Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Natl Cancer Inst ; 115(11): 1404-1419, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37195421

ABSTRACT

BACKGROUND: We investigated the role of A2B-adenosine receptor in regulating immunosuppressive metabolic stress in the tumor microenvironment. Novel A2B-adenosine receptor antagonist PBF-1129 was tested for antitumor activity in mice and evaluated for safety and immunologic efficacy in a phase I clinical trial of patients with non-small cell lung cancer. METHODS: The antitumor efficacy of A2B-adenosine receptor antagonists and their impact on the metabolic and immune tumor microenvironment were evaluated in lung, melanoma, colon, breast, and epidermal growth factor receptor-inducible transgenic cancer models. Employing electron paramagnetic resonance, we assessed changes in tumor microenvironment metabolic parameters, including pO2, pH, and inorganic phosphate, during tumor growth and evaluated the immunologic effects of PBF-1129, including its pharmacokinetics, safety, and toxicity, in patients with non-small cell lung cancer. RESULTS: Levels of metabolic stress correlated with tumor growth, metastasis, and immunosuppression. Tumor interstitial inorganic phosphate emerged as a correlative and cumulative measure of tumor microenvironment stress and immunosuppression. A2B-adenosine receptor inhibition alleviated metabolic stress, downregulated expression of adenosine-generating ectonucleotidases, increased expression of adenosine deaminase, decreased tumor growth and metastasis, increased interferon γ production, and enhanced the efficacy of antitumor therapies following combination regimens in animal models (anti-programmed cell death 1 protein vs anti-programmed cell death 1 protein plus PBF-1129 treatment hazard ratio = 11.74 [95% confidence interval = 3.35 to 41.13], n = 10, P < .001, 2-sided F test). In patients with non-small cell lung cancer, PBF-1129 was well tolerated, with no dose-limiting toxicities; demonstrated pharmacologic efficacy; modulated the adenosine generation system; and improved antitumor immunity. CONCLUSIONS: Data identify A2B-adenosine receptor as a valuable therapeutic target to modify metabolic and immune tumor microenvironment to reduce immunosuppression, enhance the efficacy of immunotherapies, and support clinical application of PBF-1129 in combination therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Receptor, Adenosine A2B/metabolism , Tumor Microenvironment , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Immunosuppression Therapy , Adenosine/metabolism , Phosphates , Cell Line, Tumor
3.
Cell Rep ; 35(1): 108944, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826903

ABSTRACT

Inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) delay progression of metastatic breast cancer. However, complete responses are uncommon and tumors eventually relapse. Here, we show that CDK4/6i can enhance efficacy of T cell-based therapies, such as adoptive T cell transfer or T cell-activating antibodies anti-OX40/anti-4-1BB, in murine breast cancer models. This effect is driven by the induction of chemokines CCL5, CXCL9, and CXCL10 in CDK4/6i-treated tumor cells facilitating recruitment of activated CD8+ T cells, but not Tregs, into the tumor. Mechanistically, chemokine induction is associated with metabolic stress that CDK4/6i treatment induces in breast cancer cells. Despite the cell cycle arrest, CDK4/6i-treated cells retain high metabolic activity driven by deregulated PI3K/mTOR pathway. This causes cell hypertrophy and increases mitochondrial content/activity associated with oxidative stress and inflammatory stress response. Our findings uncover a link between tumor metabolic vulnerabilities and anti-tumor immunity and support further development of CDK4/6i and immunotherapy combinations.


Subject(s)
Chemokines/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Mammary Neoplasms, Animal/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Female , Humans , Hypertrophy , Immunotherapy , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Prognosis , Reactive Oxygen Species/metabolism , Receptors, Chemokine/metabolism , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/metabolism
4.
Front Immunol ; 10: 1906, 2019.
Article in English | MEDLINE | ID: mdl-31456803

ABSTRACT

Multiple effector layers in the immune system ensure an optimal temporal and spatial distribution of immune defense. Cytotoxic innate lymphoid natural killers (NK) and adaptive CD8+ T lymphocytes (CTL) interact to elicit specific cytolytic outcomes. The CTL carry antigen-specific T cell receptors (TCR) to recognize cognate peptides bound with major histocompatibility complex class-I (MHC-I) or human leukocyte antigen (HLA) molecules on target cells. Upon TCR engagement with MHC-I:peptide at a threshold of avidity, T cell intracellular programs converge into cytolytic activity. By contrast, NK cells lack antigen-specific receptors but express a repertoire of highly polymorphic and polygenic inhibitory and activating receptors that bind various ligands including MHC and like molecules. A highly calibrated maturation enables NK cells to eliminate target cells with lowered or absent MHC-I or induced MHC-I-related molecules while maintaining their tolerance toward self-MHC. Both CTL and mature NK cells undergo membranous reorganization and express various effector molecules to eliminate aberrant cells undergoing a stress of transformation, infection or other pathological noxa. Here, we present the cellular modules that underlie the CTL-NK circuitry to maximize their effector cooperativity against stressed or cancerous cells.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , Cell Communication , Humans , Neoplasms/immunology
6.
J Immunother Cancer ; 7(1): 95, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940183

ABSTRACT

BACKGROUND: Notch intercellular communication instructs tissue-specific T-cell development and function. In this study, we explored the roles of dendritic cell (DC)-expressed Notch ligands in the regulation of T-cell effector function. METHODS: We generated mice with CD11c lineage-specific deletion of Notch Delta-like ligand (Dll)1 and Jagged (Jag)2. Using these genetically-ablated mice and engineered pharmacological Notch ligand constructs, the roles of various Delta-like and Jagged ligands in the regulation of T-cell-mediated immunity were investigated. We assessed tumor growth, mouse survival, cytokine production, immunophenotyping of myeloid and lymphoid populations infiltrating the tumors, expression of checkpoint molecules and T-cell function in the experimental settings of murine lung and pancreatic tumors and cardiac allograft rejection. Correlative studies were also performed for the expression of NOTCH ligands, NOTCH receptors and PD-1 on various subsets of myeloid and lymphoid cells in tumor-infiltrating immune cells analyzed from primary human lung cancers. RESULTS: Mice with CD11c lineage-specific deletion of Notch ligand gene Dll1, but not Jag2, exhibited accelerated growth of lung and pancreatic tumors concomitant with decreased antigen-specific CD8+T-cell functions and effector-memory (Tem) differentiation. Increased IL-4 but decreased IFN-γ production and elevated populations of T-regulatory and myeloid-derived suppressor cells were observed in Dll1-ablated mice. Multivalent clustered DLL1-triggered Notch signaling overcame DC Dll1 deficiency and improved anti-tumor T-cell responses, whereas the pharmacological interference by monomeric soluble DLL1 construct suppressed the rejection of mouse tumors and cardiac allograft. Moreover, monomeric soluble JAG1 treatment reduced T-regulatory cells and improved anti-tumor immune responses by decreasing the expression of PD-1 on CD8+Tem cells. A significant correlation was observed between DC-expressed Jagged and Delta-like ligands with Tem-expressed PD-1 and Notch receptors, respectively, in human lung tumor-infiltrates. CONCLUSION: Our data show the importance of specific expression of Notch ligands on DCs in the regulation of T-cell effector function. Thus, strategies incorporating selectively engineered Notch ligands could provide a novel approach of therapeutics for modulating immunity in various immunosuppressive conditions including cancer.


Subject(s)
Calcium-Binding Proteins/metabolism , Dendritic Cells/metabolism , Jagged-2 Protein/metabolism , Lung Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , 3T3 Cells , Animals , Calcium-Binding Proteins/agonists , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Communication/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Graft Rejection/immunology , Heart Transplantation/adverse effects , Humans , Jagged-2 Protein/agonists , Jagged-2 Protein/antagonists & inhibitors , Jagged-2 Protein/genetics , Lung/immunology , Lung/pathology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
7.
Oncotarget ; 8(5): 8604-8621, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28052005

ABSTRACT

Tumor-induced immune tolerance poses a major challenge for therapeutic interventions aimed to manage cancer. We explored approaches to overcome T-cell suppression in murine breast and kidney adenocarcinomas, and lung fibrosarcoma expressing immunogenic antigens. We observed that treatment with a reversible proteasome inhibitor bortezomib (1 mg/kg body weight) in tumor-bearing mice significantly enhanced the expression of lymphocyte-stimulatory cytokines IL-2, IL-12, and IL-15. Notably, bortezomib administration reduced pulmonary nodules of mammary adenocarcinoma 4T1.2 expressing hemagglutinin (HA) model antigen (4T1HA) in mice. Neutralization of IL-12 and IL-15 cytokines with a regimen of blocking antibodies pre- and post-adoptive transfer of low-avidity HA518-526-specific CD8+T-cells following intravenous injection of 4T1HA cells increased the number of pulmonary tumor nodules. This neutralization effect was counteracted by the tumor metastasis-suppressing action of bortezomib treatments. In bortezomib-treated 4T1HA tumor-bearing mice, CD4+T-cells showed increased IL-2 production, CD11c+ dendritic cells showed increased IL-12 and IL-15 production, and HA-specific activated CD8+T-cells showed enhanced expression of IFNγ, granzyme-B and transcription factor eomesodermin. We also noted a trend of increased expression of IL-2, IL-12 and IL-15 receptors as well as increased phosphorylation of STAT5 in tumor-infiltrating CD8+T-cells following bortezomib treatment. Furthermore, bortezomib-treated CD8+T-cells showed increased phosphorylation of mitogen-activated protein kinase p38, and Akt, which was abrogated by phosphatidylinositide 3-kinase (PI3K) inhibitor. These data support the therapeutic potential of bortezomib in conjunction with other immunotherapies to augment the strength of convergent signals from CD8+T-cell signaling molecules including TCR, cytokine receptors and downstream PI3K/Akt/STAT5 pathways to sustain CD8+T-cell effector function in the tumor microenvironment.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Breast Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/drug effects , Cytokines/metabolism , Fibrosarcoma/drug therapy , Kidney Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Proteasome Inhibitors/pharmacology , Tumor Microenvironment , Adenocarcinoma/enzymology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Female , Fibrosarcoma/enzymology , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Kidney Neoplasms/enzymology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred BALB C , Mice, Transgenic , Phosphatidylinositol 3-Kinase/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytokine/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Escape/drug effects
8.
Immunol Lett ; 169: 61-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26597610

ABSTRACT

Common γ chain (γC) cytokines, namely IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 are important for the proliferation, differentiation, and survival of lymphocytes that display antitumor activity, thus stimulating considerable interest for the use of cytokines in cancer immunotherapy. In this review, we will focus on the γC cytokines that demonstrate the greatest potential for immunotherapy, IL-2, IL-7, IL-15, and IL-21. We will briefly cover their biological function, potential applications in cancer therapy, and update on their use in combinatorial immune strategies for eradicating tumors and hematopoietic malignancies.


Subject(s)
Cytokines/metabolism , Immunotherapy , Interleukin Receptor Common gamma Subunit/metabolism , Lymphocytes/immunology , Neoplasms/therapy , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Clinical Trials as Topic , Combined Modality Therapy , Humans , Neoplasms/immunology
10.
Oncotarget ; 6(32): 32439-55, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26431276

ABSTRACT

The immunosuppressive tumor microenvironment usurps host antitumor immunity by multiple mechanisms including interference with the Notch system, which is important for various metazoan cell fate decisions and hematopoietic cell differentiation and function. We observed that treatment with the proteasome inhibitor bortezomib in mice bearing various solid tumors resulted in an upregulated expression of various Notch signaling components in lymphoid tissues, thereby increasing CD8+T-lymphocyte IFNγ secretion and expression of effector molecules, perforin and granzyme B, as well as the T-box transcription factor eomesodermin. Bortezomib also neutralized TGFß-mediated suppression of IFNγ and granzyme B expression in activated CD8+T-cells. Of note, bortezomib reversed tumor-induced downregulation of Notch receptors, Notch1 and Notch2, as well as increased the levels of cleaved Notch intracellular domain (NICD) and downstream targets Hes1 and Hey1 in tumor-draining CD8+T-cells. Moreover, bortezomib promoted CD8+T-cell nuclear factor-κB (NFκB) activity by increasing the total and phosphorylated levels of the IκB kinase and IκBα as well as the cytoplasmic and nuclear levels of phosphorylated p65. Even when we blocked NFκB activity by Bay-11-7082, or NICD cleavage by γ-secretase inhibitor, bortezomib significantly increased expression of Notch Hes1 and Hey1 genes as well as perforin, granzyme B and eomesodermin in activated CD8+T-cells. Data suggest that bortezomib can rescue tumor-induced dysfunction of CD8+T-cells by its intrinsic stimulatory effects promoting NICD-NFκB crosstalk. These findings provide novel insights on using bortezomib not only as an agent to sensitize tumors to cell death but also to provide lymphocyte-stimulatory effects, thereby overcoming immunosuppressive actions of tumor on anti-tumor T-cell functions.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Breast Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/drug effects , Kidney Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , NF-kappa B/metabolism , Proteasome Inhibitors/pharmacology , Receptors, Notch/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Genes, ras , Granzymes/metabolism , Homeodomain Proteins/metabolism , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Transgenic , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/metabolism , Transcription Factor HES-1 , Tumor Escape , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Cancer Res ; 75(22): 4728-41, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26404003

ABSTRACT

Activation of Notch signaling in hematopoietic cells by tumors contributes to immune escape. T-cell defects in tumors can be reversed by treating tumor-bearing mice with multivalent forms of the Notch receptor ligand DLL-1, but the immunologic correlates of this effect have not been elucidated. Here, we report mechanistic insights along with the efficacy of combinational treatments of multivalent DLL-1 with oncoprotein targeting drugs in preclinical mouse models of lung cancer. Systemic DLL-1 administration increased T-cell infiltration into tumors and elevated numbers of CD44(+)CD62L(+)CD8(+) memory T cells while decreasing the number of regulatory T cells and limiting tumor vascularization. This treatment was associated with upregulation of Notch and its ligands in tumor-infiltrating T cells enhanced expression of T-bet and phosphorylation of Stat1/2. Adoptive transfer of T cells from DLL1-treated tumor-bearing immunocompetent hosts into tumor-bearing SCID-NOD immunocompromised mice attenuated tumor growth and extended tumor-free survival in the recipients. When combined with the EGFR-targeted drug erlotinib, DLL-1 significantly improved progression-free survival by inducing robust tumor-specific T-cell immunity. In tissue culture, DLL1 induced proliferation of human peripheral T cells, but lacked proliferative or clonogenic effects on lung cancer cells. Our findings offer preclinical mechanistic support for the development of multivalent DLL1 to stimulate antitumor immunity.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium-Binding Proteins , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Female , Flow Cytometry , Humans , Immunohistochemistry , Immunotherapy/methods , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/agonists , Recombinant Fusion Proteins/pharmacology
12.
Mol Immunol ; 45(3): 661-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17869341

ABSTRACT

Immunogenic properties of the combined vaccine CombiHIVvac, comprising polyepitope HIV-1 immunogens, one being the artificial polyepitope protein TBI, containing the T- and B-cell epitopes from Env and Gag proteins, and the DNA vaccine construct pcDNA-TCI coding for the artificial protein TCI, carrying over 80 T-cell epitopes (both CD4+ CTL and CD8+ Th) from Env, Gag, Pol, and Nef proteins, are studied in this work. The data reported demonstrate clearly that a combination of two B- and T-cell immunogens (TBI and TCI) in one construct results in a synergistic increase in the antibody response to both TBI protein and the proteins from HIV-1 lysate. The level of antibodies induced by immunization with the constructs containing either immunogen alone (TBI protein or the plasmid pcDNA-TCI) was significantly lower as compared to that induced by the combined vaccine. The analysis performed suggests that the presence of CD4+ T-helper epitopes, which can be presented by MHC class II, in the protein TCI may be the main reason underlying the increased synthesis of antibodies to TBI protein due to a CD4-mediated stimulation of B-cell proliferation and differentiation.


Subject(s)
AIDS Vaccines/agonists , Antibodies, Viral/immunology , Antibody Formation/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , Viral Proteins/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antigen Presentation/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred BALB C , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Proteins/genetics
13.
Vaccine ; 25(21): 4312-23, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17418918

ABSTRACT

We have previously described designing of polyepitope immunogens TBI and TCI, to stimulate the humoral and cellular immune responses to HIV-1. Here, immunogens TBI and TCI were used to create new vaccine construct named CombiHIVvac (Combined HIV-1 vaccine). CombiHIVvac is a virus-like particles (VLP) containing the DNA vaccine pcDNA-TCI as a core encapsulated within a spermidine-polyglucin-TBI conjugate. The immunogenic and toxic properties of the candidate vaccine CombiHIVvac have been studied. CombiHIVvac induces a strong humoral and CTL responses in mice; the antibodies are highly specific and are able to neutralize HIV-1 in vitro. Preclinical study demonstrated that CombiHIVvac does not cause long-term changes in physiological, biochemical and morphological parameters in immunized animals and thus can be recommended for clinical trials.


Subject(s)
AIDS Vaccines/immunology , Epitopes/immunology , HIV-1/immunology , Vaccines, DNA/immunology , Vaccines, Virosome/immunology , AIDS Vaccines/adverse effects , AIDS Vaccines/chemistry , Animals , Blotting, Western , Cells, Cultured , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , HIV Antibodies/blood , Humans , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Models, Animal , Neutralization Tests , Vaccines, DNA/adverse effects , Vaccines, DNA/chemistry , Vaccines, Virosome/adverse effects , Vaccines, Virosome/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...