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.
Oncologist ; 25(10): 833-e1438, 2020 10.
Article in English | MEDLINE | ID: mdl-32311798

ABSTRACT

LESSONS LEARNED: Despite strong preclinical rationale, combined cobimetinib-mediated MEK inhibition and GDC-0994-mediated ERK inhibition was not tolerable on two 28-day dosing schedules in which GDC-0994 was given for 21 days continuously and cobimetinib administered over 21 days either continuously or intermittently. Adverse events were as expected for mitogen-activated protein kinase pathway inhibition, but overlapping and cumulative toxicities could not be managed on either dosing schedule. Pharmacokinetic parameters of cobimetinib and GDC-0994 given in combination were similar to those previously observed in monotherapy studies, so that there was no evidence of drug-drug interaction. Cycle 1 metabolic responses were observed by 18F-fluorodeoxyglucose-positron emission tomography but were not predictive of outcome measured by RECIST 1.1. BACKGROUND: Simultaneous targeting of multiple nodes in the mitogen-activated protein kinase (MAPK) pathway offers the prospect of enhanced activity in RAS-RAF-mutant tumors. This phase Ib trial evaluated the combination of cobimetinib (MEK inhibitor) and GDC-0994 (ERK inhibitor) in patients with locally advanced or metastatic solid tumors. METHODS: Cobimetinib and GDC-0994 were administered orally on two separate dosing schedules. Arm A consisted of concurrent cobimetinib and GDC-0994 once daily for 21 days of a 28-day cycle; Arm B consisted of intermittent dosing of cobimetinib on a 28-day cycle concurrent with GDC-0994 daily for 21 days of a 28-day cycle. RESULTS: In total, 24 patients were enrolled. For Arm A, owing to cumulative grade 1-2 toxicity, the dose of cobimetinib was decreased. For Arm B, dose increases of GDC-0994 and cobimetinib were intolerable with grade 3 dose-limiting toxicities of myocardial infarction and rash. Pharmacokinetic data did not show evidence of a drug-drug interaction. Overall, seven patients had a best overall response of stable disease (SD) and one patient with pancreatic adenocarcinoma had an unconfirmed partial response. CONCLUSION: The safety profile of MEK and ERK inhibition demonstrated classic MAPK inhibitor-related adverse events (AEs). However, overlapping AEs and cumulative toxicity could not be adequately managed on either dosing schedule, restricting the ability to further develop this combination.


Subject(s)
Adenocarcinoma , Neoplasms , Pancreatic Neoplasms , Azetidines , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases , Neoplasms/drug therapy , Piperidines , Protein Kinase Inhibitors/adverse effects
2.
Clin Cancer Res ; 26(6): 1229-1236, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31848189

ABSTRACT

PURPOSE: ERK1/2 signaling can be dysregulated in cancer. GDC-0994 is an oral inhibitor of ERK1/2. A first-in-human, phase I dose escalation study of GDC-0994 was conducted in patients with locally advanced or metastatic solid tumors. PATIENTS AND METHODS: GDC-0994 was administered once daily on a 21-day on/7-day off schedule to evaluate safety, pharmacokinetics, and preliminary signs of efficacy. Patients with pancreatic adenocarcinoma and BRAF-mutant colorectal cancer were enrolled in the expansion stage. RESULTS: Forty-seven patients were enrolled in six successive cohorts (50-800 mg). A single DLT of grade 3 rash occurred at 600 mg. The most common drug-related adverse events (AE) were diarrhea, rash, nausea, fatigue, and vomiting. Pharmacokinetic data showed dose-proportional increases in exposure, with a mean half-life of 23 hours, supportive of once daily dosing. In evaluable paired biopsies, MAPK pathway inhibition ranged from 19% to 51%. Partial metabolic responses by FDG-PET were observed in 11 of 20 patients across dose levels in multiple tumor types. Overall, 15 of 45 (33%) patients had a best overall response of stable disease and 2 patients with BRAF-mutant colorectal cancer had a confirmed partial response. CONCLUSIONS: GDC-0994 had an acceptable safety profile and pharmacodynamic effects were observed by FDG-PET and in serial tumor biopsies. Single-agent activity was observed in 2 patients with BRAF-mutant colorectal cancer.


Subject(s)
MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Adult , Aged , Dose-Response Relationship, Drug , Fatigue/chemically induced , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Neoplasms/chemically induced , Neoplasms/pathology , Patient Safety , Tissue Distribution , Vomiting/chemically induced
3.
J Cell Sci ; 127(Pt 7): 1454-63, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24481816

ABSTRACT

Coatomer (COPI)-coated vesicles mediate membrane trafficking in the early secretory pathway. There are at least three subclasses of COPI coats and two classes of Arf GTPases that couple COPI coat proteins to membranes. Whether mechanisms exist to link specific Arfs to specific COPI subcomplexes is unknown. We now demonstrate that Scy1-like protein 1 (Scyl1), a member of the Scy1-like family of catalytically inactive protein kinases, oligomerizes through centrally located HEAT repeats and uses a C-terminal RKXX-COO(-) motif to interact directly with the appendage domain of coatomer subunit γ-2 (also known as COPG2 or γ2-COP). Through a distinct site, Scyl1 interacts selectively with class II Arfs, notably Arf4, thus linking class II Arfs to γ2-bearing COPI subcomplexes. Therefore, Scyl1 functions as a scaffold for key components of COPI coats, and disruption of the scaffolding function of Scyl1 causes tubulation of the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) and the cis-Golgi, similar to that observed following the loss of Arf and Arf-guanine-nucleotide-exchange factor (GEF) function. Our data reveal that Scyl1 is a key organizer of a subset of the COPI machinery.


Subject(s)
Coat Protein Complex I/metabolism , Coatomer Protein/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Binding Sites , DNA-Binding Proteins , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Protein Transport
4.
PLoS Biol ; 11(10): e1001670, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24130457

ABSTRACT

AP-2 is the core-organizing element in clathrin-mediated endocytosis. During the formation of clathrin-coated vesicles, clathrin and endocytic accessory proteins interact with AP-2 in a temporally and spatially controlled manner, yet it remains elusive as to how these interactions are regulated. Here, we demonstrate that the endocytic protein NECAP 1, which binds to the α-ear of AP-2 through a C-terminal WxxF motif, uses an N-terminal PH-like domain to compete with clathrin for access to the AP-2 ß2-linker, revealing a means to allow AP-2-mediated coordination of accessory protein recruitment and clathrin polymerization at sites of vesicle formation. Knockdown and functional rescue studies demonstrate that through these interactions, NECAP 1 and AP-2 cooperate to increase the probability of clathrin-coated vesicle formation and to control the number, size, and cargo content of the vesicles. Together, our data demonstrate that NECAP 1 modulates the AP-2 interactome and reveal a new layer of organizational control within the endocytic machinery.


Subject(s)
Adaptor Protein Complex 2/metabolism , Clathrin-Coated Vesicles/metabolism , Clathrin/metabolism , Endocytosis , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , COS Cells , Chlorocebus aethiops , Conserved Sequence , Gene Knockdown Techniques , HEK293 Cells , Humans , Membrane Proteins/chemistry , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Transport , Synaptic Vesicles/metabolism
5.
J Biol Chem ; 286(16): 13791-800, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21330364

ABSTRACT

The DENN domain is a common, evolutionarily ancient, and conserved protein module, yet it has gone largely unstudied; until recently, little was known regarding its functional roles. New studies reveal that various DENN domains interact directly with members of the Rab family of small GTPases and that DENN domains function enzymatically as Rab-specific guanine nucleotide exchange factors. Thus, DENN domain proteins appear to be generalized regulators of Rab function. Study of these proteins will provide new insights into Rab-mediated membrane trafficking pathways.


Subject(s)
rab GTP-Binding Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Endocytosis , Genome, Human , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction
6.
J Biol Chem ; 284(2): 774-83, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-18990695

ABSTRACT

Entry of the bacterial pathogen Listeria monocytogenes into host epithelial cells is critical for infection and virulence. One major pathway for Listeria entry involves binding of the bacterial protein Internalin B to the host receptor tyrosine kinase Met (hepatocyte growth factor receptor). Activation of Met and downstream signaling cascades is critical for Listeria entry. Internalin B is composed of several structural domains including an N-terminal leucine-rich repeat that is sufficient for binding Met and stimulating downstream signal transduction. Internalin B is monomeric, whereas the leucine-rich repeat is dimeric when expressed as an isolated fragment. The different quaternary states of Internalin B and the leucine-rich repeat suggest that these two Met ligands might cause distinct biological effects. Here we demonstrate that Internalin B and the leucine-rich repeat fragment exhibit agonist properties that differentially influence Met down-regulation in lysosomes. Specifically, Met stability is increased in response to the leucine-rich repeat fragment compared with Internalin B. Interestingly, Internalin B and the leucine-rich repeat stimulate equivalent rates of clathrin-mediated Met internalization. However, the leucine-rich repeat is defective in promoting lysosomal down-regulation of Met and instead enhances receptor recycling to the cell surface. In addition, the leucine-rich repeat causes prolonged Met activation (phosphorylation) and increased cell motility compared with Internalin B. Taken together, our findings indicate that individual domains of Internalin B differentially regulate Met trafficking. The ability of the leucine-rich repeat fragment to promote Met recycling could account for the increased cell motility induced by this ligand.


