Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Publication year range
1.
J Immunol ; 188(1): 259-69, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22140262

ABSTRACT

Lasting B cell persistence depends on survival signals that are transduced by cell surface receptors. In this study, we describe a novel biological mechanism essential for survival and homeostasis of normal peripheral mature B cells and chronic lymphocytic leukemia cells, regulated by the heparin-binding cytokine, midkine (MK), and its proteoglycan receptor, the receptor-type tyrosine phosphatase ζ (RPTPζ). We demonstrate that MK initiates a signaling cascade leading to B cell survival by binding to RPTPζ. In mice lacking PTPRZ, the proportion and number of the mature B cell population are reduced. Our results emphasize a unique and critical function for MK signaling in the previously described MIF/CD74-induced survival pathway. Stimulation of CD74 with MIF leads to c-Met activation, resulting in elevation of MK expression in both normal mouse splenic B and chronic lymphocytic leukemia cells. Our results indicate that MK and RPTPζ are important regulators of the B cell repertoire. These findings could pave the way toward understanding the mechanisms shaping B cell survival and suggest novel therapeutic strategies based on the blockade of the MK/RPTPζ-dependent survival pathway.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , B-Lymphocytes/immunology , Cytokines/immunology , Histocompatibility Antigens Class II/immunology , Membrane Glycoproteins/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/immunology , Receptors, Growth Factor/immunology , Signal Transduction/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Midkine , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Signal Transduction/genetics , Spleen/immunology , Spleen/metabolism
2.
Nat Med ; 6(10): 1160-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017149

ABSTRACT

The breaking of immune tolerance against autologous angiogenic endothelial cells should be a useful approach for cancer therapy. Here we show that immunotherapy of tumors using fixed xenogeneic whole endothelial cells as a vaccine was effective in affording protection from tumor growth, inducing regression of established tumors and prolonging survival of tumor-bearing mice. Furthermore, autoreactive immunity targeting to microvessels in solid tumors was induced and was probably responsible for the anti-tumor activity. These observations may provide a new vaccine strategy for cancer therapy through the induction of an autoimmune response against the tumor endothelium in a cross-reaction.


Subject(s)
Cancer Vaccines/pharmacology , Endothelium/cytology , Endothelium/immunology , Immunotherapy/methods , Neoplasms, Experimental/therapy , Amino Acid Sequence , Animals , Antigens, CD/immunology , Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Cattle , Cells, Cultured , Cross Reactions , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Humans , Integrin alphaV , Mice , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Peptides/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor
3.
Nat Med ; 8(8): 831-40, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12091877

ABSTRACT

The therapeutic potential of placental growth factor (PlGF) and its receptor Flt1 in angiogenesis is poorly understood. Here, we report that PlGF stimulated angiogenesis and collateral growth in ischemic heart and limb with at least a comparable efficiency to vascular endothelial growth factor (VEGF). An antibody against Flt1 suppressed neovascularization in tumors and ischemic retina, and angiogenesis and inflammatory joint destruction in autoimmune arthritis. Anti-Flt1 also reduced atherosclerotic plaque growth and vulnerability, but the atheroprotective effect was not attributable to reduced plaque neovascularization. Inhibition of VEGF receptor Flk1 did not affect arthritis or atherosclerosis, indicating that inhibition of Flk1-driven angiogenesis alone was not sufficient to halt disease progression. The anti-inflammatory effects of anti-Flt1 were attributable to reduced mobilization of bone marrow-derived myeloid progenitors into the peripheral blood; impaired infiltration of Flt1-expressing leukocytes in inflamed tissues; and defective activation of myeloid cells. Thus, PlGF and Flt1 constitute potential candidates for therapeutic modulation of angiogenesis and inflammation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arteriosclerosis/drug therapy , Arthritis, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic , Pregnancy Proteins/pharmacology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antibodies, Monoclonal/pharmacology , Arteriosclerosis/immunology , Arteriosclerosis/pathology , Arthritis, Experimental/pathology , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/pathology , Disease Models, Animal , Endothelial Growth Factors/pharmacology , Female , Hematopoietic Stem Cells/drug effects , Hindlimb/blood supply , Humans , Ischemia/drug therapy , Ischemia/pathology , Joints/pathology , Lymphokines/pharmacology , Mice , Mice, Nude , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Neovascularization, Physiologic/drug effects , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/physiology , Proto-Oncogene Proteins/metabolism , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/immunology , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
4.
ScientificWorldJournal ; 11: 2491-505, 2011.
Article in English | MEDLINE | ID: mdl-22235180

ABSTRACT

The 13 kDa heparin-binding growth factor midkine (MK) was originally identified as a molecule involved in the orchestration of embryonic development. Recent studies provided evidence for a new role of MK in acute and chronic inflammatory processes. Accordingly, several inflammatory diseases including nephritis, arthritis, atherosclerosis, colitis, and autoimmune encephalitis have been shown to be alleviated in the absence of MK in animal models. Reduced leukocyte recruitment to the sites of inflammation was found to be one important mechanism attenuating chronic inflammation when MK was absent. Furthermore, MK was found to modulate expression of proinflammatory cytokines and the expansion of regulatory T-cells. Here, we review the current understanding of the role of MK in different inflammatory disorders and summarize the knowledge of MK biology.


Subject(s)
Cytokines/immunology , Inflammation/immunology , Nerve Growth Factors/immunology , Amino Acid Sequence , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Carrier Proteins/chemistry , Cytokines/chemistry , Cytokines/genetics , Humans , Inflammation/pathology , Inflammation Mediators/immunology , Membrane Glycoproteins/immunology , Midkine , Molecular Sequence Data , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Protein Structure, Secondary , Receptors, Growth Factor/immunology , Sequence Homology, Amino Acid , Signal Transduction , T-Lymphocytes, Regulatory/immunology
5.
Int Immunopharmacol ; 94: 107473, 2021 May.
Article in English | MEDLINE | ID: mdl-33611062

ABSTRACT

The immunogenicity of the breast tumor microenvironment is clinically heterogeneous. The insight into the role of tumor-infiltrating lymphocytes (TILs) might serve as a biomarker to predict a survival benefit and enable optimal patient selection for immunotherapy. In this study, we aimed at characterizing the breast cancer immune subtypes linked to CD8 T cells and associating with the patient characteristics and clinical outcomes. We analyzed the immune gene signatures of human breast cancer using The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) database profiling of 1092 breast tumor patients. We performed hierarchical clustering to the immune gene expression by applying hormone receptor status including triple negative breast cancer to categorize 66 immune-related genes in breast tumors. The separation was characterized by dividing the breast tumors into two major immune subtypes: predominant immune (PI) subtype and low immune subtype (LI). Our results showed that both PI and LI subtypes can be observed in the different hormone receptor phenotypes of breast tumors, and PI subtype accounted for 16% and LI subtype 20% in the breast tumor patients. The estimated odds for LI subtype breast tumors were significantly higher than PI subtype breast tumors in primary tumor stages. Our data demonstrated that the PI subtype breast tumors have significantly improved survival compared with LI subtype. Our findings provide a novel perspective of breast cancer immune subtypes linked to CD8 T cells. The immune subtypes will be a valuable resource for future research to identify clinically relevant biomarkers for precision immunotherapy.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Neoplasm Staging , Phenotype , Prognosis , Receptors, Cell Surface/immunology , Receptors, Growth Factor/immunology , T-Lymphocytes/immunology
6.
J Exp Med ; 195(12): 1575-84, 2002 Jun 17.
Article in English | MEDLINE | ID: mdl-12070285

ABSTRACT

The vascular endothelial growth factor (VEGF) receptor fetal liver kinase 1 (flk1; VEGFR-2, KDR) is an endothelial cell-specific receptor tyrosine kinase that mediates physiological and pathological angiogenesis. We hypothesized that an active immunotherapy approach targeting flk1 may inhibit tumor angiogenesis and metastasis. To test this hypothesis, we first evaluated whether immune responses to flk1 could be elicited in mice by immunization with dendritic cells pulsed with a soluble flk1 protein (DC-flk1). This immunization generated flk1-specific neutralizing antibody and CD8+ cytotoxic T cell responses, breaking tolerance to self-flk1 antigen. Tumor-induced angiogenesis was suppressed in immunized mice as measured in an alginate bead assay. Development of pulmonary metastases was strongly inhibited in DC-flk1-immunized mice challenged with B16 melanoma or Lewis lung carcinoma cells. DC-flk1 immunization also significantly prolonged the survival of mice challenged with Lewis lung tumors. Thus, an active immunization strategy that targets an angiogenesis-related antigen on endothelium can inhibit angiogenesis and may be a useful approach for treating angiogenesis-related diseases.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Melanoma, Experimental/blood supply , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Animals , Carcinoma, Lewis Lung/pathology , Female , Immunotherapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Receptors, Vascular Endothelial Growth Factor
7.
Invest Clin ; 51(3): 369-80, 2010 Sep.
Article in Spanish | MEDLINE | ID: mdl-21305773

ABSTRACT

The product of the proto-oncogene C-MET (the c-Met receptor) and its ligand, hepatocyte growth factor (HGF), have been implicated in the progression of gastric cancer. The aim of this study was to analyze the expression of c-Met receptor, HGF and proliferating cell nuclear antigen (PCNA) by the immunohistochemistry method of labeled streptavidin-biotin, as well as survival, and they were correlated with anatomopathological factors in stomach specimens of 40 patients, who underwent gastrectomy for gastric cancer in the Department of General Surgery, Hospital Central Universitario "Antonio María Pineda" in Barquisimeto, Venezuela, in 2001-2004. High expression of c-Met receptor and PCNA was observed in patients with advanced stages of gastric cancer (III and IV) compared with early stages (I and II) (p<0.01). There was also overexpression of the c-Met receptor in histologic variables with low degree of differentiation, deeper tumor invasion into the submucosa, liver metastases and it is reported a lower survival rate in patients with increased receptor expression (+++ and ++++) when compared with patients with the lowest expression (+ and ++) (p<0.01). The expression of HGF was constant in both, advanced and early groups. The c-Met receptor is associated with proliferation and cell migration in Venezuelan patients with gastric cancer and could be used as a prognostic factor in this pathology.


Subject(s)
Proto-Oncogene Proteins c-met/biosynthesis , Receptors, Growth Factor/biosynthesis , Stomach Neoplasms/etiology , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Male , Middle Aged , Prospective Studies , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/immunology , Receptors, Growth Factor/immunology , Stomach Neoplasms/immunology
8.
Clin Cancer Res ; 14(12): 3814-22, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559601

ABSTRACT

PURPOSE: The association hepatocyte growth factor receptor (Met) tyrosine kinase with prognosis and survival in colon cancer is unclear, due in part to the limitation of detection methods used. In particular, conventional chromagenic immunohistochemistry (IHC) has several limitations including the inability to separate compartmental measurements. Measurement of membrane, cytoplasm, and nuclear levels of Met could offer a superior approach to traditional IHC. EXPERIMENTAL DESIGN: Fluorescent-based IHC for Met was done in 583 colon cancer patients in a tissue microarray format. Using curvature and intensity-based image analysis, the membrane, nuclear, and cytoplasm were segmented. Probability distributions of Met within each compartment were determined, and an automated scoring algorithm was generated. An optimal score cutpoint was calculated using 500-fold crossvalidation of a training and test data set. For comparison with conventional IHC, a second array from the same tissue microarray block was 3,3'-diaminobenzidine immunostained for Met. RESULTS: In crossvalidated and univariate Cox analysis, the membrane relative to cytoplasm Met score was a significant predictor of survival in stage I (hazard ratio, 0.16; P = 0.006) and in stage II patients (hazard ratio, 0.34; P < or = 0.0005). Similar results were found with multivariate analysis. Met in the membrane alone was not a significant predictor of outcome in all patients or within stage. In the 3,3'-diaminobenzidine-stained array, no associations were found with Met expression and survival. CONCLUSIONS: These data indicate that the relative subcellular distribution of Met, as measured by novel automated image analysis, may be a valuable biomarker for estimating colon cancer prognosis.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Cell Membrane/metabolism , Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Cytoplasm/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Growth Factor/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Cohort Studies , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Follow-Up Studies , HeLa Cells , Humans , Middle Aged , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/immunology , Survival Analysis , Tissue Array Analysis , Tissue Distribution
9.
World J Gastroenterol ; 14(1): 1-14, 2008 Jan 07.
Article in English | MEDLINE | ID: mdl-18176955

ABSTRACT

Growth factors and their corresponding receptors are commonly overexpressed and/or dysregulated in many cancers including hepatocellular cancer (HCC). Clinical trials indicate that growth factor receptors and their related signalling pathways play important roles in HCC cancer etiology and progression, thus providing rational targets for innovative cancer therapies. A number of strategies including monoclonal antibodies, tyrosine kinase inhibitors ("small molecule inhibitors") and antisense oligonucleotides have already been evaluated for their potency to inhibit the activity and downstream signalling cascades of these receptors in HCC. First clinical trials have also shown that multi-kinase inhibition is an effective novel treatment strategy in HCC. In this respect sorafenib, an inhibitor of Raf-, VEGF- and PDGF-signalling, is the first multi-kinase inhibitor that has been approved by the FDA for the treatment of advanced HCC. Moreover, the serine-threonine kinase of mammalian target of rapamycin (mTOR) upon which the signalling of several growth factor receptors converge plays a central role in cancer cell proliferation. mTOR inhibition of HCC is currently also being studied in preclinical trials. As HCCs represent hypervascularized neoplasms, inhibition of tumour vessel formation via interfering with the VEGF/VEGFR system is another promising approach in HCC treatment. This review will summarize the current status of the various growth factor receptor-based treatment strategies and in view of the multitude of novel targeted approaches, the rationale for combination therapies for advanced HCC treatment will also be taken into account.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Receptors, Growth Factor/metabolism , Signal Transduction/physiology , Animals , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Receptors, Growth Factor/immunology
10.
J Clin Invest ; 105(8): R15-24, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10772661

ABSTRACT

Various conventional chemotherapeutic drugs can block angiogenesis or even kill activated, dividing endothelial cells. Such effects may contribute to the antitumor efficacy of chemotherapy in vivo and may delay or prevent the acquisition of drug-resistance by cancer cells. We have implemented a treatment regimen that augments the potential antivascular effects of chemotherapy, that is devoid of obvious toxic side effects, and that obstructs the development of drug resistance by tumor cells. Xenografts of 2 independent neuroblastoma cell lines were subjected to either continuous treatment with low doses of vinblastine, a monoclonal neutralizing antibody (DC101) targeting the flk-1/KDR (type 2) receptor for VEGF, or both agents together. The rationale for this combination was that any antivascular effects of the low-dose chemotherapy would be selectively enhanced in cells of newly formed vessels when survival signals mediated by VEGF are blocked. Both DC101 and low-dose vinblastine treatment individually resulted in significant but transient xenograft regression, diminished tumor vascularity, and direct inhibition of angiogenesis. Remarkably, the combination therapy resulted in full and sustained regressions of large established tumors, without an ensuing increase in host toxicity or any signs of acquired drug resistance during the course of treatment, which lasted for >6 months. This article may have been published online in advance of the print edition. The date of publication is available from the JCI website, http://www.jci.org.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Neuroblastoma/drug therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Vinblastine/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Cells, Cultured , Combined Modality Therapy , Dose-Response Relationship, Drug , Fluorescence , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic , Neuroblastoma/blood supply , Neuroblastoma/pathology , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vinblastine/adverse effects
11.
Arch Immunol Ther Exp (Warsz) ; 54(2): 85-101, 2006.
Article in English | MEDLINE | ID: mdl-16648969

ABSTRACT

Gene amplification, over-expression, and mutation of growth factors, or the receptors themselves, causes increased signaling through receptor kinases, which has been implicated in many human cancers and is associated with poor prognosis. Tumor growth has been shown to be decreased by interrupting this process of extensive growth factor-mediated signaling by directly targeting either the surface receptor or the ligand and thereby preventing cell survival and promoting apoptosis. Monoclonal antibodies have long been eyed as a potential new class of therapeutics targeting cancer and other diseases. Antibody-based therapy initially entered clinical practice when trastuzumab/Herceptin became the first clinically approved drug against an oncogene product as a well-established blocking reagent for tumors with hyperactivity of epidermal growth factor signaling pathways. In the first part of this review we explain basic terms related to the development of antibody-based drugs, give a brief historic perspective of the field, and also touch on topics such as the "humanization of antibodie" or creation of hybrid antibodies. The second part of the review gives an overview of the clinical usage of bispecific antibodies and antibodies "armed" with cytotoxic agents or enzymes. Further within this section, cancer-specific, site-specific, or signaling pathway-specific therapies are discussed in detail. Among other antibody-based therapeutic products, we discuss: Avastin (bevacizumab), CG76030, Theragyn (pemtumomab), daclizumab (Zenapax), TriAb, MDX-210, Herceptin (trastuzumab), panitumumab (ABX-EGF), mastuzimab (EMD-72000), Erbitux (certuximab, IMC225), Panorex (edrecolomab), STI571, CeaVac, Campath (alemtuizumab), Mylotarg (gemtuzumab, ozogamicin), and many others. The end of the review deliberates upon potential problems associated with cancer immunotherapy.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Design , Neoplasms/immunology , Antigens, Neoplasm/immunology , Humans , Neoplasms/therapy , Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/immunology , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction
12.
J Natl Cancer Inst ; 94(11): 819-25, 2002 Jun 05.
Article in English | MEDLINE | ID: mdl-12048269

ABSTRACT

BACKGROUND: Vascular endothelial growth factor C (VEGF-C) stimulates tumor lymphangiogenesis (i.e., formation of lymphatic vessels) and metastasis to regional lymph nodes by interacting with VEGF receptor 3 (VEGFR-3). We sought to determine whether inhibiting VEGFR-3 signaling, and thus tumor lymphangiogenesis, would inhibit tumor metastasis. METHODS: We used the highly metastatic human lung cancer cell line NCI-H460-LNM35 (LNM35) and its parental line NCI-H460-N15 (N15) with low metastatic capacity. We inserted genes by transfection and established a stable N15 cell line secreting VEGF-C and a LNM35 cell line secreting the soluble fusion protein VEGF receptor 3-immunoglobulin (VEGFR-3-Ig, which binds VEGF-C and inhibits VEGFR-3 signaling). Control lines were transfected with mock vectors. Tumor cells were implanted subcutaneously into severe combined immunodeficient mice (n = 6 in each group), and tumors and metastases were examined 6 weeks later. In another approach, recombinant adenoviruses expressing VEGFR-3-Ig (AdR3-Ig) or beta-galactosidase (AdLacZ) were injected intravenously into LNM35 tumor-bearing mice (n = 14 and 7, respectively). RESULTS: LNM35 cells expressed higher levels of VEGF-C RNA and protein than did N15 cells. Xenograft mock vector-transfected LNM35 tumors showed more intratumoral lymphatic vessels (15.3 vessels per grid; 95% confidence interval [CI] = 13.3 to 17.4) and more metastases in draining lymph nodes (12 of 12) than VEGFR-3-Ig-transfected LNM35 tumors (4.1 vessels per grid; 95% CI = 3.4 to 4.7; P<.001, two-sided t test; and four lymph nodes with metastases of 12 lymph nodes examined). Lymph node metastasis was also inhibited in AdR3-Ig-treated mice (AdR3-Ig = 0 of 28 lymph nodes; AdLacZ = 11 of 14 lymph nodes). However, metastasis to the lungs occurred in all mice, suggesting that LNM35 cells can also spread via other mechanisms. N15 tumors overexpressing VEGF-C contained more lymphatic vessels than vector-transfected tumors but did not have increased metastatic ability. CONCLUSIONS: Lymph node metastasis appears to be regulated by additional factors besides VEGF-C. Inhibition of VEGFR-3 signaling can suppress tumor lymphangiogenesis and metastasis to regional lymph nodes but not to lungs.


Subject(s)
Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Lymphatic Metastasis/prevention & control , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Animals , Antibodies/genetics , Antibodies/immunology , Blotting, Western , Cell Division , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Lymphatic Metastasis/pathology , Mice , Mice, SCID , Neoplasm Transplantation , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Signal Transduction , Time Factors , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor D , Vascular Endothelial Growth Factor Receptor-3
13.
Cancer Res ; 58(9): 1952-9, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581838

ABSTRACT

Vascular endothelial growth factor (VEGF) is an angiogenic growth factor that is a primary stimulant of the vascularization of solid tumors. VEGF production is induced by oncogenic gene mutations in the tumor cells and by hypoxic conditions inside the tumor mass. Hypoxia and the locally increased concentration of VEGF lead to an up-regulation of VEGF receptor expression on tumor endothelial cells. Therefore, in the tumor microenvironment, there is an up-regulation of both VEGF and its receptor, leading to a high concentration of occupied receptor on tumor vascular endothelium. The VEGF:receptor complex presents an attractive target for the specific delivery of drugs or other effectors to tumor endothelium. In the present study, several hybridomas that secrete monoclonal antibodies against the VEGF:receptor (Flk-1) complex or against VEGF itself have been raised. Three of the antibodies (3E7, GV39M, and 11B5) bind with high affinity to the VEGF:Flk-1 complex in ELISA and to tumor endothelium in frozen sections of human tumors, rodent tumors, and human tumor xenografts. 3E7 and GV39M localize selectively to tumor endothelium after i.v. injection into mice bearing human tumor xenografts. Additionally, one antibody (2C3) was raised that blocks the interaction between VEGF and KDR/Flk-1. 2C3 inhibits VEGF-mediated growth of endothelial cells in vitro and localizes strongly to connective tissue in tumors after injection into mice bearing human tumor xenografts. These findings suggest that 3E7, GV39M, and 2C3 are candidates for targeting and imaging the vasculature or connective tissue of tumors.


Subject(s)
Antibodies, Monoclonal/analysis , Biomarkers, Tumor/analysis , Endothelial Growth Factors/immunology , Endothelium, Vascular/immunology , Lymphokines/immunology , Neoplasms, Experimental/blood supply , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Animals , Antibodies, Blocking , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Blotting, Western , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Guinea Pigs , Humans , Immunoenzyme Techniques , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Receptors, Vascular Endothelial Growth Factor , Severe Combined Immunodeficiency/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Cancer Res ; 61(18): 6624-8, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559524

ABSTRACT

Using an orthotopic intracerebral model, we investigated whether systemic treatment with DC101, a monoclonal antibody against vascular endothelial growth factor receptor (VEGFR)-2, could inhibit angiogenesis and the growth of human glioblastoma cells in severe combined immunodeficient mice. Intraperitoneal treatment with DC101, control IgG, or PBS was initiated either on day 0 or, in another series, on day 6 after tumor cell implantation, and animals were killed approximately 2 weeks after tumor cell injection. Tumor volumes in animals treated with DC101 were reduced by 59 and 81% compared with IgG and PBS controls, respectively (P < 0.001), when treatment was initiated immediately, and similar results were obtained when treatment started on day 6. Microvessel density in tumors of DC101-treated animals was reduced by at least 40% compared with animals treated with control IgG or PBS (P < 0.01). We observed a reduction in tumor cell proliferation and an increase in apoptosis in DC101-treated animals (P < 0.001). However, in mice treated with DC101, we also noticed a striking increase in the number and total area of small satellite tumors clustered around, but distinct from, the primary. These satellites usually contained central vessel cores, and tumor cells often had migrated over long distances along the host vasculature to eventually reach the surface and spread leptomeningeally. We conclude that systemic antagonization of VEGFR-2 can inhibit glioblastoma neovascularization and growth but can lead to increased cooption of preexistent cerebral blood vessels. Therefore, a combination of different treatment modalities which also include anti-invasive therapy may be needed for an effective therapy against glioblastoma, and the use of an antibody against VEGFR-2 may be one effective component.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/prevention & control , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Division/drug effects , Cell Division/physiology , Female , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mice , Mice, Nude , Mice, SCID , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Xenograft Model Antitumor Assays
15.
Cancer Res ; 62(10): 2731-5, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019144

ABSTRACT

A number of recent preclinical studies have sparked interest in the concept of exploiting conventional chemotherapeutic drugs as antiangiogenics. Such antiangiogenic activity is achieved or optimized by metronomic-dosing protocols in which the drug is given at comparatively low doses using a frequent schedule of administration (e.g., once to three times per week) with no breaks, particularly when combined with an endothelial cell-specific antiangiogenic drug. The use of p.o. chemotherapeutic drugs is particularly suitable for this type of treatment strategy. We tested one such drug, cyclophosphamide (CTX), in a protocol wherein the drug was administered to mice at low doses, of approximately 10-40 mg/kg on a daily basis through the drinking water. CTX is typically given p.o. to patients, but it has almost always been injected when treating preclinical mouse tumor models. We found p.o. CTX to be a safe and convenient treatment with significant antitumor efficacy. Growth delays were observed for human orthotopic breast or ectopic colon cancer xenografts in nude or SCID mice. Established PC3 human prostate tumor xenografts could be induced to almost fully regress, remaining virtually nonpalpable for > or =2 months of continuous therapy, after which tumors began to grow progressively. These re-emergent tumors were not found to be drug resistant when tested in new hosts, using the same treatment protocol. Regression of spontaneously arising, late-stage pancreatic islet cell carcinomas in Rip Tag transgenic mice was also observed. The effects of continuous p.o. CTX treatment were enhanced significantly in an orthotopic, metastatic breast cancer xenograft model when used in combination with an antivascular endothelial growth factor receptor-2 blocking antibody. Maximum tolerated dose levels established for other mouse strains proved highly toxic to SCID mice, whereas daily p.o. low-dose regimens of CTX were well tolerated. Taken together, the results demonstrate the feasibility of delivering CTX in a p.o. metronomic chemotherapy regimen, which proved safe, reasonably efficacious, and potentially applicable to chronic treatment. Such a regimen may be particularly well suited for integration with antiangiogenic drugs.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Breast Neoplasms/drug therapy , Cyclophosphamide/administration & dosage , Administration, Oral , Animals , Antibodies/administration & dosage , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Division/drug effects , Dose-Response Relationship, Drug , Drinking , Female , HT29 Cells/drug effects , Humans , Male , Mice , Mice, Nude , Mice, SCID , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Cancer Res ; 60(16): 4556-60, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969807

ABSTRACT

Tumor oxygenation is critical for tumor survival as well as for response to therapy, e.g., radiation therapy. Hormone ablation therapy in certain hormone-dependent tumors and antiangiogenic therapy lead to vessel regression and have also shown beneficial effects when combined with radiation therapy. These findings are counterintuitive because vessel regression should reduce oxygen tension (pO2) in tumors, decreasing the effectiveness of radiotherapy. Here we report on the dynamics of pO2 and oxygen consumption in a hormone-dependent tumor following hormone ablation and during treatment with an anti-VEGFR-2 monoclonal antibody (mAb) or a combination of doxorubicin and cyclophosphamide; the latter combination is not known to cause vessel regression at doses used clinically. Androgen-dependent male mouse mammary carcinoma (Shionogi) was implanted into transparent dorsal skin-fold chambers in male severe combined immunodeficient mice. Thirteen days after the tumors were implanted, mice were treated with antiangiogenic therapy (anti-VEGFR-2 mAb, 1.4 mg/30 g body weight), hormone ablation by castration, or doxorubicin (6.5 mg/kg every 7 days) and cyclophosphamide (100 mg/kg every 7 days). A non-invasive in vivo method was used to measure pO2 profiles and to calculate oxygen consumption rates (Q(O2)) in tumors. Tumors treated with anti-VEGFR-2 mAb exhibited vessel regression and became hypoxic. Initial vessel regression was followed by a "second wave" of angiogenesis and increases in both pO2 and Q(O2). Hormone ablation led to tumor regression followed by an increase in pO2 coincident with regrowth. Chemotherapy led to tumor growth arrest characterized by constant Q(O2) and elevated pO2. The increased pO2 during anti-VEGFR-2 mAb and hormone ablation therapy may explain the observed beneficial effects of combining antiangiogenic or hormone therapies with radiation treatment. Thus, understanding the microenvironmental dynamics is critical for optimal scheduling of these treatment modalities.


Subject(s)
Androgens/physiology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , Orchiectomy , Oxygen/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Luminescent Measurements , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/therapy , Mice , Mice, SCID , Microscopy, Fluorescence/methods , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/blood supply , Neoplasms, Hormone-Dependent/therapy , Oxygen/blood , Oxygen Consumption , Partial Pressure , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor
17.
Cancer Res ; 61(1): 39-44, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196192

ABSTRACT

Antiangiogenic therapy can enhance radiation-induced tumor growth inhibition. However, the effects of combined antiangiogenic and radiation therapy on long-term tumor control and normal tissue response have not been reported. We treated mice bearing two different human tumor xenografts with anti-vascular endothelial growth factor receptor-2 antibody (DC101) and five dose fractions of local radiation and followed them for at least 6 months. DC101 significantly decreased the dose of radiation necessary to control 50% of tumors locally. The decrease was 1.7- and 1.3-fold for the moderately radiosensitive small cell lung carcinoma 54A and the highly radioresistant glioblastoma multiforme U87, respectively. In contrast to tumors, no increase in skin radiation reaction by the antibody was detected. Surprisingly, 44% of mice bearing 54A tumor developed clear ascites after DC101 treatment at its highest dose; this was fatal to 20% of mice. This adverse effect was seen only in mice that received whole-body irradiation 1 day before tumor implantation. The encouraging results on two human tumor xenografts suggest that vascular endothelial growth factor receptor-2 blockade merits further investigation to assess its potential as an enhancer of radiation therapy in the clinic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Carcinoma, Small Cell/therapy , Glioblastoma/therapy , Lung Neoplasms/therapy , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/immunology , Angiogenesis Inhibitors/toxicity , Animals , Antibodies, Monoclonal/toxicity , Body Weight/immunology , Body Weight/radiation effects , Carcinoma, Small Cell/blood supply , Carcinoma, Small Cell/radiotherapy , Cell Division/immunology , Cell Division/radiation effects , Combined Modality Therapy , Dose Fractionation, Radiation , Dose-Response Relationship, Immunologic , Dose-Response Relationship, Radiation , Glioblastoma/blood supply , Glioblastoma/radiotherapy , Humans , Intestinal Diseases/etiology , Intestinal Diseases/immunology , Lung Neoplasms/blood supply , Lung Neoplasms/radiotherapy , Male , Mice , Mice, Nude , Oxygen/metabolism , Radiation Tolerance/immunology , Radiation Tolerance/physiology , Receptors, Vascular Endothelial Growth Factor , Skin/radiation effects , Whole-Body Irradiation/adverse effects , Xenograft Model Antitumor Assays
18.
Cancer Res ; 61(9): 3653-9, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11325835

ABSTRACT

Vascular endothelial growth factor (VEGF) is produced by cancer cells in response to hypoxia and is the primary stimulant of vascularization in solid tumors. Endothelial cells lining the blood vessels of these tumors have a high concentration of receptor-bound VEGF on their surface, providing a target for antibody- directed cancer therapy. To obtain a cloned antibody to this target when bound to its receptor on tumor endothelium, we used phage display technology to create a single-chain Fv (sFv) antibody library from mice immunized with the 165-amino acid isoform of human VEGF-A. We selected, purified, and characterized LL4, an anti-VEGF sFv that was shown to react with receptor-bound VEGF. LL4 bound selectively to blood vessel endothelium, as shown by immunohistochemistry on tissue sections of human tumors. Furthermore, using autoradiography and grain counting of histological sections, systemically administered LL4 was shown to localize selectively to the endothelial lining of tumor blood vessels in human colorectal carcinoma xenografts in vivo. This study demonstrates the feasibility of targeting tumor vasculature using recombinant antibodies to the VEGF:receptor complex.


Subject(s)
Colorectal Neoplasms/blood supply , Endothelial Growth Factors/immunology , Immunization, Passive/methods , Immunoglobulin Variable Region/immunology , Lymphokines/immunology , Neovascularization, Pathologic/therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Endothelial Growth Factors/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Immunohistochemistry , Lymphokines/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Peptide Library , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
19.
Cancer Res ; 61(19): 7002-8, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585724

ABSTRACT

Vascular endothelial growth factor (VEGF) binds to and mediates its activity mainly through two tyrosine kinase receptors, VEGF receptor 1 [or fms-like tyrosine kinase receptor (Flt-1)] and VEGF receptor 2 [or kinase insert domain-containing receptor (KDR)]. Numerous studies have shown that overexpression of VEGF and its receptor plays an important role in tumor-associated angiogenesis and hence in both tumor growth and metastasis. We demonstrated previously that antagonistic antibodies to KDR specifically inhibited VEGF-stimulated receptor activation, cell migration, and endothelial cell mitogenesis. Here we constructed a recombinant bifunctional diabody that is capable of blocking both Flt-1 and KDR from binding to their ligands, including VEGF and placenta growth factor (PlGF). The diabody was expressed in Escherichia coli and purified by single-step affinity chromatography. The diabody retained the capacity to bind both KDR and Flt-1 and effectively blocked interaction between KDR and VEGF, Flt-1 and VEGF, and Flt-1 and PlGF. Furthermore, the diabody is a stronger inhibitor than its parent antibodies to VEGF-stimulated mitogenesis of human endothelial cells, as well as both VEGF- and PlGF-induced migration of human leukemia cells. Taken together, our results suggest that dual receptor blockade with the bifunctional diabody may prove to be a more efficient approach in inhibiting VEGF-stimulated angiogenesis.


Subject(s)
Antibodies, Bispecific/immunology , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , 3T3 Cells , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Cell Division/drug effects , Cell Movement/drug effects , Cloning, Molecular , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , HL-60 Cells/cytology , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Lymphokines/metabolism , Lymphokines/pharmacology , Mice , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
20.
Cancer Res ; 62(7): 2034-42, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11929822

ABSTRACT

Vascular endothelial growth factor (VEGF) and VEGF receptor 2 [fetal liver kinase 1 (Flk-1)/kinase insert domain-containing receptor] have been shown to play a major role in tumor angiogenesis. In this study, we investigated whether anti-Flk-1 monoclonal antibody DC101 could therapeutically inhibit growth and angiogenesis of human soft tissue sarcoma, and we explored its capacity to enhance the tumoricidal effects of doxorubicin. Treatment of well-established leiomyosarcoma SKLMS-1 and rhabdomyosarcoma RD xenografts in severe combined immunodeficient mice with DC101 resulted in significant antitumor activity. In a parallel study, we compared tumor inhibition with continuous low-dose "antiangiogenic" schedule versus once-every-2-weeks high-dose standard schedule of doxorubicin. We found that continuous low-dose treatment inhibited the tumor growth of RD xenografts about 46.5% of that with standard-schedule treatment, but that continuous low-dose treatment did not inhibit the tumor growth of SKLMS-1 xenografts. Notably, combined DC101 and continuous low-dose doxorubicin resulted in more effective growth inhibition of SKLMS-1 and RD xenografts than has been observed with any agent alone in a long-term s.c. tumor xenograft model. The combination therapy was associated with no additional toxicity to the host animal compared with low-dose doxorubicin alone. Histological examination of xenografts showed significantly reduced microvessel counts in the tumors given combined therapy compared with the tumors given either agent alone. These results are consistent with an enhanced inhibition of angiogenesis in vivo by combined DC101 and doxorubicin using Matrigel plug assay. Additionally, DC101 plus doxorubicin directly exerted enhanced inhibitory effects on endothelial cell migration, proliferation, and tube-like formation in vitro. Furthermore, the combination induced an enhanced apoptosis of endothelial cells that was associated with an increase of capase-3 activity. Thus, the inhibition of angiogenesis and induction of endothelial cell apoptosis are likely important mechanisms for the antitumor activity of combined DC101 and doxorubicin. Collectively, our data suggested that anti-VEGF receptor 2 in combination with continuous low-dose doxorubicin may provide a new therapeutic approach for human soft tissue sarcoma in the clinic.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Doxorubicin/pharmacology , Endothelium, Vascular/drug effects , Leiomyosarcoma/therapy , Neovascularization, Pathologic/therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Rhabdomyosarcoma/therapy , Soft Tissue Neoplasms/therapy , Animals , Combined Modality Therapy , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Synergism , Endothelium, Vascular/pathology , Female , Humans , Leiomyosarcoma/blood supply , Leiomyosarcoma/drug therapy , Leiomyosarcoma/pathology , Mice , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Receptors, Vascular Endothelial Growth Factor , Rhabdomyosarcoma/blood supply , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Soft Tissue Neoplasms/blood supply , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL