Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Immunotargets Ther ; 10: 155-169, 2021.
Article in English | MEDLINE | ID: mdl-34079772

ABSTRACT

NHS-IL12 is a novel immunocytokine designed for delivery of IL-12 to the tumor microenvironment (TME). NHS-IL12 consists of two molecules of IL-12 fused to a human IgG1 (NHS76) recognizing DNA/histone complexes, which are often exposed in the necrotic portions of tumors. Preclinical studies demonstrated the tumor-targeting ability and longer plasma half-life for NHS-IL12 when compared with recombinant IL-12 (rIL-12). NHS-IL12 outperformed rIL-12 in enhancing the proliferation and activation of immune as well as antigen-presenting cells, resulting in a more robust primary immune response. NHS-IL12 also reduced the number and function of suppressive myeloid cells (myeloid derived suppressor cells/macrophages) within the TME. In a murine bladder tumor model, NHS-IL12 administration led to a coordinated increase in host immunity with a reduction of immunosuppressive myeloid cells in the TME resulting in substantial reduction in tumor growth. Several preclinical studies have demonstrated increased overall anti-tumor efficacy when NHS-IL12 was combined with either immune-based therapeutics or chemotherapeutic approaches.

2.
Anticancer Res ; 40(10): 5329-5341, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988851

ABSTRACT

Investigation of the efficacy and mechanisms of human immuno-oncology agents has been hampered due to species-specific differences when utilizing preclinical mouse models. Peripheral blood mononuclear cell (PBMC) humanized mice provide a platform for investigating the modulation of the human immune-mediated antitumor response while circumventing the limitations of syngeneic model systems. Use of humanized mice has been stymied by model-specific limitations, some of which include the development of graft versus host disease, technical difficulty and cost associated with each humanized animal, and insufficient engraftment of some human immune subsets. Recent advances have addressed many of these limitations from which have emerged humanized models that are more clinically relevant. This review characterizes the expanded usage, advantages and limitations of humanized mice and provides insights into the development of the next generation of murine humanized models to further inform clinical applications of cancer immunotherapeutic agents.


Subject(s)
Immunity, Cellular/drug effects , Immunotherapy , Leukocytes, Mononuclear/immunology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Disease Models, Animal , Humans , Leukocytes, Mononuclear/drug effects , Mice , Neoplasms/immunology , Neoplasms/pathology
3.
Front Oncol ; 10: 549, 2020.
Article in English | MEDLINE | ID: mdl-32373533

ABSTRACT

The lack of serial biopsies in patients with a range of carcinomas has been one obstacle in our understanding of the mechanism of action of immuno-oncology agents as well as the elucidation of mechanisms of resistance to these novel therapeutics. While much information can be obtained from studies conducted with syngeneic mouse models, these models have limitations, including that both tumor and immune cells being targeted are murine and that many of the immuno-oncology agents being evaluated are human proteins, and thus multiple administrations are hampered by host xenogeneic responses. Some of these limitations are being overcome by the use of humanized mouse models where human peripheral blood mononuclear cells (PBMC) are engrafted into immunosuppressed mouse strains. Bintrafusp alfa (M7824) is an innovative first-in-class bifunctional fusion protein composed of the extracellular domain of the TGF-ßRII to function as a TGF-ß "trap" fused to a human IgG1 antibody blocking PD-L1. A phase I clinical trial of bintrafusp alfa showed promising anti-tumor efficacy in heavily pretreated advanced solid tumors, and multiple clinical studies are currently ongoing. There is still much to learn regarding the mechanism of action of bintrafusp alfa, including its effects on both human immune cells in the periphery and in the tumor microenvironment (TME), and any temporal effects upon multiple administrations. By using the NSG-ß2m-/- mouse strain humanized with PBMC, we demonstrate here for the first time: (a) the effects of bintrafusp alfa administration on human immune cells in the periphery vs. the TME using three different human xenograft models; (b) temporal effects upon multiple administrations of bintrafusp alfa; (c) phenotypic changes induced in the TME, and (d) variations observed in the use of multiple different PBMC donors. Also discussed are the similarities and differences in the data thus far obtained employing murine syngeneic models, from clinical trials, and in the use of this humanized mouse model. The results described here may guide the future use of this agent or similar immunotherapy agents as monotherapies or in combination therapy studies.

4.
J Immunother Cancer ; 7(1): 150, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186063

ABSTRACT

BACKGROUND: While significant strides in the treatment of metastatic bladder cancer have been made with immune checkpoint inhibitors, the treatment of carcinoma in situ and non-muscle invasive, non-metastatic (superficial) human urothelial carcinoma, also termed non-muscle invasive bladder cancer (NMIBC), remains intractable with bacillus Calmette-Guerin (BCG) employed as the standard of care. In this study, an immunocytokine, NHS-muIL12, which consists of two molecules of murine IL-12 fused to NHS76, a tumor necrosis-targeting human IgG1, was examined as an immunotherapeutic in an orthotopic MB49luc bladder tumor model. METHODS: The antitumor activity of systemic administration of NHS-muIL12 was investigated on MB49luc tumors, an aggressive, bioluminescent orthotopic bladder cancer model. Temporal studies were carried out on MB49luc bladder tumors harvested during various time points during NHS-muIL12 treatment and cellular changes associated with the reduction in tumor burden following NHS-muIL12 were determined by flow cytometry. Effects of those changes on the proliferation/activation of lymphoid cells were also determined. RESULTS: Studies revealed a significant reduction in MB49luc bladder tumor burden occurring between days 3 and 6 after the third and final systemic administration of NHS-muIL12. Temporal analyses of the MB49luc bladder tumor microenvironment (TME) initially revealed a large accumulation of myeloid-derived suppressor cells (MDSCs) and macrophages that elicited potent immunosuppression. Immunosuppression was characterized by the inability of CD4+ and CD8+ T cells to respond to broad-based immune stimulants. NHS-muIL12 administration resulted in temporal-dependent reductions in the number of MDSCs, macrophages and tumor-associated TGF-ß, which culminated in a re-ignition of CD4+ and CD8+ T cells to elicit potent antitumor responses against MB49luc bladder tumors. CONCLUSIONS: These findings provide strong evidence that the systemic administration of an immunocytokine consisting of a tumor-targeting Ig through recognition of DNA and DNA-histone complexes coupled to muIL-12 can effectively target the bladder TME; this significantly reduces the myeloid cellular compartment and reverts an immunosuppressive to an immunopermissive TME, ultimately resulting in antitumor effects. These studies provide further rationale for the employment of NHS-IL12 as an immunomodulator and clinical immunotherapeutic for NMIBC.


Subject(s)
Immunoglobulin G/genetics , Immunotherapy/methods , Interleukin-12/genetics , Recombinant Fusion Proteins/genetics , Urinary Bladder Neoplasms/drug therapy , Animals , Disease Models, Animal , Female , Humans , Mice , Tumor Microenvironment
5.
Bladder Cancer ; 4(4): 429-440, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30417054

ABSTRACT

The Fourth Annual Albert Institute Bladder Cancer Care and Research Symposium was held from September 14th-16th in Houston, Texas. The symposium covered a range of topics relevant to bladder cancer, including basic science aspects of immunology and immunotherapy that inform clinical management; intravesical therapy for non-muscle invasive disease; understanding the nuances of carcinoma in situ; and optimizing patient care and outcomes following therapy. The moving landscape of bladder cancer from an industry perspective was also discussed. In the following sections we discuss intrinsic and extrinsic factors, including the immune microenvironment and sex bias, in the context of bladder cancer; how these influence tumor development, progression, and treatment strategies; and how the interpretation of immune features in relation to molecular subtypes informs both treatment decisions and response. We conclude with a summary of key points that will need to be addressed to ensure best use of new knowledge in this area for improved clinical management of patients with bladder cancer.

6.
Oncotarget ; 8(43): 73469-73482, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088720

ABSTRACT

Immunotherapy was significantly enhanced in a murine tumor model by combining a vaccine with a fusion protein designed to target the glucocorticoid-induced tumor necrosis factor (TNF) receptor related gene (GITR) on the surface of T cells. The recombinant poxvirus-based vaccine platform included Modified Vaccinia virus Ankara (rMVA) and fowlpox (rF) vectors as the driver immunogens both engineered to express the human carcinoembryonic antigen (CEA) and three murine costimulatory molecules B7.1, ICAM-1, LFA-3 (designated TRICOM). In previous studies, mice expressing human CEA as a transgene (CEA.Tg mice) vaccinated with rMVA/rF-CEA-TRICOM overcame CEA immune tolerance by inducing anti-CEA‒specific immunity and regression of CEA-expressing tumors. The murine GITR ligand fusion protein (mGITRL-FP) consisted of a mouse IgG2a Fc region, a yeast-derived coiled GCN4 pII and the extracellular GITR-binding domain of murine GITR ligand. The design maximized valency and the potential to agonize the GITR receptor. Combined treatment of the vaccine and mGITRL-FP mediated a more robust tumor regression, leading to sustained improvement in overall survival. The enhanced immunotherapeutic effect was linked to the generation of a strong CD8+ T cell antitumor immune response. A treatment schedule with mGITRL-FP administered prior to the priming rMVA-CEA-TRICOM vaccination was of paramount importance. The mechanism of action for the enhanced antitumor effects resided in the depletion of immune cells, particularly FoxP3+ regulatory T cells, that express high GITR levels following activation. The results provide evidence that targeting GITR with mGITRL-FP in concert with a cancer vaccine represents a potential novel approach to more effective immunotherapy.

7.
Oncotarget ; 8(13): 20558-20571, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423552

ABSTRACT

The combined therapeutic potential of an immunocytokine designed to deliver IL-12 to the necrotic regions of solid tumors with an anti-PD-L1 antibody that disrupts the immunosuppressive PD-1/PD-L1 axis yielded a combinatorial benefit in multiple murine tumor models. The murine version of the immunocytokine, NHS-muIL12, consists of an antibody (NHS76) recognizing DNA/DNA-histone complexes, fused with two molecules of murine IL-12 (NHS-muIL12). By its recognition of exposed DNA, NHS-muIL12 targets IL-12 to the necrotic portions of tumors; it has a longer plasma half-life and better antitumor efficacy against murine tumors than recombinant murine IL-12. It is shown here that NHS-muIL12, in an IFN-γ‒dependent mechanism, upregulates mPD-L1 expression on mouse tumors, which could be construed as an immunosuppressive action. Yet concurrent therapy with NHS-muIL12 and an anti-PD-L1 antibody resulted in additive/synergistic antitumor effects in PD-L1‒expressing subcutaneously transplanted tumors (MC38, MB49) and in an intravesical bladder tumor model (MB49). Antitumor efficacy correlated with (a) with a higher frequency of tumor antigen-specific splenic CD8+ T cells and (b) enhanced T cell activation over a wide range of NHS-muIL12 concentrations. These findings suggest that combining NHS-muIL12 and an anti-PD-L1 antibody enhances T cell activation and T cell effector functions within the tumor microenvironment, significantly improving overall tumor regression. These results should provide the rationale to examine the combination of these agents in clinical studies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunotherapy/methods , Neoplasms, Experimental/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Disease Models, Animal , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Immunohistochemistry , Interleukin-12/immunology , Interleukin-12/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
8.
Oncotarget ; 7(52): 86359-86373, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27861156

ABSTRACT

Natural killer (NK) cells are known to play a role in mediating innate immunity, in enhancing adaptive immune responses, and have been implicated in mediating anti-tumor responses via antibody-dependent cell-mediated cytotoxicity (ADCC) by reactivity of CD16 with the Fc region of human IgG1 antibodies. The NK-92 cell line, derived from a lymphoma patient, has previously been well characterized and adoptive transfer of irradiated NK-92 cells has demonstrated safety and shown preliminary evidence of clinical benefit in cancer patients. The NK-92 cell line, devoid of CD16, has now been engineered to express the high affinity (ha) CD16 V158 FcγRIIIa receptor, as well as engineered to express IL-2; IL-2 has been shown to replenish the granular stock of NK cells, leading to enhanced perforin- and granzyme-mediated lysis of tumor cells. The studies reported here show high levels of granzyme in haNK cells, and demonstrate the effects of irradiation of haNK cells on multiple phenotypic markers, viability, IL-2 production, and lysis of a spectrum of human tumor cells. Studies also compare endogenous irradiated haNK lysis of tumor cells with that of irradiated haNK-mediated ADCC using cetuximab, trastuzumab and pertuzumab monoclonal antibodies. These studies thus provide the rationale for the potential use of irradiated haNK cells in adoptive transfer studies for a range of human tumor types. Moreover, since only approximately 10% of humans are homozygous for the high affinity V CD16 allele, these studies also provide the rationale for the use of irradiated haNK cells in combination with IgG1 anti-tumor monoclonal antibodies.


Subject(s)
Adoptive Transfer , Granzymes/immunology , Killer Cells, Natural/immunology , Receptors, IgG/genetics , Animals , Antibody-Dependent Cell Cytotoxicity , B7-H1 Antigen/analysis , Cell Line, Tumor , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Genetic Engineering , Humans , Immunoglobulin G/therapeutic use , Interleukin-2/genetics , Killer Cells, Natural/enzymology , Killer Cells, Natural/radiation effects , Male , Mice , Middle Aged , Receptors, IgG/immunology
9.
Cancer Immunol Res ; 4(5): 452-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26921031

ABSTRACT

Bacillus Calmette-Guerin (BCG) is the standard of care for intravesical therapy for carcinoma in situ and non-muscle invasive, nonmetastatic human urothelial carcinoma. Although the responsiveness to this immunotherapeutic is believed to be linked with (i) a high number of somatic mutations and (ii) a large number of tumor-infiltrating lymphocytes, recent findings of the roles that inhibitory immune receptors and their ligands play in tumor evasion may provide insights into the limitations of the effectiveness of BCG and offer new targets for immune-based therapy. In this study, an aggressive, bioluminescent orthotopic bladder cancer model, MB49 tumor cells transfected with luciferase (MB49(luc)), was used to study the antitumor effects of avelumab, an antibody to PD-L1. MB49(luc) murine tumor cells form multifocal tumors on the mucosal wall of the bladder reminiscent of non-muscle invasive, nonmetastatic urothelial carcinomas. MB49(luc) bladder tumors are highly positive for the expression of PD-L1, and avelumab administration induced significant (P < 0.05) antitumor effects. These antitumor effects were more dependent on the presence of CD4 than CD8 T cells, as determined by in vivo immune cell depletions. The findings suggest that in this bladder tumor model, interruption of the immune-suppressive PD-1/PD-L1 complex releases a local adaptive immune response that, in turn, reduces tumor growth. This bladder tumor model can be used to further identify host antitumor immune mechanisms and evaluate combinations of immune-based therapies for carcinoma in situ and non-muscle invasive, nonmetastatic urothelial carcinoma, to provide the rationale for subsequent clinical studies. Cancer Immunol Res; 4(5); 452-62. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Transitional Cell/drug therapy , Urinary Bladder Neoplasms/drug therapy , Adaptive Immunity , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Immune Tolerance/immunology , Immunotherapy/methods , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Neoplasm Transplantation , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
10.
Cancer Immunol Immunother ; 63(12): 1307-17, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25186612

ABSTRACT

The transcription factor brachyury is a major driver of epithelial to mesenchymal transition in human carcinoma cells. It is overexpressed in several human tumor types versus normal adult tissues, except for testes and thyroid. Overexpression is associated with drug resistance and poor prognosis. Previous studies identified a brachyury HLA-A2 cytotoxic T-lymphocyte epitope. The studies reported here describe an enhancer epitope of brachyury. Compared to the native epitope, the agonist epitope: (a) has enhanced binding to MHC class I, (b) increased the IFN-γ production from brachyury-specific T cells, (c) generated brachyury-specific T cells with greater levels of perforin and increased proliferation, (d) generated T cells more proficient at lysing human carcinoma cells endogenously expressing the native epitope, and (e) achieved greater brachyury-specific T-cell responses in vivo in HLA-A2 transgenic mice. These studies also report the generation of a heat-killed recombinant Saccharomyces cerevisiae (yeast) vector expressing the full-length brachyury gene encoding the agonist epitope. Compared to yeast-brachyury (native) devoid of the agonist epitope, the yeast-brachyury (agonist) enhanced the activation of brachyury-specific T cells, which efficiently lysed human carcinoma cells. In addition to providing the rationale for the recombinant yeast-brachyury (agonist) as a potential vaccine in cancer therapy, these studies also provide the rationale for the use of the agonist in (a) dendritic cell (DC) vaccines, (b) adjuvant or liposomal vaccines, (c) recombinant viral and/or bacterial vaccines, (d) protein/polypeptide vaccines, (e) activation of T cells ex vivo in adoptive therapy protocols, and (f) generation of genetically engineered targeted T cells.


Subject(s)
Epithelial-Mesenchymal Transition/immunology , Epitopes, T-Lymphocyte/immunology , Fetal Proteins/immunology , Neoplasms/immunology , T-Box Domain Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy
11.
Adv Cancer Res ; 121: 67-124, 2014.
Article in English | MEDLINE | ID: mdl-24889529

ABSTRACT

Therapeutic cancer vaccines have the potential of being integrated in the therapy of numerous cancer types and stages. The wide spectrum of vaccine platforms and vaccine targets is reviewed along with the potential for development of vaccines to target cancer cell "stemness," the epithelial-to-mesenchymal transition (EMT) phenotype, and drug-resistant populations. Preclinical and recent clinical studies are now revealing how vaccines can optimally be used with other immune-based therapies such as checkpoint inhibitors, and so-called nonimmune-based therapeutics, radiation, hormonal therapy, and certain small molecule targeted therapies; it is now being revealed that many of these traditional therapies can lyse tumor cells in a manner as to further potentiate the host immune response, alter the phenotype of nonlysed tumor cells to render them more susceptible to T-cell lysis, and/or shift the balance of effector:regulatory cells in a manner to enhance vaccine efficacy. The importance of the tumor microenvironment, the appropriate patient population, and clinical trial endpoints is also discussed in the context of optimizing patient benefit from vaccine-mediated therapy.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/therapy , Animals , Biomarkers, Tumor/physiology , Clinical Trials as Topic , Combined Modality Therapy , Disease Progression , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Neoplasms/pathology
12.
Oncotarget ; 5(7): 1869-84, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24681847

ABSTRACT

Targeted delivery of IL-12 might turn this cytokine into a safer, more effective cancer therapeutic. Here we describe a novel immunocytokine, NHS-IL12, consisting of two molecules of IL-12 fused to a tumor necrosis-targeting human IgG1 (NHS76). The addition of the human IgG1 moiety resulted in a longer plasma half-life of NHS-IL12 than recombinant IL-12, and a selective targeting to murine tumors in vivo. Data from both in vitro assays using human PBMCs and in vivo primate studies showed that IFN-gamma production by immune cells is attenuated following treatment with the immunocytokine, suggesting an improved toxicity profile than seen with recombinant IL-12 alone. NHS-IL12 was superior to recombinant IL-12 when evaluated as an anti-tumor agent in three murine tumor models. Mechanistic studies utilizing immune cell subset-depleting antibodies, flow cytometric methods, and in vitro cytotoxicity and ELISA assays all indicated that the anti-tumor effects of NHS-IL12 were primarily CD8+ T cell-dependent and likely IL-12-mediated. Combining NHS-IL12 treatment with a cancer vaccine, radiation, or chemotherapy resulted in greater anti-tumor effects than each individual therapy alone. These preclinical findings provide a rationale for the clinical testing of this immunocytokine, both as a single agent and in combination with vaccines, radiation and chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immunoglobulin G/pharmacology , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD8-Positive T-Lymphocytes , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Docetaxel , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Immunomodulation , Indoles/administration & dosage , Interleukin-12/immunology , Interleukin-12/therapeutic use , Lymphocyte Activation/drug effects , Macaca fascicularis , Membrane Glycoproteins/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Engineering , Pyrroles/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Sunitinib , Survival Rate , Taxoids/administration & dosage , Tumor Burden
13.
J Immunol ; 191(5): 2299-307, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23904158

ABSTRACT

This study affirmed that isolated CD8(+) T cells express mRNA and produce TGF-ß following cognate peptide recognition. Blockage of endogenous TGF-ß with either a TGF-ß-blocking Ab or a small molecule inhibitor of TGF-ßRI enhances the generation of CD62L(high)/CD44(high) central memory CD8(+) T cells accompanied with a robust recall response. Interestingly, the augmentation within the central memory T cell pool occurs in lieu of cellular proliferation or activation, but with the expected increase in the ratio of the Eomesoderm/T-bet transcriptional factors. Yet, the signal transduction pathway(s) seems to be noncanonical, independent of SMAD or mammalian target of rapamycin signaling. Enhancement of central memory generation by TGF-ß blockade is also confirmed in human PBMCs. The findings underscore the role(s) that autocrine TGF-ß plays in T cell homeostasis and, in particular, the balance of effector/memory and central/memory T cells. These results may provide a rationale to targeting TGF-ß signaling to enhance Ag-specific CD8(+) T cell memory against a lethal infection or cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Transforming Growth Factor beta1/immunology , Adoptive Transfer , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
14.
J Immunol ; 188(9): 4323-33, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450814

ABSTRACT

Immunologic memory involving CD8(+) T cells is a hallmark of an adaptive Ag-specific immune response and constitutes a critical component of protective immunity. Designing approaches that enhance long-term T cell memory would, for the most part, fortify vaccines and enhance host protection against infectious diseases and, perhaps, cancer immunotherapy. A better understanding of the cellular programs involved in the Ag-specific T cell response has led to new approaches that target the magnitude and quality of the memory T cell response. In this article, we show that T cells from TCR transgenic mice for the nucleoprotein of influenza virus NP68 exhibit the distinct phases--priming, expansion, contraction, and memory--of an Ag-specific T cell response when exposed in vitro to the cognate peptide. Saracatinib, a specific inhibitor of Src family kinases, administered at low doses during the expansion or contraction phases, increased CD62L(high)/CD44(high) central memory CD8(+) T cells and IFN-γ production but suppressed immunity when added during the priming phase. These effects by saracatinib were not accompanied by the expected decline of Src family kinases but were accompanied by Akt-mammalian target of rapamycin suppression and/or mediated via another pathway. Increased central memory cells by saracatinib were recapitulated in mice using a poxvirus-based influenza vaccine, thus underscoring the importance of dose and timing of the inhibitor in the context of memory T cell differentiation. Finally, vaccine plus saracatinib treatment showed better protection against tumor challenge. The immune-potentiating effects on CD8(+) T cells by a low dose of saracatinib might afford better protection from pathogens or cancer when combined with vaccine.


Subject(s)
Benzodioxoles/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Immunologic Memory/drug effects , Quinazolines/pharmacology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/enzymology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Immunologic Memory/genetics , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/metabolism , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/immunology , src-Family Kinases/metabolism
15.
Biomaterials ; 32(3): 926-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20965561

ABSTRACT

Vaccines based on recombinant proteins require adjuvant systems in order to generate Th1-type immune responses. We have developed a vaccine adjuvant system using a viscous chitosan solution and interleukin (IL)-12, a Th1-inducing cytokine. The chitosan solution is designed to create a depot of antigen and IL-12 at a subcutaneous injection site. We measured the in vivo immune response of a vaccine containing 0.25, 1, or 4 µg murine IL-12 and 75 µg ovalbumin (OVA), formulated in a 1.5% chitosan glutamate solution. The chitosan/IL-12/OVA vaccine, in comparison to chitosan/OVA, IL-12/OVA, or OVA alone, elicited greater antigen-specific CD4(+) and CD8(+) T-cell responses, as determined by CD4(+) splenocyte proliferation, Th1 cytokine release, CD8(+) T-cell interferon-γ release, and MHC class I peptide pentamer staining. The combination of chitosan and IL-12 also enhanced IgG2a and IgG2b antibody responses to OVA. Co-formulation of chitosan and IL-12 thus promoted the generation of a Th1 immune response to a model protein vaccine.


Subject(s)
Adjuvants, Immunologic/metabolism , Chitosan/immunology , Interleukin-12/immunology , Proteins/immunology , Vaccines/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL
16.
J Immunother ; 33(7): 697-705, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664357

ABSTRACT

IL-12 is a potent antitumor cytokine that exhibits significant clinical toxicities after systemic administration. We hypothesized that intratumoral (i.t.) administration of IL-12 coformulated with the biodegradable polysaccharide chitosan could enhance the antitumor activity of IL-12 while limiting its systemic toxicity. Noninvasive imaging studies monitored local retention of IL-12, with and without chitosan coformulation, after i.t. injection. Antitumor efficacy of IL-12 alone and IL-12 coformulated with chitosan (chitosan/IL-12) was assessed in mice bearing established colorectal (MC32a) and pancreatic (Panc02) tumors. Additional studies involving depletion of immune cell subsets, tumor rechallenge, and CTL activity were designed to elucidate mechanisms of regression and tumor-specific immunity. Coformulation with chitosan increased local IL-12 retention from 1 to 2 days to 5 to 6 days. Weekly i.t. injections of IL-12 alone eradicated ≤10% of established MC32a and Panc02 tumors, while i.t. chitosan/IL-12 immunotherapy caused complete tumor regression in 80% to 100% of mice. Depletion of CD4(+) or Gr-1(+) cells had no impact on chitosan/IL-12-mediated tumor regression. However, CD8(+) or NK cell depletion completely abrogated antitumor activity. I.t. chitosan/IL-12 immunotherapy generated systemic tumor-specific immunity, as >80% of mice cured with i.t. chitosan/IL-12 immunotherapy were at least partially protected from tumor rechallenge. Furthermore, CTLs from spleens of cured mice lysed MC32a and gp70 peptide-loaded targets. Chitosan/IL-12 immunotherapy increased local retention of IL-12 in the tumor microenvironment, eradicated established, aggressive murine tumors, and generated systemic tumor-specific protective immunity. Chitosan/IL-12 is a well-tolerated, effective immunotherapy with considerable potential for clinical translation.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Chitosan/administration & dosage , Colorectal Neoplasms/therapy , Immunotherapy , Pancreatic Neoplasms/therapy , Animals , Carcinoembryonic Antigen/genetics , Cells, Cultured , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic/drug effects , Immunologic Memory , Interleukin-12/administration & dosage , Interleukin-12/adverse effects , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , Remission Induction , Tumor Burden/drug effects
17.
Cancer Res ; 69(15): 6192-9, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638573

ABSTRACT

Intravesical BCG has been used successfully to treat superficial bladder cancer for three decades. However, 20% to 30% of patients will fail initial BCG therapy and 30% to 50% of patients will develop recurrent tumors within 5 years. Alternative or complementary strategies for the management of superficial bladder cancer are needed. Interleukin-12 (IL-12) is a potent T(H)1 cytokine with robust antitumor activity and the ability to potentiate immunologic memory. Unfortunately, intravesical IL-12 did not show antitumor efficacy in a recent clinical study of patients with recurrent superficial bladder cancer. We hypothesized that coformulation of IL-12 with chitosan, a biocompatible, mucoadhesive polysaccharide, could improve intravesical IL-12 delivery and provide an effective and durable alternative for the treatment of superficial bladder cancer. In antitumor studies, 88% to 100% of mice bearing orthotopic bladder tumors were cured after four intravesical treatments with chitosan/IL-12. In contrast, only 38% to 60% of mice treated with IL-12 alone and 0% treated with BCG were cured. Antitumor responses following chitosan/IL-12 treatments were durable and provided complete protection from intravesical tumor rechallenge. Urinary cytokine analysis showed that chitosan/IL-12 induced multiple T(H)1 cytokines at levels significantly higher than either IL-12 alone or BCG. Immunohistochemistry revealed moderate to intense tumor infiltration by T cells and macrophages following chitosan/IL-12 treatments. Bladder submucosa from cured mice contained residual populations of immune cells that returned to baseline levels after several months. Intravesical chitosan/IL-12 is a well-tolerated, effective immunotherapy that deserves further consideration for testing in humans for the management of superficial bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/therapy , Chitosan/administration & dosage , Interleukin-12/administration & dosage , Urinary Bladder Neoplasms/therapy , Administration, Intravesical , Animals , BCG Vaccine/administration & dosage , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/immunology , Cell Line, Tumor , Female , Immunohistochemistry , Interferon-gamma/blood , Interferon-gamma/urine , Interleukin-12/blood , Interleukin-12/urine , Luciferases/biosynthesis , Luciferases/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
18.
Clin Cancer Res ; 15(7): 2387-96, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276249

ABSTRACT

PURPOSE: IFN-alpha is a pleiotropic cytokine possessing immunomodulatory properties that may improve the efficacy of therapeutic cancer vaccines. The aim of this study was to evaluate the effectiveness and compatibility of combining recombinant IFN-alpha with poxvirus vaccines targeting the human carcinoembryonic antigen (CEA) in murine models of colorectal and pancreatic adenocarcinomas, where CEA is a self-antigen. EXPERIMENTAL DESIGN: The phenotypic and functional effects of IFN-alpha were evaluated in the draining inguinal lymph nodes of tumor-free mice. We studied the effect of the site of IFN-alpha administration (local versus distal) on antigen-specific immune responses to poxvirus vaccination. Mechanistic studies were conducted to assess the efficacy of IFN-alpha and CEA-directed poxvirus vaccines in tumor-bearing CEA transgenic mice. RESULTS: We identified a dose and schedule of IFN-alpha that induced a locoregional expansion of the draining inguinal lymph nodes and improved cellular cytotoxicity (natural killer and CD8(+)) and antigen presentation. Suppression of the vaccinia virus was avoided by administering IFN-alpha distal to the site of vaccination. The combination of IFN-alpha and vaccine inhibited tumor growth, improved survival, and elicited CEA-specific CTL responses in mice with CEA(+) adenocarcinomas. In mice with pancreatic tumors, IFN-alpha slowed tumor growth, induced CTL activity, and increased CD8(+) tumor-infiltrating lymphocytes. CONCLUSIONS: These data suggest that IFN-alpha can be used as a biological response modifier with antigen-directed poxvirus vaccines to yield significant therapeutic antitumor immune responses. This study provides the rationale and mechanistic insights to support a clinical trial of this immunotherapeutic strategy in patients with CEA-expressing carcinomas.


Subject(s)
Adenocarcinoma/drug therapy , Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Fowlpox virus/genetics , Interferon-alpha/therapeutic use , Vaccinia virus/genetics , Adenocarcinoma/immunology , Animals , Cancer Vaccines/genetics , Carcinoembryonic Antigen/genetics , Cell Line, Tumor , Combined Modality Therapy , DNA, Recombinant/analysis , Female , Histocompatibility Antigens Class I/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
19.
Vaccine ; 26(42): 5407-15, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18706954

ABSTRACT

Regular moderate exercise has been proposed to enhance immune function, but its effects on immunity and their consequences have not been well studied. Mice without (AL) or with access (AL+EX) to voluntary running wheels were vaccinated with a model antigen (ovalbumin (OVA)) via intranasal or subcutaneous routes to target the mucosal and systemic immune compartments, respectively. EX enhanced OVA-specific CD4(+) T cell cytokine production and proliferation in all lymphoid organs examined without changes in cell distribution in any organ. These results suggest that coupling moderate exercise with vaccination may enhance vaccine efficacy for the prevention and/or therapy of numerous diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Motor Activity , Vaccination , Animals , Body Composition , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Substrate Specificity
20.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594015

ABSTRACT

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Subject(s)
Antigens, Neoplasm/chemistry , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/chemistry , Gene Expression Regulation , Immunotherapy/methods , Saccharomyces cerevisiae/metabolism , Vaccines, DNA/chemistry , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Cell Proliferation , Female , Humans , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Recombinant Proteins/chemistry , Vaccines, DNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...