Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell Rep ; 34(3): 108636, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33472061

ABSTRACT

The chromatin-associated protein WDR5 is a promising pharmacological target in cancer, with most drug discovery efforts directed against an arginine-binding cavity in WDR5 called the WIN site. Despite a clear expectation that WIN site inhibitors will alter the repertoire of WDR5 interaction partners, their impact on the WDR5 interactome remains unknown. Here, we use quantitative proteomics to delineate how the WDR5 interactome is changed by WIN site inhibition. We show that the WIN site inhibitor alters the interaction of WDR5 with dozens of proteins, including those linked to phosphatidylinositol 3-kinase (PI3K) signaling. As proof of concept, we demonstrate that the master kinase PDPK1 is a bona fide high-affinity WIN site binding protein that engages WDR5 to modulate transcription of genes expressed in the G2 phase of the cell cycle. This dataset expands our understanding of WDR5 and serves as a resource for deciphering the action of WIN site inhibitors.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/chemistry , 3-Phosphoinositide-Dependent Protein Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Amino Acid Sequence , Binding Sites , Drug Discovery , G2 Phase/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Models, Molecular , Molecular Targeted Therapy , Protein Binding
2.
J Med Chem ; 63(15): 8325-8337, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32673492

ABSTRACT

The nucleotide exchange factor Son of Sevenless (SOS) catalyzes the activation of RAS by converting it from its inactive GDP-bound state to its active GTP-bound state. Recently, we have reported the discovery of small-molecule allosteric activators of SOS1 that can increase the amount of RAS-GTP in cells. The compounds can inhibit ERK phosphorylation at higher concentrations by engaging a feedback mechanism. To further study this process, we sought different chemical matter from an NMR-based fragment screen using selective methyl labeling. To aid this process, several Ile methyl groups located in different binding sites of the protein were assigned and used to categorize the NMR hits into different classes. Hit to lead optimization using an iterative structure-based design paradigm resulted in compounds with improvements in binding affinity. These improved molecules of a different chemical class increase SOS1cat-mediated nucleotide exchange on RAS and display cellular action consistent with our prior results.


Subject(s)
Guanosine Triphosphate/metabolism , SOS1 Protein/agonists , SOS1 Protein/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacology , ras Proteins/metabolism , Allosteric Regulation/drug effects , Crystallography, X-Ray , Drug Design , Drug Discovery , Humans , Molecular Docking Simulation , SOS1 Protein/chemistry
3.
J Med Chem ; 63(8): 4315-4333, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32223236

ABSTRACT

The frequent deregulation of MYC and its elevated expression via multiple mechanisms drives cells to a tumorigenic state. Indeed, MYC is overexpressed in up to ∼50% of human cancers and is considered a highly validated anticancer target. Recently, we discovered that WD repeat-containing protein 5 (WDR5) binds to MYC and is a critical cofactor required for the recruitment of MYC to its target genes and reported the first small molecule inhibitors of the WDR5-MYC interaction using structure-based design. These compounds display high binding affinity, but have poor physicochemical properties and are hence not suitable for in vivo studies. Herein, we conducted an NMR-based fragment screening to identify additional chemical matter and, using a structure-based approach, we merged a fragment hit with the previously reported sulfonamide series. Compounds in this series can disrupt the WDR5-MYC interaction in cells, and as a consequence, we observed a reduction of MYC localization to chromatin.


Subject(s)
Drug Design , Drug Discovery/methods , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism , Structure-Activity Relationship
4.
Biochemistry ; 59(8): 964-969, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32032490

ABSTRACT

KLHL-12 is a substrate specific adapter protein for a Cul3-Ring ligase complex. It is a member of the Kelch ß-propeller domain subclass of Cullin-Ring substrate recognition domains. This E3 ubiquitin ligase complex has many activities, including acting as a negative regulator of the Wnt signaling pathway by mediating ubiquitination and subsequent proteolysis of Dvl3/Dsh3. KLHL-12 is also known to mediate the polyubiquitination of the dopamine D4 receptor (D4.2), the ubiquitination of KHSRP, a protein that is involved in IRES translation, and also the ubiquitination of Sec31, which is involved in endoplasmic reticulum-Golgi transport by regulating the size of COPII coats. Earlier studies broadly defined the substrate binding regions for D4.2 and Dvl3/Dsh3 to KLHL-12. We tested several peptides from these regions and succeeded in identifying a short peptide that bound to KLHL-12 with low micromolar affinity. To better understand the sequence specificity of this peptide, we used alanine substitutions to map the important residues and obtained an X-ray structure of this peptide bound to KLHL-12. This structure and our peptide affinity measurements suggest a sequence motif for peptides that bind to the top face of KLHL-12. Understanding this binding site on KLHL-12 may contribute to efforts to find small molecule ligands that can either directly inhibit the degradation of substrate proteins or be used in targeted protein degradation strategies using PROTACs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Peptides/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Humans , Mutation , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Protein Binding , Protein Domains
5.
J Med Chem ; 62(8): 3971-3988, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30929420

ABSTRACT

Overexpression of myeloid cell leukemia-1 (Mcl-1) in cancers correlates with high tumor grade and poor survival. Additionally, Mcl-1 drives intrinsic and acquired resistance to many cancer therapeutics, including B cell lymphoma 2 family inhibitors, proteasome inhibitors, and antitubulins. Therefore, Mcl-1 inhibition could serve as a strategy to target cancers that require Mcl-1 to evade apoptosis. Herein, we describe the use of structure-based design to discover a novel compound (42) that robustly and specifically inhibits Mcl-1 in cell culture and animal xenograft models. Compound 42 binds to Mcl-1 with picomolar affinity and inhibited growth of Mcl-1-dependent tumor cell lines in the nanomolar range. Compound 42 also inhibited the growth of hematological and triple negative breast cancer xenografts at well-tolerated doses. These findings highlight the use of structure-based design to identify small molecule Mcl-1 inhibitors and support the use of 42 as a potential treatment strategy to block Mcl-1 activity and induce apoptosis in Mcl-1-dependent cancers.


Subject(s)
Antineoplastic Agents/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Small Molecule Libraries/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Azepines/chemistry , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Dynamics Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Protein Structure, Tertiary , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Xenograft Model Antitumor Assays
6.
ACS Chem Biol ; 14(3): 325-331, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30735352

ABSTRACT

Activating mutations in RAS can lead to oncogenesis by enhancing downstream signaling, such as through the MAPK and PI3K pathways. Therefore, therapeutically targeting RAS may perturb multiple signaling pathways simultaneously. One method for modulating RAS signaling is to target the activity of the guanine nucleotide exchange factor SOS1. Our laboratory has discovered compounds that bind to SOS1 and activate RAS. Interestingly, these SOS1 agonist compounds elicit biphasic modulation of ERK phosphorylation and simultaneous inhibition of AKT phosphorylation levels. Here, we utilized multiple chemically distinct compounds to elucidate whether these effects on MAPK and PI3K signaling by SOS1 agonists were mechanistically linked. In addition, we used CRISPR/Cas9 gene-editing to generate clonally derived SOS1 knockout cells and identified a potent SOS1 agonist that rapidly elicited on-target molecular effects at substantially lower concentrations than those causing off-target effects. Our findings will allow us to further define the on-target utility of SOS1 agonists.


Subject(s)
Benzimidazoles/chemistry , Indoles/chemistry , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Quinazolines/chemistry , SOS1 Protein/agonists , Benzimidazoles/metabolism , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing , Humans , Indoles/metabolism , Quinazolines/metabolism
7.
Bioorg Med Chem Lett ; 29(6): 786-790, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30728114

ABSTRACT

The PD-1 immune checkpoint pathway is a highly validated target for cancer immunotherapy. Despite the potential advantages of small molecule inhibitors over antibodies, the discovery of small molecule checkpoint inhibitors has lagged behind. To discover small molecule inhibitors of the PD-1 pathway, we have utilized a fragment-based approach. Small molecules were identified that bind to PD-L1 and crystal structures of these compounds bound to PD-L1 were obtained.


Subject(s)
B7-H1 Antigen/metabolism , Small Molecule Libraries/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/chemistry , Crystallography, X-Ray , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Protein Binding , Small Molecule Libraries/chemistry
8.
Cancer Discov ; 8(12): 1566-1581, 2018 12.
Article in English | MEDLINE | ID: mdl-30185627

ABSTRACT

Suppression of apoptosis by expression of antiapoptotic BCL2 family members is a hallmark of acute myeloblastic leukemia (AML). Induced myeloid leukemia cell differentiation protein (MCL1), an antiapoptotic BCL2 family member, is commonly upregulated in AML cells and is often a primary mode of resistance to treatment with the BCL2 inhibitor venetoclax. Here, we describe VU661013, a novel, potent, selective MCL1 inhibitor that destabilizes BIM/MCL1 association, leads to apoptosis in AML, and is active in venetoclax-resistant cells and patient-derived xenografts. In addition, VU661013 was safely combined with venetoclax for synergy in murine models of AML. Importantly, BH3 profiling of patient samples and drug-sensitivity testing ex vivo accurately predicted cellular responses to selective inhibitors of MCL1 or BCL2 and showed benefit of the combination. Taken together, these data suggest a strategy of rationally using BCL2 and MCL1 inhibitors in sequence or in combination in AML clinical trials. SIGNIFICANCE: Targeting antiapoptotic proteins in AML is a key therapeutic strategy, and MCL1 is a critical antiapoptotic oncoprotein. Armed with novel MCL1 inhibitors and the potent BCL2 inhibitor venetoclax, it may be possible to selectively induce apoptosis by combining or thoughtfully sequencing these inhibitors based on a rational evaluation of AML.See related commentary by Leber et al., p. 1511.This article is highlighted in the In This Issue feature, p. 1494.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Leukemia, Myeloid/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Pyrazines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Acute Disease , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , HL-60 Cells , Humans , Indoles/chemistry , K562 Cells , Leukemia, Myeloid/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazines/chemistry , Pyrazoles/chemistry , THP-1 Cells , U937 Cells , Xenograft Model Antitumor Assays
9.
Biochemistry ; 57(32): 4952-4958, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30011190

ABSTRACT

To test for on target toxicity of a new chemical entity, it is important to have comparable binding affinities of the compound in the target proteins from humans and the test species. To evaluate our myeloid cell leukemia-1 (Mcl-1) inhibitors, we tested them against rodent Mcl-1 and found a significant loss of binding affinity when compared to that seen with human Mcl-1. To understand the affinity loss, we used sequence alignments and structures of human Mcl-1/inhibitor complexes to identify the important differences in the amino acid sequences. One difference is human L246 (F226 in rat, F227 in mouse) in the ligand binding pocket. Mutating rat F226 to a Leu restores affinity, but the mouse F227L mutant still has a ligand affinity that is lower than that of human Mcl-1. Another mutation of mouse F267, located ∼12 Šfrom the ligand pocket, to the human/rat cysteine, F267C, improved the affinity and combined with F227L resulted in a mutant mouse protein with a binding affinity similar to that of human Mcl-1. To help understand the structural components of the affinity loss, we obtained an X-ray structure of a mouse Mcl-1/inhibitor complex and identified how the residue changes reduced compound complementarity. Finally, we tested Mcl-1 of other preclinical animal models (canine, monkey, rabbit, and ferret) that are identical to humans in terms of these two residues and found that their Mcl-1 bound our compounds with affinities comparable to that of human Mcl-1. These results have implications for understanding ligand selectivity for similar proteins and for the interpretation of preclinical toxicology studies with Mcl-1 inhibitors.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Mice , Molecular Sequence Data , Protein Binding/drug effects , Rabbits , Rats , Sequence Homology, Amino Acid , Structure-Activity Relationship
10.
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
11.
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
12.
Bioorg Med Chem ; 25(12): 3087-3092, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28428041

ABSTRACT

Amplification of the gene encoding Myeloid cell leukemia-1 (Mcl-1) is one of the most common genetic aberrations in human cancer and is associated with high tumor grade and poor survival. Recently, we reported on the discovery of high affinity Mcl-1 inhibitors that elicit mechanism-based cell activity. These inhibitors are lipophilic and contain an acidic functionality which is a common chemical profile for compounds that bind to albumin in plasma. Indeed, these Mcl-1 inhibitors exhibited reduced in vitro cell activity in the presence of serum. Here we describe the structure of a lead Mcl-1 inhibitor when bound to Human Serum Albumin (HSA). Unlike many acidic lipophilic compounds that bind to drug site 1 or 2, we found that this Mcl-1 inhibitor binds predominantly to drug site 3. Site 3 of HSA may be able to accommodate larger, more rigid compounds that do not fit into the smaller drug site 1 or 2. Structural studies of molecules that bind to this third site may provide insight into how some higher molecular weight compounds bind to albumin and could be used to aid in the design of compounds with reduced albumin binding.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Serum Albumin/metabolism , Binding Sites , Cell Line , Crystallography, X-Ray , Humans , Ligands , Molecular Docking Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding , Serum Albumin/chemistry
13.
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
14.
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
15.
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
16.
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
17.
Mol Cell ; 58(3): 440-52, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25818646

ABSTRACT

MYC is an oncoprotein transcription factor that is overexpressed in the majority of malignancies. The oncogenic potential of MYC stems from its ability to bind regulatory sequences in thousands of target genes, which depends on interaction of MYC with its obligate partner, MAX. Here, we show that broad association of MYC with chromatin also depends on interaction with the WD40-repeat protein WDR5. MYC binds WDR5 via an evolutionarily conserved "MYC box IIIb" motif that engages a shallow, hydrophobic cleft on the surface of WDR5. Structure-guided mutations in MYC that disrupt interaction with WDR5 attenuate binding of MYC at ∼80% of its chromosomal locations and disable its ability to promote induced pluripotent stem cell formation and drive tumorigenesis. Our data reveal WDR5 as a key determinant for MYC recruitment to chromatin and uncover a tractable target for the discovery of anticancer therapies against MYC-driven tumors.


Subject(s)
Carcinogenesis/metabolism , Chromatin/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Anisotropy , Binding Sites/genetics , Carcinogenesis/genetics , Chromatin/chemistry , Chromatin/genetics , Fluorescence Polarization , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
18.
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
19.
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
20.
J Med Chem ; 57(6): 2455-61, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24491171

ABSTRACT

Stapled helix peptides can serve as useful tools for inhibiting protein-protein interactions but can be difficult to optimize for affinity. Here we describe the discovery and optimization of a stapled helix peptide that binds to the N-terminal domain of the 70 kDa subunit of replication protein A (RPA70N). In addition to applying traditional optimization strategies, we employed a novel approach for efficiently designing peptides containing unnatural amino acids. We discovered hot spots in the target protein using a fragment-based screen, identified the amino acid that binds to the hot spot, and selected an unnatural amino acid to incorporate, based on the structure-activity relationships of small molecules that bind to this site. The resulting stapled helix peptide potently and selectively binds to RPA70N, does not disrupt ssDNA binding, and penetrates cells. This peptide may serve as a probe to explore the therapeutic potential of RPA70N inhibition in cancer.


Subject(s)
Peptides/chemical synthesis , Peptides/pharmacology , Replication Protein A/metabolism , Alanine/metabolism , Amino Acid Sequence , Cell Line , Crystallization , Crystallography, X-Ray , DNA, Single-Stranded/metabolism , Drug Discovery , Electrophoretic Mobility Shift Assay , Fluorescence Polarization , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Penetrance , Peptides/metabolism , Protein Conformation , Replication Protein A/drug effects , Structure-Activity Relationship , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL