Next Article in Journal
Neurobehavioral and Oxidative Stress Effects of SiO2 Nanoparticles in Zebrafish and the Protective Role of N-Acetylcysteine
Previous Article in Journal
Small Interfering RNAs Targeting VP4, VP3, 2B, or 3A Coding Regions of Enterovirus A71 Inhibit Viral Replication In Vitro
Previous Article in Special Issue
Metagenomic Microbial Signatures for Noninvasive Detection of Pancreatic Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine

by
Aneta Nowicka
1,*,
Hanna Tomczak
2,
Edyta Szałek
3,
Agnieszka Karbownik
3 and
Lidia Gil
1
1
Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, 60-569 Poznan, Poland
2
Faculty of Medicine and Health Sciences, Calisia University, 62-800 Kalisz, Poland
3
Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 60-806 Poznan, Poland
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(7), 1761; https://doi.org/10.3390/biomedicines13071761
Submission received: 7 June 2025 / Revised: 13 July 2025 / Accepted: 16 July 2025 / Published: 18 July 2025
(This article belongs to the Collection Feature Papers in Microbiology in Human Health and Disease)

Abstract

Increasing evidence demonstrates the mutualistic connection between the microbiome and acute myeloid leukemia (AML) treatment. Drugs disrupt the microbial balance and, conversely, changes in the microbiome influence therapy. A new field, pharmacomicrobiomics, examines the role of the microbiome in pharmacokinetics, pharmacodynamics, and drug toxicity. The multimodal therapeutic management of AML, along with disease-related immunosuppression, infection, and malnutrition, creates the unique microbial profile of AML patients, in which every delicate modification plays a crucial role in pharmacotherapy. While both preclinical and real-world data have confirmed a bilateral connection between standard chemotherapy and the microbiome, the impact of novel targeted therapies and immunotherapy remains unknown. Multi-omics technologies have provided qualitative and mechanistic insights into specific compositional and functional microbial signatures associated with the outcomes of AML therapy, but require a large-scale investigation to draw reliable conclusions. In this review, we outline the role of the microbiome within the therapeutic landscape of AML, focusing on the determinants of post-treatment dysbiosis and its effects on the therapeutic response and toxicity. We explore emerging strategies for microbiota modulation, highlighting their safety and efficacy. Advances in microbiome-based approaches are an inevitable step toward precision medicine in AML. However, clinical research in a well-defined group of immunocompromised patients is needed to study their variable effects on human health and determine safety issues.

1. Introduction

Millions of microorganisms inhabiting the human body and their genomes, collectively known as the microbiome, create a functional ecosystem that maintains a mutualistic balance with the host. Cumulative exposures across the lifespan—including diet, medication, environment, and aging—sculpt a unique microbial fingerprint in each individual. Although generally stable, the microbiome can undergo lasting alterations in its taxonomic composition and reduced diversity under specific conditions, potentially generating disease-associated microbial patterns with biomarker potential. Recent advancements in genomics, metagenomics, and metabolomics emphasize the microbiome’s crucial role in immunity, hormones, inflammation, metabolism, intestinal barrier function, and hematopoiesis [1,2]. A microbiome imbalance, referred to as dysbiosis, can influence the onset, progression, and response to the therapy of cancers, including acute myeloid leukemia (AML) [3]. Mechanistically, it not only influences mutagenesis, cell proliferation, and microenvironmental immunomodulation locally [4], but also provides bioactive metabolites that exert systemic health effects [5]. As the understanding of the cancer biology expands, researchers increasingly recognize the polymorphic variability of the microbiome as essential to the core hallmarks of cancer and, therefore, as a promising therapeutic target [6].
Given the significance of the microbiome in health and disease, its effect on the disposition and drug response has garnered significant attention, which is reflected in the increasing number of scientific papers in the field (51,913 publications were reported for search terms “microbiome and therapy” in PubMed.gov as of 30 May 2025). To address this knowledge gap, the emerging field of pharmacomicrobiomics has investigated how the microbiome affects drug pharmacokinetics, pharmacodynamics, and toxicity [7]. Two additional terms further refine this field: toxicomicrobiomics, which examines the microbiome’s impact on drug metabolism and toxicity, and pharmacoecology, which refers to changes in the microbiome following drug administration [8]. In cancer patients, these interactions are especially relevant as they may critically influence treatment outcomes.
AML remains one of the most challenging blood cancers, with a 5-year overall survival (OS) of 30% and a 50% rate of relapse after achieving complete remission (CR) [9]. Originating from somatic genetic and epigenetic alterations in immature myeloid progenitor cells in the bone marrow, it progresses aggressively to create subclonal populations with a high potential for resistance to treatment [10]. While extensive research has elucidated the genetic landscape of AML, we are in the infancy of understanding the microbiome’s place in AML pathogenesis and progression. Compared to healthy controls, treatment-naïve patients with AML exhibit reduced diversity in their gut microbiome [11]. One study noticed significantly higher relative abundances of Streptococcus [12]. The analysis of microbiome data in European leukemia populations identified the Rikenellaceae RC9 gut group, Anaerostipes, Slackia, and Lachnospiraceae ND3007 group as risk factors for AML [13]. However, understanding their causal relationship with AML biology requires a detailed multi-omics approach.
The pre-existing state of dysbiosis is markedly exacerbated during treatment (Table 1) [14]. Figure 1 highlights the critical points of microbiome disruption and alteration during the clinical course of AML patients.
In addition to leukemia-specific therapies, broad-spectrum antibiotics and supportive medications (analgesics, antipyretics, diuretics, antiemetics, allopurinol, laxatives, antacids, and antidepressants) used in AML markedly disrupt the microbiota [15]. Microbial shifts persist even after hospital discharge and recovery, resulting in new communities highly dissimilar from the baseline [16]. In turn, the microbiome composition affects the outcomes of the treatment. Higher alpha diversity and enrichment in health-associated taxa belonging to the genera Faecalibacterium, Ruminococcus, Blautia, and Butyricimonas at diagnosis are predictive for improved the post-induction recovery of platelet, lymphocyte, and neutrophil counts [17]. Dysbiosis has proven importance in complications of intensive therapy, including infections [18,19], by creating an environment conducive to pathogen colonization [20]. Conversely, a favorable microbiota composition has been associated with improved outcomes, reduced treatment-related complications, and enhanced responses to cancer therapies, highlighting its potential as a promising avenue for therapeutic intervention [19,21,22].
Based on previous clinical observations from different cancers, we conclude that the drug’s mode of action or side effects are partly attributable to the composition and function of the microbiome [23]. However, challenges remain in the AML setting. A key obstacle is the lack of a detailed mechanistic understanding of how treatments, especially novel therapies, interact with human biology and the microbial community, limiting further exploration in pharmacomicrobiomics. While the real-world data let us look closer at the microbiome dynamics during the treatment of AML, interventional studies remain largely in preclinical stages, leaving many questions about the safety, implications, and therapeutic relevance of microbiome-centered strategies.
This paper reviews the literature to summarize existing findings on the complex crosstalk between the microbiome and AML therapy. First, we present how AML treatments, particularly chemotherapy and antibiotics, influence the microbiota composition. Then, we focus on the role of the microbiome in modulating AML treatment outcomes, considering underlying mechanisms. Further, we look at prebiotics, probiotics, paraprobiotics, and postbiotics as potential therapeutic strategies. Finally, we outline current and future research directions while addressing the limitations of pharmacomicrobiomics in AML treatment.

2. AML Therapy Triggers Dysbiosis

In addition to anticancer therapies, supportive agents—such as antibiotics, analgesics, antiemetics, allopurinol, diuretics, laxatives, proton pump inhibitors, and psychiatric medications—as well as treatment-related gut barrier disruption and immunosuppression, contribute to dysbiosis. Reduced microbial diversity and pathogenic shifts at the phylum, genetic, and metabolic levels characterize this state.

2.1. Antileukemic Treatment

Intensive therapy in acute leukemia is prolonged, highly myelosuppressive, and causes significant mucosal barrier damage. The standard induction treatment for AML involves 3 days of daunorubicin and 7 days of cytarabine (3 + 7 regimen). Newer strategies include CPX-351, a dual-drug liposomal encapsulation of these agents, targeted therapies such as FLT3 inhibitors (midostaurin, quizartinib, and gilteritinib), IDH inhibitors (ivosidenib, olutasidenib, and enasidenib), the Hedgehog pathway inhibitor glasdegib, the BCL-2 inhibitor venetoclax, and the monoclonal antibody gemtuzumab ozogamicin. Consolidation therapy with cytarabine, followed by HSCT in intermediate and adverse-risk patients, constitutes the standard post-remission therapeutic procedure for AML [24]. For patients deemed unfit for intensive therapy, a lower-intensity regimen combining azacitidine and venetoclax remains a viable alternative (Figure 2).

2.1.1. Standard Therapy

The increasing number of studies has documented the negative impact of standard therapy on the microbiota in AML. The post-chemotherapy decline is characterized by an increase in Lactobacillus, a decrease in Blautia, and the dominance of opportunistic pathogens like Staphylococcus, Enterobacter, and Escherichia in the gut. Long-term analyses of oral and stool microbiomes during intensive chemotherapy show significant intra-patient variability in the microbial diversity, which is linked to a higher risk of infection. This variability is associated with predominant pathogenic genera such as Staphylococcus, while more stable states exhibited higher levels of beneficial microbes like Akkermansia, Subdoligranulum, and Pseudobutyrivibrio [19,25]. Microbial disruptions caused by chemotherapy are enduring, with increased levels of Bacteroides and decreased levels of Faecalibacterium and Alistipes lasting up to six months after treatment. The microbiome remains altered from its baseline composition even after antibiotic tapering and post-discharge recovery, failing to return to its original state—suggesting sustained disruption and incomplete restoration [16]. Reduced gut microbiota (GM) diversity persisted despite bone marrow recovery, and subsequent re-induction or salvage therapy further destabilized the ecosystem, leading to compromised colonization resistance, increased susceptibility to Enterococcus overgrowth, and a higher risk of infections [26]. Rattanathammethee et al. studied the microbiome in the long term to identify changes during neutropenic fever (NF). Firmicutes thrived during NF but declined after bone marrow recovery, contrasting with trends in Bacteroidetes and Proteobacteria. Compared to pretreatment, Enterococcus increased during NF, while Escherichia declined. Microbial richness was higher before treatment than during NF and remained at a low level after bone marrow recovery [27]. This study highlights a pathogenic association and suggests the potential predictive value of the GM composition for NF. In contrast to prior findings, Jing Xu et al. reported increased alpha diversity during chemotherapy when comparing the GM and metabolic profiles of AML patients—with and without chemotherapy—to those of healthy controls. The discrepancy with prior findings may be attributed to the absence of antibiotics. The distinct beta diversity profile differentiated three groups. Compared to controls, AML patients displayed an increased Firmicutes-to-Bacteroidetes ratio, with the significant enrichment of Collinsella and Coriobacteriaceae as potential biomarkers. Additionally, the Eubacterium hallii group was notably enriched in AML patients relative to those undergoing chemotherapy [28].
Medications influence not only the composition of the gut microbiota but also its metabolic activity, thereby affecting the host physiology. Hueso et al. linked the 7 + 3 to gut injury and dysbiosis, evidenced by decreased plasma citrulline (a marker of functional enterocyte mass), short-chain fatty acids (SCFAs), and the fecal bacterial load, except for Escherichia coli and Enterococcus spp. These features were associated with concurrent histologic impairment in mice with AML [29]. Pötgens et al. have recently highlighted the links between post-treatment gut microbiome changes and cachectic features in AML patients. Elevated systemic inflammation, muscle mass depletion, anorexia, and weight loss coexisted with the transient impairment of the gut barrier function and persistent reduced diversity. Lactobacillaceae and Campylobacter levels were increased at induction completion, whereas Enterococcus faecium and Staphylococcus levels were at discharge. Metabolomics analyses indicated reductions in urinary hippurate and fecal bacterial amino acid metabolites [30].
Arsenic trioxide, in combination with all-trans retinoic acid, forms the cornerstone of therapy for acute promyelocytic leukemia. After exposure, patients presented reduced diversity and notably decreased Bifidobacterium adolescentis and Lactobacillus mucosae. Additionally, arsenic trioxide prompted the development of resistance genes in Bacteroides fragilis, a prevalent gut bacterium [31].

2.1.2. Novel Therapies

Limited research has considered the microbiome in the context of novel therapies. Recently, researchers have paid close attention to CPX-351, which positively impacts the mucosal barrier, microbiome composition, and immune homeostasis in an animal AML model. The effects were mediated via the activation of the aryl hydrocarbon receptor-IL-22-IL-10 pathway and the production of immunomodulatory metabolites by anaerobes, which improve gut barrier integrity, decrease local inflammation, and regulate the local microbial community [32]. Liu W et al. assessed GM and metabolite profiles of serum and urine in 29 patients with AML before, during, and post-therapy in the following groups: chemotherapy (cytarabine + idarubicin) +/−venetoclax; azacytidine + venetoclax/gilteritinib/afatinib; and venetoclax + gilteritinib. Treatment reduced GM diversity, which did not recover until the next cycle. Enterococcus expansion was associated with a high risk of NF, whereas decreases in Anaerococcus and Dialister and an increase in Enterobacteriaceae predicted CR. The authors did not compare specific therapies [33]. A taxonomic comparative analysis in patients with colorectal, stomach, breast, lung, melanoma, and lymphoid neoplasms and AML did not identify any bacteria that differentiated patients tested before and after immunotherapy or chemotherapy in any of the groups, except for breast cancer. The patients with AML received different treatments (cytarabine + idarubicin, cytarabine + daunorubicin, or cytarabine + gemtuzumab ozogamicin), either with or without autoHSCT [34]. At ASH 2024, Italian researchers presented findings from a prospective analysis of GM and blood and fecal metabolomics in myeloid neoplasms (AML n = 198), at diagnosis and follow-ups. In treated patients, both the microbiome composition and metabolomic profiles shifted significantly, alongside gut mucosal injury from chemotherapy. Compared to the 3 + 7, hypomethylating agents +/−venetoclax better preserved the microbial balance, correlating with a lower risk of infections [35].

2.2. Allo-HSCT and Microbiome

Allo-HSCT includes a conditioning regimen, typically involving chemotherapy and radiotherapy, followed by the infusion of donor hematopoietic cells. This procedure significantly alters the composition and function of the microbiome, influencing clinical outcomes.

2.2.1. Conditioning

The impact on the intestinal microbiome depends on the specific conditioning regimen. Montassier et al. reported a post-conditioning decline in α-diversity, characterized by a significant decrease in Firmicutes and an increase in Escherichia spp., correlated with the conditioning intensity [22,36]. High-intensity myeloablative regimens, based on total body irradiation (TBI) or busulphan, showed the highest depletion of commensal bacteria and expansion of Enterococcus, while cyclophosphamide/fludarabine/TBI200 preserved pre-transplant microbial compositions most effectively [37]. Similarly, the myeloablative group showed the most profound decline in the alpha diversity, species, gene, and metabolic richness following allo-HSCT [38].
Radiation acts as a stressor to the gastrointestinal tract’s microbial ecosystem, potentially leading to mucositis, diarrhea, and fatigue in cancer patients. The systematic review of studies on ionizing radiation in animals revealed an increased abundance of Lactobacillaceae and Staphylococcaceae families, alongside decreased levels of Lachnospiraceae, Ruminococcaceae, and Clostridiaceae [39]. A murine model examining total body irradiation at 0, 4, 8, and 12 Gy revealed dose-dependent changes in intestinal tissues and GM. The abundance of Proteobacteria, Escherichia-Shigella, Eubacterium xylanophilum group, and Lactobacillus murinus correlated with the radiation dose. Dysbiosis persisted through recovery, but may be alleviated by a 14-day administration of probiotics [40]. In mice, radiotherapy and melphalan therapy increased the levels of bacteria with mucin-degrading capabilities, particularly Akkermansia spp., and fever risk, similar to cytotoxic chemotherapy in HSCT neutropenic recipients, even without antibiotics [41].
GM may enhance the radiosensitivity and strengthen anticancer responses; in contrast, broad-spectrum antibiotics reduce radiotherapy efficacy. Proposed mechanisms include direct and SCFA-mediated anti-tumor immunity, as well as the modulation of the hypoxic tumor microenvironment. The GM contributes also to radiation-induced toxicities, including diarrhea, mucositis, and pulmonary complications, highlighting its potential as a therapeutic target [42].
Rashidi et al. compared GM changes between patients with intensively treated acute leukemia and recipients of allo-HSCT. Microbial diversity decreased and Enterococcus increased in both cohorts, while Lactobacillus rose only in the leukemia group [43], emphasizing the need for a personalized approach to address distinct microbiota changes in individual patients.

2.2.2. T-Cell Depletion

Alloreactive T lymphocytes in the graft worsen dysbiosis by damaging intestinal epithelial cells. T-cell depletion can mitigate this effect. The decline in the diversity of the intestinal microbiota during allo-HSCT remains generally consistent across various graft-versus-host disease (GVHD) prophylaxis regimens, including post-cyclophosphamide-based and anti-thymocyte globulin-based approaches [44,45]. However, variability in the transplant indications, comorbidities, antimicrobial exposure, and conditioning intensity hinders the direct comparison of GVHD prophylaxis effects across clinical studies.

2.3. Antibiotics

Due to the high mortality related to infections, an appropriate antibiotic policy is a key element of AML treatment. However, the overuse or misuse of antibiotics leads to intestinal dysbiosis, which is associated with recurrent Clostridioides difficile infections (CDI), systemic infections, and worse clinical outcomes. Table 2 summarizes the clinical studies investigating the effect of antibiotics on the microbiome in AML.
Antibiotic-induced dysbiosis has been linked to treatment outcomes and complications in patients with AML with the strongest body of evidence relating to gastrointestinal GVHD. The use of broad-spectrum antibiotics such as imipenem–cilastatin and piperacillin–tazobactam for NF was associated with increased GVHD-related mortality in clinical trials and murine models [48]. Antibiotic prophylaxis and therapy promoted enterococcal dominance, correlating with a higher risk of gastrointestinal GVHD [47]. In addition to compositional changes, HSCT patients’ microbiomes showed an accumulation of antimicrobial resistance (AMR) genes, closely associated with aGVHD. Interestingly, AMR gene patterns could not be fully explained by antibiotic use alone, suggesting more complex mechanisms of resistance acquisition [53].
Of particular concern is the increased risk of infection associated with antibiotics. Enterococcal domination, exacerbated by metronidazole use, was linked to a higher incidence of vancomycin-resistant Enterococcus bacteremia [46]. Prolonged carbapenem use (>72 h) significantly reduced gut microbial alpha diversity at the time of neutrophil recovery and was associated with a higher risk of infection within the subsequent 90 days [54]. In contrast, fluoroquinolone prophylaxis was independently associated with a 55% reduction in Gram-negative BSIs, likely due to the suppression of intestinal colonization by Gammaproteobacteria [55]. Rifaximin has emerged as a potentially beneficial alternative, associated with lower enterococcal prevalence, a reduced incidence of gastrointestinal GVHD, lower transplant-related mortality, and improved OS, compared to regimens involving ciprofloxacin plus metronidazole, piperacillin–tazobactam, meropenem with vancomycin, ceftazidime, or multiple systemic antibiotics [49].
The association between specific antibiotics and AML treatment outcomes remains difficult to establish due to numerous confounding variables. By inducing dysbiosis, antibiotics may disrupt drug metabolism and impair immune function, potentially compromising the therapeutic efficacy. However, a comprehensive assessment of treatment effects must consider both the indications for and the specific use patterns of antimicrobial agents.
Prophylactic and empirical antibiotic practices for AML vary globally. Fluoroquinolones prophylaxis was recommended in 2005 by the European Conference on Infections in Leukemia (ECIL) for high-risk patients, such as those with AML or undergoing HSCT [56]. However, its common use has increased resistance to Gram-positive, Gram-negative, and CDI [57]. Given the absence of an overall mortality benefit, its clinical value remains increasingly contested [58]. Monotherapy with rifaximina offers a safer alternative to current non-selective prophylaxis in AML, preserving intestinal microbiome diversity even when used alongside broad-spectrum antibiotics [49]. It acts eubiotically by reducing bacterial virulence, inhibiting adherence to epithelial cells, and decreasing mucosal inflammation [59]. Its localized gastrointestinal activity targets aerobic Gram-negative pathogens while sparing the anaerobic microbes, representing over 99% of the gut microbiome. In clinical settings, rifaximin minimizes the disruption of GM, promoting beneficial bacteria like Bifidobacteria and Lactobacilli [59].
The current ECIL-10 recommendations for empirical therapy in AML patients include escalation (anti-pseudomonal cephalosporins, piperacillin–tazobactam, cefoperazone–sulbactam, or piperacillin plus gentamicin) and de-escalation strategies (carbapenem monotherapy, anti-pseudomonal β-lactams with aminoglycosides or quinolones, colistin with β-lactams ± rifampicin, glycopeptides, or newer agents for resistant Gram-positive infections) [60]. According to the increasing incidence of extended-spectrum beta-lactamase-producing infections, carbapenems with broad-spectrum activity against Gram-positive and Gram-negative organisms are commonly selected. However, they also pose a significant danger by reducing beneficial gut commensals, especially those that uphold the mucus layer, and contribute to the rise of antibiotic resistance [61].
Enterococcus species are common commensals of the gastrointestinal tract. Due to their intrinsic resistance to multiple antimicrobial classes, including cephalosporins, lincosamides, fluoroquinolones, aminoglycosides, clindamycin, and trimethoprim/sulfamethoxazole, Enterococcus has become the most common dominating genus in AML [46,62]. Exposure to cephalosporins, fluoroquinolones, and vancomycin is a risk factor for colonization with vancomycin-resistant Enterococcus [63,64]. Patients with Enterococcus domination tended toward E. faecalis and E. faecium bacteremia, gut GVHD, reduced OS, and increased treatment-related and relapse-related mortality [62]. Alongside fluoroquinolone-resistant Enterobacterales, extended-spectrum beta-lactamase producers, and multidrug-resistant Pseudomonas aeruginosa, these pathogens now contribute to higher mortality rates in AML patients.
Clinician adherence to recommendations against routine bacterial prophylaxis and early de-escalation remains suboptimal. Improving antibiotic regimens to minimize harm to the GM while maintaining the therapeutic efficacy presents a challenge for future research endeavors.

3. Impact of the Microbiome on the Anticancer Treatment in AML

Interindividual variability in the drug response and toxicity poses a significant challenge in cancer therapy. To address this, pharmacomicrobiomics, which explores the relationship between variations in the human microbiome and pharmacological responses, has rapidly developed in recent years, revealing numerous underlying mechanisms. The microbiome directly influences a drug’s availability and activity and the sensitivity of the host to related toxic effects [65]. Microbiome-derived metabolism refers to the biochemical transformation of xenobiotics by microbiome-produced enzymes: the activation of a prodrug or conversion to inactive or toxic metabolites. Additionally, the GM may influence drug metabolism by competing with drug molecules for metabolizing enzymes, altering the levels of the host’s drug-metabolizing enzymes in the liver and intestine, or producing enzyme-inducing metabolites derived from the diet [66]. Indirectly, the microbiome improves therapeutic outcomes by modulating the host immune system, as observed in enhanced antigen presentation and better T cell function during a PD-1/PD-L 1 blockade in immunotherapy. Responders showed higher gut microbial diversity and increased levels of Clostridiales, Ruminococcaceae, and Faecalibacterium [67]. Understanding various mechanisms of action in pharmacomicrobiomics has led to the idea of targeting the microbiome to predict responses, boost efficacy, and reduce side effects of cancer treatments.
Microbiota chemically alter the metabolism of several drugs. In an analysis by Zimmermann et al. testing the metabolic capacity of 76 gut microbial strains, 176 of 271 orally administered drugs (66%) were metabolized by at least one bacterial strain [68]. Some of these drugs are used in hematology. Variations in the microbiome phenotype influence the efficacy and toxicity profiles of drugs such as cyclophosphamide, methotrexate, platinum compounds, cisplatin, doxorubicin, cladribine, rituximab, and anti-CD19 CAR T cell therapy [21,69,70,71,72,73,74]. This is also supported by the reduced anticancer immune response related to dexamethasone and corticosteroid metabolism, which results from the adverse effects of antibiotics on the microbiome [68]. In vivo data show that E. coli can inhibit chemotherapeutics like cladribine, vidarabine, doxorubicin, idarubicin, daunorubicin, etoposide phosphate, and mitoxantrone, while enhancing the effects of drugs such as mercaptopurine, fludarabine phosphate, and 5- 5-fluorocytosine [72]. Bacteria can also influence cancer cell characteristics and modify chemotherapy sensitivity through toxin production. In AML patients, Staphylococcus aureus-derived enterotoxins A and B increase AML cell proliferation, migration, invasion, and resistance to cytarabine. By dysregulating immune-related genes, these toxins may help AML cells evade hostile environments, potentially via the endoplasmic reticulum stress signaling pathway [75]. Only a few clinical trials have focused on the microbiome’s impact on the drug response in AML patients (NCT05596968, NCT04214249).

4. Impacts of the Microbiome on Treatment Complications in AML

The findings from clinical studies in acute leukemia point to a role of the microbiota in shaping chemotherapy complications such as organ toxicity, neutropenia, infections, GVHD, and gastrointestinal dysfunction. Emerging evidence identifies alpha diversity and GM variability as potential predictors of infection risk during and after chemotherapy [19,25,54]. Pre-treatment Proteobacteria abundance correlates with NF [76] and low diversity with a high Enterococcus spp. level relates to increased systemic inflammation and intestinal epithelial integrity injury [77]. The intricate relationship between a pre-existing microbiome composition and treatment-related complications influences the overall outcomes after allo-HSCT in leukemia. For instance, increased bacterial diversity [22] and a higher relative abundance of Blautia were correlated with reduced mortality [78], whereas lower gut microbial diversity was associated with decreased OS, a higher risk of treatment-related mortality, and an increased risk of GVHD-related mortality [79]. Besides lower microbial diversity, a compositional shift characterized by a decline in beneficial bacteria (A. muciniphila, Blautia, and anti-inflammatory Clostridia [80]), as well as an increase in Enterococcus [81], was predictive of aGVHD in another study. Also, microbiota-derived metabolites, including SCFAs, tryptophan and its derivatives, choline metabolites, tyrosine, and bile acids, affected the severity and prognosis of aGVHD [82]. Finally, a higher abundance of Eubacterium limosum was linked to a reduced risk of relapse and disease progression in patients undergoing allo-HSCT [83].
Figure 3 provides a comprehensive summary of the relationships between microbiome changes, AML treatment, and clinical outcomes, highlighting potential underlying mechanisms.

5. The Therapeutic Potential of the Microbiome

Based on evidence outlining the role of the microbiota in mediating the effects of drugs [84,85,86], researchers have intensified efforts to develop strategies for manipulating the microbiome for preventive and therapeutic purposes. However, in high-risk patients, such as those with leukemia, numerous practical and ethical challenges have hindered clinical implementation.

5.1. Antibiotic Stewardship

Antimicrobial stewardship—coordinated strategies to optimize antimicrobial use, improve outcomes, reduce resistance, and limit the spread of MDRO—is essential to the comprehensive care of leukemia patients. This section discusses emerging strategies that aim to balance effective infection control with microbiome preservation, ultimately enhancing treatment outcomes.
Broad-spectrum antibiotics (vancomycin, imipenem, colistin, ampicillin, beta-lactams, quinolones, sulfonamides, streptomycin, and neomycin) have been linked to adverse effects on cancer therapy, possibly through microbiome-mediated mechanisms. Therefore, using narrow-spectrum antibiotics to selectively target harmful bacterial species, known as “targeted microbiome-sparing antibiotics,” could play an adjunctive role in modulating the therapy response.
A lack of mortality benefit and rising resistance with fluoroquinolone prophylaxis support revising current strategies, with rifaximin emerging as a promising alternative [87,88,89].
Empirical antibiotic strategies are often tailored to general institutional antimicrobial sensitivity patterns while overlooking adjustments based on an individual patient’s microbiological colonization profile. Characterizing microbiome structures upfront can help guide personalized prophylaxis and treatment.
Following current guidelines, the early de-escalation and discontinuation of antibiotics for NF are effective without worsening outcomes in hematology wards [90]. Limitations in carbapenem use resulted in decreased vancomycin-resistant E. faecium colonization, BSI, hospitalization duration, and costs in the patients with hematological malignancies unit [91]. A study in AML and MDS patients undergoing induction chemotherapy confirmed the safety of the withdrawal of empirical antibiotics after 72 h in hemodynamically stable patients [92].
Further research is needed to evaluate the effectiveness of new antibiotic compounds and microbiome-preserving strategies, such as beta-lactamase enzymes that act locally within the gastrointestinal tract, and non-specific adsorbents to sequester the antibiotics within the colon [93,94].

5.2. Diet

Diet is one of the most significant factors influencing the human microbiome. Microbiome-mediated mechanisms partially explain the dietary impact on the drug response and side effects, as seen in immune [95] and radiation-induced anti-tumor responses [96] and chemotherapy-induced mucositis [97,98]. Targeted dietary interventions may return dysbiosis to a healthy state or tend to change in favor of the oncologic patient.
In recent years, various diets, including fasting or fasting-mimicking diets, ketogenic diets, and fiber-rich Mediterranean diets, have been studied to enhance the effectiveness of cancer treatments while reducing their toxicity. Research has shown that fasting interventions are linked to increased levels of beneficial gut bacteria such as Faecalibacterium, Roseburia, Butyricoccus, and Coprococcus. These bacteria are significant producers of SCFAs. F. prausnitzii has been shown to improve chemotherapy efficacy in preclinical studies and clinical trials involving cancer patients (melanoma [67], thyroid [99], and colorectal [100,101]). Akkermansia, Roseburia, and Ruminococcaceae were all elevated in patients on a ketogenic diet [102]. Preliminary findings from a study investigating the administration of a high-fat ketogenic diet before and during induction chemotherapy in patients with AML demonstrated favorable tolerability and an increase in DNA damage within leukemic blasts. Additionally, the diet exhibited protective effects against senescence in healthy lymphocytes. However, the study did not evaluate its impact in the microbiome context [103]. A fiber-rich diet improved the efficacy of immunotherapy [104] and reduced the toxicity of 5-fluorouracil treatment [98] by prompting the gastrointestinal microbiome to produce SCFAs. In a mouse model, the Mediterranean diet, compared to the Western diet, was associated with greater microbiome diversity and higher abundances of Clostridium, Lactobacillus, Oscillospira, and Faecalibacterium, while showing lower levels of Coprococcus and Ruminococcus [105]. These results were consistent with human studies that demonstrated an increased abundance of Lachnoclostridium, Enterorhabdus, and Parabacteroides, along with enhanced SCFA production [106]. A study involving 41 pediatric HSCT patients found that a higher pre-treatment intake of soluble fiber, iron, breast milk, bazlama, yogurt, onion, parsley, and bulgur was associated with earlier neutrophil engraftment, a lower incidence of NF episodes, and shorter durations of total parenteral nutrition [107]. Despite promising findings, adhering to dietary interventions can prove challenging for malnourished AML patients undergoing aggressive treatments. Therefore, maintaining a balanced diet with an adequate nutrient intake is essential for recovery and for alleviating treatment-related complications. The low-bacterial diet, designed to minimize infection risk, has gained popularity, yet it has faced criticism. A review of 12 studies on low-bacterial diets found no significant benefits in reducing infection or mortality rates. Additionally, this diet often resulted in a diminished quality of life due to less appealing food options and limited variety, which negatively affected the overall nutritional status [108]. This diet may restrict key dietary components, particularly fiber, leading to dysbiosis and decreased SCFA production. Given the significant role of nutrition in AML patients, the current U.S.A Food and Drug Administration (FDA) guidelines on nutrition highlight the importance of maintaining food safety through proper handling rather than dietary eliminations [109]. More specific recommendations that incorporate microbiome-related changes and their impact on treatment on AML warrant further research.

5.3. Microbiota-Targeted Nutritional Approaches

Microbiota-modulating dietary modalities, including prebiotics, probiotics, paraprobiotics, and probiotics, have achieved favorable results in non-hematological patients (lung, genitourinary, colorectal, renal, prostate, melanoma, solid, gastrointestinal, pancreatic, hepatocellular, head and neck, and breast cancers [110]). In patients with AML, the effects and safety of these interventions are complicated by profound immunological disturbances, limiting research primarily to in vitro and animal studies, with only a few involving humans.

5.3.1. Probiotics and Prebiotics

Certain probiotic species may promote apoptosis in myeloid leukemia cells. Lactobacillus reuteri suppresses NF-κB-dependent proliferation, reduces survival signaling, and enhances MAPK pathway activity, thereby promoting cell death [111]. L. casei rhamnosus killed the cells of the human monocytic leukemia line, THP-1 [112]. In vitro, Lactobacillus kefiri P-IF-based kefir induced apoptosis in multidrug-resistant human myeloid leukemia cells [113]. Curcumin enhanced sensitivity to cytarabine and influenced the intestinal microbiota without directly affecting the AML cell lines in a mice model. A metagenomic analysis indicates an increase in beneficial bacteria, such as Lactobacillus acidophilus, Bifidobacterium bifidum, and Lactobacillus reuteri, alongside a decrease in pathogenic species like E. coli, A. muciniphila, and Bacteroides fragilis. Additional mechanisms may include the enhancement of the intestinal integrity and a reduction in bacterial translocation into the bloodstream [114]. Combining both probiotic and prebiotic properties, kimchi inhibited HL-60 (human acute promyelocytic leukemia) cell proliferation by inducing apoptosis and disrupting the mitochondrial membrane potential, suggesting anticancer potential [115]. Concerns about systemic infections, pathogenicity, excessive immune stimulation, toxicity, metabolic activity, and horizontal gene transfer continue to restrict the medical use of probiotic products in vulnerable patients. For instance, a study found that oral treatment with Lactobacillus species in individuals with compromised immune systems led to bacteremia [116].

5.3.2. Paraprobiotics

Researchers are exploring paraprobiotics—inactivated bacterial cells or extracts—as safer alternatives to live probiotics, with some already available commercially. Their various benefits—including the absence of risk for bacterial translocation, prevention of transferring antibiotic resistance genes, and the convenience of standardization, production, transportation, and storage—render them a viable alternative for patients with weakened immune systems [117].

5.3.3. Postbiotics

Postbiotics cover a group of different chemical compounds, including vitamins, organic acids, SCFAs, and amino acids. Their anticancer effects include selective cytotoxicity against tumor cells, antiproliferative action, apoptosis induction, antioxidant activity, gut microenvironment modulation, inflammation reduction, intestinal barrier protection, and immune response regulation [118]. SCFAs play a critical role in numerous biological processes, with acetic acid, butyric acid, and propionic acid constituting the three primary types. Among these, butyrate functions as a histone deacetylase inhibitor, effectively normalizing epigenetic imbalances. Furthermore, butyrate has been shown to modulate genes associated with apoptosis and the cell cycle, impede the proliferation of tumor cells, and counteract resistance linked to anti-apoptotic regulators, such as MCL-1 [119], a known mechanism of resistance to venetoclax [120]. The anti-cancer potential of sodium butyrate has driven trials on its efficacy in AML treatment. Pulliam et al. described the apoptotic effect of butyrate in human acute leukemia cells by the activation of caspase-3, reduction in cell viability, and lowering the concentration of the chemokines CCL2 and CCL5 [121]. Its implementation enhanced the efficacy of venetoclax [122]. Administering sodium butyrate with TRAIL enhanced the killing effect on t (8;21) AML cells [123]. Clinical trials are needed to explore the efficiency of this approach in AML patients.

5.3.4. Modulating Chemotherapy Toxicity with Pro-, Pre-, Para-, and Postbiotics

With limited options for managing chemotherapy toxicity, microbiome modulation offers a promising therapeutic strategy. In children with acute leukemia, daily supplementation with probiotic Lactobacillus rhamnosus reduced abdominal distension, intestinal constipation, and nausea during chemotherapy [124]. The Bifidobacterium breve strain Yakult diminished the incidence of fever in pediatric leukemic patients [125]. Enteral supplementation enriched with glutamine, fiber, and oligosaccharides resulted in fewer days of diarrhea and mucositis of grade 3–4 in HSCT patients [126]. The administration of resistant starch and the commercially available prebiotic mixture GFO to allo-HSCT recipients, starting from pretransplant conditioning through day 28 post-transplant, effectively mitigated mucosal injury and reduced the incidence of diarrhea. This intervention positively impacted gut microbiome diversity, increased the population of butyrate-producing bacteria, and elevated fecal butyrate concentrations after the transplant [127]. In a Phase II study involving AML patients undergoing high-dose chemotherapy and radiation followed by HSCT, the use of Lactobacillus brevis CD2 for prophylaxis was both safe and effective. Of the 31 patients enrolled, only six developed severe oral mucositis [128]. In a recent retrospective analysis, viable Bifidobacterium tablets significantly reduced the incidence and duration of grades 1–2 oral mucositis during the transplant process without impacting HSCT outcomes [129]. In contrast, the first randomized trial of HSCT patients supplemented with Lactobacillus rhamnosus GG found no significant changes in the GM or GVHD incidence [130].
Though currently classified as health products, microbiome-based interventions may soon achieve a drug status, with defined indications, dosages, and safety profiles as research advances.

5.4. The Role of the Microbiota in GVHD: From Mechanisms to Therapies

GVHD remains a major cause of morbidity and mortality following HSCT. Its pathogenesis primarily involves the presentation of host alloantigens to naïve donor T cells, which then mount an immune response characterized by progressive tissue injury. The gastrointestinal tract plays a central role as an early site of alloreactivity. Various antigen-presenting cells—such as dendritic cells, mesenchymal stromal cells, and intestinal epithelial cells—actively participate in initiating and sustaining pathogenic T cell activation and differentiation [131]. However, many of these factors may modulate GVHD through microbiome-related mechanisms, particularly within the gastrointestinal tract, the most densely populated and diverse microbial ecosystem in the human body. The dynamic interplay between treatment regimens, gut integrity, and microbiota composition forms a tightly interconnected triad that critically influences immune homeostasis. The disruption of the intestinal epithelial barrier—often resulting from chemotherapy, radiotherapy, or antibiotic exposure—facilitates the translocation of pathogen-associated molecular patterns from the microbiota, as well as damage-associated molecular patterns from injured host tissues. This influx of microbial and host-derived signals amplifies local inflammation, creating a pro-inflammatory milieu that promotes the onset and progression of GVHD [132].
Beyond epithelial barrier disruption, dysbiosis itself is a significant driver of GVHD. Both preclinical and clinical studies have proposed multiple microbiome-related mechanisms underlying the microbiome T (8;21) AML cells GVHD association: (1) the modulation of MHC class II expression [133], (2) immunological tolerance control [134], (3) immunoregulation (production of IL-17 and IL-22 by Th17 cells in the lamina propria; Th17 and Treg cells balance) [135,136], (4) the activation of Toll-like receptors [137], (5) the regulation of cytokine profiles (activation of inflammasome, production of IL-18, Th1/Th2 balance) [138], and (6) microbial metabolites—mediated mechanisms (SCFAs modulate immune responses by inhibiting NF-κB signaling, enhancing IL-10 expression, and promoting regulatory T cell differentiation) [139,140,141].
Numerous preclinical studies and clinical investigations have explored the relationship between the microbiota composition and the risk and severity of GVHD. Systematic reviews and meta-analyses in both pediatric [142] and adult populations [143] have identified certain characteristic microbial signatures associated with GVHD-related dysbiosis. The most consistent findings include reduced microbial diversity and an increased dominance of genera such as Lactobacillales, Staphylococcaceae, Enterobacteriales, and Enterococcus. These changes often coincide with a depletion of bacterial taxa linked to butyrate synthesis, notably Clostridia, Lachnospiraceae, Blautia, Bacteroides, and Akkermansia muciniphila.
Although pinpointing a single causative microorganism remains challenging, some studies have attempted to define stereotyped pathogenic microbial consortia. Nevertheless, these microbial patterns have not yet been consistently replicated across independent research groups, highlighting the complexity and heterogeneity of GVHD-related dysbiosis.
Growing scientific and clinical evidence supports microbiota-targeted strategies to mitigate GVHD. Therapeutic approaches focus on microbial metabolites such as SCFAs and 3-indoxyl sulfate [144], antibiotic use optimization (e.g., narrow-spectrum agents, a reduced duration, and novel β-lactams), and microbiome-supportive diet interventions including prebiotics, probiotics, and symbiotics [126,145]. Fecal microbiota transplantation (FMT) has a growing interest among hematologists as a promising therapeutic option for GVHD; however, further research is needed to optimize its application and confirm its safety and efficacy in immunocompromised patients [146].

5.5. FMT as Full Microbiome Restoration: A Targeted Strategy Against Clostridioides Difficile Infection

CDI poses a significant clinical challenge in patients with hematologic malignancies. Among patients with AML, reported CDI incidences range from 4.8% to 9%, while in recipients of allo-HSCT, the rate rises markedly to 14–30.4%. In conjunction with the underlying disease burden, CDI exacerbates morbidity and contributes to treatment-related mortality, which may approach 20% in this population [147].
Microbiome and CDI are closely related. Balanced GM protect against colonization with pathogens by competing for nutrients, providing inhibitory substances, and regulating the host’s immune response. An imbalance between beneficial and harmful bacteria promotes the transformation of C. difficile spores into vegetative forms. First of them are characteristic for a resistance to antibiotics and transmission, whereas vegetative cells produce toxins (toxin A, B, and binary toxin), causing disease.
While numerous studies have addressed this topic broadly, data specifically focused on patients with AML remain limited. In this population, several factors—including the use of proton pump inhibitors, immunosuppressive agents, impaired mucosal immunity, and the disruption of the intestinal epithelial barrier—may significantly increase CDI susceptibility. However, the most critical contributor remains the widespread use of broad-spectrum antibiotics, which profoundly disturb the gut microbial diversity, density, and metabolic function, thereby promoting C. difficile colonization and overgrowth. While nearly all antibiotics, including those used to treat CDI (e.g., metronidazole and vancomycin), can elevate the infection risk, cephalosporins, clindamycin, ampicillin, amoxicillin, penicillin, and fluoroquinolones are associated with the most substantial alterations in the GM [148]. Newly established ecosystem in the gut creates an environment conducive to C. difficile colonization and proliferation [149]. Conversely, recurrent C. difficile infections further exacerbate gut dysbiosis, perpetuating a harmful cycle [150]. FMT aims to restore the balance of the GM, which may facilitate the clearance of multidrug-resistant organisms, including extended-spectrum beta-lactamase-producing Escherichia coli, vancomycin-resistant Enterococcus, and carbapenem-resistant Enterobacteriaceae [151]. Currently, recurrent C. difficile infection remains the only formally approved indication for FMT in clinical guidelines for both adults and children. Nevertheless, guidelines for managing C. difficile infection in hematology populations advise against FMT, based on the absence of randomized controlled trials in immunocompromised patients and challenges related to administration [152]. Despite these concerns, emerging clinical evidence supports the safety and feasibility of FMT in selected cases, underscoring the need for broader validation in future trials [153,154].
Completed and ongoing clinical trials on the microbiome in AML, including microbiome-targeted interventions, are listed in Table 3.

6. Challenges, Research Directions, and Clinical Implications

As a result of the progress made over recent years, microbiome-focused approaches have become a game-changer for medicine. Following the leading trends in cancer therapy, integrating the microbiome in multimodal management is inevitable. Despite an improvement in the treatment outcomes, AML remains a disease with the highest mortality rates, and further advancements appear to have plateaued with the current available methods. In line with molecularly targeted therapies, individual microbiome-tailored interventions may support the transition toward personalized medicine by both optimizing the impact of conventional therapies and mitigating off-target effects. Building on both foundational knowledge and current findings, we are the first to comprehensively position the microbiome within the therapeutic landscape of AML from multiple perspectives. Due to the relatively limited and inconsistent data, a comprehensive summary is crucial for guiding the direction of future research.
Collectively, evidence from both basic science and clinical studies demonstrates a bidirectional relationship between cancer treatment and patients’ microbial profiles. Despite differences in methodologies, treatment protocols, and clinical factors that can bias the analysis, some findings remain consistent. These typically indicate a reduction in commensal flora, the dominance of a single species, and decreased intestinal microbiota diversity after treatment. The high complexity of the microbiome’s compositional and functional structure in leukemic patients prevents the unequivocal identification of a beneficial profile, but SCFA producers are generally linked to positive results. In contrast, the expansion of Enterococcus is commonly associated with bloodstream infections, GVHD, and mortality. Considering the progress in understanding complex microbiome–host interactions, we recognize that we are currently witnessing only the beginning of a boom fueled by breakthroughs in cancer treatment. The number of uncertainties highlights the growing need for extensive and long-term research to draw reliable conclusions. In this section, we explore the primary gaps and unknown frontiers of microbiome research (Figure 4).
Microbiome-focused studies face several methodological limitations. Most have evaluated participants at various stages of treatment (before, during, after induction, after HSCT, etc.) without clearly distinguishing between different regimens or drugs. Studies often fail to account for confounding factors such as diet and antibiotics. Since animal models only partially reflect the human microbiome ecosystem, clinical trials are essential to establish the true link between the microbiome and AML therapies and to identify microbiome-specific targets for treatment.
Currently, the role of microorganisms in cancer therapy goes far beyond microbiome issues. While the synergistic effect of combining traditional therapies with tumor-targeting or immunotherapy has significantly improved outcomes in oncology, challenges such as limited clinical response rates, resistance, and off-target effects of drugs continue to hinder the optimal clinical efficiency. Thanks to their unique properties, bacteria play an active role in modern strategies to overcome these drawbacks [154]. The therapeutic potential of microorganisms includes the following: (1) immunotherapeutic applications utilizing live, attenuated, or genetically modified bacteria, either alone or with conventional treatments; (2) the targeted delivery of anticancer agents; (3) the bacterial expression of tumor-specific antigens; (4) the delivery or expression of suppressor genes, anti-angiogenic genes, and suicide genes; (5) RNA interference mechanisms; and (6) the activation of pro-drugs through bacterial cleavage [155]. Both preclinical and clinical trials with novel bacterial therapeutics primarily focus on solid tumors (bladder, prostate, melanoma, lung, gastric, colon, renal, breast, and cervical cancers) with an achievable tumor microenvironment [156]. Microbial therapy for hematologic neoplasms is currently in its early stages. The difficulty in implementing bacterial-based therapies for hematopoietic neoplasms stems from the distribution of cancer cells in the bone marrow and blood circulation, making systemic chemotherapy the most relevant option. Among the few therapies of proven significance, L-asparaginase, an enzyme produced by bacteria such as Escherichia coli, Bacillus subtilis, Streptomyces, or Erwinia species, demonstrates efficacy in treating acute lymphoblastic leukemia and lymphosarcoma [157]. Meirong Li et al. reported that an attenuated Salmonella typhimurium strain, VNP20009, can induce apoptosis in leukemia cells both in vivo and in vitro, inhibit the proliferation of MLL-AF9-induced AML cells, and prolong the survival of AML-carrying mice [158]. Given the complex immune mechanisms involved in leukemia, advancing bacteria-based immunotherapy remains a promising research area. The leukemia-permissive microenvironment of the bone marrow, where pathological leukemic stem cells reprogram mesenchymal stem cells to support leukemia progression, chemoresistance, and relapse, presents a potential therapeutic target in AML.
Not only bacteria, but also other microbes such as archaea, fungi, and viruses inhabit the human body. While many efforts have been made to study bacteria, the issue of non-bacterial microbes in health and disease is still unknown, representing the ‘dark matter’ of the microbial ecosystem. Currently, the role of these non-bacterial microbes in cancer remains elusive, not to mention depicting their role in therapy. To fully understand the host–microbiome complexity, future research should explore the entire microbiome as an integrated ecosystem, focusing on both community structure and functional traits, and emphasizing interspecies interactions. Finally, comprehensive sampling and sequencing, both spatially and temporally, will complete the changing picture of the microbial ecosystem throughout treatment in AML patients.
In addition to the best-studied intestinal toxicity, the human microbiota may be associated with other chemotherapy-induced side effects. The study by Cuozzo et al. demonstrated the effectiveness of the probiotic formulation, SLAB51, in preventing paclitaxel-induced neuropathy by enhancing the expression of opioid and cannabinoid receptors in the spinal cord, reducing nerve fiber damage in the paws and regulating the concentration of proinflammatory cytokines in the serum [159]. A randomized trial involving 159 breast cancer patients yielded compelling evidence that probiotic supplementation reduced the incidence of chemotherapy-related cognitive impairment and enhanced cognitive function, potentially through the modulation of plasma metabolites [160]. Finally, animal model studies have linked the gut microbiome with therapy-induced inflammatory and neuropathic pain [161]. Very little is known about the efficiency of microbiome-mediated intervention in managing complications in AML patients. The horrendous gap in knowledge refers to some specific drug-related side effects, such as well-established anthracycline-induced cardiotoxicity, novel issues of cardiovascular adverse effects associated with midostaurin, and gemtuzumab ozogamicin-related hepatotoxicity. Further research is essential to meet these unmet needs.
The impact of the polymorphism within genes in the microbiome-mediated drug response remains largely unexplored. The manually curated Pharmacogenomics Knowledgebase (PharmGKB) identifies key human genes involved in the individualized drug response; however, it does not incorporate microbial influences on drugs [162]. There are few studies revealing interactions of drugs with known pharmacogenetics with the human gut microbiome [163]. Developing models that capture the variation in the human microbiome can provide valuable insights into xenobiotic metabolism, particularly in the context of genetic variations and metabolic interactions between the host and associated microbes. This strategy may help predict individual drug responses and guide the design of effective, low-risk personalized treatments. The study of molecular pathological epidemiology (MPE) combines the disciplines of biology to provide research frameworks that link genetic and environmental factors, including pharmacological factors, to pathologic processes. The integration of microbiology into the MPE model (microbiology-MPE) can enhance our understanding of the complex interplay between the genetic background, environment, tumor cells, immune cells, drug response, and the microbiome in AML [164].
Recent advancements in high-throughput sequencing technologies, along with innovative statistical and computational methods for multi-omics data analysis, have enabled researchers to predict drug responses. However, the analysis of the omics data for effective treatment personalization in AML remains challenging due to the biological variability of the disease and data complexity. To address these challenges, researchers from the Karolinska Institute proposed MegaFun, a computational method designed to quantify the functional aspects of the microbiome using metagenomics data, to obtain information for prognostic models. The results of this study are still pending. The application of AI in microbiome data analysis is promising and will evolve into personalization in AML management shortly [165].
Besides the beneficial influence on the efficiency of drugs in cancer, some bacteria could promote chemotherapy resistance. Targeting specific bacterial species with bacteriophages offers a novel strategy to overcome chemoresistance. Achieving this goal requires identifying host strains with therapeutic properties to optimize phage combinations for selective pathobiont elimination.
Most prior studies have focused on GM. However, intratumor bacterial signatures may also play a role in regulating cancer development and progression, as documented in the tumor microbiome of pancreatic, lung, and breast cancers [166]. Fu et al. linked tumor-resident microbiota with cancer metastasis, suggesting that circulating tumor cells carrying intratumor bacteria may influence cancer cell survival [167]. In myeloid malignancies, the bone marrow and blood microbiome may play a key role. Advances in molecular technologies have introduced the concept of a blood microbiome, assessed in both healthy individuals and cancer patients. While a core healthy blood microbiome has not been established, dysbiosis appears relevant across various conditions [168,169]. The deep sequencing of blood and bone marrow from 1870 patients (including 612 with AML) revealed distinct dysbiosis patterns across four disease subtypes, with microbial profiles linked to gene mutations and myeloblast percentages. AML patients showed a higher bacterial burden, but lower diversity, dominated by Proteobacteria. Epstein–Barr virus presence correlated with a poorer prognosis in low-risk MDS. Blood and bone marrow samples showed comparable microbial profiles [170]. The blood microbiome exhibits potential for applications in risk stratification, diagnosis, monitoring, and drug development. Nonetheless, the investigation of the bone marrow and peripheral blood microbiome in the context of AML remains largely unexplored for these diagnostic and therapeutic objectives. Significant challenges include contamination in low-biomass samples and uncertainty regarding the viability of microbes identified through NGS. Predominantly, the circulating microbes are commensals and the disruption of physiological barriers in AML may facilitate their transient entry into the bloodstream. Typically, these microbes are cleared rapidly, lacking sustained colonization or functional significance [171]. Further investigation is necessary to distinguish between viable microbes and residual, inactive microbial DNA.
Currently, microbiome-related dietary modalities are classified as health products rather than pharmaceuticals. However, with ongoing technological and scientific advancements, these interventions may achieve the status of drugs with precise indications, dosages, and adverse effect profiles. Investigating their pharmacodynamics and pharmacokinetics within the context of the variable interindividual microbiome signature will present a considerable challenge for pharmacology with potential applications in hematology.
Based on previous evidence, it appears that shortly, microbiome management will become a core element of supportive care for cancer patients, similar to antiemetic, anticoagulant, and psychological support treatments. However, researchers still need to develop safe and effective strategies. Manipulating the composition of the microbial consortium for therapeutic purposes is one of the most explored topics within the field of pharmacomicrobiomics. In the case of AML, we are only beginning to understand the complex interactions between the microbiome and treatment, especially concerning new therapies.

7. Conclusions

Building on both foundational knowledge and current findings, we are the first to comprehensively position the microbiome within the therapeutic landscape of AML from multiple perspectives. The host microbiome and AML therapy are inter-related and affect each other through various mechanisms, including drug metabolism, immunomodulation, direct anticancer effects, and changes in microbial composition. Patients with AML constitute a distinct population where numerous additional factors significantly complicate these relationships. Common side effects, resistance to treatments, and the volatile bioavailability of drugs limit the efficacy of the standard therapy, a critical element for patient outcomes. The human microbiome plays a crucial role in orchestrating all these effects. A mechanistic and functional understanding of the relationships between the microbiome and therapy in AML patients is a clue to optimizing treatment and improving outcomes. It requires complementing microbiota characterization with a detailed analysis of the resistome, metagenome, and metabolome using novel techniques. While targeting human microbes offers promising anti-cancer potential, safety concerns remain a key limitation. The dynamic and multifaceted field of pharmacomicrobiomics represents a significant advancement toward personalized medicine; however, in AML, it remains in the early stages of development, highlighting the urgent need for ongoing progress.

Author Contributions

Conceptualization, A.N.; writing—original draft preparation, A.N.; writing—review and editing, H.T., E.S. and L.G.; visualization, A.N. and A.K.; supervision, H.T., E.S. and L.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
aGVHDacute graft-versus-host disease
alloHSCTallogeneic hematopoietic stem cell transplantation
AMLacute myeloid leukemia
AMRantimicrobial resistance
ASHThe American Society of Hematology
BCL2B-cell lymphoma 2
BSIsbloodstream infections
CAR Tchimeric antigen receptor T-cell
CCL2C-C motif chemokine ligand 2
CCL5C-C motif chemokine ligand 5
CDIClostridioides difficile infection
CRcomplete remission
ECILEuropean Conference on Infections in Leukemia
FDAFood and Drug Administration
FLT3fms-related tyrosine kinase 3
FMTfecal microbiota transplantation
GM gut microbiota
GVHDgraft-versus-host disease
HSCThematopoietic stem cell transplantation
IDHisocitrate dehydrogenase-1
IL-10interleukin–10
IL-22interleukin–22
ISindoxyl sulfate
MAPKmitogen-activated protein kinase
MCL-1myeloid cell leukemia 1
n/anot applicable
NFneutropenic fever
OSoverall survival
PD-1/PD-Lprogrammed death 1/programmed death ligand 1
SCFAsshort-chain fatty acids
TBItotal body irradiation
TNFtumor necrosis factor
TRAILtumor necrosis factor-related apoptosis-inducing ligand
WGSwhole-genome shotgun

References

  1. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in Health and Diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  2. Manzo, V.E.; Bhatt, A.S. The Human Microbiome in Hematopoiesis and Hematologic Disorders. Blood 2015, 126, 311–318. [Google Scholar] [CrossRef] [PubMed]
  3. Marjanovic, N.D.; Hofree, M.; Chan, J.E.; Canner, D.; Wu, K.; Trakala, M.; Hartmann, G.G.; Smith, O.C.; Kim, J.Y.; Evans, K.V.; et al. Emergence of a High-Plasticity Cell State during Lung Cancer Evolution. Cancer Cell 2020, 38, 229–246.e13. [Google Scholar] [CrossRef]
  4. Pleguezuelos-Manzano, C.; Puschhof, J.; Rosendahl Huber, A.; van Hoeck, A.; Wood, H.M.; Nomburg, J.; Gurjao, C.; Manders, F.; Dalmasso, G.; Stege, P.B.; et al. Mutational Signature in Colorectal Cancer Caused by Genotoxic Pks+ E. coli. Nature 2020, 580, 269–273. [Google Scholar] [CrossRef]
  5. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef] [PubMed]
  6. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  7. Panebianco, C.; Andriulli, A.; Pazienza, V. Pharmacomicrobiomics: Exploiting the Drug-Microbiota Interactions in Anticancer Therapies. Microbiome 2018, 6, 92. [Google Scholar] [CrossRef]
  8. Torres-Carrillo, N.; Martínez-López, E.; Torres-Carrillo, N.M.; López-Quintero, A.; Moreno-Ortiz, J.M.; González-Mercado, A.; Gutiérrez-Hurtado, I.A. Pharmacomicrobiomics and Drug–Infection Interactions: The Impact of Commensal, Symbiotic and Pathogenic Microorganisms on a Host Response to Drug Therapy. Int. J. Mol. Sci. 2023, 24, 17100. [Google Scholar] [CrossRef]
  9. Acute Myeloid Leukemia—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/amyl.html (accessed on 20 October 2024).
  10. Cai, S.F.; Levine, R.L. Genetic and Epigenetic Determinants of AML Pathogenesis. Semin. Hematol. 2019, 56, 84–89. [Google Scholar] [CrossRef]
  11. Wang, R.; Yang, X.; Liu, J.; Zhong, F.; Zhang, C.; Chen, Y.; Sun, T.; Ji, C.; Ma, D. Gut Microbiota Regulates Acute Myeloid Leukaemia via Alteration of Intestinal Barrier Function Mediated by Butyrate. Nat. Commun. 2022, 13, 2522. [Google Scholar] [CrossRef]
  12. Yu, D.; Yu, X.; Ye, A.; Xu, C.; Li, X.; Geng, W.; Zhu, L. Profiling of Gut Microbial Dysbiosis in Adults with Myeloid Leukemia. FEBS Open Bio 2021, 11, 2050–2059. [Google Scholar] [CrossRef]
  13. Chen, G.; Kuang, Z.; Li, F.; Li, J. The Causal Relationship between Gut Microbiota and Leukemia: A Two-Sample Mendelian Randomization Study. Front. Microbiol. 2023, 14, 1293333. [Google Scholar] [CrossRef] [PubMed]
  14. Acute Myeloid Leukemia (AML): Practice Essentials, Pathophysiology, Etiology. Available online: https://emedicine.medscape.com/article/197802-overview (accessed on 20 October 2024).
  15. Franklin, S.; Aitken, S.L.; Shi, Y.; Sahasrabhojane, P.V.; Robinson, S.; Peterson, C.B.; Daver, N.; Ajami, N.A.; Kontoyiannis, D.P.; Shelburne, S.A.; et al. Oral and Stool Microbiome Coalescence and Its Association With Antibiotic Exposure in Acute Leukemia Patients. Front. Cell. Infect. Microbiol. 2022, 12, 848580. [Google Scholar] [CrossRef]
  16. Rashidi, A.; Ebadi, M.; Rehman, T.U.; Elhusseini, H.; Halaweish, H.F.; Kaiser, T.; Holtan, S.G.; Khoruts, A.; Weisdorf, D.J.; Staley, C. Lasting Shift in the Gut Microbiota in Patients with Acute Myeloid Leukemia. Blood Adv. 2022, 6, 3451–3457. [Google Scholar] [CrossRef]
  17. Salvestrini, V.; Conti, G.; D’Amico, F.; Cristiano, G.; Candela, M.; Cavo, M.; Turroni, S.; Curti, A. Gut Microbiome as a Potential Marker of Hematologic Recovery Following Induction Therapy in Acute Myeloid Leukemia Patients. Cancer Med. 2025, 14, e70501. [Google Scholar] [CrossRef] [PubMed]
  18. Goswami, M.; Bose, P.D. Gut Microbial Dysbiosis in the Pathogenesis of Leukemia: An Immune-Based Perspective. Exp. Hematol. 2024, 133, 104211. [Google Scholar] [CrossRef] [PubMed]
  19. 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]
  20. McMahon, S.; Sahasrabhojane, P.; Kim, J.; Franklin, S.; Chang, C.-C.; Jenq, R.R.; Hillhouse, A.E.; Shelburne, S.A.; Galloway-Peña, J. Contribution of the Oral and Gastrointestinal Microbiomes to Bloodstream Infections in Leukemia Patients. Microbiol. Spectr. 2023, 11, e00415-23. [Google Scholar] [CrossRef] [PubMed]
  21. 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 (1979) 2013, 342, 971–976. [Google Scholar] [CrossRef]
  22. 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]
  23. Ciernikova, S.; Sevcikova, A.; Mladosievicova, B.; Mego, M. Microbiome in Cancer Development and Treatment. Microorganisms 2023, 12, 24. [Google Scholar] [CrossRef] [PubMed]
  24. Acute Myeloid Leukemia—Guidelines Detail. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1411 (accessed on 28 May 2025).
  25. 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] [PubMed]
  26. Rashidi, A.; Kaiser, T.; Shields-Cutler, R.; Graiziger, C.; Holtan, S.G.; Rehman, T.U.; Wasko, J.; Weisdorf, D.J.; Dunny, G.; Khoruts, A.; et al. Dysbiosis Patterns during Re-Induction/Salvage versus Induction Chemotherapy for Acute Leukemia. Sci. Rep. 2019, 9, 6083. [Google Scholar] [CrossRef] [PubMed]
  27. Rattanathammethee, T.; Tuitemwong, P.; Thiennimitr, P.; Sarichai, P.; Na Pombejra, S.; Piriyakhuntorn, P.; Hantrakool, S.; Chai-Adisaksopha, C.; Rattarittamrong, E.; Tantiworawit, A.; et al. Gut Microbiota Profiles of Treatment-Naïve Adult Acute Myeloid Leukemia Patients with Neutropenic Fever during Intensive Chemotherapy. PLoS ONE 2020, 15, e0236460. [Google Scholar] [CrossRef]
  28. Xu, J.; Kang, Y.; Zhong, Y.; Ye, W.; Sheng, T.; Wang, Q.; Zheng, J.; Yang, Q.; Yi, P.; Li, Z. Alteration of Gut Microbiome and Correlated Amino Acid Metabolism Are Associated with Acute Myelocytic Leukemia Carcinogenesis. Cancer Med. 2023, 12, 16431–16443. [Google Scholar] [CrossRef]
  29. Hueso, T.; Ekpe, K.; Mayeur, C.; Gatse, A.; Joncquel-Chevallier Curt, M.; Gricourt, G.; Rodriguez, C.; Burdet, C.; Ulmann, G.; Neut, C.; et al. Impact and Consequences of Intensive Chemotherapy on Intestinal Barrier and Microbiota in Acute Myeloid Leukemia: The Role of Mucosal Strengthening. Gut Microbes 2020, 12, 1800897. [Google Scholar] [CrossRef]
  30. Pötgens, S.A.; Lecop, S.; Havelange, V.; Li, F.; Neyrinck, A.M.; Neveux, N.; Maertens, J.; Walter, J.; Schoemans, H.; Delzenne, N.M.; et al. Gut Microbiota Alterations Induced by Intensive Chemotherapy in Acute Myeloid Leukaemia Patients Are Associated with Gut Barrier Dysfunction and Body Weight Loss. Clin. Nutr. 2023, 42, 2214–2228. [Google Scholar] [CrossRef]
  31. Li, J.; Chen, X.; Zhao, S.; Chen, J. Arsenic-Containing Medicine Treatment Disturbed the Human Intestinal Microbial Flora. Toxics 2023, 11, 458. [Google Scholar] [CrossRef]
  32. Renga, G.; Nunzi, E.; Stincardini, C.; Pariano, M.; Puccetti, M.; Pieraccini, G.; Di Serio, C.; Fraziano, M.; Poerio, N.; Oikonomou, V.; et al. CPX-351 Exploits the Gut Microbiota to Promote Mucosal Barrier Function, Colonization Resistance and Immune Homeostasis. Blood J. 2024, 143, 1628–1645. [Google Scholar] [CrossRef]
  33. Liu, W.; Yang, J.; Chen, Y.; Chen, S.; Lu, L.; Li, J.; Li, J.; Liu, W.; Yang, T.; Zhang, G.; et al. Combining 16s rRNA and LC-QTOF-MS/MS to Explore the Temporal Changes of the Gut Microbiota and Metabolome with Clinical Characteristics of AML Patients. Available online: https://ssrn.com/abstract=5048723 (accessed on 15 July 2025).
  34. Kulecka, M.; Czarnowski, P.; Bałabas, A.; Turkot, M.; Kruczkowska-Tarantowicz, K.; Żeber-Lubecka, N.; Dąbrowska, M.; Paszkiewicz-Kozik, E.; Walewski, J.; Ługowska, I.; et al. Microbial and Metabolic Gut Profiling across Seven Malignancies Identifies Fecal Faecalibacillus Intestinalis and Formic Acid as Commonly Altered in Cancer Patients. Int. J. Mol. Sci. 2024, 25, 8026. [Google Scholar] [CrossRef]
  35. Campagna, A.; Clasen, F.; Portlock, T.; Zampini, M.; Ficara, F.; Crisafulli, L.; Brindisi, M.; Martano, G.; Saba, E.; Ubezio, M.; et al. Clinical Relevance of the Integrative Analysis of Gut Microbiome and Metabolomics in Myeloid Neoplasms: Correlations with Genomic Profiles, Treatment Response/Complications and Clinical Outcome. Blood 2024, 144, 104. [Google Scholar] [CrossRef]
  36. 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]
  37. Shouval, R.; Waters, N.R.; Gomes, A.L.C.; Zuanelli Brambilla, C.; Fei, T.; Devlin, S.M.; Nguyen, C.L.; Markey, K.A.; Dai, A.; Slingerland, J.B.; et al. Conditioning Regimens Are Associated with Distinct Patterns of Microbiota Injury in Allogeneic Hematopoietic Cell Transplantation. Clin. Cancer Res. 2023, 29, 165–173. [Google Scholar] [CrossRef] [PubMed]
  38. Jørgensen, M.; Nørgaard, J.C.; Ilett, E.E.; Marandi, R.Z.; Noguera-Julian, M.; Paredes, R.; Murray, D.D.; Lundgren, J.; MacPherson, C.R.; Sengeløv, H. Metabolic Potential of the Gut Microbiome Is Significantly Impacted by Conditioning Regimen in Allogeneic Hematopoietic Stem Cell Transplantation Recipients. Int. J. Mol. Sci. 2022, 23, 11115. [Google Scholar] [CrossRef]
  39. Goudarzi, M.; Mak, T.D.; Jacobs, J.P.; Moon, B.H.; Strawn, S.J.; Braun, J.; Brenner, D.J.; Fornace, A.J.; Li, H.H. An Integrated Multi-Omic Approach to Assess Radiation Injury on the Host-Microbiome Axis. Radiat Res. 2016, 186, 219. [Google Scholar] [CrossRef]
  40. Zhao, T.-S.; Xie, L.-W.; Cai, S.; Xu, J.-Y.; Zhou, H.; Tang, L.-F.; Yang, C.; Fang, S.; Li, M.; Tian, Y. Dysbiosis of Gut Microbiota Is Associated With the Progression of Radiation-Induced Intestinal Injury and Is Alleviated by Oral Compound Probiotics in Mouse Model. Front. Cell. Infect. Microbiol. 2021, 11, 717636. [Google Scholar] [CrossRef]
  41. Schwabkey, Z.I.; Wiesnoski, D.H.; Chang, C.-C.; Tsai, W.-B.; Pham, D.; Ahmed, S.S.; Hayase, T.; Ortega Turrubiates, M.R.; El-Himri, R.K.; Sanchez, C.A.; et al. Diet-Derived Metabolites and Mucus Link the Gut Microbiome to Fever after Cytotoxic Cancer Treatment. Sci. Transl. Med. 2022, 14, eabo3445. [Google Scholar] [CrossRef]
  42. Lu, L.; Li, F.; Gao, Y.; Kang, S.; Li, J.; Guo, J. Microbiome in Radiotherapy: An Emerging Approach to Enhance Treatment Efficacy and Reduce Tissue Injury. Mol. Med. 2024, 30, 105. [Google Scholar] [CrossRef]
  43. Rashidi, A.; Kaiser, T.; Graiziger, C.; Holtan, S.G.; Rehman, T.U.; Weisdorf, D.J.; Dunny, G.M.; Khoruts, A.; Staley, C. Gut Dysbiosis during Antileukemia Chemotherapy versus Allogeneic Hematopoietic Cell Transplantation. Cancer 2020, 126, 1434–1447. [Google Scholar] [CrossRef]
  44. Gu, Z.; Xiong, Q.; Wang, L.; Wang, L.; Li, F.; Hou, C.; Dou, L.; Zhu, B.; Liu, D. The Impact of Intestinal Microbiota in Antithymocyte Globulin–Based Myeloablative Allogeneic Hematopoietic Cell Transplantation. Cancer 2022, 128, 1402–1410. [Google Scholar] [CrossRef]
  45. 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. [Google Scholar] [CrossRef]
  46. 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. [Google Scholar] [CrossRef] [PubMed]
  47. 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] [PubMed]
  48. 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] [PubMed]
  49. 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]
  50. Haak, B.W.; Littmann, E.R.; Chaubard, J.L.; Pickard, A.J.; Fontana, E.; Adhi, F.; Gyaltshen, Y.; Ling, L.; Morjaria, S.M.; Peled, J.U.; et al. Impact of Gut Colonization with Butyrate-Producing Microbiota on Respiratory Viral Infection Following Allo-HCT. Blood 2018, 131, 2978–2986. [Google Scholar] [CrossRef]
  51. 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, ofz252. [Google Scholar] [CrossRef]
  52. 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]
  53. D’Amico, F.; Soverini, M.; Zama, D.; Consolandi, C.; Severgnini, M.; Prete, A.; Pession, A.; Barone, M.; Turroni, S.; Biagi, E.; et al. Gut Resistome Plasticity in Pediatric Patients Undergoing Hematopoietic Stem Cell Transplantation. Sci. Rep. 2019, 9, 5649. [Google Scholar] [CrossRef]
  54. 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]
  55. Stoma, I.; Littmann, E.R.; Peled, J.U.; Giralt, S.; van den Brink, M.R.M.; Pamer, E.G.; Taur, Y. Compositional Flux Within the Intestinal Microbiota and Risk for Bloodstream Infection With Gram-Negative Bacteria. Clin. Infect. Dis. 2021, 73, e4627–e4635. [Google Scholar] [CrossRef] [PubMed]
  56. Taplitz, R.A.; Kennedy, E.B.; Bow, E.J.; Crews, J.; Gleason, C.; Langston, A.A.; Flowers, C.R.; Hawley, D.K.; Nastoupil, L.J.; Rolston, K.V.; et al. Antimicrobial Prophylaxis for Adult Patients With Cancer-Related Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update. J. Clin. Oncol. 2018, 36, 3043–3054. [Google Scholar] [CrossRef] [PubMed]
  57. Wren, C.M.; Cowper, J.; Greer, N.; Goldin, L.; Perry, A. Effect of Reduced Fluoroquinolone Use on Cephalosporin Use, Susceptibilities and Clostridioides Difficile Infections. Antibiotics 2022, 11, 1312. [Google Scholar] [CrossRef]
  58. Mikulska, M.; Tissot, F.; Cordonnier, C.; Akova, M.; Calandra, T.; Ceppi, M.; Bruzzi, P.; Viscoli, C.; Aljurf, M.; Averbuch, D.; et al. Fluoroquinolone Prophylaxis in Haematological Cancer Patients with Neutropenia: ECIL Critical Appraisal of Previous Guidelines. J. Infect. 2018, 76, 20–37. [Google Scholar] [CrossRef]
  59. Ponziani, F.R.; Zocco, M.A.; D’Aversa, F.; Pompili, M.; Gasbarrini, A. Eubiotic Properties of Rifaximin: Disruption of the Traditional Concepts in Gut Microbiota Modulation. World J. Gastroenterol. 2017, 23, 4491. [Google Scholar] [CrossRef]
  60. Resources. Available online: https://www.ecil-leukaemia.com/en/resources/resources-ecil (accessed on 28 May 2025).
  61. Hayase, E.; Hayase, T.; Chang, C.-C.; Miyama, T.; Karmouch, J.L.; Tsai, W.-B.; Jamal, M.A.; Jenq, R.R. Carbapenem Antibiotics Promote Mucus Degradation By Bacteroides and Aggravate Graft-Versus-Host Disease. Blood 2021, 138, 85. [Google Scholar] [CrossRef]
  62. Messina, J.A.; Tan, C.Y.; Ren, Y.; Hill, L.; Bush, A.; Lew, M.; Andermann, T.; Peled, J.U.; Gomes, A.; van den Brink, M.R.M.; et al. Enterococcus Intestinal Domination Is Associated With Increased Mortality in the Acute Leukemia Chemotherapy Population. Clin. Infect. Dis. 2024, 78, 414–422. [Google Scholar] [CrossRef]
  63. Meschiari, M.; Kaleci, S.; Del Monte, M.; Dessilani, A.; Santoro, A.; Scialpi, F.; Franceschini, E.; Orlando, G.; Cervo, A.; Monica, M.; et al. Vancomycin Resistant Enterococcus Risk Factors for Hospital Colonization in Hematological Patients: A Matched Case-Control Study. Antimicrob. Resist. Infect. Control 2023, 12, 126. [Google Scholar] [CrossRef]
  64. Janjusevic, A.; Cirkovic, I.; Minic, R.; Stevanovic, G.; Soldatovic, I.; Mihaljevic, B.; Vidovic, A.; Markovic Denic, L. Predictors of Vancomycin-Resistant Enterococcus spp. Intestinal Carriage Among High-Risk Patients In University Hospitals In Serbia. Antibiotics 2022, 11, 1228. [Google Scholar] [CrossRef]
  65. Saad, R.; Rizkallah, M.R.; Aziz, R.K. Gut Pharmacomicrobiomics: The Tip of an Iceberg of Complex Interactions between Drugs and Gut-Associated Microbes. Gut Pathog. 2012, 4, 16. [Google Scholar] [CrossRef]
  66. Pant, A.; Maiti, T.K.; Mahajan, D.; Das, B. Human Gut Microbiota and Drug Metabolism. Microb. Ecol. 2023, 86, 97–111. [Google Scholar] [CrossRef]
  67. 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 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed]
  68. Zimmermann, M.; Zimmermann-Kogadeeva, M.; Wegmann, R.; Goodman, A.L. Mapping Human Microbiome Drug Metabolism by Gut Bacteria and Their Genes. Nature 2019, 570, 462–467. [Google Scholar] [CrossRef]
  69. Smith, M.; Dai, A.; Ghilardi, G.; Amelsberg, K.V.; Devlin, S.M.; Pajarillo, R.; Slingerland, J.B.; Beghi, S.; Herrera, P.S.; Giardina, P.; et al. Gut Microbiome Correlates of Response and Toxicity Following Anti-CD19 CAR T Cell Therapy. Nat. Med. 2022, 28, 713–723. [Google Scholar] [CrossRef]
  70. Zhou, B.; Xia, X.; Wang, P.; Chen, S.; Yu, C.; Huang, R.; Zhang, R.; Wang, Y.; Lu, L.; Yuan, F.; et al. Induction and Amelioration of Methotrexate-Induced Gastrointestinal Toxicity Are Related to Immune Response and Gut Microbiota. EBioMedicine 2018, 33, 122–133. [Google Scholar] [CrossRef]
