Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 7(1): 199, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31362778

ABSTRACT

BACKGROUND: Despite major advancements in immunotherapy among a number of solid tumors, response rates among ovarian cancer patients remain modest. Standard treatment for ovarian cancer is still surgery followed by taxane- and platinum-based chemotherapy. Thus, there is an urgent need to develop novel treatment options for clinical translation. METHODS: Our approach was to analyze the effects of standard chemotherapy in the tumor microenvironment of mice harboring orthotopic, syngeneic ID8-Vegf-Defb29 ovarian tumors in order to mechanistically determine a complementary immunotherapy combination. Specifically, we interrogated the molecular and cellular consequences of chemotherapy by analyzing gene expression and flow cytometry data. RESULTS: These data show that there is an immunosuppressive shift in the myeloid compartment, with increased expression of IL-10 and ARG1, but no activation of CD3+ T cells shortly after chemotherapy treatment. We therefore selected immunotherapies that target both the innate and adaptive arms of the immune system. Survival studies revealed that standard chemotherapy was complemented most effectively by a combination of anti-IL-10, 2'3'-cGAMP, and anti-PD-L1. Immunotherapy dramatically decreased the immunosuppressive myeloid population while chemotherapy effectively activated dendritic cells. Together, combination treatment increased the number of activated T and dendritic cells as well as expression of cytotoxic factors. It was also determined that the immunotherapy had to be administered concurrently with the chemotherapy to reverse the acute immunosuppression caused by chemotherapy. Mechanistic studies revealed that antitumor immunity in this context was driven by CD4+ T cells, which acquired a highly activated phenotype. Our data suggest that these CD4+ T cells can kill cancer cells directly via granzyme B-mediated cytotoxicity. Finally, we showed that this combination therapy is also effective at delaying tumor growth substantially in an aggressive model of lung cancer, which is also treated clinically with taxane- and platinum-based chemotherapy. CONCLUSIONS: This work highlights the importance of CD4+ T cells in tumor immunology. Furthermore, the data support the initiation of clinical trials in ovarian cancer that target both innate and adaptive immunity, with a focus on optimizing dosing schedules.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Gene Expression Profiling/methods , Immunity, Innate/drug effects , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Carboplatin/administration & dosage , Carboplatin/pharmacology , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-10/antagonists & inhibitors , Mice , Molecular Targeted Therapy , Nucleotides, Cyclic/administration & dosage , Nucleotides, Cyclic/pharmacology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Survival Analysis , Treatment Outcome , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
2.
Nat Commun ; 8(1): 1747, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170511

ABSTRACT

Targeted delivery of compounds to particular cell subsets can enhance therapeutic index by concentrating their action on the cells of interest. Because attempts to target tumors directly have yielded limited benefit, we instead target endogenous immune cell subsets in the circulation that can migrate actively into tumors. We describe antibody-targeted nanoparticles that bind to CD8+ T cells in the blood, lymphoid tissues, and tumors of mice. PD-1+ T cells are successfully targeted in the circulation and tumor. The delivery of an inhibitor of TGFß signaling to PD-1-expressing cells extends the survival of tumor-bearing mice, whereas free drugs have no effect at such doses. This modular platform also enables PD-1-targeted delivery of a TLR7/8 agonist to the tumor microenvironment, increasing the proportion of tumor-infiltrating CD8+ T cells and sensitizing tumors to subsequent anti-PD-1. Targeted delivery of immunotherapy to defined subsets of endogenous leukocytes may be superior to administration of free drugs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Imidazoles/administration & dosage , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL