Microbiota-based markers predictive of development of Clostridioides difficile infection

Matilda Berkell, Mohamed Mysara, Basil Britto Xavier, Cornelis H van Werkhoven, Pieter Monsieurs, Christine Lammens, Annie Ducher, Maria J G T Vehreschild, Herman Goossens, Jean de Gunzburg, Marc J M Bonten, Surbhi Malhotra-Kumar, ANTICIPATE study group, Annemarie Engbers, Marieke de Regt, Lena M Biehl, Oliver A Cornely, Nathalie Jazmati, Marie-Noelle Bouverne, Frederique Sablier-Gallis, France Mentré, Uta Merle, Andreas Stallmach, Jan Rupp, Johannes Bogner, Christoph Lübbert, Gerda Silling, Oliver Witzke, Achilleas Gikas, Sofia Maraki, George Daikos, Sotirios Tsiodras, Athanasios Skoutelis, Helen Sambatakou, Miquel Pujol, M Angeles Dominguez-Luzon, Jose M Aguado, Emilio Bouza, Javier Cobo, Jesús Rodríguez-Baño, Benito Almirante, Julian de la Torre Cisneros, Simin A Florescu, Maria Nica, Andrei Vata, Adriana Hristea, Mihaela Lupse, Delia Herghea, Deborah Postil, Olivier Barraud, Jean-Michel Molina, Victoire De Lastours, Thomas Guimard, Jean-Philippe Talarmin, Xavier Duval, Louis Bernard, Odile Launay, Matilda Berkell, Mohamed Mysara, Basil Britto Xavier, Cornelis H van Werkhoven, Pieter Monsieurs, Christine Lammens, Annie Ducher, Maria J G T Vehreschild, Herman Goossens, Jean de Gunzburg, Marc J M Bonten, Surbhi Malhotra-Kumar, ANTICIPATE study group, Annemarie Engbers, Marieke de Regt, Lena M Biehl, Oliver A Cornely, Nathalie Jazmati, Marie-Noelle Bouverne, Frederique Sablier-Gallis, France Mentré, Uta Merle, Andreas Stallmach, Jan Rupp, Johannes Bogner, Christoph Lübbert, Gerda Silling, Oliver Witzke, Achilleas Gikas, Sofia Maraki, George Daikos, Sotirios Tsiodras, Athanasios Skoutelis, Helen Sambatakou, Miquel Pujol, M Angeles Dominguez-Luzon, Jose M Aguado, Emilio Bouza, Javier Cobo, Jesús Rodríguez-Baño, Benito Almirante, Julian de la Torre Cisneros, Simin A Florescu, Maria Nica, Andrei Vata, Adriana Hristea, Mihaela Lupse, Delia Herghea, Deborah Postil, Olivier Barraud, Jean-Michel Molina, Victoire De Lastours, Thomas Guimard, Jean-Philippe Talarmin, Xavier Duval, Louis Bernard, Odile Launay

Abstract

Antibiotic-induced modulation of the intestinal microbiota can lead to Clostridioides difficile infection (CDI), which is associated with considerable morbidity, mortality, and healthcare-costs globally. Therefore, identification of markers predictive of CDI could substantially contribute to guiding therapy and decreasing the infection burden. Here, we analyze the intestinal microbiota of hospitalized patients at increased CDI risk in a prospective, 90-day cohort-study before and after antibiotic treatment and at diarrhea onset. We show that patients developing CDI already exhibit significantly lower diversity before antibiotic treatment and a distinct microbiota enriched in Enterococcus and depleted of Ruminococcus, Blautia, Prevotella and Bifidobacterium compared to non-CDI patients. We find that antibiotic treatment-induced dysbiosis is class-specific with beta-lactams further increasing enterococcal abundance. Our findings, validated in an independent prospective patient cohort developing CDI, can be exploited to enrich for high-risk patients in prospective clinical trials, and to develop predictive microbiota-based diagnostics for management of patients at risk for CDI.

Conflict of interest statement

C.H.v.W. received speaker fees from Pfizer and Merck/MSD, and non-financial research support from bioMérieux. A.D. is an employee and shareholder of Da Volterra. J.d.G. is a consultant and shareholder of Da Volterra. M.J.G.T.V. has received research grants from 3M, Astellas Pharma, Da Volterra, Gilead Sciences, Glycom, MaaT Pharma, Merck/MSD, Organobalance, Seres Therapeutics; speaker fees from Astellas Pharma, Basilea, Gilead Sciences, Merck/MSD, Organobalance, Pfizer and has been a consultant to Alb Fils Kliniken GmbH, Astellas Pharma, Da Volterra, Ferring, MaaT Pharma, Merck/MSD and Summit Therapeutics. M.B., M.M., B.B.X., C.L., P.M., M.J.M.B., H.G., S.M.-K. report no competing interests.

Figures

Fig. 1. Patient and sample flow in…
Fig. 1. Patient and sample flow in the study.
The flow chart provides an overview of participating patients in each processing step, number of samples collected at each timepoint, and reasons for sample exclusion or non-collection. D1: rectal swab sample collected upon study enrollment. D6: rectal swab collected ~6 days after initiation and at the end of antibiotic treatment. S1: stool sample collected at the first occurrence of diarrhea (variable time-point). AAD: patients with non-C. difficile antibiotic-associated diarrhea. CDI: patients with confirmed C. difficile infection. ND: non-diarrheic patients. PBL: penicillin + beta-lactamase inhibitor. OBL: other beta-lactamase antibiotics. FQN: fluoroquinolones.
Fig. 2. C. difficile and C. bartlettii…
Fig. 2. C. difficile and C. bartlettii carriage in the analyzed population at D1 and D6.
Oligotyping revealed diversity within the OTU classified as Clostridium XI wherein reads were divided into C. difficile (green) and C. bartlettii (blue). a Rectal swab samples collected at the time of study enrollment (D1), and ~6 days later at the end of treatment (D6) harbored reads classified as both C. difficile and C. bartlettii. Patients that were defined as C. difficile carriers due to the presence of C. difficile reads after oligotyping showed no clear link to clinical outcome. b Similar carriage rates of the OTU Clostridium XI were identified in patients both at D1 (5.40%, n = 51) and D6 (6.78%, n = 50) with varying relative abundances of C. difficile and C. bartlettii reads as demonstrated in the Circos plot. Patients at D6 generally show higher relative abundance of C. difficile than of C. bartlettii compared to D1. D1: rectal swab sample collected upon study enrollment. D6: rectal swab collected ~6 days after initiation and at the end of antibiotic treatment. AAD: patients with non-C. difficile antibiotic-associated diarrhea. CDI: patients with confirmed C. difficile infection. ND: non-diarrheic patients. NA: patients without known CDI status and/or early study termination. OTU: operational taxonomic unit.
Fig. 3. Characterization of microbial diversity in…
Fig. 3. Characterization of microbial diversity in baseline (D1) samples.
a CDI patients (n = 14, brown) display distinctly lower alpha diversity expressed by the Shannon index compared to AAD (n = 64, blue, p = 0.037) and ND patients (n = 669, green, p = 0.005) at D1. AAD patients similarly display lower Shannon diversity compared to ND patients, however not sufficient to be statistically significant (p = 0.087). b Both CDI and AAD patients display lower diversity expressed by the Chao1 index compared to ND patients (p = 0.001 and 0.017, respectively) at D1. Compared to patients who develop AAD, CDI patients display lower Chao1 indices (p = 0.049). c Cladogram generated by LEfSe demonstrating significantly higher abundances of Actinobacteria, Alphaproteobacteria, and Enterococcus spp. in the gut microbiota of CDI patients at baseline (D1) compared to AAD and ND patients. The cladogram shows distinctly abundant taxa of interest. For more details, see Supplementary Fig. 4. Alpha diversity indices were compared using the non-parametric two-sided Wilcoxon rank sum test followed by Bonferroni correction of p-values. Box plots indicate median (middle line), 25th, 75th percentile (box), and 5th and 95th percentile (whiskers) as well as outliers (gray single dots). AAD: patients with non-C. difficile antibiotic-associated diarrhea. CDI: patients with confirmed C. difficile infection. ND: non-diarrheic patients. LEfSe: Linear discriminant analysis effect size. LDA: linear discriminant analysis score. *p < 0.05. **p ≤ 0.01. ***p ≤ 0.001.
Fig. 4. Longitudinal analysis of the impact…
Fig. 4. Longitudinal analysis of the impact of antibiotic therapy on the intestinal microbiota.
Microbial diversity was compared prior to broad-spectrum antibiotic treatment at D1 (green) and after treatment at D6 (purple) following treatment with PBLs (n = 194), OBLs (n = 133), and FQNs (n = 63). a A distinct reduction was observed in Shannon diversity in patients treated with all antibiotic classes (p = 8.98*10−6, p = 2.06*10−5, and p = 0.007, respectively). b Similarly, distinct reductions in Chao1 diversity was observed following treatment with all antibiotic classes (p = 0.011, p = 0.001, and p = 9.26 × 10−5, respectively). c Treatment with each antibiotic class resulted in a shift in microbial composition illustrated by the Jaccard distances between the D1 and D6 samples. d The heatmap illustrates distinctly abundant taxa (LDA > 3.0) identified using LEfSe. For more details, see Supplementary Table 10. Alpha diversity indices were compared using the paired two-sided non-parametric Wilcoxon signed rank test. Box plots in indicate median (middle line), 25th, 75th percentile (box), and 5th and 95th percentile (whiskers) as well as outliers (gray single dots). PBL: penicillin + beta-lactamase inhibitor. OBL: other beta-lactam antibiotics. FQN: fluoroquinolones. LDA: Linear discriminant analysis score. D1: rectal swab sample collected upon study enrollment. D6: rectal swab collected ~6 days after initiation and at the end of antibiotic treatment. LEfSe: linear discriminant analysis effect size. LDA: linear discriminant analysis score. OTU: operational taxonomic unit. *p < 0.05, **p ≤ 0.01, ***p ≤ 0.001.
Fig. 5. Longitudinal analysis of microbial diversity…
Fig. 5. Longitudinal analysis of microbial diversity and dysbiosis in patients developing AAD.
Microbial diversity and composition in patients developing AAD in the study population (n = 26) was assessed at D1 (green), D6 (blue), S1(purple). a Gradual decrease in Shannon diversity was observed between all timepoints (from D1 to D6: p = 0.018, from D1 to S1: p = 2.74*10−5, from D6 to S1: p = 0.007). b Similar trends are observed for the Chao1 index (Friedman rank sum: p = 0.054). c Multi-dimensional scaling (MDS) shows distinct clusters for samples collected at each timepoint. d Comparison of the microbiota composition at D1 and S1 conducted using LEfSe (LDA > 2.0) shows large changes in the Firmicutes and Proteobacteria phyla for AAD patients. Proteobacteria are significantly reduced at the occurrence of AAD, and a shift is observed from the Clostridia to Bacilli class at the instance of diarrhea. The cladogram shows distinct taxa of interest. For more details, see Supplementary Fig. 5. Alpha diversity indices were compared using the paired two-sided non-parametric Wilcoxon signed rank test followed by Bonferroni correction of p-values. Box plots indicate median (middle line), 25th, 75th percentile (box), and 5th and 95th percentile (whiskers) as well as outliers (gray single dots). AAD: patients with non-C. difficile antibiotic-associated diarrhea. D1: rectal swab sample collected upon study enrollment. D6: rectal swab collected ~6 days after initiation and at the end of antibiotic treatment. S1: stool sample collected at the first occurrence of diarrhea (variable time-point). LEfSe: linear discriminant analysis effect size. LDA: Linear discriminant analysis score. *p < 0.05, **p ≤ 0.01, ***p ≤ 0.001.

References

    1. Freeman J, et al. The changing epidemiology of Clostridium difficile infections. Clin. Microbiol. Rev. 2010;23:529–549. doi: 10.1128/CMR.00082-09.
    1. Smits WK, Lyras D, Lacy DB, Wilcox MH, Kuijper EJ. Clostridium difficile infection. Nat. Rev. Dis. Prim. 2016;2:16020. doi: 10.1038/nrdp.2016.20.
    1. Magill SS, et al. Multistate point-prevalence survey of health care-associated infections. New Engl. J. Med. 2014;370:1198–1208. doi: 10.1056/NEJMoa1306801.
    1. Slimings C, Riley TV. Antibiotics and hospital-acquiredClostridium difficile infection: update of systematic review and meta-analysis. J. Antimicrob. Chemother. 2014;69:881–891. doi: 10.1093/jac/dkt477.
    1. Owens RC, Jr., Donskey CJ, Gaynes RP, Loo VG, Muto CA. Antimicrobial‐associated risk factors for Clostridium difficile infection. Clin. Infect. Dis. 2008;46:S19–S31. doi: 10.1086/521859.
    1. Ananthakrishnan AN. Clostridium difficile infection: epidemiology, risk factors and management. Nat. Rev. Gastroenterol. Hepatol. 2011;8:17–26. doi: 10.1038/nrgastro.2010.190.
    1. Pepin J, et al. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a Cohort Study during an epidemic in Quebec. Clin. Infect. Dis. 2005;41:1254–1260. doi: 10.1086/496986.
    1. Vardakas KZ, Trigkidis KK, Boukouvala E, Falagas ME. Clostridium difficile infection following systemic antibiotic administration in randomised controlled trials: a systematic review and meta-analysis. Int. J. Antimicrob. Agents. 2016;48:1–10. doi: 10.1016/j.ijantimicag.2016.03.008.
    1. Brown KA, Khanafer N, Daneman N, Fisman N. Meta-analysis of antibiotics and the risk of community-associatedClostridium difficile infection. Antimicrob. Agents Chemother. 2013;57:2326–2332. doi: 10.1128/AAC.02176-12.
    1. Battaglioli EJ, et al. Clostridioides difficile uses amino acids associated with gut microbial dysbiosis in a subset of patients with diarrhea. Sci. Transl. Med. 2018;10:eaam7019. doi: 10.1126/scitranslmed.aam7019.
    1. Manges AR, et al. Comparative metagenomic study of alterations to the intestinal microbiota and risk of nosocomial Clostridum difficile-associated disease. J. Infect. Dis. 2010;202:1877–1884. doi: 10.1086/657319.
    1. Han SH, Yi J, Kim JH, Lee SW, Moon HW. Composition of gut microbiota in patients with toxigenic Clostridioides (Clostridium) difficile: comparison between subgroups according to clinical criteria and toxin gene load. PLoS ONE. 2019;14:e0212626. doi: 10.1371/journal.pone.0212626.
    1. Seekatz AM, Rao K, Santhosh K, Young VB. Dynamics of the fecal microbiome in patients with recurrent and nonrecurrent Clostridium difficile infection. Genome Med. 2016;8:1–11. doi: 10.1186/s13073-016-0298-8.
    1. Gu S, et al. Identification of key taxa that favor intestinal colonization of Clostridium difficile in an adult Chinese population. Microbes Infect. 2016;18:30–38. doi: 10.1016/j.micinf.2015.09.008.
    1. Antharam VC, et al. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J. Clin. Microbiol. 2013;51:2884–2892. doi: 10.1128/JCM.00845-13.
    1. Schubert AM, et al. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy. MBio. 2014;5:1–9. doi: 10.1128/mBio.01021-14.
    1. Buffie CG, et al. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect. Immun. 2012;80:62–73. doi: 10.1128/IAI.05496-11.
    1. Vincent C, et al. Reductions in intestinal Clostridiales precede the development of nosocomial Clostridium difficile infection. Microbiome. 2013;1:1–11. doi: 10.1186/2049-2618-1-18.
    1. Zhang L, et al. Insight into alteration of gut microbiota in Clostridium difficile infection and asymptomatic C. difficile colonization. Anaerobe. 2015;34:1–7. doi: 10.1016/j.anaerobe.2015.03.008.
    1. Khanna S, et al. Gut microbiome predictors of treatment response and recurrence in primary Clostridium difficile infection. Aliment. Pharmacol. Ther. 2016;44:715–727. doi: 10.1111/apt.13750.
    1. Loo VG, et al. Host and pathogen factors for Clostridium difficile infection and colonization. New Engl. J. Med. 2011;365:1693–1703. doi: 10.1056/NEJMoa1012413.
    1. Palleja A, et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat. Microbiol. 2018;3:1255–1265. doi: 10.1038/s41564-018-0257-9.
    1. Jernberg C, Löfmark S, Edlund C, Jansson JK. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology. 2010;156:3216–3223. doi: 10.1099/mic.0.040618-0.
    1. Jernberg C, Löfmark S, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 2007;1:56–66. doi: 10.1038/ismej.2007.3.
    1. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl Acad. Sci. USA. 2011;108:4554–4561. doi: 10.1073/pnas.1000087107.
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280. doi: 10.1371/journal.pbio.0060280.
    1. Panda S, et al. Short-term effect of antibiotics on human gut microbiota. PLoS ONE. 2014;9:e95476. doi: 10.1371/journal.pone.0095476.
    1. Zaura E, et al. Same exposure but two radically different responses to antibiotics: resilience of the salivary microbiome versus long-term microbial shifts in feces. MBio. 2015;6:e01693–15. doi: 10.1128/mBio.01693-15.
    1. van Werkhoven, C. H. et al. Incidence and predictive biomarkers of Clostridioides difficile infection in hospitalized patients receiving broad-spectrum antibiotics. Nat. Commun. 10.1038/s41467-021-22269-y (2021).
    1. Gerritsen J, et al. Characterization of Romboutsia ilealis gen. nov., sp. nov., isolated from the gastro-intestinal tract of a rat, and proposal for the reclassification of five closely related members of the genus Clostridium into the genera Romboutsia gen. nov., Intestinibacter gen. nov., Terrisporobacter gen. nov. and Asaccharospora gen. nov. Int. J. Syst. Evol. Microbiol. 2014;64:1600–1616. doi: 10.1099/ijs.0.059543-0.
    1. McTeague M, Finegold S, Summanen P, Liu C, Song Y. Clostridium bartlettii sp. nov., isolated from human faeces. Anaerobe. 2004;10:179–184. doi: 10.1016/j.anaerobe.2004.04.004.
    1. García-Solache M, Rice LB. The Enterococcus: a model of adaptability to its environment. Clin. Microbiol. Rev. 2019;32:1–28. doi: 10.1128/CMR.00058-18.
    1. Arias CA, Murray BE. The rise of the Enterococcus: beyond vancomycin resistance. Nat. Rev. Microbiol. 2012;10:266–278. doi: 10.1038/nrmicro2761.
    1. Guzman Prieto AM, et al. Global emergence and dissemination of enterococci as nosocomial pathogens: attack of the clones? Front. Microbiol. 2016;7:1–15. doi: 10.3389/fmicb.2016.00788.
    1. Axelrad JE, et al. Gut colonization with vancomycin-resistantEnterococcus and risk for subsequent enteric infection. Gut Pathog. 2018;10:1–9. doi: 10.1186/s13099-018-0259-4.
    1. Özsoy S, İlki A. Detection of vancomycin-resistant enterococci (VRE) in stool specimens submitted for Clostridium difficile toxin testing. Braz. J. Microbiol. 2017;48:489–492. doi: 10.1016/j.bjm.2016.12.012.
    1. Vital M, Howe AC, Revealing TJ. The bacterial synthesis pathways by analyzing (Meta) genomic data. MBio. 2014;5:e00889–14. doi: 10.1128/mBio.00889-14.
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand. J. Gastroenterol. 1997;32:920–924. doi: 10.3109/00365529709011203.
    1. Crobach MJT, et al. European Society of Clinical Microbiology and Infectious Diseases: update of the diagnostic guidance document for Clostridium difficile infection. Clin. Microbiol. Infect. 2016;22:S63–S81. doi: 10.1016/j.cmi.2016.03.010.
    1. Mysara M, Njima M, Leys N, Raes J, Monsieurs P. From reads to operational taxonomic units: an ensemble processing pipeline for MiSeq amplicon sequencing data. Gigascience. 2017;6:1–10. doi: 10.1093/gigascience/giw017.
    1. Bankevich A, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. 2012;19:455–477. doi: 10.1089/cmb.2012.0021.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl. Environ. Microbiol. 2013;79:5112–5120. doi: 10.1128/AEM.01043-13.
    1. Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Pruesse E, et al. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res. 2007;35:7188–7196. doi: 10.1093/nar/gkm864.
    1. Mysara M, Leys N, Raes J, Monsieurs P. IPED: A highly efficient denoising tool for Illumina MiSeq paired-end 16S rRNA gene amplicon sequencing data. BMC Bioinforma. 2016;17:1–11. doi: 10.1186/s12859-016-1061-2.
    1. Mysara M, Saeys Y, Leys N, Raes J, Monsieurs P. CATCh, an ensemble classifier for chimera detection in 16s rRNA sequencing studies. Appl. Environ. Microbiol. 2015;81:1573–1584. doi: 10.1128/AEM.02896-14.
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods. 2013;10:996–998. doi: 10.1038/nmeth.2604.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naïve Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 2007;73:5261–5267. doi: 10.1128/AEM.00062-07.
    1. Eren AM, et al. Oligotyping: differentiating between closely related microbial taxa using 16S rRNA gene data. Methods Ecol. Evol. 2013;4:1111–1119. doi: 10.1111/2041-210X.12114.
    1. Lagkouvardos I, Fischer S, Kumar N, Clavel T. Rhea: a transparent and modular R pipeline for microbial profiling based on 16S rRNA gene amplicons. PeerJ. 2017;5:e2836. doi: 10.7717/peerj.2836.
    1. Team. RStudio: Integrated Development for R, Vol. 42, 14 (RStudio, Inc., 2015).
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Krzywinski M, et al. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–1645. doi: 10.1101/gr.092759.109.
    1. Wood DE, Lu J, Langmead B. Improved metagenomic analysis with Kraken 2. Genome Biol. 2019;20:1–13. doi: 10.1186/s13059-019-1891-0.

Source: PubMed

3
Prenumerera