Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 11(10): 2183-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22891038

ABSTRACT

The importance of the blood-brain barrier in preventing effective pharmacotherapy of glioblastoma has been controversial. The controversy stems from the fact that vascular endothelial cell tight junctions are disrupted in the tumor, allowing some systemic drug delivery. P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) efflux drugs from brain capillary endothelial cells into the blood. We tested the hypothesis that although the tight junctions are "leaky" in the core of glioblastomas, active efflux limits drug delivery to tumor-infiltrated normal brain and consequently, treatment efficacy. Malignant gliomas were induced by oncogene transfer into wild-type (WT) mice or mice deficient for Pgp and BCRP (knockout, KO). Glioma-bearing mice were orally dosed with dasatinib, a kinase inhibitor and dual BCRP/PgP substrate that is being currently tested in clinical trials. KO mice treated with dasatinib survived for twice as long as WT mice. Microdissection of the tumor core, invasive rim, and normal brain revealed 2- to 3-fold enhancement in dasatinib brain concentrations in KO mice relative to WT. Analysis of signaling showed that poor drug delivery correlated with the lack of inhibition of a dasatinib target, especially in normal brain. A majority of human glioma xenograft lines tested expressed BCRP or PgP and were sensitized to dasatinib by a dual BCRP/Pgp inhibitor, illustrating a second barrier to drug delivery intrinsic to the tumor itself. These data show that active efflux is a relevant obstacle to treating glioblastoma and provide a plausible mechanistic basis for the clinical failure of numerous drugs that are BCRP/Pgp substrates.


Subject(s)
Brain/metabolism , Glioblastoma/drug therapy , Molecular Targeted Therapy , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Thiazoles/metabolism , Thiazoles/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/metabolism , Acridines/chemistry , Acridines/pharmacology , Acridines/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/pathology , Dasatinib , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Mice , Mice, Knockout , Oncogenes , Permeability/drug effects , Pyrimidines/chemistry , Pyrimidines/pharmacology , Signal Transduction/drug effects , Survival Analysis , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacology , Tetrahydroisoquinolines/therapeutic use , Thiazoles/chemistry , Thiazoles/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Tissue Distribution/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 18(17): 4657-68, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22781551

ABSTRACT

PURPOSE: We tested the combination of a tumor lysate vaccine with a panel of costimulatory molecules to identify an immunotherapeutic approach capable of curing established murine gliomas. EXPERIMENTAL DESIGN: Glioma-bearing mice were primed with a tumor lysate vaccine, followed by systemic administration of the following costimulatory ligands: OX40L, CD80, 4-1BBL, and GITRL, which were fused to the Fc portion of human immunoglobulin. Lymphocytes and mRNA were purified from the brain tumor site for immune monitoring studies. Numerous variations of the vaccine and Fc-OX40L regimen were tested alone or in combination with temozolomide. RESULTS: Lysate vaccinations combined with Fc-OX40L led to the best overall survival, yielding cure rates of 50% to 100% depending on the timing, regimen, and combination with temozolomide. Cured mice that were rechallenged with glioma cells rejected the challenge, showing immunologic memory. Lymphocytes isolated from the draining lymph nodes of vaccine/Fc-OX40L-treated mice had superior tumoricidal function relative to all other groups. Vaccine/Fc-OX40L-treated mice exhibited a significant increase in proliferation of brain-infiltrating CD4 and CD8 T cells, as indicated by Ki67 staining. Fc-OX40L had single-agent activity in transplanted and spontaneous glioma models, and the pattern of inflammatory gene expression in the tumor predicted the degree of therapeutic response. CONCLUSIONS: These data show that Fc-OX40L has unique and potent activity against experimental gliomas and warrants further testing.


Subject(s)
Cancer Vaccines , Glioma , Immunoglobulin Fc Fragments , Immunotherapy , 4-1BB Ligand/genetics , 4-1BB Ligand/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Glioma/immunology , Glioma/metabolism , Glioma/pathology , Glioma/therapy , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunologic Memory , Ligands , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Mice , OX40 Ligand/genetics , OX40 Ligand/immunology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology
3.
Cancer Res ; 71(7): 2664-74, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21324923

ABSTRACT

Epidemiologic studies have highlighted associations between the regular use of nonsteroidal anti-inflammatory drugs (NSAID) and reduced glioma risks in humans. Most NSAIDs function as COX-2 inhibitors that prevent production of prostaglandin E2 (PGE2). Because PGE2 induces expansion of myeloid-derived suppressor cells (MDSC), we hypothesized that COX-2 blockade would suppress gliomagenesis by inhibiting MDSC development and accumulation in the tumor microenvironment (TME). In mouse models of glioma, treatment with the COX-2 inhibitors acetylsalicylic acid (ASA) or celecoxib inhibited systemic PGE2 production and delayed glioma development. ASA treatment also reduced the MDSC-attracting chemokine CCL2 (C-C motif ligand 2) in the TME along with numbers of CD11b(+)Ly6G(hi)Ly6C(lo) granulocytic MDSCs in both the bone marrow and the TME. In support of this evidence that COX-2 blockade blocked systemic development of MDSCs and their CCL2-mediated accumulation in the TME, there were defects in these processes in glioma-bearing Cox2-deficient and Ccl2-deficient mice. Conversely, these mice or ASA-treated wild-type mice displayed enhanced expression of CXCL10 (C-X-C motif chemokine 10) and infiltration of cytotoxic T lymphocytes (CTL) in the TME, consistent with a relief of MDSC-mediated immunosuppression. Antibody-mediated depletion of MDSCs delayed glioma growth in association with an increase in CXCL10 and CTLs in the TME, underscoring a critical role for MDSCs in glioma development. Finally, Cxcl10-deficient mice exhibited reduced CTL infiltration of tumors, establishing that CXCL10 limited this pathway of immunosuppression. Taken together, our findings show that the COX-2 pathway promotes gliomagenesis by directly supporting systemic development of MDSCs and their accumulation in the TME, where they limit CTL infiltration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Glioma/prevention & control , Myeloid Cells/drug effects , Alleles , Animals , Aspirin/pharmacology , CD8-Positive T-Lymphocytes/immunology , Celecoxib , Chemokine CCL2/immunology , Chemokine CXCL10/immunology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Female , Glioma/enzymology , Glioma/immunology , Glioma/pathology , Lysosomal-Associated Membrane Protein 1/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/enzymology , Myeloid Cells/immunology , Myeloid Cells/pathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Cytotoxic/immunology
4.
Clin Cancer Res ; 16(13): 3409-19, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20472682

ABSTRACT

PURPOSE: We hypothesized that the type 1 IFNs would play a pivotal role in antiglioma immunosurveillance through promotion of type 1 adaptive immunity and suppression of immunoregulatory cells. EXPERIMENTAL DESIGN: We induced de novo gliomas in Ifnar1(-/-) (deficient for type 1 IFN receptors) or wild-type mice by intracerebroventricuar transfection of NRas and a short hairpin RNA against P53 using the Sleeping Beauty transposon system. We analyzed the survival of 587 glioma patients for single nucleotide polymorphisms (SNP) in type 1 IFN-related genes. RESULTS: Ifnar1(-/-) mice exhibited accelerated tumor growth and death. Analyses of brain tumor-infiltrating lymphocytes in Ifnar1(-/-) mice revealed an increase of cells positive for CD11b(+)Ly6G(+) and CD4(+)FoxP3(+), which represent myeloid-derived suppressor cells and regulatory T cells, respectively, but a decrease of CD8(+) cytotoxic T lymphocytes (CTLs) compared with wild-type mice. Ifnar1(-/-) mouse-derived glioma tissues exhibited a decrease in mRNA for the CTL-attracting chemokine Cxcl10, but an increase of Ccl2 and Ccl22, both of which are known to attract immunoregulatory cell populations. Dendritic cells generated from the bone marrow of Ifnar1(-/-) mice failed to function as effective antigen-presenting cells. Moreover, depletion of Ly6G(+) cells prolonged the survival of mice with developing gliomas. Human epidemiologic studies revealed that SNPs in IFNAR1 and IFNA8 are associated with significantly altered overall survival of patients with WHO grade 2 to 3 gliomas. CONCLUSIONS: The novel Sleeping Beauty-induced murine glioma model led us to discover a pivotal role for the type 1 IFN pathway in antiglioma immunosurveillance and relevant human SNPs that may represent novel prognostic markers.


Subject(s)
Brain Neoplasms/economics , Brain Neoplasms/immunology , Glioma/immunology , Interferon Type I/genetics , Interferon Type I/physiology , Receptor, Interferon alpha-beta/deficiency , Adaptive Immunity , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Chemokine CXCL10 , Dendritic Cells/immunology , Glioma/genetics , Glioma/mortality , Interferon Type I/deficiency , Lymphocytes, Tumor-Infiltrating , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monitoring, Immunologic , Polymorphism, Single Nucleotide , Prognosis , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology
5.
Cancer Res ; 69(2): 431-9, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147555

ABSTRACT

Spontaneous mouse models of cancer show promise to more accurately recapitulate human disease and predict clinical efficacy. Transgenic mice or viral vectors have been required to generate spontaneous models of glioma, a lethal brain tumor, because nonviral gene transfer is typically transient. To overcome this constraint, we used the Sleeping Beauty transposable element to achieve chromosomal integration of human oncogenes into endogenous brain cells of immunocompetent mice. Genetically engineered, spontaneous brain tumors were induced with plasmid DNA in a matter of weeks in three separate mouse strains. The phenotype of tumors was influenced by the combination of oncogenes delivered, resembling human astrocytoma or glioblastoma in the majority of cases. At least five different genes can be cotransfected simultaneously including reporters, allowing measurement of tumor viability by in vivo imaging. This model can accelerate brain tumor research in a variety of ways such as generation of "humanized" models for high throughput drug screening and candidate gene validation with exceptional speed and flexibility.


Subject(s)
Brain Neoplasms/genetics , DNA/administration & dosage , Glioma/genetics , Plasmids/administration & dosage , Animals , Brain Neoplasms/pathology , DNA/genetics , Disease Models, Animal , Female , Genes, Reporter , Genes, Tumor Suppressor , Genetic Engineering/methods , Glioma/pathology , Injections, Intraventricular , Male , Mice , Oncogenes , Plasmids/genetics , Polyethyleneimine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...