Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Breast Cancer Res Treat ; 141(1): 43-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23959396

ABSTRACT

Trastuzumab is effective in the treatment of HER2/neu over-expressing breast cancer, but not all patients benefit from it. In vitro data suggest a role for HER3 in the initiation of signaling activity involving the AKT­mTOR pathway leading to trastuzumab insensitivity. We sought to investigate the potential of HER3 alone and in the context of p95HER2 (p95), a trastuzumab resistance marker, as biomarkers of trastuzumab escape. Using the VeraTag® assay platform, we developed a dual antibody proximity-based assay for the precise quantitation of HER3 total protein (H3T) from formalin-fixed paraffin-embedded (FFPE) breast tumors. We then measured H3T in 89 patients with metastatic breast cancer treated with trastuzumab-based therapy, and correlated the results with progression-free survival and overall survival using Kaplan­Meier and decision tree analyses that also included HER2 total (H2T) and p95 expression levels. Within the sub-population of patients that over-expressed HER2, high levels of HER3 and/or p95 protein expression were significantly associated with poor clinical outcomes on trastuzumab-based therapy. Based on quantitative H3T, p95, and H2T measurements, multiple subtypes of HER2-positive breast cancer were identified that differ in their outcome following trastuzumab therapy. These data suggest that HER3 and p95 are informative biomarkers of clinical outcomes on trastuzumab therapy, and that multiple subtypes of HER2-positive breast cancer may be defined by quantitative measurements of H3T, p95, and H2T.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/secondary , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Neoplasm Proteins/biosynthesis , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/classification , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , Decision Trees , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Neoplasm Proteins/genetics , Peptide Fragments/analysis , Peptide Fragments/immunology , Prognosis , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-3/genetics , Receptor, ErbB-3/immunology , Retrospective Studies , Single-Blind Method , Trastuzumab , Treatment Outcome
2.
Breast Cancer Res ; 13(2): R44, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21496232

ABSTRACT

INTRODUCTION: Ligand-bound and phosphorylated ErbB/HER heterodimers are potent signaling forms of this receptor family, and quantitative measurements of these active receptors may be predictive of patient response to targeted therapies. Using VeraTag technology, we developed and characterized quantitative assays measuring epidermal growth factor (EGF)-dependent increases in activated HER receptors in tumor cell line lysates and formalin-fixed, paraffin-embedded (FFPE) tumor sections. We demonstrated the ability of the assays to quantitatively measure changes in activated HER1 and HER2 receptor levels in cell lines following treatment with 2C4, erlotinib, and lapatinib. We utilized these assays to determine the prevalence and distribution of activated HER1, HER2, and HER1-HER2 heterodimers in 43 HER2-positive breast tumors. METHODS: Assays for activated HER1 and HER2 receptors in FFPE and cell lysate formats were developed using VeraTag technology, which requires the proximity of an antibody pair for light-dependent release of a fluorescently labeled tag, followed by capillary electrophoresis-based quantitation. RESULTS: Ligand-dependent and independent HER1-HER2 heterodimer levels measured by lysate and FFPE VeraTag assays trended with HER1 and HER2 expression levels in tumor cell lines, which was confirmed by co-immunoprecipitation. The formation of EGF-dependent HER1-HER2 heterodimers were inhibited by the HER2-targeted monoclonal antibody 2C4 and stabilized by the HER1 tyrosine kinase inhibitor (TKI) erlotinib. EGF-dependent HER1 and HER2 phosphorylation was inhibited by lapatinib and erlotinib. Further, we observed that dominant receptor signaling patterns may switch between HER1-HER1 and HER1-HER2, depending on drug mechanism of action and relative levels of HER receptors. In FFPE breast tumors that expressed both HER1 and HER2, HER1-HER2 heterodimers were detected in 25 to 50% of tumors, depending on detection method. The levels of activated phospho-HER1-HER2 heterodimers correlated with HER1 or HER2 levels in an analysis of 43 HER2-positive breast tumors. CONCLUSIONS: VeraTag lysate assays can be used as a tool for understanding the mechanism of action of targeted HER-family inhibitors in the preclinical setting, while VeraTag FFPE assays of activated HER receptors combined with total HER2 measurements (HERmark) in tumor samples may provide a more accurate prediction of clinical response to both HER1 and HER2 targeted therapies.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , ErbB Receptors/chemistry , Receptor, ErbB-2/chemistry , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Humans , Lapatinib , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , Signal Transduction , Staining and Labeling , Treatment Outcome
3.
PLoS One ; 6(1): e15932, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21283737

ABSTRACT

Aberrant activation of membrane receptors frequently occurs in human carcinomas. Detection of phosphorylated receptors is commonly used as an indicator of receptor activation in formalin-fixed paraffin embedded (FFPE) tumor specimens. FFPE is a standard method of specimen preparation used in the histological analysis of solid tumors. Due to variability in FFPE preparations and the labile nature of protein phosphorylation, measurements of phospho-proteins are unreliable and create ambiguities in clinical interpretation. Here, we describe an alternative, novel approach to measure receptor activation by detecting and quantifying ligand-receptor complexes in FFPE specimens. We used hepatocyte growth factor (HGF)-c-MET as our model ligand-receptor system. HGF is the only known ligand of the c-MET tyrosine kinase receptor and HGF binding triggers c-MET phosphorylation. Novel antibody proximity-based assays were developed and used to detect and quantify total c-MET, total HGF, and HGF-c-MET ligand-receptor interactions in FFPE cell line and tumor tissue. In glioma cells, autocrine activation of c-MET by HGF-c-MET increased basal levels of c-MET phosphorylation at tyrosine (Tyr) 1003. Furthermore, HGF-c-MET activation in glioma cell lines was verified by Surface Protein-Protein Interaction by Crosslinking ELISA (SPPICE) assay in corresponding soluble cell lysates. Finally, we profiled levels ofc-MET, HGF, and HGF-c-MET complexes in FFPE specimens of human Non-Small Cell Lung Cancer (NSCLC), Gastric Cancer, Head and Neck Squamous Cell, and Head and Neck Non-Squamous Cell carcinomas. This report describes a novel approach for the detection and quantification of ligand-receptor interactions that can be widely applied to measure receptor activation in FFPE preclinical models and archived FFPE human tissue specimens.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Hepatocyte Growth Factor/metabolism , Neoplasm Proteins/analysis , Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Biopsy , Cross-Linking Reagents , Formaldehyde , Hepatocyte Growth Factor/analysis , Humans , Ligands , Paraffin Embedding , Protein Binding , Proto-Oncogene Proteins c-met/analysis , Receptors, Growth Factor/analysis
4.
PLoS One ; 6(1): e16443, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21297994

ABSTRACT

BACKGROUND: The identification of patients for targeted antineoplastic therapies requires accurate measurement of therapeutic targets and associated signaling complexes. HER3 signaling through heterodimerization is an important growth-promoting mechanism in several tumor types and may be a principal resistance mechanism by which EGFR and HER2 expressing tumors elude targeted therapies. Current methods that can study these interactions are inadequate for formalin-fixed, paraffin-embedded (FFPE) tumor samples. METHODOLOGY AND PRINCIPAL FINDINGS: Herein, we describe a panel of proximity-directed assays capable of measuring protein-interactions and phosphorylation in FFPE samples in the HER3/PI3K/Akt pathway and examine the capability of these assays to inform on the functional state of the pathway. We used FFPE breast cancer cell line and tumor models for this study. In breast cancer cell lines we observe both ligand-dependent and independent activation of the pathway and strong correlations between measured activation of key analytes. When selected cell lines are treated with HER2 inhibitors, we not only observe the expected molecular effects based on mechanism of action knowledge, but also novel effects of HER2 inhibition on key targets in the HER receptor pathway. Significantly, in a xenograft model of delayed tumor fixation, HER3 phosphorylation is unstable, while alternate measures of pathway activation, such as formation of the HER3PI3K complex is preserved. Measurements in breast tumor samples showed correlations between HER3 phosphorylation and receptor interactions, obviating the need to use phosphorylation as a surrogate for HER3 activation. SIGNIFICANCE: This assay system is capable of quantitatively measuring therapeutically relevant responses and enables molecular profiling of receptor networks in both preclinical and tumor models.


