Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 22(1): 89-101, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36343381

ABSTRACT

4-1BB (CD137) is an activation-induced costimulatory receptor that regulates immune responses of activated CD8 T and natural killer cells, by enhancing proliferation, survival, cytolytic activity, and IFNγ production. The ability to induce potent antitumor activity by stimulating 4-1BB on tumor-specific cytotoxic T cells makes 4-1BB an attractive target for designing novel immuno-oncology therapeutics. To minimize systemic immune toxicities and enhance activity at the tumor site, we have developed a novel bispecific antibody that stimulates 4-1BB function when co-engaged with the tumor-associated antigen 5T4. ALG.APV-527 was built on the basis of the ADAPTIR bispecific platform with optimized binding domains to 4-1BB and 5T4 originating from the ALLIGATOR-GOLD human single-chain variable fragment library. The epitope of ALG.APV-527 was determined to be located at domain 1 and 2 on 4-1BB using X-ray crystallography. As shown in reporter and primary cell assays in vitro, ALG.APV-527 triggers dose-dependent 4-1BB activity mediated only by 5T4 crosslinking. In vivo, ALG.APV-527 demonstrates robust antitumor responses, by inhibiting growth of established tumors expressing human 5T4 followed by a long-lasting memory immune response. ALG.APV-527 has an antibody-like half-life in cynomolgus macaques and was well tolerated at 50.5 mg/kg. ALG.APV-527 is uniquely designed for 5T4-conditional 4-1BB-mediated antitumor activity with potential to minimize systemic immune activation and hepatotoxicity while providing efficacious tumor-specific responses in a range of 5T4-expressing tumor indications as shown by robust activity in preclinical in vitro and in vivo models. On the basis of the combined preclinical dataset, ALG.APV-527 has potential as a promising anticancer therapeutic for the treatment of 5T4-expressing tumors.


Subject(s)
Antibodies, Bispecific , Neoplasms , Single-Chain Antibodies , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antigens, Neoplasm , T-Lymphocytes , Tumor Necrosis Factor Receptor Superfamily, Member 9 , 4-1BB Ligand/metabolism
2.
Cancer Immunol Res ; 10(5): 626-640, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35319751

ABSTRACT

The CD40 receptor is an attractive target for cancer immunotherapy. Although a modest pharmacodynamic effect is seen in patients following administration of CD40-targeting monoclonal antibodies (mAb), the doses that could be safely administered do not result in a meaningful clinical response, most likely due to the limited therapeutic window associated with systemic CD40 activation. To overcome this issue, we developed a multispecific DARPin construct, α-FAPxCD40, which has conditional activity at the site of disease. α-FAPxCD40 activation of CD40 depends on binding to fibroblast activation protein (FAP), a cell-surface protease overexpressed in the stroma of solid tumors. In vitro studies demonstrated that α-FAPxCD40 potently activates human antigen-presenting cells in the presence, but not in the absence, of FAP-positive cells. After intravenous injection, a murine surrogate construct (α-mFAPxCD40) accumulated in FAP-positive tumors, elicited rejection of 88% of these tumors, and induced memory antitumor immunity. Importantly, in contrast to the mouse anti-CD40 tested in parallel, the in vivo antitumor activity of α-mFAPxCD40 was associated neither with elevated blood cytokines nor with hepatotoxicity, both of which contribute to the clinical dose-limiting toxicities of several CD40 mAb. This study demonstrates that α-(m)FAPxCD40 engages CD40 in an FAP-restricted manner, leading to tumor eradication without signs of peripheral toxicity. This distinct preclinical profile suggests that a favorable therapeutic index may be achieved in humans. It further supports the development of α-FAPxCD40, currently tested in a first-in-human clinical study in patients with solid tumors (NCT05098405).


Subject(s)
Antineoplastic Agents, Immunological , Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , CD40 Antigens , Cell Line, Tumor , Designed Ankyrin Repeat Proteins , Humans , Immunotherapy , Lymphocyte Activation , Mice , Neoplasms/drug therapy
3.
J Immunother Cancer ; 7(1): 103, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975201

ABSTRACT

BACKGROUND: The CTLA-4 blocking antibody ipilimumab has demonstrated substantial and durable effects in patients with melanoma. While CTLA-4 therapy, both as monotherapy and in combination with PD-1 targeting therapies, has great potential in many indications, the toxicities of the current treatment regimens may limit their use. Thus, there is a medical need for new CTLA-4 targeting therapies with improved benefit-risk profile. METHODS: ATOR-1015 is a human CTLA-4 x OX40 targeting IgG1 bispecific antibody generated by linking an optimized version of the Ig-like V-type domain of human CD86, a natural CTLA-4 ligand, to an agonistic OX40 antibody. In vitro evaluation of T-cell activation and T regulatory cell (Treg) depletion was performed using purified cells from healthy human donors or cell lines. In vivo anti-tumor responses were studied using human OX40 transgenic (knock-in) mice with established syngeneic tumors. Tumors and spleens from treated mice were analyzed for CD8+ T cell and Treg frequencies, T-cell activation markers and tumor localization using flow cytometry. RESULTS: ATOR-1015 induces T-cell activation and Treg depletion in vitro. Treatment with ATOR-1015 reduces tumor growth and improves survival in several syngeneic tumor models, including bladder, colon and pancreas cancer models. It is further demonstrated that ATOR-1015 induces tumor-specific and long-term immunological memory and enhances the response to PD-1 inhibition. Moreover, ATOR-1015 localizes to the tumor area where it reduces the frequency of Tregs and increases the number and activation of CD8+ T cells. CONCLUSIONS: By targeting CTLA-4 and OX40 simultaneously, ATOR-1015 is directed to the tumor area where it induces enhanced immune activation, and thus has the potential to be a next generation CTLA-4 targeting therapy with improved clinical efficacy and reduced toxicity. ATOR-1015 is also expected to act synergistically with anti-PD-1/PD-L1 therapy. The pre-clinical data support clinical development of ATOR-1015, and a first-in-human trial has started (NCT03782467).


Subject(s)
Antibodies, Bispecific/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Receptors, OX40/agonists , Urinary Bladder Neoplasms/drug therapy , Animals , Antibodies, Bispecific/therapeutic use , CHO Cells , CTLA-4 Antigen/immunology , Cell Line, Tumor/transplantation , Cricetulus , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Primary Cell Culture , Proof of Concept Study , Receptors, OX40/genetics , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
4.
Int J Cancer ; 145(5): 1189-1199, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30664811

ABSTRACT

Agonistic CD40 antibodies activate dendritic cells and can expand and activate tumor-specific T cells. Our purpose was to assess the CD40 agonistic antibody ADC-1013 in the clinical setting including intratumoral administration since preclinical studies have indicated that intratumoral is better than intravenous administration. A Phase I, open label, multicenter study was conducted in patients with advanced solid tumors who had received established treatments. A modified 3 + 3 dose-escalation was applied (every other week dosing). Twenty-three patients were treated with ADC-1013 intratumorally (dosing from 22.5 µg/kg up to 400 µg/kg) or intravenously (dosing at 75 µg/kg). The pharmacodynamic effects observed in the patients were further verified in an hCD40tg mouse model. Adverse events were mostly Common Terminology Criteria for Adverse Events (CTCAE) Grades 1 or 2 and transient. The serum concentration ADC-1013 and cytokine release (MCP-1, TNFα and IL-6) were more pronounced in patients receiving injections in deep metastases compared to patients receiving injections in superficial metastases. Treatment with ADC-1013 resulted in a marked decrease in B cell levels in peripheral blood after 24 h while remaining B cells significantly increased their expression of the cell surface activation marker CD86. Activation of antigen-presenting cells and subsequent activation of T cells were demonstrated in hCD40tg mice. Moreover, ADC-1013 treatment in this mouse model acted synergistically with a PD-1 inhibitor. The results from the first-in-human study of ADC-1013 indicate that intratumoral administration of ADC-1013 into superficial lesions is well tolerated at clinically relevant doses and associated with pharmacodynamic responses.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intralesional , Infusions, Intravenous , Macaca fascicularis , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Middle Aged , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Young Adult
5.
Ther Adv Vaccines Immunother ; 6(1): 3-17, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29998217

