Next Article in Journal
Conformational Landscape of Cytochrome P450 Reductase Interactions
Next Article in Special Issue
Microbiome-Derived Metabolites in Allogeneic Hematopoietic Stem Cell Transplantation
Previous Article in Journal
Hedgehog Signaling in Colorectal Cancer: All in the Stroma?
Previous Article in Special Issue
New Frontiers about the Role of Human Microbiota in Immunotherapy: The Immune Checkpoint Inhibitors and CAR T-Cell Therapy Era
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure

by
Mireia Uribe-Herranz
1,2,3,*,
Nela Klein-González
1,2,3,
Luis Gerardo Rodríguez-Lobato
2,4,
Manel Juan
1,2,3,5 and
Carlos Fernández de Larrea
2,4,*
1
Centre de Diagnòstic Biomèdic, Department of Immunology, Hospital Clínic de Barcelona (HCB), 08036 Barcelona, Spain
2
Institut D’ Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica (FCRB), 08036 Barcelona, Spain
3
Plataforma d’Immunoterapia Banc de Sang i Teixits–HSB, Universitat de Barcelona, 08036 Barcelona, Spain
4
Amyloidosis and Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
5
Plataforma d’Immunoteràpia Hospital Sant Joan de Déu-HCB, Esplugues de Llobregat, 08950 Barcelona, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(3), 1026; https://doi.org/10.3390/ijms22031026
Submission received: 21 December 2020 / Revised: 13 January 2021 / Accepted: 15 January 2021 / Published: 20 January 2021
(This article belongs to the Special Issue Microbiota and Cancer)

Abstract

:
Hematological malignancies, including multiple myeloma, lymphoma, and leukemia, are a heterogeneous group of neoplasms that affect the blood, bone marrow, and lymph nodes. They originate from uncontrolled growth of hematopoietic and lymphoid cells from different stages in their maturation/differentiation and account for 6.5% of all cancers around the world. During the last decade, it has been proven that the gut microbiota, more specifically the gastrointestinal commensal bacteria, is implicated in the genesis and progression of many diseases. The immune-modulating effects of the human microbiota extend well beyond the gut, mostly through the small molecules they produce. This review aims to summarize the current knowledge of the role of the microbiota in modulating the immune system, its role in hematological malignancies, and its influence on different therapies for these diseases, including autologous and allogeneic stem cell transplantation, chemotherapy, and chimeric antigen receptor T cells.

1. Introduction

The complex ecosystem formed by trillions of microorganisms living in and on the human body is known as the human microbiota. The microbial communities inhabiting inside us include bacteria, virus, fungi, yeast, and protozoa. Most of the human microbiota resides in the gastrointestinal tract, but many other microbial niches exist such as the skin, the lungs, the vagina, or the mammary glands. The gastrointestinal tract, with a projected area of 32 m2 [1], is among the largest interaction surfaces between the host, environmental factors, and antigens in the human body. The latest study sets an estimation of over 40 trillion intestinal microorganisms, bringing the ratio closer to 1:1 to somatic cells, expected to be around 30 trillion [2]. The diversity of the bacterial community that resides inside the mammalian intestine is influenced by several factors such as physiological features, chemical and nutrient gradients, and the strictly compartmentalized host immune response [3].
The bacteria that comprise the mammal gut microbiota belong primarily to four phyla: Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria. Altogether, these phyla account for over 95% of the total bacteria in the mammalian microbiota, regardless of the animal species [4]. The genetic potential of this biomass was estimated in 3.3 million non-redundant microbial genes by the ‘Metagenomics of the Human Intestinal Tract’ project. According to this study, 99.1% of the genes are of bacterial origin, followed by archaeal, eukaryotic, and viral origins. The analyzed cohort contained 1150 bacterial species abundant enough to be detected and at least 160 of such bacterial species are estimated to be in every single person [5].
The gastrointestinal microbiota is generated after birth through vertical transmission and it is then constantly shaped by environmental factors throughout life. The most relevant factors affecting the intestinal microbe composition are: the childbirth delivery method (vaginal vs. cesarean) [6,7,8]; the diet (including breastfeeding as the most significant microbiome variable during the first year of life); household exposures (especially during infancy) [9]; geographical location and genetics. However, the latter has been shown to have a minor role compared to environmental factors [10]. Medication is another important factor that shapes the microbial composition in the human body. Antibiotics are the most relevant drugs affecting gut microbiota composition [11,12], but non-antibiotic drugs like antimetabolites, antipsychotics, and calcium-channel blockers can also affect the human microbiome [13]. Antibiotics reduce the microbial diversity rapidly and significantly; around one third of the total bacterial taxa is lost in 3 days. Four weeks after a course of antibiotics, bacterial communities mostly return to their initial state, even though this return is not complete [12,14]. It has now been established that antibiotics during early life can have a profound effect upon health, increasing the risk of developing asthma [15], type 1 diabetes [16] or having long-term host metabolic effects [17,18]. According to a recent study, the most efficient therapeutic approach to fully recover the initial microbial state is an autologous fecal microbiota transplant (FMT). This approach induced a faster microbial recovery compared to the use of probiotics, which hindered a complete microbiome reconstitution [19].
The gut microbiota and the human host have co-evolved in a mutualistic association [4]. The host obtains benefits like the strengthening of the gut integrity, intestinal epithelium modulation [20], protection against pathogens [21], fat metabolism support [22], and angiogenesis [23]. Beyond these important functions however, the gut microbiota generates a complex network of metabolic pathways, as the bacterial gene set is approximately 150 times larger than the human. Therefore, one of the key functions of the gut microbiota is its enzymatic capability for enabling the acquisition of vitamins and bioactive compounds. For instance, through the fermentation of non-digestible substrates, like dietary fibers, anaerobic cecal and colonic bacteria produce short chain fatty acids (SCFA). SCFA are saturated aliphatic organic acids present mainly in the intestine, such as acetate, propionate, and butyrate [24].
The bidirectional crosstalk between the immune system and the microorganisms living in the gastrointestinal tract has established connections that extend beyond the metabolic nature. The high bacteria density present in the lower intestine is only separated from tissues and organs by a thin epithelial layer, which represents a great health challenge for the host. If commensal bacteria are not tightly controlled, a bacteria outgrowth could lead to inflammation and sepsis (commensals turn into pathobionts). Therefore, the host immune system is essential for preserving homeostasis with the microbial communities. The mucus layer, the secretion of antimicrobial peptides (AMP) and immunoglobulin A (IgA) by lamina propria plasma cells, and the presence of dendritic cells (DC) are some of the immune system mechanisms that help to stratify the luminal microbes, to minimize the interaction of the microorganisms and the immune system, and to reduce bacterial-epithelial contact [25] (Figure 1). Additionally, the immune system can modulate the gut microbiota composition at a species level [26,27,28]. The microbiota-host interaction works two ways and commensal microorganisms can shape mammalian immunity as well. Accumulating evidences show that gut flora impacts the immune system predominantly through small molecules (bacterial metabolites and other molecules) [29,30,31], which affects the balance between pro- and anti-inflammatory mechanisms [32]. For example, SCFA not only act as key metabolites for mammalian cell metabolism, but they also regulate host immunity. They are implicated in the following processes: they facilitate the extrathymic generation of regulatory T (T reg) cells [32], they regulate the function of the colonic T regs [33], and furthermore, they modulate the function of DCs, including cytokine production [34,35], through their receptor GPR43 [36]. The complete maturation of the host immune system is determined by host-specific commensals; exposure to a non-specific gut commensal or its microbe-associated molecular patterns (MAMPs) is not enough to induce intestinal immune maturation and fails to provide protection against enteric pathogens [37,38]. Mice raised under germ-free conditions have an altered gene-expression profile of the intestinal epithelial-cell layer [20], significantly smaller Peyer’s patches [39], a reduced number of CD4+ T cells [40], and a reduced number of IgA-producing plasma cells [41,42]. Several studies have shown the influence of commensal microbes on T-cell phenotype and function in the gut. For instance, the segmented filamentous bacterium (SFB) is sufficient to induce the appearance of T helper cell type 17 (Th17) CD4+ T cells in the lamina propria [43]. In mice, bacteria from the Clostridiales clusters IV, XIVa, and XVIII can direct T reg differentiation and Bacteroides fragilis can induce mucosal tolerance by defining the T reg lineage differentiation pathway [44,45] (Figure 1). Moreover, changes in microbiota density can alter host metabolism and the frequency of immune populations like lamina propria FoxP3+CD4+ T regs [46]. Beyond the effects on intestinal and local immune physiology, the gut microbiome has systemic effects [47,48]. For instance, peptidoglycan can prime systemic innate immunity by activating neutrophils [49]; polysaccharide (PSA) from B. fragilis can promote the increase in systemic T helper cell type 1 (Th1) CD4+ T cells [39]; and SCFAs can regulate extrathymic/peripheral T regs [32] and DC cytokine production and function [34,35] (Figure 1).
Commensal bacteria can prevent colonization by pathogens via multiple mechanisms, such as the production of microbial metabolites. Lactobacillus species improve the intestinal barrier function by modulating epithelial cell tight junction proteins. Clusters IV, XIVa, and XVIII of Clostridia (via TGF-β) and Bacteroides fragilis (via polysaccharide A (PSA)) promote the differentiation and expansion of regulatory T (T reg) cells and segmented filamentous bacteria (SFB) stimulate the induction of intestinal T helper cell type 17 (Th17) cells. Microbiota antigens are sampled via transepithelial dendrites of dendritic cells (DC) through M cells. These DC migrate to the mesenteric lymph node and there, they induce T-cell differentiation. Activated CD4+ T cells also prime B cells, and IgA+ secreting plasma cells then migrate to the lamina propria. Upon translocation into the circulation, bacterial metabolites from commensal microbes have multiple effects. Peptidoglycan can prime neutrophils and PSA can promote the increase in Th1 CD4+ T cells. SCFAs can regulate extrathymic/peripheral T regs, modulate leukocyte trafficking and DC cytokine production and function.
Gut microbiota has an additional important role in defining the TCR and BCR repertoire. It is known that there are circulating memory CD4+ T cells reactive against commensal bacterial lysates [50]. For instance, using a spontaneous uveitis mouse model, Horai and colleagues showed that the activation of retina-specific T cells was dependent on gut commensal microbiota [51]. Moreover, colonic T regs have a different TCR repertoire than peripheral T regs, implying a recognition of local colonic antigens [52]. Regarding the influence of the commensal microbiota on the BCR repertoire, ileum resident memory B cells expressing IgA and IgG have antigen-specificities for commensal flora [53].
Changes in the normal gut microbiota, called dysbiosis, have been reported to be an important factor in the development of many diseases. Dysbiosis contributes to autoimmune disorders, including arthritis and inflammatory bowel disease, allergic [54,55,56,57,58] and metabolic diseases, such as obesity and diabetes [59] (Figure 2). A recent study determined that 13% of global cancer incidence is due to microorganisms [60]. Moreover, some specific bacteria have been demonstrated to be involved in the process of initiation and progression of carcinogenesis at epithelial barriers [61,62], both locally and systemically. Locally, colorectal carcinogenesis and its progression [63,64] is directly associated with gut microbiota through several mechanisms. Systemically, several studies have confirmed that oncogenesis and tumor progression of breast [65,66], pancreas [67], and hepatocellular carcinoma [68] are influenced by gut microbiota. In addition, the human microbiota has been implicated in modulating the efficacy and toxicity of cancer therapy, including chemotherapy, radiotherapy, and, more recently, immunotherapy [35,69,70,71,72,73,74]. All these studies provide strong evidence for a close and complex interplay between the gut microbiome, tumor development, and anti-tumor immunotherapies as explained in the following sections.

2. Gut Microbiota in Hematological Malignancies

The changes of the gut microbiota composition have been assessed both in mouse models and in a clinical setting in adult and pediatric patients with hematological diseases. Most of these patients receive chemotherapy or immunotherapy and many of them additionally receive either antibiotic prophylaxis or other treatments that impact on the microbiota composition and diversity. This fact makes the study of the microbiota in hematological patients particularly challenging.
Ataxia-telangiectasia (A-T) is an autosomal recessive disorder associated with a high incidence of lymphoma. To study the role of intestinal bacteria in the penetrance of lymphoma, Yamamoto and coworkers used an A-T mouse model. This group studied the lymphoma incidence in different mouse colonies harboring different bacterial communities and found that Lactobacillus johnsonii was more abundant in the more cancer-resistant mouse colony and it could even reduce systemic inflammation and genotoxicity when administered orally to the more cancer-prone colony. Moreover, Ataxia-telangiectasia mutated (ATM) gene-deficient (Atm-/-) mice that were exposed to a more sterile environment lived longer and had a reduced lymphoma penetrance. This seemed to be associated with a reduced systemic inflammatory state (reduced basal leukocytes and cytokine-mediated inflammation) [75].
A study of adolescent/young adult Hodgkin lymphoma (AYAHL) survivors found that they had fewer early childhood fecal-oral exposures compared with healthy controls, which suggests reduced exposure to infections in these patients over childhood. Furthermore, suppressed Th1 activity and an increased T helper cell type 2 (Th2) response has been reported in AYAHL [76]. During childhood, the acquisition of gut microbiota diversity changes from a more immature Th2 to a Th1-orchestrated immune profile [77]. Both an increase in Th2 cytokines and IgE in AYAHL and a decrease in cytotoxic T cells and NK cells in Hodgkin lymphoma (HL) might suggest a failure to make this Th2-to-Th1 change in AYAHL. One reason could be that the observed decreased diversity of the gut microorganisms could impact the development of AYAHL [78,79]. Another study reported that AYAHL survivors seemed to have a reduced number of rare gut microorganisms compared to the unaffected twin controls [80]. As we mentioned at the beginning of this section, it is highly controversial whether this reduced microbial diversity is due to the environment as an initial risk factor, the lymphoma itself, or the treatment the patients received. However, it has been observed that gut microorganisms cause oxidative stress that can affect carcinogenesis and influence different pathways associated with lymphomagenesis [81,82,83,84,85]. Indeed, many pathogens have been directly associated with lymphomagenesis, such as Epstein-Barr virus (EBV), human herpesvirus 8 (HHV-8), human T-cell leukemia virus type 1 (HTLV-1), and Helicobacter pylori (HP), although the details regarding this are out of the scope of this review.
To study the influence of antibiotics that modulate intestinal microbiota in the efficacy of antineoplastic treatment, a group in Germany carried out a study on patients with relapsed lymphoma that were treated with cisplatin and patients with chronic lymphocytic leukemia (CLL) that were treated with cyclophosphamide from the CLL8 trial (NCT00281918) and the Cologne Cohort of Neutropenic Patients (NCT01821456). Among the 122 patients with relapsed lymphoma and the 800 patients with CLL, those treated with anti-Gram-positive antibiotics achieved a significantly lower overall response rate (ORR) and progressed significantly earlier [86]. The use of anti-Gram-positive antibiotics was independently associated with reduced progression-free survival (PFS) and overall survival (OS) in the multivariate analysis. A negative impact of anti-Gram-positive antibiotics on the efficacy of cyclophosphamide and cisplatin has also been observed in mouse models [74,87].
As mentioned before, commensal bacteria are involved in the differentiation of Th17 cells, which are characterized by their production of IL-17 and play a critical role in inflammation [88]. Calcinotto and colleagues studied the influence of intestinal microorganisms on multiple myeloma (MM) genesis in a mouse strain that develops a de novo disease mimicking MM. The commensal bacteria Prevotella heparinolytica was found to promote the differentiation of Th17 cells colonizing the gut; these cells migrated to the bone marrow (BM) where they favored tumor progression. Prevotella heparinolytica was also found to promote the progression of MM via an eosinophil-mediated inflammation [89]. A study in humans evaluated whether alterations in the intestinal flora are associated with relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and it was reported that microorganisms from the genus Eubacterium were associated with a reduced risk of MM relapse after allo-HSCT (as explained in more depth below) [90].
Decrease in bacterial diversity leads to a reduced colonization resistance against invading pathogens and has been associated with different pathological conditions such as inflammatory bowel disease, diabetes and obesity, as explained previously [91]. Several groups have reported the significant decrease in bacterial diversity both in acute lymphoblastic leukemia (ALL) and in acute myeloid leukemia (AML), and this effect has even been observed five years after the diagnosis [92,93,94,95,96,97,98].
In the following sections, we outline how the microbiota influences the response to treatment, including hematopoietic stem cell transplantation, both allogeneic and autologous.

3. The Treatment of Hematological Malignancies and Microbiota

Live wild-type bacteria can affect the efficacy of some anti-cancer agents either positively or negatively, in vitro and in vivo, most likely via enzymatic modifications (Table 1). Thus, in multiple reports, the efficacy of 20% of tested chemotherapy drugs was increased, the efficacy of 30% was decreased, and 50% were unaffected by bacteria. The cytotoxicities of cladribine, gemcitabine other commonly-used chemotherapy agents, like etoposide, and also anti-cancer antibiotics, such as doxorubicin, were decreased by bacteria. In contrast, bacteria increased fludarabine and 6-mercaptopurine cytotoxicity [99]. Moreover, these in vitro observations were replicated in an experimental mouse model where it was demonstrated that bacteria could hamper the effects of a selected drug, namely gemcitabine.
In addition to bacteria affecting drug effectiveness in patients with hematological malignancies, the microbiota is itself influenced by chemotherapeutics. Cyclophosphamide is commonly used in the treatment of patients with lymphoma, MM, and AL amyloidosis, as well as part of stem cell transplantation conditioning and chimeric antigen receptor (CAR) T cell lymphodepletive treatment [74]. These drugs can alter the composition of microbiota in the small intestine of mice, inducing the translocation of selected species of Gram-positive bacteria into secondary lymphoid organs. These bacteria stimulate the generation of a specific subset of “pathogenic” Th17 cells and memory Th1 immune responses. On the other hand, with platinum chemotherapy (used in some regimens for relapsed lymphoma, for example), infiltrating myeloid-derived cells responded poorly to therapy in germ-free mice, resulting in deficient production of reactive oxygen species and cytotoxicity after chemotherapy, as previously mentioned [87]. Thus, optimal responses to cancer therapy may require an intact commensal microbiota.

4. Allogeneic Stem Cell Transplantation

Allo-HSCT is a potentially curative modality for treating high-risk hematological malignancies (lymphomas and leukemias) and non-malignant conditions, such as aplastic anemia and inherited diseases. This procedure employs a conditioning regimen, including chemotherapy, radiation, radioimmunotherapy, and/or antibody-based immunotherapy with the goal being to reduce residual malignant cells, deplete the bone marrow hematopoietic cells, and suppress the patient’s immune system. Then, the patient receives an infusion of donor hematopoietic stem cells that reconstitutes hematopoiesis. The graft additionally contains allogeneic T-cells, which can attack residual malignant cells (graft vs. tumor effect) or healthy host tissues (graft vs. host disease (GvHD)) [100]. GvHD is still a leading cause of morbidity and mortality [101,102]. Acute GvHD (aGvHD) occurs in about 30–50% of patients and classically develops within 100 days after transplantation; however, late-onset aGvHD may develop even later [103]. Unfortunately, mortality remains high; the 2-year survival rate for patients with grade III and IV aGvHD is 25–30% and 1–2%, respectively [101]. Regarding pathophysiology, three phases have been described. Phase 1 is characterized by tissue damage due to a conditioning regiment that causes release of inflammatory cytokines and activation of host antigen-presenting cells (APC). During the second phase, host and donor APCs activate lymphocytes to produce Th1 cytokines and in phase 3, T cells migrate to target tissues and produce direct damage, especially in the skin, the gut and the liver [102,104]. Chronic GvHD (cGvHD) can occur with or without previous aGvHD and occurs in approximately 40% of patients [103]. The skin, the liver, the lungs and the gastrointestinal tract are the principal target organs [105]. The diagnosis of GvHD is predominantly based on clinical findings, supported by tissue biopsy in the case of aGvHD [106]. Strategies to reduce GvHD include optimization of HLA matching, T-cell depletion, and the use of immunosuppressive prophylaxis [107]. The first-line treatment for acute and chronic GvHD is based on corticosteroid therapy, however 35 to 50% of patients become refractory to this treatment. Responses to subsequent lines of therapy are also poor and there is no accepted standard of care treatment [108,109].
In recent years, the role of the intestinal microbiota in the development of infectious complications, GvHD, and mortality after allo-HSCT has been increasing. Allo-HSCT has been found to be related to a significant decrease in the diversity of the intestinal microbiota and it is believed to be due to the combination of various factors such as conditioning regimens, antibiotics, changes in diet, and intestinal inflammation [110,111,112]. About 50 years ago, different groups showed that, after receiving an allo-HCST, prolonged survival was observed in germ-free (gnotobiotic) animals and animals decontaminated with high-dose antibiotics compared with conventional mice due to reduced GvHD [113,114,115,116,117,118,119]. These results suggested that the microbiota plays a critical role in GvHD, and the absence of microbiota protects against it. However, these germ-free animals had an aberrant immunity and an anomalous development [120,121]. Efforts were made to bring these results into clinical practice. Diverse clinical trials and observational studies analyzed the effects of intestinal decontamination using broad spectrum antibiotics or lamina-airflow isolation rooms. However, the majority of these publications were contradictory and the outcomes were inconsistent [122,123,124,125,126,127,128,129,130]. In an attempt to narrow the antibiotic spectrum, the use of non-absorbable (rifaximin) or anti-anaerobic (metronidazole) antibiotics has been associated with a decrease in the rate and severity of GvHD [127,131,132,133]. Nevertheless, no standardized protocol for prophylactic antibiotics during allo-HSCT has been established [134].
The reduction in the diversity of the intestinal flora and domination by a single taxa at the time of neutrophil engraftment after allo-HSCT has been associated with reduced OS, increased transplant-related mortality, and GvHD-related mortality [135,136,137]. A recent publication showed that patterns of loss of diversity across different transplantation centers and geographic locations are similar. These findings highlighted that higher diversity of gut microbiota was associated with lower mortality, mainly in the subgroup of patients that received unmodified T-cell-repleted grafts [138]. These outcomes have been supported in a recent meta-analysis that highlights the importance of microbiota diversity and the drawbacks of intestinal decontamination [139].
One of the main limitations of the first studies carried out in this field is that most of them did not characterize the microbiota using high-throughput sequencing technology. Advanced molecular microbiological methods have elucidated that domination of a single taxa microorganism is associated with detrimental outcomes. For example, in mouse models the expansion of Lactobacillus and the depletion of Clostridiales were associated with worse GvHD [140]. Human studies have indicated that the presence of Blautia bacterial species is associated with reduced risk of GvHD-related mortality and increased OS [136]. This could suggest that certain microorganisms alleviate inflammation and could be used therapeutically [141]. However, a randomized clinical trial using the probiotic Lactobacillus rhamnosus GG did not show any benefit against GvHD after allo-HSCT [142]. Furthermore, lower levels of Parabacteroides and Bacteroides species were associated with aGvHD [143] and a greater abundance of Firmicutes, Proteobacteria, and Enterobacteriaceae was linked with increased mortality and GvHD [144], while a greater abundance of Lachnospiraceae and Actinomycetaceae was associated with better outcomes [135]. Similarly, monodomination of Enterococcus, Escherichia coli or Prevotella spp. was related to the presence and severity of GvHD [145,146,147,148]. Recently, an elegant study confirmed the role of fecal dominance by Enterococcus spp. in the development of aGvHD and for increased GvHD-related and overall mortality after allo-HSCT. They identified a microbiota-intrinsic mechanism dependent on lactose utilization that favors the expansion of Enterococci, whereby dietary lactose depletions attenuate the outgrowth of enterococci [149]. Contrastingly, Clostridiales have an important anti-inflammatory role, which includes upregulation of T regs, through the production of butyrate. Increased levels of butyrate aid the recovery of damaged intestinal epithelial cells after allo-HSCT and the depletion of anti-inflammatory Clostridia spp. precedes the development of GvHD [150]. Overall, there is sufficient data to confirm the role of the microbiota in immune reconstitution and immunosurveillance. In this context, Peled et al. found associations between the abundance of a bacterial group, mostly of Eubacterium limosum, and relapse after allo-HSCT [90]. However, the potential protective mechanism that could be provided by this bacterium is yet to be elucidated.
Regarding conditioning regimens, these can modify the composition of the intestinal microbiota by decreasing Firmicutes (including Blautia species), Bifidobacteria, and Clostridium cluster XIV and by increasing Enterococcus and Proteobacteria (including Escherichia species). However, it has not been possible to demonstrate a causal relationship between conditioning regimens and the diversity of intestinal bacteria due to other confounding factors such as the concurrent use of prophylactic antibiotics [151,152]. A very recent report from Memorial Sloan Kettering Cancer Center (MSKCC) described the association between neutrophil, lymphocyte, and monocyte populations during hematological recovery and the microbiota dynamics in hundreds of patients who received stem cell transplantation, with positive associations between both obligate anaerobe fermenters and Staphylococcus and immune cell dynamics [153]. The role of microbial metabolites has recently been associated with the development of GvHD [134]. Indole and indole derivates limited intestinal epithelial damage and reduced GvHD while preserving graft versus tumor activity, and this was related to the upregulation of genes associated with type I interferon responses [154]. A low urinary level of indoxyl sulfate (a metabolite of tryptophan) has been associated with the development of GvHD; therefore, it could be a potential biomarker of a disrupted microbiome, as well as risk of GvHD [155]. Low levels of propionate have been detected in stool samples from patients with GvHD [143]. In murine intestinal tissue, reduced levels of butyrate were found after allo-HSCT, and the administration of butyrate improved the integrity of the intestinal epithelial cells and reduced the severity of GvHD [156]. All these data suggest that intestinal microbiome-derived metabolites may modulate intestinal damage and mitigate GvHD [156] (Table 2).

5. Autologous Stem Cell Transplant

Autologous hematopoietic stem cell transplantation (ASCT) is a multistep procedure originally developed for the treatment of hematological malignancies. This is part of the first-line treatment for MM, and an alternative treatment modality for lymphomas and a few solid tumors. After administration of G-CSF and/or chemotherapy, hematopoietic stem cells are harvested. The infusion of these cells is used to bridge hematopoietic failure after high-dose chemotherapy, usually melphalan for MM and combination regimens for lymphoma, such as BEAM (BCNU, etoposide, cytarabine, and melphalan). In this sense, autologous stem cell support is not a “transplant”; however, the term “ASCT” is commonly used. More than half of the stem cell transplants performed in Europe are autologous, most of them for lymphoid malignancies (plasma-cell disorders (mainly MM), non-Hodgkin lymphoma, and Hodgkin lymphoma), although the procedure has been also adapted for the treatment of severe immune-mediated disorders [165,166,167,168]. The source of stem cells for 99% of all autologous transplant procedures is peripheral blood.
There is limited information about the impact of microbiota on ASCT. Preliminary results showed significant changes in the oral microbiome of 51 patients after ASCT, which returned to its pre-chemotherapy composition after three months. However, changes in microbial diversity were more pronounced and rapid in patients who developed mucositis compared to patients who did not [161]. For non-Hodgkin lymphoma, ASCT caused a decrease in Firmicutes and Actinobacteria abundance but an increase in Proteobacteria abundance in fecal samples collected after exposure to conditioning compared to baseline fecal samples [169]. A small pilot study with 15 patients with plasma-cell dyscrasia showed that microbiome composition present at baseline was associated with the incidence and severity of post-transplantation nausea, vomiting, and culture-negative neutropenic fever, as well as with the rate of neutrophil engraftment [170]. Other studies have included ASCT in the design, but the majority of them received allo-HSCT [171]. There are many aspects still to explore regarding the relationship between the microbiome and ASCT, and the association of the microbiota with other immune complications such as engraftment syndrome [172] would be of clinical interest for further studies (Table 2).

6. Antibiotics and Acute Myeloid Leukemia

The impact of microbiota composition has not been studied in detail in patients receiving induction therapy for AML. However, antibiotic prophylaxis during chemotherapy could be a factor to consider when evaluating the microbiome in these patients. A report of 60 patients, most with AML (42%) or MM (37%), showed that the treatment of neutropenic fever with beta-lactam antibiotics reduced the diversity of the gut microbiome in comparison to when the prophylactic levofloxacin was used [173]. Moreover, another study reported that baseline microbiome diversity was a strong independent predictor of infection during induction chemotherapy. Higher baseline levels of Porphyromonadaceae appeared protective against infection, while carbapenem use was associated with deeper changes in the microbiome and with infection susceptibility [161]. This microbiome-based information could be useful to design interventional strategies and optimize antibiotic administration in clinical practice for patients with AML.

7. First Line Treatment in Multiple Myeloma

Survival trends for patients with MM appear to be improving in the last decade, and these better outcomes are related to the use of novel therapeutic agents. The introduction of ASCT, followed by immunomodulatory drugs (thalidomide, lenalidomide, pomalidomide) proteasome inhibitors (bortezomib, carfilzomib, ixazomib), and monoclonal antibodies (daratumumab), has improved the OS in older patients and lowered early mortality rates [174,175,176]. A very recent report from MSKCC [163] of 34 MM patients who completed first-line therapy and were candidates for lenalidomide maintenance sought to define the connection between the patient intestinal microbiome and MM disease progression. Patients were evaluated for minimal residual disease (MRD) status after completion of induction and before the start of maintenance therapy and stool samples were obtained for DNA sequencing to identify the microbiome composition. A higher relative abundance of the butyrate producer E. hallii was observed in patients without MRD in their BM when compared to those that were MRD+. F prausnitzii was identified as another microbe potentially associated with negative MRD status after initial therapy. Future studies are needed to confirm these findings about intestinal microbiota composition and deeper treatment response. In general, information about the link between myeloma treatment and the microbiome is scarce.

8. CAR T Cells

CARs are synthetic fusion proteins that redirect lymphocytes to recognize and eliminate cells that express a target antigen on its surface. A CAR is endowed with four fundamental components: an extracellular antigen binding domain or single-chain variable fragment (scFv) derived typically from the immunoglobulin structure; a spacer or hinge region; a transmembrane domain from CD8α or CD28 molecules; and the intracellular or activation domains derived from the CD3ζ subunit of the TCR and one or two co-stimulatory domains derived from CD28 or 4-1BB molecules, among others. Synthetic engineering of T-cells expressing CARs against CD19 antigen have shown outstanding results against B-cell malignancies in clinical trials. Therefore, the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have approved Tisagenlecleucel (Kymriah) for relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (ALL) and R/R diffuse large B-cell lymphoma, and Axicabtagene Ciloleucel (Yescarta) for R/R diffuse large B-cell lymphoma as well. Recently, the FDA has also approved brexucabtagene autoleucel (Tecartus) for R/R mantle cell lymphoma [177,178,179,180,181]. The outstanding results of anti-CD19 CAR T cells have led to numerous clinical trials being carried out on diverse hematological and solid cancers. In this sense, several encouraging results have been presented using CAR T-cells/CART-cell therapy targeting BCMA in MM patients. This therapy has emerged as a potential treatment strategy for R/R MM patients, and it is expected that in the next few months the first anti-BCMA CAR will be approved [182,183].
Fecal samples from 25 patients at MSKCC were collected pre-CAR T cell infusion [164] and microbiota composition was profiled by 16S sequencing to determine the correlation between microbiome composition and the efficacy of CAR T cell therapy. The patients were adult recipients of CAR T cells who varied regarding conditioning regimen, CAR construct, and underlying diagnosis, which included hematologic and solid malignancies. The study found an increased representation of bacterial taxa in the microbiome of patients who achieved a complete remission (CR) versus those who did not. Oscillospiraceae, Ruminococcacaeae, and Lachnospiraceae were enriched in patients with CR and Peptostreptococcaceae was more abundant in patients who did not achieve a CR. A higher abundance of Lachnospiraceae was found in those who experienced some toxicity (cytokine release syndrome or neurotoxicity of grade 1 to 4), while Peptostreptococcaceae was more abundant in patients who did not have toxicity. Although very preliminary, these data indicate that features of the microbiota may correlate with outcomes of CAR T cell therapy (Table 2).

9. Outlook-Future Perspective

Gut microbiota has been studied in detail for the last decade and overall data suggest the great impact of intestinal microorganisms upon carcinogenesis, cancer proliferation, HSCT outcome, and response to anticancer therapeutics. Hence, gut microbiota modulation is an exciting and important field of research that will likely be used as a complement to existing therapies, either to enhance the efficacy of treatments or to diminish first-line treatment side effects. To date, several microbiota-targeted approaches have been used to modulate the gastrointestinal bacterial composition (Figure 2).
Dietary intervention can change the gut microbiota composition within 24–48 h at the level of species and family, but not phyla [184]. In addition, the circadian rhythm needs to be accounted for, as at least 10% of Operational Taxonomic Units (OTUs) exhibit diurnal oscillations in their abundance [185]. A Western-style diet, which is known to be linked to higher levels of inflammatory markers and with inflammation-related chronic diseases [186], has been associated with CLL. The author of the study hypothesized that the Western diet microbiota could lack the diversity to establish a balanced immune response, therefore suggesting that a percentage of CLL cases could be prevented by a change in dietary patterns [187]. Dietary intervention is also being tested in a clinical trial of subjects undergoing allo-HSCT in which a potato-based resistant starch is administered for 100 days, beginning just before the conditioning phase. The aim is to increase the butyrate level inside the intestine to mitigate GvHD [188] (Table 3).
Prebiotics, like oligosaccharides, are components that can promote the growth and function of beneficial microorganisms. They can have a direct effect inhibiting pathogen colonization [189,190]. There are a few clinical trials studying the effects of prebiotics on GvHD. A pilot phase I trial, now completed but without published results, tested the side effects and best dose of fructo-oligosaccharides in patients with hematological malignancies undergoing HSCT, with the main goal being to reduce the incidence of GvHD [188,191]. A phase I/II clinical trial, not recruiting yet, has been approved to determine whether a prebiotic, galacto-oligosaccharide, a host carbohydrate, can modulate the microbiome to prevent GvHD after allo-HSCT. During phase I, the maximum tolerated dose will be determined and in phase II, participants will be randomized to receive the prebiotic or a placebo [192]. Lysozyme is an enzyme that plays a role in defense against gastrointestinal pathogens. It is effective against Gram-positive and Gram-negative bacteria and promotes beneficial microbes and reduces detrimental microbes within gut microbial communities [193,194]. A phase I trial will study the safety and efficacy of goat milk that is genetically engineered to produce human lysozyme in the context of preventing GvHD in patients with hematological malignancies undergoing HSCT [195]. To date, the use of prebiotics as an adjuvant treatment in the hematological malignancies setting is hindered by a lack of solid data. However, great efforts are being made in this field and the results of different ongoing clinical trials are eagerly awaited (Table 3).
Probiotics are beneficial microorganisms that, in the correct amount, confer health benefits for the host. Besides restoring microbial dysbiosis and preventing colonization of pathogenic bacteria [196], probiotic byproducts can lower the intestinal infection risk and inflammation [197]. Probiotics can be directly introduced as defined microbial strains, engineered microbes, or by FMT. As a defined strain strategy, there is a randomized open label pilot study testing CBM588 (Clostridium butyricum CBM 588 Probiotic Strain). The aims of this study are the following: (1) to assess the side effects of this probiotic, and (2) to evaluate if it improves the clinical outcome in patients undergoing allo-HSCT by increasing gut microbiota diversity [198]. FMT is an effective approach to restore a dysbiotic intestinal community, therefore returning the homeostasis with the microbial communities. This is achieved by administering a fecal solution from a healthy donor into the gastrointestinal tract of a recipient. Healthy donors are selected after a thorough screening to discard harmful pathogens or family history of autoimmune, metabolic or malignant diseases. FMT can be delivered via colonoscopy, enema, nasogastric or nasojejunal tube and gastroscopy. Moreover, FMT can now be performed orally through a freeze-dried capsule containing the fecal matter. This therapeutic procedure represents an innovation in the hematological field with a remarkable potential to minimize the side effects of standard treatments like antibiotics or chemotherapy. Currently, FMT is only approved for the Clostridioides difficile infection (CDI) but there are already some promising results. Refractory immune checkpoint inhibitor-associated colitis was successfully treated with FMT in two patients [199]. At this time, over 10 clinical trials are being run to establish if FMT is a viable treatment for GvHD [200,201,202,203,204,205,206,207,208], as well as for gastrointestinal complications in AML patients [209,210], none with posted results. Importantly, some of the clinical trials are already testing FMT as a first-line treatment for severe aGvDH [201,208] or as an upfront treatment to avoid or reduce the incidence of GvHD [209,210] or multidrug-resistant bacteria (MDRB) [210] in HSCT patients. Although it has been shown that in the recurrent CDI prevention context, FMT administered orally via capsules is as effective as administered by colonoscopy [211], these clinical trials are testing several FMT administration options: oral administration through a freeze-dried capsule [198,200,202,203,206,207,209], colonoscopy/gastroscopy [201,205,208], rectal enema [210], and nasojejunal tube/gastroscopy [204] administration. While an increasing number of studies are testing FMT as a treatment, it has encountered several difficulties that have halted its application in the clinic. A patient died from a drug-resistant Escherichia coli bacteremia transmitted by FMT after an allo-HSCT [212]. Moreover, the FDA issued a safety alert in 2020 after the death of patients receiving FMT for CDI. Patients developed infections caused by enteropathogenic Escherichia coli (EPEC) and Shiga toxin-producing Escherichia coli (STEC) [213]. Hence, a better stool donor screening technique to avoid the transmission of harmful microorganisms is required and multiple clinical trials are ongoing to confirm the safety and viability of FMT (Table 3).
Postbiotics are bacterial bioactive compounds that can be delivered directly to a patient without targeting the gut microbiota composition. This is done by the exogenous administration of a specific metabolite, like SCFAs, flavonoids, the organic acid taurine, and indole derivatives. A recent study showed that higher circulating concentrations of butyrate and propionate are associated with protection from cGvHD in allo-HSCT patients [214]. On the other hand, the gut microbial metabolite trimethylamine N-oxide (TMAO) aggravates GvHD in mice, enhancing M1 macrophage polarization via NLRP3 inflammasome activation [215]. These two examples highlight the therapeutic potential of postbiotics in hematological malignancies (Table 3).
Antibiotic strategies, that include selecting more narrow-spectrum agents, are currently being tested in clinical trials in the HSCT setting in order to preserve intestinal microbiota composition and therefore reduce the incidence and severity of aGvHD [216] (Table 3).

10. Concluding Remarks

Gut microbiota can shape the immune system well beyond the gastrointestinal tract and this is key to comprehending its role in health and disease. Understanding this interplay will help us to identify novel targets in the design of new approaches to treat hematological malignancies. For instance, a possible approach to treat cancer as an adoptive T-cell therapy (ACT), like CAR-T therapy, would be the use of cross-reactive T cells. Cross-reactive T cells could recognize bacterial antigens but are able to recognize tumor-associated antigens as well. Migrating bacterial antigen-loaded DCs would travel to secondary lymphoid organs and potentially prime cross-reactive anti-tumor T cells. These T cells could be potentially used as an ACT themselves or as an adjuvant for CAR-T cell therapy. Furthermore, gastrointestinal flora modulation is a promising approach to improve the efficacy of hematological malignancies therapies, especially to avoid the serious complications related to those treatments. The clinical trials ongoing in this field will help to establish this approach as a therapeutic tool, adding a valuable personalized medicine resource in the standard care treatment of hematological malignancies (Figure 2). FMT is the most advanced option to move forward. As outlined in this review, multiple clinical trials are being developed in the setting of hematological malignancies. Several key factors must be taken into consideration, like the source of the fecal transplant, autologous vs. healthy donor, its way of administration, the dose, and the timing of infusion. It is also important to acknowledge that different hematological malignancies, or their complications, may need different approaches. Moreover, once FMT is validated as a therapeutic tool beyond CDI, it will be necessary to establish the required technology and to standardize the donor microbiota. As mentioned before, besides FMT, other strategies are being studied in pre-clinical models and clinical trials. A lot of resources are being used to uncover the most efficient way to modulate the gastrointestinal flora, but to date, the use of dietary intervention, prebiotics, probiotics, or postbiotics is hindered by a lack of solid and reproducible data. This highlights the need for more basic and preclinical research to better understand the mechanisms of action behind gut microbiota modulation.

Author Contributions

Conceptualization and Original Draft Preparation, M.U.-H. and C.F.d.L.; Writing—Review & Editing M.U.-H., N.K.-G., L.G.R.-L., M.J. and C.F.d.L. All authors have read and agreed to the published version of the manuscript.

Funding

M.U-H. as “Beatriu de Pinos” fellow (2019-BP-00204) has received funding from the Ministry of Business and Knowledge of the Government of Catalonia and the European Union’s Horizon 2020, Marie Sklodowska-Curie COFUND (BP3, under the agreement nº 801370). L.G.R.-L. as BITRECS fellow has received funding from the European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant agreement No 754550 and from “La Caixa” Foundation. This work has been supported in part by grants from the Instituto de Salud Carlos III, Spanish Ministry of Health (FIS PI18/00775, PI19/00669, ICI19/00025 and complementary grant for CONCORD-023), Fondo Europeo de Desarrollo Regional (FEDER) and 2017SGR00792 (AGAUR; Generalitat de Catalunya) and “La Caixa” Foundation (CP042702). We would like to thank Anthony Battram for his help with manuscript editing.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Helander, H.F.; Fändriks, L. Surface Area of the Digestive Tract-Revisited. Scand. J. Gastroenterol. 2014, 49, 681–689. [Google Scholar] [CrossRef] [PubMed]
  2. Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Donaldson, G.P.; Lee, S.M.; Mazmanian, S.K. Gut Biogeography of the Bacterial Microbiota. Nat. Rev. Microbiol. 2016, 14, 20–32. [Google Scholar] [CrossRef] [Green Version]
  4. Ley, R.E.; Hamady, M.; Lozupone, C.; Turnbaugh, P.; Ramey, R.R.; Bircher, J.S.; Schlegel, M.L.; Tucker, T.A.; Schrenzel, M.D.; Knight, R.; et al. Evolution of Mammals and Their Gut Microbes. Science 2008, 320, 1647–1651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A Human Gut Microbial Gene Catalogue Established by Metagenomic Sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery Mode Shapes the Acquisition and Structure of the Initial Microbiota across Multiple Body Habitats in Newborns. PNAS 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
  7. Ferretti, P.; Pasolli, E.; Tett, A.; Asnicar, F.; Gorfer, V.; Fedi, S.; Armanini, F.; Truong, D.T.; Manara, S.; Zolfo, M.; et al. Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 2018, 24, 133–145. [Google Scholar] [CrossRef]
  8. Jakobsson, H.E.; Abrahamsson, T.R.; Jenmalm, M.C.; Harris, K.; Quince, C.; Jernberg, C.; Björkstén, B.; Engstrand, L.; Andersson, A.F. Decreased Gut Microbiota Diversity, Delayed Bacteroidetes Colonisation and Reduced Th1 Responses in Infants Delivered by Caesarean Section. Gut 2014, 63, 559–566. [Google Scholar] [CrossRef] [Green Version]
  9. Stewart, C.J.; Ajami, N.J.; O’Brien, J.L.; Hutchinson, D.S.; Smith, D.P.; Wong, M.C.; Ross, M.C.; Lloyd, R.E.; Doddapaneni, H.; Metcalf, G.A.; et al. Temporal Development of the Gut Microbiome in Early Childhood from the TEDDY Study. Nature 2018, 562, 583–588. [Google Scholar] [CrossRef]
  10. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment Dominates over Host Genetics in Shaping Human Gut Microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
  11. Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J.K. Long-Term Ecological Impacts of Antibiotic Administration on the Human Intestinal Microbiota. ISME J. 2007, 1, 56–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The Pervasive Effects of an Antibiotic on the Human Gut Microbiota, as Revealed by Deep 16S RRNA Sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef] [PubMed]
  13. Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H.; et al. Extensive Impact of Non-Antibiotic Drugs on Human Gut Bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef] [PubMed]
  14. Dethlefsen, L.; Relman, D.A. Incomplete Recovery and Individualized Responses of the Human Distal Gut Microbiota to Repeated Antibiotic Perturbation. PNAS 2011, 108, 4554–4561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Marra, F.; Marra, C.A.; Richardson, K.; Lynd, L.D.; Kozyrskyj, A.; Patrick, D.M.; Bowie, W.R.; Fitzgerald, J.M. Antibiotic Use in Children Is Associated with Increased Risk of Asthma. Pediatrics 2009, 123, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  16. Livanos, A.E.; Greiner, T.U.; Vangay, P.; Pathmasiri, W.; Stewart, D.; McRitchie, S.; Li, H.; Chung, J.; Sohn, J.; Kim, S.; et al. Antibiotic-Mediated Gut Microbiome Perturbation Accelerates Development of Type 1 Diabetes in Mice. Nat. Microbiol. 2016, 1, 16140. [Google Scholar] [CrossRef]
  17. Cho, I.; Yamanishi, S.; Cox, L.; Methé, B.A.; Zavadil, J.; Li, K.; Gao, Z.; Mahana, D.; Raju, K.; Teitler, I.; et al. Antibiotics in Early Life Alter the Murine Colonic Microbiome and Adiposity. Nature 2012, 488, 621–626. [Google Scholar] [CrossRef]
  18. Cox, L.M.; Yamanishi, S.; Sohn, J.; Alekseyenko, A.V.; Leung, J.M.; Cho, I.; Kim, S.; Li, H.; Gao, Z.; Mahana, D.; et al. Altering the Intestinal Microbiota during a Critical Developmental Window Has Lasting Metabolic Consequences. Cell 2014, 158, 705–721. [Google Scholar] [CrossRef] [Green Version]
  19. Suez, J.; Zmora, N.; Zilberman-Schapira, G.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Zur, M.; Regev-Lehavi, D.; Ben-Zeev Brik, R.; Federici, S.; et al. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell 2018, 174, 1406–1423. [Google Scholar] [CrossRef] [Green Version]
  20. Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular Analysis of Commensal Host-Microbial Relationships in the Intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [Green Version]
  21. Bäumler, A.J.; Sperandio, V. Interactions between the Microbiota and Pathogenic Bacteria in the Gut. Nature 2016, 535, 85–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice. Science 2013, 341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Andriessen, E.M.; Wilson, A.M.; Mawambo, G.; Dejda, A.; Miloudi, K.; Sennlaub, F.; Sapieha, P. Gut Microbiota Influences Pathological Angiogenesis in Obesity-driven Choroidal Neovascularization. EMBO Mol. Med. 2016, 8, 1366–1379. [Google Scholar] [CrossRef] [PubMed]
  24. Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short Chain Fatty Acids in Human Large Intestine, Portal, Hepatic and Venous Blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Macpherson, A.J.; Slack, E.; Geuking, M.B.; McCoy, K.D. The Mucosal Firewalls against Commensal Intestinal Microbes. Semin. Immunopathol. 2009, 31, 145–149. [Google Scholar] [CrossRef]
  26. Elinav, E.; Strowig, T.; Kau, A.L.; Henao-Mejia, J.; Thaiss, C.A.; Booth, C.J.; Peaper, D.R.; Bertin, J.; Eisenbarth, S.C.; Gordon, J.I.; et al. NLRP6 Inflammasome Regulates Colonic Microbial Ecology and Risk for Colitis. Cell 2011, 145, 745–757. [Google Scholar] [CrossRef] [Green Version]
  27. Garrett, W.S.; Lord, G.M.; Punit, S.; Lugo-Villarino, G.; Mazmanian, S.K.; Ito, S.; Glickman, J.N.; Glimcher, L.H. Communicable Ulcerative Colitis Induced by T-Bet Deficiency in the Innate Immune System. Cell 2007, 131, 33–45. [Google Scholar] [CrossRef] [Green Version]
  28. Vijay-Kumar, M.; Aitken, J.D.; Carvalho, F.A.; Cullender, T.C.; Mwangi, S.; Srinivasan, S.; Sitaraman, S.V.; Knight, R.; Ley, R.E.; Gewirtz, A.T. Metabolic Syndrome and Altered Gut Microbiota in Mice Lacking Toll-like Receptor 5. Science 2010, 328, 228–231. [Google Scholar] [CrossRef] [Green Version]
  29. Skelly, A.N.; Sato, Y.; Kearney, S.; Honda, K. Mining the Microbiota for Microbial and Metabolite-Based Immunotherapies. Nat. Rev. Immunol. 2019, 19, 305–323. [Google Scholar] [CrossRef]
  30. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut Microbiome Structure and Metabolic Activity in Inflammatory Bowel Disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef]
  31. Blacher, E.; Levy, M.; Tatirovsky, E.; Elinav, E. Microbiome-Modulated Metabolites at the Interface of Host Immunity. J. Immunol. 2017, 198, 572–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; de Roos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites Produced by Commensal Bacteria Promote Peripheral Regulatory T-Cell Generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
  33. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-Y, M.; Glickman, J.N.; Garrett, W.S. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nastasi, C.; Candela, M.; Bonefeld, C.M.; Geisler, C.; Hansen, M.; Krejsgaard, T.; Biagi, E.; Andersen, M.H.; Brigidi, P.; Ødum, N.; et al. The Effect of Short-Chain Fatty Acids on Human Monocyte-Derived Dendritic Cells. Sci. Rep. 2015, 5, 16148. [Google Scholar] [CrossRef]
  35. Uribe-Herranz, M.; Rafail, S.; Beghi, S.; Gil-de-Gómez, L.; Verginadis, I.; Bittinger, K.; Pustylnikov, S.; Pierini, S.; Perales-Linares, R.; Blair, I.A.; et al. Gut Microbiota Modulate Dendritic Cell Antigen Presentation and Radiotherapy-Induced Antitumor Immune Response. J. Clin. Invest. 2020, 130, 466–479. [Google Scholar] [CrossRef]
  36. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.-Y.; Lannoy, V.; Decobecq, M.-E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional Characterization of Human Receptors for Short Chain Fatty Acids and Their Role in Polymorphonuclear Cell Activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
  37. Bauer, H.; Horowitz, R.E.; Levenson, S.M.; Popper, H. The Response of the Lymphatic Tissue to the Microbial Flora. Studies on Germfree Mice. Am. J. Pathol. 1963, 42, 471–483. [Google Scholar]
  38. Chung, H.; Pamp, S.J.; Hill, J.A.; Surana, N.K.; Edelman, S.M.; Troy, E.B.; Reading, N.C.; Villablanca, E.J.; Wang, S.; Mora, J.R.; et al. Gut Immune Maturation Depends on Colonization with a Host-Specific Microbiota. Cell 2012, 149, 1578–1593. [Google Scholar] [CrossRef] [Green Version]
  39. Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An Immunomodulatory Molecule of Symbiotic Bacteria Directs Maturation of the Host Immune System. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [Green Version]
  40. Talham, G.L.; Jiang, H.-Q.; Bos, N.A.; Cebra, J.J. Segmented Filamentous Bacteria Are Potent Stimuli of a Physiologically Normal State of the Murine Gut Mucosal Immune System. Infect. Immun. 1999, 67, 1992–2000. [Google Scholar] [CrossRef]
  41. Macpherson, A.J.; Hunziker, L.; McCoy, K.; Lamarre, A. IgA Responses in the Intestinal Mucosa against Pathogenic and Non-Pathogenic Microorganisms. Microbes Infect. 2001, 3, 1021–1035. [Google Scholar] [CrossRef]
  42. Shroff, K.E.; Meslin, K.; Cebra, J.J. Commensal Enteric Bacteria Engender a Self-Limiting Humoral Mucosal Immune Response While Permanently Colonizing the Gut. Infect. Immun. 1995, 63, 3904–3913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ivanov, I.I.; de Llanos Frutos, R.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific Microbiota Direct the Differentiation of Th17 Cells in the Mucosa of the Small Intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg Induction by a Rationally Selected Mixture of Clostridia Strains from the Human Microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
  45. Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ Regulatory T-Cell Development by a Commensal Bacterium of the Intestinal Microbiota. PNAS 2010, 107, 12204–12209. [Google Scholar] [CrossRef] [Green Version]
  46. Contijoch, E.J.; Britton, G.J.; Yang, C.; Mogno, I.; Li, Z.; Ng, R.; Llewellyn, S.R.; Hira, S.; Johnson, C.; Rabinowitz, K.M.; et al. Gut Microbiota Density Influences Host Physiology and Is Shaped by Host and Microbial Factors. eLife 2019, 8, e40553. [Google Scholar] [CrossRef]
  47. Belkaid, Y.; Naik, S. Compartmentalized and Systemic Control of Tissue Immunity by Commensals. Nat. Immunol. 2013, 14, 646–653. [Google Scholar] [CrossRef] [Green Version]
  48. Khosravi, A.; Yáñez, A.; Price, J.G.; Chow, A.; Merad, M.; Goodridge, H.S.; Mazmanian, S.K. Gut Microbiota Promotes Hematopoiesis Tocontrol Bacterial Infection. Cell Host Microbe 2014, 15, 374–381. [Google Scholar] [CrossRef] [Green Version]
  49. Clarke, T.B.; Davis, K.M.; Lysenko, E.S.; Zhou, A.Y.; Yu, Y.; Weiser, J.N. Recognition of Peptidoglycan from the Microbiota by Nod1 Enhances Systemic Innate Immunity. Nat. Med. 2010, 16, 228–231. [Google Scholar] [CrossRef] [Green Version]
  50. Hegazy, A.N.; West, N.R.; Stubbington, M.J.T.; Wendt, E.; Suijker, K.I.M.; Datsi, A.; This, S.; Danne, C.; Campion, S.; Duncan, S.H.; et al. Circulating and Tissue-Resident CD4+ T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology 2017, 153, 1320–1337. [Google Scholar] [CrossRef] [Green Version]
  51. Lathrop, S.K.; Bloom, S.M.; Rao, S.M.; Nutsch, K.; Lio, C.-W.; Santacruz, N.; Peterson, D.A.; Stappenbeck, T.S.; Hsieh, C.-S. Peripheral Education of the Immune System by Colonic Commensal Microbiota. Nature 2011, 478, 250–254. [Google Scholar] [CrossRef] [PubMed]
  52. Horai, R.; Zárate-Bladés, C.R.; Dillenburg-Pilla, P.; Chen, J.; Kielczewski, J.L.; Silver, P.B.; Jittayasothorn, Y.; Chan, C.-C.; Yamane, H.; Honda, K.; et al. Microbiota-Dependent Activation of an Autoreactive T Cell Receptor Provokes Autoimmunity in an Immunologically Privileged Site. Immunity 2015, 43, 343–353. [Google Scholar] [CrossRef] [Green Version]
  53. Benckert, J.; Schmolka, N.; Kreschel, C.; Zoller, M.J.; Sturm, A.; Wiedenmann, B.; Wardemann, H. The Majority of Intestinal IgA+ and IgG+ Plasmablasts in the Human Gut Are Antigen-Specific. J. Clin. Invest. 2011, 121, 1946–1955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Brusca, S.B.; Abramson, S.B.; Scher, J.U. Microbiome and Mucosal Inflammation as Extra-Articular Triggers for Rheumatoid Arthritis and Autoimmunity. Curr. Opin. Rheumatol. 2014, 26, 101–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-Omics of the Gut Microbial Ecosystem in Inflammatory Bowel Diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
  56. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut Microbiota Metabolism of Dietary Fiber Influences Allergic Airway Disease and Hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
  57. Wen, L.; Ley, R.E.; Volchkov, P.V.; Stranges, P.B.; Avanesyan, L.; Stonebraker, A.C.; Hu, C.; Wong, F.S.; Szot, G.L.; Bluestone, J.A.; et al. Innate Immunity and Intestinal Microbiota in the Development of Type 1 Diabetes. Nature 2008, 455, 1109–1113. [Google Scholar] [CrossRef] [PubMed]
  58. Wu, H.-J.; Ivanov, I.I.; Darce, J.; Hattori, K.; Shima, T.; Umesaki, Y.; Littman, D.R.; Benoist, C.; Mathis, D. Gut-Residing Segmented Filamentous Bacteria Drive Autoimmune Arthritis via T Helper 17 Cells. Immunity 2010, 32, 815–827. [Google Scholar] [CrossRef] [Green Version]
  59. Tilg, H.; Moschen, A.R. Microbiota and Diabetes: An Evolving Relationship. Gut 2014, 63, 1513–1521. [Google Scholar] [CrossRef]
  60. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global Burden of Cancer Attributable to Infections in 2018: A Worldwide Incidence Analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, H.; Sun, L. When Human Cells Meet Bacteria: Precision Medicine for Cancers Using the Microbiota. Am. J. Cancer Res. 2018, 8, 1157–1175. [Google Scholar] [PubMed]
  62. Rosshart, S.P.; Vassallo, B.G.; Angeletti, D.; Hutchinson, D.S.; Morgan, A.P.; Takeda, K.; Hickman, H.D.; McCulloch, J.A.; Badger, J.H.; Ajami, N.J.; et al. Wild Mouse Gut Microbiota Promotes Host Fitness and Improves Disease Resistance. Cell 2017, 171, 1015–1028.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Song, X.; Gao, H.; Lin, Y.; Yao, Y.; Zhu, S.; Wang, J.; Liu, Y.; Yao, X.; Meng, G.; Shen, N.; et al. Alterations in the Microbiota Drive Interleukin-17C Production from Intestinal Epithelial Cells to Promote Tumorigenesis. Immunity 2014, 40, 140–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhan, Y.; Chen, P.-J.; Sadler, W.D.; Wang, F.; Poe, S.; Núñez, G.; Eaton, K.A.; Chen, G.Y. Gut Microbiota Protects against Gastrointestinal Tumorigenesis Caused by Epithelial Injury. Cancer Res. 2013, 73, 7199–7210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lakritz, J.R.; Poutahidis, T.; Mirabal, S.; Varian, B.J.; Levkovich, T.; Ibrahim, Y.M.; Ward, J.M.; Teng, E.C.; Fisher, B.; Parry, N.; et al. Gut Bacteria Require Neutrophils to Promote Mammary Tumorigenesis. Oncotarget 2015, 6, 9387–9396. [Google Scholar] [CrossRef] [Green Version]
  66. Rao, V.P.; Poutahidis, T.; Ge, Z.; Nambiar, P.R.; Boussahmain, C.; Wang, Y.Y.; Horwitz, B.H.; Fox, J.G.; Erdman, S.E. Innate Immune Inflammatory Response against Enteric Bacteria Helicobacter Hepaticus Induces Mammary Adenocarcinoma in Mice. Cancer Res. 2006, 66, 7395–7400. [Google Scholar] [CrossRef] [Green Version]
  67. Ertz-Archambault, N.; Keim, P.; Von Hoff, D. Microbiome and Pancreatic Cancer: A Comprehensive Topic Review of Literature. World J. Gastroenterol. 2017, 23, 1899–1908. [Google Scholar] [CrossRef]
  68. Dapito, D.H.; Mencin, A.; Gwak, G.-Y.; Pradere, J.-P.; Jang, M.-K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef] [Green Version]
  69. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut Microbiome Modulates Response to Anti–PD-1 Immunotherapy in Melanoma Patients. Science 2017, eaan4236. [Google Scholar] [CrossRef] [Green Version]
  70. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The Commensal Microbiome Is Associated with Anti-PD-1 Efficacy in Metastatic Melanoma Patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [Green Version]
  71. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut Microbiome Influences Efficacy of PD-1-Based Immunotherapy against Epithelial Tumors. Science 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium Promotes Antitumor Immunity and Facilitates Anti–PD-L1 Efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Uribe-Herranz, M.; Bittinger, K.; Rafail, S.; Guedan, S.; Pierini, S.; Tanes, C.; Ganetsky, A.; Morgan, M.A.; Gill, S.; Tanyi, J.L.; et al. Gut Microbiota Modulates Adoptive Cell Therapy via CD8α Dendritic Cells and IL-12. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed]
  74. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Science 2013, 342, 971–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yamamoto, M.L.; Maier, I.; Dang, A.T.; Berry, D.; Liu, J.; Ruegger, P.M.; Yang, J.-I.; Soto, P.A.; Presley, L.L.; Reliene, R.; et al. Intestinal Bacteria Modify Lymphoma Incidence and Latency by Affecting Systemic Inflammatory State, Oxidative Stress, and Leukocyte Genotoxicity. Cancer Res. 2013, 73, 4222–4232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Salas, C.; Niembro, A.; Lozano, V.; Gallardo, E.; Molina, B.; Sánchez, S.; Ramos, S.; Carnevale, A.; Pérez-Vera, P.; Rivera Luna, R.; et al. Persistent Genomic Instability in Peripheral Blood Lymphocytes from Hodgkin Lymphoma Survivors. Environ. Mol. Mutagen. 2012, 53, 271–280. [Google Scholar] [CrossRef] [PubMed]
  77. Martin, R.; Nauta, A.J.; Ben Amor, K.; Knippels, L.M.J.; Knol, J.; Garssen, J. Early Life: Gut Microbiota and Immune Development in Infancy. Benef. Microbes 2010, 1, 367–382. [Google Scholar] [CrossRef]
  78. Poppema, S. Immunobiology and Pathophysiology of Hodgkin Lymphomas. Hematol. Am. Soc. Hematol. Educ. Program 2005, 231–238. [Google Scholar] [CrossRef] [Green Version]
  79. Blumberg, R.; Powrie, F. Microbiota, Disease, and Back to Health: A Metastable Journey. Sci. Transl. Med. 2012, 4, 137rv7. [Google Scholar] [CrossRef] [Green Version]
  80. Cozen, W.; Yu, G.; Gail, M.H.; Ridaura, V.K.; Nathwani, B.N.; Hwang, A.E.; Hamilton, A.S.; Mack, T.M.; Gordon, J.I.; Goedert, J.J. Fecal Microbiota Diversity in Survivors of Adolescent/Young Adult Hodgkin Lymphoma: A Study of Twins. Br. J. Cancer 2013, 108, 1163–1167. [Google Scholar] [CrossRef]
  81. Shaffer, A.L.; Rosenwald, A.; Staudt, L.M. Lymphoid Malignancies: The Dark Side of B-Cell Differentiation. Nat. Rev. Immunol. 2002, 2, 920–932. [Google Scholar] [CrossRef] [PubMed]
  82. Pasqualucci, L.; Neumeister, P.; Goossens, T.; Nanjangud, G.; Chaganti, R.S.; Küppers, R.; Dalla-Favera, R. Hypermutation of Multiple Proto-Oncogenes in B-Cell Diffuse Large-Cell Lymphomas. Nature 2001, 412, 341–346. [Google Scholar] [CrossRef] [PubMed]
  83. Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic Inflammation and Oxidative Stress in Human Carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386. [Google Scholar] [CrossRef] [PubMed]
  84. Epeldegui, M.; Widney, D.P.; Martínez-Maza, O. Pathogenesis of AIDS Lymphoma: Role of Oncogenic Viruses and B Cell Activation-Associated Molecular Lesions. Curr. Opin. Oncol. 2006, 18, 444–448. [Google Scholar] [CrossRef]
  85. Illes, A.; Varoczy, L.; Papp, G.; Wilson, P.C.; Alex, P.; Jonsson, R.; Kovacs, T.; Konttinen, Y.T.; Zehert, M.; Nakken, B.; et al. Aspects of B-Cell Non-Hodgkin’s Lymphoma Development: A Transition from Immune-Reactivity to Malignancy. Scand. J. Immunol. 2009, 69, 387–400. [Google Scholar] [CrossRef]
  86. Pflug, N.; Kluth, S.; Vehreschild, J.J.; Bahlo, J.; Tacke, D.; Biehl, L.; Eichhorst, B.; Fischer, K.; Cramer, P.; Fink, A.-M.; et al. Efficacy of Antineoplastic Treatment Is Associated with the Use of Antibiotics That Modulate Intestinal Microbiota. Oncoimmunology 2016, 5, e1150399. [Google Scholar] [CrossRef]
  87. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal Bacteria Control Cancer Response to Therapy by Modulating the Tumor Microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef]
  88. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23-IL-17 Immune Axis: From Mechanisms to Therapeutic Testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef]
  89. Calcinotto, A.; Brevi, A.; Chesi, M.; Ferrarese, R.; Garcia Perez, L.; Grioni, M.; Kumar, S.; Garbitt, V.M.; Sharik, M.E.; Henderson, K.J.; et al. Microbiota-Driven Interleukin-17-Producing Cells and Eosinophils Synergize to Accelerate Multiple Myeloma Progression. Nat. Commun. 2018, 9, 4832. [Google Scholar] [CrossRef]
  90. Peled, J.U.; Devlin, S.M.; Staffas, A.; Lumish, M.; Khanin, R.; Littmann, E.R.; Ling, L.; Kosuri, S.; Maloy, M.; Slingerland, J.B.; et al. Intestinal Microbiota and Relapse After Hematopoietic-Cell Transplantation. J. Clin. Oncol. 2017, 35, 1650–1659. [Google Scholar] [CrossRef] [Green Version]
  91. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the Gut Microbiota in Nutrition and Health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bai, L.; Zhou, P.; Li, D.; Ju, X. Changes in the Gastrointestinal Microbiota of Children with Acute Lymphoblastic Leukaemia and Its Association with Antibiotics in the Short Term. J. Med. Microbiol. 2017, 66, 1297–1307. [Google Scholar] [CrossRef] [PubMed]
  93. Nearing, J.T.; Connors, J.; Whitehouse, S.; Van Limbergen, J.; Macdonald, T.; Kulkarni, K.; Langille, M.G.I. Infectious Complications Are Associated With Alterations in the Gut Microbiome in Pediatric Patients With Acute Lymphoblastic Leukemia. Front. Cell. Infect. Microbiol. 2019, 9, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. van Vliet, M.J.; Tissing, W.J.E.; Dun, C.A.J.; Meessen, N.E.L.; Kamps, W.A.; de Bont, E.S.J.M.; Harmsen, H.J.M. Chemotherapy Treatment in Pediatric Patients with Acute Myeloid Leukemia Receiving Antimicrobial Prophylaxis Leads to a Relative Increase of Colonization with Potentially Pathogenic Bacteria in the Gut. Clin. Infect. Dis. 2009, 49, 262–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kaysen, A.; Heintz-Buschart, A.; Muller, E.E.L.; Narayanasamy, S.; Wampach, L.; Laczny, C.C.; Graf, N.; Simon, A.; Franke, K.; Bittenbring, J.; et al. Integrated Meta-Omic Analyses of the Gastrointestinal Tract Microbiome in Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Transl. Res. 2017, 186, 79–94. [Google Scholar] [CrossRef] [Green Version]
  96. Galloway-Peña, J.R.; Smith, D.P.; Sahasrabhojane, P.; Ajami, N.J.; Wadsworth, W.D.; Daver, N.G.; Chemaly, R.F.; Marsh, L.; Ghantoji, S.S.; Pemmaraju, N.; et al. The Role of the Gastrointestinal Microbiome in Infectious Complications during Induction Chemotherapy for Acute Myeloid Leukemia. Cancer 2016, 122, 2186–2196. [Google Scholar] [CrossRef] [Green Version]
  97. Galloway-Peña, J.R.; Smith, D.P.; Sahasrabhojane, P.; Wadsworth, W.D.; Fellman, B.M.; Ajami, N.J.; Shpall, E.J.; Daver, N.; Guindani, M.; Petrosino, J.F.; et al. Characterization of Oral and Gut Microbiome Temporal Variability in Hospitalized Cancer Patients. Genome Med. 2017, 9, 21. [Google Scholar] [CrossRef] [Green Version]
  98. Kelly, M.S.; Ward, D.V.; Severyn, C.J.; Arshad, M.; Heston, S.M.; Jenkins, K.; Martin, P.L.; McGill, L.; Stokhuyzen, A.; Bhattarai, S.K.; et al. Gut Colonization Preceding Mucosal Barrier Injury Bloodstream Infection in Pediatric Hematopoietic Stem Cell Transplantation Recipients. Biol. Blood Marrow Transpl. 2019, 25, 2274–2280. [Google Scholar] [CrossRef]
  99. Lehouritis, P.; Cummins, J.; Stanton, M.; Murphy, C.T.; McCarthy, F.O.; Reid, G.; Urbaniak, C.; Byrne, W.L.; Tangney, M. Local Bacteria Affect the Efficacy of Chemotherapeutic Drugs. Sci. Rep. 2015, 5, 14554. [Google Scholar] [CrossRef] [Green Version]
  100. Gyurkocza, B.; Rezvani, A.; Storb, R.F. Allogeneic Hematopoietic Cell Transplantation: The State of the Art. Expert Rev. Hematol. 2010, 3, 285–299. [Google Scholar] [CrossRef] [Green Version]
  101. Malard, F.; Huang, X.-J.; Sim, J.P.Y. Treatment and Unmet Needs in Steroid-Refractory Acute Graft-versus-Host Disease. Leukemia 2020, 34, 1229–1240. [Google Scholar] [CrossRef] [PubMed]
  102. DiMaggio, E. Acute Graft-versus-Host Disease: Emerging Insights and Updates into Detection, Prevention, and Treatment. Pharmacotherapy 2020, 40, 788–807. [Google Scholar] [CrossRef] [PubMed]
  103. Filipovich, A.H.; Weisdorf, D.; Pavletic, S.; Socie, G.; Wingard, J.R.; Lee, S.J.; Martin, P.; Chien, J.; Przepiorka, D.; Couriel, D.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. Diagnosis and Staging Working Group Report. Biol. Blood Marrow Transpl. 2005, 11, 945–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Harris, A.C.; Ferrara, J.L.M.; Levine, J.E. Advances in Predicting Acute GVHD. Br. J. Haematol. 2013, 160, 288–302. [Google Scholar] [CrossRef] [PubMed]
  105. Jacobsohn, D.A.; Kurland, B.F.; Pidala, J.; Inamoto, Y.; Chai, X.; Palmer, J.M.; Arai, S.; Arora, M.; Jagasia, M.; Cutler, C.; et al. Correlation between NIH Composite Skin Score, Patient-Reported Skin Score, and Outcome: Results from the Chronic GVHD Consortium. Blood 2012, 120, 2545–2552, quiz 2774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Shulman, H.M.; Cardona, D.M.; Greenson, J.K.; Hingorani, S.; Horn, T.; Huber, E.; Kreft, A.; Longerich, T.; Morton, T.; Myerson, D.; et al. NIH Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: II. The 2014 Pathology Working Group Report. Biol. Blood Marrow Transpl. 2015, 21, 589–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Ferrara, J.L.M.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-Host Disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  108. Zeiser, R.; Blazar, B.R. Acute Graft-versus-Host Disease-Biologic Process, Prevention, and Therapy. N. Engl. J. Med. 2017, 377, 2167–2179. [Google Scholar] [CrossRef]
  109. Martin, P.J.; Inamoto, Y.; Flowers, M.E.D.; Carpenter, P.A. Secondary Treatment of Acute Graft-versus-Host Disease: A Critical Review. Biol. Blood Marrow Transpl. 2012, 18, 982–988. [Google Scholar] [CrossRef] [Green Version]
  110. Taur, Y.; Xavier, J.B.; Lipuma, L.; Ubeda, C.; Goldberg, J.; Gobourne, A.; Lee, Y.J.; Dubin, K.A.; Socci, N.D.; Viale, A.; et al. Intestinal Domination and the Risk of Bacteremia in Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Infect. Dis. 2012, 55, 905–914. [Google Scholar] [CrossRef]
  111. Taur, Y.; Jenq, R.R.; Ubeda, C.; van den Brink, M.; Pamer, E.G. Role of Intestinal Microbiota in Transplantation Outcomes. Best Pract. Res. Clin. Haematol. 2015, 28, 155–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Shono, Y.; van den Brink, M.R.M. Gut Microbiota Injury in Allogeneic Haematopoietic Stem Cell Transplantation. Nat. Rev. Cancer 2018, 18, 283–295. [Google Scholar] [CrossRef] [PubMed]
  113. Connell, M.S.; Wilson, R. The treatment of x-irradiated germfree cfw and c3h mice with isologous and homologous bone marrow. Life Sci. 1965, 4, 721–729. [Google Scholar] [CrossRef]
  114. Jones, J.M.; Wilson, R.; Bealmear, P.M. Mortality and Gross Pathology of Secondary Disease in Germfree Mouse Radiation Chimeras. Radiat. Res. 1971, 45, 577–588. [Google Scholar] [CrossRef] [PubMed]
  115. van Bekkum, D.W.; Roodenburg, J.; Heidt, P.J.; van der Waaij, D. Mitigation of Secondary Disease of Allogeneic Mouse Radiation Chimeras by Modification of the Intestinal Microflora. J. Natl. Cancer Inst. 1974, 52, 401–404. [Google Scholar] [CrossRef]
  116. Bealmear, P.M.; Mirand, E.A.; Holtermann, O.A. Modification of Graft-vs-Host Disease Following Bone Marrow Transplantation in Germfree Mice. Prog. Clin. Biol. Res. 1983, 132C, 409–421. [Google Scholar]
  117. Wade, A.C.; Luckert, P.H.; Tazume, S.; Niedbalski, J.L.; Pollard, M. Characterization of Xenogeneic Mouse-to-Rat Bone Marrow Chimeras. I. Examination of Hematologic and Immunologic Function. Transplantation 1987, 44, 88–92. [Google Scholar] [CrossRef]
  118. Lampert, I.A.; Moore, R.H.; Huby, R.; Cohen, J. Observations on the Role of Endotoxin in Graft-versus-Host Disease. Prog. Clin. Biol. Res. 1988, 272, 351–359. [Google Scholar]
  119. Heidt, P.J.; Vossen, J.M. Experimental and Clinical Gnotobiotics: Influence of the Microflora on Graft-versus-Host Disease after Allogeneic Bone Marrow Transplantation. J. Med. 1992, 23, 161–173. [Google Scholar]
  120. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the Microbiota and the Immune System. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [Green Version]
  121. Round, J.L.; Mazmanian, S.K. The Gut Microbiota Shapes Intestinal Immune Responses during Health and Disease. Nat. Rev. Immunol. 2009, 9, 313–323. [Google Scholar] [CrossRef] [PubMed]
  122. Buckner, C.D.; Clift, R.A.; Sanders, J.E.; Meyers, J.D.; Counts, G.W.; Farewell, V.T.; Thomas, E.D. Protective Environment for Marrow Transplant Recipients: A Prospective Study. Ann. Intern. Med. 1978, 89, 893–901. [Google Scholar] [CrossRef] [PubMed]
  123. Petersen, F.B.; Buckner, C.D.; Clift, R.A.; Nelson, N.; Counts, G.W.; Meyers, J.D.; Thomas, E.D. Prevention of Nosocomial Infections in Marrow Transplant Patients: A Prospective Randomized Comparison of Systemic Antibiotics versus Granulocyte Transfusions. Infect. Control 1986, 7, 586–592. [Google Scholar] [CrossRef] [PubMed]
  124. Petersen, F.B.; Buckner, C.D.; Clift, R.A.; Lee, S.; Nelson, N.; Counts, G.W.; Meyers, J.D.; Sanders, J.E.; Stewart, P.S.; Bensinger, W.I. Laminar Air Flow Isolation and Decontamination: A Prospective Randomized Study of the Effects of Prophylactic Systemic Antibiotics in Bone Marrow Transplant Patients. Infection 1986, 14, 115–121. [Google Scholar] [CrossRef] [PubMed]
  125. Storb, R.; Prentice, R.L.; Buckner, C.D.; Clift, R.A.; Appelbaum, F.; Deeg, J.; Doney, K.; Hansen, J.A.; Mason, M.; Sanders, J.E.; et al. Graft-versus-Host Disease and Survival in Patients with Aplastic Anemia Treated by Marrow Grafts from HLA-Identical Siblings. Beneficial Effect of a Protective Environment. N. Engl. J. Med. 1983, 308, 302–307. [Google Scholar] [CrossRef] [PubMed]
  126. Petersen, F.B.; Buckner, C.D.; Clift, R.A.; Nelson, N.; Counts, G.W.; Meyers, J.D.; Thomas, E.D. Infectious Complications in Patients Undergoing Marrow Transplantation: A Prospective Randomized Study of the Additional Effect of Decontamination and Laminar Air Flow Isolation among Patients Receiving Prophylactic Systemic Antibiotics. Scand. J. Infect. Dis. 1987, 19, 559–567. [Google Scholar] [CrossRef]
  127. Beelen, D.W.; Haralambie, E.; Brandt, H.; Linzenmeier, G.; Müller, K.D.; Quabeck, K.; Sayer, H.G.; Graeven, U.; Mahmoud, H.K.; Schaefer, U.W. Evidence That Sustained Growth Suppression of Intestinal Anaerobic Bacteria Reduces the Risk of Acute Graft-versus-Host Disease after Sibling Marrow Transplantation. Blood 1992, 80, 2668–2676. [Google Scholar] [CrossRef]
  128. 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. [Google Scholar] [CrossRef] [Green Version]
  129. Nishi, K.; Kanda, J.; Hishizawa, M.; Kitano, T.; Kondo, T.; Yamashita, K.; Takaori-Kondo, A. Impact of the Use and Type of Antibiotics on Acute Graft-versus-Host Disease. Biol. Blood Marrow Transpl. 2018, 24, 2178–2183. [Google Scholar] [CrossRef] [Green Version]
  130. Weber, D.; Hiergeist, A.; Weber, M.; Dettmer, K.; Wolff, D.; Hahn, J.; Herr, W.; Gessner, A.; Holler, E. Detrimental Effect of Broad-Spectrum Antibiotics on Intestinal Microbiome Diversity in Patients After Allogeneic Stem Cell Transplantation: Lack of Commensal Sparing Antibiotics. Clin. Infect. Dis. 2019, 68, 1303–1310. [Google Scholar] [CrossRef] [Green Version]
  131. Vossen, J.M.; Heidt, P.J.; van den Berg, H.; Gerritsen, E.J.; Hermans, J.; Dooren, L.J. Prevention of Infection and Graft-versus-Host Disease by Suppression of Intestinal Microflora in Children Treated with Allogeneic Bone Marrow Transplantation. Eur. J. Clin. Microbiol. Infect. Dis. 1990, 9, 14–23. [Google Scholar] [CrossRef] [PubMed]
  132. Beelen, D.W.; Elmaagacli, A.; Müller, K.D.; Hirche, H.; Schaefer, U.W. Influence of Intestinal Bacterial Decontamination Using Metronidazole and Ciprofloxacin or Ciprofloxacin Alone on the Development of Acute Graft-versus-Host Disease after Marrow Transplantation in Patients with Hematologic Malignancies: Final Results and Long-Term Follow-up of an Open-Label Prospective Randomized Trial. Blood 1999, 93, 3267–3275. [Google Scholar] [PubMed]
  133. 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 Transpl. 2016, 51, 1087–1092. [Google Scholar] [CrossRef] [PubMed]
  134. Köhler, N.; Zeiser, R. Intestinal Microbiota Influence Immune Tolerance Post Allogeneic Hematopoietic Cell Transplantation and Intestinal GVHD. Front Immunol 2018, 9, 3179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. 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. [Google Scholar] [CrossRef] [PubMed]
  136. 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 Transpl. 2015, 21, 1373–1383. [Google Scholar] [CrossRef] [Green Version]
  137. Golob, J.L.; Pergam, S.A.; Srinivasan, S.; Fiedler, T.L.; Liu, C.; Garcia, K.; Mielcarek, M.; Ko, D.; Aker, S.; Marquis, 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, 1984–1991. [Google Scholar] [CrossRef]
  138. Peled, J.U.; Gomes, A.L.C.; 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. [Google Scholar] [CrossRef]
  139. Gavriilaki, M.; Sakellari, I.; Anagnostopoulos, A.; Gavriilaki, E. The Impact of Antibiotic-Mediated Modification of the Intestinal Microbiome on Outcomes of Allogeneic Hematopoietic Cell Transplantation: Systematic Review and Meta-Analysis. Biol. Blood Marrow Transpl. 2020, 26, 1738–1746. [Google Scholar] [CrossRef]
  140. Jenq, R.R.; Ubeda, C.; Taur, Y.; Menezes, C.C.; Khanin, R.; Dudakov, J.A.; Liu, C.; West, M.L.; Singer, N.V.; Equinda, M.J.; et al. Regulation of Intestinal Inflammation by Microbiota Following Allogeneic Bone Marrow Transplantation. J. Exp. Med. 2012, 209, 903–911. [Google Scholar] [CrossRef]
  141. Fredricks, D.N. The Gut Microbiota and Graft-versus-Host Disease. J. Clin. Invest. 2019, 129, 1808–1817. [Google Scholar] [CrossRef] [Green Version]
  142. Gorshein, E.; Wei, C.; Ambrosy, S.; Budney, S.; Vivas, J.; Shenkerman, A.; Manago, J.; McGrath, M.K.; Tyno, A.; Lin, Y.; et al. Lactobacillus Rhamnosus GG Probiotic Enteric Regimen Does Not Appreciably Alter the Gut Microbiome or Provide Protection against GVHD after Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Transpl. 2017, 31. [Google Scholar] [CrossRef] [PubMed]
  143. Biagi, E.; Zama, D.; Nastasi, C.; Consolandi, C.; Fiori, J.; Rampelli, S.; Turroni, S.; Centanni, M.; Severgnini, M.; Peano, C.; et al. Gut Microbiota Trajectory in Pediatric Patients Undergoing Hematopoietic SCT. Bone Marrow Transpl. 2015, 50, 992–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Laterza, L.; Rizzatti, G.; Gaetani, E.; Chiusolo, P.; Gasbarrini, A. The Gut Microbiota and Immune System Relationship in Human Graft-versus-Host Disease. Mediterr. J. Hematol. Infect. Dis. 2016, 8, e2016025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. 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 Transpl. 2014, 20, 640–645. [Google Scholar] [CrossRef] [Green Version]
  146. Stein-Thoeringer, C.; Peled, J.U.; Lazrak, A.; Slingerland, A.E.; Shono, Y.; Staffas, A.; Gomes, A.; Argyropoulos, K.; Docampo, M.; Xavier, J.; et al. 20-Domination of the Gut Microbiota with Enterococcus Species Early after Allogeneic Bone Marrow Transplantation Is an Important Contributor to the Development of Acute Graft-Versus-Host Disease (GHVD) in Mouse and Man. Biol. Blood Marrow Transpl. 2018, 24, S40–S41. [Google Scholar] [CrossRef] [Green Version]
  147. Eriguchi, Y.; Takashima, S.; Oka, H.; Shimoji, S.; Nakamura, K.; Uryu, H.; Shimoda, S.; Iwasaki, H.; Shimono, N.; Ayabe, T.; et al. Graft-versus-Host Disease Disrupts Intestinal Microbial Ecology by Inhibiting Paneth Cell Production of α-Defensins. Blood 2012, 120, 223–231. [Google Scholar] [CrossRef] [Green Version]
  148. Heimesaat, M.M.; Nogai, A.; Bereswill, S.; Plickert, R.; Fischer, A.; Loddenkemper, C.; Steinhoff, U.; Tchaptchet, S.; Thiel, E.; Freudenberg, M.A.; et al. MyD88/TLR9 Mediated Immunopathology and Gut Microbiota Dynamics in a Novel Murine Model of Intestinal Graft-versus-Host Disease. Gut 2010, 59, 1079–1087. [Google Scholar] [CrossRef]
  149. 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. [Google Scholar] [CrossRef]
  150. Simms-Waldrip, T.R.; Sunkersett, G.; Coughlin, L.A.; Savani, M.R.; Arana, C.; Kim, J.; Kim, M.; Zhan, X.; Greenberg, D.E.; Xie, Y.; 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 Transpl. 2017, 23, 820–829. [Google Scholar] [CrossRef] [Green Version]
  151. Zwielehner, J.; Lassl, C.; Hippe, B.; Pointner, A.; Switzeny, O.J.; Remely, M.; Kitzweger, E.; Ruckser, R.; Haslberger, A.G. Changes in Human Fecal Microbiota Due to Chemotherapy Analyzed by TaqMan-PCR, 454 Sequencing and PCR-DGGE Fingerprinting. PLoS ONE 2011, 6, e28654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. 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. [Google Scholar] [CrossRef] [PubMed]
  153. Schluter, J.; Peled, J.U.; Taylor, B.P.; Markey, K.A.; Smith, M.; Taur, Y.; Niehus, R.; Staffas, A.; Dai, A.; Fontana, E.; et al. The Gut Microbiota Is Associated with Immune Cell Dynamics in Humans. Nature 2020, 588, 303–307. [Google Scholar] [CrossRef] [PubMed]
  154. Swimm, A.; Giver, C.R.; DeFilipp, Z.; Rangaraju, S.; Sharma, A.; Ulezko Antonova, A.; Sonowal, R.; Capaldo, C.; Powell, D.; Qayed, M.; et al. Indoles Derived from Intestinal Microbiota Act via Type I Interferon Signaling to Limit Graft-versus-Host Disease. Blood 2018, 132, 2506–2519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Weber, D.; Oefner, P.J.; Hiergeist, A.; Koestler, J.; Gessner, A.; Weber, M.; Hahn, J.; Wolff, D.; Stämmler, F.; Spang, R.; et al. Low Urinary Indoxyl Sulfate Levels Early after Transplantation Reflect a Disrupted Microbiome and Are Associated with Poor Outcome. Blood 2015, 126, 1723–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Mathewson, N.D.; Jenq, R.; Mathew, A.V.; Koenigsknecht, M.; Hanash, A.; Toubai, T.; Oravecz-Wilson, K.; Wu, S.-R.; Sun, Y.; Rossi, C.; et al. Gut Microbiome-Derived Metabolites Modulate Intestinal Epithelial Cell Damage and Mitigate Graft-versus-Host Disease. Nat. Immunol. 2016, 17, 505–513. [Google Scholar] [CrossRef]
  157. Levine, J.E.; Huber, E.; Hammer, S.T.G.; Harris, A.C.; Greenson, J.K.; Braun, T.M.; Ferrara, J.L.M.; Holler, E. Low Paneth Cell Numbers at Onset of Gastrointestinal Graft-versus-Host Disease Identify Patients at High Risk for Nonrelapse Mortality. Blood 2013, 122, 1505–1509. [Google Scholar] [CrossRef]
  158. Ubeda, C.; Taur, Y.; Jenq, R.R.; Equinda, M.J.; Son, T.; Samstein, M.; Viale, A.; Socci, N.D.; van den Brink, M.R.M.; Kamboj, M.; et al. Vancomycin-Resistant Enterococcus Domination of Intestinal Microbiota Is Enabled by Antibiotic Treatment in Mice and Precedes Bloodstream Invasion in Humans. J. Clin. Invest. 2010, 120, 4332–4341. [Google Scholar] [CrossRef]
  159. Ubeda, C.; Bucci, V.; Caballero, S.; Djukovic, A.; Toussaint, N.C.; Equinda, M.; Lipuma, L.; Ling, L.; Gobourne, A.; No, D.; et al. Intestinal Microbiota Containing Barnesiella Species Cures Vancomycin-Resistant Enterococcus Faecium Colonization. Infect. Immun. 2013, 81, 965–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Legoff, J.; Resche-Rigon, M.; Bouquet, J.; Robin, M.; Naccache, S.N.; Mercier-Delarue, S.; Federman, S.; Samayoa, E.; Rousseau, C.; Piron, P.; et al. The Eukaryotic Gut Virome in Hematopoietic Stem Cell Transplantation: New Clues in Enteric Graft-versus-Host Disease. Nat. Med. 2017, 23, 1080–1085. [Google Scholar] [CrossRef]
  161. Laheij, A.M.G.A.; Raber-Durlacher, J.E.; Koppelmans, R.G.A.; Huysmans, M.-C.D.N.J.M.; Potting, C.; van Leeuwen, S.J.M.; Hazenberg, M.D.; Brennan, M.T.; von Bültzingslöwen, I.; Johansson, J.-E.; et al. Microbial Changes in Relation to Oral Mucositis in Autologous Hematopoietic Stem Cell Transplantation Recipients. Sci. Rep. 2019, 9, 16929. [Google Scholar] [CrossRef] [PubMed]
  162. Galloway-Peña, J.R.; Shi, Y.; Peterson, C.B.; Sahasrabhojane, P.; Gopalakrishnan, V.; Brumlow, C.E.; Daver, N.G.; Alfayez, M.; Boddu, P.C.; Khan, M.A.W.; et al. Gut Microbiome Signatures Are Predictive of Infectious Risk Following Induction Therapy for Acute Myeloid Leukemia. Clin. Infect. Dis. 2020, 71, 63–71. [Google Scholar] [CrossRef] [PubMed]
  163. Pianko, M.J.; Devlin, S.M.; Littmann, E.R.; Chansakul, A.; Mastey, D.; Salcedo, M.; Fontana, E.; Ling, L.; Tavitian, E.; Slingerland, J.B.; et al. Minimal Residual Disease Negativity in Multiple Myeloma Is Associated with Intestinal Microbiota Composition. Blood Adv. 2019, 3, 2040–2044. [Google Scholar] [CrossRef] [PubMed]
  164. Smith, M.; Littmann, E.; Slingerland, J.; Clurman, A.; Slingerland, A.E.; Taur, Y.; Peled, J.U.; Park, J.; O’Cearbhaill, R.; Mailankody, S.; et al. Intestinal Microbiome Analyses Identify Biomarkers for Patient Response to CAR T Cell Therapy. Biol. Blood Marrow Transpl. 2019, 25, S177. [Google Scholar] [CrossRef]
  165. Muraro, P.A.; Martin, R.; Mancardi, G.L.; Nicholas, R.; Sormani, M.P.; Saccardi, R. Autologous Haematopoietic Stem Cell Transplantation for Treatment of Multiple Sclerosis. Nat. Rev. Neurol. 2017, 13, 391–405. [Google Scholar] [CrossRef]
  166. Passweg, J.R.; Baldomero, H.; Chabannon, C.; Basak, G.W.; Corbacioglu, S.; Duarte, R.; Dolstra, H.; Lankester, A.C.; Mohty, M.; Montoto, S.; et al. The EBMT Activity Survey on Hematopoietic-Cell Transplantation and Cellular Therapy 2018: CAR-T’s Come into Focus. Bone Marrow Transpl. 2020, 55, 1604–1613. [Google Scholar] [CrossRef] [Green Version]
  167. Zahid, U.; Akbar, F.; Amaraneni, A.; Husnain, M.; Chan, O.; Riaz, I.B.; McBride, A.; Iftikhar, A.; Anwer, F. A Review of Autologous Stem Cell Transplantation in Lymphoma. Curr. Hematol. Malig. Rep. 2017, 12, 217–226. [Google Scholar] [CrossRef]
  168. Al Hamed, R.; Bazarbachi, A.H.; Malard, F.; Harousseau, J.-L.; Mohty, M. Current Status of Autologous Stem Cell Transplantation for Multiple Myeloma. Blood Cancer J. 2019, 9, 44. [Google Scholar] [CrossRef] [Green Version]
  169. El Jurdi, N.; Filali-Mouhim, A.; Salem, I.; Retuerto, M.; Dambrosio, N.M.; Baer, L.; Lazarus, H.M.; Caimi, P.; Cooper, B.; Tomlinson, B.; et al. Gastrointestinal Microbiome and Mycobiome Changes during Autologous Transplantation for Multiple Myeloma: Results of a Prospective Pilot Study. Biol Blood Marrow Transpl. 2019, 25, 1511–1519. [Google Scholar] [CrossRef]
  170. Kusakabe, S.; Fukushima, K.; Maeda, T.; Motooka, D.; Nakamura, S.; Fujita, J.; Yokota, T.; Shibayama, H.; Oritani, K.; Kanakura, Y. 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. [Google Scholar] [CrossRef]
  171. Montassier, E.; Gastinne, T.; Vangay, P.; Al-Ghalith, G.A.; Bruley des Varannes, S.; Massart, S.; Moreau, P.; Potel, G.; de La Cochetière, M.F.; Batard, E.; et al. Chemotherapy-Driven Dysbiosis in the Intestinal Microbiome. Aliment. Pharmacol. Ther. 2015, 42, 515–528. [Google Scholar] [CrossRef] [PubMed]
  172. Gutiérrez-García, G.; Rovira, M.; Magnano, L.; Rosiñol, L.; Bataller, A.; Suárez-Lledó, M.; Cibeira, M.T.; de Larrea, C.F.; Garrote, M.; Jorge, S.; et al. Innovative Strategies Minimize Engraftment Syndrome in Multiple Myeloma Patients with Novel Induction Therapy Following Autologous Hematopoietic Stem Cell Transplantation. Bone Marrow Transpl. 2018, 53, 1541–1547. [Google Scholar] [CrossRef]
  173. Ziegler, M.; Han, J.H.; Landsburg, D.; Pegues, D.; Reesey, E.; Gilmar, C.; Gorman, T.; Bink, K.; Moore, A.; Kelly, B.J. Impact of Levofloxacin for the Prophylaxis of Bloodstream Infection on the Gut Microbiome in Patients With Hematologic Malignancy. Open Forum Infect. Dis. 2019, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kristinsson, S.Y.; Anderson, W.F.; Landgren, O. Improved Long-Term Survival in Multiple Myeloma up to the Age of 80 Years. Leukemia 2014, 28, 1346–1348. [Google Scholar] [CrossRef] [PubMed]
  175. Kumar, S.K.; Dispenzieri, A.; Lacy, M.Q.; Gertz, M.A.; Buadi, F.K.; Pandey, S.; Kapoor, P.; Dingli, D.; Hayman, S.R.; Leung, N.; et al. Continued Improvement in Survival in Multiple Myeloma: Changes in Early Mortality and Outcomes in Older Patients. Leukemia 2014, 28, 1122–1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Thorsteinsdottir, S.; Dickman, P.W.; Landgren, O.; Blimark, C.; Hultcrantz, M.; Turesson, I.; Björkholm, M.; Kristinsson, S.Y. Dramatically Improved Survival in Multiple Myeloma Patients in the Recent Decade: Results from a Swedish Population-Based Study. Haematologica 2018, 103, e412–e415. [Google Scholar] [CrossRef] [Green Version]
  177. June, C.H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379, 64–73. [Google Scholar] [CrossRef]
  178. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  179. Schuster, S.J.; Svoboda, J.; Chong, E.A.; Nasta, S.D.; Mato, A.R.; Anak, Ö.; Brogdon, J.L.; Pruteanu-Malinici, I.; Bhoj, V.; Landsburg, D.; et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N. Engl. J. Med. 2017, 377, 2545–2554. [Google Scholar] [CrossRef]
  180. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  181. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  182. Rodríguez-Lobato, L.G.; Ganzetti, M.; Fernández de Larrea, C.; Hudecek, M.; Einsele, H.; Danhof, S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front. Oncol. 2020, 10, 1243. [Google Scholar] [CrossRef] [PubMed]
  183. Gagelmann, N.; Riecken, K.; Wolschke, C.; Berger, C.; Ayuk, F.A.; Fehse, B.; Kröger, N. Development of CAR-T Cell Therapies for Multiple Myeloma. Leukemia 2020, 34, 2317–2332. [Google Scholar] [CrossRef] [PubMed]
  184. Sonnenburg, J.L.; Bäckhed, F. Diet-Microbiota Interactions as Moderators of Human Metabolism. Nature 2016, 535, 56–64. [Google Scholar] [CrossRef] [PubMed]
  185. Thaiss, C.A.; Zeevi, D.; Levy, M.; Zilberman-Schapira, G.; Suez, J.; Tengeler, A.C.; Abramson, L.; Katz, M.N.; Korem, T.; Zmora, N.; et al. Transkingdom Control of Microbiota Diurnal Oscillations Promotes Metabolic Homeostasis. Cell 2014, 159, 514–529. [Google Scholar] [CrossRef] [Green Version]
  186. Barbaresko, J.; Koch, M.; Schulze, M.B.; Nöthlings, U. Dietary Pattern Analysis and Biomarkers of Low-Grade Inflammation: A Systematic Literature Review. Nutr. Rev. 2013, 71, 511–527. [Google Scholar] [CrossRef]
  187. Solans, M.; Castelló, A.; Benavente, Y.; Marcos-Gragera, R.; Amiano, P.; Gracia-Lavedan, E.; Costas, L.; Robles, C.; Gonzalez-Barca, E.; de la Banda, E.; et al. Adherence to the Western, Prudent, and Mediterranean Dietary Patterns and Chronic Lymphocytic Leukemia in the MCC-Spain Study. Haematologica 2018, 103, 1881–1888. [Google Scholar] [CrossRef]
  188. University of Michigan Rogel Cancer Center. Dietary Manipulation of the Microbiome-Metabolomic Axis for Mitigating GVHD in Allo HCT Patients. 2019. Available online: clinicaltrials.gov (accessed on 17 November 2020).
  189. Monteagudo-Mera, A.; Rastall, R.A.; Gibson, G.R.; Charalampopoulos, D.; Chatzifragkou, A. Adhesion Mechanisms Mediated by Probiotics and Prebiotics and Their Potential Impact on Human Health. Appl. Microbiol. Biotechnol. 2019, 103, 6463–6472. [Google Scholar] [CrossRef] [Green Version]
  190. Shoaf, K.; Mulvey, G.L.; Armstrong, G.D.; Hutkins, R.W. Prebiotic Galactooligosaccharides Reduce Adherence of Enteropathogenic Escherichia Coli to Tissue Culture Cells. Infect. Immun. 2006, 74, 6920–6928. [Google Scholar] [CrossRef] [Green Version]
  191. Fructooligosaccharides in Treating Patients With Blood Cancer Undergoing Donor Stem Cell Transplant-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02805075 (accessed on 17 November 2020).
  192. Duke University. A Phase I/II Trial of the Prebiotic Galacto-Oligosaccharide to Prevent Acute GVHD. 2020. Available online: clinicaltrials.gov (accessed on 17 November 2020).
  193. Maga, E.A.; Desai, P.T.; Weimer, B.C.; Dao, N.; Kültz, D.; Murray, J.D. Consumption of Lysozyme-Rich Milk Can Alter Microbial Fecal Populations. Appl. Environ. Microbiol. 2012, 78, 6153–6160. [Google Scholar] [CrossRef] [Green Version]
  194. Ko, K.Y.; Mendoncam, A.F.; Ismail, H.; Ahn, D.U. Ethylenediaminetetraacetate and Lysozyme Improves Antimicrobial Activities of Ovotransferrin against Escherichia Coli O157:H7. Poult. Sci. 2009, 88, 406–414. [Google Scholar] [CrossRef]
  195. Human Lysozyme Goat Milk for the Prevention of Graft Versus Host Disease in Patients With Blood Cancer Undergoing a Donor Stem Cell Transplant-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04177004 (accessed on 24 November 2020).
  196. Mills, J.P.; Rao, K.; Young, V.B. Probiotics for Prevention of Clostridium Difficile Infection. Curr. Opin. Gastroenterol. 2018, 34, 3–10. [Google Scholar] [CrossRef] [PubMed]
  197. Klaenhammer, T.R.; Kleerebezem, M.; Kopp, M.V.; Rescigno, M. The Impact of Probiotics and Prebiotics on the Immune System. Nat. Rev. Immunol. 2012, 12, 728–734. [Google Scholar] [CrossRef] [PubMed]
  198. CBM588 in Improving Clinical Outcomes in Patients Who Have Undergone Donor Hematopoietic Stem Cell Transplant-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03922035 (accessed on 24 November 2020).
  199. Wang, Y.; Wiesnoski, D.H.; Helmink, B.A.; Gopalakrishnan, V.; Choi, K.; DuPont, H.L.; Jiang, Z.-D.; Abu-Sbeih, H.; Sanchez, C.A.; Chang, C.-C.; et al. Fecal Microbiota Transplantation for Refractory Immune Checkpoint Inhibitor-Associated Colitis. Nat. Med. 2018, 24, 1804. [Google Scholar] [CrossRef] [PubMed]
  200. Jonsson Comprehensive Cancer Center An Open-Label Phase 1 Pilot Study: Fecal Microbiota Transplantation (FMT) in Severe Acute Gut Graft-Versus-Host Disease Patients. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04280471 (accessed on 24 November 2020).
  201. Medical University of Graz Fecal Microbiota Transplantation in Patients With Acute Gastrointestinal Graft-versus-Host-Disease After Allogeneic Stem Cell Transplantation. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  202. Metheny, L. Fecal Microbiota Transplantation For The Treatment Of Gastro-Intestinal Acute Graft Versus Host Disease. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  203. Moiseev, I.S. Pilot Study of Fecal Microbiota Transplantation in Combination With Ruxolitinib and Steroids for Severe Acute Intestinal Graft-versus-Host-Disease After Allogeneic Hematopoietic Stem Cell Transplantation. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  204. Qing, C. Fecal Microbiota Transplantation for Treatment of Refractory Graft Versus Host Disease—A Pilot Study. 2019. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  205. Wan, L. Fecal Microbiota Transplantation for the Treatment of Steroid Resistant/Dependent Acute Gastrointestinal Graft Versus Host Disease. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  206. Liu, Q. Investigator-Initiated Trial to Explore the Efficacy and Safety of Fecal Microbiota Transplant (FMT) Capsule in the Treatment of Steroid-Refractory Gastrointestinal Acute Graft Versus Host Disease Patients. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  207. FMT In High-Risk Acute GVHD After ALLO HCT-Full Text-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04139577 (accessed on 17 November 2020).
  208. FMT for Steroid Resistant Gut Acute GVHD-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04285424 (accessed on 17 November 2020).
  209. Masonic Cancer Center, University of Minnesota A Randomized Placebo-Controlled Clinical Trial of Fecal Microbiota Transplantation in Patients With Acute Myeloid Leukemia and Allogeneic Hematopoietic Cell Transplantation Recipients. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  210. MaaT Pharma PreventiOn of DYSbioSis Complications With Autologous Fecal Microbiota Transplantation in AcutE MyEloid Leukemia Patients Undergoing Intensive Treatment: A Feasibility and Safety Study ODYSSEE STUDY. 2020. Available online: clinicaltrials.gov (accessed on 24 November 2020).
  211. Kao, D.; Roach, B.; Silva, M.; Beck, P.; Rioux, K.; Kaplan, G.G.; Chang, H.-J.; Coward, S.; Goodman, K.J.; Xu, H.; et al. Effect of Oral Capsule- vs Colonoscopy-Delivered Fecal Microbiota Transplantation on Recurrent Clostridium Difficile Infection: A Randomized Clinical Trial. JAMA 2017, 318, 1985–1993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. DeFilipp, Z.; Bloom, P.P.; Torres Soto, M.; Mansour, M.K.; Sater, M.R.A.; Huntley, M.H.; Turbett, S.; Chung, R.T.; Chen, Y.-B.; Hohmann, E.L. Drug-Resistant E. Coli Bacteremia Transmitted by Fecal Microbiota Transplant. N. Engl. J. Med. 2019, 381, 2043–2050. [Google Scholar] [CrossRef]
  213. FDA. Fecal Microbiota for Transplantation: Safety Alert-Risk of Serious Adverse Events Likely Due to Transmission of Pathogenic Organisms. 2020. Available online: https://www.fda.gov/safety/medical-product-safety-information/fecal-microbiota-transplantation-safety-alert-risk-serious-adverse-events-likely-due-transmission (accessed on 24 November 2020).
  214. Markey, K.A.; Schluter, J.; Gomes, A.L.C.; Littmann, E.R.; Pickard, A.J.; Taylor, B.P.; Giardina, P.A.; Weber, D.; Dai, A.; Docampo, M.D.; et al. The Microbe-Derived Short-Chain Fatty Acids Butyrate and Propionate Are Associated with Protection from Chronic GVHD. Blood 2020, 136, 130–136. [Google Scholar] [CrossRef]
  215. Wu, K.; Yuan, Y.; Yu, H.; Dai, X.; Wang, S.; Sun, Z.; Wang, F.; Fei, H.; Lin, Q.; Jiang, H.; et al. The Gut Microbial Metabolite Trimethylamine N-Oxide Aggravates GVHD by Inducing M1 Macrophage Polarization in Mice. Blood 2020, 136, 501–515. [Google Scholar] [CrossRef]
  216. Optimization of Antibiotic Treatment in Hematopoietic Stem Cell Receptors-Tabular View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/record/NCT03727113 (accessed on 18 November 2020).
Figure 1. Dynamic interactions between the gut microbiota and the immune system.
Figure 1. Dynamic interactions between the gut microbiota and the immune system.
Ijms 22 01026 g001
Figure 2. Mechanisms of gut microbiota modulation, consequences, and potential solutions.
Figure 2. Mechanisms of gut microbiota modulation, consequences, and potential solutions.
Ijms 22 01026 g002
Table 1. Summary of the potential effects of gut microbiota on anticancer agents used in clinical hematology.
Table 1. Summary of the potential effects of gut microbiota on anticancer agents used in clinical hematology.
MechanismOutcomeInvolved Drug (s)
Enzymatic modification of the drugsDecreased levelsCladribine, gemcitabine, doxorubicin, Idarubicin, etoposide, mitoxantrone [99]
Increased activityFludarabine, 6-Mercaptopurine [99]
Translocation of Gram-positive bacteriaGeneration of Th17 and Th1 lymphocytesCyclophosphamide [74]
Modulation of genotoxicityReduction of DNA damage and apoptosisOxaliplatin, cisplatin [87]
Table 2. Summary of outcomes induced by type of treatment and microbiota disruption in humans.
Table 2. Summary of outcomes induced by type of treatment and microbiota disruption in humans.
TreatmentMicrobiota FeatureOutcome
Allogeneic hematopoietic stem cell transplantationDecontamination of gut anaerobesLower risk of GvHD [127,132]
Decreased duodenal Paneth cellsHigher GI GvHD and NRM [148]
Low intestinal microbiota diversityHigher TRM, lower OS and GvHD-related mortality [135,138]
Enterococcus spp. dominationIncreased GI GvHD severity [145] Increased GvHD-related and overall mortality [146,157] Associated with blood stream infections [110,158]
Blautia abundanceReduced GvHD-related mortality [136]
Clostridia spp. depletionIncreased GvHD [150]
Barnesiella spp. abundanceProtection against Enterococcus domination [159]
Akkermansia muciniphila dominationMucus degradation [128]
Lactobacillales dominationAssociated with GvHD development [140]
Eubacterium limosum abundanceLower risk of relapse or progression, higher OS [90]
Picobirnivirus presenceSevere GI GvHD [160]
Lower urinary 3-indoxyl sulfateHigher risk of GvHD, higher TRM, lower OS, higher dysbiosis [145,155]
Autologous stem cell transplantationReduction in diversity index and an increased dominance indexDevelopment of mucositis [161]
Decreased in Firmicutes and Actinobacteria and increased in ProteobacteriaDevelopment of GI mucositis [161]
ChemotherapyBaseline levels of Porphyromonadaceae Predictor of infection during induction for acute myeloid leukemia [162]
Relative abundance of E hallii Higher negative minimal residual disease rate in bone marrow for multiple myeloma [163]
CAR T-cell therapy Oscillospiraceae, Ruminococcacaeae and Lachnospiraceae enriched Association with complete remission after CAR T cell therapy [164]
Higher abundance of LachnospiraceaeDevelopment of cytokine release syndrome and/or neurotoxicity [164]
GI gastrointestinal; GvHD graft versus host disease; NRM no-relapse mortality; OS overall survival; TRM, transplant-related mortality.
Table 3. Interventional clinical trials employing microbial products of hematological malignancies treatment and its complications.
Table 3. Interventional clinical trials employing microbial products of hematological malignancies treatment and its complications.
ConditionInterventionAdministrationPrimary AimPhaseParticipantsStatusIdentifier
aGvHDFMTOrally via capsuleEfficacy, safety, and tolerabilityI10NYRNCT04280471
GI aGvHDFMTColonoscopy or gastroscopyEfficacy and safetyII30RNCT03812705
GI aGvHDFMTNasojejunal tubeEfficacy and safetyI15NYRNCT03549676
GI aGvHDFMTOrally via capsuleEfficacy and safetyI/II20RNCT04269850
GI aGvHDFMTOrally via capsuleEfficacy and safetyII17NYRNCT04059757
GI aGvHDFMTColonoscopy or gastroscopyEfficacyIII15RNCT03819803
GI aGvHDFMTOrally via capsuleEfficacy and safetyN/A10RNCT04622475
aGvHDFMTOrally via capsuleFeasibility and efficacyI11NYRNCT04139577
GI aGvHDFMTColonoscopy or duodenal nutrition tube injectionEfficacyI30RNCT04285424
Allo-HSCT, AMLFMTOrally via capsuleEfficacy and incidence of infectionsII120RNCT03678493
AMLFMTRectal enemaEvaluation in dysbiosis correction and in MDRB eradicationI/II20CNCT02928523
aGvHDFMTNot specifiedEfficacyII24CNCT03359980
Allo-HSCTFMTOrally via capsuleFeasibility and efficacyI18CNCT02733744
Allo-HSCTPotato-based dietary starchOrallyIncidence of grade II-IV GVHDII70RNCT02763033
Allo-HSCTFructo-oligosaccharides prebioticOrallySafety and tolerability I15CNCT02805075
Allo-HSCTGalacto-oligosaccharide prebioticOrallyTolerability and incidence of Grade II-IV aGVHDI/II128NYRNCT04373057
Allo-HSCTHuman lysozyme goat milk prebioticOrallySafety and tolerabilityI36NYRNCT04177004
Allo-HSCTClostridium butyricum CBM 588 Probiotic StrainOrallySafetyI36RNCT03922035
AML acute myeloid leukemia; Allo-HSCT allogeneic hematopoietic stem cell transplant; FMT fecal microbiota transplant; GI gastrointestinal; GvHD graft versus host disease; HSCT hematopoietic stem cell transplant; MDRB multidrug resistant bacteria; NYR, Not yet recruiting; R recruiting; C completed.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Uribe-Herranz, M.; Klein-González, N.; Rodríguez-Lobato, L.G.; Juan, M.; Fernández de Larrea, C. Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure. Int. J. Mol. Sci. 2021, 22, 1026. https://doi.org/10.3390/ijms22031026

AMA Style

Uribe-Herranz M, Klein-González N, Rodríguez-Lobato LG, Juan M, Fernández de Larrea C. Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure. International Journal of Molecular Sciences. 2021; 22(3):1026. https://doi.org/10.3390/ijms22031026

Chicago/Turabian Style

Uribe-Herranz, Mireia, Nela Klein-González, Luis Gerardo Rodríguez-Lobato, Manel Juan, and Carlos Fernández de Larrea. 2021. "Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure" International Journal of Molecular Sciences 22, no. 3: 1026. https://doi.org/10.3390/ijms22031026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop