Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 65(5): 537-49, 2016 May.
Article in English | MEDLINE | ID: mdl-26961085

ABSTRACT

The dramatic clinical benefit of immune checkpoint blockade for a fraction of cancer patients suggests the potential for further clinical benefit in a broader cancer patient population by combining immune checkpoint inhibitors with active immunotherapies. The anti-tumor efficacy of MVA-BN-HER2 poxvirus-based active immunotherapy alone or in combination with CTLA-4 checkpoint blockade was investigated in a therapeutic CT26-HER-2 lung metastasis mouse model. MVA-BN-HER2 immunotherapy significantly improved the median overall survival compared to untreated controls or CTLA-4 blockade alone (p < 0.001). Robust synergistic efficacy was achieved with the combination therapy (p < 0.01). Improved survival following MVA-BN-HER2 administration was accompanied by increased tumor infiltration by HER-2-specific cytotoxic T lymphocytes (CTL). These tumor-specific CTL had characteristics similar to antiviral CTL, including strong expression of activation markers and co-expression of IFNγ and TNFα. Combination with CTLA-4 blockade significantly increased the magnitude of HER-2-specific T cell responses, with a higher proportion co-expressing TNFα and/or IL-2 with IFNγ. Furthermore, in mice treated with MVA-BN-HER2 (alone or in combination with CTLA-4 blockade), the inducible T cell co-stimulator (ICOS) protein was expressed predominantly on CD4 and CD8 effector T cells but not on regulatory T cells (T(reg)). In contrast, mice left untreated or treated solely with CTLA-4 blockade harbored elevated ICOS(+) Treg, a phenotype associated with highly suppressive activity. In conclusion, poxvirus-based active immunotherapy induced robust tumor infiltration by highly efficient effector T cells. Combination with CTLA-4 immune checkpoint blockade amplified this response resulting in synergistically improved efficacy. These hypothesis-generating data may help elucidate evidence of enhanced clinical benefit from combining CTLA-4 blockade with poxvirus-based active immunotherapy.


Subject(s)
CTLA-4 Antigen/immunology , Cancer Vaccines/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/pharmacology , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Drug Synergism , Female , Flow Cytometry , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccinia virus/genetics
2.
PLoS One ; 11(2): e0150084, 2016.
Article in English | MEDLINE | ID: mdl-26910562

ABSTRACT

Poxvirus-based active immunotherapies mediate anti-tumor efficacy by triggering broad and durable Th1 dominated T cell responses against the tumor. While monotherapy significantly delays tumor growth, it often does not lead to complete tumor regression. It was hypothesized that the induced robust infiltration of IFNγ-producing T cells into the tumor could provoke an adaptive immune evasive response by the tumor through the upregulation of PD-L1 expression. In therapeutic CT26-HER-2 tumor models, MVA-BN-HER2 poxvirus immunotherapy resulted in significant tumor growth delay accompanied by a robust, tumor-infiltrating T cell response that was characterized by low to mid-levels of PD-1 expression on T cells. As hypothesized, this response was countered by significantly increased PD-L1 expression on the tumor and, unexpectedly, also on infiltrating T cells. Synergistic benefit of anti-tumor therapy was observed when MVA-BN-HER2 immunotherapy was combined with PD-1 immune checkpoint blockade. Interestingly, PD-1 blockade stimulated a second immune checkpoint molecule, LAG-3, to be expressed on T cells. Combining MVA-BN-HER2 immunotherapy with dual PD-1 plus LAG-3 blockade resulted in comprehensive tumor regression in all mice treated with the triple combination therapy. Subsequent rejection of tumors lacking the HER-2 antigen by treatment-responsive mice without further therapy six months after the original challenge demonstrated long lasting memory and suggested that effective T cell immunity to novel, non-targeted tumor antigens (antigen spread) had occurred. These data support the clinical investigation of this triple therapy regimen, especially in cancer patients harboring PD-L1neg/low tumors unlikely to benefit from immune checkpoint blockade alone.


Subject(s)
Antigens, CD/immunology , Immunity, Cellular , Immunotherapy , Neoplasms, Experimental/therapy , Poxviridae/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Cell Line, Tumor , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Poxviridae/genetics , Programmed Cell Death 1 Receptor/genetics , Lymphocyte Activation Gene 3 Protein
3.
J Immunother Cancer ; 2(1): 34, 2014.
Article in English | MEDLINE | ID: mdl-25328681

ABSTRACT

BACKGROUND: PROSTVAC®, an active immunotherapy currently studied for the treatment of metastatic castration-resistant prostate cancer (mCRPC), consists of a heterologous prime-boost regimen with two different poxvirus-based vectors to provoke productive immune responses against prostate specific antigen (PSA) as the target tumor antigen. A Phase 2 study of PROSTVAC immunotherapy showed significantly improved median overall survival by 8.5 months and is currently being validated in a global Phase 3 study (PROSPECT; NCT01322490). Here, preclinical models were explored to investigate the mechanism of action and immune signatures of anti-tumor efficacy with PROSTVAC immunotherapy with the goal to identify potential immune correlates of clinical benefit. METHODS: PROSTVAC-induced immune responses and anti-tumor efficacy were studied in male BALB/c mice. Functionality of the induced T cell response was characterized by interferon-gamma (IFNγ) ELISPOT, cytotoxic degranulation, multi-cytokine intracellular staining, and in vivo T cell depletion. Tumor infiltrating lymphocytes (TILs) were evaluated phenotypically by flow cytometry. RESULTS: The heterologous prime-boost regimen of the two PROSTVAC vectors significantly enhanced the magnitude and quality of activated PSA-specific CD4 and CD8 T cell responses compared to homologous, single vector regimens. PROSTVAC-activated CD4 and CD8 T cells were highly functional as evidenced by expression of activation markers, production of multiple cytokines, and amplified cytotoxic T cell activity. Importantly, PROSTVAC immunotherapy resulted in significant anti-tumor efficacy in a transplantable prostate cancer mouse model. Antigen-spreading occurred in PROSTVAC-treated animals that rejected PSA-expressing tumors, as shown by subsequent rejection of PSA-negative tumors. In vivo CD4 and CD8 depletion revealed that both T cell subsets contributed to anti-tumor efficacy. Characterization of TILs demonstrated that PROSTVAC immunotherapy greatly increased the intra-tumoral ratio of activated effector to regulatory T cells. CONCLUSIONS: PROSTVAC immunotherapy activates broad, highly functional T cell immunity to PSA and to endogenous tumor antigens via immune-mediated antigen spreading. These preclinical results further elucidate the mode of action of PROSTVAC immunotherapy and its potential causal relationship to extended overall survival as observed in the PROSTVAC Phase 2 study. The clinical validation is ongoing in the PROSPECT Phase 3 clinical study.

4.
Cancer Immunol Immunother ; 61(1): 19-29, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21822917

ABSTRACT

MVA-BN®-HER2 is a new candidate immunotherapy designed for the treatment of HER-2-positive breast cancer. Here, we demonstrate that a single treatment with MVA-BN®-HER2 exerts potent anti-tumor efficacy in a murine model of experimental pulmonary metastasis. This anti-tumor efficacy occurred despite a strong tumor-mediated immunosuppressive environment characterized by a high frequency of regulatory T cells (T(reg)) in the lungs of tumor-bearing mice. Immunogenicity studies showed that treatment with MVA-BN®-HER2 induced strongly Th1-dominated HER-2-specific antibody and T-cell responses. MVA-BN®-HER2-induced anti-tumor activity was characterized by an increased infiltration of lungs with highly activated, HER-2-specific, CD8+CD11c+ T cells accompanied by a decrease in the frequency of T(reg) cells in the lung, resulting in a significantly increased ratio of effector T cells to T(reg) cells. In contrast, administration of HER2 protein formulated in Complete Freund's Adjuvant (CFA) induced a strongly Th2-biased immune response to HER-2. However, this did not lead to significant infiltration of the tumor-bearing lungs by CD8+ T cells or the decrease in the frequency of T(reg) cells nor did it result in anti-tumor efficacy. In vivo depletion of CD8+ cells confirmed that CD8 T cells were required for the anti-tumor activity of MVA-BN®-HER2. Furthermore, depletion of CD4+ or CD25+ cells demonstrated that tumor-induced T(reg) cells promoted tumor growth and that CD4 effector cells also contribute to MVA-BN®-HER2-mediated anti-tumor efficacy. Taken together, our data demonstrate that treatment with MVA-BN®-HER2 controls tumor growth through mechanisms including the induction of Th1-biased HER-2-specific immune responses and the control of tumor-mediated immunosuppression.


Subject(s)
Adenocarcinoma/therapy , B-Lymphocyte Subsets/immunology , Cancer Vaccines/pharmacology , Colonic Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Cancer Vaccines/immunology , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunophenotyping , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/pathology
5.
Cancer Res ; 71(15): 5235-44, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21670078

ABSTRACT

MVA-BN-PRO (BN ImmunoTherapeutics) is a candidate immunotherapy product for the treatment of prostate cancer. It encodes 2 tumor-associated antigens, prostate-specific antigen (PSA), and prostatic acid phosphatase (PAP), and is derived from the highly attenuated modified vaccinia Ankara (MVA) virus stock known as MVA-BN. Past work has shown that the immunogenicity of antigens can be improved by targeting their localization to exosomes, which are small, 50- to 100-nm diameter vesicles secreted by most cell types. Exosome targeting is achieved by fusing the antigen to the C1C2 domain of the lactadherin protein. To test whether exosome targeting would improve the immunogenicity of PSA and PAP, 2 additional versions of MVA-BN-PRO were produced, targeting either PSA (MVA-BN-PSA-C1C2) or PAP (MVA-BN-PAP-C1C2) to exosomes, while leaving the second transgene untargeted. Treatment of mice with MVA-BN-PAP-C1C2 led to a striking increase in the immune response against PAP. Anti-PAP antibody titers developed more rapidly and reached levels that were 10- to 100-fold higher than those for mice treated with MVA-BN-PRO. Furthermore, treatment with MVA-BN-PAP-C1C2 increased the frequency of PAP-specific T cells 5-fold compared with mice treated with MVA-BN-PRO. These improvements translated into a greater frequency of tumor rejection in a PAP-expressing solid tumor model. Likewise, treatment with MVA-BN-PSA-C1C2 increased the antigenicity of PSA compared with treatment with MVA-BN-PRO and resulted in a trend of improved antitumor efficacy in a PSA-expressing tumor model. These experiments confirm that targeting antigen localization to exosomes is a viable approach for improving the therapeutic potential of MVA-BN-PRO in humans.


Subject(s)
Adenocarcinoma/immunology , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Exosomes/immunology , Immunotherapy, Active/methods , Prostatic Neoplasms/immunology , Protein Tyrosine Phosphatases/immunology , Acid Phosphatase , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Antigens, Surface/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Drug Delivery Systems , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Milk Proteins/immunology , Milk Proteins/pharmacokinetics , Prostate-Specific Antigen/administration & dosage , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Structure, Tertiary , Th1 Cells/immunology , Vaccines, Attenuated/immunology , Vaccinia virus/immunology , Xenograft Model Antitumor Assays
6.
Mol Imaging ; 3(1): 1-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15142407

ABSTRACT

Radiolabeled annexin V may provide an early indication of the success or failure of anticancer therapy on a patient-by-patient basis as an in vivo marker of tumor cell killing. An important question that remains is when, after initiation of treatment, should annexin V imaging be performed. To address this issue, we obtained simultaneous in vivo measurements of tumor burden and uptake of radiolabeled annexin V in the syngeneic orthotopic murine BCL1 lymphoma model using in vivo bioluminescence imaging (BLI) and small animal single-photon emission computed tomography (SPECT). BCL1 cells labeled for fluorescence and bioluminescence assays (BCL1-gfp/luc) were injected into mice at a dose that leads to progressive disease within two to three weeks. Tumor response was followed by BLI and SPECT before and after treatment with a single dose of 10 mg/kg doxorubicin. Biodistribution analyses revealed a biphasic increase of annexin V uptake within the tumor-bearing tissues of mice. An early peak occurring before actual tumor cells loss was observed between 1 and 5 hr after treatment, and a second longer sustained rise from 9 to 24 hr after therapy, which heralds the onset of tumor cell loss as confirmed by BLI. Multimodality imaging revealed the temporal patterns of tumor cell loss and annexin V uptake revealing a better understanding of the timing of radiolabeled annexin V uptake for its development as a marker of therapeutic efficacy.


Subject(s)
Annexin A5 , Lymphoma, B-Cell/drug therapy , Magnetic Resonance Spectroscopy/methods , Tomography, Emission-Computed, Single-Photon/methods , Animals , Annexin A5/pharmacokinetics , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival , Doxorubicin/toxicity , Female , Green Fluorescent Proteins , Injections, Intravenous , Luciferases/metabolism , Luminescent Measurements , Luminescent Proteins/metabolism , Lymphoma, B-Cell/diagnostic imaging , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Organotechnetium Compounds/pharmacokinetics , Radioactive Tracers , Radiopharmaceuticals , Retroviridae/genetics , Time Factors , Tissue Distribution , Treatment Outcome
7.
Mol Imaging ; 2(4): 313-23, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14717330

ABSTRACT

The lipid bilayer of a cell presents a significant barrier for the delivery of many molecular imaging reagents into cells at target sites in the body. Protein translocation domains (PTDs) are peptides that breach this barrier. Conjugation of PTDs to imaging agents can be utilized to facilitate the delivery of these agents through the cell wall, and in some cases, into the cell nucleus, and have potential for in vitro and in vivo applications. PTD imaging conjugates have included small molecules, peptides, proteins, DNA, metal chelates, and magnetic nanoparticles. The full potential of the use of PTDs in novel in vivo molecular probes is currently under investigation. Cells have been labeled in culture using magnetic nanoparticles derivatized with a PTD and monitored in vivo to assess trafficking patterns relative to cells expressing a target antigen. In vivo imaging of PTD-mediated gene transfer to cells of the skin has been demonstrated in living animals. Here we review several natural and synthetic PTDs that have evolved in the quest for easier translocation across biological barriers and the application of these peptide domains to in vivo delivery of imaging agents.


Subject(s)
Molecular Biology/methods , Protein Structure, Tertiary , Protein Transport/physiology , Animals , Genes, Reporter , Humans , Macromolecular Substances , Proteins/genetics , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...