Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
J Histochem Cytochem ; 49(12): 1509-18, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11724898

ABSTRACT

We describe a novel fluorescent method for the detection of receptors for chimeric proteins in tissue sections. The technique was developed using a recombinant human insulin-like growth factor (IGF-1) chimera, bearing six additional histidine residues at the carboxy-terminal end (IGF-1-His). We demonstrated that dehydration of the tissue sections was detrimental for binding and that its prevention dramatically increased sensitivity. The specificity of IGF-1-His interaction was shown by gradual abolition of the fluorescent signal in the presence of increasing concentrations of IGF-1. Combining immunofluorescence with in situ ligand binding, we showed that IGF-1-His binding corresponded to the IGF-1 receptor (IGFR-1) distribution in human fetal kidney. Moreover, incubation of the tissue sections with an anti-IGFR-1 blocking antibody abolished IGF-1-His binding, demonstrating that the interaction was mediated by the IGFR-1. The method was also used to localize the IGFR-1 in E18 rat embryo sagittal sections. The IGF-1-His binding pattern was observed in brain, cartilage, lung, skin, heart, diaphragm, and tongue, and paralleled the previously reported IGFR-1 distribution. We believe that this new non-isotopic in situ ligand binding method will facilitate rapid and accurate localization of receptors in tissue sections.


Subject(s)
Insulin-Like Growth Factor I/genetics , Receptor, IGF Type 1/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Fetus , Fluorescent Antibody Technique , Frozen Sections , Histidine/genetics , Humans , Image Processing, Computer-Assisted , Kidney/metabolism , Ligands , Organ Specificity , Rats , Recombinant Fusion Proteins/genetics
2.
Breast Dis ; 11: 103-11, 2000.
Article in English | MEDLINE | ID: mdl-15687596
3.
Semin Oncol ; 26(4 Suppl 12): 60-70, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10482195

ABSTRACT

HER2 is a ligand-less member of the human epidermal growth factor receptor or ErbB family of tyrosine kinases. In normal biological systems, HER2 functions as a co-receptor for a multitude of epidermal growth factor-like ligands that bind and activate other HER family members. HER2 overexpression is observed in a number of human adenocarcinomas and results in constitutive HER2 activation. Specific targeting of these tumors can be accomplished with antibodies directed against the extracellular domain of the HER2 protein. One of these antibodies, 4D5, has been fully humanized and is termed trastuzumab (Herceptin; Genentech, San Francisco, CA). Treatment of HER2-overexpressing breast cancer cell lines with trastuzumab results in induction of p27KIP1 and the Rb-related protein, p130, which in turn significantly reduces the number of cells undergoing S-phase. A number of other phenotypic changes are observed in vitro as a consequence of trastuzumab binding to HER2-overexpressing cells. These phenotypic changes include downmodulation of the HER2 receptor, inhibition of tumor cell growth, reversed cytokine resistance, restored E-cadherin expression levels, and reduced vascular endothelial growth factor production. Interaction of trastuzumab with the human immune system via its human immunoglobulin G1 Fc domain may potentiate its antitumor activities. In vitro studies demonstrate that trastuzumab is very effective in mediating antibody-dependent cell-mediated cytotoxicity against HER2-overexpressing tumor targets. Trastuzumab treatment of mouse xenograft models results in marked suppression of tumor growth. When given in combination with standard cytotoxic chemotherapeutic agents, trastuzumab treatment generally results in statistically superior antitumor efficacy compared with either agent given alone. Taken together, these studies suggest that the mechanism of action of trastuzumab includes antagonizing the constitutive growth-signaling properties of the HER2 system, enlisting immune cells to attack and kill the tumor target, and augmenting chemotherapy-induced cytotoxicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Complement Activation , Down-Regulation , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Receptor, ErbB-2/biosynthesis , Trastuzumab
4.
Cancer Immunol Immunother ; 47(6): 337-42, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10203064

ABSTRACT

The HER2/neu oncogene product, p185(HER2/neu), is overexpressed on the surface of many human breast cancers. Strains of transgenic mice have been developed that express the rat neu oncogene in mammary epithelial cells and develop spontaneous mammary tumors that overexpress p185neu. This model provides an ideal system for testing interventions to prevent tumor development. In this study, we immunized neu-transgenic mice with a vaccine consisting of the extracellular domain of p185neu (NeuECD). Immunized mice developed Neu-specific humoral immune responses, as measured by circulating anti-Neu antibodies in their sera, and cellular immune responses, as measured by lymphocyte proliferation to NeuECD in vitro. In addition, the subsequent development of mammary tumors was significantly lower in immunized mice than in controls and vaccine treatment was associated with a significant increase in median survival.


Subject(s)
Cancer Vaccines/immunology , Genes, erbB-2/immunology , Mammary Neoplasms, Experimental/prevention & control , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , 3T3 Cells , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/blood , Antibody Formation/immunology , Cancer Vaccines/genetics , Cell Division/immunology , Extracellular Space/immunology , Female , Lymphocyte Activation/immunology , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Transgenic , Protein Structure, Tertiary , Rats , Receptor, ErbB-2/genetics
5.
Am J Pathol ; 151(6): 1523-30, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9403702

ABSTRACT

The overexpression in tumor cells of (proto)-oncogenic receptor tyrosine kinases such as epidermal growth factor receptor (EGFR) or ErbB2/neu (also known as HER-2) is generally thought to contribute to the development of solid tumors primarily through their effects on promoting uncontrolled cell proliferation. However, agents that antagonize the function of the protein products encoded by these (proto)-oncogenes are known to behave in vivo in a cytotoxic-like manner. This implies that such oncogenes may regulate critical cell survival functions, including angiogenesis. The latter could occur as a consequence of regulation of relevant growth factors by such oncogenes. We therefore sought to determine whether EGFR or ErbB2/neu may contribute to tumor angiogenesis by examining their effects on the expression of vascular endothelial cell growth factor (VEGF)/vascular permeability factor (VPF), one of the most important of all known inducers of tumor angiogenesis. We found that in vitro treatment of EGFR-positive A431 human epidermoid carcinoma cells, which are known to be heavily dependent on VEGF/VPF in vivo as an angiogenesis growth factor, with the C225 anti-EGFR neutralizing antibody caused a dose-dependent inhibition of VEGF protein expression. Prominent suppression of VEGF/VPF expression in vivo, as well as a significant reduction in tumor blood vessel counts, were also observed in established A431 tumors shortly after injection of the antibody as few as four times into nude mice. Transformation of NIH 3T3 fibroblasts with mutant ErbB2/neu, another EGFR-like oncogenic tyrosine kinase, resulted in a significant induction of VEGF/VPF, and the magnitude of this effect was further elevated by hypoxia. Moreover, treatment of ErbB2/neu-positive SKBR-3 human breast cancer cells in vitro with a specific neutralizing anti-ErbB2/neu monoclonal antibody (4D5) resulted in a dose-dependent reduction of VEGF/VPF protein expression. Taken together, the results suggest that oncogenic properties of EGFR and ErbB2/neu may, at least in part, be mediated by stimulation of tumor angiogenesis by up-regulating potent angiogenesis growth factors such as VEGF/VPF. These genetic changes may cooperate with epigenetic/environmental effects such as hypoxia to maximally stimulate VEGF/VPF expression. Therapeutic disruption of EGFR or ErbB2/neu protein function in vivo may therefore result in partial suppression of angiogenesis, a feature that could enhance the therapeutic index of such agents in vivo and endow them with anti-tumor effects, the magnitude of which may be out of proportion with their observed cytostatic effects in monolayer tissue culture.


Subject(s)
Adenocarcinoma/metabolism , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/metabolism , Endothelial Growth Factors/metabolism , Epidermal Growth Factor/immunology , Lymphokines/metabolism , Neovascularization, Pathologic/metabolism , Receptor, ErbB-2/immunology , Signal Transduction/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neutralization Tests , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
J Biol Chem ; 272(33): 20595-602, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9252374

ABSTRACT

Thrombopoietin (TPO) is a hematopoietin important for megakaryocyte proliferation and production of blood platelets. We sought to characterize how TPO binds and activates its receptor, myeloproliferative leukemia virus receptor. The erythropoietin-like domain of TPO (TPO1-153) has been fused to the gIII coat protein of M13 bacteriophage. Forty residues were chosen for mutation to alanine using the criteria that they were charged residues or predicted to be solvent-exposed, based on a homology model. Phage enzyme-linked immunosorbent assay was used to determine affinities for binding to both the TPO receptor and five anti-TPO1-153 monoclonal antibodies. Mutations at mostly positively charged residues (Asp8, Lys14, Lys52, Lys59, Lys136, Lys138, Arg140) caused the greatest reduction in receptor-binding affinity. Most of these residues mapped to helices-1 and -4 and a loop region between helix-1 and helix-2. Two of the monoclonal antibodies that blocked TPO binding and bioactivity had determinants in helix-4. In contrast, the other three monoclonal antibodies, which were effective at blocking TPO activity but did not block initial binding of TPO to its receptor, had epitopes predominantly on helix or 3. These results suggest that TPO has two distinct receptor-binding sites that function to dimerize TPO receptors in a sequential fashion.


Subject(s)
Epitope Mapping , Neoplasm Proteins , Proto-Oncogene Proteins/analysis , Receptors, Cytokine , Thrombopoietin/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Binding Sites , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Receptors, Thrombopoietin , Sequence Alignment , Thrombopoietin/immunology , Thrombopoietin/metabolism
7.
Cancer Res ; 56(6): 1457-65, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8640840

ABSTRACT

Alterations in the expression of the epidermal growth factor (EGF) receptor ErbB family are frequently encountered in a number of human cancers. Two of these receptors, ErbB3 and ErbB4, are known to bind a family of related proteins termed heregulins (HRGs) or neu differentiation factors. In biologically relevant systems, interaction of HRG with ErbB3 or ErbB4 results in the transactivation of ErbB2. In this report, we show that ErbB2 is a critical component in mediating HRG responsiveness in a panel of human breast and ovarian tumor cell lines. Because HRGs have been reported to elicit diverse biological effects on cultured cells, including growth stimulation, growth inhibition, and induction of differentiation, we systematically examined the effect of rHRG beta 1 on tumor cell proliferation. HRG binding studies were performed with a panel of breast and ovarian tumor cell lines expressing a range of levels of ErbB2. The biological responses to HRG were also compared to EGF and to the growth-inhibitory anti-ErbB2 antibody, 4D5. In most cases, HRG stimulation of DNA synthesis correlated with positive effects on cell cycle progression and cell number and with enhancement of colony formation in soft agar. On each cell line tested, the HRG effects were distinguishable from EGF and 4D5. Our findings indicate that HRG induces cell proliferation in a number of tumor cell lines. In addition, we show that methods for measuring cell proliferation, as well as experimental conditions, are critical for determining HRGs effect on tumor cell growth in vitro.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Glycoproteins/metabolism , Neuregulin-1 , Ovarian Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/metabolism , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Count/drug effects , Cell Cycle/drug effects , Cell Division , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Humans , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-3 , Tumor Cells, Cultured
8.
Hybridoma ; 14(3): 253-60, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7590788

ABSTRACT

Transforming growth factor beta (TGF-beta) is a multifunctional peptide that controls proliferation, differentiation, and other functions in a variety of cell types. Transforming growth factor beta activities have been implicated in a variety of diseased states including arthritis, prostate cancer, and AIDS, and in the repair of tissue injury caused by trauma, burns, and surgery. We describe the development and characterization of novel murine monoclonal antibodies (MAbs) to the latency-associated peptide (LAP) of TGF-beta 1, and the subsequent development of an ELISA for the detection and quantitation of TGF-beta 1-LAP in buffer and serum matrices. Fusion of immune splenocytes with myeloma cells yielded 576 hybridomas, 110 of which were antibody secreting. Five were selected for extensive characterization. Clinically, the MAbs described here should be valuable for studying potentially abnormal production and/or function of the LAP, and its relationship to TGF-beta.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Peptide Fragments , Protein Precursors , Proteins/immunology , Transforming Growth Factor beta/immunology , Animals , Antibody Specificity , Buffers , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Mice , Mice, Inbred BALB C , Proteins/isolation & purification , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta1
9.
Int Arch Allergy Immunol ; 107(1-3): 308-12, 1995.
Article in English | MEDLINE | ID: mdl-7613156

ABSTRACT

Numerous clinical studies show that direct interference with the IgE response leads to a decrease or elimination of allergic symptoms. The aim of these studies was to design a therapy aimed at decreasing IgE levels in order to ameliorate atopic disease. To this end, a murine monoclonal antibody, MAE11, directed against IgE was identified, which had all the properties necessary to interfere with IgE responses, but lacked the harmful side effects of inducing receptor cross-linking. The antibody was selected on the basis of its ability to bind circulating IgE at the same site as the high-affinity receptor, thus blocking the binding of IgE to mast cells and basophils. To allow for possible chronic administration and to avoid the problems of antigenicity, MAE11 was humanized. The best of several humanized variants, version 25 (rhumAb-E25) was selected since it possessed binding affinity and biological activity comparable to MAE11. Clinical studies are underway to determine the safety and efficacy of this treatment for allergic rhinitis and asthma.


Subject(s)
Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunization, Passive , Immunoglobulin E/immunology , Rhinitis, Allergic, Seasonal/therapy , Allergens/immunology , Allergens/toxicity , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Basophil Degranulation Test , Basophils/metabolism , Humans , Intradermal Tests , Macaca fascicularis , Mice , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology
11.
J Biol Chem ; 270(9): 4668-74, 1995 Mar 03.
Article in English | MEDLINE | ID: mdl-7876238

ABSTRACT

The most potent known agonist for the natriuretic peptide receptor-B (NPR-B)/guanylyl cyclase-B is C-type natriuretic peptide (CNP). A homologous ligand-receptor system consists of atrial natriuretic peptide (ANP) and NPR-A/guanylyl cyclase-A. A third member of this family is NPR-C, a non-guanylyl cyclase receptor. Monoclonal antibodies were raised against NPR-B by immunizing mice with a purified receptor-IgG fusion protein consisting of the extracellular domain of NPR-B and the Fc portion of human IgG-gamma 1. One monoclonal antibody, 3G12, did not recognize NPR-A or NPR-C and bound to human and rat NPR-B. CNP binding to NPR-B and stimulation of cGMP synthesis were inhibited by 3G12. With cells isolated from either the media or adventitia layers of rat thoracic aorta, 3G12 did not interfere with ANP-stimulated cGMP synthesis, but it inhibited CNP-stimulated cGMP levels in cells from both layers. CNP (IC50 = 10 nM) and ANP (IC50 = 1 nM) caused relaxation of phenylephrine-contracted rat aortic rings. 3G12 caused a marked increase in the IC50 for CNP, from 10 nM to 140 nM, but failed to affect ANP-mediated relaxation. Therefore, our results for the first time demonstrate that CNP relaxes vascular smooth muscle by virtue of its binding to NPR-B.


Subject(s)
Aorta/physiology , Guanylate Cyclase/physiology , Proteins/physiology , Receptors, Atrial Natriuretic Factor/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Line , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Muscle Relaxation/physiology , Natriuretic Peptide, C-Type , Protein Binding , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/metabolism
12.
Proc Natl Acad Sci U S A ; 92(6): 1866-70, 1995 Mar 14.
Article in English | MEDLINE | ID: mdl-7534404

ABSTRACT

Htk is a receptor protein-tyrosine kinase that is related to the EPH subfamily of tyrosine kinases. The receptor has a wide tissue distribution including expression in several myeloid hematopoietic cell lines. Using an Htk-Fc fusion protein, a protein ligand for this receptor was expression cloned from the murine kidney mesangial cell line SV40MES 13. The Htk ligand cDNA encodes a transmembrane protein of 336 amino acids. Binding competition experiments demonstrated a Kd of 535 pM for binding of Htk-Fc to the Htk ligand. Incubation of 3T3 cells expressing Htk with COS-7 cells expressing the ligand resulted in tyrosine phosphorylation of Htk. The ligand, like its receptor, is widely expressed and may function in a variety of tissues. However, we localized hematopoietic expression of Htk to the monocytic lineage, suggesting that the ligand may play a role in differentiation and/or proliferation of these cells.


Subject(s)
Membrane Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Adult , Amino Acid Sequence , Animals , Binding, Competitive , Blotting, Northern , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Ephrin-B2 , Female , Fetal Blood , Fetus , Glomerular Mesangium/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Kidney , Kinetics , Ligands , Male , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Organ Specificity , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine , Pregnancy , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, EphB4 , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , Tyrosine/analogs & derivatives , Tyrosine/analysis
13.
J Cell Physiol ; 161(3): 597-605, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7962141

ABSTRACT

The cytotoxic effects of TNF on malignant cells are known to be mediated through high affinity surface receptors. The precise mechanism by which transformed cells are selectively killed by the activation of these receptors is yet unknown, but several intracellular signaling pathways are known to be involved. Phospholipase A2 activation by TNF-alpha has been shown to be important in the transduction of signals leading to cell death. We have used monitoring of extracellular acidification rate as a measure of cellular metabolism to follow the early time course of TNF effects on a human leukemic T cell line (CEM-SS cells). CEM-SS cells were relatively resistant to TNF cell killing but TNF caused an early stimulation of metabolism within 2-4 hr, followed by a suppression of metabolic activity occurring over 20 hr. In contrast, a TNF sensitive subclone of CEM cells (C1Ca) showed a rapid and dramatic decrease in metabolic activity corresponding to cytotoxicity within 18 hr. It was discovered that cupric o-phenanthroline markedly potentiated the effects of TNF on the resistant CEM-SS cells leading to cell death. This observation was specific for copper because ferric o-phenanthroline was without effect at the same concentration. The copper cytotoxic effect was shown to be mediated through the TNF-R1 receptor and independent of phospholipase A2 signaling.


Subject(s)
Cell Survival/drug effects , Copper/administration & dosage , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Animals , Cell Line , Drug Synergism , Flow Cytometry , Hydrogen-Ion Concentration , Leukemia, T-Cell , Metals/pharmacology , Oxidation-Reduction , Phenanthrolines/pharmacology , Quinacrine/pharmacology , Rats , Reactive Oxygen Species , Receptor Aggregation , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/toxicity
14.
Oncogene ; 9(7): 1829-38, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7911565

ABSTRACT

Approximately 30% of human breast and ovarian cancers have amplification and/or overexpression of HER-2/neu gene which encodes a cell surface growth-factor receptor. Overexpression of this receptor, p185HER-2/neu, is associated with poor outcome and may predict clinical response to chemotherapy. Antibodies to HER-2/neu receptor have a cytostatic effect in suppressing growth of cells with overexpression of p185HER-2/neu. To elicit a cytocidal effect, therapy with antireceptor antibody was used in combination with the DNA-damaging drug, cisplatin, and this combined treatment produced a synergistic decrease in cell growth. In addition, antibody mediated an increased sensitivity to cisplatin in drug-resistant ovarian carcinoma cells containing multiple copies of HER-2/neu gene. To evaluate the mechanism for this synergy, unscheduled DNA synthesis was measured in cancer cells using incorporation of [3H]thymidine and autoradiography, and formation and repair of cisplatin-induced DNA adducts was also measured. Treatment with cisplatin led to a marked, dose-dependent increase in unscheduled DNA synthesis which was significantly reduced by combined treatment with antireceptor antibody in HER-2/neu-overexpressing cells. Therapy with antibody to HER-2/neu receptor also led to a 35-40% reduction in repair of cisplatin-DNA adducts after cisplatin exposure and, as a result, promoted drug-induced killing in target cells. This phenomenon which we term receptor-enhanced chemosensitivity may provide a rationale for more selective targeting and exploitation of overexpressed growth factor receptors in cancer cells, thus leading to new strategies for clinical intervention.


