Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2021 Jul 15;89(8):e00105-21. doi: 10.1128/IAI.00105-21

Pharyngeal Microbial Signatures Are Predictive of the Risk of Fungal Pneumonia in Hematologic Patients

Claudio Costantini a,#, Emilia Nunzi a,b,#, Angelica Spolzino c, Melissa Palmieri a, Giorgia Renga a, Teresa Zelante a, Lukas Englmaier d, Katerina Coufalikova d, Zdeněk Spáčil d, Monica Borghi a, Marina M Bellet a, Enzo Acerbi e, Matteo Puccetti f, Stefano Giovagnoli f, Roberta Spaccapelo a,b, Vincenzo N Talesa a,b, Giuseppe Lomurno a, Francesco Merli g, Luca Facchini g, Antonio Spadea h, Lorella Melillo i, Katia Codeluppi g, Francesco Marchesi h, Gessica Marchesini j, Daniela Valente i, Giulia Dragonetti k, Gianpaolo Nadali j, Livio Pagano k,l, Franco Aversa c, Luigina Romani a,b,
Editor: Mairi C Noverrm
PMCID: PMC8281212  PMID: 33782152

ABSTRACT

The ability to predict invasive fungal infections (IFI) in patients with hematological malignancies is fundamental for successful therapy. Although gut dysbiosis is known to occur in hematological patients, whether airway dysbiosis also contributes to the risk of IFI has not been investigated. Nasal and oropharyngeal swabs were collected for functional microbiota characterization in 173 patients with hematological malignancies recruited in a multicenter, prospective, observational study and stratified according to the risk of developing IFI. A lower microbial richness and evenness were found in the pharyngeal microbiota of high-risk patients that were associated with a distinct taxonomic and metabolic profile. A murine model of IFI provided biologic plausibility for the finding that loss of protective anaerobes, such as Clostridiales and Bacteroidetes, along with an apparent restricted availability of tryptophan, is causally linked to the risk of IFI in hematologic patients and indicates avenues for antimicrobial stewardship and metabolic reequilibrium in IFI.

KEYWORDS: hematological malignancies, airway microbiome, antibiotics, indole-3-aldehyde, invasive fungal infection, metabolomics, tryptophan

INTRODUCTION

Invasive fungal infections (IFI) still remain a major cause of nonrelapse mortality and represent one of the significant causes of expense in the management of patients with hematological malignancies (1, 2). The possibility to determine the actual risk for IFI would be of fundamental importance to drive the clinicians toward the best therapeutic option for the patients and individualize the therapy to balance between prevention of IFI and occurrence of side effects (3). Although the introduction of molecular and serological diagnostic techniques into clinical practice has significantly improved fungal diagnostics, sensitivity and specificity are still a problem.

Currently, the host-fungus interaction is being exploited to project more efficient and reliable fungal diagnostics (4), and efforts are being devoted to the implementation of clinical models predicting the infection in high-risk patients (5). Several studies have identified associations between the gut microbiome and infection risk in allogeneic hematopoietic stem cell transplant (HSCT) and graft-versus-host disease (GvHD) (6). Loss of intestinal bacterial diversity and outgrowth of opportunistic pathogens belonging to the phylum Proteobacteria and the Enterococcus genus are common during the course of HSCT and are associated with GvHD development and treatment with broad-spectrum antibiotics (6, 7). The “gut-lung axis” is commonly invoked to explain the microbiome's influence on lung immunity and inflammation, yet the lungs harbor their own microbiome and their own mucosa. As the influence of remote (gut-lung) microbe-host interactions on lung inflammation is relatively unknown (8), the assessment of lung microbial composition may better explain the role of microbial dysbiosis in lung health and disease. One recent study has shown that following HSCT, a significantly increased relative abundance of Proteobacteria in bronchoalveolar lavage (BAL) specimens is associated with histologic, immunologic, and physiologic features of pulmonary complications (9), a finding consistent with the association of Proteobacteria domination with lung dysfunction and disease (10). However, there are limited data regarding the actual predictive capacity of lung microbiome-related dysbiosis and biomarkers. In addition, although the use of BAL specimens provides valuable information to assess the microbiota of the lower respiratory tract, it represents an invasive procedure (11). Thus, considering that spatial microbial diversity in the lungs of healthy individuals seems almost absent (12) and that the lung microbiota is a community of transiently present microorganisms derived from the upper respiratory tract, the exploitation of the upper airway microbiota as a representation of the lung microbiota (11, 13) could be an attractive option that overcomes the inherent limitations associated with the sputum (11) or the bias related to the sampling method.

Based on these premises, we have designed a multicenter, prospective, observational study, termed SNIF (survey of nasal infection), in which patients with hematologic malignancies were recruited and their nasal and pharyngeal swabs collected over a 6-month period for microbiome characterization. The study was designed to identify microbial signatures that could predict the risk of IFI as a primary objective. Here, we show that the specific risk for IFI is associated with configurations of the pharyngeal microbiome along characteristic patterns.

RESULTS

The oropharyngeal microbiota of hematologic patients.

In a first analysis of the SNIF database, we focused on the oropharyngeal microbiome, owing to its higher richness, that may increase the likelihood of uncovering associations with the risk of IFI for being the main source of the lung microbiota in adult tracts (14) and for the potential protective role in pulmonary infections (15). The taxonomic composition of the oropharyngeal microbiome is shown in Fig. 1. Consistent with previous reports (16, 17), Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, and Fusobacteria represented the major phyla, accounting for nearly the entire spectrum of bacteria detected in the oropharynx, followed by low-abundance phyla with a cumulative abundance of less than 1% (Fig. 1A). At the genus level, Streptococcus was found to be the most abundant, followed by Veillonella, Prevotella, Neisseria, Actinomyces, Haemophilus, Fusobacterium, and Rothia, common genera of the pharynx (14, 16, 17) (Fig. 1B).

FIG 1.

FIG 1

Oropharyngeal microbiome of hematologic patients is dominated by bacteria commonly associated with the oropharynx. (A) Bar plot showing bacterial composition (abundance percentage) of each sample at the phylum level. Taxa are differentiated by colors. Samples are ranked based on the abundance of Firmicutes. (B) Behavior of the cumulative proportions of taxa at genus level in the complete cohort versus number of observed taxa. The color of each taxon represents the prevalence of the taxon within the community. Taxa are ordered by increasing percentage value, and the first 10 most abundant genera are explicitly indicated by labels. These 10 taxa present a prevalence >50%.

Hematologic patients at different risks for IFI harbor distinct oropharyngeal microbiomes.

To define the potential predictive value of the oropharyngeal microbiome, each sample was associated with the overall risk of IFI based on the following criteria, i.e., the duration of neutropenia, antibiotic usage, and the dynamic risk proposed by SEIFEM (epidemiology survey of invasive fungal infections in hematological malignancies) (3). The details for risk stratification in each hematological malignancy, based on diagnosis, phase, and type of treatment, as well as the algorithm at the basis of the dynamic risk definition, are described in reference 3. Upon application of these criteria, samples were assigned to high risk (HR) for IFI if collected during a condition of prolonged neutropenia or administration of broad-spectrum antibiotics or if satisfying the conditions for dynamic high risk (dHR) according to the SEIFEM algorithm. Conversely, samples were assigned to low risk (LR) for IFI if collected outside periods of prolonged neutropenia or broad-spectrum antibiotics administration or if satisfying the conditions for dynamic low risk (dLR) according to the SEIFEM algorithm. The distribution for age, gender, and hematological disease for the HR and LR populations considered separately are shown in Fig. S1 in the supplemental material. The overlap between the different HR and LR populations is shown in Fig. S2. Specifically, 66.4% and 94% of the LR samples are identified by all three criteria or at least two of them, respectively, indicating a substantial degree of overlapping in the classification of patients as low risk for IFI (Fig. S2A). Conversely, only 14.3% and 29.6% of HR samples are identified by all three criteria or at least two of them, respectively (Fig. S2B to E). The difference is driven by a group of samples that are recognized as HR by the dynamic risk proposed by SEIFEM but not by prolonged neutropenia and administration of broad-spectrum antibiotics. This is in line with the dynamic risk factors that go beyond the occurrence of neutropenia and the use of broad-spectrum antibiotics (3). The analysis of alpha and beta diversities revealed that LR and HR samples were associated with distinct microbiota for all the criteria. Indeed, a higher richness and evenness, as measured by observed operational taxonomic units and Chao1 and Shannon indexes, were observed in the LR groups, i.e., no prolonged neutropenia or broad-spectrum antibiotics or assigned to dLR, than the HR groups (Fig. 2A). This result was expected, as the HR group is associated with a long period of neutropenia and, therefore, more intense pharmacological treatment that severely affects the microbial composition and diversity. The oropharyngeal microbiota of the two groups also showed significant differences in compositional structure, as measured by Jaccard and Bray-Curtis indexes (Fig. 2B). Indeed, principal coordinate analysis derived from Jaccard and Bray-Curtis distances revealed significant differences between the LR and HR groups (Fig. S3).

FIG 2.

FIG 2

Microbiome composition differs between LR and HR oropharyngeal samples. (A) Boxplots of observed OTUs, Chao1, and Shannon alpha diversity indexes grouped by risk of IFI according to three distinct criteria (presence or not of prolonged neutropenia, dynamic LR, or HR according to SEIFEM algorithm, and use or not of broad-spectrum antibiotics). Significance was evaluated by applying a Kruskal-Wallis test (the P value is indicated). (B) Boxplots of Jaccard and Bray-Curtis beta diversity indexes evaluating distances within (LR, green; HR, red) or between (gray) LR and HR samples according to the three distinct criteria described for panel A. Significance was evaluated by applying a Kruskal-Wallis test (the P value is indicated).

Upon applying the Adonis test to Jaccard and Bray-Curtis diversities when a linear simple multifactorial model of the dynamic risk, prolonged neutropenia, broad-spectrum antibiotics, and occurrence of mucositis was used, all the variables, except mucositis, had significantly different profiles for both metrics. Of note, the dynamic risk accounted for the largest percentage of variation (Table 1).

TABLE 1.

Multivariate analysis

Parameter Df SumsOfSqs MeanSqs F.Model R2 Pr(>F)
Jaccard
    Dynamic risk 1 2.361346588 2.361346588 7.203580256 0.024817729 0.001
    Prolonged neutropenia 1 0.721783724 0.721783724 2.201890654 0.00758594 0.001
    Broad-spectrum antibiotics 1 0.594637061 0.594637061 1.81401401 0.00624963 0.001
    Mucositis 1 0.340901612 0.340901612 1.039962593 0.003582873 0.33
    Residuals 278 91.12890092 0.327801802 NA 0.95776383 NA
    Total 282 95.1475699 NAa NA 1 NA
Bray-Curtis
    Dynamic risk 1 1.579643048 1.579643048 5.350979996 0.018505499 0.001
    Prolonged neutropenia 1 0.839875287 0.839875287 2.845045193 0.009839129 0.001
    Broad-spectrum antibiotics 1 0.608823486 0.608823486 2.062366115 0.00713236 0.003
    Mucositis 1 0.265042627 0.265042627 0.897821693 0.003104971 0.614
    Residuals 278 82.06735358 0.295206308 NA 0.961418042 NA
    Total 282 85.36073802 NA NA 1 NA
a

NA, not applicable. Df: degrees of freedom; SumsOfSqs: Sums of Squares; MeanSqs: mean squares; F.Model: F-statistics; R2: R-squared; Pr(>F): P value from F-distribution.

Collectively, these results indicate that the different criteria provide a consistent framework to identify potential signatures of the oropharyngeal microbiome that could predict the risk of IFI.

The risk of IFI is associated with distinct microbial genera.

To identify these signatures, we performed high-dimensional class comparisons using linear discriminant analysis of effect size (LEfSe) (18), by which we could detect significant differences in the bacterial communities between LR and HR groups. Despite differences at the level of some genera, a common signature could be identified that was associated with the risk of infection among the differently stratified groups. Specifically, we observed the relative predominance of members of Bacteroidetes (i.e., the Prevotella genus), Firmicutes (i.e., Clostridia with the Lachnospiranaceae family), and Actinobacteria phyla, in addition to a higher abundance of most oral taxa (Veillonella, Neisseria, Leptotrichia, and Gemella) in the LR groups. Conversely, the relative predominance of members of Firmicutes (i.e., the Staphylococcus and Enterococcus genera) variably associated with Lactobacillaceae and Gram-negative Proteobacteria (i.e., Acinetobacter and Stenotrophomonas genera) was observed in the different HR groups (Fig. 3 to 5 and Fig. S4 and S5). Similar results were obtained when samples were assigned to HR or LR for IFI if collected or not during the occurrence of mucositis (Fig. S6). The relative abundances of these genera are shown in Fig. S4.

FIG 3.

FIG 3

Oropharyngeal microbiomes have a different genus composition in function of prolonged neutropenia. Taxonomic visualization of statistically and biologically consistent differences between samples collected during the presence or absence of prolonged neutropenia. The cladogram simultaneously highlights high-level taxa and specific genera. Taxa (circles) are colored red when significantly (LEfSe, P < 0.05) associated with the presence of prolonged neutropenia, green when significantly associated with the absence of prolonged neutropenia, and yellow when not significantly associated with either group. The size of each circle is proportional to the abundance of the corresponding taxon in all samples. The histograms of the linear discriminant analysis (LDA) scores are computed for genera significantly associated with either the presence (red) or the absence (green) of prolonged neutropenia. LEfSe has been applied with default alpha values for the analysis of variance (ANOVA) and Wilcoxon tests (0.05), and the LDA effect size has been evaluated by setting the absolute value of the logarithmic LDA threshold equal to 3.5. Other LEfSe parameters have been set to the default.

FIG 4.

FIG 4

Oropharyngeal microbiomes have a different genera composition in function of dynamic risk stratification. Taxonomic visualization of statistically and biologically consistent differences between samples allocated to low risk (dLR) or high risk (dHR) for IFI by dynamic risk stratification. The cladogram simultaneously highlights high-level taxa and specific genera. Taxa (circles) are colored red when significantly (LEfSe, P < 0.05) associated with dHR, green when significantly associated with dLR, and yellow when not significantly associated with either group. The size of each circle is proportional to the abundance of the corresponding taxon in all samples. The histograms of the LDA scores are computed for genera significantly associated with either dHR (red) or dLR (green) groups. LEfSe has been applied with default alpha values for the ANOVA and Wilcoxon test (0.05) and the LDA effect size has been evaluated by setting the absolute value of the logarithmic LDA threshold equal to 3.5. Other LEfSe parameters have been set to the default.

FIG 5.

FIG 5

Oropharyngeal microbiomes have a different genera composition in function of broad-spectrum antibiotics use. Taxonomic visualization of statistically and biologically consistent differences between samples collected or not during treatments with broad-spectrum antibiotics. The cladogram simultaneously highlights high-level taxa and specific genera. Taxa (circles) are colored red when significantly (LEfSe, P < 0.05) associated with the use of broad-spectrum antibiotics, green when significantly associated with the absence of broad-spectrum antibiotics use, and yellow when not significantly associated with either group. The size of each circle is proportional to the abundance of the corresponding taxon in all samples. The histograms of the LDA scores are computed for genera significantly associated with either the use (red) or lack of use (green) of broad-spectrum antibiotics. LEfSe has been applied with default alpha values for the ANOVA and Wilcoxon test (0.05), and the LDA effect size has been evaluated by setting the absolute value of the logarithmic LDA threshold equal to 3.5. Other LEfSe parameters have been set to the default.

The risk of IFI is associated with distinct microbial metabolic activities.

To improve the predictive value of microbial signatures (19), we have assessed microbial functional activity in dHR and dLR groups. By using PICRUSt2 and the annotated MetaCyc database, abundances of functional pathways were inferred from 16S data, and LEfSe was applied to evaluate significant association with either HR or LR samples. In particular, genes involved in the biosynthesis of l-tryptophan (trp), glycolysis, and homolactic fermentation were more abundant in HR than LR patients, while genes involved in the fatty acid elongation and the starch degradation pathways (abundantly present in oral bacteria [20]) were more abundant in LR than HR patients (Fig. 6A and Fig. S7). The KEGG database confirmed the association of trp biosynthesis with the HR group (Fig. 6B and Fig. S8). The increased trp auxotrophy would also predict different availability of trp in the HR and LR groups. We measured these levels in a subgroup of pharyngeal samples from 14 consecutive patients undergoing HSCT during the study period and found that trp levels were indeed lower in HR than LR patients (Fig. 6C). Moreover, and importantly, while the levels of l-kynurenine (kyn), resulting from host catabolism of trp, were not different between the two groups, the production of indole-3-aldehyde (3-IAld), an indole that reflects the microbial consumption of trp, was significantly lower in HR than LR patients (Fig. 6C). These findings suggest an apparent restricted availability of trp for microbial consumption in HR patients. Consistent with this, amino acid biosynthetic pathways were abundantly present in HR samples (Fig. S7).

FIG 6.

FIG 6

LR and HR oropharyngeal samples differ in tryptophan metabolism. (A and B) Box plots of trp biosynthesis pathway (A) and module (B) inferred by PICRUSt2 analysis according to MetaCyc and KEGG databases, respectively. These predicted metagenome functions were indicated by LEfSe as significantly differentially represented in the high- and low-risk groups. (C) Tryptophan (trp), kynurenines (kyn), and indole-3-aldehyde (3-IAld) levels (nmol/liter) were measured in oropharyngeal samples (n = 63; LR, 27; HR, 36) and expressed as means ± standard deviations (SD). *, P < 0.05 LR versus HR, unpaired t test.

Collectively, these results reveal the existence of a transcriptionally active oropharyngeal microbiota that may impact lung immune status and suggest that not only microbial composition but also active functional activity characterizes the pharyngeal microbiota.

Validation experiments in mice.

We have resorted to a murine model of Aspergillus pneumonia for experimental validation and biological interpretation of the possible causal contribution of the discriminating taxa to the risk of IFI. For this purpose, C57BL/6 mice were intranasally infected with live A. fumigatus conidia and assessed for microbial composition and function in the lung. Using a customized 16S rRNA sequencing protocol amplifying the V3-V4 region, the most represented phyla in the lung of uninfected mice were Proteobacteria (55%), Firmicutes (31%), Actinobacteria (6%), and Bacteroidetes (4%) (Fig. S9A), a phylum distribution similar to that previously reported (21). Except for Bacteriodetes, which expanded, no major alterations were observed at the phylum level in infection (Fig. S9A). However, a significant shift in bacterial richness and diversity as well as community structure was observed. The Lactobacillaceae family (Lactobacillus genus), mostly represented in uninfected mice, slightly decreased, while strictly anaerobic bacteria belonging to both Firmicutes (Clostridiales order, Lachnospiranaceae and Clostridiaceae Family_XI and Blautia) and Bacteroidetes (Prevotellaceae, S24-7, and Bacteroides) phyla expanded (Fig. S9B). The expansion of strictly anaerobic Clostridiales occurred in the early phase of the infection, followed by a more sustained expansion of Bacteroidetes (Fig. S9C), consistent with the observation that Bacteroidetes are pH sensitive and fail to prosper in an acidic inflammatory environment (22). Within Proteobacteria, Enterobacteriaceae (Morganella and Escherichia-Shigella) expanded in infection (Fig. S9B). Strictly anaerobic bacteria are protected from fungal pneumonia, as indicated by the increased susceptibility to the infection upon treatment with metronidazole or the broad-spectrum antibiotics, meropenem and piperacillin-tazobactam, known to cause severe declines among obligate anaerobes (23) and less with ciprofloxacin (experimental protocol depicted in Fig. 7A). The decreased abundance of strict anaerobes (Clostridiales and Bacteroidetes) was associated with the expansion of facultative anaerobes of Enterobacteriaceae (Fig. 7B) and concomitantly increased fungal growth and pathology (Fig. 7C) in the lung. In agreement with these findings, Prevotella oris, a representative member of Bacteroidetes, and Peptostreptococcus anaerobius, a representative member of Clostridiales, inhibited conidial germination upon coculturing with the fungus (Fig. S10). This finding suggests that anaerobic bacteria serve a protective role in infection by attenuating fungal virulence and limiting the expansion of facultative anaerobes known to promote lung inflammation (24). Of interest, 3-IAld, more abundantly present in LR than HR patients, also protected from infection and inflammation upon administration to Aspergillus-infected mice, as indicated by reduced fungal burden and improved histopathology (Fig. 7D). 3-IAld treatment was associated with an increased expression of the aryl hydrocarbon receptor (AhR) and its target gene Cyp1a1 (Fig. 7E), in line with 3-IAld working as an agonist of AhR (25). 3-IAld treatment also induced the expression of Rorc and the production of IL-22 (Fig. 7E), indicating the expansion of protective type 3 innate lymphoid cells (ILC3) (25). Thus, murine Aspergillus pneumonia recapitulates the human microbiological findings while offering plausible explanations for the possible causal relationship between the microbial landscape, metabolic activity, and risk of IFI.

FIG 7.

FIG 7

Murine model of aspergillosis validate the human findings. (A) Schematic representation of the protocol with antibiotics and A. fumigatus infection. i.n., intranasal. (B and C) Microbial composition (by qPCR) (B) and fungal growth (means ± SD) and histology (periodic acid-Schiff staining) (C) of lung from C57BL/6 mice intranasally infected with viable resting A. fumigatus conidia and treated with antibiotics 2 weeks before and continuing for a week after the infection. Pip/Taz, piperacillin-tazobactam. (D and E) Fungal growth, histology (D), and transcription factor gene expression (RT-PCR) and cytokine production (ELISA) (E) in infected C57BL/6 mice treated with an oral formulation of indole-3-aldehyde (3-IAld) 4 times the week before the infection. Assays were done at the end of treatments. **, P < 0.01; ***, P < 0.001, treated versus untreated (none) mice. Naïve, uninfected mice.

DISCUSSION

Here, we have described the potential role of oropharyngeal microbiota in predicting the risk of IFI in hematologic patients. We have found that loss of alpha diversity, associated with decreased abundance of Clostridiales, Bacteroidetes, and common oral taxa, the relative expansion of Staphylococcus and Enterococcus, and a distinct metabolic profile characterize the HR patients. Interestingly, depletion of common taxa typically associated with oral health, including Actinomyces, Gemella, and Veillonella, has been reported in oral microbiota of patients undergoing 5-fluorouracil- or doxorubicin-based chemotherapy and negatively correlated with mucositis severity (26). Conversely, the role of Staphylococcus species in chronic inflammatory airway diseases (27) and infections in various hematological malignancies (28) is well documented. Interestingly, the expansion of Staphylococcus in oral and stool microbiome has been reported in a leukemic patient prior to invasive mucormycosis (29). The increased prevalence of the Enterococcus genus in HR patients is also of particular interest, as it is present not only in chronic obstructive pulmonary disease (30) but also in the pharyngeal microbiome of patients with hematological malignancies (31) and is causally associated with GvHD in acute myeloid leukemia (AML) patients (32). In addition, in a mouse intravenous chemotherapy model, upon 5-fluorouracil administration, C. albicans infection resulted in increased abundance of the Enterococcus genus in the oral mucosa, potentially responsible for the mucosal barrier impairment and fungal invasion (33).

The metabolic profiling helped to elucidate the possible pathogenic role of the HR-associated microbes to dysbiosis. While the glycolytic pathways are generally associated with inflammation, the increased biosynthesis of trp is of particular interest, as bacterial trp prototrophy has been associated with immune evasion and adaptation (34, 35). Indeed, on a metabolic level, the increased trp biosynthesis was associated with low levels of trp and indole observed in HR patients. Considering the influence of host and microbial trp metabolism on human health status (36, 37) as well as the occurrence of metabolic immune programming during pulmonary infections (38), these results point to a dysregulated metabolic activity linked to bacterial virulence and host inflammation (37). Fecal indole, as a potential surrogate marker for microbial diversity and specific taxa, has recently been described in HSCT recipients (39). As antibiotics alter the metabolic state of bacteria (40), it is likely that the more intense prophylactic regimen in HR patients is responsible for the observed metabolic profile in addition to the loss of bacterial diversity. In this regard, our murine results suggest that anaerobic bacteria serve a protective role in infection by attenuating fungal virulence and limiting the expansion of facultative anaerobes known to promote lung inflammation (24). In addition, they confirm the deleterious effects of anaerobe-targeting broad-spectrum antibiotics on the outcome of fungal pneumonia (23, 41, 42) and support the human findings. Indeed, the failure of strict anaerobes Clostridiales and Bacteroidetes to expand was associated with increased fungal virulence and the expansion of pathogenic Proteobacteria. Thus, although specific Prevotella species may exhibit different properties (43), the general beneficial role of Bacteriodetes in lung homeostasis (43), immune tolerance (44), and prevention of Th17 cell activation (45) is consistent with the abundance of Prevotella spp. in LR patients and loss in HR patients. Similar to what was observed in patients with AML following induction therapy (46), in our HR cohort, we did not observe the expansion of pathogenic Proteobacteria, probably because of first-line therapy with broad-spectrum piperacillin-tazobactam or carbapenems. Interestingly, however, Gammaproteobacteria that usually benefit from airway inflammation (47) and whose domination in the feces was a strong independent predictor of pulmonary complications in HSCT (7) were more abundantly present in HR than LR patients. Both Acinetobacter baumannii and Stenotrophomonas maltophilia are fatal infectious complications in HSCT (48, 49).

Overall, these results, while consistent with previous findings in gut (7), provide evidence for significant differences in oral microbial composition of patients at different risks for IFI. Supported by the high number of patients enrolled, the results clearly highlight the major contribution of neutropenia, the associated antibiotic use, and the occurrence of mucositis in the differences observed between HR and LR patients. As such, these results may pave the way for further studies to uncover associations between the many different risk factors for IFI and the changes in the microbiome. In addition, considering the tolerability and rapidity of the oropharyngeal swabbing, including the advantage of self-collection (50), this sampling method appears highly feasible and relatively low cost. Thus, if corroborated by further studies, the loss of alpha diversity associated with the loss of beneficial Clostridiales and Bacteroidetes could help delineate patients at risk of IFI, thereby providing information for antimicrobial therapy optimization. Indeed, HR patients would benefit from an antifungal prophylactic-based approach, as opposed to LR patients, for whom a fungal diagnostic-based approach is recommended to reduce overtreatment and unintended collateral damage to beneficial commensals. Discriminating patients who will benefit or not from antimicrobial prophylaxis will also help reduce the antimicrobial resistance crisis. In this regard, this study is a proof-of-concept demonstration that microbial metabolites, such as 3-IAld, known to protect from GvHD in murine HSCT (51), also reduce the risk of IFI in these patients. Indeed, 3-IAld could protect mice against aspergillosis, likely by activating AhR in ILC3 and inducing the production of interleukin-22 (IL-22). This mechanism recently has been shown to protect mice against mucosal candidiasis and dextran sodium sulfate-induced colitis (25, 52). Our results would expand the potential application of 3-IAld in the respiratory tract, in line with its suggested efficacy in cystic fibrosis (53). These wide-ranging activities are associated with a favorable safety profile (54), as expected for a molecule endogenously produced under physiological conditions, a fundamental prerequisite for clinical translation. More generally, this type of study reinforces the notion that mining the microbiota for microbial- and metabolite-based therapies could inform future interventional strategies in patients at risk for IFI.

MATERIALS AND METHODS

Study design and data collection.

The study was approved by the institutional review board of the University of Perugia, Italy, protocol number 2014-026. All subjects gave written informed consent in accordance with the Declaration of Helsinki. A total of 173 patients diagnosed with hematological malignancies were enrolled between November 2015 and November 2017. Clinical records from each patient were reviewed, and demographic and clinical data, including age, gender, disease, chemotherapeutic regimen, neutropenia, antimicrobial treatments (Table 2), transplantation type, and conditioning regimens (see Table S1 in the supplemental material), were recorded. The risk of IFI rather than the actual incidence of IFI was chosen as a surrogate primary endpoint to overcome the inherent diagnostics variability and inconsistencies of a multicenter study. In addition, as the epidemiology of IFI in hematological patients has shifted in recent years from a yeast (mainly Candida spp.) to a mold (mainly Aspergillus spp.) predominant etiology (55), the term IFI points to Aspergillus spp. as the most common causative agent in this study.

TABLE 2.

Characteristics of patients

Parameter Value(s)
Patients
    Total, n 173
    Age in yr, median (range) 54.5 (25–65)
    Female, n (%) 72 (41.6)
    Male, n (%) 101 (58.4)
    Pharyngeal swabs (no.) 945
Underlying disease, n (%)
    Acute myeloid leukemia 51 (29.5)
    Acute lymphoid leukemia 9 (5.2)
    Lymphoma 58 (33.5)
    Myeloma 44 (25.5)
    Chronic lymphatic leukemia 3 (1.7)
    Myelofibrosis 2 (1.1)
    Chronic myeloid leukemia 1 (0.6)
    Aplastic anemia 1 (0.6)
    Others 4 (2.3)
Disease status, n (%)
    At diagnosis 59 (34.1)
    Complete remission 63 (36.4)
    Partial remission 19 (11)
    Refractory/relapse 14 (8.1)
    Progression 18 (10.4)
Treatment, n (%)
    No. of cycles 424
    Myeloablative chemotherapy 201 (47.4)
    Reduced intensity chemotherapy 67 (15.8)
    Others (i.e., monoclonal antibodies, protease inhibitors, and immunomodulatory drugs) 156 (36.8)
Cycles of steroid therapy
    No. of cycles 295 (69.6)
    Duration, >7 days 94 (31.9)
Neutropenia (therapy related), n (%)
    Severe (<500 neutrophils/μl) 213 (50.2)
    Duration, >10 days 135 (63.4)
Antibiotics, no. (%)
    Total administered 458
    Prophylaxisa 105 (22.9)
    Therapy 353 (77.1)
    Broad-spectrum antibiotics (i.e., piperacillin-tazobactam or ceftazidime ± aminoglycoside or tigecycline; carbapenem ± aminoglycoside or tigecycline) 258 (73)
    Narrow-spectrum antibiotics (i.e., fluorochinolones or oral penicillin but not antimicrobial combinations) 95 (27)
Antifungals, no. (%)
    Total administered 159
    Prophylaxis (fluconazole [n = 42], micafungin [n = 17], l-AmB [n = 14], itraconazole [n = 17], voriconazole [n = 3], posaconazole [n = 11], anidulafungin [n = 1]) 105 (66)
    Therapy (voriconazole [n = 9], l-AmB [n = 18], caspofungin [n = 10], anidulafungin [n = 3], micafungin [n = 1], posaconazole [n = 1], itraconazole [n = 2], fluconazole [n = 6], isavuconazole [n = 4]) 54 (34)
a

Seventy-three of 105 (69.5) are PCP prophylaxis, and 32 of 105 (30.5) are antibacterial prophylaxis.

Sample collection, processing, and sequencing for microbial composition.

A total of 945 pharyngeal swabs were collected, on average, monthly from the diagnosis and up to 6 months (Table 2). Swabbing was performed by the treating physicians at the time of outpatient visits according to standard procedures. Swabs were stored in liquid Amies medium (Copan Diagnostics Inc.) in each participating center before delivery to the University of Perugia for sample extraction. On the day of extraction, pharyngeal samples were thawed on ice, transferred into 2-ml Eppendorf tubes, and centrifuged at 4°C for 15 min. The supernatant was collected and stored, while the pellet further processed for DNA extraction and sequencing as described in the supplemental material.

Mice, infection, and treatment.

C57BL/6 female mice, 6 to 8 weeks old (Charles River Laboratories, Calco, Italy), were anesthetized by inhalation of 3% isoflurane in oxygen before intranasal (i.n.) instillation of 2 × 107/20 μl saline viable resting conidia from the A. fumigatus AF293 strain (56), treated, and analyzed as described in the supplemental material. Murine experiments were performed according to the Italian Approved Animal Welfare Authorization 360/2015-PR and legislative degree 26/2014 regarding the animal license obtained by the Italian Ministry of Health, lasting for 5 years (2015 to 2020).

Murine 16S RNA sequencing and qPCR.

DNA was isolated from murine lung using glass beads (Sigma) followed by a QIAamp DNA minikit (Qiagen). The bacterial microbiota was evaluated by 16S rRNA. The V3-V4 region of the bacterial 16S genes was sequenced using the MiSeq platform (Illumina). Sequencing libraries were prepared using a NEXTERA XT DNA sample preparation kit (Illumina). Statistical analysis was performed as described in the supplemental material. Quantification of bacteria was done from standard curves established by quantitative PCR (qPCR) as previously described (25, 57). qPCR was performed with Power SYBR green PCR master mix (Applied Biosystems) using a StepOne Plus (Applied Biosystems) thermocycler. A short segment of the 16S rRNA gene was amplified using a conserved 16S rRNA-specific primer pair to determine the total amount of commensal bacteria. Using the same genomic DNA from each sample, group-specific primers were used to determine the amount of bacteria in the groups Enterobacteriaceae, Clostridia, and Bacteroidetes. Bacterial numbers were determined using standard curves constructed with reference bacteria specific for each bacterial group analyzed.

Real-time PCR.

Real-time PCR was performed using the iCycler iQ detection system (Bio-Rad) and iTaq universal SYBR green supermix (Bio-Rad). Total RNA was extracted using the RNeasy minikit (Qiagen, Milan, Italy) and reverse transcribed with Sensiscript reverse transcriptase (Qiagen). Amplification efficiencies were normalized against β-actin.

Cytokine and metabolite quantification.

The levels of IL-22 in lung homogenates were determined by using a specific enzyme-linked immunosorbent assay (ELISA) kit according to the manufacturer’s instructions (BioLegend). l-Tryptophan (trp), l-kynurenine (kyn), and 3-IAld were determined in pharyngeal samples by ultrahigh-performance liquid chromatography (UHPLC) and tandem mass spectrometry (MS/MS) as described in the supplemental material.

ACKNOWLEDGMENTS

We thank the SEIFEM (Epidemiology Survey of Invasive Fungal Infections in Hematological Malignancies) group for critical discussion.

A. Spolzino, F.M., L.F., A. Spadea, L.M., K.C., F.M., G.M., D.V., G.D., G.N., L.P., and F.A. contributed to the running of the clinical study and acquisition of the samples. C.C., M. I. Palmieri, G.R., T.Z., M.B., M.M.B., R.S., and G.L. contributed to the processing of the samples. E.N. performed the analysis of the human data sets. E.A. performed the analysis of the murine data sets. M. I. Palmieri, G.R., T.Z., and M.B. performed the murine experiments. L.E., K.C., and Z.S. performed the metabolomic analysis of tryptophan metabolites. M. Puccetti and S.G. contributed to the formulation of 3-IAld for oral administration. F.A. and L.R. contributed to the conception and design. C.C., E.N., A. Spolzino, V.N.T., F.A., and L.R. contributed to the interpretation of data. C.C., E.N., A. Spolzino, F.A., and L.R. were major contributors to writing the manuscript. All authors read and approved the final manuscript.

We have no potential conflicts of interest to report.

This work was supported by FunMeta Project (ERC-2011-AdG 293714), MicroTher (ERC-2018-PoC-813099), and Gilead (IN-IT-131-4525-518872.9) to L.R. and the Grant Agency of the Czech Republic (GACR No 17-24592Y) and the Czech Ministry of Education, Youth and Sports (CETOCOEN PLUS CZ.02.1.01/0.0/0.0/15_003/0000469; LM2015051 and CETOCOEN EXCELLENCE Teaming 2 project CZ.02.1.01/0.0/0.0/18_046/0015975; and Horizon2020 project 857560) to Z.S.

Footnotes

Supplemental material is available online only.

Supplemental file 1
Supplemental materials and methods; Tables S1 to S3; Fig. S1 to S10. Download IAI00105-21_Supp_1_seq10.pdf, PDF file, 4.0 MB (4.1MB, pdf)

For a commentary on this article, see https://doi.org/10.1128/IAI.00174-21.

Contributor Information

Luigina Romani, Email: luigina.romani@unipg.it.

Mairi C. Noverr, Tulane School of Medicine

REFERENCES

  • 1.Ananda-Rajah MR, Cheng A, Morrissey CO, Spelman T, Dooley M, Neville AM, Slavin M. 2011. Attributable hospital cost and antifungal treatment of invasive fungal diseases in high-risk hematology patients: an economic modeling approach. Antimicrob Agents Chemother 55:1953–1960. 10.1128/AAC.01423-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG, White TC. 2012. Hidden killers: human fungal infections. Sci Transl Med 4:165rv13. 10.1126/scitranslmed.3004404. [DOI] [PubMed] [Google Scholar]
  • 3.Pagano L, Busca A, Candoni A, Cattaneo C, Cesaro S, Fanci R, Nadali G, Potenza L, Russo D, Tumbarello M, Nosari A, Aversa F, et al. 2017. Risk stratification for invasive fungal infections in patients with hematological malignancies: SEIFEM recommendations. Blood Rev 31:17–29. 10.1016/j.blre.2016.09.002. [DOI] [PubMed] [Google Scholar]
  • 4.Oliveira-Coelho A, Rodrigues F, Campos A, Jr, Lacerda JF, Carvalho A, Cunha C. 2015. Paving the way for predictive diagnostics and personalized treatment of invasive aspergillosis. Front Microbiol 6:411. 10.3389/fmicb.2015.00411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stanzani M, Lewis RE, Fiacchini M, Ricci P, Tumietto F, Viale P, Ambretti S, Baccarani M, Cavo M, Vianelli N. 2013. A risk prediction score for invasive mold disease in patients with hematological malignancies. PLoS One 8:e75531. 10.1371/journal.pone.0075531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Staffas A, Burgos da Silva M, van den Brink MR. 2017. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood 129:927–933. 10.1182/blood-2016-09-691394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Harris B, Morjaria SM, Littmann ER, Geyer AI, Stover DE, Barker JN, Giralt SA, Taur Y, Pamer EG. 2016. Gut microbiota predict pulmonary infiltrates after allogeneic hematopoietic cell transplantation. Am J Respir Crit Care Med 194:450–463. 10.1164/rccm.201507-1491OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.McAleer JP, Kolls JK. 2018. Contributions of the intestinal microbiome in lung immunity. Eur J Immunol 48:39–49. 10.1002/eji.201646721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.O'Dwyer DN, Zhou X, Wilke CA, Xia M, Falkowski NR, Norman KC, Arnold KB, Huffnagle GB, Murray S, Erb-Downward JR, Yanik GA, Moore BB, Dickson RP. 2018. Lung dysbiosis, inflammation, and injury in hematopoietic cell transplantation. Am J Respir Crit Care Med 198:1312–1321. 10.1164/rccm.201712-2456OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rizzatti G, Lopetuso LR, Gibiino G, Binda C, Gasbarrini A. 2017. Proteobacteria: a common factor in human diseases. Biomed Res Int 2017:9351507. 10.1155/2017/9351507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Faner R, Sibila O, Agusti A, Bernasconi E, Chalmers JD, Huffnagle GB, Manichanh C, Molyneaux PL, Paredes R, Perez Brocal V, Ponomarenko J, Sethi S, Dorca J, Monso E. 2017. The microbiome in respiratory medicine: current challenges and future perspectives. Eur Respir J 49:1602086. 10.1183/13993003.02086-2016. [DOI] [PubMed] [Google Scholar]
  • 12.Dickson RP, Erb-Downward JR, Freeman CM, McCloskey L, Beck JM, Huffnagle GB, Curtis JL. 2015. Spatial variation in the healthy human lung microbiome and the adapted island model of lung biogeography. Annals ATS 12:821–830. 10.1513/AnnalsATS.201501-029OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Marsh RL, Kaestli M, Chang AB, Binks MJ, Pope CE, Hoffman LR, Smith-Vaughan HC. 2016. The microbiota in bronchoalveolar lavage from young children with chronic lung disease includes taxa present in both the oropharynx and nasopharynx. Microbiome 4:37. 10.1186/s40168-016-0182-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Man WH, de Steenhuijsen Piters WA, Bogaert D. 2017. The microbiota of the respiratory tract: gatekeeper to respiratory health. Nat Rev Microbiol 15:259–270. 10.1038/nrmicro.2017.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gao Z, Kang Y, Yu J, Ren L. 2014. Human pharyngeal microbiome may play a protective role in respiratory tract infections. Genomics Proteomics Bioinformatics 12:144–150. 10.1016/j.gpb.2014.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Human Microbiome Project Consortium. 2012. Structure, function and diversity of the healthy human microbiome. Nature 486:207–214. 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lemon KP, Klepac-Ceraj V, Schiffer HK, Brodie EL, Lynch SV, Kolter R. 2010. Comparative analyses of the bacterial microbiota of the human nostril and oropharynx. mBio 1:e00129-10. 10.1128/mBio.00129-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. 2011. Metagenomic biomarker discovery and explanation. Genome Biol 12:R60. 10.1186/gb-2011-12-6-r60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Knights D, Parfrey LW, Zaneveld J, Lozupone C, Knight R. 2011. Human-associated microbial signatures: examining their predictive value. Cell Host Microbe 10:292–296. 10.1016/j.chom.2011.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. 2012. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 3:289–306. 10.4161/gmic.19897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yun Y, Srinivas G, Kuenzel S, Linnenbrink M, Alnahas S, Bruce KD, Steinhoff U, Baines JF, Schaible UE. 2014. Environmentally determined differences in the murine lung microbiota and their relation to alveolar architecture. PLoS One 9:e113466. 10.1371/journal.pone.0113466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Marsland BJ, Gollwitzer ES. 2014. Host-microorganism interactions in lung diseases. Nat Rev Immunol 14:827–835. 10.1038/nri3769. [DOI] [PubMed] [Google Scholar]
  • 23.Morjaria S, Schluter J, Taylor BP, Littmann ER, Carter RA, Fontana E, Peled JU, van den Brink MRM, Xavier JB, Taur Y. 2019. Antibiotic-induced shifts in fecal microbiota density and composition during hematopoietic stem cell transplantation. Infect Immun 87:e00206-19. 10.1128/IAI.00206-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Scales BS, Dickson RP, Huffnagle GB. 2016. A tale of two sites: how inflammation can reshape the microbiomes of the gut and lungs. J Leukoc Biol 100:943–950. 10.1189/jlb.3MR0316-106R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D'Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L. 2013. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39:372–385. 10.1016/j.immuni.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 26.Hong BY, Sobue T, Choquette L, Dupuy AK, Thompson A, Burleson JA, Salner AL, Schauer PK, Joshi P, Fox E, Shin DG, Weinstock GM, Strausbaugh LD, Dongari-Bagtzoglou A, Peterson DE, Diaz PI. 2019. Chemotherapy-induced oral mucositis is associated with detrimental bacterial dysbiosis. Microbiome 7:66. 10.1186/s40168-019-0679-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Teufelberger AR, Broker BM, Krysko DV, Bachert C, Krysko O. 2019. Staphylococcus aureus orchestrates type 2 airway diseases. Trends Mol Med 25:696–707. 10.1016/j.molmed.2019.05.003. [DOI] [PubMed] [Google Scholar]
  • 28.Teegavarapu PS, Puppala M, Pingali SR, Patel A, Ibrahim I, Mukherjee A, Rice L, Padmanabhan Iyer S. 2015. Staphylococcus species leads the Eskape infections in various hematological malignancies. Blood 126:4623–4623. 10.1182/blood.V126.23.4623.4623. [DOI] [Google Scholar]
  • 29.Shelburne SA, Ajami NJ, Chibucos MC, Beird HC, Tarrand J, Galloway-Pena J, Albert N, Chemaly RF, Ghantoji SS, Marsh L, Pemmaraju N, Andreeff M, Shpall EJ, Wargo JA, Rezvani K, Alousi A, Bruno VM, Futreal PA, Petrosino JF, Kontoyiannis DP. 2015. Implementation of a pan-genomic approach to investigate holobiont-infecting microbe interaction: a case report of a leukemic patient with invasive mucormycosis. PLoS One 10:e0139851. 10.1371/journal.pone.0139851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fedosenko S, Karnaushkina M, Ogorodova L, Petrov V, Kulikov E, Kirillova N. 2017. Sources of origin of the oropharyngeal microbiota in patients with different phenotypes of COPD. Eur Resp J 50:OA1500. 10.1183/1393003.congress-2017.OA1500. [DOI] [Google Scholar]
  • 31.Jovanovic M, Tosic T, Jovanovic S, Stosovic R, Stevanovic G, Velebit B, Zervos MJ. 2018. Presence of the esp gene in Enterococcus faecium derived from oropharyngeal microbiota of haematology patients. Arch Oral Biol 88:54–59. 10.1016/j.archoralbio.2018.01.008. [DOI] [PubMed] [Google Scholar]
  • 32.Stein-Thoeringer CK, Nichols KB, Lazrak A, Docampo MD, Slingerland AE, Slingerland JB, Clurman AG, Armijo G, Gomes ALC, Shono Y, Staffas A, Burgos da Silva M, Devlin SM, Markey KA, Bajic D, Pinedo R, Tsakmaklis A, Littmann ER, Pastore A, Taur Y, Monette S, Arcila ME, Pickard AJ, Maloy M, Wright RJ, Amoretti LA, Fontana E, Pham D, Jamal MA, Weber D, Sung AD, Hashimoto D, Scheid C, Xavier JB, Messina JA, Romero K, Lew M, Bush A, Bohannon L, Hayasaka K, Hasegawa Y, Vehreschild M, Cross JR, Ponce DM, Perales MA, Giralt SA, Jenq RR, Teshima T, Holler E, Chao NJ, et al. 2019. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science 366:1143–1149. 10.1126/science.aax3760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bertolini M, Ranjan A, Thompson A, Diaz PI, Sobue T, Maas K, Dongari-Bagtzoglou A. 2019. Candida albicans induces mucosal bacterial dysbiosis that promotes invasive infection. PLoS Pathog 15:e1007717. 10.1371/journal.ppat.1007717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hamilton S, Bongaerts RJ, Mulholland F, Cochrane B, Porter J, Lucchini S, Lappin-Scott HM, Hinton JC. 2009. The transcriptional programme of Salmonella enterica serovar Typhimurium reveals a key role for tryptophan metabolism in biofilms. BMC Genomics 10:599. 10.1186/1471-2164-10-599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wellington S, Nag PP, Michalska K, Johnston SE, Jedrzejczak RP, Kaushik VK, Clatworthy AE, Siddiqi N, McCarren P, Bajrami B, Maltseva NI, Combs S, Fisher SL, Joachimiak A, Schreiber SL, Hung DT. 2017. A small-molecule allosteric inhibitor of Mycobacterium tuberculosis tryptophan synthase. Nat Chem Biol 13:943–950. 10.1038/nchembio.2420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Agus A, Planchais J, Sokol H. 2018. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23:716–724. 10.1016/j.chom.2018.05.003. [DOI] [PubMed] [Google Scholar]
  • 37.Dehhaghi M, Kazemi Shariat Panahi H, Guillemin GJ. 2019. Microorganisms, tryptophan metabolism, and kynurenine pathway: a complex interconnected loop influencing human health status. Int J Tryptophan Res 12:1178646919852996. 10.1177/1178646919852996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rao M, Dodoo E, Zumla A, Maeurer M. 2019. Immunometabolism and pulmonary infections: implications for protective immune responses and host-directed therapies. Front Microbiol 10:962. 10.3389/fmicb.2019.00962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Galloway-Pena JR, Peterson CB, Malik F, Sahasrabhojane PV, Shah DP, Brumlow CE, Carlin LG, Chemaly RF, Im JS, Rondon G, Felix E, Veillon L, Lorenzi PL, Alousi AM, Jenq RR, Kontoyiannis DP, Shpall EJ, Shelburne SA, Okhuysen PC. 2019. Fecal microbiome, metabolites, and stem cell transplant outcomes: a single-center pilot study. Open Forum Infect Dis 6:ofz173. 10.1093/ofid/ofz173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stokes JM, Lopatkin AJ, Lobritz MA, Collins JJ. 2019. Bacterial metabolism and antibiotic efficacy. Cell Metab 30:251–259. 10.1016/j.cmet.2019.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shono Y, Docampo MD, Peled JU, Perobelli SM, Velardi E, Tsai JJ, Slingerland AE, Smith OM, Young LF, Gupta J, Lieberman SR, Jay HV, Ahr KF, Porosnicu Rodriguez KA, Xu K, Calarfiore M, Poeck H, Caballero S, Devlin SM, Rapaport F, Dudakov JA, Hanash AM, Gyurkocza B, Murphy GF, Gomes C, Liu C, Moss EL, Falconer SB, Bhatt AS, Taur Y, Pamer EG, van den Brink MRM, Jenq RR. 2016. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci Transl Med 8:339ra71. 10.1126/scitranslmed.aaf2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Galloway-Pena JR, Shi Y, Peterson CB, Sahasrabhojane P, Gopalakrishnan V, Brumlow CE, Daver NG, Alfayez M, Boddu PC, Khan MAW, Wargo JA, Do KA, Jenq RR, Kontoyiannis DP, Shelburne SA. 2019. Gut microbiome signatures are predictive of infectious risk following induction therapy for acute myeloid leukemia. Clin Infect Dis 71:63–71. 10.1093/cid/ciz777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Larsen JM. 2017. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 151:363–374. 10.1111/imm.12760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, Nicod LP, Lloyd CM, Marsland BJ. 2014. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med 20:642–647. 10.1038/nm.3568. [DOI] [PubMed] [Google Scholar]
  • 45.Budden KF, Shukla SD, Rehman SF, Bowerman KL, Keely S, Hugenholtz P, Armstrong-James DPH, Adcock IM, Chotirmall SH, Chung KF, Hansbro PM. 2019. Functional effects of the microbiota in chronic respiratory disease. Lancet Respir Med 7:907–920. 10.1016/S2213-2600(18)30510-1. [DOI] [PubMed] [Google Scholar]
  • 46.Galloway-Pena JR, Smith DP, Sahasrabhojane P, Ajami NJ, Wadsworth WD, Daver NG, Chemaly RF, Marsh L, Ghantoji SS, Pemmaraju N, Garcia-Manero G, Rezvani K, Alousi AM, Wargo JA, Shpall EJ, Futreal PA, Guindani M, Petrosino JF, Kontoyiannis DP, Shelburne SA. 2016. The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia. Cancer 122:2186–2196. 10.1002/cncr.30039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Huffnagle GB, Dickson RP, Lukacs NW. 2017. The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol 10:299–306. 10.1038/mi.2016.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Scheich S, Koenig R, Wilke AC, Lindner S, Reinheimer C, Wichelhaus TA, Hogardt M, Kempf VAJ, Kessel J, Weber S, Martin H, Bug G, Serve H, Steffen B. 2018. Stenotrophomonas maltophilia colonization during allogeneic hematopoietic stem cell transplantation is associated with impaired survival. PLoS One 13:e0201169. 10.1371/journal.pone.0201169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Al-Anazi KA, Al-Jasser AM. 2014. Infections caused by Acinetobacter baumannii in recipients of hematopoietic stem cell transplantation. Front Oncol 4:186. 10.3389/fonc.2014.00186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Akmatov MK, Koch N, Vital M, Ahrens W, Flesch-Janys D, Fricke J, Gatzemeier A, Greiser H, Gunther K, Illig T, Kaaks R, Krone B, Kuhn A, Linseisen J, Meisinger C, Michels K, Moebus S, Nieters A, Obi N, Schultze A, Six-Merker J, Pieper DH, Pessler F. 2017. Determination of nasal and oropharyngeal microbiomes in a multicenter population-based study–findings from pretest 1 of the German National Cohort. Sci Rep 7:1855. 10.1038/s41598-017-01212-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Swimm A, Giver CR, DeFilipp Z, Rangaraju S, Sharma A, Ulezko Antonova A, Sonowal R, Capaldo C, Powell D, Qayed M, Kalman D, Waller EK. 2018. Indoles derived from intestinal microbiota act via type I interferon signaling to limit graft-versus-host disease. Blood 132:2506–2519. 10.1182/blood-2018-03-838193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Borghi M, Pariano M, Solito V, Puccetti M, Bellet MM, Stincardini C, Renga G, Vacca C, Sellitto F, Mosci P, Brancorsini S, Romani L, Costantini C. 2019. Targeting the aryl hydrocarbon receptor with indole-3-aldehyde protects from vulvovaginal candidiasis via the IL-22-IL-18 cross-talk. Front Immunol 10:2364. 10.3389/fimmu.2019.02364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Puccetti M, Paolicelli G, Oikonomou V, De Luca A, Renga G, Borghi M, Pariano M, Stincardini C, Scaringi L, Giovagnoli S, Ricci M, Romani L, Zelante T. 2018. Towards targeting the aryl hydrocarbon receptor in cystic fibrosis. Mediators Inflamm 2018:1601486. 10.1155/2018/1601486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zelante T, Puccetti M, Giovagnoli S, Romani L. 2021. Regulation of host physiology and immunity by microbial indole-3-aldehyde. Curr Opin Immunol 70:27–32. 10.1016/j.coi.2020.12.004. [DOI] [PubMed] [Google Scholar]
  • 55.Pagano L, Caira M, Candoni A, Offidani M, Fianchi L, Martino B, Pastore D, Picardi M, Bonini A, Chierichini A, Fanci R, Caramatti C, Invernizzi R, Mattei D, Mitra ME, Melillo L, Aversa F, Van Lint MT, Falcucci P, Valentini CG, Girmenia C, Nosari A. 2006. The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study. Haematologica 91:1068–1075. [PubMed] [Google Scholar]
  • 56.Romani L, Oikonomou V, Moretti S, Iannitti RG, D'Adamo MC, Villella VR, Pariano M, Sforna L, Borghi M, Bellet MM, Fallarino F, Pallotta MT, Servillo G, Ferrari E, Puccetti P, Kroemer G, Pessia M, Maiuri L, Goldstein AL, Garaci E. 2017. Thymosin alpha1 represents a potential potent single-molecule-based therapy for cystic fibrosis. Nat Med 23:590–600. 10.1038/nm.4305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Barman M, Unold D, Shifley K, Amir E, Hung K, Bos N, Salzman N. 2008. Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect Immun 76:907–915. 10.1128/IAI.01432-07. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental file 1

Supplemental materials and methods; Tables S1 to S3; Fig. S1 to S10. Download IAI00105-21_Supp_1_seq10.pdf, PDF file, 4.0 MB (4.1MB, pdf)


Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES