21 results on '"Lofgren JA"'
Search Results
2. Bispecific T cell engager (BiTE®) antibody constructs can mediate bystander tumor cell killing.
- Author
-
Ross SL, Sherman M, McElroy PL, Lofgren JA, Moody G, Baeuerle PA, Coxon A, and Arvedson T
- Subjects
- Animals, Coculture Techniques, Cytokines metabolism, Cytotoxicity, Immunologic, ErbB Receptors metabolism, Female, Heterografts, Humans, Lymphocyte Activation, Mice, Mice, Nude, Antibodies, Bispecific immunology, Bystander Effect, Neoplasms pathology, T-Lymphocytes immunology
- Abstract
For targets that are homogenously expressed, such as CD19 on cells of the B lymphocyte lineage, immunotherapies can be highly effective. Targeting CD19 with blinatumomab, a CD19/CD3 bispecific antibody construct (BiTE®), or with chimeric antigen receptor T cells (CAR-T) has shown great promise for treating certain CD19-positive hematological malignancies. In contrast, solid tumors with heterogeneous expression of the tumor-associated antigen (TAA) may present a challenge for targeted therapies. To prevent escape of TAA-negative cancer cells, immunotherapies with a local bystander effect would be beneficial. As a model to investigate BiTE®-mediated bystander killing in the solid tumor setting, we used epidermal growth factor receptor (EGFR) as a target. We measured lysis of EGFR-negative populations in vitro and in vivo when co-cultured with EGFR-positive cells, human T cells and an EGFR/CD3 BiTE® antibody construct. Bystander EGFR-negative cells were efficiently lysed by BiTE®-activated T cells only when proximal to EGFR-positive cells. Our mechanistic analysis suggests that cytokines released by BiTE®-activated T-cells induced upregulation of ICAM-1 and FAS on EGFR-negative bystander cells, contributing to T cell-induced bystander cell lysis.
- Published
- 2017
- Full Text
- View/download PDF
3. MDM2 antagonists synergize broadly and robustly with compounds targeting fundamental oncogenic signaling pathways.
- Author
-
Saiki AY, Caenepeel S, Yu D, Lofgren JA, Osgood T, Robertson R, Canon J, Su C, Jones A, Zhao X, Deshpande C, Payton M, Ledell J, Hughes PE, and Oliner JD
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Drug Screening Assays, Antitumor, Drug Synergism, Gene Expression drug effects, Humans, Antineoplastic Combined Chemotherapy Protocols pharmacology, Neoplasms metabolism, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Signal Transduction drug effects
- Abstract
While MDM2 inhibitors hold great promise as cancer therapeutics, drug resistance will likely limit their efficacy as single agents. To identify drug combinations that might circumvent resistance, we screened for agents that could synergize with MDM2 inhibition in the suppression of cell viability. We observed broad and robust synergy when combining MDM2 antagonists with either MEK or PI3K inhibitors. Synergy was not limited to cell lines harboring MAPK or PI3K pathway mutations, nor did it depend on which node of the PI3K axis was targeted. MDM2 inhibitors also synergized strongly with BH3 mimetics, BCR-ABL antagonists, and HDAC inhibitors. MDM2 inhibitor-mediated synergy with agents targeting these mechanisms was much more prevalent than previously appreciated, implying that clinical translation of these combinations could have far-reaching implications for public health. These findings highlight the importance of combinatorial drug targeting and provide a framework for the rational design of MDM2 inhibitor clinical trials.
- Published
- 2014
- Full Text
- View/download PDF
4. Design and synthesis of novel amide AKT1 inhibitors with selectivity over CDK2.
- Author
-
Ashton KS, St Jean DJ Jr, Poon SF, Lee MR, Allen JG, Zhang S, Lofgren JA, Zhang X, Fotsch C, and Hungate R
- Subjects
- Amides chemical synthesis, Amides chemistry, Chemistry Techniques, Synthetic, Crystallography, X-Ray, Cyclin-Dependent Kinase 2 metabolism, Dose-Response Relationship, Drug, Enzyme Inhibitors chemistry, Models, Molecular, Molecular Structure, Proto-Oncogene Proteins c-akt metabolism, Stereoisomerism, Structure-Activity Relationship, Amides pharmacology, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Drug Design, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors
- Abstract
Through the analysis of X-ray crystallographic information and previous SAR studies, a novel series of protein kinase B (PKB/AKT) inhibitors was developed. The compounds showed nanomolar inhibition of AKT1 and were selective against cyclin-dependent kinase 2 (CDK2)., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
5. Azole-based inhibitors of AKT/PKB for the treatment of cancer.
- Author
-
Zeng Q, Allen JG, Bourbeau MP, Wang X, Yao G, Tadesse S, Rider JT, Yuan CC, Hong FT, Lee MR, Zhang S, Lofgren JA, Freeman DJ, Yang S, Li C, Tominey E, Huang X, Hoffman D, Yamane HK, Fotsch C, Dominguez C, Hungate R, and Zhang X
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Azoles pharmacokinetics, Azoles therapeutic use, Binding Sites, Crystallography, X-Ray, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Cyclin-Dependent Kinase 2 metabolism, Drug Design, Mice, Mice, Nude, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-akt metabolism, Rats, Rats, Sprague-Dawley, Structure-Activity Relationship, Xenograft Model Antitumor Assays, Antineoplastic Agents chemistry, Azoles chemistry, Neoplasms drug therapy, Protein Kinase Inhibitors chemistry, Proto-Oncogene Proteins c-akt antagonists & inhibitors
- Abstract
Through a combination of screening and structure-based rational design, we have discovered a series of N(1)-(5-(heterocyclyl)-thiazol-2-yl)-3-(4-trifluoromethylphenyl)-1,2-propanediamines that were developed into potent ATP competitive inhibitors of AKT. Studies of linker strand-binding adenine isosteres identified SAR trends in potency and selectivity that were consistent with binding interactions observed in structures of the inhibitors bound to AKT1 and to the counter-screening target PKA. One compound was shown to have acceptable pharmacokinetic properties and to be a potent inhibitor of AKT signaling and of in vivo xenograft tumor growth in a preclinical model of glioblastoma., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
6. Comparing ELISA and surface plasmon resonance for assessing clinical immunogenicity of panitumumab.
- Author
-
Lofgren JA, Dhandapani S, Pennucci JJ, Abbott CM, Mytych DT, Kaliyaperumal A, Swanson SJ, and Mullenix MC
- Subjects
- Animals, Antibodies, Anti-Idiotypic biosynthesis, Antibodies, Anti-Idiotypic blood, Antibodies, Monoclonal metabolism, Antibody Affinity, Cell Line, Tumor, Dose-Response Relationship, Immunologic, Enzyme-Linked Immunosorbent Assay standards, Epitope Mapping, ErbB Receptors immunology, Female, Humans, Immunoglobulin G biosynthesis, Immunoglobulin G metabolism, Mice, Mice, Inbred NOD, Panitumumab, Sensitivity and Specificity, Surface Plasmon Resonance standards, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal immunology, Enzyme-Linked Immunosorbent Assay methods, Surface Plasmon Resonance methods
- Abstract
Evaluation of the immunogenicity of panitumumab, a fully human anti-epidermal growth factor receptor mAb approved for use in colorectal cancer patients, led to the development of two separate immunoassays for the detection of anti-panitumumab Abs. The first immunoassay used a bridging ELISA capable of detecting 10 ng/ml positive control anti-panitumumab Ab. The ELISA incorporated an acid dissociation step to reduce drug interference and tolerated the presence of approximately 100-fold molar excess of drug. During eight clinical trials, the ELISA detected developing Ab responses in 2 of 612 (0.3%) subjects. In one of the ELISA positive subjects, neutralizing Abs were detected using an epidermal growth factor receptor phosphorylation bioassay. The second immunoassay used a Biacore biosensor immunoassay format capable of detecting 1 mug/ml positive control Ab while tolerating the presence of equal molar amounts of drug. Although less sensitive and less tolerant to competing drug in the assay, the Biacore assay detected developing Ab responses in 25 of the 604 (4.1%) subjects. Additionally, the Biacore assay identified eight subjects who developed neutralizing Abs. Mouse mAbs with affinities ranging from 1.1 x 10(-6) to 8.4 x 10(-10) M were used to characterize both assay types. The ELISA was more sensitive for the detection of higher affinity mAbs and detected high-affinity mAbs in the presence of higher molar ratio of drug to mAb. The Biacore assay was more sensitive for detection of lower affinity mAbs and detected low affinity Abs in the presence of higher molar ratios of drug to mAb.
- Published
- 2007
- Full Text
- View/download PDF
7. Kinetic mechanism of AKT/PKB enzyme family.
- Author
-
Zhang X, Zhang S, Yamane H, Wahl R, Ali A, Lofgren JA, and Kendall RL
- Subjects
- Adenosine Triphosphate chemistry, Animals, Catalysis, Humans, Kinetics, Peptides chemistry, Phosphatidylinositol 3-Kinases metabolism, Protein Binding, Protein Isoforms, Rats, Proto-Oncogene Proteins c-akt metabolism
- Abstract
AKT/PKB is a phosphoinositide-dependent serine/threonine protein kinase that plays a critical role in the signal transduction of receptors. It also serves as an oncogene in the tumorigenesis of cancer cells when aberrantly activated by genetic lesions of the PTEN tumor suppressor, phosphatidylinositol 3-kinase, and receptor tyrosine kinase overexpression. Here we have characterized and compared kinetic mechanisms of the three AKT isoforms. Initial velocity studies revealed that all AKT isozymes follow the sequential kinetic mechanism by which an enzyme-substrate ternary complex forms before the product release. The empirically derived kinetic parameters are apparently different among the isoforms. AKT2 showed the highest Km value for ATP, and AKT3 showed the highest kcat value. The patterns of product inhibition of AKT1, AKT2, and AKT3 by ADP were all consistent with an ordered substrate addition mechanism with ATP binding to the enzymes prior to the peptide substrate. Further analysis of steady state kinetics of AKT1 in the presence of dead-end inhibitors supported the finding and suggested that the AKT family of kinases catalyzes reactions via an Ordered Bi Bi sequential mechanism with ATP binding to the enzyme prior to peptide substrate and ADP being released after the phosphopeptide product. These results suggest that ATP is an initiating factor for the catalysis of AKT enzymes and may play a role in the regulation AKT enzyme activity in cells.
- Published
- 2006
- Full Text
- View/download PDF
8. Detection of neutralizing anti-therapeutic protein antibodies in serum or plasma samples containing high levels of the therapeutic protein.
- Author
-
Lofgren JA, Wala I, Koren E, Swanson SJ, and Jing S
- Subjects
- Antibodies, Monoclonal blood, Antigen-Antibody Complex blood, Antigen-Antibody Complex isolation & purification, Cell Line, Humans, Hydrogen-Ion Concentration, Immunosorbent Techniques, In Vitro Techniques, Interleukin-8 biosynthesis, Receptors, Interleukin-1 antagonists & inhibitors, Receptors, Interleukin-1 immunology, Recombinant Proteins adverse effects, Recombinant Proteins therapeutic use, Antibodies blood, Neutralization Tests methods, Recombinant Proteins blood, Recombinant Proteins immunology
- Abstract
Neutralizing antibodies against therapeutic proteins can be potentially harmful if the antibody blocks not only the therapeutic activities of the therapeutic protein but also the normal functions of the endogenous counterpart. Detection of the neutralizing anti-therapeutic protein antibodies generally relies on bioassays measuring changes in the biologic activity of the therapeutic protein triggered by the presence of the antibody. Most of the bioassays, particularly the cell-based in vitro assays, fail to detect neutralizing anti-therapeutic protein antibodies when the remaining therapeutic protein level in the assay samples is high. The remaining therapeutic protein, either a free molecule or an immune complex with anti-therapeutic protein antibodies, can inhibit the neutralizing activity of the antibody and prevent detection. We describe the development of a procedure that uses acid dissociation and affinity adsorption to remove therapeutic protein from assay samples. With this procedure, we can detect the presence of neutralizing anti-therapeutic protein antibodies from samples containing high levels of therapeutic protein.
- Published
- 2006
- Full Text
- View/download PDF
9. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth.
- Author
-
Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI, Lofgren JA, Tindell C, Evans DP, Maiese K, Scher HI, and Sliwkowski MX
- Subjects
- Androgens metabolism, Animals, Antibodies, Monoclonal immunology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Breast Neoplasms metabolism, Cell Division drug effects, Female, Gene Expression Regulation, Neoplastic, Humans, Ligands, Male, Mice, Neoplasm Transplantation, Neuregulin-1 pharmacology, Prostatic Neoplasms metabolism, Protein Binding drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology, Receptor, ErbB-2 metabolism, Receptor, ErbB-3 metabolism, Time Factors, Transplantation, Heterologous, Tumor Cells, Cultured, Antibodies, Monoclonal therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Receptor, ErbB-2 antagonists & inhibitors, Signal Transduction drug effects
- Abstract
ErbB2 is a ligand-less member of the ErbB receptor family that functions as a coreceptor with EGFR, ErbB3, and ErbB4. Here, we describe an approach to target ErbB2's role as a coreceptor using a monoclonal antibody, 2C4, which sterically hinders ErbB2's recruitment into ErbB ligand complexes. Inhibition of ligand-dependent ErbB2 signaling by 2C4 occurs in both low- and high-ErbB2-expressing systems. Since the ErbB3 receptor contains an inactive tyrosine kinase domain, 2C4 is very effective in blocking heregulin-mediated ErbB3-ErbB2 signaling. We demonstrate that the in vitro and in vivo growth of several breast and prostate tumor models is inhibited by 2C4 treatment.
- Published
- 2002
- Full Text
- View/download PDF
10. A potential role for activated HER-2 in prostate cancer.
- Author
-
Agus DB, Akita RW, Fox WD, Lofgren JA, Higgins B, Maiese K, Scher HI, and Sliwkowski MX
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Humans, Ligands, Male, Prostate-Specific Antigen metabolism, Trastuzumab, Neoplasms, Hormone-Dependent drug therapy, Neoplasms, Hormone-Dependent metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 physiology
- Abstract
The epidermal growth factor (also known as HER or ErbB) family of receptor tyrosine kinases are important mediators of cell growth, differentiation, and survival. At present there are 10 ligands that bind directly to epidermal growth factor, HER-3, or HER-4. Although none of these ligands bind directly to HER-2, it is recruited to these receptor complexes and also becomes activated. A monoclonal antibody directed against HER-2, 2C4, inhibits the association of HER-2 with other HER family members. Ligand-activated HER-2 may also play a role in cancers, particularly those that do not overexpress HER-2 at high levels. For example, when prostate cancers progress from an androgen-dependent to an androgen-independent phenotype, epidermal growth factor pathways are frequently activated. 2C4 will inhibit the growth of both androgen-dependent and androgen-independent prostate tumors grown as xenografts in athymic mice.
- Published
- 2000
11. Nonclinical studies addressing the mechanism of action of trastuzumab (Herceptin).
- Author
-
Sliwkowski MX, Lofgren JA, Lewis GD, Hotaling TE, Fendly BM, and Fox JA
- Subjects
- 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, Antibodies, Monoclonal pharmacology, Antineoplastic Agents pharmacology, Receptor, ErbB-2 immunology
- 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.
- Published
- 1999
12. Selection of heregulin variants having higher affinity for the ErbB3 receptor by monovalent phage display.
- Author
-
Ballinger MD, Jones JT, Lofgren JA, Fairbrother WJ, Akita RW, Sliwkowski MX, and Wells JA
- Subjects
- Amino Acid Sequence, Bacteriophage M13, Binding Sites, Ligands, Models, Molecular, Molecular Sequence Data, Nuclear Magnetic Resonance, Biomolecular, Peptide Library, Protein Binding, Receptor, ErbB-3, Recombinant Proteins, Structure-Activity Relationship, Carrier Proteins chemistry, ErbB Receptors metabolism, Glycoproteins chemistry, Neuregulin-1, Proto-Oncogene Proteins metabolism
- Abstract
Heregulins (HRGs) are epidermal growth factor (egf) domain containing polypeptide growth factors that bind and activate several members of the ErbB receptor family. Although HRG can bind to ErbB3 and ErbB4 homodimers, the highest affinity and most intracellularly active receptor complexes are hetero-oligomers containing ErbB2. The HRGbeta egf domain was displayed on the surface of M13 phage to facilitate mutagenic analysis and optimize for binding to a homodimeric ErbB3-immunoglobulin (IgG) fusion. Nine libraries were constructed in which virtually the entire sequence was randomized in stretches of four to six amino acids. These were selected separately for binding to immobilized ErbB3-IgG. Analysis of the resulting sequences revealed some areas that diverged radically from the wild-type, whereas others showed strong conservation. The degree of wild-type conservation correlated strongly with the functional importance of the residues as determined by alanine scanning mutagenesis (Jones, J. T., Ballinger, M. D., Pisacane, P. I., Lofgren, J. A., Fitzpatrick, V. D., Fairbrother, W. J., Wells, J. A., and Sliwkowski, M. X. (1998) J. Biol. Chem. 273, 11667-11674). Some variants from several libraries showed significant improvements in binding affinity to the ErbB3-IgG. These optimized segments were combined in various ways in the same molecule to generate variants (containing up to 16 mutations) that had >50-fold higher affinity than wild-type HRGbeta. The optimized variants stimulated ErbB2 phophorylation on MCF7 cells at levels similar to wild-type. This indicates wild-type affinity is optimized for potency and that factors other than affinity for ErbB3 are limiting. These variants showed enhanced affinity toward the ErbB4 homodimer, suggesting these receptors use very similar binding determinants despite them having 65% sequence identity.
- Published
- 1998
- Full Text
- View/download PDF
13. Binding interaction of the heregulinbeta egf domain with ErbB3 and ErbB4 receptors assessed by alanine scanning mutagenesis.
- Author
-
Jones JT, Ballinger MD, Pisacane PI, Lofgren JA, Fitzpatrick VD, Fairbrother WJ, Wells JA, and Sliwkowski MX
- Subjects
- Alanine, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Carrier Proteins chemistry, Carrier Proteins ultrastructure, Enzyme Activation, Glycoproteins chemistry, Glycoproteins ultrastructure, Humans, Ligands, Models, Molecular, Mutagenesis, Site-Directed, Nuclear Magnetic Resonance, Biomolecular, Phosphorylation, Protein Binding, Protein Structure, Secondary, Receptor, ErbB-3, Receptor, ErbB-4, Signal Transduction, Structure-Activity Relationship, Tumor Cells, Cultured, Carrier Proteins metabolism, ErbB Receptors metabolism, Glycoproteins metabolism, Neuregulin-1, Proto-Oncogene Proteins metabolism
- Abstract
Individual residues of the heregulinbeta (HRG) egf domain were mutated to alanine and displayed monovalently on phagemid particles as gene III fusion proteins. Wild type HRGbeta egf domain displayed on phage was properly folded as evidenced by its ability to bind ErbB3 and ErbB4 receptor-IgG fusion proteins with affinities close to those measured for bacterially produced HRGbeta egf domain. Binding to ErbB3 and ErbB4 receptors was affected by mutation of residues throughout the egf domain; including the NH2 terminus (His2 and Leu3), the two beta-turns (Val15-Gly18 and Gly42-Gln46), and some discontinuous residues (including Leu3, Val4, Phe13, Val23, and Leu33) that form a patch on the major beta-sheet and the COOH-terminal region (Tyr48 and Met50-Phe53). Binding affinity was least changed by mutations throughout the Omega-loop and the second strand of the major beta-sheet. More mutants had greater affinity loss for ErbB3 compared with ErbB4 implying that it has more stringent binding requirements. Many residues important for HRG binding to its receptors correspond to critical residues for epidermal growth factor (EGF) and transforming growth factor alpha binding to the EGF receptor. Specificity may be determined in part by bulky groups that prevent binding to the unwanted receptor. All of the mutants tested were able to induce phosphorylation and mitogen-activated protein kinase activation through ErbB4 receptors and were able to modulate a transphosphorylation signal from ErbB3 to ErbB2 in MCF7 cells. An understanding of binding similarities and differences among the EGF family of ligands may facilitate the development of egf-like analogs with broad or narrow specificity.
- Published
- 1998
- Full Text
- View/download PDF
14. Growth regulation of human breast and ovarian tumor cells by heregulin: Evidence for the requirement of ErbB2 as a critical component in mediating heregulin responsiveness.
- Author
-
Lewis GD, Lofgren JA, McMurtrey AE, Nuijens A, Fendly BM, Bauer KD, and Sliwkowski MX
- Subjects
- 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, Breast Neoplasms metabolism, Carrier Proteins metabolism, Glycoproteins metabolism, Neuregulin-1, Ovarian Neoplasms metabolism, Receptor, ErbB-2 metabolism
- 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.
- Published
- 1996
15. Analysis of heregulin-induced ErbB2 phosphorylation with a high-throughput Kinase receptor activation enzyme-linked immunosorbant assay.
- Author
-
Sadick MD, Sliwkowski MX, Nuijens A, Bald L, Chiang N, Lofgren JA, and Wong WL
- Subjects
- Animals, Blotting, Western, Humans, Phosphorylation, Rabbits, Receptor Protein-Tyrosine Kinases metabolism, Sensitivity and Specificity, Tumor Cells, Cultured, Carrier Proteins pharmacology, Enzyme-Linked Immunosorbent Assay methods, Glycoproteins pharmacology, Neuregulin-1, Receptor, ErbB-2 metabolism
- Abstract
A rapid, sensitive, and high-throughput assay has been developed to quantify ligand-induced receptor tyrosine kinase activation in terms of receptor phosphorylation. The assay, termed a kinase receptor activation enzyme-linked immunosorbant assay (KIRA-ELISA), consists of two separate microtiter plates, one for cell culture, ligand stimulation, and cell lysis/receptor solubilization and the other plate for receptor capture and phosphotyrosine ELISA. The assay was developed for analysis of heregulin-induced ErbB2 activation and utilizes the stimulation of intact receptor on the adherent breast carcinoma cell line, MCF-7. Membrane proteins are solubilized via Triton X-100 lysis and the receptor is captured in ELISA wells coated with ErbB2-specific antibodies with no cross-reaction to ErbB3 or ErbB4. The degree of receptor phosphorylation is then quantified by antiphosphotyrosine ELISA. A reproducible standard curve is generated with a EC(50) of approximately 360 pM for heregulin beta 1(177-244) (HRG beta 1(177-244). When identical samples of HRG beta 1(177-244) are analyzed by both the KIRA-ELISA and quantitative antiphosphotyrosine Western blot analysis, the results correlate very closely with one another. The assay described in this report is able to specifically quantify tyrosine phosphorylation of ErbB2 that results from the interaction of HRG with ErbB3 and/or ErbB4.
- Published
- 1996
- Full Text
- View/download PDF
16. Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin.
- Author
-
Sliwkowski MX, Schaefer G, Akita RW, Lofgren JA, Fitzpatrick VD, Nuijens A, Fendly BM, Cerione RA, Vandlen RL, and Carraway KL 3rd
- Subjects
- 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, Carrier Proteins metabolism, ErbB Receptors metabolism, Gene Expression, Glycoproteins metabolism, Neuregulin-1, Proto-Oncogene Proteins metabolism, Proto-Oncogenes
- 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.
- Published
- 1994
17. Monoclonal antibody based ELISAs for measurement of activins in biological fluids.
- Author
-
Wong WL, Garg SJ, Woodruff T, Bald L, Fendly B, and Lofgren JA
- Subjects
- Activins, Animals, Antibodies, Monoclonal, Antibody Specificity, Female, Humans, Mice, Mice, Inbred BALB C, Recombinant Proteins analysis, Reproducibility of Results, Sensitivity and Specificity, Adjuvants, Immunologic analysis, Enzyme-Linked Immunosorbent Assay methods, Growth Substances analysis, Inhibins analysis, Oligopeptides, Peptides analysis
- Abstract
Two sensitive monoclonal antibody (MAb)-based enzyme-linked immunosorbent assays (ELISAs), one for activin A (homodimer of beta A subunits) and one for activin B (homodimer of beta B subunits) in plasma have been developed. The activin A ELISA had an effective range of 0.2-50 ng/ml while the activin B ELISA's range was 0.1-25 ng/ml in human serum. Both ELISAs were specific with < 0.01% cross-reactivity with related hormones and follistatin (an activin binding protein), however the presence of recombinant human follistatin caused a decrease in measured level of activin A and B spiked human samples. The assay was linear across the standard curve range with intra- and interassay coefficients of variation were less than 15%. The level of activins in female serum range from 0.3 to 10.4 ng/ml. In summary, we have developed a reliable, convenient and rapid MAb-based enzyme immunoassay for determination of activin A and B levels in human serum which are also applicable for buffer, mouse and monkey serum matrices. This assay will be useful for studying the regulation and role of activin A and B in health and disease.
- Published
- 1993
- Full Text
- View/download PDF
18. Relaxin binding in the rat heart atrium.
- Author
-
Osheroff PL, Cronin MJ, and Lofgren JA
- Subjects
- Amino Acid Sequence, Animals, Autoradiography, Binding, Competitive, Estradiol pharmacology, Female, Heart Atria, Kinetics, Male, Molecular Sequence Data, Organ Specificity, Ovariectomy, Phosphorus Radioisotopes, Rats, Rats, Inbred Strains, Receptors, G-Protein-Coupled, Receptors, Neurotransmitter drug effects, Recombinant Proteins metabolism, Reference Values, Uterus metabolism, Myocardium metabolism, Receptors, Neurotransmitter metabolism, Receptors, Peptide, Relaxin metabolism
- Abstract
Relaxin is a member of the insulin family of polypeptides that is best known as a reproductive hormone. In an effort to elucidate the mechanism of action of relaxin we previously localized the specific binding sites of a 32P-labeled relaxin in the rat uterus and brain. These studies suggested that, in addition to its classical role in pregnancy, relaxin might have other physiological functions. In the present paper we describe the specific and high-affinity binding of relaxin to the cardiac atrium of both male and female rats. The relaxin binding could not be displaced by peptides belonging to the same family [insulin, insulin-like growth factor I (IGF-I)] or by peptides that were identified in the atrium or were known to have cardiovascular functions (atrial natriuretic peptide, angiotensin II). The dissociation constant for relaxin in the atrium was estimated to be 1.4 nM, which was similar to that found in the uterus (1.3 nM) and the brain (1.4 nM). In view of the close association of relaxin with reproduction, an experiment was also performed to compare the relaxin binding in the uterus and heart after gonadectomy and sex steroid treatment. It was found that the relaxin binding in the rat uterus was diminished by 53% overall following ovariectomy but was restored to 90% of normal levels when treated with estrogen (but not with testosterone). In contrast, the relaxin binding in the rat heart was not affected by castration or sex steroid treatment. We conclude that specific and high-affinity relaxin receptors exist in the atrium of both the male and female rat heart and that these are regulated differently than the relaxin receptors in the uterus.
- Published
- 1992
- Full Text
- View/download PDF
19. In vivo regulation of FSH synthesis by inhibin and activin.
- Author
-
Carroll RS, Kowash PM, Lofgren JA, Schwall RH, and Chin WW
- Subjects
- Activins, Aging physiology, Animals, Estradiol pharmacology, Female, Follicle Stimulating Hormone genetics, Follicle Stimulating Hormone metabolism, Follicle Stimulating Hormone, beta Subunit, Male, Ovariectomy, Pituitary Gland drug effects, Pituitary Gland metabolism, RNA, Messenger metabolism, Rats, Rats, Inbred Strains, Recombinant Proteins pharmacology, Follicle Stimulating Hormone biosynthesis, Inhibins pharmacology
- Abstract
The effect of a single sc injection of the gonadal peptide, recombinant human activin A (rhActivin A), on gonadotropin synthesis and secretion was examined in adult and immature male and female rats and the effect of recombinant human inhibin A (rhInhibin A) was examined in adult male rats. Pituitary FSH beta, LH beta and alpha messenger RNA (mRNA) levels were determined by blot hybridization. Trunk blood was collected to measure serum FSH levels. Treatment with rhInhibin A (100 micrograms/kg) resulted in a decrease in FSH beta mRNA to 2% of controls levels 6 h after injection. FSH beta mRNA levels started to rebound at 10 h, but were still significantly lower than vehicle-treated controls. Serum FSH levels were significantly reduced at 2 h and were reduced further at 6 and 10 h. There were no significant changes in alpha and LH beta mRNA levels. RhActivin A, at the highest dose (500 micrograms/kg), in immature male rats had only a modest effect (1.2- and 1.3-fold increase) on FSH beta mRNA levels and FSH secretion, respectively, at 2 h. No increase in FSH synthesis and FSH secretion was observed in adult male rats. In contrast, both immature and adult-ovariectomized E2 implanted females showed a robust response to rhActivin A. In immature females, 2 h after rhActivin A (100 and 500 micrograms/kg) administration, FSH beta mRNA levels were elevated 2.0- and 2.2-fold. At this time serum FSH was also elevated. At 6 and 10 h rhActivin A significantly reduced FSH beta mRNA levels from vehicle-treated controls. In contrast, FSH secretion was elevated at 6 h and returned to baseline at 10 h. Administration of rhActivin A (500 micrograms/kg) to adult, ovariectomized-E2 females resulted in a significant increase in FSH beta mRNA levels and FSH secretion at 2 and 6 h. There were no significant changes in alpha and LH beta mRNA levels in either males or females. Thus, these in vivo studies have shown that rhInhibin A can inhibit FSH beta mRNA levels and FSH secretion in the adult male rat. RhActivin A stimulates FSH synthesis and secretion in the immature and adult ovariectomized-E2 females, but has little or no effect in immature and adult males. Hence, there is a sexual dimorphic response to rhActivin A in vivo in the rat.
- Published
- 1991
- Full Text
- View/download PDF
20. Generation of polyclonal antibodies against recombinant human activin A.
- Author
-
Lofgren JA, Schwall R, Schmelzer C, and Wong WL
- Subjects
- Activins, Animals, Antibody Formation, Antibody Specificity, Blotting, Western, Female, Goats immunology, Humans, Immunization, Male, Rabbits immunology, Radioimmunoassay, Recombinant Proteins immunology, Antibodies immunology, Inhibins immunology
- Abstract
A goat antiserum to purified recombinant human activin A (rhAct-A), a dimer formed by two beta A-subunits of inhibin, has been produced. The immunoreactivity of the antiserum has been evaluated in an antigen coated enzyme-linked immunosorbent assay, in a radioimmunoassay using iodinated rhAct-A, and by Western blot analysis. The antiserum demonstrated some cross reactivity to inhibin A, a structurally related heterodimer which contains an identical beta A-subunit coupled to a distinct, though similar, alpha subunit. A simple radioimmunoassay for rhAct-A in tissue culture supernatant has been developed with rhAct-A affinity column purified polyclonal antiserum. The assay is precise and sensitive with a range of 0.31-40 ng/ml. The cross reactivity of inhibin A in the RIA is about 4.3%. Despite its cross-reactivity this antiserum will facilitate studies of the physiology of activin A and inhibin A which includes a Western blot analysis where a molecular size distinction is accomplished.
- Published
- 1991
- Full Text
- View/download PDF
21. Preparation of biologically active 32P-labeled human relaxin. Displaceable binding to rat uterus, cervix, and brain.
- Author
-
Osheroff PL, Ling VT, Vandlen RL, Cronin MJ, and Lofgren JA
- Subjects
- Adenosine Triphosphate metabolism, Amino Acid Sequence, Animals, Biological Assay, Cyclic AMP metabolism, Cyclic AMP pharmacology, Electrophoresis, Polyacrylamide Gel, Female, Humans, Immunosorbent Techniques, Isotope Labeling, Molecular Sequence Data, Phosphorylation, Protein Kinases metabolism, Rats, Rats, Inbred Strains, Relaxin pharmacology, Uterus drug effects, Brain metabolism, Cervix Uteri metabolism, Phosphorus Radioisotopes, Relaxin metabolism, Uterus metabolism
- Abstract
Relaxin is a member of the insulin family of polypeptide hormones and is known to exert its biological effects on various parts of the mammalian reproductive system. Biologically active human relaxin has been chemically synthesized based on the nucleotide sequence obtained from an ovarian cDNA clone. In the present study synthetic human relaxin was radiolabled by phosphorylation with cAMP-dependent protein kinase and [gamma-32P]ATP to a specific activity of 5000 Ci/mmol. The phosphorylated relaxin was purified on cation exchange high performance liquid chromatography and was shown to co-migrate with relaxin on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Mass spectrometry revealed a single phosphorylated site on the B chain of relaxin. The 32P-relaxin was able to bind to a goat anti-relaxin antibody, and this binding could be displaced by unlabeled relaxin in a concentration-dependent manner. A comparison of the concentration responses of cellular cAMP production stimulated by relaxin and phosphorylated relaxin in a primary human uterine cell line showed that phosphorylation did not affect the in vitro biological efficacy of relaxin. This made it suitable for in situ autoradiographic localization of relaxin binding sites in rat uterine, cervical, and brain tissue sections. Displacement of the binding of 100 pM 32P-relaxin by 100, 10, and 3 nM unlabeled relaxin, but not by 100 nM insulin, insulin-like growth factor-I, and an insulin-like growth factor-I analog, demonstrated the high affinity and specificity of such binding. We conclude that 32P-labeled human relaxin is biologically and immunologically active and that this novel probe binds reversibly and with high affinity to classical (e.g. uterus) and unpredicted (e.g. brain) tissues.
- Published
- 1990
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.