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 Mol Biol ; 430(12): 1786-1798, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29704491

ABSTRACT

We have engineered a panel of novel Fn3 scaffold-based proteins that bind with high specificity and affinity to each of the individual mouse Fcγ receptors (mFcγR). These binders were expressed as fusions to anti-tumor antigen single-chain antibodies and mouse serum albumin, creating opsonizing agents that invoke only a single mFcγR response rather than the broader activity of natural Fc isotypes, as well as all previously reported Fc mutants. This panel isolated the capability of each of the four mFcγRs to contribute to macrophage phagocytosis of opsonized tumor cells and in vivo tumor growth control with these monospecific opsonizing fusion proteins. All activating receptors (mFcγRI, mFcγRIII, and mFcγRIV) were capable of driving specific tumor cell phagocytosis to an equivalent extent, while mFcγRII, the inhibitory receptor, did not drive phagocytosis. Monospecific opsonizing fusion proteins that bound mFcγRI alone controlled tumor growth to an extent similar to the most active IgG2a murine isotype. As expected, binding to the inhibitory mFcγRII did not delay tumor growth, but unexpectedly, mFcγRIII also failed to control tumor growth. mFcγRIV exhibited detectable but lesser tumor-growth control leading to less overall survival compared to mFcγRI. Interestingly, in vivo macrophage depletion demonstrates their importance in tumor control with mFcγRIV engagement, but not with mFcγRI. This panel of monospecific mFcγR-binding proteins provides a toolkit for isolating the functional effects of each mFcγR in the context of an intact immune system.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Fibronectins/chemistry , Melanoma, Experimental/drug therapy , Protein Engineering/methods , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , HEK293 Cells , Humans , Melanoma, Experimental/immunology , Mice , Models, Molecular , Phagocytosis , Receptors, IgG/chemistry , Structural Homology, Protein , Xenograft Model Antitumor Assays
2.
J Exp Med ; 214(6): 1679-1690, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28473400

ABSTRACT

Certain RGD-binding integrins are required for cell adhesion, migration, and proliferation and are overexpressed in most tumors, making them attractive therapeutic targets. However, multiple integrin antagonist drug candidates have failed to show efficacy in cancer clinical trials. In this work, we instead exploit these integrins as a target for antibody Fc effector functions in the context of cancer immunotherapy. By combining administration of an engineered mouse serum albumin/IL-2 fusion with an Fc fusion to an integrin-binding peptide (2.5F-Fc), significant survival improvements are achieved in three syngeneic mouse tumor models, including complete responses with protective immunity. Functional integrin antagonism does not contribute significantly to efficacy; rather, this therapy recruits both an innate and adaptive immune response, as deficiencies in either arm result in reduced tumor control. Administration of this integrin-targeted immunotherapy together with an anti-PD-1 antibody further improves responses and predominantly results in cures. Overall, this well-tolerated therapy achieves tumor specificity by redirecting inflammation to a functional target fundamental to tumorigenic processes but expressed at significantly lower levels in healthy tissues, and it shows promise for translation.


Subject(s)
Adaptive Immunity , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immunotherapy , Integrins/metabolism , Adaptive Immunity/drug effects , Animals , Antibody Formation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/pathology , Cross Reactions/drug effects , Cross Reactions/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immune Tolerance/drug effects , Inflammation/pathology , Interleukin-2/metabolism , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Peptides/metabolism , Receptors, IgG/metabolism , Serum Albumin/metabolism , Species Specificity , Tissue Distribution/drug effects , Treatment Outcome
3.
Cell Rep ; 17(10): 2503-2511, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27926855

ABSTRACT

Numerous synergistic cancer immunotherapy combinations have been identified, but the effects of relative dose timing are rarely considered. In established syngeneic mouse tumor models, we found that staggering interferon-α (IFNα) administration after, rather than before or simultaneously with, serum-persistent interleukin-2 (IL-2) and tumor-specific antibody significantly increased long-term survival. Successful combination therapy required IFNα-induced activation of cross-presenting CD8α+ dendritic cells (DCs) following the release of antigenic tumor debris by the IL-2- and antibody-mediated immune response. Due to decreased phagocytic ability post-maturation, DCs activated too early captured less antigen and could not effectively prime CD8+ T cells. Temporally programming DC activation to occur after tumoricidal activity enhanced tumor control by multiple distinct combination immunotherapies, highlighting dose schedule as an underappreciated factor that can profoundly affect the success of multi-component immunotherapies.


Subject(s)
Interferon-alpha/immunology , Interleukin-2/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Humans , Immunotherapy , Interferon-alpha/administration & dosage , Interleukin-2/administration & dosage , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Melanoma, Experimental/pathology , Mice
4.
Nat Med ; 22(12): 1402-1410, 2016 12.
Article in English | MEDLINE | ID: mdl-27775706

ABSTRACT

Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte-associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Cytokines/drug effects , Immunotherapy/methods , Interleukin-2/pharmacology , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Adaptive Immunity , Animals , Cell Line, Tumor , Cytokines/immunology , Drug Therapy, Combination , Flow Cytometry , Gene Knockout Techniques , Immunity, Innate , Immunoblotting , Intramolecular Oxidoreductases/genetics , Mice , T-Lymphocytes/immunology
5.
Cancer Cell ; 27(4): 489-501, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25873172

ABSTRACT

Cancer immunotherapies under development have generally focused on either stimulating T cell immunity or driving antibody-directed effector functions of the innate immune system such as antibody-dependent cell-mediated cytotoxicity (ADCC). We find that a combination of an anti-tumor antigen antibody and an untargeted IL-2 fusion protein with delayed systemic clearance induces significant tumor control in aggressive isogenic tumor models via a concerted innate and adaptive response involving neutrophils, NK cells, macrophages, and CD8(+) T cells. This combination therapy induces an intratumoral "cytokine storm" and extensive lymphocyte infiltration. Adoptive transfer of anti-tumor T cells together with this combination therapy leads to robust cures of established tumors and development of immunological memory.


Subject(s)
Neoplasms/therapy , Adaptive Immunity , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Drug Synergism , Half-Life , Immunity, Innate , Immunotherapy , Interleukin-2/metabolism , Interleukin-2/pharmacokinetics , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...