Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Cancer Immunol Res ; 11(4): 466-485, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36757308

ABSTRACT

Oncolytic virus therapies induce the direct killing of tumor cells and activation of conventional dendritic cells (cDC); however, cDC activation has not been optimized with current therapies. We evaluated the adenoviral delivery of engineered membrane-stable CD40L (MEM40) and IFNß to locally activate cDCs in mouse tumor models. Combined tumor MEM40 and IFNß expression induced the highest cDC activation coupled with increased lymph node migration, increased systemic antitumor CD8+ T-cell responses, and regression of established tumors in a cDC1-dependent manner. MEM40 + IFNß combined with checkpoint inhibitors led to effective control of distant tumors and lung metastases. An oncolytic adenovirus (MEM-288) expressing MEM40 + IFNß  in phase I clinical testing induced cancer cell loss concomitant with enhanced T-cell infiltration and increased systemic presence of tumor T-cell clonotypes in non-small cell lung cancer (NSCLC) patients. This approach to simultaneously target two major DC-activating pathways has the potential to significantly affect the solid tumor immunotherapy landscape.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , CD40 Ligand , CD8-Positive T-Lymphocytes , Dendritic Cells , Immunotherapy , Cell Line, Tumor
3.
J Immunol ; 206(8): 1966-1975, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33722878

ABSTRACT

Inflammation has long been associated with cancer initiation and progression; however, how inflammation causes immune suppression in the tumor microenvironment and resistance to immunotherapy is not well understood. In this study, we show that both innate proinflammatory cytokine IL-1α and immunotherapy-induced IL-1α make melanoma resistant to immunotherapy. In a mouse melanoma model, we found that tumor size was inversely correlated with response to immunotherapy. Large tumors had higher levels of IL-1α, Th2 cytokines, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), and regulatory T cells but lower levels of IL-12, Th1 cytokines, and activated T cells. We found that therapy with adenovirus-encoded CD40L (rAd.CD40L) increased tumor levels of IL-1α and PMN-MDSCs. Blocking the IL-1 signaling pathway significantly decreased rAd.CD40L-induced PMN-MDSCs and their associated PD-L1 expression in the tumor microenvironment and enhanced tumor-specific immunity. Similarly, blocking the IL-1 signaling pathway improved the antimelanoma activity of anti-PD-L1 Ab therapy. Our study suggests that blocking the IL-1α signaling pathway may increase the efficacy of immunotherapies against melanoma.


Subject(s)
Drug Resistance, Neoplasm/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Interleukin-1alpha/immunology , Melanoma, Experimental/therapy , Animals , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors/immunology , Interleukin-1alpha/metabolism , Kaplan-Meier Estimate , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
Nat Commun ; 8(1): 1447, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29129918

ABSTRACT

CD40 agonists bind the CD40 molecule on antigen-presenting cells and activate them to prime tumor-specific CD8+ T cell responses. Here, we study the antitumor activity and mechanism of action of a nonreplicating adenovirus encoding a chimeric, membrane-bound CD40 ligand (ISF35). Intratumoral administration of ISF35 in subcutaneous B16 melanomas generates tumor-specific, CD8+ T cells that express PD-1 and suppress tumor growth. Combination therapy of ISF35 with systemic anti-PD-1 generates greater antitumor activity than each respective monotherapy. Triple combination of ISF35, anti-PD-1, and anti-CTLA-4 results in complete eradication of injected and noninjected subcutaneous tumors, as well as melanoma tumors in the brain. Therapeutic efficacy is associated with increases in the systemic level of tumor-specific CD8+ T cells, and an increased ratio of intratumoral CD8+ T cells to CD4+ Tregs. These results provide a proof of concept of systemic antitumor activity after intratumoral CD40 triggering with ISF35 in combination with checkpoint blockade for multifocal cancer, including the brain.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , CD40 Antigens/agonists , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Adenoviridae/genetics , Animals , Brain/pathology , CD4-CD8 Ratio , CD40 Antigens/metabolism , CD40 Ligand/genetics , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Enzyme Activation , Female , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis
5.
Proc Natl Acad Sci U S A ; 105(8): 3047-52, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287027

ABSTRACT

We examined the sera of six patients before and after i.v. infusions of autologous chronic lymphocytic leukemia (CLL) cells transduced ex vivo with an adenovirus encoding CD154 (Ad-CD154). Five patients made high-titer antibodies against adenovirus and three made IgG reactive with a leukemia-associated surface antigen, which we identified as ROR1. Anti-ROR1 antibodies were not detected in the sera of untreated patients. We generated anti-ROR1 mAbs and found they reacted specifically with the CLL cells of all patients, but not with nonleukemic leukocytes, a wide variety of normal adult tissues, or blood mononuclear cells, including CD5(+) B cells of healthy adults. ROR1 could bind Wnt5a, which induced activation of NF-kappaB when coexpressed with ROR1 in HEK293 cells and enhanced the survival of CLL cells in vitro, an effect that could be neutralized by posttreatment anti-ROR1 antisera. We conclude that patients with CLL can break immune tolerance to ROR1, which is an oncofetal surface antigen and survival-signaling receptor in this neoplastic disease.


Subject(s)
CD40 Ligand/immunology , Immune Sera/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Proteins/metabolism , Adenoviridae/immunology , Animals , Antibodies, Monoclonal , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA Primers , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Molecular Sequence Data , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptor Tyrosine Kinase-like Orphan Receptors , Sequence Analysis, DNA , Tumor Cells, Cultured , Wnt-5a Protein
6.
Br J Haematol ; 118(2): 506-13, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12139739

ABSTRACT

Neoplastic plasma cells from patients with myeloma fail to stimulate an effective anti-myeloma immune response, which may be in part due to their deficient expression of immune accessory molecules. Attempting to alter this, we infected myeloma cell lines and patient-derived primary myeloma cells with an adenovirus encoding CD154 (Ad-CD154). Myeloma cells were made to express the CD154 transgene at multiplicity of infection (MOI) between 10 and 1000. Furthermore, infection of CD40(positive) myeloma cells with Ad-CD154, but not an adenovirus encoding an irrelevant transgene, beta-galactosidase (Ad-LacZ), induced enhanced expression of immune accessory molecules, such as CD54, HLA-DR and CD70. In addition, Ad-CD154-infected myeloma cells could activate bystander CD40(positive) antigen-presenting cells to express immune accessory molecules. Consequently, Ad-CD154 infected myeloma cells stimulated proliferation in allogeneic mixed lymphocyte reactions (MLR). Finally, co-infection of CD40(negative) myeloma cells with Ad-CD154 and an adenovirus encoding CD40 (Ad-CD40) induced expression of immune accessory molecules and enhanced the MLR stimulatory capacity of transduced myeloma cells. Collectively, these results indicate that infection of myeloma cells with Ad-CD154 or Ad-CD154/Ad-CD40 can induce changes in myeloma cells that enhance their ability to induce cellular immune activation. As such, this approach may have potential application for immune therapy of patients with this disease.


Subject(s)
CD40 Antigens/immunology , Multiple Myeloma/immunology , Adenoviridae , Adenoviridae Infections/immunology , Bone Marrow Cells/pathology , Cell Division , Gene Transfer Techniques , Genetic Vectors , Humans , Immunotherapy/methods , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Transduction, Genetic/methods , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...