Subject(s)
Breast Neoplasms/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Mice , Phosphorylation , Protein Binding , Receptor, ErbB-3/antagonists & inhibitors , Transplantation, Heterologous
5.
Patholog Res Int ; 2010: 814176, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-21151530

ABSTRACT

We report here the results of the analytical validation of assays that measure HER2 total protein (H2T) and HER2 homodimer (H2D) expression in Formalin Fixed Paraffin Embedded (FFPE) breast cancer tumors as well as cell line controls. The assays are based on the VeraTag technology platform and are commercially available through a central CAP-accredited clinical reference laboratory. The accuracy of H2T measurements spans a broad dynamic range (2-3 logs) as evaluated by comparison with cross-validating technologies. The measurement of H2T expression demonstrates a sensitivity that is approximately 7-10 times greater than conventional immunohistochemistry (IHC) (HercepTest). The HERmark assay is a quantitative assay that sensitively and reproducibly measures continuous H2T and H2D protein expression levels and therefore may have the potential to stratify patients more accurately with respect to response to HER2-targeted therapies than current methods which rely on semiquantitative protein measurements (IHC) or on indirect assessments of gene amplification (FISH).

6.
Clin Cancer Res ; 16(16): 4226-35, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20664024

ABSTRACT

PURPOSE: p95HER2 is an NH(2)-terminally truncated form of HER2 that lacks the trastuzumab binding site and is therefore thought to confer resistance to trastuzumab treatment. In this report, we introduce a new antibody that has enabled the first direct quantitative measurement of p95HER2 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissues. We sought to show that quantitative p95HER2 levels would correlate with outcome in trastuzumab-treated HER2-positive metastatic breast cancer. EXPERIMENTAL DESIGN: The novel p95HER2 antibody used here was characterized for sensitivity, specificity, and selectivity over full-length HER2. Quantitative p95HER2 levels were measured in 93 metastatic breast tumors using a VeraTag FFPE assay to determine the correlation of p95HER2 levels with outcomes. RESULTS: Within a cohort of trastuzumab-treated metastatic breast cancer patients, high levels of p95HER2 were found to correlate with shorter progression-free survival [hazard ratio (HR), 1.9; P = 0.017] and overall survival (HR, 2.2; P = 0.012) in patients with tumors selected to be HER2 positive by the VeraTag HER2 assay. For those with tumors found to be fluorescence in situ hybridization positive, elevated p95HER2 correlated similarly with shorter progression-free survival (HR, 1.8; P = 0.022) and overall survival (HR, 2.2; P = 0.009). CONCLUSIONS: We have successfully generated an antibody that can specifically detect p95HER2, and developed an assay to quantify expression in FFPE tumor specimens. Using this novel assay, we have identified a group of HER2-positive patients expressing p95HER2 that have a worse outcome while on trastuzumab. As p95HER2 retains sensitivity to kinase inhibitors, measurement of p95HER2 in breast tumor sections may be useful in guiding treatment for patients with HER2-positive breast cancer.


Subject(s)
Antibodies, Monoclonal , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, ErbB-2/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Blotting, Western , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Separation , Cohort Studies , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Formaldehyde , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Paraffin Embedding , Peptide Fragments/analysis , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , Sensitivity and Specificity , Tissue Fixation , Trastuzumab , Treatment Outcome
7.
Breast Cancer Res Treat ; 122(3): 685-97, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19859802

ABSTRACT

HER2 is gene amplified or over-expressed in 20-25% of breast cancers resulting in elevated HER2 activation. Trastuzumab (Herceptin), a humanized monoclonal antibody, targets activated HER2 and is clinically effective in HER2-over-expressing breast cancers. However, despite prolonged survival, treated breast cancer patients develop resistance. Resistance to trastuzumab occurs upon inactivation of HER2 regulatory proteins or upon up-regulation of alternative receptors. In particular, elevated levels of EGFR, present in estrogen receptor (ER) positive, trastuzumab-resistant BT-474 xenografts caused, a trastuzumab-resistant phenotype (Ritter et al. Clin Cancer Res 13:4909-4919, 2007). However, the role of EGFR in acquired trastuzumab resistance in ER negative cell models is not well defined. In this study, SKBR3 cell line clones expressing EGFR were generated to examine the role of EGFR over-expression on trastuzumab sensitivity in an, ER-negative breast carcinoma cell line. A stable clone, SKBR3/EGFR (clone 4) expressing moderate levels of EGFR remained sensitive to trastuzumab, whereas a stable clone, SKBR3/EGFR (clone 5) expressing high levels of EGFR, became resistant to trastuzumab. Depletion of EGFR by EGFR small-interfering RNAs in the SKBR3/EGFR (clone 5) reversed trastuzumab resistance. However, the SKBR3/EGFR (clone 5) cell line remained sensitive to lapatinib, an EGFR/HER2 inhibitor. Biochemical analysis using co-immunoprecipitation and proximity-based quantitative VeraTag assays demonstrated that high levels of EGFR phosphorylation, EGFR/EGFR homo-dimerization, and EGFR/HER2 hetero-dimerization were present in the trastuzumab-resistant cells. We conclude that EGFR over-expression can mediate trastuzumab resistance in both ER positive and ER negative cells and hypothesize that a threshold level of EGFR, in the absence of autocrine ligand production, is required to induce the resistant phenotype.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Antibodies, Monoclonal, Humanized , Blotting, Western , Breast Neoplasms/pathology , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Humans , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Trastuzumab , Tumor Cells, Cultured/drug effects
8.
J Nucl Med ; 50(3): 435-43, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19223400

