Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Cell ; 82(18): 3333-3349.e9, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35981542

ABSTRACT

The interaction of RB with chromatin is key to understanding its molecular functions. Here, for first time, we identify the full spectrum of chromatin-bound RB. Rather than exclusively binding promoters, as is often described, RB targets three fundamentally different types of loci (promoters, enhancers, and insulators), which are largely distinguishable by the mutually exclusive presence of E2F1, c-Jun, and CTCF. While E2F/DP facilitates RB association with promoters, AP-1 recruits RB to enhancers. Although phosphorylation in CDK sites is often portrayed as releasing RB from chromatin, we show that the cell cycle redistributes RB so that it enriches at promoters in G1 and at non-promoter sites in cycling cells. RB-bound promoters include the classic E2F-targets and are similar between lineages, but RB-bound enhancers associate with different categories of genes and vary between cell types. Thus, RB has a well-preserved role controlling E2F in G1, and it targets cell-type-specific enhancers and CTCF sites when cells enter S-phase.


Subject(s)
Chromatin , Retinoblastoma Protein , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Promoter Regions, Genetic , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factor AP-1/genetics
2.
Proc Natl Acad Sci U S A ; 113(21): E2945-54, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162353

ABSTRACT

The Wnt signaling pathways play pivotal roles in carcinogenesis. Modulation of the cell-surface abundance of Wnt receptors is emerging as an important mechanism for regulating sensitivity to Wnt ligands. Endocytosis and degradation of the Wnt receptors Frizzled (Fzd) and lipoprotein-related protein 6 (LRP6) are regulated by the E3 ubiquitin ligases zinc and ring finger 3 (ZNRF3) and ring finger protein 43 (RNF43), which are disrupted in cancer. In a genome-wide small interfering RNA screen, we identified the deubiquitylase ubiquitin-specific protease 6 (USP6) as a potent activator of Wnt signaling. USP6 enhances Wnt signaling by deubiquitylating Fzds, thereby increasing their cell-surface abundance. Chromosomal translocations in nodular fasciitis result in USP6 overexpression, leading to transcriptional activation of the Wnt/ß-catenin pathway. Inhibition of Wnt signaling using Dickkopf-1 (DKK1) or a Porcupine (PORCN) inhibitor significantly decreased the growth of USP6-driven xenograft tumors, indicating that Wnt signaling is a key target of USP6 during tumorigenesis. Our study defines an additional route to ectopic Wnt pathway activation in human disease, and identifies a potential approach to modulate Wnt signaling for therapeutic benefit.


Subject(s)
DNA-Binding Proteins/metabolism , Frizzled Receptors/metabolism , Neoplasms, Experimental/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Wnt Signaling Pathway , Animals , DNA-Binding Proteins/genetics , Frizzled Receptors/genetics , HEK293 Cells , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Neoplasms, Experimental/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
J Biol Chem ; 288(46): 33156-70, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089523

ABSTRACT

Histone deacetylase 6 (HDAC6) is well known for its ability to promote cell migration through deacetylation of its cytoplasmic substrates such as α-tubulin. However, how HDAC6 itself is regulated to control cell motility remains elusive. Previous studies have shown that one third of extracellular signal-regulated kinase (ERK) is associated with the microtubule cytoskeleton in cells. Yet, no connection between HDAC6 and ERK has been discovered. Here, for the first time, we reveal that ERK binds to and phosphorylates HDAC6 to promote cell migration via deacetylation of α-tubulin. We have identified two novel ERK-mediated phosphorylation sites: threonine 1031 and serine 1035 in HDAC6. Both sites were phosphorylated by ERK1 in vitro, whereas Ser-1035 was phosphorylated in response to the activation of EGFR-Ras-Raf-MEK-ERK signaling pathway in vivo. HDAC6-null mouse embryonic fibroblasts rescued by the nonphosphorylation mimicking mutant displayed significantly reduced cell migration compared with those rescued by the wild type. Consistently, the nonphosphorylation mimicking mutant exerted lower tubulin deacetylase activity in vivo compared with the wild type. These data indicate that ERK/HDAC6-mediated cell motility is through deacetylation of α-tubulin. Overall, our results suggest that HDAC6-mediated cell migration could be governed by EGFR-Ras-Raf-MEK-ERK signaling.


