Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
J Immunother Cancer ; 8(2)2020 09.
Article in English | MEDLINE | ID: mdl-32958686

ABSTRACT

BACKGROUND: Immunotherapies, such as immune checkpoint inhibitors and adoptive cell therapies, have revolutionized cancer treatment and resulted in complete and durable responses in some patients. Unfortunately, most immunotherapy treated patients still fail to respond. Absence of T cell infiltration to the tumor site is one of the major obstacles limiting immunotherapy efficacy against solid tumors. Thus, the development of strategies that enhance T cell infiltration and broaden the antitumor efficacy of immunotherapies is greatly needed. METHODS: We used mouse tumor models, genetically deficient mice and vascular endothelial cells (VECs) to study the requirements for T cell infiltration into tumors. RESULTS: A specific formulation of poly-IC, containing poly-lysine and carboxymethylcellulose (PICLC) facilitated the traffic and infiltration of effector CD8 T cells into the tumors that reduced tumor growth. Surprisingly, intratumoral injection of PICLC was significantly less effective in inducing tumor T cell infiltration and controlling growth of tumors as compared with systemic (intravenous or intramuscular) administration. Systemically administered PICLC, but not poly-IC stimulated tumor VECs via the double-stranded RNA cytoplasmic sensor MDA5, resulting in enhanced adhesion molecule expression and the production of type I interferon (IFN-I) and T cell recruiting chemokines. Expression of IFNαß receptor in VECs was necessary to obtain the antitumor effects by PICLC and IFN-I was found to directly stimulate the secretion of T cell recruiting chemokines by VECs indicating that this cytokine-chemokine regulatory axis is crucial for recruiting effector T cells into the tumor parenchyma. Unexpectedly, these effects of PICLC were mostly observed in tumors and not in normal tissues. CONCLUSIONS: These findings have strong implications for the improvement of all types of T cell-based immunotherapies for solid cancers. We predict that systemic administration of PICLC will improve immune checkpoint inhibitor therapy, adoptive cell therapies and therapeutic cancer vaccines.


Subject(s)
Immunotherapy/methods , Poly I-C/metabolism , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Female , Humans , Mice
2.
Cancer Immunol Res ; 6(5): 617-627, 2018 05.
Article in English | MEDLINE | ID: mdl-29483127

ABSTRACT

Peptide vaccines can be a successful and cost-effective way of generating T-cell responses against defined tumor antigens, especially when combined with immune adjuvants such as poly-IC. However, strong immune adjuvants can induce a collateral increase in numbers of irrelevant, nonspecific T cells, which limits the effectiveness of the peptide vaccines. Here, we report that providing prolonged IL2 signaling in the form of either IL2/anti-IL2 complexes or pegylated IL2 overcomes the competitive suppressive effect of irrelevant T cells, allowing the preferential expansion of antigen-specific T cells. In addition to increasing the number of tumor-reactive T cells, sustained IL2 enhanced the ability of T cells to resist PD-1-induced negative signals, increasing the therapeutic effectiveness of the vaccines against established tumors. This vaccination strategy using peptides and sustained IL2 could be taken into the clinic for the treatment of cancer. Cancer Immunol Res; 6(5); 617-27. ©2018 AACR.


Subject(s)
Cancer Vaccines/therapeutic use , Cell Proliferation/drug effects , Immunotherapy, Adoptive/methods , Interleukin-2/administration & dosage , Neoplasms/therapy , T-Lymphocytes/drug effects , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/immunology , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Drug Administration Schedule , Drug Synergism , Female , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Poly I-C/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Rats , Signal Transduction/drug effects , T-Lymphocytes/physiology , Tumor Cells, Cultured , Vaccines, Subunit/therapeutic use
3.
Cancer Immunol Immunother ; 66(2): 203-213, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27052572

ABSTRACT

The design of efficacious and cost-effective therapeutic vaccines against cancer remains both a research priority and a challenge. For more than a decade, our laboratory has been involved in the development of synthetic peptide-based anti-cancer therapeutic vaccines. We first dedicated our efforts in the identification and validation of peptide epitopes for both CD8 and CD4 T cells from tumor-associated antigens (TAAs). Because of suboptimal immune responses and lack of therapeutic benefit of peptide vaccines containing these epitopes, we have focused our recent efforts in optimizing peptide vaccinations in mouse tumor models using numerous TAA epitopes. In this focused research review, we describe how after taking lessons from the immune system's way of dealing with acute viral infections, we have designed peptide vaccination strategies capable of generating very high numbers of therapeutically effective CD8 T cells. We also discuss some of the remaining challenges to translate these findings into the clinical setting.


Subject(s)
Cancer Vaccines/immunology , Melanoma, Experimental/immunology , Vaccines, Subunit/immunology , Virus Diseases/immunology , Animals , Disease Models, Animal , Humans
4.
Leuk Res ; 38(3): 377-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24461365

ABSTRACT

The nitric oxide (NO) prodrug JS-K, a promising anti-cancer agent, consists of a diazeniumdiolate group necessary for the release of NO as well as an arylating ring. In this study, we research the mechanism by which JS-K kills a murine erythroleukemia cell line and determine the roles of NO and arylation in the process. Our studies indicate that JS-K inhibits the PI 3-kinase/Akt and MAP kinase pathways. This correlates with the activation of the tumor suppressor FoxO3a and increased expression of various caspases, leading to apoptosis. The arylating capability of JS-K appears to be sufficient for inducing these biological effects. Overall, these data suggest that JS-K kills tumor cells by arylating and inactivating signaling molecules that block the activation of a tumor suppressor.


Subject(s)
Azo Compounds/pharmacology , Cytotoxins/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Prodrugs/pharmacology , Animals , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Forkhead Box Protein O3 , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
5.
Immunopharmacol Immunotoxicol ; 29(3-4): 487-98, 2007.
Article in English | MEDLINE | ID: mdl-18075860

ABSTRACT

Langerhans cells (LC) are immature dendritic cells (DC) present in the skin epithelium. To understand the molecular and cellular mechanisms governing the inflammatory reaction in atopic dermatitis (AD), the expression of the LC specific marker CD1a, a member of major histocompatibility (MHC)-like glycoproteins, and the co-stimulatory molecules CD80 and CD86, expressed on functionally mature dendritic cells, were counted in lesional biopsies and normal epidermis by an immunohistochemical method. CD1a specific staining was observed in both normal and AD lesion specimens. CD80 and CD86 positive cells with morphological characteristics of the LC were found in lesional AD epidermis, suggesting a high level of functional maturity of these cells and their involvement in chronic inflammatory disease.


Subject(s)
Dermatitis, Atopic/metabolism , Epidermis/metabolism , Epidermis/pathology , Langerhans Cells/metabolism , Animals , Antigens, CD1/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Chronic Disease , Dermatitis, Atopic/pathology , Eczema/metabolism , Eczema/pathology , Humans , Immunoglobulins/metabolism , Immunohistochemistry , Mice
SELECTION OF CITATIONS
SEARCH DETAIL