Microbiota Changes during Allogeneic Stem Cell Transplantation: Comparison
Please note this is a comparison between Version 2 by Sirius Huang and Version 1 by Elisabetta Metafuni.

Microbiota changes during allogeneic hematopoietic stem cell transplantation has several known causes: conditioning chemotherapy and radiation, broad-spectrum antibiotic administration, modification in nutrition status and diet, and graft-versus-host disease.

  • fecal microbiota transplantation
  • graft-versus-host disease
  • allogeneic hematopoietic stem cell transplantation

1. Introduction

The intestine is the main site of bacterial, viral, and fungal colonization. All these microorganisms do not merely act as commensal organisms, but actively participate in the digestion of complex carbohydrates and interact with the host immune system in a manner that we still do not fully understand. A healthy bacterial microbiota is involved in heterogeneous activities: development and maturity of the host immune system, digestion of food, synthesis of essential amino acids, short-chain fatty acids (SCFAs), and vitamins, regulation of the immune response, and enhancement of the resistance to pathogenic infection [1]. The vast majority of the bacteria belong to the Bacteroidetes and Firmicutes phyla; these bacteria are in equilibrium with the host’s innate immune system and help to maintain homeostasis, which directly affects host health when altered [2]. The gut microbiota of each individual contains many unique strains not found in others, and inter-individual differences in microbiota composition are much larger than intra-individual variations [3].
It has been known since the 1970s that the microbiota is involved in graft-versus-host disease (GvHD) pathogenesis, as demonstrated by the van Bekkum group in 1974, where mice raised in a germ-free environment did not develop gastrointestinal (GI) GvHD. Just recently, researchers have achieved a partial understanding of the panorama and the intricacies of the microbiota and GvHD [4,5,6][4][5][6]. The microbiota-derived signals can indirectly influence regulatory T lymphocytes (Tregs) by activating innate, gut-resident, antigen-presenting cells (APCs) known as CD103 + CD11b + dendritic cells (DCs) [7]. These DCs subsequently promote adaptive anti-inflammatory responses of Treg cells by producing molecules such as transforming growth factor β (TGF-β) and the vitamin A metabolite, retinoic acid. Tregs are not only responsible for maintaining immunological tolerance in tissues but also actively contribute to tissue repair through the production of amphiregulin [8]. This mechanism is important for homeostasis and modulates the immune response against the gut flora. An impaired mechanism can lead to a pro-inflammatory state that can be common in inflammatory bowel diseases, non-alcoholic steatohepatitis, type 2 diabetes, obesity, and, with some peculiarities, in acute GvHD (aGvHD) [9,10][9][10].
To recognize the complexity of the microbiota, it is essential to acknowledge that any factors disrupting or weakening this system can predispose to infections and autoimmunity. In the setting of allogeneic hematopoietic stem cell transplantation (HSCT), damage to the microbiota is not solely due to the conditioning regimen but also to the preceding chemotherapy and antibiotic treatments, which consistently undermine the microbiota [11,12][11][12].

2. Microbiota Changes during Allogeneic Stem Cell Transplantation

Significant changes were reported in microbiota composition during the HSCT procedure, both in terms of diversity and in terms of taxonomy [13]. These changes might contribute to post-transplant outcomes, such as GvHD incidence, transplant-related mortality (TRM), infectious complications, and overall survival (OS). A reduced intestinal microbial diversity after HSCT was reported to affect survival outcomes [14], while a low microbial diversity at engraftment was significantly associated with a high risk of TRM and severely reduced OS [15,16][15][16]. Predominant genera in samples obtained from patients with reduced microbial diversity were Enterococcus, Streptococcus, Enterobacteriaceae, and Lactobacillus [4,15][4][15]. Patients who died showed an abundance of Proteobacteria including Enterobacteriaceae, while surviving patients showed an abundance of Lachnospiraceae and Actinomycetaceae [15].

2.1. Antibiotics Affect the Gut Microbiota

A patient candidate for HSCT has already received several courses of broad-spectrum antibiotics. These therapeutic interventions have profoundly influenced the composition of the patient’s gut microbiota [17]. Notably, the most significant impact on the microbiota arises from the antimicrobial regimens administered both prior to and after transplantation. Different antibiotic classes exert distinct effects on the gut microbiota, which largely depend on the activity spectrum of the drug and may promote a different proinflammatory pattern in the gut flora. Particularly, the use of broad-spectrum antibiotics leads to a reduction in microbiota diversity and richness.
It has been demonstrated in mice that treatment with levofloxacin and cefepime had selected a gut flora with high abundances of Clostridia compared with meropenem-treated mice, and this difference was consistent with fecal butyrate levels. Additionally, both levofloxacin- and cefepime-treated mice had significantly lower abundances of Bacteroides thetaiotaomicron, compared with meropenem-treated mice. Notably, meropenem usage has been associated with the proliferation of Bacteroides thetaiotaomicron, a Gram-negative obligate anaerobe, that exhibits the capacity to metabolize dietary polysaccharides and host-derived glycans, including mucin. Therefore, Clostridia regulates intestinal immunity through SCFA production, while Bacteroides thetaiotaomicron is detrimental in determining mucus integrity. This observation provides a plausible explanation for the correlation between the utilization of carbapenems, frequently employed in the context of pre-engraftment febrile neutropenia, and the heightened severity of GI GvHD [18].
Different antibiotics used in the HSCT setting might impact GvHD-related mortality, probably due to the modification induced by antibiotics in microbiota composition [4]. A study conducted by Shono et al. on 857 patients who underwent T-cell-replete HSCT showed that among the twelve most frequently used antibiotics, piperacillin-tazobactam and imipenem-cilastatin were associated with different GvHD-related mortality rates. Additionally, these antibiotics were linked to an increased incidence of grade II–IV aGvHD with a higher occurrence of upper GI GvHD. Conversely, exposure to aztreonam or cefepime correlated with reduced GvHD-related mortality. Microbial composition in subjects and mice treated with imipenem-cilastatin showed a decreased presence of Clostridiales, which is believed to regulate anti-inflammatory processes in the gastrointestinal tract, inducing Tregs via SCFAs metabolites [19]. The use of antibiotic prophylaxes like ciprofloxacin and broad-spectrum systemic antibiotics reduced commensal bacteria, favoring the overgrowth of Enterococci [20]. Antibiotic therapy is not only used for infections but is also employed as a gut decontamination strategy. Weber et al. compared two decontamination schedules: ciprofloxacin 500 mg twice a day and metronidazole 500 mg three times a day, starting 8 days before HSCT until 14 days post-engraftment (n = 200) and rifaximin 200 mg twice a day (n = 194). Results indicated that gut decontamination with rifaximin was associated with similar rates of infectious complications compared with ciprofloxacin/metronidazole but preserved a high intestinal microbiota diversity and mitigated the negative effects of systemic antibiotics on microbial composition [21]. Previously hospitalized patients compared with de novo admitted exhibited a reduced expression of predominant commensal strains together with an increased presence of Enterococci [20]. The selection of antibiotics with a narrower spectrum of activity, particularly targeting anaerobic bacteria, may mitigate intestinal GVHD by minimizing the extent of microbiota alterations [22]. The results are presented Table 1.
Table 1.
Microbiota changes due to conditioning regimen.

2.2. Conditioning Affects the Gut Microbiota

By the time a patient arrives at the time of transplantation, he/she has already received varying amounts of chemotherapy and has been exposed to several antibiotics in most cases [23]. Therefore, the patient’s intestinal microbiota has been injured, displays a reduced α-diversity, and is going to receive a conditioning regimen with or without radiotherapy. This means that on a weakened flora susceptible to damage, further depletion will occur. Although we know that chemotherapy impairs intestinal microbiota, the mechanisms by which this occurs are still not completely clear [23,24][23][24].
Data about changes in the microbiota composition after HSCT are sometimes conflicting. Holler et al. compared pre- and post-HSCT fecal microbial composition finding an increase in Enterococci [20,25][20][25] and a reduction in Firmicutes and other phyla for all patients [20,26][20][26]. Recently, Kouidhi et al. reported a comparison between HSCT patients and healthy controls. At the phylum level, Actinobacteria were more represented in the control group compared with Proteobacteria and Verrucomicrobia in the HSCT group. At the genus level, patients in the HSCT group showed a lower abundance of Faecalibacterium, Alistipes, and Prevotella 9 and a higher abundance of Bacteroides, Escherichia/Shigella, Klebsiella, and Akkermansia [27]. It is well established that a conditioning regimen involving radioactive sources can lead to dysbiosis of the microbiota, and that radiation-induced enteritis is exacerbated by this dysbiosis. This understanding is derived not only from direct observations of microbiota damage in total body irradiation (TBI) regimens but also from extensive experience in oncology and the widespread use of radiotherapy [28].
In radiation enteritis, as in chemotherapy regimens, an increased abundance of bacteria belonging to the Actinobacteria and Proteobacteria phyla is reported. These bacteria are conditional pathogens such as Escherichia coli. Conversely, microorganisms from the Firmicutes and Bacteroides phyla are reduced by radiations [29]. In a recent paper, Gu and colleagues reported microbiota changes during myeloablative transplantation, including rabbit thymoglobulin administration. Microbiota diversity began to decline from the start of the conditioning regimen and continued to diminish over the course of HSCT until day 12 after HSCT, where diversity reached the lowest value. After that, diversity gradually increased over time. Intestinal domination varied from beneficial genus Bacteroides before conditioning to pathogenic genera such as Enterococcus, Klebsiella, and Escherichia during the engraftment phase [16].
The reduction in microbiota diversity described early after HSCT is a byproduct of conditioning regimens and restoration of pre-HSCT levels is possible over time in the absence of GvHD or other modifying events [27,30][27][30]. A reduced microbial diversity after HSCT was associated with high GvHD lethality [14], while the persistence of high microbiota diversity after HSCT was associated with a lower risk of death and TRM, without an increased relapse rate [4]. Results are resumed in Table 2.
Table 2.
Microbiota changes due to conditioning regimen.

2.3. Diet Affects Microbiota

The role of diet and nutrition in HSCT patients is often underappreciated, but growing evidence suggests a link between the diet, the microbiota, and clinical outcomes. Although we do not yet know exactly how this element can be used to deliberately select a tolerogenic microbiota, some evidence suggests its potential role in the HSCT setting. It has been demonstrated that enteral nutrition, compared to parenteral nutrition, is protective against the development of GvHD and reduces TRM while increasing OS [31,32][31][32].
In this regard, it is even more intriguing evidence provided by Khuat et al., who demonstrated in preclinical models and clinical trials that obesity has a negative effect on HSCT outcomes in both mice and humans. Obesity is specifically associated with an increased risk of aGVHD with GI involvement. This effect appeared restricted to the gut and relies on increased production of pro-inflammatory cytokines by donor CD4+ T-cells. In the murine model, the pre-transplant diet and consequently the selected microbiota can influence TRM, often related to GI aGvHD. Indeed, it has been observed that mice with diet-induced obesity (DIO) exhibited increased gut permeability and translocation of endotoxins across the gut barrier, along with reduced diversity in their gut microbiota. After HSCT, these changes in DIO mice promoted aGvHD and led to a more severe clinical presentation [33].

References

  1. Nagpal, R.; Wang, S.; Ahmadi, S.; Hayes, J.; Gagliano, J.; Subashchandrabose, S.; Kitzman, D.W.; Becton, T.; Read, R.; Yadav, H. Human-origin probiotic cocktail increases short-chain fatty acid production via modulation of mice and human gut microbiome. Sci. Rep. 2018, 8, 12649.
  2. Ramirez, J.; Guarner, F.; Fernandez, L.B.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912.
  3. Allaband, C.; McDonald, D.; Vázquez-Baeza, Y.; Minich, J.J.; Tripathi, A.; Brenner, D.A.; Loomba, R.; Smarr, L.; Sandborn, W.J.; Schnabl, B.; et al. Microbiome 101: Studying, Analyzing, and Interpreting Gut Microbiome Data for Clinicians. Clin. Gastroenterol. Hepatol. 2019, 17, 218–230.
  4. Peled, J.U.; Gomes, A.L.; Devlin, S.M.; Littmann, E.R.; Taur, Y.; Sung, A.D.; Weber, D.; Hashimoto, D.; Slingerland, A.E.; Slingerland, J.B.; et al. Microbiota as Predictor of Mortality in Allogeneic Hematopoietic-Cell Transplantation. N. Engl. J. Med. 2020, 382, 822–834.
  5. Chi, M.; Jiang, T.; He, X.; Peng, H.; Li, Y.; Zhang, J.; Wang, L.; Nian, Q.; Ma, K.; Liu, C. Role of Gut Microbiota and Oxidative Stress in the Progression of Transplant-Related Complications following Hematopoietic Stem Cell Transplantation. Oxidative Med. Cell. Longev. 2023, 2023, 3532756.
  6. Zeiser, R.; Warnatz, K.; Rosshart, S.; Sagar; Tanriver, Y. GVHD, IBD, and primary immunodeficiencies: The gut as a target of immunopathology resulting from impaired immunity. Eur. J. Immunol. 2022, 52, 1406–1418.
  7. Köhler, N.; Zeiser, R. Intestinal Microbiota Influence Immune Tolerance Post Allogeneic Hematopoietic Cell Transplantation and Intestinal GVHD. Front. Immunol. 2019, 9, 3179.
  8. Gao, B.; Xiang, X. Interleukin-22 from bench to bedside: A promising drug for epithelial repair. Cell. Mol. Immunol. 2019, 16, 666–667.
  9. Duvallet, C.; Gibbons, S.M.; Gurry, T.; Irizarry, R.A.; Alm, E.J. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 2017, 8, 1784.
  10. Mohty, M.; Malard, F. IL-22, a new beacon in gastrointestinal aGVHD. Blood 2023, 141, 1369–1370.
  11. Henig, I.; Yehudai-Ofir, D.; Zuckerman, T. The clinical role of the gut microbiome and fecal microbiota transplantation in allogeneic stem cell transplantation. Haematologica 2021, 106, 933–946.
  12. D’angelo, C.R.; Sudakaran, S.; Callander, N.S. Clinical effects and applications of the gut microbiome in hematologic malignancies. Cancer 2021, 127, 679–687.
  13. Chong, P.P.; Koh, A.Y. The gut microbiota in transplant patients. Blood Rev. 2020, 39, 100614.
  14. Jenq, R.R.; Taur, Y.; Devlin, S.M.; Ponce, D.M.; Goldberg, J.D.; Ahr, K.F.; Littmann, E.R.; Ling, L.; Gobourne, A.C.; Miller, L.C.; et al. Intestinal Blautia Is Associated with Reduced Death from Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2015, 21, 1373–1383.
  15. Taur, Y.; Jenq, R.R.; Perales, M.-A.; Littmann, E.R.; Morjaria, S.; Ling, L.; No, D.; Gobourne, A.; Viale, A.; Dahi, P.B.; et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014, 124, 1174–1182.
  16. Gu, Z.; Xiong, Q.; Wang, L.; Wang, L.; Li, F.; Hou, C.; Dou, L.; Zhu, B.; Liu, D. The impact of intestinal microbiota in antithymocyte globulin–based myeloablative allogeneic hematopoietic cell transplantation. Cancer 2022, 128, 1402–1410.
  17. Shallis, R.M.; Terry, C.M.; Lim, S.H. Changes in intestinal microbiota and their effects on allogeneic stem cell transplantation. Am. J. Hematol. 2018, 93, 122–128.
  18. Hayase, E.; Hayase, T.; Jamal, M.A.; Miyama, T.; Chang, C.-C.; Ortega, M.R.; Ahmed, S.S.; Karmouch, J.L.; Sanchez, C.A.; Brown, A.N.; et al. Mucus-degrading Bacteroides link carbapenems to aggravated graft-versus-host disease. Cell 2022, 185, 3705–3719.e14.
  19. Shono, Y.; Docampo, M.D.; Peled, J.U.; Perobelli, S.M.; Velardi, E.; Tsai, J.J.; Slingerland, A.E.; Smith, O.M.; Young, L.F.; Gupta, J.; 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, 339ra71.
  20. Holler, E.; Butzhammer, P.; Schmid, K.; Hundsrucker, C.; Koestler, J.; Peter, K.; Zhu, W.; Sporrer, D.; Hehlgans, T.; Kreutz, M.; 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.
  21. Weber, D.; Oefner, P.J.; Dettmer, K.; Hiergeist, A.; Koestler, J.; Gessner, A.; Weber, M.; Stämmler, F.; Hahn, J.; Wolff, D.; et al. Rifaximin preserves intestinal microbiota balance in patients undergoing allogeneic stem cell transplantation. Bone Marrow Transplant. 2016, 51, 1087–1092.
  22. Lee, S.-E.; Lim, J.-Y.; Ryu, D.-B.; Kim, T.W.; Park, S.S.; Jeon, Y.-W.; Yoon, J.-H.; Cho, B.-S.; Eom, K.-S.; Kim, Y.-J.; et al. Alteration of the Intestinal Microbiota by Broad-Spectrum Antibiotic Use Correlates with the Occurrence of Intestinal Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2019, 25, 1933–1943.
  23. Montassier, E.; Batard, E.; Massart, S.; Gastinne, T.; Carton, T.; Caillon, J.; Le Fresne, S.; Caroff, N.; Hardouin, J.B.; Moreau, P.; et al. 16S rRNA Gene Pyrosequencing Reveals Shift in Patient Faecal Microbiota During High-Dose Chemotherapy as Conditioning Regimen for Bone Marrow Transplantation. Microb. Ecol. 2014, 67, 690–699.
  24. DeFilipp, Z.; Hohmann, E.; Jenq, R.R.; Chen, Y.-B. Fecal Microbiota Transplantation: Restoring the Injured Microbiome after Allogeneic Hematopoietic Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, e17–e22.
  25. Stein-Thoeringer, C.K.; Nichols, K.B.; Lazrak, A.; Docampo, M.D.; Slingerland, A.E.; Slingerland, J.B.; Clurman, A.G.; Armijo, G.; Gomes, A.L.C.; Shono, Y.; et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science 2019, 366, 1143–1149.
  26. Chiusolo, P.; Metafuni, E.; Sterbini, F.P.; Giammarco, S.; Masucci, L.; Leone, G.; Sica, S. Gut Microbiome Changes after Stem Cell Transplantation. Blood 2015, 126, 1953.
  27. Kouidhi, S.; Souai, N.; Zidi, O.; Mosbah, A.; Lakhal, A.; Ben Othmane, T.; Belloumi, D.; Ben Ayed, F.; Asimakis, E.; Stathopoulou, P.; et al. High Throughput Analysis Reveals Changes in Gut Microbiota and Specific Fecal Metabolomic Signature in Hematopoietic Stem Cell Transplant Patients. Microorganisms 2021, 9, 1845.
  28. Crawford, P.A.; Gordon, J.I. Microbial regulation of intestinal radiosensitivity. Proc. Natl. Acad. Sci. USA 2005, 102, 13254–13259.
  29. Jian, Y.; Zhang, D.; Liu, M.; Wang, Y.; Xu, Z.-X. The Impact of Gut Microbiota on Radiation-Induced Enteritis. Front. Cell. Infect. Microbiol. 2021, 11, 586392.
  30. Shono, Y.; Brink, M.R.M.v.D. Gut microbiota injury in allogeneic haematopoietic stem cell transplantation. Nat. Rev. Cancer 2018, 18, 283–295.
  31. Beckman, M.F.; Morton, D.S.; Mougeot, F.B.; Mougeot, J.-L.C. Allogenic stem cell transplant-associated acute graft versus host disease: A computational drug discovery text mining approach using oral and gut microbiome signatures. Support. Care Cancer 2021, 29, 1765–1779.
  32. Beckerson, J.; Szydlo, R.M.; Hickson, M.; Mactier, C.E.; Innes, A.J.; Gabriel, I.H.; Palanicawandar, R.; Kanfer, E.J.; Macdonald, D.H.; Milojkovic, D.; et al. Impact of route and adequacy of nutritional intake on outcomes of allogeneic haematopoietic cell transplantation for haematologic malignancies. Clin. Nutr. 2019, 38, 738–744.
  33. Khuat, L.T.; Le, C.T.; Pai, C.-C.S.; Shields-Cutler, R.R.; Holtan, S.G.; Rashidi, A.; Parker, S.L.; Knights, D.; Luna, J.I.; Dunai, C.; et al. Obesity induces gut microbiota alterations and augments acute graft-versus-host disease after allogeneic stem cell transplantation. Sci. Transl. Med. 2020, 12, eaay7713.
More
Video Production Service