Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota

Dae-Wook Kang, James B Adams, Devon M Coleman, Elena L Pollard, Juan Maldonado, Sharon McDonough-Means, J Gregory Caporaso, Rosa Krajmalnik-Brown, Dae-Wook Kang, James B Adams, Devon M Coleman, Elena L Pollard, Juan Maldonado, Sharon McDonough-Means, J Gregory Caporaso, Rosa Krajmalnik-Brown

Abstract

Many studies have reported abnormal gut microbiota in individuals with Autism Spectrum Disorders (ASD), suggesting a link between gut microbiome and autism-like behaviors. Modifying the gut microbiome is a potential route to improve gastrointestinal (GI) and behavioral symptoms in children with ASD, and fecal microbiota transplant could transform the dysbiotic gut microbiome toward a healthy one by delivering a large number of commensal microbes from a healthy donor. We previously performed an open-label trial of Microbiota Transfer Therapy (MTT) that combined antibiotics, a bowel cleanse, a stomach-acid suppressant, and fecal microbiota transplant, and observed significant improvements in GI symptoms, autism-related symptoms, and gut microbiota. Here, we report on a follow-up with the same 18 participants two years after treatment was completed. Notably, most improvements in GI symptoms were maintained, and autism-related symptoms improved even more after the end of treatment. Important changes in gut microbiota at the end of treatment remained at follow-up, including significant increases in bacterial diversity and relative abundances of Bifidobacteria and Prevotella. Our observations demonstrate the long-term safety and efficacy of MTT as a potential therapy to treat children with ASD who have GI problems, and warrant a double-blind, placebo-controlled trial in the future.

Conflict of interest statement

J.B.A., D.-W.K. and R.K.B. have pending/approved patents related to the use of FMT and/or probiotics for various conditions including autism (Application number: 14/403,425 (approved and active); 15/290,798 (pending)). J.B.A., R.K.B. and D.-W.K. have received research funding from Crestovo/Finch Therapeutics for FMT research. J.B.A., R.K.B. and J.G.C. have received consulting fees from Crestovo. The other authors declare no competing interests.

Figures

Figure 1
Figure 1
Changes in GI- and ASD-related symptoms of 18 children with ASD at two-year follow-up after treatment stopped. Asterisks (at the top of the box plot) indicate whether individuals (at each time point) have significantly changed since pre-treatment (Week 0 of original Phase 1 trial). Based on two-tailed Wilcoxon signed-rank test, ns indicates not significant, single asterisk indicates p < 0.05, double asterisks indicate p < 0.01, triple asterisks indicate p < 0.001. See also Supplementary Figs S1–S3.
Figure 2
Figure 2
CARS and SRS diagnostic category for ASD at baseline, 8 weeks after treatment, and two-year follow-up after treatment stopped. (a) For CARS, Minimal-to-No Symptoms (15–29.5 for ages less than 13; 15–27.5 for ages 13 or older), Mild-to-Moderate Symptoms (30–36.5 for ages less than 13; 28–34.5 for ages 13 or order), and Sever Symptoms (37 and higher for ages less than 13; 35 and higher for ages 13 or order). (b) For SRS, Normal (0–53), Mild or High Functioning autism (54–86), Clinical diagnosis of autistic disorder, Asperger’s disorder, or more severe cases of Pervasive developmental disorder not otherwise specified (PDD-NOS) (>87). (cg) Strong and significant correlations between improvements in GI symptoms (GSRS) and behavior symptoms based on % changes in 2 years.
Figure 3
Figure 3
Stool microbiota assessments at two-year follow-up after treatment stopped. (a) Faith’s phylogenetic diversity (PD) in the microbiota of 18 children with ASD as measured from stool samples. Orange lines indicate median PD of the donor samples (dashed line represents initial donor samples (n = 5), and dotted line represents maintenance dose samples (n = 2)), and green line indicates median PD of 20 neurotypical controls at week 0. ns indicates not significant, single asterisk indicates p < 0.05, double asterisks indicate p < 0.01, triple asterisks indicate p < 0.001 (two-tailed Wilcoxon signed-rank test comparing weeks 3, 10, and 18 and two-year to week 0 values). (b) Unweighted UniFrac distances between ASD gut microbiota and most relevant donor sample (initial donor sample at weeks 0 and 3, most recent maintenance dose sample at weeks 10 and 18, and 2 years). Green line indicates the median interpersonal variation between neurotypical controls and illustrates that prior to treatment the difference in gut microbiota composition between MTT recipients and donors was on the order of normal interpersonal variation. Statistics are the same as those used in (a). See also Supplementary Figs S5 and S6.
Figure 4
Figure 4
Changes in relative abundances of Bifidobacterium, Prevotella, and Desulfovibrio. ns indicates not significant, single asterisk indicates p < 0.05 and double asterisks indicate p < 0.01 (two-tailed Wilcoxon signed-rank test comparing weeks 3, 10, and 8 and two-year to week 0 values). Orange lines indicate median of the donor samples (dashed line represents initial donor samples, and dotted line represents maintenance dose samples), and green line indicates median of 20 neurotypical controls at week 0.

References

    1. Rogers GB, et al. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Molecular Psychiatry. 2016;21:738–748. doi: 10.1038/mp.2016.50.
    1. Sharon G, Sampson TR, Geschwind DH, Mazmanian SK. The central nervous system and the gut microbiome. Cell. 2016;167:915–932. doi: 10.1016/j.cell.2016.10.027.
    1. McElhanon, B. O., McCracken, C., Karpen, S. & Sharp, W. G. Gastrointestinal symptoms in autism spectrum disorder: a meta-analysis. Pediatrics133, 10.1542/peds.2013-3995 (2014).
    1. Braak H, Rub U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. Journal of Neural Transmission. 2003;110:517–536. doi: 10.1007/s00702-002-0808-2.
    1. McCarthy M, AddingtonHall J, Altmann D. The experience of dying with dementia: A retrospective study. International Journal of Geriatric Psychiatry. 1997;12:404–409. doi: 10.1002/(SICI)1099-1166(199703)12:3<404::AID-GPS529>;2-2.
    1. Hallmayer J, et al. Genetic heritability and shared environmental factors among twin pairs with autism. Archives of General Psychiatry. 2011;68:1095–1102. doi: 10.1001/archgenpsychiatry.2011.76.
    1. Finegold SM, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16:444–453. doi: 10.1016/j.anaerobe.2010.06.008.
    1. De Angelis M, et al. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. Plos One. 2013;8:18. doi: 10.1371/journal.pone.0076993.
    1. Williams BL, Hornig M, Parekh T, Lipkin WI. Application of novel PCR-based methods for detection, quantification, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances. MBio. 2012;3:e00261–11. doi: 10.1128/mBio.00261-11.
    1. Gondalia SV, et al. Molecular characterisation of gastrointestinal microbiota of children with autism (with and without gastrointestinal dysfunction) and their neurotypical siblings. Autism Research. 2012;5:419–427. doi: 10.1002/aur.1253.
    1. Son JS, et al. Comparison of fecal microbiota in children with autism spectrum disorders and neurotypical siblings in the Simons simplex sollection. Plos One. 2015;10:e0137725. doi: 10.1371/journal.pone.0137725.
    1. Hsiao EY, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopment disorders. Cell. 2013;155:1451–1463. doi: 10.1016/j.cell.2013.11.024.
    1. Kim S, et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature. 2017;549:528–532. doi: 10.1038/nature23910.
    1. Buffington SA, et al. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell. 2016;165:1762–1775. doi: 10.1016/j.cell.2016.06.001.
    1. Lacivita E, Perrone R, Margari L, Leopoldo M. Targets for drug therapy for autism spectrum disorder: challenges and future directions. Journal of Medicinal Chemistry. 2017;60:9114–9141. doi: 10.1021/acs.jmedchem.7b00965.
    1. Bagdasarian N, Rao K, Malani PN. Diagnosis and treatment of Clostridium difficile in adults a systematic review. Jama-Journal of the American Medical Association. 2015;313:398–408. doi: 10.1001/jama.2014.17103.
    1. Moayyedi P, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015;149:102–109. doi: 10.1053/j.gastro.2015.04.001.
    1. Xu MQ, et al. Fecal microbiota transplantation broadening its application beyond intestinal disorders. World Journal of Gastroenterology. 2015;21:102–111. doi: 10.3748/wjg.v21.i1.102.
    1. Kang D-W, et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5:10. doi: 10.1186/s40168-016-0225-7.
    1. Belsito KM, Law PA, Kirk KS, Landa RJ, Zimmerman AW. Lamotrigine therapy for autistic disorder: A randomized, double-blind, placebo-controlled trial. Journal of Autism and Developmental Disorders. 2001;31:175–181. doi: 10.1023/a:1010799115457.
    1. Nah YH, Young RL, Brewer N. Using the Autism Detection in Early Childhood (ADEC) and Childhood Autism Rating Scales (CARS) to predict long term outcomes in children with autism spectrum disorders. Journal of Autism and Developmental Disorders. 2014;44:2301–2310. doi: 10.1007/s10803-014-2102-1.
    1. Gattino GS, Riesgo RD, Longo D, Leite JCL, Faccini LS. Effects of relational music therapy on communication of children with autism: a randomized controlled study. Nordic Journal of Music Therapy. 2011;20:142–154. doi: 10.1080/08098131.2011.566933.
    1. Saad K, et al. Randomized controlled trial of vitamin D supplementation in children with autism spectrum disorder. Journal of Child Psychology and Psychiatry. 2018;59:20–29. doi: 10.1111/jcpp.12652.
    1. Adams JB, Johansen LJ, Powell LD, Quig D, Rubin RA. Gastrointestinal flora and gastrointestinal status in children with autism-comparisons to typical children and correlation with autism severity. Bmc Gastroenterology. 2011;11:22. doi: 10.1186/1471-230x-11-22.
    1. Horvath K, Perman JA. Autistic disorder and gastrointestinal disease. Current Opinion in Pediatrics. 2002;14:583–587. doi: 10.1097/01.mop.0000030221.71203.46.
    1. Kang DW, et al. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. Plos One. 2013;8:e68322. doi: 10.1371/journal.pone.0068322.
    1. Ilhan ZE, et al. Distinctive microbiomes and metabolites linked with weight loss after gastric bypass, but not gastric banding. Isme Journal. 2017;11:2047–2058. doi: 10.1038/ismej.2017.71.
    1. Ahn J, et al. Human gut microbiome and risk for colorectal cancer. Jnci-Journal of the National Cancer Institute. 2013;105:1907–1911. doi: 10.1093/jnci/djt300.
    1. Ott SJ, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53:685–693. doi: 10.1136/gut.2003.025403.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Applied and Environmental Microbiology. 2005;71:8228–8235. doi: 10.1128/aem.71.12.8228-8235.2005.
    1. Kang DW, et al. Differences in fecal microbial metabolites and microbiota of children with autism spectrum disorders. Anaerobe. 2017;49:121–131. doi: 10.1016/j.anaerobe.2017.12.007.
    1. Qiao YA, et al. Alterations of oral microbiota distinguish children with autism spectrum disorders from healthy controls. Scientific Reports. 2018;8:12. doi: 10.1038/s41598-018-19982-y.
    1. Esquivel-Elizondo S, Ilhan ZE, Garcia-Pena EI, Krajmalnik-Brown R. Insights into butyrate production in a controlled fermentation system via gene predictions. mSystems. 2017;2:13. doi: 10.1128/mSystems.00051-17.
    1. Donohoe DR, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metabolism. 2011;13:517–526. doi: 10.1016/j.cmet.2011.02.018.
    1. Arumugam M, et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174–180. doi: 10.1038/nature09944.
    1. Derrien M, et al. Mucin-bacterial interactions in the human oral cavity and digestive tract. Gut Microbes. 2010;1:254–268. doi: 10.4161/gmic.1.4.12778.
    1. Sandler RH, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. Journal of Child Neurology. 2000;15:429–435. doi: 10.1177/088307380001500701.
    1. Singh K, et al. Sulforaphane treatment of autism spectrum disorder (ASD) Proceedings of the National Academy of Sciences of the United States of America. 2014;111:15550–15555. doi: 10.1073/pnas.1416940111.
    1. Gotham K, Pickles A, Lord C. Trajectories of autism severity in children using standardized ADOS scores. Pediatrics. 2012;130:E1278–E1284. doi: 10.1542/peds.2011-3668.
    1. Jain KS, et al. Recent advances in proton pump inhibitors and management of acid-peptic disorders. Bioorganic & Medicinal Chemistry. 2007;15:1181–1205. doi: 10.1016/j.bmc.2006.07.068.
    1. Hamilton MJ, Weingarden AR, Sadowsky MJ, Khoruts A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am J Gastroenterol. 2012;107:761–767. doi: 10.1038/ajg.2011.482.
    1. Revicki DA, Wood M, Wiklund I, Crawley J. Reliability and validity of the gastrointestinal symptom rating scale in patients with gastroesophageal reflux disease. Quality of Life Research. 1997;7:75–83. doi: 10.1023/A:1008841022998.
    1. Adams, J. B. et al. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr11, 10.1186/1471-2431-11-111 (2011).
    1. Caporaso JG, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. Isme Journal. 2012;6:1621–1624. doi: 10.1038/ismej.2012.8.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nature Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nature Methods. 2016;13:581–583. doi: 10.1038/nmeth.3869.
    1. Bokulich NA, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nature Methods. 2013;10:57–59. doi: 10.1038/nmeth.2276.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open source tool for metagenomics. Peerj. 2016;4:e2584. doi: 10.7717/peerj.2584.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Research. 2013;41:D590–D596. doi: 10.1093/nar/gks1219.
    1. Katoh K, Standley DM. MAFFT Multiple Sequence Alignment Software Version 7: Improvements in Performance and Usability. Molecular Biology and Evolution. 2013;30:772–780. doi: 10.1093/molbev/mst010.
    1. Lane, D. Nucleic Acid Techniques in Bacterial Systematics. 115–175 (John Wiley and Sons, 1991).
    1. Price MN, Dehal PS, Arkin AP. FastTree 2-approximately maximum-likelihood trees for large alignments. Plos One. 2010;5:e9490. doi: 10.1371/journal.pone.0009490.
    1. Bokulich NA, et al. Optimizing taxonomic classification of marker-gene amplicon sequences with QIIME 2′s q2-feature-classifier plugin. Microbiome. 2018;6:90. doi: 10.1186/s40168-018-0470-z.
    1. Schopler, E., Van Bourgondien, M. E., Wellman, G. J. & Love, S. R. Childhood autism rating scale, (CARS2). (WPS Torrance, CA, 2010).
    1. Constantino, J. N. Social Responsiveness Scale (SRS). (Western Psychological Services, 2005).

Source: PubMed

3
구독하다