Subject(s)
Antibodies, Neoplasm/immunology , Breast Neoplasms/pathology , Cisplatin/pharmacology , DNA Adducts , DNA Repair , ErbB Receptors/immunology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/immunology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , DNA , DNA Repair/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , Drug Resistance , ErbB Receptors/genetics , Female , Gene Expression , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2 , Tumor Cells, Cultured
15.
J Biol Chem ; 269(20): 14661-5, 1994 May 20.
Article in English | MEDLINE | ID: mdl-7514177

ABSTRACT

The heregulin/neu differentiation factor gene products were purified and cloned based on their ability to stimulate the phosphorylation of a 185-kDa protein in human breast carcinoma cell lines known to express erbB2. However, not all cells that express erbB2 respond to heregulin, indicating that other components besides erbB2 may be required for heregulin binding. Cells that are transfected with the closely related receptor, erbB3, display a single class of lower affinity heregulin binding sites than has been previously observed on breast carcinoma cell lines. Little or no stimulation of tyrosine phosphorylation in response to heregulin occurs in cells that are transfected with erbB3 alone. Transfection of cells with erbB3 and erbB2 reconstitutes a higher affinity binding receptor, which is also capable of generating a tyrosine phosphorylation signal in response to heregulin. A monoclonal antibody to erbB2 will inhibit heregulin activation of tyrosine phosphorylation and binding in cells transfected with both receptors but not with erbB3 alone. In cells expressing erbB2 and erbB3, both proteins become tyrosine-phosphorylated upon interaction with heregulin. Direct interaction between heregulin and the two proteins was demonstrated by chemical cross-linking experiments using 125I-heregulin followed by immunoprecipitation with antibodies specific for erbB2 or erbB3.


Subject(s)
Carrier Proteins/metabolism , ErbB Receptors/metabolism , Gene Expression , Glycoproteins/metabolism , Neuregulin-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Animals , Carrier Proteins/biosynthesis , Cell Line , Chlorocebus aethiops , Cross-Linking Reagents , ErbB Receptors/analysis , ErbB Receptors/biosynthesis , Glycoproteins/biosynthesis , Humans , Iodine Radioisotopes , Kinetics , Neuregulins , Phosphorylation , Phosphotyrosine , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/biosynthesis , Radioligand Assay , Receptor, ErbB-2 , Receptor, ErbB-3 , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
J Clin Invest ; 92(6): 2569-76, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7504687

ABSTRACT

TGF-beta effects on angiogenesis, stroma formation, and immune function suggest its possible involvement in tumor progression. This hypothesis was tested using the 2G7 IgG2b, which neutralizes TGF-beta 1, -beta 2, and -beta 3, and the MDA-231 human breast cancer cell line. Inoculation of these cells in athymic mice decreases mouse spleen natural killer (NK) cell activity. Intraperitoneal injections of 2G7 starting 1 d after intraperitoneal inoculation of tumor cells suppressed intraabdominal tumor and lung metastases, whereas the nonneutralizing anti-TGF-beta 12H5 IgG2a had no effect. 2G7 transiently inhibited growth of established MDA-231 subcutaneous tumors. Histologically, both 2G7-treated and control tumors were identical. Intraperitoneal administration of 2G7 resulted in a marked increase in mouse spleen NK cell activity. 2G7 did not inhibit MDA-231 primary tumor or metastases formation, nor did it stimulate NK cell-mediated cytotoxicity in beige NK-deficient nude mice. Finally, serum-free conditioned medium from MDA-231 cells inhibited the NK cell activity of human blood lymphocytes. This inhibition was blocked by the neutralizing anti-TGF-beta 2G7 antibody but not by a nonspecific IgG2. These data support a possible role for tumor cell TGF-beta in the progression of mammary carcinomas by suppressing host immune surveillance.


Subject(s)
Antibodies/pharmacology , Breast Neoplasms/pathology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/physiology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/immunology , Cell Division , Collagen/analysis , Collagen/metabolism , Factor VIII/analysis , Factor VIII/metabolism , Female , Humans , Immunoglobulin G/classification , Immunoglobulin G/pharmacology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Recombinant Proteins/pharmacology , Spleen/immunology , Transforming Growth Factor beta/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured
17.
J Immunol ; 151(9): 4637-41, 1993 Nov 01.
Article in English | MEDLINE | ID: mdl-8409424

ABSTRACT

TNF-alpha can enhance the proliferation of human thymocytes stimulated by the comitogen Con A. To determine which of the two different TNF receptors is responsible for signaling this cellular response, we investigated the proliferation of human thymocytes in response to agonistic antibodies specific for the two TNF receptor types. In contrast to previously examined TNF activities in human cells, thymocyte proliferation was stimulated in response to rabbit polyclonal antibodies directed against the 75-kDa TNF receptor (TNF-R2), but not those directed against the 55-kDa TNF receptor (TNF-R1). Lymphotoxin (TNF-beta) was also shown to stimulate human thymocyte proliferation, demonstrating that TNF-beta can initiate a biologic response that is mediated by TNF-R2. TNF-R2-mediated T-cell proliferation was not restricted to the immature T cells within the thymus, as the anti-TNF-R2 antibodies also stimulated the proliferation of peripheral T cells. As a first step toward identifying a specific agonist of TNF-R2 with therapeutic potential, 10 anti-TNF-R2 mAb were examined for potential agonist activity. Nine of these significantly stimulated human thymocyte proliferation with maximal responses ranging from twofold to significantly greater than that obtained with TNF-alpha by itself.


Subject(s)
Lymphocyte Activation , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes/immunology , Antibodies, Monoclonal/immunology , Cells, Cultured , Child, Preschool , Humans , Infant , Molecular Weight , Tumor Necrosis Factor-alpha/pharmacology
18.
J Virol ; 67(10): 6179-91, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7690420

ABSTRACT

The binding properties of seven CD4-blocking monoclonal antibodies raised against recombinant gp120 of human immunodeficiency virus type 1 strain MN (HIV-1MN) and two CD4-blocking monoclonal antibodies to recombinant envelope glycoproteins gp120 and gp160 of substrain IIIB of HIVLAI were analyzed. With a panel of recombinant gp120s from seven diverse HIV-1 isolates, eight of the nine antibodies were found to be strain specific and one was broadly cross-reactive. Epitope mapping revealed that all nine antibodies bound to epitopes located in the fourth conserved domain (C4) of gp120. Within this region, three distinct epitopes could be identified: two were polymorphic between HIV-1 strains, and one was highly conserved. Studies with synthetic peptides demonstrated that the conserved epitope, recognized by antibody 13H8, was located between residues 431 and 439. Site-directed mutagenesis of gp120 demonstrated that residue 429 and/or 432 was critical for the binding of the seven antibodies to gp120 from HIV-1MN. Similarly, residues 423 and 429 were essential for the binding of monoclonal antibody 5C2 raised against gp120 from HIV-1IIIB. The amino acids located at positions 423 and 429 were found to vary between strains of HIV-1 as well as between molecular clones derived from the MN and LAI isolates of HIV-1. Polymorphism at these positions prevented the binding of virus-neutralizing monoclonal antibodies and raised the possibility that HIV-1 neutralization serotypes may be defined on the basis of C4 domain sequences. Analysis of the binding characteristics of the CD4-blocking antibodies demonstrated that their virus-neutralizing activity was directly proportional to their gp120-binding affinity. These studies account for the strain specificity of antibodies to the C4 domain of gp120 and demonstrate for the first time that antibodies to this region can be as effective as those directed to the principal neutralizing determinant (V3 domain) in neutralizing HIV-1 infectivity.


Subject(s)
Antibodies, Monoclonal/metabolism , CD4 Antigens/metabolism , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , HIV-1/immunology , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites , Binding Sites, Antibody , CD4 Antigens/immunology , Cell Line , Cross Reactions , Disulfides/analysis , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , HIV Envelope Protein gp120/immunology , HIV-1/genetics , Humans , Kinetics , Models, Structural , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutralization Tests , Polymerase Chain Reaction , Polymorphism, Genetic , Species Specificity
19.
Am J Respir Cell Mol Biol ; 9(4): 448-54, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8104437

ABSTRACT

p185HER2, the product of the c-erbB-2 or HER2 gene, is a membrane-bound tyrosine kinase that has structural similarity to the epidermal growth factor receptor. Functionally, interaction of HER2 with its ligand or p185HER2 antibodies affects the growth and differentiation of HER2-expressing breast cancer cell lines. As p185HER2 is also expressed in human lung cancers and human lung cancer cell lines, we hypothesized that these cell lines would also respond to p185HER2 antibodies. To test this hypothesis, we cultured human non-small cell lung cancer cell lines in the presence of a p185HER2 monoclonal antibody called 4D5. 4D5 inhibited the growth of p185HER2-expressing cell lines in a dose-dependent fashion. In addition, BEAS.2B, a p185HER2-nonexpressing bronchial epithelial cell line, was transfected with the HER2 cDNA, resulting in high-level p185HER2 expression, and growth of BEAS.HER2 was now inhibited by 4D5 exposure. Mechanistically, 4D5 appeared to have a weak agonist effect on the tyrosine kinase function of p185HER2, as exposure of p185HER2-expressing cell lines to 4D5 resulted in increased p185HER2 phosphorylation. Furthermore, inhibition of tyrosine kinase function with Genistein reversed the 4D5-induced growth inhibition. Therefore, 4D5 can regulate the growth of p185HER2-expressing lung cancer cell lines through agonist effects on p185HER2.


Subject(s)
ErbB Receptors/physiology , Lung Neoplasms/etiology , Proto-Oncogene Proteins/physiology , Adenocarcinoma , Antibodies, Neoplasm/immunology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/physiology , Bronchi/cytology , Cell Division , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Humans , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Receptor, ErbB-2 , Transfection , Tumor Cells, Cultured
20.
Cancer Immunol Immunother ; 37(4): 255-63, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8102322

ABSTRACT

The HER2 protooncogene encodes a receptor tyrosine kinase, p185HER2. The overexpression of p185HER2 has been associated with a worsened prognosis in certain human cancers. In the present work we have screened a variety of different tumor cell lines for p185HER2 expression using both enzyme-linked immunosorbent and fluorescence-activated cell sorting assays employing murine monoclonal antibodies directed against the extracellular domain of the receptor. Increased levels of p185HER2 were found in breast (5/9), ovarian (1/6), stomach (2/3) and colorectal (5/16) carcinomas, whereas all kidney and submaxillary adenocarcinoma cell lines tested were negative. Some monoclonal antibodies directed against the extracellular domain of p185HER2 inhibited growth in monolayer culture of breast and ovarian tumor cell lines overexpressing p185HER2, but had no effect on the growth of colon or gastric adenocarcinomas expressing increased levels of this receptor. The most potent growth-inhibitory anti-p185HER2 monoclonal antibody in monolayer culture, designated mumAb 4D5 (a murine IgG1 kappa antibody), was also tested in soft-agar growth assays for activity against p185HER2-overexpressing tumor cell lines of each type, with similar results. In order to increase the spectrum of tumor types potentially susceptible to monoclonal antibody-mediated anti-p185HER2 therapies, to decrease potential immunogenicity issues with the use of murine monoclonal antibodies for human therapy, and to provide the potential for antibody-mediated cytotoxic activity, a mouse/human chimeric 4D5 (chmAb 4D5) and a "humanized" 4D5 (rhu)mAb 4D5 HER2 antibody were constructed. Both engineered antibodies, in combination with human peripheral blood mononuclear cells, elicited antibody-dependent cytotoxic responses in accordance with the level of p185HER2 expression. Since this cytotoxic activity is independent of sensitivity to mumAb 4D5, the engineered monoclonal antibodies expand the potential target population for antibody-mediated therapy of human cancers characterized by the overexpression of p185HER2.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/immunology , Proto-Oncogene Proteins/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Division/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Proto-Oncogene Proteins/biosynthesis , Receptor, ErbB-2 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...