Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation

Ying Taur, Joao B Xavier, Lauren Lipuma, Carles Ubeda, Jenna Goldberg, Asia Gobourne, Yeon Joo Lee, Krista A Dubin, Nicholas D Socci, Agnes Viale, Miguel-Angel Perales, Robert R Jenq, Marcel R M van den Brink, Eric G Pamer, Ying Taur, Joao B Xavier, Lauren Lipuma, Carles Ubeda, Jenna Goldberg, Asia Gobourne, Yeon Joo Lee, Krista A Dubin, Nicholas D Socci, Agnes Viale, Miguel-Angel Perales, Robert R Jenq, Marcel R M van den Brink, Eric G Pamer

Abstract

Background: Bacteremia is a frequent complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). It is unclear whether changes in the intestinal microbiota during allo-HSCT contribute to the development of bacteremia. We examined the microbiota of patients undergoing allo-HSCT, and correlated microbial shifts with the risk of bacteremia.

Methods: Fecal specimens were collected longitudinally from 94 patients undergoing allo-HSCT, from before transplant until 35 days after transplant. The intestinal microbiota was characterized by 454 pyrosequencing of the V1-V3 region of bacterial 16S ribosomal RNA genes. Microbial diversity was estimated by grouping sequences into operational taxonomic units and calculating the Shannon diversity index. Phylogenetic classification was obtained using the Ribosomal Database Project classifier. Associations of the microbiota with clinical predictors and outcomes were evaluated.

Results: During allo-HSCT, patients developed reduced diversity, with marked shifts in bacterial populations inhabiting the gut. Intestinal domination, defined as occupation of at least 30% of the microbiota by a single predominating bacterial taxon, occurred frequently. Commonly encountered dominating organisms included Enterococcus, Streptococcus, and various Proteobacteria. Enterococcal domination was increased 3-fold by metronidazole administration, whereas domination by Proteobacteria was reduced 10-fold by fluoroquinolone administration. As a predictor of outcomes, enterococcal domination increased the risk of Vancomycin-resistant Enterococcus bacteremia 9-fold, and proteobacterial domination increased the risk of gram-negative rod bacteremia 5-fold.

Conclusions: During allo-HSCT, the diversity and stability of the intestinal flora are disrupted, resulting in domination by bacteria associated with subsequent bacteremia. Assessment of fecal microbiota identifies patients at highest risk for bloodstream infection during allo-HCST.

Figures

Figure 1.
Figure 1.
Changes in microbial diversity within the intestinal tract during allogeneic hematopoietic stem cell transplantation. Microbial diversity, quantified by the Shannon diversity index, was calculated for each fecal specimen of each patient and plotted over day of transplant (circles). A diversity trend (solid black line; 95% confidence intervals shown in gray) was constructed using moving average filtering.
Figure 2.
Figure 2.
Characterization of the intestinal microbiota during allogeneic hematopoietic stem cell transplantation. A, Genus-level taxonomy of 12 selected patients, plotted over day of transplant. Each stacked bar represents the microbial composition of a single fecal specimen, based on taxonomic classification. Shading between specimens are provided for interpolation and visualization of changes in relative abundances. Concurrent antibiotic administration (vancomycin, fluoroquinolones, metronidazole, and beta-lactams) and detected bacteremias are shown for each patient. Patients in the first row generally maintained diversity with few changes in composition; patients in the second row developed intestinal domination by Enterococcus; patients in the third row developed intestinal domination by Streptococcus; and patients in the fourth row developed domination with Proteobacteria. Note that patient 58 in the top row maintained diversity but developed Enterococcus domination shortly after metronidazole administration. (Plots of all 94 patients can be found in Supplementary Figure 3.) B, Kaplan-Meier plot of intestinal domination as a survival event. Crosshairs represent censored observations. Intestinal domination by Enterococcus, Streptococcus, and Proteobacteria was common and occurred at various times throughout the observation period.
Figure 3.
Figure 3.
Microbiota states and their transitions. A, Hierarchical clustering of all fecal specimens according to microbiota state based on phylogenetic classification. There was wide variation in phylogenetic composition; some specimens showed a relatively biodiverse membership, whereas others were largely dominated by a single bacterial taxon. B, Circos plots representing microbiota state transitions between consecutive specimens within the observation period. Subplots of transitions during specific transplant periods are shown in CE. C, Pre-transplant transitions revealing that patients in the “biodiverse microbiota” state tend to remain in that state; resilience of the “biodiverse” state, measured as the percentage of consecutive specimens that remain in that state, is 77%. D, Resilience of “biodiverse” state decreasing to 35% during the intra-transplant period (consecutive specimens straddling the day of stem cell infusion). E, Post-transplant transitions with the “biodiverse” state resilience staying low at 48%.

References

    1. Junghanss C, Marr KA, Carter RA, et al. Incidence and outcome of bacterial and fungal infections following nonmyeloablative compared with myeloablative allogeneic hematopoietic stem cell transplantation: a matched control study. Biol Blood Marrow Transplant. 2002;8:512–20.
    1. Blijlevens NM, Donnelly JP, de Pauw BE. mucosal barrier injury depends on conditioning regimen. Bone Marrow Transplant. 2005;35:707–11.
    1. van Vliet MJ, Harmsen HJ, de Bont ES, Tissing WJ. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 2010;6:e1000879.
    1. Jaffe D, Jakubowski A, Sepkowitz K, et al. Prevention of peritransplantation viridans streptococcal bacteremia with early vancomycin administration: a single-center observational cohort study. Clin Infect Dis. 2004;39:1625–32.
    1. Castagnola E, Bagnasco F, Faraci M, et al. Incidence of bacteremias and invasive mycoses in children undergoing allogeneic hematopoietic stem cell transplantation: a single center experience. Bone Marrow Transplant. 2008;41:339–47.
    1. Almyroudis NG, Fuller A, Jakubowski A, et al. Pre- and post-engraftment bloodstream infection rates and associated mortality in allogeneic hematopoietic stem cell transplant recipients. Transpl Infect Dis. 2005;7:11–7.
    1. Kamboj M, Chung D, Seo SK, et al. The changing epidemiology of vancomycin-resistant Enterococcus (VRE) bacteremia in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Biol Blood Marrow Transplant. 2010;16:1576–81.
    1. Weinstock DM, Conlon M, Iovino C, et al. Colonization, bloodstream infection, and mortality caused by vancomycin-resistant enterococcus early after allogeneic hematopoietic stem cell transplant. Biol Blood Marrow Transplant. 2007;13:615–21.
    1. Donskey CJ. The role of the intestinal tract as a reservoir and source for transmission of nosocomial pathogens. Clin Infect Dis. 2004;39:219–26.
    1. Gorbach SL. Microbiology of the gastrointestinal tract. In: Baron S, editor. Medical microbiology. 4th ed. Galveston, TX: 1996.
    1. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–8.
    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.
    1. Liu Z, Lozupone C, Hamady M, Bushman FD, Knight R. Short pyrosequencing reads suffice for accurate microbial community analysis. Nucleic Acids Res. 2007;35:e120.
    1. Relman DA. Microbial genomics and infectious diseases. N Engl J Med. 2011;365:347–57.
    1. Ubeda C, Taur Y, Jenq RR, et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest. 2010;120:4332–41.
    1. Schloss PD, Westcott SL, Ryabin T, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75:7537–41.
    1. Schloss PD, Gevers D, Westcott SL. Reducing the effects of PCR amplification and sequencing artifacts on 16S rRNA-based studies. PLoS One. 2011;6:e27310.
    1. Schloss PD. A high-throughput DNA sequence aligner for microbial ecology studies. PLoS One. 2009;4:e8230.
    1. Magurran AE. Measuring biological diversity. Malden, Ma: Blackwell Pub.; 2004.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73:5261–7.
    1. Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4680–7.
    1. Krzywinski M, Schein J, Birol I, et al. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–45.
    1. Bacigalupo A, Ballen K, Rizzo D, et al. Defining the intensity of conditioning regimens: working definitions. Biol Blood Marrow Transplant. 2009;15:1628–33.
    1. Jakubowski AA, Small TN, Kernan NA, et al. T cell-depleted unrelated donor stem cell transplantation provides favorable disease-free survival for adults with hematologic malignancies. Biol Blood Marrow Transplant. 2011
    1. Heinze G, Schemper M. A solution to the problem of monotone likelihood in Cox regression. Biometrics. 2001;57:114–9.
    1. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474:327–36.
    1. Young VB, Schmidt TM. Antibiotic-associated diarrhea accompanied by large-scale alterations in the composition of the fecal microbiota. J Clin Microbiol. 2004;42:1203–6.
    1. Vollaard EJ, Clasener HA. Colonization resistance. Antimicrob Agents Chemother. 1994;38:409–14.
    1. Endt K, Stecher B, Chaffron S, et al. The microbiota mediates pathogen clearance from the gut lumen after non-typhoidal Salmonella diarrhea. PLoS Pathog. 2010;6:e1001097..
    1. van der Waaij D, Berghuis-de Vries JM, Lekkerkerk L-V. Colonization resistance of the digestive tract in conventional and antibiotic-treated mice. J Hyg (Lond) 1971;69:405–11.
    1. Pultz NJ, Stiefel U, Subramanyan S, Helfand MS, Donskey CJ. Mechanisms by which anaerobic microbiota inhibit the establishment in mice of intestinal colonization by vancomycin-resistant Enterococcus. J Infect Dis. 2005;191:949–56.
    1. Bohnhoff M, Miller CP, Martin WR. Resistance of the mouse's intestinal tract to experimental Salmonella infection. II. Factors responsible for its loss following streptomycin treatment. J Exp Med. 1964;120:817–28.
    1. Zirakzadeh A, Gastineau DA, Mandrekar JN, Burke JP, Johnston PB, Patel R. Vancomycin-resistant enterococcal colonization appears associated with increased mortality among allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant. 2008;41:385–92.
    1. Chizuka A, Kami M, Kanda Y, et al. Value of surveillance blood culture for early diagnosis of occult bacteremia in patients on corticosteroid therapy following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2005;35:577–82.
    1. Donskey CJ, Hanrahan JA, Hutton RA, Rice LB. Effect of parenteral antibiotic administration on persistence of vancomycin-resistant Enterococcus faecium in the mouse gastrointestinal tract. J Infect Dis. 1999;180:384–90.
    1. Donskey CJ, Chowdhry TK, Hecker MT, et al. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med. 2000;343:1925–32.
    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(Suppl 1):4554–61.
    1. Bucaneve G, Micozzi A, Menichetti F, et al. Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N Engl J Med. 2005;353:977–87.
    1. Gafter-Gvili A, Fraser A, Paul M, Leibovici L. Meta-analysis: antibiotic prophylaxis reduces mortality in neutropenic patients. Ann Intern Med. 2005;142(12 Pt 1):979–95.
    1. Vossen JM, Heidt PJ, van den Berg H, Gerritsen EJ, Hermans J, Dooren LJ. Prevention of infection and graft-versus-host disease by suppression of intestinal microflora in children treated with allogeneic bone marrow transplantation. Eur J Clin Microbiol Infect Dis. 1990;9:14–23.
    1. Pollard M, Chang CF, Srivastava KK. The role of microflora in development of graft-versus-host disease. Transplant Proc. 1976;8:533–6.
    1. Chang JY, Antonopoulos DA, Kalra A, et al. Decreased diversity of the fecal Microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis. 2008;197:435–8.
    1. Hill GR, Ferrara JL. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95:2754–9.
    1. CDC. Central Line-Associated Bloodstream Infection (CLABSI) Event. Available at: . Accessed 23 October 2011.

Source: PubMed

3
Sottoscrivi