Subject(s)
Cell Movement/physiology , Histone Deacetylases/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Tubulin/metabolism , Acetylation , Animals , CHO Cells , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 3/genetics , Tubulin/genetics
4.
Cancer Discov ; 14(7): 1190-1205, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38588399

ABSTRACT

Alterations in the RAS-MAPK signaling cascade are common across multiple solid tumor types and are a driver for many cancers. NST-628 is a potent pan-RAF-MEK molecular glue that prevents the phosphorylation and activation of MEK by RAF, overcoming the limitations of traditional RAS-MAPK inhibitors and leading to deep durable inhibition of the pathway. Cellular, biochemical, and structural analyses of RAF-MEK complexes show that NST-628 engages all isoforms of RAF and prevents the formation of BRAF-CRAF heterodimers, a differentiated mechanism from all current RAF inhibitors. With a potent and durable inhibition of the RAF-MEK signaling complex as well as high intrinsic permeability into the brain, NST-628 demonstrates broad efficacy in cellular and patient-derived tumor models harboring diverse MAPK pathway alterations, including orthotopic intracranial models. Given its functional and pharmacokinetic mechanisms that are differentiated from previous therapies, NST-628 is positioned to make an impact clinically in areas of unmet patient need. Significance: This study introduces NST-628, a molecular glue having differentiated mechanism and drug-like properties. NST-628 treatment leads to broad efficacy with high tolerability and central nervous system activity across multiple RAS- and RAF-driven tumor models. NST-628 has the potential to provide transformative clinical benefits as both monotherapy and vertical combination anchor.


Subject(s)
MAP Kinase Signaling System , Neoplasms , Protein Kinase Inhibitors , Humans , Animals , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , raf Kinases/metabolism , raf Kinases/antagonists & inhibitors , Cell Line, Tumor , ras Proteins/metabolism , Xenograft Model Antitumor Assays , Brain/metabolism , Brain/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism
5.
Cell Rep ; 39(12): 110993, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732135

ABSTRACT

Although KRAS has long been considered undruggable, direct KRASG12C inhibitors have shown promising initial clinical efficacy. However, the majority of patients still fail to respond. Adaptive feedback reactivation of RAS-mitogen-activated protein kinase (MAPK) signaling has been proposed by our group and others as a key mediator of resistance, but the exact mechanism driving reactivation and the therapeutic implications are unclear. We find that upstream feedback activation of wild-type RAS, as opposed to a shift in KRASG12C to its active guanosine triphosphate (GTP)-bound state, is sufficient to drive RAS-MAPK reactivation in a KRASG12C-independent manner. Moreover, multiple receptor tyrosine kinases (RTKs) can drive feedback reactivation, potentially necessitating targeting of convergent signaling nodes for more universal efficacy. Even in colorectal cancer, where feedback is thought to be primarily epidermal growth factor receptor (EGFR)-mediated, alternative RTKs drive pathway reactivation and limit efficacy, but convergent upstream or downstream signal blockade can enhance activity. Overall, these data provide important mechanistic insight to guide therapeutic strategies targeting KRAS.


Subject(s)
Proto-Oncogene Proteins p21(ras) , Animals , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Feedback , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
6.
Cancer Discov ; 11(8): 1913-1922, 2021 08.
Article in English | MEDLINE | ID: mdl-33824136

ABSTRACT

Mutant-selective KRASG12C inhibitors, such as MRTX849 (adagrasib) and AMG 510 (sotorasib), have demonstrated efficacy in KRAS G12C-mutant cancers, including non-small cell lung cancer (NSCLC). However, mechanisms underlying clinical acquired resistance to KRASG12C inhibitors remain undetermined. To begin to define the mechanistic spectrum of acquired resistance, we describe a patient with KRAS G12C NSCLC who developed polyclonal acquired resistance to MRTX849 with the emergence of 10 heterogeneous resistance alterations in serial cell-free DNA spanning four genes (KRAS, NRAS, BRAF, MAP2K1), all of which converge to reactivate RAS-MAPK signaling. Notably, a novel KRAS Y96D mutation affecting the switch-II pocket, to which MRTX849 and other inactive-state inhibitors bind, was identified that interferes with key protein-drug interactions and confers resistance to these inhibitors in engineered and patient-derived KRAS G12C cancer models. Interestingly, a novel, functionally distinct tricomplex KRASG12C active-state inhibitor RM-018 retained the ability to bind and inhibit KRASG12C/Y96D and could overcome resistance. SIGNIFICANCE: In one of the first reports of clinical acquired resistance to KRASG12C inhibitors, our data suggest polyclonal RAS-MAPK reactivation as a central resistance mechanism. We also identify a novel KRAS switch-II pocket mutation that impairs binding and drives resistance to inactive-state inhibitors but is surmountable by a functionally distinct KRASG12C inhibitor.See related commentary by Pinnelli and Trusolino, p. 1874.This article is highlighted in the In This Issue feature, p. 1861.


Subject(s)
Acetonitriles/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Piperazines/therapeutic use , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras)/genetics
7.
Cell Rep ; 37(9): 110060, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852220

ABSTRACT

We apply genetic screens to delineate modulators of KRAS mutant pancreatic ductal adenocarcinoma (PDAC) sensitivity to ERK inhibitor treatment, and we identify components of the ATR-CHK1 DNA damage repair (DDR) pathway. Pharmacologic inhibition of CHK1 alone causes apoptotic growth suppression of both PDAC cell lines and organoids, which correlates with loss of MYC expression. CHK1 inhibition also activates ERK and AMPK and increases autophagy, providing a mechanistic basis for increased efficacy of concurrent CHK1 and ERK inhibition and/or autophagy inhibition with chloroquine. To assess how CHK1 inhibition-induced ERK activation promotes PDAC survival, we perform a CRISPR-Cas9 loss-of-function screen targeting direct/indirect ERK substrates and identify RIF1. A key component of non-homologous end joining repair, RIF1 suppression sensitizes PDAC cells to CHK1 inhibition-mediated apoptotic growth suppression. Furthermore, ERK inhibition alone decreases RIF1 expression and phenocopies RIF1 depletion. We conclude that concurrent DDR suppression enhances the efficacy of ERK and/or autophagy inhibitors in KRAS mutant PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Checkpoint Kinase 1/antagonists & inhibitors , DNA Damage , Mutation , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 26(7): 1633-1643, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31776128

ABSTRACT

PURPOSE: Although KRAS represents the most commonly mutated oncogene, it has long been considered an "undruggable" target. Novel covalent inhibitors selective for the KRASG12C mutation offer the unprecedented opportunity to target KRAS directly. However, prior efforts to target the RAS-MAPK pathway have been hampered by adaptive feedback, which drives pathway reactivation and resistance. EXPERIMENTAL DESIGN: A panel of KRASG12C cell lines were treated with the KRASG12C inhibitors ARS-1620 and AMG 510 to assess effects on signaling and viability. Isoform-specific pulldown of activated GTP-bound RAS was performed to evaluate effects on the activity of specific RAS isoforms over time following treatment. RTK inhibitors, SHP2 inhibitors, and MEK/ERK inhibitors were assessed in combination with KRASG12C inhibitors in vitro and in vivo as potential strategies to overcome resistance and enhance efficacy. RESULTS: We observed rapid adaptive RAS pathway feedback reactivation following KRASG12C inhibition in the majority of KRASG12C models, driven by RTK-mediated activation of wild-type RAS, which cannot be inhibited by G12C-specific inhibitors. Importantly, multiple RTKs can mediate feedback, with no single RTK appearing critical across all KRASG12C models. However, coinhibition of SHP2, which mediates signaling from multiple RTKs to RAS, abrogated feedback reactivation more universally, and combined KRASG12C/SHP2 inhibition drove sustained RAS pathway suppression and improved efficacy in vitro and in vivo. CONCLUSIONS: These data identify feedback reactivation of wild-type RAS as a key mechanism of adaptive resistance to KRASG12C inhibitors and highlight the potential importance of vertical inhibition strategies to enhance the clinical efficacy of KRASG12C inhibitors.See related commentary by Yaeger and Solit, p. 1538.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins p21(ras) , Feedback , Humans , Mutation , Oncogenes , Piperazines , Pyridines , Pyrimidines
9.
Cell Rep ; 26(1): 79-93.e8, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30605688

ABSTRACT

ß-Catenin-dependent WNT signal transduction governs development, tissue homeostasis, and a vast array of human diseases. Signal propagation through a WNT-Frizzled/LRP receptor complex requires proteins necessary for clathrin-mediated endocytosis (CME). Paradoxically, CME also negatively regulates WNT signaling through internalization and degradation of the receptor complex. Here, using a gain-of-function screen of the human kinome, we report that the AP2 associated kinase 1 (AAK1), a known CME enhancer, inhibits WNT signaling. Reciprocally, AAK1 genetic silencing or its pharmacological inhibition using a potent and selective inhibitor activates WNT signaling. Mechanistically, we show that AAK1 promotes clearance of LRP6 from the plasma membrane to suppress the WNT pathway. Time-course experiments support a transcription-uncoupled, WNT-driven negative feedback loop; prolonged WNT treatment drives AAK1-dependent phosphorylation of AP2M1, clathrin-coated pit maturation, and endocytosis of LRP6. We propose that, following WNT receptor activation, increased AAK1 function and CME limits WNT signaling longevity.


Subject(s)
Clathrin/metabolism , Endocytosis/physiology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Protein Serine-Threonine Kinases/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Feedback, Physiological , HEK293 Cells , Humans , Male , Mice , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors
11.
Nat Rev Clin Oncol ; 15(11): 709-720, 2018 11.
Article in English | MEDLINE | ID: mdl-30275515

ABSTRACT

RAS genes are the most commonly mutated oncogenes in cancer, but effective therapeutic strategies to target RAS-mutant cancers have proved elusive. A key aspect of this challenge is the fact that direct inhibition of RAS proteins has proved difficult, leading researchers to test numerous alternative strategies aimed at exploiting RAS-related vulnerabilities or targeting RAS effectors. In the past few years, we have witnessed renewed efforts to target RAS directly, with several promising strategies being tested in clinical trials at different stages of completion. Important advances have also been made in approaches designed to indirectly target RAS by improving inhibition of RAS effectors, exploiting synthetic lethal interactions or metabolic dependencies, using therapeutic combination strategies or harnessing the immune system. In this Review, we describe historical and ongoing efforts to target RAS-mutant cancers and outline the current therapeutic landscape in the collective quest to overcome the effects of this crucial oncogene.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , ras Proteins/genetics , Humans , Molecular Targeted Therapy , Mutation , Neoplasms/pathology , Signal Transduction/drug effects , ras Proteins/antagonists & inhibitors
12.
Disaster Med Public Health Prep ; 12(3): 345-351, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28760169

ABSTRACT

OBJECTIVE: Disasters occur without warning and can have devastating consequences. Emergency preparedness can reduce negative effects. It is especially important that parents prepare, as children are particularly vulnerable after disasters. This study tested 2 hypotheses: (1) adults with more children are likely to be better prepared than those with fewer or no children because greater caretaking is linked to greater perceived threat of disaster leading to greater preparedness and (2) the strength of this mediational link varies as a function of parental self-efficacy. METHODS: Data from an online survey about human-made disasters (terrorism) with a community convenience sample were used to test the hypothesis that perceived threat mediates the relationship between parental status (number of children cared for) and preparedness behaviors, moderated by level of self-efficacy for emergency preparedness. RESULTS: A bootstrapping analysis with relevant covariates supported the hypothesized mediating effect of threat on the relationship between parental status and preparedness. This relationship was strengthened at higher levels of parental preparedness self-efficacy. CONCLUSIONS: The results of this study are particularly relevant for preparedness interventions. Because threat leads to preparedness, particularly for parents with high self-efficacy, it is important to focus attention on factors that can improve parents' sense of self-efficacy. (Disaster Med Public Health Preparedness. 2018; 12: 345-351).


Subject(s)
Civil Defense/standards , Parenting/psychology , Perception , Self Efficacy , Terrorism/psychology , Adolescent , Adult , Aged , Civil Defense/methods , Female , Humans , Income/statistics & numerical data , Internet , Male , Middle Aged , Surveys and Questionnaires , Terrorism/statistics & numerical data
13.
Cell Rep ; 20(4): 999-1015, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28746882

ABSTRACT

Combinatorial inhibition of effector and feedback pathways is a promising treatment strategy for KRAS mutant cancers. However, the particular pathways that should be targeted to optimize therapeutic responses are unclear. Using CRISPR/Cas9, we systematically mapped the pathways whose inhibition cooperates with drugs targeting the KRAS effectors MEK, ERK, and PI3K. By performing 70 screens in models of KRAS mutant colorectal, lung, ovarian, and pancreas cancers, we uncovered universal and tissue-specific sensitizing combinations involving inhibitors of cell cycle, metabolism, growth signaling, chromatin regulation, and transcription. Furthermore, these screens revealed secondary genetic modifiers of sensitivity, yielding a SRC inhibitor-based combination therapy for KRAS/PIK3CA double-mutant colorectal cancers (CRCs) with clinical potential. Surprisingly, acquired resistance to combinations of growth signaling pathway inhibitors develops rapidly following treatment, but by targeting signaling feedback or apoptotic priming, it is possible to construct three-drug combinations that greatly delay its emergence.


Subject(s)
Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Colorectal Neoplasms/genetics , Humans , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/genetics , Signal Transduction/physiology
14.
Mol Cancer Res ; 14(10): 1009-1018, 2016 10.
Article in English | MEDLINE | ID: mdl-27418645

ABSTRACT

Recently, we identified that PREX1 overexpression is critical for metastatic but not tumorigenic growth in a mouse model of NRAS-driven melanoma. In addition, a PREX1 gene signature correlated with and was dependent on ERK MAPK activation in human melanoma cell lines. In the current study, the underlying mechanism of PREX1 overexpression in human melanoma was assessed. PREX1 protein levels were increased in melanoma tumor tissues and cell lines compared with benign nevi and normal melanocytes, respectively. Suppression of PREX1 by siRNA impaired invasion but not proliferation in vitro PREX1-dependent invasion was attributable to PREX1-mediated activation of the small GTPase RAC1 but not the related small GTPase CDC42. Pharmacologic inhibition of ERK signaling reduced PREX1 gene transcription and additionally regulated PREX1 protein stability. This ERK-dependent upregulation of PREX1 in melanoma, due to both increased gene transcription and protein stability, contrasts with the mechanisms identified in breast and prostate cancers, in which PREX1 overexpression was driven by gene amplification and HDAC-mediated gene transcription, respectively. Thus, although PREX1 expression is aberrantly upregulated and regulates RAC1 activity and invasion in these three different tumor types, the mechanisms of its upregulation are distinct and context dependent. IMPLICATIONS: This study identifies an ERK-dependent mechanism that drives PREX1 upregulation and subsequent RAC1-dependent invasion in BRAF- and NRAS-mutant melanoma. Mol Cancer Res; 14(10); 1009-18. ©2016 AACR.


Subject(s)
GTP Phosphohydrolases/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Melanoma/genetics , Membrane Proteins/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Animals , Cell Line, Tumor , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Indazoles/pharmacology , Indoles/pharmacology , MAP Kinase Signaling System , Male , Melanoma/metabolism , Mice , Piperazines/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Up-Regulation , Vemurafenib
15.
Trends Cancer ; 1(3): 183-198, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26858988

ABSTRACT

The three RAS genes comprise the most frequently mutated oncogene family in cancer. With significant and compelling evidence that continued function of mutant RAS is required for tumor maintenance, it is widely accepted that effective anti-RAS therapy will have a significant impact on cancer growth and patient survival. However, despite more than three decades of intense research and pharmaceutical industry efforts, a clinically effective anti-RAS drug has yet to be developed. With the recent renewed interest in targeting RAS, exciting and promising progress has been made. In this review, we discuss the prospects and challenges of drugging oncogenic RAS. In particular we focus on new inhibitors of RAS effector signaling and the ERK mitogen-activated protein kinase cascade.

16.
Cancer Discov ; 3(6): 690-703, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23547054

ABSTRACT

Mutations in KRAS drive the oncogenic phenotype in a variety of tumors of epithelial origin. The NF-κB transcription factor pathway is important for oncogenic RAS to transform cells and to drive tumorigenesis in animal models. Recently, TGF-ß-activated kinase 1 (TAK1), an upstream regulator of IκB kinase (IKK), which controls canonical NF-κB signaling, was shown to be important for chemoresistance in pancreatic cancer and for regulating KRAS-mutant colorectal cancer cell growth and survival. Here, we show that mutant KRAS upregulates glycogen synthase kinase 3α (GSK-3α), leading to its interaction with TAK1 to stabilize the TAK1-TAB complex to promote IKK activity. In addition, GSK-3α is required for promoting critical noncanonical NF-κB signaling in pancreatic cancer cells. Pharmacologic inhibition of GSK-3 suppresses growth of human pancreatic tumor explants, consistent with the loss of expression of oncogenic genes such as c-myc and TERT. These data identify GSK-3α as a key downstream effector of oncogenic KRAS via its ability to coordinately regulate distinct NF-κB signaling pathways.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , Glycogen Synthase Kinase 3/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Mice , NF-kappa B/genetics , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Transcription Factors , ras Proteins/genetics
17.
Cancer Chemother Pharmacol ; 63(4): 681-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18587580

ABSTRACT

Previous studies have identified interleukin 6 (IL-6) as an important cytokine with prognostic significance in ovarian cancer. Activation of the IL-6-Stat3 pathway contributes to tumor cell growth, survival and drug resistance in several cancers, including ovarian cancer. To explore potential therapeutic strategies for interrupting signaling through this pathway, we assessed the ability of CDDO-Me, a synthetic triterpenoid, to inhibit IL-6 secretion, Stat3 phosphorylation, Stat3 nuclear translocation and paclitaxel sensitivity in several cell line model systems. These studies demonstrated that CDDO-Me significantly inhibits IL-6 secretion in paclitaxel-resistant ovarian cancer cells and specifically suppresses IL-6- or oncostatin M-induced Stat3 nuclear translocation. Treatment with CDDO-Me significantly decreases the levels of Stat3, Jak2, and Src phosphorylation in ovarian and breast cancer cell lines with constitutively activated Stat3. This inhibition of the IL-6-Stat3 pathway correlated with suppression of the anti-apoptotic Stat3 target genes Bcl-X(L), survivin, and Mcl-1, and with apoptosis induction as measured by monitoring PARP and its cleavage product, as well as by quantitative measurement of the apoptosis-associated CK18Asp396. Furthermore, CDDO-Me increases the cytotoxic effects of paclitaxel in the paclitaxel-resistant ovarian cancer cell line OVCAR8(TR) (2 to 5-fold) and of cisplatin in the cisplatin-resistant ovarian cancer cell line A2780cp70 (2 to 4-fold). Our data confirm that CDDO-Me interrupts the signaling of multiple kinases involved in the IL-6-Stat3 and Src signaling pathways. Inhibition is likely achieved through multiple points within these pathways. In a model system of established acquired drug resistance, CCDO-Me is effective at partially reversing the drug-resistance phenotype.


Subject(s)
Drug Resistance, Multiple , Drug Resistance, Neoplasm , Interleukin-6/metabolism , Oleanolic Acid/analogs & derivatives , Ovarian Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Female , Humans , Interleukin-6/antagonists & inhibitors , Oleanolic Acid/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction , Tumor Cells, Cultured
18.
J Org Chem ; 72(17): 6618-20, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17655258

ABSTRACT

Isopropoxide protection of arylboronates allowed their use in metal-halogen exchange reactions. The isopropoxide-protected borate species were obtained from a boronate or in situ from dibromoarenes. meta- and para-dibromoarenes were converted via these intermediates into functionalized arylboronates in a one-pot manner.

SELECTION OF CITATIONS
SEARCH DETAIL