Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Lung Cancer ; 194: 107862, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959670

ABSTRACT

Despite the efficacy of immunotherapy in non-small cell lung cancer (NSCLC), the majority of the patients experience relapse with limited subsequent treatment options. Preclinical studies of various epithelial tumors, such as melanoma and NSCLC, have shown that harnessing the gut microbiome resulted in improvement of therapeutic responses to immunotherapy. Is this review, we summarize the role of microbiome, including lung and gut microbiome in the context of NSCLC, provide overview of the mechanisms of microbiome in efficacy and toxicity of chemotherapies and immunotherapies, and address current ongoing clinical trials for NSCLC including fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Gastrointestinal Microbiome/immunology , Lung Neoplasms/microbiology , Lung Neoplasms/therapy , Lung Neoplasms/immunology , Immunotherapy/methods , Fecal Microbiota Transplantation/methods , Animals
2.
Cell ; 187(13): 3373-3389.e16, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906102

ABSTRACT

The gut microbiota influences the clinical responses of cancer patients to immunecheckpoint inhibitors (ICIs). However, there is no consensus definition of detrimental dysbiosis. Based on metagenomics (MG) sequencing of 245 non-small cell lung cancer (NSCLC) patient feces, we constructed species-level co-abundance networks that were clustered into species-interacting groups (SIGs) correlating with overall survival. Thirty-seven and forty-five MG species (MGSs) were associated with resistance (SIG1) and response (SIG2) to ICIs, respectively. When combined with the quantification of Akkermansia species, this procedure allowed a person-based calculation of a topological score (TOPOSCORE) that was validated in an additional 254 NSCLC patients and in 216 genitourinary cancer patients. Finally, this TOPOSCORE was translated into a 21-bacterial probe set-based qPCR scoring that was validated in a prospective cohort of NSCLC patients as well as in colorectal and melanoma patients. This approach could represent a dynamic diagnosis tool for intestinal dysbiosis to guide personalized microbiota-centered interventions.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome , Immunotherapy , Lung Neoplasms , Neoplasms , Female , Humans , Male , Akkermansia , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Dysbiosis/microbiology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Lung Neoplasms/microbiology , Lung Neoplasms/drug therapy , Metagenomics/methods , Neoplasms/microbiology , Treatment Outcome
3.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928392

ABSTRACT

Lung cancer (LC) is one of the most prevalent cancers in both men and women and today is still characterized by high mortality and lethality. Several biomarkers have been identified for evaluating the prognosis of non-small cell lung cancer (NSCLC) patients and selecting the most effective therapeutic strategy for these patients. The introduction of innovative targeted therapies and immunotherapy with immune checkpoint inhibitors (ICIs) for the treatment of NSCLC both in advanced stages and, more recently, also in early stages, has revolutionized and significantly improved the therapeutic scenario for these patients. Promising evidence has also been shown by analyzing both micro-RNAs (miRNAs) and the lung/gut microbiota. MiRNAs belong to the large family of non-coding RNAs and play a role in the modulation of several key mechanisms in cells such as proliferation, differentiation, inflammation, and apoptosis. On the other hand, the microbiota (a group of several microorganisms found in human orgasms such as the gut and lungs and mainly composed by bacteria) plays a key role in the modulation of inflammation and, in particular, in the immune response. Some data have shown that the microbiota and the related microbiome can modulate miRNAs expression and vice versa by regulating several intracellular signaling pathways that are known to play a role in the pathogenesis of lung cancer. This evidence suggests that this axis is key to predicting the prognosis and effectiveness of ICIs in NSCLC treatment and could represent a new target in the treatment of NSCLC. In this review, we highlight the most recent evidence and data regarding the role of both miRNAs and the lung/gut microbiome in the prediction of prognosis and response to ICI treatment, focusing on the link between miRNAs and the microbiome. A new potential interaction based on the underlying modulated intracellular signaling pathways is also shown.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , MicroRNAs , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/genetics , MicroRNAs/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Immune Checkpoint Inhibitors/therapeutic use , Microbiota , Gastrointestinal Microbiome/drug effects , Prognosis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Animals
4.
Int J Mol Sci ; 25(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38396998

ABSTRACT

Next-generation sequencing technologies have started a new era of respiratory tract research in recent years. Alterations in the respiratory microbiome between healthy and malignant conditions have been revealed. However, the composition of the microbiome varies among studies, even in similar medical conditions. Also, there is a lack of complete knowledge about lung-gut microbiome interactions in lung cancer patients. The aim of this study was to explore the lung-gut axis in non-small-cell lung cancer (NSCLC) patients and the associations between lung-gut axis microbiota and clinical parameters (CRP, NLR, LPS, CD8, and PD-L1). Lung tissue and fecal samples were used for bacterial 16S rRNA sequencing. The results revealed, for the first time, that the bacterial richness in lung tumor tissue gradually decreased with an increase in the level of PD-L1 expression (p < 0.05). An analysis of ß-diversity indicated a significant positive correlation between the genera Romboutsia and Alistipes in both the lung tumor biopsies and stool samples from NSCLC patients (p < 0.05). Survival analysis showed that NSCLC patients with higher bacterial richness in their stool samples had prolonged overall survival (HR: 2.06, 95% CI: 1.025-4.17, p = 0.0426).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome , Lung Neoplasms , Lung , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/analysis , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Lung/microbiology , Lung/pathology , Lung Neoplasms/microbiology , RNA, Ribosomal, 16S/genetics
5.
Pol J Microbiol ; 72(4): 467-475, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38103007

ABSTRACT

Lung malignancies have a substantial impact on cancer incidence and mortality worldwide. Even though many factors involved in the development of the disease are known, many questions remain unanswered. Previous studies suggest that the intestinal microbiota may have a role in developing malignant diseases. According to some findings, the microbiota has proven to be a key modulator of carcinogenic processes and the immune response against cancer cells, potentially influencing the effectiveness of immunotherapy. In our study, we characterized culturable microorganisms associated with non-small cell lung cancer (NSCLC) that can be recovered from rectal swabs and mouthwash. In addition, we also explored differences in the culturable microbiota with two main types of NSCLC - adenocarcinoma (ADC) and squamous cell carcinoma (SCC). With 141 patients included in the study (86 ADC and 55 SCC cases), a significant difference was observed between the two types in seven bacterial species (Collinsella, Corynebacterium, Klebsiella, Lactobacillus, Neisseria, Rothia, and Streptococcus), including the site of origin. The relationship between microbial dysbiosis and lung cancer is poorly understood; future research could shed light on the links between gut microbiota and lung cancer development.


Subject(s)
Adenocarcinoma , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Microbiota , Humans , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/microbiology , Lung Neoplasms/pathology
6.
Sci Rep ; 12(1): 2210, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140255

ABSTRACT

Lung cancer (LC) and pulmonary tuberculosis (TB) are the deadliest neoplastic and bacterial infectious diseases worldwide, respectively. Clinicians and pathologists have long discussed the co-existence of LC and TB, and several epidemiologic studies have presented evidence indicating that TB could be associated with the development of LC, particularly adenocarcinoma. Nonetheless, this data remains controversial, and the mechanism which could underlie the association remains largely unexplored. Some bioinformatic studies have shown that human cancer biopsies have a very high frequency of bacterial DNA integration; since Mycobacterium Tuberculosis (MTb) is an intracellular pathogen, it could play an active role in the cellular transformation. Our group performed an exploratory study in a cohort of 88 LC patients treated at the Instituto Nacional de Cancelorogía (INCan) of Mexico City to evaluate the presence of MTb DNA in LC tissue specimens. For the first time, our results show the presence of the MTb IS6110 transposon in 40.9% (n = 36/88) of patients with lung adenocarcinomas. Additionally, through in-situ PCR we identified the presence of IS6110 in the nuclei of tumor cells. Furthermore, shotgun sequencing from two samples identified traces of MTb genomes present in tumor tissue, suggesting that similar Mtb strains could be infecting both patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/microbiology , DNA Transposable Elements/genetics , Lung Neoplasms/genetics , Lung Neoplasms/microbiology , Mycobacterium tuberculosis/genetics , Aged , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/pathology , Cohort Studies , DNA, Bacterial/genetics , Female , Humans , Lung Neoplasms/complications , Lung Neoplasms/pathology , Male , Mexico , Middle Aged , Survival Analysis , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/microbiology
7.
Gut ; 71(3): 457-466, 2022 03.
Article in English | MEDLINE | ID: mdl-34253574

ABSTRACT

OBJECTIVE: In this study, we determined whether Helicobacter pylori (H. pylori) infection dampens the efficacy of cancer immunotherapies. DESIGN: Using mouse models, we evaluated whether immune checkpoint inhibitors or vaccine-based immunotherapies are effective in reducing tumour volumes of H. pylori-infected mice. In humans, we evaluated the correlation between H. pylori seropositivity and the efficacy of the programmed cell death protein 1 (PD-1) blockade therapy in patients with non-small-cell lung cancer (NSCLC). RESULTS: In mice engrafted with MC38 colon adenocarcinoma or B16-OVA melanoma cells, the tumour volumes of non-infected mice undergoing anticytotoxic T-lymphocyte-associated protein 4 and/or programmed death ligand 1 or anti-cancer vaccine treatments were significantly smaller than those of infected mice. We observed a decreased number and activation status of tumour-specific CD8+ T cells in the tumours of infected mice treated with cancer immunotherapies independent of the gut microbiome composition. Additionally, by performing an in vitro co-culture assay, we observed that dendritic cells of infected mice promote lower tumour-specific CD8+ T cell proliferation. We performed retrospective human clinical studies in two independent cohorts. In the Dijon cohort, H. pylori seropositivity was found to be associated with a decreased NSCLC patient survival on anti-PD-1 therapy. The survival median for H. pylori seropositive patients was 6.7 months compared with 15.4 months for seronegative patients (p=0.001). Additionally, in the Montreal cohort, H. pylori seropositivity was found to be associated with an apparent decrease of NSCLC patient progression-free survival on anti-PD-1 therapy. CONCLUSION: Our study unveils for the first time that the stomach microbiota affects the response to cancer immunotherapies and that H. pylori serology would be a powerful tool to personalize cancer immunotherapy treatment.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Colonic Neoplasms/drug therapy , Helicobacter Infections/complications , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Animals , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Helicobacter pylori , Humans , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Retrospective Studies
8.
J BUON ; 26(5): 1862-1870, 2021.
Article in English | MEDLINE | ID: mdl-34761593

ABSTRACT

PURPOSE: The characteristics of pathogenic microbes are useful for understanding the microbe-driven tumorigenesis. There is a lack of studies on the lung microecology for lung cancer (LC) patients without any respiratory infection. In this work, we aimed to describe the profiles of pathogenic microbes in lung microenvironment of non-small cell lung cancer (NSCLC) patients using pathogen targeted sequencing and 16S rDNA sequencing. METHODS: A total of 22 NSCLC patients (13 adenocarcinomas and 9 squamous cell carcinomas) without any pulmonary infection were enrolled. Among them, we collected 15 pieces of tumor tissues, 5 pieces of peritumoral tissues, 6 blood serum samples, and 5 broncho-alveolar lavage fluid (BALF) samples. Pathogen targeted sequencingand16S rDNA sequencing was performed for microbial classification. RESULTS: The pathogen targeted sequencing results showed that 33, 14, 11, and 27 pathogenic microorganisms were detected in tumor tissues, peritumoral tissues, blood samples, and BALF, respectively. No common microorganisms were shared by four sample types. However, some common elements were shared by three sets: Streptococcus cristatus, Enterococcus, Staphylococcus haemolyticus, Corynebacterium pseudodiphtheria, Acinetobacter jungii, Haemophilus haemolyticus and Haemophilus parainfluenzae. Based on the 16S rDNA sequencing of two BALF samples, there were 104 OTUs found in one BALF sample and 127 OTUs in the other BALF sample; among them, there were 82 common ones, such as OTU1, OTU10, OTU101, OTU105, OTU106, and so on. Based on the above microbial classification and abundance, there might be enriched function in COG terms like COG1132, COG0438 and COG0745, and KEGG terms like K06147, K02029, and K09687. CONCLUSION: This study emphasizes the role of the microbiome in LC patients without respiratory infection. These potential biomarkers of LC based on the taxonomic composition of pathogenic microorganisms might have clinical application.


Subject(s)
Adenocarcinoma/microbiology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Squamous Cell/microbiology , Lung Neoplasms/microbiology , Tumor Microenvironment , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Respiratory Tract Infections
9.
Thorac Cancer ; 12(24): 3293-3303, 2021 12.
Article in English | MEDLINE | ID: mdl-34693651

ABSTRACT

BACKGROUND: Lung cancer has the highest mortality rate among malignant tumors, with non-small cell lung cancer (NSCLC) being the most common type. As the main component of the human microflora, the intestinal flora interacts with the human body to affect immunity, metabolism, and the formation of diseases. METHODS: Forty-five patients with advanced NSCLC who received platinum-containing dual-drug chemotherapy were enrolled in a prospective exploratory cohort study. The intestinal flora was dynamically collected at baseline and after two chemotherapy cycles. Next-generation sequencing and metagenomics were then used to analyze the species and function of the intestinal flora at all levels. RESULTS: Significant differences in the intestinal flora of patients with NSCLC were found according to sex and age. At the family level, the abundances of Streptococcaceae, Lactobacillaceae, and Leuconostocaceae after platinum-containing dual-drug chemotherapy were significantly higher compared to those before chemotherapy. At the family level, patients with chemotherapy-induced gastrointestinal reactions had a significantly higher abundance of Leuconostocaceae than those without gastrointestinal responses. Meanwhile, patients with gastrointestinal reactions had higher metabolism, human diseases, cellular processes, and environmental information processing than those who did not. At the genus level, responders had higher abundances of Bacteroides compared to nonresponders. Moreover, nonresponders had higher levels of the six major metabolic pathways compared to responders. CONCLUSIONS: The intestinal flora of Chinese patients with advanced NSCLC differed according to sex and age. Moreover, significant differences in the intestinal flora were noted after chemotherapy, which could be associated with chemotherapy-induced gastrointestinal reactions and the efficacy of chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/microbiology , Aged , China , Cohort Studies , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Metagenomics , Middle Aged , Platinum/therapeutic use , Prospective Studies
10.
Int J Mol Sci ; 22(19)2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34638770

ABSTRACT

The gut microbiota (GM) is considered to constitute a powerful "organ" capable of influencing the majority of the metabolic, nutritional, physiological, and immunological processes of the human body. To date, five microbial-mediated mechanisms have been revealed that either endorse or inhibit tumorigenesis. Although the gastrointestinal and respiratory tracts are distant physically, they have common embryonic origin and similarity in structure. The lung microbiota is far less understood, and it is suggested that the crosslink between the human microbiome and lung cancer is a complex, multifactorial relationship. Several pathways linking their respective microbiota have reinforced the existence of a gut-lung axis (GLA). Regarding implications of specific GM in lung cancer therapy, a few studies showed that the GM considerably affects immune checkpoint inhibitor (ICI) therapy by altering the differentiation of regulatory T cells and thus resulting in changes in immunomodulation mechanisms, as discovered by assessing drug metabolism directly and by assessing the host immune modulation response. Additionally, the GM may increase the efficacy of chemotherapeutic treatment in lung cancer. The mechanism underlying the role of the GLA in the pathogenesis and progression of lung cancer and its capability for diagnosis, manipulation, and treatment need to be further explored.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome/immunology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms , T-Lymphocytes, Regulatory/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/immunology , Lung Neoplasms/microbiology , Lung Neoplasms/therapy , T-Lymphocytes, Regulatory/pathology
11.
Genes (Basel) ; 12(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34573440

ABSTRACT

This study was designed to characterize the microbiomes of the lung tissues of lung cancer patients. RNA-sequencing was performed on lung tumor samples from 49 patients with lung cancer. Metatranscriptomics data were analyzed using SAMSA2 and Kraken2 software. 16S rRNA sequencing was also performed. The heterogeneous cellular landscape and immune repertoires of the lung samples were examined using xCell and TRUST4, respectively. We found that nine bacteria were significantly enriched in the lung tissues of cancer patients, and associated with reduced overall survival (OS). We also found that subjects with mutations in the epidermal growth factor receptor gene were less likely to experience the presence of Pseudomonas. aeruginosa. We found that the presence of CD8+ T-cells, CD4+ naive T-cells, dendritic cells, and CD4+ central memory T cells were associated with a good prognosis, while the presence of pro B-cells was associated with a poor prognosis. Furthermore, high clone numbers were associated with a high ImmuneScore for all immune receptor repertoires. Clone numbers and diversity were significantly higher in unpresented subjects compared to presented subjects. Our results provide insight into the microbiota of human lung cancer, and how its composition is linked to the tumor immune microenvironment, immune receptor repertoires, and OS.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/microbiology , Tumor Microenvironment/immunology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/microbiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/microbiology , Gene Expression Profiling , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Metagenome , Mutation , Pilot Projects , RNA, Ribosomal, 16S , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, RNA
12.
Cancer Sci ; 112(8): 3005-3017, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34028936

ABSTRACT

Immunotherapy against cancer, through immune checkpoint inhibitors targeting the programmed cell death-1/programmed cell death-ligand 1 axis, is particularly successful in tumors by relieving the immune escape. However, interindividual responses to immunotherapy are often heterogeneous. Therefore, it is essential to screen out predictive tumor biomarkers. In this study, we analyzed the commensal microbiota in stool samples and paired sputum samples from 75 metastatic non-small-cell lung cancer (NSCLC) patients at baseline and during treatment with immune checkpoint inhibitors. Results showed distinct microbes' signatures between the gut microbiota and paired respiratory microbiota. The alpha diversity between the gut and respiratory microbiota was uncorrelated, and only the gut microbiota alpha diversity was associated with anti-programmed cell death-1 response. Higher gut microbiota alpha diversity indicated better response and more prolonged progression-free survival. Comparison of bacterial communities between responders and nonresponders showed some favorable/unfavorable microbes enriched in responders/nonresponders, indicating that commensal microbiota had potential predictive value for the response to immune checkpoint inhibitors. Generally, some rare low abundance gut microbes and high abundance respiratory microbes lead to discrepancies in microbial composition between responders and nonresponders. A significant positive correlation was observed between the abundance of Streptococcus and CD8+ T cells. These results highlighted the intimate relationship between commensal microbiota and the response to immunotherapy in NSCLC patients. Gut microbiota and respiratory microbiota are promising biomarkers to screen suitable candidates who are likely to benefit from immune checkpoint inhibitor-based immunotherapy.


Subject(s)
Bacteria/classification , Carcinoma, Non-Small-Cell Lung/therapy , Immune Checkpoint Inhibitors/administration & dosage , Lung Neoplasms/therapy , Sequence Analysis, DNA/methods , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Carcinoma, Non-Small-Cell Lung/microbiology , Chemoradiotherapy , Female , Gastrointestinal Microbiome/drug effects , High-Throughput Nucleotide Sequencing , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/microbiology , Male , Neoplasm Metastasis , Phylogeny , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Streptococcus/isolation & purification , Survival Analysis , Treatment Outcome
13.
PLoS One ; 16(4): e0249832, 2021.
Article in English | MEDLINE | ID: mdl-33891617

ABSTRACT

Following recent findings linking the human gut microbiota to gastrointestinal cancer and its treatment, the plausible relationship between lung microbiota and pulmonary cancer is explored. This study aims at characterizing the intratumoral and adjacent healthy tissue microbiota by applying a 16S rRNA gene amplicon sequencing protocol to tissue samples of 29 non-small cancer patients. Emphasis was put on contaminant management and a comprehensive comparison of bacterial composition between cancerous and healthy adjacent tissues of lung adenocarcinoma and squamous cell carcinoma is provided. A variable degree of similarity between the two tissues of a same patient was observed. Each patient seems to possess its own bacterial signature. The two types of cancer tissue do not have a distinct bacterial profile that is shared by every patient. In addition, enteric, potentially pathogenic and pro-inflammatory bacteria were more frequently found in cancer than healthy tissue. This work brings insights into the dynamic of bacterial communities in lung cancer and provides prospective data for more targeted studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/microbiology , Lung Neoplasms/microbiology , Microbiota , Aged , Aged, 80 and over , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Female , Humans , Lung/microbiology , Male , Middle Aged , Organ Specificity , RNA, Ribosomal, 16S/genetics
14.
Nat Microbiol ; 6(3): 277-288, 2021 03.
Article in English | MEDLINE | ID: mdl-33432149

ABSTRACT

The gut microbiome can influence the development of tumours and the efficacy of cancer therapeutics1-5; however, the multi-omics characteristics of antitumour bacterial strains have not been fully elucidated. In this study, we integrated metagenomics, genomics and transcriptomics of bacteria, and analyses of mouse intestinal transcriptome and serum metabolome data to reveal an additional mechanism by which bacteria determine the efficacy of cancer therapeutics. In gut microbiome analyses of 96 samples from patients with non-small-cell lung cancer, Bifidobacterium bifidum was abundant in patients responsive to therapy. However, when we treated syngeneic mouse tumours with commercial strains of B. bifidum to establish relevance for potential therapeutic uses, only specific B. bifidum strains reduced tumour burden synergistically with PD-1 blockade or oxaliplatin treatment by eliciting an antitumour host immune response. In mice, these strains induced tuning of the immunological background by potentiating the production of interferon-γ, probably through the enhanced biosynthesis of immune-stimulating molecules and metabolites.


Subject(s)
Bifidobacterium bifidum/physiology , Immune Checkpoint Inhibitors/therapeutic use , Probiotics/therapeutic use , Tumor Burden/drug effects , Animals , Bifidobacterium bifidum/classification , Bifidobacterium bifidum/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Drug Therapy, Combination , Gastrointestinal Microbiome , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Metabolome/drug effects , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Probiotics/administration & dosage , Species Specificity , Transcriptome/drug effects , Tryptophan/metabolism
15.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33375194

ABSTRACT

Infectious diseases represent a relevant issue in lung cancer patients. Bacterial and viral infections might influence the patients' prognosis, both directly affecting the immune system and indirectly impairing the outcome of anticancer treatments, mainly immunotherapy. In this analysis, we aimed to review the current evidence in order to clarify the complex correlation between infections and lung cancer. In detail, we mainly explored the potential impact on immunotherapy outcome/safety of (1) bacterial infections, with a detailed focus on antibiotics; and (2) viral infections, discriminating among (a) human immune-deficiency virus (HIV), (b) hepatitis B/C virus (HBV-HCV), and (c) Sars-Cov-2. A series of studies suggested the prognostic impact of antibiotic therapy administration, timing, and exposure ratio in patients treated with immune checkpoint inhibitors, probably through an antibiotic-related microbiota dysbiosis. Although cancer patients with HIV, HBV, and HCV were usually excluded from clinical trials evaluating immunotherapy, some retrospective and prospective trials performed in these patient subgroups reported similar results compared to those described in not-infected patients, with a favorable safety profile. Moreover, patients with thoracic cancers are particularly at risk of COVID-19 severe outcomes and mortality. Few reports speculated about the prognostic implications of anticancer therapy, including immunotherapy, in lung cancer patients with concomitant Sars-Cov-2 infection, showing, to date, inconsistent results. The correlation between infectious diseases and immunotherapy remains to be further explored and clarified in the context of dedicated trials. In clinical practice, the accurate and prompt multidisciplinary management of lung cancer patients with infections should be encouraged in order to select the best treatment options for these patients, avoiding unexpected toxicities, while maintaining the anticancer effect.


Subject(s)
Bacterial Infections/complications , COVID-19/complications , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/complications , Lung Neoplasms/therapy , Virus Diseases/complications , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/therapy , Anti-Bacterial Agents/administration & dosage , Bacterial Infections/drug therapy , Bacterial Infections/pathology , COVID-19/pathology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/virology , HIV/drug effects , Hepatitis B/complications , Hepatitis B/immunology , Hepatitis B/pathology , Hepatitis C/complications , Hepatitis C/drug therapy , Hepatitis C/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/microbiology , Lung Neoplasms/virology , Microbiota/drug effects , Microbiota/immunology , COVID-19 Drug Treatment
16.
PLoS One ; 15(11): e0242544, 2020.
Article in English | MEDLINE | ID: mdl-33237921

ABSTRACT

OBJECTIVES: Multidrug-resistant organisms (MDRO) are considered an emerging threat worldwide. Data covering the clinical impact of MDRO colonization in patients with solid malignancies, however, is widely missing. We sought to determine the impact of MDRO colonization in patients who have been diagnosed with Non-small cell lung cancer (NSCLC) who are at known high-risk for invasive infections. MATERIALS AND METHODS: Patients who were screened for MDRO colonization within a 90-day period after NSCLC diagnosis of all stages were included in this single-center retrospective study. RESULTS: Two hundred and ninety-five patients were included of whom 24 patients (8.1%) were screened positive for MDRO colonization (MDROpos) at first diagnosis. Enterobacterales were by far the most frequent MDRO detected with a proportion of 79.2% (19/24). MDRO colonization was present across all disease stages and more present in patients with concomitant diabetes mellitus. Median overall survival was significantly inferior in the MDROpos study group with a median OS of 7.8 months (95% CI, 0.0-19.9 months) compared to a median OS of 23.9 months (95% CI, 17.6-30.1 months) in the MDROneg group in univariate (p = 0.036) and multivariate analysis (P = 0.02). Exploratory analyses suggest a higher rate of non-cancer-related-mortality in MDROpos patients compared to MDROneg patients (p = 0.002) with an increased rate of fatal infections in MDROpos patients (p = 0.0002). CONCLUSIONS: MDRO colonization is an independent risk factor for inferior OS in patients diagnosed with NSCLC due to a higher rate of fatal infections. Empirical antibiotic treatment approaches should cover formerly detected MDR commensals in cases of (suspected) invasive infections.


Subject(s)
Bacteria/isolation & purification , Carcinoma, Non-Small-Cell Lung/microbiology , Drug Resistance, Multiple, Bacterial , Lung Neoplasms/microbiology , Adult , Aged , Aged, 80 and over , Bacteria/drug effects , Bacterial Infections/complications , Bacterial Infections/drug therapy , Bacterial Infections/mortality , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/therapy , Cause of Death , Comorbidity , Female , Humans , Length of Stay/statistics & numerical data , Lung Neoplasms/complications , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Male , Middle Aged , Nose/microbiology , Patient Admission/statistics & numerical data , Pharynx/microbiology , Rectum/microbiology , Retrospective Studies , Risk Factors
17.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33227982

ABSTRACT

Several studies in recent times have linked gut microbiome (GM) diversity to the pathogenesis of cancer and its role in disease progression through immune response, inflammation and metabolism modulation. This study focused on the use of network analysis and weighted gene co-expression network analysis (WGCNA) to identify the biological interaction between the gut ecosystem and its metabolites that could impact the immunotherapy response in non-small cell lung cancer (NSCLC) patients undergoing second-line treatment with anti-PD1. Metabolomic data were merged with operational taxonomic units (OTUs) from 16S RNA-targeted metagenomics and classified by chemometric models. The traits considered for the analyses were: (i) condition: disease or control (CTRLs), and (ii) treatment: responder (R) or non-responder (NR). Network analysis indicated that indole and its derivatives, aldehydes and alcohols could play a signaling role in GM functionality. WGCNA generated, instead, strong correlations between short-chain fatty acids (SCFAs) and a healthy GM. Furthermore, commensal bacteria such as Akkermansia muciniphila, Rikenellaceae, Bacteroides, Peptostreptococcaceae, Mogibacteriaceae and Clostridiaceae were found to be more abundant in CTRLs than in NSCLC patients. Our preliminary study demonstrates that the discovery of microbiota-linked biomarkers could provide an indication on the road towards personalized management of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Lung Neoplasms/genetics , Metabolome/immunology , Akkermansia/classification , Akkermansia/genetics , Akkermansia/isolation & purification , Alcohols/metabolism , Aldehydes/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Bacteroides/classification , Bacteroides/genetics , Bacteroides/isolation & purification , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/microbiology , Clostridiaceae/classification , Clostridiaceae/genetics , Clostridiaceae/isolation & purification , Databases, Genetic , Disease Progression , Drug Monitoring/methods , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/genetics , Humans , Immunotherapy/methods , Indoles/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/microbiology , Metabolome/genetics , Metagenomics/methods , Peptostreptococcus/classification , Peptostreptococcus/genetics , Peptostreptococcus/isolation & purification , Precision Medicine/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , RNA, Ribosomal, 16S/genetics , Signal Transduction
18.
Zhonghua Nei Ke Za Zhi ; 59(11): 887-893, 2020 Nov 01.
Article in Chinese | MEDLINE | ID: mdl-33120493

ABSTRACT

Objective: To analyze clinical characteristics and monitor microbiome changes in patients with anti-PD-1 associated colitis. Methods: Two patients with non-small cell lung cancer who developed colitis after treated with anti-PD-1 antibodies were retrospectively analyzed in Peking Union Medical College Hospital from January 2019 to January 2020. The clinical symptoms, endoscopic and pathological manifestations, as well microbiome changes were analyzed and compared during pre-treatment, post-treatment and relapse. Results: The main clinical manifestations included diarrhea, elevated inflammatory indicators, colonic mucosal diffuse hyperemic edema with erosion by endoscopy. Changes in the structure of crypts were common pathological characteristics. Glucocorticoids were effective agents, which achieved clinical remission and mucosal healing. The microbiome composition of OTUs was different. After glucocorticoid treatment, the alpha diversity Observed species, Shannon, Simpson, Chao1, ACE indexes all decreased. The Firmicutes decreased with Bacteroidetes increasing in phylum level; while the Bacteroides increased with Ruminococcaceae decreasing in genus level. Lactobacillus was the potentially beneficial genus. Conclusion: Patients developing anti-PD-1 associated colitis have characteristic clinical and pathological manifestations. Glucocorticoids are effective treatment. The fecal microbiome diversity, relative abundance of major phylum and genus have changed after treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/complications , Colitis/diagnosis , Gastrointestinal Microbiome , Lung Neoplasms/complications , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Bacteria/classification , Carcinoma, Non-Small-Cell Lung/microbiology , Colitis/microbiology , Glucocorticoids/therapeutic use , Humans , Lung Neoplasms/microbiology , Retrospective Studies
19.
Anticancer Res ; 40(9): 4807-4818, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878769

ABSTRACT

The microbiome is extremely important for human health; more recently its role in the context of cancer became clear. Microbial effects range from enhancing cancer immunity and cancer therapy efficacy, to promoting cancer progression and inhibiting treatment efficacy. These broad implications led researchers to investigate these specific interactions, as well as how modification of the microbiome can improve cancer survival and treatment efficacy. While these interactions are better established for cancers such as gastric cancer, they are far less understood in others. As non-small cell lung cancer (NSCLC) makes up the majority of lung cancer cases, and is among the top causes of cancer deaths worldwide, understanding the mechanisms by which the microbiome may impact progression and treatment is crucial to improve patient survival and treatment response. A literature review was conducted to reveal the crosslink between human microbiome and lung cancer. This includes immune priming, induction of pro- or anti-tumor response, and the local effects of intra-tumoral microbiota. Overall, this is a complex multifactorial relationship, and there are broad implications as to how this knowledge can improve cancer treatment. Solutions include manipulation of the microbiome using probiotics, bacterial vaccines and antibiotics. Bacteria biomarkers may also be used as a diagnostic tool.


Subject(s)
Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/microbiology , Lung Neoplasms/therapy , Microbiota/physiology , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Bacteria/drug effects , Bacteria/isolation & purification , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Immunomodulation , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Microbiota/drug effects , Treatment Outcome
20.
Sci Rep ; 10(1): 9094, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499509

ABSTRACT

Chemotherapy remains the gold standard for advanced cancer. Pemetrexed, a chemotherapeutic agent used in non-small cell lung cancer, can induce significant side effects in patients. Although microbiota's role in the efficacy and/or toxicity of chemotherapy agents has been demonstrated, the impacts of pemetrexed on the gut microbiota and on gastrointestinal inflammation remain unknown. The objective of this study was to evaluate the impact of pemetrexed and the tumor graft on the gut microbiota composition in immunodeficient mice. The faecal microbiota composition was studied with metabarcoding before, 24-h and one week after treatment. The colon epithelial barrier integrity was evaluated by histological examination, intestinal permeability measurement, and selected cytokines quantification. The tumor graft induced some variations in the microbiota composition. Pemetrexed further increased the relative abundance of Enterobacteriaceae and 3 families from the Firmicutes phylum: Enterococcaceae, Lactobacillaceae and Streptococcaceae. Pemetrexed also significantly altered the epithelial barrier integrity, which was associated with early inflammation. This pilot study shows that the association of a lung tumor graft with pemetrexed causes an alteration in the microbiota composition. Such information increases our knowledge about the impact of chemotherapy on the microbiota, which could help to minimize side effects and improve therapeutic effectiveness in the future.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Lung Neoplasms/drug therapy , Lung Neoplasms/microbiology , Neoplasm Transplantation , Pemetrexed/adverse effects , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Female , Heterografts , Inflammation , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lung Neoplasms/pathology , Mice, SCID , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL