Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Appl Immunohistochem Mol Morphol ; 17(1): 57-67, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18815565

ABSTRACT

The inappropriate expression of the c-MET cell surface receptor in many human solid tumors necessitates the development of companion diagnostics to identify those patients who could benefit from c-MET targeted therapies. Tumor tissues are formalin fixed and paraffin embedded (FFPE) for histopathologic evaluation, making the development of an antibody against c-MET that accurately and reproducibly detects the protein in FFPE samples an urgent need. We have developed a monoclonal antibody (mAb), designated MET4, from a panel of MET-avid mAbs, based on its specific staining pattern in FFPE preparations. The accuracy of MET4 immunohistochemistry (MET4-IHC) was assessed by comparing MET4-IHC in FFPE cell pellets with immunoblotting analysis. The technical reproducibility of MET4-IHC possessed a percentage coefficient of variability of 6.25% in intra-assay and interassay testing. Comparison with other commercial c-MET antibody detection reagents demonstrated equal specificity and increased sensitivity for c-MET detection in prostate tissues. In cohorts of ovarian cancers and gliomas, MET4 reacted with ovarian cancers of all histologic subtypes (strong staining in 25%) and with 63% of gliomas. In addition, MET4 bound c-MET on the surfaces of cultured human cancer cells and tumor xenografts. In summary, the MET4 mAb accurately and reproducibly measures c-MET expression by IHC in FFPE tissues and can be used for molecular imaging in vivo. These properties encourage further development of MET4 as a multipurpose molecular diagnostics reagent to help to guide appropriate selection of patients being considered for treatment with c-MET-antagonistic drugs.


Subject(s)
Antibodies, Monoclonal , Biomarkers, Tumor/analysis , Neoplasms/pathology , Proto-Oncogene Proteins c-met/analysis , Biomarkers, Tumor/standards , Female , Formaldehyde , Glioma , Humans , Immunohistochemistry , Male , Neoplasms/chemistry , Neoplasms/diagnosis , Ovarian Neoplasms , Paraffin Embedding , Tissue Fixation
3.
J Transl Med ; 6: 77, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-19055779

ABSTRACT

Animal models greatly facilitate understanding of cancer and importantly, serve pre-clinically for evaluating potential anti-cancer therapies. We developed an invasive orthotopic human glioblastoma multiforme (GBM) mouse model that enables real-time tumor ultrasound imaging and pre-clinical evaluation of anti-neoplastic drugs such as 17-(allylamino)-17-demethoxy geldanamycin (17AAG). Clinically, GBM metastasis rarely happen, but unexpectedly most human GBM tumor cell lines intrinsically possess metastatic potential. We used an experimental lung metastasis assay (ELM) to enrich for metastatic cells and three of four commonly used GBM lines were highly metastatic after repeated ELM selection (M2). These GBM-M2 lines grew more aggressively orthotopically and all showed dramatic multifold increases in IL6, IL8, MCP-1 and GM-CSF expression, cytokines and factors that are associated with GBM and poor prognosis. DBM2 cells, which were derived from the DBTRG-05MG cell line were used to test the efficacy of 17AAG for treatment of intracranial tumors. The DMB2 orthotopic xenografts form highly invasive tumors with areas of central necrosis, vascular hyperplasia and intracranial dissemination. In addition, the orthotopic tumors caused osteolysis and the skull opening correlated to the tumor size, permitting the use of real-time ultrasound imaging to evaluate antitumor drug activity. We show that 17AAG significantly inhibits DBM2 tumor growth with significant drug responses in subcutaneous, lung and orthotopic tumor locations. This model has multiple unique features for investigating the pathobiology of intracranial tumor growth and for monitoring systemic and intracranial responses to antitumor agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzoquinones/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/pathology , Lactams, Macrocyclic/therapeutic use , Xenograft Model Antitumor Assays , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Glioblastoma/blood supply , Humans , Imaging, Three-Dimensional , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis
4.
Mol Cancer Ther ; 7(3): 648-58, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18319331

ABSTRACT

We hypothesized that signaling through multiple mitogen-activated protein kinase (MAPK) kinase (MKK) pathways is essential for the growth and vascularization of soft-tissue sarcomas, which are malignant tumors derived from mesenchymal tissues. We tested this using HT-1080, NCI, and Shac fibrosarcoma-derived cell lines and anthrax lethal toxin (LeTx), a bacterial toxin that inactivates MKKs. Western blots confirmed that LeTx treatment reduced the levels of phosphorylated extracellular signal-regulated kinase and p38 MAPK in vitro. Although short treatments with LeTx only modestly affected cell proliferation, sustained treatment markedly reduced cell numbers. LeTx also substantially inhibited the extracellular release of angioproliferative factors including vascular endothelial growth factor, interleukin-8, and basic fibroblast growth factor. Similar results were obtained with cell lines derived from malignant fibrous histiocytomas, leiomyosarcomas, and liposarcomas. In vivo, LeTx decreased MAPK activity and blocked fibrosarcoma growth. Growth inhibition correlated with decreased cellular proliferation and extensive necrosis, and it was accompanied by a decrease in tumor mean vessel density as well as a reduction in serum expression of angioproliferative cytokines. Vital imaging using high-resolution ultrasound enhanced with contrast microbubbles revealed that the effects of LeTx on tumor perfusion were remarkably rapid (<24 h) and resulted in a marked reduction of perfusion within the tumor but not in nontumor tissues. These results are consistent with our initial hypothesis and lead us to propose that MKK inhibition by LeTx is a broadly effective strategy for targeting neovascularization in fibrosarcomas and other similar proliferative lesions.


Subject(s)
Cell Division , Fibrosarcoma/blood supply , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic , Signal Transduction , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Fibrosarcoma/pathology , Humans
5.
Clin Cancer Res ; 13(20): 6049-55, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17947467

ABSTRACT

PURPOSE: Aberrant c-Met expression has been implicated in most types of human cancer. We are developing Met-directed imaging and therapeutic agents. EXPERIMENTAL DESIGN: To seek peptides that bind specifically to receptor Met, the Met-expressing cell lines S114 and SK-LMS-1 were used for biopanning with a random peptide phage display library. Competition ELISA, fluorescence-activated cell sorting analysis, an internalization assay, and a cell proliferation assay were used to characterize a Met-binding peptide in vitro. To evaluate the utility of the peptide as a diagnostic agent in vivo, 125I-labeled peptide was injected i.v. into nude mice bearing s.c. xenografts of the Met-expressing and hepatocyte growth factor (HGF)/scatter factor-expressing SK-LMS-1/HGF, and total body scintigrams were obtained between 1 and 24 h postinjection. RESULTS: One Met-binding peptide (YLFSVHWPPLKA), designated Met-pep1, reacts with Met on the cell surface and competes with HGF/scatter factor binding to Met in a dose-dependent manner. Met-pep1 is internalized by Met-expressing cells after receptor binding. Met-pep1 inhibits human leiomyosarcoma SK-LMS-1 cell proliferation in vitro. In SK-LMS-1 mouse xenografts, tumor-associated activity was imaged as early as 1 h postinjection and remained visible in some animals as late as 24 h postinjection. CONCLUSIONS: Met-pep1 specifically interacts with Met: it is internalized by Met-expressing cells and inhibits tumor cell proliferation in vitro; it is a potential diagnostic agent for tumor imaging.


Subject(s)
Gene Expression Regulation, Neoplastic , Peptide Library , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hepatocyte Growth Factor/metabolism , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Transplantation , Peptides/chemistry
6.
Clin Cancer Res ; 11(19 Pt 2): 7064s-7069s, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203803

ABSTRACT

PURPOSE: Met, an oncogene product and receptor tyrosine kinase, is a keystone molecule for malignant progression in solid human tumors. We are developing Met-directed imaging and therapeutic agents, including anti-Met monoclonal antibodies (MetSeek). In this study, we compared two antibodies, Met5 and Met3, for nuclear imaging of human and canine Met-expressing tumor xenografts in nude mice. EXPERIMENTAL DESIGN: Xenografts representing cancers of three different human tissue origins and metastatic canine prostate cancer were raised s.c. in host athymic nude mice. Animals were injected i.v. with I-125-Met5 or I-125-Met3, posterior total body gamma camera images were acquired for several days postinjection, and quantitative region-of-interest activity analysis was done. RESULTS: PC-3, SK-LMS-1/HGF, and CNE-2 xenografts imaged with I-125-Met5 were compared with PC-3, SK-LMS-1/HGF, and DU145 xenografts imaged with I-125-Met3. Nuclear imaging contrast was qualitatively similar for I-125-Met5 and I-125-Met3 in PC-3 and SK-LMS-1/HGF host mice. However, by region-of-interest analysis, the set of human tumors imaged with I-125-Met3 exhibited a pattern of rapid initial tumor uptake followed by a continuous decline in activity, whereas the set of human tumors imaged with I-125-Met5 showed slow initial uptake, peak tumor-associated activity at 1 day postinjection, and persistence of activity in xenografts for at least 5 days. GN4 canine prostate cancer xenografts were readily imaged with I-125-Met5. CONCLUSIONS: We conclude that radioiodinated Met3 and Met5 offer qualitatively similar nuclear images in xenograft-bearing mice, but quantitative considerations indicate that Met5 might be more useful for radioimmunotherapy. Moreover, canine prostate cancer seems to be a suitable model for second-stage preclinical evaluation of Met5.


Subject(s)
Antibodies, Monoclonal/chemistry , Neoplasms/diagnosis , Neoplasms/pathology , Proto-Oncogene Proteins c-met/biosynthesis , Animals , Dogs , Female , Humans , Image Processing, Computer-Assisted , Male , Mice , Mice, Nude , Nasopharyngeal Neoplasms/pathology , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-met/physiology , RNA, Small Interfering/metabolism , Radioimmunotherapy/methods , Radionuclide Imaging , Time Factors
7.
Mol Biotechnol ; 31(1): 41-54, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16118414

ABSTRACT

Inappropriate expression of the receptor tyrosine kinase Met and its ligand hepatocyte growth factor (HGF)/scatter factor (SF) is usually associated with an aggressive solid tumor phenotype (angiogenesis, invasiveness, and metastasis) and poor clinical prognosis. We report here the design and construction of a large, human naïve antigen-binding fragment (Fab) phage-display library with a diversity of 2.0 x 109, which allows rapid isolation of antigen-specific human antibody fragments. A Fab fragment specifically against Met (designated hFab-Met-1) was successively selected from this library by using biopanning on Met-transfected cell line S114. The specificity of hFab-Met-1 was characterized by immunoprecipitation, Western blotting, and flow cytometry. The results demonstrate that hFab-Met-1 reacts with the extracellular domain of Met in its native conformation. Moreover, functional analysis by Madine-Darby canine kidney cell scattering and urokinase-type plasminogen activator assays demonstrated that hFab-Met-1 is not an agonist to HGF/Met signaling compared with a murine intact monoclonal antibody (MAb) Met5. To confirm that hFab-Met-1 interacts with Met-expressing tumors in vivo, I-125-labeled hFab-Met-1 was nuclear-imaged in a mouse xenograft of Met- and HGF/SF-expressing human leiomyosarcoma. Total body scintigrams were obtained between 1 and 48 h postinjection (PI). Tumor-associated activity was imaged as early as 1 h PI, and remained visible in some animals as late as 24 h PI. As expected, activity was highest in the kidneys in early images, whereas thyroid activity became predominant in later images. In conclusion, hFab-Met-1 interacts with Met both in vitro and in vivo, and is a promising candidate for clinical diagnosis and therapeutics.


Subject(s)
Antibodies, Monoclonal , Immunoglobulin Fab Fragments/genetics , Peptide Library , Proto-Oncogene Proteins c-met/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Hepatocyte Growth Factor , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/therapeutic use , Immunoprecipitation , Leiomyosarcoma/diagnostic imaging , Leiomyosarcoma/drug therapy , Neoplasm Invasiveness , Polymerase Chain Reaction , Proto-Oncogene Proteins c-met/genetics , Radioimmunodetection , Radiopharmaceuticals , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Proc Natl Acad Sci U S A ; 102(30): 10528-33, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16024725

ABSTRACT

Invasive and proliferative phenotypes are fundamental components of malignant disease, yet basic questions persist about whether tumor cells can express both phenotypes simultaneously and, if so, what are their properties. Suitable in vitro models that allow characterization of cells that are purely invasive are limited because proliferation is required for cell maintenance. Here, we describe glioblastoma cells that are highly invasive in response to hepatocyte growth factor/scatter factor (HGF/SF). From this cell population, we selected subclones that were highly proliferative or displayed both invasive and proliferative phenotypes. The biological activities of invasion, migration, urokinase-type plasminogen activation, and branching morphogenesis exclusively partitioned with the highly invasive cells, whereas the highly proliferative subcloned cells uniquely displayed anchorage independent growth in soft agar and were highly tumorigenic as xenografts in immune-compromised mice. In response to HGF/SF, the highly invasive cells signal through the MAPK pathway, whereas the selection of the highly proliferative cells coselected for signaling through Myc. Moreover, in subcloned cells displaying both invasive and proliferative phenotypes, both signaling pathways are activated by HGF/SF. These results show how the mitogen-activated protein kinase and Myc pathways can cooperate to confer both invasive and proliferative phenotypes on tumor cells and provide a system for studying how transitions between invasion and proliferation can contribute to malignant progression.


Subject(s)
Cell Proliferation , Glioblastoma/pathology , Phenotype , Signal Transduction/physiology , Animals , Carcinogenicity Tests , Cell Line, Tumor , Female , Glioblastoma/physiopathology , Hepatocyte Growth Factor , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness
9.
Oncogene ; 24(23): 3697-707, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15782129

ABSTRACT

Induction of the urokinase-type plasminogen activator (uPA) by hepatocyte growth factor/scatter factor (HGF/SF) plays an important role in tumor cell invasion and metastasis that is mediated through the Met receptor tyrosine kinase. Geldanamycins (GA) are antitumor drugs that bind and inhibit HSP90 chaperone activity at nanomolar concentrations (nM-GAi) by preventing proper folding and functioning of certain oncoproteins. Previously, we have shown that a subset of GA derivatives exhibit exquisite potency, inhibiting HGF/SF-induced uPA-plasmin activation at femtomolar concentrations (fM-GAi) in canine MDCK cells. Here, we report that (1) inhibition of HGF/SF-induced uPA activity by fM-GAi is not uncommon, in that several human tumor glioblastoma cell lines (DBTRG, U373 and SNB19), as well as SK-LMS-1 human leiomyosarcoma cells are also sensitive to fM-GAi; (2) fM-GAi drugs only display inhibitory activity against HGF/SF-induced uPA activity (rather than basal activity), and only when the observed magnitude of uPA activity induction by HGF/SF is at least 1.5 times basal uPA activity; and (3) not only do fM-GAi derivatives strongly inhibit uPA activity but they also block MDCK cell scattering and in vitro invasion of human glioblastoma cells at similarly low drug concentrations. These effects of fM-GAi drugs on the Met-activated signaling pathway occur at concentrations well below those required to measurably affect Met expression or cell proliferation. We also examined the effect of Radicicol (RA), a drug with higher affinity than GA for HSP90. RA displays uPA activity inhibition at nanomolar levels, but not at lower concentrations, indicating that HSP90 is not likely the fM-GAi molecular target. Thus, we show that certain GA drugs (fM-GAi) in an HGF/SF-dependent manner block uPA-plasmin activation in tumor cells at femtomolar levels. This inhibition can also be observed in scattering and in vitro invasion assays. Our findings also provide strong circumstantial evidence for a novel non-HSP90 molecular target that is involved in HGF/SF-mediated tumor cell invasion.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Hepatocyte Growth Factor/antagonists & inhibitors , Quinones/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Benzoquinones , Cell Line , Dogs , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Neoplasm Invasiveness , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/metabolism
10.
Oncogene ; 24(1): 101-6, 2005 Jan 06.
Article in English | MEDLINE | ID: mdl-15531925

ABSTRACT

Downstream signaling that results from the interaction of hepatocyte growth factor/scatter factor (HGF/SF) with the receptor tyrosine kinase Met plays critical roles in tumor development, progression, and metastasis. This ligand-receptor pair is an attractive target for new diagnostic and therapeutic agents, preclinical development of which requires suitable animal models. The growth of heterotopic and orthotopic Met-expressing human tumor xenografts in conventional strains of immunocompromised mice inadequately replicates the paracrine stimulation by human HGF/SF (hHGF/SF) that occurs in humans with cancer. We have therefore generated a mouse strain transgenic for hHGF/SF (designated hHGF-Tg) on a severe combined immunodeficiency (SCID) background. We report here that the presence of ectopically expressed hHGF/SF ligand significantly enhances growth of heterotopic subcutaneous xenografts derived from human Met-expressing cancer cells, including the lines SK-LMS-1 (human leiomyosarcoma), U118 (human glioblastoma), and DU145 (human prostate carcinoma), but not that of M14-Mel xenografts (human melanoma that expresses insignificant levels of Met). Our results indicate that ectopic hHGF/SF can specifically activate Met in human tumor xenografts. This new hHGF-Tg strain of mice should provide a powerful tool for evaluating drugs and diagnostic agents that target the various pathways influenced by Met activity.


Subject(s)
Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Growth Factor/metabolism , Transplantation, Heterologous , Animals , Hepatocyte Growth Factor/genetics , Humans , Immunocompromised Host/immunology , Mice , Mice, SCID , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/genetics , Time Factors , Tumor Cells, Cultured
11.
Clin Cancer Res ; 9(10 Pt 2): 3839S-44S, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14506181

ABSTRACT

PURPOSE: Inappropriate expression of the receptor tyrosine kinase Met and its ligand is associated with an aggressive phenotype and poor clinical prognosis for a wide variety of solid human tumors. We are developing imaging and therapeutic agents that target this receptor-ligand complex. In this study, we evaluated the ability of radioiodinated anti-Met monoclonal antibodies from a single hybridoma clone to image human Met-expressing tumor xenografts. EXPERIMENTAL DESIGN: Xenografts of four different tissue origins were raised s.c. in host athymic nude mice. Animals received i.v. injections of I-125-Met3, posterior total body gamma camera images were acquired for several days after injection, and quantitative region-of-interest activity analysis was performed. RESULTS: The autocrine Met-expressing tumors S-114 and SK-LMS-1/HGF and the paracrine Met-expressing human prostate carcinoma PC-3 were satisfactorily imaged with I-125-Met3. By region-of-interest analysis, mean initial tumor-associated activities in S-114, SK-LMS-1/HGF, and PC-3 were 18.6 +/- 2.1, 7.2 +/- 2.2, and 5.4 +/- 2.6% estimated injected activity, and the mean ratios of tumor:total body activity at 3 days after injection were 0.32 +/- 0.13, 0.15 +/- 0.06, and 0.10 +/- 0.04, respectively. Human melanoma xenografts, however, accounted for < or =3% of injected or total body activity. We observed a direct rank order correlation between relative levels of Met3-derived radioactivity in xenografts and relative quantities of Met expressed by the respective cultured tumor cell lines. CONCLUSIONS: We conclude that I-125-Met3 is effective for imaging human Met-expressing xenografts of different tissue origins, and we infer that I-125-Met3 distinguishes human tumor xenografts according to their levels of Met expression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Radioimmunodetection/methods , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Hybridomas , Immunohistochemistry , Ligands , Mice , Mice, Nude , Neoplasm Transplantation , Phenotype , Protein Binding , Proto-Oncogene Proteins c-met/chemistry , Time Factors
12.
J Cell Biochem Suppl ; 39: 184-93, 2002.
Article in English | MEDLINE | ID: mdl-12552618

ABSTRACT

The availability of a test to assess the likelihood that a given tumor will invade or metastasize would be a useful development in clinical oncology. We propose that multimodality imaging of tumor expression of Met could serve as a prototype for metastatic risk stratification (MRS). Met, a receptor protein tyrosine kinase, is expressed by most solid tumors, and aberrant expression of Met is associated with poor clinical prognosis. We summarize the current status and predict the future direction of research in four areas of molecular imaging and cancer therapy that exploit Met.


Subject(s)
Diagnostic Imaging/methods , Neoplasm Metastasis/diagnosis , Neoplasms/diagnosis , Proto-Oncogene Proteins c-met/analysis , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Humans , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Prognosis , Proto-Oncogene Proteins c-met/metabolism
13.
Mol Imaging ; 1(1): 56-62, 2002.
Article in English | MEDLINE | ID: mdl-12920861

ABSTRACT

Inappropriate expression of the c-met-protooncogene product (Met) and/or of its ligand, hepatocyte growth factor/scatter factor (HGF/SF), has been correlated with poor prognosis in a variety of human solid tumors. We are developing animal models for nuclear imaging of Met and HGF/SF expression in tumors in vivo. We radioiodinated a mixture of monoclonal antibodies (MAbs) that bind to human HGF/SF and to the external ligand-binding domain of human Met, and then injected the I-125-MAb mixture intravenously into mice bearing tumors either autocrine for human HGF/SF and human Met or autocrine-paracrine for murine HGF/SF and murine Met. Serial total body gamma camera images were obtained, and regional activity was determined by quantitative region-of-interest (ROI) analysis. Tumors autocrine for human HGF/SF and Met demonstrated significantly more rapid uptake and more rapid clearance of the I-125-MAb mixture than tumors expressing one or both murine homologues, reaching a mean tumor to total body activity ratio of > 0.3 by 1 day postinjection. We conclude that radioimmunodetection of tumors autocrine for human HGF/SF and Met is feasible with an I-125-MAb mixture reactive against the ligand-receptor pair.


Subject(s)
Hepatocyte Growth Factor/analysis , Neoplasms/diagnostic imaging , Proto-Oncogene Proteins c-met/analysis , Radioimmunodetection/methods , Animals , Antibodies, Monoclonal , Binding Sites , Humans , Iodine Radioisotopes , Mice , Mice, Transgenic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...