Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 14(3): 649-58, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25567130

ABSTRACT

p53 is a critical tumor suppressor and is the most frequently inactivated gene in human cancer. Inhibition of the interaction of p53 with its negative regulator MDM2 represents a promising clinical strategy to treat p53 wild-type tumors. AMG 232 is a potential best-in-class inhibitor of the MDM2-p53 interaction and is currently in clinical trials. We characterized the activity of AMG 232 and its effect on p53 signaling in several preclinical tumor models. AMG 232 binds the MDM2 protein with picomolar affinity and robustly induces p53 activity, leading to cell-cycle arrest and inhibition of tumor cell proliferation. AMG 232 treatment inhibited the in vivo growth of several tumor xenografts and led to complete and durable regression of MDM2-amplified SJSA-1 tumors via growth arrest and induction of apoptosis. Therapeutic combination studies of AMG 232 with chemotherapies that induce DNA damage and p53 activity resulted in significantly superior antitumor efficacy and regression, and markedly increased activation of p53 signaling in tumors. These preclinical data support the further evaluation of AMG 232 in clinical trials as both a monotherapy and in combination with standard-of-care cytotoxics.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Piperidones/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , HT29 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Xenograft Model Antitumor Assays/methods
2.
J Med Chem ; 57(24): 10499-511, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25384157

ABSTRACT

Structure-based rational design and extensive structure-activity relationship studies led to the discovery of AMG 232 (1), a potent piperidinone inhibitor of the MDM2-p53 association, which is currently being evaluated in human clinical trials for the treatment of cancer. Further modifications of 1, including replacing the carboxylic acid with a 4-amidobenzoic acid, afforded AM-7209 (25), featuring improved potency (KD from ITC competition was 38 pM, SJSA-1 EdU IC50 = 1.6 nM), remarkable pharmacokinetic properties, and in vivo antitumor activity in both the SJSA-1 osteosarcoma xenograft model (ED50 = 2.6 mg/kg QD) and the HCT-116 colorectal carcinoma xenograft model (ED50 = 10 mg/kg QD). In addition, 25 possesses distinct mechanisms of elimination compared to 1.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Drug Discovery , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-mdm2/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
3.
Lab Invest ; 94(11): 1296-308, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25243900

ABSTRACT

Sustained angiogenesis is essential for tumor growth as it provides the tumor with a network of blood vessels that supply both oxygen and essential nutrients. Limiting tumor-associated angiogenesis is a proven strategy for the treatment of human cancer. To date, the rapid detection and quantitation of tumor-associated endothelial cell (TAEC) proliferation has been challenging, largely due to the low frequency of endothelial cells (ECs) within the tumor microenvironment. In this report, we address this problem using a new multiparametric flow cytometry method capable of rapid and precise quantitation of proliferation by measuring bromodeoxyuridine (BrdUrd) uptake in mouse TAECs from established human tumor xenografts. We determined the basal proliferation labeling index of TAECs in two human tumor xenografts representing two distinct histologies, COLO 205 (colorectal cancer) and U-87 (glioblastoma). We then investigated the effects of two large-molecule antiangiogenic agents targeting different biochemical pathways. Blocking angiopoietin-Tie2 signaling with the peptide-Fc fusion protein, trebananib (AMG 386), inhibited proliferation of TAECs, whereas blocking Dll4-Notch signaling with an anti-Dll4-specific antibody induced hyperproliferation of TAECs. These pharmacodynamic studies highlight the sensitivity and utility of this flow cytometry-based method and demonstrate the value of this assay to rapidly assess the in vivo proliferative effects of angiogenesis-targeted agents on both the tumor and the associated vasculature.


Subject(s)
Antibodies, Neutralizing/pharmacology , Endothelial Cells/drug effects , Flow Cytometry/methods , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Receptor, TIE-2/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/therapeutic use , Bromodeoxyuridine , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Female , Glioblastoma/pathology , Humans , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Recombinant Fusion Proteins/therapeutic use , Xenograft Model Antitumor Assays
4.
ACS Med Chem Lett ; 5(8): 894-9, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147610

ABSTRACT

Continued optimization of the N-substituent in the piperidinone series provided potent piperidinone-pyridine inhibitors 6, 7, 14, and 15 with improved pharmacokinetic properties in rats. Reducing structure complexity of the N-alkyl substituent led to the discovery of 23, a potent and simplified inhibitor of MDM2. Compound 23 exhibits excellent pharmacokinetic properties and substantial in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft mouse model.

5.
Oncotarget ; 5(8): 2030-43, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24810962

ABSTRACT

While MDM2 inhibitors hold great promise as cancer therapeutics, drug resistance will likely limit their efficacy as single agents. To identify drug combinations that might circumvent resistance, we screened for agents that could synergize with MDM2 inhibition in the suppression of cell viability. We observed broad and robust synergy when combining MDM2 antagonists with either MEK or PI3K inhibitors. Synergy was not limited to cell lines harboring MAPK or PI3K pathway mutations, nor did it depend on which node of the PI3K axis was targeted. MDM2 inhibitors also synergized strongly with BH3 mimetics, BCR-ABL antagonists, and HDAC inhibitors. MDM2 inhibitor-mediated synergy with agents targeting these mechanisms was much more prevalent than previously appreciated, implying that clinical translation of these combinations could have far-reaching implications for public health. These findings highlight the importance of combinatorial drug targeting and provide a framework for the rational design of MDM2 inhibitor clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Gene Expression/drug effects , Humans
6.
Transl Oncol ; 7(2): 188-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24704536

ABSTRACT

Angiopoietin 2 (Ang2) is a secreted glycoprotein upregulated at sites of angiogenesis and has been implicated in cancer neovascularization. Recent studies have suggested efficacy of combined Ang and vascular endothelial growth factor receptor (VEGFR) inhibition for patients with metastatic renal cell carcinoma (mRCC). We measured Ang2 expression in human tissue and plasma, and tested the effect of dual Ang1/2 (trebananib; AMG386) or Ang2 alone (L1-7) inhibition with VEGFR inhibition on murine RCC growth and blood flow. Ang2 levels were higher in human tumors than normal tissues with RCC ranking highest for Ang2 expression across all tumor types tested. Plasma Ang2 was significantly higher in patients with mRCC compared to controls or patients with stage I disease. Plasma Ang2 decreased with sunitinib treatment and increased at time of disease progression. In the RCC mouse, dual Ang1/2 and Ang2 inhibition improved the activity of sunitinib. Combined Ang1/2 and VEGFR inhibition prevented the resumption of blood flow associated with sunitinib resistance. Thus, Ang2 inhibition, independent of Ang1 inhibition, improves the activity of sunitinib and plasma Ang2 increases in the setting of progression on sunitinib possibly contributing to resistance. Further, arterial spin-labeled perfusion magnetic resonance imaging might be a non-invasive marker of the antiangiogenic activity of Ang inhibitors.

7.
J Med Chem ; 57(7): 2963-88, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24601644

ABSTRACT

We previously reported the discovery of potent and selective morpholinone and piperidinone inhibitors of the MDM2-p53 interaction. These inhibitors have in common a carboxylic acid moiety that engages in an electrostatic interaction with MDM2-His96. Our continued search for potent and diverse inhibitors led to the discovery of novel replacements for these acids uncovering new interactions with the MDM2 protein. In particular, using pyridine or thiazole as isosteres of the carboxylic acid moiety resulted in very potent analogues. From these, AM-6761 (4) emerged as a potent inhibitor with remarkable biochemical (HTRF IC50 = 0.1 nM) and cellular potency (SJSA-1 EdU IC50 = 16 nM), as well as favorable pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 11 mg/kg. Optimization efforts toward the discovery of these inhibitors as well as the new interactions observed with the MDM2 protein are described herein.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Carboxylic Acids/pharmacology , Cell Proliferation/drug effects , Myocytes, Smooth Muscle/drug effects , Piperidones/pharmacology , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Acetates/chemistry , Animals , Bone Neoplasms/drug therapy , Carboxylic Acids/chemistry , Cells, Cultured , Crystallography, X-Ray , Drug Design , Female , Humans , Hydrogen Bonding , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Osteosarcoma/drug therapy , Piperidones/chemistry , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
J Med Chem ; 57(6): 2472-88, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24548297

ABSTRACT

We previously reported the discovery of AMG 232, a highly potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Our continued search for potent and diverse analogues led to the discovery of novel morpholinone MDM2 inhibitors. This change to a morpholinone core has a significant impact on both potency and metabolic stability compared to the piperidinone series. Within this morpholinone series, AM-8735 emerged as an inhibitor with remarkable biochemical potency (HTRF IC50 = 0.4 nM) and cellular potency (SJSA-1 EdU IC50 = 25 nM), as well as pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 41 mg/kg. Lead optimization toward the discovery of this inhibitor as well as key differences between the morpholinone and the piperidinone series will be described herein.


Subject(s)
Acetates/chemical synthesis , Acetates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Morpholines/chemical synthesis , Morpholines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/chemistry , Animals , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Humans , Indicators and Reagents , Mice , Models, Molecular , Molecular Conformation , Morpholines/pharmacokinetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
J Med Chem ; 57(4): 1454-72, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24456472

ABSTRACT

We recently reported the discovery of AM-8553 (1), a potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Continued research investigation of the N-alkyl substituent of this series, focused in particular on a previously underutilized interaction in a shallow cleft on the MDM2 surface, led to the discovery of a one-carbon tethered sulfone which gave rise to substantial improvements in biochemical and cellular potency. Further investigation produced AMG 232 (2), which is currently being evaluated in human clinical trials for the treatment of cancer. Compound 2 is an extremely potent MDM2 inhibitor (SPR KD = 0.045 nM, SJSA-1 EdU IC50 = 9.1 nM), with remarkable pharmacokinetic properties and in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft model (ED50 = 9.1 mg/kg).


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Piperidones/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Acetates/chemistry , Administration, Oral , Antineoplastic Agents/chemistry , Biological Availability , Crystallography, X-Ray , Drug Discovery , Humans , Piperidones/chemistry , Protein Conformation
10.
J Med Chem ; 56(10): 4053-70, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23597064

ABSTRACT

Structural analysis of both the MDM2-p53 protein-protein interaction and several small molecules bound to MDM2 led to the design and synthesis of tetrasubstituted morpholinone 10, an MDM2 inhibitor with a biochemical IC50 of 1.0 µM. The cocrystal structure of 10 with MDM2 inspired two independent optimization strategies and resulted in the discovery of morpholinones 16 and 27 possessing distinct binding modes. Both analogues were potent MDM2 inhibitors in biochemical and cellular assays, and morpholinone 27 (IC50 = 0.10 µM) also displayed suitable PK profile for in vivo animal experiments. A pharmacodynamic (PD) experiment in mice implanted with human SJSA-1 tumors showed p21(WAF1) mRNA induction (2.7-fold over vehicle) upon oral dosing of 27 at 300 mg/kg.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Circular Dichroism , Crystallography , Crystallography, X-Ray , Drug Design , Female , Humans , Indicators and Reagents , Mice , Mice, Nude , Models, Molecular , Morpholines/chemical synthesis , Morpholines/pharmacology , Piperidines/chemical synthesis , Piperidines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
J Med Chem ; 55(11): 4936-54, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22524527

ABSTRACT

Structure-based rational design led to the discovery of novel inhibitors of the MDM2-p53 protein-protein interaction. The affinity of these compounds for MDM2 was improved through conformational control of both the piperidinone ring and the appended N-alkyl substituent. Optimization afforded 29 (AM-8553), a potent and selective MDM2 inhibitor with excellent pharmacokinetic properties and in vivo efficacy.


Subject(s)
Acetates/chemical synthesis , Antineoplastic Agents/chemical synthesis , Piperidones/chemical synthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Acetates/pharmacokinetics , Acetates/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Hepatocytes/metabolism , Humans , Macaca fascicularis , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Neoplasm Transplantation , Piperidones/pharmacokinetics , Piperidones/pharmacology , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , rho GTP-Binding Proteins/biosynthesis
12.
Bioorg Med Chem Lett ; 21(9): 2752-5, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21123063

ABSTRACT

Human murine double minute 2 (MDM2) is a negative regulator of p53, which plays an important role in cell cycle and apoptosis. We report several optimizations to the synthesis of the chromenotriazolopyrimidine series of MDM2-p53 protein-protein interaction inhibitors. Additionally, the in vitro and in vivo stability, pharmacokinetic properties and solubility were improved through N-substitution.


Subject(s)
Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrimidines/chemical synthesis , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Drug Stability , Humans , Infusion Pumps , Inhibitory Concentration 50 , Molecular Structure , Pyrimidines/pharmacokinetics , Rats , Solubility , Structure-Activity Relationship
13.
Mol Cancer Ther ; 9(10): 2641-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20937592

ABSTRACT

AMG 386 is an investigational first-in-class peptide-Fc fusion protein (peptibody) that inhibits angiogenesis by preventing the interaction of angiopoietin-1 (Ang1) and Ang2 with their receptor, Tie2. Although the therapeutic value of blocking Ang2 has been shown in several models of tumorigenesis and angiogenesis, the potential benefit of Ang1 antagonism is less clear. To investigate the consequences of Ang1 neutralization, we have developed potent and selective peptibodies that inhibit the interaction between Ang1 and its receptor, Tie2. Although selective Ang1 antagonism has no independent effect in models of angiogenesis-associated diseases (cancer and diabetic retinopathy), it induces ovarian atrophy in normal juvenile rats and inhibits ovarian follicular angiogenesis in a hormone-induced ovulation model. Surprisingly, the activity of Ang1 inhibitors seems to be unmasked in some disease models when combined with Ang2 inhibitors, even in the context of concurrent vascular endothelial growth factor inhibition. Dual inhibition of Ang1 and Ang2 using AMG 386 or a combination of Ang1- and Ang2-selective peptibodies cooperatively suppresses tumor xenograft growth and ovarian follicular angiogenesis; however, Ang1 inhibition fails to augment the suppressive effect of Ang2 inhibition on tumor endothelial cell proliferation, corneal angiogenesis, and oxygen-induced retinal angiogenesis. In no case was Ang1 inhibition shown to (a) confer superior activity to Ang2 inhibition or dual Ang1/2 inhibition or (b) antagonize the efficacy of Ang2 inhibition. These results imply that Ang1 plays a context-dependent role in promoting postnatal angiogenesis and that dual Ang1/2 inhibition is superior to selective Ang2 inhibition for suppression of angiogenesis in some postnatal settings.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Cell Division/drug effects , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Cornea/blood supply , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental/blood supply , Ovarian Follicle/blood supply , Rats , Rats, Sprague-Dawley
14.
Cancer Res ; 70(6): 2213-23, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197469

