Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Oncogene ; 31(30): 3505-15, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22105361

ABSTRACT

Epidermal growth factor (EGF)-like growth factors control tumor progression as well as evasion from the toxic effects of chemotherapy. Accordingly, antibodies targeting the cognate receptors, such as EGFR/ErbB-1 and the co-receptor HER2/ErbB-2, are widely used to treat cancer patients, but agents that target the EGF-like growth factors are not available. To circumvent the existence of 11 distinct ErbB ligands, we constructed a soluble fusion protein (hereinafter: TRAP-Fc) comprising truncated extracellular domains of EGFR/ErbB-1 and ErbB-4. The recombinant TRAP-Fc retained high-affinity ligand binding to EGF-like growth factors and partially inhibited growth of a variety of cultured tumor cells. Consistently, TRAP-Fc displayed an inhibitory effect in xenograft models of human cancer, as well as synergy with chemotherapy. Additionally, TRAP-Fc inhibited invasive growth of mammary tumor cells and reduced their metastatic seeding in the lungs of animals. Taken together, the activities displayed by TRAP-Fc reinforce critical roles of EGF-like growth factors in tumor progression, and they warrant further tests of TRAP-Fc in preclinical models.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , ErbB Receptors/chemistry , Lung Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/secondary , Mice , Mice, Nude , Mice, SCID , Receptor, ErbB-2/chemistry , Receptor, ErbB-4 , Recombinant Fusion Proteins/chemistry , Xenograft Model Antitumor Assays
2.
Br J Cancer ; 91(6): 1190-4, 2004 Sep 13.
Article in English | MEDLINE | ID: mdl-15305194

ABSTRACT

ErbB2 and EGFR are attractive oncology therapeutic targets as their overexpression in tumors predicts a poorer clinical outcome in a variety of epithelial malignancies. However, clinical results with therapeutic compounds targeting these receptors have been mixed. Therefore, there is a need for improved predictive biomarkers for these targeted therapeutics. In this study we analysed tissue microarrays of patients treated with combination chemotherapy and Herceptin for expression or phosphorylation of signalling proteins associated with erbB receptors to identify protein biomarkers that are predictive of breast cancer patient response. A comparison of expression or phosphorylation of these markers with patient outcome revealed that response to Herceptin depended not only on expression levels of erbB2 but also on expression of EGFR, expression of erbB ligands, expression of other receptors and phosphorylation of downstream proteins. Elucidating the biological effects of EGFR/erbB2 targeted therapeutics will enable patient tumor profiling to identify likely responders and the determination of biologically effective doses that allows chronic administration of these agents in order to maximise efficacy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Disease Progression , ErbB Receptors/genetics , Female , Humans , Retrospective Studies , Signal Transduction , Trastuzumab
3.
Pathol Oncol Res ; 7(3): 171-7, 2001.
Article in English | MEDLINE | ID: mdl-11692142

ABSTRACT

The purpose of this study was to investigate the frequency of expression of the erbB/HER family of growth factor receptors, their ligand heregulin, and the two different signaling pathways p38 and mitogen-activated protein kinase (MAPK), as well as the status of HER-2 phosphorylation in tumor specimens from patients with primary breast cancer. The level of expression of these proteins was measured by quantitative immunohistochemistry combined with microscope-based image analysis in paraffin-embedded breast cancer tissue from 35 patients. The frequency of expression was: EGFR (51%), HER-2 (54%), P-HER-2 (48%), HER-3 (48%), HER-4 (57%), heregulin (48%), p38 (17%), MAPK (48%). There was evidence of associations among the coexpression of heregulin, EGFR, HER-2, and HER-3. Also, there was evidence of a positive association between P-MAPK and HER-4. HER-3 was expressed at high levels in patients younger than 50 years of age. There was a trend for expression of higher levels of HER-4 in tumors larger than 2 cm. The expression of EGFR, HER-2, heregulin, p38 and MAPK was independent of age, tumor size, number of lymph nodes involved or hormone receptor status. The HER family of growth factor receptors appear to be regulated independently in invasive breast cancer. Assessing the expression of multiple tumor markers by quantitative immuno-histochemistry is feasible. Further research is needed to determine the prognostic and predictive roles of the various associations between HER receptors, their ligands and signal transduction molecules in patients with early-stage breast cancer.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-2/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Neoplasm Staging , Signal Transduction , p38 Mitogen-Activated Protein Kinases
4.
Am J Ophthalmol ; 131(6): 788-91, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11384577

ABSTRACT

PURPOSE: To evaluate choroidal melanomas in enucleated eyes for the presence of type I estrogen receptors. METHODS: Fourteen consecutive eyes with large choroidal melanomas (defined as >16-mm basal diameter and >8 mm thickness) from 14 patients (eight women and six men with a mean age of 57 years; range, 25--74 years) enucleated in accordance with the Collaborative Ocular Melanoma Study (COMS) protocol were investigated. Immunohistochemical techniques were employed to label the choroidal melanomas for the presence of type I estrogen receptors. Each specimen was then evaluated in a masked fashion by an experienced ophthalmic pathologist for positive nuclear staining. RESULTS: No tumors showed immunohistochemical evidence of a type I estrogen receptor. CONCLUSION: Type I estrogen receptors are not present in choroidal melanoma. Estrogens are not likely to influence choroidal melanoma growth through traditional receptors.


Subject(s)
Choroid Neoplasms/metabolism , Melanoma/metabolism , Receptors, Estrogen/metabolism , Adult , Aged , Breast Neoplasms/metabolism , Carcinoma/metabolism , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Single-Blind Method , Staining and Labeling
5.
Oncogene ; 20(2): 147-55, 2001 Jan 11.
Article in English | MEDLINE | ID: mdl-11313944

ABSTRACT

The anti-cancer agent paclitaxel (Taxol) stabilizes microtubules leading to G2/M cell cycle arrest and apoptotic cell death. In order to analyse the molecular mechanisms of Taxol-induced cytotoxicity, we studied the involvement of mitogen-activated protein kinases (MAPK) ERK and p38 as well as the p53 pathways in Taxol-induced apoptosis. The human breast carcinoma cell line MCF7 and its derivatives, MCF7/HER-2 and MDD2, were used in the study. We found that Taxol treatment strongly activated ERK, p38 MAP kinase and p53 in MAP kinase MCF7 cells prior to apoptosis. PD98059 or SB203580, specific inhibitors of ERK and p38 kinase activities, significantly decreased apoptosis, leaving the surviving cells arrested in G2/M. These inhibitors did not significantly affect Taxol-induced alterations in the cell cycle regulatory proteins Rb, p53, p21/Waf1 and Cdk-2. In addition, inactivation of p53 did not affect cellular sensitivity to Taxol killing. However, cells with inactivated p53, unlike cells harboring wild type p53, failed to arrest in G2/M after treatment with Taxol and continued to divide or go into apoptosis. Our data show that both ERK and p38 MAP kinase cascades are essential for apoptotic response to Taxol-induced cellular killing and are independent of p53 activity. However, p53 may serve as a survival factor in breast carcinoma cells treated with Taxol by blocking cells in G2/M phase of the cell cycle.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , CDC2-CDC28 Kinases , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System/drug effects , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , G2 Phase/drug effects , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Mitosis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Rats , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , p38 Mitogen-Activated Protein Kinases
6.
Breast Dis ; 11: 63-75, 2000.
Article in English | MEDLINE | ID: mdl-15687593

ABSTRACT

Amplification and overexpression of the Her2/neu (c-erbB-2) growth factor receptor occurs in approximately 25% of early stage breast cancers. HER2/neu has been established as an important prognostic factor in early stage breast cancer in large patient populations and has been associated with shorter overall and disease free survival. New data are emerging to suggest that HER2/neu may be useful not only as a prognostic factor but also as a predictive marker for response to chemotherapeutics, anti-estrogens, and therapeutic regimens using anti-HER2/neu monoclonal antibodies. However, little is known of how other erbB receptors tyrosine kinases affect breast cancer biological behavior and response to therapy. In this review, we highlight recent data on Her2/neu as a prognostic and predictive marker of response to therapy, as well as how the other receptors of the erbB family affect the biological behavior of breast cancer.

7.
Mol Cell Biol ; 18(10): 6090-101, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742126

ABSTRACT

The recently isolated second family of neuregulins, NRG2, shares its primary receptors, ErbB-3 and ErbB-4, and induction of mammary cell differentiation with NRG1 isoforms, suggesting functional redundancy of the two growth factor families. To address this possibility, we analyzed receptor specificity of NRGs by using an engineered cellular system. The activity of isoform-specific but partly overlapping patterns of specificities that collectively activate all eight ligand-stimulatable ErbB dimers was revealed. Specifically, NRG2-alpha [corrected], like NRG1-beta [corrected], emerges as a narrow-specificity ligand, whereas NRG2-beta [corrected] is a pan-ErbB ligand that binds with different affinities to all receptor combinations, including those containing ErbB-1, but excluding homodimers of ErbB-2. The latter protein, however, displayed cooperativity with the direct NRG receptors. Apparently, signaling by all NRGs is funneled through the mitogen-activated protein kinase (MAPK). However, the duration and potency of MAPK activation depend on the identity of the stimulatory ligand-receptor ternary complex. We conclude that the NRG-ErbB network represents a complex and nonredundant machinery developed for fine-tuning of signal transduction.


Subject(s)
ErbB Receptors/metabolism , Glycoproteins/metabolism , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation , ErbB Receptors/biosynthesis , Glycoproteins/pharmacology , Isomerism , Ligands , Nerve Growth Factors/pharmacology , Neuregulins , Phosphorylation , Proto-Oncogene Proteins/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3 , Receptor, ErbB-4
8.
J Biol Chem ; 273(17): 10496-505, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9553109

ABSTRACT

The ErbB signaling network consists of four transmembrane receptor tyrosine kinases and more than a dozen ligands sharing an epidermal growth factor (EGF) motif. The multiplicity of ErbB-specific ligands is incompletely understood in terms of signal specificity because all ErbB molecules signal through partially overlapping pathways. Here we addressed the action of epiregulin, a recently isolated ligand of ErbB-1. By employing a set of factor-dependent cell lines engineered to express individual ErbBs or their combinations, we found that epiregulin is the broadest specificity EGF-like ligand so far characterized: not only does it stimulate homodimers of both ErbB-1 and ErbB-4, it also activates all possible heterodimeric ErbB complexes. Consistent with its relaxed selectivity, epiregulin binds the various receptor combinations with an affinity that is approximately 100-fold lower than the affinity of ligands with more stringent selectivity, including EGF. Nevertheless, epiregulin's action upon most receptor combinations transmits a more potent mitogenic signal than does EGF. This remarkable discrepancy between binding affinity and bioactivity is permitted by a mechanism that prevents receptor down-regulation, and results in a weak, but prolonged, state of receptor activation.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Animals , CHO Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Cell Line , Cricetinae , Dimerization , Down-Regulation , Enzyme Activation , Epiregulin , Ligands , Phosphorylation , Signal Transduction , Tyrosine/metabolism
9.
Oncogene ; 16(9): 1209-15, 1998 Mar 05.
Article in English | MEDLINE | ID: mdl-9528863

ABSTRACT

Betacellulin (BTC) is a member of the EGF ligand family that directly binds to both EGFR and HER4 and induces the growth of certain epithelial cell types. Fusion proteins composed of the terminal 48 or 50 amino acids of mature betacellulin and a binding defective form of Pseudomonas exotoxin (BTC-TX48 and BTC-TX50, respectively), have been produced. BTC-TX50 induced tyrosine phosphorylation of both EGFR and HER4, whereas BTC-TX48 induced phosphorylation of HER4 but to a much lesser extent EGFR, indicating that the presence of two additional amino acid residues, Arg62 and Lys63, contribute to full kinase activity. BTC-TX50 was up to 300-fold more active at inhibiting protein synthesis than BTC-TX48 on cell lines expressing EGFR, most likely due to the >tenfold higher affinity of BTC-TX50. MDA-MB-453 breast carcinoma cells which express HER4 but not EGFR, were not sensitive to either BTC-TX form. These data indicate that despite the ability of BTC-TX to bind and phosphorylate HER4, it was only cytotoxic to cells expressing EGFR. The inability of BTC-TX to kill cells was likely due to its failure to internalize through HER4.


Subject(s)
Bacterial Toxins/pharmacokinetics , Bacterial Toxins/toxicity , ErbB Receptors/physiology , Growth Substances/pharmacokinetics , Growth Substances/toxicity , Intercellular Signaling Peptides and Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Bacterial Toxins/chemistry , Betacellulin , Breast Neoplasms , Carcinoma, Squamous Cell , Cell Survival/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/drug effects , Female , Growth Substances/chemistry , Humans , KB Cells , Lymphoma, B-Cell , Mice , Molecular Sequence Data , Ovarian Neoplasms , Pseudomonas , Receptor, ErbB-4 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Tumor Cells, Cultured
10.
Oncogene ; 16(10): 1249-58, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9546426

ABSTRACT

The ErbB-1 receptor tyrosine kinase binds to six different growth factors, whose prototype is the epidermal growth factor (EGF). Two homologous epithelial receptors, ErbB-3 and ErbB-4, bind all isoforms of another family of growth factors, the Neu differentiation factors (NDFs/neuregulins). The fourth member of the ErbB family, ErbB-2, acts as the preferred heterodimeric partner of ligand-occupied complexes of the three other ErbB proteins. Here we report that at high concentrations, EGF can induce cell growth and differentiation in the absence of ErbB-1. This function is shared by betacellulin, but not by three other ligands, including the transforming growth factor alpha (TGFalpha). The functional receptor was identified as a heterodimer between ErbB-3 and ErbB-2, a previously identified oncogenic complex. When singly expressed, neither ErbB-3 nor ErbB-2 can mediate signaling by EGF. In addition, when co-expressed, blocking either receptor by using site-specific antibodies inhibited EGF and betacellulin activities, indicating strict cooperativity between ErbB-3 and ErbB-2. Through analysis of chimeras between EGF and TGFalpha, we identified the middle portion of EGF (loop B) as the site that enables activation of ErbB-2/ErbB-3. In conclusion, cooperative and promiscuous binding of stroma-derived growth factors by the epithelium-expressed ErbB-2/ErbB-3 heterodimer may be significant to cancer development. The mechanistic implications of our results for a model that attributes receptor dimerization to ligand bivalency, as well as to a recently proposed mechanism of secondary dimerization, are discussed.


Subject(s)
ErbB Receptors/metabolism , Growth Substances/pharmacology , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Betacellulin , Binding Sites , Breast Neoplasms , Cell Differentiation/drug effects , Cell Division/drug effects , Dimerization , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/chemistry , Female , Growth Substances/metabolism , Hematopoietic Stem Cells , Humans , Ligands , Mice , Phosphorylation , Proto-Oncogene Proteins/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-3 , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism
11.
Biochem Soc Symp ; 63: 199-210, 1998.
Article in English | MEDLINE | ID: mdl-9513724

ABSTRACT

Members of the epidermal growth factor (EGF) family of tyrosine kinase receptors are involved in the regulation of cell growth and differentiation, and are found to be expressed in many types of cancers. Activation of these receptors can be elicited by multiple ligands, resulting in the formation of a spectrum of heterodimer complexes and a number of biological outcomes. A clear demonstration of biological activation by a single complex has been difficult to address because of the endogenous expression of HERs (human EGF-like receptors) in many cell lines. We have generated a collection of cell lines expressing all HERs alone or in all pairwise combinations in a clone of NIH 3T3 cells (3T3-7d) devoid of detectable EGF receptor family members. Transformation, as measured by growth in soft agar, only occurred in cells expressing two different HER family members. Transformation with activated Neu and the rate of in vivo tumour formation were also correlated with the expression of multiple HERs in the same cell. To further our understanding of the role of heterodimer signalling, we demonstrated that, within a breast carcinoma cell line, activation of HER-3 results in cellular differentiation, prolonged activation of extracellular-signal-related kinase 1 (ERK1) activity and an increase in p21CIP1/WAF1 nuclear staining. In contrast, activation of HER-4 is mitogenic, induces transient activation of ERK1 activity and decreases the nuclear staining of p21CIP1/WAF1. These differences in biochemical and biological responses are correlated with the contrasting abilities of HER-3 and HER-4 to be down-regulated from the cell surface. The cell-surface localization of HER-3 does not change in response to ligand, whereas activation of HER-4 results in a loss of cell-surface staining followed by accumulation into a perinuclear compartment.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , Cell Division , ErbB Receptors/physiology , 3T3 Cells , Animals , ErbB Receptors/genetics , Humans , Mice , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3 , Receptor, ErbB-4 , Transfection , Tumor Cells, Cultured
12.
Cancer Res ; 57(23): 5217-20, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9393737

ABSTRACT

DNA damage leads to the stabilization of p53 protein and its translocation to the nucleus, resulting in activation or suppression of p53-responsive genes. However, a significant proportion of cell nuclei remain negative for p53 and p53-inducible cyclin-dependent kinase inhibitor p21waf1 after a single dose of gamma-irradiation. Quantitation of DNA content in p53-positive and -negative nuclei 4-6 h after 10 Gy of gamma-irradiation of human breast carcinoma MCF7 cells, fibrosarcoma HT1080 cells, and diploid skin fibroblasts showed that p53 and p21waf1 nuclear accumulation occurs predominantly in the G1 phase and at the beginning of the S phase of the cell cycle. The majority of the nuclei in late S phase and in G2-M phase remained p53- and p21waf1-negative. This suggests that there is a cell cycle window during which p53 can accumulate in the nucleus and activate expression of p21waf1. To determine whether cell cycle-dependent distribution of p53 is caused by cytoplasmic modifications of p53 protein or by properties of the nucleus, p53 localization was analyzed in multinucleated cells obtained by polyethylene glycol-mediated cell fusion. Dramatic differences in p53 accumulation were found among the nuclei in individual multinucleated cells. Distribution of p53-positive and -negative nuclei among the phases of the cell cycle was similar to that observed in a regular cell population. These results suggest that the observed differences in p53 accumulation in the nuclei of irradiated cells are determined by cell cycle-dependent nuclear functions. In contrast to p53, p21waf1 was equally distributed among the nuclei of multinucleated cells regardless of the stage of the cell cycle, indicating that the observed phenomenon is specific for p53.


Subject(s)
Cell Cycle/radiation effects , Cell Nucleus/radiation effects , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms , Cell Cycle/physiology , Cell Fusion , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , Cyclins/biosynthesis , Enzyme Inhibitors , Female , Fibroblasts , Fibrosarcoma , Gamma Rays , Humans , Polyethylene Glycols , Skin , Tumor Cells, Cultured , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/radiation effects
13.
J Clin Oncol ; 15(10): 3249-57, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9336362

ABSTRACT

PURPOSE: In addition to tumor size, grade, location, and the presence of metastases, other factors may be useful in prognostication for adults with soft tissue sarcoma (STS). This study examines the relationship of MDR-1 mRNA, p-glycoprotein (P-gp), Ki-67 expression, and DNA content expression to clinical outcome in adults with STS. PATIENTS AND METHODS: Snap-frozen STS specimens from 65 patients were analyzed and compared with clinical outcomes. Immunohistochemistry was performed for the Ki-67 antigen and P-gp. DNA content was determined using the Feulgen reaction and quantitated using image analysis. MDR-1 mRNA expression was determined using a reverse-transcriptase polymerase chain reaction (RT-PCR)-based assay. RESULTS: P-glycoprotein expression was found by immunohistochemistry in 48% of cases with 5-year overall (54% v 14%, P = .07) and disease-free survival rates (32% v 18%, P = .039) higher in high-grade tumors that did not express P-gp. MDR-1 mRNA was detected in 51% of cases and no patient with high levels of MDR-1 mRNA expression was a long-term survivor. Patients with diploid tumors had significantly better survival than those with nondiploid tumors (51% v 31%, P = .03). High levels of Ki-67 were associated with poorer overall survival (46% v 31%, P = .04). On multivariate analysis, American Joint Committee on Cancer (AJCC) staging, DNA content, Ki-67, and P-gp staining were significant prognostic factors for 5-year overall and disease-free survival. CONCLUSION: P-gp expression, high-level Ki-67 expression, and nondiploid DNA content are independent prognostic indicators that correlate with poor outcomes in STS patients. However, MDR-1 mRNA was not found to be predictive of survival. These newer markers are useful additions to AJCC staging for prognostication for patients with STS. Such markers may be useful in selecting high-risk STS patients who could benefit from systemic adjuvant therapy.


Subject(s)
Biomarkers, Tumor/analysis , Sarcoma/mortality , Soft Tissue Neoplasms/mortality , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Adolescent , Adult , Aged , Aged, 80 and over , Child , DNA, Neoplasm/analysis , Disease-Free Survival , Female , Genes, MDR , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Middle Aged , Prognosis , Sarcoma/chemistry , Sarcoma/pathology , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/pathology , Survival Rate
14.
Anal Quant Cytol Histol ; 19(4): 316-28, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9267565

ABSTRACT

OBJECTIVE: To summarize the current literature on the association between biologic prognostic factors and therapeutic implications. STUDY DESIGN: To illustrate how these biologic factors are determined and how they can affect treatment, three patients' biologic profiles and their implications for the patients' choice of therapeutic approaches were analyzed. Immunohistochemical techniques combined with image analysis was used to evaluate estrogen receptors, progesterone receptors, proliferation index and erbB-2. Visual assessment was used to evaluate P glycoprotein (MDR1), EGFR and p53. RESULTS: Data from the literature stress the importance of biologic profiles for defining tumor behavior and patient management. The examples of patients' biologic factors illustrated the possible importance of these factors for helping to design treatment. CONCLUSION: Today the data on the association of patient response to chemotherapy and molecular markers are only starting to accumulate. A larger database is needed for a more precise estimation of response probability in order to help physicians decide between treatment options.


Subject(s)
Breast Neoplasms/pathology , Adult , Aged , Breast Neoplasms/chemistry , Breast Neoplasms/therapy , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Middle Aged , Oncogene Proteins/analysis , Predictive Value of Tests , Proto-Oncogene Proteins c-bcl-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Treatment Outcome , Tumor Suppressor Protein p53/analysis
15.
EMBO J ; 16(6): 1391-400, 1997 Mar 17.
Article in English | MEDLINE | ID: mdl-9135154

ABSTRACT

To analyze the involvement of p53-dependent transcriptional activation in normal development and in response to DNA damage in vivo, we created transgenic mice with a lacZ reporter gene under the control of a p53-responsive promoter. Five independent strains showed similar patterns of transgene expression. In untreated animals, lacZ expression was limited to the developing nervous system of embryos and newborn mice and was strongly decreased in the adult brain. gamma-irradiation or adriamycin treatment induced lacZ expression in the majority of cells of early embryos and in the spleen, thymus and small intestine in adult mice. Transgene expression was p53 dependent and coincided with the sites of strong p53 accumulation. The lacZ-expressing tissues and early embryos, unlike other adult tissues and late embryos, are characterized by high levels of p53 mRNA expression and respond to DNA damage by massive apoptotic cell death. Analysis of p53-null mice showed that this apoptosis is p53 dependent. These data suggest that p53 activity, monitored by the reporter lacZ transgene, is the determinant of radiation and drug sensitivity in vivo and indicate the importance of tissue and stage specificity of p53 regulation at the level of mRNA expression.


Subject(s)
Lac Operon , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Base Sequence , DNA Damage , DNA Primers/genetics , Doxorubicin/pharmacology , Drug Resistance/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/radiation effects , Genes, Reporter , Genes, p53 , Male , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Tolerance/genetics , Tissue Distribution , Transcriptional Activation/drug effects , Transcriptional Activation/radiation effects , Transfection
16.
Oncogene ; 12(12): 2535-47, 1996 Jun 20.
Article in English | MEDLINE | ID: mdl-8700512

ABSTRACT

Previously we reported that neu differentiation factor (NDF)/heregulin (HRG) elevates tyrosine phosphorylation of its receptors erbB-3, erbB-4, and erbB-2 (through heterodimer formation). We also showed that both NDF/HRG and antibodies to erbB-2 can arrest growth and induce differentiation in breast cancer cells. In this study, we report on the mechanism of NDF/HRG-induced cellular effects. We show that NDF/HRG and antibodies to erbB-2 receptors up-regulate expression of p53 by stabilizing the protein. This is accompanied by up-regulation of the p53 inducible gene, p21CIP1/WAF1, in a variety of cell lines: MCF7 and their derivatives (MCF7/HER2, MN1 and MCF-7-puro), ZR75T and LnCap cells. The induction of p21 is further enhanced when cells are treated with both NDF/HRG and DNA-damaging chemotherapeutic agents (i.e. doxorubicin). The NDF/HRG mediated induction of p21 is dependent on wildtype p53, as it fails to occur in cells expressing dominant negative p53 (MDD2). Furthermore, p21 induction is capable of inactivating cdk2 complexes as measured by Histone H1 phosphorylation assays. Finally, we show that in primary cultures of breast and other cancers, p21 is significantly induced in response to NDF/HRG treatment. Collectively, these observations suggest that the mechanism of breast cancer cell growth inhibition and differentiation via erbB receptors activation is through a p53-mediated pathway.


Subject(s)
Breast Neoplasms/genetics , CDC2-CDC28 Kinases , Cyclins/genetics , Glycoproteins/pharmacology , Prostatic Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Cyclins/drug effects , Doxorubicin/pharmacology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Enzyme Inhibitors/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/drug effects , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic , Genes, p53 , Glycoproteins/genetics , Humans , Male , Neuregulins , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-3 , Receptor, ErbB-4 , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Up-Regulation
17.
J Biol Chem ; 271(13): 7620-9, 1996 Mar 29.
Article in English | MEDLINE | ID: mdl-8631797

ABSTRACT

The group of subtype I transmembrane tyrosine kinases includes the epidermal growth factor (EGF) receptor (ErbB-1), an orphan receptor (ErbB-2), and two receptors for the Neu differentiation factor (NDF/heregulin), namely: ErbB-3 and ErbB-4. Here we addressed the distinct functions of the two NDF receptors by using an immunological approach. Two sets of monoclonal antibodies (mAbs) to ErbB-3 and ErbB-4 were generated through immunization with recombinant ectodomains of the corresponding receptors that were fused to immunoglobulin. We found that the shared ligand binds to highly immunogenic, but immunologically distinct sites of ErbB-3 and ErbB-4. NDF receptors differed also in their kinase activities; whereas the catalytic activity of ErbB-4 was activable by mAbs, ErbB-3 underwent no activation by mAbs in living cells. Likewise, down-regulation of ErbB-4, but not ErbB-3, was induced by certain mAbs. By using the generated mAbs, we found that the major NDF receptor on mammary epithelial cells is a heterodimer of ErbB-3 with ErbB-2, whereas an ErbB-1/ErbB-2 heterodimer, or an ErbB-1 homodimer, is the predominant species that binds EGF. Consistent with ErbB-2 being a shared receptor subunit, its tyrosine phosphorylation was increased by both heterologous ligands and it mediated a trans-inhibitory effect of NDF on EGF binding. Last, we show that the effect of NDF on differentiation of breast tumor cells can be mimicked by anti-ErbB-4 antibodies, but not by mAbs to ErbB-3. Nevertheless, an ErbB-3-specific mAb partially inhibited the effect of NDF on cellular differentiation. These results suggest that homodimers of ErbB-4 are biologically active, but heterodimerization of the kinase-defective ErbB-3, probably with ErbB-2, is essential for transmission of NDF signals through ErbB-3.


Subject(s)
Antibodies, Monoclonal , ErbB Receptors/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Base Sequence , Breast Neoplasms , CHO Cells , Cell Differentiation/physiology , Clone Cells , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , Cyclins/biosynthesis , DNA Primers , Epidermal Growth Factor/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/immunology , Female , Glycoproteins/metabolism , Humans , Hybridomas , Immunohistochemistry , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/biosynthesis , Macromolecular Substances , Mammals , Mice , Mice, Inbred BALB C/immunology , Molecular Sequence Data , Neuregulins , Phosphorylation , Phosphotyrosine/analysis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/immunology , Receptor, ErbB-3 , Receptor, ErbB-4 , Restriction Mapping , Transfection , Tumor Cells, Cultured
18.
Am J Pathol ; 148(2): 549-58, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8579117

ABSTRACT

The neu/erbB-2/HER-2 proto-oncogene is amplified and/or overexpressed in up to 30% of mammary carcinomas and has been variably correlated with poor prognosis. The signaling activity of the encoded receptor tyrosine kinase is regulated by interactions with other type 1 receptors and their ligands. We have used a novel approach, phosphorylation-sensitive anti-Neu antibodies, to quantify signaling by Neu and epidermal growth factor receptor in a panel of frozen sections of mammary carcinoma specimens. We also determined the relationship of Neu, phosphorylated Neu (and epidermal growth factor receptor), and phosphotyrosine to the expression of Neu-related receptors (epidermal growth factor receptor, HER-3, and HER-4) and to prognostic factors (estrogen and progesterone receptor). We found that tyrosine phosphorylation of Neu (and hence signaling activity) is highly variable among mammary carcinomas. Neu and HER-4 were associated with divergent correlates, suggesting that they have profoundly different biological activities. These results have implications for etiology of mammary carcinoma for clinical evaluation of mammary carcinoma patients, and for development of Neu-targeted therapeutic strategies.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Steroid/metabolism , Biomarkers, Tumor , Breast Neoplasms/pathology , Female , Frozen Sections , Genes, erbB-2 , Humans , Immunohistochemistry , Phosphorylation , Phosphotyrosine/metabolism , Prognosis , Proto-Oncogene Mas , Receptor, ErbB-4 , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies
19.
Cancer Res ; 55(17): 3890-6, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7543819

ABSTRACT

The amplification and overexpression of the HER-2/neu proto-oncogene, which encodes the tyrosine kinase receptor p185neu, have been observed frequently in tumors from human breast cancer patients and are correlated with poor prognosis. To explore the potential of chemotherapy directed at the tyrosine kinase of p185neu, we have found that emodin (3-methyl-1,6,8-trihydroxyanthraquinone), a tyrosine kinase inhibitor, suppresses autophosphorylation and transphosphorylation activities of HER-2/neu tyrosine kinase, resulting in tyrosine hypophosphorylation of p185neu in HER-2/neu-overexpressing breast cancer cells. Emodin, at a 40-microM concentration, which repressed tyrosine kinase of p185neu, efficiently inhibited both anchorage-dependent and anchorage-independent growth of HER-2/neu-overexpressing breast cancer cells. However, the inhibition was much less effective for those cells expressing basal levels of p185neu under the same conditions. Emodin also induced differentiation of HER-2/neu-overexpressing breast cancer cells by exhibiting a morphological maturation property of large lacy nuclei surrounded by sizable flat cytoplasm and by showing a measurable production of large lipid droplets, which is a marker of mature breast cells. Therefore, our results indicate that emodin inhibits HER-2/neu tyrosine kinase activity and preferentially suppresses growth and induces differentiation of HER-2/neu-overexpressing cancer cells. These results may have chemotherapeutic implications for using emodin to target HER-2/neu-overexpressing cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Emodin/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Line, Transformed , Drug Screening Assays, Antitumor , Humans , Phosphorylation/drug effects , Phosphotyrosine , Proto-Oncogene Mas , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/metabolism
20.
J Biol Chem ; 270(13): 7625-30, 1995 Mar 31.
Article in English | MEDLINE | ID: mdl-7535774

ABSTRACT

We have constructed, expressed, and purified a fusion protein, HAR-TX beta 2, consisting of heregulin-beta 2 fused to a binding-defective form of Pseudomonas exotoxin A, PE40. The fusion protein was found to induce receptor tyrosine phosphorylation in CEM cells transfected with HER4 alone or in combination with HER2 but not in cells transfected with HER2 or HER1 alone. The phosphorylation of receptor tyrosines was both dose-dependent and saturable in amounts similar to those shown to be active for native heregulin. HAR-TX beta 2 was specifically cytotoxic toward a variety of carcinoma cell lines in the ng/ml range. However, some tumor cell lines were found to be insensitive to the cytotoxic action of the fusion protein even at > 2 micrograms/ml. Relative amounts of HER4, HER3, and HER2 were determined on seven cell lines sensitive and four cell lines insensitive to HAR-TX beta 2. All lines that express HER4 were killed by HAR-TX beta 2, while none lacking HER4 were affected. HAR-TX beta 2 was able to bind to and signal via tyrosine phosphorylation in cell lines that co-express HER2 and HER3 in the absence of HER4 without inducing cytotoxicity. Thus HAR-TX beta 2 may prove to be a useful reagent for the targeting and elimination of HER4-positive tumor cells.


Subject(s)
ADP Ribose Transferases , Cell Survival/drug effects , ErbB Receptors/biosynthesis , Exotoxins/pharmacology , Gene Expression/drug effects , Glycoproteins/pharmacology , Immunotoxins/pharmacology , Virulence Factors , Amino Acid Sequence , Animals , Bacterial Toxins/pharmacology , Base Sequence , Breast Neoplasms , Cell Line , DNA Primers , Exotoxins/biosynthesis , Exotoxins/isolation & purification , Female , Glycoproteins/biosynthesis , Glycoproteins/isolation & purification , Humans , Immunotoxins/isolation & purification , Kidney/metabolism , Kinetics , Leukemia, T-Cell , Lung Neoplasms , Male , Molecular Sequence Data , Neuregulins , Ovarian Neoplasms , Phosphorylation , Phosphotyrosine , Polymerase Chain Reaction , Prostatic Neoplasms , Pseudomonas aeruginosa , RNA, Messenger/metabolism , Rats , Receptor, ErbB-4 , Recombinant Fusion Proteins , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL