Next Article in Journal
Correlation between Language Development and Motor Skills, Physical Activity, and Leisure Time Behaviour in Preschool-Aged Children
Next Article in Special Issue
Oral Health Preventive Program in Patients with Autism Spectrum Disorder
Previous Article in Journal
Identification of a Novel FAM83H Mutation and Management of Hypocalcified Amelogenesis Imperfecta in Early Childhood
Previous Article in Special Issue
Quantitative Assessment of Sensory Integration and Balance in Children with Autism Spectrum Disorders: Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Research-Based Intervention (RBI) for Autism Spectrum Disorder: Looking beyond Traditional Models and Outcome Measures for Clinical Trials

by
Antonio Narzisi
1,*,
Yurena Alonso-Esteban
2,
Gabriele Masi
1 and
Francisco Alcantud-Marín
2
1
Department of Child Psychiatry and Psychopharmacology, IRCCS Stella Maris Foundation, 56812 Pisa, Italy
2
Department of Developmental and Educational Psychology, University of Valencia, Avenida de Blasco Ibáñez, 13, 46010 Valencia, Spain
*
Author to whom correspondence should be addressed.
Children 2022, 9(3), 430; https://doi.org/10.3390/children9030430
Submission received: 23 February 2022 / Revised: 14 March 2022 / Accepted: 16 March 2022 / Published: 18 March 2022
(This article belongs to the Special Issue Autism and Children)

Abstract

:
The rising prevalence of Autism Spectrum Disorders (ASD) has led to a quickly increasing need for effective interventions. Several criteria and measures have been developed to critically assess these interventions with particular focus on the evaluation of the efficacy. Given the huge diversity of ASD symptoms and the different levels of severity across individuals, identifying a one size fits all intervention approach is challenging, and the question What works and for whom? Remains still unanswered. Why do we seem to be dragging our feet on this fundamental issue? The main aim of this paper is to answer this question through four non-alternative points. First, there are a scarce number of studies with a solid methodology. Secondly, most trials on intervention efficacy for ASD are designed exclusively in terms of behavioral outcomes. Thirdly, there is a reduced use of biologically oriented outcome measures. Fourthly, in most clinical trials, appropriate practices emerging from research evidence are not systematically applied. A strong effort to improve the methodology of clinical trials is mandatory for the future of autism research. The development of a research-based intervention (RBI) perspective aimed at better integrating: (a) evidence-based approaches; (b) more sensitive behavioral outcome measures; and (c) biomarkers, with the aim of increasing a more detailed clustering of phenotypes, may strongly improve our approach to a precision medicine.

The rising prevalence of Autism Spectrum Disorders (ASD) [1] has led to a quickly increasing need for effective interventions. Timely interventions aim to prevent or minimize the developmental effects of early impairments [2]. Currently, a large number of methods and techniques are available, with different levels of scientific evidence [3,4]. Several criteria and measures have been developed to critically assess these interventions [5,6,7,8,9,10], with particular focus on the evaluation of the efficacy [11,12,13,14,15]. However, despite the rapidly growing number of studies, most of them fall below the threshold of adequate methodological quality in conformity with the Cochrane risk of bias tool [16]. It is organized into a definite set of domains of bias, focusing on different facets of trial design, conduct, as well as reporting the risk of bias in RCT contained in Cochrane Reviews.
Given the huge diversity of ASD symptoms and the different levels of severity across individuals, identifying a “one size fits all” intervention approach is challenging, and the question “What works and for whom?” [15] remains still unanswered. In other words, in this field we are still far from the target of “precision medicine”. Why do we seem to be dragging our feet on this fundamental issue? The main aim of this paper is to answer this question through four non-alternative points:
First, as previously reported, there are a scarce number of studies with a solid methodology, and this issue strongly limits the generalization of the findings [16]. These limits, in line with the Cochrane risk of bias tool, can be summarized as follows: (a) small sample size; (b) small number of multi-center studies; (c) the absence of long-term research on outcome effects; (d) the reduced adaptation of some evidence-based interventions outside of the country and research group in which they were developed; and (e) the excessive number of novel intervention models. It is emblematic that, currently, only 12% of published papers meet the criteria for high methodological quality [16]. This means that research in the arena of intervention in ASD, apart from a few rare examples [9,17], remains below cutting-edge levels [18,19].
Secondly, (a) most trials on intervention efficacy for ASD are designed exclusively in terms of behavioral outcomes. Despite the wide range of psychometric measures providing useful information (e.g., about IQ, language), they cannot assess subtle changes in the quality of the core symptomatology of the disorder (e.g., parent–child interaction, social-communication behaviors); (b) the differences in the behavioral outcome measures used by studies do not allow for comparison between trials.
Over the years, the low reliability of outcome measures and the lack of objective markers to identify subgroups of young children with different levels of response to specific interventions has prompted researchers to develop more sensitive methodological designs. The systematic adoption of instruments aimed at studying subtle changes in children and in parent–child interactions may be helpful at further improving current outcome measures. Among the well-conducted research in ASD [20,21] the ADOS-BOSCC (Brief Observation of Social Communication Change) [19] was drawn up specifically as an outcome measure in early intervention trials, to detect subtle clinical changes especially in the Social Communication domain (e.g., eye contact, facial expressions, gestures, vocalizations, social overtures, social responses, requesting and engagement) [18,22]. The ADOS-BOSCC is based on videotaped observation of parent–child naturalistic social interactions and free play [19]. Findings about ADOS-BOSCC: (a) support it as a promising test for recordings modifications in social communication behaviors as a result of a behavioral treatment [23]; and (b) indicate strong reliability and validity also in children with ASD who have reduced use of language. Findings in addition claim that the ADOS-BOSCC may be more sensitive in detecting subtle changes in social communication, compared to other instruments [22]. Unfortunately, due to time-consuming, training and costs, few trials have used ADOS-BOSCC as a supplementary outcome measure [21,24].
The findings of a seminal research carried out by Green et al. in 2010, replicated in 2016 [25], showed that the DCMA (Dyadic Communication Measure for Autism) could be used to study, from a punctiform perspective, the parental child interaction. [12,26]. It provides a reliable and naturalistic assessment of the interactive exchanges between parent and child in a play setting [12,26,27]. The interaction is then coded from video-tapes, and three levels are observed: (a) parent synchrony and responsiveness; (b) child communicative initiations, responses, and communicative functions; (c) amount of mutual shared attention between parent and child [12,26,27]. However, these measures of parent–child interactions are not yet systematically included in most clinical trials, with few exceptions [28,29].
Thirdly, a reliable response to the question about what works and for whom is difficult, given the reduced use of biologically oriented outcome measures. In recent years, biomarker research has become an increasing goal in the field of ASD. Although much effort is being made, there are still no reliable and valid biological or brain imaging markers that would allow us to study subtle changes more objectively during the intervention [30,31,32].
To address the question, it would be essential to implement studies including, alongside behavioral outcome measures, also biologically oriented outcome measures, such as EEG, eye-tracking, f-MRI, and wearable devices for neurovegetative parameters. Integrating the results of biological and behavioral outcome measures using artificial intelligence may help to identifying subtypes of ASD with different responses to specific interventions, with the aim to develop and monitor specific biological therapies [31].
In terms of biological-oriented outcome measures, the research has shown that visual patterning, explored through eye-tracking, could be a promising measure to monitor subtle response to the intervention, to predict outcomes, and to determine unique features of the child’s performance that fit with the proposed mechanisms of change [33]. Over the past two decades, eye-tracking device has been widely applied in ASD research [34], namely, to study joint attention [35], social attention [36,37], visual preference [38], responses to dyadic bids [39], theory of mind abilities [40], facial expression recognition [41], and attentional preferences at both the semantic [42] and perceptual levels [43].
Since the literature has identified unordinary models of functional brain connectivity in young children with ASD based on electrophysiological measures [44,45,46], the EEG and fMRI should also be more closely considered in trials using biologically oriented outcome measures. Studies from animal models have reported multiple atypicality in functional brain connectivity, known as connectopathy [46,47]. Zerbi et al. carried out a cross-etiological investigation of fMRI-based connectivity in the mice, showing that different ASD-associated etiologies cause a broad spectrum of connectional abnormalities, with different, often diverging, connectivity signatures [47]. These findings suggest that etiological variability is a key determinant of connectivity heterogeneity in ASD, accounting for conflicting findings in different clinical populations.
Following the wave of atypical connectivity, postmortem studies have revealed increased density of excitatory synapses in the brain of individuals with ASD, with a putative link to aberrant mTORdependent synaptic pruning [46,47]. These observations raise the question of whether an excess of synapses may cause aberrant functional connectivity in ASD. Using resting state fMRI, electrophysiology and in silico modelling in Tsc2 haploinsufficient mice, Pagani et al. showed that mTOR-dependent increased spine density is associated with ASD-like stereotypies and cortico-striatal hyperconnectivity [47]. These deficits are completely rescued by pharmacological inhibition of mTOR. Pagani et al. showed that the identified transcriptomic signature is predominantly expressed in a subset of children with ASD, thereby defining a segregable autism subtype [47]. The findings of Pagani et al. causally link mTOR-related synaptic pathology to large-scale network aberrations, revealing a unifying multi-scale framework that mechanistically integrates developmental synaptopathy and functional hyperconnectivity in ASD. Thus, the identification of etiologically relevant connectivity subtypes could improve diagnostic label accuracy in the non-syndromic ASD population and paves the way for personalized treatment approaches.
In 2012, a trial by Dawson et al. [11] showed that the behavioral changes in preschoolers with ASD after a Naturalistic Developmental Behavioral Intervention (NDBI) [17] were associated with the normalization of brain activity patterns, parallel to the improvements in social behavior.
In 2016, Yang et al. demonstrated that functional MRI may predict responses to evidence-based behavioral intervention [48]. In their study, Yang et al. [48] identified neural predictors of pre-intervention activity levels in response to biological versus scrambled movement in neural circuits that support social information processing and social motivation/reward. The predictive value of their findings in ASD children was supported by cross-validated multivariate pattern analysis. The implications of these findings are far-reaching and should greatly accelerate progress towards more accurate and effective interventions for the core deficits of ASD [48].
Fourthly, in most clinical trials, appropriate practices emerging from research evidence are not systematically applied. Following the advices of Zwaigenbaum et al. [49], effective intervention research and practice should: (a) include naturalistic behavioral and developmental interventions; (b) involve parents in the treatment setting; (c) act on secondary and core ASD deficits; (d) take into account the socio-cultural facets of families involved in treatment and their affordability as possible variables moderating outcomes; (e) include subjects come from many different countries to evaluate how factors of cultural affiliation might affect therapeutic compliance; (f) apply a rigorous research methodology and be faithful to the implementation of the model; (g) examine active ingredients of effective treatments such as treatment hours; and (h) use a standardized outcome assessment protocol.
In addition to the above suggestions, another best practice aimed to capture subtle changes in children and their interacting families is the technique of video-feedback [12,50]. It typically consists of videotaped play sessions between parent and child. These recordings are then observed by the parent along with a clinician. The role of the clinician is to help the parent reflect on the child’s behaviors as well as the parent’s own behaviors. The goal of video-feedback is to support the parent in being able to pick up on the child’s subtle communicative cues and develop effective interaction strategies [46]. Literature showed that the video-feedback can be integrated into parent mediated interventions to increase the child’s language skills [51], make parents less stressed and more competent in their role [52], increase parental synchronous acts [12], reduce, in the long term, autistic symptomatology [24], and decrease parental asynchronous acts [52].
Although substantial advancement continues to be made within the past decade in the study of ASD, the translational research about the efficacy and effectiveness of interventions is hampered by extreme heterogeneity in models, as well as in outcome measures. These limits do not allow for an adequate generalization of findings and a significant progress in precision medicine. The lack of a systematic integration of behavioral and biomarker outcomes in clinical trials targeting the core symptomatology of ASD decrease our potential for new knowledge (i.e., from pharmacological studies) [53]. A strong effort to improve the methodology of clinical trials is mandatory for the future of autism research. As reported by Ruggeri et al. [54], recognized biomarkers, neuropsychological assessments, electrophysiological measurements, and functional brain imaging will be linked with novel biomarkers identified from omics data (e.g., proteomic, epigenomic) to generate multimarker panels. Management of these data and analysis methods using artificial intelligence techniques will be central to identifying these biomarker panels.

Conclusions

The development of a research-based intervention (RBI) perspective aimed at better integrating: (a) evidence-based approaches; (b) more sensitive behavioral outcome measures; and (c) biomarkers, with the aim of increasing a more detailed clustering of phenotypes, may strongly improve our approach to a precision medicine. This target could help us to better identify the most effective intervention solution for children and improve and accelerate the development of effective treatments (including innovative drugs) for the core deficits of ASD.

Author Contributions

Conceptualization, A.N. Writing—review and editing, A.N., Y.A.-E., G.M. and F.A.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been partially supported by grant-RC 2.06 and the 5 × 1000 voluntary contributions, Italian Ministry of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

There are no data in this paper.

Conflicts of Interest

G.M. has received institutional research grants from Lundbeck and Humana, was on an advisory board for Angelini, and has been a speaker for Angelini, FB Health, Janssen, Lundbeck, and Otsuka. All the other authors do not have conflict of interest to declare.

References

  1. Maenner, M.J.; Shaw, K.A.; Bakian, A.V.; Bilder, D.A.; Durkin, M.S.; Esler, A.; Furnier, S.M.; Hallas, L.; Hall-Lande, J.; Hudson, A.; et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2018. MMWR Surveill. Summ. 2021, 70, 1–16. [Google Scholar] [CrossRef]
  2. Francis, K.; Karantanos, G.; Al-Ozairi, A.; AlKhadhari, S. Prevention in Autism Spectrum Disorder: A Lifelong Focused Approach. Brain Sci. 2021, 11, 151. [Google Scholar] [CrossRef] [PubMed]
  3. Green, V.; Pituch, K.; Itchon, J.; Choi, A.; O’Reilly, M.; Sigafoos, J. Internet survey of treatments used by parents of children with autism. Res. Dev. Disabil. 2006, 27, 70–84. [Google Scholar] [CrossRef] [PubMed]
  4. Narzisi, A.; Colombi, C.; Balottin, U.; Muratori, F. Non-pharmacological treatments in autism spectrum disorders: An overview on early interventions for pre-schoolers. Curr. Clin. Pharmacol. 2014, 9, 17–26. [Google Scholar] [CrossRef] [PubMed]
  5. Cook, B.; Buysse, V.; Klingner, J. CEC’s standards for classifying the evidence base of practices in special education. Remedial Spec. Educ. 2014, 36, 220–234. [Google Scholar] [CrossRef]
  6. Harris, R.; Helfand, M.; Woolf, S.; Lohr, K.; Mulrow, C.; Teutsch, S.; Atkins, D. Current methods of the US Preventive Services Task Force: A review of the process. Am. J. Prev. Med. 2001, 20, 21–35. [Google Scholar] [CrossRef]
  7. Kemdall, T.; Megnin-Viggars, O.; Gould, N.; Taylor, C.; Burt, L.; Baird, G. Management of autism in children and young people: Summary of NICE and SCIE guidance. Br. Med. J. 2013, 347, f4865. [Google Scholar] [CrossRef] [Green Version]
  8. Manglione, M.; Gans, D.; Das, L.; Timbie, J.; Kasari, C.; Technical Expert Panel, & HRSA AIRB Network. Nonmedical Interventions for Children With ASD: Recommended Guidelines and Further Research Needs. Pediatrics 2012, 130, S169. [Google Scholar] [CrossRef] [Green Version]
  9. Reichow, B.; Hume, K.; Barton, E.; Boyd, B. Early intensive behavioral intervention (EIBI) for young children with autism spectrum disorders (ASD). Cochrane Database Syst. Rev. 2018, 5, 1–16. [Google Scholar] [CrossRef]
  10. Simith, T.; Iadarola, S. Evidence base update for autism spectrum disorder. J. Clin. Child Adolesc. Psychol. 2015, 44, 897–922. [Google Scholar] [CrossRef] [Green Version]
  11. Dawson, G.; Jones, E.; Merkle, K.; Venema, K.; Lowy, R.; Faja, S.; Webb, S. Early Behavioral Intervention Is Associated with Normalized Brain Activity in Young Children With Autism. J. Am. Acad. Child Adolesc. Psychiatry 2012, 51, 1150–1159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Green, J.; Charman, T.; McConachie, H.; Aldred, C.; Slonims, V.; Howlin, P.; Le Couteur, A.; Leadbitter, K.; Hudry, K.; Byford, S.; et al. Parent-mediated communication-focused treatment in children with autism (PACT): A randomised controlled trial. Lancet 2010, 375, 2152–2160. [Google Scholar] [CrossRef] [Green Version]
  13. Landa, R. Efficacy of early interventions for infants and young children with, and at risk for, autism spectrum disorders. Int. Rev. Psychiatry 2018, 30, 25–39. [Google Scholar] [CrossRef] [PubMed]
  14. Nahmias, A.; Pellecchia, M.; Stahmer, A.; Mandell, D. Effectiveness of community-based early intervention for children with autism spectrum disorder: A meta-analysis. J. Child Psychol. Psychiatry 2019, 60, 1200–1209. [Google Scholar] [CrossRef] [PubMed]
  15. Vivanti, G.; Stahmer, A. Early intervention for autism: Are we prioritizing feasibility at the expense of effectiveness? A cautionary note. Autism 2018, 22, 770–773. [Google Scholar] [CrossRef] [Green Version]
  16. French, L.; Kennedy, E. Annual Research Review: Early intervention for infants and young children with, or at-risk of, autism spectrum disorder: A systematic review. J. Child Psychol. Psychiatry 2018, 59, 444–456. [Google Scholar] [CrossRef] [Green Version]
  17. Schreibman, L.; Dawson, G.; Stahmer, A.C.; Landa, R.; Rogers, S.J.; McGee, G.G.; Kasari, C.; Ingersoll, B.; Kaiser, A.P.; Bruinsma, Y.; et al. Naturalistic Developmental Behavioral Interventions: Empirically Validated Treatments for Autism Spectrum Disorder. J. Autism Dev. Disord. 2015, 45, 2411–2428. [Google Scholar] [CrossRef] [Green Version]
  18. Curruthers, S.; Charman, T.; El Hawi, N.; Ah Kim, Y.; Randle, R.; Lord, C.; PACT Consortium. Utility of the Autism Diagnostic Observation Schedule and the Brief Observation of Social and Communication Change for Measuring Outcomes for a Parent-Mediated Early Autism Intervention. Autism Res. 2021, 14, 411–425. [Google Scholar] [CrossRef] [PubMed]
  19. Grzadzinski, R.; Carr, T.; Colombi, C.; McGuire, K.; Dufek, S.; Pickles, A.; Lord, C. Measuring Changes in Social Communication Behaviors: Preliminary Development of the Brief Observation of Social Communication Change (BOSCC). J. Autism Dev. Disord. 2016, 46, 264–2479. [Google Scholar] [CrossRef] [Green Version]
  20. Green, J.; Aldred, C.; Charman, T.; Le Couteur, A.; Emsley, R.A.; Grahame, V.; Howlin, P.; Humphrey, N.; Leadbitter, K.; McConachie, H.; et al. Paediatric Autism Communication Therapy-Generalised (PACT-G) against treatment as usual for reducing symptom severity in young children with autism spectrum disorder: Study protocol for a randomised controlled trial. Trials 2018, 19, 514. [Google Scholar] [CrossRef]
  21. McClure, L.A.; Lee, N.L.; Sand, K.; Vivanti, G.; Fein, D.; Stahmer, A.; Robins, D.L. Connecting the Dots: A cluster-randomized clinical trial integrating standardized autism spectrum disorders screening, high-quality treatment, and long-term outcomes. Trials 2021, 22, 319. [Google Scholar] [PubMed]
  22. Kim, S.; Grzadzinski, R.; Martinez, K.; Lord, C. Measuring treatment response in children with autism spectrum disorder: Applications of the Brief Observation of Social Communication Change to the Autism Diagnostic Observation Schedule. Autism 2019, 23, 1176–1185. [Google Scholar] [CrossRef] [PubMed]
  23. Gengoux, G.W.; Abrams, D.A.; Schuck, R.; Millan, M.E.; Libove, R.; Ardel, C.M.; Phillips, J.M.; Fox, M.; Frazier, T.W.; Hardan, A.Y. A Pivotal Response Treatment Package for Children with Autism Spectrum Disorder: An RCT. Pediatrics 2019, 144, e20190178. [Google Scholar] [CrossRef] [PubMed]
  24. Green, J.; Garg, S. Annual Research Review: The state of autism intervention science: Progress, target psychological and biological mechanisms and future prospects. J. Child Psychol. Psychiatry 2018, 59, 424–443. [Google Scholar] [CrossRef] [Green Version]
  25. Pickles, A.; Le Couteur, A.; Leadbitter, K.; Salomone, E.; Cole-Fletcher, R.; Tobin, H.; Gammer, I.; Lowry, J.; Vamvakas, G.; Byford, S.; et al. Parent-mediated social communication therapy for young children with autism (PACT): Long-term follow-up of a randomised controlled trial. Lancet 2016, 388, 2501–2509. [Google Scholar]
  26. Aldred, C.; Green, J.; Adams, C. A new social communication intervention for children with autism: Pilot randomised controlled treatment study suggesting effectiveness. J. Child Psychol. Psychiatry 2004, 45, 1420–1430. [Google Scholar] [CrossRef]
  27. Aldred, C.; Green, J.; Emsley, R.; McConachie, M. Brief report: Mediation of treatment effect in a communication intervention for pre-school children with autism. J. Autism Dev. Disord. 2011, 42, 447–454. [Google Scholar] [CrossRef]
  28. Oono, I.P.; Honey, E.J.; McConachie, H. Parent-mediated early intervention for young children with autism spectrum disorders (ASD). Cochrane Database Syst. Rev. 2013, 30, CD009774. [Google Scholar]
  29. Rahman, A.; Divan, G.; Hamdani, S.U.; Vajaratkar, V.; Taylor, C.; Leadbitter, K.; Aldred, C.; Minhas, A.; Cardozo, P.; Emsley, R.; et al. Effectiveness of the parent-mediated intervention for children with autism spectrum disorder in south Asia in India and Pakistan (PASS): A randomised controlled trial. Lancet Psychiatry 2016, 3, 128–136. [Google Scholar] [CrossRef]
  30. Wang, Y.; Wang, P.; Xu, X.; Godstein, J.; McConkie, A.; Cheung, S.; Jiang, Y. Genetics of Autism Spectrum Disorders: The Opportunity and Challenge in the Genetics Clinic. In The Molecular Basis of Autism; Fantemi, H., Ed.; Springer: New York, NY, USA, 2015; pp. 33–66. [Google Scholar]
  31. Muglia, P.; Filosi, M.; Da Ros, L.; Kam-Thong, T.; Nardocci, F.; Trabetti, E.; Ratti, E.; Rizzini, P.; Zuddas, A.; Bernardina, B.D.; et al. The Italian autism network (ITAN): A resource for molecular genetics and biomarker investigations. BMC Psychiatry 2018, 18, 369. [Google Scholar] [CrossRef] [Green Version]
  32. Lord, C.; Charman, T.; Havdahl, A.; Carbone, P.; Anagnostou, E.; Boyd, B.; Carr, T.; de Vries, P.J.; Dissanayake, C.; Divan, G.; et al. The Lancet Commission on the future of care and clinical research in autism. Lancet 2022, 399, 271–334. [Google Scholar] [CrossRef]
  33. Bradshaw, J.; Shic, F.; Holden, A.; Horowitz, E.; Barrett, A.; German, T.; Vernon, T. The Use of Eye Tracking as a Biomarker of Treatment Outcome in a Pilot Randomized Clinical Trial for Young Children with Autism. Autism Res. 2019, 12, 779–793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Mastergeorge, A.M.; Kahathuduwa, C.; Blume, J. Eye-Tracking in Infants and Young Children at Risk for Autism Spectrum Disorder: A Systematic Review of Visual Stimuli in Experimental Paradigms. J. Autism Dev. Disord. 2021, 51, 2578–2599. [Google Scholar] [CrossRef] [PubMed]
  35. Nyström, P.; Thorup, E.; Bölte, S.; Falck-Ytter, T. Joint Attention in Infancy and the Emergence of Autism. Biol. Psychiatry 2019, 86, 631–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Billeci, L.; Narzisi, A.; Campatelli, G.; Crifaci, G.; Calderoni, S.; Gagliano, A.; Calzone, C.; Colombi, C.; Pioggia, G.; Muratori, F.; et al. Disentangling the initiation from the response in joint attention: An eye-tracking study in toddlers with autism spectrum disorders. Transl. Psychiatry 2016, 6, e808. [Google Scholar] [CrossRef] [Green Version]
  37. Muratori, F.; Billeci, L.; Calderoni, S.; Boncoddo, M.; Lattarulo, C.; Costanzo, V.; Turi, M.; Colombi, C.; Narzisi, A. How Attention to Faces and Objects Changes Over Time in Toddlers with Autism Spectrum Disorders: Preliminary Evidence from An Eye Tracking Study. Brain Sci. 2019, 9, 344. [Google Scholar] [CrossRef] [Green Version]
  38. Pierce, K.; Marinero, S.; Hazin, R.; McKenna, B.; Barnes, C.C.; Malige, A. 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, 657–666. [Google Scholar] [CrossRef] [Green Version]
  39. Murias, M.; Major, S.; Davlantis, K.; Franz, L.; Harris, A.; Rardin, B.; Sabatos-DeVito, M.; Dawson, G. Validation of eye-tracking measures of social attention as a potential biomarker for autism clinical trials. Autism Res. 2018, 11, 166–174. [Google Scholar] [CrossRef]
  40. Crehan, E.T.; Althoff, R.R. Me looking at you, looking at me: The stare-in-the-crowd effect and autism spectrum disorder. J. Psychiatr. Res. 2021, 140, 101–109. [Google Scholar] [CrossRef]
  41. Wang, Y.; Peng, S.; Shao, Z.; Feng, T. Active Viewing Facilitates Gaze to the Eye Region in Young Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2022, 7. [Google Scholar] [CrossRef]
  42. Barone, R.; Spampinato, C.; Pino, C.; Palermo, F.; Scuderi, A.; Zavattieri, A.; Gulisano, M.; Giordano, D.; Rizzo, R. Online comprehension across different semantic categories in preschool children with autism spectrum disorder. PLoS ONE 2019, 14, e0211802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Stevenson, R.A.; Philipp-Muller, A.; Hazlett, N.; Wang, Z.Y.; Luk, J.; Lee, J.; Black, K.R.; Yeung, L.K.; Shafai, F.; Segers, M.; et al. Conjunctive Visual Processing Appears Abnormal in Autism. Front. Psychol. 2019, 9, 2668. [Google Scholar] [CrossRef]
  44. Shou, G.; Mosconi, M.W.; Wang, J.; Ethridge, L.E.; Sweeney, J.A.; Ding, L. Electrophysiological signatures of atypical intrinsic brain connectivity networks in autism. J. Neural Eng. 2017, 14, 046010. [Google Scholar] [CrossRef] [PubMed]
  45. Gomot, M.; Blanc, R.; Clery, H.; Roux, S.; Barthelemy, C.; Bruneau, N. Candidate Electrophysiological Endophenotypes of Hyper-Reactivity to Change in Autism. J. Autism Dev. Disord. 2011, 41, 705–714. [Google Scholar] [CrossRef]
  46. Pagani, M.; Barsotti, N.; Bertero, A.; Trakoshis, S.; Ulysse, L.; Locarno, A.; Miseviciute, I.; De Felice, A.; Canella, C.; Supekar, K.; et al. mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity. Nat. Commun. 2021, 12, 6084. [Google Scholar] [CrossRef] [PubMed]
  47. Zerbi, V.; Pagani, M.; Markicevic, M.; Matteoli, M.; Pozzi, D.; Fagiolini, M.; Bozzi, Y.; Galbusera, A.; Scattoni, M.L.; Provenzano, G.; et al. Brain mapping across 16 autism mouse models reveals a spectrum of functional connectivity subtypes. Mol. Psychiatry 2021, 26, 7610–7620. [Google Scholar] [CrossRef]
  48. Yang, D.; Beam, D.; Pelphrey, K.; Abdullahi, S.; Jou, R. Cortical morphological markers in children with autism: A structural magnetic resonance imaging study of thickness, area, volume, and gyrification. Mol. Autism 2016, 7, 11. [Google Scholar] [CrossRef] [Green Version]
  49. Zwaigenbaum, L.; Bauman, M.L.; Choueiri, R.; Kasari, C.; Carter, A.; Granpeesheh, D.; Mailloux, Z.; Smith Roley, S.; Wagner, S.; Fein, D.; et al. Early Intervention for Children with Autism Spectrum Disorder Under 3 Years of Age: Recommendations for Practice and Research. Pediatrics 2015, 136 (Suppl. 1), S60–S81. [Google Scholar] [CrossRef] [Green Version]
  50. Klein, C.B.; Swain, D.M.; Vibert, B.; Clark-Whitney, E.; Lemelman, A.R.; Giordano, J.A.; Winter, J.; Kim, S.H. Implementation of Video Feedback Within a Community Based Naturalistic Developmental Behavioral Intervention Program for Toddlers with ASD: Pilot Study. Front. Psychiatry 2021, 12, 763367. [Google Scholar] [CrossRef]
  51. Whitehouse, A.J.; Varcin, K.J.; Alvares, G.A.; Barbaro, J.; Bent, C.; Boutrus, M.; Chetcuti, L.; Cooper, M.N.; Clark, A.; Davidson, E.; et al. Pre-emptive intervention versus treatment as usual for infants showing early behavioural risk signs of autism spectrum disorder: A single-blind, randomised controlled trial. Lancet Child Adolesc. Health 2019, 3, 605–615. [Google Scholar] [CrossRef] [Green Version]
  52. Poslawsky, I.E.; Naber, F.B.; Bakermans-Kranenburg, M.J.; Van Daalen, E.; Van Engeland, H.; Van Ijzendoorn, M.H. Video-feedback Intervention to promote Positive Parenting adapted to Autism (VIPP-AUTI): A randomized controlled trial. Autism 2015, 19, 588–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hong, M.P.; Erickson, C.A. Investigational drugs in early-stage clinical trials for autism spectrum disorder. Expert Opin. Investig. Drugs 2019, 28, 709–718. [Google Scholar] [CrossRef] [PubMed]
  54. Ruggeri, B.; Sarkans, U.; Schumann, G.; Persico, A. Biomarkers in autism spectrum disorder: The old and the new. Psychopharmacology 2014, 231, 1201–1216. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Narzisi, A.; Alonso-Esteban, Y.; Masi, G.; Alcantud-Marín, F. Research-Based Intervention (RBI) for Autism Spectrum Disorder: Looking beyond Traditional Models and Outcome Measures for Clinical Trials. Children 2022, 9, 430. https://doi.org/10.3390/children9030430

AMA Style

Narzisi A, Alonso-Esteban Y, Masi G, Alcantud-Marín F. Research-Based Intervention (RBI) for Autism Spectrum Disorder: Looking beyond Traditional Models and Outcome Measures for Clinical Trials. Children. 2022; 9(3):430. https://doi.org/10.3390/children9030430

Chicago/Turabian Style

Narzisi, Antonio, Yurena Alonso-Esteban, Gabriele Masi, and Francisco Alcantud-Marín. 2022. "Research-Based Intervention (RBI) for Autism Spectrum Disorder: Looking beyond Traditional Models and Outcome Measures for Clinical Trials" Children 9, no. 3: 430. https://doi.org/10.3390/children9030430

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop