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 ; 11: 922, 2020.
Article in English | MEDLINE | ID: mdl-32499782

ABSTRACT

Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immunity , Mice , Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
2.
Cell Rep ; 30(3): 714-724.e5, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968248

ABSTRACT

Due to limited current therapies, metastases are the primary cause of mortality in cancer patients. Here, we employ a fusion compound of the cytokine LIGHT and a vascular targeting peptide (LIGHT-VTP) that homes to angiogenic blood vessels in primary tumors. We show in primary mouse lung cancer that normalization of tumor vasculature by LIGHT-VTP prevents cancer cell intravasation. Further, LIGHT-VTP efficiently targets pathological blood vessels in the pre-metastatic niche, reducing vascular hyper-permeability and extracellular matrix (ECM) deposition, thus blocking metastatic lung colonization. Moreover, we demonstrate that mouse and human metastatic melanoma deposits are targetable by VTP. In overt melanoma metastases, LIGHT-VTP normalizes intra-metastatic blood vessels and increases GrzB+ effector T cells. Successful treatment induces high endothelial venules (HEVs) and lymphocyte clusters, which sensitize refractory lung metastases to anti-PD-1 checkpoint inhibitors. These findings demonstrate an important application for LIGHT-VTP therapy in preventing metastatic development as well as exerting anti-tumor effects in established metastases.


Subject(s)
Immunotherapy , Lung/blood supply , Lung/pathology , Neovascularization, Pathologic/pathology , Animals , Humans , Immunity , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymph Nodes/pathology , Male , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice, Inbred C57BL , Neoadjuvant Therapy , Neoplasm Metastasis , Peptides/therapeutic use , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use
3.
Cancer Gene Ther ; 17(10): 730-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20559332

ABSTRACT

Primary tumor cells genetically modified to express a collection of immunological ligands on their surface may have the utility as therapeutic autologous cancer vaccines. However, genetic modification of primary tumor cells is not only cost, labor and time intensive, but also has safety repercussions. As an alternative, we developed the ProtEx technology that involves generation of immunological ligands with core streptavidin (SA) and their display on biotinylated cells in a rapid and efficient manner. We herein demonstrate that TC-1 tumor cells can be rapidly and efficiently engineered to codisplay on their surface two costimulatory proteins, SA-4-1BBL and SA-LIGHT, simultaneously. Vaccination with irradiated TC-1 cells codisplaying both chimeric proteins showed 100% efficacy in a prophylactic and >55% efficacy in a therapeutic tumor setting. In contrast, vaccination with TC-1 cells engineered with either protein alone showed significantly reduced efficacy in the prophylactic setting. Vaccine efficacy was associated with the generation of primary and memory T-cell and antibody responses against the tumor without detectable signs of autoimmunity. Engineering tumor cells in a rapid and effective manner to simultaneously display on their surface a collection of immunostimulatory proteins with additive/synergistic functions presents a novel alternative approach to gene therapy with considerable potential for cancer immunotherapy.


Subject(s)
4-1BB Ligand/therapeutic use , Cancer Vaccines/genetics , Tumor Cells, Cultured/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , 4-1BB Ligand/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Genetic Therapy , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
4.
Cell Mol Immunol ; 7(4): 296-305, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20418899

ABSTRACT

Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Immunity/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokines/metabolism , Female , Gene Transfer Techniques , Humans , Immunization , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/prevention & control , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/immunology
5.
Science ; 316(5822): 285-8, 2007 Apr 13.
Article in English | MEDLINE | ID: mdl-17431181

ABSTRACT

Hyperlipidemia, one of the most important risk factors for coronary heart disease, is often associated with inflammation. We identified lymphotoxin (LT) and LIGHT, tumor necrosis factor cytokine family members that are primarily expressed on lymphocytes, as critical regulators of key enzymes that control lipid metabolism. Dysregulation of LIGHT expression on T cells resulted in hypertriglyceridemia and hypercholesterolemia. In low-density lipoprotein receptor-deficient mice, which lack the ability to control lipid levels in the blood, inhibition of LT and LIGHT signaling with a soluble lymphotoxin beta receptor decoy protein attenuated the dyslipidemia. These results suggest that the immune system directly influences lipid metabolism and that LT modulating agents may represent a novel therapeutic route for the treatment of dyslipidemia.


Subject(s)
Lipid Metabolism , Liver/metabolism , Lymphotoxin beta Receptor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Cells, Cultured , Dyslipidemias/drug therapy , Dyslipidemias/etiology , Dyslipidemias/metabolism , Female , Homeostasis , Humans , Hypercholesterolemia/etiology , Lipids/blood , Lymphotoxin beta Receptor/therapeutic use , Lymphotoxin-alpha/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...