Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Proc Natl Acad Sci U S A ; 110(32): E2987-96, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23882082

ABSTRACT

Binding of hepatocyte growth factor (HGF) to the receptor tyrosine kinase MET is implicated in the malignant process of multiple cancers, making disruption of this interaction a promising therapeutic strategy. However, targeting MET with bivalent antibodies can mimic HGF agonism via receptor dimerization. To address this limitation, we have developed onartuzumab, an Escherichia coli-derived, humanized, and affinity-matured monovalent monoclonal antibody against MET, generated using the knob-into-hole technology that enables the antibody to engage the receptor in a one-to-one fashion. Onartuzumab potently inhibits HGF binding and receptor phosphorylation and signaling and has antibody-like pharmacokinetics and antitumor activity. Biochemical data and a crystal structure of a ternary complex of onartuzumab antigen-binding fragment bound to a MET extracellular domain fragment, consisting of the MET Sema domain fused to the adjacent Plexins, Semaphorins, Integrins domain (MET Sema-PSI), and the HGF ß-chain demonstrate that onartuzumab acts specifically by blocking HGF α-chain (but not ß-chain) binding to MET. These data suggest a likely binding site of the HGF α-chain on MET, which when dimerized leads to MET signaling. Onartuzumab, therefore, represents the founding member of a class of therapeutic monovalent antibodies that overcomes limitations of antibody bivalency for targets impacted by antibody crosslinking.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Mice, SCID , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Neoplasms/pathology , Protein Binding/drug effects , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Sequence Homology, Amino Acid
2.
PLoS Genet ; 9(2): e1003228, 2013.
Article in English | MEDLINE | ID: mdl-23459311

ABSTRACT

The alveolar compartment, the fundamental gas exchange unit in the lung, is critical for tissue oxygenation and viability. We explored hepatocyte growth factor (HGF), a pleiotrophic cytokine that promotes epithelial proliferation, morphogenesis, migration, and resistance to apoptosis, as a candidate mediator of alveolar formation and regeneration. Mice deficient in the expression of the HGF receptor Met in lung epithelial cells demonstrated impaired airspace formation marked by a reduction in alveolar epithelial cell abundance and survival, truncation of the pulmonary vascular bed, and enhanced oxidative stress. Administration of recombinant HGF to tight-skin mice, an established genetic emphysema model, attenuated airspace enlargement and reduced oxidative stress. Repair in the TSK/+ mouse was punctuated by enhanced akt and stat3 activation. HGF treatment of an alveolar epithelial cell line not only induced proliferation and scattering of the cells but also conferred protection against staurosporine-induced apoptosis, properties critical for alveolar septation. HGF promoted cell survival was attenuated by akt inhibition. Primary alveolar epithelial cells treated with HGF showed improved survival and enhanced antioxidant production. In conclusion, using both loss-of-function and gain-of-function maneuvers, we show that HGF signaling is necessary for alveolar homeostasis in the developing lung and that augmentation of HGF signaling can improve airspace morphology in murine emphysema. Our studies converge on prosurvival signaling and antioxidant protection as critical pathways in HGF-mediated airspace maintenance or repair. These findings support the exploration of HGF signaling enhancement for diseases of the airspace.


Subject(s)
Hepatocyte Growth Factor , Homeostasis , Proto-Oncogene Proteins c-met , Pulmonary Alveoli , Animals , Cell Movement/genetics , Cell Proliferation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hepatocyte Growth Factor/administration & dosage , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Mice , Morphogenesis/genetics , Morphogenesis/physiology , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiology , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/physiopathology , Signal Transduction , Tissue Survival/genetics
3.
Clin Cancer Res ; 14(23): 7733-40, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047100