ABSTRACT

Following the clinical success of immune checkpoint antibodies targeting CTLA-4, PD-1 or PD-L1 in cancer treatment, bispecific antibodies are now emerging as a growing class of immunotherapies with potential to further improve clinical efficacy and safety. We describe three classes of immunotherapeutic bispecific antibodies: (a) cytotoxic effector cell redirectors; (b) tumor-targeted immunomodulators; and (c) dual immunomodulators. Cytotoxic effector cell redirectors are dominated by T-cell redirecting compounds, bispecific compounds engaging a tumor-associated antigen and the T-cell receptor/CD3 complex, thereby redirecting T-cell cytotoxicity to malignant cells. This is the most established class of bispecific immunotherapies, with two compounds having reached the market and numerous compounds in clinical development. Tumor-targeted immunomodulators are bispecific compounds binding to a tumor-associated antigen and an immunomodulating receptor, such as CD40 or 4-1BB. Such compounds are usually designed to be inactive until binding the tumor antigen, thereby localizing immune stimulation to the tumor environment, while minimizing immune activation elsewhere. This is expected to induce powerful activation of tumor-specific T cells with reduced risk of immune-related adverse events. Finally, dual immunomodulators are bispecific compounds that bind two distinct immunomodulating targets, often combining targeting of PD-1 or PD-L1 with that of LAG-3 or TIM-3. The rationale is to induce superior tumor immunity compared to monospecific antibodies to the same targets. In this review, we describe each of these classes of bispecific antibodies, and present examples of compounds in development.

6.
Clin Cancer Res ; 21(5): 1115-26, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25316820

ABSTRACT

PURPOSE: Local administration of immune-activating antibodies may increase the efficacy and reduce the immune-related adverse events associated with systemic immunotherapy of cancer. Here, we report the development and affinity maturation of a fully human agonistic CD40 antibody (IgG1), ADC-1013. EXPERIMENTAL DESIGN: We have used molecular engineering to generate an agonistic antibody with high affinity for CD40. The functional activity of ADC-1013 was investigated in human and murine in vitro models. The in vivo effect was investigated in two separate bladder cancer models, both using human xenograft tumors in immune deficient NSG mice and using a syngeneic bladder cancer model in a novel human CD40 transgenic mouse. RESULTS: Activation of dendritic cells (DC) by ADC-1013 results in upregulation of the costimulatory molecules CD80 and CD86, and secretion of IL12. ADC-1013 also activates DCs from human CD40 transgenic mice, and peptide-pulsed and ADC-1013-stimulated DCs induce antigen-specific T-cell proliferation in vitro. In vivo, treatment with ADC-1013 in a syngeneic bladder cancer model, negative for hCD40, induces significant antitumor effects and long-term tumor-specific immunity. Furthermore, ADC-1013 demonstrates significant antitumor effects in a human bladder cancer transplanted into immunodeficient NSG mice. CONCLUSIONS: Our data demonstrate that ADC-1013 induces long-lasting antitumor responses and immunologic memory mediated by CD40 stimulation. To the best of our knowledge, ADC-1013 represents the first immunomodulatory antibody developed for local immunotherapy of cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , CD40 Antigens/agonists , T-Lymphocytes/immunology , Urinary Bladder Neoplasms/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , CD40 Antigens/chemistry , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Models, Molecular , Molecular Conformation , Protein Binding , T-Lymphocytes/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 23(4): 502-15, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597564

ABSTRACT

We isolated a tumor B-cell-targeting antibody, BI-505, from a highly diversified human phage-antibody library, using a pioneering "function-first" approach involving screening for (1) specificity for a tumor B cell surface receptor, (2) induction of tumor programmed cell death, and (3) enhanced in vivo antitumor activity compared to currently used treatments. BI-505 bound to intercellular adhesion molecule-1, identifying a previously unrecognized role for this receptor as a therapeutic target in cancer. The BI-505 epitope was strongly expressed on the surface of multiple myeloma cells from both newly diagnosed and relapsed patients. BI-505 had potent macrophage-dependent antimyeloma activity and conferred enhanced survival compared to currently used treatments in advanced experimental models of multiple myeloma.


Subject(s)
Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , Intercellular Adhesion Molecule-1/immunology , Macrophages/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , B-Lymphocytes/metabolism , Cell Line, Tumor , Epitopes/biosynthesis , Epitopes/immunology , Female , Humans , Macrophages/metabolism , Male , Mice , Mice, SCID , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Peptide Library , Receptors, IgG/immunology , Xenograft Model Antitumor Assays
8.
Biochim Biophys Acta ; 1783(3): 429-37, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18164266

ABSTRACT

We have previously shown that angiomotin (Amot) plays an important role in growth factor-induced migration of endothelial cells in vitro. Genetic knock-down of Amot in zebrafish also results in inhibition of migration of intersegmental vessels in vivo. Amot is expressed as two different isoforms, p80-Amot and p130-Amot. Here we have analyzed the expression of the two Amot isoforms during retinal angiogenesis in vivo and demonstrate that p80-Amot is expressed during the migratory phase. In contrast, p130-Amot is expressed during the period of blood vessel stabilization and maturation. We also show that the N-terminal domain of p130-Amot serves as a targeting domain responsible for localization of p130-Amot to actin and tight junctions. We further show that the relative expression levels of p80-Amot and p130-Amot regulate a switch between a migratory and a non-migratory cell phenotype where the migratory function of p80-Amot is dominant over the stabilization and maturation function of p130-Amot. Our data indicates that homo-oligomerization of p80-Amot and hetero-oligomerization of both isoforms are critical for this regulation.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Endothelial Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Microfilament Proteins/physiology , Angiomotins , Animals , Animals, Newborn , CHO Cells , Cell Communication/physiology , Cells, Cultured , Cricetinae , Cricetulus , Dimerization , Dogs , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Neovascularization, Physiologic/physiology , Protein Binding , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Transport , Retinal Vessels/growth & development , Retinal Vessels/metabolism
9.
FASEB J ; 22(3): 880-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17984175

ABSTRACT

We have previously shown that angiomotin (Amot) mediates angiostatin inhibition of endothelial migration and tube formation in vitro. A crucial role of angiomotin in regulating endothelial cell motility is indicated by the findings that knockdown of Amot in zebrafish reduces the number of filopodia of endothelial tip cells and severely impairs the migration of intersegmental vessels. In addition, targeting angiomotin using DNA vaccination inhibits angiogenesis and tumor growth in vivo. In this report, we have generated antibodies that, similar to angiostatin, bind to angiomotin on the endothelial cell surface. These antibodies inhibited FGF-2 and vascular endothelial growth factor (VEGF) -induced endothelial migration in the Boyden chamber assay. Furthermore, the anti-Amot B06 antibody significantly reduced the number of endothelial filopodia and inhibited vessel migration during retinal angiogenesis in vivo. We also show that systemic or local treatment with this antibody inhibits pathological blood vessel formation associated with tumor growth or laser-induced choroid neovascularization of the eye. These findings provide a rationale for using angiomotin antibodies for specifically targeting endothelial migration in angiogenesis-dependent diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Endothelium/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Angiomotins , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Aorta/cytology , Cell Movement/drug effects , Cells, Cultured , Endothelium/cytology , Endothelium/drug effects , Humans , Mice , Pseudopodia/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Retina/cytology
10.
Proc Natl Acad Sci U S A ; 103(24): 9208-13, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16754857

ABSTRACT

Endogenous angiogenesis inhibitors have shown promise in preclinical trials, but clinical use has been hindered by low half-life in circulation and high production costs. Here, we describe a strategy that targets the angiostatin receptor angiomotin (Amot) by DNA vaccination. The vaccination procedure generated antibodies that detected Amot on the endothelial cell surface. Purified Ig bound to the endothelial cell membrane and inhibited endothelial cell migration. In vivo, DNA vaccination blocked angiogenesis in the matrigel plug assay and prevented growth of transplanted tumors for up to 150 days. We further demonstrate that a combination of DNA vaccines encoding Amot and the extracellular and transmembrane domains of the human EGF receptor 2 (Her-2)/neu oncogene inhibited breast cancer progression and impaired tumor vascularization in Her-2/neu transgenic mice. No toxicity or impairment of normal blood vessels could be detected. This work shows that DNA vaccination targeting Amot may be used to mimic the effect of angiostatin.


Subject(s)
Intercellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Neoplasms/pathology , Neovascularization, Pathologic , Vaccines, DNA , Angiomotins , Animals , Antibodies/metabolism , Cell Line , Cell Movement/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microfilament Proteins , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Gastroenterology ; 130(4): 1301-10, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16618420

ABSTRACT

BACKGROUND & AIMS: Recent studies indicate that kringle 1-5 has a potent and specific antiangiogenic activity. Here, we investigated the antitumor effect of kringle 1-5 gene transfer on hepatocellular carcinoma in mice. METHODS: The inhibitory effect of kringle 1-5 protein on proliferation of bovine capillary endothelial cells was evaluated by a tetrazolium-based assay. To study tumor growth, intrahepatic metastasis, and survival, liposome/kringle 1-5 complementary DNA complexes were injected intravenously in nude mice preimplanted with 1 of 3 hepatoma cell lines into the liver. Production of kringle 1-5 was tested by immunohistochemistry and Western blotting. Intratumoral vessel density was quantified. Expression of vascular endothelial growth factor, angiopoietin-1, and angiopoietin-2 in tumors was examined by Western blotting. Serum alanine aminotransferase and alpha-fetoprotein levels and body weights were measured. RESULTS: Proliferation of bovine capillary endothelial cells was inhibited by purified kringle 1-5 in a dose-dependent manner. Gene transfer of kringle 1-5 caused a significant reduction in vessel density with suppression of tumor growth of the 3 hepatoma cell lines and serum alpha-fetoprotein levels, prolonged the survival period, and reduced the number of intrahepatic metastases. Among the analyzed angiogenic factors, kringle 1-5 reduced angiopoietin-2 expression levels. Expression of kringle 1-5 protein was detected on hepatoma cells and hepatocytes in the liver. However, it did not alter serum alanine aminotransferase levels and body weights, suggesting kringle 1-5 lacks severe side effects. CONCLUSIONS: Antiangiogenic gene therapy with kringle 1-5 complementary DNA is a promising safe and effective strategy for suppression of growth of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Transfer Techniques , Kringles/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Alanine Transaminase/blood , Animals , Body Weight/drug effects , COS Cells , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/physiopathology , Cattle , Cell Proliferation/drug effects , Chlorocebus aethiops , DNA, Complementary , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Liver/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/physiopathology , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/pathology , Prognosis , Survival Analysis , Transfection
12.
FEBS J ; 273(9): 2000-11, 2006 May.
Article in English | MEDLINE | ID: mdl-16640563

ABSTRACT

Angiomotin, an 80 kDa protein expressed in endothelial cells, promotes cell migration and invasion, and stabilizes tube formation in vitro. Angiomotin belongs to a new protein family with two additional members, Amotl-1 and Amotl-2, which are characterized by conserved coiled-coil domains and C-terminal PDZ binding motifs. Here, we report the identification of a 130 kDa splice isoform of angiomotin that is expressed in different cell types including vascular endothelial cells, as well as cytotrophoblasts of the placenta. p130-Angiomotin consists of a cytoplasmic N-terminal extension that mediates its association with F-actin. Transfection of p130-angiomotin into endothelial cells induces actin fiber formation and changes cell shape. The p130-angiomotin protein remained associated with actin after destabilization of actin fibers with cytochalasin B. In contrast to p80-angiomotin, p130-angiomotin does not promote cell migration and did not respond to angiostatin. We propose that p80- and p130-angiomotin play coordinating roles in tube formation by affecting cell migration and cell shape, respectively.


Subject(s)
Actins/metabolism , Cell Shape/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Alternative Splicing , Amino Acid Sequence , Angiomotins , Angiostatins/physiology , Animals , Cell Line , Cell Line, Transformed , Cell Movement/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Molecular Sequence Data , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology
13.
J Biol Chem ; 280(41): 34859-69, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16043488

ABSTRACT

We have previously identified angiomotin by its ability to bind to and mediate the anti-angiogenic properties of angiostatin. In vivo and in vitro data indicate an essential role of angiomotin in endothelial cell motility. Here we show that angiostatin binds angiomotin on the cell surface and provide evidence for a transmembrane model for the topology of both p80 and p130 angiomotin isoforms. Immunofluorescence analysis shows that angiomotin co-localized with ZO-1 in cell-cell contacts in endothelial cells in vitro and in angiogenic blood vessels of the postnatal mouse retina in vivo. Transfection of p80 as well as p130 angiomotin in Chinese hamster ovary cells resulted in junctional localization of both isoforms. Furthermore, p130 angiomotin could recruit ZO-1 to actin stress fibers. The p130 but not p80 isoform could be coprecipitated with MAGI-1b, a component of endothelial tight junctions. Paracellular permeability, as measured by diffusion of fluorescein isothiocyanate-dextran, was reduced by p80 and p130 angiomotin expression with 70 and 88%, respectively, compared with control. Angiostatin did not have any effect on cell permeability but inhibited the migration of angiomotin-expressing cells in the Boyden chamber assay. We conclude that angiomotin, in addition to controlling cell motility, may play a role in the assembly of endothelial cell-cell junctions.


Subject(s)
Carrier Proteins/physiology , Endothelial Cells/cytology , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/physiology , Alternative Splicing , Angiomotins , Angiopoietin-Like Protein 1 , Angiostatins/chemistry , Angiostatins/metabolism , Animals , Biotinylation , Blotting, Western , CHO Cells , Capillaries/cytology , Cattle , Cell Communication , Cell Membrane/metabolism , Cell Movement , Computational Biology , Cricetinae , Cross-Linking Reagents/pharmacology , Diffusion , Endothelium, Vascular/cytology , Fluorescein-5-isothiocyanate/pharmacology , HeLa Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Microfilament Proteins , Microscopy, Fluorescence , Octoxynol , Permeability , Phosphoproteins/metabolism , Plasmids/metabolism , Polyethylene Glycols/pharmacology , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Proteins/metabolism , Retina/metabolism , Retinoblastoma-Like Protein p130/metabolism , Time Factors , Trypsin/pharmacology , Zonula Occludens-1 Protein
14.
Cancer Res ; 64(10): 3679-86, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15150128

ABSTRACT

We have recently reported the identification of kringle 1-5 (K1-5) of plasminogen as a potent and specific inhibitor of angiogenesis and tumor growth. Here, we show that K1-5 bound to endothelial cell surface ATP synthase and triggered caspase-mediated endothelial cell apoptosis. Induction of endothelial apoptosis involved sequential activation of caspases-8, -9, and -3. Administration of neutralizing antibodies directed against the alpha- and beta-subunits of ATP synthase to endothelial cells attenuated activation of these caspases. Furthermore, inhibitors of caspases-3, -8, and -9 also remarkably blocked K1-5-induced endothelial cell apoptosis and antiangiogenic responses. In a mouse tumor model, we show that caspase-3 inhibitors abolished the antitumor activity of K1-5 by protecting the tumor vasculature undergoing apoptosis. These results suggest that the specificity of the antiendothelial effect of K1-5 is attributable, at least in part, to its interaction with the endothelial cell surface ATP synthase and that the caspase-mediated endothelial apoptosis is essential for the angiostatic activity of K1-5. Thus, our findings provide a mechanistic insight with respect to the angiostatic action and signaling pathway of K1-5 and angiostatin.


Subject(s)
ATP Synthetase Complexes/metabolism , Caspases/metabolism , Endothelium, Vascular/enzymology , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Kringles , Neovascularization, Pathologic/drug therapy , Plasminogen/pharmacology , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase Inhibitors , Chick Embryo , Cornea/blood supply , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Fibrosarcoma/enzymology , Humans , Isoenzymes , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Plasminogen/metabolism
15.
Proc Natl Acad Sci U S A ; 101(8): 2476-81, 2004 Feb 24.
Article in English | MEDLINE | ID: mdl-14983034

