Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(3): 1734-1746, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38267212

ABSTRACT

Fibroblast growth factor receptor (FGFR) alterations are present as oncogenic drivers and bypass mechanisms in many forms of cancer. These alterations can include fusions, amplifications, rearrangements, and mutations. Acquired drug resistance to current FGFR inhibitors often results in disease progression and unfavorable outcomes for patients. Genomic profiling of tumors refractory to current FGFR inhibitors in the clinic has revealed several acquired driver alterations that could be the target of next generation therapeutics. Herein, we describe how structure-based drug design (SBDD) was used to enable the discovery of the potent and kinome selective pan-FGFR inhibitor KIN-3248, which is active against many acquired resistance mutations. KIN-3248 is currently in phase I clinical development for the treatment of advanced tumors harboring FGFR2 and/or FGFR3 gene alterations.


Subject(s)
Neoplasms , Receptor, Fibroblast Growth Factor, Type 2 , Humans , Receptor, Fibroblast Growth Factor, Type 2/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Mutation , Disease Progression , Protein Kinase Inhibitors/adverse effects , Receptor, Fibroblast Growth Factor, Type 3
2.
J Med Chem ; 67(3): 1747-1757, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38230963

ABSTRACT

RAF, a core signaling component of the MAPK kinase cascade, is often mutated in various cancers, including melanoma, lung, and colorectal cancers. The approved inhibitors were focused on targeting the BRAFV600E mutation that results in constitutive activation of kinase signaling through the monomeric protein (Class I). However, these inhibitors also paradoxically activate kinase signaling of RAF dimers, resulting in increased MAPK signaling in normal tissues. Recently, significant attention has turned to targeting RAF alterations that activate dimeric signaling (class II and III BRAF and NRAS). However, the discovery of a potent and selective inhibitor with biopharmaceutical properties suitable to sustain robust target inhibition in the clinical setting has proven challenging. Herein, we report the discovery of exarafenib (15), a highly potent and selective inhibitor that intercepts the RAF protein in the dimer compatible αC-helix-IN conformation and demonstrates anti-tumor efficacy in preclinical models with BRAF class I, II, and III and NRAS alterations.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor , Melanoma/pathology , MAP Kinase Signaling System , Mutation
3.
PLoS One ; 7(5): e36753, 2012.
Article in English | MEDLINE | ID: mdl-22586492

ABSTRACT

Receptor tyrosine kinases and integrins play an essential role in tumor cell invasion and metastasis. We previously showed that EGF and other growth factors induce human carcinoma cell invasion and metastasis mediated by integrin αvß5 that is prevented by Src blockade. MUC1, a transmembrane glycoprotein, is expressed in most epithelial tumors as a heterodimer consisting of an extracellular and a transmembrane subunit. The MUC1 cytoplasmic domain of the transmembrane subunit (MUC1.CD) translocates to the nucleus where it promotes the transcription of a metastatic gene signature associated with epithelial to mesenchymal transition. Here, we demonstrate a requirement for MUC1 in carcinoma cell metastasis dependent on EGFR and Src without affecting primary tumor growth. EGF stimulates Src-dependent MUC1 cleavage and nuclear localization leading to the expression of genes linked to metastasis. Moreover, expression of MUC1.CD results in its nuclear localization and is sufficient for transcription of the metastatic gene signature and tumor cell metastasis. These results demonstrate that EGFR and Src activity contribute to carcinoma cell invasion and metastasis mediated by integrin αvß5 in part by promoting proteolytic cleavage of MUC1 and highlight the ability of MUC1.CD to promote metastasis in a context-dependent manner. Our findings may have implications for the use and future design of targeted therapies in cancers known to express EGFR, Src, or MUC1.


Subject(s)
Epidermal Growth Factor , ErbB Receptors , Mucin-1 , Neoplasm Invasiveness/genetics , Protein-Tyrosine Kinases , Receptors, Vitronectin , Animals , CSK Tyrosine-Protein Kinase , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Chick Embryo , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mucin-1/genetics , Mucin-1/metabolism , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Signal Transduction , src-Family Kinases
4.
Blood ; 119(9): 2149-58, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22134168

ABSTRACT

Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvß3 on VSMCs. Once expressed, integrin αvß3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvß3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Integrins/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Notch/metabolism , Animals , Arteries/metabolism , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Integrins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Protein Binding , RNA, Messenger/metabolism , Receptors, Notch/genetics , Serrate-Jagged Proteins , Signal Transduction/physiology , von Willebrand Factor/metabolism
5.
Nat Med ; 17(12): 1641-5, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081024

ABSTRACT

RAF kinases regulate cell proliferation and survival and can be dysregulated in tumors. The role of RAF in cell proliferation has been linked to its ability to activate mitogen-activated protein kinase kinase 1 (MEK) and mitogen-activated protein kinase 1 (ERK). Here we identify a MEK-independent role for RAF in tumor growth. Specifically, in mitotic cells, CRAF becomes phosphorylated on Ser338 and localizes to the mitotic spindle of proliferating tumor cells in vitro as well as in murine tumor models and in biopsies from individuals with cancer. Treatment of tumors with allosteric inhibitors, but not ATP-competitive RAF inhibitors, prevents CRAF phosphorylation on Ser338 and localization to the mitotic spindle and causes cell-cycle arrest at prometaphase. Furthermore, we identify phospho-Ser338 CRAF as a potential biomarker for tumor progression and a surrogate marker for allosteric RAF blockade. Mechanistically, CRAF, but not BRAF, associates with Aurora kinase A (Aurora-A) and Polo-like kinase 1 (Plk1) at the centrosomes and spindle poles during G2/M. Indeed, allosteric or genetic inhibition of phospho-Ser338 CRAF impairs Plk1 activation and accumulation at the kinetochores, causing prometaphase arrest, whereas a phospho-mimetic Ser338D CRAF mutant potentiates Plk1 activation, mitosis and tumor progression in mice. These findings show a previously undefined role for RAF in tumor progression beyond the RAF-MEK-ERK paradigm, opening new avenues for targeting RAF in cancer.


Subject(s)
MAP Kinase Kinase 1/metabolism , Mitosis , Neoplasms/pathology , Proto-Oncogene Proteins c-raf/metabolism , Animals , Aurora Kinase A , Aurora Kinases , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Centrosome/metabolism , Disease Models, Animal , Female , Humans , Kinetochores/metabolism , MAP Kinase Kinase 1/genetics , Mice , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction , Spindle Apparatus/metabolism , Polo-Like Kinase 1
6.
Mol Cancer Ther ; 10(6): 972-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21518727

ABSTRACT

Although nanoparticle-based drug delivery formulations can improve the effectiveness and safety of certain anticancer drugs, many drugs, due to their chemical composition, are unsuitable for nanoparticle loading. Here, we describe a targeted nanogel drug delivery platform that can (i) encapsulate a wide range of drug chemotypes, including biological, small molecule, and cytotoxic agents; (ii) display targeting ligands and polymeric coatings on the surface; (iii) enhance drug retention within the nanogel core after photo-cross-linking; and (iv) retain therapeutic activity after lyophilization allowing for long-term storage. For therapeutic studies, we used integrin αvß3-targeted lipid-coated nanogels with cross-linked human serum albumin in the core for carrying therapeutic cargoes. These particles exhibited potent activity in tumor cell viability assays with drugs of distinct chemotype, including paclitaxel, docetaxel, bortezomib, 17-AAG, sorafenib, sunitinib, bosutinib, and dasatinib. Treatment of orthotopic breast and pancreas tumors in mice with taxane-loaded nanogels produced a 15-fold improvement in antitumor activity relative to Abraxane by blocking both primary tumor growth and spontaneous metastasis. With a modifiable surface and core, the lipid-coated nanogel represents a platform technology that can be easily adapted for specific drug delivery applications to treat a wide range of malignant diseases.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Pancreatic Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethyleneimine/administration & dosage , Polyethyleneimine/chemistry , Albumin-Bound Paclitaxel , Albumins/administration & dosage , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Freeze Drying/methods , Humans , Integrin alphaVbeta3/metabolism , Lipids/chemistry , Mice , Mice, Nude , Nanogels , Paclitaxel/administration & dosage , Pancreatic Neoplasms/metabolism , Polyethylene Glycols/chemical synthesis , Polyethyleneimine/chemical synthesis , Polymers/chemistry , Taxoids/administration & dosage
7.
Nat Med ; 16(8): 909-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20676106

ABSTRACT

Although it is well established that tumors initiate an angiogenic switch, the molecular basis of this process remains incompletely understood. Here we show that the miRNA miR-132 acts as an angiogenic switch by targeting p120RasGAP in the endothelium and thereby inducing neovascularization. We identified miR-132 as a highly upregulated miRNA in a human embryonic stem cell model of vasculogenesis and found that miR-132 was highly expressed in the endothelium of human tumors and hemangiomas but was undetectable in normal endothelium. Ectopic expression of miR-132 in endothelial cells in vitro increased their proliferation and tube-forming capacity, whereas intraocular injection of an antagomir targeting miR-132, anti-miR-132, reduced postnatal retinal vascular development in mice. Among the top-ranking predicted targets of miR-132 was p120RasGAP, which we found to be expressed in normal but not tumor endothelium. Endothelial expression of miR-132 suppressed p120RasGAP expression and increased Ras activity, whereas a miRNA-resistant version of p120RasGAP reversed the vascular response induced by miR-132. Notably, administration of anti-miR-132 inhibited angiogenesis in wild-type mice but not in mice with an inducible deletion of Rasa1 (encoding p120RasGAP). Finally, vessel-targeted nanoparticle delivery of anti-miR-132 restored p120RasGAP expression in the tumor endothelium, suppressed angiogenesis and decreased tumor burden in an orthotopic xenograft mouse model of human breast carcinoma. We conclude that miR-132 acts as an angiogenic switch by suppressing endothelial p120RasGAP expression, leading to Ras activation and the induction of neovascularization, whereas the application of anti-miR-132 inhibits neovascularization by maintaining vessels in the resting state.


Subject(s)
Endothelium, Vascular/pathology , MicroRNAs/physiology , Neovascularization, Pathologic/genetics , p120 GTPase Activating Protein/genetics , Animals , Antibodies, Monoclonal/pharmacology , Cell Proliferation , Cells, Cultured , Drug Evaluation, Preclinical , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Retinal Artery/drug effects , Retinal Artery/metabolism , Retinal Artery/pathology , Up-Regulation/genetics , Up-Regulation/physiology , p120 GTPase Activating Protein/metabolism
8.
Proc Natl Acad Sci U S A ; 107(9): 4299-304, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20154271

ABSTRACT

Kinases are known to regulate fundamental processes in cancer including tumor proliferation, metastasis, neovascularization, and chemoresistance. Accordingly, kinase inhibitors have been a major focus of drug development, and several kinase inhibitors are now approved for various cancer indications. Typically, kinase inhibitors are selected via high-throughput screening using catalytic kinase domains at low ATP concentration, and this process often yields ATP mimetics that lack specificity and/or function poorly in cells where ATP levels are high. Molecules targeting the allosteric site in the inactive kinase conformation (type II inhibitors) provide an alternative for developing selective inhibitors that are physiologically active. By applying a rational design approach using a constrained amino-triazole scaffold predicted to stabilize kinases in the inactive state, we generated a series of selective type II inhibitors of PDGFRbeta and B-RAF, important targets for pericyte recruitment and endothelial cell survival, respectively. These molecules were designed in silico and screened for antivascular activity in both cell-based models and a Tg(fli1-EGFP) zebrafish embryogenesis model. Dual inhibition of PDGFRbeta and B-RAF cellular signaling demonstrated synergistic antiangiogenic activity in both zebrafish and murine models of angiogenesis, and a combination of previously characterized PDGFRbeta and RAF inhibitors validated the synergy. Our lead compound was selected as an orally active molecule with favorable pharmacokinetic properties which demonstrated target inhibition in vivo leading to suppression of murine orthotopic tumors in both the kidney and pancreas.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/pathology , Cell Division/drug effects , Kidney Neoplasms/pathology , Neovascularization, Pathologic , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Administration, Oral , Angiogenesis Inhibitors/therapeutic use , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Zebrafish
9.
Proc Natl Acad Sci U S A ; 107(5): 2189-94, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080647

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies. To discover functionally relevant modulators of pancreatic neoplasia, we performed activity-based proteomic profiling on primary human ductal adenocarcinomas. Here, we identify retinoblastoma-binding protein 9 (RBBP9) as a tumor-associated serine hydrolase that displays elevated activity in pancreatic carcinomas. Whereas RBBP9 is expressed in normal and malignant tissues at similar levels, its elevated activity in tumor cells promotes anchorage-independent growth in vitro as well as pancreatic carcinogenesis in vivo. At the molecular level, RBBP9 activity overcomes TGF-beta-mediated antiproliferative signaling by reducing Smad2/3 phosphorylation, a previously unknown role for a serine hydrolase in cancer biology. Conversely, loss of endogenous RBBP9 or expression of mutationally inactive RBBP9 leads to elevated Smad2/3 phosphorylation, implicating this serine hydrolase as an essential suppressor of TGF-beta signaling. Finally, RBBP9-mediated suppression of TGF-beta signaling is required for E-cadherin expression as loss of the serine hydrolase activity leads to a reduction in E-cadherin levels and a concomitant decrease in the integrity of tumor cell-cell junctions. These data not only define a previously uncharacterized serine hydrolase activity associated with epithelial neoplasia, but also demonstrate the potential benefit of functional proteomics in the identification of new therapeutic targets.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/enzymology , Amino Acid Sequence , Animals , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/genetics , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Phosphorylation , Proteomics , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
10.
J Cell Sci ; 122(Pt 11): 1812-22, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435802

ABSTRACT

In this study, we present data showing that Cdc42-dependent lumen formation by endothelial cells (ECs) in three-dimensional (3D) collagen matrices involves coordinated signaling by PKCepsilon in conjunction with the Src-family kinases (SFKs) Src and Yes. Activated SFKs interact with Cdc42 in multiprotein signaling complexes that require PKCepsilon during this process. Src and Yes are differentially expressed during EC lumen formation and siRNA suppression of either kinase, but not Fyn or Lyn, results in significant inhibition of EC lumen formation. Concurrent with Cdc42 activation, PKCepsilon- and SFK-dependent signaling converge to activate p21-activated kinase (Pak)2 and Pak4 in steps that are also required for EC lumen formation. Pak2 and Pak4 further activate two Raf kinases, B-Raf and C-Raf, leading to ERK1 and ERK2 (ERK1/2) activation, which all seem to be necessary for EC lumen formation. This work reveals a multicomponent kinase signaling pathway downstream of integrin-matrix interactions and Cdc42 activation involving PKCepsilon, Src, Yes, Pak2, Pak4, B-Raf, C-Raf and ERK1/2 to control EC lumen formation in 3D collagen matrices.


Subject(s)
Endothelium , Protein Kinase C-epsilon/metabolism , Signal Transduction/physiology , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , raf Kinases/metabolism , src-Family Kinases/metabolism , Cells, Cultured , Collagen/metabolism , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium/cytology , Endothelium/physiology , Enzyme Activation , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Multiprotein Complexes/metabolism , Protein Kinase C-epsilon/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tissue Culture Techniques , Tissue Scaffolds , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases/genetics , raf Kinases/genetics , src-Family Kinases/genetics
11.
Proc Natl Acad Sci U S A ; 105(27): 9343-8, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18607000

ABSTRACT

Integrin alphanubeta3 is found on a subset of tumor blood vessels where it is associated with angiogenesis and malignant tumor growth. We designed an alphanubeta3-targeted nanoparticle (NP) encapsulating the cytotoxic drug doxorubicin (Dox) for targeted drug delivery to the alphanubeta3-expressing tumor vasculature. We observed real-time targeting of this NP to tumor vessels and noted selective apoptosis in regions of the alphanubeta3-expressing tumor vasculature. In clinically relevant pancreatic and renal cell orthotopic models of spontaneous metastasis, targeted delivery of Dox produced an antimetastatic effect. In fact, alphanubeta3-mediated delivery of this drug to the tumor vasculature resulted in a 15-fold increase in antimetastatic activity without producing drug-associated weight loss as observed with systemic administration of the free drug. These findings reveal that NP-based delivery of cytotoxic drugs to the alphanubeta3-positive tumor vasculature represents an approach for treating metastatic disease.


Subject(s)
Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Delivery Systems , Nanoparticles , Neoplasm Metastasis/drug therapy , Neoplasms/blood supply , Neoplasms/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/pharmacology , Endothelium/drug effects , Endothelium/pathology , Integrin alphaVbeta3/metabolism , Kidney Neoplasms/pathology , Mice , Neovascularization, Pathologic/pathology , Oligopeptides/pharmacology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...