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.
ACS Chem Biol ; 19(1): 173-184, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38193430

ABSTRACT

Small molecules that induce protein degradation hold the potential to overcome several limitations of the currently available inhibitors. Monovalent or molecular glue degraders, in particular, enable the benefits of protein degradation without the disadvantages of high molecular weight and the resulting challenge in drug development that are associated with bivalent molecules like Proteolysis Targeting Chimeras. One key challenge in designing monovalent degraders is how to build in the degrader activity─how can we convert an inhibitor into a degrader? If degradation activity requires very specific molecular features, it will be difficult to find new degraders and challenging to optimize those degraders toward drugs. Herein, we demonstrate that an unexpectedly wide range of modifications to the degradation-inducing group of the cyclin K degrader CR8 are tolerated, including both aromatic and nonaromatic groups. We used these findings to convert the pan-CDK inhibitors dinaciclib and AT-7519 to Cyclin K degraders, leading to a novel dinaciclib-based compound with improved degradation activity compared to CR8 and confirm the mechanism of degradation. These results suggest that general design principles can be generated for the development and optimization of monovalent degraders.


Subject(s)
Cyclins , Proteolysis , Cell Cycle Checkpoints , Cyclins/metabolism
2.
J Med Chem ; 66(8): 5892-5906, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37026591

ABSTRACT

B-cell lymphoma 6 (BCL6) is a transcriptional repressor and oncogenic driver of diffuse large B-cell lymphoma (DLBCL). Here, we report the optimization of our previously reported tricyclic quinolinone series for the inhibition of BCL6. We sought to improve the cellular potency and in vivo exposure of the non-degrading isomer, CCT373567, of our recently published degrader, CCT373566. The major limitation of our inhibitors was their high topological polar surface areas (TPSA), leading to increased efflux ratios. Reducing the molecular weight allowed us to remove polarity and decrease TPSA without considerably reducing solubility. Careful optimization of these properties, as guided by pharmacokinetic studies, led to the discovery of CCT374705, a potent inhibitor of BCL6 with a good in vivo profile. Modest in vivo efficacy was achieved in a lymphoma xenograft mouse model after oral dosing.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Quinolones , Animals , Humans , Mice , Cell Line, Tumor , Disease Models, Animal , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-6/chemistry , Transcription Factors
3.
RSC Med Chem ; 13(6): 731-736, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35814929

ABSTRACT

Various BRAF kinase inhibitors were developed to treat cancers carrying the BRAFV600E mutation. First-generation BRAF inhibitors could lead to paradoxical activation of the MAPK pathway, limiting their clinical usefulness. Here, we show the development of two series of BRAFV600E-targeting PROTACs and demonstrate that the exchange of the inhibitor scaffold from vemurafenib to paradox-breaker ligands resulted in BRAFV600E degraders that did not cause paradoxical ERK activation.

4.
J Med Chem ; 65(12): 8191-8207, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35653645

ABSTRACT

The transcriptional repressor BCL6 is an oncogenic driver found to be deregulated in lymphoid malignancies. Herein, we report the optimization of our previously reported benzimidazolone molecular glue-type degrader CCT369260 to CCT373566, a highly potent probe suitable for sustained depletion of BCL6 in vivo. We observed a sharp degradation SAR, where subtle structural changes conveyed the ability to induce degradation of BCL6. CCT373566 showed modest in vivo efficacy in a lymphoma xenograft mouse model following oral dosing.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , Animals , Humans , Mice , Proto-Oncogene Proteins c-bcl-6/metabolism
5.
J Cell Sci ; 132(14)2019 07 15.
Article in English | MEDLINE | ID: mdl-31217285

ABSTRACT

Growth factor-induced signal transduction pathways are tightly regulated at multiple points intracellularly, but how cells monitor levels of extracellular ligand and translate this information into appropriate downstream responses remains unclear. Understanding signalling dynamics is thus a key challenge in determining how cells respond to external cues. Here, we demonstrate that different TGF-ß family ligands, namely activin A and BMP4, signal with distinct dynamics, which differ profoundly from those of TGF-ß itself. The signalling dynamics are driven by differences in the localisation and internalisation of receptors for each ligand, which in turn determine the capability of cells to monitor levels of extracellular ligand. By using mathematical modelling, we demonstrate that the distinct receptor behaviours and signalling dynamics observed may be primarily driven by differences in ligand-receptor affinity. Furthermore, our results provide a clear rationale for the different mechanisms of pathway regulation found in vivo for each of these growth factors.


Subject(s)
Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Ligands , Mice , Models, Biological , NIH 3T3 Cells , Protein Biosynthesis , Smad Proteins/metabolism
6.
Cell Rep ; 25(7): 1841-1855.e5, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428352

ABSTRACT

Signal transduction pathways stimulated by secreted growth factors are tightly regulated at multiple levels between the cell surface and the nucleus. The trafficking of cell surface receptors is emerging as a key step for regulating appropriate cellular responses, with perturbations in this process contributing to human diseases, including cancer. For receptors recognizing ligands of the transforming growth factor ß (TGF-ß) family, little is known about how trafficking is regulated or how this shapes signaling dynamics. Here, using whole genome small interfering RNA (siRNA) screens, we have identified the ESCRT (endosomal sorting complex required for transport) machinery as a crucial determinant of signal duration. Downregulation of ESCRT components increases the outputs of TGF-ß signaling and sensitizes cells to low doses of ligand in their microenvironment. This sensitization drives an epithelial-to-mesenchymal transition (EMT) in response to low doses of ligand, and we demonstrate a link between downregulation of the ESCRT machinery and cancer survival.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Down-Regulation , Epithelial-Mesenchymal Transition , Genome, Human , Humans , Lysosomes/metabolism , Mice , Multivesicular Bodies/metabolism , Neoplasms/pathology , Phosphorylation , Prognosis , Protein Transport , Proteolysis , Smad2 Protein/metabolism , Survival Analysis , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
7.
Elife ; 62017 02 13.
Article in English | MEDLINE | ID: mdl-28191871

ABSTRACT

NODAL/Activin signaling orchestrates key processes during embryonic development via SMAD2. How SMAD2 activates programs of gene expression that are modulated over time however, is not known. Here we delineate the sequence of events that occur from SMAD2 binding to transcriptional activation, and the mechanisms underlying them. NODAL/Activin signaling induces dramatic chromatin landscape changes, and a dynamic transcriptional network regulated by SMAD2, acting via multiple mechanisms. Crucially we have discovered two modes of SMAD2 binding. SMAD2 can bind pre-acetylated nucleosome-depleted sites. However, it also binds to unacetylated, closed chromatin, independently of pioneer factors, where it induces nucleosome displacement and histone acetylation. For a subset of genes, this requires SMARCA4. We find that long term modulation of the transcriptional responses requires continued NODAL/Activin signaling. Thus SMAD2 binding does not linearly equate with transcriptional kinetics, and our data suggest that SMAD2 recruits multiple co-factors during sustained signaling to shape the downstream transcriptional program.


Subject(s)
Activins/metabolism , Chromatin/metabolism , Gene Expression Regulation, Developmental , Nodal Protein/metabolism , Signal Transduction , Smad2 Protein/metabolism , Transcription, Genetic , Animals , Mice , Protein Binding
8.
Sci Signal ; 6(305): ra106, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24327760

ABSTRACT

Understanding the complex dynamics of growth factor signaling requires both mechanistic and kinetic information. Although signaling dynamics have been studied for pathways downstream of receptor tyrosine kinases and G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors, they have not been investigated for the transforming growth factor-ß (TGF-ß) superfamily pathways. Using an integrative experimental and mathematical modeling approach, we dissected the dynamic behavior of the TGF-ß to Smad pathway, which is mediated by type I and type II receptor serine/threonine kinases, in response to acute, chronic, and repeated ligand stimulations. TGF-ß exposure produced a transient response that attenuated over time, resulting in desensitized cells that were refractory to further acute stimulation. This loss of signaling competence depended on ligand binding, but not on receptor activity, and was restored only after the ligand had been depleted. Furthermore, TGF-ß binding triggered the rapid depletion of signaling-competent receptors from the cell surface, with the type I and type II receptors exhibiting different degradation and trafficking kinetics. A computational model of TGF-ß signal transduction from the membrane to the nucleus that incorporates our experimental findings predicts that autocrine signaling, such as that associated with tumorigenesis, severely compromises the TGF-ß response, which we confirmed experimentally. Thus, we have shown that the long-term signaling behavior of the TGF-ß pathway is determined by receptor dynamics, does not require TGF-ß-induced gene expression, and influences context-dependent responses in vivo.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism , Blotting, Western , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Kinetics , Ligands , Models, Biological , Phosphorylation , Protein Binding , Protein Transport/drug effects , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction/drug effects , Smad2 Protein/metabolism , Transforming Growth Factor beta1/pharmacology
9.
Proc Natl Acad Sci U S A ; 110(27): E2490-9, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23766371

ABSTRACT

The genetic and morphological development of colorectal cancer is a paradigm for tumorigenesis. However, the dynamics of clonal evolution underpinning carcinogenesis remain poorly understood. Here we identify multipotential stem cells within human colorectal adenomas and use methylation patterns of nonexpressed genes to characterize clonal evolution. Numerous individual crypts from six colonic adenomas and a hyperplastic polyp were microdissected and characterized for genetic lesions. Clones deficient in cytochrome c oxidase (CCO(-)) were identified by histochemical staining followed by mtDNA sequencing. Topographical maps of clone locations were constructed using a combination of these data. Multilineage differentiation within clones was demonstrated by immunofluorescence. Methylation patterns of adenomatous crypts were determined by clonal bisulphite sequencing; methylation pattern diversity was compared with a mathematical model to infer to clonal dynamics. Individual adenomatous crypts were clonal for mtDNA mutations and contained both mucin-secreting and neuroendocrine cells, demonstrating that the crypt contained a multipotent stem cell. The intracrypt methylation pattern was consistent with the crypts containing multiple competing stem cells. Adenomas were epigenetically diverse populations, suggesting that they were relatively mitotically old populations. Intratumor clones typically showed less diversity in methylation pattern than the tumor as a whole. Mathematical modeling suggested that recent clonal sweeps encompassing the whole adenoma had not occurred. Adenomatous crypts within human tumors contain actively dividing stem cells. Adenomas appeared to be relatively mitotically old populations, pocketed with occasional newly generated subclones that were the result of recent rapid clonal expansion. Relative stasis and occasional rapid subclone growth may characterize colorectal tumorigenesis.


Subject(s)
Adenoma/pathology , Cell Lineage/genetics , Colorectal Neoplasms/pathology , Multipotent Stem Cells/pathology , Neoplastic Stem Cells/pathology , Adenoma/genetics , Adenoma/metabolism , Cell Differentiation/genetics , Clone Cells/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA, Mitochondrial/genetics , DNA, Neoplasm/genetics , Epigenesis, Genetic , Humans , Models, Biological , Multipotent Stem Cells/metabolism , Mutation , Neoplastic Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...