  71. 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] [PubMed]
  72. 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]
  73. Zhao, B.; Zhou, B.; Dong, C.; Zhang, R.; Xie, D.; Tian, Y.; Yang, L. Lactobacillus Reuteri Alleviates Gastrointestinal Toxicity of Rituximab by Regulating the Proinflammatory T Cells in Vivo. Front. Microbiol. 2021, 12, 645500. [Google Scholar] [CrossRef]
  74. Zhao, L.; Xing, C.; Sun, W.; Hou, G.; Yang, G.; Yuan, L. Lactobacillus Supplementation Prevents Cisplatin-Induced Cardiotoxicity Possibly by Inflammation Inhibition. Cancer Chemother. Pharmacol. 2018, 82, 999–1008. [Google Scholar] [CrossRef] [PubMed]
  75. Turk, S.; Yanpar, H.; Baesmat, A.S.; Canli, S.D.; Cinar, O.E.; Malkan, U.Y.; Turk, C.; Haznedaroglu, I.C.; Ucar, G. Enterotoxins A and B Produced by Staphylococcus Aureus Increase Cell Proliferation, Invasion and Cytarabine Resistance in Acute Myeloid Leukemia Cell Lines. Heliyon 2023, 9, e19743. [Google Scholar] [CrossRef]
  76. Hakim, H.; Dallas, R.; Wolf, J.; Tang, L.; Schultz-Cherry, S.; Darling, V.; Johnson, C.; Karlsson, E.A.; Chang, T.-C.; Jeha, S.; et al. Gut Microbiome Composition Predicts Infection Risk During Chemotherapy in Children With Acute Lymphoblastic Leukemia. Clin. Infect. Dis. 2018, 67, 541–548. [Google Scholar] [CrossRef] [PubMed]
  77. De Pietri, S.; Ingham, A.C.; Frandsen, T.L.; Rathe, M.; Krych, L.; Castro-Mejía, J.L.; Nielsen, D.S.; Nersting, J.; Wehner, P.S.; Schmiegelow, K.; et al. Gastrointestinal Toxicity during Induction Treatment for Childhood Acute Lymphoblastic Leukemia: The Impact of the Gut Microbiota. Int. J. Cancer 2020, 147, 1953–1962. [Google Scholar] [CrossRef] [PubMed]
  78. 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. [Google Scholar] [CrossRef] [PubMed]
  79. Yue, X.Y.; Zhou, H.; Wang, S.F.; Chen, X.; Xiao, H.W. Gut Microbiota, Microbiota-derived Metabolites, and Graft-versus-host Disease. Cancer Med. 2024, 13, e6799. [Google Scholar] [CrossRef]
  80. Ilett, E.E.; Jørgensen, M.; Noguera-Julian, M.; Nørgaard, J.C.; Daugaard, G.; Helleberg, M.; Paredes, R.; Murray, D.D.; Lundgren, J.; MacPherson, C.; et al. Associations of the Gut Microbiome and Clinical Factors with Acute GVHD in Allogeneic HSCT Recipients. Blood Adv. 2020, 4, 5797–5809. [Google Scholar] [CrossRef]
  81. 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 (1979) 2019, 366, 1143–1149. [Google Scholar] [CrossRef]
  82. Fujiwara, H.; Docampo, M.D.; Riwes, M.; Peltier, D.; Toubai, T.; Henig, I.; Wu, S.J.; Kim, S.; Taylor, A.; Brabbs, S.; et al. Microbial Metabolite Sensor GPR43 Controls Severity of Experimental GVHD. Nat. Commun. 2018, 9, 3674. [Google Scholar] [CrossRef]
  83. 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. [Google Scholar] [CrossRef]
  84. Spanogiannopoulos, P.; Bess, E.N.; Carmody, R.N.; Turnbaugh, P.J. The Microbial Pharmacists within Us: A Metagenomic View of Xenobiotic Metabolism. Nat. Rev. Microbiol. 2016, 14, 273–287. [Google Scholar] [CrossRef]
  85. Haiser, H.J.; Gootenberg, D.B.; Chatman, K.; Sirasani, G.; Balskus, E.P.; Turnbaugh, P.J. Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella Lenta. Science 2013, 341, 295–298. [Google Scholar] [CrossRef]
  86. Forslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Krogh Pedersen, H.; et al. Disentangling Type 2 Diabetes and Metformin Treatment Signatures in the Human Gut Microbiota. Nature 2015, 528, 262–266. [Google Scholar] [CrossRef]
  87. Klastersky, J.; de Naurois, J.; Rolston, K.; Rapoport, B.; Maschmeyer, G.; Aapro, M.; Herrstedt, J. on behalf of the ESMO Guidelines Committee Management of Febrile Neutropaenia: ESMO Clinical Practice Guidelines. Ann. Oncol. 2016, 27, v111–v118. [Google Scholar] [CrossRef] [PubMed]
  88. Shbaklo, N.; Vicentini, C.; Busca, A.; Giaccone, L.; Dellacasa, C.; Dogliotti, I.; Lupia, T.; Zotti, C.M.; Corcione, S.; De Rosa, F.G. Cost-Effectiveness of Targeted Prophylaxis among Allogenic Stem Cell Transplant Recipients. Pharmaceuticals 2023, 16, 466. [Google Scholar] [CrossRef] [PubMed]
  89. Slavin, M.A.; Lingaratnam, S.; Mileshkin, L.; Booth, D.L.; Cain, M.J.; Ritchie, D.S.; Wei, A.; Thursky, K.A. Use of Antibacterial Prophylaxis for Patients with Neutropenia. Australian Consensus Guidelines 2011 Steering Committee. Intern. Med. J. 2011, 41, 102–109. [Google Scholar] [CrossRef] [PubMed]
  90. Averbuch, D.; Orasch, C.; Cordonnier, C.; Livermore, D.M.; Mikulska, M.; Viscoli, C.; Gyssens, I.C.; Kern, W.V.; Klyasova, G.; Marchetti, O.; et al. European Guidelines for Empirical Antibacterial Therapy for Febrile Neutropenic Patients in the Era of Growing Resistance: Summary of the 2011 4th European Conference on Infections in Leukemia. Haematologica 2013, 98, 1826–1835. [Google Scholar] [CrossRef] [PubMed]
  91. Webb, B.J.; Majers, J.; Healy, R.; Jones, P.B.; Butler, A.M.; Snow, G.; Forsyth, S.; Lopansri, B.K.; Ford, C.D.; Hoda, D. Antimicrobial Stewardship in a Hematological Malignancy Unit: Carbapenem Reduction and Decreased Vancomycin-Resistant Enterococcus Infection. Clin. Infect. Dis. 2020, 71, 960–967. [Google Scholar] [CrossRef]
  92. Schauwvlieghe, A.; Dunbar, A.; Storme, E.; Vlak, A.; Aerts, R.; Maertens, J.; Sciot, B.; Van Der Wel, T.; Papageorgiou, G.; Moors, I.; et al. Stopping Antibiotic Therapy after 72 h in Patients with Febrile Neutropenia Following Intensive Chemotherapy for AML/MDS (Safe Study): A Retrospective Comparative Cohort Study. EClinicalMedicine 2021, 35, 100855. [Google Scholar] [CrossRef]
  93. de Gunzburg, J.; Ducher, A.; Modess, C.; Wegner, D.; Oswald, S.; Dressman, J.; Augustin, V.; Feger, C.; Andremont, A.; Weitschies, W.; et al. Targeted Adsorption of Molecules in the Colon with the Novel Adsorbent-Based Medicinal Product, DAV132: A Proof of Concept Study in Healthy Subjects. J. Clin. Pharmacol. 2015, 55, 10–16. [Google Scholar] [CrossRef] [PubMed]
  94. Connelly, S.; Fanelli, B.; Hasan, N.A.; Colwell, R.R.; Kaleko, M. Oral Metallo-Beta-Lactamase Protects the Gut Microbiome From Carbapenem-Mediated Damage and Reduces Propagation of Antibiotic Resistance in Pigs. Front. Microbiol. 2019, 10, 101. [Google Scholar] [CrossRef]
  95. Jing, N.; Wang, L.; Zhuang, H.; Jiang, G.; Liu, Z. Ultrafine Jujube Powder Enhances the Infiltration of Immune Cells during Anti-PD-L1 Treatment against Murine Colon Adenocarcinoma. Cancers 2021, 13, 3987. [Google Scholar] [CrossRef]
  96. 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 2018, 359, 91–97. [Google Scholar] [CrossRef]
  97. Leles, G.; Quintanilha, M.F.; Souza, R.O.; Miranda, V.C.; Rocha, V.M.; Trindade, L.M.; Jesus, L.C.L.; Mendes, V.; Andre, L.C.; D’Auriol-Souza, M.M.; et al. Effects of Dietary Fibre Intake in Chemotherapy-Induced Mucositis in Murine Model. Br. J. Nutr. 2021, 126, 853–864. [Google Scholar] [CrossRef]
  98. Wang, C.; Yang, S.; Gao, L.; Wang, L.; Cao, L. Carboxymethyl Pachyman (CMP) Reduces Intestinal Mucositis and Regulates the Intestinal Microflora in 5-Fluorouracil-Treated CT26 Tumour-Bearing Mice. Food Funct. 2018, 9, 2695–2704. [Google Scholar] [CrossRef]
  99. Fernandes, A.; Oliveira, A.; Carvalho, A.L.; Soares, R.; Barata, P. Faecalibacterium Prausnitzii in Differentiated Thyroid Cancer Patients Treated with Radioiodine. Nutrients 2023, 15, 2680. [Google Scholar] [CrossRef] [PubMed]
  100. Forslund, S.K. Fasting Intervention and Its Clinical Effects on the Human Host and Microbiome. J. Intern. Med. 2022, 293, 166–183. [Google Scholar] [CrossRef] [PubMed]
  101. Dong, J.; Wang, B.; Xiao, Y.; Liu, J.; Wang, Q.; Xiao, H.; Jin, Y.; Liu, Z.; Chen, Z.; Li, Y.; et al. Roseburia Intestinalis Sensitizes Colorectal Cancer to Radiotherapy through the Butyrate/OR51E1/RALB Axis. Cell Rep. 2024, 43, 113846. [Google Scholar] [CrossRef] [PubMed]
  102. Kong, C.; Yan, X.; Liu, Y.; Huang, L.; Zhu, Y.; He, J.; Gao, R.; Kalady, M.F.; Goel, A.; Qin, H.; et al. Ketogenic Diet Alleviates Colitis by Reduction of Colonic Group 3 Innate Lymphoid Cells through Altering Gut Microbiome. Signal Transduct. Target. Ther. 2021, 6, 154. [Google Scholar] [CrossRef]
  103. Cagigas, M.L.; Rajakumar, G.; Skarratt, K.; Hayward, R.; Pelaia, T.; Ardjmand, A.; Vaughan, L.; Fontana, L.; Fuller, S. PB1796: ADMINISTRATION OF A KETOGENIC DIET FOR CHEMOTHERAPY PROTECTION IN ACUTE LEUKEMIA PATIENTS: IS IT FEASIBLE, SAFE, AND VALUABLE? PRELIMINARY RESULTS OF THE RANDOMIZED CONTROLLED TRIAL LEU-KETO STUDY. Hemasphere 2023, 7, e189423f. [Google Scholar] [CrossRef]
  104. Spencer, C.N.; McQuade, J.L.; Gopalakrishnan, V.; McCulloch, J.A.; Vetizou, M.; Cogdill, A.P.; Khan, M.A.W.; Zhang, X.; White, M.G.; Peterson, C.B.; et al. Dietary Fiber and Probiotics Influence the Gut Microbiome and Melanoma Immunotherapy Response. Science (1979) 2021, 374, 1632–1640. [Google Scholar] [CrossRef]
  105. Nagpal, R.; Shively, C.A.; Appt, S.A.; Register, T.C.; Michalson, K.T.; Vitolins, M.Z.; Yadav, H. Gut Microbiome Composition in Non-Human Primates Consuming a Western or Mediterranean Diet. Front. Nutr. 2018, 5, 28. [Google Scholar] [CrossRef]
  106. Pagliai, G.; Russo, E.; Niccolai, E.; Dinu, M.; Di Pilato, V.; Magrini, A.; Bartolucci, G.; Baldi, S.; Menicatti, M.; Giusti, B.; et al. Influence of a 3-Month Low-Calorie Mediterranean Diet Compared to the Vegetarian Diet on Human Gut Microbiota and SCFA: The CARDIVEG Study. Eur. J. Nutr. 2020, 59, 2011–2024. [Google Scholar] [CrossRef] [PubMed]
  107. Tavil, B.; Koksal, E.; Yalcin, S.S.; Uckan, D. Pretransplant Nutritional Habits and Clinical Outcome in Children Undergoing Hematopoietic Stem Cell Transplant. Exp. Clin. Transplant. 2012, 10, 55–61. [Google Scholar] [CrossRef] [PubMed]
  108. Matteucci, S.; De Pasquale, G.; Pastore, M.; Morenghi, E.; Pipitone, V.; Soekeland, F.; Caccialanza, R.; Mazzoleni, B.; Mancin, S. Low-Bacterial Diet in Cancer Patients: A Systematic Review. Nutrients 2023, 15, 3171. [Google Scholar] [CrossRef] [PubMed]
  109. Food Safety for Older Adults and People with Cancer, Diabetes, HIV/AIDS, Organ Transplants, and Autoimmune Diseases|FDA. Available online: https://www.fda.gov/food/people-risk-foodborne-illness/food-safety-older-adults-and-people-cancer-diabetes-hivaids-organ-transplants-and-autoimmune (accessed on 27 October 2024).
  110. Ciernikova, S.; Sevcikova, A.; Drgona, L.; Mego, M. Modulating the Gut Microbiota by Probiotics, Prebiotics, Postbiotics, and Fecal Microbiota Transplantation: An Emerging Trend in Cancer Patient Care. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2023, 1878, 188990. [Google Scholar] [CrossRef]
  111. Iyer, C.; Kosters, A.; Sethi, G.; Kunnumakkara, A.B.; Aggarwal, B.B.; Versalovic, J. Probiotic Lactobacillus Reuteri Promotes TNF-Induced Apoptosis in Human Myeloid Leukemia-Derived Cells by Modulation of NF-KappaB and MAPK Signalling. Cell Microbiol. 2008, 10, 1442–1452. [Google Scholar] [CrossRef]
  112. Wang, Y.; Xue, J.; Zhou, X.; You, M.; Du, Q.; Yang, X.; He, J.; Zou, J.; Cheng, L.; Li, M.; et al. Oral Microbiota Distinguishes Acute Lymphoblastic Leukemia Pediatric Hosts from Healthy Populations. PLoS ONE 2014, 9, e102116. [Google Scholar] [CrossRef]
  113. Ghoneum, M.; Gimzewski, J. Apoptotic Effect of a Novel Kefir Product, PFT, on Multidrug-Resistant Myeloid Leukemia Cells via a Hole-Piercing Mechanism. Int. J. Oncol. 2014, 44, 830–837. [Google Scholar] [CrossRef]
  114. Liu, J.; Luo, W.; Chen, Q.; Chen, X.; Zhou, G.; Sun, H. Curcumin Sensitizes Response to Cytarabine in Acute Myeloid Leukemia by Regulating Intestinal Microbiota. Cancer Chemother. Pharmacol. 2022, 89, 243–253. [Google Scholar] [CrossRef]
  115. Kwak, S.-H.; Cho, Y.-M.; Noh, G.-M.; Om, A.-S. Cancer Preventive Potential of Kimchi Lactic Acid Bacteria (Weissella cibaria, Lactobacillus plantarum). J. Cancer Prev. 2014, 19, 253. [Google Scholar] [CrossRef]
  116. Ambesh, P.; Stroud, S.; Franzova, E.; Gotesman, J.; Sharma, K.; Wolf, L.; Kamholz, S. Recurrent Lactobacillus Bacteremia in a Patient With Leukemia. J. Investig. Med. High Impact Case Rep. 2017, 5, 2324709617744233. [Google Scholar] [CrossRef]
  117. Martín, R.; Langella, P. Emerging Health Concepts in the Probiotics Field: Streamlining the Definitions. Front. Microbiol. 2019, 10, 1047. [Google Scholar] [CrossRef]
  118. Balendra, V.; Rosenfeld, R.; Amoroso, C.; Castagnone, C.; Rossino, M.G.; Garrone, O.; Ghidini, M. Postbiotics as Adjuvant Therapy in Cancer Care. Nutrients 2024, 16, 2400. [Google Scholar] [CrossRef] [PubMed]
  119. Mederle, A.L.; Semenescu, A.; Drăghici, G.A.; Dehelean, C.A.; Vlăduț, N.V.; Nica, D.V. Sodium Butyrate: A Multifaceted Modulator in Colorectal Cancer Therapy. Medicina 2025, 61, 136. [Google Scholar] [CrossRef] [PubMed]
  120. Condoluci, A.; Rossi, D. Mechanisms of Resistance to Venetoclax. Blood 2022, 140, 2094–2096. [Google Scholar] [CrossRef]
  121. Pulliam, S.R.; Pellom, S.T.; Shanker, A.; Adunyah, S.E. Butyrate Regulates the Expression of Inflammatory and Chemotactic Cytokines in Human Acute Leukemic Cells during Apoptosis. Cytokine 2016, 84, 74–87. [Google Scholar] [CrossRef]
  122. Kawakatsu, R.; Tadagaki, K.; Yamasaki, K.; Yoshida, T. Venetoclax Efficacy on Acute Myeloid Leukemia Is Enhanced by the Combination with Butyrate. Sci. Rep. 2024, 14, 4975. [Google Scholar] [CrossRef]
  123. Yoshida, T.; Yamasaki, K.; Tadagaki, K.; Kuwahara, Y.; Matsumoto, A.; Sofovic, A.E.; Kondo, N.; Sakai, T.; Okuda, T. Tumor Necrosis Factor related Apoptosis inducing Ligand Is a Novel Transcriptional Target of Runt related Transcription Factor 1. Int. J. Oncol. 2022, 60, 6. [Google Scholar] [CrossRef]
  124. Reyna-Figueroa, J.; Barrón-Calvillo, E.; García-Parra, C.; Galindo-Delgado, P.; Contreras-Ochoa, C.; Lagunas-Martínez, A.; Campos-Romero, F.H.; Silva-Estrada, J.A.; Limón-Rojas, A.E. Probiotic Supplementation Decreases Chemotherapy-Induced Gastrointestinal Side Effects in Patients With Acute Leukemia. J. Pediatr. Hematol. Oncol. 2019, 41, 468–472. [Google Scholar] [CrossRef]
  125. Wada, M.; Nagata, S.; Saito, M.; Shimizu, T.; Yamashiro, Y.; Matsuki, T.; Asahara, T.; Nomoto, K. Effects of the Enteral Administration of Bifidobacterium Breve on Patients Undergoing Chemotherapy for Pediatric Malignancies. Support. Care Cancer 2010, 18, 751–759. [Google Scholar] [CrossRef] [PubMed]
  126. Iyama, S.; Sato, T.; Tatsumi, H.; Hashimoto, A.; Tatekoshi, A.; Kamihara, Y.; Horiguchi, H.; Ibata, S.; Ono, K.; Murase, K.; et al. Efficacy of Enteral Supplementation Enriched with Glutamine, Fiber, and Oligosaccharide on Mucosal Injury Following Hematopoietic Stem Cell Transplantation. Case Rep. Oncol. 2014, 7, 692. [Google Scholar] [CrossRef]
  127. Yoshifuji, K.; Inamoto, K.; Kiridoshi, Y.; Takeshita, K.; Sasajima, S.; Shiraishi, Y.; Yamashita, Y.; Nisaka, Y.; Ogura, Y.; Takeuchi, R.; et al. Prebiotics Protect against Acute Graft-versus-Host Disease and Preserve the Gut Microbiota in Stem Cell Transplantation. Blood Adv. 2020, 4, 4607–4617. [Google Scholar] [CrossRef] [PubMed]
  128. Sharma, A.; Tilak, T.; Bakhshi, S.; Raina, V.; Kumar, L.; Chaudhary, S.P.; Sahoo, R.K.; Gupta, R.; Thulkar, S. Lactobacillus Brevis CD2 Lozenges Prevent Oral Mucositis in Patients Undergoing High Dose Chemotherapy Followed by Haematopoietic Stem Cell Transplantation. ESMO Open 2016, 1, 138. [Google Scholar] [CrossRef] [PubMed]
  129. Guo, J.; Zhang, H.; Lu, X.; Xia, L. Viable Bifidobacterium Tablets for the Prevention of Chemotherapy-/Radiation-Induced Mucositis in Patients Undergoing Haematopoietic Stem Cell Transplantation. Support. Care Cancer 2023, 31, 282. [Google Scholar] [CrossRef] [PubMed]
  130. 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, e12947. [Google Scholar] [CrossRef]
  131. 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] [PubMed]
  132. Hill, G.R.; Ferrara, J.L.M. The Primacy of the Gastrointestinal Tract as a Target Organ of Acute Graft-versus-Host Disease: Rationale for the Use of Cytokine Shields in Allogeneic Bone Marrow Transplantation. Blood 2000, 95, 2754–2759. [Google Scholar] [CrossRef]
  133. Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; De los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and Their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 180861. [Google Scholar] [CrossRef]
  134. Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef]
  135. Van der Velden, W.J.F.M.; Herbers, A.H.E.; Netea, M.G.; Blijlevens, N.M.A. Mucosal Barrier Injury, Fever and Infection in Neutropenic Patients with Cancer: Introducing the Paradigm Febrile Mucositis. Br. J. Haematol. 2014, 167, 441–452. [Google Scholar] [CrossRef]
  136. Cheng, H.; Guan, X.; Chen, D.; Ma, W. The Th17/Treg Cell Balance: A Gut Microbiota-Modulated Story. Microorganisms 2019, 7, 583. [Google Scholar] [CrossRef]
  137. Vaishnava, S.; Behrendt, C.L.; Ismail, A.S.; Eckmann, L.; Hooper, L.V. Paneth Cells Directly Sense Gut Commensals and Maintain Homeostasis at the Intestinal Host-Microbial Interface. Proc. Natl. Acad. Sci. USA 2008, 105, 20858–20863. [Google Scholar] [CrossRef]
  138. Koyama, M.; Hill, G.R. Alloantigen Presentation and Graft-versus-Host Disease: Fuel for the Fire. Blood 2016, 127, 2963–2970. [Google Scholar] [CrossRef] [PubMed]
  139. 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]
  140. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The Microbial Metabolite Butyrate Regulates Intestinal Macrophage Function via Histone Deacetylase Inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef]
  141. 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]
  142. Todor, S.B.; Ichim, C. Microbiome Modulation in Pediatric Leukemia: Impact on Graft-Versus-Host Disease and Treatment Outcomes: A Narrative Review. Children 2025, 12, 166. [Google Scholar] [CrossRef]
  143. Pinzon-Leal, P.; Gutierrez-Barbosa, H.; Medina-Moreno, S.; Zapata, J.C. The Microbiome, Inflammation, and GVHD Axis: The Balance Between the “Gut” and the Bad. Immuno 2025, 5, 10. [Google Scholar] [CrossRef]
  144. 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]
  145. Nigam, M.; Panwar, A.S.; Singh, R.K. Orchestrating the Fecal Microbiota Transplantation: Current Technological Advancements and Potential Biomedical Application. Front. Med. Technol. 2022, 4, 961569. [Google Scholar] [CrossRef]
  146. Bieganska, E.A.; Kosinski, P.; Wolski, M. Possible Applications of Fecal Microbiota Transplantation in the Pediatric Population: A Systematic Review. Biomedicines 2025, 13, 1393. [Google Scholar] [CrossRef] [PubMed]
  147. Zerdan, M.B.; Niforatos, S.; Nasr, S.; Nasr, D.; Ombada, M.; John, S.; Dutta, D.; Lim, S.H. Fecal Microbiota Transplant for Hematologic and Oncologic Diseases: Principle and Practice. Cancers 2022, 14, 691. [Google Scholar] [CrossRef]
  148. Mayer, E.F.; Maron, G.; Dallas, R.H.; Ferrolino, J.; Tang, L.; Sun, Y.; Danziger-Isakov, L.; Paulsen, G.C.; Fisher, B.T.; Vora, S.B.; et al. A Multicenter Study to Define the Epidemiology and Outcomes of Clostridioides Difficile Infection in Pediatric Hematopoietic Cell and Solid Organ Transplant Recipients. Am. J. Transplant. 2020, 20, 2133–2142. [Google Scholar] [CrossRef]
  149. Webb, B.J.; Brunner, A.; Ford, C.D.; Gazdik, M.A.; Petersen, F.B.; Hoda, D. Fecal Microbiota Transplantation for Recurrent Clostridium Difficile Infection in Hematopoietic Stem Cell Transplant Recipients. Transpl. Infect. Dis. 2016, 18, 628–633. [Google Scholar] [CrossRef] [PubMed]
  150. Seong, H.; Lee, S.K.; Cheon, J.H.; Yong, D.E.; Koh, H.; Kang, Y.K.; Jeong, W.Y.; Lee, W.J.; Sohn, Y.; Cho, Y.; et al. Fecal Microbiota Transplantation for Multidrug-Resistant Organism: Efficacy and Response Prediction. J. Infect. 2020, 81, 719–725. [Google Scholar] [CrossRef] [PubMed]
  151. Patel, P.; Robinson, P.D.; Fisher, B.T.; Phillips, R.; Morgan, J.E.; Lehrnbecher, T.; Kuczynski, S.; Koenig, C.; Haeusler, G.M.; Esbenshade, A.; et al. Guideline for the Management of Clostridioides Difficile Infection in Pediatric Patients with Cancer and Hematopoietic Cell Transplantation Recipients: 2024 Update. EClinicalMedicine 2024, 72, 102604. [Google Scholar] [CrossRef]
  152. Kelly, C.R.; Ihunnah, C.; Fischer, M.; Khoruts, A.; Surawicz, C.; Afzali, A.; Aroniadis, O.; Barto, A.; Borody, T.; Giovanelli, A.; et al. Fecal Microbiota Transplant for Treatment of Clostridium Difficile Infection in Immunocompromised Patients. Am. J. Gastroenterol. 2014, 109, 1065–1071. [Google Scholar] [CrossRef] [PubMed]
  153. Conover, K.R.; Absah, I.; Ballal, S.; Brumbaugh, D.; Cho, S.; Cardenas, M.C.; Knackstedt, E.D.; Goyal, A.; Jensen, M.K.; Kaplan, J.L.; et al. Fecal Microbiota Transplantation for Clostridioides Difficile Infection in Immunocompromised Pediatric Patients. J. Pediatr. Gastroenterol. Nutr. 2023, 76, 440–446. [Google Scholar] [CrossRef]
  154. Nallar, S.C.; Xu, D.Q.; Kalvakolanu, D.V. Bacteria and Genetically Modified Bacteria as Cancer Therapeutics: Current Advances and Challenges. Cytokine 2017, 89, 160–172. [Google Scholar] [CrossRef]
  155. Forbes, N.S. Engineering the Perfect (Bacterial) Cancer Therapy. Nat. Rev. Cancer 2010, 10, 785–794. [Google Scholar] [CrossRef]
  156. Wang, J.; Ghosh, D.; Maniruzzaman, M. Using Bugs as Drugs: Administration of Bacteria-Related Microbes to Fight Cancer. Adv. Drug. Deliv. Rev. 2023, 197, 114825. [Google Scholar] [CrossRef]
  157. Mayakrishnan, V.; Kannappan, P.; Tharmalingam, N.; Bose, R.J.C.; Madheswaran, T.; Ramasamy, M. Bacterial Cancer Therapy: A Turning Point for New Paradigms. Drug. Discov. Today 2022, 27, 2043–2050. [Google Scholar] [CrossRef]
  158. Li, M.; Lu, M.; Lai, Y.; Zhang, X.; Li, Y.; Mao, P.; Liang, Z.; Mu, Y.; Lin, Y.; Zhao, A.Z.; et al. Inhibition of Acute Leukemia with Attenuated Salmonella Typhimurium Strain VNP20009. Biomed. Pharmacother. 2020, 129, 110425. [Google Scholar] [CrossRef]
  159. Cuozzo, M.; Castelli, V.; Avagliano, C.; Cimini, A.; D’angelo, M.; Cristiano, C.; Russo, R. Effects of Chronic Oral Probiotic Treatment in Paclitaxel-Induced Neuropathic Pain. Biomedicines 2021, 9, 346. [Google Scholar] [CrossRef]
  160. Juan, Z.; Chen, J.; Ding, B.; Yongping, L.; Liu, K.; Wang, L.; Le, Y.; Liao, Q.; Shi, J.; Huang, J.; et al. Probiotic Supplement Attenuates Chemotherapy-Related Cognitive Impairment in Patients with Breast Cancer: A Randomised, Double-Blind, and Placebo-Controlled Trial. Eur. J. Cancer 2022, 161, 10–22. [Google Scholar] [CrossRef]
  161. Shen, S.; Lim, G.; You, Z.; DIng, W.; Huang, P.; Ran, C.; Doheny, J.; Caravan, P.; Tate, S.; Hu, K.; et al. Gut Microbiota Is Critical for the Induction of Chemotherapy-Induced Pain. Nat. Neurosci. 2017, 20, 1213–1216. [Google Scholar] [CrossRef]
  162. Whirl-Carrillo, M.; McDonagh, E.M.; Hebert, J.M.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Altman, R.B.; Klein, T.E. Pharmacogenomics Knowledge for Personalized Medicine. Clin. Pharmacol. Ther. 2012, 92, 414–417. [Google Scholar] [CrossRef]
  163. Hitchings, R.; Kelly, L. Predicting and Understanding the Human Microbiome’s Impact on Pharmacology. Trends Pharmacol. Sci. 2019, 40, 495. [Google Scholar] [CrossRef]
  164. Mima, K.; Kosumi, K.; Baba, Y.; Hamada, T.; Baba, H.; Ogino, S. The Microbiome, Genetics, and Gastrointestinal Neoplasms: The Evolving Field of Molecular Pathological Epidemiology to Analyze the Tumor-Immune-Microbiome Interaction. Hum. Genet. 2021, 140, 725–746. [Google Scholar] [CrossRef]
  165. Trac, Q.T. Statistical and Computational Methodologies for Omics Data Analyses and Drug Response Prediction; Karolinska Institutet: Stockholm, Sweden, 2024. [Google Scholar] [CrossRef]
  166. Chen, Y.; Wu, F.H.; Wu, P.Q.; Xing, H.Y.; Ma, T. The Role of The Tumor Microbiome in Tumor Development and Its Treatment. Front. Immunol. 2022, 13, 935846. [Google Scholar] [CrossRef]
  167. Fu, A.; Yao, B.; Dong, T.; Chen, Y.; Yao, J.; Liu, Y.; Li, H.; Bai, H.; Liu, X.; Zhang, Y.; et al. Tumor-Resident Intracellular Microbiota Promotes Metastatic Colonization in Breast Cancer. Cell 2022, 185, 1356–1372.e26. [Google Scholar] [CrossRef]
  168. Castillo, D.J.; Rifkin, R.F.; Cowan, D.A.; Potgieter, M. The Healthy Human Blood Microbiome: Fact or Fiction? Front. Cell Infect. Microbiol. 2019, 9, 449041. [Google Scholar] [CrossRef]
  169. Cheng, H.S.; Tan, S.P.; Wong, D.M.K.; Koo, W.L.Y.; Wong, S.H.; Tan, N.S. The Blood Microbiome and Health: Current Evidence, Controversies, and Challenges. Int. J. Mol. Sci. 2023, 24, 5633. [Google Scholar] [CrossRef]
  170. Woerner, J.; Huang, Y.; Hutter, S.; Gurnari, C.; Sánchez, J.M.H.; Wang, J.; Huang, Y.; Schnabel, D.; Aaby, M.; Xu, W.; et al. Circulating Microbial Content in Myeloid Malignancy Patients Is Associated with Disease Subtypes and Patient Outcomes. Nat. Commun. 2022, 13, 1038. [Google Scholar] [CrossRef]
  171. Tan, C.C.S.; Ko, K.K.K.; Chen, H.; Liu, J.; Loh, M.; Chia, M.; Nagarajan, N. No Evidence for a Common Blood Microbiome Based on a Population Study of 9770 Healthy Humans. Nat. Microbiol. 2023, 8, 973–985. [Google Scholar] [CrossRef]
Figure 1. Critical timepoints of microbiome disruption and alteration during the clinical course of AML patients. Red circles indicate critical disruption points for the microbiome in AML patients where targeted therapeutic intervention and future research may be most beneficial (hospital administration, treatment, neutropenic fever, and infection). AML: acute myeloid leukemia; GM: gut microbiome; SCFAs: short-chain fatty acids.
Figure 1. Critical timepoints of microbiome disruption and alteration during the clinical course of AML patients. Red circles indicate critical disruption points for the microbiome in AML patients where targeted therapeutic intervention and future research may be most beneficial (hospital administration, treatment, neutropenic fever, and infection). AML: acute myeloid leukemia; GM: gut microbiome; SCFAs: short-chain fatty acids.
Biomedicines 13 01761 g001
Figure 2. Treatment algorithm of AML according to NCCN 2025 recommendation. After initial disease biology and clinical evaluation, patients are stratified to intensive or lower-intensity therapy, followed by consolidation with/without HSCT and maintenance. Therapy options are presented. AML: acute myeloid leukemia; allo-HSCT: allogenic hematopoietic stem cell transplantation; CMML: chronic myelomonocytic leukemia; CPX-351: liposomal formulation of a fixed combination of daunorubicin and cytarabine; FLT3: FMS-like tyrosine kinase 3; G-CSF: granulocyte colony-stimulating factor; GO: gemtuzumab ozogamicin; HMA: hypomethylating agent; IC: intensive chemotherapy; IDH: isocitrate dehydrogenase; IVO: ivosidenib; LDAC: low-dose cytarabine; MDS: myelodysplastic syndrome; VEN: venetoclax.
Figure 2. Treatment algorithm of AML according to NCCN 2025 recommendation. After initial disease biology and clinical evaluation, patients are stratified to intensive or lower-intensity therapy, followed by consolidation with/without HSCT and maintenance. Therapy options are presented. AML: acute myeloid leukemia; allo-HSCT: allogenic hematopoietic stem cell transplantation; CMML: chronic myelomonocytic leukemia; CPX-351: liposomal formulation of a fixed combination of daunorubicin and cytarabine; FLT3: FMS-like tyrosine kinase 3; G-CSF: granulocyte colony-stimulating factor; GO: gemtuzumab ozogamicin; HMA: hypomethylating agent; IC: intensive chemotherapy; IDH: isocitrate dehydrogenase; IVO: ivosidenib; LDAC: low-dose cytarabine; MDS: myelodysplastic syndrome; VEN: venetoclax.
Biomedicines 13 01761 g002
Figure 3. Interactions between microbiome alterations, AML treatment, and clinical outcomes. A comprehensive overview of the inter-relationships between microbiome-disrupting factors, AML treatment, resulting dysbiosis, and associated clinical outcomes, with an emphasis on potential underlying mechanisms. AML: acute myeloid leukemia; GVHD: graft-versus-host disease; HSCT: hematopoietic stem cell transplantation.
Figure 3. Interactions between microbiome alterations, AML treatment, and clinical outcomes. A comprehensive overview of the inter-relationships between microbiome-disrupting factors, AML treatment, resulting dysbiosis, and associated clinical outcomes, with an emphasis on potential underlying mechanisms. AML: acute myeloid leukemia; GVHD: graft-versus-host disease; HSCT: hematopoietic stem cell transplantation.
Biomedicines 13 01761 g003
Figure 4. Current challenges, emerging directions, and future applications of the microbiome in AML treatment. Orange rectangles highlight key limitations—methodology: (1) variability in treatment regimens and drugs, (2) lack of adjustment for confounding factors such as diet and antibiotic use, (3) discrepancies in sequencing technologies and analytical pipelines, and (4) absence of standardized diagnostic criteria; safety: microbiome-based interventions in immunocompromised individuals; data interpretation: (1) high inter-individual and population variability and (2) uncertainty regarding microbial viability; and clinical relevance. Green rectangles outline key research needs and directions—study design: (1) rigorous experimental frameworks, (2) multi-spatial and temporal assessment, (3) in vitro and in vivo models, (4) longitudinal cohorts, (5) mechanistic investigations, and (6) clinical interventional trials; non-bacterial microbiome: (1) archaea, (2) fungi, and (3) viruses; bone marrow and blood microbiome; microbial therapy in AML: (1) immunotherapeutic applications utilizing live, attenuated, or genetically modified bacteria, either alone or with conventional treatments, (2) targeted delivery of anticancer agents, (3) bacterial expression of tumor-specific antigens, (4) delivery or expression of suppressor genes, anti-angiogenic genes, and suicide genes, (5) RNA interference mechanisms, and (6) activation of pro-drugs through bacterial cleavage; microbiome in novel AML treatment methods: (1) immunotherapy and (2) targeted therapies; toxicomicrobiomics: (1) neuropathy, (2) cardiotoxicity, (3) hepatotoxicity, and (4) mental disorder; microbiology-MPE; multi-omics approaches: (1) genomics, (2) epigenomics, (3) transcriptomics, (4) proteomics, and (5) metabolomics; drug–response and toxicity prognostic models; chemotherapy resistance; targeted microbiome modulation; microbiome-related dietary modalities; and antibiotic stewardship. Blue circles represent future prospects for microbiome integration in AML treatment: personalized treatment; improved therapeutic outcomes; reduction of adverse effects. Microbiology-MPE: microbiology–molecular pathological epidemiology.
Figure 4. Current challenges, emerging directions, and future applications of the microbiome in AML treatment. Orange rectangles highlight key limitations—methodology: (1) variability in treatment regimens and drugs, (2) lack of adjustment for confounding factors such as diet and antibiotic use, (3) discrepancies in sequencing technologies and analytical pipelines, and (4) absence of standardized diagnostic criteria; safety: microbiome-based interventions in immunocompromised individuals; data interpretation: (1) high inter-individual and population variability and (2) uncertainty regarding microbial viability; and clinical relevance. Green rectangles outline key research needs and directions—study design: (1) rigorous experimental frameworks, (2) multi-spatial and temporal assessment, (3) in vitro and in vivo models, (4) longitudinal cohorts, (5) mechanistic investigations, and (6) clinical interventional trials; non-bacterial microbiome: (1) archaea, (2) fungi, and (3) viruses; bone marrow and blood microbiome; microbial therapy in AML: (1) immunotherapeutic applications utilizing live, attenuated, or genetically modified bacteria, either alone or with conventional treatments, (2) targeted delivery of anticancer agents, (3) bacterial expression of tumor-specific antigens, (4) delivery or expression of suppressor genes, anti-angiogenic genes, and suicide genes, (5) RNA interference mechanisms, and (6) activation of pro-drugs through bacterial cleavage; microbiome in novel AML treatment methods: (1) immunotherapy and (2) targeted therapies; toxicomicrobiomics: (1) neuropathy, (2) cardiotoxicity, (3) hepatotoxicity, and (4) mental disorder; microbiology-MPE; multi-omics approaches: (1) genomics, (2) epigenomics, (3) transcriptomics, (4) proteomics, and (5) metabolomics; drug–response and toxicity prognostic models; chemotherapy resistance; targeted microbiome modulation; microbiome-related dietary modalities; and antibiotic stewardship. Blue circles represent future prospects for microbiome integration in AML treatment: personalized treatment; improved therapeutic outcomes; reduction of adverse effects. Microbiology-MPE: microbiology–molecular pathological epidemiology.
