Vol 53, No 6 (2022)
Review article
Published online: 2022-11-22

open access

Page views 3424
Article views/downloads 429
Get Citation

Connect on Social Media

Connect on Social Media

Influence of gut microbiota on efficacy and adverse effects of treatment of lymphoproliferative disorders

Klaudia Zielonka1, Marcin Jasiński12, Krzysztof Jamroziak1
Acta Haematol Pol 2022;53(6):363-375.

Abstract

Gut microbiota has aroused great interest because of its influence on the human body's homeostasis. In addition, multiple reports have indicated its role in the pathogenesis of various diseases. Interestingly, gut microbiota can affect hematological disorders by participating in lymphomagenesis. Patients with lymphoproliferative disorders undergo many procedures that alter their unique microbiota composition and lead to dysbiosis. However, this can have a biased effect as many studies have highlighted gut microbiota’s activity in chemotherapy efficacy, for instance by either enhancing the anti-malignant effects of cyclophosphamide or by diminishing the activity of doxorubicin or cladribine. This review aimed to summarize gut microbiota’s influence on chemotherapy’s outcomes on treatment-related side effects in lymphoproliferative disorders, antimicrobial regimens, and possible gut microbiota modifications to enhance treatment outcomes.

REVIEW ARTICLE

Acta Haematologica Polonica 2022

Number 6, Volume 53, pages 375

DOI: 10.5603/AHP.a2022.0053

ISSN 0001–5814

e-ISSN 2300–7117

Influence of gut microbiota on efficacy and adverse effects of treatment of lymphoproliferative disorders

Klaudia Zielonka1*Marcin Jasiński12Krzysztof Jamroziak1
1Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warszawa, Poland
2Doctoral School, Medical University of Warsaw, Warszawa, Poland

*Address for correspondence: Klaudia Zielonka, Department
of Hematology, Transplantation and Internal Medicine,
Medical University of Warsaw, Banacha 1A, 02–091 Warszawa, Poland,
e-mail: klaudiazielonka99@gmail.com

Copyright © 2022

The Polish Society of Haematologists and Transfusiologists,
Insitute of Haematology and Transfusion Medicine.

All rights reserved.

Received: 27.07.2022 Accepted: 02.10.2022

Copyright © 2022

The Polish Society of Haematologists and Transfusiologists,
Insitute of Haematology and Transfusion Medicine.

All rights reserved.

Abstract
Gut microbiota has aroused great interest because of its influence on the human body’s homeostasis. In addition, multiple reports have indicated its role in the pathogenesis of various diseases. Interestingly, gut microbiota can affect hematological disorders by participating in lymphomagenesis. Patients with lymphoproliferative disorders undergo many procedures that alter their unique microbiota composition and lead to dysbiosis. However, this can have a biased effect as many studies have highlighted gut microbiota’s activity in chemotherapy efficacy, for instance by either enhancing the anti-malignant effects of cyclophosphamide or by diminishing the activity of doxorubicin or cladribine. This review aimed to summarize gut microbiota’s influence on chemotherapy’s outcomes on treatment-related side effects in lymphoproliferative disorders, antimicrobial regimens, and possible gut microbiota modifications to enhance treatment outcomes.
Key words: microbiota, chemotherapy, lymphoproliferative disorders
Acta Haematologica Polonica 2022; 53, 6: 363–375

Introduction

The term ‘gut microbiota’ (GMB) refers to a composition of microbes, including bacteria, viruses, yeast, protozoa, fungi, and archaea, that exist within the human gastrointestinal (GI) tract [1]. Acquired at birth, GMB plays a fundamental role in the development of the immune system and its further homeostasis. The GI tract is the biggest lymphoid organ and the most extensive surface in touch with an external environment [2]. As much as 70% of human immune cells are there, and every day they identify a variety of new antigens. Therefore, a relationship between the GMB and the immune system is required to balance excessive responses to antigen and infectious complications. Furthermore, protective activities to combat pathogens, vitamins and amino acid synthesis, and the structure of the GI tract barrier, are influenced by GMB [3]. One thousand or more species inhabit the GI tract, with the exact composition unique to each individual and appearing to remain stable throughout life [4].

The main phyla in the GI tract are Firmicutes and Bacteroidetes, which are dominant; less abundant are Proteobacteria, Actinobacteria, and Verrucomicrobia [5]. Nevertheless, lifestyle, diet, xenobiotics, physical injury, and disease can alter its composition and lead to dysbiosis, so the state of disruption in the composition and functions of microbiota is triggered by a host of external factors [6].

Hematological patients are especially prone to develop dysbiosis due to hospitalization, infection, malnutrition, treatment, and eventually hematopoietic stem cell transplantation (HSCT).

Interestingly, multiple studies have demonstrated substantial crosstalk between the GMB and the innate immune system in influencing response to tumors, therapy outcome, and patients’ overall survival (OS) [7]. Therefore, this review aimed to highlight the implications of GMB on the efficacy and adverse effects of therapy for lymphoproliferative malignancies.

The composition of GMB in the human body is set out in Table I.

Table I. Composition of gut microbiota in healthy humans (based on [5, 8–10])

Microorganism of human GMB

Dominant phyla
of GMB

Most abundant genus
of GMB

Additional information

Bacteria

Bacteroidetes

Bacteroides

Tanarella

Parabacteroides

Allstipes

Prevotella

Phyla dominate in human GMB and represent 90% of GMB

Each phylum comprises ~30% of bacteria in feces and mucus overlying intestinal epithelium

Substantial numbers of Firmicutes are related to known butyrate-producing bacteria [5, 10]

Firmicutes

Clostridium (represents 95% of Firmicutes phyla)

Lactobacillus

Bacillus

Enterococcus

Ruminicoccus

Actinobacteria

Bifidobacterium

Corynebacterium

Atobium

Phylum is mainly represented by Bifidobacterium genus [10]

Proteobacteria

Escherichia

Shigella

Helicobacter

Less abundant phylum which may comprise ~0.1% of bacteria in strict anerobic environment of colon
Increase of phylum is observed in GMB of people aged over 70 [8]

Fusobacteria

Fusobacterium

Verrucomicrobia

Akkermansia

Methanogenic archaea

Mainly single phylotype Methanobrevibacter smithii

Eukarya

Mainly yeasts

Viruses

Primarily phage

Chronic lymphocytic leukemia

Despite the introduction of novel targeted therapies in chronic lymphocytic leukemia (CLL), such as BCl-2 antagonists and Bruton’s tyrosine kinase (BTK) inhibitors, the FCR protocol combining an anti-CD20 monoclonal antibody with chemotherapeutic agents (fludarabine, cyclophosphamide, and rituximab) is still widely used in fit patients without TP53 gene abnormalities [11]. It has been revealed that the clinical activity of FCR is due to the drugs’ pleiotropic effects. Interestingly, the efficacy and toxicity of FCR components may also be influenced by GMB. For instance, the antineoplastic activity of cyclophosphamide is enhanced by a potent immunomodulatory effect [12]. High doses used in CLL treatment induce intestinal epithelium damage and mucositis. However, a study by Viaud et al. [13] demonstrated that the event is essential for the proper activity of cyclophosphamide. The disrupted barrier resulted in the translocation of commensal Gram-positive bacteria into the secondary lymphoid organs. Subsequently, the commensals enhanced T helper 17 (Th17) cells and memory Th1 immune response, which promoted antitumor activity. Conversely, germ-free mice with tumors were resistant to cyclophosphamide, indicating microbiota’s significant role in the antitumor activity of cyclophosphamide [13]. Furthermore, the analysis outlined Enterococcus hirae and Barnesiella intestinihominis as being responsible for the anti-malignant feature of cyclophosphamide [14].

This finding was assessed in a clinical study by Pflug et al. [15]. A group of patients with CLL treated with anti-Gram-positive antibiotics developed a response to the treatment, progressed significantly earlier (median progression-free survival (PFS) 14.1 vs. 44.1 months, p <0.001), and had reduced OS (median OS 56.1 vs. 91.7 months,
p <0.001) [15]. Independent multivariate analysis confirmed a direct correlation between anti-Gram-positive antibiotics administration and PFS, suggesting the beneficial role of GMB in the antitumor effect of cyclophosphamide [15]. Furthermore, it has been postulated that fludarabine cytotoxicity could also be altered due to dysbiosis, although the mechanism is different [16]. This comprises enzymatic modification of the drug by Gram-negative non-pathogenic
Escherichia coli and gram-positive Listeria welshimeri, resulting in increased antineoplastic activity [16]. Hence, bacterial infections might lead to biotransformation of the fludarabine and enhance its anticancer effect and toxicity.

It is common knowledge that bacteria and viruses participate in lymphomagenesis. However, a study by Hoogeboom et al. [17] presented an influence of fungi on the development of a subtype of CLL with mutated immunoglobulin heavy chain variable gene (IGHV). In the subtype, the IGHV gene encodes B-cell receptors (BCRs), which are significantly specific for β-(١-٦)-glucan, an antigen of yeasts and filamentous fungi [17]. Significantly, CLL cells derived from patients with no known history of fungal infection proliferated in response to the β-(١-٦)-glucan [17]. This suggests the possible participation of fungi in the pathogenesis of CLL, similar to that observed in Helicobacter pylori responsible for gastric mucosa-associated lymphoid tissue (MALT) lymphoma [18]. It would also be interesting to verify whether the fungi infection or the antifungal treatment affects the future outcomes of chemotherapy.

Multiple myeloma

Multiple myeloma (MM) is linked to a state of immunosuppression and immune impairment, and it is assumed that dysbiosis and chronic antigen stimulation could be crucial for the pathogenesis of MM [19]. In addition, the abovementioned studies presented the influence of GMB on response to immunotherapies in CLL. In MM, immunomodulatory drugs are a mainstay of therapy; therefore there was a clear need to determine the effects of GMB on the treatment outcome.

It was recently shown that the composition of harvested microbiota of newly diagnosed patients with MM differs from healthy individuals. Compared to a control group, fecal species were abundant with nitrogen-recycling bacteria such as Klebsiella and Streptococcus [20]. The mechanism of dysbiosis in some MM patients is a result of renal insufficiency, which is one of the symptoms of MM [20]. Impaired renal function leads to the accumulation of metabolites in serum, such as ammonium, which cannot be converted into urea, and hinder its excretion with urine. In MM, ammonium enters the GI tract and stimulates the outgrowth of nitrogen-recycling bacteria. The bacteria prevent ammonium accumulation by hydrolysis of urea and uric acid in the GI tract and de novo synthesis of L-glutamine promoted by Klebsiella [20]. In addition, glutamine is essential for the growth of MM cells and is known for its effect on tumorigenesis [21]. To assess the influence of nitrogen-recycling bacteria on the course of MM, fecal microbiota transplantation (FMT) of MM patients was performed on mice. Those mice which underwent FMT presented severe progression of the disease [20].

Infections in MM are the leading cause of death and may significantly accelerate progression and reduce survival [22]. These findings suggest the potential role of antimicrobial strategies in MM, which could slow progression. The suggestion that GMB influences the progression of MM was confirmed by Calcinotto et al. [23]. In this study, commensal bacteria contributed to the accumulation of cells producing interleukin (IL)-17 in the bone marrow and led to MM’s progression. Prevotella heparinolytica, a GMB component, significantly enhanced the tumor and gut inflammation. Additionally, activation of T cells resulted in their migration to the MM environment, and bone marrow ensued with eosinophil, which enhanced inflammation and consequently resulted in tumor progression. This event can be considered one of the factors responsible for the progression of smoldering MM to MM [23].

Proteasome inhibitors (PIs) are potent agents in the treatment of MM. Nevertheless, they present gastrointestinal adverse effects provoked by dysregulation of the
NF-
κB pathway. Therefore, it is vital to assess whether microbiota, which also influences the NF-κB pathway, may alleviate the side effects of PIs [24, 25]. Another side effect of PIs, mainly bortezomib, is severe peripheral neuropathy characterized by paraesthesia and reduced sensitization. Unfortunately, there is no prevention to PIs-induced neuropathy, and aggravation of the symptoms often leads to tapering off or even withdrawal of PIs [26]. Little is known about the possible mechanism of PI-dependent polyneuropathy; however, activation of astrocytes seems to induce hypersensitivity. Astrocyte activation may be initiated by various factors, including those outside the central nervous system such as bacterial lipopolysaccharides (LPS), contributing to the activation of toll-like receptor 2 (TLR2) and then production of proinflammatory cytokines and chemokines. Interestingly, modulation of the process may be done by activating aryl hydrocarbon receptors located in astrocytes with tryptophan-derived ligand synthesized by Lactobacillus, Clostridium sporogenes, and Peptostreptococcus [27]. However, this is only a hypothesis, and experimental studies are essential to verify if targeting GMB may actually alleviate PIs-induced peripheral neuropathy.

Minimal residual disease (MRD) status after autologous HSCT (auto-HSCT) in MM patients is the principal factor indicative of outcome after the treatment. Intriguingly, Eubacterium halli has been found to be present in higher amounts among MRD(–) patients after auto-HSCT [28]. Therefore, it is clinically relevant to perform additional tests on larger groups to define more species associated with MRD, which could assess treatment efficacy during therapy [28].

Non-Hodgkin lymphoma

This heterogeneous group of diseases is influenced by microbiota in various manners. Manifold organisms may contribute to lymphomagenesis. Persistent inflammation induced by Delftia, Chlamydophila psittaci, and Helicobacter pylori — commensal of approximately half of the human population, may be associated with MALT lymphoma in the conjunctiva, ocular region, and stomach respectively [15, 29, 30]. Eradication of Helicobacter pylori and Chlamydophila psittaci with an antibacterial regimen results in regression of the lymphomas without chemotherapeutic agents [31, 32]. Interestingly, Helicobacter heilmannii induced lymphoma development in mouse models by the formation of infiltration of lymphocytes in the gastric mucosa [33]. Single reports have mentioned Borrelia burgdorferi infection preceding lymphomas [34, 35]. Ataxia–telangiectasia mice exposed to a more sterile environment had a reduced risk of lymphoma [36].

GMB influences not only lymphomagenesis but is essential for anti-malignant effects of chemotherapeutics. Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma. DLBCL is linked to substantially poor outcomes; consequently, 40% of patients who develop complete response will relapse. Great efforts have been made to understand the resistance to the treatment. There are hypotheses concerning altered tumor microenvironment, deficiency in immune cells, and upregulation of inhibitory checkpoint molecules [37]. Studies have revealed that the GMB of patients with DLBCL differed significantly before treatment completion [38]. Furthermore, aggressive DLBCL was characterized by more aberrant and reduced diversity than was indolent lymphoma [39]. Interestingly, the patients who responded to immunotherapy tend to have higher pre-treatment diversity with a distinct composition of GMB and an abundance of Dorea formicigenerans (associated with indolent lymphoma) and Faecalibacterium prausnitzii, known for being related to the better response of melanoma to anti-programmed-cell death 1 (anti-PD1) immunotherapy [39]. The whole GMB composition has been considered a predictor of treatment response. The diversity of GMB has been altered by chemotherapy compared to baseline diversity before treatment, with a marked increase in Bacteroidetes and a concomitant decrease in Firmicutes at the phylum level [39]. The group was small; however, similar studies performed on mouse models revealed that Bifidobacterium upregulated the level of tumor-specific CD8+ T cell and interferon γ secretion and, consequently, increased the sensitivity to anti-PD1 and antibody CTLA4 [40]. Conversely, antibiotics used one month prior to anti-PD1 therapy for epithelial tumors resulted in lower OS (median 20.6 vs. 11.5 months, p <0.001) and PFS (median 4.1 vs. 3.5 months, p = 0.017) [40]. This needs to be further studied in a larger group of patients with lymphoma, as resistance to immunotherapy in melanoma patients has been overcome with probiotics and FMT [40, 41].

Attenuation of the efficacy of chemotherapy comprising platinum salts observed in mouse models has been assessed clinically [42]. Accordingly, earlier use of anti-Gram-positive antibiotics was associated with reduced PFS (median 2.3 vs. 11.5 months, p = 0.001) and OS (median 5.6 vs. 96.8 months, p <0.001) in a group of patients with relapsed lymphoma treated with platinum-based chemotherapy [15]. Platinum genotoxicity needs reactive oxygen species produced by tumor-associated inflammatory cells stimulated by commensals [42]. Additionally, dysbiosis guides chemoresistance to oxaliplatin, although in an unknown manner. On the contrary, in tumor cell lines infected with Mycoplasma hyorhinis, degradation of gemcitabine and a decrease in the antitumoral efficacy induced by mycoplasma thymidine phosphorylase have been observed [43, 44]. Intra-tumor Gammaproteobacteria exerted the same effect, and ciprofloxacin’s administration reversed this effect and elicited anti-malignant activity of the drug [44]. There are more examples of other chemotherapeutics affected by bacteria; for instance, doxorubicin and cladribine activity were significantly decreased by Escherichia coli [16]. However, there has been a lack of clinical studies investigating the correlation of GMB composition with treatment outcomes in hematological patients.

Moreover, doxorubicin is responsible for dysbiosis and affects the integrity of intestinal mucosa, which can lead to fatal infections [45]. Stimulation of NOD2, a critical part of the immune response, by bacterial muramyl dipeptide may have a protective role in mucosal damage induced by doxorubicin [45, 46]. Furthermore, GMB has alleviated the adverse effect of cisplatin. D-methionine, a prebiotic, has antioxidative and anti-inflammatory features and, by promoting the growth of Lachnospiracae and Lactobacillus, can regulate microbiota and reverse cisplatin-induced ototoxicity [47, 48]. Therefore, a hypothesis arises that supplementation could help to avoid cardiac dysfunction and the ototoxic effect of cisplatin. A mouse-performed study showed that Lactobacillus supplementation alleviated weight loss and cardiotoxicity evoked by cisplatin and reduced inflammation [49]. Moreover, in mouse models, an increase of Lactobacillus johnsonii was coherent with delayed development of B-cell lymphoma and oral supplementation was linked to decreased systemic inflammation and genotoxicity [50].

Further interesting findings concerned the accumulation of one of the short-chain fatty acids (SCFAs) — butyrate [51]. Mice fed with a high-fiber diet presented a raised level of butyrate and consequently had a significantly lower risk of lymphoma. Additionally, there was a correlation with tumor growth, as it may lead to apoptosis of lymphoma cells [51]. However, this finding must be treated with caution. Humans have altered the metabolism of SCFAs, and due to degradation in the liver it is undetectable in serum, so the anti-lymphoma effect might not be revealed [52].

Rituximab, an anti-CD20 antibody, markedly affects intestinal microbiota in mice. It is important to emphasize that after treatment with rituximab, GMB composition was reduced in Lactobacillus reuteri [53]. Consequently, the absence of Lactobacillus reuteri may be related to intestinal damage, a severe adverse event of rituximab administration, and inflammatory cell infiltration in the gut mucosa. As a result, administration of antibiotics can lead to rituximab-induced mucositis, and oral supplementation of Lactobacillus reuteri could restore the alleviating effect on gut inflammation [53].

A possible mechanism suggests hampering inflammatory reactions by Lactobacillus reuteri. However, this in vitro observation needs further confirmation via in vivo and clinical studies [53].

Hodgkin lymphoma

Undoubtedly, childhood plays a crucial role in determining GMB diversity because that is when the transition of Th2 response to Th1 takes place. Notably, a study performed on adolescent/young adult Hodgkin lymphoma (HL) patients revealed that their total assemblage of microorganisms was less diverse than that of a healthy population [54]. Moreover, their Th1 response was suppressed while Th2 was enhanced. This suggests that this immunological shift did not happen. Considering possible causes of this condition, less early-childhood fecal-oral exposure due to a more hygienic environment has been postulated as the primary cause [54]. First, however, it is vital to determine whether the less diverse microbiota is not an effect of previous therapy, which is a common cause of the impoverishment of commensals in an oncological patient [54].

Doxorubicin is one of the anthracyclines with a potent antitumor effect. However, its use is limited due to the induction of cardiomyopathy. Additionally, the agent leads to dysbiosis. Interesting findings suggest that dysbiosis contributes to cardiotoxicity. In the analysis, antibiotics administered to mice resulted in depletion of the altered GMB, thus contributing to the alleviation of cardiac failure, inhibition of cardiac cell apoptosis, and decrease in cardiac enzymes activity [55]. FMT obtained a similar effect implemented a day after doxorubicin administration. Then attenuation of doxorubicin-induced dysbiosis and heart function improvement were assessed by left ventricular ejection fraction, and loss of cardiac fibrosis was observed [56]. This must be evaluated in clinical practice, as it raises the possibility of dealing with anthracycline-induced cardiotoxicity.

Programmed death-ligand 1 (PD-L1) blockade presents a highly remarkable outcome in classical HL [57]. Interestingly, the responsiveness and effectiveness of anti-PD1 immunotherapy are significantly determined by GMB [58, 59]. This has been assessed clinically in patients with refractory melanoma, in whom FMT from responders to anti-PD1 therapy to non-responders reprogrammed the resistance to anti-PD1 [59]. Activated and differentiated CD8+ cells, more abundant in the tumor microenvironment of the responders, were the result of both immunotherapy of anti-PD-L1 and FMT, which combated myeloid-induced immunosuppression [59]. An extensive analysis performed on humans revealed that prior use of antibiotics was linked to a poorer outcome of immunotherapy [60]. There raises the question as to which moment is the most appropriate to implement antibiotics. One study indicated that one month prior was enough to diminish the effect of immunotherapy [61]. Hence, similar studies should be performed in a population with HL to establish antimicrobial management in immunotherapy.

Specific bacterial species of Actinobacteria and Firmicutes, detected in patients with melanoma, have been responsible for improving the efficacy of PD-1 [41, 59]. Interestingly, responders had elevated levels of products of bacterial catabolism, which could be treated as biomarkers of microbiome diversity [62]. Notably, the products of catabolism correlated with the presence of taxa associated with response to anti-PD1 [59]. The establishment of key commensals in HL patients is relevant. Patients who are refractory to anti-PD-1 could be treated with FMT and gain response to immunotherapy [59]. It is worth noting that today’s trends for using supplementation probiotics by patients themselves may also lead to worsening response to treatment. Patients anticipating the therapy should be warned about the possible effects of using over-the-counter drugs on their own [63].

