Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 13(7): 2387-96, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27248573

ABSTRACT

Antibody-drug conjugates (ADC) rely on the target-binding specificity of an antibody to selectively deliver potent drugs to cancer cells. IgG antibody half-life is regulated by neonatal Fc receptor (FcRn) binding. Histidine 435 of human IgG was mutated to alanine (H435A) to explore the effect of FcRn binding on the pharmacokinetics, efficacy, and tolerability of two separate maytansine-based ADC pairs with noncleavable linkers, (c-DM1 and c-H435A-DM1) and (7v-Cys-may and 7v-H435A-Cys-may). The in vitro cell-killing potency of each pair of ADCs was similar, demonstrating that H435A showed no measurable impact on ADC bioactivity. The H435A mutant antibodies showed no detectable binding to human or mouse FcRn in vitro, whereas their counterpart wild-type IgG ADCs were found to bind to FcRn at pH = 6.0. In xenograft bearing SCID mice expressing mouse FcRn, the AUC of 7v-Cys-may was 1.6-fold higher than that of 7v-H435A-may, yet the observed efficacy was similar. More severe thrombocytopenia was observed with 7v-H435A-Cys-may as compared to 7v-Cys-may at multiple dose levels. The AUC of c-DM1 was approximately 3-fold higher than that of c-H435A-DM1 in 786-0 xenograft bearing SCID mice, which led to a 3-fold difference in efficacy by dose. Murine FcRn knockout, human FcRn transgenic line 32 SCID animals bearing 786-0 xenografts showed an amplified exposure difference between c-DM1 and c-H435A-DM1 as compared to murine FcRn expressing SCID mice, leading to a 10-fold higher dose required for efficacy despite a 6-fold higher AUC of the c-H435A-DM1. The accelerated clearance observed for the noncleavable maytansine ADCs with the H435A FcRn mutation led to reduced efficacy at equivalent doses and exacerbation of clinical pathology parameters (decreased tolerability) at equivalent doses. The results show that reduced ADC clearance mediated by FcRn modulation can improve therapeutic index.


Subject(s)
Antibodies/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/pharmacology , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Animals , Antibodies/genetics , CD27 Ligand/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Histocompatibility Antigens Class I/genetics , Humans , Immunoconjugates/chemistry , Maytansine/metabolism , Mice , Mice, SCID , Receptors, Fc/genetics
2.
Cancer Res ; 75(24): 5329-40, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26631267

ABSTRACT

Antibody-drug conjugates (ADC) target cytotoxic drugs to antigen-positive cells for treating cancer. After internalization, ADCs with noncleavable linkers are catabolized to amino acid-linker-warheads within the lysosome, which then enter the cytoplasm by an unknown mechanism. We hypothesized that a lysosomal transporter was responsible for delivering noncleavable ADC catabolites into the cytoplasm. To identify candidate transporters, we performed a phenotypic shRNA screen with an anti-CD70 maytansine-based ADC. This screen revealed the lysosomal membrane protein SLC46A3, the genetic attenuation of which inhibited the potency of multiple noncleavable antibody-maytansine ADCs, including ado-trastuzumab emtansine. In contrast, the potencies of noncleavable ADCs carrying the structurally distinct monomethyl auristatin F were unaffected by SLC46A3 attenuation. Structure-activity experiments suggested that maytansine is a substrate for SLC46A3. Notably, SLC46A3 silencing led to relative increases in catabolite concentrations in the lysosome. Taken together, our results establish SLC46A3 as a direct transporter of maytansine-based catabolites from the lysosome to the cytoplasm, prompting further investigation of SLC46A3 as a predictive response marker in breast cancer specimens.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Immunoconjugates/metabolism , Maytansine/metabolism , Membrane Transport Proteins/metabolism , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Line, Tumor , Cytoplasm/metabolism , Drug Delivery Systems , Humans , Immunoconjugates/administration & dosage , Lysosomes/metabolism , Maytansine/administration & dosage
3.
Drug Metab Dispos ; 43(9): 1341-4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26101225

ABSTRACT

Antibody drug conjugates are emerging as a powerful class of antitumor agents with efficacy across a range of cancers; therefore, understanding the disposition of this class of therapeutic is crucial. Reported here is a method of enriching a specific organelle (lysosome) to understand the catabolism of an anti-CD70 Ab-MCC-DM1, an antibody drug conjugate with a noncleavable linker. With such techniques a higher degree of concentration-activity relationship can be established for in vitro cell lines; this can aid in understanding the resultant catabolite concentrations necessary to exert activity.


Subject(s)
Immunoconjugates/metabolism , Lysosomes/metabolism , Pharmaceutical Preparations/metabolism , CD27 Ligand/immunology , Cell Line, Tumor , Humans
4.
Mol Cancer Ther ; 14(7): 1614-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931519

ABSTRACT

Epidermal growth factor receptor variant III (EGFRvIII) is a cancer-specific deletion mutant observed in approximately 25% to 50% of glioblastoma multiforme (GBM) patients. An antibody drug conjugate, AMG 595, composed of the maytansinoid DM1 attached to a highly selective anti-EGFRvIII antibody via a noncleavable linker, was developed to treat EGFRvIII-positive GBM patients. AMG 595 binds to the cell surface and internalizes into the endo-lysosomal pathway of EGFRvIII-expressing cells. Incubation of AMG 595 with U251 cells expressing EGFRvIII led to potent growth inhibition. AMG 595 treatment induced significant tumor mitotic arrest, as measured by phospho-histone H3, in GBM subcutaneous xenografts expressing EGFRvIII. A single intravenous injection of AMG 595 at 17 mg/kg (250 µg DM1/kg) generated complete tumor regression in the U251vIII subcutaneous xenograft model. AMG 595 mediated tumor regression in the D317 subcutaneous xenograft model that endogenously expresses EGFRvIII. Finally, AMG 595 treatment inhibited the growth of D317 xenografts orthotopically implanted into the brain as determined by magnetic resonance imaging. These results demonstrate that AMG 595 is a promising candidate to evaluate in EGFRvIII-expressing GBM patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/drug therapy , ErbB Receptors/immunology , Glioblastoma/drug therapy , Immunoconjugates/pharmacology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Immunohistochemistry , Injections, Intravenous , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacology , Mice, Nude , Mice, SCID , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology
5.
J Biol Chem ; 290(12): 7535-62, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25583986

ABSTRACT

Producing pure and well behaved bispecific antibodies (bsAbs) on a large scale for preclinical and clinical testing is a challenging task. Here, we describe a new strategy for making monovalent bispecific heterodimeric IgG antibodies in mammalian cells. We applied an electrostatic steering mechanism to engineer antibody light chain-heavy chain (LC-HC) interface residues in such a way that each LC strongly favors its cognate HC when two different HCs and two different LCs are co-expressed in the same cell to assemble a functional bispecific antibody. We produced heterodimeric IgGs from transiently and stably transfected mammalian cells. The engineered heterodimeric IgG molecules maintain the overall IgG structure with correct LC-HC pairings, bind to two different antigens with comparable affinity when compared with their parental antibodies, and retain the functionality of parental antibodies in biological assays. In addition, the bispecific heterodimeric IgG derived from anti-HER2 and anti-EGF receptor (EGFR) antibody was shown to induce a higher level of receptor internalization than the combination of two parental antibodies. Mouse xenograft BxPC-3, Panc-1, and Calu-3 human tumor models showed that the heterodimeric IgGs strongly inhibited tumor growth. The described approach can be used to generate tools from two pre-existent antibodies and explore the potential of bispecific antibodies. The asymmetrically engineered Fc variants for antibody-dependent cellular cytotoxicity enhancement could be embedded in monovalent bispecific heterodimeric IgG to make best-in-class therapeutic antibodies.


