Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Thorac Oncol ; 11(10): 1736-44, 2016 10.
Article in English | MEDLINE | ID: mdl-27448761

ABSTRACT

INTRODUCTION: A randomized phase 2 study was designed to compare the combination of ficlatuzumab (AV-299), a humanized hepatocyte growth factor-neutralizing monoclonal antibody, plus gefitinib versus gefitinib monotherapy in a pulmonary adenocarcinoma population clinically enriched for EFGR tyrosine kinase inhibitor-sensitizing mutations. METHODS: A total of 188 patients were randomized 1:1 to receive either gefitinib or ficlatuzumab plus gefitinib treatment. Patients who demonstrated disease control in the single-agent gefitinib arm were allowed to cross over to ficlatuzumab plus gefitinib treatment upon disease progression. Molecular analyses included tumor EGFR mutation status and retrospective proteomic testing using VeriStrat, a multivariate test based on mass spectrometry. RESULTS: The addition of ficlatuzumab to gefitinib did not provide significant improvement over gefitinib monotherapy for the primary end point of overall response rate or the secondary end points of progression-free survival and overall survival. In the subgroup classified as VeriStrat poor, the addition of ficlatuzumab to gefitinib showed significant improvement in both progression-free survival and overall survival in both the intent-to-treat population and the subgroup with EGFR tyrosine kinase inhibitor-sensitizing mutations. For all patients, the most frequent adverse events were diarrhea, dermatitis acneiform, and paronychia. CONCLUSIONS: Although the trial showed no significant benefit from the addition of ficlatuzumab to gefitinib in the overall population of Asian patients with advanced-stage pulmonary adenocarcinoma, the biomarker data suggest that patients classified as VeriStrat poor may benefit from ficlatuzumab combination therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Asian People , Disease-Free Survival , Female , Gefitinib , Humans , Lung Neoplasms/pathology , Male , Neoplasm Staging
2.
Cancer Res ; 76(16): 4841-4849, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27287719

ABSTRACT

Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) suppress normal hematopoietic activity in part by enabling a pathogenic inflammatory milieu in the bone marrow. In this report, we show that elevation of angiopoietin-1 in myelodysplastic CD34(+) stem-like cells is associated with higher risk disease and reduced overall survival in MDS and AML patients. Increased angiopoietin-1 expression was associated with a transcriptomic signature similar to known MDS/AML stem-like cell profiles. In seeking a small-molecule inhibitor of this pathway, we discovered and validated pexmetinib (ARRY-614), an inhibitor of the angiopoietin-1 receptor Tie-2, which was also found to inhibit the proinflammatory kinase p38 MAPK (which is overactivated in MDS). Pexmetinib inhibited leukemic proliferation, prevented activation of downstream effector kinases, and abrogated the effects of TNFα on healthy hematopoietic stem cells. Notably, treatment of primary MDS specimens with this compound stimulated hematopoiesis. Our results provide preclinical proof of concept for pexmetinib as a Tie-2/p38 MAPK dual inhibitor applicable to the treatment of MDS/AML. Cancer Res; 76(16); 4841-9. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Indazoles/pharmacology , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/pathology , Receptor, TIE-2/antagonists & inhibitors , Urea/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Angiopoietin-1/metabolism , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Gene Knockdown Techniques , Humans , Male , Mice , Proportional Hazards Models , Urea/pharmacology
3.
Cancer Biol Ther ; 12(9): 788-96, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21892012

ABSTRACT

Inhibition of the checkpoint kinase Chk1, both as a monotherapy and in combination with DNA damaging cytotoxics, is a promising therapeutic approach for the treatment of a wide array of human cancers. However, much remains to be elucidated in regard to the patient populations that will respond best to a Chk1 inhibitor and the optimal therapeutics to combine with a Chk1 inhibitor. In an effort to discover sensitizing mutations and novel combination strategies for Chk1 inhibition, an siRNA screen was performed in combination with the selective Chk1 inhibitor AR458323. This screen employed a custom made library of siRNAs targeting 195 genes, most of which are involved in cell-cycle control or DNA damage repair. One of the most prominent and consistent hits across runs of the screen performed in three different cancer cell lines was Wee1 kinase. MK-1775 is a small molecule inhibitor of Wee1 that is currently in early stage clinical trials. In confirmation of the results obtained from the siRNA screen, AR458323 and MK-1775 synergistically inhibited proliferation in multiple cancer cell types. This antiproliferative effect correlated with a synergistic induction of apoptosis. In cellular mechanistic studies, the combination of the two molecules resulted in dramatic decreases in inhibitory phosphorylation of cyclin-dependent kinases, an increase in DNA damage, alterations in cell-cycle profile, and collapse of DNA synthesis. In conclusion, the clinical combination of a Chk1 inhibitor and a Wee1 inhibitor holds promise as an effective treatment strategy for cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1 , DNA Replication/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Neoplasms/enzymology , Neoplasms/genetics , Nuclear Proteins/genetics , Phosphorylation/drug effects , Protein Kinases/genetics , Protein-Tyrosine Kinases/genetics , Pyrimidinones , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
5.
Genetics ; 168(3): 1159-76, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15579677

ABSTRACT

The retrovirus-like element Ty3 of Saccharomyces cerevisiae integrates at the transcription initiation region of RNA polymerase III. To identify host genes that affect transposition, a collection of insertion mutants was screened using a genetic assay in which insertion of Ty3 activates expression of a tRNA suppressor. Fifty-three loci were identified in this screen. Corresponding knockout mutants were tested for the ability to mobilize a galactose-inducible Ty3, marked with the HIS3 gene. Of 42 mutants tested, 22 had phenotypes similar to those displayed in the original assay. The proteins encoded by the defective genes are involved in chromatin dynamics, transcription, RNA processing, protein modification, cell cycle regulation, nuclear import, and unknown functions. These mutants were induced for Ty3 expression and assayed for Gag3p protein, integrase, cDNA, and Ty3 integration upstream of chromosomal tDNA(Val(AAC)) genes. Most mutants displayed differences from the wild type in one or more intermediates, although these were typically not as severe as the genetic defect. Because a relatively large number of genes affecting retrotransposition can be identified in yeast and because the majority of these genes have mammalian homologs, this approach provides an avenue for the identification of potential antiviral targets.


Subject(s)
Recombination, Genetic/physiology , Retroelements , Saccharomyces cerevisiae/genetics , Adaptor Proteins, Signal Transducing , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , DNA, Complementary , Mutation , RNA, Transfer, Val/genetics , RNA-Directed DNA Polymerase/genetics , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics
6.
J Biol Chem ; 279(34): 36059-71, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15218037

ABSTRACT

Efficient human immunodeficiency virus type 1 (HIV-1) budding requires an interaction between the PTAP late domain in the viral p6(Gag) protein and the cellular protein TSG101. In yeast, Vps23p/TSG101 binds both Vps28p and Vps37p to form the soluble ESCRT-I complex, which functions in sorting ubiquitylated protein cargoes into multivesicular bodies. Human cells also contain ESCRT-I, but the VPS37 component(s) have not been identified. Bioinformatics and yeast two-hybrid screening methods were therefore used to identify four novel human proteins (VPS37A-D) that share weak but significant sequence similarity with yeast Vps37p and to demonstrate that VPS37A and VPS37B bind TSG101. Detailed studies produced four lines of evidence that human VPS37B is a Vps37p ortholog. 1) TSG101 bound to several different sites on VPS37B, including a putative coiled-coil region and a PTAP motif. 2) TSG101 and VPS28 co-immunoprecipitated with VPS37B-FLAG, and the three proteins comigrated together in soluble complexes of the correct size for human ESCRT-I ( approximately 350 kDa). 3) Like TGS101, VPS37B became trapped on aberrant endosomal compartments in the presence of VPS4A proteins lacking ATPase activity. 4) Finally, VPS37B could recruit TSG101/ESCRT-I activity and thereby rescue the budding of both mutant Gag particles and HIV-1 viruses lacking native late domains. Further studies of ESCRT-I revealed that TSG101 mutations that inhibited PTAP or VPS28 binding blocked HIV-1 budding. Taken together, these experiments define new components of the human ESCRT-I complex and characterize several TSG101 protein/protein interactions required for HIV-1 budding and infectivity.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Products, gag/metabolism , HIV-1/physiology , Transcription Factors/metabolism , Binding Sites , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport , Gene Expression Regulation, Viral , HIV Infections/virology , Humans , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Vesicular Transport Proteins/genetics , Virus Replication
7.
J Cell Biol ; 162(3): 425-34, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12900394

ABSTRACT

The HIV-1 Gag protein recruits the cellular factor Tsg101 to facilitate the final stages of virus budding. A conserved P(S/T)AP tetrapeptide motif within Gag (the "late domain") binds directly to the NH2-terminal ubiquitin E2 variant (UEV) domain of Tsg101. In the cell, Tsg101 is required for biogenesis of vesicles that bud into the lumen of late endosomal compartments called multivesicular bodies (MVBs). However, the mechanism by which Tsg101 is recruited from the cytoplasm onto the endosomal membrane has not been known. Now, we report that Tsg101 binds the COOH-terminal region of the endosomal protein hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs; residues 222-777). This interaction is mediated, in part, by binding of the Tsg101 UEV domain to the Hrs 348PSAP351 motif. Importantly, Hrs222-777 can recruit Tsg101 and rescue the budding of virus-like Gag particles that are missing native late domains. These observations indicate that Hrs normally functions to recruit Tsg101 to the endosomal membrane. HIV-1 Gag apparently mimics this Hrs activity, and thereby usurps Tsg101 and other components of the MVB vesicle fission machinery to facilitate viral budding.


Subject(s)
DNA-Binding Proteins/metabolism , Eukaryotic Cells/virology , Gene Products, gag/metabolism , HIV/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Virus Replication/physiology , Virus Shedding/physiology , Cell Line , Endosomal Sorting Complexes Required for Transport , Endosomes/metabolism , Endosomes/ultrastructure , Endosomes/virology , Eukaryotic Cells/metabolism , HIV/pathogenicity , HIV/ultrastructure , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Microscopy, Electron , Molecular Mimicry/physiology , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Transport Vesicles/virology , gag Gene Products, Human Immunodeficiency Virus
8.
J Virol ; 77(9): 5439-50, 2003 May.
Article in English | MEDLINE | ID: mdl-12692245

ABSTRACT

The human immunodeficiency virus type 1 initially assembles and buds as an immature particle that is organized by the viral Gag polyprotein. Gag is then proteolyzed to produce the smaller capsid protein CA, which forms the central conical capsid that surrounds the RNA genome in the mature, infectious virus. To define CA surfaces that function at different stages of the viral life cycle, a total of 48 different alanine-scanning surface mutations in CA were tested for their effects on Gag protein expression, processing, particle production and morphology, capsid assembly, and infectivity. The 27 detrimental mutations fall into three classes: 13 mutations significantly diminished or altered particle production, 9 mutations failed to assemble normal capsids, and 5 mutations supported normal viral assembly but were nevertheless reduced more than 20-fold in infectivity. The locations of the assembly-defective mutations implicate three different CA surfaces in immature particle assembly: one surface encompasses helices 4 to 6 in the CA N-terminal domain (NTD), a second surrounds the crystallographically defined CA dimer interface in the C-terminal domain (CTD), and a third surrounds the loop preceding helix 8 at the base of the CTD. Mature capsid formation required a distinct surface encompassing helices 1 to 3 in the NTD, in good agreement with a recent structural model for the viral capsid. Finally, the identification of replication-defective mutants with normal viral assembly phenotypes indicates that CA also performs important nonstructural functions at early stages of the viral life cycle.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , HIV-1/metabolism , Amino Acid Sequence , Capsid/metabolism , Capsid Proteins/genetics , Cell Line , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HIV-1/genetics , HIV-1/pathogenicity , HeLa Cells , Humans , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Virion/genetics , Virion/metabolism , Virion/ultrastructure , Virus Assembly
9.
Trends Cell Biol ; 12(12): 569-79, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12495845

ABSTRACT

To spread infection, enveloped viruses must bud from infected host cells. Recent research indicates that HIV and other enveloped RNA viruses bud by appropriating the cellular machinery that is normally used to create vesicles that bud into late endosomal compartments called multivesicular bodies. This new model of virus budding has many potential implications for cell biology and viral pathogenesis.


Subject(s)
Cell Membrane/virology , RNA Viruses/growth & development , Virus Assembly , Cytoplasmic Vesicles/virology , HIV/growth & development , Humans , Molecular Mimicry , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL