Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
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
2.
Neoplasia ; 32: 100824, 2022 10.
Article in English | MEDLINE | ID: mdl-35914370

ABSTRACT

Non-small cell lung cancer (NSCLC) is a heterogeneous disease with genetic and environmental parameters that influence cell metabolism. Because of the complex interplay of environmental factors within the tumor microenvironment (TME) and the profound impact of these factors on the metabolic activities of tumor and immune cells, there is an emerging interest to advance the understanding of these diverse metabolic phenotypes in the TME. High levels of adenosine are characteristic of the TME, and adenosine can have a significant impact on both tumor cell growth and the immune response. Consistent with this, we showed in NSCLC data from TCGA that high expression of the A2BR leads to worse outcome and that expression of A2BR may be different for different mutation backgrounds. We then investigated the metabolic reprogramming of tumor cells and immune cells (T and dendritic cells) by adenosine. We used A2AR and A2BR antagonism or agonism as well as receptor knockout animals to explore whether these treatments altered specific immune compartments or conferred specific therapeutic vulnerabilities. Using the seahorse assay, we found that an A2BR antagonist modulates oxidative stress homeostasis in NSCLC cell lines. In addition, we found distinct metabolic roles of A2AR and A2BR receptors in T cell activation and dendritic cell maturation. These data suggest potential mechanisms and therapeutic benefits of A2 receptor antagonist therapy in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenosine , Animals , Receptor, Adenosine A2A , Receptor, Adenosine A2B , Tumor Microenvironment
3.
Front Immunol ; 11: 1958, 2020.
Article in English | MEDLINE | ID: mdl-32922403

ABSTRACT

Understanding and targeting Notch signaling effectively has long been valued in the field of cancer and other immune disorders. Here, we discuss key discoveries at the intersection of Notch signaling, cancer and immunology. While there is a plethora of Notch targeting agents tested in vitro, in vivo and in clinic, undesirable off-target effects and therapy-related toxicities have been significant obstacles. We make a case for the clinical application of ligand-derived and affinity modifying compounds as novel therapeutic agents and discuss major research findings with an emphasis on Notch ligand-specific modulation of immune responses.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/therapy , Receptors, Notch/antagonists & inhibitors , Animals , Antineoplastic Agents/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Ligands , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Tumor Escape , Tumor Microenvironment
4.
Clin Cancer Res ; 26(3): 669-678, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31672772

ABSTRACT

PURPOSE: EGF-like domain 7 (EGFL7) is a secreted protein and recently has been shown to play an important role in acute myeloid leukemia (AML); however, the underlying mechanism by which EGFL7 promotes leukemogenesis is largely unknown. EXPERIMENTAL DESIGN: Using an antibody interaction array, we measured the ability of EGFL7 to bind directly approximately 400 proteins expressed by primary AML blasts. Primary patient samples were stimulated in vitro with recombinant EGFL7 (rEGFL7) or anti-EGFL7 blocking antibody to assess alterations in downstream signaling and the ability to effect blast differentiation and survival. We treated three independent AML models with anti-EGFL7 or IgG1 control to determine whether anti-EGFL7 could prolong survival in vivo. RESULTS: We found EGFL7 significantly binds several signaling proteins important for normal and malignant hematopoiesis including NOTCH. Stimulation of AML blasts with rEGFL7 reduced NOTCH intracellular domain and NOTCH target gene expression while treatment with an anti-EGFL7 blocking antibody resulted in reactivation of NOTCH signaling, increased differentiation, and apoptosis. Competitive ligand-binding assays showed rEGFL7 inhibits DELTA-like (DLL) 4-mediated NOTCH activation while anti-EGFL7 combined with DLL4 significantly increased NOTCH activation and induced apoptosis. Using three different AML mouse models, we demonstrated that in vivo treatment with anti-EGFL7 alone results in increased survival. CONCLUSIONS: Our data demonstrate that EGFL7 contributes to NOTCH silencing in AML by antagonizing canonical NOTCH ligand binding. Reactivation of NOTCH signaling in vivo using anti-EGFL7 results in prolonged survival of leukemic mice, supporting the use of EGFL7 as a novel therapeutic target in AML.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Calcium-Binding Proteins/metabolism , EGF Family of Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/antagonists & inhibitors , Animals , Apoptosis , Calcium-Binding Proteins/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , EGF Family of Proteins/genetics , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/metabolism , Signal Transduction
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.
Nat Commun ; 9(1): 3198, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097569

ABSTRACT

EGFR tyrosine kinase inhibitors cause dramatic responses in EGFR-mutant lung cancer, but resistance universally develops. The involvement of ß-catenin in EGFR TKI resistance has been previously reported, however, the precise mechanism by which ß-catenin activation contributes to EGFR TKI resistance is not clear. Here, we show that EGFR inhibition results in the activation of ß-catenin signaling in a Notch3-dependent manner, which facilitates the survival of a subset of cells that we call "adaptive persisters". We previously reported that EGFR-TKI treatment rapidly activates Notch3, and here we describe the physical association of Notch3 with ß-catenin, leading to increased stability and activation of ß-catenin. We demonstrate that the combination of EGFR-TKI and a ß-catenin inhibitor inhibits the development of these adaptive persisters, decreases tumor burden, improves recurrence free survival, and overall survival in xenograft models. These results supports combined EGFR-TKI and ß-catenin inhibition in patients with EGFR mutant lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Receptor, Notch3/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/genetics , Humans , Lung Neoplasms/blood , Lung Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Plasminogen Activator Inhibitor 1/blood , Protein Stability/drug effects , Transcription Factors/metabolism , beta Catenin/antagonists & inhibitors
9.
Sci Rep ; 7: 41233, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28117423

ABSTRACT

Noninvasive in vivo assessment of chemical tumor microenvironment (TME) parameters such as oxygen (pO2), extracellular acidosis (pHe), and concentration of interstitial inorganic phosphate (Pi) may provide unique insights into biological processes in solid tumors. In this work, we employ a recently developed multifunctional trityl paramagnetic probe and electron paramagnetic resonance (EPR) technique for in vivo concurrent assessment of these TME parameters in various mouse models of cancer. While the data support the existence of hypoxic and acidic regions in TME, the most dramatic differences, about 2-fold higher concentrations in tumors vs. normal tissues, were observed for interstitial Pi - the only parameter that also allowed for discrimination between non-metastatic and highly metastatic tumors. Correlation analysis between [Pi], pO2, pHe and tumor volumes reveal an association of high [Pi] with changes in tumor metabolism and supports different mechanisms of protons and Pi accumulation in TME. Our data identifies interstitial inorganic phosphate as a new TME marker for tumor progression. Pi association with tumor metabolism, buffer-mediated proton transport, and a requirement of high phosphorus content for the rapid growth in the "growth rate hypothesis" may underline its potential role in tumorigenesis and tumor progression.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Disease Progression , Phosphates/analysis , Tumor Microenvironment , Acidosis/metabolism , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Electron Spin Resonance Spectroscopy , Female , Heterografts , Humans , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/metabolism , Mice, Transgenic , Oxygen/analysis , Oxygen/metabolism , Phosphates/metabolism , Tumor Burden
10.
Am J Physiol Renal Physiol ; 310(5): F416-25, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26719361

ABSTRACT

Kidney disease, a common complication of diabetes, associates with poor prognosis. Our previous animal model studies linked aquaporin (AQP)11 to acute kidney injury, hyperglycemia-induced renal impairment, and kidney disease in diabetes. Here, we report the AQP11 rs2276415 variant as a genetic factor placing type 2 diabetic patients at greater risk for the development of kidney disease. We performed two independent retrospective case-control studies in 1,075 diabetic and 1,619 nondiabetic individuals who were identified in the Synthetic Derivative Database with DNA samples in the BioVU DNA repository at Vanderbilt University (Nashville, TN). A χ(2)-test and multivariable logistic regression analysis with adjustments for age, sex, baseline serum creatinine, and underlying comorbid disease covariates showed a significant association between rs2276415 and the prevalence of any event of acute kidney injury and chronic kidney disease (CKD) in diabetic patients but not in patients without diabetes. This result was replicated in the second independent study. Diabetic CKD patients over 55 yrs old with the minor AQP11 allele had a significantly faster progression of estimated glomerular filtration rate decline than patients with the wild-type genotype. Three-dimensional structural analysis suggested a functional impairment of AQP11 with rs2276415, which could place diabetic patients at a higher risk for kidney disease. These studies identified rs2276415 as a candidate genetic factor predisposing patients with type 2 diabetes to CKD.


Subject(s)
Acute Kidney Injury/genetics , Aquaporins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/genetics , Acute Kidney Injury/diagnosis , Acute Kidney Injury/epidemiology , Aged , Aquaporins/chemistry , Aquaporins/metabolism , Chi-Square Distribution , Databases, Genetic , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/epidemiology , Disease Progression , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Glomerular Filtration Rate , Humans , Linear Models , Logistic Models , Male , Middle Aged , Models, Molecular , Multivariate Analysis , Phenotype , Prevalence , Protein Conformation , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Retrospective Studies , Risk Assessment , Risk Factors , Structure-Activity Relationship
12.
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
13.
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
14.
Prog Tumor Res ; 42: 95-109, 2015.
Article in English | MEDLINE | ID: mdl-26383097

ABSTRACT

Metastatic lung cancer is the most common cause of cancer mortality globally in both men and women, with 5-year survival of less than 5%. Standard treatment approaches for metastatic lung cancer are based on chemotherapy, with radiation and surgery used for local control, but these rarely result in relapse-free survivals longer than 2-3 years, although they may provide symptom relief. Thus, additional tools are needed to treat this disease. In this chapter, we discuss the various immune-based cancer treatments for lung cancer patients that are being developed, and the increasing awareness that therapies targeted at overcoming immune evasion mechanisms may be essential to clinical efficacy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Immunotherapy , Lung Neoplasms/drug therapy , Animals , Female , Humans , Lung Neoplasms/immunology , Male
15.
Mol Pharmacol ; 85(1): 62-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24136993

ABSTRACT

Interstitial adenosine stimulates neovascularization in part through A2B adenosine receptor-dependent upregulation of vascular endothelial growth factor (VEGF). In the current study, we tested the hypothesis that A2B receptors upregulate JunB, which can contribute to stimulation of VEGF production. Using the human microvascular endothelial cell line, human mast cell line, mouse cardiac Sca1-positive stromal cells, and mouse Lewis lung carcinoma (LLC) cells, we found that adenosine receptor-dependent upregulation of VEGF production was associated with an increase in VEGF transcription, activator protein-1 (AP-1) activity, and JunB accumulation in all cells investigated. Furthermore, the expression of JunB, but not the expression of other genes encoding transcription factors from the Jun family, was specifically upregulated. In LLC cells expressing A2A and A2B receptor transcripts, only the nonselective adenosine agonist NECA (5'-N-ethylcarboxamidoadenosine), but not the selective A2A receptor agonist CGS21680 [2-p-(2-carboxyethyl) phenylethylamino-5'-N-ethylcarboxamidoadenosine], significantly increased JunB reporter activity and JunB nuclear accumulation, which were inhibited by the A2B receptor antagonist PSB603 [(8-[4-[4-((4-chlorophenzyl)piperazide-1-sulfonyl)phenyl]]-1-propylxanthine]. Using activators and inhibitors of intracellular signaling, we demonstrated that A2B receptor-dependent accumulation of JunB protein and VEGF secretion share common intracellular pathways. NECA enhanced JunB binding to the murine VEGF promoter, whereas mutation of the high-affinity AP-1 site (-1093 to -1086) resulted in a loss of NECA-dependent VEGF reporter activity. Finally, NECA-dependent VEGF secretion and reporter activity were inhibited by the expression of a dominant negative JunB or by JunB knockdown. Thus, our data suggest an important role of the A2B receptor-dependent upregulation of JunB in VEGF production and possibly other AP-1-regulated events.


Subject(s)
Receptor, Adenosine A2B/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Adenosine/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Humans , Mice , Mutation , Promoter Regions, Genetic , RNA Interference , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
16.
Am J Physiol Renal Physiol ; 304(10): F1295-307, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23486012

ABSTRACT

Aquaporin 11 (AQP11) is a newly described member of the protein family of transport channels. AQP11 associates with the endoplasmic reticulum (ER) and is highly expressed in proximal tubular epithelial cells in the kidney. Previously, we identified and characterized a recessive mutation of the highly conserved Cys227 to Ser227 in mouse AQP11 that caused proximal tubule (PT) injury and kidney failure in mutant mice. The current study revealed induction of ER stress, unfolded protein response, and apoptosis as molecular mechanisms of this PT injury. Cys227Ser mutation interfered with maintenance of AQP11 oligomeric structure. AQP11 is abundantly expressed in the S1 PT segment, a site of major renal glucose flux, and Aqp11 mutant mice developed PT-specific mitochondrial injury. Glucose increased AQP11 protein expression in wild-type kidney and upregulation of AQP11 expression by glucose in vitro was prevented by phlorizin, an inhibitor of sodium-dependent glucose transport across PT. Total AQP11 levels in heterozygotes were higher than in wild-type mice but were not further increased in response to glucose. In Aqp11 insufficient PT cells, glucose potentiated increases in reactive oxygen species (ROS) production. ROS production was also elevated in Aqp11 mutation carriers. Phenotypically normal mice heterozygous for the Aqp11 mutation repeatedly treated with glucose showed increased blood urea nitrogen levels that were prevented by the antioxidant sulforaphane or by phlorizin. Our results indicate an important role for AQP11 to prevent glucose-induced oxidative stress in proximal tubules.


Subject(s)
Aquaporins/genetics , Endoplasmic Reticulum/metabolism , Kidney/metabolism , Oxidative Stress/genetics , Renal Insufficiency/genetics , Animals , Aquaporins/metabolism , Cell Line , Endoplasmic Reticulum Stress/physiology , Mice , Mutation , Reactive Oxygen Species/metabolism , Renal Insufficiency/metabolism , Up-Regulation
17.
J Immunol ; 187(11): 6120-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22039302

ABSTRACT

Extracellular adenosine and purine nucleotides are elevated in many pathological situations associated with the expansion of CD11b(+)Gr1(+) myeloid-derived suppressor cells (MDSCs). Therefore, we tested whether adenosinergic pathways play a role in MDSC expansion and functions. We found that A(2B) adenosine receptors on hematopoietic cells play an important role in accumulation of intratumoral CD11b(+)Gr1(high) cells in a mouse Lewis lung carcinoma model in vivo and demonstrated that these receptors promote preferential expansion of the granulocytic CD11b(+)Gr1(high) subset of MDSCs in vitro. Flow cytometry analysis of MDSCs generated from mouse hematopoietic progenitor cells revealed that the CD11b(+)Gr-1(high) subset had the highest levels of CD73 (ecto-5'-nucleotidase) expression (Δmean fluorescence intensity [MFI] of 118.5 ± 16.8), followed by CD11b(+)Gr-1(int) (ΔMFI of 57.9 ± 6.8) and CD11b(+)Gr-1(-/low) (ΔMFI of 12.4 ± 1.0) subsets. Even lower levels of CD73 expression were found on Lewis lung carcinoma tumor cells (ΔMFI of 3.2 ± 0.2). The high levels of CD73 expression in granulocytic CD11b(+)Gr-1(high) cells correlated with high levels of ecto-5'-nucleotidase enzymatic activity. We further demonstrated that the ability of granulocytic MDSCs to suppress CD3/CD28-induced T cell proliferation was significantly facilitated in the presence of the ecto-5'-nucleotidase substrate 5'-AMP. We propose that generation of adenosine by CD73 expressed at high levels on granulocytic MDSCs may promote their expansion and facilitate their immunosuppressive activity.


Subject(s)
Adenosine/metabolism , Granulocytes/metabolism , Myeloid Cells/metabolism , 5'-Nucleotidase/immunology , 5'-Nucleotidase/metabolism , Adenosine/immunology , Animals , CD11b Antigen/immunology , CD11b Antigen/metabolism , Carcinoma, Lewis Lung , Cell Differentiation/immunology , Cell Proliferation , Cell Separation , Female , Flow Cytometry , Granulocytes/cytology , Granulocytes/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P1/immunology , Receptors, Purinergic P1/metabolism
18.
Cancer Res ; 71(19): 6122-31, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21825014

ABSTRACT

Deficiencies in immune function that accumulate during cancer immunoediting lead to a progressive escape from host immunosurveillance. Therapies that correct or overcome these defects could have a powerful impact on cancer management, but current knowledge of the types and mechanisms of immune escape is still incomplete. Here, we report a novel mechanism of escape from T-cell immunity that is caused by reduction in levels of the Delta family Notch ligands DLL1 and DLL4 in hematopoietic microenvironments. An important mediator of this effect was an elevation in the levels of circulating VEGF. Selective activation of the DLL1-Notch signaling pathway in bone marrow precursors enhanced T-cell activation and inhibited tumor growth. Conversely, tumor growth led to inhibition of Delta family ligand signaling through Notch in the hematopoietic environment, resulting in suppressed T-cell function. Overall, our findings uncover a novel mechanism of tumoral immune escape and suggest that a soluble multivalent form of DLL1 may offer a generalized therapeutic intervention to stimulate T-cell immunity and suppress tumor growth.


Subject(s)
Immunologic Surveillance , Intercellular Signaling Peptides and Proteins/immunology , Lung Neoplasms/immunology , Receptor, Notch1/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Adaptor Proteins, Signal Transducing , Animals , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins , Cell Line, Tumor , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/blood supply , Lymphocyte Activation , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Neovascularization, Pathologic , Receptor, Notch1/metabolism , Signal Transduction , Tumor Escape/genetics , Tumor Microenvironment/immunology , Vascular Endothelial Growth Factor A
19.
J Thorac Oncol ; 5(9): 1410-5, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20683208

ABSTRACT

INTRODUCTION: Targeting of cancer by chemotherapy in combination with anti-vascular endothelial growth factor (VEGF) therapy has demonstrated not only the clinical efficacy but also a higher risk of serious hematologic complications including neutropenia. The purpose of the study was to elucidate the molecular mechanisms responsible for the development of neutropenia during the combination treatment. METHODS: Mouse model and in vitro studies were undertaken to determine the effect of interference with VEGF signaling by VEGF-specific agents or a multitargeted VEGF receptor (VEGFR) tyrosine kinase inhibitor on proliferation of hematopoietic progenitor cell (HPC) and repopulation of the hematopoietic compartment after myeloablation. RESULTS: The studies demonstrated that blockage of VEGFR1 or VEGFR2 signaling decreased HPC proliferation and impaired repopulation of the hematopoietic compartment after myelosuppression by slowing the progression of HPC through the cell cycle. The combination of cytotoxic drugs and VEGFR tyrosine kinase inhibitor had an additive inhibitory effect and decreased proliferation of HPC significantly stronger than either agent alone. CONCLUSIONS: Signaling through both VEGFR1 and VEGFR2 is required for normal reconstitution of the hematopoietic compartment after cytotoxic chemotherapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/drug effects , Cell Proliferation/drug effects , Hematopoietic Stem Cells/pathology , Hematopoietic System/drug effects , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Blotting, Western , Cell Cycle/drug effects , Female , Flow Cytometry , Fluorouracil/administration & dosage , Hematopoietic Stem Cells/metabolism , Hematopoietic System/physiology , Indoles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pyrroles/administration & dosage , Signal Transduction/drug effects , Survival Rate , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
20.
Biochem Biophys Res Commun ; 392(2): 201-6, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20060810

ABSTRACT

The Forkhead/Fox transcription factor Foxc2 is a critical regulator of vascular development. However, the role of Foxc2 in pathological angiogenesis in cancer remains unknown. Here we show that FoxC2 is highly expressed in human breast and colonic tumors and in the tumor endothelium in human and mouse melanomas. Using the B16 melanoma tumor model, we investigated the function of Foxc2 in tumor angiogenesis. After subcutaneous injection of B16 melanoma cells, primary tumor growth as well as neovascularization was markedly reduced in mice lacking one copy of the Foxc2 gene (Foxc2+/-). Consistently, expression levels of several angiogenic factors, including vascular endothelial growth factor (Vegf), matrix metallopeptidase 2 (Mmp2), and platelet-derived growth factor-B (Pdgfb), were significantly decreased in B16 tumors grown in Foxc2+/- mice, and tumor blood vessels formed in Foxc2+/- mice showed reduced coverage of mural cells and endothelial cell apoptosis. In addition, the tumor tissue in Foxc2+/- mice had an accumulation of necrotic cells. Taken together, these findings demonstrate that haplodeficiency of Foxc2 results in impaired formation of tumor blood vessels as well as reduced tumor growth and thereby provide evidence that Foxc2 is critical for tumor development and angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , Forkhead Transcription Factors/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Animals , Apoptosis , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Gene Flow , Heterozygote , Humans , Matrix Metalloproteinase 2/genetics , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Mice, Mutant Strains , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL