Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
BMC Microbiol ; 24(1): 235, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38956452

RESUMEN

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.


Asunto(s)
Bilis , Carcinoma Ductal Pancreático , Microbiota , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Bilis/microbiología , Masculino , Femenino , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Microbiota/genética , Persona de Mediana Edad , Anciano , Disbiosis/microbiología , Supervivencia sin Progresión , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios Prospectivos , ARN Ribosómico 16S/genética
2.
Front Immunol ; 15: 1434771, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39044834

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático , Microbioma Gastrointestinal , Neoplasias Pancreáticas , Humanos , Microbioma Gastrointestinal/inmunología , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/inmunología , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/inmunología , Animales , Probióticos/uso terapéutico , Trasplante de Microbiota Fecal , Inmunoterapia/métodos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico
3.
Gan To Kagaku Ryoho ; 51(6): 603-607, 2024 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-39009514

RESUMEN

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.


Asunto(s)
Microbiota , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/inmunología , Disbiosis/microbiología , Disbiosis/inmunología , Disbiosis/terapia , Microambiente Tumoral
4.
J Med Virol ; 96(7): e29809, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39016466

RESUMEN

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.


Asunto(s)
Heces , Microbioma Gastrointestinal , Metagenómica , Neoplasias Pancreáticas , Viroma , Humanos , Neoplasias Pancreáticas/virología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/microbiología , Microbioma Gastrointestinal/genética , Metagenómica/métodos , Heces/virología , Heces/microbiología , Virus/aislamiento & purificación , Virus/genética , Virus/clasificación , Metagenoma , Bacterias/aislamiento & purificación , Bacterias/clasificación , Bacterias/genética , Persona de Mediana Edad , Masculino , Femenino , Anciano , Estudios de Casos y Controles
5.
Gut Microbes ; 16(1): 2374596, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39024520

RESUMEN

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.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Microbioma Gastrointestinal , Neoplasias Pancreáticas , Microambiente Tumoral , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/microbiología , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/uso terapéutico , Animales , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/clasificación , Medicina de Precisión , Resistencia a Antineoplásicos
6.
Gut Microbes ; 16(1): 2375483, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38972056

RESUMEN

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.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Aprendizaje Automático , Metástasis de la Neoplasia , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Masculino , Femenino , ARN Ribosómico 16S/genética , Persona de Mediana Edad , Heces/microbiología , Anciano , Metagenómica
7.
Pharmacol Res ; 204: 107197, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692467

RESUMEN

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.


Asunto(s)
Progresión de la Enfermedad , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , China/epidemiología , Masculino , Femenino , Animales , Pronóstico , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Bacillus/genética , Bacillus/aislamiento & purificación , Persona de Mediana Edad , Anciano , Microambiente Tumoral , Probióticos/uso terapéutico , Ratones , Microbiota , Línea Celular Tumoral , Microbioma Gastrointestinal
8.
Indian J Med Microbiol ; 49: 100611, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38735644

RESUMEN

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.


Asunto(s)
Antibacterianos , Neoplasias Pancreáticas , Sepsis , Vibrio cholerae no O1 , Humanos , Masculino , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/microbiología , Vibrio cholerae no O1/aislamiento & purificación , Vibrio cholerae no O1/clasificación , Vibrio cholerae no O1/patogenicidad , Vibrio cholerae no O1/genética , Sepsis/microbiología , Sepsis/diagnóstico , Persona de Mediana Edad , Antibacterianos/uso terapéutico , Cólera/microbiología , Cólera/diagnóstico , Cólera/complicaciones , Vibriosis/diagnóstico , Vibriosis/microbiología
9.
PLoS One ; 19(4): e0302361, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38687802

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático , Microbiota , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/diagnóstico , Microbiota/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/diagnóstico , Femenino , Masculino , Boca/microbiología , Persona de Mediana Edad , Farmacorresistencia Microbiana/genética , Anciano , Genómica/métodos
10.
Gut ; 73(7): 1131-1141, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38429112

RESUMEN

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.


Asunto(s)
Microbiota , Neoplasias Intraductales Pancreáticas , Neoplasias Pancreáticas , ARN Ribosómico 16S , Humanos , Masculino , Femenino , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología , Anciano , Persona de Mediana Edad , Neoplasias Intraductales Pancreáticas/microbiología , Neoplasias Intraductales Pancreáticas/patología , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/patología , Líquido Quístico/microbiología , Adenocarcinoma Mucinoso/microbiología , Adenocarcinoma Mucinoso/patología , Anciano de 80 o más Años , Páncreas/microbiología , Adulto
11.
Biochim Biophys Acta Rev Cancer ; 1878(5): 188943, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37355177

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the lethal cancers in the world and its 5-year survival rate is <10%. Due to the unique TME and dense tissue structure, its curative efficacy is far from satisfactory,the immunotherapy is even more invalid. According to the recent studies, the gut and tumor microbiota have been proved to play a key role in the development, progression and prognosis of PDAC. Based on the differences of microbiome composition observed in PDAC patients and normal pancreas, many researches have been made focusing on the latent communication between gut and intra-tumor microbiota and PDAC. In this review, we will demonstrate the potential mechanism of the oncogenic effects of GM and IM and their crucial effects on modulating the TME. Besides, we focus on their interaction with chemotherapeutic and immunotherapeutic drugs and inducing the drug resistance, thus enlightening the promising role to be used to monitor the occurrence of PDAC, accurately modulate the immune environment to promote the therapeutic efficacy and predict the prognosis.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Microambiente Tumoral , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/terapia , Humanos , Animales , Microbioma Gastrointestinal , Carcinogénesis , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/terapia , Bacterias/clasificación , Metástasis de la Neoplasia , Antineoplásicos/uso terapéutico
12.
Nature ; 615(7950): 168-174, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36813961

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is expected to be the second most deadly cancer by 2040, owing to the high incidence of metastatic disease and limited responses to treatment1,2. Less than half of all patients respond to the primary treatment for PDAC, chemotherapy3,4, and genetic alterations alone cannot explain this5. Diet is an environmental factor that can influence the response to therapies, but its role in PDAC is unclear. Here, using shotgun metagenomic sequencing and metabolomic screening, we show that the microbiota-derived tryptophan metabolite indole-3-acetic acid (3-IAA) is enriched in patients who respond to treatment. Faecal microbiota transplantation, short-term dietary manipulation of tryptophan and oral 3-IAA administration increase the efficacy of chemotherapy in humanized gnotobiotic mouse models of PDAC. Using a combination of loss- and gain-of-function experiments, we show that the efficacy of 3-IAA and chemotherapy is licensed by neutrophil-derived myeloperoxidase. Myeloperoxidase oxidizes 3-IAA, which in combination with chemotherapy induces a downregulation of the reactive oxygen species (ROS)-degrading enzymes glutathione peroxidase 3 and glutathione peroxidase 7. All of this results in the accumulation of ROS and the downregulation of autophagy in cancer cells, which compromises their metabolic fitness and, ultimately, their proliferation. In humans, we observed a significant correlation between the levels of 3-IAA and the efficacy of therapy in two independent PDAC cohorts. In summary, we identify a microbiota-derived metabolite that has clinical implications in the treatment of PDAC, and provide a motivation for considering nutritional interventions during the treatment of patients with cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Microbiota , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Carcinoma Ductal Pancreático/dietoterapia , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/microbiología , Glutatión Peroxidasa/metabolismo , Neoplasias Pancreáticas/dietoterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/microbiología , Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Triptófano/metabolismo , Triptófano/farmacología , Triptófano/uso terapéutico , Neutrófilos/enzimología , Autofagia , Metagenoma , Metabolómica , Trasplante de Microbiota Fecal , Ácidos Indolacéticos/farmacología , Ácidos Indolacéticos/uso terapéutico , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Neoplasias Pancreáticas
13.
Curr Oncol ; 29(12): 9833-9854, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36547187

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is still hampered by a dismal prognosis. A better understanding of the tumor microenvironment within the pancreas and of the factors affecting its composition is of utmost importance for developing new diagnostic and treatment tools. In this context, the complement system plays a prominent role. Not only has it been shown to shape a T cell-mediated immune response, but it also directly affects proliferation and apoptosis of the tumor cells, influencing angiogenesis, metastatic spread and therapeutic resistance. This makes complement proteins appealing not only as early biomarkers of PDAC development, but also as therapeutic targets. Fungal dysbiosis is currently the new kid on the block in tumorigenesis with cancer-associated mycobiomes extracted from several cancer types. For PDAC, colonization with the yeast Malassezia seems to promote cancer progression, already in precursor lesions. One responsible mechanism appears to be complement activation via the lectin pathway. In the present article, we review the role of the complement system in tumorigenesis, presenting observations that propose it as the missing link between fungal dysbiosis and PDAC development. We also present the results of a small pilot study supporting the crucial interplay between the complement system and Malassezia colonization in PDAC pathogenesis.


Asunto(s)
Carcinogénesis , Carcinoma Ductal Pancreático , Disbiosis , Malassezia , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/microbiología , Carcinoma Ductal Pancreático/patología , Proteínas del Sistema Complemento/metabolismo , Disbiosis/microbiología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología , Proyectos Piloto , Pronóstico , Microambiente Tumoral
14.
Adv Mater ; 34(49): e2206765, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36082582

RESUMEN

The limited drug penetration and robust bacteria-mediated drug inactivation in pancreatic cancer result in the failure of chemotherapy. To fight against these issues, a dual-cascade responsive nanoparticle (sNP@G/IR) that can sequentially trigger deep penetration, killing of intratumor bacteria, and controlled release of chemo-drug, is reported. sNP@G/IR consists of a hyaluronic acid (HA) shell and glutathione (GSH)-responsive polymer-core (NP@G/IR), that encapsulates gemcitabine (Gem) and photothermal agent (IR1048). The polymer core, as an antibiotic alternative, is tailored to exert optimal antibacterial activity and selectivity. sNP@G/IR actively homes in on the tumor due to the CD44 targeting of the HA shell, which is subsequently degraded by the hyaluronidase in the extracellular matrix. The resultant NP@G/IR in decreased size and reversed charge facilitates deep tumor penetration. After cellular endocytosis, the exposed guanidine on NP@G/IR kills intracellular bacteria through disrupting cell membranes. Intracellular GSH further triggers the controlled release of the cargo. Thus, the protected Gem eventually induces cell apoptosis. Under laser irradiation, the hyperthermia of IR1048 helps further elimination of tumors and bacteria. Moreover, sNP@G/IR activates immune response, thereby reinforcing anticancer capacity. Therefore, this dual-cascade responsive sNP@G/IR eliminates tumor-resident intracellular bacteria and augments drug delivery efficacy, providing a new avenue for improving cancer therapy.


Asunto(s)
Infecciones Bacterianas , Nanopartículas , Neoplasias Pancreáticas , Humanos , Preparaciones de Acción Retardada , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/microbiología , Polímeros , Nanopartículas/uso terapéutico , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico
15.
Int Immunol ; 34(9): 447-454, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35863313

RESUMEN

Bacterial dysbiosis is evolving as an advocate for carcinogenesis and has been associated with pancreatic cancer progression and survival outcomes. The gut and pancreas of cancer patients harbor a unique microbiome that differs significantly from that of healthy individuals. We believe that the pancreatic cancer microbiome regulates tumorigenesis by altering host cell function and modulating immune cells, skewing them toward an immunosuppressive phenotype. Moreover, altering this pathogenic microbiome may enhance the efficacy of current therapies in pancreatic cancer and improve survival outcomes. This review highlights the findings on microbial modulation across various pre-clinical and clinical studies and provides insight into the potential of targeting the microbiome for pancreatic cancer therapy.


Asunto(s)
Microbiota , Neoplasias Pancreáticas , Carcinogénesis , Disbiosis , Humanos , Páncreas/microbiología , Páncreas/patología , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
16.
Microbiome ; 10(1): 93, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35701831

RESUMEN

Pancreatic cancer is a deadly disease with limited diagnostic and treatment options. Not all populations are affected equally, as disparities exist in pancreatic cancer prevalence, treatment and outcomes. Recently, next-generation sequencing has facilitated a more comprehensive analysis of the human oral microbiome creating opportunity for its application in precision medicine. Oral microbial shifts occur in patients with pancreatic cancer, which may be appreciated years prior to their diagnosis. In addition, pathogenic bacteria common in the oral cavity have been found within pancreatic tumors. Despite these findings, much remains unknown about how or why the oral microbiome differs in patients with pancreatic cancer. As individuals develop, their oral microbiome reflects both their genotype and environmental influences. Genetics, race/ethnicity, smoking, socioeconomics and age affect the composition of the oral microbiota, which may ultimately play a role in pancreatic carcinogenesis. Multiple mechanisms have been proposed to explain the oral dysbiosis found in patients with pancreatic cancer though they have yet to be confirmed. With a better understanding of the interplay between the oral microbiome and pancreatic cancer, improved diagnostic and therapeutic approaches may be implemented to reduce healthcare disparities. Video Abstract.


Asunto(s)
Microbiota , Neoplasias Pancreáticas , Disbiosis/microbiología , Humanos , Microbiota/genética , Boca/microbiología , Neoplasias Pancreáticas/microbiología , Medicina de Precisión , Neoplasias Pancreáticas
17.
Sci Rep ; 12(1): 1783, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110595

RESUMEN

As oral or intestinal bacteria have been found in pancreatic cystic fluid and tumors, understanding bacterial migration from the duodenum into the pancreas via hepato-pancreatic duct is critical. Mathematical models of migration of aerobic bacteria from the duodenum to the pancreas with tumors were developed. Additionally, the bacterial distributions under the pH gradient and those under flow were measured in double-layer flow based microfluidic device and T-shaped cylinders. Migration of aerobic bacteria from the duodenum into pancreas is counteracted by bile and pancreatic juice flow but facilitated by pH-taxis from acidic duodenum fluid toward more favorable slightly alkaline pH in pancreatic juice. Additionally, the reduced flow velocity in cancer patients, due to compressed pancreatic duct by solid tumor, facilitates migration. Moreover, measured distribution of GFP E. coli under the pH gradient in a microfluidic device validated pH-tactic behaviors. Furthermore, Pseudomonas fluorescens in hydrochloride solution, but not in bicarbonate solution, migrated upstream against bicarbonate flow of > 20 µm/s, with an advancement at approximately 50 µm/s.


Asunto(s)
Bacterias Aerobias/fisiología , Movimiento Celular , Duodeno/microbiología , Páncreas/microbiología , Jugo Pancreático/microbiología , Neoplasias Pancreáticas/microbiología , Humanos , Concentración de Iones de Hidrógeno
18.
Sci Rep ; 12(1): 107, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997106

RESUMEN

A new approach by investigating the intra-tumoral microbiome raised great interest because they may influence the host immune response and natural history of the disease. However, previous studies on the intra-tumoral microbiome of pancreatic ductal adenocarcinoma (PDAC) were mostly based on examining the formalin-fixed paraffin-embedded tumor specimens. This study aims to investigate the feasibility of using endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) as a complementary procedure of surgical biopsy to obtain adequate fresh pancreatic cancer tissue for intra-tumoral microbial research. This was a prospective pilot study performed at a single tertiary referral center. We obtained pancreatic cancer tissue by EUS-FNB and surgical biopsy, respectively. We amplified the V3-V4 hyper-variable region of bacterial 16S ribosomal ribonucleic acid (rRNA) genes, constructed a pair-end library, and performed high-throughput sequencing. From August 2020 to November 2020, nine eligible patients with PDAC were enrolled in this study. The intra-tumoral microbiome profile was successfully generated from the PDAC cancer tissue obtained by EUS-FNB as well as by surgical biopsy. There was no significant difference in intra-tumoral alpha-diversity or bacterial taxonomic composition between tissues obtained by EUS-FNB and by surgical biopsy. EUS-FNB can collect sufficient fresh cancer tissue for microbiome analyses without complication. The intra-tumoral microbiome profile in tissues obtained by EUS-FNB had similar alpha-diversity and taxonomic profiles with those obtained by surgical biopsy. It implicated, except for surgical biopsy, EUS-FNB can be another valid and valuable tool for studying intra-tumoral microbiome in patients with resectable and unresectable PDAC.


Asunto(s)
Bacterias/crecimiento & desarrollo , Carcinoma Ductal Pancreático/microbiología , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Microbiota , Neoplasias Pancreáticas/microbiología , Ribotipificación , Microambiente Tumoral , Anciano , Bacterias/genética , Carcinoma Ductal Pancreático/patología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Proyectos Piloto , Valor Predictivo de las Pruebas , Estudios Prospectivos
19.
Br J Cancer ; 126(2): 287-296, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34718358

RESUMEN

BACKGROUND: African Americans have the highest pancreatic cancer incidence of any racial/ethnic group in the United States. The oral microbiome was associated with pancreatic cancer risk in a recent study, but no such studies have been conducted in African Americans. Poor oral health, which can be a cause or effect of microbial populations, was associated with an increased risk of pancreatic cancer in a single study of African Americans. METHODS: We prospectively investigated the oral microbiome in relation to pancreatic cancer risk among 122 African-American pancreatic cancer cases and 354 controls. DNA was extracted from oral wash samples for metagenomic shotgun sequencing. Alpha and beta diversity of the microbial profiles were calculated. Multivariable conditional logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for associations between microbes and pancreatic cancer risk. RESULTS: No associations were observed with alpha or beta diversity, and no individual microbial taxa were differentially abundant between cases and control, after accounting for multiple comparisons. Among never smokers, there were elevated ORs for known oral pathogens: Porphyromonas gingivalis (OR = 1.69, 95% CI: 0.80-3.56), Prevotella intermedia (OR = 1.40, 95% CI: 0.69-2.85), and Tannerella forsythia (OR = 1.36, 95% CI: 0.66-2.77). CONCLUSIONS: Previously reported associations between oral taxa and pancreatic cancer were not present in this African-American population overall.


Asunto(s)
Población Negra/genética , Microbiota , Boca/microbiología , Neoplasias Pancreáticas/patología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/microbiología , Estudios Prospectivos , Factores de Riesgo , Estados Unidos/epidemiología
20.
Eur Rev Med Pharmacol Sci ; 25(23): 7275-7284, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34919227

RESUMEN

OBJECTIVE: The aim of this paper was to discuss the potency of gut microbiome as a non-invasive predictive biomarker for early detection of pancreatic cancer and hepatocellular carcinoma. MATERIALS AND METHODS: We analysed the available up-to-date literature (PubMed, Embase, Google Scholar databases) regarding the link between gut microbiome and early detection of pancreatic cancer, as well as hepatocellular carcinoma. The following search linked to gut microbiome and aforementioned cancers was used: 'gut microbiome', 'gut microbiota', 'pancreatic cancer', 'pancreatic ductal adenocarcinoma', hepatocellular carcinoma', 'microbial biomarkers', 'fungal microbiota', 'mycobiota'.  The search was conducted in English. RESULTS: The association between gut microbiota imbalance and development of pancreatic cancer and hepatocellular carcinoma has been recognized during last several years. The most common type of pancreatic cancer is pancreatic ductal adenocarcinoma, whose carcinogenesis is strongly related to oral microbial dysbiosis, H. pylori infection, bactibilia, hepatotropic viruses, and intrapancreatic microbiota. It is known that gut-liver axis exists and may affect hepatocarcinogenesis. Currently, the treatment strategies of these cancers are strongly limited and there are not well-recognized screening tools to early diagnose them. The growing attention towards the use of gut microbiome as a predictive non-invasive biomarker to detect pancreatic cancer and hepatocellular carcinoma in early stage has been observed. CONCLUSIONS: To conclude, the field regarding the link between gut microbiome as a non-invasive biomarkers and early detection of pancreatic cancer and hepatocellular carcinoma exists, however, it is not well-investigated. Additionally, many of the studies were conducted with small sample sizes, whereas biomarkers are ethnicity-dependent and should be validated in wide range of populations. Nevertheless, these aspects are promising and open up new diagnostic options.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Microbioma Gastrointestinal , Neoplasias Hepáticas/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Animales , Biomarcadores de Tumor , Carcinoma Hepatocelular/microbiología , Carcinoma Hepatocelular/patología , Disbiosis/diagnóstico , Detección Precoz del Cáncer/métodos , Humanos , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/patología , Neoplasias Pancreáticas/microbiología , Neoplasias Pancreáticas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...