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.
Front Immunol ; 15: 1323198, 2024.
Article in English | MEDLINE | ID: mdl-38384463

ABSTRACT

One of the most deadly and aggressive cancers in the world, pancreatic ductal adenocarcinoma (PDAC), typically manifests at an advanced stage. PDAC is becoming more common, and by the year 2030, it is expected to overtake lung cancer as the second greatest cause of cancer-related death. The poor prognosis can be attributed to a number of factors, including difficulties in early identification, a poor probability of curative radical resection, limited response to chemotherapy and radiotherapy, and its immunotherapy resistance. Furthermore, an extensive desmoplastic stroma that surrounds PDAC forms a mechanical barrier that prevents vascularization and promotes poor immune cell penetration. Phenotypic heterogeneity, drug resistance, and immunosuppressive tumor microenvironment are the main causes of PDAC aggressiveness. There is a complex and dynamic interaction between tumor cells in PDAC with stromal cells within the tumour immune microenvironment. The immune suppressive microenvironment that promotes PDAC aggressiveness is contributed by a range of cellular and humoral factors, which itself are modulated by the cancer. In this review, we describe the role of innate and adaptive immune cells, complex tumor microenvironment in PDAC, humoral factors, innate immune-mediated therapeutic advances, and recent clinical trials in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tumor Microenvironment , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Immunotherapy
2.
Cancer Res ; 77(24): 7049-7058, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29055013

ABSTRACT

T cell-based immunotherapies are a promising approach for patients with advanced cancers. However, various obstacles limit T-cell efficacy, including suboptimal T-cell receptor (TCR) activation and an immunosuppressive tumor environment. Here, we developed a fusion protein by linking CD8α and MyD88 (CD8α:MyD88) to enhance CD8+ T-cell responses to weakly immunogenic and poorly expressed tumor antigens. CD8α:MyD88-engineered T cells exhibited increased proliferation and expression of effector and costimulatory molecules in a tumor antigen-dependent manner. These effects were accompanied by elevated activation of TCR and Toll-like receptor signaling-related proteins. CD8α:MyD88-expressing T cells improved antitumor responses in mice. Enhanced antitumor activity was associated with a unique tumor cytokine/chemokine signature, improved T-cell infiltration, reduced markers of T-cell exhaustion, elevated levels of proteins associated with antigen presentation, and fewer macrophages with an immunosuppressive phenotype in tumors. Given these observations, CD8α:MyD88 represents a unique and versatile approach to help overcome immunosuppression and enhance T-cell responses to tumor antigens. Cancer Res; 77(24); 7049-58. ©2017 AACR.


Subject(s)
Antigens, Neoplasm/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Myeloid Differentiation Factor 88/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Recombinant Fusion Proteins/genetics
3.
Cancer Immunol Res ; 4(8): 708-16, 2016 08.
Article in English | MEDLINE | ID: mdl-27267778

ABSTRACT

The activation of TLR-MyD88 (Toll-like receptor-myeloid differentiation factor 88) signaling within T cells functions as a potent costimulatory signal that boosts antitumor and antiviral responses. However, the molecular mechanisms underlying the costimulatory processes are poorly understood. We compared microarray gene analysis data between TLR1-TLR2-stimulated and unstimulated T-cell receptor transgenic "pmel" and MyD88(-/-) pmel CD8(+) T cells and identified changes in the expression of several TNF family members. In particular, TLR stimulation increased 4-1BB levels in pmel but not in MyD88(-/-)pmel T cells. A link between 4-1BB and TLR1-TLR2 signaling in CD8(+) T cells was highlighted by the suboptimal responses of 4-1BB(-/-) T cells to TLR1-TLR2 agonist, but their normal response to CD28 or OX40 costimulation. Blocking 4-1BB signaling with antibodies also hindered the costimulatory effects of the TLR1-TLR2 agonist. The elevated levels of 4-1BB transcripts in TLR1-TLR2-stimulated cells were not due to increased mRNA stability nor increased histone activation, but instead were associated with increased binding of p65 and c-Jun to two distinct 4-1BB promoter sites. Combining TLR1-TLR2 ligand with an agonistic antibody to 4-1BB enhanced the antitumor activity in mice with established melanoma tumors. These studies reveal that the costimulatory effects of TLR1-TLR2 signaling in CD8(+) T cells are in part mediated by 4-1BB and are important for mounting an effective antitumor immune response. Cancer Immunol Res; 4(8); 708-16. ©2016 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Signal Transduction , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , Cell Survival/genetics , Cytotoxicity, Immunologic , Gene Expression , Gene Expression Profiling , Immunomodulation , Melanoma/immunology , Melanoma/metabolism , Melanoma, Experimental , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
J Clin Invest ; 125(3): 1081-97, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25642772

ABSTRACT

Signaling via the MyD88/IRAK pathway in T cells is indispensable for cell survival; however, it is not known whether this pathway functions in the progression of T acute lymphoblastic leukemia (T-ALL). Here, we determined that compared with thymic and peripheral T cells, T-ALL cells from patients have elevated levels of IRAK1 and IRAK4 mRNA as well as increased total and phosphorylated protein. Targeted inhibition of IRAK1 and IRAK4, either with shRNA or with a pharmacological IRAK1/4 inhibitor, dramatically impeded proliferation of T-ALL cells isolated from patients and T-ALL cells in a murine leukemia model; however, IRAK1/4 inhibition had little effect on cell death. We screened several hundred FDA-approved compounds and identified a set of drugs that had enhanced cytotoxic activity when combined with IRAK inhibition. Administration of an IRAK1/4 inhibitor or IRAK knockdown in combination with either ABT-737 or vincristine markedly reduced leukemia burden in mice and prolonged survival. IRAK1/4 signaling activated the E3 ubiquitin ligase TRAF6, increasing K63-linked ubiquitination and enhancing stability of the antiapoptotic protein MCL1; therefore, IRAK inhibition reduced MCL1 stability and sensitized T-ALL to combination therapy. These studies demonstrate that IRAK1/4 signaling promotes T-ALL progression through stabilization of MCL1 and suggest that impeding this pathway has potential as a therapeutic strategy to enhance chemotherapeutic efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , HEK293 Cells , Humans , Jurkat Cells , MCF-7 Cells , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Stability , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , TNF Receptor-Associated Factor 6/metabolism , Vincristine/pharmacology , Vincristine/therapeutic use , Xenograft Model Antitumor Assays
5.
J Leukoc Biol ; 93(6): 847-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23475577

ABSTRACT

Various TLR agonists are currently under investigation in clinical trials for their ability to orchestrate antitumor immunity. The antitumor responses are largely attributed to their aptitude to stimulate APCs such as DCs which in turn, activate tumor-specific T cell responses. However, there is a potential for TLR signaling to occur on cells other than professional APCs that could negate antitumor responses or even worse, promote tumor growth. The impetus for this review is twofold. First, there is accumulating data demonstrating that the engagement of TLRs on different T cell subsets and different cancer types could promote tumor growth or conversely, contribute to antitumor responses. Second, the efficacy of TLR agonists as monotherapies to treat cancer patients has been limited. In this review, we discuss how TLR signaling within different T cell subsets and cancer cells can potentially impact the generation of antitumor responses. Based on evidence from preclinical models and clinical trials, we draw attention to several criteria that we believe must be considered when selecting TLR agonists for developing effective immunotherapeutic strategies against cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Immunotherapy/methods , Neoplasms/therapy , Toll-Like Receptors/immunology , Animals , Clinical Trials as Topic , Humans , Neoplasms/immunology , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...