Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer J ; 29(2): 49-56, 2023.
Article in English | MEDLINE | ID: mdl-36957973

ABSTRACT

ABSTRACT: Changes in the gut microbiome have been increasingly shown to accompany oncogenesis across various tumors. Similarly, microbial dysbiosis was found to be associated with pancreatic cancer progression and survival outcomes, expanding the field of tumor microenvironment research in pancreatic cancer. Mechanistic studies in pancreatic cancer models implicate components of the gut and pancreatic cancer microbiome in regulating tumorigenesis by altering cancer cell signaling, modulating immune function, and influencing the efficacy of current therapies in pancreatic cancer. This review discusses the outcomes of microbial modulation across various preclinical and clinical studies and highlights ongoing trials targeting the microbiome for pancreatic cancer therapy.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/therapy , Carcinogenesis , Cell Transformation, Neoplastic , Tumor Microenvironment , Pancreatic Neoplasms
2.
Int Immunol ; 34(9): 447-454, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35863313

ABSTRACT

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.


Subject(s)
Microbiota , Pancreatic Neoplasms , Carcinogenesis , Dysbiosis , Humans , Pancreas/microbiology , Pancreas/pathology , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
6.
Cancer Discov ; 8(4): 403-416, 2018 04.
Article in English | MEDLINE | ID: mdl-29567829

ABSTRACT

We found that the cancerous pancreas harbors a markedly more abundant microbiome compared with normal pancreas in both mice and humans, and select bacteria are differentially increased in the tumorous pancreas compared with gut. Ablation of the microbiome protects against preinvasive and invasive pancreatic ductal adenocarcinoma (PDA), whereas transfer of bacteria from PDA-bearing hosts, but not controls, reverses tumor protection. Bacterial ablation was associated with immunogenic reprogramming of the PDA tumor microenvironment, including a reduction in myeloid-derived suppressor cells and an increase in M1 macrophage differentiation, promoting TH1 differentiation of CD4+ T cells and CD8+ T-cell activation. Bacterial ablation also enabled efficacy for checkpoint-targeted immunotherapy by upregulating PD-1 expression. Mechanistically, the PDA microbiome generated a tolerogenic immune program by differentially activating select Toll-like receptors in monocytic cells. These data suggest that endogenous microbiota promote the crippling immune-suppression characteristic of PDA and that the microbiome has potential as a therapeutic target in the modulation of disease progression.Significance: We found that a distinct and abundant microbiome drives suppressive monocytic cellular differentiation in pancreatic cancer via selective Toll-like receptor ligation leading to T-cell anergy. Targeting the microbiome protects against oncogenesis, reverses intratumoral immune tolerance, and enables efficacy for checkpoint-based immunotherapy. These data have implications for understanding immune suppression in pancreatic cancer and its reversal in the clinic. Cancer Discov; 8(4); 403-16. ©2018 AACR.See related commentary by Riquelme et al., p. 386This article is highlighted in the In This Issue feature, p. 371.


Subject(s)
Carcinogenesis , Microbiota , Monocytes/physiology , Pancreatic Neoplasms/microbiology , Toll-Like Receptors/metabolism , Animals , Bacteria , Cell Differentiation , Female , Humans , Male , Mice , Monocytes/immunology , Monocytes/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Signal Transduction
7.
J Exp Med ; 214(6): 1711-1724, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28442553

ABSTRACT

The tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance, which enables disease to progress unabated by adaptive immunity. However, the drivers of this tolerogenic program are incompletely defined. In this study, we found that NLRP3 promotes expansion of immune-suppressive macrophages in PDA. NLRP3 signaling in macrophages drives the differentiation of CD4+ T cells into tumor-promoting T helper type 2 cell (Th2 cell), Th17 cell, and regulatory T cell populations while suppressing Th1 cell polarization and cytotoxic CD8+ T cell activation. The suppressive effects of NLRP3 signaling were IL-10 dependent. Pharmacological inhibition or deletion of NLRP3, ASC (apoptosis-associated speck-like protein containing a CARD complex), or caspase-1 protected against PDA and was associated with immunogenic reprogramming of innate and adaptive immunity within the TME. Similarly, transfer of PDA-entrained macrophages or T cells from NLRP3-/- hosts was protective. These data suggest that targeting NLRP3 holds the promise for the immunotherapy of PDA.


Subject(s)
Adaptive Immunity , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Caspase 1/deficiency , Caspase 1/metabolism , Cell Differentiation , Cell Proliferation , Cellular Reprogramming , Gene Deletion , Humans , Immunosuppression Therapy , Mice, Inbred C57BL , Nod2 Signaling Adaptor Protein/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , T-Lymphocytes/immunology , Tumor Microenvironment , Pancreatic Neoplasms
8.
Nat Med ; 23(5): 556-567, 2017 May.
Article in English | MEDLINE | ID: mdl-28394331

ABSTRACT

The progression of pancreatic oncogenesis requires immune-suppressive inflammation in cooperation with oncogenic mutations. However, the drivers of intratumoral immune tolerance are uncertain. Dectin 1 is an innate immune receptor crucial for anti-fungal immunity, but its role in sterile inflammation and oncogenesis has not been well defined. Furthermore, non-pathogen-derived ligands for dectin 1 have not been characterized. We found that dectin 1 is highly expressed on macrophages in pancreatic ductal adenocarcinoma (PDA). Dectin 1 ligation accelerated the progression of PDA in mice, whereas deletion of Clec7a-the gene encoding dectin 1-or blockade of dectin 1 downstream signaling was protective. We found that dectin 1 can ligate the lectin galectin 9 in mouse and human PDA, which results in tolerogenic macrophage programming and adaptive immune suppression. Upon disruption of the dectin 1-galectin 9 axis, CD4+ and CD8+ T cells, which are dispensable for PDA progression in hosts with an intact signaling axis, become reprogrammed into indispensable mediators of anti-tumor immunity. These data suggest that targeting dectin 1 signaling is an attractive strategy for developing an immunotherapy for PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Galectins/metabolism , Lectins, C-Type/genetics , Pancreatic Neoplasms/genetics , Tumor Escape/genetics , Animals , Blotting, Western , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Gene Knockdown Techniques , Humans , Immune Tolerance/genetics , Immunohistochemistry , Immunoprecipitation , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Pancreatic Ducts/cytology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism , Tumor Escape/immunology
10.
J Immunol ; 197(7): 2816-27, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27559045

ABSTRACT

Con A hepatitis is regarded as a T cell-mediated model of acute liver injury. Mincle is a C-type lectin receptor that is critical in the immune response to mycobacteria and fungi but does not have a well-defined role in preclinical models of non-pathogen-mediated inflammation. Because Mincle can ligate the cell death ligand SAP130, we postulated that Mincle signaling drives intrahepatic inflammation and liver injury in Con A hepatitis. Acute liver injury was assessed in the murine Con A hepatitis model using C57BL/6, Mincle(-/-), and Dectin-1(-/-) mice. The role of C/EBPß and hypoxia-inducible factor-1α (HIF-1α) signaling was assessed using selective inhibitors. We found that Mincle was highly expressed in hepatic innate inflammatory cells and endothelial cells in both mice and humans. Furthermore, sterile Mincle ligands and Mincle signaling intermediates were increased in the murine liver in Con A hepatitis. Most significantly, Mincle deletion or blockade protected against Con A hepatitis, whereas Mincle ligation exacerbated disease. Bone marrow chimeric and adoptive transfer experiments suggested that Mincle signaling in infiltrating myeloid cells dictates disease phenotype. Conversely, signaling via other C-type lectin receptors did not alter disease course. Mechanistically, we found that Mincle blockade decreased the NF-κß-related signaling intermediates C/EBPß and HIF-1α, both of which are necessary in macrophage-mediated inflammatory responses. Accordingly, Mincle deletion lowered production of nitrites in Con A hepatitis and inhibition of both C/EBPß and HIF-1α reduced the severity of liver disease. Our work implicates a novel innate immune driver of Con A hepatitis and, more broadly, suggests a potential role for Mincle in diseases governed by sterile inflammation.


Subject(s)
Concanavalin A/immunology , Hepatitis/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , Signal Transduction/immunology , Animals , Disease Models, Animal , Hepatitis/metabolism , Humans , Inflammation/immunology , Lectins, C-Type/deficiency , Leukocytes, Mononuclear , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitrites/metabolism
11.
Cell ; 166(6): 1485-1499.e15, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27569912

ABSTRACT

Inflammation is paramount in pancreatic oncogenesis. We identified a uniquely activated γδT cell population, which constituted ∼40% of tumor-infiltrating T cells in human pancreatic ductal adenocarcinoma (PDA). Recruitment and activation of γδT cells was contingent on diverse chemokine signals. Deletion, depletion, or blockade of γδT cell recruitment was protective against PDA and resulted in increased infiltration, activation, and Th1 polarization of αßT cells. Although αßT cells were dispensable to outcome in PDA, they became indispensable mediators of tumor protection upon γδT cell ablation. PDA-infiltrating γδT cells expressed high levels of exhaustion ligands and thereby negated adaptive anti-tumor immunity. Blockade of PD-L1 in γδT cells enhanced CD4(+) and CD8(+) T cell infiltration and immunogenicity and induced tumor protection suggesting that γδT cells are critical sources of immune-suppressive checkpoint ligands in PDA. We describe γδT cells as central regulators of effector T cell activation in cancer via novel cross-talk.


Subject(s)
Carcinogenesis/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/physiopathology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Carcinogenesis/pathology , Cells, Cultured , Chemokines/immunology , Epithelial Cells/physiology , Female , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Tumor Microenvironment/immunology
12.
Nature ; 532(7598): 245-9, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27049944

ABSTRACT

Neoplastic pancreatic epithelial cells are believed to die through caspase 8-dependent apoptotic cell death, and chemotherapy is thought to promote tumour apoptosis. Conversely, cancer cells often disrupt apoptosis to survive. Another type of programmed cell death is necroptosis (programmed necrosis), but its role in pancreatic ductal adenocarcinoma (PDA) is unclear. There are many potential inducers of necroptosis in PDA, including ligation of tumour necrosis factor receptor 1 (TNFR1), CD95, TNF-related apoptosis-inducing ligand (TRAIL) receptors, Toll-like receptors, reactive oxygen species, and chemotherapeutic drugs. Here we report that the principal components of the necrosome, receptor-interacting protein (RIP)1 and RIP3, are highly expressed in PDA and are further upregulated by the chemotherapy drug gemcitabine. Blockade of the necrosome in vitro promoted cancer cell proliferation and induced an aggressive oncogenic phenotype. By contrast, in vivo deletion of RIP3 or inhibition of RIP1 protected against oncogenic progression in mice and was associated with the development of a highly immunogenic myeloid and T cell infiltrate. The immune-suppressive tumour microenvironment associated with intact RIP1/RIP3 signalling depended in part on necroptosis-induced expression of the chemokine attractant CXCL1, and CXCL1 blockade protected against PDA. Moreover, cytoplasmic SAP130 (a subunit of the histone deacetylase complex) was expressed in PDA in a RIP1/RIP3-dependent manner, and Mincle--its cognate receptor--was upregulated in tumour-infiltrating myeloid cells. Ligation of Mincle by SAP130 promoted oncogenesis, whereas deletion of Mincle protected against oncogenesis and phenocopied the immunogenic reprogramming of the tumour microenvironment that was induced by RIP3 deletion. Cellular depletion suggested that whereas inhibitory macrophages promote tumorigenesis in PDA, they lose their immune-suppressive effects when RIP3 or Mincle is deleted. Accordingly, T cells, which are not protective against PDA progression in mice with intact RIP3 or Mincle signalling, are reprogrammed into indispensable mediators of anti-tumour immunity in the absence of RIP3 or Mincle. Our work describes parallel networks of necroptosis-induced CXCL1 and Mincle signalling that promote macrophage-induced adaptive immune suppression and thereby enable PDA progression.


Subject(s)
Carcinogenesis , Chemokine CXCL1/metabolism , Immune Tolerance , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Necrosis , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL1/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Progression , Female , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lectins, C-Type/immunology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Up-Regulation , Gemcitabine
13.
Gastroenterology ; 150(7): 1659-1672.e5, 2016 06.
Article in English | MEDLINE | ID: mdl-26946344

ABSTRACT

BACKGROUND & AIMS: The role of radiation therapy in the treatment of patients with pancreatic ductal adenocarcinoma (PDA) is controversial. Randomized controlled trials investigating the efficacy of radiation therapy in patients with locally advanced unresectable PDA have reported mixed results, with effects ranging from modest benefit to worse outcomes compared with control therapies. We investigated whether radiation causes inflammatory cells to acquire an immune-suppressive phenotype that limits the therapeutic effects of radiation on invasive PDAs and accelerates progression of preinvasive foci. METHODS: We investigated the effects of radiation therapy in p48(Cre);LSL-Kras(G12D) (KC) and p48(Cre);LSLKras(G12D);LSL-Trp53(R172H) (KPC) mice, as well as in C57BL/6 mice with orthotopic tumors grown from FC1242 cells derived from KPC mice. Some mice were given neutralizing antibodies against macrophage colony-stimulating factor 1 (CSF1 or MCSF) or F4/80. Pancreata were exposed to doses of radiation ranging from 2 to 12 Gy and analyzed by flow cytometry. RESULTS: Pancreata of KC mice exposed to radiation had a higher frequency of advanced pancreatic intraepithelial lesions and more foci of invasive cancer than pancreata of unexposed mice (controls); radiation reduced survival time by more than 6 months. A greater proportion of macrophages from radiation treated invasive and preinvasive pancreatic tumors had an immune-suppressive, M2-like phenotype compared with control mice. Pancreata from mice exposed to radiation had fewer CD8(+) T cells than controls, and greater numbers of CD4(+) T cells of T-helper 2 and T-regulatory cell phenotypes. Adoptive transfer of T cells from irradiated PDA to tumors of control mice accelerated tumor growth. Radiation induced production of MCSF by PDA cells. A neutralizing antibody against MCSF prevented radiation from altering the phenotype of macrophages in tumors, increasing the anti-tumor T-cell response and slowing tumor growth. CONCLUSIONS: Radiation treatment causes macrophages murine PDA to acquire an immune-suppressive phenotype and disabled T-cell-mediated anti-tumor responses. MCSF blockade negates this effect, allowing radiation to have increased efficacy in slowing tumor growth.


Subject(s)
Adenoma/immunology , Carcinoma, Pancreatic Ductal/immunology , Macrophages/radiation effects , Pancreatic Neoplasms/immunology , T-Lymphocytes/immunology , Adenoma/radiotherapy , Animals , Carcinoma, Pancreatic Ductal/radiotherapy , Disease Models, Animal , Mice , Mice, Inbred C57BL , Pancreas/immunology , Pancreas/radiation effects , Pancreatic Neoplasms/radiotherapy , T-Lymphocytes/radiation effects
14.
J Leukoc Biol ; 100(1): 185-94, 2016 07.
Article in English | MEDLINE | ID: mdl-26747838

ABSTRACT

Regulation of Toll-like receptor responses is critical for limiting tissue injury and autoimmunity in both sepsis and sterile inflammation. We found that Mincle, a C-type lectin receptor, regulates proinflammatory Toll-like receptor 4 signaling. Specifically, Mincle ligation diminishes Toll-like receptor 4-mediated inflammation, whereas Mincle deletion or knockdown results in marked hyperresponsiveness to lipopolysaccharide in vitro, as well as overwhelming lipopolysaccharide-mediated inflammation in vivo. Mechanistically, Mincle deletion does not up-regulate Toll-like receptor 4 expression or reduce interleukin 10 production after Toll-like receptor 4 ligation; however, Mincle deletion decreases production of the p38 mitogen-activated protein kinase-dependent inhibitory intermediate suppressor of cytokine signaling 1, A20, and ABIN3 and increases expression of the Toll-like receptor 4 coreceptor CD14. Blockade of CD14 mitigates the increased sensitivity of Mincle(-/-) leukocytes to Toll-like receptor 4 ligation. Collectively, we describe a major role for Mincle in suppressing Toll-like receptor 4 responses and implicate its importance in nonmycobacterial models of inflammation.


Subject(s)
Inflammation/etiology , Lectins, C-Type/deficiency , Membrane Proteins/deficiency , Spleen/immunology , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Inflammation/metabolism , Inflammation/pathology , Lectins, C-Type/genetics , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Toll-Like Receptor 4/antagonists & inhibitors
15.
Cell Rep ; 13(9): 1909-1921, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655905

ABSTRACT

Dectin-1 is a C-type lectin receptor critical in anti-fungal immunity, but Dectin-1 has not been linked to regulation of sterile inflammation or oncogenesis. We found that Dectin-1 expression is upregulated in hepatic fibrosis and liver cancer. However, Dectin-1 deletion exacerbates liver fibro-inflammatory disease and accelerates hepatocarcinogenesis. Mechanistically, we found that Dectin-1 protects against chronic liver disease by suppressing TLR4 signaling in hepatic inflammatory and stellate cells. Accordingly, Dectin-1(-/-) mice exhibited augmented cytokine production and reduced survival in lipopolysaccharide (LPS)-mediated sepsis, whereas Dectin-1 activation was protective. We showed that Dectin-1 inhibits TLR4 signaling by mitigating TLR4 and CD14 expression, which are regulated by Dectin-1-dependent macrophage colony stimulating factor (M-CSF) expression. Our study suggests that Dectin-1 is an attractive target for experimental therapeutics in hepatic fibrosis and neoplastic transformation. More broadly, our work deciphers critical cross-talk between pattern recognition receptors and implicates a role for Dectin-1 in suppression of sterile inflammation, inflammation-induced oncogenesis, and LPS-mediated sepsis.


Subject(s)
Lectins, C-Type/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Toll-Like Receptor 4/metabolism , Animals , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Chemokine CCL2/blood , Cytokines/metabolism , Diethylnitrosamine/toxicity , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inflammation , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Sepsis/etiology , Signal Transduction/drug effects , Thioacetamide/toxicity , Toll-Like Receptor 4/antagonists & inhibitors , Up-Regulation/drug effects
16.
J Exp Med ; 212(12): 2077-94, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26481685

ABSTRACT

Modulation of Toll-like receptor (TLR) signaling can have protective or protumorigenic effects on oncogenesis depending on the cancer subtype and on specific inflammatory elements within the tumor milieu. We found that TLR9 is widely expressed early during the course of pancreatic transformation and that TLR9 ligands are ubiquitous within the tumor microenvironment. TLR9 ligation markedly accelerates oncogenesis, whereas TLR9 deletion is protective. We show that TLR9 activation has distinct effects on the epithelial, inflammatory, and fibrogenic cellular subsets in pancreatic carcinoma and plays a central role in cross talk between these compartments. Specifically, TLR9 activation can induce proinflammatory signaling in transformed epithelial cells, but does not elicit oncogene expression or cancer cell proliferation. Conversely, TLR9 ligation induces pancreatic stellate cells (PSCs) to become fibrogenic and secrete chemokines that promote epithelial cell proliferation. TLR9-activated PSCs mediate their protumorigenic effects on the epithelial compartment via CCL11. Additionally, TLR9 has immune-suppressive effects in the tumor microenvironment (TME) via induction of regulatory T cell recruitment and myeloid-derived suppressor cell proliferation. Collectively, our work shows that TLR9 has protumorigenic effects in pancreatic carcinoma which are distinct from its influence in extrapancreatic malignancies and from the mechanistic effects of other TLRs on pancreatic oncogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Chemokine CCL11/metabolism , Chemokines/metabolism , Epithelial Cells/metabolism , Immunoblotting , Ligands , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Oligodeoxyribonucleotides/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Toll-Like Receptor 9/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
17.
PLoS One ; 10(7): e0132786, 2015.
Article in English | MEDLINE | ID: mdl-26172047

ABSTRACT

Cancer cachexia is a debilitating condition characterized by a combination of anorexia, muscle wasting, weight loss, and malnutrition. This condition affects an overwhelming majority of patients with pancreatic cancer and is a primary cause of cancer-related death. However, few, if any, effective therapies exist for both treatment and prevention of this syndrome. In order to develop novel therapeutic strategies for pancreatic cancer cachexia, appropriate animal models are necessary. In this study, we developed and validated a syngeneic, metastatic, murine model of pancreatic cancer cachexia. Using our model, we investigated the ability of transforming growth factor beta (TGF-ß) blockade to mitigate the metabolic changes associated with cachexia. We found that TGF-ß inhibition using the anti-TGF-ß antibody 1D11.16.8 significantly improved overall mortality, weight loss, fat mass, lean body mass, bone mineral density, and skeletal muscle proteolysis in mice harboring advanced pancreatic cancer. Other immunotherapeutic strategies we employed were not effective. Collectively, we validated a simplified but useful model of pancreatic cancer cachexia to investigate immunologic treatment strategies. In addition, we showed that TGF-ß inhibition can decrease the metabolic changes associated with cancer cachexia and improve overall survival.


Subject(s)
Cachexia/metabolism , Cachexia/mortality , Disease Models, Animal , Immunotherapy , Pancreatic Neoplasms/complications , Transforming Growth Factor beta/immunology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Body Composition , Cachexia/complications , Cachexia/therapy , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Muscular Atrophy/complications , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...