Next Article in Journal
Cone-Beam Computed Tomography Evaluation of Bone Density After Sagittal Split Osteotomy Using the Novel Modification of Low Z Plasty Technique
Previous Article in Journal
Single-Center Experience with the HeartMate 3 Assist Device in Pediatric Patients with Cardiomyopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fecal Microbiota Transplantation for Autism Spectrum Disorder in Children: Results from a Prospective Open-Label Controlled Observational Study

by
Dominykas Varnas
1,*,
Arnas Kunevičius
2,
Aurelijus Burokas
2 and
Vaidotas Urbonas
1
1
Clinic of Children’s Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
2
Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
*
Author to whom correspondence should be addressed.
Medicina 2026, 62(1), 65; https://doi.org/10.3390/medicina62010065 (registering DOI)
Submission received: 7 November 2025 / Revised: 23 December 2025 / Accepted: 26 December 2025 / Published: 28 December 2025
(This article belongs to the Section Pediatrics)

Abstract

Background and Objectives: Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder with an increasing global incidence. Gut microbiota dysbiosis is believed to be playing a role in ASD pathogenesis. Fecal microbiota transplantation (FMT) is emerging as a potential therapeutic strategy to alleviate ASD-related and gastrointestinal symptoms, but data in pediatric ASD populations remain limited. Materials and Methods: We conducted a prospective, single-center, open-label controlled study to evaluate the efficacy of colonoscopic FMT in children with ASD. Participants were allocated to two groups: an intervention group that underwent a single FMT procedure and a control group. Gastrointestinal Symptoms Rating Scale (GSRS), Autism Diagnostic Observation Schedule (ADOS), Childhood Autism Rating Scale (CARS), Child Behavior Checklist (CBCL), and Parent Global Impression (PGI-R) scales were assessed for both groups at baseline and at set time points. Results: 30 participants were enrolled, with 15 in each group. At 8 weeks, no significant between-group differences were observed for the prespecified primary endpoint, change in ADOS scores. The intervention group showed significantly greater improvements in CARS (p < 0.001), PGI-R (p < 0.001), CBCL Internalizing Problems (p = 0.001), and GSRS (p = 0.037) compared with controls; CARS and PGI-R improvements persisted at 6 months. Within the intervention group, sustained improvements were noted in CARS, GSRS, and PGI-R up to 18 months. No serious adverse events were observed; three mild, self-limited adverse events were recorded following FMT. Conclusions: Colonoscopic FMT was associated with significant short-term improvements in gastrointestinal and caregiver-reported ASD symptoms (CARS), but not in ADOS scores. Some effects persisted long-term. However, due to a lack of blinding and possible selection bias, these findings should be interpreted as exploratory. Larger randomized controlled trials are needed to confirm efficacy and optimize protocols.

1. Introduction

ASD is a neurodevelopmental disorder characterized by persistent deficits in social interaction and communication, along with restricted, repetitive, and inflexible patterns of behavior, interests, or activities that are atypical for the individual’s age and sociocultural context [1]. The diagnosis can usually be established in children as early as 18–24 months of age [2]. It is a prevalent neurodevelopmental disorder with a number of cases estimated at around 28.3 million globally as of 2019 [3]. Over the last two decades, the prevalence of ASD has been increasing steadily from 1 in 150 [4] to 1 in 36 as of 2020 in the USA alone [5]. Predominantly, males are affected more often, with a male-to-female ratio of 3:1 [6]. There is no curative treatment for core ASD symptoms. The main treatment strategies involve behavioral and psychological interventions, while pharmacological options provide only a partial relief to adverse adaptive behaviors and comorbidities [7].
ASD pathogenesis is multifactorial and complex. Hundreds of genes have been identified to play a role in autism through neural development and synaptic function [8]. However, these genes can account for just 10–20% of cases [9]. More than 50% of neurobiological changes could be attributed to non-heritable factors [10]. Environmental triggers [11], immune system dysregulation, neuroinflammation [12], and the gut microbiota dysbiosis [13] are all implicated in ASD pathogenesis. Even though numerous advances have been made in recent years, the precise mechanisms, the interplay of different risk factors, and the influence on neurodevelopment remain incompletely understood [14,15].
Patients with ASD often exhibit a high comorbidity with gastrointestinal symptoms (GIS). GIS are found in about half the children with ASD. This rate is three to four times more than in neurotypical children (NC) [16,17,18]. Children with ASD were approximately six times more likely to experience multiple GIS than typically developing children. Children with ASD and co-occurring GIS exhibit more behavioral challenges—including self-injury, sensory sensitivities, sleep disturbances, attention difficulties, and aggressive behaviors—than those without GIS [19].
The gut microbiota has been proposed to play an important role in ASD [20,21]. Intestinal microbiota composition differs between children with ASD and neurotypical children [22,23,24]. This difference is seen in both human and animal models [25,26,27]. An important pilot study by Sandler et al., though limited in scope, demonstrated a short-term improvement in ASD symptoms in children during treatment with a minimally absorbed oral antibiotic (vancomycin). This study is among the earliest to provide evidence of the potential effectiveness of gut microbiota modulation therapy in children with ASD [28]. While the concept of the gut microbiota and brain connection was longstanding [29], one of the earliest mentions of the term “gut–brain axis” (GBA) is in a 2011 study by Grenham et al., in which the authors explored the complex bidirectional communication network between the central nervous system (CNS) and the gastrointestinal microbiota [30]. Since then, several possible mechanisms underlying the role of the microbiota and GBA in the pathogenesis of ASD have been elucidated [31,32].
The intestinal barrier is a semipermeable barrier and plays a pivotal role in ASD pathogenesis and in human health [33]. Increased intestinal permeability (sometimes referred to as “leaky gut”) allows bacterial products, allergens, and toxins to enter the gut, triggering local inflammation and reaching systemic circulation. De Magistris et al. found that 36.7% of children with ASD exhibited abnormal intestinal permeability, compared to less than 5% of controls [34]. Bacterial components such as lipopolysaccharides (LPS) can trigger low-grade neuroinflammation that can induce changes in the CNS [35]. Healthy gut microbiota maintains intestinal barrier integrity through multiple mechanisms. Beneficial bacteria, such as Lactobacillus and Bifidobacterium, promote tight junction stability. Akkermansia muciniphila enhances mucin production and reinforces the mucus layer, preventing pathogen adhesion to the epithelial surface. Commensal bacteria regulate gut-associated lymphoid tissue, promote regulatory T cell activity, and limit Gram-negative bacterial overgrowth. This helps reduce LPS leakage into circulation [36].
The gut microbiota produces different metabolites, including short-chain fatty acids (SCFAs) [37]. Alterations in the composition of the gut microbiota and its metabolites have been observed in both individuals with ASD and ASD animal models [38,39,40]. Studies reveal that fecal microbiota alterations resulting in an imbalance of certain SCFAs-producing bacteria can lead to exacerbation of ASD symptoms [41,42]. SCFAs modulate the inflammatory status of intestinal epithelial cells and inhibit LPS-induced cytokine production. They also play roles in energy metabolism, influence hormone secretion, and strengthen gut barrier integrity [43,44,45,46,47]. SCFAs act locally but can also cross the blood–brain barrier (BBB) via monocarboxylate transporters [48].
Certain gut bacteria synthesize neurotransmitters or their precursors that influence CNS function [49]. One of them is γ-Aminobutyric acid (GABA), mainly produced by Lactobacillus spp. and Bifidobacterium spp. [50,51], which is a primary inhibitory neurotransmitter. It controls neuronal excitability, participating in motility, immune cell activity, and secretory gastrointestinal functions [52]. Dysregulation of GABA signaling is a well-documented contributor to the pathophysiology of various neurological disorders, including ASD [53]. Gut microbiota alterations can dysregulate catecholaminergic systems, contributing to behavioral symptoms such as altered reward processing, attention deficits, heightened stress reactivity, and repetitive behaviors [54,55]. Dopamine pathway dysfunction has been implicated in ASD, particularly in relation to social motivation, executive function, and cognitive flexibility [56]. Norepinephrine imbalances are associated with sensory hypersensitivity and autonomic dysfunction. This causes challenges in sensory processing, autonomic regulation, and experiencing heightened responses to sensory input [57]. Additionally, epinephrine’s role in the stress response system suggests that microbial influences on catecholamine production may lead to difficulties in tolerating novel or benign environmental stressors [58]. Serotonin (5-HT) is involved in the regulation of gastrointestinal tract motility and secretion [59]. In the CNS, 5-HT signaling pathways are implicated in regulating mood, sleep, social behavior, cognitive flexibility, and neurodevelopment, including sensory development [60], all of which contribute to the pathophysiology of ASD [61]. While 95% of serotonin is produced by enterochromatophilic cells located within the intestinal mucosa [62], some gut microorganisms (Escherichia spp., Candida spp., Enterococcus spp.) can either produce or, mostly, indirectly influence the metabolism of 5-HT [63,64,65].
Unlike SCFAs, due to the BBB, neurotransmitters are unable to cross into the CNS (with the exception of low quantities of GABA) [66]. Their effects on the CNS are mediated by several mechanisms. Dysbiosis in the gut can alter tryptophan metabolism, reducing its availability as a serotonin precursor in the brain [67]. The vagus nerve provides bidirectional communication between the gut and the brain. Neurotransmitters produced in the gut can activate vagal afferents, which then modulate CNS function. Vagal nerve stimulation has been shown to improve core symptoms of ASD [68,69]. Additionally, a healthy gut microbiota is essential in preserving BBB integrity. SCFAs and their ability to modulate BBB permeability are well-known [70], but other microbial products, namely trimethylamine N-oxide, LPS, and secondary bile acid metabolites, also contribute [71,72]. A Disturbed BBB is not able to effectively prevent translocation of immune cells, inflammatory mediators, and microglial activation, all of which result in increased neuroinflammation, which is an important pathophysiological aspect of ASD [73,74].
Patients with ASD have dysfunction of the hypothalamic–pituitary–adrenal (HPA) axis and the autonomic nervous system, resulting in altered responses to stress and cortisol levels characteristic of ASD [58]. Gut microbiota-related mediators (prostaglandins, cytokines, microbial antigens) activate the HPA axis or suppress it, while the neurotransmitters they produce can, through the vagal nerve, activate the nucleus of the solitary tract and, in turn, the HPA axis [75]. Studies were conducted with germ-free animals that displayed exaggerated HPA axis responses to stress, which were normalized by introducing specific microbial species into their gut [76].
In light of this, microbiota modulation arose as a possible therapy to alter ASD and comorbid symptom severity. Probiotic/prebiotic supplementation has been researched in multiple studies, but current evidence is inconclusive due to a lack of high-quality randomized, double-blind, and placebo-controlled trials. Recommendations cannot be made regarding the efficacy of probiotics/prebiotics in ASD [77,78,79,80]. Given the limitations of probiotics, alternative microbiota-based interventions have gained attention. FMT is an established method for treating Clostridioides difficile infection [81] and is being researched for possible therapeutic uses in inflammatory bowel diseases, irritable bowel syndrome, metabolic syndrome, Parkinson’s disease, depression, and other conditions [82]. Unlike conventional probiotics, which are limited to one or several bacteria, FMT contains over a thousand bacterial species [83] as well as native gut viruses, archaea, fungi, and metabolites. A landmark study by Kang et al. showed promising FMT results by reducing autism and GI symptoms in children with ASD [84]. The effect was sustained even at 2 years of follow-up [85]. However, there is still a lack of understanding about the optimal strategy, including delivery route, donor screening, stool preparation, clinical indications, safety, and robust data regarding efficacy [82,86].
We present data from the first prospective, single-center, open-label, controlled study in Europe on the efficacy of FMT in pediatric patients with ASD.

