Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 7(1): 132, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31113479

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors are not effective for pancreatic ductal adenocarcinoma (PDAC) as single agents. Vaccine therapy may sensitize PDACs to checkpoint inhibitor treatments. Annexin A2 (ANXA2) is a pro-metastasis protein, previously identified as a relevant PDAC antigen that is expressed by a GM-CSF-secreting allogenic whole pancreatic tumor cell vaccine (GVAX) to induce an anti-ANXA2 antibody response in patients with PDAC. We hypothesized that an ANXA2-targeting vaccine approach not only provokes an immune response but also mounts anti-tumor effects. METHODS: We developed a Listeria-based, ANXA2-targeting cancer immunotherapy (Lm-ANXA2) and investigated its effectiveness within two murine models of PDAC. RESULTS: We show that Lm-ANXA2 prolonged the survival in a transplant model of mouse PDACs. More importantly, priming with the Lm-ANXA2 treatment prior to administration of anti-PD-1 antibodies increased cure rates in the implanted PDAC model and resulted in objective tumor responses and prolonged survival in the genetically engineered spontaneous PDAC model. In tumors treated with Lm-ANXA2 followed by anti-PD-1 antibody, the T cells specific to ANXA2 had significantly increased INFγ expression. CONCLUSIONS: For the first time, a listeria vaccine-based immunotherapy was shown to be able to induce a tumor antigen-specific T cell response within the tumor microenvironment of a "cold" tumor such as PDAC and sensitize the tumor to checkpoint inhibitor therapy. Moreover, this combination immunotherapy led to objective tumor responses and survival benefit in the mice with spontaneously developed PDAC tumors. Therefore, our study supports developing Lm-ANXA2 as a therapeutic agent in combination with anti-PD-1 antibody for PDAC treatment.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/therapy , Listeria/immunology , Pancreatic Neoplasms/therapy , Animals , Annexin A2/antagonists & inhibitors , Annexin A2/genetics , Annexin A2/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
2.
J Clin Invest ; 129(4): 1742-1755, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30747725

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents an immune quiescent tumor that is resistant to immune checkpoint inhibitors. Previously, our group has shown that a GM-CSF-secreting allogenic pancreatic tumor cell vaccine (GVAX) may prime the tumor microenvironment by inducing intratumoral T cell infiltration. Here, we show that untreated PDACs express minimal indoleamine-2,3-dioxygenase (IDO1); however, GVAX therapy induced IDO1 expression on tumor epithelia as well as vaccine-induced tertiary lymphoid aggregates. IDO1 expression plays a role in regulating the polarization of Th1, Th17, and possibly T regulatory cells in PDAC tumors. IDO1 inhibitor enhanced antitumor efficacy of GVAX in a murine model of PDACs. The combination of vaccine and IDO1 inhibitor enhanced intratumoral T cell infiltration and function, but adding anti-PD-L1 antibody to the combination did not offer further synergy and in fact may have had a negative interaction, decreasing the number of intratumoral effector T cells. Additionally, IDO1 inhibitor in the presence of vaccine therapy did not significantly modulate intratumoral myeloid-derived suppressor cells quantitatively, but diminished their suppressive effect on CD8+ proliferation. Our study supports the combination of IDO1 inhibitor and vaccine therapy; however, it does not support the combination of IDO1 inhibitor and anti-PD-1/PD-L1 antibody for T cell-inflamed tumors such as PDACs treated with vaccine therapy.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines/pharmacology , Carcinoma, Pancreatic Ductal/therapy , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Mice , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
3.
Mol Cancer Ther ; 16(11): 2399-2409, 2017 11.
Article in English | MEDLINE | ID: mdl-28864680

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most chemotherapy- and radiotherapy-resistant tumors. The c-Met and Hedgehog (Hh) pathways have been shown previously by our group to be key regulatory pathways in the primary tumor growth and metastases formation. Targeting both the HGF/c-Met and Hh pathways has shown promising results in preclinical studies; however, the benefits were not readily translated into clinical trials with PDAC patients. In this study, utilizing mouse models of PDAC, we showed that inhibition of either HGF/c-Met or Hh pathways sensitize the PDAC tumors to gemcitabine, resulting in decreased primary tumor volume as well as significant reduction of metastatic tumor burden. However, prolonged treatment of single HGF/c-Met or Hh inhibitor leads to resistance to these single inhibitors, likely because the single c-Met treatment leads to enhanced expression of Shh, and vice versa. Targeting both the HGF/c-Met and Hh pathways simultaneously overcame the resistance to the single-inhibitor treatment and led to a more potent antitumor effect in combination with the chemotherapy treatment. Mol Cancer Ther; 16(11); 2399-409. ©2017 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Hepatocyte Growth Factor/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Benzamides/administration & dosage , Biphenyl Compounds/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Hepatocyte Growth Factor/genetics , Humans , Imidazoles , Mice , Mice, Transgenic , Neoplasm Metastasis , Proto-Oncogene Proteins c-met/genetics , Pyridines/administration & dosage , Triazines , Tumor Microenvironment/drug effects , Gemcitabine
4.
Cancer Res ; 77(1): 41-52, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27821486

ABSTRACT

Understanding how stromal signals regulate the development of pancreatic ductal adenocarcinoma (PDAC) may suggest novel therapeutic interventions in this disease. In this study, we assessed the metastatic role of stromal signals suggested to be important in the PDAC microenvironment. Src and IGF-1R phosphorylated the prometastatic molecule Annexin A2 (AnxA2) at Y23 and Y333 in response to stromal signals HGF and IGF-1, respectively, and IGF-1 expression was regulated by the Sonic Hedgehog (Shh) pathway. Both Shh and HGF were heterogeneously expressed in PDAC stroma, and only dual inhibition of these pathways could significantly suppress AnxA2 phosphorylation, PDAC growth, and metastasis. Taken together, our results illuminate tumor-stromal interactions, which drive metastasis, and provide a mechanism-based rationale for a stroma-directed therapy for PDAC. Cancer Res; 77(1); 41-52. ©2016 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Tumor Microenvironment/physiology , Animals , Blotting, Western , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Heterografts , Humans , Immunohistochemistry , Mice , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism , Tandem Mass Spectrometry
5.
Cancer Res ; 76(18): 5395-404, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27496707

ABSTRACT

Stromal fibrosis is a prominent histologic characteristic of pancreatic ductal adenocarcinoma (PDAC), but how stromal fibroblasts are regulated in the tumor microenvironment (TME) to support tumor growth is largely unknown. Here we show that PDAC cells can induce DNA methylation in cancer-associated fibroblasts (CAF). Upon direct contact with PDAC cells, DNA methylation of SOCS1 and other genes is induced in mesenchymal stem cells or in CAF that lack SOCS1 methylation at baseline. Silencing or decitabine treatment to block the DNA methylation enzyme DNMT1 inhibited methylation of SOCS1. In contrast, SOCS1 gene methylation and downregulation in CAF activated STAT3 and induced insulin-like growth factor-1 expression to support PDAC cell growth. Moreover, CAF facilitated methylation-dependent growth of PDAC tumor xenografts in mice. The ability of patient-derived CAF with SOCS1 methylation to promote PDAC growth was more robust than CAF without SOCS1 methylation. Overall, our results reveal how PDAC cells can reprogram CAF to modify tumor-stromal interactions in the TME, which promote malignant growth and progression. Cancer Res; 76(18); 5395-404. ©2016 AACR.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/genetics , DNA Methylation/genetics , Pancreatic Neoplasms/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Tumor Microenvironment
6.
Cancer Biol Med ; 12(3): 201-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26487965

ABSTRACT

Immune checkpoint inhibitors are increasingly drawing much attention in the therapeutic development for cancer treatment. However, many cancer patients do not respond to treatments with immune checkpoint inhibitors, partly because of the lack of tumor-infiltrating effector T cells. Cancer vaccines may prime patients for treatments with immune checkpoint inhibitors by inducing effector T-cell infiltration into the tumors and immune checkpoint signals. The combination of cancer vaccine and an immune checkpoint inhibitor may function synergistically to induce more effective antitumor immune responses, and clinical trials to test the combination are currently ongoing.

7.
Sci Signal ; 8(388): ra77, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26243191

ABSTRACT

Most patients with pancreatic ductal adenocarcinoma (PDA) present with metastatic disease at the time of diagnosis or will recur with metastases after surgical treatment. Semaphorin-plexin signaling mediates the migration of neuronal axons during development and of blood vessels during angiogenesis. The expression of the gene encoding semaphorin 3D (Sema3D) is increased in PDA tumors, and the presence of antibodies against the pleiotropic protein annexin A2 (AnxA2) in the sera of some patients after surgical resection of PDA is associated with longer recurrence-free survival. By knocking out AnxA2 in a transgenic mouse model of PDA (KPC) that recapitulates the progression of human PDA from premalignancy to metastatic disease, we found that AnxA2 promoted metastases in vivo. The expression of AnxA2 promoted the secretion of Sema3D from PDA cells, which coimmunoprecipitated with the co-receptor plexin D1 (PlxnD1) on PDA cells. Mouse PDA cells in which SEMA3D was knocked down or ANXA2-null PDA cells exhibited decreased invasive and metastatic potential in culture and in mice. However, restoring Sema3D in AnxA2-null cells did not entirely rescue metastatic behavior in culture and in vivo, suggesting that AnxA2 mediates additional prometastatic mechanisms. Patients with primary PDA tumors that have abundant Sema3D have widely metastatic disease and decreased survival compared to patients with tumors that have relatively low Sema3D abundance. Thus, AnxA2 and Sema3D may be new therapeutic targets and prognostic markers of metastatic PDA.


Subject(s)
Annexin A2/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Semaphorins/genetics , Signal Transduction/genetics , Animals , Annexin A2/metabolism , Autocrine Communication/genetics , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence/classification , Neoplasm Metastasis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Survival Analysis , Tumor Cells, Cultured , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...