Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
EMBO J ; 43(5): 666-694, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38279026

ABSTRACT

The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.


Subject(s)
Anaphase-Promoting Complex-Cyclosome , Neoplasms , Humans , Anaphase-Promoting Complex-Cyclosome/genetics , Dyneins , Kinesins/genetics , Kinetochores , Mitosis , Neoplasms/genetics
2.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38151625

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Subject(s)
Kinesins , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Kinesins/genetics , Kinesins/metabolism , Mitosis/genetics , Cell Line , M Phase Cell Cycle Checkpoints
3.
J Med Chem ; 65(6): 4972-4990, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35286090

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer that results from errors in chromosome segregation during mitosis. Targeting of CIN-associated vulnerabilities is an emerging therapeutic strategy in drug development. KIF18A, a mitotic kinesin, has been shown to play a role in maintaining bipolar spindle integrity and promotes viability of CIN cancer cells. To explore the potential of KIF18A, a series of inhibitors was identified. Optimization of an initial hit led to the discovery of analogues that could be used as chemical probes to interrogate the role of KIF18A inhibition. Compounds 23 and 24 caused significant mitotic arrest in vivo, which was sustained for 24 h. This would be followed by cell death either in mitosis or in the subsequent interphase. Furthermore, photoaffinity labeling experiments reveal that this series of inhibitors binds at the interface of KIF18A and tubulin. This study represents the first disclosure of KIF18A inhibitors with in vivo activity.


Subject(s)
Kinesins , Neoplasms , Cell Death , Humans , Mitosis , Neoplasms/drug therapy , Neoplasms/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
4.
Mol Cancer Ther ; 17(12): 2575-2585, 2018 12.
Article in English | MEDLINE | ID: mdl-30266802

ABSTRACT

Aurora kinase A and B have essential and non-overlapping roles in mitosis, with elevated expression in a subset of human cancers, including acute myeloid leukemia (AML). In this study, pan-aurora kinase inhibitor (AKI) AMG 900 distinguishes itself as an anti-leukemic agent that is more uniformly potent against a panel of AML cell lines than are isoform-selective AKIs and classic AML drugs. AMG 900 inhibited AML cell growth by inducing polyploidization and/or apoptosis. AMG 900 and aurora-B-selective inhibitor AZD1152-hQPA showed comparable cellular effects on AML lines that do not harbor a FLT3-ITD mutation. AMG 900 was active against P-glycoprotein-expressing AML cells resistant to AZD1152-hQPA and was effective at inducing expression of megakaryocyte-lineage markers (CD41, CD42) on human CHRF-288-11 cells and mouse Jak2 V617F cells. In MOLM-13 cells, inhibition of p-histone H3 by AMG 900 was associated with polyploidy, extra centrosomes, accumulation of p53 protein, apoptosis, and cleavage of Bcl-2 protein. Co-administration of cytarabine (Ara-C) with AMG 900 potentiated cell killing in a subset of AML lines, with evidence of attenuated polyploidization. AMG 900 inhibited the proliferation of primary human bone marrow cells in culture, with a better proliferation recovery profile relative to classic antimitotic drug docetaxel. In vivo, AMG 900 significantly reduced tumor burden in a systemic MOLM-13 xenograft model where we demonstrate the utility of 3'-deoxy-3'-18F-fluorothymidine [18F]FLT positron emission tomographic (PET)-CT imaging to measure the antiproliferative effects of AMG 900 in skeletal tissues in mice.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Mitosis/drug effects , Phthalazines/pharmacology , Animals , Apoptosis/drug effects , Aurora Kinases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase/drug effects , Humans , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Organophosphates/pharmacology , Polyploidy , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolines/pharmacology , Tumor Burden , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
J Med Chem ; 58(13): 5189-207, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25970324

ABSTRACT

Efforts to improve upon the physical properties and metabolic stability of Aurora kinase inhibitor 14a revealed that potency against multidrug-resistant cell lines was compromised by increased polarity. Despite its high in vitro metabolic intrinsic clearance, 23r (AMG 900) showed acceptable pharmacokinetic properties and robust pharmacodynamic activity. Projecting from in vitro data to in vivo target coverage was not practical due to disjunctions between enzyme and cell data, complex and apparently contradictory indicators of binding kinetics, and unmeasurable free fraction in plasma. In contrast, it was straightforward to relate pharmacokinetics to pharmacodynamics and efficacy by following the time above a threshold concentration. On the basis of its oral route of administration, a selectivity profile that favors Aurora-driven pharmacology and its activity against multidrug-resistant cell lines, 23r was identified as a potential best-in-class Aurora kinase inhibitor. In phase 1 dose expansion studies with G-CSF support, 23r has shown promising single agent activity.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Drug Discovery , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms/enzymology , Neoplasms/pathology , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
J Transl Med ; 12: 307, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25367255