Hematopoietic stem cell transplantation

Although its severe complications such as graft-versus-host disease (GvHD) and engraftment syndrome, HSCT including autologous (auto-HSCT), haploidentical, and allogeneic (allo-HSCT) types, remains a powerful method in a variety of hematological malignancies. The source of hematopoietic stem cells may be peripheral blood, bone marrow or umbilical cord [64]. HSCT is preceded by multiple procedures, chemotherapy, anti-infective treatment, and conditioning which can alter the diversity and abundance of GMB, and consequently destroy the beneficial commensals [65]. Interestingly, patients whose diversity or microbiota composition did not change significantly through the process of HSCT (both auto- and allo-) were characterized by fewer complications that contributed to better 2-year OS in contrast to the group with a loss of diversity with a mortality rate of 66.7% [65]. Moreover, antimicrobial prophylaxis is an inevitable part of the event due to the risk of neutropenic fever or infectious complications during the whole HSCT procedure. Nevertheless, studies have had contradictory results on the administration of antibiotics on gut microbial diversity. Kusakabe et al. [65] suggested that carbapenems, cephems, and glycopeptides did not significantly contribute to the loss of diversity and alterations of GMB. Further studies are essential to evaluate correlations among antibiotics to assess the outcome of HSCT and OS and implement protocols in the course of HSCT. For instance, in allo-HSCT, rifaximin was linked to reduced 1-year transplant-related mortality and increased OS (log-rank = 0.008, p = 0.008), unlike ciprofloxacin and metronidazole, due to its protective feature on microbiota diversity, and did not affect the infectious complications at the same time [66]. There is a need to evaluate similar studies for auto-HSCT. The effects of GMB on HSCT are summarized in Table II.

Table II. Summary of known effects of gut microbiota on efficacy and toxicity of cellular therapies used in lymphoproliferative diseases

Organism

Mechanism and/or clinical relevance

Auto-HSCT

Firmicutes and Acinetobacter

Depletion results in loss of butyrate production

Butyrate reduces intestinal permeability leading to anti-inflammatory effect and as a result attenuates chemotherapy-induced mucositis in mice

Depletion led to enhancing mucositis [67]

Proteobacteria

Increased amount during dysbiosis

Increased amount may be considered as a biomarker of dysbiosis [67, 68]

Blautia and Ruminococcus

Presence on day +7 was associated with enhancement and severity of emesis after melphalan conditioning, biomarker of emesis after melphalan conditioning [69]

Tenericutes

Presence is considered a protective factor against nausea [69]

Glomerella

Presence negatively correlated with time to neutrophil engraftment, and development of early culture-negative neutropenic fever [69]

Allo-HSCT

Enterococcus

Increased amount associated with development of GvHD and GvHD-related mortality, conditioning protocols, diet, intestinal mucositis, and antimicrobial prophylaxis enhanced amount of Enterococcus [65, 70]

Eubacterium limosum

Presence associated with reduced risk of relapse after allo-HSCT [71]

Candida albicans

Higher density was associated with worse outcomes of HSCT [72]

Low diversity of microbiota in time of neutrophil engraftment

Increased transplant-related mortality, independent factor of allo-HSCT mortality [73], autologous FMT in time of neutrophil engraftments correlated with better 3-year survival [74]

Clostridiales

A urinary 3-indoxyl produced by Clostridiales was predictive of outcome after HSCT [75]

Responsible for production of butyrate, which acts as an energy source for intestinal epithelial cells and mitigates intestinal effects of GvHD [76]

Butyrate-induced enhancement of Treg cells and restoration of intestinal cells, mitigation of inflammation and GvHD [77], oral supplementation with fructooligosaccharide prebiotics resulted in production of SCAFs, led to proliferation of T regulators, and therefore, decreased risk of GvHD [78]

Clostridiales supplementation was considered to alleviate gastrointestinal symptoms of GvHD [77]

aGvHD

Actinobacteria and Firmicutes

Presence in stool during neutrophil recovery time may serve as a biomarker of development of severe aGvHD [79]

Blautia

Presence beneficial for OS, associated with reduced GvHD mortality [80]

CAR-T

Enterococci

Overgrowth of Enterococci leads to direct stimulation of TLR2 receptors, enhancement of IL-2, TNFα, IFNγ and CD8+ T cells production and severe CRS [81, 82]

Ruminococcus, Bacteroides and Faecalibacterium

Higher amount associated with a complete response at day 100

Implementation of piperacillin/tazobactam, imipenem/cilastatin and meropenem (PIM) four weeks prior to CAR-T CD19 correlated with reduced OS and increased risk of neurotoxicity syndrome; no correlations with CRS were seen [83]

Veillonellales

Higher abundance associated with a decreased complete response at day 100 [83]

Autologous HSCT

Auto-HSCT accounts for approximately half of transplants in Europe. Nowadays, the main indications of auto-HSCT concern lymphoproliferative disorders. The recipient is simultaneously the donor of the stem cells, and the infusion of stem cells is preceded by high-dose conditioning chemotherapy [64]. Unlike allo-HSCT, an association of GMB on auto-HSCT has not been fully elucidated. One of the most common complications of auto-HSCT is mucositis [84]. Intestinal barrier disruption leads to translocation and may induce bloodstream infections [85], which are the main cause of mortality and morbidity in HSCT patients [68, 86].

For a long time, scientists have tried to elucidate the pathogenesis of mucositis. However, few hypotheses postulated the importance of GMB due to its inhibitory effect on the NF-κB pathway and diminishing inflammatory cytokines levels [86]. This was further confirmed in a group of patients with NHL receiving carmustine, etoposide, cytarabine, and melphalan (BEAM) conditioning. Furthermore, depletion in Firmicutes members, influencing NF-κB pathway and Acinetobacter members inhibiting inflammation were observed [67, 87]. Both species are responsible for butyrate production, which causes an anti-inflammatory effect by reducing intestinal permeability and attenuating chemotherapy-induced mucositis in mice [52, 88, 89]. On the other hand, Proteobacteria species were elevated and considered a biomarker of dysbiosis [67, 90]. This led to intestinal barrier disruption and the translocation of bacteria.

Interestingly, there is a correlation between bacteria diversity and complications related to auto-HSCT. High dose melphalan conditioning in plasmatic cell disorders affects the intestinal microbiota variety, and the detection of Blautia and Ruminococcus on day +7 is associated with enhancement and severity of emesis after melphalan conditioning [69]. On the other hand, oral bacteriome at baseline was predictive of nausea, especially the presence of phyla Tenericutes which was regarded as a protective factor [69]. Additionally, bacterial composition at baseline correlated with time to neutrophil engraftment [69]. Development of early culture-negative neutropenic fever was linked to oral fungal genus with the emphasis on Glomerella, whose presence negatively correlated with time to neutrophil engraftment (p = 0.03) [69]. The evaluation of diversity GMB through total procedure auto-HSCT showed only subtle changes of butyrate-producing bacteria and other commensals after auto-HSCT, apart from Lachnospiraceae, of which amounts substantially decreased [61]. Further studies on a larger group are essential to evaluate the possibility of species as a biomarker of complications due to auto-HSCT.

Allogeneic HSCT

In allo-HSCT, stem cells are harvested from related or unrelated donors. It is mainly used to treat acute leukemias and plays a minor role in lymphoproliferative disorders.

Multiple studies have indicated an unquestionable impact of GMB diversity on allo-HSCT [73]. Loss of diversity in GBM is an inevitable event during allo-HSCT. Conditioning protocols, diet, intestinal mucositis, and antimicrobial prophylaxis lead to the impoverishment of beneficial species and the enhancement of Enterococcus, associated with developing GvHD and GvHD-related mortality [65, 70]. A few independent studies have suggested that pre-HSCT microbiota seems to have no significant impact on the future outcome of HSCT, whereas GMB after HSCT is important from the very first day of transplant [65, 70, 72].

There is robust evidence that enhanced microbiota variability correlates with increased overall survival after HSCT, with a 3-year survival of 67% in a high diversity group as opposed to 36% for a low diversity group [73]. Peled et al. [71] indicated that the presence of Eubacterium limosum was associated with a reduced risk of relapse [hazard ratio (HR) 0.82 per 10-fold increase in abundance; 95% confidence interval (CI), 0.71 to 0.95, p = 0.009]. Fungal commensals did not determine the efficacy of HSCT, although a higher density of Candida albicans was associated with a worse outcome depicted in lower OS (p = 0.0008), disease-free survival (p = 0.0064), and GvHD-free (p = 0.026) [72]. Low microbiota diversity, especially during the time of neutrophil engraftment, is significantly linked to worse outcomes contributing to increased transplant-related mortality, and can be considered an independent factor of allo-HSCT mortality (transplant-related mortality: adjusted HR 5.25, p = 0.014) [73]. One analysis indicated that correlation in neutrophil, lymphocyte and monocyte populations during hematological recovery and microbiota dynamics, with reconstitution of GMB, may have beneficial effects on white blood cell (WBCs) counts. Observation of patients who received autologous FMT at the time of neutrophil engraftments showed higher WBCs up to 100 days after FMT and further correlated with better survival. The striking effect of Staphylococcus on lymphocytes and Faecalibacterium on neutrophils has been observed; nevertheless, evaluation of species responsible for the better efficacy of HSCT is relevant for clinical practice [74].

There is a need for better predictive biomarkers for the outcome of HSCT. Notably, increased urinary 3-indoxyl sulphate (3-IS), a fermented product of commensal colonic Clostridiales, and predicts outcomes after HSCT. Studies by Weber at al. revealed that low 3-IS levels within the first 10 days after HSCT correlated with substantially increased transplant-related mortality (p = 5.017) and decreased OS (p = 5.05) one year after HSCT [66, 75] This may suggest that microbiota elements after HSCT could serve as biomarkers of survival after HSCT.

Acute GvHD (aGvHD) is a severe complication of allo-HSCT. Intestinal GvHD affects 54% of patients who developed aGvHD and significantly contributes to increased mortality among patients [91]. There are a few postulated mechanisms of aGvHD regarding microbiota disturbance as a possible factor [80]. Animal models of GvHD showed monodominance of Entereobacteriales with a concomitant decrease of Clostridiales. Interestingly, Clostridiales are important producers of SCFAs, including butyrate. Therefore, Clostridiales supplementation has been considered to alleviate gastrointestinal symptoms of GvHD [77]. In addition, butyrate serves as an energy source for intestinal epithelial cells, and it raises protected intestinal cells and mitigates intestinal effects of GvHD [76]. Another mechanism involving the protective role of butyrate in intestinal aGvHD outlined butyrate-induced enhancement of Treg cells and subsequent restoration of intestinal cells. This resulted in the mitigation of both inflammation and GvHD [77]. Moreover, oral supplementation with fructooligosaccharide prebiotics resulted in the production of SCAFs, led to a proliferation of Tregs, and decreased the risk of GvHD [78].

Considering the mortality rate in aGvHD, biomarkers indicative of aGvHD may help stratify risk in patients. Furthermore, it could be a key in the differential diagnosis between aGvHD and other HSCT complications [92]. Additionally, the neutrophil recovery time is significant for predicting aGvHD and the presence of Actinobacteria and oral Firmicutes in the stool during neutrophil recovery time may serve as a biomarker of the development of severe aGvHD [79].

Intestinal mucositis leads to difficulties with oral intake, something which requires the implementation of parenteral nutrition. Consequently, this may contribute to malnourishment of intestinal cells and reduction in GMB diversity compared to enteral nourishment, eventually triggering aGvHD [93]. Additionally, parenteral nutrition leads to the replenishment of Blautia, which has been associated with reduced GvHD mortality [HR (95% CI) 0.18 (0.05–
–0.63),
p = 0.007] and beneficial for OS (p <0.001) [94].

Chimeric antigen receptor T-cell

CD19-targeted chimeric antigen receptor-modified (CAR) T-cell is a novel therapy registered in lymphoproliferative disorders that is revolutionizing current management. In the long term, CD19-targeted CAR T-cell therapy is inefficacious in most patients who relapse or develop CAR-T-
-related toxicity [82]. Considering bacterial effects on other immunotherapy mentioned in HL [59–61], it seems highly probable that there is a
correlation between CAR-T and GMB. GMB may be one of the determinants responsible for the failure of CAR-T therapy, although little is known yet on this subject. Nevertheless, recent articles have suggested that microbiota and antibiotics do influence CAR-T therapy. Schubert et al. [81] put forward an interesting hypothesis suggesting that overgrowth of Enterococci in intestines may lead to severe cytokine release syndrome (CRS) induced by CAR-T. Direct stimulation of Enterococci to TLR2 receptors, which enables co-stimulatory signal enhancing the production of IL-2, TNFα and IFNγ in murine and human CD8+ T cells, contributed to an increase in polyfunctional T cells against tumor cells and the possibility of CRS [81, 82]. Investigations performed on mice suggested that long-term broad-spectrum antibiotics therapy contributing to loss in bacterial diversity did not influence the outcome of CD19-CAR-T. This is only partially consistent with a retrospective study concerning the use of antibiotics on the efficacy of CD19 CAR-T [95]. Implementation of piperacillin/
/tazobactam, imipenem/cilastatin and meropenem therapy four weeks prior to CAR-T CD19 correlated with reduced OS in patients with NHL (HR = 1.71,
p = 0.011) [83]. Significantly, antibiotics seemed to have no impact on PFS in that cohort. Besides the influence on OS, exposure to these antibiotics was associated with increased immune effector cell-associated neurotoxicity syndrome; however, no correlations with CRS were seen [83]. The impact of GMB on CAR-T is set out in Table II. Further studies concerning microbiota’s influence on CAR-T efficacy are essential.

Figure 1. Gut microbiota and non-Hodgkin lymphoma (created with BioRender.com); MALT — mucosa-associated lymphoid tissue; FMT — fecal microbiota transplant; ROS — reactive oxygen species

Conclusions and perspectives for future

GMB is thought to have tremendous effects on chemotherapy efficacy and outcomes by cooperating with the immune system [13, 15, 59]. However, since many of these results have been obtained from in vitro or in vivo experiments or retrospective patient cohorts, it would be essential to confirm and quantify the influence of GMB on the effects of chemotherapeutic agents, immunotherapy, and HSCT in prospective clinical trials.

In the future, determining the composition of GMB prior to and during treatment may be helpful in further management. Moreover, GMB can serve as a biomarker for patients especially at risk of poor outcomes, and may lead to modifications in their treatment protocol or antimicrobial prophylaxis [39, 62].

Therefore, every effort should be made to maintain microbiota’s primary composition and abundance. However, therapeutic interventions with modifications to reestablish baseline GMB composition such as FMT should be considered in situations resulting in dysbiosis.

By way of illustration, FMT augmented anti-PD-1 immunotherapy and had excellent outcomes in patients with steroid-refractory or dependent type aGvHD as a second-line treatment with or without ruxolitinib [96, 97].

Authors’ contributions

Conceptualization: KZ and KJ. Writing — original draft preparation: KZ. Writing — review and editing: MJ and KJ. Supervision: KJ. All authors read and agreed to published version of manuscript.

Conflicts of interest

None.

Financial support

None.

Ethics

The work described in this article has been carried out in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki) for experiments involving humans; EU Directive 2010/63/EU for animal experiments; uniform requirements for manuscripts submitted to Biomedical journals.

References

  1. Lee KA, Luong MK, Shaw H, et al. The gut microbiome: what the oncologist ought to know. Br J Cancer. 2021; 125(9): 1197–1209, doi: 10.1038/s41416-021-01467-x, indexed in Pubmed: 34262150.
  2. Peaudecerf L, Rocha B. Role of the gut as a primary lymphoid organ. Immunol Lett. 2011; 140(1-2): 1–6, doi: 10.1016/j.imlet.2011.05.009, indexed in Pubmed: 21704078.
  3. Adak A, Khan MR. An insight into gut microbiota and its functionalities. Cell Mol Life Sci. 2019; 76(3): 473–493, doi: 10.1007/s00018-018-2943-4, indexed in Pubmed: 30317530.
  4. Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010; 464(7285): 59–65, doi: 10.1038/nature08821.
  5. Bäckhed F, Ley R, Sonnenburg J, et al. Host-bacterial mutualism in the human intestine. Science. 2005; 307(5717): 1915–1920, doi: 10.1126/science.1104816.
  6. Levy M, Kolodziejczyk A, Thaiss C, et al. Dysbiosis and the immune system. Nat Rev Immunol. 2017; 17(4): 219–232, doi: 10.1038/nri.2017.7.
  7. Xu JYi, Liu MM, Tao TT, et al. The role of gut microbiota in tumorigenesis and treatment. Biomed Pharmacother. 2021; 138: 111444, doi: 10.1016/j.biopha.2021.111444, indexed in Pubmed: 33662679.
  8. Rinninella E, Raoul P, Cintoni M, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019; 7(1), doi: 10.3390/microorganisms7010014, indexed in Pubmed: 30634578.
  9. Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature. 2012; 489(7415): 220–
    –230, doi: 10.1038/nature11550, indexed in Pubmed: 22972295.
  10. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005; 308(5728): 1635–1638, doi: 10.1126/science.1110591, indexed in Pubmed: 15831718.
  11. Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018; 131(25): 2745–2760, doi: 10.1182/blood-2017-09-806398, indexed in Pubmed: 29540348.
  12. Bass KK, Mastrangelo MJ. Immunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer. Cancer Immunol Immunother. 1998; 47(1): 1–12, doi: 10.1007/s002620050498, indexed in Pubmed: 9755873.
  13. Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013; 342(6161): 971–976, doi: 10.1126/science.1240537.
  14. Daillère R, Vétizou M, Waldschmitt N, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016; 45(4): 931–943, doi: 10.1016/j.immuni.2016.09.009.
  15. Pflug N, Kluth S, Vehreschild JJ, et al. Efficacy of antineoplastic treatment is associated with the use of antibiotics that modulate intestinal microbiota. Oncoimmunology. 2016; 5(6): e1150399, doi: 10.1080/2162402X.2016.1150399, indexed in Pubmed: 27471619.
  16. Lehouritis P, Cummins J, Stanton M, et al. Local bacteria affect the efficacy of chemotherapeutic drugs. Sci Rep. 2015; 5: 14554, doi: 10.1038/srep14554, indexed in Pubmed: 26416623.
  17. Hoogeboom R, van Kessel KPM, Hochstenbach F, et al. A mutated B cell chronic lymphocytic leukemia subset that recognizes and responds to fungi. J Exp Med. 2013; 210(1): 59–70, doi: 10.1084/jem.20121801, indexed in Pubmed: 23296468.
  18. Hooi JKY, Lai WY, Ng WK, et al. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology. 2017; 153(2): 420–429, doi: 10.1053/j.gastro.2017.04.022, indexed in Pubmed: 28456631.
  19. Jasiński M, Biliński J, Basak GW. The role of the crosstalk between gut microbiota and immune cells in the pathogenesis and treatment of multiple myeloma. Front Immunol. 2022; 13: 853540, doi: 10.3389/fimmu.2022.853540, indexed in Pubmed: 35432306.
  20. Jian X, Zhu Y, Ouyang J, et al. Alterations of gut microbiome accelerate multiple myeloma progression by increasing the relative abundances of nitrogen-recycling bacteria. Microbiome. 2020; 8(1): 74, doi: 10.1186/s40168-020-00854-5, indexed in Pubmed: 32466801.
  21. Bolzoni M, Chiu M, Accardi F, et al. Dependence on glutamine uptake and glutamine addiction characterize myeloma cells: a new attractive target. Blood. 2016; 128(5): 667–679, doi: 10.1182/blood-2016-01-690743, indexed in Pubmed: 27268090.
  22. Nucci M, Anaissie E. Infections in patients with multiple myeloma. Semin Hematol. 2009; 46(3): 277–288, doi: 10.1053/j.seminhematol.2009.03.006, indexed in Pubmed: 19549580.
  23. Calcinotto A, Brevi A, Chesi M, et al. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat Commun. 2018; 9(1): 4832, doi: 10.1038/s41467-018-07305-8, indexed in Pubmed: 30510245.
  24. Alkharabsheh O, Sidiqi MH, Aljama MA, et al. The human microbiota in multiple myeloma and proteasome inhibitors. Acta Haematol. 2020; 143(2): 118–123, doi: 10.1159/000500976, indexed in Pubmed: 31311009.
  25. Stansborough RL, Gibson RJ. Proteasome inhibitor-induced gastrointestinal toxicity. Curr Opin Support Palliat Care. 2017; 11(2): 133–137, doi: 10.1097/SPC.0000000000000266, indexed in Pubmed: 28333868.
  26. Yamamoto S, Egashira N. Pathological mechanisms of bortezomib-induced peripheral neuropathy. Int J Mol Sci. 2021; 22(2), doi: 10.3390/ijms22020888, indexed in Pubmed: 33477371.
  27. Zhong S, Zhou Z, Liang Y, et al. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol. 2019; 45(4): 369–393, doi: 10.1080/1040841X.2019.1608905, indexed in Pubmed: 31106639.
  28. Pianko MJ, Devlin SM, Littmann ER, et al. Minimal residual disease negativity in multiple myeloma is associated with intestinal microbiota composition. Blood Adv. 2019; 3(13): 2040–2044, doi: 10.1182/bloodadvances.2019032276, indexed in Pubmed: 31289031.
  29. Asao K, Hashida N, Ando S, et al. Conjunctival dysbiosis in mucosa-associated lymphoid tissue lymphoma. Sci Rep. 2019; 9(1): 8424, doi: 10.1038/s41598-019-44861-5, indexed in Pubmed: 31182732.
  30. Travaglino A, Pace M, Varricchio S, et al. Prevalence of Chlamydia psittaci, Chlamydia pneumoniae, and Chlamydia trachomatis Determined by Molecular Testing in Ocular Adnexa Lymphoma Specimens. Am J Clin Pathol. 2020; 153(4): 427–434, doi: 10.1093/ajcp/aqz181, indexed in Pubmed: 31755895.
  31. Schmelz R, Miehlke S, Thiede C, et al. Sequential H. pylori eradication and radiation therapy with reduced dose compared to standard dose for gastric MALT lymphoma stages IE & II1E: a prospective randomized trial. J Gastroenterol. 2019; 54(5): 388–395, doi: 10.1007/s00535-018-1517-4, indexed in Pubmed: 30327875.
  32. Ferreri AJM, Ponzoni M, Guidoboni M, et al. Bacteria-eradicating therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter prospective trial. J Natl Cancer Inst. 2006; 98(19): 1375–1382, doi: 10.1093/jnci/djj373, indexed in Pubmed: 17018784.
  33. O’Rourke JL, Dixon MF, Jack A, et al. Gastric B-cell mucosa-associated lymphoid tissue (MALT) lymphoma in an animal model of ‚Helicobacter heilmannii’ infection. J Pathol. 2004; 203(4): 896–903, doi: 10.1002/path.1593, indexed in Pubmed: 15258991.
  34. Chang CM, Landgren O, Koshiol J, et al. Borrelia and subsequent risk of solid tumors and hematologic malignancies in Sweden. Int
    J Cancer. 2012; 131(9): 2208–2209, doi: 10.1002/ijc.27483, indexed in Pubmed: 22322900.
  35. Schöllkopf C, Melbye M, Munksgaard L, et al. Borrelia infection and risk of non-Hodgkin lymphoma. Blood. 2008; 111(12): 5524–5529, doi: 10.1182/blood-2007-08-109611, indexed in Pubmed: 18424667.
  36. Yamamoto ML, Maier I, Dang AT, et al. Intestinal bacteria modify lymphoma incidence and latency by affecting systemic inflammatory state, oxidative stress, and leukocyte genotoxicity. Cancer Res. 2013; 73(14): 4222–4232, doi: 10.1158/0008-5472.CAN-13-0022, indexed in Pubmed: 23860718.
  37. He MY, Kridel R. Treatment resistance in diffuse large B-cell lymphoma. Leukemia. 2021; 35(8): 2151–2165, doi: 10.1038/s41375-021-01285-3, indexed in Pubmed: 34017074.
  38. Yuan Li, Wang W, Zhang W, et al. Gut microbiota in untreated diffuse large B cell lymphoma patients. Front Microbiol. 2021; 12: 646361, doi: 10.3389/fmicb.2021.646361, indexed in Pubmed: 33927704.
  39. Diefenbach CS, Peters BA, Li H, et al. Microbial dysbiosis is associated with aggressive histology and adverse clinical outcome in B-cell non-Hodgkin lymphoma. Blood Adv. 2021; 5(5): 1194–1198, doi: 10.1182/bloodadvances.2020003129, indexed in Pubmed: 33635332.
  40. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015; 350(6264): 1084–1089, doi: 10.1126/science.aac4255.
  41. Routy B, Chatelier ELe, Derosa L, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018; 359(6371): 91–97, doi: 10.1126/science.aan3706.
  42. Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013; 342(6161): 967–970, doi: 10.1126/science.1240527, indexed in Pubmed: 24264989.
  43. Vande Voorde J, Sabuncuoğlu S, Noppen S, et al. Nucleoside-catabolizing enzymes in mycoplasma-infected tumor cell cultures compromise the cytostatic activity of the anticancer drug gemcitabine. J Biol Chem. 2014; 289(19): 13054–13065, doi: 10.1074/jbc.M114.558924, indexed in Pubmed: 24668817.
  44. Geller LT, Barzily-Rokni M, Danino T, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017; 357(6356): 1156–1160, doi: 10.1126/science.aah5043.
  45. Rigby RJ, Carr J, Orgel K, et al. Intestinal bacteria are necessary for doxorubicin-induced intestinal damage but not for doxorubicin-induced apoptosis. Gut Microbes. 2016; 7(5): 414–423, doi: 10.1080/19490976.2016.1215806, indexed in Pubmed: 27459363.
  46. Nigro G, Rossi R, Commere PH, et al. The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe. 2014; 15(6): 792–798, doi: 10.1016/j.chom.2014.05.003, indexed in Pubmed: 24882705.
  47. Gopal KV, Wu C, Shrestha B, et al. d-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol. 2012; 34(5): 495–504, doi: 10.1016/j.ntt.2012.06.002, indexed in Pubmed: 22732230.
  48. Wu CH, Ko JL, Liao JM, et al. D-methionine alleviates cisplatin-induced mucositis by restoring the gut microbiota structure and improving intestinal inflammation. Ther Adv Med Oncol. 2019; 11: 1758835918821021, doi: 10.1177/1758835918821021, indexed in Pubmed: 30792823.
  49. Zhao L, Xing C, Sun W, et al. Lactobacillus supplementation prevents cisplatin-induced cardiotoxicity possibly by inflammation inhibition. Cancer Chemother Pharmacol. 2018; 82(6): 999–1008, doi: 10.1007/s00280-018-3691-8, indexed in Pubmed: 30276453.
  50. Yamamoto ML, Schiestl RH. Intestinal microbiome and lymphoma development. Cancer J. 2014; 20(3): 190–194, doi: 10.1097/PPO.0000000000000047, indexed in Pubmed: 24855006.
  51. Wei W, Sun W, Yu S, et al. Butyrate production from high-fiber diet protects against lymphoma tumor. Leuk Lymphoma. 2016; 57(10): 2401–2408, doi: 10.3109/10428194.2016.1144879.
  52. Pryde SE, Duncan S, Hold G, et al. The microbiology of butyrate formation in the human colon. FEMS Microbiology Letters. 2002; 217(2): 133–139, doi: 10.1111/j.1574-6968.2002.tb11467.x.
  53. Zhao B, Zhou B, Dong C, et al. Alleviates gastrointestinal toxicity of rituximab by regulating the proinflammatory T cells . Front Microbiol. 2021; 12: 645500, doi: 10.3389/fmicb.2021.645500, indexed in Pubmed: 34712207.
  54. Cozen W, Yu G, Gail MH, et al. Fecal microbiota diversity in survivors of adolescent/young adult Hodgkin lymphoma: a study of twins. Br J Cancer. 2013; 108(5): 1163–1167, doi: 10.1038/bjc.2013.60, indexed in Pubmed: 23443674.
  55. Huang J, Wei S, Jiang C, et al. Involvement of abnormal gut microbiota composition and function in doxorubicin-induced cardiotoxicity. Front Cell Infect Microbiol, 2022; 12, doi: 10.3389/fcimb.2022.808837.
  56. Zhu H. Doxorubicin-induced cardiotoxicity. Cardiotoxicity. 2018, doi: 10.5772/intechopen.78791.
  57. Xu-Monette ZY, Zhou J, Young KH. PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood. 2018; 131(1): 68–83, doi: 10.1182/blood-2017-07-740993, indexed in Pubmed: 29118007.
  58. Dzutsev A, Goldszmid RS, Viaud S, et al. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol. 2015; 45(1): 17–31, doi: 10.1002/eji.201444972, indexed in Pubmed: 25328099.
  59. Davar D, Dzutsev AK, McCulloch JA, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021; 371(6529): 595–602, doi: 10.1126/science.abf3363, indexed in Pubmed: 33542131.
  60. Elkrief A, Derosa L, Kroemer G, et al. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: a new independent prognostic factor? Ann Oncol. 2019; 30(10): 1572–1579, doi: 10.1093/annonc/mdz206, indexed in Pubmed: 31268133.
  61. Derosa L, Hellmann MD, Spaziano M, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol. 2018; 29(6): 1437–1444, doi: 10.1093/annonc/mdy103.
  62. Pallister T, Jackson MA, Martin TC, et al. Hippurate as a metabolomic marker of gut microbiome diversity: modulation by diet and relationship to metabolic syndrome. Sci Rep. 2017; 7(1): 13670, doi: 10.1038/s41598-017-13722-4, indexed in Pubmed:
    29057986.
  63. Spencer CN, Gopalakrishnan V, McQuade J. Abstract 2838: The gut microbiome (GM) and immunotherapy response are influenced by host lifestyle factors Atlanta, GA, Conference: Proceedings: AACR Annual Meeting 2019; March 29-April 3, 2019.
  64. Duarte RF, Labopin M, Bader P, et al. European Society for Blood and Marrow Transplantation (EBMT). Indications for haematopoietic stem cell transplantation for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2019. Bone Marrow Transplant. 2019; 54(10): 1525–1552, doi: 10.1038/s41409-019-0516-2, indexed in Pubmed: 30953028.
  65. Kusakabe S, Fukushima K, Maeda T, 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(3): 438–449, doi: 10.1111/bjh.16205, indexed in Pubmed: 31566729.
  66. Weber D, Oefner PJ, Dettmer K, et al. Rifaximin preserves intestinal microbiota balance in patients undergoing allogeneic stem cell transplantation. Bone Marrow Transplantation. 2016; 51(8): 1087–1092, doi: 10.1038/bmt.2016.66.
  67. Montassier E, Gastinne T, Vangay P, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther. 2015; 42(5): 515–528, doi: 10.1111/apt.13302.
  68. Sahin U, Kocak Toprak S, Ataca Atilla P, et al. An overview of infectious complications after allogeneic hematopoietic stem cell transplantation. J Infect Chemother. 2016; 22(8): 505–514, doi: 10.1016/j.jiac.2016.05.006, indexed in Pubmed: 27344206.
  69. El Jurdi N, Filali-Mouhim A, Salem I, et al. Gastrointestinal microbiome and mycobiome changes during autologous transplantation for multiple myeloma: results of a prospective pilot study. Biol Blood Marrow Transplant. 2019; 25(8): 1511–1519, doi: 10.1016/j.bbmt.2019.04.007, indexed in Pubmed: 30959164.
  70. Doki N, Suyama M, Sasajima S, et al. Clinical impact of pre-transplant gut microbial diversity on outcomes of allogeneic hematopoietic stem cell transplantation. Ann Hematol. 2017; 96(9): 1517–
    –1523, doi: 10.1007/s00277-017-3069-8, indexed in Pubmed:
    28733895.
  71. Peled JU, Devlin SM, Staffas A, et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J Clin Oncol. 2017; 35(15): 1650–1659, doi: 10.1200/JCO.2016.70.3348, indexed in Pubmed: 28296584.
  72. Malard F, Lavelle A, Battipaglia G, et al. Impact of gut fungal and bacterial communities on the outcome of allogeneic hematopoietic cell transplantation. Mucosal Immunol. 2021; 14(5): 1127–1132, doi: 10.1038/s41385-021-00429-z.
  73. Taur Y, Jenq RR, Perales MA, 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, indexed in Pubmed: 24939656.
  74. Schluter J, Peled JU, Taylor BP, et al. The gut microbiota is associated with immune cell dynamics in humans. Nature. 2020; 588(7837): 303–307, doi: 10.1038/s41586-020-2971-8, indexed in Pubmed: 33239790.
  75. Weber D, Oefner PJ, Hiergeist A, et al. Low urinary indoxyl sulfate levels early after transplantation reflect a disrupted microbiome and are associated with poor outcome. Blood. 2015; 126(14): 1723–1728, doi: 10.1182/blood-2015-04-638858, indexed in Pubmed: 26209659.
  76. Mathewson ND, Jenq R, Mathew A, et al. Gut microbiome–derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat Immunol. 2016; 17(5): 505–513, doi: 10.1038/ni.3400.
  77. Simms-Waldrip TR, Sunkersett G, Coughlin LA, 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, indexed in Pubmed: 28192251.
  78. Andermann TM, Fouladi F, Tamburini FB, et al. A fructo-oligosaccharide prebiotic is well tolerated in adults undergoing allogeneic hematopoietic stem cell transplantation: a phase I dose-escalation trial. Transplant Cell Ther. 2021; 27(11): 932.e1–932.e11, doi: 10.1016/j.jtct.2021.07.009, indexed in Pubmed: 34274493.
  79. Golob JL, Pergam SA, Srinivasan S, et al. Stool microbiota at neutrophil recovery is predictive for severe acute graft vs host disease After Hematopoietic Cell Transplantation. Clin Infect Dis. 2017; 65(12): 1984–1991, doi: 10.1093/cid/cix699, indexed in Pubmed: 29020185.
  80. Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012; 209(5): 903–911, doi: 10.1084/jem.20112408.
  81. Schubert ML, Rohrbach R, Schmitt M, et al. The potential role of the intestinal micromilieu and individual microbes in the immunobiology of chimeric antigen receptor T-cell therapy. Front Immunol. 2021; 12: 670286, doi: 10.3389/fimmu.2021.670286, indexed in Pubmed: 34135898.
  82. Salerno F, Heeren JFv, Guislain A, et al. Costimulation through TLR2 drives polyfunctional CD8(+) T cell responses. J Immunol. 2019; 202(3): 714–723, doi: 10.4049/jimmunol.1801026, indexed in Pubmed: 30578304.
  83. Smith M, Dai A, Ghilardi G, et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 2022; 28(4): 713–723, doi: 10.1038/s41591-022-01702-9, indexed in Pubmed: 35288695.
  84. Fanning SR, Rybicki L, Kalaycio M, et al. Severe mucositis is associated with reduced survival after autologous stem cell transplantation for lymphoid malignancies. Br J Haematol. 2006; 135(3): 374–381, doi: 10.1111/j.1365-2141.2006.06323.x, indexed in Pubmed: 16995885.
  85. Wang C, Li Q, Ren J. Microbiota-immune interaction in the pathogenesis of gut-derived infection. Front Immunol. 2019; 10: 1873, doi: 10.3389/fimmu.2019.01873, indexed in Pubmed: 31456801.
  86. van Vliet MJ, Harmsen HJM, de Bont ES, et al. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 2010; 6(5): e1000879, doi: 10.1371/journal.ppat.1000879, indexed in Pubmed: 20523891.
  87. Lakhdari O, Tap J, Béguet-Crespel, F, et al. Identification of NF-κB modulation capabilities within human intestinal commensal bacteria. J Biomed Biotechnol. 2011; 2011: 282356, doi: 10.1155/2011/­282356, indexed in Pubmed: 21765633.
  88. Segain JP, Raingeard de la Blétière D, Bourreille A, et al. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease. Gut. 2000; 47(3): 397–403, doi: 10.1136/gut.47.3.397, indexed in Pubmed: 10940278.
  89. Ferreira TM, Leonel AJ, Melo MA, et al. Oral supplementation of butyrate reduces mucositis and intestinal permeability associated with 5-fluorouracil administration. Lipids. 2012; 47(7): 669–678, doi: 10.1007/s11745-012-3680-3, indexed in Pubmed: 22648862.
  90. Shin NR, Whon TW, Bae JW. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015; 33(9): 496–503, doi: 10.1016/j.tibtech.2015.06.011, indexed in Pubmed:
    26210164.
  91. Martin PJ, Schoch G, Fisher L, et al. A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment. Blood. 1990; 76(8): 1464–1472, doi: 10.1182/blood.v76.8.1464.bloodjournal7681464.
  92. Giaccone L, Faraci DG, Butera S, et al. Biomarkers for acute and chronic graft versus host disease: state of the art. Expert Rev Hematol. 2021; 14(1): 79–96, doi: 10.1080/17474086.2021.1860001, indexed in Pubmed: 33297779.
  93. Andersen S, Staudacher H, Weber N, et al. Pilot study investigating the effect of enteral and parenteral nutrition on the gastrointestinal microbiome post-allogeneic transplantation. Br J Haematol. 2020; 188(4): 570–581, doi: 10.1111/bjh.16218, indexed in Pubmed: 31612475.
  94. Jenq RR, Taur Y, Devlin SM, 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, indexed in Pubmed: 25977230.
  95. Kuczma MP, Ding ZC, Li T, et al. The impact of antibiotic usage on the efficacy of chemoimmunotherapy is contingent on the source of tumor-reactive T cells. Oncotarget. 2017; 8(67): 111931–111942, doi: 10.18632/oncotarget.22953, indexed in Pubmed: 29340102.
  96. Biliński J, Jasiński M, Tomaszewska A, et al. Fecal microbiota transplantation with ruxolitinib as a treatment modality for steroid-refractory/dependent acute, gastrointestinal graft-versus-host disease: a case series. Am J Hematol. 2021; 96(12): E461–E463, doi: 10.1002/ajh.26365, indexed in Pubmed: 34587331.
  97. Qi X, Li X, Zhao Ye, et al. Treating steroid refractory intestinal acute graft-vs-host disease with fecal microbiota transplantation: a pilot study. Front Immunol. 2018; 9: 2195, doi: 10.3389/fimmu.2018.02195, indexed in Pubmed: 30319644.