Subject(s)
Bacterial Proteins/metabolism , Leucine/metabolism , Listeria monocytogenes/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Cell Line , Clathrin/metabolism , Dogs , Endocytosis , Enzyme Activation/drug effects , Humans , Listeria monocytogenes/genetics , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics
7.
Cell Microbiol ; 9(10): 2497-516, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17848169

ABSTRACT

The intracellular bacterial pathogen Listeria monocytogenes causes food-borne illnesses leading to gastroenteritis, meningitis or abortion. Listeria induces its internalization into some mammalian cells through binding of the bacterial surface protein InlB to its host receptor, the Met Receptor Tyrosine Kinase. InlB-induced activation of Met stimulates host signal transduction pathways that culminate in cell surface changes driving pathogen engulfment. One mammalian protein with the potential to couple Met to downstream signalling is the adaptor CrkII. CrkII contains an unusual carboxyl-terminal SH3 domain (SH3C) that promotes entry of Listeria. However, binding partners or downstream effectors of SH3C remain unknown. Here, we use RNA interference and overexpression studies to demonstrate that SH3C affects bacterial uptake, at least in part, through stimulation of host phosphatidylinositide (PI) 3-kinase. Experiments with latex beads coated with InlB protein indicated that one potential role of SH3C and PI 3 kinase is to promote changes in the F-actin cytoskeleton necessary for particle engulfment. Taken together, our results indicate that the CrkII SH3C domain engages a cellular ligand that regulates PI 3 kinase activity and host cell surface rearrangements.


Subject(s)
Listeria monocytogenes/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-crk/physiology , src Homology Domains , Actins/physiology , Bacterial Proteins/metabolism , Cell Membrane/physiology , Chromones/pharmacology , Endocytosis , Enzyme Activation , HeLa Cells , Host-Pathogen Interactions , Humans , Membrane Proteins/metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
8.
Traffic ; 6(6): 459-73, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15882443

ABSTRACT

Increased hepatocyte growth factor receptor (HGFR) signaling correlates closely with neoplastic invasion and metastatic potential of many human cancers. Hepatocyte growth factor receptor signaling is initiated by binding the physiological ligand HGF or the internalin B (InlB) protein of Listeria monocytogenes. Subsequent degradation of endocytosed HGFR terminates receptor signaling. Previously reported discrepancies in InlB and HGF-induced HGFR signaling could reflect differences in receptor internalization and degradation in response to these distinct ligands. We report that soluble InlB and HGF are mechanistically equivalent in triggering clathrin-dependent endocytosis and lysosomal degradation of HGFR. After internalization, InlB and HGF colocalize with Rab5, EEA1 and the transferrin receptor in classical early endosomes. Hepatocyte growth factor receptor internalization was prevented by overexpression of dominant negative mutants of dynamin 1 and epidermal growth factor phosphorylation substrate 15, but not caveolin 1, the GTPase Arf6 or the cholesterol-chelating drug Nystatin. Thus, HGFR internalization is principally clathrin-mediated and is not regulated by clathrin- independent pathways. Phosphatidylinositol 3-kinase signaling and HGF-regulated tyrosine kinase substrate were not required for ligand-triggered internalization of HGFR but were essential for subsequent lysosomal degradation. Thus, soluble InlB and HGF induce HGFR endocytosis and degradation by indistinguishable mechanisms, suggesting that InlB may be exploited to regulate pathogenic HGFR signaling.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Hepatocyte Growth Factor/metabolism , Membrane Proteins/metabolism , Membrane Proteins/physiology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing , Biotin/chemistry , Biotinylation , Calcium-Binding Proteins/metabolism , Cell Line , Cell Line, Tumor , Cholesterol/metabolism , Clathrin/metabolism , Dynamin I/metabolism , Endocytosis , Endosomes/metabolism , GTP Phosphohydrolases/metabolism , Genes, Dominant , Humans , Intracellular Signaling Peptides and Proteins , Ligands , Listeria/metabolism , Lysosomes/metabolism , Membrane Microdomains/metabolism , Microscopy, Confocal , Nystatin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Plasmids/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Signal Transduction , Time Factors , Transfection
9.
Cell Microbiol ; 7(3): 443-57, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15679846

ABSTRACT

The bacterial surface protein InlB mediates internalization of Listeria monocytogenes into mammalian cells through interaction with the host receptor tyrosine kinase, Met. InlB/Met interaction results in activation of the host phosphoinositide (PI) 3-kinase p85-p110, an event required for bacterial entry. p85-p110 activation coincides with tyrosine phosphorylation of the host adaptor Gab1, and formation of complexes between Gab1 and the p85 regulatory subunit of PI 3-kinase. When phosphorylated in response to agonists, Gab1 is known to recruit several Src-homology 2 (SH2) domain-containing proteins including p85, the tyrosine phosphatase Shp2 and the adaptor CrkII. Here, we demonstrate that Gab1.p85 and Gab1.CrkII complexes promote entry of Listeria. Overexpression of wild-type Gab1 stimulated entry, whereas Gab1 alleles unable to recruit all SH2 proteins known to bind wild-type Gab1 inhibited internalization. Further analysis with Gab1 alleles defective in binding individual effectors suggested that recruitment of p85 and CrkII are critical for entry. Consistent with this data, overexpression of wild-type CrkII stimulated bacterial uptake. Experiments with mutant CrkII alleles indicated that both the first and second SH3 domains of this adaptor participate in entry, with the second domain playing the most critical role. Taken together, these findings demonstrate novel roles for Gab1 and CrkII in Listeria internalization.


Subject(s)
Bacterial Proteins/physiology , Listeria monocytogenes/physiology , Proto-Oncogene Proteins/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chlorocebus aethiops , Intracellular Signaling Peptides and Proteins/metabolism , Listeria monocytogenes/pathogenicity , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-crk , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , Vero Cells , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...