Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
ChemMedChem ; 16(7): 1126-1132, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33314657

ABSTRACT

Rev1 is a protein scaffold of the translesion synthesis (TLS) pathway, which employs low-fidelity DNA polymerases for replication of damaged DNA. The TLS pathway helps cancers tolerate DNA damage induced by genotoxic chemotherapy, and increases mutagenesis in tumors, thus accelerating the onset of chemoresistance. TLS inhibitors have emerged as potential adjuvant drugs to enhance the efficacy of first-line chemotherapy, with the majority of reported inhibitors targeting protein-protein interactions (PPIs) of the Rev1 C-terminal domain (Rev1-CT). We previously identified phenazopyridine (PAP) as a scaffold to disrupt Rev1-CT PPIs with Rev1-interacting regions (RIRs) of TLS polymerases. To explore the structure-activity relationships for this scaffold, we developed a protocol for co-crystallization of compounds that target the RIR binding site on Rev1-CT with a triple Rev1-CT/Rev7R124A /Rev3-RBM1 complex, and solved an X-ray crystal structure of Rev1-CT bound to the most potent PAP analogue. The structure revealed an unexpected binding pose of the compound and informed changes to the scaffold to improve its affinity for Rev1-CT. We synthesized eight additional PAP derivatives, with modifications to the scaffold driven by the structure, and evaluated their binding to Rev1-CT by microscale thermophoresis (MST). Several second-generation PAP derivatives showed an affinity for Rev1-CT that was improved by over an order of magnitude, thereby validating the structure-based assumptions that went into the compound design.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Phenazopyridine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Phenazopyridine/chemical synthesis , Phenazopyridine/chemistry , Structure-Activity Relationship
2.
ChemMedChem ; 14(17): 1610-1617, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31361935

ABSTRACT

Translesion synthesis (TLS) has emerged as a mechanism through which several forms of cancer develop acquired resistance to first-line genotoxic chemotherapies by allowing replication to continue in the presence of damaged DNA. Small molecules that inhibit TLS hold promise as a novel class of anticancer agents that can serve to enhance the efficacy of these front-line therapies. We previously used a structure-based rational design approach to identify the phenazopyridine scaffold as an inhibitor of TLS that functions by disrupting the protein-protein interaction (PPI) between the C-terminal domain of the TLS DNA polymerase Rev1 (Rev1-CT) and the Rev1 interacting regions (RIR) of other TLS DNA polymerases. To continue the identification of small molecules that disrupt the Rev1-CT/RIR PPI, we generated a pharmacophore model based on the phenazopyridine scaffold and used it in a structure-based virtual screen. In vitro analysis of promising hits identified several new chemotypes with the ability to disrupt this key TLS PPI. In addition, several of these compounds were found to enhance the efficacy of cisplatin in cultured cells, highlighting their anti-TLS potential.


Subject(s)
Azo Compounds/pharmacology , DNA-Directed DNA Polymerase/metabolism , Nucleotidyltransferases/metabolism , Protein Binding/drug effects , Pyridines/pharmacology , Animals , DNA-Directed DNA Polymerase/chemistry , Drug Evaluation, Preclinical , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Nucleotidyltransferases/chemistry , Protein Domains
3.
Bioorg Med Chem ; 26(14): 4301-4309, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30037752

ABSTRACT

Translesion synthesis (TLS) is a DNA damage tolerance mechanism that allows replicative bypass of DNA lesions, including DNA adducts formed by cancer chemotherapeutics. Previous studies demonstrated that suppression of TLS can increase sensitivity of cancer cells to first-line chemotherapeutics and decrease mutagenesis linked to the onset of chemoresistance, marking the TLS pathway as an emerging therapeutic target. TLS is mediated by a heteroprotein complex consisting of specialized DNA polymerases, including the Y-family DNA polymerase Rev1. Previously, we developed a screening assay to identify the first small molecules that disrupt the protein-protein interaction between the C-terminal domain of Rev1 (Rev1-CT) and the Rev1-interacting region (RIR) present in multiple DNA polymerases involved in TLS. Herein we report additional hit scaffolds that inhibit this key TLS PPI. In addition, through a series of biochemical, computational, and cellular studies we have identified preliminary structure-activity relationships and determined initial pharmacokinetic parameters for our original hits.


Subject(s)
Antineoplastic Agents/pharmacology , Nuclear Proteins/antagonists & inhibitors , Nucleotidyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Thiophenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Protein Binding/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
4.
ACS Chem Biol ; 12(7): 1903-1912, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28541665

ABSTRACT

Translesion synthesis (TLS) is an important mechanism through which proliferating cells tolerate DNA damage during replication. The mutagenic Rev1/Polζ-dependent branch of TLS helps cancer cells survive first-line genotoxic chemotherapy and introduces mutations that can contribute to the acquired resistance so often observed with standard anticancer regimens. As such, inhibition of Rev1/Polζ-dependent TLS has recently emerged as a strategy to enhance the efficacy of first-line chemotherapy and reduce the acquisition of chemoresistance by decreasing tumor mutation rate. The TLS DNA polymerase Rev1 serves as an integral scaffolding protein that mediates the assembly of the active multiprotein TLS complexes. Protein-protein interactions (PPIs) between the C-terminal domain of Rev1 (Rev1-CT) and the Rev1-interacting region (RIR) of other TLS DNA polymerases play an essential role in regulating TLS activity. To probe whether disrupting the Rev1-CT/RIR PPI is a valid approach for developing a new class of targeted anticancer agents, we designed a fluorescence polarization-based assay that was utilized in a pilot screen for small molecule inhibitors of this PPI. Two small molecule scaffolds that disrupt this interaction were identified, and secondary validation assays confirmed that compound 5 binds to Rev1-CT at the RIR interface. Finally, survival and mutagenesis assays in mouse embryonic fibroblasts and human fibrosarcoma HT1080 cells treated with cisplatin and ultraviolet light indicate that these compounds inhibit mutagenic Rev1/Polζ-dependent TLS in cells, validating the Rev1-CT/RIR PPI for future anticancer drug discovery and identifying the first small molecule inhibitors of TLS that target Rev1-CT.


Subject(s)
DNA-Directed DNA Polymerase , Drug Delivery Systems , Nuclear Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleotidyltransferases/metabolism , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Drug Discovery , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Molecular Dynamics Simulation , Nuclear Proteins/genetics , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleotidyltransferases/genetics , Proteins/chemistry , Small Molecule Libraries/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL