Microbiota dynamics in a randomized trial of gut decontamination during allogeneic hematopoietic cell transplantation

Christopher J Severyn, Benjamin A Siranosian, Sandra Tian-Jiao Kong, Angel Moreno, Michelle M Li, Nan Chen, Christine N Duncan, Steven P Margossian, Leslie E Lehmann, Shan Sun, Tessa M Andermann, Olga Birbrayer, Sophie Silverstein, Carol G Reynolds, Soomin Kim, Niaz Banaei, Jerome Ritz, Anthony A Fodor, Wendy B London, Ami S Bhatt, Jennifer S Whangbo, Christopher J Severyn, Benjamin A Siranosian, Sandra Tian-Jiao Kong, Angel Moreno, Michelle M Li, Nan Chen, Christine N Duncan, Steven P Margossian, Leslie E Lehmann, Shan Sun, Tessa M Andermann, Olga Birbrayer, Sophie Silverstein, Carol G Reynolds, Soomin Kim, Niaz Banaei, Jerome Ritz, Anthony A Fodor, Wendy B London, Ami S Bhatt, Jennifer S Whangbo

Abstract

BACKGROUNDGut decontamination (GD) can decrease the incidence and severity of acute graft-versus-host disease (aGVHD) in murine models of allogeneic hematopoietic cell transplantation (HCT). In this pilot study, we examined the impact of GD on gut microbiome composition and the incidence of aGVHD in HCT patients.METHODSWe randomized 20 patients undergoing allogeneic HCT to receive (GD) or not receive (no-GD) oral vancomycin-polymyxin B from day -5 through neutrophil engraftment. We evaluated shotgun metagenomic sequencing of serial stool samples to compare the composition and diversity of the gut microbiome between study arms. We assessed clinical outcomes in the 2 arms and performed strain-specific analyses of pathogens that caused bloodstream infections (BSI).RESULTSThe 2 arms did not differ in the predefined primary outcome of Shannon diversity of the gut microbiome at 2 weeks post-HCT (genus, P = 0.8; species, P = 0.44) or aGVHD incidence (P = 0.58). Immune reconstitution of T cell and B cell subsets was similar between groups. Five patients in the no-GD arm had 8 BSI episodes versus 1 episode in the GD arm (P = 0.09). The BSI-causing pathogens were traceable to the gut in 7 of 8 BSI episodes in the no-GD arm, including Staphylococcus species.CONCLUSIONWhile GD did not differentially affect Shannon diversity or clinical outcomes, our findings suggest that GD may protect against gut-derived BSI in HCT patients by decreasing the prevalence or abundance of gut pathogens.TRIAL REGISTRATIONClinicalTrials.gov NCT02641236.FUNDINGNIH, Damon Runyon Cancer Research Foundation, V Foundation, Sloan Foundation, Emerson Collective, and Stanford Maternal & Child Health Research Institute.

Keywords: Bacterial infections; Hematology; Infectious disease; Molecular genetics; Stem cell transplantation.

Figures

Figure 1. Study flow diagram.
Figure 1. Study flow diagram.
ClinicalTrials.gov Identifier NCT02641236. Self-reported racial and ethnic categories in Supplemental Table 11. ANC, absolute neutrophil count.
Figure 2. Study design.
Figure 2. Study design.
Twenty patients undergoing allo-HCT were randomized to 2 arms, 10 patients with GD and 10 patients with no GD. The GD arm received vancomycin-polymyxin B starting day –5 through engraftment (median neutrophil engraftment day +25, see Supplemental Figure 1) and was analyzed as intention-to-treat (Supplemental Table 1). The no-GD arm had the same stool and blood collection time points and did not receive oral vancomycin-polymyxin B. Black circles show time of stool collections, including pretransplant, weekly until engraftment, and monthly until day +100. An additional cohort of 2 healthy sibling donors serve as a stool control comparison group (Supplemental Figure 8). For immune reconstitution studies, blood samples (red circles) were collected at pretransplant, at 2 weeks, monthly for the first 3 months, and then at months 6, 9, and 12.
