, Sullivan EV, et al.: A quantitative magnetic resonance imaging study of changes in brain morphology from infancy to late adulthood. Arch Neurol 1994; 51:874–887CrossrefGoogle Scholar

80 Huttenlocher PR: Synaptic density in human frontal cortex—developmental changes and effects of aging. Brain Res 1979; 163:195–205CrossrefGoogle Scholar

81 Petanjek Z, Judaš M, Šimic G, et al.: Extraordinary neoteny of synaptic spines in the human prefrontal cortex. Proc Natl Acad Sci USA 2011; 108:13281–13286CrossrefGoogle Scholar

82 Kang HJ, Kawasawa YI, Cheng F, et al.: Spatio-temporal transcriptome of the human brain. Nature 2011; 478:483–489CrossrefGoogle Scholar

83 Sacrey L-AR, Bennett JA, Zwaigenbaum L: Early infant development and intervention for autism spectrum disorder. J Child Neurol 2015; 30:1921–1929CrossrefGoogle Scholar

84 Jones W, Klin A: Attention to eyes is present but in decline in 2–6-month-old infants later diagnosed with autism. Nature 2013; 504:427–431CrossrefGoogle Scholar

85 Constantino JN, Kennon-McGill S, Weichselbaum C, et al.: Infant viewing of social scenes is under genetic control and is atypical in autism. Nature 2017; 547:340–344CrossrefGoogle Scholar

86 Klin, A., Jones, W., Schultz, R., et al.: The enactive mind, or from actions to cognition: Lessons from autism. Philos Trans R Soc B Biol Sci 2003; 358:345–360CrossrefGoogle Scholar

87 Klin A, Lin DJ, Gorrindo P, et al.: Two-year-olds with autism orient to non-social contingencies rather than biological motion. Nature 2009; 459:257–261CrossrefGoogle Scholar

88 Kampe KK, Frith CD, Dolan RJ, et al.: Reward value of attractiveness and gaze. Nature 2001; 413:589CrossrefGoogle Scholar

89 Farroni T, Johnson MH, Csibra G: Mechanisms of eye gaze perception during infancy. J Cogn Neurosci 2004; 16:1320–1326CrossrefGoogle Scholar

90 Adolphs R, Gosselin F, Buchanan TW, et al.: A mechanism for impaired fear recognition after amygdala damage. Nature 2005; 433:68–72CrossrefGoogle Scholar

91 Farroni T, Csibra G, Simion F, et al.: Eye contact detection in humans from birth. Proc Natl Acad Sci USA 2002; 99:9602–9605CrossrefGoogle Scholar

92 Farroni T, Massaccesi S, Menon E, et al.: Direct gaze modulates face recognition in young infants. Cognition 2007; 102:396–404CrossrefGoogle Scholar

93 Haith MM, Bergman T, Moore MJ: Eye contact and face scanning in early infancy. Science 1977; 198:853–855CrossrefGoogle Scholar

94 Klin A, Klaiman C, Jones W: Reducing age of autism diagnosis: developmental social neuroscience meets public health challenge. Rev Neurol 2015; 60(Suppl 1):S3–S11Google Scholar

95 Klin A, Wetherby AM, Woods J, et al.: Toward innovative, cost-effective, and systemic solutions to improve outcomes and well-being of military families affected by autism spectrum disorder. Yale J Biol Med 2015; 88:73–79Google Scholar

96 Gandal MJ, Leppa V, Won H, et al.: The road to precision psychiatry: translating genetics into disease mechanisms. Nat Neurosci 2016; 19:1397–1407CrossrefGoogle Scholar

97 Geschwind DH: Genetics of autism spectrum disorders. Trends Cogn Sci 2011; 15:409–416CrossrefGoogle Scholar

98 Lord C, Rutter M, DiLavore P, et al.: ADOS Toddler Module: ADOS-T: Manual. Torrance, CA, Western Psychological Services, 2008Google Scholar

99 Rutter M, Le Couteur A, Lord C: ADI-R: Autism Diagnostic Interview—Revised. Torrance, CA, Western Psychological Services, 2003Google Scholar

100 Insel TR: The challenge of translation in social neuroscience: a review of oxytocin, vasopressin, and affiliative behavior. Neuron 2010; 65:768–779CrossrefGoogle Scholar

101 Johnson S, Hollis C, Kochhar P, et al.: Autism spectrum disorders in extremely preterm children. J Pediatr 2010; 156:525–531CrossrefGoogle Scholar

102 Limperopoulos C, Bassan H, Sullivan NR, et al.: Positive screening for autism in ex-preterm infants: prevalence and risk factors. Pediatrics 2008; 121:758–765CrossrefGoogle Scholar

103 Marino BS, Lipkin PH, Newburger JW, et al.: Neurodevelopmental outcomes in children with congenital heart disease: evaluation and management: a scientific statement from the American Heart Association. Circulation 2012; 126:1143–1172CrossrefGoogle Scholar

104 Lord C, Petkova E, Hus V, et al.: A multisite study of the clinical diagnosis of different autism spectrum disorders. Arch Gen Psychiatry 2012; 69:306–313CrossrefGoogle Scholar

105 Pierce K, Marinero S, Hazin R, et al.: Eye tracking reveals abnormal visual preference for geometric images as an early biomarker of an autism spectrum disorder subtype associated with increased symptom severity. Biol Psychiatry 2016; 79:657–666CrossrefGoogle Scholar

106 Schreibman L, Dawson G, Stahmer AC, et al.: Naturalistic developmental behavioral interventions: empirically validated treatments for autism spectrum disorder. J Autism Dev Disord 2015; 45:2411–2428CrossrefGoogle Scholar

107 Zwaigenbaum L, Bauman ML, Choueiri R, et al.: Early identification and interventions for autism spectrum disorder: executive summary. Pediatrics 2015; 136(Suppl 1):S1–S9CrossrefGoogle Scholar

108 Robins DL, Casagrande K, Barton M, et al.: Validation of the Modified Checklist for Autism in Toddlers, Revised With Follow-Up (M-CHAT-R/F). Pediatrics 2014; 133:37–45CrossrefGoogle Scholar

109 Daniels AM, Mandell DS: Explaining differences in age at autism spectrum disorder diagnosis: a critical review. Autism 2014; 18:583–597CrossrefGoogle Scholar