Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Immunity ; 57(7): 1514-1532.e15, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38788712

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.


Subject(s)
Immunogenic Cell Death , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Humans , Animals , Mice , Proteolysis/drug effects , Cell Line, Tumor , Signal Transduction/drug effects , Immunogenic Cell Death/drug effects , Necroptosis/drug effects , Necroptosis/immunology , Neoplasms/immunology , Neoplasms/drug therapy , Mice, Inbred C57BL , Antineoplastic Agents/pharmacology , Immunotherapy/methods
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.
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
4.
J Med Chem ; 65(12): 8169-8190, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35657291

ABSTRACT

To identify new chemical series with enhanced binding affinity to the BTB domain of B-cell lymphoma 6 protein, we targeted a subpocket adjacent to Val18. With no opportunities for strong polar interactions, we focused on attaining close shape complementarity by ring fusion onto our quinolinone lead series. Following exploration of different sized rings, we identified a conformationally restricted core which optimally filled the available space, leading to potent BCL6 inhibitors. Through X-ray structure-guided design, combined with efficient synthetic chemistry to make the resulting novel core structures, a >300-fold improvement in activity was obtained by the addition of seven heavy atoms.


Subject(s)
BTB-POZ Domain , Protein Binding , Proto-Oncogene Proteins c-bcl-6
5.
Diabetes Obes Metab ; 22(11): 1985-1994, 2020 11.
Article in English | MEDLINE | ID: mdl-32519798

ABSTRACT

AIM: To test the hypothesis that glucokinase activators (GKAs) induce hepatic adaptations that alter intra-hepatocyte metabolite homeostasis. METHODS: C57BL/6 mice on a standard rodent diet were treated with a GKA (AZD1656) acutely or chronically. Hepatocytes were isolated from the mice after 4 or 8 weeks of treatment for analysis of cellular metabolites and gene expression in response to substrate challenge. RESULTS: Acute exposure of mice to AZD1656 or a liver-selective GKA (PF-04991532), before a glucose tolerance test, or challenge of mouse hepatocytes with GKAs ex vivo induced various Carbohydrate response element binding protein (ChREBP) target genes, including Carbohydrate response element binding protein beta isoform (ChREBP-ß), Gckr and G6pc. Both glucokinase activation and ChREBP target gene induction by PF-04991532 were dependent on the chirality of the molecule, confirming a mechanism linked to glucokinase activation. Hepatocytes from mice treated with AZD1656 for 4 or 8 weeks had lower basal glucose 6-phosphate levels and improved ATP homeostasis during high substrate challenge. They also had raised basal ChREBP-ß mRNA and AMPK-α mRNA (Prkaa1, Prkaa2) and progressively attenuated substrate induction of some ChREBP target genes and Prkaa1 and Prkaa2. CONCLUSIONS: Chronic GKA treatment of C57BL/6 mice for 8 weeks activates liver ChREBP and improves the resilience of hepatocytes to compromised ATP homeostasis during high-substrate challenge. These changes are associated with raised mRNA levels of ChREBP-ß and both catalytic subunits of AMP-activated protein kinase.


Subject(s)
Glucokinase , Liver , Adenosine Triphosphate , Animals , Carrier Proteins/genetics , Glucokinase/genetics , Glucokinase/metabolism , Glucose , Glucose-6-Phosphate , Hepatocytes/metabolism , Homeostasis , Liver/metabolism , Mice , Mice, Inbred C57BL , Response Elements
6.
Org Biomol Chem ; 16(11): 1843-1850, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29469144

ABSTRACT

ATAD2 is an ATPase that is overexpressed in a variety of cancers and associated with a poor patient prognosis. This protein has been suggested to function as a cofactor for a range of transcription factors, including the proto-oncogene MYC and the androgen receptor. ATAD2 comprises an ATPase domain, implicated in chromatin remodelling, and a bromodomain which allows it to interact with acetylated histone tails. Dissection of the functional roles of these two domains would benefit from the availability of selective, cell-permeable pharmacological probes. An in silico evaluation of the 3D structures of various bromodomains suggested that developing small molecule ligands for the bromodomain of ATAD2 is likely to be challenging, although recent reports have shown that ATAD2 bromodomain ligands can be identified. We report a structure-guided fragment-based approach to identify lead compounds for ATAD2 bromodomain inhibitor development. Our findings indicate that the ATAD2 bromodomain can accommodate fragment hits (Mr < 200) that yield productive structure-activity relationships, and structure-guided design enabled the introduction of selectivity over BRD4.


Subject(s)
ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Drug Design , Nuclear Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Cell Cycle Proteins , Computer-Aided Design , DNA-Binding Proteins/chemistry , Humans , Ligands , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/metabolism , Nuclear Proteins/chemistry , Protein Binding , Protein Domains/drug effects , Proto-Oncogene Mas , Transcription Factors/chemistry
7.
ChemMedChem ; 12(12): 895-900, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28423228

ABSTRACT

The catalytic activity of DNA-dependent protein kinase (DNA-PK) is critical to its ability to repair lethal DNA double-strand breaks (DSBs). This includes repair of DSB lesions resulting from oxidative stress, oncogene-induced transcription, or following therapeutic treatment of cancer cells. Armed with this knowledge, many attempts have been made to identify small-molecule inhibitors of DNA-PK activity as an approach to induce tumour chemo- and radiosensitisation. This review examines the structures of known reversible and irreversible inhibitors, including those based on chromen-4-one, arylmorpholine, and benzaldehyde scaffolds. DNA-PK catalytic inhibitors, such as VX-984 (8-[(1S)-2-[[6-(4,6-dideuterio-2-methylpyrimidin-5-yl)pyrimidin-4-yl]amino]-1-methylethyl]quinoline-4-carboxamide) and M3814 ((S)-[2-chloro-4-fluoro-5-(7-morpholinoquinazolin-4-yl)phenyl]-(6-methoxypyridazin-3-yl)methanol), have now progressed into clinical development which should help to further advance our understanding of whether this approach is a promising therapeutic strategy for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , DNA-Activated Protein Kinase/metabolism , Humans , Models, Molecular , Molecular Structure , Neoplasms/pathology , Protein Kinase Inhibitors/chemistry
8.
Chem Sci ; 7(4): 2821-2826, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-28660059

ABSTRACT

Inhibitors of sulfatase-2 are putative anticancer agents, but the discovery of potent small molecules targeting this enzyme has proved challenging. Based on molecular modelling, two series of sulfatase-2 inhibitors have been developed with biphenyl and biphenyl ether scaffolds judiciously substituted with sulfamate, carboxylate and other polar groups (e.g. amino). Inhibition of aryl sulfatase A and B was also determined. The biphenyl ether derivatives were less selective for sulfatase-2 over aryl sulfatase B than the biphenyl series. All biphenyl ether derivatives inhibited aryl sulfatase A, whereas only amino derivatives inhibited aryl sulfatase B significantly. In the biphenyl series few derivatives exhibited activity against aryl sulfatase B. The trichloroethylsulfamate group was identified as a new pharmacophore enabling potent inhibition of all of the sulfatases studied.

SELECTION OF CITATIONS
SEARCH DETAIL