Mortality and microbial diversity after allogeneic hematopoietic stem cell transplantation: secondary analysis of a randomized nutritional intervention trial

Kristin J Skaarud, Johannes R Hov, Simen H Hansen, Martin Kummen, Jørgen Valeur, Ingebjørg Seljeflot, Asta Bye, Vemund Paulsen, Knut E A Lundin, Marius Trøseid, Geir E Tjønnfjord, Per Ole Iversen, Kristin J Skaarud, Johannes R Hov, Simen H Hansen, Martin Kummen, Jørgen Valeur, Ingebjørg Seljeflot, Asta Bye, Vemund Paulsen, Knut E A Lundin, Marius Trøseid, Geir E Tjønnfjord, Per Ole Iversen

Abstract

Gut mucosal barrier injury is common following allogeneic hematopoietic stem cell transplantation (allo-HSCT) and associated with poor clinical outcomes. Diet is critical for microbial diversity, but whether nutritional support affects microbiota and outcome after allo-HSCT is unknown. We present a secondary analysis of a randomized controlled nutritional intervention trial during allo-HSCT. We investigated if the intervention influenced gut microbiota, short-chain fatty acids (SCFAs), and markers of gut barrier functions, and if these parameters were associated with clinical outcomes. Fecal specimens were available from 47 recipients, and subjected to 16S rRNA gene sequencing. We found no significant differences between the intervention group and controls in investigated parameters. We observed a major depletion of microbiota, SCFAs, and altered markers of gut barrier function from baseline to 3 weeks post-transplant. One-year mortality was significantly higher in patients with lower diversity at 3 weeks post-HSCT, but not related to diversity at baseline. The relative abundance of Blautia genus at 3 weeks was higher in survivors. Fecal propionic acid was associated with survival. Markers of gut barrier functions were less strongly associated with clinical outcomes. Possibly, other strategies than dietary intervention are needed to prevent negative effects of gut microbiota and clinical outcomes after allo-HSCT.ClinicalTrials.gov (NCT01181076).

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Gut microbial diversity and the intervention and control group at baseline and 3 weeks. (a) Observed operational taxonomic units (OTUs) and groups, (b) Shannon diversity index and groups. Data are observed operational taxonomic units (OTUs) and Shannon diversity index given as individual values (dots) and box plots (median and interquartile range) with minimum and maximum values.
Figure 2
Figure 2
Fecal total SCFAs and the intervention and control group at baseline and 3 weeks. Data are total SCFAs given as individual values (dots) and box plots (median and interquartile range) with minimum and maximum values.
Figure 3
Figure 3
Markers of gut barrier functions and the intervention and control group at baseline and 3 weeks. (a) Intestinal fatty acid binding protein (I-FABP) and groups, (b) lipopolysaccharide binding protein (LBP) and groups. Data are I-FABP and LBP given as individual values (dots) and box plots (median and interquartile range) with minimum and maximum values.
Figure 4
Figure 4
Gut microbiota diversity and one-year survival. (a) Observed operational taxonomic units (OTUs) at baseline and 3 weeks and one-year survival, (b) Shannon diversity index at baseline and 3 weeks and one-year survival, (c) Loss of observed OTUs from baseline to 3 weeks and one-year survival. Data are observed OTUs and Shannon diversity index, and given as individual values (dots) and box plots (median and interquartile range) with minimum and maximum values.
Figure 5
Figure 5
Blautia abundance and one-year survival. (a) Blautia abundance at 3 weeks and one-year survival, (b) and (c) Drop in Blautia abundance from baseline to 3 weeks and one-year survival. Data are Blautia abundance given as individual values (dots) and box plots (median and interquartile range) with minimum and maximum values. Lines connect individual data points.

References

    1. Shallis RM, Terry CM, Lim SH. Changes in intestinal microbiota and their effects on allogeneic stem cell transplantation. Am. J. Hematol. 2018;93:122–128. doi: 10.1002/ajh.24896.
    1. Rashidi A, et al. Pretransplant serum citrulline predicts acute graft-versus-host disease. Biol. Blood Marrow Transplant. 2018;24:2190–2196. doi: 10.1016/j.bbmt.2018.06.036.
    1. Ye H, Lv M, Zhao X, Zhao X, Huang X. Plasma level of lipopolysaccharide-binding protein is indicative of acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Int. J. Hematol. 2012;95:680–688. doi: 10.1007/s12185-012-1076-z.
    1. Derikx JP, et al. Loss of enterocyte mass is accompanied by diminished turnover of enterocytes after myeloablative therapy in haematopoietic stem-cell transplant recipients. Ann. Oncol. 2009;20:337–342. doi: 10.1093/annonc/mdn579.
    1. Peled JU, et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 2020;382:822–834. doi: 10.1056/NEJMoa1900623.
    1. Rafei H, Jenq RR. Microbiome-intestine cross talk during acute graft-versus-host disease. Blood. 2020;136:401–409. doi: 10.1182/blood.2019000950.
    1. Jenq RR, et al. Intestinal blautia is associated with reduced death from graft-versus-host disease. Biol. Blood Marrow Transplant. 2015;21:1373–1383. doi: 10.1016/j.bbmt.2015.04.016.
    1. Stein-Thoeringer CK, et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science. 2019;366:1143–1149. doi: 10.1126/science.aax3760.
    1. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi: 10.1038/nature12820.
    1. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019;16:461–478. doi: 10.1038/s41575-019-0157-3.
    1. Romick-Rosendale LE, et al. Antibiotic exposure and reduced short chain fatty acid production after hematopoietic stem cell transplant. Biol. Blood Marrow Transplant. 2018;24:2418–2424. doi: 10.1016/j.bbmt.2018.07.030.
    1. Mathewson ND, et al. Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat. Immunol. 2016;17:505–513. doi: 10.1038/ni.3400.
    1. Fujiwara H, et al. Microbial metabolite sensor GPR43 controls severity of experimental GVHD. Nat. Commun. 2018;9:3674. doi: 10.1038/s41467-018-06048-w.
    1. Markey KA, et al. The microbe-derived short-chain fatty acids butyrate and propionate are associated with protection from chronic GVHD. Blood. 2020;136:130–136. doi: 10.1182/blood.2019003369.
    1. Peric Z, et al. Variability of nutritional practices in peritransplant period after allogeneic hematopoietic stem cell transplantation: A survey by the Complications and Quality of Life Working Party of the EBMT. Bone Marrow Transplant. 2018 doi: 10.1038/s41409-018-0137-1.
    1. Moody, K. J. B. o. B. & Transplantation, M. Neutropenic dietary restrictions for hematopoietic stem cell patients: time for a change. 25, e223–e225 (2019).
    1. D'Amico F, et al. Enteral nutrition in pediatric patients undergoing hematopoietic SCT promotes the recovery of gut microbiome homeostasis. Nutrients. 2019;11:2958. doi: 10.3390/nu11122958.
    1. Skaarud KJ, et al. Effects of individualized nutrition after allogeneic hematopoietic stem cell transplantation following myeloablative conditioning; A randomized controlled trial. Clin. Nutr. ESPEN. 2018;28:59–66. doi: 10.1016/j.clnesp.2018.08.002.
    1. Skaarud KJ, et al. Body weight, body composition and survival after 1 year: Follow-up of a nutritional intervention trial in allo-HSCT recipients. Bone Marrow Transplant. 2019 doi: 10.1038/s41409-019-0638-6.
    1. Torfoss D, et al. Benzylpenicillin plus an aminoglycoside versus meropenem in neutropenic lymphoma and leukaemia patients with a suspected bacterial infection: a randomized, controlled trial. Clin. Microbiol. Infect. 2017;23:179–187. doi: 10.1016/j.cmi.2016.10.019.
    1. Przepiorka D, et al. 1994 consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15:825–828.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: Improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–596. doi: 10.1093/nar/gks1219.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Valeur J, et al. Fecal fermentation in irritable bowel syndrome: Influence of dietary restriction of fermentable oligosaccharides, disaccharides. Monosaccharides Polyols. Digest. 2016;94:50–56. doi: 10.1159/000448280.
    1. Primec M, Mičetić-Turk D, Langerholc T. Analysis of short-chain fatty acids in human feces: A scoping review. Anal. Biochem. 2017;526:9–21. doi: 10.1016/j.ab.2017.03.007.
    1. Høverstad T, Fausa O, Bjørneklett A, Bøhmer T. Short-chain fatty acids in the normal human feces. Scand. J. Gastroenterol. 1984;19:375–381. doi: 10.1080/00365521.1984.12005738.
    1. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, Vienna, Austria. v. 3.6.1, 2020).
    1. Seguy D, et al. Better outcome of patients undergoing enteral tube feeding after myeloablative conditioning for allogeneic stem cell transplantation. Transplantation. 2012;94:287–294. doi: 10.1097/TP.0b013e3182558f60.
    1. Mattsson J, Westin S, Edlund S, Remberger M. Poor oral nutrition after allogeneic stem cell transplantation correlates significantly with severe graft-versus-host disease. Bone Marrow Transplant. 2006;38:629–633. doi: 10.1038/sj.bmt.1705493.
    1. Sefcick A, Anderton D, Byrne JL, Teahon K, Russell NH. Naso-jejunal feeding in allogeneic bone marrow transplant recipients: Results of a pilot study. Bone Marrow Transplant. 2001;28:1135–1139. doi: 10.1038/sj.bmt.1703301.
    1. Seguy D, et al. Enteral feeding and early outcomes of patients undergoing allogeneic stem cell transplantation following myeloablative conditioning. Transplantation. 2006;82:835–839. doi: 10.1097/01.tp.0000229419.73428.ff.
    1. Andersen S, et al. Pilot study investigating the effect of enteral and parenteral nutrition on the gastrointestinal microbiome post-allogeneic transplantation. Br. J. Haematol. 2019 doi: 10.1111/bjh.16218.
    1. Iyama S, et al. Efficacy of enteral supplementation enriched with glutamine, fiber, and oligosaccharide on mucosal injury following hematopoietic stem cell transplantation. Case Rep. Oncol. 2014;7:692–699. doi: 10.1159/000368714.
    1. Taur Y, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–1182. doi: 10.1182/blood-2014-02-554725.
    1. Holler E, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: Loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transplant. 2014;20:640–645. doi: 10.1016/j.bbmt.2014.01.030.
    1. Kusakabe S, et al. Pre- and post-serial metagenomic analysis of gut microbiota as a prognostic factor in patients undergoing haematopoietic stem cell transplantation. Br. J. Haematol. 2020;188:438–449. doi: 10.1111/bjh.16205.
    1. Carreras E, Diaz-Ricart M. The role of the endothelium in the short-term complications of hematopoietic SCT. Bone Marrow Transplant. 2011;46:1495–1502. doi: 10.1038/bmt.2011.65.

Source: PubMed

3
Sottoscrivi