2. Materials and Methods

2.1. Objective

The objective of this study was to evaluate the effectiveness of the FMT procedure as an investigatory treatment for children with ASD, focusing on its impact on both GIS and ASD symptoms.

2.2. Study Design and Ethical Considerations

This open-label study was performed at a tertiary university hospital between 2022 and 2024 and involved two groups of children with ASD aged 3–6 years. A total of 39 children were assessed for eligibility; nine did not participate in the study due to not meeting inclusion criteria (n = 4) or declining participation prior to informed consent (n = 5), and no study-related data were collected from these patients. The final study population consisted of 15 children in the intervention group who underwent a single FMT (ASDi group) and 15 children in the control group (ASDc), who did not receive FMT.
This study was conducted in accordance with the Declaration of Helsinki. It was approved by the Vilnius Regional Bioethics Committee, Vilnius University. Approval number 2022/4˗1400˗893. Written informed consent was obtained from all parents or legal guardians prior to the enrollment of each participant. Approval date: 12 April 2022.
Participants were not assigned to receive FMT by the research protocol. In our institution, FMT is performed under a pre-existing hospital clinical protocol that operates independently of any research activity. Decisions regarding whether a child should undergo FMT were made exclusively by a multidisciplinary clinical consilium based on individual clinical indications, independent of study.
Parents of children with ASD who were referred to our hospital to explore the possibility of FMT were informed about the procedure and were separately invited to participate in the present observational study. Children who underwent FMT under the clinical protocol and whose parents provided informed consent for study participation were included in the intervention group.
The control group consisted of children with ASD receiving standard care at our hospital during the same period. Parents of these children were also invited to participate in the study, but their children did not receive FMT. No randomization or matching was applied (Figure 1).
At our institution, standard care for children with ASD consists of individualized, multidisciplinary non-pharmacological interventions, including kinesitherapy, speech and language therapy (logotherapy), occupational therapy (ergotherapy), structured teaching, and art therapy. The selection, duration, and intensity of these interventions are tailored to each child and determined by treating developmental–behavioral pediatricians. Both groups were advised to continue their usual care throughout the study period. No protocol-mandated restrictions or changes to behavioral or pharmacological treatments were imposed as part of the study, and any therapy or medication decisions during follow-up were made at the discretion of the treating clinicians as part of routine care.
Children whose parents declined participation in the study continued to receive clinically indicated treatment; however, no study-related data were collected from these patients.
Patients were excluded from the study if they used antibiotic within 2 months or probiotic within 1 month prior to the procedure, or patient has been diagnosed inflammatory bowel disease (IBD), infectious gastrointestinal disease within the past 3 months, celiac disease, eosinophilic esophagitis, malnutrition, clinically significant structural brain abnormalities, or a history of abdominal surgery within the past 6 months.
To assess gastrointestinal symptoms, parents completed a modified GSRS [84]. The GSRS is a self-report questionnaire that utilizes a 7-point Likert scale, where responses range from 1 (asymptomatic) to 7 (very severe), with intermediate categories: 2 (slight), 3 (mild), 4 (moderate), 5 (moderate to severe), and 6 (severe).
Assessment of ASD symptoms was performeed using CARS and ADOS. CARS is a 15-item health care professional-rated measure with total scores ranging from 15 to 60, where higher scores indicate greater ASD symptoms [87]. ADOS is a standardized, semi-structured assessment administered by a single trained clinician [88]. The ADOS involves direct observation of social interaction, communication, play, and restricted/repetitive behaviors. To enable comparison across modules and developmental levels, raw scores were converted to calibrated severity scores (normalized ADOS scores), which provide a standardized measure of autism symptom severity.
The PGI-R is a parent-reported instrument designed to capture perceived changes in overall functioning and specific symptom domains in children with ASD [89]. It consists of 11 domains, including social interaction, communication, behavior, daily functioning, and other relevant developmental aspects. Each domain is rated on a 7-point scale of −3 to +3 (−3 = much worse, 0 = no change, and +3 = much better). The final score is the average score of all 11 domains. At baseline, all participants were assigned a PGI-R score of 0, reflecting no perceived change from the child’s usual condition at study entry and serving as the reference anchor for subsequent assessments. The PGI-R was selected because it provides a clinically meaningful, parent-centered assessment of behavioral and functional changes that may not be fully captured by clinician-administered scales.
Finally, behavioral and emotional functioning in children was assessed using the CBCL 1.5–5, part of the ASEBA [90]. It evaluates multiple domains, including internalizing, externalizing, and total behavioral problems, as reported by parents. Each item is rated on a 3-point scale (0 = not true, 1 = somewhat or sometimes true, 2 = very true or often true).

2.2.1. ASD Intervention Group

Before FMT, parents were asked to complete the GSRS and CBCL questionnaires. ASD symptoms were evaluated by a psychologist using the ADOS and CARS during a structured consultation.
After the FMT procedure, the GSRS and PGI-R questionnaires were completed at 1, 2, 3, 4, 8 weeks, and 6 and 18 months post-procedure. The CBCL questionnaire was asked to be completed at 2, 8 weeks, 6 months, and 18 months post-procedure. The ASD symptoms were evaluated using ADOS and CARS at 2, 8 weeks, 6 months, and 18 months post-procedure.

2.2.2. ASD Control Group

At the time of inclusion in the study, parents were asked to complete the GSRS and CBCL questionnaires. A psychologist evaluated ADOS and CARS scores. Follow-up assessments were conducted at 8 weeks and 6 months post-intervention. Parents completed the GSRS, CBCL, and PGI-R questionnaires, and autism symptoms were further evaluated by a psychologist using the ADOS and CARS.
This clinical study was approved by the local Ethics Committee for Scientific Research. Written informed consent was obtained from parents or caregivers before enrollment.

2.3. Intervention

For the FMT procedure, patients were prepared with 1.5–3 L polyethylene glycol 4000 (Fortrans) or sodium picosulphate (Picoprep) solution (weight-based dosage) the day before the procedure and in the morning.
No antibiotic pretreatment was administered prior to fecal microbiota transplantation, in accordance with institutional practice and in the absence of standardized pediatric recommendations.

2.3.1. Donor Eligibility and Exclusion Criteria

Fecal microbiota donors were three unrelated healthy children aged 3–7 years. Donors were required to be asymptomatic at the time of donation, and to have normal bowel habits and stool consistency. Donors with a history of food allergy were excluded. Other exclusion criteria included the use of antibiotics, probiotics, or any other medications 30 days prior to donation. Donors with a personal or family history of chronic diseases were excluded. Donor eligibility was assessed using a structured medical interview.
All donor screening investigations were performed within four weeks prior to fecal microbiota transplantation. Donors were screened once before stool donation, and only those with negative results on all screening tests were eligible to participate.

2.3.2. Stool Screening Panel

Stool screening included microscopic examination for protozoa and helminth ova and fecal occult blood testing. Viral testing was performed for rotavirus, norovirus, and adenovirus. Bacteriological (PCR) testing included Campylobacter spp., Salmonella spp., toxin-producing Escherichia coli, Shigella spp., Vibrio spp., Yersinia spp., Listeria monocytogenes, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), and detection of Clostridioides difficile toxins A and B.

2.3.3. Blood Screening Panel

Blood samples were obtained from all donors to exclude transmissible infections. Serological testing included: Hepatitis A virus IgM, Hepatitis B surface antigen (HBsAg), Hepatitis C virus antibodies (anti-HCV), Cytomegalovirus (CMV) IgM, Epstein–Barr virus (EBV) IgM, Human immunodeficiency virus (HIV) testing, Syphilis screening using the rapid plasma reagin (RPR) test.
Additional laboratory testing included: Complete blood count with erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP).

2.3.4. Stool Collection, Handling, and Administration

Stool samples were collected from donors and stored at +4 °C. In all cases, the interval between stool sample collection and FMT was less than 8 h. Stool samples were mixed with 150–200 mL of 0.9% NaCl and manually homogenized. The suspension was filtered to remove particulate matter. After preparation, the fecal suspension was administered within 1 h.
Prepared fecal microbiota suspension (final volume approximately 150–250 mL, depending on stool quantity) was administered during colonoscopy with delivery to the cecum. All procedures were performed under general anesthesia using sevoflurane administered via face mask. The total duration of the fecal microbiota transplantation procedure ranged from 10 to 20 min. No additional antiemetic or antispasmodic medications were administered during the procedure.

2.4. Statistical Analysis

Descriptive statistics were calculated for all dependent variables at each assessment time point. Variables are presented as means ± standard deviations (SD) with 95% confidence intervals (CI), while non-normally distributed variables are reported as medians with interquartile ranges (IQR).
All statistical analyses were performed using IBM SPSS Statistics (version 27; IBM Corp., Armonk, NY, USA) and Python (version 3.10; Python Software Foundation, Wilmington, DE, USA) for effect size and correction computations. The primary objective was to evaluate the effects of FMT on behavioral and gastrointestinal symptoms in children with ASD. The primary outcome was the change in normalized ADOS score at 8 weeks post-intervention.
Due to the small sample size (n = 15 per group), nonparametric tests were used throughout. Baseline characteristics and outcome scores were compared between the intervention and control groups using the Mann–Whitney U test.
Between-group comparisons at 8 weeks and 6 months were based on change scores from baseline (follow-up minus baseline). For outcomes with significant baseline imbalance (CBCL externalizing and total subscales, GSRS), a rank-based analysis of covariance (Quade test) was applied to adjust for baseline values.
Within-group longitudinal changes in the intervention group were assessed using the Friedman test. If significant, post hoc comparisons were conducted using the Wilcoxon signed-rank test in a predefined hierarchical order: Baseline vs. 2 weeks, followed by 8 weeks, 6 months, and 18 months. Testing stopped at the first non-significant comparison to control the familywise error rate, and no additional corrections (e.g., Bonferroni) were applied. Although all post hoc Wilcoxon comparisons are presented for completeness and transparency, only comparisons up to the first non-significant result within each outcome were considered confirmatory under the hierarchical gatekeeping procedure; subsequent comparisons are reported descriptively and should be interpreted as exploratory.
This gatekeeping procedure was chosen to balance statistical rigor with interpretability. For PGI-R, the default baseline score was 0, and comparisons were made relative to the anchor point.
Effect sizes for Wilcoxon tests were calculated using the formula ( r = Z N ) , where Z is the Wilcoxon test statistic and N is the number of observations (N = 15). Two-sided p-values < 0.05 were considered statistically significant.
No a priori sample size or power calculation was performed, as this study was conceived as an exploratory observational investigation conducted within a clinical care framework rather than as a confirmatory randomized trial.

3. Results

Baseline Characteristics and Group Comparisons
At baseline, no statistically significant differences were observed between the intervention and control groups on primary measures of autism severity, including normalized ADOS scores (p = 0.25) and CARS scores (p = 0.367). However, significant differences were noted in GSRS (Median IQR19.0 [17.0–23.0] vs. 27.0 [23.0–38.0]), CBCL Externalizing (10.0 [7.0–20.0] vs. 23.0 [11.0–27.0]) and CBCL Total scores (31.0 [27.0–50.0] vs. 49.0 [40.0–58.0]), with the intervention group reporting more severe symptoms at baseline (Table 1).
Descriptive statistics for both intervention and control groups can be found in Appendix A (Table A1 and Table A2, respectively).
Between-Group Differences at 8 Weeks and 6 Months
Mann–Whitney U tests of change scores revealed that, at 8 weeks post-intervention, the intervention group demonstrated significantly greater improvements in CARS by (p < 0.001), PGI-R (p < 0.001), and CBCL Internalizing Problems (p = 0.001) scores compared to the ASDc group. These effects were sustained at 6 months for CARS (p = 0.045) and PGI-R (p = 0.007), but not for CBCL Internalizing Problems (p= 0.25). No statistically significant group differences were observed in ADOS score at both time points (p = 0.061 and p = 0.367) (Table 2). Although no statistically significant between-group differences were observed for ADOS change scores, the corresponding effect sizes were moderate at 8 weeks (r ≈ 0.44) and small-to-moderate at 6 months (r ≈ 0.26), indicating that the non-significant findings may reflect limited statistical power rather than the absence of a clinically meaningful effect.
Due to baseline differences, rank-based ANCOVA (Quade test) was performed. After adjustment, the CBCL total problems subscale and GSRS showed significant between-group differences at 8 weeks (p = 0.001 and p = 0.037, respectively), but the effect was not observed at 6 months. No significant difference was observed on CBCL Externalizing Problems at both time points (Table 3, Figure 2).
Longitudinal symptom changes within the ASDi group
Friedman tests showed significant changes over time across all outcomes in the ASDi group (Table 4). Post hoc Wilcoxon signed-rank tests were then applied in a hierarchical fashion to compare baseline scores with each follow-up time point (2 weeks, 8 weeks, 6 months, and 18 months) for all outcome measures (Table 5, Figure 3). Reported are Z statistics, p-values, and effect sizes (r). Significant improvements were observed in CARS, GSRS, PGI-R, and CBCL subscales, with varying persistence across time points.
ADOS scores significantly improved at 2 weeks (p = 0.031) and 8 weeks (p = 0.016), but there were no significant changes at 6 or 18 months. CARS scores showed sustained improvement across all time points (p < 0.01).
CBCL internalizing and total problems improved consistently through 18 months. CBCL externalizing scores showed significant early reductions at 2 and 8 weeks (p = 0.001), but these gains were not sustained at 6 or 18 months.
GSRS scores demonstrated significant reductions in gastrointestinal symptoms from baseline to all post-intervention time points.
PGI-R scores improved significantly at all follow-up visits compared with baseline (2 weeks: Z = −3.11, p = 0.001, r = 0.80; 8 weeks: Z = −4.02, p < 0.001; 6 months: Z = −3.62, p < 0.001, r = 0.94; 18 months: Z = −2.68, p = 0.004, r = 0.69). Improvement was observed in 14 of 15 participants, resulting in extremely large effect sizes. At 8 weeks, the effect size calculation exceeded 1.0 due to the ceiling effect rather than error.

Safety Outcomes

Three adverse events were recorded following the FMT. Two patients experienced transient diarrhea accompanied by mild abdominal discomfort on the day of the procedure. Symptoms resolved spontaneously on the same day without intervention. One patient developed subfebrile fever and pharyngitis symptoms three days after the procedure, which resolved in an ambulatory setting without treatment and was considered unlikely to be related to FMT. No serious adverse events, procedural complications, or hospitalizations occurred. No unexpected clinical findings were observed during the 18-month follow-up period.

4. Discussion

Several previous open-label studies suggested that FMT can improve both gastrointestinal and ASD-related symptoms [84,91,92,93,94,95,96]. However, these studies lacked a control group. Since no standardized intervention model for FMT in children with ASD exists, patient preparation procedures (antibiotics, bowel cleansing, gastric acid suppression), administration routes of FMT, and donor selection varied significantly.
We decided not to use antibiotic pretreatment because the role of antibiotics in facilitating FMT engraftment remains uncertain, with conflicting evidence and no consensus regarding optimal regimens, particularly in pediatric populations with ASD [81,97].
To the best of our knowledge, only two randomized controlled trials (RCTs) have been published with conflicting results. A study by Wang et al. [98] reported improvement in both GI and ASD symptoms, while Wan et al. [99] found no significant ASD-related symptom improvement.
These discrepancies may be explained by differences in study design and patient selection. Wang et al. employed a multi-step intervention, including antibiotic pretreatment and bowel cleansing before repeated oral FMT capsule administration, and reported consistent improvements in both gastrointestinal and ASD symptoms [98]. In contrast, Wan et al. administered two short courses of oral FMT capsules without antibiotics and did not observe significant differences from placebo on primary (SRS-2) or secondary (Vineland-3, ABC) outcomes, despite within-group improvements in both arms [99]. Unlike in Wang and Wan studies, we used one-time FMT via colonoscopy, because it ensures transplant administration to the cecum, and the effectiveness in treating C. difficile colitis of this route is comparable to capsules and superior to other administration routes [100]. Importantly, we also used the CARS as one of our outcomes, consistent with Wang et al., which facilitates comparison between studies. In addition, we included the ADOS, a gold-standard, clinician-administered measure that was not applied in any of the mentioned trials. This allowed us to capture both caregiver-reported improvements and clinician-rated outcomes, revealing a divergence that may reflect expectancy bias or differences in sensitivity between instruments.
The primary endpoint, change in ADOS scores at 8 weeks, did not differ significantly between groups. However, several secondary outcomes showed meaningful improvement in the intervention group compared with untreated controls. Children receiving FMT demonstrated greater reductions in CARS scores at both 8 weeks and 6 months, alongside significant improvements in caregiver-reported PGI-R scores. GSRS and CBCL Internalizing and Total Problems improved at the 8-week time point only. Improvements evident at the 8-week time point but not maintained at 6 months may reflect transient changes in gut microbiota after FMT, with partial reversion over time in the absence of repeated dosing. Alternatively, expectancy effects in an open-label design may be strongest shortly after treatment but attenuate during longer follow-up.
The decision to administer a single colonoscopic FMT was driven by practical and methodological considerations. At present, there are no consensus recommendations regarding the optimal number or frequency of FMT procedures for children with ASD [82,86]. Colonoscopic delivery requires general anesthesia and carries a procedural burden, making repeated administrations less feasible, especially in pediatric populations. A single FMT allowed assessment of the clinical impact of a single, clearly defined intervention.
While no differences were detected in ADOS scores between groups, CARS scores showed significant improvements following FMT. Both scales measure autism severity, yet they differ in sensitivity and context. ADOS is examiner-administered, based on structured tasks, which may not fully reflect improvements in daily behavior. ADOS was primarily developed as a diagnostic instrument rather than an outcome measure, and its standardized, time-limited observational format may limit sensitivity to short-term behavioral change [101].
Differences in the psychometric properties of these instruments may also contribute. ADOS has relatively coarse scoring ranges (items scored 0–3, then summed), and total scores are mapped to calibrated severity scores. This compresses variability and may reduce sensitivity to small treatment-related shifts. Previous works noted that ADOS total and calibrated severity scores tend to remain relatively stable over short follow-up intervals, which may further constrain their ability to capture subtle treatment-related improvements over weeks to a few months [102,103,104].
In contrast, CARS includes 15 items scored on a 7-point scale, offering greater resolution to detect subtle improvements. Additionally, CARS incorporates caregiver interview data alongside direct observation, allowing ratings to reflect behavior across a broader range of daily situations like emotional responses and adaptability that may have been influenced not only by changes in core ASD symptoms, but also by improvements in emotional regulation, adaptability, and reduction of GI-related distress [101]. However, CARS is more subjective and may be influenced by expectancy bias, particularly in an open-label design where caregivers were aware of treatment assignment. Placebo responses are well documented in autism intervention studies, with larger placebo effects consistently observed when outcomes are reported by caregivers rather than clinicians [104]. Such expectancy-related effects may preferentially influence global or context-dependent measures such as CARS, whereas structured clinician-administered instruments, such as ADOS, conducted in standardized settings, may be less sensitive to these influences [105]. This difference between scales may partly explain why CARS, but not ADOS, showed significance between groups.
Beyond differences in psychometric properties and possible expectancy effects, the discrepancy between caregiver-reported improvements (CARS, PGI-R) and the lack of significant between-group effects on ADOS may also be viewed in relation to sensory regulation and neurophysiological stress in ASD. Children with ASD and co-occurring sensory processing difficulties often show heightened stress responses. Reductions in physical or sensory discomfort are more often noticed by caregivers in everyday contexts than by clinicians using structured observational tools. It is relevant since improvements in gastrointestinal symptoms following FMT may plausibly reduce ongoing visceral discomfort and associated stress. In turn, parents may observe changes in emotional regulation, adaptability, and daily functioning that are less readily captured by ADOS scores, particularly over relatively short follow-up periods. Although neurophysiological stress markers were not assessed in the present study, this framework offers a plausible mechanistic explanation for the greater sensitivity of parent-based instruments [106].
These considerations indicate that the discrepancy between ADOS and CARS reflects a divergence that may arise from differences in instrument sensitivity, expectancy bias, and measurement context, or all of these, rather than necessarily indicating inconsistency in the observed clinical response.
Analyses within the ASDi group revealed sustained improvements in multiple domains, with CARS, GSRS, and PGI-R persisting up to 18 months post-treatment. However, the control group was followed for only 6 months. Due to this, these long-term findings cannot be attributed solely to the intervention. Long-term developmental trajectories in ASD are highly heterogeneous and influenced by baseline severity, comorbidities, and concurrent interventions, as demonstrated in previous longitudinal studies [107,108]. In addition, although a hierarchical gatekeeping procedure was applied to control the familywise error rate within longitudinal analyses, the evaluation of multiple secondary outcomes across several domains and time points increases the potential for residual Type I error, further supporting an exploratory interpretation of these findings.
These observations support the long-term benefits reported in the follow-up study by Kang et al. [85]. There are no other studies with a long follow-up. Since within-group comparisons cannot fully rule out the placebo effect or natural disease development, these results should be interpreted cautiously.
Finally, we chose the colonoscopic delivery route for FMT as oral routes require either a tube placement or swallowing a significant amount of capsules for several days, which is problematic for children with ASD. The only study comparing the effectiveness of FMT in children with ASD was by Li et al. [91], in which no significant difference was observed between oral or colonoscopic FMT administration.

Limitations

This study has several methodological limitations. Although outcomes were assessed prospectively, assignment to FMT or standard care was not randomized and followed routine clinical decision-making under established hospital protocol, with parental agreement. Therefore, selection bias remains a possibility.
Baseline differences between groups should also be considered when interpreting the results. The intervention group showed higher GSRS and CBCL scores at the beginning of the study, raising the possibility that some of the observed improvement may partly reflect regression to the mean rather than a true treatment effect. We addressed this by applying baseline-adjusted rank-based ANCOVA (Quade test) for outcomes with significant baseline differences. Nevertheless, statistical adjustment cannot fully eliminate regression to the mean in a non-randomized study.
Follow-up frequency differed between the intervention and control groups. Patients who underwent FMT were monitored more frequently, particularly early after the procedure, reflecting clinical safety considerations given the invasive nature of the intervention. The control group received standard care, and intensive short-term monitoring was not required, thereby avoiding unnecessary burden. This asymmetry in follow-up schedules may have introduced observation bias, particularly for caregiver-reported outcomes, and should be considered when interpreting the results.
No microbiome sequencing or biomarker analyses were performed in this study; therefore, mechanistic interpretations regarding gut–brain pathways remain speculative and cannot be directly inferred from the present data.
While a comparison group was included, the absence of randomization and blinding limits causal inference and increases susceptibility to expectancy bias. The absence of blinding is particularly relevant for caregiver-reported outcomes such as CARS, PGI-R, GSRS, and CBCL, which may overestimate treatment effects. In addition, the small, clinically heterogeneous study population limits generalizability. No a priori power calculation was performed, and with only 15 participants per group, the study was likely underpowered to detect small-to-moderate between-group differences, particularly for clinician-administered measures such as ADOS. Accordingly, the findings should be interpreted as exploratory and hypothesis-generating, underscoring the need for larger, randomized, and blinded trials.

5. Conclusions

In this open-label controlled study, the prespecified primary endpoint—change in clinician-administered ADOS scores at 8 weeks—was not significantly different between children who underwent FMT and controls. Secondary outcomes, including gastrointestinal symptoms and caregiver-reported measures of autism symptom severity (CARS), showed improvements in the intervention group. While the persistence of improvements in some domains up to 18 months is encouraging, the absence of randomization and blinding underscores the need for cautious interpretation. Taken together, these findings should be regarded as exploratory and hypothesis-generating, supporting the rationale for larger, blinded, and methodologically rigorous trials with standardized protocols to determine efficacy and the best routes and FMT preparations.

Author Contributions

Conceptualization, D.V., A.B., and V.U.; methodology, D.V. and V.U.; formal analysis, D.V. and A.K.; investigation, D.V.; data curation, D.V. and A.K.; visualization, D.V. and A.K.; writing—original draft preparation, D.V. and A.K.; writing—review and editing, A.B. and V.U.; supervision, V.U.; funding acquisition, V.U. and A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This project has received funding from the European Regional Development Fund (project No. 01.2.2-LMT-K-718-03-0099 under grant agreement with the Research Council of Lithuania (LMTLT).

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki, and approved by Vilnius Regional Bioethics Committee (Approval No.: 2022/4-1400-893). Approved 12 April 2022.

Informed Consent Statement

Written informed consent was obtained from all parents or legal guardians prior to the enrollment of each participant.

Data Availability Statement

The raw de-identified data presented in this study are available on reasonable request from the corresponding author. The data are not publicly available due to ethical restrictions related to pediatric patient confidentiality.

Acknowledgments

We thank all the participating children and their families for their cooperation and contribution of samples. We also express our gratitude to Akvilė Marcikonytė for conducting the psychological evaluations, and to our endoscopists, Julija Červinskienė and Benas Prušinskas, for their assistance in performing the fecal microbiota transplantation procedures.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ASDAutism spectrum disorder
FMTFecal microbiota transplantation
GSRSGastrointestinal Symptoms Rating Scale
ADOSAutism Diagnostic Observation Schedule
CARSChildhood Autism Rating Scale
ASEBAAchenbach System of Empirically Based Assessment
PGI-RParent Global Impression
GISGastrointestinal symptoms
NCNeurotypical children
GBAGut–brain axis
CNSCentral nervous system
LPSLipopolysaccharides
SCFAsShort-chain fatty acids
BBBBlood–brain barrier
GABAγ-Aminobutyric acid
5-HTSerotonin
HPAHypothalamic–pituitary–adrenal
ASDiAutism spectrum disorder intervention
ASDcAutism spectrum disorder control
IBDInflammatory bowel disease
RCTRandomized controlled trial

Appendix A

Table A1. Descriptive statistics, intervention group (ASDi).
Table A1. Descriptive statistics, intervention group (ASDi).
GroupVariableTime PointMean ± SDMedian [IQR]
ASDiADOSBaseline7.9 ± 2.08.0 [6.0–10.0]
ASDiCARSBaseline38.8 ± 3.339.0 [37.0–41.0]
ASDiGSRSBaseline29.7 ± 9.027.0 [23.0–38.0]
ASDiCBCL Internalizing ProblemsBaseline23.6 ± 5.724.0 [19.0–28.0]
ASDiCBCL Externalizing ProblemsBaseline21.1 ± 10.923.0 [11.0–27.0]
ASDiCBCL Total ProblemsBaseline49.1 ± 12.349.0 [40.0–58.0]
ASDiGSRS1 week21.9 ± 8.319.0 [17.0–26.0]
ASDiGSRS2 weeks21.7 ± 6.519.0 [17.0–23.0]
ASDiCBCL Internalizing Problems2 weeks21.0 ± 5.322.0 [18.0–24.0]
ASDiCBCL Externalizing Problems2 weeks18.6 ± 10.217.0 [10.0–22.0]
ASDiCBCL Total Problems2 weeks43.7 ± 12.743.0 [34.0–49.0]
ASDiADOS2 weeks7.5 ± 2.18.0 [5.0–10.0]
ASDiCARS2 weeks35.3 ± 3.436.0 [32.0–38.0]
ASDiGSRS3 weeks22.5 ± 5.921.0 [17.0–25.0]
ASDiGSRS4 weeks24.7 ± 9.121.0 [18.0–29.0]
ASDiGSRS8 weeks22.3 ± 5.221.0 [18.0–26.0]
ASDiCBCL Internalizing Problems8 weeks21.5 ± 5.620.0 [18.0–26.0]
ASDiCBCL Externalizing Problems8 weeks19.1 ± 10.220.0 [10.0–22.0]
ASDiCBCL Total Problems8 weeks44.9 ± 12.944.0 [34.0–52.0]
ASDiADOS8 weeks7.5 ± 2.17.0 [5.0–10.0]
ASDiCARS8 weeks35.3 ± 3.435.0 [32.5–38.0]
ASDiGSRS6 months24.4 ± 5.723.0 [20.0–28.0]
ASDiCBCL Internalizing Problems6 months22.7 ± 5.723.0 [19.0–28.0]
ASDiCBCL Externalizing Problems6 months20.5 ± 10.621.0 [10.0–26.0]
ASDiCBCL Total Problems6 months47.3 ± 12.648.0 [40.0–56.0]
ASDiADOS6 months7.7 ± 2.28.0 [5.0–10.0]
ASDiCARS6 months36.9 ± 3.738.0 [33.0–40.0]
ASDiGSRS18 months27.0 ± 7.825.0 [20.0–35.0]
ASDiCBCL Internalizing Problems18 months22.8 ± 5.623.0 [19.0–27.0]
ASDiCBCL Externalizing Problems18 months20.6 ± 10.621.0 [11.0–28.0]
ASDiCBCL Total problems18 months47.6 ± 12.648.0 [41.0–55.0]
ASDiADOS18 months7.7 ± 2.18.0 [6.0–10.0]
ASDiCARS18 months37.4 ± 3.537.0 [34.0–41.0]
ASDiPGI-R1 week0.55 ± 0.510.45 [0.27–0.91]
ASDiPGI-R2 weeks0.66 ± 0.60.45 [0.27–0.91]
ASDiPGI-R3 weeks0.69 ± 0.550.55 [0.27–0.91]
ASDiPGI-R4 weeks0.67 ± 0.420.64 [0.27–0.82]
ASDiPGI-R8 weeks0.65 ± 0.460.45 [0.36–0.82]
ASDiPGI-R6 months0.35 ± 0.360.36 [0.09–0.73]
ASDiPGI-R18 months0.25 ± 0.360.18 [0.00–0.27]
Table A2. Descriptive statistics control group (ASDc).
Table A2. Descriptive statistics control group (ASDc).
GroupVariableTime PointMean ± SDMedian [IQR]
ASDcADOSBaseline6.9 ± 2.46.0 [5.0–10.0]
ASDcCARSBaseline37.1 ± 4.538.0 [34.0–41.0]
ASDcGSRSBaseline21.0 ± 5.619.0 [17.0–23.0]
ASDcCBCL Internalizing ProblemsBaseline21.7 ± 5.721.0 [16.0–27.0]
ASDcCBCL Externalizing ProblemsBaseline13.2 ± 8.010.0 [7.0–20.0]
ASDcCBCL Total ProblemsBaseline37.4 ± 12.531.0 [27.0–50.0]
ASDcGSRS8 weeks21.0 ± 6.819.0 [17.0–22.0]
ASDcCBCL Internalizing Problems8 weeks21.7 ± 5.121.0 [18.0–27.0]
ASDcCBCL Externalizing Problems8 weeks12.5 ± 7.810.0 [6.0–18.0]
ASDcCBCL Total Problems8 weeks36.6 ± 11.931.0 [27.0–48.0]
ASDcADOS8 weeks6.8 ± 2.46.0 [5.0–9.0]
ASDcCARS8 weeks37.4 ± 4.738.0 [34.0–41.0]
ASDcGSRS6 months21.0 ± 6.819.0 [17.0–22.0]
ASDcCBCL Internalizing Problems6 months21.5 ± 5.421.0 [16.0–26.0]
ASDcCBCL Externalizing Problems6 months12.7 ± 7.810.0 [7.0–19.0]
ASDcCBCL Total Problems6 months36.7 ± 12.230.0 [26.0–50.0]
ASDcADOS6 months6.9 ± 2.46.0 [5.0–10.0]
ASDcCARS6 months37.6 ± 5.437.0 [33.0–41.0]
ASDcPGI-R8 weeks0.04 ± 0.060.00 [0.0–0.09]
ASDcPGI-R6 months0.07 ± 0.070.09 [0.0–0.09]

References

  1. World Health Organization. International Classification of Diseases, 11th Revision (ICD-11) for Mortality and Morbidity Statistics, 2024–01Release. World Health Organization, 2024. Available online: https://icd.who.int/browse/2024-01/mms/en#437815624 (accessed on 3 January 2025).
  2. Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism spectrum disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef] [PubMed]
  3. Li, Y.-A.; Chen, Z.-J.; Li, X.-D.; Gu, M.-H.; Xia, N.; Gong, C.; Zhou, Z.-W.; Yasin, G.; Xie, H.-Y.; Wei, X.-P.; et al. Epidemiology of autism spectrum disorders: Global burden of disease 2019 and bibliometric analysis of risk factors. Front. Pediatr. 2022, 10, 972809. [Google Scholar] [CrossRef] [PubMed]
  4. Centers for Disease Control and Prevention. Prevalence of Autism Spectrum Disorders—Autism and Developmental Disabilities Monitoring Network, Six Sites, United States, 2007. Available online: https://www.cdc.gov/mmwr/preview/mmwrhtml/ss5601a1.htm (accessed on 3 January 2025).
  5. Maenner, M.J.; Warren, Z.; Williams, A.R.; Amoakohene, E.; Bakian, A.V.; Bilder, D.A.; Durkin, M.S.; Fitzgerald, R.T.; Furnier, S.M.; Hughes, M.M.; et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2020. MMWR Surveill. Summ. 2023, 72, 1–14. [Google Scholar] [CrossRef] [PubMed]
  6. Li, Z.; Yang, L.; Chen, H.; Fang, Y.; Zhang, T.; Yin, X.; Man, J.; Yang, X.; Lu, M. Global, regional and national burden of autism spectrum disorder from 1990 to 2019: Results from the Global Burden of Disease Study 2019. Epidemiol. Psychiatr. Sci. 2022, 31, e33. [Google Scholar] [CrossRef]
  7. Sharma, S.R.; Gonda, X.; Tarazi, F.I. Autism Spectrum Disorder: Classification, diagnosis and therapy. Pharmacol. Ther. 2018, 190, 91–104. [Google Scholar] [CrossRef]
  8. Jiang, C.-C.; Lin, L.-S.; Long, S.; Ke, X.-Y.; Fukunaga, K.; Lu, Y.-M.; Han, F. Signalling pathways in autism spectrum disorder: Mechanisms and therapeutic implications. Signal Transduct. Target. Ther. 2022, 7, 229. [Google Scholar] [CrossRef]
  9. Rylaarsdam, L.; Guemez-Gamboa, A. Genetic Causes and Modifiers of Autism Spectrum Disorder. Front. Cell Neurosci. 2019, 13, 385. [Google Scholar] [CrossRef]
  10. Cheroni, C.; Caporale, N.; Testa, G. Autism spectrum disorder at the crossroad between genes and environment: Contributions, convergences, and interactions in ASD developmental pathophysiology. Mol. Autism 2020, 11, 69. [Google Scholar] [CrossRef]
  11. Duque-Cartagena, T.; Dalla, M.D.B.; Mundstock, E.; Neto, F.K.; Espinoza, S.A.R.; De Moura, S.K.; Zanirati, G.; Padoin, A.V.; Jimenez, J.G.P.; Stein, A.T.; et al. Environmental pollutants as risk factors for autism spectrum disorders: A systematic review and meta-analysis of cohort studies. BMC Public Health 2024, 24, 2388. [Google Scholar] [CrossRef]
  12. Meltzer, A.; Van De Water, J. The Role of the Immune System in Autism Spectrum Disorder. Neuropsychopharmacology 2017, 42, 284–298. [Google Scholar] [CrossRef]
  13. Hughes, H.K.; Rose, D.; Ashwood, P. The Gut Microbiota and Dysbiosis in Autism Spectrum Disorders. Curr. Neurol. Neurosci. Rep. 2018, 18, 81. [Google Scholar] [CrossRef] [PubMed]
  14. Horecka-Lewitowicz, A.; Lewitowicz, W.; Wawszczak-Kasza, M.; Lim, H.; Lewitowicz, P. Autism Spectrum Disorder Pathogenesis—A Cross-Sectional Literature Review Emphasizing Molecular Aspects. Int. J. Mol. Sci. 2024, 25, 11283. [Google Scholar] [CrossRef] [PubMed]
  15. Beopoulos, A.; Géa, M.; Fasano, A.; Iris, F. Autism spectrum disorders pathogenesis: Toward a comprehensive model based on neuroanatomic and neurodevelopment considerations. Front. Neurosci. 2022, 16, 988735. [Google Scholar] [CrossRef] [PubMed]
  16. Holingue, C.; Newill, C.; Lee, L.; Pasricha, P.J.; Daniele Fallin, M. Gastrointestinal symptoms in autism spectrum disorder: A review of the literature on ascertainment and prevalence. Autism Res. 2018, 11, 24–36. [Google Scholar] [CrossRef]
  17. McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal Symptoms in Autism Spectrum Disorder: A Meta-analysis. Pediatrics 2014, 133, 872–883. [Google Scholar] [CrossRef]
  18. Leader, G.; Abberton, C.; Cunningham, S.; Gilmartin, K.; Grudzien, M.; Higgins, E.; Joshi, L.; Whelan, S.; Mannion, A. Gastrointestinal Symptoms in Autism Spectrum Disorder: A Systematic Review. Nutrients 2022, 14, 1471. [Google Scholar] [CrossRef]
  19. Restrepo, B.; Angkustsiri, K.; Taylor, S.L.; Rogers, S.J.; Cabral, J.; Heath, B.; Hechtman, A.; Solomon, M.; Ashwood, P.; Amaral, D.G.; et al. Developmental–behavioral profiles in children with autism spectrum disorder and co-occurring gastrointestinal symptoms. Autism Res. 2020, 13, 1778–1789. [Google Scholar] [CrossRef]
  20. Cryan, J.F. Microbiome and Brain Development: A Tale of Two Systems. Ann. Nutr. Metab. 2025, 81, 34–46. [Google Scholar] [CrossRef]
  21. Burokas, A.; Moloney, R.D.; Dinan, T.G.; Cryan, J.F. Microbiota Regulation of the Mammalian Gut–Brain Axis. In Advances in Applied Microbiology; Elsevier: Amsterdam, The Netherlands, 2015; pp. 1–62. [Google Scholar] [CrossRef]
  22. Andreo-Martínez, P.; Rubio-Aparicio, M.; Sánchez-Meca, J.; Veas, A.; Martínez-González, A.E. A Meta-analysis of Gut Microbiota in Children with Autism. J. Autism Dev. Disord. 2022, 52, 1374–1387. [Google Scholar] [CrossRef]
  23. Iglesias-Vázquez, L.; Van Ginkel Riba, G.; Arija, V.; Canals, J. Composition of Gut Microbiota in Children with Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 792. [Google Scholar] [CrossRef]
  24. Yang, C.; Xiao, H.; Zhu, H.; Du, Y.; Wang, L. Revealing the gut microbiome mystery: A meta-analysis revealing differences between individuals with autism spectrum disorder and neurotypical children. Biosci. Trends 2024, 18, 233–249. [Google Scholar] [CrossRef]
  25. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549, 528–532. [Google Scholar] [CrossRef]
  26. Golubeva, A.V.; Joyce, S.A.; Moloney, G.; Burokas, A.; Sherwin, E.; Arboleya, S.; Flynn, I.; Khochanskiy, D.; Moya-Pérez, A.; Peterson, V.; et al. Microbiota-related Changes in Bile Acid & Tryptophan Metabolism are Associated with Gastrointestinal Dysfunction in a Mouse Model of Autism. eBioMedicine 2017, 24, 166–178. [Google Scholar] [CrossRef]
  27. Butler, M.I.; Bastiaanssen, T.F.S.; Long-Smith, C.; Morkl, S.; Berding, K.; Ritz, N.L.; Strain, C.; Patangia, D.; Patel, S.; Stanton, C.; et al. The gut microbiome in social anxiety disorder: Evidence of altered composition and function. Transl. Psychiatry 2023, 13, 95. [Google Scholar] [CrossRef]
  28. Sandler, R.H.; Finegold, S.M.; Bolte, E.R.; Buchanan, C.P.; Maxwell, A.P.; Väisänen, M.-L.; Nelson, M.N.; Wexler, H.M. Short-Term Benefit From Oral Vancomycin Treatment of Regressive-Onset Autism. J. Child. Neurol. 2000, 15, 429–435. [Google Scholar] [CrossRef]
  29. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef]
  30. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain?Gut?Microbe Communication in Health and Disease. Front. Physio 2011, 2, 94. [Google Scholar] [CrossRef]
  31. Alharthi, A.; Alhazmi, S.; Alburae, N.; Bahieldin, A. The Human Gut Microbiome as a Potential Factor in Autism Spectrum Disorder. Int. J. Mol. Sci. 2022, 23, 1363. [Google Scholar] [CrossRef]
  32. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  33. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef] [PubMed]
  34. De Magistris, L.; Familiari, V.; Pascotto, A.; Sapone, A.; Frolli, A.; Iardino, P.; Carteni, M.; De Rosa, M.; Francavilla, R.; Riegler, G.; et al. Alterations of the Intestinal Barrier in Patients with Autism Spectrum Disorders and in Their First-degree Relatives. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 418–424. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, J.; Bi, W.; Xiao, S.; Lan, X.; Cheng, X.; Zhang, J.; Lu, D.; Wei, W.; Wang, Y.; Li, H.; et al. Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci. Rep. 2019, 9, 5790. [Google Scholar] [CrossRef] [PubMed]
  36. Chernikova, M.A.; Flores, G.D.; Kilroy, E.; Labus, J.S.; Mayer, E.A.; Aziz-Zadeh, L. The Brain-Gut-Microbiome System: Pathways and Implications for Autism Spectrum Disorder. Nutrients 2021, 13, 4497. [Google Scholar] [CrossRef] [PubMed]
  37. Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S.; Giustina, A.; Gazzaruso, C. Microbiota and metabolic diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef]
  38. Zhang, S.; Zhao, J.; Xie, F.; He, H.; Johnston, L.J.; Dai, X.; Wu, C.; Ma, X. Dietary fiber-derived short-chain fatty acids: A potential therapeutic target to alleviate obesity-related nonalcoholic fatty liver disease. Obes. Rev. 2021, 22, e13316. [Google Scholar] [CrossRef]
  39. He, J.; Gong, X.; Hu, B.; Lin, L.; Lin, X.; Gong, W.; Zhang, B.; Cao, M.; Xu, Y.; Xia, R.; et al. Altered Gut Microbiota and Short-chain Fatty Acids in Chinese Children with Constipated Autism Spectrum Disorder. Sci. Rep. 2023, 13, 19103. [Google Scholar] [CrossRef]
  40. Choi, J.; Lee, S.; Won, J.; Jin, Y.; Hong, Y.; Hur, T.-Y.; Kim, J.-H.; Lee, S.-R.; Hong, Y. Pathophysiological and neurobehavioral characteristics of a propionic acid-mediated autism-like rat model. PLoS ONE 2018, 13, e0192925. [Google Scholar] [CrossRef]
  41. Korteniemi, J.; Karlsson, L.; Aatsinki, A. Systematic review: Autism spectrum disorder and the gut microbiota. Acta Psychiatr. Scand. 2023, 148, 242–254. [Google Scholar] [CrossRef]
  42. Lagod, P.P.; Naser, S.A. The Role of Short-Chain Fatty Acids and Altered Microbiota Composition in Autism Spectrum Disorder: A Comprehensive Literature Review. Int. J. Mol. Sci. 2023, 24, 17432. [Google Scholar] [CrossRef]
  43. Boets, E.; Gomand, S.V.; Deroover, L.; Preston, T.; Vermeulen, K.; De Preter, V.; Hamer, H.M.; Van Den Mooter, G.; De Vuyst, L.; Courtin, C.M.; et al. Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: A stable isotope study. J. Physiol. 2017, 595, 541–555. [Google Scholar] [CrossRef]
  44. Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.K.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [PubMed]
  45. Peng, L.; Li, Z.-R.; Green, R.S.; Holzmanr, I.R.; Lin, J. Butyrate Enhances the Intestinal Barrier by Facilitating Tight Junction Assembly via Activation of AMP-Activated Protein Kinase in Caco-2 Cell Monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [PubMed]
  46. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed]
  47. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
  48. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  49. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  50. Lyte, M. Microbial Endocrinology and the Microbiota-Gut-Brain Axis. In Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease; Lyte, M., Cryan, J.F., Eds.; Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2014; pp. 3–24. [Google Scholar] [CrossRef]
  51. Otaru, N.; Ye, K.; Mujezinovic, D.; Berchtold, L.; Constancias, F.; Cornejo, F.A.; Krzystek, A.; De Wouters, T.; Braegger, C.; Lacroix, C.; et al. GABA Production by Human Intestinal Bacteroides spp.: Prevalence, Regulation, and Role in Acid Stress Tolerance. Front. Microbiol. 2021, 12, 656895. [Google Scholar] [CrossRef]
  52. Auteri, M.; Zizzo, M.G.; Serio, R. GABA and GABA receptors in the gastrointestinal tract: From motility to inflammation. Pharmacol. Res. 2015, 93, 11–21. [Google Scholar] [CrossRef]
  53. Braat, S.; Kooy, R.F. The GABAA Receptor as a Therapeutic Target for Neurodevelopmental Disorders. Neuron 2015, 86, 1119–1130. [Google Scholar] [CrossRef]
  54. Koevoet, D.; Deschamps, P.K.H.; Kenemans, J.L. Catecholaminergic and cholinergic neuromodulation in autism spectrum disorder: A comparison to attention-deficit hyperactivity disorder. Front. Neurosci. 2023, 16, 1078586. [Google Scholar] [CrossRef]
  55. Averina, O.V.; Danilenko, V.N. Human intestinal microbiota: Role in development and functioning of the nervous system. Microbiology 2017, 86, 5–24. [Google Scholar] [CrossRef] [PubMed]
  56. Pavăl, D. The dopamine hypothesis of autism spectrum disorder: A comprehensive analysis of the evidence. In International Review of Neurobiology; Elsevier: Amsterdam, The Netherlands, 2023; pp. 1–42. [Google Scholar] [CrossRef]
  57. Beversdorf, D.Q. The Role of the Noradrenergic System in Autism Spectrum Disorders, Implications for Treatment. Semin. Pediatr. Neurol. 2020, 35, 100834. [Google Scholar] [CrossRef] [PubMed]
  58. Makris, G.; Agorastos, A.; Chrousos, G.P.; Pervanidou, P. Stress System Activation in Children and Adolescents with Autism Spectrum Disorder. Front. Neurosci. 2022, 15, 756628. [Google Scholar] [CrossRef] [PubMed]
  59. Mawe, G.M.; Hoffman, J.M. Serotonin signalling in the gut—Functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473–486. [Google Scholar] [CrossRef]
  60. Foster, J.A.; Rinaman, L.; Cryan, J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress 2017, 7, 124–136. [Google Scholar] [CrossRef]
  61. Muller, C.L.; Anacker, A.M.J.; Veenstra-VanderWeele, J. The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 2016, 321, 24–41. [Google Scholar] [CrossRef]
  62. Terry, N.; Margolis, K.G. Serotonergic Mechanisms Regulating the GI Tract: Experimental Evidence and Therapeutic Relevance. In Gastrointestinal Pharmacology; Greenwood-Van Meerveld, B., Ed.; Handbook of Experimental Pharmacology; Springer International Publishing: Cham, Switzerland, 2016; pp. 319–342. [Google Scholar] [CrossRef]
  63. Dinan, T.G.; Stilling, R.M.; Stanton, C.; Cryan, J.F. Collective unconscious: How gut microbes shape human behavior. J. Psychiatr. Res. 2015, 63, 1–9. [Google Scholar] [CrossRef]
  64. Dezfouli, M.A.; Rashidi, S.K.; Yazdanfar, N.; Khalili, H.; Goudarzi, M.; Saadi, A.; Kiani Deh Kiani, A. The emerging roles of neuroactive components produced by gut microbiota. Mol. Biol. Rep. 2025, 52, 1. [Google Scholar] [CrossRef]
  65. Potter, K.; Gayle, E.J.; Deb, S. Effect of gut microbiome on serotonin metabolism: A personalized treatment approach. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 397, 2589–2602. [Google Scholar] [CrossRef]
  66. Hepsomali, P.; Groeger, J.A.; Nishihira, J.; Scholey, A. Effects of Oral Gamma-Aminobutyric Acid (GABA) Administration on Stress and Sleep in Humans: A Systematic Review. Front. Neurosci. 2020, 14, 923. [Google Scholar] [CrossRef]
  67. Gao, K.; Mu, C.; Farzi, A.; Zhu, W. Tryptophan Metabolism: A Link Between the Gut Microbiota and Brain. Adv. Nutr. 2020, 11, 709–723. [Google Scholar] [CrossRef] [PubMed]
  68. Jin, Y.; Kong, J. Transcutaneous Vagus Nerve Stimulation: A Promising Method for Treatment of Autism Spectrum Disorders. Front. Neurosci. 2017, 10, 609. [Google Scholar] [CrossRef] [PubMed]
  69. Van Hoorn, A.; Carpenter, T.; Oak, K.; Laugharne, R.; Ring, H.; Shankar, R. Neuromodulation of autism spectrum disorders using vagal nerve stimulation. J. Clin. Neurosci. 2019, 63, 8–12. [Google Scholar] [CrossRef] [PubMed]
  70. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed]
  71. Hoyles, L.; Pontifex, M.G.; Rodriguez-Ramiro, I.; Anis-Alavi, M.A.; Jelane, K.S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A.L.; Carding, S.R.; et al. Regulation of blood–brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235. [Google Scholar] [CrossRef]
  72. Parker, A.; Fonseca, S.; Carding, S.R. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2020, 11, 135–157. [Google Scholar] [CrossRef]
  73. Fiorentino, M.; Sapone, A.; Senger, S.; Camhi, S.S.; Kadzielski, S.M.; Buie, T.M.; Kelly, D.L.; Cascella, N.; Fasano, A. Blood–brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol. Autism 2016, 7, 49. [Google Scholar] [CrossRef]
  74. Eshraghi, R.S.; Davies, C.; Iyengar, R.; Perez, L.; Mittal, R.; Eshraghi, A.A. Gut-Induced Inflammation during Development May Compromise the Blood-Brain Barrier and Predispose to Autism Spectrum Disorder. J. Clin. Med. 2020, 10, 27. [Google Scholar] [CrossRef]
  75. Misiak, B.; Łoniewski, I.; Marlicz, W.; Frydecka, D.; Szulc, A.; Rudzki, L.; Samochowiec, J. The HPA axis dysregulation in severe mental illness: Can we shift the blame to gut microbiota? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 102, 109951. [Google Scholar] [CrossRef]
  76. Rusch, J.A.; Layden, B.T.; Dugas, L.R. Signalling cognition: The gut microbiota and hypothalamic-pituitary-adrenal axis. Front. Endocrinol. 2023, 14, 1130689. [Google Scholar] [CrossRef]
  77. Mamun, A.A.; Geng, P.; Wang, S.; Shao, C.; Xiao, J. IUPHAR review: Targeted therapies of signaling pathways based on the gut microbiome in autism spectrum disorders: Mechanistic and therapeutic applications. Pharmacol. Res. 2025, 211, 107559. [Google Scholar] [CrossRef] [PubMed]
  78. Kotowska, M.; Kołodziej, M.; Szajewska, H.; Łukasik, J. The impact of probiotics on core autism symptoms—A systematic review and meta-analysis of randomized clinical trials. Clin. Nutr. ESPEN 2024, 63, 893–902. [Google Scholar] [CrossRef] [PubMed]
  79. He, X.; Liu, W.; Tang, F.; Chen, X.; Song, G. Effects of Probiotics on Autism Spectrum Disorder in Children: A Systematic Review and Meta-Analysis of Clinical Trials. Nutrients 2023, 15, 1415. [Google Scholar] [CrossRef] [PubMed]
  80. Barba-Vila, O.; García-Mieres, H.; Ramos, B. Probiotics in autism spectrum disorders: A systematic review of clinical studies and future directions. Nutr. Rev. 2025, 83, 329–343. [Google Scholar] [CrossRef]
  81. Davidovics, Z.H.; Michail, S.; Nicholson, M.R.; Kociolek, L.K.; Pai, N.; Hansen, R.; Schwerd, T.; Maspons, A.; Shamir, R.; Szajewska, H.; et al. Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection and Other Conditions in Children: A Joint Position Paper From the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 130–143. [Google Scholar] [CrossRef]
  82. Lee, J.Y.; Kim, Y.; Kim, J.; Kim, J.K. Fecal Microbiota Transplantation: Indications, Methods, and Challenges. J. Microbiol. 2024, 62, 1057–1074. [Google Scholar] [CrossRef]
  83. Liu, C.; Du, M.-X.; Abuduaini, R.; Yu, H.-Y.; Li, D.-H.; Wang, Y.-J.; Zhou, N.; Jiang, M.-Z.; Niu, P.-X.; Han, S.-S.; et al. Enlightening the taxonomy darkness of human gut microbiomes with a cultured biobank. Microbiome 2021, 9, 119. [Google Scholar] [CrossRef]
  84. Kang, D.-W.; Adams, J.B.; Gregory, A.C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S.; et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study. Microbiome 2017, 5, 10. [Google Scholar] [CrossRef]
  85. Kang, D.-W.; Adams, J.B.; Coleman, D.M.; Pollard, E.L.; Maldonado, J.; McDonough-Means, S.; Caporaso, J.G.; Krajmalnik-Brown, R. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Sci. Rep. 2019, 9, 5821. [Google Scholar] [CrossRef]
  86. Wang, J.; Yang, R.; Zhong, H.; Liu, Y.-J. Fecal microbiota transplants in pediatric autism: Opportunities and challenges. World J. Pediatr. 2024, 20, 1201–1204. [Google Scholar] [CrossRef]
  87. Vaughan, C.A. Test Review: E. Schopler, M.E. Van Bourgondien, G.J. Wellman, & S. R. Love Childhood Autism Rating Scale (2nd ed.). Los Angeles, CA: Western Psychological Services, 2010. J. Psychoeduc. Assess. 2011, 29, 489–493. [Google Scholar] [CrossRef]
  88. Lord, C.; Rutter, M.; Goode, S.; Heemsbergen, J.; Jordan, H.; Mawhood, L.; Schopler, E. Autism Diagnostic Observation Schedule. 2016. Available online: https://psycnet.apa.org/doiLanding?doi=10.1037%2Ft54175-000 (accessed on 15 October 2025).
  89. Adams, J.B.; Audhya, T.; McDonough-Means, S.; Rubin, R.A.; Quig, D.; Geis, E.; Gehn, E.; Loresto, M.; Mitchell, J.; Atwood, S.; et al. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr. 2011, 11, 111. [Google Scholar] [CrossRef]
  90. Achenbach, T.M.; Rescorla, L.A. Manual for the ASEBA Preschool Forms & Profiles: An Integrated System of Multi-Informant Assessment; University of Vermont, Research Center for Children, Youth, & Families: Burlington, VT, USA, 2000. [Google Scholar]
  91. Li, N.; Chen, H.; Cheng, Y.; Xu, F.; Ruan, G.; Ying, S.; Tang, W.; Chen, L.; Chen, M.; Lv, L.; et al. Fecal Microbiota Transplantation Relieves Gastrointestinal and Autism Symptoms by Improving the Gut Microbiota in an Open-Label Study. Front. Cell Infect. Microbiol. 2021, 11, 759435. [Google Scholar] [CrossRef]
  92. Zhang, Y.; Zhang, J.; Pan, Z.; He, X. Effects of Washed Fecal Bacteria Transplantation in Sleep Quality, Stool Features and Autism Symptomatology: A Chinese Preliminary Observational Study. Neuropsychiatr. Dis. Treat. 2022, 18, 1165–1173. [Google Scholar] [CrossRef] [PubMed]
  93. Pan, Z.-Y.; Zhong, H.-J.; Huang, D.-N.; Wu, L.-H.; He, X.-X. Beneficial Effects of Repeated Washed Microbiota Transplantation in Children with Autism. Front. Pediatr. 2022, 10, 928785. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, N.-H.; Liu, H.-Q.; Zheng, J.-Y.; Zhu, M.-L.; Wu, L.-H.; Pan, H.-F.; He, X.-X. Fresh Washed Microbiota Transplantation Alters Gut Microbiota Metabolites to Ameliorate Sleeping Disorder Symptom of Autistic Children. J. Microbiol. 2023, 61, 741–753. [Google Scholar] [CrossRef]
  95. Chen, Q.; Wu, C.; Xu, J.; Ye, C.; Chen, X.; Tian, H.; Zong, N.; Zhang, S.; Li, L.; Gao, Y.; et al. Donor-recipient intermicrobial interactions impact transfer of subspecies and fecal microbiota transplantation outcome. Cell Host Microbe 2024, 32, 349–365.e4. [Google Scholar] [CrossRef]
  96. Li, Y.; Xiao, P.; Cao, R.; Le, J.; Xu, Q.; Xiao, F.; Ye, L.; Wang, X.; Wang, Y.; Zhang, T. Effects and microbiota changes following oral lyophilized fecal microbiota transplantation in children with autism spectrum disorder. Front. Pediatr. 2024, 12, 1369823. [Google Scholar] [CrossRef]
  97. Zhang, Y.J.; Bousvaros, A.; Docktor, M.; Kaplan, A.L.; Rufo, P.A.; Leier, M.; Weatherly, M.; Zimmerman, L.; Nguyen, L.T.T.; Barton, B.; et al. Higher Alpha Diversity and Lactobacillus Blooms Are Associated with Better Engraftment after Fecal Microbiota Transplant in Inflammatory Bowel Disease. Sci. Rep. 2024, 14, 18188. [Google Scholar] [CrossRef]
  98. Wang, L.; Yu, L.; Liu, Z.; Che, C.; Wang, Y.; Zhao, Y.; Zhu, M.; Yang, G.; Cao, A. FMT intervention decreases urine 5-HIAA levels: A randomized double-blind controlled study. Front. Med. 2024, 11, 1411089. [Google Scholar] [CrossRef]
  99. Wan, L.; Wang, H.; Liang, Y.; Zhang, X.; Yao, X.; Zhu, G.; Cai, J.; Liu, G.; Liu, X.; Niu, Q.; et al. Effect of oral faecal microbiota transplantation intervention for children with autism spectrum disorder: A randomised, double-blind, placebo-controlled trial. Clin. Transl. Med. 2024, 14, e70006. [Google Scholar] [CrossRef]
  100. Ramai, D.; Zakhia, K.; Fields, P.J.; Ofosu, A.; Patel, G.; Shahnazarian, V.; Lai, J.K.; Dhaliwal, A.; Reddy, M.; Chang, S. Fecal Microbiota Transplantation (FMT) with Colonoscopy Is Superior to Enema and Nasogastric Tube While Comparable to Capsule for the Treatment of Recurrent Clostridioides difficile Infection: A Systematic Review and Meta-Analysis. Dig. Dis. Sci. 2021, 66, 369–380. [Google Scholar] [CrossRef] [PubMed]
  101. McConachie, H.; Parr, J.R.; Glod, M.; Hanratty, J.; Livingstone, N.; Oono, I.P.; Robalino, S.; Baird, G.; Beresford, B.; Charman, T.; et al. Systematic Review of Tools to Measure Outcomes for Young Children with Autism Spectrum Disorder. Health Technol. Assess. 2015, 19, 1–506. [Google Scholar] [CrossRef] [PubMed]
  102. Thurman, A.J.; Nunnally, A.D.; Nguyen, V.; Berry-Kravis, E.; Sterling, A.; Edgin, J.; Hamilton, D.; Aschkenasy, J.; Abbeduto, L. Short-Term and Long-Term Stability of the Autism Diagnostic Observation Schedule (ADOS-2) Calibrated Comparison Scores (CCS) and Classification Scores in Youth with Down Syndrome or Fragile X Syndrome with Intellectual Disability. J. Autism Dev. Disord. 2024; published online. [Google Scholar] [CrossRef] [PubMed]
  103. Elias, R.; Lord, C. Diagnostic Stability in Individuals with Autism Spectrum Disorder: Insights from a Longitudinal Follow-up Study. Child. Psychol. Psychiatry 2022, 63, 973–983. [Google Scholar] [CrossRef]
  104. Siafis, S.; Çıray, O.; Schneider-Thoma, J.; Bighelli, I.; Krause, M.; Rodolico, A.; Ceraso, A.; Deste, G.; Huhn, M.; Fraguas, D.; et al. Placebo Response in Pharmacological and Dietary Supplement Trials of Autism Spectrum Disorder (ASD): Systematic Review and Meta-Regression Analysis. Mol. Autism 2020, 11, 66. [Google Scholar] [CrossRef]
  105. Curie, A.; Oberlander, T.F.; Jensen, K.B. Placebo Effects in Children with Autism Spectrum Disorder. Dev. Med. Child Neuro 2023, 65, 1316–1320. [Google Scholar] [CrossRef]
  106. Pergantis, P.; Bamicha, V.; Doulou, A.; Christou, A.I.; Bardis, N.; Skianis, C.; Drigas, A. Assistive and Emerging Technologies to Detect and Reduce Neurophysiological Stress and Anxiety in Children and Adolescents with Autism and Sensory Processing Disorders: A Systematic Review. Technologies 2025, 13, 144. [Google Scholar] [CrossRef]
  107. Mysore, A.; Kaku, S.M. Predictors of Outcome in Autism Spectrum Disorders: A Perspective for Clinicians and Therapists. Indian J. Psychol. Med. 2025, 47, 290–294. [Google Scholar] [CrossRef]
  108. Posar, A. Long-Term Outcome of Autism Spectrum Disorder. Turk. Pediatr. Ars. 2019, 54, 207–212. [Google Scholar] [CrossRef]
Figure 1. The CONSORT-style flow diagram shows participant flow through study enrollment, clinical allocation, and in-person follow-up assessments. Parents’ decision to decline study participation did not affect clinical care; however, no study-related data were collected from these patients.
Figure 1. The CONSORT-style flow diagram shows participant flow through study enrollment, clinical allocation, and in-person follow-up assessments. Parents’ decision to decline study participation did not affect clinical care; however, no study-related data were collected from these patients.
Medicina 62 00065 g001
Figure 2. Between-group comparison between the ASDi and control (ASDc) groups at baseline, 8 weeks, and 6 months. (AG) Panels depict scores in ADOS, CARS, CBCL subscales (Internalizing, Externalizing, and Total Problems), GSRS, and PGI-R, respectively. The ASDi group demonstrated significantly greater improvement in CARS, PGI-R, GSRS, CBCL Internalizing, and CBCL Total scores at 8 weeks compared with controls. Improvement was sustained at 6 months in CARS, CBCL Internalizing, and PGI-R scores. Data are presented as mean ± SD. * p ≤ 0.05, *** p ≤ 0.001.
Figure 2. Between-group comparison between the ASDi and control (ASDc) groups at baseline, 8 weeks, and 6 months. (AG) Panels depict scores in ADOS, CARS, CBCL subscales (Internalizing, Externalizing, and Total Problems), GSRS, and PGI-R, respectively. The ASDi group demonstrated significantly greater improvement in CARS, PGI-R, GSRS, CBCL Internalizing, and CBCL Total scores at 8 weeks compared with controls. Improvement was sustained at 6 months in CARS, CBCL Internalizing, and PGI-R scores. Data are presented as mean ± SD. * p ≤ 0.05, *** p ≤ 0.001.
Medicina 62 00065 g002
Figure 3. ASDi group symptom scores after FMT. Panels (AG) show trajectories of ADOS, CARS, GSRS, PGI-R, and CBCL subscales (Internalizing, Externalizing, and Total Problems) across baseline, 2 weeks, 8 weeks, 6 months, and 18 months. GSRS and PGI-R have additional timepoints at 1, 3, and 4 weeks. Significant reductions were observed in CARS, GSRS, and CBCL Internalizing scores, with sustained improvement in PGI-R ratings throughout follow-up (Wilcoxon signed-rank test p < 0.01). Data are presented as median (interquartile range).
Figure 3. ASDi group symptom scores after FMT. Panels (AG) show trajectories of ADOS, CARS, GSRS, PGI-R, and CBCL subscales (Internalizing, Externalizing, and Total Problems) across baseline, 2 weeks, 8 weeks, 6 months, and 18 months. GSRS and PGI-R have additional timepoints at 1, 3, and 4 weeks. Significant reductions were observed in CARS, GSRS, and CBCL Internalizing scores, with sustained improvement in PGI-R ratings throughout follow-up (Wilcoxon signed-rank test p < 0.01). Data are presented as median (interquartile range).
Medicina 62 00065 g003
Table 1. The comparison of baseline characteristics of participants between study groups.
Table 1. The comparison of baseline characteristics of participants between study groups.
MeasureControl Group
Mean (SD)
Intervention Group
Mean (SD)
Median [IQR]
Control vs. Intervention
p
Age (years)4.40 (0.83)5.13 (1.36)4.0 [4.0–5.0] vs. 5.0 [4.0–6.0]0.116
ADOS6.87 (2.45)7.93 (1.98)6.0 [5.0–10.0] vs. 8.0 [6.0–10.0]0.250
CARS37.13 (4.50)38.83 (3.34)38.0 [34.0–41.0] vs. 39.0 [37.0–41.0]0.367
GSRS21.00 (5.59)29.73 (8.97)19.0 [17.0–23.0] vs. 27.0 [23.0–38.0]0.002
CBCL Internalizing21.73 (5.68)23.60 (5.71)21.0 [16.0–27.0] vs. 24.0 [19.0–28.0]0.285
CBCL Externalizing13.20 (7.99)21.07 (10.89)10.0 [7.0–20.0] vs. 23.0 [11.0–27.0]0.026
CBCL Total Problems37.40 (12.55)49.07 (12.35)31.0 [27.0–50.0] vs. 49.0 [40.0–58.0]0.015
Data are presented as mean (SD) and median [IQR]. p-values were obtained using the Mann–Whitney U test.
Table 2. Between-group comparison of change scores (Mann–Whitney U) at 8-week and 6-month time points.
Table 2. Between-group comparison of change scores (Mann–Whitney U) at 8-week and 6-month time points.
MeasureTime PointMedian (ASDc)IQR (ASDc)Median (ASDi)IQR (ASDi)p (MWU)
ADOS8 weeks0.0[−1.00–1.00]−1.0[−2.00–0.00]0.061
ADOS6 months−0.5[−1.00–0.00]−1.0[−3.00–0.00]0.367
CARS8 weeks0.0[−1.00–1.00]−4.0[−5.00–3.00]<0.001
CARS6 months−1.0[−2.00–0.00]−5.0[−7.00–3.00]0.045
PGI-R8 weeks1.0[1.00–2.00]3.0[3.00–4.00]<0.001
PGI-R6 months2.0[1.00–3.00]3.0[3.00–4.00]0.007
CBCL Internalizing8 weeks0.0[−2.00–2.00]−4.0[−5.00–2.00]0.001
CBCL Internalizing6 months−3.0[−4.00–2.00]−4.0[−6.00–1.00]0.250
Data are presented as median [interquartile range]. Between-group comparisons were performed using the Mann–Whitney U test. Bold values indicate statistically significant results (p < 0.05).
Table 3. Between-group comparison (Quade Rank ANCOVA, Baseline-Adjusted Outcomes) of change scores at 8-week and 6-month time points.
Table 3. Between-group comparison (Quade Rank ANCOVA, Baseline-Adjusted Outcomes) of change scores at 8-week and 6-month time points.
MeasureTime PointAdjusted pPartial Eta2
GSRS8 weeks0.0370.152
GSRS6 months0.2400.051
CBCL Externalizing8 weeks0.3090.038
CBCL Externalizing6 months0.9470.000
CBCL Total8 weeks0.0010.323
CBCL Total6 months0.4020.026
Bold values indicate statistically significant results (p < 0.05).
Table 4. Over time ASDi group symptoms (Friedman test).
Table 4. Over time ASDi group symptoms (Friedman test).
MeasureTotal Time PointsFriedman χ2dfp-Value
ADOS517.64740.0014
CARS533.2244<0.0001
CBCL Internalizing537.824<0.0001
CBCL Externalizing525.1924<0.0001
CBCL Total Problems534.7564<0.0001
GSRS728.8586<0.0001
PGI-R722.48460.001
Friedman test used to assess within-group changes across time points in the ASDi group (Baseline, 2 weeks, 8 weeks, 6 months, and 18 months). χ2—Chi-square statistic; df—degrees of freedom. Bold values indicate statistically significant results (p < 0.05).
Table 5. ASDi group Wilcoxon hierarchical post hoc tests.
Table 5. ASDi group Wilcoxon hierarchical post hoc tests.
OutcomeComparisonZp-ValueEffect Size (r)
ADOSBaseline vs. 2 w−2.4490.0310.632
ADOSBaseline vs. 8 w−2.6460.0160.683
ADOSBaseline vs. 6 m−20.1250.516
ADOSBaseline vs. 18 m−1.7320.250.447
CARSBaseline vs. 2 w−3.192<0.0010.824
CARSBaseline vs. 8 w−3.208<0.0010.828
CARSBaseline vs. 6 m−2.5580.0080.66
CARSBaseline vs. 18 m−2.4530.0120.633
CBCL InternalizingBaseline vs. 2 w−3.319<0.0010.857
CBCL InternalizingBaseline vs. 8 w−3.078<0.0010.795
CBCL InternalizingBaseline vs. 6 m−2.5850.0080.667
CBCL InternalizingBaseline vs. 18 m−2.4890.0160.643
CBCL ExternalizingBaseline vs. 2 w−3.0520.0010.788
CBCL ExternalizingBaseline vs. 8 w−3.090.0010.798
CBCL ExternalizingBaseline vs. 6 m−1.830.0930.473
CBCL ExternalizingBaseline vs. 18 m−1.6150.1430.417
CBCL TotalBaseline vs. 2 w−3.301<0.0010.852
CBCL TotalBaseline vs. 8 w−3.31<0.0010.855
CBCL TotalBaseline vs. 6 m−2.9120.0030.752
CBCL TotalBaseline vs. 18 m−2.5030.0110.646
GSRSBaseline vs. 2 w−3.413<0.0010.881
GSRSBaseline vs. 8 w−3.306<0.0010.854
GSRSBaseline vs. 6 m−2.8820.0030.744
GSRSBaseline vs. 18 m−2.6680.0070.689
PGI-RBaseline vs. 2 w−3.1050.0010.802
PGI-RBaseline vs. 8 w−4.023<0.0011.039
PGI-RBaseline vs. 6 m−3.623<0.0010.935
PGI-RBaseline vs. 18 m−2.6770.0040.691
Post hoc Wilcoxon signed-rank tests were conducted hierarchically within the ASDi group to compare baseline scores with follow-up time points (2 weeks, 8 weeks, 6 months, 18 months). Significant results are shown in bold. All post hoc comparisons are reported for transparency. In accordance with the predefined hierarchical gatekeeping strategy, only comparisons occurring prior to the first non-significant result were considered confirmatory; subsequent comparisons are exploratory and reported descriptively. Bold values indicate statistically significant results (p < 0.05).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Varnas, D.; Kunevičius, A.; Burokas, A.; Urbonas, V. Fecal Microbiota Transplantation for Autism Spectrum Disorder in Children: Results from a Prospective Open-Label Controlled Observational Study. Medicina 2026, 62, 65. https://doi.org/10.3390/medicina62010065

AMA Style

Varnas D, Kunevičius A, Burokas A, Urbonas V. Fecal Microbiota Transplantation for Autism Spectrum Disorder in Children: Results from a Prospective Open-Label Controlled Observational Study. Medicina. 2026; 62(1):65. https://doi.org/10.3390/medicina62010065

Chicago/Turabian Style

Varnas, Dominykas, Arnas Kunevičius, Aurelijus Burokas, and Vaidotas Urbonas. 2026. "Fecal Microbiota Transplantation for Autism Spectrum Disorder in Children: Results from a Prospective Open-Label Controlled Observational Study" Medicina 62, no. 1: 65. https://doi.org/10.3390/medicina62010065

APA Style

Varnas, D., Kunevičius, A., Burokas, A., & Urbonas, V. (2026). Fecal Microbiota Transplantation for Autism Spectrum Disorder in Children: Results from a Prospective Open-Label Controlled Observational Study. Medicina, 62(1), 65. https://doi.org/10.3390/medicina62010065

Article Metrics

Back to TopTop