ABSTRACT

Obesity is a risk factor for the development of many severe human diseases such as cardiovascular disorders, diabetes, and cancer, which are tightly linked to angiogenesis. The adipose tissue produces several growth factors/hormones including leptin, tumor necrosis factor alpha, and adiponectin. It has been found that adiponectin levels are reduced in obesity. Here, we report a unique function of adiponectin as a negative regulator of angiogenesis. In vitro, adiponectin potently inhibits endothelial cell proliferation and migration. In the chick chorioallantoic membrane and the mouse corneal angiogenesis assays, adiponectin remarkably prevents new blood vessel growth. Further, we demonstrate that the antiendothelial mechanisms involve activation of caspase-mediated endothelial cell apoptosis. Adiponectin induces a cascade activation of caspases-8, -9, and -3, which leads to cell death. In a mouse tumor model, adiponectin significantly inhibits primary tumor growth. Impaired tumor growth is associated with decreased neovascularization, leading to significantly increased tumor cell apoptosis. These data demonstrate induction of endothelial apoptosis as an unique mechanism of adiponectin-induced antiangiogenesis. Adiponectin, as a direct endogenous angiogenesis inhibitor, may have therapeutic implications in the treatment of angiogenesis-dependent diseases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Endothelium, Vascular/cytology , Intercellular Signaling Peptides and Proteins , Proteins/pharmacology , Adiponectin , Allantois/blood supply , Allantois/drug effects , Animals , Apoptosis/drug effects , Capillaries , Caspases/metabolism , Cattle , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Collagen/pharmacology , Endothelium, Vascular/drug effects , Female , Fibrosarcoma/blood supply , Humans , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor Receptor-2/physiology
16.
FASEB J ; 17(6): 764-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12586745

ABSTRACT

Recent studies show that stable expression of the human telomerase catalytic subunit, hTERT, alone can lead several types of normal human somatic cells to bypass replicative senescence and become immortal. The molecular mechanisms by which telomerase immortalizes cells are not fully understood, although a key function of telomerase is to maintain a critical length of telomeres in order to preserve the stability and integrity of the genome. Here we report that stable transfection of hTERT alone was sufficient to allow bovine capillary endothelial (BCE) cells to bypass senescence and acquire immortality. Surprisingly, telomere lengths in these stable transfectants were progressively shortened during an increasing number of population doublings (PDLs), despite high telomerase activity. The expression of the cyclin-dependent kinase inhibitors (CDKIs) p16INK4A and p21CIP1/WAF1 was concomitantly repressed, and the retinoblastoma protein (pRb) was maintained in a hyperphosphorylated state in the telomerase-expressing cells. Re-expression of p16INK4A in these cells by either treatment with a demethylating agent or by adenovirus-mediated expression reinduced a senescence-like phenotype, suggesting that the inactivation of p16INK4A was due to DNA methylation and was crucial for the immortalization process. In agreement with this finding, the expression levels of the prototypic DNA methyltransferase DNMT1 were elevated in the hTERT-positive cells.


Subject(s)
Azacitidine/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelium, Vascular/metabolism , Retinoblastoma Protein/metabolism , Telomerase/metabolism , Animals , Azacitidine/pharmacology , Cattle , Cell Line, Transformed , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Modification Methylases/antagonists & inhibitors , DNA-Binding Proteins , Decitabine , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Humans , Telomerase/genetics , Telomere/genetics , Telomere/metabolism , Transfection
17.
Curr Med Chem Anticancer Agents ; 2(6): 667-81, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12678719

ABSTRACT

The quiescent vascular system in the adult body represents the balanced net outcome of overproduction of endogenous angiogenesis inhibitors and reduced levels of angiogenic factors. While these inhibitors are expressed under physiological conditions, they are also generated in association with tumor growth. Angiostatin is such a specific angiogenesis inhibitor produced by tumors. It inhibits primary and metastatic tumor growth by blocking tumor angiogenesis. Encouraged by its potent anti-tumor activity, angiostatin is in clinical trials for cancer therapy. Angiostatin contains the first four triple loop structures, known as kringle domains, of plasminogen. The disulfide bond-linked kringle architectures are essential for the antiangiogenic activity of angiostatin. Based on this initial finding, recent work shows that kringle fragments of several other proteins also inhibit angiogenesis. Thus, kringle domains may provide a structural basis for identification of novel angiogenesis inhibitors. Surprisingly, most kringles only inhibit angiogenesis when cleaved as fragments from their parental proteins that lack antiangiogenic activity. These findings suggest that they are cryptic fragments hidden in large protein molecules. Thus, proteolytic processing plays a critical role in down regulation of angiogenesis. The kringle structure may provide the first example of a conserved architecture that specifically inhibits blood vessel growth. This review will focus on the structural and functional relationships of kringle domains in regulation of angiogenesis and tumor growth.


Subject(s)
Angiogenesis Inhibitors/chemistry , Kringles , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...