Fecal microbiota transplantation (FMT) is highly effective against recurrent Clostridioides difficile infection and is considered a promising treatment for other microbiome-related disorders, but a comprehensive understanding of microbial engraftment dynamics is lacking, which prevents informed applications of this therapeutic approach. Here, we performed an integrated shotgun metagenomic systematic meta-analysis of new and publicly available stool microbiomes collected from 226 triads of donors, pre-FMT recipients and post-FMT recipients across eight different disease types. By leveraging improved metagenomic strain-profiling to infer strain sharing, we found that recipients with higher donor strain engraftment were more likely to experience clinical success after FMT (P = 0.017) when evaluated across studies. Considering all cohorts, increased engraftment was noted in individuals receiving FMT from multiple routes (for example, both via capsules and colonoscopy during the same treatment) as well as in antibiotic-treated recipients with infectious diseases compared with antibiotic-naive patients with noncommunicable diseases. Bacteroidetes and Actinobacteria species (including Bifidobacteria) displayed higher engraftment than Firmicutes except for six under-characterized Firmicutes species. Cross-dataset machine learning predicted the presence or absence of species in the post-FMT recipient at 0.77 average AUROC in leave-one-dataset-out evaluation, and highlighted the relevance of microbial abundance, prevalence and taxonomy to infer post-FMT species presence. By exploring the dynamics of microbiome engraftment after FMT and their association with clinical variables, our study uncovered species-specific engraftment patterns and presented machine learning models able to predict donors that might optimize post-FMT specific microbiome characteristics for disease-targeted FMT protocols.Coupling microbial metagenomics with machine learning enables prediction of donor strain engraftment after fecal microbiota transplantation (FMT) for a range of diseases, and may help tailor design of FMT to optimize microbial engraftment and achieve clinical outcomes.

Ianiro, G., Punčochář, M., Karcher, N., Porcari, S., Armanini, F., Asnicar, F., Beghini, F., Blanco-Míguez, A., Cumbo, F., Manghi, P., Pinto, F., Masucci, L., Quaranta, G., De Giorgi, S., Sciumè, G. D., Bibbò, S., Del Chierico, F., Putignani, L., Sanguinetti, M., Gasbarrini, A., Valles-Colomer, M., Cammarota, G., Segata, N., Variability of strain engraftment and predictability of microbiome composition after fecal microbiota transplantation across different diseases, <<NATURE MEDICINE>>, 2022; 28 (9): 1913-1923. [doi:10.1038/s41591-022-01964-3] [https://hdl.handle.net/10807/231511]

Variability of strain engraftment and predictability of microbiome composition after fecal microbiota transplantation across different diseases

Ianiro, Gianluca
Primo
;
Porcari, Serena;Masucci, Luca;Quaranta, Gianluca;Putignani, Lorenza;Sanguinetti, Maurizio;Gasbarrini, Antonio;Cammarota, Giovanni;
2022

Abstract

Fecal microbiota transplantation (FMT) is highly effective against recurrent Clostridioides difficile infection and is considered a promising treatment for other microbiome-related disorders, but a comprehensive understanding of microbial engraftment dynamics is lacking, which prevents informed applications of this therapeutic approach. Here, we performed an integrated shotgun metagenomic systematic meta-analysis of new and publicly available stool microbiomes collected from 226 triads of donors, pre-FMT recipients and post-FMT recipients across eight different disease types. By leveraging improved metagenomic strain-profiling to infer strain sharing, we found that recipients with higher donor strain engraftment were more likely to experience clinical success after FMT (P = 0.017) when evaluated across studies. Considering all cohorts, increased engraftment was noted in individuals receiving FMT from multiple routes (for example, both via capsules and colonoscopy during the same treatment) as well as in antibiotic-treated recipients with infectious diseases compared with antibiotic-naive patients with noncommunicable diseases. Bacteroidetes and Actinobacteria species (including Bifidobacteria) displayed higher engraftment than Firmicutes except for six under-characterized Firmicutes species. Cross-dataset machine learning predicted the presence or absence of species in the post-FMT recipient at 0.77 average AUROC in leave-one-dataset-out evaluation, and highlighted the relevance of microbial abundance, prevalence and taxonomy to infer post-FMT species presence. By exploring the dynamics of microbiome engraftment after FMT and their association with clinical variables, our study uncovered species-specific engraftment patterns and presented machine learning models able to predict donors that might optimize post-FMT specific microbiome characteristics for disease-targeted FMT protocols.Coupling microbial metagenomics with machine learning enables prediction of donor strain engraftment after fecal microbiota transplantation (FMT) for a range of diseases, and may help tailor design of FMT to optimize microbial engraftment and achieve clinical outcomes.
2022
Inglese
Ianiro, G., Punčochář, M., Karcher, N., Porcari, S., Armanini, F., Asnicar, F., Beghini, F., Blanco-Míguez, A., Cumbo, F., Manghi, P., Pinto, F., Masucci, L., Quaranta, G., De Giorgi, S., Sciumè, G. D., Bibbò, S., Del Chierico, F., Putignani, L., Sanguinetti, M., Gasbarrini, A., Valles-Colomer, M., Cammarota, G., Segata, N., Variability of strain engraftment and predictability of microbiome composition after fecal microbiota transplantation across different diseases, <<NATURE MEDICINE>>, 2022; 28 (9): 1913-1923. [doi:10.1038/s41591-022-01964-3] [https://hdl.handle.net/10807/231511]
File in questo prodotto:
File Dimensione Formato  
Ianiro et al - Nat Med 2022.pdf

accesso aperto

Licenza: Creative commons
Dimensione 15.54 MB
Formato Adobe PDF
15.54 MB Adobe PDF Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/10807/231511
Citazioni
  • ???jsp.display-item.citation.pmc??? 56
  • Scopus 106
  • ???jsp.display-item.citation.isi??? 98
social impact