Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
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
2.
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
3.
Gut ; 73(7): 1131-1141, 2024 Jun 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
4.
Biochim Biophys Acta Rev Cancer ; 1878(5): 188943, 2023 09.
Article in English | MEDLINE | ID: mdl-37355177

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tumor Microenvironment , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/therapy , Humans , Animals , Gastrointestinal Microbiome , Carcinogenesis , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/therapy , Bacteria/classification , Neoplasm Metastasis , Antineoplastic Agents/therapeutic use
5.
Nature ; 615(7950): 168-174, 2023 03.
Article in English | MEDLINE | ID: mdl-36813961

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal , Microbiota , Pancreatic Neoplasms , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/diet therapy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/microbiology , Glutathione Peroxidase/metabolism , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/microbiology , Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan/therapeutic use , Neutrophils/enzymology , Autophagy , Metagenome , Metabolomics , Fecal Microbiota Transplantation , Indoleacetic Acids/pharmacology , Indoleacetic Acids/therapeutic use , Disease Models, Animal , Germ-Free Life , Pancreatic Neoplasms
6.
Curr Oncol ; 29(12): 9833-9854, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36547187

ABSTRACT

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.


Subject(s)
Carcinogenesis , Carcinoma, Pancreatic Ductal , Dysbiosis , Malassezia , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Complement System Proteins/metabolism , Dysbiosis/microbiology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Pilot Projects , Prognosis , Tumor Microenvironment
7.
Sci Rep ; 12(1): 107, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997106

ABSTRACT

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.


Subject(s)
Bacteria/growth & development , Carcinoma, Pancreatic Ductal/microbiology , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Microbiota , Pancreatic Neoplasms/microbiology , Ribotyping , Tumor Microenvironment , Aged , Bacteria/genetics , Carcinoma, Pancreatic Ductal/pathology , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Pilot Projects , Predictive Value of Tests , Prospective Studies
8.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884776

ABSTRACT

Pancreatic cancer (PC) is an aggressive disease with a high mortality and poor prognosis. The human microbiome is a key factor in many malignancies, having the ability to alter host metabolism and immune responses and participate in tumorigenesis. Gut microbes have an influence on physiological functions of the healthy pancreas and are themselves controlled by pancreatic secretions. An altered oral microbiota may colonize the pancreas and cause local inflammation by the action of its metabolites, which may lead to carcinogenesis. The mechanisms behind dysbiosis and PC development are not completely clear. Herein, we review the complex interactions between PC tumorigenesis and the microbiota, and especially the question, whether and how an altered microbiota induces oncogenomic changes, or vice versa, whether cancer mutations have an impact on microbiota composition. In addition, the role of the microbiota in drug efficacy in PC chemo- and immunotherapies is discussed. Possible future scenarios are the intentional manipulation of the gut microbiota in combination with therapy or the utilization of microbial profiles for the noninvasive screening and monitoring of PC.


Subject(s)
Carcinoma, Pancreatic Ductal/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Pancreas/microbiology , Pancreatic Neoplasms/microbiology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Transformation, Neoplastic/pathology , Humans , Mouth/microbiology , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy
9.
Int J Oncol ; 59(6)2021 Dec.
Article in English | MEDLINE | ID: mdl-34738624

ABSTRACT

A large body of evidence has revealed that the microbiome serves a role in all aspects of cancer, particularly cancer treatment. To date, studies investigating the relationship between the microbiome and systemic therapy for pancreatic ductal adenocarcinoma (PDAC) are lacking. PDAC is a high­mortality malignancy (5­year survival rate; <9% for all stages). Systemic therapy is one of the most important treatment choices for all patients; however, resistance or toxicity can affect its efficacy. Studies have supported the hypothesis that the microbiome is closely associated with the response to systemic therapy in PDAC, including the induction of drug resistance, or toxicity and therapy­related changes in microbiota composition. The present review comprehensively summarized the role of the microbiome in systemic therapy for PDAC and the associated molecular mechanisms in an attempt to provide a novel direction for the improvement of treatment response and proposed potential directions for in­depth research.


Subject(s)
Antineoplastic Agents/therapeutic use , Bacteria/drug effects , Carcinoma, Pancreatic Ductal/pathology , Microbiota , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/microbiology , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms
10.
BMC Microbiol ; 21(1): 280, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34656097

ABSTRACT

BACKGROUND: Intra-tumor microbiota have been implicated in pancreatic ductal adenocarcinoma (PDAC) development, treatment response and post-treatment survivorship. Moreover, therapeutic interventions targeting microbiota may improve the response to chemotherapy and immunotherapy, further emphasizing the critical need to understand the origins of and growth of bacteria within the pancreatic tumor microenvironment. Here, we studied the role of several clinical factors on the bacterial colonization of PDAC. RESULTS: We obtained matched tumor and normal pancreatic tissue specimens from 27 patients who had undergone surgical resection for PDAC between 2011 and 2015 from the University of Minnesota Biological Materials Procurement Network (BioNet). We found that 26 (48%) out of 54 pancreatic tissue samples harbored detectable bacterial communities using real-time PCR targeting the 16S rRNA gene. Bacterial colonization was detected significantly more frequently in samples from patients who had pancreatic head tumors, underwent Whipple procedure, or had preoperative biliary stent placement. There was also a significantly greater relative abundance of microbiota from the family Enterobacteriaceae among samples from patients who underwent biliary stent placement or neoadjuvant treatment with a combination of Gemcitabine and Paclitaxel. CONCLUSIONS: These findings suggest that biliary stent placement and neoadjuvant chemotherapy are associated with specific alterations that promote the infiltration and growth of intra-tumor bacteria in the setting of PDAC. Further studies exploring whether specific bacterial communities could contribute to increased chemoresistance will be essential for optimizing medical therapies in the future.


Subject(s)
Carcinoma, Pancreatic Ductal/microbiology , Microbiota , Neoadjuvant Therapy , Pancreatic Neoplasms/microbiology , Stents , Aged , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Enterobacteriaceae/isolation & purification , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Paclitaxel/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Stents/adverse effects , Tumor Microenvironment , Gemcitabine
11.
Cancer Med ; 10(15): 5041-5050, 2021 08.
Article in English | MEDLINE | ID: mdl-34250759

ABSTRACT

Recent studies defined a potentially important role of the microbiome in modulating pancreatic ductal adenocarcinoma (PDAC) and responses to therapies. We hypothesized that antibiotic usage may predict outcomes in patients with PDAC. We retrospectively analyzed clinical data of patients with resectable or metastatic PDAC seen at MD Anderson Cancer from 2003 to 2017. Demographic, chemotherapy regimen and antibiotic use, duration, type, and reason for indication were recorded. A total of 580 patients with PDAC were studied, 342 resected and 238 metastatic patients, selected retrospectively from our database. Antibiotic use, for longer than 48 hrs, was detected in 209 resected patients (61%) and 195 metastatic ones (62%). On resectable patients, we did not find differences in overall survival (OS) or progression-free survival (PFS), based on antibiotic intake. However, in the metastatic cohort, antibiotic consumption was associated with a significantly longer OS (13.3 months vs. 9.0 months, HR 0.48, 95% CI 0.34-0.7, p = 0.0001) and PFS (4.4 months vs. 2 months, HR 0.48, 95% CI 0.34-0.68, p = <0.0001). In multivariate analysis, the impact of ATB remained significant for PFS (HR 0.59, p = 0.005) and borderline statistically significant for OS (HR 0.69, p = 0.06). When we analyzed by chemotherapy regimen, we found that patients who received gemcitabine-based chemotherapy as first-line therapy (n = 118) had significantly prolonged OS (HR 0.4, p 0.0013) and PFS (HR 0.55, p 0.02) if they received antibiotics, while those receiving 5FU-based chemotherapy (n = 98) had only prolonged PFS (HR 0.54, p = 0.03). Antibiotics-associated modulation of the microbiome is associated with better outcomes in patients with metastatic PDAC.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Infections , Carcinoma, Pancreatic Ductal/therapy , Gastrointestinal Microbiome/drug effects , Pancreatic Neoplasms/therapy , Progression-Free Survival , Adult , Aged , Aged, 80 and over , Bacterial Infections/drug therapy , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Epidemiologic Methods , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Multivariate Analysis , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Retrospective Studies , Time Factors , Treatment Outcome , Gemcitabine
12.
Pancreas ; 50(2): 119-129, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33565788

ABSTRACT

ABSTRACT: Microorganisms can help maintain homeostasis in humans by providing nutrition, maintaining hormone balance, and regulating inflammatory responses. In the case of imbalances, these microbes can cause various diseases, even malignancy. Pancreatic cancer (PC) is characterized by high tumor invasiveness, distant metastasis, and insensitivity to traditional chemotherapeutic drugs, and it is confirmed that PC is closely related to microorganisms. Recently, most studies based on clinical samples or case reports discussed the positive or negative relationships between microorganisms and PC. However, the specific mechanisms are blurry, especially the involved immunological pathways, and the roles of beneficial flora have usually been ignored. We reviewed studies published through September 2020 as identified using PubMed, MEDLINE, and Web of Science. We mainly introduced the traits of oral, gastrointestinal, and intratumoral microbes in PC and summarized the roles of these microbes in tumorigenesis and tumoral development through immunological pathways, in addition to illustrating the relationships between metabolic diseases with PC by microorganism. In addition, we identified microorganisms as biomarkers for early diagnosis and immunotherapy. This review will be significant for greater understanding the effect of microorganisms in PC and provide more meaningful guidance for future clinical applications.


Subject(s)
Bacteria/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/microbiology , Gastrointestinal Microbiome , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/microbiology , Tumor Microenvironment/immunology , Animals , Bacteria/pathogenicity , Carcinoma, Pancreatic Ductal/therapy , Dysbiosis , Host-Pathogen Interactions , Humans , Immunotherapy , Pancreatic Neoplasms/therapy
13.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188484, 2021 01.
Article in English | MEDLINE | ID: mdl-33246025

ABSTRACT

Pancreatic ductal adenocarcinoma is one of the most lethal malignancies and is known for its high resistance and low response to treatment. Cancer treatments can reshape the microbiota and in turn, the microbiota influences the therapeutic efficacy by regulating immune response and metabolism. This crosstalk is bidirectional, heterogeneous, and dynamic. In this review, we elaborated on the interactions between the microbiota and therapeutic resistance in pancreatic ductal adenocarcinoma. Regulating the microbiota in pancreatic tumor microenvironment may not only generate direct anti-cancer but also synergistic effects with other treatments, providing new directions in cancer therapy.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/genetics , Gastrointestinal Microbiome/genetics , Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/microbiology , Drug Resistance, Neoplasm/immunology , Gastrointestinal Microbiome/immunology , Humans , Immunity/drug effects , Immunity/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Sci Rep ; 10(1): 20319, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230218

ABSTRACT

Pancreatic cancer is one of the most lethal and chemo-resistant cancers worldwide. Growing evidence supports the theory that the gut microbiota plays an essential role in modulating the host response to anti-cancer therapy. The present study aimed to explore the effect of probiotics as an adjuvant during chemotherapy for pancreatic cancer. An LSL-KrasG12D/--Pdx-1-Cre mouse model of pancreatic ductal adenocarcinoma (PDAC) was created to study the effects of using four-week multi-strain probiotics (Lactobacillus paracasei GMNL-133 and Lactobacillus reuteri GMNL-89) as an adjuvant therapy for controlling cancer progression. At 12 weeks of age, pancreatitis was induced in the mice by two intraperitoneal injection with caerulein (25 µg/kg 2 days apart). Over the next 4 weeks the mice were treated with intraperitoneal injections of gemcitabine in combination with the oral administration of probiotics. The pancreas was then harvested for analysis. Following caerulein treatment, the pancreases of the LSL-KrasG12D/--Pdx-1-Cre transgenic mice exhibited more extensive pancreatic intraepithelial neoplasia (PanIN) formation. Combined treatment with gemcitabine and probiotics revealed a lower grade of PanIN formation and a decrease in the expression of vimentin and Ki-67. Mice that received gemcitabine in combination with probiotics had lower aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Notably, the use of high-dose probiotics alone without gemcitabine also had an inhibitory effect on PanIN changes and serum liver enzyme elevation. These findings suggest that probiotics are able to make standard chemotherapy more effective and could help improve the patient's tolerance of chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Pancreatic Ductal/diet therapy , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Lactobacillus , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Probiotics/administration & dosage , Administration, Oral , Animals , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/microbiology , Ceruletide/adverse effects , Deoxycytidine/administration & dosage , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/microbiology , Treatment Outcome , Gemcitabine
15.
Cell ; 181(2): 424-441.e21, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32234521

ABSTRACT

KRAS mutant pancreatic ductal adenocarcinoma (PDAC) is characterized by a desmoplastic response that promotes hypovascularity, immunosuppression, and resistance to chemo- and immunotherapies. We show that a combination of MEK and CDK4/6 inhibitors that target KRAS-directed oncogenic signaling can suppress PDAC proliferation through induction of retinoblastoma (RB) protein-mediated senescence. In preclinical mouse models of PDAC, this senescence-inducing therapy produces a senescence-associated secretory phenotype (SASP) that includes pro-angiogenic factors that promote tumor vascularization, which in turn enhances drug delivery and efficacy of cytotoxic gemcitabine chemotherapy. In addition, SASP-mediated endothelial cell activation stimulates the accumulation of CD8+ T cells into otherwise immunologically "cold" tumors, sensitizing tumors to PD-1 checkpoint blockade. Therefore, in PDAC models, therapy-induced senescence can establish emergent susceptibilities to otherwise ineffective chemo- and immunotherapies through SASP-dependent effects on the tumor vasculature and immune system.


Subject(s)
Aging/physiology , Carcinoma, Pancreatic Ductal/pathology , Vascular Remodeling/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/microbiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Humans , Immunotherapy/methods , MAP Kinase Signaling System/physiology , Mice , Pancreatic Neoplasms/pathology , Retinoblastoma Protein/immunology , Signal Transduction/genetics , Tumor Microenvironment , Vascular Remodeling/genetics
16.
Carcinogenesis ; 41(5): 561-570, 2020 07 10.
Article in English | MEDLINE | ID: mdl-31369062

ABSTRACT

The lack of tools for early detection of pancreatic ductal adenocarcinoma (PDAC) is directly correlated with the abysmal survival rates in patients. In addition to several potential detection tools under active investigation, we tested the gut microbiome and its metabolic complement as one of the earliest detection tools that could be useful in patients at high risk for PDAC. We used a combination of 16s rRNA pyrosequencing and whole-genome sequencing of gut fecal microbiota in a genetically engineered PDAC murine model (KRASG12DTP53R172HPdxCre or KPC). Metabolic reconstruction of microbiome was done using the HUMAnN2 pipeline. Serum polyamine levels were measured from murine and patient samples using chromogenic assay. Our results showed a Proteobacterial and Firmicutes dominance in gut microbiota in early stages of PDAC development. Upon in silico reconstruction of active metabolic pathways within the altered microbial flora, polyamine and nucleotide biosynthetic pathways were significantly elevated. These metabolic products are known to be actively assimilated by the host and eventually utilized by rapidly dividing cells for proliferation validating their importance in the context of tumorigenesis. In KPC mice, as well as PDAC patients, we show significantly elevated serum polyamine concentrations. Therefore, at the early stages of tumorigenesis, there is a strong correlation between microbial changes and release of metabolites that foster host tumorigenesis, thereby fulfilling the 'vicious cycle hypothesis' of the role of microbiome in health and disease states. Our results provide a potential, precise, noninvasive tool for early detection of PDAC, which may result in improved outcomes.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnosis , Dysbiosis/complications , Early Detection of Cancer , Gastrointestinal Microbiome , Pancreatic Neoplasms/diagnosis , Polyamines/metabolism , Animals , Carcinogenesis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Dysbiosis/microbiology , Feces/microbiology , Female , Humans , Male , Mice , Mice, Transgenic , Mutation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
17.
Nat Rev Gastroenterol Hepatol ; 17(1): 53-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31811279

ABSTRACT

Diseases intrinsic to the pancreas such as pancreatitis, pancreatic cancer and type 1 diabetes mellitus impart substantial health and financial burdens on society but identification of novel mechanisms contributing to these pathologies are slow to emerge. A novel area of research suggests that pancreatic-specific disorders might be modulated by the gut microbiota, either through a local (direct pancreatic influence) or in a remote (nonpancreatic) fashion. In this Perspectives, we examine literature implicating microorganisms in diseases of the pancreas, specifically pancreatitis, type 1 diabetes mellitus and pancreatic ductal adenocarcinoma. We also discuss evidence of an inherent pancreatic microbiota and the influence of the intestinal microbiota as it relates to disease association and development. In doing so, we address pitfalls in the current literature and areas of investigation that are needed to advance a developing field of research that has clinical potential to reduce the societal burden of pancreatic diseases.


Subject(s)
Carcinoma, Pancreatic Ductal/microbiology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/physiology , Pancreas/microbiology , Pancreatic Neoplasms/microbiology , Pancreatitis/microbiology , Bacterial Translocation , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Humans , Microbiota , Pancreas/physiology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
18.
Trends Cancer ; 5(11): 670-676, 2019 11.
Article in English | MEDLINE | ID: mdl-31735286

ABSTRACT

Late-stage pancreatic cancer harbors a fibrotic and immune-excluded tumor microenvironment that impedes immunotherapy success. A key to unlocking pancreatic cancer immunotherapy may be treating early-stage pancreatic cancer, when peripancreatic inflammation promoted by the microbiome potentiates oncogenic signaling and suppresses innate and adaptive immunity. Hence, understanding the role of microbiota in pancreatic cancer initiation, progression, and immunosuppression is crucial. We propose that not only are microbiota targets for immunomodulation in this disease, but also that microbiome profiling has a potential role in pancreatic cancer screening. Furthermore, combining microbiome profiling with liquid and tissue biopsy may validate the early pancreatic cancer treatment approach of microbiome modulation and immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Gastrointestinal Microbiome/physiology , Immunotherapy/methods , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/therapy , Adaptive Immunity , Carcinoma, Pancreatic Ductal/microbiology , Humans , Immunomodulation , Pancreatic Neoplasms/pathology , Tumor Microenvironment/immunology
19.
Nature ; 574(7777): 264-267, 2019 10.
Article in English | MEDLINE | ID: mdl-31578522

ABSTRACT

Bacterial dysbiosis accompanies carcinogenesis in malignancies such as colon and liver cancer, and has recently been implicated in the pathogenesis of pancreatic ductal adenocarcinoma (PDA)1. However, the mycobiome has not been clearly implicated in tumorigenesis. Here we show that fungi migrate from the gut lumen to the pancreas, and that this is implicated in the pathogenesis of PDA. PDA tumours in humans and mouse models of this cancer displayed an increase in fungi of about 3,000-fold compared to normal pancreatic tissue. The composition of the mycobiome of PDA tumours was distinct from that of the gut or normal pancreas on the basis of alpha- and beta-diversity indices. Specifically, the fungal community that infiltrated PDA tumours was markedly enriched for Malassezia spp. in both mice and humans. Ablation of the mycobiome was protective against tumour growth in slowly progressive and invasive models of PDA, and repopulation with a Malassezia species-but not species in the genera Candida, Saccharomyces or Aspergillus-accelerated oncogenesis. We also discovered that ligation of mannose-binding lectin (MBL), which binds to glycans of the fungal wall to activate the complement cascade, was required for oncogenic progression, whereas deletion of MBL or C3 in the extratumoral compartment-or knockdown of C3aR in tumour cells-were both protective against tumour growth. In addition, reprogramming of the mycobiome did not alter the progression of PDA in Mbl- (also known as Mbl2) or C3-deficient mice. Collectively, our work shows that pathogenic fungi promote PDA by driving the complement cascade through the activation of MBL.


Subject(s)
Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Carcinogenesis , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Gastrointestinal Microbiome/immunology , Mannose-Binding Lectin/immunology , Mycobiome/immunology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Case-Control Studies , Complement Activation , Complement C3/deficiency , Complement C3/immunology , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred C57BL
20.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G763-G772, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31545922

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC)'s growing incidence has been linked to the rise in obesity and type 2 diabetes mellitus. In previous work, we have shown that metformin can prevent the increased incidence of PDAC in a KrasG12D mouse model subjected to a diet high in fat and calories (HFCD). One potential way that metformin can affect the host is through alterations in the gut microbiome. Therefore, we investigated microbial associations with PDAC development and metformin use in the same mouse model. Lox-Stop-Lox Kras G12D/+ (LSL-Kras G12D/+); p48-Cre (KC) mice were given control diet, HFCD, or HFCD with 5 mg/mL metformin in drinking water for 3 mo. At the end of the 3 mo, 16S rRNA sequencing was performed to characterize microbiome composition of duodenal mucosal, duodenal luminal, and cecal luminal samples. KC mice on an HFCD demonstrated depletion of intact acini and formation of advanced pancreatic intraepithelial neoplasia. This effect was completely abrogated by metformin treatment. HFCD was associated with significant changes in microbial composition and diversity in the duodenal mucosa and lumen, much of which was prevented by metformin. In particular, Clostridium sensu stricto was negatively correlated with percent intact acini and seemed to be inhibited by the addition of metformin while on an HFCD. Administration of metformin eliminated PDAC formation in KC mice. This change was associated with significant microbial changes in both the mucosal and luminal microbiome of the duodenum. This suggests that the microbiome may be a potential mediator of the chemopreventive effects of metformin.NEW & NOTEWORTHY Pancreatic ductal adenocarcinoma (PDAC)'s growing incidence has been linked to the rise in obesity and type 2 diabetes mellitus. Administration of metformin eliminated PDAC formation in KC mice with diet-induced obesity. This change was associated with significant microbial changes in both the mucosal and luminal microbiome of the duodenum. This suggests that the microbiome may be a potential mediator of the chemopreventive effects of metformin.


Subject(s)
Carcinoma, Pancreatic Ductal , Duodenum , Gastrointestinal Microbiome/drug effects , Metformin/pharmacology , Animals , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Disease Models, Animal , Duodenum/microbiology , Duodenum/pathology , Hypoglycemic Agents/pharmacology , Mice , Obesity/etiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...