ABSTRACT

Inhibition of angiopoietin-2 (Ang2) can slow tumor growth, but the underlying mechanism is not fully understood. Because Ang2 is expressed in growing blood vessels and promotes angiogenesis driven by vascular endothelial growth factor (VEGF), we asked whether the antitumor effect of Ang2 inhibition results from reduced sprouting angiogenesis and whether the effect is augmented by inhibition of VEGF from tumor cells. Using Colo205 human colon carcinomas in nude mice as a model, we found that selective inhibition of Ang2 by the peptide-Fc fusion protein L1-7(N) reduced the number of vascular sprouts by 46% and tumor growth by 62% over 26 days. Strikingly, when the Ang2 inhibitor was combined with a function-blocking anti-VEGF antibody, the number of sprouts was reduced by 82%, tumor vascularity was reduced by 67%, and tumor growth slowed by 91% compared with controls. The reduction in tumor growth was accompanied by decreased cell proliferation and increased apoptosis. We conclude that inhibition of Ang2 slows tumor growth by limiting the expansion of the tumor vasculature by sprouting angiogenesis, in a manner that is complemented by concurrent inhibition of VEGF and leads to reduced proliferation and increased apoptosis of tumor cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiopoietin-2/biosynthesis , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Growth Processes/drug effects , Colonic Neoplasms/pathology , Drug Synergism , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Rats , Receptors, Fc/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
15.
J Med Chem ; 52(22): 7044-53, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19856920

ABSTRACT

Tumor protein 53 (p53) is a critical regulator of cell cycle and apoptosis that is frequently disabled in human tumors. In many tumor types, p53 is deleted or mutated, but in others p53 is inactivated by overexpression or amplification of its negative regulator mouse double minute 2 (MDM2). A high-throughput screening effort identified 6,7-bis(4-bromophenyl)-7,12-dihydro-6H-chromeno[4,3-d][1,2,4]triazolo[1,5-a]pyrimidine as a potent inhibitor of the MDM2-p53 protein-protein interaction. This screening hit was found to be chemically unstable and difficult to handle due to poor DMSO solubility. Co-crystallization with the target protein helped to direct further optimization and provided a tractable lead series of novel MDM2-p53 inhibitors. In cellular assays, these compounds were shown to upregulate p53 protein levels and p53 signaling and to cause p53-dependent inhibition of proliferation and apoptosis.


Subject(s)
Drug Discovery , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , HCT116 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Protein Binding/drug effects , Stereoisomerism , Structure-Activity Relationship
16.
Cancer Cell ; 6(5): 507-16, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542434

ABSTRACT

Angiopoietin-2 (Ang2) exhibits broad expression in the remodeling vasculature of human tumors but very limited expression in normal tissues, making it an attractive candidate target for antiangiogenic cancer therapy. To investigate the functional consequences of blocking Ang2 activity, we generated antibodies and peptide-Fc fusion proteins that potently and selectively neutralize the interaction between Ang2 and its receptor, Tie2. Systemic treatment of tumor-bearing mice with these Ang2-blocking agents resulted in tumor stasis, followed by elimination of all measurable tumor in a subset of animals. These effects were accompanied by reduced endothelial cell proliferation, consistent with an antiangiogenic therapeutic mechanism. Anti-Ang2 therapy also prevented VEGF-stimulated neovascularization in a rat corneal model of angiogenesis. These results imply that specific Ang2 inhibition may represent an effective antiangiogenic strategy for treating patients with solid tumors.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Antibodies/pharmacology , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Animals , Cell Proliferation/drug effects , Cornea/blood supply , Endothelial Cells/drug effects , Female , Mice , Mice, Nude , Neoplasm Transplantation , Neutralization Tests , Receptors, Fc , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...