ABSTRACT

UNLABELLED: To optimize in vivo tissue uptake kinetics and clearance of engineered monoclonal antibody (mAb) fragments for radiotherapeutic and radiodiagnostic applications, we compared the biodistribution and tumor localization of four (111)In- and (86)Y-labeled antibody formats, derived from a single antimindin/RG-1 mAb, in a prostate tumor model. The IgG, diabody, single-chain variable domain (scFv), and novel miniantibody formats, composed of the human IgE-C(H)4 and a modified IgG1 hinge linked to scFv domains, were compared. METHODS: Antibodies were first derivatized with the bifunctional chelator CHX-A''-diethylenetriamine pentaacetic acid and then bound to the radiometal to create radiolabeled immunoconjugates. Human LNCaP xenografts were grown in nude mice, and (111)In- or (86)Y-labeled antibodies were administered intravenously. Tissues were harvested at different times, and the level of antibody deposition was determined by measuring radioactivity. Whole-body small-animal PET of mice receiving (86)Y-labeled antibodies was performed at 6 time points and colocalized with simultaneous micro-CT imaging. RESULTS: The biodistributions of (111)In and (86)Y antibodies were quite similar. The blood, tumor, kidney, and liver tissues contained varying levels of radioactivity. The antibody accumulation in the tumor correlated with molecular size. The IgG steadily increased with time to 24.1 percentage injected dose per gram (%ID/g) at 48 h. The miniantibody accumulated at a similar rate to reach a lower level (14.2 %ID/g) at 48 h but with a higher tumor-to-blood ratio than the IgG. Tumor accumulation of the diabody peaked at 3 h, reaching a much lower level (3.7 %ID/g). A combination of rapid clearance and lower relative affinity of the scFv precluded deposition in the tumor. Small-animal PET results correlated well with the biodistribution results, with similar tumor localization patterns. CONCLUSION: The larger antibody formats (IgG and miniantibody) gave higher tumor uptake levels than did the smaller formats (diabody and scFv). These larger formats may be more suitable for radioimmunotherapy applications, evidenced by the preclinical efficacy previously shown by a report on the IgG format. The smaller formats were rapidly cleared from circulation, and the diabody, which accumulated in the tumor, may be more suitable for radiodiagnostic applications.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Chelating Agents/chemistry , Humans , Indium Radioisotopes , Isothiocyanates/chemistry , Male , Mice , Mice, Nude , Neoplasm Transplantation , Pentetic Acid/analogs & derivatives , Pentetic Acid/chemistry , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , Tissue Distribution , Tomography, X-Ray Computed , Transplantation, Heterologous , Yttrium Radioisotopes
9.
Front Biosci ; 12: 5052-9, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17569629

ABSTRACT

Hepsin is a membrane serine protease expressed in several human tissues including the liver, kidney, prostate, and thyroid. The physiological function of hepsin remains unknown. In vitro studies have shown that hepsin activates blood clotting factors VII, XII, and IX, pro-urokinase (pro-uPA), and pro-hepatocyte growth factor (pro-HGF). Recently, hepsin has been identified as one of the most up-regulated genes in prostate cancer. The hepsin up-regulation appears to correlate with the disease progression. In a mouse model of prostate cancer, hepsin overexpression promotes cancer progression and metastasis. In culture, anti-hepsin antibodies inhibited the invasion of human prostate cancer cells. This review will outline the molecular biology and biochemistry of hepsin and highlight recent data of hepsin in prostate cancer.


Subject(s)
Prostatic Neoplasms/enzymology , Serine Endopeptidases/physiology , Animals , Cell Proliferation , Humans , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Up-Regulation
10.
Cancer Res ; 66(7): 3611-9, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16585186

ABSTRACT

Hepsin is a type II transmembrane serine protease that is expressed in normal liver, and at lower levels in kidney, pancreas, and testis. Several studies have shown that hepsin mRNA is significantly elevated in most prostate tumors, as well as a significant fraction of ovarian and renal cell carcinomas and hepatomas. Although the overexpression of mRNA in these tumors has been extensively documented, there has been conflicting literature on whether hepsin plays a role in tumor cell growth and progression. Early literature implied a role for hepsin in human tumor cell proliferation, whereas recent studies with a transgenic mouse model for prostate cancer support a role for hepsin in tumor progression and metastases. To evaluate this issue further, we have expressed an activatable form of hepsin, and have generated a set of monoclonal antibodies that neutralize enzyme activity. The neutralizing antibodies inhibit hepsin enzymatic activity in biochemical and cell-based assays. Selected neutralizing and nonneutralizing antibodies were used in cell-based assays with tumor cells to evaluate the effect of antibodies on tumor cell growth and invasion. Neutralizing antibodies failed to inhibit the growth of prostate, ovarian, and hepatoma cell lines in culture. However, potent inhibitory effects of the antibodies were seen on invasion of ovarian and prostate cells in transwell-based invasion assays. These results support a role for hepsin in tumor cell progression but not in primary tumor growth. Consistent with this, immunohistochemical experiments with a mouse monoclonal antibody reveal progressively increased staining of prostate tumors with advanced disease, and in particular, extensive staining of bone metastatic lesions.


Subject(s)
Antibodies, Monoclonal/pharmacology , Ovarian Neoplasms/enzymology , Prostatic Neoplasms/enzymology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cloning, Molecular , Female , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Proteinase Inhibitors/immunology
11.
Cancer Res ; 65(18): 8397-405, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166318

ABSTRACT

Gene expression analysis showed that a human mindin homologue, mindin/RG-1, is expressed selectively in prostate tissues and that its expression level is elevated in some prostate tumors. Mindin/RG-1 protein expression is maintained in >80% of prostate cancers metastatic to bone or lymph nodes as well as in locally recurrent tumors in androgen-unresponsive patients. In contrast, mindin/RG-1 expression in other normal tissues is significantly lower than that seen in the prostate. A fully human antibody, 19G9, was generated against mindin/RG-1 protein and was shown to accumulate at high abundance in LNCaP tumor xenografts. Conjugates of this antibody with the chelator CHX-A''-DTPA were generated and radiolabeled with either 111In, 90Y, or 86Y. Small animal positron emission tomography imaging with the 86Y-radiolabeled conjugate showed very specific accumulation of the antibody in LNCaP tumor xenografts with clear tumor delineation apparent at 4 hours. The therapeutic efficacy of [90Y]-CHX-A''-DTPA-19G9 was evaluated in mice bearing LNCaP xenografts. A dose-finding study identified a nontoxic therapeutic dose to be approximately 75 microCi. Significant antitumor effects were seen with a single administration of radiolabeled antibody to animals bearing 200 to 400 mm3 tumors. Inhibition of tumor growth was observed in all treated animals over a 49-day period. At 49 days posttreatment, slow tumor growth recurred but this could be prevented for an additional 40-day period by a second administration of a 75 microCi dose at day 49. We conclude that [90Y]-CHX-A''-DTPA-19G9 is a novel antibody conjugate that has considerable promise for therapy of metastatic prostate cancer in androgen-unresponsive patients.


Subject(s)
Antibodies, Monoclonal/immunology , Extracellular Matrix Proteins/immunology , Immunotoxins/immunology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , CHO Cells , Cricetinae , Dose-Response Relationship, Immunologic , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Isothiocyanates/immunology , Isothiocyanates/pharmacokinetics , Isothiocyanates/pharmacology , Male , Molecular Sequence Data , Pentetic Acid/analogs & derivatives , Pentetic Acid/immunology , Pentetic Acid/pharmacokinetics , Pentetic Acid/pharmacology , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tissue Distribution , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/administration & dosage , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/pharmacology
12.
Cancer Res ; 65(7): 2846-53, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805286

ABSTRACT

Radiotherapy is an effective approach for the treatment of local prostate cancer. However, once prostate cancer metastasizes, radiotherapy cannot be used due to the distribution of multiple metastases to lymph nodes and bones. In contrast, radioimmunotherapy should still be efficacious in metastatic prostate cancer as radioisotopes are brought to tumor cells by targeting antibodies. Here we identify and validate a prostate-expressed molecule, tomoregulin, as a target for radioimmunotherapy of prostate cancer. Tomoregulin is a transmembrane protein selectively expressed in the brain, prostate, and prostate cancer, but not expressed in other normal tissues. Immunohistochemical studies of tomoregulin protein in clinical samples show its location in the luminal epithelium of normal prostate, benign prostatic hyperplasia, and prostatic intraepithelial neoplasia. More importantly, the tomoregulin protein is expressed in primary prostate tumors and in their lymph node and bone metastases. The nature of tomoregulin as a transmembrane protein and its tissue-specific expression make tomoregulin an attractive target for radioimmunotherapy, in which tomoregulin-specific antibodies will deliver a radioisotope to prostate tumor cells and metastases. Indeed, biodistribution studies using a prostate tumor xenograft model showed that the (111)In-labeled anti-tomoregulin antibody 2H8 specifically recognizes tomoregulin protein in vivo, leading to a strong tumor-specific accumulation of the antibody. In efficacy studies, a single i.p. dose of 150 microCi (163 microg) (90)Y-labeled 2H8 substantially inhibits the growth rate of established LNCaP human prostate tumor xenograft in nude mice but produces no overt toxicity despite cross-reactivity of 2H8 with mouse tomoregulin. Our data clearly validate tomoregulin as a target for radioimmunotherapy of prostate cancer.


Subject(s)
Immunotoxins/therapeutic use , Indium Radioisotopes/therapeutic use , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Ytterbium/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Cell Line, Tumor , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Prostate/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radioimmunotherapy , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Tissue Distribution , Xenograft Model Antitumor Assays
13.
Mol Cancer Ther ; 1(14): 1293-304, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12516962

ABSTRACT

Drug discovery strategies are needed that can rapidly exploit multiple therapeutic targets associated with the complex gene expression changes that characterize a polygenic disease such as cancer. We report a new cell-based high-throughput technology for screening chemical libraries against several potential cancer target genes in parallel. Multiplex gene expression (MGE) analysis provides direct and quantitative measurement of multiple endogenous mRNAs using a multiplexed detection system coupled to reverse transcription-PCR. A multiplex assay for six genes overexpressed in cancer cells was used to screen 9000 chemicals and known drugs in the human prostate cancer cell line PC-3. Active compounds that modulated gene expression levels were identified, and IC50 values were determined for compounds that bind DNA, cell surface receptors, and components of intracellular signaling pathways. A class of steroids related to the cardiac glycosides was identified that potently inhibited the plasma membrane Na(+)K(+)-ATPase resulting in the inhibition of four of the prostate target genes including transcription factors Hoxb-13, hPSE/PDEF, hepatocyte nuclear factor-3alpha, and the inhibitor of apoptosis, survivin. Representative compounds selectively induced apoptosis in PC-3 cells compared with the nonmetastatic cell line BPH-1. The multiplex assay distinguished potencies among structural variants, enabling structure-activity analysis suitable for chemical optimization studies. A second multiplex assay for five toxicological markers, Hsp70, Gadd153, Gadd45, O6-methylguanine-DNA methyltransferase, and cyclophilin, detected compounds that caused DNA damage and cellular stress and was a more sensitive and specific indicator of potential toxicity than measurement of cell viability. MGE analysis facilitates rapid drug screening and compound optimization, the simultaneous measurement of toxicological end points, and gene function analysis.


Subject(s)
Drug Design , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Up-Regulation , Actins/metabolism , Apoptosis , CCAAT-Enhancer-Binding Proteins/metabolism , Coloring Agents/pharmacology , Cyclophilins/metabolism , DNA/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins , Kinetics , Male , Models, Chemical , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proteins/metabolism , RNA, Messenger/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Structure-Activity Relationship , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Transcription Factor CHOP , Transcription Factors/metabolism , Tumor Cells, Cultured , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...