Abstract
The gut microbiota makes critical contributions to host homeostasis, and its role in the treatment of acute myeloid leukemia (AML) has attracted attention. We investigated whether the gut microbiome is affected by AML, and whether such changes are associated with hallmarks of cachexia. Biological samples and clinical data were collected from 30 antibiotic- free AML patients at diagnosis and matched volunteers (1:1) in a multicenter, cross-sectional, prospective study. The composition and functional potential of the fecal microbiota were analyzed using shotgun metagenomics. Fecal, blood, and urinary metabolomics analyses were performed. AML patients displayed muscle weakness, anorexia, signs of altered gut function, and glycemic disorders. The composition of the fecal microbiota differed between patients with AML and control subjects, with an increase in oral bacteria. Alterations in bacterial functions and fecal metabolome support an altered redox status in the gut microbiota, which may contribute to the altered redox status observed in patients with AML. Eubacterium eligens, reduced 3-fold in AML patients, was strongly correlated with muscle strength and citrulline, a marker of enterocyte mass and function. Blautia and Parabacteroides, increased in patients with AML, were correlated with anorexia. Several bacterial taxa and metabolites (e.g., Blautia, Prevotella, phenylacetate, and hippurate) previously associated with glycemic disorders were altered. Our work revealed important perturbations in the gut microbiome of AML patients at diagnosis, which are associated with muscle strength, altered redox status, and anorexia. These findings pave the way for future mechanistic work to explore the function and therapeutic potential of the bacteria identified in this study.
Introduction
The role of the gut microbiota in acute myeloid leukemia (AML) has attracted the attention of the scientific community. The complex microbiota community inhabiting the human gastrointestinal tract cultivates an intricate and mutually beneficial relationship with its host. The gut microbiota synthesizes essential metabolites and releases bioactive compounds, which can influence host physiology.1 Fifteen percent of the metabolites found in mammalian blood are derived from the gut microbiota,2 underlying the profound imbalance between the host and gut microbiota metabolism. The gut microbiota can also regulate host metabolism and immunity through the regulation of gut function.3 Therefore, gut microbiota is considered a key regulator of host metabolism and inflammatory status in different patho-physiological contexts, including metabolic syndrome and insulin resistance.4
AML is a clonal disorder of hematopoietic stem cells, resulting in impaired production of the myeloid blood cell lineage. The standard treatment for younger AML patients (<65 years) is intensive induction chemotherapy followed by consolidation chemotherapy and, in some cases, allogeneic hematopoietic stem cell transplantation. The gut microbiota seems to control aspects of hematopoiesis through mechanisms including metabolite production as well as circulating microbial DNA.5 AML is also characterized by an altered host immunity, this latter being widely influenced by the gut microbiota. Despite the expected links between AML and gut microbiota, little is known about the influence of AML on the composition of the gut microbiota, whereas more is known about the impact of chemotherapy.
Indeed, patients undergoing induction chemotherapy experience loss of microbiota diversity.6 ,7 The reduction in some beneficial bacterial species, such as Faecalibacterium prausnitzii and Bifidobacterium spp., was found to persist even after completion of chemotherapy.8 A higher microbial diversity at diagnosis was linked to a reduced risk of infectious complications following induction chemotherapy, suggesting that microbiota profiling could be useful in infectious risk stratification.9 Much of the recent work investigating the gut microbiota in the context of AML has focused on its relationship with the development of graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation. Higher α-diversity was associated with better survival10 and higher levels of Blautia with reduced GvHD-related mortality.11 The presence of Eubac-terium limosum is also associated with a reduced risk of disease relapse.12 During the post-transplant period, the presence of antibiotic-resistant bacteria or domination by some bacteria, such as Enterococcus spp. and Proteobacteria, is associated with a higher risk of infection.13,14 These observations have encouraged the evaluation of strategies to enhance microbial diversity. In this context, a recent study investigated the safety and diversity-enhancing ability of autologous fecal microbiota transfer in patients with AML receiving intensive chemotherapy and antibiotics.15 Fecal material collected at the time of diagnosis was used for fecal microbiota transfer. This transfer appeared to be safe and could restore microbial richness and diversity based on α-diversity indices. Many unknowns remain concerning the composition and activity of gut microbiota at diagnosis and its potential influence on the host in AML. In a mouse model of leukemia, we showed that gut microbiota composition was altered alongside the gut barrier and that reversing these changes through administration of probiotics improved features of cachexia.16,17 Cachexia is a multifactorial systemic syndrome characterized by substantial weight loss (coming from atrophy of the skeletal muscle and adipose tissue), often accompanied by anorexia, muscle weakness, insulin resistance, and inflammation, which occurs in approximately 50-80% of patients with cancer.18 Although cachexia affects 40% of patients with hematologic cancers and is often witnessed in clinics, studies focusing on AML-related cachexia are scarce.19 Therefore, we launched an academic, multicenter, cross-sectional, prospective study to evaluate gut microbiota composition and activity in AML patients and volunteers matched for age, sex, body mass index (BMI) and smoking status. Thirty patients newly diagnosed with AML were recruited before therapeutic intervention. We collected information on the patients’ food habits and hallmarks of cachexia. We also assessed the inflammatory status of the patients, as well as metabolic and gut function markers. Through a multiomics integrative approach combining metagenomics, fecal, blood, and urinary metabolomics, and clinical data, we investigated the gut microbiota of AML patients at diagnosis and links to hallmarks of cachexia.
Methods
Subjects
Thirty patients newly diagnosed with AML were recruited between December 2015 and December 2019 from three Belgian University hospitals (Saint-Luc Brussels [n=13], UZ Leuven [n=15], and UZ Gent [n=2]). The exclusion criteria included antibiotic consumption within the preceding 30 days, chronic intestinal diseases, obesity (BMI >30), pregnancy, gastric bypass, and treatment with antidiabetic or hypoglycemic drugs.
Control subjects from the general population were recruited between December 2017 and January 2020 based on the same inclusion/exclusion criteria, except for the AML diagnosis. They were matched (1:1) for age, sex, BMI, and smoking status, all factors known to affect the gut microbiota. This study followed the ethical guidelines set out in the Declaration of Helsinki, was approved by the “Comité d’éthique Hospitalo-facultaire des Cliniques Universitaires Saint-Luc” (B403201317128), and all participants provided written informed consent. A full description of the study design is provided in the Online Supplementary Supporting Information. The study was retrospectively registered on March 20, 2018 on Clinicaltrials.gov: NCT03881826.
Samples, data collection and analyses
All biological sampling and data collection were performed at diagnosis (i.e., before the initiation of chemotherapy and antibiotic treatment). The full details are provided in the Online Supplementary Supporting Information.
Biochemical, gut microbiota, proton nuclear magnetic resonance (1H-NMR) and statistical analyses are detailed in the Online Supplementary Supporting Information. Statistical significance was set at P<0.05 and q-value <0.1.
Results
Control subjects and acute myeloid leukemia patients have similar anthropometric characteristics and habits
As ensured by one-to-one matching, the group of controls and the group of AML patients did not show any differences in terms of age, sex, BMI, body composition, and percentage of smokers (Table 1). They also had similar habits in terms of daily alcohol consumption. The food frequency ques-tionnaire20 revealed no differences in the overall dietary scores. No differences in drug exposure, food supplement consumption, and transit scores were detected between the groups (Online Supplementary Table S1, Online Supplementary Figure S1).
Patients with acute myeloid leukemia have specific gut microbiota alterations
The composition of the gut microbiota was assessed in the same stool samples using two independent techniques (16S rRNA gene sequencing and metagenomics). Gut microbiota composition was not differentially clustered between controls and AML patients, as revealed by principal component analysis at the genus level (Figure 1A, Online Supplementary Figure S2A). Permutational multivariate analysis of variance (PERMANOVA) revealed that 3.1% of the variation in gut microbiota composition, as assessed at the genus level by shotgun metagenomics, was explained by AML (Figure 1A, B). At the species level, the disease explained 3.2% of the variation of the dataset (Online Supplementary Figure S3A). We also assessed the explanatory power of other factors previously shown to explain most of the variation in gut microbiota composition in the Belgian Flemish Gut Flora Project, a cohort of 1,106 Belgian volunteers,21 namely BMI, hemoglobin, and age (Figure 1B, Online Supplementary Figure S3B). Disease presence had the highest explanatory power, close to BMI (3.1% in this cohort and ~3% in the Belgian Flemish Gut Flora Project) while age and sex were not significant. Total bacterial levels were similar in the two groups (Online Supplementary Figure S2B), as were the α-diversity indices (Online Supplementary Figure S2C). However, the univariate analyses revealed specific changes. Three genera, namely Actinomyces, Blautia and Parabacteroides, and one species, Parvimonas micra, were increased in the feces of patients with AML, whereas Eu-bacterium eligens was decreased (Figure 1C). These changes, highlighted by shotgun metagenomics, were confirmed by 16S rRNA gene sequencing, with the exception of Blautia (Online Supplementary Figure S2D, Online Supplementary Table S2). Coprococcus and Prevotella were reduced in AML patients as assessed by 16S rRNA gene sequencing (q=0.109 using shotgun metagenomics) (Online Supplementary Figure S2D, Online Supplementary Table S2). The relative abundances of oral species increased more than two-fold, while obligate relative abundances of anaerobe genera were reduced (Figure 1D).
Principal component analysis of metagenomic functions (Figure 1E) confirmed the compositional analyses, with no clear global functional changes between the two groups. However, 22 functions were significantly different (Figure 1F, Online Supplementary Table S3). Of these 22 functions, only four were decreased in AML patients, whereas the others were increased. The contributions of the species and genera to each of these functions are shown in Online Supplementary Figure S4. A visual inspection of these graphs indicates that for the majority of the functions, the changes in their functional abundances observed in AML patients cannot be ascribed to specific species/genera, thereby suggesting that functional changes in the gut microbio-ta may be independent of compositional changes. These compositional and functional changes were sufficient to predict disease status in a testing set with an accuracy of 87% based on the top altered bacteria and 77% based on the altered functions using random forest models (Online Supplementary Figure S5).
Acute myeloid leukemia patients demonstrate anorexia, muscle weakness and glycemic disorders
Patients with AML displayed changes in hemoglobin concentration, white blood cell count, and C-reactive protein, albumin, and glycemia levels characteristic of the disease (Figure 2, Online Supplementary Figure S6). The higher C-re-active protein and lower albumin levels also led to a higher modified Glasgow prognostic score (Figure 2A). In addition to reflecting a higher inflammatory status, the modified Glasgow prognostic score has recently been associated with adverse outcomes in patients with newly diagnosed AML.22 Appetite score and muscle strength were reduced in patients with AML (Figure 2B). Fasted blood glucose and insulin levels, as well as HOMA-IR2, were higher in AML patients (Figure 2C), collectively reflecting and confirming alterations of glucose metabolism in AML.23 This appears especially the case for a subset of patients who, intriguingly, presented a specific microbial signature (Online Supplementary Figures S7 and S8; Online Supplementary Results and Discussion).
In addition to an increase in C-reactive protein, the levels of other inflammatory markers (i.e., interleukin [IL]6), IL8), monocyte chemoattractant protein 1, and tumor necrosis factor α) were elevated in AML patients as compared to the levels in control subjects (Figure 3A). Together with the increase in IL10 and reduction in transforming growth factor β1 (TGFβ1), this confirms alterations in cytokine levels in AML patients24 and a high inflammatory status. We also reported an increase in growth differentiation factor-15 (GDF15), a member of the TGFβ superfamily, and fibroblast growth factor 21 (FGF21) in AML patients (Figure 3B). GDF15 mediates tumor-induced anorexia and body weight loss25 while FGF21 has been implicated in fasting-induced muscle atrophy and weakness in mice.26
Plasma levels of citrulline, a non-proteinogenic amino acid reflecting enterocyte mass and function,27 were decreased in patients with AML (Figure 3C). Together with the increased level of lipopolysaccharide-binding protein (Figure 3C), the reduction in citrulline levels in AML patients suggests an alteration of gut function.
Patients with acute myeloid leukemia have specific fecal, blood and urinary metabolome alterations
Principal component analysis performed on fecal, blood, and urinary metabolites did not reveal a clear clustering between controls and AML (Online Supplementary Figure S9), with AML samples being more dispersed than the control samples. PERMANOVA revealed that 11.2% and 2.9% of the variation in blood and urine metabolomics, respectively, can be explained by the presence of AML (P<0.05). The metabolites driving the separation between both groups were identified using multivariate discriminant analysis (Online Supplementary Figure S9). Univariate analysis also revealed significantly altered metabolites (Figure 4, Online Supplementary Table S4). In AML feces, two sugars (glucose and galactose), three alcohols (methanol, ethanol, and glycerol) and two amino acids (phenylalanine and threonine) decreased. Bacterial amino acid-derived metabolites were modified: 3-phenylpropionate decreased, while phenylacetate increased. In AML blood, two sugars (glucose and mannose), as well as ethanol, increased. Amino acids (alanine, asparagine, citrulline, cystine, glutamine, methionine, sarcosine, and threonine) decreased, except for glutamate, which increased. Uridine and urea levels were decreased.
Lactate levels increased, while acetate, fumarate, and succinate levels decreased. Amino acid catabolic products, such as 3-hydroxyisovalerate, 2-hydroxybutyrate, formate, and dimethylamine, were increased. Ascorbate, also known as vitamin C, was decreased in AML patients. In the urine of AML patients, the metabolites were mainly increased. This was the case for most amino acids (2-aminobutyrate, 3-aminoisobutyrate, carnitine, isoleucine, leucine, taurine, threonine, and tyrosine) and nucleic base-related metabolites (hypoxanthine and pseudouridine), with the exception of uracil, which decreased. Formate, maleate, and Sumiki’s acid also increased, whereas hippurate and trigonelline decreased. Consistent with the alterations in glucose metabolism mentioned earlier, glucose levels were also increased in the AML urine samples.
Altered bacteria and functions in acute myeloid leukemia patients correlate with several fecal, blood and urinary metabolites
Next, we performed correlations between the top altered bacterial taxa/functions and fecal, blood, and urinary metabolite levels. The genus Eubacterium, as well as the associated species E. eligens and E. hallii, showed the strongest correlations with fecal metabolites, especially malonate (negative correlation) and glycerol (positive correlation) (Figure 5). Several blood metabolites were strongly correlated with the different bacterial taxa (Figure 5). Urea was negatively correlated with the Blautia genus and positively correlated with the Sutterellaceae family and E. eligens. Ascorbate levels were negatively correlated with Clostridium spiroforme. 3-hydroxyisobutyrate correlated negatively with the Erysipelatoclostridium genus, and 2-hydroxybutyrate correlated positively with the Parabacteroides genus. The correlations were weaker for urinary metabolites (Online Supplementary Figure S10). E. eligens displayed the highest number of correlations (all negative), mainly with amino acids. Parabacteroides also showed several positive correlations. Most of these correlations were maintained when corrected for age (Online Supplementary Figure S11). Among those correlations, fecal malonate and glycerol correlations with Eubacterium, E. eligens, and E. hallii were also found in the healthy control group alone (Online Supplementary Figure S12). In the healthy control group, blood urea also correlated with Blautia as well as E. eligens, and 3-hydroxyisobutyrate correlated with the Erysipelatoclostridium genus (Online Supplementary Figures S13 and S14).
Interestingly, when looking at the correlations between altered microbial functions and fecal, blood, and urinary metabolomes, some fecal and blood metabolites demonstrated strong correlations with many different functions (Online Supplementary Figure S15). For instance, in feces, glycerol showed strong negative correlations with almost all increased functions in the AML group. In blood, several metabolites showed multiple correlations with bacterial function. Some were positive (e.g., correlation with formate) and some were negative (e.g., correlations with ascorbate and propylene glycol). In contrast, ferredoxinnitrite reductase was negatively correlated with many urinay metabolites, whereas glycerol dehydratase was positively correlated with similar metabolites (Online Supplementary Figure S16).
Altered bacteria and functions in acute myeloid leukemia patients associate with clinical, metabolic and inflammatory parameters
We then looked for associations between altered bacterial taxa and functions and the collected clinical, dietary, inflammatory, and metabolic parameters in the whole cohort and exclusively in the AML group to delineate the strongest correlations. We found that many taxa, including Parvimonas micra, Actinomyces, Parabacteroides and Blautia, were positively associated with inflammatory markers in the entire cohort (Figure 6). E. eligens, decreased in AML patients, was negatively associated with inflammation and positively associated with citrulline and muscle strength; the latter correlation was maintained within the AML group (Online Supplementary Figure S17). Consistent with these findings, muscle strength was strongly correlated with blood citrulline (rho=0.3522, P=0.0058) and C-reactive protein levels (rho= -0.4585, P=0.0002). Looking at the negative correlations, we noticed that Blautia was inversely associated with muscle strength. Some parameters, such as hemoglobin, IL6 and IL8 as well as TGFβ1 and citrulline, were associated with many different functions (Online Supplementary Figure S18), with some associations maintained when considering exclusively the AML patients (Online Supplementary Figure S19). Most of these correlations were maintained when corrected for age (Online Supplementary Figure S20).
Discussion
This study highlights alterations in gut microbiota composition and activity, hallmarks of cachexia and metabolic and inflammatory disturbances in patients newly diagnosed with AML, with several associations found between these alterations (Figure 7).
Altered bacterial functions in AML patients include genes catalyzing redox reactions, suggesting an altered redox status in the gut microbiota of such patients. Two elements from fecal metabolomics further support the hypothesis of a redox imbalance in AML gut microbiota towards a more pro-oxidative status. First, phenylalanine, which is decreased in the feces of patients with AML, can be metabolized by the gut microbiota into 3-phenylpropionate and phenylacetate. 3-phenylpropionate, reduced in AML patients, is produced through a reductive pathway, while phenylacetate, which is increased in AML patients, arises through an oxidative pathway. 28 Along these lines, fecal phenylacetate was associated with a higher pro-oxidant and pro-inflammatory status in elderly volunteers29 whereas blood phenylacetate was elevated in cachectic cancer patients.30 Second, glycerol levels were reduced in AML feces. This decrease could result from oxidative stress on bacterial membranes, requiring increased lipid synthesis. This is supported by the increase in lipid synthesis functions (glycerol dehydratase and lipoate protein ligase), as well as by the correlations between glycerol and redox and lipid synthesis functions. Notably, the reduction in the relative abundance of obligate anaerobic genera may reflect increased oxygen levels in the intestine, contributing to the altered bacterial redox status. Several elements from blood and urinary metabolomics indicate the presence of oxidative stress on the host side, which is consistent with the findings of previous studies.31 First, taurine, a non-pro-teinogenic amino acid with antioxidant properties, was more excreted in the urine of patients with AML. Second, ascor-bate, another antioxidant, was decreased in the blood of AML patients compared with its levels in control subjects. Interestingly, a few preclinical studies have demonstrated that the gut microbiota affects the host antioxidant defense system.32 Considering all these elements, we propose that the altered activity and composition of the gut microbiota observed in AML patients could contribute to the oxidative stress present in these patients. This hypothesis is in line with the results of a previous study arguing for a microbial role in the oxidative stress present in some patients with acute leukemia under chemotherapy. Specifically, the authors proposed oxidative stress as a mediator involved in Akkermansia-related neutropenic fever.33
In addition to their link with the gut microbiota redox pathways, some metabolites pointed out previously could be linked to altered glycemic homeostasis in AML patients. Indeed, an increase in systemic phenylacetate was causally associated with insulin resistance34 and 2-hydroxybutyrate, a marker of insulin resistance,35 was increased in the blood of our cohort of AML patients. Hippurate, a glycine conjugate of microbial benzoate, was decreased in the urine of AML patients. This metabolite is commonly seen as a general marker of metabolic health and is causally associated with glucose tolerance.36 Hippurate and Prevotella increased following the administration of fibre.37 Their positive correlation in the current study suggests that reduced hippurate levels may be explained by a decrease in Prevotella. Blautia has been inconsistently associated with different aspects of metabolic syndrome.38 This genus is increased in anorectic patients.39 Consistent with this finding, in our study, Blautia increased in AML patients and correlated negatively with appetite and positively with the pro-anorexigenic cytokine GDF15. Collectively, our data reinforce previous observations of the contribution of the gut microbiota to metabolic derangements in preclinical models of acute leukemia40 and suggest that the gut microbiota could influence glucose metabolism and energy intake in AML patients, strengthening the relevance of gut microbiota alterations in this pathological context.
Among all the alterations and correlations with the most altered bacteria, E. eligens stands out. This species, reduced three-fold in leukemic patients compared to controls, was strongly correlated with muscle strength in the entire cohort and when considering exclusively AML patients. Based on urinary metabolomics, we found that E. eligens was widely associated with the urinary excretion of amino acids and amino acid metabolites. Higher urinary excretion of carni-tine and 2-aminobutyrate was previously associated with cachexia and muscle loss.41 Blood metabolomics revealed a strong association between E. eligens and urea. Such an asso ciation with urea, a metabolite involved in the regulation of ammonium levels, may be related to the urinary excretion of amino acids, representing a loss of ammonium. The increase in 8-oxoguanine deaminase and caffeine dehydro-genase, which are two functions strongly correlated with urea, could also indicate greater ammonium use by the gut microbiota. In elderly individuals, E. eligens was positively associated with markers of lower frailty,42 reinforcing the relevance of the association of this bacterial species with muscle strength. Considering the growing awareness of the importance of muscle wasting in AML outcomes,19 gut microbiota modulation could be an innovative strategy to help patients fight muscle weakness.
E. eligens was also correlated with blood levels of citrulline. Reduction in citrulline levels following intensive chemotherapy was previously reported in AML mice and patients43 and low plasma citrulline levels before allogeneic hematopoietic stem cell transplantation were associated with a higher risk of gastrointestinal GvHD and non-relapse mortality in pa-tients.44 A decrease in citrulline combined with an increase in systemic lipopolysaccharide-binding protein levels suggests an alteration of the gut function in AML patients at diagnosis. Gut permeability was not assessed at the functional level in this study as the procedure for ingestible probes is incompatible with the clinical management of patients at diagnosis. An alteration in intestinal functions, such as absorption and gut barrier, could potentially contribute to the inflammation, metabolic disorders and oxidative stress (this last through reduced ascorbate absorption) found in these patients.
Parabacteroides, especially P. merdae and P. distasonis, were increased in patients with leukemia. These two genera were reported to be affected in many pathologies38 and were ascribed both beneficial and harmful effects. An increase in Parabacteroides was also found in leukemic mice and was attributed to a reduction in food intake.17 Consistently, in our dataset, P. merdae correlated negatively with appetite. In mice with acute leukemia, we found a bloom in members of the Enterobacteriaceae family.17 In our AML cohort, Enterobacteriaceae levels did not change (Online Supplementary Figure S21). Prevotella was significantly less abundant in leukemic patients, which was mainly explained by a sixfold reduction in P. copri, a genus also reduced in cachectic patients.45 Actinomyces and P. micra were increased in our cohort of AML patients. These two oral bacteria were previously found to be increased in colorectal cancer pa-tients.38,46 In line with these studies, the relative abundance of oral species increased more than two-fold. Translocation of oral bacteria to the gut has been associated with many severe diseases, such as inflammatory bowel disease and liver cirrhosis and can promote intestinal inflammation.47 Reciprocally, we hypothesize that modifications of the microbiota composition observed upon AML are triggered by inflammation, a key driver of microbial dysbiosis.48,49 This hypothesis is coherent with the positive correlations found between inflammatory cytokines and bacteria increased in AML. Based on preclinical studies,50 we further speculate that this dysbiosis contributes to disease progression, hence the notion that AML and dysbiosis may be co-evolving.
Our clinical data are consistent with our previous preclinical observations.16,17 In both settings, we observed metabolic and inflammatory alterations, impaired gut function, and altered gut microbiota composition and function. We previously showed that counteracting microbial alterations using rationally selected prebiotics and/or probiotics, including fibers, improved gut function, and counteracted muscle atrophy in leukemic mice.16,17 These observations further support the therapeutic potential of gut microbiota-based adjuvant treatments.
This study has several limitations. The sample size was limited, and the data from this exploratory pilot study will need to be confirmed in a larger cohort. As the primary objective of this study was to evaluate gut microbiota composition and function, we enforced several exclusion criteria for metabolic and pathological situations already known to be associated with changes in gut microbiota. The exclusion of patients treated with antibiotics within 30 days and obese patients led us to exclude a considerable proportion of the patients referred to the hospital centers involved in the study. Impedancemetry is less precise than computed tomography scanning for evaluating body composition, but computed tomography scanning was not compatible with the clinical management of patients at diagnosis. In addition, the analysis of skeletal muscle function would have benefited from tests of whole-body strength and range of movement.
In conclusion, our work reveals important alterations in the gut microbiota composition and function of AML patients at diagnosis before any therapeutic intervention. This finding may be of clinical importance considering that autologous fecal material transfer is emerging as an option for these patients.15 Our findings call for caution when using autologous fecal material transfer during the therapeutic care of AML patients and go in favor of heterologous transfer to increase the gut microbiota diversity and richness in these patients. Whether transfer of fecal material would also be of benefit in cases of cachexia will need to be determined. Gut microbiota alterations are associated with hallmarks of cachexia (muscle weakness, anorexia, and inflammation), redox status, and signs of gut dysfunction. However, association does not imply causality. Therefore, our findings will constitute the basis of future mechanistic studies exploring the contribution and therapeutic potential of the bacteria identified in this study, such as Eubacterium eligens.
Footnotes
- Received August 25, 2023
- Accepted March 19, 2024
Correspondence
Disclosures
HS reports having received personal fees from Incyte, Janssen, Novartis, Sanofi and the Belgian Hematological Society (BHS), as well as research grants from Novartis and the BHS, all paid to her institution. She has also received non-financial support (travel grants) from Gilead, Pfizer, the European Society for Blood and Marrow Transplantation and the Center for International Bone Marrow Transplantation Research. None of the potential conflicts of interest are relevant to this project. All the other authors declare that they have no conflicts of interest to disclose.
Funding
This work was funded by an ESPEN Research fellowship awarded to LBB, the Fonds de la Recherche Scientifique (FRS)-FNRS under Grant MIS F.4512.20, the Fonds Wetenschappelijk Onderzoek (FWO) – Vlaanderen, and the FRS-FNRS under EOS Project N. 40007505. This work would not have been possible without the support of the Télévie (Intercachec-tomics Consortium) and the Louvain Foundation. LBB is a Collen-Francqui Research Professor (Francqui Foundation) and the recipient of subsidies from the Fonds Spéciaux de la Recherche (FSR; UCLouvain, including the Action de Recherche Concertée LIPOCAN [19-24.096]) and from the Walloon Region in the context of funding of the strategic axis FRFS-WELBIO (40009849). NMD is a recipient of grants from the FRS-FNRS (PINT-MULTI R.8013.19 [NEURON-ERANET, call 2019] and PDR T.0068.19). The funders had no role in the study design, data collection and analysis, interpretation of results, decision to publish, or preparation of the manuscript.
Acknowledgments
The authors are grateful to all the study participants. The authors are also grateful to Myriam Cleeren, Jill Pannecoucke, Vera Ciku, Dr. Martina Sboarina, Dr. Morgane Thibaut, Isabelle Blave and Dr. Julie Vanacker, as well as the data nurses and the medical team involved in the study, for their help in the collection of biological samples and clinical data. SAP and LBB thank Martin Nicolas and Bouazza Es Saadi for providing technical assistance during sample analyses and Pascaline Incourt for her help with the analyses of the food questionnaires. SAP and LBB also thank Dr. Nicolas Joudiou and the UCLouvain-LDRI Nuclear and Electron Spin Technologies (NEST) platform for providing easy access and skilled assistance for the nuclear magnetic resonance spectrometry and Prof. Ana Beloqui for access to a Meso Scale Discovery microplate reader. The authors thank Dr. Inge Huybrechts and Dr. Ernst Rietzschel for sharing the food frequency questionnaires. The authors also acknowledge the Centre d’Expertise et de Services Génome Québec for their sequencing services and support (shotgun metagenomics), and the University of Minnesota Genomics Center (for 16S rRNA gene sequencing). This research has benefitted from a statistical consult with Statistical Methodology and Computing Service, a technological platform at UCLouvain – SMCS/LIDAM, UCLouvain.
References
- Postler TS, Ghosh S. Understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab. 2017; 26(1):110-130. https://doi.org/10.1016/j.cmet.2017.05.008PubMedPubMed CentralGoogle Scholar
- Visconti A, Le Roy CI, Rosa F. Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019; 10(1):4505. https://doi.org/10.1038/s41467-019-12476-zPubMedPubMed CentralGoogle Scholar
- Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020; 20(1):40-54. https://doi.org/10.1038/s41577-019-0198-4PubMedGoogle Scholar
- Delzenne NM, Knudsen C, Beaumont M, Rodriguez J, Neyrinck AM, Bindels LB. Contribution of the gut microbiota to the regulation of host metabolism and energy balance: a focus on the gut-liver axis. Proc Nutr Soc. 2019; 78(3):319-328. https://doi.org/10.1017/S0029665118002756PubMedGoogle Scholar
- Manzo VE, Bhatt AS. The human microbiome in hematopoiesis and hematologic disorders. Blood. 2015; 126(3):311-318. https://doi.org/10.1182/blood-2015-04-574392PubMedPubMed CentralGoogle Scholar
- Rashidi A, Kaiser T, Graiziger C. Gut dysbiosis during antileukemia chemotherapy versus allogeneic hematopoietic cell transplantation. Cancer. 2020; 126(7):1434-1447. https://doi.org/10.1002/cncr.32641PubMedGoogle Scholar
- D’Angelo CR, Sudakaran S, Callander NS. Clinical effects and applications of the gut microbiome in hematologic malignancies. Cancer. 2021; 127(5):679-687. https://doi.org/10.1002/cncr.33400PubMedGoogle Scholar
- Galloway-Pena JR, Smith DP, Sahasrabhojane P. The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia. Cancer. 2016; 122(14):2186-2196. https://doi.org/10.1002/cncr.30039PubMedPubMed CentralGoogle Scholar
- Galloway-Pena JR, Shi Y, Peterson CB. Gut microbiome signatures are predictive of infectious risk following induction therapy for acute myeloid leukemia. Clin Infect Dis. 2020; 71(1):63-71. https://doi.org/10.1093/cid/ciz777PubMedPubMed CentralGoogle Scholar
- Taur Y, Jenq RR, Perales MA. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014; 124(7):1174-1182. https://doi.org/10.1182/blood-2014-02-554725PubMedPubMed CentralGoogle Scholar
- Jenq RR, Taur Y, Devlin SM. Intestinal Blautia is associated with reduced death from graft-versus-host disease. Biol Blood Marrow Transplant. 2015; 21(8):1373-1383. https://doi.org/10.1016/j.bbmt.2015.04.016PubMedPubMed CentralGoogle Scholar
- Peled JU, Devlin SM, Staffas A. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J Clin Oncol. 2017; 35(15):1650-1659. https://doi.org/10.1200/JCO.2016.70.3348PubMedPubMed CentralGoogle Scholar
- Bilinski J, Robak K, Peric Z. Impact of gut colonization by antibiotic-resistant bacteria on the outcomes of allogeneic hematopoietic stem cell transplantation: a retrospective, single-center study. Biol Blood Marrow Transplant. 2016; 22(6):1087-1093. https://doi.org/10.1016/j.bbmt.2016.02.009PubMedGoogle Scholar
- Harris B, Morjaria SM, Littmann ER. Gut microbiota predict pulmonary infiltrates after allogeneic hematopoietic cell transplantation. Am J Respir Crit Care Med. 2016; 194(4):450-463. https://doi.org/10.1164/rccm.201507-1491OCPubMedPubMed CentralGoogle Scholar
- Malard F, Vekhoff A, Lapusan S. Gut microbiota diversity after autologous fecal microbiota transfer in acute myeloid leukemia patients. Nat Commun. 2021; 12(1):3084. https://doi.org/10.1038/s41467-021-23376-6PubMedPubMed CentralGoogle Scholar
- Bindels LB, Beck R, Schakman O. Restoring specific lactobacilli levels decreases inflammation and muscle atrophy markers in an acute leukemia mouse model. PLoS One. 2012; 7(6):e37971. https://doi.org/10.1371/journal.pone.0037971PubMedPubMed CentralGoogle Scholar
- Bindels LB, Neyrinck AM, Claus SP. Synbiotic approach restores intestinal homeostasis and prolongs survival in leukaemic mice with cachexia. ISME J. 2016; 10(6):1456-1470. https://doi.org/10.1038/ismej.2015.209PubMedPubMed CentralGoogle Scholar
- Argiles JM, Lopez-Soriano FJ, Stemmler B, Busquets S. Cancer-associated cachexia - understanding the tumour macroenvironment and microenvironment to improve management. Nat Rev Clin Oncol. 2023; 20(4):250-264. https://doi.org/10.1038/s41571-023-00734-5PubMedGoogle Scholar
- Campelj DG, Timpani CA, Rybalka E. Cachectic muscle wasting in acute myeloid leukaemia: a sleeping giant with dire clinical consequences. J Cachexia Sarcopenia Muscle. 2022; 13(1):42-54. https://doi.org/10.1002/jcsm.12880PubMedPubMed CentralGoogle Scholar
- Hoebeeck LI, Rietzschel ER, Langlois M. The relationship between diet and subclinical atherosclerosis: results from the Asklepios study. Eur J Clin Nutr. 2011; 65(5):606-613. https://doi.org/10.1038/ejcn.2010.286PubMedGoogle Scholar
- Falony G, Joossens M, Vieira-Silva S. Population-level analysis of gut microbiome variation. Science. 2016; 352(6285):560-564. https://doi.org/10.1126/science.aad3503PubMedGoogle Scholar
- Heini AD, Hugo R, Berger MD, Novak U, Bacher U, Pabst T. Simple acute phase protein score to predict long-term survival in patients with acute myeloid leukemia. Hematol Oncol. 2020; 38(1):74-81. https://doi.org/10.1002/hon.2696PubMedGoogle Scholar
- Chen WL, Wang JH, Zhao AH. A distinct glucose metabolism signature of acute myeloid leukemia with prognostic value. Blood. 2014; 124(10):1645-1654. https://doi.org/10.1182/blood-2014-02-554204PubMedPubMed CentralGoogle Scholar
- Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): a focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev. 2018; 43:8-15. https://doi.org/10.1016/j.cytogfr.2018.08.004PubMedGoogle Scholar
- Johnen H, Lin S, Kuffner T. Tumor-induced anorexia and weight loss are mediated by the TGF-β superfamily cytokine MIC-1. Nat Med. 2007; 13(11):1333-1340. https://doi.org/10.1038/nm1677PubMedGoogle Scholar
- Oost LJ, Kustermann M, Armani A, Blaauw B, Romanello V. Fibroblast growth factor 21 controls mitophagy and muscle mass. J Cachexia Sarcopenia Muscle. 2019; 10(3):630-642. https://doi.org/10.1002/jcsm.12409PubMedPubMed CentralGoogle Scholar
- Fragkos KC, Forbes A. Citrulline as a marker of intestinal function and absorption in clinical settings: a systematic review and meta-analysis. United European Gastroenterol J. 2018; 6(2):181-191. https://doi.org/10.1177/2050640617737632PubMedPubMed CentralGoogle Scholar
- Dodd D, Spitzer MH, Van Treuren W. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017; 551(7682):648-652. https://doi.org/10.1038/nature24661PubMedPubMed CentralGoogle Scholar
- Gutierrez-Diaz I, Fernandez-Navarro T, Salazar N. Could fecal phenylacetic and phenylpropionic acids be used as indicators of health status?. J Agric Food Chem. 2018; 66(40):10438-10446. https://doi.org/10.1021/acs.jafc.8b04102PubMedGoogle Scholar
- Yang QJ, Zhao JR, Hao J. Serum and urine metabolomics study reveals a distinct diagnostic model for cancer cachexia. J Cachexia Sarcopenia Muscle. 2018; 9(1):71-85. https://doi.org/10.1002/jcsm.12246PubMedPubMed CentralGoogle Scholar
- Trombetti S, Cesaro E, Catapano R. Oxidative stress and ROS-mediated signaling in leukemia: novel promising perspectives to eradicate chemoresistant cells in myeloid leukemia. Int J Mol Sci. 2021; 22(5):2470. https://doi.org/10.3390/ijms22052470PubMedPubMed CentralGoogle Scholar
- Uchiyama J, Akiyama M, Hase K, Kumagai Y, Kim YG. Gut microbiota reinforce host antioxidant capacity via the generation of reactive sulfur species. Cell Rep. 2022; 38(10):110479. https://doi.org/10.1016/j.celrep.2022.110479PubMedGoogle Scholar
- Rashidi A, Ebadi M, Rehman TU. Altered microbiota-host metabolic cross talk preceding neutropenic fever in patients with acute leukemia. Blood Adv. 2021; 5(20):3937-3950. https://doi.org/10.1182/bloodadvances.2021004973PubMedPubMed CentralGoogle Scholar
- Hoyles L, Fernandez-Real JM, Federici M. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat Med. 2018; 24(7):1070-1080. https://doi.org/10.1038/s41591-018-0061-3PubMedPubMed CentralGoogle Scholar
- Vangipurapu J, Fernandes Silva L, Kuulasmaa T, Smith U, Laakso M. Microbiota-related metabolites and the risk of type 2 diabetes. Diabetes Care. 2020; 43(6):1319-1325. https://doi.org/10.2337/dc19-2533PubMedGoogle Scholar
- Brial F, Chilloux J, Nielsen T. Human and preclinical studies of the host-gut microbiome co-metabolite hippurate as a marker and mediator of metabolic health. Gut. 2021; 70(11):2105-2114. https://doi.org/10.1136/gutjnl-2020-323314PubMedPubMed CentralGoogle Scholar
- Dewulf EM, Cani PD, Claus SP. Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut. 2013; 62(8):1112-1121. https://doi.org/10.1136/gutjnl-2012-303304PubMedPubMed CentralGoogle Scholar
- Cheng L, Hu Y, Sun J, Zhou M, Jiang Q. DincRNA: a comprehensive web-based bioinformatics toolkit for exploring disease associations and ncRNA function. Bioinformatics. 2018; 34(11):1953-1956. https://doi.org/10.1093/bioinformatics/bty002PubMedGoogle Scholar
- Kleiman SC, Watson HJ, Bulik-Sullivan EC. The intestinal microbiota in acute anorexia nervosa and during renourishment: relationship to depression, anxiety, and eating disorder psychopathology. Psychosom Med. 2015; 77(9):969-981. https://doi.org/10.1097/PSY.0000000000000247PubMedPubMed CentralGoogle Scholar
- Ye H, Adane B, Khan N. Subversion of systemic glucose metabolism as a mechanism to support the growth of leukemia cells. Cancer Cell. 2018; 34(4):659-673. https://doi.org/10.1016/j.ccell.2018.08.016PubMedPubMed CentralGoogle Scholar
- Szefel J, Kruszewski WJ, Ciesielski M. L-carnitine and cancer cachexia. I. L-carnitine distribution and metabolic disorders in cancer cachexia. Oncol Rep. 2012; 28(1):319-323. https://doi.org/10.3892/or.2012.1804PubMedGoogle Scholar
- Ghosh TS, Rampelli S, Jeffery IB. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: the NU-AGE 1-year dietary intervention across five European countries. Gut. 2020; 69(7):1218-1228. https://doi.org/10.1136/gutjnl-2019-319654PubMedPubMed CentralGoogle Scholar
- Hueso T, Ekpe K, Mayeur C. Impact and consequences of intensive chemotherapy on intestinal barrier and microbiota in acute myeloid leukemia: the role of mucosal strengthening. Gut Microbes. 2020; 12(1):1800897. https://doi.org/10.1080/19490976.2020.1800897PubMedPubMed CentralGoogle Scholar
- Hueso T, Gauthier J, Joncquel Chevalier-Curt M. Association between low plasma level of citrulline before allogeneic hematopoietic cell transplantation and severe gastrointestinal graft vs host disease. Clin Gastroenterol Hepatol. 2018; 16(6):908-917. https://doi.org/10.1016/j.cgh.2017.12.024PubMedGoogle Scholar
- Ni Y, Lohinai Z, Heshiki Y. Distinct composition and metabolic functions of human gut microbiota are associated with cachexia in lung cancer patients. ISME J. 2021; 15(11):3207-3220. https://doi.org/10.1038/s41396-021-00998-8PubMedPubMed CentralGoogle Scholar
- Dai Z, Coker OO, Nakatsu G. Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome. 2018; 6(1):70. https://doi.org/10.1186/s40168-018-0451-2PubMedPubMed CentralGoogle Scholar
- Atarashi K, Suda W, Luo C. Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science. 2017; 358(6361):359-365. https://doi.org/10.1126/science.aan4526PubMedPubMed CentralGoogle Scholar
- Zeng MY, Inohara N, Nunez G. Mechanisms of inflammationdriven bacterial dysbiosis in the gut. Mucosal Immunol. 2017; 10(1):18-26. https://doi.org/10.1038/mi.2016.75PubMedPubMed CentralGoogle Scholar
- Bindels LB, Neyrinck AM, Loumaye A. Increased gut permeability in cancer cachexia: mechanisms and clinical relevance. Oncotarget. 2018; 9(26):18224-18238. https://doi.org/10.18632/oncotarget.24804PubMedPubMed CentralGoogle Scholar
- Wang R, Yang X, Liu J. Gut microbiota regulates acute myeloid leukaemia via alteration of intestinal barrier function mediated by butyrate. Nat Commun. 2022; 13(1):2522. https://doi.org/10.1038/s41467-022-30240-8PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.