Biomedicines 13 01761 g004
Table 1. Medications for AML.
Table 1. Medications for AML.
GroupTypesDrugs
ChemotherapyAntimetabolitesCytarabine
Fludarabine
Cladribine
Clofarabine
Thioguanine
Decitabine
Methotrexate
Azacitidine
alkylating agentsCyclophosphamide
anti-microtubule agentsvincristine
topoisomerase inhibitorsdoxorubicin
mitoxantrone
cytotoxic antibioticsidarubicin
daunorubicin
mitoxantrone
doxorubicin
targeted therapyinhibitor of the b-cell lymphoma 2 regulator proteinvenetoclax
fms-related tyrosine kinase 3 inhibitorsmidostaurin
quizartinib
gilteritinib
sorafenib
menin inhibitorrevumenib
isocitrate dehydrogenase inhibitorsenasidenib
ivosidenib
olutasidenib
antibody drug conjugategemtuzumab ozogamicin
hedgehog pathway inhibitorglasdegib
Otherhypomethylating agentsazacitidine
decitabine
dexamethasone
Steroidsprednisone
Retinoidsall-trans retinoic acid
arsenic trioxide
hydroxycarbamide
combinations of antineoplastic agentsdaunorubicin hydrochloride and cytarabine liposome
cytarabine, daunorubicin hydrochloride, and etoposide phosphate
Table 2. Summary of studies related to the effect of antibiotics on the microbiome in AML.
Table 2. Summary of studies related to the effect of antibiotics on the microbiome in AML.
RefStudy GroupSampling Time PointMethodology (Material)AntibioticsUseComparatorMain Results
Alpha DiversityCompositionResistomeMetabolites
[46]n = 94 HSCT recipients (AML n = 44)serial analyses from before HSCT to 35 days post-HSCT16S ribosomal RNA gene sequencing (fecal specimens)vancomycinPtemporal variations of microbiota profilesn/an/a
ciprofloxacin, levofloxacinPtemporal variations of microbiota profiles10-fold ↓ in Proteobacteria dominancen/an/a
metronidazoleTtemporal variations of microbiota profiles3-fold ↑ in enterococcal dominancen/an/a
cephalosporins, beta-lactam–beta-lactamase combinations, carbapenemsTtemporal variations of microbiota profilesn/an/a
[47]n = 34 HSCT recipients (AML n = 14)serial analyses from before HSCT to day 28 post-HSCTnext-generation sequencing (fecal specimens), strain-specific enterococcal PCR (fecal specimens), liquid chromatography–tandem mass spectrometry of urinary IS (urine)trimethoprim/sulfamethoxazole followed by ciprofloxacin + metronidazolePtemporal variations of microbiota profiles ↑ in the proportion of E. faecalis and E. faecium; ↓ in other Firmicutes and phylan/a↓ 3-IS levels
systemic antibioticsTtemporal variations of microbiota profiles↑ mainly in E. faecalis and, to a lesser extent, E. faeciumn/a↓ 3-IS levels
[48]n = 857 HSCT recipients (AML n = 277)prior to and after the initiation of a specific antibiotic treatment16S ribosomal RNA gene sequencing (fecal specimens)vancomycin + ciprofloxacinPcomparison with othern/aminor perturbationsn/an/a
iv or oral trimethoprim/sulfamethoxazoleTcomparison with othern/aminor perturbationsn/an/a
oral atovaquoneTcomparison with othern/aminor perturbationsn/an/a
imipenem–cilastatinTcomparison with othern/aminor perturbationsn/an/a
piperacillin–tazobactamTcomparison with othern/agreater ↓ in Bacteroidetes and Lactobacillus; trend towards a ↓ in Clostridia and Actinobacteria (no statistical significance); ↓ in Enterococcus, Akkermansia, and Erysipelotrichia (similar to aztreonam and cefepime)n/an/a
aztreonamTcomparison with othern/a↓ in Enterococcus, Akkermansia, and Erysipelotrichia (similar to piperacillin–tazobactam)n/an/a
cefepimeTcomparison with othern/a↓ in Enterococcus, Akkermansia, and Erysipelotrichia (similar to piperacillin–tazobactam)n/an/a
[49]n = 394 HSCT recipients (acute leukemia n = 219)serial analyses from before conditioning and weekly within the first 28 days after HSCT3-IS (urine), 16S ribosomal RNA gene sequencing (fecal specimens)rifaximinPciprofloxacin–metronidazolen/a↓ in E. faecium and E. faecalisn/a↑ 3-IS levels
[50]n = 360 HSCT recipients (AML n = 197)serial analyses from before HSCT to the time of stem cell engraftment16S ribosomal RNA gene sequencing (fecal specimens), concentrations of SCFAs using targeted metabolomics methodology (fecal samples)metronidazoleTtemporal variations of microbiota profilesn/a↓ in butyrate-producing bacterian/abutyrate, acetate, propionate, and desamino-tyrosine correlated with the abundance of butyrate-producing bacteria
beta-lactamsTtemporal variations of microbiota profilesn/a↓ in butyrate-producing bacterian/a
vancomycinTtemporal variations of microbiota profilesn/an/a
fluoroquinolonesTtemporal variations of microbiota profilesn/an/a
[51]n = 60 (AML n = 26)within 7 days after antibiotics exposure16S ribosomal RNA gene sequencing (fecal specimens)levofloxacinPno antibioticstrend toward ↓ dominance of non-Bacteroidetes; ↓ in Proteobacteria; ↑ in Lachnospiraceae, Ruminococcaceae, Blautian/an/a
oral vancomycinPno antibiotics↓ in Bacteroidetes; ↑ risk of dominance of non-Bacteroidetesn/an/a
cefepime, piperacillin–tazobactam, meropenemTno antibioticstrend toward ↑ Enterococcus; ↓ in Clostridia and Blautian/an/a
[52]n = 161 HSCT recipients (AML n = 87)serial analyses within the first 10 days after HSCT3-IS (urine), 16S ribosomal RNA gene sequencing (fecal specimens)rifaximinPrifaximin with/withouTsystemic antibiotics vs. ciprofloxacin–metronidazole with/without systemic antibiotics↑ in Clostridium cluster XIVa (CCXIVa) abundance; ↓ in enterococcal load (not statistically significant)n/a↑ 3-IS levels
ciprofloxacin + metronidazolePtemporal variations of microbiota profilesdomination of Akkermansia, Eubacterium, or Enterococcusn/a↓ 3-IS levels
piperacillin–tazobactam, meropenem + vancomycin, ceftazidime, vancomycinTtemporal variations of microbiota profilesdomination of Akkermansia, Eubacterium, or Enterococcusn/a↓ 3-IS levels (except vancomycin alone)
systemic antibioticsTtemporal variations of microbiota profiles↑ mainly in E. faecalis and, to a lesser extent, E. faeciumn/a↓ 3-IS levels
[53]n = 8 HSCT recipients (AML n = 8)serial analyses from before HSCT to 85 days post-HSCTWGS metagenome sequencing (fecal specimens)fluoroquinolonesPtemporal variations of the gut resistome in each individualn/aARU26—↑ in Bacteroides sp. D1, Prevotella intermedia, Capnocytophaga ochracea, and Bacteroides fragilis species; ARU38—↑ in B. fragilis and Bacteroides sp.↑ trend for AMR genes: ARU4 (tetracycline inhibitor), ARU26 (β-lactamase CFXA3), and ARU38 (erythromycin resistance); consolidation of AMR genes present before transplanting and acquisition of new AMR genes, particularly in aGvHD-positive patients, extending beyond the antibiotics used during treatmentn/a
beta-lactamsTtemporal variations of the gut resistome in each individualn/a n/a
[54]n = 97 AMLserial analyses from baseline to neutrophil recovery during induction chemotherapy16S ribosomal RNA gene sequencing (oral swabs and fecal specimens)carbapenemTtemporal variations of microbiota profiles↓ when carbapenems for >72 hn/an/an/a
cephalosporinTtemporal variations of microbiota profilesn/an/an/a
piperacillin–tazobactamTtemporal variations of microbiota profilesn/an/an/a
[43]n = 20 HSCT recipients (AML n = 10); n = 20 intensively treated acute leukemia (AML = 16)acute leukemia—serial analyses from day 1 of chemotherapy until day 28 or discharge; HSCT—serial analyses from the day of transplantation until day 14 after transplantation.16S ribosomal RNA gene sequencing (fecal specimens)levofloxacinPcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibiotics↓ in Enterococcus domination (both groups)n/an/a
cephalosporins third-generation or higherTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibiotics↑ in Lactobacillus domination (acute leukemia); ↑ in Enterococcus domination (acute leukemia)n/an/a
iv vancomycinTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibiotics↑ in Lactobacillus domination (acute leukemia)n/an/a
oral vancomycinTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibioticsno impact on dominationn/an/a
piperacillin–tazobactamTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibioticsno impact on dominationn/an/a
carbapenemsTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibioticsno impact on dominationn/an/a
metronidazol/clindamycinTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibiotics↑ in Enterococcus domination (acute leukemia)n/an/a
linezolid/daptomycinTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibioticsno impact on dominationn/an/a
cefepime + iv vancomycinTcomparison of variations of microbiota profiles in 2 cohorts of patients↓ in both cohorts; n/a for particular antibioticsno impact on dominationn/an/a
[55]n = 708 HSCT recipients (AML n = 360)serial analyses from the start of pretransplant conditioning until engraftment16S ribosomal RNA gene sequencing (fecal specimens)ciprofloxacinPno antibioticsn/a↓ in BSIs and intestinal colonization by Gram-negative microbes, including Klebsiella, Citrobacter, Enterobacter, and Desulfovibrio; ↑ in Escherichia, Pseudomonas, and Stenotrophomonas; ↑ in breakthrough with E. coli in intestinal colonization and BSIs↑ in fluoroquinolone resistancen/a
AMR: antimicrobial resistance. BSIs: bloodstream infections. IS: indoxyl sulfate. n/a: not applicable. P: prophylaxis. SCFA: short-chain fatty acids. T: treatment. WGS: whole-genome shotgun. ↓: decreased. ↔: stable. ↑: increased.
Table 3. Completed and ongoing clinical trials on the microbiome in AML.
Table 3. Completed and ongoing clinical trials on the microbiome in AML.
ClinicalTrials.Gov IDPurposeInterventionOutcome MeasureStudy PopulationStudy DesignStateResults
NCT06899581to assess the impact of leukemia treatment on GM and its recovery trajectorydietary supplement: enteral nutrition food-derived ingredientGM, alpha and beta-diversity, Firmicutes to Bacteroidetes ratio, compositionChildobservational, case–control, prospectiveRecruitingno results posted
NCT02928523 (ODYSSEE)to assess autologous FMT efficacy in preventing dysbiosis complications in AML patients receiving intensive treatmentautologous FMT MaaT011 post-chemotherapydysbiosis correction (microbiota alpha and beta diversity), MDRB eradication (bacterial culture), biological parametersAdultinterventional,
phase 1 and
phase 2, single group assignment, open label
completedrestoration of GM diversity and community
recovery of microbial richness and diversity to baseline levels
safety
NCT03959241to assess whether GM diversity at neutrophil engraftment predicts one-year non-relapse mortality in patients receiving reduced-intensity allo-HSCTtacrolimus/methotrexate versus post-transplant cyclophosphamide/tacrolimus/mycophenolate mofetil in non-myeloablative/reduced intensity conditioning allogeneic peripheral blood stem cell transplantationpercentage of participants with GVHD/relapse or progression-free survival at one yearAdultinterventional, phase 3, randomized, parallel assignment 1:1, open labelcompletedno results on GM finding posted
NCT02949427to characterize oral and nasal microbiota, including fungi, before and after chemotherapy, and supportive carechemotherapy and supportive carediversity index of oronasal mycobiome and microbiome, relative abundance of the oronasal fungal microbiome and microbiome4 years to 21 yearsobservational, cohort, prospectivecompletedno results posted
NCT04940468to determine whether dietary intervention to increase fiber and decrease fat reduces C. difficile infection recurrence in a cohort of oncology patientsdiet higher in fiber and lower in fatC. difficile toxins A and B, fecal microbiome (16S rRNA, shotgun metagenomic sequencing)9 years and olderinterventional, randomized, parallel assignment, open labelrecruitingno results posted
NCT06355583to test the ability to restore GM to healthier levels in patients with blood cancers scheduled to have HSCTcapsule with communities of dried, intestinal microorganisms from screened, pooled human stool samples (IMT) swallowed 2 weeks before HSCTtolerability and acceptability of IMT, GM diversity (richness and evenness), health, infective/microbiological, and hematological outcomes (days of fever, admission to intensive care unit, survival, non-relapsed mortality, and incidence of GVHD)18 years and olderinterventional, phase 2, randomized, parallel assignment, masking: triple (participant, care provider, investigator)recruitingno results posted
NCT04214249to correlate microbial/metabolome changes at baseline and changes with clinical response (immune-checkpoint expression, kinetics of immune cell subset recovery, and programming) in the standard of care and experimental armpembrolizumab in combination with conventional intensive chemotherapy as frontline therapy in patients with AMLrate of MRD negative—complete response/complete remission with incomplete recovery, immune cell subsets, PD-1 and PD-L1 expression, protein signatures, T cell receptor sequencing, GM18 years to 75 yearsinterventional, phase 2, randomized, parallel assignment, open labelactive, not recruitingno results posted
NCT05596981to investigate the effect of sorafenib maintenance therapy in FLT3-ITD-positive AML patients after allo-HSCT on GMsorafenibvariation of GM composition and diversity (16s rRNA sequencing of serial stool samples), variation of gut barrier integrity (serum levels of zonulin, I-FABP, and citrulline or other potential candidates), treatment outcomes, GVHD18 years to 65 yearsobservational, cohort, perspectiverecruitingno results posted
NCT03678493to asses efficacy of FMT in AML patients and allo-HSCT recipients3 treatments of oral capsule FMT vs. placebo after each exposure to antibacterial antibioticsnumber of infections, FMT engraftment, GVHD18 years and olderinterventional, randomized, double-blind, placebo-controlled, open-labelcompletedsafety, intestinal dysbiosis amelioration, no decrease in infections
NCT04629430to see whether HSCT patients can consistently eat a diet rich in prebioticsprebiotic foods/drinksfrequency of participants ingesting the required diet, GVHD, incidence of C. difficile infection, patient weight, number of days to neutrophil engraftment18 years and olderinterventional, single group assignment, open labelcompletedno results posted
allo-HSCT, allogenic hematopoietic stem cell transplant; FMT, fecal microbiota transplantation; GM, gut microbiota; GVHD, graft-versus-host disease; MDRB, multidrug-resistant bacteria; MRD, minimal residual disease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nowicka, A.; Tomczak, H.; Szałek, E.; Karbownik, A.; Gil, L. Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine. Biomedicines 2025, 13, 1761. https://doi.org/10.3390/biomedicines13071761

AMA Style

Nowicka A, Tomczak H, Szałek E, Karbownik A, Gil L. Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine. Biomedicines. 2025; 13(7):1761. https://doi.org/10.3390/biomedicines13071761

Chicago/Turabian Style

Nowicka, Aneta, Hanna Tomczak, Edyta Szałek, Agnieszka Karbownik, and Lidia Gil. 2025. "Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine" Biomedicines 13, no. 7: 1761. https://doi.org/10.3390/biomedicines13071761

APA Style

Nowicka, A., Tomczak, H., Szałek, E., Karbownik, A., & Gil, L. (2025). Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine. Biomedicines, 13(7), 1761. https://doi.org/10.3390/biomedicines13071761

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