ABSTRACT

BACKGROUND: The Aurora family of serine-threonine kinases are essential regulators of cell division in mammalian cells. Aurora-A and -B expression and kinase activity is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis. AMG 900 is a highly potent and selective pan-aurora kinase inhibitor that has entered clinical evaluation in adult patients with advanced cancers. In mice, oral administration of AMG 900 blocks the phosphorylation of histone H3 on serine-10 (p-Histone H3), a proximal substrate of aurora-B and inhibits the growth of multiple human tumor xenografts, including multidrug-resistant models. METHODS: In order to establish a preclinical pharmacokinetic-pharmacodynamic (PK-PD) relationship for AMG 900 that could be translated to the clinic, we used flow cytometry and laser scanning cytometry detection platforms to assess the effects on p-Histone H3 inhibition in terms of sensitivity, precision, and specificity, in human tumor xenografts in conjunction with mouse skin and bone marrow tissues. Mice with established COLO 205 tumors were administered AMG 900 at 3.75, 7.5, and 15 mg/kg and assessed after 3 hours. RESULTS: Significant suppression of p-Histone H3 in mouse skin was only observed at 15 mg/kg (p <0.0001), whereas in mouse bone marrow and in tumor a dose-dependent inhibition was achieved at all three doses (p ≤ 0.00015). These studies demonstrate that AMG 900 inhibits p-Histone H3 in tumors and surrogate tissues (although tissues such as skin may be less sensitive for assessing PD effects). To further extend our work, we evaluated the feasibility of measuring p-Histone H3 using fine-needle aspirate (FNA) tumor xenograft biopsies. Treatment with AMG 900 significantly inhibited p-Histone H3 (>99% inhibition, p <0.0001) in COLO 205 tumors. Lastly, we illustrate this LSC-based approach can detect p-Histone H3 positive cells using mock FNAs from primary human breast tumor tissues. CONCLUSION: Phosphorylation of histone H3 is a useful biomarker to determine the pharmacodynamics (PD) activity of AMG 900. FNA biopsies may be a viable approach for assessing AMG 900 PD effects in the clinic.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Histones/metabolism , Organ Specificity/drug effects , Phthalazines/pharmacology , Xenograft Model Antitumor Assays , Adult , Animals , Aurora Kinases/metabolism , Biopsy, Fine-Needle , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice, Nude , Phosphorylation/drug effects , Phthalazines/blood
7.
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
8.
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
9.
Mol Cancer Ther ; 12(11): 2356-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990115

ABSTRACT

Breast cancer is the most prevalent malignancy affecting women and ranks second in cancer-related deaths, in which death occurs primarily from metastatic disease. Triple-negative breast cancer (TNBC) is a more aggressive and metastatic subtype of breast cancer that is initially responsive to treatment of microtubule-targeting agents (MTA) such as taxanes. Recently, we reported the characterization of AMG 900, an orally bioavailable, potent, and highly selective pan-Aurora kinase inhibitor that is active in multidrug-resistant cell lines. In this report, we investigate the activity of AMG 900 alone and in combination with two distinct classes of MTAs (taxanes and epothilones) in multidrug-resistant TNBC cell lines and xenografts. In TNBC cells, AMG 900 inhibited phosphorylation of histone H3 on Ser(10), a proximal substrate of Aurora-B, and induced polyploidy and apoptosis. Furthermore, AMG 900 potentiated the antiproliferative effects of paclitaxel and ixabepilone at low nanomolar concentrations. In mice, AMG 900 significantly inhibited the growth of MDA-MB-231 (F(11); parental), MDA-MB-231 (F(11)) PTX-r (paclitaxel-resistant variant), and DU4475 xenografts. The combination of AMG 900 with docetaxel enhanced tumor inhibition in MDA-MB-231 (F(11)) xenografts compared with either monotherapy. Notably, combining AMG 900 with ixabepilone resulted in regressions of MDA-MB-231 (F(11)) PTX-r xenografts, in which more than 50% of the tumors failed to regrow 75 days after the cessation of drug treatment. These findings suggest that AMG 900, alone and in combination with MTAs, may be an effective intervention strategy for the treatment of metastatic breast cancer and provide potential therapeutic options for patients with multidrug-resistant tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Neoplasm Metastasis/pathology , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols , Aurora Kinases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Epothilones/pharmacology , Female , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Paclitaxel/pharmacology , Phosphorylation/drug effects , Polyploidy , Triple Negative Breast Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
10.
Cancer Res ; 70(23): 9846-54, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20935223

ABSTRACT

In mammalian cells, the aurora kinases (aurora-A, -B, and -C) play essential roles in regulating cell division. The expression of aurora-A and -B is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis, making them attractive targets for anticancer therapy. AMG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser(10), a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adult , Animals , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasms/enzymology , Neoplasms/pathology , Organophosphates/pharmacology , Paclitaxel/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Quinazolines/pharmacology , Xenograft Model Antitumor Assays
11.
J Med Chem ; 53(17): 6368-77, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20684549

ABSTRACT

The discovery of aurora kinases as essential regulators of cell division has led to intense interest in identifying small molecule aurora kinase inhibitors for the potential treatment of cancer. A high-throughput screening effort identified pyridinyl-pyrimidine 6a as a moderately potent dual inhibitor of aurora kinases -A and -B. Optimization of this hit resulted in an anthranilamide lead (6j) that possessed improved enzyme and cellular activity and exhibited a high level of kinase selectivity. However, this anthranilamide and subsequent analogues suffered from a lack of oral bioavailability. Converting the internally hydrogen-bonded six-membered pseudo-ring of the anthranilamide to a phthalazine (8a-b) led to a dramatic improvement in oral bioavailability (38-61%F) while maintaining the potency and selectivity characteristics of the anthranilamide series. In a COLO 205 tumor pharmacodynamic assay measuring phosphorylation of the aurora-B substrate histone H3 at serine 10 (p-histone H3), oral administration of 8b at 50 mg/kg demonstrated significant reduction in tumor p-histone H3 for at least 6 h.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phthalazines/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , Biological Availability , Blood Proteins/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Histones/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Phthalazines/pharmacokinetics , Phthalazines/pharmacology , Protein Binding , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Transplantation, Heterologous
12.
Cancer Res ; 66(8): 4299-308, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618755

ABSTRACT

In eukaryotic cells, cyclin-dependent kinase (CDK) complexes regulate the temporal progression of cells through the cell cycle. Deregulation in the cell cycle is an essential component in the evolution of cancer. Here, we validate CDK1 and CDK2 as potential therapeutic targets using novel selective small-molecule inhibitors of cyclin B1/CDK1 and cyclin E2/CDK2 enzyme complexes (CDKi). Flow cytometry-based methods were developed to assess intracellular retinoblastoma (Rb) phosphorylation to show inhibition of the CDK pathway. Tumor cells treated with CDK inhibitors showed an overall decrease in cell proliferation, accumulation of cells in G1 and G2, and apoptosis in a cell line-specific manner. Although CDK inhibitors activate p53, the inhibitors were equipotent in arresting the cell cycle in isogenic breast and colon tumor cells lacking p53, suggesting the response is independent of p53. In vivo, the CDK inhibitors prevented the growth of colon and prostate tumors, blocked proliferation of tumor cells, and inhibited Rb phosphorylation. The discovery and evaluation of novel potent and selective CDK1 and CDK2 inhibitors will help delineate the role that CDK complexes play in regulating tumorigenesis.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclin B/antagonists & inhibitors , Cyclin B/metabolism , Cyclin B1 , Cyclin E/antagonists & inhibitors , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Female , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Mice , Mice, Nude , Neoplasms/pathology , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Xenograft Model Antitumor Assays
13.
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
14.
Oncogene ; 21(55): 8529-34, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12466974

ABSTRACT

The increased expression of G(1) cyclins has been associated with the many types of human tumors. In primary solid tumors however, the expression and activity of cyclin E2, the newest member of the G(1) cyclin family, is largely unknown. In this study we have analysed the expression of the E-type cyclins in primary solid tumors from breast, lung, uterus, ovary, colon, and rectal tissues. Relative gene expression was analysed by quantitative real-time reverse transcription polymerase chain reaction (Taqman). The levels of cyclin E1 and cyclin E2 were significantly elevated (23 vs 38%, respectively) in primary breast tumor samples relative to normal breast tissue controls. We also observed an inverse correlation between the expression of cyclin E1/E2 and estrogen receptor in breast tumors. Our results demonstrate that the expression and associated catalytic activity for both cyclin E1 and cyclin E2 is elevated in primary breast tumors when compared to normal breast tissue. The increased level of cyclin E2 in breast tumors suggests that, similar to cyclin E1, it may contribute to the pathogenesis of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Colorectal Neoplasms , Cyclin E , Female , Gene Expression Regulation, Enzymologic , Genital Neoplasms, Female , Humans , Lung Neoplasms , Organ Specificity , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Int J Biochem Cell Biol ; 34(4): 315-20, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11854029

ABSTRACT

The eukaryotic cell cycle is regulated by a family of serine/threonine protein kinases known as cyclin-dependent kinases (CDKs). The activation of a CDK is dependent on its association with a cyclin regulatory subunit. The formation of distinct cyclin-CDK complexes controls the progression through the first gap phase (G(1)) and initiation of DNA synthesis (S phase). These complexes are in turn regulated by protein phosphorylation and cyclin-dependent kinase inhibitors (CKIs). Cyclin E2 has emerged as the second member of the E-type cyclin family. Cyclin E2-associated kinase activity is regulated in a cell cycle dependent manner with peak activity at the G(1) to S transition. Ectopic expression of cyclin E2 in human cells accelerates G(1), suggesting that cyclin E2 is rate limiting for G(1) progression. Although the pattern and level of cyclin E2 expression in some primary tumor and normal tissue RNAs are distinct from cyclin E1, both E-type cyclins appear to have inherent functional redundancies. This functional redundancy has facilitated the rapid characterization of cyclin E2 and uncovered unique features associated with each E-type cyclin.


Subject(s)
Cell Cycle , Cyclins/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Conserved Sequence , Cyclin-Dependent Kinases/physiology , Cyclins/chemistry , Forecasting , Humans , Molecular Sequence Data , Neoplasms/etiology , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...