Subject(s)
Immunoglobulin G/chemistry , Static Electricity , Amino Acids/chemistry , Animals , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line , Cricetulus , Dimerization , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology , Mice , Protein Engineering , Surface Plasmon Resonance
6.
J Biol Chem ; 289(6): 3571-90, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24311787

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is mediated through the engagement of the Fc segment of antibodies with Fcγ receptors (FcγRs) on immune cells upon binding of tumor or viral antigen. The co-crystal structure of FcγRIII in complex with Fc revealed that Fc binds to FcγRIII asymmetrically with two Fc chains contacting separate regions of the FcγRIII by utilizing different residues. To fully explore this asymmetrical nature of the Fc-FcγR interaction, we screened more than 9,000 individual clones in Fc heterodimer format in which different mutations were introduced at the same position of two Fc chains using a high throughput competition AlphaLISA® assay. To this end, we have identified a panel of novel Fc variants with significant binding improvement to FcγRIIIA (both Phe-158 and Val-158 allotypes), increased ADCC activity in vitro, and strong tumor growth inhibition in mice xenograft human tumor models. Compared with previously identified Fc variants in conventional IgG format, Fc heterodimers with asymmetrical mutations can achieve similar or superior potency in ADCC-mediated tumor cell killing and demonstrate improved stability in the CH2 domain. Fc heterodimers also allow more selectivity toward activating FcγRIIA than inhibitory FcγRIIB. Afucosylation of Fc variants further increases the affinity of Fc to FcγRIIIA, leading to much higher ADCC activity. The discovery of these Fc variants will potentially open up new opportunities of building the next generation of therapeutic antibodies with enhanced ADCC effector function for the treatment of cancers and infectious diseases.


Subject(s)
Antibodies, Neoplasm , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G , Protein Engineering , Receptors, IgG/immunology , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Cell Line, Tumor , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Mice, SCID , Mutation , Neoplasms , Receptors, IgG/genetics , Xenograft Model Antitumor Assays
7.
Mol Cancer ; 11: 47, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22830443

ABSTRACT

BACKGROUND: Successful treatment of solid tumors relies on the ability of drugs to penetrate into the tumor tissue. METHODS: We examined the correlation of panitumumab (an anti-epidermal growth factor [EGFR] antibody) tumor penetration and EGFR saturation, a potential obstacle in large molecule drug delivery, using pharmacokinetics, pharmacodynamics, and tumor growth rate in an A431 epidermoid carcinoma xenograft model of human cancer. To determine receptor saturation, receptor occupancy, and levels of proliferation markers, immunohistochemical and flow cytometric methods were used. Pharmacokinetic data and modeling were used to calculate growth characteristics of panitumumab-treated tumors. RESULTS: Treatment with panitumumab in vivo inhibited pEGFR, Ki67 and pMAPK levels vs control. Tumor penetration and receptor saturation were dose- and time-dependent, reaching 100% and 78%, respectively. Significant tumor inhibition and eradication (p < 0.05) were observed; plasma concentration associated with tumor eradication was estimated to be 0.2 µg/ml. The tumor inhibition model was able to describe the mean tumor growth and death rates. CONCLUSIONS: These data demonstrate that the antitumor activity of panitumumab correlates with its ability to penetrate into tumor tissue, occupy and inhibit activation of EGFR, and inhibit markers of proliferation and MAPK signaling.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Neoplasms/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Ligands , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Panitumumab , Phosphorylation/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Invest Ophthalmol Vis Sci ; 53(4): 2170-80, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22410553

ABSTRACT

PURPOSE: To determine whether systemic treatment with AMG 386, a selective angiopoietin 1/2-neutralizing peptibody, inhibits neovascular processes in animal models of ocular disease. METHODS: AMG 386 was tested in a laser-induced choroidal neovascularization (CNV) model in monkeys using fluorescein angiography. The biodistribution of (125)I-AMG 386 was determined in cynomolgus monkeys by whole-body autoradiography and radioanalysis of ocular tissues. A murine retinopathy of prematurity (ROP) model was used to examine the effect of AMG 386 on established and newly formed retinal vessels, either as a single agent or when combined with VEGF inhibition.AMG 386 pharmacokinetics were evaluated in each model. RESULTS: In the CNV model, AMG 386 significantly decreased fluorescent angiographic leakage and reduced fibroplasia, indicating an impaired healing response consistent with angiogenesis blockade. Radiolabeled AMG 386 was widely distributed across ocular tissues, with highest concentrations in the choroid, cornea, retinal pigmented epithelium, iris/ciliary body, and sclera. In the ROP model, AMG 386 prevented pathologic retinal angiogenesis when administered from P8 to P16 but transiently impeded regression of these abnormal vessels when administered from P17 to P23. Combining AMG 386 with VEGF inhibition led to cooperative prevention of retinal angiogenesis in this model. No AMG 386-related ocular toxicities occurred, and no treatment-related clinical observations were made in any of the studies. CONCLUSIONS: In this study, AMG 386 inhibited angiogenesis in animal models of CNV and ROP, supporting investigation of AMG 386 for the treatment of ocular neovascular diseases in the clinical setting.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Recombinant Fusion Proteins/pharmacology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Angiogenesis Inhibitors/pharmacokinetics , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Animals , Animals, Newborn , Autoradiography , Capillary Permeability/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Eye/metabolism , Female , Fluorescein Angiography , Humans , In Situ Hybridization , Infant, Newborn , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacokinetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/drug effects , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Tissue Distribution
9.
J Immunol ; 187(9): 4392-402, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22013205

ABSTRACT

Herpesvirus Saimiri gene 13 (HVS13) exhibits 57% identity with the predicted sequence of a T cell-derived molecule termed CTLA8. Recombinant HVS13 and CTLA8 stimulate transcriptional factor NF-kappaB activity and Interleukin-6 (IL-6) secretion in fibroblasts, and costimulate T cell proliferation. An HVS13.Fc fusion protein was used to isolate a cDNA encoding a novel receptor that also binds CTLA8. This receptor is unrelated to previously identified cytokine receptor families. A recombinant soluble receptor inhibited T cell proliferation and IL-2 production induced by PHA, concanavalin A (conA), and anti-TCR MAb. These results define CTLA8 and HVS13 as novel cytokines that bind to a novel cytokine receptor. We propose to call these molecules IL-17, vIL-17, and IL-17R, respectively.


Subject(s)
Herpesvirus 2, Saimiriine/immunology , Interleukin-17/history , Receptors, Interleukin-17/history , Repressor Proteins/history , Trans-Activators/history , Amino Acid Sequence , Animals , Aotidae , Base Sequence , Cell Line, Tumor , History, 20th Century , Humans , Mice , Molecular Sequence Data , Protein Binding/immunology , Rats
10.
Blood ; 118(14): 3811-7, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21841160

ABSTRACT

X-linked hyper IgM syndrome (XHM) is a combined immune deficiency disorder caused by genetic alterations in CD40 ligand. The purpose of this study was to investigate the safety and efficacy of recombinant CD40 ligand (rCD40L) in the treatment of the disease. Three children were administered rCD40L subcutaneously 3 times per week at 0.03 mg/kg for 22 weeks, and after a 12-week drug-free interval, the dose was increased to 0.05 mg/kg for an additional 22 weeks of treatment. Although specific antibody responses to T cell-dependent antigens was lacking, administration of rCD40 resulted in acquisition of the capacity to mount cutaneous delayed type hypersensitivity reactions that disappeared during the drug-free interval as well as the postbiologic follow-up period. With rCD40L treatment, patient T cells developed a new capacity to respond to T-cell mitogens with synthesis of IFN-γ and TNF-α. Intracellular cytokine staining studies showed that both CD4(+) and CD8(+) T cells participated in this response. Finally, CD40L therapy was associated with changes in lymph node size and architecture based on comparison of biopsies taken before and after therapy. This clinical study showed that rCD40L is capable of improving T cell-immune function in patients with XHM.


Subject(s)
CD40 Ligand/therapeutic use , Hyper-IgM Immunodeficiency Syndrome, Type 1/immunology , Hyper-IgM Immunodeficiency Syndrome, Type 1/therapy , Recombinant Proteins/therapeutic use , Adolescent , Animals , CD40 Ligand/administration & dosage , CD40 Ligand/adverse effects , CD40 Ligand/immunology , Child , Follow-Up Studies , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/pathology , Immunotherapy , Interferon-gamma/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/immunology
11.
Prostate ; 70(16): 1799-808, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20583134

ABSTRACT

BACKGROUND: Angiopoietin-2 is expressed in prostate cancer (PCa) bone, liver, and lymph node metastases, whereas, its competitor angiopoietin-1 has limited expression in these tissues. Therefore, we hypothesized that the inhibition of angiopoietin-2 activity in PCa will impede angiogenesis, tumor growth, and alter bone response in vivo. METHODS: To test our hypothesis we used L1-10, a peptide-Fc fusion that inhibits interactions between angiopoietin-2 and its receptor tie2. We blocked angiopoietin-2 activity using L1-10 in established subcutaneous and intra-tibial LuCaP 23.1 xenografts. We then determined the effect of L1-10 on survival, tumor growth, serum PSA, proliferation, microvessel density, and angiogenesis-associated gene expression in subcutaneous tumors. We also determined serum PSA, tumor area, and bone response in intra-tibial tumors. RESULTS: The administration of L1-10 decreased tumor volume and serum PSA, and increased survival in SCID mice bearing subcutaneous LuCaP 23.1 tumors. Histomorphometric analysis, showed a further significant decrease in tumor epithelial area within the L1-10 treated LuCaP 23.1 subcutaneous tumors (P=0.0063). There was also a significant decrease in cell proliferation (P=0.012), microvessel density (P=0.012), and a significant increase in ANGPT-2 and HIF-1α mRNA expression (P≤0.05) associated with L1-10 treatment. Alternatively, in LuCaP 23.1 intra-tibial tumors L1-10 treatment did not significantly change serum PSA, tumor area or bone response. CONCLUSIONS: Our results demonstrate that inhibiting angiopoietin-2 activity impedes angiogenesis and growth of LuCaP 23.1 PCa xenografts. Based on these data, we hypothesize that angiopoietin-2 inhibition in combination with other therapies may represent a potential therapy for patients with metastatic disease.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Prostatic Neoplasms/pathology , Angiopoietin-2/genetics , Animals , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/pathology , Cell Division , Cell Survival , DNA Primers , Humans , Male , Mice , Mice, SCID , Microcirculation , Polymerase Chain Reaction , Prostate/blood supply , Prostate-Specific Antigen/blood , Radiography , Receptor, TIE-2/genetics , Skin Neoplasms/pathology , Tibia/diagnostic imaging , Tibia/pathology , Transplantation, Heterologous
12.
J Biol Chem ; 280(23): 21955-64, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15781451

ABSTRACT

Dendritic cells (DCs) are a phenotypically and functionally heterogenous population of leukocytes with distinct subsets serving a different set of specialized immune functions. Here we applied an in vitro whole cell panning approach using antibody phage display technology to identify cell-surface epitopes specifically expressed on human blood BDCA3(+) DCs. A single-chain antibody fragment (anti-1F12 scFv) was isolated that recognizes a conserved surface antigen expressed on both human BDCA3(+) DCs and mouse CD8alpha(+) DCs. We demonstrate that anti-1F12 scFv binds Nectin-like protein 2 (Necl2, Tslc1, SynCaM, SgIGSF, or Igsf4), an adhesion molecule involved in tumor suppression, synapse formation, and spermatogenesis. Thus, Necl2 defines a specialized subset of DCs in both mouse and human. We further show that Necl2 binds Class-I-restricted T-cell-associated molecule (CRTAM), a receptor primarily expressed on activated cytotoxic lymphocytes. When present on antigen presenting cells, Necl2 regulates IL-22 expression by activated CD8(+) T-cells. We propose that Necl2/CRTAM molecular pair could regulate a large panel of cell/cell interactions both within and outside of the immune system.


Subject(s)
Dendritic Cells/cytology , Immunoglobulins/metabolism , Immunoglobulins/physiology , Membrane Proteins/physiology , T-Lymphocytes/metabolism , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Cell Aggregation , Cell Line, Tumor , Cell Membrane/metabolism , Cell Separation , Coculture Techniques , Electrophoresis, Polyacrylamide Gel , Female , Flow Cytometry , Humans , Immune System/physiology , Immunoblotting , Immunoprecipitation , Interleukins/biosynthesis , Lentivirus/genetics , Leukocytes/metabolism , Ligands , Lymphocytes/cytology , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Octoxynol/pharmacology , Peptide Library , Phenotype , Polymerase Chain Reaction , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Spleen/metabolism , Interleukin-22
13.
Neoplasia ; 5(5): 445-56, 2003.
Article in English | MEDLINE | ID: mdl-14670182

ABSTRACT

Elevated expression of Eph receptors has long been correlated with the growth of solid tumors. However, the functional role of this family of receptor tyrosine kinases in carcinogenesis and tumor angiogenesis has not been well characterized. Here we report that soluble EphA receptors inhibit tumor angiogenesis and tumor progression in vivo in the RIP-Tag transgenic model of vascular endothelial growth factor (VEGF)-dependent multistage pancreatic islet cell carcinoma. Soluble EphA receptors delivered either by a transgene or an osmotic minipump inhibited the formation of angiogenic islet, a premalignant lesion, and reduced tumor volume of solid islet cell carcinoma. EphA2-Fc or EphA3-Fc treatment resulted in decreased tumor volume but increased tumor and endothelial cell apoptosis in vivo. In addition, soluble EphA receptors inhibited VEGF and betaTC tumor cell-conditioned medium-induced endothelial cell migration in vitro and VEGF-induced cornea angiogenesis in vivo. A dominant negative EphA2 mutant inhibited--whereas a gain-of-function EphA2 mutant enhanced--tumor cell-induced endothelial cell migration, suggesting that EphA2 receptor activation is required for tumor cell-endothelial cell interaction. These data provide functional evidence for EphA class receptor regulation of VEGF-dependent tumor angiogenesis, suggesting that the EphA signaling pathway may represent an attractive novel target for antiangiogenic therapy in cancer.


Subject(s)
Carcinoma/metabolism , Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Eph Family/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Coculture Techniques , DNA, Complementary/metabolism , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neovascularization, Pathologic , Receptor, EphA2/metabolism , Signal Transduction , Time Factors , Transgenes
14.
Lab Invest ; 83(11): 1637-45, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14615417

ABSTRACT

The objective of the study was to determine the role of the angiopoietins in the regulation of gelatinase expression during angiogenesis, and whether inhibition of the angiopoietin/Tek interaction in vivo can suppress the extent of retinal neovascularization. Retinal microvascular endothelial cells were treated with angiopoietins and examined for the production of gelatinases. The effects of inhibiting angiopoietin binding to the Tie-2 receptor was studied in newborn mice with experimentally induced retinal neovascularization. Animals were treated with an ip injection of the Tie-2 antagonist, muTek delta Fc, while oxygen-exposed mice treated with similar concentrations of murine IgG were used as controls. The effect of muTek delta Fc on the gelatinase expression in the retina was examined by real-time RT-PCR analysis. The stimulation of cultured retinal endothelial cells with Ang-1 and -2 resulted in the increased expression of matrix metalloproteinase (MMP)-9. Ang-2 expression was up-regulated in experimental animals during the period of angiogenesis and was the greatest on Day 17 (the time of maximal angiogenic response). Histologic analysis of mice treated with the Tie-2 antagonist, muTek delta Fc, showed significant (87%; p = 0.001) inhibition of retinal neovascularization, and the response was dose-dependent. In vitro binding data support the fact that both Ang-1 and Ang-2 bind with high avidity to muTek delta Fc. The RT-PCR analysis of the retinas of the Tek-treated animals showed a similar (80%; p = 0.001) inhibition of the MMP-9 expression, which correlated with the decrease in angiogenesis. The up-regulation of gelatinases in microvascular endothelial cells by Ang-2 may be an important early response during the development of retinal neovascularization. Inhibition of the binding activity of the angiopoietins in vivo suppressed retinal neovascularization concomitant with a reduction in the expression of MMP-9.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Angiopoietin-2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Neovascularization, Physiologic/physiology , Receptor, TIE-2/metabolism , Retinal Vessels/metabolism , Angiopoietin-1/pharmacology , Angiopoietin-2/pharmacology , Animals , Animals, Newborn , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/enzymology , Immunoglobulin Fc Fragments/pharmacology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Microcirculation , RNA, Messenger/metabolism , Receptor, TIE-2/immunology , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Up-Regulation
15.
Blood ; 100(1): 217-23, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12070030

ABSTRACT

CD40 is present on both normal and neoplastic B-lineage cells. CD40 stimulation of normal B cells has been shown to promote normal growth and differentiation, whereas aggressive histology B lymphomas are growth inhibited. The inhibition of neoplastic B-cell growth is believed to occur via activation-induced cell death in which stimuli that typically promote the growth of normal cells prevent the growth of their neoplastic counterparts. We show here that CD40 stimulation using either a soluble recombinant human CD40 ligand (srhCD40L) or anti-CD40 monoclonal antibody resulted in apoptosis of human Burkitt lymphoma cell lines. Additional studies examining the mechanism of CD40-mediated death revealed an increase in bax messenger RNA with a subsequent increase in Bax protein in the mitochondria of the treated cells. In vitro exposure of the cells to bax antisense oligonucleotides resulted in a significant decline in Bax protein levels and partial protection from CD40-mediated death, indicating that induction of Bax was at least one mechanism underlying this inhibitory effect of CD40 stimulation on lymphomas. When immunodeficient mice bearing Burkitt lymphoma were treated with srhCD40L, significant increases in survival were observed indicating a direct antitumor effect as a result of CD40 stimulation in vivo. Overall, these results demonstrate that CD40 ligation of aggressive histology B-lymphoma cells results in inhibition both in vitro and in vivo and thus may be of potential clinical use in their treatment.


Subject(s)
CD40 Antigens/physiology , CD40 Ligand/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins c-bcl-2 , Animals , CD40 Antigens/metabolism , CD40 Ligand/administration & dosage , Cell Death/drug effects , Gene Expression Regulation , Humans , Lymphocyte Activation/drug effects , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred NOD , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Survival Rate , Tumor Cells, Cultured/drug effects , bcl-2-Associated X Protein
16.
J Immunol ; 168(2): 661-70, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777959

ABSTRACT

Dendritic cells (DCs) are bone marrow-derived APCs that display unique properties aimed at stimulating naive T cells. Several members of the TNF/TNFR families have been implicated in T cell functions. In this study, we examined the role that Ox40 costimulation might play on the ability of DCs to regulate CD4(+) and CD8(+) T cell responses in vivo. Administration of anti-mouse Ox40 mAb enhanced the Th response induced by immunization with Ag-pulsed DCs, and introduced a bias toward a Th1 immune response. However, anti-Ox40 treatment enhanced the production of Th2 cytokines in IFN-gamma(-/-) mice after immunization with Ag-pulsed DCs, suggesting that the production of IFN-gamma during the immune response could interfere with the development of Th2 lymphocytes induced by DCs. Coadministration of anti-Ox40 with DCs during Ag rechallenge enhanced both Th1 and Th2 responses induced during a primary immunization with DCs, and did not reverse an existing Th2 response. This suggests that Ox40 costimulation amplifies an ongoing immune response, regardless of Th differentiation potential. In an OVA-TCR class II-restricted adoptive transfer system, anti-Ox40 treatment greatly enhanced the level of cytokine secretion per Ag-specific CD4(+) T cell induced by immunization with DCs. In an OVA-TCR class I-restricted adoptive transfer system, administration of anti-Ox40 strongly enhanced expansion, IFN-gamma secretion, and cytotoxic activity of Ag-specific CD8(+) T cells induced by immunization with DCs. Thus, by enhancing immune responses induced by DCs in vivo, the Ox40 pathway might be a target for immune intervention in therapeutic settings that use DCs as Ag-delivery vehicles.


Subject(s)
Dendritic Cells/transplantation , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Emulsions , Female , Hemocyanins/administration & dosage , Injections, Intravenous , Injections, Subcutaneous , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lymphocyte Activation/genetics , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/administration & dosage , Receptors, Antigen, T-Cell/deficiency , Receptors, Antigen, T-Cell/genetics , Receptors, OX40 , T-Lymphocyte Subsets/transplantation , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...