Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 14(2): 375-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487917

ABSTRACT

Resistance to FLT3 inhibitors is a serious clinical issue in treating acute myelogenous leukemia (AML). AMG 925, a dual FLT3/CDK4 inhibitor, has been developed to overcome this resistance. It is hypothesized that the combined inhibition of FLT3 and CDK4 may reduce occurrence of the FLT3 resistance mutations, and thereby prolong clinical responses. To test this hypothesis, we attempted to isolate AML cell clones resistant to AMG 925 or to FLT3 inhibitors. After a selection of over 8 months with AMG 925, we could only isolate partially resistant clones. No new mutations in FLT3 were found, but a 2- to 3-fold increase in total FLT3 protein was detected and believed to contribute to the partial resistance. In contrast, selection with the FLT3 inhibitors sorafenib or AC220 (Quizartinib), led to a resistance and the appearance of a number of mutations in FLT3 kinase domains, including the known hot spot sites D835 and F691. However, when AC220 was combined with the CDK4 inhibitor PD0332991 (palbociclib) at 0.1 µmol/L or higher, no resistance mutations were obtained, indicating that the CDK4-inhibiting activity of AMG 925 contributed to the failure to develop drug resistance. AMG 925 was shown to potently inhibit the FLT3 inhibitor-resistant mutation D835Y/V. This feature of AMG 925 was also considered to contribute to the lack of resistance mutations to the compound. Together, our data suggest that AMG 925 has the potential to reduce resistance mutations in FLT3 and may prolong clinical responses.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Leukemia, Myeloid, Acute/pathology , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Apoptosis/drug effects , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Clone Cells , Cyclin-Dependent Kinase 4/metabolism , Humans , Mutation/genetics , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/metabolism
2.
J Med Chem ; 57(8): 3430-49, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24641103

ABSTRACT

We describe the structural optimization of a lead compound 1 that exhibits dual inhibitory activities against FLT3 and CDK4. A series of pyrido[4',3':4,5]pyrrolo[2,3-d]pyrimidine derivatives was synthesized, and SAR analysis, using cell-based assays, led to the discovery of 28 (AMG 925), a potent and orally bioavailable dual inhibitor of CDK4 and FLT3, including many FLT3 mutants reported to date. Compound 28 inhibits the proliferation of a panel of human tumor cell lines including Colo205 (Rb(+)) and U937 (FLT3(WT)) and induced cell death in MOLM13 (FLT3(ITD)) and even in MOLM13 (FLT3(ITD, D835Y)), which exhibits resistance to a number of FLT3 inhibitors currently under clinical development. At well-tolerated doses, compound 28 leads to significant growth inhibition of MOLM13 xenografts in nude mice, and the activity correlates with inhibition of STAT5 and Rb phosphorylation.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/chemical synthesis , Naphthyridines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Dogs , Drug Discovery , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Macaca fascicularis , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats , Structure-Activity Relationship , U937 Cells , fms-Like Tyrosine Kinase 3/genetics
3.
Mol Cancer Ther ; 13(4): 880-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24526162

ABSTRACT

Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard-of-care treatment, the disease eventually relapses in a major proportion of patients. Activating Fms-like tyrosine kinase 3 (FLT3) mutations are found in approximately 30% of patients with AML. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here the preclinical evaluation of AMG 925, a potent, selective, and bioavailable FLT3/cyclin-dependent kinase 4 (CDK4) dual kinase inhibitor. AMG 925 inhibited AML xenograft tumor growth by 96% to 99% without significant body weight loss. The antitumor activity of AMG 925 correlated with the inhibition of STAT5 and RB phosphorylation, the pharmacodynamic markers for inhibition of FLT3 and CDK4, respectively. In addition, AMG 925 was also found to inhibit FLT3 mutants (e.g., D835Y) that are resistant to the current FLT3 inhibitors (e.g., AC220 and sorafenib). CDK4 is a cyclin D-dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4-RB pathway in cancer development has been well established. CDK4-specific inhibitors are being developed for treating RB-positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve and prolong clinical responses.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Naphthyridines/pharmacokinetics , Naphthyridines/therapeutic use , Neoplasms, Experimental , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacology , Signal Transduction/drug effects , Sorafenib , U937 Cells , Xenograft Model Antitumor Assays
4.
Discov Med ; 16(88): 149-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24099669

ABSTRACT

Cancer immunotherapy attempts to exploit the capability of the immune system to attack malignant cells. Recent results suggest that clinical responses in patients point to this new mechanism as potentially beneficial in harnessing the immune system for combating established malignancies. These checkpoint-related immunotherapies rely on engaging a subset of T cells in anti-tumor immune responses. BiTE® (Bi-specific T cell engager) represents a distinct modality that directly engages any T cell and a specific antigen expressing tumor cell. The approach offers the advantage of engaging T cells and patient tumor cells that differentially express a specific cell surface antigen. The specificity confers redirected tumor cell killing and recent clinical data with the BiTE blinatumomab show evidence of clinical remissions. The characteristics of a suitable BiTE with the benefit of CD3 mediated T cell recognition and articulation of tumor specific antigens combined in this therapeutic modality is described here.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Humans
5.
PLoS One ; 8(7): e68328, 2013.
Article in English | MEDLINE | ID: mdl-23861887

ABSTRACT

Sphingosine kinases (SPHKs) are enzymes that phosphorylate the lipid sphingosine, leading to the formation of sphingosine-1-phosphate (S1P). In addition to the well established role of extracellular S1P as a mitogen and potent chemoattractant, SPHK activity has been postulated to be an important intracellular regulator of apoptosis. According to the proposed rheostat theory, SPHK activity shifts the intracellular balance from the pro-apoptotic sphingolipids ceramide and sphingosine to the mitogenic S1P, thereby determining the susceptibility of a cell to apoptotic stress. Despite numerous publications with supporting evidence, a clear experimental confirmation of the impact of this mechanism on tumor cell viability in vitro and in vivo has been hampered by the lack of suitable tool reagents. Utilizing a structure based design approach, we developed potent and specific SPHK1/2 inhibitors. These compounds completely inhibited intracellular S1P production in human cells and attenuated vascular permeability in mice, but did not lead to reduced tumor cell growth in vitro or in vivo. In addition, siRNA experiments targeting either SPHK1 or SPHK2 in a large panel of cell lines failed to demonstrate any statistically significant effects on cell viability. These results show that the SPHK rheostat does not play a major role in tumor cell viability, and that SPHKs might not be attractive targets for pharmacological intervention in the area of oncology.


Subject(s)
Neoplasms/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Capillary Permeability/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Humans , Inhibitory Concentration 50 , Mice , Neoplasms/genetics , Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Interference , Tumor Burden/drug effects , Tumor Stem Cell Assay , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
6.
PLoS One ; 7(12): e50920, 2012.
Article in English | MEDLINE | ID: mdl-23300529

ABSTRACT

Endoglin (ENG), a co-receptor for several TGFß-family cytokines, is expressed in dividing endothelial cells alongside ALK1, the ACVRL1 gene product. ENG and ACVRL1 are both required for angiogenesis and mutations in either gene are associated with Hereditary Hemorrhagic Telangectasia, a rare genetic vascular disorder. ENG and ALK1 function in the same genetic pathway but the relative contribution of TGFß and BMP9 to SMAD1/5/8 activation and the requirement of ENG as a co-mediator of SMAD phosphorylation in endothelial cells remain debated. Here, we show that BMP9 and TGFß1 induce distinct SMAD phosphorylation responses in primary human endothelial cells and that, unlike BMP9, TGFß only induces SMAD1/5/8 phosphorylation in a subset of immortalized mouse endothelial cell lines, but not in primary human endothelial cells. We also demonstrate, using siRNA depletion of ENG and novel anti-ENG antibodies, that ENG is required for BMP9/pSMAD1 signaling in all human and mouse endothelial cells tested. Finally, anti-ENG antibodies that interfere with BMP9/pSMAD1 signaling, but not with TGFß1/pSMAD3 signaling, also decrease in vitro HUVEC endothelial tube formation and inhibit BMP9 binding to recombinant ENG in vitro. Our data demonstrate that BMP9 signaling inhibition is a key and previously unreported mechanism of action of TRC105, an anti-angiogenic anti-Endoglin antibody currently evaluated in clinical trials.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Growth Differentiation Factors/metabolism , Receptors, Cell Surface/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/genetics , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Endoglin , Endothelium, Vascular/cytology , Flow Cytometry , Growth Differentiation Factor 2 , Growth Differentiation Factors/genetics , Humans , Mice , Phosphorylation , Protein Binding , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins/genetics , Transforming Growth Factor beta/genetics
7.
Bioorg Med Chem Lett ; 21(18): 5206-9, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21840217

ABSTRACT

A bis-amide antagonist of Smoothened, a seven-transmembrane receptor in the Hedgehog signaling pathway, was discovered via high throughput screening. In vitro and in vivo experiments demonstrated that the bis-amide was susceptible to N-acyl transferase mediated amide scission. Several bioisosteric replacements of the labile amide that maintained in vitro potency were identified and shown to be metabolically stable in vitro and in vivo.


Subject(s)
Acyltransferases/antagonists & inhibitors , Amides/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Acyltransferases/metabolism , Amides/chemistry , Amides/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , High-Throughput Screening Assays , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 20(15): 4607-10, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20594845

ABSTRACT

Pyridopyridazine antagonists of the hedgehog signaling pathway are described. Designed to optimize our previously described phthalazine smoothened antagonists, a representative compound eliminates a PXR liability while retaining potency and in vitro metabolic stability. Moreover, the compound has improved efficacy in a hedgehog/smoothened signaling mouse pharmacodynamic model.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Phthalazines/chemistry , Piperazines/chemistry , Pyridazines/chemistry , Receptors, Steroid/chemistry , Animals , Hedgehog Proteins/metabolism , Humans , Mice , Microsomes, Liver/metabolism , Phthalazines/chemical synthesis , Phthalazines/pharmacokinetics , Piperazines/chemical synthesis , Piperazines/pharmacokinetics , Pregnane X Receptor , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Steroid/metabolism , Signal Transduction , Smoothened Receptor , Structure-Activity Relationship , Tylosin/analogs & derivatives
9.
Bioorg Med Chem Lett ; 20(12): 3618-22, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20493695

ABSTRACT

The Hedgehog (Hh) signaling pathway regulates cell proliferation and differentiation in developing tissues, and abnormal activation of the Hh pathway has been linked to several tumor subsets. As a transducer of Hh signaling, the GPCR-like protein Smoothened (Smo) is a promising target for disruption of unregulated Hh signaling. A series of 1-amino-4-arylphthalazines was developed as potent and orally bioavailable inhibitors of Smo. A representative compound from this class demonstrated significant tumor volume reduction in a mouse medulloblastoma model.


Subject(s)
Phthalazines/chemistry , Phthalazines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Cytochrome P-450 Enzyme System/drug effects , Drug Design , Hedgehog Proteins , Humans , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Mice , Phthalazines/chemical synthesis , Signal Transduction , Smoothened Receptor
10.
Bioorg Med Chem Lett ; 18(3): 1037-41, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18178086

ABSTRACT

The LPA(2) protein is overexpressed in many tumor cells. We report the optimization of a series of LPA(2) antagonists using calcium mobilization assay (aequorin assay) that led to the discovery of the first reported inhibitors selective for LPA(2). Key compounds were evaluated in vitro for inhibition of LPA(2) mediated Erk activation and proliferation of HCT-116 cells. These compounds could be used to evaluate the benefits of LPA(2) inhibition both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Combinatorial Chemistry Techniques , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Humans , Molecular Structure , Tumor Cells, Cultured
11.
Cancer Res ; 66(1): 283-9, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397241

ABSTRACT

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


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Deletion , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Receptors, Growth Factor/genetics , Alternative Splicing , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Down-Regulation , Enzyme Activation , Exons , Female , Humans , Introns , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/metabolism , Signal Transduction , Ubiquitin/metabolism
12.
Bioorg Med Chem Lett ; 16(6): 1716-20, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16384702

ABSTRACT

The syntheses of potent small molecule inhibitors of the CDK2/cyclinA recruitment site are described. Structure-activity trends of nanomolar octapeptides were examined through amino-acid substitution and truncation of the sequence resulting in the identification of a smaller, albeit significantly less potent, tetrapeptide lead. These losses in affinity were recovered by side-chain optimization and by rigidification of the peptide backbone using a combination of solid-phase parallel synthesis and structure-based design. Finally, two guanidine functionalities were replaced to improve drug-like properties, resulting in neutral small molecules equal in activity to that of the peptide lead.


Subject(s)
Cyclin A/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Peptide Fragments/pharmacology , Amino Acid Substitution , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Guanidine/chemistry , Humans , Models, Molecular , Peptide Fragments/chemistry , Protein Conformation
13.
Cell Cycle ; 4(5): 683-5, 2005 May.
Article in English | MEDLINE | ID: mdl-15846105

ABSTRACT

Ligand dependent activity of receptor tyrosine kinases is critical for modulating downstream signaling and cell proliferation. In normal cellular context, hepatocyte growth factor (HGF) regulates MET kinase activation and mediates cell proliferation, migration and motility. Recent elucidation of the MET extracellular domain suggests that the Sema domain, which bears structural similarity to other Semaphorins and Plexin family members, plays a critical role in ligand mediated receptor activation. Overexpression of MET which is observed in many cancers leads to ligand independent receptor dimerization and activation. Evidence to support a role for the Sema domain in cancer and therapeutic implications of targeting the Met Sema domain are discussed in this review.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation , Dimerization , Drug Delivery Systems , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic , Hepatocyte Growth Factor/metabolism , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/genetics , Semaphorins/chemistry
14.
Cancer Cell ; 6(1): 75-84, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15261143

ABSTRACT

Hepatocyte growth factor (HGF) binds the extracellular domain and activates the Met receptor to induce mitogenesis, morphogenesis, and motility. The extracellular domain of Met is comprised of Sema, PSI, and four IPT subdomains. We investigated the contribution of these subdomains to Met receptor dimerization. Our observations indicate that the Sema domain is necessary for dimerization in addition to HGF binding. Treatment of Met-overexpressing tumor cells with recombinant Sema in the presence or absence of HGF results in decreased Met-mediated signal transduction, cell motility, and migration, behaving in a manner similar to an antagonistic anti-Met Fab. These data suggest that the Sema domain of Met may not only represent a novel anticancer therapeutic target but also acts as a biotherapeutic itself.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Cross-Linking Reagents , Dimerization , Female , Hepatocyte Growth Factor/genetics , Humans , Mice , Mice, Nude , Precipitin Tests , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/immunology , Sequence Deletion , Tumor Cells, Cultured
15.
Cancer Res ; 63(5): 1020-4, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12615717

ABSTRACT

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


Subject(s)
Antineoplastic Agents/pharmacology , CDC2-CDC28 Kinases , Cyclin A/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3T3 Cells , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/physiology , Binding Sites , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , Cyclin D , Cyclin-Dependent Kinase 2 , Cyclins/physiology , Female , Genes, erbB-2/genetics , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Rats , Retinoblastoma Protein/physiology , Signal Transduction/physiology
16.
J Biol Chem ; 277(49): 47804-9, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12372819

ABSTRACT

Hepatocyte growth factor (HGF), the ligand for the receptor tyrosine kinase c-Met, is composed of an alpha-chain containing four Kringle domains (K1-K4) and a serine protease domain-like beta-chain. Receptor activation by HGF is contingent upon prior proteolytic conversion of the secreted inactive single chain form (pro-HGF) into the biologically active two chain form by a single cleavage at the Arg(494)-Val(495) bond. By screening a panel of serine proteases we identified two new HGF activators, plasma kallikrein and coagulation factor XIa (FXIa). The concentrations of kallikrein and FXIa to cleave 50% (EC(50)) of (125)I-labeled pro-HGF during a 4-h period were 10 and 17 nm. Unlike other known activators, both FXIa and kallikrein processed pro-HGF by cleavage at two sites. Using N-terminal sequencing they were identified as the normal cleavage site Arg(494)-Val(495) and the novel site Arg(424)-His(425) located in the K4 domain of the alpha-chain. The identity of this unusual second cleavage site was firmly established by use of the double mutant HGF(R424A/R494E), which was completely resistant to cleavage by kallikrein and FXIa. Experiments with another mutant form, HGF(Arg(494) --> Glu), indicated that cleavage at the K4 site was independent of a prior cleavage at the primary, kinetically preferred Arg(494)-Val(495) site. The cleavage at the K4 site had no obvious consequences on HGF function, because it was fully capable of phosphorylating the c-Met receptor of A549 cells. This may be explained by the disulfide bond network in K4, which holds the cleaved alpha-chain together. In conclusion, the ability of plasma kallikrein and FXIa to activate pro-HGF in vitro raises the possibility that mediators of inflammation and blood coagulation may also regulate processes that involve the HGF/c-Met pathway, such as tissue repair and angiogenesis.


Subject(s)
Factor XIa/chemistry , Hepatocyte Growth Factor/metabolism , Kallikreins/blood , Animals , Arginine/chemistry , Binding Sites , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Factor XIIa/antagonists & inhibitors , Factor XIa/antagonists & inhibitors , Factor XIa/metabolism , Histidine/chemistry , Humans , Kallikreins/antagonists & inhibitors , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/metabolism , Recombinant Proteins/metabolism , Time Factors , Tumor Cells, Cultured , Valine/chemistry
17.
Cancer Res ; 62(19): 5485-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12359757

ABSTRACT

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


Subject(s)
Antibodies, Monoclonal/pharmacology , Prostatic Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Androgens/physiology , Animals , Cell Division/drug effects , Dose-Response Relationship, Immunologic , Female , Humans , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/therapy , Neuregulin-1/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Recombinant Proteins/pharmacology , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Nat Genet ; 30(4): 446-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11912493

ABSTRACT

In a wide variety of animal species, oocyte maturation is arrested temporarily at prophase of meiosis I (ref. 1). Resumption of meiosis requires activation of cyclin-dependent kinase-1 (CDK1, p34cdc2), one component of maturation-promoting factor (MPF). The dual specificity phosphatases Cdc25a, Cdc25b and Cdc25c are activators of cyclin-dependent kinases; consequently, they are postulated to regulate cell-cycle progression in meiosis and mitosis as well as the DNA-damage response. We generated Cdc25b-deficient (Cdc25b-/-) mice and found that they are viable. As compared with wildtype cells, fibroblasts from Cdc25b-/- mice grew vigorously in culture and arrested normally in response to DNA damage. Female Cdc25b-/- mice were sterile, and Cdc25b-/- oocytes remained arrested at prophase with low MPF activity. Microinjection of wildtype Cdc25b mRNA into Cdc25b-/- oocytes caused activation of MPF and resumption of meiosis. Thus, Cdc25b-/- female mice are sterile because of permanent meiotic arrest resulting from the inability to activate MPF. Cdc25b is therefore essential for meiotic resumption in female mice. Mice lacking Cdc25b provide the first genetic model for studying the mechanisms regulating prophase arrest in vertebrates.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Meiosis , Oocytes/physiology , cdc25 Phosphatases/genetics , cdc25 Phosphatases/physiology , Animals , Blotting, Southern , Blotting, Western , Cells, Cultured , DNA Damage , Female , MAP Kinase Signaling System , Male , Mesothelin , Mice , Microscopy, Fluorescence , Mitosis , Models, Genetic , Oocytes/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...