ABSTRACT

PURPOSE: Apomab is a fully human monoclonal antibody that induces programmed cell death through the proapoptotic receptor DR5 in various cancer cells but not in normal cells. Several lung cancer cell lines express DR5 and exhibit apoptosis in response to apomab in vitro. EXPERIMENTAL DESIGN: We investigated the efficacy of apomab and its interaction with chemotherapy in xenograft models based on human NCI-H460 non-small-cell lung carcinoma cells. In an established model of s.c. tumor xenografts, apomab or Taxol plus carboplatin chemotherapy delayed tumor progression, whereas combined treatment caused tumor regression and a substantially longer growth delay. To test apomab activity in a setting that may more closely mimic lung cancer pathology in patients, we developed a lung orthotopic model. RESULTS: In this model, microcomputed tomography imaging showed that apomab, chemotherapy, or combination treatment significantly inhibited tumor growth compared with vehicle, whereas the combination caused greater inhibition in tumor growth relative to chemotherapy or apomab. Similarly, histologic analysis revealed that apomab, chemotherapy, or the combination significantly reduced tumor size compared with vehicle, whereas the combination induced significantly greater reduction in tumor size than did chemotherapy or apomab. Furthermore, combined treatment improved 105-day survival relative to vehicle (P = 0.0023) as well as to apomab (P = 0.0445) or chemotherapy (P = 0.0415). CONCLUSION: These results show a positive interaction of apomab with chemotherapy, evidenced by significant inhibition of tumor growth as well as improved survival, thus supporting further investigation of this therapeutic approach in lung cancer patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Animals , Apoptosis/drug effects , Carboplatin/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Immunoblotting , In Situ Nick-End Labeling , Mice , Mice, Nude , Paclitaxel/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Res ; 68(22): 9280-90, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010901

ABSTRACT

HER2 is a validated target in breast cancer therapy. Two drugs are currently approved for HER2-positive breast cancer: trastuzumab (Herceptin), introduced in 1998, and lapatinib (Tykerb), in 2007. Despite these advances, some patients progress through therapy and succumb to their disease. A variation on antibody-targeted therapy is utilization of antibodies to deliver cytotoxic agents specifically to antigen-expressing tumors. We determined in vitro and in vivo efficacy, pharmacokinetics, and toxicity of trastuzumab-maytansinoid (microtubule-depolymerizing agents) conjugates using disulfide and thioether linkers. Antiproliferative effects of trastuzumab-maytansinoid conjugates were evaluated on cultured normal and tumor cells. In vivo activity was determined in mouse breast cancer models, and toxicity was assessed in rats as measured by body weight loss. Surprisingly, trastuzumab linked to DM1 through a nonreducible thioether linkage (SMCC), displayed superior activity compared with unconjugated trastuzumab or trastuzumab linked to other maytansinoids through disulfide linkers. Serum concentrations of trastuzumab-MCC-DM1 remained elevated compared with other conjugates, and toxicity in rats was negligible compared with free DM1 or trastuzumab linked to DM1 through a reducible linker. Potent activity was observed on all HER2-overexpressing tumor cells, whereas nontransformed cells and tumor cell lines with normal HER2 expression were unaffected. In addition, trastuzumab-DM1 was active on HER2-overexpressing, trastuzumab-refractory tumors. In summary, trastuzumab-DM1 shows greater activity compared with nonconjugated trastuzumab while maintaining selectivity for HER2-overexpressing tumor cells. Because trastuzumab linked to DM1 through a nonreducible linker offers improved efficacy and pharmacokinetics and reduced toxicity over the reducible disulfide linkers evaluated, trastuzumab-MCC-DM1 was selected for clinical development.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Immunotoxins/therapeutic use , Maytansine/analogs & derivatives , Receptor, ErbB-2/analysis , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/toxicity , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/toxicity , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Maytansine/toxicity , Mice , Rats , Rats, Sprague-Dawley , Trastuzumab , Xenograft Model Antitumor Assays
5.
Cancer Res ; 68(11): 4360-8, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519697

ABSTRACT

The hepatocyte growth factor (HGF) and its receptor, c-Met, have been implicated in driving proliferation, invasion, and poor prognosis in pancreatic cancer. Here, we investigated the expression of HGF and c-Met in primary pancreatic cancers and described in vitro and in vivo models in which MetMAb, a monovalent antibody against c-Met, was evaluated. First, expression of HGF and MET mRNA was analyzed in 59 primary pancreatic cancers and 51 normal samples, showing that both factors are highly expressed in pancreatic cancer. We next examined HGF responsiveness in pancreatic cancer lines to select lines that proliferate in response to HGF. Based on these studies, two lines were selected for further in vivo model development: BxPC-3 (c-Met(+), HGF(-)) and KP4 (c-Met(+), HGF(+)) cells. As BxPC-3 cells are responsive to exogenous HGF, s.c. tumor xenografts were grown in a paracrine manner with purified human HGF provided by osmotic pumps, wherein MetMAb treatment significantly inhibited tumor growth. KP4 cells are autocrine for HGF and c-Met, and MetMAb strongly inhibited s.c. tumor growth. To better model pancreatic cancer and to enable long-term survival studies, an orthotopic model of KP4 was established. MetMAb significantly inhibited orthotopic KP4 tumor growth in 4-week studies monitored by ultrasound and also improved survival in 90-day studies. MetMAb significantly reduced c-Met phosphorylation in orthotopic KP4 tumors with a concomitant decrease in Ki-67 staining. These data suggest that the HGF/c-Met axis plays an important role in the progression of pancreatic cancer and that targeting c-Met therein may have therapeutic value.


Subject(s)
Adenocarcinoma/pathology , Antibodies, Monoclonal/immunology , Pancreatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/immunology , Adenocarcinoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Nude , Neovascularization, Pathologic/immunology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Phosphorylation , Signal Transduction , Survival Analysis
6.
Magn Reson Med ; 60(1): 64-72, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18421695

ABSTRACT

Tumor heterogeneity complicates the quantification of tumor microvascular characteristics assessed by dynamic contrast-enhanced MRI (DCE-MRI). To address this issue a novel approach was developed that combines DCE-MRI with diffusion-based multispectral (MS) analysis to quantify the microvascular characteristics of specific tumor tissue populations. Diffusion-based MS segmentation (feature space: apparent diffusion coefficient, T(2) and proton density) was performed to identify tumor tissue populations and the DCE-MRI characteristics were determined for each tissue class. The ability of this MS DCE-MRI technique to detect microvascular changes due to treatment with an antibody (G6-31) to vascular endothelial growth factor-A (VEGF) was evaluated in a tumor xenograft mouse model. Anti-VEGF treatment resulted in a significant reduction in K(trans) for the MS viable tumor tissue class (-0.0034 +/- 0.0022 min(-1), P < 0.01) at 24 hr posttreatment that differ significantly from the change observed in the control group (0.0002 +/- 0.0025 min(-1)). Viable tumor K(trans) for the anti-VEGF group was also reduced 62% relative to the pretreatment values (P < 0.01). Necrotic tissue classes were found to add only noise to DCE-MRI estimates. This approach provides a means to measure physiological parameters within the viable tumor and address the issue of tumor heterogeneity that complicates DCE-MRI analysis.


Subject(s)
Magnetic Resonance Imaging/methods , Neoplasms, Experimental/blood supply , Animals , Antibodies/therapeutic use , Female , Mice , Mice, Nude , Microcirculation/anatomy & histology , Tissue Survival , Vascular Endothelial Growth Factor A/immunology
7.
Cancer Res ; 67(1): 254-61, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17210705

ABSTRACT

Association with albumin as a means to improve biodistribution and tumor deposition of a Fab was investigated using AB.Fab4D5, a bifunctional molecule derived from trastuzumab (HERCEPTIN) capable of binding albumin and tumor antigen HER2 (erbB2) simultaneously. AB.Fab4D5 was compared with trastuzumab and a trastuzumab-derived Fab (Fab4D5) for the ability to target tumors overexpressing HER2 in mouse mammary tumor virus/HER2 allograft models. Biodistribution was monitored using intravital microscopy, histology, and integrated single-photon emission computed tomography/computed tomography analysis. Fab4D5 tumor deposition was characterized by rapid but transient appearance in tumor at 2 h with little retention, followed by rapid accumulation in kidney by 6 h. Trastuzumab was slow to accumulate in tumors and slow to clear from normal tissues, although significant tumor deposition was achieved by 24 h. In contrast, AB.Fab4D5 was observed at 2 h in tumor and its presence was sustained beyond 24 h similar to trastuzumab. Intravital microscopy revealed that at peak tumor accumulation, tumor cell staining by AB.Fab4D5 was more uniform than for Fab4D5 or trastuzumab. Similar tumor deposition was achieved for both AB.Fab4D5 and trastuzumab at 48 h (35.9 +/- 1.8% and 38.2 +/- 3.1% injected dose/g); however, AB.Fab4D5 targeted tumors more rapidly and quickly cleared from blood, leading to a lower overall normal tissue exposure. Importantly, unlike Fab4D5, AB.Fab4D5 did not accumulate in kidney, suggesting that association with albumin leads to an altered route of clearance and metabolism. Rapid targeting, excellent tumor deposition and retention, coupled with high tumor to blood ratios may make AB.Fab an exceptional molecule for imaging and cancer therapy.


Subject(s)
Albumins/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Immunoconjugates/pharmacokinetics , Immunoglobulin Fragments/metabolism , Mammary Neoplasms, Experimental/metabolism , Albumins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Female , Humans , Image Processing, Computer-Assisted , Immunoconjugates/chemistry , Immunoglobulin Fragments/chemistry , Mammary Neoplasms, Experimental/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Radionuclide Imaging , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/metabolism , Trastuzumab
8.
Clin Cancer Res ; 12(20 Pt 1): 6144-52, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17062691

ABSTRACT

PURPOSE: Expression of the receptor tyrosine kinase c-Met and its ligand scatter factor/hepatocyte growth factor (SF/HGF) are strongly increased in glioblastomas, where they promote tumor proliferation, migration, invasion, and angiogenesis. We used a novel one-armed anti-c-Met antibody to inhibit glioblastoma growth in vivo. EXPERIMENTAL DESIGN: U87 glioblastoma cells (c-Met and SF/HGF positive) or G55 glioblastoma cells (c-Met positive and SF/HGF negative) were used to generate intracranial orthotopic xenografts in nude mice. The one-armed 5D5 (OA-5D5) anti-c-Met antibody was infused intratumorally using osmotic minipumps. Following treatment, tumor volumes were measured and tumors were analyzed histologically for extracellular matrix (ECM) components and proteases relevant to tumor invasion. Microarray analyses were done to determine the effect of the antibody on invasion-related genes. RESULTS: U87 tumor growth, strongly driven by SF/HGF, was inhibited > 95% with OA-5D5 treatment. In contrast, G55 tumors, which are not SF/HGF driven, did not respond to OA-5D5, suggesting that the antibody can have efficacy in SF/HGF-activated tumors. In OA-5D5-treated U87 tumors, cell proliferation was reduced > 75%, microvessel density was reduced > 90%, and apoptosis was increased > 60%. Furthermore, OA-5D5 treatment decreased tumor cell density > 2-fold, with a consequent increase in ECM deposition and increased immunoreactivity for laminin, fibronectin, and tenascin. Microarray studies showed no increase in these ECM factors, rather down-regulation of urokinase-type plasminogen activator and matrix metalloproteinase 16 in glioblastoma cells treated with OA-5D5. CONCLUSIONS: Local treatment with OA-5D5 can almost completely inhibit intracerebral glioblastoma growth when SF/HGF is driving tumor growth. The mechanisms of tumor inhibition include antiproliferative, antiangiogenic, and proapoptotic effects.


Subject(s)
Antibodies/pharmacology , Cell Division/drug effects , Glioblastoma/pathology , Proto-Oncogene Proteins c-met/immunology , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism , Transplantation, Heterologous
9.
Cancer Res ; 66(1): 283-9, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397241

ABSTRACT

Activating mutations in receptor tyrosine kinases play a critical role in oncogenesis. Despite evidence that Met kinase is deregulated in human cancer, the role of activating mutations in cancers other than renal papillary carcinoma has not been well defined. Here we report the identification of somatic intronic mutations of Met kinase that lead to an alternatively spliced transcript in lung cancer, which encodes a deletion of the juxtamembrane domain resulting in the loss of Cbl E3-ligase binding. The mutant receptor exhibits decreased ubiquitination and delayed down-regulation correlating with elevated, distinct Met expression in primary tumors harboring the deleted receptor. As a consequence, phospho-Met and downstream mitogen-activated protein kinase activation is sustained on ligand stimulation. Cells expressing the Met deletion reveal enhanced ligand-mediated proliferation and significant in vivo tumor growth. A hepatocyte growth factor competitive Met antagonist inhibits receptor activation and proliferation in tumor cells harboring the Met deletion, suggesting the important role played by ligand-dependent Met activation and the potential for anticancer therapy. These results support a critical role for Met in lung cancer and somatic mutation-driven splicing of an oncogene that leads to a different mechanism for tyrosine kinase activation through altered receptor down-regulation in human cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Deletion , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Receptors, Growth Factor/genetics , Alternative Splicing , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Down-Regulation , Enzyme Activation , Exons , Female , Humans , Introns , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/metabolism , Signal Transduction , Ubiquitin/metabolism
10.
Curr Pharm Des ; 10(20): 2525-33, 2004.
Article in English | MEDLINE | ID: mdl-15320761

ABSTRACT

Hepatocyte growth factor (HGF) is a cytokine whose multipotent actions are mediated by c-Met receptor. This review focuses on effects of HGF on myocardial infarction (MI) and heart failure. Circulating concentrations of HGF and myocardial concentrations of HGF and c-Met mRNA and protein are substantially increased following acute MI. HGF has been shown to be cardioprotective towards acute cardiac ischemia-reperfusion injury. Gene transfection of HGF into rat hearts attenuates acute ischemia injury. Administration of HGF protein reduces infarct size and increases cardiac performance in a rat model of acute ischemia/reperfusion. In contrast, acute blockade of endogenous HGF increases infarct size and mortality. These acute effects of HGF appear to be related to angiogenic and anti-apoptotic mechanisms. Recent studies demonstrate that post-MI treatment with HGF gene or protein attenuates chronic cardiac remodeling and dysfunction. In rats, HGF gene transfer following large MI results in preserved cardiac function and geometry in association with angiogenesis and reduced apoptosis, and treatment with recombinant HGF also significantly improves cardiac performance measured 8 weeks after MI. In mice, post-MI HGF gene therapy improves cardiac remodeling and dysfunction through hypertrophy of cardiomyocytes, infarct wall thickening, preservation of vessels, and antifibrosis. In addition, gene transfer of HGF improves cardiac remodeling, angiogenesis and regional myocardial function in the chronic ischemic myocardium of dogs. Together, these preclinical data highlight the significant acute and chronic cardioprotective effects of HGF following ischemic heart failure. Clinical trials are needed to investigate the therapeutic potential of HGF for postinfarction heart failure in humans.


Subject(s)
Heart Failure/drug therapy , Hepatocyte Growth Factor/therapeutic use , Myocardial Infarction/drug therapy , Angiotensin II/physiology , Angiotensin II/therapeutic use , Animals , Disease Models, Animal , Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/physiology , Humans
11.
Cancer Res ; 64(14): 4900-5, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256461

ABSTRACT

Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) is a tumor necrosis factor superfamily member that induces apoptosis through the death receptors DR4 and/or DR5 in various cancer cell types but not in most normal cells. Several lung cancer cell lines express DR4 and DR5 and undergo apoptosis in vitro in response to Apo2L/TRAIL. We investigated the efficacy of recombinant soluble human Apo2L/TRAIL and its interaction with chemotherapy in xenograft models based on human NCI-H460 non-small cell lung carcinoma cells. In vitro, Taxol enhanced caspase activation and apoptosis induction by Apo2L/TRAIL. In vivo, Apo2L/TRAIL or Taxol plus carboplatin chemotherapy partially delayed progression of established subcutaneous tumor xenografts, whereas combined treatment caused tumor regression and a substantially longer growth delay. Apo2L/TRAIL, chemotherapy, or the combination of both inhibited growth of preformed orthotopic lung parenchymal tumors versus control by 60%, 57%, or 97%, respectively (all P < 0.01; n = 8-10). Furthermore, combination treatment improved day-90 survival relative to control (7 of 15 versus 1 of 15; P = 0.0003 by Mantel-Cox) as well as to Apo2L/TRAIL (3 of 14; P = 0.031) or chemotherapy (3 of 15; P = 0.035). These studies provide evidence for in vivo activity of Apo2L/TRAIL against lung tumor xenografts and underscore the potential of this ligand for advancing current lung cancer treatment strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis Regulatory Proteins , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Membrane Glycoproteins/administration & dosage , Mice , Mice, Nude , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 10(7): 2499-511, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15073130

ABSTRACT

PURPOSE: This study examined the effectiveness of early and prolonged mu4D5 (the murine form of trastuzumab/Herceptin) treatment in transgenic mice that overexpress human HER2 (huHER2), under the murine mammary tumor virus promoter, as a model of huHER2-overexpressing breast cancer. EXPERIMENTAL DESIGN: Mice were randomly assigned to one of three treatment groups and received i.p. injections from 17 weeks of age until either 52 weeks of age or morbidity. Fourteen mice received 100 mg/kg mu4D5, 14 mice received 100 mg/kg antiherpes simplex virus glycoprotein D control antibody, and 11 mice received a diluent control. RESULTS: High levels of huHER2 expression were detectable in mammary glands of young virgin founder mice. Mammary adenocarcinomas were frequently found in female founders and progeny at an average age of 28 weeks, with some progressing to metastatic disease. The incidence of mammary tumors was significantly reduced, and tumor growth inhibition was observed in mice receiving mu4D5 compared with control mice. In addition, Harderian gland neoplasms, highly associated with overexpression of huHER2 in this transgenic line, were entirely absent in the mu4D5 treatment group, indicating down-regulation of huHER2 in vivo activity. CONCLUSIONS: Early intervention with mu4D5 was of benefit in our transgenic mice at high risk for developing huHER2-overexpressing breast cancer. This study suggests a potential benefit of early treatment with Herceptin in HER2-positive primary breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Mammary Tumor Virus, Mouse/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Base Sequence , Disease Progression , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/secondary , Mammary Neoplasms, Animal , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Neoplasm Metastasis , Neoplasms, Experimental , Promoter Regions, Genetic , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transgenes , Trastuzumab
13.
Magn Reson Med ; 51(3): 542-51, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15004796

ABSTRACT

Tumor heterogeneity complicates the quantification of a therapeutic response by MRI. To address this issue, a novel approach has been developed that combines MR diffusion imaging with multispectral (MS) analysis to quantify tumor tissue populations. K-means (KM) clustering of the apparent diffusion coefficient (ADC), T2, and proton density (M0) was employed to estimate the volumes of viable tumor tissue, necrosis, and neighboring subcutaneous adipose tissue in a human colorectal tumor xenograft mouse model. In a second set of experiments, the temporal evolution of the MS tissue classes in response to therapeutic intervention Apo2L/TRAIL and CPT-11 was observed. The multiple parameters played complementary roles in identifying the various tissues. The ADC was the dominant parameter for identifying regions of necrosis, whereas T2 identified two necrotic subpopulations, and M0 contributed to the differentiation of viable tumor from subcutaneous adipose tissue. MS viable tumor estimates (mean volume = 275 +/- 147 mm(3)) were highly correlated (r = 0.81, P < 0.01) with histological estimates (117 +/- 51 mm(3)). In the treatment study, MS viable tumor volume (at day 10) was 77 +/- 67 mm(3) for the Apo2L/TRAIL+CPT-11 group, and was significantly reduced relative to the control group (292 +/- 127 mm(3), P < 0.01). This method shows promise as a means of detecting an early therapeutic response in vivo.


Subject(s)
Camptothecin/analogs & derivatives , Colonic Neoplasms/pathology , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Skin Neoplasms/pathology , Adipose Tissue/pathology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis Regulatory Proteins , Camptothecin/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Disease Models, Animal , Female , Humans , Irinotecan , Ligands , Membrane Glycoproteins/therapeutic use , Mice , Mice, Nude , Necrosis , Neoplasm Transplantation , Remission Induction , Skin Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Topoisomerase I Inhibitors , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/therapeutic use , fas Receptor/therapeutic use
14.
Cancer Gene Ther ; 10(11): 840-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14605670

ABSTRACT

Engineered retroviruses are widely used vectors for cancer gene therapy approaches. However, the ability to target cells of therapeutic interest while controlling the expression of the transferred genes would improve both the efficiency and the safety of viral vectors. In this study, we investigated the ability of a retroviral amphotropic envelope displaying single-chain variable-fragment (scFv) directed against the c-Met receptor, to target the entry of recombinant retroviruses to human hepatocarcinoma cells. Four single-chain antibody fragments directed against the c-Met receptor were generated and inserted into the viral envelope protein as an N-terminal fusion. The modified envelopes were incorporated into virus particles and one of the chimeric viruses, 3D6-Env, transduced preferentially human hepatoma cells rather than proliferating human hepatocytes. In another construct, the urokinase cleavage site was inserted between the scFv moiety and the envelope. Chimeric scFv-urokinase-Env viruses transduced hepatoma cells with a similar efficiency to that of the control virus and their infectivity in human hepatocytes remained low. These results indicate that amphotropic retroviruses with engineered envelopes to display scFv directed against the c-Met receptor can efficiently and selectively deliver genes into hepatoma cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Transfer Techniques , Immunoglobulin Variable Region/genetics , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-met/immunology , Antibodies/genetics , Antibodies/immunology , Carcinoma, Hepatocellular/virology , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hepatocytes/physiology , Hepatocytes/virology , Humans , Protein Engineering/methods , Retroviridae/genetics , Retroviridae/pathogenicity , Transduction, Genetic , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Virion
15.
J Biol Chem ; 278(48): 47654-9, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14500721

ABSTRACT

Stanniocalcin 1 (STC1) is a secreted glycoprotein originally described as a hormone involved in calcium and phosphate homeostasis in bony fishes. We recently identified the mammalian homolog of this molecule to be highly up-regulated in an in vitro model of angiogenesis, as well as focally and intensely expressed at sites of pathological angiogenesis (e.g. tumor vasculature). In the present study, we report that STC1 is a selective modulator of hepatocyte growth factor (HGF)-induced endothelial migration and morphogenesis, but not proliferation. STC1 did not inhibit proliferative or migratory responses to vascular endothelial growth factor or basic fibroblast growth factor. The mechanism of STC1 inhibitory effects on HGF-induced endothelial migration seem to occur secondary to receptor activation because STC1 did not inhibit HGF-induced c-met receptor phosphorylation, but did block HGF-induced focal adhesion kinase activation. In the mouse femoral artery ligation model of angiogenesis, STC1 expression closely paralleled that of the endothelial marker CD31, and the peak level of STC1 expression occurred after an increase in HGF expression. We propose that STC1 may play a selective modulatory role in angiogenesis, possibly serving as a "stop signal" or stabilizing factor contributing to the maturation of newly formed blood vessels. HGF is a mesenchyme-derived pleiotropic factor with mitogenic, motogenic, and morphogenic activities on a number of different cell types. HGF effects are mediated through a specific tyrosine kinase, c-met, and aberrant HGF and c-met expression are frequently observed in a variety of tumors. Recent studies have shown HGF to be a potent growth factor implicated in wound healing, tissue regeneration, and angiogenesis.


Subject(s)
Endothelium/metabolism , Glycoproteins/physiology , Hepatocyte Growth Factor/metabolism , Neovascularization, Physiologic , Animals , Antibodies, Monoclonal/chemistry , Cell Division , Cell Movement , Cells, Cultured , Collagen/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Endothelium, Vascular/cytology , Enzyme Activation , Fibroblast Growth Factor 2/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Genetic Vectors , Humans , Laminin/pharmacology , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proteoglycans/pharmacology , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/metabolism , Time Factors , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
16.
Cancer Res ; 63(5): 1020-4, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12615717

ABSTRACT

A central function of the tumor suppressor retinoblastoma (Rb) is its ability to repress E2F transcriptional activity. Many cancers harbor inactivated Rb and consequently deregulated E2F. RXL peptides inhibit E2F recruitment and phosphorylation by CDK2/cyclin A. Here we report that RXL peptides selectively kill tumor cells with deregulated Rb/cyclin D pathways. We extend these observations to tumor models and demonstrate inhibition of tumor growth in SV40 large T transformed Balb/c 3T3 grafts and in HER2 transgenic tumors. Moreover, our observations reveal that RXL peptide-treated tumors undergo apoptosis. Our results indicate that RXL motif-based inhibitors will provide selective antiproliferative agents with in vivo efficacy in tumors with deregulated Rb/cyclin D pathways.


Subject(s)
Antineoplastic Agents/pharmacology , CDC2-CDC28 Kinases , Cyclin A/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3T3 Cells , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/physiology , Binding Sites , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , Cyclin D , Cyclin-Dependent Kinase 2 , Cyclins/physiology , Female , Genes, erbB-2/genetics , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Rats , Retinoblastoma Protein/physiology , Signal Transduction/physiology
17.
J Pharmacol Exp Ther ; 304(2): 654-60, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538818

ABSTRACT

Plasma levels of hepatocyte growth factor (HGF) are increased within hours of cardiac ischemia/reperfusion in rats, and HGF has been shown to be cardioprotective toward acute ischemic injury. Myocardial levels of HGF mRNA and protein are increased for several days after myocardial infarction (MI), however, indicating a possible additional protective effect of HGF toward the progression of MI to heart failure. The purpose of this study was to determine whether HGF administration during the time course of endogenous cardiac HGF induction would lead to long-term improvement in cardiac function in rats with MI. MI was induced by 2-h occlusion of the left coronary artery, followed by reperfusion. HGF was given by intravenous infusion at 0.45 mg/kg/day for 6 days beginning on the day after surgery. Cardiac function and hemodynamic parameters were measured by using indwelling catheters and perivascular flow probes in conscious animals 8 weeks post-MI. Myocardial infarcts were approximately 30% of the left ventricle, and there was no difference in infarct size between the vehicle-treated and HGF-treated groups. Compared with untreated sham-operated rats, vehicle-treated MI animals had significantly lower cardiac index and stroke volume index and higher systemic vascular resistance, indicating heart failure developed. Treatment with HGF caused a significant increase in cardiac index and stroke volume index and a reduction in systemic vascular resistance in rats with MI, restoring these parameters close to those observed in sham-operated control animals. These results provide direct evidence that HGF may be of benefit to cardiovascular function in ischemic cardiomyopathy.


Subject(s)
Heart Failure/drug therapy , Hepatocyte Growth Factor/therapeutic use , Myocardial Infarction/drug therapy , Animals , Drug Administration Schedule , Gene Expression/drug effects , Gene Expression/physiology , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Function Tests/drug effects , Heart Function Tests/methods , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/pharmacology , Male , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
18.
Cancer Res ; 62(19): 5485-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12359757

ABSTRACT

Hormone-independent tumor growth and metastasis are associated with increased mortality in human prostate cancer. In this study, we evaluate a potential role for ligand-mediated activation of HER2 receptor tyrosine kinase in androgen-independent prostate cancers. HER2, HER3, and epidermal growth factor receptor were detected in the androgen-independent cell line 22Rv1. Heregulin stimulation results in receptor phosphorylation and cell proliferation that is inhibited by increasing concentrations of anti-HER2 recombinant humanized monoclonal antibody (rhuMAb) 2C4. Furthermore, inhibition of tumor growth was observed in xenografts derived from 22Rv1 cells when treated with rhuMAb 2C4 in a dose-dependent manner. These studies provide a framework, both in vitro and in vivo, to examine the molecular mechanisms of ligand-driven HER2 activation in androgen-independent tumorigenesis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Prostatic Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Androgens/physiology , Animals , Cell Division/drug effects , Dose-Response Relationship, Immunologic , Female , Humans , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/therapy , Neuregulin-1/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Recombinant Proteins/pharmacology , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Nat Med ; 8(3): 274-81, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11875499

ABSTRACT

The importance of Bax for induction of tumor apoptosis through death receptors remains unclear. Here we show that Bax can be essential for death receptor--mediated apoptosis in cancer cells. Bax-deficient human colon carcinoma cells were resistant to death-receptor ligands, whereas Bax-expressing sister clones were sensitive. Bax was dispensable for apical death-receptor signaling events including caspase-8 activation, but crucial for mitochondrial changes and downstream caspase activation. Treatment of colon tumor cells deficient in DNA mismatch repair with the death-receptor ligand apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selected in vitro or in vivo for refractory subclones with Bax frameshift mutations including deletions at a novel site. Chemotherapeutic agents upregulated expression of the Apo2L/TRAIL receptor DR5 and the Bax homolog Bak in Baxminus sign/minus sign cells, and restored Apo2L/TRAIL sensitivity in vitro and in vivo. Thus, Bax mutation in mismatch repair--deficient tumors can cause resistance to death receptor--targeted therapy, but pre-exposure to chemotherapy rescues tumor sensitivity.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins , BH3 Interacting Domain Death Agonist Protein , Camptothecin/pharmacology , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Enzyme Activation , Etoposide/pharmacology , Fas-Associated Death Domain Protein , Female , Flow Cytometry , Humans , Membrane Glycoproteins/therapeutic use , Mice , Mice, Nude , Mitochondria/metabolism , Mutation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Random Allocation , TNF-Related Apoptosis-Inducing Ligand , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/therapeutic use , bcl-2-Associated X Protein
20.
J Biol Chem ; 277(16): 14329-35, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11832495

ABSTRACT

Novel drug targets can be identified by differential analysis of RNA transcripts isolated from cancer cell lines and tissues. We have extended this approach by analyzing differences in gene expression resulting from the drug treatment of transformed and nontransformed cells. A mouse mammary epithelial cell line (C57MG), which conditionally expresses the Wnt-1 proto-oncogene, was left untreated or treated with retinoic acid in the presence or absence of Wnt-1 expression. The experiment was performed in triplicate, and RNA extracted from the four samples was analyzed by hybridization to over 12,000 unique oligonucleotide probe sets. Reproducible alterations in gene expression that occurred in response to retinoic acid, Wnt-1, or retinoic acid plus Wnt-1 relative to untreated cells were identified. Greater attention was given to genes encoding cell surface antigens that were selectively up-regulated by the combination of Wnt-1 and retinoic acid. These genes included the tumor necrosis factor family 4-1BB ligand, ephrin B1, stra6, autotaxin, and ISLR. Administration of retinoic acid to mice bearing tumors driven by activation of the Wnt-1/beta-catenin pathway resulted in increased expression of stra6 in the tumors but not in normal tissue. In principal, the therapeutic index of antibodies directed against these antigens should be enhanced by co-administration of retinoic acid.


Subject(s)
Antigens, Neoplasm/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Tretinoin/metabolism , Zebrafish Proteins , Animals , Blotting, Northern , Blotting, Western , Cell Line , Cells, Cultured , Humans , Mice , Neoplasm Transplantation , Protein Binding , Proto-Oncogene Mas , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/pharmacology , Tumor Cells, Cultured , Up-Regulation , Wnt Proteins , Wnt1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...