Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 7096, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154092

ABSTRACT

The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1ß expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.


Subject(s)
Gallium , Lacticaseibacillus rhamnosus , Microbiota , Pancreatic Neoplasms , Polyphenols , Probiotics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/microbiology , Animals , Probiotics/administration & dosage , Mice , Female , Humans , Lacticaseibacillus rhamnosus/immunology , Polyphenols/pharmacology , Microbiota/immunology , Microbiota/drug effects , Cell Line, Tumor , Immunotherapy/methods , Mice, Inbred C57BL , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , T-Lymphocytes, Cytotoxic/immunology
2.
Front Immunol ; 15: 1408770, 2024.
Article in English | MEDLINE | ID: mdl-39119339

ABSTRACT

Introduction: Gut microbiota (GM) influences the occurrence and development of pancreatic cancer (PC), potentially through the involvement of inflammatory cytokines (IC) and immune cells (IM). We aimed to investigate the causal impact of the gut microbiota (GM) on pancreatic cancer (PC) and identify potential IC and IM mediators. Methods: The summary statistics data from whole-genome association studies of gut microbiota, immune cells, inflammatory cytokines, and four types of pancreatic tumors (MNP: Malignant neoplasm of pancreas; BNP: Benign neoplasm of pancreas; ADCP: Adenocarcinoma and ductal carcinoma of pancreas; NTCP: Neuroendocrine tumor and carcinoma of pancreas). Two-sample univariable Mendelian randomization (UVMR), multivariable Mendelian randomization (MVMR), and mediation analysis were employed to assess the causal relationship between gut microbiota (GM) and pancreatic cancer (PC), as well as potential IC and IM mediators. Results: The two-sample UVMR analysis showed causal relationships between 20 gut microbiota species and pancreatic cancer, with pancreatic cancer affecting the abundance of 37 gut microbiota species. Mediation analysis revealed that Interleukin-6 (IL-6), "CD4 on naive CD4+ T cell" and "SSC-A on HLA DR+ Natural Killer" mediated the causal effects of gut microbiota on pancreatic cancer. Conclusion: This Mendelian randomization study demonstrates causal relationships between several specific gut microbiota and pancreatic cancer, as well as potential mediators (IC, IM).


Subject(s)
Cytokines , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/microbiology , Gastrointestinal Microbiome/immunology , Genome-Wide Association Study , Risk Factors , Inflammation Mediators/metabolism
3.
BMC Microbiol ; 24(1): 235, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38956452

ABSTRACT

BACKGROUND: Patients with pancreatic ductal adenocarcinoma (PDAC) display an altered oral, gastrointestinal, and intra-pancreatic microbiome compared to healthy individuals. However, knowledge regarding the bile microbiome and its potential impact on progression-free survival in PDACs remains limited. METHODS: Patients with PDAC (n = 45), including 20 matched pairs before and after surgery, and benign controls (n = 16) were included prospectively. The characteristics of the microbiomes of the total 81 bile were revealed by 16  S-rRNA gene sequencing. PDAC patients were divided into distinct groups based on tumor marker levels, disease staging, before and after surgery, as well as progression free survival (PFS) for further analysis. Disease diagnostic model was formulated utilizing the random forest algorithm. RESULTS: PDAC patients harbor a unique and diverse bile microbiome (PCoA, weighted Unifrac, p = 0.038), and the increasing microbial diversity is correlated with dysbiosis according to key microbes and microbial functions. Aliihoeflea emerged as the genus displaying the most significant alteration among two groups (p < 0.01). Significant differences were found in beta diversity of the bile microbiome between long-term PFS and short-term PFS groups (PCoA, weighted Unifrac, p = 0.005). Bacillota and Actinomycetota were identified as altered phylum between two groups associated with progression-free survival in all PDAC patients. Additionally, we identified three biomarkers as the most suitable set for the random forest model, which indicated a significantly elevated likelihood of disease occurrence in the PDAC group (p < 0.0001). The area under the receiver operating characteristic (ROC) curve reached 80.8% with a 95% confidence interval ranging from 55.0 to 100%. Due to the scarcity of bile samples, we were unable to conduct further external verification. CONCLUSION: PDAC is characterized by an altered microbiome of bile ducts. Biliary dysbiosis is linked with progression-free survival in all PDACs. This study revealed the alteration of the bile microbiome in PDACs and successfully developed a diagnostic model for PDAC.


Subject(s)
Bile , Carcinoma, Pancreatic Ductal , Microbiota , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Bile/microbiology , Male , Female , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Microbiota/genetics , Middle Aged , Aged , Dysbiosis/microbiology , Progression-Free Survival , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Prospective Studies , RNA, Ribosomal, 16S/genetics
4.
Gan To Kagaku Ryoho ; 51(6): 603-607, 2024 Jun.
Article in Japanese | MEDLINE | ID: mdl-39009514

ABSTRACT

An association between periodontal disease and the development of pancreatic cancer has been pointed out since before. Advances in genome analysis technology have revealed that a pancreatic cancer-specific microbiome is formed in the intestines and tumors of pancreatic cancer patients and modifies the progression of pancreatic cancer. Disturbance of microbiome( dysbiosis)suppresses anti-tumor immunity against pancreatic cancer, promoting cancer progression. Therefore, attempts are being made to correct dysbiosis by administration of probiotics or transplantation of microbiome, which is especially activating immune checkpoint inhibitors against cancer. In addition, specific intratumor bacteria has been identified that create an immunosuppressive microenvironment through crosstalk with pancreatic cancer cells. In the future, analysis of the microbiome distribution in pancreatic cancers may determine the following treatment strategy as an individualized treatment. We hope that innovations in omics technology will reveal more detailed functions of microbiome and lead to the development of effective treatments for pancreatic cancer.


Subject(s)
Microbiota , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Dysbiosis/microbiology , Dysbiosis/immunology , Dysbiosis/therapy , Tumor Microenvironment
5.
J Med Virol ; 96(7): e29809, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39016466

ABSTRACT

Pancreatic cancer (PC) is a highly aggressive malignancy with a poor prognosis, making early diagnosis crucial for improving patient outcomes. While the gut microbiome, including bacteria and viruses, is believed to be essential in cancer pathogenicity, the potential contribution of the gut virome to PC remains largely unexplored. In this study, we conducted a comparative analysis of the gut viral compositional and functional profiles between PC patients and healthy controls, based on fecal metagenomes from two publicly available data sets comprising a total of 101 patients and 82 healthy controls. Our results revealed a decreasing trend in the gut virome diversity of PC patients with disease severity. We identified significant alterations in the overall viral structure of PC patients, with a meta-analysis revealing 219 viral operational taxonomic units (vOTUs) showing significant differences in relative abundance between patients and healthy controls. Among these, 65 vOTUs were enriched in PC patients, and 154 were reduced. Host prediction revealed that PC-enriched vOTUs preferentially infected bacterial members of Veillonellaceae, Enterobacteriaceae, Fusobacteriaceae, and Streptococcaceae, while PC-reduced vOTUs were more likely to infect Ruminococcaceae, Lachnospiraceae, Clostridiaceae, Oscillospiraceae, and Peptostreptococcaceae. Furthermore, we constructed random forest models based on the PC-associated vOTUs, achieving an optimal average area under the curve (AUC) of up to 0.879 for distinguishing patients from controls. Through additional 10 public cohorts, we demonstrated the reproducibility and high specificity of these viral signatures. Our study suggests that the gut virome may play a role in PC development and could serve as a promising target for PC diagnosis and therapeutic intervention. Future studies should further explore the underlying mechanisms of gut virus-bacteria interactions and validate the diagnostic models in larger and more diverse populations.


Subject(s)
Feces , Gastrointestinal Microbiome , Metagenomics , Pancreatic Neoplasms , Virome , Humans , Pancreatic Neoplasms/virology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/microbiology , Gastrointestinal Microbiome/genetics , Metagenomics/methods , Feces/virology , Feces/microbiology , Viruses/isolation & purification , Viruses/genetics , Viruses/classification , Metagenome , Bacteria/isolation & purification , Bacteria/classification , Bacteria/genetics , Middle Aged , Male , Female , Aged , Case-Control Studies
6.
Front Immunol ; 15: 1434771, 2024.
Article in English | MEDLINE | ID: mdl-39044834

ABSTRACT

The gut microbiome plays a significant role in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), influencing oncogenesis, immune responses, and treatment outcomes. Studies have identified microbial species like Porphyromonas gingivalis and Fusobacterium nucleatum, that promote PDAC progression through various mechanisms. Additionally, the gut microbiome affects immune cell activation and response to immunotherapy, including immune checkpoint inhibitors and CAR-T therapy. Specific microbes and their metabolites play a significant role in the effectiveness of immune checkpoint inhibitors (ICIs). Alterations in the gut microbiome can either enhance or diminish responses to PD-1/PD-L1 and CTLA-4 blockade therapy. Additionally, bacterial metabolites like trimethylamine N-oxide (TMAO) and lipopolysaccharide (LPS) impact antitumor immunity, offering potential targets to augment immunotherapy responses. Modulating the microbiome through fecal microbiota transplantation, probiotics, prebiotics, dietary changes, and antibiotics shows promise in PDAC treatment, although outcomes are highly variable. Dietary modifications, particularly high-fiber diets and specific fat consumption, influence microbiome composition and impact cancer risk. Combining microbiome-based therapies with existing treatments holds potential for improving PDAC therapy outcomes, but further research is needed to optimize their effectiveness.


Subject(s)
Carcinoma, Pancreatic Ductal , Gastrointestinal Microbiome , Pancreatic Neoplasms , Humans , Gastrointestinal Microbiome/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/immunology , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Animals , Probiotics/therapeutic use , Fecal Microbiota Transplantation , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use
7.
Gut Microbes ; 16(1): 2375483, 2024.
Article in English | MEDLINE | ID: mdl-38972056

ABSTRACT

Pancreatic cancer has a dismal prognosis, as it is often diagnosed at stage IV of the disease and is characterized by metastatic spread. Gut microbiota and its metabolites have been suggested to influence the metastatic spread by modulating the host immune system or by promoting angiogenesis. To date, the gut microbial profiles of metastatic and non-metastatic patients need to be explored. Taking advantage of the 16S metagenomic sequencing and the PEnalized LOgistic Regression Analysis (PELORA) we identified clusters of bacteria with differential abundances between metastatic and non-metastatic patients. An overall increase in Gram-negative bacteria in metastatic patients compared to non-metastatic ones was identified using this method. Furthermore, to gain more insight into how gut microbes can predict metastases, a machine learning approach (iterative Random Forest) was performed. Iterative Random Forest analysis revealed which microorganisms were characterized by a different level of relative abundance between metastatic and non-metastatic patients and established a functional relationship between the relative abundance and the probability of having metastases. At the species level, the following bacteria were found to have the highest discriminatory power: Anaerostipes hadrus, Coprobacter secundus, Clostridium sp. 619, Roseburia inulinivorans, Porphyromonas and Odoribacter at the genus level, and Rhodospirillaceae, Clostridiaceae and Peptococcaceae at the family level. Finally, these data were intertwined with those from a metabolomics analysis on fecal samples of patients with or without metastasis to better understand the role of gut microbiota in the metastatic process. Artificial intelligence has been applied in different areas of the medical field. Translating its application in the field of gut microbiota analysis may help fully exploit the potential information contained in such a large amount of data aiming to open up new supportive areas of intervention in the management of cancer.


Subject(s)
Bacteria , Gastrointestinal Microbiome , Machine Learning , Neoplasm Metastasis , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Male , Female , RNA, Ribosomal, 16S/genetics , Middle Aged , Feces/microbiology , Aged , Metagenomics
8.
Gut Microbes ; 16(1): 2374596, 2024.
Article in English | MEDLINE | ID: mdl-39024520

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by late diagnosis, rapid progression, and a high mortality rate. Its complex biology, characterized by a dense, stromal tumor environment with an immunosuppressive milieu, contributes to resistance against standard treatments like chemotherapy and radiation. This comprehensive review explores the dynamic role of the microbiome in modulating chemotherapy efficacy and outcomes in PDAC. It delves into the microbiome's impact on drug metabolism and resistance, and the interaction between microbial elements, drugs, and human biology. We also highlight the significance of specific bacterial species and microbial enzymes in influencing drug action and the immune response in the tumor microenvironment. Cutting-edge methodologies, including artificial intelligence, low-biomass microbiome analysis and patient-derived organoid models, are discussed, offering insights into the nuanced interactions between microbes and cancer cells. The potential of microbiome-based interventions as adjuncts to conventional PDAC treatments are discussed, paving the way for personalized therapy approaches. This review synthesizes recent research to provide an in-depth understanding of how the microbiome affects chemotherapy efficacy. It focuses on elucidating key mechanisms and identifying existing knowledge gaps. Addressing these gaps is crucial for enhancing personalized medicine and refining cancer treatment strategies, ultimately improving patient outcomes.


Subject(s)
Antineoplastic Agents , Carcinoma, Pancreatic Ductal , Gastrointestinal Microbiome , Pancreatic Neoplasms , Tumor Microenvironment , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/microbiology , Gastrointestinal Microbiome/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/microbiology , Tumor Microenvironment/drug effects , Antineoplastic Agents/therapeutic use , Animals , Bacteria/metabolism , Bacteria/drug effects , Bacteria/genetics , Bacteria/classification , Precision Medicine , Drug Resistance, Neoplasm
9.
Cancer Sci ; 115(8): 2738-2750, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38888048

ABSTRACT

Pancreatic head cancer (PHC) and pancreatic body/tail cancer (PBTC) have distinct clinical and biological behaviors. The microbial and metabolic differences in PHC and PBTC have not been studied. The pancreatic microbiota and metabolome of 15 PHC and 8 PBTC tissues and their matched nontumor tissues were characterized using 16S rRNA amplicon sequencing and untargeted metabolomics. At the genus level, Bradyrhizobium was increased while Corynebacterium and Ruminococcus were decreased in the PHC tissues (Head T) compared with the matched nontumor tissues (Head N) significantly. Shuttleworthia, Bacillus, and Bifidobacterium were significantly decreased in the PBTC tissues (Body/Tail T) compared with the matched nontumor tissues (Body/Tail N). Significantly, Ileibacterium was increased whereas Pseudoxanthomonas was decreased in Head T and Body/Tail T, and Lactobacillus was increased in Head T but decreased in Body/Tail T. A total of 102 discriminative metabolites were identified between Head T and Head N, which were scattered through linoleic acid metabolism and purine metabolism pathways. However, there were only four discriminative metabolites between Body/Tail T and Body/Tail N, which were related to glycerophospholipid metabolism and autophagy pathways. The differential metabolites in PHC and PBTC were commonly enriched in alpha-linolenic acid metabolism and choline metabolism in cancer pathways. Eubacterium decreased in Head T was positively correlated with decreased linoleic acid while negatively correlated with increased arachidyl carnitine and stearoylcarnitine. Bacillus decreased in Body/Tail T was negatively correlated with increased L-carnitine. These microbiota and metabolites deserve further investigations to reveal their roles in the pathogenesis of PHC and PBTC, providing clues for future treatments.


Subject(s)
Pancreatic Neoplasms , RNA, Ribosomal, 16S , Humans , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/metabolism , Male , Middle Aged , Female , Aged , RNA, Ribosomal, 16S/genetics , Metabolome , Microbiota , Metabolomics/methods , Pancreas/metabolism , Pancreas/microbiology , Corynebacterium/metabolism , Corynebacterium/genetics
10.
Pharmacol Res ; 204: 107197, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692467

ABSTRACT

The existing body of research underscores the critical impact of intratumoral microbiomes on the progression of pancreatic ductal adenocarcinoma (PDAC), particularly in reshaping the tumor microenvironment and influencing gemcitabine resistance. However, peritumoral tissues' microbiome, distinct from PDAC tumors, remain understudied, and Western-centric analyses overlooking potential variations in dietary-influenced microbiomes. Our study addresses this gap by 16 S rRNA sequencing of PDAC tumors and matched peritumoral tissues from Chinese Mainland patients. Our research has uncovered that the microbiome composition within tumors and paired peritumoral tissues exhibits a high degree of similarity, albeit with certain discrepancies. Notably, Exiguobacterium is found to be more abundant within the tumor tissues. Further investigations have revealed that a lower Exiguobacterium/Bacillus ratio in both the tumor and peritumoral tissues of PDAC patients is indicative of a more favorable prognosis. Further exploration utilizing an orthotopic tumor model demonstrates that the probiotic Bacillus Coagulans impedes PDAC progression, accompanied by an increased infiltration of inflammatory neutrophils in tumors. Additionally, in the subgroup with a low Exiguobacterium/Bacillus ratio, whole-exome sequencing reveals elevated missense mutations in ABL2 and MSH2. The elevated expression of ABL2 and MSH2 has been correlated with poorer prognostic outcomes in PDAC patients. Together, these insights shed light on risk factors influencing PDAC progression and unveil potential therapeutic targets, alongside probiotic intervention strategies.


Subject(s)
Disease Progression , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , China/epidemiology , Male , Female , Animals , Prognosis , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Bacillus/genetics , Bacillus/isolation & purification , Middle Aged , Aged , Tumor Microenvironment , Probiotics/therapeutic use , Mice , Microbiota , Cell Line, Tumor , Gastrointestinal Microbiome
11.
Indian J Med Microbiol ; 49: 100611, 2024.
Article in English | MEDLINE | ID: mdl-38735644

ABSTRACT

Non-O1/non-O139 Vibrio cholerae, a comparably poorly studied pathogen is culpable of sporadic but serious infections. We report a case of non O1 non O139 Vibrio cholerae septicemia in a middle aged male recently diagnosed with carcinoma pancreas. He underwent biliary tract interventional procedure for hematemesis three weeks before the presentation. Now, he presented with fever, abdominal pain, hematemesis and melena. Endoscopy revealed severe portal hypertensive gastropathy and mild hemobilia. Blood culture grew Vibrio cholerae, identified as non O1 non O139 by serogrouping. He recovered successfully with timely diagnosis, appropriate antibiotics and supportive measures.


Subject(s)
Anti-Bacterial Agents , Pancreatic Neoplasms , Sepsis , Vibrio cholerae non-O1 , Humans , Male , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/microbiology , Vibrio cholerae non-O1/isolation & purification , Vibrio cholerae non-O1/classification , Vibrio cholerae non-O1/pathogenicity , Vibrio cholerae non-O1/genetics , Sepsis/microbiology , Sepsis/diagnosis , Middle Aged , Anti-Bacterial Agents/therapeutic use , Cholera/microbiology , Cholera/diagnosis , Cholera/complications , Vibrio Infections/diagnosis , Vibrio Infections/microbiology
12.
PLoS One ; 19(4): e0302361, 2024.
Article in English | MEDLINE | ID: mdl-38687802

ABSTRACT

Growing evidence has increasingly suggested a potential linkage between the oral microbiome and various diseases, including pancreatic ductal adenocarcinoma (PDAC). However, the utilization of gene-level information derived from the oral microbiome for diagnosing PDAC remains unexplored. In this study, we sought to investigate the novel potential of leveraging genomic signatures associated with antibiotic resistance genes (ARGs) within the oral microbiome for the diagnosis of PDAC. By conducting an analysis of oral microbiome samples obtained from PDAC patients, we successfully identified specific ARGs that displayed distinct sequence abundance profiles correlated with the presence of PDAC. In the healthy group, three ARGs were found to be enriched, whereas 21 ARGs were enriched in PDAC patients. Remarkably, these ARGs from oral microbiome exhibited promising diagnostic capabilities for PDAC (AUROC = 0.79), providing a non-invasive and early detection method. Our findings not only provide novel modal data for diagnosing PDAC but also shed light on the intricate interplay between the oral microbiome and PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Microbiota , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/diagnosis , Microbiota/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/diagnosis , Female , Male , Mouth/microbiology , Middle Aged , Drug Resistance, Microbial/genetics , Aged , Genomics/methods
13.
Gut ; 73(7): 1131-1141, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38429112

ABSTRACT

OBJECTIVE: This study aims to validate the existence of a microbiome within intraductal papillary mucinous neoplasm (IPMN) that can be differentiated from the taxonomically diverse DNA background of next-generation sequencing procedures. DESIGN: We generated 16S rRNA amplicon sequencing data to analyse 338 cyst fluid samples from 190 patients and 19 negative controls, the latter collected directly from sterile syringes in the operating room. A subset of samples (n=20) and blanks (n=5) were spiked with known concentrations of bacterial cells alien to the human microbiome to infer absolute abundances of microbial traces. All cyst fluid samples were obtained intraoperatively and included IPMNs with various degrees of dysplasia as well as other cystic neoplasms. Follow-up culturing experiments were conducted to assess bacterial growth for microbiologically significant signals. RESULTS: Microbiome signatures of cyst fluid samples were inseparable from those of negative controls, with no difference in taxonomic diversity, and microbial community composition. In a patient subgroup that had recently undergone invasive procedures, a bacterial signal was evident. This outlier signal was not characterised by higher taxonomic diversity but by an increased dominance index of a gut-associated microbe, leading to lower taxonomic evenness compared with the background signal. CONCLUSION: The 'microbiome' of IPMNs and other pancreatic cystic neoplasms does not deviate from the background signature of negative controls, supporting the concept of a sterile environment. Outlier signals may appear in a small fraction of patients following recent invasive endoscopic procedures. No associations between microbial patterns and clinical or cyst parameters were apparent.


Subject(s)
Microbiota , Pancreatic Intraductal Neoplasms , Pancreatic Neoplasms , RNA, Ribosomal, 16S , Humans , Male , Female , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Aged , Middle Aged , Pancreatic Intraductal Neoplasms/microbiology , Pancreatic Intraductal Neoplasms/pathology , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Cyst Fluid/microbiology , Adenocarcinoma, Mucinous/microbiology , Adenocarcinoma, Mucinous/pathology , Aged, 80 and over , Pancreas/microbiology , Adult
14.
Biomol Biomed ; 24(4): 952-958, 2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38153528

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy, often associated with a poor prognosis for patients. One of the major challenges in managing PDAC is the difficulty in early diagnosis, owing to the limited and invasive nature of current diagnostic methods. Recent studies have identified the oral microbiome as a potential source of non-invasive biomarkers for diseases, including PDAC. In this study, we focused on leveraging the differential expression of virulence factors (VFs) encoded by the oral microbiome to create a diagnostic tool for PDAC. We observed a higher alpha diversity in VF categories among PDAC patients compared to healthy controls. We then identified a panel of VF categories that were significantly upregulated in PDAC patients, these being associated with bacterial adherence, exoenzyme production, and nutritional/metabolic processes. Moreover, Streptococcus-derived VFs were notably enriched in PDAC patients. We developed a diagnostic model using random forest analysis based on the levels of these VFs. The model's diagnostic accuracy was evaluated using receiver operating characteristic (ROC) curve analysis, with an area under the curve (AUC) of 0.88, indicating high accuracy in differentiating PDAC patients from healthy controls. Our findings suggest that VFs encoded by the oral microbiome hold potential as diagnostic tools for PDAC, offering a non-invasive approach that could significantly enhance early detection and prognosis, ultimately leading to improved patient outcomes.


Subject(s)
Carcinoma, Pancreatic Ductal , Microbiota , Pancreatic Neoplasms , Virulence Factors , Humans , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/diagnosis , Virulence Factors/metabolism , Male , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/diagnosis , Female , Middle Aged , Aged , Mouth/microbiology , ROC Curve , Case-Control Studies , Biomarkers, Tumor
15.
Article in English | WPRIM (Western Pacific) | ID: wpr-149192

ABSTRACT

Several species of Helicobacter colonize the hepatobiliary tract of animals and cause hepatobiliary diseases. The aim of this study is to investigate Helicobacter found in the biliary tract diseases of humans. Thirty-two bile samples (15 from bile duct cancer, 6 from pancreatic head cancer, and 11 from intrahepatic duct stone) were obtained by percutaneous transhepatic biliary drainage. Polymerase chain reaction analysis using Helicobacter specific urease A gene and 16S rRNA primers, bile pH measurement, and Helicobacter culture were performed. Helicobacter DNA was detected in 37.5%, and 31.3% by PCR with ureA gene, and 16S rRNA, respectively. The bile pH was not related to the presence of Helicobacter. The cultures were not successful. In conclusion, Helicobacter can be detected in the bile of patients with bile duct diseases. The possibility of pathogenesis of biliary tract diseases in humans by these organisms will be further investigated.


Subject(s)
Adult , Aged , Aged, 80 and over , Humans , Adenocarcinoma/microbiology , Bile/microbiology , Bile Duct Diseases/microbiology , Bile Duct Neoplasms/microbiology , Cholelithiasis/microbiology , DNA Primers , DNA, Bacterial , Helicobacter/isolation & purification , Helicobacter/growth & development , Helicobacter/genetics , Hydrogen-Ion Concentration , Middle Aged , Pancreatic Neoplasms/microbiology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL