Long-Term Bacterial and Fungal Dynamics following Oral Lyophilized Fecal Microbiota Transplantation in Clostridioides difficile Infection

Craig Haifer, Sudarshan Paramsothy, Thomas J Borody, Annabel Clancy, Rupert W Leong, Nadeem O Kaakoush, Craig Haifer, Sudarshan Paramsothy, Thomas J Borody, Annabel Clancy, Rupert W Leong, Nadeem O Kaakoush

Abstract

Oral lyophilized fecal microbiota transplantation (FMT) is effective in recurrent Clostridioides difficile infection (CDI); however, limited data exist on its efficacy in primary CDI and long-term microbial engraftment. Patients with primary or recurrent CDI were prospectively enrolled to receive oral FMT. Changes in the bacterial and fungal communities were characterized prior to and up to 6 months following treatment. A total of 37 patients with CDI (15 primary, 22 recurrent) were treated with 6 capsules each containing 0.35-g lyophilized stool extract. A total of 33 patients (89%) had sustained CDI cure, of whom 3 required a second course. There were no safety signals identified. FMT significantly increased bacterial diversity and shifted composition toward donor profiles in responders but not in nonresponders, with robust donor contribution observed to 6 months following FMT (P < 0.001). Responders showed consistent decreases in Enterobacteriaceae and increases in Faecalibacterium sp. to levels seen in donors. Mycobiome profiling revealed an association with FMT failure and increases in one Penicillium taxon, as well as coexclusion relationships between Candida sp. and bacterial taxa enriched in both donors and responders. Primary CDI was associated with more robust changes in the bacterial community than those with recurrent disease. Oral FMT leads to durable microbial engraftment in patients with primary and recurrent CDI, with several microbial taxa being associated with therapy outcome. Novel coexclusion relationships between bacterial and fungal species support the clinical relevance of transkingdom dynamics.IMPORTANCE Clostridioides difficile infection (CDI) is a substantial health concern worldwide, complicated by patterns of increasing antibiotic resistance that may impact primary treatment. Orally administered fecal microbiota transplantation (FMT) is efficacious in the management of recurrent CDI, with specific bacterial species known to influence clinical outcomes. To date, little is known about the efficacy of FMT in primary CDI and the impact of the mycobiome on therapeutic outcomes. We performed matched bacterial and fungal sequencing on longitudinal samples from a cohort of patients treated with oral FMT for primary and recurrent CDI. We validated many bacterial signatures following oral therapy, confirmed engraftment of donor microbiome out to 6 months following therapy, and demonstrated coexclusion relationships between Candida albicans and two bacterial species in the gut microbiota, which has potential significance beyond CDI, including in the control of gut colonization by this fungal species.

Keywords: Clostridioides difficile infection; fecal microbiota transplantation; microbiome; mycobiome.

Copyright © 2021 Haifer et al.

Figures

FIG 1
FIG 1
Changes to the bacterial communities. Both primary and recurrent CDI are included. Two patients did not provide baseline samples. (A) Shannon’s diversity (H´) indices in donors and responders to FMT. Significance was tested using ANOVA with Tukey’s multiple-comparison test, and P-W0 was found to be statistically significantly different from all other groups. No other comparisons were significant. (B) Shannon’s diversity (H´) index in donors and nonresponders to FMT. Patient samples at recurrence were labeled in black and with patient number. P34 had persistent disease. Significance was tested using ANOVA with Tukey’s multiple-comparison test, and P-W4 was found to be significantly different from donors B and C. No other comparisons were significant. (C) Principal-coordinate analysis of responders to FMT and donors. Bray-Curtis resemblance matrix was generated from square-root-transformed relative abundances of bacterial OTUs. All patient subgroups (P-) were significantly different from the donors (DON) when tested using pairwise PERMANOVA (P < 0.005 for all). P-W0 was significantly different from all other patient subgroups (P < 0.006 for all). No other comparisons were significant. ANOSIM confirmed the pairwise PERMANOVA results. (D) Principal-coordinate analysis of nonresponders to FMT and donors. Bray-Curtis resemblance matrix was generated from square-root-transformed relative abundances of bacterial OTUs. Dotted lines indicate samples corresponding to the same patient unless otherwise indicated. All patient subgroups (P-) were significantly different from the donors (DON) when tested using pairwise PERMANOVA (P < 0.004 for all). No other comparisons were significant. ANOSIM confirmed the pairwise PERMANOVA results. (E) Heatmap of mean relative abundance of bacterial OTUs found to be consistently significantly different between responders’ baseline and all post-FMT samples as well as responders’ baseline and donor samples. OTUs were not found to be significantly different in nonresponders but were included for comparison. Tests were performed using LEfSe, and a strict cutoff LDA score of >4 and P value of <0.05 were applied. (F) Heatmap of mean relative abundance of bacterial OTUs found to be consistently significantly different between nonresponders’ baseline and all post-FMT samples as well as nonresponders’ baseline and donor samples. Tests performed using LEfSe and a cutoff LDA score of >3.5 and P value of <0.05 were applied.
FIG 2
FIG 2
Mycobiome diversity and composition. (A) Relative abundance of Penicillium OTU14 which was significantly different between responders and nonresponders at baseline. Testing was performed using LEfSe (LDA score, 3.68; P < 0.05). (B) Species richness (d) at baseline (P-W0) and week 1 of FMT (P-W1) for responders (-Resp) and nonresponders (-Non). Significance was tested using Welch’s t test for each of the two comparisons reported. (C) Coexclusion relationships between Candida OTU2 with similarity to Candida albicans and two bacterial taxa (OTU18 and OTU34). Nonparametric relationships were identified using the MINE framework. OTU18 was classified to Dorea, and OTU34 was classified to Clostridium XVIII. (D) Heatmaps of mean relative abundances of bacterial OTU18 and OTU34 across responders’ baseline and post-FMT samples as well as donor samples.
FIG 3
FIG 3
Donor contribution to the patient bacterial component of the microbiome. (A) Contribution was determined using SourceTracker with donor samples assigned as specific sources (one-to-one) to their matched patient samples (sinks). Significance was tested using ANOVA with Tukey’s multiple-comparison tests. Donor contribution to the baseline sample was significantly lower than the post-FMT samples with the exception of P-W12. (B) Donor contribution was stratified according to response (-Resp) or lack of response (-Non) to FMT. Contribution was determined using SourceTracker with donor samples assigned as specific sources to their matched patient samples (sinks). Patient samples at recurrence were labeled in black and with patient number. Significance was tested using ANOVA with Tukey’s multiple-comparison tests. Only baseline samples of responders (P-W0-Resp) were significantly different from other groups (denoted by asterisks above groups).*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
FIG 4
FIG 4
Bacterial communities in donor samples. (A) Principal-coordinate analysis of Bray-Curtis resemblance matrix generated from square-root-transformed relative abundances of bacterial OTUs. All donors (A, B, C, and D) were found to be significantly different from each other using pairwise PERMANOVA (P < 0.024 for all) except for donors B and C (PERMANOVA: t = 1.46, P = 0.054; ANOSIM: r = 0.288, P = 0.087). (B) Heatmap of mean relative abundance of bacterial OTUs found to be significantly different between donor samples that led to therapy success and those that led to therapy failure. Tests were performed using LEfSe and a cutoff LDA score of >3.5 and P value of <0.05 were applied.
FIG 5
FIG 5
Differences in the bacterial communities between types of C. difficile infection. Only responders were included in this analysis. Severe C. difficile infections or those with persisting disease despite antibiotics were excluded due to low numbers leaving primary (-P) and recurrent (-R) infections. (A) Shannon’s diversity (H´) index across different sample groups. Significance was tested using ANOVA with Tukey’s multiple-comparison test, and only P-W0-P was found to be statistically significantly different from other post-FMT groups. (B) Principal-coordinate analysis of Bray-Curtis resemblance matrix generated from square-root-transformed relative abundances of bacterial OTUs. All patient subgroups (P-) were significantly different from the donors (DON) when tested using pairwise PERMANOVA (P < 0.046 for all). P-W0-P was consistently significantly different from all other post-FMT sample groups in primary CDI (P < 0.015 for all). This result could not be replicated in the patients with recurrent CDI. (C) Heatmap of mean relative abundance of bacterial OTUs found to be significantly different between patients with primary and recurrent CDI at baseline. Tests were performed using LEfSe, and a strict cutoff LDA score of >4 and P value of <0.05 were applied.

References

    1. Kelly CP. 2012. Can we identify patients at high risk of recurrent Clostridium difficile infection? Clin Microbiol Infect 18:21–27. doi:10.1111/1469-0691.12046.
    1. Haifer C, Kelly CR, Paramsothy S, Andresen D, Papanicolas LE, McKew GL, Borody TJ, Kamm M, Costello SP, Andrews JM, Begun J, Chan HT, Connor S, Ghaly S, Johnson PD, Lemberg DA, Paramsothy R, Redmond A, Sheorey H, van der Poorten D, Leong RW. 2020. Australian consensus statements for the regulation, production and use of faecal microbiota transplantation in clinical practice. Gut 69:801–810. doi:10.1136/gutjnl-2019-320260.
    1. Cammarota G, Ianiro G, Tilg H, Rajilic-Stojanovic M, Kump P, Satokari R, Sokol H, Arkkila P, Pintus C, Hart A, Segal J, Aloi M, Masucci L, Molinaro A, Scaldaferri F, Gasbarrini G, Lopez-Sanroman A, Link A, de Groot P, de Vos WM, Hogenauer C, Malfertheiner P, Mattila E, Milosavljevic T, Nieuwdorp M, Sanguinetti M, Simren M, Gasbarrini A, The European FMT Working Group. 2017. European consensus conference on faecal microbiota transplantation in clinical practice. Gut 66:569–580. doi:10.1136/gutjnl-2016-313017.
    1. Staley C, Hamilton MJ, Vaughn BP, Graiziger CT, Newman KM, Kabage AJ, Sadowsky MJ, Khoruts A. 2017. Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study. Am J Gastroenterol 112:940–947. doi:10.1038/ajg.2017.6.
    1. De Zoysa P, Kingston-Smith H, Maistry P, Jayewardene AF, Borody TJ. 2017. Treatment-naïve ulcerative colitis patient treated with lyophilized full spectrum microbiota: a case study. Am J Gastroenterol 112:S1095–S1096. doi:10.14309/00000434-201710001-01984.
    1. Ghimire S, Roy C, Wongkuna S, Antony L, Maji A, Keena MC, Foley A, Scaria J. 2020. Identification of Clostridioides difficile-inhibiting gut commensals using culturomics, phenotyping, and combinatorial community assembly. mSystems 5:e00620-19. doi:10.1128/mSystems.00620-19.
    1. Staley C, Kaiser T, Vaughn BP, Graiziger CT, Hamilton MJ, Rehman TU, Song K, Khoruts A, Sadowsky MJ. 2018. Predicting recurrence of Clostridium difficile infection following encapsulated fecal microbiota transplantation. Microbiome 6:166. doi:10.1186/s40168-018-0549-6.
    1. Mullish BH, McDonald JAK, Pechlivanis A, Allegretti JR, Kao D, Barker GF, Kapila D, Petrof EO, Joyce SA, Gahan CGM, Glegola-Madejska I, Williams HRT, Holmes E, Clarke TB, Thursz MR, Marchesi JR. 2019. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection. Gut 68:1791–1800. doi:10.1136/gutjnl-2018-317842.
    1. McDonald JAK, Mullish BH, Pechlivanis A, Liu Z, Brignardello J, Kao D, Holmes E, Li JV, Clarke TB, Thursz MR, Marchesi JR. 2018. Inhibiting growth of Clostridioides difficile by restoring valerate, produced by the intestinal microbiota. Gastroenterology 155:1495–1507.e15. doi:10.1053/j.gastro.2018.07.014.
    1. He M, Miyajima F, Roberts P, Ellison L, Pickard DJ, Martin MJ, Connor TR, Harris SR, Fairley D, Bamford KB, D'Arc S, Brazier J, Brown D, Coia JE, Douce G, Gerding D, Kim HJ, Koh TH, Kato H, Senoh M, Louie T, Michell S, Butt E, Peacock SJ, Brown NM, Riley T, Songer G, Wilcox M, Pirmohamed M, Kuijper E, Hawkey P, Wren BW, Dougan G, Parkhill J, Lawley TD. 2013. Emergence and global spread of epidemic healthcare-associated Clostridium difficile. Nat Genet 45:109–113. doi:10.1038/ng.2478.
    1. Camacho-Ortiz A, Gutiérrez-Delgado EM, Garcia-Mazcorro JF, Mendoza-Olazarán S, Martínez-Meléndez A, Palau-Davila L, Baines SD, Maldonado-Garza H, Garza-González E. 2017. Randomized clinical trial to evaluate the effect of fecal microbiota transplant for initial Clostridium difficile infection in intestinal microbiome. PLoS One 12:e0189768. doi:10.1371/journal.pone.0189768.
    1. Juul FE, Garborg K, Bretthauer M, Skudal H, Oines MN, Wiig H, Rose O, Seip B, Lamont JT, Midtvedt T, Valeur J, Kalager M, Holme O, Helsingen L, Loberg M, Adami HO. 2018. Fecal microbiota transplantation for primary Clostridium difficile infection. N Engl J Med 378:2535–2536. doi:10.1056/NEJMc1803103.
    1. Hocquart M, Lagier JC, Cassir N, Saidani N, Eldin C, Kerbaj J, Delord M, Valles C, Brouqui P, Raoult D, Million M. 2018. Early fecal microbiota transplantation improves survival in severe Clostridium difficile infections. Clin Infect Dis 66:645–650. doi:10.1093/cid/cix762.
    1. Stewart DB, Sr, Wright JR, Fowler M, McLimans CJ, Tokarev V, Amaniera I, Baker O, Wong HT, Brabec J, Drucker R, Lamendella R. 2019. Integrated meta-omics reveals a fungus-associated bacteriome and distinct functional pathways in Clostridioides difficile infection. mSphere 4:e00454-19. doi:10.1128/mSphere.00454-19.
    1. Sangster W, Hegarty JP, Schieffer KM, Wright JR, Hackman J, Toole DR, Lamendella R, Stewart DB, Sr. 2016. Bacterial and fungal microbiota changes distinguish C. difficile infection from other forms of diarrhea: results of a prospective inpatient study. Front Microbiol 7:789. doi:10.3389/fmicb.2016.00789.
    1. Zuo T, Wong SH, Cheung CP, Lam K, Lui R, Cheung K, Zhang F, Tang W, Ching JYL, Wu JCY, Chan PKS, Sung JJY, Yu J, Chan FKL, Ng SC. 2018. Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat Commun 9:3663. doi:10.1038/s41467-018-06103-6.
    1. Collins DA, Sohn KM, Wu Y, Ouchi K, Ishii Y, Elliott B, Riley TV, Tateda K, Clostridioides difficile Asia-Pacific Study Group. 2020. Clostridioides difficile infection in the Asia-Pacific region. Emerg Microbes Infect 9:42–52. doi:10.1080/22221751.2019.1702480.
    1. Jiang ZD, Jenq RR, Ajami NJ, Petrosino JF, Alexander AA, Ke S, Iqbal T, DuPont AW, Muldrew K, Shi Y, Peterson C, Do KA, DuPont HL. 2018. Safety and preliminary efficacy of orally administered lyophilized fecal microbiota product compared with frozen product given by enema for recurrent Clostridium difficile infection: a randomized clinical trial. PLoS One 13:e0205064. doi:10.1371/journal.pone.0205064.
    1. Paramsothy S, Borody TJ, Lin E, Finlayson S, Walsh AJ, Samuel D, van den Bogaerde J, Leong RW, Connor S, Ng W, Mitchell HM, Kaakoush N, Kamm MA. 2015. Donor recruitment for fecal microbiota transplantation. Inflamm Bowel Dis 21:1600–1606. doi:10.1097/MIB.0000000000000405.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, Knight R. 2011. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A 108:4516–4522. doi:10.1073/pnas.1000080107.
    1. Deshpande NP, Riordan SM, Castano-Rodríguez N, Wilkins MR, Kaakoush NO. 2018. Signatures within the esophageal microbiome are associated with host genetics, age, and disease. Microbiome 6:227. doi:10.1186/s40168-018-0611-4.
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, Sahl JW, Stres B, Thallinger GG, Van Horn DJ, Weber CF. 2009. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 75:7537–7541. doi:10.1128/AEM.01541-09.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. 2013. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 79:5112–5120. doi:10.1128/AEM.01043-13.
    1. Bissett A, Fitzgerald A, Court L, Meintjes T, Mele PM, Reith F, Dennis PG, Breed MF, Brown B, Brown MV, Brugger J, Byrne M, Caddy-Retalic S, Carmody B, Coates DJ, Correa C, Ferrari BC, Gupta VV, Hamonts K, Haslem A, Hugenholtz P, Karan M, Koval J, Lowe AJ, Macdonald S, McGrath L, Martin D, Morgan M, North KI, Paungfoo-Lonhienne C, Pendall E, Phillips L, Pirzl R, Powell JR, Ragan MA, Schmidt S, Seymour N, Snape I, Stephen JR, Stevens M, Tinning M, Williams K, Yeoh YK, Zammit CM, Young A. 2016. Introducing BASE: the Biomes of Australian Soil Environments soil microbial diversity database. Gigascience 5:21. doi:10.1186/s13742-016-0126-5.
    1. Ihrmark K, Bodeker IT, Cruz-Martinez K, Friberg H, Kubartova A, Schenck J, Strid Y, Stenlid J, Brandstrom-Durling M, Clemmensen KE, Lindahl BD. 2012. New primers to amplify the fungal ITS2 region–evaluation by 454-sequencing of artificial and natural communities. FEMS Microbiol Ecol 82:666–677. doi:10.1111/j.1574-6941.2012.01437.x.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. 2011. Metagenomic biomarker discovery and explanation. Genome Biol 12:R60. doi:10.1186/gb-2011-12-6-r60.
    1. Knights D, Kuczynski J, Charlson ES, Zaneveld J, Mozer MC, Collman RG, Bushman FD, Knight R, Kelley ST. 2011. Bayesian community-wide culture-independent microbial source tracking. Nat Methods 8:761–763. doi:10.1038/nmeth.1650.
    1. Feng K, Zhang Z, Cai W, Liu W, Xu M, Yin H, Wang A, He Z, Deng Y. 2017. Biodiversity and species competition regulate the resilience of microbial biofilm community. Mol Ecol 26:6170–6182. doi:10.1111/mec.14356.
    1. Reshef DN, Reshef YA, Finucane HK, Grossman SR, McVean G, Turnbaugh PJ, Lander ES, Mitzenmacher M, Sabeti PC. 2011. Detecting novel associations in large data sets. Science 334:1518–1524. doi:10.1126/science.1205438.

Source: PubMed

3
Abonnieren