Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Med Chem ; 62(24): 11232-11259, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31724864

ABSTRACT

The treatment of tumors driven by overexpression or amplification of MYC oncogenes remains a significant challenge in drug discovery. Here, we present a new strategy toward the inhibition of MYC via the disruption of the protein-protein interaction between MYC and its chromatin cofactor WD Repeat-Containing Protein 5. Blocking the association of these proteins is hypothesized to disrupt the localization of MYC to chromatin, thus disrupting the ability of MYC to sustain tumorigenesis. Utilizing a high-throughput screening campaign and subsequent structure-guided design, we identify small-molecule inhibitors of this interaction with potent in vitro binding affinity and report structurally related negative controls that can be used to study the effect of this disruption. Our work suggests that disruption of this protein-protein interaction may provide a path toward an effective approach for the treatment of multiple tumors and anticipate that the molecules disclosed can be used as starting points for future efforts toward compounds with improved drug-like properties.


Subject(s)
Drug Discovery , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Salicylic Acid/chemistry , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-myc/metabolism , WD40 Repeats
2.
J Med Chem ; 61(13): 5623-5642, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29889518

ABSTRACT

WDR5 is a chromatin-regulatory scaffold protein overexpressed in various cancers and a potential epigenetic drug target for the treatment of mixed-lineage leukemia. Here, we describe the discovery of potent and selective WDR5-WIN-site inhibitors using fragment-based methods and structure-based design. NMR-based screening of a large fragment library identified several chemically distinct hit series that bind to the WIN site within WDR5. Members of a 6,7-dihydro-5 H-pyrrolo[1,2- a]imidazole fragment class were expanded using a structure-based design approach to arrive at lead compounds with dissociation constants <10 nM and micromolar cellular activity against an AML-leukemia cell line. These compounds represent starting points for the discovery of clinically useful WDR5 inhibitors for the treatment of cancer.


Subject(s)
Drug Design , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Imidazoles/chemistry , Imidazoles/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Structure-Activity Relationship
3.
Anal Biochem ; 548: 44-52, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29444450

ABSTRACT

K-RAS is mutated in approximately 30% of human cancers, resulting in increased RAS signaling and tumor growth. Thus, RAS is a highly validated therapeutic target, especially in tumors of the pancreas, lung and colon. Although directly targeting RAS has proven to be challenging, it may be possible to target other proteins involved in RAS signaling, such as the guanine nucleotide exchange factor Son of Sevenless (SOS). We have previously reported on the discovery of small molecules that bind to SOS1, activate SOS-mediated nucleotide exchange on RAS, and paradoxically inhibit ERK phosphorylation (Burns et al., PNAS, 2014). Here, we describe the discovery of additional, structurally diverse small molecules that also bind to SOS1 in the same pocket and elicit similar biological effects. We tested >160,000 compounds in a fluorescence-based assay to assess their effects on SOS-mediated nucleotide exchange. X-Ray structures revealed that these small molecules bind to the CDC25 domain of SOS1. Compounds that elicited high levels of nucleotide exchange activity in vitro increased RAS-GTP levels in cells, and inhibited phospho ERK levels at higher treatment concentrations. The identification of structurally diverse SOS1 binding ligands may assist in the discovery of new molecules designed to target RAS-driven tumors.


Subject(s)
MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/metabolism , HeLa Cells , Humans , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , SOS1 Protein/chemistry , SOS1 Protein/genetics
4.
J Med Chem ; 61(6): 2410-2421, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29323899

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1), an antiapoptotic member of the Bcl-2 family of proteins, has emerged as an attractive target for cancer therapy. Mcl-1 upregulation is often found in many human cancers and is associated with high tumor grade, poor survival, and resistance to chemotherapy. Here, we describe a series of potent and selective tricyclic indole diazepinone Mcl-1 inhibitors that were discovered and further optimized using structure-based design. These compounds exhibit picomolar binding affinity and mechanism-based cellular efficacy, including growth inhibition and caspase induction in Mcl-1-sensitive cells. Thus, they represent useful compounds to study the implication of Mcl-1 inhibition in cancer and serve as potentially useful starting points toward the discovery of anti-Mcl-1 therapeutics.


Subject(s)
Azepines/chemical synthesis , Azepines/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Apoptosis , Caspases/metabolism , Cell Division/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Enzyme Activators/chemical synthesis , Enzyme Activators/pharmacology , Humans , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Structure-Activity Relationship
5.
FEBS Lett ; 591(1): 240-251, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878989

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1) is an antiapoptotic member of the Bcl-2 family of proteins that when overexpressed is associated with high tumor grade, poor survival, and resistance to chemotherapy. Mcl-1 is amplified in many human cancers, and knockdown of Mcl-1 using RNAi can lead to apoptosis. Thus, Mcl-1 is a promising cancer target. Here, we describe the discovery of picomolar Mcl-1 inhibitors that cause caspase activation, mitochondrial depolarization, and selective growth inhibition. These compounds represent valuable tools to study the role of Mcl-1 in cancer and serve as useful starting points for the discovery of clinically useful Mcl-1 inhibitors. PDB ID CODES: Comp. 2: 5IEZ; Comp. 5: 5IF4.


Subject(s)
Antineoplastic Agents/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Bcl-2-Like Protein 11/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Design , Drug Discovery , Humans , Immunoprecipitation , Membrane Potential, Mitochondrial/drug effects , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , bcl-X Protein/metabolism
6.
J Med Chem ; 59(5): 2054-66, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26878343

ABSTRACT

Myeloid cell leukemia-1 (Mcl-1) is a member of the Bcl-2 family of proteins responsible for the regulation of programmed cell death. Amplification of Mcl-1 is a common genetic aberration in human cancer whose overexpression contributes to the evasion of apoptosis and is one of the major resistance mechanisms for many chemotherapies. Mcl-1 mediates its effects primarily through interactions with pro-apoptotic BH3 containing proteins that achieve high affinity for the target by utilizing four hydrophobic pockets in its binding groove. Here we describe the discovery of Mcl-1 inhibitors using fragment-based methods and structure-based design. These novel inhibitors exhibit low nanomolar binding affinities to Mcl-1 and >500-fold selectivity over Bcl-xL. X-ray structures of lead Mcl-1 inhibitors when complexed to Mcl-1 provided detailed information on how these small-molecules bind to the target and were used extensively to guide compound optimization.


Subject(s)
Drug Discovery , Indoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Sulfonamides/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
7.
ChemMedChem ; 11(8): 893-9, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26748787

ABSTRACT

Replication protein A (RPA) is an essential single-stranded DNA (ssDNA)-binding protein that initiates the DNA damage response pathway through protein-protein interactions (PPIs) mediated by its 70N domain. The identification and use of chemical probes that can specifically disrupt these interactions is important for validating RPA as a cancer target. A high-throughput screen (HTS) to identify new chemical entities was conducted, and 90 hit compounds were identified. From these initial hits, an anthranilic acid based series was optimized by using a structure-guided iterative medicinal chemistry approach to yield a cell-penetrant compound that binds to RPA70N with an affinity of 812 nm. This compound, 2-(3- (N-(3,4-dichlorophenyl)sulfamoyl)-4-methylbenzamido)benzoic acid (20 c), is capable of inhibiting PPIs mediated by this domain.


Subject(s)
Replication Protein A/antagonists & inhibitors , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/pharmacology , Anisotropy , Dose-Response Relationship, Drug , Fluorescence Polarization , High-Throughput Screening Assays , Models, Molecular , Molecular Structure , Structure-Activity Relationship , ortho-Aminobenzoates/chemical synthesis
8.
J Med Chem ; 58(9): 3794-805, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25844895

ABSTRACT

Myeloid cell leukemia-1 (Mcl-1) is an antiapoptotic member of the Bcl-2 family of proteins that is overexpressed and amplified in many cancers. Overexpression of Mcl-1 allows cancer cells to evade apoptosis and contributes to the resistance of cancer cells to be effectively treated with various chemotherapies. From an NMR-based screen of a large fragment library, several distinct chemical scaffolds that bind to Mcl-1 were discovered. Here, we describe the discovery of potent tricyclic 2-indole carboxylic acid inhibitors that exhibit single digit nanomolar binding affinity to Mcl-1 and greater than 1700-fold selectivity over Bcl-xL and greater than 100-fold selectivity over Bcl-2. X-ray structures of these compounds when complexed to Mcl-1 provide detailed information on how these small-molecules bind to the target, which was used to guide compound optimization.


Subject(s)
Heterocyclic Compounds, 3-Ring/chemistry , Indoles/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Crystallography, X-Ray , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Indoles/chemical synthesis , Indoles/pharmacology , K562 Cells , Models, Molecular , Molecular Conformation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Structure-Activity Relationship , bcl-X Protein/chemistry , bcl-X Protein/metabolism
9.
J Biomol NMR ; 60(1): 11-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25087006

ABSTRACT

K-Ras is a well-validated cancer target but is considered to be "undruggable" due to the lack of suitable binding pockets. We previously discovered small molecules that bind weakly to K-Ras but wanted to improve their binding affinities by identifying ligands that bind near our initial hits that we could link together. Here we describe an approach for identifying second site ligands that uses a cysteine residue to covalently attach a compound for tight binding to the first site pocket followed by a fragment screen for binding to a second site. This approach could be very useful for targeting Ras and other challenging drug targets.


Subject(s)
Drug Discovery/methods , Models, Molecular , Proto-Oncogene Proteins p21(ras)/chemistry , Cysteine/chemistry , Cysteine/metabolism , Ligands , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Binding , Proto-Oncogene Proteins p21(ras)/metabolism
10.
Proc Natl Acad Sci U S A ; 111(9): 3401-6, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550516

ABSTRACT

Aberrant activation of the small GTPase Ras by oncogenic mutation or constitutively active upstream receptor tyrosine kinases results in the deregulation of cellular signals governing growth and survival in ∼30% of all human cancers. However, the discovery of potent inhibitors of Ras has been difficult to achieve. Here, we report the identification of small molecules that bind to a unique pocket on the Ras:Son of Sevenless (SOS):Ras complex, increase the rate of SOS-catalyzed nucleotide exchange in vitro, and modulate Ras signaling pathways in cells. X-ray crystallography of Ras:SOS:Ras in complex with these molecules reveals that the compounds bind in a hydrophobic pocket in the CDC25 domain of SOS adjacent to the Switch II region of Ras. The structure-activity relationships exhibited by these compounds can be rationalized on the basis of multiple X-ray cocrystal structures. Mutational analyses confirmed the functional relevance of this binding site and showed it to be essential for compound activity. These molecules increase Ras-GTP levels and disrupt MAPK and PI3K signaling in cells at low micromolar concentrations. These small molecules represent tools to study the acute activation of Ras and highlight a pocket on SOS that may be exploited to modulate Ras signaling.


Subject(s)
Indoles/metabolism , Models, Molecular , Multiprotein Complexes/metabolism , Piperidines/metabolism , Protein Conformation , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , SOS1 Protein/metabolism , Chromatography, Liquid , Chromatography, Thin Layer , Crystallography, X-Ray , Fluorescence Polarization , HeLa Cells , Humans , Ligands , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Multiprotein Complexes/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , SOS1 Protein/chemistry
11.
J Med Chem ; 56(22): 9242-50, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24147804

ABSTRACT

Replication protein A (RPA), the major eukaryotic single-stranded DNA (ssDNA)-binding protein, is involved in nearly all cellular DNA transactions. The RPA N-terminal domain (RPA70N) is a recruitment site for proteins involved in DNA-damage response and repair. Selective inhibition of these protein-protein interactions has the potential to inhibit the DNA-damage response and to sensitize cancer cells to DNA-damaging agents without affecting other functions of RPA. To discover a potent, selective inhibitor of the RPA70N protein-protein interactions to test this hypothesis, we used NMR spectroscopy to identify fragment hits that bind to two adjacent sites in the basic cleft of RPA70N. High-resolution X-ray crystal structures of RPA70N-ligand complexes revealed how these fragments bind to RPA and guided the design of linked compounds that simultaneously occupy both sites. We have synthesized linked molecules that bind to RPA70N with submicromolar affinity and minimal disruption of RPA's interaction with ssDNA.


Subject(s)
Drug Discovery , Replication Protein A/metabolism , DNA, Single-Stranded/metabolism , Drug Design , Drug Evaluation, Preclinical , Ligands , Models, Molecular , Protein Binding/drug effects , Protein Structure, Tertiary , Replication Protein A/chemistry
12.
J Med Chem ; 56(1): 15-30, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23244564

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1), a member of the Bcl-2 family of proteins, is overexpressed and amplified in various cancers and promotes the aberrant survival of tumor cells that otherwise would undergo apoptosis. Here we describe the discovery of potent and selective Mcl-1 inhibitors using fragment-based methods and structure-based design. NMR-based screening of a large fragment library identified two chemically distinct hit series that bind to different sites on Mcl-1. Members of the two fragment classes were merged together to produce lead compounds that bind to Mcl-1 with a dissociation constant of <100 nM with selectivity for Mcl-1 over Bcl-xL and Bcl-2. Structures of merged compounds when complexed to Mcl-1 were obtained by X-ray crystallography and provide detailed information about the molecular recognition of small-molecule ligands binding Mcl-1. The compounds represent starting points for the discovery of clinically useful Mcl-1 inhibitors for the treatment of a wide variety of cancers.


Subject(s)
Antineoplastic Agents/chemistry , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Small Molecule Libraries/chemistry , Crystallography, X-Ray , Databases, Factual , Drug Design , Humans , Indoles/chemical synthesis , Indoles/chemistry , Ligands , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Molecular Structure , Myeloid Cell Leukemia Sequence 1 Protein , Protein Binding , Proto-Oncogene Proteins c-bcl-2/chemistry , Structure-Activity Relationship , bcl-X Protein/chemistry
13.
Anal Biochem ; 393(2): 176-81, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19595984

ABSTRACT

Obtaining highly purified proteins is essential to begin investigating their functional and structural properties. The steps that are typically involved in purifying proteins can include an initial capture, intermediate purification, and a final polishing step. Completing these steps can take several days and require frequent attention to ensure success. Our goal was to design automated protocols that would allow the purification of proteins with minimal operator intervention. Separate methods have been produced and tested that automate the sample loading, column washing, sample elution and peak collection steps for ion exchange, metal affinity, hydrophobic interaction, and gel filtration chromatography. These individual methods are designed to be coupled and run sequentially in any order to achieve a flexible and fully automated protein purification protocol.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/isolation & purification , Chromatography, Liquid/methods , Recombinant Proteins/isolation & purification , Automation , Bacterial Proteins/biosynthesis , Bacterial Proteins/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Recombinant Proteins/biosynthesis , Streptococcus pneumoniae/enzymology , Transformation, Bacterial , Vibrio cholerae/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...