Figure 3. Shannon diversity is similar between…
Figure 3. Shannon diversity is similar between the GD and no-GD groups based on intention-to-treat analysis at the species taxonomic level.
Samples from patients undergoing GD (red) and no GD (blue). (A) Shannon diversity over time analyzed at the species level using local polynomial regression fitting (LOESS, locally estimated scatterplot smoothing of the mean Shannon diversity) showing similarity between the 2 groups. n = 48 samples from 10 patients in GD arm, n = 51 samples from 10 patients in no-GD arm. (B) Shannon diversity of individual patients from pretransplant (before GD antibiotics) to 2 weeks after HCT connected with a line. Boxes shown are the median with hinges at the 25% and 75%. All comparisons not significant (see Supplemental Table 4 for details) using Wilcoxon rank-sum test. n = 10 GD arm, n = 10 no-GD arm.
Figure 4. Immune reconstitution.
Figure 4. Immune reconstitution.
Peripheral blood samples at pretransplant, monthly for the first 3 months, and then months 6, 9, and 12. Shown are the median values ± interquartile range, along with the number of patients sampled at each time point below each graph. (A) CD4+ T helper cells, (B) Treg/Tcon, (C) CD8+ cytotoxic T cells, (D) CD4+ Tcon naive cells, (E) CD19+ B cells, (F) CD56+CD3– natural killer cells. For CD19+ B cells at 12 months, an uncorrected P = 0.016 with a Wilcoxon rank-sum test was not significant when tested against a stringent Bonferroni-adjusted α level of 0.0045 (0.05/11 biomarkers tested).
Figure 5. Cumulative incidence of BSI during…
Figure 5. Cumulative incidence of BSI during the first 100 days of transplant.
Patients are separated by treatment group with GD (dashed red line, n = 10 patients) and no-GD (solid blue line, n = 10 patients). In the 6 patients with a BSI, 5 BSIs occurred within the first 31 days; 1 patient in the no-GD arm had BSI on day +85 relative to the first transplant (on day +6 of the second transplant). P = 0.0483, Gray’s test.
Figure 6. Bacterial abundance in the gut…
Figure 6. Bacterial abundance in the gut around the time of BSI.
Multiple pathogens are present in the gut around the time of the BSI. Days relative to the course of the transplant shown on the x axis (from top to bottom on the y axis), antibiotic administration, α (Shannon) diversity, relative abundance of microbes in the stool samples at the genus taxonomic level (with organisms listed by color according to the key at the lower left), and relative abundance in the gut of the BSI-causing organism with the date of the BSI shown as an asterisk (*). Note: y axis is a different scale between abundance plots for focused organisms found in the BSI. (A) Patient C04 had 2 Staphylococcus aureus BSIs 89 days apart (day +5 and day +94) and a Klebsiella oxytoca BSI on day +18. (B) E. coli BSI on day +8 in patient C10. (C) Staphylococcus epidermidis BSI on day +23 in patient C20. (D) Patient C22 received 2 transplants and had low abundance of Rothia dentrocariosa in the gut during the first transplant; Rothia was not detectable in the gut after day +15 of the first transplant, with a Rothia BSI on day +6 of the second transplant (day +85 relative to the first transplant). An Enterococcus faecium BSI occurred on day +20 of the second transplant (day +99 relative to the first transplant). Antifungal and antiviral medications are shown in Supplemental Figure 13. Information on patients C03 and C11 may be found in Supplemental Figure 11. Azithro, azithromycin; Cipro, ciprofloxacin; Clinda, clindamycin; Levo, levofloxacin; Mero, meropenem; PipTazo, piperacillin/tazobactam; TMP/SMX, trimethoprim/sulfamethoxazole (cotrimoxazole).

References

    1. Storb R, et al. Graft-versus-host disease and survival in patients with aplastic anemia treated by marrow grafts from HLA-identical siblings. Beneficial effect of a protective environment. N Engl J Med. 1983;308(6):302–307. doi: 10.1056/NEJM198302103080602.
    1. Van Bekkum DW, et al. Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora. J Nat Cancer Inst. 1974;52(2):401–404. doi: 10.1093/jnci/52.2.401.
    1. Buckner CD, et al. Protective environment for marrow transplant recipients: a prospective study. Ann Intern Med. 1978;89(6):893–901. doi: 10.7326/0003-4819-89-6-893.
    1. Navari RM, et al. Prophylaxis of infection in patients with aplastic anemia receiving allogeneic marrow transplants. Am J Med. 1984;76(4):564–572. doi: 10.1016/0002-9343(84)90274-2.
    1. Vossen JM, et al. Complete suppression of the gut microbiome prevents acute graft-versus-host disease following allogeneic bone marrow transplantation. PLoS One. 2014;9(9):e105706. doi: 10.1371/journal.pone.0105706.
    1. Fredricks DN. The gut microbiota and graft-versus-host disease. J Clin Invest. 2019;129(5):1808–1817. doi: 10.1172/JCI125797.
    1. Whangbo J, et al. Antibiotic-mediated modification of the intestinal microbiome in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2017;52(2):183–190. doi: 10.1038/bmt.2016.206.
    1. Beelen DW, et al. Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host disease after marrow transplantation in patients with hematologic malignancies: final results and long-term follow-up of an open-label prospective randomized trial. Blood. 1999;93(10):3267–3275. doi: 10.1182/blood.V93.10.3267.410k22_3267_3275.
    1. Elgarten CW, et al. Broad-spectrum antibiotics and risk of graft-versus-host disease in pediatric patients undergoing transplantation for acute leukemia: association of carbapenem use with the risk of acute graft-versus-host disease. Transplant Cell Ther. 2021;27(2):177.e1–177.e8. doi: 10.1016/j.jtct.2020.10.012.
    1. Shono Y, et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci Transl Med. 2016;8(339):339ra71.
    1. Simms-Waldrip TR, et al. Antibiotic-induced depletion of anti-inflammatory clostridia is associated with the development of graft-versus-host disease in pediatric stem cell transplantation patients. Biol Blood Marrow Transplant. 2017;23(5):820–829. doi: 10.1016/j.bbmt.2017.02.004.
    1. Dandoy CE, et al. Incidence, risk factors, and outcomes of patients who develop mucosal barrier injury-laboratory confirmed bloodstream infections in the first 100 days after allogeneic hematopoietic stem cell transplant. JAMA Netw Open. 2020;3(1):e1918668. doi: 10.1001/jamanetworkopen.2019.18668.
    1. Dandoy CE, et al. Outcomes after bloodstream infection in hospitalized pediatric hematology/oncology and stem cell transplant patients. Pediatr Blood Cancer. 2019;66(12):e27978.
    1. Castagnola E, 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(4):339–347. doi: 10.1038/sj.bmt.1705921.
    1. Heston SM, et al. Microbiology of bloodstream infections in children after hematopoietic stem cell transplantation: a single-center experience over two decades (1997–2017) Open Forum Infect Dis. 2020;7(11):ofaa465. doi: 10.1093/ofid/ofaa465.
    1. Taur Y, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55(7):905–914. doi: 10.1093/cid/cis580.
    1. Goudie A, et al. Attributable cost and length of stay for central line-associated bloodstream infections. Pediatrics. 2014;133(6):e1525–e1532. doi: 10.1542/peds.2013-3795.
    1. Montassier E, et al. Pretreatment gut microbiome predicts chemotherapy-related bloodstream infection. Genome Med. 2016;8(1):49. doi: 10.1186/s13073-016-0301-4.
    1. Kelly MS, et al. Gut colonization preceding mucosal barrier injury bloodstream infection in pediatric hematopoietic stem cell transplant recipients. Biol Blood Marrow Transplant. 2019;25(11):2274–2280. doi: 10.1016/j.bbmt.2019.07.019.
    1. Andersen H, et al. Development of an infection risk index for microbiome targeted intervention in children at high-risk of multidrug-resistant bloodstream infections. Biol Blood Marrow Transplantat. 2019;25(suppl 3):S73–S74.
    1. Hakim H, et al. Gut microbiome composition predicts infection risk during chemotherapy in children with acute lymphoblastic leukemia. Clin Infect Dis. 2018;67(4):541–548. doi: 10.1093/cid/ciy153.
    1. Sleijfer DT, et al. Infection prevention in granulocytopenic patients by selective decontamination of the digestive tract. Eur J Cancer. 1980;16(6):859–869. doi: 10.1016/0014-2964(80)90140-1.
    1. Silvestri L, et al. Selective decontamination of the digestive tract: the mechanism of action is control of gut overgrowth. Intensive Care Med. 2012;38(11):1738–1750. doi: 10.1007/s00134-012-2690-1.
    1. Wittekamp BH, et al. Decontamination strategies and bloodstream infections with antibiotic-resistant microorganisms in ventilated patients: a randomized clinical trial. JAMA. 2018;320(20):2087–2098. doi: 10.1001/jama.2018.13765.
    1. Bekker V, et al. Dynamics of the gut microbiota in children receiving selective or total gut decontamination treatment during hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2019;25(6):1164–1171. doi: 10.1016/j.bbmt.2019.01.037.
    1. Tamburini FB, et al. Precision identification of diverse bloodstream pathogens in the gut microbiome. Nat Med. 2018;24(12):1809–1814. doi: 10.1038/s41591-018-0202-8.
    1. Pont S, et al. Bacterial behavior in human blood reveals complement evaders with some persister-like features. PLoS Pathog. 2020;16(12):e1008893. doi: 10.1371/journal.ppat.1008893.
    1. Sharon I, et al. Time series community genomics analysis reveals rapid shifts in bacterial species, strains, and phage during infant gut colonization. Genome Res. 2013;23(1):111–120. doi: 10.1101/gr.142315.112.
    1. Zhao S, et al. Adaptive evolution within gut microbiomes of healthy people. Cell Host Microbe. 2019;25(5):656–667. doi: 10.1016/j.chom.2019.03.007.
    1. Olm MR, et al. inStrain profiles population microdiversity from metagenomic data and sensitively detects shared microbial strains. Nat Biotechnol. 2021;39(6):727–736. doi: 10.1038/s41587-020-00797-0.
    1. Taur Y, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124(7):1174–1182. doi: 10.1182/blood-2014-02-554725.
    1. Wood DE, et al. Improved metagenomic analysis with Kraken 2. Genome Biol. 2019;20(1):257. doi: 10.1186/s13059-019-1891-0.
    1. Jenq RR, et al. Intestinal blautia is associated with reduced death from graft-versus-host disease. Biol Blood Marrow Transplant. 2015;21(8):1373–1383. doi: 10.1016/j.bbmt.2015.04.016.
    1. Konopinski MK. Shannon diversity index: a call to replace the original Shannon’s formula with unbiased estimator in the population genetics studies. PeerJ. 2020;8:e9391. doi: 10.7717/peerj.9391.
    1. Morjaria S, et al. Antibiotic-induced shifts in fecal microbiota density and composition during hematopoietic stem cell transplantation. Infect Immun. 2019;87(9):e00206-19.
    1. Zhai B, et al. High-resolution mycobiota analysis reveals dynamic intestinal translocation preceding invasive candidiasis. Nat Med. 2020;26(1):59–64. doi: 10.1038/s41591-019-0709-7.
    1. Siranosian BA, et al. Rare transmission of commensal and pathogenic bacteria in the gut microbiome of hospitalized adults. Nat Commun. 2022;13(1):586. doi: 10.1038/s41467-022-28048-7.
    1. CDC. Bloodstream Infection Event (Central Line-Associated Bloodstream Infection and Non-central Line Associated Bloodstream Infection). Accessed March 3, 2022.
    1. Gjaerde LK, et al. Gut decontamination during allogeneic hematopoietic stem cell transplantation and the risk of acute graft-versus-host disease. Bone Marrow Transplant. 2018;53(8):1061–1064. doi: 10.1038/s41409-018-0131-7.
    1. Li H, et al. Mucosal or systemic microbiota exposures shape the Bcell repertoire. Nature. 2020;584(7820):274–278. doi: 10.1038/s41586-020-2564-6.
    1. Lindner C, et al. Diversification of memory B cells drives the continuous adaptation of secretory antibodies to gut microbiota. Nat Immunol. 2015;16(8):880–888. doi: 10.1038/ni.3213.
    1. Lindner C, et al. Age, microbiota, and T cells shape diverse individual IgA repertoires in the intestine. J Exp Med. 2012;209(2):365–377. doi: 10.1084/jem.20111980.
    1. Wesemann DR, et al. Microbial colonization influences early B-lineage development in the gut lamina propria. Nature. 2013;501(7465):112–115. doi: 10.1038/nature12496.
    1. Daneman N, et al. Effect of selective decontamination on antimicrobial resistance in intensive care units: a systematic review and meta-analysis. Lancet Infect Dis. 2013;13(4):328–341. doi: 10.1016/S1473-3099(12)70322-5.
    1. Rommes H, et al, eds. Selective Decontamination of the Digestive Tract (SDD). Springer; 2021.
    1. Gluckman E, et al. Prophylaxis of bacterial infections after bone marrow transplantation. A randomized prospective study comparing oral broad-spectrum nonabsorbable antibiotics (vancomycin-tobramycin-colistin) to absorbable antibiotics (ofloxacin-amoxicillin) Chemotherapy. 1991;37(suppl 1):33–38.
    1. Kimura S-i, et al. Antibiotic prophylaxis in hematopoietic stem cell transplantation. A meta-analysis of randomized controlled trials. J Infect. 2014;69(1):13–25. doi: 10.1016/j.jinf.2014.02.013.
    1. Alexander S, et al. Effect of levofloxacin prophylaxis on bacteremia in children with acute leukemia or undergoing hematopoietic stem cell transplantation: a randomized clinical trial. JAMA. 2018;320(10):995–1004. doi: 10.1001/jama.2018.12512.
    1. Dandoy CE, Alonso PB. MBI-LCBI and CLABSI: more than scrubbing the line. Bone Marrow Transplant. 2019;54(12):1932–1939. doi: 10.1038/s41409-019-0489-1.
    1. Epstein L, et al. Mucosal barrier injury laboratory-confirmed bloodstream infections (MBI-LCBI): descriptive analysis of data reported to National Healthcare Safety Network (NHSN), 2013. Infect Control Hosp Epidemiol. 2016;37(1):2–7. doi: 10.1017/ice.2015.245.
    1. Metzger KE, et al. The burden of mucosal barrier injury laboratory-confirmed bloodstream infection among hematology, oncology, and stem cell transplant patients. Infect Control Hosp Epidemiol. 2015;36(2):119–124. doi: 10.1017/ice.2014.38.
    1. Stoma I, et al. Compositional flux within the intestinal microbiota and risk for bloodstream infection with gram-negative bacteria. Clin Infect Dis. 2021;73(11):e4627–e4635. doi: 10.1093/cid/ciaa068.
    1. Severyn CJ, et al. Microbiota modification in hematology: still at the bench or ready for the bedside? Blood Adv. 2019;3(21):3461–3472. doi: 10.1182/bloodadvances.2019000365.
    1. Alho AC, et al. Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. Blood. 2016;127(5):646–657. doi: 10.1182/blood-2015-10-672345.
    1. Humphries RM, et al. Multicenter evaluation of colistin broth disk elution and colistin agar test: a report from the clinical and laboratory standards institute. J Clin Microbiol. 2019;57(11):e01269–19.
    1. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing. Accessed March 3, 2022.
    1. European Committee on Antimicrobial Susptibility Testing. Colistin: rationale for the EUCAST clinical breakpoints, version 1.0. Accessed March 3, 2022.
    1. Illumina. Effects of Index Misassignment on Multiplexing and Downstream Analysis. Accessed March 3, 2022.
    1. Siranosian B, et al. bhattlab/bhattlab_workflows: v1.0.1. Accessed March 3, 2022.
    1. Petersen KR, et al. Super deduper, fast PCR duplicate detection in fastq files. Paper presented at: Proceedings of the 6th ACM Conference on Bioinformatics, Computational Biology and Health Informatics; September 9–12, 2015; Atlanta, Georgia, USA. Accessed March 3, 2022.
    1. Babraham Bioinformatics. Trim Galore! Accessed March 3, 2022.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26(5):589–595. doi: 10.1093/bioinformatics/btp698.
    1. Babraham Bioinformatics. FastQC. Accessed March 3, 2022.
    1. Koster J, Rahmann S. Snakemake-a scalable bioinformatics workflow engine. Bioinformatics. 2018;34(20):3600. doi: 10.1093/bioinformatics/bty350.
    1. Lu J BF, et al. Bracken: estimating species abundance in metagenomics data. PeerJ Comput Sci. 2017;3:e104. doi: 10.7717/peerj-cs.104.
    1. Siranosian B, Moss E. bhattlab/kraken2_classification: a mostly finished pipleline. Accessed March 3, 2022.
    1. Oksanen J, et al. Ordination methods, diversity analysis and other funtions for community and vegetation ecologists. Version 2.5-7. Accessed March 3, 2022.
    1. Nurk S, et al. metaSPAdes: a new versatile metagenomic assembler. Genome Res. 2017;27(5):824–834. doi: 10.1101/gr.213959.116.
    1. Alneberg J, et al. Binning metagenomic contigs by coverage and composition. Nat Methods. 2014;11(11):1144–1146. doi: 10.1038/nmeth.3103.
    1. Kang DD, et al. MetaBAT 2: an adaptive binning algorithm for robust and efficient genome reconstruction from metagenome assemblies. PeerJ. 2019;7:e7359. doi: 10.7717/peerj.7359.
    1. Wu Y-W, et al. MaxBin 2.0: an automated binning algorithm to recover genomes from multiple metagenomic datasets. Bioinformatics. 2015;32(4):605–607.
    1. Sieber CMK, et al. Recovery of genomes from metagenomes via a dereplication, aggregation and scoring strategy. Nat Microbiol. 2018;3(7):836–843. doi: 10.1038/s41564-018-0171-1.
    1. Olm MR, et al. dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication. ISME J. 2017;11(12):2864–2868. doi: 10.1038/ismej.2017.126.
    1. Gurevich A, et al. QUAST: quality assessment tool for genome assemblies. Bioinformatics. 2013;29(8):1072–1075. doi: 10.1093/bioinformatics/btt086.
    1. Bowers RM, et al. Minimum information about a single amplified genome (MISAG) and a metagenome-assembled genome (MIMAG) of bacteria and archaea. Nat Biotechnol. 2017;35(8):725–231. doi: 10.1038/nbt.3893.
    1. Nayfach S, et al. New insights from uncultivated genomes of the global human gut microbiome. Nature. 2019;568(7753):505–510. doi: 10.1038/s41586-019-1058-x.
    1. Alcock BP, et al. CARD 2020: antibiotic resistome surveillance with the comprehensive antibiotic resistance database. Nucleic Acids Res. 2020;48(d1):D517–D525.
    1. Wickham H, ed. ggplot2: Elegant Graphics for Data Analysis. Springer; 2016.
    1. Kang JB, et al. Intestinal microbiota domination under extreme selective pressures characterized by metagenomic read cloud sequencing and assembly. BMC Bioinformatics. 2019;20(16):585.

Source: PubMed

3
Subscribe